1
|
Wu N, Wu Z, Wang Y, Zhang A, Peng Y, Cheng Y, Lei H, Liu S, Zhao J, Li T, Zhou G. A diagnostic model for non-invasive urothelial cancer early detection based on methylation of urinary tumor DNA. Cancer Cell Int 2025; 25:148. [PMID: 40234942 PMCID: PMC12001437 DOI: 10.1186/s12935-025-03766-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2024] [Accepted: 03/26/2025] [Indexed: 04/17/2025] Open
Abstract
BACKGROUND Diagnostic methods for urothelial cancer (UC) are often invasive, while urinary cytology, a non-invasive alternative, suffers from limited sensitivity. This study aimed to identify differentially methylated markers in urinary tumor DNA and develop a diagnostic method to enhance the sensitivity of non-invasive UC detection. METHODS Whole-genome bisulfite sequencing and deep methylation sequencing were employed to identify significantly hypermethylated UC-associated genes in clinical samples and public UC datasets. Further screening was conducted using tumor biopsies and urine samples from patients, leading to the selection of three hypermethylated UC markers. A diagnostic model based on these markers was constructed and validated in a cohort (N = 432) comprising patients with UC, other cancers, benign lesions, and non-UC urinary tract diseases. RESULTS Validation in a cohort of 432 subjects demonstrated that the UC diagnostic model, incorporating three hypermethylated markers (VIM, TMEM220, and PPM1N), achieved an overall sensitivity of 94.44% in 108 UC patients. Specificities were 96.34%, 90.76%, and 87.72% in 191 non-neoplastic individuals, 76 patients with benign lesions, and 57 patients with other cancers, respectively, resulting in an overall specificity of 93.52%. Methylation level analysis revealed significantly higher methylation (P < 0.001) for three markers in UC samples compared to non-UC samples. Furthermore, the model exhibited sensitivities of 80% and 88.57% for detecting stage 0a/0is and stage I UC, respectively. CONCLUSIONS The UC diagnostic model demonstrates excellent diagnostic performance, particularly in the early detection of UC. This non-invasive approach, characterized by high sensitivity and specificity, holds significant potential for further clinical evaluation and development as a reliable tool for UC diagnosis using urine samples.
Collapse
Affiliation(s)
- Ningning Wu
- BioChain (Beijing) Science & Technology Inc., Beijing, 102600, China
| | - Zhen Wu
- BioChain (Beijing) Science & Technology Inc., Beijing, 102600, China
| | - Yanwen Wang
- Department of Urology, The First Affiliated Hospital of Dali University, 32 Carlsberg Avenue, Dali City, 671000, Yunnan Province, China
| | - Anqi Zhang
- Central Laboratory, Liaocheng People's Hospital, Liaocheng, 252000, Shandong Province, China
| | - Yongfei Peng
- BioChain (Beijing) Science & Technology Inc., Beijing, 102600, China
| | - Yan Cheng
- Central Laboratory, Liaocheng People's Hospital, Liaocheng, 252000, Shandong Province, China
| | - Hongsong Lei
- Department of Urology, The Second Affiliated Hospital of Kunming Medical University, 374 Yunnan-Burma Road, Wuhua District, Kunming, 650000, Yunnan Province, China
| | - Siwen Liu
- BioChain (Beijing) Science & Technology Inc., Beijing, 102600, China
| | - Jie Zhao
- BioChain (Beijing) Science & Technology Inc., Beijing, 102600, China.
| | - Tianbao Li
- BioChain (Beijing) Science & Technology Inc., Beijing, 102600, China.
| | - Guangpeng Zhou
- BioChain (Beijing) Science & Technology Inc., Beijing, 102600, China.
| |
Collapse
|
2
|
Li X, Liu Q, Wang L, Bu T, Yang X, Gao S, Yun D, Sun F. PPM1G dephosphorylates α-catenin to maintain the integrity of adherens junctions and regulates apoptosis in Sertoli cells. Mol Cell Endocrinol 2025; 600:112493. [PMID: 39952314 DOI: 10.1016/j.mce.2025.112493] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/17/2024] [Revised: 02/08/2025] [Accepted: 02/10/2025] [Indexed: 02/17/2025]
Abstract
Protein phosphatase, Mg2+/Mn2+ dependent, 1G (PPM1G) regulates protein function via dephosphorylation. PPM1G participates in the assembly of adherens junctions by dephosphorylating α-catenin. Here, we demonstrated through siRNA transfection and intratesticular injection that PPM1G is critical for maintaining blood-testis barrier function and regulating Sertoli cell apoptosis. We observed that upon knocking down Ppm1g in rat testes, the function of the blood testis barrier was compromised, and the localization of α-catenin and β-catenin became aberrant. Further investigation in rat Sertoli cells revealed that after Ppm1g knockdown, the level of phosphorylated α-catenin increased, and it failed to properly aggregate at the cell membrane; instead, it was mislocalized to the cytoplasm. The actin to which catenin is attached also exhibited a disordered arrangement in the absence of PPM1G. Additionally, through RNA sequencing and bioinformatics analysis, we identified genes associated with Sertoli cell dysfunction induced by Ppm1g knockdown and identified a set of genes involved in regulating intercellular junctions. Subsequent validation revealed that after Ppm1g knockdown, the expression of the junction-related protein JAM2 was reduced, and Sertoli cells underwent apoptosis. Overall, we identified a gene, Ppm1g, which may be involved in maintaining the normal function of the blood-testis barrier and influencing the survival of Sertoli cells by regulating apoptotic pathways.
Collapse
Affiliation(s)
- Xinyao Li
- Department of General Surgery, Gongli Hospital of Shanghai Pudong New Area, Shanghai, 200135, China
| | - Qian Liu
- Institute of Reproductive Medicine, Medical School of Nantong University, Nantong, 226001, Jiangsu, China
| | - Lingling Wang
- Institute of Reproductive Medicine, Medical School of Nantong University, Nantong, 226001, Jiangsu, China
| | - Tiao Bu
- Department of Anesthesiology, Affiliated Hospital of Guangdong Medical University, Guangdong, 57 South Renming Avenue, Xiashan District, Zhanjiang City, 524000, Guandong Province, China
| | - Xiwen Yang
- School of Basic Medical Science, Key Laboratory of Fertility Preservation and Maintenance of Ministry of Education, Key Laboratory of Reproduction and Genetics of Ningxia Hui Autonomous Region, Ningxia Medical University, Yinchuan, 750004, China
| | - Sheng Gao
- Institute of Reproductive Medicine, Medical School of Nantong University, Nantong, 226001, Jiangsu, China
| | - Damin Yun
- Institute of Reproductive Medicine, Medical School of Nantong University, Nantong, 226001, Jiangsu, China.
| | - Fei Sun
- School of Basic Medical Science, Key Laboratory of Fertility Preservation and Maintenance of Ministry of Education, Key Laboratory of Reproduction and Genetics of Ningxia Hui Autonomous Region, Ningxia Medical University, Yinchuan, 750004, China; Department of Urology and Andrology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, 310016, Zhejiang, China.
| |
Collapse
|
3
|
Leng Y, Luan Z, Li Z, Ma Y, Zhou Y, Liu J, Liu S, Tian T, Feng W, Liu Y, Shi Q, Huang C, Zhao X, Wang W, Liu A, Wang T, Ren Q, Liu J, Huang Q, Zhang Y, Yin B, Chen J, Yang L, Zhao S, Bao R, Ji X, Xu Y, Liu L, Zhou J, Chen M, Ma W, Shen L, Zhang T, Zhao H. PPM1F regulates ovarian cancer progression by affecting the dephosphorylation of ITGB1. Clin Transl Oncol 2025; 27:1013-1025. [PMID: 39133386 DOI: 10.1007/s12094-024-03614-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Accepted: 07/09/2024] [Indexed: 08/13/2024]
Abstract
PPM1F has been shown to play diverse biological functions in the progression of multiple tumors. PPM1F controls the T788/T789 phosphorylation switch of ITGB1 and regulates integrin activity. However, the impacts of PPM1F and ITGB1 on ovarian cancer (OV) progression remain unclear. Whether there is such a regulatory relationship between PPM1F and ITGB1 in ovarian cancer has not been studied. Therefore, the purpose of this study is to elucidate the function and the mechanism of PPM1F in ovarian cancer. The expression level and the survival curve of PPM1F were analyzed by databases. Gain of function and loss of function were applied to explore the function of PPM1F in ovarian cancer. A tumor formation assay in nude mice showed that knockdown of PPM1F inhibited tumor formation. We tested the effect of PPM1F on ITGB1 dephosphorylation in ovarian cancer cells by co-immunoprecipitation and western blotting. Loss of function was applied to investigate the function of ITGB1 in ovarian cancer. ITGB1-mut overexpression promotes the progression of ovarian cancer. Rescue assays showed the promoting effect of ITGB1-wt on ovarian cancer is attenuated due to the dephosphorylation of ITGB1-wt by PPM1F. PPM1F and ITGB1 play an oncogene function in ovarian cancer. PPM1F regulates the phosphorylation of ITGB1, which affects the occurrence and development of ovarian cancer.
Collapse
Affiliation(s)
- Yahui Leng
- School of Basic Medicine, Hubei University of Medicine, Shiyan, Hubei, China
- Department of Obstetrics and Gynecology, The Third Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Biomedical Research Institute, Hubei University of Medicine, Shiyan, 442000, Hubei, China
- Department of Clinical OncologyU, Taihe Hospital, Hubei University of Medicine, 30 Renmin South Road, Shiyan, 442000, Hubei, China
| | - Zhenzi Luan
- School of Basic Medicine, Hubei University of Medicine, Shiyan, Hubei, China
- Department of Obstetrics and Gynecology, The Third Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Biomedical Research Institute, Hubei University of Medicine, Shiyan, 442000, Hubei, China
- Department of Clinical OncologyU, Taihe Hospital, Hubei University of Medicine, 30 Renmin South Road, Shiyan, 442000, Hubei, China
| | - Zihang Li
- School of Basic Medicine, Hubei University of Medicine, Shiyan, Hubei, China
- Department of Obstetrics and Gynecology, The Third Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Biomedical Research Institute, Hubei University of Medicine, Shiyan, 442000, Hubei, China
- Department of Clinical OncologyU, Taihe Hospital, Hubei University of Medicine, 30 Renmin South Road, Shiyan, 442000, Hubei, China
| | - Yongqing Ma
- School of Basic Medicine, Hubei University of Medicine, Shiyan, Hubei, China
- Department of Obstetrics and Gynecology, The Third Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Biomedical Research Institute, Hubei University of Medicine, Shiyan, 442000, Hubei, China
- Department of Clinical OncologyU, Taihe Hospital, Hubei University of Medicine, 30 Renmin South Road, Shiyan, 442000, Hubei, China
| | - Yang Zhou
- School of Basic Medicine, Hubei University of Medicine, Shiyan, Hubei, China
- Department of Obstetrics and Gynecology, The Third Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Biomedical Research Institute, Hubei University of Medicine, Shiyan, 442000, Hubei, China
- Department of Clinical OncologyU, Taihe Hospital, Hubei University of Medicine, 30 Renmin South Road, Shiyan, 442000, Hubei, China
| | - Jiaqi Liu
- School of Basic Medicine, Hubei University of Medicine, Shiyan, Hubei, China
- Department of Obstetrics and Gynecology, The Third Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Biomedical Research Institute, Hubei University of Medicine, Shiyan, 442000, Hubei, China
- Department of Clinical OncologyU, Taihe Hospital, Hubei University of Medicine, 30 Renmin South Road, Shiyan, 442000, Hubei, China
| | - Song Liu
- School of Basic Medicine, Hubei University of Medicine, Shiyan, Hubei, China
- Department of Obstetrics and Gynecology, The Third Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Biomedical Research Institute, Hubei University of Medicine, Shiyan, 442000, Hubei, China
- Department of Clinical OncologyU, Taihe Hospital, Hubei University of Medicine, 30 Renmin South Road, Shiyan, 442000, Hubei, China
| | - Tian Tian
- School of Basic Medicine, Hubei University of Medicine, Shiyan, Hubei, China
- Department of Obstetrics and Gynecology, The Third Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Biomedical Research Institute, Hubei University of Medicine, Shiyan, 442000, Hubei, China
- Department of Clinical OncologyU, Taihe Hospital, Hubei University of Medicine, 30 Renmin South Road, Shiyan, 442000, Hubei, China
| | - Wenxiao Feng
- School of Basic Medicine, Hubei University of Medicine, Shiyan, Hubei, China
- Department of Obstetrics and Gynecology, The Third Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Biomedical Research Institute, Hubei University of Medicine, Shiyan, 442000, Hubei, China
- Department of Clinical OncologyU, Taihe Hospital, Hubei University of Medicine, 30 Renmin South Road, Shiyan, 442000, Hubei, China
| | - Yanni Liu
- School of Basic Medicine, Hubei University of Medicine, Shiyan, Hubei, China
- Department of Obstetrics and Gynecology, The Third Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Biomedical Research Institute, Hubei University of Medicine, Shiyan, 442000, Hubei, China
- Department of Clinical OncologyU, Taihe Hospital, Hubei University of Medicine, 30 Renmin South Road, Shiyan, 442000, Hubei, China
| | - Qin Shi
- School of Basic Medicine, Hubei University of Medicine, Shiyan, Hubei, China
- Department of Obstetrics and Gynecology, The Third Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Biomedical Research Institute, Hubei University of Medicine, Shiyan, 442000, Hubei, China
- Department of Clinical OncologyU, Taihe Hospital, Hubei University of Medicine, 30 Renmin South Road, Shiyan, 442000, Hubei, China
| | - Chengyang Huang
- School of Basic Medicine, Hubei University of Medicine, Shiyan, Hubei, China
- Department of Obstetrics and Gynecology, The Third Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Biomedical Research Institute, Hubei University of Medicine, Shiyan, 442000, Hubei, China
- Department of Clinical OncologyU, Taihe Hospital, Hubei University of Medicine, 30 Renmin South Road, Shiyan, 442000, Hubei, China
| | - Xuan Zhao
- The Second Clinical College, Xi'an Medical University, Xi'an, China
| | - Wenlong Wang
- School of Basic Medicine, Hubei University of Medicine, Shiyan, Hubei, China
- Department of Obstetrics and Gynecology, The Third Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Biomedical Research Institute, Hubei University of Medicine, Shiyan, 442000, Hubei, China
- Department of Clinical OncologyU, Taihe Hospital, Hubei University of Medicine, 30 Renmin South Road, Shiyan, 442000, Hubei, China
| | - Ao Liu
- School of Basic Medicine, Hubei University of Medicine, Shiyan, Hubei, China
- Department of Obstetrics and Gynecology, The Third Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Biomedical Research Institute, Hubei University of Medicine, Shiyan, 442000, Hubei, China
- Department of Clinical OncologyU, Taihe Hospital, Hubei University of Medicine, 30 Renmin South Road, Shiyan, 442000, Hubei, China
| | - Tianhang Wang
- School of Basic Medicine, Hubei University of Medicine, Shiyan, Hubei, China
- Department of Obstetrics and Gynecology, The Third Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Biomedical Research Institute, Hubei University of Medicine, Shiyan, 442000, Hubei, China
- Department of Clinical OncologyU, Taihe Hospital, Hubei University of Medicine, 30 Renmin South Road, Shiyan, 442000, Hubei, China
| | - Qiulei Ren
- School of Basic Medicine, Hubei University of Medicine, Shiyan, Hubei, China
- Department of Obstetrics and Gynecology, The Third Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Biomedical Research Institute, Hubei University of Medicine, Shiyan, 442000, Hubei, China
- Department of Clinical OncologyU, Taihe Hospital, Hubei University of Medicine, 30 Renmin South Road, Shiyan, 442000, Hubei, China
| | - Jiakun Liu
- School of Basic Medicine, Hubei University of Medicine, Shiyan, Hubei, China
- Department of Obstetrics and Gynecology, The Third Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Biomedical Research Institute, Hubei University of Medicine, Shiyan, 442000, Hubei, China
- Department of Clinical OncologyU, Taihe Hospital, Hubei University of Medicine, 30 Renmin South Road, Shiyan, 442000, Hubei, China
| | - Qian Huang
- School of Basic Medicine, Hubei University of Medicine, Shiyan, Hubei, China
- Department of Obstetrics and Gynecology, The Third Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Biomedical Research Institute, Hubei University of Medicine, Shiyan, 442000, Hubei, China
- Department of Clinical OncologyU, Taihe Hospital, Hubei University of Medicine, 30 Renmin South Road, Shiyan, 442000, Hubei, China
| | - Yaling Zhang
- School of Basic Medicine, Hubei University of Medicine, Shiyan, Hubei, China
- Department of Obstetrics and Gynecology, The Third Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Biomedical Research Institute, Hubei University of Medicine, Shiyan, 442000, Hubei, China
- Department of Clinical OncologyU, Taihe Hospital, Hubei University of Medicine, 30 Renmin South Road, Shiyan, 442000, Hubei, China
| | - Bin Yin
- School of Basic Medicine, Hubei University of Medicine, Shiyan, Hubei, China
- Department of Obstetrics and Gynecology, The Third Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Biomedical Research Institute, Hubei University of Medicine, Shiyan, 442000, Hubei, China
- Department of Clinical OncologyU, Taihe Hospital, Hubei University of Medicine, 30 Renmin South Road, Shiyan, 442000, Hubei, China
| | - Jialin Chen
- School of Basic Medicine, Hubei University of Medicine, Shiyan, Hubei, China
- Department of Obstetrics and Gynecology, The Third Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Biomedical Research Institute, Hubei University of Medicine, Shiyan, 442000, Hubei, China
- Department of Clinical OncologyU, Taihe Hospital, Hubei University of Medicine, 30 Renmin South Road, Shiyan, 442000, Hubei, China
| | - Liangliang Yang
- School of Basic Medicine, Hubei University of Medicine, Shiyan, Hubei, China
- Department of Obstetrics and Gynecology, The Third Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Biomedical Research Institute, Hubei University of Medicine, Shiyan, 442000, Hubei, China
- Department of Clinical OncologyU, Taihe Hospital, Hubei University of Medicine, 30 Renmin South Road, Shiyan, 442000, Hubei, China
| | - Shiyun Zhao
- School of Basic Medicine, Hubei University of Medicine, Shiyan, Hubei, China
- Department of Obstetrics and Gynecology, The Third Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Biomedical Research Institute, Hubei University of Medicine, Shiyan, 442000, Hubei, China
- Department of Clinical OncologyU, Taihe Hospital, Hubei University of Medicine, 30 Renmin South Road, Shiyan, 442000, Hubei, China
| | - Ruoyi Bao
- School of Basic Medicine, Hubei University of Medicine, Shiyan, Hubei, China
- Department of Obstetrics and Gynecology, The Third Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Biomedical Research Institute, Hubei University of Medicine, Shiyan, 442000, Hubei, China
- Department of Clinical OncologyU, Taihe Hospital, Hubei University of Medicine, 30 Renmin South Road, Shiyan, 442000, Hubei, China
| | - Xingyu Ji
- School of Basic Medicine, Hubei University of Medicine, Shiyan, Hubei, China
- Department of Obstetrics and Gynecology, The Third Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Biomedical Research Institute, Hubei University of Medicine, Shiyan, 442000, Hubei, China
- Department of Clinical OncologyU, Taihe Hospital, Hubei University of Medicine, 30 Renmin South Road, Shiyan, 442000, Hubei, China
| | - Yuewen Xu
- School of Basic Medicine, Hubei University of Medicine, Shiyan, Hubei, China
- Department of Obstetrics and Gynecology, The Third Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Biomedical Research Institute, Hubei University of Medicine, Shiyan, 442000, Hubei, China
- Department of Clinical OncologyU, Taihe Hospital, Hubei University of Medicine, 30 Renmin South Road, Shiyan, 442000, Hubei, China
| | - Liaoyuan Liu
- School of Basic Medicine, Hubei University of Medicine, Shiyan, Hubei, China
- Department of Obstetrics and Gynecology, The Third Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Biomedical Research Institute, Hubei University of Medicine, Shiyan, 442000, Hubei, China
- Department of Clinical OncologyU, Taihe Hospital, Hubei University of Medicine, 30 Renmin South Road, Shiyan, 442000, Hubei, China
| | - Junsuo Zhou
- School of Basic Medicine, Hubei University of Medicine, Shiyan, Hubei, China
- Department of Obstetrics and Gynecology, The Third Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Biomedical Research Institute, Hubei University of Medicine, Shiyan, 442000, Hubei, China
- Department of Clinical OncologyU, Taihe Hospital, Hubei University of Medicine, 30 Renmin South Road, Shiyan, 442000, Hubei, China
| | - Miao Chen
- School of Basic Medicine, Hubei University of Medicine, Shiyan, Hubei, China
- Department of Obstetrics and Gynecology, The Third Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Biomedical Research Institute, Hubei University of Medicine, Shiyan, 442000, Hubei, China
- Department of Clinical OncologyU, Taihe Hospital, Hubei University of Medicine, 30 Renmin South Road, Shiyan, 442000, Hubei, China
| | - Wenhui Ma
- School of Basic Medicine, Hubei University of Medicine, Shiyan, Hubei, China
- Department of Obstetrics and Gynecology, The Third Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Biomedical Research Institute, Hubei University of Medicine, Shiyan, 442000, Hubei, China
- Department of Clinical OncologyU, Taihe Hospital, Hubei University of Medicine, 30 Renmin South Road, Shiyan, 442000, Hubei, China
| | - Li Shen
- School of Basic Medicine, Hubei University of Medicine, Shiyan, Hubei, China.
- Department of Obstetrics and Gynecology, The Third Affiliated Hospital of Chongqing Medical University, Chongqing, China.
- Biomedical Research Institute, Hubei University of Medicine, Shiyan, 442000, Hubei, China.
- Department of Clinical OncologyU, Taihe Hospital, Hubei University of Medicine, 30 Renmin South Road, Shiyan, 442000, Hubei, China.
| | - Te Zhang
- School of Basic Medicine, Hubei University of Medicine, Shiyan, Hubei, China.
- Department of Obstetrics and Gynecology, The Third Affiliated Hospital of Chongqing Medical University, Chongqing, China.
- Biomedical Research Institute, Hubei University of Medicine, Shiyan, 442000, Hubei, China.
- Department of Clinical OncologyU, Taihe Hospital, Hubei University of Medicine, 30 Renmin South Road, Shiyan, 442000, Hubei, China.
| | - Hongyan Zhao
- School of Basic Medicine, Hubei University of Medicine, Shiyan, Hubei, China.
- Department of Obstetrics and Gynecology, The Third Affiliated Hospital of Chongqing Medical University, Chongqing, China.
- Biomedical Research Institute, Hubei University of Medicine, Shiyan, 442000, Hubei, China.
- Department of Clinical OncologyU, Taihe Hospital, Hubei University of Medicine, 30 Renmin South Road, Shiyan, 442000, Hubei, China.
| |
Collapse
|
4
|
Zhu GW, Chen H, Liu SY, Lin PH, Lin CL, Ye JX. PPM1B degradation mediated by TRIM25 ubiquitination modulates cell cycle and promotes gastric cancer growth. Sci Rep 2025; 15:6160. [PMID: 39979355 PMCID: PMC11842632 DOI: 10.1038/s41598-025-89519-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2024] [Accepted: 02/05/2025] [Indexed: 02/22/2025] Open
Abstract
Protein phosphatase PPM1B, a member of the serine/threonine phosphatase family, has been implicated in various human cancers. In this study, our objective was to investigate the role of PPM1B in GC growth and explore the underlying mechanisms. Our findings revealed that PPM1B expression was downregulated in GC tissues, and higher levels of PPM1B expression were associated with improved overall survival in GC patients. Overexpression of PPM1B significantly inhibited cell proliferation, induced G1 phase cell cycle arrest, and suppressed tumor growth. Conversely, knockdown or knockout of PPM1B yielded opposite effects. Mechanistically, we identified that PPM1B exerted its inhibitory role in GC cell growth and cell cycle regulation through the TRIM25/PPM1B/CDK2 signaling pathway. Specifically, we demonstrated that TRIM25 physically interacts with PPM1B, leading to enhanced degradation of PPM1B and subsequent modulation of CDK2 phosphorylation and GC cell growth. PPM1B emerges as a potential prognostic biomarker and therapeutic target in GC. These findings hold clinical significance by offering opportunities to improve diagnosis and treatment strategies for GC patients. Furthermore, this study provides novel insights into the pathogenesis and progression of GC, expanding our understanding of this disease.
Collapse
Affiliation(s)
- Guang-Wei Zhu
- Department of Gastrointestinal Surgery 2 Section, the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350005, China
- Department of Gastrointestinal Surgery, Binhai Campus of the First Affiliated Hospital, National Regional Medical Center, Fujian Medical University, Fuzhou, 350212, China
- Fujian Key Laboratory of Precision Medicine for Cancer, the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350005, China
| | - Hui Chen
- Department of Gastrointestinal Surgery 2 Section, the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350005, China
- Department of Gastrointestinal Surgery, Binhai Campus of the First Affiliated Hospital, National Regional Medical Center, Fujian Medical University, Fuzhou, 350212, China
- Fujian Key Laboratory of Precision Medicine for Cancer, the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350005, China
| | - Song-Yi Liu
- Department of Gastrointestinal Surgery 2 Section, the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350005, China
- Department of Gastrointestinal Surgery, Binhai Campus of the First Affiliated Hospital, National Regional Medical Center, Fujian Medical University, Fuzhou, 350212, China
- Fujian Key Laboratory of Precision Medicine for Cancer, the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350005, China
| | - Peng-Hang Lin
- Department of Gastrointestinal Surgery 2 Section, the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350005, China
- Department of Gastrointestinal Surgery, Binhai Campus of the First Affiliated Hospital, National Regional Medical Center, Fujian Medical University, Fuzhou, 350212, China
- Fujian Key Laboratory of Precision Medicine for Cancer, the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350005, China
- Key Laboratory of Ministry of Education for Gastrointestinal Cancer, Fujian Medical University, Fuzhou, 350000, China
| | - Chun-Lin Lin
- Department of Gastrointestinal Surgery 2 Section, the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350005, China
- Department of Gastrointestinal Surgery, Binhai Campus of the First Affiliated Hospital, National Regional Medical Center, Fujian Medical University, Fuzhou, 350212, China
- Fujian Key Laboratory of Precision Medicine for Cancer, the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350005, China
- Key Laboratory of Ministry of Education for Gastrointestinal Cancer, Fujian Medical University, Fuzhou, 350000, China
| | - Jian-Xin Ye
- Department of Gastrointestinal Surgery 2 Section, the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350005, China.
- Department of Gastrointestinal Surgery, Binhai Campus of the First Affiliated Hospital, National Regional Medical Center, Fujian Medical University, Fuzhou, 350212, China.
- Fujian Key Laboratory of Precision Medicine for Cancer, the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350005, China.
| |
Collapse
|
5
|
Hu L, Shi X, Yuan X, Liu D, Zheng D, Li Y, Shi F, Zhang M, Su S, Zhang CZ. PPM1G-mediated TBL1X mRNA splicing promotes cell migration in hepatocellular carcinoma. Cancer Sci 2025; 116:67-80. [PMID: 39462759 PMCID: PMC11711060 DOI: 10.1111/cas.16372] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2024] [Revised: 09/29/2024] [Accepted: 10/04/2024] [Indexed: 10/29/2024] Open
Abstract
The progression of hepatocellular carcinoma (HCC) is coincident with aberrant splicing of numerous tumor-related genes. Identification of the tumor-specific splice variants that facilitate HCC metastasis may provide a more comprehensive insight into the mechanisms of HCC metastasis. Through RNA sequencing and bioinformatic analyses, PPM1G was identified as a biomarker associated with HCC metastasis. Our data mapped a transcriptome-wide landscape of alternative splicing events modulated by PPM1G in HCC. Notably, we characterized the exon six-skipping transcript of TBL1X as an onco-splice variant regulated by PPM1G. Experimental validation revealed the enrichment of TBL1X-S in response to PPM1G overexpression. Moreover, mRNA stability analyses revealed that PPM1G prolonged the half-life of the TBL1X-S transcript. Both PPM1G and TBL1X-S exhibited metastasis-promoting phenotypes, with PPM1G-driven metastasis in HCC being partially dependent on TBL1X-S. Mechanistically, different TBL1X splice variants showed varying affinities for ZEB1, with TBL1X-S significantly enhancing ZEB1 activation and repressing CDH1 transcription, potentially accelerating the epithelial-mesenchymal transition (EMT) process. In conclusion, our study highlights the biological role of PPM1G and TBL1X-S in tumor metastasis. The PPM1G/TBL1X-S signaling axis presents a new view for investigating liver cancer metastasis mechanisms.
Collapse
Affiliation(s)
- Liling Hu
- MOE Key Laboratory of Tumor Molecular Biology and State Key Laboratory of Bioactive Molecules and Druggability Assessment, Institute of Life and Health Engineering, College of Life Science and TechnologyJinan UniversityGuangzhouChina
| | - Xinyu Shi
- MOE Key Laboratory of Tumor Molecular Biology and State Key Laboratory of Bioactive Molecules and Druggability Assessment, Institute of Life and Health Engineering, College of Life Science and TechnologyJinan UniversityGuangzhouChina
| | - Xiaoyi Yuan
- MOE Key Laboratory of Tumor Molecular Biology and State Key Laboratory of Bioactive Molecules and Druggability Assessment, Institute of Life and Health Engineering, College of Life Science and TechnologyJinan UniversityGuangzhouChina
| | - Danya Liu
- MOE Key Laboratory of Tumor Molecular Biology and State Key Laboratory of Bioactive Molecules and Druggability Assessment, Institute of Life and Health Engineering, College of Life Science and TechnologyJinan UniversityGuangzhouChina
| | - Dandan Zheng
- MOE Key Laboratory of Tumor Molecular Biology and State Key Laboratory of Bioactive Molecules and Druggability Assessment, Institute of Life and Health Engineering, College of Life Science and TechnologyJinan UniversityGuangzhouChina
| | - Yuying Li
- MOE Key Laboratory of Tumor Molecular Biology and State Key Laboratory of Bioactive Molecules and Druggability Assessment, Institute of Life and Health Engineering, College of Life Science and TechnologyJinan UniversityGuangzhouChina
| | - Fujin Shi
- MOE Key Laboratory of Tumor Molecular Biology and State Key Laboratory of Bioactive Molecules and Druggability Assessment, Institute of Life and Health Engineering, College of Life Science and TechnologyJinan UniversityGuangzhouChina
| | - Meifang Zhang
- Department of PathologySun Yat‐sen University Cancer CenterGuangzhouChina
| | - Shu‐Guang Su
- Department of PathologyThe Affiliated Hexian Memorial Hospital of Southern Medical UniversityGuangzhouChina
| | - Chris Zhiyi Zhang
- MOE Key Laboratory of Tumor Molecular Biology and State Key Laboratory of Bioactive Molecules and Druggability Assessment, Institute of Life and Health Engineering, College of Life Science and TechnologyJinan UniversityGuangzhouChina
| |
Collapse
|
6
|
Bo T, Fujii J. Primary Roles of Branched Chain Amino Acids (BCAAs) and Their Metabolism in Physiology and Metabolic Disorders. Molecules 2024; 30:56. [PMID: 39795113 PMCID: PMC11721030 DOI: 10.3390/molecules30010056] [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: 11/19/2024] [Revised: 12/25/2024] [Accepted: 12/25/2024] [Indexed: 01/13/2025] Open
Abstract
Leucine, isoleucine, and valine are collectively known as branched chain amino acids (BCAAs) and are often discussed in the same physiological and pathological situations. The two consecutive initial reactions of BCAA catabolism are catalyzed by the common enzymes referred to as branched chain aminotransferase (BCAT) and branched chain α-keto acid dehydrogenase (BCKDH). BCAT transfers the amino group of BCAAs to 2-ketoglutarate, which results in corresponding branched chain 2-keto acids (BCKAs) and glutamate. BCKDH performs an oxidative decarboxylation of BCKAs, which produces their coenzyme A-conjugates and NADH. BCAT2 in skeletal muscle dominantly catalyzes the transamination of BCAAs. Low BCAT activity in the liver reduces the metabolization of BCAAs, but the abundant presence of BCKDH promotes the metabolism of muscle-derived BCKAs, which leads to the production of glucose and ketone bodies. While mutations in the genes responsible for BCAA catabolism are involved in rare inherited disorders, an aberrant regulation of their enzymatic activities is associated with major metabolic disorders such as diabetes, cardiovascular disease, and cancer. Therefore, an understanding of the regulatory process of metabolic enzymes, as well as the functions of the BCAAs and their metabolites, make a significant contribution to our health.
Collapse
Affiliation(s)
- Tomoki Bo
- Laboratory Animal Center, Institute for Promotion of Medical Science Research, Faculty of Medicine, Yamagata University, Yamagata 990-9585, Japan
| | - Junichi Fujii
- Department of Biochemistry and Molecular Biology, Graduate School of Medical Science, Yamagata University, Yamagata 990-9585, Japan
| |
Collapse
|
7
|
Mao Y, Feng J. Phosphatase activity-based PPM1K: a key player in the regulation of mitochondrial function and its multifaceted impact in diseases. Mol Cell Biochem 2024:10.1007/s11010-024-05188-6. [PMID: 39695034 DOI: 10.1007/s11010-024-05188-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2024] [Accepted: 12/07/2024] [Indexed: 12/20/2024]
Abstract
PPM1K is a significant metal-dependent phosphatase predominantly located in the mitochondrial matrix, where it plays a crucial role in the metabolism of branched-chain amino acids (BCAAs). It is implicated in cellular function and development across various tissues and is associated with diseases like Alzheimer's, cardiomyopathy, and maple syrup urine disease (MSUD). This article reviews PPM1K's impact on mitochondrial function and cellular metabolism, as well as its role in disease progression. The regulation of PPM1K expression and activity by various factors is complex and highlights its therapeutic potential. PPM1K's dysfunction can lead to the accumulation of BCAAs and the excessive opening of the mitochondrial permeability transition pore (MPTP), disrupting physiological metabolism and function. It also regulates the degradation of BCAAs by acting as a specific phosphatase for the E1α subunit of the BCKD complex. Outside the mitochondria, PPM1K suppresses de novo fatty acid synthesis and promotes fatty acid oxidation by dephosphorylating ACL. Furthermore, PPM1K has anti-inflammatory effects and modulates immune cell infiltration in tumor tissues. The expression and activity of PPM1K are influenced by factors such as BCAA concentration, fructose intake, and drug treatments, making it a promising target for therapeutic applications and further basic research.
Collapse
Affiliation(s)
- Yuanling Mao
- Tianjin Medical University General Hospital, 154 Anshan Road, Heping District, Tianjin, 300052, China
| | - Jing Feng
- Tianjin Medical University General Hospital, 154 Anshan Road, Heping District, Tianjin, 300052, China.
| |
Collapse
|
8
|
Etingov I, Pintel DJ. Minute virus of mice NS1 redirects casein kinase 2 specificity to suppress the ATR DNA damage response pathway during infection. J Virol 2024; 98:e0055924. [PMID: 39565137 DOI: 10.1128/jvi.00559-24] [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: 03/25/2024] [Accepted: 10/10/2024] [Indexed: 11/21/2024] Open
Abstract
During infection the autonomous parvovirus minute virus of mice (MVM) generates extensive DNA damage which facilitates virus replication and induces a cellular DNA damage response (DDR) driven by the ataxia telangiectasia mutated (ATM) kinase. Atypically, the ataxia telangiectasia and Rad-3-related (ATR) DDR pathway remains inactive. Upon DNA damage ATR is normally recruited to single-stranded DNA sequences formed at genomic DNA damage sites, and while within a multiprotein complex activates, via phosphorylation, the key DDR regulator checkpoint kinase 1 (Chk1). Inactivation of ATR during MVM infection leads to the accumulation of damaged DNA and enhancement of virus replication. Although ATR is inactivated, we show that during infection, the Chk1 activation pathway downstream of the initial ATR activating events remained functional. Activation of ATR, and consequently of Chk1, requires interaction with TopBP1, which itself is maintained in proximity to ATR by interaction with the phosphorylated S387 residue of Rad9, part of the Rad9-Hus1-Rad1 (911) complex. Both MVM infection and MVM NS1 overexpression inhibited Rad9 S387 phosphorylation and subsequent ATR activation. ATR inactivation during infection was suppressed by expression of Rad9 bearing a phosphomimetic 387 residue, indicating that this site, and the function it served, was the target of NS1 inhibition. NS1 interaction with CK2α and CK2α enzymatic activity was both required to prevent ATR activation, indicating MVM retargeted this kinase's activity during infection. Inhibition of the protein phosphatase 2C (PP2C) prevented Rad9 S387 dephosphorylation and Chk1 inactivation during MVM infection and NS1 overexpression revealing its role in the pathway's suppression. IMPORTANCE Infection by the parvovirus minute virus of mice (MVM) causes significant DNA damage and induces a potent DNA damage response (DDR) which the virus exploits to further its replication. The cell responds to infection with an ATM-regulated DDR; however, atypically, the ATR-regulated DDR pathway is disabled during infection. This prevents Chk1 activation, thus allowing the accumulation of damaged DNA which facilitates virus replication. We describe here how MVM, and specifically the main viral replication protein NS1, inhibits ATR activation. Activation of ATR, and consequently Chk1, requires TopBP1 localization into the activating complex via its interaction with a phosphorylated residue of Rad9. We show that NS1 redirects casein kinase 2 to activate a phosphatase in the PP2C family which causes dephosphorylation of this critical residue, thus inhibiting ATR activation. This work provides mechanistic insight into one of the ways by which parvoviruses modify the host DDR response to facilitate their replication.
Collapse
Affiliation(s)
- Igor Etingov
- Department of Molecular Microbiology and Immunology, University of Missouri-Columbia, School of Medicine, Bond Life Sciences Center, Columbia, Missouri, USA
| | - David J Pintel
- Department of Molecular Microbiology and Immunology, University of Missouri-Columbia, School of Medicine, Bond Life Sciences Center, Columbia, Missouri, USA
| |
Collapse
|
9
|
Wang X, Shang W, Li M, Cao F, Wang D, Wang M, Lu Y, Zhang H, Shen F, Liu J. Identification and characterization of CmPP2C31 playing a positive role in the abiotic stress resistance of Chinese chestnut via an integrated strategy. FRONTIERS IN PLANT SCIENCE 2024; 15:1491269. [PMID: 39735773 PMCID: PMC11671270 DOI: 10.3389/fpls.2024.1491269] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Accepted: 11/25/2024] [Indexed: 12/31/2024]
Abstract
Chinese chestnut (Castanea mollissima Blume) is an important economic forest tree species and mainly cultivated in mountainous areas and wastelands, subjecting it to various abiotic stresses. The protein phosphatase 2C (PP2C) genes contributes largely to stress responses in plants. However, the characteristics and functions of PP2C genes in C. mollissima remain unknown. This study provides comprehensive analyses (including phylogenetic, synteny, RNA-seq, transgenic and yeast one-hybrid methods) revealing the characteristics of CmPP2C gene, which plays an important role in response to abiotic stress. Here, we identified 68 CmPP2Cs in the Chinese chestnut genome, and analyzed their characteristics and phylogenetic relationships. Furthermore, synteny analysis revealed that segmental and tandem duplication drove the expansion of the CmPP2C family to adapt to natural environmental pressures. RNA sequencing and co-expression analyses indicated that four hub CmPP2Cs in two key modules probably play important roles in the resistance to abiotic stress in chestnut. Among them, CmPP2C31 was significantly down-regulated under drought stress. Transgenic experiments via pollen magnetofection revealed that CmPP2C31 could positively and significantly regulate the drought resistance of Chinese chestnut seedlings. Subcellular localization showed that CmPP2C31 was a nuclear protein. Yeast one-hybrid assays suggested that EVM0007407 could regulate CmPP2C31 expression by binding to its promoter, thereby participating in abiotic stress resistance. These findings in our study provided detailed information on the CmPP2C family genes and laid a foundation for further elucidating the molecular mechanism of resistance to abiotic stress chestnut.
Collapse
Affiliation(s)
- Xuan Wang
- Engineering Research Center of Chestnut Industry Technology, Ministry of Education, Hebei Normal University of Science and Technology, Qinhuangdao, Hebei, China
- Hebei Key Laboratory of Horticultural Germplasm Excavation and Innovative Utilization, College of Horticulture Science and Technology, Hebei Normal University of Science and Technology, Changli, Hebei, China
| | - Wenli Shang
- Engineering Research Center of Chestnut Industry Technology, Ministry of Education, Hebei Normal University of Science and Technology, Qinhuangdao, Hebei, China
- Hebei Key Laboratory of Horticultural Germplasm Excavation and Innovative Utilization, College of Horticulture Science and Technology, Hebei Normal University of Science and Technology, Changli, Hebei, China
| | - Mingyuan Li
- Rural Revitalization Research Center, Hebei Normal University of Science and Technology, Qinhuangdao, Hebei, China
| | - Fei Cao
- Engineering Research Center of Chestnut Industry Technology, Ministry of Education, Hebei Normal University of Science and Technology, Qinhuangdao, Hebei, China
- Hebei Key Laboratory of Horticultural Germplasm Excavation and Innovative Utilization, College of Horticulture Science and Technology, Hebei Normal University of Science and Technology, Changli, Hebei, China
| | - Dongsheng Wang
- Engineering Research Center of Chestnut Industry Technology, Ministry of Education, Hebei Normal University of Science and Technology, Qinhuangdao, Hebei, China
| | - Meng Wang
- Engineering Research Center of Chestnut Industry Technology, Ministry of Education, Hebei Normal University of Science and Technology, Qinhuangdao, Hebei, China
| | - Yi Lu
- Engineering Research Center of Chestnut Industry Technology, Ministry of Education, Hebei Normal University of Science and Technology, Qinhuangdao, Hebei, China
| | - Haie Zhang
- Engineering Research Center of Chestnut Industry Technology, Ministry of Education, Hebei Normal University of Science and Technology, Qinhuangdao, Hebei, China
| | - Fei Shen
- Institute of Biotechnology, Beijing Academy of Agriculture and Forestry Sciences, Beijing, China
| | - Jing Liu
- Engineering Research Center of Chestnut Industry Technology, Ministry of Education, Hebei Normal University of Science and Technology, Qinhuangdao, Hebei, China
| |
Collapse
|
10
|
Zhang W, Li Y, Li H, Liu X, Zheng T, Li G, Liu B, Lv T, Wei Z, Xing C, Jia S, Meng A, Wu X. Znf706 regulates germ plasm assembly and primordial germ cell development in zebrafish. J Genet Genomics 2024:S1673-8527(24)00308-4. [PMID: 39571790 DOI: 10.1016/j.jgg.2024.11.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2024] [Revised: 11/14/2024] [Accepted: 11/14/2024] [Indexed: 01/14/2025]
Abstract
The cell fate of primordial germ cells (PGCs) in zebrafish is pre-determined by maternally deposited germ plasm, which is packaged into ribonucleoprotein complex in oocytes and inherited into PGCs-fated cells in embryos. However, the maternal factors regulating the assembly of germ plasm and PGC development remain poorly understood. In this study, we report that the maternal transcription factor Znf706 regulates the assembly of germ plasm factors into a granule-like structure localized perinuclearly in PGCs during migration. Maternal and zygotic mutants of znf706 (MZznf706) exhibit deficient germ plasm scattering at the early embryonic stage, decreased PGC numbers with some mislocation during PGC migration, and a lower female ratio in adulthood. Notably, the implementation of Znf706 CUT&Tag and RNA-seq on immature oocytes uncovers that Znf706 in stage I oocytes may promote transcription of several mitochondrial genes in addition to other functions. Hence, we propose that Znf706 is implicated in germ plasm assembly and PGC development in zebrafish.
Collapse
Affiliation(s)
- Weiying Zhang
- Laboratory of Molecular Developmental Biology, State Key Laboratory of Membrane Biology, Tsinghua-Peking Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Yaqi Li
- Laboratory of Molecular Developmental Biology, State Key Laboratory of Membrane Biology, Tsinghua-Peking Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Han Li
- Laboratory of Molecular Developmental Biology, State Key Laboratory of Membrane Biology, Tsinghua-Peking Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Xin Liu
- Laboratory of Molecular Developmental Biology, State Key Laboratory of Membrane Biology, Tsinghua-Peking Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Tao Zheng
- Laboratory of Molecular Developmental Biology, State Key Laboratory of Membrane Biology, Tsinghua-Peking Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Guangyuan Li
- Laboratory of Molecular Developmental Biology, State Key Laboratory of Membrane Biology, Tsinghua-Peking Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Boqi Liu
- State Key Laboratory of Membrane Biology, Tsinghua University-Peking University Joint Center for Life Sciences, Beijing Frontier Research Center for Biological Structure, School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Tong Lv
- Laboratory of Molecular Developmental Biology, State Key Laboratory of Membrane Biology, Tsinghua-Peking Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Zihang Wei
- Laboratory of Molecular Developmental Biology, State Key Laboratory of Membrane Biology, Tsinghua-Peking Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Cencan Xing
- School of Chemistry and Biological Engineering, University of Science and Technology Beijing, Beijing 100083, China
| | - Shunji Jia
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
| | - Anming Meng
- Laboratory of Molecular Developmental Biology, State Key Laboratory of Membrane Biology, Tsinghua-Peking Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing 100084, China; Guangzhou Laboratory, Guangzhou, Guangdong 510320, China.
| | - Xiaotong Wu
- Laboratory of Molecular Developmental Biology, State Key Laboratory of Membrane Biology, Tsinghua-Peking Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing 100084, China.
| |
Collapse
|
11
|
Patel A, Fréville A, Rey JA, Flynn HR, Koussis K, Skehel MJ, Blackman MJ, Baker DA. Plasmodium falciparum protein phosphatase PP7 is required for early ring-stage development. mBio 2024; 15:e0253924. [PMID: 39387582 PMCID: PMC11559042 DOI: 10.1128/mbio.02539-24] [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: 08/22/2024] [Accepted: 09/13/2024] [Indexed: 10/15/2024] Open
Abstract
We previously reported that the Plasmodium falciparum putative serine/threonine protein phosphatase 7 (PP7) is a high-confidence substrate of the cAMP-dependent protein kinase (PKA). Here we explore the function of PP7 in asexual P. falciparum blood stage parasites. We show that conditional disruption of PP7 leads to a severe growth arrest. We show that PP7 is a calcium-dependent phosphatase that interacts with calmodulin and calcium-dependent protein kinase 1 (CDPK1), consistent with a role in calcium signaling. Notably, PP7 was found to be dispensable for erythrocyte invasion, but was crucial for ring-stage development, with PP7-null parasites arresting shortly following invasion and showing no transition to ameboid forms. Phosphoproteomic analysis revealed that PP7 may regulate certain PKAc substrates. Its interaction with calmodulin and CDPK1 further emphasizes a role in calcium signaling, while its impact on early ring development and PKAc substrate phosphorylation underscores its importance in parasite development. IMPORTANCE Plasmodium falciparum causes malaria and is responsible for more than 600,000 deaths each year. Although effective drugs are available to treat disease, the spread of drug-resistant parasites endangers their future efficacy. It is hoped that a better understanding of the biology of malaria parasites will help us to discover new drugs to tackle the resistance problem. Our work is focused on the cell signaling mechanisms that control the development of the parasite throughout its complex life cycle. All signal transduction pathways are ultimately regulated by reversible protein phosphorylation by protein kinase and protein phosphatase enzymes. In this study, we investigate the function of calcium-dependent protein phosphatase PP7 and show that it is essential for the development of ring-stage parasites following the invasion of human erythrocytes. Our results contribute to the understanding of the erythrocytic stages of the parasite life cycle that cause malaria pathology.
Collapse
Affiliation(s)
- Avnish Patel
- Department of Infection Biology, London School of Hygiene & Tropical Medicine, London, United Kingdom
| | - Aline Fréville
- Department of Infection Biology, London School of Hygiene & Tropical Medicine, London, United Kingdom
| | - Joshua A. Rey
- Department of Infection Biology, London School of Hygiene & Tropical Medicine, London, United Kingdom
| | - Helen R. Flynn
- Proteomics Laboratory, The Francis Crick Institute, London, United Kingdom
| | - Konstantinos Koussis
- Malaria Biochemistry Laboratory, The Francis Crick Institute, London, United Kingdom
| | - Mark J. Skehel
- Proteomics Laboratory, The Francis Crick Institute, London, United Kingdom
| | - Michael J. Blackman
- Department of Infection Biology, London School of Hygiene & Tropical Medicine, London, United Kingdom
- Malaria Biochemistry Laboratory, The Francis Crick Institute, London, United Kingdom
| | - David A. Baker
- Department of Infection Biology, London School of Hygiene & Tropical Medicine, London, United Kingdom
| |
Collapse
|
12
|
Ma M, Yang X, Zhang Y, Wang S, Jin C, Xia W, Chen W, Cai B, Zheng C. PPM1J regulates meat quality feature and glycerophospholipids composition in broiler by modulating protein dephosphorylation. NPJ Sci Food 2024; 8:89. [PMID: 39511232 PMCID: PMC11544016 DOI: 10.1038/s41538-024-00335-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Accepted: 10/30/2024] [Indexed: 11/15/2024] Open
Abstract
The quality of broiler meat affects consumers' purchasing decisions. Numerous studies have shown that phosphorylation of proteins in muscle can affect muscle quality. Here, metabolomics and transcriptomics were used to systematically identify the genetic regulation of differences in meat flavor among different broiler. By constructing the meat flavor-related metabolite-gene networks, we identified that protein phosphatase magnesium/manganese-dependent 1J (PPM1J), which is known to regulate a range of biological processes by modulating reversible protein phosphorylation, was a differentially expressed gene with the highest connectivity to meat flavor-related metabolites. Gain- and loss-of-function analysis revealed that PPM1J induced muscular atrophy, improved meat quality and regulated the composition of glycerophospholipids. More importantly, phosphoproteome and metabolome results found that PPM1J participates in the regulation of meat quality feature and glycerophospholipids composition by catalyzing protein dephosphorylation. Our study provides a basis for further understanding the molecular mechanism of meat quality feature and glycerophospholipids composition in broiler.
Collapse
Affiliation(s)
- Manting Ma
- Institute of Animal Science, Guangdong Academy of Agricultural Sciences, State Key Laboratory of Swine and Poultry Breeding Industry, Key Laboratory of Animal Nutrition and Feed Science in South China, Ministry of Agriculture and Rural Affairs, Guangdong Key Laboratory of Animal Breeding and Nutrition, Guangzhou, China
| | - Xin Yang
- State Key Laboratory of Swine and Poultry Breeding Industry, Lingnan Guangdong Laboratory of Agriculture, College of Animal Science, South China Agricultural University, Guangzhou, China
- Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, and Key Laboratory of Chicken Genetics, Breeding and Reproduction, Ministry of Agriculture, Guangzhou, China
| | - Yanan Zhang
- Institute of Animal Science, Guangdong Academy of Agricultural Sciences, State Key Laboratory of Swine and Poultry Breeding Industry, Key Laboratory of Animal Nutrition and Feed Science in South China, Ministry of Agriculture and Rural Affairs, Guangdong Key Laboratory of Animal Breeding and Nutrition, Guangzhou, China
| | - Shuang Wang
- Institute of Animal Science, Guangdong Academy of Agricultural Sciences, State Key Laboratory of Swine and Poultry Breeding Industry, Key Laboratory of Animal Nutrition and Feed Science in South China, Ministry of Agriculture and Rural Affairs, Guangdong Key Laboratory of Animal Breeding and Nutrition, Guangzhou, China
| | - Chenglong Jin
- Institute of Animal Science, Guangdong Academy of Agricultural Sciences, State Key Laboratory of Swine and Poultry Breeding Industry, Key Laboratory of Animal Nutrition and Feed Science in South China, Ministry of Agriculture and Rural Affairs, Guangdong Key Laboratory of Animal Breeding and Nutrition, Guangzhou, China
| | - Weiguang Xia
- Institute of Animal Science, Guangdong Academy of Agricultural Sciences, State Key Laboratory of Swine and Poultry Breeding Industry, Key Laboratory of Animal Nutrition and Feed Science in South China, Ministry of Agriculture and Rural Affairs, Guangdong Key Laboratory of Animal Breeding and Nutrition, Guangzhou, China
| | - Wei Chen
- Institute of Animal Science, Guangdong Academy of Agricultural Sciences, State Key Laboratory of Swine and Poultry Breeding Industry, Key Laboratory of Animal Nutrition and Feed Science in South China, Ministry of Agriculture and Rural Affairs, Guangdong Key Laboratory of Animal Breeding and Nutrition, Guangzhou, China
| | - Bolin Cai
- State Key Laboratory of Swine and Poultry Breeding Industry, Lingnan Guangdong Laboratory of Agriculture, College of Animal Science, South China Agricultural University, Guangzhou, China.
- Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, and Key Laboratory of Chicken Genetics, Breeding and Reproduction, Ministry of Agriculture, Guangzhou, China.
| | - Chuntian Zheng
- Institute of Animal Science, Guangdong Academy of Agricultural Sciences, State Key Laboratory of Swine and Poultry Breeding Industry, Key Laboratory of Animal Nutrition and Feed Science in South China, Ministry of Agriculture and Rural Affairs, Guangdong Key Laboratory of Animal Breeding and Nutrition, Guangzhou, China.
| |
Collapse
|
13
|
Song M, Xu M, Zhang Q, Fan T, Xu J, Hang C, Cheng C, Ou X, Gong C, Lu Q. PPM1G promotes autophagy and progression of pancreatic cancer via upregulating HMGB1. Cell Signal 2024; 123:111342. [PMID: 39121976 DOI: 10.1016/j.cellsig.2024.111342] [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: 04/21/2024] [Revised: 07/24/2024] [Accepted: 08/06/2024] [Indexed: 08/12/2024]
Abstract
Pancreatic cancer remains one of the most aggressive and lethal malignancies worldwide, with a dismal 5-year relative survival rates of only 12%. Therefore, it is urgent to discover the key molecular markers to improve the therapeutic outcomes in pancreatic cancer. Herein, we first demonstrated that PPM1G is upregulated in pancreatic cancer and that PPM1G depletion decreases pancreatic cancer cell growth in vitro and in vivo. High PPM1G expression was linked to short overall survival of pancreatic cancer patients, which was further validated in the TCGA database. Moreover, by detecting Beclin 1, LC3-II, and SQSTM1/p62 expressions and observing autolysosome under transmission electron microscope, we discovered that PPM1G is a novel positive regulator of macroautophagy/autophagy. Furthermore, by using immunoprecipitation-mass spectrometry (IP-MS) analysis and following systemic molecular biology experiment, we demonstrated PPM1G promotes the autophagy and proliferation of pancreatic cancer by directly upregulating HMGB1. Additionally, patients with both high PPM1G and high HMGB1 exhibited poorer prognosis in our cohort. This study preliminarily investigated the possibility of PPM1G as a potential therapeutic target and prognostic biomarker in pancreatic cancer patients.
Collapse
Affiliation(s)
- Mingyang Song
- Department of Gastroenterology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450000, China; Department of Gastroenterology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing 210009, China
| | - Min Xu
- Department of Human Anatomy, School of Medicine, Southeast University, Nanjing 210009, China
| | - Qi Zhang
- Department of Gastroenterology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing 210009, China
| | - Tingyu Fan
- Department of Gastroenterology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing 210009, China
| | - Jiajia Xu
- Department of Clinical Pathology, Zhongda Hospital, Southeast University, Nanjing 210009, China
| | - Cheng Hang
- Department of Gastroenterology, Taicang Affiliated Hospital of Soochow University, The First People's Hospital of Taicang, Jiangsu 215400, China
| | - Cuie Cheng
- Department of Gastroenterology, Affiliated Changshu Hospital of Nantong University, Suzhou 215500, China
| | - Xilong Ou
- Department of Gastroenterology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing 210009, China.
| | - Chen Gong
- Department of Gastroenterology, Taicang Affiliated Hospital of Soochow University, The First People's Hospital of Taicang, Jiangsu 215400, China.
| | - Qin Lu
- Department of Gastroenterology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing 210009, China.
| |
Collapse
|
14
|
Fabra MA, Paredes-Fuentes AJ, Torralba Carnerero M, Moreno Férnandez de Ayala DJ, Arroyo Luque A, Sánchez Cuesta A, Staiano C, Sanchez-Pintos P, Luz Couce M, Tomás M, Marco-Hernández AV, Orellana C, Martínez F, Roselló M, Caro A, Oltra Soler JS, Monfort S, Sánchez A, Rausell D, Vitoria I, Del Toro M, Garcia-Cazorla A, Julia-Palacios NA, Jou C, Yubero D, López LC, Hernández Camacho JD, López Lluch G, Ballesteros Simarro M, Rodríguez Aguilera JC, Calvo GB, Cascajo Almenara MV, Artuch R, Santos-Ocaña C. New variants expand the neurological phenotype of COQ7 deficiency. J Inherit Metab Dis 2024; 47:1047-1068. [PMID: 38973597 DOI: 10.1002/jimd.12776] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2024] [Revised: 06/05/2024] [Accepted: 06/24/2024] [Indexed: 07/09/2024]
Abstract
The protein encoded by COQ7 is required for CoQ10 synthesis in humans, hydroxylating 3-demethoxyubiquinol (DMQ10) in the second to last steps of the pathway. COQ7 mutations lead to a primary CoQ10 deficiency syndrome associated with a pleiotropic neurological disorder. This study shows the clinical, physiological, and molecular characterization of four new cases of CoQ10 primary deficiency caused by five mutations in COQ7, three of which have not yet been described, inducing mitochondrial dysfunction in all patients. However, the specific combination of the identified variants in each patient generated precise pathophysiological and molecular alterations in fibroblasts, which would explain the differential in vitro response to supplementation therapy. Our results suggest that COQ7 dysfunction could be caused by specific structural changes that affect the interaction with COQ9 required for the DMQ10 presentation to COQ7, the substrate access to the active site, and the maintenance of the active site structure. Remarkably, patients' fibroblasts share transcriptional remodeling, supporting a modification of energy metabolism towards glycolysis, which could be an adaptive mechanism against CoQ10 deficiency. However, transcriptional analysis of mitochondria-associated pathways showed distinct and dramatic differences between patient fibroblasts, which correlated with the extent of pathophysiological and neurological alterations observed in the probands. Overall, this study suggests that the combination of precise genetic diagnostics and the availability of new structural models of human proteins could help explain the origin of phenotypic pleiotropy observed in some genetic diseases and the different responses to available therapies.
Collapse
Affiliation(s)
- María Alcázar Fabra
- Departamento de Fisiología, Anatomía y Biología Celular, CABD, Universidad Pablo de Olavide, Sevilla, Spain
- Unidad U729 CIBERER, Instituto de Salud Carlos III, Sevilla, Spain
| | - Abraham J Paredes-Fuentes
- Hospital San Joan de Deu, Barcelona, Spain
- Unidad U703 CIBERER, Instituto de Salud Carlos III, Barcelona, Spain
| | - Manuel Torralba Carnerero
- Departamento de Fisiología, Anatomía y Biología Celular, CABD, Universidad Pablo de Olavide, Sevilla, Spain
| | - Daniel J Moreno Férnandez de Ayala
- Departamento de Fisiología, Anatomía y Biología Celular, CABD, Universidad Pablo de Olavide, Sevilla, Spain
- Unidad U729 CIBERER, Instituto de Salud Carlos III, Sevilla, Spain
| | - Antonio Arroyo Luque
- Departamento de Fisiología, Anatomía y Biología Celular, CABD, Universidad Pablo de Olavide, Sevilla, Spain
- Unidad U729 CIBERER, Instituto de Salud Carlos III, Sevilla, Spain
| | - Ana Sánchez Cuesta
- Departamento de Fisiología, Anatomía y Biología Celular, CABD, Universidad Pablo de Olavide, Sevilla, Spain
- Unidad U729 CIBERER, Instituto de Salud Carlos III, Sevilla, Spain
| | - Carmine Staiano
- Departamento de Fisiología, Anatomía y Biología Celular, CABD, Universidad Pablo de Olavide, Sevilla, Spain
- Unidad U729 CIBERER, Instituto de Salud Carlos III, Sevilla, Spain
| | - Paula Sanchez-Pintos
- Unidad de Diagnóstico y Tratamiento de Enfermedades Metabólicas Congénitas, Hospital de Santiago de Compostela, IDIS, CIBERER, MetabERN, Santiago de Compostela, Spain
- GCV14/ER/5 CIBERER, Instituto de Salud Carlos III, Santiago de Compostela, Spain
| | - María Luz Couce
- Unidad de Diagnóstico y Tratamiento de Enfermedades Metabólicas Congénitas, Hospital de Santiago de Compostela, IDIS, CIBERER, MetabERN, Santiago de Compostela, Spain
- GCV14/ER/5 CIBERER, Instituto de Salud Carlos III, Santiago de Compostela, Spain
| | - Miguel Tomás
- Hospital Universitari i Politècnic La Fe, Servicio de Neuropediatría, Valencia, Spain
| | | | - Carmen Orellana
- Hospital Universitari i Politècnic La Fe, Unidad de Genética, Valencia, Spain
| | - Francisco Martínez
- Hospital Universitari i Politècnic La Fe, Unidad de Genética, Valencia, Spain
| | - Mónica Roselló
- Hospital Universitari i Politècnic La Fe, Unidad de Genética, Valencia, Spain
| | - Alfonso Caro
- Hospital Universitari i Politècnic La Fe, Unidad de Genética, Valencia, Spain
| | | | - Sandra Monfort
- Hospital Universitari i Politècnic La Fe, Unidad de Genética, Valencia, Spain
| | - Alejandro Sánchez
- Hospital Universitari i Politècnic La Fe, Unidad de Genética, Valencia, Spain
| | - Dolores Rausell
- Hospital Universitari i Politècnic La Fe, Servicio de Análisis Clínicos, Valencia, Spain
| | - Isidro Vitoria
- Hospital Universitari i Politècnic La Fe, Unidad de Metabolopatías, Valencia, Spain
| | - Mireia Del Toro
- Pediatric Neurology Unit, Hospital Universitari Vall d'Hebron, CIBERER, MetabERN, Barcelona, Spain
- Instituto de Salud Carlos III, Barcelona, Spain
| | - Angels Garcia-Cazorla
- Hospital San Joan de Deu, Barcelona, Spain
- Unidad U703 CIBERER, Instituto de Salud Carlos III, Barcelona, Spain
| | - Natalia A Julia-Palacios
- Hospital San Joan de Deu, Barcelona, Spain
- Unidad U703 CIBERER, Instituto de Salud Carlos III, Barcelona, Spain
| | - Cristina Jou
- Hospital San Joan de Deu, Barcelona, Spain
- Unidad U703 CIBERER, Instituto de Salud Carlos III, Barcelona, Spain
| | - Delia Yubero
- Hospital San Joan de Deu, Barcelona, Spain
- Unidad U703 CIBERER, Instituto de Salud Carlos III, Barcelona, Spain
| | - Luis Carlos López
- Departamento de Fisiología, Facultad de Medicina, Centro de Investigación Biomédica, Instituto de Biotecnología, Universidad de Granada, Granada, Spain
| | - Juan Diego Hernández Camacho
- Departamento de Fisiología, Anatomía y Biología Celular, CABD, Universidad Pablo de Olavide, Sevilla, Spain
- Unidad U729 CIBERER, Instituto de Salud Carlos III, Sevilla, Spain
| | - Guillermo López Lluch
- Departamento de Fisiología, Anatomía y Biología Celular, CABD, Universidad Pablo de Olavide, Sevilla, Spain
- Unidad U729 CIBERER, Instituto de Salud Carlos III, Sevilla, Spain
| | - Manuel Ballesteros Simarro
- Departamento de Fisiología, Anatomía y Biología Celular, CABD, Universidad Pablo de Olavide, Sevilla, Spain
- Unidad U729 CIBERER, Instituto de Salud Carlos III, Sevilla, Spain
| | - Juan Carlos Rodríguez Aguilera
- Departamento de Fisiología, Anatomía y Biología Celular, CABD, Universidad Pablo de Olavide, Sevilla, Spain
- Unidad U729 CIBERER, Instituto de Salud Carlos III, Sevilla, Spain
| | - Gloria Brea Calvo
- Departamento de Fisiología, Anatomía y Biología Celular, CABD, Universidad Pablo de Olavide, Sevilla, Spain
- Unidad U729 CIBERER, Instituto de Salud Carlos III, Sevilla, Spain
| | - María Victoria Cascajo Almenara
- Departamento de Fisiología, Anatomía y Biología Celular, CABD, Universidad Pablo de Olavide, Sevilla, Spain
- Unidad U729 CIBERER, Instituto de Salud Carlos III, Sevilla, Spain
| | - Rafael Artuch
- Hospital San Joan de Deu, Barcelona, Spain
- Unidad U703 CIBERER, Instituto de Salud Carlos III, Barcelona, Spain
| | - Carlos Santos-Ocaña
- Departamento de Fisiología, Anatomía y Biología Celular, CABD, Universidad Pablo de Olavide, Sevilla, Spain
- Unidad U729 CIBERER, Instituto de Salud Carlos III, Sevilla, Spain
| |
Collapse
|
15
|
Zhao L, Wei X, Chen F, Yuan L, Chen B, Li R. N6-methyladenosine RNA methyltransferase CpMTA1 mediates CpAphA mRNA stability through a YTHDF1-dependent m6A modification in the chestnut blight fungus. PLoS Pathog 2024; 20:e1012476. [PMID: 39159278 PMCID: PMC11361730 DOI: 10.1371/journal.ppat.1012476] [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: 02/01/2024] [Revised: 08/29/2024] [Accepted: 08/04/2024] [Indexed: 08/21/2024] Open
Abstract
In eukaryotic cells, N6-methyladenosine (m6A) is the most prevalent RNA epigenetic modification that plays crucial roles in multiple biological processes. Nevertheless, the functions and regulatory mechanisms of m6A in phytopathogenic fungi are poorly understood. Here, we showed that CpMTA1, an m6A methyltransferase in Cryphonectria parasitica, plays a crucial role in fungal phenotypic traits, virulence, and stress tolerance. Furthermore, the acid phosphatase gene CpAphA was implicated to be a target of CpMTA1 by integrated analysis of m6A-seq and RNA-seq, as in vivo RIP assay data confirmed that CpMTA1 directly interacts with CpAphA mRNA. Deletion of CpMTA1 drastically lowered the m6A level of CpAphA and reduced its mRNA expression. Moreover, we found that an m6A reader protein CpYTHDF1 recognizes CpAphA mRNA and increases its stability. Typically, the levels of CpAphA mRNA and protein exhibited a positive correlation with CpMTA1 and CpYTHDF1. Importantly, site-specific mutagenesis demonstrated that the m6A sites, A1306 and A1341, of CpAphA mRNA are important for fungal phenotypic traits and virulence in C. parasitica. Together, our findings demonstrate the essential role of the m6A methyltransferase CpMTA1 in C. parasitica, thereby advancing our understanding of fungal gene regulation through m6A modification.
Collapse
Affiliation(s)
- Lijiu Zhao
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-bioresources, Guangxi Research Center for Microbial and Enzyme Engineering Technology, College of Life Science and Technology, Guangxi University, Nanning, China
| | - Xiangyu Wei
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-bioresources, Guangxi Research Center for Microbial and Enzyme Engineering Technology, College of Life Science and Technology, Guangxi University, Nanning, China
| | - Fengyue Chen
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-bioresources, Guangxi Research Center for Microbial and Enzyme Engineering Technology, College of Life Science and Technology, Guangxi University, Nanning, China
| | - Luying Yuan
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-bioresources, Guangxi Research Center for Microbial and Enzyme Engineering Technology, College of Life Science and Technology, Guangxi University, Nanning, China
| | - Baoshan Chen
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-bioresources, Guangxi Research Center for Microbial and Enzyme Engineering Technology, College of Life Science and Technology, Guangxi University, Nanning, China
- Guangxi Key Laboratory of Sugarcane Biology, College of Agriculture, Guangxi University, Nanning, China
| | - Ru Li
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-bioresources, Guangxi Research Center for Microbial and Enzyme Engineering Technology, College of Life Science and Technology, Guangxi University, Nanning, China
- Guangxi Key Laboratory of Sugarcane Biology, College of Agriculture, Guangxi University, Nanning, China
| |
Collapse
|
16
|
Kumar JP, Kosek D, Durell SR, Miller Jenkins LM, Debnath S, Coussens NP, Hall MD, Appella DH, Dyda F, Mazur SJ, Appella E. Crystal structure and mechanistic studies of the PPM1D serine/threonine phosphatase catalytic domain. J Biol Chem 2024; 300:107561. [PMID: 39002674 PMCID: PMC11342775 DOI: 10.1016/j.jbc.2024.107561] [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: 11/29/2023] [Revised: 06/28/2024] [Accepted: 07/03/2024] [Indexed: 07/15/2024] Open
Abstract
Protein phosphatase 1D (PPM1D, Wip1) is induced by the tumor suppressor p53 during DNA damage response signaling and acts as an oncoprotein in several human cancers. Although PPM1D is a potential therapeutic target, insights into its atomic structure were challenging due to flexible regions unique to this family member. Here, we report the first crystal structure of the PPM1D catalytic domain to 1.8 Å resolution. The structure reveals the active site with two Mg2+ ions bound, similar to other structures. The flap subdomain and B-loop, which are crucial for substrate recognition and catalysis, were also resolved, with the flap forming two short helices and three short β-strands that are followed by an irregular loop. Unexpectedly, a nitrogen-oxygen-sulfur bridge was identified in the catalytic domain. Molecular dynamics simulations and kinetic studies provided further mechanistic insights into the regulation of PPM1D catalytic activity. In particular, the kinetic experiments demonstrated a magnesium concentration-dependent lag in PPM1D attaining steady-state velocity, a feature of hysteretic enzymes that show slow transitions compared with catalytic turnover. All combined, these results advance the understanding of PPM1D function and will support the development of PPM1D-targeted therapeutics.
Collapse
Affiliation(s)
- Jay Prakash Kumar
- Laboratory of Cell Biology, NCI, National Institutes of Health, Bethesda, Maryland, United States
| | - Dalibor Kosek
- Laboratory of Molecular Biology, NIDDK, National Institutes of Health, Bethesda, Maryland, United States
| | - Stewart R Durell
- Laboratory of Cell Biology, NCI, National Institutes of Health, Bethesda, Maryland, United States
| | - Lisa M Miller Jenkins
- Laboratory of Cell Biology, NCI, National Institutes of Health, Bethesda, Maryland, United States
| | - Subrata Debnath
- Laboratory of Cell Biology, NCI, National Institutes of Health, Bethesda, Maryland, United States
| | - Nathan P Coussens
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, Maryland, United States
| | - Matthew D Hall
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, Maryland, United States
| | - Daniel H Appella
- Laboratory of Bioorganic Chemistry, NIDDK, National Institutes of Health, Bethesda, Maryland, United States
| | - Fred Dyda
- Laboratory of Molecular Biology, NIDDK, National Institutes of Health, Bethesda, Maryland, United States
| | - Sharlyn J Mazur
- Laboratory of Cell Biology, NCI, National Institutes of Health, Bethesda, Maryland, United States
| | - Ettore Appella
- Laboratory of Cell Biology, NCI, National Institutes of Health, Bethesda, Maryland, United States.
| |
Collapse
|
17
|
Tang Q, Ojiro R, Ozawa S, Zou X, Nakahara J, Nakao T, Koyanagi M, Jin M, Yoshida T, Shibutani M. DNA methylation-altered genes in the rat hippocampal neurogenic niche after continuous exposure to amorphous curcumin. J Chem Neuroanat 2024; 137:102414. [PMID: 38490283 DOI: 10.1016/j.jchemneu.2024.102414] [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: 11/05/2023] [Revised: 03/09/2024] [Accepted: 03/12/2024] [Indexed: 03/17/2024]
Abstract
Rat offspring who are exposed to an amorphous formula of curcumin (CUR) from the embryonic stage have anti-anxiety-like behaviors, enhanced fear extinction learning, and increased synaptic plasticity in the hippocampal dentate gyrus (DG). In the present study, we investigated the links between genes with altered methylation status in the neurogenic niche and enhanced neural functions after CUR exposure. We conducted methylation and RNA sequencing analyses of the DG of CUR-exposed rat offspring on day 77 after delivery. Methylation status and transcript levels of candidate genes were validated using methylation-sensitive high-resolution melting and real-time reverse-transcription PCR, respectively. In the CUR group, we confirmed the hypermethylation and downregulation of Gpr150, Mmp23, Rprml, and Pcdh8 as well as the hypomethylation and upregulation of Ppm1j, Fam222a, and Opn3. Immunohistochemically, reprimo-like+ hilar cells and protocadherin-8+ granule cells were decreased and opsin-3+ hilar cells were increased by CUR exposure. Both reprimo-like and opsin-3 were partially expressed on subpopulations of glutamic acid decarboxylase 67+ γ-aminobutyric acid-ergic interneurons. Furthermore, the transcript levels of genes involved in protocadherin-8-mediated N-cadherin endocytosis were altered with CUR exposure; this was accompanied by Ctnnb1 and Syp upregulation and Mapk14, Map2k3, and Grip1 downregulation, suggesting that CUR-induced enhanced synaptic plasticity is associated with cell adhesion. Together, our results indicate that functionally different genes have altered methylation and expression in different neuronal populations of the hippocampal neurogenic niche, thus enhancing synaptic plasticity after CUR exposure.
Collapse
Affiliation(s)
- Qian Tang
- Laboratory of Veterinary Pathology, Tokyo University of Agriculture and Technology, 3-5-8 Saiwai-cho, Fuchu-shi, Tokyo 183-8509, Japan; Cooperative Division of Veterinary Sciences, Graduate School of Agriculture, Tokyo University of Agriculture and Technology, 3-5-8 Saiwai-cho, Fuchu-shi, Tokyo 183-8509, Japan
| | - Ryota Ojiro
- Laboratory of Veterinary Pathology, Tokyo University of Agriculture and Technology, 3-5-8 Saiwai-cho, Fuchu-shi, Tokyo 183-8509, Japan; Cooperative Division of Veterinary Sciences, Graduate School of Agriculture, Tokyo University of Agriculture and Technology, 3-5-8 Saiwai-cho, Fuchu-shi, Tokyo 183-8509, Japan
| | - Shunsuke Ozawa
- Laboratory of Veterinary Pathology, Tokyo University of Agriculture and Technology, 3-5-8 Saiwai-cho, Fuchu-shi, Tokyo 183-8509, Japan; Cooperative Division of Veterinary Sciences, Graduate School of Agriculture, Tokyo University of Agriculture and Technology, 3-5-8 Saiwai-cho, Fuchu-shi, Tokyo 183-8509, Japan
| | - Xinyu Zou
- Laboratory of Veterinary Pathology, Tokyo University of Agriculture and Technology, 3-5-8 Saiwai-cho, Fuchu-shi, Tokyo 183-8509, Japan; Cooperative Division of Veterinary Sciences, Graduate School of Agriculture, Tokyo University of Agriculture and Technology, 3-5-8 Saiwai-cho, Fuchu-shi, Tokyo 183-8509, Japan
| | - Junta Nakahara
- Laboratory of Veterinary Pathology, Tokyo University of Agriculture and Technology, 3-5-8 Saiwai-cho, Fuchu-shi, Tokyo 183-8509, Japan
| | - Tomohiro Nakao
- Emulsion Laboratory, San-Ei Gen F.F.I., Inc., 1-1-11 Sanwa-cho, Toyonaka-shi, Osaka 561-8588, Japan
| | - Mihoko Koyanagi
- Global Scientific and Regulatory Affairs, San-Ei Gen F.F.I., Inc., 1-1-11 Sanwa-cho, Toyonaka-shi, Osaka 561-8588, Japan
| | - Meilan Jin
- Laboratory of Veterinary Pathology, College of Veterinary Medicine, Southwest University, No. 2 Tiansheng Road, BeiBei District, Chongqing 400715, PR China
| | - Toshinori Yoshida
- Laboratory of Veterinary Pathology, Tokyo University of Agriculture and Technology, 3-5-8 Saiwai-cho, Fuchu-shi, Tokyo 183-8509, Japan; Cooperative Division of Veterinary Sciences, Graduate School of Agriculture, Tokyo University of Agriculture and Technology, 3-5-8 Saiwai-cho, Fuchu-shi, Tokyo 183-8509, Japan
| | - Makoto Shibutani
- Laboratory of Veterinary Pathology, Tokyo University of Agriculture and Technology, 3-5-8 Saiwai-cho, Fuchu-shi, Tokyo 183-8509, Japan; Cooperative Division of Veterinary Sciences, Graduate School of Agriculture, Tokyo University of Agriculture and Technology, 3-5-8 Saiwai-cho, Fuchu-shi, Tokyo 183-8509, Japan.
| |
Collapse
|
18
|
Zheng L, Zhao S, Li Y, Xu J, Yan W, Guo B, Xu J, Jiang L, Zhang Y, Wei H, Jiang Q. Engineered MgO nanoparticles for cartilage-bone synergistic therapy. SCIENCE ADVANCES 2024; 10:eadk6084. [PMID: 38457498 PMCID: PMC10923500 DOI: 10.1126/sciadv.adk6084] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Accepted: 02/02/2024] [Indexed: 03/10/2024]
Abstract
The emerging therapeutic strategies for osteoarthritis (OA) are shifting toward comprehensive approaches that target periarticular tissues, involving both cartilage and subchondral bone. This shift drives the development of single-component therapeutics capable of acting on multiple tissues and cells. Magnesium, an element essential for maintaining skeletal health, shows promise in treating OA. However, the precise effects of magnesium on cartilage and subchondral bone are not yet clear. Here, we investigated the therapeutic effect of Mg2+ on OA, unveiling its protective effects on both cartilage and bone at the cellular and animal levels. The beneficial effect on the cartilage-bone interaction is primarily mediated by the PI3K/AKT pathway. In addition, we developed poly(lactic-co-glycolic acid) (PLGA) microspheres loaded with nano-magnesium oxide modified with stearic acid (SA), MgO&SA@PLGA, for intra-articular injection. These microspheres demonstrated remarkable efficacy in alleviating OA in rat models, highlighting their translational potential in clinical applications.
Collapse
Affiliation(s)
- Liming Zheng
- Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, 321 Zhongshan Road; State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University; Branch of National Clinical Research Center for Orthopedics, Sports Medicine and Rehabilitation; Institute of Medical 3D Printing, Nanjing University; Jiangsu Engineering Research Center for 3D Bioprinting, Nanjing 210008, Jiangsu, PR China
- Department of Biomedical Engineering, College of Engineering and Applied Sciences, Nanjing National Laboratory of Microstructures, Jiangsu Key Laboratory of Artificial Functional Materials, Nanjing University; State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Chemistry and Biomedicine Innovation Center (ChemBIC), Nanjing University, Nanjing 210023, Jiangsu, PR China
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine; Orthopedics Research Institute of Zhejiang University; Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province; Clinical Research Center of Motor System Disease of Zhejiang Province, Hangzhou, Zhejiang, 310000, PR China
- School of Life Science and Technology, ShanghaiTech University, Shanghai, 201210, PR China
| | - Sheng Zhao
- Department of Biomedical Engineering, College of Engineering and Applied Sciences, Nanjing National Laboratory of Microstructures, Jiangsu Key Laboratory of Artificial Functional Materials, Nanjing University; State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Chemistry and Biomedicine Innovation Center (ChemBIC), Nanjing University, Nanjing 210023, Jiangsu, PR China
| | - Yixuan Li
- Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, 321 Zhongshan Road; State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University; Branch of National Clinical Research Center for Orthopedics, Sports Medicine and Rehabilitation; Institute of Medical 3D Printing, Nanjing University; Jiangsu Engineering Research Center for 3D Bioprinting, Nanjing 210008, Jiangsu, PR China
| | - Jiankun Xu
- Musculoskeletal Research Laboratory, Department of Orthopedics and Traumatology, The Chinese University of Hong Kong, Hong Kong 999077, PR China
| | - Wenjin Yan
- Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, 321 Zhongshan Road; State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University; Branch of National Clinical Research Center for Orthopedics, Sports Medicine and Rehabilitation; Institute of Medical 3D Printing, Nanjing University; Jiangsu Engineering Research Center for 3D Bioprinting, Nanjing 210008, Jiangsu, PR China
| | - Baosheng Guo
- Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, 321 Zhongshan Road; State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University; Branch of National Clinical Research Center for Orthopedics, Sports Medicine and Rehabilitation; Institute of Medical 3D Printing, Nanjing University; Jiangsu Engineering Research Center for 3D Bioprinting, Nanjing 210008, Jiangsu, PR China
| | - Jianbin Xu
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine; Orthopedics Research Institute of Zhejiang University; Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province; Clinical Research Center of Motor System Disease of Zhejiang Province, Hangzhou, Zhejiang, 310000, PR China
| | - Lifeng Jiang
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine; Orthopedics Research Institute of Zhejiang University; Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province; Clinical Research Center of Motor System Disease of Zhejiang Province, Hangzhou, Zhejiang, 310000, PR China
| | - Yifeng Zhang
- Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, 321 Zhongshan Road; State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University; Branch of National Clinical Research Center for Orthopedics, Sports Medicine and Rehabilitation; Institute of Medical 3D Printing, Nanjing University; Jiangsu Engineering Research Center for 3D Bioprinting, Nanjing 210008, Jiangsu, PR China
- School of Life Science and Technology, ShanghaiTech University, Shanghai, 201210, PR China
| | - Hui Wei
- Department of Biomedical Engineering, College of Engineering and Applied Sciences, Nanjing National Laboratory of Microstructures, Jiangsu Key Laboratory of Artificial Functional Materials, Nanjing University; State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Chemistry and Biomedicine Innovation Center (ChemBIC), Nanjing University, Nanjing 210023, Jiangsu, PR China
| | - Qing Jiang
- Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, 321 Zhongshan Road; State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University; Branch of National Clinical Research Center for Orthopedics, Sports Medicine and Rehabilitation; Institute of Medical 3D Printing, Nanjing University; Jiangsu Engineering Research Center for 3D Bioprinting, Nanjing 210008, Jiangsu, PR China
| |
Collapse
|
19
|
Asanoma K, Yagi H, Onoyama I, Cui L, Hori E, Kawakami M, Maenohara S, Hachisuga K, Tomonobe H, Kodama K, Yasunaga M, Ohgami T, Okugawa K, Yahata H, Kitao H, Kato K. The BHLHE40‒PPM1F‒AMPK pathway regulates energy metabolism and is associated with the aggressiveness of endometrial cancer. J Biol Chem 2024; 300:105695. [PMID: 38301894 PMCID: PMC10904277 DOI: 10.1016/j.jbc.2024.105695] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2023] [Revised: 01/03/2024] [Accepted: 01/17/2024] [Indexed: 02/03/2024] Open
Abstract
BHLHE40 is a basic helix-loop-helix transcription factor that is involved in multiple cell activities including differentiation, cell cycle, and epithelial-to-mesenchymal transition. While there is growing evidence to support the functions of BHLHE40 in energy metabolism, little is known about the mechanism. In this study, we found that BHLHE40 expression was downregulated in cases of endometrial cancer of higher grade and advanced disease. Knockdown of BHLHE40 in endometrial cancer cells resulted in suppressed oxygen consumption and enhanced extracellular acidification. Suppressed pyruvate dehydrogenase (PDH) activity and enhanced lactated dehydrogenase (LDH) activity were observed in the knockdown cells. Knockdown of BHLHE40 also led to dephosphorylation of AMPKα Thr172 and enhanced phosphorylation of pyruvate dehydrogenase E1 subunit alpha 1 (PDHA1) Ser293 and lactate dehydrogenase A (LDHA) Tyr10. These results suggested that BHLHE40 modulates PDH and LDH activity by regulating the phosphorylation status of PDHA1 and LDHA. We found that BHLHE40 enhanced AMPKα phosphorylation by directly suppressing the transcription of an AMPKα-specific phosphatase, PPM1F. Our immunohistochemical study showed that the expression of BHLHE40, PPM1F, and phosphorylated AMPKα correlated with the prognosis of endometrial cancer patients. Because AMPK is a central regulator of energy metabolism in cancer cells, targeting the BHLHE40‒PPM1F‒AMPK axis may represent a strategy to control cancer development.
Collapse
Affiliation(s)
- Kazuo Asanoma
- Department of Obstetrics and Gynecology, Faculty of Medical Sciences, Kyushu University, Fukuoka, Japan.
| | - Hiroshi Yagi
- Department of Obstetrics and Gynecology, Faculty of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Ichiro Onoyama
- Department of Obstetrics and Gynecology, Faculty of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Lin Cui
- Department of Obstetrics and Gynecology, Faculty of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Emiko Hori
- Department of Obstetrics and Gynecology, Faculty of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Minoru Kawakami
- Department of Obstetrics and Gynecology, Faculty of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Shoji Maenohara
- Department of Obstetrics and Gynecology, Faculty of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Kazuhisa Hachisuga
- Department of Obstetrics and Gynecology, Faculty of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Hiroshi Tomonobe
- Department of Obstetrics and Gynecology, Faculty of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Keisuke Kodama
- Department of Obstetrics and Gynecology, Faculty of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Masafumi Yasunaga
- Department of Obstetrics and Gynecology, Faculty of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Tatsuhiro Ohgami
- Department of Obstetrics and Gynecology, Faculty of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Kaoru Okugawa
- Department of Obstetrics and Gynecology, Faculty of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Hideaki Yahata
- Department of Obstetrics and Gynecology, Faculty of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Hiroyuki Kitao
- Oral Medicine Research Center, Fukuoka Dental College, Fukuoka, Japan
| | - Kiyoko Kato
- Department of Obstetrics and Gynecology, Faculty of Medical Sciences, Kyushu University, Fukuoka, Japan
| |
Collapse
|
20
|
Sun Y, Cao Y, Wan H, Memetimin A, Cao Y, Li L, Wu C, Wang M, Chen S, Li Q, Ma Y, Dong M, Jiang H. A mitophagy sensor PPTC7 controls BNIP3 and NIX degradation to regulate mitochondrial mass. Mol Cell 2024; 84:327-344.e9. [PMID: 38151018 DOI: 10.1016/j.molcel.2023.11.038] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Revised: 09/15/2023] [Accepted: 11/29/2023] [Indexed: 12/29/2023]
Abstract
Mitophagy mediated by BNIP3 and NIX critically regulates mitochondrial mass. Cellular BNIP3 and NIX levels are tightly controlled by SCFFBXL4-mediated ubiquitination to prevent excessive mitochondrial loss and lethal disease. Here, we report that knockout of PPTC7, a mitochondrial matrix protein, hyperactivates BNIP3-/NIX-mediated mitophagy and causes perinatal lethality that is rescued by NIX knockout in mice. Biochemically, the PPTC7 precursor is trapped by BNIP3 and NIX to the mitochondrial outer membrane, where PPTC7 scaffolds assembly of a substrate-PPTC7-SCFFBXL4 holocomplex to degrade BNIP3 and NIX, forming a homeostatic regulatory loop. PPTC7 possesses an unusually weak mitochondrial targeting sequence to facilitate its outer membrane retention and mitophagy control. Starvation upregulates PPPTC7 expression in mouse liver to repress mitophagy, which critically maintains hepatic mitochondrial mass, bioenergetics, and gluconeogenesis. Collectively, PPTC7 functions as a mitophagy sensor that integrates homeostatic and physiological signals to dynamically control BNIP3 and NIX degradation, thereby maintaining mitochondrial mass and cellular homeostasis.
Collapse
Affiliation(s)
- Yuqiu Sun
- Tsinghua Institute of Multidisciplinary Biomedical Research, Tsinghua University, Beijing 102206, China; National Institute of Biological Sciences, Beijing 102206, China; Beijing Key Laboratory of Cell Biology for Animal Aging, Beijing 102206, China
| | - Yu Cao
- College of Life Sciences, Beijing Normal University, Beijing 100875, China; National Institute of Biological Sciences, Beijing 102206, China; Beijing Key Laboratory of Cell Biology for Animal Aging, Beijing 102206, China
| | - Huayun Wan
- National Institute of Biological Sciences, Beijing 102206, China; Beijing Key Laboratory of Cell Biology for Animal Aging, Beijing 102206, China
| | - Adalet Memetimin
- National Institute of Biological Sciences, Beijing 102206, China
| | - Yang Cao
- National Institute of Biological Sciences, Beijing 102206, China
| | - Lin Li
- National Institute of Biological Sciences, Beijing 102206, China
| | - Chongyang Wu
- National Institute of Biological Sciences, Beijing 102206, China
| | - Meng Wang
- National Institute of Biological Sciences, Beijing 102206, China
| | - She Chen
- Tsinghua Institute of Multidisciplinary Biomedical Research, Tsinghua University, Beijing 102206, China; National Institute of Biological Sciences, Beijing 102206, China
| | - Qi Li
- Tsinghua Institute of Multidisciplinary Biomedical Research, Tsinghua University, Beijing 102206, China; National Institute of Biological Sciences, Beijing 102206, China
| | - Yan Ma
- Tsinghua Institute of Multidisciplinary Biomedical Research, Tsinghua University, Beijing 102206, China; National Institute of Biological Sciences, Beijing 102206, China
| | - Mengqiu Dong
- Tsinghua Institute of Multidisciplinary Biomedical Research, Tsinghua University, Beijing 102206, China; National Institute of Biological Sciences, Beijing 102206, China; Beijing Key Laboratory of Cell Biology for Animal Aging, Beijing 102206, China
| | - Hui Jiang
- Tsinghua Institute of Multidisciplinary Biomedical Research, Tsinghua University, Beijing 102206, China; National Institute of Biological Sciences, Beijing 102206, China; Beijing Key Laboratory of Cell Biology for Animal Aging, Beijing 102206, China.
| |
Collapse
|
21
|
Li YM, He HW, Zhang N. Targeting Protein Phosphatases for the Treatment of Chronic Liver Disease. Curr Drug Targets 2024; 25:171-189. [PMID: 38213163 DOI: 10.2174/0113894501278886231221092522] [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: 10/12/2023] [Revised: 11/27/2023] [Accepted: 11/29/2023] [Indexed: 01/13/2024]
Abstract
There exists a huge number of patients suffering from chronic liver disease worldwide. As a disease with high incidence and mortality worldwide, strengthening the research on the pathogenesis of chronic liver disease and the development of novel drugs is an important issue related to the health of all human beings. Phosphorylation modification of proteins plays a crucial role in cellular signal transduction, and phosphatases are involved in the development of liver diseases. Therefore, this article summarized the important role of protein phosphatases in chronic liver disease with the aim of facilitating the development of drugs targeting protein phosphatases for the treatment of chronic liver disease.
Collapse
Affiliation(s)
- Yi-Ming Li
- NHC Key Laboratory of Biotechnology for Microbial Drugs, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, China
| | - Hong-Wei He
- NHC Key Laboratory of Biotechnology for Microbial Drugs, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, China
| | - Na Zhang
- NHC Key Laboratory of Biotechnology for Microbial Drugs, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, China
| |
Collapse
|
22
|
Liu K, Liu J, Zhang X, Liu D, Yao W, Bu Y, Chen B. Identification of a Novel CD8 + T cell exhaustion-related gene signature for predicting survival in hepatocellular carcinoma. BMC Cancer 2023; 23:1185. [PMID: 38049741 PMCID: PMC10694949 DOI: 10.1186/s12885-023-11648-x] [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: 08/31/2023] [Accepted: 11/16/2023] [Indexed: 12/06/2023] Open
Abstract
BACKGROUND Hepatocellular carcinoma (HCC) is a major health concern, necessitating a deeper understanding of its prognosis and underlying mechanisms. This study aimed to investigate the mechanism and prognostic value of CD8+ T Cell exhaustion (CD8+ TEX)-related genes in HCC and construct a survival prognosis prediction model for patients with HCC. METHODS CD8+ TEX-related genes associated with HCC prognosis were analysed and identified, and a prognostic prediction model was constructed using the 'least absolute shrinkage and selection operator' Cox regression model. Immunohistochemistry was used to verify the expression of the model genes in HCC tissues. A nomogram was constructed based on risk scores and clinical features, and its predictive efficacy was verified. The expression of STAM, ANXA5, and MAD2L2 in HCC cell lines was detected by western blotting; subsequently, these genes were knocked down in HCC cell lines by small interfering RNA, and their effects on the proliferation and migration of HCC cell lines were detected by colony formation assay, cck8, wound healing, and transwell assays. RESULTS Six genes related to CD8+ TEX were included in the risk-prediction model. The prognosis of patients with HCC in the low-risk group was significantly better than that of those in the high-risk group. Cox regression analysis revealed that the risk score was an independent risk factor for the prognosis of patients with HCC. The differentially expressed genes in patients with high-risk HCC were mainly enriched in the nucleotide-binding oligomerization domain-containing protein-like receptor, hypoxia-inducible factor-1, and tumour programmed cell death protein (PD)-1/PD-L1 immune checkpoint pathways. The CD8+ TEX-related genes STAM, ANXA5, and MAD2L2 were knocked down in HCC cell lines to significantly inhibit cell proliferation and migration. The prediction results of the nomogram based on the risk score showed a good fit and application value. CONCLUSION The prediction model based on CD8+ TEX-related genes can predict the prognosis of HCC and provide a theoretical basis for the early identification of patients with poor HCC prognosis.
Collapse
Affiliation(s)
- Kejun Liu
- Department of Hepatobiliary Surgery, General Hospital of Ningxia Medical University, Yinchuan, 750004, China
- School of Clinical Medicine, Ningxia Medical University, Yinchuan, 750004, China
| | - Junhao Liu
- School of Clinical Medicine, Ningxia Medical University, Yinchuan, 750004, China
- Department of Hepatobiliary Surgery, People's Hospital of Ningxia Hui Autonomous Region, Yinchuan, 750002, China
| | - Xusheng Zhang
- Department of Hepatobiliary Surgery, General Hospital of Ningxia Medical University, Yinchuan, 750004, China
- School of Clinical Medicine, Ningxia Medical University, Yinchuan, 750004, China
| | - Di Liu
- Department of Hepatobiliary Surgery, General Hospital of Ningxia Medical University, Yinchuan, 750004, China
- School of Clinical Medicine, Ningxia Medical University, Yinchuan, 750004, China
| | - Weijie Yao
- Department of Hepatobiliary Surgery, General Hospital of Ningxia Medical University, Yinchuan, 750004, China
- School of Clinical Medicine, Ningxia Medical University, Yinchuan, 750004, China
| | - Yang Bu
- School of Clinical Medicine, Ningxia Medical University, Yinchuan, 750004, China.
- Department of Hepatobiliary Surgery, People's Hospital of Ningxia Hui Autonomous Region, Yinchuan, 750002, China.
| | - Bendong Chen
- Department of Hepatobiliary Surgery, General Hospital of Ningxia Medical University, Yinchuan, 750004, China.
- School of Clinical Medicine, Ningxia Medical University, Yinchuan, 750004, China.
| |
Collapse
|
23
|
Wang L, Xie Y, Myrzagali S, Pu W, Liu E. Metal ions as effectual tools for cancer with traditional Chinese medicine. ACUPUNCTURE AND HERBAL MEDICINE 2023; 3:296-308. [DOI: 10.1097/hm9.0000000000000083] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
Abstract
Malignant tumor has become a major threat affecting human health, and is one of the main causes of human death. Recent studies have shown that many traditional Chinese medicines (TCM) have good anti-tumor activity, which may improve the therapeutic effect of routine treatment and quality of life with lower toxicity. However, the efficacy of TCM alone for the treatment of tumors is limited. Metal ions are essential substances for maintaining normal physiological activities. This article summarized the multiple mechanisms in which metal ions are involved in the prevention and treatment of tumors in TCM.
Collapse
Affiliation(s)
- Lei Wang
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Yingqiu Xie
- Department of Biology, School of Sciences and Humanities, Nazarbayev University, Astana, Kazakhstan
| | - Sandugash Myrzagali
- Department of Biology, School of Sciences and Humanities, Nazarbayev University, Astana, Kazakhstan
| | - Weiling Pu
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Erwei Liu
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| |
Collapse
|
24
|
Liu J, Meng F, Wang W, Wu M, Zhang Y, Cui M, Qiu C, Hu F, Zhao D, Wang D, Liu C, Liu D, Xu Z, Wang Y, Li W, Li C. Medial prefrontal cortical PPM1F alters depression-related behaviors by modifying p300 activity via the AMPK signaling pathway. CNS Neurosci Ther 2023; 29:3624-3643. [PMID: 37309288 PMCID: PMC10580341 DOI: 10.1111/cns.14293] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Revised: 05/16/2023] [Accepted: 05/22/2023] [Indexed: 06/14/2023] Open
Abstract
AIMS Protein phosphatase Mg2+/Mn2+-dependent 1F (PPM1F) is a serine/threonine phosphatase, and its dysfunction in depression in the hippocampal dentate gyrus has been previously identified. Nevertheless, its role in depression of another critical emotion-controlling brain region, the medial prefrontal cortex (mPFC), remains unclear. We explored the functional relevance of PPM1F in the pathogenesis of depression. METHODS The gene expression levels and colocalization of PPM1F in the mPFC of depressed mice were measured by real-time PCR, western blot and immunohistochemistry. An adeno-associated virus strategy was applied to determine the impact of knockdown or overexpression of PPM1F in the excitatory neurons on depression-related behaviors under basal and stress conditions in both male and female mice. The neuronal excitability, expression of p300 and AMPK phosphorylation levels in the mPFC after knockdown of PPM1F were measured by electrophysiological recordings, real-time PCR and western blot. The depression-related behavior induced by PPM1F knockdown after AMPKα2 knockout or the antidepressant activity of PPM1F overexpression after inhibiting acetylation activity of p300 was evaluated. RESULTS Our results indicate that the expression levels of PPM1F were largely decreased in the mPFC of mice exposed to chronic unpredictable stress (CUS). Behavioral alterations relevant to depression emerged with short hairpin RNA (shRNA)-mediated genetic knockdown of PPM1F in the mPFC, while overexpression of PPM1F produced antidepressant activity and ameliorated behavioral responses to stress in CUS-exposed mice. Molecularly, PPM1F knockdown decreased the excitability of pyramidal neurons in the mPFC, and restoring this low excitability decreased the depression-related behaviors induced by PPM1F knockdown. PPM1F knockdown reduced the expression of CREB-binding protein (CBP)/E1A-associated protein (p300), a histone acetyltransferase (HAT), and induced hyperphosphorylation of AMPK, resulting in microglial activation and upregulation of proinflammatory cytokines. Conditional knockout of AMPK revealed an antidepressant phenotype, which can also block depression-related behaviors induced by PPM1F knockdown. Furthermore, inhibiting the acetylase activity of p300 abolished the beneficial effects of PPM1F elevation on CUS-induced depressive behaviors. CONCLUSION Our findings demonstrate that PPM1F in the mPFC modulates depression-related behavioral responses by regulating the function of p300 via the AMPK signaling pathway.
Collapse
|
25
|
Zhu Z, Yang M, Yang G, Zhang B, Cao X, Yuan J, Ge F, Wang S. PP2C phosphatases Ptc1 and Ptc2 dephosphorylate PGK1 to regulate autophagy and aflatoxin synthesis in the pathogenic fungus Aspergillus flavus. mBio 2023; 14:e0097723. [PMID: 37754565 PMCID: PMC10653812 DOI: 10.1128/mbio.00977-23] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Accepted: 08/08/2023] [Indexed: 09/28/2023] Open
Abstract
IMPORTANCE Aspergillus flavus is a model filamentous fungus that can produce aflatoxins when it infects agricultural crops. This study evaluated the protein phosphatase 2C (PP2C) family as a potential drug target with important physiological functions and pathological significance in A. flavus. We found that two redundant PP2C phosphatases, Ptc1 and Ptc2, regulate conidia development, aflatoxin synthesis, autophagic vesicle formation, and seed infection. The target protein phosphoglycerate kinase 1 (PGK1) that interacts with Ptc1 and Ptc2 is essential to regulate metabolism and the autophagy process. Furthermore, Ptc1 and Ptc2 regulate the phosphorylation level of PGK1 S203, which is important for influencing aflatoxin synthesis. Our results provide a potential target for interdicting the toxicity of A. flavus.
Collapse
Affiliation(s)
- Zhuo Zhu
- State Key Laboratory of Ecological Pest Control for Fujian and Taiwan Crops, Key Laboratory of Biopesticide and Chemical Biology of Education Ministry, Key Laboratory of Pathogenic Fungi, School of Life Sciences, Fujian Agriculture and Forestry University, Fuzhou, China
- Mycotoxins of Fujian Province, School of Life Sciences, Fujian Agriculture and Forestry University, Fuzhou, China
| | - Mingkun Yang
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, China
| | - Guang Yang
- State Key Laboratory of Ecological Pest Control for Fujian and Taiwan Crops, Key Laboratory of Biopesticide and Chemical Biology of Education Ministry, Key Laboratory of Pathogenic Fungi, School of Life Sciences, Fujian Agriculture and Forestry University, Fuzhou, China
| | - Bei Zhang
- State Key Laboratory of Ecological Pest Control for Fujian and Taiwan Crops, Key Laboratory of Biopesticide and Chemical Biology of Education Ministry, Key Laboratory of Pathogenic Fungi, School of Life Sciences, Fujian Agriculture and Forestry University, Fuzhou, China
| | - Xiaohong Cao
- State Key Laboratory of Ecological Pest Control for Fujian and Taiwan Crops, Key Laboratory of Biopesticide and Chemical Biology of Education Ministry, Key Laboratory of Pathogenic Fungi, School of Life Sciences, Fujian Agriculture and Forestry University, Fuzhou, China
| | - Jun Yuan
- State Key Laboratory of Ecological Pest Control for Fujian and Taiwan Crops, Key Laboratory of Biopesticide and Chemical Biology of Education Ministry, Key Laboratory of Pathogenic Fungi, School of Life Sciences, Fujian Agriculture and Forestry University, Fuzhou, China
| | - Feng Ge
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, China
| | - Shihua Wang
- State Key Laboratory of Ecological Pest Control for Fujian and Taiwan Crops, Key Laboratory of Biopesticide and Chemical Biology of Education Ministry, Key Laboratory of Pathogenic Fungi, School of Life Sciences, Fujian Agriculture and Forestry University, Fuzhou, China
| |
Collapse
|
26
|
Chen Y, Zhao H, Wang Y, Qiu X, Gao G, Zhu A, Chen P, Wang X, Chen K, Chen J, Chen P, Chen J. Genome-Wide Identification and Expression Analysis of BnPP2C Gene Family in Response to Multiple Stresses in Ramie ( Boehmeria nivea L.). Int J Mol Sci 2023; 24:15282. [PMID: 37894962 PMCID: PMC10607689 DOI: 10.3390/ijms242015282] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Revised: 10/04/2023] [Accepted: 10/12/2023] [Indexed: 10/29/2023] Open
Abstract
The protein phosphatase 2C (PP2C), a key regulator of the ABA signaling pathway, plays important roles in plant growth and development, hormone signaling, and abiotic stress response. Although the PP2C gene family has been identified in many species, systematic analysis was still relatively lacking in ramie (Boehmeria nivea L.). In the present study, we identified 63 BnPP2C genes from the ramie genome, using bioinformatics analysis, and classified them into 12 subfamilies, and this classification was consistently supported by their gene structures and conserved motifs. In addition, we observed that the functional differentiation of the BnPP2C family of genes was restricted and that fragment replication played a major role in the amplification of the BnPP2C gene family. The promoter cis-regulatory elements of BnPP2C genes were mainly involved in light response regulation, phytohormone synthesis, transport and signaling, environmental stress response and plant growth and development regulation. We identified BnPP2C genes with tissue specificity, using ramie transcriptome data from different tissues, in rhizome leaves and bast fibers. The qRT-PCR results showed that the BnPP2C1, BnPP2C26 and BnPP2C27 genes had a strong response to drought, high salt and ABA, and there were a large number of stress-responsive elements in the promoter region of BnPP2C1 and BnPP2C26. The results suggested that BnPP2C1 and BnPP2C26 could be used as the candidate genes for drought and salt tolerance in ramie. These results provide a reference for further studies on the function of the PP2C gene and advance the development of the mechanism of ramie stress response, with a view to providing candidate genes for the molecular breeding of ramie for drought and salt tolerance.
Collapse
Affiliation(s)
- Yu Chen
- Institute of Bast Fiber Crops, Chinese Academy of Agricultural Sciences, Changsha 410221, China; (Y.C.); (H.Z.); (Y.W.); (X.Q.); (G.G.); (A.Z.); (P.C.); (X.W.); (K.C.); (J.C.)
- College of Agriculture, Guangxi University, Nanning 530004, China
| | - Haohan Zhao
- Institute of Bast Fiber Crops, Chinese Academy of Agricultural Sciences, Changsha 410221, China; (Y.C.); (H.Z.); (Y.W.); (X.Q.); (G.G.); (A.Z.); (P.C.); (X.W.); (K.C.); (J.C.)
| | - Yue Wang
- Institute of Bast Fiber Crops, Chinese Academy of Agricultural Sciences, Changsha 410221, China; (Y.C.); (H.Z.); (Y.W.); (X.Q.); (G.G.); (A.Z.); (P.C.); (X.W.); (K.C.); (J.C.)
| | - Xiaojun Qiu
- Institute of Bast Fiber Crops, Chinese Academy of Agricultural Sciences, Changsha 410221, China; (Y.C.); (H.Z.); (Y.W.); (X.Q.); (G.G.); (A.Z.); (P.C.); (X.W.); (K.C.); (J.C.)
| | - Gang Gao
- Institute of Bast Fiber Crops, Chinese Academy of Agricultural Sciences, Changsha 410221, China; (Y.C.); (H.Z.); (Y.W.); (X.Q.); (G.G.); (A.Z.); (P.C.); (X.W.); (K.C.); (J.C.)
| | - Aiguo Zhu
- Institute of Bast Fiber Crops, Chinese Academy of Agricultural Sciences, Changsha 410221, China; (Y.C.); (H.Z.); (Y.W.); (X.Q.); (G.G.); (A.Z.); (P.C.); (X.W.); (K.C.); (J.C.)
| | - Ping Chen
- Institute of Bast Fiber Crops, Chinese Academy of Agricultural Sciences, Changsha 410221, China; (Y.C.); (H.Z.); (Y.W.); (X.Q.); (G.G.); (A.Z.); (P.C.); (X.W.); (K.C.); (J.C.)
| | - Xiaofei Wang
- Institute of Bast Fiber Crops, Chinese Academy of Agricultural Sciences, Changsha 410221, China; (Y.C.); (H.Z.); (Y.W.); (X.Q.); (G.G.); (A.Z.); (P.C.); (X.W.); (K.C.); (J.C.)
| | - Kunmei Chen
- Institute of Bast Fiber Crops, Chinese Academy of Agricultural Sciences, Changsha 410221, China; (Y.C.); (H.Z.); (Y.W.); (X.Q.); (G.G.); (A.Z.); (P.C.); (X.W.); (K.C.); (J.C.)
| | - Jia Chen
- Institute of Bast Fiber Crops, Chinese Academy of Agricultural Sciences, Changsha 410221, China; (Y.C.); (H.Z.); (Y.W.); (X.Q.); (G.G.); (A.Z.); (P.C.); (X.W.); (K.C.); (J.C.)
| | - Peng Chen
- College of Agriculture, Guangxi University, Nanning 530004, China
| | - Jikang Chen
- Institute of Bast Fiber Crops, Chinese Academy of Agricultural Sciences, Changsha 410221, China; (Y.C.); (H.Z.); (Y.W.); (X.Q.); (G.G.); (A.Z.); (P.C.); (X.W.); (K.C.); (J.C.)
- National Breeding Center or Bast Fiber Crops, MARA, Changsha 410221, China
| |
Collapse
|
27
|
Xia X, Pi W, Chen M, Wang W, Cai D, Wang X, Lan Y, Yang H. Emerging roles of PHLPP phosphatases in lung cancer. Front Oncol 2023; 13:1216131. [PMID: 37576883 PMCID: PMC10414793 DOI: 10.3389/fonc.2023.1216131] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Accepted: 07/12/2023] [Indexed: 08/15/2023] Open
Abstract
Pleckstrin homologous domain leucine-rich repeating protein phosphatases (PHLPPs) were originally identified as protein kinase B (Akt) kinase hydrophobic motif specific phosphatases to maintain the cellular homeostasis. With the continuous expansion of PHLPPs research, imbalanced-PHLPPs were mainly found as a tumor suppressor gene of a variety of solid tumors. In this review, we simply described the history and structures of PHLPPs and summarized the recent achievements in emerging roles of PHLPPs in lung cancer by 1) the signaling pathways affected by PHLPPs including Phosphoinositide 3-kinase (PI3K)/AKT, RAS/RAF/mitogen-activated protein kinase (MEK)/extracellular signal-regulated kinase (ERK) and Protein kinase C (PKC) signaling cascades. 2) function of PHLPPs regulatory factor USP46 and miR-190/miR-215, 3) the potential roles of PHLPPs in disease prognosis, Epidermal growth factor receptors (EGFR)- tyrosine kinase inhibitor (TKI) resistance and DNA damage, 4) and the possible function of PHLPPs in radiotherapy, ferroptosis and inflammation response. Therefore, PHLPPs can be considered as either biomarker or prognostic marker for lung cancer treatment.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Haihua Yang
- Key Laboratory of Radiation Oncology of Taizhou, Radiation Oncology Institute of Enze Medical Health Academy, Department of Radiation Oncology, Taizhou Hospital Affiliated to Wenzhou Medical University, Taizhou, Zhejiang, China
| |
Collapse
|
28
|
Liu R, Wu J, Guo H, Yao W, Li S, Lu Y, Jia Y, Liang X, Tang J, Zhang H. Post-translational modifications of histones: Mechanisms, biological functions, and therapeutic targets. MedComm (Beijing) 2023; 4:e292. [PMID: 37220590 PMCID: PMC10200003 DOI: 10.1002/mco2.292] [Citation(s) in RCA: 53] [Impact Index Per Article: 26.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Revised: 05/05/2023] [Accepted: 05/09/2023] [Indexed: 05/25/2023] Open
Abstract
Histones are DNA-binding basic proteins found in chromosomes. After the histone translation, its amino tail undergoes various modifications, such as methylation, acetylation, phosphorylation, ubiquitination, malonylation, propionylation, butyrylation, crotonylation, and lactylation, which together constitute the "histone code." The relationship between their combination and biological function can be used as an important epigenetic marker. Methylation and demethylation of the same histone residue, acetylation and deacetylation, phosphorylation and dephosphorylation, and even methylation and acetylation between different histone residues cooperate or antagonize with each other, forming a complex network. Histone-modifying enzymes, which cause numerous histone codes, have become a hot topic in the research on cancer therapeutic targets. Therefore, a thorough understanding of the role of histone post-translational modifications (PTMs) in cell life activities is very important for preventing and treating human diseases. In this review, several most thoroughly studied and newly discovered histone PTMs are introduced. Furthermore, we focus on the histone-modifying enzymes with carcinogenic potential, their abnormal modification sites in various tumors, and multiple essential molecular regulation mechanism. Finally, we summarize the missing areas of the current research and point out the direction of future research. We hope to provide a comprehensive understanding and promote further research in this field.
Collapse
Affiliation(s)
- Ruiqi Liu
- Cancer CenterDepartment of Radiation OncologyZhejiang Provincial People's HospitalAffiliated People's HospitalHangzhou Medical CollegeHangzhouZhejiangChina
- Graduate DepartmentBengbu Medical College, BengbuAnhuiChina
| | - Jiajun Wu
- Graduate DepartmentBengbu Medical College, BengbuAnhuiChina
- Otolaryngology & Head and Neck CenterCancer CenterDepartment of Head and Neck SurgeryZhejiang Provincial People's HospitalAffiliated People's Hospital, Hangzhou Medical CollegeHangzhouZhejiangChina
| | - Haiwei Guo
- Otolaryngology & Head and Neck CenterCancer CenterDepartment of Head and Neck SurgeryZhejiang Provincial People's HospitalAffiliated People's Hospital, Hangzhou Medical CollegeHangzhouZhejiangChina
| | - Weiping Yao
- Cancer CenterDepartment of Radiation OncologyZhejiang Provincial People's HospitalAffiliated People's HospitalHangzhou Medical CollegeHangzhouZhejiangChina
- Graduate DepartmentBengbu Medical College, BengbuAnhuiChina
| | - Shuang Li
- Cancer CenterDepartment of Radiation OncologyZhejiang Provincial People's HospitalAffiliated People's HospitalHangzhou Medical CollegeHangzhouZhejiangChina
- Graduate DepartmentJinzhou Medical UniversityJinzhouLiaoningChina
| | - Yanwei Lu
- Cancer CenterDepartment of Radiation OncologyZhejiang Provincial People's HospitalAffiliated People's HospitalHangzhou Medical CollegeHangzhouZhejiangChina
| | - Yongshi Jia
- Cancer CenterDepartment of Radiation OncologyZhejiang Provincial People's HospitalAffiliated People's HospitalHangzhou Medical CollegeHangzhouZhejiangChina
| | - Xiaodong Liang
- Cancer CenterDepartment of Radiation OncologyZhejiang Provincial People's HospitalAffiliated People's HospitalHangzhou Medical CollegeHangzhouZhejiangChina
- Graduate DepartmentBengbu Medical College, BengbuAnhuiChina
| | - Jianming Tang
- Department of Radiation OncologyThe First Hospital of Lanzhou UniversityLanzhou UniversityLanzhouGansuChina
| | - Haibo Zhang
- Cancer CenterDepartment of Radiation OncologyZhejiang Provincial People's HospitalAffiliated People's HospitalHangzhou Medical CollegeHangzhouZhejiangChina
| |
Collapse
|
29
|
Ozcelik F, Arslan S, Ozguc Caliskan B, Kardas F, Ozkul Y, Dundar M. PPM1K defects cause mild maple syrup urine disease: The second case in the literature. Am J Med Genet A 2023; 191:1360-1365. [PMID: 36706222 DOI: 10.1002/ajmg.a.63129] [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: 10/09/2022] [Revised: 01/11/2023] [Accepted: 01/13/2023] [Indexed: 01/29/2023]
Abstract
Maple syrup urine disease (MSUD) is an inborn error of metabolism caused by the insufficient catabolism of branched-chain amino acids. BCKDHA, BCKDHB, DBT, and DLD encode the subunits of the branched-chain α-ketoacid dehydrogenase complex, which is responsible for the catabolism of these amino acids. Biallelic pathogenic variants in BCKDHA, BCKDHB, or DBT are characteristic of MSUD. In addition, a patient with a PPM1K defect was previously reported. PPM1K dephosphorylates and activates the enzyme complex. We report a patient with MSUD with mild findings and elevated BCAA levels carrying a novel homozygous start-loss variant in PPM1K. Our study offers further evidence that PPM1K variants cause mild MSUD.
Collapse
Affiliation(s)
- Firat Ozcelik
- Department of Medical Genetics, Erciyes University, Kayseri, Turkey
| | - Sezai Arslan
- Division of Nutrition and Metabolism, Department of Pediatrics, Erciyes University, Kayseri, Turkey
| | | | - Fatih Kardas
- Division of Nutrition and Metabolism, Department of Pediatrics, Erciyes University, Kayseri, Turkey
| | - Yusuf Ozkul
- Department of Medical Genetics, Erciyes University, Kayseri, Turkey
| | - Munis Dundar
- Department of Medical Genetics, Erciyes University, Kayseri, Turkey
| |
Collapse
|
30
|
Kaspers MS, Pogenberg V, Pett C, Ernst S, Ecker F, Ochtrop P, Groll M, Hedberg C, Itzen A. Dephosphocholination by Legionella effector Lem3 functions through remodelling of the switch II region of Rab1b. Nat Commun 2023; 14:2245. [PMID: 37076474 PMCID: PMC10115812 DOI: 10.1038/s41467-023-37621-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2022] [Accepted: 03/22/2023] [Indexed: 04/21/2023] Open
Abstract
Bacterial pathogens often make use of post-translational modifications to manipulate host cells. Legionella pneumophila, the causative agent of Legionnaires disease, secretes the enzyme AnkX that uses cytidine diphosphate-choline to post-translationally modify the human small G-Protein Rab1 with a phosphocholine moiety at Ser76. Later in the infection, the Legionella enzyme Lem3 acts as a dephosphocholinase, hydrolytically removing the phosphocholine. While the molecular mechanism for Rab1 phosphocholination by AnkX has recently been resolved, structural insights into the activity of Lem3 remained elusive. Here, we stabilise the transient Lem3:Rab1b complex by substrate mediated covalent capture. Through crystal structures of Lem3 in the apo form and in complex with Rab1b, we reveal Lem3's catalytic mechanism, showing that it acts on Rab1 by locally unfolding it. Since Lem3 shares high structural similarity with metal-dependent protein phosphatases, our Lem3:Rab1b complex structure also sheds light on how these phosphatases recognise protein substrates.
Collapse
Affiliation(s)
- Marietta S Kaspers
- Institute of Biochemistry and Signal Transduction, University Medical Centre Hamburg-Eppendorf (UKE), Martinistr. 52, 20246, Hamburg, Germany
| | - Vivian Pogenberg
- Institute of Biochemistry and Signal Transduction, University Medical Centre Hamburg-Eppendorf (UKE), Martinistr. 52, 20246, Hamburg, Germany
| | - Christian Pett
- Chemical Biology Center (KBC), Department of Chemistry, Umeå University, Linnaeus väg 10, 90187, Umeå, Sweden
| | - Stefan Ernst
- Center for Integrated Protein Science Munich (CIPSM), Department Chemistry, Technical University of Munich, Lichtenbergstrasse 4, 85747, Garching, Germany
| | - Felix Ecker
- Center for Protein Assemblies, Technical University of Munich, Ernst-Otto-Fischer-Str. 8, 85748, Garching, Germany
| | - Philipp Ochtrop
- Chemical Biology Center (KBC), Department of Chemistry, Umeå University, Linnaeus väg 10, 90187, Umeå, Sweden
| | - Michael Groll
- Center for Protein Assemblies, Technical University of Munich, Ernst-Otto-Fischer-Str. 8, 85748, Garching, Germany
| | - Christian Hedberg
- Chemical Biology Center (KBC), Department of Chemistry, Umeå University, Linnaeus väg 10, 90187, Umeå, Sweden
| | - Aymelt Itzen
- Institute of Biochemistry and Signal Transduction, University Medical Centre Hamburg-Eppendorf (UKE), Martinistr. 52, 20246, Hamburg, Germany.
- Centre for Structural Systems Biology, University Medical Centre Hamburg-Eppendorf (UKE), Martinistr. 52, 20246, Hamburg, Germany.
| |
Collapse
|
31
|
Shimizu K, Inoue KI, Oishi T, Takada M, Fukada Y, Imai H. Diurnal variation in declarative memory and the involvement of SCOP in cognitive functions in nonhuman primates. Mol Brain 2023; 16:31. [PMID: 36966302 PMCID: PMC10039603 DOI: 10.1186/s13041-023-01022-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Accepted: 03/22/2023] [Indexed: 03/27/2023] Open
Abstract
Cognitive functions depend on the time of day in various organisms. Previously, we found that 24-h recognition memory performance of nocturnal mice changes diurnally through SCOP protein-dependent regulation. It remains unknown whether diurnal change and SCOP-dependent regulation of memory performance are conserved across species with diurnal/nocturnal habits. We tested whether the memory performance of diurnal Japanese macaques depends on the time of day. The memory association between bitter taste of drinking water and the nozzle color of the water bottle was established during the light period of the day to evaluate of memory performance for macaques. Here we found diurnal variation of declarative memory in Japanese macaques. The middle of the daytime is the most effective time for memory performance during the light period. To assess whether SCOP is involved in declarative memory performance, we interfered with SCOP expression by using lentiviral vector expressing shRNA against Scop in the hippocampus of Japanese macaques. Scop knockdown in the hippocampus abrogated the memory performance in the middle of the daytime. Our results implicate that SCOP in the hippocampus is necessary for the diurnal rhythm of the memory system and that the SCOP-dependent memory regulation system may be conserved in mammals.
Collapse
Affiliation(s)
- Kimiko Shimizu
- Department of Biological Sciences, School of Science, The University of Tokyo, Tokyo, 113-0033, Japan.
- Department of Pathological Cell Biology, Medical Research Institute, Tokyo Medical and Dental University, Tokyo, 113-8510, Japan.
- Laboratory of Animal Resources, Graduate School of Medicine, Center for Disease Biology and Integrative Medicine, The University of Tokyo, Tokyo, 113-0033, Japan.
| | - Ken-Ichi Inoue
- Systems Neuroscience Section, Center for the Evolutionary Origins of Human Behavior, Kyoto University, Inuyama, Aichi, 484-8506, Japan
| | - Takao Oishi
- Systems Neuroscience Section, Center for the Evolutionary Origins of Human Behavior, Kyoto University, Inuyama, Aichi, 484-8506, Japan
| | - Masahiko Takada
- Systems Neuroscience Section, Center for the Evolutionary Origins of Human Behavior, Kyoto University, Inuyama, Aichi, 484-8506, Japan
| | - Yoshitaka Fukada
- Department of Biological Sciences, School of Science, The University of Tokyo, Tokyo, 113-0033, Japan.
- Laboratory of Animal Resources, Graduate School of Medicine, Center for Disease Biology and Integrative Medicine, The University of Tokyo, Tokyo, 113-0033, Japan.
| | - Hiroo Imai
- Molecular Biology Section, Center for the Evolutionary Origins of Human Behavior, Kyoto University, Inuyama, Aichi, 484-8506, Japan.
| |
Collapse
|
32
|
Zeng R, Wang L, Zhang Y, Yang Y, Yang J, Qin Y. Exploring the immunological role and prognostic potential of PPM1M in pan-cancer. Medicine (Baltimore) 2023; 102:e32758. [PMID: 36961170 PMCID: PMC10036021 DOI: 10.1097/md.0000000000032758] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/20/2022] [Accepted: 01/05/2023] [Indexed: 03/25/2023] Open
Abstract
BACKGROUND PPM1M is a member of the metal-dependent protein phosphatase family, and its role in the immunization process has not been studied in depth. In this study, we investigated the role of PPM1M in pan-cancer. METHODS Samples of cancer and normal tissues were obtained from the cancer genome atlas and genotype-tissue expression. Kaplan-Meier survival curves and Cox regression were used to analyze the effect of PPM1M on prognosis. Functional and pathway enrichment analyses were performed using the R package "clusterProfiler" to explore the role of PPM1M. The Sanger Box database was used to analyze the relationship between PPM1M and tumor immune checkpoint, tumor mutational burden, and microsatellite instability. The Tumor Immune Estimation Resource 2 database and CIBERSORT method were used to analyze the relationship between PPM1M and tumor-infiltrating immune cells. Finally, the cBioPortal database was used to analyze the genomic variation in PPM1M. RESULTS Among the variety of tumors, the expression of PPM1M was higher in normal tissues than in cancerous tissues. The expression of PPM1M is closely associated with patient prognosis, tumor immune checkpoint, tumor mutational burden, and microsatellite instability. PPM1M is closely associated with the infiltration of immune cells into the tumor microenvironment. In addition, PPM1M is involved in the regulation of several immune-related pathways. CONCLUSION In pan-cancer, PPM1M affects patient prognosis and may be a potential immunological biomarker. Furthermore, PPM1M may be a potential therapeutic target in tumor immunology.
Collapse
Affiliation(s)
- Rongruo Zeng
- Department of Health Management, The People’s Hospital of Guangxi Zhuang Autonomous Region & Research Center of Health Management, Guangxi Academy of Medical Sciences, Nanning, Guangxi, People’s Republic of China
- Department of Pathology, Guangxi Medical University Cancer Hospital, Nanning, Guangxi, People’s Republic of China
| | - Lulu Wang
- Department of Health Management, The People’s Hospital of Guangxi Zhuang Autonomous Region & Research Center of Health Management, Guangxi Academy of Medical Sciences, Nanning, Guangxi, People’s Republic of China
| | - Yuxu Zhang
- Department of International Medicine Services, The People’s Hospital of Guangxi Zhuang Autonomous Region, Nanning, Guangxi, People’s Republic of China
| | - Ye Yang
- Department of Rehabilitation Medicine, Guangxi Medical University, Nanning, Guangxi, People’s Republic of China
| | - Jie Yang
- Department of Health Management, The People’s Hospital of Guangxi Zhuang Autonomous Region & Research Center of Health Management, Guangxi Academy of Medical Sciences, Nanning, Guangxi, People’s Republic of China
| | - Yan Qin
- Department of Health Management, The People’s Hospital of Guangxi Zhuang Autonomous Region & Research Center of Health Management, Guangxi Academy of Medical Sciences, Nanning, Guangxi, People’s Republic of China
| |
Collapse
|
33
|
Li Z, Chen R, Li Y, Zhou Q, Zhao H, Zeng K, Zhao B, Lu Z. A comprehensive overview of PPM1B: From biological functions to diseases. Eur J Pharmacol 2023; 947:175633. [PMID: 36863552 DOI: 10.1016/j.ejphar.2023.175633] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2022] [Revised: 02/08/2023] [Accepted: 02/28/2023] [Indexed: 03/04/2023]
Abstract
Reversible phosphorylation of proteins is an important mechanism that regulates cellular processes, which are precisely regulated by protein kinases and phosphatases. PPM1B is a metal ion-dependent serine/threonine protein phosphatase, which regulates multiple biological functions by targeting substrate dephosphorylation, such as cell cycle, energy metabolism, inflammatory responses. In this review, we summarized the occurrent understandings of PPM1B focused on its regulation of signaling pathways, related diseases, and small-molecular inhibitors, which may provide new insights for the identification of PPM1B inhibitors and the treatment of PPM1B-related diseases.
Collapse
Affiliation(s)
- Zhongyao Li
- School of Pharmacy and Pharmaceutical Sciences, Institute of Materia Medica, Shandong First Medical University, Shandong Academy of Medical Sciences, NHC Key Laboratory of Biotechnology Drugs (Shandong Academy of Medical Sciences), Key Lab for Rare & Uncommon Diseases of Shandong Province, Ji'nan, 250117, Shandong, China
| | - Ruoyu Chen
- Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Ji'nan, 250012, Shandong, China
| | - Yanxia Li
- Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Ji'nan, 250012, Shandong, China
| | - Qian Zhou
- Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Ji'nan, 250012, Shandong, China
| | - Huanxin Zhao
- School of Pharmacy and Pharmaceutical Sciences, Institute of Materia Medica, Shandong First Medical University, Shandong Academy of Medical Sciences, NHC Key Laboratory of Biotechnology Drugs (Shandong Academy of Medical Sciences), Key Lab for Rare & Uncommon Diseases of Shandong Province, Ji'nan, 250117, Shandong, China
| | - Kewu Zeng
- School of Pharmacy and Pharmaceutical Sciences, Institute of Materia Medica, Shandong First Medical University, Shandong Academy of Medical Sciences, NHC Key Laboratory of Biotechnology Drugs (Shandong Academy of Medical Sciences), Key Lab for Rare & Uncommon Diseases of Shandong Province, Ji'nan, 250117, Shandong, China.
| | - Baobing Zhao
- Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Ji'nan, 250012, Shandong, China; Department of Pharmacology, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Ji'nan, 250012, Shandong, China; NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Ji'nan, 250012, Shandong, China.
| | - Zhiyuan Lu
- School of Pharmacy and Pharmaceutical Sciences, Institute of Materia Medica, Shandong First Medical University, Shandong Academy of Medical Sciences, NHC Key Laboratory of Biotechnology Drugs (Shandong Academy of Medical Sciences), Key Lab for Rare & Uncommon Diseases of Shandong Province, Ji'nan, 250117, Shandong, China.
| |
Collapse
|
34
|
Ge Q, Shi Z, Zou KA, Ying J, Chen J, Yuan W, Wang W, Xiao L, Lin X, Chen D, Feng XH, Wang PE, Tong P, Jin H. Protein phosphatase PPM1A inhibition attenuates osteoarthritis via regulating TGF-β/Smad2 signaling in chondrocytes. JCI Insight 2023; 8:166688. [PMID: 36752205 PMCID: PMC9926971 DOI: 10.1172/jci.insight.166688] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Accepted: 12/21/2022] [Indexed: 02/09/2023] Open
Abstract
TGF-β signaling is crucial for modulating osteoarthritis (OA), and protein phosphatase magnesium-dependent 1A (PPM1A) has been reported as a phosphatase of SMAD2 and regulates TGF-β signaling, while the role of PPM1A in cartilage homeostasis and OA development remains largely unexplored. In this study, we found increased PPM1A expression in OA chondrocytes and confirmed the interaction between PPM1A and phospho-SMAD2 (p-SMAD2). Importantly, our data show that PPM1A KO substantially protected mice treated with destabilization of medial meniscus (DMM) surgery against cartilage degeneration and subchondral sclerosis. Additionally, PPM1A ablation reduced the cartilage catabolism and cell apoptosis after the DMM operation. Moreover, p-SMAD2 expression in chondrocytes from KO mice was higher than that in WT controls with DMM induction. However, intraarticular injection with SD-208, repressing TGF-β/SMAD2 signaling, dramatically abolished protective phenotypes in PPM1A-KO mice. Finally, a specific pharmacologic PPM1A inhibitor, Sanguinarine chloride (SC) or BC-21, was able to ameliorate OA severity in C57BL/6J mice. In summary, our study identified PPM1A as a pivotal regulator of cartilage homeostasis and demonstrated that PPM1A inhibition attenuates OA progression via regulating TGF-β/SMAD2 signaling in chondrocytes and provided PPM1A as a potential target for OA treatment.
Collapse
Affiliation(s)
- Qinwen Ge
- Institute of Orthopaedics and Traumatology, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China.,The First College of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, China
| | - Zhenyu Shi
- Institute of Orthopaedics and Traumatology, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China.,Department of Orthopaedic Surgery, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China
| | - Kai-ao Zou
- Institute of Orthopaedics and Traumatology, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China.,The First College of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, China
| | - Jun Ying
- Institute of Orthopaedics and Traumatology, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China.,Department of Orthopaedic Surgery, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China
| | - Jiali Chen
- Institute of Orthopaedics and Traumatology, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China
| | - Wenhua Yuan
- Institute of Orthopaedics and Traumatology, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China
| | - Weidong Wang
- Institute of Orthopaedics and Traumatology, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China.,Department of Orthopedics, The Second Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China
| | - Luwei Xiao
- Institute of Orthopaedics and Traumatology, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China
| | - Xia Lin
- Department of Hepatobiliary and Pancreatic Surgery and Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Di Chen
- Research Center for Human Tissues and Organs Degeneration, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Xin-Hua Feng
- The MOE Key Laboratory of Biosystems Homeostasis & Protection and Zhejiang Provincial Key Laboratory of Cancer Molecular Cell Biology, Life Sciences Institute and
| | - Ping-er Wang
- Institute of Orthopaedics and Traumatology, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China
| | - Peijian Tong
- Institute of Orthopaedics and Traumatology, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China.,Department of Orthopaedic Surgery, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China
| | - Hongting Jin
- Institute of Orthopaedics and Traumatology, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China.,Life Sciences Institute, Zhejiang University, Hangzhou, China
| |
Collapse
|
35
|
Zhuang Y, Lan S, Zhong W, Huang F, Peng J, Zhang S. Comprehensive Analysis of PPMs in Pancreatic Adenocarcinoma Indicates the Value of PPM1K in the Tumor Microenvironment. Cancers (Basel) 2023; 15:cancers15020474. [PMID: 36672423 PMCID: PMC9856814 DOI: 10.3390/cancers15020474] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2022] [Revised: 12/28/2022] [Accepted: 01/04/2023] [Indexed: 01/15/2023] Open
Abstract
Early metastasis and resistance to traditional therapy are responsible for the poor prognosis of pancreatic adenocarcinoma patients. Metal-dependent protein phosphatases (PPMs) have been proven to play a crucial role in the initiation and progression of various tumors. Nevertheless, the expression and function of distinct PPMs in pancreatic adenocarcinoma have not been fully elucidated. In this study, we investigated the mRNA expression level, prognostic value, and the relationship between the expression of PPMs and the tumor microenvironment in pancreatic adenocarcinoma using Oncomine, TCGA and GTEx, GEO, Kaplan-Meier plotter, STRING, GeneMANIA, and HPA databases and R packages. GO and KEGG analysis revealed that PPMs and their differential co-expression genes are attributed to cell-cell adhesion and immune cell infiltration. Among these, PPM1K was downregulated in the tissue and peripheral blood of PAAD patients, whose expression level was negatively related to poor prognosis. Further to this, PPM1K was found to play a role in the epithelial-mesenchymal transition and immune infiltration. ROC curves showed that PPM1K had a good predictive value for pancreatic adenocarcinoma. The knockdown of PPM1K markedly promoted the proliferation and migration of pancreatic cancer cells, confirming its role in tumor suppressor activity in PAAD. This study demonstrates the potential clinical utility of PPM1K in tumor immunotherapy and brings about novel insights into the prognostic value of PPM1K in pancreatic adenocarcinoma.
Collapse
Affiliation(s)
- Yanyan Zhuang
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China
- Department of Gastroenterology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, No. 107, Yanjiangxi Road, Guangzhou 510120, China
| | - Sihua Lan
- Department of Gastroenterology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, No. 107, Yanjiangxi Road, Guangzhou 510120, China
| | - Wa Zhong
- Department of Gastroenterology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, No. 107, Yanjiangxi Road, Guangzhou 510120, China
| | - Fengting Huang
- Department of Gastroenterology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, No. 107, Yanjiangxi Road, Guangzhou 510120, China
| | - Juanfei Peng
- Department of Gastroenterology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, No. 107, Yanjiangxi Road, Guangzhou 510120, China
| | - Shineng Zhang
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China
- Department of Gastroenterology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, No. 107, Yanjiangxi Road, Guangzhou 510120, China
- Correspondence:
| |
Collapse
|
36
|
Jie W, Rui-Fen Z, Zhong-Xiang H, Yan W, Wei-Na L, Yong-Ping M, Jing S, Jing-Yi C, Wan-Hong L, Xiao-Hua H, Zhi L, Yan S. Inhibition of cell proliferation by Tas of foamy viruses through cell cycle arrest or apoptosis underlines the different mechanisms of virus-host interactions. Virulence 2022; 13:342-354. [PMID: 35132916 PMCID: PMC8837258 DOI: 10.1080/21505594.2022.2029329] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Revised: 12/22/2021] [Accepted: 01/11/2022] [Indexed: 11/22/2022] Open
Abstract
Foamy viruses belong to the Spumaretrovirinae subfamily member of the Retroviridae family and produce nonpathogenic infection to hosts in the natural conditions. However, infections of foamy viruses can dramatically cause severe cytopathic effects in vitro. To date, the exact molecular mechanism has remained unclear which implied the tremendous importance of virus-host cell immune reactions. In this study, we found that the transactivator Tas in two foamy viruses isolated from Old World Monkey (OWM) induced obvious inhibition of cell proliferation via the upregulation of Foxo3a expression. It was mediated by the generation of ROS and the initiation of ER stress, and ultimately, the mitochondrial apoptosis pathway was triggered. Notably, PFV Tas contributed to the accumulation of G0/G1 phase cycle arrest induced by the activation of the p53 signaling pathway and the nuclear transportation of HDAC4 via upregulating PPM1E expression. Together, these results demonstrated the different survival strategies by which foamy virus can hijack host cell cytokines and regulate virus-host cell interactions.
Collapse
Affiliation(s)
- Wei Jie
- College of Life Sciences, Shaanxi Normal University, Xi’an, P. R. China
| | - Zhang Rui-Fen
- College of Life Sciences, Shaanxi Normal University, Xi’an, P. R. China
| | - Hu Zhong-Xiang
- College of Life Sciences, Shaanxi Normal University, Xi’an, P. R. China
| | - Wu Yan
- College of Life Sciences, Shaanxi Normal University, Xi’an, P. R. China
| | - Liu Wei-Na
- College of Life Sciences, Shaanxi Normal University, Xi’an, P. R. China
| | - Ma Yong-Ping
- College of Life Sciences, Shaanxi Normal University, Xi’an, P. R. China
| | - Song Jing
- College of Life Sciences, Shaanxi Normal University, Xi’an, P. R. China
| | - Chen Jing-Yi
- College of Life Sciences, Shaanxi Normal University, Xi’an, P. R. China
| | - Liu Wan-Hong
- School of Medicine, Wuhan University, Wuhan, P. R. China
| | - He Xiao-Hua
- School of Medicine, Wuhan University, Wuhan, P. R. China
| | - Li Zhi
- College of Life Sciences, Shaanxi Normal University, Xi’an, P. R. China
| | - Sun Yan
- College of Life Sciences, Shaanxi Normal University, Xi’an, P. R. China
| |
Collapse
|
37
|
Emerging roles of PHLPP phosphatases in the nervous system. Mol Cell Neurosci 2022; 123:103789. [PMID: 36343848 DOI: 10.1016/j.mcn.2022.103789] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Revised: 10/15/2022] [Accepted: 10/22/2022] [Indexed: 11/06/2022] Open
Abstract
It has been more than a decade since the discovery of a novel class of phosphatase, the Pleckstrin Homology (PH) domain Leucine-rich repeat Protein Phosphatases (PHLPP). Over time, they have been recognized as crucial regulators of various cellular processes, such as memory formation, cellular survival and proliferation, maintenance of circadian rhythm, and others, with any deregulation in their expression or cellular localization causing havoc in any cellular system. With the ever-growing number of downstream substrates across multiple tissue systems, a web is emerging wherein the central point is PHLPP. A slight nick in the normal signaling cascade of the two isoforms of PHLPP, namely PHLPP1 and PHLPP2, has been recently found to invoke a variety of neurological disorders including Alzheimer's disease, epileptic seizures, Parkinson's disease, and others, in the neuronal system. Improper regulation of the two isoforms has also been associated with various disease pathologies such as diabetes, cardiovascular disorders, cancer, musculoskeletal disorders, etc. In this review, we have summarized all the current knowledge about PHLPP1 (PHLPP1α and PHLPP1β) and PHLPP2 and their emerging roles in regulating various neuronal signaling pathways to pave the way for a better understanding of the complexities. This would in turn aid in providing context for the development of possible future therapeutic strategies.
Collapse
|
38
|
Wang C, Yao C, Sun Y, Chen J, Ge Y, Wang Y, Wang F, Wang L, Lin Y, Yao S. Identification and verification of a novel epigenetic-related gene signature for predicting the prognosis of hepatocellular carcinoma. Front Genet 2022; 13:897123. [DOI: 10.3389/fgene.2022.897123] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Accepted: 11/14/2022] [Indexed: 12/05/2022] Open
Abstract
Hepatocellular carcinoma (HCC) is a common malignant tumor with a poor prognosis. Epigenetic dysregulation is now considered to be related to hepatocarcinogenesis. However, it is unclear how epigenetic-related genes (ERGs) contribute to the prognosis of HCC. In this study, we used the TCGA database to identify prognostic ERGs that were differentially expressed in HCC patients. Then, using least absolute shrinkage and selection operator (LASSO) regression analysis, a six-gene signature was constructed, and patients were divided into high- and low-risk groups. Validation was performed on HCC patients from the ICGC database. Patients in the high-risk group had a significantly lower chance of survival than those in the low-risk group (p < 0.001 in both databases). The predictive ability of the signature was determined by the receiver operating characteristic (ROC) curve. The risk score was then shown to be an independent prognostic factor for the overall survival (OS) of HCC patients based on the results of univariate and multivariate analyses. We also created a practical nomogram combining the prognostic model with other clinical features. Moreover, functional enrichment analysis revealed that these genes are linked to tumor immunity. In conclusion, our findings showed that a novel six-gene signature related to epigenetics can accurately predict the occurrence and prognosis of HCC.
Collapse
|
39
|
MdPP2C24/37, Protein Phosphatase Type 2Cs from Apple, Interact with MdPYL2/12 to Negatively Regulate ABA Signaling in Transgenic Arabidopsis. Int J Mol Sci 2022; 23:ijms232214375. [PMID: 36430851 PMCID: PMC9696740 DOI: 10.3390/ijms232214375] [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: 10/09/2022] [Revised: 11/15/2022] [Accepted: 11/16/2022] [Indexed: 11/22/2022] Open
Abstract
The phytohormone abscisic acid (ABA) plays an important role in the ability of plants to cope with drought stress. As core members of the ABA signaling pathway, protein phosphatase type 2Cs (PP2Cs) have been reported in many species. However, the functions of MdPP2Cs in apple (Malus domestica) are unclear. In this study, we identified two PP2C-encoding genes, MdPP2C24/37, with conserved PP2C catalytic domains, using sequence alignment. The nucleus-located MdPP2C24/37 genes were induced by ABA or mannitol in apple. Genetic analysis revealed that overexpression of MdPP2C24/37 in Arabidopsis thaliana led to plant insensitivity to ABA or mannitol treatment, in terms of inhibiting seed germination and overall seedling establishment. The expression of stress marker genes was upregulated in MdPP2C24/37 transgenic lines. At the same time, MdPP2C24/37 transgenic lines displayed inhibited ABA-mediated stomatal closure, which led to higher water loss rates. Moreover, when exposed to drought stress, chlorophyll levels decreased and MDA and H2O2 levels accumulated in the MdPP2C24/37 transgenic lines. Further, MdPP2C24/37 interacted with MdPYL2/12 in vitro and vivo. The results indicate that MdPP2C24/37 act as negative regulators in response to ABA-mediated drought resistance.
Collapse
|
40
|
Transcriptome Sequencing Analysis of circRNA in Skeletal Muscle between Fast- and Slow-Growing Chickens at Embryonic Stages. Animals (Basel) 2022; 12:ani12223166. [PMID: 36428392 PMCID: PMC9686870 DOI: 10.3390/ani12223166] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2022] [Revised: 11/10/2022] [Accepted: 11/14/2022] [Indexed: 11/18/2022] Open
Abstract
Skeletal muscle growth has always been the focus of the broiler industry, and circRNAs play a significant role in this process. We collected leg muscles of slow- and fast-growing Bian chicken embryos in the study at 14 (S14 and F14) and 20 (S20 and F20) days for RNA-seq. Finally, 123 and 121 differentially expressed circRNAs (DECs) were identified in S14 vs. F14 and S20 vs. F20, respectively. GO enrichment analysis for DECs obtained important biological process (BP) terms including nicotinate nucleotide biosynthetic process, nicotinate nucleotide salvage, and NAD salvage in S20 vs. F20 and protein mannosylation in S14 vs. F14. KEGG pathway analysis showed Wnt signaling pathway, Tight junction, Ubiquitin mediated proteolysis, and Notch signaling pathway were enriched in the top 20. Based on the GO and KEGG analysis results, we found some significant host genes and circRNAs such as NAPRT and novel_circ_0004547, DVL1 and novel_circ_0003578, JAK2 and novel_circ_0010289, DERA and novel_circ_0003082, etc. Further analysis found 19 co-differentially expressed circRNAs between the two comparison groups. We next constructed a circRNA-miRNA network for them, and some candidate circRNA-miRNA pairs related to skeletal muscle were obtained, such as novel_circ_0002153-miR-12219-5p, novel_circ_0003578-miR-3064-3p, and novel_circ_0010661-miR-12260-3p. These results would help to reveal the mechanism for circRNAs in skeletal muscle and also provide some guidance for the breeding of broilers.
Collapse
|
41
|
Vaneynde P, Verbinnen I, Janssens V. The role of serine/threonine phosphatases in human development: Evidence from congenital disorders. Front Cell Dev Biol 2022; 10:1030119. [PMID: 36313552 PMCID: PMC9608770 DOI: 10.3389/fcell.2022.1030119] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2022] [Accepted: 09/27/2022] [Indexed: 11/23/2022] Open
Abstract
Reversible protein phosphorylation is a fundamental regulation mechanism in eukaryotic cell and organismal physiology, and in human health and disease. Until recently, and unlike protein kinases, mutations in serine/threonine protein phosphatases (PSP) had not been commonly associated with disorders of human development. Here, we have summarized the current knowledge on congenital diseases caused by mutations, inherited or de novo, in one of 38 human PSP genes, encoding a monomeric phosphatase or a catalytic subunit of a multimeric phosphatase. In addition, we highlight similar pathogenic mutations in genes encoding a specific regulatory subunit of a multimeric PSP. Overall, we describe 19 affected genes, and find that most pathogenic variants are loss-of-function, with just a few examples of gain-of-function alterations. Moreover, despite their widespread tissue expression, the large majority of congenital PSP disorders are characterised by brain-specific abnormalities, suggesting a generalized, major role for PSPs in brain development and function. However, even if the pathogenic mechanisms are relatively well understood for a small number of PSP disorders, this knowledge is still incomplete for most of them, and the further identification of downstream targets and effectors of the affected PSPs is eagerly awaited through studies in appropriate in vitro and in vivo disease models. Such lacking studies could elucidate the exact mechanisms through which these diseases act, and possibly open up new therapeutic avenues.
Collapse
Affiliation(s)
- Pieter Vaneynde
- Laboratory of Protein Phosphorylation and Proteomics, Department of Cellular and Molecular Medicine, University of Leuven (KU Leuven), Leuven, Belgium
- Leuven Brain Institute (LBI), Leuven, Belgium
| | - Iris Verbinnen
- Laboratory of Protein Phosphorylation and Proteomics, Department of Cellular and Molecular Medicine, University of Leuven (KU Leuven), Leuven, Belgium
- Leuven Brain Institute (LBI), Leuven, Belgium
| | - Veerle Janssens
- Laboratory of Protein Phosphorylation and Proteomics, Department of Cellular and Molecular Medicine, University of Leuven (KU Leuven), Leuven, Belgium
- Leuven Brain Institute (LBI), Leuven, Belgium
- *Correspondence: Veerle Janssens,
| |
Collapse
|
42
|
Soto-Verdugo J, Siva-Parra J, Hernández-Kelly LC, Ortega A. Acute Manganese Exposure Modifies the Translation Machinery via PI3K/Akt Signaling in Glial Cells. ASN Neuro 2022; 14:17590914221131452. [PMID: 36203371 PMCID: PMC9551334 DOI: 10.1177/17590914221131452] [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] [Indexed: 11/06/2022] Open
Abstract
SUMMARY STATEMENT We demonstrate herein that short-term exposure of radial glia cells to Manganese, a neurotoxic metal, induces an effect on protein synthesis, altering the protein repertoire of these cells.
Collapse
Affiliation(s)
| | | | | | - Arturo Ortega
- Arturo Ortega, Departamento de Toxicología,
Centro de Investigación y de Estudios Avanzados del Instituto Politécnico
Nacional, México City, México, 07360.
| |
Collapse
|
43
|
Tian Y, Liu Y, Wang Q, Wen J, Wu Y, Han J, Man C. Stress-Induced Immunosuppression Affects Immune Response to Newcastle Disease Virus Vaccine via Circulating miRNAs. Animals (Basel) 2022; 12:ani12182376. [PMID: 36139236 PMCID: PMC9495071 DOI: 10.3390/ani12182376] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Revised: 09/06/2022] [Accepted: 09/08/2022] [Indexed: 11/16/2022] Open
Abstract
Simple Summary Circulating miRNAs play important roles in immune response and stress-induced immunosuppression, but the function and mechanism of stress-induced immunosuppression affecting the NDV vaccine immune response remain unknown. In our study, key timepoints, functions, mechanisms, and potential biomarkers of circulating miRNAs involved in immune response and immunosuppression were discovered, providing a theoretical basis for studying the roles of circulating miRNAs in immune regulation. Abstract Studies have shown that circulating microRNAs (miRNAs) are important players in the immune response and stress-induced immunosuppression. However, the function and mechanism of stress-induced immunosuppression affecting the immune response to the Newcastle disease virus (NDV) vaccine remain largely unknown. This study analyzed the changes of 15 NDV-related circulating miRNAs at different immune stages by qRT-PCR, aiming to explore the key timepoints, potential biomarkers, and mechanisms for the functional regulation of candidate circulating miRNAs under immunosuppressed conditions. The results showed that stress-induced immunosuppression induced differential expressions of the candidate circulating miRNAs, especially at 2 days post immunization (dpi), 14 dpi, and 28 dpi. In addition, stress-induced immunosuppression significantly affected the immune response to NDV vaccine, which was manifested by significant changes in candidate circulating miRNAs at 2 dpi, 5 dpi, and 21 dpi. The featured expressions of candidate circulating miRNAs indicated their potential application as biomarkers in immunity and immunosuppression. Bioinformatics analysis revealed that the candidate circulating miRNAs possibly regulated immune function through key targeted genes, such as Mg2+/Mn2+-dependent 1A (PPM1A) and Nemo-like kinase (NLK), in the MAPK signaling pathway. This study provides a theoretical reference for studying the function and mechanism of circulating miRNAs in immune regulation.
Collapse
|
44
|
Sisi C, Jieru D, Peidong C, Zhaolong Z, Yihang W, Shuwen C, Yan T, Tianyu W, Guiyan Y. Transcriptome-wide identification of walnut PP2C family genes in response to external stimulus. BMC Genomics 2022; 23:640. [PMID: 36076184 PMCID: PMC9461273 DOI: 10.1186/s12864-022-08856-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Accepted: 08/23/2022] [Indexed: 11/11/2022] Open
Abstract
Walnut is an important economic tree species while confronting with global environmental stress, resulting in decline in quality and yield. Therefore, it is urgent to elucidate the molecular mechanism for the regulation of walnut response to adversity. The protein phosphatase 2C (PP2C) gene family participates in cellular processes in eukaryotes through reversible phosphorylation of proteins and signal transduction regulation. However, the stress response function of PP2C genes was far to be clarified. Therefore, to understand the stress response mechanism of walnut tree, in this study, a total of 41 PP2C genes with complete ORFs were identified from Juglans regia, whose basic bio-information and expression patterns in response to multiple stresses and ABA were confirmed. The results showed that the ORFs of JrPP2Cs were 495 ~ 3231 bp in length, the predicted JrPP2C proteins contained 164 to 1076 amino acids and the molecular weights were 18,581.96 ~ 118,853.34 Da, the pI was 4.55 ~ 9.58. These JrPP2C genes were unevenly distributed on 14 chromosomes, among which Chr11 and Chr13 contained the most genes. Phylogenetic analysis found that these JrPP2C proteins were classed into 9 subfamilies, among which group F covered most JrPP2Cs. The JrPP2Cs in the same subfamily exhibited similarities in the composition of conserved domains, amino acid sequences of motifs and exon/intron organization in DNA sequences. Each JrPP2C includes 4 ~ 10 motifs and each motif contained 15 ~ 37 amino acids. Among the motifs, motif1, motif2, motif3 and motif8 were most abundant. Most of the JrPP2C genes diversely response to osmotic, cadmium, and Colletotrichum gloeosporioide stress as well as ABA treatments, among which JrPP2C28, JrPP2C17, JrPP2C09, JrPP2C36 were more obvious and deserves further attention. All these results indicated that JrPP2C genes play potential vital roles in plant response to multiple stimulus, and are possibly involved in ABA-dependent signaling pathway.
Collapse
Affiliation(s)
- Chen Sisi
- Labortory of Walnut Research Center, College of Forestry, Northwest A & F University, Yangling, 712100, Shaanxi, China
| | - Deng Jieru
- Labortory of Walnut Research Center, College of Forestry, Northwest A & F University, Yangling, 712100, Shaanxi, China
| | - Cheng Peidong
- Labortory of Walnut Research Center, College of Forestry, Northwest A & F University, Yangling, 712100, Shaanxi, China
| | - Zhang Zhaolong
- Labortory of Walnut Research Center, College of Forestry, Northwest A & F University, Yangling, 712100, Shaanxi, China
| | - Wang Yihang
- Labortory of Walnut Research Center, College of Forestry, Northwest A & F University, Yangling, 712100, Shaanxi, China
| | - Chen Shuwen
- Labortory of Walnut Research Center, College of Forestry, Northwest A & F University, Yangling, 712100, Shaanxi, China
| | - Tang Yan
- Labortory of Walnut Research Center, College of Forestry, Northwest A & F University, Yangling, 712100, Shaanxi, China
| | - Wang Tianyu
- Labortory of Walnut Research Center, College of Forestry, Northwest A & F University, Yangling, 712100, Shaanxi, China
| | - Yang Guiyan
- Labortory of Walnut Research Center, College of Forestry, Northwest A & F University, Yangling, 712100, Shaanxi, China. .,Key Laboratory of Economic Plant Resources Development and Utilization in Shaanxi Province, College of Forestry, Northwest A & F University, Yangling, 712100, Shaanxi, China.
| |
Collapse
|
45
|
Role of serine/threonine protein phosphatase PrpN in the life cycle of Bacillus anthracis. PLoS Pathog 2022; 18:e1010729. [PMID: 35913993 PMCID: PMC9371265 DOI: 10.1371/journal.ppat.1010729] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2021] [Revised: 08/11/2022] [Accepted: 07/07/2022] [Indexed: 11/30/2022] Open
Abstract
Reversible protein phosphorylation at serine/threonine residues is one of the most common protein modifications, widely observed in all kingdoms of life. The catalysts controlling this modification are specific serine/threonine kinases and phosphatases that modulate various cellular pathways ranging from growth to cellular death. Genome sequencing and various omics studies have led to the identification of numerous serine/threonine kinases and cognate phosphatases, yet the physiological relevance of many of these proteins remain enigmatic. In Bacillus anthracis, only one ser/thr phosphatase, PrpC, has been functionally characterized; it was reported to be non-essential for bacterial growth and survival. In the present study, we characterized another ser/thr phosphatase (PrpN) of B. anthracis by various structural and functional approaches. To examine its physiological relevance in B. anthracis, a null mutant strain of prpN was generated and shown to have defects in sporulation and reduced synthesis of toxins (PA and LF) and the toxin activator protein AtxA. We also identified CodY, a global transcriptional regulator, as a target of PrpN and ser/thr kinase PrkC. CodY phosphorylation strongly controlled its binding to the promoter region of atxA, as shown using phosphomimetic and phosphoablative mutants. In nutshell, the present study reports phosphorylation-mediated regulation of CodY activity in the context of anthrax toxin synthesis in B. anthracis by a previously uncharacterized ser/thr protein phosphatase–PrpN. Reversible protein phosphorylation at specific ser/thr residues causes conformational changes in the protein structure, thereby modulating its cellular activity. In B. anthracis, though the role of ser/thr phosphorylation is implicated in various cellular pathways including pathogenesis, till date only one STP (PrpC) has been functionally characterized. This manuscript reports functional characterization of another STP (PrpN) in B. anthracis and with the aid of a null mutant strain (BAS ΔprpN) we provide important insight regarding the role of PrpN in the life cycle of B. anthracis. We have also identified the global transcriptional regulator, CodY as a target of PrpN and PrkC, and for the first time showed the physiological relevance of CodY phosphorylation status in the regulation of anthrax toxin synthesis.
Collapse
|
46
|
Development of Antibody-like Proteins Targeting the Oncogenic Ser/Thr Protein Phosphatase PPM1D. Processes (Basel) 2022. [DOI: 10.3390/pr10081501] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
PPM1D, a protein Ser/Thr phosphatase, is overexpressed in various cancers and functions as an oncogenic protein by inactivating the p53 pathway. Therefore, molecules that bind PPM1D are expected to be useful anti-cancer agents. In this study, we constructed a phage display library based on the antibody-like small molecule protein adnectin and screened for PPM1D-specific binding molecules. We identified two adnectins, PMDB-1 and PMD-24, that bind PPM1D specific B-loop and PPM1D430 as targets, respectively. Specificity analyses of these recombinant proteins using other Ser/Thr protein phosphatases showed that these molecules bind to only PPM1D. Expression of PMDB-1 in breast cancer-derived MCF-7 cells overexpressing endogenous PPM1D stabilized p53, indicating that PMDB-1 functions as an inhibitor of PPM1D. Furthermore, MTT assay exhibited that MCF-7 cells expressing PMDB-1 showed inhibition of cell proliferation. These data suggest that the adnectin PMDB-1 identified in this study can be used as a lead compound for anti-cancer drugs targeting intracellular PPM1D.
Collapse
|
47
|
Bailly C. Anti-inflammatory and anticancer p-terphenyl derivatives from fungi of the genus Thelephora. Bioorg Med Chem 2022; 70:116935. [PMID: 35901638 DOI: 10.1016/j.bmc.2022.116935] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Revised: 06/22/2022] [Accepted: 07/11/2022] [Indexed: 02/08/2023]
Abstract
Fungi from the genus Thelephora have been exploited to identify bioactive compounds. The main natural products characterized are para-terphenyl derivatives, chiefly represented by the lead anti-inflammatory compound vialinin A isolated from species T. vialis and T. terrestris. Different series of p-terphenyls have been identified, including vialinins, ganbajunins, terrestrins, telephantins and other products. Their mechanism of action is not always clearly identified, and different potential molecule targets have been proposed. The lead vialinin A functions as a protease inhibitor, efficiently targeting ubiquitin-specific peptidases USP4/5 and sentrin-specific protease SENP1 which are prominent anti-inflammatory and anticancer targets. Protease inhibition is coupled with a powerful inhibition of the cellular production of tumor necrosis factor TNFα. Other mechanisms contributing to the anti-inflammatory or anti-proliferative action of these p-terphenyl compounds have been invoked, including the formation of cytotoxic copper complexes for derivatives bearing a catechol central unit such vialinin A, terrestrin B and telephantin O. These p-terphenyl compounds could be further exploited to design novel anticancer agents, as evidenced with the parent compound terphenyllin (essentially found in Aspergillus species) which has revealed marked antitumor and anti-metastatic effects in xenograft models of gastric and pancreatic cancer. This review shed light on the structural and functional diversity of p-terphenyls compounds isolated from Thelephora species, their molecular targets and pharmacological properties.
Collapse
Affiliation(s)
- Christian Bailly
- OncoWitan, Scientific Consulting Office, Lille (Wasquehal) 59290, France.
| |
Collapse
|
48
|
Zhang HL, Wang XC, Liu R. Zinc in Regulating Protein Kinases and Phosphatases in Neurodegenerative Diseases. Biomolecules 2022; 12:biom12060785. [PMID: 35740910 PMCID: PMC9220840 DOI: 10.3390/biom12060785] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Revised: 05/27/2022] [Accepted: 06/02/2022] [Indexed: 12/12/2022] Open
Abstract
Zinc is essential for human growth and development. As a trace nutrient, zinc plays important roles in numerous signal transduction pathways involved in distinct physiologic or pathologic processes. Protein phosphorylation is a posttranslational modification which regulates protein activity, degradation, and interaction with other molecules. Protein kinases (PKs) and phosphatases (PPs), with their effects of adding phosphate to or removing phosphate from certain substrates, are master regulators in controlling the phosphorylation of proteins. In this review, we summarize the disturbance of zinc homeostasis and role of zinc disturbance in regulating protein kinases and protein phosphatases in neurodegenerative diseases, with the focus of that in Alzheimer’s disease, providing a new perspective for understanding the mechanisms of these neurologic diseases.
Collapse
|
49
|
Pan J, Zhou L, Zhang C, Xu Q, Sun Y. Targeting protein phosphatases for the treatment of inflammation-related diseases: From signaling to therapy. Signal Transduct Target Ther 2022; 7:177. [PMID: 35665742 PMCID: PMC9166240 DOI: 10.1038/s41392-022-01038-3] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Revised: 04/28/2022] [Accepted: 05/25/2022] [Indexed: 11/09/2022] Open
Abstract
Inflammation is the common pathological basis of autoimmune diseases, metabolic diseases, malignant tumors, and other major chronic diseases. Inflammation plays an important role in tissue homeostasis. On one hand, inflammation can sense changes in the tissue environment, induce imbalance of tissue homeostasis, and cause tissue damage. On the other hand, inflammation can also initiate tissue damage repair and maintain normal tissue function by resolving injury and restoring homeostasis. These opposing functions emphasize the significance of accurate regulation of inflammatory homeostasis to ameliorate inflammation-related diseases. Potential mechanisms involve protein phosphorylation modifications by kinases and phosphatases, which have a crucial role in inflammatory homeostasis. The mechanisms by which many kinases resolve inflammation have been well reviewed, whereas a systematic summary of the functions of protein phosphatases in regulating inflammatory homeostasis is lacking. The molecular knowledge of protein phosphatases, and especially the unique biochemical traits of each family member, will be of critical importance for developing drugs that target phosphatases. Here, we provide a comprehensive summary of the structure, the "double-edged sword" function, and the extensive signaling pathways of all protein phosphatases in inflammation-related diseases, as well as their potential inhibitors or activators that can be used in therapeutic interventions in preclinical or clinical trials. We provide an integrated perspective on the current understanding of all the protein phosphatases associated with inflammation-related diseases, with the aim of facilitating the development of drugs that target protein phosphatases for the treatment of inflammation-related diseases.
Collapse
Affiliation(s)
- Jie Pan
- State Key Laboratory of Pharmaceutical Biotechnology, Chemistry and Biomedicine Innovation Center (ChemBIC), Department of Biotechnology and Pharmaceutical Sciences, School of Life Science, Nanjing University, 163 Xianlin Avenue, Nanjing, 210023, China
| | - Lisha Zhou
- State Key Laboratory of Pharmaceutical Biotechnology, Chemistry and Biomedicine Innovation Center (ChemBIC), Department of Biotechnology and Pharmaceutical Sciences, School of Life Science, Nanjing University, 163 Xianlin Avenue, Nanjing, 210023, China
| | - Chenyang Zhang
- State Key Laboratory of Pharmaceutical Biotechnology, Chemistry and Biomedicine Innovation Center (ChemBIC), Department of Biotechnology and Pharmaceutical Sciences, School of Life Science, Nanjing University, 163 Xianlin Avenue, Nanjing, 210023, China
| | - Qiang Xu
- State Key Laboratory of Pharmaceutical Biotechnology, Chemistry and Biomedicine Innovation Center (ChemBIC), Department of Biotechnology and Pharmaceutical Sciences, School of Life Science, Nanjing University, 163 Xianlin Avenue, Nanjing, 210023, China
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, 221004, Jiangsu, China
| | - Yang Sun
- State Key Laboratory of Pharmaceutical Biotechnology, Chemistry and Biomedicine Innovation Center (ChemBIC), Department of Biotechnology and Pharmaceutical Sciences, School of Life Science, Nanjing University, 163 Xianlin Avenue, Nanjing, 210023, China.
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, 221004, Jiangsu, China.
| |
Collapse
|
50
|
Grimm TM, Herbinger M, Krüger L, Müller S, Mayer TU, Hauck CR. Lockdown, a selective small-molecule inhibitor of the integrin phosphatase PPM1F, blocks cancer cell invasion. Cell Chem Biol 2022; 29:930-946.e9. [PMID: 35443151 DOI: 10.1016/j.chembiol.2022.03.011] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Revised: 03/04/2022] [Accepted: 03/23/2022] [Indexed: 12/18/2022]
Abstract
Phosphatase PPM1F is a regulator of cell adhesion by fine-tuning integrin activity and actin cytoskeleton structures. Elevated expression of this enzyme in human tumors is associated with high invasiveness, enhanced metastasis, and poor prognosis. Thus, PPM1F is a target for pharmacological intervention, yet inhibitors of this enzyme are lacking. Here, we use high-throughput screening to identify Lockdown, a reversible and non-competitive PPM1F inhibitor. Lockdown is selective for PPM1F, because this compound does not inhibit other protein phosphatases in vitro and does not induce additional phenotypes in PPM1F knockout cells. Importantly, Lockdown-treated glioblastoma cells fully re-capitulate the phenotype of PPM1F-deficient cells as assessed by increased phosphorylation of PPM1F substrates and corruption of integrin-dependent cellular processes. Ester modification yields LockdownPro with increased membrane permeability and prodrug-like properties. LockdownPro suppresses tissue invasion by PPM1F-overexpressing human cancer cells, validating PPM1F as a therapeutic target and providing an access point to control tumor cell dissemination.
Collapse
Affiliation(s)
- Tanja M Grimm
- Lehrstuhl Zellbiologie, Department of Biology, University of Konstanz, Maildrop 621, Universitätsstrasse 10, 78467 Konstanz, Germany; Konstanz Research School Chemical Biology, University of Konstanz, Universitätsstrasse 10, 78467 Konstanz, Germany
| | - Marleen Herbinger
- Lehrstuhl Zellbiologie, Department of Biology, University of Konstanz, Maildrop 621, Universitätsstrasse 10, 78467 Konstanz, Germany
| | - Lena Krüger
- Department of Chemistry, University of Konstanz, Universitätsstrasse 10, 78467 Konstanz, Germany; Konstanz Research School Chemical Biology, University of Konstanz, Universitätsstrasse 10, 78467 Konstanz, Germany
| | - Silke Müller
- Lehrstuhl Molekulare Genetik, Department of Biology, University of Konstanz, Universitätsstrasse 10, 78467 Konstanz, Germany; Screening Center, Department of Biology, University of Konstanz, Universitätsstrasse 10, 78467 Konstanz, Germany
| | - Thomas U Mayer
- Lehrstuhl Molekulare Genetik, Department of Biology, University of Konstanz, Universitätsstrasse 10, 78467 Konstanz, Germany; Screening Center, Department of Biology, University of Konstanz, Universitätsstrasse 10, 78467 Konstanz, Germany; Konstanz Research School Chemical Biology, University of Konstanz, Universitätsstrasse 10, 78467 Konstanz, Germany
| | - Christof R Hauck
- Lehrstuhl Zellbiologie, Department of Biology, University of Konstanz, Maildrop 621, Universitätsstrasse 10, 78467 Konstanz, Germany; Konstanz Research School Chemical Biology, University of Konstanz, Universitätsstrasse 10, 78467 Konstanz, Germany.
| |
Collapse
|