1
|
Hu B, Gong H, Nie L, Zhang J, Li Y, Liu D, Zhang H, Zhang H, Han L, Yang C, Li M, Xu W, Nakamura Y, Shi L, Ye M, Hillyer CD, Mohandas N, Liang L, Sheng Y, Liu J. Lysine succinylation precisely controls normal erythropoiesis. Haematologica 2025; 110:397-413. [PMID: 39415677 PMCID: PMC11788629 DOI: 10.3324/haematol.2024.285752] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Accepted: 10/04/2024] [Indexed: 10/19/2024] Open
Abstract
Lysine succinylation (Ksu) has recently emerged as a protein modification that regulates diverse functions in various biological processes. However, the systemic, precise role of lysine succinylation in erythropoiesis remains to be fully elucidated. In this study, we noted a prominent increase of succinyl-CoA and lysine succinylation during human erythroid differentiation. To explore the functional significance of succinylation, we inhibited succinylation by either knocking down key succinyltransferases or overexpressing desuccinylases. Succinylation inhibition led to suppressed cell proliferation, increased apoptosis, and disrupted erythroid differentiation. In vivo overexpression of the desuccinylase SIRT5 delayed erythroid differentiation. Furthermore, integrative proteome and succinylome analysis identified 939 succinylated proteins with 3,562 Ksu sites, distributed across various cellular compartments and involved in multiple cellular processes. Significantly, inconsistencies were observed between protein expression levels and succinylation levels, indicating that the succinylation of certain proteins may function independently of expression. Mechanistically, we implicated KAT2A-mediated succinylation of histone H3 K79, leading to chromatin remodeling and, subsequently, regulation of erythropoiesis. Specifically, we identified CYCS as a key regulator of erythropoiesis, a function that depends on its succinylation sites K28/K40. Taken together, our comprehensive investigation of the succinylation landscape during erythropoiesis provides valuable insights into its regulatory role and offers potential implications for erythroid-related diseases.
Collapse
Affiliation(s)
- Bin Hu
- Department of Hematology, The Second Xiangya Hospital, Molecular Biology Research Center, School of Life Sciences, Hunan Province Key Laboratory of Basic and Applied Hematology, Central South University, Hunan
| | - Han Gong
- Department of Hematology, The Second Xiangya Hospital, Molecular Biology Research Center, School of Life Sciences, Hunan Province Key Laboratory of Basic and Applied Hematology, Central South University, Hunan
| | - Ling Nie
- Department of Hematology, Xiangya Hospital, Central South University, Hunan
| | - Ji Zhang
- The Affiliated Nanhua Hospital, Department of Clinical Laboratory, Hengyang Medical School, University of South China, Hunan
| | - Yanan Li
- Department of Hematology, The Second Xiangya Hospital, Molecular Biology Research Center, School of Life Sciences, Hunan Province Key Laboratory of Basic and Applied Hematology, Central South University, Hunan
| | - Dandan Liu
- Department of Hematology, The Second Xiangya Hospital, Molecular Biology Research Center, School of Life Sciences, Hunan Province Key Laboratory of Basic and Applied Hematology, Central South University, Hunan
| | - Huifang Zhang
- Department of Hematology, The Second Xiangya Hospital, Molecular Biology Research Center, School of Life Sciences, Hunan Province Key Laboratory of Basic and Applied Hematology, Central South University, Hunan
| | - Haihang Zhang
- Department of Hematology, The Second Xiangya Hospital, Molecular Biology Research Center, School of Life Sciences, Hunan Province Key Laboratory of Basic and Applied Hematology, Central South University, Hunan
| | - Lu Han
- Department of Hematology, The Second Xiangya Hospital, Molecular Biology Research Center, School of Life Sciences, Hunan Province Key Laboratory of Basic and Applied Hematology, Central South University, Hunan
| | - Chaoying Yang
- Department of Hematology, The Second Xiangya Hospital, Molecular Biology Research Center, School of Life Sciences, Hunan Province Key Laboratory of Basic and Applied Hematology, Central South University, Hunan
| | - Maohua Li
- Department of Hematology, The Second Xiangya Hospital, Molecular Biology Research Center, School of Life Sciences, Hunan Province Key Laboratory of Basic and Applied Hematology, Central South University, Hunan
| | - Wenwen Xu
- Department of Hematology, The Second Xiangya Hospital, Molecular Biology Research Center, School of Life Sciences, Hunan Province Key Laboratory of Basic and Applied Hematology, Central South University, Hunan
| | - Yukio Nakamura
- Cell Engineering Division, RIKEN BioResource Center, Tsukuba, Ibaraki
| | - Lihong Shi
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College; CAMS Center for Stem Cell Medicine, PUMC Department of Stem Cell and Regenerative Medicine, Tianjin
| | - Mao Ye
- Molecular Science and Biomedicine Laboratory (MBL), State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Biology, College of Chemistry and Chemical Engineering, Aptamer Engineering Center of Hunan Province, Hunan University, Hunan
| | | | - Narla Mohandas
- Research Laboratory of Red Cell Physiology, New York Blood Center, New York
| | - Long Liang
- Department of Hematology, The Second Xiangya Hospital, Molecular Biology Research Center, School of Life Sciences, Hunan Province Key Laboratory of Basic and Applied Hematology, Central South University, Hunan.
| | - Yue Sheng
- Department of Hematology, The Second Xiangya Hospital, Molecular Biology Research Center, School of Life Sciences, Hunan Province Key Laboratory of Basic and Applied Hematology, Central South University, Hunan.
| | - Jing Liu
- Department of Hematology, The Second Xiangya Hospital, Molecular Biology Research Center, School of Life Sciences, Hunan Province Key Laboratory of Basic and Applied Hematology, Central South University, Hunan.
| |
Collapse
|
2
|
Yang J, Pan C, Pan Y, Hu A, Zhao P, Chen M, Song H, Li Y, Hao X. A Carbon 21 Steroidal Glycoside with Pregnane Skeleton from Cynanchum atratum Bunge Promotes Megakaryocytic and Erythroid Differentiation in Erythroleukemia HEL Cells through Regulating Platelet-Derived Growth Factor Receptor Beta and JAK2/STAT3 Pathway. Pharmaceuticals (Basel) 2024; 17:628. [PMID: 38794198 PMCID: PMC11125340 DOI: 10.3390/ph17050628] [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: 03/12/2024] [Revised: 04/25/2024] [Accepted: 04/26/2024] [Indexed: 05/26/2024] Open
Abstract
Erythroleukemia is a rare form of acute myeloid leukemia (AML). Its molecular pathogenesis remains vague, and this disease has no specific therapeutic treatments. Previously, our group isolated a series of Carbon 21 (C-21) steroidal glycosides with pregnane skeleton from the root of Cynanchum atratum Bunge. Among them, we found that a compound, named BW18, can induce S-phase cell cycle arrest and apoptosis via the mitogen-activated protein kinase (MAPK) pathway in human chronic myeloid leukemia K562 cells. However, its anti-tumor activity against erythroleukemia remains largely unknown. In this study, we aimed to investigate the anti-erythroleukemia activity of BW18 and the underlying molecular mechanisms. Our results demonstrated that BW18 exhibited a good anti-erythroleukemia activity in the human erythroleukemia cell line HEL and an in vivo xenograft mouse model. In addition, BW18 induced cell cycle arrest at the G2/M phase and promoted megakaryocytic and erythroid differentiation in HEL cells. Furthermore, RNA sequencing (RNA-seq) and rescue assay demonstrated that overexpression of platelet-derived growth factor receptor beta (PDGFRB) reversed BW18-induced megakaryocytic differentiation in HEL cells, but not erythroid differentiation. In addition, the network pharmacology analysis, the molecular docking and cellular thermal shift assay (CETSA) revealed that BW18 could inactivate Janus tyrosine kinase 2 (JAK2)/signal transducer and activator of transcription 3 (STAT3) pathway, which might mediate BW18-induced erythroid differentiation. Taken together, our findings elucidated a novel role of PDGFRB in regulating erythroleukemia differentiation and highlighted BW18 as an attractive lead compound for erythroleukemia treatment.
Collapse
Affiliation(s)
- Jue Yang
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang 561113, China; (J.Y.); (C.P.); (Y.P.); (A.H.); (P.Z.); (M.C.)
- Key Laboratory of Endemic and Ethnic Diseases, Ministry of Education, Key Laboratory of Medical Molecular Biology of Guizhou Province, Guizhou Medical University, Guiyang 561113, China
- Natural Products Research Center of Guizhou Province, Guiyang 550014, China
- School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang 561113, China
| | - Chaolan Pan
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang 561113, China; (J.Y.); (C.P.); (Y.P.); (A.H.); (P.Z.); (M.C.)
- Key Laboratory of Endemic and Ethnic Diseases, Ministry of Education, Key Laboratory of Medical Molecular Biology of Guizhou Province, Guizhou Medical University, Guiyang 561113, China
- Natural Products Research Center of Guizhou Province, Guiyang 550014, China
- School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang 561113, China
| | - Yang Pan
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang 561113, China; (J.Y.); (C.P.); (Y.P.); (A.H.); (P.Z.); (M.C.)
- Key Laboratory of Endemic and Ethnic Diseases, Ministry of Education, Key Laboratory of Medical Molecular Biology of Guizhou Province, Guizhou Medical University, Guiyang 561113, China
- Natural Products Research Center of Guizhou Province, Guiyang 550014, China
- School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang 561113, China
| | - Anlin Hu
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang 561113, China; (J.Y.); (C.P.); (Y.P.); (A.H.); (P.Z.); (M.C.)
- Key Laboratory of Endemic and Ethnic Diseases, Ministry of Education, Key Laboratory of Medical Molecular Biology of Guizhou Province, Guizhou Medical University, Guiyang 561113, China
- Natural Products Research Center of Guizhou Province, Guiyang 550014, China
| | - Peng Zhao
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang 561113, China; (J.Y.); (C.P.); (Y.P.); (A.H.); (P.Z.); (M.C.)
- Key Laboratory of Endemic and Ethnic Diseases, Ministry of Education, Key Laboratory of Medical Molecular Biology of Guizhou Province, Guizhou Medical University, Guiyang 561113, China
- Natural Products Research Center of Guizhou Province, Guiyang 550014, China
- School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang 561113, China
| | - Meijun Chen
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang 561113, China; (J.Y.); (C.P.); (Y.P.); (A.H.); (P.Z.); (M.C.)
- Key Laboratory of Endemic and Ethnic Diseases, Ministry of Education, Key Laboratory of Medical Molecular Biology of Guizhou Province, Guizhou Medical University, Guiyang 561113, China
- Natural Products Research Center of Guizhou Province, Guiyang 550014, China
- School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang 561113, China
| | - Hui Song
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang 561113, China; (J.Y.); (C.P.); (Y.P.); (A.H.); (P.Z.); (M.C.)
- Key Laboratory of Endemic and Ethnic Diseases, Ministry of Education, Key Laboratory of Medical Molecular Biology of Guizhou Province, Guizhou Medical University, Guiyang 561113, China
| | - Yanmei Li
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang 561113, China; (J.Y.); (C.P.); (Y.P.); (A.H.); (P.Z.); (M.C.)
- Key Laboratory of Endemic and Ethnic Diseases, Ministry of Education, Key Laboratory of Medical Molecular Biology of Guizhou Province, Guizhou Medical University, Guiyang 561113, China
- Natural Products Research Center of Guizhou Province, Guiyang 550014, China
- School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang 561113, China
| | - Xiaojiang Hao
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang 561113, China; (J.Y.); (C.P.); (Y.P.); (A.H.); (P.Z.); (M.C.)
- Key Laboratory of Endemic and Ethnic Diseases, Ministry of Education, Key Laboratory of Medical Molecular Biology of Guizhou Province, Guizhou Medical University, Guiyang 561113, China
- Natural Products Research Center of Guizhou Province, Guiyang 550014, China
- School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang 561113, China
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming 650204, China
| |
Collapse
|
3
|
Ma JK, Su LD, Feng LL, Li JL, Pan L, Danzeng Q, Li Y, Shang T, Zhan XL, Chen SY, Ying S, Hu JR, Chen XQ, Zhang Q, Liang T, Lu XJ. TFPI from erythroblasts drives heme production in central macrophages promoting erythropoiesis in polycythemia. Nat Commun 2024; 15:3976. [PMID: 38729948 PMCID: PMC11087540 DOI: 10.1038/s41467-024-48328-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Accepted: 04/26/2024] [Indexed: 05/12/2024] Open
Abstract
Bleeding and thrombosis are known as common complications of polycythemia for a long time. However, the role of coagulation system in erythropoiesis is unclear. Here, we discover that an anticoagulant protein tissue factor pathway inhibitor (TFPI) plays an essential role in erythropoiesis via the control of heme biosynthesis in central macrophages. TFPI levels are elevated in erythroblasts of human erythroblastic islands with JAK2V617F mutation and hypoxia condition. Erythroid lineage-specific knockout TFPI results in impaired erythropoiesis through decreasing ferrochelatase expression and heme biosynthesis in central macrophages. Mechanistically, the TFPI interacts with thrombomodulin to promote the downstream ERK1/2-GATA1 signaling pathway to induce heme biosynthesis in central macrophages. Furthermore, TFPI blockade impairs human erythropoiesis in vitro, and normalizes the erythroid compartment in mice with polycythemia. These results show that erythroblast-derived TFPI plays an important role in the regulation of erythropoiesis and reveal an interplay between erythroblasts and central macrophages.
Collapse
Affiliation(s)
- Jun-Kai Ma
- Department of Physiology and Department of Hepatobiliary and Pancreatic Surgery of the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310058, China
| | - Li-Da Su
- Neuroscience Care Unit, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, 310009, China
| | - Lin-Lin Feng
- Department of Physiology and Department of Hepatobiliary and Pancreatic Surgery of the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310058, China
- State Key Laboratory for Quality and Safety of Agro-products, Ningbo University, Ningbo, 315211, China
| | - Jing-Lin Li
- Department of Physiology and Department of Hepatobiliary and Pancreatic Surgery of the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310058, China
| | - Li Pan
- The General Hospital of Tibet Military Area Command, Lhasa, China
| | - Qupei Danzeng
- Department of Tibetan Medicine; University of Tibetan Medicine, Lhasa, 540100, China
| | - Yanwei Li
- Core Facilities, Zhejiang University School of Medicine, Hangzhou, China
| | - Tongyao Shang
- Department of Physiology and Department of Hepatobiliary and Pancreatic Surgery of the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310058, China
| | - Xiao-Lin Zhan
- Department of Physiology and Department of Hepatobiliary and Pancreatic Surgery of the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310058, China
- State Key Laboratory for Quality and Safety of Agro-products, Ningbo University, Ningbo, 315211, China
| | - Si-Ying Chen
- Department of Physiology and Department of Hepatobiliary and Pancreatic Surgery of the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310058, China
- State Key Laboratory for Quality and Safety of Agro-products, Ningbo University, Ningbo, 315211, China
| | - Shibo Ying
- School of Public Health, Hangzhou Medical College, Hangzhou, 310013, China
| | - Jian-Rao Hu
- School of Life and Environmental Sciences, Hangzhou Normal University, Hangzhou, 311121, Zhejiang, China
| | - Xue Qun Chen
- Zhejiang University, School of Brain Science and Brain Medicine, Hangzhou, China
| | - Qi Zhang
- Department of Hepatobiliary and Pancreatic Surgery, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310058, China.
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310058, China.
| | - Tingbo Liang
- Department of Hepatobiliary and Pancreatic Surgery, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310058, China.
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310058, China.
| | - Xin-Jiang Lu
- Department of Physiology and Department of Hepatobiliary and Pancreatic Surgery of the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310058, China.
| |
Collapse
|
4
|
Meng Y, Lin W, Wang N, Wei X, Mei P, Wang X, Zhang C, Huang Q, Liao Y. USP7-mediated ERβ stabilization mitigates ROS accumulation and promotes osimertinib resistance by suppressing PRDX3 SUMOylation in non-small cell lung carcinoma. Cancer Lett 2024; 582:216587. [PMID: 38097136 DOI: 10.1016/j.canlet.2023.216587] [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: 09/15/2023] [Revised: 11/27/2023] [Accepted: 12/04/2023] [Indexed: 12/20/2023]
Abstract
Osimertinib resistance is regarded as a major obstacle limiting survival benefits for patients undergoing treatment of epidermal growth factor receptor (EGFR)-mutant non-small cell lung cancer (NSCLC). However, the underlying mechanisms of acquired resistance remain unclear. In this study, we report that estrogen receptor β (ERβ) is highly expressed in osimertinib-resistant NSCLC and plays a pivotal role in promoting osimertinib resistance. We further identified ubiquitin-specific protease 7 (USP7) as a critical binding partner that deubiquitinates and upregulates ERβ in NSCLC. ERβ promotes osimertinib resistance by mitigating reactive oxygen species (ROS) accumulation. We found that ERβ mechanistically suppresses peroxiredoxin 3 (PRDX3) SUMOylation and thus confers osimertinib resistance onto NSCLC. Furthermore, we provide evidence showing that depletion of ERβ induces ROS accumulation and reverses osimertinib resistance in NSCLC both in vitro and in vivo. Thus, our results demonstrate that USP7-mediated ERβ stabilization suppresses PRDX3 SUMOylation to mitigate ROS accumulation and promote osimertinib resistance, suggesting that targeting ERβ may be an effective therapeutic strategy to overcome osimertinib resistance in NSCLC.
Collapse
Affiliation(s)
- Yunchong Meng
- Department of Thoracic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430022, China
| | - Wei Lin
- Department of Thoracic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430022, China
| | - Na Wang
- Department of Pathology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430022, China
| | - Xiao Wei
- Cancer Biology Research Center (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China
| | - Peiyuan Mei
- Department of Thoracic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430022, China
| | - Xiaojun Wang
- Department of Thoracic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430022, China
| | - Chi Zhang
- Department of Thoracic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430022, China
| | - Quanfu Huang
- Department of Thoracic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430022, China.
| | - Yongde Liao
- Department of Thoracic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430022, China.
| |
Collapse
|
5
|
Park HB, Baek KH. Current and future directions of USP7 interactome in cancer study. Biochim Biophys Acta Rev Cancer 2023; 1878:188992. [PMID: 37775071 DOI: 10.1016/j.bbcan.2023.188992] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Revised: 09/14/2023] [Accepted: 09/23/2023] [Indexed: 10/01/2023]
Abstract
The ubiquitin-proteasome system (UPS) is an essential protein quality controller for regulating protein homeostasis and autophagy. Ubiquitination is a protein modification process that involves the binding of one or more ubiquitins to substrates through a series of enzymatic processes. These include ubiquitin-activating enzymes (E1), ubiquitin-conjugating enzymes (E2), and ubiquitin ligases (E3). Conversely, deubiquitination is a reverse process that removes ubiquitin from substrates via deubiquitinating enzymes (DUBs). Dysregulation of ubiquitination-related enzymes can lead to various human diseases, including cancer, through the modulation of protein ubiquitination. The most structurally and functionally studied DUB is the ubiquitin-specific protease 7 (USP7). Both the TRAF and UBL domains of USP7 are known to bind to the [P/A/E]-X-X-S or K-X-X-X-K motif of substrates. USP7 has been shown to be involved in cancer pathogenesis by binding with numerous substrates. Recently, a novel substrate of USP7 was discovered through a systemic analysis of its binding motif. This review summarizes the currently discovered substrates and cellular functions of USP7 in cancer and suggests putative substrates of USP7 through a comprehensive systemic analysis.
Collapse
Affiliation(s)
- Hong-Beom Park
- Department of Convergence, CHA University, Gyeonggi-Do 13488, Republic of Korea
| | - Kwang-Hyun Baek
- Department of Convergence, CHA University, Gyeonggi-Do 13488, Republic of Korea; International Ubiquitin Center(,) CHA University, Gyeonggi-Do 13488, Republic of Korea.
| |
Collapse
|
6
|
de Carvalho LGA, Komoto TT, Moreno DA, Goes JVC, de Oliveira RTG, de Lima Melo MM, Roa MEGV, Gonçalves PG, Montefusco-Pereira CV, Pinheiro RF, Ribeiro Junior HL. USP15-USP7 Axis and UBE2T Differential Expression May Predict Pathogenesis and Poor Prognosis in De Novo Myelodysplastic Neoplasm. Int J Mol Sci 2023; 24:10058. [PMID: 37373211 PMCID: PMC10298103 DOI: 10.3390/ijms241210058] [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: 04/05/2023] [Revised: 04/17/2023] [Accepted: 04/20/2023] [Indexed: 06/29/2023] Open
Abstract
The aim of this study was to evaluate the expression of USP7, USP15, UBE2O, and UBE2T genes in Myelodysplastic neoplasm (MDS) to identify possible targets of ubiquitination and deubiquitination in MDS pathobiology. To achieve this, eight datasets from the Gene Expression Omnibus (GEO) database were integrated, and the expression relationship of these genes was analyzed in 1092 MDS patients and healthy controls. Our results showed that UBE2O, UBE2T, and USP7 were upregulated in MDS patients compared with healthy individuals, but only in mononucleated cells collected from bone marrow samples (p < 0.001). In contrast, only the USP15 gene showed a downregulated expression compared with healthy individuals (p = 0.03). Additionally, the upregulation of UBE2T expression was identified in MDS patients with chromosomal abnormalities compared with patients with normal karyotypes (p = 0.0321), and the downregulation of UBE2T expression was associated with MDS hypoplastic patients (p = 0.033). Finally, the USP7 and USP15 genes were strongly correlated with MDS (r = 0.82; r2 = 0.67; p < 0.0001). These findings suggest that the differential expression of the USP15-USP7 axis and UBE2T may play an important role in controlling genomic instability and the chromosomal abnormalities that are a striking characteristic of MDS.
Collapse
Affiliation(s)
- Luiz Gustavo Almeida de Carvalho
- Center for Research and Drug Development (NPDM), Federal University of Ceara, Fortaleza 60020-181, CE, Brazil; (L.G.A.d.C.); (J.V.C.G.); (M.M.d.L.M.); (C.V.M.-P.); (R.F.P.)
- Post-Graduate Program in Translational Medicine, Federal University of Ceara, Fortaleza 60020-181, CE, Brazil
| | - Tatiana Takahasi Komoto
- Molecular Oncology Research Center, Barretos Cancer Hospital, Barretos 14784-390, SP, Brazil; (T.T.K.); (D.A.M.); (P.G.G.)
| | - Daniel Antunes Moreno
- Molecular Oncology Research Center, Barretos Cancer Hospital, Barretos 14784-390, SP, Brazil; (T.T.K.); (D.A.M.); (P.G.G.)
| | - João Vitor Caetano Goes
- Center for Research and Drug Development (NPDM), Federal University of Ceara, Fortaleza 60020-181, CE, Brazil; (L.G.A.d.C.); (J.V.C.G.); (M.M.d.L.M.); (C.V.M.-P.); (R.F.P.)
- Post-Graduate Program of Pathology, Federal University of Ceara, Fortaleza 60020-181, CE, Brazil
| | - Roberta Taiane Germano de Oliveira
- Center for Research and Drug Development (NPDM), Federal University of Ceara, Fortaleza 60020-181, CE, Brazil; (L.G.A.d.C.); (J.V.C.G.); (M.M.d.L.M.); (C.V.M.-P.); (R.F.P.)
- Post-Graduate Program in Medical Science, Federal University of Ceara, Fortaleza 60020-181, CE, Brazil
| | - Mayara Magna de Lima Melo
- Center for Research and Drug Development (NPDM), Federal University of Ceara, Fortaleza 60020-181, CE, Brazil; (L.G.A.d.C.); (J.V.C.G.); (M.M.d.L.M.); (C.V.M.-P.); (R.F.P.)
- Post-Graduate Program in Medical Science, Federal University of Ceara, Fortaleza 60020-181, CE, Brazil
| | | | - Paola Gyuliane Gonçalves
- Molecular Oncology Research Center, Barretos Cancer Hospital, Barretos 14784-390, SP, Brazil; (T.T.K.); (D.A.M.); (P.G.G.)
- Department of Pathology, School of Medicine, Universidade Estadual Paulista, Botucatu 18618-970, SP, Brazil
| | - Carlos Victor Montefusco-Pereira
- Center for Research and Drug Development (NPDM), Federal University of Ceara, Fortaleza 60020-181, CE, Brazil; (L.G.A.d.C.); (J.V.C.G.); (M.M.d.L.M.); (C.V.M.-P.); (R.F.P.)
| | - Ronald Feitosa Pinheiro
- Center for Research and Drug Development (NPDM), Federal University of Ceara, Fortaleza 60020-181, CE, Brazil; (L.G.A.d.C.); (J.V.C.G.); (M.M.d.L.M.); (C.V.M.-P.); (R.F.P.)
- Post-Graduate Program in Translational Medicine, Federal University of Ceara, Fortaleza 60020-181, CE, Brazil
- Post-Graduate Program of Pathology, Federal University of Ceara, Fortaleza 60020-181, CE, Brazil
- Post-Graduate Program in Medical Science, Federal University of Ceara, Fortaleza 60020-181, CE, Brazil
| | - Howard Lopes Ribeiro Junior
- Center for Research and Drug Development (NPDM), Federal University of Ceara, Fortaleza 60020-181, CE, Brazil; (L.G.A.d.C.); (J.V.C.G.); (M.M.d.L.M.); (C.V.M.-P.); (R.F.P.)
- Post-Graduate Program in Translational Medicine, Federal University of Ceara, Fortaleza 60020-181, CE, Brazil
- Molecular Oncology Research Center, Barretos Cancer Hospital, Barretos 14784-390, SP, Brazil; (T.T.K.); (D.A.M.); (P.G.G.)
- Post-Graduate Program in Medical Science, Federal University of Ceara, Fortaleza 60020-181, CE, Brazil
| |
Collapse
|
7
|
Angiotensin-converting enzyme inhibitor promotes angiogenesis through Sp1/Sp3-mediated inhibition of notch signaling in male mice. Nat Commun 2023; 14:731. [PMID: 36759621 PMCID: PMC9911748 DOI: 10.1038/s41467-023-36409-z] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Accepted: 01/31/2023] [Indexed: 02/11/2023] Open
Abstract
Angiogenesis is a critical pathophysiological process involved in organ growth and various diseases. Transcription factors Sp1/Sp3 are necessary for fetal development and tumor growth. Sp1/Sp3 proteins were downregulated in the capillaries of the gastrocnemius in patients with critical limb ischemia samples. Endothelial-specific Sp1/Sp3 knockout reduces angiogenesis in retinal, pathological, and tumor models and induced activation of the Notch1 pathway. Further, the inactivation of VEGFR2 signaling by Notch1 contributes to the delayed angiogenesis phenotype. Mechanistically, endothelial Sp1 binds to the promoter of Notch1 and inhibits its transcription, which is enhanced by Sp3. The proangiogenic effect of ACEI is abolished in Sp1/Sp3-deletion male mice. We identify USP7 as an ACEI-activated deubiquitinating enzyme that translocated into the nucleus binding to Sp1/Sp3, which are deacetylated by HDAC1. Our findings demonstrate a central role for endothelial USP7-Sp1/Sp3-Notch1 signaling in pathophysiological angiogenesis in response to ACEI treatment.
Collapse
|
8
|
Sherpa D, Mueller J, Karayel Ö, Xu P, Yao Y, Chrustowicz J, Gottemukkala KV, Baumann C, Gross A, Czarnecki O, Zhang W, Gu J, Nilvebrant J, Sidhu SS, Murray PJ, Mann M, Weiss MJ, Schulman BA, Alpi AF. Modular UBE2H-CTLH E2-E3 complexes regulate erythroid maturation. eLife 2022; 11:e77937. [PMID: 36459484 PMCID: PMC9718529 DOI: 10.7554/elife.77937] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Accepted: 11/24/2022] [Indexed: 12/05/2022] Open
Abstract
The development of haematopoietic stem cells into mature erythrocytes - erythropoiesis - is a controlled process characterized by cellular reorganization and drastic reshaping of the proteome landscape. Failure of ordered erythropoiesis is associated with anaemias and haematological malignancies. Although the ubiquitin system is a known crucial post-translational regulator in erythropoiesis, how the erythrocyte is reshaped by the ubiquitin system is poorly understood. By measuring the proteomic landscape of in vitro human erythropoiesis models, we found dynamic differential expression of subunits of the CTLH E3 ubiquitin ligase complex that formed maturation stage-dependent assemblies of topologically homologous RANBP9- and RANBP10-CTLH complexes. Moreover, protein abundance of CTLH's cognate E2 ubiquitin conjugating enzyme UBE2H increased during terminal differentiation, and UBE2H expression depended on catalytically active CTLH E3 complexes. CRISPR-Cas9-mediated inactivation of CTLH E3 assemblies or UBE2H in erythroid progenitors revealed defects, including spontaneous and accelerated erythroid maturation as well as inefficient enucleation. Thus, we propose that dynamic maturation stage-specific changes of UBE2H-CTLH E2-E3 modules control the orderly progression of human erythropoiesis.
Collapse
Affiliation(s)
- Dawafuti Sherpa
- Department of Molecular Machines and Signaling, Max Planck Institute of BiochemistryMartinsriedGermany
| | - Judith Mueller
- Department of Molecular Machines and Signaling, Max Planck Institute of BiochemistryMartinsriedGermany
| | - Özge Karayel
- Department of Proteomics and Signal Transduction, Max Planck Institute of BiochemistryMartinsriedGermany
| | - Peng Xu
- Cyrus Tang Medical Institute, National Clinical Research Centre for Hematologic Diseases, Collaborative Innovation Centre of Hematology, State Key Laboratory of Radiation Medicine and Protection, Soochow UniversitySuzhouChina
- Department of Hematology, St. Jude Children’s Research HospitalMemphisUnited States
| | - Yu Yao
- Department of Hematology, St. Jude Children’s Research HospitalMemphisUnited States
| | - Jakub Chrustowicz
- Department of Molecular Machines and Signaling, Max Planck Institute of BiochemistryMartinsriedGermany
| | - Karthik V Gottemukkala
- Department of Molecular Machines and Signaling, Max Planck Institute of BiochemistryMartinsriedGermany
| | - Christine Baumann
- Department of Molecular Machines and Signaling, Max Planck Institute of BiochemistryMartinsriedGermany
| | - Annette Gross
- Department of Molecular Machines and Signaling, Max Planck Institute of BiochemistryMartinsriedGermany
- Department of Immunoregulation, Max Planck Institute of BiochemistryMartinsriedGermany
| | - Oliver Czarnecki
- Department of Molecular Machines and Signaling, Max Planck Institute of BiochemistryMartinsriedGermany
| | - Wei Zhang
- Donnelly Centre for Cellular and Biomolecular Research, University of TorontoTorontoCanada
| | - Jun Gu
- Donnelly Centre for Cellular and Biomolecular Research, University of TorontoTorontoCanada
- Department of Molecular Genetics, University of TorontoTorontoCanada
| | - Johan Nilvebrant
- Donnelly Centre for Cellular and Biomolecular Research, University of TorontoTorontoCanada
- Department of Molecular Genetics, University of TorontoTorontoCanada
| | - Sachdev S Sidhu
- Donnelly Centre for Cellular and Biomolecular Research, University of TorontoTorontoCanada
- Department of Molecular Genetics, University of TorontoTorontoCanada
| | - Peter J Murray
- Department of Immunoregulation, Max Planck Institute of BiochemistryMartinsriedGermany
| | - Matthias Mann
- Department of Proteomics and Signal Transduction, Max Planck Institute of BiochemistryMartinsriedGermany
| | - Mitchell J Weiss
- Department of Hematology, St. Jude Children’s Research HospitalMemphisUnited States
| | - Brenda A Schulman
- Department of Molecular Machines and Signaling, Max Planck Institute of BiochemistryMartinsriedGermany
| | - Arno F Alpi
- Department of Molecular Machines and Signaling, Max Planck Institute of BiochemistryMartinsriedGermany
| |
Collapse
|
9
|
Caulier A, Jankovsky N, Gautier EF, El Nemer W, Guitton C, Ouled-Haddou H, Guillonneau F, Mayeux P, Salnot V, Bruce J, Picard V, Garçon L. Red blood cell proteomics reveal remnant protein biosynthesis and folding pathways in PIEZO1-related hereditary xerocytosis. Front Physiol 2022; 13:960291. [PMID: 36531183 PMCID: PMC9751340 DOI: 10.3389/fphys.2022.960291] [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: 06/02/2022] [Accepted: 10/21/2022] [Indexed: 11/25/2023] Open
Abstract
Hereditary xerocytosis is a dominant red cell membrane disorder characterized by an increased leak of potassium from the inside to outside the red blood cell membrane, associated with loss of water leading to red cell dehydration and chronic hemolysis. 90% of cases are related to heterozygous gain of function mutations in PIEZO1, encoding a mechanotransductor that translates a mechanical stimulus into a biological signaling. Data are still required to understand better PIEZO1-HX pathophysiology. Recent studies identified proteomics as an accurate and high-input tool to study erythroid progenitors and circulating red cell physiology. Here, we isolated red blood cells from 5 controls and 5 HX patients carrying an identified and pathogenic PIEZO1 mutation and performed a comparative deep proteomic analysis. A total of 603 proteins were identified among which 56 were differentially expressed (40 over expressed and 16 under expressed) between controls and HX with a homogenous expression profile within each group. We observed relevant modifications in the protein expression profile related to PIEZO1 mutations, identifying two main "knots". The first contained both proteins of the chaperonin containing TCP1 complex involved in the assembly of unfolded proteins, and proteins involved in translation. The second contained proteins involved in ubiquitination. Deregulation of proteins involved in protein biosynthesis was also observed in in vitro-produced reticulocytes after Yoda1 exposure. Thus, our work identifies significant changes in the protein content of PIEZO1-HX erythrocytes, revealing a "PIEZO1 signature" and identifying potentially targetable pathways in this disease characterized by a heterogeneous clinical expression and contra-indication of splenectomy.
Collapse
Affiliation(s)
- Alexis Caulier
- HEMATIM, CURS, Amiens and Laboratoire d’Hématologie, CHU Amiens, UPJV, Amiens, France
| | - Nicolas Jankovsky
- HEMATIM, CURS, Amiens and Laboratoire d’Hématologie, CHU Amiens, UPJV, Amiens, France
| | - Emilie Fleur Gautier
- 3P5 Proteom’IC, Institut Cochin, INSERM, CNRS, Université Paris Cité, Paris, France
- Institut Imagine-INSERM U1163, Necker Hospital, University of Paris, Paris, France
- Laboratoire d’excellence GR-Ex, Paris, France
| | | | - Corinne Guitton
- Laboratoire d’Hématologie et Filière MCGRE, CHU Bicêtre, Le Kremlin-Bicêtre, France
| | - Hakim Ouled-Haddou
- HEMATIM, CURS, Amiens and Laboratoire d’Hématologie, CHU Amiens, UPJV, Amiens, France
| | - François Guillonneau
- 3P5 Proteom’IC, Institut Cochin, INSERM, CNRS, Université Paris Cité, Paris, France
| | - Patrick Mayeux
- 3P5 Proteom’IC, Institut Cochin, INSERM, CNRS, Université Paris Cité, Paris, France
| | - Virginie Salnot
- 3P5 Proteom’IC, Institut Cochin, INSERM, CNRS, Université Paris Cité, Paris, France
| | - Johanna Bruce
- 3P5 Proteom’IC, Institut Cochin, INSERM, CNRS, Université Paris Cité, Paris, France
| | - Véronique Picard
- Laboratoire d’Hématologie et Filière MCGRE, CHU Bicêtre, Le Kremlin-Bicêtre, France
- Laboratoire d’Hématologie, Faculté de Pharmacie, Université Paris Saclay, Amiens, France
| | - Loïc Garçon
- HEMATIM, CURS, Amiens and Laboratoire d’Hématologie, CHU Amiens, UPJV, Amiens, France
- INSERM U1134, INTS, Paris, France
- Laboratoire d’Hématologie et Filière MCGRE, CHU Bicêtre, Le Kremlin-Bicêtre, France
| |
Collapse
|
10
|
The deubiquitinase USP7 promotes HNSCC progression via deubiquitinating and stabilizing TAZ. Cell Death Dis 2022; 13:677. [PMID: 35931679 PMCID: PMC9356134 DOI: 10.1038/s41419-022-05113-z] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Revised: 07/13/2022] [Accepted: 07/18/2022] [Indexed: 01/21/2023]
Abstract
Dysregulated abundance, location and transcriptional output of Hippo signaling effector TAZ have been increasingly linked to human cancers including head neck squamous cell carcinoma (HNSCC). TAZ is subjected to ubiquitination and degradation mediated by E3 ligase β-TRCP. However, the deubiquitinating enzymes and mechanisms responsible for its protein stability remain underexplored. Here, we exploited customized deubiquitinases siRNA and cDNA library screen strategies and identified USP7 as a bona fide TAZ deubiquitinase in HNSCC. USP7 promoted cell proliferation, migration, invasion in vitro and tumor growth by stabilizing TAZ. Mechanistically, USP7 interacted with, deubiquitinated and stabilized TAZ by selectively removing its K48-linked ubiquitination chain independent of canonical Hippo kinase cascade. USP7 potently antagonized β-TRCP-mediated ubiquitin-proteasomal degradation of TAZ and enhanced its nuclear retention and transcriptional output. Importantly, overexpression of USP7 correlated with TAZ upregulation, tumor aggressiveness and unfavorable prognosis in HNSCC patients. Pharmacological inhibition of USP7 significantly suppressed tumor growth in both xenograft and PDX models. Collectively, these findings identify USP7 as an essential regulator of TAZ and define USP7-TAZ signaling axis as a novel biomarker and potential therapeutic target for HNSCC.
Collapse
|
11
|
Ren K, Li E, Ji P. Proteome remodeling and organelle clearance in mammalian terminal erythropoiesis. Curr Opin Hematol 2022; 29:137-143. [PMID: 35441599 DOI: 10.1097/moh.0000000000000707] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
PURPOSE OF REVIEW The differentiation from colony forming unit-erythroid (CFU-E) cells to mature enucleated red blood cells is named terminal erythropoiesis in mammals. Apart from enucleation, several unique features during these developmental stages include proteome remodeling and organelle clearance that are important to achieve hemoglobin enrichment. Here, we review the recent advances in the understanding of novel regulatory mechanisms in these processes, focusing on the master regulators that link these major events during terminal erythropoiesis. RECENT FINDINGS Comprehensive proteomic studies revealed a mismatch of protein abundance to their corresponding transcript abundance, which indicates that the proteome remodeling is regulated in a complex way from transcriptional control to posttranslational modifications. Key regulators in organelle clearance were also found to play critical roles in proteome remodeling. SUMMARY These studies demonstrate that the complexity of terminal erythropoiesis is beyond the conventional transcriptomic centric perspective. Posttranslational modifications such as ubiquitination are critical in terminal erythroid proteome remodeling that is also closely coupled with organelle clearance.
Collapse
Affiliation(s)
- Kehan Ren
- Department of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | | | | |
Collapse
|
12
|
Ren Y, Zhu J, Han Y, Li P, Wu J, Qu H, Zhang Z, Fang X. Regulatory association of long noncoding RNAs and chromatin accessibility facilitates erythroid differentiation. Blood Adv 2021; 5:5396-5409. [PMID: 34644394 PMCID: PMC9153002 DOI: 10.1182/bloodadvances.2021005167] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Accepted: 09/13/2021] [Indexed: 01/17/2023] Open
Abstract
Erythroid differentiation is a dynamic process regulated by multiple factors, whereas the interaction between long noncoding RNAs (lncRNAs) and chromatin accessibility and its influence on erythroid differentiation remains unclear. To elucidate this interaction, we used hematopoietic stem cells, multipotent progenitor cells, common myeloid progenitor cells, megakaryocyte-erythroid progenitor cells, and erythroblasts from human cord blood as an erythroid differentiation model to explore the coordinated regulatory functions of lncRNAs and chromatin accessibility by integrating RNA-seq and ATAC-seq data. We revealed that the integrated network of chromatin accessibility and lncRNAs exhibits stage-specific changes throughout the erythroid differentiation process and that the changes at the erythroblast stage of maturation are dramatic. We identified a subset of stage-specific lncRNAs and transcription factors (TFs) that associate with chromatin accessibility during erythroid differentiation, in which lncRNAs are key regulators of terminal erythroid differentiation via an lncRNA-TF-gene network. LncRNA PCED1B-AS1 was revealed to regulate terminal erythroid differentiation by coordinating GATA1 dynamically binding to the chromatin and interacting with the cytoskeleton network during erythroid differentiation. DANCR, another lncRNA that is highly expressed at the megakaryocyte-erythroid progenitor cell stage, was verified to promote erythroid differentiation by compromising megakaryocyte differentiation and coordinating with chromatin accessibility and TFs, such as RUNX1. Overall, our results identify the associated network of lncRNAs and chromatin accessibility in erythropoiesis and provide novel insights into erythroid differentiation and abundant resources for further study.
Collapse
Affiliation(s)
| | - Junwei Zhu
- CAS Key Laboratory of Genome Science & Information, Beijing Institute of Genomics, Chinese Academy of Sciences/China National Center for Bioinformation, Beijing, China
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China
- Beijing Key Laboratory of Genome and Precision Medicine Technologies, Beijing, China
| | - Yuanyuan Han
- Guizhou University, Medical College, Guiyang, China, China; and
| | - Pin Li
- CAS Key Laboratory of Genome Science & Information, Beijing Institute of Genomics, Chinese Academy of Sciences/China National Center for Bioinformation, Beijing, China
| | - Jing Wu
- CAS Key Laboratory of Genome Science & Information, Beijing Institute of Genomics, Chinese Academy of Sciences/China National Center for Bioinformation, Beijing, China
- Sino-Danish College, University of Chinese Academy of Sciences, Beijing, China
| | - Hongzhu Qu
- CAS Key Laboratory of Genome Science & Information, Beijing Institute of Genomics, Chinese Academy of Sciences/China National Center for Bioinformation, Beijing, China
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China
- Beijing Key Laboratory of Genome and Precision Medicine Technologies, Beijing, China
| | | | - Xiangdong Fang
- CAS Key Laboratory of Genome Science & Information, Beijing Institute of Genomics, Chinese Academy of Sciences/China National Center for Bioinformation, Beijing, China
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China
- Beijing Key Laboratory of Genome and Precision Medicine Technologies, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
13
|
Cui D, Feng Y, Qian R. Up-regulation of microRNA miR-101-3p enhances sensitivity to cisplatin via regulation of small interfering RNA (siRNA) Anti-human AGT4D and autophagy in non-small-cell lung carcinoma (NSCLC). Bioengineered 2021; 12:8435-8446. [PMID: 34694211 PMCID: PMC8806688 DOI: 10.1080/21655979.2021.1982274] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
The emergence of drug resistance hinders the treatment of malignant tumors, and autophagy plays an important role in tumor chemotherapy resistance. However, its mechanism in non-small cell lung cancer (NSCLC) has not been well-researched. We aim to investigate the role of miR-101-3p in cisplatin-resistant via regulation of autophagy-related protein 4D (ATG4D) and autophagy. Cell viability, apoptosis, fluorescence intensity of GFP-LC3 and RFP-GFP-LC3 were determined using Cell Counting Kit-8 (CCK-8) assay, flow cytometry, and Laser scanning confocal microscope analysis, respectively. The levels of LC3II/LC3I, P62 and ATG4D were detected by Western blot. The results showed that the sensitivity to cisplatin in NSCLC cells was up-regulated by miR-101-3p mimics treatment, inducing promoting cell apoptosis and inhibiting autophagy. Further mechanistic study identified that ATG4D was a direct target of miR-101-3p. Moreover, ATG4D siRNA also could reverse miR-101-3p inhibitor-induced the up-regulation of ATG4D and the ration of LC3II/LC3I, the down-regulation of p62 expression. Our findings indicated that miR-101-3p could regulate sensitivity to cisplatin of NSNCC cells by regulating autophagy mediated by ATG4D. Therefore, miR-101-3p may act as a potential therapeutic target for the treatment of NSCLC.
Collapse
Affiliation(s)
- Dong Cui
- Department of Thoracic Surgery, Henan Provincial Chest Hospital, Zhengzhou, China
| | - Yu Feng
- Department of Thoracic Surgery, Henan Provincial Chest Hospital, Zhengzhou, China
| | - Rulin Qian
- Department of Thoracic Surgery, Henan Provincial Chest Hospital, Zhengzhou, China
| |
Collapse
|
14
|
Barabino SML, Citterio E, Ronchi AE. Transcription Factors, R-Loops and Deubiquitinating Enzymes: Emerging Targets in Myelodysplastic Syndromes and Acute Myeloid Leukemia. Cancers (Basel) 2021; 13:cancers13153753. [PMID: 34359655 PMCID: PMC8345071 DOI: 10.3390/cancers13153753] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Revised: 07/21/2021] [Accepted: 07/23/2021] [Indexed: 12/19/2022] Open
Abstract
Simple Summary The advent of DNA massive sequencing technologies has allowed for the first time an extensive look into the heterogeneous spectrum of genes and mutations underpinning myelodysplastic syndromes (MDSs) and acute myeloid leukemia (AML). In this review, we wish to explore the most recent advances and the rationale for the potential therapeutic interest of three main actors in myelo-leukemic transformation: transcription factors that govern myeloid differentiation; RNA splicing factors, which ensure proper mRNA maturation and whose mutations increase R-loops formation; and deubiquitinating enzymes, which contribute to genome stability in hematopoietic stem cells (HSCs). Abstract Myeloid neoplasms encompass a very heterogeneous family of diseases characterized by the failure of the molecular mechanisms that ensure a balanced equilibrium between hematopoietic stem cells (HSCs) self-renewal and the proper production of differentiated cells. The origin of the driver mutations leading to preleukemia can be traced back to HSC/progenitor cells. Many properties typical to normal HSCs are exploited by leukemic stem cells (LSCs) to their advantage, leading to the emergence of a clonal population that can eventually progress to leukemia with variable latency and evolution. In fact, different subclones might in turn develop from the original malignant clone through accumulation of additional mutations, increasing their competitive fitness. This process ultimately leads to a complex cancer architecture where a mosaic of cellular clones—each carrying a unique set of mutations—coexists. The repertoire of genes whose mutations contribute to the progression toward leukemogenesis is broad. It encompasses genes involved in different cellular processes, including transcriptional regulation, epigenetics (DNA and histones modifications), DNA damage signaling and repair, chromosome segregation and replication (cohesin complex), RNA splicing, and signal transduction. Among these many players, transcription factors, RNA splicing proteins, and deubiquitinating enzymes are emerging as potential targets for therapeutic intervention.
Collapse
|
15
|
Bang IH, Park D, Lee Y, Cho H, Park B, Bae EJ. Sirtuin 6 promotes eosinophil differentiation by activating GATA-1 transcription factor. Aging Cell 2021; 20:e13418. [PMID: 34125994 PMCID: PMC8282249 DOI: 10.1111/acel.13418] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2021] [Revised: 04/19/2021] [Accepted: 05/24/2021] [Indexed: 12/16/2022] Open
Abstract
There is evidence emerging that exposure to cold temperatures enhances alternative activation of macrophages in white adipose tissue (WAT), which promotes adipocyte beiging and adaptive thermogenesis. Although we recently reported that NAD+‐dependent deacetylase sirtuin 6 (Sirt6) drives alternatively activated (M2) macrophage polarization, the role of myeloid Sirt6 in adaptive thermogenesis had remained elusive. In this study, we demonstrate that myeloid Sirt6 deficiency impaired both thermogenic responses and M2 macrophage infiltration in subcutaneous WAT (scWAT) during cold exposure. Moreover, the infiltration of Siglec‐F‐positive eosinophils in scWAT and Th2 cytokines levels was reduced in myeloid Sirt6 knockout mice. An ex vivo bone marrow‐derived cell culture experiment indicated that Sirt6 was required for eosinophil differentiation independent of its deacetylase activity. Data from our in vitro experiments show that Sirt6 acted as a transcriptional cofactor of GATA‐1, independent of its catalytic function as a deacetylase or ADP‐ribosyltransferase. Specifically, Sirt6 physically interacted with GATA‐1, and enhanced GATA‐1’s acetylation and transcriptional activity by facilitating its cooperation with p300. Overall, our results suggest that myeloid Sirt6 plays an important role in eosinophil differentiation and fat beiging/adaptive thermogenesis, which is at least in part due to its ability to bind GATA‐1 and stimulate its transcriptional activity.
Collapse
Affiliation(s)
- In Hyuk Bang
- Department of Biochemistry and Molecular Biology Chonbuk National University Medical School Jeonju Korea
| | - Dami Park
- Department of Biochemistry and Molecular Biology Chonbuk National University Medical School Jeonju Korea
| | - Youngyi Lee
- Department of Biochemistry and Molecular Biology Chonbuk National University Medical School Jeonju Korea
| | - Hwangeui Cho
- College of Pharmacy Chonbuk National University Jeonju Korea
| | - Byung‐Hyun Park
- Department of Biochemistry and Molecular Biology Chonbuk National University Medical School Jeonju Korea
| | - Eun Ju Bae
- College of Pharmacy Chonbuk National University Jeonju Korea
| |
Collapse
|
16
|
Lee CS, Kim S, Hwang G, Song J. Deubiquitinases: Modulators of Different Types of Regulated Cell Death. Int J Mol Sci 2021; 22:4352. [PMID: 33919439 PMCID: PMC8122337 DOI: 10.3390/ijms22094352] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Revised: 04/16/2021] [Accepted: 04/19/2021] [Indexed: 02/06/2023] Open
Abstract
The mechanisms and physiological implications of regulated cell death (RCD) have been extensively studied. Among the regulatory mechanisms of RCD, ubiquitination and deubiquitination enable post-translational regulation of signaling by modulating substrate degradation and signal transduction. Deubiquitinases (DUBs) are involved in diverse molecular pathways of RCD. Some DUBs modulate multiple modalities of RCD by regulating various substrates and are powerful regulators of cell fate. However, the therapeutic targeting of DUB is limited, as the physiological consequences of modulating DUBs cannot be predicted. In this review, the mechanisms of DUBs that regulate multiple types of RCD are summarized. This comprehensive summary aims to improve our understanding of the complex DUB/RCD regulatory axis comprising various molecular mechanisms for diverse physiological processes. Additionally, this review will enable the understanding of the advantages of therapeutic targeting of DUBs and developing strategies to overcome the side effects associated with the therapeutic applications of DUB modulators.
Collapse
Affiliation(s)
- Choong-Sil Lee
- Integrated OMICS for Biomedical Science, World Class University, Yonsei University, Seoul 120-749, Korea;
| | - Seungyeon Kim
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul 120-749, Korea; (S.K.); (G.H.)
| | - Gyuho Hwang
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul 120-749, Korea; (S.K.); (G.H.)
| | - Jaewhan Song
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul 120-749, Korea; (S.K.); (G.H.)
| |
Collapse
|
17
|
Xu P, Scott DC, Xu B, Yao Y, Feng R, Cheng L, Mayberry K, Wang YD, Bi W, Palmer LE, King MT, Wang H, Li Y, Fan Y, Alpi AF, Li C, Peng J, Papizan J, Pruett-Miller SM, Spallek R, Bassermann F, Cheng Y, Schulman BA, Weiss MJ. FBXO11-mediated proteolysis of BAHD1 relieves PRC2-dependent transcriptional repression in erythropoiesis. Blood 2021; 137:155-167. [PMID: 33156908 PMCID: PMC7820877 DOI: 10.1182/blood.2020007809] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2020] [Accepted: 10/15/2020] [Indexed: 12/15/2022] Open
Abstract
The histone mark H3K27me3 and its reader/writer polycomb repressive complex 2 (PRC2) mediate widespread transcriptional repression in stem and progenitor cells. Mechanisms that regulate this activity are critical for hematopoietic development but are poorly understood. Here we show that the E3 ubiquitin ligase F-box only protein 11 (FBXO11) relieves PRC2-mediated repression during erythroid maturation by targeting its newly identified substrate bromo adjacent homology domain-containing 1 (BAHD1), an H3K27me3 reader that recruits transcriptional corepressors. Erythroblasts lacking FBXO11 are developmentally delayed, with reduced expression of maturation-associated genes, most of which harbor bivalent histone marks at their promoters. In FBXO11-/- erythroblasts, these gene promoters bind BAHD1 and fail to recruit the erythroid transcription factor GATA1. The BAHD1 complex interacts physically with PRC2, and depletion of either component restores FBXO11-deficient erythroid gene expression. Our studies identify BAHD1 as a novel effector of PRC2-mediated repression and reveal how a single E3 ubiquitin ligase eliminates PRC2 repression at many developmentally poised bivalent genes during erythropoiesis.
Collapse
Affiliation(s)
| | | | - Beisi Xu
- Department of Computational Biology
| | | | | | | | | | | | | | | | | | - Hong Wang
- Center for Proteomics and Metabolomics, St. Jude Children's Research Hospital, Memphis, TN
| | - Yuxin Li
- Center for Proteomics and Metabolomics, St. Jude Children's Research Hospital, Memphis, TN
| | | | - Arno F Alpi
- Department of Molecular Machines and Signaling, Max Planck Institute of Biochemistry, Martinsried, Germany
| | | | - Junmin Peng
- Department of Structural Biology
- Center for Proteomics and Metabolomics, St. Jude Children's Research Hospital, Memphis, TN
- Department of Development Neurobiology
| | | | - Shondra M Pruett-Miller
- Center for Advanced Genome Engineering, and
- Department of Cell and Molecular Biology, St. Jude Children's Research Hospital, Memphis, TN; and
| | - Ria Spallek
- Department of Medicine III and
- TranslaTUM, Center for Translational Cancer Research, Technical University of Munich, Munich, Germany
| | - Florian Bassermann
- Department of Medicine III and
- TranslaTUM, Center for Translational Cancer Research, Technical University of Munich, Munich, Germany
| | - Yong Cheng
- Department of Hematology
- Department of Computational Biology
| | - Brenda A Schulman
- Department of Structural Biology
- Department of Molecular Machines and Signaling, Max Planck Institute of Biochemistry, Martinsried, Germany
| | | |
Collapse
|
18
|
Cui L, Song W, Zeng Y, Wu Q, Fan Z, Huang T, Zeng B, Zhang M, Ni Q, Li Y, Wang T, Li D, Mao X, Lian T, Yang D, Yang M, Fan X. Deubiquitinase USP7 regulates Drosophila aging through ubiquitination and autophagy. Aging (Albany NY) 2020; 12:23082-23095. [PMID: 33221768 PMCID: PMC7746378 DOI: 10.18632/aging.104067] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2020] [Accepted: 08/14/2020] [Indexed: 02/04/2023]
Abstract
Ubiquitination-mediated protein degradation is the selective degradation of diverse forms of damaged proteins that are tagged with ubiquitin, while deubiquitinating enzymes reverse ubiquitination-mediated protein degradation by removing the ubiquitin chain from the target protein. The interactions of ubiquitinating and deubiquitinating enzymes are required to maintain protein homeostasis. The ubiquitin-specific protease USP7 is a deubiquitinating enzyme that indirectly plays a role in repairing DNA damage and development. However, the mechanism of its participation in aging has not been fully explored. Regarding this issue, we found that USP7 was necessary to maintain the normal lifespan of Drosophila melanogaster, and knockdown of dusp7 shortened the lifespan and reduced the ability of Drosophila to cope with starvation, oxidative stress and heat stress. Furthermore, we showed that the ability of USP7 to regulate aging depends on the autophagy and ubiquitin signaling pathways. Furthermore, 2,5-dimethyl-celecoxib (DMC), a derivative of celecoxib, can partially restore the shortened lifespan and aberrant phenotypes caused by dusp7 knockdown. Our results suggest that USP7 is an important factor involved in the regulation of aging, and related components in this regulatory pathway may become new targets for anti-aging treatments.
Collapse
Affiliation(s)
- Lang Cui
- Institute of Animal Genetics and Breeding, Sichuan Agricultural University, Chengdu, Sichuan, P. R. China
| | - Wenhao Song
- Institute of Animal Genetics and Breeding, Sichuan Agricultural University, Chengdu, Sichuan, P. R. China
| | - Yao Zeng
- Institute of Animal Genetics and Breeding, Sichuan Agricultural University, Chengdu, Sichuan, P. R. China
| | - Qi Wu
- Institute of Animal Genetics and Breeding, Sichuan Agricultural University, Chengdu, Sichuan, P. R. China
| | - Ziqiang Fan
- Institute of Animal Genetics and Breeding, Sichuan Agricultural University, Chengdu, Sichuan, P. R. China
| | - Tiantian Huang
- Institute of Animal Genetics and Breeding, Sichuan Agricultural University, Chengdu, Sichuan, P. R. China
| | - Bo Zeng
- Institute of Animal Genetics and Breeding, Sichuan Agricultural University, Chengdu, Sichuan, P. R. China.,Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Mingwang Zhang
- Institute of Animal Genetics and Breeding, Sichuan Agricultural University, Chengdu, Sichuan, P. R. China.,Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Qingyong Ni
- Institute of Animal Genetics and Breeding, Sichuan Agricultural University, Chengdu, Sichuan, P. R. China.,Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Yan Li
- Institute of Animal Genetics and Breeding, Sichuan Agricultural University, Chengdu, Sichuan, P. R. China.,Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Tao Wang
- Institute of Animal Genetics and Breeding, Sichuan Agricultural University, Chengdu, Sichuan, P. R. China.,Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Diyan Li
- Institute of Animal Genetics and Breeding, Sichuan Agricultural University, Chengdu, Sichuan, P. R. China.,Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Xueping Mao
- Institute of Animal Genetics and Breeding, Sichuan Agricultural University, Chengdu, Sichuan, P. R. China.,Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Ting Lian
- Institute of Animal Genetics and Breeding, Sichuan Agricultural University, Chengdu, Sichuan, P. R. China.,Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Deying Yang
- Institute of Animal Genetics and Breeding, Sichuan Agricultural University, Chengdu, Sichuan, P. R. China.,Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Mingyao Yang
- Institute of Animal Genetics and Breeding, Sichuan Agricultural University, Chengdu, Sichuan, P. R. China.,Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Xiaolan Fan
- Institute of Animal Genetics and Breeding, Sichuan Agricultural University, Chengdu, Sichuan, P. R. China.,Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, Sichuan, China
| |
Collapse
|
19
|
Zhao Y, Li X, Tian G, Zhao X, Wong J, Shen Y, Wu J. Ubiquitin-Specific-Processing Protease 7 Regulates Female Germline Stem Cell Self-Renewal Through DNA Methylation. Stem Cell Rev Rep 2020; 17:938-951. [PMID: 33151468 PMCID: PMC8166723 DOI: 10.1007/s12015-020-10076-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/27/2020] [Indexed: 11/29/2022]
Abstract
Ubiquitin-specific-processing protease 7 (Usp7) is a key deubiquitinase controlling epigenetic modification and regulating the self-renewal, proliferation, and differentiation of stem cells. However, the functions and mechanisms of action of Usp7 on female germline stem cells (FGSCs) are unknown. Here, we demonstrated that Usp7 regulated FGSC self-renewal via DNA methylation. The results of Cell Counting Kit-8 and 5-ethynyl-20-deoxyuridine assays showed that the viability and proliferation of FGSCs were negatively regulated by Usp7. Moreover, Usp7 downregulated the expression of self-renewal genes, such as Oct4, Etv5, Foxo1, and Akt, but upregulated the expression of differentiation-related genes including Stra8 and Sycp3. Mechanistically, RNA-seq results showed that Usp7 negatively regulated FGSC self-renewal but positively modulated differentiation in FGSCs. Meanwhile, both overexpression and knockdown of Usp7 resulted in significant changes in DNA methylation and histone modification in FGSCs. Additionally, RNA-seq and MeDIP-seq analyses showed that Usp7 regulates the self-renewal and differentiation of FGSCs mainly through DNA methylation rather than histone modification, which was also confirmed by a rescue assay. Our study not only offers a novel method to research FGSC self-renewal and differentiation in view of epigenetic modifications, but also provides a deep understanding of FGSC development. Graphical Abstract ![]()
Collapse
Affiliation(s)
- Yongqiang Zhao
- Renji Hospital, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Bio-X Institutes, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Xiaoyong Li
- Renji Hospital, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Bio-X Institutes, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Geng Tian
- Renji Hospital, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Bio-X Institutes, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Xinyan Zhao
- Key Laboratory of Fertility Preservation and Maintenance of Ministry of Education, Ningxia Medical University, Yinchuan, 750004, China
| | - Jiemin Wong
- Shanghai Key Laboratory of Regulatory Biology, Fengxian District Central Hospital-ECNU Joint Center of Translational Medicine, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, 200241, China
| | - Yue Shen
- Key Laboratory of Fertility Preservation and Maintenance of Ministry of Education, Ningxia Medical University, Yinchuan, 750004, China
| | - Ji Wu
- Renji Hospital, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Bio-X Institutes, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200240, China. .,Key Laboratory of Fertility Preservation and Maintenance of Ministry of Education, Ningxia Medical University, Yinchuan, 750004, China.
| |
Collapse
|
20
|
Das S, Chandrasekaran AP, Jo KS, Ko NR, Oh SJ, Kim KS, Ramakrishna S. HAUSP stabilizes Cdc25A and protects cervical cancer cells from DNA damage response. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2020; 1867:118835. [PMID: 32860838 DOI: 10.1016/j.bbamcr.2020.118835] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/21/2020] [Revised: 08/14/2020] [Accepted: 08/19/2020] [Indexed: 12/21/2022]
Abstract
Resistance to DNA-damaging agents is one of the main reasons for the low survival of cervical cancer patients. Previous reports have suggested that the Cdc25A oncoprotein significantly affects the level of susceptibility to DNA-damaging agents, but the molecular mechanism remains unclear. In this study, we used Western blot and flow cytometry analyses to demonstrate that the deubiquitinating enzyme HAUSP stabilizes Cdc25A protein level. Furthermore, in a co-immunoprecipitation assay, we found that HAUSP interacts with and deubiquitinates Cdc25A both exogenously and endogenously. HAUSP extends the half-life of the Cdc25A protein by circumventing turnover. HAUSP knockout in HeLa cells using the CRISPR/Cas9 system caused a significant delay in Cdc25A-mediated cell cycle progression, cell migration, and colony formation and attenuated tumor progression in a mouse xenograft model. Furthermore, HAUSP-mediated stabilization of the Cdc25A protein produced enhanced resistance to DNA-damaging agents. Overall, our study suggests that targeting Cdc25A and HAUSP could be a promising combinatorial approach to halt progression and minimize antineoplastic resistance in cervical cancer.
Collapse
Affiliation(s)
- Soumyadip Das
- Graduate School of Biomedical Science and Engineering, Hanyang University, Seoul, South Korea
| | | | - Ki-Sang Jo
- Graduate School of Biomedical Science and Engineering, Hanyang University, Seoul, South Korea
| | - Na Re Ko
- Biomedical Research Center, Asan Institute for Life Sciences, Seoul, South Korea; Department of Nuclear Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea
| | - Seung Jun Oh
- Department of Nuclear Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea.
| | - Kye-Seong Kim
- Graduate School of Biomedical Science and Engineering, Hanyang University, Seoul, South Korea; College of Medicine, Hanyang University, Seoul, South Korea.
| | - Suresh Ramakrishna
- Graduate School of Biomedical Science and Engineering, Hanyang University, Seoul, South Korea; College of Medicine, Hanyang University, Seoul, South Korea.
| |
Collapse
|
21
|
de la Vega E, González N, Cabezas F, Montecino F, Blanco N, Olguín H. USP7-dependent control of myogenin stability is required for terminal differentiation in skeletal muscle progenitors. FEBS J 2020; 287:4659-4677. [PMID: 32115872 DOI: 10.1111/febs.15269] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2019] [Revised: 01/31/2020] [Accepted: 02/27/2020] [Indexed: 12/29/2022]
Abstract
Satellite cells (SCs) are myogenic progenitors responsible for skeletal muscle regeneration and maintenance. Upon activation, SCs enter a phase of robust proliferation followed by terminal differentiation. Underlying this myogenic progression, the sequential expression of muscle regulatory transcription factors (MRFs) and the downregulation of transcription factor paired box gene 7 (Pax7) are key steps regulating SC fate. In addition to transcriptional regulation, post-translational control of Pax7 and the MRFs provides another layer of spatiotemporal control to the myogenic process. In this context, previous work showed that Pax7 is ubiquitinated by the E3 ligase neural precursor cell-expressed developmentally downregulated protein 4 and interacts with several proteins related to the ubiquitin-proteasome system, including the deubiquitinase ubiquitin-specific protease 7 (USP7). Although USP7 functions in diverse cellular contexts, its role(s) during myogenesis remains poorly explored. Here, we show that USP7 is transiently expressed in adult muscle progenitors, correlating with the onset of myogenin expression, while it is downregulated in newly formed myotubes/myofibers. Acute inhibition of USP7 activity upon muscle injury results in persistent expression of early regeneration markers and a significant reduction in the diameter of regenerating myofibers. At the molecular level, USP7 downregulation or pharmacological inhibition impairs muscle differentiation by affecting myogenin stability. Co-immunoprecipitation and in vitro activity assays indicate that myogenin is a novel USP7 target for deubiquitination. These results suggest that USP7 regulates SC myogenic progression by enhancing myogenin stability.
Collapse
Affiliation(s)
- Eduardo de la Vega
- Laboratory of Tissue Repair and Adult Stem Cells, Molecular and Cell Biology Department, Faculty of Biological Sciences, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Natalia González
- Laboratory of Tissue Repair and Adult Stem Cells, Molecular and Cell Biology Department, Faculty of Biological Sciences, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Felipe Cabezas
- Laboratory of Tissue Repair and Adult Stem Cells, Molecular and Cell Biology Department, Faculty of Biological Sciences, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Fabián Montecino
- Laboratory of Tissue Repair and Adult Stem Cells, Molecular and Cell Biology Department, Faculty of Biological Sciences, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Natasha Blanco
- Laboratory of Tissue Repair and Adult Stem Cells, Molecular and Cell Biology Department, Faculty of Biological Sciences, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Hugo Olguín
- Laboratory of Tissue Repair and Adult Stem Cells, Molecular and Cell Biology Department, Faculty of Biological Sciences, Pontificia Universidad Católica de Chile, Santiago, Chile
| |
Collapse
|
22
|
Deng J, Liang L, Yi H, Su T, Yang Z, Nie L, Liu J. USP7 inhibition inhibits proliferation and induces megakaryocytic differentiation in MDS cells by upregulating gelsolin. Br J Haematol 2020; 190:418-429. [PMID: 32130729 DOI: 10.1111/bjh.16549] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Accepted: 01/31/2020] [Indexed: 01/02/2023]
Abstract
Myelodysplastic syndrome (MDS), a largely incurable hematological malignancy, is driven by complex genetic and epigenetic alterations from an aberrant clone of hematopoietic stem/progenitor cells (HSPCs). Ubiquitin-specific protease 7 (USP7) has been demonstrated to have an important oncogenic role in the development of several cancer types, but its role in MDS is unknown. Here, we demonstrate that USP7 expression is elevated in MDS cell lines and patient samples. The USP7-selective small-molecule inhibitors P5091 and P22077 inhibited cell proliferation and induced megakaryocytic differentiation in both cell lines and primary cells. Furthermore, pharmacological inhibition of USP7 markedly suppressed the growth of MDS cell lines in xenograft mouse models. To explore the mechanisms underlying the observed phenotypic changes, we employed RNA-seq to compare the differences in genes after USP7 inhibitor treatment and found that gelsolin (GSN) expression was increased significantly after USP7 inhibitor treatment. Furthermore, knockdown of GSN attenuated the proliferation inhibition, apoptosis induction and megakaryocyte differentiation induced by USP7 inhibitors in MDS cells. Collectively, our findings identify previously unknown roles of USP7 and suggest that the USP7/GSN axis may be a potential therapeutic target in MDS.
Collapse
Affiliation(s)
- Jinghui Deng
- Hematology Department, Hunan Province Key Laboratory of Basic and Applied Hematology, Xiangya Hospital, Central South University.,Hunan Province Key Laboratory of Basic and Applied Hematology, School of Life Sciences, Central South University
| | - Long Liang
- Hematology Department, Hunan Province Key Laboratory of Basic and Applied Hematology, Xiangya Hospital, Central South University.,Hunan Province Key Laboratory of Basic and Applied Hematology, School of Life Sciences, Central South University
| | - Hui Yi
- Hunan Province Key Laboratory of Basic and Applied Hematology, School of Life Sciences, Central South University
| | - Tao Su
- Hematology Department, Hunan Province Key Laboratory of Basic and Applied Hematology, Xiangya Hospital, Central South University
| | - Zhou Yang
- The First Affiliated Hospital of Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Ling Nie
- Hematology Department, Hunan Province Key Laboratory of Basic and Applied Hematology, Xiangya Hospital, Central South University
| | - Jing Liu
- Hunan Province Key Laboratory of Basic and Applied Hematology, School of Life Sciences, Central South University
| |
Collapse
|
23
|
Narla A, Mohandas N. Staying hydrated is important also for erythroblasts. Haematologica 2020; 105:528-529. [PMID: 32115411 DOI: 10.3324/haematol.2019.233999] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Affiliation(s)
- Anupama Narla
- Department of Pediatrics, Stanford University, School of Medicine, Stanford, CA
| | - Narla Mohandas
- Laboratory of Red Cell Physiology, New York Blood Center, New York, NY, USA
| |
Collapse
|
24
|
Chromatin occupancy and epigenetic analysis reveal new insights into the function of the GATA1 N terminus in erythropoiesis. Blood 2020; 134:1619-1631. [PMID: 31409672 DOI: 10.1182/blood.2019001234] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2019] [Accepted: 08/05/2019] [Indexed: 12/13/2022] Open
Abstract
Mutations in GATA1, which lead to expression of the GATA1s isoform that lacks the GATA1 N terminus, are seen in patients with Diamond-Blackfan anemia (DBA). In our efforts to better understand the connection between GATA1s and DBA, we comprehensively studied erythropoiesis in Gata1s mice. Defects in yolks sac and fetal liver hematopoiesis included impaired terminal maturation and reduced numbers of erythroid progenitors. RNA-sequencing revealed that both erythroid and megakaryocytic gene expression patterns were altered by the loss of the N terminus, including aberrant upregulation of Gata2 and Runx1. Dysregulation of global H3K27 methylation was found in the erythroid progenitors upon loss of N terminus of GATA1. Chromatin-binding assays revealed that, despite similar occupancy of GATA1 and GATA1s, there was a striking reduction of H3K27me3 at regulatory elements of the Gata2 and Runx1 genes. Consistent with the observation that overexpression of GATA2 has been reported to impair erythropoiesis, we found that haploinsufficiency of Gata2 rescued the erythroid defects of Gata1s fetuses. Together, our integrated genomic analysis of transcriptomic and epigenetic signatures reveals that, Gata1 mice provide novel insights into the role of the N terminus of GATA1 in transcriptional regulation and red blood cell maturation which may potentially be useful for DBA patients.
Collapse
|
25
|
Gutiérrez L, Caballero N, Fernández-Calleja L, Karkoulia E, Strouboulis J. Regulation of GATA1 levels in erythropoiesis. IUBMB Life 2019; 72:89-105. [PMID: 31769197 DOI: 10.1002/iub.2192] [Citation(s) in RCA: 76] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2019] [Accepted: 10/14/2019] [Indexed: 12/15/2022]
Abstract
GATA1 is considered as the "master" transcription factor in erythropoiesis. It regulates at the transcriptional level all aspects of erythroid maturation and function, as revealed by gene knockout studies in mice and by genome-wide occupancies in erythroid cells. The GATA1 protein contains two zinc finger domains and an N-terminal transactivation domain. GATA1 translation results in the production of the full-length protein and of a shorter variant (GATA1s) lacking the N-terminal transactivation domain, which is functionally deficient in supporting erythropoiesis. GATA1 protein abundance is highly regulated in erythroid cells at different levels, including transcription, mRNA translation, posttranslational modifications, and protein degradation, in a differentiation-stage-specific manner. Maintaining high GATA1 protein levels is essential in the early stages of erythroid maturation, whereas downregulating GATA1 protein levels is a necessary step in terminal erythroid differentiation. The importance of maintaining proper GATA1 protein homeostasis in erythropoiesis is demonstrated by the fact that both GATA1 loss and its overexpression result in lethal anemia. Importantly, alterations in any of those GATA1 regulatory checkpoints have been recognized as an important cause of hematological disorders such as dyserythropoiesis (with or without thrombocytopenia), β-thalassemia, Diamond-Blackfan anemia, myelodysplasia, or leukemia. In this review, we provide an overview of the multilevel regulation of GATA1 protein homeostasis in erythropoiesis and of its deregulation in hematological disease.
Collapse
Affiliation(s)
- Laura Gutiérrez
- Platelet Research Lab, Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Oviedo, Spain.,Department of Medicine, Universidad de Oviedo, Oviedo, Spain
| | - Noemí Caballero
- Platelet Research Lab, Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Oviedo, Spain
| | - Luis Fernández-Calleja
- Platelet Research Lab, Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Oviedo, Spain
| | - Elena Karkoulia
- Institute of Molecular Biology and Biotechnology, Foundation of Research & Technology Hellas, Heraklion, Crete, Greece
| | - John Strouboulis
- Cancer Comprehensive Center, School of Cancer and Pharmaceutical Sciences, King's College London, London, United Kingdom
| |
Collapse
|
26
|
Peng Y, Liu Y, Gao Y, Yuan B, Qi X, Fu Y, Zhu Q, Cao T, Zhang S, Yin L, Li X. USP7 is a novel Deubiquitinase sustaining PLK1 protein stability and regulating chromosome alignment in mitosis. J Exp Clin Cancer Res 2019; 38:468. [PMID: 31730000 PMCID: PMC6858727 DOI: 10.1186/s13046-019-1457-8] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2019] [Accepted: 10/17/2019] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND The deubiquitinase USP7 has been identified as an oncogene with key roles in tumorigenesis and therapeutic resistance for a series of cancer types. Recently small molecular inhibitors have been developed to target USP7. However, the anticancer mechanism of USP7 inhibitors is still elusive. METHODS Cell viability or clonogenicity was tested by violet crystal assay. Cell apoptosis or cell cycle was analyzed by flow cytometry, and chromosome misalignment was observed by a fluorescent microscopy. The protein interaction of PLK1 and USP7 was detected by tandem affinity purification and high throughput proteomics, and further confirmed by co-immunoprecipitation, GST pull-down and protein co-localization. The correlation between USP7 level of tumor tissues and taxane-resistance was evaluated. RESULTS Pharmacological USP7 inhibition by P5091 retarded cell proliferation and induced cell apoptosis. Further studies showed that P5091 induced cell cycle arrest at G2/M phase, and particularly induced chromosome misalignment, indicating the key roles of USP7 in mitosis. USP7 protein was detected in the PLK1-interacted protein complex. USP7 interacts with PLK1 protein through its PBD domain by catalytic activity. USP7 as a deubiquitinase sustained PLK1 protein stability via the C223 site, and inversely, USP7 inhibition by P5091 promoted the protein degradation of PLK1 through the ubiquitination-proteasome pathway. By overexpressing PLK1, USP7 that had been depleted by RNAi ceased to induce chromosome misalignment in mitosis and again supported cell proliferation and cell survival. Both USP7 and PLK1 were overexpressed in taxane-resistant cancer cells, and negatively correlated with the MP scores in tumor tissues. Either USP7 or PLK1 knockdown by RNAi significantly sensitized taxane-resistant cells to taxane cell killing. CONCLUSION This is the first report that PLK1 is a novel substrate of USP7 deubiquitinase, and that USP7 sustained the protein stability of PLK1. USP7 inhibition induces cell apoptosis and cell cycle G2/M arrest, and overcomes taxane resistance by inducing the protein degradation of PLK1, resulting in chromosome misalignment in mitosis.
Collapse
Affiliation(s)
- Yuchong Peng
- Center for Molecular Medicine, Xiangya Hospital, Central South University, Changsha, China
- Hunan Key Laboratory of Molecular Radiation Oncology, Xiangya Hospital, Central South University, Changsha, China
| | - Youhong Liu
- Center for Molecular Medicine, Xiangya Hospital, Central South University, Changsha, China
- Hunan Key Laboratory of Molecular Radiation Oncology, Xiangya Hospital, Central South University, Changsha, China
| | - Yingxue Gao
- Center for Molecular Medicine, Xiangya Hospital, Central South University, Changsha, China
- Hunan Key Laboratory of Molecular Radiation Oncology, Xiangya Hospital, Central South University, Changsha, China
| | - Bowen Yuan
- Department of Pathology, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Xuli Qi
- Center for Molecular Medicine, Xiangya Hospital, Central South University, Changsha, China
- Hunan Key Laboratory of Molecular Radiation Oncology, Xiangya Hospital, Central South University, Changsha, China
| | - Yuxin Fu
- Center for Molecular Medicine, Xiangya Hospital, Central South University, Changsha, China
- Hunan Key Laboratory of Molecular Radiation Oncology, Xiangya Hospital, Central South University, Changsha, China
| | - Qianling Zhu
- Center for Molecular Medicine, Xiangya Hospital, Central South University, Changsha, China
- Hunan Key Laboratory of Molecular Radiation Oncology, Xiangya Hospital, Central South University, Changsha, China
| | - Tuoyu Cao
- Center for Molecular Medicine, Xiangya Hospital, Central South University, Changsha, China
- Hunan Key Laboratory of Molecular Radiation Oncology, Xiangya Hospital, Central South University, Changsha, China
| | - Songwei Zhang
- Center for Molecular Medicine, Xiangya Hospital, Central South University, Changsha, China
- Hunan Key Laboratory of Molecular Radiation Oncology, Xiangya Hospital, Central South University, Changsha, China
| | - Linglong Yin
- Center for Molecular Medicine, Xiangya Hospital, Central South University, Changsha, China
- Hunan Key Laboratory of Molecular Radiation Oncology, Xiangya Hospital, Central South University, Changsha, China
| | - Xiong Li
- Center for Molecular Medicine, Xiangya Hospital, Central South University, Changsha, China
- Hunan Key Laboratory of Molecular Radiation Oncology, Xiangya Hospital, Central South University, Changsha, China
- School of Clinical Pharmacy, Guangdong Pharmacology University, Guangzhou, China
- The First Affiliated Hospital, Guangdong Pharmacology University, 19 Nonglinxia Road, Yuexiu District, Guangzhou, Guangdong China
| |
Collapse
|
27
|
Ling T, Crispino JD. GATA1 mutations in red cell disorders. IUBMB Life 2019; 72:106-118. [PMID: 31652397 DOI: 10.1002/iub.2177] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2019] [Accepted: 09/18/2019] [Indexed: 01/01/2023]
Abstract
GATA1 is an essential regulator of erythroid cell gene expression and maturation. In its absence, erythroid progenitors are arrested in differentiation and undergo apoptosis. Much has been learned about GATA1 function through animal models, which include genetic knockouts as well as ones with decreased levels of expression. However, even greater insights have come from the finding that a number of rare red cell disorders, including Diamond-Blackfan anemia, are associated with GATA1 mutations. These mutations affect the amino-terminal zinc finger (N-ZF) and the amino-terminus of the protein, and in both cases can alter the DNA-binding activity, which is primarily conferred by the third functional domain, the carboxyl-terminal zinc finger (C-ZF). Here we discuss the role of GATA1 in erythropoiesis with an emphasis on the mutations found in human patients with red cell disorders.
Collapse
Affiliation(s)
- Te Ling
- Division of Hematology/Oncology, Northwestern University, Chicago, Illinois
| | - John D Crispino
- Division of Hematology/Oncology, Northwestern University, Chicago, Illinois
| |
Collapse
|
28
|
Wang Z, Kang W, You Y, Pang J, Ren H, Suo Z, Liu H, Zheng Y. USP7: Novel Drug Target in Cancer Therapy. Front Pharmacol 2019; 10:427. [PMID: 31114498 PMCID: PMC6502913 DOI: 10.3389/fphar.2019.00427] [Citation(s) in RCA: 85] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2019] [Accepted: 04/04/2019] [Indexed: 12/22/2022] Open
Abstract
Ubiquitin specific protease 7 (USP7) is one of the deubiquitinating enzymes (DUB) that erases ubiquitin and protects substrate protein from degradation. Full activity of USP7 requires the C-terminal Ub-like domains fold back onto the catalytic domain, allowing the remodeling of the active site to a catalytically competent state by the C-terminal peptide. Until now, numerous proteins have been identified as substrates of USP7, which play a key role in cell cycle, DNA repair, chromatin remodeling, and epigenetic regulation. Aberrant activation or overexpression of USP7 may promote oncogenesis and viral disease, making it a target for therapeutic intervention. Currently, several synthetic small molecules have been identified as inhibitors of USP7, and applied in the treatment of diverse diseases. Hence, USP7 may be a promising therapeutic target for the treatment of cancer.
Collapse
Affiliation(s)
- Zhiru Wang
- School of Pharmaceutical Sciences, Zhenghzou University, Zhengzhou, China.,Collaborative Innovation Centre of New Drug Research and Safety Evaluation, Henan Province, and Key Laboratory of Advanced Drug Preparation Technologies, Zhengzhou University, and Key Laboratory of Henan Province for Drug Quality and Evaluation, Ministry of Education of China, Zhengzhou, China.,Pathology, Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Wenting Kang
- School of Pharmaceutical Sciences, Zhenghzou University, Zhengzhou, China.,Collaborative Innovation Centre of New Drug Research and Safety Evaluation, Henan Province, and Key Laboratory of Advanced Drug Preparation Technologies, Zhengzhou University, and Key Laboratory of Henan Province for Drug Quality and Evaluation, Ministry of Education of China, Zhengzhou, China
| | - Yinghua You
- School of Pharmaceutical Sciences, Zhenghzou University, Zhengzhou, China.,Collaborative Innovation Centre of New Drug Research and Safety Evaluation, Henan Province, and Key Laboratory of Advanced Drug Preparation Technologies, Zhengzhou University, and Key Laboratory of Henan Province for Drug Quality and Evaluation, Ministry of Education of China, Zhengzhou, China
| | - Jingru Pang
- School of Pharmaceutical Sciences, Zhenghzou University, Zhengzhou, China.,Collaborative Innovation Centre of New Drug Research and Safety Evaluation, Henan Province, and Key Laboratory of Advanced Drug Preparation Technologies, Zhengzhou University, and Key Laboratory of Henan Province for Drug Quality and Evaluation, Ministry of Education of China, Zhengzhou, China
| | - Hongmei Ren
- School of Pharmaceutical Sciences, Zhenghzou University, Zhengzhou, China.,Collaborative Innovation Centre of New Drug Research and Safety Evaluation, Henan Province, and Key Laboratory of Advanced Drug Preparation Technologies, Zhengzhou University, and Key Laboratory of Henan Province for Drug Quality and Evaluation, Ministry of Education of China, Zhengzhou, China
| | - Zhenhe Suo
- Pathology, Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Hongmin Liu
- School of Pharmaceutical Sciences, Zhenghzou University, Zhengzhou, China.,Collaborative Innovation Centre of New Drug Research and Safety Evaluation, Henan Province, and Key Laboratory of Advanced Drug Preparation Technologies, Zhengzhou University, and Key Laboratory of Henan Province for Drug Quality and Evaluation, Ministry of Education of China, Zhengzhou, China
| | - Yichao Zheng
- School of Pharmaceutical Sciences, Zhenghzou University, Zhengzhou, China.,Collaborative Innovation Centre of New Drug Research and Safety Evaluation, Henan Province, and Key Laboratory of Advanced Drug Preparation Technologies, Zhengzhou University, and Key Laboratory of Henan Province for Drug Quality and Evaluation, Ministry of Education of China, Zhengzhou, China
| |
Collapse
|