1
|
Sundararajan R, Hegde SR, Panda AK, Christie J, Gadewal N, Venkatraman P. Loss of correlated proteasomal subunit expression selectively promotes the 20S High state which underlies luminal breast tumorigenicity. Commun Biol 2025; 8:55. [PMID: 39814910 PMCID: PMC11735796 DOI: 10.1038/s42003-024-07432-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Accepted: 12/19/2024] [Indexed: 01/18/2025] Open
Abstract
Why cancer cells disproportionately accumulate polyubiquitinated proteotoxic proteins despite high proteasomal activity is an outstanding question. While mis-regulated ubiquitination is a contributing factor, here we show that a structurally-perturbed and sub-optimally functioning proteasome is at the core of altered proteostasis in tumors. By integrating the gene coexpression signatures of proteasomal subunits in breast cancer (BrCa) patient tissues with the atomistic details of 26S holocomplex, we find that the transcriptional deregulation induced-stoichiometric imbalances perpetuate with disease severity. As seen in luminal BrCa cell lines, this imbalance limits the number of double-capped 19S-20S-19S holocomplexes (30S) formed and promotes free 20S catalytic core accumulation that is widely-believed to confer survival advantage to tumors. By retaining connectivity with key tumor 19S:20S interface nodes, the PSMD9 19S subunit chaperone emerges as a crucial regulator of 26S/30S:20S ratios sustaining tumor cell proteasome function. Disrupting this connectivity by depleting PSMD9 in MCF7 cells introduces structural anomalies in the proteasome, and shifts dependence from 20SHigh to a deregulated 26SHigh state invoking anti-tumor responses which opens up clinically-relevant therapeutic possibilities.
Collapse
Affiliation(s)
- Rangapriya Sundararajan
- Protein Interactome Laboratory for Structural and Functional Biology, Advanced Centre for Treatment, Research and Education in Cancer, Navi Mumbai, India.
- Department of Computer Science and Engineering, Indian Institute of Technology, Bombay, Mumbai, India.
- Center for Cell and Gene Therapy, Texas Children's Hospital, Baylor College of Medicine, Houston, TX, USA.
| | - Shubhada R Hegde
- Institute of Bioinformatics and Applied Biotechnology, Bangalore, India
- School of Biosciences, Chanakya University, Bangalore, India
| | - Ashish Kumar Panda
- Protein Interactome Laboratory for Structural and Functional Biology, Advanced Centre for Treatment, Research and Education in Cancer, Navi Mumbai, India
- Homi Bhabha National Institute, Mumbai, India
| | - Joel Christie
- Protein Interactome Laboratory for Structural and Functional Biology, Advanced Centre for Treatment, Research and Education in Cancer, Navi Mumbai, India
- Homi Bhabha National Institute, Mumbai, India
| | - Nikhil Gadewal
- Bioinformatics Center, Advanced Centre for Treatment, Research and Education in Cancer, Navi Mumbai, India
| | - Prasanna Venkatraman
- Protein Interactome Laboratory for Structural and Functional Biology, Advanced Centre for Treatment, Research and Education in Cancer, Navi Mumbai, India.
- Homi Bhabha National Institute, Mumbai, India.
| |
Collapse
|
2
|
Dong Q, Wang D, Song C, Gong C, Liu Y, Zhou X, Yue J, Hu Y, Liu H, Zhu L, Niu X, Zheng T, Zhang X, Jin J, Wang T, Ju R, Wang C, Jiang Q, Gao T, Jin Y, Li P, Wang Y, Zhang C, Wang GF, Cao C, Liu X. ABL1-mediated phosphorylation promotes FOXM1-related tumorigenicity by Increasing FOXM1 stability. Cell Death Differ 2024; 31:1285-1301. [PMID: 39060421 PMCID: PMC11445503 DOI: 10.1038/s41418-024-01339-w] [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: 07/06/2023] [Revised: 06/20/2024] [Accepted: 07/01/2024] [Indexed: 07/28/2024] Open
Abstract
The transcription factor FOXM1, which plays critical roles in cell cycle progression and tumorigenesis, is highly expressed in rapidly proliferating cells and various tumor tissues, and high FOXM1 expression is related to a poor prognosis. However, the mechanism responsible for FOXM1 dysregulation is not fully understood. Here, we show that ABL1, a nonreceptor tyrosine kinase, contributes to the high expression of FOXM1 and FOXM1-dependent tumor development. Mechanistically, ABL1 directly binds FOXM1 and mediates FOXM1 phosphorylation at multiple tyrosine (Y) residues. Among these phospho-Y sites, pY575 is indispensable for FOXM1 stability as phosphorylation at this site protects FOXM1 from ubiquitin-proteasomal degradation. The interaction of FOXM1 with CDH1, a coactivator of the E3 ubiquitin ligase anaphase-promoting complex/cyclosome (APC/C), which is responsible for FOXM1 degradation, is significantly inhibited by Y575 phosphorylation. The phospho-deficient FOXM1(Y575F) mutant exhibited increased ubiquitination, a shortened half-life, and consequently a substantially decreased abundance. Compared to wild-type cells, a homozygous Cr-Y575F cell line expressing endogenous FOXM1(Y575F) that was generated by CRISPR/Cas9 showed obviously delayed mitosis progression, impeded colony formation and inhibited xenotransplanted tumor growth. Overall, our study demonstrates that ABL1 kinase is involved in high FOXM1 expression, providing clear evidence that ABL1 may act as a therapeutic target for the treatment of tumors with high FOXM1 expression.
Collapse
Affiliation(s)
- Qincai Dong
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Biotechnology, 100850, Beijing, China
| | - Di Wang
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Biotechnology, 100850, Beijing, China
| | - Caiwei Song
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Biotechnology, 100850, Beijing, China
| | - Chunxue Gong
- Institute of Health Sciences, Anhui University, Hefei, 230601, China
| | - Yue Liu
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Biotechnology, 100850, Beijing, China
| | - Xinwei Zhou
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Biotechnology, 100850, Beijing, China
| | - Junjie Yue
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Biotechnology, 100850, Beijing, China
| | - Yong Hu
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Biotechnology, 100850, Beijing, China
| | - Hainan Liu
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Biotechnology, 100850, Beijing, China
| | - Lin Zhu
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Biotechnology, 100850, Beijing, China
| | - Xiayang Niu
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Biotechnology, 100850, Beijing, China
| | - Tong Zheng
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Biotechnology, 100850, Beijing, China
| | - Xun Zhang
- Institute of Health Sciences, Anhui University, Hefei, 230601, China
| | - Jing Jin
- Institute of Health Sciences, Anhui University, Hefei, 230601, China
| | - Tingting Wang
- Institute of Health Sciences, Anhui University, Hefei, 230601, China
| | - Ruixia Ju
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Biotechnology, 100850, Beijing, China
| | - Chen Wang
- Institute of Health Sciences, Anhui University, Hefei, 230601, China
| | - Qian Jiang
- Institute of Health Sciences, Anhui University, Hefei, 230601, China
| | - Ting Gao
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Biotechnology, 100850, Beijing, China
| | - Yanwen Jin
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Biotechnology, 100850, Beijing, China
| | - Ping Li
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Biotechnology, 100850, Beijing, China
| | - Yan Wang
- Clinical Biobank Center, Medical Innovation Research Division, Chinese PLA General Hospital, 100853, Beijing, China
| | - Chunmei Zhang
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, 200241, China
| | - Guang-Fei Wang
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Biotechnology, 100850, Beijing, China.
| | - Cheng Cao
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Biotechnology, 100850, Beijing, China.
| | - Xuan Liu
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Biotechnology, 100850, Beijing, China.
| |
Collapse
|
3
|
Xiong J, Pang X, Song X, Yang L, Pang C. The coherence between PSMC6 and α-ring in the 26S proteasome is associated with Alzheimer's disease. Front Mol Neurosci 2024; 16:1330853. [PMID: 38357597 PMCID: PMC10864545 DOI: 10.3389/fnmol.2023.1330853] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Accepted: 12/22/2023] [Indexed: 02/16/2024] Open
Abstract
Alzheimer's disease (AD) is a heterogeneous age-dependent neurodegenerative disorder. Its hallmarks involve abnormal proteostasis, which triggers proteotoxicity and induces neuronal dysfunction. The 26S proteasome is an ATP-dependent proteolytic nanomachine of the ubiquitin-proteasome system (UPS) and contributes to eliminating these abnormal proteins. This study focused on the relationship between proteasome and AD, the hub genes of proteasome, PSMC6, and 7 genes of α-ring, are selected as targets to study. The following three characteristics were observed: 1. The total number of proteasomes decreased with AD progression because the proteotoxicity damaged the expression of proteasome proteins, as evidenced by the downregulation of hub genes. 2. The existing proteasomes exhibit increased activity and efficiency to counterbalance the decline in total proteasome numbers, as evidenced by enhanced global coordination and reduced systemic disorder of proteasomal subunits as AD advances. 3. The synergy of PSMC6 and α-ring subunits is associated with AD. Synergistic downregulation of PSMC6 and α-ring subunits reflects a high probability of AD risk. Regarding the above discovery, the following hypothesis is proposed: The aggregation of pathogenic proteins intensifies with AD progression, then proteasome becomes more active and facilitates the UPS selectively targets the degradation of abnormal proteins to maintain CNS proteostasis. In this paper, bioinformatics and support vector machine learning methods are applied and combined with multivariate statistical analysis of microarray data. Additionally, the concept of entropy was used to detect the disorder of proteasome system, it was discovered that entropy is down-regulated continually with AD progression against system chaos caused by AD. Another conception of the matrix determinant was used to detect the global coordination of proteasome, it was discovered that the coordination is enhanced to maintain the efficiency of degradation. The features of entropy and determinant suggest that active proteasomes resist the attack caused by AD like defenders, on the one hand, to protect themselves (entropy reduces), and on the other hand, to fight the enemy (determinant reduces). It is noted that these are results from biocomputing and need to be supported by further biological experiments.
Collapse
Affiliation(s)
- Jing Xiong
- College of Computer Science, Sichuan Normal University, Chengdu, China
| | - Xinping Pang
- West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, China
| | - Xianghu Song
- College of Computer Science, Sichuan Normal University, Chengdu, China
| | - Lin Yang
- College of Computer Science, Sichuan Normal University, Chengdu, China
| | - Chaoyang Pang
- College of Computer Science, Sichuan Normal University, Chengdu, China
| |
Collapse
|
4
|
Zhang N, Zhou Z, Huang Y, Wang G, Tang Z, Lu J, Wang C, Ni X. Reduced hydrogen sulfide production contributes to adrenal insufficiency induced by hypoxia via modulation of NLRP3 inflammasome activation. Redox Rep 2023; 28:2163354. [PMID: 36661247 PMCID: PMC9869992 DOI: 10.1080/13510002.2022.2163354] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
Objective: Adrenocortical responsiveness is critical for maintaining glucocorticoids production and homeostasis during stress. We sought to investigate adrenocortical responsiveness during hypoxia in mice and the mechanisms responsible for the regulation of adrenal responsiveness.Methods: (1) Adult male WT mice were randomly divided into four groups: normoxia, hypoxia (24h), hypoxia (72h), hypoxia (72h) + GYY4137(hydrogen sulfide (H2S) donor, 133mmol/kg/day); (2) WT mice were randomly divided into four groups: sham, adrenalectomy (ADX), sham+hypoxia, ADX+hypoxia; (3) Cse-/- mice were randomly divided into two groups: Cse-/-, Cse-/- +GYY4137.Results: The circulatory level of corticosteroid induced by ACTH stimulation was significantly reduced in the mice with hypoxia compared with control mice. The mortality rate induced by lipopolysaccharide (LPS) increased during hypoxia. Cystathionine-γ-lyase (CSE) expression was significantly reduced in adrenal glands during hypoxia. GYY4137 treatment significantly increased adrenal responsiveness and attenuated NLRP3 inflammasome activation in mice treated by hypoxia and Cse-/- mice. Furthermore, The sulfhydrated level of PSMA7 in adrenal gland was decreased in the mice with hypoxia and Cse-/- mice. PSMA7 was S-sulfhydrated at cysteine 70. Blockage of S-sulfhydration of PSMA7 increased NLRP3 expression in adrenocortical cells.Conclusion: Reduced H2S production mediated hypo-adrenocortical responsiveness and NLRP3 inflammasome activation via PAMA7 S-sulfhydration during hypoxia.
Collapse
Affiliation(s)
- Ningning Zhang
- National Clinical Research Center for Geriatric Disorders, Central South University Xiangya Hospital, Changsha, People’s Republic of China,International Collaborative Research Center for Medical Metabolomics, Central South University Xiangya Hospital, Changsha, People’s Republic of China,Department of Physiology, Navy Medical University, Shanghai, People’s Republic of China
| | - Zhan Zhou
- National Clinical Research Center for Geriatric Disorders, Central South University Xiangya Hospital, Changsha, People’s Republic of China,International Collaborative Research Center for Medical Metabolomics, Central South University Xiangya Hospital, Changsha, People’s Republic of China
| | - Yan Huang
- Department of Physiology, Navy Medical University, Shanghai, People’s Republic of China
| | - Gang Wang
- Department of Physiology, Navy Medical University, Shanghai, People’s Republic of China
| | - Zhengshan Tang
- National Clinical Research Center for Geriatric Disorders, Central South University Xiangya Hospital, Changsha, People’s Republic of China,International Collaborative Research Center for Medical Metabolomics, Central South University Xiangya Hospital, Changsha, People’s Republic of China
| | - Jianqiang Lu
- The Key Laboratory of Exercise and Health Sciences of Ministry of Education, School of Kinesiology, Shanghai University of Sport, Shanghai, People’s Republic of China
| | - Changnan Wang
- Department of Physiology, Navy Medical University, Shanghai, People’s Republic of China, Changnan Wang Department of Physiology, Navy Medical University, Shanghai200433, People’s Republic of China; Xin Ni
| | - Xin Ni
- National Clinical Research Center for Geriatric Disorders, Central South University Xiangya Hospital, Changsha, People’s Republic of China,International Collaborative Research Center for Medical Metabolomics, Central South University Xiangya Hospital, Changsha, People’s Republic of China,Department of Physiology, Navy Medical University, Shanghai, People’s Republic of China, Changnan Wang Department of Physiology, Navy Medical University, Shanghai200433, People’s Republic of China; Xin Ni
| |
Collapse
|
5
|
Okabe S, Gotoh A. Effect of asciminib and vitamin K2 on Abelson tyrosine-kinase-inhibitor-resistant chronic myelogenous leukemia cells. BMC Cancer 2023; 23:827. [PMID: 37670241 PMCID: PMC10478393 DOI: 10.1186/s12885-023-11304-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2023] [Accepted: 08/16/2023] [Indexed: 09/07/2023] Open
Abstract
BACKGROUND Abelson (ABL) tyrosine kinase inhibitors (TKIs) are effective against chronic myeloid leukemia (CML); however, many patients develop resistance during ABL TKI therapy. Vitamin K2 (VK2) is a crucial fat-soluble vitamin used to activate hepatic coagulation factors and treat osteoporosis. Although VK2 has demonstrated impressive anticancer activity in various cancer cell lines, it is not known whether VK2 enhances the effects of asciminib, which specifically targets the ABL myristoyl pocket (STAMP) inhibitor. METHOD In this work, we investigated whether VK2 contributed to the development of CML cell lines. We also investigated the efficacy of asciminib and VK2 by using K562, ponatinib-resistant K562 (K562 PR), Ba/F3 BCR-ABL, and T315I point mutant Ba/F3 (Ba/F3 T315I) cells. RESULTS Based on data from the Gene Expression Omnibus (GEO) database, gamma-glutamyl carboxylase (GGCX) and vitamin K epoxide reductase complex subunit 1 (VKORC1) were elevated in imatinib-resistant patients (GSE130404). UBIA Prenyltransferase Domain Containing 1 (UBIAD1) was decreased, and K562 PR cells were resistant to ponatinib. In contrast, asciminib inhibited CML cells and ponatinib resistance in a dose-dependent manner. CML cells were suppressed by VK2. Caspase 3/7 activity was also elevated, as was cellular cytotoxicity. Asciminib plus VK2 therapy induced a significantly higher level of cytotoxicity than use of each drug alone. Asciminib and VK2 therapy altered the mitochondrial membrane potential. CONCLUSIONS Asciminib and VK2 are suggested as a novel treatment for ABL-TKI-resistant cells since they increase treatment efficacy. Additionally, this treatment option has intriguing clinical relevance for patients who are resistant to ABL TKIs.
Collapse
Affiliation(s)
- Seiichi Okabe
- Department of Hematology, Tokyo Medical University, 6-7-1 Nishi-shinjuku, Shinjuku-ku, Tokyo, 160-0023, Japan.
| | - Akihiko Gotoh
- Department of Hematology, Tokyo Medical University, 6-7-1 Nishi-shinjuku, Shinjuku-ku, Tokyo, 160-0023, Japan
| |
Collapse
|
6
|
Kim Y, Kim EK, Chey Y, Song MJ, Jang HH. Targeted Protein Degradation: Principles and Applications of the Proteasome. Cells 2023; 12:1846. [PMID: 37508510 PMCID: PMC10378610 DOI: 10.3390/cells12141846] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Revised: 07/10/2023] [Accepted: 07/12/2023] [Indexed: 07/30/2023] Open
Abstract
The proteasome is a multi-catalytic protease complex that is involved in protein quality control via three proteolytic activities (i.e., caspase-, trypsin-, and chymotrypsin-like activities). Most cellular proteins are selectively degraded by the proteasome via ubiquitination. Moreover, the ubiquitin-proteasome system is a critical process for maintaining protein homeostasis. Here, we briefly summarize the structure of the proteasome, its regulatory mechanisms, proteins that regulate proteasome activity, and alterations to proteasome activity found in diverse diseases, chemoresistant cells, and cancer stem cells. Finally, we describe potential therapeutic modalities that use the ubiquitin-proteasome system.
Collapse
Affiliation(s)
- Yosup Kim
- Department of Biochemistry, College of Medicine, Gachon University, Incheon 21999, Republic of Korea
| | - Eun-Kyung Kim
- Department of Biochemistry, College of Medicine, Gachon University, Incheon 21999, Republic of Korea
| | - Yoona Chey
- Department of Biochemistry, College of Medicine, Gachon University, Incheon 21999, Republic of Korea
| | - Min-Jeong Song
- Department of Biochemistry, College of Medicine, Gachon University, Incheon 21999, Republic of Korea
| | - Ho Hee Jang
- Department of Biochemistry, College of Medicine, Gachon University, Incheon 21999, Republic of Korea
- Department of Health Sciences and Technology, Gachon Advanced Institute for Health Sciences and Technology (GAIHST), Gachon University, Incheon 21999, Republic of Korea
- Lee Gil Ya Cancer and Diabetes Institute, Gachon University, Incheon 21999, Republic of Korea
| |
Collapse
|
7
|
Lu F, Zhou J, Chen Q, Zhu J, Zheng X, Fang N, Qiao L. PSMA5 contributes to progression of lung adenocarcinoma in association with the JAK/STAT pathway. Carcinogenesis 2022; 43:624-634. [PMID: 35605971 DOI: 10.1093/carcin/bgac046] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Revised: 04/29/2022] [Accepted: 05/21/2022] [Indexed: 11/12/2022] Open
Abstract
Proteasome dysregulation is a common feature of cancer and a critical risk for tumorigenesis. However, the characteristics of proteasome components in tumor development and metastasis are poorly understood. PSMA5, an α5 subunit of the 20S core proteasome, is associated with the degradation of intracellular proteins. Increasing evidence indicated it is involved in tumor development, but the underlying mechanism has remained unknown. Here, we show that PSMA5 is up-regulated in lung adenocarcinoma (LUAD) cells and clinical LUAD tissues. Moreover, its up-regulation is positively associated with lymph node metastasis and the poor prognosis of LUAD patients. PSMA5 knockdown inhibited the proliferation, invasion and metastasis of LUAD cells in vitro and in vivo, induced apoptosis of LUAD cells and sensitized LUAD cells to cisplatin. Further investigations revealed that PSMA5 overexpression inhibited cell apoptosis by activating the janus kinase (JAK)/signal transducer and activator of transcription (STAT) signaling pathway in LUAD cells. In total, our results demonstrate that PSMA5 may function as a prognostic factor in LUAD. In addition, PSMA5 is a promising therapeutic target for LUAD, as its depletion induces cell apoptosis by inhibiting the JAK/STAT pathway.
Collapse
Affiliation(s)
- Feng Lu
- Joint National Laboratory for Antibody Drug Engineering, the First Affiliated Hospital, School of Basic Medical Sciences, Henan University, Kaifeng, China.,Department of Immunology, School of Basic Medical Sciences, Henan University, Kaifeng, China
| | - Jing Zhou
- Joint National Laboratory for Antibody Drug Engineering, the First Affiliated Hospital, School of Basic Medical Sciences, Henan University, Kaifeng, China
| | - Qing Chen
- Joint National Laboratory for Antibody Drug Engineering, the First Affiliated Hospital, School of Basic Medical Sciences, Henan University, Kaifeng, China
| | - Jianling Zhu
- Joint National Laboratory for Antibody Drug Engineering, the First Affiliated Hospital, School of Basic Medical Sciences, Henan University, Kaifeng, China
| | - Xiaowei Zheng
- Joint National Laboratory for Antibody Drug Engineering, the First Affiliated Hospital, School of Basic Medical Sciences, Henan University, Kaifeng, China.,Department of Clinical Laboratory, Puyang Hospital of Traditional Chinese Medicine, Puyang, China
| | - Na Fang
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Henan University, Kaifeng, China
| | - Ling Qiao
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Henan University, Kaifeng, China
| |
Collapse
|
8
|
Chen L, Zhang Y, Shu X, Chen Q, Wei T, Wang H, Wang X, Wu Q, Zhang X, Liu X, Zheng S, Huang L, Xiao J, Jiang C, Yang B, Wang Z, Guo X. Proteasome regulation by reversible tyrosine phosphorylation at the membrane. Oncogene 2021; 40:1942-1956. [PMID: 33603165 PMCID: PMC7990385 DOI: 10.1038/s41388-021-01674-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2020] [Revised: 12/21/2020] [Accepted: 01/21/2021] [Indexed: 01/30/2023]
Abstract
Reversible phosphorylation has emerged as an important mechanism for regulating 26S proteasome function in health and disease. Over 100 phospho-tyrosine sites of the human proteasome have been detected, and yet their function and regulation remain poorly understood. Here we show that the 19S subunit Rpt2 is phosphorylated at Tyr439, a strictly conserved residue within the C-terminal HbYX motif of Rpt2 that is essential for 26S proteasome assembly. Unexpectedly, we found that Y439 phosphorylation depends on Rpt2 membrane localization mediated by its N-myristoylation. Multiple receptors tyrosine kinases can trigger Rpt2-Y439 phosphorylation by activating Src, a N-myristoylated tyrosine kinase. Src directly phosphorylates Rpt2-Y439 in vitro and negatively regulates 26S proteasome activity at cellular membranes, which can be reversed by the membrane-associated isoform of protein tyrosine phosphatase nonreceptor type 2 (PTPN2). In H1975 lung cancer cells with activated Src, blocking Rpt2-Y439 phosphorylation by the Y439F mutation conferred partial resistance to the Src inhibitor saracatinib both in vitro and in a mouse xenograft tumor model, and caused significant changes of cellular responses to saracatinib at the proteome level. Our study has defined a novel mechanism involved in the spatial regulation of proteasome function and provided new insights into tyrosine kinase inhibitor-based anticancer therapies.
Collapse
Affiliation(s)
- Lu Chen
- Zhejiang Provincial Key Laboratory for Cancer Molecular Cell Biology, Life Sciences Institute, Zhejiang University, Hangzhou, China
| | - Yanan Zhang
- Zhejiang Provincial Key Laboratory for Cancer Molecular Cell Biology, Life Sciences Institute, Zhejiang University, Hangzhou, China
| | - Xin Shu
- Zhejiang Provincial Key Laboratory for Cancer Molecular Cell Biology, Life Sciences Institute, Zhejiang University, Hangzhou, China
| | - Qiong Chen
- Zhejiang Provincial Key Laboratory for Cancer Molecular Cell Biology, Life Sciences Institute, Zhejiang University, Hangzhou, China
| | - Tiantian Wei
- Academy for Advanced Interdisciplinary Studies, Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, China
| | - Heman Wang
- Zhejiang Provincial Key Laboratory for Cancer Molecular Cell Biology, Life Sciences Institute, Zhejiang University, Hangzhou, China
| | - Xiaorong Wang
- Departments of Physiology and Biophysics and of Developmental and Cell Biology, University of California, Irvine, CA, USA
| | - Qirou Wu
- Zhejiang Provincial Key Laboratory for Cancer Molecular Cell Biology, Life Sciences Institute, Zhejiang University, Hangzhou, China
| | - Xiaomei Zhang
- Zhejiang Provincial Key Laboratory for Cancer Molecular Cell Biology, Life Sciences Institute, Zhejiang University, Hangzhou, China
| | - Xiaoyan Liu
- Zhejiang Provincial Key Laboratory for Cancer Molecular Cell Biology, Life Sciences Institute, Zhejiang University, Hangzhou, China
| | - Suya Zheng
- Zhejiang Provincial Key Laboratory for Cancer Molecular Cell Biology, Life Sciences Institute, Zhejiang University, Hangzhou, China
| | - Lan Huang
- Departments of Physiology and Biophysics and of Developmental and Cell Biology, University of California, Irvine, CA, USA
| | - Junyu Xiao
- Academy for Advanced Interdisciplinary Studies, Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, China
| | - Chao Jiang
- Zhejiang Provincial Key Laboratory for Cancer Molecular Cell Biology, Life Sciences Institute, Zhejiang University, Hangzhou, China
| | - Bing Yang
- Zhejiang Provincial Key Laboratory for Cancer Molecular Cell Biology, Life Sciences Institute, Zhejiang University, Hangzhou, China
| | - Zhiping Wang
- Center for Neuroscience and Department of Neurology of Second Affiliated Hospital, NHC and CAMS Key Laboratory of Medical Neurobiology, Zhejiang University School of Medicine, Hangzhou, China.
| | - Xing Guo
- Zhejiang Provincial Key Laboratory for Cancer Molecular Cell Biology, Life Sciences Institute, Zhejiang University, Hangzhou, China.
| |
Collapse
|
9
|
Expanding the role of proteasome homeostasis in Parkinson's disease: beyond protein breakdown. Cell Death Dis 2021; 12:154. [PMID: 33542205 PMCID: PMC7862491 DOI: 10.1038/s41419-021-03441-0] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Revised: 01/13/2021] [Accepted: 01/15/2021] [Indexed: 02/06/2023]
Abstract
Proteasome is the principal hydrolytic machinery responsible for the great majority of protein degradation. The past three decades have testified prominent advances about proteasome involved in almost every aspect of biological processes. Nonetheless, inappropriate increase or decrease in proteasome function is regarded as a causative factor in several diseases. Proteasome abundance and proper assembly need to be precisely controlled. Indeed, various neurodegenerative diseases including Parkinson's disease (PD) share a common pathological feature, intracellular protein accumulation such as α-synuclein. Proteasome activation may effectively remove aggregates and prevent the neurodegeneration in PD, which provides a potential application for disease-modifying treatment. In this review, we build on the valuable discoveries related to different types of proteolysis by distinct forms of proteasome, and how its regulatory and catalytic particles promote protein elimination. Additionally, we summarize the emerging ideas on the proteasome homeostasis regulation by targeting transcriptional, translational, and post-translational levels. Given the imbalanced proteostasis in PD, the strategies for intensifying proteasomal degradation are advocated as a promising approach for PD clinical intervention.
Collapse
|
10
|
JANNUZZI AT, ARSLAN S, ALPERTUNGA B, KARADEMİR YILMAZ B. Proteasomal system related stress response in different cancer cell lines. CLINICAL AND EXPERIMENTAL HEALTH SCIENCES 2020. [DOI: 10.33808/clinexphealthsci.802815] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
|
11
|
Mishra S, Dunkerly-Eyring BL, Keceli G, Ranek MJ. Phosphorylation Modifications Regulating Cardiac Protein Quality Control Mechanisms. Front Physiol 2020; 11:593585. [PMID: 33281625 PMCID: PMC7689282 DOI: 10.3389/fphys.2020.593585] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Accepted: 09/28/2020] [Indexed: 12/12/2022] Open
Abstract
Many forms of cardiac disease, including heart failure, present with inadequate protein quality control (PQC). Pathological conditions often involve impaired removal of terminally misfolded proteins. This results in the formation of large protein aggregates, which further reduce cellular viability and cardiac function. Cardiomyocytes have an intricately collaborative PQC system to minimize cellular proteotoxicity. Increased expression of chaperones or enhanced clearance of misfolded proteins either by the proteasome or lysosome has been demonstrated to attenuate disease pathogenesis, whereas reduced PQC exacerbates pathogenesis. Recent studies have revealed that phosphorylation of key proteins has a potent regulatory role, both promoting and hindering the PQC machinery. This review highlights the recent advances in phosphorylations regulating PQC, the impact in cardiac pathology, and the therapeutic opportunities presented by harnessing these modifications.
Collapse
Affiliation(s)
- Sumita Mishra
- Division of Cardiology, Department of Medicine, The Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Brittany L Dunkerly-Eyring
- Department of Pharmacology and Molecular Sciences, Johns Hopkins University, Baltimore, MD, United States
| | - Gizem Keceli
- Division of Cardiology, Department of Medicine, The Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Mark J Ranek
- Division of Cardiology, Department of Medicine, The Johns Hopkins University School of Medicine, Baltimore, MD, United States
| |
Collapse
|
12
|
Tsimokha AS, Artamonova TO, Diakonov EE, Khodorkovskii MA, Tomilin AN. Post-Translational Modifications of Extracellular Proteasome. Molecules 2020; 25:molecules25153504. [PMID: 32752045 PMCID: PMC7435879 DOI: 10.3390/molecules25153504] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Revised: 07/27/2020] [Accepted: 07/30/2020] [Indexed: 12/29/2022] Open
Abstract
The ubiquitin-proteasome system (UPS) is one of the major protein degradation pathways in eukaryotic cells. Abnormal functioning of this system has been observed in cancer and neurological diseases. The 20S proteasomes, essential components of the UPS, are present not only within the cells but also in the extracellular space, and their concentration in blood plasma has been found to be elevated and dependent upon the disease state, being of prognostic significance in patients suffering from cancer, liver diseases, and autoimmune diseases. However, functions of extracellular proteasomes and mechanisms of their release by cells remain largely unknown. The main mechanism of proteasome activity regulation is provided by modulation of their composition and post-translational modifications (PTMs). Moreover, diverse PTMs of proteins are known to participate in the loading of specific elements into extracellular vesicles. Since previous studies have revealed that the transport of extracellular proteasomes may occur via extracellular vesicles, we have set out to explore the PTMs of extracellular proteasomes in comparison to cellular counterparts. In this work, cellular and extracellular proteasomes were affinity purified and separated by SDS-PAGE for subsequent trypsinization and matrix-assisted laser desorption/ionization (MALDI) Fourier transform ion cyclotron resonance (FT-ICR) mass spectrometry (MS) analysis. In total, we could identify 64 and 55 PTM sites in extracellular and cellular proteasomes, respectively, including phosphorylation, ubiquitination, acetylation, and succinylation. We observed novel sites of acetylation at K238 and K192 of the proteasome subunits β2 and β3, respectively, that are specific for extracellular proteasomes. Moreover, cellular proteasomes show specific acetylation at K227 of α2 and ubiquitination at K201 of β3. Interestingly, succinylation of β6 at the residue K228 seems not to be present exclusively in extracellular proteasomes, whereas both extracellular and cellular proteasomes may also be acetylated at this site. The same situation takes place at K201 of the β3 subunit where ubiquitination is seemingly specific for cellular proteasomes. Moreover, crosstalk between acetylation, ubiquitination, and succinylation has been observed in the subunit α3 of both proteasome populations. These data will serve as a basis for further studies, aimed at dissection of the roles of extracellular proteasome-specific PTMs in terms of the function of these proteasomes and mechanism of their transport into extracellular space.
Collapse
Affiliation(s)
- Anna S. Tsimokha
- Institute of Cytology of the Russian Academy of Sciences, 4 Tikhoretsky Ave., 194064 Saint-Petersburg, Russia; (T.O.A.); (E.E.D.); (M.A.K.); (A.N.T.)
- Correspondence: ; Tel.: +7-812-297-1829; Fax: +7-812-297-0341
| | - Tatiana O. Artamonova
- Institute of Cytology of the Russian Academy of Sciences, 4 Tikhoretsky Ave., 194064 Saint-Petersburg, Russia; (T.O.A.); (E.E.D.); (M.A.K.); (A.N.T.)
- Institute of Nanobiotechnologies, Peter the Great St. Petersburg Polytechnic University, 29 Polytechnicheskaya Str., 195251 Saint-Petersburg, Russia
| | - Egor E. Diakonov
- Institute of Cytology of the Russian Academy of Sciences, 4 Tikhoretsky Ave., 194064 Saint-Petersburg, Russia; (T.O.A.); (E.E.D.); (M.A.K.); (A.N.T.)
| | - Mikhail A. Khodorkovskii
- Institute of Cytology of the Russian Academy of Sciences, 4 Tikhoretsky Ave., 194064 Saint-Petersburg, Russia; (T.O.A.); (E.E.D.); (M.A.K.); (A.N.T.)
- Institute of Nanobiotechnologies, Peter the Great St. Petersburg Polytechnic University, 29 Polytechnicheskaya Str., 195251 Saint-Petersburg, Russia
| | - Alexey N. Tomilin
- Institute of Cytology of the Russian Academy of Sciences, 4 Tikhoretsky Ave., 194064 Saint-Petersburg, Russia; (T.O.A.); (E.E.D.); (M.A.K.); (A.N.T.)
| |
Collapse
|
13
|
Chen NX, Liu K, Liu X, Zhang XX, Han DY. Induction and Regulation of the Immunoproteasome Subunit β5i (PSMB8) in Laryngeal and Hypopharyngeal Carcinoma Cells. Med Sci Monit 2020; 26:e923621. [PMID: 32680979 PMCID: PMC7366787 DOI: 10.12659/msm.923621] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Background The ubiquitin–proteasome pathway (UPP) is closely associated with the occurrence and progression of cancer, and the 5i immunoproteasome subunit is an important antitumor target in UPP. This study aimed to characterize the regulation of the immunoproteasome subunit β5i (PSMB8) in JHU-011 laryngeal carcinoma cells and FaDu hypopharyngeal carcinoma cells to explore a new target for the treatment of laryngeal and hypopharyngeal carcinomas. Material/Methods JHU-011 and FaDu cells were used as effector cells in this study. By means of 60Co γ-irradiation, the construction of stable cell lines of the silenced proto-oncogene c-Abl, and the addition of exogenous tyrosine kinase inhibitor (TKI) and activator, the transcription and protein expression levels of PSMB8 and its alternatively spliced isoforms in both cell lines were detected by real-time fluorescence quantitative polymerase chain reaction (RT-PCR) and Western blot. Results Ionizing radiation upregulated the transcription level of the alternatively spliced isoform of PSMB8, E2, in both cell lines, thereby upregulating the mRNA and protein levels of PSMB8. The silencing of the proto-oncogene c-Abl and the activation and inhibition of its kinetic kinase product can affect the transcription and protein levels of PSMB8. Conclusions Ionizing radiation can significantly upregulate the mRNA and protein levels of PSMB8, which happens through the upregulation of its splicing isoform E2. The proto-oncogene c-Abl and its kinetic kinase protein product can regulate the transcription and protein expression levels of PSMB8 and its alternatively spliced isoforms.
Collapse
Affiliation(s)
- Nan-Xiang Chen
- College of Otolaryngology, Head and Neck Surgery, Chinese People's Liberation Army (PLA) General Hospital, Chinese PLA Medical School, Beijing, China (mainland)
| | - Kun Liu
- College of Otolaryngology, Head and Neck Surgery, Chinese People's Liberation Army (PLA) General Hospital, Chinese PLA Medical School, Beijing, China (mainland)
| | - Xuan Liu
- Department of Biotechnology, Beijing Institute of Biotechnology, Beijing, China (mainland)
| | - Xin-Xin Zhang
- College of Otolaryngology, Head and Neck Surgery, Chinese People's Liberation Army (PLA) General Hospital, Chinese PLA Medical School, Beijing, China (mainland)
| | - Dong-Yi Han
- College of Otolaryngology, Head and Neck Surgery, Chinese People's Liberation Army (PLA) General Hospital, Chinese PLA Medical School, Beijing, China (mainland)
| |
Collapse
|
14
|
Wang X, Meul T, Meiners S. Exploring the proteasome system: A novel concept of proteasome inhibition and regulation. Pharmacol Ther 2020; 211:107526. [PMID: 32173559 DOI: 10.1016/j.pharmthera.2020.107526] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2019] [Accepted: 03/08/2020] [Indexed: 12/13/2022]
Abstract
The proteasome is a well-identified therapeutic target for cancer treatment. It acts as the main protein degradation system in the cell and degrades key mediators of cell growth, survival and function. The term "proteasome" embraces a whole family of distinct complexes, which share a common proteolytic core, the 20S proteasome, but differ by their attached proteasome activators. Each of these proteasome complexes plays specific roles in the control of cellular function. In addition, distinct proteasome interacting proteins regulate proteasome activity in subcellular compartments and in response to cellular signals. Proteasome activators and regulators may thus serve as building blocks to fine-tune proteasome function in the cell according to cellular needs. Inhibitors of the proteasome, e.g. the FDA approved drugs Velcade™, Kyprolis™, Ninlaro™, inactivate the catalytic 20S core and effectively block protein degradation of all proteasome complexes in the cell resulting in inhibition of cell growth and induction of apoptosis. Efficacy of these inhibitors, however, is hampered by their pronounced cytotoxic side-effects as well as by the emerging development of resistance to catalytic proteasome inhibitors. Targeted inhibition of distinct buiding blocks of the proteasome system, i.e. proteasome activators or regulators, represents an alternative strategy to overcome these limitations. In this review, we stress the importance of the diversity of the proteasome complexes constituting an entire proteasome system. Our building block concept provides a rationale for the defined targeting of distinct proteasome super-complexes in disease. We thereby aim to stimulate the development of innovative therapeutic approaches beyond broad catalytic proteasome inhibition.
Collapse
Affiliation(s)
- Xinyuan Wang
- Comprehensive Pneumology Center (CPC), University Hospital of the Ludwig-Maximilians-University (LMU) and Helmholtz Zentrum München, German Center for Lung Research (DZL), 81377 Munich, Germany
| | - Thomas Meul
- Comprehensive Pneumology Center (CPC), University Hospital of the Ludwig-Maximilians-University (LMU) and Helmholtz Zentrum München, German Center for Lung Research (DZL), 81377 Munich, Germany
| | - Silke Meiners
- Comprehensive Pneumology Center (CPC), University Hospital of the Ludwig-Maximilians-University (LMU) and Helmholtz Zentrum München, German Center for Lung Research (DZL), 81377 Munich, Germany.
| |
Collapse
|
15
|
Coux O, Zieba BA, Meiners S. The Proteasome System in Health and Disease. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1233:55-100. [DOI: 10.1007/978-3-030-38266-7_3] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
16
|
Xia S, Tang Q, Wang X, Zhang L, Jia L, Wu D, Xu P, Zhang X, Tang G, Yang T, Feng Z, Lu L. Overexpression of PSMA7 predicts poor prognosis in patients with gastric cancer. Oncol Lett 2019; 18:5341-5349. [PMID: 31612044 PMCID: PMC6781669 DOI: 10.3892/ol.2019.10879] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2018] [Accepted: 07/19/2019] [Indexed: 12/27/2022] Open
Abstract
Gastric cancer (GC) is the fourth most common tumor and the second most common cause of cancer-associated mortality worldwide. Current tumor biomarkers for GC, such as serum carcinoembryonic antigen and carbohydrate antigen 19-9, are not ideal due to their limited role as prognostic indicators for GC. Proteasome subunit α7 (PSMA7) is a multifunctional protein, which has been revealed to be involved in the development and progression of various types of malignancy. However, little is known about the role of PSMA7 in GC. In the present study, PSMA7 was identified to be overexpressed at the mRNA and protein levels in GC tissues, compared with in non-tumor tissues, using reverse transcription-quantitative PCR and immunohistochemistry. Furthermore, PSMA7 expression is associated with tumor invasion, lymph node metastasis, distant metastasis, and Tumor-Node-Metastasis stage. Univariate and multivariate Cox regression analysis identified that PSMA7 expression is an independent prognostic factor for poor survival. Kaplan-Meier survival curves revealed that high PSMA7 expression is associated with a poor prognosis in patients with GC. Overall, the results of the present study suggested that PSMA7 may be a promising biomarker for the prognosis of GC, and may represent a new diagnostic marker and molecular therapeutic target for GC.
Collapse
Affiliation(s)
- Shujing Xia
- Department of Pathology, Nanjing Medical University, Nanjing, Jiangsu 211166, P.R. China.,Department of Gastroenterology, Affiliated Xinghua People's Hospital of Yangzhou University Medical College, Xinghua, Jiangsu 225700, P.R. China.,Department of Gastroenterology, Shanghai General Hospital, Nanjing Medical University, Shanghai 200080, P.R. China
| | - Qi Tang
- Key Laboratory of Antibody Technique of Ministry of Health, Nanjing Medical University, Nanjing, Jiangsu 211166, P.R. China
| | - Xudong Wang
- The Clinical Bio-Bank, Affiliated Hospital of Nantong University, Nantong, Jiangsu 226019, P.R. China
| | - Lili Zhang
- Department of Gastroenterology, Affiliated Xinghua People's Hospital of Yangzhou University Medical College, Xinghua, Jiangsu 225700, P.R. China
| | - Lizhou Jia
- Department of Pathology, Nanjing Medical University, Nanjing, Jiangsu 211166, P.R. China
| | - Duo Wu
- Department of Gastrointestinal Surgery, Affiliated Xinghua People's Hospital of Yangzhou University Medical College, Xinghua, Jiangsu 225700, P.R. China
| | - Pingxiang Xu
- Department of Gastrointestinal Surgery, Affiliated Xinghua People's Hospital of Yangzhou University Medical College, Xinghua, Jiangsu 225700, P.R. China
| | - Xiumei Zhang
- Department of Pathology, Affiliated Xinghua People's Hospital of Yangzhou University Medical College, Xinghua, Jiangsu 225700, P.R. China
| | - Genxiong Tang
- Department of Pathology, Nanjing Medical University, Nanjing, Jiangsu 211166, P.R. China
| | - Tingting Yang
- Department of Pathology, Nanjing Medical University, Nanjing, Jiangsu 211166, P.R. China
| | - Zhenqing Feng
- Department of Pathology, Nanjing Medical University, Nanjing, Jiangsu 211166, P.R. China.,Key Laboratory of Antibody Technique of Ministry of Health, Nanjing Medical University, Nanjing, Jiangsu 211166, P.R. China.,Jiangsu Collaborative Innovation Center for Cancer Personalized Medicine, Cancer Center, Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China.,Key Laboratory of Cancer Biomarkers, Prevention and Treatment, Cancer Center, Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
| | - Lungen Lu
- Department of Gastroenterology, Shanghai General Hospital, Nanjing Medical University, Shanghai 200080, P.R. China
| |
Collapse
|
17
|
Kors S, Geijtenbeek K, Reits E, Schipper-Krom S. Regulation of Proteasome Activity by (Post-)transcriptional Mechanisms. Front Mol Biosci 2019; 6:48. [PMID: 31380390 PMCID: PMC6646590 DOI: 10.3389/fmolb.2019.00048] [Citation(s) in RCA: 72] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2019] [Accepted: 06/11/2019] [Indexed: 12/23/2022] Open
Abstract
Intracellular protein synthesis, folding, and degradation are tightly controlled processes to ensure proper protein homeostasis. The proteasome is responsible for the degradation of the majority of intracellular proteins, which are often targeted for degradation via polyubiquitination. However, the degradation rate of proteins is also affected by the capacity of proteasomes to recognize and degrade these substrate proteins. This capacity is regulated by a variety of proteasome modulations including (1) changes in complex composition, (2) post-translational modifications, and (3) altered transcription of proteasomal subunits and activators. Various diseases are linked to proteasome modulation and altered proteasome function. A better understanding of these modulations may offer new perspectives for therapeutic intervention. Here we present an overview of these three proteasome modulating mechanisms to give better insight into the diversity of proteasomes.
Collapse
Affiliation(s)
- Suzan Kors
- Department of Medical Biology, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
| | - Karlijne Geijtenbeek
- Department of Medical Biology, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
| | - Eric Reits
- Department of Medical Biology, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
| | - Sabine Schipper-Krom
- Department of Medical Biology, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
| |
Collapse
|
18
|
夏 淑, 张 莉, 成 凤, 冯 振, 陆 伦. [Expression of PSMA 7 and its effect on proliferation, invasion, migration and tumorigenesis of gastric cancer]. NAN FANG YI KE DA XUE XUE BAO = JOURNAL OF SOUTHERN MEDICAL UNIVERSITY 2019; 39:387-393. [PMID: 31068280 PMCID: PMC6743988 DOI: 10.12122/j.issn.1673-4254.2019.04.02] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 03/02/2019] [Indexed: 11/24/2022]
Abstract
OBJECTIVE To study the expression of PSMA7 and its effect on proliferation, invasion and migration of gastric cancer and subcutaneous tumorigenesis in nude mice. >and subcutaneous tumorigenesis in nude mice. METHODS Specimens of tumor tissues and paired adjacent tissues were collected from 60 patients with gastric cancer for detecting the expression levels of PSMA7 using immunohistochemical method. Gastric cancer cell line SGC7901 was transfected with a lentiviral vector to inhibit PSMA7 expression, and the changes in cell proliferation and invasion were observed using cell counting kit-8 (CCK-8), clone formation assay and Transwell assay. A BALB/c mouse model bearing subcutaneous gastric cancer xenograft was established using SGC7901 cells with stable PSMA7 knockdown to assess the effect of low expression of PSMA7 on xenograft growth. RESULTS Gastric cancer tissues expressed significantly higher levels of PSMA7 than the paired adjacent tissues (P < 0.05). In SGC7901 cells, interference of PSMA7 expression significantly inhibited the cell proliferation and invasion (P < 0.05). In the tumor-bearing BALB/c mice, the xenografts derived from SGC7901 cells with PSMA7 expression interference showed significant growth suppression as compared with the control xenografts (P < 0.05). CONCLUSIONS PPSMA 7 is overexpressed in gastric cancer tissues, and PSMA7 knockdown inhibits the proliferation, invasion, migration and subcutaneous tumorigenesis of gastric cancer cells in nude mice.
Collapse
Affiliation(s)
- 淑晶 夏
- 南京医科大学病理系Department of Pathology, Nanjing Medical University, Nanjing 211166, China
- 国家卫健委抗体技术重点实验室,南京 211166Key Laboratory of Antibody Technology of National Health Commission, Nanjing 211166, China
- 南京医科大学附属上海一院临床医学院消化科,上海 201600Department of Gastroenterology, Shanghai General Hospital Affiliated to Nanjing Medical University, Shanghai 201600, China
- 兴化市人民医院消化内科,江苏 兴化 225700Department of Gastroenterology, Xinghua People's Hospital, Xinghua 225700, China
| | - 莉莉 张
- 南京医科大学附属上海一院临床医学院消化科,上海 201600Department of Gastroenterology, Shanghai General Hospital Affiliated to Nanjing Medical University, Shanghai 201600, China
| | - 凤干 成
- 南京医科大学附属上海一院临床医学院消化科,上海 201600Department of Gastroenterology, Shanghai General Hospital Affiliated to Nanjing Medical University, Shanghai 201600, China
| | - 振卿 冯
- 南京医科大学病理系Department of Pathology, Nanjing Medical University, Nanjing 211166, China
- 兴化市人民医院消化内科,江苏 兴化 225700Department of Gastroenterology, Xinghua People's Hospital, Xinghua 225700, China
| | - 伦根 陆
- 国家卫健委抗体技术重点实验室,南京 211166Key Laboratory of Antibody Technology of National Health Commission, Nanjing 211166, China
| |
Collapse
|
19
|
Mahdavi M, Nassiri M, Kooshyar MM, Vakili‐Azghandi M, Avan A, Sandry R, Pillai S, Lam AK, Gopalan V. Hereditary breast cancer; Genetic penetrance and current status with BRCA. J Cell Physiol 2018; 234:5741-5750. [DOI: 10.1002/jcp.27464] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2018] [Accepted: 08/29/2018] [Indexed: 12/14/2022]
Affiliation(s)
- Morteza Mahdavi
- Institute of Biotechnology, Ferdowsi University of Mashhad Mashhad Iran
| | | | | | | | - Amir Avan
- Metabolic Syndrome Research Center, Mashhad University of Medical Sciences Mashhad Iran
- Cancer Research Center and Surgical Oncology Research Center, Mashhad University of Medical Sciences Mashhad Iran
- School of Medicine, Griffith University Gold Coast QLD
| | - Ryan Sandry
- Department of Modern Sciences and Technologies Faculty of Medicine, Mashhad University of Medical Sciences Mashhad Iran
| | - Suja Pillai
- School of Biomedical Sciences, Faculty of Medicine, The University of Queensland Brisbane Australia
| | | | - Vinod Gopalan
- Department of Modern Sciences and Technologies Faculty of Medicine, Mashhad University of Medical Sciences Mashhad Iran
| |
Collapse
|
20
|
Sugimoto K, Hiwasa T, Shibuya K, Hirano S, Beppu M, Isose S, Arai K, Takiguchi M, Kuwabara S, Mori M. Novel autoantibodies against the proteasome subunit PSMA7 in amyotrophic lateral sclerosis. J Neuroimmunol 2018; 325:54-60. [PMID: 30390597 DOI: 10.1016/j.jneuroim.2018.09.013] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2018] [Revised: 09/27/2018] [Accepted: 09/30/2018] [Indexed: 12/12/2022]
Abstract
OBJECTIVE To identify autoantibodies using sera from ALS patients and elucidate their roles in disease pathology. METHODS An immunological screening was performed with a phage expression library SEREX method using sera from 3 ALS patients to identify ALS-related autoantibodies. Levels of antibodies identified by SEREX were measured in 33 ALS patients and 30 normal controls (NCs) by AlphaLISA using recombinant non-full-length proteins. The results were then validated by ELISA using full-length proteins in 71 ALS patients, 30 NCs and 34 disease controls (DCs). The relationship between the titres and clinical profiles of ALS patients were examined. RESULTS Four autoantibodies identified by SEREX were proteasome subunit alpha type 7 (PSMA7), vimentin, hydroxymethylbilane synthase and TBC1 domain family member 2 (TBC1D2). AlphaLISA revealed that only the anti-PSMA7 and anti-TBC1D2 levels were significantly different between the ALS and NCs groups. ELISA showed that only the levels of antibody against PSMA7, involved in protein degradation by the ubiquitin-proteasome pathway (UPP), were higher in the ALS group than both the NC (P < .01) and DC (P = .034) groups. Anti-PSMA7 levels tended to be negatively correlated with the logarithm of disease duration (P = .052) and were significantly positively correlated with the logarithm of creatine kinase levels (P = .011). The anti-PSMA7 antibody levels were different between patients with and without dysphagia (P < .01). CONCLUSIONS Serum anti-PSMA7 antibody might be a disease-promoting factor in early-stage ALS and might be a biomarker of ALS. Anti-PSMA7 autoantibody might contribute to the pathogenesis of ALS, possibly via its role in the UPP.
Collapse
Affiliation(s)
- Kazuo Sugimoto
- Department of Neurology, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Takaki Hiwasa
- Department of Biochemistry and Genetics, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Kazutomo Shibuya
- Department of Neurology, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Shigeki Hirano
- Department of Neurology, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Minako Beppu
- Department of Neurology, Graduate School of Medicine, Chiba University, Chiba, Japan; Department of Molecular Diagnosis, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Sagiri Isose
- Department of Neurology, National Hospital Organization Chiba-East-Hospital, Chiba, Japan
| | - Kimihito Arai
- Department of Neurology, National Hospital Organization Chiba-East-Hospital, Chiba, Japan
| | - Masaki Takiguchi
- Department of Biochemistry and Genetics, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Satoshi Kuwabara
- Department of Neurology, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Masahiro Mori
- Department of Neurology, Graduate School of Medicine, Chiba University, Chiba, Japan.
| |
Collapse
|
21
|
Nath SR, Yu Z, Gipson TA, Marsh GB, Yoshidome E, Robins DM, Todi SV, Housman DE, Lieberman AP. Androgen receptor polyglutamine expansion drives age-dependent quality control defects and muscle dysfunction. J Clin Invest 2018; 128:3630-3641. [PMID: 29809168 DOI: 10.1172/jci99042] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2017] [Accepted: 05/24/2018] [Indexed: 12/28/2022] Open
Abstract
Skeletal muscle has emerged as a critical, disease-relevant target tissue in spinal and bulbar muscular atrophy, a degenerative disorder of the neuromuscular system caused by a CAG/polyglutamine (polyQ) expansion in the androgen receptor (AR) gene. Here, we used RNA-sequencing (RNA-Seq) to identify pathways that are disrupted in diseased muscle using AR113Q knockin mice. This analysis unexpectedly identified substantially diminished expression of numerous ubiquitin/proteasome pathway genes in AR113Q muscle, encoding approximately 30% of proteasome subunits and 20% of E2 ubiquitin conjugases. These changes were age, hormone, and glutamine length dependent and arose due to a toxic gain of function conferred by the mutation. Moreover, altered gene expression was associated with decreased levels of the proteasome transcription factor NRF1 and its activator DDI2 and resulted in diminished proteasome activity. Ubiquitinated ADRM1 was detected in AR113Q muscle, indicating the occurrence of stalled proteasomes in mutant mice. Finally, diminished expression of Drosophila orthologues of NRF1 or ADRM1 promoted the accumulation of polyQ AR protein and increased toxicity. Collectively, these data indicate that AR113Q muscle develops progressive proteasome dysfunction that leads to the impairment of quality control and the accumulation of polyQ AR protein, key features that contribute to the age-dependent onset and progression of this disorder.
Collapse
Affiliation(s)
- Samir R Nath
- Department of Pathology.,Medical Scientist Training Program, and.,Cellular and Molecular Biology Graduate Program, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | | | - Theresa A Gipson
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | - Gregory B Marsh
- Department of Pharmacology, Wayne State University School of Medicine, Detroit, Michigan, USA
| | | | - Diane M Robins
- Department of Human Genetics, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Sokol V Todi
- Department of Pharmacology, Wayne State University School of Medicine, Detroit, Michigan, USA
| | - David E Housman
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | | |
Collapse
|
22
|
Bustamante HA, González AE, Cerda-Troncoso C, Shaughnessy R, Otth C, Soza A, Burgos PV. Interplay Between the Autophagy-Lysosomal Pathway and the Ubiquitin-Proteasome System: A Target for Therapeutic Development in Alzheimer's Disease. Front Cell Neurosci 2018; 12:126. [PMID: 29867359 PMCID: PMC5954036 DOI: 10.3389/fncel.2018.00126] [Citation(s) in RCA: 63] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2018] [Accepted: 04/20/2018] [Indexed: 12/14/2022] Open
Abstract
Alzheimer’s disease (AD) is the most common cause of age-related dementia leading to severe irreversible cognitive decline and massive neurodegeneration. While therapeutic approaches for managing symptoms are available, AD currently has no cure. AD associates with a progressive decline of the two major catabolic pathways of eukaryotic cells—the autophagy-lysosomal pathway (ALP) and the ubiquitin-proteasome system (UPS)—that contributes to the accumulation of harmful molecules implicated in synaptic plasticity and long-term memory impairment. One protein recently highlighted as the earliest initiator of these disturbances is the amyloid precursor protein (APP) intracellular C-terminal membrane fragment β (CTFβ), a key toxic agent with deleterious effects on neuronal function that has become an important pathogenic factor for AD and a potential biomarker for AD patients. This review focuses on the involvement of regulatory molecules and specific post-translational modifications (PTMs) that operate in the UPS and ALP to control a single proteostasis network to achieve protein balance. We discuss how these aspects can contribute to the development of novel strategies to strengthen the balance of key pathogenic proteins associated with AD.
Collapse
Affiliation(s)
- Hianara A Bustamante
- Institute of Physiology, Faculty of Medicine, Universidad Austral de Chile, Valdivia, Chile.,Center for Interdisciplinary Studies on the Nervous System (CISNe), Universidad Austral de Chile, Valdivia, Chile
| | - Alexis E González
- Institute of Physiology, Faculty of Medicine, Universidad Austral de Chile, Valdivia, Chile.,Fundación Ciencia y Vida, Santiago, Chile
| | - Cristobal Cerda-Troncoso
- Centro de Biología Celular y Biomedicina (CEBICEM), Facultad de Medicina y Ciencia, Universidad San Sebastián, Santiago, Chile
| | - Ronan Shaughnessy
- Centro de Biología Celular y Biomedicina (CEBICEM), Facultad de Medicina y Ciencia, Universidad San Sebastián, Santiago, Chile.,Center for Aging and Regeneration (CARE), Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Carola Otth
- Center for Interdisciplinary Studies on the Nervous System (CISNe), Universidad Austral de Chile, Valdivia, Chile.,Institute of Clinical Microbiology, Faculty of Medicine, Universidad Austral de Chile, Valdivia, Chile
| | - Andrea Soza
- Centro de Biología Celular y Biomedicina (CEBICEM), Facultad de Medicina y Ciencia, Universidad San Sebastián, Santiago, Chile.,Center for Aging and Regeneration (CARE), Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Patricia V Burgos
- Institute of Physiology, Faculty of Medicine, Universidad Austral de Chile, Valdivia, Chile.,Centro de Biología Celular y Biomedicina (CEBICEM), Facultad de Medicina y Ciencia, Universidad San Sebastián, Santiago, Chile.,Center for Aging and Regeneration (CARE), Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| |
Collapse
|
23
|
Liu H, Cui Y, Wang GF, Dong Q, Yao Y, Li P, Cao C, Liu X. The nonreceptor tyrosine kinase c-Abl phosphorylates Runx1 and regulates Runx1-mediated megakaryocyte maturation. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2018; 1865:1060-1072. [PMID: 29730354 DOI: 10.1016/j.bbamcr.2018.05.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/28/2017] [Revised: 04/28/2018] [Accepted: 05/02/2018] [Indexed: 02/07/2023]
Abstract
The transcription factor Runx1 is an essential regulator of definitive hematopoiesis, megakaryocyte (MK) maturation, and lymphocyte differentiation. Runx1 mutations that interfere with its transcriptional activity are often present in leukemia patients. Recent work demonstrated that the transcriptional activity of Runx1 is regulated by kinase-mediated phosphorylation. In this study, we showed that c-Abl, but not Arg tyrosine kinase, associated with Runx1 both in cultured cells and in vitro. c-Abl-mediated tyrosine phosphorylation in the Runx1 transcription inhibition domain negatively regulated the transcriptional activity of Runx1 and inhibited Runx1-mediated MK maturation. Consistent with these findings, increased numbers of MKs were detected in the spleens and bone marrow of abl gene conditional knockout mice. Our findings demonstrate an important role of c-Abl kinase in Runx1-mediated MK maturation and platelet formation and provide a potential mechanism of Abl kinase-regulated hematopoiesis.
Collapse
Affiliation(s)
- Hainan Liu
- Beijing Institute of Biotechnology, 27 Taiping Rd, Haidian District, Beijing 100850, China
| | - Yan Cui
- Department of Laboratory Animal Science, Third Military Medical University, Chongqing 400038, China
| | - Guang-Fei Wang
- Beijing Institute of Biotechnology, 27 Taiping Rd, Haidian District, Beijing 100850, China
| | - Qincai Dong
- Beijing Institute of Biotechnology, 27 Taiping Rd, Haidian District, Beijing 100850, China
| | - Yebao Yao
- Beijing Institute of Biotechnology, 27 Taiping Rd, Haidian District, Beijing 100850, China
| | - Ping Li
- Beijing Institute of Biotechnology, 27 Taiping Rd, Haidian District, Beijing 100850, China
| | - Cheng Cao
- Beijing Institute of Biotechnology, 27 Taiping Rd, Haidian District, Beijing 100850, China.
| | - Xuan Liu
- Beijing Institute of Biotechnology, 27 Taiping Rd, Haidian District, Beijing 100850, China.
| |
Collapse
|
24
|
Sun S, Liu S, Zhang Z, Zeng W, Sun C, Tao T, Lin X, Feng XH. Phosphatase UBLCP1 controls proteasome assembly. Open Biol 2018; 7:rsob.170042. [PMID: 28539385 PMCID: PMC5451543 DOI: 10.1098/rsob.170042] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2017] [Accepted: 04/20/2017] [Indexed: 12/26/2022] Open
Abstract
Ubiquitin-like domain-containing C-terminal domain phosphatase 1 (UBLCP1), an FCP/SCP phosphatase family member, was identified as the first proteasome phosphatase. UBLCP1 binds to proteasome subunit Rpn1 and dephosphorylates the proteasome in vitro. However, it is still unclear which proteasome subunit(s) are the bona fide substrate(s) of UBLCP1 and the precise mechanism for proteasome regulation remains elusive. Here, we show that UBLCP1 selectively binds to the 19S regulatory particle (RP) through its interaction with Rpn1, but not the 20S core particle (CP) or the 26S proteasome holoenzyme. In the RP, UBLCP1 dephosphorylates the subunit Rpt1, impairs its ATPase activity, and consequently disrupts the 26S proteasome assembly, yet it has no effects on the RP assembly from precursor complexes. The Rpn1-binding and phosphatase activities of UBLCP1 are essential for its function on Rpt1 dephosphorylation and proteasome activity both in vivo and in vitro. Our study establishes the essential role of the UBLCP1/Rpn1/Rpt1 complex in regulating proteasome assembly.
Collapse
Affiliation(s)
- Shuangwu Sun
- Life Sciences Institute and Innovation Center for Cell Signaling Network, Zhejiang University, Hangzhou, Zhejiang, People's Republic of China
| | - Sisi Liu
- Life Sciences Institute and Innovation Center for Cell Signaling Network, Zhejiang University, Hangzhou, Zhejiang, People's Republic of China
| | - Zhengmao Zhang
- Michael E. DeBakey, Department of Surgery, Houston, TX, USA.,Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA
| | - Wang Zeng
- Life Sciences Institute and Innovation Center for Cell Signaling Network, Zhejiang University, Hangzhou, Zhejiang, People's Republic of China
| | - Chuang Sun
- Michael E. DeBakey, Department of Surgery, Houston, TX, USA
| | - Tao Tao
- State Key Laboratory of Stress Cell Biology, School of Life Sciences, Xiamen University, Xiamen, Fujian, People's Republic of China
| | - Xia Lin
- Michael E. DeBakey, Department of Surgery, Houston, TX, USA.,Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA
| | - Xin-Hua Feng
- Life Sciences Institute and Innovation Center for Cell Signaling Network, Zhejiang University, Hangzhou, Zhejiang, People's Republic of China .,Michael E. DeBakey, Department of Surgery, Houston, TX, USA.,Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA
| |
Collapse
|
25
|
Guo X, Huang X, Chen MJ. Reversible phosphorylation of the 26S proteasome. Protein Cell 2017; 8:255-272. [PMID: 28258412 PMCID: PMC5359188 DOI: 10.1007/s13238-017-0382-x] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2016] [Accepted: 01/26/2017] [Indexed: 01/09/2023] Open
Abstract
The 26S proteasome at the center of the ubiquitin-proteasome system (UPS) is essential for virtually all cellular processes of eukaryotes. A common misconception about the proteasome is that, once made, it remains as a static and uniform complex with spontaneous and constitutive activity for protein degradation. Recent discoveries have provided compelling evidence to support the exact opposite insomuch as the 26S proteasome undergoes dynamic and reversible phosphorylation under a variety of physiopathological conditions. In this review, we summarize the history and current understanding of proteasome phosphorylation, and advocate the idea of targeting proteasome kinases/phosphatases as a new strategy for clinical interventions of several human diseases.
Collapse
Affiliation(s)
- Xing Guo
- The Life Sciences Institute of Zhejiang University, Hangzhou, 310058, China.
| | - Xiuliang Huang
- Ministry of Education Key Laboratory of Protein Science, School of Life Sciences, Tsinghua University, Beijing, 100084, China
| | - Mark J Chen
- Department of Bioinformatics and Computational Biology, Genentech Inc., South San Francisco, CA, 94080, USA
| |
Collapse
|
26
|
Howell LA, Tomko RJ, Kusmierczyk AR. Putting it all together: intrinsic and extrinsic mechanisms governing proteasome biogenesis. ACTA ACUST UNITED AC 2017. [DOI: 10.1007/s11515-017-1439-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
|
27
|
Im E, Chung KC. Precise assembly and regulation of 26S proteasome and correlation between proteasome dysfunction and neurodegenerative diseases. BMB Rep 2017; 49:459-73. [PMID: 27312603 PMCID: PMC5227139 DOI: 10.5483/bmbrep.2016.49.9.094] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2016] [Indexed: 11/20/2022] Open
Abstract
Neurodegenerative diseases (NDs) often involve the formation of abnormal and toxic protein aggregates, which are thought to be the primary factor in ND occurrence and progression. Aged neurons exhibit marked increases in aggregated protein levels, which can lead to increased cell death in specific brain regions. As no specific drugs/therapies for treating the symptoms or/and progression of NDs are available, obtaining a complete understanding of the mechanism underlying the formation of protein aggregates is needed for designing a novel and efficient removal strategy. Intracellular proteolysis generally involves either the lysosomal or ubiquitin-proteasome system. In this review, we focus on the structure and assembly of the proteasome, proteasome-mediated protein degradation, and the multiple dynamic regulatory mechanisms governing proteasome activity. We also discuss the plausibility of the correlation between changes in proteasome activity and the occurrence of NDs. [BMB Reports 2016; 49(9): 459-473]
Collapse
Affiliation(s)
- Eunju Im
- Department of Systems Biology, College of Life Science and Biotechnology, Yonsei University, Seoul 03722, Korea
| | - Kwang Chul Chung
- Department of Systems Biology, College of Life Science and Biotechnology, Yonsei University, Seoul 03722, Korea
| |
Collapse
|
28
|
Torsello B, Bianchi C, Meregalli C, Di Stefano V, Invernizzi L, De Marco S, Bovo G, Brivio R, Strada G, Bombelli S, Perego RA. Arg tyrosine kinase modulates TGF-β1 production in human renal tubular cells under high-glucose conditions. J Cell Sci 2016; 129:2925-36. [PMID: 27298228 DOI: 10.1242/jcs.183640] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2015] [Accepted: 04/23/2016] [Indexed: 01/15/2023] Open
Abstract
Renal tubular cells are involved in the tubular interstitial fibrosis observed in diabetic nephropathy. It is debated whether epithelial-mesenchymal transition (EMT) affects tubular cells, which under high-glucose conditions overproduce transforming growth factor-β (TGF-β), a fibrogenic cytokine involved in interstitial fibrosis development. Our study investigated the involvement of non-receptor tyrosine kinase Arg (also called Abl2) in TGF-β production. Human primary tubular cell cultures exposed to high-glucose conditions were used. These cells showed an elongated morphology, stress fibers and vimentin increment but maintained most of the epithelial marker expression and distribution. In these cells exposed to high glucose, which overexpressed and secreted active TGF-β1, Arg protein and activity was downregulated. A further TGF-β1 increase was induced by Arg silencing with siRNA, as with the Arg tyrosine kinase inhibitor Imatinib. In the cells exposed to high glucose, reactive oxygen species (ROS)-dependent Arg kinase downregulation induced both RhoA activation, through p190RhoGAPA (also known as ARHGAP35) modulation, and proteasome activity inhibition. These data evidence a new specific involvement of Arg kinase into the regulation of TGF-β1 expression in tubular cells under high-glucose conditions and provide cues for new translational approaches in diabetic nephropathy.
Collapse
Affiliation(s)
- Barbara Torsello
- School of Medicine and Surgery, Milano-Bicocca University, Monza 20900, Italy
| | - Cristina Bianchi
- School of Medicine and Surgery, Milano-Bicocca University, Monza 20900, Italy
| | - Chiara Meregalli
- School of Medicine and Surgery, Milano-Bicocca University, Monza 20900, Italy
| | - Vitalba Di Stefano
- School of Medicine and Surgery, Milano-Bicocca University, Monza 20900, Italy
| | - Lara Invernizzi
- School of Medicine and Surgery, Milano-Bicocca University, Monza 20900, Italy
| | - Sofia De Marco
- School of Medicine and Surgery, Milano-Bicocca University, Monza 20900, Italy
| | - Giorgio Bovo
- Anatomo-Pathology Unit, San Gerardo Hospital, Monza 20900, Italy
| | - Rinaldo Brivio
- Clinical Pathology Unit, San Gerardo Hospital, Monza 20900, Italy
| | - Guido Strada
- Urology Unit, Bassini ICP Hospital, Milano 20092, Italy
| | - Silvia Bombelli
- School of Medicine and Surgery, Milano-Bicocca University, Monza 20900, Italy
| | - Roberto A Perego
- School of Medicine and Surgery, Milano-Bicocca University, Monza 20900, Italy
| |
Collapse
|
29
|
Riz I, Hawley TS, Hawley RG. KLF4-SQSTM1/p62-associated prosurvival autophagy contributes to carfilzomib resistance in multiple myeloma models. Oncotarget 2016; 6:14814-31. [PMID: 26109433 PMCID: PMC4558117 DOI: 10.18632/oncotarget.4530] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2015] [Accepted: 05/22/2015] [Indexed: 11/25/2022] Open
Abstract
Multiple myeloma (MM) is an incurable clonal plasma cell malignancy. Because of a high rate of immunoglobulin synthesis, the endoplasmic reticulum of MM cells is subjected to elevated basal levels of stress. Consequently, proteasome inhibitors, which exacerbate this stress by inhibiting ubiquitin-proteasome-mediated protein degradation, are an important new class of chemotherapeutic agents being used to combat this disease. However, MM cells still develop resistance to proteasome inhibitors such as carfilzomib. Toward this end, we have established carfilzomib-resistant derivatives of MM cell lines. We found that resistance to carfilzomib was associated with elevated levels of prosurvival autophagy, and Kruppel-like factor 4 (KLF4) was identified as a contributing factor. Expression levels as well as nuclear localization of KLF4 protein were elevated in MM cells with acquired carfilzomib resistance. Chromatin immunoprecipitations indicated that endogenous KLF4 bound to the promoter regions of the SQSTM1 gene encoding the ubiquitin-binding adaptor protein sequestosome/p62 that links the proteasomal and autophagic protein degradation pathways. Ectopic expression of KLF4 induced upregulation of SQSTM1. On the other hand, inhibitors of autophagy sensitized MM cells to carfilzomib, even in carfilzomib-resistant derivatives having increased expression of the multidrug resistance protein P-glycoprotein. Thus, we report here a novel function for KLF4, one of the Yamanaka reprogramming factors, as being a contributor to autophagy gene expression which moderates preclinical proteasome inhibitor efficacy in MM.
Collapse
Affiliation(s)
- Irene Riz
- Department of Anatomy and Regenerative Biology, The George Washington University, Washington, DC, USA
| | - Teresa S Hawley
- Flow Cytometry Core Facility, The George Washington University, Washington, DC, USA
| | - Robert G Hawley
- Department of Anatomy and Regenerative Biology, The George Washington University, Washington, DC, USA
| |
Collapse
|
30
|
Padmanabhan A, Vuong SAT, Hochstrasser M. Assembly of an Evolutionarily Conserved Alternative Proteasome Isoform in Human Cells. Cell Rep 2016; 14:2962-74. [PMID: 26997268 DOI: 10.1016/j.celrep.2016.02.068] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2015] [Revised: 01/25/2016] [Accepted: 02/16/2016] [Indexed: 11/17/2022] Open
Abstract
Targeted intracellular protein degradation in eukaryotes is largely mediated by the proteasome. Here, we report the formation of an alternative proteasome isoform in human cells, previously found only in budding yeast, that bears an altered subunit arrangement in the outer ring of the proteasome core particle. These proteasomes result from incorporation of an additional α4 (PSMA7) subunit in the position normally occupied by α3 (PSMA4). Assembly of "α4-α4" proteasomes depends on the relative cellular levels of α4 and α3 and on the proteasome assembly chaperone PAC3. The oncogenic tyrosine kinases ABL and ARG and the tumor suppressor BRCA1 regulate cellular α4 levels and formation of α4-α4 proteasomes. Cells primed to assemble α4-α4 proteasomes exhibit enhanced resistance to toxic metal ions. Taken together, our results establish the existence of an alternative mammalian proteasome isoform and suggest a potential role in enabling cells to adapt to environmental stresses.
Collapse
Affiliation(s)
- Achuth Padmanabhan
- Department of Molecular Biophysics and Biochemistry, Yale University, 266 Whitney Avenue, New Haven, CT 06520, USA
| | - Simone Anh-Thu Vuong
- Department of Molecular Biophysics and Biochemistry, Yale University, 266 Whitney Avenue, New Haven, CT 06520, USA
| | - Mark Hochstrasser
- Department of Molecular Biophysics and Biochemistry, Yale University, 266 Whitney Avenue, New Haven, CT 06520, USA.
| |
Collapse
|
31
|
Qi J, Ronai ZA. Dysregulation of ubiquitin ligases in cancer. Drug Resist Updat 2015; 23:1-11. [PMID: 26690337 DOI: 10.1016/j.drup.2015.09.001] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2015] [Revised: 08/31/2015] [Accepted: 09/02/2015] [Indexed: 02/08/2023]
Abstract
Ubiquitin ligases (UBLs) are critical components of the ubiquitin proteasome system (UPS), which governs fundamental processes regulating normal cellular homeostasis, metabolism, and cell cycle in response to external stress signals and DNA damage. Among multiple steps of the UPS system required to regulate protein ubiquitination and stability, UBLs define specificity, as they recognize and interact with substrates in a temporally- and spatially-regulated manner. Such interactions are required for substrate modification by ubiquitin chains, which marks proteins for recognition and degradation by the proteasome or alters their subcellular localization or assembly into functional complexes. UBLs are often deregulated in cancer, altering substrate availability or activity in a manner that can promote cellular transformation. Such deregulation can occur at the epigenetic, genomic, or post-translational levels. Alterations in UBL can be used to predict their contributions, affecting tumor suppressors or oncogenes in select tumors. Better understanding of mechanisms underlying UBL expression and activities is expected to drive the development of next generation modulators that can serve as novel therapeutic modalities. This review summarizes our current understanding of UBL deregulation in cancer and highlights novel opportunities for therapeutic interventions.
Collapse
Affiliation(s)
- Jianfei Qi
- University of Maryland School of Medicine, Baltimore, 21201, USA.
| | - Ze'ev A Ronai
- Sanford Burnham Prebys Medical Discovery Institute, La Jolla, 92037, USA.
| |
Collapse
|