1
|
He L, He J, Jiang T, Gong R, Wan X, Duan M, Chen Z, Cheng Y. Inhibition of UCH-L1 enhances immunotherapy efficacy in triple-negative breast cancer by stabilizing PD-L1. Eur J Pharmacol 2025; 1000:177743. [PMID: 40389130 DOI: 10.1016/j.ejphar.2025.177743] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2025] [Revised: 05/15/2025] [Accepted: 05/16/2025] [Indexed: 05/21/2025]
Abstract
Recent research indicates that programmed death 1 (PD-1) and programmed death-ligand 1 (PD-L1) inhibitors show promise in treating triple-negative breast cancer (TNBC), but their efficacy is lower than anticipated, especially when used alone. Therefore, enhancing the anti-tumor immune response strategy for TNBC is crucial. Ubiquitin carboxy-terminal hydrolase L1 (UCH-L1), involved in tumor cell regulation and a potential therapeutic target, has an undefined role in TNBC immunotherapy. In this study, we explored the inverse correlation between UCH-L1 and PD-L1 in TNBC patient tissues. Through in vitro experiments, we found that UCH-L1 negatively regulates PD-L1 by stabilizing the E3 ubiquitin ligase ariadne-1 homolog (ARIH1), which promotes PD-L1 ubiquitination and degradation. Further analysis in Balb/c mice xenograft tumors showed that UCH-L1 correlates with GZMB+/CD8+ T cell infiltration in TNBC, suggesting potential synergistic effects when combining UCH-L1 inhibitors with PD-L1 antibodies. Overall, in TNBC, UCH-L1 stabilizes ARIH1, leading to low PD-L1 expression, which may explain the limited effectiveness of immunotherapy in TNBC patients. Our mouse experiments showed improved therapeutic effects when combining UCH-L1 inhibitors with PD-L1 antibodies. These findings offer a new avenue for immunotherapy in TNBC patients.
Collapse
Affiliation(s)
- Linhao He
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, 410011, China; Hunan Provincial Engineering Research Centre of Translational Medicine and Innovative Drug, Changsha, 410011, China
| | - Jiaying He
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, 410011, China
| | - Ting Jiang
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, 410011, China; Hunan Provincial Engineering Research Centre of Translational Medicine and Innovative Drug, Changsha, 410011, China
| | - Rong Gong
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, 410011, China; Hunan Provincial Engineering Research Centre of Translational Medicine and Innovative Drug, Changsha, 410011, China
| | - Xiaoya Wan
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, 410011, China; Hunan Provincial Engineering Research Centre of Translational Medicine and Innovative Drug, Changsha, 410011, China
| | - Mingwu Duan
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, 410011, China; Hunan Provincial Engineering Research Centre of Translational Medicine and Innovative Drug, Changsha, 410011, China
| | - Zonglin Chen
- Clinical Research Center for Breast Disease in Hunan Province, Changsha, 410011, China
| | - Yan Cheng
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, 410011, China; Hunan Provincial Engineering Research Centre of Translational Medicine and Innovative Drug, Changsha, 410011, China; Clinical Research Center for Breast Disease in Hunan Province, Changsha, 410011, China; NHC Key Laboratory of Cancer Proteomics & State Local Joint Engineering Laboratory for Anticancer Drugs, Xiangya Hospital, Central South University, Changsha, 410008, China; Key Laboratory of Diabetes Immunology (Central South University), Ministry of Education, Changsha, 410011, China.
| |
Collapse
|
2
|
Mei Y, Shen P, Yang Z, Fu S, Shi Y, Cui X, Wang J, Zhang S, Zhou C, Wang X. Ubiquitin Receptor RPN13-Mediated " Candidatus Liberibacter asiaticus" Virulence Effector Degradation to Positively Regulate Immunity. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2025; 73:8809-8821. [PMID: 40192613 DOI: 10.1021/acs.jafc.4c12314] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/11/2025]
Abstract
Ubiquitin-proteasome is a conserved mechanism that regulates cellular responses and disease resistance in plants. However, the regulation role of ubiquitin-proteasome in the pathogenicity of "Candidatus Liberibacter asiaticus" (CLas), the causal agent of citrus Huanglongbing, one of the most serious citrus diseases, remains poorly defined. In this study, we identified a CLas effector, SDE5640 (CLIBASIA_05640), which downregulates salicylic acid signaling pathway genes and partial 26S proteasome genes in SDE5640-transgenic citrus shoots. SDE5640 suppresses proteasome activity to promote bacterial infection. Intriguingly, RPN13, a known component of the proteasome, was first identified as a new ubiquitin receptor. Overexpression of NbRPN13 leads to the degradation of SDE5640 via the 26S proteasome system. We further revealed that SDE5640 can be ubiquitinated in planta and that NbRPN13 promotes the degradation of SDE5640 by binding to ubiquitinated SDE5640. Furthermore, transient coexpression of SDE5640 and CsRPN13 enhances citrus resistance against Xanthomonas. Together, these results demonstrate that RPN13 recognizes the ubiquitination site of SDE5640 and specifically degrades it using the ubiquitin-proteasome system, which provides evidence for the role of ubiquitin-proteasome in CLas-citrus interactions.
Collapse
Affiliation(s)
- Yalin Mei
- Integrative Science Center of Germplasm Creation in Western China (CHONGQING) Science City, Citrus Research Institute, Southwest University, Chongqing 400712, China
- National Citrus Engineering Research Center, Citrus Research Institute, Southwest University, Chongqing 400712, China
| | - Pan Shen
- Integrative Science Center of Germplasm Creation in Western China (CHONGQING) Science City, Citrus Research Institute, Southwest University, Chongqing 400712, China
- College of Bioengineering, Jingchu University of Technology, Jingmen, 448000 Hubei, China
| | - Zaiyu Yang
- Integrative Science Center of Germplasm Creation in Western China (CHONGQING) Science City, Citrus Research Institute, Southwest University, Chongqing 400712, China
- National Citrus Engineering Research Center, Citrus Research Institute, Southwest University, Chongqing 400712, China
| | - Shimin Fu
- Integrative Science Center of Germplasm Creation in Western China (CHONGQING) Science City, Citrus Research Institute, Southwest University, Chongqing 400712, China
- National Citrus Engineering Research Center, Citrus Research Institute, Southwest University, Chongqing 400712, China
| | - Yaqian Shi
- Integrative Science Center of Germplasm Creation in Western China (CHONGQING) Science City, Citrus Research Institute, Southwest University, Chongqing 400712, China
- National Citrus Engineering Research Center, Citrus Research Institute, Southwest University, Chongqing 400712, China
| | - Xuejin Cui
- National Citrus Engineering Research Center, Citrus Research Institute, Southwest University, Chongqing 400712, China
| | - Jiajun Wang
- Integrative Science Center of Germplasm Creation in Western China (CHONGQING) Science City, Citrus Research Institute, Southwest University, Chongqing 400712, China
- National Citrus Engineering Research Center, Citrus Research Institute, Southwest University, Chongqing 400712, China
| | - Shushe Zhang
- Integrative Science Center of Germplasm Creation in Western China (CHONGQING) Science City, Citrus Research Institute, Southwest University, Chongqing 400712, China
- National Citrus Engineering Research Center, Citrus Research Institute, Southwest University, Chongqing 400712, China
| | - Changyong Zhou
- Integrative Science Center of Germplasm Creation in Western China (CHONGQING) Science City, Citrus Research Institute, Southwest University, Chongqing 400712, China
- National Citrus Engineering Research Center, Citrus Research Institute, Southwest University, Chongqing 400712, China
| | - Xuefeng Wang
- Integrative Science Center of Germplasm Creation in Western China (CHONGQING) Science City, Citrus Research Institute, Southwest University, Chongqing 400712, China
- National Citrus Engineering Research Center, Citrus Research Institute, Southwest University, Chongqing 400712, China
| |
Collapse
|
3
|
Wang Y, Wang Y, Iriki T, Hashimoto E, Inami M, Hashimoto S, Watanabe A, Takano H, Motosugi R, Hirayama S, Sugishita H, Gotoh Y, Yao R, Hamazaki J, Murata S. The DYT6 dystonia causative protein THAP1 is responsible for proteasome activity via PSMB5 transcriptional regulation. Nat Commun 2025; 16:1600. [PMID: 39952963 PMCID: PMC11828994 DOI: 10.1038/s41467-025-56867-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Accepted: 02/04/2025] [Indexed: 02/17/2025] Open
Abstract
The proteasome plays a pivotal role in protein degradation, and its impairment is associated with various pathological conditions, including neurodegenerative diseases. It is well understood that Nrf1 coordinates the induction of all proteasome genes in response to proteasome dysfunction. However, the molecular mechanism regulating the basal expression of the proteasome remains unclear. Here we identify the transcription factor THAP1, the causative gene of DYT6 dystonia, as a regulator of proteasome activity through a genome-wide genetic screen. We demonstrated that THAP1 directly regulates the expression of the PSMB5 gene, which encodes the central protease subunit β5. Depletion of THAP1 disrupts proteasome assembly, leading to reduced proteasome activity and the accumulation of ubiquitinated proteins. These findings uncover a regulatory mechanism for the proteasome and suggest a potential role for proteasome dysfunction in the pathogenesis of dystonia.
Collapse
Affiliation(s)
- Yan Wang
- Laboratory of Protein Metabolism, Graduate School of Pharmaceutical Sciences, the University of Tokyo, Bunkyo-ku, Tokyo, Japan
- The Affiliated Kangning Hospital of Ningbo University, No. 1 Zhuangyunan Road, Ningbo, China
| | - Yi Wang
- Laboratory of Protein Metabolism, Graduate School of Pharmaceutical Sciences, the University of Tokyo, Bunkyo-ku, Tokyo, Japan
| | - Tomohiro Iriki
- Laboratory of Protein Metabolism, Graduate School of Pharmaceutical Sciences, the University of Tokyo, Bunkyo-ku, Tokyo, Japan
| | - Eiichi Hashimoto
- Laboratory of Protein Metabolism, Graduate School of Pharmaceutical Sciences, the University of Tokyo, Bunkyo-ku, Tokyo, Japan
| | - Maki Inami
- Laboratory of Protein Metabolism, Graduate School of Pharmaceutical Sciences, the University of Tokyo, Bunkyo-ku, Tokyo, Japan
| | - Sota Hashimoto
- Laboratory of Protein Metabolism, Graduate School of Pharmaceutical Sciences, the University of Tokyo, Bunkyo-ku, Tokyo, Japan
| | - Ayako Watanabe
- One-stop Sharing Facility Center for Future Drug Discoveries, Graduate School of Pharmaceutical Sciences, the University of Tokyo, Bunkyo-ku, Tokyo, Japan
| | - Hiroshi Takano
- Department of Cell Biology, Cancer Institute, Japanese Foundation for Cancer Research, Koto-ku, Tokyo, Japan
| | - Ryo Motosugi
- Laboratory of Protein Metabolism, Graduate School of Pharmaceutical Sciences, the University of Tokyo, Bunkyo-ku, Tokyo, Japan
| | - Shoshiro Hirayama
- Laboratory of Protein Metabolism, Graduate School of Pharmaceutical Sciences, the University of Tokyo, Bunkyo-ku, Tokyo, Japan
| | - Hiroki Sugishita
- Laboratory of Molecular Biology, Graduate School of Pharmaceutical Sciences, the University of Tokyo, Bunkyo-ku, Tokyo, Japan
- International Research Center for Neurointelligence (WPI-IRCN), the University of Tokyo, Bunkyo-ku, Tokyo, Japan
| | - Yukiko Gotoh
- Laboratory of Molecular Biology, Graduate School of Pharmaceutical Sciences, the University of Tokyo, Bunkyo-ku, Tokyo, Japan
- International Research Center for Neurointelligence (WPI-IRCN), the University of Tokyo, Bunkyo-ku, Tokyo, Japan
| | - Ryoji Yao
- Department of Cell Biology, Cancer Institute, Japanese Foundation for Cancer Research, Koto-ku, Tokyo, Japan
| | - Jun Hamazaki
- Laboratory of Protein Metabolism, Graduate School of Pharmaceutical Sciences, the University of Tokyo, Bunkyo-ku, Tokyo, Japan
| | - Shigeo Murata
- Laboratory of Protein Metabolism, Graduate School of Pharmaceutical Sciences, the University of Tokyo, Bunkyo-ku, Tokyo, Japan.
| |
Collapse
|
4
|
Bao Y, Cruz G, Zhang Y, Qiao Y, Mannan R, Hu J, Yang F, Gondal M, Shahine M, Kang S, Mahapatra S, Chu A, Choi JE, Yu J, Lin H, Miner SJ, Robinson DR, Wu YM, Zheng Y, Cao X, Su F, Wang R, Hosseini N, Cieslik M, Kryczek I, Vaishampayan U, Zou W, Chinnaiyan AM. The UBA1-STUB1 Axis Mediates Cancer Immune Escape and Resistance to Checkpoint Blockade. Cancer Discov 2025; 15:363-381. [PMID: 39540840 PMCID: PMC11803397 DOI: 10.1158/2159-8290.cd-24-0435] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Revised: 09/12/2024] [Accepted: 10/15/2024] [Indexed: 11/16/2024]
Abstract
SIGNIFICANCE Our study reveals UBA1 as a predictive biomarker for clinical outcomes in ICB cohorts, mediating cancer immune evasion and ICB resistance. We further highlight JAK1 stabilization as a key mechanism of UBA1 inhibition and nominate the UBA1-STUB1 axis as an immuno-oncology therapeutic target to improve the efficacy of ICB.
Collapse
Affiliation(s)
- Yi Bao
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, Michigan
- Department of Pathology, University of Michigan, Ann Arbor, Michigan
| | - Gabriel Cruz
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, Michigan
- Department of Pathology, University of Michigan, Ann Arbor, Michigan
| | - Yuping Zhang
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, Michigan
- Department of Pathology, University of Michigan, Ann Arbor, Michigan
| | - Yuanyuan Qiao
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, Michigan
- Department of Pathology, University of Michigan, Ann Arbor, Michigan
- Rogel Cancer Center, University of Michigan, Ann Arbor, Michigan
| | - Rahul Mannan
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, Michigan
- Department of Pathology, University of Michigan, Ann Arbor, Michigan
| | - Jing Hu
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, Michigan
- Department of Pathology, University of Michigan, Ann Arbor, Michigan
- Department of Pathology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Fan Yang
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, Michigan
- Department of Pathology, University of Michigan, Ann Arbor, Michigan
| | - Mahnoor Gondal
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, Michigan
- Department of Pathology, University of Michigan, Ann Arbor, Michigan
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, Michigan
| | - Miriam Shahine
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, Michigan
| | - Sarah Kang
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, Michigan
| | - Somnath Mahapatra
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, Michigan
- Department of Pathology, University of Michigan, Ann Arbor, Michigan
| | - Alec Chu
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, Michigan
- Department of Pathology, University of Michigan, Ann Arbor, Michigan
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, Michigan
| | - Jae Eun Choi
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, Michigan
- Department of Pathology, University of Michigan, Ann Arbor, Michigan
| | - Jiali Yu
- Department of Surgery, University of Michigan, Ann Arbor, Michigan
- Center of Excellence for Cancer Immunology and Immunotherapy, University of Michigan, Ann Arbor, Michigan
| | - Heng Lin
- Department of Surgery, University of Michigan, Ann Arbor, Michigan
- Center of Excellence for Cancer Immunology and Immunotherapy, University of Michigan, Ann Arbor, Michigan
| | - Stephanie J. Miner
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, Michigan
- Department of Pathology, University of Michigan, Ann Arbor, Michigan
| | - Dan R. Robinson
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, Michigan
- Department of Pathology, University of Michigan, Ann Arbor, Michigan
| | - Yi-Mi Wu
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, Michigan
- Department of Pathology, University of Michigan, Ann Arbor, Michigan
| | - Yang Zheng
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, Michigan
- Department of Pathology, University of Michigan, Ann Arbor, Michigan
| | - Xuhong Cao
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, Michigan
- Department of Pathology, University of Michigan, Ann Arbor, Michigan
| | - Fengyun Su
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, Michigan
- Department of Pathology, University of Michigan, Ann Arbor, Michigan
| | - Rui Wang
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, Michigan
- Department of Pathology, University of Michigan, Ann Arbor, Michigan
| | - Noshad Hosseini
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, Michigan
- Department of Pathology, University of Michigan, Ann Arbor, Michigan
| | - Marcin Cieslik
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, Michigan
- Department of Pathology, University of Michigan, Ann Arbor, Michigan
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, Michigan
| | - Ilona Kryczek
- Department of Surgery, University of Michigan, Ann Arbor, Michigan
- Center of Excellence for Cancer Immunology and Immunotherapy, University of Michigan, Ann Arbor, Michigan
| | - Ulka Vaishampayan
- Rogel Cancer Center, University of Michigan, Ann Arbor, Michigan
- Division of Hematology and Oncology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan
| | - Weiping Zou
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, Michigan
- Department of Pathology, University of Michigan, Ann Arbor, Michigan
- Rogel Cancer Center, University of Michigan, Ann Arbor, Michigan
- Department of Surgery, University of Michigan, Ann Arbor, Michigan
- Center of Excellence for Cancer Immunology and Immunotherapy, University of Michigan, Ann Arbor, Michigan
| | - Arul M. Chinnaiyan
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, Michigan
- Department of Pathology, University of Michigan, Ann Arbor, Michigan
- Rogel Cancer Center, University of Michigan, Ann Arbor, Michigan
- Howard Hughes Medical Institute, University of Michigan, Ann Arbor, Michigan
- Department of Urology, University of Michigan, Ann Arbor, Michigan
| |
Collapse
|
5
|
Wang J, Kjellgren A, DeMartino GN. PI31 is a positive regulator of 20S immunoproteasome assembly. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.15.633194. [PMID: 39868238 PMCID: PMC11761684 DOI: 10.1101/2025.01.15.633194] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 01/28/2025]
Abstract
PI31 (Proteasome Inhibitor of 31,000 Da) is a 20S proteasome-binding protein originally identified as an inhibitor of in vitro 20S proteasome activity. Although recent studies have provided a detailed structural basis for this activity, the physiologic significance of PI31-mediated proteasome inhibition remains uncertain and alternative cellular roles for PI31 have been described. Here we report a role for PI31 as a positive regulator for the assembly of the 20S immuno-proteasome (20Si), a compositionally and functionally distinct isoform of the proteasome that is poorly inhibited by PI31. Genetic ablation of PI31 in mammalian cells had no effect on the cellular content or activity of constitutively expressed proteasomes but reduced the cellular content and activity of interferon-γ-induced immuno-proteasomes. This selective effect is a consequence of defective 20Si assembly, as indicated by the accumulation of 20Si assembly intermediates. Our results highlight a distinction in the assembly pathways of constitutive and immuno-proteasomes and indicate that PI31 plays a chaperone-like role for the selective assembly of 20S immunoproteasomes.
Collapse
Affiliation(s)
| | - Abbey Kjellgren
- Department of Physiology, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX 75390-9040
| | - George N. DeMartino
- Department of Physiology, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX 75390-9040
| |
Collapse
|
6
|
Zheng M, Chen X, Xu Z, Ye Z, Zhong L, Chen Z, Chen G, Cai B. Comprehensive analysis of PSMG3 in pan-cancer and validation of its role in hepatocellular carcinoma. Clin Transl Oncol 2025; 27:319-332. [PMID: 38967739 DOI: 10.1007/s12094-024-03580-8] [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: 05/04/2024] [Accepted: 06/19/2024] [Indexed: 07/06/2024]
Abstract
BACKGROUND Proteasome assembly chaperone 3 (PSMG3), a subunit of proteasome, has been found to be associated with lung cancer. However, the role of PSMG3 in other cancers has not been elucidated. The objective of this study was to explore the immune role of PSMG3 in pan-cancer and confirm the oncogenic significance in liver hepatocellular carcinoma (LIHC). METHODS We examined the differential expression of PSMG3 across various cancer types using data from The Cancer Genome Atlas (TCGA) and the Genotype-Tissue Expression (GTEx) databases. We investigated the prognostic value of PSMG3 and examined its relationship with tumor mutation burden (TMB), microsatellite instability (MSI), and immune infiltration. The functional enrichment analysis was performed to explore the potential molecular mechanism of PSMG3. To elucidate the biological function of PSMG3, we conducted in vitro experiments using liver cancer cell lines. RESULTS PSMG3 was highly expressed in most cancers. The high PSMG3 expression value of PSMG3 was closely related to poor prognosis. We observed correlations between PSMG3 and TMB, and MSI immune infiltration. PSMG3 may be involved in metabolic reprogramming, cell cycle, and PPAR pathways. The over-expression of PSMG3 promoted the proliferation, migration, and invasion capabilities of liver cancer cells. CONCLUSION Our study demonstrated that PSMG3 was a pivotal oncogene in multiple cancers. PSMG3 contributed to the progression and immune infiltration in pan-cancer, especially in LIHC.
Collapse
Affiliation(s)
- Mengli Zheng
- Department of Gastroenterology, The Tenth Affiliated Hospital, Southern Medical University (Dongguan People's Hospital), Dongguan, China
| | - Xiaochun Chen
- Department of Gastroenterology, The Tenth Affiliated Hospital, Southern Medical University (Dongguan People's Hospital), Dongguan, China
| | - Zhe Xu
- Department of Gastroenterology, The Tenth Affiliated Hospital, Southern Medical University (Dongguan People's Hospital), Dongguan, China
| | - Zhitao Ye
- Department of Nephrology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Lixian Zhong
- Department of Gastroenterology, The Tenth Affiliated Hospital, Southern Medical University (Dongguan People's Hospital), Dongguan, China
| | - Zhicao Chen
- Department of Gastroenterology, The Tenth Affiliated Hospital, Southern Medical University (Dongguan People's Hospital), Dongguan, China
| | - Guiquan Chen
- Department of Gastroenterology, The Tenth Affiliated Hospital, Southern Medical University (Dongguan People's Hospital), Dongguan, China.
| | - Boyong Cai
- Department of Gastroenterology, The Tenth Affiliated Hospital, Southern Medical University (Dongguan People's Hospital), Dongguan, China.
| |
Collapse
|
7
|
Luo D, Li X, Wei L, Yu Y, Hazaisihan Y, Tao L, Li S, Jia W. Ubiquitin-related gene markers predict immunotherapy response and prognosis in patients with epithelial ovarian carcinoma. Sci Rep 2024; 14:25239. [PMID: 39448713 PMCID: PMC11502900 DOI: 10.1038/s41598-024-76945-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2024] [Accepted: 10/17/2024] [Indexed: 10/26/2024] Open
Abstract
Epithelial ovarian carcinoma (EOC) is the most fatal among female reproductive system tumors. The immune tumor microenvironment and ubiquitin-proteasome pathway are closely related to the proliferation, invasion, and response to chemotherapy in EOC. However, their specific roles in EOC have not been fully elucidated. Therefore, we aimed to recognize potential prognostic markers and novel therapeutic targets for EOC. We constructed the ubiquitin-related signature risk model comprising HSP90AB1, FBXO9, SIGMAR1, STAT1, SH3KBP1, EPB41L2, DNAJB6, VPS18, PPM1G, AKAP12, FRK, and PYGB, specifically for patients with EOC. The high-risk model presented a worse prognosis, primarily associated with the B-cell receptor signaling pathway, ECM receptor interaction, focal adhesion, and actin cytoskeleton regulations. Analysis of the immune landscape revealed a higher abundance of B cells, M2 macrophages, neutrophil CD4 T cells, cancer-associated fibroblasts, macrophage neutrophils, and fibroblasts in the high-risk group. It also exhibited lower tumor mutation burden, mRNAsi, and EREG-mRNAsi and reduced sensitivity to other chemotherapy drugs, except dasatinib. These findings serve as a valuable indicator for personalized treatment strategies and clinical stratification in managing patients with EOC. Additionally, our study will serve as a foundation for future mechanistic research to explore the association between the ubiquitin-proteasome pathway and EOC.
Collapse
Affiliation(s)
- Donglin Luo
- First Affiliated Hospital, Department of Pathology, Key Laboratory for Xinjiang Endemic and Ethnic Diseases, Shihezi University, Shihezi University School of Medicine, Shihezi, China
| | - Xiaoning Li
- First Affiliated Hospital, Department of Pathology, Key Laboratory for Xinjiang Endemic and Ethnic Diseases, Shihezi University, Shihezi University School of Medicine, Shihezi, China
| | - Li Wei
- First Affiliated Hospital, Department of Pathology, Key Laboratory for Xinjiang Endemic and Ethnic Diseases, Shihezi University, Shihezi University School of Medicine, Shihezi, China
| | - Yankun Yu
- First Affiliated Hospital, Department of Pathology, Key Laboratory for Xinjiang Endemic and Ethnic Diseases, Shihezi University, Shihezi University School of Medicine, Shihezi, China
| | - Yeernaer Hazaisihan
- First Affiliated Hospital, Department of Pathology, Key Laboratory for Xinjiang Endemic and Ethnic Diseases, Shihezi University, Shihezi University School of Medicine, Shihezi, China
| | - Lin Tao
- First Affiliated Hospital, Department of Pathology, Key Laboratory for Xinjiang Endemic and Ethnic Diseases, Shihezi University, Shihezi University School of Medicine, Shihezi, China
| | - Siyuan Li
- First Affiliated Hospital, Department of Pathology, Key Laboratory for Xinjiang Endemic and Ethnic Diseases, Shihezi University, Shihezi University School of Medicine, Shihezi, China.
| | - Wei Jia
- First Affiliated Hospital, Department of Pathology, Key Laboratory for Xinjiang Endemic and Ethnic Diseases, Shihezi University, Shihezi University School of Medicine, Shihezi, China.
| |
Collapse
|
8
|
Engineering modular enzymes using DNA origami. NATURE NANOTECHNOLOGY 2024; 19:1440-1441. [PMID: 39117908 DOI: 10.1038/s41565-024-01739-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/10/2024]
|
9
|
Yan G, Zhu T, Zhou J, Li X, Wen Z, Miuhuitijiang B, Zhang Z, Du Y, Li C, Shi X, Tan W. GOLM1 promotes prostate cancer progression via interaction with PSMD1 and enhancing AR-driven transcriptional activation. J Cell Mol Med 2024; 28:e70186. [PMID: 39470578 PMCID: PMC11520440 DOI: 10.1111/jcmm.70186] [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: 05/20/2024] [Revised: 10/20/2024] [Accepted: 10/22/2024] [Indexed: 10/30/2024] Open
Abstract
Aberrant transcriptional activation of the androgen receptor (AR) is a predominant cause of prostate cancer (PCa), including both in the initial and androgen-independent stages. Our study highlights Golgi membrane protein 1 (GOLM1) as a key regulator of AR-driven transcriptional activity in PCa progression. Utilizing local clinical data and TCGA data, we have established a robust association between GOLM1 and AR target genes, and further demonstrated that GOLM1 can enhance the expression of AR target genes. We discovered that GOLM1 interacts with PSMD1, a component of the 19S regulatory complex in the 26S proteasome, using mass spectrometry and Co-IP analysis. It is well known that ubiquitin-proteasome plays a vital role in AR expression and transcriptional regulation. Our findings demonstrate that GOLM1 enhances ubiquitin proteasome activity by binding to PSMD1, thereby facilitating AR-driven transcriptional activity and PCa progression. These results indicate that GOLM1 and its associated proteins may become potential therapeutic targets for PCa characterized by dysregulated AR-driven transcriptional activation.
Collapse
Affiliation(s)
- Guang Yan
- Department of Urology, Nanfang HospitalSouthern Medical UniversityGuangzhouChina
- Department of Andrology, Shanghai Seventh People's HospitalShanghai University of Traditional Chinese MedicineShanghaiChina
| | - Tianhang Zhu
- Department of Urology, Nanfang HospitalSouthern Medical UniversityGuangzhouChina
| | - Jiawei Zhou
- Department of Urology, Nanfang HospitalSouthern Medical UniversityGuangzhouChina
| | - Xia Li
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Department of Biochemistry and Molecular BiologyThe Fourth Military Medical UniversityXi'anShaanxiChina
| | - Zonghua Wen
- Department of PathologyShenzhen University General HospitalShenzhenChina
| | | | - Zhiyong Zhang
- Department of Urology, Nanfang HospitalSouthern Medical UniversityGuangzhouChina
| | - Yuejun Du
- Department of Urology, Nanfang HospitalSouthern Medical UniversityGuangzhouChina
| | - Chengyao Li
- Department of Transfusion Medicine, School of Laboratory Medicine and BiotechnologySouthern Medical UniversityGuangzhouChina
| | - Xiaojun Shi
- Department of Urology, Nanfang HospitalSouthern Medical UniversityGuangzhouChina
| | - Wanlong Tan
- Department of Urology, Nanfang HospitalSouthern Medical UniversityGuangzhouChina
| |
Collapse
|
10
|
Huang J, Jaekel A, van den Boom J, Podlesainski D, Elnaggar M, Heuer-Jungemann A, Kaiser M, Meyer H, Saccà B. A modular DNA origami nanocompartment for engineering a cell-free, protein unfolding and degradation pathway. NATURE NANOTECHNOLOGY 2024; 19:1521-1531. [PMID: 39075293 PMCID: PMC11486656 DOI: 10.1038/s41565-024-01738-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Accepted: 05/28/2024] [Indexed: 07/31/2024]
Abstract
Within the cell, chemical reactions are often confined and organized through a modular architecture. This facilitates the targeted localization of molecular species and their efficient translocation to subsequent sites. Here we present a cell-free nanoscale model that exploits compartmentalization strategies to carry out regulated protein unfolding and degradation. Our synthetic model comprises two connected DNA origami nanocompartments (each measuring 25 nm × 41 nm × 53 nm): one containing the protein unfolding machine, p97, and the other housing the protease chymotrypsin. We achieve the unidirectional immobilization of p97 within the first compartment, establishing a gateway mechanism that controls substrate recruitment, translocation and processing within the second compartment. Our data show that, whereas spatial confinement increases the rate of the individual reactions by up to tenfold, the physical connection of the compartmentalized enzymes into a chimera efficiently couples the two reactions and reduces off-target proteolysis by almost sixfold. Hence, our modular approach may serve as a blueprint for engineering artificial nanofactories with reshaped catalytic performance and functionalities beyond those observed in natural systems.
Collapse
Affiliation(s)
- J Huang
- Bionanotechnology, CENIDE and ZMB, University of Duisburg-Essen, Essen, Germany
| | - A Jaekel
- Bionanotechnology, CENIDE and ZMB, University of Duisburg-Essen, Essen, Germany
| | - J van den Boom
- Molecular Biology, ZMB, University of Duisburg-Essen, Essen, Germany
| | - D Podlesainski
- Chemical Biology, ZMB, University of Duisburg-Essen, Essen, Germany
| | - M Elnaggar
- Max Planck Institute of Biochemistry, Martinsried, Germany
| | | | - M Kaiser
- Chemical Biology, ZMB, University of Duisburg-Essen, Essen, Germany
| | - H Meyer
- Molecular Biology, ZMB, University of Duisburg-Essen, Essen, Germany.
| | - B Saccà
- Bionanotechnology, CENIDE and ZMB, University of Duisburg-Essen, Essen, Germany.
| |
Collapse
|
11
|
Quiroz-Castañeda RE, Aguilar-Díaz H, Coronado-Villanueva E, Catalán-Ochoa DI, Amaro-Estrada I. Molecular Identification and Bioinformatics Analysis of Anaplasma marginale Moonlighting Proteins as Possible Antigenic Targets. Pathogens 2024; 13:845. [PMID: 39452716 PMCID: PMC11510912 DOI: 10.3390/pathogens13100845] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Revised: 09/13/2024] [Accepted: 09/27/2024] [Indexed: 10/26/2024] Open
Abstract
BACKGROUND Diseases of veterinary importance, such as bovine Anaplasmosis, cause significant economic losses. Due to this, the study of various proteins of the causal agent Anaplasma marginale has focused on surface proteins. However, a vaccine for this disease is not yet available. To this end, in this work, moonlighting proteins (MLPs) are presented as an alternative approach for the design of immunogens against A. marginale. METHODS The proteins of the strain MEX-15-099-01 were analyzed, and its MLPs were identified. Subsequently, four virulence-associated MLP genes were selected and identified using PCR. The proteins were analyzed using a structural homology approach and the collection of B-cell epitopes was predicted for each MLP. Finally, a pair of AmEno peptides were synthesized and the antigenic potential was tested using an iELISA. RESULTS Our bioinformatics analysis revealed the potential of AmEno, AmGroEl, AmEF-Tu, and AmDnaK proteins as promising candidates for designing immunogens. The PCR allowed the gene sequence identification in the genome of the strain MEX-15-099-01. Notably, AmEno-derived synthetic peptides showed antigenicity in an ELISA. CONCLUSIONS Our study has shed light on the potential use of MLPs for immunogen design, demonstrating the antigenic potential of AmEno.
Collapse
|
12
|
Sun L, Liu Z, Wu Z, Wu Z, Qiu B, Liu S, Hu J, Yin X. PSMD11 promotes the proliferation of hepatocellular carcinoma by regulating the ubiquitination degradation of CDK4. Cell Signal 2024; 121:111279. [PMID: 38944255 DOI: 10.1016/j.cellsig.2024.111279] [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: 04/01/2024] [Revised: 06/14/2024] [Accepted: 06/26/2024] [Indexed: 07/01/2024]
Abstract
BACKGROUND The 26S proteasome non-ATPase regulatory subunit 11 is a multiprotein complex involved in the ATP-dependent degradation of ubiquitinated proteins, and PSMD11 plays a key role in the regulation of embryonic stem cell proteasome activity. However, the role of PSMD11 in hepatocellular carcinoma has not been studied. In this study, it was found that the expression of PSMD11 in HCC tissues was significantly higher than that in para-cancerous tissues, and was associated with poor prognosis. The results of in vitro experiments showed that PSMD11 knockdown could effectively inhibit the proliferation and apoptosis of hepatoma cell lines, and flow cytometry showed that the G0/G1 phase was significantly prolonged. Through protein spectrometry, immunoprecipitation and in vitro experiments, it was found that PSMD11 can promote the proliferation of hepatocellular carcinoma through regulating the ubiquitination of CDK4 and enhancing its protein stability. This study explores the mechanism of action of PSMD11 in hepatocellular carcinoma and provides new insights for the treatment of hepatocellular carcinoma.
Collapse
Affiliation(s)
- Liang Sun
- Department of Hepatobiliary and Pancreatic Surgery, the Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Zitao Liu
- Department of Gastrointestinal Surgery, the Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Zhengyi Wu
- Department of Hepatobiliary and Pancreatic Surgery, the Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Zhipeng Wu
- Department of Hepatobiliary and Pancreatic Surgery, the Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Bingbing Qiu
- Department of Hepatobiliary and Pancreatic Surgery, the Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Shuiqiu Liu
- Department of Hepatobiliary and Pancreatic Surgery, the Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Junwen Hu
- Department of Hepatobiliary and Pancreatic Surgery, the Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China.
| | - Xiangbao Yin
- Department of Hepatobiliary and Pancreatic Surgery, the Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China.
| |
Collapse
|
13
|
Matias AC, Viegas AR, Couto A, Lourenço-Marques C, Aragão C, Castanho S, Gamboa M, Candeias-Mendes A, Soares F, Modesto T, Pousão-Ferreira P, Ribeiro L. Effect of dietary l-glutamine supplementation on the intestinal physiology and growth during Solea senegalensis larval development. Comp Biochem Physiol B Biochem Mol Biol 2024; 272:110961. [PMID: 38387740 DOI: 10.1016/j.cbpb.2024.110961] [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: 09/20/2023] [Revised: 02/19/2024] [Accepted: 02/19/2024] [Indexed: 02/24/2024]
Abstract
The maturation of the intestinal digestive and absorptive functions might limit the amount of absorbed nutrients to fulfil the high requirements of the fast-growing marine fish larva. Glutamine (Gln) has been described to improve intestinal epithelium functions, due to its involvement in energy metabolism and protein synthesis. The purpose of this study was to evaluate dietary 0.2% Gln supplementation on aspects of intestinal physiology, protein metabolism and growth-related genes expression in Senegalese sole larvae. Experiment was carried out between 12 and 33 days post hatching (DPH) and fish were divided into two experimental groups, one fed Artemia spp. (CTRL) and the other fed Artemia spp. supplemented with Gln (GLN). GLN diet had two times more Gln than the CTRL diet. Samples were collected at 15, 19, 26 and 33 DPH for biometry, histology, and digestive enzymes activity, and at 33 DPH for gene expression, protein metabolism and AA content determination. Growth was significantly higher for Senegalese sole fed GLN diet, supported by differences on protein metabolism and growth-related gene expression. Slight differences were observed between treatments regarding the intestinal physiology. Overall, GLN diet seems to be directed to enhance protein metabolism leading to higher larval growth.
Collapse
Affiliation(s)
- Ana Catarina Matias
- IPMA - Portuguese Institute for the Sea and Atmosphere, EPPO - Aquaculture Research Station, Av. Parque Natural da Ria Formosa, s/n, 8700-194 Olhão, Portugal.
| | - Ana Rita Viegas
- IPMA - Portuguese Institute for the Sea and Atmosphere, EPPO - Aquaculture Research Station, Av. Parque Natural da Ria Formosa, s/n, 8700-194 Olhão, Portugal
| | - Ana Couto
- CIIMAR - Interdisciplinary Centre of Marine and Environmental Research, University of Porto, Terminal de Cruzeiros do Porto de Leixões, Av. General Norton de Matos s/n, 4450-208 Matosinhos, Portugal
| | - Cátia Lourenço-Marques
- IPMA - Portuguese Institute for the Sea and Atmosphere, EPPO - Aquaculture Research Station, Av. Parque Natural da Ria Formosa, s/n, 8700-194 Olhão, Portugal
| | - Cláudia Aragão
- CCMAR - Centre of Marine Sciences, University of Algarve, Campus de Gambelas, 8005-139 Faro, Portugal
| | - Sara Castanho
- IPMA - Portuguese Institute for the Sea and Atmosphere, EPPO - Aquaculture Research Station, Av. Parque Natural da Ria Formosa, s/n, 8700-194 Olhão, Portugal
| | - Margarida Gamboa
- IPMA - Portuguese Institute for the Sea and Atmosphere, EPPO - Aquaculture Research Station, Av. Parque Natural da Ria Formosa, s/n, 8700-194 Olhão, Portugal
| | - Ana Candeias-Mendes
- IPMA - Portuguese Institute for the Sea and Atmosphere, EPPO - Aquaculture Research Station, Av. Parque Natural da Ria Formosa, s/n, 8700-194 Olhão, Portugal
| | - Florbela Soares
- IPMA - Portuguese Institute for the Sea and Atmosphere, EPPO - Aquaculture Research Station, Av. Parque Natural da Ria Formosa, s/n, 8700-194 Olhão, Portugal
| | - Teresa Modesto
- CCMAR - Centre of Marine Sciences, University of Algarve, Campus de Gambelas, 8005-139 Faro, Portugal
| | - Pedro Pousão-Ferreira
- IPMA - Portuguese Institute for the Sea and Atmosphere, EPPO - Aquaculture Research Station, Av. Parque Natural da Ria Formosa, s/n, 8700-194 Olhão, Portugal
| | - Laura Ribeiro
- IPMA - Portuguese Institute for the Sea and Atmosphere, EPPO - Aquaculture Research Station, Av. Parque Natural da Ria Formosa, s/n, 8700-194 Olhão, Portugal
| |
Collapse
|
14
|
Ott C. Mapping the interplay of immunoproteasome and autophagy in different heart failure phenotypes. Free Radic Biol Med 2024; 218:149-165. [PMID: 38570171 DOI: 10.1016/j.freeradbiomed.2024.03.026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Revised: 03/25/2024] [Accepted: 03/30/2024] [Indexed: 04/05/2024]
Abstract
Proper protein degradation is required for cellular protein homeostasis and organ function. Particularly, in post-mitotic cells, such as cardiomyocytes, unbalanced proteolysis due to inflammatory stimuli and oxidative stress contributes to organ dysfunction. To ensure appropriate protein turnover, eukaryotic cells exert two main degradation systems, the ubiquitin-proteasome-system and the autophagy-lysosome-pathway. It has been shown that proteasome activity affects the development of cardiac dysfunction differently, depending on the type of heart failure. Studies analyzing the inducible subtype of the proteasome, the immunoproteasome (i20S), demonstrated that the i20S plays a double role in diseased hearts. While i20S subunits are increased in cardiac hypertrophy, atrial fibrillation and partly in myocarditis, the opposite applies to diabetic cardiomyopathy and ischemia/reperfusion injury. In addition, the i20S appears to play a role in autophagy modulation depending on heart failure phenotype. This review summarizes the current literature on the i20S in different heart failure phenotypes, emphasizing the two faces of i20S in injured hearts. A selection of established i20S inhibitors is introduced and signaling pathways linking the i20S to autophagy are highlighted. Mapping the interplay of the i20S and autophagy in different types of heart failure offers potential approaches for developing treatment strategies against heart failure.
Collapse
Affiliation(s)
- Christiane Ott
- German Institute of Human Nutrition Potsdam-Rehbruecke, Department of Molecular Toxicology, Arthur-Scheunert-Allee 114-116, 14558, Nuthetal, Germany; DZHK (German Center for Cardiovascular Research), Partner Site Berlin, Berlin, Germany.
| |
Collapse
|
15
|
Wang J, Kjellgren A, DeMartino GN. Differential Interactions of the Proteasome Inhibitor PI31 with Constitutive and Immuno-20S Proteasomes. Biochemistry 2024; 63:1000-1015. [PMID: 38577872 DOI: 10.1021/acs.biochem.3c00707] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/06/2024]
Abstract
PI31 (Proteasome Inhibitor of 31,000 Da) is a 20S proteasome binding protein originally identified as an in vitro inhibitor of 20S proteasome proteolytic activity. Recently reported cryo-electron microscopy structures of 20S-PI31 complexes have revealed that the natively disordered proline-rich C-terminus of PI31 enters the central chamber in the interior of the 20S proteasome and interacts directly with the proteasome's multiple catalytic threonine residues in a manner predicted to inhibit their enzymatic function while evading its own proteolysis. Higher eukaryotes express an alternative form of the 20S proteasome (termed "immuno-proteasome") that features genetically and functionally distinct catalytic subunits. The effect of PI31 on immuno-proteasome function is unknown. We examine the relative inhibitory effects of PI31 on purified constitutive (20Sc) and immuno-(20Si) 20S proteasomes in vitro and show that PI31 inhibits 20Si hydrolytic activity to a significantly lesser degree than that of 20Sc. Unlike 20Sc, 20Si hydrolyzes the carboxyl-terminus of PI31 and this effect contributes to the reduced inhibitory activity of PI31 toward 20Si. Conversely, loss of 20Sc inhibition by PI31 point mutants leads to PI31 degradation by 20Sc. These results demonstrate unexpected differential interactions of PI31 with 20Sc and 20Si and document their functional consequences.
Collapse
Affiliation(s)
- Jason Wang
- Department of Physiology, UT Southwestern Medical Center, 5323 Harry Hines Blvd, Dallas, Texas 75390-9040, United States
| | - Abbey Kjellgren
- Department of Physiology, UT Southwestern Medical Center, 5323 Harry Hines Blvd, Dallas, Texas 75390-9040, United States
| | - George N DeMartino
- Department of Physiology, UT Southwestern Medical Center, 5323 Harry Hines Blvd, Dallas, Texas 75390-9040, United States
| |
Collapse
|
16
|
Bozhüyük KAJ, Präve L, Kegler C, Schenk L, Kaiser S, Schelhas C, Shi YN, Kuttenlochner W, Schreiber M, Kandler J, Alanjary M, Mohiuddin TM, Groll M, Hochberg GKA, Bode HB. Evolution-inspired engineering of nonribosomal peptide synthetases. Science 2024; 383:eadg4320. [PMID: 38513038 DOI: 10.1126/science.adg4320] [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: 12/23/2022] [Accepted: 02/09/2024] [Indexed: 03/23/2024]
Abstract
Many clinically used drugs are derived from or inspired by bacterial natural products that often are produced through nonribosomal peptide synthetases (NRPSs), megasynthetases that activate and join individual amino acids in an assembly line fashion. In this work, we describe a detailed phylogenetic analysis of several bacterial NRPSs that led to the identification of yet undescribed recombination sites within the thiolation (T) domain that can be used for NRPS engineering. We then developed an evolution-inspired "eXchange Unit between T domains" (XUT) approach, which allows the assembly of NRPS fragments over a broad range of GC contents, protein similarities, and extender unit specificities, as demonstrated for the specific production of a proteasome inhibitor designed and assembled from five different NRPS fragments.
Collapse
Affiliation(s)
- Kenan A J Bozhüyük
- Max Planck Institute for Terrestrial Microbiology, Department of Natural Products in Organismic Interactions, 35043 Marburg, Germany
- Molecular Biotechnology, Department of Biosciences, Goethe-University Frankfurt, 60438 Frankfurt, Germany
- Myria Biosciences AG, Tech Park Basel, Hochbergstrasse 60C, 4057 Basel, Switzerland
| | - Leonard Präve
- Max Planck Institute for Terrestrial Microbiology, Department of Natural Products in Organismic Interactions, 35043 Marburg, Germany
- Molecular Biotechnology, Department of Biosciences, Goethe-University Frankfurt, 60438 Frankfurt, Germany
| | - Carsten Kegler
- Max Planck Institute for Terrestrial Microbiology, Department of Natural Products in Organismic Interactions, 35043 Marburg, Germany
- Molecular Biotechnology, Department of Biosciences, Goethe-University Frankfurt, 60438 Frankfurt, Germany
| | - Leonie Schenk
- Max Planck Institute for Terrestrial Microbiology, Department of Natural Products in Organismic Interactions, 35043 Marburg, Germany
- Molecular Biotechnology, Department of Biosciences, Goethe-University Frankfurt, 60438 Frankfurt, Germany
| | - Sebastian Kaiser
- Max Planck Institute for Terrestrial Microbiology, Department of Natural Products in Organismic Interactions, 35043 Marburg, Germany
- Evolutionary Biochemistry Group, Max Planck Institute for Terrestrial Microbiology, 35043 Marburg, Germany
| | - Christian Schelhas
- Max Planck Institute for Terrestrial Microbiology, Department of Natural Products in Organismic Interactions, 35043 Marburg, Germany
| | - Yan-Ni Shi
- Molecular Biotechnology, Department of Biosciences, Goethe-University Frankfurt, 60438 Frankfurt, Germany
| | - Wolfgang Kuttenlochner
- Chair of Biochemistry, Center for Protein Assemblies, Technical University of Munich, Ernst-Otto-Fischer-Straße 8, 85748 Garching, Germany
| | - Max Schreiber
- Max Planck Institute for Terrestrial Microbiology, Department of Natural Products in Organismic Interactions, 35043 Marburg, Germany
- Molecular Biotechnology, Department of Biosciences, Goethe-University Frankfurt, 60438 Frankfurt, Germany
| | - Joshua Kandler
- Molecular Biotechnology, Department of Biosciences, Goethe-University Frankfurt, 60438 Frankfurt, Germany
| | - Mohammad Alanjary
- Bioinformatics Group, Wageningen University, Droevendaalsesteeg 1, 6708PB Wageningen, The Netherlands
| | - T M Mohiuddin
- Molecular Biotechnology, Department of Biosciences, Goethe-University Frankfurt, 60438 Frankfurt, Germany
| | - Michael Groll
- Chair of Biochemistry, Center for Protein Assemblies, Technical University of Munich, Ernst-Otto-Fischer-Straße 8, 85748 Garching, Germany
| | - Georg K A Hochberg
- Evolutionary Biochemistry Group, Max Planck Institute for Terrestrial Microbiology, 35043 Marburg, Germany
- Center for Synthetic Microbiology (SYNMIKRO), Phillips University Marburg, 35043 Marburg, Germany
- Department of Chemistry, Phillips University Marburg, 35043 Marburg, Germany
| | - Helge B Bode
- Max Planck Institute for Terrestrial Microbiology, Department of Natural Products in Organismic Interactions, 35043 Marburg, Germany
- Molecular Biotechnology, Department of Biosciences, Goethe-University Frankfurt, 60438 Frankfurt, Germany
- Center for Synthetic Microbiology (SYNMIKRO), Phillips University Marburg, 35043 Marburg, Germany
- Department of Chemistry, Phillips University Marburg, 35043 Marburg, Germany
- LOEWE Centre for Translational Biodiversity Genomics (LOEWE-TBG) & Senckenberg Gesellschaft für Naturforschung, 60325 Frankfurt, Germany
| |
Collapse
|
17
|
Sahoo MP, Lavy T, Cohen N, Sahu I, Kleifeld O. Activity-Guided Proteomic Profiling of Proteasomes Uncovers a Variety of Active (and Inactive) Proteasome Species. Mol Cell Proteomics 2024; 23:100728. [PMID: 38296025 PMCID: PMC10907802 DOI: 10.1016/j.mcpro.2024.100728] [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: 09/02/2023] [Revised: 01/11/2024] [Accepted: 01/24/2024] [Indexed: 02/29/2024] Open
Abstract
Proteasomes are multisubunit, multicatalytic protein complexes present in eukaryotic cells that degrade misfolded, damaged, or unstructured proteins. In this study, we used an activity-guided proteomic methodology based on a fluorogenic peptide substrate to characterize the composition of proteasome complexes in WT yeast and the changes these complexes undergo upon the deletion of Pre9 (Δα3) or of Sem1 (ΔSem1). A comparison of whole-cell proteomic analysis to activity-guided proteasome profiling indicates that the amounts of proteasomal proteins and proteasome interacting proteins in the assembled active proteasomes differ significantly from their total amounts in the cell as a whole. Using this activity-guided profiling approach, we characterized the changes in the abundance of subunits of various active proteasome species in different strains, quantified the relative abundance of active proteasomes across these strains, and charted the overall distribution of different proteasome species within each strain. The distributions obtained by our mass spectrometry-based quantification were markedly higher for some proteasome species than those obtained by activity-based quantification alone, suggesting that the activity of some of these species is impaired. The impaired activity appeared mostly among 20SBlm10 proteasome species which account for 20% of the active proteasomes in WT. To identify the factors behind this impaired activity, we mapped and quantified known proteasome-interacting proteins. Our results suggested that some of the reduced activity might be due to the association of the proteasome inhibitor Fub1. Additionally, we provide novel evidence for the presence of nonmature and therefore inactive proteasomal protease subunits β2 and β5 in the fully assembled proteasomes.
Collapse
Affiliation(s)
| | - Tali Lavy
- Faculty of Biology, Technion-Israel Institute of Technology, Technion City, Haifa, Israel
| | - Noam Cohen
- Faculty of Biology, Technion-Israel Institute of Technology, Technion City, Haifa, Israel
| | - Indrajit Sahu
- Faculty of Biology, Technion-Israel Institute of Technology, Technion City, Haifa, Israel
| | - Oded Kleifeld
- Faculty of Biology, Technion-Israel Institute of Technology, Technion City, Haifa, Israel.
| |
Collapse
|
18
|
Liu J, Shen J, Zong J, Fan Y, Cui J, Peng D, Jin Y. Lithium Chloride Promotes Endogenous Synthesis of CLA in Bovine Mammary Epithelial Cells. Biol Trace Elem Res 2024; 202:513-526. [PMID: 37099221 DOI: 10.1007/s12011-023-03679-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Accepted: 04/18/2023] [Indexed: 04/27/2023]
Abstract
Although conjugated linoleic acid (CLA) can promote human health, its content in milk is insufficient to have a significant impact. The majority of the CLA in milk is produced endogenously by the mammary gland. However, research on improving its content through nutrient-induced endogenous synthesis is relatively scarce. Previous research found that the key enzyme, stearoyl-CoA desaturase (SCD) for the synthesis of CLA, can be expressed more actively in bovine mammary epithelial cells (MAC-T) when lithium chloride (LiCl) is present. This study investigated whether LiCl can encourage CLA synthesis in MAC-T cells. The results showed that LiCl effectively increased SCD and proteasome α5 subunit (PSMA5) protein expression in MAC-T cells as well as the content of CLA and its endogenous synthesis index. LiCl enhanced the expression of proliferator-activated receptor-γ (PPARγ), sterol regulatory element-binding protein 1 (SREBP1), and its downstream enzymes acetyl CoA carboxylase (ACC), fatty acid synthase (FASN), lipoprotein lipase (LPL), and Perilipin 2 (PLIN2). The addition of LiCl significantly enhanced p-GSK-3β, β-catenin, p-β-catenin protein expression, hypoxia-inducible factor-1α (HIF-1α), and downregulation factor genes for mRNA expression (P < 0.05). These findings highlight that LiCl can increase the expression of SCD and PSMA5 by activating the transcription of HIF-1α, Wnt/β-catenin, and the SREBP1 signaling pathways to promote the conversion of trans-vaccenic acid (TVA) to the endogenous synthesis of CLA. This data suggests that the exogenous addition of nutrients can increase CLA content in milk through pertinent signaling pathways.
Collapse
Affiliation(s)
- Jiayi Liu
- Jilin Provincial Key Laboratory of Livestock and Poultry Feed and Feeding in the Northeastern Frigid Area, College of Animal Sciences, Jilin University, Changchun, 130062, China
| | - Jinglin Shen
- Jilin Provincial Key Laboratory of Livestock and Poultry Feed and Feeding in the Northeastern Frigid Area, College of Animal Sciences, Jilin University, Changchun, 130062, China
| | - Jinxin Zong
- Jilin Provincial Key Laboratory of Livestock and Poultry Feed and Feeding in the Northeastern Frigid Area, College of Animal Sciences, Jilin University, Changchun, 130062, China
| | - Yating Fan
- Jilin Provincial Key Laboratory of Livestock and Poultry Feed and Feeding in the Northeastern Frigid Area, College of Animal Sciences, Jilin University, Changchun, 130062, China
| | - Junhao Cui
- Jilin Provincial Key Laboratory of Livestock and Poultry Feed and Feeding in the Northeastern Frigid Area, College of Animal Sciences, Jilin University, Changchun, 130062, China
| | - Dongqiao Peng
- Jilin Provincial Key Laboratory of Livestock and Poultry Feed and Feeding in the Northeastern Frigid Area, College of Animal Sciences, Jilin University, Changchun, 130062, China
| | - Yongcheng Jin
- Jilin Provincial Key Laboratory of Livestock and Poultry Feed and Feeding in the Northeastern Frigid Area, College of Animal Sciences, Jilin University, Changchun, 130062, China.
| |
Collapse
|
19
|
Liu Q, Maqbool A, Mirkin FG, Singh Y, Stevenson CEM, Lawson DM, Kamoun S, Huang W, Hogenhout SA. Bimodular architecture of bacterial effector SAP05 that drives ubiquitin-independent targeted protein degradation. Proc Natl Acad Sci U S A 2023; 120:e2310664120. [PMID: 38039272 DOI: 10.1073/pnas.2310664120] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Accepted: 10/19/2023] [Indexed: 12/03/2023] Open
Abstract
In eukaryotes, targeted protein degradation (TPD) typically depends on a series of interactions among ubiquitin ligases that transfer ubiquitin molecules to substrates leading to degradation by the 26S proteasome. We previously identified that the bacterial effector protein SAP05 mediates ubiquitin-independent TPD. SAP05 forms a ternary complex via interactions with the von Willebrand Factor Type A (vWA) domain of the proteasomal ubiquitin receptor Rpn10 and the zinc-finger (ZnF) domains of the SQUAMOSA-PROMOTER BINDING PROTEIN-LIKE (SPL) and GATA BINDING FACTOR (GATA) transcription factors (TFs). This leads to direct TPD of the TFs by the 26S proteasome. Here, we report the crystal structures of the SAP05-Rpn10vWA complex at 2.17 Å resolution and of the SAP05-SPL5ZnF complex at 2.20 Å resolution. Structural analyses revealed that SAP05 displays a remarkable bimodular architecture with two distinct nonoverlapping surfaces, a "loop surface" with three protruding loops that form electrostatic interactions with ZnF, and a "sheet surface" featuring two β-sheets, loops, and α-helices that establish polar interactions with vWA. SAP05 binding to ZnF TFs involves single amino acids responsible for multiple contacts, while SAP05 binding to vWA is more stable due to the necessity of multiple mutations to break the interaction. In addition, positioning of the SAP05 complex on the 26S proteasome points to a mechanism of protein degradation. Collectively, our findings demonstrate how a small bacterial bimodular protein can bypass the canonical ubiquitin-proteasome proteolysis pathway, enabling ubiquitin-independent TPD in eukaryotic cells. This knowledge holds significant potential for the creation of TPD technologies.
Collapse
Affiliation(s)
- Qun Liu
- Department of Crop Genetics, John Innes Centre, Norwich Research Park, Norwich NR4 7UH, United Kingdom
| | - Abbas Maqbool
- Department of Biochemistry and Metabolism, John Innes Centre, Norwich Research Park, Norwich NR4 7UH, United Kingdom
- The Sainsbury Laboratory, University of East Anglia, Norwich NR4 7UH, United Kingdom
| | - Federico G Mirkin
- Department of Crop Genetics, John Innes Centre, Norwich Research Park, Norwich NR4 7UH, United Kingdom
| | - Yeshveer Singh
- Department of Crop Genetics, John Innes Centre, Norwich Research Park, Norwich NR4 7UH, United Kingdom
| | - Clare E M Stevenson
- Department of Biochemistry and Metabolism, John Innes Centre, Norwich Research Park, Norwich NR4 7UH, United Kingdom
| | - David M Lawson
- Department of Biochemistry and Metabolism, John Innes Centre, Norwich Research Park, Norwich NR4 7UH, United Kingdom
| | - Sophien Kamoun
- The Sainsbury Laboratory, University of East Anglia, Norwich NR4 7UH, United Kingdom
| | - Weijie Huang
- National Key Laboratory of Plant Molecular Genetics, Shanghai Centre for Plant Stress Biology, Centre for Excellence in Molecular Plant Sciences, Chinese Academy of Sciences, Shanghai 20032, China
| | - Saskia A Hogenhout
- Department of Crop Genetics, John Innes Centre, Norwich Research Park, Norwich NR4 7UH, United Kingdom
| |
Collapse
|
20
|
Lee H, Kim S, Lee D. The versatility of the proteasome in gene expression and silencing: Unraveling proteolytic and non-proteolytic functions. BIOCHIMICA ET BIOPHYSICA ACTA. GENE REGULATORY MECHANISMS 2023; 1866:194978. [PMID: 37633648 DOI: 10.1016/j.bbagrm.2023.194978] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Revised: 08/02/2023] [Accepted: 08/21/2023] [Indexed: 08/28/2023]
Abstract
The 26S proteasome consists of a 20S core particle and a 19S regulatory particle and critically regulates gene expression and silencing through both proteolytic and non-proteolytic functions. The 20S core particle mediates proteolysis, while the 19S regulatory particle performs non-proteolytic functions. The proteasome plays a role in regulating gene expression in euchromatin by modifying histones, activating transcription, initiating and terminating transcription, mRNA export, and maintaining transcriptome integrity. In gene silencing, the proteasome modulates the heterochromatin formation, spreading, and subtelomere silencing by degrading specific proteins and interacting with anti-silencing factors such as Epe1, Mst2, and Leo1. This review discusses the proteolytic and non-proteolytic functions of the proteasome in regulating gene expression and gene silencing-related heterochromatin formation. This article is part of a special issue on the regulation of gene expression and genome integrity by the ubiquitin-proteasome system.
Collapse
Affiliation(s)
- Hyesu Lee
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology, Daejeon 34141, South Korea
| | - Sungwook Kim
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology, Daejeon 34141, South Korea
| | - Daeyoup Lee
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology, Daejeon 34141, South Korea.
| |
Collapse
|
21
|
Sagathia V, Patel C, Beladiya J, Patel S, Sheth D, Shah G. Tankyrase: a promising therapeutic target with pleiotropic action. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2023; 396:3363-3374. [PMID: 37338576 DOI: 10.1007/s00210-023-02576-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Accepted: 06/13/2023] [Indexed: 06/21/2023]
Abstract
Tankyrase 1 (TNKS1) and tankyrase 2 (TNKS2) enzymes belong to the poly (ADP-ribose) polymerase (PARP) family participates in process of poly-ADP-ribosylation of different target proteins which leads to ubiquitin-mediated proteasomal degradation. Tankyrases are also involved in the pathophysiology of many diseases, especially cancer. Their functions include cell cycle homeostasis (primarily in mitosis), telomere maintenance, Wnt signaling pathway regulation, and insulin signaling (particularly GLUT4 translocation). Studies have implicated that genetic changes, mutations in the tankyrase coding sequence, or up regulation and down regulation of tankyrase are reflected in the numerous disease conditions. Investigations are pursued to develop putative molecules that target tankyrase in various diseases such as cancer, obesity, osteoarthritis, fibrosis, cherubism, and diabetes, thereby providing a new therapeutic treatment option. In the present review, we described the structure and function of tankyrase along with its role in different disease conditions. Furthermore, we also presented cumulative experimental evidences of different drugs acting on tankyrase.
Collapse
Affiliation(s)
- Vrunda Sagathia
- Department of Pharmacology, L. M. College of Pharmacy, Ahmedabad, 380009, Gujarat, India
| | - Chirag Patel
- Department of Pharmacology, L. M. College of Pharmacy, Ahmedabad, 380009, Gujarat, India.
| | - Jayesh Beladiya
- Department of Pharmacology, L. M. College of Pharmacy, Ahmedabad, 380009, Gujarat, India
| | - Sandip Patel
- Department of Pharmacology, L. M. College of Pharmacy, Ahmedabad, 380009, Gujarat, India
| | - Devang Sheth
- Department of Pharmacology, L. M. College of Pharmacy, Ahmedabad, 380009, Gujarat, India
| | - Gaurang Shah
- Department of Pharmacology, L. M. College of Pharmacy, Ahmedabad, 380009, Gujarat, India
| |
Collapse
|
22
|
Loy CA, Muli CS, Ali EMH, Xie D, Ahmed MH, Beth Post C, Trader DJ. Discovery of a non-covalent ligand for Rpn-13, a therapeutic target for hematological cancers. Bioorg Med Chem Lett 2023; 95:129485. [PMID: 37714498 PMCID: PMC10639113 DOI: 10.1016/j.bmcl.2023.129485] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2023] [Revised: 09/11/2023] [Accepted: 09/12/2023] [Indexed: 09/17/2023]
Abstract
The ubiquitin-proteasome system serves as the major proteolytic degradation pathway in eukaryotic cells. Many inhibitors that covalently bind to the proteasome's active sites have been developed for hematological cancers, but resistance can arise in patients. To overcome limitations of active-site proteasome inhibitors, we and others have focused on developing ligands that target subunits on the 19S regulatory particle (19S RP). One such 19S RP subunit, Rpn-13, is a ubiquitin receptor required for hematological cancers to rapidly degrade proteins to avoid apoptosis. Reported Rpn-13 inhibitors covalently bind to the Rpn-13's Pru domain and have been effective anti-hematological cancer agents. Here, we describe the discovery of TCL-1, a non-covalent binder to the Pru domain. Optimization of TCL-1's carboxylate group to an ester increases its cytotoxicity in hematological cancer cell lines. Altogether, our data provides a new scaffold for future medicinal chemistry optimization to target Rpn-13 therapeutically.
Collapse
Affiliation(s)
- Cody A Loy
- Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, 575 West Stadium Avenue, West Lafayette, IN 47907, United States
| | - Christine S Muli
- Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, 575 West Stadium Avenue, West Lafayette, IN 47907, United States
| | - Eslam M H Ali
- Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, 575 West Stadium Avenue, West Lafayette, IN 47907, United States
| | - Dan Xie
- Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, 575 West Stadium Avenue, West Lafayette, IN 47907, United States
| | | | - Carol Beth Post
- Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, 575 West Stadium Avenue, West Lafayette, IN 47907, United States
| | - Darci J Trader
- Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, 575 West Stadium Avenue, West Lafayette, IN 47907, United States.
| |
Collapse
|
23
|
Yang K, Halima A, Chan TA. Antigen presentation in cancer - mechanisms and clinical implications for immunotherapy. Nat Rev Clin Oncol 2023; 20:604-623. [PMID: 37328642 DOI: 10.1038/s41571-023-00789-4] [Citation(s) in RCA: 89] [Impact Index Per Article: 44.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/25/2023] [Indexed: 06/18/2023]
Abstract
Over the past decade, the emergence of effective immunotherapies has revolutionized the clinical management of many types of cancers. However, long-term durable tumour control is only achieved in a fraction of patients who receive these therapies. Understanding the mechanisms underlying clinical response and resistance to treatment is therefore essential to expanding the level of clinical benefit obtained from immunotherapies. In this Review, we describe the molecular mechanisms of antigen processing and presentation in tumours and their clinical consequences. We examine how various aspects of the antigen-presentation machinery (APM) shape tumour immunity. In particular, we discuss genomic variants in HLA alleles and other APM components, highlighting their influence on the immunopeptidomes of both malignant cells and immune cells. Understanding the APM, how it is regulated and how it changes in tumour cells is crucial for determining which patients will respond to immunotherapy and why some patients develop resistance. We focus on recently discovered molecular and genomic alterations that drive the clinical outcomes of patients receiving immune-checkpoint inhibitors. An improved understanding of how these variables mediate tumour-immune interactions is expected to guide the more precise administration of immunotherapies and reveal potentially promising directions for the development of new immunotherapeutic approaches.
Collapse
Affiliation(s)
- Kailin Yang
- Department of Radiation Oncology, Taussig Cancer Center, Cleveland Clinic, Cleveland, OH, USA
| | - Ahmed Halima
- Department of Radiation Oncology, Taussig Cancer Center, Cleveland Clinic, Cleveland, OH, USA
| | - Timothy A Chan
- Department of Radiation Oncology, Taussig Cancer Center, Cleveland Clinic, Cleveland, OH, USA.
- Center for Immunotherapy and Precision Immuno-Oncology, Cleveland Clinic, Cleveland, OH, USA.
- National Center for Regenerative Medicine, Cleveland, OH, USA.
- Case Comprehensive Cancer Center, Cleveland, OH, USA.
| |
Collapse
|
24
|
Iriki T, Iio H, Yasuda S, Masuta S, Kato M, Kosako H, Hirayama S, Endo A, Ohtake F, Kamiya M, Urano Y, Saeki Y, Hamazaki J, Murata S. Senescent cells form nuclear foci that contain the 26S proteasome. Cell Rep 2023; 42:112880. [PMID: 37541257 DOI: 10.1016/j.celrep.2023.112880] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Revised: 05/26/2023] [Accepted: 07/13/2023] [Indexed: 08/06/2023] Open
Abstract
The proteasome plays a central role in intracellular protein degradation. Age-dependent decline in proteasome activity is associated with cellular senescence and organismal aging; however, the mechanism by which the proteasome plays a role in senescent cells remains elusive. Here, we show that nuclear foci that contain the proteasome and exhibit liquid-like properties are formed in senescent cells. The formation of senescence-associated nuclear proteasome foci (SANPs) is dependent on ubiquitination and RAD23B, similar to previously known nuclear proteasome foci, but also requires proteasome activity. RAD23B knockdown suppresses SANP formation and increases mitochondrial activity, leading to reactive oxygen species production without affecting other senescence traits such as cell-cycle arrest and cell morphology. These findings suggest that SANPs are an important feature of senescent cells and uncover a mechanism by which the proteasome plays a role in senescent cells.
Collapse
Affiliation(s)
- Tomohiro Iriki
- Laboratory of Protein Metabolism, Graduate School of Pharmaceutical Sciences, the University of Tokyo, Bunkyo-ku, Tokyo 1130033, Japan
| | - Hiroaki Iio
- Laboratory of Protein Metabolism, Graduate School of Pharmaceutical Sciences, the University of Tokyo, Bunkyo-ku, Tokyo 1130033, Japan
| | - Shu Yasuda
- Department of Hygienic Chemistry and Medical Research Laboratories, School of Pharmaceutical Sciences, Kitasato University, Minato-ku, Tokyo 1088641, Japan
| | - Shun Masuta
- Laboratory of Protein Metabolism, Graduate School of Pharmaceutical Sciences, the University of Tokyo, Bunkyo-ku, Tokyo 1130033, Japan
| | - Masakazu Kato
- Faculty of Pharmaceutical Sciences, Teikyo Heisei University, Nakano-ku, Tokyo 1648530, Japan
| | - Hidetaka Kosako
- Division of Cell Signaling, Fujii Memorial Institute of Medical Sciences, Tokushima University, Kuramoto-cho, Tokushima 7708503, Japan
| | - Shoshiro Hirayama
- Laboratory of Protein Metabolism, Graduate School of Pharmaceutical Sciences, the University of Tokyo, Bunkyo-ku, Tokyo 1130033, Japan
| | - Akinori Endo
- Laboratory of Protein Metabolism, Tokyo Metropolitan Institute of Medical Science, Setagaya-ku, Tokyo 1568506, Japan
| | - Fumiaki Ohtake
- Institute for Advanced Life Sciences, Hoshi University, Shinagawa-ku, Tokyo 1428501, Japan
| | - Mako Kamiya
- Department of Life Science and Technology, Tokyo Institute of Technology, Midori-ku, Yokohama 2268501, Japan
| | - Yasuteru Urano
- Laboratory of Chemical Biology and Molecular Imaging, Graduate School of Medicine, the University of Tokyo, Bunkyo-ku, Tokyo 1130033, Japan; Laboratory of Chemical Biology and Molecular Imaging, Graduate School of Pharmaceutical Sciences, the University of Tokyo, Bunkyo-ku, Tokyo 1130033, Japan
| | - Yasushi Saeki
- Division of Protein Metabolism, the Institute of Medical Science, the University of Tokyo, Minato-ku, Tokyo 1088639, Japan
| | - Jun Hamazaki
- Laboratory of Protein Metabolism, Graduate School of Pharmaceutical Sciences, the University of Tokyo, Bunkyo-ku, Tokyo 1130033, Japan
| | - Shigeo Murata
- Laboratory of Protein Metabolism, Graduate School of Pharmaceutical Sciences, the University of Tokyo, Bunkyo-ku, Tokyo 1130033, Japan.
| |
Collapse
|
25
|
Lee MH, Ratanachan D, Wang Z, Hack J, Abdulrahman L, Shamlin NP, Kalayjian M, Nesseler JP, Ganapathy E, Nguyen C, Ratikan JA, Cacalano NA, Austin D, Damoiseaux R, DiPardo B, Graham DS, Kalbasi A, Sayer JW, McBride WH, Schaue D. Adaptation of the Tumor Antigen Presentation Machinery to Ionizing Radiation. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2023; 211:693-705. [PMID: 37395687 PMCID: PMC10435044 DOI: 10.4049/jimmunol.2100793] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Accepted: 08/18/2022] [Indexed: 07/04/2023]
Abstract
Ionizing radiation (IR) can reprogram proteasome structure and function in cells and tissues. In this article, we show that IR can promote immunoproteasome synthesis with important implications for Ag processing and presentation and tumor immunity. Irradiation of a murine fibrosarcoma (FSA) induced dose-dependent de novo biosynthesis of the immunoproteasome subunits LMP7, LMP2, and Mecl-1, in concert with other changes in the Ag-presentation machinery (APM) essential for CD8+ T cell-mediated immunity, including enhanced expression of MHC class I (MHC-I), β2-microglobulin, transporters associated with Ag processing molecules, and their key transcriptional activator NOD-like receptor family CARD domain containing 5. In contrast, in another less immunogenic, murine fibrosarcoma (NFSA), LMP7 transcripts and expression of components of the immunoproteasome and the APM were muted after IR, which affected MHC-I expression and CD8+ T lymphocyte infiltration into NFSA tumors in vivo. Introduction of LMP7 into NFSA largely corrected these deficiencies, enhancing MHC-I expression and in vivo tumor immunogenicity. The immune adaptation in response to IR mirrored many aspects of the response to IFN-γ in coordinating the transcriptional MHC-I program, albeit with notable differences. Further investigations showed divergent upstream pathways in that, unlike IFN-γ, IR failed to activate STAT-1 in either FSA or NFSA cells while heavily relying on NF-κB activation. The IR-induced shift toward immunoproteasome production within a tumor indicates that proteasomal reprogramming is part of an integrated and dynamic tumor-host response that is specific to the stressor and the tumor and therefore is of clinical relevance for radiation oncology.
Collapse
Affiliation(s)
- Mi-Heon Lee
- Department of Radiation Oncology, Biostatistics and Radiology at the University of California, Los Angeles (UCLA), Los Angeles, CA, USA
| | - Duang Ratanachan
- Department of Radiation Oncology, Biostatistics and Radiology at the University of California, Los Angeles (UCLA), Los Angeles, CA, USA
| | - Zitian Wang
- Department of Radiation Oncology, Biostatistics and Radiology at the University of California, Los Angeles (UCLA), Los Angeles, CA, USA
| | - Jacob Hack
- Department of Radiation Oncology, Biostatistics and Radiology at the University of California, Los Angeles (UCLA), Los Angeles, CA, USA
| | - Lobna Abdulrahman
- Department of Radiation Oncology, Biostatistics and Radiology at the University of California, Los Angeles (UCLA), Los Angeles, CA, USA
| | - Nicholas P. Shamlin
- Department of Radiation Oncology, Biostatistics and Radiology at the University of California, Los Angeles (UCLA), Los Angeles, CA, USA
| | - Mirna Kalayjian
- Department of Radiation Oncology, Biostatistics and Radiology at the University of California, Los Angeles (UCLA), Los Angeles, CA, USA
| | - Jean Philippe Nesseler
- Department of Radiation Oncology, Biostatistics and Radiology at the University of California, Los Angeles (UCLA), Los Angeles, CA, USA
| | - Ekambaram Ganapathy
- Department of Radiation Oncology, Biostatistics and Radiology at the University of California, Los Angeles (UCLA), Los Angeles, CA, USA
| | - Christine Nguyen
- Department of Radiation Oncology, Biostatistics and Radiology at the University of California, Los Angeles (UCLA), Los Angeles, CA, USA
| | - Josephine A. Ratikan
- Department of Radiation Oncology, Biostatistics and Radiology at the University of California, Los Angeles (UCLA), Los Angeles, CA, USA
| | - Nicolas A. Cacalano
- Department of Radiation Oncology, Biostatistics and Radiology at the University of California, Los Angeles (UCLA), Los Angeles, CA, USA
| | - David Austin
- Department of Molecular and Medical Pharmacology, Biostatistics and Radiology at the University of California, Los Angeles (UCLA), Los Angeles, CA, USA
| | - Robert Damoiseaux
- Department of Molecular and Medical Pharmacology, Biostatistics and Radiology at the University of California, Los Angeles (UCLA), Los Angeles, CA, USA
- Department of Bioengineering, Biostatistics and Radiology at the University of California, Los Angeles (UCLA), Los Angeles, CA, USA
- Department of CNSI, Biostatistics and Radiology at the University of California, Los Angeles (UCLA), Los Angeles, CA, USA
- Department of Jonsson Comprehensive Cancer Center, Biostatistics and Radiology at the University of California, Los Angeles (UCLA), Los Angeles, CA, USA
| | - Benjamin DiPardo
- Department of Surgery, Biostatistics and Radiology at the University of California, Los Angeles (UCLA), Los Angeles, CA, USA
| | - Danielle S. Graham
- Department of Surgery, Biostatistics and Radiology at the University of California, Los Angeles (UCLA), Los Angeles, CA, USA
| | - Anusha Kalbasi
- Department of Radiation Oncology, Biostatistics and Radiology at the University of California, Los Angeles (UCLA), Los Angeles, CA, USA
- Department of Jonsson Comprehensive Cancer Center, Biostatistics and Radiology at the University of California, Los Angeles (UCLA), Los Angeles, CA, USA
- Department of Surgery, Biostatistics and Radiology at the University of California, Los Angeles (UCLA), Los Angeles, CA, USA
| | - James W. Sayer
- Department of Jonsson Comprehensive Cancer Center, Biostatistics and Radiology at the University of California, Los Angeles (UCLA), Los Angeles, CA, USA
- School of Public Health, Biostatistics and Radiology at the University of California, Los Angeles (UCLA), Los Angeles, CA, USA
| | - William H. McBride
- Department of Radiation Oncology, Biostatistics and Radiology at the University of California, Los Angeles (UCLA), Los Angeles, CA, USA
- Department of Jonsson Comprehensive Cancer Center, Biostatistics and Radiology at the University of California, Los Angeles (UCLA), Los Angeles, CA, USA
| | - Dörthe Schaue
- Department of Radiation Oncology, Biostatistics and Radiology at the University of California, Los Angeles (UCLA), Los Angeles, CA, USA
- Department of Jonsson Comprehensive Cancer Center, Biostatistics and Radiology at the University of California, Los Angeles (UCLA), Los Angeles, CA, USA
| |
Collapse
|
26
|
Hsu HC, Wang J, Kjellgren A, Li H, DeMartino GN. Ηigh-resolution structure of mammalian PI31-20S proteasome complex reveals mechanism of proteasome inhibition. J Biol Chem 2023; 299:104862. [PMID: 37236357 PMCID: PMC10319324 DOI: 10.1016/j.jbc.2023.104862] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Revised: 05/08/2023] [Accepted: 05/18/2023] [Indexed: 05/28/2023] Open
Abstract
Proteasome-catalyzed protein degradation mediates and regulates critical aspects of many cellular functions and is an important element of proteostasis in health and disease. Proteasome function is determined in part by the types of proteasome holoenzymes formed between the 20S core particle that catalyzes peptide bond hydrolysis and any of multiple regulatory proteins to which it binds. One of these regulators, PI31, was previously identified as an in vitro 20S proteasome inhibitor, but neither the molecular mechanism nor the possible physiologic significance of PI31-mediated proteasome inhibition has been clear. Here we report a high-resolution cryo-EM structure of the mammalian 20S proteasome in complex with PI31. The structure shows that two copies of the intrinsically disordered carboxyl terminus of PI31 are present in the central cavity of the closed-gate conformation of the proteasome and interact with proteasome catalytic sites in a manner that blocks proteolysis of substrates but resists their own degradation. The two inhibitory polypeptide chains appear to originate from PI31 monomers that enter the catalytic chamber from opposite ends of the 20S cylinder. We present evidence that PI31 can inhibit proteasome activity in mammalian cells and may serve regulatory functions for the control of cellular proteostasis.
Collapse
Affiliation(s)
- Hao-Chi Hsu
- Department of Structural Biology, Van Andel Institute, Grand Rapids, Michigan, USA
| | - Jason Wang
- Department of Physiology, UT Southwestern Medical Center, Dallas, Texas, USA
| | - Abbey Kjellgren
- Department of Physiology, UT Southwestern Medical Center, Dallas, Texas, USA
| | - Huilin Li
- Department of Structural Biology, Van Andel Institute, Grand Rapids, Michigan, USA.
| | - George N DeMartino
- Department of Physiology, UT Southwestern Medical Center, Dallas, Texas, USA.
| |
Collapse
|
27
|
Hsu HC, Wang J, Kjellgren A, Li H, DeMartino GN. High-resolution structure of mammalian PI31â€"20S proteasome complex reveals mechanism of proteasome inhibition. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.04.03.535455. [PMID: 37066326 PMCID: PMC10103979 DOI: 10.1101/2023.04.03.535455] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/03/2023]
Abstract
Proteasome-catalyzed protein degradation mediates and regulates critical aspects of many cellular functions and is an important element of proteostasis in health and disease. Proteasome function is determined in part by the types of proteasome holoenzymes formed between the 20S core particle that catalyzes peptide bond hydrolysis and any of multiple regulatory proteins to which it binds. One of these regulators, PI31, was previously identified as an in vitro 20S proteasome inhibitor, but neither the molecular mechanism nor the possible physiologic significance of PI31-mediated proteasome inhibition has been clear. Here we report a high- resolution cryo-EM structure of the mammalian 20S proteasome in complex with PI31. The structure shows that two copies of the intrinsically-disordered carboxyl-terminus of PI31 are present in the central cavity of the closed-gate conformation of the proteasome and interact with proteasome catalytic sites in a manner that blocks proteolysis of substrates but resists their own degradation. The two inhibitory polypeptide chains appear to originate from PI31 monomers that enter the catalytic chamber from opposite ends of the 20S cylinder. We present evidence that PI31 can inhibit proteasome activity in mammalian cells and may serve regulatory functions for the control of cellular proteostasis.
Collapse
|
28
|
Sato B, Kim J, Morohoshi K, Kang W, Miyado K, Tsuruta F, Kawano N, Chiba T. Proteasome-Associated Proteins, PA200 and ECPAS, Are Essential for Murine Spermatogenesis. Biomolecules 2023; 13:biom13040586. [PMID: 37189334 DOI: 10.3390/biom13040586] [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: 02/22/2023] [Revised: 03/16/2023] [Accepted: 03/22/2023] [Indexed: 05/17/2023] Open
Abstract
Proteasomes are highly sophisticated protease complexes that degrade non-lysosomal proteins, and their proper regulation ensures various biological functions such as spermatogenesis. The proteasome-associated proteins, PA200 and ECPAS, are predicted to function during spermatogenesis; however, male mice lacking each of these genes sustain fertility, raising the possibility that these proteins complement each other. To address this issue, we explored these possible roles during spermatogenesis by producing mice lacking these genes (double-knockout mice; dKO mice). Expression patterns and quantities were similar throughout spermatogenesis in the testes. In epididymal sperm, PA200 and ECPAS were expressed but were differentially localized to the midpiece and acrosome, respectively. Proteasome activity was considerably reduced in both the testes and epididymides of dKO male mice, resulting in infertility. Mass spectrometric analysis revealed LPIN1 as a target protein for PA200 and ECPAS, which was confirmed via immunoblotting and immunostaining. Furthermore, ultrastructural and microscopic analyses demonstrated that the dKO sperm displayed disorganization of the mitochondrial sheath. Our results indicate that PA200 and ECPAS work cooperatively during spermatogenesis and are essential for male fertility.
Collapse
Affiliation(s)
- Ban Sato
- Master's and Doctoral Program in Biology, Faculty of Life and Environmental Sciences, University of Tsukuba, 1-1-1 Tennodai, Tsukuba 305-8577, Japan
- Laboratory of Regulatory Biology, Department of Life Sciences, School of Agriculture, Meiji University, 1-1-1 Higashimita, Kawasaki 214-8571, Japan
| | - Jiwoo Kim
- College of Biological Sciences, School of Life and Environmental Sciences, University of Tsukuba, 1-1-1 Tennodai, Tsukuba 305-8577, Japan
| | - Kazunori Morohoshi
- Laboratory of Regulatory Biology, Department of Life Sciences, School of Agriculture, Meiji University, 1-1-1 Higashimita, Kawasaki 214-8571, Japan
| | - Woojin Kang
- Department of Reproductive Biology, National Research Institute for Child Health and Development, 2-10-1 Okura, Setagaya 157-8535, Japan
| | - Kenji Miyado
- Department of Reproductive Biology, National Research Institute for Child Health and Development, 2-10-1 Okura, Setagaya 157-8535, Japan
| | - Fuminori Tsuruta
- Master's and Doctoral Program in Biology, Faculty of Life and Environmental Sciences, University of Tsukuba, 1-1-1 Tennodai, Tsukuba 305-8577, Japan
| | - Natsuko Kawano
- Laboratory of Regulatory Biology, Department of Life Sciences, School of Agriculture, Meiji University, 1-1-1 Higashimita, Kawasaki 214-8571, Japan
| | - Tomoki Chiba
- Master's and Doctoral Program in Biology, Faculty of Life and Environmental Sciences, University of Tsukuba, 1-1-1 Tennodai, Tsukuba 305-8577, Japan
| |
Collapse
|
29
|
Teixeira VON, Bartikoski BJ, do Espirito Santo RC, Alabarse PVG, Ghannan K, Silva JMS, Filippin LI, Visioli F, Martinez-Gamboa L, Feist E, Xavier RM. The role of proteasome in muscle wasting of experimental arthritis. Adv Rheumatol 2023; 63:14. [PMID: 36949513 DOI: 10.1186/s42358-023-00292-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Accepted: 03/05/2023] [Indexed: 03/24/2023] Open
Abstract
BACKGROUND Rheumatoid arthritis is an autoimmune inflammatory disease that often leads patients to muscle impairment and physical disability. This study aimed to evaluate changes in the activity of proteasome system in skeletal muscles of mice with collagen-induced arthritis (CIA) and treated with etanercept or methotrexate. METHODS Male DBA1/J mice were divided into four groups (n = 8 each): CIA-Vehicle (treated with saline), CIA-ETN (treated with etanercept, 5.5 mg/kg), CIA-MTX (treated with methotrexate, 35 mg/kg) and CO (healthy control group). Mice were treated two times a week for 6 weeks. Clinical score and hind paw edema were measured. Muscles were weighted after euthanasia and used to quantify proteasome activity, gene (MuRF-1, PMSα4, PSMβ5, PMSβ6, PSMβ7, PSMβ8, PSMβ9, and PSMβ10), and protein (PSMβ1, PSMβ5, PSMβ1i, PSMβ5i) expression of proteasome subunits. RESULTS Both treatments slowed disease development, but only CIA-ETN maintained muscle weight compared to CIA-MTX and CIA-Vehicle groups. Etanercept treatment showed caspase-like activity of 26S proteasome similar to CO group, while CIA-Vehicle and CIA-MTX had higher activity compared to CO group (p: 0.0057). MuRF-1 mRNA expression was decreased after etanercept administration compared to CIA-Vehicle and CO groups (p: 0.002, p: 0.007, respectively). PSMβ8 and PSMβ9 mRNA levels were increased in CIA-Vehicle and CIA-MTX compared to CO group, while CIA-ETN presented no difference from CO. PMSβ6 mRNA expression was higher in CIA-Vehicle and CIA-MTX groups than in CO group. Protein levels of the PSMβ5 subunit were increased in CO group compared to CIA-Vehicle; after both etanercept and methotrexate treatments, PSMβ5 expression was higher than in CIA-Vehicle group and did not differ from CO group expression (p: 0.0025, p: 0.001, respectively). The inflammation-induced subunit β1 (LMP2) was enhanced after methotrexate treatment compared to CO group (p: 0.043). CONCLUSIONS The results of CIA-Vehicle show that arthritis increases muscle proteasome activation by enhanced caspase-like activity of 26S proteasome and increased PSMβ8 and PSMβ9 mRNA levels. Etanercept treatment was able to maintain the muscle weight and to modulate proteasome so that its activity and gene expression were compared to CO after TNF inhibition. The protein expression of inflammation-induced proteasome subunit was increased in muscle of CIA-MTX group but not following etanercept treatment. Thus, anti-TNF treatment may be an interesting approach to attenuate the arthritis-related muscle wasting.
Collapse
Affiliation(s)
- Vivian Oliveira Nunes Teixeira
- Medical Sciences Program, Medicine Department, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
- Laboratório de Doenças Autoimunes, Hospital de Clínicas de Porto Alegre, Ramiro Barcelos Street, Santa Cecília, Porto Alegre, 2350, Brazil
| | - Bárbara Jonson Bartikoski
- Medical Sciences Program, Medicine Department, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
- Laboratório de Doenças Autoimunes, Hospital de Clínicas de Porto Alegre, Ramiro Barcelos Street, Santa Cecília, Porto Alegre, 2350, Brazil
| | - Rafaela Cavalheiro do Espirito Santo
- Medical Sciences Program, Medicine Department, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil.
- Laboratório de Doenças Autoimunes, Hospital de Clínicas de Porto Alegre, Ramiro Barcelos Street, Santa Cecília, Porto Alegre, 2350, Brazil.
| | - Paulo Vinícius Gil Alabarse
- Medical Sciences Program, Medicine Department, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
- Laboratório de Doenças Autoimunes, Hospital de Clínicas de Porto Alegre, Ramiro Barcelos Street, Santa Cecília, Porto Alegre, 2350, Brazil
- University of California San Diego Medical Center Library, University of California San Diego School of Medicine, San Diego, USA
| | - Khetam Ghannan
- Schwerpunkt Rheumatologie und Klinische Immunologie, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Jordana Miranda Souza Silva
- Medical Sciences Program, Medicine Department, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
- Laboratório de Doenças Autoimunes, Hospital de Clínicas de Porto Alegre, Ramiro Barcelos Street, Santa Cecília, Porto Alegre, 2350, Brazil
| | - Lidiane Isabel Filippin
- Laboratório de Doenças Autoimunes, Hospital de Clínicas de Porto Alegre, Ramiro Barcelos Street, Santa Cecília, Porto Alegre, 2350, Brazil
- Health and Human Development Department, Universidade La Salle, Canoas, Brazil
| | - Fernanda Visioli
- Patology Department, Faculdade de Odontologia, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Lorena Martinez-Gamboa
- Schwerpunkt Rheumatologie und Klinische Immunologie, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Eugen Feist
- Schwerpunkt Rheumatologie und Klinische Immunologie, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Ricardo Machado Xavier
- Medical Sciences Program, Medicine Department, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
- Laboratório de Doenças Autoimunes, Hospital de Clínicas de Porto Alegre, Ramiro Barcelos Street, Santa Cecília, Porto Alegre, 2350, Brazil
| |
Collapse
|
30
|
Lim SM, Nahm M, Kim SH. Proteostasis and Ribostasis Impairment as Common Cell Death Mechanisms in Neurodegenerative Diseases. J Clin Neurol 2023; 19:101-114. [PMID: 36854331 PMCID: PMC9982182 DOI: 10.3988/jcn.2022.0379] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2022] [Revised: 12/07/2022] [Accepted: 12/07/2022] [Indexed: 03/02/2023] Open
Abstract
The cellular homeostasis of proteins (proteostasis) and RNA metabolism (ribostasis) are essential for maintaining both the structure and function of the brain. However, aging, cellular stress conditions, and genetic contributions cause disturbances in proteostasis and ribostasis that lead to protein misfolding, insoluble aggregate deposition, and abnormal ribonucleoprotein granule dynamics. In addition to neurons being primarily postmitotic, nondividing cells, they are more susceptible to the persistent accumulation of abnormal aggregates. Indeed, defects associated with the failure to maintain proteostasis and ribostasis are common pathogenic components of age-related neurodegenerative diseases, including Alzheimer's disease, Parkinson's disease, and amyotrophic lateral sclerosis. Furthermore, the neuronal deposition of misfolded and aggregated proteins can cause both increased toxicity and impaired physiological function, which lead to neuronal dysfunction and cell death. There is recent evidence that irreversible liquid-liquid phase separation (LLPS) is responsible for the pathogenic aggregate formation of disease-related proteins, including tau, α-synuclein, and RNA-binding proteins, including transactive response DNA-binding protein 43, fused in sarcoma, and heterogeneous nuclear ribonucleoprotein A1. Investigations of LLPS and its control therefore suggest that chaperone/disaggregase, which reverse protein aggregation, are valuable therapeutic targets for effective treatments for neurological diseases. Here we review and discuss recent studies to highlight the importance of understanding the common cell death mechanisms of proteostasis and ribostasis in neurodegenerative diseases.
Collapse
Affiliation(s)
- Su Min Lim
- Cell Therapy Center and Department of Neurology, College of Medicine, Hanyang University, Seoul, Korea
| | - Minyeop Nahm
- Dementia Research Group, Korea Brain Research Institute, Daegu, Korea
| | - Seung Hyun Kim
- Cell Therapy Center and Department of Neurology, College of Medicine, Hanyang University, Seoul, Korea.
| |
Collapse
|
31
|
Dermatologic Manifestations of Noninflammasome-Mediated Autoinflammatory Diseases. JID INNOVATIONS 2023; 3:100176. [PMID: 36876221 PMCID: PMC9982332 DOI: 10.1016/j.xjidi.2022.100176] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Revised: 11/14/2022] [Accepted: 11/21/2022] [Indexed: 12/23/2022] Open
Abstract
Autoinflammatory diseases (AIDs) arise from disturbances that alter interactions of immune cells and tissues. They give rise to prominent (auto)inflammation in the absence of aberrant autoantibodies and/or autoreactive T cells. AIDs that are predominantly caused by changes in the inflammasome pathways, such as the NLRP3- or pyrin-associated inflammasome, have gained substantial attention over the last years. However, AIDs resulting primarily from other changes in the defense system of the innate immune system are less well-studied. These noninflammasome-mediated AIDs relate to, for example, disturbance in the TNF or IFN signaling pathways or aberrations in genes affecting the IL-1RA. The spectrum of clinical signs and symptoms of these conditions is vast. Thus, recognizing early cutaneous signs constitutes an important step in differential diagnoses for dermatologists and other physicians. This review provides an overview of the pathogenesis, clinical presentation, and available treatment options highlighting dermatologic aspects of noninflammasome-mediated AIDs.
Collapse
Key Words
- AID, autoinflammatory disease
- ANCA, antineutrophil cytoplasmic antibody
- AOSD, adult-onset Still disease
- BASDAI, Bath Ankylosing Spondylitis Activity Index
- CANDLE, chronic atypical neutrophilic dermatosis with lipodystrophy and elevated temperature
- CAPS, cryopyrin-associated periodic syndrome
- CRD, cysteine-rich domain
- DIRA, deficiency of IL-1RA
- DITRA, deficiency of IL-36RA
- ER, endoplasmic reticulum
- ESR, erythrocyte sedimentation rate
- FMF, familial Mediterranean fever
- M-CSF, macrophage colony-stimulating factor
- MAS, macrophage activation syndrome
- NET, neutrophil extracellular trap
- NOS, nitrous oxide
- NSAID, nonsteroidal anti-inflammatory drug
- NUD, neutrophilic urticarial dermatosis
- PFAPA, periodic fever, aphthous stomatitis, pharyngitis, and adenitis
- PKR, protein kinase R
- PRAAS, proteosome-associated autoinflammatory disease
- SAPHO, synovitis, acne, pustulosis, hyperostosis, osteitis syndrome
- SAVI, STING-associated vasculopathy with onset in infancy
- STAT, signal transducer and activator of transcription
- SchS, Schnitzler syndrome
- TNFR, TNF receptor
- TRAPS, TNF receptor‒associated autoinflammatory disease
- Th17, T helper 17
- VAS, Visual Analog Scale
- sTNFR, soluble TNF receptor
Collapse
|
32
|
Zhan W, Li D, Saha P, Wang R, Zhang H, Ajay AK, Deban C, Sukenick G, Azzi J, Lin G. Discovery of Highly Selective Inhibitors of the Human Constitutive Proteasome β5c Chymotryptic Subunit. J Med Chem 2023; 66:1172-1185. [PMID: 36608337 PMCID: PMC10157300 DOI: 10.1021/acs.jmedchem.2c00733] [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] [Indexed: 01/07/2023]
Abstract
We describe our discovery and development of potent and highly selective inhibitors of human constitutive proteasome chymotryptic activity (β5c). Structure-activity relationship studies of the novel class of inhibitors focused on optimization of N-cap, C-cap, and side chain of the chemophore asparagine. Compound 32 is the most potent and selective β5c inhibitor in this study. A docking study provides a structure rationale for potency and selectivity. Kinetic studies show a reversible and noncompetitive inhibition mechanism. It enters the cells to engage the proteasome target, potently and selectively kills multiple myeloma cells, and does so by synergizing with a β5i-selective inhibitor.
Collapse
Affiliation(s)
- Wenhu Zhan
- Department of Microbiology & Immunology, Weill Cornell Medicine, 1300 York Ave., New York, NY 10065, United States
| | - Daqiang Li
- Department of Microbiology & Immunology, Weill Cornell Medicine, 1300 York Ave., New York, NY 10065, United States
| | - Priya Saha
- Department of Microbiology & Immunology, Weill Cornell Medicine, 1300 York Ave., New York, NY 10065, United States
| | - Rong Wang
- NMR Analytical Core Facility, Memorial Sloan Kettering Cancer Center, New York, NY 10065, United States
| | - Hao Zhang
- Department of Microbiology & Immunology, Weill Cornell Medicine, 1300 York Ave., New York, NY 10065, United States
| | - Amrendra K. Ajay
- Renal Division, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Christa Deban
- Transplantation Research Center, Renal Division, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - George Sukenick
- NMR Analytical Core Facility, Memorial Sloan Kettering Cancer Center, New York, NY 10065, United States
| | - Jamil Azzi
- Renal Division, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Gang Lin
- Department of Microbiology & Immunology, Weill Cornell Medicine, 1300 York Ave., New York, NY 10065, United States
| |
Collapse
|
33
|
The β-Grasp Domain of Proteasomal ATPase Mpa Makes Critical Contacts with the Mycobacterium tuberculosis 20S Core Particle to Facilitate Degradation. mSphere 2022; 7:e0027422. [PMID: 35993699 PMCID: PMC9599533 DOI: 10.1128/msphere.00274-22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Mycobacterium tuberculosis possesses a Pup-proteasome system analogous to the eukaryotic ubiquitin-proteasome pathway. We have previously shown that the hexameric mycobacterial proteasome ATPase (Mpa) recruits pupylated protein substrates via interactions between amino-terminal coiled-coils in Mpa monomers and the degradation tag Pup. However, it is unclear how Mpa rings interact with a proteasome due to the presence of a carboxyl-terminal β-grasp domain unique to Mpa homologues that makes the interaction highly unstable. Here, we describe newly identified critical interactions between Mpa and 20S core proteasomes. Interestingly, the Mpa C-terminal GQYL motif binds the 20S core particle activation pocket differently than the same motif of the ATP-independent proteasome accessory factor PafE. We further found that the β-hairpin of the Mpa β-grasp domain interacts variably with the H0 helix on top of the 20S core particle via a series of ionic and hydrogen-bond interactions. Individually mutating several involved residues reduced Mpa-mediated protein degradation both in vitro and in vivo. IMPORTANCE The Pup-proteasome system in Mycobacterium tuberculosis is critical for this species to cause lethal infections in mice. Investigating the molecular mechanism of how the Mpa ATPase recruits and unfolds pupylated substrates to the 20S proteasomal core particle for degradation will be essential to fully understand how degradation is regulated, and the structural information we report may be useful for the development of new tuberculosis chemotherapies.
Collapse
|
34
|
Itagi P, Kante A, Lagunes L, Deeds EJ. Understanding the separation of timescales in bacterial proteasome core particle assembly. Biophys J 2022; 121:3975-3986. [PMID: 36016496 PMCID: PMC9674962 DOI: 10.1016/j.bpj.2022.08.022] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Revised: 08/10/2022] [Accepted: 08/19/2022] [Indexed: 11/29/2022] Open
Abstract
The 20S proteasome core particle (CP) is a molecular machine that is a key component of cellular protein degradation pathways. Like other molecular machines, it is not synthesized in an active form but rather as a set of subunits that assemble into a functional complex. The CP is conserved across all domains of life and is composed of 28 subunits, 14 α and 14 β, arranged in four stacked seven-member rings (α7β7β7α7). While details of CP assembly vary across species, the final step in the assembly process is universally conserved: two half proteasomes (HPs; α7β7) dimerize to form the CP. In the bacterium Rhodococcus erythropolis, experiments have shown that the formation of the HP is completed within minutes, while the dimerization process takes hours. The N-terminal propeptide of the β subunit, which is autocatalytically cleaved off after CP formation, plays a key role in regulating this separation of timescales. However, the detailed molecular mechanism of how the propeptide achieves this regulation is unclear. In this work, we used molecular dynamics simulations to characterize HP conformations and found that the HP exists in two states: one where the propeptide interacts with key residues in the HP dimerization interface and likely blocks dimerization, and one where this interface is free. Furthermore, we found that a propeptide mutant that dimerizes extremely slowly is essentially always in the nondimerizable state, while the wild-type rapidly transitions between the two. Based on these simulations, we designed a propeptide mutant that favored the dimerizable state in molecular dynamics simulations. In vitro assembly experiments confirmed that this mutant dimerizes significantly faster than wild-type. Our work thus provides unprecedented insight into how this critical step in CP assembly is regulated, with implications both for efforts to inhibit proteasome assembly and for the evolution of hierarchical assembly pathways.
Collapse
Affiliation(s)
- Pushpa Itagi
- Center for Computational Biology, University of Kansas, Lawrence, Kansas; Institute for Quantitative and Computational Biosciences, UCLA, Los Angeles, California
| | - Anupama Kante
- Department of Molecular Biosciences, University of Kansas, Lawrence, Kansas; Department of Integrative Biology and Physiology, UCLA, Los Angeles, California
| | - Leonila Lagunes
- Department of Integrative Biology and Physiology, UCLA, Los Angeles, California
| | - Eric J Deeds
- Department of Integrative Biology and Physiology, UCLA, Los Angeles, California; Institute for Quantitative and Computational Biosciences, UCLA, Los Angeles, California.
| |
Collapse
|
35
|
Sengupta M, Pluciennik A, Merry DE. The role of ubiquitination in spinal and bulbar muscular atrophy. Front Mol Neurosci 2022; 15:1020143. [PMID: 36277484 PMCID: PMC9583669 DOI: 10.3389/fnmol.2022.1020143] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Accepted: 09/20/2022] [Indexed: 11/13/2022] Open
Abstract
Spinal and bulbar muscular atrophy (SBMA) is a neurodegenerative and neuromuscular genetic disease caused by the expansion of a polyglutamine-encoding CAG tract in the androgen receptor (AR) gene. The AR is an important transcriptional regulator of the nuclear hormone receptor superfamily; its levels are regulated in many ways including by ubiquitin-dependent degradation. Ubiquitination is a post-translational modification (PTM) which plays a key role in both AR transcriptional activity and its degradation. Moreover, the ubiquitin-proteasome system (UPS) is a fundamental component of cellular functioning and has been implicated in diseases of protein misfolding and aggregation, including polyglutamine (polyQ) repeat expansion diseases such as Huntington's disease and SBMA. In this review, we discuss the details of the UPS system, its functions and regulation, and the role of AR ubiquitination and UPS components in SBMA. We also discuss aspects of the UPS that may be manipulated for therapeutic effect in SBMA.
Collapse
Affiliation(s)
| | | | - Diane E. Merry
- Department of Biochemistry and Molecular Biology, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA, United States
| |
Collapse
|
36
|
Zhang W, Roy Burman SS, Chen J, Donovan KA, Cao Y, Shu C, Zhang B, Zeng Z, Gu S, Zhang Y, Li D, Fischer ES, Tokheim C, Shirley Liu X. Machine Learning Modeling of Protein-intrinsic Features Predicts Tractability of Targeted Protein Degradation. GENOMICS, PROTEOMICS & BIOINFORMATICS 2022; 20:882-898. [PMID: 36494034 PMCID: PMC10025769 DOI: 10.1016/j.gpb.2022.11.008] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Revised: 10/25/2022] [Accepted: 11/04/2022] [Indexed: 12/12/2022]
Abstract
Targeted protein degradation (TPD) has rapidly emerged as a therapeutic modality to eliminate previously undruggable proteins by repurposing the cell's endogenous protein degradation machinery. However, the susceptibility of proteins for targeting by TPD approaches, termed "degradability", is largely unknown. Here, we developed a machine learning model, model-free analysis of protein degradability (MAPD), to predict degradability from features intrinsic to protein targets. MAPD shows accurate performance in predicting kinases that are degradable by TPD compounds [with an area under the precision-recall curve (AUPRC) of 0.759 and an area under the receiver operating characteristic curve (AUROC) of 0.775] and is likely generalizable to independent non-kinase proteins. We found five features with statistical significance to achieve optimal prediction, with ubiquitination potential being the most predictive. By structural modeling, we found that E2-accessible ubiquitination sites, but not lysine residues in general, are particularly associated with kinase degradability. Finally, we extended MAPD predictions to the entire proteome to find 964 disease-causing proteins (including proteins encoded by 278 cancer genes) that may be tractable to TPD drug development.
Collapse
Affiliation(s)
- Wubing Zhang
- Department of Data Science, Dana-Farber Cancer Institute, Boston, MA 02215, USA; Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA
| | - Shourya S Roy Burman
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA 02215, USA; Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA 02115, USA
| | - Jiaye Chen
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA 02115, USA
| | - Katherine A Donovan
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA 02215, USA; Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA 02115, USA
| | - Yang Cao
- Center of Growth, Metabolism, and Aging, Key Laboratory of Bio-resource and Eco-environment, Ministry of Education, College of Life Sciences, Sichuan University, Chengdu 610064, China
| | - Chelsea Shu
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA 02215, USA; Research Scholar Initiative, Graduate School of Arts and Sciences, Harvard University, Cambridge, MA 02138, USA
| | - Boning Zhang
- Department of Data Science, Dana-Farber Cancer Institute, Boston, MA 02215, USA; Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA
| | - Zexian Zeng
- Department of Data Science, Dana-Farber Cancer Institute, Boston, MA 02215, USA; Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA
| | - Shengqing Gu
- Department of Data Science, Dana-Farber Cancer Institute, Boston, MA 02215, USA; Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA
| | - Yi Zhang
- Department of Data Science, Dana-Farber Cancer Institute, Boston, MA 02215, USA; Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA
| | - Dian Li
- Department of Data Science, Dana-Farber Cancer Institute, Boston, MA 02215, USA; Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA
| | - Eric S Fischer
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA 02215, USA; Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA 02115, USA.
| | - Collin Tokheim
- Department of Data Science, Dana-Farber Cancer Institute, Boston, MA 02215, USA; Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA.
| | - X Shirley Liu
- Department of Data Science, Dana-Farber Cancer Institute, Boston, MA 02215, USA; Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA.
| |
Collapse
|
37
|
Abdalla OHMH, Mascarenhas B, Cheng HYM. Death of a Protein: The Role of E3 Ubiquitin Ligases in Circadian Rhythms of Mice and Flies. Int J Mol Sci 2022; 23:ijms231810569. [PMID: 36142478 PMCID: PMC9502492 DOI: 10.3390/ijms231810569] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2022] [Revised: 09/07/2022] [Accepted: 09/08/2022] [Indexed: 12/04/2022] Open
Abstract
Circadian clocks evolved to enable organisms to anticipate and prepare for periodic environmental changes driven by the day–night cycle. This internal timekeeping mechanism is built on autoregulatory transcription–translation feedback loops that control the rhythmic expression of core clock genes and their protein products. The levels of clock proteins rise and ebb throughout a 24-h period through their rhythmic synthesis and destruction. In the ubiquitin–proteasome system, the process of polyubiquitination, or the covalent attachment of a ubiquitin chain, marks a protein for degradation by the 26S proteasome. The process is regulated by E3 ubiquitin ligases, which recognize specific substrates for ubiquitination. In this review, we summarize the roles that known E3 ubiquitin ligases play in the circadian clocks of two popular model organisms: mice and fruit flies. We also discuss emerging evidence that implicates the N-degron pathway, an alternative proteolytic system, in the regulation of circadian rhythms. We conclude the review with our perspectives on the potential for the proteolytic and non-proteolytic functions of E3 ubiquitin ligases within the circadian clock system.
Collapse
Affiliation(s)
- Osama Hasan Mustafa Hasan Abdalla
- Department of Biology, University of Toronto Mississauga, Mississauga, ON L5L 1C6, Canada
- Department of Cell & Systems Biology, University of Toronto, Toronto, ON M5S 3G5, Canada
| | - Brittany Mascarenhas
- Department of Biology, University of Toronto Mississauga, Mississauga, ON L5L 1C6, Canada
- Department of Cell & Systems Biology, University of Toronto, Toronto, ON M5S 3G5, Canada
| | - Hai-Ying Mary Cheng
- Department of Biology, University of Toronto Mississauga, Mississauga, ON L5L 1C6, Canada
- Department of Cell & Systems Biology, University of Toronto, Toronto, ON M5S 3G5, Canada
- Correspondence:
| |
Collapse
|
38
|
Fu M, Liu Y, Wang G, Wang P, Zhang J, Chen C, Zhao M, Zhang S, Jiao J, Ouyang X, Yu Y, Wen B, He C, Wang J, Zhou D, Xiong X. A protein–protein interaction map reveals that the Coxiella burnetii effector CirB inhibits host proteasome activity. PLoS Pathog 2022; 18:e1010660. [PMID: 35816513 PMCID: PMC9273094 DOI: 10.1371/journal.ppat.1010660] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Accepted: 06/09/2022] [Indexed: 12/19/2022] Open
Abstract
Coxiella burnetii is the etiological agent of the zoonotic disease Q fever, which is featured by its ability to replicate in acid vacuoles resembling the lysosomal network. One key virulence determinant of C. burnetii is the Dot/Icm system that transfers more than 150 effector proteins into host cells. These effectors function to construct the lysosome-like compartment permissive for bacterial replication, but the functions of most of these effectors remain elusive. In this study, we used an affinity tag purification mass spectrometry (AP-MS) approach to generate a C. burnetii-human protein-protein interaction (PPI) map involving 53 C. burnetii effectors and 3480 host proteins. This PPI map revealed that the C. burnetii effector CBU0425 (designated CirB) interacts with most subunits of the 20S core proteasome. We found that ectopically expressed CirB inhibits hydrolytic activity of the proteasome. In addition, overexpression of CirB in C. burnetii caused dramatic inhibition of proteasome activity in host cells, while knocking down CirB expression alleviated such inhibitory effects. Moreover, we showed that a region of CirB that spans residues 91–120 binds to the proteasome subunit PSMB5 (beta 5). Finally, PSMB5 knockdown promotes C. burnetii virulence, highlighting the importance of proteasome activity modulation during the course of C. burnetii infection. As the causative agent of Q fever, C. burnetii colonizes host cells by transferring effector proteins into the host cytoplasm through its Dot/Icm secretion system to construct a replicative vacuole. The function of effectors remains largely unknown. Here, we performed a large-scale AP-MS screen to analyze the interactions among C. burnetii effectors and human proteins. These analyses found that CirB functions as an inhibitor of host proteasome activity, revealing that proteasome activity is important for intracellular survival of C. burnetii. Our data have laid the foundation for future exploring the molecular mechanisms underlying the roles of C. burnetii effectors in its virulence and for the identification of novel potential drug targets for the development of novel therapeutic treatment for C. burnetii infection.
Collapse
Affiliation(s)
- Mengjiao Fu
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Academy of Military Medicine Sciences, Fengtai, Beijing,China
| | - Yuchen Liu
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences, Beijing Institute of Lifeomics, Beijing, China
| | - Guannan Wang
- Beijing Advanced Innovation Center for Soft Matter Science and Engineering, Beijing University of Chemical Technology, Beijing, China
| | - Peng Wang
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Academy of Military Medicine Sciences, Fengtai, Beijing,China
| | - Jianing Zhang
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Academy of Military Medicine Sciences, Fengtai, Beijing,China
| | - Chen Chen
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, California, United States of America
| | - Mingliang Zhao
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Academy of Military Medicine Sciences, Fengtai, Beijing,China
| | - Shan Zhang
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Academy of Military Medicine Sciences, Fengtai, Beijing,China
| | - Jun Jiao
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Academy of Military Medicine Sciences, Fengtai, Beijing,China
| | - Xuan Ouyang
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Academy of Military Medicine Sciences, Fengtai, Beijing,China
| | - Yonghui Yu
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Academy of Military Medicine Sciences, Fengtai, Beijing,China
| | - Bohai Wen
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Academy of Military Medicine Sciences, Fengtai, Beijing,China
| | - Chengzhi He
- Beijing Advanced Innovation Center for Soft Matter Science and Engineering, Beijing University of Chemical Technology, Beijing, China
| | - Jian Wang
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences, Beijing Institute of Lifeomics, Beijing, China
| | - Dongsheng Zhou
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Academy of Military Medicine Sciences, Fengtai, Beijing,China
- * E-mail: , (DZ); (XX)
| | - Xiaolu Xiong
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Academy of Military Medicine Sciences, Fengtai, Beijing,China
- * E-mail: , (DZ); (XX)
| |
Collapse
|
39
|
Su L, Mosquera J, Mabesoone MFJ, Schoenmakers SMC, Muller C, Vleugels MEJ, Dhiman S, Wijker S, Palmans ARA, Meijer EW. Dilution-induced gel-sol-gel-sol transitions by competitive supramolecular pathways in water. Science 2022; 377:213-218. [DOI: 10.1126/science.abn3438] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Fascinating properties are displayed by synthetic multicomponent supramolecular systems that comprise a manifold of competitive interactions, thereby mimicking natural processes. We present the integration of two reentrant phase transitions based on an unexpected dilution-induced assembly process using supramolecular polymers and surfactants. The co-assembly of the water-soluble benzene-1,3,5-tricarboxamide (BTA-EG
4
) and a surfactant at a specific ratio yielded small-sized aggregates. These interactions were modeled using the competition between self-sorting and co-assembly of both components. The small-sized aggregates were transformed into supramolecular polymer networks by a twofold dilution in water without changing their ratio. Kinetic experiments show the in situ growth of micrometer-long fibers in the dilution process. We were able to create systems that undergo fully reversible hydrogel-solution-hydrogel-solution transitions upon dilution by introducing another orthogonal interaction.
Collapse
Affiliation(s)
- Lu Su
- Institute for Complex Molecular Systems, Laboratory of Macromolecular and Organic Chemistry, Eindhoven University of Technology, 5600 MB Eindhoven, Netherlands
- Leiden Academic Centre for Drug Research, Leiden University, 2333 CC Leiden, Netherlands
| | - Jesús Mosquera
- Institute for Complex Molecular Systems, Laboratory of Macromolecular and Organic Chemistry, Eindhoven University of Technology, 5600 MB Eindhoven, Netherlands
- Centro de Investigacións Científicas Avanzadas, Universidade da Coruña (CICA), A Coruña, Spain
| | - Mathijs F. J. Mabesoone
- Institute for Complex Molecular Systems, Laboratory of Macromolecular and Organic Chemistry, Eindhoven University of Technology, 5600 MB Eindhoven, Netherlands
| | - Sandra M. C. Schoenmakers
- Institute for Complex Molecular Systems, Laboratory of Macromolecular and Organic Chemistry, Eindhoven University of Technology, 5600 MB Eindhoven, Netherlands
| | - Cyprien Muller
- Institute for Complex Molecular Systems, Laboratory of Macromolecular and Organic Chemistry, Eindhoven University of Technology, 5600 MB Eindhoven, Netherlands
| | - Marle E. J. Vleugels
- Institute for Complex Molecular Systems, Laboratory of Macromolecular and Organic Chemistry, Eindhoven University of Technology, 5600 MB Eindhoven, Netherlands
| | - Shikha Dhiman
- Institute for Complex Molecular Systems, Laboratory of Macromolecular and Organic Chemistry, Eindhoven University of Technology, 5600 MB Eindhoven, Netherlands
| | - Stefan Wijker
- Institute for Complex Molecular Systems, Laboratory of Macromolecular and Organic Chemistry, Eindhoven University of Technology, 5600 MB Eindhoven, Netherlands
| | - Anja R. A. Palmans
- Institute for Complex Molecular Systems, Laboratory of Macromolecular and Organic Chemistry, Eindhoven University of Technology, 5600 MB Eindhoven, Netherlands
| | - E. W. Meijer
- Institute for Complex Molecular Systems, Laboratory of Macromolecular and Organic Chemistry, Eindhoven University of Technology, 5600 MB Eindhoven, Netherlands
- School of Chemistry and RNA Institute, University of New South Wales, Sydney, NSW 2052, Australia
| |
Collapse
|
40
|
Enenkel C, Kang RW, Wilfling F, Ernst OP. Intracellular localization of the proteasome in response to stress conditions. J Biol Chem 2022; 298:102083. [PMID: 35636514 PMCID: PMC9218506 DOI: 10.1016/j.jbc.2022.102083] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Revised: 05/13/2022] [Accepted: 05/16/2022] [Indexed: 12/15/2022] Open
Abstract
The ubiquitin–proteasome system fulfills an essential role in regulating protein homeostasis by spatially and temporally controlling proteolysis in an ATP- and ubiquitin-dependent manner. However, the localization of proteasomes is highly variable under diverse cellular conditions. In yeast, newly synthesized proteasomes are primarily localized to the nucleus during cell proliferation. Yeast proteasomes are transported into the nucleus through the nuclear pore either as immature subcomplexes or as mature enzymes via adapter proteins Sts1 and Blm10, while in mammalian cells, postmitotic uptake of proteasomes into the nucleus is mediated by AKIRIN2, an adapter protein essentially required for nuclear protein degradation. Stressful growth conditions and the reversible halt of proliferation, that is quiescence, are associated with a decline in ATP and the reorganization of proteasome localization. Cellular stress leads to proteasome accumulation in membraneless granules either in the nucleus or in the cytoplasm. In quiescence, yeast proteasomes are sequestered in an ubiquitin-dependent manner into motile and reversible proteasome storage granules in the cytoplasm. In cancer cells, upon amino acid deprivation, heat shock, osmotic stress, oxidative stress, or the inhibition of either proteasome activity or nuclear export, reversible proteasome foci containing polyubiquitinated substrates are formed by liquid–liquid phase separation in the nucleus. In this review, we summarize recent literature revealing new links between nuclear transport, ubiquitin signaling, and the intracellular organization of proteasomes during cellular stress conditions.
Collapse
Affiliation(s)
- Cordula Enenkel
- Department of Biochemistry, University of Toronto, Toronto, Ontario, Canada.
| | - Ryu Won Kang
- Department of Biochemistry, University of Toronto, Toronto, Ontario, Canada
| | - Florian Wilfling
- Mechanisms of Cellular Quality Control, Max-Planck-Institute of Biophysics, Frankfurt, Germany
| | - Oliver P Ernst
- Department of Biochemistry, University of Toronto, Toronto, Ontario, Canada; Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
41
|
Ahn MY, Seo DH, Kim WT. PUB22 and PUB23 U-box E3 ubiquitin ligases negatively regulate 26S proteasome activity under proteotoxic stress conditions. JOURNAL OF INTEGRATIVE PLANT BIOLOGY 2022; 64:625-631. [PMID: 34964269 DOI: 10.1111/jipb.13209] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Accepted: 12/25/2021] [Indexed: 06/14/2023]
Abstract
The mechanism regulating proteasomal activity under proteotoxic stress conditions remains unclear. Here, we showed that arsenite-induced proteotoxic stress resulted in upregulation of Arabidopsis homologous PUB22 and PUB23 U-box E3 ubiquitin ligases and that pub22pub23 double mutants displayed arsenite-insensitive seed germination and root growth phenotypes. PUB22/PUB23 downregulated 26S proteasome activity by promoting the dissociation of the 19S regulatory particle from the holo-proteasome complex, resulting in intracellular accumulation of UbG76V -GFP, an artificial substrate of the proteasome complex, and insoluble poly-ubiquitinated proteins. These results suggest that PUB22/PUB23 play a critical role in arsenite-induced proteotoxic stress response via negative regulation of 26S proteasome integrity.
Collapse
Affiliation(s)
- Min Yong Ahn
- Department of Systems Biology, Division of Life Science, Yonsei University, Seoul, 03722, Korea
- Institute of Life Science and Biotechnology, Yonsei University, Seoul, 03722, Korea
| | - Dong Hye Seo
- Department of Systems Biology, Division of Life Science, Yonsei University, Seoul, 03722, Korea
- Institute of Life Science and Biotechnology, Yonsei University, Seoul, 03722, Korea
| | - Woo Taek Kim
- Department of Systems Biology, Division of Life Science, Yonsei University, Seoul, 03722, Korea
- Institute of Life Science and Biotechnology, Yonsei University, Seoul, 03722, Korea
| |
Collapse
|
42
|
The effect of nutrient deprivation on proteasome activity in 4-week-old mice and 24-week-old mice. J Nutr Biochem 2022; 105:108993. [DOI: 10.1016/j.jnutbio.2022.108993] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Revised: 10/20/2021] [Accepted: 02/03/2022] [Indexed: 11/21/2022]
|
43
|
Hasan AM, Jyoti MMS, Rana MR, Rezanujjaman M, Tokumoto T. Purification and Identification of the 20S Proteasome Complex from Zebrafish. Zebrafish 2022; 19:18-23. [PMID: 35171713 DOI: 10.1089/zeb.2021.0064] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
The proteasome is a large polymeric protease complex responsible for degradation of intracellular proteins and generation of peptides. In this study, we purified a native 20S proteasome protein complex from zebrafish (Danio rerio) from the whole body. The cytosolic fraction of zebrafish hydrolyzed Suc-Leu-Leu-Val-Tyr-MCA (Suc-LLVY-MCA), a well-known substrate for the proteasome, in the presence of sodium dodecyl sulfate. From the cytosolic fraction, the 20S proteasome was purified using five column chromatography steps: DEAE cellulose, Q-Sepharose, Sephacryl S-300 gel, hydroxylapatite, and phenyl Sepharose. Electrophoresis and Western blot analyses showed that zebrafish 20S proteasome subunits have molecular masses ranging from 22 to 33 kDa. The subunit composition of the purified 20S proteasome was identified by matrix-assisted laser desorption/ionization-time of flight mass spectrometry (MALDI-TOF/MS) analysis after two-dimensional polyacrylamide gel electrophoresis (2D-PAGE) separation. Fourteen kinds of 20S subunits were found. As a special characteristic of zebrafish, two proteins of the α1 subunit were identified. In addition, the results suggested that the α8 subunit is in the 20S complex instead of the α4 subunit. In this study, we demonstrated the subunit composition of the 20S proteasome complex present in zebrafish cells.
Collapse
Affiliation(s)
- Ali Md Hasan
- Integrated Bioscience Section, Graduate School of Science and Technology, National University Corporation, Shizuoka University, Shizuoka, Japan
| | - Md Maisum Sarwar Jyoti
- Integrated Bioscience Section, Graduate School of Science and Technology, National University Corporation, Shizuoka University, Shizuoka, Japan
| | - Md Rubel Rana
- Integrated Bioscience Section, Graduate School of Science and Technology, National University Corporation, Shizuoka University, Shizuoka, Japan
| | - Md Rezanujjaman
- Integrated Bioscience Section, Graduate School of Science and Technology, National University Corporation, Shizuoka University, Shizuoka, Japan
| | - Toshinobu Tokumoto
- Integrated Bioscience Section, Graduate School of Science and Technology, National University Corporation, Shizuoka University, Shizuoka, Japan
| |
Collapse
|
44
|
Cassioli C, Baldari CT. Lymphocyte Polarization During Immune Synapse Assembly: Centrosomal Actin Joins the Game. Front Immunol 2022; 13:830835. [PMID: 35222415 PMCID: PMC8873515 DOI: 10.3389/fimmu.2022.830835] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Accepted: 01/20/2022] [Indexed: 11/13/2022] Open
Abstract
Interactions among immune cells are essential for the development of adaptive immune responses. The immunological synapse (IS) provides a specialized platform for integration of signals and intercellular communication between T lymphocytes and antigen presenting cells (APCs). In the T cell the reorganization of surface molecules at the synaptic interface is initiated by T cell receptor binding to a cognate peptide-major histocompatibility complex on the APC surface and is accompanied by a polarized remodelling of the cytoskeleton and centrosome reorientation to a subsynaptic position. Although there is a general agreement on polarizing signals and mechanisms driving centrosome reorientation during IS assembly, the primary events that prepare for centrosome repositioning remain largely unexplored. It has been recently shown that in resting lymphocytes a local polymerization of filamentous actin (F-actin) at the centrosome contributes to anchoring this organelle to the nucleus. During early stages of IS formation centrosomal F-actin undergoes depletion, allowing for centrosome detachment from the nucleus and its polarization towards the synaptic membrane. We recently demonstrated that in CD4+ T cells the reduction in centrosomal F-actin relies on the activity of a centrosome-associated proteasome and implicated the ciliopathy-related Bardet-Biedl syndrome 1 protein in the dynein-dependent recruitment of the proteasome 19S regulatory subunit to the centrosome. In this short review we will feature our recent findings that collectively provide a new function for BBS proteins and the proteasome in actin dynamics, centrosome polarization and T cell activation.
Collapse
|
45
|
How do protein aggregates escape quality control in neurodegeneration? Trends Neurosci 2022; 45:257-271. [DOI: 10.1016/j.tins.2022.01.006] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Revised: 01/16/2022] [Accepted: 01/27/2022] [Indexed: 02/07/2023]
|
46
|
Functional Differences between Proteasome Subtypes. Cells 2022; 11:cells11030421. [PMID: 35159231 PMCID: PMC8834425 DOI: 10.3390/cells11030421] [Citation(s) in RCA: 42] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 01/11/2022] [Accepted: 01/14/2022] [Indexed: 12/30/2022] Open
Abstract
Four proteasome subtypes are commonly present in mammalian tissues: standard proteasomes, which contain the standard catalytic subunits β1, β2 and β5; immunoproteasomes containing the immuno-subunits β1i, β2i and β5i; and two intermediate proteasomes, containing a mix of standard and immuno-subunits. Recent studies revealed the expression of two tissue-specific proteasome subtypes in cortical thymic epithelial cells and in testes: thymoproteasomes and spermatoproteasomes. In this review, we describe the mechanisms that enable the ATP- and ubiquitin-dependent as well as the ATP- and ubiquitin-independent degradation of proteins by the proteasome. We focus on understanding the role of the different proteasome subtypes in maintaining protein homeostasis in normal physiological conditions through the ATP- and ubiquitin-dependent degradation of proteins. Additionally, we discuss the role of each proteasome subtype in the ATP- and ubiquitin-independent degradation of disordered proteins. We also discuss the role of the proteasome in the generation of peptides presented by MHC class I molecules and the implication of having different proteasome subtypes for the peptide repertoire presented at the cell surface. Finally, we discuss the role of the immunoproteasome in immune cells and its modulation as a potential therapy for autoimmune diseases.
Collapse
|
47
|
Frayssinhes JYA, Cerruti F, Laulin J, Cattaneo A, Bachi A, Apcher S, Coux O, Cascio P. PA28γ-20S proteasome is a proteolytic complex committed to degrade unfolded proteins. Cell Mol Life Sci 2021; 79:45. [PMID: 34913092 PMCID: PMC11071804 DOI: 10.1007/s00018-021-04045-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2020] [Revised: 11/15/2021] [Accepted: 11/17/2021] [Indexed: 02/07/2023]
Abstract
PA28γ is a nuclear activator of the 20S proteasome that, unlike the 19S regulatory particle, stimulates hydrolysis of several substrates in an ATP- and ubiquitin-independent manner and whose exact biological functions and molecular mechanism of action still remain elusive. In an effort to shed light on these important issues, we investigated the stimulatory effect of PA28γ on the hydrolysis of different fluorogenic peptides and folded or denatured full-length proteins by the 20S proteasome. Importantly, PA28γ was found to dramatically enhance breakdown rates by 20S proteasomes of several naturally or artificially unstructured proteins, but not of their native, folded counterparts. Furthermore, these data were corroborated by experiments in cell lines with a nucleus-tagged myelin basic protein. Finally, mass spectrometry analysis of the products generated during proteasomal degradation of two proteins demonstrated that PA28γ does not increase, but rather decreases, the variability of peptides that are potentially suitable for MHC class I antigen presentation. These unexpected findings indicate that global stimulation of the degradation of unfolded proteins may represent a more general feature of PA28γ and suggests that this proteasomal activator might play a broader role in the pathway of protein degradation than previously believed.
Collapse
Affiliation(s)
| | - Fulvia Cerruti
- Department of Veterinary Sciences, University of Turin, Largo P. Braccini 2, 10095, Grugliasco, Turin, Italy
| | - Justine Laulin
- Université Paris-Saclay, Institut Gustave Roussy, Inserm, Immunologie Des Tumeurs et Immunothérapie, Villejuif, France
| | | | - Angela Bachi
- The FIRC Institute of Molecular Oncology (IFOM), 20139, Milan, Italy
| | - Sebastien Apcher
- Université Paris-Saclay, Institut Gustave Roussy, Inserm, Immunologie Des Tumeurs et Immunothérapie, Villejuif, France
| | - Olivier Coux
- Centre de Recherche de Biologie Cellulaire de Montpellier (CRBM), CNRS UMR 5237, Université de Montpellier, 1919 Route de Mende, 34293, Montpellier, France
| | - Paolo Cascio
- Department of Veterinary Sciences, University of Turin, Largo P. Braccini 2, 10095, Grugliasco, Turin, Italy.
| |
Collapse
|
48
|
Abstract
Proteasomes are compartmentalized, ATP-dependent, N-terminal nucleophile hydrolases that play essentials roles in intracellular protein turnover. They are present in all 3 kingdoms. Pharmacological inhibition of proteasomes is detrimental to cell viability. Proteasome inhibitor rugs revolutionize the treatment of multiple myeloma. Proteasomes in pathogenic microbes such as Mycobacterium tuberculosis (Mtb), Plasmodium falciparum (Pf), and other parasites and worms have been validated as therapeutic targets. Starting with Mtb proteasome, efforts in developing inhibitors selective for microbial proteasomes have made great progress lately. In this review, we describe the strategies and pharmacophores that have been used in developing proteasome inhibitors with potency and selectivity that spare human proteasomes and highlight the development of clinical proteasome inhibitor candidates for treatment of leishmaniasis and Chagas disease. Finally, we discuss the future challenges and therapeutical potentials of the microbial proteasome inhibitors.
Collapse
Affiliation(s)
- Hao Zhang
- Department of Microbiology & Immunology, Weill Cornell Medicine, New York, New York, United States of America
| | - Gang Lin
- Department of Microbiology & Immunology, Weill Cornell Medicine, New York, New York, United States of America
- * E-mail:
| |
Collapse
|
49
|
An P, Zhang LJ, Peng W, Chen YY, Liu QP, Luan X, Zhang H. Natural products are an important source for proteasome regulating agents. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2021; 93:153799. [PMID: 34715511 DOI: 10.1016/j.phymed.2021.153799] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/26/2021] [Revised: 09/14/2021] [Accepted: 10/07/2021] [Indexed: 06/13/2023]
Abstract
BACKGROUND Natural medicines have a long history in the prevention and treatment of various diseases in East Asian region, especially in China. Modern research has proved that the pharmacological effects of numerous natural medicines involve the participation of ubiquitin proteasome system (UPS). UPS can degrade the unwanted and damaged proteins widely distributed in the nucleus and cytoplasm of various eukaryotes. PURPOSE The objective of the present study was to review and discuss the regulatory effects of natural products and extracts on proteasome components, which may help to find new proteasome regulators for drug development and clinical applications. METHODS The related information was compiled using the major scientific databases, such as CNKI, Elsevier, ScienceDirect, PubMed, SpringerLink, Wiley Online, and GeenMedical. The keywords "natural product" and "proteasome" were applied to extract the literature. Nature derived extracts, compounds and their derivatives involved in proteasome regulation were included, and the publications related to synthetic proteasome agents were excluded. RESULTS The pharmacological effects of more than 80 natural products and extracts derived from phytomedicines related to the proteasome regulation were reviewed. These natural products were classified according to their chemical properties. We also summarized some laws of action of natural products as proteasome regulators in the treatment of diseases, and listed the action characteristics of the typical natural products. CONCLUSION Natural products derived from nature can induce the degradation of damaged proteins through UPS or act as regulators to directly regulate the activity of proteasome. But few proteasome modulators are applied clinically. Summary of known rules for proteasome modulators will contribute to discover, modify and synthesize more proteasome modulators for clinical applications.
Collapse
Affiliation(s)
- Pei An
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, No. 1200, Cailun Road, Pudong New Area, Shanghai 201203, China
| | - Li-Jun Zhang
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, No. 1200, Cailun Road, Pudong New Area, Shanghai 201203, China
| | - Wei Peng
- School of pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Yu-Ying Chen
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, No. 1200, Cailun Road, Pudong New Area, Shanghai 201203, China
| | - Qiu-Ping Liu
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, No. 1200, Cailun Road, Pudong New Area, Shanghai 201203, China
| | - Xin Luan
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, No. 1200, Cailun Road, Pudong New Area, Shanghai 201203, China.
| | - Hong Zhang
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, No. 1200, Cailun Road, Pudong New Area, Shanghai 201203, China.
| |
Collapse
|
50
|
Parihar N, Bhatt LK. Deubiquitylating enzymes: potential target in autoimmune diseases. Inflammopharmacology 2021; 29:1683-1699. [PMID: 34792672 DOI: 10.1007/s10787-021-00890-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2021] [Accepted: 10/28/2021] [Indexed: 12/28/2022]
Abstract
The ubiquitin-proteasome pathway is responsible for the turnover of different cellular proteins, such as transport proteins, presentation of antigens to the immune system, control of the cell cycle, and activities that promote cancer. The enzymes which remove ubiquitin, deubiquitylating enzymes (DUBs), play a critical role in central and peripheral immune tolerance to prevent the development of autoimmune diseases and thus present a potential therapeutic target for the treatment of autoimmune diseases. DUBs function by removing ubiquitin(s) from target protein and block ubiquitin chain elongation. The addition and removal of ubiquitin molecules have a significant impact on immune responses. DUBs and E3 ligases both specifically cleave target protein and modulate protein activity and expression. The balance between ubiquitylation and deubiquitylation modulates protein levels and also protein interactions. Dysregulation of the ubiquitin-proteasome pathway results in the development of various autoimmune diseases such as inflammatory bowel diseases (IBD), psoriasis, multiple sclerosis (MS), systemic lupus erythematosus (SLE) and rheumatoid arthritis (RA). This review summarizes the current understanding of ubiquitination in autoimmune diseases and focuses on various DUBs responsible for the progression of autoimmune diseases.
Collapse
Affiliation(s)
- Niraj Parihar
- Department of Pharmacology, SVKM's Dr. Bhanuben Nanavati College of Pharmacy, Vile Parle (W), Mumbai, India
| | - Lokesh Kumar Bhatt
- Department of Pharmacology, SVKM's Dr. Bhanuben Nanavati College of Pharmacy, Vile Parle (W), Mumbai, India.
| |
Collapse
|