1
|
Chen J, Zhou Q, Su L, Ni L. Mitochondrial dysfunction: the hidden catalyst in chronic kidney disease progression. Ren Fail 2025; 47:2506812. [PMID: 40441691 PMCID: PMC12123951 DOI: 10.1080/0886022x.2025.2506812] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2025] [Accepted: 05/10/2025] [Indexed: 06/02/2025] Open
Abstract
Chronic kidney disease (CKD) represents a global health epidemic, with approximately one-third of affected individuals ultimately necessitating renal replacement therapy or transplantation. The kidney, characterized by its exceptionally high energy demands, exhibits significant sensitivity to alterations in energy supply and mitochondrial function. In CKD, a compromised capacity for mitochondrial ATP synthesis has been documented. As research advances, the multifaceted roles of mitochondria, extending beyond their traditional functions in oxygen sensing and energy production, are increasingly acknowledged. Empirical studies have demonstrated a strong association between mitochondrial dysfunction and the pathogenesis of fibrosis and cellular apoptosis in CKD. Targeting mitochondrial dysfunction holds substantial therapeutic promise, with emerging insights into its epigenetic regulation in CKD, particularly involving non-coding RNAs and DNA methylation. This article presents a comprehensive review of contemporary research on mitochondrial dysfunction in relation to the onset and progression of CKD. It elucidates the associated molecular mechanisms across various renal cell types and proposes novel research avenues for CKD treatment.
Collapse
Affiliation(s)
- Jinhu Chen
- Department of Nephrology, Huanggang Central Hospital of Yangtze University, Huanggang, China
- Department of Nephrology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Qiuyuan Zhou
- Department of Nephrology, Zhongnan Hospital of Wuhan University, Wuhan, China
- Department of Pathology, Liang Ping People’s Hospital of Chongqing, Chongqing, People’s Republic of China
| | - Lianjiu Su
- Department of Critical Care Medicine, Zhongnan Hospital of Wuhan University, Wuhan, China
- Clinical Research Center of Hubei Critical Care Medicine, Wuhan, China
- Department of Cardiology, David Geffen School of Medicine, University of California, Los Angeles, CA, USA
| | - Lihua Ni
- Department of Nephrology, Zhongnan Hospital of Wuhan University, Wuhan, China
| |
Collapse
|
2
|
Xu Y, Chen Y, Bai N, Su Y, Ye Y, Zhang R, Yang Y, Liu C, Hu C, Pan J. Deubiquitinating enzyme USP2 regulates brown adipose tissue thermogenesis via controlling EBF2 stabilization. Mol Metab 2025; 96:102139. [PMID: 40189098 PMCID: PMC12020889 DOI: 10.1016/j.molmet.2025.102139] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2024] [Revised: 03/23/2025] [Accepted: 04/01/2025] [Indexed: 04/14/2025] Open
Abstract
OBJECTIVE The activation of brown adipose tissue (BAT) promotes energy expenditure is recognized as a promising therapeutic strategy for combating obesity. The deubiquitinating enzyme family members are widely involved in the process of energy metabolism. However, the specific deubiquitinating enzyme member that affects the BAT thermogenesis remains largely unexplored. METHODS Adeno-associated virus, lentivirus and small molecule inhibitor were applied to generate USP2 gain- or loss-of-function both in vivo and in vitro. OxyMax comprehensive laboratory animal monitoring system, seahorse and transmission electron microscopy were used to determine the energy metabolism. Quantitative proteomics, immunofluorescence staining and co-immunoprecipitation were performed to reveal the potential substrates of USP2. RESULTS USP2 is upregulated upon thermogenic activation in adipose, and has a close correlation with UCP1 mRNA levels in human adipose tissue. BAT-specific Usp2 knockdown or systemic USP2 inhibition resulted in impaired thermogenic programs both in vivo and in vitro. Conversely, overexpression of Usp2 in BAT conferred protection against high-fat diet-induced obesity and associated metabolic disorders. Proteome-wide analysis identified EBF2 as the substrate of USP2 that mediates the thermogenic function of USP2 in BAT. CONCLUSIONS Our data demonstrated the vital role of USP2 in regulating BAT activation and systemic energy homeostasis. Activation of USP2-EBF2 interaction could be a potential therapeutic strategy against obesity.
Collapse
Affiliation(s)
- Yuejie Xu
- Department of Endocrinology and Metabolism, Shanghai Diabetes Institute, Shanghai Clinical Center for Diabetes, Shanghai Key Laboratory of Diabetes Mellitus, Shanghai Key Clinical Center for Metabolic Disease, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - Ying Chen
- Jinzhou Medical University Graduate Training Base (Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine), Jinzhou, 121001, China
| | - Ningning Bai
- Department of Endocrinology and Metabolism, Shanghai Diabetes Institute, Shanghai Clinical Center for Diabetes, Shanghai Key Laboratory of Diabetes Mellitus, Shanghai Key Clinical Center for Metabolic Disease, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - Yingying Su
- Department of Endocrinology and Metabolism, Shanghai Diabetes Institute, Shanghai Clinical Center for Diabetes, Shanghai Key Laboratory of Diabetes Mellitus, Shanghai Key Clinical Center for Metabolic Disease, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - Yafen Ye
- Department of Endocrinology and Metabolism, Shanghai Diabetes Institute, Shanghai Clinical Center for Diabetes, Shanghai Key Laboratory of Diabetes Mellitus, Shanghai Key Clinical Center for Metabolic Disease, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - Rong Zhang
- Department of Endocrinology and Metabolism, Shanghai Diabetes Institute, Shanghai Clinical Center for Diabetes, Shanghai Key Laboratory of Diabetes Mellitus, Shanghai Key Clinical Center for Metabolic Disease, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - Ying Yang
- Department of Endocrinology and Metabolism, Shanghai Diabetes Institute, Shanghai Clinical Center for Diabetes, Shanghai Key Laboratory of Diabetes Mellitus, Shanghai Key Clinical Center for Metabolic Disease, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - Caizhi Liu
- Department of Endocrinology and Metabolism, Shanghai Diabetes Institute, Shanghai Clinical Center for Diabetes, Shanghai Key Laboratory of Diabetes Mellitus, Shanghai Key Clinical Center for Metabolic Disease, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China.
| | - Cheng Hu
- Department of Endocrinology and Metabolism, Shanghai Diabetes Institute, Shanghai Clinical Center for Diabetes, Shanghai Key Laboratory of Diabetes Mellitus, Shanghai Key Clinical Center for Metabolic Disease, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China.
| | - Jiemin Pan
- Department of Endocrinology and Metabolism, Shanghai Diabetes Institute, Shanghai Clinical Center for Diabetes, Shanghai Key Laboratory of Diabetes Mellitus, Shanghai Key Clinical Center for Metabolic Disease, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China.
| |
Collapse
|
3
|
Shen C, Wang X, Gu L, Cui X, Zhu W, Wang Y, Zhang X, Chen X. USP41 plays carcinogenic roles in human cutaneous melanoma through PI3K/Akt signaling pathway. Arch Dermatol Res 2025; 317:768. [PMID: 40392309 DOI: 10.1007/s00403-025-04114-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2025] [Revised: 02/18/2025] [Accepted: 02/25/2025] [Indexed: 05/22/2025]
Abstract
Cutaneous melanoma is a malignant tumor with a high mortality rate. Ubiquitin-specific protease 41 (USP41) has recently been reported to be overexpressed in various malignancies. However, its role in melanoma remains unclear. Gene Expression Profiling Interactive Analysis (GEPIA) was used to perform pan-cancer analysis using data from the the Cancer Genome Atlas (TCGA) and the Genotype-Tissue Expression (GTEx) databases. Melanoma tissue microarray (TMA), clinical patient tissues, and cells were used to explore USP41 expression profiles by immunohistochemistry (IHC), RT-qPCR or Western blotting. Small interfering RNAs (siRNAs) were used to knock down USP41 in melanoma cells. Cell proliferation, migration, and invasion were assessed using CCK-8, EdU staining, wound healing, and transwell assays, respectively. Cell apoptosis was detected by flow cytometry and TUNEL staining. GEPIA revealed that USP41 is highly expressed in most human cancers, including melanoma. USP41 is overexpressed in melanoma tumor tissues and cells. IHC showed that USP41 was positively stained in melanoma tissues and was significantly correlated with the TNM stage of melanoma. USP41 knockdown inhibited cell proliferation, migration, and invasion while promoting cell apoptosis and inhibiting phosphorylated PI3K, AKT, and mTOR in the PI3K/AKT signaling pathway. The results indicate that USP41 may play a carcinogenic role in melanoma partly via the PI3K/AKT signaling pathway, suggesting that USP41 may be an effective therapeutic target for the treatments of cutaneous melanoma.
Collapse
Affiliation(s)
- Congcong Shen
- Department of Dermatology, Affiliated Hospital of Nantong University, 20 Xisi Road, Nantong, 226001, China
| | - Xin Wang
- Department of Dermatology, Affiliated Hospital of Nantong University, 20 Xisi Road, Nantong, 226001, China
| | - Lixiong Gu
- Department of Dermatology, Affiliated Hospital of Nantong University, 20 Xisi Road, Nantong, 226001, China
| | - Xiaomei Cui
- Department of Dermatology, Affiliated Hospital of Nantong University, 20 Xisi Road, Nantong, 226001, China
| | - Wenyan Zhu
- Department of Dermatology, Affiliated Hospital of Nantong University, 20 Xisi Road, Nantong, 226001, China
| | - Yixiao Wang
- Department of Dermatology, Affiliated Hospital of Nantong University, 20 Xisi Road, Nantong, 226001, China
| | - Xin Zhang
- Department of Dermatology, Affiliated Hospital of Nantong University, 20 Xisi Road, Nantong, 226001, China
| | - Xiaodong Chen
- Department of Dermatology, Affiliated Hospital of Nantong University, 20 Xisi Road, Nantong, 226001, China.
| |
Collapse
|
4
|
Li C, Zhao J, Ping H, Li Y, Lei R, Pan B, Liang G. OsbHLH062 negatively regulates Fe homeostasis by enhancing OsHRZ1 targeting OsPRIs for degradation. THE NEW PHYTOLOGIST 2025. [PMID: 40346870 DOI: 10.1111/nph.70207] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Accepted: 04/17/2025] [Indexed: 05/12/2025]
Abstract
Iron (Fe) is crucial for plant growth and development, and many transcription factors are involved in the maintenance of Fe homeostasis. In rice Fe signaling, the basic helix-loop-helix (bHLH) IVc proteins (OsPRI1/OsbHLH060, OsPRI2/OsbHLH058, OsPRI3/OsbHLH059, and OsPRI4/OsbHLH057) act as positive regulators, while Oryza sativa IRON-RELATED BHLH TRANSCRIPTION FACTOR 3 (OsIRO3/OsbHLH063) functions as a negative regulator. Additionally, HEMERYTHRIN MOTIF-CONTAINING REALLY INTERESTING NEW GENE AND ZINC-FINGER PROTEIN1 (OsHRZ1) plays a role in targeting OsPRIs for degradation. Here, we investigated the role of OsbHLH062 in response to Fe deficiency. We revealed that OsbHLH062 directly binds to the promoter region of Oryza sativa IRON-RELATED bHLH TRANSCRIPTION FACTOR 2 (OsIRO2), thereby repressing its activity through the recruitment of corepressors TOPLESS/TOPLESSRELATED (OsTPL/OsTPRs). When both OsbHLH062 and OsIRO3 are mutated simultaneously, it results in stunted root growth, reduced plant height, elevated Fe concentration, and the upregulation of Fe deficiency inducible genes. OsbHLH062 is found to be localized in both the cytoplasm and the nucleus. The interaction of OsbHLH062 with OsPRIs promotes its accumulation in the nucleus. Our findings also reveal that both OsbHLH062 and OsIRO3 physically interact with OsHRZ1, which in turn enhances the interaction between OsHRZ1 and OsPRI3, leading to the degradation of OsPRI3 by OsHRZ1. These findings shed light on a new function of OsbHLH062 and OsIRO3 as potential cofactors of OsHRZ1, which contribute to the reduction in OsPRI proteins and, consequently, the repression of Fe signaling pathways.
Collapse
Affiliation(s)
- Chenyang Li
- State Key Laboratory of Plant Diversity and Specialty Crops, Xishuangbanna Tropical Botanical Garden, Chinese Academy of Sciences, Kunming, Yunnan, 650223, China
| | - Junhui Zhao
- State Key Laboratory of Plant Diversity and Specialty Crops, Xishuangbanna Tropical Botanical Garden, Chinese Academy of Sciences, Kunming, Yunnan, 650223, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Huaqian Ping
- State Key Laboratory of Plant Diversity and Specialty Crops, Xishuangbanna Tropical Botanical Garden, Chinese Academy of Sciences, Kunming, Yunnan, 650223, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Yang Li
- State Key Laboratory of Plant Diversity and Specialty Crops, Xishuangbanna Tropical Botanical Garden, Chinese Academy of Sciences, Kunming, Yunnan, 650223, China
| | - Rihua Lei
- State Key Laboratory of Plant Diversity and Specialty Crops, Xishuangbanna Tropical Botanical Garden, Chinese Academy of Sciences, Kunming, Yunnan, 650223, China
| | - Bangzhen Pan
- State Key Laboratory of Plant Diversity and Specialty Crops, Xishuangbanna Tropical Botanical Garden, Chinese Academy of Sciences, Kunming, Yunnan, 650223, China
| | - Gang Liang
- State Key Laboratory of Plant Diversity and Specialty Crops, Xishuangbanna Tropical Botanical Garden, Chinese Academy of Sciences, Kunming, Yunnan, 650223, China
| |
Collapse
|
5
|
Chen YF, Teng YC, Yang JH, Kao CH, Tsai TF. Cisd1 synergizes with Cisd2 to modulate protein processing by maintaining mitochondrial and ER homeostasis. Aging (Albany NY) 2025; 17:206249. [PMID: 40349253 DOI: 10.18632/aging.206249] [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: 05/22/2024] [Accepted: 03/24/2025] [Indexed: 05/14/2025]
Abstract
Connection and crosstalk among the organelles critically contribute to cellular functions. Destruction of any kind of organelle is likely to induce a series of intracellular disorders and finally lead to cell death. Because of its subcellular locations, CDGSH iron-sulfur domain-containing protein 1 (Cisd1) and Cisd2 have functions that are related to maintaining mitochondria and ER homeostasis. As previous reports have shown, Cisd2 knockout mice have a decreased body weight and poor survival rate, and the primary defects were conducted in skeletal muscle. Our previous findings indicated that Cisd1 deletion causes a range of skeletal muscle defects in mice with Cisd2 deficiency, including mitochondrial degeneration, endoplasmic reticulum (ER) stress, and alteration of protein process, as well as programmed cell death. In Cisd1 and Cisd2 deficient condition, the whole of the protein biosynthesis was damaged, including translation, modification, transport, and degradation. Changes in the immune response, redox regulation, and metabolism were also present in Cisd1 and Cisd2 double knockout mice. Overall, we have demonstrated that Cisd1 and Cisd2 knockout have a synergistic effect on skeletal muscles, and that Cisd2 plays a more critical role than Cisd1. These synergistic effects impact signaling regulation and interrupt the crosstalk and homeostasis of organelles. This creates severe disorders in various tissues and organs.
Collapse
Affiliation(s)
- Yi-Fan Chen
- International Master Program for Translation Science, College of Medical Science and Technology, Taipei Medical University, New Taipei City 23564, Taiwan
- The Ph.D. Program for Translational Medicine, College of Medical Science and Technology, Taipei Medical University, Taipei 11529, Taiwan
- TMU Research Center of Cancer Translational Medicine, Taipei Medical University, Taipei 11031, Taiwan
- International Ph.D. Program for Translational Science, College of Medical Science and Technology, Taipei Medical University, Taipei 11031, Taiwan
- Master Program in Clinical Genomics and Proteomics, School of Pharmacy, Taipei Medical University, Taipei 11031, Taiwan
| | - Yuan-Chi Teng
- Department of Life Sciences and Institute of Genome Sciences, National Yang Ming Chiao Tung University, Taipei 112, Taiwan
- Department of Medical Research, Taipei Veterans General Hospital, Taipei 112, Taiwan
| | - Jian-Hsin Yang
- Department of Life Sciences and Institute of Genome Sciences, National Yang Ming Chiao Tung University, Taipei 112, Taiwan
| | - Cheng-Heng Kao
- Center of General Education, Chang Gung University, Taoyuan 333, Taiwan
| | - Ting-Fen Tsai
- Department of Life Sciences and Institute of Genome Sciences, National Yang Ming Chiao Tung University, Taipei 112, Taiwan
- Center for Healthy Longevity and Aging Sciences, National Yang Ming Chiao Tung University, Taipei 112, Taiwan
- Institute of Molecular and Genomic Medicine, National Health Research Institutes, Zhunan 350, Taiwan
| |
Collapse
|
6
|
Pan M, Fu Z, Hou H, Yang C, Li J. Proteolysis-Targeting Chimera (PROTAC): A Revolutionary Tool for Chemical Biology Research. SMALL METHODS 2025:e2500402. [PMID: 40342226 DOI: 10.1002/smtd.202500402] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/26/2025] [Revised: 04/24/2025] [Indexed: 05/11/2025]
Abstract
Proteolysis-targeting chimera (PROTAC) technology is a revolutionary tool for drug discovery that simultaneously recruits E3 ligase and the protein of interest to induce ubiquitination and subsequent proteasomal degradation. Since the inaugural PROTAC prototype emerged in 2001, this modality has garnered significant interest across academia and industry, catalyzing transformative applications in drug discovery and chemical biology. The field has evolved from foundational investigations into molecular design, structural optimization, and protein target extension to address more sophisticated challenges, such as structural analysis of ternary complexes, expansion of diversified therapeutic indications, and clinical translation studies. Recent progress across chemical, pharmaceutical, and biochemical sciences has reshaped PROTAC design paradigms, which in turn expanded the chemical biology toolkit. In this review, pivotal milestones are systematically chronicled in PROTAC development, evaluate emerging strategies for diversifying E3 ligase utilization and expanding the scope of degradable targets, and summarize a series of instrumental and biochemical methodologies that propelled sequential breakthroughs. Additionally, forward-looking trajectories are proposed to address current limitations and accelerate the clinical maturation of PROTAC-based therapeutics.
Collapse
Affiliation(s)
- Meichen Pan
- Beijing Life Science Academy, Beijing, 102209, China
- School of Biomedical Sciences, Hunan University, Changsha, Hunan, 410082, China
| | - Zhongliang Fu
- Beijing Life Science Academy, Beijing, 102209, China
- School of Biomedical Sciences, Hunan University, Changsha, Hunan, 410082, China
| | - Hongwei Hou
- Beijing Life Science Academy, Beijing, 102209, China
| | - Chunrong Yang
- New Cornerstone Science Laboratory, Department of Chemistry, Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology, Tsinghua University, Beijing, 100084, China
- Beijing Life Science Academy, Beijing, 102209, China
| | - Jinghong Li
- New Cornerstone Science Laboratory, Department of Chemistry, Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology, Tsinghua University, Beijing, 100084, China
- Beijing Life Science Academy, Beijing, 102209, China
- Center for Bioanalytical Chemistry, Hefei National Laboratory of Physical Science at Microscale, University of Science and Technology of China, Hefei, 230026, China
| |
Collapse
|
7
|
Ji XL, Zhao LL, Liu B, Yuan YB, Han Y, You CX, An JP. MdZFP7 integrates JA and GA signals via interaction with MdJAZ2 and MdRGL3a in regulating anthocyanin biosynthesis and undergoes degradation by the E3 ubiquitin ligase MdBRG3. JOURNAL OF INTEGRATIVE PLANT BIOLOGY 2025; 67:1339-1363. [PMID: 39936840 DOI: 10.1111/jipb.13862] [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: 07/27/2024] [Accepted: 01/10/2025] [Indexed: 02/13/2025]
Abstract
Jasmonic acid (JA) and gibberellin (GA) coordinate many aspects of plant growth and development, including anthocyanin biosynthesis. However, the crossover points of JA and GA signals and the pathways through which they interact to regulate anthocyanin biosynthesis are poorly understood. Here, we investigated the molecular mechanism by which the zinc finger protein (ZFP) transcription factor Malus domestica ZFP7 (MdZFP7) regulates anthocyanin biosynthesis by integrating JA and GA signals at the transcriptional and post-translational levels. MdZFP7 is a positive regulator of anthocyanin biosynthesis, which fulfills its role by directly activating the expression of MdMYB1 and enhancing the transcriptional activation of MdWRKY6 on the target genes MdDFR and MdUF3GT. MdZFP7 integrates JA and GA signals by interacting with the JA repressor apple JASMONATE ZIM-DOMAIN2 (MdJAZ2) and the GA repressor apple REPRESSOR-of-ga1-3-like 3a (MdRGL3a). MdJAZ2 weakens the transcriptional activation of MdMYB1 by MdZFP7 and disrupts the MdZFP7-MdWRKY6 interaction, thereby reducing the anthocyanin biosynthesis promoted by MdZFP7. MdRGL3a contributes to the stimulation of anthocyanin biosynthesis by MdZFP7 by sequestering MdJAZ2 from the MdJAZ2-MdZFP7 complex. The E3 ubiquitin ligase apple BOI-related E3 ubiquitin-protein ligase 3 (MdBRG3), which is antagonistically regulated by JA and GA, targets the ubiquitination degradation of MdZFP7. The MdBRG3-MdZFP7 module moves the crosstalk of JA and GA signals from the realm of transcriptional regulation and into the protein post-translational modification. In conclusion, this study not only elucidates the node-role of MdZFP7 in the integration of JA and GA signals, but also describes the transcriptional and post-translational regulatory network of anthocyanin biosynthesis with MdZFP7 as the hub.
Collapse
Affiliation(s)
- Xing-Long Ji
- College of Horticulture, Qingdao Agricultural University, Qingdao, 266109, China
| | - Ling-Ling Zhao
- Yantai Academy of Agricultural Sciences, Yantai, 265599, China
| | - Baoyou Liu
- Yantai Academy of Agricultural Sciences, Yantai, 265599, China
| | - Yong-Bing Yuan
- College of Horticulture, Qingdao Agricultural University, Qingdao, 266109, China
| | - Yuepeng Han
- State Key Laboratory of Plant Diversity and Specialty Crops, CAS Key Laboratory of Plant Germplasm Enhancement and Specialty Agriculture, Wuhan Botanical Garden of Chinese Academy of Sciences, Wuhan, 430074, China
- Hubei Hongshan Laboratory, Wuhan, 430070, China
| | - Chun-Xiang You
- College of Horticulture Science and Engineering, Shandong Agricultural University, Tai'an, 271018, China
| | - Jian-Ping An
- State Key Laboratory of Plant Diversity and Specialty Crops, CAS Key Laboratory of Plant Germplasm Enhancement and Specialty Agriculture, Wuhan Botanical Garden of Chinese Academy of Sciences, Wuhan, 430074, China
- Hubei Hongshan Laboratory, Wuhan, 430070, China
| |
Collapse
|
8
|
Wu Z, Li Y, Dong J, Qin JJ. An updated review on the role of small molecules in mediating protein degradation. Eur J Med Chem 2025; 287:117370. [PMID: 39933402 DOI: 10.1016/j.ejmech.2025.117370] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2024] [Revised: 01/25/2025] [Accepted: 02/03/2025] [Indexed: 02/13/2025]
Abstract
Targeted protein degradation (TPD) technologies, inspired by physiological processes, have recently provided new directions for drug development. Unlike conventional drug development focusing on targeting the active sites of disease-related proteins, TPD can utilize any nook or cranny of a protein to drive degradation through the cell's inherent destruction mechanism. It offers various advantages such as stronger pharmacological effects, an expanded range of drug targets, and higher selectivity. Based on the ubiquitin-proteasome system and the lysosomal degradation pathway, a variety of TPD strategies have been developed including PROTAC, PROTAB, and AUTOTAC. These TPD strategies have continuously enriched the toolbox for targeted protein degradation and expanded the scope of application, providing new ideas for biological research and drug discovery. This review attempts to introduce up-to-date research progress in the TPD strategies, focusing mainly on their design concepts, advantages, potential applications, and challenges, which may provide some inspiration for drug design, drug discovery, and clinical application for biologists and chemists.
Collapse
Affiliation(s)
- Zumei Wu
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Yulong Li
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Jinyun Dong
- Center for Innovative Drug Research, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, 310022, China.
| | - Jiang-Jiang Qin
- Center for Innovative Drug Research, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, 310022, China.
| |
Collapse
|
9
|
Salomonsson J, Sjöstrand L, Eskilson A, Derbyshire D, D'Arcy P, Sunnerhagen M, Ahlner A. Dynamic networks connect the USP14 active site region with the proteasome interaction surface. Protein Sci 2025; 34:e70077. [PMID: 40095364 PMCID: PMC11912437 DOI: 10.1002/pro.70077] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2024] [Revised: 01/03/2025] [Accepted: 02/06/2025] [Indexed: 03/19/2025]
Abstract
Ubiquitin-specific protease 14 (USP14) is a member of the USP family responsible for the catalytic removal of ubiquitin (Ub) from proteins directed to the proteasome, implicated in the pathogenesis of neurodegeneration and cancer. Crystallography and cryo-EM analysis have identified loop regions crucial for the deubiquitinase activity of USP14, specifically those involved in Ub and proteasome binding. However, the structural changes in USP14 upon ligand binding to these regions are minimal, indicating significant yet uncharacterized dynamic contributions to its function. In this study, through structural and dynamical NMR experiments and functional evaluation, we demonstrate that small mutations designed to impact Ub binding and catalytic activity without disturbing the USP structure display both local and long-range effects. The affected residues connect the catalytic site and the Ub binding region with the proteasome interaction surface through a network of loops, which show varied dynamics on the ps-ms time scale. Collectively, our findings experimentally reveal different aspects of dynamic connections within USP14, suggesting the presence of allosteric networks that link enzyme activity with regulatory function. The identification of coupled clusters of possible allostery participants in the free USP domain provides new insights into the dynamic regulation of USP14, with potential implications for understanding its role in cellular processes.
Collapse
Affiliation(s)
| | - Linda Sjöstrand
- Department of Biomedical and Clinical SciencesLinköping UniversityLinköpingSweden
| | - Arvid Eskilson
- Department of PhysicsChemistry and Biology, Linköping UniversityLinköpingSweden
| | - Dean Derbyshire
- Department of PhysicsChemistry and Biology, Linköping UniversityLinköpingSweden
| | - Pádraig D'Arcy
- Department of Biomedical and Clinical SciencesLinköping UniversityLinköpingSweden
| | - Maria Sunnerhagen
- Department of PhysicsChemistry and Biology, Linköping UniversityLinköpingSweden
| | - Alexandra Ahlner
- Department of PhysicsChemistry and Biology, Linköping UniversityLinköpingSweden
| |
Collapse
|
10
|
Zanfardino P, Amati A, Perrone M, Petruzzella V. The Balance of MFN2 and OPA1 in Mitochondrial Dynamics, Cellular Homeostasis, and Disease. Biomolecules 2025; 15:433. [PMID: 40149969 PMCID: PMC11940761 DOI: 10.3390/biom15030433] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2025] [Revised: 03/10/2025] [Accepted: 03/11/2025] [Indexed: 03/29/2025] Open
Abstract
Mitochondrial dynamics, governed by fusion and fission, are crucial for maintaining cellular homeostasis, energy production, and stress adaptation. MFN2 and OPA1, key regulators of mitochondrial fusion, play essential roles beyond their structural functions, influencing bioenergetics, intracellular signaling, and quality control mechanisms such as mitophagy. Disruptions in these processes, often caused by MFN2 or OPA1 mutations, are linked to neurodegenerative diseases like Charcot-Marie-Tooth disease type 2A (CMT2A) and autosomal dominant optic atrophy (ADOA). This review explores the molecular mechanisms underlying mitochondrial fusion, the impact of MFN2 and OPA1 dysfunction on oxidative phosphorylation and autophagy, and their role in disease progression. Additionally, we discuss the divergent cellular responses to MFN2 and OPA1 mutations, particularly in terms of proliferation, senescence, and metabolic signaling. Finally, we highlight emerging therapeutic strategies to restore mitochondrial integrity, including mTOR modulation and autophagy-targeted approaches, with potential implications for neurodegenerative disorders.
Collapse
Affiliation(s)
| | | | | | - Vittoria Petruzzella
- Department of Translational Biomedicine and Neurosciences (DiBraiN), University of Bari Aldo Moro, Piazza Giulio Cesare, 70124 Bari, Italy; (P.Z.); (A.A.); (M.P.)
| |
Collapse
|
11
|
Hurła M, Pikor D, Banaszek-Hurła N, Drelichowska A, Dorszewska J, Kozubski W, Kacprzak E, Paul M. Unraveling the Role of Proteinopathies in Parasitic Infections. Biomedicines 2025; 13:610. [PMID: 40149586 PMCID: PMC11940292 DOI: 10.3390/biomedicines13030610] [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: 02/03/2025] [Revised: 02/26/2025] [Accepted: 02/27/2025] [Indexed: 03/29/2025] Open
Abstract
Proteinopathies, characterized by the misfolding, aggregation, and deposition of proteins, are hallmarks of various neurodegenerative and systemic diseases. Increasingly, research has highlighted the role of protein misfolding in parasitic infections, unveiling intricate interactions between host and parasite that exacerbate disease pathology and contribute to chronic outcomes. The life cycles of parasitic protozoa, including Plasmodium, Toxoplasmosis, and Leishmania species, are complicated and involve frequent changes between host and vector environments. Their proteomes are severely stressed during these transitions, which calls for highly specialized protein quality control systems. In order to survive harsh intracellular conditions during infection, these parasites have been demonstrated to display unique adaptations in the unfolded protein response, a crucial pathway controlling endoplasmic reticulum stress. In addition to improving parasite survival, these adaptations affect host cell signaling and metabolism, which may jeopardize cellular homeostasis. By causing oxidative stress, persistent inflammation, and disturbance of cellular proteostasis, host-parasite interactions also contribute to proteinopathy. For instance, Plasmodium falciparum disrupts normal protein homeostasis and encourages the accumulation of misfolded proteins by influencing host redox systems involved in protein folding. In addition to interfering with host chaperone systems, the parasitic secretion of effector proteins exacerbates protein misfolding and aggregate formation. Autophagy, apoptosis regulation, organelle integrity, and other vital cellular processes are all disrupted by these pathological protein aggregates. Long-term misfolding and aggregation can cause irreversible tissue damage, which can worsen the clinical course of illnesses like visceral leishmaniasis, cerebral malaria, and toxoplasmosis. Treating parasite-induced proteinopathies is a potentially fruitful area of therapy. According to recent research, autophagy modulators, proteasome enhancers, and small-molecule chaperones may be repurposed to lessen these effects. Pharmacological agents that target the UPR, for example, have demonstrated the ability to decrease parasite survival while also reestablishing host protein homeostasis. Targeting the proteins secreted by parasites that disrupt host proteostasis may also offer a novel way to stop tissue damage caused by proteinopathies. In conclusion, the intersection of protein misfolding and parasitic infections represents a rapidly advancing field of research. Dissecting the molecular pathways underpinning these processes offers unprecedented opportunities for developing innovative therapies. These insights could not only transform the management of parasitic diseases but also contribute to a broader understanding of proteinopathies in infectious and non-infectious diseases alike.
Collapse
Affiliation(s)
- Mikołaj Hurła
- Department of Tropical and Parasitic Diseases, Central University Hospital, Przybyszewskiego 49, 61-701 Poznan, Poland
| | - Damian Pikor
- Department of Internal Medicine, University of Medical Sciences, Przybyszewskiego 49, 60-355 Poznan, Poland
| | - Natalia Banaszek-Hurła
- Laboratory of Neurobiology, Department of Neurology, Poznan University of Medical Sciences, 60-355 Poznan, Poland
| | - Alicja Drelichowska
- Student Scientific Society of Poznan, University of Medical Sciences, 60-806 Poznan, Poland
| | - Jolanta Dorszewska
- Laboratory of Neurobiology, Department of Neurology, Poznan University of Medical Sciences, 60-355 Poznan, Poland
| | - Wojciech Kozubski
- Chair and Department of Neurology, Poznan University of Medical Sciences, 60-355 Poznan, Poland
| | - Elżbieta Kacprzak
- Department of Tropical and Parasitic Diseases, Central University Hospital, Przybyszewskiego 49, 61-701 Poznan, Poland
| | - Małgorzata Paul
- Department of Internal Medicine, University of Medical Sciences, Przybyszewskiego 49, 60-355 Poznan, Poland
| |
Collapse
|
12
|
Ou L, Setegne MT, Elliot J, Shen F, Dassama LMK. Protein-Based Degraders: From Chemical Biology Tools to Neo-Therapeutics. Chem Rev 2025; 125:2120-2183. [PMID: 39818743 PMCID: PMC11870016 DOI: 10.1021/acs.chemrev.4c00595] [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: 08/08/2024] [Revised: 12/26/2024] [Accepted: 12/30/2024] [Indexed: 01/19/2025]
Abstract
The nascent field of targeted protein degradation (TPD) could revolutionize biomedicine due to the ability of degrader molecules to selectively modulate disease-relevant proteins. A key limitation to the broad application of TPD is its dependence on small-molecule ligands to target proteins of interest. This leaves unstructured proteins or those lacking defined cavities for small-molecule binding out of the scope of many TPD technologies. The use of proteins, peptides, and nucleic acids (otherwise known as "biologics") as the protein-targeting moieties in degraders addresses this limitation. In the following sections, we provide a comprehensive and critical review of studies that have used proteins and peptides to mediate the degradation and hence the functional control of otherwise challenging disease-relevant protein targets. We describe existing platforms for protein/peptide-based ligand identification and the drug delivery systems that might be exploited for the delivery of biologic-based degraders. Throughout the Review, we underscore the successes, challenges, and opportunities of using protein-based degraders as chemical biology tools to spur discoveries, elucidate mechanisms, and act as a new therapeutic modality.
Collapse
Affiliation(s)
- Lisha Ou
- Department
of Chemistry, Stanford University, Stanford, California 94305, United States
- Sarafan
ChEM-H Institute, Stanford University, Stanford, California 94305, United States
| | - Mekedlawit T. Setegne
- Department
of Chemistry, Stanford University, Stanford, California 94305, United States
- Sarafan
ChEM-H Institute, Stanford University, Stanford, California 94305, United States
| | - Jeandele Elliot
- Department
of Chemical Engineering, Stanford University, Stanford, California 94305, United States
| | - Fangfang Shen
- Department
of Chemistry, Stanford University, Stanford, California 94305, United States
| | - Laura M. K. Dassama
- Department
of Chemistry, Stanford University, Stanford, California 94305, United States
- Sarafan
ChEM-H Institute, Stanford University, Stanford, California 94305, United States
- Department
of Microbiology & Immunology, Stanford
School of Medicine, Stanford, California 94305, United States
| |
Collapse
|
13
|
Li Y, Wu Y, Gao S, Sun T, Jiang C. PROTAC delivery in tumor immunotherapy: Where are we and where are we going? J Control Release 2025; 378:116-144. [PMID: 39637991 DOI: 10.1016/j.jconrel.2024.11.076] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2024] [Revised: 11/22/2024] [Accepted: 11/28/2024] [Indexed: 12/07/2024]
Abstract
Immunotherapy has emerged as a pioneering therapeutic modality, particularly within the realm of oncology, where Chimeric Antigen Receptor T-cell (CAR-T) therapy has manifested significant efficacy in the treatment of hematological malignancies. Nonetheless, the extension of immunotherapy to solid tumors poses a considerable challenge. This challenge is largely attributed to the intrinsic "cold" characteristics of certain tumors, which are defined by scant T-cell infiltration and a diminished immune response. Additionally, the impediment is exacerbated by the elusive nature of numerous targets within the tumor microenvironment, notably those deemed "undruggable" by small molecule inhibitors. This scenario underscores an acute necessity for the inception of innovative therapeutic strategies aimed at countering the resistance mechanisms underlying immune evasion in cold tumors, thereby amplifying the efficacy of cancer immunotherapy. Among the promising strategies is the deployment of Proteolysis Targeting Chimeras (PROTACs), which facilitate the targeted degradation of proteins. PROTACs present unique advantages and have become indispensable in oncology. However, they concurrently grapple with challenges such as solubility issues, permeability barriers, and the classical Hook effect. Notably, advanced delivery systems have been instrumental in surmounting these obstacles. This review commences with an analysis of the factors contributing to the suboptimal responses to immunotherapy in cold tumors. Subsequently, it delivers a thorough synthesis of immunotherapeutic concepts tailored for these tumors, clarifying the integral role of PROTACs in their management and delineating the trajectory of PROTAC technology from bench-side investigation to clinical utilization, facilitated by drug delivery systems. Ultimately, the review extrapolates the prospective future of this approach, aspiring to present novel insights that could catalyze progress in immunotherapy for the treatment of cold tumors.
Collapse
Affiliation(s)
- Yiyang Li
- Key Laboratory of Smart Drug DeliveryMinistry of Education, Minhang Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Department of Pharmaceutics, School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Yike Wu
- Key Laboratory of Smart Drug DeliveryMinistry of Education, Minhang Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Department of Pharmaceutics, School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Sihan Gao
- Key Laboratory of Smart Drug DeliveryMinistry of Education, Minhang Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Department of Pharmaceutics, School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Tao Sun
- Key Laboratory of Smart Drug DeliveryMinistry of Education, Minhang Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Department of Pharmaceutics, School of Pharmacy, Fudan University, Shanghai 201203, China; Quzhou Fudan Institute, Quzhou 324003, China.
| | - Chen Jiang
- Key Laboratory of Smart Drug DeliveryMinistry of Education, Minhang Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Department of Pharmaceutics, School of Pharmacy, Fudan University, Shanghai 201203, China; Department of Digestive Diseases, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou 350212, China.
| |
Collapse
|
14
|
Ibrahim S, Umer Khan M, Khurram I, Rehman R, Rauf A, Ahmad Z, Aljohani ASM, Al Abdulmonem W, Quradha MM. Navigating PROTACs in Cancer Therapy: Advancements, Challenges, and Future Horizons. Food Sci Nutr 2025; 13:e70011. [PMID: 39898116 PMCID: PMC11786021 DOI: 10.1002/fsn3.70011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2024] [Revised: 01/02/2025] [Accepted: 01/07/2025] [Indexed: 02/04/2025] Open
Abstract
Proteolysis Targeting Chimeras (PROTACs) have revolutionized cancer therapy by offering a selective and innovative approach to degrade key oncogenic proteins associated with various malignancies. These hybrid molecules exploit the ubiquitin-proteasome system, facilitating the degradation of target proteins through an event-driven mechanism, thereby overcoming drug resistance and enhancing selectivity. With diverse targets including androgen receptors, BTK, estrogen receptors, BET proteins, and BRAF, PROTACs offer a versatile strategy for personalized cancer treatment. Advantages of PROTACs over traditional small molecule inhibitors include their ability to operate at lower concentrations, catalyzing the degradation of multiple proteins of interest with reduced cytotoxicity. Notably, PROTACs address challenges associated with traditionally "undruggable" targets, expanding the therapeutic landscape of cancer therapy. Ongoing preclinical and clinical studies highlight the transformative potential of PROTACs, with promising results in prostate, breast, lung, melanoma, and colorectal cancers. Despite their potential, challenges persist in optimizing physicochemical properties and enhancing bioavailability. Further research is needed to refine PROTAC design and address complexities in molecule development. Nevertheless, the development of oral androgen receptor PROTACs represents a significant milestone, demonstrating the feasibility and efficacy of this innovative therapeutic approach. This review provides a comprehensive overview of PROTACs in cancer therapy, emphasizing their mechanism of action, advantages, and challenges. As PROTAC research progresses, continued exploration in both preclinical and clinical settings will be crucial to unlocking their full therapeutic potential and shaping the future of personalized cancer treatment.
Collapse
Affiliation(s)
- Saooda Ibrahim
- Institute of Molecular Biology and BiotechnologyThe University of LahoreLahorePakistan
- Centre for Applied Molecular BiologyUniversity of the PunjabLahorePakistan
| | - Muhammad Umer Khan
- Institute of Molecular Biology and BiotechnologyThe University of LahoreLahorePakistan
| | - Iqra Khurram
- Institute of Molecular Biology and BiotechnologyThe University of LahoreLahorePakistan
- Centre for Applied Molecular BiologyUniversity of the PunjabLahorePakistan
| | - Raima Rehman
- Institute of Molecular Biology and BiotechnologyThe University of LahoreLahorePakistan
| | - Abdur Rauf
- Department of ChemistryUniversity of SwabiSwabiKhyber PakhtunkhwaPakistan
| | - Zubair Ahmad
- Department of ChemistryUniversity of SwabiSwabiKhyber PakhtunkhwaPakistan
| | - Abdullah S. M. Aljohani
- Department of Medical BiosciencesCollege of Veterinary Medicine, Qassim UniversityBuraydahSaudi Arabia
| | - Waleed Al Abdulmonem
- Department of PathologyCollege of Medicine, Qassim UniversityBuraydahSaudi Arabia
| | | |
Collapse
|
15
|
Chen P, Hu JJ, Liu Y, Cao B, Song XJ. VCP controls KCC2 degradation through FAF1 recruitment and accelerates emergence from anesthesia. Proc Natl Acad Sci U S A 2025; 122:e2414016122. [PMID: 39793039 PMCID: PMC11725920 DOI: 10.1073/pnas.2414016122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2024] [Accepted: 11/26/2024] [Indexed: 01/12/2025] Open
Abstract
Ubiquitin-proteasomal degradation of K+/Cl- cotransporter 2 (KCC2) in the ventral posteromedial nucleus (VPM) has been demonstrated to serve as a common mechanism by which the brain emerges from anesthesia and regains consciousness. Ubiquitin-proteasomal degradation of KCC2 during anesthesia is driven by E3 ligase Fbxl4. However, the mechanism by which ubiquitinated KCC2 is targeted to the proteasome has not been elucidated. We report in cultured neuro-2a cells that the valosin-containing protein (VCP) transported ubiquitinated KCC2 to the proteasome and in mice in vivo experiments that inhibition of VCP restored KCC2 expression in the VPM and enhanced the effects of anesthesia. In cultured neuro-2a cells, propofol-induced degradation of KCC2 was inhibited by VCP inhibitor DBeQ and VCP knockout plasmid sgRNA(VCP). Propofol-induced enhanced interaction between VCP and KCC2 was inhibited by knockout of Fbxl4 or Fas-associated factor 1 (FAF1). In in vivo studies, pharmacological or genetic inhibition of VCP in the VPM significantly prevented KCC2 degradation and enhanced propofol anesthesia; these effects were abrogated by a KCC2 antagonist VU0463271. These results demonstrate that the VCP controls ubiquitin-proteasomal degradation of KCC2 dependent on FAF1 recruitment and serves as a mechanism for the ubiquitin-proteasomal degradation of KCC2, which is responsible for the subsequent emergence from anesthesia.
Collapse
Affiliation(s)
- Peng Chen
- Department of Medical Neuroscience, SUSTech Center for Pain Medicine, School of Medicine, Southern University of Science and Technology, Shenzhen518055, China
| | - Jiang-Jian Hu
- Department of Medical Neuroscience, SUSTech Center for Pain Medicine, School of Medicine, Southern University of Science and Technology, Shenzhen518055, China
| | - Yuexin Liu
- Department of Medical Neuroscience, SUSTech Center for Pain Medicine, School of Medicine, Southern University of Science and Technology, Shenzhen518055, China
| | - Boxu Cao
- Department of Medical Neuroscience, SUSTech Center for Pain Medicine, School of Medicine, Southern University of Science and Technology, Shenzhen518055, China
| | - Xue-Jun Song
- Department of Medical Neuroscience, SUSTech Center for Pain Medicine, School of Medicine, Southern University of Science and Technology, Shenzhen518055, China
| |
Collapse
|
16
|
Sabeh P, Dumas SA, Maios C, Daghar H, Korzeniowski M, Rousseau J, Lines M, Guerin A, Millichap JJ, Landsverk M, Grebe T, Lindstrom K, Strober J, Ait Mouhoub T, Zweier C, Steinraths M, Hebebrand M, Callewaert B, Abou Jamra R, Kautza-Lucht M, Wegler M, Kruszka P, Kumps C, Banne E, Waberski MB, Dieux A, Raible S, Krantz I, Medne L, Pechter K, Villard L, Guerrini R, Bianchini C, Barba C, Mei D, Blanc X, Kallay C, Ranza E, Yang XR, O'Heir E, Donald KA, Murugasen S, Bruwer Z, Calikoglu M, Mathews JM, Lesieur-Sebellin M, Baujat G, Derive N, Pierson TM, Murrell JR, Shillington A, Ormieres C, Rondeau S, Reis A, Fernandez-Jaen A, Au PYB, Sweetser DA, Briere LC, Couque N, Perrin L, Schymick J, Gueguen P, Lefebvre M, Van Andel M, Juusola J, Antonarakis SE, Parker JA, Burnett BG, Campeau PM. Heterozygous UBR5 variants result in a neurodevelopmental syndrome with developmental delay, autism, and intellectual disability. Am J Hum Genet 2025; 112:75-86. [PMID: 39721588 PMCID: PMC11739880 DOI: 10.1016/j.ajhg.2024.11.009] [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: 12/08/2023] [Revised: 11/19/2024] [Accepted: 11/19/2024] [Indexed: 12/28/2024] Open
Abstract
E3 ubiquitin ligases have been linked to developmental diseases including autism, Angelman syndrome (UBE3A), and Johanson-Blizzard syndrome (JBS) (UBR1). Here, we report variants in the E3 ligase UBR5 in 29 individuals presenting with a neurodevelopmental syndrome that includes developmental delay, autism, intellectual disability, epilepsy, movement disorders, and/or genital anomalies. Their phenotype is distinct from JBS due to the absence of exocrine pancreatic insufficiency and the presence of autism, epilepsy, and, in some probands, a movement disorder. E3 ubiquitin ligases are responsible for transferring ubiquitin to substrate proteins to regulate a variety of cellular functions, including protein degradation, protein-protein interactions, and protein localization. Knocking out ubr-5 in C. elegans resulted in a lower movement score compared to the wild type, supporting a role for UBR5 in neurodevelopment. Using an in vitro autoubiquitination assay and confocal microscopy for the human protein, we found decreased ubiquitination activity and altered cellular localization in several variants found in our cohort compared to the wild type. In conclusion, we found that variants in UBR5 cause a neurodevelopmental syndrome that can be associated with a movement disorder, reinforcing the role of the UBR protein family in a neurodevelopmental disease that differs from previously described ubiquitin-ligase-related syndromes. We also provide evidence for the pathogenic potential loss of UBR5 function with functional experiments in C. elegans and in vitro ubiquitination assays.
Collapse
Affiliation(s)
- Pascale Sabeh
- Department of Genetics, CHU Sainte-Justine, Montréal, QC, Canada
| | - Samantha A Dumas
- Department of Anatomy, Physiology, and Genetics, Uniformed Services University of the Health Sciences, F. Edward Herbert School of Medicine, Bethesda, MD 20814, USA
| | - Claudia Maios
- Department of Neuroscience, Université de Montréal, CRCHUM, Montréal, QC, Canada
| | - Hiba Daghar
- Department of Neuroscience, Université de Montréal, CRCHUM, Montréal, QC, Canada
| | - Marek Korzeniowski
- Department of Anatomy, Physiology, and Genetics, Uniformed Services University of the Health Sciences, F. Edward Herbert School of Medicine, Bethesda, MD 20814, USA
| | - Justine Rousseau
- Department of Genetics, CHU Sainte-Justine, Montréal, QC, Canada
| | - Matthew Lines
- Department of Clinical and Metabolic Genetics, Alberta Children's Hospital, Calgary, AB, Canada
| | - Andrea Guerin
- Division of Medical Genetics, Department of Pediatrics, Queen's University, Kingston, ON, Canada
| | - John J Millichap
- Department of Neurology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Megan Landsverk
- Sanford Research, Pediatrics and Rare Diseases Group, University of South Dakota Sanford School of Medicine, Sioux Falls, SD, USA
| | - Theresa Grebe
- Department of Genetics and Metabolism, Phoenix Children Hospital, Phoenix, AZ, USA
| | - Kristin Lindstrom
- BioMarin Pharmaceutical, Inc., 105 Digital Dr., Novato, CA 94949, USA
| | - Jonathan Strober
- Department of Child Neurology, University of California, San Francisco, San Francisco, CA, USA
| | - Tarik Ait Mouhoub
- Department of Genetic, University Hospital Center of Reims, Reims, France
| | - Christiane Zweier
- Department of Human Genetics, Inselspital Bern, University of Bern, Bern, Switzerland
| | - Michelle Steinraths
- Department of Medical Genetics, University of British Columbia, Vancouver, BC, Canada
| | - Moritz Hebebrand
- Institute of Human Genetics, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Bert Callewaert
- Center for Medical Genetics, Ghent University Hospital, Ghent, Belgium
| | - Rami Abou Jamra
- Institute of Human Genetics, University of Leipzig Medical Center, Leipzig, Germany
| | - Monika Kautza-Lucht
- Institute of Human Genetics, University Hospital of Schleswig-Holstein, Kiel, Germany
| | - Meret Wegler
- Institute of Human Genetics, University of Leipzig Medical Center, Leipzig, Germany
| | | | - Candy Kumps
- Center for Medical Genetics, Ghent University Hospital, Ghent, Belgium
| | - Ehud Banne
- The Genetic Institute, Wolfson Medical Center, Holon, Israel
| | | | - Anne Dieux
- Service de génétique clinique du CHU de Lille, Lille, France
| | - Sarah Raible
- Division of Human Genetics, The Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Ian Krantz
- Division of Human Genetics, The Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Livija Medne
- Division of Genomic Diagnostics, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Kieran Pechter
- Division of Genomic Diagnostics, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Laurent Villard
- Aix Marseille University, INSERM, Marseille Medical Genetics Center, MMG, Marseille, France; University of Florence, Florence, Italy
| | - Renzo Guerrini
- Neuroscience and Human Genetics Department, Meyer Children's Hospital IRCCS, Florence, Italy; University of Florence, Florence, Italy
| | - Claudia Bianchini
- Neuroscience and Human Genetics Department, Meyer Children's Hospital IRCCS, Florence, Italy; University of Florence, Florence, Italy
| | - Carmen Barba
- Neuroscience and Human Genetics Department, Meyer Children's Hospital IRCCS, Florence, Italy; University of Florence, Florence, Italy
| | - Davide Mei
- Neuroscience and Human Genetics Department, Meyer Children's Hospital IRCCS, Florence, Italy
| | - Xavier Blanc
- Medigenome, Swiss Institute of Genomic Medicine, Geneva, Switzerland
| | | | - Emmanuelle Ranza
- Medigenome, Swiss Institute of Genomic Medicine, Geneva, Switzerland
| | - Xiao-Ru Yang
- Department of Medical Genetics, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Emily O'Heir
- Center for Mendelian Genomics, Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Kirsten A Donald
- Department of Paediatrics and Child Health, Red Cross War Memorial Children's Hospital, University of Cape Town, Rondebosch, South Africa
| | - Serini Murugasen
- Department of Paediatrics and Child Health, Red Cross War Memorial Children's Hospital, University of Cape Town, Rondebosch, South Africa
| | - Zandre Bruwer
- Department of Paediatrics and Child Health, Red Cross War Memorial Children's Hospital, University of Cape Town, Rondebosch, South Africa
| | - Muge Calikoglu
- University of North Carolina, Department of Pediatrics, Division of Genetics and Metabolism, Chapel Hill, NC, USA
| | - Jennifer M Mathews
- University of North Carolina, Department of Pediatrics, Division of Genetics and Metabolism, Chapel Hill, NC, USA
| | | | - Geneviève Baujat
- University of North Carolina, Department of Pediatrics, Division of Genetics and Metabolism, Chapel Hill, NC, USA
| | - Nicolas Derive
- Laboratoire de Biologie Médicale Multi-Sites SeqOIA, Paris, France
| | - Tyler Mark Pierson
- Departments of Pediatrics and of Neurology, Guerin Children's Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA; Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Jill R Murrell
- Department of Pathology and Laboratory Medicine, Children's Hospital of the University of Pennsylvania, Philadelphia, PA, USA
| | - Amelle Shillington
- Division of Human Genetics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Clothilde Ormieres
- Service de médecine génomique des maladies rares, Necker Hospital, Paris, France
| | - Sophie Rondeau
- Service de médecine génomique des maladies rares, Necker Hospital, Paris, France
| | - André Reis
- Institute of Human Genetics, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | | | - Ping Yee Billie Au
- Department of Medical Genetics, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - David A Sweetser
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Lauren C Briere
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Nathalie Couque
- Département de Génétique - UF de Génétique Moléculaire Hôpital Robert Debré, Paris, France
| | - Laurence Perrin
- Département de Génétique - UF de Génétique Moléculaire Hôpital Robert Debré, Paris, France
| | | | - Paul Gueguen
- Laboratoire de Biologie Médicale Multi-Sites SeqOIA, Paris, France
| | - Mathilde Lefebvre
- UF de génétique clinique, Centre Hospitalier Régional d'Orléans, Orléans, France
| | | | | | | | - J Alex Parker
- Department of Neuroscience, Université de Montréal, CRCHUM, Montréal, QC, Canada
| | - Barrington G Burnett
- Department of Anatomy, Physiology, and Genetics, Uniformed Services University of the Health Sciences, F. Edward Herbert School of Medicine, Bethesda, MD 20814, USA.
| | | |
Collapse
|
17
|
Feng X, Wang Z, Cen M, Zheng Z, Wang B, Zhao Z, Zhong Z, Zou Y, Lv Q, Li S, Huang L, Huang H, Qiu X. Deciphering potential molecular mechanisms in clear cell renal cell carcinoma based on the ubiquitin-conjugating enzyme E2 related genes: Identifying UBE2C correlates to infiltration of regulatory T cells. Biofactors 2025; 51:e2143. [PMID: 39614426 DOI: 10.1002/biof.2143] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Accepted: 10/28/2024] [Indexed: 12/01/2024]
Abstract
Renal clear cell carcinoma (ccRCC) is a highly aggressive and common form of kidney cancer, with limited treatment options for advanced stages. Recent studies have highlighted the importance of the ubiquitin-proteasome system in tumor progression, particularly the role of ubiquitin-conjugating enzyme E2 (UBE2) family members. However, the prognostic significance of UBE2-related genes (UBE2RGs) in ccRCC remains unclear. In this study, bulk RNA-sequencing and single-cell RNA-sequencing data from ccRCC patients were retrieved from the Cancer Genome Atlas and Gene Expression Omnibus databases. Differential expression analysis was performed to identify UBE2RGs associated with ccRCC. A combination of 10 machine learning methods was applied to develop an optimal prognostic model, and its predictive performance was evaluated using area under the curve (AUC) values for 1-, 3-, and 5-year overall survival (OS) in both training and validation cohorts. Functional enrichment analyses of gene ontology and Kyoto Encyclopedia of Genes and Genomes were conducted to explore the biological pathways involved. Correlation analysis was conducted to investigate the association between the risk score and tumor mutational burden (TMB) and immune cell infiltration. Immunotherapy and chemotherapy sensitivity were assessed by immunophenoscore and tumor immune, dysfunction, and exclusion scores to identify potential predictive significance. In vitro, knockdown of the key gene UBE2C in 786-O cells by specific small interfering RNA to validate its impact on apoptosis, migration, cell cycle, migration, invasion of tumor cells, and induction of regulatory T cells (Tregs). Analysis of sc-RNA revealed that UBE2 activity was significantly upregulated in malignant cells, suggesting its role in tumor progression. A three-gene prognostic model comprising UBE2C, UBE2D3, and UBE2T was constructed by Lasoo Cox regression and demonstrated robust predictive accuracy, with AUC values of 0.745, 0.766, and 0.771 for 1-, 3-, and 5-year survival, respectively. The model was validated as an independent prognostic factor in ccRCC. Patients in the high-risk group had a worse prognosis, higher TMB scores, and low responsiveness to immunotherapy. Additionally, immune infiltration and chemotherapy sensitivity analyses revealed that UBE2RGs are associated with various immune cells and drugs, suggesting that UBE2RGs could be a potential therapeutic target for ccRCC. In vitro experiments confirmed that the reduction of UBE2C led to an increase in apoptosis rate, as well as a decrease in tumor cell invasion and metastasis abilities. Additionally, si-UBE2C cells reduced the release of the cytokine Transforming Growth Factor-beta 1 (TGF-β1), leading to a decreased ratio of Tregs in the co-culture system. This study presents a novel three-gene prognostic model based on UBE2RGs that demonstrates significant predictive value for OS, immunotherapy, and chemotherapy in ccRCC patients. The findings underscore the potential of UBE2 family members as biomarkers and therapeutic targets in ccRCC, warranting further investigation in prospective clinical trials.
Collapse
Affiliation(s)
- Xiaoqiang Feng
- Center of Stem Cell and Regenerative Medicine, Gaozhou People's Hospital, Gaozhou, Guangdong, China
| | - Zhenwei Wang
- Department of Urology, The Affiliated Guangdong Second Provincial General Hospital of Jinan University, Guangzhou, Guangdong, China
- Guangdong Second Provincial General Hospital, Guangzhou, Guangdong, China
| | - Meini Cen
- Department of Rehabilitation Medicine, The Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, China
| | - Zongtai Zheng
- Department of Urology, The Affiliated Guangdong Second Provincial General Hospital of Jinan University, Guangzhou, Guangdong, China
- Guangdong Second Provincial General Hospital, Guangzhou, Guangdong, China
| | - Bangqi Wang
- Department of Urology, The Affiliated Guangdong Second Provincial General Hospital of Jinan University, Guangzhou, Guangdong, China
- Guangdong Second Provincial General Hospital, Guangzhou, Guangdong, China
| | - Zongxiang Zhao
- Department of Urology, The Affiliated Guangdong Second Provincial General Hospital of Jinan University, Guangzhou, Guangdong, China
- Guangdong Second Provincial General Hospital, Guangzhou, Guangdong, China
| | - Zhihui Zhong
- Center of Stem Cell and Regenerative Medicine, Gaozhou People's Hospital, Gaozhou, Guangdong, China
| | - Yesong Zou
- Department of Urology, The Affiliated Guangdong Second Provincial General Hospital of Jinan University, Guangzhou, Guangdong, China
- Guangdong Second Provincial General Hospital, Guangzhou, Guangdong, China
| | - Qian Lv
- Department of Urology, The Affiliated Guangdong Second Provincial General Hospital of Jinan University, Guangzhou, Guangdong, China
- Guangdong Second Provincial General Hospital, Guangzhou, Guangdong, China
| | - Shiyu Li
- Department of Microbiology and Immunology, Institute of Geriatric Immunology, School of Medicine, Jinan University, Guangzhou, Guangdong, China
| | - Li Huang
- Center of Stem Cell and Regenerative Medicine, Gaozhou People's Hospital, Gaozhou, Guangdong, China
| | - Hai Huang
- Department of Urology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Department of Urology, Ruijin Hospital Lu Wan Branch, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiaofu Qiu
- Department of Urology, The Affiliated Guangdong Second Provincial General Hospital of Jinan University, Guangzhou, Guangdong, China
- Guangdong Second Provincial General Hospital, Guangzhou, Guangdong, China
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, Guangdong, China
| |
Collapse
|
18
|
An Y, Guo C, Wang X, Liu J, Li Z, Ding J, Zhang Q, Zhou H, Xia B, Wang J, Yu Y, Wan C, Wang J, Dai J. USP18 Promotes Cholesterol Efflux and Mitigates Atherosclerosis by Deubiquitinating ABCG1. J Cell Mol Med 2025; 29:e70320. [PMID: 39804798 PMCID: PMC11728483 DOI: 10.1111/jcmm.70320] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Revised: 12/11/2024] [Accepted: 12/13/2024] [Indexed: 01/16/2025] Open
Abstract
Deubiquitinating enzymes (DUBs) are integral regulators of protein stability. Among these, Ubiquitin-specific protease 18 (USP18) has emerged as a potential therapeutic target for heart failure. However, its precise role in atherosclerosis remains to be comprehensively understood. This study endeavours to examine the impact of USP18 on atherosclerosis and elucidate its corresponding molecular mechanisms. Our studies indicate an elevated expression of USP18 in human coronary atherosclerotic plaques. Notably, the knockdown of USP18 significantly exacerbated lipid accumulation in macrophages. This knockdown effect impaired cholesterol efflux and influenced the downregulation of ATP-binding cassette transporter G1 (ABCG1) expression, achieved by altering the ubiquitination level of ABCG1. Comprehensive mechanistic studies unveiled that USP18 directly affiliates with ABCG1, reducing its ubiquitination and consequently bolstering ABCG1 stability within macrophages. Furthermore, in vivo studies elucidated that the knockdown of USP18 notably elevated atherosclerotic lesions and diminished ABCG1 levels in the plaques of Apoe-/- mice. In summary, our results suggested that USP18 plays a crucial role in managing the progression of atherosclerosis by strengthening the expression of ABCG1 protein through its deubiquitinating effect on ABCG1.
Collapse
Affiliation(s)
- Yang An
- School of Forensic MedicineGuizhou Medical UniversityGuiyangChina
| | - Chuxian Guo
- Department of CardiologyThe First Affiliated Hospital of Jinan UniversityGuangzhouChina
| | - Xiaoli Wang
- School of Forensic MedicineGuizhou Medical UniversityGuiyangChina
| | - Jiangjin Liu
- School of Forensic MedicineGuizhou Medical UniversityGuiyangChina
| | - Zhu Li
- School of Forensic MedicineGuizhou Medical UniversityGuiyangChina
| | - Jiuyang Ding
- School of Forensic MedicineGuizhou Medical UniversityGuiyangChina
| | - Qiaojun Zhang
- School of Forensic MedicineGuizhou Medical UniversityGuiyangChina
| | - Hongmei Zhou
- School of Forensic MedicineGuizhou Medical UniversityGuiyangChina
| | - Bing Xia
- School of Forensic MedicineGuizhou Medical UniversityGuiyangChina
| | - Jiawen Wang
- School of Forensic MedicineGuizhou Medical UniversityGuiyangChina
| | - Yanni Yu
- School of Forensic MedicineGuizhou Medical UniversityGuiyangChina
| | - Changwu Wan
- School of Forensic MedicineGuizhou Medical UniversityGuiyangChina
| | - Jie Wang
- School of Forensic MedicineGuizhou Medical UniversityGuiyangChina
| | - Jialin Dai
- School of Forensic MedicineGuizhou Medical UniversityGuiyangChina
| |
Collapse
|
19
|
Shinno K, Miura Y, Iijima KM, Suzuki E, Ando K. Axonal distribution of mitochondria maintains neuronal autophagy during aging via eIF2β. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.20.576435. [PMID: 38293064 PMCID: PMC10827206 DOI: 10.1101/2024.01.20.576435] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/01/2024]
Abstract
Neuronal aging and neurodegenerative diseases are accompanied by proteostasis collapse, while cellular factors that trigger it are not identified. Impaired mitochondrial transport in the axon is another feature of aging and neurodegenerative diseases. Using Drosophila, we found that genetic depletion of axonal mitochondria causes dysregulation of protein degradation. Axons with mitochondrial depletion showed abnormal protein accumulation and autophagic defects. Lowering neuronal ATP levels by blocking glycolysis did not reduce autophagy, suggesting that autophagic defects are associated with mitochondrial distribution. We found that eIF2β was increased by the depletion of axonal mitochondria via proteome analysis. Phosphorylation of eIF2α, another subunit of eIF2, was lowered, and global translation was suppressed. Neuronal overexpression of eIF2β phenocopied the autophagic defects and neuronal dysfunctions, and lowering eIF2β expression rescued those perturbations caused by depletion of axonal mitochondria. These results indicate the mitochondria-eIF2β axis maintains proteostasis in the axon, of which disruption may underly the onset and progression of age-related neurodegenerative diseases.
Collapse
Affiliation(s)
- Kanako Shinno
- Department of Biological Sciences, Graduate School of Science, Tokyo Metropolitan University, Hachioji, Tokyo, 192-0397, Japan
| | - Yuri Miura
- Research Team for Mechanism of Aging, Tokyo Metropolitan Institute for Geriatrics and Gerontology, Itabashi, Tokyo, 173-0015, Japan
| | - Koichi M. Iijima
- Department of Alzheimer’s Disease Research, National Center for Geriatrics and Gerontology, Obu, Aichi, 474-8511, Japan
- Department of Experimental Gerontology, Graduate School of Pharmaceutical Sciences, Nagoya City University, Nagoya, Aichi, 467-8603, Japan
| | - Emiko Suzuki
- Department of Biological Sciences, Graduate School of Science, Tokyo Metropolitan University, Hachioji, Tokyo, 192-0397, Japan
- Gene Network Laboratory, National Institute of Genetics and Department of Genetics, SOKENDAI, Mishima, Shizuoka, 411-8540, Japan
| | - Kanae Ando
- Department of Biological Sciences, Graduate School of Science, Tokyo Metropolitan University, Hachioji, Tokyo, 192-0397, Japan
- Department of Biological Sciences, School of Science, Tokyo Metropolitan University, Hachioji, Tokyo, 192-0397, Japan
| |
Collapse
|
20
|
Sirera J, Sarlak S, Teisseire M, Carminati A, Nicolini VJ, Savy C, Brest P, Juel T, Bontoux C, Deckert M, Ohanna M, Giuliano S, Dufies M, Pages G, Luciano F. Disrupting USP39 deubiquitinase function impairs the survival and migration of multiple myeloma cells through ZEB1 degradation. J Exp Clin Cancer Res 2024; 43:335. [PMID: 39736693 DOI: 10.1186/s13046-024-03241-2] [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: 07/12/2024] [Accepted: 11/26/2024] [Indexed: 01/01/2025] Open
Abstract
BACKGROUND Multiple Myeloma (MM) is the second most common hematological malignancy, characterized by the accumulation of monoclonal plasmocytes in the bone marrow. Despite advancements with proteasome inhibitors, immunomodulatory agents, and CD38-targeting antibodies, MM remains largely incurable due to resistant clones and frequent relapses. The success of the proteasome inhibitor bortezomib (BTZ) in MM treatment highlights the critical role of the ubiquitin-proteasome system (UPS) in this disease. Deubiquitinases (DUBs), which regulate protein stability, interactions, and localization by removing ubiquitin modifications, have emerged as promising therapeutic targets in various cancers, including MM. METHODS Through a comprehensive loss-of-function screen, we identified USP39 as a critical survival factor for MM cells. Gene Set Enrichment Analysis (GSEA) was employed to correlate USP39 mRNA levels with clinical outcomes in MM patients. USP39 protein expression was evaluated via immunohistochemistry (IHC) on bone marrow samples from MM patients and healthy controls. The impact of USP39 knockdown via SiRNA was assessed through in vitro assays measuring cellular metabolism, clonogenic capacity, cell cycle progression, apoptosis, and sensitivity to BTZ. Co-immunoprecipitation and deubiquitination assays were conducted to elucidate the interaction and regulation of ZEB1 by USP39. Finally, in vitro and in vivo zebrafish experiments were used to characterize the biological consequences of ZEB1 regulation by USP39. RESULTS Our study found that elevated USP39 mRNA levels are directly associated with shorter survival in MM patients. USP39 protein expression is significantly higher in MM patient plasmocytes compared to healthy individuals. USP39 knockdown inhibits clonogenic capacity, induces cell cycle arrest, triggers apoptosis, and overcomes BTZ resistance. Gain-of-function assays revealed that USP39 stabilizes the transcription factor ZEB1, enhancing the proliferation and the trans-migratory potential of MM cells. CONCLUSIONS Our findings highlight the critical role of the deubiquitinase USP39, suggesting that the USP39/ZEB1 axis could serve as a potential diagnostic marker and therapeutic target in MM.
Collapse
Affiliation(s)
- Jessy Sirera
- Institute for Research On Cancer and Aging of Nice (IRCAN), CNRS UMR 7284, INSERM U1081, University Côte d'Azur, Nice, France
| | - Saharnaz Sarlak
- Institute for Research On Cancer and Aging of Nice (IRCAN), CNRS UMR 7284, INSERM U1081, University Côte d'Azur, Nice, France
| | - Manon Teisseire
- Institute for Research On Cancer and Aging of Nice (IRCAN), CNRS UMR 7284, INSERM U1081, University Côte d'Azur, Nice, France
| | - Alexandrine Carminati
- Centre Méditerranéen de Médecine Moléculaire (C3M), INSERM, University Côte d'Azur, Nice, France
| | - Victoria J Nicolini
- Institute for Research On Cancer and Aging of Nice (IRCAN), CNRS UMR 7284, INSERM U1081, University Côte d'Azur, Nice, France
| | - Coline Savy
- Centre Méditerranéen de Médecine Moléculaire (C3M), INSERM, University Côte d'Azur, Nice, France
| | - Patrick Brest
- Institute for Research On Cancer and Aging of Nice (IRCAN), CNRS UMR 7284, INSERM U1081, University Côte d'Azur, Nice, France
| | - Thierry Juel
- Institute for Research On Cancer and Aging of Nice (IRCAN), CNRS UMR 7284, INSERM U1081, University Côte d'Azur, Nice, France
| | - Christophe Bontoux
- Laboratory of Clinical and Experimental Pathology, University Côte d'Azur, Pasteur Hospital, Hospital-integrated Biobank (BB-0033-00025), FHU OncoAge, IHU RespirERA, Centre Hospitalier Universitaire de Nice, Nice, 06001, France
- Department of Pathology, University Hospital of Toulouse, Cancer Biobank, Cancer University Institute of Toulouse-Oncopole, Toulouse, 31059, France
| | - Marcel Deckert
- Centre Méditerranéen de Médecine Moléculaire (C3M), INSERM, University Côte d'Azur, Nice, France
| | - Mickael Ohanna
- Centre Méditerranéen de Médecine Moléculaire (C3M), INSERM, University Côte d'Azur, Nice, France
| | - Sandy Giuliano
- Institute for Research On Cancer and Aging of Nice (IRCAN), CNRS UMR 7284, INSERM U1081, University Côte d'Azur, Nice, France
| | - Maeva Dufies
- Institute for Research On Cancer and Aging of Nice (IRCAN), CNRS UMR 7284, INSERM U1081, University Côte d'Azur, Nice, France
| | - Gilles Pages
- Institute for Research On Cancer and Aging of Nice (IRCAN), CNRS UMR 7284, INSERM U1081, University Côte d'Azur, Nice, France
| | - Frederic Luciano
- Institute for Research On Cancer and Aging of Nice (IRCAN), CNRS UMR 7284, INSERM U1081, University Côte d'Azur, Nice, France.
| |
Collapse
|
21
|
Zang N, Li X, Zhang Z, Liu W, Qi L, Yang Y, Sun Q, Yin Z, Wang A. Transcription factors PuNAC37/PuWRKY74 and E3 ubiquitin ligase PuRDUF2 inhibit volatile ester synthesis in 'Nanguo' pear. PLANT PHYSIOLOGY 2024; 197:kiae635. [PMID: 39607735 DOI: 10.1093/plphys/kiae635] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Revised: 10/31/2024] [Accepted: 11/04/2024] [Indexed: 11/29/2024]
Abstract
As major contributors to flavor in many fruit species, volatile esters are useful for investigating the regulation of the biosynthesis pathways of volatile aroma compounds in plants. Ethylene promotes the biosynthesis of volatile esters during fruit storage while accelerating fruit ripening; thus, the ethylene perception inhibitor 1-methylcyclopropene (1-MCP) is employed to prolong the shelf life of fruits. However, the mechanisms by which 1-MCP regulates volatiles synthesis remain unclear. In this study, we analyzed the pathway of 1-MCP-mediated volatile ester synthesis in 'Nanguo' pear (Pyrus ussuriensis). 1-MCP significantly decreased volatile ester synthesis during storage. Comparative transcriptome analysis showed that the genes encoding two transcription factors (PuNAC37 and PuWRKY74) and a RING-type E3 ubiquitin ligase that have a domain of unknown function (PuRDUF2) were expressed at high levels, whereas ALCOHOL ACYLTRANSFERASE 1 (PuAAT1), encoding an enzyme responsible for volatile ester synthesis, was expressed at low levels in 1-MCP-treated fruit. Moreover, PuNAC37 inhibited the expression of PuWRKY74 via transcriptional regulation, whereas PuNAC37 and PuWRKY74, after directly binding to the promoter of PuAAT1, synergistically inhibited its expression in 1-MCP-treated fruit. In addition, in vitro and in vivo ubiquitination experiments revealed that PuRDUF2 functions as an E3 ubiquitin ligase that ubiquitinates PuAAT1, causing its degradation via the 26S proteasome pathway following 1-MCP treatment. Subsequent PuAAT1 degradation resulted in a reduction of volatile esters during fruit storage. Our findings provide insights into the complex transcriptional regulation of volatile ester formation in 'Nanguo' pears and contribute to future research on AAT protein ubiquitination in other species.
Collapse
Affiliation(s)
- Nannan Zang
- Key Laboratory of Fruit Postharvest Biology of Liaoning Province, College of Horticulture, Shenyang Agricultural University, Shenyang 110866, China
- Key Laboratory of Protected Horticulture, National & Local Joint Engineering Research Center of Northern Horticultural Facilities Design & Application Technology, Shenyang 110866, China
- College of Horticulture, Shenyang Agricultural University, Shenyang 110866, China
| | - Xiaojing Li
- Key Laboratory of Fruit Postharvest Biology of Liaoning Province, College of Horticulture, Shenyang Agricultural University, Shenyang 110866, China
- Key Laboratory of Protected Horticulture, National & Local Joint Engineering Research Center of Northern Horticultural Facilities Design & Application Technology, Shenyang 110866, China
- College of Horticulture, Shenyang Agricultural University, Shenyang 110866, China
- Research Institute of Pomology, Chinese Academy of Agricultural Sciences, Xinghai South Street 98, Xingcheng 125100, China
| | - Zhuoran Zhang
- Key Laboratory of Fruit Postharvest Biology of Liaoning Province, College of Horticulture, Shenyang Agricultural University, Shenyang 110866, China
- Key Laboratory of Protected Horticulture, National & Local Joint Engineering Research Center of Northern Horticultural Facilities Design & Application Technology, Shenyang 110866, China
- College of Horticulture, Shenyang Agricultural University, Shenyang 110866, China
| | - Weiting Liu
- Key Laboratory of Fruit Postharvest Biology of Liaoning Province, College of Horticulture, Shenyang Agricultural University, Shenyang 110866, China
- Key Laboratory of Protected Horticulture, National & Local Joint Engineering Research Center of Northern Horticultural Facilities Design & Application Technology, Shenyang 110866, China
- College of Horticulture, Shenyang Agricultural University, Shenyang 110866, China
| | - Liyong Qi
- Key Laboratory of Fruit Postharvest Biology of Liaoning Province, College of Horticulture, Shenyang Agricultural University, Shenyang 110866, China
- Key Laboratory of Protected Horticulture, National & Local Joint Engineering Research Center of Northern Horticultural Facilities Design & Application Technology, Shenyang 110866, China
- College of Horticulture, Shenyang Agricultural University, Shenyang 110866, China
- Research Institute of Pomology, Chinese Academy of Agricultural Sciences, Xinghai South Street 98, Xingcheng 125100, China
| | - Yueming Yang
- Key Laboratory of Fruit Postharvest Biology of Liaoning Province, College of Horticulture, Shenyang Agricultural University, Shenyang 110866, China
- Key Laboratory of Protected Horticulture, National & Local Joint Engineering Research Center of Northern Horticultural Facilities Design & Application Technology, Shenyang 110866, China
- College of Horticulture, Shenyang Agricultural University, Shenyang 110866, China
| | - Qitian Sun
- Key Laboratory of Fruit Postharvest Biology of Liaoning Province, College of Horticulture, Shenyang Agricultural University, Shenyang 110866, China
- Key Laboratory of Protected Horticulture, National & Local Joint Engineering Research Center of Northern Horticultural Facilities Design & Application Technology, Shenyang 110866, China
- College of Horticulture, Shenyang Agricultural University, Shenyang 110866, China
| | - Zepeng Yin
- Key Laboratory of Fruit Postharvest Biology of Liaoning Province, College of Horticulture, Shenyang Agricultural University, Shenyang 110866, China
- Key Laboratory of Protected Horticulture, National & Local Joint Engineering Research Center of Northern Horticultural Facilities Design & Application Technology, Shenyang 110866, China
- College of Horticulture, Shenyang Agricultural University, Shenyang 110866, China
| | - Aide Wang
- Key Laboratory of Fruit Postharvest Biology of Liaoning Province, College of Horticulture, Shenyang Agricultural University, Shenyang 110866, China
- Key Laboratory of Protected Horticulture, National & Local Joint Engineering Research Center of Northern Horticultural Facilities Design & Application Technology, Shenyang 110866, China
- College of Horticulture, Shenyang Agricultural University, Shenyang 110866, China
| |
Collapse
|
22
|
Wu W, Yang J, Yu T, Zou Z, Huang X. The Role and Mechanism of TRIM Proteins in Gastric Cancer. Cells 2024; 13:2107. [PMID: 39768197 PMCID: PMC11674240 DOI: 10.3390/cells13242107] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2024] [Revised: 12/13/2024] [Accepted: 12/17/2024] [Indexed: 01/11/2025] Open
Abstract
Tripartite motif (TRIM) family proteins, distinguished by their N-terminal region that includes a Really Interesting New Gene (RING) domain with E3 ligase activity, two B-box domains, and a coiled-coil region, have been recognized as significant contributors in carcinogenesis, primarily via the ubiquitin-proteasome system (UPS) for degrading proteins. Mechanistically, these proteins modulate a variety of signaling pathways, including Wnt/β-catenin, PI3K/AKT, and TGF-β/Smad, contributing to cellular regulation, and also impact cellular activities through non-signaling mechanisms, including modulation of gene transcription, protein degradation, and stability via protein-protein interactions. Currently, growing evidence indicates that TRIM proteins emerge as potential regulators in gastric cancer, exhibiting both tumor-suppressive and oncogenic roles. Given their critical involvement in cellular processes and the notable challenges of gastric cancer, exploring the specific contributions of TRIM proteins to this disease is necessary. Consequently, this review elucidates the roles and mechanisms of TRIM proteins in gastric cancer, emphasizing their potential as therapeutic targets and prognostic factors.
Collapse
Affiliation(s)
- Wangxi Wu
- The National Engineering Research Center for Bioengineering Drugs and the Technologies, Jiangxi Provincial Key Laboratory of Bioengineering Drugs, Institute of Translational Medicine, Jiangxi Medical College, Nanchang University, Nanchang 330031, China; (W.W.); (T.Y.)
- The Queen Mary School, Jiangxi Medical College, Nanchang University, Nanchang 330031, China; (J.Y.); (Z.Z.)
| | - Jinyu Yang
- The Queen Mary School, Jiangxi Medical College, Nanchang University, Nanchang 330031, China; (J.Y.); (Z.Z.)
| | - Tian Yu
- The National Engineering Research Center for Bioengineering Drugs and the Technologies, Jiangxi Provincial Key Laboratory of Bioengineering Drugs, Institute of Translational Medicine, Jiangxi Medical College, Nanchang University, Nanchang 330031, China; (W.W.); (T.Y.)
| | - Zhuoling Zou
- The Queen Mary School, Jiangxi Medical College, Nanchang University, Nanchang 330031, China; (J.Y.); (Z.Z.)
| | - Xuan Huang
- The National Engineering Research Center for Bioengineering Drugs and the Technologies, Jiangxi Provincial Key Laboratory of Bioengineering Drugs, Institute of Translational Medicine, Jiangxi Medical College, Nanchang University, Nanchang 330031, China; (W.W.); (T.Y.)
- Chongqing Research Institute, Nanchang University, Chongqing 400010, China
| |
Collapse
|
23
|
Park S, Park HW, Seo DB, Yoo DS, Bae S. In vitro hair growth-promoting effects of araliadiol via the p38/PPAR-γ signaling pathway in human hair follicle stem cells and dermal papilla cells. Front Pharmacol 2024; 15:1482898. [PMID: 39691387 PMCID: PMC11649413 DOI: 10.3389/fphar.2024.1482898] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Accepted: 11/18/2024] [Indexed: 12/19/2024] Open
Abstract
Background Scalp hair plays a crucial role in social communication by expressing personal appearance and self-identity. Consequently, hair loss often leads to a perception of unattractiveness, negatively impacting an individual's life and mental health. Currently, the use of Food and Drug Administration (FDA)-approved drugs for hair loss is associated with several side effects, highlighting the need for identifying new drug candidates, such as plant-derived phytochemicals, to overcome these issues. Objective This study investigated the hair growth-promoting effects of araliadiol, a polyacetylene compound found in plants such as Centella asiatica. Methods We employed an in vitro model comprising human hair follicle stem cells (HHFSCs) and human dermal papilla cells (HDPCs) to evaluate the hair growth-promoting effects of araliadiol. The proliferation-stimulating effects of araliadiol were assessed using water-soluble tetrazolium salt assay, adenosine triphosphate content assay, and crystal violet staining assay. In addition, we performed luciferase reporter assay, polymerase chain reaction analysis, cell fractionation, Western blot analysis, and enzyme-linked immunosorbent assay (ELISA) to elucidate the mechanism underlying the hair growth-inductive effects of araliadiol. Results Araliadiol exhibited both proliferation- and hair growth-promoting effects in HHFSCs and HDPCs. Specifically, it increased the protein expression of cyclin B1 and Ki67. In HHFSCs, it elevated the expression of hair growth-promoting factors, including CD34, vascular endothelial growth factor (VEGF), and angiopoietin-like 4. Similarly, araliadiol increased the expression of hair growth-inductive proteins such as fibroblast growth factor 7, VEGF, noggin, and insulin-like growth factor 1 in HDPCs. Subsequent Western blot analysis and ELISA using inhibitors such as GW9662 and SB202190 confirmed that these hair growth-promoting effects were dependent on the p38/PPAR-γ signaling in both HHFSCs and HDPCs. Conclusion Araliadiol promotes hair growth through the p38/PPAR-γ signaling pathway in human hair follicle cells. Therefore, araliadiol can be considered a novel drug candidate for the treatment of alopecia.
Collapse
Affiliation(s)
- Seokmuk Park
- Department of Cosmetics Engineering, Konkuk University, Seoul, Republic of Korea
| | | | | | | | - Seunghee Bae
- Department of Cosmetics Engineering, Konkuk University, Seoul, Republic of Korea
| |
Collapse
|
24
|
Zhou C, Bao G, Chen Y. TRIM46 accelerates H1N1 influenza virus-induced ferroptosis and inflammatory response by regulating SLC7A11 ubiquitination. J Bioenerg Biomembr 2024; 56:631-643. [PMID: 39531094 DOI: 10.1007/s10863-024-10043-w] [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: 07/06/2024] [Accepted: 10/29/2024] [Indexed: 11/16/2024]
Abstract
Influenza A (H1N1) virus is an acute respiratory infection responsible for enormous morbidity and mortality worldwide. The tripartite motif-containing protein 46 (TRIM46) has an antiviral function that inhibits various viral infections. This study is designed to explore the role and mechanism of TRIM46 in the progress of H1N1 infection. Herein, we infected A549 or 16HBE cells with the H1N1 virus at different times to assess TRIM46 and solute carrier family 7 member 11 (SLC7A11) expression. TRIM46 and Influenza A nucleoprotein mRNA levels were detected by real-time quantitative polymerase chain reaction (RT-qPCR). TRIM46, solute carrier family 7 member 11 (SLC7A11), and Nucleoprotein protein levels were detected using protein level were detected by western blot assay. Cell virulence was determined using Virulence assay (TCID50) assay. Cell viability was determined using Cell Counting Kit-8 (CCK-8) assay. Reactive oxygen species (ROS), intracellular iron content, Malondialdehyde (MDA), and Glutathione (GSH) levels were determined using special assay kits. The stability of SLC7A11 was assessed by Cycloheximide (CHX) assay. Interaction between TRIM46 and SLC7A11 was verified using Co-immunoprecipitation (CoIP) assay. The biological role of TRIM46 was assessed in H1N1 virus-challenged lung injury mice in vivo. TRIM46 level was significantly increased during H1N1 virus infection, and SLC7A11 expression was decreased. TRIM46 downregulation could suppress H1N1 virus replication and relieve H1N1 infection-induced ferroptosis and inflammation in A549 or 16HBE cells. Mechanistically, TRIM46 could promote SLC7A11 ubiquitination and decrease its stability. TRIM46 knockdown repressed H1N1 virus-induced lung injury in vivo. TRIM46 could contribute to influenza A H1N1 virus infection by promoting SLC7A11 ubiquitination in A549 cells, which indicates that targeting TRIM46 may improve the prognosis of patients.
Collapse
Affiliation(s)
- Chao Zhou
- Department of Infection, The People's Hospital of Danyang, Affiliated Danyang Hospital of Nantong University, Danyang, China
| | - Genchong Bao
- Department of Neurology, The People's Hospital of Danyang, Danyang, China
| | - Yanfei Chen
- Department of ICU, Danyang Hospital of Traditional Chinese Medicine, No. 38, Yunyang Road, Danyang, 212300, China.
| |
Collapse
|
25
|
Shi JJ, Chen RY, Liu YJ, Li CY, Yu J, Tu FY, Sheng JX, Lu JF, Zhang LL, Yang GJ, Chen J. Unraveling the role of ubiquitin-conjugating enzyme 5 (UBC5) in disease pathogenesis: A comprehensive review. Cell Signal 2024; 124:111376. [PMID: 39236836 DOI: 10.1016/j.cellsig.2024.111376] [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: 08/01/2024] [Revised: 08/22/2024] [Accepted: 08/30/2024] [Indexed: 09/07/2024]
Abstract
While certain members of ubiquitin-coupled enzymes (E2s) have garnered attention as potential therapeutic targets across diverse diseases, research progress on Ubiquitin-Conjugating Enzyme 5 (UBC5)-a pivotal member of the E2s family involved in crucial cellular processes such as apoptosis, DNA repair, and signal transduction-has been relatively sluggish. Previous findings suggest that UBC5 plays a vital role in the ubiquitination of various target proteins implicated in diseases and homeostasis, particularly in various cancer types. This review comprehensively introduces the structure and biological functions of UBC5, with a specific focus on its contributions to the onset and advancement of diverse diseases. It suggests that targeting UBC5 holds promise as a therapeutic approach for disease therapy. Recent discoveries highlighting the high homology between UBC5, UBC1, and UBC4 have provided insight into the mechanism of UBC5 in protein degradation and the regulation of cellular functions. As our comprehension of the structural distinctions among UBC5 and its homologues, namely UBC1 and UBC4, advances, our understanding of UBC5's functional significance also expands.
Collapse
Affiliation(s)
- Jin-Jin Shi
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, School of Marine Sciences, Ningbo University, Ningbo 315211, China
| | - Ru-Yi Chen
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, School of Marine Sciences, Ningbo University, Ningbo 315211, China
| | - Yan-Jun Liu
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, School of Marine Sciences, Ningbo University, Ningbo 315211, China
| | - Chang-Yun Li
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, School of Marine Sciences, Ningbo University, Ningbo 315211, China
| | - Jing Yu
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, School of Marine Sciences, Ningbo University, Ningbo 315211, China
| | - Fei-Yang Tu
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, School of Marine Sciences, Ningbo University, Ningbo 315211, China
| | - Jian-Xiang Sheng
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, School of Marine Sciences, Ningbo University, Ningbo 315211, China
| | - Jian-Fei Lu
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, School of Marine Sciences, Ningbo University, Ningbo 315211, China
| | - Le-Le Zhang
- School of Basic Medical Sciences, Chengdu University, Chengdu 610106, China.
| | - Guan-Jun Yang
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, School of Marine Sciences, Ningbo University, Ningbo 315211, China.
| | - Jiong Chen
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, School of Marine Sciences, Ningbo University, Ningbo 315211, China.
| |
Collapse
|
26
|
Sogbein O, Paul P, Umar M, Chaari A, Batuman V, Upadhyay R. Bortezomib in cancer therapy: Mechanisms, side effects, and future proteasome inhibitors. Life Sci 2024; 358:123125. [PMID: 39413903 DOI: 10.1016/j.lfs.2024.123125] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2024] [Revised: 06/07/2024] [Accepted: 10/08/2024] [Indexed: 10/18/2024]
Abstract
The ubiquitin-proteasome pathway (UPP) regulates protein stability and normal cellular functions with the help of autocatalytic proteasome complex. Studies have linked aberrant proteasome activity to malignant cells and found that proteasome inhibitors play a significant role as therapeutic drugs for various types of cancer, specifically multiple myeloma and mantle cell lymphoma. Bortezomib, the first FDA-approved proteasome inhibitor for treating different stages of multiple myeloma, acts on cancer cells by inhibiting the 26S proteasome, modulating NF-κB, phosphorylating Bcl-2, upregulating of NOXA, blocking p53 degradation, activating caspase, generating reactive oxygen species (ROS), and inhibiting angiogenesis. However, its efficacy is limited due to side effects such as peripheral neuropathy (PN), thrombotic microangiopathy (TMA), and acute interstitial nephritis (AIN). Therefore, a better understanding of its precise mechanism of action may help mitigate these side effects. In this review, we have discussed the proposed mechanisms of action and off target effects of Bortezomib, along with the prospects of next generation potential proteasome inhibitor drugs in the treatment of cancer.
Collapse
Affiliation(s)
- Olusola Sogbein
- Department of Medicine, Tulane University School of Medicine, New Orleans, LA 70112, USA
| | - Pradipta Paul
- Weill Cornell Medicine-Qatar, Education City, Qatar Foundation, P.O. Box 24144, Qatar
| | - Meenakshi Umar
- Department of Medicine, Tulane University School of Medicine, New Orleans, LA 70112, USA
| | - Ali Chaari
- Weill Cornell Medicine-Qatar, Education City, Qatar Foundation, P.O. Box 24144, Qatar
| | - Vecihi Batuman
- Department of Medicine, Tulane University School of Medicine, New Orleans, LA 70112, USA.
| | - Rohit Upadhyay
- Department of Medicine, Tulane University School of Medicine, New Orleans, LA 70112, USA.
| |
Collapse
|
27
|
Mitra P, Deshmukh AS. Proteostasis is a key driver of the pathogenesis in Apicomplexa. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2024; 1871:119824. [PMID: 39168412 DOI: 10.1016/j.bbamcr.2024.119824] [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: 01/31/2024] [Revised: 08/08/2024] [Accepted: 08/12/2024] [Indexed: 08/23/2024]
Abstract
Proteostasis, including protein folding mediated by molecular chaperones, protein degradation, and stress response pathways in organelles like ER (unfolded protein response: UPR), are responsible for cellular protein quality control. This is essential for cell survival as it regulates and reprograms cellular processes. Here, we underscore the role of the proteostasis pathway in Apicomplexan parasites with respect to their well-characterized roles as well as potential roles in many parasite functions, including survival, multiplication, persistence, and emerging drug resistance. In addition to the diverse physiological importance of proteostasis in Apicomplexa, we assess the potential of the pathway's components as chemotherapeutic targets.
Collapse
Affiliation(s)
- Pallabi Mitra
- BRIC-Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, Kerala, India.
| | | |
Collapse
|
28
|
Ashok A, Ashwathnarayan A, Bhaskar S, Shekar S, Kalathur G, Prasanna J, Kumar A. Inhibition of proteasome activity facilitates definitive endodermal specification of pluripotent stem cells by influencing YAP signalling. Life Sci 2024; 358:123160. [PMID: 39433087 DOI: 10.1016/j.lfs.2024.123160] [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: 05/27/2024] [Revised: 08/03/2024] [Accepted: 10/17/2024] [Indexed: 10/23/2024]
Abstract
AIMS The knowledge of the molecular players that regulate the generation of endoderm cells is imperative to obtain homogenous population of pancreatic β-cells from stem cells. The Ubiquitin proteasome system (UPS) has been envisaged as a crucial intracellular protein degradation system, but its role in the generation of β-cells remains elusive. Hence, it would be appropriate to unravel the potential role of UPS in endoderm specification and utilize the understanding to generate β-cells from pluripotent stem cells. MATERIALS AND METHODS The pluripotent stem cells (mESCs, miPSCs and hIPSCs) were subjected to differentiation towards pancreatic β-cells and assessed the proteasomal activity during endodermal differentiation. Pharmacologic agents MG132 and IU-1 were employed to inhibit and activate proteasomal activity respectively at the definitive endoderm stage to investigate its impact on the generation of β-cells. The expression of stage-specific genes were analyzed at transcript and protein levels. We also explored the role of unfolded protein response and UPS-regulated signalling pathways in endodermal differentiation. KEY FINDINGS We observed decreased proteasomal activity specifically during endoderm, but not during the generation of other lineages. Extraneous proteasomal inhibition enhanced the expression of endodermal genes while increasing the proteasomal activity hindered definitive endodermal differentiation. Proteasomal inhibition at the definitive endodermal stage culminated in an enriched generation of insulin-positive cells. Elevated endodermal gene expression was consistent in mESCs and hIPSCs upon proteasomal inhibition. Mechanistic insight revealed the proteasome-inhibited enhanced endodermal differentiation to be via modulating the YAP pathway. SIGNIFICANCE Our study unravels the specific involvement of UPS in endoderm cell generation from pluripotent stem cells and paves the way for obtaining potential definitive endodermal cells for plausible cellular therapy in the future.
Collapse
Affiliation(s)
- Akshaya Ashok
- Manipal Institute of Regenerative Medicine, Bangalore, Manipal Academy of Higher Education, Manipal, India
| | - Ashwini Ashwathnarayan
- Manipal Institute of Regenerative Medicine, Bangalore, Manipal Academy of Higher Education, Manipal, India
| | - Smitha Bhaskar
- Manipal Institute of Regenerative Medicine, Bangalore, Manipal Academy of Higher Education, Manipal, India
| | - Spandana Shekar
- Manipal Institute of Regenerative Medicine, Bangalore, Manipal Academy of Higher Education, Manipal, India
| | - Guruprasad Kalathur
- Division of Reproductive Biology, Department of Reproductive Science, Kasturba Medical College, Manipal, Manipal Academy of Higher Education, Manipal 576104, India
| | - Jyothi Prasanna
- Manipal Institute of Regenerative Medicine, Bangalore, Manipal Academy of Higher Education, Manipal, India
| | - Anujith Kumar
- Manipal Institute of Regenerative Medicine, Bangalore, Manipal Academy of Higher Education, Manipal, India.
| |
Collapse
|
29
|
Zimmermann A, Reitnauer D, Yundung Y, Menges AL, Meuli L, Pelisek J, Reutersberg B. Different Proteins as Biomarkers for Sac Shrinkage After Endovascular Aortic Repair of Abdominal Aortic Aneurysms. J Cardiovasc Dev Dis 2024; 11:374. [PMID: 39590217 PMCID: PMC11594911 DOI: 10.3390/jcdd11110374] [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: 09/18/2024] [Revised: 11/15/2024] [Accepted: 11/18/2024] [Indexed: 11/28/2024] Open
Abstract
BACKGROUND This study aims to identify circulating biomarkers by using proteomic analysis associated with sac shrinkage or expansion in patients undergoing endovascular aneurysm repair (EVAR) for abdominal aortic aneurysms (AAAs). METHODS Plasma samples were analysed from 32 patients treated with EVAR between 10/2009 and 10/2020. Patients were divided into two groups based on postoperative sac behaviour: sac shrinkage (≥5 mm reduction) and no shrinkage (stabilisation or expansion). Proteomic analysis was performed using high-resolution liquid chromatography-tandem mass spectrometry (LC-MS/MS), with abundant protein depletion to enhance the detection of low-abundant proteins. RESULTS Of the 32 patients, 20 exhibited sac shrinkage, and 12 showed no shrinkage. Proteomic analysis identified 632 proteins, with significant differential abundance observed after adjusting for relevant clinical parameters. Notably, neurogranin (NRGN) levels were significantly associated with hypertension and smoking, while casein alpha S1 (CSN1S1) levels varied with statin use. Differentially abundant proteins related to aortic diameter included calpastatin, SCUBE3, and ubiquitin-conjugating enzyme E2, among others. CONCLUSIONS Proteomic profiling revealed distinct biomarker patterns associated with sac behaviour in EVAR-treated AAA patients. These findings suggest potential therapeutic targets for enhancing EVAR outcomes and underscore the need for further investigation into the biological mechanisms underlying aneurysm sac shrinkage and stability.
Collapse
Affiliation(s)
- Alexander Zimmermann
- Department of Vascular Surgery, University Hospital Zurich, 8091 Zurich, Switzerland; (D.R.); (A.-L.M.); (L.M.); (J.P.); (B.R.)
| | | | | | | | | | | | | |
Collapse
|
30
|
Meng T, He J, Huo Q, Wang Y, Ren Q, Kang Y. Association of Stress Defense System With Fine Particulate Matter Exposure: Mechanism Analysis and Application Prospects. J Appl Toxicol 2024. [PMID: 39538419 DOI: 10.1002/jat.4724] [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: 08/01/2024] [Revised: 10/29/2024] [Accepted: 10/31/2024] [Indexed: 11/16/2024]
Abstract
The association between the stress defense system and exposure to fine particulate matter (PM2.5) is a hot topic in the field of environmental health. PM2.5 pollution is an increasingly serious issue, and its impact on health cannot be ignored. The stress defense system is an important biological mechanism for maintaining cell and internal environment homeostasis, playing a crucial role in PM2.5-induced damage and diseases. The association between PM2.5 exposure and activation of the stress defense system has been reported. Moderate PM2.5 exposure rapidly mobilizes the stress defense system, while excessive PM2.5 exposure may exceed its compensatory and coping abilities, resulting in system imbalance and dysfunction that triggers pathological changes in cells and tissues, thereby increasing the risk of chronic diseases, such as respiratory diseases, cardiovascular diseases, and cancer. This detailed review focuses on the composition, function, and regulatory mechanisms of the antioxidant defense system, autophagy system, ubiquitin-proteasome system, and inflammatory response system, which are all components of the stress defiance system. In particular, the influence of PM2.5 exposure on each of these defense systems and their roles in responding to PM2.5-induced damage was investigated to provide an in-depth understanding of the pathogenesis of PM2.5 exposure, accurately assess potential hazards, and formulate prevention and intervention strategies for health damage caused by PM2.5 exposure.
Collapse
Affiliation(s)
- Tao Meng
- Institute of Brain Science, Datong Key Laboratory of Molecular and Cellular Immunology, Shanxi Datong University, Datong, China
- Doctoral Innovation Station of Shanxi Province, Key Laboratory of TCM Prevention and Treatment of Dementia Disease, The Fifth People's Hospital of Datong, Datong, China
| | - Jing He
- Institute of Brain Science, Datong Key Laboratory of Molecular and Cellular Immunology, Shanxi Datong University, Datong, China
| | - Qianru Huo
- Institute of Brain Science, Datong Key Laboratory of Molecular and Cellular Immunology, Shanxi Datong University, Datong, China
| | - Yajie Wang
- Institute of Brain Science, Datong Key Laboratory of Molecular and Cellular Immunology, Shanxi Datong University, Datong, China
| | - Qingchun Ren
- Institute of Brain Science, Datong Key Laboratory of Molecular and Cellular Immunology, Shanxi Datong University, Datong, China
| | - Yihui Kang
- Institute of Brain Science, Datong Key Laboratory of Molecular and Cellular Immunology, Shanxi Datong University, Datong, China
| |
Collapse
|
31
|
Hesham HM, Dokla EME, Elrazaz EZ, Lasheen DS, Abou El Ella DA. FLT3-PROTACs for combating AML resistance: Analytical overview on chimeric agents developed, challenges, and future perspectives. Eur J Med Chem 2024; 277:116717. [PMID: 39094274 DOI: 10.1016/j.ejmech.2024.116717] [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: 05/16/2024] [Revised: 07/13/2024] [Accepted: 07/24/2024] [Indexed: 08/04/2024]
Abstract
The urgent and unmet medical demand of acute myeloid leukemia (AML) patients has driven the drug discovery process for expansion of the landscape of AML treatment. Despite the several agents developed for treatment of AML, more than 60 % of treated patients undergo relapse again after re-emission, thus, no complete cure for this complex disease has been reached yet. Targeted oncoprotein degradation is a new paradigm that can be employed to solve drug resistance, disease relapse, and treatment failure in complex diseases as AML, the most lethal hematological malignancy. AML is an aggressive blood cancer form and the most common type of acute leukemia, with bad outcomes and a very poor 5-year survival rate. FLT3 mutations occur in about 30 % of AML cases and FLT3-ITD is associated with poor prognosis of this disease. Prevalent FLT3 mutations include internal tandem duplication and point mutations (e.g., D835) in the tyrosine kinase domain, which induce FLT3 kinase activation and result in survival and proliferation of AML cells again. Currently approved FLT3 inhibitors suffer from limited clinical efficacy due to FLT3 reactivation by mutations, therefore, alternative new treatments are highly needed. Proteolysis-targeting chimera (PROTAC) is a bi-functional molecule that consists of a ligand of the protein of interest, FLT3 inhibitor in our case, that is covalently linked to an E3 ubiquitin ligase ligand. Upon FLT3-specific PROTAC binding to FLT3, the PROTAC can recruit E3 for FLT3 ubiquitination, which is subsequently subjected to proteasome-mediated degradation. In this review we tried to address the question if PROTAC technology has succeeded in tackling the disease relapse and treatment failure of AML. Next, we explored the latest FLT3-targeting PROTACs developed in the past few years such as quizartinib-based PROTACs, dovitinib-based PROTACs, gilteritinib-based PROTACs, and others. Then, we followed with a deep analysis of their advantages regarding potency improvement and overcoming AML drug resistance. Finally, we discussed the challenges facing these chimeric molecules with proposed future solutions to circumvent them.
Collapse
Affiliation(s)
- Heba M Hesham
- Pharmaceutical Chemistry Department, Faculty of Pharmacy, Ain Shams University, Abbassia, 11566, Cairo, Egypt
| | - Eman M E Dokla
- Pharmaceutical Chemistry Department, Faculty of Pharmacy, Ain Shams University, Abbassia, 11566, Cairo, Egypt.
| | - Eman Z Elrazaz
- Pharmaceutical Chemistry Department, Faculty of Pharmacy, Ain Shams University, Abbassia, 11566, Cairo, Egypt
| | - Deena S Lasheen
- Pharmaceutical Chemistry Department, Faculty of Pharmacy, Ain Shams University, Abbassia, 11566, Cairo, Egypt
| | - Dalal A Abou El Ella
- Pharmaceutical Chemistry Department, Faculty of Pharmacy, Ain Shams University, Abbassia, 11566, Cairo, Egypt.
| |
Collapse
|
32
|
Bonelli S, Lo Pinto M, Ye Y, Müller SA, Lichtenthaler SF, Scilabra SD. Proteomic Characterization of Ubiquitin Carboxyl-Terminal Hydrolase 19 Deficient Cells Reveals a Role for USP19 in the Secretion of Lysosomal Proteins. Mol Cell Proteomics 2024; 23:100854. [PMID: 39389361 PMCID: PMC11617723 DOI: 10.1016/j.mcpro.2024.100854] [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: 04/08/2024] [Revised: 09/26/2024] [Accepted: 10/04/2024] [Indexed: 10/12/2024] Open
Abstract
Ubiquitin carboxyl-terminal hydrolase 19 (USP19) is a unique deubiquitinase, characterized by multiple variants generated by alternative splicing. Several variants bear a C-terminal transmembrane domain that anchors them to the endoplasmic reticulum. Other than regulating protein stability by preventing proteasome degradation, USP19 has been reported to rescue substrates from endoplasmic reticulum-associated protein degradation in a catalytic-independent manner, promote autophagy, and address proteins to lysosomal degradation via endosomal microautophagy. USP19 has recently emerged as the protein responsible for the unconventional secretion of misfolded proteins including Parkinson's disease-associated protein α-synuclein. Despite mounting evidence that USP19 plays crucial roles in several biological processes, the underlying mechanisms are unclear due to lack of information on the physiological substrates of USP19. Herein, we used high-resolution quantitative proteomics to analyze changes in the secretome and cell proteome induced by the loss of USP19 to identify proteins whose secretion or turnover is regulated by USP19. We found that ablation of USP19 induced significant proteomic alterations both in and out of the cell. Loss of USP19 impaired the release of several lysosomal proteins, including legumain (LGMN) and several cathepsins. In order to understand the underlaying mechanism, we dissected the USP19-regulated secretion of LGMN in several cell types. We found that LGMN was not a deubiquitinase substrate of USP19 and that its USP19-dependent release did not require their direct interaction. LGMN secretion occurred by a mechanism that involved the Golgi apparatus, autophagosome formation, and lysosome function. This mechanism resembled the recently described "lysosomal exocytosis," by which lysosomal hydrolases are secreted, when ubiquitination of p62 is increased in cells lacking deubiquitinases such as USP15 and USP17. In conclusion, our proteomic characterization of USP19 has identified a collection of proteins in the secretome and within the cell that are regulated by USP19, which link USP19 to the secretion of lysosomal proteins, including LGMN.
Collapse
Affiliation(s)
- Simone Bonelli
- Proteomics Group of Ri.MED Foundation, Research Department IRCCS ISMETT (Istituto Mediterraneo per i Trapianti e Terapie ad Alta Specializzazione), Palermo, Italy; Department of Biological, Chemical and Pharmaceutical Sciences and Technologies (STEBICEF), University of Palermo, Palermo, Italy
| | - Margot Lo Pinto
- Proteomics Group of Ri.MED Foundation, Research Department IRCCS ISMETT (Istituto Mediterraneo per i Trapianti e Terapie ad Alta Specializzazione), Palermo, Italy
| | - Yihong Ye
- Laboratory of Molecular Biology, National Institute of Diabetes, Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Stephan A Müller
- Neuroproteomics Department, German Center for Neurodegenerative Diseases (DZNE), Munich, Germany; Neuroproteomics, School of Medicine and Health, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Stefan F Lichtenthaler
- Neuroproteomics Department, German Center for Neurodegenerative Diseases (DZNE), Munich, Germany; Neuroproteomics, School of Medicine and Health, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany; Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Simone Dario Scilabra
- Proteomics Group of Ri.MED Foundation, Research Department IRCCS ISMETT (Istituto Mediterraneo per i Trapianti e Terapie ad Alta Specializzazione), Palermo, Italy.
| |
Collapse
|
33
|
Lee DH, Lee HJ, Yang G, Kim DY, Kim JU, Yook TH, Lee JH, Kim HJ. A novel treatment strategy targeting cellular pathways with natural products to alleviate sarcopenia. Phytother Res 2024; 38:5033-5051. [PMID: 39099170 DOI: 10.1002/ptr.8301] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2023] [Revised: 07/08/2024] [Accepted: 07/12/2024] [Indexed: 08/06/2024]
Abstract
Sarcopenia is a condition marked by a significant reduction in muscle mass and strength, primarily due to the aging process, which critically impacts muscle protein dynamics, metabolic functions, and overall physical functionality. This condition leads to increased body fat and reduced daily activity, contributing to severe health issues and a lower quality of life among the elderly. Recognized in the ICD-10-CM only in 2016, sarcopenia lacks definitive treatment options despite its growing prevalence and substantial social and economic implications. Given the aging global population, addressing sarcopenia has become increasingly relevant and necessary. The primary causes include aging, cachexia, diabetes, and nutritional deficiencies, leading to imbalances in protein synthesis and degradation, mitochondrial dysfunction, and hormonal changes. Exercise remains the most effective intervention, but it is often impractical for individuals with limited mobility, and pharmacological options such as anabolic steroids and myostatin inhibitors are not FDA-approved and are still under investigation. This review is crucial as it examines the potential of natural products as a novel treatment strategy for sarcopenia, targeting multiple mechanisms involved in its pathogenesis. By exploring natural products' multi-targeted effects, this study aims to provide innovative and practical solutions for sarcopenia management. Therefore, this review indicates significant improvements in muscle mass and function with the use of specific natural compounds, suggesting promising alternatives for those unable to engage in regular physical activity.
Collapse
Affiliation(s)
- Da Hee Lee
- College of Korean Medicine, Woosuk University, Jeonju-si, Republic of Korea
| | - Hye Jin Lee
- College of Korean Medicine, Woosuk University, Jeonju-si, Republic of Korea
| | - Gabsik Yang
- College of Korean Medicine, Woosuk University, Jeonju-si, Republic of Korea
| | - Dae Yong Kim
- College of Korean Medicine, Woosuk University, Jeonju-si, Republic of Korea
| | - Jong Uk Kim
- College of Korean Medicine, Woosuk University, Jeonju-si, Republic of Korea
| | - Tae Han Yook
- College of Korean Medicine, Woosuk University, Jeonju-si, Republic of Korea
| | - Jun Ho Lee
- College of Korean Medicine, Woosuk University, Jeonju-si, Republic of Korea
- Da Capo Co., Ltd., Jeonju-si, Republic of Korea
| | - Hong Jun Kim
- College of Korean Medicine, Woosuk University, Jeonju-si, Republic of Korea
| |
Collapse
|
34
|
Bickels Nuri R, Feldmesser E, Fridmann-Sirkis Y, Keren-Shaul H, Nevo R, Minsky A, Reich Z. Acanthamoeba polyphaga de novo transcriptome and its dynamics during Mimivirus infection. Sci Rep 2024; 14:25894. [PMID: 39472705 PMCID: PMC11522460 DOI: 10.1038/s41598-024-76078-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Accepted: 10/10/2024] [Indexed: 11/02/2024] Open
Abstract
Mimivirus bradfordmassiliense (Mimivirus) is a giant virus that infects Acanthamoeba species - opportunistic human pathogens. Long- and short-read sequencing were used to generate a de novo transcriptome of the host and followed the dynamics of both host and virus transcriptomes over the course of infection. The assembled transcriptome of the host included 22,604 transcripts and 13,043 genes, with N50 = 2,372 nucleotides. Functional enrichment analysis revealed major changes in the host transcriptome, namely, enrichment in downregulated genes associated with cytoskeleton homeostasis and DNA replication, repair, and nucleotide synthesis. These modulations, together with those implicated by other enriched processes, indicate cell cycle arrest, which was demonstrated experimentally. We also observed upregulation of host genes associated with transcription, secretory pathways and, as reported here for the first time, peroxisomes and the ubiquitin-proteasome system. In Mimivirus, the early stages of infection were marked by upregulated genes related to DNA replication, transcription, translation, and nucleotide metabolism, and in later stages, enrichment in genes associated with lipid metabolism, carbohydrates, and proteases. Some of the changes observed in the amoebal transcriptome likely point to Mimivirus infection causing dismantling of host cytoskeleton and translocation of endoplasmic reticulum membranes to viral factory areas.
Collapse
Affiliation(s)
- Reut Bickels Nuri
- Departments of Chemical and Structural Biology and Biomolecular Sciences, Weizmann Institute of Science, 7610001, Rehovot, Israel.
| | - Ester Feldmesser
- Bioinformatics Unit, Department of Life Sciences Core Facilities, Weizmann Institute of Science, 7610001, Rehovot, Israel
| | - Yael Fridmann-Sirkis
- Protein Analysis Unit, Department of Life Sciences Core Facilities, Weizmann Institute of Science, 7610001, Rehovot, Israel
| | - Hadas Keren-Shaul
- Genomics unit, Department of Life Sciences Core Facilities- The Nancy & Stephen Grand Israel National Center for Personalized Medicine, Weizmann Institute of Science, 7610001, Rehovot, Israel
| | - Reinat Nevo
- Department of Biomolecular Sciences, Weizmann Institute of Science, 7610001, Rehovot, Israel
| | - Abraham Minsky
- Department of Chemical and Structural biology, Weizmann Institute of Science, 7610001, Rehovot, Israel
| | - Ziv Reich
- Department of Biomolecular Sciences, Weizmann Institute of Science, 7610001, Rehovot, Israel.
| |
Collapse
|
35
|
Shen S, Liu R, Huang J, Sun Y, Tan Q, Luo Q, Liu R. MAT1A activation of glycolysis to promote NSCLC progression depends on stabilizing CCND1. Cell Death Dis 2024; 15:768. [PMID: 39438468 PMCID: PMC11496809 DOI: 10.1038/s41419-024-07113-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Revised: 09/18/2024] [Accepted: 09/25/2024] [Indexed: 10/25/2024]
Abstract
Non-small cell lung cancer (NSCLC) remains a cause for concern as the leading cause of cancer-related death worldwide. Amidst ongoing debates on the role and mechanisms of methionine adenosyltransferase 1A (MAT1A) in cancer, our study sheds light on its significance in NSCLC. Leveraging TCGA database and immunohistochemical staining, we systematically analyzed MAT1A expression in NSCLC, uncovering its marked upregulation. To unravel the functional and mechanistic underpinnings, we implemented stable knockdown of MAT1A in NSCLC cell lines. Our findings converged to demonstrate that suppression of MAT1A expression effectively impeded the proliferation and migratory capabilities of NSCLC cells, while concurrently enhancing apoptosis. Mechanistically, we discovered that MAT1A depletion accelerated the degradation of CCND1, a key cell cycle regulator, through S-phase kinase-associated protein 2 (SKP2)-mediated ubiquitination. Notably, CCND1 emerged as a crucial MAT1A partner, jointly orchestrating glycolytic metabolism in NSCLC cells. This intricate interplay suggests that MAT1A promotes NSCLC progression by safeguarding CCND1 protein stability and activating glycolytic pathways, thereby sustaining tumorigenesis. In summary, our study not only identifies MAT1A as a prognostic marker for poor survival in NSCLC patients but also elucidates its mechanistic contributions to cancer progression. These findings pave the way for the development of targeted therapies aimed at disrupting the deleterious MAT1A-CCND1-glycolysis axis in NSCLC.
Collapse
Affiliation(s)
- Shengping Shen
- Shanghai Lung Tumor Clinical Medicine Center, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, 200030, PR China
| | - Ruili Liu
- Department of Stomatology, Ordos Central Hospital, Ordos, 017000, Inner Mongolia, PR China
| | - Jiazheng Huang
- Shanghai Lung Tumor Clinical Medicine Center, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, 200030, PR China
| | - Yingjia Sun
- Shanghai Lung Tumor Clinical Medicine Center, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, 200030, PR China
| | - Qiang Tan
- Shanghai Lung Tumor Clinical Medicine Center, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, 200030, PR China
| | - Qingquan Luo
- Shanghai Lung Tumor Clinical Medicine Center, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, 200030, PR China
| | - Ruijun Liu
- Shanghai Lung Tumor Clinical Medicine Center, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, 200030, PR China.
| |
Collapse
|
36
|
Yi H, Shi H, Mao W, Yin J, Ma Y, Xu L, Jing L, He M, Zhu X, Lu X, Xiong Q, Tang Y, Hou Q, Song L, Wang L, Li W, Yu H, Chen X, Li J, Wang J. E3 ubiquitin ligase IPI1 controls rice immunity and flowering via both E3 ligase-dependent and -independent pathways. Dev Cell 2024; 59:2719-2730.e4. [PMID: 39025062 DOI: 10.1016/j.devcel.2024.06.014] [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: 09/12/2023] [Revised: 04/19/2024] [Accepted: 06/19/2024] [Indexed: 07/20/2024]
Abstract
Immunity and flowering are energy-consuming processes. However, the mechanism underlying the balance between immunity and flowering remains to be elucidated. Here, we report that the E3 ligase ideal plant architecture 1 interactor 1 (IPI1) controls rice immunity and flowering via two different pathways, one dependent on and another independent of its E3 ligase activity. We found that IPI1, a RING-finger E3 ligase, interacts with another E3 ligase, AvrPiz-t-interacting protein 6 (APIP6), and protects APIP6 from degradation by preventing APIP6's self-ubiquitination. Stabilization of APIP6 by IPI1 requires no IPI1 E3 ligase activity and leads to degradation of APIP6 substrates via the ubiquitin-proteasome system (UPS). Meanwhile, IPI1 directly ubiquitinates OsELF3-1 and OsELF3-2, two homologs of EARLY FLOWERING3 (ELF3), targeting them for degradation via the 26S proteasome. IPI1 knockout plants display early flowering but compromised resistance to rice blast. Thus, IPI1 balances rice immunity and flowering via both E3 ligase-dependent and -independent pathways.
Collapse
Affiliation(s)
- Hong Yi
- State Key Laboratory of Crop Gene Exploration and Utilization in Southwest China, Sichuan Agricultural University, Chengdu 611130, Sichuan, China
| | - Hui Shi
- State Key Laboratory of Crop Gene Exploration and Utilization in Southwest China, Sichuan Agricultural University, Chengdu 611130, Sichuan, China
| | - Wei Mao
- State Key Laboratory of Crop Gene Exploration and Utilization in Southwest China, Sichuan Agricultural University, Chengdu 611130, Sichuan, China
| | - Junjie Yin
- State Key Laboratory of Crop Gene Exploration and Utilization in Southwest China, Sichuan Agricultural University, Chengdu 611130, Sichuan, China
| | - Yanyan Ma
- State Key Laboratory of Crop Gene Exploration and Utilization in Southwest China, Sichuan Agricultural University, Chengdu 611130, Sichuan, China
| | - Li Xu
- State Key Laboratory of Crop Gene Exploration and Utilization in Southwest China, Sichuan Agricultural University, Chengdu 611130, Sichuan, China
| | - Linjie Jing
- State Key Laboratory of Crop Gene Exploration and Utilization in Southwest China, Sichuan Agricultural University, Chengdu 611130, Sichuan, China
| | - Min He
- State Key Laboratory of Crop Gene Exploration and Utilization in Southwest China, Sichuan Agricultural University, Chengdu 611130, Sichuan, China
| | - Xiaobo Zhu
- State Key Laboratory of Crop Gene Exploration and Utilization in Southwest China, Sichuan Agricultural University, Chengdu 611130, Sichuan, China
| | - Xiang Lu
- State Key Laboratory of Crop Gene Exploration and Utilization in Southwest China, Sichuan Agricultural University, Chengdu 611130, Sichuan, China
| | - Qing Xiong
- State Key Laboratory of Crop Gene Exploration and Utilization in Southwest China, Sichuan Agricultural University, Chengdu 611130, Sichuan, China
| | - Yongyan Tang
- State Key Laboratory of Crop Gene Exploration and Utilization in Southwest China, Sichuan Agricultural University, Chengdu 611130, Sichuan, China
| | - Qingqing Hou
- State Key Laboratory of Crop Gene Exploration and Utilization in Southwest China, Sichuan Agricultural University, Chengdu 611130, Sichuan, China
| | - Li Song
- State Key Laboratory of Crop Gene Exploration and Utilization in Southwest China, Sichuan Agricultural University, Chengdu 611130, Sichuan, China
| | - Long Wang
- State Key Laboratory of Crop Gene Exploration and Utilization in Southwest China, Sichuan Agricultural University, Chengdu 611130, Sichuan, China
| | - Weitao Li
- State Key Laboratory of Crop Gene Exploration and Utilization in Southwest China, Sichuan Agricultural University, Chengdu 611130, Sichuan, China; Rice Research Institute, Sichuan Agricultural University, Chengdu 611130, China
| | - Hong Yu
- State Key Laboratory of Plant Genomics and National Center for Plant Gene Research (Beijing), Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
| | - Xuewei Chen
- State Key Laboratory of Crop Gene Exploration and Utilization in Southwest China, Sichuan Agricultural University, Chengdu 611130, Sichuan, China
| | - Jiayang Li
- State Key Laboratory of Plant Genomics and National Center for Plant Gene Research (Beijing), Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China; Yazhou Bay Seed Laboratory, Sanya 572025, Hainan, China
| | - Jing Wang
- State Key Laboratory of Crop Gene Exploration and Utilization in Southwest China, Sichuan Agricultural University, Chengdu 611130, Sichuan, China.
| |
Collapse
|
37
|
Zhang W, Zhang K. Understanding the Biological Basis of Polygenic Risk Scores and Disparities in Prostate Cancer: A Comprehensive Genomic Analysis. Cancer Inform 2024; 23:11769351241276319. [PMID: 39444678 PMCID: PMC11497523 DOI: 10.1177/11769351241276319] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Accepted: 07/14/2024] [Indexed: 10/25/2024] Open
Abstract
Objectives For prostate cancer (PCa), hundreds of risk variants have been identified. It remains unknown whether the polygenic risk score (PRS) that combines the effects of these variants is also a sufficiently informative metric with relevance to the molecular mechanisms of carcinogenesis in prostate. We aimed to understand the biological basis of PRS and racial disparities in the cancer. Methods We performed a comprehensive analysis of the data generated (deposited in) by several genomic and/or transcriptomic projects (databases), including the GTEx, TCGA, 1000 Genomes, GEO and dbGap. PRS was constructed from 260 PCa risk variants that were identified by a recent trans-ancestry meta-analysis and contained in the GTEx dataset. The dosages of risk variants and the multi-ancestry effects on PCa incidence estimated by the meta-analysis were used in calculating individual PRS values. Results The following novel results were obtained from our analyses. (1) In normal prostate samples from healthy European Americans (EAs), the expression levels of 540 genes (termed PRS genes) were associated with the PRS (P < .01). (2) Ubiquitin-proteasome system in high-PRS individuals' prostates was more active than that in low-PRS individuals' prostates. (3) Nine PRS genes play roles in the cancer progression-relevant parts, which are frequently hit by somatic mutations in PCa, of PI3K-Akt/RAS-MAPK/mTOR signaling pathways. (4) The expression profiles of the top significant PRS genes in tumor samples were capable of predicting malignant PCa relapse after prostatectomy. (5) The transcriptomic differences between African American and EA samples were incompatible with the patterns of the aforementioned associations between PRS and gene expression levels. Conclusions This study provided unique insights into the relationship between PRS and the molecular mechanisms of carcinogenesis in prostate. The new findings, alongside the moderate but significant heritability of PCa susceptibility contributed by the risk variants, suggest the aptness and inaptness of PRS for explaining PCa and disparities.
Collapse
Affiliation(s)
- Wensheng Zhang
- Bioinformatics Core of Xavier NIH RCMI Center of Cancer Research, Xavier University of Louisiana, New Orleans, LA, USA
| | - Kun Zhang
- Bioinformatics Core of Xavier NIH RCMI Center of Cancer Research, Xavier University of Louisiana, New Orleans, LA, USA
- Department of Computer Science, Xavier University of Louisiana, New Orleans, LA, USA
| |
Collapse
|
38
|
Krull KK, Ali SA, Krijgsveld J. Enhanced feature matching in single-cell proteomics characterizes IFN-γ response and co-existence of cell states. Nat Commun 2024; 15:8262. [PMID: 39327420 PMCID: PMC11427561 DOI: 10.1038/s41467-024-52605-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: 01/11/2024] [Accepted: 09/17/2024] [Indexed: 09/28/2024] Open
Abstract
Proteome analysis by data-independent acquisition (DIA) has become a powerful approach to obtain deep proteome coverage, and has gained recent traction for label-free analysis of single cells. However, optimal experimental design for DIA-based single-cell proteomics has not been fully explored, and performance metrics of subsequent data analysis tools remain to be evaluated. Therefore, we here formalize and comprehensively evaluate a DIA data analysis strategy that exploits the co-analysis of low-input samples with a so-called matching enhancer (ME) of higher input, to increase sensitivity, proteome coverage, and data completeness. We assess the matching specificity of DIA-ME by a two-proteome model, and demonstrate that false discovery and false transfer are maintained at low levels when using DIA-NN software, while preserving quantification accuracy. We apply DIA-ME to investigate the proteome response of U-2 OS cells to interferon gamma (IFN-γ) in single cells, and recapitulate the time-resolved induction of IFN-γ response proteins as observed in bulk material. Moreover, we uncover co- and anti-correlating patterns of protein expression within the same cell, indicating mutually exclusive protein modules and the co-existence of different cell states. Collectively our data show that DIA-ME is a powerful, scalable, and easy-to-implement strategy for single-cell proteomics.
Collapse
Affiliation(s)
- Karl K Krull
- German Cancer Research Center (DKFZ), Heidelberg, Division of Proteomics of Stem Cells and Cancer, Heidelberg, Germany
- Heidelberg University, Faculty of Biosciences, Heidelberg, Germany
- Heidelberg University, Medical Faculty, Heidelberg, Germany
| | - Syed Azmal Ali
- German Cancer Research Center (DKFZ), Heidelberg, Division of Proteomics of Stem Cells and Cancer, Heidelberg, Germany
| | - Jeroen Krijgsveld
- German Cancer Research Center (DKFZ), Heidelberg, Division of Proteomics of Stem Cells and Cancer, Heidelberg, Germany.
- Heidelberg University, Medical Faculty, Heidelberg, Germany.
| |
Collapse
|
39
|
Hernández-Carralero E, Quinet G, Freire R. ATXN3: a multifunctional protein involved in the polyglutamine disease spinocerebellar ataxia type 3. Expert Rev Mol Med 2024; 26:e19. [PMID: 39320846 PMCID: PMC11440613 DOI: 10.1017/erm.2024.10] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Revised: 01/24/2024] [Accepted: 03/15/2024] [Indexed: 09/26/2024]
Abstract
ATXN3 is a ubiquitin hydrolase (or deubiquitinase, DUB), product of the ATXN3 gene, ubiquitously expressed in various cell types including peripheral and neuronal tissues and involved in several cellular pathways. Importantly, the expansion of the CAG trinucleotides within the ATXN3 gene leads to an expanded polyglutamine domain in the encoded protein, which has been associated with the onset of the spinocerebellar ataxia type 3, also known as Machado-Joseph disease, the most common dominantly inherited ataxia worldwide. ATXN3 has therefore been under intensive investigation for decades. In this review, we summarize the main functions of ATXN3 in proteostasis, DNA repair and transcriptional regulation, as well as the emerging role in regulating chromatin structure. The mentioned molecular functions of ATXN3 are also reviewed in the context of the pathological expanded form of ATXN3.
Collapse
Affiliation(s)
- Esperanza Hernández-Carralero
- Fundación Canaria Instituto de Investigación Sanitaria de Canarias (FIISC), Unidad de Investigación, Hospital Universitario de Canarias, La Laguna, Santa Cruz de Tenerife, Spain
- Instituto de Tecnologías Biomédicas, Centro de Investigaciones Biomédicas de Canarias, Facultad de Medicina, Campus Ciencias de la Salud, Universidad de La Laguna, Santa Cruz de Tenerife, Spain
| | - Grégoire Quinet
- Fundación Canaria Instituto de Investigación Sanitaria de Canarias (FIISC), Unidad de Investigación, Hospital Universitario de Canarias, La Laguna, Santa Cruz de Tenerife, Spain
| | - Raimundo Freire
- Fundación Canaria Instituto de Investigación Sanitaria de Canarias (FIISC), Unidad de Investigación, Hospital Universitario de Canarias, La Laguna, Santa Cruz de Tenerife, Spain
- Instituto de Tecnologías Biomédicas, Centro de Investigaciones Biomédicas de Canarias, Facultad de Medicina, Campus Ciencias de la Salud, Universidad de La Laguna, Santa Cruz de Tenerife, Spain
- Faculty of Health Sciences, Universidad Fernando Pessoa Canarias, Las Palmas de Gran Canaria, Spain
| |
Collapse
|
40
|
Wang A, Shen X, Liang N, Xie Z, Tian Z, Zhang L, Guo J, Wei F, Shi G, Wei X. Integrated cytological and transcriptomic analyses provide new insights into restoration of pollen viability in synthetic allotetraploid Brassica carinata. PLANT CELL REPORTS 2024; 43:234. [PMID: 39292285 DOI: 10.1007/s00299-024-03325-5] [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: 05/02/2024] [Accepted: 09/10/2024] [Indexed: 09/19/2024]
Abstract
KEY MESSAGE Upregulation of genes involved in DNA damage repair and sperm cell differentiation leads to restoration of pollen viability in synthetic allotetraploid B. carinata after chromosome doubling. Apart from the well-known contribution of polyploidy to crop improvement, polyploids can also be induced for other purposes, such as to restore the viability of sterile hybrids. The mechanism related to viability transition between the sterile allodiploid and the fertile allotetraploid after chromosome doubling are not well understood. Here, we synthesised allodiploid B. carinata (2n = 2x = 17) and allotetraploid B. carinata (2n = 4x = 34) as models to investigate the cytological and transcriptomic differences during pollen development. The results showed that after chromosome doubling, the recovery of pollen viability in allotetraploid was mainly reflected in the stabilisation of microtubule spindle morphology, normal meiotic chromosome behaviour, and normal microspore development. Interestingly, the deposition and degradation of synthetic anther tapetum were not affected by polyploidy. Transcription analysis showed that the expression of genes related to DNA repair (DMC1, RAD51, RAD17, SPO11-2), cell cycle differentiation (CYCA1;2, CYCA2;3) and ubiquitination proteasome pathway (UBC4, PIRH2, CDC53) were positively up-regulated during pollen development of synthetic allotetraploid B. carinata. In summary, these results provide some refreshing updates about the ploidy-related restoration of pollen viability in newly synthesised allotetraploid B. carinata.
Collapse
Affiliation(s)
- Ao Wang
- Henan International Joint Laboratory of Crop Gene Resources and Improvements, School of Agricultural Sciences, Zhengzhou University, Zhengzhou, 450001, Henan, China
| | - Xiaohan Shen
- Henan International Joint Laboratory of Crop Gene Resources and Improvements, School of Agricultural Sciences, Zhengzhou University, Zhengzhou, 450001, Henan, China
- Institute of Horticulture, Henan Academy of Agricultural Sciences, Graduate T & R Base of Zhengzhou University, Zhengzhou, 450002, Henan, China
| | - Niannian Liang
- Henan International Joint Laboratory of Crop Gene Resources and Improvements, School of Agricultural Sciences, Zhengzhou University, Zhengzhou, 450001, Henan, China
| | - Zhengqing Xie
- Henan International Joint Laboratory of Crop Gene Resources and Improvements, School of Agricultural Sciences, Zhengzhou University, Zhengzhou, 450001, Henan, China
| | - Zhaoran Tian
- Henan International Joint Laboratory of Crop Gene Resources and Improvements, School of Agricultural Sciences, Zhengzhou University, Zhengzhou, 450001, Henan, China
- Institute of Horticulture, Henan Academy of Agricultural Sciences, Graduate T & R Base of Zhengzhou University, Zhengzhou, 450002, Henan, China
| | - Luyue Zhang
- Henan International Joint Laboratory of Crop Gene Resources and Improvements, School of Agricultural Sciences, Zhengzhou University, Zhengzhou, 450001, Henan, China
| | - Jialin Guo
- Henan International Joint Laboratory of Crop Gene Resources and Improvements, School of Agricultural Sciences, Zhengzhou University, Zhengzhou, 450001, Henan, China
| | - Fang Wei
- Henan International Joint Laboratory of Crop Gene Resources and Improvements, School of Agricultural Sciences, Zhengzhou University, Zhengzhou, 450001, Henan, China.
| | - Gongyao Shi
- Henan International Joint Laboratory of Crop Gene Resources and Improvements, School of Agricultural Sciences, Zhengzhou University, Zhengzhou, 450001, Henan, China.
| | - Xiaochun Wei
- Institute of Horticulture, Henan Academy of Agricultural Sciences, Graduate T & R Base of Zhengzhou University, Zhengzhou, 450002, Henan, China.
| |
Collapse
|
41
|
Wang Q, Zhu Y, Pei J. Targeting EGFR with molecular degraders as a promising strategy to overcome resistance to EGFR inhibitors. Future Med Chem 2024; 16:1923-1944. [PMID: 39206853 PMCID: PMC11485768 DOI: 10.1080/17568919.2024.2389764] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Accepted: 07/31/2024] [Indexed: 09/04/2024] Open
Abstract
Abnormal activation of EGFR is often associated with various malignant tumors, making it an important target for antitumor therapy. However, traditional targeted inhibitors have several limitations, such as drug resistance and side effects. Many studies have focused on the development of EGFR degraders to overcome this resistance and enhance the therapeutic effect on tumors. Proteolysis targeting chimeras (PROTAC) and Lysosome-based degradation techniques have made significant progress in degrading EGFR. This review provides a summary of the structural and function of EGFR, the resistance, particularly the research progress and activity of EGFR degraders via the proteasome and lysosome. Furthermore, this review aims to provide insights for the development of the novel EGFR degraders.
Collapse
Affiliation(s)
- Qiangfeng Wang
- Department of Medical Oncology, The First Affiliated Hospital, Zhejiang University School of Medicine, 310003, Hangzhou, Zhejiang, China
| | - Yumeng Zhu
- State Key Laboratory of Biotherapy & Cancer Center, West China Hospital, Collaborative Innovation Center of Biotherapy, Sichuan University, Chengdu, 610041, China
| | - Junping Pei
- Shandong Laboratory of Yantai Drug Discovery, Bohai Rim Advanced Research Institute for Drug Discovery, Yantai, Shandong, 264117, China
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| |
Collapse
|
42
|
Guo X, Chen Z, Miao Y, Zhang G, Miao L. VMA21: unveiling a novel oncogene that facilitates immune evasion in triple-negative breast cancer through TCIRG1 protein stability regulation. Am J Cancer Res 2024; 14:4096-4111. [PMID: 39267677 PMCID: PMC11387875 DOI: 10.62347/ngsd3193] [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: 05/11/2024] [Accepted: 08/24/2024] [Indexed: 09/15/2024] Open
Abstract
BACKGROUND VMA21 has been shown to be dysregulated in a number of cancers. However, no study has yet explored whether VMA21 is involved in the regulation of triple-negative breast cancer (TNBC), especially from the level of immune escape. METHODS The Gene Expression Omnibus (GEO) database was accessed to obtain the microarray dataset identified as GSE38959, which was then subjected to an analysis aimed at identifying genes that are differentially expressed (DEGs). The researchers examined the expression of VMA21 in TNBC cell lines. After knockdown of VMA21 in TNBC cells, cell proliferation, invasion, and migration were assessed by clone formation, cell scratch, and Transwell assay, respectively. The effect of VMA21 on immune cell function was explored by cell co-culture method, which was used to assess how TNBC cells with suppressed VMA21 expression affected CD8+ T cytotoxic potential and cytokine secretion. The effect of VMA21 on TCIRG1 protein stability and ubiquitination was assessed using immunoprecipitation. The effects of VMA21 knockdown on tumor xenograft growth and CD8+ T cell immune infiltration in mice were further evaluated. RESULTS VMA21 expression is significantly elevated across various cell lines of TNBC. Furthermore, the perturbation of VMA21 notably suppresses key cell viability parameters in TNBC cells, including their proliferation, invasiveness, and migratory abilities. The modulation of VMA21 in TNBC cells can lead to a substantial augmentation in CD8+ T cell effectiveness. VMA21 stabilizes TCIRG1 protein expression by inhibiting its ubiquitination degradation. Further, VMA21 is involved in regulating TNBC cell proliferation, invasion and immune escape by promoting TCIRG1 expression. CONCLUSIONS VMA21 is able to regulate TCIRG1 protein stability by binding to TCIRG1 protein in the form of ubiquitination, which ultimately promotes the malignant behavior as well as immune escape of TNBC cells.
Collapse
Affiliation(s)
- Xiangyang Guo
- Department of Breast Surgery, Shanxi Province Cancer Hospital/Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Sciences/Cancer Hospital Affiliated to Shanxi Medical University Taiyuan 030013, Shanxi, P. R. China
| | - Zhiqiang Chen
- Department of Breast Surgery, Shanxi Province Cancer Hospital/Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Sciences/Cancer Hospital Affiliated to Shanxi Medical University Taiyuan 030013, Shanxi, P. R. China
| | - Yongmin Miao
- Department of Breast Surgery, Shanxi Province Cancer Hospital/Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Sciences/Cancer Hospital Affiliated to Shanxi Medical University Taiyuan 030013, Shanxi, P. R. China
| | - Guochen Zhang
- Department of Breast Surgery, Shanxi Province Cancer Hospital/Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Sciences/Cancer Hospital Affiliated to Shanxi Medical University Taiyuan 030013, Shanxi, P. R. China
| | - Lifeng Miao
- Department of Breast Surgery, Shanxi Province Cancer Hospital/Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Sciences/Cancer Hospital Affiliated to Shanxi Medical University Taiyuan 030013, Shanxi, P. R. China
| |
Collapse
|
43
|
Tyrna P, Procyk G, Szeleszczuk Ł, Młynarczuk-Biały I. Different Strategies to Overcome Resistance to Proteasome Inhibitors-A Summary 20 Years after Their Introduction. Int J Mol Sci 2024; 25:8949. [PMID: 39201634 PMCID: PMC11354503 DOI: 10.3390/ijms25168949] [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: 07/02/2024] [Revised: 08/09/2024] [Accepted: 08/14/2024] [Indexed: 09/02/2024] Open
Abstract
Proteasome inhibitors (PIs), bortezomib, carfilzomib, and ixazomib, are the first-line treatment for multiple myeloma (MM). They inhibit cytosolic protein degradation in cells, which leads to the accumulation of misfolded and malfunctioned proteins in the cytosol and endoplasmic reticulum, resulting in cell death. Despite being a breakthrough in MM therapy, malignant cells develop resistance to PIs via different mechanisms. Understanding these mechanisms drives research toward new anticancer agents to overcome PI resistance. In this review, we summarize the mechanism of action of PIs and how MM cells adapt to these drugs to develop resistance. Finally, we explore these mechanisms to present strategies to interfere with PI resistance. The strategies include new inhibitors of the ubiquitin-proteasome system, drug efflux inhibitors, autophagy disruption, targeting stress response mechanisms, affecting survival and cell cycle regulators, bone marrow microenvironment modulation, and immunotherapy. We list potential pharmacological targets examined in in vitro, in vivo, and clinical studies. Some of these strategies have already provided clinicians with new anti-MM medications, such as panobinostat and selinexor. We hope that further exploration of the subject will broaden the range of therapeutic options and improve patient outcomes.
Collapse
Affiliation(s)
- Paweł Tyrna
- Histology and Embryology Students’ Science Association, Department of Histology and Embryology, Faculty of Medicine, Warsaw Medical University, Chalubinskiego 5, 02-004 Warsaw, Poland;
| | - Grzegorz Procyk
- 1st Chair and Department of Cardiology, Medical University of Warsaw, Banacha 1A, 02-097 Warsaw, Poland;
| | - Łukasz Szeleszczuk
- Department of Organic and Physical Chemistry, Faculty of Pharmacy, Medical University of Warsaw, Banacha 1 Str., 02-093 Warsaw, Poland;
| | - Izabela Młynarczuk-Biały
- Department of Histology and Embryology, Faculty of Medicine, Warsaw Medical University, Chalubinskiego 5, 02-004 Warsaw, Poland
| |
Collapse
|
44
|
Galla MS, Sharma N, Mishra P, Shankaraiah N. Recent insights of PROTAC developments in inflammation-mediated and autoimmune targets: a critical review. RSC Med Chem 2024; 15:2585-2600. [PMID: 39149114 PMCID: PMC11324044 DOI: 10.1039/d4md00142g] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Accepted: 05/08/2024] [Indexed: 08/17/2024] Open
Abstract
According to the mounting evidence in the literature, pro-inflammatory mediators/targets activate multiple signalling pathways to trigger illnesses that are ultimately responsible for acute pain, chronic inflammatory diseases, and several auto-immune disorders. Conventional drugs have been ruled out since proteolysis-targeting chimeras (PROTACs) are poised to overcome the limitations of traditional therapies. These heterobifunctional molecules help to degrade the targeted proteins of interest through ubiquitination. This review encompasses current and future aspects of PROTACs in inflammation-mediated and autoimmune targets. Different key points are highlighted and discussed, such as why PROTACs are preferred in this disease area, drawbacks and lessons learnt from the past, the role of linkers in establishing crucial degradation, in vitro findings, pharmacokinetics, in silico parameters, limitations of PROTACs in clinical settings, and future outcomes.
Collapse
Affiliation(s)
- Mary Sravani Galla
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research (NIPER) Hyderabad 500037 India
| | - Nitika Sharma
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research (NIPER) Hyderabad 500037 India
| | - Priyanka Mishra
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research (NIPER) Hyderabad 500037 India
| | - Nagula Shankaraiah
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research (NIPER) Hyderabad 500037 India
| |
Collapse
|
45
|
Yan M, Mei Y, Zhang T, Liu Z, Su L, Xiao Y, Zhong X, Lu Y. USP7 cardiomyocyte specific knockout causes disordered mitochondrial biogenesis and dynamics and early neonatal lethality in mice. Int J Cardiol 2024; 408:132149. [PMID: 38723908 DOI: 10.1016/j.ijcard.2024.132149] [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: 01/26/2024] [Revised: 04/22/2024] [Accepted: 05/06/2024] [Indexed: 05/12/2024]
Abstract
BACKGROUND Ubiquitination is an enzymatic modification involving ubiquitin chains, that can be reversed by deubiquitination (DUB) enzymes. Ubiquitin-specific protease 7 (USP7), which is also known as herpes virus-associated ubiquitin-specific protease (HAUSP), has been shown to play a vital role in cardiovascular diseases. However, the underlying molecular mechanism by which USP7 regulates cardiomyocyte function has not been reported. METHODS To understand the physiological function of USP7 in the heart, we constructed cardiomyocyte-specific USP7 conditional knockout mice. RESULTS We found that homozygous knockout mice died approximately three weeks after birth, while heterozygous knockout mice grew normally into adulthood. Severe cardiac dysfunction, hypertrophy, fibrosis, and cell apoptosis were observed in cardiomyocyte-specific USP7 knockout mice, and these effects were accompanied by disordered mitochondrial dynamics and cardiometabolic-related proteins. CONCLUSIONS In summary, we investigated changes in the growth status and cardiac function of cardiomyocyte-specific USP7 knockout mice, and preliminarily explored the underlying mechanism.
Collapse
Affiliation(s)
- Meiling Yan
- Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine, China; Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education of China, Guangzhou, China; Guangdong Key Laboratory of Metabolic Disease Prevention and Treatment of Traditional Chinese Medicine, China; Institute of Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou, China.
| | - Yu Mei
- Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine, China; Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education of China, Guangzhou, China; Guangdong Key Laboratory of Metabolic Disease Prevention and Treatment of Traditional Chinese Medicine, China; Institute of Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou, China
| | - Tianjun Zhang
- People's Hospital of Huadu District, Xinhua Road, Huadu District, Guangzhou 510800, China
| | - Zhou Liu
- Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine, China; Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education of China, Guangzhou, China; Guangdong Key Laboratory of Metabolic Disease Prevention and Treatment of Traditional Chinese Medicine, China; Institute of Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou, China
| | - Liyan Su
- Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine, China; Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education of China, Guangzhou, China; Guangdong Key Laboratory of Metabolic Disease Prevention and Treatment of Traditional Chinese Medicine, China; Institute of Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou, China
| | - Yang Xiao
- Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine, China; Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education of China, Guangzhou, China; Guangdong Key Laboratory of Metabolic Disease Prevention and Treatment of Traditional Chinese Medicine, China; Institute of Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou, China
| | - Xunlong Zhong
- Department of Pharmacy, The Second Affiliated Hospital of Guangzhou Medical University, 250 Changgang East Road, Guangzhou 510260, China
| | - Yanjie Lu
- Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine, China; Department of Pharmacology, State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Medicine Research, Ministry of Education, College of Pharmacy, Harbin Medical University, Harbin 150081, China,; Translational Medicine Research and Cooperation Center of Northern China, Heilongjiang Academy of Medical Sciences, Harbin Medical University, Harbin 150081, China; Research Unit of Noninfectious Chronic Diseases in Frigid Zone (2019RU070), Chinese Academy of Medical Sciences, Harbin 150081, China.
| |
Collapse
|
46
|
Prajapati SK, Pathak A, Samaiya PK. Alzheimer's disease: from early pathogenesis to novel therapeutic approaches. Metab Brain Dis 2024; 39:1231-1254. [PMID: 39046584 DOI: 10.1007/s11011-024-01389-6] [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: 12/06/2023] [Accepted: 07/15/2024] [Indexed: 07/25/2024]
Abstract
The mainstay behind Alzheimer's disease (AD) remains unknown due to the elusive pathophysiology of the disease. Beta-amyloid and phosphorylated Tau is still widely incorporated in various research studies while studying AD. However, they are not sufficient. Therefore, many scientists and researchers have dug into AD studies to deliver many innovations in this field. Many novel biomarkers, such as phosphoglycerate-dehydrogenase, clusterin, microRNA, and a new peptide ratio (Aβ37/Aβ42) in cerebral-spinal fluid, plasma glial-fibrillary-acidic-protein, and lipid peroxidation biomarkers, are mushrooming. They are helping scientists find breakthroughs and substantiating their research on the early detection of AD. Neurovascular unit dysfunction in AD is a significant discovery that can help us understand the relationship between neuronal activity and cerebral blood flow. These new biomarkers are promising and can take these AD studies to another level. There have also been big steps forward in diagnosing and finding AD. One example is self-administered-gerocognitive-examination, which is less expensive and better at finding AD early on than mini-mental-state-examination. Quantum brain sensors and electrochemical biosensors are innovations in the detection field that must be explored and incorporated into the studies. Finally, novel innovations in AD studies like nanotheranostics are the future of AD treatment, which can not only diagnose and detect AD but also offer treatment. Non-pharmacological strategies to treat AD have also yielded interesting results. Our literature review spans from 1957 to 2022, capturing research and trends in the field over six decades. This review article is an update not only on the recent advances in the search for credible biomarkers but also on the newer detection techniques and therapeutic approaches targeting AD.
Collapse
Affiliation(s)
- Santosh Kumar Prajapati
- Bhavdiya Institute of Pharmaceutical Sciences and Research, Ayodhya, UP, India
- Department of Neurosurgery and Brain Repair, University of South Florida, Tampa, FL, 33613, USA
| | - Arjit Pathak
- Department of Pharmacy Shri G.S. Institute of Technology and Science, Indore, 452003, Madhya Pradesh, India
| | - Puneet K Samaiya
- Department of Pharmacy Shri G.S. Institute of Technology and Science, Indore, 452003, Madhya Pradesh, India.
| |
Collapse
|
47
|
Shokeen K, Baroi MK, Chahar M, Das D, Saini H, Kumar S. Arginyltransferase 1 (ATE1)-mediated proteasomal degradation of viral haemagglutinin protein: a unique host defence mechanism. J Gen Virol 2024; 105. [PMID: 39207120 DOI: 10.1099/jgv.0.002020] [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] [Indexed: 09/04/2024] Open
Abstract
The extensive protein production in virus-infected cells can disrupt protein homeostasis and activate various proteolytic pathways. These pathways utilize post-translational modifications (PTMs) to drive the ubiquitin-mediated proteasomal degradation of surplus proteins. Protein arginylation is the least explored PTM facilitated by arginyltransferase 1 (ATE1) enzyme. Several studies have provided evidence supporting its importance in multiple physiological processes, including ageing, stress, nerve regeneration, actin formation and embryo development. However, its function in viral pathogenesis is still unexplored. The present work utilizes Newcastle disease virus (NDV) as a model to establish the role of the ATE1 enzyme and its activity in pathogenesis. Our data indicate a rise in levels of N-arginylated cellular proteins in the infected cells. Here, we also explore the haemagglutinin-neuraminidase (HN) protein of NDV as a presumable target for arginylation. The data indicate that the administration of Arg amplifies the arginylation process, resulting in reduced stability of the HN protein. ATE1 enzyme activity inhibition and gene expression knockdown studies were also conducted to analyse modulation in HN protein levels, which further substantiated the findings. Moreover, we also observed Arg addition and probable ubiquitin modification to the HN protein, indicating engagement of the proteasomal degradation machinery. Lastly, we concluded that the enhanced levels of the ATE1 enzyme could transfer the Arg residue to the N-terminus of the HN protein, ultimately driving its proteasomal degradation.
Collapse
Affiliation(s)
- Kamal Shokeen
- Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati, India
| | - Malay Kumar Baroi
- Department of Chemistry, Indian Institute of Technology Guwahati, Guwahati, India
| | - Manjeet Chahar
- Centre for Medical Biotechnology, Maharshi Dayanand University, Rohtak, India
| | - Debapratim Das
- Department of Chemistry, Indian Institute of Technology Guwahati, Guwahati, India
| | - Harimohan Saini
- Centre for Medical Biotechnology, Maharshi Dayanand University, Rohtak, India
| | - Sachin Kumar
- Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati, India
| |
Collapse
|
48
|
Waltho A, Popp O, Lenz C, Pluska L, Lambert M, Dötsch V, Mertins P, Sommer T. K48- and K63-linked ubiquitin chain interactome reveals branch- and length-specific ubiquitin interactors. Life Sci Alliance 2024; 7:e202402740. [PMID: 38803224 PMCID: PMC11109483 DOI: 10.26508/lsa.202402740] [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: 03/27/2024] [Revised: 05/08/2024] [Accepted: 05/08/2024] [Indexed: 05/29/2024] Open
Abstract
The ubiquitin (Ub) code denotes the complex Ub architectures, including Ub chains of different lengths, linkage types, and linkage combinations, which enable ubiquitination to control a wide range of protein fates. Although many linkage-specific interactors have been described, how interactors are able to decode more complex architectures is not fully understood. We conducted a Ub interactor screen, in humans and yeast, using Ub chains of varying lengths, as well as homotypic and heterotypic branched chains of the two most abundant linkage types-lysine 48-linked (K48) and lysine 63-linked (K63) Ub. We identified some of the first K48/K63-linked branch-specific Ub interactors, including histone ADP-ribosyltransferase PARP10/ARTD10, E3 ligase UBR4, and huntingtin-interacting protein HIP1. Furthermore, we revealed the importance of chain length by identifying interactors with a preference for Ub3 over Ub2 chains, including Ub-directed endoprotease DDI2, autophagy receptor CCDC50, and p97 adaptor FAF1. Crucially, we compared datasets collected using two common deubiquitinase inhibitors-chloroacetamide and N-ethylmaleimide. This revealed inhibitor-dependent interactors, highlighting the importance of inhibitor consideration during pulldown studies. This dataset is a key resource for understanding how the Ub code is read.
Collapse
Affiliation(s)
- Anita Waltho
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
- Institute for Biology, Humboldt-University zu Berlin, Berlin, Germany
| | - Oliver Popp
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
| | - Christopher Lenz
- Institute for Biophysical Chemistry and Center for Biomolecular Magnetic Resonance, Goethe University, Frankfurt, Germany
| | - Lukas Pluska
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
- Institute for Biology, Humboldt-University zu Berlin, Berlin, Germany
| | - Mahil Lambert
- Institute for Biophysical Chemistry and Center for Biomolecular Magnetic Resonance, Goethe University, Frankfurt, Germany
| | - Volker Dötsch
- Institute for Biophysical Chemistry and Center for Biomolecular Magnetic Resonance, Goethe University, Frankfurt, Germany
| | - Philipp Mertins
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
| | - Thomas Sommer
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
- Institute for Biology, Humboldt-University zu Berlin, Berlin, Germany
| |
Collapse
|
49
|
Ghasemi N, Azizi H. Exploring Myc puzzle: Insights into cancer, stem cell biology, and PPI networks. Gene 2024; 916:148447. [PMID: 38583818 DOI: 10.1016/j.gene.2024.148447] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2023] [Revised: 03/13/2024] [Accepted: 04/04/2024] [Indexed: 04/09/2024]
Abstract
"The grand orchestrator," "Universal Amplifier," "double-edged sword," and "Undruggable" are just some of the Myc oncogene so-called names. It has been around 40 years since the discovery of the Myc, and it remains in the mainstream of cancer treatment drugs. Myc is part of basic helix-loop-helix leucine zipper (bHLH-LZ) superfamily proteins, and its dysregulation can be seen in many malignant human tumors. It dysregulates critical pathways in cells that are connected to each other, such as proliferation, growth, cell cycle, and cell adhesion, impacts miRNAs action, intercellular metabolism, DNA replication, differentiation, microenvironment regulation, angiogenesis, and metastasis. Myc, surprisingly, is used in stem cell research too. Its family includes three members, MYC, MYCN, and MYCL, and each dysfunction was observed in different cancer types. This review aims to introduce Myc and its function in the body. Besides, Myc deregulatory mechanisms in cancer cells, their intricate aspects will be discussed. We will look at promising drugs and Myc-based therapies. Finally, Myc and its role in stemness, Myc pathways based on PPI network analysis, and future insights will be explained.
Collapse
Affiliation(s)
- Nima Ghasemi
- Faculty of Biotechnology, Amol University of Special Modern Technologies, Amol, Iran
| | - Hossein Azizi
- Faculty of Biotechnology, Amol University of Special Modern Technologies, Amol, Iran.
| |
Collapse
|
50
|
Chen Y, Liu Y, Li Y, Yao C, Qu J, Tang J, Chen G, Han Y. Acute exposure to polystyrene nanoplastics induces unfolded protein response and global protein ubiquitination in lungs of mice. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2024; 280:116580. [PMID: 38865938 DOI: 10.1016/j.ecoenv.2024.116580] [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: 02/20/2024] [Revised: 05/21/2024] [Accepted: 06/08/2024] [Indexed: 06/14/2024]
Abstract
Inhaling microplastics (MPs) and nanoplastics (NPs) in the air can damage lung function. Xenobiotics in the body can cause endoplasmic reticulum (ER) stress, and the unfolded protein response (UPR) activation alleviates ER stress. Degradation of unfolded or misfolded proteins is an important pathway for recovering cellular homeostasis. The UPR and protein degradation induced by MPs/NPs in lung tissues are not well understood. Here, we investigated the UPR and protein ubiquitination in the lungs of mice exposed to polystyrene (PS)-NPs and their possible molecular mechanisms leading to protein ubiquitination. Mice were intratracheally administered with 5.6, 17, and 51 mg/kg PS-NPs once for 24 h. Exposure to PS-NPs elevated protein ubiquitination in the lungs of mice in a dose-dependent manner. PS-NPs activated three branches of UPR including inositol-requiring protein 1α (IRE1α), eukaryotic translation initiator factor 2α (eIF2α), and activating transcription factor 6α (ATF6α) in the lungs of mice. However, activated IRE1α did not trigger X-box binding protein 1 (XBP1) mRNA splicing. Exposure to PS-NPs induced an increase in the levels of E3 ubiquitin ligase hydroxymethyl glutaryl-coenzyme A reductase degradation protein 1 (HRD1) and carboxy terminus of Hsc70 interacting protein (CHIP) in the lungs of mice and BEAS-2B cells. ATF6α siRNA inhibited the levels of HRD1 and CHIP proteins induced by PS-NPs in BEAS-2B cells. These results suggest that ATF6α plays a critical role in increasing ubiquitination of unfolded or misfolded proteins by alleviating PS-NPs induced ER stress through UPR to achieve ER homeostasis in the lungs of mice.
Collapse
Affiliation(s)
- Yanhong Chen
- Department of Occupational Medicine and Environmental Toxicology, College of Public Health, Nantong University, Nantong, Jiangsu 226019, China
| | - Yingqi Liu
- Department of Occupational Medicine and Environmental Toxicology, College of Public Health, Nantong University, Nantong, Jiangsu 226019, China; Wujiang Center for Disease Control and Prevention, Suzhou, Jiangsu 215299, China
| | - Yanli Li
- Department of Occupational Medicine and Environmental Toxicology, College of Public Health, Nantong University, Nantong, Jiangsu 226019, China
| | - Chenjuan Yao
- Department of Molecular Oral Physiology, Institute of Health Biosciences, University of Tokushima Graduate School, Tokushima-Shi, Tokushima 770-8504, Japan
| | - Jianhua Qu
- Department of Occupational Medicine and Environmental Toxicology, College of Public Health, Nantong University, Nantong, Jiangsu 226019, China
| | - Juan Tang
- Department of Occupational Medicine and Environmental Toxicology, College of Public Health, Nantong University, Nantong, Jiangsu 226019, China
| | - Gang Chen
- Department of Occupational Medicine and Environmental Toxicology, College of Public Health, Nantong University, Nantong, Jiangsu 226019, China.
| | - Yu Han
- Department of Occupational Medicine and Environmental Toxicology, College of Public Health, Nantong University, Nantong, Jiangsu 226019, China.
| |
Collapse
|