1
|
Wang H, Sun X, Cui K, Wang Y, Sun Y. Ubiquitin specific peptidase 13 protects against inflammation-associated joint injury in collagen-induced rheumatoid arthritis mice by targeting TRAF6. Int Immunopharmacol 2025; 156:114617. [PMID: 40252462 DOI: 10.1016/j.intimp.2025.114617] [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: 08/08/2024] [Revised: 04/01/2025] [Accepted: 04/02/2025] [Indexed: 04/21/2025]
Abstract
Rheumatoid arthritis (RA) is a chronic inflammatory autoimmune disease with complex pathogenesis. Ubiquitin-specific protease 13 (USP13), a member of the deubiquitinating enzyme (DUB) superfamily, plays diverse roles in cellular events. This study investigates the role of USP13 in RA, revealing its significant downregulation in peripheral blood mononuclear cells (PBMCs) from RA patients compared to healthy individuals. USP13 expression negatively correlates with RA characteristics, including erythrocyte sedimentation rate (ESR), C-reactive protein (CRP), and rheumatoid factor (RF). Consistent with these findings, USP13 levels were reduced in knee joint synovial tissues of collagen-induced arthritis (CIA) mice and lipopolysaccharide (LPS)-stimulated bone marrow-derived macrophages (BMDMs). Mechanistically, USP13 directly interacts with tumor necrosis factor receptor-associated factor 6 (TRAF6), reducing its K63-linked polyubiquitination and thereby inhibiting TRAF6 expression. This interaction restrains nuclear factor κB (NF-κB) signaling, leading to a marked attenuation of LPS-induced inflammatory responses in BMDMs. Importantly, the anti-inflammatory effects of USP13 over-expression are largely dependent on TRAF6 suppression. In vitro, USP13 over-expression in BMDMs inhibits the proliferation of fibroblast-like synoviocytes (FLS) and osteoclastogenesis. In addition, USP13-overexpressing BMDMs in CIA mice significantly alleviates RA development, as evidenced by reduced synovial hyperplasia, inflammatory cell infiltration, cartilage destruction, and bone loss. These findings highlight the essential role of USP13 in macrophages during RA progression and reveal its therapeutic potential by targeting TRAF6 signaling.
Collapse
Affiliation(s)
- Haili Wang
- Department of General Medince, Affiliated Hospital of Beihua University, Jilin 132011, China
| | - Xinyi Sun
- Department of General Medince, Affiliated Hospital of Beihua University, Jilin 132011, China
| | - Kai Cui
- Department of Oncology, Affiliated Hospital of Beihua University, Jilin 132011, China
| | - Yan Wang
- Geriatric Department, Jilin Integrated Traditional Chinese and Western Medicine Hospital of Jilin, Jilin 132011, China
| | - Yuman Sun
- Department of General Medince, Affiliated Hospital of Beihua University, Jilin 132011, China.
| |
Collapse
|
2
|
Deng S, Zhang Z, Liu L, Xu C, Zhang D, Dong L, Gao C, Wang X, Fan Z. The E3 ligase c-Cbl modulates microglial phenotypes and contributes to Parkinson's disease pathology. Cell Death Discov 2025; 11:184. [PMID: 40246829 PMCID: PMC12006326 DOI: 10.1038/s41420-025-02482-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2024] [Revised: 04/03/2025] [Accepted: 04/07/2025] [Indexed: 04/19/2025] Open
Abstract
Microglial activation, particularly the polarization between classical (M1 phenotype) and alternative (M2 phenotype) states, plays pivotal roles in the immune pathogenesis of Parkinson's disease (PD), with the M1 phenotype exerting neurotoxic effects and the M2 phenotype conferring neuroprotection. Modulating microglial polarization toward the M2 phenotype holds therapeutic potential for PD. This study investigated the role of c-Cbl, an E3 ubiquitin ligase implicated in modulating microglial phenotypes and protecting dopaminergic neurons. Our findings revealed that c-Cbl-/- mice exhibited motor deficits, reduced striatal dopamine levels, and progressive dopaminergic neuron loss in the substantia nigra (SN). Genetic ablation of c-Cbl significantly increased proinflammatory cytokine release and microglial activation in the SN, accompanied by a phenotypic shift from M2 to M1 polarization. Furthermore, stereotaxic c-Cbl knockdown in the SN exacerbated behavioral impairments and accelerated dopaminergic neuron degeneration in the MPTP-induced mouse model of PD. At the molecular level, c-Cbl deletion promoted M1 polarization of microglia through dysregulation of the PI3K/Akt signaling pathway, thereby impairing dopaminergic neuronal survival. Collectively, this study demonstrates that c-Cbl knockout recapitulates PD-like pathology and drives microglial activation. Our results establish that c-Cbl orchestrates the transition from neurotoxic M1 to neuroprotective M2 microglial phenotypes, highlighting its central role in PD immunopathogenesis. These findings suggest c-Cbl as a promising therapeutic target for modulating microglial polarization and alleviating PD symptoms.
Collapse
Affiliation(s)
- Shumin Deng
- Department of Pharmacology, School of Basic Medical Sciences, Capital Medical University, Beijing, PR China
| | - Zhiyuan Zhang
- Department of Neurobiology, School of Basic Medical Sciences, Capital Medical University, Beijing, PR China
| | - Lu Liu
- Department of Pharmacology, School of Basic Medical Sciences, Capital Medical University, Beijing, PR China
| | - Chen Xu
- Department of Pharmacology, School of Basic Medical Sciences, Capital Medical University, Beijing, PR China
| | - Di Zhang
- Department of Pharmacology, School of Basic Medical Sciences, Capital Medical University, Beijing, PR China
| | - Lin Dong
- Department of Pathology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, PR China
| | - Chunyan Gao
- Department of Clinical Medicine, Yanjing Medical College, Capital Medical University, Beijing, PR China
| | - Xiaomin Wang
- Department of Neurobiology, School of Basic Medical Sciences, Capital Medical University, Beijing, PR China
| | - Zheng Fan
- Department of Pharmacology, School of Basic Medical Sciences, Capital Medical University, Beijing, PR China.
- Co-innovation Center of Neuroregeneration, Nantong University, Nantong, Jiangsu, PR China.
| |
Collapse
|
3
|
Gu H, Yu W, Feng P, Zeng C, Cao Q, Chen F, Wang Z, Shen H, Wu Y, Wang S. Circular RNA circSTX12 regulates osteo-adipogenic balance and proliferation of BMSCs in senile osteoporosis. Cell Mol Life Sci 2025; 82:149. [PMID: 40192802 PMCID: PMC11977094 DOI: 10.1007/s00018-025-05684-y] [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: 06/20/2024] [Revised: 01/03/2025] [Accepted: 03/28/2025] [Indexed: 04/10/2025]
Abstract
Increased adipogenic differentiation and decreased osteogenic differentiation of bone marrow mesenchymal stem cells (BMSCs) along with slow self-renewal are pivotal causes for decreased bone formation in senile osteoporosis. Circular RNAs (circRNAs) play important roles in cell proliferation and differentiation, and are closely related to osteoporosis. Whether circRNAs orchestrate the adipo-osteogenic balance and the proliferation of BMSCs in osteoporosis remains unclear. We found in this study that circSTX12 was abnormally upregulated in bone sections from osteoporosis patients and in BMSCs from aged mice, as well as in later-generation human BMSCs in culture. Knockdown of circSTX12 in BMSCs resulted in enhanced osteogenesis, decreased adipogenesis, and increased proliferation capacity; circSTX12 overexpression had the opposite effect. RNA pull-down and mass spectrometry revealed the interactions between circSTX12 with CBL and LMO7. At the molecular level, circSTX12 regulated cell fate in BMSCs by competitively binding to CBL, reducing the ubiquitination-mediated degradation of MST1 and thereby activating the Hippo pathway, a key regulator of adipo-osteogenic balance. Knockdown of circSTX12 promoted the nuclear localization of YAP. In addition, our findings suggest that LMO7 mediates circSTX12-induced BMSCs proliferation by regulating the transcription of CCNA2, CCNH, and CCND1. In vivo, injection of antisense oligonucleotides (ASOs) to knockdown circSTX12 promoted bone formation in aged mice. Our results provide evidence for circSTX12 as a regulator of adipo-osteogenic differentiation and proliferation of BMSCs through binding to CBL and LMO7, respectively. Targeting circSTX12 may be a novel approach for osteoporosis treatment.
Collapse
Affiliation(s)
- Huimin Gu
- Center for Biotherapy, Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, 518033, P. R. China
- Guangdong Provincial Clinical Research Center for Orthopedic Diseases, The Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, 518033, P. R. China
| | - Wenhui Yu
- Department of Orthopedics, Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, 518033, P. R. China
- Guangdong Provincial Clinical Research Center for Orthopedic Diseases, The Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, 518033, P. R. China
| | - Pei Feng
- Center for Biotherapy, Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, 518033, P. R. China
- Guangdong Provincial Clinical Research Center for Orthopedic Diseases, The Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, 518033, P. R. China
| | - Chenying Zeng
- Center for Biotherapy, Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, 518033, P. R. China
- Guangdong Provincial Clinical Research Center for Orthopedic Diseases, The Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, 518033, P. R. China
| | - Qian Cao
- Center for Biotherapy, Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, 518033, P. R. China
- Guangdong Provincial Clinical Research Center for Orthopedic Diseases, The Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, 518033, P. R. China
| | - Fenglei Chen
- Department of Orthopedics, Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, 518033, P. R. China
- Guangdong Provincial Clinical Research Center for Orthopedic Diseases, The Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, 518033, P. R. China
| | - Ziming Wang
- Department of Orthopedics, Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, 518033, P. R. China
- Guangdong Provincial Clinical Research Center for Orthopedic Diseases, The Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, 518033, P. R. China
| | - Huiyong Shen
- Department of Orthopedics, Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, 518033, P. R. China.
- Guangdong Provincial Clinical Research Center for Orthopedic Diseases, The Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, 518033, P. R. China.
| | - Yanfeng Wu
- Center for Biotherapy, Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, 518033, P. R. China.
- Guangdong Provincial Clinical Research Center for Orthopedic Diseases, The Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, 518033, P. R. China.
| | - Shan Wang
- Center for Biotherapy, Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, 518033, P. R. China.
- Guangdong Provincial Clinical Research Center for Orthopedic Diseases, The Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, 518033, P. R. China.
| |
Collapse
|
4
|
Mohapatra B, Lavudi K, Kokkanti RR, Patnaik S. Regulation of NLRP3/TRIM family signaling in gut inflammation and colorectal cancer. Biochim Biophys Acta Rev Cancer 2025; 1880:189271. [PMID: 39864469 DOI: 10.1016/j.bbcan.2025.189271] [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: 09/19/2024] [Revised: 01/13/2025] [Accepted: 01/16/2025] [Indexed: 01/28/2025]
Abstract
CRC (Colorectal cancer) ranks among the most prevalent tumors in humans and remains a leading cause of cancer-related mortality worldwide. Numerous studies have highlighted the connection between inflammasome over-activation and the initiation and progression of CRC. The activation of the NLRP3 (NOD-like receptor family, pyrin domain containing 3) inflammasome is dependent on the nuclear NF-kβ (Nuclear Factor kappa-light-chain-enhancer of activated B cells) pathway, leading to the maturation and release of inflammatory cytokines such as IL-1ß (Interleukin 1 beta) and IL-18 (Interleukin 18). While inflammation is crucial for defense mechanisms and tissue repair, excessive information can pose significant risks. Mounting evidence suggests that overactivation of the inflammasome contributes to the pathogenesis of inflammatory diseases. Consequently, there is a concerted effort to tightly regulate inflammasome activity and mitigate excessive inflammatory responses, particularly in conditions such as IBD (Inflammatory Bowel Disease), which includes Ulcerative Colitis and Crohn's Disease. The tripartite motif (TRIM) protein family, characterized by a conserved structure and rapid evolutionary diversification, includes members with critical roles in ubiquitination and other regulatory functions. Their importance in modulating inflammatory responses is widely acknowledged. This article aims to investigate the interplay between TRIM proteins and the NLRP3 Inflammasome in CRC and gut inflammation, offering insights for future research endeavors and potential therapeutic strategies.
Collapse
Affiliation(s)
- Bibhashee Mohapatra
- School of Biotechnology, Kalinga Institute of Industrial Technology (KIIT) Deemed to be University, Bhubaneswar, Odisha 751024, India
| | - Kousalya Lavudi
- Department of Radiation Oncology, College of Medicine, The Ohio State University, Columbus, OH 43210, United States; Comprehensive cancer center, The Ohio State University, Columbus, OH, United States
| | - Rekha Rani Kokkanti
- School of Biotechnology, Kalinga Institute of Industrial Technology (KIIT) Deemed to be University, Bhubaneswar, Odisha 751024, India
| | - Srinivas Patnaik
- School of Biotechnology, Kalinga Institute of Industrial Technology (KIIT) Deemed to be University, Bhubaneswar, Odisha 751024, India.
| |
Collapse
|
5
|
Zhao Q, Chen X, Mai B, Che F, Zhang Z, Kang P, Hou C, Lu L, Xu L. Stigmasterol mitigates estrogen-deficiency-induced osteoporosis through inhibition of phosphorylated p65 and MAPK signaling pathways. Front Pharmacol 2024; 15:1527494. [PMID: 39741634 PMCID: PMC11685749 DOI: 10.3389/fphar.2024.1527494] [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: 11/13/2024] [Accepted: 12/02/2024] [Indexed: 01/03/2025] Open
Abstract
Background Osteoporosis is a pervasive bone metabolic disorder characterized by the progressive degeneration of bone microstructure. Osteoclasts are playing a pivotal role in bone remodeling and resorption. Consequently, modulating osteoclast activity, particularly curbing their overactivation, has become a crucial strategy in clinical treatments. Stigmasterol (STG), a plant-derived phytosterol, has shown promise in inhibiting osteoclastic activity, although its precise biological mechanisms require further scientific investigation. Therefore, this study aims to explore the potential mechanisms by which STG inhibits osteoclasts and to further assess its impact on osteoporosis by establishing an Ovariectomy (OVX) model. Methods Initially, osteoclast differentiation was induced in vitro using RANKL (Receptor Activator of Nuclear Factor Kappa-B Ligand) on RAW 264.7 cells, followed by TRAP staining and F-actin banding to observe the effects of various concentrations of STG during osteoclast differentiation. The osteoclast-specific gene and protein expression changes were further analyzed using Real-Time PCR (qPCR) and Western blot, exploring the RANKL-mediated NF-κB and MAPK signaling pathways. An OVX model was established in vivo to examine changes in bone mass through Micro-CT and Hematoxylin and eosin (H&E) staining, and to assess osteoclast formation and characteristic protein expression through TRAP staining and Immunohistochemistry staining. Results In vitro experiments revealed that STG significantly inhibited osteoclast activity, as evidenced by reductions in osteoclast numbers and spreading areas, and a marked suppression of F-actin formation. On the molecular level, this compound effectively downregulated key osteoclast markers such as NFATc1, Acp5, c-Fos, and ΜMP9 in both gene and protein expressions. Western blot analysis showed that STG notably inhibited the phosphorylation of the p65 subunit in the NF-κB pathway, thus affecting the pathway's activity. Further validation through OVX model indicated significant protective effects of STG against bone loss, as demonstrated by Micro-CT. Histopathological staining confirmed STG's efficacy in reducing bone surface area and volume loss. Additionally, TRAP staining showed significant reductions in osteoclast number and surface area in the STG group compared to the OVX group, underscoring STG's potential therapeutic role in bone metabolism regulation. Conclusion The findings reveal that STG effectively inhibits the phosphorylation of the p65 protein in the NF-κB pathway, and influences the MAPK signaling pathway, thereby reducing osteoclast formation and preserving bone mass. These mechanisms provide a crucial molecular basis for its potential therapeutic application in treating osteoporosis.
Collapse
Affiliation(s)
- Qiangqiang Zhao
- Lingnan Medical Research Center, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
- Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Xingling Chen
- Lingnan Medical Research Center, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
- Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Bin Mai
- Lingnan Medical Research Center, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
- Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Feihong Che
- Lingnan Medical Research Center, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
- Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Zhen Zhang
- Lingnan Medical Research Center, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
- Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Pan Kang
- Lingnan Medical Research Center, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
- Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Chengyu Hou
- Lingnan Medical Research Center, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
- Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Lu Lu
- Lingnan Medical Research Center, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
- Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Liangliang Xu
- Lingnan Medical Research Center, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
- Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou, China
| |
Collapse
|
6
|
Du Y, Chen H, Zhou L, Guo Q, Gong S, Feng S, Guan Q, Shi P, Lv T, Guo Y, Yang C, Sun P, Li K, Xu S, Li L. REGγ is essential to maintain bone homeostasis by degrading TRAF6, preventing osteoporosis. Proc Natl Acad Sci U S A 2024; 121:e2405265121. [PMID: 39536082 PMCID: PMC11588133 DOI: 10.1073/pnas.2405265121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Accepted: 10/14/2024] [Indexed: 11/16/2024] Open
Abstract
Primary osteoporosis, manifesting as decreased bone mass and increased bone fragility, is a "silent disease" that is often ignored until a bone breaks. Accordingly, it is urgent to develop reliable biomarkers and novel therapeutic strategies for osteoporosis treatment. Here, we identified REGγ as a potential biomarker of osteoporotic populations through proteomics analysis. Next, we demonstrated that REGγ deficiency increased osteoclast activity and triggered bone mass loss in REGγ knockout (KO) and bone marrow-derive macrophage (BMM)-conditional REGγ KO mice. However, the osteoclast activity decreased in BMM-conditional REGγ overexpression mice. Mechanistically, we defined that REGγ-20S proteasome directly degraded TRAF6 to inhibit bone absorption in a ubiquitin-independent pathway. More importantly, BMM-conditional Traf6 KO with REGγ KO mice could "rescue" the osteoporosis phenotypes. Based on NIP30 (a REGγ "inhibitor") dephosphorylation by CKII inhibition activated the ubiquitin-independent degradation of TRAF6, we selected TTP22, an inhibitor of CKII, and defined that TTP22 could alleviate osteoporosis in vitro and in vivo. Overall, our study reveals a unique function of NIP30/REGγ/TRAF6 axis in osteoporosis and provides a potential therapeutic drug TTP22 for osteoporosis.
Collapse
Affiliation(s)
- Yingying Du
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences, School of Life Sciences, East China Normal University, Shanghai200241, China
| | - Hui Chen
- Department of Trauma-Emergency and Critical Care Medicine, Shanghai Fifth People’s Hospital, Fudan University, Shanghai200240, China
- Joint Center for Translational Medicine, Shanghai Fifth People’s Hospital, Fudan University and School of Life Science, East China Normal University, Shanghai200241, China
- School of Life Sciences, East China Normal University, Shanghai200241, China
| | - Lei Zhou
- Department of Orthopedics, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai200080, China
| | - Qunfeng Guo
- Department of Orthopedics, Changzheng Hospital, Naval Medical University, Shanghai200003, China
| | - Shuangming Gong
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences, School of Life Sciences, East China Normal University, Shanghai200241, China
| | - Siyuan Feng
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences, School of Life Sciences, East China Normal University, Shanghai200241, China
| | - Qiujing Guan
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences, School of Life Sciences, East China Normal University, Shanghai200241, China
| | - Peilin Shi
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences, School of Life Sciences, East China Normal University, Shanghai200241, China
| | - Tongxin Lv
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences, School of Life Sciences, East China Normal University, Shanghai200241, China
| | - Yilan Guo
- The Key Laboratory of Adolescent Health Assessment and Exercise Intervention of the Ministry of Education, East China Normal University, Shanghai200241, China
| | - Cheng Yang
- Department of Orthopedics, Changzheng Hospital, Naval Medical University, Shanghai200003, China
| | - Peng Sun
- The Key Laboratory of Adolescent Health Assessment and Exercise Intervention of the Ministry of Education, East China Normal University, Shanghai200241, China
| | - Kun Li
- Health Science Center, East China Normal University, Shanghai200241, China
| | - Shuogui Xu
- Department of Emergency and Trauma, The First Affiliated Hospital of Naval Medical University, Shanghai200433, China
| | - Lei Li
- Joint Center for Translational Medicine, Shanghai Fifth People’s Hospital, Fudan University and School of Life Science, East China Normal University, Shanghai200241, China
- School of Life Sciences, East China Normal University, Shanghai200241, China
- Chongqing Key Laboratory of Precision Optics, Chongqing Institute of East China Normal University, Chongqing401120, China
- East China Normal University, Shanghai Frontiers Science Center of Genome Editing and Cell Therapy, Shanghai200241, China
| |
Collapse
|
7
|
Liu F, Chen J, Li K, Li H, Zhu Y, Zhai Y, Lu B, Fan Y, Liu Z, Chen X, Jia X, Dong Z, Liu K. Ubiquitination and deubiquitination in cancer: from mechanisms to novel therapeutic approaches. Mol Cancer 2024; 23:148. [PMID: 39048965 PMCID: PMC11270804 DOI: 10.1186/s12943-024-02046-3] [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: 04/17/2024] [Accepted: 06/15/2024] [Indexed: 07/27/2024] Open
Abstract
Ubiquitination, a pivotal posttranslational modification of proteins, plays a fundamental role in regulating protein stability. The dysregulation of ubiquitinating and deubiquitinating enzymes is a common feature in various cancers, underscoring the imperative to investigate ubiquitin ligases and deubiquitinases (DUBs) for insights into oncogenic processes and the development of therapeutic interventions. In this review, we discuss the contributions of the ubiquitin-proteasome system (UPS) in all hallmarks of cancer and progress in drug discovery. We delve into the multiple functions of the UPS in oncology, including its regulation of multiple cancer-associated pathways, its role in metabolic reprogramming, its engagement with tumor immune responses, its function in phenotypic plasticity and polymorphic microbiomes, and other essential cellular functions. Furthermore, we provide a comprehensive overview of novel anticancer strategies that leverage the UPS, including the development and application of proteolysis targeting chimeras (PROTACs) and molecular glues.
Collapse
Affiliation(s)
- Fangfang Liu
- Tianjian Laboratory of Advanced Biomedical Sciences, Academy of Medical Sciences, College of Medicine, Zhengzhou University, Zhengzhou, Henan, 450001, China
- China-US (Henan) Hormel Cancer Institute, Zhengzhou, Henan, 450000, China
| | - Jingyu Chen
- Department of Pediatric Medicine, School of Third Clinical Medical Sciences, Zhengzhou University, Zhengzhou, 450000, China
| | - Kai Li
- Department of Clinical Medicine, School of First Clinical Medical Sciences, Zhengzhou University, Zhengzhou, 450000, China
| | - Haochen Li
- Department of Clinical Medicine, School of First Clinical Medical Sciences, Zhengzhou University, Zhengzhou, 450000, China
| | - Yiyi Zhu
- Department of Clinical Medicine, School of First Clinical Medical Sciences, Zhengzhou University, Zhengzhou, 450000, China
| | - Yubo Zhai
- Department of Pathophysiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450000, China
| | - Bingbing Lu
- Department of Pathophysiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450000, China
| | - Yanle Fan
- China-US (Henan) Hormel Cancer Institute, Zhengzhou, Henan, 450000, China
| | - Ziyue Liu
- Department of Pathophysiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450000, China
| | - Xiaojie Chen
- School of Basic Medicine, Henan University of Science and Technology, Luoyang, China
| | - Xuechao Jia
- Henan International Joint Laboratory of TCM Syndrome and Prescription in Signaling, Traditional Chinese Medicine (Zhong Jing) School, Henan University of Chinese Medicine, Zhengzhou, Henan, China.
| | - Zigang Dong
- Tianjian Laboratory of Advanced Biomedical Sciences, Academy of Medical Sciences, College of Medicine, Zhengzhou University, Zhengzhou, Henan, 450001, China.
- China-US (Henan) Hormel Cancer Institute, Zhengzhou, Henan, 450000, China.
- Department of Pathophysiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450000, China.
| | - Kangdong Liu
- Tianjian Laboratory of Advanced Biomedical Sciences, Academy of Medical Sciences, College of Medicine, Zhengzhou University, Zhengzhou, Henan, 450001, China.
- Department of Pathophysiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450000, China.
| |
Collapse
|
8
|
Chen Z, Joseph D, Ding M, Bhujbal SP, Rajan RPS, Kim E, Park SW, Lee S, Lee TH. Synthesis and evaluation of 2-NMPA derivatives as potential agents for prevention of osteoporosis in vitro and in vivo. Eur J Med Chem 2023; 260:115767. [PMID: 37651877 DOI: 10.1016/j.ejmech.2023.115767] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Revised: 07/25/2023] [Accepted: 08/25/2023] [Indexed: 09/02/2023]
Abstract
Abnormal osteoclast differentiation causes various bone disorders such as osteoporosis. Targeting the formation and activation of osteoclasts has been recognized as an effective approach for preventing osteoporosis. Herein, we synthesized eleven 2-NMPA derivatives which are (2-(2-chlorophenoxy)-N-(4-alkoxy-2-morpholinophenyl) acetamides, and evaluated their suppression effects on osteoclastogenesis in vitro by using TRAP-staining assay. Among the synthesized eleven novel 2-NMPAs, 4-(2-(2-chlorophenoxy)acetamido)-3-morpholinophenyl trifluoromethanesulfonate (11b), 4-(2-(2-chlorophenoxy) acetamido)-3-morpholinophenyl-3-(N-(2-oxo-2-((2-(phenylthio) phenyl) amino) ethyl)methylsulfonamido)benzoate (11d), and 4-(2-(2-chlorophenoxy) acetamido)-3-morpholinophenyl 4-acetamidobenzenesulfonate (11h) displayed highly inhibitory bioactivity on the differentiation of primary osteoclasts. 11h was selected for further investigation of the inhibitory effects and potential mechanism involved in the suppression of osteoclastogenesis. In vitro analysis suggested that 11h inhibited osteoclastogenesis with an IC50 of 358.29 nM, decreased the formation of F-action belts and bone resorption, without interfering cell viability and osteoblast differentiation. Furthermore, the mRNA expressions of osteoclast-specific genes such as Acp5, Nfatc1, Dc-stamp, Atp6v0d2, Mmp9, and Ctsk significantly decreased following 11h treatment. RANKL-induced osteoclast-specific proteins analysis demonstrated that 11h suppressed osteoclast differentiation by downregulating of RANKL-mediated TRAF6 expression, followed by inactivation of PI3K/AKT and IκBα/NF-κB signaling pathways. Finally, 11h inhibited ovariectomy-induced bone loss in vivo. Therefore, the current work highlighted the therapeutic potential of 11h as an anti-osteoporosis lead compound.
Collapse
Affiliation(s)
- Zhihao Chen
- BioMedical Sciences Graduate Program (BMSGP), Chonnam National University, Gwangju, 61186, Republic of Korea
| | - Devaneyan Joseph
- Department of Chemistry, Chonnam National University, Gwangju, 61186, Republic of Korea
| | - Mina Ding
- BioMedical Sciences Graduate Program (BMSGP), Chonnam National University, Gwangju, 61186, Republic of Korea
| | - Swapnil Pandurang Bhujbal
- Institute of Pharmaceutical Science and Technology, Hanyang University, Ansan, 426-791, Republic of Korea
| | | | - Eunae Kim
- Department of Pharmacy, College of Pharmacy, Chosun University, Gwangju, 61452, Republic of Korea
| | - Sang-Wook Park
- Department of Oral Biochemistry, Dental Science Research Institute, School of Dentistry, Chonnam National University, Gwangju, 61186, Republic of Korea
| | - Sunwoo Lee
- Department of Chemistry, Chonnam National University, Gwangju, 61186, Republic of Korea.
| | - Tae-Hoon Lee
- Department of Oral Biochemistry, Dental Science Research Institute, School of Dentistry, Chonnam National University, Gwangju, 61186, Republic of Korea.
| |
Collapse
|
9
|
Zhang R, Yang W, Zhu H, Zhai J, Xue M, Zheng C. NLRC4 promotes the cGAS-STING signaling pathway by facilitating CBL-mediated K63-linked polyubiquitination of TBK1. J Med Virol 2023; 95:e29013. [PMID: 37537877 DOI: 10.1002/jmv.29013] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Revised: 06/28/2023] [Accepted: 07/20/2023] [Indexed: 08/05/2023]
Abstract
TANK-binding kinase 1 (TBK1) is crucial in producing type Ⅰ interferons (IFN-Ⅰ) that play critical functions in antiviral innate immunity. The tight regulation of TBK1, especially its activation, is very important. Here we identify NLRC4 as a positive regulator of TBK1. Ectopic expression of NLRC4 facilitates the activation of the IFN-β promoter, the mRNA levels of IFN-β, ISG54, and ISG56, and the nuclear translocation of interferon regulatory factor 3 induced by cGAS and STING. Consistently, under herpes simplex virus-1 (HSV-1) infection, knockdown or knockout of NLRC4 in BJ cells and primary peritoneal macrophages from Nlrc4-deficient (Nlrc4-/- ) mice show attenuated Ifn-β, Isg54, and Isg56 mRNA transcription, TBK1 phosphorylation, and augmented viral replications. Moreover, Nlrc4-/- mice show higher mortality upon HSV-1 infection. Mechanistically, NLRC4 facilitates the interaction between TBK1 and the E3 ubiquitin ligase CBL to enhance the K63-linked polyubiquitination of TBK1. Our study elucidates a previously uncharacterized function for NLRC4 in upregulating the cGAS-STING signaling pathway and antiviral innate immunity.
Collapse
Affiliation(s)
- Rongzhao Zhang
- Department of Immunology, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, China
| | - Wenxian Yang
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, China
| | - Huifang Zhu
- Neonatal/Pediatric Intensive Care Unit, Children's Medical Center, First Affiliated Hospital of Gannan Medical University, Ganzhou, China
| | - Jingbo Zhai
- Key Laboratory of Zoonose Prevention and Control at Universities of Inner Mongolia Autonomous Region, Medical College, Inner Mongolia Minzu University, Tongliao, China
| | - Mengzhou Xue
- Department of Cerebrovascular Diseases, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Chunfu Zheng
- Department of Immunology, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, China
- Department of Microbiology, Immunology, and Infectious Diseases, University of Calgary, Calgary, Alberta, Canada
| |
Collapse
|
10
|
Zhen H, Huang M, Zheng M, Gao L, Guo Z, Pang Q, Jin G, Zhou Z. WTAP regulates stem cells via TRAF6 to maintain planarian homeostasis and regeneration. Int J Biol Macromol 2023:124932. [PMID: 37268082 DOI: 10.1016/j.ijbiomac.2023.124932] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Revised: 04/23/2023] [Accepted: 05/06/2023] [Indexed: 06/04/2023]
Abstract
WTAP, a highly conserved Wilms' tumor 1 interacting protein, is involved in a variety of biological processes. However, functional studies of WTAP in planarians have not been reported. In this study, we examined the spatiotemporal expression pattern of planarian DjWTAP and investigated its functions in planarians regeneration and homeostasis. Knocking-down DjWTAP resulted in severe morphological defects leading to lethality within 20 days. Silencing DjWTAP promoted the proliferation of PiwiA+ cells but impaired the lineage differentiation of epidermal, neural, digestive, and excretory cell types, suggesting a critical role for DjWTAP in stem cell self-renewal and differentiation in planarian. To further investigate the mechanisms underlying the defective differentiation, RNA-seq was employed to determine the transcriptomic alterations upon DjWTAP RNA interference. Histone 4 (H4), Histone-lysine N-methyltransferase-SETMAR like, and TNF receptor-associated factor 6 (TRAF6), were significantly upregulated in response to DjWTAP RNAi. Knocking-down TRAF6 largely rescued the defective tissue homeostasis and regeneration resulted from DjWTAP knockdown in planarians, suggesting that DjWTAP maintains planarian regeneration and homeostasis via TRAF6.
Collapse
Affiliation(s)
- Hui Zhen
- Guangdong Cardiovascular Institute, Medical Research Center, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Mujie Huang
- Guangdong Cardiovascular Institute, Medical Research Center, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Mingyue Zheng
- Guangdong Cardiovascular Institute, Medical Research Center, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Lili Gao
- Laboratory of Developmental and Evolutionary Biology, Shandong University of Technology, Zibo, China
| | - Zepeng Guo
- Guangdong Cardiovascular Institute, Medical Research Center, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Qiuxiang Pang
- Laboratory of Developmental and Evolutionary Biology, Shandong University of Technology, Zibo, China.
| | - Guoxiang Jin
- Guangdong Cardiovascular Institute, Medical Research Center, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China; Guangdong Provincial Geriatrics Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong 510080, China.
| | - Zhongjun Zhou
- Guangdong Cardiovascular Institute, Medical Research Center, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China; School of Biomedical Sciences, The University of Hong Kong, Hong Kong; Reproductive Medicine Center, The University of Hong Kong-Shenzhen Hospital, Shenzhen, CHINA.
| |
Collapse
|
11
|
Cao B, Zhao Y, Luo Q, Chen Y, Xu T, Sun Y. Vinculin B inhibits NF-κB signaling pathway by targeting MyD88 in miiuy croaker, Miichthys miiuy. FISH & SHELLFISH IMMUNOLOGY 2023; 135:108683. [PMID: 36931481 DOI: 10.1016/j.fsi.2023.108683] [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: 11/04/2022] [Revised: 02/14/2023] [Accepted: 03/11/2023] [Indexed: 06/18/2023]
Abstract
Myeloid differentiation factor 88 (MyD88) is the canonical adaptor for inflammatory signaling pathways downstream from members of the Toll-like receptor (TLR) and interleukin-1 (IL-1) receptor families, which activates the NF-κB signaling pathway and regulates immune and inflammatory responses. In this study, we found that Vinculin B (Vclb) is an inhibitor in the NF-κB signaling pathway, and its inhibitory effect was enhanced by LPS induction. Furthermore, Vclb inhibits NF-κB activation by targeting MyD88, thereby suppressing the production of inflammatory cytokines. Mechanistically, Vclb inhibits the NF-κB signaling pathway by targeting MyD88 ubiquitin-proteasome pathway. In summary, our study reveals that Vclb inhibits NF-κB signaling activation and mediates innate immunity in teleosts via the ubiquitin-proteasome pathway of MyD88.
Collapse
Affiliation(s)
- Baolan Cao
- Laboratory of Fish Molecular Immunology, College of Fisheries and Life Science, Shanghai Ocean University, Shanghai, China
| | - Yan Zhao
- Laboratory of Fish Molecular Immunology, College of Fisheries and Life Science, Shanghai Ocean University, Shanghai, China
| | - Qiang Luo
- Laboratory of Fish Molecular Immunology, College of Fisheries and Life Science, Shanghai Ocean University, Shanghai, China
| | - Ya Chen
- Laboratory of Fish Molecular Immunology, College of Fisheries and Life Science, Shanghai Ocean University, Shanghai, China
| | - Tianjun Xu
- Laboratory of Fish Molecular Immunology, College of Fisheries and Life Science, Shanghai Ocean University, Shanghai, China; Laboratory of Marine Biology and Biotechnology, Qingdao National Laboratory for Marine Science and Technology, Qingdao, China.
| | - Yuena Sun
- Laboratory of Fish Molecular Immunology, College of Fisheries and Life Science, Shanghai Ocean University, Shanghai, China; National Pathogen Collection Center for Aquatic Animals, Shanghai Ocean University, China; Key Laboratory of Freshwater Aquatic Genetic Resources, Ministry of Agriculture and Rural Affairs, Shanghai Ocean University, China.
| |
Collapse
|
12
|
Sampson C, Wang Q, Otkur W, Zhao H, Lu Y, Liu X, Piao H. The roles of E3 ubiquitin ligases in cancer progression and targeted therapy. Clin Transl Med 2023; 13:e1204. [PMID: 36881608 PMCID: PMC9991012 DOI: 10.1002/ctm2.1204] [Citation(s) in RCA: 66] [Impact Index Per Article: 33.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Revised: 02/07/2023] [Accepted: 02/13/2023] [Indexed: 03/08/2023] Open
Abstract
Ubiquitination is one of the most important post-translational modifications which plays a significant role in conserving the homeostasis of cellular proteins. In the ubiquitination process, ubiquitin is conjugated to target protein substrates for degradation, translocation or activation, dysregulation of which is linked to several diseases including various types of cancers. E3 ubiquitin ligases are regarded as the most influential ubiquitin enzyme owing to their ability to select, bind and recruit target substrates for ubiquitination. In particular, E3 ligases are pivotal in the cancer hallmarks pathways where they serve as tumour promoters or suppressors. The specificity of E3 ligases coupled with their implication in cancer hallmarks engendered the development of compounds that specifically target E3 ligases for cancer therapy. In this review, we highlight the role of E3 ligases in cancer hallmarks such as sustained proliferation via cell cycle progression, immune evasion and tumour promoting inflammation, and in the evasion of apoptosis. In addition, we summarise the application and the role of small compounds that target E3 ligases for cancer treatment along with the significance of targeting E3 ligases as potential cancer therapy.
Collapse
Affiliation(s)
- Chibuzo Sampson
- CAS Key Laboratory of Separation Science for Analytical ChemistryDalian Institute of Chemical PhysicsChinese Academy of SciencesDalianChina
- University of Chinese Academy of SciencesBeijingChina
| | - Qiuping Wang
- CAS Key Laboratory of Separation Science for Analytical ChemistryDalian Institute of Chemical PhysicsChinese Academy of SciencesDalianChina
| | - Wuxiyar Otkur
- CAS Key Laboratory of Separation Science for Analytical ChemistryDalian Institute of Chemical PhysicsChinese Academy of SciencesDalianChina
| | - Haifeng Zhao
- Department of OrthopedicsDalian Second People's HospitalDalianChina
| | - Yun Lu
- CAS Key Laboratory of Separation Science for Analytical ChemistryDalian Institute of Chemical PhysicsChinese Academy of SciencesDalianChina
- Department of StomatologyDalian Medical UniversityDalianChina
| | - Xiaolong Liu
- CAS Key Laboratory of Separation Science for Analytical ChemistryDalian Institute of Chemical PhysicsChinese Academy of SciencesDalianChina
| | - Hai‐long Piao
- CAS Key Laboratory of Separation Science for Analytical ChemistryDalian Institute of Chemical PhysicsChinese Academy of SciencesDalianChina
- University of Chinese Academy of SciencesBeijingChina
| |
Collapse
|
13
|
IFT80 negatively regulates osteoclast differentiation via association with Cbl-b to disrupt TRAF6 stabilization and activation. Proc Natl Acad Sci U S A 2022; 119:e2201490119. [PMID: 35733270 PMCID: PMC9245634 DOI: 10.1073/pnas.2201490119] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
Osteoclasts (OCs) are the sole bone resorbing cells indispensable for bone remodeling. Hence, understanding of novel signaling modulators regulating OC formation is clinically important. Intraflagellar transport (IFT) proteins are important for cilia, cell signaling, and organ development. It remains unclear whether IFT80 plays a role in OCs. This study uncovers an intriguing role of IFT80 in OCs where the ciliary protein regulates the stability of critical OC factor TRAF6 via Cbl-b and thereby contributes to the maintenance of OC numbers. These findings provide further basis for understanding and delineating the role of IFT proteins in OCs that may provide new strategies for treatment of osteolytic diseases. Excess bone loss due to increased osteoclastogenesis is a significant clinical problem. Intraflagellar transport (IFT) proteins have been reported to regulate cell growth and differentiation. The role of IFT80, an IFT complex B protein, in osteoclasts (OCs) is completely unknown. Here, we demonstrate that deletion of IFT80 in the myeloid lineage led to increased OC formation and activity accompanied by severe bone loss in mice. IFT80 regulated OC formation by associating with Casitas B-lineage lymphoma proto-oncogene-b (Cbl-b) to promote protein stabilization and proteasomal degradation of tumor necrosis factor (TNF) receptor–associated factor 6 (TRAF6). IFT80 knockdown resulted in increased ubiquitination of Cbl-b and higher TRAF6 levels, thereby hyperactivating the receptor activator of nuclear factor-κβ (NF-κβ) ligand (RANKL) signaling axis and increased OC formation. Ectopic overexpression of IFT80 rescued osteolysis in a calvarial model of bone loss. We have thus identified a negative function of IFT80 in OCs.
Collapse
|
14
|
Kuassivi ON, Abiven H, Satie AP, Cartron M, Mahé D, Aubry F, Mathieu R, Rebours V, Le Tortorec A, Dejucq-Rainsford N. Human Testicular Germ Cells, a Reservoir for Zika Virus, Lack Antiviral Response Upon Zika or Poly(I:C) Exposure. Front Immunol 2022; 13:909341. [PMID: 35784373 PMCID: PMC9248283 DOI: 10.3389/fimmu.2022.909341] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Accepted: 05/19/2022] [Indexed: 11/13/2022] Open
Abstract
Zika virus (ZIKV) is an emerging teratogenic arbovirus that persists in semen and is sexually transmitted. We previously demonstrated that ZIKV infects the human testis and persists in testicular germ cells (TGCs) for several months after patients’ recovery. To decipher the mechanisms underlying prolonged ZIKV replication in TGCs, we compared the innate immune response of human testis explants and isolated TGCs to ZIKV and to Poly(I:C), a viral RNA analog. Our results demonstrate the weak innate responses of human testis to both ZIKV and Poly(I:C) as compared with other tissues or species. TGCs failed to up-regulate antiviral effectors and type I IFN upon ZIKV or Poly(I:C) stimulation, which might be due to a tight control of PRR signaling, as evidenced by the absence of activation of the downstream effector IRF3 and elevated expression of repressors. Importantly, exogenous IFNβ boosted the innate immunity of TGCs and inhibited ZIKV replication in the testis ex vivo, raising hopes for the prevention of ZIKV infection and persistence in this organ.
Collapse
Affiliation(s)
- Ohiniba Nadège Kuassivi
- Université de Rennes, INSERM, Ecole des Hautes Etudes en Santé Publique (EHESP), Institut de recherche en santé, environnement et travail (Irset) – UMR_S1085, Rennes, France
| | - Hervé Abiven
- Université de Rennes, INSERM, Ecole des Hautes Etudes en Santé Publique (EHESP), Institut de recherche en santé, environnement et travail (Irset) – UMR_S1085, Rennes, France
| | - Anne-Pascale Satie
- Université de Rennes, INSERM, Ecole des Hautes Etudes en Santé Publique (EHESP), Institut de recherche en santé, environnement et travail (Irset) – UMR_S1085, Rennes, France
| | - Matéo Cartron
- Université de Rennes, INSERM, Ecole des Hautes Etudes en Santé Publique (EHESP), Institut de recherche en santé, environnement et travail (Irset) – UMR_S1085, Rennes, France
| | - Dominique Mahé
- Université de Rennes, INSERM, Ecole des Hautes Etudes en Santé Publique (EHESP), Institut de recherche en santé, environnement et travail (Irset) – UMR_S1085, Rennes, France
| | - Florence Aubry
- Université de Rennes, INSERM, Ecole des Hautes Etudes en Santé Publique (EHESP), Institut de recherche en santé, environnement et travail (Irset) – UMR_S1085, Rennes, France
| | - Romain Mathieu
- Service d’Urologie, Centre Hospitalier Universitaire de Rennes, Rennes, France
| | - Valérie Rebours
- Unité de Coordination Hospitalière des Prélèvements d’Organes et de Tissus, Centre Hospitalier Universitaire de Rennes, Rennes, France
| | - Anna Le Tortorec
- Université de Rennes, INSERM, Ecole des Hautes Etudes en Santé Publique (EHESP), Institut de recherche en santé, environnement et travail (Irset) – UMR_S1085, Rennes, France
| | - Nathalie Dejucq-Rainsford
- Université de Rennes, INSERM, Ecole des Hautes Etudes en Santé Publique (EHESP), Institut de recherche en santé, environnement et travail (Irset) – UMR_S1085, Rennes, France
- *Correspondence: Nathalie Dejucq-Rainsford,
| |
Collapse
|
15
|
Chen Z, Zeng Y, Wei Y, Wang Q, Liu M, Zhang B, Liu J, Zhu Q, Xu S. Influenza D virus Matrix protein 1 restricts the type I interferon response by degrading TRAF6. Virology 2022; 568:1-11. [DOI: 10.1016/j.virol.2022.01.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Revised: 01/06/2022] [Accepted: 01/09/2022] [Indexed: 01/04/2023]
|
16
|
TRAF6 Phosphorylation Prevents Its Autophagic Degradation and Re-Shapes LPS-Triggered Signaling Networks. Cancers (Basel) 2021; 13:cancers13143618. [PMID: 34298830 PMCID: PMC8303406 DOI: 10.3390/cancers13143618] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Revised: 07/07/2021] [Accepted: 07/13/2021] [Indexed: 11/20/2022] Open
Abstract
Simple Summary Here, we reveal that basal turnover and autophagy-induced decay of the ubiquitin E3 ligase TRAF6 is antagonized by IKKε-mediated phosphorylation at five serines. Phosphoproteomic experiments show that TRAF6 and its phosphorylation contribute to the remodeling of LPS- and autophagyinduced signaling networks, revealing an intricate link between inflammatory and metabolic processes that are frequently dysregulated in cancer. Abstract The ubiquitin E3 ligase TNF Receptor Associated Factor 6 (TRAF6) participates in a large number of different biological processes including innate immunity, differentiation and cell survival, raising the need to specify and shape the signaling output. Here, we identify a lipopolysaccharide (LPS)-dependent increase in TRAF6 association with the kinase IKKε (inhibitor of NF-κB kinase subunit ε) and IKKε-mediated TRAF6 phosphorylation at five residues. The reconstitution of TRAF6-deficient cells, with TRAF6 mutants representing phosphorylation-defective or phospho-mimetic TRAF6 variants, showed that the phospho-mimetic TRAF6 variant was largely protected from basal ubiquitin/proteasome-mediated degradation, and also from autophagy-mediated decay in autolysosomes induced by metabolic perturbation. In addition, phosphorylation of TRAF6 and its E3 ligase function differentially shape basal and LPS-triggered signaling networks, as revealed by phosphoproteome analysis. Changes in LPS-triggered phosphorylation networks of cells that had experienced autophagy are partially dependent on TRAF6 and its phosphorylation status, suggesting an involvement of this E3 ligase in the interplay between metabolic and inflammatory circuits.
Collapse
|
17
|
Chondroprotection and Molecular Mechanism of Action of Phytonutraceuticals on Osteoarthritis. Molecules 2021; 26:molecules26082391. [PMID: 33924083 PMCID: PMC8074261 DOI: 10.3390/molecules26082391] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Revised: 04/16/2021] [Accepted: 04/17/2021] [Indexed: 02/06/2023] Open
Abstract
Osteoarthritis (OA) is a degenerative joint disease and an important cause of incapacitation. There is a lack of drugs and effective treatments that stop or slow the OA progression. Modern pharmacological treatments, such as analgesics, have analgesic effects but do not affect the course of OA. Long-term use of these drugs can lead to serious side effects. Given the OA nature, it is likely that lifelong treatment will be required to stop or slow its progression. Therefore, there is an urgent need for disease-modifying OA treatments that are also safe for clinical use over long periods. Phytonutraceuticals are herbal products that provide a therapeutic effect, including disease prevention, which not only have favorable safety characteristics but may have an alleviating effect on the OA and its symptoms. An estimated 47% of OA patients use alternative drugs, including phytonutraceuticals. The review studies the efficacy and action mechanism of widely used phytonutraceuticals, analyzes the available experimental and clinical data on the effect of some phytonutraceuticals (phytoflavonoids, polyphenols, and bioflavonoids) on OA, and examines the known molecular effect and the possibility of their use for chondroprotection.
Collapse
|
18
|
Budroni V, Versteeg GA. Negative Regulation of the Innate Immune Response through Proteasomal Degradation and Deubiquitination. Viruses 2021; 13:584. [PMID: 33808506 PMCID: PMC8066222 DOI: 10.3390/v13040584] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2021] [Revised: 03/26/2021] [Accepted: 03/27/2021] [Indexed: 12/25/2022] Open
Abstract
The rapid and dynamic activation of the innate immune system is achieved through complex signaling networks regulated by post-translational modifications modulating the subcellular localization, activity, and abundance of signaling molecules. Many constitutively expressed signaling molecules are present in the cell in inactive forms, and become functionally activated once they are modified with ubiquitin, and, in turn, inactivated by removal of the same post-translational mark. Moreover, upon infection resolution a rapid remodeling of the proteome needs to occur, ensuring the removal of induced response proteins to prevent hyperactivation. This review discusses the current knowledge on the negative regulation of innate immune signaling pathways by deubiquitinating enzymes, and through degradative ubiquitination. It focusses on spatiotemporal regulation of deubiquitinase and E3 ligase activities, mechanisms for re-establishing proteostasis, and degradation through immune-specific feedback mechanisms vs. general protein quality control pathways.
Collapse
Affiliation(s)
| | - Gijs A. Versteeg
- Max Perutz Labs, Department of Microbiology, Immunobiology, and Genetics, University of Vienna, Vienna Biocenter (VBC), 1030 Vienna, Austria;
| |
Collapse
|
19
|
Hu CF, Liao XY, Xu DD, Ruan YB, Gao FG. K48-Linked Ubiquitination Contributes to Nicotine-Augmented Bone Marrow-Derived Dendritic-Cell-Mediated Adaptive Immunity. Vaccines (Basel) 2021; 9:vaccines9030278. [PMID: 33808531 PMCID: PMC8003133 DOI: 10.3390/vaccines9030278] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2021] [Revised: 03/15/2021] [Accepted: 03/17/2021] [Indexed: 01/23/2023] Open
Abstract
K48-linked ubiquitination determining antigen degradation and the endosomal recruitments of p97 and Sec61 plays vital roles in dendritic cell (DC) cross-presentation. Our previous studies revealed that nicotine treatment increases bone marrow-derived dendritic cell (BM-DC) cross-presentation and promotes BM-DC-based cytotoxic T lymphocyte (CTL) priming. But the effect of nicotine on K48-linked ubiquitination and the mechanism of nicotine-increased BM-DC cross-presentation are still uncertain. In this study, we first demonstrated that ex vivo nicotine administration obviously increased K48-linked ubiquitination in BM-DC. Then, we found that K48-linked ubiquitination was essential for nicotine-augmented cross-presentation, BM-DC-based CTL priming, and thereby the superior cytolytic capacity of DC-activated CTL. Importantly, K48-linked ubiquitination was verified to be necessary for nicotine-augmented endosomal recruitments of p97 and Sec61. Importantly, mannose receptor (MR), which is an important antigenic receptor for cross-presentation, was exactly catalyzed with K48-linked ubiquitination by the treatment with nicotine. Thus, these data suggested that K48-linked ubiquitination contributes to the superior adaptive immunity of nicotine-administrated BM-DC. Regulating K48-linked ubiquitination might have therapeutic potential for DC-mediated immune therapy.
Collapse
Affiliation(s)
- Chun Fang Hu
- Department of Basic Medical Sciences, School of Medicine, Xiamen University, Xiamen 361102, China; (C.F.H.); (X.Y.L.); (D.D.X.)
| | - Xiao Yan Liao
- Department of Basic Medical Sciences, School of Medicine, Xiamen University, Xiamen 361102, China; (C.F.H.); (X.Y.L.); (D.D.X.)
| | - Dan Dan Xu
- Department of Basic Medical Sciences, School of Medicine, Xiamen University, Xiamen 361102, China; (C.F.H.); (X.Y.L.); (D.D.X.)
| | - Yi Bin Ruan
- Technology Center, China Tobacco Guizhou Industrial Co., Ltd., Guiyang 550003, China
- Correspondence: (Y.B.R.); (F.G.G.)
| | - Feng Guang Gao
- Department of Basic Medical Sciences, School of Medicine, Xiamen University, Xiamen 361102, China; (C.F.H.); (X.Y.L.); (D.D.X.)
- Correspondence: (Y.B.R.); (F.G.G.)
| |
Collapse
|
20
|
Di Q, Zhao X, Zhang R, Ma X, Liang X, Li X, Gao J, Tang H, Chen W, Xiao W. Novel clerodane-type diterpenoid Cintelactone A suppresses lipopolysaccharide -induced inflammation by promoting ubiquitination, proteasomal degradation of TRAF6. Pharmacol Res 2021; 164:105386. [PMID: 33352228 DOI: 10.1016/j.phrs.2020.105386] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Revised: 12/13/2020] [Accepted: 12/14/2020] [Indexed: 12/29/2022]
Abstract
Cellular inflammation is the underlying cause of several diseases and development of a safe and effective anti-inflammatory drug is need-of-the hour for treatment of diseases like lung inflammation. Callicarpa integerrima Champ. is a well-known herbal medicine with hemostatic and anti-inflammatory functions. However, the exact ingredient exhibiting anti-inflammatory activity in C. integerrima Champ. is largely unknown. Here, we first isolated, purified and characterized a novel clerodane-type diterpenoid Cintelactone A (CA) from C. integerrima Champ. We demonstrated that CA could significantly inhibit lipopolysaccharide (LPS)-induced pro-inflammatory cytokines and mediators production both in mouse peritoneal macrophages and THP1 cells. Consistently, CA also relieved inflammation and reduced LPS-induced lung injury in mice. We systematically elucidated the mechanism of action as well. CA interacted with Arg78 of tumor necrosis factor receptor-associated factor 6 (TRAF6) by hydrogen bonding. It further promoted the K48-linked ubiquitination and proteasomal degradation of TRAF6, and suppressed the activation of NF-κB and MAPKs signaling pathways. Collectively, our study reveals that new clerodane-type diterpenoid CA suppresses LPS-induced inflammation by promoting TRAF6 degradation, suggesting that CA as the potential therapeutic candidate for the treatment of inflammation associated diseases.
Collapse
Affiliation(s)
- Qianqian Di
- Guangdong Provincial Key Laboratory of Regional Immunity and Diseases, Department of Immunology, Shenzhen University School of Medicine, Shenzhen, 518060, China
| | - Xibao Zhao
- Guangdong Provincial Key Laboratory of Regional Immunity and Diseases, Department of Immunology, Shenzhen University School of Medicine, Shenzhen, 518060, China
| | - Ruihan Zhang
- Key Laboratory of Medicinal Chemistry for Natural Resource of Ministry of Education and Yunnan Province, School of Chemical Science and Technology, Yunnan University, Kunming, 650091, China
| | - Xingyu Ma
- Guangdong Provincial Key Laboratory of Regional Immunity and Diseases, Department of Immunology, Shenzhen University School of Medicine, Shenzhen, 518060, China
| | - Xinxin Liang
- Key Laboratory of Medicinal Chemistry for Natural Resource of Ministry of Education and Yunnan Province, School of Chemical Science and Technology, Yunnan University, Kunming, 650091, China
| | - Xiaoli Li
- Key Laboratory of Medicinal Chemistry for Natural Resource of Ministry of Education and Yunnan Province, School of Chemical Science and Technology, Yunnan University, Kunming, 650091, China
| | - Junbo Gao
- Key Laboratory of Medicinal Chemistry for Natural Resource of Ministry of Education and Yunnan Province, School of Chemical Science and Technology, Yunnan University, Kunming, 650091, China
| | - Haimei Tang
- Guangdong Provincial Key Laboratory of Regional Immunity and Diseases, Department of Immunology, Shenzhen University School of Medicine, Shenzhen, 518060, China
| | - Weilin Chen
- Guangdong Provincial Key Laboratory of Regional Immunity and Diseases, Department of Immunology, Shenzhen University School of Medicine, Shenzhen, 518060, China.
| | - Weilie Xiao
- Key Laboratory of Medicinal Chemistry for Natural Resource of Ministry of Education and Yunnan Province, School of Chemical Science and Technology, Yunnan University, Kunming, 650091, China.
| |
Collapse
|
21
|
TRIM Proteins in Inflammation: from Expression to Emerging Regulatory Mechanisms. Inflammation 2021; 44:811-820. [PMID: 33415537 DOI: 10.1007/s10753-020-01394-8] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2020] [Revised: 10/07/2020] [Accepted: 12/07/2020] [Indexed: 02/06/2023]
Abstract
Inflammation is an immune response to exogenous or endogenous insults that helps to maintain the tissue homeostasis under stressful conditions. Depending on the differential types of insults, inflammation is classified into microbial, autoimmune, metabolic, allergic, and physical inflammation. With regard to its involvement in the pathogenesis of most of human diseases, dissecting the key molecules in the regulation of inflammatory process is vital for the prevention and therapeutics of human diseases. Tripartite motif (TRIM) proteins are a versatile family of E3 ligases, which are composed of > 80 distinct members in humans recognized for their roles in antiviral responses. Recently, a large number of studies have shown the regulatory roles of TRIM proteins in mediating the inflammation. Herein in this review, we discuss the aberrations of TRIM proteins in autoimmune and autoinflammatory diseases, with a focus on the regulation of different components of inflammatory process, including inflammasome, NF-κB signaling, type I IFN (interferon) production, and Th1/Th17 cell differentiation. Importantly, elucidation of the mechanism underlying the regulation of inflammation by TRIMs provides insights into the use of TRIMs as therapeutic targets for disease treatment.
Collapse
|
22
|
Zhou C, Lu C, Pu H, Li D, Zhang L. TRAF6 promotes IL-4-induced M2 macrophage activation by stabilizing STAT6. Mol Immunol 2020; 127:223-229. [DOI: 10.1016/j.molimm.2020.09.001] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Revised: 08/27/2020] [Accepted: 09/02/2020] [Indexed: 12/13/2022]
|
23
|
Molecular and Cellular Pathways Contributing to Joint Damage in Rheumatoid Arthritis. Mediators Inflamm 2020; 2020:3830212. [PMID: 32256192 PMCID: PMC7103059 DOI: 10.1155/2020/3830212] [Citation(s) in RCA: 120] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2020] [Accepted: 02/24/2020] [Indexed: 01/19/2023] Open
Abstract
Rheumatoid arthritis is a chronic autoimmune syndrome associated with several genetic, epigenetic, and environmental factors affecting the articular joints contributing to cartilage and bone damage. Although etiology of this disease is not clear, several immune pathways, involving immune (T cells, B cells, dendritic cells, macrophages, and neutrophils) and nonimmune (fibroblasts and chondrocytes) cells, participate in the secretion of many proinflammatory cytokines, chemokines, proteases (MMPs, ADAMTS), and other matrix lysing enzymes that could disturb the immune balance leading to cartilage and bone damage. The presence of autoantibodies preceding the clinical onset of arthritis and the induction of bone erosion early in the disease course clearly suggest that initiation events damaging the cartilage and bone start very early during the autoimmune phase of the arthritis development. During this process, several signaling molecules (RANKL-RANK, NF-κB, MAPK, NFATc1, and Src kinase) are activated in the osteoclasts, cells responsible for bone resorption. Hence, comprehensive knowledge on pathogenesis is a prerequisite for prevention and development of targeted clinical treatment for RA patients that can restore the immune balance improving clinical therapy.
Collapse
|
24
|
Fan Q, Wang Q, Cai R, Yuan H, Xu M. The ubiquitin system: orchestrating cellular signals in non-small-cell lung cancer. Cell Mol Biol Lett 2020; 25:1. [PMID: 31988639 PMCID: PMC6966813 DOI: 10.1186/s11658-019-0193-6] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2019] [Accepted: 11/25/2019] [Indexed: 02/07/2023] Open
Abstract
The ubiquitin system, known as a common feature in eukaryotes, participates in multiple cellular processes, such as signal transduction, cell-cycle progression, receptor trafficking and endocytosis, and even the immune response. In lung cancer, evidence has revealed that aberrant events in ubiquitin-mediated processes can cause a variety of pathological outcomes including tumorigenesis and metastasis. Likewise, ubiquitination on the core components contributing to the activity of cell signaling controls bio-signal turnover and cell final destination. Given this, inhibitors targeting the ubiquitin system have been developed for lung cancer therapies and have shown great prospects for clinical application. However, the exact biological effects and physiological role of the drugs used in lung cancer therapies are still not clearly elucidated, which might seriously impede the progress of treatment. In this work, we summarize current research advances in cell signal regulation processes mediated through the ubiquitin system during the development of lung cancer, with the hope of improving the therapeutic effects by means of aiming at efficient targets.
Collapse
Affiliation(s)
- Qiang Fan
- 1Department of Oncology, Shanghai 9th People's Hospital, Shanghai Jiao Tong University School of Medicine, 280 Mohe Road, Shanghai, China.,2Department of General Surgery, Shanghai 9th People's Hospital, Shanghai Jiao Tong University School of Medicine, 280 Mohe Road, Shanghai, China
| | - Qian Wang
- 1Department of Oncology, Shanghai 9th People's Hospital, Shanghai Jiao Tong University School of Medicine, 280 Mohe Road, Shanghai, China
| | - Renjie Cai
- 1Department of Oncology, Shanghai 9th People's Hospital, Shanghai Jiao Tong University School of Medicine, 280 Mohe Road, Shanghai, China.,2Department of General Surgery, Shanghai 9th People's Hospital, Shanghai Jiao Tong University School of Medicine, 280 Mohe Road, Shanghai, China
| | - Haihua Yuan
- 1Department of Oncology, Shanghai 9th People's Hospital, Shanghai Jiao Tong University School of Medicine, 280 Mohe Road, Shanghai, China
| | - Ming Xu
- 1Department of Oncology, Shanghai 9th People's Hospital, Shanghai Jiao Tong University School of Medicine, 280 Mohe Road, Shanghai, China
| |
Collapse
|