1
|
Ma Y, Jia R, Chen S, Ma J, Yin L, Pan X, He Y, Wu T, Zhao Z, Ma L, Wu S, Wang H, Liang G, Huang S, Sun X. Ubiquitin‐Proteasome System in Periodontitis: Mechanisms and Clinical Implications. Cell Prolif 2024. [DOI: 10.1111/cpr.13781] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Accepted: 11/09/2024] [Indexed: 01/03/2025] Open
Abstract
ABSTRACTThe progression of periodontitis, a bacteria‐driven inflammatory and bone‐destructive disease, involves myriad cellular and molecular mechanisms. Protein regulation significantly influences the pathogenesis and management of periodontitis. However, research regarding its regulatory role in periodontitis remains relatively limited. The ubiquitin‐proteasome system (UPS), which mainly involves ubiquitination by E3 ubiquitin ligases (E3s) and deubiquitination by deubiquitinating enzymes (DUBs), is the primary intracellular and non‐lysosomal mechanism of protein degradation. Recent studies have provided compelling evidence to support the involvement of UPS in periodontitis progression. Increasing evidence indicated that E3s, such as CUL3, Nedd4‐2, Synoviolin, FBXL19, PDLIM2, TRIMs and TRAFs, modulate inflammatory responses and bone resorption in periodontitis through multiple classical signalling pathways, including NLRP3, GSDMD, NF‐κB, Wnt/β‐catenin and Nrf2. Meanwhile, DUBs, including OTUD1, A20, CYLD, UCH‐L1 and USPs, also broadly modulate periodontitis progression by regulating signalling pathways such as NF‐κB, Wnt/β‐catenin, NLRP3, and BMP2. Therefore, the modulation of E3s and DUBs has proven to be an effective therapy against periodontitis. This review provides a comprehensive overview of the regulatory role of ubiquitinating and deubiquitinating enzymes in periodontitis progression and the underlying mechanisms. Finally, we summarise several chemical and genetic methods that regulate UPS enzymes and pave the way for the development of targeted therapies for periodontitis.
Collapse
Affiliation(s)
- Yilin Ma
- Institute of Stomatology, School and Hospital of Stomatology Wenzhou Medical University Wenzhou China
| | - Ruiwei Jia
- Institute of Stomatology, School and Hospital of Stomatology Wenzhou Medical University Wenzhou China
| | - Shuhong Chen
- Institute of Stomatology, School and Hospital of Stomatology Wenzhou Medical University Wenzhou China
| | - Jun Ma
- Institute of Stomatology, School and Hospital of Stomatology Wenzhou Medical University Wenzhou China
| | - Lei Yin
- Institute of Stomatology, School and Hospital of Stomatology Wenzhou Medical University Wenzhou China
| | - Xingbei Pan
- Institute of Stomatology, School and Hospital of Stomatology Wenzhou Medical University Wenzhou China
| | - Yunuo He
- Institute of Stomatology, School and Hospital of Stomatology Wenzhou Medical University Wenzhou China
| | - Tong Wu
- Institute of Stomatology, School and Hospital of Stomatology Wenzhou Medical University Wenzhou China
| | - Zheyu Zhao
- Institute of Stomatology, School and Hospital of Stomatology Wenzhou Medical University Wenzhou China
| | - Lulu Ma
- Institute of Stomatology, School and Hospital of Stomatology Wenzhou Medical University Wenzhou China
| | - Shengzhuang Wu
- Institute of Stomatology, School and Hospital of Stomatology Hangzhou Medical College Zhejiang Hangzhou China
| | - Huining Wang
- Department of Periodontics, School and Hospital of Stomatology Wenzhou Medical University Wenzhou China
| | - Guang Liang
- Institute of Stomatology, School and Hospital of Stomatology Hangzhou Medical College Zhejiang Hangzhou China
| | - Shengbin Huang
- Institute of Stomatology, School and Hospital of Stomatology Wenzhou Medical University Wenzhou China
- Department of Prosthodontics, School and Hospital of Stomatology Wenzhou Medical University Wenzhou China
| | - Xiaoyu Sun
- Institute of Stomatology, School and Hospital of Stomatology Wenzhou Medical University Wenzhou China
- Department of Periodontics, School and Hospital of Stomatology Wenzhou Medical University Wenzhou China
| |
Collapse
|
2
|
Lu H, Li Z, Zhu L, Xu P, Wang H, Li Y, Zhao W. Fabrication and Temporal Dependency Osteogenic Regulation of Dual-Scale Hierarchical Microstructures on Medical Metal Surface. Adv Healthc Mater 2024; 13:e2402369. [PMID: 39175381 DOI: 10.1002/adhm.202402369] [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: 07/13/2024] [Revised: 08/09/2024] [Indexed: 08/24/2024]
Abstract
The structural characteristics at the interface of bone implants can guide biological regulation. In this study, a dual-scale hierarchical microstructure is proposed and customized using hybrid machining to achieve temporal dependency osteogenic regulation. It is observed that osteoblasts induced by dual-scale hierarchical structure exhibit adequate protrusion development and rapid cell attachment through the modulation of mechanical forces in the cell growth environment, and further promot the upregulation of the cell membrane receptor PDGFR-α, which is related to cell proliferation. Afterward, transcriptomic analysis reveals that during the differentiation stage, the DSH structure regulates cellular signaling cascades primarily through integrin adhesion mechanisms and then accelerates osteogenic differentiation by activating the TGF-β pathway and cAMP signaling pathway. Furthermore, the calcium nodules are preferentially deposited within the lower honeycomb-like channels, thereby endowing the proposed dual-scale hierarchical structure with the potential to induce oriented deposition and improve the long-term stability of the implant.
Collapse
Affiliation(s)
- Hao Lu
- School of Mechanical Engineering and Automation, Northeastern University, Shenyang, 110819, China
| | - Zhijun Li
- Department of Developmental Cell Biology, Key Laboratory of Cell Biology, Ministry of Public Health, and Key Laboratory of Medical Cell Biology, Ministry of Education, China Medical University, Shenyang, China
| | - Lida Zhu
- School of Mechanical Engineering and Automation, Northeastern University, Shenyang, 110819, China
| | - Peihua Xu
- School of Mechanical Engineering and Automation, Northeastern University, Shenyang, 110819, China
| | - Hai Wang
- Shenyang Lebuy Vacuum Tech. Co., Ltd, Shenyang, Liaoning, China
| | - Yonghao Li
- Shenyang Lebuy Vacuum Tech. Co., Ltd, Shenyang, Liaoning, China
| | - Weidong Zhao
- Department of Developmental Cell Biology, Key Laboratory of Cell Biology, Ministry of Public Health, and Key Laboratory of Medical Cell Biology, Ministry of Education, China Medical University, Shenyang, China
| |
Collapse
|
3
|
Yang X, Sun J, Ning Y, Wang J, Xu J, Zhang S. Role of the CTCF/p300 axis in osteochondrogenic-like differentiation of polyploid giant cancer cells with daughter cells. Cell Commun Signal 2024; 22:546. [PMID: 39548585 PMCID: PMC11566548 DOI: 10.1186/s12964-024-01933-y] [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: 07/01/2024] [Accepted: 11/07/2024] [Indexed: 11/18/2024] Open
Abstract
BACKGROUND Polyploid giant cancer cells (PGCCs) have properties of cancer stem cells (CSCs). PGCCs with daughter cells (PDCs) undergo epithelial-mesenchymal transition and show enhanced cellular plasticity. This study aimed to elucidate the mechanisms underlying the osteo/chondrogenic-like differentiation of PDCs, which may be exploited therapeutically by transdifferentiation into post-mitotic and functional cells. METHODS Cobalt chloride was used to induce PGCC formation in MDA-MB-231 and HEY cells, and PDCs were cultured in osteo/chondrogenic differentiation media. Alcian blue staining was used to confirm osteo/chondrogenic differentiation, and the cell cycle was detected using flow cytometry. The expression of osteo/chondrogenic differentiation-related proteins was compared, and a co-immunoprecipitation assay was used to demonstrate the interactions between proteins. Bioinformatic analysis was used to explore the regulatory mechanism of osteo/chondrogenic differentiation, and a dual-luciferase reporter assay was performed to validate the interaction between transcriptional factors and target genes. Animal xenograft models were used to confirm the osteo/chondrogenic differentiation of PDCs. RESULTS When cultured in osteo/chondrogenic medium, the stemness of PDCs decreased, and the expression of osteo/chondrogenic-related markers increased. This osteo/chondrogenic-like process was regulated by the transforming growth factor-β pathway in a time-dependent manner. A concurrent increase in the expression of histone acetyltransferase p300 and the transcription factor CCCTC-binding factor (CTCF) was observed. Co-immunoprecipitation assays revealed that p300 acetylated the osteo/chondrogenic marker RUNT-related transcription factor 2 (RUNX2). Analysis of chromatin immunoprecipitation sequencing datasets revealed that both CTCF and histone H3 lysine 27 acetylation (H3K27ac) were enriched in the promoter region of E1A-associated protein p300 (P300). The four predicted binding sites for CTCF and P300 were validated using dual-luciferase reporter assays. We examined the interaction between CTCF and H3K27ac and found that these two proteins had a combined effect on the transactivation of P300. CONCLUSION CTCF, in synergy with H3K27ac, amplified the expression of P300, facilitating acetyl group transfer to RUNX2. This acetylation stabilized RUNX2 and promoted osteo/chondrogenic differentiation, thereby reducing the incidence of PDC malignancies.
Collapse
Affiliation(s)
- Xiaohui Yang
- Nankai University School of Medicine, Nankai University, Tianjin, 300071, P.R. China
| | - Jie Sun
- Nankai University School of Medicine, Nankai University, Tianjin, 300071, P.R. China
- Department of Pathology, Tianjin Union Medical Center, Tianjin, 300121, P.R. China
| | - Yidi Ning
- Nankai University School of Medicine, Nankai University, Tianjin, 300071, P.R. China
| | - Jiangping Wang
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, P.R. China
| | - Jing Xu
- Department of General Surgery, Tianjin Union Medical Center, Nankai University, Tianjin, China
| | - Shiwu Zhang
- Department of Pathology, Tianjin Union Medical Center, Tianjin, 300121, P.R. China.
| |
Collapse
|
4
|
Xu Y, Chang L, Chen Y, Dan Z, Zhou L, Tang J, Deng L, Tang G, Li C. USP26 Combats Age-Related Declines in Self-Renewal and Multipotent Differentiation of BMSC by Maintaining Mitochondrial Homeostasis. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2406428. [PMID: 39377219 PMCID: PMC11600297 DOI: 10.1002/advs.202406428] [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: 06/11/2024] [Revised: 09/24/2024] [Indexed: 10/09/2024]
Abstract
Age-related declines in self-renewal and multipotency of bone marrow mesenchymal stem cells (BMSCs) limit their applications in tissue engineering and clinical therapy. Thus, understanding the mechanisms behind BMSC senescence is crucial for maintaining the rejuvenation and multipotent differentiation capabilities of BMSCs. This study reveals that impaired USP26 expression in BMSCs leads to mitochondrial dysfunction, ultimately resulting in aging and age-related declines in the self-renewal and multipotency of BMSCs. Specifically, decreased USP26 expression results in decreased protein levels of Sirtuin 2 due to its ubiquitination degradation, which leads to mitochondrial dysfunction in BMSCs and ultimately resulting in aging and age-related declines in self-renewal and multilineage differentiation potentials. Additionally, decreased USP26 expression in aging BMSCs is a result of dampened hypoxia-inducible factor 1α (HIF-1α) expression. HIF-1α facilitates USP26 transcriptional expression by increasing USP26 promoter activity through binding to the -191 - -198 bp and -262 - -269 bp regions on the USP26 promoter. Therefore, the identification of USP26 as being correlated with aging and age-related declines in self-renewal and multipotency of BMSCs, along with understanding its expression and action mechanisms, suggests that USP26 represents a novel therapeutic target for combating aging and age-related declines in the self-renewal and multipotent differentiation of BMSCs.
Collapse
Affiliation(s)
- Yiming Xu
- Department of OrthopedicsShanghai Key Laboratory for Prevention and Treatment of Bone and Joint DiseasesShanghai Institute of Traumatology and OrthopedicsRuijin HospitalShanghai Jiao Tong University School of Medicine197 Ruijin 2nd RoadShanghai200025China
| | - Leilei Chang
- Department of OrthopedicsShanghai Key Laboratory for Prevention and Treatment of Bone and Joint DiseasesShanghai Institute of Traumatology and OrthopedicsRuijin HospitalShanghai Jiao Tong University School of Medicine197 Ruijin 2nd RoadShanghai200025China
| | - Yong Chen
- Department of OrthopedicsKunshan Hospital of Chinese MedicineAffiliated Hospital of Yangzhou UniversitySuzhouJiangsu Province215300China
- Institute of Traumatology and OrthopedicsKunshan Hospital of Chinese MedicineAffiliated Hospital of Yangzhou UniversitySuzhouJiangsu Province215300China
| | - Zhou Dan
- Department of OrthopedicsShanghai Key Laboratory for Prevention and Treatment of Bone and Joint DiseasesShanghai Institute of Traumatology and OrthopedicsRuijin HospitalShanghai Jiao Tong University School of Medicine197 Ruijin 2nd RoadShanghai200025China
| | - Li Zhou
- Department of OrthopedicsKunshan Hospital of Chinese MedicineAffiliated Hospital of Yangzhou UniversitySuzhouJiangsu Province215300China
- Institute of Traumatology and OrthopedicsKunshan Hospital of Chinese MedicineAffiliated Hospital of Yangzhou UniversitySuzhouJiangsu Province215300China
| | - Jiyuan Tang
- Department of OrthopedicsShanghai Key Laboratory for Prevention and Treatment of Bone and Joint DiseasesShanghai Institute of Traumatology and OrthopedicsRuijin HospitalShanghai Jiao Tong University School of Medicine197 Ruijin 2nd RoadShanghai200025China
| | - Lianfu Deng
- Department of OrthopedicsShanghai Key Laboratory for Prevention and Treatment of Bone and Joint DiseasesShanghai Institute of Traumatology and OrthopedicsRuijin HospitalShanghai Jiao Tong University School of Medicine197 Ruijin 2nd RoadShanghai200025China
| | - Guoqing Tang
- Department of OrthopedicsKunshan Hospital of Chinese MedicineAffiliated Hospital of Yangzhou UniversitySuzhouJiangsu Province215300China
- Institute of Traumatology and OrthopedicsKunshan Hospital of Chinese MedicineAffiliated Hospital of Yangzhou UniversitySuzhouJiangsu Province215300China
| | - Changwei Li
- Department of OrthopedicsShanghai Key Laboratory for Prevention and Treatment of Bone and Joint DiseasesShanghai Institute of Traumatology and OrthopedicsRuijin HospitalShanghai Jiao Tong University School of Medicine197 Ruijin 2nd RoadShanghai200025China
| |
Collapse
|
5
|
Monroe DG, Javeed N, Rowsey JL, Ruan M, McCabe CE, Piatkowski BT, Roy A, Bobbili MR, Grillari J, Khosla S. Isolation and characterization of bone mesenchymal cell small extracellular vesicles using a novel mouse model. J Bone Miner Res 2024; 39:1633-1643. [PMID: 39173022 PMCID: PMC11523127 DOI: 10.1093/jbmr/zjae135] [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: 05/09/2024] [Revised: 08/06/2024] [Accepted: 08/21/2024] [Indexed: 08/24/2024]
Abstract
Extracellular vesicles (EVs) are key mediators of cell-cell communication and are involved in transferring specific biomolecular cargo to recipient cells to regulate their physiological functions. A major challenge in the understanding of EV function in vivo is the difficulty ascertaining the origin of the EV particles. The recent development of the "Snorkel-tag," which includes EV-membrane-targeted CD81 fused to a series of extra-vesicular protein tags, can be used to mark EVs originating from a specific source for subsequent isolation and characterization. We developed an in vivo mouse model, termed "CAGS-Snorkel," which expresses the Snorkel-tag under the control of the Cre-lox system, and crossed this mouse with either Prx1-Cre (mesenchymal progenitors) or Ocn-Cre (osteoblasts/osteocytes) and isolated Snorkel-tagged EVs from the mouse bone marrow plasma using a magnetic bead affinity column. miRNA-sequencing was performed on the isolated EVs, and although similar profiles were observed, a few key miRNAs involved in bone metabolism (miR-106b-5p, miRs-19b-3p, and miRs-219a-5p) were enriched in the Ocn-derived relative to the Prx1-derived EV subpopulations. To characterize the effects of these small EVs on a bone cell target, cultured mouse bone marrow stromal cells were treated with Prx1 or Ocn EVs, and mRNA-sequencing was performed. Pathways involved in ossification, bone development, and extracellular matrix interactions were regulated by both EV subpopulations, whereas a few pathways including advanced glycation end-products signaling were uniquely regulated in the Ocn EV subpopulation, underlying important biological effects of specific EV subpopulations within the bone marrow microenvironment. These data demonstrate that EV isolation in vivo using the CAGS-Snorkel mouse model is a useful tool in characterizing the cargo and understanding the biology of tissue-specific EVs. Moreover, while bone mesenchymal cell populations share a common EV secretory profile, we uncover key differences based on the stage of osteoblastic differentiation that may have important biological consequences.
Collapse
Affiliation(s)
- David G Monroe
- Robert and Arlene Kogod Center on Aging and Division of Endocrinology, Mayo Clinic College of Medicine, Rochester, MN 55905, United States
| | - Naureen Javeed
- Department of Physiology and Biomedical Engineering, Mayo Clinic College of Medicine and Science, Rochester, MN 55905, United States
| | - Jennifer L Rowsey
- Robert and Arlene Kogod Center on Aging and Division of Endocrinology, Mayo Clinic College of Medicine, Rochester, MN 55905, United States
| | - Ming Ruan
- Robert and Arlene Kogod Center on Aging and Division of Endocrinology, Mayo Clinic College of Medicine, Rochester, MN 55905, United States
| | - Chantal E McCabe
- Department of Quantitative Health Sciences, Mayo Clinic College of Medicine, Rochester, MN 55905, United States
| | - Bryan T Piatkowski
- Department of Quantitative Health Sciences, Mayo Clinic College of Medicine, Rochester, MN 55905, United States
| | - Abhishek Roy
- Department of Physiology and Biomedical Engineering, Mayo Clinic College of Medicine and Science, Rochester, MN 55905, United States
| | - Madhusudhan R Bobbili
- Institute of Molecular Biotechnology, BOKU University, 1180, Vienna, Austria
- Ludwig Boltzmann Institute for Traumatology, The Research Institute in Cooperation with AUVA, 1200, Vienna, Austria
| | - Johannes Grillari
- Institute of Molecular Biotechnology, BOKU University, 1180, Vienna, Austria
- Ludwig Boltzmann Institute for Traumatology, The Research Institute in Cooperation with AUVA, 1200, Vienna, Austria
| | - Sundeep Khosla
- Robert and Arlene Kogod Center on Aging and Division of Endocrinology, Mayo Clinic College of Medicine, Rochester, MN 55905, United States
| |
Collapse
|
6
|
Liang W, Feng R, Li X, Duan X, Feng S, Chen J, Li Y, Chen J, Liu Z, Wang X, Ruan G, Tang S, Ding C, Huang B, Zou Z, Chen T. A RANKL-UCHL1-sCD13 negative feedback loop limits osteoclastogenesis in subchondral bone to prevent osteoarthritis progression. Nat Commun 2024; 15:8792. [PMID: 39389988 PMCID: PMC11466963 DOI: 10.1038/s41467-024-53119-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2023] [Accepted: 09/27/2024] [Indexed: 10/12/2024] Open
Abstract
Abnormal subchondral bone remodeling plays a pivotal role in the progression of osteoarthritis (OA). Here, we analyzed subchondral bone samples from OA patients and observed a significant upregulation of ubiquitin carboxy-terminal hydrolase L1 (UCHL1) specifically in subchondral bone osteoclasts. Notably, we found a strong correlation between UCHL1 expression and osteoclast activity in the subchondral bone during OA progression in both human and murine models. Conditional UCHL1 deletion in osteoclast precursors exacerbated OA progression, while its overexpression, mediated by adeno-associated virus 9, alleviated this process in male mice. Mechanistically, RANKL stimulates UCHL1 expression in osteoclast precursors, subsequently stabilizing CD13, augmenting soluble CD13 (sCD13) release, and triggering an autocrine inhibitory effect on the MAPK pathway, thereby suppressing osteoclast formation. These findings unveil a previously unidentified negative feedback loop, RANKL-UCHL1-sCD13, that modulates osteoclast formation and presents a potential therapeutic target for OA.
Collapse
Affiliation(s)
- Wenquan Liang
- Department of Cell Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China.
- Department of Anatomy, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China.
| | - Ru Feng
- Department of Rehabilitation medicine, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
- School of Rehabilitation, Capital Medical University, Beijing, China
- Department of Spinal and Neural Functional Reconstruction, China Rehabilitation Research Center, Beijing, China
| | - Xiaojia Li
- Department of Rehabilitation medicine, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
| | - Xingwei Duan
- Department of Cell Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Shourui Feng
- State Key Laboratory of Biocontrol and Guangdong Provincial Key Laboratory of Plant Resources, School of Life Sciences, Sun Yat-sen University, Guangzhou, China
| | - Jun Chen
- Department of Rehabilitation Medicine, Nanfang Hospital of Southern Medical University, Guangzhou, China
| | - Yicheng Li
- Department of Rehabilitation medicine, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
| | - Junqi Chen
- Department of Rehabilitation medicine, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
| | - Zezheng Liu
- Department of Orthopaedics, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
| | - Xiaogang Wang
- Department of Orthopaedics, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
| | - Guangfeng Ruan
- Clinical Research Centre, Guangzhou First People's Hospital, Guangzhou, China
| | - Su'an Tang
- Clinical Research Centre, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Changhai Ding
- Clinical Research Centre, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Bin Huang
- Department of Orthopaedics, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China.
| | - Zhipeng Zou
- Department of Cell Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China.
| | - Tianyu Chen
- Department of Orthopaedics, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China.
- Department of Orthopaedic Surgery, Third Hospital of Hebei Medical University, Shijiazhuang, China.
| |
Collapse
|
7
|
Wu Y, Mohd Sani SB, Peng K, Lin T, Tan C, Huang X, Li Z. Research progress of the Otubains subfamily in hepatocellular carcinoma. Biomed Pharmacother 2024; 179:117348. [PMID: 39208669 DOI: 10.1016/j.biopha.2024.117348] [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: 05/14/2024] [Revised: 08/14/2024] [Accepted: 08/22/2024] [Indexed: 09/04/2024] Open
Abstract
In cancer research, oncogenesis can be affected by modulating the deubiquitination pathway. Ubiquitination regulates proteins post-translationally in variety of physiological processes. The Otubain Subfamily includes OTUB1 (ovarian tumor-associated proteinase B1) and OTUB2(ovarian tumor-associated proteinase B2). They are deubiquitinating enzymes, which are research hotspots in tumor immunotherapy, with their implications extending across the spectrum of tumor development. Understanding their important role in tumorigenesis, includ-ing hepatocellular carcinoma (HCC) is crucial. HCC has alarming global incidence rates and mortality statistics, ranking among the top five prevalent cancers in Malaysia1. Numerous studies have consistently indicated significant expression of OTUB1 and OTUB2 in HCC cells. In addition, OTUB1 has important biological functions in cancer, suggesting its important role in tumorigenesis. However, the mechanism underlying the action of OTUB1 and OTUB2 in liver cancer remains inadequately explored. Therefore, Otubain Subfamily, as potential molecular target, holds promise for advancing HCC treatments. However, further clinical studies are required to verify its efficacy and application prospects.
Collapse
Affiliation(s)
- Yanming Wu
- Department of Biomedical Sciences, Advanced Medical and Dental Institute, Universiti Sains Malaysia, Bertam, Kepala Batas, Penang 13200, Malaysia.
| | - Sa'udah Badriah Mohd Sani
- Department of Biomedical Sciences, Advanced Medical and Dental Institute, Universiti Sains Malaysia, Bertam, Kepala Batas, Penang 13200, Malaysia.
| | - Ke Peng
- Department of Neurology, School of Clinical Medicine, The First Affiliated Hospital of Yangtze University, Jingzhou, Hubei 434000, China.
| | - Tao Lin
- Department of General Surgery, Anyang People's Hospital, Anyang, Henan 450000, China.
| | - Chenghao Tan
- Department of Social Science, Universiti Sain Malaysia, Gelugor, Penang 11700, Malaysia.
| | | | - Zhengrui Li
- Shanghai Jiao Tong University School of Medicine, Shanghai 200240, China.
| |
Collapse
|
8
|
Yuan R, Li Y, Li X, Fu Y, Ning A, Wang D, Zhang R, Yu S, Xu Q. Transcriptome analysis to explore the mechanism of downregulated TNIK influencing the effect of risperidone. Front Pharmacol 2024; 15:1431923. [PMID: 39268461 PMCID: PMC11391209 DOI: 10.3389/fphar.2024.1431923] [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: 05/13/2024] [Accepted: 08/12/2024] [Indexed: 09/15/2024] Open
Abstract
Background Risperidone is one of the most reliable and effective antipsychotics for schizophrenia treatment. However, the mechanism of action of risperidone is not yet fully understood. Traf2 and Nck-interacting protein kinase (TNIK), a schizophrenia susceptibility gene, is associated with risperidone treatment response. Our previous in vitro experiments confirmed that downregulated TNIK affected the effect of risperidone on downstream targets. However, the effect of downregulated TNIK on risperidone-induced molecular expression remains to be further explored. Methods Transcriptome analysis was performed on U251 cells subjected to risperidone, TNIK siRNA, and no treatment, respectively. Compared to the no-treatment group, two groups of DEGs were screened out and then intersected with the schizophrenia-related genes to screen the cross-talk genes. Those DEGs were analyzed using GO and KEGG. STRING and Cytoscape were used to construct a protein-protein interaction (PPI) network for the cross-talk gene. Results The results showed that the parathyroid hormone synthesis, secretion, and action were significantly enriched after risperidone treatment. Downregulated TNIK could have an impact on the collagen-containing extracellular matrix, signaling receptor activator activity, and PI3K-Akt signaling pathway. Interestingly, bone mineralization function and calcium signaling pathway were enriched in the cross-talk genes. Additionally, FGFR2, FGF1, and FGFR might be the potential targets for TNIK affecting the effects of risperidone. Conclusion The study indicated that risperidone primarily influences functions and/or pathways associated with bone metabolism, potentially contributing to the adverse effect of osteoporosis. Our study may offer a novel perspective on investigating the mechanisms underlying the adverse effects of risperidone.
Collapse
Affiliation(s)
- Ruixue Yuan
- Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yaojing Li
- 958 Hospital of PLA ARMY, Chongqing, China
| | - Xiangyi Li
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, Shanghai, China
| | - Yingmei Fu
- Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ailing Ning
- Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Dongxiang Wang
- Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ran Zhang
- Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Shunying Yu
- Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Qingqing Xu
- Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
9
|
Wei Y, Wu B, Liu M, Cui CP. The Discovery of a Specific CKIP-1 Ligand for the Potential Treatment of Disuse Osteoporosis. Int J Mol Sci 2024; 25:8870. [PMID: 39201556 PMCID: PMC11354310 DOI: 10.3390/ijms25168870] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Revised: 07/30/2024] [Accepted: 08/03/2024] [Indexed: 09/02/2024] Open
Abstract
Bone homeostasis relies on the delicate balance between osteoblast-mediated bone formation and osteoclast-mediated bone resorption. The casein kinase 2 interacting protein-1 (CKIP-1), a specific CK2α subunit-interacting protein, has been documented as one of the crucial negative regulators of bone formation. CKIP-1 siRNA therapy has constraints that limit its use in clinical applications. Therefore, it is necessary to explore effective targeting strategies for CKIP-1. In this study, we observed an upregulation of CKIP-1 protein expression in the microgravity environment, while its ubiquitination levels decreased. We further investigated the interaction between CKIP-1 and VHL and found that VHL enhanced CKIP-1 degradation through the ubiquitylation-proteasome system (UPS). Additionally, we discovered a small molecule ligand, named C77, through DNA-encoded library (DEL) screening, which binds to CKIP-1 both in vivo and in vitro, as confirmed by Surface Plasmon Resonance (SPR) and the Cellular Thermal shift assay (CETSA), respectively. Our findings demonstrated the potential of VHL and C77 as guiding factors in the development of CKIP-1-based Proteolysis-Targeting Chimeras (PROTACs), which could be future therapeutic interventions in disuse osteoporosis.
Collapse
Affiliation(s)
| | | | | | - Chun-Ping Cui
- State Key Laboratory of Medical Proteomics, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing 100850, China; (Y.W.); (B.W.); (M.L.)
| |
Collapse
|
10
|
Zhang HR, Wang YH, Xiao ZP, Yang G, Xu YR, Huang ZT, Wang WZ, He F. E3 ubiquitin ligases: key regulators of osteogenesis and potential therapeutic targets for bone disorders. Front Cell Dev Biol 2024; 12:1447093. [PMID: 39211390 PMCID: PMC11358089 DOI: 10.3389/fcell.2024.1447093] [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: 06/11/2024] [Accepted: 08/05/2024] [Indexed: 09/04/2024] Open
Abstract
Ubiquitination is a crucial post-translational modification of proteins that mediates the degradation or functional regulation of specific proteins. This process participates in various biological processes such as cell growth, development, and signal transduction. E3 ubiquitin ligases play both positive and negative regulatory roles in osteogenesis and differentiation by ubiquitination-mediated degradation or stabilization of transcription factors, signaling molecules, and cytoskeletal proteins. These activities affect the proliferation, differentiation, survival, and bone formation of osteoblasts (OBs). In recent years, advances in genomics, transcriptomics, and proteomics have led to a deeper understanding of the classification, function, and mechanisms of action of E3 ubiquitin ligases. This understanding provides new insights and approaches for revealing the molecular regulatory mechanisms of bone formation and identifying therapeutic targets for bone metabolic diseases. This review discusses the research progress and significance of the positive and negative regulatory roles and mechanisms of E3 ubiquitin ligases in the process of osteogenic differentiation. Additionally, the review highlights the role of E3 ubiquitin ligases in bone-related diseases. A thorough understanding of the role and mechanisms of E3 ubiquitin ligases in osteogenic differentiation could provide promising therapeutic targets for bone tissue engineering based on stem cells.
Collapse
Affiliation(s)
- Heng-Rui Zhang
- School of Medicine, Kunming University of Science and Technology, Kunming, Yunnan, China
- Department of Orthopedic, Qujing Affiliated Hospital of Kunming Medical University, Qujing, Yunnan, China
| | - Yang-Hao Wang
- Department of Pathology, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
- Department of Orthopedics, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
| | - Zhen-Ping Xiao
- Department of Orthopedic, Qujing Affiliated Hospital of Kunming Medical University, Qujing, Yunnan, China
- Department of Pain and Rehabilitation, The Second Affiliated Hospital of University of South China, Hengyang, Hunan, China
| | - Guang Yang
- Department of Trauma Surgery, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
| | - Yun-Rong Xu
- Department of Orthopedic, Qujing Affiliated Hospital of Kunming Medical University, Qujing, Yunnan, China
| | - Zai-Tian Huang
- Department of Orthopedic, Qujing Affiliated Hospital of Kunming Medical University, Qujing, Yunnan, China
| | - Wei-Zhou Wang
- Department of Orthopedics, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
| | - Fei He
- Department of Orthopedic, Qujing Affiliated Hospital of Kunming Medical University, Qujing, Yunnan, China
| |
Collapse
|
11
|
Fan X, Zhang R, Xu G, Fan P, Luo W, Cai C, Ge RL. Role of ubiquitination in the occurrence and development of osteoporosis (Review). Int J Mol Med 2024; 54:68. [PMID: 38940355 PMCID: PMC11232666 DOI: 10.3892/ijmm.2024.5392] [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: 03/29/2024] [Accepted: 06/14/2024] [Indexed: 06/29/2024] Open
Abstract
The ubiquitin (Ub)‑proteasome system (UPS) plays a pivotal role in maintaining protein homeostasis and function to modulate various cellular processes including skeletal cell differentiation and bone homeostasis. The Ub ligase E3 promotes the transfer of Ub to the target protein, especially transcription factors, to regulate the proliferation, differentiation and survival of bone cells, as well as bone formation. In turn, the deubiquitinating enzyme removes Ub from modified substrate proteins to orchestrate bone remodeling. As a result of abnormal regulation of ubiquitination, bone cell differentiation exhibits disorder and then bone homeostasis is affected, consequently leading to osteoporosis. The present review discussed the role and mechanism of UPS in bone remodeling. However, the specific mechanism of UPS in the process of bone remodeling is still not fully understood and further research is required. The study of the mechanism of action of UPS can provide new ideas and methods for the prevention and treatment of osteoporosis. In addition, the most commonly used osteoporosis drugs that target ubiquitination processes in the clinic are discussed in the current review.
Collapse
Affiliation(s)
- Xiaoxia Fan
- Research Center for High Altitude Medicine, Qinghai University, Xining, Qinghai 810000, P.R. China
- Key Laboratory of The Ministry of High Altitude Medicine, Qinghai University, Xining, Qinghai 810000, P.R. China
- Key Laboratory of Applied Fundamentals of High Altitude Medicine, (Qinghai-Utah Joint Key Laboratory of Plateau Medicine), Qinghai University, Xining, Qinghai 810000, P.R. China
- Laboratory for High Altitude Medicine of Qinghai Province, Qinghai University, Xining, Qinghai 810000, P.R. China
- Qinghai Provincial People's Hospital, Department of Endocrinology, Xining, Qinghai 810000, P.R. China
| | - Rong Zhang
- Research Center for High Altitude Medicine, Qinghai University, Xining, Qinghai 810000, P.R. China
- Key Laboratory of The Ministry of High Altitude Medicine, Qinghai University, Xining, Qinghai 810000, P.R. China
- Key Laboratory of Applied Fundamentals of High Altitude Medicine, (Qinghai-Utah Joint Key Laboratory of Plateau Medicine), Qinghai University, Xining, Qinghai 810000, P.R. China
- Laboratory for High Altitude Medicine of Qinghai Province, Qinghai University, Xining, Qinghai 810000, P.R. China
| | - Guocai Xu
- Research Center for High Altitude Medicine, Qinghai University, Xining, Qinghai 810000, P.R. China
- Key Laboratory of The Ministry of High Altitude Medicine, Qinghai University, Xining, Qinghai 810000, P.R. China
- Key Laboratory of Applied Fundamentals of High Altitude Medicine, (Qinghai-Utah Joint Key Laboratory of Plateau Medicine), Qinghai University, Xining, Qinghai 810000, P.R. China
- Laboratory for High Altitude Medicine of Qinghai Province, Qinghai University, Xining, Qinghai 810000, P.R. China
| | - Peiyun Fan
- Qinghai Provincial People's Hospital, Department of Endocrinology, Xining, Qinghai 810000, P.R. China
| | - Wei Luo
- Qinghai Provincial People's Hospital, Department of Endocrinology, Xining, Qinghai 810000, P.R. China
| | - Chunmei Cai
- Research Center for High Altitude Medicine, Qinghai University, Xining, Qinghai 810000, P.R. China
- Key Laboratory of The Ministry of High Altitude Medicine, Qinghai University, Xining, Qinghai 810000, P.R. China
- Key Laboratory of Applied Fundamentals of High Altitude Medicine, (Qinghai-Utah Joint Key Laboratory of Plateau Medicine), Qinghai University, Xining, Qinghai 810000, P.R. China
- Laboratory for High Altitude Medicine of Qinghai Province, Qinghai University, Xining, Qinghai 810000, P.R. China
| | - Ri-Li Ge
- Research Center for High Altitude Medicine, Qinghai University, Xining, Qinghai 810000, P.R. China
- Key Laboratory of The Ministry of High Altitude Medicine, Qinghai University, Xining, Qinghai 810000, P.R. China
- Key Laboratory of Applied Fundamentals of High Altitude Medicine, (Qinghai-Utah Joint Key Laboratory of Plateau Medicine), Qinghai University, Xining, Qinghai 810000, P.R. China
- Laboratory for High Altitude Medicine of Qinghai Province, Qinghai University, Xining, Qinghai 810000, P.R. China
| |
Collapse
|
12
|
Wu S, Lei X, Zhu Z, Liu Z, Gao Y, Wei J, Qin Q. Grouper OTUB1 and OTUB2 promote red-spotted grouper nervous necrosis virus (RGNNV) replication by inhibiting the host innate immune response. FISH & SHELLFISH IMMUNOLOGY 2024; 151:109715. [PMID: 38909637 DOI: 10.1016/j.fsi.2024.109715] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Revised: 06/19/2024] [Accepted: 06/19/2024] [Indexed: 06/25/2024]
Abstract
Red-spotted grouper nervous necrosis virus (RGNNV) is a major viral pathogen of grouper and is able to antagonize interferon responses through multiple strategies, particularly evading host immune responses by inhibiting interferon responses. Ovarian tumor (OTU) family proteins are an important class of DUBs and the underlying mechanisms used to inhibit interferon pathway activation are unknown. In the present study, primers were designed based on the transcriptome data, and the ovarian tumor (OTU) domain-containing ubiquitin aldehyde-binding protein 1 (OTUB1) and OTUB2 genes of Epinephelus coioides (EcOTUB1 and EcOTUB2) were cloned and characterized. The homology alignment showed that both EcOTUB1 and EcOTUB2 were most closely related to E. lanceolatus with 98 % identity. Both EcOTUB1 and EcOTUB2 were distributed to varying degrees in grouper tissues, and the transcript levels were significantly up-regulated following RGNNV stimulation. Both EcOTUB1 and EcOTUB2 promoted replication of RGNNV in vitro, and inhibited the promoter activities of interferon stimulated response element (ISRE), nuclear transcription factors kappaB (NF-κB) and IFN3, and the expression levels of interferon related genes and proinflammatory factors. Co-immunoprecipitation experiments showed that both EcOTUB1 and EcOTUB2 could interact with TRAF3 and TRAF6, indicating that EcOTUB1 and EcOTUB2 may play important roles in interferon signaling pathway. The results will provide a theoretical reference for the development of novel disease prevention and control techniques.
Collapse
Affiliation(s)
- Siting Wu
- College of Marine Sciences, South China Agricultural University, Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou, 510642, China; Nansha-South China Agricultural University Fishery Research Institute, Guangzhou, Guangzhou, 511400, China
| | - Xiaoxia Lei
- College of Marine Sciences, South China Agricultural University, Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou, 510642, China; Nansha-South China Agricultural University Fishery Research Institute, Guangzhou, Guangzhou, 511400, China
| | - Zheng Zhu
- College of Marine Sciences, South China Agricultural University, Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou, 510642, China; Nansha-South China Agricultural University Fishery Research Institute, Guangzhou, Guangzhou, 511400, China
| | - Zetian Liu
- College of Marine Sciences, South China Agricultural University, Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou, 510642, China; Nansha-South China Agricultural University Fishery Research Institute, Guangzhou, Guangzhou, 511400, China
| | - Yanfei Gao
- Guangdong Haiyuan Agricultural Technology Co., Ltd, Yangjiang, 529800, China
| | - Jingguang Wei
- College of Marine Sciences, South China Agricultural University, Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou, 510642, China; Nansha-South China Agricultural University Fishery Research Institute, Guangzhou, Guangzhou, 511400, China.
| | - Qiwei Qin
- College of Marine Sciences, South China Agricultural University, Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou, 510642, China; Nansha-South China Agricultural University Fishery Research Institute, Guangzhou, Guangzhou, 511400, China.
| |
Collapse
|
13
|
Guo M, Li X, Tao W, Teng F, Li C. Vibrio splendidus infection promotes circRNA-FGL1-regulated coelomocyte apoptosis via competitive binding to Myc with the deubiquitinase OTUB1 in Apostichopus japonicus. PLoS Pathog 2024; 20:e1012463. [PMID: 39146353 PMCID: PMC11349225 DOI: 10.1371/journal.ppat.1012463] [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: 05/10/2024] [Revised: 08/27/2024] [Accepted: 07/30/2024] [Indexed: 08/17/2024] Open
Abstract
Circular RNAs (circRNAs) are involved in various physiological and pathological processes in both vertebrates and invertebrates. However, most studies on circRNAs have focused on their roles as endogenous competitive RNAs. Here, we report a novel function of circRNA derived from the Fibrinogen-like protein 1 gene (circ-FGL1) that inhibits coelomocyte apoptosis via competing with the deubiquitinase AjOTUB1 to bind AjMyc in Apostichopus japonicus during Vibrio splendidus infection. The results showed that circ-FGL1 is significantly downregulated in coelomocytes of V. splendidus-induced A. japonicus and negatively regulates coelomocyte apoptosis through the AjBax-AjCyt c pathway. Mechanistically, the deubiquitinase AjOTUB1 and circ-FGL1 could interact with the transcription factor protein AjMyc in the same region with circ-FGL1/AjMyc having greater affinity. Under normal conditions, high levels of circ-FGL1 bind directly to AjMyc, inhibiting the deubiquitylation of AjMyc by AjOTUB1 and leading to the degradation of AjMyc. After V. splendidus infection, AjMyc disassociates from the depressed expression of circ-FGL1, promoting its deubiquitylation by binding to the induced deubiquitinase AjOTUB1 to inhibit its degradation. AjMyc is then transferred to the nucleus and promotes the transcription of AjCyt c and AjBax to induce coelomocyte apoptosis. The new finding will expand our present outstanding on the functional role of circRNAs and suggest new therapeutic targets for the treatment of echinoderms during bacterial invasion.
Collapse
Affiliation(s)
- Ming Guo
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, School of Marine Sciences, Ningbo University, Ningbo, China
| | - Xin 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, China
| | - Wenjun Tao
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, School of Marine Sciences, Ningbo University, Ningbo, China
| | - Fei Teng
- College of Mathematics and Computer, Jilin Normal University, Siping, Jilin, China
| | - Chenghua 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, China
| |
Collapse
|
14
|
Zhang Y, Yang Y, Qi X, Cui P, Kang Y, Liu H, Wei Z, Wang H. SLC14A1 and TGF-β signaling: a feedback loop driving EMT and colorectal cancer metachronous liver metastasis. J Exp Clin Cancer Res 2024; 43:208. [PMID: 39061061 PMCID: PMC11282742 DOI: 10.1186/s13046-024-03114-8] [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: 12/01/2023] [Accepted: 07/01/2024] [Indexed: 07/28/2024] Open
Abstract
BACKGROUND Colorectal cancer (CRC) metachronous liver metastasis is a significant clinical challenge, largely attributable to the late detection and the intricate molecular mechanisms that remain poorly understood. This study aims to elucidate the role of Solute Carrier Family 14 Member 1 (SLC14A1) in the pathogenesis and progression of CRC metachronous liver metastasis. METHODS We conducted a comprehensive analysis of CRC patient data from The Cancer Genome Atlas and GSE40967 databases, focusing on the differential expression of genes associated with non-metachronous liver metastasis and metachronous liver metastasis. Functional assays, both in vitro and in vivo, were performed to assess the biological impact of SLC14A1 modulation in CRC cells. Gene set enrichment analysis, molecular assays and immunohistochemical analyses on clinical specimens were employed to unravel the underlying mechanisms through which SLC14A1 exerts its effects. RESULTS SLC14A1 was identified as a differentially expressed gene, with its overexpression significantly correlating with poor relapse-free and overall survival. Mechanistically, elevated SLC14A1 levels enhanced CRC cell invasiveness and migratory abilities, corroborated by upregulated TGF-β/Smad signaling and Epithelial-Mesenchymal Transition. SLC14A1 interacted with TβRII and stabilized TβRII protein, impeding its Smurf1-mediated K48-linked ubiquitination and degradation, amplifying TGF-β/Smad signaling. Furthermore, TGF-β1 reciprocally elevated SLC14A1 mRNA expression, with Snail identified as a transcriptional regulator, binding downstream of SLC14A1's transcription start site, establishing a positive feedback loop. Clinically, SLC14A1, phosphorylated Smad2, and Snail were markedly upregulated in CRC patients with metachronous liver metastasis, underscoring their potential as prognostic markers. CONCLUSIONS Our findings unveil SLC14A1 as a critical regulator in CRC metachronous liver metastasis, providing novel insights into the molecular crosstalk between SLC14A1 and TGF-β/Smad signaling. These discoveries not only enhance our understanding of CRC metachronous liver metastasis pathogenesis, but also highlight SLC14A1 as a promising target for therapeutic intervention and predictive marker.
Collapse
Affiliation(s)
- Yixun Zhang
- Department of Colorectal Surgery, Cancer Hospital Affiliated to Shanxi Medical University, Shanxi Province Cancer Hospital, Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Sciences, Taiyuan, China
| | - Yumeng Yang
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Capital Medical University, No.10 Xitoutiao, You An Men Wai, Beijing, 100069, China
- Laboratory for Clinical Medicine, Capital Medical University, No.10 Xitoutiao, You An Men Wai, Beijing, China
| | - Xuan Qi
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Capital Medical University, No.10 Xitoutiao, You An Men Wai, Beijing, 100069, China
- Laboratory for Clinical Medicine, Capital Medical University, No.10 Xitoutiao, You An Men Wai, Beijing, China
| | - Peng Cui
- Department of General Surgery, State Key Lab of Digestive Health, National Clinical Research Center for Digestive Diseases, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Yi Kang
- Department of Gastroenterology, Cancer Hospital Affiliated to Shanxi Medical University, Shanxi Province Cancer Hospital, Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Sciences, Taiyuan, China
| | - Haiyi Liu
- Department of Colorectal Surgery, Cancer Hospital Affiliated to Shanxi Medical University, Shanxi Province Cancer Hospital, Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Sciences, Taiyuan, China
| | - Zhigang Wei
- Hepatobiliary and Pancreatic Surgery and Liver Transplantation Center, First Hospital of Shanxi Medical University, 85 Jiefang Nan Lu, Taiyuan, 030001, Shanxi, China.
| | - Haibo Wang
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Capital Medical University, No.10 Xitoutiao, You An Men Wai, Beijing, 100069, China.
- Laboratory for Clinical Medicine, Capital Medical University, No.10 Xitoutiao, You An Men Wai, Beijing, China.
- Beijing Laboratory of Oral Health, Capital Medical University, No.10 Xitoutiao, You An Men Wai, Beijing, China.
| |
Collapse
|
15
|
Zhu S, Chen W, Masson A, Li YP. Cell signaling and transcriptional regulation of osteoblast lineage commitment, differentiation, bone formation, and homeostasis. Cell Discov 2024; 10:71. [PMID: 38956429 PMCID: PMC11219878 DOI: 10.1038/s41421-024-00689-6] [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: 07/07/2023] [Accepted: 05/04/2024] [Indexed: 07/04/2024] Open
Abstract
The initiation of osteogenesis primarily occurs as mesenchymal stem cells undergo differentiation into osteoblasts. This differentiation process plays a crucial role in bone formation and homeostasis and is regulated by two intricate processes: cell signal transduction and transcriptional gene expression. Various essential cell signaling pathways, including Wnt, BMP, TGF-β, Hedgehog, PTH, FGF, Ephrin, Notch, Hippo, and Piezo1/2, play a critical role in facilitating osteoblast differentiation, bone formation, and bone homeostasis. Key transcriptional factors in this differentiation process include Runx2, Cbfβ, Runx1, Osterix, ATF4, SATB2, and TAZ/YAP. Furthermore, a diverse array of epigenetic factors also plays critical roles in osteoblast differentiation, bone formation, and homeostasis at the transcriptional level. This review provides an overview of the latest developments and current comprehension concerning the pathways of cell signaling, regulation of hormones, and transcriptional regulation of genes involved in the commitment and differentiation of osteoblast lineage, as well as in bone formation and maintenance of homeostasis. The paper also reviews epigenetic regulation of osteoblast differentiation via mechanisms, such as histone and DNA modifications. Additionally, we summarize the latest developments in osteoblast biology spurred by recent advancements in various modern technologies and bioinformatics. By synthesizing these insights into a comprehensive understanding of osteoblast differentiation, this review provides further clarification of the mechanisms underlying osteoblast lineage commitment, differentiation, and bone formation, and highlights potential new therapeutic applications for the treatment of bone diseases.
Collapse
Affiliation(s)
- Siyu Zhu
- Division in Cellular and Molecular Medicine, Department of Pathology and Laboratory Medicine, Tulane University School of Medicine, Tulane University, New Orleans, LA, USA
| | - Wei Chen
- Division in Cellular and Molecular Medicine, Department of Pathology and Laboratory Medicine, Tulane University School of Medicine, Tulane University, New Orleans, LA, USA.
| | - Alasdair Masson
- Division in Cellular and Molecular Medicine, Department of Pathology and Laboratory Medicine, Tulane University School of Medicine, Tulane University, New Orleans, LA, USA
| | - Yi-Ping Li
- Division in Cellular and Molecular Medicine, Department of Pathology and Laboratory Medicine, Tulane University School of Medicine, Tulane University, New Orleans, LA, USA.
| |
Collapse
|
16
|
Zhang XN, Zhang YJ, Wang L, Hong SJ, Zhang CL, Zhao XL, Zeng T. NLRP3 inflammasome activation triggers severe inflammatory liver injury in N, N-dimethylformamide-exposed mice. THE SCIENCE OF THE TOTAL ENVIRONMENT 2024; 929:172653. [PMID: 38649053 DOI: 10.1016/j.scitotenv.2024.172653] [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: 02/06/2024] [Revised: 04/17/2024] [Accepted: 04/19/2024] [Indexed: 04/25/2024]
Abstract
N,N-dimethylformamide (DMF) is a widely utilized chemical solvent with various industrial applications. Previous studies have indicated that the liver is the most susceptible target to DMF exposure, whereas the underlying mechanisms remain to be elucidated. This study aimed to investigate the role of NLRP3 inflammasome in DMF-induced liver injury in mice by using two NLRP3 inflammasome inhibitors, Nlrp3-/- mice, Nfe2l2-/- mice, and a macrophage-depleting agent. RNA sequencing revealed that endoplasmic reticulum (ER) stress and NLRP3 inflammasome-associated pathways were activated in the mouse liver after acute DMF exposure, which was validated by Western blotting. Interestingly, DMF-induced liver injury was effectively suppressed by two inflammasome inhibitors, MCC950 and Dapansutrile. In addition, knockout of Nlrp3 markedly attenuated DMF-induced liver injury without affecting the metabolism of DMF. Furthermore, silencing Nfe2l2 aggravated the liver injury and the NLRP3 inflammasome activation in mouse liver. Finally, the depletion of hepatic macrophages by clodronate liposomes significantly reduced the liver damage caused by DMF. These results suggest that NLRP3 inflammasome activation is the upstream molecular event in the development of acute liver injury induced by DMF.
Collapse
Affiliation(s)
- Xiu-Ning Zhang
- Institute of Toxicology, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China
| | - Yan-Jing Zhang
- Institute of Toxicology, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China
| | - Lin Wang
- Institute of Toxicology, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China
| | - Shu-Jun Hong
- Institute of Toxicology, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China
| | - Cui-Li Zhang
- Institute of Toxicology, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China
| | - Xiu-Lan Zhao
- Institute of Toxicology, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China.
| | - Tao Zeng
- Institute of Toxicology, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China.
| |
Collapse
|
17
|
Chen Q, Wu Z, Shi Y, Li Z, Yang J, Qu M, Zhang S, Wang Z, Ji N, Li J, Shen Y, Xie L, Chen Q. Loss of PA28γ exacerbates imbalanced differentiation of bone marrow stromal cells during bone formation and bone healing in mice. J Bone Miner Res 2024; 39:326-340. [PMID: 38477820 DOI: 10.1093/jbmr/zjae012] [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: 08/24/2023] [Revised: 01/04/2024] [Accepted: 01/10/2024] [Indexed: 03/14/2024]
Abstract
Proteasome activator subunit 3 (PA28γ) is a member of the proteasome activator family, which mainly regulates the degradation and stability of proteins. Studies have shown that it plays crucial roles in lipid formation, stemness maintenance, and blood vessel formation. However, few studies have clarified the association between PA28γ and bone diseases. Herein, we identified PA28γ as a previously unknown regulator of bone homeostasis that coordinates bone formation and lipid accumulation. PA28γ-knockout mice presented with the characteristics of low bone mass and accumulation of lipids. Suppressed expression of PA28γ restrained the osteogenic differentiation and enhanced the adipogenic differentiation of bone marrow stromal cells (BMSCs). Overexpression of PA28γ promoted osteogenic differentiation and inhibited adipogenic differentiation of BMSCs. Mechanistically, PA28γ interacted with Wnt5α, and the two interactors appeared to be positively correlated. PA28γ mainly activated the downstream Wnt/β-catenin signaling pathway, which affects BMSCs differentiation homeostasis. Deletion of Wnt5α significantly delayed the promotion of osteogenic differentiation and partially alleviated the inhibitory effect of adipogenic differentiation of BMSCs in the PA28γ-overexpressing group. Furthermore, we demonstrated that PA28γ-knockout mice had an inhibited rate of bone healing in a drill-hole femoral bone defect model in vivo. Therefore, our results confirm the effects of PA28γ on bone formation and bone defect repair, indicating that PA28γ mainly interacts with Wnt5α to activate the Wnt/β-catenin signaling pathway regulating BMSCs differentiation homeostasis. Our results reveal the function of PA28γ in bone diseases and provide a new theoretical basis for expanding the treatment of bone diseases.
Collapse
Affiliation(s)
- Qian Chen
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Research Unit of Oral Carcinogenesis and Management, Chinese Academy of Medical Sciences, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, China
- The Affiliated Hospital of Stomatology, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou 310006, P. R. China
| | - ZuPing Wu
- The Affiliated Hospital of Stomatology, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou 310006, P. R. China
| | - YuJie Shi
- Department of Stomatology, The First Affiliated Hospital with Nanjing Medical University, Nanjing 210029, P. R. China
| | - ZaiYe Li
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Research Unit of Oral Carcinogenesis and Management, Chinese Academy of Medical Sciences, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, China
| | - JiaKang Yang
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Research Unit of Oral Carcinogenesis and Management, Chinese Academy of Medical Sciences, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, China
| | - MoYuan Qu
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Research Unit of Oral Carcinogenesis and Management, Chinese Academy of Medical Sciences, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, China
| | - ShiYu Zhang
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Research Unit of Oral Carcinogenesis and Management, Chinese Academy of Medical Sciences, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, China
| | - Zheng Wang
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Research Unit of Oral Carcinogenesis and Management, Chinese Academy of Medical Sciences, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, China
| | - Ning Ji
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Research Unit of Oral Carcinogenesis and Management, Chinese Academy of Medical Sciences, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, China
| | - Jing Li
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Research Unit of Oral Carcinogenesis and Management, Chinese Academy of Medical Sciences, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, China
| | - YingQiang Shen
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Research Unit of Oral Carcinogenesis and Management, Chinese Academy of Medical Sciences, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, China
| | - Liang Xie
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Research Unit of Oral Carcinogenesis and Management, Chinese Academy of Medical Sciences, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, China
| | - Qianming Chen
- The Affiliated Hospital of Stomatology, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou 310006, P. R. China
| |
Collapse
|
18
|
Fu Y, Li L, Zhang X, Deng Z, Wu Y, Chen W, Liu Y, He S, Wang J, Xie Y, Tu Z, Lyu Y, Wei Y, Wang S, Cui CP, Liu CH, Zhang L. Systematic HOIP interactome profiling reveals critical roles of linear ubiquitination in tissue homeostasis. Nat Commun 2024; 15:2974. [PMID: 38582895 PMCID: PMC10998861 DOI: 10.1038/s41467-024-47289-2] [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: 08/03/2023] [Accepted: 03/27/2024] [Indexed: 04/08/2024] Open
Abstract
Linear ubiquitination catalyzed by HOIL-1-interacting protein (HOIP), the key component of the linear ubiquitination assembly complex, plays fundamental roles in tissue homeostasis by executing domain-specific regulatory functions. However, a proteome-wide analysis of the domain-specific interactome of HOIP across tissues is lacking. Here, we present a comprehensive mass spectrometry-based interactome profiling of four HOIP domains in nine mouse tissues. The interaction dataset provides a high-quality HOIP interactome resource with an average of approximately 90 interactors for each bait per tissue. HOIP tissue interactome presents a systematic understanding of linear ubiquitination functions in each tissue and also shows associations of tissue functions to genetic diseases. HOIP domain interactome characterizes a set of previously undefined linear ubiquitinated substrates and elucidates the cross-talk among HOIP domains in physiological and pathological processes. Moreover, we show that linear ubiquitination of Integrin-linked protein kinase (ILK) decreases focal adhesion formation and promotes the detachment of Shigella flexneri-infected cells. Meanwhile, Hoip deficiency decreases the linear ubiquitination of Smad ubiquitination regulatory factor 1 (SMURF1) and enhances its E3 activity, finally causing a reduced bone mass phenotype in mice. Overall, our work expands the knowledge of HOIP-interacting proteins and provides a platform for further discovery of linear ubiquitination functions in tissue homeostasis.
Collapse
Affiliation(s)
- Yesheng Fu
- State Key Laboratory of Medical Proteomics, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing, 100850, China
| | - Lei Li
- State Key Laboratory of Medical Proteomics, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing, 100850, China
| | - Xin Zhang
- State Key Laboratory of Medical Proteomics, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing, 100850, China
| | - Zhikang Deng
- State Key Laboratory of Medical Proteomics, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing, 100850, China
| | - Ying Wu
- State Key Laboratory of Medical Proteomics, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing, 100850, China
| | - Wenzhe Chen
- State Key Laboratory of Medical Proteomics, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing, 100850, China
| | - Yuchen Liu
- State Key Laboratory of Medical Proteomics, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing, 100850, China
| | - Shan He
- State Key Laboratory of Medical Proteomics, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing, 100850, China
| | - Jian Wang
- State Key Laboratory of Medical Proteomics, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing, 100850, China
| | - Yuping Xie
- State Key Laboratory of Medical Proteomics, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing, 100850, China
| | - Zhiwei Tu
- State Key Laboratory of Medical Proteomics, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing, 100850, China
| | - Yadi Lyu
- State Key Laboratory of Medical Proteomics, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing, 100850, China
| | - Yange Wei
- State Key Laboratory of Medical Proteomics, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing, 100850, China
| | - Shujie Wang
- State Key Laboratory of Medical Proteomics, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing, 100850, China
| | - Chun-Ping Cui
- State Key Laboratory of Medical Proteomics, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing, 100850, China
| | - Cui Hua Liu
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, 100101, China.
- Savaid Medical School, University of Chinese Academy of Sciences, Beijing, 101408, China.
| | - Lingqiang Zhang
- State Key Laboratory of Medical Proteomics, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing, 100850, China.
| |
Collapse
|
19
|
Campos Alonso M, Knobeloch KP. In the moonlight: non-catalytic functions of ubiquitin and ubiquitin-like proteases. Front Mol Biosci 2024; 11:1349509. [PMID: 38455765 PMCID: PMC10919355 DOI: 10.3389/fmolb.2024.1349509] [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: 12/04/2023] [Accepted: 02/05/2024] [Indexed: 03/09/2024] Open
Abstract
Proteases that cleave ubiquitin or ubiquitin-like proteins (UBLs) are critical players in maintaining the homeostasis of the organism. Concordantly, their dysregulation has been directly linked to various diseases, including cancer, neurodegeneration, developmental aberrations, cardiac disorders and inflammation. Given their potential as novel therapeutic targets, it is essential to fully understand their mechanisms of action. Traditionally, observed effects resulting from deficiencies in deubiquitinases (DUBs) and UBL proteases have often been attributed to the misregulation of substrate modification by ubiquitin or UBLs. Therefore, much research has focused on understanding the catalytic activities of these proteins. However, this view has overlooked the possibility that DUBs and UBL proteases might also have significant non-catalytic functions, which are more prevalent than previously believed and urgently require further investigation. Moreover, multiple examples have shown that either selective loss of only the protease activity or complete absence of these proteins can have different functional and physiological consequences. Furthermore, DUBs and UBL proteases have been shown to often contain domains or binding motifs that not only modulate their catalytic activity but can also mediate entirely different functions. This review aims to shed light on the non-catalytic, moonlighting functions of DUBs and UBL proteases, which extend beyond the hydrolysis of ubiquitin and UBL chains and are just beginning to emerge.
Collapse
Affiliation(s)
- Marta Campos Alonso
- Institute of Neuropathology, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Klaus-Peter Knobeloch
- Institute of Neuropathology, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- CIBSS—Centre for Integrative Biological Signalling Studies, University of Freiburg, Freiburg, Germany
| |
Collapse
|
20
|
Wu M, Sun L, Song T. OTUB1-mediated inhibition of ubiquitination: a growing list of effectors, multiplex mechanisms, and versatile functions. Front Mol Biosci 2024; 10:1261273. [PMID: 38264570 PMCID: PMC10803509 DOI: 10.3389/fmolb.2023.1261273] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Accepted: 12/19/2023] [Indexed: 01/25/2024] Open
Abstract
Protein ubiquitination plays a pivotal role in protein homeostasis. Ubiquitination may regulate the stability, activity, protein-protein interaction, and localization of a protein. Ubiquitination is subject to regulation by two groups of counteracting enzymes, the E3 ubiquitin ligases and deubiquitinases. Consistently, deubiquitinases are involved in essentially all biological processes. OTUB1, an OTU-family deubiquitinase, is a critical regulator of development, cancer, DNA damage response, and immune response. OTUB1 antagonizes the ubiquitination of a wide-spectrum of proteins through at least two different mechanisms. Besides direct deubiquitination, OTUB1 can also inhibit ubiquitination by non-canonically blocking ubiquitin transfer from certain ubiquitin-conjugases (E2). In this review, we start with a general background of protein ubiquitination and deubiquitination. Next, we introduce the basic characteristics of OTUB1 and then elaborate on the updated biological functions of OTUB1. Afterwards, we discuss potential mechanisms underlying the versatility and specificity of OTUB1 functions. In the end, we discuss the perspective that OTUB1 can be a potential therapeutic target for cancer.
Collapse
Affiliation(s)
- Miaomiao Wu
- Deparment of Obstetrics and Gynecology, Shuyang Hospital of Traditional Chinese Medicine, Suqian, China
| | - Lidong Sun
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Cell Architecture Research Institute, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Tanjing Song
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Cell Architecture Research Institute, Huazhong University of Science and Technology, Wuhan, Hubei, China
| |
Collapse
|
21
|
Shi S, Guo D, Ye L, Li T, Fei Q, Lin M, Yu X, Jin K, Wu W. Knockdown of TACC3 inhibits tumor cell proliferation and increases chemosensitivity in pancreatic cancer. Cell Death Dis 2023; 14:778. [PMID: 38012214 PMCID: PMC10682013 DOI: 10.1038/s41419-023-06313-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: 07/09/2023] [Revised: 11/06/2023] [Accepted: 11/15/2023] [Indexed: 11/29/2023]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a highly malignant digestive tract tumor with limited clinical treatments. Transforming acidic coiled-coil-containing protein 3 (TACC3) is a component of the centrosome axis and a member of the TACC family, which affect mitosis and regulate chromosome stability and are involved in tumor development and progression. However, the role of TACC3 in PDAC remains elusive. In this study, by exploiting the TCGA database, we found that high TACC3 expression in PDAC is associated with poor prognosis. shRNA-mediated TACC3 knockdown caused S phase arrest of the cell cycle and inhibited proliferation in PDAC cell lines. Through RNA sequencing and protein co-immunoprecipitation combined with mass spectrometry, KIF11 was identified as a protein that interacts with TACC3. TACC3 stabilizes and regulates KIF11 protein expression levels in PDAC cells through physical interaction. Knockdown of TACC3 or KIF11 resulted in abnormal spindle formation during cell division both in vitro and in vivo. Pharmacological inhibition of TACC3 or KIF11 can suppress tumor cell proliferation and promote apoptosis. Our studies further demonstrated that high expression of TACC3 and KIF11 mediated the resistance of PDAC to gemcitabine, and deficiency of TACC3 or KIF11 increased the sensitivity of PDAC cells to chemotherapy. In conclusion, our study reveals the fundamental role of TACC3 expression in PDAC cell proliferation and chemoresistance, suggesting that TACC3 can be used as a molecular marker to evaluate the prognosis of PDAC.
Collapse
Affiliation(s)
- Saimeng Shi
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
- Shanghai Pancreatic Cancer Institute, Shanghai, 200032, China
- Pancreatic Cancer Institute, Fudan University, Shanghai, 200032, China
| | - Duancheng Guo
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
- Shanghai Pancreatic Cancer Institute, Shanghai, 200032, China
- Pancreatic Cancer Institute, Fudan University, Shanghai, 200032, China
| | - Longyun Ye
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
- Shanghai Pancreatic Cancer Institute, Shanghai, 200032, China
- Pancreatic Cancer Institute, Fudan University, Shanghai, 200032, China
| | - Tianjiao Li
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
- Shanghai Pancreatic Cancer Institute, Shanghai, 200032, China
- Pancreatic Cancer Institute, Fudan University, Shanghai, 200032, China
| | - Qinglin Fei
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
- Shanghai Pancreatic Cancer Institute, Shanghai, 200032, China
- Pancreatic Cancer Institute, Fudan University, Shanghai, 200032, China
| | - Mengxiong Lin
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
- Shanghai Pancreatic Cancer Institute, Shanghai, 200032, China
- Pancreatic Cancer Institute, Fudan University, Shanghai, 200032, China
| | - Xianjun Yu
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032, China.
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China.
- Shanghai Pancreatic Cancer Institute, Shanghai, 200032, China.
- Pancreatic Cancer Institute, Fudan University, Shanghai, 200032, China.
| | - Kaizhou Jin
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032, China.
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China.
- Shanghai Pancreatic Cancer Institute, Shanghai, 200032, China.
- Pancreatic Cancer Institute, Fudan University, Shanghai, 200032, China.
| | - Weiding Wu
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032, China.
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China.
- Shanghai Pancreatic Cancer Institute, Shanghai, 200032, China.
- Pancreatic Cancer Institute, Fudan University, Shanghai, 200032, China.
| |
Collapse
|
22
|
Zhou Y, Zhu P, Shen S, Wang Y, Li B, Guo B, Li H. Overexpression of fibroblast growth factor receptor 2 in bone marrow mesenchymal stem cells enhances osteogenesis and promotes critical cranial bone defect regeneration. Front Cell Dev Biol 2023; 11:1208239. [PMID: 37266455 PMCID: PMC10229770 DOI: 10.3389/fcell.2023.1208239] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Accepted: 05/09/2023] [Indexed: 06/03/2023] Open
Abstract
Background: Reconstruction of cranial bone defects is one of the most challenging problems in reconstructive surgery, and several biological tissue engineering methods have been used to promote bone repair, such as genetic engineering of bone marrow mesenchymal stem cells (BMSCs). Fibroblast growth factor receptor 2 (Fgfr2) is an important regulator of bone construction and can be used as a potential gene editing site. However, its role in the osteogenesis process of BMSCs remains unclear. This article clarifies the function of Fgfr2 in BMSCs and explores the role of Fgfr2-overexpressed BMSCs carried by light-induced porous hydrogel (GelMA) in the repair of cranial bone defects. Methods: Lenti-virus was used to overexpress Fgfr2 in BMSCs, and cell counting kit-8, transwell, and flow cytometry assays were conducted to investigate the proliferation, migration, and characteristics. After 0, 3, 7, and 10 days of osteogenic or chondrogenic induction, the changes in osteogenic and chondrogenic ability were detected by real-time PCR, western blot, alkaline phosphatase staining, alizarin Red staining, and alcian blue staining. To investigate the viability of BMSCs carried by GelMA, calcein and propyl iodide staining were carried out as well. Finally, a critical cranial bone defect model was established in 6-week-old male mice and micro-computerized tomography, masson staining, and immunohistochemistry of OCN were conducted to test the bone regeneration properties of implanting Fgfr2-overexpressed BMSCs with GelMA in cranial bone defects over 6 weeks. Results: Overexpression of Fgfr2 in BMSCs significantly promoted cell proliferation and migration and increased the percentage of CD200+CD105+ cells. After osteogenic and chondrogenic induction, Fgfr2 overexpression enhanced both osteogenic and chondrogenic ability. Furthermore, in cranial bone defect regeneration, BMSCs carried by light-induced GelMA showed favorable biocompatibility, and Fgfr2-overexpressed BMSCs induced superior cranial bone regeneration compared to a normal BMSCs group and an untreated blank group. Conclusion: In vitro, Fgfr2 enhanced the proliferation, migration, and stemness of BMSCs and promoted osteogenesis and chondrogenesis after parallel induction. In vivo, BMSCs with Fgfr2 overexpression carried by GelMA showed favorable performance in treating critical cranial bone defects. This study clarifies the multiple functions of Fgfr2 in BMSCs and provides a new method for future tissue engineering.
Collapse
Affiliation(s)
- Yiwen Zhou
- Department of Orthodontics, Nanjing Stomatological Hospital, Medical School of Nanjing University, Nanjing, China
- Medical School of Nanjing University, Nanjing, China
| | - Peixiang Zhu
- Department of Orthodontics, Nanjing Stomatological Hospital, Medical School of Nanjing University, Nanjing, China
- Medical School of Nanjing University, Nanjing, China
| | - Siyu Shen
- Medical School of Nanjing University, Nanjing, China
| | - Yanyi Wang
- Department of Orthodontics, Nanjing Stomatological Hospital, Medical School of Nanjing University, Nanjing, China
- Medical School of Nanjing University, Nanjing, China
| | - Baochao Li
- Department of Orthodontics, Nanjing Stomatological Hospital, Medical School of Nanjing University, Nanjing, China
- Medical School of Nanjing University, Nanjing, China
| | - Baosheng Guo
- Medical School of Nanjing University, Nanjing, China
| | - Huang Li
- Department of Orthodontics, Nanjing Stomatological Hospital, Medical School of Nanjing University, Nanjing, China
- Medical School of Nanjing University, Nanjing, China
| |
Collapse
|