1
|
Feng X, Wei G, Su Y, Xian Y, Liu Z, Gao Y, Liang J, Lian H, Xu J, Zhao J, Liu Q, Song F. Active fraction of Polyrhachis vicina (Rogers) inhibits osteoclastogenesis by targeting Trim38 mediated proteasomal degradation of TRAF6. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 132:155890. [PMID: 39033726 DOI: 10.1016/j.phymed.2024.155890] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/02/2024] [Revised: 05/25/2024] [Accepted: 07/15/2024] [Indexed: 07/23/2024]
Abstract
BACKGROUND Reactive Oxygen Species (ROS) is a key factor in the pathogenesis of osteoporosis (OP) primarily characterized by excessive osteoclast activity. Active fraction of Polyrhachis vicina Rogers (AFPR) exerts antioxidant effects and possesses extensive promising therapeutic effects in various conditions, however, its function in osteoclastogenesis and OP is unknown. PURPOSE The aim of this study is to elucidate the cellular and molecular mechanisms of AFPR in OP. STUDY DESIGN AND METHODS CCK8 assay was used to evaluate the cell viability under AFPR treatment. TRAcP staining, podosome belts staining and bone resorption were used to test the effect of AFPR on osteoclastogenesis. Immunofluorescence staining was used to observe the effect of AFPR on ROS production. si-RNA transfection, coimmunoprecipitation and Western-blot were used to clarify the underlying mechanisms. Further, an ovariectomy (OVX) -induced OP mice model was used to identify the effect of AFPR on bone loss using Micro-CT scanning and histological examination. RESULTS In the present study, AFPR inhibited osteoclast differentiation and bone resorption induced by nuclear factor-κB receptor activator (NF-κB) ligand (RANKL) in dose-/ time-dependent with no cytotoxicity. Meanwhile, AFPR decreased RANKL-mediated ROS levels and enhanced ROS scavenging enzymes. Mechanistically, AFPR promoted proteasomal degradation of TRAF6 by significantly upregulating its K48-linked ubiquitination, subsequently inhibiting NFATc1 activity. We further observed that tripartite motif protein 38 (TRIM38) could mediate the ubiquitination of TRAF6 in response to RANKL. Moreover, TRIM38 could negatively regulate the RANKL pathway by binding to TRAF6 and promoting K48-linked polyubiquitination. In addition, TRIM38 deficiency rescued the inhibition of AFPR on ROS and NFATc1 activity and osteoclastogenesis. In line with these results, AFPR reduced OP caused by OVX through ameliorating osteoclastogenesis. CONCLUSION AFPR alleviates ovariectomized-induced bone loss via suppressing ROS and NFATc1 by targeting Trim38 mediated proteasomal degradation of TRAF6. The research offers innovative perspectives on AFPR's suppressive impact in vivo OVX mouse model and in vitro, and clarifies the fundamental mechanism.
Collapse
Affiliation(s)
- Xiaoliang Feng
- Guangxi Key Laboratory of Regenerative Medicine, Orthopaedics Trauma and Hand Surgery, the First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, PR China; Collaborative Innovation Centre of Regenerative Medicine and Medical BioResource Development and Application Co-constructed by the Province and Ministry, Guangxi Medical University, Nanning, Guangxi, PR China
| | - Guining Wei
- Department of Pharmacology, Guangxi Institute of Chinese Medicine and Pharmaceutical Science, Nanning, 530022, PR China
| | - Yuangang Su
- Guangxi Key Laboratory of Regenerative Medicine, Orthopaedics Trauma and Hand Surgery, the First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, PR China; Collaborative Innovation Centre of Regenerative Medicine and Medical BioResource Development and Application Co-constructed by the Province and Ministry, Guangxi Medical University, Nanning, Guangxi, PR China
| | - Yansi Xian
- Guangxi Key Laboratory of Regenerative Medicine, Orthopaedics Trauma and Hand Surgery, the First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, PR China; Collaborative Innovation Centre of Regenerative Medicine and Medical BioResource Development and Application Co-constructed by the Province and Ministry, Guangxi Medical University, Nanning, Guangxi, PR China
| | - Zhijuan Liu
- Guangxi Key Laboratory of Regenerative Medicine, Orthopaedics Trauma and Hand Surgery, the First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, PR China; Collaborative Innovation Centre of Regenerative Medicine and Medical BioResource Development and Application Co-constructed by the Province and Ministry, Guangxi Medical University, Nanning, Guangxi, PR China
| | - Yijie Gao
- Guangxi Key Laboratory of Regenerative Medicine, Orthopaedics Trauma and Hand Surgery, the First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, PR China; Collaborative Innovation Centre of Regenerative Medicine and Medical BioResource Development and Application Co-constructed by the Province and Ministry, Guangxi Medical University, Nanning, Guangxi, PR China
| | - Jiamin Liang
- Guangxi Key Laboratory of Regenerative Medicine, Orthopaedics Trauma and Hand Surgery, the First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, PR China; Collaborative Innovation Centre of Regenerative Medicine and Medical BioResource Development and Application Co-constructed by the Province and Ministry, Guangxi Medical University, Nanning, Guangxi, PR China
| | - Haoyu Lian
- Guangxi Key Laboratory of Regenerative Medicine, Orthopaedics Trauma and Hand Surgery, the First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, PR China; Collaborative Innovation Centre of Regenerative Medicine and Medical BioResource Development and Application Co-constructed by the Province and Ministry, Guangxi Medical University, Nanning, Guangxi, PR China
| | - Jiake Xu
- School of Biomedical Sciences, the University of Western Australia, Perth, Australia; Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, PR China
| | - Jinmin Zhao
- Guangxi Key Laboratory of Regenerative Medicine, Orthopaedics Trauma and Hand Surgery, the First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, PR China; Collaborative Innovation Centre of Regenerative Medicine and Medical BioResource Development and Application Co-constructed by the Province and Ministry, Guangxi Medical University, Nanning, Guangxi, PR China
| | - Qian Liu
- Guangxi Key Laboratory of Regenerative Medicine, Orthopaedics Trauma and Hand Surgery, the First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, PR China.
| | - Fangming Song
- Guangxi Key Laboratory of Regenerative Medicine, Orthopaedics Trauma and Hand Surgery, the First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, PR China; Collaborative Innovation Centre of Regenerative Medicine and Medical BioResource Development and Application Co-constructed by the Province and Ministry, Guangxi Medical University, Nanning, Guangxi, PR China.
| |
Collapse
|
2
|
Zhang K, Lin G, Nie Z, Jin S, Bing X, Li Z, Li M. TRIM38 suppresses migration, invasion, metastasis, and proliferation in non-small cell lung cancer (NSCLC) via regulating the AMPK/NF-κB/NLRP3 pathway. Mol Cell Biochem 2024; 479:2069-2079. [PMID: 37566200 DOI: 10.1007/s11010-023-04823-y] [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: 02/09/2023] [Accepted: 07/25/2023] [Indexed: 08/12/2023]
Abstract
Accumulating data have revealed the pivotal function of tripartite motif protein 38 (TRIM38) in tumors. In view of this, this investigation aims to explore the function and potential mechanism of TRIM38 in non-small cell lung cancer (NSCLC). A xenotypic tumor model was established in vivo by subcutaneously injecting NSCLC cells (2 × 106 cells) in tail vein of each mouse. Relative expression of TRIM38 mRNA was detected via quantitative real-time polymerase chain reaction (qRT-PCR). For exploring the role of TRIM38 in vivo and in vitro, mice or NSCLC cells were divided into two groups: the vector group and the TRIM38 overexpression group. Also, protein expression levels of TRIM38, Vimentin, E-cadherin, and N-cadherin were determined using western blotting and immunohistochemistry staining. Tumor nodules of mouse lung tissues were assessed via performing H&E staining. Moreover, proliferation of NSCLC cells was evaluated through colony formation and CCK-8 assays. Further, migration and invasion of NSCLC cells were assessed through wound healing and transwell assays. Protein levels of pathway-related proteins including p-p65, p65, IκB, p-IκB, p-AMPK, AMPK, and NLRP3 were examined through western blotting analysis. Tumor lung tissues of mice and NSCLC cells showed low protein and mRNA expression of TRIM38. Functionally, up-regulation of TRIM38 reduced the number of tumor nodules and suppressed epithelial-to-mesenchymal transition (EMT) in lung tissues of mice. Furthermore, up-regulation of TRIM38 in NSCLC cells inhibited migration, invasion, EMT, and proliferation. With respect to the mechanism, in vivo experiments, the inhibitory effects of TRIM38 overexpression on tumor nodules, and EMT were reversed by AMPK inhibitor. In vitro experiments, TRIM38 overexpression caused down-regulation of p-IκB and p-p65 as well as up-regulation of p-AMPK. The inhibitory effects of TRIM38 overexpression on migration, proliferation, invasion, and EMT of NSCLC cells were reversed by overexpression of NLRP3. Concurrently, AMPK inhibitor enhanced the TRIM38-overexpressed NSCLC cell's abilities in migration, clone formation, invasion, and proliferation. TRIM38 regulated the AMPK/NF-κB/NLRP3 pathway to suppress the NSCLC's progression and development.
Collapse
Affiliation(s)
- Kaihua Zhang
- Department of Thoracic Surgery, China Aerospace Science & Industry Corporation 731 Hospital, No. 3, Zhen Gang Nan Li, Yun Gang, Feng Tai District, Beijing, 100074, China
| | - Guihu Lin
- Department of Thoracic Surgery, China Aerospace Science & Industry Corporation 731 Hospital, No. 3, Zhen Gang Nan Li, Yun Gang, Feng Tai District, Beijing, 100074, China
| | - Zhenkai Nie
- Department of Thoracic Surgery, China Aerospace Science & Industry Corporation 731 Hospital, No. 3, Zhen Gang Nan Li, Yun Gang, Feng Tai District, Beijing, 100074, China
| | - Shan Jin
- Department of Thoracic Surgery, China Aerospace Science & Industry Corporation 731 Hospital, No. 3, Zhen Gang Nan Li, Yun Gang, Feng Tai District, Beijing, 100074, China
| | - Xiaohan Bing
- Department of Thoracic Surgery, China Aerospace Science & Industry Corporation 731 Hospital, No. 3, Zhen Gang Nan Li, Yun Gang, Feng Tai District, Beijing, 100074, China
| | - Zhantao Li
- Department of Thoracic Surgery, China Aerospace Science & Industry Corporation 731 Hospital, No. 3, Zhen Gang Nan Li, Yun Gang, Feng Tai District, Beijing, 100074, China
| | - Mingru Li
- Department of Thoracic Surgery, China Aerospace Science & Industry Corporation 731 Hospital, No. 3, Zhen Gang Nan Li, Yun Gang, Feng Tai District, Beijing, 100074, China.
| |
Collapse
|
3
|
Lai Z, Li M, Yang X, Xian Z. Knockdown of the UL-16 binding protein 1 promotes osteoblast differentiation of human mesenchymal stem cells by activating the SMAD2/3 pathway. BMC Musculoskelet Disord 2024; 25:213. [PMID: 38481217 PMCID: PMC10936096 DOI: 10.1186/s12891-024-07341-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/18/2023] [Accepted: 03/06/2024] [Indexed: 03/17/2024] Open
Abstract
Osteoporosis is caused by the imbalance of osteoblasts and osteoclasts. The regulatory mechanisms of differentially expressed genes (DEGs) in pathogenesis of osteoporosis are of significant and needed to be further investigated. GSE100609 dataset downloaded from Gene Expression Omnibus (GEO) database was used to identified DEGs in osteoporosis patients. KEGG analysis was conducted to demonstrate signaling pathways related to enriched genes. Osteoporosis patients and the human mesenchymal stem cells (hMSCs) were obtained for in vivo and in vitro resaerch. Lentivirus construction and viral infection was used to knockdown genes. mRNA expression and protein expression were detected via qRT-PCR and western blot assay separately. Alkaline phosphatase (ALP) activity detection, alizarin Red S (ARS) staining, and expression of bone morphogenetic protein 2 (BMP2), osteocalcin (OCN) and Osterix were evaluated to determine osteoblast differentiation capacity. UL-16 binding protein 1 (ULBP1) gene was upregulated in osteoporosis and downregulated in differentiated hMSCs. Knockdown of ULBP1 increased ALP activity, mineralization ability evaluated by ARS staining, expression of BMP2, OCN and Osterix in differentiated hMSCs. Furthermore, rescue experiment demonstrated that suppressed ULBP1 boosted osteoblast differentiation by activating TNF-β signaling pathway. Knockdown of ULBP1 gene could promoted osteoblast differentiation by activating TNF-β signaling pathway in differentiated hMSCs. ULBP1 may be a the Achilles' heel of osteoporosis, and suppression of ULBP1 could be a promising treatment for osteoporosis.
Collapse
Affiliation(s)
- Zhen Lai
- Department of Orthopedic Surgery, Huadu District People's Hospital of Guangzhou, 48 Xinhua Road, Xinhua Street, Huadu District, Guangzhou, 510800, Guangdong, China.
| | - Mingming Li
- Shiling Town Health Center, 19 Qiling Street, Huadu District, Guangzhou, 510800, Guangdong, China
| | - Xiaodong Yang
- Department of Orthopedic Surgery, Huadu District People's Hospital of Guangzhou, 48 Xinhua Road, Xinhua Street, Huadu District, Guangzhou, 510800, Guangdong, China
| | - Zhenjie Xian
- Department of Orthopedic Surgery, Huadu District People's Hospital of Guangzhou, 48 Xinhua Road, Xinhua Street, Huadu District, Guangzhou, 510800, Guangdong, China
| |
Collapse
|
4
|
Li JY, Wang TT, Ma L, Zhang Y, Zhu D. Silencing of Jumonji domain-containing 1C inhibits the osteogenic differentiation of bone marrow mesenchymal stem cells via nuclear factor-κB signaling. World J Stem Cells 2024; 16:151-162. [PMID: 38455099 PMCID: PMC10915961 DOI: 10.4252/wjsc.v16.i2.151] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/05/2023] [Revised: 12/20/2023] [Accepted: 01/17/2024] [Indexed: 02/26/2024] Open
Abstract
BACKGROUND Osteoporosis is a common metabolic bone disorder induced by an imbalance between osteoclastic activity and osteogenic activity. During osteoporosis, bone mesenchymal stem cells (BMSCs) exhibit an increased ability to differentiate into adipocytes and a decreased ability to differentiate into osteoblasts, resulting in bone loss. Jumonji domain-containing 1C (JMJD1C) has been demonstrated to suppress osteoclastogenesis. AIM To examine the effect of JMJD1C on the osteogenesis of BMSCs and the potential underlying mechanism. METHODS BMSCs were isolated from mouse bone marrow tissues. Oil Red O staining, Alizarin red staining, alkaline phosphatase staining and the expression of adipogenic and osteogenic-associated genes were assessed to determine the differentiation of BMSCs. Bone marrow-derived macrophages (BMMs) were incubated with receptor activator of nuclear factor-kappa Β ligand to induce osteoclast differentiation, and osteoclast differentiation was confirmed by tartrate-resistant acid phosphatase staining. Other related genes were measured via reverse transcription coupled to the quantitative polymerase chain reaction and western blotting. Enzyme-linked immunosorbent assays were used to measure the levels of inflammatory cytokines, including tumor necrosis factor alpha, interleukin-6 and interleukin-1 beta. RESULTS The osteogenic and adipogenic differentiation potential of BMSCs isolated from mouse bone marrow samples was evaluated. JMJD1C mRNA and protein expression was upregulated in BMSCs after osteoblast induction, while p-nuclear factor-κB (NF-κB) and inflammatory cytokines were not significantly altered. Knockdown of JMJD1C repressed osteogenic differentiation and enhanced NF-κB activation and inflammatory cytokine release in BMSCs. Moreover, JMJD1C expression decreased during BMM osteoclast differentiation. CONCLUSION The JMJD1C/NF-κB signaling pathway is potentially involved in BMSC osteogenic differentiation and may play vital roles in the pathogenesis of osteoporosis.
Collapse
Affiliation(s)
- Jing-Yi Li
- Department of Medical Cosmetology, Beijing Tiantan Hospital, Capital Medical University, Beijing 100070, China
| | - Ting-Ting Wang
- Department of General Gerontology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan Province, China
| | - Li Ma
- Department of Plastic Surgery, China-Japan Friendship Hospital, Beijing 100029, China
| | - Yu Zhang
- Senior Department of Hematology, The Fifth Medical Centre, General Hospital of Chinese People's Liberation Army, Beijing 100071, China
| | - Di Zhu
- Department of Orthopaedic Surgery, Beijing Tiantan Hospital, Capital Medical University, Beijing 100070, China.
| |
Collapse
|
5
|
Liu RX, Gu RH, Li ZP, Hao ZQ, Hu QX, Li ZY, Wang XG, Tang W, Wang XH, Zeng YK, Li ZW, Dong Q, Zhu XF, Chen D, Zhao KW, Zhang RH, Zha ZG, Zhang HT. Trim21 depletion alleviates bone loss in osteoporosis via activation of YAP1/β-catenin signaling. Bone Res 2023; 11:56. [PMID: 37884520 PMCID: PMC10603047 DOI: 10.1038/s41413-023-00296-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2023] [Revised: 08/26/2023] [Accepted: 09/22/2023] [Indexed: 10/28/2023] Open
Abstract
Despite the diverse roles of tripartite motif (Trim)-containing proteins in the regulation of autophagy, the innate immune response, and cell differentiation, their roles in skeletal diseases are largely unknown. We recently demonstrated that Trim21 plays a crucial role in regulating osteoblast (OB) differentiation in osteosarcoma. However, how Trim21 contributes to skeletal degenerative disorders, including osteoporosis, remains unknown. First, human and mouse bone specimens were evaluated, and the results showed that Trim21 expression was significantly elevated in bone tissues obtained from osteoporosis patients. Next, we found that global knockout of the Trim21 gene (KO, Trim21-/-) resulted in higher bone mass compared to that of the control littermates. We further demonstrated that loss of Trim21 promoted bone formation by enhancing the osteogenic differentiation of bone marrow mesenchymal stem cells (BMSCs) and elevating the activity of OBs; moreover, Trim21 depletion suppressed osteoclast (OC) formation of RAW264.7 cells. In addition, the differentiation of OCs from bone marrow-derived macrophages (BMMs) isolated from Trim21-/- and Ctsk-cre; Trim21f/f mice was largely compromised compared to that of the littermate control mice. Mechanistically, YAP1/β-catenin signaling was identified and demonstrated to be required for the Trim21-mediated osteogenic differentiation of BMSCs. More importantly, the loss of Trim21 prevented ovariectomy (OVX)- and lipopolysaccharide (LPS)-induced bone loss in vivo by orchestrating the coupling of OBs and OCs through YAP1 signaling. Our current study demonstrated that Trim21 is crucial for regulating OB-mediated bone formation and OC-mediated bone resorption, thereby providing a basis for exploring Trim21 as a novel dual-targeting approach for treating osteoporosis and pathological bone loss.
Collapse
Affiliation(s)
- Ri-Xu Liu
- Department of Bone and Joint Surgery, the First Affiliated Hospital of Jinan University; Key Laboratory of Regenerative Medicine of Ministry of Education, Jinan University, Guangzhou, 510630, Guangdong, China
- Department of Orthopedic and Spine Surgery, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, Guangdong, China
| | - Rong-He Gu
- School of Basic Medical Sciences of Guangxi Medical University, the Fifth Affiliated Hospital of Guangxi Medical University, Nanning, 530022, Guangxi, China
| | - Zhi-Peng Li
- Department of Bone and Joint Surgery, the First Affiliated Hospital of Jinan University; Key Laboratory of Regenerative Medicine of Ministry of Education, Jinan University, Guangzhou, 510630, Guangdong, China
| | - Zhi-Quan Hao
- Department of Bone and Joint Surgery, the First Affiliated Hospital of Jinan University; Key Laboratory of Regenerative Medicine of Ministry of Education, Jinan University, Guangzhou, 510630, Guangdong, China
| | - Qin-Xiao Hu
- Department of Bone and Joint Surgery, the First Affiliated Hospital of Jinan University; Key Laboratory of Regenerative Medicine of Ministry of Education, Jinan University, Guangzhou, 510630, Guangdong, China
| | - Zhen-Yan Li
- Department of Bone and Joint Surgery, the First Affiliated Hospital of Jinan University; Key Laboratory of Regenerative Medicine of Ministry of Education, Jinan University, Guangzhou, 510630, Guangdong, China
| | - Xiao-Gang Wang
- Key Laboratory of Big Data-Based Precision Medicine, School of Engineering Medicine, Beihang University, 100191, Beijing, China
| | - Wang Tang
- Department of Bone and Joint Surgery, the First Affiliated Hospital of Jinan University; Key Laboratory of Regenerative Medicine of Ministry of Education, Jinan University, Guangzhou, 510630, Guangdong, China
| | - Xiao-He Wang
- Department of Bone and Joint Surgery, the First Affiliated Hospital of Jinan University; Key Laboratory of Regenerative Medicine of Ministry of Education, Jinan University, Guangzhou, 510630, Guangdong, China
| | - Yu-Kai Zeng
- Department of Bone and Joint Surgery, the First Affiliated Hospital of Jinan University; Key Laboratory of Regenerative Medicine of Ministry of Education, Jinan University, Guangzhou, 510630, Guangdong, China
| | - Zhen-Wei Li
- Department of Bone and Joint Surgery, the First Affiliated Hospital of Jinan University; Key Laboratory of Regenerative Medicine of Ministry of Education, Jinan University, Guangzhou, 510630, Guangdong, China
| | - Qiu Dong
- Department of Bone and Joint Surgery, the First Affiliated Hospital of Jinan University; Key Laboratory of Regenerative Medicine of Ministry of Education, Jinan University, Guangzhou, 510630, Guangdong, China
| | - Xiao-Feng Zhu
- Guangdong Provincial Key Laboratory of Traditional Chinese Medicine Informatization, College of Pharmacy, Jinan University, Guangzhou, 510630, Guangdong, China
| | - Di Chen
- Research Center for Computer-aided Drug Discovery, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, 518005, Shenzhen, China
| | - Ke-Wei Zhao
- Guangzhou Key Laboratory of Chinese Medicine Research on Prevention and Treatment of Osteoporosis, the Third Affiliated Hospital of Guangzhou University of Chinese Medicine, 510375, Guangzhou, China
| | - Rong-Hua Zhang
- Guangdong Provincial Key Laboratory of Traditional Chinese Medicine Informatization, College of Pharmacy, Jinan University, Guangzhou, 510630, Guangdong, China.
| | - Zhen-Gang Zha
- Department of Bone and Joint Surgery, the First Affiliated Hospital of Jinan University; Key Laboratory of Regenerative Medicine of Ministry of Education, Jinan University, Guangzhou, 510630, Guangdong, China.
| | - Huan-Tian Zhang
- Department of Bone and Joint Surgery, the First Affiliated Hospital of Jinan University; Key Laboratory of Regenerative Medicine of Ministry of Education, Jinan University, Guangzhou, 510630, Guangdong, China.
| |
Collapse
|
6
|
Yao X, Dong R, Hu S, Liu Z, Hu F, Cheng X, Wang X, Ma T, Tian S, Zhang XJ, Hu Y, Bai L, Li H, Zhang P. Tripartite motif 38 alleviates the pathological process of NAFLD/NASH by promoting TAB2 degradation. J Lipid Res 2023:100382. [PMID: 37116711 PMCID: PMC10394331 DOI: 10.1016/j.jlr.2023.100382] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Revised: 04/18/2023] [Accepted: 04/19/2023] [Indexed: 04/30/2023] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD) has become the most prevalent chronic liver disease worldwide, without any FDA-approved pharmacological intervention in clinic. The TRIM (tripartite motif-containing) family plays essential roles in innate immune and hepatic inflammation. TRIM38, as one of the important members in TRIM family, was largely reported to be involved in the regulation of innate immune and inflammatory responses. However, the functional roles of TRIM38 in NAFLD remains largely unknown. Here, the expression of TRIM38 was first detected in liver samples of both NAFLD mice model and patients diagnosed with NAFLD. We found TRIM38 expression was downregulated in NAFLD liver tissues compared with normal liver tissues. Genetic TRIM38 knockout in vivo showed that TRIM38 depletion deteriorated the HFD and HFHC diet-induced hepatic steatosis and HFHC diet-induced liver inflammation and fibrosis. In particular, we found that the effects of hepatocellular lipid accumulation and inflammation induced by palmitic acid and oleic acid (PA+OA) was aggravated by TRIM38 depletion but mitigated by TRIM38 overexpression in vitro. Mechanically, RNA-seq analysis demonstrated that TRIM38 ameliorated NASH progression by attenuating the activating of mitogen-activated protein kinase (MAPK) signaling pathway. We further found that TRIM38 interacted with TGF-β-activated kinase 1 (TAK1) binding protein 2 (TAB2) and promoted its protein degradation, thus inhibiting the TAK1-MAPK signal cascades. In summary, our study revealed that TRIM38 could suppress hepatic steatosis, inflammatory and fibrosis in NAFLD via promoting TAB2 degradation. TRIM38 could be a potential target for NAFLD treatment.
Collapse
Affiliation(s)
- Xinxin Yao
- Basic Medical School, Wuhan University, Wuhan, China; Institute of Model Animal, Wuhan University, Wuhan, China
| | - Ruixiang Dong
- Basic Medical School, Wuhan University, Wuhan, China; Institute of Model Animal, Wuhan University, Wuhan, China
| | - Sha Hu
- Basic Medical School, Wuhan University, Wuhan, China; Institute of Model Animal, Wuhan University, Wuhan, China
| | - Zhen Liu
- Institute of Model Animal, Wuhan University, Wuhan, China
| | - Fengjiao Hu
- Institute of Model Animal, Wuhan University, Wuhan, China; Medical Science Research Center, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Xu Cheng
- Gannan Innovation and Translational Medicine Research Institute, Ganzhou, China; Key Laboratory of Cardiovascular Disease Prevention and Control, Ministry of Education, First Affiliated Hospital of Gannan Medical University, Gannan Medical University, Ganzhou, China
| | - Xiaoming Wang
- Basic Medical School, Wuhan University, Wuhan, China; Institute of Model Animal, Wuhan University, Wuhan, China
| | - Tengfei Ma
- Department of Neurology, Huanggang Central Hospital, Huanggang, China
| | - Song Tian
- Basic Medical School, Wuhan University, Wuhan, China; Institute of Model Animal, Wuhan University, Wuhan, China
| | - Xiao-Jing Zhang
- Basic Medical School, Wuhan University, Wuhan, China; Institute of Model Animal, Wuhan University, Wuhan, China
| | - Yufeng Hu
- Gannan Innovation and Translational Medicine Research Institute, Ganzhou, China; Key Laboratory of Cardiovascular Disease Prevention and Control, Ministry of Education, First Affiliated Hospital of Gannan Medical University, Gannan Medical University, Ganzhou, China
| | - Lan Bai
- Gannan Innovation and Translational Medicine Research Institute, Ganzhou, China; Key Laboratory of Cardiovascular Disease Prevention and Control, Ministry of Education, First Affiliated Hospital of Gannan Medical University, Gannan Medical University, Ganzhou, China.
| | - Hongliang Li
- Basic Medical School, Wuhan University, Wuhan, China; Institute of Model Animal, Wuhan University, Wuhan, China; Medical Science Research Center, Zhongnan Hospital of Wuhan University, Wuhan, China; Gannan Innovation and Translational Medicine Research Institute, Ganzhou, China; Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China.
| | - Peng Zhang
- Basic Medical School, Wuhan University, Wuhan, China; Institute of Model Animal, Wuhan University, Wuhan, China.
| |
Collapse
|
7
|
Multiple Genetic Loci Associated with Pug Dog Thoracolumbar Myelopathy. Genes (Basel) 2023; 14:genes14020385. [PMID: 36833311 PMCID: PMC9957375 DOI: 10.3390/genes14020385] [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: 12/21/2022] [Revised: 01/24/2023] [Accepted: 01/27/2023] [Indexed: 02/04/2023] Open
Abstract
Pug dogs with thoracolumbar myelopathy (PDM) present with a specific clinical phenotype that includes progressive pelvic limb ataxia and paresis, commonly accompanied by incontinence. Vertebral column malformations and lesions, excessive scar tissue of the meninges, and central nervous system inflammation have been described. PDM has a late onset and affects more male than female dogs. The breed-specific presentation of the disorder suggests that genetic risk factors are involved in the disease development. To perform a genome-wide search for PDM-associated loci, we applied a Bayesian model adapted for mapping complex traits (BayesR) and a cross-population extended haplotype homozygosity test (XP-EHH) in 51 affected and 38 control pugs. Nineteen associated loci (harboring 67 genes in total, including 34 potential candidate genes) and three candidate regions under selection (with four genes within or next to the signal) were identified. The multiple candidate genes identified have implicated functions in bone homeostasis, fibrotic scar tissue, inflammatory responses, or the formation, regulation, and differentiation of cartilage, suggesting the potential relevance of these processes to the pathogenesis of PDM.
Collapse
|
8
|
E3 Ubiquitin Ligases: Potential Therapeutic Targets for Skeletal Pathology and Degeneration. Stem Cells Int 2022; 2022:6948367. [PMID: 36203882 PMCID: PMC9532118 DOI: 10.1155/2022/6948367] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Revised: 06/06/2022] [Accepted: 09/03/2022] [Indexed: 11/18/2022] Open
Abstract
The ubiquitination-proteasome system (UPS) is crucial in regulating a variety of cellular processes including proliferation, differentiation, and survival. Ubiquitin protein ligase E3 is the most critical molecule in the UPS system. Dysregulation of the UPS system is associated with many conditions. Over the past few decades, there have been an increasing number of studies focusing on the UPS system and how it affects bone metabolism. Multiple E3 ubiquitin ligases have been found to mediate osteogenesis or osteolysis through a variety of pathways. In this review, we describe the mechanisms of UPS, especially E3 ubiquitin ligases on bone metabolism. To date, many E3 ubiquitin ligases have been found to regulate osteogenesis or osteoclast differentiation. We review the classification of these E3 enzymes and the mechanisms that influence upstream and downstream molecules and transduction pathways. Finally, this paper reviews the discovery of the relevant UPS inhibitors, drug molecules, and noncoding RNAs so far and prospects the future research and treatment.
Collapse
|
9
|
Shen H, Gong Q, Zhang J, Wang H, Qiu Q, Zhang J, Luo D. TRIM46 aggravated high glucose-induced hyper permeability and inflammatory response in human retinal capillary endothelial cells by promoting IκBα ubiquitination. EYE AND VISION 2022; 9:35. [PMID: 36064447 PMCID: PMC9443035 DOI: 10.1186/s40662-022-00305-2] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Accepted: 08/15/2022] [Indexed: 11/10/2022]
Abstract
Background Diabetic retinopathy (DR) as a severe diabetic complication contributes to blindness. The increased permeability of retinal capillary endothelial cells (RCECs) as well as the production of inflammatory markers are closely related to DR occurrence. We recently revealed that TRIM46 promotes high glucose (HG)-caused ferroptosis in human RCECs (HRCECs). The current study aims to explore the molecular mechanism of how TRIM46 plays its role in DR progression. Methods Western blot was utilized to determine protein expression. The cell counting kit-8 assay was used to observe cell viability. The permeability of the cell layer was determined by measuring the transepithelial electrical resistance and fluorescein isothiocyanate (FITC)-dextran leak. Enzyme-linked immunosorbent assay was used to quantify the protein level of pro-inflammatory cytokines and co-immunoprecipitation was employed to verify the relationship between TRIM46 and IκBα. Results HG dramatically upregulated TRIM46 protein expression in a dose-dependent way. Silencing TRIM46 effectively reversed HG-induced cell growth inhibition, cell cycle arrest, hyper permeability and pro-inflammatory cytokines secretion in HRCECs, while overexpression of TRIM46 exhibited an opposite effect. Furthermore, TRIM46 was able to interact with IκBα and promote the ubiquitination and degradation of IκBα. IκBα overexpression recovered the effects of TRIM46 overexpression in HRCECs. Furthermore, inhibiting the activation of NF-κB partially recovered HG-induced HRCEC injury, whereas TRIM46 overexpression reversed these effects. Conclusion This study demonstrates that TRIM46 interacts with IκBα to activate the NF-κB signaling pathway, thereby enhancing cell proliferation inhibition, hyper permeability and the inflammatory response of HRCECs in a HG state. Supplementary Information The online version contains supplementary material available at 10.1186/s40662-022-00305-2.
Collapse
|
10
|
Xu X, Li Y, Shi L, He K, Sun Y, Ding Y, Meng B, Zhang J, Xiang L, Dong J, Liu M, Zhang J, Xiang L, Xiang G. Myeloid-derived growth factor (MYDGF) protects bone mass through inhibiting osteoclastogenesis and promoting osteoblast differentiation. EMBO Rep 2022; 23:e53509. [PMID: 35068044 PMCID: PMC8892248 DOI: 10.15252/embr.202153509] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2021] [Revised: 12/15/2021] [Accepted: 12/21/2021] [Indexed: 12/13/2022] Open
Abstract
Whether bone marrow regulates bone metabolism through endocrine and paracrine mechanism remains largely unknown. Here, we found that (i) myeloid cell-specific myeloid-derived growth factor (MYDGF) deficiency decreased bone mass and bone strength in young and aged mice; (ii) myeloid cell-specific MYDGF restoration prevented decreases in bone mass and bone strength in MYDGF knockout mice; moreover, myeloid cell-derived MYDGF improved the progress of bone defects healing, prevented ovariectomy (OVX)-induced bone loss and age-related osteoporosis; (iii) MYDGF inhibited osteoclastogenesis and promoted osteoblast differentiation in vivo and in vitro; and (iv) PKCβ-NF-κB and MAPK1/3-STAT3 pathways were involved in the regulation of MYDGF on bone metabolism. Thus, we concluded that myeloid cell-derived MYDGF is a positive regulator of bone homeostasis by inhibiting bone resorption and promoting bone formation. MYDGF may become a potential novel therapeutic drug for osteoporosis, and bone marrow may become a potential therapeutic target for bone metabolic disorders.
Collapse
Affiliation(s)
- Xiaoli Xu
- Department of EndocrinologyGeneral Hospital of Central Theater CommandWuhanChina,The First School of Clinical MedicineSouthern Medical UniversityGuangzhouChina
| | - Yixiang Li
- Department of Hematology and Medical OncologySchool of MedicineEmory UniversityAtlantaGAUSA
| | - Lingfeng Shi
- Department of EndocrinologyGeneral Hospital of Central Theater CommandWuhanChina,The First School of Clinical MedicineSouthern Medical UniversityGuangzhouChina
| | - Kaiyue He
- Department of EndocrinologyGeneral Hospital of Central Theater CommandWuhanChina,The First School of Clinical MedicineSouthern Medical UniversityGuangzhouChina
| | - Ying Sun
- Department of EndocrinologyGeneral Hospital of Central Theater CommandWuhanChina
| | - Yan Ding
- Department of EndocrinologyGeneral Hospital of Central Theater CommandWuhanChina,The First School of Clinical MedicineSouthern Medical UniversityGuangzhouChina
| | - Biying Meng
- Department of EndocrinologyGeneral Hospital of Central Theater CommandWuhanChina,The First School of Clinical MedicineSouthern Medical UniversityGuangzhouChina
| | - Jiajia Zhang
- Department of EndocrinologyGeneral Hospital of Central Theater CommandWuhanChina
| | - Lin Xiang
- Department of EndocrinologyGeneral Hospital of Central Theater CommandWuhanChina
| | - Jing Dong
- Department of EndocrinologyGeneral Hospital of Central Theater CommandWuhanChina
| | - Min Liu
- Department of EndocrinologyGeneral Hospital of Central Theater CommandWuhanChina
| | - Junxia Zhang
- Department of EndocrinologyGeneral Hospital of Central Theater CommandWuhanChina,The First School of Clinical MedicineSouthern Medical UniversityGuangzhouChina
| | - Lingwei Xiang
- Centers for Surgery and Public HealthBrigham and Women's HospitalBostonMAUSA
| | - Guangda Xiang
- Department of EndocrinologyGeneral Hospital of Central Theater CommandWuhanChina,The First School of Clinical MedicineSouthern Medical UniversityGuangzhouChina
| |
Collapse
|
11
|
Yin H, Guo X, Chen Y, Zeng Y, Mo X, Hong S, He H, Li J, Steinmetz R, Liu Q. TAB2 deficiency induces dilated cardiomyopathy by promoting RIPK1-dependent apoptosis and necroptosis. J Clin Invest 2022; 132:152297. [PMID: 34990405 PMCID: PMC8843707 DOI: 10.1172/jci152297] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Accepted: 01/04/2022] [Indexed: 02/01/2023] Open
Abstract
Mutations in TGF-β-activated kinase 1 binding protein 2 (TAB2) have been implicated in the pathogenesis of dilated cardiomyopathy and/or congenital heart disease in humans, but the underlying mechanisms are currently unknown. Here, we identified an indispensable role for TAB2 in regulating myocardial homeostasis and remodeling by suppressing receptor-interacting protein kinase 1 (RIPK1) activation and RIPK1-dependent apoptosis and necroptosis. Cardiomyocyte-specific deletion of Tab2 in mice triggered dilated cardiomyopathy with massive apoptotic and necroptotic cell death. Moreover, Tab2-deficient mice were also predisposed to myocardial injury and adverse remodeling after pathological stress. In cardiomyocytes, deletion of TAB2 but not its close homolog TAB3 promoted TNF-α-induced apoptosis and necroptosis, which was rescued by forced activation of TAK1 or inhibition of RIPK1 kinase activity. Mechanistically, TAB2 critically mediates RIPK1 phosphorylation at Ser321 via a TAK1-dependent mechanism, which prevents RIPK1 kinase activation and the formation of RIPK1-FADD-caspase-8 apoptotic complex or RIPK1-RIPK3 necroptotic complex. Strikingly, genetic inactivation of RIPK1 with Ripk1-K45A knockin effectively rescued cardiac remodeling and dysfunction in Tab2-deficient mice. Together, these data demonstrated that TAB2 is a key regulator of myocardial homeostasis and remodeling by suppressing RIPK1-dependent apoptosis and necroptosis. Our results also suggest that targeting RIPK1-mediated cell death signaling may represent a promising therapeutic strategy for TAB2 deficiency-induced dilated cardiomyopathy.
Collapse
Affiliation(s)
- Haifeng Yin
- Department of Physiology and Biophysics, University of Washington, Seattle, Washington, USA
| | - Xiaoyun Guo
- Department of Physiology and Biophysics, University of Washington, Seattle, Washington, USA
| | - Yi Chen
- Department of Physiology and Biophysics, University of Washington, Seattle, Washington, USA
| | - Yachang Zeng
- Department of Physiology and Biophysics, University of Washington, Seattle, Washington, USA
| | - Xiaoliang Mo
- Department of Physiology and Biophysics, University of Washington, Seattle, Washington, USA
| | - Siqi Hong
- Department of Physiology and Biophysics, University of Washington, Seattle, Washington, USA
| | - Hui He
- Department of Physiology and Biophysics, University of Washington, Seattle, Washington, USA
| | - Jing Li
- Department of Physiology and Biophysics, University of Washington, Seattle, Washington, USA
| | - Rachel Steinmetz
- Department of Physiology and Biophysics, University of Washington, Seattle, Washington, USA
| | - Qinghang Liu
- Department of Physiology and Biophysics, University of Washington, Seattle, Washington, USA
| |
Collapse
|
12
|
Xian J, Liang D, Zhao C, Chen Y, Zhu Q. TRIM21 inhibits the osteogenic differentiation of mesenchymal stem cells by facilitating K48 ubiquitination-mediated degradation of Akt. Exp Cell Res 2022; 412:113034. [DOI: 10.1016/j.yexcr.2022.113034] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2021] [Revised: 01/11/2022] [Accepted: 01/13/2022] [Indexed: 01/18/2023]
|
13
|
Micale L, Morlino S, Carbone A, Carissimo A, Nardella G, Fusco C, Palumbo O, Schirizzi A, Russo F, Mazzoccoli G, Breckpot J, De Luca C, Ferraris A, Giunta C, Grammatico P, Haanpää MK, Mancano G, Forzano G, Cacchiarelli D, Van Esch H, Callewaert B, Rohrbach M, Castori M. Loss-of-function variants in exon 4 of TAB2 cause a recognizable multisystem disorder with cardiovascular, facial, cutaneous, and musculoskeletal involvement. Genet Med 2021; 24:439-453. [PMID: 34906501 DOI: 10.1016/j.gim.2021.10.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Revised: 09/02/2021] [Accepted: 10/15/2021] [Indexed: 11/19/2022] Open
Abstract
PURPOSE This study aimed to describe a multisystemic disorder featuring cardiovascular, facial, musculoskeletal, and cutaneous anomalies caused by heterozygous loss-of-function variants in TAB2. METHODS Affected individuals were analyzed by next-generation technologies and genomic array. The presumed loss-of-function effect of identified variants was assessed by luciferase assay in cells transiently expressing TAB2 deleterious alleles. In available patients' fibroblasts, variant pathogenicity was further explored by immunoblot and osteoblast differentiation assays. The transcriptomic profile of fibroblasts was investigated by RNA sequencing. RESULTS A total of 11 individuals from 8 families were heterozygotes for a novel TAB2 variant. In total, 7 variants were predicted to be null alleles and 1 was a missense change. An additional subject was heterozygous for a 52 kb microdeletion involving TAB2 exons 1 to 3. Luciferase assay indicated a decreased transcriptional activation mediated by NF-κB signaling for all point variants. Immunoblot analysis showed a reduction of TAK1 phosphorylation while osteoblast differentiation was impaired. Transcriptomic analysis identified deregulation of multiple pleiotropic pathways, such as TGFβ-, Ras-MAPK-, and Wnt-signaling networks. CONCLUSION Our data defined a novel disorder associated with loss-of-function or, more rarely, hypomorphic alleles in a restricted linker region of TAB2. The pleiotropic manifestations in this disorder partly recapitulate the 6q25.1 (TAB2) microdeletion syndrome and deserve the definition of cardio-facial-cutaneous-articular syndrome.
Collapse
Affiliation(s)
- Lucia Micale
- Division of Medical Genetics, Fondazione IRCCS-Casa Sollievo della Sofferenza, San Giovanni Rotondo, Italy
| | - Silvia Morlino
- Division of Medical Genetics, Fondazione IRCCS-Casa Sollievo della Sofferenza, San Giovanni Rotondo, Italy
| | - Annalucia Carbone
- Unit of Chronobiology, Division of Internal Medicine, Fondazione IRCCS-Casa Sollievo della Sofferenza, San Giovanni Rotondo, Italy
| | - Annamaria Carissimo
- Institute for Applied Mathematics "Mauro Picone" National Research Council, Naples, Italy
| | - Grazia Nardella
- Division of Medical Genetics, Fondazione IRCCS-Casa Sollievo della Sofferenza, San Giovanni Rotondo, Italy
| | - Carmela Fusco
- Division of Medical Genetics, Fondazione IRCCS-Casa Sollievo della Sofferenza, San Giovanni Rotondo, Italy
| | - Orazio Palumbo
- Division of Medical Genetics, Fondazione IRCCS-Casa Sollievo della Sofferenza, San Giovanni Rotondo, Italy
| | - Annalisa Schirizzi
- Division of Medical Genetics, Fondazione IRCCS-Casa Sollievo della Sofferenza, San Giovanni Rotondo, Italy
| | - Federica Russo
- Division of Medical Genetics, Fondazione IRCCS-Casa Sollievo della Sofferenza, San Giovanni Rotondo, Italy
| | - Gianluigi Mazzoccoli
- Unit of Chronobiology, Division of Internal Medicine, Fondazione IRCCS-Casa Sollievo della Sofferenza, San Giovanni Rotondo, Italy
| | - Jeroen Breckpot
- Center for Human Genetics, University Hospital Leuven, Leuven, Belgium
| | - Chiara De Luca
- Center for Human Genetics, University Hospital Leuven, Leuven, Belgium
| | - Alessandro Ferraris
- Laboratory of Medical Genetics, Department of Molecular Medicine, Sapienza University, San Camillo-Forlanini Hospital, Rome, Italy
| | - Cecilia Giunta
- Division of Metabolism and Children's Research Center, University Children's Hospital Zurich and University of Zurich, Zurich, Switzerland
| | - Paola Grammatico
- Laboratory of Medical Genetics, Department of Molecular Medicine, Sapienza University, San Camillo-Forlanini Hospital, Rome, Italy
| | - Maria K Haanpää
- Department of Clinical Genetics and Genomics, Turku University Hospital and University of Turku, Turku, Finland
| | - Giorgia Mancano
- Medical Genetics Unit, Meyer Children's University Hospital, Florence, Italy
| | - Giulia Forzano
- Medical Genetics Unit, University of Florence, Florence, Italy
| | - Davide Cacchiarelli
- Telethon Institute of Genetics and Medicine (TIGEM), Naples, Italy; Department of Translational Medicine, University of Naples "Federico II", Naples, Italy
| | - Hilde Van Esch
- Center for Human Genetics, University Hospital Leuven, Leuven, Belgium
| | - Bert Callewaert
- Center for Medical Genetics and Department of Biomolecular Medicine, Ghent University Hospital, Ghent, Belgium
| | - Marianne Rohrbach
- Division of Metabolism and Children's Research Center, University Children's Hospital Zurich and University of Zurich, Zurich, Switzerland
| | - Marco Castori
- Division of Medical Genetics, Fondazione IRCCS-Casa Sollievo della Sofferenza, San Giovanni Rotondo, Italy.
| |
Collapse
|
14
|
TRIM proteins in fibrosis. Biomed Pharmacother 2021; 144:112340. [PMID: 34678729 DOI: 10.1016/j.biopha.2021.112340] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Revised: 10/12/2021] [Accepted: 10/13/2021] [Indexed: 02/06/2023] Open
Abstract
Fibrosis is an outcome of tissue repair after different types of injuries. The homeostasis of extracellular matrix is broken, and excessive deposition occurs, affecting the normal function of tissues and organs, which could become prostrated in serious cases.Finding a suitable target to regulate the repair process and reduce the damage caused by fibrosis is a hot research topic at present. The TRIM family is number of one of the E3 ubiquitin ligase subfamilies and participates in various biological processes including intracellular signal transduction, apoptosis, autophagy, and immunity by regulating the ubiquitination of target proteins. For the past few years, the important role of TRIM in the occurrence and development of fibrosis has been gradually revealed. In this review, we focus on the recent emerging topics on TRIM proteins in the regulation of fibrosis, fibrosis-related cytokines and pathways.
Collapse
|
15
|
Hu S, Li Y, Wang B, Peng K. TRIM38 protects chondrocytes from IL-1β-induced apoptosis and degeneration via negatively modulating nuclear factor (NF)-κB signaling. Int Immunopharmacol 2021; 99:108048. [PMID: 34426118 DOI: 10.1016/j.intimp.2021.108048] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2021] [Revised: 07/27/2021] [Accepted: 08/02/2021] [Indexed: 12/14/2022]
Abstract
Tripartite motif protein 38 (TRIM38) has been documented as a vital modulator of inflammation. However, the relevance of TRIM38 in osteoarthritis is not yet known. In this work, we aimed to explore any possible effects of TRIM38 on interleukin-1β (IL-1β)-stimulated chondrocytes, an in vitro cellular model of osteoarthritis. Analyzing our data showed significant decreases in the levels of TRIM38 in chondrocytes following IL-1β stimulation. Gain-of-function studies revealed that overexpression of TRIM38 markedly increased the viability of IL-1β-stimulated chondrocytes while decreasing their rate of apoptosis and degeneration. Conversely, depletion of TRIM38 enhanced the sensitivity of chondrocytes to IL-1β-induced injury. Further research demonstrated that TRIM38 was capable of inhibiting IL-1β-induced activation of nuclear factor (NF)-κB signaling. Reactivation of NF-κB markedly reversed TRIM38-overexpression-mediated effects, while inhibition of NF-κB significantly abolished TRIM38-depletion-induced effects in IL-1β-stimulated chondrocytes. In summary, the findings of this work suggest that TRIM38 is capable of ameliorating IL-1β-induced apoptosis and degeneration of chondrocytes via suppression of NF-κB signaling. Our work indicates a potential role of TRIM38 in osteoarthritis and proposes it as a new therapeutic target for osteoarthritis.
Collapse
Affiliation(s)
- Shouye Hu
- Department of Joint Surgery, Xi'an Hong Hui Hospital, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi Province 710054, PR China
| | - Yanqi Li
- Department of Respiratory, Xi'an Children's Hospital, Xi'an, Shaanxi Province 710003, PR China
| | - Bo Wang
- Department of Joint Surgery, Xi'an Hong Hui Hospital, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi Province 710054, PR China
| | - Kan Peng
- Department of Joint Surgery, Xi'an Hong Hui Hospital, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi Province 710054, PR China.
| |
Collapse
|
16
|
Guan S, Li L, Chen WS, Jiang WY, Ding Y, Zhao LL, Shi YF, Wang J, Gui Q, Xu CC, Cheng Y, Zhang W. Circular RNA WHSC1 exerts oncogenic properties by regulating miR-7/TAB2 in lung cancer. J Cell Mol Med 2021; 25:9784-9795. [PMID: 34551195 PMCID: PMC8505844 DOI: 10.1111/jcmm.16925] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Revised: 05/28/2021] [Accepted: 09/01/2021] [Indexed: 12/24/2022] Open
Abstract
Circular RNA is a newly discovered member of non‐coding RNA (ncRNA) and regulates the target gene by acting as a micro‐RNA sponge. It plays vital roles in various diseases. However, the functions of circular RNA in non‐small cell lung cancer (NSCLC) remain still unclear. Our data showed that circ‐WHSC1 was highly expressed in NSCLC cells and tissues. Both in vitro and in vivo experiments showed that circ‐WHSC1 promoted NSCLC proliferation. circ‐WHSC1 also promoted the migration and invasion of lung cancer cells. Through bioinformatic analysis and functional experiments, we showed that circ‐WHSC1 could act as a sponge for micro‐RNA‐7 (miR‐7) and regulate the expression of TAB2 (TGF‐beta activated kinase one binding protein two). Inhibition of the circ‐WHSC1/miR‐7/TAB2 pathway could effectively attenuate lung cancer progression. In summary, this study confirmed the existence and oncogenic function of circ‐WHSC1 in NSCLC. The research suggests that the circ‐WHSC1/miR‐7/TAB2 axis might be a potential target for NSCLC therapy.
Collapse
Affiliation(s)
- Sisi Guan
- Department of Geriatrics, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Li Li
- Department of Geriatrics, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Wen-Shu Chen
- Department of Thoracic Surgery, Shengli Clinical Medical College of Fujian Medical University, Fuzhou, China
| | - Wen-Yang Jiang
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, China
| | - Yun Ding
- Department of Thoracic Surgery, Shengli Clinical Medical College of Fujian Medical University, Fuzhou, China
| | - Li-Lan Zhao
- Department of Thoracic Surgery, Shengli Clinical Medical College of Fujian Medical University, Fuzhou, China
| | - Yi-Fan Shi
- Department of Thoracic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Jun Wang
- Department of Thoracic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Qi Gui
- Department of Thoracic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Cheng-Cheng Xu
- Department of Oncology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Yang Cheng
- Department of Geriatrics, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Wenjuan Zhang
- Department of Geriatrics, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
17
|
Gu Z, Chen X, Yang W, Qi Y, Yu H, Wang X, Gong Y, Chen Q, Zhong B, Dai L, Qi S, Zhang Z, Zhang H, Hu H. The SUMOylation of TAB2 mediated by TRIM60 inhibits MAPK/NF-κB activation and the innate immune response. Cell Mol Immunol 2021; 18:1981-1994. [PMID: 33184450 PMCID: PMC8322076 DOI: 10.1038/s41423-020-00564-w] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Accepted: 09/27/2020] [Indexed: 02/05/2023] Open
Abstract
Activation of the TAK1 signalosome is crucial for mediating the innate immune response to pathogen invasion and is regulated by multiple layers of posttranslational modifications, including ubiquitination, SUMOylation, and phosphorylation; however, the underlying molecular mechanism is not fully understood. In this study, TRIM60 negatively regulated the formation and activation of the TAK1 signalosome. Deficiency of TRIM60 in macrophages led to enhanced MAPK and NF-κB activation, accompanied by elevated levels of proinflammatory cytokines but not IFN-I. Immunoprecipitation-mass spectrometry assays identified TAB2 as the target of TRIM60 for SUMOylation rather than ubiquitination, resulting in impaired formation of the TRAF6/TAB2/TAK1 complex and downstream MAPK and NF-κB pathways. The SUMOylation sites of TAB2 mediated by TRIM60 were identified as K329 and K562; substitution of these lysines with arginines abolished the SUMOylation of TAB2. In vivo experiments showed that TRIM60-deficient mice showed an elevated immune response to LPS-induced septic shock and L. monocytogenes infection. Our data reveal that SUMOylation of TAB2 mediated by TRIM60 is a novel mechanism for regulating the innate immune response, potentially paving the way for a new strategy to control antibacterial immune responses.
Collapse
Affiliation(s)
- Zhiwen Gu
- Department of Rheumatology and Immunology, National Clinical Research Center for Geriatrics, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, and Collaborative Innovation Center of Biotherapy, Chengdu, 610041, China
| | - Xueying Chen
- Department of Rheumatology and Immunology, National Clinical Research Center for Geriatrics, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, and Collaborative Innovation Center of Biotherapy, Chengdu, 610041, China
| | - Wenyong Yang
- Department of Rheumatology and Immunology, National Clinical Research Center for Geriatrics, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, and Collaborative Innovation Center of Biotherapy, Chengdu, 610041, China
| | - Yu Qi
- Department of Rheumatology and Immunology, National Clinical Research Center for Geriatrics, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, and Collaborative Innovation Center of Biotherapy, Chengdu, 610041, China
| | - Hui Yu
- Department of Rheumatology and Immunology, National Clinical Research Center for Geriatrics, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, and Collaborative Innovation Center of Biotherapy, Chengdu, 610041, China
| | - Xiaomeng Wang
- Department of Virology, College of Life Sciences, Department of Immunology, Medical Research Institute, Wuhan University, Wuhan, 430072, China
| | - Yanqiu Gong
- Department of General Practice and Lab of PTM, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, and Collaborative Innovation Center of Biotherapy, Chengdu, 610041, China
| | - Qianqian Chen
- Department of Urology, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, and Collaborative Innovation Center of Biotherapy, Chengdu, 610041, China
| | - Bo Zhong
- Department of Virology, College of Life Sciences, Department of Immunology, Medical Research Institute, Wuhan University, Wuhan, 430072, China
| | - Lunzhi Dai
- Department of General Practice and Lab of PTM, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, and Collaborative Innovation Center of Biotherapy, Chengdu, 610041, China
| | - Shiqian Qi
- Department of Urology, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, and Collaborative Innovation Center of Biotherapy, Chengdu, 610041, China
| | - Zhiqiang Zhang
- Immunobiology and Transplant Science Center, Houston Methodist Hospital, Houston, TX, USA, 77030.
| | - Huiyuan Zhang
- Department of Rheumatology and Immunology, National Clinical Research Center for Geriatrics, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, and Collaborative Innovation Center of Biotherapy, Chengdu, 610041, China.
| | - Hongbo Hu
- Department of Rheumatology and Immunology, National Clinical Research Center for Geriatrics, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, and Collaborative Innovation Center of Biotherapy, Chengdu, 610041, China.
| |
Collapse
|
18
|
Wang R, Zheng Z, Mao S, Zhang W, Liu J, Li C, Liu S, Yao X. Construction and Validation of a Novel Eight-Gene Risk Signature to Predict the Progression and Prognosis of Bladder Cancer. Front Oncol 2021; 11:632459. [PMID: 34268106 PMCID: PMC8276675 DOI: 10.3389/fonc.2021.632459] [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: 11/23/2020] [Accepted: 06/07/2021] [Indexed: 11/13/2022] Open
Abstract
The progression from non-muscle-invasive bladder cancer (NMIBC) to muscle-invasive bladder cancer (MIBC) increases the risk of death. It is therefore important to find new relevant molecular models that will allow for effective prediction of the progression and prognosis of bladder cancer (BC). Using RNA-Sequence data of 49 BC patients in Shanghai tenth people's hospital (STPH) and weighted gene co-expression network analysis methods, a co-expression network of genes was developed and three key modules associated with malignant progression were selected. Based on the genes in three key modules, an eight-gene risk signature was established using univariate Cox regression and the Least absolute shrinkage and selection operator Cox model in The Cancer Genome Atlas Program (TCGA) and validated in validation sets. Subsequently, a nomogram based on the risk signature was constructed for prognostic prediction. The mRNA and protein expression levels of eight genes in cell lines and tissues were further investigated. The novel eight-gene risk signature was closely related to the malignant clinical features of BC and could predict the prognosis of patients in the training dataset (TCGA) and four validation sets (GSE32894, GSE13507, IMvigor210 trial, and STPH). The nomogram showed good prognostic prediction and calibration. The mRNA and protein expression levels of the eight genes were differentially expressed in cell lines and tissues. In our study, we established a novel eight-gene risk signature that could predict the progression and prognoses of BC patients.
Collapse
Affiliation(s)
- Ruiliang Wang
- Department of Urology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Zongtai Zheng
- Department of Urology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Shiyu Mao
- Department of Urology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Wentao Zhang
- Department of Urology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Ji Liu
- Department of Urology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Cheng Li
- Department of Urology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Shenghua Liu
- Department of Urology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Xudong Yao
- Department of Urology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| |
Collapse
|
19
|
Jin Z, Zhu Z. The role of TRIM proteins in PRR signaling pathways and immune-related diseases. Int Immunopharmacol 2021; 98:107813. [PMID: 34126340 DOI: 10.1016/j.intimp.2021.107813] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2021] [Revised: 05/13/2021] [Accepted: 05/23/2021] [Indexed: 12/25/2022]
Abstract
Pattern recognition receptors (PRRs) are a kind of recognition molecules mainly expressed on innate immune cells. PRRs recognize one or more kinds of pathogen-associated molecular patterns (PAMPs), inducing the production of interleukin (IL), tumor necrosis factor (TNF), interferon (IFN) and other related cytokines to aggravate immune-related diseases. PPR signaling pathways play an important role in both innate and adaptive immune system, and they are easy to be activated or regulated. Tripartite motif (TRIM) proteins are a group of highly conserved proteins in structure. Most of TRIM proteins contain RING domain, which is thought to play a role in ubiquitination. TRIM proteins are involved in viral immunity, inflammatory response, autophagy, and tumor growth. In this review, we focus on the regulation of TRIM proteins on PRR signaling pathways and their roles in immune-related diseases.
Collapse
Affiliation(s)
- Zheng Jin
- Department of Immunology, College of Basic Medical Sciences, Jilin University, Changchun, Jilin Province, China
| | - Zhenhua Zhu
- Department of Orthopaedic Trauma, The Third Affiliated Hospital of Southern, Medical University, Guangzhou, Guangdong Province, China.
| |
Collapse
|
20
|
Huang C, Liu Q, Tang Q, Jing X, Wu T, Zhang J, Zhang G, Zhou J, Zhang Z, Zhao Y, Huang H, Xia Y, Yan J, Xiao J, Li Y, He J. Hepatocyte-specific deletion of Nlrp6 in mice exacerbates the development of non-alcoholic steatohepatitis. Free Radic Biol Med 2021; 169:110-121. [PMID: 33857628 DOI: 10.1016/j.freeradbiomed.2021.04.008] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Revised: 04/06/2021] [Accepted: 04/07/2021] [Indexed: 02/08/2023]
Abstract
OBJECTIVE Previous studies have established that deficiency in Nucleotide-binding and oligomerization domain (NOD)-like receptor family pyrin domain containing 6 (Nlrp6) changes the configuration of the gut microbiota, which leads to hepatic steatosis. Here, we aimed to determine the hepatic function of Nlrp6 in lipid metabolism and inflammation and its role in the development of non-alcoholic steatohepatitis (NASH). METHODS Nlrp6Loxp/Loxp and hepatocyte-specific Nlrp6-knockout mice were fed a high-fat diet (HFD) or methionine-choline deficient (MCD) diet to induce fatty liver or steatohepatitis, respectively. Primary hepatocytes were isolated to further explore the underlying mechanisms in vitro. In addition, we used adenovirus to overexpress Nlrp6 in ob/ob mice to demonstrate its role in NASH. RESULTS Hepatic Nlrp6 expression was downregulated in NASH patients and in obese mice. Hepatocyte-specific Nlrp6 deficiency promoted HFD- or MCD diet-induced lipid accumulation and inflammation, whereas Nlrp6 overexpression in ob/ob mice had beneficial effects. In vitro studies demonstrated that knockdown of Nlrp6 aggravated hepatic steatosis and inflammation in hepatocytes, but its overexpression markedly attenuated these abnormalities. Moreover, both in vitro and in vivo study demonstrated that Nlrp6 inhibited Cd36-mediated lipid uptake. Nlrp6 deficiency-enhanced fatty acid uptake was blocked by a Cd36 inhibitor in hepatocytes. Nlrp6 ablation increased the expression of proinflammatory cytokines, likely as a result of increased NF-κB phosphorylation and activation. Mechanistically, Nlrp6 promoted the degradation of transforming growth factor-β-activated kinase 1 (TAK1)-binding protein 2/3 (TAB2/3) via a lysosomal-dependent pathway, which suppressed NF-κB activation. CONCLUSIONS Nlrp6 may play a key role in the pathological process of NASH by inhibiting Cd36 and NF-κB pathways. It may be a potential therapeutic target for NASH.
Collapse
Affiliation(s)
- Cuiyuan Huang
- Department of Pharmacy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China; Laboratory of Clinical Pharmacy and Adverse Drug Reaction, National Clinical Research Center for Geriatrics,West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Qinhui Liu
- Laboratory of Clinical Pharmacy and Adverse Drug Reaction, National Clinical Research Center for Geriatrics,West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Qin Tang
- Laboratory of Clinical Pharmacy and Adverse Drug Reaction, National Clinical Research Center for Geriatrics,West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Xiandan Jing
- Laboratory of Clinical Pharmacy and Adverse Drug Reaction, National Clinical Research Center for Geriatrics,West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Tong Wu
- Department of Pharmacy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China; Laboratory of Clinical Pharmacy and Adverse Drug Reaction, National Clinical Research Center for Geriatrics,West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Jinhang Zhang
- Department of Pharmacy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China; Laboratory of Clinical Pharmacy and Adverse Drug Reaction, National Clinical Research Center for Geriatrics,West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Guorong Zhang
- Department of Pharmacy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China; Laboratory of Clinical Pharmacy and Adverse Drug Reaction, National Clinical Research Center for Geriatrics,West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Jian Zhou
- Department of Pharmacy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China; Laboratory of Clinical Pharmacy and Adverse Drug Reaction, National Clinical Research Center for Geriatrics,West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Zijing Zhang
- Department of Pharmacy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China; Laboratory of Clinical Pharmacy and Adverse Drug Reaction, National Clinical Research Center for Geriatrics,West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Yingnan Zhao
- Department of Pharmacy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China; Laboratory of Clinical Pharmacy and Adverse Drug Reaction, National Clinical Research Center for Geriatrics,West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Hui Huang
- Department of Pharmacy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China; Laboratory of Clinical Pharmacy and Adverse Drug Reaction, National Clinical Research Center for Geriatrics,West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Yan Xia
- Department of Pharmacy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China; Laboratory of Clinical Pharmacy and Adverse Drug Reaction, National Clinical Research Center for Geriatrics,West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Jiamin Yan
- Department of Pharmacy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China; Laboratory of Clinical Pharmacy and Adverse Drug Reaction, National Clinical Research Center for Geriatrics,West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Jia Xiao
- Clinical Research Institute, First Affiliated Hospital of Jinan University, Guangzhou 510632, China.
| | - Yanping Li
- Laboratory of Clinical Pharmacy and Adverse Drug Reaction, National Clinical Research Center for Geriatrics,West China Hospital, Sichuan University, Chengdu, Sichuan Province, China.
| | - Jinhan He
- Department of Pharmacy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China; Laboratory of Clinical Pharmacy and Adverse Drug Reaction, National Clinical Research Center for Geriatrics,West China Hospital, Sichuan University, Chengdu, Sichuan Province, 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: 18] [Impact Index Per Article: 6.0] [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
|
Xu YR, Lei CQ. TAK1-TABs Complex: A Central Signalosome in Inflammatory Responses. Front Immunol 2021; 11:608976. [PMID: 33469458 PMCID: PMC7813674 DOI: 10.3389/fimmu.2020.608976] [Citation(s) in RCA: 89] [Impact Index Per Article: 29.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Accepted: 11/09/2020] [Indexed: 12/14/2022] Open
Abstract
Transforming growth factor-β (TGF-β)-activated kinase 1 (TAK1) is a member of the MAPK kinase kinase (MAPKKK) family and has been implicated in the regulation of a wide range of physiological and pathological processes. TAK1 functions through assembling with its binding partners TAK1-binding proteins (TAB1, TAB2, and TAB3) and can be activated by a variety of stimuli such as tumor necrosis factor α (TNFα), interleukin-1β (IL-1β), and toll-like receptor ligands, and they play essential roles in the activation of NF-κB and MAPKs. Numerous studies have demonstrated that post-translational modifications play important roles in properly controlling the activity, stability, and assembly of TAK1-TABs complex according to the indicated cellular environment. This review focuses on the recent advances in TAK1-TABs-mediated signaling and the regulations of TAK1-TABs complex by post-translational modifications.
Collapse
Affiliation(s)
- Yan-Ran Xu
- Hubei Key Laboratory of Cell Homeostasis, Frontier Science Center for Immunology and Metabolism, State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan, China
| | - Cao-Qi Lei
- Hubei Key Laboratory of Cell Homeostasis, Frontier Science Center for Immunology and Metabolism, State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan, China
| |
Collapse
|
23
|
Hage A, Rajsbaum R. To TRIM or not to TRIM: the balance of host-virus interactions mediated by the ubiquitin system. J Gen Virol 2020; 100:1641-1662. [PMID: 31661051 DOI: 10.1099/jgv.0.001341] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
The innate immune system responds rapidly to protect against viral infections, but an overactive response can cause harmful damage. To avoid this, the response is tightly regulated by post-translational modifications (PTMs). The ubiquitin system represents a powerful PTM machinery that allows for the reversible linkage of ubiquitin to activate and deactivate a target's function. A precise enzymatic cascade of ubiquitin-activating, conjugating and ligating enzymes facilitates ubiquitination. Viruses have evolved to take advantage of the ubiquitin pathway either by targeting factors to dampen the antiviral response or by hijacking the system to enhance their replication. The tripartite motif (TRIM) family of E3 ubiquitin ligases has garnered attention as a major contributor to innate immunity. Many TRIM family members limit viruses either indirectly as components in innate immune signalling, or directly by targeting viral proteins for degradation. In spite of this, TRIMs and other ubiquitin ligases can be appropriated by viruses and repurposed as valuable tools in viral replication. This duality of function suggests a new frontier of research for TRIMs and raises new challenges for discerning the subtleties of these pro-viral mechanisms. Here, we review current findings regarding the involvement of TRIMs in host-virus interactions. We examine ongoing developments in the field, including novel roles for unanchored ubiquitin in innate immunity, the direct involvement of ubiquitin ligases in promoting viral replication, recent controversies on the role of ubiquitin and TRIM25 in activation of the pattern recognition receptor RIG-I, and we discuss the implications these studies have on future research directions.
Collapse
Affiliation(s)
- Adam Hage
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Ricardo Rajsbaum
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX 77555, USA.,Institute for Human Infections and Immunity, University of Texas Medical Branch, Galveston, TX 77555, USA
| |
Collapse
|
24
|
Xu Z, Zhao F, Chen H, Xu S, Fan F, Shi P, Tu M, Wang Z, Du M. Nutritional properties and osteogenic activity of enzymatic hydrolysates of proteins from the blue mussel (Mytilus edulis). Food Funct 2019; 10:7745-7754. [DOI: 10.1039/c9fo01656b] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Seafood provides a range of health benefits due to its nutritional and bioactive components. The proteins and peptides from Mytilus edulis have good bone growth promoting activities.
Collapse
Affiliation(s)
- Zhe Xu
- School of Food Science and Technology
- National Engineering Research Center of Seafood
- Dalian Polytechnic University
- Dalian, 116034
- China
| | - Fujunzhu Zhao
- Food Science Department
- College of Agriculture Science
- Pennsylvania State University, Commonwealth of Pennsylvania
- PA 16802
- United States
| | - Hui Chen
- School of Food Science and Technology
- National Engineering Research Center of Seafood
- Dalian Polytechnic University
- Dalian, 116034
- China
| | - Shiqi Xu
- School of Food Science and Technology
- National Engineering Research Center of Seafood
- Dalian Polytechnic University
- Dalian, 116034
- China
| | - Fengjiao Fan
- College of Food Science and Engineering
- Nanjing University of Finance and Economics
- Nanjing
- China
| | - Pujie Shi
- School of Food Science and Technology
- National Engineering Research Center of Seafood
- Dalian Polytechnic University
- Dalian, 116034
- China
| | - Maolin Tu
- School of Food Science and Technology
- National Engineering Research Center of Seafood
- Dalian Polytechnic University
- Dalian, 116034
- China
| | - Ziye Wang
- School of Food Science and Technology
- National Engineering Research Center of Seafood
- Dalian Polytechnic University
- Dalian, 116034
- China
| | - Ming Du
- School of Food Science and Technology
- National Engineering Research Center of Seafood
- Dalian Polytechnic University
- Dalian, 116034
- China
| |
Collapse
|