1
|
Lin Y, Feng H, Wang Y, Liu S, Hu P, Wang J, Cao H. The E3 ubiquitin ligase RNF182 regulates the induction of innate immune response against GCRV by mediating the ubiquitination of RIG-I in grass carp (Ctenopharyngodon idella) and rare minnow (Gobiocypris rarus). FISH & SHELLFISH IMMUNOLOGY 2025; 161:110244. [PMID: 40049566 DOI: 10.1016/j.fsi.2025.110244] [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: 10/29/2024] [Revised: 01/05/2025] [Accepted: 02/28/2025] [Indexed: 03/16/2025]
Abstract
Innate immunity is the first line of antiviral or antimicrobial defence for the host. A cytoplasmic viral RNA sensor, which is known as retinoic acid-inducible gene 1 (RIG-I), makes a vital impact on the production of type I interferons (IFN) and eliminating RNA virus. This study indicated that E3 ubiquitin ligase RING finger protein 182 (RNF182) inhibited the antiviral activity of type I IFN in grass carp reovirus (GCRV)-infected cells by directly interplaying with RIG-I. The CiE3RNF182 cDNA encode a polypeptide of 158 amino acids. Cellular distribution analysis results suggested that cytoplasm was the main site of CiE3RNF182 location. Real-time quantitative PCR showed universal expression of CiE3RNF182 in all investigated tissues, with extremely high expression in liver. During virus infection, the CiE3RNF182 associates with the CiRIG-I and then induces the Lys-33-linked ubiquitin to the Lys33 residues of CiRIG-I to trigger its degradation, causing the inhibition of CiRIG-I downstream signalling. Furthermore, we obtained CRISPR/Cas9-mediated generation of E3RNF182-null rare minnows, finding that E3RNF182 deletion facilitates the survival ratio of GCRV-infected rare minnows. Additionally, the E3RNF182-/- rare minnows exhibited significantly lower relative copy number of GCRV compared to the wild-type group. In summary, our findings demonstrate the function of E3 ligase in controlling the anti-GCRV innate immunity through RIG-I in fish.
Collapse
Affiliation(s)
- Yusheng Lin
- Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, 430072, China; University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Haohao Feng
- Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, 430072, China; University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Yuxuan Wang
- Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, 430072, China
| | - Shuai Liu
- Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, 430072, China; University of Chinese Academy of Sciences, Beijing, 100049, China; College of Fisheries and Life Science, Dalian Ocean University, Dalian, 116023, China
| | - Pengcheng Hu
- Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, 430072, China; University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Jing Wang
- Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, 430072, China; University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Hong Cao
- Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, 430072, China; University of Chinese Academy of Sciences, Beijing, 100049, China.
| |
Collapse
|
2
|
Kim D, Nam HJ, Baek SH. Ubiquitination of transcription factors in cancer: unveiling therapeutic potential. Mol Oncol 2025. [PMID: 40227962 DOI: 10.1002/1878-0261.70033] [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: 11/13/2024] [Revised: 03/05/2025] [Accepted: 03/21/2025] [Indexed: 04/16/2025] Open
Abstract
Transcription factors, pivotal in gene expression regulation, are essential in cancer progression. Their function is meticulously regulated by post-translational modifications, including ubiquitination. This process, which marks proteins for degradation, can either enhance or inhibit the function of transcription factors, contingent on the context. In cancers, dysregulated ubiquitination of transcription factors contributes to the hallmark of uncontrolled growth and survival of tumors. For example, tumor suppressors such as p53 might be degraded prematurely due to abnormal ubiquitination, causing genomic instability. On the other hand, oncogenic transcription factors may gain stability via ubiquitination, thus facilitating tumorigenesis. Targeting the ubiquitin-proteasome system (UPS) therefore could be a viable therapeutic approach in cancer. Emerging treatments aim to block the ubiquitination of oncogenic transcription factors or to stabilize tumor suppressors. This review underscores the critical impact of transcription factor-altered ubiquitination on cancer progression. Additionally, it outlines innovative therapeutic approaches that involve inhibitors or drugs directed at specific ubiquitin E3 ligases and deubiquitinases (DUBs) that regulate transcription factor activity.
Collapse
Affiliation(s)
- Dongha Kim
- Department of Anatomy, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Hye Jin Nam
- Therapeutics and Biotechnology Division, Korea Research Institute of Chemical Technology, Daejeon, Korea
- Department of Medicinal Chemistry and Pharmacology, University of Science and Technology, Daejeon, Korea
| | - Sung Hee Baek
- Creative Research Initiatives Center for Epigenetic Code and Diseases, School of Biological Sciences, Seoul National University, Korea
| |
Collapse
|
3
|
Lu J, Zhang J, Jiang H, Hu Z, Zhang Y, He L, Yang J, Xie Y, Wu D, Li H, Zeng K, Tan P, Xiao Q, Song Z, Pan C, Bai X, Yu X. Vangl2 suppresses NF-κB signaling and ameliorates sepsis by targeting p65 for NDP52-mediated autophagic degradation. eLife 2024; 12:RP87935. [PMID: 39269442 PMCID: PMC11398866 DOI: 10.7554/elife.87935] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/15/2024] Open
Abstract
Van Gogh-like 2 (Vangl2), a core planar cell polarity component, plays an important role in polarized cellular and tissue morphology induction, growth development, and cancer. However, its role in regulating inflammatory responses remains elusive. Here, we report that Vangl2 is upregulated in patients with sepsis and identify Vangl2 as a negative regulator of The nuclear factor-kappaB (NF-κB) signaling by regulating the protein stability and activation of the core transcription component p65. Mice with myeloid-specific deletion of Vangl2 (Vangl2ΔM) are hypersusceptible to lipopolysaccharide (LPS)-induced septic shock. Vangl2-deficient myeloid cells exhibit enhanced phosphorylation and expression of p65, therefore, promoting the secretion of proinflammatory cytokines after LPS stimulation. Mechanistically, NF-κB signaling-induced-Vangl2 recruits E3 ubiquitin ligase PDLIM2 to catalyze K63-linked ubiquitination on p65, which serves as a recognition signal for cargo receptor NDP52-mediated selective autophagic degradation. Taken together, these findings demonstrate Vangl2 as a suppressor of NF-κB-mediated inflammation and provide insights into the crosstalk between autophagy and inflammatory diseases.
Collapse
Affiliation(s)
- Jiansen Lu
- Department of Joint Surgery, the Fifth Affiliated Hospital, Southern Medical UniversityGuangzhouChina
- Department of Immunology, School of Basic Medical Sciences, Southern Medical UniversityGuangzhouChina
| | - Jiahuan Zhang
- Department of Clinical Laboratory Medicine, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical UniversityGuangzhouChina
| | - Huaji Jiang
- Department of Immunology, School of Basic Medical Sciences, Southern Medical UniversityGuangzhouChina
- Department of Orthopaedics, Yuebei People's Hospital Affiliated to Medical College of Shantou UniversityShaoguanChina
| | - Zhiqiang Hu
- Department of Immunology, School of Basic Medical Sciences, Southern Medical UniversityGuangzhouChina
| | - Yufen Zhang
- Department of Immunology, School of Basic Medical Sciences, Southern Medical UniversityGuangzhouChina
| | - Lian He
- Department of Pharmacology, School of Medicine, Southern University of Science and TechnologyShenzhenChina
- Institute of Biosciences and Technology, College of Medicine, Texas A&M UniversityHoustonUnited States
| | - Jianwu Yang
- Department of Immunology, School of Basic Medical Sciences, Southern Medical UniversityGuangzhouChina
| | - Yingchao Xie
- Department of Immunology, School of Basic Medical Sciences, Southern Medical UniversityGuangzhouChina
| | - Dan Wu
- Department of Immunology, School of Basic Medical Sciences, Southern Medical UniversityGuangzhouChina
| | - Hongyu Li
- Department of Immunology, School of Basic Medical Sciences, Southern Medical UniversityGuangzhouChina
| | - Ke Zeng
- Department of Immunology, School of Basic Medical Sciences, Southern Medical UniversityGuangzhouChina
| | - Peng Tan
- Department of Pharmacology, School of Medicine, Southern University of Science and TechnologyShenzhenChina
- Klarman Cell Observatory, Broad Institute of MIT and HarvardCambridgeUnited States
| | - Qingyue Xiao
- Department of Cell Biology, School of Basic Medical Sciences, Southern Medical UniversityGuangzhouChina
| | - Zijing Song
- Department of Cell Biology, School of Basic Medical Sciences, Southern Medical UniversityGuangzhouChina
| | - Chenglong Pan
- Department of Joint Surgery, the Fifth Affiliated Hospital, Southern Medical UniversityGuangzhouChina
| | - Xiaochun Bai
- Department of Cell Biology, School of Basic Medical Sciences, Southern Medical UniversityGuangzhouChina
| | - Xiao Yu
- Department of Immunology, School of Basic Medical Sciences, Southern Medical UniversityGuangzhouChina
- Department of Clinical Laboratory Medicine, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical UniversityGuangzhouChina
- Guangdong Provincial Key Lab of Single Cell Technology and Application, Southern Medical UniversityGuangzhouChina
| |
Collapse
|
4
|
Liao HX, Mao X, Wang L, Wang N, Ocansey DKW, Wang B, Mao F. The role of mesenchymal stem cells in attenuating inflammatory bowel disease through ubiquitination. Front Immunol 2024; 15:1423069. [PMID: 39185411 PMCID: PMC11341407 DOI: 10.3389/fimmu.2024.1423069] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Accepted: 07/22/2024] [Indexed: 08/27/2024] Open
Abstract
Inflammatory bowel disease (IBD), a condition of the digestive tract and one of the autoimmune diseases, is becoming a disease of significant global public health concern and substantial clinical burden. Various signaling pathways have been documented to modulate IBD, but the exact activation and regulatory mechanisms have not been fully clarified; thus, a need for constant exploration of the molecules and pathways that play key roles in the development of IBD. In recent years, several protein post-translational modification pathways, such as ubiquitination, phosphorylation, methylation, acetylation, and glycolysis, have been implicated in IBD. An aberrant ubiquitination in IBD is often associated with dysregulated immune responses and inflammation. Mesenchymal stem cells (MSCs) play a crucial role in regulating ubiquitination modifications through the ubiquitin-proteasome system, a cellular machinery responsible for protein degradation. Specifically, MSCs have been shown to influence the ubiquitination of key signaling molecules involved in inflammatory pathways. This paper reviews the recent research progress in MSC-regulated ubiquitination in IBD, highlighting their therapeutic potential in treating IBD and offering a promising avenue for developing targeted interventions to modulate the immune system and alleviate inflammatory conditions.
Collapse
Affiliation(s)
- Hong Xi Liao
- Key Laboratory of Medical Science and Laboratory Medicine of Jiangsu Province, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu, China
- Department of Laboratory Medicine, Lianyungang Clinical College, Jiangsu University, Lianyungang, Jiangsu, China
| | - Xiaojun Mao
- The People’s Hospital of Danyang, Affiliated Danyang Hospital of Nantong University, Zhenjiang, Jiangsu, China
| | - Lan Wang
- Department of Laboratory Medicine, Danyang Blood Station, Zhenjiang, Jiangsu, China
| | - Naijian Wang
- Department of Laboratory Medicine, Lianyungang Clinical College, Jiangsu University, Lianyungang, Jiangsu, China
| | - Dickson Kofi Wiredu Ocansey
- Key Laboratory of Medical Science and Laboratory Medicine of Jiangsu Province, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu, China
| | - Bo Wang
- Key Laboratory of Medical Science and Laboratory Medicine of Jiangsu Province, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu, China
| | - Fei Mao
- Key Laboratory of Medical Science and Laboratory Medicine of Jiangsu Province, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu, China
- Department of Laboratory Medicine, Lianyungang Clinical College, Jiangsu University, Lianyungang, Jiangsu, China
| |
Collapse
|
5
|
Zhang J, Yu Y, Zou X, Du Y, Liang Q, Gong M, He Y, Luo J, Wu D, Jiang X, Sinclair M, Tajkhorshid E, Chen HZ, Hou Z, Zheng Y, Chen LF, Yang XD. WSB1/2 target chromatin-bound lysine-methylated RelA for proteasomal degradation and NF-κB termination. Nucleic Acids Res 2024; 52:4969-4984. [PMID: 38452206 PMCID: PMC11109945 DOI: 10.1093/nar/gkae161] [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: 06/06/2023] [Revised: 02/08/2024] [Accepted: 02/22/2024] [Indexed: 03/09/2024] Open
Abstract
Proteasome-mediated degradation of chromatin-bound NF-κB is critical in terminating the transcription of pro-inflammatory genes and can be triggered by Set9-mediated lysine methylation of the RelA subunit. However, the E3 ligase targeting methylated RelA remains unknown. Here, we find that two structurally similar substrate-recognizing components of Cullin-RING E3 ligases, WSB1 and WSB2, can recognize chromatin-bound methylated RelA for polyubiquitination and proteasomal degradation. We showed that WSB1/2 negatively regulated a subset of NF-κB target genes via associating with chromatin where they targeted methylated RelA for ubiquitination, facilitating the termination of NF-κB-dependent transcription. WSB1/2 specifically interacted with methylated lysines (K) 314 and 315 of RelA via their N-terminal WD-40 repeat (WDR) domains, thereby promoting ubiquitination of RelA. Computational modeling further revealed that a conserved aspartic acid (D) at position 158 within the WDR domain of WSB2 coordinates K314/K315 of RelA, with a higher affinity when either of the lysines is methylated. Mutation of D158 abolished WSB2's ability to bind to and promote ubiquitination of methylated RelA. Together, our study identifies a novel function and the underlying mechanism for WSB1/2 in degrading chromatin-bound methylated RelA and preventing sustained NF-κB activation, providing potential new targets for therapeutic intervention of NF-κB-mediated inflammatory diseases.
Collapse
Affiliation(s)
- Jie Zhang
- Hongqiao Institute of Medicine, Tongren Hospital/Faculty of Basic Medicine, Shanghai Jiaotong University School of Medicine, Shanghai 200025, China
| | - Yuanyuan Yu
- The Research Center for Traditional Chinese Medicine, Shanghai Institute of Infectious Diseases and Biosecurity, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
- Center for Traditional Chinese Medicine and Immunology Research, School of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Xiuqun Zou
- Hongqiao Institute of Medicine, Tongren Hospital/Faculty of Basic Medicine, Shanghai Jiaotong University School of Medicine, Shanghai 200025, China
| | - Yaning Du
- Hongqiao Institute of Medicine, Tongren Hospital/Faculty of Basic Medicine, Shanghai Jiaotong University School of Medicine, Shanghai 200025, China
| | - Qiankun Liang
- The Research Center for Traditional Chinese Medicine, Shanghai Institute of Infectious Diseases and Biosecurity, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
- Center for Traditional Chinese Medicine and Immunology Research, School of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Mengyao Gong
- The Research Center for Traditional Chinese Medicine, Shanghai Institute of Infectious Diseases and Biosecurity, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
- Center for Traditional Chinese Medicine and Immunology Research, School of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Yurong He
- The Research Center for Traditional Chinese Medicine, Shanghai Institute of Infectious Diseases and Biosecurity, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
- Center for Traditional Chinese Medicine and Immunology Research, School of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Junqi Luo
- The Research Center for Traditional Chinese Medicine, Shanghai Institute of Infectious Diseases and Biosecurity, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
- Center for Traditional Chinese Medicine and Immunology Research, School of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Dandan Wu
- Shanghai Institute of Immunology, and Department of Immunology and Microbiology, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Xiaoli Jiang
- Shanghai Institute of Immunology, and Department of Immunology and Microbiology, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Matt Sinclair
- Theoretical and Computational Biophysics Group, NIH Center for Macromolecular Modeling and Visualization, Beckman Institute for Advanced Science and Technology, and Center for Biophysics and Quantitative Biology, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
- Department of Biochemistry, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - Emad Tajkhorshid
- Theoretical and Computational Biophysics Group, NIH Center for Macromolecular Modeling and Visualization, Beckman Institute for Advanced Science and Technology, and Center for Biophysics and Quantitative Biology, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
- Department of Biochemistry, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - Hong-Zhuan Chen
- The Research Center for Traditional Chinese Medicine, Shanghai Institute of Infectious Diseases and Biosecurity, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
- Shuguang lab of Future Health, Shanghai Frontiers Science Center of TCM Chemical Biology, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Zhaoyuan Hou
- Hongqiao Institute of Medicine, Tongren Hospital/Faculty of Basic Medicine, Shanghai Jiaotong University School of Medicine, Shanghai 200025, China
- Linyi University-Shanghai Jiaotong University Joint Institute of Translational Medicine, Linyi University, Shandong 276000, China
| | - Yuejuan Zheng
- The Research Center for Traditional Chinese Medicine, Shanghai Institute of Infectious Diseases and Biosecurity, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
- Center for Traditional Chinese Medicine and Immunology Research, School of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Lin-Feng Chen
- Department of Biochemistry, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - Xiao-Dong Yang
- The Research Center for Traditional Chinese Medicine, Shanghai Institute of Infectious Diseases and Biosecurity, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
- Center for Traditional Chinese Medicine and Immunology Research, School of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| |
Collapse
|
6
|
Bo Z, Li X, Wang S, Zhang C, Guo M, Cao Y, Zhang X, Wu Y. Suppression of NF-κB signaling by Pseudorabies virus DNA polymerase processivity factor UL42 via recruiting SOCS1 to promote the ubiquitination degradation of p65. Vet Microbiol 2023; 287:109896. [PMID: 37931575 DOI: 10.1016/j.vetmic.2023.109896] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Revised: 10/18/2023] [Accepted: 10/21/2023] [Indexed: 11/08/2023]
Abstract
The NF-κB pathway is a critical signaling involved in the regulation of the inflammatory and innate immune responses. Previous studies have shown that Pseudorabies Virus (PRV), a porcine alpha herpesvirus, could lead to the phosphorylation and nucleus translocation of p65 while inhibiting the expression of NF-κB-dependent inflammatory cytokines, which indicated that there may be unknown mechanisms downstream of p65 that downregulate the activation of NF-κB signaling. Here, we found that PRV DNA polymerase factor UL42 inhibited TNFα-, LPS-, IKKα-, IKKβ-, and p65-mediated transactivation of NF-κB signaling, which demonstrated UL42 worked either at or downstream of p65. In addition, it was found that the DNA-binding activity of UL42 was required for inhibition of NF-κB signaling. Importantly, it was revealed that UL42 could induce the ubiquitination degradation of p65 by upregulating the suppressor of cytokine signaling 1 (SOCS1). Additionally, it was found that UL42 could promote the K6/K29-linked ubiquitination of p65. Finally, knockdown of SOCS1 attenuated the replication of PRV and led to a significant increase of the inflammatory cytokines. Taken together, our findings uncovered a novel mechanism that PRV-UL42 could upregulated SOCS1 to promote the ubiquitination degradation of p65 to prevent excessive inflammatory response during PRV infection.
Collapse
Affiliation(s)
- Zongyi Bo
- Joint International Research Laboratory of Agriculture and Agri-Product Safety, The Ministry of Education of China, Yangzhou University, Yangzhou 225009, China; Jiangsu Co-Innovation Center for the Prevention and Control of Animal Infectious Disease and Zoonoses, College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, China
| | - Xiaojuan Li
- Joint International Research Laboratory of Agriculture and Agri-Product Safety, The Ministry of Education of China, Yangzhou University, Yangzhou 225009, China; Jiangsu Co-Innovation Center for the Prevention and Control of Animal Infectious Disease and Zoonoses, College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, China
| | - Shixu Wang
- Joint International Research Laboratory of Agriculture and Agri-Product Safety, The Ministry of Education of China, Yangzhou University, Yangzhou 225009, China; Jiangsu Co-Innovation Center for the Prevention and Control of Animal Infectious Disease and Zoonoses, College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, China
| | - Chengcheng Zhang
- Jiangsu Co-Innovation Center for the Prevention and Control of Animal Infectious Disease and Zoonoses, College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, China
| | - Mengjiao Guo
- Jiangsu Co-Innovation Center for the Prevention and Control of Animal Infectious Disease and Zoonoses, College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, China
| | - Yongzhong Cao
- Joint International Research Laboratory of Agriculture and Agri-Product Safety, The Ministry of Education of China, Yangzhou University, Yangzhou 225009, China; Jiangsu Co-Innovation Center for the Prevention and Control of Animal Infectious Disease and Zoonoses, College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, China
| | - Xiaorong Zhang
- Jiangsu Co-Innovation Center for the Prevention and Control of Animal Infectious Disease and Zoonoses, College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, China
| | - Yantao Wu
- Joint International Research Laboratory of Agriculture and Agri-Product Safety, The Ministry of Education of China, Yangzhou University, Yangzhou 225009, China; Jiangsu Co-Innovation Center for the Prevention and Control of Animal Infectious Disease and Zoonoses, College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, China.
| |
Collapse
|
7
|
Wu Z, Chen H, Ke S, Mo L, Qiu M, Zhu G, Zhu W, Liu L. Identifying potential biomarkers of idiopathic pulmonary fibrosis through machine learning analysis. Sci Rep 2023; 13:16559. [PMID: 37783761 PMCID: PMC10545744 DOI: 10.1038/s41598-023-43834-z] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Accepted: 09/28/2023] [Indexed: 10/04/2023] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is the most common and serious type of idiopathic interstitial pneumonia, characterized by chronic, progressive, and low survival rates, while unknown disease etiology. Until recently, patients with idiopathic pulmonary fibrosis have a poor prognosis, high mortality, and limited treatment options, due to the lack of effective early diagnostic and prognostic tools. Therefore, we aimed to identify biomarkers for idiopathic pulmonary fibrosis based on multiple machine-learning approaches and to evaluate the role of immune infiltration in the disease. The gene expression profile and its corresponding clinical data of idiopathic pulmonary fibrosis patients were downloaded from Gene Expression Omnibus (GEO) database. Next, the differentially expressed genes (DEGs) with the threshold of FDR < 0.05 and |log2 foldchange (FC)| > 0.585 were analyzed via R package "DESeq2" and GO enrichment and KEGG pathways were run in R software. Then, least absolute shrinkage and selection operator (LASSO) logistic regression, support vector machine-recursive feature elimination (SVM-RFE) and random forest (RF) algorithms were combined to screen the key potential biomarkers of idiopathic pulmonary fibrosis. The diagnostic performance of these biomarkers was evaluated through receiver operating characteristic (ROC) curves. Moreover, the CIBERSORT algorithm was employed to assess the infiltration of immune cells and the relationship between the infiltrating immune cells and the biomarkers. Finally, we sought to understand the potential pathogenic role of the biomarker (SLAIN1) in idiopathic pulmonary fibrosis using a mouse model and cellular model. A total of 3658 differentially expressed genes of idiopathic pulmonary fibrosis were identified, including 2359 upregulated genes and 1299 downregulated genes. FHL2, HPCAL1, RNF182, and SLAIN1 were identified as biomarkers of idiopathic pulmonary fibrosis using LASSO logistic regression, RF, and SVM-RFE algorithms. The ROC curves confirmed the predictive accuracy of these biomarkers both in the training set and test set. Immune cell infiltration analysis suggested that patients with idiopathic pulmonary fibrosis had a higher level of B cells memory, Plasma cells, T cells CD8, T cells follicular helper, T cells regulatory (Tregs), Macrophages M0, and Mast cells resting compared with the control group. Correlation analysis demonstrated that FHL2 was significantly associated with the infiltrating immune cells. qPCR and western blotting analysis suggested that SLAIN1 might be a signature for the diagnosis of idiopathic pulmonary fibrosis. In this study, we identified four potential biomarkers (FHL2, HPCAL1, RNF182, and SLAIN1) and evaluated the potential pathogenic role of SLAIN1 in idiopathic pulmonary fibrosis. These findings may have great significance in guiding the understanding of disease mechanisms and potential therapeutic targets in idiopathic pulmonary fibrosis.
Collapse
Affiliation(s)
- Zenan Wu
- The Clinical Medical School, Jiangxi University of Traditional Chinese Medicine, Nanchang, Jiangxi, China
| | - Huan Chen
- The Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, China
| | - Shiwen Ke
- The Affiliated Hospital of Jiangxi University of Traditional Chinese Medicine, Nanchang, China
| | - Lisha Mo
- The Affiliated Hospital of Jiangxi University of Traditional Chinese Medicine, Nanchang, China
| | - Mingliang Qiu
- The Affiliated Hospital of Jiangxi University of Traditional Chinese Medicine, Nanchang, China
| | - Guoshuang Zhu
- The Clinical Medical School, Jiangxi University of Traditional Chinese Medicine, Nanchang, Jiangxi, China
| | - Wei Zhu
- The Second Clinical Medical School, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Liangji Liu
- The Affiliated Hospital of Jiangxi University of Traditional Chinese Medicine, Nanchang, China.
| |
Collapse
|
8
|
Zeng X, Tang X, Chen X, Wen H. RNF182 induces p65 ubiquitination to affect PDL1 transcription and suppress immune evasion in lung adenocarcinoma. Immun Inflamm Dis 2023; 11:e864. [PMID: 37249301 PMCID: PMC10201958 DOI: 10.1002/iid3.864] [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: 12/28/2022] [Revised: 04/06/2023] [Accepted: 04/21/2023] [Indexed: 05/31/2023] Open
Abstract
BACKGROUND The RING finger (RNF) proteins are a large group of ubiquitin ligases whose aberrant expression is often associated with disease progression. This study examines the function of RNF protein 182 (RNF182) in lung adenocarcinoma (LUAD) cells and its impact on p65 and programmed death ligand 1 (PDL1) regulation. METHODS Expression of RNF182, p65, and PDL1 in LUAD tissues and cells was measured using immunohistochemistry, reverse transcription quantitative polymerase chain reaction (RT-qPCR), and/or western blot (WB) assays. LUAD cells were induced to overexpress RNF182 and p65, followed by cell counting kit-8, colony formation, Transwell, and flow cytometry assays to evaluate the cells' malignant phenotype. Coimmunoprecipitation and WB assays were used to verify RNF182's effect on p65 ubiquitination. Chromatin immunoprecipitation-qPCR and luciferase assays were used to analyze p65's transcriptional regulation of PDL1. Coculture of LUAD with CD8+ cytotoxic T cells was performed to detect lactate dehydrogenase release and interferon-γ and interleukin-2 concentrations. LUAD cells were implanted in mice to analyze tumorigenicity. RESULTS RNF182 was poorly expressed, while p65 and PDL1 were highly expressed in LUAD tissues and cells. RNF182 overexpression suppressed the malignant properties of LUAD cells, and it promoted p65 ubiquitination and protein degradation. p65 activated PDL1 transcription. Overexpression of RNF182 suppressed the PDL1 expression, increased the cytotoxicity in LUAD cells cocultured with CD8+ T cells, and suppressed the tumorigenesis of cancer cells in vivo. However, these tumor-suppressive effects of RNF182 on LUAD cells were blocked by p65 restoration. CONCLUSION This research demonstrates that RNF182 induces p65 ubiquitination to suppress PDL1 transcription and immunosuppression in LUAD.
Collapse
Affiliation(s)
- Xingdu Zeng
- Department of Respiratory MedicineThe First Affiliated Hospital of Gannan Medical UniversityGanzhouJiangxiPeople's Republic of China
| | - Xiaoyuan Tang
- Department of Respiratory MedicineThe First Affiliated Hospital of Gannan Medical UniversityGanzhouJiangxiPeople's Republic of China
| | - Xingxiang Chen
- Department of Respiratory MedicineThe First Affiliated Hospital of Gannan Medical UniversityGanzhouJiangxiPeople's Republic of China
| | - Huilan Wen
- Department of Respiratory MedicineThe First Affiliated Hospital of Gannan Medical UniversityGanzhouJiangxiPeople's Republic of China
| |
Collapse
|
9
|
Shetty AC, Sivinski J, Cornell J, Sadzewicz L, Mahurkar A, Wang XQ, Colloca L, Lin W, Kane MA, Seneviratne C. Peripheral blood transcriptomic profiling indicates molecular mechanisms commonly regulated by binge-drinking and placebo-effects. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2023:2023.03.21.23287501. [PMID: 36993621 PMCID: PMC10055573 DOI: 10.1101/2023.03.21.23287501] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/30/2023]
Abstract
Molecular changes associated with alcohol consumption arise from complex interactions between pharmacological effects of alcohol, psychological/placebo context surrounding drinking, and other environmental and biological factors. The goal of this study was to tease apart molecular mechanisms regulated by pharmacological effects of alcohol - particularly at binge-drinking, from underlying placebo effects. Transcriptome-wide RNA-seq analyses were performed on peripheral blood samples collected from healthy heavy social drinkers (N=16) enrolled in a 12-day randomized, double-blind, cross-over human laboratory trial testing three alcohol doses: Placebo, moderate (0.05g/kg (men), 0.04g/kg (women)), and binge (1g/kg (men), 0.9g/kg (women)), administered in three 4-day experiments, separated by minimum of 7-day washout periods. Effects of beverage doses on the normalized gene expression counts were analyzed within each experiment compared to its own baseline using paired-t-tests. Differential expression of genes (DEGs) across experimental sequences in which each beverage dose was administered, as well as responsiveness to regular alcohol compared to placebo (i.e., pharmacological effects), were analyzed using generalized linear mixed-effects models. The 10% False discovery rate-adjusted DEGs varied across experimental sequences in response to all three beverage doses. We identified and validated 22 protein coding DEGs potentially responsive to pharmacological effects of binge and medium doses, of which 11 were selectively responsive to binge dose. Binge-dose significantly impacted the Cytokine-cytokine receptor interaction pathway (KEGG: hsa04060) across all experimental-sequences that it was administered in, and during dose-extending placebo. Medium dose and placebo impacted pathways hsa05322, hsa04613, and hsa05034, in the first two and last experimental sequences, respectively. In summary, our findings add novel, and confirm previously reported data supporting dose-dependent effects of alcohol on molecular mechanisms and suggest that the placebo effects may induce molecular responses within the same pathways regulated by alcohol. Innovative study designs are required to validate molecular correlates of placebo effects underlying drinking.
Collapse
|
10
|
Liu Y, Ouyang L, Mao C, Chen Y, Liu N, Chen L, Shi Y, Xiao D, Liu S, Tao Y. Inhibition of RNF182 mediated by Bap promotes non-small cell lung cancer progression. Front Oncol 2023; 12:1009508. [PMID: 36686776 PMCID: PMC9853554 DOI: 10.3389/fonc.2022.1009508] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Accepted: 11/25/2022] [Indexed: 01/08/2023] Open
Abstract
Introduction Ubiquitylation that mediated by ubiquitin ligases plays multiple roles not only in proteasome-mediated protein degradation but also in various cellular process including DNA repair, signal transduction and endocytosis. RING finger (RNF) proteins form the majority of these ubiquitin ligases. Recent studies have demonstrated the important roles of RNF finger proteins in tumorigenesis and tumor progression. Benzo[a]pyrene (BaP) is one of the most common environmental carcinogens causing lung cancer. The molecular mechanism of Bap carcinogenesis remains elusive. Considering the critical roles of RNF proteins in tumorigenesis and tumor progression, we speculate on whether Bap regulates RNF proteins resulting in carcinogenesis. Methods We used GEO analysis to identify the potential RING finger protein family member that contributes to Bap-induced NSCLC. We next used RT-qPCR, Western blot and ChIP assay to investigate the potential mechanism of Bap inhibits RNF182. BGS analyses were used to analyze the methylation level of RNF182. Results Here we reported that the carcinogen Bap suppresses the expression of ring finger protein 182 (RNF182) in non-small cell lung cancer (NSCLC) cells, which is mediated by abnormal hypermethylation in an AhR independent way and transcriptional regulation in an AhR dependent way. Furthermore, RNF182 exhibits low expression and hypermethylation in tumor tissues. RNF182 also significantly suppresses cell proliferation and induces cell cycle arrest in NSCLC cell lines. Conclusion These results demonstrated that Bap inhibits RNF182 expression to promote lung cancer tumorigenesis through activating AhR and promoting abnormal methylation.
Collapse
Affiliation(s)
- Yating Liu
- Department of Pathology, Xiangya Hospital, Central South University, Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Hunan, Changsha, China,National Health Commission (NHC) Key Laboratory of Carcinogenesis (Central South University), Cancer Research Institute and School of Basic Medicine, Central South University, Changsha, Hunan, China,Postdoctoral Research Station of Clinical Medicine & Department of Hematology and Critical Care Medicine, The 3rd Xiangya Hospital, Central South University, Changsha, China
| | - Lianlian Ouyang
- Department of Pathology, Xiangya Hospital, Central South University, Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Hunan, Changsha, China,National Health Commission (NHC) Key Laboratory of Carcinogenesis (Central South University), Cancer Research Institute and School of Basic Medicine, Central South University, Changsha, Hunan, China
| | - Chao Mao
- Department of Pathology, Xiangya Hospital, Central South University, Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Hunan, Changsha, China,National Health Commission (NHC) Key Laboratory of Carcinogenesis (Central South University), Cancer Research Institute and School of Basic Medicine, Central South University, Changsha, Hunan, China
| | - Yuanbing Chen
- Department of Pathology, Xiangya Hospital, Central South University, Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Hunan, Changsha, China,National Health Commission (NHC) Key Laboratory of Carcinogenesis (Central South University), Cancer Research Institute and School of Basic Medicine, Central South University, Changsha, Hunan, China
| | - Na Liu
- Department of Pathology, Xiangya Hospital, Central South University, Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Hunan, Changsha, China,National Health Commission (NHC) Key Laboratory of Carcinogenesis (Central South University), Cancer Research Institute and School of Basic Medicine, Central South University, Changsha, Hunan, China
| | - Ling Chen
- Department of Pathology, Xiangya Hospital, Central South University, Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Hunan, Changsha, China,National Health Commission (NHC) Key Laboratory of Carcinogenesis (Central South University), Cancer Research Institute and School of Basic Medicine, Central South University, Changsha, Hunan, China
| | - Ying Shi
- Department of Pathology, Xiangya Hospital, Central South University, Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Hunan, Changsha, China,National Health Commission (NHC) Key Laboratory of Carcinogenesis (Central South University), Cancer Research Institute and School of Basic Medicine, Central South University, Changsha, Hunan, China
| | - Desheng Xiao
- Department of Pathology, Xiangya Hospital, Central South University, Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Hunan, Changsha, China
| | - Shuang Liu
- Department of Oncology, Institute of Medical Sciences, National Clinical Research, Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China,*Correspondence: Shuang Liu, ; Yongguang Tao,
| | - Yongguang Tao
- Department of Pathology, Xiangya Hospital, Central South University, Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Hunan, Changsha, China,National Health Commission (NHC) Key Laboratory of Carcinogenesis (Central South University), Cancer Research Institute and School of Basic Medicine, Central South University, Changsha, Hunan, China,Hunan Key Laboratory of Early Diagnosis and Precision Therapy in Lung Cancer, Second Xiangya Hospital, Central South University, Changsha, China,*Correspondence: Shuang Liu, ; Yongguang Tao,
| |
Collapse
|
11
|
Huang W, Ji N, Zhao X, Guo J, Feng J, Chen K, Wu Y, Wang J, Zou J. RNA-seq analysis of a zebrafish caudal fin cell line in response to infection with spring viraemia of carp virus. AQUACULTURE AND FISHERIES 2022. [DOI: 10.1016/j.aaf.2022.11.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/04/2022]
|
12
|
The RING finger protein family in health and disease. Signal Transduct Target Ther 2022; 7:300. [PMID: 36042206 PMCID: PMC9424811 DOI: 10.1038/s41392-022-01152-2] [Citation(s) in RCA: 77] [Impact Index Per Article: 25.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Revised: 07/31/2022] [Accepted: 08/09/2022] [Indexed: 02/05/2023] Open
Abstract
Ubiquitination is a highly conserved and fundamental posttranslational modification (PTM) in all eukaryotes regulating thousands of proteins. The RING (really interesting new gene) finger (RNF) protein, containing the RING domain, exerts E3 ubiquitin ligase that mediates the covalent attachment of ubiquitin (Ub) to target proteins. Multiple reviews have summarized the critical roles of the tripartite-motif (TRIM) protein family, a subgroup of RNF proteins, in various diseases, including cancer, inflammatory, infectious, and neuropsychiatric disorders. Except for TRIMs, since numerous studies over the past decades have delineated that other RNF proteins also exert widespread involvement in several diseases, their importance should not be underestimated. This review summarizes the potential contribution of dysregulated RNF proteins, except for TRIMs, to the pathogenesis of some diseases, including cancer, autoimmune diseases, and neurodegenerative disorder. Since viral infection is broadly involved in the induction and development of those diseases, this manuscript also highlights the regulatory roles of RNF proteins, excluding TRIMs, in the antiviral immune responses. In addition, we further discuss the potential intervention strategies targeting other RNF proteins for the prevention and therapeutics of those human diseases.
Collapse
|
13
|
Liu Y, Ouyang L, Mao C, Chen Y, Li T, Liu N, Wang Z, Lai W, Zhou Y, Cao Y, Liu S, Liang Y, Wang M, Liu S, Chen L, Shi Y, Xiao D, Tao Y. PCDHB14 promotes ferroptosis and is a novel tumor suppressor in hepatocellular carcinoma. Oncogene 2022; 41:3570-3583. [PMID: 35688944 DOI: 10.1038/s41388-022-02370-2] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Revised: 05/19/2022] [Accepted: 05/30/2022] [Indexed: 12/20/2022]
Abstract
Liver cancer, a result of multifactorial interplay between heredity and the environment, is one of the leading causes of cancer-related death worldwide. Hepatocellular carcinoma (HCC) is the most common histologic type of primary liver cancer. Here, we reported that deficiency in PCDHB14, a member of the cadherin superfamily, participates in the progression of HCC. We found that PCDHB14 is inactivated by aberrant methylation of its promoter in HCC patients and that PCDHB14 functions as a tumor suppressor to promote cell cycle arrest, inhibit cell proliferation, and induce ferroptosis. Furthermore, PCDHB14 ablation dramatically enhanced diethylenenitrite-induced HCC development. Mechanistically, PCDHB14 is induced by p53, and increased PCDHB14 downregulates the expression of SLC7A11, which is critical for ferroptosis. This effect is mediated by accelerated p65 protein degradation resulting from PCDHB14 promoting E3 ubiquitin ligase RNF182-mediated ubiquitination of p65 to block p65 binding to the promoter of SLC7A11. This study reports the new discovery that PCDHB14 serves as a potential prognostic marker for HCC.
Collapse
Affiliation(s)
- Yating Liu
- Postdoctoral Research Station of Clinical Medicine & Department of Hematology and Critical Care Medicine, the 3rd Xiangya Hospital, Central South University, Changsha, 410000, P. R. China.,Department of Pathology, Xiangya Hospital, Central South University, Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Changsha, Hunan, 410078, P. R. China.,NHC Key Laboratory of Carcinogenesis (Central South University), Cancer Research Institute and School of Basic Medicine, Central South University, Changsha, Hunan, 410078, P. R. China
| | - Lianlian Ouyang
- Department of Pathology, Xiangya Hospital, Central South University, Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Changsha, Hunan, 410078, P. R. China.,NHC Key Laboratory of Carcinogenesis (Central South University), Cancer Research Institute and School of Basic Medicine, Central South University, Changsha, Hunan, 410078, P. R. China
| | - Chao Mao
- Department of Pathology, Xiangya Hospital, Central South University, Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Changsha, Hunan, 410078, P. R. China.,NHC Key Laboratory of Carcinogenesis (Central South University), Cancer Research Institute and School of Basic Medicine, Central South University, Changsha, Hunan, 410078, P. R. China
| | - Yuanbing Chen
- Department of Pathology, Xiangya Hospital, Central South University, Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Changsha, Hunan, 410078, P. R. China.,NHC Key Laboratory of Carcinogenesis (Central South University), Cancer Research Institute and School of Basic Medicine, Central South University, Changsha, Hunan, 410078, P. R. China
| | - Tiansheng Li
- Department of Pathology, Xiangya Hospital, Central South University, Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Changsha, Hunan, 410078, P. R. China.,NHC Key Laboratory of Carcinogenesis (Central South University), Cancer Research Institute and School of Basic Medicine, Central South University, Changsha, Hunan, 410078, P. R. China
| | - Na Liu
- Department of Pathology, Xiangya Hospital, Central South University, Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Changsha, Hunan, 410078, P. R. China.,NHC Key Laboratory of Carcinogenesis (Central South University), Cancer Research Institute and School of Basic Medicine, Central South University, Changsha, Hunan, 410078, P. R. China
| | - Zuli Wang
- Department of Pathology, Xiangya Hospital, Central South University, Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Changsha, Hunan, 410078, P. R. China.,NHC Key Laboratory of Carcinogenesis (Central South University), Cancer Research Institute and School of Basic Medicine, Central South University, Changsha, Hunan, 410078, P. R. China
| | - Weiwei Lai
- Department of Pathology, Xiangya Hospital, Central South University, Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Changsha, Hunan, 410078, P. R. China.,NHC Key Laboratory of Carcinogenesis (Central South University), Cancer Research Institute and School of Basic Medicine, Central South University, Changsha, Hunan, 410078, P. R. China
| | - Yanling Zhou
- Department of Pathology, Xiangya Hospital, Central South University, Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Changsha, Hunan, 410078, P. R. China.,NHC Key Laboratory of Carcinogenesis (Central South University), Cancer Research Institute and School of Basic Medicine, Central South University, Changsha, Hunan, 410078, P. R. China
| | - Ya Cao
- Department of Pathology, Xiangya Hospital, Central South University, Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Changsha, Hunan, 410078, P. R. China
| | - Shuang Liu
- Department of Oncology, Institute of Medical Sciences, National Clinical Research, Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, P. R. China
| | - Yinming Liang
- School of Laboratory Medicine, Xinxiang Medical University, Xinxiang, 453003, P. R. China
| | - Min Wang
- Department of Pathology, Xiangya Hospital, Central South University, Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Changsha, Hunan, 410078, P. R. China.,NHC Key Laboratory of Carcinogenesis (Central South University), Cancer Research Institute and School of Basic Medicine, Central South University, Changsha, Hunan, 410078, P. R. China
| | - Shouping Liu
- Department of Pathology, Xiangya Hospital, Central South University, Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Changsha, Hunan, 410078, P. R. China.,NHC Key Laboratory of Carcinogenesis (Central South University), Cancer Research Institute and School of Basic Medicine, Central South University, Changsha, Hunan, 410078, P. R. China
| | - Ling Chen
- Department of Pathology, Xiangya Hospital, Central South University, Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Changsha, Hunan, 410078, P. R. China.,NHC Key Laboratory of Carcinogenesis (Central South University), Cancer Research Institute and School of Basic Medicine, Central South University, Changsha, Hunan, 410078, P. R. China
| | - Ying Shi
- Department of Pathology, Xiangya Hospital, Central South University, Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Changsha, Hunan, 410078, P. R. China. .,NHC Key Laboratory of Carcinogenesis (Central South University), Cancer Research Institute and School of Basic Medicine, Central South University, Changsha, Hunan, 410078, P. R. China.
| | - Desheng Xiao
- Department of Pathology, Xiangya Hospital, Central South University, Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Changsha, Hunan, 410078, P. R. China. .,NHC Key Laboratory of Carcinogenesis (Central South University), Cancer Research Institute and School of Basic Medicine, Central South University, Changsha, Hunan, 410078, P. R. China.
| | - Yongguang Tao
- Department of Pathology, Xiangya Hospital, Central South University, Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Changsha, Hunan, 410078, P. R. China. .,NHC Key Laboratory of Carcinogenesis (Central South University), Cancer Research Institute and School of Basic Medicine, Central South University, Changsha, Hunan, 410078, P. R. China. .,Hunan Key Laboratory of Early Diagnosis and Precision Therapy in Lung Cancer, Second Xiangya Hospital, Central South University, Changsha, 410011, P. R. China.
| |
Collapse
|
14
|
Ye G, Luo H, Zhang T, Lan T, Ling B, Qi Z. Knockdown of RNF183 suppressed proliferation of lung adenocarcinoma cells via inactivating the STAT3 signaling pathway. Cell Cycle 2022; 21:948-960. [PMID: 35104174 PMCID: PMC9037501 DOI: 10.1080/15384101.2022.2035617] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2021] [Accepted: 01/21/2022] [Indexed: 02/03/2023] Open
Abstract
Proteins of the RNF183 (RING finger 183) family proteins have been reported to be of great importance in tumor the initiation and progression. However, the biological role and regulatory mechanism of RNF183 in non small cell lung cancer (NSCLC) development and progression are poorly defined. Hence, lung adenocarcinoma (LUAD) cell proliferation, cell apoptosis and cell cycle were measured using Cell Counting Kit-8 and flow cytometry analysis, respectively. The correlation between RNF183 and SHP2 (Src homology-2 domain-containing protein tyrosine phosphatase) was measured using coimmunoprecipitation and ubiquitination analysis in vitro. Tumor growth of NSCLC cells in vivo was measured using the nude mouse xenograft model. In this study, we verify that elevated RNF183 expression in tumor tissues of LUAD, origin from the TCGA, GEPIA, TIMER, and UALCAN database. RNF183 regulates apoptosis and cell cycle in vitro and tumor growth in vivo by activating the STAT3 pathway through ubiquitination of SHP2, a negative feedback regulator of the STAT3 pathway. Taken together, our results demonstrate that RNF183 regulates proliferation, apoptosis, and cell cycle in LUAD cells via modulation of SHP2/STAT3 signaling, suggesting the potential for targeting the RNF183-SHP2/STAT3 pathway for use in LUAD treatment.
Collapse
Affiliation(s)
- Guangbin Ye
- Medical College of Guangxi University, Nanning, Guangxi, PR China
- School of Basic Medical Sciences, Youjiang Medical University for Nationalities, Baise, Guangxi, PR China
| | - Hongcheng Luo
- Medical College of Guangxi University, Nanning, Guangxi, PR China
| | - Tingting Zhang
- Medical College of Guangxi University, Nanning, Guangxi, PR China
| | - Tianshu Lan
- Key Laboratory of Functional and Clinical Translational Medicine, Fujian Province University, Xiamen Medical College, Xiamen, Fujian, PR China
| | - Bo Ling
- School of Pharmacy, Youjiang Medical University for Nationalities, Baise, Guangxi, PR China
| | - Zhongquan Qi
- Medical College of Guangxi University, Nanning, Guangxi, PR China
- Key Laboratory of Functional and Clinical Translational Medicine, Fujian Province University, Xiamen Medical College, Xiamen, Fujian, PR China
| |
Collapse
|
15
|
Jiang H, Lin X, Liang W, Li Y, Yu X. Friedelin Alleviates the Pathogenesis of Collagenase-Induced Tendinopathy in Mice by Promoting the Selective Autophagic Degradation of p65. Nutrients 2022; 14:1673. [PMID: 35458235 PMCID: PMC9031956 DOI: 10.3390/nu14081673] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Revised: 03/29/2022] [Accepted: 04/13/2022] [Indexed: 02/06/2023] Open
Abstract
With the development of an aging population, tendinopathy has become a common musculoskeletal disease in the elderly with a high recurrence rate and no curative treatment. The inflammation mediated by NF-κB signaling plays an important role in tendon senescence and degeneration. Friedelin (FR) is a triterpenoid derived from green plants, which has a variety of pharmacological functions, such as analgesia, anti-inflammation, antioxidation, and anti-tumor functions. However, the role and mechanism of FR in tendinopathy are unclear. Here, we found that FR improved the mechanical strength of the Achilles tendon, restored the orderly arrangement of collagen fibers, reduced inflammatory cell infiltration, and promoted tenogenesis, thereby blocking the progression of tendinopathy. Mechanistically, FR promoted the autophagic degradation of p65 by enhancing the interaction between p62 and p65 and effectively inhibited the activation of the NF-κB pathway, thus alleviating the inflammatory response of tenocytes. In addition, FR recruited E3 ubiquitin enzyme RNF182 to increase the K48-linked ubiquitination of p65 and promoted p62-mediated autophagic degradation. Furthermore, blocking ubiquitination reversed the degradation of p65 by FR. Therefore, these findings identify the new pharmacological mechanism of the anti-inflammatory effect of FR and provide a new candidate drug for the treatment of tendinopathy.
Collapse
Affiliation(s)
- Huaji Jiang
- Department of Immunology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
- Department of Orthopaedics, Yuebei People’s Hospital Affiliated to Medical College of Shantou University, Shaoguan 512026, China
| | - Xuemei Lin
- Department of Pediatric Orthopedics, Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, Guangzhou 510623, China;
| | - Wei Liang
- Department of Orthopaedics, Yuebei People’s Hospital Affiliated to Medical College of Shantou University, Shaoguan 512026, China;
| | - Yiqiang Li
- Department of Pediatric Orthopedics, Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, Guangzhou 510623, China
| | - Xiao Yu
- Department of Immunology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
- Guangdong Provincial Key Lab of Single Cell Technology and Application, Southern Medical University, Guangzhou 510515, China
| |
Collapse
|
16
|
Zhang TY, Chen T, Hu WY, Li JC, Guo MY. Ammonia induces autophagy via circ-IFNLR1/miR-2188-5p/RNF182 axis in tracheas of chickens. Biofactors 2022; 48:416-427. [PMID: 34652043 DOI: 10.1002/biof.1795] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Accepted: 10/03/2021] [Indexed: 12/18/2022]
Abstract
Ammonia (NH3 ), an air pollutant in the living environment, has many toxic effects on various tissues and organs. However, the underlying mechanisms of NH3 -induced tracheal cell autophagy remains poorly understood. In present study, chickens and LMH cells were used as NH3 exposure models to investigate toxic effects. The change of tracheal tissues ultrastructure showed that NH3 exposure induced autolysosomes. The differential expression of 12 circularRNAs (circRNAs) was induced by NH3 exposure using circRNAs transcriptome analysis in broiler tracheas. We further found that circ-IFNLR1 was down-regulated, and miR-2188-5p was up-regulated in tracheal tissues under NH3 exposure. Bioinformatics analysis and dual luciferase reporter system showed that circ-IFNLR1 bound directly to miR-2188-5p and regulated each other, and miR-2188-5p regulated RNF182. Overexpression of miR-2188-5p caused autophagy and its inhibition partially reversed autophagy in LMH cells which were caused by ammonia stimulation or knockdown of circ-IFNLR1. The expressions of three autophagy-related genes (LC3, Beclin 1, and BNIP3) were observably up-regulated. Our results indicated that NH3 exposure caused autophagy through circ-IFNLR1/miR-2188-5p/RNF182. These results provided new insights for the study of ammonia on environmental toxicology on ceRNA and circRNAs in vivo and vitro.
Collapse
Affiliation(s)
- Tian-Yi Zhang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
| | - Ting Chen
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
| | - Wan-Ying Hu
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
| | - Ji-Chang Li
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
| | - Meng-Yao Guo
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
| |
Collapse
|
17
|
Borden ES, Adams AC, Buetow KH, Wilson MA, Bauman JE, Curiel-Lewandrowski C, Chow HHS, LaFleur BJ, Hastings KT. Shared Gene Expression and Immune Pathway Changes Associated with Progression from Nevi to Melanoma. Cancers (Basel) 2021; 14:cancers14010003. [PMID: 35008167 PMCID: PMC8749980 DOI: 10.3390/cancers14010003] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2021] [Revised: 12/16/2021] [Accepted: 12/20/2021] [Indexed: 02/07/2023] Open
Abstract
Simple Summary Melanoma is a deadly skin cancer, and the incidence of melanoma is rising. Chemoprevention, using small molecule drugs to prevent the development of cancer, is a key strategy that could reduce the burden of melanoma on society. The long-term goal of our study is to develop a gene signature biomarker of progression from nevi to melanoma. We found that a small number of genes can distinguish nevi from melanoma and identified shared genes and immune-related pathways that are associated with progression from nevi to melanoma across independent datasets. This study demonstrates (1) a novel approach to aid melanoma chemoprevention trials by using a gene signature as a surrogate endpoint and (2) the feasibility of determining a gene signature biomarker of melanoma progression. Abstract There is a need to identify molecular biomarkers of melanoma progression to assist the development of chemoprevention strategies to lower melanoma incidence. Using datasets containing gene expression for dysplastic nevi and melanoma or melanoma arising in a nevus, we performed differential gene expression analysis and regularized regression models to identify genes and pathways that were associated with progression from nevi to melanoma. A small number of genes distinguished nevi from melanoma. Differential expression of seven genes was identified between nevi and melanoma in three independent datasets. C1QB, CXCL9, CXCL10, DFNA5 (GSDME), FCGR1B, and PRAME were increased in melanoma, and SCGB1D2 was decreased in melanoma, compared to dysplastic nevi or nevi that progressed to melanoma. Further supporting an association with melanomagenesis, these genes demonstrated a linear change in expression from benign nevi to dysplastic nevi to radial growth phase melanoma to vertical growth phase melanoma. The genes associated with melanoma progression showed significant enrichment of multiple pathways related to the immune system. This study demonstrates (1) a novel application of bioinformatic approaches to aid clinical trials of melanoma chemoprevention and (2) the feasibility of determining a gene signature biomarker of melanomagenesis.
Collapse
Affiliation(s)
- Elizabeth S. Borden
- Department of Basic Medical Sciences, University of Arizona College of Medicine Phoenix, Phoenix, AZ 85004, USA; (E.S.B.); (A.C.A.)
- Phoenix Veterans Affairs Health Care System, Phoenix, AZ 85012, USA
| | - Anngela C. Adams
- Department of Basic Medical Sciences, University of Arizona College of Medicine Phoenix, Phoenix, AZ 85004, USA; (E.S.B.); (A.C.A.)
- Phoenix Veterans Affairs Health Care System, Phoenix, AZ 85012, USA
| | - Kenneth H. Buetow
- School of Life Sciences, Arizona State University, Tempe, AZ 85281, USA; (K.H.B.); (M.A.W.)
- Center for Evolution and Medicine, Arizona State University, Tempe, AZ 85281, USA
| | - Melissa A. Wilson
- School of Life Sciences, Arizona State University, Tempe, AZ 85281, USA; (K.H.B.); (M.A.W.)
- Center for Evolution and Medicine, Arizona State University, Tempe, AZ 85281, USA
| | - Julie E. Bauman
- Department of Medicine, University of Arizona College of Medicine Tucson, Tucson, AZ 85724, USA; (J.E.B.); (C.C.-L.); (H.-H.S.C.)
- University of Arizona Cancer Center, University of Arizona, Tucson, AZ 85724, USA
| | - Clara Curiel-Lewandrowski
- Department of Medicine, University of Arizona College of Medicine Tucson, Tucson, AZ 85724, USA; (J.E.B.); (C.C.-L.); (H.-H.S.C.)
- University of Arizona Cancer Center, University of Arizona, Tucson, AZ 85724, USA
| | - H.-H. Sherry Chow
- Department of Medicine, University of Arizona College of Medicine Tucson, Tucson, AZ 85724, USA; (J.E.B.); (C.C.-L.); (H.-H.S.C.)
- University of Arizona Cancer Center, University of Arizona, Tucson, AZ 85724, USA
| | | | - Karen Taraszka Hastings
- Department of Basic Medical Sciences, University of Arizona College of Medicine Phoenix, Phoenix, AZ 85004, USA; (E.S.B.); (A.C.A.)
- Phoenix Veterans Affairs Health Care System, Phoenix, AZ 85012, USA
- University of Arizona Cancer Center, University of Arizona, Tucson, AZ 85724, USA
- Correspondence: ; Tel.: +1-602-827-2106
| |
Collapse
|
18
|
Chen Y, Lin J, Zhao Y, Ma X, Yi H. Toll-like receptor 3 (TLR3) regulation mechanisms and roles in antiviral innate immune responses. J Zhejiang Univ Sci B 2021; 22:609-632. [PMID: 34414698 PMCID: PMC8377577 DOI: 10.1631/jzus.b2000808] [Citation(s) in RCA: 106] [Impact Index Per Article: 26.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Revised: 04/09/2021] [Accepted: 04/09/2021] [Indexed: 01/08/2023]
Abstract
Toll-like receptor 3 (TLR3) is a member of the TLR family, mediating the transcriptional induction of type I interferons (IFNs), proinflammatory cytokines, and chemokines, thereby collectively establishing an antiviral host response. Studies have shown that unlike other TLR family members, TLR3 is the only RNA sensor that is utterly dependent on the Toll-interleukin-1 receptor (TIR)-domain-containing adaptor-inducing IFN-β (TRIF). However, the details of how the TLR3-TRIF signaling pathway works in an antiviral response and how it is regulated are unclear. In this review, we focus on recent advances in understanding the antiviral mechanism of the TRIF pathway and describe the essential characteristics of TLR3 and its antiviral effects. Advancing our understanding of TLR3 may contribute to disease diagnosis and could foster the development of novel treatments for viral diseases.
Collapse
Affiliation(s)
- Yujuan Chen
- College of Veterinary Medicine, Southwest University, Chongqing 402460, China
- Chongqing Veterinary Science Engineering Research Center, Chongqing 402460, China
| | - Junhong Lin
- College of Veterinary Medicine, Southwest University, Chongqing 402460, China
- Chongqing Veterinary Science Engineering Research Center, Chongqing 402460, China
| | - Yao Zhao
- College of Veterinary Medicine, Southwest University, Chongqing 402460, China
- Chongqing Veterinary Science Engineering Research Center, Chongqing 402460, China
| | - Xianping Ma
- College of Veterinary Medicine, Southwest University, Chongqing 402460, China
- Chongqing Veterinary Science Engineering Research Center, Chongqing 402460, China
| | - Huashan Yi
- College of Veterinary Medicine, Southwest University, Chongqing 402460, China.
- Chongqing Veterinary Science Engineering Research Center, Chongqing 402460, China.
- Immunology Research Center, Medical Research Institute, Southwest University, Chongqing 402460, China.
| |
Collapse
|
19
|
Transcriptome sequencing provides insights into the mechanism of hypoxia adaption in bighead carp (Hypophthalmichthys nobilis). COMPARATIVE BIOCHEMISTRY AND PHYSIOLOGY D-GENOMICS & PROTEOMICS 2021; 40:100891. [PMID: 34404015 DOI: 10.1016/j.cbd.2021.100891] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Revised: 08/03/2021] [Accepted: 08/03/2021] [Indexed: 12/13/2022]
Abstract
Hypoxia negatively affects the behavior, immunology, physiology, and growth of fish. Therefore, uncovering the genetic mechanisms underlying hypoxia adaptation and tolerance in fish prior to any genetic improvement is essential. Bighead carp is one of the most important freshwater fish species in aquaculture worldwide; however, this species does not have a strong ability to tolerate hypoxia. In this study, the dissolved oxygen level (0.6 mg/L) was maintained above the asphyxiation point of bighead carp for a long time to simulate hypoxia stress. The liver, gills, and heart were sampled before (0 h) and after (1 h, 2 h, 4 h) the hypoxia tests. Glutathione peroxidase (GPx) and catalase (CAT) activities and malondialdehyde (MDA) levels in the liver were significantly (p < 0.05) elevated at 1 h after hypoxic stress. By observing tissue morphology, the cell structure of the liver and gill tissues was found to change to varying degrees before and after hypoxia stress. Transcriptome sequencing was performed on 36 samples of gill, liver, and heart at four time points, and a total of 293.55G of data was obtained. In the early phase (0-1 h), differentially expressed genes (DEGs, 807 genes upregulated, 654 genes downregulated) were mainly enriched in signal transduction, such as cytokine-cytokine receptor interactions and ECM-receptor interactions. In the middle phase (0-2 h), DEGs (1201 genes upregulated and 2036 genes downregulated) were mainly enriched in regulation and adaptation, such as the MAPK and FoxO signaling pathways. Finally, in the later phase (0-4 h), DEGs (3975 genes upregulated and 4412 genes downregulated) were mainly enriched in tolerance and apoptosis, such as the VEGF signaling pathway and apoptosis. The genes with the most remarkable upregulation at different time points in the three tissues had some similarities. Genetic differences in these genes may be responsible for the differences in hypoxia tolerance among individuals. Altogether, our study provides new insights into the molecular mechanisms of hypoxia adaptation in fish. Further, the key regulatory genes identified provide genetic resources for breeding hypoxia-tolerant bighead carp species.
Collapse
|
20
|
Budroni V, Versteeg GA. Negative Regulation of the Innate Immune Response through Proteasomal Degradation and Deubiquitination. Viruses 2021; 13:584. [PMID: 33808506 PMCID: PMC8066222 DOI: 10.3390/v13040584] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2021] [Revised: 03/26/2021] [Accepted: 03/27/2021] [Indexed: 12/25/2022] Open
Abstract
The rapid and dynamic activation of the innate immune system is achieved through complex signaling networks regulated by post-translational modifications modulating the subcellular localization, activity, and abundance of signaling molecules. Many constitutively expressed signaling molecules are present in the cell in inactive forms, and become functionally activated once they are modified with ubiquitin, and, in turn, inactivated by removal of the same post-translational mark. Moreover, upon infection resolution a rapid remodeling of the proteome needs to occur, ensuring the removal of induced response proteins to prevent hyperactivation. This review discusses the current knowledge on the negative regulation of innate immune signaling pathways by deubiquitinating enzymes, and through degradative ubiquitination. It focusses on spatiotemporal regulation of deubiquitinase and E3 ligase activities, mechanisms for re-establishing proteostasis, and degradation through immune-specific feedback mechanisms vs. general protein quality control pathways.
Collapse
Affiliation(s)
| | - Gijs A. Versteeg
- Max Perutz Labs, Department of Microbiology, Immunobiology, and Genetics, University of Vienna, Vienna Biocenter (VBC), 1030 Vienna, Austria;
| |
Collapse
|
21
|
Bale S, Varga J, Bhattacharyya S. Role of RP105 and A20 in negative regulation of toll-like receptor activity in fibrosis: potential targets for therapeutic intervention. AIMS ALLERGY AND IMMUNOLOGY 2021. [DOI: 10.3934/allergy.2021009] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
|
22
|
Li K, Zhang Z, Mei Y, Yang Q, Qiao S, Ni C, Yao Y, Li X, Li M, Wei D, Fu W, Guo X, Huang X, Yang H. Metallothionein-1G suppresses pancreatic cancer cell stemness by limiting activin A secretion via NF-κB inhibition. Theranostics 2021; 11:3196-3212. [PMID: 33537082 PMCID: PMC7847690 DOI: 10.7150/thno.51976] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2020] [Accepted: 12/15/2020] [Indexed: 12/15/2022] Open
Abstract
Resistance to chemotherapy is a long-standing problem in the management of cancer, and cancer stem cells are regarded as the main source of this resistance. This study aimed to investigate metallothionein (MT)-1G involvement in the regulation of cancer stemness and provide a strategy to overcome chemoresistance in pancreatic ductal adenocarcinoma (PDAC). Methods: MT1G was identified as a critical factor related with gemcitabine resistance in PDAC cells by mRNA microarray. Its effects on PDAC stemness were evaluated through sphere formation and tumorigenicity. LC-MS/MS analysis of conditional medium revealed that activin A, a NF-κB target, was a major protein secreted from gemcitabine resistant PDAC cells. Both loss-of-function and gain-of-function approaches were used to validate that MT1G inhibited NF-κB-activin A pathway. Orthotopic pancreatic tumor model was employed to explore the effects on gemcitabine resistance with recombinant follistatin to block activin A. Results: Downregulation of MT1G due to hypermethylation of its promoter is related with pancreatic cancer stemness. Secretome analysis revealed that activin A, a NF-κB target, was highly secreted by drug resistant cells. It promotes pancreatic cancer stemness in Smad4-dependent or independent manners. Mechanistically, MT1G negatively regulates NF-κB signaling and promotes the degradation of NF-κB p65 subunit by enhancing the expression of E3 ligase TRAF7. Blockade of activin A signaling with follistatin could overcome gemcitabine resistance. Conclusions: MT1G suppresses PDAC stemness by limiting activin A secretion via NF-κB inhibition. The blockade of the activin A signaling with follistatin may provide a promising therapeutic strategy for overcoming gemcitabine resistance in PDAC.
Collapse
Affiliation(s)
- Kai Li
- School of Life Science and Technology, Harbin Institute of Technology, Harbin, 150001, China
| | - Zhicheng Zhang
- Department of General Surgery, Fourth Affiliated Hospital of Harbin Medical University, Harbin, 150001, China
| | - Yu Mei
- School of Life Science and Technology, Harbin Institute of Technology, Harbin, 150001, China
| | - Qingzhu Yang
- School of Life Science and Technology, Harbin Institute of Technology, Harbin, 150001, China
| | - Shupei Qiao
- School of Life Science and Technology, Harbin Institute of Technology, Harbin, 150001, China
| | - Cheng Ni
- School of Life Science and Technology, Harbin Institute of Technology, Harbin, 150001, China
| | - Yao Yao
- School of Life Science and Technology, Harbin Institute of Technology, Harbin, 150001, China
| | - Xinyuan Li
- School of Life Science and Technology, Harbin Institute of Technology, Harbin, 150001, China
| | - Mengmeng Li
- School of Life Science and Technology, Harbin Institute of Technology, Harbin, 150001, China
| | - Dongdong Wei
- School of Life Science and Technology, Harbin Institute of Technology, Harbin, 150001, China
| | - Wangjun Fu
- School of Life Science and Technology, Harbin Institute of Technology, Harbin, 150001, China
| | - Xuefei Guo
- School of Life Science and Technology, Harbin Institute of Technology, Harbin, 150001, China
| | - Xuemei Huang
- School of Life Science and Technology, Harbin Institute of Technology, Harbin, 150001, China
| | - Huanjie Yang
- School of Life Science and Technology, Harbin Institute of Technology, Harbin, 150001, China
| |
Collapse
|
23
|
Geng R, Zheng Y, Zhao L, Huang X, Qiang R, Zhang R, Guo X, Li R. RNF183 Is a Prognostic Biomarker and Correlates With Tumor Purity, Immune Infiltrates in Uterine Corpus Endometrial Carcinoma. Front Genet 2020; 11:595733. [PMID: 33324448 PMCID: PMC7726321 DOI: 10.3389/fgene.2020.595733] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Accepted: 11/02/2020] [Indexed: 12/21/2022] Open
Abstract
RNF183, a member of the E3 ubiquitin ligase, has been shown to involve in carcinogenesis and proposed as one of the biomarkers in Uterine Corpus Endometrial Carcinoma (UCEC). However, no research focused on the role of RNF183 in UCEC. We analyzed the expression and immune infiltration of RNF183 in UCEC. TIMER, UALCAN, and GEPIA were used to analyze the gene expression of RNF183. We emplored Kaplan-Meier Plotter to examine the overall survival and progression-free survival of RNF183, and applied GeneMANIA to identify RNF183-related functional networks. LinkedOmics was helpful to identify the differential gene expression of RNF183, and to further analyze gene ontology and the genome pathways in the Kyoto Protocol. Finally, we used TIMER to investigate the immune infiltration of RNF183 in UCEC. Otherwise, we partly verified the results of bioinformatics analysis that RNF183 controlled ERα expression in ERα-positive Ishikawa cells dependent on its RING finger domain. We also found that ERα increased the stability of RNF183 through the post-translational mechanism. Together, patients with a high level of RNF183 harbor favorable overall and progression-free survival. High expression of RNF183 was associated with a low stage, endometrioid, and TP53 Non-Mutant status in endometrial cancer. The RNF183 expression was greater at higher expression and the tumor stage was greater at the lower level. On the side of immunization, high level of RNF183 in UCEC is negatively related to tumor purity, infiltrating levels of CD4 + T cells, neutrophils, and dendritic cells. Besides, the expression of RNF183 in UCEC is significantly correlated with the expression of several immune cell markers, including B cell, M1 macrophage marker, M2 Macrophage, Dendritic cell, Th1 markers, Th2 markers, Treg markers, and T cell exhaustion markers, indicating its role in regulating tumor immunity. These results suggested that RNF183 may be considered as a novel prognostic factor in endometrial cancer and an early diagnostic indicator for patients with UCEC.
Collapse
Affiliation(s)
- Rong Geng
- Department of Gynecology and Obstetrics, The First Affiliated Hospital, Jinan University, Guangzhou, China.,Department of Gynecology, Affiliated Foshan Maternity & Child Healthcare Hospital, Southern Medical University, Foshan, China.,Foshan Maternal and Children Healthy Research Institute, Affiliated Foshan Maternity & Child Healthcare Hospital, Southern Medical University, Foshan, China
| | - Yuhua Zheng
- Department of Gynecology, Affiliated Foshan Maternity & Child Healthcare Hospital, Southern Medical University, Foshan, China
| | - Lijie Zhao
- Department of Gynecology, Affiliated Foshan Maternity & Child Healthcare Hospital, Southern Medical University, Foshan, China
| | - Xiaobin Huang
- Department of Gynecology, Affiliated Foshan Maternity & Child Healthcare Hospital, Southern Medical University, Foshan, China
| | - Rong Qiang
- Department of Gynecology, Affiliated Foshan Maternity & Child Healthcare Hospital, Southern Medical University, Foshan, China
| | - Rujian Zhang
- Department of Gynecology, Affiliated Foshan Maternity & Child Healthcare Hospital, Southern Medical University, Foshan, China
| | - Xiaoling Guo
- Department of Gynecology, Affiliated Foshan Maternity & Child Healthcare Hospital, Southern Medical University, Foshan, China
| | - Ruiman Li
- Department of Gynecology and Obstetrics, The First Affiliated Hospital, Jinan University, Guangzhou, China
| |
Collapse
|
24
|
Identification and verification of EOMEs regulated network in Alopecia areata. Int Immunopharmacol 2020; 84:106544. [DOI: 10.1016/j.intimp.2020.106544] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2020] [Revised: 04/22/2020] [Accepted: 04/23/2020] [Indexed: 01/25/2023]
|
25
|
The Role of Tissue-Specific Ubiquitin Ligases, RNF183, RNF186, RNF182 and RNF152, in Disease and Biological Function. Int J Mol Sci 2020; 21:ijms21113921. [PMID: 32486221 PMCID: PMC7313026 DOI: 10.3390/ijms21113921] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Revised: 05/27/2020] [Accepted: 05/28/2020] [Indexed: 12/25/2022] Open
Abstract
Ubiquitylation plays multiple roles not only in proteasome-mediated protein degradation but also in various other cellular processes including DNA repair, signal transduction, and endocytosis. Ubiquitylation is mediated by ubiquitin ligases, which are predicted to be encoded by more than 600 genes in humans. RING finger (RNF) proteins form the majority of these ubiquitin ligases. It has also been predicted that there are 49 RNF proteins containing transmembrane regions in humans, several of which are specifically localized to membrane compartments in the secretory and endocytic pathways. Of these, RNF183, RNF186, RNF182, and RNF152 are closely related genes with high homology. These genes share a unique common feature of exhibiting tissue-specific expression patterns, such as in the kidney, nervous system, and colon. The products of these genes are also reported to be involved in various diseases such as cancers, inflammatory bowel disease, Alzheimer's disease, and chronic kidney disease, and in various biological functions such as apoptosis, endoplasmic reticulum stress, osmotic stress, nuclear factor-kappa B (NF-κB), mammalian target of rapamycin (mTOR), and Notch signaling. This review summarizes the current knowledge of these tissue-specific ubiquitin ligases, focusing on their physiological roles and significance in diseases.
Collapse
|