1
|
He Y, Yang Z, Guo D, Luo C, Liu Q, Xian L, Yang F, Huang C, Wei Q. The multifaceted nature of SUMOylation in heart disease and its therapeutic potential. Mol Cell Biochem 2025:10.1007/s11010-025-05286-z. [PMID: 40287894 DOI: 10.1007/s11010-025-05286-z] [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: 11/05/2024] [Accepted: 04/06/2025] [Indexed: 04/29/2025]
Abstract
SUMOylation (SUMO), a crucial post-translational modification, is implicated in the regulation of diverse biological processes and plays a pivotal role in both the maintenance of cardiac function and progression and treatment of heart disease. Here, we reviewed the mechanisms by which SUMO-related various aspects of cardiac function and disease, including cardiac hypertrophy, heart failure, ischemia-reperfusion injury, and myocardial infarction. Furthermore, we highlight its potential as a therapeutic target.
Collapse
Affiliation(s)
- Ying He
- The Fourth Affiliated Hospital of Guangxi Medical University, Liuzhou, Guangxi, China
- The Second Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Zhijie Yang
- The Fourth Affiliated Hospital of Guangxi Medical University, Liuzhou, Guangxi, China
| | - Dan Guo
- Medical College, Guangxi University, Nanning, Guangxi, China
| | - Cheng Luo
- The Fourth Affiliated Hospital of Guangxi Medical University, Liuzhou, Guangxi, China
| | - Qiaoqiao Liu
- The Fourth Affiliated Hospital of Guangxi Medical University, Liuzhou, Guangxi, China
| | - Lei Xian
- The Second Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Fan Yang
- The Fourth Affiliated Hospital of Guangxi Medical University, Liuzhou, Guangxi, China.
- Liuzhou Key Laboratory of Primary Cardiomyopathy in Prevention and Treatment, Liuzhou, Guangxi, China.
| | - Chusheng Huang
- The Second Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China.
| | - Qingjun Wei
- The Second Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China.
| |
Collapse
|
2
|
Che Y, Liu YT, Wang ZP, Feng YZ, Xia HX, Yuan Y, Zhou H, Qiu HL, Hu ML, Wang SS, Tang QZ. Cardiac tumour necrosis factor receptor-associated factor 7 mediates the ubiquitination of apoptosis signal-regulating kinase 1 and aggravates cardiac hypertrophy. Cardiovasc Res 2024; 120:2031-2046. [PMID: 39373232 DOI: 10.1093/cvr/cvae217] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/18/2022] [Revised: 05/14/2023] [Accepted: 07/14/2024] [Indexed: 10/08/2024] Open
Abstract
AIMS Cardiac remodelling is a common pathophysiological process in the development of various cardiovascular diseases, but there is still a lack of effective interventions. Tumour necrosis receptor-associated factor 7 (TRAF7) belongs to the tumour necrosis factor receptor-associated factor family and plays an important role in biological processes. Previous studies have shown that TRAF7 mutations lead to congenital defects and malformations of the heart. However, the molecular mechanisms of TRAF7 in the underlying pathogenesis of pathological cardiac hypertrophy remain unknown. We aim to study the molecular mechanisms and effects of TRAF7 in cardiac remodelling and whether it has the potential to become a therapeutic target for cardiac remodelling. METHODS AND RESULTS The pressure overload-induced cardiac hypertrophy model in mice was established via transverse aortic constriction (TAC) surgery, and cardiomyocytes were treated with phenylephrine (PE) to induce hypertrophic phenotype. Levels of cardiac dysfunction and remodelling were measured with echocardiography and tissue or cell staining. RNA sequencing, western blot, qRT-PCR, co-immunoprecipitation, and in vivo ubiquitination assays were used to explore the molecular mechanisms. The results showed that the expression of TRAF7 increased gradually during the development of hypertrophy. Accordingly, TRAF7 significantly exacerbated the PE-induced enlargement of primary neonatal Sprague-Dawley rat cardiomyocytes, whereas TRAF7 knockdown alleviated the hypertrophic phenotype in primary cardiomyocytes. Cardiac-specific overexpression of TRAF7 accelerated hypertrophic phenotype in mice and cardiac-specific Traf7 conditional knockout mice improved hypertrophic phenotype induced by TAC. Mechanistically, TRAF7 directly interacted with apoptosis signal-regulating kinase-1 (ASK1) and promoted ASK1 phosphorylation by mediating the K63-linked ubiquitination of ASK1 in response to PE stimulation, which then promoted ASK1 activation and downstream signalling during cardiac hypertrophy. Notably, the pro-hypertrophic effect of TRAF7 was largely blocked by GS4997 in vitro and cardiac-specific Ask1 conditional knockout in vivo. CONCLUSION In summary, we identified TRAF7 as an essential regulator during cardiac hypertrophy, and modulation of the regulatory axis between TRAF7 and ASK1 could be a novel therapeutic strategy to prevent this pathological process.
Collapse
MESH Headings
- Animals
- Ubiquitination
- Myocytes, Cardiac/enzymology
- Myocytes, Cardiac/metabolism
- Myocytes, Cardiac/pathology
- Ventricular Remodeling
- Signal Transduction
- MAP Kinase Kinase Kinase 5/metabolism
- MAP Kinase Kinase Kinase 5/genetics
- Mice, Inbred C57BL
- Disease Models, Animal
- Cells, Cultured
- Male
- Apoptosis
- Mice, Knockout
- Ventricular Function, Left
- Hypertrophy, Left Ventricular/enzymology
- Hypertrophy, Left Ventricular/metabolism
- Hypertrophy, Left Ventricular/pathology
- Hypertrophy, Left Ventricular/physiopathology
- Hypertrophy, Left Ventricular/genetics
- Rats, Sprague-Dawley
- Cardiomegaly/metabolism
- Cardiomegaly/enzymology
- Cardiomegaly/pathology
- Cardiomegaly/genetics
- Cardiomegaly/physiopathology
- Rats
- Phosphorylation
Collapse
Affiliation(s)
- Yan Che
- Department of Cardiology, Renmin Hospital of Wuhan University, Jiefang Road 238, Wuhan 430060, PR China
- Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan University, Jiefang Road 238, Wuhan 430060, PR China
| | - Yu-Ting Liu
- Department of Cardiology, Renmin Hospital of Wuhan University, Jiefang Road 238, Wuhan 430060, PR China
- Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan University, Jiefang Road 238, Wuhan 430060, PR China
| | - Zhao-Peng Wang
- Department of Cardiology, Renmin Hospital of Wuhan University, Jiefang Road 238, Wuhan 430060, PR China
- Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan University, Jiefang Road 238, Wuhan 430060, PR China
- Department of Cardiology, Taikang Tongji (Wuhan) Hospital, Wuhan 420060, PR China
| | - Yi-Zhou Feng
- Department of Cardiology, Renmin Hospital of Wuhan University, Jiefang Road 238, Wuhan 430060, PR China
- Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan University, Jiefang Road 238, Wuhan 430060, PR China
| | - Hong-Xia Xia
- Department of Cardiology, Renmin Hospital of Wuhan University, Jiefang Road 238, Wuhan 430060, PR China
- Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan University, Jiefang Road 238, Wuhan 430060, PR China
| | - Yuan Yuan
- Department of Cardiology, Renmin Hospital of Wuhan University, Jiefang Road 238, Wuhan 430060, PR China
- Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan University, Jiefang Road 238, Wuhan 430060, PR China
| | - Heng Zhou
- Department of Cardiology, Renmin Hospital of Wuhan University, Jiefang Road 238, Wuhan 430060, PR China
- Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan University, Jiefang Road 238, Wuhan 430060, PR China
| | - Hong-Liang Qiu
- Department of Cardiology, Renmin Hospital of Wuhan University, Jiefang Road 238, Wuhan 430060, PR China
- Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan University, Jiefang Road 238, Wuhan 430060, PR China
| | - Man-Li Hu
- Gannan Medical University, Ganzhou 341000, PR China
| | - Sha-Sha Wang
- Department of Cardiology, Renmin Hospital of Wuhan University, Jiefang Road 238, Wuhan 430060, PR China
- Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan University, Jiefang Road 238, Wuhan 430060, PR China
| | - Qi-Zhu Tang
- Department of Cardiology, Renmin Hospital of Wuhan University, Jiefang Road 238, Wuhan 430060, PR China
- Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan University, Jiefang Road 238, Wuhan 430060, PR China
| |
Collapse
|
3
|
Han J, Wu J, Kou WT, Xie LN, Tang YL, Zhi DL, Li P, Chen DQ. New insights into SUMOylation and NEDDylation in fibrosis. Front Pharmacol 2024; 15:1476699. [PMID: 39697538 PMCID: PMC11652140 DOI: 10.3389/fphar.2024.1476699] [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: 08/06/2024] [Accepted: 11/12/2024] [Indexed: 12/20/2024] Open
Abstract
Fibrosis is the outcome of any abnormal tissue repair process that results in normal tissue replacement with scar tissue, leading to persistent tissue damage and cellular injury. During the process of fibrosis, many cytokines and chemokines are involved, and their activities are controlled by post-translational modifications, especially SUMOylation and NEDDylation. Both these modifications entail a three-step process of activation, conjugation, and ligation that involves three kinds of enzymes, namely, E1 activating, E2 conjugating, and E3 ligase enzymes. SUMOylation participates in organ fibrosis by modulating FXR, PML, TGF-β receptor I, Sirt3, HIF-1α, and Sirt1, while NEDDylation influences organ fibrosis by regulating cullin3, NIK, SRSF3, and UBE2M. Further investigations exhibit the therapeutic potentials of SUMOylation/NEDDylation activators and inhibitors against organ fibrosis, especially ginkgolic acid in SUMOylation and MLN4924 in NEDDylation. These results demonstrate the therapeutic effects of SUMOylation and NEDDylation against organ fibrosis and highlight their activators as well as inhibitors as potential candidates. In the future, deeper investigations of SUMOylation and NEDDylation are needed to identify novel substrates against organ fibrosis; moreover, clinical investigations are needed to determine the therapeutic effects of their activators and inhibitors that can benefit patients. This review highlights that SUMOylation and NEDDylation function as potential therapeutic targets for organ fibrosis.
Collapse
Affiliation(s)
- Jin Han
- Northwest University Chang An Hospital, Faculty of Life Sciences and Medicine, Northwest University, Xi’an, Shaanxi, China
- Department of Nephrology, Chang An District Hospital, Xi’an, Shaanxi, China
| | - Jun Wu
- School of Pharmacy, Shandong College of Traditional Chinese Medicine, Yantai, Shandong, China
| | - Wen-Tao Kou
- Northwest University Chang An Hospital, Faculty of Life Sciences and Medicine, Northwest University, Xi’an, Shaanxi, China
- Department of Nephrology, Chang An District Hospital, Xi’an, Shaanxi, China
| | - Li-Na Xie
- Northwest University Chang An Hospital, Faculty of Life Sciences and Medicine, Northwest University, Xi’an, Shaanxi, China
- Department of Nephrology, Chang An District Hospital, Xi’an, Shaanxi, China
| | - Ya-Li Tang
- Northwest University Chang An Hospital, Faculty of Life Sciences and Medicine, Northwest University, Xi’an, Shaanxi, China
- Department of Nephrology, Chang An District Hospital, Xi’an, Shaanxi, China
| | - Da-Long Zhi
- Northwest University Chang An Hospital, Faculty of Life Sciences and Medicine, Northwest University, Xi’an, Shaanxi, China
- Department of Nephrology, Chang An District Hospital, Xi’an, Shaanxi, China
| | - Ping Li
- Beijing Key Lab for Immune-Mediated Inflammatory Diseases, Institute of Clinical Medical Sciences, China-Japan Friendship Hospital, Beijing, China
| | - Dan-Qian Chen
- Northwest University Chang An Hospital, Faculty of Life Sciences and Medicine, Northwest University, Xi’an, Shaanxi, China
- Department of Nephrology, Chang An District Hospital, Xi’an, Shaanxi, China
| |
Collapse
|
4
|
Zenke K, Sugimoto R, Watanabe S, Muroi M. NF-κB p105-mediated nuclear translocation of ERK is required for full activation of IFNγ-induced iNOS expression. Cell Signal 2024; 124:111424. [PMID: 39304100 DOI: 10.1016/j.cellsig.2024.111424] [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: 06/11/2024] [Revised: 08/30/2024] [Accepted: 09/17/2024] [Indexed: 09/22/2024]
Abstract
Inducible nitric oxidase (iNOS) encoded by Nos2 is a representative IFNγ-inducible effector molecule that plays an important role in both innate and adaptive immunity. In the present study, we demonstrated that full-length NF-κB p105 (p105), which is a precursor of NF-κB p50 (p50), is required for full activation of IFNγ-induced iNOS expression in the RAW264.7 mouse macrophage cell line. In comparison to wild-type (WT) RAW264.7 cells, p105 KO RAW264.7 (p105 KO) cells completely lost IFNγ-induced iNOS expression. Despite the limited effect of exogenous expression of p50 in p105 KO cells on IFNγ-induced Nos2 promoter activity, p105 expression fully restored IFNγ-induced Nos2 promoter activity to a level comparable to that of WT cells, suggesting an important role for full-length p105 in IFNγ-induced iNOS expression. While the expression and phosphorylation of JAK1 and STAT1 were rather enhanced in p105 KO cells, the phosphorylation of c-Jun downstream of MAPK signaling was decreased. IFNγ-induced phosphorylation of ERK, a kinase for IFNγ-induced c-Jun phosphorylation, was not significantly reduced in p105 KO cells, although the nuclear activity of ERK was significantly decreased due to its reduced translocation to the nucleus. Expression of iNOS, nuclear translocation of ERK, and phosphorylation of c-Jun were restored by stable supplementation of p105 in p105 KO cells. These results suggest that p105 is required for the nuclear translocation of ERK and the subsequent phosphorylation of c-Jun, which are necessary for full activation of IFNγ-induced iNOS expression. Reduced nuclear translocation of ERK in p105 KO cells was also observed in the activation of ERK following serum starvation, further suggesting that the involvement of p105 in ERK nuclear translocation is not limited to IFNγ-stimulated cells but is a more general function of p105.
Collapse
Affiliation(s)
- Kosuke Zenke
- Research Institute of Pharmaceutical Sciences, Musashino University, 1-1-20 Shinmachi, Nishitokyo-shi, Tokyo 202-8585, Japan
| | - Rino Sugimoto
- Research Institute of Pharmaceutical Sciences, Musashino University, 1-1-20 Shinmachi, Nishitokyo-shi, Tokyo 202-8585, Japan
| | - Sachiko Watanabe
- Research Institute of Pharmaceutical Sciences, Musashino University, 1-1-20 Shinmachi, Nishitokyo-shi, Tokyo 202-8585, Japan
| | - Masashi Muroi
- Research Institute of Pharmaceutical Sciences, Musashino University, 1-1-20 Shinmachi, Nishitokyo-shi, Tokyo 202-8585, Japan.
| |
Collapse
|
5
|
Ruan Z, Li Y, Chen Y. HECTD3 promotes NLRP3 inflammasome and pyroptosis to exacerbate diabetes-related cognitive impairment by stabilising MALT1 to regulate JNK pathway. Arch Physiol Biochem 2024; 130:373-384. [PMID: 35913790 DOI: 10.1080/13813455.2022.2093377] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Accepted: 06/17/2022] [Indexed: 11/02/2022]
Abstract
BACKGROUND HECTD3 (HECT domain E3 ubiquitin protein ligase 3) exerts biological activities in neuroinflammation of distinct diseases, such as autoimmune encephalomyelitis and donations after heart death. However, the effect of HECTD3 on diabetes-associated cognitive decline (DACD) remains unclear. METHODS Wild-type or HECTD3-knockout rats were administered with streptozotocin to establish diabetic model. Pathological changes in the hippocampus were assessed by NISSL and haematoxylin and eosin staining. Morris water maze test was used to assess cognitive function. Neuronal survival and inflammation were investigated by immunofluorescence staining and ELISA assay. NLRP3 inflammasome and pyroptosis were assessed by western blot, immunofluorescence and flow cytometry assays. RESULTS HECTD3 was up-regulated in hippocampus of streptozotocin-induced diabetic rats and high glucose-induced PC12 cells. Knockout of HECTD3 increased the number of neurons and improved the learning and memory function. Moreover, knockout of HECTD3 promoted in vivo neuronal survival, and reduced levels of IL-1β, TNF-α, and IL-6 in the hippocampus. Silencing of HECTD3 increased cell viability, and reduced IL-1β, TNF-α, and IL-6 in high glucose-induced PC12 cells. Fluorescence intensities of NLRP3, GSDMD-N and caspase-1 were reduced in HECTD3-knockout diabetic rats, and knockdown of HECTD3 down-regulated protein expression of NLRP3, GSDMD-N, caspase-1, IL-1β, and IL-18 in high glucose-induced PC12 cells to suppress the pyroptosis. HECTD3 promoted the stability of mucosa-associated lymphoid tissue 1 (MALT1) through up-regulation of c-JUN and phospho (p)-JNK in high glucose-induced PC12 cells. Over-expression of MALT1 attenuated neuroprotective effects of HECTD3 silencing on high glucose-induced PC12 cells. CONCLUSION HECTD3 silencing exerted neuroprotective effect against DACD through MALT1-mediated JNK signalling.HighlightsHECTD3 was up-regulated in hippocampus of streptozotocin-induced diabetic rats and high glucose-induced PC12.Knockout of HECTD3 promoted in vivo neuronal survival, reduced inflammation and pyroptosis, and improved the learning and memory function in diabetic rats.Knockout of HECTD3 suppressed the activation of NLRP3 inflammasome in diabetic rats.Silencing of HECTD3 exerted neuroprotective effects through MALT1-mediated JNK signalling.
Collapse
Affiliation(s)
- Zhongfan Ruan
- Department of Neurology, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, China
| | - Yan Li
- Department of Anesthesiology, The Affiliated Nanhua Hospital, Hengyang Medical School, University of South China, Hengyang, China
| | - Yanfang Chen
- Department of Neurology, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, China
| |
Collapse
|
6
|
Zhao Y, Huang J, Zhao K, Li M, Wang S. Ubiquitination and deubiquitination in the regulation of N 6-methyladenosine functional molecules. J Mol Med (Berl) 2024; 102:337-351. [PMID: 38289385 DOI: 10.1007/s00109-024-02417-9] [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: 05/06/2023] [Revised: 10/17/2023] [Accepted: 01/09/2024] [Indexed: 02/21/2024]
Abstract
N6 methyladenosine (m6A) is the most prevalent RNA epigenetic modification, regulated by methyltransferases and demethyltransferases and recognized by methylation-related reading proteins to impact mRNA splicing, translocation, stability, and translation efficiency. It significantly affects a variety of activities, including stem cell maintenance and differentiation, tumor formation, immune regulation, and metabolic disorders. Ubiquitination refers to the specific modification of target proteins by ubiquitin molecule in response to a series of enzymes. E3 ligases connect ubiquitin to target proteins and usually lead to protein degradation. On the contrary, deubiquitination induced by deubiquitinating enzymes (DUBs) can separate ubiquitin and regulate the stability of protein. Recent studies have emphasized the potential importance of ubiquitination and deubiquitination in controlling the function of m6A modification. In this review, we discuss the impact of ubiquitination and deubiquitination on m6A functional molecules in diseases, such as metabolism, cellular stress, and tumor growth.
Collapse
Affiliation(s)
- Yue Zhao
- Department of Laboratory Medicine, Affiliated Hospital, Jiangsu University, Jiefang Road No 438, Zhenjiang, 212002, China
- Department of Immunology, Jiangsu Key Laboratory of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, China
| | - Jiaojiao Huang
- Department of Immunology, Jiangsu Key Laboratory of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, China
| | - Kexin Zhao
- Department of Immunology, Jiangsu Key Laboratory of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, China
| | - Min Li
- Department of Immunology, Jiangsu Key Laboratory of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, China
| | - Shengjun Wang
- Department of Laboratory Medicine, Affiliated Hospital, Jiangsu University, Jiefang Road No 438, Zhenjiang, 212002, China.
- Department of Immunology, Jiangsu Key Laboratory of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, China.
| |
Collapse
|
7
|
Chen X, Ma J, Wang ZW, Wang Z. The E3 ubiquitin ligases regulate inflammation in cardiovascular diseases. Semin Cell Dev Biol 2024; 154:167-174. [PMID: 36872193 DOI: 10.1016/j.semcdb.2023.02.008] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Revised: 02/19/2023] [Accepted: 02/20/2023] [Indexed: 03/06/2023]
Abstract
Accumulating evidence has illustrated that the E3 ubiquitin ligases critically participate in the development and progression of cardiovascular diseases. Dysregulation of E3 ubiquitin ligases exacerbates cardiovascular diseases. Blockade or activation of E3 ubiquitin ligases mitigates cardiovascular performance. Therefore, in this review, we mainly introduced the critical role and underlying molecular mechanisms of E3 ubiquitin ligase NEDD4 family in governing the initiation and progression of cardiovascular diseases, including ITCH, WWP1, WWP2, Smurf1, Smurf2, Nedd4-1 and Nedd4-2. Moreover, the functions and molecular insights of other E3 ubiquitin ligases, such as F-box proteins, in cardiovascular disease development and malignant progression are described. Furthermore, we illustrate several compounds that alter the expression of E3 ubiquitin ligases to alleviate cardiovascular diseases. Therefore, modulation of E3 ubiquitin ligases could be a novel and promising strategy for improvement of therapeutic efficacy of deteriorative cardiovascular diseases.
Collapse
Affiliation(s)
- Xiao Chen
- Department of Cardiology, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
| | - Jia Ma
- Department of Biochemistry and Molecular Biology, School of Laboratory Medicine, Bengbu Medical College, Bengbu, Anhui, 233030, China
| | - Zhi-Wei Wang
- The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, China.
| | - Zhiting Wang
- Department of Cardiology, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, China.
| |
Collapse
|
8
|
Lan L, Peng S, Zhang R, He H, Yang Y, Xi B, Zhang J. Serum proteomic biomarker investigation of vascular depression using data-independent acquisition: a pilot study. Front Aging Neurosci 2024; 16:1341374. [PMID: 38384936 PMCID: PMC10879412 DOI: 10.3389/fnagi.2024.1341374] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Accepted: 01/22/2024] [Indexed: 02/23/2024] Open
Abstract
Background Vascular depression (VaD) is a depressive disorder closely associated with cerebrovascular disease and vascular risk factors. It remains underestimated owing to challenging diagnostics and limited information regarding the pathophysiological mechanisms of VaD. The purpose of this study was to analyze the proteomic signatures and identify the potential biomarkers with diagnostic significance in VaD. Methods Deep profiling of the serum proteome of 35 patients with VaD and 36 controls was performed using liquid chromatography-tandem mass spectrometry (LC-MS/MS). Functional enrichment analysis of the quantified proteins was based on Gene Ontology (GO), Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway, and Reactome databases. Machine learning algorithms were used to screen candidate proteins and develop a protein-based model to effectively distinguish patients with VaD. Results There were 29 up-regulated and 31 down-regulated proteins in the VaD group compared to the controls (|log2FC| ≥ 0.26, p ≤ 0.05). Enrichment pathways analyses showed that neurobiological processes related to synaptic vesicle cycle and axon guidance may be dysregulated in VaD. Extrinsic component of synaptic vesicle membrane was the most enriched term in the cellular components (CC) terms. 19 candidate proteins were filtered for further modeling. A nomogram was developed with the combination of HECT domain E3 ubiquitin protein ligase 3 (HECTD3), Nidogen-2 (NID2), FTO alpha-ketoglutarate-dependent dioxygenase (FTO), Golgi membrane protein 1 (GOLM1), and N-acetylneuraminate lyase (NPL), which could be used to predict VaD risk with favorable efficacy. Conclusion This study offers a comprehensive and integrated view of serum proteomics and contributes to a valuable proteomics-based diagnostic model for VaD.
Collapse
Affiliation(s)
- Liuyi Lan
- Department of Neurology, Zhongnan Hospital, Wuhan University, Wuhan, China
| | - Sisi Peng
- Department of Neuropsychology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Ran Zhang
- Department of Neurology, Zhongnan Hospital, Wuhan University, Wuhan, China
| | - Haoying He
- Department of Neurology, Zhongnan Hospital, Wuhan University, Wuhan, China
| | - Yong Yang
- SpecAlly Life Technology Co., Ltd., Wuhan, China
| | - Bing Xi
- SpecAlly Life Technology Co., Ltd., Wuhan, China
| | - Junjian Zhang
- Department of Neurology, Zhongnan Hospital, Wuhan University, Wuhan, China
| |
Collapse
|
9
|
Yang Q, Xie Z, Lai B, Cheng G, Liao B, Wan J, Deng M. Identification and verification of atrial fibrillation hub genes caused by primary mitral regurgitation. Medicine (Baltimore) 2023; 102:e35851. [PMID: 37960721 PMCID: PMC10637477 DOI: 10.1097/md.0000000000035851] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Accepted: 10/09/2023] [Indexed: 11/15/2023] Open
Abstract
In the clinic, atrial fibrillation (AF) is a common arrhythmia. Despite constant innovation in treatments for AF, they remain limited by a lack of knowledge of the underlying mechanism responsible for AF. In this study, we examined the molecular mechanisms associated with primary mitral regurgitation (MR) in AF using several bioinformatics techniques. Limma was used to identify differentially expressed genes (DEGs) associated with AF using microarray data from the GSE115574 dataset. WGCNA was used to identify significant module genes. A functional enrichment analysis for overlapping genes between the DEGs and module genes was done and several AF hub genes were identified from a protein-protein interaction (PPI) network. Receiver operating characteristic (ROC) curves were generated to evaluate the validity of the hub genes. We examined 306 DEGs and 147 were upregulated and 159 were downregulated. WGCNA analysis revealed black and ivory modules that contained genes associated with AF. Functional enrichment analysis revealed various biological process terms related to AF. The AUCs for the 8 hub genes screened by the PPI network analysis were > 0.7, indicating satisfactory diagnostic accuracy. The 8 AF-related hub genes included SYT13, VSNL1, GNAO1, RGS4, RALYL, CPLX1, CHGB, and CPLX3. Our findings provide novel insight into the molecular mechanisms of AF and may lead to the development of new treatments.
Collapse
Affiliation(s)
- Qi Yang
- Department of Cardiovascular Surgery, Affiliated Hospital of Southwest Medical University, Jiangyang District, Luzhou, Sichuan Province, China
- Metabolic Vascular Diseases Key Laboratory of Sichuan Province, Luzhou, China
- Key Laboratory of Medical Electrophysiology, Ministry of Education & Medical Electrophysiological Key Laboratory of Sichuan Province, (Collaborative Innovation Center for Prevention of Cardiovascular Diseases), Institute of Cardiovascular Research, Southwest Medical University, Luzhou, China
| | - Zixin Xie
- Department of Cardiovascular Surgery, Affiliated Hospital of Southwest Medical University, Jiangyang District, Luzhou, Sichuan Province, China
- Metabolic Vascular Diseases Key Laboratory of Sichuan Province, Luzhou, China
- Key Laboratory of Medical Electrophysiology, Ministry of Education & Medical Electrophysiological Key Laboratory of Sichuan Province, (Collaborative Innovation Center for Prevention of Cardiovascular Diseases), Institute of Cardiovascular Research, Southwest Medical University, Luzhou, China
| | - Banghui Lai
- Department of Cardiovascular Surgery, Affiliated Hospital of Southwest Medical University, Jiangyang District, Luzhou, Sichuan Province, China
- Metabolic Vascular Diseases Key Laboratory of Sichuan Province, Luzhou, China
- Key Laboratory of Medical Electrophysiology, Ministry of Education & Medical Electrophysiological Key Laboratory of Sichuan Province, (Collaborative Innovation Center for Prevention of Cardiovascular Diseases), Institute of Cardiovascular Research, Southwest Medical University, Luzhou, China
| | - Gang Cheng
- Department of Cardiovascular Surgery, Affiliated Hospital of Southwest Medical University, Jiangyang District, Luzhou, Sichuan Province, China
- Metabolic Vascular Diseases Key Laboratory of Sichuan Province, Luzhou, China
- Key Laboratory of Medical Electrophysiology, Ministry of Education & Medical Electrophysiological Key Laboratory of Sichuan Province, (Collaborative Innovation Center for Prevention of Cardiovascular Diseases), Institute of Cardiovascular Research, Southwest Medical University, Luzhou, China
| | - Bin Liao
- Department of Cardiovascular Surgery, Affiliated Hospital of Southwest Medical University, Jiangyang District, Luzhou, Sichuan Province, China
- Metabolic Vascular Diseases Key Laboratory of Sichuan Province, Luzhou, China
- Key Laboratory of Medical Electrophysiology, Ministry of Education & Medical Electrophysiological Key Laboratory of Sichuan Province, (Collaborative Innovation Center for Prevention of Cardiovascular Diseases), Institute of Cardiovascular Research, Southwest Medical University, Luzhou, China
| | - Juyi Wan
- Department of Cardiovascular Surgery, Affiliated Hospital of Southwest Medical University, Jiangyang District, Luzhou, Sichuan Province, China
- Metabolic Vascular Diseases Key Laboratory of Sichuan Province, Luzhou, China
- Key Laboratory of Medical Electrophysiology, Ministry of Education & Medical Electrophysiological Key Laboratory of Sichuan Province, (Collaborative Innovation Center for Prevention of Cardiovascular Diseases), Institute of Cardiovascular Research, Southwest Medical University, Luzhou, China
| | - Mingbin Deng
- Department of Cardiovascular Surgery, Affiliated Hospital of Southwest Medical University, Jiangyang District, Luzhou, Sichuan Province, China
- Metabolic Vascular Diseases Key Laboratory of Sichuan Province, Luzhou, China
- Key Laboratory of Medical Electrophysiology, Ministry of Education & Medical Electrophysiological Key Laboratory of Sichuan Province, (Collaborative Innovation Center for Prevention of Cardiovascular Diseases), Institute of Cardiovascular Research, Southwest Medical University, Luzhou, China
| |
Collapse
|
10
|
Huang J, Yu Z, Li X, Yang M, Fang Q, Li Z, Wang C, Chen T, Cao X. E3 ligase HECTD3 promotes RNA virus replication and virus-induced inflammation via K33-linked polyubiquitination of PKR. Cell Death Dis 2023; 14:396. [PMID: 37402711 DOI: 10.1038/s41419-023-05923-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Revised: 06/17/2023] [Accepted: 06/23/2023] [Indexed: 07/06/2023]
Abstract
Uncontrolled viral replication and excessive inflammation are the main causes of death in the host infected with virus. Hence inhibition of intracellular viral replication and production of innate cytokines, which are the key strategies of hosts to fight virus infections, need to be finely tuned to eliminate viruses while avoid harmful inflammation. The E3 ligases in regulating virus replication and subsequent innate cytokines production remain to be fully characterized. Here we report that the deficiency of the E3 ubiquitin-protein ligase HECTD3 results in accelerated RNA virus clearance and reduced inflammatory response both in vitro and in vivo. Mechanistically, HECTD3 interacts with dsRNA-dependent protein kinase R (PKR) and mediates Lys33-linkage of PKR, which is the first non-proteolytic ubiquitin modification for PKR. This process disrupts the dimerization and phosphorylation of PKR and subsequent EIF2α activation, which results in the acceleration of virus replication, but promotes the formation of PKR-IKK complex and subsequent inflammatory response. The finding suggests HECTD3 is the potential therapeutic target for simultaneously restraining RNA virus replication and virus-induced inflammation once pharmacologically inhibited.
Collapse
Affiliation(s)
- Jiaying Huang
- Institute of Immunology, Zhejiang University School of Medicine, Hangzhou, 310058, China
| | - Zhou Yu
- National Key Laboratory of Immunity and Inflammation, Suzhou Institute of Systems Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Suzhou, 215123, Jiangsu, China.
- National Key Laboratory of Immunity and Inflammation & Institute of Immunology, Navy Medical University, Shanghai, 200433, China.
| | - Xuelian Li
- National Key Laboratory of Immunity and Inflammation, Suzhou Institute of Systems Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Suzhou, 215123, Jiangsu, China
| | - Mingjin Yang
- National Key Laboratory of Immunity and Inflammation & Institute of Immunology, Navy Medical University, Shanghai, 200433, China
| | - Qian Fang
- National Key Laboratory of Immunity and Inflammation & Institute of Immunology, Navy Medical University, Shanghai, 200433, China
| | - Zheng Li
- National Key Laboratory of Immunity and Inflammation & Institute of Immunology, Navy Medical University, Shanghai, 200433, China
| | - Chunmei Wang
- National Key Laboratory of Immunity and Inflammation, Suzhou Institute of Systems Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Suzhou, 215123, Jiangsu, China
| | - Taoyong Chen
- National Key Laboratory of Immunity and Inflammation & Institute of Immunology, Navy Medical University, Shanghai, 200433, China
| | - Xuetao Cao
- Institute of Immunology, Zhejiang University School of Medicine, Hangzhou, 310058, China.
- National Key Laboratory of Immunity and Inflammation, Suzhou Institute of Systems Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Suzhou, 215123, Jiangsu, China.
- National Key Laboratory of Immunity and Inflammation & Institute of Immunology, Navy Medical University, Shanghai, 200433, China.
- Institute of Immunology, College of Life Science, Nankai University, Tianjin, 300071, China.
| |
Collapse
|
11
|
Xiao J, Sun F, Wang YN, Liu B, Zhou P, Wang FX, Zhou HF, Ge Y, Yue TT, Luo JH, Yang CL, Rong SJ, Xiong ZZ, Ma S, Zhang Q, Xun Y, Yang CG, Luan Y, Wang SG, Wang CY, Wang ZH. UBC9 deficiency enhances immunostimulatory macrophage activation and subsequent antitumor T cell response in prostate cancer. J Clin Invest 2023; 133:158352. [PMID: 36626227 PMCID: PMC9927932 DOI: 10.1172/jci158352] [Citation(s) in RCA: 33] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Accepted: 01/05/2023] [Indexed: 01/11/2023] Open
Abstract
The role of tumor-associated macrophages (TAMs), along with the regulatory mechanisms underlying distinct macrophage activation states, remains poorly understood in prostate cancer (PCa). Herein, we report that PCa growth in mice with macrophage-specific Ubc9 deficiency is substantially suppressed compared with that in wild-type littermates, an effect partially ascribed to the augmented CD8+ T cell response. Biochemical and molecular analyses revealed that signal transducer and activator of transcription 4 (STAT4) is a crucial UBC9-mediated SUMOylation target, with lysine residue 350 (K350) as the major modification site. Site-directed mutation of STAT4 (K350R) enhanced its nuclear translocation and stability, thereby facilitating the proinflammatory activation of macrophages. Importantly, administration of the UBC9 inhibitor 2-D08 promoted the antitumor effect of TAMs and increased the expression of PD-1 on CD8+ T cells, supporting a synergistic antitumor efficacy once it combined with the immune checkpoint blockade therapy. Together, our results demonstrate that ablation of UBC9 could reverse the immunosuppressive phenotype of TAMs by promoting STAT4-mediated macrophage activation and macrophage-CD8+ T cell crosstalk, which provides valuable insights to halt the pathogenic process of tumorigenesis.
Collapse
Affiliation(s)
- Jun Xiao
- Department of Urology;,Department of Thyroid and Breast Surgery;,Department of Respiratory and Critical Care Medicine, Center for Biomedical Research, NHC Key Laboratory of Respiratory Diseases; and
| | - Fei Sun
- Department of Respiratory and Critical Care Medicine, Center for Biomedical Research, NHC Key Laboratory of Respiratory Diseases; and
| | - Ya-Nan Wang
- Department of Urology;,Department of Respiratory and Critical Care Medicine, Center for Biomedical Research, NHC Key Laboratory of Respiratory Diseases; and
| | - Bo Liu
- Department of Oncology; Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | | | - Fa-Xi Wang
- Department of Respiratory and Critical Care Medicine, Center for Biomedical Research, NHC Key Laboratory of Respiratory Diseases; and
| | - Hai-Feng Zhou
- Department of Respiratory and Critical Care Medicine, Center for Biomedical Research, NHC Key Laboratory of Respiratory Diseases; and,Department of Integrated Traditional Chinese and Western Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | | | - Tian-Tian Yue
- Department of Respiratory and Critical Care Medicine, Center for Biomedical Research, NHC Key Laboratory of Respiratory Diseases; and
| | - Jia-Hui Luo
- Department of Respiratory and Critical Care Medicine, Center for Biomedical Research, NHC Key Laboratory of Respiratory Diseases; and
| | - Chun-Liang Yang
- Department of Respiratory and Critical Care Medicine, Center for Biomedical Research, NHC Key Laboratory of Respiratory Diseases; and
| | - Shan-Jie Rong
- Department of Respiratory and Critical Care Medicine, Center for Biomedical Research, NHC Key Laboratory of Respiratory Diseases; and
| | | | | | - Qi Zhang
- Department of Critical Care Medicine, Zhongnan Hospital, Wuhan University, Wuhan, China
| | | | | | | | | | - Cong-Yi Wang
- Department of Respiratory and Critical Care Medicine, Center for Biomedical Research, NHC Key Laboratory of Respiratory Diseases; and
| | | |
Collapse
|
12
|
SUMOylation targeting mitophagy in cardiovascular diseases. J Mol Med (Berl) 2022; 100:1511-1538. [PMID: 36163375 DOI: 10.1007/s00109-022-02258-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Revised: 09/13/2022] [Accepted: 09/14/2022] [Indexed: 12/14/2022]
Abstract
Small ubiquitin-like modifier (SUMO) plays a key regulatory role in cardiovascular diseases, such as cardiac hypertrophy, hypertension, atherosclerosis, and cardiac ischemia-reperfusion injury. As a multifunctional posttranslational modification molecule in eukaryotic cells, SUMOylation is essentially associated with the regulation of mitochondrial dynamics, especially mitophagy, which is involved in the progression and development of cardiovascular diseases. SUMOylation targeting mitochondrial-associated proteins is admittedly considered to regulate mitophagy activation and mitochondrial functions and dynamics, including mitochondrial fusion and fission. SUMOylation triggers mitochondrial fusion to promote mitochondrial dysfunction by modifying Fis1, OPA1, MFN1/2, and DRP1. The interaction between SUMO and DRP1 induces SUMOylation and inhibits lysosomal degradation of DRP1, which is further involved in the regulation of mitochondrial fission. Both SUMOylation and deSUMOylation contribute to the initiation and activation of mitophagy by regulating the conjugation of MFN1/2 SERCA2a, HIF1α, and PINK1. SUMOylation mediated by the SUMO molecule has attracted much attention due to its dual roles in the development of cardiovascular diseases. In this review, we systemically summarize the current understanding underlying the expression, regulation, and structure of SUMO molecules; explore the biochemical functions of SUMOylation in the initiation and activation of mitophagy; discuss the biological roles and mechanisms of SUMOylation in cardiovascular diseases; and further provide a wider explanation of SUMOylation and deSUMOylation research to provide a possible therapeutic strategy for cardiovascular diseases. Considering the precise functions and exact mechanisms of SUMOylation in mitochondrial dysfunction and mitophagy will provide evidence for future experimental research and may serve as an effective approach in the development of novel therapeutic strategies for cardiovascular diseases. Regulation and effect of SUMOylation in cardiovascular diseases via mitophagy. SUMOylation is involved in multiple cardiovascular diseases, including cardiac hypertrophy, hypertension, atherosclerosis, and cardiac ischemia-reperfusion injury. Since it is expressed in multiple cells associated with cardiovascular disease, SUMOylation can be regulated by numerous ligases, including the SENP family proteins PIAS1, PIASy/4, UBC9, and MAPL. SUMOylation regulates the activation and degradation of PINK1, SERCA2a, PPARγ, ERK5, and DRP1 to mediate mitochondrial dynamics, especially mitophagy activation. Mitophagy activation regulated by SUMOylation further promotes or inhibits ventricular diastolic dysfunction, perfusion injury, ventricular remodelling and ventricular noncompaction, which contribute to the development of cardiovascular diseases.
Collapse
|
13
|
Li S, Liang Y, Zou J, Cai Z, Yang H, Yang J, Zhang Y, Lin H, Zhang G, Tan M. SUMOylation of microtubule-cleaving enzyme KATNA1 promotes microtubule severing and neurite outgrowth. J Biol Chem 2022; 298:102292. [PMID: 35868557 PMCID: PMC9403493 DOI: 10.1016/j.jbc.2022.102292] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Revised: 07/10/2022] [Accepted: 07/11/2022] [Indexed: 12/01/2022] Open
Abstract
Katanin p60 ATPase-containing subunit A1 (KATNA1) is a microtubule-cleaving enzyme that regulates the development of neural protrusions through cytoskeletal rearrangements. However, the mechanism underlying the linkage of the small ubiquitin-like modifier (SUMO) protein to KATNA1 and how this modification regulates the development of neural protrusions is unclear. Here we discovered, using mass spectrometry analysis, that SUMO-conjugating enzyme UBC9, an enzyme necessary for the SUMOylation process, was present in the KATNA1 interactome. Moreover, GST-pull down and co-immunoprecipitation assays confirmed that KATNA1 and SUMO interact. We further demonstrated using immunofluorescence experiments that KATNA1 and the SUMO2 isoform colocalized in hippocampal neurites. We also performed a bioinformatics analysis of KATNA1 protein sequences to identify three potentially conserved SUMOylation sites (K77, K157, and K330) among vertebrates. Mutation of K330, but not K77 or K157, abolished KATNA1-induced microtubule severing and decreased the level of binding observed for KATNA1 and SUMO2. Cotransfection of SUMO2 and wildtype KATNA1 in COS7 cells increased microtubule severing, whereas no effect was observed after cotransfection with the K330R KATNA1 mutant. Furthermore, in cultured hippocampal neurons, overexpression of wildtype KATNA1 significantly promoted neurite outgrowth, whereas the K330R mutant eliminated this effect. Taken together, our results demonstrate that the K330 site in KATNA1 is modified by SUMOylation and SUMOylation of KATNA1 promotes microtubule dynamics and hippocampal neurite outgrowth.
Collapse
Affiliation(s)
- Shaojin Li
- Department of Orthopaedics, the First Affiliated Hospital of Jinan University, Guangzhou 510630, China
| | - Yaozhong Liang
- Department of Orthopaedics, the First Affiliated Hospital of Jinan University, Guangzhou 510630, China
| | - Jianyu Zou
- Department of Orthopaedics, the First Affiliated Hospital of Jinan University, Guangzhou 510630, China
| | - Zhenbin Cai
- Department of Orthopaedics, the First Affiliated Hospital of Jinan University, Guangzhou 510630, China
| | - Hua Yang
- Department of Orthopaedics, the First Affiliated Hospital of Jinan University, Guangzhou 510630, China
| | - Jie Yang
- Department of Orthopaedics, the First Affiliated Hospital of Jinan University, Guangzhou 510630, China
| | - Yunlong Zhang
- Department of Orthopaedics, the First Affiliated Hospital of Jinan University, Guangzhou 510630, China
| | - Hongsheng Lin
- Department of Orthopaedics, the First Affiliated Hospital of Jinan University, Guangzhou 510630, China.
| | - Guowei Zhang
- Department of Orthopaedics, the First Affiliated Hospital of Jinan University, Guangzhou 510630, China.
| | - Minghui Tan
- Department of Orthopaedics, the First Affiliated Hospital of Jinan University, Guangzhou 510630, China.
| |
Collapse
|
14
|
Gao A, Zou J, Mao Z, Zhou H, Zeng G. SUMO2-mediated SUMOylation of SH3GLB1 promotes ionizing radiation-induced hypertrophic cardiomyopathy through mitophagy activation. Eur J Pharmacol 2022; 924:174980. [PMID: 35487252 DOI: 10.1016/j.ejphar.2022.174980] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2021] [Revised: 04/05/2022] [Accepted: 04/22/2022] [Indexed: 12/25/2022]
Abstract
Hypertrophic cardiomyopathy (HC) is characterized by the enlargement of individual cardiomyocytes, which is a typical pathophysiological process that occurs in various cardiovascular diseases. Ionizing radiation (IR) is an important independent risk factor for hypertrophic cardiomyopathy, but the underlying molecular mechanism is still unclear. In the present study, we aimed to clarify the role of IR in promoting cardiac hypertrophy and investigate the mechanism by which the SUMO2-mediated SUMOylation of SH3GLB1 affects mitophagy in IR-induced cardiac hypertrophy. In vivo, IR promoted cardiac hypertrophy by activating mitophagy. In vitro, IR upregulated PINK1 and Parkin protein expression and damaged mitochondrial morphological structure. We further demonstrated that SH3GLB1 deficiency inhibited mitophagy activation and restored mitochondrial cristae, revealing a regulatory role of SH3GLB1 in cardiac hypertrophy. IR promoted interactions between SH3GLB1 and mitochondrial membrane proteins, such as MFN1/2, TOM20 and Drp1, further indicating that the mechanism by which SH3GLB1 functions in cardiac hypertrophy might involve mitophagy. A bioinformatics prediction found that SUMO2 could SUMOylate SH3GLB1 at position K82. Consistent with this finding, both co-IP assays and laser confocal microscopy showed that IR promoted the interaction and colocalization of SUMO2 and SH3GLB1. In summary, our study identifies IR as an important factor that promotes hypertrophic cardiomyopathy by accelerating the activation of mitophagy through the SUMO2-mediated SUMOylation of SH3GLB1; thus, IR exerts dual therapeutic effects in the treatment of thoracic tumours with long-term radiotherapy. Additionally, this study provides novel treatment strategies and targets for preventing the hypertrophic cardiomyopathy caused by thoracic tumour radiotherapy. Furthermore, SH3GLB1 may be a promising experimental target for the development of strategies for treating cardiovascular diseases caused by IR.
Collapse
Affiliation(s)
- Anbo Gao
- Clinical Research Institute, The Second Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, 421002, Hunan, China; Department of Cardiovascular Medicine, The Second Affiliated Hospital, Hengyang Medical School, University of South China, Key Laboratory of Heart Failure Prevention & Treatment of Hengyang, Clinical Medicine Research Center of Arteriosclerotic Disease of Hunan Province, Hengyang, 421002, Hunan, China
| | - Jin Zou
- Department of Cardiovascular Medicine, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, 421002, Hunan, China
| | - Zhenjiang Mao
- Department of Gastroenterology, The Second Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, 421002, Hunan, China
| | - Hong Zhou
- Department of Radiology, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, 421002, Hunan, China.
| | - Gaofeng Zeng
- Department of Cardiovascular Medicine, The Second Affiliated Hospital, Hengyang Medical School, University of South China, Key Laboratory of Heart Failure Prevention & Treatment of Hengyang, Clinical Medicine Research Center of Arteriosclerotic Disease of Hunan Province, Hengyang, 421002, Hunan, China.
| |
Collapse
|
15
|
Nath M, Romaine SP, Koekemoer A, Hamby S, Webb TR, Nelson CP, Castellanos‐Uribe M, Papakonstantinou M, Anker SD, Lang CC, Metra M, Zannad F, Filippatos G, van Veldhuisen DJ, Cleland JG, Ng LL, May ST, Marelli‐Berg F, Voors AA, Timmons JA, Samani NJ. Whole blood transcriptomic profiling identifies molecular pathways related to cardiovascular mortality in heart failure. Eur J Heart Fail 2022; 24:1009-1019. [PMID: 35570197 PMCID: PMC9546237 DOI: 10.1002/ejhf.2540] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/24/2021] [Revised: 03/08/2022] [Accepted: 03/18/2022] [Indexed: 12/01/2022] Open
Abstract
AIMS Chronic heart failure (CHF) is a systemic syndrome with a poor prognosis and a need for novel therapies. We investigated whether whole blood transcriptomic profiling can provide new mechanistic insights into cardiovascular (CV) mortality in CHF. METHODS AND RESULTS Transcriptome profiles were generated at baseline from 944 CHF patients from the BIOSTAT-CHF study, of whom 626 survived and 318 died from a CV cause during a follow-up of 21 months. Multivariable analysis, including adjustment for cell count, identified 1153 genes (6.5%) that were differentially expressed between those that survived or died and strongly related to a validated clinical risk score for adverse prognosis. The differentially expressed genes mainly belonged to five non-redundant pathways: adaptive immune response, proteasome-mediated ubiquitin-dependent protein catabolic process, T-cell co-stimulation, positive regulation of T-cell proliferation, and erythrocyte development. These five pathways were selectively related (RV coefficients >0.20) with seven circulating protein biomarkers of CV mortality (fibroblast growth factor 23, soluble ST2, adrenomedullin, hepcidin, pentraxin-3, WAP 4-disulfide core domain 2, and interleukin-6) revealing an intricate relationship between immune and iron homeostasis. The pattern of survival-associated gene expression matched with 29 perturbagen-induced transcriptome signatures in the iLINCS drug-repurposing database, identifying drugs, approved for other clinical indications, that were able to reverse in vitro the molecular changes associated with adverse prognosis in CHF. CONCLUSION Systematic modelling of the whole blood protein-coding transcriptome defined molecular pathways that provide a link between clinical risk factors and adverse CV prognosis in CHF, identifying both established and new potential therapeutic targets.
Collapse
Affiliation(s)
- Mintu Nath
- Department of Cardiovascular SciencesUniversity of Leicester and NIHR Leicester Biomedical Research CentreGlenfield Hospital, LeicesterUK
- Institute of Applied Health SciencesUniversity of AberdeenAberdeenUK
| | - Simon P.R. Romaine
- Department of Cardiovascular SciencesUniversity of Leicester and NIHR Leicester Biomedical Research CentreGlenfield Hospital, LeicesterUK
| | - Andrea Koekemoer
- Department of Cardiovascular SciencesUniversity of Leicester and NIHR Leicester Biomedical Research CentreGlenfield Hospital, LeicesterUK
| | - Stephen Hamby
- Department of Cardiovascular SciencesUniversity of Leicester and NIHR Leicester Biomedical Research CentreGlenfield Hospital, LeicesterUK
| | - Thomas R. Webb
- Department of Cardiovascular SciencesUniversity of Leicester and NIHR Leicester Biomedical Research CentreGlenfield Hospital, LeicesterUK
| | - Christopher P. Nelson
- Department of Cardiovascular SciencesUniversity of Leicester and NIHR Leicester Biomedical Research CentreGlenfield Hospital, LeicesterUK
| | | | - Manolo Papakonstantinou
- Department of Cardiovascular SciencesUniversity of Leicester and NIHR Leicester Biomedical Research CentreGlenfield Hospital, LeicesterUK
| | - Stefan D. Anker
- German Centre for Cardiovascular Research (DZHK) Partner Site Berlin, Charité – Universitätsmedizin BerlinBerlinGermany
| | - Chim C. Lang
- Division of Molecular and Clinical Medicine, School of MedicineUniversity of DundeeDundeeUK
| | - Marco Metra
- Department of Medical and Surgical Specialties, Radiological Sciences and Public HealthUniversity of BresciaBresciaItaly
| | - Faiez Zannad
- Clinical Investigation Center 1433, Centre Hospitalier Regional et Universitaire de NancyVandoeuvre les NancyFrance
| | | | - Dirk J. van Veldhuisen
- Department of Cardiology, University of GroningenUniversity Medical Center GroningenGroningenThe Netherlands
| | - John G. Cleland
- National Heart and Lung Institute, Royal Brompton and Harefield Hospitals, Imperial College, London, UK and Robertson Centre for Biostatistics and Clinical TrialsUniversity of GlasgowGlasgowUK
| | - Leong L. Ng
- Department of Cardiovascular SciencesUniversity of Leicester and NIHR Leicester Biomedical Research CentreGlenfield Hospital, LeicesterUK
| | - Sean T. May
- School of BiosciencesUniversity of Nottingham, Sutton Bonington CampusLoughboroughUK
| | | | - Adriaan A. Voors
- Department of Cardiology, University of GroningenUniversity Medical Center GroningenGroningenThe Netherlands
| | - James A. Timmons
- Barts & The London School of MedicineQueen Mary University of LondonLondonUK
- Augur Precision Medicine LtdStirling University Innovation ParkUK
| | - Nilesh J. Samani
- Department of Cardiovascular SciencesUniversity of Leicester and NIHR Leicester Biomedical Research CentreGlenfield Hospital, LeicesterUK
| |
Collapse
|
16
|
Deng Y, Yu C, Chen L, Zhang X, Lei Q, Liu Q, Cai G, Liu F. ARV-771 Acts as an Inducer of Cell Cycle Arrest and Apoptosis to Suppress Hepatocellular Carcinoma Progression. Front Pharmacol 2022; 13:858901. [PMID: 35600879 PMCID: PMC9114478 DOI: 10.3389/fphar.2022.858901] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Accepted: 04/19/2022] [Indexed: 11/13/2022] Open
Abstract
Hepatocellular carcinoma (HCC) is the most commonly diagnosed liver cancer with limited treatment options and extremely poor prognosis worldwide. Recently, the proteolysis targeting chimeras (PROTACs), which aim to induce proteasome-mediated degradation of interesting proteins via recruiting E3 ligases, have become the advanced tools and attractive molecules for cancer treatment. However, the anticancer effects of PROTACs in HCC remain to be clarified. Here, we evaluate the anticancer activity of ARV-771, a previously reported PROTAC compound designed for bromodomain and extra-terminal domain (BET) proteins, in HCC. We show that ARV-771 suppresses the cell viability and colony formation of HCC cells via arresting cell cycle progression and triggering apoptosis. Further investigations reveal that ARV-771 notably downregulates multiple non-proteasomal deubiquitinases which are critical to the development of cancers. Additionally, HCC cells can decrease their sensitivity to ARV-771 via activating the MEK/ERK and p38 MAPKs. ARV-771 also inhibits HCC progression in vivo. Moreover, we show that ARV-771 and sorafenib, a Raf inhibitor that clinically used for targeted therapy of liver cancer, can synergistically inhibit the growth of HCC cells. Overall, this study not only explores the anticancer activity of ARV-771 and its underlying mechanisms in HCC, but also deepens our understanding of deubiquitinases, MAPKs, cell cycle, and apoptosis induction in cancer therapy.
Collapse
Affiliation(s)
- Yuanfei Deng
- Department of Pathology, The First People’s Hospital of Foshan, Foshan, China
- *Correspondence: Yuanfei Deng, ; Gengxi Cai, ; Fang Liu,
| | - Cuifu Yu
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Degradation, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, China
| | - Lushi Chen
- Health Management Center, The First People’s Hospital of Foshan, Foshan, China
| | - Xin Zhang
- Department of Pathology, The First People’s Hospital of Foshan, Foshan, China
| | - Qiucheng Lei
- Department of Hepatopancreatic Surgery, The First People’s Hospital of Foshan, Foshan, China
| | - Qing Liu
- Department of Pathology, The First People’s Hospital of Foshan, Foshan, China
| | - Gengxi Cai
- Department of Breast Surgery, The First People’s Hospital of Foshan, Foshan, China
- *Correspondence: Yuanfei Deng, ; Gengxi Cai, ; Fang Liu,
| | - Fang Liu
- Department of Pathology, The First People’s Hospital of Foshan, Foshan, China
- *Correspondence: Yuanfei Deng, ; Gengxi Cai, ; Fang Liu,
| |
Collapse
|
17
|
Pan Z, Zhao R, Li B, Qi Y, Qiu W, Guo Q, Zhang S, Zhao S, Xu H, Li M, Gao Z, Fan Y, Xu J, Wang H, Wang S, Qiu J, Wang Q, Guo X, Deng L, Zhang P, Xue H, Li G. EWSR1-induced circNEIL3 promotes glioma progression and exosome-mediated macrophage immunosuppressive polarization via stabilizing IGF2BP3. Mol Cancer 2022; 21:16. [PMID: 35031058 PMCID: PMC8759291 DOI: 10.1186/s12943-021-01485-6] [Citation(s) in RCA: 191] [Impact Index Per Article: 63.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Accepted: 12/20/2021] [Indexed: 12/13/2022] Open
Abstract
Background Gliomas are the most common malignant primary brain tumours with a highly immunosuppressive tumour microenvironment (TME) and poor prognosis. Circular RNAs (circRNA), a newly found type of endogenous noncoding RNA, characterized by high stability, abundance, conservation, have been shown to play an important role in the pathophysiological processes and TME remodelling of various tumours. Methods CircRNA sequencing analysis was performed to explore circRNA expression profiles in normal and glioma tissues. The biological function of a novel circRNA, namely, circNEIL3, in glioma development was confirmed both in vitro and in vivo. Mechanistically, RNA pull-down, mass spectrum, RNA immunoprecipitation (RIP), luciferase reporter, and co-immunoprecipitation assays were conducted. Results We identified circNEIL3, which could be cyclized by EWS RNA-binding protein 1(EWSR1), to be upregulated in glioma tissues and to correlate positively with glioma malignant progression. Functionally, we confirmed that circNEIL3 promotes tumorigenesis and carcinogenic progression of glioma in vitro and in vivo. Mechanistically, circNEIL3 stabilizes IGF2BP3 (insulin-like growth factor 2 mRNA binding protein 3) protein, a known oncogenic protein, by preventing HECTD4-mediated ubiquitination. Moreover, circNEIL3 overexpression glioma cells drives macrophage infiltration into the tumour microenvironment (TME). Finally, circNEIL3 is packaged into exosomes by hnRNPA2B1 and transmitted to infiltrated tumour associated macrophages (TAMs), enabling them to acquire immunosuppressive properties by stabilizing IGF2BP3 and in turn promoting glioma progression. Conclusions This work reveals that circNEIL3 plays a nonnegligible multifaceted role in promoting gliomagenesis, malignant progression and macrophage tumour-promoting phenotypes polarization, highlighting that circNEIL3 is a potential prognostic biomarker and therapeutic target in glioma. Supplementary Information The online version contains supplementary material available at 10.1186/s12943-021-01485-6.
Collapse
Affiliation(s)
- Ziwen Pan
- Department of Neurosurgery, Qilu Hospital, Cheeloo College of Medicine and Institute of Brain and Brain-Inspired Science, Shandong University, 107 Wenhua Western Road; Jinan, Shandong 250012, China, Jinan, 250012, Shandong, China.,Shandong Key Laboratory of Brain Function Remodeling, Jinan, Shandong, China
| | - Rongrong Zhao
- Department of Neurosurgery, Qilu Hospital, Cheeloo College of Medicine and Institute of Brain and Brain-Inspired Science, Shandong University, 107 Wenhua Western Road; Jinan, Shandong 250012, China, Jinan, 250012, Shandong, China.,Shandong Key Laboratory of Brain Function Remodeling, Jinan, Shandong, China
| | - Boyan Li
- Department of Neurosurgery, Qilu Hospital, Cheeloo College of Medicine and Institute of Brain and Brain-Inspired Science, Shandong University, 107 Wenhua Western Road; Jinan, Shandong 250012, China, Jinan, 250012, Shandong, China.,Shandong Key Laboratory of Brain Function Remodeling, Jinan, Shandong, China
| | - Yanhua Qi
- Department of Neurosurgery, Qilu Hospital, Cheeloo College of Medicine and Institute of Brain and Brain-Inspired Science, Shandong University, 107 Wenhua Western Road; Jinan, Shandong 250012, China, Jinan, 250012, Shandong, China.,Shandong Key Laboratory of Brain Function Remodeling, Jinan, Shandong, China
| | - Wei Qiu
- Department of Neurosurgery, Qilu Hospital, Cheeloo College of Medicine and Institute of Brain and Brain-Inspired Science, Shandong University, 107 Wenhua Western Road; Jinan, Shandong 250012, China, Jinan, 250012, Shandong, China.,Shandong Key Laboratory of Brain Function Remodeling, Jinan, Shandong, China
| | - Qindong Guo
- Department of Neurosurgery, Qilu Hospital, Cheeloo College of Medicine and Institute of Brain and Brain-Inspired Science, Shandong University, 107 Wenhua Western Road; Jinan, Shandong 250012, China, Jinan, 250012, Shandong, China.,Shandong Key Laboratory of Brain Function Remodeling, Jinan, Shandong, China
| | - Shouji Zhang
- Department of Neurosurgery, Qilu Hospital, Cheeloo College of Medicine and Institute of Brain and Brain-Inspired Science, Shandong University, 107 Wenhua Western Road; Jinan, Shandong 250012, China, Jinan, 250012, Shandong, China.,Shandong Key Laboratory of Brain Function Remodeling, Jinan, Shandong, China
| | - Shulin Zhao
- Department of Neurosurgery, Qilu Hospital, Cheeloo College of Medicine and Institute of Brain and Brain-Inspired Science, Shandong University, 107 Wenhua Western Road; Jinan, Shandong 250012, China, Jinan, 250012, Shandong, China.,Shandong Key Laboratory of Brain Function Remodeling, Jinan, Shandong, China
| | - Hao Xu
- Shandong Key Laboratory of Brain Function Remodeling, Jinan, Shandong, China.,Department of Neurosurgery, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, Shandong, China
| | - Ming Li
- Shandong Key Laboratory of Brain Function Remodeling, Jinan, Shandong, China.,Department of Neurosurgery, Taian Central Hospital, Taian, Shandong, China
| | - Zijie Gao
- Department of Neurosurgery, Qilu Hospital, Cheeloo College of Medicine and Institute of Brain and Brain-Inspired Science, Shandong University, 107 Wenhua Western Road; Jinan, Shandong 250012, China, Jinan, 250012, Shandong, China.,Shandong Key Laboratory of Brain Function Remodeling, Jinan, Shandong, China
| | - Yang Fan
- Department of Neurosurgery, Qilu Hospital, Cheeloo College of Medicine and Institute of Brain and Brain-Inspired Science, Shandong University, 107 Wenhua Western Road; Jinan, Shandong 250012, China, Jinan, 250012, Shandong, China.,Shandong Key Laboratory of Brain Function Remodeling, Jinan, Shandong, China
| | - Jianye Xu
- Department of Neurosurgery, Qilu Hospital, Cheeloo College of Medicine and Institute of Brain and Brain-Inspired Science, Shandong University, 107 Wenhua Western Road; Jinan, Shandong 250012, China, Jinan, 250012, Shandong, China.,Shandong Key Laboratory of Brain Function Remodeling, Jinan, Shandong, China
| | - Huizhi Wang
- Department of Neurosurgery, Qilu Hospital, Cheeloo College of Medicine and Institute of Brain and Brain-Inspired Science, Shandong University, 107 Wenhua Western Road; Jinan, Shandong 250012, China, Jinan, 250012, Shandong, China.,Shandong Key Laboratory of Brain Function Remodeling, Jinan, Shandong, China
| | - Shaobo Wang
- Department of Neurosurgery, Qilu Hospital, Cheeloo College of Medicine and Institute of Brain and Brain-Inspired Science, Shandong University, 107 Wenhua Western Road; Jinan, Shandong 250012, China, Jinan, 250012, Shandong, China.,Shandong Key Laboratory of Brain Function Remodeling, Jinan, Shandong, China
| | - Jiawei Qiu
- Department of Neurosurgery, Qilu Hospital, Cheeloo College of Medicine and Institute of Brain and Brain-Inspired Science, Shandong University, 107 Wenhua Western Road; Jinan, Shandong 250012, China, Jinan, 250012, Shandong, China.,Shandong Key Laboratory of Brain Function Remodeling, Jinan, Shandong, China
| | - Qingtong Wang
- Department of Neurosurgery, Qilu Hospital, Cheeloo College of Medicine and Institute of Brain and Brain-Inspired Science, Shandong University, 107 Wenhua Western Road; Jinan, Shandong 250012, China, Jinan, 250012, Shandong, China.,Shandong Key Laboratory of Brain Function Remodeling, Jinan, Shandong, China
| | - Xing Guo
- Department of Neurosurgery, Qilu Hospital, Cheeloo College of Medicine and Institute of Brain and Brain-Inspired Science, Shandong University, 107 Wenhua Western Road; Jinan, Shandong 250012, China, Jinan, 250012, Shandong, China.,Shandong Key Laboratory of Brain Function Remodeling, Jinan, Shandong, China
| | - Lin Deng
- Department of Neurosurgery, Qilu Hospital, Cheeloo College of Medicine and Institute of Brain and Brain-Inspired Science, Shandong University, 107 Wenhua Western Road; Jinan, Shandong 250012, China, Jinan, 250012, Shandong, China.,Shandong Key Laboratory of Brain Function Remodeling, Jinan, Shandong, China
| | - Ping Zhang
- Department of Neurosurgery, Qilu Hospital, Cheeloo College of Medicine and Institute of Brain and Brain-Inspired Science, Shandong University, 107 Wenhua Western Road; Jinan, Shandong 250012, China, Jinan, 250012, Shandong, China.,Shandong Key Laboratory of Brain Function Remodeling, Jinan, Shandong, China
| | - Hao Xue
- Department of Neurosurgery, Qilu Hospital, Cheeloo College of Medicine and Institute of Brain and Brain-Inspired Science, Shandong University, 107 Wenhua Western Road; Jinan, Shandong 250012, China, Jinan, 250012, Shandong, China. .,Shandong Key Laboratory of Brain Function Remodeling, Jinan, Shandong, China.
| | - Gang Li
- Department of Neurosurgery, Qilu Hospital, Cheeloo College of Medicine and Institute of Brain and Brain-Inspired Science, Shandong University, 107 Wenhua Western Road; Jinan, Shandong 250012, China, Jinan, 250012, Shandong, China. .,Shandong Key Laboratory of Brain Function Remodeling, Jinan, Shandong, China.
| |
Collapse
|
18
|
The Role of HECT-Type E3 Ligase in the Development of Cardiac Disease. Int J Mol Sci 2021; 22:ijms22116065. [PMID: 34199773 PMCID: PMC8199989 DOI: 10.3390/ijms22116065] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Revised: 05/26/2021] [Accepted: 06/01/2021] [Indexed: 12/12/2022] Open
Abstract
Despite advances in medicine, cardiac disease remains an increasing health problem associated with a high mortality rate. Maladaptive cardiac remodeling, such as cardiac hypertrophy and fibrosis, is a risk factor for heart failure; therefore, it is critical to identify new therapeutic targets. Failing heart is reported to be associated with hyper-ubiquitylation and impairment of the ubiquitin–proteasome system, indicating an importance of ubiquitylation in the development of cardiac disease. Ubiquitylation is a post-translational modification that plays a pivotal role in protein function and degradation. In 1995, homologous to E6AP C-terminus (HECT) type E3 ligases were discovered. E3 ligases are key enzymes in ubiquitylation and are classified into three families: really interesting new genes (RING), HECT, and RING-between-RINGs (RBRs). Moreover, 28 HECT-type E3 ligases have been identified in human beings. It is well conserved in evolution and is characterized by the direct attachment of ubiquitin to substrates. HECT-type E3 ligase is reported to be involved in a wide range of human diseases and health. The role of HECT-type E3 ligases in the development of cardiac diseases has been uncovered in the last decade. There are only a few review articles summarizing recent advancements regarding HECT-type E3 ligase in the field of cardiac disease. This study focused on cardiac remodeling and described the role of HECT-type E3 ligases in the development of cardiac disease. Moreover, this study revealed that the current knowledge could be exploited for the development of new clinical therapies.
Collapse
|
19
|
Zhu Y, Zhang C, Huang M, Lin J, Fan X, Ni T. TRIM26 Induces Ferroptosis to Inhibit Hepatic Stellate Cell Activation and Mitigate Liver Fibrosis Through Mediating SLC7A11 Ubiquitination. Front Cell Dev Biol 2021; 9:644901. [PMID: 33869196 PMCID: PMC8044755 DOI: 10.3389/fcell.2021.644901] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Accepted: 02/16/2021] [Indexed: 12/12/2022] Open
Abstract
Hepatic stellate cells (HSCs) are activated by inflammatory mediators to secrete extracellular matrix for collagen deposition, leading to liver fibrosis. Ferroptosis is iron- and lipid hydroperoxide-dependent programmed cell death, which has recently been targeted for inhibiting liver fibrogenic processes. Tripartite motif-containing protein 26 (TRIM26) is an E3 ubiquitin ligase that functions as a tumor suppressor in hepatocellular carcinoma, while little is known about its function in liver fibrosis. In the present study, the differential expression of TRIM26 in normal and fibrotic liver tissues was examined based on both online databases and specimens collected from patient cohort. The effects of TRIM26 on HSCs ferroptosis were examined in vitro through evaluating cell proliferation, lipid peroxidation, and expression of key ferroptosis-related factors. In vivo function of TRIM26 in liver fibrosis was examined based on CCl4-induced mice model. We found that TRIM26 was downregulated in fibrotic liver tissues. The overexpression of TRIM26 inhibited HSCs proliferation, promoted lipid peroxidation, manipulated ferroptosis-related factor expressions, and counteracted the effect of iron inhibitor deferoxamine. Moreover, TRIM26 physically interacted with solute carrier family-7 member-11 (SLC7A11), a critical protein for lipid reactive oxygen species (ROS) scavenging, and mediated its ubiquitination. In addition, TRIM26 overexpression induced HSCs ferroptosis and mitigated CCl4-induced liver fibrosis in mice. In conclusion, TRIM26 promotes HSCs ferroptosis to suppress liver fibrosis through mediating the ubiquitination of SLC7A11. The TRIM26-targeted SLC7A11 suppression can be a novel therapeutic strategy for liver fibrosis.
Collapse
Affiliation(s)
- Yiming Zhu
- Department of General Surgery, Shanghai Ninth People's Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Chihao Zhang
- Department of General Surgery, Shanghai Ninth People's Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Mingzhe Huang
- Department of General Surgery, Shanghai Ninth People's Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Jiayun Lin
- Department of General Surgery, Shanghai Ninth People's Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Xiao Fan
- Department of General Surgery, Shanghai Ninth People's Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Tao Ni
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|