1
|
Huang R, Cui H, Yahya Ali Alshami MA, Fu C, Jiang W, Cai M, Zhou S, Zhu X, Hu C. LOX-1 rewires glutamine ammonia metabolism to drive liver fibrosis. Mol Metab 2025; 96:102132. [PMID: 40180177 PMCID: PMC12004974 DOI: 10.1016/j.molmet.2025.102132] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/23/2025] [Revised: 03/21/2025] [Accepted: 03/26/2025] [Indexed: 04/05/2025] Open
Abstract
OBJECTIVE Liver fibrosis is a crucial condition for evaluating the prognosis of chronic liver disease. Lectin-1ike oxidized low density lipoprotein receptor-1 (LOX-1) has been shown potential research value and therapeutic targeting possibilities in different fibrotic diseases. However, the role of LOX-1 and the underlying mechanisms in liver fibrosis progression remain unclear. METHODS LOX-1 expression was detected in liver tissues from patients and rodents with liver fibrosis. LOX-1 knockout rats were subjected to CCl4 or methionine and choline-deficient diet (MCD) to induce liver fibrosis. Transcriptomic and metabolomics analysis were used to investigate the involvement and mechanism of LOX-1 on liver fibrosis. RESULTS We found that LOX-1 exacerbated liver fibrosis by promoting hepatic stellate cells (HSCs) activation. LOX-1 deletion reversed the development of liver fibrosis. We further verified that LOX-1 drove liver fibrosis by reprogramming glutamine metabolism through mediating isoform switching of glutaminase (GLS). Mechanistically, we revealed the crucial role of the LOX-1/OCT1/GLS1 axis in the pathogenesis of liver fibrosis. Moreover, LOX-1 rewired ammonia metabolism by regulating glutamine metabolism-urea cycle to drive the progression of liver fibrosis. CONCLUSIONS Our findings uncover the pivotal role of LOX-1 in the progression of liver fibrosis, enrich the pathological significance of LOX-1 regulation of hepatic ammonia metabolism, and provide an insight into promising targets for the therapeutic strategy of liver fibrosis, demonstrating the potential clinical value of targeting LOX-1 in antifibrotic therapy.
Collapse
Affiliation(s)
- Ruihua Huang
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha 410013, China
| | - Hanyu Cui
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha 410013, China
| | | | - Chuankui Fu
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha 410013, China
| | - Wei Jiang
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha 410013, China
| | - Mingyuan Cai
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha 410013, China
| | - Shuhan Zhou
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha 410013, China
| | - Xiaoyun Zhu
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha 410013, China; Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Yantai University, Yantai, China.
| | - Changping Hu
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha 410013, China; School of Pharmacy, Changzhi Medical College, Changzhi 046000, Shanxi, China; Hunan Provincial Key Laboratory of Cardiovascular Research, Changsha 410013 China.
| |
Collapse
|
2
|
Lu Z, Xu J, Li J. The Transcription Factor ATF2 Accelerates Clear Cell Renal Cell Carcinoma Progression Through Activating the PLEKHO1/NUS1 Pathway. Mol Carcinog 2025; 64:617-628. [PMID: 39777695 DOI: 10.1002/mc.23868] [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: 09/12/2024] [Revised: 11/27/2024] [Accepted: 12/02/2024] [Indexed: 01/11/2025]
Abstract
Clear cell renal cell carcinoma (ccRCC) is a common malignant cancer with high mortality rate. Activating transcription factor 2 (ATF2) and pleckstrin homology domain containing O1 (PLEKHO1) were reported to participate in numerous cancers. However, their roles and the detailed mechanisms in ccRCC development remain largely unknown. RT-qPCR and western blot were used to measure the levels of PLEKHO1, ATF2, and nuclear undecaprenyl pyrophosphate synthase 1 (NUS1). Cell proliferation, apoptosis, invasion, migration and stemness were evaluated using CCK-8 assay, flow cytometry, transwell invasion assay, wound-healing assay and sphere formation assay, respectively. Dual-luciferase reporter assay was conducted to verify the relationship between ATF2 and PLEKHO1. The interaction between PLEKHO1 and NUS1 was proved by Co-IP assay. Xenograft models were utilized to evaluate the tumorigenic capability of ccRCC cells upon PLEKHO1 knockdown. PLEKHO1, ATF2 and NUS1 expression were significantly elevated in ccRCC, and PLEKHO1 might be a prognosis biomarker for ccRCC. PLEKHO1 depletion significantly inhibited cell proliferation, invasion, migration, stemness, and induced cell apoptosis in ccRCC cells. ATF2 activated PLEKHO1 expression via transcription regulation, and PLEKHO1 overexpression could reverse the suppressive effects of ATF2 knockdown on the malignant behaviors of ccRCC cells. Moreover, PLEKHO1 directly bound to NUS1, and PLEKHO1 depletion markedly restrained ccRCC progression through targeting NUS1 in vitro and in vivo. Our findings suggested that ATF2 transcriptionally activated PLEKHO1 to promote the development of ccRCC via regulating NUS1 expression.
Collapse
Affiliation(s)
- Zheng Lu
- Gravel Center, Nanyang First People's Hospital, Nanyang, China
| | - Jinge Xu
- Department of Urology, The Fourth Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Junyu Li
- Department of Urology, The Fourth Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| |
Collapse
|
3
|
Lian Z, Zhao Y, Wen W, Zhu Z, Wang W, Zhang Z, Liu P, Favoreel HW, Li X. Distinct effects of glucocorticoid on pseudorabies virus infection in neuron-like and epithelial cells. J Virol 2025; 99:e0147224. [PMID: 39853115 PMCID: PMC11852744 DOI: 10.1128/jvi.01472-24] [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: 08/22/2024] [Accepted: 01/02/2025] [Indexed: 01/26/2025] Open
Abstract
Pseudorabies virus (PRV) is a porcine neurotropic alphaherpesvirus that infects peripheral tissues of its host, spreads into the nervous system, and establishes a life-long latency in neuronal cells. During productive infection, PRV replicates rapidly and causes pseudorabies or Aujeszky's disease. Reactivation from latent infection in the nervous system may lead to anterograde axonal transport of progeny virions, leading to recurrent infection of the epithelial layer and virus spread. Dexamethasone (DEX), a member of the glucocorticoid family that is widely used in clinical treatment as a high-efficiency glucocorticoid receptor (GR) agonist, is known to trigger reactivation of alphaherpesviruses like PRV and the closely related bovine alphaherpesvirus 1. In the current study, two cell type-dependent distinct regulatory mechanisms of glucocorticoid during PRV infection are described. In neuron-like cells, DEX upregulates expression of PRV IE180 and promotes viral productive infection. In addition, we found that GR activates the IE180 promoter by binding multiple GR response elements. The amino acids A465, P631, and I634 in GR were found to be critical for IE180 promoter activation. The impact of DEX on PRV productive infection in epithelial cells was also investigated. Interestingly, DEX was found to downregulate IE180 expression and suppress PRV infection in epithelial cells. Mechanistically, in epithelial cells, activation of the IE180 promoter by the VP16/Oct-1 (octamer-binding transcription factor 1) complex was suppressed by DEX-mediated degradation of Oct-1 in epithelial cells. In summary, our work reveals two distinct, cell type-dependent biological functions of glucocorticoid during PRV infection in neuron-like and epithelial cells, respectively.IMPORTANCEPseudorabies virus (PRV) can infect mucosal epithelium and the peripheral nervous system of its host, resulting in acute infection in epithelial cells and neuronal cells. In this study, we describe that glucocorticoid promotes PRV replication in neuron-like cells while it suppresses productive infection in epithelial cells through distinct regulations of the viral transactivator IE180, thereby revealing a cell type-dependent regulatory mechanism of glucocorticoid on PRV infection. Therefore, our findings provide a new perspective on the role of glucocorticoids during PRV infection.
Collapse
Affiliation(s)
- Zhengmin Lian
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, College of Veterinary Medicine, Yangzhou University, Yangzhou, China
| | - Yuan Zhao
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, College of Veterinary Medicine, Yangzhou University, Yangzhou, China
| | - Wei Wen
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, College of Veterinary Medicine, Yangzhou University, Yangzhou, China
| | - Zhenbang Zhu
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, College of Veterinary Medicine, Yangzhou University, Yangzhou, China
| | - Wenqiang Wang
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, College of Veterinary Medicine, Yangzhou University, Yangzhou, China
| | - Zhendong Zhang
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, College of Veterinary Medicine, Yangzhou University, Yangzhou, China
| | - Panrao Liu
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, College of Veterinary Medicine, Yangzhou University, Yangzhou, China
| | - Herman W. Favoreel
- Department of Translational Physiology, Infectiology and Public Health, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium
| | - Xiangdong Li
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, College of Veterinary Medicine, Yangzhou University, Yangzhou, China
- Joint International Research Laboratory of Agriculture and Agri-Product Safety, the Ministry of Education of China, Yangzhou University, Yangzhou, China
| |
Collapse
|
4
|
Chen T, Wang Y, Yang JL, Ni J, You K, Li X, Song Y, Wang X, Li J, Shen X, Fan Y, You Y. Gentisic acid prevents the development of atherosclerotic lesions by inhibiting SNX10-mediated stabilization of LRP6. Pharmacol Res 2024; 210:107516. [PMID: 39603572 DOI: 10.1016/j.phrs.2024.107516] [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: 07/22/2024] [Revised: 11/19/2024] [Accepted: 11/20/2024] [Indexed: 11/29/2024]
Abstract
Atherosclerotic-related acute cardiovascular events remain a leading cause of mortality worldwide, yet there are currently no pharmacological interventions available to address plaque formation or plaque rupture (PR). Here we reported that gentisic acid (GA) exerted potent therapeutic effects on plaque formation and PR in a dose-dependent manner by inhibiting LRP6-mediated macrophage apoptosis. By using the CETSA assay and DARTS assay, we identified sorting nexin 10 (SNX10) as the direct target of GA. The binding of GA to SNX10 disrupts the interaction between SNX10 and LRP6, leading to the degradation of LRP6. The downregulation of LRP6 then significantly attenuated the activation of Wnt/β-catenin pathway to exert an inhibitory effect on apoptosis. Moreover, the specific depletion of SNX10 in macrophages significantly reduced LRP6 levels and subsequently macrophage apoptosis both in vivo and in vitro. In conclusion, our findings not only suggest that GA may serve as a potential therapeutic candidate for the prevention of atherosclerosis and acute cardiovascular events caused by PR, but also confirm the druggability of SNX10 as a promising therapeutic target for atherosclerotic rupture.
Collapse
Affiliation(s)
- Tongqing Chen
- Department of Pharmacology, School of Pharmacy, Fudan University, Shanghai, China
| | - Yiming Wang
- Department of Cardiology, Worldwide Medical Center, Huashan Hospital, Fudan University, Shanghai, China
| | - Jia-Lin Yang
- Institute of Fudan-Minhang Academic Health System, Minhang Hospital, Fudan University, Shanghai, China
| | - Jiahui Ni
- Department of Pharmacology, School of Pharmacy, Fudan University, Shanghai, China
| | - Keyuan You
- Department of Pharmacology, School of Pharmacy, Fudan University, Shanghai, China
| | - Xuesong Li
- Institute of Fudan-Minhang Academic Health System, Minhang Hospital, Fudan University, Shanghai, China
| | - Yuping Song
- Institute of Fudan-Minhang Academic Health System, Minhang Hospital, Fudan University, Shanghai, China
| | - Xu Wang
- Department of Pharmacology, School of Pharmacy, Fudan University, Shanghai, China
| | - Jian Li
- Department of Cardiology, Worldwide Medical Center, Huashan Hospital, Fudan University, Shanghai, China.
| | - Xiaoyan Shen
- Department of Pharmacology, School of Pharmacy, Fudan University, Shanghai, China; Innovative Center for New Drug Development of Immune Inflammatory Diseases, Ministry of Education, Shanghai, China.
| | - Yujuan Fan
- Institute of Fudan-Minhang Academic Health System, Minhang Hospital, Fudan University, Shanghai, China.
| | - Yan You
- Department of Pharmacology, School of Pharmacy, Fudan University, Shanghai, China; Innovative Center for New Drug Development of Immune Inflammatory Diseases, Ministry of Education, Shanghai, China.
| |
Collapse
|
5
|
Cao Y, Yang Y, Guo C, Zong J, Li M, Li X, Yu T. Role of RNA-binding Proteins in Regulating Cell Adhesion and Progression of the Atherosclerotic Plaque and Plaque Erosion. Curr Atheroscler Rep 2024; 27:8. [PMID: 39576410 DOI: 10.1007/s11883-024-01250-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/15/2024] [Indexed: 11/24/2024]
Abstract
PURPOSE OF REVIEW RNA-binding proteins (RBPs) have emerged as crucial regulators of post-transcriptional processes, influencing the fate of RNA. This review delves into the biological functions of RBPs and their role in alternative splicing concerning atherosclerosis (AS), highlighting their participation in essential cellular processes. Our goal is to offer new insights for cardiovascular disease research and treatment. RECENT FINDING Dysregulation of RBPs is associated with various human diseases, including autoimmune and neurological disorders. The role of RBPs in the pathogenesis of AS is progressively being elucidated, as they influence plaque formation and disease progression by regulating cell function and gene expression. RBPs play intricate biological roles in regulating pre-mRNA, including editing, splicing, stability and translation. Alternative splicing has been demonstrated to enhance biological complexity and diversity. Our findings indicate that alternative splicing is extensively involved in the pathogenesis of AS. The dysregulated expression of specific RBPs in AS is linked to the production of adhesion molecules and vascular endothelium damage. Further research on RBPs could pave the way for the development of novel therapeutic targets.
Collapse
Affiliation(s)
- Ying Cao
- Clinical Laboratory, Central Laboratory, Qingdao Hiser Hospital Affiliated of Qingdao University (Qingdao Traditional Chinese Medicine Hospital), Qingdao, 266000, People's Republic of China
| | - Yanyan Yang
- Department of Immunology, School of Basic Medicine, Qingdao University, No. 308 Ningxia Road, Qingdao, 266000, People's Republic of China
| | - Chuan Guo
- Industrial Synergy Innovation Center, Linyi Vocational University of Science and Technology, Linyi, 276000, People's Republic of China
| | - Jinbao Zong
- Clinical Laboratory, Central Laboratory, Qingdao Hiser Hospital Affiliated of Qingdao University (Qingdao Traditional Chinese Medicine Hospital), Qingdao, 266000, People's Republic of China
| | - Min Li
- Clinical Laboratory, Central Laboratory, Qingdao Hiser Hospital Affiliated of Qingdao University (Qingdao Traditional Chinese Medicine Hospital), Qingdao, 266000, People's Republic of China
| | - Xiaolu Li
- Department of Cardiac Ultrasound, The Affiliated Hospital of Qingdao University, Qingdao, 266000, People's Republic of China
| | - Tao Yu
- Clinical Laboratory, Central Laboratory, Qingdao Hiser Hospital Affiliated of Qingdao University (Qingdao Traditional Chinese Medicine Hospital), Qingdao, 266000, People's Republic of China.
- Department of Cardiac Ultrasound, The Affiliated Hospital of Qingdao University, Qingdao, 266000, People's Republic of China.
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, No. 38 Dengzhou Road, Qingdao, 266021, People's Republic of China.
| |
Collapse
|
6
|
Xie J, Chen X, Zhou G. CKIP-1 silencing suppresses OSCC via mitochondrial homeostasis-associated TFAM/cGAS-STING signalling axis. J Cell Mol Med 2024; 28:e70006. [PMID: 39169452 PMCID: PMC11338841 DOI: 10.1111/jcmm.70006] [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: 08/25/2023] [Revised: 05/11/2024] [Accepted: 08/05/2024] [Indexed: 08/23/2024] Open
Abstract
Limited effective targets have challenged the treatment of oral squamous cell carcinoma (OSCC). Casein kinase 2 interacting protein 1 (CKIP-1) is a scaffold protein involved in various diseases. However, the role of CKIP-1 in OSCC remains unclear. The aim of this study was to explore the regulatory role of CKIP-1 in OSCC, as well as the involved mechanism. First, higher expression of CKIP-1 in OSCC tissues and cell lines were found. Series of gain- and loss-of-function experiments demonstrated suppressed malignant behaviours and enhanced apoptosis of OSCC cells when CKIP-1 was silenced. Also, inhibited tumour growth in CKIP-1-silenced group were proved. Further, mitochondrial transcription factor A (TFAM) downregulation, increased ROS production, decreased mitochondrial membrane potential and cGAS-STING activation in CKIP-1-silenced group were observed. The involvement of mitochondrial homeostasis-related TFAM/cGAS-STING axis in CKIP-1-silenced OSCC cells was finally demonstrated by tetramethylpyrazine (TMP) that inhibits TFAM degradation. Taken together, our study demonstrated that CKIP-1 silencing could significantly antagonize OSCC via TFAM/cGAS-STING axis, which may provide a candidate target for OSCC treatment.
Collapse
Affiliation(s)
- Ji‐Rong Xie
- State Key Laboratory of Oral and Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School and Hospital of StomatologyWuhan UniversityWuhanChina
| | - Xiao‐Jie Chen
- State Key Laboratory of Oral and Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School and Hospital of StomatologyWuhan UniversityWuhanChina
- Department of Oral Medicine, School and Hospital of StomatologyWuhan UniversityWuhanChina
| | - Gang Zhou
- State Key Laboratory of Oral and Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School and Hospital of StomatologyWuhan UniversityWuhanChina
- Department of Oral Medicine, School and Hospital of StomatologyWuhan UniversityWuhanChina
| |
Collapse
|
7
|
Li X, Zhao Y, Xue M, Hu H, Yin J, Cheng W, Shi Y, Wang Y, Yan S. CKIP-1 mediates CK2 translocation to regulate Nav1.5 and Kir2.1 channel complexes in cardiomyocytes. J Biochem Mol Toxicol 2024; 38:e23780. [PMID: 39056188 DOI: 10.1002/jbt.23780] [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: 11/23/2023] [Revised: 05/09/2024] [Accepted: 07/15/2024] [Indexed: 07/28/2024]
Abstract
Sodium and potassium channels, especially Nav1.5 and Kir2.1, play key roles in the formation of action potentials in cardiomyocytes. These channels interact with, and are regulated by, synapse-associated protein 97 (SAP97). However, the regulatory role of SAP97 in myocyte remains incompletely understood. Here, we investigate the function of SAP97 phosphorylation in the regulation of Nav1.5 and Kir2.1 channel complexes and the upstream regulation of SAP97. We found that SAP97 is phosphorylated by casein kinase II (CK2) in vitro. In addition, transfection of casein kinase 2 interacting protein-1 (CKIP-1) into cardiomyocytes to drive CK2 from the nucleus to the cytoplasm, increased SAP97 phosphorylation and Nav1.5 and Kir2.1 current activity. These findings demonstrated that CKIP-1 modulates the subcellular translocation of CK2, which regulates Nav1.5 and Kir2.1 channel complex formation and activity in cardiomyocytes.
Collapse
Affiliation(s)
- Xinran Li
- Department of Cardiology, Shandong Medicine and Health Key Laboratory of Cardiac Electrophysiology and Arrhythmia, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, People's Republic of China
| | - Yingzhu Zhao
- Department of Cardiology, Shandong Medicine and Health Key Laboratory of Cardiac Electrophysiology and Arrhythmia, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, People's Republic of China
| | - Mei Xue
- Department of Cardiology, Shandong Medicine and Health Key Laboratory of Cardiac Electrophysiology and Arrhythmia, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, People's Republic of China
| | - Hesheng Hu
- Department of Cardiology, Shandong Medicine and Health Key Laboratory of Cardiac Electrophysiology and Arrhythmia, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, People's Republic of China
| | - Jie Yin
- Department of Cardiology, Shandong Medicine and Health Key Laboratory of Cardiac Electrophysiology and Arrhythmia, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, People's Republic of China
| | - Wenjuan Cheng
- Department of Cardiology, Shandong Medicine and Health Key Laboratory of Cardiac Electrophysiology and Arrhythmia, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, People's Republic of China
| | - Yugen Shi
- Department of Cardiology, Shandong Medicine and Health Key Laboratory of Cardiac Electrophysiology and Arrhythmia, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, People's Republic of China
| | - Ye Wang
- Department of Cardiology, Shandong Medicine and Health Key Laboratory of Cardiac Electrophysiology and Arrhythmia, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, People's Republic of China
| | - Suhua Yan
- Department of Cardiology, Shandong Medicine and Health Key Laboratory of Cardiac Electrophysiology and Arrhythmia, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, People's Republic of China
| |
Collapse
|
8
|
Yang Y, Wang J, Tian Y, Li M, Xu S, Zhang L, Luo X, Tan Y, Liang H, Chen M. Equisetin protects from atherosclerosis in vivo by binding to STAT3 and inhibiting its activity. Pharmacol Res 2024; 206:107289. [PMID: 38960011 DOI: 10.1016/j.phrs.2024.107289] [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: 05/08/2024] [Revised: 06/21/2024] [Accepted: 06/28/2024] [Indexed: 07/05/2024]
Abstract
Atherosclerosis is a chronic inflammatory vascular disease characterized by lipid metabolism disorder and lipid accumulation. Equisetin (EQST) is a hemiterpene compound isolated from fungus of marine sponge origin, which has antibacterial, anti-inflammatory, lipid-lowering, and weight loss effects. Whether EQST has anti-atherosclerotic activity has not been reported. In this study, we revealed that EQST displayed anti- atherosclerosis effects through inhibiting macrophage inflammatory response, lipid uptake and foam cell formation in vitro, and finally ameliorated high-fat diet (HFD)-induced atherosclerosis in AopE-/- mice in vivo. Mechanistically, EQST directly bound to STAT3 with high-affinity by forming hydrophobic bonds at GLN247 and GLN326 residues, as well as hydrogen bonds at ARG325 and THR346 residues. EQST interacted with STAT3 physically, and functionally inhibited the transcription activity of STAT3, thereby regulating atherosclerosis. Therefore, these results supports EQST as a candidate for developing anti-atherosclerosis therapeutic agent.
Collapse
Affiliation(s)
- Yuting Yang
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources (Ministry of Education of China), Collaborative Innovation Center for Guangxi Ethnic Medicine, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin 541004, China
| | - Jingzhu Wang
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources (Ministry of Education of China), Collaborative Innovation Center for Guangxi Ethnic Medicine, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin 541004, China
| | - Yang Tian
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources (Ministry of Education of China), Collaborative Innovation Center for Guangxi Ethnic Medicine, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin 541004, China
| | - Min Li
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources (Ministry of Education of China), Collaborative Innovation Center for Guangxi Ethnic Medicine, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin 541004, China
| | - Shaohua Xu
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources (Ministry of Education of China), Collaborative Innovation Center for Guangxi Ethnic Medicine, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin 541004, China
| | - Lijun Zhang
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources (Ministry of Education of China), Collaborative Innovation Center for Guangxi Ethnic Medicine, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin 541004, China; GXNU & GLHCWM Joint Medical Research Center, Guangxi Normal University, Guilin 541004, China
| | - Xiaowei Luo
- Institute of Marine Drugs, Guangxi University of Chinese Medicine, Nanning 530200, China
| | - Yanhui Tan
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources (Ministry of Education of China), Collaborative Innovation Center for Guangxi Ethnic Medicine, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin 541004, China.
| | - Hong Liang
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources (Ministry of Education of China), Collaborative Innovation Center for Guangxi Ethnic Medicine, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin 541004, China.
| | - Ming Chen
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources (Ministry of Education of China), Collaborative Innovation Center for Guangxi Ethnic Medicine, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin 541004, China; GXNU & GLHCWM Joint Medical Research Center, Guangxi Normal University, Guilin 541004, China.
| |
Collapse
|
9
|
Xue Q, Ma Y, Shao H. Bacillus amyloliquefaciens Protect Against Atherosclerosis Through Alleviating Foam Cell Formation and Macrophage Polarization. Curr Microbiol 2024; 81:263. [PMID: 38997545 DOI: 10.1007/s00284-024-03775-w] [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: 07/07/2023] [Accepted: 11/01/2023] [Indexed: 07/14/2024]
Abstract
This study was to investigate the therapeutic effect of Bacillus amyloliquefaciens (Ba) on atherosclerosis (AS). THP-1 monocyte was differentiated to THP-1 macrophage (THP-M) through phorbol 12-myristate 13-acetate. After pre-treatment by 108 cfu/ml Ba lasting 6 h, THP-M was induced with 100 mg/l ox-LDL lasting 48 h to form macrophage foam cell (THP-F). RT-qPCR and flow cytometry were employed to determine the polarization of THP-M and THP-F. ApoE-/- mice with high-fat and high-cholesterol diet were used for constructing an AS model to evaluate the effect of Ba on AS. Our in vitro results showed that Ba vegetative cells pre-treatment distinctly inhibited the levels of iNOS and CD16/CD32 (M1 macrophage markers), and increased the levels of FIZZ1, Ym1, Arg1, CD163, and CD206 (M2 macrophage markers), indicating that Ba pre-treatment promoted anti-inflammatory M2-like polarization both in THP-M and THP-F. Meanwhile, it also suppressed cholesterol uptake, esterification, and hydrolysis, and efflux by THP-M and THP-F. Additionally, our animal experiments demonstrated that Ba vegetative cells treatment suppressed high cholesterol, hyperglycemia, hyperlipidemia, and the release of inflammatory factors (TNF-α, IL-6 and IL-1β) in ApoE-/- AS mice. In a word, our results indicated that Ba may protect against AS through alleviating foam cell formation and macrophage polarization through targeting certain stages of AS.
Collapse
Affiliation(s)
- Qi Xue
- Department of Cardiology, Zhejiang Provincial People's Hospital, No. 158 Shangtang Road, Hangzhou, 310014, Zhejiang, China
| | - Yuan Ma
- Department of Cardiology, Zhejiang Provincial People's Hospital, No. 158 Shangtang Road, Hangzhou, 310014, Zhejiang, China
| | - Hong Shao
- Department of Cardiology, Zhejiang Provincial People's Hospital, No. 158 Shangtang Road, Hangzhou, 310014, Zhejiang, China.
| |
Collapse
|
10
|
Chen Q, Wu Z, Shi Y, Li Z, Yang J, Qu M, Zhang S, Wang Z, Ji N, Li J, Shen Y, Xie L, Chen Q. Loss of PA28γ exacerbates imbalanced differentiation of bone marrow stromal cells during bone formation and bone healing in mice. J Bone Miner Res 2024; 39:326-340. [PMID: 38477820 DOI: 10.1093/jbmr/zjae012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/24/2023] [Revised: 01/04/2024] [Accepted: 01/10/2024] [Indexed: 03/14/2024]
Abstract
Proteasome activator subunit 3 (PA28γ) is a member of the proteasome activator family, which mainly regulates the degradation and stability of proteins. Studies have shown that it plays crucial roles in lipid formation, stemness maintenance, and blood vessel formation. However, few studies have clarified the association between PA28γ and bone diseases. Herein, we identified PA28γ as a previously unknown regulator of bone homeostasis that coordinates bone formation and lipid accumulation. PA28γ-knockout mice presented with the characteristics of low bone mass and accumulation of lipids. Suppressed expression of PA28γ restrained the osteogenic differentiation and enhanced the adipogenic differentiation of bone marrow stromal cells (BMSCs). Overexpression of PA28γ promoted osteogenic differentiation and inhibited adipogenic differentiation of BMSCs. Mechanistically, PA28γ interacted with Wnt5α, and the two interactors appeared to be positively correlated. PA28γ mainly activated the downstream Wnt/β-catenin signaling pathway, which affects BMSCs differentiation homeostasis. Deletion of Wnt5α significantly delayed the promotion of osteogenic differentiation and partially alleviated the inhibitory effect of adipogenic differentiation of BMSCs in the PA28γ-overexpressing group. Furthermore, we demonstrated that PA28γ-knockout mice had an inhibited rate of bone healing in a drill-hole femoral bone defect model in vivo. Therefore, our results confirm the effects of PA28γ on bone formation and bone defect repair, indicating that PA28γ mainly interacts with Wnt5α to activate the Wnt/β-catenin signaling pathway regulating BMSCs differentiation homeostasis. Our results reveal the function of PA28γ in bone diseases and provide a new theoretical basis for expanding the treatment of bone diseases.
Collapse
Affiliation(s)
- Qian Chen
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Research Unit of Oral Carcinogenesis and Management, Chinese Academy of Medical Sciences, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, China
- The Affiliated Hospital of Stomatology, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou 310006, P. R. China
| | - ZuPing Wu
- The Affiliated Hospital of Stomatology, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou 310006, P. R. China
| | - YuJie Shi
- Department of Stomatology, The First Affiliated Hospital with Nanjing Medical University, Nanjing 210029, P. R. China
| | - ZaiYe Li
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Research Unit of Oral Carcinogenesis and Management, Chinese Academy of Medical Sciences, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, China
| | - JiaKang Yang
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Research Unit of Oral Carcinogenesis and Management, Chinese Academy of Medical Sciences, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, China
| | - MoYuan Qu
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Research Unit of Oral Carcinogenesis and Management, Chinese Academy of Medical Sciences, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, China
| | - ShiYu Zhang
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Research Unit of Oral Carcinogenesis and Management, Chinese Academy of Medical Sciences, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, China
| | - Zheng Wang
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Research Unit of Oral Carcinogenesis and Management, Chinese Academy of Medical Sciences, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, China
| | - Ning Ji
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Research Unit of Oral Carcinogenesis and Management, Chinese Academy of Medical Sciences, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, China
| | - Jing Li
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Research Unit of Oral Carcinogenesis and Management, Chinese Academy of Medical Sciences, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, China
| | - YingQiang Shen
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Research Unit of Oral Carcinogenesis and Management, Chinese Academy of Medical Sciences, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, China
| | - Liang Xie
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Research Unit of Oral Carcinogenesis and Management, Chinese Academy of Medical Sciences, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, China
| | - Qianming Chen
- The Affiliated Hospital of Stomatology, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou 310006, P. R. China
| |
Collapse
|
11
|
Shen S, Sun T, Ding X, Gu X, Wang Y, Ma X, Li Z, Gao H, Ge S, Feng Q. The exoprotein Gbp of Fusobacterium nucleatum promotes THP-1 cell lipid deposition by binding to CypA and activating PI3K-AKT/MAPK/NF-κB pathways. J Adv Res 2024; 57:93-105. [PMID: 37100345 PMCID: PMC10918358 DOI: 10.1016/j.jare.2023.04.007] [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: 01/09/2023] [Revised: 04/07/2023] [Accepted: 04/11/2023] [Indexed: 04/28/2023] Open
Abstract
INTRODUCTION Growing evidence has shown the correlation between periodontitis and atherosclerosis, while our knowledge on the pathogenesis of periodontitis-promoting atherosclerosis is far from sufficient. OBJECTIVES Illuminate the pathogenic effects of Fusobacterium nucleatum (F. nucleatum) on intracellular lipid deposition in THP-1-derived macrophages and elucidate the underlying pathogenic mechanism of how F. nucleatum promoting atherosclerosis. METHODS AND RESULTS F. nucleatum was frequently detected in different kinds of atherosclerotic plaques and its abundance was positively correlated with the proportion of macrophages. In vitro assays showed F. nucleatum could adhere to and invade THP-1 cells, and survive continuously in macrophages for 24 h. F. nucleatum stimulation alone could significantly promote cellular inflammation, lipid uptake and inhibit lipid outflow. The dynamic gene expression of THP-1 cells demonstrated that F. nucleatum could time-serially induce the over-expression of multiple inflammatory related genes and activate NF-κB, MAPK and PI3K-AKT signaling pathways. The exoprotein of F. nucleatum, D-galactose-binding protein (Gbp), acted as one of the main pathogenic proteins to interact with the Cyclophilin A (CypA) of THP-1 cells and induced the activation of the NF- κB, MAPK and PI3K-AKT signaling pathways. Furthermore, use of six candidate drugs targeting to the key proteins in NF- κB, MAPK and PI3K-AKT pathways could dramatically decrease F. nucleatum induced inflammation and lipid deposition in THP-1 cells. CONCLUSIONS This study suggests that the periodontal pathogen F. nucleatum can activate macrophage PI3K-AKT/MAPK/NF-κB signal pathways, promotes inflammation, enhances cholesterol uptake, reduces lipid excretion, and promotes lipid deposition, which may be one of its main strategies promoting the development of atherosclerosis.
Collapse
Affiliation(s)
- Song Shen
- Department of Human Microbiome & Periodontology & Implantology & Orthodontics, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration & Shandong Provincial Clinical Research Center for Oral Diseases, Jinan 250012, China
| | - Tianyong Sun
- Department of Human Microbiome & Periodontology & Implantology & Orthodontics, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration & Shandong Provincial Clinical Research Center for Oral Diseases, Jinan 250012, China
| | - Xiangjiu Ding
- Department of Vascular Surgery, General Surgery, Qilu Hospital of Shandong University, Jinan 250012, China
| | - Xiufeng Gu
- Department of Human Microbiome & Periodontology & Implantology & Orthodontics, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration & Shandong Provincial Clinical Research Center for Oral Diseases, Jinan 250012, China
| | - Yushang Wang
- Department of Human Microbiome & Periodontology & Implantology & Orthodontics, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration & Shandong Provincial Clinical Research Center for Oral Diseases, Jinan 250012, China
| | - Xiaomei Ma
- Department of Human Microbiome & Periodontology & Implantology & Orthodontics, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration & Shandong Provincial Clinical Research Center for Oral Diseases, Jinan 250012, China
| | - Zixuan Li
- Department of Human Microbiome & Periodontology & Implantology & Orthodontics, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration & Shandong Provincial Clinical Research Center for Oral Diseases, Jinan 250012, China
| | - Haiting Gao
- Department of Human Microbiome & Periodontology & Implantology & Orthodontics, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration & Shandong Provincial Clinical Research Center for Oral Diseases, Jinan 250012, China
| | - Shaohua Ge
- Department of Human Microbiome & Periodontology & Implantology & Orthodontics, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration & Shandong Provincial Clinical Research Center for Oral Diseases, Jinan 250012, China.
| | - Qiang Feng
- Department of Human Microbiome & Periodontology & Implantology & Orthodontics, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration & Shandong Provincial Clinical Research Center for Oral Diseases, Jinan 250012, China; State Key Laboratory of Microbial Technology, Shandong University, Qingdao 266237, China.
| |
Collapse
|
12
|
Li Y, Zhou M, Li H, Dai C, Yin L, Liu C, Li Y, Zhang E, Dong X, Ji H, Hu Q. Macrophage P2Y6 receptor deletion attenuates atherosclerosis by limiting foam cell formation through phospholipase Cβ/store-operated calcium entry/calreticulin/scavenger receptor A pathways. Eur Heart J 2024; 45:268-283. [PMID: 38036416 DOI: 10.1093/eurheartj/ehad796] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Revised: 10/16/2023] [Accepted: 11/17/2023] [Indexed: 12/02/2023] Open
Abstract
BACKGROUND AND AIMS Macrophage-derived foam cells play a causal role during the pathogenesis of atherosclerosis. P2Y6 receptor (P2Y6R) highly expressed has been considered as a disease-causing factor in atherogenesis, but the detailed mechanism remains unknown. This study aims to explore P2Y6R in regulation of macrophage foaming, atherogenesis, and its downstream pathways. Furthermore, the present study sought to find a potent P2Y6R antagonist and investigate the feasibility of P2Y6R-targeting therapy for atherosclerosis. METHODS The P2Y6R expression was examined in human atherosclerotic plaques and mouse artery. Atherosclerosis animal models were established in whole-body P2Y6R or macrophage-specific P2Y6R knockout mice to evaluate the role of P2Y6R. RNA sequencing, DNA pull-down experiments, and proteomic approaches were performed to investigate the downstream mechanisms. High-throughput Glide docking pipeline from repurposing drug library was performed to find potent P2Y6R antagonists. RESULTS The P2Y6R deficiency alleviated atherogenesis characterized by decreasing plaque formation and lipid deposition of the aorta. Mechanically, deletion of macrophage P2Y6R significantly inhibited uptake of oxidized low-density lipoprotein through decreasing scavenger receptor A expression mediated by phospholipase Cβ/store-operated calcium entry pathways. More importantly, P2Y6R deficiency reduced the binding of scavenger receptor A to CALR, accompanied by dissociation of calreticulin and STIM1. Interestingly, thiamine pyrophosphate was found as a potent P2Y6R antagonist with excellent P2Y6R antagonistic activity and binding affinity, of which the pharmacodynamic effect and mechanism on atherosclerosis were verified. CONCLUSIONS Macrophage P2Y6R regulates phospholipase Cβ/store-operated calcium entry/calreticulin signalling pathway to increase scavenger receptor A protein level, thereby improving foam cell formation and atherosclerosis, indicating that the P2Y6R may be a potential therapeutic target for intervention of atherosclerotic diseases using P2Y6R antagonists including thiamine pyrophosphate.
Collapse
Affiliation(s)
- Yehong Li
- Department of Pharmacology, School of Pharmacy, China Pharmaceutical University, Longmian Avenue 639, Nanjing 211198, China
| | - Mengze Zhou
- Department of Pharmacology, School of Pharmacy, China Pharmaceutical University, Longmian Avenue 639, Nanjing 211198, China
| | - Huanqiu Li
- College of Pharmaceutical Sciences, Soochow University, Suzhou, China
| | - Chen Dai
- Experimental Teaching Center of Life Science, College of Life Sciences, Nanjing Agricultural University, Nanjing 210095, China
| | - Li Yin
- Department of Pharmacology, School of Pharmacy, China Pharmaceutical University, Longmian Avenue 639, Nanjing 211198, China
| | - Chunxiao Liu
- Department of Pharmacology, School of Pharmacy, China Pharmaceutical University, Longmian Avenue 639, Nanjing 211198, China
| | - Yuxin Li
- Department of Pharmacology, School of Pharmacy, China Pharmaceutical University, Longmian Avenue 639, Nanjing 211198, China
| | - Enming Zhang
- Department of Pharmacology, School of Pharmacy, China Pharmaceutical University, Longmian Avenue 639, Nanjing 211198, China
| | - Xinli Dong
- Department of Pharmacology, School of Pharmacy, China Pharmaceutical University, Longmian Avenue 639, Nanjing 211198, China
| | - Hui Ji
- Department of Pharmacology, School of Pharmacy, China Pharmaceutical University, Longmian Avenue 639, Nanjing 211198, China
| | - Qinghua Hu
- Department of Pharmacology, School of Pharmacy, China Pharmaceutical University, Longmian Avenue 639, Nanjing 211198, China
| |
Collapse
|
13
|
Cao LN, Wang YY, Hou XY, Zheng HD, Wei RZ, Zhao RR, Shen WY, Yang Y, Chu JF, Tian GY, Xiao J, Tian T. New insights on the association of weight loss with the reduction in carotid intima-media thickness among patients with obesity: an updated systematic review and meta-analysis. Public Health 2024; 226:248-254. [PMID: 38091813 DOI: 10.1016/j.puhe.2023.11.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2023] [Revised: 10/09/2023] [Accepted: 11/07/2023] [Indexed: 01/15/2024]
Abstract
OBJECTIVES Carotid intima-media thickness (CIMT) is a noninvasive marker of atherosclerosis, a typical pathologic process underlying cardiovascular diseases (CVDs). It is essential to explore the relationships between weight loss and the reduction of CIMT. STUDY DESIGN This was an updated systematic review and meta-analysis. METHODS A systematic literature search was conducted to collect relevant clinical trials. The pooled results of meta-analyses were assessed by weighted mean difference (WMD) and the corresponding 95 % confidence interval (95% CI). RESULTS Thirty-three articles involving 2273 participants were collected in this meta-analysis. Among all participants with obesity, the pooled mean of weight loss was -23.26 kg (95% CI: -27.71 to -18.81), and the pooled mean change of CIMT was -0.06 mm (95% CI: -0.08 to -0.04). Compared with Non-surgical interventions, Surgical ones could lead to much higher weight loss (Pbetween groups < 0.001). A more significant CIMT reduction was identified among Surgical intervention patients than among Non-surgical intervention participants (Pbetween groups < 0.001). CONCLUSIONS Effective interventions, especially Surgical interventions, could reduce the weight of patients with obesity, followed by the decline of CIMT, which might further disturb atherosclerosis progression and lower CVD risk.
Collapse
Affiliation(s)
- L N Cao
- The Department of Epidemiology and Medical Statistics, School of Public Health, Nantong University, Nantong 226019, China
| | - Y Y Wang
- The Department of Epidemiology and Medical Statistics, School of Public Health, Nantong University, Nantong 226019, China
| | - X Y Hou
- The Center for Disease Control and Prevention of Nantong, Nantong 226007, China
| | - H D Zheng
- The Department of Epidemiology and Medical Statistics, School of Public Health, Nantong University, Nantong 226019, China
| | - R Z Wei
- The Department of Epidemiology and Medical Statistics, School of Public Health, Nantong University, Nantong 226019, China
| | - R R Zhao
- The Department of Oncology, Jiangdu People's Hospital of Yangzhou, Yangzhou 225202, China
| | - W Y Shen
- The Department of Epidemiology and Medical Statistics, School of Public Health, Nantong University, Nantong 226019, China
| | - Y Yang
- The Department of Epidemiology and Medical Statistics, School of Public Health, Nantong University, Nantong 226019, China
| | - J F Chu
- The Department of Oncology, Jiangdu People's Hospital of Yangzhou, Yangzhou 225202, China
| | - G Y Tian
- The Department of Oncology, Jiangdu People's Hospital of Yangzhou, Yangzhou 225202, China.
| | - J Xiao
- The Department of Epidemiology and Medical Statistics, School of Public Health, Nantong University, Nantong 226019, China.
| | - T Tian
- The Department of Epidemiology and Medical Statistics, School of Public Health, Nantong University, Nantong 226019, China.
| |
Collapse
|
14
|
Xiong J, Zhang L, Chen G, Dong P, Tong J, Hua L, Li N, Wen L, Zhu L, Chang W, Jin Y. Associations of CKIP-1 and LOX-1 polymorphisms with the risk of type 2 diabetes mellitus with hypertension among Chinese adults. Acta Diabetol 2024; 61:43-52. [PMID: 37668684 DOI: 10.1007/s00592-023-02175-z] [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: 04/29/2023] [Accepted: 08/18/2023] [Indexed: 09/06/2023]
Abstract
AIMS Type 2 diabetes mellitus (T2DM) and hypertension are common high-incidence diseases, closely related, and have common pathogenic basis such as oxidative stress. Casein kinase 2 interacting protein-1 (CKIP-1) and low-density lipoprotein receptor (LOX-1) are considered to be important factors affect the level of oxidative stress in the body. The main purpose of this study was to explore the relationship between CKIP-1 (rs6693817 A > T, rs2306235 C > G) and LOX-1 (rs1050283 G > A, rs11053646 C > G) polymorphisms and the risk of hypertension and diabetes, and try to find new candidate genes for diabetes and diabetes with hypertension etiology in Chinese population. METHODS 574 T2DM patients and 597 controls frequently matched by age and sex were selected for genotyping of CKIP-1 (rs6693817 A > T, rs2306235 C > G) and LOX-1 gene (rs1050283 G > A, rs11053646 C > G). Logistic regression was used to analyze the correlation between different genotypes and the risk of T2DM and T2DM with hypertension, and the results were expressed as odds ratio (OR) and 95% confidence interval (95% CI). RESULTS We found that the risk of T2DM in the AA + AT genotype of rs6693817 was higher than that in the TT genotype in Chinese population (OR = 1.318, 95%CI: 1.011-1.717, P = 0.041), and the difference was still significant after adjustment (OR = 1.370, 95%CI: 1.043-1.799, Padjusted = 0.024), the difference of heterozygotes (AT vs TT: OR = 1.374, 95%CI: 1.026-1.840, Padjusted = 0.033) was statistically significant. But after Bonferroni correction, the significance of the above sites disappeared. And rs6693817 was associated with the risk of T2DM combined with hypertension before and after adjustment in dominant model (OR = 1.424, 95% CI: 1.038-1.954, P = 0.028; OR = 1.460, 95% CI: 1.057-2.015, Padjusted = 0.021, respectively) and in heterozygote model (OR = 1.499, 95% CI: 1.069-2.102, P = 0.019; OR = 1.562, 95% CI: 1.106-2.207, Padjusted = 0.011, respectively). However, only the statistical significance of the heterozygous model remained after Bonferroni correction. rs2306235, rs1050283 and rs11053646 were not significantly correlated with T2DM and T2DM combined with hypertension risk (P > 0.05). CONCLUSIONS The results suggest that CKIP-1 rs6693817 is related to the susceptibility of Chinese people to T2DM with hypertension, providing a new genetic target for the treatment of diabetes with hypertension with in the future.
Collapse
Affiliation(s)
- Jiajie Xiong
- Department of Epidemiology and Health Statistics, School of Public Health, Wannan Medical College, Wuhu, 241002, Anhui, China
| | - Liu Zhang
- Department of Hospital Infection Management Office, Wuhu Hospital of Traditional Chinese Medicine, Wuhu, 241000, Anhui, China
| | - Guimei Chen
- School of Health Management, Anhui Medical University, Hefei, 230032, Anhui, China
| | - Pu Dong
- Department of Epidemiology and Health Statistics, School of Public Health, Wannan Medical College, Wuhu, 241002, Anhui, China
| | - Jiani Tong
- Department of Epidemiology and Health Statistics, School of Public Health, Wannan Medical College, Wuhu, 241002, Anhui, China
| | - Long Hua
- Department of Epidemiology and Health Statistics, School of Public Health, Wannan Medical College, Wuhu, 241002, Anhui, China
| | - Ning Li
- Department of Epidemiology and Health Statistics, School of Public Health, Wannan Medical College, Wuhu, 241002, Anhui, China
| | - Liying Wen
- Department of Epidemiology and Health Statistics, School of Public Health, Wannan Medical College, Wuhu, 241002, Anhui, China
| | - Lijun Zhu
- Department of Epidemiology and Health Statistics, School of Public Health, Wannan Medical College, Wuhu, 241002, Anhui, China
| | - Weiwei Chang
- Department of Epidemiology and Health Statistics, School of Public Health, Wannan Medical College, Wuhu, 241002, Anhui, China.
| | - Yuelong Jin
- Department of Epidemiology and Health Statistics, School of Public Health, Wannan Medical College, Wuhu, 241002, Anhui, China.
| |
Collapse
|
15
|
Thomas T, Salcedo-Tacuma D, Smith DM. Structure, Function, and Allosteric Regulation of the 20S Proteasome by the 11S/PA28 Family of Proteasome Activators. Biomolecules 2023; 13:1326. [PMID: 37759726 PMCID: PMC10526260 DOI: 10.3390/biom13091326] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Revised: 08/24/2023] [Accepted: 08/24/2023] [Indexed: 09/29/2023] Open
Abstract
The proteasome, a complex multi-catalytic protease machinery, orchestrates the protein degradation essential for maintaining cellular homeostasis, and its dysregulation also underlies many different types of diseases. Its function is regulated by many different mechanisms that encompass various factors such as proteasome activators (PAs), adaptor proteins, and post-translational modifications. This review highlights the unique characteristics of proteasomal regulation through the lens of a distinct family of regulators, the 11S, REGs, or PA26/PA28. This ATP-independent family, spanning from amoebas to mammals, exhibits a common architectural structure; yet, their cellular biology and criteria for protein degradation remain mostly elusive. We delve into their evolution and cellular biology, and contrast their structure and function comprehensively, emphasizing the unanswered questions regarding their regulatory mechanisms and broader roles in proteostasis. A deeper understanding of these processes will illuminate the roles of this regulatory family in biology and disease, thus contributing to the advancement of therapeutic strategies.
Collapse
Affiliation(s)
- Taylor Thomas
- Department of Biochemistry and Molecular Medicine, School of Medicine, West Virginia University, 64 Medical Center Drive, Morgantown, WV 26506, USA
| | - David Salcedo-Tacuma
- Department of Biochemistry and Molecular Medicine, School of Medicine, West Virginia University, 64 Medical Center Drive, Morgantown, WV 26506, USA
| | - David M. Smith
- Department of Biochemistry and Molecular Medicine, School of Medicine, West Virginia University, 64 Medical Center Drive, Morgantown, WV 26506, USA
- Department of Neuroscience, Rockefeller Neuroscience Institute, West Virginia University, Morgantown, WV 26506, USA
| |
Collapse
|
16
|
Li C, Song Z, Gao P, Duan W, Liu X, Liang S, Gong Q, Guo J. Transaldolase inhibits CD36 expression by modulating glutathione-p38 signaling, exerting protective effects against macrophage foam cell formation. Acta Biochim Biophys Sin (Shanghai) 2023; 55:1496-1505. [PMID: 37528662 PMCID: PMC10520467 DOI: 10.3724/abbs.2023146] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Accepted: 05/19/2023] [Indexed: 08/03/2023] Open
Abstract
In atherosclerosis, macrophage-derived foam cell formation is considered to be a hallmark of the pathological process; this occurs via the uptake of modified lipoproteins. In the present study, we aim to determine the role of transaldolase in foam cell formation and atherogenesis and reveal the mechanisms underlying its role. Bone marrow-derived macrophages (BMDMs) isolated from mice successfully form foam cells after treatment with oxidized low-density lipoprotein (80 μg/mL). Elevated transaldolase levels in the foam cell model are assessed by quantitative polymerase chain reaction and western blot analysis. Transaldolase overexpression and knockdown in BMDMs are achieved via plasmid transfection and small interfering RNA technology, respectively. We find that transaldolase overexpression effectively attenuates, whereas transaldolase knockdown accelerates, macrophage-derived foam cell formation through the inhibition or activation of cholesterol uptake mediated by the scavenger receptor cluster of differentiation 36 (CD36) in a p38 mitogen-activated protein kinase (MAPK) signaling-dependent manner. Transaldolase-mediated glutathione (GSH) homeostasis is identified as the upstream regulator of p38 MAPK-mediated CD36-dependent cholesterol uptake in BMDMs. Transaldolase upregulates GSH production, thereby suppressing p38 activity and reducing the CD36 level, ultimately preventing foam cell formation and atherosclerosis. Thus, our findings indicate that the transaldolase-GSH-p38-CD36 axis may represent a promising therapeutic target for atherosclerosis.
Collapse
Affiliation(s)
- Chengyi Li
- Department of ImmunologySchool of MedicineYangtze UniversityJingzhou434023China
| | - Zihao Song
- Department of ImmunologySchool of MedicineYangtze UniversityJingzhou434023China
| | - Pengyue Gao
- Department of ImmunologySchool of MedicineYangtze UniversityJingzhou434023China
| | - Wei Duan
- Department of OncologyJingzhou Hospital Affiliated to Yangtze UniversityJingzhou434023China
| | - Xiu Liu
- Department of Cardiovascular SurgeryNanfang HospitalSouthern Medical UniversityGuangzhou510515China
| | - Sijia Liang
- Department of Pharmacologyand Cardiac & Cerebral Vascular Research CenterZhongshan School of MedicineSun Yat-Sen UniversityGuangzhou510080China
| | - Quan Gong
- Department of ImmunologySchool of MedicineYangtze UniversityJingzhou434023China
| | - Jiawei Guo
- Department of ImmunologySchool of MedicineYangtze UniversityJingzhou434023China
| |
Collapse
|
17
|
Huang X, Xiao J, Wang H, Peng Y, Liu H, Ma L, Wang X, Cao Z. CKIP-1 mediates P. gingivalis-suppressed osteogenic/cementogenic differentiation of periodontal ligament cells partially via p38 signaling pathway. J Oral Microbiol 2023; 15:2236427. [PMID: 37483640 PMCID: PMC10360982 DOI: 10.1080/20002297.2023.2236427] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Revised: 07/03/2023] [Accepted: 07/08/2023] [Indexed: 07/25/2023] Open
Abstract
OBJECTIVES Casein kinase 2 interacting protein-1 (CKIP-1) is a versatile player involved in various biological processes. However, whether CKIP-1 mediates the osteogenic/cementogenic differentiation of periodontal ligament cells (PDLCs) under Porphyromonas gingivalis (Pg) stimulation remains unknown. MATERIAL AND METHODS The effect of Pg on PDLC differentiation was first verified. CKIP-1 expression in Pg-infected PDLCs or in PDL of apical periodontitis (AP) mice was detected. The changes of CKIP-1 during PDLC differentiation was also determined. PDLC differentiation capacity in CKIP-1 knockout (KO) mice and CKIP-1-silenced PDLCs with or without Pg stimulation were further studied. Inhibitor was finally applied to verify the involvement of p38 signaling pathway in PDLC differentiation. RESULTS The suppression effect of Pg on PDLC differentiation was demonstrated. CKIP-1 increased in the PDL of AP mice and Pg-induced PDLCs, and decreased gradually during PDLC differentiation. Increased OSX and RUNX2 expression in PDL were observed in CKIP-1 KO mice. Also, CKIP-1 silencing facilitated and rescued Pg-inhibited PDLC differentiation. Inhibitor for p38 signaling pathway blocked CKIP-1 silencing-facilitated PDLC differentiation. CONCLUSIONS CKIP-1 mediated the osteogenic/cementogenic differentiation of PDLCs partially through p38 signaling pathway, which may provide evidence for the regeneration of periodontal hard tissues damaged by Pg.
Collapse
Affiliation(s)
- Xin Huang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, China
- Department of Periodontology, School & Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Junhong Xiao
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Huiyi Wang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Yan Peng
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Heyu Liu
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Li Ma
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, China
- Department of Periodontology, School & Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Xiaoxuan Wang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, China
- Department of Periodontology, School & Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Zhengguo Cao
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, China
- Department of Periodontology, School & Hospital of Stomatology, Wuhan University, Wuhan, China
| |
Collapse
|
18
|
Wei D, Tang L, Su L, Zeng S, Telushi A, Lang X, Zhang Y, Qin M, Qiu L, Zhong C, Yu J. Edgeworthia gardneri (Wall.) Meisn. extract protects against myocardial infarction by inhibiting NF-κB-and MAPK-mediated endothelial inflammation. Front Cardiovasc Med 2022; 9:1013013. [PMID: 36606274 PMCID: PMC9808090 DOI: 10.3389/fcvm.2022.1013013] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2022] [Accepted: 12/05/2022] [Indexed: 12/24/2022] Open
Abstract
Background Experimental and clinical evidence has demonstrated a pivotal role of inflammation in the pathogenesis of ischemic heart disease, and targeting inflammation has been shown to provide clinical benefits for patients with coronary disease. Endothelial cells constitute the majority of non-cardiomyocytes in the heart. Endothelial pro-inflammatory activation is recognized as a critical component in the pathophysiology of cardiovascular disease. The dried flowers of Edgeworthia gardneri (Wall.) Meisn. (EG) have been widely used as Tibetan folk medicine to ameliorate a range of metabolic disorders, such as diabetes mellitus, hyperlipidemia, hypertension, and obesity. However, its role in modulating endothelial inflammation and ischemic heart disease has not been evaluated. Methods and results Herein, using a preclinical rat model of coronary artery ligation-induced myocardial infarction (MI), we demonstrated that systemic administration of EG extract (EEEG) attenuated ischemic cardiac injury. EEEG reduced myocardial infarct size, improved cardiac function, and ameliorated adverse cardiac remodeling. Moreover, the cardioprotective effects of EEEG were associated with decreased MI-induced myocardial inflammation. Consistent with the anti-inflammatory role of EEEG in vivo, EEEG attenuated TNF-α-stimulated human umbilical vein endothelial cells (HUVECs) activation and monocyte-endothelial cell firm adhesion in vitro. Mechanistically, our data showed that EEEG's mode of action suppresses the activation of NF-κB, ERK, and p38 MAPK signaling pathways in ECs. Importantly, we demonstrated that EEEG inhibits endothelial inflammation in an NF-κB- and p38 MAPK-dependent manner using pharmacological inhibitors. Conclusion Collectively, this study identified EG as a potential therapeutic agent in attenuating endothelial inflammation and managing ischemic cardiovascular disease.
Collapse
Affiliation(s)
- Dan Wei
- Center for Translational Medicine, Jiangxi University of Chinese Medicine, Nanchang, Jiangxi, China
| | - Le Tang
- Center for Translational Medicine, Jiangxi University of Chinese Medicine, Nanchang, Jiangxi, China
| | - Lingqing Su
- Center for Translational Medicine, Jiangxi University of Chinese Medicine, Nanchang, Jiangxi, China
| | - Sufen Zeng
- Center for Translational Medicine, Jiangxi University of Chinese Medicine, Nanchang, Jiangxi, China
| | - Ajdora Telushi
- Department of Cardiovascular Sciences and Center for Metabolic Disease Research, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, United States
| | - Xiaoya Lang
- Center for Translational Medicine, Jiangxi University of Chinese Medicine, Nanchang, Jiangxi, China
| | - Yanli Zhang
- The National Pharmaceutical Engineering Center for Solid Preparation in Chinese Herbal Medicine, Jiangxi University of Chinese Medicine, Nanchang, Jiangxi, China
| | - Manman Qin
- Center for Translational Medicine, Jiangxi University of Chinese Medicine, Nanchang, Jiangxi, China
| | - Liang Qiu
- Center for Translational Medicine, Jiangxi University of Chinese Medicine, Nanchang, Jiangxi, China
| | - Chao Zhong
- Center for Translational Medicine, Jiangxi University of Chinese Medicine, Nanchang, Jiangxi, China,*Correspondence: Chao Zhong,
| | - Jun Yu
- Department of Cardiovascular Sciences and Center for Metabolic Disease Research, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, United States,Jun Yu,
| |
Collapse
|
19
|
Xie C, Li Z, Hua Y, Sun S, Zhong L, Chen Q, Feng H, Ji N, Li T, Zhou X, Zeng X, Tang Z, Sun C, Li J, Chen Q. Identification of a BRAF/PA28γ/MEK1 signaling axis and its role in epithelial-mesenchymal transition in oral submucous fibrosis. Cell Death Dis 2022; 13:701. [PMID: 35961969 PMCID: PMC9374740 DOI: 10.1038/s41419-022-05152-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2022] [Revised: 07/26/2022] [Accepted: 07/29/2022] [Indexed: 01/21/2023]
Abstract
Oral submucous fibrosis (OSF) is a chronic and insidious oral potentially malignant disorder associated with a 4-17% risk of oral squamous cell carcinoma (OSCC). Our previous study found that proteasomal activator 28 gamma (PA28γ) is frequently overexpressed in oral squamous cell carcinoma and negatively correlated with poor patient prognosis. However, the role of PA28γ in the occurrence and development of OSF remains unclear. Here, we screened PA28γ-related genes and investigated their function in OSF. We demonstrated that the expression of PA28γ was positively associated with MEK1 and gradually elevated from normal to progressive stages of OSF tissue. Arecoline, a pathogenic component of OSF, could upregulate the protein levels of PA28γ and phosphorylated MEK1 and contribute to epithelial to mesenchymal transition (EMT) in epithelial cells. Notably, PA28γ could interact with MEK1 and upregulate its phosphorylation level. Furthermore, arecoline upregulated BRAF, which can interact with PA28γ and upregulate its protein level. Additionally, BRAF, PA28γ, and MEK1 could form protein complexes and then enhance the MEK1/ERK signaling pathways. The concrete mechanism of the protein stability of PA28γ is that BRAF mediates its degradation by inhibiting its ubiquitination. These findings underscore the instrumental role of PA28γ in the BRAF/MEK1 pathway and enhanced EMT through MEK1/ERK activation in OSF.
Collapse
Affiliation(s)
- Changqing Xie
- grid.13291.380000 0001 0807 1581State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Chinese Academy of Medical Sciences Research Unit of Oral Carcinogenesis and Management, West China Hospital of Stom atology, Sichuan University, Chengdu, Sichuan 610041 People’s Republic of China ,grid.216417.70000 0001 0379 7164Department of Oral and Maxillofacial Surgery, Xiangya Stomatological Hospital & School of Stomatology, Postdoctoral Research Workstation, Cancer Research Institute and School of Basic Medicine, Central South University, Changsha, Hunan 410078 People’s Republic of China
| | - Zaiye Li
- grid.13291.380000 0001 0807 1581State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Chinese Academy of Medical Sciences Research Unit of Oral Carcinogenesis and Management, West China Hospital of Stom atology, Sichuan University, Chengdu, Sichuan 610041 People’s Republic of China
| | - Yufei Hua
- grid.13291.380000 0001 0807 1581State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Chinese Academy of Medical Sciences Research Unit of Oral Carcinogenesis and Management, West China Hospital of Stom atology, Sichuan University, Chengdu, Sichuan 610041 People’s Republic of China
| | - Silu Sun
- grid.13291.380000 0001 0807 1581State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Chinese Academy of Medical Sciences Research Unit of Oral Carcinogenesis and Management, West China Hospital of Stom atology, Sichuan University, Chengdu, Sichuan 610041 People’s Republic of China
| | - Liang Zhong
- grid.13291.380000 0001 0807 1581State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Chinese Academy of Medical Sciences Research Unit of Oral Carcinogenesis and Management, West China Hospital of Stom atology, Sichuan University, Chengdu, Sichuan 610041 People’s Republic of China
| | - Qian Chen
- grid.13291.380000 0001 0807 1581State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Chinese Academy of Medical Sciences Research Unit of Oral Carcinogenesis and Management, West China Hospital of Stom atology, Sichuan University, Chengdu, Sichuan 610041 People’s Republic of China
| | - Hui Feng
- grid.13291.380000 0001 0807 1581State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Chinese Academy of Medical Sciences Research Unit of Oral Carcinogenesis and Management, West China Hospital of Stom atology, Sichuan University, Chengdu, Sichuan 610041 People’s Republic of China ,grid.216417.70000 0001 0379 7164Department of Oral and Maxillofacial Surgery, Xiangya Stomatological Hospital & School of Stomatology, Postdoctoral Research Workstation, Cancer Research Institute and School of Basic Medicine, Central South University, Changsha, Hunan 410078 People’s Republic of China
| | - Ning Ji
- grid.13291.380000 0001 0807 1581State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Chinese Academy of Medical Sciences Research Unit of Oral Carcinogenesis and Management, West China Hospital of Stom atology, Sichuan University, Chengdu, Sichuan 610041 People’s Republic of China
| | - Taiwen Li
- grid.13291.380000 0001 0807 1581State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Chinese Academy of Medical Sciences Research Unit of Oral Carcinogenesis and Management, West China Hospital of Stom atology, Sichuan University, Chengdu, Sichuan 610041 People’s Republic of China
| | - Xikun Zhou
- grid.13291.380000 0001 0807 1581State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, Sichuan 610041 People’s Republic of China
| | - Xin Zeng
- grid.13291.380000 0001 0807 1581State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Chinese Academy of Medical Sciences Research Unit of Oral Carcinogenesis and Management, West China Hospital of Stom atology, Sichuan University, Chengdu, Sichuan 610041 People’s Republic of China
| | - Zhangui Tang
- grid.216417.70000 0001 0379 7164Department of Oral and Maxillofacial Surgery, Xiangya Stomatological Hospital & School of Stomatology, Postdoctoral Research Workstation, Cancer Research Institute and School of Basic Medicine, Central South University, Changsha, Hunan 410078 People’s Republic of China
| | - Chongkui Sun
- grid.13291.380000 0001 0807 1581State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Chinese Academy of Medical Sciences Research Unit of Oral Carcinogenesis and Management, West China Hospital of Stom atology, Sichuan University, Chengdu, Sichuan 610041 People’s Republic of China
| | - Jing Li
- grid.13291.380000 0001 0807 1581State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Chinese Academy of Medical Sciences Research Unit of Oral Carcinogenesis and Management, West China Hospital of Stom atology, Sichuan University, Chengdu, Sichuan 610041 People’s Republic of China
| | - Qianming Chen
- grid.13291.380000 0001 0807 1581State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Chinese Academy of Medical Sciences Research Unit of Oral Carcinogenesis and Management, West China Hospital of Stom atology, Sichuan University, Chengdu, Sichuan 610041 People’s Republic of China
| |
Collapse
|
20
|
Regulation of Life & Death by REGγ. Cells 2022; 11:cells11152281. [PMID: 35892577 PMCID: PMC9330691 DOI: 10.3390/cells11152281] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Revised: 07/17/2022] [Accepted: 07/20/2022] [Indexed: 01/27/2023] Open
Abstract
REGγ, a proteasome activator belonging to the 11S (otherwise known as REG, PA28, or PSME) proteasome activator family, is widely present in many eukaryotes. By binding to the 20S catalytic core particle, REGγ acts as a molecular sieve to selectively target proteins for degradation in an ATP- and ubiquitin-independent manner. This non-canonical proteasome pathway directly regulates seemingly unrelated cellular processes including cell growth and proliferation, apoptosis, DNA damage response, immune response, and metabolism. By affecting different pathways, REGγ plays a vital role in the regulation of cellular life and death through the maintenance of protein homeostasis. As a promoter of cellular growth and a key regulator of several tumor suppressors, many recent studies have linked REGγ overexpression with tumor formation and suggested the REGγ-proteasome as a potential target of new cancer-drug development. This review will present an overview of the major functions of REGγ as it relates to the regulation of cellular life and death, along with new mechanistic insights into the regulation of REGγ.
Collapse
|
21
|
Miao G, Zhao X, Chan SL, Zhang L, Li Y, Zhang Y, Zhang L, Wang B. Vascular smooth muscle cell c-Fos is critical for foam cell formation and atherosclerosis. Metabolism 2022; 132:155213. [PMID: 35513168 DOI: 10.1016/j.metabol.2022.155213] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Revised: 04/19/2022] [Accepted: 04/25/2022] [Indexed: 10/18/2022]
Abstract
BACKGROUND Hyperlipidemia-induced vascular smooth muscle cell (VSMC)-derived foam cell formation is considered a crucial event in the development of atherosclerosis. Since c-Fos emerges as a key modulator of lipid metabolism, we investigated whether c-Fos plays a role in hyperlipidemia-induced VSMC-derived foam cell formation and atherosclerosis. APPROACH AND RESULTS c-Fos expression was observed in VSMCs in atherosclerotic plaques from patients and western diet-fed atherosclerosis-prone LDLR-/- and ApoE-/- mice by immunofluorescence staining. To ascertain c-Fos's function in atherosclerosis development, VSMC-specific c-Fos deficient mice in ApoE-/- background were established. Western diet-fed c-FosVSMCKOApoE-/- mice exhibited a significant reduction of atherosclerotic lesion formation as measured by hematoxylin and eosin staining, accompanied by decreased lipid deposition within aortic roots as determined by Oil red O staining. Primary rat VSMCs were isolated to examine the role of c-Fos in lipid uptake and foam cell formation. oxLDL stimulation resulted in VSMC-derived foam cell formation and elevated intracellular mitochondrial reactive oxygen species (mtROS), c-Fos and LOX-1 levels, whereas specific inhibition of mtROS, c-Fos or LOX-1 lessened lipid accumulation in oxLDL-stimulated VSMCs. Mechanistically, oxLDL acts through mtROS to enhance transcription activity of c-Fos to facilitate the expression of LOX-1, exerting a feedforward mechanism with oxLDL to increase lipid uptake and propel VSMC-derived foam cell formation and atherogenesis. CONCLUSION Our study demonstrates a fundamental role of mtROS/c-Fos/LOX-1 signaling pathway in promoting oxLDL uptake and VSMC-derived foam cell formation during atherosclerosis. c-Fos may represent a promising therapeutic target amenable to clinical translation in the future.
Collapse
Affiliation(s)
- Guolin Miao
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China; Institute of Cardiovascular Sciences and Key Laboratory of Molecular Cardiovascular Sciences, Ministry of Education, Peking University, Beijing, China
| | - Xi Zhao
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China; State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, China
| | - Siu-Lung Chan
- Vascular Biology Program, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Lijun Zhang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Yaohua Li
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, China
| | - Yuke Zhang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Lijun Zhang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China.
| | - Beibei Wang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China.
| |
Collapse
|
22
|
Huang X, Ma L, Wang X, Wang H, Peng Y, Gao X, Huang H, Chen Y, Zhang Y, Cao Z. Ckip-1 Mediates P. gingivalis-Suppressed Cementoblast Mineralization. J Dent Res 2022; 101:599-608. [PMID: 34875910 DOI: 10.1177/00220345211054744] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Porphyromonas gingivalis is responsible for the destruction of cementum in patients with periodontitis and periapical periodontitis. However, research about the effects of P. gingivalis on cementoblast mineralization and the underlying mechanism is still lacking. Casein kinase 2 interacting protein 1 (Ckip-1) is a scaffold protein that interacts with various proteins and signals to regulate different cell functions, such as cell morphology, apoptosis, and differentiation. In this study, we verified the suppressive effects of P. gingivalis and lipopolysaccharide (Pg-LPS) on OCCM-30 mineralization. We also showed that Ckip-1 gradually decreased during OCCM-30 mineralization but increased with the aggravation of Pg-induced inflammation. However, it remained unchanged when cells were stimulated with Pg-LPS, regardless of the concentration and incubation time. Then, more cellular cementum and enhanced Osterix expression were observed in Ckip-1 knockout mice when compared with the wild-type mice. Meanwhile, Ckip-1 silencing significantly enhanced cementoblast mineralization with or without P. gingivalis-associated inflammation. The trend was opposite when Ckip-1 was overexpressed. Finally, we found that the p38, Akt, and Wnt pathways were activated, while the Erk1/2 pathway was inhibited when Ckip-1 was silenced. The opposite results were also observed in the Ckip-1 overexpression group. Furthermore, we proved that cell mineralization was weakened when p38, Akt inhibitors were applied and strengthened when the Erk1/2 pathway was inhibited. In summary, Ckip-1 is upregulated underP. gingivalis-induced inflammation and negatively regulates cementoblast mineralization partially through mitogen-activated protein kinases and Akt signaling pathways, which may contribute to the restoration of cementum destroyed by P. gingivalis.
Collapse
Affiliation(s)
- X Huang
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST KLOS) & Key Laboratory of Oral Biomedicine Ministry of Education (KLOBME), School & Hospital of Stomatology, Wuhan University, Wuhan, China
| | - L Ma
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST KLOS) & Key Laboratory of Oral Biomedicine Ministry of Education (KLOBME), School & Hospital of Stomatology, Wuhan University, Wuhan, China
| | - X Wang
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST KLOS) & Key Laboratory of Oral Biomedicine Ministry of Education (KLOBME), School & Hospital of Stomatology, Wuhan University, Wuhan, China
- Department of Periodontology, School & Hospital of Stomatology, Wuhan University, Wuhan, China
| | - H Wang
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST KLOS) & Key Laboratory of Oral Biomedicine Ministry of Education (KLOBME), School & Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Y Peng
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST KLOS) & Key Laboratory of Oral Biomedicine Ministry of Education (KLOBME), School & Hospital of Stomatology, Wuhan University, Wuhan, China
| | - X Gao
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST KLOS) & Key Laboratory of Oral Biomedicine Ministry of Education (KLOBME), School & Hospital of Stomatology, Wuhan University, Wuhan, China
| | - H Huang
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST KLOS) & Key Laboratory of Oral Biomedicine Ministry of Education (KLOBME), School & Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Y Chen
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST KLOS) & Key Laboratory of Oral Biomedicine Ministry of Education (KLOBME), School & Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Y Zhang
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST KLOS) & Key Laboratory of Oral Biomedicine Ministry of Education (KLOBME), School & Hospital of Stomatology, Wuhan University, Wuhan, China
- Medical Research Institute, School of Medicine, Wuhan University, Wuhan, China
| | - Z Cao
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST KLOS) & Key Laboratory of Oral Biomedicine Ministry of Education (KLOBME), School & Hospital of Stomatology, Wuhan University, Wuhan, China
- Department of Periodontology, School & Hospital of Stomatology, Wuhan University, Wuhan, China
| |
Collapse
|
23
|
Wu D, Hu Q, Wang Y, Jin M, Tao Z, Wan J. Identification of HMOX1 as a Critical Ferroptosis-Related Gene in Atherosclerosis. Front Cardiovasc Med 2022; 9:833642. [PMID: 35498043 PMCID: PMC9046663 DOI: 10.3389/fcvm.2022.833642] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Accepted: 03/11/2022] [Indexed: 01/12/2023] Open
Abstract
Ferroptosis is a novel form of programmed iron-dependent cell death. The ferroptosis-related genes (FRGs) have been recognized as biomarkers for cancers. Increasing evidence has indicated that ferroptosis is involved in the process of atherosclerosis. However, the potential FRGs used for the diagnosis, prognosis and therapy for atherosclerosis are still unclear. We aimed to identify the ferroptosis-related differentially expressed genes (DEGs) of atherosclerosis. We downloaded the mRNA-sequencing data of patients with atherosclerosis from the Gene Expression Omnibus (GEO) database. HMOX1 was identified as an essential ferroptosis-related DEG by bioinformatic analysis of the GSE28829 and GSE43292 datasets. The pro-ferroptotic effect of HMOX1 was validated through cell experiments. Then we conducted a single-gene analysis of HMOX1 and found that high-expression of HMOX1 in atherosclerotic plaques was accompanied by matrix metalloproteinases (MMPs) producing and M0 macrophages infiltration. Taken together, our present study suggested HMOX1 as a potential diagnostic biomarker for atherosclerosis and provided more evidence about the vital role of ferroptosis in atherosclerosis progression.
Collapse
Affiliation(s)
- Daiqian Wu
- Department of Cardiology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Qian Hu
- Department of Gastroenterology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Yuqing Wang
- School of Basic Medical Sciences, Wuhan University, Wuhan, China
| | - Mengying Jin
- Department of Cardiology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Ziqi Tao
- Department of Cardiology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Jing Wan
- Department of Cardiology, Zhongnan Hospital of Wuhan University, Wuhan, China
- *Correspondence: Jing Wan
| |
Collapse
|
24
|
Gui Y, Zheng H, Cao RY. Foam Cells in Atherosclerosis: Novel Insights Into Its Origins, Consequences, and Molecular Mechanisms. Front Cardiovasc Med 2022; 9:845942. [PMID: 35498045 PMCID: PMC9043520 DOI: 10.3389/fcvm.2022.845942] [Citation(s) in RCA: 104] [Impact Index Per Article: 34.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Accepted: 03/17/2022] [Indexed: 12/12/2022] Open
Abstract
Foam cells play a vital role in the initiation and development of atherosclerosis. This review aims to summarize the novel insights into the origins, consequences, and molecular mechanisms of foam cells in atherosclerotic plaques. Foam cells are originated from monocytes as well as from vascular smooth muscle cells (VSMC), stem/progenitor cells, and endothelium cells. Novel technologies including lineage tracing and single-cell RNA sequencing (scRNA-seq) have revolutionized our understanding of subtypes of monocyte- and VSMC-derived foam cells. By using scRNA-seq, three main clusters including resident-like, inflammatory, and triggering receptor expressed on myeloid cells-2 (Trem2 hi ) are identified as the major subtypes of monocyte-derived foam cells in atherosclerotic plaques. Foam cells undergo diverse pathways of programmed cell death including apoptosis, autophagy, necroptosis, and pyroptosis, contributing to the necrotic cores of atherosclerotic plaques. The formation of foam cells is affected by cholesterol uptake, efflux, and esterification. Novel mechanisms including nuclear receptors, non-coding RNAs, and gut microbiota have been discovered and investigated. Although the heterogeneity of monocytes and the complexity of non-coding RNAs make obstacles for targeting foam cells, further in-depth research and therapeutic exploration are needed for the better management of atherosclerosis.
Collapse
Affiliation(s)
- Yuzhou Gui
- Shanghai Xuhui Central Hospital, Zhongshan-Xuhui Hospital, Fudan University, Shanghai, China
- Shanghai Engineering Research Center of Phase I Clinical Research and Quality Consistency Evaluation for Drugs, Shanghai, China
| | - Hongchao Zheng
- Department of Cardiovascular, Shanghai Xuhui Central Hospital, Zhongshan-Xuhui Hospital, Fudan University, Shanghai, China
| | - Richard Y. Cao
- Department of Cardiovascular, Shanghai Xuhui Central Hospital, Zhongshan-Xuhui Hospital, Fudan University, Shanghai, China
| |
Collapse
|
25
|
Li L, Liu S, Tan J, Wei L, Wu D, Gao S, Weng Y, Chen J. Recent advance in treatment of atherosclerosis: Key targets and plaque-positioned delivery strategies. J Tissue Eng 2022; 13:20417314221088509. [PMID: 35356091 PMCID: PMC8958685 DOI: 10.1177/20417314221088509] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Atherosclerosis, a chronic inflammatory disease of vascular wall, is a progressive pathophysiological process with lipids oxidation/depositing initiation and innate/adaptive immune responses. The coordination of multi systems covering oxidative stress, dysfunctional endothelium, diseased lipid uptake, cell apoptosis, thrombotic and pro-inflammatory responding as well as switched SMCs contributes to plaque growth. In this circumstance, inevitably, targeting these processes is considered to be effective for treating atherosclerosis. Arriving, retention and working of payload candidates mediated by targets in lesion direct ultimate therapeutic outcomes. Accumulating a series of scientific studies and clinical practice in the past decades, lesion homing delivery strategies including stent/balloon/nanoparticle-based transportation worked as the potent promotor to ensure a therapeutic effect. The objective of this review is to achieve a very brief summary about the effective therapeutic methods cooperating specifical targets and positioning-delivery strategies in atherosclerosis for better outcomes.
Collapse
Affiliation(s)
- Li Li
- Key Laboratory of Advanced Technology of Materials, Ministry of Education, Southwest Jiaotong University, Chengdu, PR China
| | - Sainan Liu
- Key Laboratory of Advanced Technology of Materials, Ministry of Education, Southwest Jiaotong University, Chengdu, PR China
| | - Jianying Tan
- Key Laboratory of Advanced Technology of Materials, Ministry of Education, Southwest Jiaotong University, Chengdu, PR China
| | - Lai Wei
- Key Laboratory of Advanced Technology of Materials, Ministry of Education, Southwest Jiaotong University, Chengdu, PR China
| | - Dimeng Wu
- Chengdu Daxan Innovative Medical Tech. Co., Ltd., Chengdu, PR China
| | - Shuai Gao
- Chengdu Daxan Innovative Medical Tech. Co., Ltd., Chengdu, PR China
| | - Yajun Weng
- Key Laboratory of Advanced Technology of Materials, Ministry of Education, Southwest Jiaotong University, Chengdu, PR China
| | - Junying Chen
- Key Laboratory of Advanced Technology of Materials, Ministry of Education, Southwest Jiaotong University, Chengdu, PR China
| |
Collapse
|
26
|
Bezhaeva T, Karper J, Quax PHA, de Vries MR. The Intriguing Role of TLR Accessory Molecules in Cardiovascular Health and Disease. Front Cardiovasc Med 2022; 9:820962. [PMID: 35237675 PMCID: PMC8884272 DOI: 10.3389/fcvm.2022.820962] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Accepted: 01/19/2022] [Indexed: 12/12/2022] Open
Abstract
Activation of Toll like receptors (TLR) plays an important role in cardiovascular disease development, progression and outcomes. Complex TLR mediated signaling affects vascular and cardiac function including tissue remodeling and repair. Being central components of both innate and adaptive arms of the immune system, TLRs interact as pattern recognition receptors with a series of exogenous ligands and endogenous molecules or so-called danger associated molecular patterns (DAMPs) that are released upon tissue injury and cellular stress. Besides immune cells, a number of structural cells within the cardiovascular system, including endothelial cells, smooth muscle cells, fibroblasts and cardiac myocytes express TLRs and are able to release or sense DAMPs. Local activation of TLR-mediated signaling cascade induces cardiovascular tissue repair but in a presence of constant stimuli can overshoot and cause chronic inflammation and tissue damage. TLR accessory molecules are essential in guiding and dampening these responses toward an adequate reaction. Furthermore, accessory molecules assure specific and exclusive TLR-mediated signal transduction for distinct cells and pathways involved in the pathogenesis of cardiovascular diseases. Although much has been learned about TLRs activation in cardiovascular remodeling, the exact role of TLR accessory molecules is not entirely understood. Deeper understanding of the role of TLR accessory molecules in cardiovascular system may open therapeutic avenues aiming at manipulation of inflammatory response in cardiovascular disease. The present review outlines accessory molecules for membrane TLRs that are involved in cardiovascular disease progression. We first summarize the up-to-date knowledge on TLR signaling focusing on membrane TLRs and their ligands that play a key role in cardiovascular system. We then survey the current evidence of the contribution of TLRs accessory molecules in vascular and cardiac remodeling including myocardial infarction, heart failure, stroke, atherosclerosis, vein graft disease and arterio-venous fistula failure.
Collapse
Affiliation(s)
- Taisiya Bezhaeva
- Department of Surgery and Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, Netherlands
| | - Jacco Karper
- Department of Cardiology, Wilhelmina Hospital Assen, Assen, Netherlands
| | - Paul H. A. Quax
- Department of Surgery and Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, Netherlands
| | - Margreet R. de Vries
- Department of Surgery and Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, Netherlands
- *Correspondence: Margreet R. de Vries
| |
Collapse
|
27
|
Gao F, Bai R, Qin W, Liang B, Yang Z, Yang H. Angiotensin II induces the expression of periostin to promote foam cell formation in oxLDL-treated macrophages. Int J Cardiol 2022; 347:46-53. [PMID: 34793856 DOI: 10.1016/j.ijcard.2021.11.009] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Revised: 09/21/2021] [Accepted: 11/07/2021] [Indexed: 11/05/2022]
Abstract
A matricellular protein periostin has been documented to promote macrophage recruitment in atherosclerotic lesions. However, the role of periostin in macrophage foam cell formation is still unknown. In this study, we examined the expression and function of periostin in cholesterol homeostasis in macrophages. The role of periostin in mediating Ang II-induced foam cell formation was also investigated. The mechanism by which Ang II induced the expression of periostin was explored. It was found that oxLDL treatment significantly increased the expression and secretion of periostin in THP-1 macrophages. Knockdown of periostin blocked oxLDL-induced lipid accumulation and enhanced cholesterol efflux. In contrast, treatment with recombinant periostin protein enhanced oxLDL-induced macrophage foam cell formation. Ang II caused a time-dependent induction of periostin in THP-1 macrophages, which was ascribed to Twist2-mediated transactivation of periostin. Ang II treatment significantly augmented lipid accumulation in THP-1 macrophages, and knockdown of periostin blocked the effect of Ang II on foam cell formation. Moreover, periostin depletion restored cholesterol efflux in Ang II-treated THP-1 macrophages. Clinically, there was a significant positive correlation between serum periostin and Ang II levels in patients with atherosclerosis. Collectively, we show that periostin is essential for Ang II-induced enhancement of macrophage foam cell formation via suppression of cholesterol efflux.
Collapse
Affiliation(s)
- Fen Gao
- Departments of Cardiology, The Second Hospital of Shanxi Medical University, Taiyuan, China
| | - Rui Bai
- Departments of Cardiology, The Second Hospital of Shanxi Medical University, Taiyuan, China
| | - Weiwei Qin
- Departments of Cardiology, The Second Hospital of Shanxi Medical University, Taiyuan, China
| | - Bin Liang
- Departments of Cardiology, The Second Hospital of Shanxi Medical University, Taiyuan, China
| | - Zhiming Yang
- Departments of Cardiology, The Second Hospital of Shanxi Medical University, Taiyuan, China
| | - Huiyu Yang
- Departments of Cardiology, The Second Hospital of Shanxi Medical University, Taiyuan, China.
| |
Collapse
|
28
|
Zhu B, Zhang Z, Wang X, Zhang W, Shi H, Song Z, Ding S, Yang X. Abnormal histidine metabolism promotes macrophage lipid accumulation under Ox-LDL condition. Biochem Biophys Res Commun 2022; 588:161-167. [PMID: 34954523 DOI: 10.1016/j.bbrc.2021.12.069] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Revised: 12/17/2021] [Accepted: 12/18/2021] [Indexed: 11/02/2022]
Abstract
Distinct macrophage populations exert highly heterogeneity and perform various functions, among which, a crucial function of lipid metabolism is highlighted. However, the role of histidine metabolism disorder in macrophage lipid metabolism remains elusive. Addressed this question, we sorted and cultured the bone marrow-derived macrophages (BMDMs) of histidine decarboxylase (Hdc) knockout (Hdc-/-) mice with an in vitro oxidized low-density lipoprotein (ox-LDL) model, and detected the intracellular lipids by Oil Red O staining as well as lipid probe staining. Astemizole, a canonical and long-acting histamine H1 receptor (H1R) antagonist, was applied to elucidate the impact of antagonizing the H1R-dependent signaling pathway on macrophage lipid metabolism. Subsequently, the differential expressed genes were screened and analyzed in the bone marrow-derived CD11b+ immature myeloid cells of Hdc-/- and Hdc+/+ mice with a high fat diet by the microarray study. The expression levels of cholesterol metabolism-related genes were examined by qRT-PCR to explore underlying mechanisms. Lastly, we used a high-sensitivity histidine probe to detect the intracellular histidine in the BMDMs after oxidative stress. The results revealed that histidine metabolism disorder and histamine deficiency aggravated lipid accumulation in the ox-LDL-treated BMDMs. The expression level of H1R gene in the BMDMs was down-regulated after ox-LDL stimulation. The disruption of the H1R-dependent signaling pathway by astemizole further exacerbated ox-LDL-induced lipid deposition in the BMDMs partly by up-regulating scavenger receptor class A (SR-A) for lipid intake, down-regulating neutral cholesteryl ester hydrolase (nCEH) for cholesterol esterification and down-regulating ATP-binding cassette transporters A1 (ABCA1) and ABCG1 for reverse cholesterol transport. The intracellular histidine increased under ox-LDL condition, which was further increased by Hdc knockout. Collectively, these results partially reveal the relationship between histidine metabolism and lipid metabolism in the BMDMs and offer a novel strategy for lipid metabolism disorder-associated diseases.
Collapse
Affiliation(s)
- Baoling Zhu
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Institutes of Biomedical Sciences, Fudan University, Shanghai, 200032, China; Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Zhiwei Zhang
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Institutes of Biomedical Sciences, Fudan University, Shanghai, 200032, China; Reproductive Medicine Center, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Xiangfei Wang
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Institutes of Biomedical Sciences, Fudan University, Shanghai, 200032, China; Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Weiwei Zhang
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Institutes of Biomedical Sciences, Fudan University, Shanghai, 200032, China
| | - Hongyu Shi
- Department of Cardiology, Wusong Hospital of Zhongshan Hospital, Fudan University, Shanghai, 200940, China
| | - Zhifeng Song
- Department of Oral Mucosa and Periodontitis, Shanghai Stomatological Hospital, Fudan University, Shanghai, 200433, China
| | - Suling Ding
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Institutes of Biomedical Sciences, Fudan University, Shanghai, 200032, China.
| | - Xiangdong Yang
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Institutes of Biomedical Sciences, Fudan University, Shanghai, 200032, China; Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China; Department of Cardiology, Wusong Hospital of Zhongshan Hospital, Fudan University, Shanghai, 200940, China.
| |
Collapse
|
29
|
Li BW, Liu Y, Zhang L, Guo XQ, Wen C, Zhang F, Luo XY, Xia YP. Cytotoxin-associated gene A (CagA) promotes aortic endothelial inflammation and accelerates atherosclerosis through the NLRP3/caspase-1/IL-1β axis. FASEB J 2021; 35:e21942. [PMID: 34670018 DOI: 10.1096/fj.202100695rr] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Revised: 08/18/2021] [Accepted: 09/07/2021] [Indexed: 11/11/2022]
Abstract
Atherosclerosis is a chronic inflammatory disease. Pathophysiological similarities between chronic infections and atherosclerosis triggered interests between these conditions. The seroepidemiological study showed that Helicobacter pylori strains that express cytotoxin-associated gene A (CagA), an oncoprotein and a major virulence factor, was positively correlated with atherosclerosis and related clinical events. Nevertheless, the underlying mechanism is poorly understood. In this study, the seroprevalence of infection by H. pylori and by strains express CagA assessed by enzyme-linked immunosorbent assay (ELISA) showed that the prevalence of CagA strains rather than H. pylori in patients was positively correlated with atherogenesis. Correspondingly, we found that CagA augmented the growth of plaque of ApoE-/- mice in the early stage of atherosclerosis and promoted the expression of adhesion molecules and inflammatory cytokines in mouse aortic endothelial cells (MAECs). Mechanistically, both si-NLRP3 and si-IL-1β mitigated the promoting effect of CagA on the inflammatory activation of HAECs. In vivo, the inhibition of NLRP3 by MCC950 significantly attenuated the promoting effect of CagA on plaque growth of ApoE-/- mice. We also propose NLRP3 as a potential therapeutic target for CagA-positive H. pylori infection-related atherosclerosis and emphasize the importance of inflammation in atherosclerosis pathology.
Collapse
Affiliation(s)
- Bo-Wei Li
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yu Liu
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Lei Zhang
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiao-Qing Guo
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Cheng Wen
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Feng Zhang
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xue-Ying Luo
- Affiliated Wuhan Mental Health Center, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yuan-Peng Xia
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
30
|
Niu Q, Shen S, He J, Wang L. CKIP-1 contributes to osteogenic differentiation of mouse bone marrow mesenchymal stem cells. Regen Med 2021; 16:847-859. [PMID: 34498492 DOI: 10.2217/rme-2020-0119] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Background: Osteogenesis greatly depends on the differentiation of bone marrow mesenchymal stem cells (BMSCs). CKIP-1 is considered to be a negative regulator of BMSCs. Methods: We established a CKIP-1 knockout mouse model, then isolated and cultured BMSCs from wild-type and knockout groups. Results: Our data demonstrated that CKIP-1 knockout significantly increased bone structure in the experimental mouse model and enhanced BMSC proliferation. CKIP-1 knockout contributed to osteoblastic and adipogenic differentiation. Furthermore, CKIP-1 regulated osteogenesis in BMSCs via the MAPK signaling pathway, and BMSCs from the CKIP-1 knockout mice were effective in repairing the skull defect null mice. Conclusion: Our results concluded that silencing of CKIP-1 promoted osteogenesis in experimental mice and increased BMSCs differentiation via upregulation of the MAPK signaling pathway.
Collapse
Affiliation(s)
- Qiannan Niu
- State Key Laboratory of Military Stomatology, National Clinical Research Center for Oral Diseases, Shaanxi International Joint Research Center for Oral Diseases, Department of Orthodontics, The Hospital of Stomatology, The Fourth Military Medical University, No.145 West Changle Road, Xi'an, 710000, Shaanxi, China
| | - Shuning Shen
- Department of Stomatology, No.984 Hospital of PLA, Beijing, 100094, China
| | - Jiaojiao He
- State Key Laboratory of Military Stomatology, National Clinical Research Center for Oral Diseases, Shaanxi International Joint Research Center for Oral Diseases, Department of Orthodontics, The Hospital of Stomatology, The Fourth Military Medical University, No.145 West Changle Road, Xi'an, 710000, Shaanxi, China
| | - Lei Wang
- State Key Laboratory of Military Stomatology, National Clinical Research Center for Oral Diseases, Shaanxi International Joint Research Center for Oral Diseases, Department of Orthodontics, The Hospital of Stomatology, The Fourth Military Medical University, No.145 West Changle Road, Xi'an, 710000, Shaanxi, China
| |
Collapse
|
31
|
Zhang M, Liu J, Gao R, Hu Y, Lu L, Liu C, Ai L, Pan J, Tian L, Fan J. Interleukin-36γ aggravates macrophage foam cell formation and atherosclerosis progression in ApoE knockout mice. Cytokine 2021; 146:155630. [PMID: 34246054 DOI: 10.1016/j.cyto.2021.155630] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Revised: 06/17/2021] [Accepted: 06/18/2021] [Indexed: 12/13/2022]
Abstract
Atherosclerosis-related cardiovascular diseases are the leading cause of mortality worldwide. Macrophage-derived foam cell formation is a critical early event in atherogenesis. However, the molecular pathways involved in this disease have not been fully elucidated. Interleukin (IL)-36 plays a crucial role in inflammation, and this study was conducted to investigate the possible role of IL-36γ in the pathogenesis and regulation of atherosclerosis. In this study, we show that IL-36γ regulates inflammatory responses and lipoprotein metabolic processes in macrophages and exerts its atherosclerosis-promoting effects by increasing macrophage foam cell formation and uptake of oxidized low-density lipoproteins. Mechanistically, IL-36γ specifically upregulates expression of the scavenger receptor CD36 through the phosphoinositide 3-kinase pathway in macrophages. These results contribute to our understanding of IL-36γ as a novel regulator of foam cell formation and atherogenesis progression.
Collapse
Affiliation(s)
- Minghua Zhang
- Clinical Pharmacy Laboratory, Chinese PLA General Hospital, Beijing 100853, China
| | - Jing Liu
- Institute of Geriatrics, National Clinical Research Center of Geriatrics Disease, Second Medical Center of Chinese PLA General Hospital, Beijing 100853, China
| | - Rong Gao
- Air Force Medical Center, PLA, Beijing 100142, China
| | - Yazhuo Hu
- Institute of Geriatrics, National Clinical Research Center of Geriatrics Disease, Second Medical Center of Chinese PLA General Hospital, Beijing 100853, China
| | - Li Lu
- Department of General Surgery, Shanghai Children's Hospital, Shanghai Jiaotong University, Shanghai 200040, China
| | - Chuanbin Liu
- Institute of Geriatrics, National Clinical Research Center of Geriatrics Disease, Second Medical Center of Chinese PLA General Hospital, Beijing 100853, China
| | - Lunna Ai
- Institute of Geriatrics, National Clinical Research Center of Geriatrics Disease, Second Medical Center of Chinese PLA General Hospital, Beijing 100853, China
| | - Jingkun Pan
- Institute of Geriatrics, National Clinical Research Center of Geriatrics Disease, Second Medical Center of Chinese PLA General Hospital, Beijing 100853, China
| | - Lei Tian
- Institute of Geriatrics, National Clinical Research Center of Geriatrics Disease, Second Medical Center of Chinese PLA General Hospital, Beijing 100853, China
| | - Jiao Fan
- Institute of Geriatrics, National Clinical Research Center of Geriatrics Disease, Second Medical Center of Chinese PLA General Hospital, Beijing 100853, China.
| |
Collapse
|
32
|
Zhao Y, Ling S, Zhong G, Li Y, Li J, Du R, Jin X, Zhao D, Liu Z, Kan G, Chang YZ, Li Y. Casein Kinase-2 Interacting Protein-1 Regulates Physiological Cardiac Hypertrophy via Inhibition of Histone Deacetylase 4 Phosphorylation. Front Physiol 2021; 12:678863. [PMID: 34211403 PMCID: PMC8239235 DOI: 10.3389/fphys.2021.678863] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Accepted: 05/06/2021] [Indexed: 11/14/2022] Open
Abstract
Different kinds of mechanical stimuli acting on the heart lead to different myocardial phenotypes. Physiological stress, such as exercise, leads to adaptive cardiac hypertrophy, which is characterized by a normal cardiac structure and improved cardiac function. Pathological stress, such as sustained cardiac pressure overload, causes maladaptive cardiac remodeling and, eventually, heart failure. Casein kinase-2 interacting protein-1 (CKIP-1) is an important regulator of pathological cardiac remodeling. However, the role of CKIP-1 in physiological cardiac hypertrophy is unknown. We subjected wild-type (WT) mice to a swimming exercise program for 21 days, which caused an increase in myocardial CKIP-1 protein and mRNA expression. We then subjected CKIP-1 knockout (KO) mice and myocardial-specific CKIP-1-overexpressing mice to the 21-day swimming exercise program. Histological and echocardiography analyses revealed that CKIP-1 KO mice underwent pathological cardiac remodeling after swimming, whereas the CKIP-1-overexpressing mice had a similar cardiac phenotype to the WT controls. Histone deacetylase 4 (HDAC4) is a key molecule in the signaling cascade associated with pathological hypertrophy; the phosphorylation levels of HDAC4 were markedly higher in CKIP-1 KO mouse hearts after the swimming exercise program. The phosphorylation levels of HDAC4 did not change after swimming in the hearts of CKIP-1-overexpressing or WT mice. Our results indicate that swimming, a mechanical stress that leads to physiological hypertrophy, triggers pathological cardiac remodeling in CKIP-1 KO mice. CKIP-1 is necessary for physiological cardiac hypertrophy in vivo, and for modulating the phosphorylation level of HDAC4 after physiological stress. Genetically engineering CKIP-1 expression affected heart health in response to exercise.
Collapse
Affiliation(s)
- Yinlong Zhao
- Key Laboratory of Molecular and Cellular Biology of Ministry of Education, College of Life Science, Hebei Normal University, Shijiazhuang, China.,State Key Laboratory of Space Medicine Fundamentals and Application, China Astronaut Research and Training Center, Beijing, China
| | - Shukuan Ling
- State Key Laboratory of Space Medicine Fundamentals and Application, China Astronaut Research and Training Center, Beijing, China
| | - Guohui Zhong
- State Key Laboratory of Space Medicine Fundamentals and Application, China Astronaut Research and Training Center, Beijing, China.,School of Aerospace Medicine, Fourth Military Medical University, Xi'an, China
| | - Yuheng Li
- State Key Laboratory of Space Medicine Fundamentals and Application, China Astronaut Research and Training Center, Beijing, China
| | - Jianwei Li
- State Key Laboratory of Space Medicine Fundamentals and Application, China Astronaut Research and Training Center, Beijing, China
| | - Ruikai Du
- State Key Laboratory of Space Medicine Fundamentals and Application, China Astronaut Research and Training Center, Beijing, China
| | - Xiaoyan Jin
- State Key Laboratory of Space Medicine Fundamentals and Application, China Astronaut Research and Training Center, Beijing, China
| | - Dingsheng Zhao
- State Key Laboratory of Space Medicine Fundamentals and Application, China Astronaut Research and Training Center, Beijing, China
| | - Zizhong Liu
- State Key Laboratory of Space Medicine Fundamentals and Application, China Astronaut Research and Training Center, Beijing, China
| | - Guanghan Kan
- State Key Laboratory of Space Medicine Fundamentals and Application, China Astronaut Research and Training Center, Beijing, China
| | - Yan-Zhong Chang
- Key Laboratory of Molecular and Cellular Biology of Ministry of Education, College of Life Science, Hebei Normal University, Shijiazhuang, China
| | - Yingxian Li
- State Key Laboratory of Space Medicine Fundamentals and Application, China Astronaut Research and Training Center, Beijing, China
| |
Collapse
|
33
|
Toll-Like Receptor 4 Mediated Oxidized Low-Density Lipoprotein-Induced Foam Cell Formation in Vascular Smooth Muscle Cells via Src and Sirt1/3 Pathway. Mediators Inflamm 2021; 2021:6639252. [PMID: 33927570 PMCID: PMC8052139 DOI: 10.1155/2021/6639252] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Revised: 02/25/2021] [Accepted: 03/16/2021] [Indexed: 11/18/2022] Open
Abstract
Oxidized low-density lipoprotein (oxLDL) induced a foam-cell-like phenotype of the vascular smooth muscle cells (VSMCs), leading to the inflammatory responses incorporating Toll-like receptor- (Tlr-) mediated cellular alterations. However, the role of Tlr4 in foam cell formation and underlying molecular pathways has not been comprehensively elucidated. To further investigate the mechanism, VSMCs were incubated with different doses of oxLDL, and then, the lipid, reactive oxygen species (ROS) accumulation, Tlr family genes, and the foam cell phenotype were explored. We observed that oxLDL induced foam cell-like phenotype in VSMCs and led to lipid and ROS accumulation in a dose-dependent manner. Furthermore, in the Tlr family, Tlr4 demonstrated the strongest upregulation under oxLDL stimulation. Simultaneously, oxLDL induced activation of Src, higher expression of Nox2, and lower expression of Mnsod, Sirt1, and Sirt3. By interfering the TLR4 expression, the phenotype alteration, lipid accumulation in VSMCs, and Src kinase activation induced by oxLDL were abolished. After interfering Src activation, the oxLDL-induced lipid accumulation and foam cell phenotype in VSMCs were also alleviated. Furthermore, the ROS accumulation, upregulated Nox2 expression, downregulated Sirt1, Sirt3, and Mnsod expression in VSMCs under oxLDL stimulation were also relieved after the knockdown of Tlr4. Additionally, overexpression of Sirt1 and Sirt3 ameliorated the ROS accumulation and foam cell-like marker expression in VSMCs. These results demonstrated that beyond its familiar role in regulating inflammation response, Tlr4 is a critical regulator in oxLDL-induced foam cell formation in VSMCs via regulating Src kinase activation as well as Sirt1 and Sirt3 expression.
Collapse
|
34
|
Casein Kinase 2-Interacting Protein-1 Alleviates High Glucose-Reduced Autophagy, Oxidative Stress, and Apoptosis in Retinal Pigment Epithelial Cells via Activating the p62/KEAP1/NRF2 Signaling Pathway. J Ophthalmol 2021; 2021:6694050. [PMID: 33628480 PMCID: PMC7892229 DOI: 10.1155/2021/6694050] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Revised: 12/18/2020] [Accepted: 01/05/2021] [Indexed: 01/11/2023] Open
Abstract
Background Casein kinase 2-interacting protein-1 (CKIP-1) has been proved to be associated with complications of diabetes. Diabetic retinopathy is a main diabetic complication which usually leads to blindness. The current study aims to investigate the role of CKIP-1 in high glucose-treated retinal pigment epithelial (RPE) cells which is a component of blood-retinal barriers. Methods The RPE cells, ARPE-19, are treated with high glucose to mimic the diabetic stimulation. CKIP-1 was overexpressed in ARPE-19 cells to evaluate its effects on autophagy, oxidative stress, and apoptosis induced by high glucose treatment, using Western blot, immunofluorescence, and flow cytometry assays, respectively. Results CKIP-1 was expressed at a lower level in high glucose-treated cells than in normal glucose cells. Overexpression of CKIP-1 enhanced the Nrf2 translocation to the nucleus. Furthermore, high glucose-induced autophagy, oxidative stress, and apoptosis were inhibited after overexpression of CKIP-1. Also, CKIP-1 regulates the p62/Keap1/Nrf2 signaling, which might be the potential mechanism in this model. Conclusion In conclusion, CKIP-1 may be a potential therapeutic target that protects RPE cells from injury and subsequent diabetic retinopathy induced by high glucose.
Collapse
|
35
|
Cascio P. PA28γ: New Insights on an Ancient Proteasome Activator. Biomolecules 2021; 11:228. [PMID: 33562807 PMCID: PMC7915322 DOI: 10.3390/biom11020228] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2021] [Revised: 02/02/2021] [Accepted: 02/03/2021] [Indexed: 02/06/2023] Open
Abstract
PA28 (also known as 11S, REG or PSME) is a family of proteasome regulators whose members are widely present in many of the eukaryotic supergroups. In jawed vertebrates they are represented by three paralogs, PA28α, PA28β, and PA28γ, which assemble as heptameric hetero (PA28αβ) or homo (PA28γ) rings on one or both extremities of the 20S proteasome cylindrical structure. While they share high sequence and structural similarities, the three isoforms significantly differ in terms of their biochemical and biological properties. In fact, PA28α and PA28β seem to have appeared more recently and to have evolved very rapidly to perform new functions that are specifically aimed at optimizing the process of MHC class I antigen presentation. In line with this, PA28αβ favors release of peptide products by proteasomes and is particularly suited to support adaptive immune responses without, however, affecting hydrolysis rates of protein substrates. On the contrary, PA28γ seems to be a slow-evolving gene that is most similar to the common ancestor of the PA28 activators family, and very likely retains its original functions. Notably, PA28γ has a prevalent nuclear localization and is involved in the regulation of several essential cellular processes including cell growth and proliferation, apoptosis, chromatin structure and organization, and response to DNA damage. In striking contrast with the activity of PA28αβ, most of these diverse biological functions of PA28γ seem to depend on its ability to markedly enhance degradation rates of regulatory protein by 20S proteasome. The present review will focus on the molecular mechanisms and biochemical properties of PA28γ, which are likely to account for its various and complex biological functions and highlight the common features with the PA28αβ paralog.
Collapse
Affiliation(s)
- Paolo Cascio
- Department of Veterinary Sciences, University of Turin, Largo P. Braccini 2, 10095 Grugliasco, Italy
| |
Collapse
|
36
|
Lv JJ, Wang H, Cui HY, Liu ZK, Zhang RY, Lu M, Li C, Yong YL, Liu M, Zhang H, Zhang TJ, Zhang K, Li G, Nan G, Zhang C, Guo SP, Wang L, Chen ZN, Bian H. Blockade of Macrophage CD147 Protects Against Foam Cell Formation in Atherosclerosis. Front Cell Dev Biol 2021; 8:609090. [PMID: 33490072 PMCID: PMC7820343 DOI: 10.3389/fcell.2020.609090] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Accepted: 12/07/2020] [Indexed: 11/13/2022] Open
Abstract
The persistence of macrophage-derived foam cells in the artery wall fuels atherosclerosis development. However, the mechanism of foam cell formation regulation remains elusive. We are committed to determining the role that CD147 might play in macrophage foam cell formation during atherosclerosis. In this study, we found that CD147 expression was primarily increased in mouse and human atherosclerotic lesions that were rich in macrophages and could be upregulated by ox-LDL. High-throughput compound screening indicated that ox-LDL-induced CD147 upregulation in macrophages was achieved through PI3K/Akt/mTOR signaling. Genetic deletion of macrophage CD147 protected against foam cell formation by impeding cholesterol uptake, probably through the scavenger receptor CD36. The opposite effect was observed in primary macrophages isolated from macrophage-specific CD147-overexpressing mice. Moreover, bioinformatics results indicated that CD147 suppression might exert an atheroprotective effect via various processes, such as cholesterol biosynthetic and metabolic processes, LDL and plasma lipoprotein clearance, and decreased platelet aggregation and collagen degradation. Our findings identify CD147 as a potential target for prevention and treatment of atherosclerosis in the future.
Collapse
Affiliation(s)
- Jian-Jun Lv
- Department of Cell Biology, National Translational Science Center for Molecular Medicine, Fourth Military Medical University, Xi'an, China
| | - Hao Wang
- Department of Cell Biology, National Translational Science Center for Molecular Medicine, Fourth Military Medical University, Xi'an, China
| | - Hong-Yong Cui
- Department of Cell Biology, National Translational Science Center for Molecular Medicine, Fourth Military Medical University, Xi'an, China
| | - Ze-Kun Liu
- Department of Cell Biology, National Translational Science Center for Molecular Medicine, Fourth Military Medical University, Xi'an, China
| | - Ren-Yu Zhang
- Department of Cell Biology, National Translational Science Center for Molecular Medicine, Fourth Military Medical University, Xi'an, China
| | - Meng Lu
- Department of Cell Biology, National Translational Science Center for Molecular Medicine, Fourth Military Medical University, Xi'an, China
| | - Can Li
- Department of Cell Biology, National Translational Science Center for Molecular Medicine, Fourth Military Medical University, Xi'an, China
| | - Yu-Le Yong
- Department of Cell Biology, National Translational Science Center for Molecular Medicine, Fourth Military Medical University, Xi'an, China
| | - Man Liu
- Department of Cell Biology, National Translational Science Center for Molecular Medicine, Fourth Military Medical University, Xi'an, China
| | - Hai Zhang
- Department of Cell Biology, National Translational Science Center for Molecular Medicine, Fourth Military Medical University, Xi'an, China
| | - Tian-Jiao Zhang
- Department of Cell Biology, National Translational Science Center for Molecular Medicine, Fourth Military Medical University, Xi'an, China
| | - Kun Zhang
- Department of Cell Biology, National Translational Science Center for Molecular Medicine, Fourth Military Medical University, Xi'an, China.,School of Science, College of Life Science and Bioengineering, Beijing Jiaotong University, Beijing, China
| | - Gang Li
- Department of Cell Biology, National Translational Science Center for Molecular Medicine, Fourth Military Medical University, Xi'an, China.,Institutes of Biomedicine and Department of Cell Biology, Jinan University, Guangzhou, China
| | - Gang Nan
- Department of Cell Biology, National Translational Science Center for Molecular Medicine, Fourth Military Medical University, Xi'an, China
| | - Cong Zhang
- Department of Cell Biology, National Translational Science Center for Molecular Medicine, Fourth Military Medical University, Xi'an, China
| | - Shuang-Ping Guo
- Department of Pathology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Ling Wang
- College of Military Preventive Medicine, Fourth Military Medical University, Xi'an, China
| | - Zhi-Nan Chen
- Department of Cell Biology, National Translational Science Center for Molecular Medicine, Fourth Military Medical University, Xi'an, China
| | - Huijie Bian
- Department of Cell Biology, National Translational Science Center for Molecular Medicine, Fourth Military Medical University, Xi'an, China
| |
Collapse
|
37
|
Li L, Xie P, Lin W, Liu J, Chen J, Guo Z, Bin C, An W, Zhang C, Zhan Y. CKIP-1 augments autophagy in steatotic hepatocytes by inhibiting Akt/mTOR signal pathway. Exp Cell Res 2020; 397:112341. [PMID: 33191205 DOI: 10.1016/j.yexcr.2020.112341] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2020] [Revised: 09/26/2020] [Accepted: 10/21/2020] [Indexed: 12/23/2022]
Abstract
Nonalcoholic fatty liver disease (NAFLD), which is characterized by aberrant accumulation of intrahepatic triglycerides and lipid droplets (LDs) in the liver cells, is becoming increasingly prevalent at an alarming rate worldwide. LDs can be consumed by either hydrolysis or autophagy, which is shown to be of importance in the regulation of hepatic lipid metabolism. We have shown that deficiency of pleckstrin homology domain-containing casein kinase 2 interacting protein-1 (CKIP-1), a scaffold protein that interacts with various proteins in multiple signal pathways, in mice aggravates high-fat diet induced fatty liver. However, its underlying mechanisms remain largely unknown. In this study, we found that the mRNA and protein levels of CKIP-1 decreased dramatically in steatotic HepG2 cells induced by oleic acid (OA) treatment. Coincidently, hepatic autophagy was also dynamically regulated in steatotic HepG2 cells. In addition, overexpression of CKIP-1 activated autophagy by suppression of Akt/mTOR signaling, which in turn reduced lipid accumulation. Moreover, these phenomena were reversed in CKIP-1-shRNA transfected steatotic hepatocytes. To further evaluate the potential role of CKIP-1 in autophagy, we determined the level of autophagy related proteins in CKIP-1 knockout mice. These results supported our findings in vitro. In summary, we found CKIP-1 to be a positive regulator of hepatic autophagy and a promising therapeutic target for treatment of NAFLD.
Collapse
Affiliation(s)
- Li Li
- Department of Gastroenterology, Beijing Tongren Hospital, Capital Medical University, Beijing, 100730, China
| | - Ping Xie
- Department of Cell Biology, Municipal Laboratory for Liver Protection and Regulation of Regeneration, Capital Medical University, Beijing, 100069, China
| | - Wenjun Lin
- Department of Gastroenterology, Beijing Tongren Hospital, Capital Medical University, Beijing, 100730, China
| | - Jinsheng Liu
- Department of Anesthesiology, Beijing Tongren Hospital, Capital Medical University, Beijing, 100730, China
| | - Jing Chen
- Department of Gastroenterology, Beijing Tongren Hospital, Capital Medical University, Beijing, 100730, China
| | - Zihao Guo
- Department of Gastroenterology, Beijing Tongren Hospital, Capital Medical University, Beijing, 100730, China
| | - Chuxuan Bin
- Department of Gastroenterology, Beijing Tongren Hospital, Capital Medical University, Beijing, 100730, China
| | - Wei An
- Department of Cell Biology, Municipal Laboratory for Liver Protection and Regulation of Regeneration, Capital Medical University, Beijing, 100069, China.
| | - Chuan Zhang
- Department of Gastroenterology, Beijing Tongren Hospital, Capital Medical University, Beijing, 100730, China.
| | - Yutao Zhan
- Department of Gastroenterology, Beijing Tongren Hospital, Capital Medical University, Beijing, 100730, China.
| |
Collapse
|
38
|
Wang C, Yang W, Liang X, Song W, Lin J, Sun Y, Guan X. MicroRNA-761 modulates foam cell formation and inflammation through autophagy in the progression of atherosclerosis. Mol Cell Biochem 2020; 474:135-146. [PMID: 32772311 DOI: 10.1007/s11010-020-03839-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2020] [Accepted: 07/14/2020] [Indexed: 02/08/2023]
Abstract
Macrophage-derived foam cells formation is the initial stage of atherosclerosis, and lipid-laden macrophage accumulation is also considered as the symbol of unstable plaque. Autophagy is a subcellular process responsible for the degradation of damaged organelles and aggregated proteins in cells (Grootaert in Oxid Med Cell Longev: 7687083, 2018). Macrophage autophagy plays an important role in atherosclerosis under various stress conditions, and microRNAs are involved in this complicated process. The present study was programmed to explore the effects of microRNA-761 on macrophage-derived foam cell formation, focusing on the role of autophagy in this pathological process. The differentiated human THP-1 macrophages were used in the study. THP-1-derived macrophages were treated with miR-761 mimics or inhibitors and cultured with oxidized low-density lipoprotein to mimic the lipid-rich environment in blood vessel. The expression of miR-761 and mRNA levels of IL-1β and IL-18 were analyzed by quantitative real-time PCR. The effect of miR-761 on autophagy was evaluated by the protein levels of Beclin1, p62/SQSTM1, microtubule-associated protein light chain 3, mammalian target of rapamycin (mTOR), and unc-51-like autophagy activating kinase 1 (ULK1), determined by immunoblot and autophagic flux detected by fluorescent staining. The secretion of IL-1β and IL-18 was tested by enzyme-linked immunosorbent reaction kit. Lipid accumulation in foam cells was detected by oil red "O" staining. We demonstrated that miR-761 was able to repress foam cell formation and reduce the production of atherogenic inflammatory cytokines IL-1β and IL-18 in an autophagy-dependent manner in atherosclerosis, possibly via mTOR-ULK1 signaling pathway. In summary, we described an athero-protective function of miR-761 in macrophages incubated with excess ox-LDL and identified an important novel modulator of mTOR signaling and autophagy in macrophage-derived foam cells. This finding may provide a potential target for the prevention and early treatment in high-risk group of atherosclerosis.
Collapse
Affiliation(s)
- Chao Wang
- Department of Laboratory Diagnostics, First Affiliated Hospital of Harbin Medical University, 23 Youzheng Street, NanGang, Harbin, 150001, Heilongjiang, People's Republic of China
| | - Wei Yang
- Department of Laboratory Diagnostics, First Affiliated Hospital of Harbin Medical University, 23 Youzheng Street, NanGang, Harbin, 150001, Heilongjiang, People's Republic of China
| | - Xiaofei Liang
- Department of Laboratory Diagnostics, First Affiliated Hospital of Harbin Medical University, 23 Youzheng Street, NanGang, Harbin, 150001, Heilongjiang, People's Republic of China
| | - Wei Song
- Department of Laboratory Diagnostics, First Affiliated Hospital of Harbin Medical University, 23 Youzheng Street, NanGang, Harbin, 150001, Heilongjiang, People's Republic of China
| | - Jing Lin
- Department of Laboratory Diagnostics, First Affiliated Hospital of Harbin Medical University, 23 Youzheng Street, NanGang, Harbin, 150001, Heilongjiang, People's Republic of China
| | - Yan Sun
- Department of Laboratory Diagnostics, First Affiliated Hospital of Harbin Medical University, 23 Youzheng Street, NanGang, Harbin, 150001, Heilongjiang, People's Republic of China
| | - Xiuru Guan
- Department of Laboratory Diagnostics, First Affiliated Hospital of Harbin Medical University, 23 Youzheng Street, NanGang, Harbin, 150001, Heilongjiang, People's Republic of China.
| |
Collapse
|
39
|
Myriocin and d-PDMP ameliorate atherosclerosis in ApoE-/- mice via reducing lipid uptake and vascular inflammation. Clin Sci (Lond) 2020; 134:439-458. [PMID: 32091078 DOI: 10.1042/cs20191028] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2019] [Revised: 02/17/2020] [Accepted: 02/21/2020] [Indexed: 02/05/2023]
Abstract
Sphingolipids have been implicated in the etiology of atherosclerosis. The commonly used sphingolipid inhibitors, myriocin (a ceramide inhibitor) and d-PDMP (d-threo-1-phenyl-2-decanoylamino-3-morpholino-1-propanol, a glycosphingolipid inhibitor), have shown therapeutic potential but their efficacy and their underlying mechanisms remain unclear. Here, apolipoprotein E-deficient (apoE-/-) mice were fed a high-fat diet (HFD) and treated with a control, myriocin, d-PDMP, or atorvastatin for 12 weeks. We analyzed the effects of these drugs on the size and detailed composition of atherosclerotic plaques. Molecular biological approaches were used to explore how the inhibitors affect lipid metabolism and foam-cell formation. Treatment with myriocin or d-PDMP led to smaller and less vulnerable atherosclerotic lesions and was almost as effective as atorvastatin. Sphingolipid inhibitors down-regulated the expression of monocyte chemotactic protein 1 (MCP-1) and its receptor chemoattractant cytokine receptor 2 (CCR2), which play a key role in monocyte recruitment. They also decreased pro-inflammatory Ly-6chigh monocytes and influenced the uptake of modified LDL by down-regulating the expression of cluster of differentiation 36 (CD36) and lectin-like oxidized LDL (ox-LDL) receptor-1 (LOX-1). The inhibitors exhibited the advantage of maintaining normal glucose homeostasis compared with atorvastatin. These findings reveal for the first time that the modulation of sphingolipid synthesis can effectively alleviate atherosclerosis progression by preventing lipid uptake and reducing inflammatory responses in the arterial walls.
Collapse
|
40
|
Treatment of atherosclerosis by macrophage-biomimetic nanoparticles via targeted pharmacotherapy and sequestration of proinflammatory cytokines. Nat Commun 2020; 11:2622. [PMID: 32457361 PMCID: PMC7251120 DOI: 10.1038/s41467-020-16439-7] [Citation(s) in RCA: 350] [Impact Index Per Article: 70.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2019] [Accepted: 04/30/2020] [Indexed: 12/27/2022] Open
Abstract
Vascular disease remains the leading cause of death and disability, the etiology of which often involves atherosclerosis. The current treatment of atherosclerosis by pharmacotherapy has limited therapeutic efficacy. Here we report a biomimetic drug delivery system derived from macrophage membrane coated ROS-responsive nanoparticles (NPs). The macrophage membrane not only avoids the clearance of NPs from the reticuloendothelial system, but also leads NPs to the inflammatory tissues, where the ROS-responsiveness of NPs enables specific payload release. Moreover, the macrophage membrane sequesters proinflammatory cytokines to suppress local inflammation. The synergistic effects of pharmacotherapy and inflammatory cytokines sequestration from such a biomimetic drug delivery system lead to improved therapeutic efficacy in atherosclerosis. Comparison to macrophage internalized with ROS-responsive NPs, as a live-cell based drug delivery system for treatment of atherosclerosis, suggests that cell membrane coated drug delivery approach is likely more suitable for dealing with an inflammatory disease than the live-cell approach.
Collapse
|
41
|
Fan Y, Yang J, Li H, Li H, Zhang S, Li X, Song Y, Dang W, Liu L, Cao X, Wang X, Nandakumar KS, Shen X, You Y. WITHDRAWN: SNX10 deficiency restricts foam cell formation and protects against atherosclerosis by suppressing CD36-Lyn axis. Can J Cardiol 2020:S0828-282X(20)30456-6. [PMID: 32428616 DOI: 10.1016/j.cjca.2020.05.010] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2019] [Revised: 04/24/2020] [Accepted: 05/06/2020] [Indexed: 12/11/2022] Open
Abstract
This article has been withdrawn at the request of the author. The Publisher apologizes for any inconvenience this may cause. The full Elsevier Policy on Article Withdrawal can be found at https://www.elsevier.com/about/our-business/policies/article-withdrawal
Collapse
Affiliation(s)
- Yujuan Fan
- Institute of Fudan-Minhang Academic Health System, Minhang Hospital, Fudan University, Shanghai, China; Department of Pharmacology, School of Pharmacy, Fudan University, Shanghai, China
| | - Jialin Yang
- Institute of Fudan-Minhang Academic Health System, Minhang Hospital, Fudan University, Shanghai, China; Department of Pharmacology, School of Pharmacy, Fudan University, Shanghai, China
| | - Hui Li
- School of Kinesiology, Shanghai University of Sport, Shanghai, China
| | - HaiDong Li
- Department of Pharmacology, School of Pharmacy, Fudan University, Shanghai, China
| | - Sulin Zhang
- Department of Pharmacology, School of Pharmacy, Fudan University, Shanghai, China
| | - Xuesong Li
- Institute of Fudan-Minhang Academic Health System, Minhang Hospital, Fudan University, Shanghai, China; Department of Pharmacology, School of Pharmacy, Fudan University, Shanghai, China
| | - Yuping Song
- Institute of Fudan-Minhang Academic Health System, Minhang Hospital, Fudan University, Shanghai, China; Department of Pharmacology, School of Pharmacy, Fudan University, Shanghai, China
| | - Wenzhen Dang
- Department of Pharmacology, School of Pharmacy, Fudan University, Shanghai, China
| | - Lixin Liu
- Department of Pharmacology, School of Pharmacy, Fudan University, Shanghai, China
| | - Xinyue Cao
- Department of Pharmacology, School of Pharmacy, Fudan University, Shanghai, China
| | - Xu Wang
- Department of Pharmacology, School of Pharmacy, Fudan University, Shanghai, China
| | | | - Xiaoyan Shen
- Institute of Fudan-Minhang Academic Health System, Minhang Hospital, Fudan University, Shanghai, China; Department of Pharmacology, School of Pharmacy, Fudan University, Shanghai, China.
| | - Yan You
- Institute of Fudan-Minhang Academic Health System, Minhang Hospital, Fudan University, Shanghai, China; Department of Pharmacology, School of Pharmacy, Fudan University, Shanghai, China; National Institute of Allergy and Infectious, National Institute of Health, Rockville, USA.
| |
Collapse
|
42
|
Coux O, Zieba BA, Meiners S. The Proteasome System in Health and Disease. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1233:55-100. [DOI: 10.1007/978-3-030-38266-7_3] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
43
|
Jia Q, Cao H, Shen D, Yan L, Chen C, Xing S. Fisetin, via CKIP-1/REGγ, limits oxidized LDL-induced lipid accumulation and senescence in RAW264.7 macrophage-derived foam cells. Eur J Pharmacol 2019; 865:172748. [DOI: 10.1016/j.ejphar.2019.172748] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2019] [Revised: 10/16/2019] [Accepted: 10/17/2019] [Indexed: 11/25/2022]
|
44
|
Tone K, Stappers MHT, Willment JA, Brown GD. C-type lectin receptors of the Dectin-1 cluster: Physiological roles and involvement in disease. Eur J Immunol 2019; 49:2127-2133. [PMID: 31580478 PMCID: PMC6916577 DOI: 10.1002/eji.201847536] [Citation(s) in RCA: 61] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2019] [Revised: 08/12/2019] [Accepted: 09/27/2019] [Indexed: 12/27/2022]
Abstract
C-type lectin receptors (CLRs) are essential for multicellular existence, having diverse functions ranging from embryonic development to immune function. One subgroup of CLRs is the Dectin-1 cluster, comprising of seven receptors including MICL, CLEC-2, CLEC-12B, CLEC-9A, MelLec, Dectin-1, and LOX-1. Reflecting the larger CLR family, the Dectin-1 cluster of receptors has a broad range of ligands and functions, but importantly, is involved in numerous pathophysiological processes that regulate health and disease. Indeed, these receptors have been implicated in development, infection, regulation of inflammation, allergy, transplantation tolerance, cancer, cardiovascular disease, arthritis, and other autoimmune diseases. In this mini-review, we discuss the latest advancements in elucidating the function(s) of each of the Dectin-1 cluster CLRs, focussing on their physiological roles and involvement in disease.
Collapse
Affiliation(s)
- Kazuya Tone
- Aberdeen Fungal Group, University of Aberdeen, Institute of Medical Sciences, Aberdeen, Scotland
| | - Mark H T Stappers
- Medical Research Council Centre for Medical Mycology, University of Exeter, Exeter, Devon, England
| | - Janet A Willment
- Aberdeen Fungal Group, University of Aberdeen, Institute of Medical Sciences, Aberdeen, Scotland.,Medical Research Council Centre for Medical Mycology, University of Exeter, Exeter, Devon, England
| | - Gordon D Brown
- Aberdeen Fungal Group, University of Aberdeen, Institute of Medical Sciences, Aberdeen, Scotland.,Medical Research Council Centre for Medical Mycology, University of Exeter, Exeter, Devon, England
| |
Collapse
|
45
|
Physiological functions of CKIP-1: From molecular mechanisms to therapy implications. Ageing Res Rev 2019; 53:100908. [PMID: 31082489 DOI: 10.1016/j.arr.2019.05.002] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2019] [Revised: 05/07/2019] [Accepted: 05/09/2019] [Indexed: 02/07/2023]
Abstract
The casein kinase 2 interacting protein-1 (CKIP-1, also known as PLEKHO1) is initially identified as a specific CK2α subunit-interacting protein. Subsequently, various proteins, including CPα, PAK1, Arp2/3, HDAC1, c-Jun, ATM, Smurf1, Rpt6, Akt, IFP35, TRAF6, REGγ and CARMA1, were reported to interact with CKIP-1. Owing to the great diversity of interacted proteins, CKIP-1 exhibits multiple biologic functions in cell morphology, cell differentiation and cell apoptosis. Besides, these functions are subcellular localization, cell type, and regulatory signaling dependent. CKIP-1 is involved in biological processes consisting of bone formation, tumorigenesis and immune regulation. Importantly, deregulation of CKIP-1 results in osteoporosis, tumor, and atherosclerosis. In this review, we introduce the molecular functions, biological processes and promising of therapeutic strategies. Through summarizing the intrinsic mechanisms, we expect to open new therapeutic avenues for CKIP-1.
Collapse
|
46
|
Yang S, Xia YP, Luo XY, Chen SL, Li BW, Ye ZM, Chen SC, Mao L, Jin HJ, Li YN, Hu B. Exosomal CagA derived from Helicobacter pylori-infected gastric epithelial cells induces macrophage foam cell formation and promotes atherosclerosis. J Mol Cell Cardiol 2019; 135:40-51. [PMID: 31352044 DOI: 10.1016/j.yjmcc.2019.07.011] [Citation(s) in RCA: 51] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/01/2019] [Revised: 07/16/2019] [Accepted: 07/24/2019] [Indexed: 12/11/2022]
Abstract
BACKGROUND Seroepidemiological studies have highlighted a positive relation between CagA-positive Helicobacter pylori (H. pylori), atherosclerosis and related clinic events. However, this link has not been well validated. The present study was designed to explore the role of H. pylori PMSS1 (a CagA-positive strain that can translocate CagA into host cells) and exosomal CagA in the progression of atherosclerosis. METHODS To evaluate whether H. pylori accelerates or even induces atherosclerosis, H. pylori-infected C57/BL6 mice and ApoE-/- mice were maintained under different dietary conditions. To identify the role of H. pylori-infected gastric epithelial cells-derived exosomes (Hp-GES-EVs) and exosomal CagA in atherosclerosis, ApoE-/- mice were given intravenous or intraperitoneal injections of saline, GES-EVs, Hp-GES-EVs, and recombinant CagA protein (rCagA). FINDINGS CagA-positive H. pylori PMSS1 infection does not induce but promotes macrophage-derived foam cell formation and augments atherosclerotic plaque growth and instability in two animal models. Meanwhile, circulating Hp-GES-EVs are taken up in aortic plaque, and CagA is secreted in Hp-GES-EVs. Furthermore, the CagA-containing EVs and rCagA exacerbates macrophage-derived foam cell formation and lesion development in vitro and in vivo, recapitulating the pro-atherogenic effects of CagA-positive H. pylori. Mechanistically, CagA suppresses the transcription of cholesterol efflux transporters by downregulating the expression of transcriptional factors PPARγ and LXRα and thus enhances foam cell formation. INTERPRETATION These results may provide new insights into the role of exosomal CagA in the pathogenesis of CagA-positive H. pylori infection-related atherosclerosis. It is suggested that preventing and eradicating CagA-positive H. pylori infection could reduce the incidence of atherosclerosis and related events.
Collapse
Affiliation(s)
- Shuai Yang
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yuan-Peng Xia
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xue-Ying Luo
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Shao-Li Chen
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Bo-Wei Li
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zi-Ming Ye
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Department of Neurology, The First Affiliated Hospital, Guangxi Medical University, Nanning, Guangxi, China
| | - Sheng-Cai Chen
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ling Mao
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hui-Juan Jin
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ya-Nan Li
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Bo Hu
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| |
Collapse
|
47
|
Vázquez-Arreguín K, Bensard C, Schell JC, Swanson E, Chen X, Rutter J, Tantin D. Oct1/Pou2f1 is selectively required for colon regeneration and regulates colon malignancy. PLoS Genet 2019; 15:e1007687. [PMID: 31059499 PMCID: PMC6522070 DOI: 10.1371/journal.pgen.1007687] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2018] [Revised: 05/16/2019] [Accepted: 04/16/2019] [Indexed: 12/22/2022] Open
Abstract
The transcription factor Oct1/Pou2f1 promotes poised gene expression states, mitotic stability, glycolytic metabolism and other characteristics of stem cell potency. To determine the effect of Oct1 loss on stem cell maintenance and malignancy, we deleted Oct1 in two different mouse gut stem cell compartments. Oct1 deletion preserved homeostasis in vivo and the ability to establish organoids in vitro, but blocked the ability to recover from treatment with dextran sodium sulfate, and the ability to maintain organoids after passage. In a chemical model of colon cancer, loss of Oct1 in the colon severely restricted tumorigenicity. In contrast, loss of one or both Oct1 alleles progressively increased tumor burden in a colon cancer model driven by loss-of-heterozygosity of the tumor suppressor gene Apc. The different outcomes are consistent with prior findings that Oct1 promotes mitotic stability, and consistent with differentially expressed genes between the two models. Oct1 ChIPseq using HCT116 colon carcinoma cells identifies target genes associated with mitotic stability, metabolism, stress response and malignancy. This set of gene targets overlaps significantly with genes differentially expressed in the two tumor models. These results reveal that Oct1 is selectively required for recovery after colon damage, and that Oct1 has potent effects in colon malignancy, with outcome (pro-oncogenic or tumor suppressive) dictated by tumor etiology.
Collapse
Affiliation(s)
- Karina Vázquez-Arreguín
- Department of Pathology and Huntsman Cancer Institute, University of Utah School of Medicine, Salt Lake City, UT, United States of America
| | - Claire Bensard
- Department of Biochemistry, University of Utah School of Medicine, Salt Lake City, UT, United States of America
| | - John C. Schell
- Department of Biochemistry, University of Utah School of Medicine, Salt Lake City, UT, United States of America
| | - Eric Swanson
- Department of Pathology and Huntsman Cancer Institute, University of Utah School of Medicine, Salt Lake City, UT, United States of America
| | - Xinjian Chen
- Department of Pathology and Huntsman Cancer Institute, University of Utah School of Medicine, Salt Lake City, UT, United States of America
| | - Jared Rutter
- Department of Biochemistry, University of Utah School of Medicine, Salt Lake City, UT, United States of America
- Howard Hughes Medical Institute, Salt Lake City, Utah, United States of America
| | - Dean Tantin
- Department of Pathology and Huntsman Cancer Institute, University of Utah School of Medicine, Salt Lake City, UT, United States of America
| |
Collapse
|