1
|
Zhang Y, Zhang D, Xie Z, Xia T, Zou L, Wang T, Zhong L, Zeng Z, Wang L, Chen G, Liang X. Integrated transcriptomic and metabolomic analysis reveals the effects of EMMPRIN on nucleotide metabolism and 1C metabolism in AS mouse BMDMs. Front Mol Biosci 2025; 11:1460186. [PMID: 40125455 PMCID: PMC11927532 DOI: 10.3389/fmolb.2024.1460186] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2024] [Accepted: 12/27/2024] [Indexed: 03/25/2025] Open
Abstract
Background Extracellular matrix metalloproteinase inducer (EMMPRIN) has been considered as a key promoting factor in atherosclerosis (AS). Some studies have shown that regulating EMMPRIN expression in bone marrow-derived macrophages (BMDMs) of ApoE-/- mice can affect plaque stability, but the mechanism was not clear. Methods AS model mice were built from high-fat-feeding ApoE -/- mice, and were divided into siE group and CON group. The BMDMs and aortas from AS mice were harvested following in vivo treatment with either EMMPRIN short interfering (si)RNA (siEMMPRIN) or negative control siRNA. Transcriptomic and metabolomic profiles were analyzed using RNA-sequencing and Liquid chromatography-tandem mass spectrometry (LC-MS/MS), respectively. The efficacy of siEMMPRIN was assessed through real-time quantitative polymerase chain reaction (RT-qPCR) and Western blotting (WB). Immunofluorescence staining was employed to measure EMMPRIN expression within aortic atherosclerotic plaques. Cell proliferation was monitored using the Cell Counting Kit-8 (CCK8), while flow cytometry was utilized to analyze the cell cycle. Additionally, seahorse analysis and oil red O staining were conducted to verify glucose and lipid metabolism, respectively. Results A total of 3,282 differentially expressed metabolites (DEMs) and 16,138 differentially expressed genes (DEGs) were identified between the CON group and siE group. The nucleotide metabolism and one-carbon (1C) metabolism were identified as major altered pathways at both the transcriptional and metabolic levels. Metabolomic results identified increased levels of glycine, serine, betaine and S-adenosyl-L-methionine (SAM) to S-adenosyl-L-homocysteine (SAH) ratio and decreased levels of dimethylglycine (DMG) and SAH in 1C metabolism, accompanied by the accumulation of nucleotides, nucleosides, and bases in nucleotide metabolism. Transcriptomics results shown that Dnmt, Mthfd2 and Dhfr were downregulated, while Mthfr were upregulated in 1C metabolism. And numerous genes involved in de novo nucleotide synthesis, pentose phosphate pathway (PPP) and dNTP production were significantly inhibited, which may be associated with decreased BMDMs proliferation and cell cycle arrest in the G0/G1 phase in siE group. Multi-omics results also showed changes in glucose and lipid metabolism. Seahorse assay confirmed reduced glycolysis and oxidative phosphorylation (OXPHOS) levels and the Oil Red O staining confirmed the decrease of lipid droplets in siE group. Conclusion The integrated metabolomic and transcriptomic analysis suggested that nucleotide metabolism and 1C metabolism may be major metabolic pathways affected by siEMMPRIN in AS mouse BMDMs. Our study contributes to a better understanding of the role of EMMPRIN in AS development.
Collapse
Affiliation(s)
- Yun Zhang
- First Clinical College, Chongqing Medical University, Chongqing, China
| | - Diyuan Zhang
- Second Clinical College, Chongqing Medical University, Chongqing, China
| | - Zulong Xie
- Department of Cardiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Tianli Xia
- Department of Cardiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Lili Zou
- Department of Cardiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Tao Wang
- Department of Cardiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Li Zhong
- Department of Cardiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Zhuo Zeng
- First Clinical College, Chongqing Medical University, Chongqing, China
| | - Lingying Wang
- First Clinical College, Chongqing Medical University, Chongqing, China
| | - Guozhu Chen
- Department of Cardiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Xing Liang
- Department of Cardiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
2
|
Ajoolabady A, Pratico D, Lin L, Mantzoros CS, Bahijri S, Tuomilehto J, Ren J. Inflammation in atherosclerosis: pathophysiology and mechanisms. Cell Death Dis 2024; 15:817. [PMID: 39528464 PMCID: PMC11555284 DOI: 10.1038/s41419-024-07166-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Revised: 08/26/2024] [Accepted: 10/17/2024] [Indexed: 11/16/2024]
Abstract
Atherosclerosis imposes a heavy burden on cardiovascular health due to its indispensable role in the pathogenesis of cardiovascular disease (CVD) such as coronary artery disease and heart failure. Ample clinical and experimental evidence has corroborated the vital role of inflammation in the pathophysiology of atherosclerosis. Hence, the demand for preclinical research into atherosclerotic inflammation is on the horizon. Indeed, the acquisition of an in-depth knowledge of the molecular and cellular mechanisms of inflammation in atherosclerosis should allow us to identify novel therapeutic targets with translational merits. In this review, we aimed to critically discuss and speculate on the recently identified molecular and cellular mechanisms of inflammation in atherosclerosis. Moreover, we delineated various signaling cascades and proinflammatory responses in macrophages and other leukocytes that promote plaque inflammation and atherosclerosis. In the end, we highlighted potential therapeutic targets, the pros and cons of current interventions, as well as anti-inflammatory and atheroprotective mechanisms.
Collapse
Affiliation(s)
- Amir Ajoolabady
- Department of Biomedical Engineering, University of Alabama at Birmingham, Birmingham, AL, 35294, USA
| | - Domenico Pratico
- Alzheimer's Center at Temple, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, 19140, USA
| | - Ling Lin
- Shanghai Institute of Cardiovascular Diseases, Department of Cardiology, Zhongshan Hospital Fudan University, Shanghai, 200032, China
- National Clinical Research Center for Interventional Medicine, Shanghai, 200032, China
| | | | - Suhad Bahijri
- Diabetes Research Group, King Abdulaziz University, Jeddah, Saudi Arabia.
| | - Jaakko Tuomilehto
- Diabetes Research Group, King Abdulaziz University, Jeddah, Saudi Arabia.
- Department of Public Health, University of Helsinki, Helsinki, Finland.
- Health Promotion Unit, Finnish Institute for Health and Welfare, Helsinki, Finland.
| | - Jun Ren
- Shanghai Institute of Cardiovascular Diseases, Department of Cardiology, Zhongshan Hospital Fudan University, Shanghai, 200032, China.
- National Clinical Research Center for Interventional Medicine, Shanghai, 200032, China.
| |
Collapse
|
3
|
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
|
4
|
Lv JJ, Wang H, Zhang C, Zhang TJ, Wei HL, Liu ZK, Ma YH, Yang Z, He Q, Wang LJ, Duan LL, Chen ZN, Bian H. CD147 Sparks Atherosclerosis by Driving M1 Phenotype and Impairing Efferocytosis. Circ Res 2024; 134:165-185. [PMID: 38166463 DOI: 10.1161/circresaha.123.323223] [Citation(s) in RCA: 23] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2023] [Accepted: 12/18/2023] [Indexed: 01/04/2024]
Abstract
BACKGROUND Atherosclerosis is a globally prevalent chronic inflammatory disease with high morbidity and mortality. The development of atherosclerotic lesions is determined by macrophages. This study aimed to investigate the specific role of myeloid-derived CD147 (cluster of differentiation 147) in atherosclerosis and its translational significance. METHODS AND RESULTS We generated mice with a myeloid-specific knockout of CD147 and mice with restricted CD147 overexpression, both in an apoE-deficient (ApoE-/-) background. Here, the myeloid-specific deletion of CD147 ameliorated atherosclerosis and inflammation. Consistent with our in vivo data, macrophages isolated from myeloid-specific CD147 knockout mice exhibited a phenotype shift from proinflammatory to anti-inflammatory macrophage polarization in response to lipopolysaccharide/IFN (interferon)-γ. These macrophages demonstrated a weakened proinflammatory macrophage phenotype, characterized by reduced production of NO and reactive nitrogen species derived from iNOS (inducible NO synthase). Mechanistically, the TRAF6 (tumor necrosis factor receptor-associated factor 6)-IKK (inhibitor of κB kinase)-IRF5 (IFN regulatory factor 5) signaling pathway was essential for the effect of CD147 on proinflammatory responses. Consistent with the reduced size of the necrotic core, myeloid-specific CD147 deficiency diminished the susceptibility of iNOS-mediated late apoptosis, accompanied by enhanced efferocytotic capacity mediated by increased secretion of GAS6 (growth arrest-specific 6) in proinflammatory macrophages. These findings were consistent in a mouse model with myeloid-restricted overexpression of CD147. Furthermore, we developed a new atherosclerosis model in ApoE-/- mice with humanized CD147 transgenic expression and demonstrated that the administration of an anti-human CD147 antibody effectively suppressed atherosclerosis by targeting inflammation and efferocytosis. CONCLUSIONS Myeloid CD147 plays a crucial role in the growth of plaques by promoting inflammation in a TRAF6-IKK-IRF5-dependent manner and inhibiting efferocytosis by suppressing GAS6 during proinflammatory conditions. Consequently, the use of anti-human CD147 antibodies presents a complementary therapeutic approach to the existing lipid-lowering strategies for treating atherosclerotic diseases.
Collapse
Affiliation(s)
- Jian-Jun Lv
- Department of Cell Biology, National Translational Science Center for Molecular Medicine (J.-J.L., H.W., C.Z., T.-J.Z., H.-L.W., Z.-K.L., Y.-H.M., Q.H., L.-J.W., Z.-N.C., H.B.), Fourth Military Medical University, Xi'an, China
| | - Hao Wang
- Department of Cell Biology, National Translational Science Center for Molecular Medicine (J.-J.L., H.W., C.Z., T.-J.Z., H.-L.W., Z.-K.L., Y.-H.M., Q.H., L.-J.W., Z.-N.C., H.B.), Fourth Military Medical University, Xi'an, China
| | - Cong Zhang
- Department of Cell Biology, National Translational Science Center for Molecular Medicine (J.-J.L., H.W., C.Z., T.-J.Z., H.-L.W., Z.-K.L., Y.-H.M., Q.H., L.-J.W., Z.-N.C., H.B.), Fourth Military Medical University, Xi'an, China
| | - Tian-Jiao Zhang
- Department of Cell Biology, National Translational Science Center for Molecular Medicine (J.-J.L., H.W., C.Z., T.-J.Z., H.-L.W., Z.-K.L., Y.-H.M., Q.H., L.-J.W., Z.-N.C., H.B.), Fourth Military Medical University, Xi'an, China
| | - Hao-Lin Wei
- Department of Cell Biology, National Translational Science Center for Molecular Medicine (J.-J.L., H.W., C.Z., T.-J.Z., H.-L.W., Z.-K.L., Y.-H.M., Q.H., L.-J.W., Z.-N.C., H.B.), Fourth Military Medical University, Xi'an, China
| | - Ze-Kun Liu
- Department of Cell Biology, National Translational Science Center for Molecular Medicine (J.-J.L., H.W., C.Z., T.-J.Z., H.-L.W., Z.-K.L., Y.-H.M., Q.H., L.-J.W., Z.-N.C., H.B.), Fourth Military Medical University, Xi'an, China
| | - Yi-Hui Ma
- Department of Cell Biology, National Translational Science Center for Molecular Medicine (J.-J.L., H.W., C.Z., T.-J.Z., H.-L.W., Z.-K.L., Y.-H.M., Q.H., L.-J.W., Z.-N.C., H.B.), Fourth Military Medical University, Xi'an, China
| | - Zhi Yang
- Department of Radiation Oncology, Xijing Hospital (Z.Y.), Fourth Military Medical University, Xi'an, China
| | - Qian He
- Department of Cell Biology, National Translational Science Center for Molecular Medicine (J.-J.L., H.W., C.Z., T.-J.Z., H.-L.W., Z.-K.L., Y.-H.M., Q.H., L.-J.W., Z.-N.C., H.B.), Fourth Military Medical University, Xi'an, China
| | - Li-Juan Wang
- Department of Cell Biology, National Translational Science Center for Molecular Medicine (J.-J.L., H.W., C.Z., T.-J.Z., H.-L.W., Z.-K.L., Y.-H.M., Q.H., L.-J.W., Z.-N.C., H.B.), Fourth Military Medical University, Xi'an, China
| | - Li-Li Duan
- Department of Gastrointestinal Surgery, State Key Laboratory of Cancer Biology, National Clinical Research Center for Digestive Diseases, and Xijing Hospital of Digestive Diseases (L.-L.D.), Fourth Military Medical University, Xi'an, China
| | - Zhi-Nan Chen
- Department of Cell Biology, National Translational Science Center for Molecular Medicine (J.-J.L., H.W., C.Z., T.-J.Z., H.-L.W., Z.-K.L., Y.-H.M., Q.H., L.-J.W., Z.-N.C., H.B.), Fourth Military Medical University, Xi'an, China
| | - Huijie Bian
- Department of Cell Biology, National Translational Science Center for Molecular Medicine (J.-J.L., H.W., C.Z., T.-J.Z., H.-L.W., Z.-K.L., Y.-H.M., Q.H., L.-J.W., Z.-N.C., H.B.), Fourth Military Medical University, Xi'an, China
| |
Collapse
|
5
|
Yao C, Liu X, Tang Y, Wang C, Duan C, Liu X, Chen M, Zhou Y, Tang E, Xiang Y, Li Y, Ji A, Cai T. Lipopolysaccharide induces inflammatory microglial activation through CD147-mediated matrix metalloproteinase expression. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2023; 30:35352-35365. [PMID: 36534246 PMCID: PMC9761036 DOI: 10.1007/s11356-022-24292-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Accepted: 11/14/2022] [Indexed: 06/17/2023]
Abstract
Microglia-mediated neuroinflammation plays a vital role in the pathophysiological processes of multiple neurodegenerative diseases. Lipopolysaccharide (LPS) is an environmental poison that can induce inflammatory microglial activation. Matrix metalloproteinases (MMPs) are vital factors regulating microglial activation, and CD147 is a key MMP inducer, which can induce inflammation by inducing MMPs. However, whether it is involved in the regulation of microglial activation has not been reported. In this study, the role of CD147 in LPS-induced microglial inflammatory activation was investigated by establishing in vivo and in vitro models. The results suggested that LPS-induced microglial activation was accompanied by the induction of CD147 expression while the inhibition of CD147 expression could inhibit LPS-induced microglial inflammatory activation. In addition, the results also indicated that the role of CD147 in LPS-induced pro-inflammatory activation of microglia was related to its downstream MMP-3, MMP-8, and autophagy. Furthermore, the inhibition of MMP-3, MMP-8, and autophagy attenuated LPS-induced inflammatory activation of microglia. At the same time, there was a certain interaction between MMPs and autophagy, which is shown that inhibiting the expression of MMPs could inhibit autophagy, whereas inhibiting autophagy could inhibit the expression of MMPs. Taken together, we provided the first evidence that CD147/MMPs can be involved in LPS-induced inflammatory activation of microglia through an autophagy-dependent manner.
Collapse
Affiliation(s)
- Chunyan Yao
- Department of Epidemiology, College of Preventive Medicine, Army Medical University, Third Military Medical University), Chongqing, China
| | - Xiaoling Liu
- Department of Epidemiology, College of Preventive Medicine, Army Medical University, Third Military Medical University), Chongqing, China
| | - Yan Tang
- Experimental Teaching Center, School of Public Health, Southwest Medical University, Luzhou, China
| | - Chunmei Wang
- Experimental Teaching Center, School of Public Health, Southwest Medical University, Luzhou, China
| | - Chenggang Duan
- Department of Pathophysiology, Southwest Medical University, Luzhou, China
| | - Xiaoyan Liu
- The Research Center for Preclinical Medicine, Southwest Medical University, Luzhou, China
| | - Mingliang Chen
- Department of Chemical Defense Medicine, College of Preventive Medicine, Army Medical University, Third Military Medical University), Chongqing, China
| | - Yumeng Zhou
- Department of Epidemiology, College of Preventive Medicine, Army Medical University, Third Military Medical University), Chongqing, China
| | - Enjie Tang
- Department of Epidemiology, College of Preventive Medicine, Army Medical University, Third Military Medical University), Chongqing, China
| | - Ying Xiang
- Department of Epidemiology, College of Preventive Medicine, Army Medical University, Third Military Medical University), Chongqing, China
| | - Yafei Li
- Department of Epidemiology, College of Preventive Medicine, Army Medical University, Third Military Medical University), Chongqing, China
| | - Ailing Ji
- Department of Preventive Medicine & Chongqing Engineering Research Center of Pharmaceutical Sciences, Chongqing Medical and Pharmaceutical College, Chongqing, China
| | - Tongjian Cai
- Department of Epidemiology, College of Preventive Medicine, Army Medical University, Third Military Medical University), Chongqing, China.
| |
Collapse
|
6
|
Han JM, Jung HJ. Cyclophilin A/CD147 Interaction: A Promising Target for Anticancer Therapy. Int J Mol Sci 2022; 23:ijms23169341. [PMID: 36012604 PMCID: PMC9408992 DOI: 10.3390/ijms23169341] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Revised: 08/12/2022] [Accepted: 08/18/2022] [Indexed: 11/16/2022] Open
Abstract
Cyclophilin A (CypA), which has peptidyl-prolyl cis-trans isomerase (PPIase) activity, regulates multiple functions of cells by binding to its extracellular receptor CD147. The CypA/CD147 interaction plays a crucial role in the progression of several diseases, including inflammatory diseases, coronavirus infection, and cancer, by activating CD147-mediated intracellular downstream signaling pathways. Many studies have identified CypA and CD147 as potential therapeutic targets for cancer. Their overexpression promotes growth, metastasis, therapeutic resistance, and the stem-like properties of cancer cells and is related to the poor prognosis of patients with cancer. This review aims to understand the biology and interaction of CypA and CD147 and to review the roles of the CypA/CD147 interaction in cancer pathology and the therapeutic potential of targeting the CypA/CD147 axis. To validate the clinical significance of the CypA/CD147 interaction, we analyzed the expression levels of PPIA and BSG genes encoding CypA and CD147, respectively, in a wide range of tumor types using The Cancer Genome Atlas (TCGA) database. We observed a significant association between PPIA/BSG overexpression and poor prognosis, such as a low survival rate and high cancer stage, in several tumor types. Furthermore, the expression of PPIA and BSG was positively correlated in many cancers. Therefore, this review supports the hypothesis that targeting the CypA/CD147 interaction may improve treatment outcomes for patients with cancer.
Collapse
Affiliation(s)
- Jang Mi Han
- Department of Life Science and Biochemical Engineering, Graduate School, Sun Moon University, Asan 31460, Korea
| | - Hye Jin Jung
- Department of Life Science and Biochemical Engineering, Graduate School, Sun Moon University, Asan 31460, Korea
- Department of Pharmaceutical Engineering and Biotechnology, Sun Moon University, Asan 31460, Korea
- Genome-Based BioIT Convergence Institute, Sun Moon University, Asan 31460, Korea
- Correspondence: ; Tel.: +82-41-530-2354; Fax: +82-41-530-2939
| |
Collapse
|