1
|
Shen Z, Zhao M, Lu J, Chen H, Zhang Y, Chen S, Wang Z, Wang M, Liu X, Fu G, Huang H. Integrated multi-omic high-throughput strategies across-species identified potential key diagnostic, prognostic, and therapeutic targets for atherosclerosis under high glucose conditions. Mol Cell Biochem 2025; 480:1785-1805. [PMID: 39223351 DOI: 10.1007/s11010-024-05097-8] [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: 03/05/2024] [Accepted: 08/18/2024] [Indexed: 09/04/2024]
Abstract
Diabetes is a well-known risk factor for atherosclerosis (AS), but the underlying molecular mechanism remains unknown. The dysregulated immune response is an important reason. High glucose is proven to induce foam cell formation under lipidemia situations in clinical patients. Exploring the potential regulatory programs of accelerated foam cell formation stimulated by high glucose is meaningful. Macrophage-derived foam cells were induced in vitro, and high-throughput sequencing was performed. Coexpression gene modules were constructed using weighted gene co-expression network analysis (WGCNA). Highly related modules were identified. Hub genes were identified by multiple integrative strategies. The potential roles of selected genes were further validated in bulk-RNA and scRNA datasets of human plaques. By transfection of the siRNA, the role of the screened gene during foam cell formation was further explored. Two modules were found to be both positively related to high glucose and ox-LDL. Further enrichment analyses confirmed the association between the brown module and AS. The high correlation between the brown module and macrophages was identified and 4 hub genes (Aldoa, Creg1, Lgmn, and Pkm) were screened. Further validation in external bulk-RNA and scRNA revealed the potential diagnostic and therapeutic value of selected genes. In addition, the survival analysis confirmed the prognostic value of Aldoa while knocking down Aldoa expression alleviated the foam cell formation in vitro. We systematically investigated the synergetic effects of high glucose and ox-LDL during macrophage-derived foam cell formation and identified that ALDOA might be an important diagnostic, prognostic, and therapeutic target in these patients.
Collapse
Affiliation(s)
- Zhida Shen
- Key Laboratory of Cardiovascular Intervention and Regenerative Medicine of Zhejiang Province, Department of Cardiology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, 310020, Zhejiang, China
- Engineering Research Center for Cardiovascular Innovative Devices of Zhejiang Province, Sir Run Run Shaw Hospital, Hangzhou, China
| | - Meng Zhao
- Key Laboratory of Cardiovascular Intervention and Regenerative Medicine of Zhejiang Province, Department of Cardiology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, 310020, Zhejiang, China
| | - Jiangting Lu
- Key Laboratory of Cardiovascular Intervention and Regenerative Medicine of Zhejiang Province, Department of Cardiology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, 310020, Zhejiang, China
| | - Huanhuan Chen
- Department of General Surgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, 310020, Zhejiang, China
| | - Yicheng Zhang
- Department of Neurosurgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, 310020, Zhejiang, China
| | - Songzan Chen
- Key Laboratory of Cardiovascular Intervention and Regenerative Medicine of Zhejiang Province, Department of Cardiology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, 310020, Zhejiang, China
| | - Zhaojing Wang
- Key Laboratory of Cardiovascular Intervention and Regenerative Medicine of Zhejiang Province, Department of Cardiology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, 310020, Zhejiang, China
| | - Meihui Wang
- Key Laboratory of Cardiovascular Intervention and Regenerative Medicine of Zhejiang Province, Department of Cardiology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, 310020, Zhejiang, China
| | - Xianglan Liu
- Key Laboratory of Cardiovascular Intervention and Regenerative Medicine of Zhejiang Province, Department of Cardiology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, 310020, Zhejiang, China
| | - Guosheng Fu
- Key Laboratory of Cardiovascular Intervention and Regenerative Medicine of Zhejiang Province, Department of Cardiology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, 310020, Zhejiang, China.
- Engineering Research Center for Cardiovascular Innovative Devices of Zhejiang Province, Sir Run Run Shaw Hospital, Hangzhou, China.
| | - He Huang
- Key Laboratory of Cardiovascular Intervention and Regenerative Medicine of Zhejiang Province, Department of Cardiology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, 310020, Zhejiang, China.
- Engineering Research Center for Cardiovascular Innovative Devices of Zhejiang Province, Sir Run Run Shaw Hospital, Hangzhou, China.
| |
Collapse
|
2
|
Dogan Z, Senyigit A, Durmus S, Duvarcı C, Gelişgen R, Uzun H, Tabak O. The relationship between oxLDL, sLOX-1, PCSK9 and carotid intima-media thickness in patients with prediabetes and type 2 diabetes. Sci Rep 2025; 15:4554. [PMID: 39915665 PMCID: PMC11802860 DOI: 10.1038/s41598-025-89220-9] [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: 11/09/2024] [Accepted: 02/04/2025] [Indexed: 02/09/2025] Open
Abstract
Affection of vascular structures is a known complication in diabetes and prediabetes. Subclinical atherosclerosis begins to develop years before both cardiovascular and cerebrovascular diseases become symptomatic. The aim of this study was to evaluate the relationship between circulating oxidized LDL (oxLDL), soluble lectin-like oxidized LDL (sLOX)-1 and proprotein convertase subtilisin/kexin type 9 (PCSK9) levels and carotid intima-media thickness (CIMT) in patients with prediabetes and T2DM. We recruited 50 T2DM patients with macrovascular complications, 50 T2DM patients with uncomplicated, 50 prediabetes patients, and 50 healthy participants. The common carotid arteries were visualized by high-resolution B-mode carotid artery ultrasonography. Measurement of serum oxLDL, sLOX-1 and PCSK9 levels were assessed by using a commercially available human enzyme-linked immunosorbent assay (ELISA) method. Homeostasis model assessment for insulin resistance (HOMA-IR) was also calculated. Circulating LDL-C, oxLDL, sLOX-1 and PCSK9 levels were significantly higher in T2DM patients with macrovascular complications compared to control group, prediabetes and uncomplicated diabetes. There was significant difference, especially between the controls and the T2DM patients with macrovascular complications. The CIMT increased progressively from control through to T2DM. Both right and left CIMT also showed significant differences between the groups of prediabetes versus uncomplicated diabetes at p < 0.001 showing progressive increase in vascular involvement with progression of disease. There was a positive correlation between PCSK9 levels with LDL-C, oxLDL, sLOX-1, and CIMT. ROC curve analyses showed that PCSK9, as well as oxLDL and LOX-1, was a strong predictor of T2DM with high sensitivity and specificity. In this study, we found that oxLDL, sLOX-1 and PCSK9 levels were significantly associated with carotid intima-media thickness. Our findings suggest that these biomarkers may be used as potential markers for the assessment of cardiovascular risk in patients with prediabetes and type 2 diabetes.
Collapse
Affiliation(s)
- Zeki Dogan
- Department of Cardiology, Faculty of Medicine, Istanbul Atlas University, Istanbul, Turkey.
| | - Abdulhalim Senyigit
- Department of Internal Medicine, Faculty of Medicine, Istanbul Atlas University, Istanbul, Turkey
| | - Sinem Durmus
- Department of Medical Biochemistry, Faculty of Medicine, Katip Celebi University, Izmir, Turkey
| | - Canan Duvarcı
- Department of Medical Biochemistry, Cerrahpasa Faculty of Medicine, Istanbul University- Cerrahpasa, Istanbul, Turkey
| | - Remise Gelişgen
- Department of Medical Biochemistry, Cerrahpasa Faculty of Medicine, Istanbul University- Cerrahpasa, Istanbul, Turkey
| | - Hafize Uzun
- Department of Medical Biochemistry, Faculty of Medicine, Istanbul Atlas University, Istanbul, Turkey
| | - Omur Tabak
- Department of Internal Medicine, Kanuni Sultan Suleyman Training and Research Hospital, Health Sciences University, Istanbul, Turkey
| |
Collapse
|
3
|
Aziz M, Jandeleit-Dahm KA, Khan AW. Interplay between epigenetic mechanisms and transcription factors in atherosclerosis. Atherosclerosis 2024; 395:117615. [PMID: 38917706 DOI: 10.1016/j.atherosclerosis.2024.117615] [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: 12/03/2023] [Revised: 05/26/2024] [Accepted: 06/04/2024] [Indexed: 06/27/2024]
Abstract
Cardiovascular diseases (CVD), including coronary heart disease and stroke, comprise the number one cause of mortality worldwide. A major contributor to CVD is atherosclerosis, which is a low-grade inflammatory disease of vasculature that involves a pathological build-up of plaque within the arterial walls. Studies have shown that regulation of gene expression via transcription factors and epigenetic mechanisms play a fundamental role in transcriptomic changes linked to the development of atherosclerosis. Chromatin remodeling is a reversible phenomenon and studies have supported the clinical application of chromatin-modifying agents for the prevention and treatment of CVD. In addition, pre-clinical studies have identified multiple transcription factors as potential therapeutic targets in combating atherosclerotic CVD. Although interaction between transcription factors and epigenetic mechanisms facilitate gene regulation, a limited number of studies appreciate this crosstalk in the context of CVD. Here, we reviewed this gene regulatory mechanism underappreciated in atherosclerosis, which will highlight the mechanisms underlying novel therapeutics targeting epigenetic modifiers and transcription factors in atherosclerosis.
Collapse
Affiliation(s)
- Misbah Aziz
- Department of Diabetes, Central Clinical School, Monash University, Melbourne, Australia
| | - Karin Am Jandeleit-Dahm
- Department of Diabetes, Central Clinical School, Monash University, Melbourne, Australia; German Diabetes Centre, Leibniz Centre for Diabetes Research at the Heinrich Heine University, Dusseldorf, Germany
| | - Abdul Waheed Khan
- Department of Diabetes, Central Clinical School, Monash University, Melbourne, Australia.
| |
Collapse
|
4
|
Sánchez-León ME, Loaeza-Reyes KJ, Matias-Cervantes CA, Mayoral-Andrade G, Pérez-Campos EL, Pérez-Campos-Mayoral L, Hernández-Huerta MT, Zenteno E, Pérez-Cervera Y, Pina-Canseco S. LOX-1 in Cardiovascular Disease: A Comprehensive Molecular and Clinical Review. Int J Mol Sci 2024; 25:5276. [PMID: 38791315 PMCID: PMC11121106 DOI: 10.3390/ijms25105276] [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: 04/03/2024] [Revised: 05/06/2024] [Accepted: 05/08/2024] [Indexed: 05/26/2024] Open
Abstract
LOX-1, ORL-1, or lectin-like oxidized low-density lipoprotein receptor 1 is a transmembrane glycoprotein that binds and internalizes ox-LDL in foam cells. LOX-1 is the main receptor for oxidized low-density lipoproteins (ox-LDL). The LDL comes from food intake and circulates through the bloodstream. LOX-1 belongs to scavenger receptors (SR), which are associated with various cardiovascular diseases. The most important and severe of these is the formation of atherosclerotic plaques in the intimal layer of the endothelium. These plaques can evolve into complicated thrombi with the participation of fibroblasts, activated platelets, apoptotic muscle cells, and macrophages transformed into foam cells. This process causes changes in vascular endothelial homeostasis, leading to partial or total obstruction in the lumen of blood vessels. This obstruction can result in oxygen deprivation to the heart. Recently, LOX-1 has been involved in other pathologies, such as obesity and diabetes mellitus. However, the development of atherosclerosis has been the most relevant due to its relationship with cerebrovascular accidents and heart attacks. In this review, we will summarize findings related to the physiologic and pathophysiological processes of LOX-1 to support the detection, diagnosis, and prevention of those diseases.
Collapse
Affiliation(s)
- Maria Eugenia Sánchez-León
- Centro de Investigación Facultad de Medicina-UNAM-UABJO, Universidad Autónoma “Benito Juárez” de Oaxaca, Oaxaca 68020, Mexico; (M.E.S.-L.); (K.J.L.-R.); (C.A.M.-C.); (G.M.-A.); (L.P.-C.-M.)
| | - Karen Julissa Loaeza-Reyes
- Centro de Investigación Facultad de Medicina-UNAM-UABJO, Universidad Autónoma “Benito Juárez” de Oaxaca, Oaxaca 68020, Mexico; (M.E.S.-L.); (K.J.L.-R.); (C.A.M.-C.); (G.M.-A.); (L.P.-C.-M.)
- Centro de Estudios en Ciencias de la Salud y la Enfermedad, Facultad de Odontología, Universidad Autónoma “Benito Juárez” de Oaxaca, Oaxaca 68020, Mexico
| | - Carlos Alberto Matias-Cervantes
- Centro de Investigación Facultad de Medicina-UNAM-UABJO, Universidad Autónoma “Benito Juárez” de Oaxaca, Oaxaca 68020, Mexico; (M.E.S.-L.); (K.J.L.-R.); (C.A.M.-C.); (G.M.-A.); (L.P.-C.-M.)
| | - Gabriel Mayoral-Andrade
- Centro de Investigación Facultad de Medicina-UNAM-UABJO, Universidad Autónoma “Benito Juárez” de Oaxaca, Oaxaca 68020, Mexico; (M.E.S.-L.); (K.J.L.-R.); (C.A.M.-C.); (G.M.-A.); (L.P.-C.-M.)
| | | | - Laura Pérez-Campos-Mayoral
- Centro de Investigación Facultad de Medicina-UNAM-UABJO, Universidad Autónoma “Benito Juárez” de Oaxaca, Oaxaca 68020, Mexico; (M.E.S.-L.); (K.J.L.-R.); (C.A.M.-C.); (G.M.-A.); (L.P.-C.-M.)
| | - María Teresa Hernández-Huerta
- Consejo Nacional de Humanidades, Ciencias y Tecnologías, Facultad de Medicina y Cirugía, Universidad Autónoma “Benito Juárez” de Oaxaca, Oaxaca 68120, Mexico;
| | - Edgar Zenteno
- Facultad de Medicina, Universidad Nacional Autónoma de México, Mexico City 04510, Mexico;
| | - Yobana Pérez-Cervera
- Centro de Investigación Facultad de Medicina-UNAM-UABJO, Universidad Autónoma “Benito Juárez” de Oaxaca, Oaxaca 68020, Mexico; (M.E.S.-L.); (K.J.L.-R.); (C.A.M.-C.); (G.M.-A.); (L.P.-C.-M.)
- Centro de Estudios en Ciencias de la Salud y la Enfermedad, Facultad de Odontología, Universidad Autónoma “Benito Juárez” de Oaxaca, Oaxaca 68020, Mexico
| | - Socorro Pina-Canseco
- Centro de Investigación Facultad de Medicina-UNAM-UABJO, Universidad Autónoma “Benito Juárez” de Oaxaca, Oaxaca 68020, Mexico; (M.E.S.-L.); (K.J.L.-R.); (C.A.M.-C.); (G.M.-A.); (L.P.-C.-M.)
| |
Collapse
|
5
|
Cervantes J, Kanter JE. Monocyte and macrophage foam cells in diabetes-accelerated atherosclerosis. Front Cardiovasc Med 2023; 10:1213177. [PMID: 37378396 PMCID: PMC10291141 DOI: 10.3389/fcvm.2023.1213177] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Accepted: 05/30/2023] [Indexed: 06/29/2023] Open
Abstract
Diabetes results in an increased risk of atherosclerotic cardiovascular disease. This minireview will discuss whether monocyte and macrophage lipid loading contribute to this increased risk, as monocytes and macrophages are critically involved in the progression of atherosclerosis. Both uptake and efflux pathways have been described as being altered by diabetes or conditions associated with diabetes, which may contribute to the increased accumulation of lipids seen in macrophages in diabetes. More recently, monocytes have also been described as lipid-laden in response to elevated lipids, including triglyceride-rich lipoproteins, the class of lipids often elevated in the setting of diabetes.
Collapse
Affiliation(s)
| | - Jenny E. Kanter
- Department of Medicine, UW Medicine Diabetes Institute, University of Washington, Seattle, WA, United States
| |
Collapse
|
6
|
Wang J, Han Y, Huang F, Tang L, Mu J, Liang Y. Diabetic macrophage small extracellular vesicles-associated miR-503/IGF1R axis regulates endothelial cell function and affects wound healing. Front Immunol 2023; 14:1104890. [PMID: 37287964 PMCID: PMC10243549 DOI: 10.3389/fimmu.2023.1104890] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Accepted: 04/28/2023] [Indexed: 06/09/2023] Open
Abstract
Diabetic foot ulcer (DFU) is a break in the skin of the foot caused by diabetes. It is one of the most serious and debilitating complications of diabetes. The previous study suggested that dominant M1 polarization during DFU could be the leading reason behind impaired wound healing. This study concluded that macrophage M1 polarization predominates in DFU skin tissue. iNOS was increased in HG-induced M1-polarized macrophages; conversely, Arg-1 was decreased. Macrophage pellets after HG stimulation can impair endothelial cell (EC) function by inhibiting cell viability, tube formation and cell migration, indicating M1 macrophage-derived small extracellular vesicles (sEVs) -mediated HUVEC dysfunction. sEVs miR-503 was significantly upregulated in response to HG stimulation, but inhibition of miR-503 in HG-stimulated macrophages attenuated M1 macrophage-induced HUVEC dysfunction. ACO1 interacted with miR-503 and mediated the miR-503 package into sEVs. Under HG stimulation, sEVs miR-503 taken in by HUVECs targeted IGF1R in HUVECs and inhibited IGF1R expression. In HUVECs, miR-503 inhibition improved HG-caused HUVEC dysfunction, whereas IGF1R knockdown aggravated HUVEC dysfunction; IGF1R knockdown partially attenuated miR-503 inhibition effects on HUVECs. In the skin wound model in control or STZ-induced diabetic mice, miR-503-inhibited sEVs improved, whereas IGF1R knockdown further hindered wound healing. Therefore, it can be inferred from the results that the M1 macrophage-derived sEVs miR-503 targets IGF1R in HUVECs, inhibits IGF1R expression, leads to HUVEC dysfunction, and impedes wound healing in diabetic patients, while packaging miR-503 as an M1 macrophage-derived sEVs may be mediated by ACO1.
Collapse
Affiliation(s)
- Jianqiang Wang
- Molecular Nutrition Branch, National Engineering Research Center of Rice and By-Product Deep Processing/College of Food Science and Engineering, Central South University of Forestry and Technology, Changsha, Hunan, China
| | - Yuanshan Han
- Scientific Research Department, The First Hospital of Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Fang Huang
- Molecular Nutrition Branch, National Engineering Research Center of Rice and By-Product Deep Processing/College of Food Science and Engineering, Central South University of Forestry and Technology, Changsha, Hunan, China
| | - Liuhuan Tang
- Molecular Nutrition Branch, National Engineering Research Center of Rice and By-Product Deep Processing/College of Food Science and Engineering, Central South University of Forestry and Technology, Changsha, Hunan, China
| | - Jianfei Mu
- Molecular Nutrition Branch, National Engineering Research Center of Rice and By-Product Deep Processing/College of Food Science and Engineering, Central South University of Forestry and Technology, Changsha, Hunan, China
| | - Ying Liang
- Molecular Nutrition Branch, National Engineering Research Center of Rice and By-Product Deep Processing/College of Food Science and Engineering, Central South University of Forestry and Technology, Changsha, Hunan, China
| |
Collapse
|
7
|
Lin MH, Cheng PC, Hsiao PJ, Chen SC, Hung CH, Kuo CH, Huang SK, Clair Chiou HY. The GLP-1 receptor agonist exenatide ameliorates neuroinflammation, locomotor activity, and anxiety-like behavior in mice with diet-induced obesity through the modulation of microglial M2 polarization and downregulation of SR-A4. Int Immunopharmacol 2023; 115:109653. [PMID: 36587502 DOI: 10.1016/j.intimp.2022.109653] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2022] [Revised: 12/16/2022] [Accepted: 12/26/2022] [Indexed: 12/31/2022]
Abstract
Obesity is associated with multiple comorbidities, such as metabolic abnormalities and cognitive dysfunction. Moreover, accumulating evidence indicates that neurodegenerative disorders are associated with chronic neuroinflammation. GLP-1 receptor agonists (RAs) have been extensively studied as a treatment for type 2 diabetes. Emerging evidence has demonstrated a protective effect of GLP-1 RAs on neurodegenerative disease, which is independent of its glucose-lowering effects. In this study, we aimed to examine the effects of a long-acting GLP-1 RA, exenatide, on high-fat diet (HFD)-induced neuroinflammation and related brain function impairment. First, mice treated with exenatide exhibited significantly reduced HFD-increased body weight and blood glucose. In an open field test, exenatide treatment ameliorated the reduction in local motor activity and anxiety in HFD-fed mice. Moreover, HFD induced astrogliosis, microgliosis, and upregulation of IL-1β, IL-6 and TNF-α in hippocampus and cortex. Exenatide treatment reduced HFD-induced astrogliosis and IL-1β and TNF-α expressions. Moreover, exenatide increased phosphor-ERK and M2-type microglia marker arginase-1 expression in the hippocampus and cortex. In addition, we found that scavenger receptor-A4 protein expression was induced by HFD and was subsequently inhibited by exenatide. SR-A4 knockout reversed the locomotor activity impairment but not the anxiety behavior caused by HFD consumption. SR-A4 knockout also reduced HFD-induced neuroinflammation, as shown by the reduced expression of GFAP and IBA-1 compared with that in wild-type control mice. These results demonstrate that exenatide decreases HFD-increased neuroinflammation and promotes anti-inflammatory M2 differentiation. The inhibition of SR-A4 by exenatide exerts anti-inflammatory activity.
Collapse
Affiliation(s)
- Ming-Hong Lin
- Department of Microbiology and Immunology, School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan; Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan; M.Sc. Program in Tropical Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan; Department of Medical Research, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 807, Taiwan.
| | - Po-Ching Cheng
- Department of Molecular Parasitology and Tropical Diseases, School of Medicine, College of Medicine, Taipei Medical University, Taipei 110, Taiwan; Center for International Tropical Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei 110, Taiwan.
| | - Pi-Jung Hsiao
- Division of Endocrinology and Metabolism, E-Da Hospital, I-Shou University, Kaohsiung, Taiwan; School of Medicine, College of Medicine, I-Shou University, Kaohsiung, Taiwan.
| | - Szu-Chia Chen
- Department of Internal Medicine, Kaohsiung Municipal Siaogang Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan; Division of Nephrology, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan; Research Center for Environmental Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan.
| | - Chih-Hsing Hung
- Research Center for Environmental Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan; Department of Pediatrics, School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan; Department of Pediatrics, Kaohsiung Municipal Siaogang Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan.
| | - Chao-Hung Kuo
- Department of Internal Medicine, Kaohsiung Municipal Siaogang Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan; Division of Gastroenterology, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan.
| | - Shau-Ku Huang
- National Institute of Environmental Health Sciences, National Health Research Institutes, Miaoli County 350, Taiwan; Department of Medicine, Division of Allergy and Clinical Immunology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.
| | - Hsin-Ying Clair Chiou
- Center of Teaching and Research, Kaohsiung Municipal Siaogang Hospital, Kaohsiung Medical University, Kaohsiung 812, Taiwan; Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 807, Taiwan; Department of Applied Chemistry, National Chi Nan University, Nantou, Taiwan.
| |
Collapse
|
8
|
Inflammatory gene silencing in activated monocytes by a cholesterol tagged-miRNA/siRNA: a novel approach to ameliorate diabetes induced inflammation. Cell Tissue Res 2022; 389:219-240. [PMID: 35604451 DOI: 10.1007/s00441-022-03637-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2021] [Accepted: 05/09/2022] [Indexed: 11/02/2022]
Abstract
There is a major unmet need for the development of effective therapies for diabetes induced inflammation. Increased adenosine-uridine rich elements (AREs) containing mRNAs of inflammatory molecules are reported in inflamed monocytes. Destabilizing these inflammatory mRNAs by the miR-16 could reduce inflammation. DNA microarrays and in vitro cell studies showed that exogenous miR16 and its mimic treatment, in LPS/PMA induced monocytes, significantly downregulated several ARE containing inflammatory cytokine mRNAs similar to those seen in the normal monocytes. Ingenuity pathway analyses showed exogenous miR-16 or its synthetic mimic treatment alleviates inflammatory responses. To selectively target uptake, especially to inflamed cells, one of the CD36 substrate cholesterol was tagged to miR16/siRNA. Cholesterol tagged miR-16/ARE-siRNA showed enhanced uptake in CD36 expressing inflamed cells. In LPS or PMA, treated monocytes, candidate genes expressions levels such as IL-6, IL-8, IL-12β, IP-10, and TNF-α mRNA were increased, as measured by RT-qPCR as seen in primary monocytes of diabetes patients. Exogenous miR16 or ARE-siRNA transfection reduced mRNAs of pro-inflammatory cytokines levels in monocyte, and its adhesion. Increased uptake of cholesterol tagged miR-16 through the CD36 receptor was observed. This destabilizes numerous inflammatory ARE containing mRNAs and alleviates inflammatory responses. Cholesterol-tagged miR-16 and its mimic are novel anti-inflammatory molecules that can be specifically targeted to, via through CD36 expressing, "inflamed" cells and thus serve as therapeutic candidates to alleviate inflammatory diseases.
Collapse
|
9
|
Li X, Feng JL, Chen ZL, Bao BH, Dai HH, Meng FC, Deng S, Wang B, Li HS, Wang JS. Mechanism by which Huoxue Tongluo Qiwei Decoction improves the erectile function of rats with diabetic erectile dysfunction. JOURNAL OF ETHNOPHARMACOLOGY 2022; 283:114674. [PMID: 34560214 DOI: 10.1016/j.jep.2021.114674] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Revised: 09/19/2021] [Accepted: 09/20/2021] [Indexed: 06/13/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Huoxue Tongluo Qiwei Decoction is a classical herbal formula, which can improve the symptoms of erectile dysfunction (ED) patients and has a good therapeutic effect on patients with diabetic erectile dysfunction (DIED). The main function of Huoxue Tongluo Qiwei Decoction is to stimulate the blood circulation and dredge collaterals, remove blood stasis, and calm wind. RATIONALE To further explore the mechanism of Huoxue Tongluo Qiwei Decoction in the treatment of DIED, related animal experiments were designed. MATERIALS AND METHODS The chemical constituents of Huoxue Tongluo Qiwei Decoction were identified with the help of high-performance liquid chromatography-tandem mass spectrometry (HPLC-MS/MS). A rat model was induced by streptozotocin (STZ) and screened by apomorphine (APO). Serum sE-selectin, lysyl oxidase-1 (LOX-1), malondialdehyde (MDA) and other markers of vascular endothelial injury and related indicators of oxidative stress were studied through enzyme-linked immunosorbent assay (ELISA). The endothelial cells and ultrastructure of the corpus cavernosum were examined by electron microscopy and HE staining. The expression of protein and mRNA was detected by western blotting (WB) and real-time quantitative polymerase chain reaction (RT-qPCR). RESULTS The results of the study revealed that the sE-selectin, LOX-1, intercellular adhesion molecule-1 (sICAM-1), endothelial microparticles (EMPs), P-selectin (CD62P), and MDA levels in the serum of group M rats were considerably higher than rats of group K, while the superoxide dismutase (SOD) level showed a significant decrease. In addition, the PKC pathway was activated, and the expression of related proteins and mRNA was increased. After 8 weeks of intervention with Huoxue Tongluo Qiwei Decoction and LY333531, serum level of sE-selectin, LOX-1, sICAM-1, EMPs, CD62P and MDA in L, D and G groups were remarkably lower than group M while SOD level increased significantly, protein kinase C (PKC) pathway was inhibited with the improved erectile function of rats. CONCLUSION Huoxue Tongluo Qiwei Decoction can inhibit the expression of protein and mRNA of the PKCβ signaling pathway related molecules in DIED rats to cure the injury of vascular endothelial, enhance antioxidant capacity, and prevent the activation of platelet, thus improving erectile function in rats with DIED.
Collapse
Affiliation(s)
- Xiao Li
- The First Affiliated Hospital, Henan University of Chinese Medicine, Zhengzhou, 450000, China.
| | - Jun-Long Feng
- First Clinical Medical College, Beijing University of Chinese Medicine, Beijing, 100029, China.
| | - Zi-Long Chen
- First Clinical Medical College, Beijing University of Chinese Medicine, Beijing, 100029, China; Department of Andrology, Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, 518033, China.
| | - Bing-Hao Bao
- Department of Andrology, China Japan Friendship Hospital, Beijing, 100029, China.
| | - Heng-Heng Dai
- First Clinical Medical College, Beijing University of Chinese Medicine, Beijing, 100029, China.
| | - Fan-Chao Meng
- First Clinical Medical College, Beijing University of Chinese Medicine, Beijing, 100029, China.
| | - Sheng Deng
- First Clinical Medical College, Beijing University of Chinese Medicine, Beijing, 100029, China.
| | - Bin Wang
- First Clinical Medical College, Beijing University of Chinese Medicine, Beijing, 100029, China; Andrology Department, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, 100700, China.
| | - Hai-Song Li
- First Clinical Medical College, Beijing University of Chinese Medicine, Beijing, 100029, China; Andrology Department, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, 100700, China.
| | - Ji-Sheng Wang
- First Clinical Medical College, Beijing University of Chinese Medicine, Beijing, 100029, China; Andrology Department, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, 100700, China.
| |
Collapse
|
10
|
Calcium Dobesilate Modulates PKCδ-NADPH Oxidase- MAPK-NF-κB Signaling Pathway to Reduce CD14, TLR4, and MMP9 Expression during Monocyte-to-Macrophage Differentiation: Potential Therapeutic Implications for Atherosclerosis. Antioxidants (Basel) 2021; 10:antiox10111798. [PMID: 34829669 PMCID: PMC8615002 DOI: 10.3390/antiox10111798] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Accepted: 11/08/2021] [Indexed: 01/18/2023] Open
Abstract
Monocyte-to-macrophage differentiation results in the secretion of various inflammatory mediators and oxidative stress molecules necessary for atherosclerosis pathogenesis. Consequently, this differentiation represents a potential clinical target in atherosclerosis. Calcium dobesilate (CaD), an established vasoactive and angioprotective drug in experimental models of diabetic microvascular complications reduces oxidative stress and inhibits inflammation via diverse molecular targets; however, its effect on monocytes/macrophages is poorly understood. In this study, we investigated the anti-inflammatory mechanism of CaD during phorbol 12-myristate 13-acetate (PMA)-induced monocyte-to-macrophage differentiation in in vitro models of sepsis (LPS) and hyperglycemia, using THP-1 monocytic cell line. CaD significantly suppressed CD14, TLR4, and MMP9 expression and activity, lowering pro-inflammatory mediators, such as IL1β, TNFα, and MCP-1. The effects of CaD translated through to studies on primary human macrophages. CaD inhibited reactive oxygen species (ROS) generation, PKCδ, MAPK (ERK1/2 and p38) phosphorylation, NOX2/p47phox expression, and membrane translocation. We used hydrogen peroxide (H2O2) to mimic oxidative stress, demonstrating that CaD suppressed PKCδ activation via its ROS-scavenging properties. Taken together, we demonstrate for the first time that CaD suppresses CD14, TLR4, MMP9, and signature pro-inflammatory cytokines, in human macrophages, via the downregulation of PKCδ/NADPH oxidase/ROS/MAPK/NF-κB-dependent signaling pathways. Our data present novel mechanisms of how CaD alleviates metabolic and infectious inflammation.
Collapse
|
11
|
Barraclough JY, Patel S, Yu J, Neal B, Arnott C. The Role of Sodium Glucose Cotransporter-2 Inhibitors in Atherosclerotic Cardiovascular Disease: A Narrative Review of Potential Mechanisms. Cells 2021; 10:cells10102699. [PMID: 34685677 PMCID: PMC8534746 DOI: 10.3390/cells10102699] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Revised: 09/30/2021] [Accepted: 09/30/2021] [Indexed: 12/16/2022] Open
Abstract
Sodium glucose cotransporter 2 (SGLT2) inhibitors are a class of medication with broad cardiovascular benefits in those with type 2 diabetes, chronic kidney disease, and heart failure. These include reductions in major adverse cardiac events and cardiovascular death. The mechanisms that underlie their benefits in atherosclerotic cardiovascular disease (ASCVD) are not well understood, but they extend beyond glucose lowering. This narrative review summarises the ASCVD benefits of SGLT2 inhibitors seen in large human outcome trials, as well as the mechanisms of action explored in rodent and small human studies. Potential pathways include favourable alterations in lipid metabolism, inflammation, and endothelial function. These all require further investigation in large human clinical trials with mechanistic endpoints, to further elucidate the disease modifying benefits of this drug class and those who will benefit most from it.
Collapse
Affiliation(s)
- Jennifer Y. Barraclough
- The George Institute for Global Health, University of New South Wales, Sydney, NSW 2042, Australia; (J.Y.B.); (J.Y.); (B.N.)
- Department of Cardiology, Royal Prince Alfred Hospital, Sydney, NSW 2050, Australia;
| | - Sanjay Patel
- Department of Cardiology, Royal Prince Alfred Hospital, Sydney, NSW 2050, Australia;
- Sydney Medical School, University of Sydney, Sydney, NSW 2042, Australia
| | - Jie Yu
- The George Institute for Global Health, University of New South Wales, Sydney, NSW 2042, Australia; (J.Y.B.); (J.Y.); (B.N.)
| | - Bruce Neal
- The George Institute for Global Health, University of New South Wales, Sydney, NSW 2042, Australia; (J.Y.B.); (J.Y.); (B.N.)
| | - Clare Arnott
- The George Institute for Global Health, University of New South Wales, Sydney, NSW 2042, Australia; (J.Y.B.); (J.Y.); (B.N.)
- Department of Cardiology, Royal Prince Alfred Hospital, Sydney, NSW 2050, Australia;
- Sydney Medical School, University of Sydney, Sydney, NSW 2042, Australia
- Correspondence: ; Tel.: +61-2-8052-4300
| |
Collapse
|
12
|
Nyandwi JB, Ko YS, Jin H, Yun SP, Park SW, Kim HJ. Rosmarinic Acid Increases Macrophage Cholesterol Efflux through Regulation of ABCA1 and ABCG1 in Different Mechanisms. Int J Mol Sci 2021; 22:8791. [PMID: 34445501 PMCID: PMC8395905 DOI: 10.3390/ijms22168791] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Revised: 08/06/2021] [Accepted: 08/13/2021] [Indexed: 01/12/2023] Open
Abstract
Lipid dysregulation in diabetes mellitus escalates endothelial dysfunction, the initial event in the development and progression of diabetic atherosclerosis. In addition, lipid-laden macrophage accumulation in the arterial wall plays a significant role in the pathology of diabetes-associated atherosclerosis. Therefore, inhibition of endothelial dysfunction and enhancement of macrophage cholesterol efflux is the important antiatherogenic mechanism. Rosmarinic acid (RA) possesses beneficial properties, including its anti-inflammatory, antioxidant, antidiabetic and cardioprotective effects. We previously reported that RA effectively inhibits diabetic endothelial dysfunction by inhibiting inflammasome activation in endothelial cells. However, its effect on cholesterol efflux remains unknown. Therefore, in this study, we aimed to assess the effect of RA on cholesterol efflux and its underlying mechanisms in macrophages. RA effectively reduced oxLDL-induced cholesterol contents under high glucose (HG) conditions in macrophages. RA enhanced ATP-binding cassette transporter A1 (ABCA1) and G1 (ABCG1) expression, promoting macrophage cholesterol efflux. Mechanistically, RA differentially regulated ABCA1 expression through JAK2/STAT3, JNK and PKC-p38 and ABCG1 expression through JAK2/STAT3, JNK and PKC-ERK1/2/p38 in macrophages. Moreover, RA primarily stabilized ABCA1 rather than ABCG1 protein levels by impairing protein degradation. These findings suggest RA as a candidate therapeutic to prevent atherosclerotic cardiovascular disease complications related to diabetes by regulating cholesterol efflux in macrophages.
Collapse
Affiliation(s)
- Jean-Baptiste Nyandwi
- Department of Pharmacology, Institute of Health Sciences, College of Medicine, Gyeongsang National University, Jinju 52727, Korea; (J.-B.N.); (Y.S.K.); (H.J.); (S.P.Y.); (S.W.P.)
- Department of Convergence Medical Science (BK21 Plus), Gyeongsang National University, Jinju 52727, Korea
- Department of Pharmacy, School of Medicine and Pharmacy, College of Medicine and Health Sciences, University of Rwanda, Kigali 4285, Rwanda
| | - Young Shin Ko
- Department of Pharmacology, Institute of Health Sciences, College of Medicine, Gyeongsang National University, Jinju 52727, Korea; (J.-B.N.); (Y.S.K.); (H.J.); (S.P.Y.); (S.W.P.)
| | - Hana Jin
- Department of Pharmacology, Institute of Health Sciences, College of Medicine, Gyeongsang National University, Jinju 52727, Korea; (J.-B.N.); (Y.S.K.); (H.J.); (S.P.Y.); (S.W.P.)
| | - Seung Pil Yun
- Department of Pharmacology, Institute of Health Sciences, College of Medicine, Gyeongsang National University, Jinju 52727, Korea; (J.-B.N.); (Y.S.K.); (H.J.); (S.P.Y.); (S.W.P.)
| | - Sang Won Park
- Department of Pharmacology, Institute of Health Sciences, College of Medicine, Gyeongsang National University, Jinju 52727, Korea; (J.-B.N.); (Y.S.K.); (H.J.); (S.P.Y.); (S.W.P.)
- Department of Convergence Medical Science (BK21 Plus), Gyeongsang National University, Jinju 52727, Korea
| | - Hye Jung Kim
- Department of Pharmacology, Institute of Health Sciences, College of Medicine, Gyeongsang National University, Jinju 52727, Korea; (J.-B.N.); (Y.S.K.); (H.J.); (S.P.Y.); (S.W.P.)
- Department of Convergence Medical Science (BK21 Plus), Gyeongsang National University, Jinju 52727, Korea
| |
Collapse
|
13
|
Wang JS, Li X, Chen ZL, Feng JL, Bao BH, Deng S, Dai HH, Meng FC, Wang B, Li HS. Effect of leech-centipede medicine on improving erectile function in DIED rats via PKC signalling pathway-related molecules. JOURNAL OF ETHNOPHARMACOLOGY 2021; 267:113463. [PMID: 33049347 DOI: 10.1016/j.jep.2020.113463] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Revised: 10/05/2020] [Accepted: 10/07/2020] [Indexed: 06/11/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Leeches (pinyin name Shui Zhi; Latin scientific name Hirudo; Hirudinea; Hirudinidae) and centipedes (pinyin name Wu Gong; Latin scientific name Scolopendridae; Chilopoda; Scolopendridae) are traditional Chinese medicines, and they belong to the family entomology. A combination of leech and centipede is used as an effective medicine to promote blood circulation and remove blood stasis in traditional Chinese medicine, and "leech-centipede" medicine has been used in many prescriptions to treat diabetic vascular disease, including diabetic erectile dysfunction (DIED). However, its specific mechanism remains unclear and requires in-depth study. AIM OF THE STUDY This study aimed to investigate the mechanism of "leech-centipede" medicine to improve erectile dysfunction-associated diabetes by detecting PKC pathway-related molecules. MATERIALS AND METHODS The active ingredients of "leech-centipede" medicine were identified using high performance liquid chromatography (HPLC). Fifty male SPF rats were injected with streptozotocin to induce the DM model. Eight weeks later, the DMED model was validated with apomorphine. The DIED rats were divided into five groups-T,P,DD,DZ, and DG-and were separately treated with tadalafil, pathway inhibitor LY333531 and low-, medium-, and high-dose "leech-centipede" medicine for 8 weeks. After treatment, the blood glucose level was measured, erectile function with apomorphine was assessed, the LOX-1, sE-selectin, sICAM-1, SOD, and MDA in serum was evaluated by enzyme-linked immunosorbent assay, and flow cytometry was performed. After the collection of penile tissue, the related protein and mRNA expression was assessed by Western blotting and PCR, and the tissue and ultrastructure were analysed by HE staining, immunohistochemistry and scanning electron microscopy. RESULTS After treatment, the erectile function of rats was significantly improved in the T,P,DD,DZ, and DG groups compared with that in the model group. Thus, "leech-centipede" medicine can significantly reduce the levels of LOX-1, sE-selectin, sICAM-1, EMPs and CD62P to protect vascular endothelial function and anti-platelet activation, improving DIED rat erectile function. Additionally, "leech-centipede" medicine can increase SOD expression and decrease MDA expression, reducing the possibility of oxidative stress injury in DIED rats and improving the antioxidant capacity. Moreover, "leech-centipede" therapy can dramatically reduce the protein and mRNA expression of DAG, PKCβ, NF-κB, and ICAM-1, improve vascular endothelial injury in DIED rats and inhibit abnormal platelet activation. CONCLUSION "leech-centipede" medicine can improve erectile dysfunction by inhibiting the expression of PKC pathway-related molecules in DIED rats and protects endothelial function and anti-platelet activation.
Collapse
Affiliation(s)
- Ji-Sheng Wang
- First Clinical Medical College, Beijing University of Chinese Medicine, Beijing, 100029, China; Andrology Department, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, 100700, China.
| | - Xiao Li
- The First Affiliated Hospital, Henan University of Chinese Medicine, Zhengzhou, 450000, China.
| | - Zi-Long Chen
- First Clinical Medical College, Beijing University of Chinese Medicine, Beijing, 100029, China; Andrology Department, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, 100700, China.
| | - Jun-Long Feng
- First Clinical Medical College, Beijing University of Chinese Medicine, Beijing, 100029, China; Andrology Department, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, 100700, China.
| | - Bing-Hao Bao
- First Clinical Medical College, Beijing University of Chinese Medicine, Beijing, 100029, China; Andrology Department, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, 100700, China.
| | - Sheng Deng
- First Clinical Medical College, Beijing University of Chinese Medicine, Beijing, 100029, China; Andrology Department, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, 100700, China.
| | - Heng-Heng Dai
- First Clinical Medical College, Beijing University of Chinese Medicine, Beijing, 100029, China; Andrology Department, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, 100700, China.
| | - Fan-Chao Meng
- First Clinical Medical College, Beijing University of Chinese Medicine, Beijing, 100029, China; Andrology Department, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, 100700, China.
| | - Bin Wang
- Andrology Department, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, 100700, China.
| | - Hai-Song Li
- Andrology Department, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, 100700, China.
| |
Collapse
|
14
|
Mentrup T, Cabrera-Cabrera F, Schröder B. Proteolytic Regulation of the Lectin-Like Oxidized Lipoprotein Receptor LOX-1. Front Cardiovasc Med 2021; 7:594441. [PMID: 33553253 PMCID: PMC7856673 DOI: 10.3389/fcvm.2020.594441] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2020] [Accepted: 12/21/2020] [Indexed: 11/13/2022] Open
Abstract
The lectin-like oxidized-LDL (oxLDL) receptor LOX-1, which is broadly expressed in vascular cells, represents a key mediator of endothelial activation and dysfunction in atherosclerotic plaque development. Being a member of the C-type lectin receptor family, LOX-1 can bind different ligands, with oxLDL being the best characterized. LOX-1 mediates oxLDL uptake into vascular cells and by this means can promote foam cell formation. In addition, LOX-1 triggers multiple signaling pathways, which ultimately induce a pro-atherogenic and pro-fibrotic transcriptional program. However, the molecular mechanisms underlying this signal transduction remain incompletely understood. In this regard, proteolysis has recently emerged as a regulatory mechanism of LOX-1 function. Different proteolytic cleavages within the LOX-1 protein can initiate its turnover and control the cellular levels of this receptor. Thereby, cleavage products with individual biological functions and/or medical significance are produced. Ectodomain shedding leads to the release of a soluble form of the receptor (sLOX1) which has been suggested to have diagnostic potential as a biomarker. Removal of the ectodomain leaves behind a membrane-bound N-terminal fragment (NTF), which despite being devoid of the ligand-binding domain is actively involved in signal transduction. Degradation of this LOX-1 NTF, which represents an athero-protective mechanism, critically depends on the aspartyl intramembrane proteases Signal peptide peptidase-like 2a and b (SPPL2a/b). Here, we present an overview of the biology of LOX-1 focusing on how proteolytic cleavages directly modulate the function of this receptor and, what kind of pathophysiological implications this has in cardiovascular disease.
Collapse
Affiliation(s)
| | | | - Bernd Schröder
- Institute for Physiological Chemistry, Technische Universität Dresden, Dresden, Germany
| |
Collapse
|
15
|
AGE-RAGE Axis Stimulates Oxidized LDL Uptake into Macrophages through Cyclin-Dependent Kinase 5-CD36 Pathway via Oxidative Stress Generation. Int J Mol Sci 2020; 21:ijms21239263. [PMID: 33291667 PMCID: PMC7730944 DOI: 10.3390/ijms21239263] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Revised: 11/28/2020] [Accepted: 12/01/2020] [Indexed: 12/11/2022] Open
Abstract
Advanced glycation end products (AGEs) are localized in macrophage-derived foam cells within atherosclerotic lesions, which could be associated with the increased risk of atherosclerotic cardiovascular disease under diabetic conditions. Although foam cell formation of macrophages has been shown to be enhanced by AGEs, the underlying molecular mechanism remains unclear. Since cyclin-dependent kinase 5 (Cdk5) is reported to modulate inflammatory responses in macrophages, we investigated whether Cdk5 could be involved in AGE-induced CD36 gene expression and foam cell formation of macrophages. AGEs significantly increased Dil-oxidized low-density lipoprotein (ox-LDL) uptake, and Cdk5 and CD36 gene expression in U937 human macrophages, all of which were inhibited by DNA aptamer raised against RAGE (RAGE-aptamer). Cdk5 and CD36 gene expression levels were correlated with each other. An antioxidant, N-acetyl-l-cysteine, mimicked the effects of RAGE-aptamer on AGE-exposed U937 cells. A selective inhibitor of Cdk5, (R)-DRF053, attenuated the AGE-induced Dil-ox-LDL uptake and CD36 gene expression, whereas anti-CD36 antibody inhibited the Dil-ox-LDL uptake but not Cdk5 gene expression. The present study suggests that AGEs may stimulate ox-LDL uptake into macrophages through the Cdk5–CD36 pathway via RAGE-mediated oxidative stress.
Collapse
|
16
|
Liu H, Xu S, Li G, Lou D, Fu X, Lu Q, Hao L, Zhang J, Mei J, Sui Z, Lou Y. Sarpogrelate and rosuvastatin synergistically ameliorate aortic damage induced by hyperlipidemia in apolipoprotein E-deficient mice. Exp Ther Med 2020; 20:170. [PMID: 33093907 PMCID: PMC7571328 DOI: 10.3892/etm.2020.9300] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Accepted: 06/17/2020] [Indexed: 11/06/2022] Open
Abstract
The current study aimed to investigate whether sarpogrelate and rosuvastatin possess anti-arterial injury, and attempted to elucidate the mechanism of action underlying this activity. Sarpogrelate, a 5-hydroxytryptamine type 2A antagonist, is extensively used to prevent arterial thrombosis; however, its effects on atherosclerosis remain unknown. In the present study, sarpogrelate combined with rosuvastatin or rosuvastatin alone were administered to male ApoE-/- mice fed a high-fat diet (HFD) for 8 weeks. Metabolic parameters in the blood samples were analyzed using an automatic analyzer. Aortic tissues were stained with hematoxylin and eosin for morphological analysis. The expression levels of oxidized-low density lipoprotein (LDL) specific scavenging receptors, lectin-like oxidized low-density lipoprotein receptor-1 (LOX-1) and cluster of differentiation 68 were detected via immunostaining. mRNA expression levels of interleukin (IL)-1β, IL-6 and tumor necrosis factor-α were determined via reverse transcription-quantitative PCR analysis, while protein expression levels of LOX-1 and phosphor(p)-ERK were determined via western blot analysis. The results demonstrated that sarpogrelate combined with rosuvastatin treatment significantly decreased total cholesterol and LDL cholesterol levels in the serum, and alleviated intimal hyperplasia and lipid deposition, accompanied by decreased inflammatory cell infiltration and lower expression levels of inflammatory cytokines, compared with rosuvastatin monotherapy or HFD treatment. Furthermore, sarpogrelate combined with rosuvastatin treatment significantly decreased the expression levels of LOX-1 and p-ERK. Taken together, these results suggest that the positive effects of sarpogrelate combined with rosuvastatin treatment on aortic injury may be associated with the regulation of the LOX-1/p-ERK signaling pathway. Sarpogrelate and rosuvastatin synergistically decreased aortic damage in ApoE-/- HFD mice, and thus provide a basis for the treatment of aortic injury caused by hyperlipidemia with sarpogrelate.
Collapse
Affiliation(s)
- Hongyang Liu
- Department of Heart Intensive Care Unit, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning 116001, P.R. China
| | - Siwei Xu
- Department of Cardiology, Affiliated Zhongshan Hospital of Dalian University, Dalian, Liaoning 116001, P.R. China
| | - Guihua Li
- Department of Cardiology, The Second Affiliated Hospital of Dalian Medical University, Dalian, Liaoning 116023, P.R. China
| | - Dayuan Lou
- Department of Cardiology, The Second Affiliated Hospital of Dalian Medical University, Dalian, Liaoning 116023, P.R. China
| | - Xiaodan Fu
- Department of Cardiology, The Second Affiliated Hospital of Dalian Medical University, Dalian, Liaoning 116023, P.R. China
| | - Qin Lu
- Department of Cardiology, The Second Affiliated Hospital of Dalian Medical University, Dalian, Liaoning 116023, P.R. China
| | - Liman Hao
- Department of Cardiology, Jiche Hospital of Dalian, Dalian, Liaoning 116021, P.R. China
| | - Jingsi Zhang
- Department of Cardiology, The Second Affiliated Hospital of Dalian Medical University, Dalian, Liaoning 116023, P.R. China
| | - Jiajie Mei
- Department of Cardiology, The Second Affiliated Hospital of Dalian Medical University, Dalian, Liaoning 116023, P.R. China
| | - Zheng Sui
- Department of Cardiology, The Second Affiliated Hospital of Dalian Medical University, Dalian, Liaoning 116023, P.R. China
| | - Yu Lou
- Department of Cardiology, The Second Affiliated Hospital of Dalian Medical University, Dalian, Liaoning 116023, P.R. China
| |
Collapse
|
17
|
Chen S, Xia Y, He F, Fu J, Xin Z, Deng B, He L, Zhou X, Ren W. Serine Supports IL-1β Production in Macrophages Through mTOR Signaling. Front Immunol 2020; 11:1866. [PMID: 32973770 PMCID: PMC7481448 DOI: 10.3389/fimmu.2020.01866] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Accepted: 07/13/2020] [Indexed: 12/13/2022] Open
Abstract
Intracellular metabolic programs tightly regulate the functions of macrophages, and previous studies have shown that serine mainly shapes the macrophage function via one-carbon metabolism. However, it is unknown whether serine modulates the macrophage function independent of one-carbon metabolism. Here, we find that serine deprivation lowers interleukin (IL)-1β production and inflammasome activation, as well as reprograms the transcriptomic and metabolic profile in M1 macrophages. Intriguingly, supplementation of formate, glycine, dNTPs, and glucose cannot rescue the production of IL-1β from serine-deprived macrophages. Mechanistically, serine deprivation inhibits macrophage IL-1β production through inhibition of mechanistic target of rapamycin (mTOR) signaling. Of note, the macrophages from mice feeding serine-free diet have lower IL-1β production, and these mice also show less inflammation after LPS challenge. Collectively, our data highlight a new regulatory mechanism for serine to modulate the macrophage function.
Collapse
Affiliation(s)
- Siyuan Chen
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, Guangdong Laboratory of Lingnan Modern Agriculture, Guangdong Provincial Key Laboratory of Animal Nutrition Control, National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Yaoyao Xia
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, Guangdong Laboratory of Lingnan Modern Agriculture, Guangdong Provincial Key Laboratory of Animal Nutrition Control, National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Fang He
- College of Animal Science and Technology, Southwest University, Chongqing, China
| | - Jian Fu
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, Guangdong Laboratory of Lingnan Modern Agriculture, Guangdong Provincial Key Laboratory of Animal Nutrition Control, National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Zhongquan Xin
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, Guangdong Laboratory of Lingnan Modern Agriculture, Guangdong Provincial Key Laboratory of Animal Nutrition Control, National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Baichuan Deng
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, Guangdong Laboratory of Lingnan Modern Agriculture, Guangdong Provincial Key Laboratory of Animal Nutrition Control, National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Liuqin He
- Hunan International Joint Laboratory of Animal Intestinal Ecology and Health, Laboratory of Animal Nutrition and Human Health, College of Life Sciences, Hunan Normal University, Changsha, China
| | - Xihong Zhou
- Key Laboratory of Agro-ecological Processes in Subtropical Region, Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic Process, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, China
| | - Wenkai Ren
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, Guangdong Laboratory of Lingnan Modern Agriculture, Guangdong Provincial Key Laboratory of Animal Nutrition Control, National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, China
| |
Collapse
|
18
|
Terasaki M, Yashima H, Mori Y, Saito T, Matsui T, Hiromura M, Kushima H, Osaka N, Ohara M, Fukui T, Hirano T, Yamagishi SI. A Dipeptidyl Peptidase-4 Inhibitor Inhibits Foam Cell Formation of Macrophages in Type 1 Diabetes via Suppression of CD36 and ACAT-1 Expression. Int J Mol Sci 2020; 21:ijms21134811. [PMID: 32646003 PMCID: PMC7369823 DOI: 10.3390/ijms21134811] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Revised: 06/27/2020] [Accepted: 07/06/2020] [Indexed: 01/15/2023] Open
Abstract
Dipeptidyl peptidase-4 (DPP-4) inhibitors have been reported to play a protective role against atherosclerosis in both animal models and patients with type 2 diabetes (T2D). However, since T2D is associated with dyslipidemia, hypertension and insulin resistance, part of which are ameliorated by DPP-4 inhibitors, it remains unclear whether DPP-4 inhibitors could have anti-atherosclerotic properties directly by attenuating the harmful effects of hyperglycemia. Therefore, we examined whether a DPP-4 inhibitor, teneligliptin, could suppress oxidized low-density lipoprotein (ox-LDL) uptake, foam cell formation, CD36 and acyl-coenzyme A: cholesterol acyltransferase-1 (ACAT-1) gene expression of macrophages isolated from streptozotocin-induced type 1 diabetes (T1D) mice and T1D patients as well as advanced glycation end product (AGE)-exposed mouse peritoneal macrophages and THP-1 cells. Foam cell formation, CD36 and ACAT-1 gene expression of macrophages derived from T1D mice or patients increased compared with those from non-diabetic controls, all of which were inhibited by 10 nmol/L teneligliptin. AGEs mimicked the effects of T1D; teneligliptin attenuated all the deleterious effects of AGEs in mouse macrophages and THP-1 cells. Our present findings suggest that teneligliptin may inhibit foam cell formation of macrophages in T1D via suppression of CD36 and ACAT-1 gene expression partly by attenuating the harmful effects of AGEs.
Collapse
Affiliation(s)
- Michishige Terasaki
- Department of Medicine, Division of Diabetes, Metabolism, and Endocrinology, Showa University School of Medicine, Tokyo 142-8666, Japan; (H.Y.); (Y.M.); (T.S.); (M.H.); (H.K.); (N.O.); (M.O.); (T.F.); (S.Y.)
- Correspondence: ; Tel.: +81-3-3784-8947; Fax: +81-3-3784-8948
| | - Hironori Yashima
- Department of Medicine, Division of Diabetes, Metabolism, and Endocrinology, Showa University School of Medicine, Tokyo 142-8666, Japan; (H.Y.); (Y.M.); (T.S.); (M.H.); (H.K.); (N.O.); (M.O.); (T.F.); (S.Y.)
| | - Yusaku Mori
- Department of Medicine, Division of Diabetes, Metabolism, and Endocrinology, Showa University School of Medicine, Tokyo 142-8666, Japan; (H.Y.); (Y.M.); (T.S.); (M.H.); (H.K.); (N.O.); (M.O.); (T.F.); (S.Y.)
| | - Tomomi Saito
- Department of Medicine, Division of Diabetes, Metabolism, and Endocrinology, Showa University School of Medicine, Tokyo 142-8666, Japan; (H.Y.); (Y.M.); (T.S.); (M.H.); (H.K.); (N.O.); (M.O.); (T.F.); (S.Y.)
| | - Takanori Matsui
- Department of Pathophysiology and Therapeutics of Diabetic Vascular Complications, Kurume University School of Medicine, Kurume 830-0011, Japan;
| | - Munenori Hiromura
- Department of Medicine, Division of Diabetes, Metabolism, and Endocrinology, Showa University School of Medicine, Tokyo 142-8666, Japan; (H.Y.); (Y.M.); (T.S.); (M.H.); (H.K.); (N.O.); (M.O.); (T.F.); (S.Y.)
| | - Hideki Kushima
- Department of Medicine, Division of Diabetes, Metabolism, and Endocrinology, Showa University School of Medicine, Tokyo 142-8666, Japan; (H.Y.); (Y.M.); (T.S.); (M.H.); (H.K.); (N.O.); (M.O.); (T.F.); (S.Y.)
| | - Naoya Osaka
- Department of Medicine, Division of Diabetes, Metabolism, and Endocrinology, Showa University School of Medicine, Tokyo 142-8666, Japan; (H.Y.); (Y.M.); (T.S.); (M.H.); (H.K.); (N.O.); (M.O.); (T.F.); (S.Y.)
| | - Makoto Ohara
- Department of Medicine, Division of Diabetes, Metabolism, and Endocrinology, Showa University School of Medicine, Tokyo 142-8666, Japan; (H.Y.); (Y.M.); (T.S.); (M.H.); (H.K.); (N.O.); (M.O.); (T.F.); (S.Y.)
| | - Tomoyasu Fukui
- Department of Medicine, Division of Diabetes, Metabolism, and Endocrinology, Showa University School of Medicine, Tokyo 142-8666, Japan; (H.Y.); (Y.M.); (T.S.); (M.H.); (H.K.); (N.O.); (M.O.); (T.F.); (S.Y.)
| | - Tsutomu Hirano
- Diabetes Center, Ebina General Hospital, Ebina 243-0433, Japan;
| | - Sho-ichi Yamagishi
- Department of Medicine, Division of Diabetes, Metabolism, and Endocrinology, Showa University School of Medicine, Tokyo 142-8666, Japan; (H.Y.); (Y.M.); (T.S.); (M.H.); (H.K.); (N.O.); (M.O.); (T.F.); (S.Y.)
| |
Collapse
|
19
|
Balzer MS, Helmke A, Ackermann M, Casper J, Dong L, Hiss M, Kiyan Y, Rong S, Timrott K, von Vietinghoff S, Wang L, Haller H, Shushakova N. Protein kinase C beta deficiency increases glucose-mediated peritoneal damage via M1 macrophage polarization and up-regulation of mesothelial protein kinase C alpha. Nephrol Dial Transplant 2020; 34:947-960. [PMID: 30247663 DOI: 10.1093/ndt/gfy282] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2018] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Peritoneal membrane (PM) damage during peritoneal dialysis (PD) is mediated largely by high glucose (HG)-induced pro-inflammatory and neo-angiogenic processes, resulting in PM fibrosis and ultrafiltration failure. We recently demonstrated a crucial role for protein kinase C (PKC) isoform α in mesothelial cells. METHODS In this study we investigate the role of PKCβ in PM damage in vitro using primary mouse peritoneal macrophages (MPMΦ), human macrophages (HMΦ) and immortalized mouse peritoneal mesothelial cells (MPMCs), as well as in vivo using a chronic PD mouse model. RESULTS We demonstrate that PKCβ is the predominant classical PKC isoform expressed in primary MPMΦ and its expression is up-regulated in vitro under HG conditions. After in vitro lipopolysaccharides stimulation PKCβ-/- MPMΦ demonstrates increased levels of interleukin 6 (IL-6), tumour necrosis factor α, and monocyte chemoattractant protein-1 and drastically decrease IL-10 release compared with wild-type (WT) cells. In vivo, catheter-delivered treatment with HG PD fluid for 5 weeks induces PKCβ up-regulation in omentum of WT mice and results in inflammatory response and PM damage characterized by fibrosis and neo-angiogenesis. In comparison to WT mice, all pathological changes are strongly aggravated in PKCβ-/- animals. Underlying molecular mechanisms involve a pro-inflammatory M1 polarization shift of MPMΦ and up-regulation of PKCα in MPMCs of PKCβ-/- mice. Finally, we demonstrate PKCβ involvement in HG-induced polarization processes in HMΦ. CONCLUSIONS PKCβ as the dominant PKC isoform in MPMΦ is up-regulated by HG PD fluid and exerts anti-inflammatory effects during PD through regulation of MPMΦ M1/M2 polarization and control of the dominant mesothelial PKC isoform α.
Collapse
Affiliation(s)
- Michael S Balzer
- Department of Nephrology and Hypertension, Hannover Medical School, Hannover, Germany
| | - Alexandra Helmke
- Department of Nephrology and Hypertension, Hannover Medical School, Hannover, Germany
| | - Martina Ackermann
- Department of Nephrology and Hypertension, Hannover Medical School, Hannover, Germany.,Phenos, Hannover, Germany
| | - Janis Casper
- Department of Nephrology and Hypertension, Hannover Medical School, Hannover, Germany
| | - Lei Dong
- Department of Nephrology and Hypertension, Hannover Medical School, Hannover, Germany
| | - Marcus Hiss
- Department of Nephrology and Hypertension, Hannover Medical School, Hannover, Germany
| | - Yulia Kiyan
- Department of Nephrology and Hypertension, Hannover Medical School, Hannover, Germany
| | - Song Rong
- Department of Nephrology and Hypertension, Hannover Medical School, Hannover, Germany
| | - Kai Timrott
- Department for General, Abdominal and Transplant Surgery, Hannover Medical School, Hannover, Germany
| | | | - Le Wang
- Department of Nephrology and Hypertension, Hannover Medical School, Hannover, Germany.,Department of Nephrology, Tongji Medical College, Wuhan, China
| | - Hermann Haller
- Department of Nephrology and Hypertension, Hannover Medical School, Hannover, Germany
| | - Nelli Shushakova
- Department of Nephrology and Hypertension, Hannover Medical School, Hannover, Germany.,Phenos, Hannover, Germany
| |
Collapse
|
20
|
Sagar D, Gaddipati R, Ongstad EL, Bhagroo N, An LL, Wang J, Belkhodja M, Rahman S, Manna Z, Davis MA, Hasni S, Siegel R, Sanjuan M, Grimsby J, Kolbeck R, Karathanasis S, Sims GP, Gupta R. LOX-1: A potential driver of cardiovascular risk in SLE patients. PLoS One 2020; 15:e0229184. [PMID: 32182251 PMCID: PMC7077835 DOI: 10.1371/journal.pone.0229184] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2019] [Accepted: 02/02/2020] [Indexed: 12/27/2022] Open
Abstract
Traditional cardiovascular disease (CVD) risk factors, such as hypertension, dyslipidemia and diabetes do not explain the increased CVD burden in systemic lupus erythematosus (SLE). The oxidized-LDL receptor, LOX-1, is an inflammation-induced receptor implicated in atherosclerotic plaque formation in acute coronary syndrome, and here we evaluated its role in SLE-associated CVD. SLE patients have increased sLOX-1 levels which were associated with elevated proinflammatory HDL, oxLDL and hsCRP. Interestingly, increased sLOX-1 levels were associated with patients with early disease onset, low disease activity, increased IL-8, and normal complement and hematological measures. LOX-1 was increased on patient-derived monocytes and low-density granulocytes, and activation with oxLDL and immune-complexes increased membrane LOX-1, TACE activity, sLOX-1 release, proinflammatory cytokine production by monocytes, and triggered the formation of neutrophil extracellular traps which can promote vascular injury. In conclusion, perturbations in the lipid content in SLE patients' blood activate LOX-1 and promote inflammatory responses. Increased sLOX-1 levels may be an indicator of high CVD risk, and blockade of LOX-1 may provide a therapeutic opportunity for ameliorating atherosclerosis in SLE patients.
Collapse
Affiliation(s)
- Divya Sagar
- Respiratory, Inflammation and Autoimmune, Biopharmaceuticals R&D, AstraZeneca, Gaithersburg, Maryland, United States of America
| | - Ranjitha Gaddipati
- Cardiovascular, Renal and Metabolism, Biopharmaceuticals R&D, AstraZeneca, Gaithersburg, Maryland, United States of America
| | - Emily L. Ongstad
- Cardiovascular, Renal and Metabolism, Biopharmaceuticals R&D, AstraZeneca, Gaithersburg, Maryland, United States of America
| | - Nicholas Bhagroo
- Cardiovascular, Renal and Metabolism, Biopharmaceuticals R&D, AstraZeneca, Gaithersburg, Maryland, United States of America
| | - Ling-Ling An
- Respiratory, Inflammation and Autoimmune, Biopharmaceuticals R&D, AstraZeneca, Gaithersburg, Maryland, United States of America
| | - Jingya Wang
- Respiratory, Inflammation and Autoimmune, Biopharmaceuticals R&D, AstraZeneca, Gaithersburg, Maryland, United States of America
| | - Mehdi Belkhodja
- Cardiovascular, Renal and Metabolism, Biopharmaceuticals R&D, AstraZeneca, Gaithersburg, Maryland, United States of America
| | - Saifur Rahman
- Respiratory, Inflammation and Autoimmune, Biopharmaceuticals R&D, AstraZeneca, Gaithersburg, Maryland, United States of America
| | - Zerai Manna
- National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institute of Health, Bethesda, Maryland, United States of America
| | - Michael A. Davis
- National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institute of Health, Bethesda, Maryland, United States of America
| | - Sarfaraz Hasni
- National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institute of Health, Bethesda, Maryland, United States of America
| | - Richard Siegel
- National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institute of Health, Bethesda, Maryland, United States of America
| | - Miguel Sanjuan
- Respiratory, Inflammation and Autoimmune, Biopharmaceuticals R&D, AstraZeneca, Gaithersburg, Maryland, United States of America
| | - Joseph Grimsby
- Cardiovascular, Renal and Metabolism, Biopharmaceuticals R&D, AstraZeneca, Gaithersburg, Maryland, United States of America
| | - Roland Kolbeck
- Respiratory, Inflammation and Autoimmune, Biopharmaceuticals R&D, AstraZeneca, Gaithersburg, Maryland, United States of America
| | - Sotirios Karathanasis
- Cardiovascular, Renal and Metabolism, Biopharmaceuticals R&D, AstraZeneca, Gaithersburg, Maryland, United States of America
| | - Gary P. Sims
- Respiratory, Inflammation and Autoimmune, Biopharmaceuticals R&D, AstraZeneca, Gaithersburg, Maryland, United States of America
- * E-mail:
| | - Ruchi Gupta
- Cardiovascular, Renal and Metabolism, Biopharmaceuticals R&D, AstraZeneca, Gaithersburg, Maryland, United States of America
| |
Collapse
|
21
|
Markstad H, Edsfeldt A, Yao Mattison I, Bengtsson E, Singh P, Cavalera M, Asciutto G, Björkbacka H, Fredrikson GN, Dias N, Volkov P, Orho-Melander M, Nilsson J, Engström G, Gonçalves I. High Levels of Soluble Lectinlike Oxidized Low-Density Lipoprotein Receptor-1 Are Associated With Carotid Plaque Inflammation and Increased Risk of Ischemic Stroke. J Am Heart Assoc 2020; 8:e009874. [PMID: 30744454 PMCID: PMC6405674 DOI: 10.1161/jaha.118.009874] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
Background When the lectinlike oxidized low-density lipoprotein (ox LDL) receptor-1 ( LOX -1), a scavenger receptor for ox LDL , binds ox LDL , processes leading to endothelial dysfunction and inflammation are promoted. We aimed to study release mechanisms of LOX -1 and how circulating levels of soluble LOX -1 ( sLOX -1) relate to plaque inflammation and future risk for ischemic stroke. Methods and Results Endothelial cells and leukocytes were used to study release of sLOX -1. Plasma levels of sLOX -1 were determined in 4703 participants in the Malmö Diet and Cancer cohort. Incidence of ischemic stroke was monitored. For 202 patients undergoing carotid endarterectomy, levels of sLOX -1 were analyzed in plasma and plaque homogenates and related to plaque inflammation factors. Endothelial cells released sLOX -1 when exposed to ox LDL . A total of 257 subjects experienced stroke during a mean follow-up of 16.5 years. Subjects in the highest tertile of sLOX -1 had a stroke hazard ratio of 1.75 (95% CI, 1.28-2.39) compared with those in the lowest tertile after adjusting for age and sex. The patients undergoing carotid endarterectomy had a significant association between plasma sLOX -1 and the plaque content of sLOX -1 ( r=0.209, P=0.004). Plaques with high levels of sLOX -1 had more ox LDL , proinflammatory cytokines, and matrix metalloproteinases. Conclusions Our findings demonstrate that ox LDL induces the release of sLOX -1 from endothelial cells and that circulating levels of sLOX -1 correlate with carotid plaque inflammation and risk for ischemic stroke. These observations provide clinical support to experimental studies implicating LOX -1 in atherosclerosis and its possible role as target for cardiovascular intervention.
Collapse
Affiliation(s)
- Hanna Markstad
- 1 Center for Medical Imaging and Physiology Skåne University Hospital Lund University Lund Sweden.,2 Experimental Cardiovascular Research Unit Clinical Research Center Clinical Sciences Malmö Lund University Malmö Sweden
| | - Andreas Edsfeldt
- 2 Experimental Cardiovascular Research Unit Clinical Research Center Clinical Sciences Malmö Lund University Malmö Sweden.,3 Department of Cardiology Skåne University Hospital Malmö Sweden
| | - Ingrid Yao Mattison
- 2 Experimental Cardiovascular Research Unit Clinical Research Center Clinical Sciences Malmö Lund University Malmö Sweden
| | - Eva Bengtsson
- 2 Experimental Cardiovascular Research Unit Clinical Research Center Clinical Sciences Malmö Lund University Malmö Sweden
| | - Pratibha Singh
- 2 Experimental Cardiovascular Research Unit Clinical Research Center Clinical Sciences Malmö Lund University Malmö Sweden
| | - Michele Cavalera
- 2 Experimental Cardiovascular Research Unit Clinical Research Center Clinical Sciences Malmö Lund University Malmö Sweden
| | - Giuseppe Asciutto
- 4 Vascular Center, Malmö, Sweden Skåne University Hospital Malmö Sweden
| | - Harry Björkbacka
- 2 Experimental Cardiovascular Research Unit Clinical Research Center Clinical Sciences Malmö Lund University Malmö Sweden
| | - Gunilla Nordin Fredrikson
- 2 Experimental Cardiovascular Research Unit Clinical Research Center Clinical Sciences Malmö Lund University Malmö Sweden
| | - Nuno Dias
- 4 Vascular Center, Malmö, Sweden Skåne University Hospital Malmö Sweden
| | - Petr Volkov
- 5 Clinical Sciences Malmö Lund University Malmö Sweden
| | | | - Jan Nilsson
- 2 Experimental Cardiovascular Research Unit Clinical Research Center Clinical Sciences Malmö Lund University Malmö Sweden
| | | | - Isabel Gonçalves
- 2 Experimental Cardiovascular Research Unit Clinical Research Center Clinical Sciences Malmö Lund University Malmö Sweden.,3 Department of Cardiology Skåne University Hospital Malmö Sweden
| |
Collapse
|
22
|
Pennig J, Scherrer P, Gissler MC, Anto-Michel N, Hoppe N, Füner L, Härdtner C, Stachon P, Wolf D, Hilgendorf I, Mullick A, Bode C, Zirlik A, Goldberg IJ, Willecke F. Glucose lowering by SGLT2-inhibitor empagliflozin accelerates atherosclerosis regression in hyperglycemic STZ-diabetic mice. Sci Rep 2019; 9:17937. [PMID: 31784656 PMCID: PMC6884628 DOI: 10.1038/s41598-019-54224-9] [Citation(s) in RCA: 57] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2019] [Accepted: 10/26/2019] [Indexed: 12/12/2022] Open
Abstract
Diabetes worsens atherosclerosis progression and leads to a defect in repair of arteries after cholesterol reduction, a process termed regression. Empagliflozin reduces blood glucose levels via inhibition of the sodium glucose cotransporter 2 (SGLT-2) in the kidney and has been shown to lead to a marked reduction in cardiovascular events in humans. To determine whether glucose lowering by empagliflozin accelerates atherosclerosis regression in a mouse model, male C57BL/6J mice were treated intraperitoneally with LDLR- and SRB1- antisense oligonucleotides and fed a high cholesterol diet for 16 weeks to induce severe hypercholesterolemia and atherosclerosis progression. At week 14 all mice were rendered diabetic by streptozotocin (STZ) injections. At week 16 a baseline group was sacrificed and displayed substantial atherosclerosis of the aortic root. In the remaining mice, plasma cholesterol was lowered by switching to chow diet and treatment with LDLR sense oligonucleotides to induce atherosclerosis regression. These mice then received either empagliflozin or vehicle for three weeks. Atherosclerotic plaques in the empagliflozin treated mice were significantly smaller, showed decreased lipid and CD68+ macrophage content, as well as greater collagen content. Proliferation of plaque resident macrophages and leukocyte adhesion to the vascular wall were significantly decreased in empagliflozin-treated mice. In summary, plasma glucose lowering by empagliflozin improves plaque regression in diabetic mice.
Collapse
Affiliation(s)
- Jan Pennig
- University Heart Center Freiburg-Bad Krozingen, Cardiology and Angiology I, University of Freiburg, Freiburg, Germany
| | - Philipp Scherrer
- University Heart Center Freiburg-Bad Krozingen, Cardiology and Angiology I, University of Freiburg, Freiburg, Germany
| | - Mark Colin Gissler
- University Heart Center Freiburg-Bad Krozingen, Cardiology and Angiology I, University of Freiburg, Freiburg, Germany
| | - Nathaly Anto-Michel
- University Heart Center Freiburg-Bad Krozingen, Cardiology and Angiology I, University of Freiburg, Freiburg, Germany
| | - Natalie Hoppe
- University Heart Center Freiburg-Bad Krozingen, Cardiology and Angiology I, University of Freiburg, Freiburg, Germany
| | - Lisa Füner
- University Heart Center Freiburg-Bad Krozingen, Cardiology and Angiology I, University of Freiburg, Freiburg, Germany
| | - Carmen Härdtner
- University Heart Center Freiburg-Bad Krozingen, Cardiology and Angiology I, University of Freiburg, Freiburg, Germany
| | - Peter Stachon
- University Heart Center Freiburg-Bad Krozingen, Cardiology and Angiology I, University of Freiburg, Freiburg, Germany
| | - Dennis Wolf
- University Heart Center Freiburg-Bad Krozingen, Cardiology and Angiology I, University of Freiburg, Freiburg, Germany
| | - Ingo Hilgendorf
- University Heart Center Freiburg-Bad Krozingen, Cardiology and Angiology I, University of Freiburg, Freiburg, Germany
| | - Adam Mullick
- Ionis Pharmaceuticals, Carlsbad, California, USA
| | - Christoph Bode
- University Heart Center Freiburg-Bad Krozingen, Cardiology and Angiology I, University of Freiburg, Freiburg, Germany
| | - Andreas Zirlik
- University Heart Center Freiburg-Bad Krozingen, Cardiology and Angiology I, University of Freiburg, Freiburg, Germany.,Division of Cardiology, Medical University of Graz, Graz, Austria
| | - Ira J Goldberg
- Department of Medicine, New York University Langone Health, New York, NY, USA
| | - Florian Willecke
- University Heart Center Freiburg-Bad Krozingen, Cardiology and Angiology I, University of Freiburg, Freiburg, Germany. .,Klinik für Allgemeine und Interventionelle Kardiologie/Angiologie, Herz- und Diabeteszentrum Nordrhein-Westfalen, Universitätsklinik der Ruhr-Universität Bochum, Bochum, Germany.
| |
Collapse
|
23
|
Shiotsugu S, Okinaga T, Habu M, Yoshiga D, Yoshioka I, Nishihara T, Ariyoshi W. The Biological Effects of Interleukin-17A on Adhesion Molecules Expression and Foam Cell Formation in Atherosclerotic Lesions. J Interferon Cytokine Res 2019; 39:694-702. [DOI: 10.1089/jir.2019.0034] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Affiliation(s)
- Shohei Shiotsugu
- Division of Infections and Molecular Biology, Department of Health Promotion, Kyushu Dental University, Kitakyushu, Fukuoka, Japan
- Division of Oral Medicine, Department of Science and Physical Functions, Kyushu Dental University, Kitakyushu, Fukuoka, Japan
| | | | - Manabu Habu
- Division of Oral and Maxillofacial Surgery, Department of Science and Physical Functions, Kyushu Dental University, Kitakyushu, Fukuoka, Japan
| | - Daigo Yoshiga
- Division of Oral Medicine, Department of Science and Physical Functions, Kyushu Dental University, Kitakyushu, Fukuoka, Japan
| | - Izumi Yoshioka
- Division of Oral Medicine, Department of Science and Physical Functions, Kyushu Dental University, Kitakyushu, Fukuoka, Japan
| | - Tatsuji Nishihara
- Division of Infections and Molecular Biology, Department of Health Promotion, Kyushu Dental University, Kitakyushu, Fukuoka, Japan
| | - Wataru Ariyoshi
- Division of Infections and Molecular Biology, Department of Health Promotion, Kyushu Dental University, Kitakyushu, Fukuoka, Japan
| |
Collapse
|
24
|
Herrero-Fernandez B, Gomez-Bris R, Somovilla-Crespo B, Gonzalez-Granado JM. Immunobiology of Atherosclerosis: A Complex Net of Interactions. Int J Mol Sci 2019; 20:E5293. [PMID: 31653058 PMCID: PMC6862594 DOI: 10.3390/ijms20215293] [Citation(s) in RCA: 85] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2019] [Revised: 10/21/2019] [Accepted: 10/22/2019] [Indexed: 02/07/2023] Open
Abstract
Cardiovascular disease is the leading cause of mortality worldwide, and atherosclerosis the principal factor underlying cardiovascular events. Atherosclerosis is a chronic inflammatory disease characterized by endothelial dysfunction, intimal lipid deposition, smooth muscle cell proliferation, cell apoptosis and necrosis, and local and systemic inflammation, involving key contributions to from innate and adaptive immunity. The balance between proatherogenic inflammatory and atheroprotective anti-inflammatory responses is modulated by a complex network of interactions among vascular components and immune cells, including monocytes, macrophages, dendritic cells, and T, B, and foam cells; these interactions modulate the further progression and stability of the atherosclerotic lesion. In this review, we take a global perspective on existing knowledge about the pathogenesis of immune responses in the atherosclerotic microenvironment and the interplay between the major innate and adaptive immune factors in atherosclerosis. Studies such as this are the basis for the development of new therapies against atherosclerosis.
Collapse
Affiliation(s)
- Beatriz Herrero-Fernandez
- LamImSys Lab. Instituto de Investigación Hospital 12 de Octubre (imas12), 28041 Madrid, Spain.
- Departamento de Fisiología. Facultad de Medicina. Universidad Autónoma de Madrid (UAM), 28029 Madrid, Spain.
| | - Raquel Gomez-Bris
- LamImSys Lab. Instituto de Investigación Hospital 12 de Octubre (imas12), 28041 Madrid, Spain.
| | | | - Jose Maria Gonzalez-Granado
- LamImSys Lab. Instituto de Investigación Hospital 12 de Octubre (imas12), 28041 Madrid, Spain.
- Departamento de Fisiología. Facultad de Medicina. Universidad Autónoma de Madrid (UAM), 28029 Madrid, Spain.
- Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), 28029 Madrid, Spain.
- CIBER de Enfermedades Cardiovasculares, 28029 Madrid, Spain.
| |
Collapse
|
25
|
Wang D, Yang Y, Lei Y, Tzvetkov NT, Liu X, Yeung AWK, Xu S, Atanasov AG. Targeting Foam Cell Formation in Atherosclerosis: Therapeutic Potential of Natural Products. Pharmacol Rev 2019; 71:596-670. [PMID: 31554644 DOI: 10.1124/pr.118.017178] [Citation(s) in RCA: 146] [Impact Index Per Article: 24.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Foam cell formation and further accumulation in the subendothelial space of the vascular wall is a hallmark of atherosclerotic lesions. Targeting foam cell formation in the atherosclerotic lesions can be a promising approach to treat and prevent atherosclerosis. The formation of foam cells is determined by the balanced effects of three major interrelated biologic processes, including lipid uptake, cholesterol esterification, and cholesterol efflux. Natural products are a promising source for new lead structures. Multiple natural products and pharmaceutical agents can inhibit foam cell formation and thus exhibit antiatherosclerotic capacity by suppressing lipid uptake, cholesterol esterification, and/or promoting cholesterol ester hydrolysis and cholesterol efflux. This review summarizes recent findings on these three biologic processes and natural products with demonstrated potential to target such processes. Discussed also are potential future directions for studying the mechanisms of foam cell formation and the development of foam cell-targeted therapeutic strategies.
Collapse
Affiliation(s)
- Dongdong Wang
- The Second Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang, China (D.W., X.L.); Department of Molecular Biology, Institute of Genetics and Animal Breeding of the Polish Academy of Sciences, Jastrzębiec, Poland (D.W., Y.Y., Y.L., A.G.A.); Department of Pharmacognosy, University of Vienna, Vienna, Austria (A.G.A.); Institute of Clinical Chemistry, University Hospital Zurich, Schlieren, Switzerland (D.W.); Institute of Molecular Biology "Roumen Tsanev," Department of Biochemical Pharmacology and Drug Design, Bulgarian Academy of Sciences, Sofia, Bulgaria (N.T.T.); Pharmaceutical Institute, University of Bonn, Bonn, Germany (N.T.T.); Aab Cardiovascular Research Institute, Department of Medicine, University of Rochester, Rochester, New York (S.X.); Oral and Maxillofacial Radiology, Applied Oral Sciences and Community Dental Care, Faculty of Dentistry, The University of Hong Kong, Hong Kong, China (A.W.K.Y.); and Institute of Neurobiology, Bulgarian Academy of Sciences, Sofia, Bulgaria (A.G.A.)
| | - Yang Yang
- The Second Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang, China (D.W., X.L.); Department of Molecular Biology, Institute of Genetics and Animal Breeding of the Polish Academy of Sciences, Jastrzębiec, Poland (D.W., Y.Y., Y.L., A.G.A.); Department of Pharmacognosy, University of Vienna, Vienna, Austria (A.G.A.); Institute of Clinical Chemistry, University Hospital Zurich, Schlieren, Switzerland (D.W.); Institute of Molecular Biology "Roumen Tsanev," Department of Biochemical Pharmacology and Drug Design, Bulgarian Academy of Sciences, Sofia, Bulgaria (N.T.T.); Pharmaceutical Institute, University of Bonn, Bonn, Germany (N.T.T.); Aab Cardiovascular Research Institute, Department of Medicine, University of Rochester, Rochester, New York (S.X.); Oral and Maxillofacial Radiology, Applied Oral Sciences and Community Dental Care, Faculty of Dentistry, The University of Hong Kong, Hong Kong, China (A.W.K.Y.); and Institute of Neurobiology, Bulgarian Academy of Sciences, Sofia, Bulgaria (A.G.A.)
| | - Yingnan Lei
- The Second Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang, China (D.W., X.L.); Department of Molecular Biology, Institute of Genetics and Animal Breeding of the Polish Academy of Sciences, Jastrzębiec, Poland (D.W., Y.Y., Y.L., A.G.A.); Department of Pharmacognosy, University of Vienna, Vienna, Austria (A.G.A.); Institute of Clinical Chemistry, University Hospital Zurich, Schlieren, Switzerland (D.W.); Institute of Molecular Biology "Roumen Tsanev," Department of Biochemical Pharmacology and Drug Design, Bulgarian Academy of Sciences, Sofia, Bulgaria (N.T.T.); Pharmaceutical Institute, University of Bonn, Bonn, Germany (N.T.T.); Aab Cardiovascular Research Institute, Department of Medicine, University of Rochester, Rochester, New York (S.X.); Oral and Maxillofacial Radiology, Applied Oral Sciences and Community Dental Care, Faculty of Dentistry, The University of Hong Kong, Hong Kong, China (A.W.K.Y.); and Institute of Neurobiology, Bulgarian Academy of Sciences, Sofia, Bulgaria (A.G.A.)
| | - Nikolay T Tzvetkov
- The Second Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang, China (D.W., X.L.); Department of Molecular Biology, Institute of Genetics and Animal Breeding of the Polish Academy of Sciences, Jastrzębiec, Poland (D.W., Y.Y., Y.L., A.G.A.); Department of Pharmacognosy, University of Vienna, Vienna, Austria (A.G.A.); Institute of Clinical Chemistry, University Hospital Zurich, Schlieren, Switzerland (D.W.); Institute of Molecular Biology "Roumen Tsanev," Department of Biochemical Pharmacology and Drug Design, Bulgarian Academy of Sciences, Sofia, Bulgaria (N.T.T.); Pharmaceutical Institute, University of Bonn, Bonn, Germany (N.T.T.); Aab Cardiovascular Research Institute, Department of Medicine, University of Rochester, Rochester, New York (S.X.); Oral and Maxillofacial Radiology, Applied Oral Sciences and Community Dental Care, Faculty of Dentistry, The University of Hong Kong, Hong Kong, China (A.W.K.Y.); and Institute of Neurobiology, Bulgarian Academy of Sciences, Sofia, Bulgaria (A.G.A.)
| | - Xingde Liu
- The Second Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang, China (D.W., X.L.); Department of Molecular Biology, Institute of Genetics and Animal Breeding of the Polish Academy of Sciences, Jastrzębiec, Poland (D.W., Y.Y., Y.L., A.G.A.); Department of Pharmacognosy, University of Vienna, Vienna, Austria (A.G.A.); Institute of Clinical Chemistry, University Hospital Zurich, Schlieren, Switzerland (D.W.); Institute of Molecular Biology "Roumen Tsanev," Department of Biochemical Pharmacology and Drug Design, Bulgarian Academy of Sciences, Sofia, Bulgaria (N.T.T.); Pharmaceutical Institute, University of Bonn, Bonn, Germany (N.T.T.); Aab Cardiovascular Research Institute, Department of Medicine, University of Rochester, Rochester, New York (S.X.); Oral and Maxillofacial Radiology, Applied Oral Sciences and Community Dental Care, Faculty of Dentistry, The University of Hong Kong, Hong Kong, China (A.W.K.Y.); and Institute of Neurobiology, Bulgarian Academy of Sciences, Sofia, Bulgaria (A.G.A.)
| | - Andy Wai Kan Yeung
- The Second Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang, China (D.W., X.L.); Department of Molecular Biology, Institute of Genetics and Animal Breeding of the Polish Academy of Sciences, Jastrzębiec, Poland (D.W., Y.Y., Y.L., A.G.A.); Department of Pharmacognosy, University of Vienna, Vienna, Austria (A.G.A.); Institute of Clinical Chemistry, University Hospital Zurich, Schlieren, Switzerland (D.W.); Institute of Molecular Biology "Roumen Tsanev," Department of Biochemical Pharmacology and Drug Design, Bulgarian Academy of Sciences, Sofia, Bulgaria (N.T.T.); Pharmaceutical Institute, University of Bonn, Bonn, Germany (N.T.T.); Aab Cardiovascular Research Institute, Department of Medicine, University of Rochester, Rochester, New York (S.X.); Oral and Maxillofacial Radiology, Applied Oral Sciences and Community Dental Care, Faculty of Dentistry, The University of Hong Kong, Hong Kong, China (A.W.K.Y.); and Institute of Neurobiology, Bulgarian Academy of Sciences, Sofia, Bulgaria (A.G.A.)
| | - Suowen Xu
- The Second Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang, China (D.W., X.L.); Department of Molecular Biology, Institute of Genetics and Animal Breeding of the Polish Academy of Sciences, Jastrzębiec, Poland (D.W., Y.Y., Y.L., A.G.A.); Department of Pharmacognosy, University of Vienna, Vienna, Austria (A.G.A.); Institute of Clinical Chemistry, University Hospital Zurich, Schlieren, Switzerland (D.W.); Institute of Molecular Biology "Roumen Tsanev," Department of Biochemical Pharmacology and Drug Design, Bulgarian Academy of Sciences, Sofia, Bulgaria (N.T.T.); Pharmaceutical Institute, University of Bonn, Bonn, Germany (N.T.T.); Aab Cardiovascular Research Institute, Department of Medicine, University of Rochester, Rochester, New York (S.X.); Oral and Maxillofacial Radiology, Applied Oral Sciences and Community Dental Care, Faculty of Dentistry, The University of Hong Kong, Hong Kong, China (A.W.K.Y.); and Institute of Neurobiology, Bulgarian Academy of Sciences, Sofia, Bulgaria (A.G.A.)
| | - Atanas G Atanasov
- The Second Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang, China (D.W., X.L.); Department of Molecular Biology, Institute of Genetics and Animal Breeding of the Polish Academy of Sciences, Jastrzębiec, Poland (D.W., Y.Y., Y.L., A.G.A.); Department of Pharmacognosy, University of Vienna, Vienna, Austria (A.G.A.); Institute of Clinical Chemistry, University Hospital Zurich, Schlieren, Switzerland (D.W.); Institute of Molecular Biology "Roumen Tsanev," Department of Biochemical Pharmacology and Drug Design, Bulgarian Academy of Sciences, Sofia, Bulgaria (N.T.T.); Pharmaceutical Institute, University of Bonn, Bonn, Germany (N.T.T.); Aab Cardiovascular Research Institute, Department of Medicine, University of Rochester, Rochester, New York (S.X.); Oral and Maxillofacial Radiology, Applied Oral Sciences and Community Dental Care, Faculty of Dentistry, The University of Hong Kong, Hong Kong, China (A.W.K.Y.); and Institute of Neurobiology, Bulgarian Academy of Sciences, Sofia, Bulgaria (A.G.A.)
| |
Collapse
|
26
|
Stankova T, Delcheva G, Maneva A, Vladeva S. Serum Levels of Carbamylated LDL and Soluble Lectin-Like Oxidized Low-Density Lipoprotein Receptor-1 Are Associated with Coronary Artery Disease in Patients with Metabolic Syndrome. MEDICINA (KAUNAS, LITHUANIA) 2019; 55:E493. [PMID: 31443320 PMCID: PMC6722918 DOI: 10.3390/medicina55080493] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/01/2019] [Revised: 08/11/2019] [Accepted: 08/13/2019] [Indexed: 01/06/2023]
Abstract
Background and objectives: Lectin-like oxidized low density lipoprotein receptor-1 (LOX-1) has been recognized as the primary receptor for carbamylated low-density lipoproteins (cLDL) and is increasingly being viewed as a critical mediator of vascular inflammation and atherosclerosis. The aim of the current study was to evaluate the possible role of circulating cLDL and soluble LOX-1 (sLOX-1) as potential biomarkers of metabolic syndrome (MetS) as well as of coronary artery disease (CAD) among MetS patients. Materials and Methods: The serum levels of cLDL and sLOX-1 were measured by ELISA in 30 MetS patients without CAD, 30 MetS patients with CAD, and 30 healthy controls. Results: Patients with MetS had significantly higher serum levels of both cLDL and sLOX-1 than the healthy controls but lower in comparison to MetS + CAD subjects. Serum sLOX-1 concentration correlated significantly with fasting glucose (rs = 0.414, p = 0.001) and high-density lipoprotein (HDL)-cholesterol (rs = -0.273, p = 0.035) in the whole MetS cohort, whereas it correlated with cLDL only in the MetS + CAD subgroup (rs = 0.396, p = 0.030). The receiver-operating characteristic (ROC) curves of cLDL and sLOX-1 for MetS diagnosis had area under the curve (AUC) values of 0.761 and 0.692, respectively. AUC values of cLDL and sLOX-1 for CAD diagnosis among MetS patients were 0.811 and 0.739. Elevated serum levels of cLDL and sLOX-1 were associated with a higher risk of MetS development [odds ratio (OR) 24.28, 95% confidence interval (CI): 5.86-104.61, p < 0.001 and OR 4.75; 95% CI: 1.58-14.25, p = 0.009] as well as with presence of CAD among MetS subjects (OR 11.23; 95% CI: 3.10-40.71, p < 0.001 and OR 4.03; 95% CI: 1.73-11.84, p = 0.019, respectively). Conclusions: The present study underscores the potential of cLDL and sLOX-1 as promising biomarkers for diagnosis and risk assessment of MetS and CAD among the MetS population.
Collapse
Affiliation(s)
- Teodora Stankova
- Department of Biochemistry, Faculty of Pharmacy, Medical University-Plovdiv, 4002 Plovdiv, Bulgaria.
| | - Ginka Delcheva
- Department of Biochemistry, Faculty of Pharmacy, Medical University-Plovdiv, 4002 Plovdiv, Bulgaria
| | - Ana Maneva
- Department of Biochemistry, Faculty of Pharmacy, Medical University-Plovdiv, 4002 Plovdiv, Bulgaria
| | - Stefka Vladeva
- Clinic of Endocrinology and Metabolic Disorders, University Hospital "Kaspela", 4001 Plovdiv, Bulgaria
| |
Collapse
|
27
|
Upregulated LOX-1 Receptor: Key Player of the Pathogenesis of Atherosclerosis. Curr Atheroscler Rep 2019; 21:38. [DOI: 10.1007/s11883-019-0801-y] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
28
|
Zeya B, Chandra NC. LOX-1: Its cytotopographical variance and disease stress. J Biochem Mol Toxicol 2019; 33:e22375. [PMID: 31332899 DOI: 10.1002/jbt.22375] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2019] [Revised: 06/27/2019] [Accepted: 07/02/2019] [Indexed: 11/06/2022]
Abstract
Lectin-like oxidized low-density lipoprotein receptor-1 (LOX-1) is a canonical receptor for oxidized LDL (oxLDL) among the known modified LDL particles. Topographical variance on LOX-1 expression in different cell types and its influence on the atherogenic potential of the particular cell type is the main focus of this review. Characteristic features of LOX-1 on the atherogenic potential of aortic endothelial cells, macrophages, platelets, and vascular smooth muscle cells have been discussed. Nonspecificity of ligands, besides oxLDL, is also the highlight of this review to show the chameleon characteristics in the functional activity of the receptor protein. Induction of LOX-1 has been reported in diseases like atherosclerosis, diabetes, and hypertension, as well as in the inflammatory response of immune reactions. The expression of LOX-1 is upregulated by the vicious cycle of stimulatory response from proatherogenic molecules.
Collapse
Affiliation(s)
- Bushra Zeya
- Department of Biochemistry, All India Institute of Medical Sciences, Patna, India
| | - Nimai C Chandra
- Department of Biochemistry, All India Institute of Medical Sciences, Patna, India
| |
Collapse
|
29
|
Chen W, Li X, Wang J, Song N, Zhu A, Jia L. miR-378a Modulates Macrophage Phagocytosis and Differentiation through Targeting CD47-SIRPα Axis in Atherosclerosis. Scand J Immunol 2019; 90:e12766. [PMID: 30929259 DOI: 10.1111/sji.12766] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2018] [Revised: 03/23/2019] [Accepted: 03/25/2019] [Indexed: 01/13/2023]
Abstract
BACKGROUND Signal regulatory protein alpha (SIRPa) is an essential signalling molecule that modulates inflammatory responses in macrophages. However, the regulation of SIRPs and its dynamic changes in macrophages under inflammatory stimulation in atherosclerosis remain uncertain. OBJECTIVE The study aimed to identify the miRNAs that regulate SIRPa transcription and their roles in modulating phagocytosis, differentiation and cholesterol efflux in macrophages. METHODS ApoE knockout mice were fed with a high-fat diet for 12 weeks. Intimal lesion areas and lipid accumulation were assessed by haematoxylin and eosin (HE) and oil red O staining. The expression of mRNAs/miRNAs was assessed by RNA-seq (RNA sequencing) and RT-qPCR (real-time quantitative polymerase chain reaction). The identification of miR-378a associated with SIRPa regulation in macrophages induced by ox-LDL was confirmed by RT-qPCR and Western blot. The phagocytosis and differentiation of macrophages were detected to figure out the role of miR-378a and SIRPa. RESULTS SIRPa was proved to be a target of miR-378a. Reduced miR-378a can promote the expression of SIRPa. RNA-seq data showed that the levels of mRNA associated with macrophage phenotypes and SIRPa-CD47 axis were increasing significantly with a decreasing phagocytic phenotype in ApoE-/- mice vs wild-type (WT) mice (P < 0.01). The level of miR-378a was reduced in the aorta of ApoE-/- mice vs WT mice. The experiment in vitro showed that overexpression of miR-378a in macrophages decreased the level of Sirpa mRNA obviously vs control (P < 0.01). The phagocytic activity of miR-378a-transfected macrophages was promoted vs control (P < 0.05). miR-378a significantly depleted Sirpa levels in oxidized low-density lipoprotein (ox-LDL)-stimulated macrophages (P < 0.05), and depletion of miR-378a reversed Sirpa reduction obviously (P < 0.05). miR-378a promoted the secretion of TNF-a and IL-6 indirectly. CONCLUSION It has been demonstrated that miR-378a regulates SIRPa-mediated phagocytosis and polarization of macrophages by a direct or indirect way. This research may provide a new path to promote reverse cholesterol transport of macrophages and hinder the progress of atherosclerosis.
Collapse
Affiliation(s)
- Wenna Chen
- Key Laboratory of Ministry of Education for TCM Viscera-State Theory and Applications, Liaoning University of Traditional Chinese Medicine, Shenyang, China.,Department of Medical Science of Laboratory, Liaoning University of Traditional Chinese Medicine, Shenyang, China
| | - Ximing Li
- Key Laboratory of Ministry of Education for TCM Viscera-State Theory and Applications, Liaoning University of Traditional Chinese Medicine, Shenyang, China
| | - Junyan Wang
- The First Medical College, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Nan Song
- Key Laboratory of Ministry of Education for TCM Viscera-State Theory and Applications, Liaoning University of Traditional Chinese Medicine, Shenyang, China
| | - Aisong Zhu
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Lianqun Jia
- Key Laboratory of Ministry of Education for TCM Viscera-State Theory and Applications, Liaoning University of Traditional Chinese Medicine, Shenyang, China
| |
Collapse
|
30
|
Azhdari M, Karandish M, Mansoori A. Metabolic benefits of curcumin supplementation in patients with metabolic syndrome: A systematic review and meta-analysis of randomized controlled trials. Phytother Res 2019; 33:1289-1301. [PMID: 30941814 DOI: 10.1002/ptr.6323] [Citation(s) in RCA: 58] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2018] [Revised: 01/19/2019] [Accepted: 01/31/2019] [Indexed: 12/16/2022]
Abstract
The finding of studies on the effect of curcumin extract on metabolic factor in patients with metabolic syndrome has had arguable results. This systematic review with meta-analysis of randomized controlled trials (RCT) aimed to analyze the effect of curcumin/turmeric on metabolic factors in patients with metabolic syndrome. The PICO strategy was used to establish the guiding question of this review. Several databases for RCT were searched until September 2018. Of the 144 articles initially identified, seven trials met the eligibility criteria. A random-effects model with a mean weight difference (WMD) and a 95% confidence interval was performed for quantitative data synthesis. Pooled estimates of WMD were calculated between intervention and control groups using random-effects model in the presence of high level of heterogeneity between the studies. The results showed significant improvement of fasting blood glucose (p = 0.01), triglycerides (p < 0.001), high-density lipoprotein cholesterol (p = 0.003), and diastolic blood pressure (p = 0.007) levels. Curcumin was not associated with a significant change in waist circumference measurement (p = 0.6) and systolic blood pressure level (p = 0.269). Curcumin supplementation improves some components of metabolic syndrome.
Collapse
Affiliation(s)
- Maryam Azhdari
- Nutrition and metabolic Diseases Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran.,Department of clinical biochemistry, Faculty of Medicine, Shahid Sadoughi University of Medical Sciences and Health Services, Yazd, Iran
| | - Majid Karandish
- Health Research Institute, Diabetes Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Anahita Mansoori
- Nutrition and metabolic Diseases Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| |
Collapse
|
31
|
Mohammadi A, Blesso CN, Barreto GE, Banach M, Majeed M, Sahebkar A. Macrophage plasticity, polarization and function in response to curcumin, a diet-derived polyphenol, as an immunomodulatory agent. J Nutr Biochem 2019; 66:1-16. [PMID: 30660832 DOI: 10.1016/j.jnutbio.2018.12.005] [Citation(s) in RCA: 144] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2018] [Revised: 11/04/2018] [Accepted: 12/12/2018] [Indexed: 12/19/2022]
Abstract
Monocytes and macrophages are important cells of the innate immune system that have diverse functions, including defense against invading pathogens, removal of dead cells by phagocytosis, antigen presentation in the context of MHC class I and class II molecules, and production of various pro-inflammatory cytokines and chemokines such as IL-1β, IL-6, TNF-α and MCP-1. In addition, pro-inflammatory (M1) and anti-inflammatory (M2) macrophages clearly play important roles in the progression of several inflammatory diseases. Therefore, therapies that target macrophage polarization and function by either blocking their trafficking to sites of inflammation, or skewing M1 to M2 phenotype polarization may hold clinical promise in several inflammatory diseases. Dietary-derived polyphenols have potent natural anti-oxidative properties. Within this group of polyphenols, curcumin has been shown to suppress macrophage inflammatory responses. Curcumin significantly reduces co-stimulatory molecules and also inhibits MAPK activation and the translocation of NF-κB p65. Curcumin can also polarize/repolarize macrophages toward the M2 phenotype. Curcumin-treated macrophages have been shown to be highly efficient at antigen capture and endocytosis via the mannose receptor. These novel findings provide new perspectives for the understanding of the immunopharmacological role of curcumin, as well as its therapeutic potential for impacting macrophage polarization and function in the context of inflammation-related disease. However, the precise effects of curcumin on the migration, differentiation, polarization and immunostimulatory functions of macrophages remain unknown. Therefore, in this review, we summarized whether curcumin can influence macrophage polarization, surface molecule expression, cytokine and chemokine production and their underlying pathways in the prevention of inflammatory diseases.
Collapse
Affiliation(s)
- Asadollah Mohammadi
- Cellular & Molecular Research Center, Kurdistan University of Medical Sciences, Sanandaj, Iran
| | | | - George E Barreto
- Departamento de Nutrición y Bioquímica, Facultad de Ciencias, Pontificia Universidad Javeriana, Bogotá D.C., Colombia; Instituto de Ciencias Biomédicas, Universidad Autónoma de Chile, Santiago, Chile
| | - Maciej Banach
- Department of Hypertension, WAM University Hospital in Lodz, Medical University of Lodz, Zeromskiego 113, Lodz, Poland; Polish Mother's Memorial Hospital Research Institute (PMMHRI), Lodz, Poland
| | | | - Amirhossein Sahebkar
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Neurogenic Inflammation Research Center, Mashhad University of Medical Sciences, Mashhad, Iran; School of Medicine, University of Western Australia, Perth, Australia.
| |
Collapse
|
32
|
Ma S, Bai Z, Wu H, Wang W. The DPP-4 inhibitor saxagliptin ameliorates ox-LDL-induced endothelial dysfunction by regulating AP-1 and NF-κB. Eur J Pharmacol 2019; 851:186-193. [PMID: 30639312 DOI: 10.1016/j.ejphar.2019.01.008] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2018] [Revised: 12/16/2018] [Accepted: 01/08/2019] [Indexed: 12/12/2022]
Abstract
Diabetes-associated cardiovascular complications are the leading cause of death for diabetic patients. Dipeptidyl peptidase 4 (DPP-4) inhibitor agents, known as gliptins, are a class of potent anti-glycemic agents developed to treat diabetes. Recently, gliptins have been shown to have independent cardiovascular benefits. In this study, we revealed the protective role of saxagliptin in vascular endothelial cells. Our data show that saxagliptin suppresses oxidized low-density lipoprotein cholesterol (ox-LDL)-induced expression of its receptor lectin-like ox-LDL receptor-1 (LOX-1). Saxagliptin treatment reduces ox-LDL-induced production of cytokines and vascular adhesion molecules including tumor necrosis factor (TNF-α), interleukin-1β (IL-1β), vascular cell adhesion molecule 1 (VCAM-1), and intercellular cell adhesion molecule-1 (ICAM-1). The presence of saxagliptin suppressed ox-LDL-induced adhesion of monocytes to endothelial cells in co-culture adhesion experiments. Moreover, saxagliptin mitigated ox-LDL-induced production of reactive oxygen species and suppressed elevated expression of endothelial nicotinamide adenine dinucleotide phosphate oxidase subunit (NOX-4) induced by ox-LDL. Mechanistically, saxagliptin exerted inhibitory effects against ox-LDL-induced phosphorylation of JNK kinase, expression of the activator protein 1 (AP-1) subunits c-Jun/c-fos, and AP-1 promoter activity. Saxagliptin also suppressed nuclear factor κB (NF-κB) p65 accumulation and inhibited its promoter activity. Our data elaborate the molecular mechanism of saxagliptin-mediated endothelial protection and indicate that saxagliptin could have vascular benefits independent on its anti-glycemic function.
Collapse
Affiliation(s)
- Suxia Ma
- Cardiology Department 2, Shangqiu First People's Hospital, Shangqiu, Henan 476100, China
| | - Zhifeng Bai
- Cardiology Department 2, Shangqiu First People's Hospital, Shangqiu, Henan 476100, China.
| | - Huiying Wu
- Cardiology Department, Fuwai Central China Cardiovascular Hospital, Zhengzhou, Henan 450007, China
| | - Wei Wang
- Cardiology Department, Fuwai Central China Cardiovascular Hospital, Zhengzhou, Henan 450007, China
| |
Collapse
|
33
|
Tian K, Ogura S, Little PJ, Xu SW, Sawamura T. Targeting LOX-1 in atherosclerosis and vasculopathy: current knowledge and future perspectives. Ann N Y Acad Sci 2018; 1443:34-53. [PMID: 30381837 DOI: 10.1111/nyas.13984] [Citation(s) in RCA: 67] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2018] [Revised: 09/12/2018] [Accepted: 09/24/2018] [Indexed: 12/11/2022]
Abstract
LOX-1 (lectin-like oxidized low-density lipoprotein receptor-1; also known as OLR1) is the dominant receptor that recognizes and internalizes oxidized low-density lipoproteins (ox-LDLs) in endothelial cells. Several genetic variants of LOX-1 are associated with the risk and severity of coronary artery disease. The LOX-1-ox-LDL interaction induces endothelial dysfunction, leukocyte adhesion, macrophage-derived foam cell formation, smooth muscle cell proliferation and migration, and platelet activation. LOX-1 activation eventually leads to the rupture of atherosclerotic plaques and acute cardiovascular events. In addition, LOX-1 can be cleaved to generate soluble LOX-1 (sLOX-1), which is a useful diagnostic and prognostic marker for atherosclerosis-related diseases in human patients. Of therapeutic relevance, several natural products and clinically used drugs have emerged as LOX-1 inhibitors that have antiatherosclerotic actions. We hereby provide an updated overview of role of LOX-1 in atherosclerosis and associated vascular diseases, with an aim to highlighting the potential of LOX-1 as a novel theranostic tool for cardiovascular disease prevention and treatment.
Collapse
Affiliation(s)
- Kunming Tian
- Department of Preventive Medicine, School of Public Health, Zunyi Medical University, Zunyi, Guizhou, China
| | - Sayoko Ogura
- Division of Laboratory Medicine, Department of Pathology and Microbiology, Nihon University School of Medicine, Tokyo, Japan
| | - Peter J Little
- School of Pharmacy, The University of Queensland, Wooloongabba, Queensland, Australia.,Department of Pharmacy, Xinhua College of Sun Yat-sen University, Guangzhou, China
| | - Suo-Wen Xu
- Aab Cardiovascular Research Institute, University of Rochester, Rochester, New York
| | - Tatsuya Sawamura
- Department of Physiology, School of Medicine, Shinshu University, Nagano, Japan.,Research Center for Next Generation Medicine, Shinshu University, Nagano, Japan
| |
Collapse
|
34
|
Suzuki T, Takebayashi K, Hara K, Tsuchiya T, Inukai T. Association between angiopoietin-like protein 2 and lectin-like oxidized low-density lipoprotein receptor 1 ligand containing apolipoprotein B in patients with type 2 diabetes. J Int Med Res 2018; 46:4167-4180. [PMID: 30157689 PMCID: PMC6166345 DOI: 10.1177/0300060518791067] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Objective This study was performed to evaluate the association of the serum level of
angiopoietin-like protein 2 (ANGPTL2) with circulating inflammatory markers
and oxidized and modified low-density lipoprotein (LDL) cholesterol as
evaluated by lectin-like oxidized LDL receptor 1 ligand containing
apolipoprotein B (LAB) in patients with type 2 diabetes. Methods The study included 70 patients with type 2 diabetes hospitalized for glycemic
control and 9 control subjects. Results The serum level of ANGPTL2 was significantly higher in the patients with type
2 diabetes than in the healthy controls. There was a significant positive
correlation between ANGPTL2 and the high-sensitivity C-reactive protein,
fibrinogen, and LAB levels and a significant negative correlation between
ANGPTL2 and the estimated glomerular filtration rate (eGFR). Conclusions These results suggest that the serum ANGPTL2 level has a close positive
association with inflammatory markers, especially fibrinogen and oxidized
and modified LDL as evaluated by LAB. The data also suggest that the serum
ANGPTL2 level is influenced by renal function as reflected by the eGFR.
Collapse
Affiliation(s)
- Tatsuhiko Suzuki
- Department of Internal Medicine, Dokkyo Medical University Saitama Medical Center, Koshigaya, Saitama, Japan
| | - Kohzo Takebayashi
- Department of Internal Medicine, Dokkyo Medical University Saitama Medical Center, Koshigaya, Saitama, Japan
| | - Kenji Hara
- Department of Internal Medicine, Dokkyo Medical University Saitama Medical Center, Koshigaya, Saitama, Japan
| | - Takafumi Tsuchiya
- Department of Internal Medicine, Dokkyo Medical University Saitama Medical Center, Koshigaya, Saitama, Japan
| | - Toshihiko Inukai
- Department of Internal Medicine, Dokkyo Medical University Saitama Medical Center, Koshigaya, Saitama, Japan
| |
Collapse
|
35
|
Kim Y, Clifton P. Curcumin, Cardiometabolic Health and Dementia. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2018; 15:ijerph15102093. [PMID: 30250013 PMCID: PMC6210685 DOI: 10.3390/ijerph15102093] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/19/2018] [Revised: 09/15/2018] [Accepted: 09/20/2018] [Indexed: 02/07/2023]
Abstract
Current research indicates curcumin [diferuloylmethane; a polyphenolic compound isolated from the rhizomes of the dietary spice turmeric (Curcuma longa)] exerts a beneficial effect on health which may be partly attributable to its anti-oxidative and anti-inflammatory properties. The aim of this review is to examine potential mechanisms of the actions of curcumin in both animal and human studies. Curcumin modulates relevant molecular target pathways to improve glucose and lipid metabolism, suppress inflammation, stimulate antioxidant enzymes, facilitate insulin signalling and reduce gut permeability. Curcumin also inhibits Aβ and tau accumulation in animal models and enhances mitochondria and synaptic function. In conclusion, in high-dose animal studies and in vitro, curcumin exerts a potential beneficial effect on cardiometabolic disease. However, human studies are relatively unconvincing. More intervention studies should be conducted with the new curcumin formulation with improved oral bioavailability.
Collapse
Affiliation(s)
- Yoona Kim
- Department of Food and Nutrition/Institute of Agriculture and Life Science, Gyeongsang National University, Jinju 52828, Korea.
| | - Peter Clifton
- School of Pharmacy and Medical Sciences, University of South Australia, General Post Office Box 2471, Adelaide, SA 5001, Australia.
| |
Collapse
|
36
|
Grosick R, Alvarado-Vazquez PA, Messersmith AR, Romero-Sandoval EA. High glucose induces a priming effect in macrophages and exacerbates the production of pro-inflammatory cytokines after a challenge. J Pain Res 2018; 11:1769-1778. [PMID: 30237731 PMCID: PMC6136416 DOI: 10.2147/jpr.s164493] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Introduction Painful diabetic neuropathy is associated with chronic inflammation, in which macrophages are the key effectors. We utilized an in vitro approach to determine the effects of high glucose on macrophage phenotype. Materials and methods We exposed human THP-1 macrophages to normal glucose (5 mM) and a clinically relevant high glucose environment (15 mM) and measured the expression and concentration of molecules associated with a diabetic cellular phenotype. Results We found that THP-1 macrophages in high glucose conditions did not influence the basal expression of cyclooxygenase-2, Toll-like receptor-4, or class A scavenger receptor mRNA, or the concentrations of the cytokines interleukin (IL)-6, monocyte chemoattractant protein (MCP)-1, and IL-10, but induced a priming effect on tumor necrosis factor (TNF)-α. Then, we stimulated THP-1 macrophages with a strong pro-inflammatory stimulus lipopolysaccharide (LPS; 5 µg/mL). After stimulation with LPS, we observed an exacerbated increase in TNF-α, IL-6, and MCP-1 concentration in the high glucose condition compared to the normal glucose environment. THP-1 macrophages in high glucose conditions developed tolerance to IL-10 anti-inflammatory effects (TNF-α production) when challenged with LPS. Conclusion Our in vitro approach allows the study of macrophages as potential targets for therapeutic purposes since it compares them to primary human macrophages exposed to high glucose and macrophages from patients with diabetes or complications of painful diabetic neuropathy (i.e. ulcers, adipocytes, and pancreas).
Collapse
Affiliation(s)
- Rachel Grosick
- Department of Pharmaceutical and Administrative Science, Presbyterian College School of Pharmacy, Clinton, SC, USA
| | | | - Amy R Messersmith
- Department of Pharmaceutical and Administrative Science, Presbyterian College School of Pharmacy, Clinton, SC, USA
| | | |
Collapse
|
37
|
Xu J, Zhu L, Liu H, Li M, Liu Y, Yang F, Pei Z. Thymoquinone reduces cardiac damage caused by hypercholesterolemia in apolipoprotein E-deficient mice. Lipids Health Dis 2018; 17:173. [PMID: 30049280 PMCID: PMC6062953 DOI: 10.1186/s12944-018-0829-y] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2017] [Accepted: 07/18/2018] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Hypercholesterolemia is a well-established risk factor for cardiac damage, which can lead to cardiovascular diseases. Many studies have shown that thymoquinone protected rats from doxorubicin-induced cardiotoxicity and cardiac damage. The aim of this study was to investigate the possible protective effects of thymoquinone against cardiac damage in apolipoprotein E knockout (ApoE-/-) mice. METHODS Eight-week-old male ApoE-/- mice were randomly divided into three groups: control group fed a normal diet (ND group), a high cholesterol diet (HD group) or HD mixed with thymoquinone (HD + TQ group). All groups were fed the different diets for 8 weeks. Blood samples were obtained from the inferior vena cava and collected in serum tubes. The samples were then stored at - 80 °C until used. Coronal sections of heart tissues were fixed in 10% formalin and then embedded in paraffin for histological evaluation. The remainder of the heart tissues was snap-frozen in liquid nitrogen for mRNA or immunohistochemical analysis. RESULTS The metabolic characteristics of total cholesterol (TC), low-density lipoprotein-cholesterol (LDL-c), and high-sensitivity C-reactive protein (hs-CRP) were lower in ApoE-/-HD + TQ mice than in ApoE-/- HD mice. Lectin-like oxidized low-density lipoprotein receptor-1 (LOX-1) gene and protein expression was lower in the heart tissue of ApoE-/-HD + TQ mice than in those of ApoE-/-HD mice. Furthermore, the levels of macrophages and pro-inflammatory cytokines were lower in the cardiac tissues of ApoE-/-HD + TQ mice than in those of ApoE-/-HD mice. CONCLUSIONS These results indicate that thymoquinone may provide a potential therapeutic target for cardiac damage caused by hypercholesterolemia.
Collapse
Affiliation(s)
- Jingyi Xu
- Department of Endocrinology, Affiliated Zhongshan Hospital of Dalian University, No. 6 Jiefang Street, Dalian, China
| | - Liyue Zhu
- Rehabilitation Center, Zhejiang Hospital, No. 12 Lingyin Road, Hangzhou, Zhejiang, China
| | - Hongyang Liu
- Department of Heart Intensive Care Unit, the First Affiliated Hospital of Dalian Medical University, No.193 Lianhe Road, Dalian, China
| | - Mengye Li
- Department of Special Medical Unit, the First Affiliated Hospital of Dalian Medical University, No. 193 Lianhe Road, Dalian, China
| | - Yingshu Liu
- Department of Endocrinology Dalian Municipal Central Hospital, No. 42 Xuegong Road, Dalian, China
| | - Fan Yang
- Department of Cardiology, Affiliated Zhongshan Hospital of Dalian University, No. 6 Jiefang Street, Dalian, 116001, China
| | - Zuowei Pei
- Department of Cardiology, Affiliated Zhongshan Hospital of Dalian University, No. 6 Jiefang Street, Dalian, 116001, China.
| |
Collapse
|
38
|
Mortazavi-Jahromi SS, Alizadeh S, Javanbakht MH, Mirshafiey A. Cardioprotective effect of β-d-mannuronic acid (M2000) as a novel NSAID on gene expression of oxLDL scavenger receptors in the experimental diabetic model. Immunopharmacol Immunotoxicol 2018; 40:284-289. [PMID: 29619884 DOI: 10.1080/08923973.2018.1455209] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
Abstract
CONTEXT The investigations have shown that patients with diabetes have the elevated levels of glucose and oxLDL. These two play an important role in increased expression levels of oxLDL scavenger receptors on the surface of macrophages and endothelial cells that leads to deposition of oxLDL and macrophages in vascular walls. OBJECTIVE The present study intends to show the effects of β-d-mannuronic acid (M2000) on the expression profile of ox-LDL scavenger receptors (including SR-A, LOX-1, CD36, and CD68) in an experimental model of diabetes. MATERIALS AND METHODS Eighteen Sprague-Dawley rats were randomly divided into three 6-member groups of the healthy control, diabetic control, and treated rats by M2000. Diabetes was induced in rats by intraperitoneal (IP) administration of 60 mg/kg streptozotocin. The treated rats were given daily intraperitoneal injections of M2000 with a dose of 25 mg/kg for 28 days and at the end of the 28th day, their aortas were removed. The qRT-PCR technique was then used to evaluate the expression levels of the proposed gene. RESULTS The gene expression levels of the SR-A, LOX-1, CD36, and CD68 significantly declined in the diabetic group that received M2000 compared with untreated diabetic rats. CONCLUSIONS The M2000, as a novel NSAID is able to modify by lowering the gene expression levels of SR-A, LOX-1, CD36, and CD68 in treated rats compared to the untreated diabetic group, which may play an important role in preventing the complications that could lead to a cardioprotective efficacy.
Collapse
Affiliation(s)
- Seyed Shahabeddin Mortazavi-Jahromi
- a Department of Immunology, School of Public Health , Tehran University of Medical Sciences , Tehran , Iran.,b Department of Cellular and Molecular Biology , Kish International Campus, University of Tehran , Kish , Iran
| | - Shahab Alizadeh
- c Department of Cellular and Molecular Nutrition, School of Nutritional Sciences and Dietetics , Tehran University of Medical Sciences , Tehran , Iran
| | - Mohammad Hassan Javanbakht
- c Department of Cellular and Molecular Nutrition, School of Nutritional Sciences and Dietetics , Tehran University of Medical Sciences , Tehran , Iran
| | - Abbas Mirshafiey
- a Department of Immunology, School of Public Health , Tehran University of Medical Sciences , Tehran , Iran
| |
Collapse
|
39
|
Ooi BK, Goh BH, Yap WH. Oxidative Stress in Cardiovascular Diseases: Involvement of Nrf2 Antioxidant Redox Signaling in Macrophage Foam Cells Formation. Int J Mol Sci 2017; 18:ijms18112336. [PMID: 29113088 PMCID: PMC5713305 DOI: 10.3390/ijms18112336] [Citation(s) in RCA: 92] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2017] [Revised: 10/11/2017] [Accepted: 10/23/2017] [Indexed: 02/07/2023] Open
Abstract
Oxidative stress is an important risk factor contributing to the pathogenesis of cardiovascular diseases. Oxidative stress that results from excessive reactive oxygen species (ROS) production accounts for impaired endothelial function, a process which promotes atherosclerotic lesion or fatty streaks formation (foam cells). Nuclear factor erythroid 2-related factor 2 (Nrf2) is a transcription factor involved in cellular redox homeostasis. Upon exposure to oxidative stress, Nrf2 is dissociated from its inhibitor Keap-1 and translocated into the nucleus, where it results in the transcriptional activation of cell defense genes. Nrf2 has been demonstrated to be involved in the protection against foam cells formation by regulating the expression of antioxidant proteins (HO-1, Prxs, and GPx1), ATP-binding cassette (ABC) efflux transporters (ABCA1 and ABCG1) and scavenger receptors (scavenger receptor class B (CD36), scavenger receptor class A (SR-A) and lectin-type oxidized LDL receptor (LOX-1)). However, Nrf2 has also been reported to exhibit pro-atherogenic effects. A better understanding on the mechanism of Nrf2 in oxidative stress-induced cardiac injury, as well as the regulation of cholesterol uptake and efflux, are required before it can serve as a novel therapeutic target for cardiovascular diseases prevention and treatment.
Collapse
Affiliation(s)
- Bee Kee Ooi
- School of Biosciences, Taylor's University, Subang Jaya, Selangor Darul Ehsan 47500, Malaysia.
| | - Bey Hing Goh
- School of Pharmacy, Monash University Malaysia, Bandar Sunway, Selangor Darul Ehsan 47500, Malaysia.
| | - Wei Hsum Yap
- School of Biosciences, Taylor's University, Subang Jaya, Selangor Darul Ehsan 47500, Malaysia.
| |
Collapse
|
40
|
Oxidative Stress in Cardiovascular Diseases: Involvement of Nrf2 Antioxidant Redox Signaling in Macrophage Foam Cells Formation. Int J Mol Sci 2017. [PMID: 29113088 DOI: 10.3390/ijms18112336.] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Oxidative stress is an important risk factor contributing to the pathogenesis of cardiovascular diseases. Oxidative stress that results from excessive reactive oxygen species (ROS) production accounts for impaired endothelial function, a process which promotes atherosclerotic lesion or fatty streaks formation (foam cells). Nuclear factor erythroid 2-related factor 2 (Nrf2) is a transcription factor involved in cellular redox homeostasis. Upon exposure to oxidative stress, Nrf2 is dissociated from its inhibitor Keap-1 and translocated into the nucleus, where it results in the transcriptional activation of cell defense genes. Nrf2 has been demonstrated to be involved in the protection against foam cells formation by regulating the expression of antioxidant proteins (HO-1, Prxs, and GPx1), ATP-binding cassette (ABC) efflux transporters (ABCA1 and ABCG1) and scavenger receptors (scavenger receptor class B (CD36), scavenger receptor class A (SR-A) and lectin-type oxidized LDL receptor (LOX-1)). However, Nrf2 has also been reported to exhibit pro-atherogenic effects. A better understanding on the mechanism of Nrf2 in oxidative stress-induced cardiac injury, as well as the regulation of cholesterol uptake and efflux, are required before it can serve as a novel therapeutic target for cardiovascular diseases prevention and treatment.
Collapse
|
41
|
Hofmann A, Brunssen C, Morawietz H. Contribution of lectin-like oxidized low-density lipoprotein receptor-1 and LOX-1 modulating compounds to vascular diseases. Vascul Pharmacol 2017; 107:S1537-1891(17)30171-4. [PMID: 29056472 DOI: 10.1016/j.vph.2017.10.002] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2017] [Revised: 09/29/2017] [Accepted: 10/11/2017] [Indexed: 12/31/2022]
Abstract
The lectin-like oxidized low-density lipoprotein receptor-1 (LOX-1) is the major receptor for binding and uptake of oxidized low-density lipoprotein (oxLDL) in endothelial cells. LOX-1 is also expressed in macrophages, smooth muscle cells and platelets. Following internalization of oxLDL, LOX-1 initiates a vicious cycle from activation of pro-inflammatory signaling pathways, thus promoting an increased reactive oxygen species formation and secretion of pro-inflammatory cytokines. LOX-1 plays a pivotal role in the development of endothelial dysfunction, foam cell and advanced lesions formation as well as in myocardial ischemia. Furthermore, it is known that LOX-1 plays a pivotal role in mitochondrial DNA damage, vascular cell apoptosis, and autophagy. A large number of studies provide evidence of a LOX-1's role in endothelial dysfunction, hypertension, diabetes, and obesity. In addition, novel insights into LOX-1 ligands and the activated signaling pathways have been gained. Recent studies have shown an interaction of LOX-1 with microRNA's, thus providing novel tools to regulate LOX-1 function. Because LOX-1 is increased in atherosclerotic plaques and contributes to endothelial dysfunction, several compounds were tested in vivo and in vitro to modulate the LOX-1 expression in therapeutic approaches.
Collapse
Affiliation(s)
- Anja Hofmann
- Division of Vascular Endothelium and Microcirculation, Department of Medicine III, University Hospital and Medical Faculty Carl Gustav Carus Dresden, Technische Universität Dresden, Dresden, Germany
| | - Coy Brunssen
- Division of Vascular Endothelium and Microcirculation, Department of Medicine III, University Hospital and Medical Faculty Carl Gustav Carus Dresden, Technische Universität Dresden, Dresden, Germany
| | - Henning Morawietz
- Division of Vascular Endothelium and Microcirculation, Department of Medicine III, University Hospital and Medical Faculty Carl Gustav Carus Dresden, Technische Universität Dresden, Dresden, Germany.
| |
Collapse
|
42
|
Rizwan H, Mohanta J, Si S, Pal A. Gold nanoparticles reduce high glucose-induced oxidative-nitrosative stress regulated inflammation and apoptosis via tuberin-mTOR/NF-κB pathways in macrophages. Int J Nanomedicine 2017; 12:5841-5862. [PMID: 28860752 PMCID: PMC5566318 DOI: 10.2147/ijn.s141839] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Hyperglycemia is a risk factor for cardiovascular mortality and morbidity, and directly responsible for exacerbating macrophage activation and atherosclerosis. We showed that gold nanoparticles (AuNPs) reduce the high glucose (HG)-induced atherosclerosis-related complications in macrophages via oxidative-nitrosative stress-regulated inflammation and apoptosis. The effects of AuNPs on oxidative-nitrosative stress markers such as cellular antioxidants were attenuated by HG exposure, leading to reduction in the accumulation of reactive oxygen/nitrogen species in cellular compartments. Further, these abnormalities of antioxidants level and reactive oxygen/nitrogen species accumulations initiate cellular stress, resulting in the activation of nuclear factor κB (NF-κB) via ERK1/2mitogen-activated protein kinase (MAPK)/Akt/tuberin-mammalian target of rapamycin (mTOR) pathways. The activated NF-κB stimulates inflammatory mediators, which subsequently subdue biomolecules damage, leading to aggravation of the inflammatory infiltration and immune responses. Treatment of AuNPs inhibits the intracellular redox-sensitive signaling pathways, inflammation, and apoptosis in macrophages. Together, our results indicate that AuNPs may modulate HG-induced oxidative-nitrosative stress. These effects may be sealed tight due to the fact that AuNPs treatment reduces the activation of NF-κB by ERK1/2MAPK/Akt/tuberin-mTOR pathways-mediated inflammatory genes expression and cellular stress responses, which may be beneficial for minimizing the atherosclerosis.
Collapse
Affiliation(s)
- Huma Rizwan
- School of Biotechnology, KIIT University, Bhubaneswar, India
| | - Jagdeep Mohanta
- School of Applied Sciences, KIIT University, Bhubaneswar, India
| | - Satyabrata Si
- School of Biotechnology, KIIT University, Bhubaneswar, India.,School of Applied Sciences, KIIT University, Bhubaneswar, India
| | - Arttatrana Pal
- Department of Zoology, School of Life Sciences, Mahatma Gandhi Central University, Bihar, India
| |
Collapse
|
43
|
Chistiakov DA, Melnichenko AA, Myasoedova VA, Grechko AV, Orekhov AN. Mechanisms of foam cell formation in atherosclerosis. J Mol Med (Berl) 2017; 95:1153-1165. [DOI: 10.1007/s00109-017-1575-8] [Citation(s) in RCA: 287] [Impact Index Per Article: 35.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2017] [Revised: 07/04/2017] [Accepted: 07/28/2017] [Indexed: 12/21/2022]
|
44
|
Zhang Y, Wang N, Zhu L, Liu Y, Pei Z, Wang G, Luo L, Liu H. The Dipeptidyl Peptidase-4 Inhibitor Teneligliptin Reduces Aortic Damage from Hypercholesterolaemia in Apolipoprotein E-Deficient Mice. Biomed Hub 2017; 2:1-9. [PMID: 31988912 PMCID: PMC6945959 DOI: 10.1159/000473869] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2017] [Accepted: 03/27/2017] [Indexed: 01/10/2023] Open
Abstract
Objective Hypercholesterolaemia is a well-established risk factor for blood vessel damage, which can lead to cardiovascular diseases. An abundance of clinical data show that dipeptidyl peptidase-4 inhibitors protect against aortic damage in patients with diabetes. The goal of this study was to investigate the possible protective effects of teneligliptin against aortic damage in apolipoprotein E knockout (ApoE-/-) mice. Methods Eight-week-old male ApoE-/- mice were randomly divided into 3 groups: a control group fed a normal diet, a high-cholesterol diet (HD group), and an HD diet mixed with teneligliptin (HD + Tene group), and all the groups were fed with the different treatments for 6 weeks. Results and Conclusion The metabolic characteristics of total cholesterol, low-density lipoprotein-cholesterol, and high-sensitivity C-reactive protein were lower in ApoE-/- HD + Tene mice than in ApoE-/- HD mice. Lectin-like oxidized low-density lipoprotein receptor-1 (LOX-1) gene and protein expression were lower in the aortic tissue of ApoE-/- HD + Tene mice than in ApoE-/- HD mice. IL-6 and TNF-α gene expression were lower in ApoE-/- HD + Tene mice than in ApoE-/- HD mice. These results indicate that teneligliptin may provide a potential therapeutic target for the aortic damage from hypercholesterolaemia.
Collapse
Affiliation(s)
- Ying Zhang
- Department of Cardiology, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Nan Wang
- Department of Cardiology, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Liyue Zhu
- Rehabilitation Center, Zhejiang Hospital, Hangzhou, China
| | - Yingshu Liu
- Department of Endocrinology, Dalian Municipal Central Hospital, Dalian, China
| | - Zuowei Pei
- Department of Cardiology, Affiliated Zhongshan Hospital of Dalian University, Dalian, China
| | - Guan Wang
- International Medical Department, Affiliated Zhongshan Hospital of Dalian University, Dalian, China
| | - Lin Luo
- Department of Central Intensive Care Unit (ICU), The Second Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Hongyang Liu
- Department of Heart Intensive Care Unit, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| |
Collapse
|
45
|
Wang P, He LY, Shen GD, Li RL, Yang JL. Inhibitory effects of Dioscin on atherosclerosis and foam cell formation in hyperlipidemia rats. Inflammopharmacology 2017; 25:633-642. [DOI: 10.1007/s10787-017-0341-4] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2016] [Accepted: 03/22/2017] [Indexed: 01/10/2023]
|
46
|
Terasaki M, Hiromura M, Mori Y, Kohashi K, Kushima H, Ohara M, Watanabe T, Andersson O, Hirano T. Combination Therapy with a Sodium-Glucose Cotransporter 2 Inhibitor and a Dipeptidyl Peptidase-4 Inhibitor Additively Suppresses Macrophage Foam Cell Formation and Atherosclerosis in Diabetic Mice. Int J Endocrinol 2017; 2017:1365209. [PMID: 28408925 PMCID: PMC5376482 DOI: 10.1155/2017/1365209] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/08/2016] [Accepted: 02/12/2017] [Indexed: 12/02/2022] Open
Abstract
Dipeptidyl peptidase-4 inhibitors (DPP-4is), in addition to their antihyperglycemic roles, have antiatherosclerotic effects. We reported that sodium-glucose cotransporter 2 inhibitors (SGLT2is) suppress atherosclerosis in a glucose-dependent manner in diabetic mice. Here, we investigated the effects of combination therapy with SGLT2i and DPP-4i on atherosclerosis in diabetic mice. SGLT2i (ipragliflozin, 1.0 mg/kg/day) and DPP-4i (alogliptin, 8.0 mg/kg/day), either alone or in combination, were administered to db/db mice or streptozotocin-induced diabetic apolipoprotein E-null (Apoe-/- ) mice. Ipragliflozin and alogliptin monotherapies improved glucose intolerance; however, combination therapy did not show further improvement. The foam cell formation of peritoneal macrophages was suppressed by both the ipragliflozin and alogliptin monotherapies and was further enhanced by combination therapy. Although foam cell formation was closely associated with HbA1c levels in all groups, DPP-4i alone or the combination group showed further suppression of foam cell formation compared with the control or SGLT2i group at corresponding HbA1c levels. Both ipragliflozin and alogliptin monotherapies decreased scavenger receptors and increased cholesterol efflux regulatory genes in peritoneal macrophages, and combination therapy showed additive changes. In diabetic Apoe-/- mice, combination therapy showed the greatest suppression of plaque volume in the aortic root. In conclusion, combination therapy with SGLT2i and DPP4i synergistically suppresses macrophage foam cell formation and atherosclerosis in diabetic mice.
Collapse
Affiliation(s)
- Michishige Terasaki
- Department of Medicine, Division of Diabetes, Metabolism, and Endocrinology, Showa University School of Medicine, Tokyo, Japan
- Department of Cell and Molecular Biology, Karolinska Institutet, Stockholm, Sweden
| | - Munenori Hiromura
- Department of Medicine, Division of Diabetes, Metabolism, and Endocrinology, Showa University School of Medicine, Tokyo, Japan
| | - Yusaku Mori
- Department of Medicine, Division of Diabetes, Metabolism, and Endocrinology, Showa University School of Medicine, Tokyo, Japan
| | - Kyoko Kohashi
- Department of Medicine, Division of Diabetes, Metabolism, and Endocrinology, Showa University School of Medicine, Tokyo, Japan
| | - Hideki Kushima
- Department of Medicine, Division of Diabetes, Metabolism, and Endocrinology, Showa University School of Medicine, Tokyo, Japan
| | - Makoto Ohara
- Department of Medicine, Division of Diabetes, Metabolism, and Endocrinology, Showa University School of Medicine, Tokyo, Japan
| | - Takuya Watanabe
- Laboratory of Cardiovascular Medicine, Tokyo University of Pharmacy and Life Sciences, Tokyo, Japan
| | - Olov Andersson
- Department of Cell and Molecular Biology, Karolinska Institutet, Stockholm, Sweden
| | - Tsutomu Hirano
- Department of Medicine, Division of Diabetes, Metabolism, and Endocrinology, Showa University School of Medicine, Tokyo, Japan
- *Tsutomu Hirano:
| |
Collapse
|
47
|
Pei Z, Zhu L, Liu Y, Li N, Yang G, Liu H. Thymoquinone reduces kidney damage in apolipoprotein E-deficient mice fed a high-cholesterol diet. RSC Adv 2017. [DOI: 10.1039/c7ra07040c] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Hypercholesterolemia is a well-established risk factor for kidney injury that can lead to chronic kidney disease (CKD). Thymoquinone may be a potential therapeutic agent for kidney damage from hypercholesterolemia.
Collapse
Affiliation(s)
- Zuowei Pei
- Department of Cardiology
- Affiliated Zhongshan Hospital of Dalian University
- Dalian
- China
| | - Liyue Zhu
- Rehabilitation Center
- Zhejiang Hospital
- Hangzhou
- China
| | - Yingshu Liu
- Department of Endocrinology Dalian Municipal Central Hospital
- Dalian
- China
| | - Nan Li
- Department of Emergency
- The First Affiliated Hospital of Dalian Medical University
- Dalian
- China
| | - Guang Yang
- Department of Heart Intensive Care Unit
- The First Affiliated Hospital of Dalian Medical University
- Dalian 116011
- China
| | - Hongyang Liu
- Department of Heart Intensive Care Unit
- The First Affiliated Hospital of Dalian Medical University
- Dalian 116011
- China
| |
Collapse
|
48
|
Liu H, Li N, Liu Y, Xing J, Feng S, Li M, Liu J, Gao H, Lu Y, Liu H. The dipeptidyl peptidase-4 inhibitor teneligliptin reduces kidney damage from hypercholesterolemia in apolipoprotein E-deficient mice. RSC Adv 2017. [DOI: 10.1039/c6ra26718a] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Hypercholesterolemia is a well-established risk factor for kidney injury that can lead to chronic kidney disease (CKD).
Collapse
Affiliation(s)
- Hui Liu
- Department of Emergency
- The First Affiliated Hospital of Dalian Medical University
- Dalian
- China
| | - Nan Li
- Department of Emergency
- The First Affiliated Hospital of Dalian Medical University
- Dalian
- China
| | - Ying Liu
- Department of Emergency
- The First Affiliated Hospital of Dalian Medical University
- Dalian
- China
| | - Jing Xing
- Department of Emergency
- The First Affiliated Hospital of Dalian Medical University
- Dalian
- China
| | - Shuai Feng
- Department of Otolaryngology
- The First Affiliated Hospital of China Medical University
- Shenyang
- China
| | - Mengye Li
- Department of Special Medical Unit
- The First Affiliated Hospital of Dalian Medical University
- Dalian
- China
| | - Jinping Liu
- Department of Geratology
- Dalian Friendship Hospital Affiliated to Dalian Medical University
- Dalian
- China
| | - Huiling Gao
- College of Life and Health Sciences
- Northeastern University
- Shenyang
- China
| | - Yan Lu
- Department of Cardiology
- The First Affiliated Hospital of Dalian Medical University
- Dalian
- China
| | - Hongyang Liu
- Department of Heart Intensive Care Unit
- The First Affiliated Hospital of Dalian Medical University
- Dalian
- China
| |
Collapse
|
49
|
Wallner S, Grandl M, Liebisch G, Peer M, Orsó E, Sigrüner A, Sobota A, Schmitz G. oxLDL and eLDL Induced Membrane Microdomains in Human Macrophages. PLoS One 2016; 11:e0166798. [PMID: 27870891 PMCID: PMC5117723 DOI: 10.1371/journal.pone.0166798] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2016] [Accepted: 11/03/2016] [Indexed: 12/14/2022] Open
Abstract
Background Extravasation of macrophages and formation of lipid-laden foam cells are key events in the development and progression of atherosclerosis. The degradation of atherogenic lipoproteins subsequently leads to alterations in cellular lipid metabolism that influence inflammatory signaling. Especially sphingolipids and ceramides are known to be involved in these processes. We therefore analyzed monocyte derived macrophages during differentiation and after loading with enzymatically (eLDL) and oxidatively (oxLDL) modified low-density lipoproteins (LDL). Methods Primary human monocytes were isolated from healthy, normolipidemic blood donors using leukapheresis and counterflow elutriation. On the fourth day of MCSF-induced differentiation eLDL (40 μg/ml) or oxLDL (80 μg/ml) were added for 48h. Lipid species were analyzed by quantitative tandem mass spectrometry. Taqman qPCR was performed to investigate transcriptional changes in enzymes involved in sphingolipid metabolism. Furthermore, membrane lipids were studied using flow cytometry and confocal microscopy. Results MCSF dependent phagocytic differentiation of blood monocytes had only minor effects on the sphingolipid composition. Levels of total sphingomyelin and total ceramide remained unchanged, while lactosylceramides, cholesterylesters and free cholesterol decreased. At the species level most ceramide species showed a reduction upon phagocytic differentiation. Loading with eLDL preferentially increased cellular cholesterol while loading with oxLDL increased cellular ceramide content. Activation of the salvage pathway with a higher mRNA expression of acid and neutral sphingomyelinase, neutral sphingomyelinase activation associated factor and glucosylceramidase as well as increased surface expression of SMPD1 were identified as potentially underlying mechanisms. Moreover, flow-cytometric analysis revealed a higher cell-surface-expression of ceramide, lactosylceramide (CDw17), globotriaosylceramide (CD77), dodecasaccharide-ceramide (CD65s) and GM1 ganglioside upon oxLDL loading. ApoE in contrast to apoA-I preferentially bound to the ceramide enriched surfaces of oxLDL loaded cells. Confocal microscopy showed a co-localization of acid sphingomyelinase with ceramide rich membrane microdomains. Conclusion eLDL leads to the formation of lipid droplets and preferentially induces cholesterol/sphingomyelin rich membrane microdomains while oxLDL promotes the development of cholesterol/ceramide rich microdomains via activation of the salvage pathway.
Collapse
Affiliation(s)
- Stefan Wallner
- Institute for Clinical Chemistry and Laboratory Medicine, University Hospital Regensburg, Regensburg, Germany
| | - Margot Grandl
- Institute for Clinical Chemistry and Laboratory Medicine, University Hospital Regensburg, Regensburg, Germany
| | - Gerhard Liebisch
- Institute for Clinical Chemistry and Laboratory Medicine, University Hospital Regensburg, Regensburg, Germany
| | - Markus Peer
- Institute for Clinical Chemistry and Laboratory Medicine, University Hospital Regensburg, Regensburg, Germany
| | - Evelyn Orsó
- Institute for Clinical Chemistry and Laboratory Medicine, University Hospital Regensburg, Regensburg, Germany
| | - Alexander Sigrüner
- Institute for Clinical Chemistry and Laboratory Medicine, University Hospital Regensburg, Regensburg, Germany
| | - Andrzej Sobota
- Department of Cell Biology, Nencki Institute of Experimental Biology, Warsaw, Poland
| | - Gerd Schmitz
- Institute for Clinical Chemistry and Laboratory Medicine, University Hospital Regensburg, Regensburg, Germany
- * E-mail:
| |
Collapse
|
50
|
Ramachandran S, Vinitha A, Kartha CC. Cyclophilin A enhances macrophage differentiation and lipid uptake in high glucose conditions: a cellular mechanism for accelerated macro vascular disease in diabetes mellitus. Cardiovasc Diabetol 2016; 15:152. [PMID: 27809851 PMCID: PMC5094075 DOI: 10.1186/s12933-016-0467-5] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/17/2016] [Accepted: 10/25/2016] [Indexed: 12/22/2022] Open
Abstract
Background Vascular disease in diabetes is initiated by monocyte adhesion to vascular endothelium, transmigration and formation of foam cells. Increasing clinical evidence supports a role for the secretory protein, cyclophilin A in diabetic vascular disease. The means by which cyclophilin A contributes to vascular lesion development in diabetes is however largely unknown. Methods In this study we investigated using THP1 cells and human monocytes whether cyclophilin A under hyperglycemic conditions, functions in the inflammatory cascade as a chemoattractant and increases lipid uptake by formation of foam cells invitro. We developed an invitro model of monocytes cultured in 20 mm glucose (high glucose) equivalent to 360 mg/dL of plasma glucose levels. These monocytes were then differentiated into macrophages using PMA and subsequently transformed to lipid laden foam cells using oxidized low density lipoproteins in the presence and absence of cyclophilin A. This cellular model was used to study monocyte to macrophage differentiation, transmigration and foam cell formation. A similar cellular model using siRNA mediated transient elimination of the cyclophilin A gene as well as chemical inhibitors were used to further confirm the role of cyclophilin A in the differentiation and foam cell formation process. Results Cyclophilin A effectively increased migration of high glucose treated monocytes to the endothelial cell monolayer (p < 0.0001). In the presence of cyclophilin A, differentiated macrophages, when treated with oxLDL had a 36 percent increase in intracellular lipid accumulation (p = 0.01) when compared to cells treated with oxLDL alone. An increased flux of reactive oxygen species was also observed (p = 0.01). Inflammatory cytokines such as TNF-α, MCP-1 and cyclophilin A were significantly increased. Silencing cyclophilin A in THP-1 cells and human monocytes using siRNA or chemical inhibitor, TMN355 resulted in decrease in lipid uptake by 65–75% even after exposure to oxidized LDL. The expression of scavenger receptors expressed during differentiation process, CD36 and LOX-1 were decreased (p < 0.0001). Levels of extracellular cyclophilin A and other inflammatory cytokines such as TNF-α and MCP-1also significantly reduced. Conclusions Taken together, we describe here a possible cellular basis by which cyclophilin A may accelerate atherogenesis in diabetes mellitus. Electronic supplementary material The online version of this article (doi:10.1186/s12933-016-0467-5) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Surya Ramachandran
- Cardiovascular Diseases and Diabetes Biology, Rajiv Gandhi Centre for Biotechnology, Trivandrum, India.
| | - Anandan Vinitha
- Cardiovascular Diseases and Diabetes Biology, Rajiv Gandhi Centre for Biotechnology, Trivandrum, India
| | | |
Collapse
|