1
|
Snijckers RPM, Foks AC. Adaptive immunity and atherosclerosis: aging at its crossroads. Front Immunol 2024; 15:1350471. [PMID: 38686373 PMCID: PMC11056569 DOI: 10.3389/fimmu.2024.1350471] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Accepted: 03/28/2024] [Indexed: 05/02/2024] Open
Abstract
Adaptive immunity plays a profound role in atherosclerosis pathogenesis by regulating antigen-specific responses, inflammatory signaling and antibody production. However, as we age, our immune system undergoes a gradual functional decline, a phenomenon termed "immunosenescence". This decline is characterized by a reduction in proliferative naïve B- and T cells, decreased B- and T cell receptor repertoire and a pro-inflammatory senescence associated secretory profile. Furthermore, aging affects germinal center responses and deteriorates secondary lymphoid organ function and structure, leading to impaired T-B cell dynamics and increased autoantibody production. In this review, we will dissect the impact of aging on adaptive immunity and the role played by age-associated B- and T cells in atherosclerosis pathogenesis, emphasizing the need for interventions that target age-related immune dysfunction to reduce cardiovascular disease risk.
Collapse
Affiliation(s)
| | - Amanda C. Foks
- Division of BioTherapeutics, Leiden Academic Centre for Drug Research, Leiden University, Leiden, Netherlands
| |
Collapse
|
2
|
Lee MW, Koh JS, Kang S, Ryu H, Song IC, Lee HJ, Yun HJ, Kim SY, Kim SS, Jo DY. Abdominal aortic calcification in patients newly diagnosed with essential thrombocythemia. Blood Res 2023; 58:173-180. [PMID: 37853439 PMCID: PMC10758625 DOI: 10.5045/br.2023.2023125] [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: 07/05/2023] [Revised: 09/04/2023] [Accepted: 09/27/2023] [Indexed: 10/20/2023] Open
Abstract
Background Although atherosclerosis is likely to be involved in the development of arterial thrombotic events in patients with essential thrombocythemia (ET), abdominal aortic calcification (AAC) has rarely been investigated. We evaluated the prevalence and clinical relevance of AAC at the time of ET diagnosis. Methods This retrospective study included patients newly diagnosed with ET who underwent abdominal computed tomography (CT) at the time of diagnosis between January 2002 and December 2021 at Chungnam National University Hospital, Daejeon, Korea. CT images were reviewed and an aortic calcification score was assigned. Results Of the 94 patients (median age, 62 yr; range, 18‒90 yr), AAC was detected in 62 (66.0%). AAC was most commonly mild (33.0%), followed by moderate (22.7%) and severe (5.3%). Old age [odds ratio (OR), 34.37; 95% confidence interval (CI), 12.32‒95.91; P<0.001] was an independent risk factor for AAC. The patients with AAC had a higher WBC count (11.8±4.7 vs. 9.7±2.9×109/L, P=0.017), higher neutrophil-to-lymphocyte ratio (4.3±2.7 vs. 3.1±1.5, P=0.039), and higher JAK2V617F positivity (81.5% vs. 58.8%, P=0.020) compared to those without AAC. AAC was an independent risk factor for arterial thrombotic vascular events that occurred before or at diagnosis of ET (OR, 4.12; 95% CI, 1.11‒15.85; P=0.034). Conclusion AAC is common in patients with ET and is associated with arterial thrombotic events.
Collapse
Affiliation(s)
- Myung-Won Lee
- Division of Hematology/Oncology, Department of Internal Medicine, Chungnam National University College of Medicine, Daejeon, Korea
| | - Jeong Suk Koh
- Division of Hematology/Oncology, Department of Internal Medicine, Chungnam National University College of Medicine, Daejeon, Korea
| | - Sora Kang
- Division of Hematology/Oncology, Department of Internal Medicine, Chungnam National University College of Medicine, Daejeon, Korea
| | - Hyewon Ryu
- Division of Hematology/Oncology, Department of Internal Medicine, Chungnam National University College of Medicine, Daejeon, Korea
| | - Ik-Chan Song
- Division of Hematology/Oncology, Department of Internal Medicine, Chungnam National University College of Medicine, Daejeon, Korea
| | - Hyo-Jin Lee
- Division of Hematology/Oncology, Department of Internal Medicine, Chungnam National University College of Medicine, Daejeon, Korea
| | - Hwan-Jung Yun
- Division of Hematology/Oncology, Department of Internal Medicine, Chungnam National University College of Medicine, Daejeon, Korea
| | - Seon Young Kim
- Department of Laboratory Medicine, Chungnam National University College of Medicine, Daejeon, Korea
| | - Seong Soo Kim
- Department of Thoracic Radiology, Chungnam National University College of Medicine, Daejeon, Korea
| | - Deog-Yeon Jo
- Division of Hematology/Oncology, Department of Internal Medicine, Chungnam National University College of Medicine, Daejeon, Korea
| |
Collapse
|
3
|
Xiong X, Duan Z, Zhou H, Huang G, Niu L, Jin Y, Luo Z, Li W. The Increased TIGIT-Expressing CD3 +CD56 + Cells Are Associated with Coronary Artery Disease and Its Inflammatory Environment. Inflammation 2023; 46:2024-2036. [PMID: 37491572 DOI: 10.1007/s10753-023-01859-6] [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: 03/11/2023] [Revised: 05/24/2023] [Accepted: 06/13/2023] [Indexed: 07/27/2023]
Abstract
We aimed to examine the correlation of T-cell immunoglobulin and ITIM domain (TIGIT)-expressing CD3 + CD56 + cells (TNKS) with coronary artery disease (CAD), atherosclerotic lesion progression, and inflammatory environment. A total of 199 subjects, including 98 patients with acute coronary syndrome (ACS), 52 patients with chronic coronary syndrome (CCS), and 49 control subjects, were recruited in the study. The TIGIT-expressing TNKS were quantified by flow cytometric analysis; the severity of coronary artery lesions was evaluated by the Gensini score. Whole blood cells were stimulated with interleukin-2 (IL-2), interleukin-7 (IL-7), and interleukin-15 (IL-15) in presence or absence of STAT, PI3K, and P38 MAPK inhibitors, respectively. The TIGIT-expressing TNKS was significantly increased in patients with CAD, ACS, and CCS compared to the control group (P < 0.05). The TIGIT-expressing TNKS were independent predictors of CAD, ACS and CCS (P < 0.05). The TIGIT-expressing TNKS were positively associated with Gensini score (P < 0.05). The TIGIT-expressing TNKS was positively correlated with age, and being male (P < 0.05). The inflammatory microenviroment with increased IL-2, IL-7, and IL-15 contributed to upregulation of TIGIT expression in TNKS. PI3K and P38 MAPK inhibitors could inhibit the upregulation of TIGIT expression in TNKS induced by IL-2, IL-7, and IL-15. The TIGIT-expressing TNKS may be involved in common pathogenesis of ACS and CCS, and atherosclerotic lesion progression. Meanwhile, the increased TIGIT-expressing TNKS might be associated with a proatherogenic microenvironment or inflammatory microenvironment. PI3K and P38 MAPK signaling pathways were involved in the regulation of TIGIT expression.
Collapse
Affiliation(s)
- Xinlin Xiong
- Department of Cardiology, The Affiliated Hospital of Guizhou Medical University, Guiyang City, People's Republic of China
- Department of cardiology, Clinical Medical College & Affiliated Hospital of Chengdu University, Chengdu City, People's Republic of China
| | - Zonggang Duan
- Department of Cardiology, The Affiliated Hospital of Guizhou Medical University, Guiyang City, People's Republic of China
| | - Haiyan Zhou
- Department of Cardiology, The Affiliated Hospital of Guizhou Medical University, Guiyang City, People's Republic of China
| | - Guangwei Huang
- Department of Cardiology, The Affiliated Hospital of Guizhou Medical University, Guiyang City, People's Republic of China
| | - Li Niu
- Department of Cardiology, The Affiliated Hospital of Guizhou Medical University, Guiyang City, People's Republic of China
| | - Yingzhu Jin
- Department of Cardiology, The Affiliated Hospital of Guizhou Medical University, Guiyang City, People's Republic of China
| | - Zhenhua Luo
- Guizhou University School of Medicine, Guizhou University, Guiyang City, People's Republic of China.
- Department of Central Lab, Guizhou Provincial People's Hospital, The Affiliated People's Hospital of Guizhou Medical University, Guiyang City, People's Republic of China.
| | - Wei Li
- Department of Cardiology, The Affiliated Hospital of Guizhou Medical University, Guiyang City, People's Republic of China.
| |
Collapse
|
4
|
Busnelli M, Manzini S, Colombo A, Franchi E, Lääperi M, Laaksonen R, Chiesa G. Effect of Diets on Plasma and Aorta Lipidome: A Study in the apoE Knockout Mouse Model. Mol Nutr Food Res 2023; 67:e2200367. [PMID: 36419336 DOI: 10.1002/mnfr.202200367] [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: 06/06/2022] [Revised: 09/20/2022] [Indexed: 11/27/2022]
Abstract
SCOPE Specific lipid molecules circulating in plasma at low concentrations have emerged as biomarkers of atherosclerotic risk. The aim of the present study is that of evaluating, in an athero-prone mouse model, how different diets can affect plasma and aorta lipidome. METHODS AND RESULTS Thirty-six apoE knockout mice are divided in three groups and feed 12 weeks with diets differing for cholesterol and fatty acid content. Atherosclerosis is measured at the aortic sinus and aorta. Lipids are quantified in plasma and aorta with mass spectrometry. The cholesterol content of the diets is the main driver of lipid accumulation in plasma and aorta. The fatty acid composition of the diets affects plasma levels both of essential (linoleic acid) and nonessential (myristic and arachidonic acid) ones. Lipidomics show a comparable distribution, in plasma and aorta, of the main lipid components of oxidized LDL, including cholesteryl esters and lysophosphatidylcholines. Interestingly, lactosylceramide, glucosyl/galactosylceramide, and individual ceramide species are found to accumulate in diseased aortic segments. CONCLUSION Both the cholesterol and fatty acid content of the diets profoundly affect plasma lipidome. Aorta lipidome is likewise affected with the accumulation of specific lipids known as markers of atherosclerosis.
Collapse
Affiliation(s)
- Marco Busnelli
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, via Balzaretti, 9, Milan, 20133, Italy
| | - Stefano Manzini
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, via Balzaretti, 9, Milan, 20133, Italy
| | - Alice Colombo
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, via Balzaretti, 9, Milan, 20133, Italy
| | - Elsa Franchi
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, via Balzaretti, 9, Milan, 20133, Italy
| | | | - Reijo Laaksonen
- Zora Biosciences Oy, Espoo, 02150, Finland.,Finnish Cardiovascular Research Center, University of Tampere, Tampere, 33520, Finland
| | - Giulia Chiesa
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, via Balzaretti, 9, Milan, 20133, Italy
| |
Collapse
|
5
|
Karamanavi E, McVey DG, van der Laan SW, Stanczyk PJ, Morris GE, Wang Y, Yang W, Chan K, Poston RN, Luo J, Zhou X, Gong P, Jones PD, Cao J, Kostogrys RB, Webb TR, Pasterkamp G, Yu H, Xiao Q, Greer PA, Stringer EJ, Samani NJ, Ye S. The FES Gene at the 15q26 Coronary-Artery-Disease Locus Inhibits Atherosclerosis. Circ Res 2022; 131:1004-1017. [PMID: 36321446 PMCID: PMC9770135 DOI: 10.1161/circresaha.122.321146] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Revised: 10/16/2022] [Accepted: 10/17/2022] [Indexed: 12/05/2022]
Abstract
BACKGROUND Genome-wide association studies have discovered a link between genetic variants on human chromosome 15q26.1 and increased coronary artery disease (CAD) susceptibility; however, the underlying pathobiological mechanism is unclear. This genetic locus contains the FES (FES proto-oncogene, tyrosine kinase) gene encoding a cytoplasmic protein-tyrosine kinase involved in the regulation of cell behavior. We investigated the effect of the 15q26.1 variants on FES expression and whether FES plays a role in atherosclerosis. METHODS AND RESULTS Analyses of isogenic monocytic cell lines generated by CRISPR (clustered regularly interspaced short palindromic repeats)-mediated genome editing showed that monocytes with an engineered 15q26.1 CAD risk genotype had reduced FES expression. Small-interfering-RNA-mediated knockdown of FES promoted migration of monocytes and vascular smooth muscle cells. A phosphoproteomics analysis showed that FES knockdown altered phosphorylation of a number of proteins known to regulate cell migration. Single-cell RNA-sequencing revealed that in human atherosclerotic plaques, cells that expressed FES were predominately monocytes/macrophages, although several other cell types including smooth muscle cells also expressed FES. There was an association between the 15q26.1 CAD risk genotype and greater numbers of monocytes/macrophage in human atherosclerotic plaques. An animal model study demonstrated that Fes knockout increased atherosclerotic plaque size and within-plaque content of monocytes/macrophages and smooth muscle cells, in apolipoprotein E-deficient mice fed a high fat diet. CONCLUSIONS We provide substantial evidence that the CAD risk variants at the 15q26.1 locus reduce FES expression in monocytes and that FES depletion results in larger atherosclerotic plaques with more monocytes/macrophages and smooth muscle cells. This study is the first demonstration that FES plays a protective role against atherosclerosis and suggests that enhancing FES activity could be a potentially novel therapeutic approach for CAD intervention.
Collapse
Affiliation(s)
- Elisavet Karamanavi
- Department of Cardiovascular Sciences, University of Leicester, and National Institute for Health Research Leicester Biomedical Research Centre, Leicester, United Kingdom (E.K., D.G.M., P.J.S., G.E.M., P.G., P.D.J., T.R.W., E.J.S., N.J.S., S.Y.)
| | - David G. McVey
- Department of Cardiovascular Sciences, University of Leicester, and National Institute for Health Research Leicester Biomedical Research Centre, Leicester, United Kingdom (E.K., D.G.M., P.J.S., G.E.M., P.G., P.D.J., T.R.W., E.J.S., N.J.S., S.Y.)
| | - Sander W. van der Laan
- Central Diagnostic Laboratory, University of Utrecht, The Netherlands (S.W.v.d.L., G.P.)
| | - Paulina J. Stanczyk
- Department of Cardiovascular Sciences, University of Leicester, and National Institute for Health Research Leicester Biomedical Research Centre, Leicester, United Kingdom (E.K., D.G.M., P.J.S., G.E.M., P.G., P.D.J., T.R.W., E.J.S., N.J.S., S.Y.)
| | - Gavin E. Morris
- Department of Cardiovascular Sciences, University of Leicester, and National Institute for Health Research Leicester Biomedical Research Centre, Leicester, United Kingdom (E.K., D.G.M., P.J.S., G.E.M., P.G., P.D.J., T.R.W., E.J.S., N.J.S., S.Y.)
| | - Yifan Wang
- Cardiovascular Disease Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore (Y.W., H.Y., S.Y.)
| | - Wei Yang
- Shantou University Medical College, China (W.Y., J.C., S.Y.)
| | - Kenneth Chan
- William Harvey Research Institute, Queen Mary University of London, United Kingdom (K.C., R.N.P., J.L., X.Z., Q.X.)
| | - Robin N. Poston
- William Harvey Research Institute, Queen Mary University of London, United Kingdom (K.C., R.N.P., J.L., X.Z., Q.X.)
| | - Jun Luo
- William Harvey Research Institute, Queen Mary University of London, United Kingdom (K.C., R.N.P., J.L., X.Z., Q.X.)
| | - Xinmiao Zhou
- William Harvey Research Institute, Queen Mary University of London, United Kingdom (K.C., R.N.P., J.L., X.Z., Q.X.)
| | - Peng Gong
- Department of Cardiovascular Sciences, University of Leicester, and National Institute for Health Research Leicester Biomedical Research Centre, Leicester, United Kingdom (E.K., D.G.M., P.J.S., G.E.M., P.G., P.D.J., T.R.W., E.J.S., N.J.S., S.Y.)
| | - Peter D. Jones
- Department of Cardiovascular Sciences, University of Leicester, and National Institute for Health Research Leicester Biomedical Research Centre, Leicester, United Kingdom (E.K., D.G.M., P.J.S., G.E.M., P.G., P.D.J., T.R.W., E.J.S., N.J.S., S.Y.)
| | - Junjun Cao
- Shantou University Medical College, China (W.Y., J.C., S.Y.)
| | - Renata B. Kostogrys
- Department of Human Nutrition, University of Agriculture in Kraków, Poland (R.B.K.)
| | - Tom R. Webb
- Department of Cardiovascular Sciences, University of Leicester, and National Institute for Health Research Leicester Biomedical Research Centre, Leicester, United Kingdom (E.K., D.G.M., P.J.S., G.E.M., P.G., P.D.J., T.R.W., E.J.S., N.J.S., S.Y.)
| | - Gerard Pasterkamp
- Central Diagnostic Laboratory, University of Utrecht, The Netherlands (S.W.v.d.L., G.P.)
| | - Haojie Yu
- Cardiovascular Disease Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore (Y.W., H.Y., S.Y.)
| | - Qingzhong Xiao
- William Harvey Research Institute, Queen Mary University of London, United Kingdom (K.C., R.N.P., J.L., X.Z., Q.X.)
| | - Peter A. Greer
- Department of Pathology and Molecular Medicine, Queen’s University, Kingston, Canada (P.A.G.)
| | - Emma J. Stringer
- Department of Cardiovascular Sciences, University of Leicester, and National Institute for Health Research Leicester Biomedical Research Centre, Leicester, United Kingdom (E.K., D.G.M., P.J.S., G.E.M., P.G., P.D.J., T.R.W., E.J.S., N.J.S., S.Y.)
| | - Nilesh J. Samani
- Department of Cardiovascular Sciences, University of Leicester, and National Institute for Health Research Leicester Biomedical Research Centre, Leicester, United Kingdom (E.K., D.G.M., P.J.S., G.E.M., P.G., P.D.J., T.R.W., E.J.S., N.J.S., S.Y.)
| | - Shu Ye
- Department of Cardiovascular Sciences, University of Leicester, and National Institute for Health Research Leicester Biomedical Research Centre, Leicester, United Kingdom (E.K., D.G.M., P.J.S., G.E.M., P.G., P.D.J., T.R.W., E.J.S., N.J.S., S.Y.)
- Cardiovascular Disease Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore (Y.W., H.Y., S.Y.)
- Shantou University Medical College, China (W.Y., J.C., S.Y.)
| |
Collapse
|
6
|
Dong XH, Lu ZF, Kang CM, Li XH, Haworth KE, Ma X, Lu JB, Liu XH, Fang FC, Wang CS, Ye JH, Zheng L, Wang Q, Ye S, Hu YW. The Long Noncoding RNA RP11-728F11.4 Promotes Atherosclerosis. Arterioscler Thromb Vasc Biol 2021; 41:1191-1204. [PMID: 33406853 DOI: 10.1161/atvbaha.120.315114] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
OBJECTIVE Noncoding RNAs are emerging as important players in gene regulation and cardiovascular diseases. Their roles in the pathogenesis of atherosclerosis are not fully understood. The purpose of this study was to determine the role played by a previously uncharacterized long noncoding RNA, RP11-728F11.4, in the development of atherosclerosis and the mechanisms by which it acts. Approach and Results: Expression microarray analysis revealed that atherosclerotic plaques had increased expression of RP11-728F11.4 as well as the cognate gene FXYD6 (FXYD domain containing ion transport regulator 6), which encodes a modulator of Na+/K+-ATPase. In vitro experiments showed that RP11-728F11.4 interacted with the RNA-binding protein EWSR1 (Ewings sarcoma RNA binding protein-1) and upregulated FXYD6 expression. Lentivirus-induced overexpression of RP11-728F11.4 in cultured monocytes-derived macrophages resulted in higher Na+/K+-ATPase activity, intracellular cholesterol accumulation, and increased proinflammatory cytokine production. The effects of RP11-728F11.4 were enhanced by siRNA-mediated knockdown of EWSR1 and reduced by downregulation of FXYD domain containing ion transport regulator 6. In vivo experiments in apoE knockout mice fed a Western diet demonstrated that RP11-728F11.4 increased proinflammatory cytokine production and augmented atherosclerotic lesions. CONCLUSIONS RP11-728F11.4 promotes atherosclerosis, with an influence on cholesterol homeostasis and proinflammatory molecule production, thus representing a potential therapeutic target. Graphic Abstract: A graphic abstract is available for this article.
Collapse
MESH Headings
- Animals
- Atherosclerosis/etiology
- Atherosclerosis/genetics
- Atherosclerosis/metabolism
- Cells, Cultured
- Cholesterol/metabolism
- Cytokines/metabolism
- Disease Models, Animal
- Endothelial Cells/metabolism
- Female
- Gene Knockdown Techniques
- Humans
- Ion Channels/genetics
- Ion Channels/metabolism
- Lipid Metabolism
- Male
- Mice
- Mice, Inbred C57BL
- Mice, Knockout, ApoE
- Middle Aged
- Plaque, Atherosclerotic/etiology
- Plaque, Atherosclerotic/genetics
- Plaque, Atherosclerotic/pathology
- RNA, Long Noncoding/genetics
- RNA, Long Noncoding/metabolism
- RNA-Binding Protein EWS/antagonists & inhibitors
- RNA-Binding Protein EWS/genetics
- RNA-Binding Protein EWS/metabolism
- Sodium-Potassium-Exchanging ATPase/metabolism
- Up-Regulation
Collapse
Affiliation(s)
- Xian-Hui Dong
- Department of Clinical Laboratory, Guangzhou Women and Children Medical Center, Guangzhou Medical University, China (X.-H.D., Y.-W.H.)
| | - Zhi-Feng Lu
- Laboratory Medicine Center (Z.-F.L., C.-M.K., X.-H. Li, X.-H. Liu, L.Z., Q.W., Y.-W.H.), Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Chun-Min Kang
- Laboratory Medicine Center (Z.-F.L., C.-M.K., X.-H. Li, X.-H. Liu, L.Z., Q.W., Y.-W.H.), Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Xue-Heng Li
- Laboratory Medicine Center (Z.-F.L., C.-M.K., X.-H. Li, X.-H. Liu, L.Z., Q.W., Y.-W.H.), Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Kim E Haworth
- Department of Cardiovascular Sciences and NIHR (National Institute for Health Research) Leicester Biomedical Research Centre, University of Leicester, United Kingdom (K.E.H., S.Y.)
| | - Xin Ma
- Department of Anesthesiology (X.M.), Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Jing-Bo Lu
- Department of Vascular Surgery (J.-B.L.), Nanfang Hospital, Southern Medical University, Guangzhou, China
- Department of Vascular Surgery, Shenzhen Sun Yat-Sen Cardiovascular Hospital, China (J.-B.L.)
| | - Xue-Hui Liu
- Laboratory Medicine Center (Z.-F.L., C.-M.K., X.-H. Li, X.-H. Liu, L.Z., Q.W., Y.-W.H.), Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Fu-Chun Fang
- Department of Stomatology (F.-C.F.), Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Claire S Wang
- Gonville and Caius College, University of Cambridge, United Kindgom (C.S.W.)
| | - John H Ye
- University Hospitals of Leicester NHS Trust, United Kingdom (J.H.Y.)
| | - Lei Zheng
- Laboratory Medicine Center (Z.-F.L., C.-M.K., X.-H. Li, X.-H. Liu, L.Z., Q.W., Y.-W.H.), Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Qian Wang
- Laboratory Medicine Center (Z.-F.L., C.-M.K., X.-H. Li, X.-H. Liu, L.Z., Q.W., Y.-W.H.), Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Shu Ye
- Department of Cardiovascular Sciences and NIHR (National Institute for Health Research) Leicester Biomedical Research Centre, University of Leicester, United Kingdom (K.E.H., S.Y.)
- Shantou University Medical College, China (S.Y.)
| | - Yan-Wei Hu
- Department of Clinical Laboratory, Guangzhou Women and Children Medical Center, Guangzhou Medical University, China (X.-H.D., Y.-W.H.)
- Laboratory Medicine Center (Z.-F.L., C.-M.K., X.-H. Li, X.-H. Liu, L.Z., Q.W., Y.-W.H.), Nanfang Hospital, Southern Medical University, Guangzhou, China
| |
Collapse
|
7
|
Chronic treatment with cinnamaldehyde prevents spontaneous atherosclerotic plaque development in ovariectomized LDLr-/- female mice. PHARMANUTRITION 2020. [DOI: 10.1016/j.phanu.2020.100205] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
|
8
|
Fu Y, Yuan PP, Cao YG, Ke YY, Zhang Q, Hou Y, Zhang YL, Feng WS, Zheng XK. Geniposide in Gardenia jasminoides var. radicans Makino modulates blood pressure via inhibiting WNK pathway mediated by the estrogen receptors. J Pharm Pharmacol 2020; 72:1956-1969. [DOI: 10.1111/jphp.13361] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Accepted: 07/25/2020] [Indexed: 12/15/2022]
Abstract
Abstract
Objectives
To investigate the effects of geniposide in an iridoid found in Gardenia jasminoides var. radicans Makino (GJRM) in spontaneous hypertensive rat (SHR) and explore the possible mechanisms.
Methods
In this study, we detected the content of geniposide in GJRM by high-performance liquid chromatography (HPLC). Then, we used acute diuretic experiments to determine whether geniposide has diuretic effect. Moreover, we carried out experiments on SHR to further study the mechanism of hypertension, while real-time PCR, Western blot and immunohistochemistry were used for the experiments in vivo test. Hypotonic model was used for in vitro test.
Key findings
Our data showed that the content of geniposide in the extract of GJRM is 27.54%. Meanwhile, 50 mg/kg geniposide showed the strongest effect on promoting urine volume. Further study indicated that the extract of GJRM and geniposide could significantly reduce blood pressure and promote the excretion of urine and Na+ in SHR. In addition, geniposide significantly inhibited the activation of the with-no-lysine kinase (WNK) signalling pathway and significantly increases the protein expressions of estrogen receptor α (ERα), estrogen receptor β (ERβ) and G protein-coupled receptor 30 (GPR30) in SHR. In hypotonic model, geniposide significantly inhibits the phosphorylation of NKCC and NCC and could be antagonistic to estrogen receptor antagonists.
Conclusions
Collectively, we would suggest that geniposide may potentially be utilized as an adjunct to existing thiazide and thiazide-like diuretics to control hypertension, mainly through inhibiting the activation of the WNK signalling pathway mediated by the estrogen receptor.
Collapse
Affiliation(s)
- Yang Fu
- Henan University of Chinese Medicine, Zhengzhou, Henan, China
- The Engineering and Technology Center for Chinese Medicine Development of Henan Province, Zhengzhou, Henan, China
| | - Pei-pei Yuan
- Henan University of Chinese Medicine, Zhengzhou, Henan, China
- The Engineering and Technology Center for Chinese Medicine Development of Henan Province, Zhengzhou, Henan, China
| | - Yan-gang Cao
- Henan University of Chinese Medicine, Zhengzhou, Henan, China
- The Engineering and Technology Center for Chinese Medicine Development of Henan Province, Zhengzhou, Henan, China
| | - Ying-ying Ke
- Henan University of Chinese Medicine, Zhengzhou, Henan, China
- The Engineering and Technology Center for Chinese Medicine Development of Henan Province, Zhengzhou, Henan, China
| | - Qi Zhang
- Henan University of Chinese Medicine, Zhengzhou, Henan, China
- The Engineering and Technology Center for Chinese Medicine Development of Henan Province, Zhengzhou, Henan, China
| | - Ying Hou
- Henan University of Chinese Medicine, Zhengzhou, Henan, China
- The Engineering and Technology Center for Chinese Medicine Development of Henan Province, Zhengzhou, Henan, China
| | - Yan-li Zhang
- Henan University of Chinese Medicine, Zhengzhou, Henan, China
- The Engineering and Technology Center for Chinese Medicine Development of Henan Province, Zhengzhou, Henan, China
| | - Wei-sheng Feng
- Henan University of Chinese Medicine, Zhengzhou, Henan, China
- The Engineering and Technology Center for Chinese Medicine Development of Henan Province, Zhengzhou, Henan, China
| | - Xiao-ke Zheng
- Henan University of Chinese Medicine, Zhengzhou, Henan, China
- The Engineering and Technology Center for Chinese Medicine Development of Henan Province, Zhengzhou, Henan, China
| |
Collapse
|
9
|
Muramatsu M, Nakagawa S, Osawa T, Toyono T, Uemura A, Kidoya H, Takakura N, Usui T, Ryeom S, Minami T. Loss of Down Syndrome Critical Region-1 Mediated-Hypercholesterolemia Accelerates Corneal Opacity Via Pathological Neovessel Formation. Arterioscler Thromb Vasc Biol 2020; 40:2425-2439. [PMID: 32787520 DOI: 10.1161/atvbaha.120.315003] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
OBJECTIVE The calcineurin-NFAT (nuclear factor for activated T cells)-DSCR (Down syndrome critical region)-1 pathway plays a crucial role as the downstream effector of VEGF (vascular endothelial growth factor)-mediated tumor angiogenesis in endothelial cells. A role for DSCR-1 in different organ microenvironment such as the cornea and its role in ocular diseases is not well understood. Corneal changes can be indicators of various disease states and are easily detected through ocular examinations. Approach and Results: The presentation of a corneal arcus or a corneal opacity due to lipid deposition in the cornea often indicates hyperlipidemia and in most cases, hypercholesterolemia. Although the loss of Apo (apolipoprotein) E has been well characterized and is known to lead to elevated serum cholesterol levels, there are few corneal changes observed in ApoE-/- mice. In this study, we show that the combined loss of ApoE and DSCR-1 leads to a dramatic increase in serum cholesterol levels and severe corneal opacity with complete penetrance. The cornea is normally maintained in an avascular state; however, loss of Dscr-1 is sufficient to induce hyper-inflammatory and -oxidative condition, increased corneal neovascularization, and lymphangiogenesis. Furthermore, immunohistological analysis and genome-wide screening revealed that loss of Dscr-1 in mice triggers increased immune cell infiltration and upregulation of SDF (stromal derived factor)-1 and its receptor, CXCR4 (C-X-C motif chemokine ligand receptor-4), potentiating this signaling axis in the cornea, thereby contributing to pathological corneal angiogenesis and opacity. CONCLUSIONS This study is the first demonstration of the critical role for the endogenous inhibitor of calcineurin, DSCR-1, and pathological corneal angiogenesis in hypercholesterolemia induced corneal opacity.
Collapse
Affiliation(s)
- Masashi Muramatsu
- Division of Molecular and Vascular Biology, IRDA, Kumamoto University, Japan (M.M., T.M.)
| | - Suguru Nakagawa
- Division of Genome Science (S.N.), RCAST, the University of Tokyo, Japan.,Department Ophthalmology, Graduate School of Medicine, the University of Tokyo, Japan (S.N., T.T., T.U.)
| | - Tsuyoshi Osawa
- Integrative Nutriomics (T.O.), RCAST, the University of Tokyo, Japan
| | - Tetsuya Toyono
- Department Ophthalmology, Graduate School of Medicine, the University of Tokyo, Japan (S.N., T.T., T.U.)
| | - Akiyoshi Uemura
- Department Retinal Vascular Biology, Nagoya City University Graduate School of Medical Sciences, Japan (A.U.)
| | - Hiroyasu Kidoya
- Department Signal Transduction, RIMD, Osaka University, Japan (H.K., N.T.)
| | - Nobuyuki Takakura
- Department Signal Transduction, RIMD, Osaka University, Japan (H.K., N.T.)
| | - Tomohiko Usui
- Department Ophthalmology, Graduate School of Medicine, the University of Tokyo, Japan (S.N., T.T., T.U.)
| | - Sandra Ryeom
- Department Cancer Biology, University of Pennsylvania (S.R.)
| | - Takashi Minami
- Division of Molecular and Vascular Biology, IRDA, Kumamoto University, Japan (M.M., T.M.)
| |
Collapse
|
10
|
Impact of Sex and Age on the Mevalonate Pathway in the Brain: A Focus on Effects Induced by Maternal Exposure to Exogenous Compounds. Metabolites 2020; 10:metabo10080304. [PMID: 32722471 PMCID: PMC7463490 DOI: 10.3390/metabo10080304] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Revised: 07/19/2020] [Accepted: 07/23/2020] [Indexed: 12/21/2022] Open
Abstract
The mevalonate pathway produces cholesterol and other compounds crucial for numerous cellular processes. It is well known that age and sex modulate this pathway in the liver. Recently, similar effects were also noted in different brain areas, suggesting that alterations of the mevalonate pathway are at the root of marked sex-specific disparities in some neurodevelopmental disorders related to disturbed cholesterol homeostasis. Here, we show how the mevalonate pathway is modulated in a sex-, age- and region-specific manner, and how maternal exposure to exogenous compounds can disturb the regulation of this pathway in the brain, possibly inducing functional alterations.
Collapse
|
11
|
Lavin Plaza B, Phinikaridou A, Andia ME, Potter M, Lorrio S, Rashid I, Botnar RM. Sustained Focal Vascular Inflammation Accelerates Atherosclerosis in Remote Arteries. Arterioscler Thromb Vasc Biol 2020; 40:2159-2170. [PMID: 32673527 PMCID: PMC7447189 DOI: 10.1161/atvbaha.120.314387] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
OBJECTIVE Evidence from preclinical and clinical studies has demonstrated that myocardial infarction promotes atherosclerosis progression. The impact of focal vascular inflammation on the progression and phenotype of remote atherosclerosis remains unknown. Approach and Results: We used a novel ApoE-/- knockout mouse model of sustained arterial inflammation, initiated by mechanical injury in the abdominal aorta. Using serial in vivo molecular MRI and ex vivo histology and flow cytometry, we demonstrate that focal arterial inflammation triggered by aortic injury, accelerates atherosclerosis in the remote brachiocephalic artery. The brachiocephalic artery atheroma had distinct histological features including increased plaque size, plaque permeability, necrotic core to collagen ratio, infiltration of more inflammatory monocyte subsets, and reduced collagen content. We also found that arterial inflammation following focal vascular injury evoked a prolonged systemic inflammatory response manifested as a persistent increase in serum IL-6 (interleukin 6). Finally, we demonstrate that 2 therapeutic interventions-pravastatin and minocycline-had distinct anti-inflammatory effects at the plaque and systemic level. CONCLUSIONS We show for the first time that focal arterial inflammation in response to vascular injury enhances systemic vascular inflammation, accelerates remote atheroma progression and induces plaques more inflamed, lipid-rich, and collagen-poor in the absence of ischemic myocardial injury. This inflammatory cascade is modulated by pravastatin and minocycline treatments, which have anti-inflammatory effects at both plaque and systemic levels that mitigate atheroma progression.
Collapse
Affiliation(s)
- Begoña Lavin Plaza
- From the School of Biomedical Engineering and Imaging Sciences, King's College London, United Kingdom (B.L.P., A.P., M.P., S.L., I.R., R.M.B.)
| | - Alkystis Phinikaridou
- From the School of Biomedical Engineering and Imaging Sciences, King's College London, United Kingdom (B.L.P., A.P., M.P., S.L., I.R., R.M.B.)
| | - Marcelo E Andia
- Radiology Department & Millennium Nucleus for Cardiovascular Magnetic Resonance (M.E.A.), Pontificia Universidad Católica de Chile
| | - Myles Potter
- From the School of Biomedical Engineering and Imaging Sciences, King's College London, United Kingdom (B.L.P., A.P., M.P., S.L., I.R., R.M.B.)
| | - Silvia Lorrio
- From the School of Biomedical Engineering and Imaging Sciences, King's College London, United Kingdom (B.L.P., A.P., M.P., S.L., I.R., R.M.B.)
| | - Imran Rashid
- From the School of Biomedical Engineering and Imaging Sciences, King's College London, United Kingdom (B.L.P., A.P., M.P., S.L., I.R., R.M.B.).,Case Cardiovascular Research Institute, Case Western Reserve University, Cleveland, OH (I.R.)
| | - Rene M Botnar
- From the School of Biomedical Engineering and Imaging Sciences, King's College London, United Kingdom (B.L.P., A.P., M.P., S.L., I.R., R.M.B.).,Escuela de Ingeniería (R.M.B.), Pontificia Universidad Católica de Chile
| |
Collapse
|
12
|
A Timing Effect of 17-β Estradiol on Atherosclerotic Lesion Development in Female ApoE -/- Mice. Int J Mol Sci 2020; 21:ijms21134710. [PMID: 32630298 PMCID: PMC7369926 DOI: 10.3390/ijms21134710] [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: 06/04/2020] [Revised: 06/25/2020] [Accepted: 06/29/2020] [Indexed: 11/18/2022] Open
Abstract
Differences in size or composition of existing plaques at the initiation of estrogen (E2) therapy may underpin evidence of increased risk of atherosclerosis-associated clinical sequelae. We investigated whether E2 had divergent effects on actively-growing versus established-advanced atherosclerotic lesions. Eight weeks of subcutaneous bi-weekly injections of 3 µg/g 17β-estradiol (n = 18) or vehicle control (n = 22) were administered to female Apolipoprotein null-mice aged 25- or 45 weeks old. Histological assessment of lesion size within the brachiocephalic artery was conducted. Lesion composition was also assessed with acellular, calcification and fibrosis areas measured and other cellular features (intimal thickening, foam cells, lipid pools and cholesterol) scored (0–3) for severity. The comparison showed increased lesion size and calcified area with advancing age but no effect of E2. However, subtle changes in composition were observed following E2. Within the younger group, E2 increased intima thickening and acceleration of calcification. In the older group, E2 increased the thickness of the lesion cap. Therefore, this study shows different effects of E2 depending on the underlying stage of lesion development at the time of initiation of treatment. These divergent changes help explain the controversy of the adverse effects of E2 treatment in cardiovascular disease.
Collapse
|
13
|
Abstract
Atherosclerosis is a chronic inflammatory vascular disease and the predominant cause of heart attack and ischemic stroke. Despite the well-known sexual dimorphism in the incidence and complications of atherosclerosis, there are relatively limited data in the clinical and preclinical literature to rigorously address mechanisms underlying sex as a biological variable in atherosclerosis. In multiple histological and imaging studies, overall plaque burden and markers of inflammation appear to be greater in men than women and are predictive of cardiovascular events. However, while younger women are relatively protected from cardiovascular disease, by the seventh decade, the incidence of myocardial infarction in women ultimately surpasses that of men, suggesting an interaction between sex and age. Most preclinical studies in animal atherosclerosis models do not examine both sexes, and even in those that do, well-powered direct statistical comparisons for sex as an independent variable remain rare. This article reviews the available data. Overall, male animals appear to have more inflamed yet smaller plaques compared to female animals. Plaque inflammation is often used as a surrogate end point for plaque vulnerability in animals. The available data support the notion that rather than plaque size, plaque inflammation may be more relevant in assessing sex-specific mechanisms since the findings correlate with the sex difference in ischemic events and mortality and thus may be more reflective of the human condition. Overall, the number of preclinical studies directly comparing plaque inflammation between the sexes is extremely limited relative to the vast literature exploring atherosclerosis mechanisms. Failure to include both sexes and to address age in mechanistic atherosclerosis studies are missed opportunities to uncover underlying sex-specific mechanisms. Understanding the mechanisms driving sex as a biological variable in atherosclerotic disease is critical to future precision medicine strategies to mitigate what is still the leading cause of death of men and women worldwide.
Collapse
Affiliation(s)
- Joshua J. Man
- Molecular Cardiology Research Institute, Tufts Medical Center, Boston, MA
- Graduate School of Biomedical Sciences, Tufts University School of Medicine, Boston, MA
| | - Joshua A. Beckman
- Cardiovascular Division, Vanderbilt University Medical Center, Nashville, TN
| | - Iris Z. Jaffe
- Molecular Cardiology Research Institute, Tufts Medical Center, Boston, MA
| |
Collapse
|
14
|
Lin K, Chen H, Chen X, Qian J, Huang S, Huang W. Efficacy of Curcumin on Aortic Atherosclerosis: A Systematic Review and Meta-Analysis in Mouse Studies and Insights into Possible Mechanisms. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2020; 2020:1520747. [PMID: 31998433 PMCID: PMC6973199 DOI: 10.1155/2020/1520747] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/29/2019] [Revised: 11/30/2019] [Accepted: 12/19/2019] [Indexed: 02/08/2023]
Abstract
Since the first report in 2005, accumulating interests have been focused on the effect of curcumin in atherosclerosis with discrepancies. Therefore, we conducted a systematic review and meta-analysis to comprehensively estimate its effect against atherosclerosis. Literature search was performed on the database of PubMed, EMBASE, and Cochrane Library to identify relevant studies which estimated the effect of curcumin in atherosclerosis. Reporting effects on aortic lesion area was the primary outcome while effects on serum lipid profiles and circulating inflammatory markers were the secondary outcome. A total of 10 studies including 14 independent pairwise experiments were included in our analysis. We clarified that curcumin could significantly reduce aortic atherosclerotic lesion area (SMD = -0.89, 95% CI: -1.36 to -0.41, P = 0.0003), decrease serum lipid profiles (Tc, MD = -1.005, 95% CI: -1.885 to -0.124, P = 0.025; TG, MD = -0.045, 95% CI: -0.088 to -0.002, P = 0.042; LDL-c, MD = -0.523, 95% CI: -0.896 to -0.149, P = 0.006) as well as plasma inflammatory indicators (TNF-α, MD = -56.641, 95% CI: -86.848 to -26.433, P < 0.001; IL-1β, MD = -5.089, 95% CI: -8.559 to -1.619, P = 0.004). Dose-response meta-analysis predicted effective dosage of curcumin between 0 and 347 mg/kg BW per day, which was safe and nontoxic according to the existing publications. The underlying mechanisms were also discussed and might be associated with the modulation of lipid transport and inflammation in cells within artery walls as well as indirect modulations in other tissues. Clinical evidence from nonatherosclerosis populations revealed that curcumin would lower the lipid profiles and inflammatory responses as it has in a mouse model. However, standard preclinical animal trial designs are still needed; further studies focusing on the optimal dose of curcumin against atherosclerosis and RCTs directly in atherosclerosis patients are also warranted.
Collapse
Affiliation(s)
- Ke Lin
- Department of Cardiology, The Key Lab of Cardiovascular Disease of Wenzhou, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province 325035, China
| | - Huaijun Chen
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang Province 310000, China
| | - Xiaojun Chen
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang Province 325035, China
| | - Jinfu Qian
- Department of Cardiology, The Key Lab of Cardiovascular Disease of Wenzhou, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province 325035, China
| | - Shushi Huang
- Department of Cardiology, The Key Lab of Cardiovascular Disease of Wenzhou, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province 325035, China
| | - Weijian Huang
- Department of Cardiology, The Key Lab of Cardiovascular Disease of Wenzhou, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province 325035, China
| |
Collapse
|
15
|
Ni Y, Liu M, Yu H, Chen Y, Liu Y, Chen S, Ruan J, Da A, Zhang Y, Wang T. Desmethylbellidifolin From Gentianella acuta Ameliorate TNBS-Induced Ulcerative Colitis Through Antispasmodic Effect and Anti-Inflammation. Front Pharmacol 2019; 10:1104. [PMID: 31616306 PMCID: PMC6764246 DOI: 10.3389/fphar.2019.01104] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2019] [Accepted: 08/28/2019] [Indexed: 12/24/2022] Open
Abstract
Desmethylbellidifolin (DMB) is a natural xanthone extracted from Gentianella acuta, which is used as the antidiarrhea drug in traditional Mongolian medicines. It remains unknown whether DMB can ameliorate ulcerative colitis (UC). In this study, trinitrobenzenesulfonic acid (TNBS)-induced colitis rats were treated with G. acuta extract (GAE) or DMB for 10 days. Body weight, food and water intake, rectal bleeding score, diarrhea score, and histopathological parameters were measured. Rat colon were collected to determine myeloperoxidase, nitric oxide levels, and inflammatory cytokines expression. In addition, the role of DMB on lipopolysaccharide stimulated RAW264.7 cell inflammatory response and intestine smooth muscle contraction was determined. The results showed that GAE and DMB treatment could significantly alleviate TNBS-induced UC. Colon morphological alteration, nitric oxide level, and inflammatory cytokines level, such as nitric oxide synthase, interleukin-6, tumor necrosis factor-α, and cyclooxygenase-2, were decreased. In addition, DMB attenuated lipopolysaccharide-induced nitric oxide release and proinflammatory cytokine expression in RAW264.7 cells. In isolated mice intestinal tissue, DMB also reduced the intestine smooth muscle spontaneous contraction and inhibited KCl, acetylcholine, BaCl2, or histamine-induced intestine smooth muscle active tension, while the active frequency was unaffected. Our results demonstrated that GAE and its active constituent DMB could inhibit TNBS-induced UC, reducing inflammatory response and alleviate colon muscle spasm, suggesting that DMB may be a good candidate for subsequent development as a multitargeting drug for UC treatment.
Collapse
Affiliation(s)
- Yajuan Ni
- Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Mengyang Liu
- Tianjin State Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Haiyang Yu
- Tianjin State Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Yue Chen
- Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Yanxia Liu
- Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Tianjin University of Traditional Chinese Medicine, Ministry of Education, Tianjin, China
| | - Suyile Chen
- Alxa League Mongolian Medical Hospital, Bayanhaote, China
| | - Jingya Ruan
- Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Tianjin University of Traditional Chinese Medicine, Ministry of Education, Tianjin, China
| | | | - Yi Zhang
- Tianjin State Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Tao Wang
- Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| |
Collapse
|
16
|
Efficacy of Terpenoid in Attenuating Aortic Atherosclerosis in Apolipoprotein-E Deficient Mice: A Meta-Analysis of Animal Studies. BIOMED RESEARCH INTERNATIONAL 2019; 2019:2931831. [PMID: 31392210 PMCID: PMC6662500 DOI: 10.1155/2019/2931831] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/29/2019] [Revised: 06/10/2019] [Accepted: 06/17/2019] [Indexed: 12/09/2022]
Abstract
Background The apolipoprotein E knockout (ApoE -/-) mouse model is well established for the study of terpenoids in the prevention of atherosclerosis. Studies investigating the clinical benefit of terpenoids in humans are scarce. This systematic review and meta-analysis evaluated the effects of terpenoid administration on atherosclerotic lesion area in ApoE -/- mice. Methods A comprehensive literature search using PubMed, Embase, and the Cochrane Library databases was performed to identify studies that assessed the effects of terpenoids on atherosclerosis in ApoE -/- mice. The primary outcome was atherosclerotic lesion area, and study quality was estimated using SYRCLE's risk of bias tool. Results The meta-analysis included 25 studies. Overall, terpenoids significantly reduced atherosclerotic lesion area when compared to vehicle control (P<0.00001; SMD: -0.55; 95% CI: -0.72, -0.39). In terpenoid type and dose subgroup analyses, sesquiterpenoid (P=0.002; SMD -0.93; 95% CI: -1.52, -0.34), diterpenoid (P=0.01; SMD: -0.30; 95% CI: -0.54, -0.06), triterpenoid (P<0.00001; SMD: -0.66; 95% CI: -0.94, -0.39), tetraterpenoid (P<0.0001; SMD: -1.81; 95% CI: -2.70, -0.91), low dose (P=0.0001; SMD: -0.51; 95% CI: -0.76, -0.25), medium dose (P<0.0001; SMD: -0.48; 95% CI: -0.72, -0.24), and high dose (P=0.002; SMD: -1.07; 95% CI: -1.74, -0.40) significantly decreased atherosclerotic lesion area when compared to vehicle control. PROSPERO register number is CRD42019121176. Conclusion Sesquiterpenoid, diterpenoid, triterpenoid, and tetraterpenoid have potential as antiatherosclerotic agents with a wide range of doses. This systematic review provides a reference for research programs aimed at the development of terpenoid-based clinical drugs.
Collapse
|
17
|
Zhang Y, Li M, Li X, Zhang T, Qin M, Ren L. Isoquinoline Alkaloids and Indole Alkaloids Attenuate Aortic Atherosclerosis in Apolipoprotein E Deficient Mice: A Systematic Review and Meta-Analysis. Front Pharmacol 2018; 9:602. [PMID: 29922166 PMCID: PMC5996168 DOI: 10.3389/fphar.2018.00602] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2018] [Accepted: 05/21/2018] [Indexed: 12/19/2022] Open
Abstract
Background: Several studies have attempted to relate the bioactive alkaloid with atherosclerotic cardiovascular diseases prevention in animal models, providing inconsistent results. Moreover, the direct anti-atherosclerotic effects of alkaloid have hardly been studied in patients. Therefore, the aim of this systematic review was to assess the reported effects of alkaloids on aortic atherosclerosis in ApoE−/− mouse models. Methods: Pubmed and Embase were searched to identify studies which estimated the effect of isolated alkaloids on atherosclerosis in apolipoprotein E deficient mice. Study quality was assessed using SYRCLE's risk of bias tool. We conducted a meta-analysis across 14 studies using a random-effect model to determine the overall effect of the alkaloids, and performed subgroup analyses to compare the effects of the isoquinolone alkaloids and indole alkaloids. Results: The quality of the included studies was low in the majority of included studies. We clarified that alkaloid administration was significantly associated with reduced aortic atherosclerotic lesion area (SMD −3.19, 95% CI −3.88, −2.51). It is important to remark that the experimental characteristics of studies were quite diverse, and the methodological variability could also contribute to heterogeneity. Subgroup analyses suggested that the isoquinoline alkaloids (SMD −4.19, 95% CI −5.18, −3.20), and the indole alkaloids (SMD −2.73, 95% CI −3.56, −1.90) obviously decreased atherosclerotic burden. Conclusion: Isoquinoline alkaloids and indole alkaloids appear to have a direct anti-atherosclerotic effect in ApoE−/− mice. Besides the limitations of animal modal studies, this systematic review could provide an important reference for future preclinical animal trials of good quality and clinical development.
Collapse
Affiliation(s)
- Yibing Zhang
- Department of Pharmacology and Toxicology, School of Pharmaceutical Sciences, Jilin University, Changchun, China.,Department of Ophthalmology, First Hospital of Jilin University, Changchun, China
| | - Min Li
- Department of Pharmacology and Toxicology, School of Pharmaceutical Sciences, Jilin University, Changchun, China
| | - Xiangjun Li
- Department of Pharmacology and Toxicology, School of Pharmaceutical Sciences, Jilin University, Changchun, China
| | - Tong Zhang
- Department of Pharmacology and Toxicology, School of Pharmaceutical Sciences, Jilin University, Changchun, China
| | - Meng Qin
- Department of Pharmacology and Toxicology, School of Pharmaceutical Sciences, Jilin University, Changchun, China
| | - Liqun Ren
- Department of Pharmacology and Toxicology, School of Pharmaceutical Sciences, Jilin University, Changchun, China
| |
Collapse
|
18
|
Szpak D, Izem L, Verbovetskiy D, Soloviev DA, Yakubenko VP, Pluskota E. α Mβ 2 Is Antiatherogenic in Female but Not Male Mice. THE JOURNAL OF IMMUNOLOGY 2018; 200:2426-2438. [PMID: 29459405 DOI: 10.4049/jimmunol.1700313] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/27/2017] [Accepted: 01/24/2018] [Indexed: 11/19/2022]
Abstract
Atherosclerosis is a complex inflammatory process characterized by monocyte recruitment into the arterial wall, their differentiation into macrophages, and lipid accumulation. Because integrin αMβ2 (CD11b/CD18) mediates multiple diverse functions of leukocytes, we examined its role in atherogenesis. αM-/-/ApoE-/- and ApoE-/- mice were fed a control or high fat diet for 3 or 16 wk to induce atherogenesis. Unexpectedly, αM deficiency accelerated development of atherosclerosis in female but not in male mice. The size of aortic root lesions was 3-4.5-fold larger in female αM-/-/ApoE-/- than in ApoE-/- mice. Monocyte and macrophage content within the lesions was increased 2.5-fold in female αM-/-/ApoE-/- mice due to enhanced proliferation. αMβ2 elimination promoted gender-dependent foam cell formation due to enhanced uptake of cholesterol by αM-/-/ApoE-/- macrophages. This difference was attributed to enhanced expression of lipid uptake receptors, CD36 and scavenger receptor A1 (SR-A1), in female mice. Macrophages from female αM-/-/ApoE-/- mice showed dramatically reduced expression of FoxM1 transcription factor and estrogen receptors (ER) α and β. As their antagonists inhibited the effect of 17β-estradiol (E2), E2 decreased CD36, SR-A1, and foam cell formation in ApoE-/- macrophages in an ERα- and ERβ-dependent manner. However, female αM-/-/ApoE-/- macrophages failed to respond to E2 and maintained elevated CD36, SR-A1, and lipid accumulation. FoxM1 inhibition in ApoE-/- macrophages reduced ERs and enhanced CD36 and SR-A1 expression, whereas FoxM1 overexpression in αM-/-/ApoE-/- macrophages reversed their proatherogenic phenotype. We demonstrate a new, surprising atheroprotective role of αMβ2 in female ApoE-/- mice. αMβ2 maintains ER expression in macrophages and E2-dependent inhibition of foam cell formation.
Collapse
Affiliation(s)
- Dorota Szpak
- Department of Molecular Cardiology, Cleveland Clinic, Cleveland, OH 44195
| | - Lahoucine Izem
- Department of Molecular and Cellular Medicine, Cleveland Clinic, Cleveland, OH 44195; and
| | | | - Dmitry A Soloviev
- Department of Molecular Cardiology, Cleveland Clinic, Cleveland, OH 44195
| | - Valentin P Yakubenko
- Department of Biomedical Sciences, Quillen College of Medicine, Johnson City, TN 37614
| | - Elzbieta Pluskota
- Department of Molecular Cardiology, Cleveland Clinic, Cleveland, OH 44195;
| |
Collapse
|
19
|
Kawakami R, Nozato Y, Nakagami H, Ikeda Y, Shimamura M, Yoshida S, Sun J, Kawano T, Takami Y, Noma T, Rakugi H, Minamino T, Morishita R. Development of vaccine for dyslipidemia targeted to a proprotein convertase subtilisin/kexin type 9 (PCSK9) epitope in mice. PLoS One 2018; 13:e0191895. [PMID: 29438441 PMCID: PMC5811007 DOI: 10.1371/journal.pone.0191895] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2017] [Accepted: 01/12/2018] [Indexed: 01/07/2023] Open
Abstract
Proprotein convertase subtilisin/kexin type 9 (PCSK9) regulates expression of low-density lipoprotein (LDL) receptors via receptor internalization and subsequent lysosomal degradation. Thus, an anti-PCSK9 antibody is well known as an anti-hyperlipidemia drug. Here, we aimed to develop vaccine for a long-term treatment of dyslipidemia targeted to PCSK9. In This study, we designed a peptide vaccine for mouse PCSK-9, which consisted of short peptides conjugated to keyhole limpet hemocyanin (KLH) as a carrier protein. Vaccines were administered to male apolipoprotein E (ApoE) deficient mice with adjuvants and significantly elicited an antibody response against PCSK9. The PCSK9 vaccines were administered to mice three times in 2-week intervals, and antibody titers and lipoprotein levels were evaluated up to 24 weeks after the first immunization to determine the therapeutic effect. Anti-PCSK9 antibody titers reached peak levels 6 weeks after the first immunization, and theses titers were maintained for up to 24 weeks. Decreased plasma levels of total cholesterol, very low-density lipoprotein (VLDL), and chylomicron (CM) were maintained for up to 24 weeks. Immunized mice exhibited a significant increase in cell-surface LDL receptor expression. Stimulation with KLH, but not PCSK9, induced the production of INF-gamma and interleukin-4 (IL-4), as determined with ELISPOT assays, thus indicating that PCSK9 vaccine did not elicit T-cell activation in our vaccine system. The present anti-PCSK9 vaccine induced long-lasting anti-PCSK9 antibody production and improved lipoprotein profiles. Thus, anti-PCSK9 vaccine could become a new option for the treatment of dyslipidemia as a long-acting therapy in future.
Collapse
Affiliation(s)
- Ryo Kawakami
- Department of Cardiorenal and Cerebrovascular Medicine, Faculty of Medicine, Kagawa University, Kagawa, Japan.,Department of Clinical Gene Therapy, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Yoichi Nozato
- Department of Geriatric and General Medicine, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Hironori Nakagami
- Department of Health Development and Medicine, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Yuka Ikeda
- Department of Clinical Gene Therapy, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Munehisa Shimamura
- Department of Health Development and Medicine, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Shota Yoshida
- Department of Health Development and Medicine, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Jiao Sun
- Department of Health Development and Medicine, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Tomohiro Kawano
- Department of Neurology, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Yoichi Takami
- Department of Geriatric and General Medicine, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Takahisa Noma
- Department of Cardiorenal and Cerebrovascular Medicine, Faculty of Medicine, Kagawa University, Kagawa, Japan
| | - Hiromi Rakugi
- Department of Geriatric and General Medicine, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Tetsuo Minamino
- Department of Cardiorenal and Cerebrovascular Medicine, Faculty of Medicine, Kagawa University, Kagawa, Japan
| | - Ryuichi Morishita
- Department of Clinical Gene Therapy, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| |
Collapse
|
20
|
Sfyri P, Matsakas A. Crossroads between peripheral atherosclerosis, western-type diet and skeletal muscle pathophysiology: emphasis on apolipoprotein E deficiency and peripheral arterial disease. J Biomed Sci 2017; 24:42. [PMID: 28688452 PMCID: PMC5502081 DOI: 10.1186/s12929-017-0346-8] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2017] [Accepted: 06/07/2017] [Indexed: 12/16/2022] Open
Abstract
Atherosclerosis is a chronic inflammatory process that, in the presence of hyperlipidaemia, promotes the formation of atheromatous plaques in large vessels of the cardiovascular system. It also affects peripheral arteries with major implications for a number of other non-vascular tissues such as the skeletal muscle, the liver and the kidney. The aim of this review is to critically discuss and assimilate current knowledge on the impact of peripheral atherosclerosis and its implications on skeletal muscle homeostasis. Accumulating data suggests that manifestations of peripheral atherosclerosis in skeletal muscle originates in a combination of increased i)-oxidative stress, ii)-inflammation, iii)-mitochondrial deficits, iv)-altered myofibre morphology and fibrosis, v)-chronic ischemia followed by impaired oxygen supply, vi)-reduced capillary density, vii)- proteolysis and viii)-apoptosis. These structural, biochemical and pathophysiological alterations impact on skeletal muscle metabolic and physiologic homeostasis and its capacity to generate force, which further affects the individual's quality of life. Particular emphasis is given on two major areas representing basic and applied science respectively: a)-the abundant evidence from a well-recognised atherogenic model; the Apolipoprotein E deficient mouse and the role of a western-type diet and b)-on skeletal myopathy and oxidative stress-induced myofibre damage from human studies on peripheral arterial disease. A significant source of reactive oxygen species production and oxidative stress in cardiovascular disease is the family of NADPH oxidases that contribute to several pathologies. Finally, strategies targeting NADPH oxidases in skeletal muscle in an attempt to attenuate cellular oxidative stress are highlighted, providing a better understanding of the crossroads between peripheral atherosclerosis and skeletal muscle pathophysiology.
Collapse
Affiliation(s)
- Peggy Sfyri
- Molecular Physiology Laboratory, Centre for Atherothrombotic & Metabolic Disease, Hull York Medical School, University of Hull, Cottingham Road, Hull, HU6 7RX, United Kingdom
| | - Antonios Matsakas
- Molecular Physiology Laboratory, Centre for Atherothrombotic & Metabolic Disease, Hull York Medical School, University of Hull, Cottingham Road, Hull, HU6 7RX, United Kingdom.
| |
Collapse
|
21
|
Sarduy R, Brito V, Castillo A, Soto Y, Griñán T, Marleau S, Vázquez AM. Dose-Dependent Induction of an Idiotypic Cascade by Anti-Glycosaminoglycan Monoclonal Antibody in apoE -/- Mice: Association with Atheroprotection. Front Immunol 2017; 8:232. [PMID: 28316603 PMCID: PMC5334371 DOI: 10.3389/fimmu.2017.00232] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2016] [Accepted: 02/17/2017] [Indexed: 01/07/2023] Open
Abstract
Atherosclerosis, the underlying pathology of most cardiovascular diseases, is triggered by the retention of apolipoprotein B (apoB)-containing lipoproteins in the arterial wall through electrostatic interactions with glycosaminoglycan (GAG) side chains of proteoglycans. Previously, we reported the antiatherogenic properties of the chimeric monoclonal antibody (mAb) chP3R99-LALA, which binds sulfated GAGs, inhibits low-density lipoprotein (LDL)–chondroitin sulfate (CS) association, and abrogates LDL oxidation and foam cell formation. In preventive and therapeutic settings, apoE-deficient (apoE−/−) mice immunized with 50 μg of this mAb showed reduced atherosclerotic lesions related with the induction of autologous anti-GAG antibodies. Knowing that age and sex are major non-modifiable risk factors in the development of atherosclerosis, the present study aimed to assess the influence of these variables on the capacity of chP3R99-LALA mAb to generate an anti-CS antibody response. Also, we aimed at defining the impact of the dose of chP3R99-LALA on the anti-CS antibody induction and the atheroprotective effect of this mAb in apoE−/− mice. Neither age nor sex had an impact in the IgG anti-CS antibody response induced by s.c. immunization with this mAb. Moreover, chP3R99-LALA mAb reduced atherosclerotic lesions to a similar extent in both young male and female apoE−/− mice fed a hypercholesterolemic diet and, in middle-aged female apoE−/− mice, with spontaneous lesions. On the other hand, increasing the dose of chP3R99-LALA (200 vs. 50 μg) elicited an anti-idiotype antibody cascade characterized by higher levels of anti-idiotype (Ab2), anti-anti-idiotype (Ab3), and anti-CS antibody responses. Moreover, this dose increment resulted in a striking reduction of aortic atherosclerotic lesions in immunized mice.
Collapse
Affiliation(s)
- Roger Sarduy
- Division of Immunobiology, Center of Molecular Immunology , Havana , Cuba
| | - Victor Brito
- Division of Immunobiology, Center of Molecular Immunology , Havana , Cuba
| | - Adriana Castillo
- Division of Immunobiology, Center of Molecular Immunology , Havana , Cuba
| | - Yosdel Soto
- Division of Immunobiology, Center of Molecular Immunology , Havana , Cuba
| | - Tania Griñán
- Division of Immunobiology, Center of Molecular Immunology , Havana , Cuba
| | - Sylvie Marleau
- Faculté de Pharmacie, Université de Montréal , Montréal, QC , Canada
| | - Ana María Vázquez
- Innovation Managing Direction, Center of Molecular Immunology , Havana , Cuba
| |
Collapse
|