1
|
Bulnes JF, González L, Velásquez L, Orellana MP, Venturelli PM, Martínez G. Role of inflammation and evidence for the use of colchicine in patients with acute coronary syndrome. Front Cardiovasc Med 2024; 11:1356023. [PMID: 38993522 PMCID: PMC11236697 DOI: 10.3389/fcvm.2024.1356023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Accepted: 05/29/2024] [Indexed: 07/13/2024] Open
Abstract
Acute Coronary Syndrome (ACS) significantly contributes to cardiovascular death worldwide. ACS may arise from the disruption of an atherosclerotic plaque, ultimately leading to acute ischemia and myocardial infarction. In the pathogenesis of atherosclerosis, inflammation assumes a pivotal role, not solely in the initiation and complications of atherosclerotic plaque formation, but also in the myocardial response to ischemic insult. Acute inflammatory processes, coupled with time to reperfusion, orchestrate ischemic and reperfusion injuries, dictating infarct magnitude and acute left ventricular (LV) remodeling. Conversely, chronic inflammation, alongside neurohumoral activation, governs persistent LV remodeling. The interplay between chronic LV remodeling and recurrent ischemic episodes delineates the progression of the disease toward heart failure and cardiovascular death. Colchicine exerts anti-inflammatory properties affecting both the myocardium and atherosclerotic plaque by modulating the activity of monocyte/macrophages, neutrophils, and platelets. This modulation can potentially result in a more favorable LV remodeling and forestalls the recurrence of ACS. This narrative review aims to delineate the role of inflammation across the different phases of ACS pathophysiology and describe the mechanistic underpinnings of colchicine, exploring its purported role in modulating each of these stages.
Collapse
Affiliation(s)
- Juan Francisco Bulnes
- División de Enfermedades Cardiovasculares, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Leticia González
- Centro de Imágenes Biomédicas, Departamento de Radiología, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Leonardo Velásquez
- División de Enfermedades Cardiovasculares, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - María Paz Orellana
- División de Enfermedades Cardiovasculares, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Paula Muñoz Venturelli
- Centro de Estudios Clínicos, Instituto de Ciencias e Innovación en Medicina (ICIM), Facultad de Medicina Clínica Alemana, Universidad del Desarrollo, Santiago, Chile
| | - Gonzalo Martínez
- División de Enfermedades Cardiovasculares, Pontificia Universidad Católica de Chile, Santiago, Chile
- Heart Research Institute, Sydney, NSW, Australia
| |
Collapse
|
2
|
Zuo X, Ding X, Zhang Y, Kang YJ. Reversal of atherosclerosis by restoration of vascular copper homeostasis. Exp Biol Med (Maywood) 2024; 249:10185. [PMID: 38978540 PMCID: PMC11228934 DOI: 10.3389/ebm.2024.10185] [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: 04/03/2024] [Accepted: 06/04/2024] [Indexed: 07/10/2024] Open
Abstract
Atherosclerosis has traditionally been considered as a disorder characterized by the accumulation of cholesterol and thrombotic materials within the arterial wall. However, it is now understood to be a complex inflammatory disease involving multiple factors. Central to the pathogenesis of atherosclerosis are the interactions among monocytes, macrophages, and neutrophils, which play pivotal roles in the initiation, progression, and destabilization of atherosclerotic lesions. Recent advances in our understanding of atherosclerosis pathogenesis, coupled with results obtained from experimental interventions, lead us to propose the hypothesis that atherosclerosis may be reversible. This paper outlines the evolution of this hypothesis and presents corroborating evidence that supports the potential for atherosclerosis regression through the restoration of vascular copper homeostasis. We posit that these insights may pave the way for innovative therapeutic approaches aimed at the reversal of atherosclerosis.
Collapse
Affiliation(s)
- Xiao Zuo
- Tasly Stem Cell Biology Laboratory, Tasly Biopharmaceutical Co., Tianjin, China
| | - Xueqin Ding
- Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Yaya Zhang
- Tasly Stem Cell Biology Laboratory, Tasly Biopharmaceutical Co., Tianjin, China
| | - Y James Kang
- Tasly Stem Cell Biology Laboratory, Tasly Biopharmaceutical Co., Tianjin, China
- Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|
3
|
Zapolski T, Kornecki W, Jaroszyński A. The Influence of Balneotherapy Using Salty Sulfide-Hydrogen Sulfide Water on Selected Markers of the Cardiovascular System: A Prospective Study. J Clin Med 2024; 13:3526. [PMID: 38930055 PMCID: PMC11204439 DOI: 10.3390/jcm13123526] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Revised: 06/10/2024] [Accepted: 06/11/2024] [Indexed: 06/28/2024] Open
Abstract
Background: The sulfide-hydrogen sulfide brine balneotherapy (HSBB), including a combination of dissolved hydrogen sulfide (H2S) gas, inorganic sulfur ions (S2-), and hydrosulfide ions (HS-), is one of the most important and most effective forms of spa treatment in patients with osteoarticular disorders (OADs). Some cardiovascular diseases (CVDs) are often considered to be contraindications to HSBB since the presence of thiol groups may lead to an increased quantity of reactive oxygen species (ROS), which damage the vascular endothelium, and endothelial dysfunction is considered to be the main cause of atherosclerosis. However, there are a number of literature reports suggesting this theory to be false. H2S is a member of the endogenous gaseous transmitter family and, since it is a relatively recent addition, it has the least well-known biological properties. H2S-NO interactions play an important role in oxidative stress in CVDs. The general objective of this study was to assess the cardiovascular safety of HSBB and analyze the effect of HSBB on selected cardiovascular risk markers. Methods: A total of 100 patients at the age of 76.3 (±7.5) years from the Włókniarz Sanatorium in Busko-Zdrój were initially included in the study. The following parameters were assessed: age, sex, height, body weight, body surface area (BSA), body mass index (BMI), systolic (SBP) and diastolic blood pressure (DBP), heart rate, the diagnosis of OAD that was the indication for balneotherapy, creatinine (CREAT), glomerular filtration rate (GFR), lipid panel, C-reactive protein (CRP), uric acid (UA), and fibrinogen (FIBR) and cardiovascular markers: (cardiac troponin T (cTnT), N-terminal pro-B-type natriuretic peptide (NT-proBNP). Results: A significant decrease in DBP and a trend towards SBP reduction were observed over the course of the study. A significant decrease was observed in CRP levels decreasing from 2.7 (±3.6) mg/L to 2.06 (±1.91) mg/L, whereas FIBR rose significantly from 2.95 (±0.59) g/L to 3.23 (±1.23) g/L. LDL-C levels decreased slightly, statistically significant, from 129.36 (±40.67) mg/dL to 123.74 (±36.14) mg/dL. HSBB did not affect the levels of evaluated cardiovascular biomarkers, namely NT-proBNP (137.41 (±176.52) pg/mL vs. 142.89 (±182.82) pg/mL; p = 0.477) and cTnT (9.64 (±4.13) vs. 9.65 (±3.91) ng/L; p = 0.948). A multiple regression analysis of pre-balneotherapy and post-balneotherapy values showed cTnT levels to be independently correlated only with CREAT levels and GFR values. None of the assessed parameters independently correlated with the NT-proBNP level. Conclusions: HSBB resulted in a statistically significant improvement in a subclinical pro-inflammatory state. HSBB has a beneficial effect in modifying key cardiovascular risk factors by reducing LDL-C levels and DBP values. HSBB has a neutral effect on cardiovascular ischemia/injury. Despite slightly elevated baseline levels of the biochemical marker of HF (NT-proBNP), HSBB causes no further increase in this marker. The use of HSBB in patients with OAD has either a neutral effect or a potentially beneficial effect on the cardiovascular system, which may constitute grounds for further studies to verify the current cardiovascular contraindications for this form of therapy.
Collapse
Affiliation(s)
- Tomasz Zapolski
- Department of Cardiology, Medical University of Lublin, 20-093 Lublin, Poland
| | | | - Andrzej Jaroszyński
- Department of Internal Medicine and Family Medicine, Collegium Medicum, Jan Kochanowski University in Kielce, 25-369 Kielce, Poland;
| |
Collapse
|
4
|
Stasinopoulou M, Kostomitsopoulos N, Kadoglou NPE. The Anti-Atherosclerotic Effects of Endothelin Receptor Antagonist, Bosentan, in Combination with Atorvastatin-An Experimental Study. Int J Mol Sci 2024; 25:6614. [PMID: 38928320 PMCID: PMC11203450 DOI: 10.3390/ijms25126614] [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: 05/13/2024] [Revised: 06/12/2024] [Accepted: 06/13/2024] [Indexed: 06/28/2024] Open
Abstract
Bosentan, an endothelin receptor antagonist (ERA), has potential anti-atherosclerotic properties. We investigated the complementary effects of bosentan and atorvastatin on the progression and composition of the atherosclerotic lesions in diabetic mice. Forty-eight male ApoE-/- mice were fed high-fat diet (HFD) for 14 weeks. At week 8, diabetes was induced with streptozotocin, and mice were randomized into four groups: (1) control/COG: no intervention; (2) ΒOG: bosentan 100 mg/kg/day per os; (3) ATG: atorvastatin 20 mg/kg/day per os; and (4) BO + ATG: combined administration of bosentan and atorvastatin. The intra-plaque contents of collagen, elastin, monocyte chemoattractant protein-1 (MCP-1), tumor necrosis factor-a (TNF-a), matrix metalloproteinases (MMP-2, -3, -9), and TIMP-1 were determined. The percentage of lumen stenosis was significantly lower across all treated groups: BOG: 19.5 ± 2.2%, ATG: 12.8 ± 4.8%, and BO + ATG: 9.1 ± 2.7% compared to controls (24.6 ± 4.8%, p < 0.001). The administration of both atorvastatin and bosentan resulted in significantly higher collagen content and thicker fibrous cap versus COG (p < 0.01). All intervention groups showed lower relative intra-plaque concentrations of MCP-1, MMP-3, and MMP-9 and a higher TIMP-1concentration compared to COG (p < 0.001). Importantly, latter parameters presented lower levels when bosentan was combined with atorvastatin compared to COG (p < 0.05). Bosentan treatment in diabetic, atherosclerotic ApoE-/- mice delayed the atherosclerosis progression and enhanced plaques' stability, showing modest but additive effects with atorvastatin, which are promising in atherosclerotic cardiovascular diseases.
Collapse
Affiliation(s)
- Marianna Stasinopoulou
- Center of Clinical, Experimental Surgery, and Translational Research, Biomedical Research Foundation, Academy of Athens, 115 27 Athens, Greece; (M.S.); (N.K.)
| | - Nikolaos Kostomitsopoulos
- Center of Clinical, Experimental Surgery, and Translational Research, Biomedical Research Foundation, Academy of Athens, 115 27 Athens, Greece; (M.S.); (N.K.)
| | | |
Collapse
|
5
|
Zhen J, Li X, Yu H, Du B. High-density lipoprotein mimetic nano-therapeutics targeting monocytes and macrophages for improved cardiovascular care: a comprehensive review. J Nanobiotechnology 2024; 22:263. [PMID: 38760755 PMCID: PMC11100215 DOI: 10.1186/s12951-024-02529-x] [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: 12/12/2023] [Accepted: 05/03/2024] [Indexed: 05/19/2024] Open
Abstract
The prevalence of cardiovascular diseases continues to be a challenge for global health, necessitating innovative solutions. The potential of high-density lipoprotein (HDL) mimetic nanotherapeutics in the context of cardiovascular disease and the intricate mechanisms underlying the interactions between monocyte-derived cells and HDL mimetic showing their impact on inflammation, cellular lipid metabolism, and the progression of atherosclerotic plaque. Preclinical studies have demonstrated that HDL mimetic nanotherapeutics can regulate monocyte recruitment and macrophage polarization towards an anti-inflammatory phenotype, suggesting their potential to impede the progression of atherosclerosis. The challenges and opportunities associated with the clinical application of HDL mimetic nanotherapeutics, emphasize the need for additional research to gain a better understanding of the precise molecular pathways and long-term effects of these nanotherapeutics on monocytes and macrophages to maximize their therapeutic efficacy. Furthermore, the use of nanotechnology in the treatment of cardiovascular diseases highlights the potential of nanoparticles for targeted treatments. Moreover, the concept of theranostics combines therapy and diagnosis to create a selective platform for the conversion of traditional therapeutic medications into specialized and customized treatments. The multifaceted contributions of HDL to cardiovascular and metabolic health via highlight its potential to improve plaque stability and avert atherosclerosis-related problems. There is a need for further research to maximize the therapeutic efficacy of HDL mimetic nanotherapeutics and to develop targeted treatment approaches to prevent atherosclerosis. This review provides a comprehensive overview of the potential of nanotherapeutics in the treatment of cardiovascular diseases, emphasizing the need for innovative solutions to address the challenges posed by cardiovascular diseases.
Collapse
Affiliation(s)
- Juan Zhen
- The First Hospital of Jilin University, Changchun, 130021, China
| | - Xiangjun Li
- School of Pharmaceutical Science, Jilin University, Changchun, 130021, China
| | - Haitao Yu
- The First Hospital of Jilin University, Changchun, 130021, China
| | - Bing Du
- The First Hospital of Jilin University, Changchun, 130021, China.
| |
Collapse
|
6
|
Chatterjee N, Komaravolu RK, Durant CP, Wu R, McSkimming C, Drago F, Kumar S, Valentin-Guillama G, Miller YI, McNamara CA, Ley K, Taylor A, Alimadadi A, Hedrick CC. Single Cell High Dimensional Analysis of Human Peripheral Blood Mononuclear Cells Reveals Unique Intermediate Monocyte Subsets Associated with Sex Differences in Coronary Artery Disease. Int J Mol Sci 2024; 25:2894. [PMID: 38474140 PMCID: PMC10932111 DOI: 10.3390/ijms25052894] [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: 01/17/2024] [Revised: 02/23/2024] [Accepted: 02/25/2024] [Indexed: 03/14/2024] Open
Abstract
Monocytes are associated with human cardiovascular disease progression. Monocytes are segregated into three major subsets: classical (cMo), intermediate (iMo), and nonclassical (nMo). Recent studies have identified heterogeneity within each of these main monocyte classes, yet the extent to which these subsets contribute to heart disease progression is not known. Peripheral blood mononuclear cells (PBMC) were obtained from 61 human subjects within the Coronary Assessment of Virginia (CAVA) Cohort. Coronary atherosclerosis severity was quantified using the Gensini Score (GS). We employed high-dimensional single-cell transcriptome and protein methods to define how human monocytes differ in subjects with low to severe coronary artery disease. We analyzed 487 immune-related genes and 49 surface proteins at the single-cell level using Antibody-Seq (Ab-Seq). We identified six subsets of myeloid cells (cMo, iMo, nMo, plasmacytoid DC, classical DC, and DC3) at the single-cell level based on surface proteins, and we associated these subsets with coronary artery disease (CAD) incidence based on Gensini score (GS) in each subject. Only frequencies of iMo were associated with high CAD (GS > 32), adj.p = 0.024. Spearman correlation analysis with GS from each subject revealed a positive correlation with iMo frequencies (r = 0.314, p = 0.014) and further showed a robust sex-dependent positive correlation in female subjects (r = 0.663, p = 0.004). cMo frequencies did not correlate with CAD severity. Key gene pathways differed in iMo among low and high CAD subjects and between males and females. Further single-cell analysis of iMo revealed three iMo subsets in human PBMC, distinguished by the expression of HLA-DR, CXCR3, and CD206. We found that the frequency of immunoregulatory iMo_HLA-DR+CXCR3+CD206+ was associated with CAD severity (adj.p = 0.006). The immunoregulatory iMo subset positively correlated with GS in both females (r = 0.660, p = 0.004) and males (r = 0.315, p = 0.037). Cell interaction analyses identified strong interactions of iMo with CD4+ effector/memory T cells and Tregs from the same subjects. This study shows the importance of iMo in CAD progression and suggests that iMo may have important functional roles in modulating CAD risk, particularly among females.
Collapse
Affiliation(s)
- Nandini Chatterjee
- La Jolla Institute of Immunology, La Jolla, CA 92037, USA; (N.C.); (K.L.)
| | - Ravi K. Komaravolu
- Department of Medicine, Immunology Center of Georgia, Augusta University, Augusta, GA 30912, USA; (R.K.K.)
| | | | - Runpei Wu
- La Jolla Institute of Immunology, La Jolla, CA 92037, USA; (N.C.); (K.L.)
| | - Chantel McSkimming
- Beirne Carter Immunology Center, University of Virginia, Charlottesville, VA 22904, USA (A.T.)
| | - Fabrizio Drago
- Beirne Carter Immunology Center, University of Virginia, Charlottesville, VA 22904, USA (A.T.)
| | - Sunil Kumar
- Department of Medicine, Immunology Center of Georgia, Augusta University, Augusta, GA 30912, USA; (R.K.K.)
| | - Gabriel Valentin-Guillama
- Department of Medicine, Immunology Center of Georgia, Augusta University, Augusta, GA 30912, USA; (R.K.K.)
| | - Yury I. Miller
- Division of Endocrinology, University of California San Diego, La Jolla, CA 92093, USA
| | - Coleen A. McNamara
- Beirne Carter Immunology Center, University of Virginia, Charlottesville, VA 22904, USA (A.T.)
| | - Klaus Ley
- La Jolla Institute of Immunology, La Jolla, CA 92037, USA; (N.C.); (K.L.)
- Department of Medicine, Immunology Center of Georgia, Augusta University, Augusta, GA 30912, USA; (R.K.K.)
| | - Angela Taylor
- Beirne Carter Immunology Center, University of Virginia, Charlottesville, VA 22904, USA (A.T.)
| | - Ahmad Alimadadi
- La Jolla Institute of Immunology, La Jolla, CA 92037, USA; (N.C.); (K.L.)
- Department of Medicine, Immunology Center of Georgia, Augusta University, Augusta, GA 30912, USA; (R.K.K.)
| | - Catherine C. Hedrick
- La Jolla Institute of Immunology, La Jolla, CA 92037, USA; (N.C.); (K.L.)
- Department of Medicine, Immunology Center of Georgia, Augusta University, Augusta, GA 30912, USA; (R.K.K.)
| |
Collapse
|
7
|
Janssen H, Koekkoek LL, Swirski FK. Effects of lifestyle factors on leukocytes in cardiovascular health and disease. Nat Rev Cardiol 2024; 21:157-169. [PMID: 37752350 DOI: 10.1038/s41569-023-00931-w] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 09/01/2023] [Indexed: 09/28/2023]
Abstract
Exercise, stress, sleep and diet are four distinct but intertwined lifestyle factors that influence the cardiovascular system. Abundant epidemiological, clinical and preclinical studies have underscored the importance of managing stress, having good sleep hygiene and responsible eating habits and exercising regularly. We are born with a genetic blueprint that can protect us against or predispose us to a particular disease. However, lifestyle factors build upon and profoundly influence those predispositions. Studies in the past 10 years have shown that the immune system in general and leukocytes in particular are particularly susceptible to environmental perturbations. Lifestyle factors such as stress, sleep, diet and exercise affect leukocyte behaviour and function and thus the immune system at large. In this Review, we explore the various mechanisms by which lifestyle factors modulate haematopoiesis and leukocyte migration and function in the context of cardiovascular health. We pay particular attention to the role of the nervous system as the key executor that connects environmental influences to leukocyte behaviour.
Collapse
Affiliation(s)
- Henrike Janssen
- Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Laura L Koekkoek
- Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Filip K Swirski
- Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- The Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| |
Collapse
|
8
|
Yennemadi AS, Jordan N, Diong S, Keane J, Leisching G. The Link Between Dysregulated Immunometabolism and Vascular Damage: Implications for the Development of Atherosclerosis in Systemic Lupus Erythematosus and Other Rheumatic Diseases. J Rheumatol 2024; 51:234-241. [PMID: 38224981 DOI: 10.3899/jrheum.2023-0833] [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] [Accepted: 11/30/2023] [Indexed: 01/17/2024]
Abstract
A bimodal pattern of mortality in systemic lupus erythematosus (SLE) exists. Early-stage deaths are predominantly caused by infection, whereas later-stage deaths are mainly caused by atherosclerotic disease. Further, although SLE-related mortality has reduced considerably in recent years, cardiovascular (CV) events remain one of the leading causes of death in people with SLE. Accelerated atherosclerosis in SLE is attributed to both an increase in traditional CV risk factors and the inflammatory effects of SLE itself. Many of these changes occur within the microenvironment of the vascular-immune interface, the site of atherosclerotic plaque development. Here, an intimate interaction between endothelial cells, vascular smooth muscle cells, and immune cells dictates physiological vs pathological responses to a chronic type 1 interferon environment. Low-density neutrophils (LDNs) have also been implicated in eliciting vasculature-damaging effects at such lesion sites. These changes are thought to be governed by dysfunctional metabolism of immune cells in this niche due at least in part to the chronic induction of type 1 interferons. Understanding these novel pathophysiological mechanisms and metabolic pathways may unveil potential innovative pharmacological targets and therapeutic opportunities for atherosclerosis, as well as shed light on the development of premature atherosclerosis in patients with SLE who develop CV events.
Collapse
Affiliation(s)
- Anjali S Yennemadi
- A.S. Yennemadi, MSc, J. Keane, MD, G. Leisching, PhD, TB Immunology Group, Department of Clinical Medicine, Trinity Translational Medicine Institute, School of Medicine, Trinity College Dublin, University of Dublin
| | - Natasha Jordan
- N. Jordan, PhD, Department of Rheumatology, St. James's Hospital
| | - Sophie Diong
- S. Diong, MD, Department of Dermatology, St. James's Hospital, Dublin, Ireland
| | - Joseph Keane
- A.S. Yennemadi, MSc, J. Keane, MD, G. Leisching, PhD, TB Immunology Group, Department of Clinical Medicine, Trinity Translational Medicine Institute, School of Medicine, Trinity College Dublin, University of Dublin
| | - Gina Leisching
- A.S. Yennemadi, MSc, J. Keane, MD, G. Leisching, PhD, TB Immunology Group, Department of Clinical Medicine, Trinity Translational Medicine Institute, School of Medicine, Trinity College Dublin, University of Dublin;
| |
Collapse
|
9
|
Wang Q, Wang T, Lio C, Yu X, Chen X, Liu L, Wu Y, Huang H, Qing L, Luo P. Surface hydrolysis-designed AuNPs-zwitterionic-glucose as a novel tool for targeting macrophage visualization and delivery into infarcted hearts. J Control Release 2023; 356:678-690. [PMID: 36898530 DOI: 10.1016/j.jconrel.2023.03.008] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2022] [Revised: 02/27/2023] [Accepted: 03/05/2023] [Indexed: 03/12/2023]
Abstract
Macrophages, innate immune cells, are key players in the maintenance of myocardial homeostasis under normal conditions and tissue repair after injury. The infiltration of macrophages into the injured heart makes them a potentially appealing vehicle for noninvasive imaging and targeted drug delivery of myocardial infarction (MI). In this study, we demonstrated the use of surface hydrolysis-designed AuNPs-zwitterionic-glucose to label macrophages and track their infiltration into isoproterenol hydrochloride (ISO)-induced MI sites noninvasively using CT. The AuNPs-zwitterionic-glucose did not affect the viability or cytokine release of macrophages and were highly taken up by these cells. The in vivo CT images were obtained on Day 4, Day 6, Day 7, and Day 9, and the attenuation was seen to increase in the heart over time compared to the Day 4 scan. In vitro analysis also confirmed the presence of macrophages around injured cardiomyocytes. Additionally, we also addressed the concern of cell tracking or merely AuNP tracking, which is the inherent problem for any form of nanoparticle-labeled cell tracking by using zwitterionic and glucose-functionalized AuNPs. The glucose coated on the surface of AuNPs-zwit-glucose will be hydrolyzed in macrophages, forming only zwitterionic protected AuNPs that cannot be taken up again by endogenous cells in vivo. This will greatly improve the accuracy and precision of imaging and target delivery. We believe this is the first study to noninvasively visualize the infiltration of macrophages into MI hearts using CT, which could be used for imaging and evaluating the possibility of macrophage-mediated delivery in infarcted hearts.
Collapse
Affiliation(s)
- Qianlong Wang
- State Key Laboratories for Quality Research in Chinese Medicines, Macau University of Science and Technology, Macau 999078, China; Chengdu Institute of Biology, Chinese Academy of Sciences, Chengdu 610041, China; School of Materials Science and Engineering, Tsinghua University, Beijing 100084, China
| | - Tiantian Wang
- State Key Laboratories for Quality Research in Chinese Medicines, Macau University of Science and Technology, Macau 999078, China; Chengdu Institute of Biology, Chinese Academy of Sciences, Chengdu 610041, China
| | - Chonkit Lio
- State Key Laboratories for Quality Research in Chinese Medicines, Macau University of Science and Technology, Macau 999078, China
| | - Xina Yu
- State Key Laboratories for Quality Research in Chinese Medicines, Macau University of Science and Technology, Macau 999078, China
| | - Xiaoyi Chen
- State Key Laboratories for Quality Research in Chinese Medicines, Macau University of Science and Technology, Macau 999078, China
| | - Lancong Liu
- State Key Laboratories for Quality Research in Chinese Medicines, Macau University of Science and Technology, Macau 999078, China
| | - Youjiao Wu
- State Key Laboratories for Quality Research in Chinese Medicines, Macau University of Science and Technology, Macau 999078, China
| | - Hui Huang
- Department of Cardiology, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen 518000, China
| | - Linsen Qing
- Chengdu Institute of Biology, Chinese Academy of Sciences, Chengdu 610041, China.
| | - Pei Luo
- State Key Laboratories for Quality Research in Chinese Medicines, Macau University of Science and Technology, Macau 999078, China.
| |
Collapse
|
10
|
Sonawane AR, Aikawa E, Aikawa M. Connections for Matters of the Heart: Network Medicine in Cardiovascular Diseases. Front Cardiovasc Med 2022; 9:873582. [PMID: 35665246 PMCID: PMC9160390 DOI: 10.3389/fcvm.2022.873582] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Accepted: 04/19/2022] [Indexed: 01/18/2023] Open
Abstract
Cardiovascular diseases (CVD) are diverse disorders affecting the heart and vasculature in millions of people worldwide. Like other fields, CVD research has benefitted from the deluge of multiomics biomedical data. Current CVD research focuses on disease etiologies and mechanisms, identifying disease biomarkers, developing appropriate therapies and drugs, and stratifying patients into correct disease endotypes. Systems biology offers an alternative to traditional reductionist approaches and provides impetus for a comprehensive outlook toward diseases. As a focus area, network medicine specifically aids the translational aspect of in silico research. This review discusses the approach of network medicine and its application to CVD research.
Collapse
Affiliation(s)
- Abhijeet Rajendra Sonawane
- Center for Interdisciplinary Cardiovascular Sciences, Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, United States
- Center for Excellence in Vascular Biology, Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, United States
| | - Elena Aikawa
- Center for Interdisciplinary Cardiovascular Sciences, Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, United States
- Center for Excellence in Vascular Biology, Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, United States
| | - Masanori Aikawa
- Center for Interdisciplinary Cardiovascular Sciences, Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, United States
- Center for Excellence in Vascular Biology, Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, United States
| |
Collapse
|
11
|
Su C, Han Y, Qu B, Zhang C, Liang T, Gao F, Hou G. CD93 in macrophages: A novel target for atherosclerotic plaque imaging? J Cell Mol Med 2022; 26:2152-2162. [PMID: 35166040 PMCID: PMC8995462 DOI: 10.1111/jcmm.17237] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Revised: 01/29/2022] [Accepted: 02/02/2022] [Indexed: 12/14/2022] Open
Abstract
Noninvasive imaging atherosclerotic (AS) plaque is of great importance for early diagnosis. Recently, CD93 in MΦ was linked to atherosclerosis development. Herein, we have investigated whether CD93 in MΦ is a potential novel target for atherosclerotic plaque imaging. CD93hi and CD93lo MΦ were prepared with or without LPS stimulation, before biological activity was evaluated. A rat AS model was produced with left carotid artery clamped. Whole‐body/ex vivo phosphor autoradiography of the artery and biodistribution were investigated after incorporation of 3H‐2‐DG into CD93hi and CD93lo MΦ or after 125I‐α‐CD93 (125I‐anti‐CD93mAb) injection. The plaque tissue was subjected to CD93/CD68 immunofluorescence/immunohistochemistry staining. CD93hi and CD93lo MΦ cells were successfully prepared without significant effect on bioactivity after incorporative labelled with 3H‐2‐DG. The AS model was successfully established. Biodistribution studies showed that adoptive transfer of 3H‐2‐DG‐CD93hi MΦ or 125I‐ α‐CD93 injection resulted in accumulation of radioactivity within the atherosclerotic plaque in the clamped left carotid artery. T/NT (target/non‐target, left/right carotid artery) ratio was higher in the 3H‐2‐DG‐CD93hi MΦ adoptive transfer group than in the 3H‐2‐DG‐CD93lo MΦ group (p < .05). Plaque radioactivity in the 125I‐α‐CD93 injection group was significantly higher than in the 125I‐IgG control group (p < .01). The higher radioactivity accumulated in the clamped left carotid artery was confirmed by phosphor autoradiography. More importantly, CD93/CD68 double‐positive MΦ accumulated at the atherosclerotic plaque in 3H‐2‐DG‐CD93hi MΦ adoptive transfer group, which correlated with plaque radioactivity (r = .99, p < .01). In summary, both adoptive‐transferred 3H‐2‐DG‐labelled CD93hi MΦ and 125I‐α‐CD93 injection specifically targeted CD93 in atherosclerotic plaque. CD93 is a potential target in atherosclerotic plaque imaging.
Collapse
Affiliation(s)
- Chen Su
- Key Laboratory for Experimental Teratology of the Ministry of Education and Research Center for Experimental Nuclear Medicine, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Yeming Han
- Radiology Department, Qilu Hospital of Shandong University, Jinan, China
| | - Bin Qu
- Key Laboratory for Experimental Teratology of the Ministry of Education and Research Center for Experimental Nuclear Medicine, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Chao Zhang
- Key Laboratory for Experimental Teratology of the Ministry of Education and Research Center for Experimental Nuclear Medicine, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Ting Liang
- Key Laboratory for Experimental Teratology of the Ministry of Education and Research Center for Experimental Nuclear Medicine, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Feng Gao
- Key Laboratory for Experimental Teratology of the Ministry of Education and Research Center for Experimental Nuclear Medicine, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Guihua Hou
- Key Laboratory for Experimental Teratology of the Ministry of Education and Research Center for Experimental Nuclear Medicine, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| |
Collapse
|
12
|
P2X 4 deficiency reduces atherosclerosis and plaque inflammation in mice. Sci Rep 2022; 12:2801. [PMID: 35181718 PMCID: PMC8857235 DOI: 10.1038/s41598-022-06706-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Accepted: 01/19/2022] [Indexed: 12/25/2022] Open
Abstract
Extracellular adenosine-5′-triphosphate (ATP) acts as an import signaling molecule mediating inflammation via purinergic P2 receptors. ATP binds to the purinergic receptor P2X4 and promotes inflammation via increased expression of pro-inflammatory cytokines. Because of the central role of inflammation, we assumed a functional contribution of the ATP-P2X4-axis in atherosclerosis. Expression of P2X4 was increased in atherosclerotic aortic arches from low-density lipoprotein receptor-deficient mice being fed a high cholesterol diet as assessed by real-time polymerase chain reaction and immunohistochemistry. To investigate the functional role of P2X4 in atherosclerosis, P2X4-deficient mice were crossed with low-density lipoprotein receptor-deficient mice and fed high cholesterol diet. After 16 weeks, P2X4-deficient mice developed smaller atherosclerotic lesions compared to P2X4-competent mice. Furthermore, intravital microscopy showed reduced ATP-induced leukocyte rolling at the vessel wall in P2X4-deficient mice. Mechanistically, we found a reduced RNA expression of CC chemokine ligand 2 (CCL-2), C-X-C motif chemokine-1 (CXCL-1), C-X-C motif chemokine-2 (CXCL-2), Interleukin-6 (IL-6) and tumor necrosis factor α (TNFα) as well as a decreased nucleotide-binding oligomerization domain-like receptor protein 3 (NLRP3)-inflammasome priming in atherosclerotic plaques from P2X4-deficient mice. Moreover, bone marrow derived macrophages isolated from P2X4-deficient mice revealed a reduced ATP-mediated release of CCL-2, CC chemokine ligand 5 (CCL-5), Interleukin-1β (IL-1β) and IL-6. Additionally, P2X4-deficient mice shared a lower proportion of pro-inflammatory Ly6Chigh monocytes and a higher proportion of anti-inflammatory Ly6Clow monocytes, and expressend less endothelial VCAM-1. Finally, increased P2X4 expression in human atherosclerotic lesions from carotid endarterectomy was found, indicating the importance of potential implementations of this study’s findings for human atherosclerosis. Collectively, P2X4 deficiency reduced experimental atherosclerosis, plaque inflammation and inflammasome priming, pointing to P2X4 as a potential therapeutic target in the fight against atherosclerosis.
Collapse
|
13
|
Lipoprotein(a), Immune Cells and Cardiovascular Outcomes in Patients with Premature Coronary Heart Disease. J Pers Med 2022; 12:jpm12020269. [PMID: 35207757 PMCID: PMC8876319 DOI: 10.3390/jpm12020269] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Revised: 02/08/2022] [Accepted: 02/10/2022] [Indexed: 11/16/2022] Open
Abstract
The detection of lipoprotein(a) [Lp(a)] in the artery wall at the stage of lipid-bands formation may indicate that it participates in the atherosclerosis local nonspecific inflammatory process. Innate immune cells are involved in atherogenesis, with monocytes playing a major role in the initiation of atherosclerosis, while neutrophils can contribute to plaque destabilization. This work studies the relationship between Lp(a), immune blood cells and major adverse cardiovascular events (MACE) in patients with the early manifestation of coronary heart disease (CHD). The study included 200 patients with chronic CHD, manifested up to the age of 55 in men and 60 in women. An increased Lp(a) concentration [hyperLp(a)] was shown to predict cardiovascular events in patients with premature CHD with long-term follow-up. According to the logistic regression analysis results, an increase in the monocyte count with OR = 4.58 (95% CI 1.04–20.06) or lymphocyte-to-monocyte ratio with OR = 0.82 (0.68–0.99), (p < 0.05 for both) was associated with MACE in patients with early CHD, regardless of gender, age, classical risk factors, atherogenic lipoproteins concentration and statin intake. The combination of an increased monocyte count and hyperLp(a) significantly increased the proportion of patients with early CHD with subsequent development of MACE (p = 0.02, ptrend = 0.003). The odds of cardiovascular events in patients with early CHD manifestation were highest in patients with an elevated lymphocyte-to-monocyte ratio and an elevated Lp(a) level. A higher neutrophil blood count and an elevated neutrophil-to-lymphocyte ratio determined the faster development of MACE in patients with a high Lp(a) concentration. The data obtained in this study suggest that the high atherothrombogenicity of Lp(a) is associated with the “inflammatory” component and the innate immune cells involvement in this process. Thus, the easily calculated immunological ratios of blood cells and Lp(a) concentrations can be considered simple predictors of future cardiovascular events.
Collapse
|
14
|
Tritz R, Hudson FZ, Harris V, Ghoshal P, Singla B, Lin H, Csanyi G, Stansfield BK. MEK inhibition exerts temporal and myeloid cell-specific effects in the pathogenesis of neurofibromatosis type 1 arteriopathy. Sci Rep 2021; 11:24345. [PMID: 34934133 PMCID: PMC8692602 DOI: 10.1038/s41598-021-03750-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Accepted: 12/09/2021] [Indexed: 11/18/2022] Open
Abstract
Mutations in the NF1 tumor suppressor gene are linked to arteriopathy. Nf1 heterozygosity (Nf1+/–) results in robust neointima formation, similar to humans, and myeloid-restricted Nf1+/– recapitulates this phenotype via MEK-ERK activation. Here we define the contribution of myeloid subpopulations to NF1 arteriopathy. Neutrophils from WT and Nf1+/– mice were functionally assessed in the presence of MEK and farnesylation inhibitors in vitro and neutrophil recruitment to lipopolysaccharide was assessed in WT and Nf1+/– mice. Littermate 12–15 week-old male wildtype and Nf1+/– mice were subjected to carotid artery ligation and provided either a neutrophil depleting antibody (1A8), liposomal clodronate to deplete monocytes/macrophages, or PD0325901 and neointima size was assessed 28 days after injury. Bone marrow transplant experiments assessed monocyte/macrophage mobilization during neointima formation. Nf1+/– neutrophils exhibit enhanced proliferation, migration, and adhesion via p21Ras activation of MEK in vitro and in vivo. Neutrophil depletion suppresses circulating Ly6Clow monocytes and enhances neointima size, while monocyte/macrophage depletion and deletion of CCR2 in bone marrow cells abolish neointima formation in Nf1+/– mice. Taken together, these findings suggest that neurofibromin-MEK-ERK activation in circulating neutrophils and monocytes during arterial remodeling is nuanced and points to important cross-talk between these populations in the pathogenesis of NF1 arteriopathy.
Collapse
Affiliation(s)
- Rebekah Tritz
- Vascular Biology Center, Augusta University, Augusta, GA, USA
| | - Farlyn Z Hudson
- Vascular Biology Center, Augusta University, Augusta, GA, USA
| | - Valerie Harris
- Vascular Biology Center, Augusta University, Augusta, GA, USA
| | | | - Bhupesh Singla
- Vascular Biology Center, Augusta University, Augusta, GA, USA
| | - Huiping Lin
- Vascular Biology Center, Augusta University, Augusta, GA, USA
| | - Gabor Csanyi
- Vascular Biology Center, Augusta University, Augusta, GA, USA.,Department of Pharmacology and Toxicology, Augusta University, Augusta, GA, USA
| | - Brian K Stansfield
- Vascular Biology Center, Augusta University, Augusta, GA, USA. .,Division of Neonatology, Department of Pediatrics, Medical College of Georgia at Augusta University, Augusta University, 1120 15th St, BIW6033, Augusta, GA, 30912, USA.
| |
Collapse
|
15
|
The Association of Lipoprotein(a) and Circulating Monocyte Subsets with Severe Coronary Atherosclerosis. J Cardiovasc Dev Dis 2021; 8:jcdd8060063. [PMID: 34206012 PMCID: PMC8228191 DOI: 10.3390/jcdd8060063] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Revised: 05/21/2021] [Accepted: 05/25/2021] [Indexed: 01/02/2023] Open
Abstract
Background and aims: Chronic inflammation associated with the uncontrolled activation of innate and acquired immunity plays a fundamental role in all stages of atherogenesis. Monocytes are a heterogeneous population and each subset contributes differently to the inflammatory process. A high level of lipoprotein(a) (Lp(a)) is a proven cardiovascular risk factor. The aim of the study was to investigate the association between the increased concentration of Lp(a) and monocyte subpopulations in patients with a different severity of coronary atherosclerosis. Methods: 150 patients (124 males) with a median age of 60 years undergoing a coronary angiography were enrolled. Lipids, Lp(a), autoantibodies, blood cell counts and monocyte subpopulations (classical, intermediate, non-classical) were analyzed. Results: The patients were divided into two groups depending on the Lp(a) concentration: normal Lp(a) < 30 mg/dL (n = 82) and hyperLp(a) ≥ 30 mg/dL (n = 68). Patients of both groups were comparable by risk factors, autoantibody levels and blood cell counts. In patients with hyperlipoproteinemia(a) the content (absolute and relative) of non-classical monocytes was higher (71.0 (56.6; 105.7) vs. 62.2 (45.7; 82.4) 103/mL and 17.7 (13.0; 23.3) vs. 15.1 (11.4; 19.4) %, respectively, p < 0.05). The association of the relative content of non-classical monocytes with the Lp(a) concentration retained a statistical significance when adjusted for gender and age (r = 0.18, p = 0.03). The severity of coronary atherosclerosis was associated with the Lp(a) concentration as well as the relative and absolute (p < 0.05) content of classical monocytes. The high content of non-classical monocytes (OR = 3.5, 95% CI 1.2–10.8) as well as intermediate monocytes (OR = 8.7, 2.5–30.6) in patients with hyperlipoproteinemia(a) were associated with triple-vessel coronary disease compared with patients with a normal Lp(a) level and a low content of monocytes. Conclusion: Hyperlipoproteinemia(a) and a decreased quantity of classical monocytes were associated with the severity of coronary atherosclerosis. The expansion of CD16+ monocytes (intermediate and non-classical) in the presence of hyperlipoproteinemia(a) significantly increased the risk of triple-vessel coronary disease.
Collapse
|
16
|
Weng R, Liu S, Gu X, Zhong Z. Characterization of the B cell receptor repertoire of patients with acute coronary syndrome. Genes Genomics 2021; 44:19-28. [PMID: 33974240 DOI: 10.1007/s13258-021-01110-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2020] [Accepted: 04/29/2021] [Indexed: 01/01/2023]
Abstract
BACKGROUND Acute coronary syndrome (ACS) is a complex cardiovascular disease whose development involves the dysregulation of adaptive immune responses. Though it has been proven that T cells associate with inflammation in the development of ACS, the function of B cells in disease remains unclear. OBJECTIVE The aim of this study was to reveal the diversity of the B cell receptor (BCR) repertoire of patients with ACS. METHODS We conducted a pilot study to sequence the immune repertoire of peripheral blood mononuclear cells (PBMCs) from patients with ACS, including acute myocardial infarction (AMI) and unstable angina (UA), and quantitatively characterized BCR repertoires by bioinformatics analysis. RESULTS We found that patients with AMI and UA had lower BCR repertoire diversity compared with controls with normal coronary arteries (NCA). Lower percentages of productive unique BCR nt sequences and higher percentages of top 200 unique BCR sequences were identified in AMI and UA patients than NCA controls. Patients had various preferential usage of V and J genes from B cell clones in accordance with the disease severity of coronary arteries. AMI patients had distinct CDR3 amino acids, and their frequency differed among patients with ACS. CONCLUSIONS Our results indicate that differential BCR signatures represent an imprint of distinct repertoires among ACS patients. This study thereby opens up the prospect of studying disease-relevant B cells to better understand and treat ACS.
Collapse
Affiliation(s)
- Ruiqiang Weng
- Center for Precision Medicine, Meizhou People's Hospital (Huangtang Hospital), Meizhou Hospital Affiliated to Sun Yat-sen University, No. 63 Huangtang Road, Meijiang District, Meizhou, 514031, People's Republic of China
- Guangdong Provincial Engineering and Technological Research Center for Molecular Diagnostics of Cardiovascular Diseases, Meizhou, 514031, People's Republic of China
- Guangdong Provincial Key Laboratory of Precision Medicine and Clinical Translational Research of Hakka Population, Meizhou, 514031, People's Republic of China
| | - Sudong Liu
- Center for Precision Medicine, Meizhou People's Hospital (Huangtang Hospital), Meizhou Hospital Affiliated to Sun Yat-sen University, No. 63 Huangtang Road, Meijiang District, Meizhou, 514031, People's Republic of China
- Guangdong Provincial Engineering and Technological Research Center for Molecular Diagnostics of Cardiovascular Diseases, Meizhou, 514031, People's Republic of China
- Guangdong Provincial Key Laboratory of Precision Medicine and Clinical Translational Research of Hakka Population, Meizhou, 514031, People's Republic of China
| | - Xiaodong Gu
- Center for Precision Medicine, Meizhou People's Hospital (Huangtang Hospital), Meizhou Hospital Affiliated to Sun Yat-sen University, No. 63 Huangtang Road, Meijiang District, Meizhou, 514031, People's Republic of China
- Guangdong Provincial Engineering and Technological Research Center for Molecular Diagnostics of Cardiovascular Diseases, Meizhou, 514031, People's Republic of China
- Guangdong Provincial Key Laboratory of Precision Medicine and Clinical Translational Research of Hakka Population, Meizhou, 514031, People's Republic of China
| | - Zhixiong Zhong
- Center for Precision Medicine, Meizhou People's Hospital (Huangtang Hospital), Meizhou Hospital Affiliated to Sun Yat-sen University, No. 63 Huangtang Road, Meijiang District, Meizhou, 514031, People's Republic of China.
- Guangdong Provincial Engineering and Technological Research Center for Molecular Diagnostics of Cardiovascular Diseases, Meizhou, 514031, People's Republic of China.
- Center for Cardiovascular Diseases, Meizhou People's Hospital (Huangtang Hospital), Meizhou Hospital Affiliated to Sun Yat-sen University, Meizhou, 514031, People's Republic of China.
| |
Collapse
|
17
|
Makowski LM, Leffers M, Waltenberger J, Pardali E. Transforming growth factor-β1 signalling triggers vascular endothelial growth factor resistance and monocyte dysfunction in type 2 diabetes mellitus. J Cell Mol Med 2021; 25:5316-5325. [PMID: 33942489 PMCID: PMC8178271 DOI: 10.1111/jcmm.16543] [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: 11/23/2020] [Revised: 03/07/2021] [Accepted: 03/24/2021] [Indexed: 12/01/2022] Open
Abstract
Type 2 diabetes mellitus (T2DM) leads to monocyte dysfunction associated with atherogenesis and defective arteriogenesis. Transforming growth factor (TGF)-β1, placenta growth factor (PlGF)-1 and vascular endothelial growth factor (VEGF)A play important roles in atherogenesis and arteriogenesis. VEGF-receptor (VEGFR)-mediated monocyte migration is inhibited in T2DM (VEGFA resistance), while TGF-β1-induced monocyte migration is fully functional. Therefore, we hypothesize that TGF-β antagonises the VEGFA responses in human monocytes. We demonstrate that monocytes from T2DM patients have an increased migratory response towards low concentrations of TGF-β1, while PlGF-1/VEGFA responses are mitigated. Mechanistically, this is due to increased expression of type II TGF-β receptor in monocytes under high-glucose conditions and increased expression of soluble (s)VEGFR1, which is known to interfere with VEGFA signalling. VEGFA resistance in monocytes from T2DM patients can be rescued by either experimental down-regulation of TGF-β receptor expression in vitro or by functional blocking of TGF-β signalling using either a TGF-β receptor kinase inhibitor or a TGF-β neutralizing antibody. Our data demonstrate that both T2DM and high-glucose potentiate the TGF-β pathway. TGF-β signalling impairs VEGFR-mediated responses in T2DM monocytes and in this way contributes to mononuclear cell dysfunction, provide novel insights into T2DM vascular dysfunction.
Collapse
Affiliation(s)
- Lena-Maria Makowski
- Department of Cardiology I-Coronary and Peripheral Vascular Disease, Heart Failure, University Hospital Münster, Cardiolology, Münster, Germany
| | - Merle Leffers
- Department of Cardiology I-Coronary and Peripheral Vascular Disease, Heart Failure, University Hospital Münster, Cardiolology, Münster, Germany
| | - Johannes Waltenberger
- Department of Cardiology I-Coronary and Peripheral Vascular Disease, Heart Failure, University Hospital Münster, Cardiolology, Münster, Germany.,Cells-in-Motion Cluster of Excellence (EXC 1003-CiM), University of Münster, Münster, Germany.,Department of Cardiovascular Medicine, Medical Faculty, University of Münster, Münster, Germany
| | - Evangelia Pardali
- Department of Cardiology I-Coronary and Peripheral Vascular Disease, Heart Failure, University Hospital Münster, Cardiolology, Münster, Germany.,Cells-in-Motion Cluster of Excellence (EXC 1003-CiM), University of Münster, Münster, Germany
| |
Collapse
|
18
|
Patel VK, Williams H, Li SCH, Fletcher JP, Medbury HJ. Monocyte Subset Recruitment Marker Profile Is Inversely Associated With Blood ApoA1 Levels. Front Immunol 2021; 12:616305. [PMID: 33717107 PMCID: PMC7952433 DOI: 10.3389/fimmu.2021.616305] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Accepted: 01/12/2021] [Indexed: 12/30/2022] Open
Abstract
Dyslipidemia promotes development of the atherosclerotic plaques that characterise cardiovascular disease. Plaque progression requires the influx of monocytes into the vessel wall, but whether dyslipidemia is associated with an increased potential of monocytes to extravasate is largely unknown. Here (using flow cytometry) we examined recruitment marker expression on monocytes from generally healthy individuals who differed in lipid profile. Comparisons were made between monocyte subsets, participants and relative to participants’ lipid levels. Monocyte subsets differed significantly in their expression of recruitment markers, with highest expression being on either the classical or non-classical subsets. However, these inter-subset differences were largely overshadowed by considerable inter-participant differences with some participants having higher levels of recruitment markers on all three monocyte subsets. Furthermore, when the expression of one recruitment marker was high, so too was that of most of the other markers, with substantial correlations evident between the markers. The inter-participant differences were explained by lipid levels. Most notably, there was a significant inverse correlation for most markers with ApoA1 levels. Our results indicate that dyslipidemia, in particular low levels of ApoA1, is associated with an increased potential of all monocyte subsets to extravasate, and to do so using a wider repertoire of recruitment markers than currently appreciated.
Collapse
Affiliation(s)
- Vyoma K Patel
- Vascular Biology Research Centre, Department of Surgery, Westmead Hospital, Westmead, NSW, Australia.,The University of Sydney, Westmead Clinical School, Westmead, NSW, Australia
| | - Helen Williams
- Vascular Biology Research Centre, Department of Surgery, Westmead Hospital, Westmead, NSW, Australia.,The University of Sydney, Westmead Clinical School, Westmead, NSW, Australia
| | - Stephen C H Li
- Western Sydney University, Blacktown/Mt Druitt Clinical School, Blacktown Hospital, Blacktown, NSW, Australia.,Institute of Clinical Pathology and Medical Research, Westmead Hospital, Westmead, NSW, Australia
| | - John P Fletcher
- Vascular Biology Research Centre, Department of Surgery, Westmead Hospital, Westmead, NSW, Australia.,The University of Sydney, Westmead Clinical School, Westmead, NSW, Australia
| | - Heather J Medbury
- Vascular Biology Research Centre, Department of Surgery, Westmead Hospital, Westmead, NSW, Australia.,The University of Sydney, Westmead Clinical School, Westmead, NSW, Australia
| |
Collapse
|
19
|
Tang D, Geng F, Yu C, Zhang R. Recent Application of Zebrafish Models in Atherosclerosis Research. Front Cell Dev Biol 2021; 9:643697. [PMID: 33718384 PMCID: PMC7947229 DOI: 10.3389/fcell.2021.643697] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Accepted: 02/05/2021] [Indexed: 12/13/2022] Open
Abstract
Atherosclerotic cardiovascular disease is one of the leading causes of death worldwide. Establishing animal models of atherosclerosis is of great benefit for studying its complicated pathogenesis and screening and evaluating related drugs. Although researchers have generated a variety of models for atherosclerosis study in rabbits, mice and rats, the limitations of these models make it difficult to monitor the development of atherosclerosis, and these models are unsuitable for large scale screening of potential therapeutic targets. On the contrast, zebrafish can fulfill these purposes thanks to their fecundity, rapid development ex utero, embryonic transparency, and conserved lipid metabolism process. Thus, zebrafish have become a popular alternative animal model for atherosclerosis research. In this mini review, we summarize different zebrafish models used to study atherosclerosis, focusing on the latest applications of these models to the dynamic monitoring of atherosclerosis progression, mechanistic study of therapeutic intervention and drug screening, and assessment of the impacts of other risk factors.
Collapse
Affiliation(s)
- Dandan Tang
- School of Basic Medical Sciences, Wuhan University, Wuhan, China
| | - Fang Geng
- School of Life Sciences, Fudan University, Shanghai, China
| | - Chunxiao Yu
- School of Basic Medical Sciences, Wuhan University, Wuhan, China
| | - Ruilin Zhang
- School of Basic Medical Sciences, Wuhan University, Wuhan, China
| |
Collapse
|
20
|
Yahara Y, Ma X, Gracia L, Alman BA. Monocyte/Macrophage Lineage Cells From Fetal Erythromyeloid Progenitors Orchestrate Bone Remodeling and Repair. Front Cell Dev Biol 2021; 9:622035. [PMID: 33614650 PMCID: PMC7889961 DOI: 10.3389/fcell.2021.622035] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Accepted: 01/12/2021] [Indexed: 12/21/2022] Open
Abstract
A third of the population sustains a bone fracture, and the pace of fracture healing slows with age. The slower pace of repair is responsible for the increased morbidity in older individuals who sustain a fracture. Bone healing progresses through overlapping phases, initiated by cells of the monocyte/macrophage lineage. The repair process ends with remodeling. This last phase is controlled by osteoclasts, which are bone-specific multinucleated cells also of the monocyte/macrophage lineage. The slower rate of healing in aging can be rejuvenated by macrophages from young animals, and secreted proteins from macrophage regulate undifferentiated mesenchymal cells to become bone-forming osteoblasts. Macrophages can derive from fetal erythromyeloid progenitors or from adult hematopoietic progenitors. Recent studies show that fetal erythromyeloid progenitors are responsible for the osteoclasts that form the space in bone for hematopoiesis and the fetal osteoclast precursors reside in the spleen postnatally, traveling through the blood to participate in fracture repair. Differences in secreted proteins between macrophages from old and young animals regulate the efficiency of osteoblast differentiation from undifferentiated mesenchymal precursor cells. Interestingly, during the remodeling phase osteoclasts can form from the fusion between monocyte/macrophage lineage cells from the fetal and postnatal precursor populations. Data from single cell RNA sequencing identifies specific markers for populations derived from the different precursor populations, a finding that can be used in future studies. Here, we review the diversity of macrophages and osteoclasts, and discuss recent finding about their developmental origin and functions, which provides novel insights into their roles in bone homeostasis and repair.
Collapse
Affiliation(s)
- Yasuhito Yahara
- Department of Orthopaedic Surgery, Duke University School of Medicine, Durham, NC, United States.,Department of Orthopaedic Surgery, Faculty of Medicine, University of Toyama, Toyama, Japan.,Department of Molecular and Medical Pharmacology, Faculty of Medicine, University of Toyama, Toyama, Japan
| | - Xinyi Ma
- Department of Orthopaedic Surgery, Duke University School of Medicine, Durham, NC, United States.,Department of Cell Biology, Duke University School of Medicine, Durham, NC, United States
| | - Liam Gracia
- Department of Orthopaedic Surgery, Duke University School of Medicine, Durham, NC, United States.,Department of Cell Biology, Duke University School of Medicine, Durham, NC, United States
| | - Benjamin A Alman
- Department of Orthopaedic Surgery, Duke University School of Medicine, Durham, NC, United States.,Department of Cell Biology, Duke University School of Medicine, Durham, NC, United States
| |
Collapse
|
21
|
Natural killer cells, gamma delta T cells and classical monocytes are associated with systolic blood pressure in the multi-ethnic study of atherosclerosis (MESA). BMC Cardiovasc Disord 2021; 21:45. [PMID: 33482725 PMCID: PMC7821496 DOI: 10.1186/s12872-021-01857-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2020] [Accepted: 01/06/2021] [Indexed: 12/12/2022] Open
Abstract
Background Hypertension is a major source of cardiovascular morbidity and mortality. Recent evidence from mouse models, genetic, and cross-sectional human studies suggest increased proportions of selected immune cell subsets may be associated with levels of systolic blood pressure (SBP).
Methods We assayed immune cells from cryopreserved samples collected at the baseline examination (2000–2002) from 1195 participants from the multi-ethnic study of atherosclerosis (MESA). We used linear mixed models, with adjustment for age, sex, race/ethnicity, smoking, exercise, body mass index, education, diabetes, and cytomegalovirus titers, to estimate the associations between 30 immune cell subsets (4 of which were a priori hypotheses) and repeated measures of SBP (baseline and up to four follow-up measures) over 10 years. The analysis provides estimates of the association with blood pressure level. Results The mean age of the MESA participants at baseline was 64 ± 10 years and 53% were male. A one standard deviation (1-SD) increment in the proportion of γδ T cells was associated with 2.40 mmHg [95% confidence interval (CI) 1.34–3.42] higher average systolic blood pressure; and for natural killer cells, a 1-SD increment was associated with 1.88 mmHg (95% CI 0.82–2.94) higher average level of systolic blood pressure. A 1-SD increment in classical monocytes (CD14++CD16−) was associated with 2.01 mmHG (95% CI 0.79–3.24) lower average systolic blood pressure. There were no associations of CD4+ T helper cell subsets with average systolic blood pressure. Conclusion These findings suggest that the innate immune system plays a role in levels of SBP whereas there were no associations with adaptive immune cells.
Collapse
|
22
|
Zhao R, Wan P, Shariati-Ievari S, Aliani M, Shen GX. North American Wild Rice-Attenuated Hyperglycemia in High-Fat-Induced Obese Mice: Involvement of AMP-Activated Protein Kinase. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2020; 68:8855-8862. [PMID: 32689799 DOI: 10.1021/acs.jafc.0c03776] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Previous studies indicated that North American wild rice (WIR) reduced atherosclerosis and vascular inflammation in low-density lipoprotein receptor knockout mice. The effects of WIR on hyperglycemia in diabetic animal models have not been documented. The present study aims to determine the impact of WIR on glucose metabolism in high-fat (HF)-induced diabetic mice and a key modulator. Male C57 BL/J6 mice were treated with a control diet and a HF diet supplemented with 26% (weight/weight, a substitute for carbohydrates in the diet) of WIR or white rice (WHR) (n = 8/group) for 11 weeks. HF + WHR diet significantly increased fasting plasma glucose, cholesterol, triglycerides, insulin, insulin resistance, monocyte adhesion, and the levels of relevant inflammatory mediators (tumor necrotic factor-α, plasminogen activator inhibitor-1, and monocyte chemotactic protein-1) in mice compared to the control diet (p < 0.01). HF + WIR significantly reduced HF diet-induced metabolic and inflammatory changes compared to the HF + WHR diet (p < 0.01). Metabolomics analysis indicated that an array of metabolites related to glucose metabolism was significantly more abundant in WIR than in WHR, including adenosine 5'-monophosphate (AMP), a potent agonist for AMP-activated protein kinase or AMPK. WIR normalized HF diet-induced reduction in the abundance of phospho-AMPKα in skeletal muscle, liver, and adipose tissue from the mice. The findings for the first time demonstrated that WIR decreased HF diet-induced hyperglycemia in mice compared to WHR. The metabolic benefits of WIR may result, at least in part, from the activation of AMPKα in insulin-sensitive tissue in the mice.
Collapse
Affiliation(s)
- Ruozhi Zhao
- Department of Internal Medicine, University of Manitoba, Winnipeg R3E 3P4, Canada
| | - Peng Wan
- Food and Human Nutritional Sciences, University of Manitoba, Winnipeg R3T 2N2, Canada
- College of Food Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Shiva Shariati-Ievari
- The Canadian Centre for Agri-Food Research in Health and Medicine, Division of Neurodegenerative, St. Boniface Hospital Albrechtsen Research, Winnipeg R2H 0G1, Canada
| | - Michel Aliani
- Food and Human Nutritional Sciences, University of Manitoba, Winnipeg R3T 2N2, Canada
- The Canadian Centre for Agri-Food Research in Health and Medicine, Division of Neurodegenerative, St. Boniface Hospital Albrechtsen Research, Winnipeg R2H 0G1, Canada
| | - Garry X Shen
- Department of Internal Medicine, University of Manitoba, Winnipeg R3E 3P4, Canada
- Food and Human Nutritional Sciences, University of Manitoba, Winnipeg R3T 2N2, Canada
| |
Collapse
|
23
|
Regal-McDonald K, Patel RP. Selective Recruitment of Monocyte Subsets by Endothelial N-Glycans. THE AMERICAN JOURNAL OF PATHOLOGY 2020; 190:947-957. [PMID: 32084367 DOI: 10.1016/j.ajpath.2020.01.006] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/01/2019] [Revised: 12/17/2019] [Accepted: 01/07/2020] [Indexed: 12/26/2022]
Abstract
Monocyte rolling, adhesion, and transmigration across the endothelium are mediated by specific interactions between surface adhesion molecules. This process is fundamental to innate immunity and to inflammatory disease, including atherosclerosis, where monocyte egress into the intimal space is central to formation of fatty plaques. Monocytes are a heterogeneous population of three distinct subsets of cells, all of which play different roles in atherosclerosis progression. However, it is not well understood how interactions between different monocyte subsets and the endothelium are regulated. Furthermore, it is appreciated that endothelial adhesion molecules are heavily N-glycosylated, but beyond regulating protein trafficking to the cell surface, whether and if so how these N-glycans contribute to monocyte recruitment is not known. This review discusses how changes in endothelial N-glycosylation may impact vascular and monocytic inflammation. It will also discuss how regulating N-glycoforms on the endothelial surface may allow for the recruitment of specific monocyte subsets to sites of inflammation, and how further understanding in this area may lead to the development of glyco-specific therapeutics in the treatment of cardiovascular disease.
Collapse
Affiliation(s)
- Kellie Regal-McDonald
- Department of Pathology, University of Alabama at Birmingham, Birmingham, Alabama; Center for Free Radical Biology, University of Alabama at Birmingham, Birmingham, Alabama
| | - Rakesh P Patel
- Department of Pathology, University of Alabama at Birmingham, Birmingham, Alabama; Center for Free Radical Biology, University of Alabama at Birmingham, Birmingham, Alabama.
| |
Collapse
|
24
|
Mossanen JC, Jansen TU, Pracht J, Liepelt A, Buendgens L, Stoppe C, Goetzenich A, Simon TP, Autschbach R, Marx G, Tacke F. Elevated circulating CD14 ++CD16 + intermediate monocytes are independently associated with extracardiac complications after cardiac surgery. Sci Rep 2020; 10:947. [PMID: 31969629 PMCID: PMC6976615 DOI: 10.1038/s41598-020-57700-9] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2019] [Accepted: 12/31/2019] [Indexed: 12/11/2022] Open
Abstract
Elective cardiac surgery has low procedural complications. However, about 40% of patients develop extracardiac complications including delirium and acute kidney injury. We hypothesized that inflammatory processes and immune cell activation might be associated with these complications. We therefore prospectively included 104 patients undergoing cardiac surgery in our study. We assessed peripheral blood leukocyte populations by flow cytometry and circulating cytokines before operation, after surgery and at days one and four post-operatively. Patients undergoing cardiac surgery showed significantly elevated leukocytes and neutrophils after surgery. On the contrary, monocytes decreased after surgery and significantly increased at days 1 and 4, particularly classical (Mon1,CD14++CD16-) and intermediate (Mon2,CD14++CD16+) monocytes. While peripheral leukocyte subsets were unaltered in patients with infectious (n = 15) or cardiac complications (n = 31), post-operative leukocytes (p = 0.0016), neutrophils (p = 0.0061) and Mon2 (p = 0.0007) were clearly raised in patients developing extracardiac complications (n = 35). Using multiple logistic regression analyses, patient's age, ICU days, number of blood transfusions and elevated post-surgery Mon2 independently predicted extracardiac complications. Our findings demonstrate that elevated Mon2 after cardiac surgery are associated with an increased risk for extracardiac complications. These findings might improve the risk estimation after cardiac operations and the role of Mon2 for inflammation in cardiac surgery.
Collapse
Affiliation(s)
- Jana C Mossanen
- Department of Medicine III, University Hospital Aachen, Aachen, Germany.
- Department of Intensive and Intermediate Care, University Hospital Aachen, Aachen, Germany.
| | - Tobias U Jansen
- Department of Medicine III, University Hospital Aachen, Aachen, Germany
| | - Jessica Pracht
- Department of Medicine III, University Hospital Aachen, Aachen, Germany
- Department of Intensive and Intermediate Care, University Hospital Aachen, Aachen, Germany
| | - Anke Liepelt
- Department of Medicine III, University Hospital Aachen, Aachen, Germany
| | - Lukas Buendgens
- Department of Medicine III, University Hospital Aachen, Aachen, Germany
| | - Christian Stoppe
- Department of Intensive and Intermediate Care, University Hospital Aachen, Aachen, Germany
| | - Andreas Goetzenich
- Department of Thoracic and Cardiovascular Surgery, University Hospital Aachen, Aachen, Germany
| | - Tim-Philipp Simon
- Department of Intensive and Intermediate Care, University Hospital Aachen, Aachen, Germany
| | - Rüdiger Autschbach
- Department of Thoracic and Cardiovascular Surgery, University Hospital Aachen, Aachen, Germany
| | - Gernot Marx
- Department of Intensive and Intermediate Care, University Hospital Aachen, Aachen, Germany
| | - Frank Tacke
- Department of Medicine III, University Hospital Aachen, Aachen, Germany
- Department of Hepatology & Gastroenterology, Charité University Medical Center, Berlin, Germany
| |
Collapse
|
25
|
Cansby E, Magnusson E, Nuñez-Durán E, Amrutkar M, Pedrelli M, Parini P, Hoffmann J, Ståhlman M, Howell BW, Marschall HU, Borén J, Mahlapuu M. STK25 Regulates Cardiovascular Disease Progression in a Mouse Model of Hypercholesterolemia. Arterioscler Thromb Vasc Biol 2019; 38:1723-1737. [PMID: 29930001 DOI: 10.1161/atvbaha.118.311241] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Objective- Recent cohort studies have shown that nonalcoholic fatty liver disease (NAFLD), and especially nonalcoholic steatohepatitis (NASH), associate with atherosclerosis and cardiovascular disease, independently of conventional cardiometabolic risk factors. However, the mechanisms underlying the pathophysiological link between NAFLD/NASH and cardiovascular disease still remain unclear. Our previous studies have identified STK25 (serine/threonine protein kinase 25) as a critical determinant in ectopic lipid storage, meta-inflammation, and progression of NAFLD/NASH. The aim of this study was to assess whether STK25 is also one of the mediators in the complex molecular network controlling the cardiovascular disease risk. Approach and Results- Atherosclerosis was induced in Stk25 knockout and transgenic mice, and their wild-type littermates, by gene transfer of gain-of-function mutant of PCSK9 (proprotein convertase subtilisin/kexin type 9), which induces the downregulation of hepatic LDLR (low-density lipoprotein receptor), combined with an atherogenic western-type diet. We found that Stk25-/- mice displayed reduced atherosclerosis lesion area as well as decreased lipid accumulation, macrophage infiltration, collagen formation, and oxidative stress in aortic lesions compared with wild-type littermates, independently from alterations in dyslipidemia. Reciprocally, Stk25 transgenic mice presented aggravated plaque formation and maturation compared with wild-type littermates despite similar levels of fasting plasma cholesterol. We also found that STK25 protein was expressed in all layers of the aorta, suggesting a possible direct role in cardiovascular disease. Conclusions- This study provides the first evidence that STK25 plays a critical role in regulation of cardiovascular disease risk and suggests that pharmacological inhibition of STK25 function may provide new possibilities for prevention/treatment of atherosclerosis.
Collapse
Affiliation(s)
- Emmelie Cansby
- From the Lundberg Laboratory for Diabetes Research (E.C., E.M., E.N.-D., J.H., M.M.)
| | - Elin Magnusson
- From the Lundberg Laboratory for Diabetes Research (E.C., E.M., E.N.-D., J.H., M.M.)
| | - Esther Nuñez-Durán
- From the Lundberg Laboratory for Diabetes Research (E.C., E.M., E.N.-D., J.H., M.M.)
| | - Manoj Amrutkar
- Department of Molecular and Clinical Medicine, Institute of Medicine, University of Gothenburg and Sahlgrenska University Hospital, Sweden; Department of Hepato-Pancreato-Biliary Surgery, Institute of Clinical Medicine, University of Oslo, Norway (M.A.)
| | | | - Paolo Parini
- Department of Laboratory Medicine (M.P., P.P.).,Department of Medicine, Metabolism Unit (P.P.)
| | - Jenny Hoffmann
- From the Lundberg Laboratory for Diabetes Research (E.C., E.M., E.N.-D., J.H., M.M.)
| | | | - Brian W Howell
- Karolinska Institute, Stockholm, Sweden; and Department of Neuroscience and Physiology, State University of New York Upstate Medical University, Syracuse (B.W.H.)
| | | | - Jan Borén
- Wallenberg Laboratory (M.S., H.-U.M., J.B.)
| | - Margit Mahlapuu
- From the Lundberg Laboratory for Diabetes Research (E.C., E.M., E.N.-D., J.H., M.M.)
| |
Collapse
|
26
|
Centa M, Prokopec KE, Garimella MG, Habir K, Hofste L, Stark JM, Dahdah A, Tibbitt CA, Polyzos KA, Gisterå A, Johansson DK, Maeda NN, Hansson GK, Ketelhuth DFJ, Coquet JM, Binder CJ, Karlsson MCI, Malin S. Acute Loss of Apolipoprotein E Triggers an Autoimmune Response That Accelerates Atherosclerosis. Arterioscler Thromb Vasc Biol 2019; 38:e145-e158. [PMID: 29880490 DOI: 10.1161/atvbaha.118.310802] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Objective- Dyslipidemia is a component of the metabolic syndrome, an established risk factor for atherosclerotic cardiovascular disease, and is also observed in various autoimmune and chronic inflammatory conditions. However, there are limited opportunities to study the impact of acquired dyslipidemia on cardiovascular and immune pathology. Approach and Results- We designed a model system that allows for the conversion to a state of acute hyperlipidemia in adult life, so that the consequences of such a transition could be observed, through conditionally deleting APOE (apolipoprotein E) in the adult mouse. The transition to hypercholesterolemia was accompanied by adaptive immune responses, including the expansion of T lymphocyte helper cell 1, T follicular helper cell, and T regulatory subsets and the formation of germinal centers. Unlike steady-state Apoe-/- mice, abrupt loss of APOE induced rapid production of antibodies recognizing rheumatoid disease autoantigens. Genetic ablation of the germinal center reduced both autoimmunity and atherosclerosis, indicating that the immune response that follows loss of APOE is independent of atherosclerosis but nevertheless promotes plaque development. Conclusions- Our findings suggest that immune activation in response to hyperlipidemia could contribute to a wide range of inflammatory autoimmune diseases, including atherosclerosis.
Collapse
Affiliation(s)
- Monica Centa
- From the Department of Medicine and Center for Molecular Medicine, Karolinska University Hospital (M.C., K.E.P., K.H., L.H., A.D., K.A.P., A.G., D.K.J., G.K.H., D.F.J.K., S.M.)
| | - Kajsa E Prokopec
- From the Department of Medicine and Center for Molecular Medicine, Karolinska University Hospital (M.C., K.E.P., K.H., L.H., A.D., K.A.P., A.G., D.K.J., G.K.H., D.F.J.K., S.M.)
| | - Manasa G Garimella
- Department of Microbiology, Tumor, and Cell Biology (M.G.G., J.M.S., C.A.T., J.M.C., M.C.I.K.), Karolinska Institutet, Stockholm, Sweden
| | - Katrin Habir
- From the Department of Medicine and Center for Molecular Medicine, Karolinska University Hospital (M.C., K.E.P., K.H., L.H., A.D., K.A.P., A.G., D.K.J., G.K.H., D.F.J.K., S.M.)
| | - Lisa Hofste
- From the Department of Medicine and Center for Molecular Medicine, Karolinska University Hospital (M.C., K.E.P., K.H., L.H., A.D., K.A.P., A.G., D.K.J., G.K.H., D.F.J.K., S.M.)
| | - Julian M Stark
- Department of Microbiology, Tumor, and Cell Biology (M.G.G., J.M.S., C.A.T., J.M.C., M.C.I.K.), Karolinska Institutet, Stockholm, Sweden
| | - Albert Dahdah
- From the Department of Medicine and Center for Molecular Medicine, Karolinska University Hospital (M.C., K.E.P., K.H., L.H., A.D., K.A.P., A.G., D.K.J., G.K.H., D.F.J.K., S.M.)
| | - Chris A Tibbitt
- Department of Microbiology, Tumor, and Cell Biology (M.G.G., J.M.S., C.A.T., J.M.C., M.C.I.K.), Karolinska Institutet, Stockholm, Sweden
| | - Konstantinos A Polyzos
- From the Department of Medicine and Center for Molecular Medicine, Karolinska University Hospital (M.C., K.E.P., K.H., L.H., A.D., K.A.P., A.G., D.K.J., G.K.H., D.F.J.K., S.M.)
| | - Anton Gisterå
- From the Department of Medicine and Center for Molecular Medicine, Karolinska University Hospital (M.C., K.E.P., K.H., L.H., A.D., K.A.P., A.G., D.K.J., G.K.H., D.F.J.K., S.M.)
| | - Daniel K Johansson
- From the Department of Medicine and Center for Molecular Medicine, Karolinska University Hospital (M.C., K.E.P., K.H., L.H., A.D., K.A.P., A.G., D.K.J., G.K.H., D.F.J.K., S.M.)
| | - Nobuyo N Maeda
- Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill (N.N.M.)
| | - Göran K Hansson
- From the Department of Medicine and Center for Molecular Medicine, Karolinska University Hospital (M.C., K.E.P., K.H., L.H., A.D., K.A.P., A.G., D.K.J., G.K.H., D.F.J.K., S.M.)
| | - Daniel F J Ketelhuth
- From the Department of Medicine and Center for Molecular Medicine, Karolinska University Hospital (M.C., K.E.P., K.H., L.H., A.D., K.A.P., A.G., D.K.J., G.K.H., D.F.J.K., S.M.)
| | - Jonathan M Coquet
- Department of Microbiology, Tumor, and Cell Biology (M.G.G., J.M.S., C.A.T., J.M.C., M.C.I.K.), Karolinska Institutet, Stockholm, Sweden
| | - Christoph J Binder
- Department of Laboratory Medicine, Medical University of Vienna, Austria (C.J.B.).,CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna (C.J.B.)
| | - Mikael C I Karlsson
- Department of Microbiology, Tumor, and Cell Biology (M.G.G., J.M.S., C.A.T., J.M.C., M.C.I.K.), Karolinska Institutet, Stockholm, Sweden
| | - Stephen Malin
- From the Department of Medicine and Center for Molecular Medicine, Karolinska University Hospital (M.C., K.E.P., K.H., L.H., A.D., K.A.P., A.G., D.K.J., G.K.H., D.F.J.K., S.M.)
| |
Collapse
|
27
|
Danvin A, Quillard T, Espitia O, Charrier C, Guyomarch B, Gouëffic Y, Maurel B. Impact of Femoral Ossification on Local and Systemic Cardiovascular Patients' Condition. Ann Vasc Surg 2019; 60:335-345. [PMID: 31200045 DOI: 10.1016/j.avsg.2019.03.035] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2018] [Revised: 02/22/2019] [Accepted: 03/01/2019] [Indexed: 12/22/2022]
Abstract
BACKGROUND Vascular calcifications are associated with a high cardiovascular morbi-mortality in the coronary territory. In parallel, femoral arteries are more calcified and develop osteoid metaplasia (OM). This study was conducted to assess the predictive value of OM and local inflammation on the occurrence of mid- and long-term adverse cardiovascular events. METHOD Between 2008 and 2015, 86 atheromatous samples were harvested during femoral endarterectomy on 81 patients and processed for histomorphological analyses of calcifications and inflammation (monocytes and B cells). Histological findings were compared with the long-term follow-up of patients, including major adverse cardiac event (MACE), major adverse limb event (MALE), and mortality. Frequencies were presented as percentage, and continuous data, as mean and standard deviation. A P-value < 0.05 was considered statistically significant. RESULTS Median follow-up was 42.4 months (26.9-58.8). Twenty-eight percent of patients underwent a MACE; a MALE occurred in 18 (21%) limbs. Survival rate was 87.2% at 36 months. OM was found in 41 samples (51%), without any significant impact on the occurrence of MACE, MALE, or mortality. Preoperative white blood cell formulae revealed a higher rate of neutrophils associated with MACE (P = 0.04) and MALE (P = 0.0008), correlated with higher B cells counts in plaque samples. CONCLUSIONS OM is part of femoral calcifications in almost 50% of the cases but does not seem to be an independent predictive variable for MACE or MALE. However, a higher rate of B cell infiltration of the plaque and preoperative neutrophil blood count may be predictive of adverse events during follow-up.
Collapse
Affiliation(s)
- Aurore Danvin
- CHU Nantes, l'institut du thorax, service de chirurgie vasculaire, Nantes, France
| | - Thibaut Quillard
- Laboratoire de Physiopathologie de la Résorption Osseuse, Inserm UMR S 1238, Nantes, France
| | - Olivier Espitia
- Laboratoire de Physiopathologie de la Résorption Osseuse, Inserm UMR S 1238, Nantes, France; CHU Nantes, unité de médicine vasculaire, Nantes, France; Université de Nantes, Nantes, France
| | - Céline Charrier
- Laboratoire de Physiopathologie de la Résorption Osseuse, Inserm UMR S 1238, Nantes, France
| | - Béatrice Guyomarch
- CHU Nantes, institut du thorax, INSERM, CNRS, UNIV Nantes, Nantes, France
| | - Yann Gouëffic
- CHU Nantes, l'institut du thorax, service de chirurgie vasculaire, Nantes, France; Laboratoire de Physiopathologie de la Résorption Osseuse, Inserm UMR S 1238, Nantes, France; Université de Nantes, Nantes, France
| | - Blandine Maurel
- CHU Nantes, l'institut du thorax, service de chirurgie vasculaire, Nantes, France; Laboratoire de Physiopathologie de la Résorption Osseuse, Inserm UMR S 1238, Nantes, France.
| |
Collapse
|
28
|
Libby P, Kobold S. Inflammation: a common contributor to cancer, aging, and cardiovascular diseases-expanding the concept of cardio-oncology. Cardiovasc Res 2019; 115:824-829. [PMID: 30830168 PMCID: PMC6452304 DOI: 10.1093/cvr/cvz058] [Citation(s) in RCA: 118] [Impact Index Per Article: 19.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/25/2019] [Accepted: 02/28/2019] [Indexed: 12/19/2022] Open
Abstract
Inflammation participates in the pathogenesis of both cancer and cardiovascular disease. This review examines the mechanistic commonalities between these two scourges of humanity through the lens of inflammation biology. Inflammatory pathways contribute to the initiation, the progression, and the complication of both malignant tumours and atherosclerotic plaques. Modulation of inflammatory pathways have proven transformative in the treatment of cancers and have crossed the threshold of clinical reality as treatments to reduce the risk of cardiovascular events. The finding that clonal haematopoiesis drives both leukaemia and cardiovascular events provides yet another link between these two seemingly disparate diseases. The nascent specialty of cardio-oncology has initially focused on the cardiovascular complications of cancer therapies. The recognition of a more profound pathophysiologic connection between cancer and cardiovascular diseases should expand the concept of cardio-oncology. Embracing the mechanistic connection and transcending traditional barriers between disciplines offers immense opportunities for speeding innovative research that can address the growing burden of both cancer and cardiovascular disease.
Collapse
Affiliation(s)
- Peter Libby
- Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, 77 Avenue Louis Pasteur, Boston, MA, USA
| | - Sebastian Kobold
- Division of Clinical Pharmacology, Department of Medicine IV, Center of Integrated Protein Science Munich (CIPS-M), Klinikum der Universität München, Munich, Germany
| |
Collapse
|
29
|
Haghikia A, Li XS, Liman TG, Bledau N, Schmidt D, Zimmermann F, Kränkel N, Widera C, Sonnenschein K, Haghikia A, Weissenborn K, Fraccarollo D, Heimesaat MM, Bauersachs J, Wang Z, Zhu W, Bavendiek U, Hazen SL, Endres M, Landmesser U. Gut Microbiota-Dependent Trimethylamine N-Oxide Predicts Risk of Cardiovascular Events in Patients With Stroke and Is Related to Proinflammatory Monocytes. Arterioscler Thromb Vasc Biol 2018; 38:2225-2235. [PMID: 29976769 PMCID: PMC6202215 DOI: 10.1161/atvbaha.118.311023] [Citation(s) in RCA: 225] [Impact Index Per Article: 32.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2017] [Accepted: 06/19/2018] [Indexed: 12/13/2022]
Abstract
Objective- Gut microbiota-dependent metabolites, in particular trimethylamine N-oxide (TMAO), have recently been reported to promote atherosclerosis and thrombosis. Here, we examined for the first time the relation of TMAO and the risk of incident cardiovascular events in patients with recent first-ever ischemic stroke in 2 independent prospective cohorts. Moreover, the link between TMAO and proinflammatory monocytes as a potential contributing factor for cardiovascular risk in stroke patients was studied. Approach and Results- In a first study (n=78), higher TMAO plasma levels were linked with an increased risk of incident cardiovascular events including myocardial infarction, recurrent stroke, and cardiovascular death (fourth quartile versus first quartile; hazard ratio, 2.31; 95% CI, 1.25-4.23; P<0.01). In the second independent validation cohort (n=593), high TMAO levels again heralded marked increased risk of adverse cardiovascular events (fourth quartile versus first quartile; hazard ratio, 5.0; 95% CI, 1.7-14.8; P<0.01), and also after adjustments for cardiovascular risk factors including hypertension, diabetes mellitus, LDL (low-density lipoprotein) cholesterol, and estimated glomerular filtration rate (hazard ratio, 3.3; 95% CI, 1.2-10.9; P=0.04). A significant correlation was also found between TMAO levels and percentage of proinflammatory intermediate CD14++CD16+ monocytes ( r=0.70; P<0.01). Moreover, in mice fed a diet enriched with choline to increase TMAO synthesis, levels of proinflammatory murine Ly6Chigh monocytes were higher than in the chow-fed control group (choline: 9.2±0.5×103 per mL versus control: 6.5±0.5×103 per mL; P<0.01). This increase was abolished in mice with depleted gut microbiota (choline+antibiotics: 5.4±0.7×103 per mL; P<0.001 versus choline). Conclusions- The present study demonstrates for the first time a graded relation between TMAO levels and the risk of subsequent cardiovascular events in patients with recent prior ischemic stroke. Our data support the notion that TMAO-related increase of proinflammatory monocytes may add to elevated cardiovascular risk of patients with increased TMAO levels.
Collapse
Affiliation(s)
- Arash Haghikia
- Department of Cardiology, Charité-Universitätsmedizin Berlin, Campus Benjamin Franklin, Berlin, Germany
- DZHK (German Center for Cardiovascular Research), partner site Berlin, Berlin, Germany
| | - Xinmin S. Li
- Department of Cellular and Molecular Medicine, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Thomas G. Liman
- Department of Neurology, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Nils Bledau
- Cardiology and Angiology, Hannover Medical School, Hannover, Germany
| | - David Schmidt
- Department of Cardiology, Charité-Universitätsmedizin Berlin, Campus Benjamin Franklin, Berlin, Germany
| | - Friederike Zimmermann
- Department of Cardiology, Charité-Universitätsmedizin Berlin, Campus Benjamin Franklin, Berlin, Germany
| | - Nicolle Kränkel
- Department of Cardiology, Charité-Universitätsmedizin Berlin, Campus Benjamin Franklin, Berlin, Germany
- DZHK (German Center for Cardiovascular Research), partner site Berlin, Berlin, Germany
| | - Christian Widera
- Department of Cardiology, Heart Center Oldenburg, European Medical School Oldenburg-Groningen, Carl von Ossietzky University Oldenburg, Oldenburg, Germany
| | | | - Aiden Haghikia
- Department of Neurology, Ruhr-University Bochum, Bochum, Germany
| | | | | | - Markus M. Heimesaat
- Institute of Microbiology and Infection Immunology, Charité-Universitätsmedizin Berlin, Berlin, Germany
- Berlin Institute of Health (BIH), Berlin, Germany
| | - Johann Bauersachs
- Cardiology and Angiology, Hannover Medical School, Hannover, Germany
| | - Zeneng Wang
- Department of Cellular and Molecular Medicine, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Weifei Zhu
- Department of Cellular and Molecular Medicine, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Udo Bavendiek
- Cardiology and Angiology, Hannover Medical School, Hannover, Germany
| | - Stanley L. Hazen
- Department of Cellular and Molecular Medicine, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA
- Department of Cardiovascular Medicine, Heart and Vascular Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Matthias Endres
- DZHK (German Center for Cardiovascular Research), partner site Berlin, Berlin, Germany
- Department of Neurology, Charité-Universitätsmedizin Berlin, Berlin, Germany
- Berlin Institute of Health (BIH), Berlin, Germany
- Center for Stroke Research Berlin, Charité-Universitätsmedizin Berlin, Berlin, Germany
- German Center for Neurodegenerative Diseases (DZNE), Berlin, Germany
| | - Ulf Landmesser
- Department of Cardiology, Charité-Universitätsmedizin Berlin, Campus Benjamin Franklin, Berlin, Germany
- DZHK (German Center for Cardiovascular Research), partner site Berlin, Berlin, Germany
- Berlin Institute of Health (BIH), Berlin, Germany
| |
Collapse
|
30
|
Differential Interaction of Platelet-Derived Extracellular Vesicles with Leukocyte Subsets in Human Whole Blood. Sci Rep 2018; 8:6598. [PMID: 29700367 PMCID: PMC5920058 DOI: 10.1038/s41598-018-25047-x] [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: 01/08/2018] [Accepted: 04/05/2018] [Indexed: 12/18/2022] Open
Abstract
Secretion and exchange of biomolecules via extracellular vesicles (EVs) are crucial mechanisms in intercellular communication, and the roles of EVs in infection, inflammation, or thrombosis have been increasingly recognized. EVs have emerged as central players in immune regulation and can enhance or suppress the immune response, depending on the state of donor and recipient cells. We investigated the interaction of blood cell-derived EVs with leukocyte subpopulations (monocytes and their subsets, granulocytes, B cells, T cells, and NK cells) directly in whole blood using a combination of flow cytometry, imaging flow cytometry, cell sorting, and high resolution confocal microscopy. Platelet-derived EVs constituted the majority of circulating EVs and were preferentially associated with granulocytes and monocytes, while they scarcely interacted with lymphocytes. Further flow cytometric differentiation of monocyte subsets provided clear indications for a preferential association of platelet-derived EVs with intermediate (CD14++CD16+) monocytes in whole blood.
Collapse
|
31
|
Cignarella A, Tedesco S, Cappellari R, Fadini GP. The continuum of monocyte phenotypes: Experimental evidence and prognostic utility in assessing cardiovascular risk. J Leukoc Biol 2018; 103:1021-1028. [PMID: 29603382 DOI: 10.1002/jlb.5ru1217-477rr] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2017] [Revised: 02/23/2018] [Accepted: 02/23/2018] [Indexed: 12/15/2022] Open
Abstract
The monocyte-macrophage cell lineage represents a major player in innate immunity, and is involved in many physiologic and pathologic conditions. Particularly, monocyte-macrophages play a very important role in atherosclerosis and cardiovascular disease. Monocyte heterogeneity is well recognized but the biologic and clinical meaning of the various monocyte subtypes is not entirely understood. Traditionally, monocytes can be divided in classical, intermediate, and nonclassical based on expression of the surface antigens CD14 and CD16. While macrophage diversity is now well recognized to organize as a continuum, monocyte subsets have long been considered as separated entities. However, mounting evidence obtained by tracking the ontology of human monocytes help clarifying that monocytes mature from classical to nonclassical ones, through an intermediate phenotype. This concept is therefore best depicted as a continuum, whereas the subdivision into discrete CD14/CD16 subsets appears an oversimplification. In this review, we discuss the evidence supporting the existence of a monocyte continuum along with the technical challenges of monocyte characterization. In particular, we describe the advantage of considering monocytes along a continuous distribution for the evaluation of cardiovascular risk. We make the point that small transition along the monocyte continuum better reflects cardiovascular risk than a simplified analysis of discrete monocyte subsets. Recognizing the monocyte continuum can be helpful to model other pathophysiologic conditions where these cells are involved.
Collapse
Affiliation(s)
| | | | | | - Gian Paolo Fadini
- Department of Medicine, University of Padova, Padova, Italy
- Venetian Institute of Molecular Medicine, Padova, Italy
| |
Collapse
|
32
|
Zhang B, Qi X, Zhao Y, Li R, Zhang C, Chang HM, Pang Y, Qiao J. Elevated CD14++CD16+ Monocytes in Hyperhomocysteinemia-Associated Insulin Resistance in Polycystic Ovary Syndrome. Reprod Sci 2018; 25:1629-1636. [DOI: 10.1177/1933719118756772] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Affiliation(s)
- Bochun Zhang
- Department of Obstetrics and Gynecology, Center for Reproductive Medicine, Peking University Third Hospital, Beijing, China
- Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Beijing, China
- Key Laboratory of Assisted Reproduction, Ministry of Education, Beijing, China
| | - Xinyu Qi
- Department of Obstetrics and Gynecology, Center for Reproductive Medicine, Peking University Third Hospital, Beijing, China
- Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Beijing, China
- Key Laboratory of Assisted Reproduction, Ministry of Education, Beijing, China
| | - Yue Zhao
- Department of Obstetrics and Gynecology, Center for Reproductive Medicine, Peking University Third Hospital, Beijing, China
- Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Beijing, China
- Key Laboratory of Assisted Reproduction, Ministry of Education, Beijing, China
| | - Rong Li
- Department of Obstetrics and Gynecology, Center for Reproductive Medicine, Peking University Third Hospital, Beijing, China
- Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Beijing, China
- Key Laboratory of Assisted Reproduction, Ministry of Education, Beijing, China
| | - Chunmei Zhang
- Department of Obstetrics and Gynecology, Center for Reproductive Medicine, Peking University Third Hospital, Beijing, China
- Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Beijing, China
- Key Laboratory of Assisted Reproduction, Ministry of Education, Beijing, China
| | - Hsun-Ming Chang
- Department of Obstetrics and Gynecology, Center for Reproductive Medicine, Peking University Third Hospital, Beijing, China
- Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Beijing, China
- Key Laboratory of Assisted Reproduction, Ministry of Education, Beijing, China
| | - Yanli Pang
- Department of Obstetrics and Gynecology, Center for Reproductive Medicine, Peking University Third Hospital, Beijing, China
- Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Beijing, China
- Key Laboratory of Assisted Reproduction, Ministry of Education, Beijing, China
| | - Jie Qiao
- Department of Obstetrics and Gynecology, Center for Reproductive Medicine, Peking University Third Hospital, Beijing, China
- Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Beijing, China
- Key Laboratory of Assisted Reproduction, Ministry of Education, Beijing, China
| |
Collapse
|
33
|
Simion V, Haemmig S, Feinberg MW. LncRNAs in vascular biology and disease. Vascul Pharmacol 2018; 114:145-156. [PMID: 29425892 DOI: 10.1016/j.vph.2018.01.003] [Citation(s) in RCA: 130] [Impact Index Per Article: 18.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2017] [Revised: 01/09/2018] [Accepted: 01/16/2018] [Indexed: 12/14/2022]
Abstract
Accumulating studies indicate that long non-coding RNAs (lncRNAs) play important roles in the regulation of diverse biological processes involved in homeostatic control of the vessel wall in health and disease. However, our knowledge of the mechanisms by which lncRNAs control gene expression and cell signaling pathways is still nascent. Furthermore, only a handful of lncRNAs has been functionally evaluated in response to pathophysiological stimuli or in vascular disease states. For example, lncRNAs may regulate endothelial dysfunction by modulating endothelial cell proliferation (e.g. MALAT1, H19) or angiogenesis (e.g. MEG3, MANTIS). LncRNAs have also been implicated in modulating vascular smooth muscle cell (VSMC) phenotypes or vascular remodeling (e.g. ANRIL, SMILR, SENCR, MYOSLID). Finally, emerging studies have implicated lncRNAs in leukocytes activation (e.g. lincRNA-Cox2, linc00305, THRIL), macrophage polarization (e.g. GAS5), and cholesterol metabolism (e.g. LeXis). This review summarizes recent findings on the expression, mechanism, and function of lncRNAs implicated in a range of vascular disease states from mice to human subjects. An improved understanding of lncRNAs in vascular disease may provide new pathophysiological insights and opportunities for the generation of a new class of RNA-based biomarkers and therapeutic targets.
Collapse
Affiliation(s)
- Viorel Simion
- Department of Medicine, Cardiovascular Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Stefan Haemmig
- Department of Medicine, Cardiovascular Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Mark W Feinberg
- Department of Medicine, Cardiovascular Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
34
|
Spronk HMH, Padro T, Siland JE, Prochaska JH, Winters J, van der Wal AC, Posthuma JJ, Lowe G, d'Alessandro E, Wenzel P, Coenen DM, Reitsma PH, Ruf W, van Gorp RH, Koenen RR, Vajen T, Alshaikh NA, Wolberg AS, Macrae FL, Asquith N, Heemskerk J, Heinzmann A, Moorlag M, Mackman N, van der Meijden P, Meijers JCM, Heestermans M, Renné T, Dólleman S, Chayouâ W, Ariëns RAS, Baaten CC, Nagy M, Kuliopulos A, Posma JJ, Harrison P, Vries MJ, Crijns HJGM, Dudink EAMP, Buller HR, Henskens YMC, Själander A, Zwaveling S, Erküner O, Eikelboom JW, Gulpen A, Peeters FECM, Douxfils J, Olie RH, Baglin T, Leader A, Schotten U, Scaf B, van Beusekom HMM, Mosnier LO, van der Vorm L, Declerck P, Visser M, Dippel DWJ, Strijbis VJ, Pertiwi K, Ten Cate-Hoek AJ, Ten Cate H. Atherothrombosis and Thromboembolism: Position Paper from the Second Maastricht Consensus Conference on Thrombosis. Thromb Haemost 2018; 118:229-250. [PMID: 29378352 DOI: 10.1160/th17-07-0492] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Atherothrombosis is a leading cause of cardiovascular mortality and long-term morbidity. Platelets and coagulation proteases, interacting with circulating cells and in different vascular beds, modify several complex pathologies including atherosclerosis. In the second Maastricht Consensus Conference on Thrombosis, this theme was addressed by diverse scientists from bench to bedside. All presentations were discussed with audience members and the results of these discussions were incorporated in the final document that presents a state-of-the-art reflection of expert opinions and consensus recommendations regarding the following five topics: 1. Risk factors, biomarkers and plaque instability: In atherothrombosis research, more focus on the contribution of specific risk factors like ectopic fat needs to be considered; definitions of atherothrombosis are important distinguishing different phases of disease, including plaque (in)stability; proteomic and metabolomics data are to be added to genetic information. 2. Circulating cells including platelets and atherothrombosis: Mechanisms of leukocyte and macrophage plasticity, migration, and transformation in murine atherosclerosis need to be considered; disease mechanism-based biomarkers need to be identified; experimental systems are needed that incorporate whole-blood flow to understand how red blood cells influence thrombus formation and stability; knowledge on platelet heterogeneity and priming conditions needs to be translated toward the in vivo situation. 3. Coagulation proteases, fibrin(ogen) and thrombus formation: The role of factor (F) XI in thrombosis including the lower margins of this factor related to safe and effective antithrombotic therapy needs to be established; FXI is a key regulator in linking platelets, thrombin generation, and inflammatory mechanisms in a renin-angiotensin dependent manner; however, the impact on thrombin-dependent PAR signaling needs further study; the fundamental mechanisms in FXIII biology and biochemistry and its impact on thrombus biophysical characteristics need to be explored; the interactions of red cells and fibrin formation and its consequences for thrombus formation and lysis need to be addressed. Platelet-fibrin interactions are pivotal determinants of clot formation and stability with potential therapeutic consequences. 4. Preventive and acute treatment of atherothrombosis and arterial embolism; novel ways and tailoring? The role of protease-activated receptor (PAR)-4 vis à vis PAR-1 as target for antithrombotic therapy merits study; ongoing trials on platelet function test-based antiplatelet therapy adjustment support development of practically feasible tests; risk scores for patients with atrial fibrillation need refinement, taking new biomarkers including coagulation into account; risk scores that consider organ system differences in bleeding may have added value; all forms of oral anticoagulant treatment require better organization, including education and emergency access; laboratory testing still needs rapidly available sensitive tests with short turnaround time. 5. Pleiotropy of coagulation proteases, thrombus resolution and ischaemia-reperfusion: Biobanks specifically for thrombus storage and analysis are needed; further studies on novel modified activated protein C-based agents are required including its cytoprotective properties; new avenues for optimizing treatment of patients with ischaemic stroke are needed, also including novel agents that modify fibrinolytic activity (aimed at plasminogen activator inhibitor-1 and thrombin activatable fibrinolysis inhibitor.
Collapse
Affiliation(s)
- H M H Spronk
- Laboratory for Clinical Thrombosis and Haemostasis, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University Medical Center, Maastricht, The Netherlands
| | - T Padro
- Cardiovascular Research Center (ICCC), Hospital Sant Pau, Barcelona, Spain
| | - J E Siland
- Department of Cardiology, University Medical Center Groningen, Groningen, The Netherlands
| | - J H Prochaska
- Center for Cardiology/Center for Thrombosis and Hemostasis/DZHK, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - J Winters
- Department of Physiology, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, The Netherlands
| | - A C van der Wal
- Department of Pathology, Academic Medical Center (AMC), Amsterdam, The Netherlands
| | - J J Posthuma
- Laboratory for Clinical Thrombosis and Haemostasis, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University Medical Center, Maastricht, The Netherlands
| | - G Lowe
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, Scotland
| | - E d'Alessandro
- Department of Physiology, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, The Netherlands.,Department of Pathology, Academic Medical Center (AMC), Amsterdam, The Netherlands
| | - P Wenzel
- Department of Cardiology, Universitätsmedizin Mainz, Mainz, Germany
| | - D M Coenen
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, The Netherlands
| | - P H Reitsma
- Einthoven Laboratory, Leiden University Medical Center, Leiden, The Netherlands
| | - W Ruf
- Center for Cardiology/Center for Thrombosis and Hemostasis/DZHK, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - R H van Gorp
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, The Netherlands
| | - R R Koenen
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, The Netherlands
| | - T Vajen
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, The Netherlands
| | - N A Alshaikh
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, The Netherlands
| | - A S Wolberg
- Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, North Carolina, United States
| | - F L Macrae
- Thrombosis and Tissue Repair Group, Division of Cardiovascular and Diabetes Research, Leeds Institute of Cardiovascular and Metabolic Medicine, School of Medicine, University of Leeds, Leeds, UK
| | - N Asquith
- Thrombosis and Tissue Repair Group, Division of Cardiovascular and Diabetes Research, Leeds Institute of Cardiovascular and Metabolic Medicine, School of Medicine, University of Leeds, Leeds, UK
| | - J Heemskerk
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, The Netherlands
| | - A Heinzmann
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, The Netherlands
| | - M Moorlag
- Synapse, Maastricht, The Netherlands
| | - N Mackman
- Department of Medicine, UNC McAllister Heart Institute, University of North Carolina, Chapel Hill, North Carolina, United States
| | - P van der Meijden
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, The Netherlands
| | - J C M Meijers
- Department of Plasma Proteins, Sanquin, Amsterdam, The Netherlands
| | - M Heestermans
- Einthoven Laboratory, Leiden University Medical Center, Leiden, The Netherlands
| | - T Renné
- Department of Molecular Medicine and Surgery, Karolinska Institutet and University Hospital, Stockholm, Sweden.,Institute of Clinical Chemistry and Laboratory Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - S Dólleman
- Department of Nephrology, Leiden University Medical Centre, Leiden, The Netherlands
| | - W Chayouâ
- Synapse, Maastricht, The Netherlands
| | - R A S Ariëns
- Thrombosis and Tissue Repair Group, Division of Cardiovascular and Diabetes Research, Leeds Institute of Cardiovascular and Metabolic Medicine, School of Medicine, University of Leeds, Leeds, UK
| | - C C Baaten
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, The Netherlands
| | - M Nagy
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, The Netherlands
| | - A Kuliopulos
- Tufts University School of Graduate Biomedical Sciences, Biochemistry/Developmental, Molecular and Chemical Biology, Tufts University School of Medicine, Boston, Massachusetts
| | - J J Posma
- Laboratory for Clinical Thrombosis and Haemostasis, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University Medical Center, Maastricht, The Netherlands
| | - P Harrison
- Institute of Inflammation and Ageing, University of Birmingham, Birmingham, United Kingdom
| | - M J Vries
- Laboratory for Clinical Thrombosis and Haemostasis, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University Medical Center, Maastricht, The Netherlands
| | - H J G M Crijns
- Department of Cardiology, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University Medical Center, Maastricht, The Netherlands
| | - E A M P Dudink
- Department of Cardiology, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University Medical Center, Maastricht, The Netherlands
| | - H R Buller
- Department of Vascular Medicine, Academic Medical Center (AMC), Amsterdam, The Netherlands
| | - Y M C Henskens
- Laboratory for Clinical Thrombosis and Haemostasis, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University Medical Center, Maastricht, The Netherlands
| | - A Själander
- Department of Public Health and Clinical Medicine, Umeå University, Umeå, Sweden
| | - S Zwaveling
- Laboratory for Clinical Thrombosis and Haemostasis, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University Medical Center, Maastricht, The Netherlands.,Synapse, Maastricht, The Netherlands
| | - O Erküner
- Department of Cardiology, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University Medical Center, Maastricht, The Netherlands
| | - J W Eikelboom
- Department of Medicine, McMaster University, Hamilton, Ontario, Canada
| | - A Gulpen
- Laboratory for Clinical Thrombosis and Haemostasis, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University Medical Center, Maastricht, The Netherlands
| | - F E C M Peeters
- Department of Cardiology, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University Medical Center, Maastricht, The Netherlands
| | - J Douxfils
- Department of Pharmacy, Thrombosis and Hemostasis Center, Faculty of Medicine, Namur University, Namur, Belgium
| | - R H Olie
- Laboratory for Clinical Thrombosis and Haemostasis, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University Medical Center, Maastricht, The Netherlands
| | - T Baglin
- Department of Haematology, Addenbrookes Hospital Cambridge, Cambridge, United Kingdom
| | - A Leader
- Laboratory for Clinical Thrombosis and Haemostasis, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University Medical Center, Maastricht, The Netherlands.,Davidoff Cancer Center, Rabin Medical Center, Institute of Hematology, Sackler Faculty of Medicine, Tel Aviv University, Petah Tikva, Tel Aviv, Israel
| | - U Schotten
- Center for Cardiology/Center for Thrombosis and Hemostasis/DZHK, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - B Scaf
- Laboratory for Clinical Thrombosis and Haemostasis, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University Medical Center, Maastricht, The Netherlands.,Department of Physiology, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, The Netherlands
| | - H M M van Beusekom
- Department of Experimental Cardiology, Erasmus Medical Center, Rotterdam, The Netherlands
| | - L O Mosnier
- Department of Molecular and Experimental Medicine, The Scripps Research Institute, La Jolla, United States
| | | | - P Declerck
- Department of Pharmaceutical and Pharmacological Sciences, University of Leuven, Leuven, Belgium
| | | | - D W J Dippel
- Department of Neurology, Erasmus MC, Rotterdam, The Netherlands
| | | | - K Pertiwi
- Department of Cardiovascular Pathology, University of Amsterdam, Academic Medical Center, Amsterdam, The Netherlands
| | - A J Ten Cate-Hoek
- Laboratory for Clinical Thrombosis and Haemostasis, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University Medical Center, Maastricht, The Netherlands
| | - H Ten Cate
- Laboratory for Clinical Thrombosis and Haemostasis, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University Medical Center, Maastricht, The Netherlands
| |
Collapse
|
35
|
Patel VK, Williams H, Li SCH, Fletcher JP, Medbury HJ. Monocyte inflammatory profile is specific for individuals and associated with altered blood lipid levels. Atherosclerosis 2017; 263:15-23. [PMID: 28570862 DOI: 10.1016/j.atherosclerosis.2017.05.026] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/03/2017] [Revised: 05/18/2017] [Accepted: 05/19/2017] [Indexed: 01/06/2023]
Abstract
BACKGROUND AND AIMS Atherogenesis is dependent upon monocyte influx into the vessel wall. In humans, three monocyte subsets exist, the number and function of which are significantly altered in cardiovascular disease (CVD). Whether such alterations arise in individuals with a perturbed lipid profile remains largely unanswered, but is important to delineate, as adoption of a pro-inflammatory state may promote plaque formation. Here, we compared the inflammatory status of monocyte subsets and determined whether monocyte inflammatory changes are evident in individuals with a perturbed lipid profile. METHODS Monocyte subset cytokine production, inflammatory and anti-inflammatory marker expression were determined by whole blood flow cytometry and related to participants' lipid levels. RESULTS The intermediate and non-classical monocytes were more inflammatory than classicals as seen by their higher cytokine production (TNF-α, IL-1β, IL-6) and M1 marker (CD86) expression, but lower levels of M2 markers (CD93, CD163). More importantly, a considerable variation was seen between participants, with all monocytes of one individual being more inflammatory than those of another. Many inter-individual differences were related to participants' lipid levels. IL-1β production correlated negatively with Apo A1 and HDL-C. CD86 and TLR2 correlated positively with Chol:HDL-C but negatively with HDL-C and Apo A1:Apo B. Interestingly, CD163 expression correlated positively with Chol:HDL-C but negatively with Apo A1:Apo B. CONCLUSIONS Our data indicates that priming of all monocytes to an inflammatory state occurs in individuals with a perturbed lipid profile, overriding the normal functional distinction attributed to the different monocyte subsets. As such, all monocytes may be important in CVD.
Collapse
Affiliation(s)
- Vyoma K Patel
- Westmead Hospital, Department of Surgery, Vascular Biology Research Centre, Westmead, NSW, Australia; The University of Sydney, Western Clinical School, Westmead, NSW, Australia
| | - Helen Williams
- Westmead Hospital, Department of Surgery, Vascular Biology Research Centre, Westmead, NSW, Australia; The University of Sydney, Western Clinical School, Westmead, NSW, Australia
| | - Stephen C H Li
- The University of Sydney, Western Clinical School, Westmead, NSW, Australia; Institute of Clinical Pathology and Medical Research, Westmead Hospital, Westmead, NSW, Australia
| | - John P Fletcher
- Westmead Hospital, Department of Surgery, Vascular Biology Research Centre, Westmead, NSW, Australia; The University of Sydney, Western Clinical School, Westmead, NSW, Australia
| | - Heather J Medbury
- Westmead Hospital, Department of Surgery, Vascular Biology Research Centre, Westmead, NSW, Australia; The University of Sydney, Western Clinical School, Westmead, NSW, Australia.
| |
Collapse
|
36
|
Haynes A, Linden MD, Robey E, Naylor LH, Cox KL, Lautenschlager NT, Green DJ. Relationship between monocyte-platelet aggregation and endothelial function in middle-aged and elderly adults. Physiol Rep 2017; 5:e13189. [PMID: 28533260 PMCID: PMC5449553 DOI: 10.14814/phy2.13189] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2017] [Accepted: 02/09/2017] [Indexed: 01/22/2023] Open
Abstract
Low-grade inflammation, endothelial dysfunction, and platelet hyper-reactivity to agonists are associated with an increased risk of cardiovascular events. In vitro and animal studies infer an inverse mechanistic relationship between platelet activation and the production of endothelium-derived nitric oxide and prostacyclin. This concept is supported by evidence of an inverse relationship between endothelial function and platelet activation in high-risk cardiac patients. The aim of this study was to investigate what relationship, if any, exists between platelet and endothelial function in healthy, middle-aged, and elderly adults. In 51 participants (18 male, 33 post menopausal female), endothelial function was assessed by flow-mediated dilation (FMD). Platelet function was assessed by flow cytometric determination of glycoprotein IIb/IIIa activation (measured by PAC-1 binding), granule exocytosis (measured by surface P-selectin expression), and monocyte-platelet aggregates (MPAs), with and without stimulation by canonical platelet agonists adenosine diphosphate (ADP), arachidonic acid (AA), and collagen. Correlation analysis indicated there was no significant (all P => 0.05) relationship between FMD and any marker of in vivo platelet activation (MPAs R = 0.193, PAC-1 R = -0.113, anti-CD62P R = -0.078) or inducible platelet activation by ADP (MPA R = -0.128, anti-CD62P R = -0.237), AA (MPA R = -0.122, PAC-1 R = -0.045, anti-CD62P R = -0.142), or collagen (MPA R = 0.136, PAC-1 R = 0.174, anti-CD62P R = -0.077). Our findings contrast with two previous studies performed in high-risk cardiac patients, which reported inverse relationships between platelet activation and endothelial function, suggesting that some compensatory redundancy may exist in the relationship between platelet and endothelial function in preclinical populations.
Collapse
Affiliation(s)
- Andrew Haynes
- School of Sport ScienceExercise and HealthUniversity of Western AustraliaCrawleyWestern AustraliaAustralia
| | - Matthew D. Linden
- School of Pathology and Laboratory MedicineUniversity of Western AustraliaCrawleyWestern Australia
| | - Elisa Robey
- School of Sport ScienceExercise and HealthUniversity of Western AustraliaCrawleyWestern AustraliaAustralia
| | - Louise H. Naylor
- School of Sport ScienceExercise and HealthUniversity of Western AustraliaCrawleyWestern AustraliaAustralia
| | - Kay L. Cox
- School of Sport ScienceExercise and HealthUniversity of Western AustraliaCrawleyWestern AustraliaAustralia
- School of Medicine and Pharmacology (Royal Perth Hospital Unit)University of Western AustraliaCrawleyWestern AustraliaAustralia
| | - Nicola T. Lautenschlager
- Academic Unit for Psychiatry of Old AgeDepartment of PsychiatryUniversity of MelbourneMelbourneVictoriaAustralia
- NorthWestern Mental HealthMelbourne HealthParkvilleVictoriaAustralia
- School of Clinical Neurosciences and the Western Australia Centre for Health and AgeingUniversity of Western AustraliaCrawleyWestern AustraliaAustralia
| | - Daniel J. Green
- School of Sport ScienceExercise and HealthUniversity of Western AustraliaCrawleyWestern AustraliaAustralia
- Research Institute for Sport and Exercise ScienceLiverpool John Moores UniversityLiverpoolUnited Kingdom
- Principal Research FellowNational Health and Medical Research CouncilCanberraAustralian Capital TerritoryAustralia
| |
Collapse
|
37
|
Kingery JR, Hamid T, Lewis RK, Ismahil MA, Bansal SS, Rokosh G, Townes TM, Ildstad ST, Jones SP, Prabhu SD. Leukocyte iNOS is required for inflammation and pathological remodeling in ischemic heart failure. Basic Res Cardiol 2017; 112:19. [PMID: 28238121 DOI: 10.1007/s00395-017-0609-2] [Citation(s) in RCA: 59] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/08/2016] [Accepted: 02/23/2017] [Indexed: 12/18/2022]
Abstract
In the failing heart, iNOS is expressed by both macrophages and cardiomyocytes. We hypothesized that inflammatory cell-localized iNOS exacerbates left ventricular (LV) remodeling. Wild-type (WT) C57BL/6 mice underwent total body irradiation and reconstitution with bone marrow from iNOS-/- mice (iNOS-/-c) or WT mice (WTc). Chimeric mice underwent coronary ligation to induce large infarction and ischemic heart failure (HF), or sham surgery. After 28 days, as compared with WTc sham mice, WTc HF mice exhibited significant (p < 0.05) mortality, LV dysfunction, hypertrophy, fibrosis, oxidative/nitrative stress, inflammatory activation, and iNOS upregulation. These mice also exhibited a ~twofold increase in circulating Ly6Chi pro-inflammatory monocytes, and ~sevenfold higher cardiac M1 macrophages, which were primarily CCR2- cells. In contrast, as compared with WTc HF mice, iNOS-/-c HF mice exhibited significantly improved survival, LV function, hypertrophy, fibrosis, oxidative/nitrative stress, and inflammatory activation, without differences in overall cardiac iNOS expression. Moreover, iNOS-/-c HF mice exhibited lower circulating Ly6Chi monocytes, and augmented cardiac M2 macrophages, but with greater infiltrating monocyte-derived CCR2+ macrophages vs. WTc HF mice. Lastly, upon cell-to-cell contact with naïve cardiomyocytes, peritoneal macrophages from WT HF mice depressed contraction, and augmented cardiomyocyte oxygen free radicals and peroxynitrite. These effects were not observed upon contact with macrophages from iNOS-/- HF mice. We conclude that leukocyte iNOS is obligatory for local and systemic inflammatory activation and cardiac remodeling in ischemic HF. Activated macrophages in HF may directly induce cardiomyocyte contractile dysfunction and oxidant stress upon cell-to-cell contact; this juxtacrine response requires macrophage-localized iNOS.
Collapse
Affiliation(s)
- Justin R Kingery
- Department of Medicine, University of Louisville, Louisville, KY, USA.,Department of Medicine, Weill Cornell Medical College, New York, NY, USA
| | - Tariq Hamid
- Department of Medicine, University of Louisville, Louisville, KY, USA.,Division of Cardiovascular Disease, Department of Medicine, University of Alabama at Birmingham, and Birmingham VAMC, Birmingham, AL, USA
| | - Robert K Lewis
- Department of Medicine, University of Louisville, Louisville, KY, USA.,Department of Medicine, Duke University School of Medicine, Durham, NC, USA
| | - Mohamed Ameen Ismahil
- Department of Medicine, University of Louisville, Louisville, KY, USA.,Division of Cardiovascular Disease, Department of Medicine, University of Alabama at Birmingham, and Birmingham VAMC, Birmingham, AL, USA
| | - Shyam S Bansal
- Division of Cardiovascular Disease, Department of Medicine, University of Alabama at Birmingham, and Birmingham VAMC, Birmingham, AL, USA
| | - Gregg Rokosh
- Division of Cardiovascular Disease, Department of Medicine, University of Alabama at Birmingham, and Birmingham VAMC, Birmingham, AL, USA
| | - Tim M Townes
- Department of Biochemistry & Molecular Genetics, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Suzanne T Ildstad
- Department of Surgery, University of Louisville, Louisville, KY, USA
| | - Steven P Jones
- Department of Medicine, University of Louisville, Louisville, KY, USA
| | - Sumanth D Prabhu
- Department of Medicine, University of Louisville, Louisville, KY, USA. .,Division of Cardiovascular Disease, Department of Medicine, University of Alabama at Birmingham, and Birmingham VAMC, Birmingham, AL, USA.
| |
Collapse
|
38
|
Urbanski K, Ludew D, Filip G, Filip M, Sagan A, Szczepaniak P, Grudzien G, Sadowski J, Jasiewicz-Honkisz B, Sliwa T, Kapelak B, McGinnigle E, Mikolajczyk T, Guzik TJ. CD14 +CD16 ++ "nonclassical" monocytes are associated with endothelial dysfunction in patients with coronary artery disease. Thromb Haemost 2017; 117:971-980. [PMID: 28229168 DOI: 10.1160/th16-08-0614] [Citation(s) in RCA: 57] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2016] [Accepted: 01/23/2016] [Indexed: 11/05/2022]
Abstract
Endothelial dysfunction and inflammation are key mechanisms of vascular disease. We hypothesised that heterogeneity of monocyte subpopulations may be related to the development of vascular dysfunction in coronary artery disease (CAD). Therefore, we examined the relationships between monocyte subsets (CD14++CD16- "classical - Mon1", CD14++CD16+ "intermediate - Mon2" and CD14+CD16++ "nonclassical - Mon3"), endothelial function and risk factor profiles in 130 patients with CAD undergoing coronary artery bypass grafting. This allowed for direct nitric oxide (NO) bioavailability assessment using isometric tension studies ex vivo (acetylcholine; ACh- and sodium-nitropruside; SNP-dependent) in segments of internal mammary arteries. The expression of CD14 and CD16 antigens and activation markers were determined in peripheral blood mononuclear cells using flow cytometry. Patients with high CD14+CD16++ "nonclassical" and low CD14++CD16- "classical" monocytes presented impaired endothelial function. High frequency of CD14+CD16++ "nonclassical" monocytes was associated with increased vascular superoxide production. Furthermore, endothelial dysfunction was associated with higher expression of activation marker CD11c selectively on CD14+CD16++ monocytes. Nonclassical and classical monocyte frequencies remained independent predictors of endothelial dysfunction when major risk factors for atherosclerosis were taken into account (β=0.18 p=0.04 and β=-0.19 p=0.03, respectively). In summary, our data indicate that CD14+CD16++ "nonclassical" monocytes are associated with more advanced vascular dysfunction measured as NO- bioavailability and vascular reactive oxygen species production.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | | | - Tomasz J Guzik
- Tomasz J. Guzik, MD, PhD, FRCP, BHF Centre for Excellence, Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, UK, E-mail:
| |
Collapse
|
39
|
Arfvidsson J, Ahlin F, Vargas KG, Thaler B, Wojta J, Huber K. Monocyte subsets in myocardial infarction: A review. Int J Cardiol 2016; 231:47-53. [PMID: 28062145 DOI: 10.1016/j.ijcard.2016.12.182] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/19/2016] [Revised: 11/07/2016] [Accepted: 12/27/2016] [Indexed: 01/12/2023]
Abstract
BACKGROUND Monocytes form an important part of the human innate immune system by taking part in inflammatory reactions. With time, monocytes have gained interest in the role they may play during the event of myocardial infarction (MI). The current paradigm suggests that monocytes consist of three subdivisions which differ in phenotypic and dynamic patterns after an MI. In the inflammation that ensues, the different subsets have been shown to have an impact on reparative processes and patient recovery. METHODS & RESULTS We searched Medline and Embase until April 5, 2016, for observational studies or clinical trials regarding monocyte functions and dynamics in MI. Apart from studies in humans, extensive work has been done in mice in an effort to understand the complex nature of monocyte dynamics. Animal models might add useful information on mapping these processes. CONCLUSION The question still remains whether animal data can, to a certain degree, be extrapolated to monocyte functions during human MI. This review aims to summarize current available evidence on both mice and men with particular focus on the understanding of monocyte subsets dynamics and effects in human MI.
Collapse
Affiliation(s)
- John Arfvidsson
- 3rd Department of Medicine, Cardiology and Intensive Care Medicine, Wilhelminen Hospital, Vienna, Austria; Faculty of Medicine and Health Sciences, Linköping University, Linköping, Sweden.
| | - Fredrik Ahlin
- 3rd Department of Medicine, Cardiology and Intensive Care Medicine, Wilhelminen Hospital, Vienna, Austria; Faculty of Medicine and Health Sciences, Linköping University, Linköping, Sweden
| | - Kris G Vargas
- 3rd Department of Medicine, Cardiology and Intensive Care Medicine, Wilhelminen Hospital, Vienna, Austria
| | - Barbara Thaler
- Department of Internal Medicine II, Medical University of Vienna, Vienna, Austria
| | - Johann Wojta
- Department of Internal Medicine II, Medical University of Vienna, Vienna, Austria; Ludwig Boltzmann Cluster for Cardiovascular Research, Vienna, Austria
| | - Kurt Huber
- 3rd Department of Medicine, Cardiology and Intensive Care Medicine, Wilhelminen Hospital, Vienna, Austria; Ludwig Boltzmann Cluster for Cardiovascular Research, Vienna, Austria; Sigmund Freud Private University, Medical School, Vienna, Austria
| |
Collapse
|
40
|
Wildgruber M, Aschenbrenner T, Wendorff H, Czubba M, Glinzer A, Haller B, Schiemann M, Zimmermann A, Berger H, Eckstein HH, Meier R, Wohlgemuth WA, Libby P, Zernecke A. The "Intermediate" CD14 ++CD16 + monocyte subset increases in severe peripheral artery disease in humans. Sci Rep 2016; 6:39483. [PMID: 27991581 PMCID: PMC5171878 DOI: 10.1038/srep39483] [Citation(s) in RCA: 73] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2016] [Accepted: 11/23/2016] [Indexed: 11/20/2022] Open
Abstract
Monocytes are key players in atherosclerotic. Human monocytes display a considerable heterogeneity and at least three subsets can be distinguished. While the role of monocyte subset heterogeneity has already been well investigated in coronary artery disease (CAD), the knowledge about monocytes and their heterogeneity in peripheral artery occlusive disease (PAOD) still is limited. Therefore, we aimed to investigate monocyte subset heterogeneity in patients with PAOD. Peripheral blood was obtained from 143 patients suffering from PAOD (Rutherford stage I to VI) and three monocyte subsets were identified by flow cytometry: CD14++CD16− classical monocytes, CD14+CD16++ non-classical monocytes and CD14++CD16+ intermediate monocytes. Additionally the expression of distinct surface markers (CD106, CD162 and myeloperoxidase MPO) was analyzed. Proportions of CD14++CD16+ intermediate monocyte levels were significantly increased in advanced stages of PAOD, while classical and non-classical monocytes displayed no such trend. Moreover, CD162 and MPO expression increased significantly in intermediate monocyte subsets in advanced disease stages. Likewise, increased CD162 and MPO expression was noted in CD14++CD16− classical monocytes. These data suggest substantial dynamics in monocyte subset distributions and phenotypes in different stages of PAOD, which can either serve as biomarkers or as potential therapeutic targets to decrease the inflammatory burden in advanced stages of atherosclerosis.
Collapse
Affiliation(s)
- Moritz Wildgruber
- Institut für diagnostische und interventionelle Radiologie, Klinikum rechts der Isar, Technische Universität München, Germany.,Institut für Klinische Radiologie, Universitätsklinikum Münster, Germany
| | - Teresa Aschenbrenner
- Institut für diagnostische und interventionelle Radiologie, Klinikum rechts der Isar, Technische Universität München, Germany
| | - Heiko Wendorff
- Klinik für vaskuläre und endovaskuläre Chirurgie, Klinikum rechts der Isar, Technische Universität München, Germany
| | - Maria Czubba
- Institut für diagnostische und interventionelle Radiologie, Klinikum rechts der Isar, Technische Universität München, Germany
| | - Almut Glinzer
- Institut für diagnostische und interventionelle Radiologie, Klinikum rechts der Isar, Technische Universität München, Germany.,Klinik für vaskuläre und endovaskuläre Chirurgie, Klinikum rechts der Isar, Technische Universität München, Germany
| | - Bernhard Haller
- Institut für medizinische Statistik und Epidemiologie, Klinikum rechts der Isar, Technische Universität München, Germany
| | - Matthias Schiemann
- Institut für medizinische Mikrobiologie, Immunologie und Hygiene, Technische Universität München, Germany.,Klinische Kooperationsgemeinschaft "Immunmonitoring", Helmholtz Zentrum München (Neuherberg) und Technische Universität München, Germany
| | - Alexander Zimmermann
- Klinik für vaskuläre und endovaskuläre Chirurgie, Klinikum rechts der Isar, Technische Universität München, Germany
| | - Hermann Berger
- Institut für diagnostische und interventionelle Radiologie, Klinikum rechts der Isar, Technische Universität München, Germany
| | - Hans-Henning Eckstein
- Klinik für vaskuläre und endovaskuläre Chirurgie, Klinikum rechts der Isar, Technische Universität München, Germany
| | - Reinhard Meier
- Institut für Radiologie, Universitätsklinikum Ulm, Germany
| | | | - Peter Libby
- Cardiovascular Division, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, USA
| | - Alma Zernecke
- Institut für Klinische Biochemie und Pathobiochemie, Universitätsklinikum Würzburg, Germany
| |
Collapse
|
41
|
Kratz JD, Chaddha A, Bhattacharjee S, Goonewardena SN. Atherosclerosis and Nanotechnology: Diagnostic and Therapeutic Applications. Cardiovasc Drugs Ther 2016; 30:33-9. [PMID: 26809711 DOI: 10.1007/s10557-016-6649-2] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Over the past several decades, tremendous advances have been made in the understanding, diagnosis, and treatment of coronary artery disease (CAD). However, with shifting demographics and evolving risk factors we now face new challenges that must be met in order to further advance are management of patients with CAD. In parallel with advances in our mechanistic appreciation of CAD and atherosclerosis, nanotechnology approaches have greatly expanded, offering the potential for significant improvements in our diagnostic and therapeutic management of CAD. To realize this potential we must go beyond to recognize new frontiers including knowledge gaps between understanding atherosclerosis to the translation of targeted molecular tools. This review highlights nanotechnology applications for imaging and therapeutic advancements in CAD.
Collapse
Affiliation(s)
- Jeremy D Kratz
- Michigan Nanotechnology Institute for Medicine and Biological Sciences, Ann Arbor, MI, 48109, USA.,Department of Internal Medicine, University of Wisconsin-Madison, Madison, WI, 53705, USA
| | - Ashish Chaddha
- Department of Internal Medicine, University of Wisconsin-Madison, Madison, WI, 53705, USA
| | - Somnath Bhattacharjee
- Michigan Nanotechnology Institute for Medicine and Biological Sciences, Ann Arbor, MI, 48109, USA.,Division of Cardiovascular Medicine, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Sascha N Goonewardena
- Michigan Nanotechnology Institute for Medicine and Biological Sciences, Ann Arbor, MI, 48109, USA. .,Division of Cardiovascular Medicine, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, 48109, USA.
| |
Collapse
|
42
|
Brilliant violet fluorochromes in simultaneous multicolor flow cytometry-fluorescence in situ hybridization measurement of monocyte subsets and telomere length in heart failure. J Transl Med 2016; 96:1223-1230. [PMID: 27617397 DOI: 10.1038/labinvest.2016.100] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2016] [Revised: 08/18/2016] [Accepted: 08/22/2016] [Indexed: 12/28/2022] Open
Abstract
Conventional analytical methods to determine telomere length (TL) have been replaced by more precise and reproducible procedures, such as fluorescence in situ hybridization coupled with flow cytometry (flow-FISH). However, simultaneous measurement of TL and cell phenotype remains difficult. Relatively expensive and time-consuming cell-sorting purification is needed to counteract the loss, due to stringent FISH conditions, of prehybridization fluorescence by the organic fluorochromes conventionally used in the phenotyping step. Here, we sought to assess whether the newly developed Brilliant Violet (BV) dyes are valuable to specifically and simultaneously assess the distribution and telomere attrition of monocyte subsets circulating in the blood of a cohort of patients with heart failure. We performed flow-FISH on blood samples from 28 patients with heart failure. To differentiate among monocyte subsets, we used BV and conventional fluorochromes conjugated to antibodies against CD86, CD14, CD16, and CD15. We simultaneously assessed the TLs of the monocyte subsets with a telomere-specific peptide nucleic acid probe labeled with fluorescein isothiocyanate. The BV dyes completely tolerated the harsh conditions required for adequate DNA denaturation and simultaneously provided accurate identification of monocyte subpopulations and respective TLs. The presented protocol may be faster and less expensive than those used currently for purposes such as establishing associations among patient categories, disease progression, monocyte heterogeneity, and aging in the context of heart failure.
Collapse
|
43
|
Pardali E, Schmitz T, Borgscheiper A, Iking J, Stegger L, Waltenberger J. Cryopreservation of primary human monocytes does not negatively affect their functionality or their ability to be labelled with radionuclides: basis for molecular imaging and cell therapy. EJNMMI Res 2016; 6:77. [PMID: 27778311 PMCID: PMC5078113 DOI: 10.1186/s13550-016-0232-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2016] [Accepted: 10/13/2016] [Indexed: 12/24/2022] Open
Abstract
Background Circulating white blood cells crucially contribute to maintenance and repair of solid organs. Therefore, certain cell populations such as monocytes are attractive targets for use in molecular imaging and cell imaging, e.g. after labelling with radionuclides, as well as for cell therapies. However, the preparation of monocytes may require freezing and thawing to preserve cells for timely and standardised applications. Additional modifications of these cells such as radioisotope labelling are necessary prior to their application in vivo. We therefore tested the hypothesis whether cryopreservation of freshly isolated circulating human monocytes affects their functional phenotype or their suitability for radionuclide labelling. Results CD14+CD16− monocytes were isolated from human peripheral blood. They were either directly used for cellular assays and labelling or frozen down using cryoprotectants. In the latter case, cells were thawed prior to further use and analysed for survival, chemotactic responses to various growth factors and adhesion on endothelial cells. In addition, both fresh and cryopreserved monocytes were labelled with radiotracers followed by assessment of survival and chemotactic responses. In all functional assays performed, cryopreserved monocytes did not significantly differ from freshly isolated monocytes with regard to their functionality. Cryopreservation did not affect cell survival. There was no effect on the chemotactic response of monocytes towards different growth factors. Likewise, adhesion properties remained unchanged following cryopreservation. Moreover, the labelling efficiency was similar for freshly isolated and cryopreserved monocytes. Labelling did not negatively affect monocyte survival and function. Conclusions Our data indicate that cryopreservation of freshly isolated human primary monocytes is feasible and does not negatively affect their functionality when used for labelling and functional assessment.
Collapse
Affiliation(s)
- Evangelia Pardali
- Department of Cardiovascular Medicine, University Hospital Münster, Albert-Schweitzer-Campus 1, Building A1, 48149, Münster, Germany
| | - Timo Schmitz
- Department of Cardiovascular Medicine, University Hospital Münster, Albert-Schweitzer-Campus 1, Building A1, 48149, Münster, Germany
| | - Andreas Borgscheiper
- Department of Cardiovascular Medicine, University Hospital Münster, Albert-Schweitzer-Campus 1, Building A1, 48149, Münster, Germany
| | - Janette Iking
- Department of Cardiovascular Medicine, University Hospital Münster, Albert-Schweitzer-Campus 1, Building A1, 48149, Münster, Germany
| | - Lars Stegger
- Department of Nuclear Medicine, University Hospital Münster, 48149, Münster, Germany
| | - Johannes Waltenberger
- Department of Cardiovascular Medicine, University Hospital Münster, Albert-Schweitzer-Campus 1, Building A1, 48149, Münster, Germany. .,Cells-in-Motion Cluster of Excellence (EXC 1003 - CiM), University of Münster, 48149, Münster, Germany.
| |
Collapse
|
44
|
Wildgruber M, Czubba M, Aschenbrenner T, Wendorff H, Hapfelmeier A, Glinzer A, Schiemann M, Zimmermann A, Eckstein HH, Berger H, Wohlgemuth WA, Meier R, Libby P, Zernecke A. Increased intermediate CD14 ++CD16 ++ monocyte subset levels associate with restenosis after peripheral percutaneous transluminal angioplasty. Atherosclerosis 2016; 253:128-134. [PMID: 27615596 DOI: 10.1016/j.atherosclerosis.2016.09.002] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/06/2016] [Revised: 08/31/2016] [Accepted: 09/01/2016] [Indexed: 11/17/2022]
Abstract
BACKGROUND AND AIMS We aimed at studying the association of three major human monocyte subsets after percutaneous transluminal angioplasty (PTA) in patients with femoropopliteal disease. METHODS We prospectively studied 67 sequential patients (40 male, 27 female; mean age 71 ± 11 years) treated with femoropopliteal angioplasty. Multi-color flow cytometry characterized monocyte subsets from venous blood for expression of CD14 and CD16 and intracellular myeloperoxidase (MPO) prior to, and 3, 6 and 12 months post PTA. Analyses tested associations between monocyte subsets and risk for restenosis. RESULTS 16/67 patients (24%) developed restenosis within 12 months after PTA. Patients with hyperlipidemia had increased risk for restenosis (HR = 1.7, 95% CI 0.7-2.9, p = 0.001). Increased baseline monocytes associated with an increased risk of late restenosis (HR = 4.9, 95% CI: 1.3-18.6, p = 0.047). CD14++CD16++ 'intermediate' monocytes assessed at baseline, and after 3, 6, and 12 months significantly associated with the risk for subsequent restenosis: HR = 3.9 (95% CI: 2.4-6.5, p = 0.029), HR = 5.7 (95% CI = 0.7-44.7, p = 0.013), HR = 6.5 (95% CI: 2.5-16.9, p = 0.001) and HR = 1.5 (95% CI = 1.4-15.5 p = 0.001), respectively. Moreover, the probability for freedom of restenosis decreased with increased levels of intermediate subsets at 12 months after PTA. Additionally, intracellular MPO expression in CD14++CD16++ measured at 3, 6 and 12 months associated with an increased restenosis risk (HR = 1.5, 95% CI: 0.8-2.1, p = 0.214, HR = 1.9, 95% CI: 1.0-2.3 p = 0.051 and HR = 1.4, 95% CI: 1.0-1.8, p = 0.052). CONCLUSIONS Our results imply altered innate immunity after angioplasty. Elevated CD14++CD16++ intermediate monocyte frequencies and increased MPO expression may identify individuals at heightened risk for restenosis.
Collapse
Affiliation(s)
- Moritz Wildgruber
- Institut für diagnostische und interventionelle Radiologie, Klinikum rechts der Isar, Technische Universität München, Germany; Institut für klinische Radiologie, Universitätsklinikum Münster, Germany.
| | - Maria Czubba
- Institut für diagnostische und interventionelle Radiologie, Klinikum rechts der Isar, Technische Universität München, Germany
| | - Teresa Aschenbrenner
- Institut für diagnostische und interventionelle Radiologie, Klinikum rechts der Isar, Technische Universität München, Germany
| | - Heiko Wendorff
- Klinik für vaskuläre und endovaskuläre Chirurgie, Klinikum rechts der Isar, Technische Universität München, Germany
| | - Alexander Hapfelmeier
- Institut für Medizinische Statistik und Epidemiologie, Klinikum rechts der Isar, Technische Universität München, Germany
| | - Almut Glinzer
- Institut für diagnostische und interventionelle Radiologie, Klinikum rechts der Isar, Technische Universität München, Germany; Klinik für vaskuläre und endovaskuläre Chirurgie, Klinikum rechts der Isar, Technische Universität München, Germany
| | - Matthias Schiemann
- Institut für medizinische Mikrobiologie, Immunologie und Hygiene, Technische Universität München, Germany; Klinische Kooperationsgemeinschaft, "Immunmonitoring", Helmholtz Zentrum München und Technische Universität München, München, Germany
| | - Alexander Zimmermann
- Klinik für vaskuläre und endovaskuläre Chirurgie, Klinikum rechts der Isar, Technische Universität München, Germany
| | - Hans-Henning Eckstein
- Klinik für vaskuläre und endovaskuläre Chirurgie, Klinikum rechts der Isar, Technische Universität München, Germany
| | - Hermann Berger
- Institut für diagnostische und interventionelle Radiologie, Klinikum rechts der Isar, Technische Universität München, Germany
| | | | - Reinhard Meier
- Klinik für diagnostische und interventionelle Radiologie, Universitätsklinikum Ulm, Germany
| | - Peter Libby
- Cardiovascular Division, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, USA
| | - Alma Zernecke
- Institut für Klinische Biochemie und Pathobiochemie, Universitätsklinikum Würzburg, Germany
| |
Collapse
|
45
|
Li D, Liu Y, Zhang X, Lv H, Pang W, Sun X, Gan LM, Hammock BD, Ai D, Zhu Y. Inhibition of soluble epoxide hydrolase alleviated atherosclerosis by reducing monocyte infiltration in Ldlr(-/-) mice. J Mol Cell Cardiol 2016; 98:128-37. [PMID: 27496380 DOI: 10.1016/j.yjmcc.2016.08.001] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/26/2016] [Revised: 07/05/2016] [Accepted: 08/01/2016] [Indexed: 01/08/2023]
Abstract
RATIONALE Circulating monocytes play pivotal roles in chronic inflammatory diseases. Epoxyeicosatrienoic acids (EETs), metabolites of arachidonic acid, are known to have anti-inflammatory effects and are hydrolyzed by soluble epoxide hydrolase (sEH). OBJECTIVE We aimed to investigate the effect of sEH inhibition in atherogenesis. METHODS AND RESULTS Mice with low-density lipoprotein receptor deficiency (Ldlr(-/-)) with or without sEH inhibitor, and Ldlr/sEH double-knockout (DK) mice were fed a Western-type diet (WTD) for 6weeks to induce arteriosclerosis. Both sEH inhibition and gene depletion decreased the WTD-induced hyperlipidemia, plaque area and macrophage infiltration in mice arterial wall. Ly6C(hi) infiltration of monocytes remained similar in blood, spleen and bone marrow of DK mice, but was decreased in aortic lesions. To further assess the role of sEH or EETs in monocyte/macrophage infiltration in atherogenesis, we transplanted DK bone marrow into Ldlr(-/-) recipients, and then fed mice the WTD. Aortic lesions and Ly6C(hi) monocyte infiltration were reduced in mice with transplanted bone marrow of DK mice without diminishing the cholesterol level. Furthermore, sEH inhibition or gene depletion increased the ratio of EETs/DHETs and diminished the expression of P-selectin glycoprotein ligand 1 (PSGL-1) in mice peripheral-blood mononuclear cells. Monocyte adhesion to P-selectin and to tumor necrosis factor α-activated endothelial cells was also diminished by sEH inhibition. CONCLUSION sEH inhibition and gene depletion attenuated atherosclerosis in mice by decreasing the infiltration of monocytes into the artery wall. EET and PSGL-1 may play pivotal roles in monocyte/macrophage infiltration and atherogenesis.
Collapse
Affiliation(s)
- Dan Li
- Department of Physiology and Pathophysiology, Peking University Health Science Center, Beijing 100191, China
| | - Yajin Liu
- Collaborative Innovation Center of Tianjin for Medical Epigenetics and Department of Physiology and Pathophysiology, Tianjin Medical University, Tianjin 300070, China
| | - Xu Zhang
- Collaborative Innovation Center of Tianjin for Medical Epigenetics and Department of Physiology and Pathophysiology, Tianjin Medical University, Tianjin 300070, China
| | - Huizhen Lv
- Collaborative Innovation Center of Tianjin for Medical Epigenetics and Department of Physiology and Pathophysiology, Tianjin Medical University, Tianjin 300070, China
| | - Wei Pang
- Department of Physiology and Pathophysiology, Peking University Health Science Center, Beijing 100191, China
| | - Xiaoli Sun
- Department of Physiology and Pathophysiology, Peking University Health Science Center, Beijing 100191, China
| | - Li-Ming Gan
- Innovative Medicines & Early Development, AstraZeneca R&D, SE-431 83 Mölndal, Sweden
| | - Bruce D Hammock
- Department of Entomology and Nematology UC Davis Comprehensive Cancer Center, University of California, Davis, Davis, CA 95616, USA
| | - Ding Ai
- Collaborative Innovation Center of Tianjin for Medical Epigenetics and Department of Physiology and Pathophysiology, Tianjin Medical University, Tianjin 300070, China
| | - Yi Zhu
- Department of Physiology and Pathophysiology, Peking University Health Science Center, Beijing 100191, China; Collaborative Innovation Center of Tianjin for Medical Epigenetics and Department of Physiology and Pathophysiology, Tianjin Medical University, Tianjin 300070, China.
| |
Collapse
|
46
|
Libby P, Nahrendorf M, Swirski FK. Leukocytes Link Local and Systemic Inflammation in Ischemic Cardiovascular Disease: An Expanded "Cardiovascular Continuum". J Am Coll Cardiol 2016; 67:1091-1103. [PMID: 26940931 DOI: 10.1016/j.jacc.2015.12.048] [Citation(s) in RCA: 257] [Impact Index Per Article: 28.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2015] [Revised: 12/01/2015] [Accepted: 12/14/2015] [Indexed: 12/23/2022]
Abstract
Physicians have traditionally viewed ischemic heart disease in a cardiocentric manner: plaques grow in arteries until they block blood flow, causing acute coronary and other ischemic syndromes. Recent research provides new insight into the integrative biology of inflammation as it contributes to ischemic cardiovascular disease. These results have revealed hitherto unsuspected inflammatory signaling networks at work in these disorders that link the brain, autonomic nervous system, bone marrow, and spleen to the atherosclerotic plaque and to the infarcting myocardium. A burgeoning clinical published data indicates that such inflammatory networks-far from a mere laboratory curiosity-operate in our patients and can influence aspects of ischemic cardiovascular disease that determine decisively clinical outcomes. These new findings enlarge the circle of the traditional "cardiovascular continuum" beyond the heart and vessels to include the nervous system, the spleen, and the bone marrow.
Collapse
Affiliation(s)
- Peter Libby
- Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts.
| | - Matthias Nahrendorf
- Center for Systems Biology, Massachusetts General Hospital, Boston, Massachusetts
| | - Filip K Swirski
- Center for Systems Biology, Massachusetts General Hospital, Boston, Massachusetts
| |
Collapse
|
47
|
Thaler B, Hohensinner PJ, Krychtiuk KA, Matzneller P, Koller L, Brekalo M, Maurer G, Huber K, Zeitlinger M, Jilma B, Wojta J, Speidl WS. Differential in vivo activation of monocyte subsets during low-grade inflammation through experimental endotoxemia in humans. Sci Rep 2016; 6:30162. [PMID: 27444882 PMCID: PMC4957086 DOI: 10.1038/srep30162] [Citation(s) in RCA: 73] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2015] [Accepted: 06/28/2016] [Indexed: 02/07/2023] Open
Abstract
Human monocytes are a heterogeneous cell population, which can be divided into a classical (CD14++CD16−), a non-classical (CD14+CD16+), and an intermediate (CD14++CD16+) subset. We hypothesized that low-grade inflammation may differentially affect monocyte subsets. We used a human lipopolysaccharide (LPS) infusion model to mimic low-grade inflammation to identify, which monocyte subsets are preferentially activated under these conditions. Monocyte subsets were identified by staining for CD14 and CD16, activation status of monocytes was analyzed by staining for CD11b and a novel in situ mRNA hybridization approach to detect IL-6 and IL-8 specific mRNA at the single-cell level by flow cytometry. After LPS challenge, cell numbers of monocyte subsets dropped after 2 h with cell numbers recovering after 6 h. Distribution of monocyte subsets was skewed dramatically towards the intermediate subset after 24 h. Furthermore, intermediate monocytes displayed the largest increase of CD11b expression after 2 h. Finally, IL-6 and IL-8 mRNA levels increased in intermediate and non-classical monocytes after 6 h whereas these mRNA levels in classical monocytes changed only marginally. In conclusion, our data indicates that the main responding subset of monocytes to standardized low-grade inflammation induced by LPS in humans is the CD14++CD16+ intermediate subset followed by the CD14+CD16+ non-classical monocyte subset. Circulating classical monocytes showed comparably less reaction to LPS challenge in vivo.
Collapse
Affiliation(s)
- B Thaler
- Department of Internal Medicine II, Medical University of Vienna, Vienna, Austria
| | - P J Hohensinner
- Department of Internal Medicine II, Medical University of Vienna, Vienna, Austria
| | - K A Krychtiuk
- Department of Internal Medicine II, Medical University of Vienna, Vienna, Austria.,Ludwig Boltzmann Cluster for Cardiovascular Research, Vienna, Austria
| | - P Matzneller
- Department of Clinical Pharmacology, Medical University of Vienna, Vienna, Austria
| | - L Koller
- Department of Internal Medicine II, Medical University of Vienna, Vienna, Austria
| | - M Brekalo
- Department of Internal Medicine II, Medical University of Vienna, Vienna, Austria
| | - G Maurer
- Department of Internal Medicine II, Medical University of Vienna, Vienna, Austria
| | - K Huber
- 3rd Medical Department for Cardiology and Emergency Medicine, Wilhelminen Hospital, Vienna, Austria
| | - M Zeitlinger
- Department of Clinical Pharmacology, Medical University of Vienna, Vienna, Austria
| | - B Jilma
- Department of Clinical Pharmacology, Medical University of Vienna, Vienna, Austria
| | - J Wojta
- Department of Internal Medicine II, Medical University of Vienna, Vienna, Austria.,Ludwig Boltzmann Cluster for Cardiovascular Research, Vienna, Austria.,Core Facilities, Medical University of Vienna, Vienna, Austria
| | - W S Speidl
- Department of Internal Medicine II, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
48
|
Crea F, Liuzzo G. Anti-inflammatory treatment of acute coronary syndromes: the need for precision medicine. Eur Heart J 2016; 37:2414-6. [PMID: 27252447 DOI: 10.1093/eurheartj/ehw207] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Affiliation(s)
- Filippo Crea
- Institute of Cardiology, Catholic University, Rome, Italy
| | | |
Collapse
|
49
|
Decano JL, Mattson PC, Aikawa M. Macrophages in Vascular Inflammation: Origins and Functions. Curr Atheroscler Rep 2016; 18:34. [PMID: 27125207 DOI: 10.1007/s11883-016-0585-2] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
|
50
|
Zhang P, Weaver JC, Chen G, Beretov J, Atsumi T, Qi M, Bhindi R, Qi JC, Madigan MC, Giannakopoulos B, Krilis SA. The Fifth Domain of Beta 2 Glycoprotein I Protects from Natural IgM Mediated Cardiac Ischaemia Reperfusion Injury. PLoS One 2016; 11:e0152681. [PMID: 27031114 PMCID: PMC4816326 DOI: 10.1371/journal.pone.0152681] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2015] [Accepted: 03/17/2016] [Indexed: 11/25/2022] Open
Abstract
Reperfusion after a period of ischemia results in reperfusion injury (IRI) which involves activation of the inflammatory cascade. In cardiac IRI, IgM natural antibodies (NAb) play a prominent role through binding to altered neoepitopes expressed on damaged cells. Beta 2 Glycoprotein I (β2GPI) is a plasma protein that binds to neoepitopes on damaged cells including anionic phospholipids through its highly conserved Domain V. Domain I of β2GPI binds circulating IgM NAbs and may provide a link between the innate immune system, IgM NAb binding and cardiac IRI. This study was undertaken to investigate the role of Β2GPI and its Domain V in cardiac IRI using wild-type (WT), Rag-1 -/- and β2GPI deficient mice. Compared with control, treatment with Domain V prior to cardiac IRI prevented binding of endogenous β2GPI to post-ischemic myocardium and resulted in smaller myocardial infarction size in both WT and β2GPI deficient mice. Domain V treatment in WT mice also resulted in less neutrophil infiltration, less apoptosis and improved ejection fraction at 24 h. Rag-1 -/- antibody deficient mice reconstituted with IgM NAbs confirmed that Domain V prevented IgM NAb induced cardiac IRI. Domain V remained equally effective when delivered at the time of reperfusion which has therapeutic clinical relevance.Based upon this study Domain V may function as a universal inhibitor of IgM NAb binding in the setting of cardiac IRI, which offers promise as a new therapeutic strategy in the treatment of cardiac IRI.
Collapse
Affiliation(s)
- Peng Zhang
- Department of Infectious Diseases, Immunology and Sexual Health, St George Hospital, Sydney, Australia
- Department of Medicine, University of New South Wales, Sydney, Australia
- Department of Cardiothoracic Surgery, Tianjin Medical University General Hospital, Tianjin, China
| | - James C. Weaver
- Department of Infectious Diseases, Immunology and Sexual Health, St George Hospital, Sydney, Australia
- Department of Medicine, University of New South Wales, Sydney, Australia
- Department of Cardiology, St George Hospital, Sydney, Australia
| | - Gang Chen
- Department of Infectious Diseases, Immunology and Sexual Health, St George Hospital, Sydney, Australia
- Department of Medicine, University of New South Wales, Sydney, Australia
| | - Julia Beretov
- Anatomical Pathology, SEALS St George Hospital, Sydney, Australia
| | - Tatsuya Atsumi
- Division of Rheumatology, Endocrinology and Nephrology, Hokkaido University Graduate School of Medicine, Sapporo, Japan
| | - Miao Qi
- Department of Infectious Diseases, Immunology and Sexual Health, St George Hospital, Sydney, Australia
- Department of Medicine, University of New South Wales, Sydney, Australia
| | - Ravinay Bhindi
- Department of Cardiology, Royal North Shore Hospital, Sydney, Australia
| | - Jian C. Qi
- Department of Infectious Diseases, Immunology and Sexual Health, St George Hospital, Sydney, Australia
- Department of Medicine, University of New South Wales, Sydney, Australia
| | - Michele C. Madigan
- Save Sight Institute, University of Sydney and Sydney Eye Hospital, Sydney, Australia
- School of Optometry and Vision Science, University of New South Wales, Sydney, Australia
| | - Bill Giannakopoulos
- Department of Infectious Diseases, Immunology and Sexual Health, St George Hospital, Sydney, Australia
- Department of Medicine, University of New South Wales, Sydney, Australia
- Department of Rheumatology, St George Hospital, Sydney, Australia
| | - Steven A. Krilis
- Department of Infectious Diseases, Immunology and Sexual Health, St George Hospital, Sydney, Australia
- Department of Medicine, University of New South Wales, Sydney, Australia
- * E-mail:
| |
Collapse
|