51
|
Taghizadeh E, Taheri F, Renani PG, Reiner Ž, Navashenaq JG, Sahebkar A. Macrophage: A Key Therapeutic Target in Atherosclerosis? Curr Pharm Des 2019; 25:3165-3174. [DOI: 10.2174/1381612825666190830153056] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2019] [Accepted: 08/22/2019] [Indexed: 12/19/2022]
Abstract
Background:
Atherosclerosis is a chronic inflammatory disease and a leading cause of coronary artery
disease, peripheral vascular disease and stroke. Lipid-laden macrophages are derived from circulating monocytes
and form fatty streaks as the first step of atherogenesis.
Methods:
An electronic search in major databases was performed to review new therapeutic opportunities for
influencing the inflammatory component of atherosclerosis based on monocytes/macrophages targeting.
Results:
In the past two decades, macrophages have been recognized as the main players in atherogenesis but also
in its thrombotic complications. There is a growing interest in immunometabolism and recent studies on metabolism
of macrophages have created new therapeutic options to treat atherosclerosis. Targeting recruitment, polarization,
cytokine profile extracellular matrix remodeling, cholesterol metabolism, oxidative stress, inflammatory
activity and non-coding RNAs of monocyte/macrophage have been proposed as potential therapeutic approaches
against atherosclerosis.
Conclusion:
Monocytes/macrophages have a crucial role in progression and pathogenesis of atherosclerosis.
Therefore, targeting monocyte/macrophage therapy in order to achieve anti-inflammatory effects might be a good
option for prevention of atherosclerosis.
Collapse
Affiliation(s)
- Eskandar Taghizadeh
- Cellular and Molecular Research Center, Yasuj University of Medical Sciences, Yasuj, Iran
| | - Forough Taheri
- Sharekord Branch, Islamic Azad University, Sharekord, Iran
| | | | - Željko Reiner
- University Hospital Centre Zagreb, School of Medicine, University of Zagreb, Department of Internal Medicine, Zagreb, Croatia
| | - Jamshid G. Navashenaq
- Immunogenetic and Cell Culture Department, Immunology Research Center, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | | |
Collapse
|
52
|
Bakogiannis C, Sachse M, Stamatelopoulos K, Stellos K. Platelet-derived chemokines in inflammation and atherosclerosis. Cytokine 2019; 122:154157. [DOI: 10.1016/j.cyto.2017.09.013] [Citation(s) in RCA: 92] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2017] [Revised: 07/31/2017] [Accepted: 09/11/2017] [Indexed: 12/16/2022]
|
53
|
Lebas H, Yahiaoui K, Martos R, Boulaftali Y. Platelets Are at the Nexus of Vascular Diseases. Front Cardiovasc Med 2019; 6:132. [PMID: 31572732 PMCID: PMC6749018 DOI: 10.3389/fcvm.2019.00132] [Citation(s) in RCA: 54] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2019] [Accepted: 08/23/2019] [Indexed: 12/17/2022] Open
Abstract
Platelets are important actors of cardiovascular diseases (CVD). Current antiplatelet drugs that inhibit platelet aggregation have been shown to be effective in CVD treatment. However, the management of bleeding complications is still an issue in vascular diseases. While platelets can act individually, they interact with vascular cells and leukocytes at sites of vascular injury and inflammation. The main goal remains to better understand platelet mechanisms in thrombo-inflammatory diseases and provide new lines of safe treatments. Beyond their role in hemostasis and thrombosis, recent studies have reported the role of several aspects of platelet functions in CVD progression. In this review, we will provide a comprehensive overview of platelet mechanisms involved in several vascular diseases.
Collapse
Affiliation(s)
- Héloïse Lebas
- Laboratory of Vascular Translational Science, U1148 Institut National de la Santé et de la Recherche Médicale (INSERM), Sorbonne Paris Cite, Univ Paris Diderot, Paris, France
| | - Katia Yahiaoui
- Laboratory of Vascular Translational Science, U1148 Institut National de la Santé et de la Recherche Médicale (INSERM), Sorbonne Paris Cite, Univ Paris Diderot, Paris, France
| | - Raphaël Martos
- Laboratory of Vascular Translational Science, U1148 Institut National de la Santé et de la Recherche Médicale (INSERM), Sorbonne Paris Cite, Univ Paris Diderot, Paris, France
| | - Yacine Boulaftali
- Laboratory of Vascular Translational Science, U1148 Institut National de la Santé et de la Recherche Médicale (INSERM), Sorbonne Paris Cite, Univ Paris Diderot, Paris, France
| |
Collapse
|
54
|
Factors Associated with RANTES Concentration in Cardiovascular Disease Patients. BIOMED RESEARCH INTERNATIONAL 2019; 2019:3026453. [PMID: 31396527 PMCID: PMC6668548 DOI: 10.1155/2019/3026453] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 04/10/2019] [Revised: 06/09/2019] [Accepted: 07/10/2019] [Indexed: 12/28/2022]
Abstract
Objective The aim of the study was to establish, by means of linear regressions analysis, whether RANTES and CCL2 have a relationship with age, sex, heart rate, ejection fraction, white blood cells count, monocyte count, platelet count, mean platelet volume, hsCRP concentration, creatinine and eGFR value, applied treatments, and coronary risk factors in polish cardiovascular disease patients. Methods Plasma chemokines concentrations were measured by ELISA method (R&D Systems Europe Ltd., Abingdon, England) in 115 cardiovascular disease patients (83 myocardial infarction/AMI and 32 stable angina/SA) and in the control group (N=25). Results Univariate linear regression analysis found that (1) for men mean RANTES plasma level is 1.56 times higher as compared to women; (2) if patient's age increases by 1 year, the mean RANTES concentration value increases by 1.4%; (3) if CCL2 concentration increases by 10 pg/mL, the mean RANTES concentration value increases by 3.3%; (4) if hsCRP concentration increases by 1 mg/L, the mean RANTES concentration value increases by 1.0%. By means of multiple linear regression analysis we found that (1) for men the mean plasma RANTES concentration value increases 1.89 times as compared to women; (2) if CCL2 concentration increases by 10 pg/mL, the mean RANTES concentration value increases by 3.4%; (3) if MPV increases by 1 fL, the mean RANTES concentration value increases by 12%, if other model parameters are fixed. For CCL2 we did not obtain statistically significant linear regression models. Conclusion Due to high variability of obtained CCL2 concentrations, it seems that RANTES better reflects the presence of the atherosclerotic lesion than CCL2. RANTES as a marker of atherosclerotic process may be an important therapeutic target, and the assessment of RANTES concentration should be interpreted depending on patient's sex, age, platelet hyperactivity state, hsCRP, and CCL2 concentration.
Collapse
|
55
|
van der Vorst EPC, Peters LJF, Müller M, Gencer S, Yan Y, Weber C, Döring Y. G-Protein Coupled Receptor Targeting on Myeloid Cells in Atherosclerosis. Front Pharmacol 2019; 10:531. [PMID: 31191301 PMCID: PMC6540917 DOI: 10.3389/fphar.2019.00531] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2019] [Accepted: 04/29/2019] [Indexed: 12/11/2022] Open
Abstract
Atherosclerosis, the underlying cause of the majority of cardiovascular diseases (CVDs), is a lipid-driven, inflammatory disease of the large arteries. Gold standard therapy with statins and the more recently developed proprotein convertase subtilisin/kexin type 9 (PCSK9) inhibitors have improved health conditions among CVD patients by lowering low density lipoprotein (LDL) cholesterol. Nevertheless, a substantial part of these patients is still suffering and it seems that 'just' lipid lowering is insufficient. The results of the Canakinumab Anti-inflammatory Thrombosis Outcome Study (CANTOS) have now proven that inflammation is a key driver of atherosclerosis and that targeting inflammation improves CVD outcomes. Therefore, the identification of novel drug targets and development of novel therapeutics that block atherosclerosis-specific inflammatory pathways have to be promoted. The inflammatory processes in atherosclerosis are facilitated by a network of immune cells and their subsequent responses. Cell networking is orchestrated by various (inflammatory) mediators which interact, bind and induce signaling. Over the last years, G-protein coupled receptors (GPCRs) emerged as important players in recognizing these mediators, because of their diverse functions in steady state but also and specifically during chronic inflammatory processes - such as atherosclerosis. In this review, we will therefore highlight a selection of these receptors or receptor sub-families mainly expressed on myeloid cells and their role in atherosclerosis. More specifically, we will focus on chemokine receptors, both classical and atypical, formyl-peptide receptors, the chemerin receptor 23 and the calcium-sensing receptor. When information is available, we will also describe the consequences of their targeting which may hold promising options for future treatment of CVD.
Collapse
Affiliation(s)
- Emiel P. C. van der Vorst
- Institute for Cardiovascular Prevention, Ludwig-Maximilians-University Munich, Munich, Germany
- Department of Pathology, Cardiovascular Research Institute Maastricht, Maastricht University Medical Centre, Maastricht, Netherlands
- Institute for Molecular Cardiovascular Research/Interdisciplinary Center for Clinical Research, RWTH Aachen University, Aachen, Germany
- Munich Heart Alliance, German Centre for Cardiovascular Research, Munich, Germany
| | - Linsey J. F. Peters
- Institute for Cardiovascular Prevention, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Madeleine Müller
- Institute for Cardiovascular Prevention, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Selin Gencer
- Institute for Cardiovascular Prevention, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Yi Yan
- Institute for Cardiovascular Prevention, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Christian Weber
- Institute for Cardiovascular Prevention, Ludwig-Maximilians-University Munich, Munich, Germany
- Munich Heart Alliance, German Centre for Cardiovascular Research, Munich, Germany
- Department of Biochemistry, Cardiovascular Research Institute Maastricht, Maastricht University Medical Centre, Maastricht, Netherlands
| | - Yvonne Döring
- Institute for Cardiovascular Prevention, Ludwig-Maximilians-University Munich, Munich, Germany
- Munich Heart Alliance, German Centre for Cardiovascular Research, Munich, Germany
| |
Collapse
|
56
|
Blin MG, Bachelier R, Fallague K, Moussouni K, Aurrand-Lions M, Fernandez S, Guillet B, Robert S, Foucault-Bertaud A, Bardin N, Blot-Chabaud M, Dignat-George F, Leroyer AS. CD146 deficiency promotes plaque formation in a mouse model of atherosclerosis by enhancing RANTES secretion and leukocyte recruitment. J Mol Cell Cardiol 2019; 130:76-87. [PMID: 30928429 DOI: 10.1016/j.yjmcc.2019.03.017] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/06/2018] [Revised: 02/26/2019] [Accepted: 03/25/2019] [Indexed: 11/18/2022]
Abstract
AIMS The progression of atherosclerosis is based on the continued recruitment of leukocytes in the vessel wall. The previously described role of CD146 in leukocyte infiltration suggests an involvement for this adhesion molecule in the inflammatory response. In this study, we investigated the role of CD146 in leukocyte recruitment by using an experimental model of atherogenesis. METHODS AND RESULTS The role of CD146 was explored in atherosclerosis by crossing CD146-/- mice with ApoE-/- mice. CD146 -/-/ApoE -/- and ApoE -/- mice were fed a Western diet for 24 weeks and were monitored for aortic wall thickness using high frequency ultrasound. The arterial wall was significantly thicker in CD146-deficient mice. After 24 weeks of Western diet, a significant increase of atheroma in both total aortic lesion and aortic sinus of CD146-null mice was observed. In addition, atherosclerotic lesions were more inflammatory since plaques from CD146-deficient mice contained more neutrophils and macrophages. This was due to up-regulation of RANTES secretion by macrophages in CD146-deficient atherosclerotic arteries. This prompted us to further address the function of CD146 in leukocyte recruitment during acute inflammation by using a second experimental model of peritonitis induced by thioglycollate. Neutrophil recruitment was significantly increased in CD146-deficient mice 12 h after peritonitis induction and associated with higher RANTES levels in the peritoneal cavity. In CD146-null macrophages, we also showed that increased RANTES production was dependent on constitutive inhibition of the p38-MAPK signaling pathway. Finally, Maraviroc, a RANTES receptor antagonist, was able to reduce atherosclerotic lesions and neutrophilia in CD146-deficient mice to the same level as that found in ApoE -/- mice. CONCLUSIONS Our data indicate that CD146 deficiency is associated with the upregulation of RANTES production and increased inflammation of atheroma, which could influence the atherosclerotic plaque fate. Thus, these data identify CD146 agonists as potential new therapeutic candidates for atherosclerosis treatment.
Collapse
Affiliation(s)
- Muriel G Blin
- Aix-Marseille Univ., INSERM 1263, INRA 1260, C2VN, Marseille, France
| | - Richard Bachelier
- Aix-Marseille Univ., INSERM 1263, INRA 1260, C2VN, Marseille, France
| | - Karim Fallague
- Aix-Marseille Univ., INSERM 1263, INRA 1260, C2VN, Marseille, France
| | - Karima Moussouni
- Aix-Marseille Univ., INSERM 1263, INRA 1260, C2VN, Marseille, France
| | - Michel Aurrand-Lions
- Aix Marseille Univ., CNRS, INSERM, Institut Paoli-Calmettes, CRCM, Marseille, France
| | - Samantha Fernandez
- Aix-Marseille Univ., CERIMED, Secteur Nucléaire Pré-clinique, Timone, 13005 Marseille, France
| | - Benjamin Guillet
- Aix-Marseille Univ., INSERM 1263, INRA 1260, C2VN, Marseille, France; Aix-Marseille Univ., CERIMED, Secteur Nucléaire Pré-clinique, Timone, 13005 Marseille, France; Assistance Publique-Hôpitaux de Marseille, Hôpital Nord, Marseille, France
| | - Stéphane Robert
- Aix-Marseille Univ., INSERM 1263, INRA 1260, C2VN, Marseille, France
| | | | - Nathalie Bardin
- Aix-Marseille Univ., INSERM 1263, INRA 1260, C2VN, Marseille, France; Assistance Publique-Hôpitaux de Marseille, Hôpital de la Conception, 13385 Marseille, France
| | | | - Françoise Dignat-George
- Aix-Marseille Univ., INSERM 1263, INRA 1260, C2VN, Marseille, France; Assistance Publique-Hôpitaux de Marseille, Hôpital de la Conception, 13385 Marseille, France
| | - Aurélie S Leroyer
- Aix-Marseille Univ., INSERM 1263, INRA 1260, C2VN, Marseille, France.
| |
Collapse
|
57
|
Chuang LP, Chen NH, Lin SW, Hu HC, Kao KC, Li LF, Yang CT, Huang CC, Pang JHS. Monocytic C-C chemokine receptor 5 expression increases in in vitro intermittent hypoxia condition and in severe obstructive sleep apnea patients. Sleep Breath 2019; 23:1177-1186. [PMID: 30778913 PMCID: PMC6867987 DOI: 10.1007/s11325-019-01797-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2018] [Revised: 01/26/2019] [Accepted: 01/31/2019] [Indexed: 12/14/2022]
Abstract
Purpose Obstructive sleep apnea (OSA) patients have higher risk of cardiovascular disease. C-C chemokine receptor 5 (CCR5), as an important receptor for monocyte recruitment and the initiation of atherosclerosis, was studied under intermittent hypoxia and in OSA patients. Methods The expression and function of CCR5 regulated by intermittent hypoxia in monocytic THP-1 cells were investigated in an in vitro intermittent hypoxia culture system. The expression levels of protein and mRNA were analyzed by western blot and RT/real-time PCR analysis. Cell adhesion assay and transwell filter migration assay were carried out to investigate the adhesion and chemotaxis of monocytes. In addition, the mRNA expression of CCR5 in monocytes isolated from peripheral blood of 72 adults was analyzed. Results Intermittent hypoxia upregulated the expression of CCR5 in THP-1 cells and enhanced the adhesion and chemotaxis of monocytes to vascular endothelial cells mediated by RANTES. The CCR5 expression induced by intermittent hypoxia was inhibited by inhibitor for p42/44 MAPK. Besides, the expression of CCR5 in monocytes increased along the AHI value especially in severe OSA patients that was statistically significant compared with mild and moderate OSA groups. Conclusions This study demonstrated the increased monocytic CCR5 gene expression in patients with severe OSA. Intermittent hypoxia, the characteristic of OSA, induced monocytic CCR5 gene expression and the enhanced RANTES-mediated chemotaxis and adhesion through p42/44 MAPK signal pathways.
Collapse
Affiliation(s)
- Li-Pang Chuang
- Sleep Center, Department of Pulmonary and Critical Care Medicine, Chang Gung Memorial Hospital, Taoyuan City, Taiwan
- School of Medicine, College of Medicine, Chang Gung University, Taoyuan City, Taiwan
| | - Ning-Hung Chen
- Sleep Center, Department of Pulmonary and Critical Care Medicine, Chang Gung Memorial Hospital, Taoyuan City, Taiwan
| | - Shih-Wei Lin
- Sleep Center, Department of Pulmonary and Critical Care Medicine, Chang Gung Memorial Hospital, Taoyuan City, Taiwan
| | - Han-Chung Hu
- Sleep Center, Department of Pulmonary and Critical Care Medicine, Chang Gung Memorial Hospital, Taoyuan City, Taiwan
| | - Kuo-Chin Kao
- Sleep Center, Department of Pulmonary and Critical Care Medicine, Chang Gung Memorial Hospital, Taoyuan City, Taiwan
| | - Li-Fu Li
- Sleep Center, Department of Pulmonary and Critical Care Medicine, Chang Gung Memorial Hospital, Taoyuan City, Taiwan
| | - Cheng-Ta Yang
- Sleep Center, Department of Pulmonary and Critical Care Medicine, Chang Gung Memorial Hospital, Taoyuan City, Taiwan
| | - Chung-Chi Huang
- Sleep Center, Department of Pulmonary and Critical Care Medicine, Chang Gung Memorial Hospital, Taoyuan City, Taiwan
| | - Jong-Hwei S Pang
- Graduate Institute of Clinical Medical Sciences, College of Medicine, Chang Gung University, Taoyuan City, Taiwan.
- Department of Physical Medicine and Rehabilitation, Chang Gung Memorial Hospital, Linkou, Taoyuan City, Taiwan.
| |
Collapse
|
58
|
Ahmad SF, Ansari MA, Nadeem A, Bakheet SA, Alotaibi MR, Alasmari AF, Alshammari MA, Al-Mazroua HA, Attia SM. DAPTA, a C-C chemokine receptor 5 (CCR5) antagonist attenuates immune aberrations by downregulating Th9/Th17 immune responses in BTBR T + Itpr3tf/J mice. Eur J Pharmacol 2019; 846:100-108. [PMID: 30658114 DOI: 10.1016/j.ejphar.2019.01.016] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2018] [Revised: 01/14/2019] [Accepted: 01/14/2019] [Indexed: 01/06/2023]
Abstract
Autism spectrum disorder (ASD) is a prevalent neurodevelopmental disorder characterized by deficits in social interaction, communication, and repetitive behaviors. BTBR T+ Itpr3tf/J (BTBR) mice, a preclinical autistic model featuring ASD symptoms as defined by social relations, was used in this study. We evaluated the potentially protective effect of D-Ala-peptide T-amide (DAPTA), a selective C-C chemokine receptor 5 (CCR5) antagonist, in BTBR mice. CCR5 is considered a potential therapeutic target in different neurodegenerative disorders. BTBR and C57 mice were intraperitoneally (i.p) treated with the DAPTA (0.01 mg/kg, i.p, once daily) for 7 days. We examined the effect of DAPTA by evaluating marble burying and administering repetitive behavior tests. We employed flow cytometry to assess the effect of DAPTA on CCR5+, CD4+CCR5+, CCR5+IL-6+, CCR5+IL-9+, CCR5+IL-17A+, CCR5+RORγT+, CCR5+IL-10+, and CCR5+Foxp3+ in spleen cells. We further explored the effects of DAPTA on IL-6, IL-9, IL-17A, RORγT, IL-10, and Foxp3 protein and mRNA expression levels in the brain tissues. DAPTA administration significantly decreased marble burying and repetitive behavior in BTBR mice. Additionally, DAPTA treatment inhibited CCR5+, CD4+CCR5+, CCR5+IL-6+, CCR5+IL-9+, CCR5+IL-17A+, CCR5+RORγT+, and upregulated CCR5+IL-10+, and CCR5+Foxp3+ production. We further observed that DAPTA downregulated IL-6, IL-9, IL-17A, and RORγT, and increased IL-10 and Foxp3 protein and mRNA expression. Therefore, our results suggest that DAPTA administration represents a potential treatment strategy for patients with ASD.
Collapse
Affiliation(s)
- Sheikh F Ahmad
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia.
| | - Mushtaq A Ansari
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Ahmed Nadeem
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Saleh A Bakheet
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Moureq R Alotaibi
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Abdullah F Alasmari
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Musaad A Alshammari
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Haneen A Al-Mazroua
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Sabry M Attia
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia; Department of Pharmacology and Toxicology, College of Pharmacy, Al-Azhar University, Cairo, Egypt
| |
Collapse
|
59
|
Jones DP, Patel J. Therapeutic Approaches Targeting Inflammation in Cardiovascular Disorders. BIOLOGY 2018; 7:biology7040049. [PMID: 30453474 PMCID: PMC6315639 DOI: 10.3390/biology7040049] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 10/23/2018] [Accepted: 11/06/2018] [Indexed: 12/22/2022]
Abstract
Cardiovascular disease is a leading cause of morbidity and mortality in the Western world and represents an enormous global health burden. Significant advances have been made in the conservative, medical and surgical management across the range of cardiovascular diseases however the inflammatory components of these diseases have traditionally been neglected. Inflammation is certainly a key component of atherosclerosis, a chronic inflammatory condition, but it is at least correlative and predictive of risk in many other aspects of cardiovascular medicine ranging from heart failure to outcomes following reperfusion strategies. Inflammation therefore represents significant potential for future risk stratification of patients as well as offering new therapeutic targets across cardiovascular medicine. This review explores the role of inflammation in several of the major aspects of cardiovascular medicine focusing on current and possible future examples of the targeting of inflammation in prognosis and therapy. It concludes that future directions of cardiovascular research and clinical practice should seek to identify cohorts of patients with a significant inflammatory component to their cardiovascular condition or reaction to cardiovascular intervention. These patients might benefit from therapeutic strategies mounted against the inflammatory components implicated in their condition.
Collapse
Affiliation(s)
- Daniel P Jones
- Bristol Medical School, University of Bristol, Canynge Hall, 39 Whatley Road, Bristol BS8 2PS, UK.
| | - Jyoti Patel
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, British Heart Foundation Centre of Research Excellence, University of Oxford, John Radcliffe Hospital, Oxford OX3 9DU, UK.
- Wellcome Trust Centre for Human Genetics, Radcliffe Department of Medicine, University of Oxford, Oxford OX3 7BN, UK.
| |
Collapse
|
60
|
Viafara-Garcia SM, Gualtero DF, Avila-Ceballos D, Lafaurie GI. Eikenella corrodens lipopolysaccharide stimulates the pro-atherosclerotic response in human coronary artery endothelial cells and monocyte adhesion. Eur J Oral Sci 2018; 126:476-484. [PMID: 30357941 DOI: 10.1111/eos.12580] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Eikenella corrodens is a gram-negative bacterium, and although primarily associated with periodontal infections or infective endocarditis, it has been identified in coronary atheromatous plaques. The effect of its lipopolysaccharide (LPS) on human coronary artery endothelial cells (HCAECs) is unknown. Our aim was to examine the mechanism underlying the inflammatory response in HCAECs stimulated with E. corrodens-LPS and to evaluate monocyte adhesion. Endothelial responses were determined by measuring the levels of chemokines and cytokines using flow cytometry. The surface expression of intercellular adhesion molecule 1 (ICAM-1) was determined using a cell-based ELISA, and the adhesion of THP-1 monocytes to HCAECs was also monitored. The involvement of toll-like receptors (TLRs) 2 and 4 was examined using TLR-neutralizing antibodies, and activation of extracellular signal-regulated kinase (ERK)1/2 and nuclear factor-kappa B (NF-κB) p65 were measured by western blotting and ELISA, respectively. Eikenella corrodens-LPS increased secretion of interleukin-8 (IL-8), monocyte chemotactic protein 1 (MCP-1), and granulocyte-macrophage colony-stimulating factor (GM-CSF), and expression of ICAM-1 on the surface of HCAECs, consistent with the increased adhesion of THP-1 cells. Moreover, E. corrodens-LPS interacted with TLR4, a key receptor able to maintain the levels of IL-8, MCP-1, and GM-CSF in HCAECs. Phosphorylation of ERK1/2 and activation of NF-κB p65 were also increased. The results indicate that E. corrodens-LPS activates HCAECs through TLR4, ERK, and NF-κB p65, triggering a pro-atherosclerotic endothelial response and enhancing monocyte adhesion.
Collapse
Affiliation(s)
- Sergio M Viafara-Garcia
- Unit of Basic Oral Investigation UIBO, School of Dentistry, Universidad El Bosque, Bogotá, Colombia
| | - Diego F Gualtero
- Unit of Basic Oral Investigation UIBO, School of Dentistry, Universidad El Bosque, Bogotá, Colombia
| | - Daniel Avila-Ceballos
- Unit of Basic Oral Investigation UIBO, School of Dentistry, Universidad El Bosque, Bogotá, Colombia
| | - Gloria I Lafaurie
- Unit of Basic Oral Investigation UIBO, School of Dentistry, Universidad El Bosque, Bogotá, Colombia
| |
Collapse
|
61
|
Pasquin S, Laplante V, Kouadri S, Milasan A, Mayer G, Tormo AJ, Savin V, Sharma M, Martel C, Gauchat JF. Cardiotrophin-like Cytokine Increases Macrophage–Foam Cell Transition. THE JOURNAL OF IMMUNOLOGY 2018; 201:2462-2471. [DOI: 10.4049/jimmunol.1800733] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/24/2018] [Accepted: 08/16/2018] [Indexed: 11/19/2022]
|
62
|
Platelet communication with the vascular wall: role of platelet-derived microparticles and non-coding RNAs. Clin Sci (Lond) 2018; 132:1875-1888. [PMID: 30185611 DOI: 10.1042/cs20180580] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2018] [Revised: 07/31/2018] [Accepted: 08/09/2018] [Indexed: 12/24/2022]
Abstract
Platelets play an important role in vascular homeostasis through their interaction with circulating blood cells as well as the vascular wall. Platelet-mediated communication with other cells can take the form of direct cell-cell interactions via membrane receptors or indirectly through the release of different soluble factors stored in their granules as well as through the release of microparticles. The latter carry different proteins and RNAs which are transferred to the target cells. The aim of this review is to discuss the role of platelet-derived factors, adhesion molecules as well as RNAs as mediators of the cross-talk between platelets and the vessel wall.
Collapse
|
63
|
Gonzalez LL, Garrie K, Turner MD. Type 2 diabetes - An autoinflammatory disease driven by metabolic stress. Biochim Biophys Acta Mol Basis Dis 2018; 1864:3805-3823. [PMID: 30251697 DOI: 10.1016/j.bbadis.2018.08.034] [Citation(s) in RCA: 64] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2018] [Accepted: 08/27/2018] [Indexed: 02/06/2023]
Abstract
Type 2 diabetes has traditionally been viewed as a metabolic disorder characterised by chronic high glucose levels, insulin resistance, and declining insulin secretion from the pancreas. Modern lifestyle, with abundant nutrient supply and reduced physical activity, has resulted in dramatic increases in the rates of obesity-associated disease conditions, including diabetes. The associated excess of nutrients induces a state of systemic low-grade chronic inflammation that results from production and secretion of inflammatory mediators from the expanded pool of activated adipocytes. Here, we review the mechanisms by which obesity induces adipose tissue dysregulation, detailing the roles of adipose tissue secreted factors and their action upon other cells and tissues central to glucose homeostasis and type 2 diabetes. Furthermore, given the emerging importance of adipokines, cytokines and chemokines in disease progression, we suggest that type 2 diabetes should now be viewed as an autoinflammatory disease, albeit one that is driven by metabolic dysregulation.
Collapse
Affiliation(s)
- Laura L Gonzalez
- Interdisciplinary Biomedical Research Centre, School of Science and Technology, Nottingham Trent University, Clifton, Nottingham NG11 8NS, United Kingdom
| | - Karin Garrie
- Interdisciplinary Biomedical Research Centre, School of Science and Technology, Nottingham Trent University, Clifton, Nottingham NG11 8NS, United Kingdom
| | - Mark D Turner
- Interdisciplinary Biomedical Research Centre, School of Science and Technology, Nottingham Trent University, Clifton, Nottingham NG11 8NS, United Kingdom.
| |
Collapse
|
64
|
Vajen T, Koenen RR, Werner I, Staudt M, Projahn D, Curaj A, Sönmez TT, Simsekyilmaz S, Schumacher D, Möllmann J, Hackeng TM, Hundelshausen PV, Weber C, Liehn EA. Blocking CCL5-CXCL4 heteromerization preserves heart function after myocardial infarction by attenuating leukocyte recruitment and NETosis. Sci Rep 2018; 8:10647. [PMID: 30006564 PMCID: PMC6045661 DOI: 10.1038/s41598-018-29026-0] [Citation(s) in RCA: 66] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2017] [Accepted: 06/29/2018] [Indexed: 12/13/2022] Open
Abstract
Myocardial infarction (MI) is a major cause of death in Western countries and finding new strategies for its prevention and treatment is thus of high priority. In a previous study, we have demonstrated a pathophysiologic relevance for the heterophilic interaction of CCL5 and CXCL4 in the progression of atherosclerosis. A specifically designed compound (MKEY) to block this CCL5-CXCR4 interaction is investigated as a potential therapeutic in a model of myocardial ischemia/reperfusion (I/R) damage. 8 week-old male C57BL/6 mice were intravenously treated with MKEY or scrambled control (sMKEY) from 1 day before, until up to 7 days after I/R. By using echocardiography and intraventricular pressure measurements, MKEY treatment resulted in a significant decrease in infarction size and preserved heart function as compared to sMKEY-treated animals. Moreover, MKEY treatment significantly reduced the inflammatory reaction following I/R, as revealed by specific staining for neutrophils and monocyte/macrophages. Interestingly, MKEY treatment led to a significant reduction of citrullinated histone 3 in the infarcted tissue, showing that MKEY can prevent neutrophil extracellular trap formation in vivo. Disrupting chemokine heterodimers during myocardial I/R might have clinical benefits, preserving the therapeutic benefit of blocking specific chemokines, and in addition, reducing the inflammatory side effects maintaining normal immune defence.
Collapse
Affiliation(s)
- Tanja Vajen
- Cardiovascular Research Institute Maastricht (CARIM), Department of Biochemistry, Maastricht University, Maastricht, The Netherlands
| | - Rory R Koenen
- Cardiovascular Research Institute Maastricht (CARIM), Department of Biochemistry, Maastricht University, Maastricht, The Netherlands.
- Institute for Cardiovascular Prevention (IPEK), LMU Munich, Munich, Germany.
| | - Isabella Werner
- Institute for Molecular Cardiovascular Research (IMCAR), RWTH Aachen University, Aachen, Germany
| | - Mareike Staudt
- Institute for Molecular Cardiovascular Research (IMCAR), RWTH Aachen University, Aachen, Germany
| | - Delia Projahn
- Institute for Cardiovascular Prevention (IPEK), LMU Munich, Munich, Germany
- Institute for Molecular Cardiovascular Research (IMCAR), RWTH Aachen University, Aachen, Germany
| | - Adelina Curaj
- Institute for Molecular Cardiovascular Research (IMCAR), RWTH Aachen University, Aachen, Germany
- Department of Experimental Molecular Imaging, RWTH Aachen University, Aachen, Germany
- Victor Babes National Institute of Pathology, Bucharest, Romania
| | - Tolga Taha Sönmez
- Institute for Molecular Cardiovascular Research (IMCAR), RWTH Aachen University, Aachen, Germany
- Department of Oral and Maxillofacial Surgery, Karlsruhe City Hospital of Freiburg University, Freiburg, Germany
- Department of Oral and Maxillofacial Surgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Sakine Simsekyilmaz
- Institute for Molecular Cardiovascular Research (IMCAR), RWTH Aachen University, Aachen, Germany
| | - David Schumacher
- Institute for Molecular Cardiovascular Research (IMCAR), RWTH Aachen University, Aachen, Germany
| | - Julia Möllmann
- Institute for Molecular Cardiovascular Research (IMCAR), RWTH Aachen University, Aachen, Germany
- Department of Cardiology, Pulmonology, Angiology and Intensive Care, University Hospital Aachen, Aachen, Germany
| | - Tilman M Hackeng
- Cardiovascular Research Institute Maastricht (CARIM), Department of Biochemistry, Maastricht University, Maastricht, The Netherlands
| | - Philipp von Hundelshausen
- Institute for Cardiovascular Prevention (IPEK), LMU Munich, Munich, Germany
- DZHK (German Centre for Cardiovascular Research), partner site Munich Heart Alliance, Munich, Germany
| | - Christian Weber
- Cardiovascular Research Institute Maastricht (CARIM), Department of Biochemistry, Maastricht University, Maastricht, The Netherlands
- Institute for Cardiovascular Prevention (IPEK), LMU Munich, Munich, Germany
- DZHK (German Centre for Cardiovascular Research), partner site Munich Heart Alliance, Munich, Germany
| | - Elisa A Liehn
- Institute for Molecular Cardiovascular Research (IMCAR), RWTH Aachen University, Aachen, Germany
- Department of Cardiology, Pulmonology, Angiology and Intensive Care, University Hospital Aachen, Aachen, Germany
- Human Genetic Laboratory, University of Medicine and Pharmacy, Craiova, Romania
| |
Collapse
|
65
|
Fernandez DM, Clemente JC, Giannarelli C. Physical Activity, Immune System, and the Microbiome in Cardiovascular Disease. Front Physiol 2018; 9:763. [PMID: 30013482 PMCID: PMC6036301 DOI: 10.3389/fphys.2018.00763] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2018] [Accepted: 05/30/2018] [Indexed: 12/23/2022] Open
Abstract
Cardiovascular health is a primary research focus, as it is a leading contributor to mortality and morbidity worldwide, and is prohibitively costly for healthcare. Atherosclerosis, the main driver of cardiovascular disease, is now recognized as an inflammatory disorder. Physical activity (PA) may have a more important role in cardiovascular health than previously expected. This review overviews the contribution of PA to cardiovascular health, the inflammatory role of atherosclerosis, and the emerging evidence of the microbiome as a regulator of inflammation.
Collapse
Affiliation(s)
- Dawn M. Fernandez
- Department of Medicine, Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Jose C. Clemente
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, United States
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Chiara Giannarelli
- Department of Medicine, Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, NY, United States
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, United States
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| |
Collapse
|
66
|
Sweet DR, Fan L, Hsieh PN, Jain MK. Krüppel-Like Factors in Vascular Inflammation: Mechanistic Insights and Therapeutic Potential. Front Cardiovasc Med 2018; 5:6. [PMID: 29459900 PMCID: PMC5807683 DOI: 10.3389/fcvm.2018.00006] [Citation(s) in RCA: 78] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2017] [Accepted: 01/17/2018] [Indexed: 12/19/2022] Open
Abstract
The role of inflammation in vascular disease is well recognized, involving dysregulation of both circulating immune cells as well as the cells of the vessel wall itself. Unrestrained vascular inflammation leads to pathological remodeling that eventually contributes to atherothrombotic disease and its associated sequelae (e.g., myocardial/cerebral infarction, embolism, and critical limb ischemia). Signaling events during vascular inflammation orchestrate widespread transcriptional programs that affect the functions of vascular and circulating inflammatory cells. The Krüppel-like factors (KLFs) are a family of transcription factors central in regulating vascular biology in states of homeostasis and disease. Given their abundance and diversity of function in cells associated with vascular inflammation, understanding the transcriptional networks regulated by KLFs will further our understanding of the pathogenesis underlying several pervasive health concerns (e.g., atherosclerosis, stroke, etc.) and consequently inform the treatment of cardiovascular disease. Within this review, we will discuss the role of KLFs in coordinating protective and deleterious responses during vascular inflammation, while addressing the potential targeting of these critical transcription factors in future therapies.
Collapse
Affiliation(s)
- David R Sweet
- Case Cardiovascular Research Institute, Case Western Reserve University, Harrington Heart and Vascular Institute, University Hospitals Cleveland Medical Center, Cleveland, OH, United States.,Department of Pathology, Case Western Reserve University, Cleveland, OH, United States
| | - Liyan Fan
- Case Cardiovascular Research Institute, Case Western Reserve University, Harrington Heart and Vascular Institute, University Hospitals Cleveland Medical Center, Cleveland, OH, United States.,Department of Pathology, Case Western Reserve University, Cleveland, OH, United States
| | - Paishiun N Hsieh
- Case Cardiovascular Research Institute, Case Western Reserve University, Harrington Heart and Vascular Institute, University Hospitals Cleveland Medical Center, Cleveland, OH, United States.,Department of Pathology, Case Western Reserve University, Cleveland, OH, United States
| | - Mukesh K Jain
- Case Cardiovascular Research Institute, Case Western Reserve University, Harrington Heart and Vascular Institute, University Hospitals Cleveland Medical Center, Cleveland, OH, United States
| |
Collapse
|
67
|
Goikuria H, Vandenbroeck K, Alloza I. Inflammation in human carotid atheroma plaques. Cytokine Growth Factor Rev 2018; 39:62-70. [PMID: 29396056 DOI: 10.1016/j.cytogfr.2018.01.006] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2017] [Accepted: 01/11/2018] [Indexed: 12/31/2022]
Abstract
Inflammation in carotid atherosclerotic plaque is linked to plaque rupture and cerebrovascular accidents. The balance between pro- and anti-inflammatory mediators governs development of the plaque, and may mediate enhancement of lesion broadening or, on the contrary, delay progression. In addition to macrophages and endothelial cells, smooth muscle cells (SMCs), which are the dominant cell subset in advanced plaques, are crucial players in carotid atherosclerosis development given their ability to differentiate into distinct phenotypes in reponse to specific signals received from the environment of the lesion. Carotid atheroma SMCs actively contribute to the inflammation in the lesion because of their acquired capacity to produce inflammatory mediators. We review the successive stages of carotid atheroma plaque formation via fatty streak early-stage toward more advanced rupture-prone lesions and document involvement of cytokines and chemokines and their cellular sources and targets in plaque progression and rupture.
Collapse
Affiliation(s)
- Haize Goikuria
- Neurogenomiks, Neuroscience Department, Faculty of Medicine and Odontology, Basque Country University (UPV/EHU), 48940 Leioa, Spain; ACHUCARRO, Basque Centre for Neuroscience, Science Park of the Basque Country University (UPV/EHU), SEDE Building, 3rd, 48940 Leioa, Spain
| | - Koen Vandenbroeck
- Neurogenomiks, Neuroscience Department, Faculty of Medicine and Odontology, Basque Country University (UPV/EHU), 48940 Leioa, Spain; ACHUCARRO, Basque Centre for Neuroscience, Science Park of the Basque Country University (UPV/EHU), SEDE Building, 3rd, 48940 Leioa, Spain; Ikerbasque, Basque Foundation for Science, 48013 Bilbao, Spain
| | - Iraide Alloza
- Neurogenomiks, Neuroscience Department, Faculty of Medicine and Odontology, Basque Country University (UPV/EHU), 48940 Leioa, Spain; ACHUCARRO, Basque Centre for Neuroscience, Science Park of the Basque Country University (UPV/EHU), SEDE Building, 3rd, 48940 Leioa, Spain.
| |
Collapse
|
68
|
Wolf D, Stachon P, Bode C, Zirlik A. Inflammatory mechanisms in atherosclerosis. Hamostaseologie 2017; 34:63-71. [DOI: 10.5482/hamo-13-09-0050] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2013] [Accepted: 12/05/2013] [Indexed: 01/13/2023] Open
Abstract
SummaryThroughout the last two decades inflammation has been recognized as the central mechanism underlying atherogenesis. A multitude of basic science work demonstrates the pivotal role of inflammatory processes during every step of atherosclerotic plaque formation: From initiation via propagation to complication.This review describes some of the key mechanisms involved with a particular focus on the diverse group of inflammatory cells and their subsets that distinctly contribute to atherogenic and anti-atherogenic phenomena. Furthermore, we summarize the controlling action of a tight network of co-stimulatory molecules and cytokines orchestrating the inflammatory and anti-inflammatory effector functions. Finally, the current status of clinical trials evaluating anti-inflammatory/ immune-modulatory treatment strategies is summarized and an outlook for future therapeutic implications is provided.
Collapse
|
69
|
Lv JX, Kong Q, Ma X. Current advances in circulating inflammatory biomarkers in atherosclerosis and related cardio-cerebrovascular diseases. Chronic Dis Transl Med 2017; 3:207-212. [PMID: 29354803 PMCID: PMC5747494 DOI: 10.1016/j.cdtm.2017.09.002] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2017] [Indexed: 12/30/2022] Open
Abstract
Atherosclerosis (AS) is a systemic chronic disease affecting both the coronary and cerebral arteries. Inflammation plays a key role in the initiation and progression of AS, and numerous inflammatory factors have been proposed as potential biomarkers. This article reviews recent research in studies on major circulating inflammatory biomarkers to identify surrogates that may reflect processes associated with AS development and the risk of AS-related vascular events, such as Von Willebrand factor, lectin-like oxidized low-density-lipoprotein receptor-1, soluble urokinase plasminogen activator receptor, regulated upon activation, normal T-cell expressed and secreted, and microparticles, which may provide new perspectives for clinical AS evaluation and risk stratification.
Collapse
Affiliation(s)
- Jun-Xuan Lv
- Department of Neurology, Xuanwu Hospital of Capital Medical University, National Clinical Research Center for Geriatric Disorders, Clinical Center for Cardio-cerebrovascular Disease Capital Medical University, Beijing 100053, China
| | - Qi Kong
- Department of Neurology, Xuanwu Hospital of Capital Medical University, National Clinical Research Center for Geriatric Disorders, Clinical Center for Cardio-cerebrovascular Disease Capital Medical University, Beijing 100053, China
| | - Xin Ma
- Department of Neurology, Xuanwu Hospital of Capital Medical University, National Clinical Research Center for Geriatric Disorders, Clinical Center for Cardio-cerebrovascular Disease Capital Medical University, Beijing 100053, China
| |
Collapse
|
70
|
Flierl U, Schäfer A. Fractalkine – a local inflammatory marker aggravating platelet activation at the vulnerable plaque. Thromb Haemost 2017; 108:457-63. [DOI: 10.1160/th12-04-0271] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2012] [Accepted: 05/30/2012] [Indexed: 01/10/2023]
Abstract
SummaryChemokines play an important role in inducing chemotaxis of cells, piloting white blood cells in immune surveillance and are crucial parts in the development and progression of atherosclerosis. Platelets are mandatory players in the initiation of atherosclerotic lesion formation and are susceptible targets for and producers of chemokines. Several chemokine receptors on platelets have been described previously, amongst them CX3CR1, the receptor for fractalkine. The unique chemokine fractalkine (CX3CL1, FKN) exists as a soluble as well as a membrane-anchored glycoprotein. Its essential role in the formation of atherosclerotic lesions and atherosclerosis progression has been impressively described in mouse models. Moreover, fractalkine induces platelet activation and adhesion via a functional fractalkine receptor (CX3CR1) expressed on the platelet surface. Platelet activation via the FKN/CX3CR1-axis triggers leukocyte adhesion to activated endothelium, and fractalkine-induced platelet P-selectin is mandatory for leukocyte recruitment under arterial flow conditions. This review summarises the role of fractalkine as a potential local inflammatory mediator which influences platelet activation in the setting of atherosclerosis. Beyond that, aspects of a potential interaction between fractalkine and platelet responsiveness to antiplatelet drugs are described. Furthermore, the possible impact of high-density lipoprotein cholesterol (HDL-C) on atherosclerosis progression, platelet activation and fractalkine signalling are discussed.
Collapse
|
71
|
Larsen SB, Grove EL, Würtz M, Neergaard-Petersen S, Hvas AM, Kristensen SD. The influence of low-grade inflammation on platelets in patients with stable coronary artery disease. Thromb Haemost 2017; 114:519-29. [DOI: 10.1160/th14-12-1007] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2014] [Accepted: 05/07/2015] [Indexed: 12/19/2022]
Abstract
SummaryInflammation is likely to be involved in all stages of atherosclerosis. Numerous inflammatory biomarkers are currently being studied, and even subtle increases in inflammatory biomarkers have been associated with increased risk of cardiovascular events in patients with coronary artery disease (CAD). Low-grade inflammation may influence both platelet production and platelet activation potentially leading to enhanced platelet aggregation. Thrombopoietin is considered the primary regulator of platelet production, but it likely acts in conjunction with numerous cytokines, of which many have altered levels in CAD. Previous studies have shown that high-sensitive C-reactive protein (CRP) independently predicts increased platelet aggregation in stable CAD patients. Increased levels of CRP, fibrinogen, interleukin-6, stromal cell-derived factor-1, CXC motif ligand 16, macrophage migration inhibitory factor, RANTES, calprotectin, and copeptin have been associated with increased risk of cardiovascular events in CAD patients. Additionally, some of these inflammatory markers have been associated with enhanced platelet activation and aggregation. However, CRP and other inflammatory markers provide only limited additional predictive value to classical risk factors such as smoking, blood pressure, and cholesterol levels. Existing data do not clarify whether inflammation simply accompanies CAD and increased production and aggregation of platelets, or whether a causal relationship exists. In this review, we provide a comprehensive overview of inflammatory markers in stable CAD with particular emphasis on platelet production, activation, and aggregation in CAD patients.
Collapse
|
72
|
Suffee N, Le Visage C, Hlawaty H, Aid-Launais R, Vanneaux V, Larghero J, Haddad O, Oudar O, Charnaux N, Sutton A. Pro-angiogenic effect of RANTES-loaded polysaccharide-based microparticles for a mouse ischemia therapy. Sci Rep 2017; 7:13294. [PMID: 29038476 PMCID: PMC5643514 DOI: 10.1038/s41598-017-13444-7] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2017] [Accepted: 08/09/2017] [Indexed: 12/20/2022] Open
Abstract
Peripheral arterial disease results from the chronic obstruction of arteries leading to critical hindlimb ischemia. The aim was to develop a new therapeutic strategy of revascularization by using biodegradable and biocompatible polysaccharides-based microparticles (MP) to treat the mouse hindlimb ischemia. For this purpose, we deliver the pro-angiogenic chemokine Regulated upon Activation, Normal T-cell Expressed and Secreted (RANTES)/CCL5 in the mouse ischemic hindlimb, in solution or incorporated into polysaccharide-based microparticles. We demonstrate that RANTES-loaded microparticles improve the clinical score, induce the revascularization and the muscle regeneration in injured mice limb. To decipher the mechanisms underlying RANTES effects in vivo, we demonstrate that RANTES increases the spreading, the migration of human endothelial progenitor cells (EPC) and the formation of vascular network. The main receptors of RANTES i.e. CCR5, syndecan-4 and CD44 expressed at endothelial progenitor cell surface are involved in RANTES-induced in vitro biological effects on EPC. By using two RANTES mutants, [E66A]-RANTES with impaired ability to oligomerize, and [44AANA47]-RANTES mutated in the main RANTES-glycosaminoglycan binding site, we demonstrate that both chemokine oligomerization and binding site to glycosaminoglycans are essential for RANTES-induced angiogenesis in vitro. Herein we improved the muscle regeneration and revascularization after RANTES-loaded MP local injection in mice hindlimb ischemia.
Collapse
Affiliation(s)
- N Suffee
- INSERM, U1148, Laboratory for Vascular Translational Science, UFR SMBH, Université Paris 13, Sorbonne Paris Cité, Bobigny, France
| | - C Le Visage
- Inserm, UMR 1229, RMeS, Regenerative Medicine and Skeleton, Université de Nantes, ONIRIS, Nantes, France
| | - H Hlawaty
- INSERM, U1148, Laboratory for Vascular Translational Science, UFR SMBH, Université Paris 13, Sorbonne Paris Cité, Bobigny, France
| | - R Aid-Launais
- INSERM, U1148, Laboratory for Vascular Translational Science, UFR SMBH, Université Paris 13, Sorbonne Paris Cité, Bobigny, France
| | - V Vanneaux
- APHP, Hôpital Saint-Louis, Unité de Thérapie Cellulaire, Paris, France; Université Paris Diderot, Sorbonne Paris Cité, F-75475, Paris, France.,Inserm UMR1160 et CIC de Biothérapies, Institut Universitaire d'Hématologie, Hôpital Saint-Louis, Paris, France
| | - J Larghero
- APHP, Hôpital Saint-Louis, Unité de Thérapie Cellulaire, Paris, France; Université Paris Diderot, Sorbonne Paris Cité, F-75475, Paris, France.,Inserm UMR1160 et CIC de Biothérapies, Institut Universitaire d'Hématologie, Hôpital Saint-Louis, Paris, France
| | - O Haddad
- INSERM, U1148, Laboratory for Vascular Translational Science, UFR SMBH, Université Paris 13, Sorbonne Paris Cité, Bobigny, France
| | - O Oudar
- INSERM, U1148, Laboratory for Vascular Translational Science, UFR SMBH, Université Paris 13, Sorbonne Paris Cité, Bobigny, France
| | - N Charnaux
- INSERM, U1148, Laboratory for Vascular Translational Science, UFR SMBH, Université Paris 13, Sorbonne Paris Cité, Bobigny, France.,Laboratoire de Biochimie, Hôpital Jean Verdier, AP-HP, Bondy, France
| | - A Sutton
- INSERM, U1148, Laboratory for Vascular Translational Science, UFR SMBH, Université Paris 13, Sorbonne Paris Cité, Bobigny, France. .,Laboratoire de Biochimie, Hôpital Jean Verdier, AP-HP, Bondy, France.
| |
Collapse
|
73
|
Nosalski R, Guzik TJ. Perivascular adipose tissue inflammation in vascular disease. Br J Pharmacol 2017; 174:3496-3513. [PMID: 28063251 PMCID: PMC5610164 DOI: 10.1111/bph.13705] [Citation(s) in RCA: 287] [Impact Index Per Article: 35.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2016] [Revised: 12/29/2016] [Accepted: 01/04/2017] [Indexed: 12/11/2022] Open
Abstract
Perivascular adipose tissue (PVAT) plays a critical role in the pathogenesis of cardiovascular disease. In vascular pathologies, perivascular adipose tissue increases in volume and becomes dysfunctional, with altered cellular composition and molecular characteristics. PVAT dysfunction is characterized by its inflammatory character, oxidative stress, diminished production of vaso-protective adipocyte-derived relaxing factors and increased production of paracrine factors such as resistin, leptin, cytokines (IL-6 and TNF-α) and chemokines [RANTES (CCL5) and MCP-1 (CCL2)]. These adipocyte-derived factors initiate and orchestrate inflammatory cell infiltration including primarily T cells, macrophages, dendritic cells, B cells and NK cells. Protective factors such as adiponectin can reduce NADPH oxidase superoxide production and increase NO bioavailability in the vessel wall, while inflammation (e.g. IFN-γ or IL-17) induces vascular oxidases and eNOS dysfunction in the endothelium, vascular smooth muscle cells and adventitial fibroblasts. All of these events link the dysfunctional perivascular fat to vascular dysfunction. These mechanisms are important in the context of a number of cardiovascular disorders including atherosclerosis, hypertension, diabetes and obesity. Inflammatory changes in PVAT's molecular and cellular responses are uniquely different from classical visceral or subcutaneous adipose tissue or from adventitia, emphasizing the unique structural and functional features of this adipose tissue compartment. Therefore, it is essential to develop techniques for monitoring the characteristics of PVAT and assessing its inflammation. This will lead to a better understanding of the early stages of vascular pathologies and the development of new therapeutic strategies focusing on perivascular adipose tissue. LINKED ARTICLES This article is part of a themed section on Molecular Mechanisms Regulating Perivascular Adipose Tissue - Potential Pharmacological Targets? To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v174.20/issuetoc.
Collapse
Affiliation(s)
- Ryszard Nosalski
- Institute of Cardiovascular and Medical SciencesUniversity of GlasgowScotlandUK
- Department of Internal and Agricultural MedicineJagiellonian University, Collegium MedicumKrakowPoland
| | - Tomasz J Guzik
- Institute of Cardiovascular and Medical SciencesUniversity of GlasgowScotlandUK
- Department of Internal and Agricultural MedicineJagiellonian University, Collegium MedicumKrakowPoland
| |
Collapse
|
74
|
Bahramsoltani R, Ebrahimi F, Farzaei MH, Baratpourmoghaddam A, Ahmadi P, Rostamiasrabadi P, Rasouli Amirabadi AH, Rahimi R. Dietary polyphenols for atherosclerosis: A comprehensive review and future perspectives. Crit Rev Food Sci Nutr 2017; 59:114-132. [PMID: 28812379 DOI: 10.1080/10408398.2017.1360244] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
Atherosclerosis is one of the most prevalent reasons for premature death in adults. Despite the several conventional drugs in the market; many patients are not completely treated. Here we comprehensively review current clinical evidence regarding the efficacy of dietary polyphenols in atherosclerosis and related complications. PubMed, Cochrane library and Scopus were searched from inception until August 2016 to obtain clinical trials in which polyphenols were evaluated in cardiovascular parameters related to atherosclerosis. From total of 13031 results, 49 clinical trials were finally included. Tyrosol derivatives from virgin olive oil, catechins and theaflavins from green and black tea, cocoa polyphenols, and red grape resveratrol, as well as anthocyanins were the most studied polyphenolic compounds which could regulate lipid profile, inflammation and oxidative stress, blood pressure, endothelial function, and cell adhesion molecules. The most important limitations of the included trials were small sample size, short follow up, and unqualified methodology. Future well-designed clinical trials are necessary to provide better level of evidence for clinical decision making.
Collapse
Affiliation(s)
- Roodabeh Bahramsoltani
- a Department of Traditional Pharmacy, School of Traditional Medicine , Tehran University of Medical Sciences , Tehran , Iran.,b PhytoPharmacology Interest Group (PPIG) , Universal Scientific Education and Research Network (USERN) , Tehran , Iran
| | - Farnaz Ebrahimi
- c Pharmacy Student's Research Committee, School of Pharmacy , Isfahan University of Medical Sciences , Isfahan , Iran
| | - Mohammad Hosein Farzaei
- d Pharmaceutical Sciences Research Center , Kermanshah University of Medical Sciences , Kermanshah , Iran.,e Medical Biology Research Center , Kermanshah University of Medical Sciences , Kermanshah , Iran
| | - Armaghan Baratpourmoghaddam
- c Pharmacy Student's Research Committee, School of Pharmacy , Isfahan University of Medical Sciences , Isfahan , Iran
| | - Pardis Ahmadi
- c Pharmacy Student's Research Committee, School of Pharmacy , Isfahan University of Medical Sciences , Isfahan , Iran
| | | | | | - Roja Rahimi
- a Department of Traditional Pharmacy, School of Traditional Medicine , Tehran University of Medical Sciences , Tehran , Iran.,b PhytoPharmacology Interest Group (PPIG) , Universal Scientific Education and Research Network (USERN) , Tehran , Iran
| |
Collapse
|
75
|
Abstract
Cardiovascular disease (CVD) is a major health problem globally. The high incidence and case fatality of CVD are, to a large extent, a consequence of its late diagnosis and lack of highly sensitive and specific markers. Only a very small number of biomarkers, such as troponin, detect late disease. There is some evidence of an association and dysregulation between specific cytokines in the pathogenesis of CVD. These molecules are involved in inflammatory and immune mechanisms associated with atherogenesis. Several molecular/cellular pathways that include STAT, MAPK, and SMAD are modulated by cytokines. Against this background, microRNAs (miRNAs) are a class of noncoding RNAs with important roles in pathological events, leading to atherosclerotic CVD. It has been shown that the latter could affect cytokine production and contribute to progression of atherosclerotic CVD. Moreover, modulation of miRNAs appears to inhibit cardiomyocyte apoptosis, attenuate infarct size, and reduce cardiac dysfunction. This review highlights several recent preclinical and clinical studies on the role of cytokines in CVD, novel miRNA-based therapeutic approaches for therapeutic intervention, and potential circulating cytokines that have promise as biomarkers in CVD.
Collapse
Affiliation(s)
- Hamed Mirzaei
- School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Gordon A Ferns
- Brighton & Sussex Medical School, Sussex, United Kingdom
| | - Amir Avan
- School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran; School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran; Cancer Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.
| | - Majid G Mobarhan
- School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran; Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
76
|
Personalized Medicine: New Perspectives for the Diagnosis and the Treatment of Renal Diseases. Int J Mol Sci 2017; 18:ijms18061248. [PMID: 28604601 PMCID: PMC5486071 DOI: 10.3390/ijms18061248] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2017] [Revised: 05/19/2017] [Accepted: 05/27/2017] [Indexed: 12/29/2022] Open
Abstract
The prevalence of renal diseases is rising and reaching 5-15% of the adult population. Renal damage is associated with disturbances of body homeostasis and the loss of equilibrium between exogenous and endogenous elements including drugs and metabolites. Studies indicate that renal diseases are influenced not only by environmental but also by genetic factors. In some cases the disease is caused by mutation in a single gene and at that time severity depends on the presence of one or two mutated alleles. In other cases, renal disease is associated with the presence of alteration within a gene or genes, but environmental factors are also necessary for the development of disease. Therefore, it seems that the analysis of genetic aspects should be a natural component of clinical and experimental studies. The goal of personalized medicine is to determine the right drug, for the right patient, at the right time. Whole-genome examinations may help to change the approach to the disease and the patient resulting in the creation of "personalized medicine" with new diagnostic and treatment strategies designed on the basis of genetic background of each individual. The identification of high-risk patients in pharmacogenomics analyses will help to avoid many unwarranted side effects while optimizing treatment efficacy for individual patients. Personalized therapies for kidney diseases are still at the preliminary stage mainly due to high costs of such analyses and the complex nature of human genome. This review will focus on several areas of interest: renal disease pathogenesis, diagnosis, treatment, rate of progression and the prediction of prognosis.
Collapse
|
77
|
Kufareva I, Gustavsson M, Zheng Y, Stephens BS, Handel TM. What Do Structures Tell Us About Chemokine Receptor Function and Antagonism? Annu Rev Biophys 2017; 46:175-198. [PMID: 28532213 PMCID: PMC5764094 DOI: 10.1146/annurev-biophys-051013-022942] [Citation(s) in RCA: 76] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Chemokines and their cell surface G protein-coupled receptors are critical for cell migration, not only in many fundamental biological processes but also in inflammatory diseases and cancer. Recent X-ray structures of two chemokines complexed with full-length receptors provided unprecedented insight into the atomic details of chemokine recognition and receptor activation, and computational modeling informed by new experiments leverages these insights to gain understanding of many more receptor:chemokine pairs. In parallel, chemokine receptor structures with small molecules reveal the complicated and diverse structural foundations of small molecule antagonism and allostery, highlight the inherent physicochemical challenges of receptor:chemokine interfaces, and suggest novel epitopes that can be exploited to overcome these challenges. The structures and models promote unique understanding of chemokine receptor biology, including the interpretation of two decades of experimental studies, and will undoubtedly assist future drug discovery endeavors.
Collapse
Affiliation(s)
- Irina Kufareva
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, La Jolla, California 92093; ,
| | - Martin Gustavsson
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, La Jolla, California 92093; ,
| | - Yi Zheng
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, La Jolla, California 92093; ,
| | - Bryan S Stephens
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, La Jolla, California 92093; ,
| | - Tracy M Handel
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, La Jolla, California 92093; ,
| |
Collapse
|
78
|
Jang YJ, Park B, Lee HW, Park HJ, Koo HJ, Kim BO, Sohn EH, Um SH, Pyo S. Sinigrin attenuates the progression of atherosclerosis in ApoE -/- mice fed a high-cholesterol diet potentially by inhibiting VCAM-1 expression. Chem Biol Interact 2017; 272:28-36. [PMID: 28483571 DOI: 10.1016/j.cbi.2017.05.006] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2017] [Accepted: 05/04/2017] [Indexed: 10/19/2022]
Abstract
Atherosclerosis is a complex inflammatory disease associated with elevated levels of atherogenic molecules for leukocyte recruitment. Sinigrin (2-propenylglucosinolate) is found mainly in broccoli, brussels sprouts, and black mustard seeds. Recently, sinigrin has received attention for its role in disease prevention and health promotion. In this study, we examined the effect of sinigrin on development of atherosclerosis in ApoE-/- mice and the expression of adhesion molecules in vascular smooth muscle cells (VSMCs). The serum concentrations of lactate dehydrogenase (LDH), triglyceride (TG), total cholesterol (TC), low density lipoprotein (LDL), calcium (Ca2+), and pro-inflammatory cytokines were reduced by sinigrin treatment in ApoE-/- mice. In addition, oral administration of sinigrin attenuated the mRNA expression of vascular cell adhesion molecule-1 (VCAM-1), intercellular adhesion molecule-1 (ICAM-1), C-C motif chemokine ligand 2 (CCL2), and CCL5 on aorta tissues and 3-hydroxy-3-methylglutaryl-coenzyme A reductase (HMGR), liver X receptor (LXR), sterol regulatory element-binding protein-2 (SREBP-2), and low density lipoprotein receptor (LDLR) on liver tissues in ApoE-/- mice. To provide a potential mechanism underlying the action of sinigrin, we evaluated the in vitro effect of sinigrin on the expression of the VCAM-1 in TNF-α-induced VSMCs. The increased expression of VCAM-1 by TNF-α stimulation was significantly suppressed by the treatment of sinigrin (1-100 μg/ml) and sinigrin inhibited the nuclear translocation of NF-κB and the phosphorylation of p38 MAPK and JNK pathways, suggesting that sinigrin decreases the TNF-α-stimulated VCAM-1 expression through the suppression of NF-κB and MAP kinases signaling pathways. Overall, sinigrin has the potential to be used in reducing the risks of atherosclerosis.
Collapse
Affiliation(s)
- Yeon Jeong Jang
- School of Pharmacy, Sungkyunkwan University, Suwon, 16419, Republic of Korea
| | - Bongkyun Park
- School of Pharmacy, Sungkyunkwan University, Suwon, 16419, Republic of Korea
| | - Hee-Weon Lee
- School of Pharmacy, Sungkyunkwan University, Suwon, 16419, Republic of Korea
| | - Hye Jin Park
- School of Pharmacy, Sungkyunkwan University, Suwon, 16419, Republic of Korea
| | - Hyun Jung Koo
- Department of Medicinal & Industrial Crops, Korea National College of Agriculture and Fisheries, Jeonju, 54874, Republic of Korea
| | - Byung Oh Kim
- School of Food Sciences & Biotechnology, College of Agriculture & Life Sciences, Kyungpook National University, Daegu, 41566, Republic of Korea
| | - Eun-Hwa Sohn
- Department of Herbal Medicine Resources, Kangwon National University, Samcheok, 25913, Republic of Korea
| | - Sung Hee Um
- Department of Molecular Cell Biology, School of Medicine, Sungkyunkwan University, Suwon, 16419, Republic of Korea.
| | - Suhkneung Pyo
- School of Pharmacy, Sungkyunkwan University, Suwon, 16419, Republic of Korea.
| |
Collapse
|
79
|
Feng X, Gao X, Jia Y, Zhang H, Xu Y. Atorvastatin Decreased Circulating RANTES Levels in Impaired Glucose Tolerance Patients with Hypercholesterolemia: An Interventional Study. Diabetes Ther 2017; 8:309-319. [PMID: 28120261 PMCID: PMC5380490 DOI: 10.1007/s13300-017-0227-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/19/2016] [Indexed: 01/27/2023] Open
Abstract
INTRODUCTION Impaired glucose tolerance (IGT) is the major cause of the development of both type 2 diabetes and atherosclerosis. Regulated upon activation, normal T cells expressed and secreted (RANTES), a proinflammatory chemokine, is associated with atherosclerosis. We investigated the effect of atorvastatin on circulating RANTES in IGT patients with hypercholesterolemia. METHODS This study evaluated cross-sectional and interventional studies of 32 IGT patients with hypercholesterolemia (group A) and 32 controls (group B). Group A was treated with atorvastatin (20 mg/day) for 8 weeks. Platelet-free plasma (PFP) RANTES and clinical characteristics were examined. RESULTS PFP RANTES was significantly higher in group A compared with group B (9.76 ± 3.10 vs 6.43 ± 2.16 ng/ml, P < 0.001). PFP RANTES was positively correlated with total cholesterol (TC) (r = 0.589, P < 0.001), low-density lipoprotein cholesterol (LDL-C) (r = 0.583, P < 0.001), triglycerides (TG) (r = 0.450, P < 0.001), fasting blood glucose (FBG) (r = 0.469, P < 0.001), 2-hour postchallenge glucose (2hPG) (r = 0.397, P = 0.001), glycosylated hemoglobin (HbA1c) (r = 0.353, P = 0.004), and high sensitivity C-reactive protein (hsCRP) (r = 0.616, P < 0.001), and negatively related to high-density lipoprotein cholesterol (HDL-C) (r = -0.272, P = 0.029). After controlling for confounders, LDL-C (β = 2.109, P < 0.001) and hsCRP (β = 0.272, P = 0.029) were independently related to RANTES. After atorvastatin treatment, PFP RANTES significantly decreased in group A compared with baseline (from 9.76 ± 3.10 to 7.48 ± 2.78 ng/ml, P < 0.001). CONCLUSIONS Atorvastatin decreased circulating RANTES in IGT patients with hypercholesterolemia, indicating that statins may play an important role in inhibiting inflammatory responses in patients with IGT.
Collapse
Affiliation(s)
- Xiaomeng Feng
- Department of Endocrinology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, 100020, China
| | - Xia Gao
- Department of Endocrinology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, 100020, China
| | - Yumei Jia
- Department of Endocrinology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, 100020, China
| | - Heng Zhang
- Department of Endocrinology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, 100020, China
| | - Yuan Xu
- Department of Endocrinology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, 100020, China.
| |
Collapse
|
80
|
Li Y, Liu X, Duan W, Tian H, Zhu G, He H, Yao S, Yi S, Song W, Tang H. Batf3-dependent CD8α + Dendritic Cells Aggravates Atherosclerosis via Th1 Cell Induction and Enhanced CCL5 Expression in Plaque Macrophages. EBioMedicine 2017; 18:188-198. [PMID: 28411140 PMCID: PMC5405198 DOI: 10.1016/j.ebiom.2017.04.008] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2016] [Revised: 03/24/2017] [Accepted: 04/04/2017] [Indexed: 01/18/2023] Open
Abstract
Dendritic cells (DCs) play an important role in controlling T cell-mediated adaptive immunity in atherogenesis. However, the role of the basic leucine zipper transcription factor, ATF-like 3 (Batf3)-dependent CD8α+ DC subset in atherogenesis remains unclear. Here we show that Batf3-/-Apoe-/- mice, lacking CD8α+ DCs, exhibited a significant reduction in atherogenesis and T help 1 (Th1) cells compared with Apoe-/- controls. Then, we found that CD8α+ DCs preferentially induce Th1 cells via secreting interleukin-12 (IL-12), and that the expression of interferon-gamma (IFN-γ)or chemokine (C-C motif) ligand 5 (CCL5) in aorta were significantly decreased in Batf3-/-Apoe-/- mice. We further demonstrated that macrophages were the major CCL5-expressing cells in the plaque, which was significantly reduced in Batf3-/-Apoe-/- mice. Furthermore, we found CCL5 expression in macrophages was promoted by IFN-γ. Finally, we showed that Batf3-/-Apoe-/- mice displayed decreased infiltration of leukocytes in the plaque. Thus, CD8α+ DCs aggravated atherosclerosis, likely by inducing Th1 cell response, which promoted CCL5 expression in macrophages and increased infiltration of leukocytes and lesion inflammation.
Collapse
Affiliation(s)
- Yalin Li
- Institute of Immunology, Taishan Medical University, Taian, Shandong, China
| | - Xueyan Liu
- Institute of Immunology, Taishan Medical University, Taian, Shandong, China
| | - Wei Duan
- Institute of Immunology, Taishan Medical University, Taian, Shandong, China
| | - Hua Tian
- Key Laboratory of Atherosclerosis in Universities of Shandong and Institute of Atherosclerosis, Taishan Medical University, Shandong, China
| | - Guangming Zhu
- Institute of Immunology, Taishan Medical University, Taian, Shandong, China
| | - Hao He
- Institute of Immunology, Taishan Medical University, Taian, Shandong, China
| | - Shutong Yao
- Key Laboratory of Atherosclerosis in Universities of Shandong and Institute of Atherosclerosis, Taishan Medical University, Shandong, China
| | - Shuying Yi
- Institute of Immunology, Taishan Medical University, Taian, Shandong, China
| | - Wengang Song
- Institute of Immunology, Taishan Medical University, Taian, Shandong, China.
| | - Hua Tang
- Institute of Immunology, Taishan Medical University, Taian, Shandong, China.
| |
Collapse
|
81
|
Leukocyte Trafficking in Cardiovascular Disease: Insights from Experimental Models. Mediators Inflamm 2017; 2017:9746169. [PMID: 28465628 PMCID: PMC5390637 DOI: 10.1155/2017/9746169] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2017] [Accepted: 03/01/2017] [Indexed: 11/17/2022] Open
Abstract
Chemokine-induced leukocyte migration into the vessel wall is an early pathological event in the progression of atherosclerosis, the underlying cause of myocardial infarction. The immune-inflammatory response, mediated by both the innate and adaptive immune cells, is involved in the initiation, recruitment, and resolution phases of cardiovascular disease progression. Activation of leukocytes via inflammatory mediators such as chemokines, cytokines, and adhesion molecules is instrumental in these processes. In this review, we highlight leukocyte activation with the main focus being on the mechanisms of chemokine-mediated recruitment in atherosclerosis and the response postmyocardial infarction with key examples from experimental models of cardiovascular inflammation.
Collapse
|
82
|
Ravindran D, Ridiandries A, Vanags LZ, Henriquez R, Cartland S, Tan JTM, Bursill CA. Chemokine binding protein 'M3' limits atherosclerosis in apolipoprotein E-/- mice. PLoS One 2017; 12:e0173224. [PMID: 28282403 PMCID: PMC5345809 DOI: 10.1371/journal.pone.0173224] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2016] [Accepted: 02/17/2017] [Indexed: 02/02/2023] Open
Abstract
Chemokines are important in macrophage recruitment and the progression of atherosclerosis. The ‘M3’ chemokine binding protein inactivates key chemokines involved in atherosclerosis (e.g. CCL2, CCL5 and CX3CL1). We aimed to determine the effect of M3 on plaque development and composition. In vitro chemotaxis studies confirmed that M3 protein inhibited the activity of chemokines CCL2, CCL5 and CX3CL1 as primary human monocyte migration as well as CCR2-, CCR5- and CX3CR1-directed migration was attenuated by M3. In vivo, adenoviruses encoding M3 (AdM3) or green fluorescence protein (AdGFP; control) were infused systemically into apolipoprotein (apo)-E-/- mice. Two models of atherosclerosis development were used in which the rate of plaque progression was varied by diet including: (1) a ‘rapid promotion’ model (6-week high-fat-fed) and (2) a ‘slow progression’ model (12-week chow-fed). Plasma chemokine activity was suppressed in AdM3-infused mice as indicated by significantly less monocyte migration towards AdM3 mouse plasma ex vivo (29.56%, p = 0.014). In the ‘slow progression’ model AdM3 mice had reduced lesion area (45.3%, p = 0.035) and increased aortic smooth muscle cell α-actin expression (60.3%, p = 0.014). The reduction in lesion size could not be explained by changes in circulating inflammatory monocytes as they were higher in the AdM3 group. In the ‘rapid promotion’ model AdM3 mice had no changes in plaque size but reduced plaque macrophage content (46.8%, p = 0.006) and suppressed lipid deposition in thoracic aortas (66.9%, p<0.05). There was also a reduction in phosphorylated p65, the active subunit of NF-κb, in the aortas of AdM3 mice (37.3%, p<0.0001). M3 inhibited liver CCL2 concentrations in both models with no change in CCL5 or systemic chemokine levels. These findings show M3 causes varying effects on atherosclerosis progression and plaque composition depending on the rate of lesion progression. Overall, our studies support a promising role for chemokine inhibition with M3 for the treatment of atherosclerosis.
Collapse
Affiliation(s)
- Dhanya Ravindran
- Heart Research Institute, Newtown, Sydney, New South Wales, Australia
- Sydney Medical School, University of Sydney, Camperdown, Sydney, New South Wales, Australia
| | - Anisyah Ridiandries
- Heart Research Institute, Newtown, Sydney, New South Wales, Australia
- Sydney Medical School, University of Sydney, Camperdown, Sydney, New South Wales, Australia
| | - Laura Z. Vanags
- Heart Research Institute, Newtown, Sydney, New South Wales, Australia
- Sydney Medical School, University of Sydney, Camperdown, Sydney, New South Wales, Australia
| | - Rodney Henriquez
- Heart Research Institute, Newtown, Sydney, New South Wales, Australia
| | - Siân Cartland
- Heart Research Institute, Newtown, Sydney, New South Wales, Australia
- Sydney Medical School, University of Sydney, Camperdown, Sydney, New South Wales, Australia
| | - Joanne T. M. Tan
- Heart Research Institute, Newtown, Sydney, New South Wales, Australia
- Sydney Medical School, University of Sydney, Camperdown, Sydney, New South Wales, Australia
| | - Christina A. Bursill
- Heart Research Institute, Newtown, Sydney, New South Wales, Australia
- Sydney Medical School, University of Sydney, Camperdown, Sydney, New South Wales, Australia
- * E-mail:
| |
Collapse
|
83
|
Giordano S, Zhao X, Chen YF, Litovsky SH, Hage FG, Townes TM, Sun CW, Wu LC, Oparil S, Xing D. Induced Pluripotent Stem Cell-Derived Endothelial Cells Overexpressing Interleukin-8 Receptors A/B and/or C-C Chemokine Receptors 2/5 Inhibit Vascular Injury Response. Stem Cells Transl Med 2017; 6:1168-1177. [PMID: 28233474 PMCID: PMC5442847 DOI: 10.1002/sctm.16-0316] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2016] [Revised: 09/14/2016] [Accepted: 10/31/2016] [Indexed: 12/24/2022] Open
Abstract
Recruitment of neutrophils and monocytes/macrophages to the site of vascular injury is mediated by binding of chemoattractants to interleukin (IL) 8 receptors RA and RB (IL8RA/B) C‐C chemokine receptors (CCR) 2 and 5 expressed on neutrophil and monocyte/macrophage membranes. Endothelial cells (ECs) derived from rat‐induced pluripotent stem cells (RiPS) were transduced with adenovirus containing cDNA of IL8RA/B and/or CCR2/5. We hypothesized that RiPS‐ECs overexpressing IL8RA/B (RiPS‐IL8RA/B‐ECs), CCR2/5 (RiPS‐CCR2/5‐ECs), or both receptors (RiPS‐IL8RA/B+CCR2/5‐ECs) will inhibit inflammatory responses and neointima formation in balloon‐injured rat carotid artery. Twelve‐week‐old male Sprague‐Dawley rats underwent balloon injury of the right carotid artery and intravenous infusion of (a) saline vehicle, (b) control RiPS‐Null‐ECs (ECs transduced with empty virus), (c) RiPS‐IL8RA/B‐ECs, (d) RiPS‐CCR2/5‐ECs, or (e) RiPS‐IL8RA/B+CCR2/5‐ECs. Inflammatory mediator expression and leukocyte infiltration were measured in injured and uninjured arteries at 24 hours postinjury by enzyme‐linked immunosorbent assay (ELISA) and immunohistochemistry, respectively. Neointima formation was assessed at 14 days postinjury. RiPS‐ECs expressing the IL8RA/B or CCR2/5 homing device targeted the injured arteries and decreased injury‐induced inflammatory cytokine expression, neutrophil/macrophage infiltration, and neointima formation. Transfused RiPS‐ECs overexpressing IL8RA/B and/or CCR2/5 prevented inflammatory responses and neointima formation after vascular injury. Targeted delivery of iPS‐ECs with a homing device to inflammatory mediators in injured arteries provides a novel strategy for the treatment of cardiovascular diseases. Stem Cells Translational Medicine2017;6:1168–1177
Collapse
Affiliation(s)
- Samantha Giordano
- Vascular Biology and Hypertension Program, Division of Cardiovascular Disease, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Xiangmin Zhao
- Vascular Biology and Hypertension Program, Division of Cardiovascular Disease, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Yiu-Fai Chen
- Vascular Biology and Hypertension Program, Division of Cardiovascular Disease, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Silvio H Litovsky
- Division of Anatomic Pathology, Department of Pathology, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Fadi G Hage
- Vascular Biology and Hypertension Program, Division of Cardiovascular Disease, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA.,Section of Cardiology, Birmingham Veterans Affairs Medical Center, Birmingham, Alabama, USA
| | - Tim M Townes
- Department of Biochemistry and Molecular Genetics, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Chiao-Wang Sun
- Department of Biochemistry and Molecular Genetics, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Li-Chen Wu
- Department of Biochemistry and Molecular Genetics, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Suzanne Oparil
- Vascular Biology and Hypertension Program, Division of Cardiovascular Disease, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Dongqi Xing
- Vascular Biology and Hypertension Program, Division of Cardiovascular Disease, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
| |
Collapse
|
84
|
Lozhkin A, Vendrov AE, Pan H, Wickline SA, Madamanchi NR, Runge MS. NADPH oxidase 4 regulates vascular inflammation in aging and atherosclerosis. J Mol Cell Cardiol 2017; 102:10-21. [PMID: 27986445 PMCID: PMC5625334 DOI: 10.1016/j.yjmcc.2016.12.004] [Citation(s) in RCA: 81] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/23/2016] [Revised: 12/08/2016] [Accepted: 12/12/2016] [Indexed: 01/10/2023]
Abstract
We recently reported that increased NADPH oxidase 4 (NOX4) expression and activity during aging results in enhanced cellular and mitochondrial oxidative stress, vascular inflammation, dysfunction, and atherosclerosis. The goal of the present study was to elucidate the molecular mechanism(s) for these effects and determine the importance of NOX4 modulation of proinflammatory gene expression in mouse vascular smooth muscle cells (VSMCs). A novel peptide-mediated siRNA transfection approach was used to inhibit Nox4 expression with minimal cellular toxicity. Using melittin-derived peptide p5RHH, we achieved significantly higher transfection efficiency (92% vs. 85% with Lipofectamine) and decreased toxicity (p<0.001 vs. Lipofectamine in MTT and p<0.0001 vs. Lipofectamine in LDH assays) in VSMCs. TGFβ1 significantly upregulates Nox4 mRNA (p<0.01) and protein (p<0.01) expression in VSMCs. p5RHH-mediated Nox4 siRNA transfection greatly attenuated TGFβ1-induced upregulation of Nox4 mRNA (p<0.01) and protein (p<0.0001) levels and decreased hydrogen peroxide production (p<0.0001). Expression of pro-inflammatory genes Ccl2, Ccl5, Il6, and Vcam1 was significantly upregulated in VSMCs in several settings cells isolated from aged vs. young wild-type mice, in atherosclerotic arteries of Apoe-/- mice, and atherosclerotic human carotid arteries and correlated with NOX4 expression. p5RHH-mediated Nox4 siRNA transfection significantly attenuated the expression of these pro-inflammatory genes in TGFβ1-treated mouse VSMCs, with the highest degree of inhibition in the expression of Il6. p5RHH peptide-mediated knockdown of TGFβ-activated kinase 1 (TAK1, also known as Map3k7), Jun, and Rela, but not Nfkb2, downregulated TGFβ1-induced Nox4 expression in VSMCs. Together, these data demonstrate that increased expression and activation of NOX4, which might result from increased TGFβ1 levels seen during aging, induces a proinflammatory phenotype in VSMCs, enhancing atherosclerosis.
Collapse
Affiliation(s)
- Andrey Lozhkin
- Frankel Cardiovascular Center, Department of Internal Medicine, University of Michigan, Ann Arbor 48109, MI, USA
| | - Aleksandr E Vendrov
- Frankel Cardiovascular Center, Department of Internal Medicine, University of Michigan, Ann Arbor 48109, MI, USA
| | - Hua Pan
- Division of Cardiology, Department of Medicine, Washington University School of Medicine, Saint Louis 63110, MO, USA
| | - Samuel A Wickline
- Division of Cardiology, Department of Medicine, Washington University School of Medicine, Saint Louis 63110, MO, USA
| | - Nageswara R Madamanchi
- Frankel Cardiovascular Center, Department of Internal Medicine, University of Michigan, Ann Arbor 48109, MI, USA
| | - Marschall S Runge
- Frankel Cardiovascular Center, Department of Internal Medicine, University of Michigan, Ann Arbor 48109, MI, USA.
| |
Collapse
|
85
|
Ciesielska A, Sas-Nowosielska H, Kwiatkowska K. Bis(monoacylglycero)phosphate inhibits TLR4-dependent RANTES production in macrophages. Int J Biochem Cell Biol 2016; 83:15-26. [PMID: 27939812 DOI: 10.1016/j.biocel.2016.12.003] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2016] [Revised: 11/14/2016] [Accepted: 12/06/2016] [Indexed: 01/10/2023]
Abstract
Toll-like receptor 4 (TLR4) is the receptor for bacterial lipopolysaccharide (LPS) triggering production of pro-inflammatory cytokines which help eradicate the bacteria but could also be harmful when overproduced. The signaling activity of TLR4 is modulated by cholesterol level in cellular membranes, which in turn is affected by bis(monoacylglycero)phosphate (BMP), a phospholipid enriched in late endosomes. We found that exogenously added BMP isomers become incorporated into the plasma membrane and intracellular vesicles of macrophages and strongly reduced LPS-stimulated production of a chemokine RANTES, which was correlated with inhibition of interferon regulatory factor 3 (IRF3) controlling Rantes expression. To investigate the mechanism underlying the influence of BMP on TLR4 signaling we applied Laurdan and studied the impact of BMP incorporation on lipid packing, a measure for membrane order. Enrichment of model and cellular membranes with BMP significantly reduced their order and the reduction was maintained during stimulation of cells with LPS. This effect of BMP was abolished by enrichment of macrophages with cholesterol. In parallel, the inhibitory effect of BMP exerted on the TLR4-dependent phosphorylation of IRF3 was also reversed. Taken together our results indicate that BMP reduces the order of macrophage membranes which contributes to the inhibition of TLR4-dependent RANTES production.
Collapse
Affiliation(s)
- Anna Ciesielska
- Laboratory of Molecular Membrane Biology, Department of Cell Biology, Nencki Institute of Experimental Biology of the Polish Academy of Sciences, 3 Pasteur St., 02-093 Warsaw, Poland
| | - Hanna Sas-Nowosielska
- Laboratory for Imaging Tissue Structure and Function, Neurobiology Center, Nencki Institute of Experimental Biology of the Polish Academy of Sciences, 3 Pasteur St., 02-093 Warsaw, Poland
| | - Katarzyna Kwiatkowska
- Laboratory of Molecular Membrane Biology, Department of Cell Biology, Nencki Institute of Experimental Biology of the Polish Academy of Sciences, 3 Pasteur St., 02-093 Warsaw, Poland.
| |
Collapse
|
86
|
Abstract
Platelets are small, anucleate circulating cells that possess a dynamic repertoire of functions spanning the hemostatic, inflammatory, and immune continuum. Once thought to be merely cell fragments with responses limited primarily to acute hemostasis and vascular wall repair, platelets are now increasingly recognized as key sentinels and effector cells regulating host responses to many inflammatory and infectious cues. Platelet granules, including α-granules and dense-granules, store hundreds of factors and secrete these mediators in response to activating signals. The cargo packaged and stored within platelet granules orchestrates communication between platelets and other circulating cells, augments host defense mechanisms to invading pathogens and tumor cells, and - in some settings - drives dysregulated and injurious responses. This focused review will highlight several of the established and emerging mechanisms and roles of platelet secretion in inflammatory and infectious diseases.
Collapse
Affiliation(s)
- Bhanu K Manne
- a The University of Utah Molecular Medicine Program , Salt Lake City , Utah , USA
| | | | - Matthew T Rondina
- a The University of Utah Molecular Medicine Program , Salt Lake City , Utah , USA.,c Department of Internal Medicine , Salt Lake City , Utah , USA.,d The GRECC, George E. Wahlen Salt Lake City VAMC , Salt Lake City , Utah , USA
| |
Collapse
|
87
|
Jabir NR, Firoz CK, Kamal MA, Damanhouri GA, Alama MN, Alzahrani AS, Almehdar HA, Tabrez S. Assessment of genetic diversity in IL-6 and RANTES promoters and their level in Saudi coronary artery disease patients. J Clin Lab Anal 2016; 31. [PMID: 27862306 DOI: 10.1002/jcla.22092] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2016] [Accepted: 10/07/2016] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND The present study consisted of a total of 200 subjects (100 confirmed coronary artery disease (CAD) patients), both men and women, and 100 healthy control individuals. METHODS Serum concentration of IL-6 and RANTES were measured by enzyme-linked immunosorbent assay kit. For SNPs analysis, sanger method of DNA sequencing was followed. RESULTS We observed variable numbers of SNP sites at -174 G/C, -572 G/C, and -597 G/A in IL-6 and -28 C/G and -109 C/T in RANTES promoters in CAD patients compared with control individuals. However, the observed changes in the number of SNPs were found to be non-significant compared with control individuals. The IL-6 level was found to be significantly (P<.001) elevated in CAD patients compared with control. Moreover, RANTES serum level did not show any significant change in CAD patients. CONCLUSION Based on our result, it is quite clear that inflammation has a role in the pathogenesis of CAD but does not lead to significant changes at the genetic level in our population. As far as our knowledge goes, this is the first report that shows the genetic diversity in IL-6 and RANTES promoters and their respective levels in Saudi CAD patients.
Collapse
Affiliation(s)
- Nasimudeen R Jabir
- King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Chelapram K Firoz
- King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Mohammad A Kamal
- King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Ghazi A Damanhouri
- King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Mohammed Nabil Alama
- Department of Cardiology, Faculty of Medicine, King Abdulaziz University Hospital, Jeddah, Saudi Arabia
| | - Anas S Alzahrani
- Department of Internal Medicine, Faculty of Medicine, University of Jeddah, Jeddah, Saudi Arabia
| | - Hussein A Almehdar
- Department of Biology, Faculty of Science, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Shams Tabrez
- King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
| |
Collapse
|
88
|
Chang TT, Chen JW. Emerging role of chemokine CC motif ligand 4 related mechanisms in diabetes mellitus and cardiovascular disease: friends or foes? Cardiovasc Diabetol 2016; 15:117. [PMID: 27553774 PMCID: PMC4995753 DOI: 10.1186/s12933-016-0439-9] [Citation(s) in RCA: 72] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/25/2016] [Accepted: 08/12/2016] [Indexed: 12/14/2022] Open
Abstract
Chemokines are critical components in pathology. The roles of chemokine CC motif ligand 4 (CCL4) and its receptor are associated with diabetes mellitus (DM) and atherosclerosis cardiovascular diseases. However, due to the complexity of these diseases, the specific effects of CCL4 remain unclear, although recent reports have suggested that multiple pathways are related to CCL4. In this review, we provide an overview of the role and potential mechanisms of CCL4 and one of its major receptors, fifth CC chemokine receptor (CCR5), in DM and cardiovascular diseases. CCL4-related mechanisms, including CCL4 and CCR5, might provide potential therapeutic targets in DM and/or atherosclerosis cardiovascular diseases.
Collapse
Affiliation(s)
- Ting-Ting Chang
- Institute of Pharmacology, National Yang-Ming University, Taipei, Taiwan, R.O.C
| | - Jaw-Wen Chen
- Institute of Pharmacology, National Yang-Ming University, Taipei, Taiwan, R.O.C. .,Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan, R.O.C. .,Cardiovascular Research Center, National Yang-Ming University, Taipei, Taiwan, R.O.C. .,Division of Clinical Research, Department of Medical Research, Taipei Veterans General Hospital, Taipei, Taiwan, R.O.C.
| |
Collapse
|
89
|
Di Marco E, Gray S, Chew P, Kennedy K, Cooper M, Schmidt H, Jandeleit-Dahm K. Differential effects of NOX4 and NOX1 on immune cell-mediated inflammation in the aortic sinus of diabetic ApoE−/− mice. Clin Sci (Lond) 2016; 130:1363-1374. [DOI: 10.1042/cs20160249] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/30/2023]
Abstract
Oxidative stress and inflammation are central mediators of atherosclerosis particularly in the context of diabetes. The potential interactions between the major producers of vascular reactive oxygen species (ROS), NADPH oxidase (NOX) enzymes and immune-inflammatory processes remain to be fully elucidated. In the present study we investigated the roles of the NADPH oxidase subunit isoforms, NOX4 and NOX1, in immune cell activation and recruitment to the aortic sinus atherosclerotic plaque in diabetic ApoE−/− mice. Plaque area analysis showed that NOX4- and NOX1-derived ROS contribute to atherosclerosis in the aortic sinus following 10 weeks of diabetes. Immunohistochemical staining of the plaques revealed that NOX4-derived ROS regulate T-cell recruitment. In addition, NOX4-deficient mice showed a reduction in activated CD4+ T-cells in the draining lymph nodes of the aortic sinus coupled with reduced pro-inflammatory gene expression in the aortic sinus. Conversely, NOX1-derived ROS appeared to play a more important role in macrophage accumulation. These findings demonstrate distinct roles for NOX4 and NOX1 in immune-inflammatory responses that drive atherosclerosis in the aortic sinus of diabetic mice.
Collapse
Affiliation(s)
- Elyse Di Marco
- Diabetic Complications Division, Baker IDI Heart & Diabetes Institute, Melbourne, Australia
- Department of Medicine, Monash University, Melbourne, Australia
| | - Stephen P. Gray
- Diabetic Complications Division, Baker IDI Heart & Diabetes Institute, Melbourne, Australia
- Department of Medicine, Monash University, Melbourne, Australia
| | - Phyllis Chew
- Diabetic Complications Division, Baker IDI Heart & Diabetes Institute, Melbourne, Australia
| | - Kit Kennedy
- Diabetic Complications Division, Baker IDI Heart & Diabetes Institute, Melbourne, Australia
| | - Mark E. Cooper
- Diabetic Complications Division, Baker IDI Heart & Diabetes Institute, Melbourne, Australia
| | - Harald H.H.W. Schmidt
- Department of Pharmacology & Cardiovascular Research Institute Maastricht (CARIM), Faculty of Medicine, Health & Life Science, Maastricht University, The Netherlands
| | - Karin A.M. Jandeleit-Dahm
- Diabetic Complications Division, Baker IDI Heart & Diabetes Institute, Melbourne, Australia
- Department of Medicine, Monash University, Melbourne, Australia
| |
Collapse
|
90
|
Czepluch FS, Meier J, Binder C, Hasenfuss G, Schäfer K. CCL5 deficiency reduces neointima formation following arterial injury and thrombosis in apolipoprotein E-deficient mice. Thromb Res 2016; 144:136-43. [DOI: 10.1016/j.thromres.2016.06.013] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2016] [Revised: 06/12/2016] [Accepted: 06/14/2016] [Indexed: 01/21/2023]
|
91
|
Krikke M, Tesselaar K, Arends JE, Drylewicz J, Otto SA, van Lelyveld SFL, Visseren FJL, Hoepelman AIM. Maraviroc Intensification Improves Endothelial Function in Abacavir-Treated Patients, an Open-Label Randomized Cross-Over Pilot Study. Infect Dis Ther 2016; 5:389-404. [PMID: 27300170 PMCID: PMC5019971 DOI: 10.1007/s40121-016-0115-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2016] [Indexed: 12/30/2022] Open
Abstract
Background The increased risk of abacavir in cardiovascular disease (CVD) in HIV-infected patients is still being debated. Maraviroc, a CCR5 blocker, has been shown to decrease immune activation and monocyte infiltration in atherosclerotic plaques in murine experiments. Therefore, we examined the effect of maraviroc intensification on flow-mediated dilatation (FMD) in abacavir-treated HIV-infected patients and its effect on immunological and inflammatory parameters. Methods A open-label prospective crossover study with a duration of 16 weeks: 8 weeks of intervention (maraviroc intensification) and 8 weeks of control (unchanged cART regimen). FMD, HIV-specific variables, expression of HIV co-receptors, markers of inflammation and coagulation and cellular markers of immune activation were measured at weeks 0, 8 and 16. The changes (Δ) in these variables were compared between intervention and control periods using non-parametric tests. To evaluate the relation with the change in FMD, linear regression modeling was used. Results Twenty-one male patients with suppressed plasma HIV-RNA, on cART, had a known HIV infection for 9.2 years (IQR 6.9–13.5) with abacavir use for 6.5 years (2.8–9.3). A significantly increased FMD of 0.73% (IQR −0.25 to 1.70) was seen after maraviroc intensification compared to a decrease of −0.42% (IQR −1.89 to 0.25; p = 0.049) in the control period. There was a negative relation between ΔFMD with ΔD-dimer (β −22.70, 95% CI −39.27; −6.13, p = 0.011) and ΔCD95+ CD4+ T cells (β −0.16, 95% CI −0.28; −0.04, p = 0.013), adjusted for age and duration of HIV. Conclusion Maraviroc intensification modestly improves endothelial function in HIV-infected patients on an abacavir-containing regimen. Trial registration NCT01389063. Electronic supplementary material The online version of this article (doi:10.1007/s40121-016-0115-0) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Maaike Krikke
- Department of Internal Medicine and Infectious Diseases, University Medical Center Utrecht (UMCU), Heidelberglaan 100, F02.126, P.O. Box 85500, 3508 GA, Utrecht, The Netherlands.
- Laboratory of Translational Immunology, Department of Immunology, University Medical Center Utrecht (UMCU), Lundlaan 6, KC02.085.2, P.O. Box 85090, 3508 AB, Utrecht, The Netherlands.
| | - Kiki Tesselaar
- Laboratory of Translational Immunology, Department of Immunology, University Medical Center Utrecht (UMCU), Lundlaan 6, KC02.085.2, P.O. Box 85090, 3508 AB, Utrecht, The Netherlands
| | - Joop E Arends
- Department of Internal Medicine and Infectious Diseases, University Medical Center Utrecht (UMCU), Heidelberglaan 100, F02.126, P.O. Box 85500, 3508 GA, Utrecht, The Netherlands
| | - Julia Drylewicz
- Laboratory of Translational Immunology, Department of Immunology, University Medical Center Utrecht (UMCU), Lundlaan 6, KC02.085.2, P.O. Box 85090, 3508 AB, Utrecht, The Netherlands
| | - Sigrid A Otto
- Laboratory of Translational Immunology, Department of Immunology, University Medical Center Utrecht (UMCU), Lundlaan 6, KC02.085.2, P.O. Box 85090, 3508 AB, Utrecht, The Netherlands
| | - Steven F L van Lelyveld
- Department of Internal Medicine and Infectious Diseases, University Medical Center Utrecht (UMCU), Heidelberglaan 100, F02.126, P.O. Box 85500, 3508 GA, Utrecht, The Netherlands
- Department of Internal Medicine and Gastroenterology, Spaarne Gasthuis, Boerhaavelaan 22, 2035 RC, Haarlem, The Netherlands
| | - Frank J L Visseren
- Department of Vascular Medicine, University Medical Center Utrecht (UMCU), Heidelberglaan 100, F02.126, P.O. Box 85500, 3508 GA, Utrecht, The Netherlands
| | - Andy I M Hoepelman
- Department of Internal Medicine and Infectious Diseases, University Medical Center Utrecht (UMCU), Heidelberglaan 100, F02.126, P.O. Box 85500, 3508 GA, Utrecht, The Netherlands
| |
Collapse
|
92
|
Martorell S, Hueso L, Gonzalez-Navarro H, Collado A, Sanz MJ, Piqueras L. Vitamin D Receptor Activation Reduces Angiotensin-II-Induced Dissecting Abdominal Aortic Aneurysm in Apolipoprotein E-Knockout Mice. Arterioscler Thromb Vasc Biol 2016; 36:1587-97. [PMID: 27283745 DOI: 10.1161/atvbaha.116.307530] [Citation(s) in RCA: 58] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2015] [Accepted: 05/27/2016] [Indexed: 01/03/2023]
Abstract
OBJECTIVE Abdominal aortic aneurysm (AAA) is a vascular disorder characterized by chronic inflammation of the aortic wall. Low concentrations of vitamin D3 are associated with AAA development; however, the potential direct effect of vitamin D3 on AAA remains unknown. This study evaluates the effect of oral treatment with the vitamin D3 receptor (VDR) ligand, calcitriol, on dissecting AAA induced by angiotensin-II (Ang-II) infusion in apoE(-/-) mice. APPROACH AND RESULTS Oral treatment with calcitriol reduced Ang-II-induced dissecting AAA formation in apoE(-/-) mice, which was unrelated to systolic blood pressure or plasma cholesterol concentrations. Immunohistochemistry and reverse-transcription polymerase chain reaction analysis demonstrated a significant increase in macrophage infiltration, neovessel formation, matrix metalloproteinase-2 and matrix metalloproteinase-9, chemokine (CCL2 [(C-C motif) ligand 2], CCL5 [(C-C motif) ligand 5], and CXCL1 [(C-X-C motif) ligand 1]) and vascular endothelial growth factor expression in suprarenal aortic walls of apoE(-/-) mice infused with Ang-II, and all were significantly reduced by cotreatment with calcitriol. Phosphorylation of extracellular signal-regulated kinases 1/2, p38 mitogen-activated protein kinase, and nuclear factor-κB was also decreased in the suprarenal aortas of apoE(-/-) mice cotreated with calcitriol. These effects were accompanied by a marked increase in VDR-retinoid X receptor (RXR) interaction in the aortas of calcitriol-treated mice. In vitro, VDR activation by calcitriol in human endothelial cells inhibited Ang-II-induced leukocyte-endothelial cell interactions, morphogenesis, and production of endothelial proinflammatory and angiogenic chemokines through VDR-RXR interactions, and knockdown of VDR or RXR abolished the inhibitory effects of calcitriol. CONCLUSIONS VDR activation reduces dissecting AAA formation induced by Ang-II in apoE(-/-) mice and may constitute a novel therapeutic strategy to prevent AAA progression.
Collapse
Affiliation(s)
- Sara Martorell
- From the Institute of Health Research-INCLIVA, Department of Pharmacology, Valencia, Spain (S.M., L.H., H.G.-N., A.C., M.-J.S., L.P.); Faculty of Medicine, Department of Pharmacology, University of Valencia, Valencia, Spain (M.-J.S.); and Diabetes and Associated Metabolic Disorders Unit, CIBERDEM, Madrid, Spain (H.G.-N.)
| | - Luisa Hueso
- From the Institute of Health Research-INCLIVA, Department of Pharmacology, Valencia, Spain (S.M., L.H., H.G.-N., A.C., M.-J.S., L.P.); Faculty of Medicine, Department of Pharmacology, University of Valencia, Valencia, Spain (M.-J.S.); and Diabetes and Associated Metabolic Disorders Unit, CIBERDEM, Madrid, Spain (H.G.-N.)
| | - Herminia Gonzalez-Navarro
- From the Institute of Health Research-INCLIVA, Department of Pharmacology, Valencia, Spain (S.M., L.H., H.G.-N., A.C., M.-J.S., L.P.); Faculty of Medicine, Department of Pharmacology, University of Valencia, Valencia, Spain (M.-J.S.); and Diabetes and Associated Metabolic Disorders Unit, CIBERDEM, Madrid, Spain (H.G.-N.)
| | - Aida Collado
- From the Institute of Health Research-INCLIVA, Department of Pharmacology, Valencia, Spain (S.M., L.H., H.G.-N., A.C., M.-J.S., L.P.); Faculty of Medicine, Department of Pharmacology, University of Valencia, Valencia, Spain (M.-J.S.); and Diabetes and Associated Metabolic Disorders Unit, CIBERDEM, Madrid, Spain (H.G.-N.)
| | - Maria-Jesus Sanz
- From the Institute of Health Research-INCLIVA, Department of Pharmacology, Valencia, Spain (S.M., L.H., H.G.-N., A.C., M.-J.S., L.P.); Faculty of Medicine, Department of Pharmacology, University of Valencia, Valencia, Spain (M.-J.S.); and Diabetes and Associated Metabolic Disorders Unit, CIBERDEM, Madrid, Spain (H.G.-N.).
| | - Laura Piqueras
- From the Institute of Health Research-INCLIVA, Department of Pharmacology, Valencia, Spain (S.M., L.H., H.G.-N., A.C., M.-J.S., L.P.); Faculty of Medicine, Department of Pharmacology, University of Valencia, Valencia, Spain (M.-J.S.); and Diabetes and Associated Metabolic Disorders Unit, CIBERDEM, Madrid, Spain (H.G.-N.).
| |
Collapse
|
93
|
Li J, McArdle S, Gholami A, Kimura T, Wolf D, Gerhardt T, Miller J, Weber C, Ley K. CCR5+T-bet+FoxP3+ Effector CD4 T Cells Drive Atherosclerosis. Circ Res 2016; 118:1540-52. [PMID: 27021296 PMCID: PMC4867125 DOI: 10.1161/circresaha.116.308648] [Citation(s) in RCA: 109] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/01/2016] [Accepted: 03/28/2016] [Indexed: 12/20/2022]
Abstract
RATIONALE CD4 T cells are involved in the pathogenesis of atherosclerosis, but atherosclerosis-specific CD4 T cells have not been described. Moreover, the chemokine(s) that regulates T-cell trafficking to the atherosclerotic lesions is also unknown. OBJECTIVE In Apoe(-/-) mice with mature atherosclerotic lesions (5 months of high fat diet), we find that most aortic T cells express CCR5 and interferon-γ with a unique combination of cell surface markers (CD4(+)CD25(-)CD44(hi)CD62L(lo)) and transcription factors (FoxP3(+)T-bet(+)). We call these cells CCR5Teff. We investigated the role of CCR5 in regulating T-cell homing to the atherosclerotic aorta and the functionality of the CCR5Teff cells. METHODS AND RESULTS CCR5Teff cells are exclusively found in the aorta and para-aortic lymph nodes of Apoe(-/-) mice. They do not suppress T-cell proliferation in vitro and are less potent than regulatory T cells at inhibiting cytokine secretion. Blocking or knocking out CCR5 or its ligand CCL5 significantly blocks T-cell homing to atherosclerotic aortas. Transcriptomic analysis shows that CCR5Teff cells are more similar to effector T cells than to regulatory T cells. They secrete interferon-γ, interleukin-2, interleukin-10, and tumor necrosis factor. Adoptive transfer of these CCR5Teff cells significantly increases atherosclerosis. CONCLUSIONS CCR5 is specifically needed for CD4 T-cell homing to the atherosclerotic plaques. CCR5(+)CD4 T cells express an unusual combination of transcription factors, FoxP3 and T-bet. Although CCR5Teff express FoxP3, we showed that they are not regulatory and adoptive transfer of these cells exacerbates atherosclerosis.
Collapse
Affiliation(s)
- Jie Li
- From the Division of Inflammation Biology (J.L., S.M., T.K., D.W., T.G., J.M., K.L.) and Bioinformatics Core (A.G.), La Jolla Institute for Allergy & Immunology, CA; Institute for Cardiovascular Prevention, Ludwig-Maximilians-University Munich, Munich, Germany (C.W.); and DZHK (German Centre for Cardiovascular Research), partner site Munich Heart Alliance, Munich, Germany (C.W.)
| | - Sara McArdle
- From the Division of Inflammation Biology (J.L., S.M., T.K., D.W., T.G., J.M., K.L.) and Bioinformatics Core (A.G.), La Jolla Institute for Allergy & Immunology, CA; Institute for Cardiovascular Prevention, Ludwig-Maximilians-University Munich, Munich, Germany (C.W.); and DZHK (German Centre for Cardiovascular Research), partner site Munich Heart Alliance, Munich, Germany (C.W.)
| | - Amin Gholami
- From the Division of Inflammation Biology (J.L., S.M., T.K., D.W., T.G., J.M., K.L.) and Bioinformatics Core (A.G.), La Jolla Institute for Allergy & Immunology, CA; Institute for Cardiovascular Prevention, Ludwig-Maximilians-University Munich, Munich, Germany (C.W.); and DZHK (German Centre for Cardiovascular Research), partner site Munich Heart Alliance, Munich, Germany (C.W.)
| | - Takayuki Kimura
- From the Division of Inflammation Biology (J.L., S.M., T.K., D.W., T.G., J.M., K.L.) and Bioinformatics Core (A.G.), La Jolla Institute for Allergy & Immunology, CA; Institute for Cardiovascular Prevention, Ludwig-Maximilians-University Munich, Munich, Germany (C.W.); and DZHK (German Centre for Cardiovascular Research), partner site Munich Heart Alliance, Munich, Germany (C.W.)
| | - Dennis Wolf
- From the Division of Inflammation Biology (J.L., S.M., T.K., D.W., T.G., J.M., K.L.) and Bioinformatics Core (A.G.), La Jolla Institute for Allergy & Immunology, CA; Institute for Cardiovascular Prevention, Ludwig-Maximilians-University Munich, Munich, Germany (C.W.); and DZHK (German Centre for Cardiovascular Research), partner site Munich Heart Alliance, Munich, Germany (C.W.)
| | - Teresa Gerhardt
- From the Division of Inflammation Biology (J.L., S.M., T.K., D.W., T.G., J.M., K.L.) and Bioinformatics Core (A.G.), La Jolla Institute for Allergy & Immunology, CA; Institute for Cardiovascular Prevention, Ludwig-Maximilians-University Munich, Munich, Germany (C.W.); and DZHK (German Centre for Cardiovascular Research), partner site Munich Heart Alliance, Munich, Germany (C.W.)
| | - Jacqueline Miller
- From the Division of Inflammation Biology (J.L., S.M., T.K., D.W., T.G., J.M., K.L.) and Bioinformatics Core (A.G.), La Jolla Institute for Allergy & Immunology, CA; Institute for Cardiovascular Prevention, Ludwig-Maximilians-University Munich, Munich, Germany (C.W.); and DZHK (German Centre for Cardiovascular Research), partner site Munich Heart Alliance, Munich, Germany (C.W.)
| | - Christian Weber
- From the Division of Inflammation Biology (J.L., S.M., T.K., D.W., T.G., J.M., K.L.) and Bioinformatics Core (A.G.), La Jolla Institute for Allergy & Immunology, CA; Institute for Cardiovascular Prevention, Ludwig-Maximilians-University Munich, Munich, Germany (C.W.); and DZHK (German Centre for Cardiovascular Research), partner site Munich Heart Alliance, Munich, Germany (C.W.)
| | - Klaus Ley
- From the Division of Inflammation Biology (J.L., S.M., T.K., D.W., T.G., J.M., K.L.) and Bioinformatics Core (A.G.), La Jolla Institute for Allergy & Immunology, CA; Institute for Cardiovascular Prevention, Ludwig-Maximilians-University Munich, Munich, Germany (C.W.); and DZHK (German Centre for Cardiovascular Research), partner site Munich Heart Alliance, Munich, Germany (C.W.).
| |
Collapse
|
94
|
Feng X, Gao X, Jia Y, Zhang H, Xu Y, Wang G. PPAR-α Agonist Fenofibrate Decreased RANTES Levels in Type 2 Diabetes Patients with Hypertriglyceridemia. Med Sci Monit 2016; 22:743-51. [PMID: 26944934 PMCID: PMC4784549 DOI: 10.12659/msm.897307] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Background Regulated upon activation, normal T cells expressed and secreted (RANTES) is associated with inflammation and atherosclerosis. We investigated the effect of fenofibrate, a peroxisome proliferator-activated receptor-α (PPAR-α) agonist, on RANTES in type 2 diabetes mellitus (T2DM) patients with hypertriglyceridemia. Material/Methods This study evaluated cross-sectional and interventional studies of 25 T2DM patients with hypertriglyceridemia (group A) and 32 controls (group B). Group A was treated with fenofibrate (200 mg/day) for 8 weeks. Serum RANTES and clinical characteristics were examined. Results Serum RANTES was significantly higher in group A compared with group B (59.04±16.74 vs. 38.57±12.98 ng/ml, P<0.001) and correlated with triglycerides (TG) (r=0.535, P<0.001), fasting blood glucose (FBG) (r=0.485, P<0.001), glycosylated hemoglobin (HbA1c) (r=0.485, P<0.001), homocysteine (Hcy) (r=0.520, P<0.001), and high-sensitivity C-reactive protein (hsCRP) (r=0.701, P<0.001). In multiple regression analysis after controlling for confounders, increased hsCRP levels (β=7.430, P<0.001) and T2DM with hypertriglyceridemia (β=11.496, P=0.002) were independently related to high serum RANTES levels. After 8 weeks of fenofibrate treatment, serum RANTES significantly decreased in group A compared with baseline (52.75±17.41 vs. 59.04±16.74 ng/ml, P=0.018). Conclusions Fenofibrate decreased serum RANTES levels in T2DM patients with hypertriglyceridemia, indicating that PPAR-α agonists may play an important role in inhibiting inflammatory responses.
Collapse
Affiliation(s)
- Xiaomeng Feng
- Department of Endocrinology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China (mainland)
| | - Xia Gao
- Department of Endocrinology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China (mainland)
| | - Yumei Jia
- Department of Endocrinology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China (mainland)
| | - Heng Zhang
- Department of Endocrinology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China (mainland)
| | - Yuan Xu
- Department of Endocrinology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China (mainland)
| | - Guang Wang
- Department of Endocrinology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China (mainland)
| |
Collapse
|
95
|
Mikolajczyk TP, Nosalski R, Szczepaniak P, Budzyn K, Osmenda G, Skiba D, Sagan A, Wu J, Vinh A, Marvar PJ, Guzik B, Podolec J, Drummond G, Lob HE, Harrison DG, Guzik TJ. Role of chemokine RANTES in the regulation of perivascular inflammation, T-cell accumulation, and vascular dysfunction in hypertension. FASEB J 2016; 30:1987-99. [PMID: 26873938 PMCID: PMC4836375 DOI: 10.1096/fj.201500088r] [Citation(s) in RCA: 192] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2015] [Accepted: 01/27/2016] [Indexed: 12/21/2022]
Abstract
Recent studies have emphasized the role of perivascular inflammation in cardiovascular disease. We studied mechanisms of perivascular leukocyte infiltration in angiotensin II (Ang II)-induced hypertension and their links to vascular dysfunction. Chronic Ang II infusion in mice increased immune cell content of T cells (255 ± 130 to 1664 ± 349 cells/mg; P < 0.01), M1 and M2 macrophages, and dendritic cells in perivascular adipose tissue. In particular, the content of T lymphocytes bearing CC chemokine receptor (CCR) 1, CCR3, and CCR5 receptors for RANTES chemokine was increased by Ang II (CCR1, 15.6 ± 1.5% vs. 31 ± 5%; P < 0.01). Hypertension was associated with an increase in perivascular adipose tissue expression of the chemokine RANTES (relative quantification, 1.2 ± 0.2 vs. 3.5 ± 1.1; P < 0.05), which induced T-cell chemotaxis and vascular accumulation of T cells expressing the chemokine receptors CCR1, CCR3, and CCR5. Mechanistically, RANTES−/− knockout protected against vascular leukocyte, and in particular T lymphocyte infiltration (26 ± 5% in wild type Ang II vs. 15 ± 4% in RANTES−/−), which was associated with protection from endothelial dysfunction induced by Ang II. This effect was linked with diminished infiltration of IFN-γ-producing CD8+ and double-negative CD3+CD4−CD8− T cells in perivascular space and reduced vascular oxidative stress while FoxP3+ T-regulatory cells were unaltered. IFN-γ ex vivo caused significant endothelial dysfunction, which was reduced by superoxide anion scavenging. In a human cohort, a significant inverse correlation was observed between circulating RANTES levels as a biomarker and vascular function measured as flow-mediated dilatation (R = −0.3, P < 0.01) or endothelial injury marker von Willebrand factor (R = +0.3; P < 0.01). Thus, chemokine RANTES is important in the regulation of vascular dysfunction through modulation of perivascular inflammation.—Mikolajczyk, T. P., Nosalski, R., Szczepaniak, P., Budzyn, K., Osmenda, G., Skiba, D., Sagan, A., Wu, J., Vinh, A., Marvar, P. J., Guzik, B., Podolec, J., Drummond, G., Lob, H. E., Harrison, D. G., Guzik, T. J. Role of chemokine RANTES in the regulation of perivascular inflammation, T-cell accumulation, and vascular dysfunction in hypertension.
Collapse
Affiliation(s)
- Tomasz P Mikolajczyk
- Department of Internal Medicine, Jagiellonian University, Cracow, Poland British Heart Foundation Centre for Excellence, Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, United Kingdom
| | - Ryszard Nosalski
- Department of Internal Medicine, Jagiellonian University, Cracow, Poland British Heart Foundation Centre for Excellence, Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, United Kingdom
| | - Piotr Szczepaniak
- Department of Internal Medicine, Jagiellonian University, Cracow, Poland British Heart Foundation Centre for Excellence, Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, United Kingdom
| | - Klaudia Budzyn
- Department of Pharmacology, Monash University, Melbourne, Victoria, Australia
| | - Grzegorz Osmenda
- Department of Internal Medicine, Jagiellonian University, Cracow, Poland
| | - Dominik Skiba
- Department of Internal Medicine, Jagiellonian University, Cracow, Poland British Heart Foundation Centre for Excellence, Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, United Kingdom
| | - Agnieszka Sagan
- Department of Internal Medicine, Jagiellonian University, Cracow, Poland British Heart Foundation Centre for Excellence, Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, United Kingdom
| | - Jing Wu
- Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - Antony Vinh
- Department of Pharmacology, Monash University, Melbourne, Victoria, Australia
| | - Paul J Marvar
- Department of Pharmacology and Physiology, George Washington University, Washington, D.C., USA
| | - Bartlomiej Guzik
- Department of Internal Medicine, Jagiellonian University, Cracow, Poland
| | - Jakub Podolec
- Department of Internal Medicine, Jagiellonian University, Cracow, Poland
| | - Grant Drummond
- Department of Pharmacology, Monash University, Melbourne, Victoria, Australia
| | - Heinrich E Lob
- Department of Biomedical Sciences, Cornell University, Ithaca, New York, USA
| | - David G Harrison
- Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - Tomasz J Guzik
- Department of Internal Medicine, Jagiellonian University, Cracow, Poland British Heart Foundation Centre for Excellence, Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, United Kingdom
| |
Collapse
|
96
|
Hellberg S, Silvola JMU, Kiugel M, Liljenbäck H, Metsälä O, Viljanen T, Metso J, Jauhiainen M, Saukko P, Nuutila P, Ylä-Herttuala S, Knuuti J, Roivainen A, Saraste A. Type 2 diabetes enhances arterial uptake of choline in atherosclerotic mice: an imaging study with positron emission tomography tracer ¹⁸F-fluoromethylcholine. Cardiovasc Diabetol 2016; 15:26. [PMID: 26852231 PMCID: PMC4744438 DOI: 10.1186/s12933-016-0340-6] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/18/2015] [Accepted: 01/18/2016] [Indexed: 01/13/2023] Open
Abstract
Background Diabetes is a risk factor for atherosclerosis associated with oxidative stress, inflammation and cell proliferation. The purpose of this study was to evaluate arterial choline uptake and its relationship to atherosclerotic inflammation in diabetic and non-diabetic hypercholesterolemic mice. Methods Low-density lipoprotein-receptor deficient mice expressing only apolipoprotein B100, with or without type 2 diabetes caused by pancreatic overexpression of insulin-like growth factor II (IGF-II/LDLR−/−ApoB100/100 and LDLR−/−ApoB100/100) were studied. Distribution kinetics of choline analogue 18F-fluoromethylcholine (18F-FMCH) was assessed in vivo by positron emission tomography (PET) imaging. Then, aortic uptakes of 18F-FMCH and glucose analogue 18F-fluorodeoxyglucose (18F-FDG), were assessed ex vivo by gamma counting and autoradiography of tissue sections. The 18F-FMCH uptake in atherosclerotic plaques was further compared with macrophage infiltration and the plasma levels of cytokines and metabolic markers. Results The aortas of all hypercholesterolemic mice showed large, macrophage-rich atherosclerotic plaques. The plaque burden and densities of macrophage subtypes were similar in diabetic and non-diabetic animals. The blood clearance of 18F-FMCH was rapid. Both the absolute 18F-FMCH uptake in the aorta and the aorta-to-blood uptake ratio were higher in diabetic than in non-diabetic mice. In autoradiography, the highest 18F-FMCH uptake co-localized with macrophage-rich atherosclerotic plaques. 18F-FMCH uptake in plaques correlated with levels of total cholesterol, insulin, C-peptide and leptin. In comparison with 18F-FDG, 18F-FMCH provided similar or higher plaque-to-background ratios in diabetic mice. Conclusions Type 2 diabetes enhances the uptake of choline that reflects inflammation in atherosclerotic plaques in mice. PET tracer 18F-FMCH is a potential tool to study vascular inflammation associated with diabetes. Electronic supplementary material The online version of this article (doi:10.1186/s12933-016-0340-6) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Sanna Hellberg
- Turku PET Centre, University of Turku, Kiinamyllynkatu 4-8, 20520, Turku, Finland.
| | - Johanna M U Silvola
- Turku PET Centre, University of Turku, Kiinamyllynkatu 4-8, 20520, Turku, Finland.
| | - Max Kiugel
- Turku PET Centre, University of Turku, Kiinamyllynkatu 4-8, 20520, Turku, Finland.
| | - Heidi Liljenbäck
- Turku PET Centre, University of Turku, Kiinamyllynkatu 4-8, 20520, Turku, Finland. .,Turku Center for Disease Modeling, University of Turku, Kiinamyllynkatu 10, 20520, Turku, Finland.
| | - Olli Metsälä
- Turku PET Centre, University of Turku, Kiinamyllynkatu 4-8, 20520, Turku, Finland.
| | - Tapio Viljanen
- Turku PET Centre, University of Turku, Kiinamyllynkatu 4-8, 20520, Turku, Finland.
| | - Jari Metso
- Genomics and Biomarkers Unit, National Institute for Health and Welfare, Haartmaninkatu 8, 00250, Helsinki, Finland.
| | - Matti Jauhiainen
- Genomics and Biomarkers Unit, National Institute for Health and Welfare, Haartmaninkatu 8, 00250, Helsinki, Finland.
| | - Pekka Saukko
- Department of Pathology and Forensic Medicine, University of Turku, Kiinamyllynkatu 10, 20520, Turku, Finland.
| | - Pirjo Nuutila
- Turku PET Centre, University of Turku, Kiinamyllynkatu 4-8, 20520, Turku, Finland. .,Turku PET Centre, Turku University Hospital, Kiinamyllynkatu 4-8, 20520, Turku, Finland.
| | - Seppo Ylä-Herttuala
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Neulaniementie 2, 70210, Kuopio, Finland. .,Science Service Center, Kuopio University Hospital, Puijonlaaksontie 2, 70210, Kuopio, Finland.
| | - Juhani Knuuti
- Turku PET Centre, University of Turku, Kiinamyllynkatu 4-8, 20520, Turku, Finland. .,Turku PET Centre, Turku University Hospital, Kiinamyllynkatu 4-8, 20520, Turku, Finland.
| | - Anne Roivainen
- Turku PET Centre, University of Turku, Kiinamyllynkatu 4-8, 20520, Turku, Finland. .,Turku Center for Disease Modeling, University of Turku, Kiinamyllynkatu 10, 20520, Turku, Finland. .,Turku PET Centre, Turku University Hospital, Kiinamyllynkatu 4-8, 20520, Turku, Finland.
| | - Antti Saraste
- Turku PET Centre, University of Turku, Kiinamyllynkatu 4-8, 20520, Turku, Finland. .,Turku PET Centre, Turku University Hospital, Kiinamyllynkatu 4-8, 20520, Turku, Finland. .,Heart Center, Turku University Hospital, Hämeentie 11, 20520, Turku, Finland.
| |
Collapse
|
97
|
Vacca M, Di Eusanio M, Cariello M, Graziano G, D'Amore S, Petridis FD, D'orazio A, Salvatore L, Tamburro A, Folesani G, Rutigliano D, Pellegrini F, Sabbà C, Palasciano G, Di Bartolomeo R, Moschetta A. Integrative miRNA and whole-genome analyses of epicardial adipose tissue in patients with coronary atherosclerosis. Cardiovasc Res 2015; 109:228-39. [PMID: 26645979 DOI: 10.1093/cvr/cvv266] [Citation(s) in RCA: 72] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/01/2015] [Accepted: 11/14/2015] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Epicardial adipose tissue (EAT) is an atypical fat depot surrounding the heart with a putative role in the development of atherosclerosis. METHODS AND RESULTS We profiled genes and miRNAs in perivascular EAT and subcutaneous adipose tissue (SAT) of metabolically healthy patients without coronary artery disease (CAD) vs. metabolic patients with CAD. Compared with SAT, a specific tuning of miRNAs and genes points to EAT as a tissue characterized by a metabolically active and pro-inflammatory profile. Then, we depicted both miRNA and gene signatures of EAT in CAD, featuring a down-regulation of genes involved in lipid metabolism, mitochondrial function, nuclear receptor transcriptional activity, and an up-regulation of those involved in antigen presentation, chemokine signalling, and inflammation. Finally, we identified miR-103-3p as candidate modulator of CCL13 in EAT, and a potential biomarker role for the chemokine CCL13 in CAD. CONCLUSION EAT in CAD is characterized by changes in the regulation of metabolism and inflammation with miR-103-3p/CCL13 pair as novel putative actors in EAT function and CAD.
Collapse
Affiliation(s)
- Michele Vacca
- Department of Interdisciplinary Medicine, University of Bari 'Aldo Moro', Piazza Giulio Cesare 11, 70124 Bari, Italy Fondazione Mario Negri Sud, Santa Maria Imbaro (CH), Italy Ageing Research Center (CeSI), 'G. d'Annunzio' University Foundation, Chieti, Italy
| | - Marco Di Eusanio
- Cardiovascular Department, 'S.Orsola Malpighi' Hospital, University of Bologna, Bologna, Italy
| | - Marica Cariello
- Department of Interdisciplinary Medicine, University of Bari 'Aldo Moro', Piazza Giulio Cesare 11, 70124 Bari, Italy National Cancer Research Center IRCCS 'Giovanni Paolo II', Bari, Italy
| | - Giusi Graziano
- Department of Interdisciplinary Medicine, University of Bari 'Aldo Moro', Piazza Giulio Cesare 11, 70124 Bari, Italy National Cancer Research Center IRCCS 'Giovanni Paolo II', Bari, Italy
| | - Simona D'Amore
- Department of Interdisciplinary Medicine, University of Bari 'Aldo Moro', Piazza Giulio Cesare 11, 70124 Bari, Italy Fondazione Mario Negri Sud, Santa Maria Imbaro (CH), Italy National Cancer Research Center IRCCS 'Giovanni Paolo II', Bari, Italy
| | | | | | | | | | - Gianluca Folesani
- Cardiovascular Department, 'S.Orsola Malpighi' Hospital, University of Bologna, Bologna, Italy
| | | | | | - Carlo Sabbà
- Department of Interdisciplinary Medicine, University of Bari 'Aldo Moro', Piazza Giulio Cesare 11, 70124 Bari, Italy
| | - Giuseppe Palasciano
- Department of Interdisciplinary Medicine, University of Bari 'Aldo Moro', Piazza Giulio Cesare 11, 70124 Bari, Italy
| | - Roberto Di Bartolomeo
- Cardiovascular Department, 'S.Orsola Malpighi' Hospital, University of Bologna, Bologna, Italy
| | - Antonio Moschetta
- Department of Interdisciplinary Medicine, University of Bari 'Aldo Moro', Piazza Giulio Cesare 11, 70124 Bari, Italy Fondazione Mario Negri Sud, Santa Maria Imbaro (CH), Italy National Cancer Research Center IRCCS 'Giovanni Paolo II', Bari, Italy
| |
Collapse
|
98
|
Relationship of Genetic Polymorphisms of the Chemokine, CCL5, and Its Receptor, CCR5, with Coronary Artery Disease in Taiwan. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2015; 2015:851683. [PMID: 26688689 PMCID: PMC4672136 DOI: 10.1155/2015/851683] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/06/2015] [Revised: 06/16/2015] [Accepted: 11/04/2015] [Indexed: 01/13/2023]
Abstract
The chemokine receptor CCR5 polymorphism, which confers resistance to HIV infection, has been associated with reduced risk of cardiovascular disease. However, the association of the chemokine, CCL5, and its receptor, CCR5, polymorphism and coronary artery disease (CAD) in the Taiwanese has not been studied. In this study, 483 subjects who received elective coronary angiography were recruited from Chung Shan Medical University Hospital. CCL5-403 and CCR5-59029 were determined by polymerase chain reaction-restriction fragment length polymorphism. We found that CCL5-403 with TT genotype frequencies was significantly associated with the risk of CAD group (odds ratio = 3.063 and p = 0.012). Moreover, the frequencies of CCR5-59029 with GG or GA genotype were higher than AA genotype in acute coronary syndrome individuals (odds ratio = 1.853, CI = 1.176–2.921, p = 0.008). In conclusion, we found that CCL5-403 polymorphism may increase genetic susceptibility of CAD. CCL5-403 or CCR5-59029 single nucleotide polymorphism may include genotype score and it may predict cardiovascular event.
Collapse
|
99
|
Abstract
The immune reactions that regulate atherosclerotic plaque inflammation involve chemokines, lipid mediators and costimulatory molecules. Chemokines are a family of chemotactic cytokines that mediate immune cell recruitment and control cell homeostasis and activation of different immune cell types and subsets. Chemokine production and activation of chemokine receptors form a positive feedback mechanism to recruit monocytes, neutrophils and lymphocytes into the atherosclerotic plaque. In addition, chemokine signalling affects immune cell mobilization from the bone marrow. Targeting several of the chemokines and/or chemokine receptors reduces experimental atherosclerosis, whereas specific chemokine pathways appear to be involved in plaque regression. Leukotrienes are lipid mediators that are formed locally in atherosclerotic lesions from arachidonic acid. Leukotrienes mediate immune cell recruitment and activation within the plaque as well as smooth muscle cell proliferation and endothelial dysfunction. Antileukotrienes decrease experimental atherosclerosis, and recent observational data suggest beneficial clinical effects of leukotriene receptor antagonism in cardiovascular disease prevention. By contrast, other lipid mediators, such as lipoxins and metabolites of omega-3 fatty acids, have been associated with the resolution of inflammation. Costimulatory molecules play a central role in fine-tuning immunological reactions and mediate crosstalk between innate and adaptive immunity in atherosclerosis. Targeting these interactions is a promising approach for the treatment of atherosclerosis, but immunological side effects are still a concern. In summary, targeting chemokines, leukotriene receptors and costimulatory molecules could represent potential therapeutic strategies to control atherosclerotic plaque inflammation.
Collapse
Affiliation(s)
- M Bäck
- Translational Cardiology, Department of Medicine, Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden.,Department of Cardiology, Karolinska University Hospital, Stockholm, Sweden
| | - C Weber
- Institute for Cardiovascular Prevention (IPEK), Ludwig Maximilians University, Munich, Germany.,German Center for Cardiovascular Research (DZHK), partner site Munich Heart Alliance, Munich, Germany.,Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), University of Maastricht, Maastricht, The Netherlands
| | - E Lutgens
- Institute for Cardiovascular Prevention (IPEK), Ludwig Maximilians University, Munich, Germany.,German Center for Cardiovascular Research (DZHK), partner site Munich Heart Alliance, Munich, Germany.,Department of Medical Biochemistry, Subdivision of Experimental Vascular Biology, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| |
Collapse
|
100
|
Abstract
The human immunodeficiency virus-1 (HIV-1) enters target cells by binding its envelope glycoprotein gp120 to the CD4 receptor and/or coreceptors such as C-C chemokine receptor type 5 (CCR5; R5) and C-X-C chemokine receptor type 4 (CXCR4; X4), and R5-tropic viruses predominate during the early stages of infection. CCR5 antagonists bind to CCR5 to prevent viral entry. Maraviroc (MVC) is the only CCR5 antagonist currently approved by the United States Food and Drug Administration, the European Commission, Health Canada, and several other countries for the treatment of patients infected with R5-tropic HIV-1. MVC has been shown to be effective at inhibiting HIV-1 entry into cells and is well tolerated. With expanding MVC use by HIV-1-infected humans, different clinical outcomes post-approval have been observed with MVC monotherapy or combination therapy with other antiretroviral drugs, with MVC use in humans infected with dual-R5- and X4-tropic HIV-1, infected with different HIV-1 genotype or infected with HIV-2. This review discuss the role of CCR5 in HIV-1 infection, the development of the CCR5 antagonist MVC, its pharmacokinetics, pharmacodynamics, drug-drug interactions, and the implications of these interactions on treatment outcomes, including viral mutations and drug resistance, and the mechanisms associated with the development of resistance to MVC. This review also discusses available studies investigating the use of MVC in the treatment of other diseases such as cancer, graft-versus-host disease, and inflammatory diseases.
Collapse
Affiliation(s)
- Shawna M Woollard
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, USA
| | - Georgette D Kanmogne
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, USA
| |
Collapse
|