101
|
Kapelouzou A, Giaglis S, Peroulis M, Katsimpoulas M, Moustardas P, Aravanis CV, Kostakis A, Karayannakos PE, Cokkinos DV. Overexpression of Toll-Like Receptors 2, 3, 4, and 8 Is Correlated to the Vascular Atherosclerotic Process in the Hyperlipidemic Rabbit Model: The Effect of Statin Treatment. J Vasc Res 2017; 54:156-169. [PMID: 28478461 DOI: 10.1159/000457797] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2016] [Accepted: 01/21/2017] [Indexed: 12/28/2022] Open
Abstract
BACKGROUND Atherosclerosis is the major cause of cardiovascular disease; hypercholesterolemia is a major risk factor. We hypothesized that specific TLR members (TLR2, TLR3, TLR4, TLR8) may play a role in atherosclerosis progression and its accompanying inflammatory response. We determined the association of atherosclerotic lesions and TLR mRNA expression in different aortic sites. We also assessed the effects of fluvastatin (Flu) treatment on TLR expression and plaque characteristics. METHODS Male rabbits, fed with an atherogenic diet for a duration of 3 months, were screened for advanced atherosclerotic lesions in the aorta. Additional animals received normal diet or normal diet plus Flu for 1 additional month. TLR mRNA expression in various thoracic and abdominal aortic segments was assessed, together with atherosclerotic changes. RESULTS After high lipid diet, the atherosclerotic burden increased more in the abdominal than in the thoracic aorta; TLR2, 3, 4, and 8 also increased significantly. Flu decreased atherosclerotic plaque, calcium deposition, lipid cores, intraplaque hemorrhage, erythrocyte membranes, endothelial cells, and macrophage infiltration, while increasing smooth muscle cells in plaques of both aortic segments; it also lowered TLR2, 3, 4, and 8 expression in all aortic segments to a stronger degree than resumption of normal diet. There was a strong association between blood and tissue parameters during experimental period and finally a strong correlation found between these parameters with mRNA of TLR2, 3, 4, and 8 in various stages. CONCLUSION For the first time TLR2, 3, 4, and 8 mRNA expression is prospectively explored after hypercholesterolemic diet in the rabbit model. TLR2, 3, 4, and 8 mRNA expression is strongly upregulated and correlates with the progression of atherosclerosis in the aorta. Flu significantly inhibited this progress and reduced inflammation via TLR downregulation which was strongly associated with regression of plaque morphology and atherosclerosis promoting factors.
Collapse
Affiliation(s)
- Alkistis Kapelouzou
- Center for Clinical, Experimental Surgery and Translational Research, Biomedical Research Foundation of the Academy of Athens, Athens, Greece
| | | | | | | | | | | | | | | | | |
Collapse
|
102
|
Liu CL, Santos MM, Fernandes C, Liao M, Iamarene K, Zhang JY, Sukhova GK, Shi GP. Toll-like receptor 7 deficiency protects apolipoprotein E-deficient mice from diet-induced atherosclerosis. Sci Rep 2017; 7:847. [PMID: 28405010 PMCID: PMC5429799 DOI: 10.1038/s41598-017-00977-0] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2016] [Accepted: 03/20/2017] [Indexed: 02/08/2023] Open
Abstract
Toll-like receptor 7 (TLR7) mediates autoantigen and viral RNA-induced cytokine production. Increased TLR7 expression in human atherosclerotic lesions suggests its involvement in atherogenesis. Here we demonstrated TLR7 expression in macrophages, smooth muscle cells (SMCs), and endothelial cells from mouse atherosclerotic lesions. To test a direct participation of TLR7 in atherosclerosis, we crossbred TLR7-deficient (Tlr7 -/-) mice with apolipoprotein E-deficient (Apoe -/-) mice and produced Apoe -/- Tlr7 -/- and Apoe -/- Tlr7 +/+ littermates, followed by feeding them an atherogenic diet to produce atherosclerosis. Compared to Apoe -/- Tlr7 +/+ mice, Apoe -/- Tlr7 -/- mice showed reduced aortic arch and sinus lesion areas. Reduced atherosclerosis in Apoe -/- Tlr7 -/- mice did not affect lesion macrophage-positive area and CD4+ T-cell number per lesion area, but reduced lesion expression of inflammatory markers major histocompatibility complex-class II and IL6, lesion matrix-degrading proteases cathepsin S and matrix metalloproteinase-9, and systemic serum amyloid A levels. TLR7 deficiency also reduced aortic arch SMC loss and lesion intima and media cell apoptosis. However, TLR7 deficiency did not affect aortic wall elastin fragmentation and collagen contents, or plasma lipoproteins. Therefore, TLR7 contributes to atherogenesis in Apoe -/- mice by regulating lesion and systemic inflammation. A TLR7 antagonist may mitigate atherosclerosis.
Collapse
Affiliation(s)
- Cong-Lin Liu
- Department of Cardiology, Institute of Clinical Medicine, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.,Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, 02115, USA
| | - Marcela M Santos
- Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, 02115, USA
| | - Cleverson Fernandes
- Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, 02115, USA
| | - Mengyang Liao
- Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, 02115, USA
| | - Karine Iamarene
- Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, 02115, USA
| | - Jin-Ying Zhang
- Department of Cardiology, Institute of Clinical Medicine, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Galina K Sukhova
- Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, 02115, USA
| | - Guo-Ping Shi
- Department of Cardiology, Institute of Clinical Medicine, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, China. .,Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, 02115, USA.
| |
Collapse
|
103
|
Familtseva A, Jeremic N, Kunkel GH, Tyagi SC. Toll-like receptor 4 mediates vascular remodeling in hyperhomocysteinemia. Mol Cell Biochem 2017; 433:177-194. [PMID: 28386844 DOI: 10.1007/s11010-017-3026-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2016] [Accepted: 04/01/2017] [Indexed: 01/12/2023]
Abstract
Although hyperhomocysteinemia (HHcy) is known to promote downstream pro-inflammatory cytokine elevation, the precise mechanism is still unknown. One of the possible receptors that could have significant attention in the field of hypertension is toll-like receptor 4 (TLR-4). TLR-4 is a cellular membrane protein that is ubiquitously expressed in all cell types of the vasculature. Its mutation can attenuate the effects of HHcy-mediated vascular inflammation and mitochondria- dependent cell death that suppresses hypertension. In this review, we observed that HHcy induces vascular remodeling through immunological adaptation, promoting inflammatory cytokine up-regulation (IL-1β, IL-6, TNF-α) and initiation of mitochondrial dysfunction leading to cell death and chronic vascular inflammation. The literature suggests that HHcy promotes TLR-4-driven chronic vascular inflammation and mitochondria-mediated cell death inducing peripheral vascular remodeling. In the previous studies, we have characterized the role of TLR-4 mutation in attenuating vascular remodeling in hyperhomocysteinemia. This review includes, but is not limited to, the physiological synergistic aspects of the downstream elevation of cytokines found within the vascular inflammatory cascade. These events subsequently induce mitochondrial dysfunction defined by excessive mitochondrial fission and mitochondrial apoptosis contributing to vascular remodeling followed by hypertension.
Collapse
Affiliation(s)
- Anastasia Familtseva
- Department of Physiology, School of Medicine, Health Sciences Centre, University of Louisville, A-1215, 500, South Preston Street, Louisville, KY, 40202, USA
| | - Nevena Jeremic
- Department of Physiology, School of Medicine, Health Sciences Centre, University of Louisville, A-1215, 500, South Preston Street, Louisville, KY, 40202, USA.
| | - George H Kunkel
- Department of Physiology, School of Medicine, Health Sciences Centre, University of Louisville, A-1215, 500, South Preston Street, Louisville, KY, 40202, USA
| | - Suresh C Tyagi
- Department of Physiology, School of Medicine, Health Sciences Centre, University of Louisville, A-1215, 500, South Preston Street, Louisville, KY, 40202, USA
| |
Collapse
|
104
|
Watanabe R, Zhang H, Berry G, Goronzy JJ, Weyand CM. Immune checkpoint dysfunction in large and medium vessel vasculitis. Am J Physiol Heart Circ Physiol 2017; 312:H1052-H1059. [PMID: 28314758 DOI: 10.1152/ajpheart.00024.2017] [Citation(s) in RCA: 79] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/19/2017] [Revised: 03/02/2017] [Accepted: 03/11/2017] [Indexed: 12/31/2022]
Abstract
Giant cell arteritis (GCA) is a granulomatous vasculitis of the aorta and its medium-sized branch vessels. CD4 T cells, macrophages, and dendritic cells (DCs) build granulomatous infiltrates that injure the vessel wall and elicit a maladaptive response to injury. Pathological consequences include fragmentation of elastic membranes, destruction of the medial layer, microvascular neoangiogenesis, massive outgrowth of myofibroblasts, and lumen-occlusive intimal hyperplasia. Antigens have been suspected to drive the local activation of vasculitogenic CD4 T cells, but recent data have suggested a more generalized defect in the threshold setting of such T cells, rendering them hyperreactive. Under physiological conditions, immune checkpoints provide negative signals to curb T cell activation and prevent inflammation-associated tissue destruction. This protective mechanism is disrupted in GCA. Vessel wall DCs fail to express the immunoinhibitory ligand programmed cell death ligand-1, leaving lesional T cells unchecked. Consequently, programmed cell death protein-1-positive CD4 T cells can enter the immunoprivileged vessel wall, where they produce a broad spectrum of inflammatory cytokines (interferon-γ, IL-17, and IL-21) and have a direct role in driving intimal hyperplasia and intramural neoangiogenesis. The deficiency of the programmed cell death protein-1 immune checkpoint in GCA, promoting unopposed T cell immunity, contrasts with checkpoint hyperactivity in cancer patients in whom excessive programmed cell death ligand-1 expression paralyzes the function of antitumor T cells. Excessive checkpoint activity is the principle underlying cancer-immune evasion and is therapeutically targeted by immunotherapy with checkpoint inhibitors. Such checkpoint inhibitors, which unleash anticancer T cells and induce immune-related toxicity, may lead to drug-induced vasculitis.
Collapse
Affiliation(s)
- Ryu Watanabe
- Division of Immunology and Rheumatology, Department of Medicine, Stanford University School of Medicine, Stanford, California; and
| | - Hui Zhang
- Division of Immunology and Rheumatology, Department of Medicine, Stanford University School of Medicine, Stanford, California; and
| | - Gerald Berry
- Department of Pathology, Stanford University School of Medicine, Stanford, California
| | - Jörg J Goronzy
- Division of Immunology and Rheumatology, Department of Medicine, Stanford University School of Medicine, Stanford, California; and
| | - Cornelia M Weyand
- Division of Immunology and Rheumatology, Department of Medicine, Stanford University School of Medicine, Stanford, California; and
| |
Collapse
|
105
|
Jiang D, Yang Y, Li D. Lipopolysaccharide induced vascular smooth muscle cells proliferation: A new potential therapeutic target for proliferative vascular diseases. Cell Prolif 2017; 50. [PMID: 28150467 DOI: 10.1111/cpr.12332] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2016] [Accepted: 12/30/2016] [Indexed: 12/12/2022] Open
Abstract
Vascular smooth muscle cells (VSMCs) proliferation is involved in vascular atherosclerosis and restenosis. Recent studies have demonstrated that lipopolysaccharide (LPS) promotes VSMCs proliferation, but the signalling pathways which are involved are not completely understood. The purpose of this review was to summarize the existing knowledge of the role and molecular mechanisms involved in controlling VSMCs proliferation stimulated by LPS and mediated by toll-like receptor 4 (TLR4) signalling pathways. Moreover, the potential inhibitors of TLR4 signalling for VSMCs proliferation in proliferative vascular diseases are discussed.
Collapse
Affiliation(s)
- Dehua Jiang
- Institute of Cardiovascular Disease Research, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Yu Yang
- Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Dongye Li
- Institute of Cardiovascular Disease Research, Xuzhou Medical University, Xuzhou, Jiangsu, China.,Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China
| |
Collapse
|
106
|
Immunoinhibitory checkpoint deficiency in medium and large vessel vasculitis. Proc Natl Acad Sci U S A 2017; 114:E970-E979. [PMID: 28115719 DOI: 10.1073/pnas.1616848114] [Citation(s) in RCA: 146] [Impact Index Per Article: 20.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Giant cell arteritis (GCA) causes autoimmune inflammation of the aorta and its large branches, resulting in aortic arch syndrome, blindness, and stroke. CD4+ T cells and macrophages form organized granulomatous lesions in the walls of affected arteries, destroy the tunica media, and induce ischemic organ damage through rapid intimal hyperplasia and luminal occlusion. Pathogenic mechanisms remain insufficiently understood; specifically, it is unknown whether the unopposed activation of the immune system is because of deficiency of immunoinhibitory checkpoints. Transcriptome analysis of GCA-affected temporal arteries revealed low expression of the coinhibitory ligand programmed death ligand-1 (PD-L1) concurrent with enrichment of the programmed death-1 (PD-1) receptor. Tissue-residing and ex vivo-generated dendritic cells (DC) from GCA patients were PD-L1lo, whereas the majority of vasculitic T cells expressed PD-1, suggesting inefficiency of the immunoprotective PD-1/PD-L1 immune checkpoint. DC-PD-L1 expression correlated inversely with clinical disease activity. In human artery-SCID chimeras, PD-1 blockade exacerbated vascular inflammation, enriched for PD-1+ effector T cells, and amplified tissue production of multiple T-cell effector cytokines, including IFN-γ, IL-17, and IL-21. Arteries infiltrated by PD-1+ effector T cells developed microvascular neoangiogenesis as well as hyperplasia of the intimal layer, implicating T cells in the maladaptive behavior of vessel wall endogenous cells. Thus, in GCA, a breakdown of the tissue-protective PD1/PD-L1 checkpoint unleashes vasculitic immunity and regulates the pathogenic remodeling of the inflamed arterial wall.
Collapse
|
107
|
Bomfim GF, Rodrigues FL, Carneiro FS. Are the innate and adaptive immune systems setting hypertension on fire? Pharmacol Res 2017; 117:377-393. [PMID: 28093357 DOI: 10.1016/j.phrs.2017.01.010] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/05/2016] [Revised: 12/06/2016] [Accepted: 01/09/2017] [Indexed: 02/08/2023]
Abstract
Hypertension is the most common chronic cardiovascular disease and is associated with several pathological states, being an important cause of morbidity and mortality around the world. Low-grade inflammation plays a key role in hypertension and the innate and adaptive immune systems seem to contribute to hypertension development and maintenance. Hypertension is associated with vascular inflammation, increased vascular cytokines levels and infiltration of immune cells in the vasculature, kidneys and heart. However, the mechanisms that trigger inflammation and immune system activation in hypertension are completely unknown. Cells from the innate immune system express pattern recognition receptors (PRR), which detect conserved pathogen-associated molecular patterns (PAMPs) and damage-associated molecular patterns (DAMPs) that induce innate effector mechanisms to produce endogenous signals, such as inflammatory cytokines and chemokines, to alert the host about danger. Additionally, antigen-presenting cells (APC) act as sentinels that are activated by PAMPs and DAMPs to sense the presence of the antigen/neoantigen, which ensues the adaptive immune system activation. In this context, different lymphocyte types are activated and contribute to inflammation and end-organ damage in hypertension. This review will focus on experimental and clinical evidence demonstrating the contribution of the innate and adaptive immune systems to the development of hypertension.
Collapse
Affiliation(s)
- Gisele F Bomfim
- Institute of Health Sciences, Federal University of Mato Grosso, Sinop, MT, Brazil
| | - Fernanda Luciano Rodrigues
- Department of Physiology, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, SP, Brazil
| | - Fernando S Carneiro
- Department of Pharmacology, Ribeirao Preto Medical School, University of Sao Paulo, Av Bandeirantes, 3900, 14049-900 Ribeirao Preto, SP, Brazil.
| |
Collapse
|
108
|
Ozde C, Korkmaz A, Kundi H, Oflar E, Ungan I, Xankisi V, Nurlu N. Relationship Between Plasma Levels of Soluble CD40 Ligand and the Presence and Severity of Isolated Coronary Artery Ectasia. Clin Appl Thromb Hemost 2016; 24:379-386. [PMID: 27879468 DOI: 10.1177/1076029616680476] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
OBJECTIVE We aimed to investigate whether soluble CD40 ligand (CD40L) levels are higher in patients with isolated coronary artery ectasia (CAE) compared to patients with angiographically normal coronary arteries and those with stable coronary artery disease (CAD). MATERIALS AND METHODS In all, 55 patients with isolated CAE without stenosis, 55 with stable CAD, and 55 control participants with angiographically normal coronary arteries were included. The CAE severity was determined according to the Markis classification. Plasma levels of soluble CD40 ligand were measured by enzyme-linked immunosorbent assay. RESULTS The baseline characteristics of the 3 groups were similar. Plasma levels of soluble CD40 ligand were significantly higher in patients with CAE and CAD than in controls (2.6 ± 3.1 ng/mL and 2.0 ± 3.1 ng/mL vs 1.8 ± 2.1 ng/mL, P = .004). No difference was found between the CAE and CAD groups. Soluble CD40 ligand level was significantly higher in the type 1 Markis subgroup than that in the type 3 or type 4 subgroups ( P = .01). A receiver operating characteristic curve analysis revealed that soluble CD40 ligand level >1.2 ng/mL identified patients with isolated CAE. CONCLUSION Significantly higher levels of soluble CD40 ligand were detected in patients with CAE than that in control participants with normal coronary arteries, suggesting that soluble CD40 ligand may be involved in the pathogenesis of CAE. The CD40-CD40 ligand system likely plays a role in the pathogenesis of CAE.
Collapse
Affiliation(s)
- Cem Ozde
- 1 Department of Cardiology, Cankiri State Hospital, Cankiri, Turkey
| | - Ahmet Korkmaz
- 2 Department of Cardiology, Ankara Numune Education and Research Hospital, Ankara, Turkey
| | - Harun Kundi
- 2 Department of Cardiology, Ankara Numune Education and Research Hospital, Ankara, Turkey
| | - Ersan Oflar
- 3 Department of Cardiology, Istanbul Bakırkoy Dr. Sadi Konuk Training and Research Hospital, Istanbul, Turkey
| | - Ismail Ungan
- 3 Department of Cardiology, Istanbul Bakırkoy Dr. Sadi Konuk Training and Research Hospital, Istanbul, Turkey
| | - Vusal Xankisi
- 3 Department of Cardiology, Istanbul Bakırkoy Dr. Sadi Konuk Training and Research Hospital, Istanbul, Turkey
| | - Nilhan Nurlu
- 4 Department of Clinical Biochemistry, Istanbul Gaziosmanpaşa Taksim Training and Research Hospital, Istanbul, Turkey
| |
Collapse
|
109
|
Hara T, Nakashima Y, Sakai Y, Nishio H, Motomura Y, Yamasaki S. Kawasaki disease: a matter of innate immunity. Clin Exp Immunol 2016; 186:134-143. [PMID: 27342882 PMCID: PMC5054572 DOI: 10.1111/cei.12832] [Citation(s) in RCA: 87] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/22/2016] [Indexed: 12/26/2022] Open
Abstract
Kawasaki disease (KD) is an acute systemic vasculitis of childhood that does not have a known cause or aetiology. The epidemiological features (existence of epidemics, community outbreaks and seasonality), unique age distribution and clinical symptoms and signs of KD suggest that the disease is caused by one or more infectious environmental triggers. However, KD is not transmitted person-to-person and does not occur in clusters within households, schools or nurseries. KD is a self-limited illness that is not associated with the production of autoantibodies or the deposition of immune complexes, and it rarely recurs. Regarding the underlying pathophysiology of KD, innate immune activity (the inflammasome) is believed to play a role in the development of KD vasculitis, based on the results of studies with animal models and the clinical and laboratory findings of KD patients. Animal studies have demonstrated that innate immune pathogen-associated molecular patterns (PAMPs) can cause vasculitis independently of acquired immunity and have provided valuable insights regarding the underlying mechanisms of this phenomenon. To validate this concept, we recently searched for KD-specific PAMPs and identified such molecules with high specificity and sensitivity. These molecules have structures similar to those of microbe-associated molecular patterns (MAMPs), as shown by liquid chromatography-tandem mass spectrometry. We propose herein that KD is an innate immune disorder resulting from the exposure of a genetically predisposed individual to microbe-derived innate immune stimulants and that it is not a typical infectious disease.
Collapse
Affiliation(s)
- T Hara
- Fukuoka Children's Hospital.
- Department of Pediatrics, Graduate School of Medical Sciences.
- Division of Molecular Immunology, Research Center for Infectious Diseases, Medical Institute of Bioregulation, Kyushu University, Fukuoka, Japan.
| | - Y Nakashima
- Department of Pediatrics, Graduate School of Medical Sciences
| | - Y Sakai
- Department of Pediatrics, Graduate School of Medical Sciences
| | - H Nishio
- Department of Pediatrics, Graduate School of Medical Sciences
| | - Y Motomura
- Department of Pediatrics, Graduate School of Medical Sciences
- Division of Molecular Immunology, Research Center for Infectious Diseases, Medical Institute of Bioregulation, Kyushu University, Fukuoka, Japan
| | - S Yamasaki
- Division of Molecular Immunology, Research Center for Infectious Diseases, Medical Institute of Bioregulation, Kyushu University, Fukuoka, Japan
| |
Collapse
|
110
|
Xiao Y, Zhou J, Wei X, Sun Y, Zhang L, Feng J, Feng R, Zhao Z, Jing Z. Outcomes of different treatments on Takayasu's arteritis. J Thorac Dis 2016; 8:2495-2503. [PMID: 27747001 DOI: 10.21037/jtd.2016.08.12] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
BACKGROUND Takayasu's arteritis (TA) is a nonspecific chronic inflammation of the aorta and its branches. This study compared the outcomes of surgical treatments including bypass surgery, cutting balloon angioplasty and conventional balloon angioplasty to TA patients exhibiting supra-aortic arterial (SAA). METHODS This retrospective study was conducted on 42 TA patients, obtained from hospital database, who underwent surgical therapy due to SAA lesions from January 2010 to March 2015. Ten patients were reconstructed using cutting balloon angioplasty, 16 patients received conventional balloon angioplasty and 16 patients from bypass surgery. The primary patency, recurrent symptoms, re-intervention, early (<30 days) and late complications associated with treatment were evaluated. RESULTS In the conventional balloon angioplasty group, two patients were converted to bypass surgery as the guidewire could not traverse the lesions. The follow-up at 30.07±17.96 months (range, 1-60 months) showed restenosis or occlusion development in 40.9% arteries in conventional balloon angioplasty, compared with 6.3% after bypass surgery (P=0.018). The restenosis or occlusion rate between cutting balloon angioplasty and conventional balloon angioplasty groups were insignificant (P=0.738). In the re-intervention, three out of four (75%) treated by cutting balloon angioplasty were patent as compared to the three out of nine arteries (33.3%) dealt with by conventional angioplasty that was patent (P=0.266). Intracerebral hemorrhage (n=1) was developed in the bypass surgery group. Mortality was not observed in any of the groups. CONCLUSIONS Cutting balloon angioplasty can be considered as a safe, effective, and less-invasive alternative for non-diffuse SAA lesions, especially in young TA patients. However, bypass surgery has better primary patency rate than endovascular treatment.
Collapse
Affiliation(s)
- Yu Xiao
- Department of Vascular Surgery, Changhai Hospital, Second Military Medical University, Shanghai 200433, China
| | - Jian Zhou
- Department of Vascular Surgery, Changhai Hospital, Second Military Medical University, Shanghai 200433, China
| | - Xiaolong Wei
- Department of Vascular Surgery, Changhai Hospital, Second Military Medical University, Shanghai 200433, China
| | - Yudong Sun
- Department of Vascular Surgery, Changhai Hospital, Second Military Medical University, Shanghai 200433, China
| | - Lei Zhang
- Department of Vascular Surgery, Changhai Hospital, Second Military Medical University, Shanghai 200433, China
| | - Jiaxuan Feng
- Department of Vascular Surgery, Changhai Hospital, Second Military Medical University, Shanghai 200433, China
| | - Rui Feng
- Department of Vascular Surgery, Changhai Hospital, Second Military Medical University, Shanghai 200433, China
| | - Zhiqing Zhao
- Department of Vascular Surgery, Changhai Hospital, Second Military Medical University, Shanghai 200433, China
| | - Zaiping Jing
- Department of Vascular Surgery, Changhai Hospital, Second Military Medical University, Shanghai 200433, China
| |
Collapse
|
111
|
Goulopoulou S, McCarthy CG, Webb RC. Toll-like Receptors in the Vascular System: Sensing the Dangers Within. Pharmacol Rev 2016; 68:142-67. [PMID: 26721702 DOI: 10.1124/pr.114.010090] [Citation(s) in RCA: 170] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Toll-like receptors (TLRs) are components of the innate immune system that respond to exogenous infectious ligands (pathogen-associated molecular patterns, PAMPs) and endogenous molecules that are released during host tissue injury/death (damage-associated molecular patterns, DAMPs). Interaction of TLRs with their ligands leads to activation of downstream signaling pathways that induce an immune response by producing inflammatory cytokines, type I interferons (IFN), and other inflammatory mediators. TLR activation affects vascular function and remodeling, and these molecular events prime antigen-specific adaptive immune responses. Despite the presence of TLRs in vascular cells, the exact mechanisms whereby TLR signaling affects the function of vascular tissues are largely unknown. Cardiovascular diseases are considered chronic inflammatory conditions, and accumulating data show that TLRs and the innate immune system play a determinant role in the initiation and development of cardiovascular diseases. This evidence unfolds a possibility that targeting TLRs and the innate immune system may be a novel therapeutic goal for these conditions. TLR inhibitors and agonists are already in clinical trials for inflammatory conditions such as asthma, cancer, and autoimmune diseases, but their study in the context of cardiovascular diseases is in its infancy. In this article, we review the current knowledge of TLR signaling in the cardiovascular system with an emphasis on atherosclerosis, hypertension, and cerebrovascular injury. Furthermore, we address the therapeutic potential of TLR as pharmacological targets in cardiovascular disease and consider intriguing research questions for future study.
Collapse
Affiliation(s)
- Styliani Goulopoulou
- Institute for Cardiovascular and Metabolic Diseases, Department of Obstetrics and Gynecology, University of North Texas Health Science Center, Fort Worth, Texas; and Department of Physiology, Augusta University, Augusta, Georgia
| | - Cameron G McCarthy
- Institute for Cardiovascular and Metabolic Diseases, Department of Obstetrics and Gynecology, University of North Texas Health Science Center, Fort Worth, Texas; and Department of Physiology, Augusta University, Augusta, Georgia
| | - R Clinton Webb
- Institute for Cardiovascular and Metabolic Diseases, Department of Obstetrics and Gynecology, University of North Texas Health Science Center, Fort Worth, Texas; and Department of Physiology, Augusta University, Augusta, Georgia
| |
Collapse
|
112
|
Familtseva A, Chaturvedi P, Kalani A, Jeremic N, Metreveli N, Kunkel GH, Tyagi SC. Toll-like receptor 4 mutation suppresses hyperhomocysteinemia-induced hypertension. Am J Physiol Cell Physiol 2016; 311:C596-C606. [PMID: 27488663 DOI: 10.1152/ajpcell.00088.2016] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2016] [Accepted: 08/01/2016] [Indexed: 01/06/2023]
Abstract
Hyperhomocysteinemia (HHcy) has been observed to promote hypertension, but the mechanisms are unclear. Toll-like receptor 4 (TLR-4) is a cellular membrane protein that is ubiquitously expressed in all cell types of the vasculature. TLR-4 activation has been known to promote inflammation that has been associated with the pathogenesis of hypertension. In this study we hypothesize that HHcy induces hypertension by TLR-4 activation, which promotes inflammatory cytokine (IL-1β, IL-6, and TNF-α) upregulation and initiation of mitochondria-dependent apoptosis, leading to cell death and chronic vascular inflammation. To test this hypothesis, we used C57BL/6J (WT) mice, cystathionine β-synthase (CBS)-deficient (CBS+/-) mice with genetic mild HHcy, C3H/HeJ (C3H) mice with TLR-4 mutation, and mice with combined genetic HHcy and TLR-4 mutation (CBS+/-/C3H). Ultrasonography of the superior mesenteric artery (SMA) detected an increase in wall-to-lumen ratio, resistive index (RI), and pulsatility index (PI). Tail cuff blood pressure (BP) measurement revealed elevated BP in CBS+/- mice. RI, PI, and wall-to-lumen ratio of the SMA in CBS+/-/C3H mice were similar to the control group, and BP was significantly alleviated. TLR-4, IL-1β, IL-6, and TNF-α expression were upregulated in the SMA of CBS+/- mice and reduced in the SMA of CBS+/-/C3H mice. Molecules involved in the mitochondria-mediated cell death pathway (BAX, caspase-9, and caspase-3) were upregulated in CBS+/- mice and attenuated in CBS+/-/C3H mice. We conclude that HHcy promotes TLR-4-driven chronic vascular inflammation and mitochondria-mediated cell death, inducing hypertension. TLR-4 mutation attenuates vascular inflammation and cell death, which suppress hypertension.
Collapse
Affiliation(s)
- Anastasia Familtseva
- Department of Physiology, School of Medicine, University of Louisville, Louisville, Kentucky
| | - Pankaj Chaturvedi
- Department of Physiology, School of Medicine, University of Louisville, Louisville, Kentucky
| | - Anuradha Kalani
- Department of Physiology, School of Medicine, University of Louisville, Louisville, Kentucky
| | - Nevena Jeremic
- Department of Physiology, School of Medicine, University of Louisville, Louisville, Kentucky
| | - Naira Metreveli
- Department of Physiology, School of Medicine, University of Louisville, Louisville, Kentucky
| | - George H Kunkel
- Department of Physiology, School of Medicine, University of Louisville, Louisville, Kentucky
| | - Suresh C Tyagi
- Department of Physiology, School of Medicine, University of Louisville, Louisville, Kentucky
| |
Collapse
|
113
|
Dejaco C, Duftner C, Buttgereit F, Matteson EL, Dasgupta B. The spectrum of giant cell arteritis and polymyalgia rheumatica: revisiting the concept of the disease. Rheumatology (Oxford) 2016; 56:506-515. [DOI: 10.1093/rheumatology/kew273] [Citation(s) in RCA: 67] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2015] [Indexed: 01/17/2023] Open
|
114
|
Maugeri N, Rovere-Querini P, Manfredi AA. Disruption of a Regulatory Network Consisting of Neutrophils and Platelets Fosters Persisting Inflammation in Rheumatic Diseases. Front Immunol 2016; 7:182. [PMID: 27242789 PMCID: PMC4871869 DOI: 10.3389/fimmu.2016.00182] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2016] [Accepted: 04/29/2016] [Indexed: 12/16/2022] Open
Abstract
A network of cellular interactions that involve blood leukocytes and platelets maintains vessel homeostasis. It plays a critical role in the response to invading microbes by recruiting intravascular immunity and through the generation of neutrophil extracellular traps (NETs) and immunothrombosis. Moreover, it enables immune cells to respond to remote chemoattractants by crossing the endothelial barrier and reaching sites of infection. Once the network operating under physiological conditions is disrupted, the reciprocal activation of cells in the blood and the vessel walls determines the vascular remodeling via inflammatory signals delivered to stem/progenitor cells. A deregulated leukocyte/mural cell interaction is an early critical event in the natural history of systemic inflammation. Despite intense efforts, the signals that initiate and sustain the immune-mediated vessel injury, or those that enforce the often-prolonged phases of clinical quiescence in patients with vasculitis, have only been partially elucidated. Here, we discuss recent evidence that implicates the prototypic damage-associated molecular pattern/alarmin, the high mobility group box 1 (HMGB1) protein in systemic vasculitis and in the vascular inflammation associated with systemic sclerosis. HMGB1 could represent a player in the pathogenesis of rheumatic diseases and an attractive target for molecular interventions.
Collapse
Affiliation(s)
- Norma Maugeri
- San Raffaele Scientific Institute, Università Vita Salute San Raffaele , Milano , Italy
| | | | - Angelo A Manfredi
- San Raffaele Scientific Institute, Università Vita Salute San Raffaele , Milano , Italy
| |
Collapse
|
115
|
Watanabe R, Goronzy JJ, Berry G, Liao YJ, Weyand CM. Giant Cell Arteritis: From Pathogenesis to Therapeutic Management. CURRENT TREATMENT OPTIONS IN RHEUMATOLOGY 2016; 2:126-137. [PMID: 27298757 DOI: 10.1007/s40674-016-0043-x] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Affiliation(s)
- Ryu Watanabe
- Division of Immunology and Rheumatology, Department of Medicine, Stanford University School of Medicine, Stanford, CA
| | - Jörg J Goronzy
- Division of Immunology and Rheumatology, Department of Medicine, Stanford University School of Medicine, Stanford, CA
| | - Gerald Berry
- Department of Pathology, Stanford University School of Medicine, Stanford, CA
| | - Y Joyce Liao
- Department of Ophthalmology, Stanford Byers Eye Institute, Stanford, CA
| | - Cornelia M Weyand
- Division of Immunology and Rheumatology, Department of Medicine, Stanford University School of Medicine, Stanford, CA
| |
Collapse
|
116
|
Salvador B, Arranz A, Francisco S, Córdoba L, Punzón C, Llamas MÁ, Fresno M. Modulation of endothelial function by Toll like receptors. Pharmacol Res 2016; 108:46-56. [PMID: 27073018 DOI: 10.1016/j.phrs.2016.03.038] [Citation(s) in RCA: 56] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/22/2016] [Revised: 03/30/2016] [Accepted: 03/30/2016] [Indexed: 12/23/2022]
Abstract
Endothelial cells (EC) are able to actively control vascular permeability, coagulation, blood pressure and angiogenesis. Most recently, a role for endothelial cells in the immune response has been described. Therefore, the endothelium has a dual role controlling homeostasis but also being the first line for host defence and tissue damage repair thanks to its ability to mount an inflammatory response. Endothelial cells have been shown to express pattern-recognition receptors (PRR) including Toll-like receptors (TLR) that are activated in response to stimuli within the bloodstream including pathogens and damage signals. TLRs are strategic mediators of the immune response in endothelial cells but they also regulate the angiogenic process critical for tissue repair. Nevertheless, endothelial activation and angiogenesis can contribute to some pathologies. Thus, inappropriate endothelial activation, also known as endothelial dysfunction, through TLRs contributes to tissue damage during autoimmune and inflammatory diseases such as atherosclerosis, hypertension, ischemia and diabetes associated cardiovascular diseases. Also TLR induced angiogenesis is required for the growth of some tumors, atherosclerosis and rheumatoid arthritis, among others. In this review we discuss the importance of various TLRs in modulating the activation of endothelial cells and their importance in immunity to infection and vascular disease as well as their potential as therapeutic targets.
Collapse
Affiliation(s)
| | - Alicia Arranz
- Centro de Biología Molecular "Severo Ochoa", Universidad Autónoma de Madrid, Madrid, Spain.
| | - Sara Francisco
- Diomune SL, Parque Científico de Madrid, Madrid, Spain; Centro de Biología Molecular "Severo Ochoa", Universidad Autónoma de Madrid, Madrid, Spain.
| | - Laura Córdoba
- Diomune SL, Parque Científico de Madrid, Madrid, Spain.
| | - Carmen Punzón
- Diomune SL, Parque Científico de Madrid, Madrid, Spain.
| | | | - Manuel Fresno
- Diomune SL, Parque Científico de Madrid, Madrid, Spain; Centro de Biología Molecular "Severo Ochoa", Universidad Autónoma de Madrid, Madrid, Spain.
| |
Collapse
|
117
|
Schmidt J, Duhaut P. Atteinte aortique dans la maladie de Horton. Rev Med Interne 2016; 37:239-44. [DOI: 10.1016/j.revmed.2015.12.018] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2015] [Revised: 11/12/2015] [Accepted: 12/21/2015] [Indexed: 01/16/2023]
|
118
|
O'Neill L, Molloy ES. The role of toll like receptors in giant cell arteritis. Rheumatology (Oxford) 2016; 55:1921-1931. [PMID: 26893518 DOI: 10.1093/rheumatology/kew001] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2015] [Revised: 01/07/2016] [Indexed: 12/21/2022] Open
Abstract
GCA is a common primary systemic vasculitis that results in granulomatous inflammation of medium to large arteries. Both innate and adaptive immune mechanisms combine to drive intimal hyperplasia, luminal stenosis and ultimately occlusion. While the pathogenesis of GCA is incompletely understood, the activation of resident adventitial dendritic cells via toll like receptors (TLRs) appears to be a crucial inciting event. Here we explore the role of TLRs in the pathogenesis of GCA, including their effects on dendritic cell and T cell activation and recruitment, putative infectious triggers for GCA and the potential of TLR inhibition as a novel therapeutic strategy in GCA.
Collapse
Affiliation(s)
- Lorraine O'Neill
- Centre for Arthritis and Rheumatic Diseases, St Vincent's University Hospital, Dublin Academic Medical Centre, Elm Park, Dublin, 4, Ireland
| | - Eamonn S Molloy
- Centre for Arthritis and Rheumatic Diseases, St Vincent's University Hospital, Dublin Academic Medical Centre, Elm Park, Dublin, 4, Ireland
| |
Collapse
|
119
|
Frohman L, Wong ABC, Matheos K, Leon-Alvarado LG, Danesh-Meyer HV. New developments in giant cell arteritis. Surv Ophthalmol 2016; 61:400-21. [PMID: 26774550 DOI: 10.1016/j.survophthal.2016.01.001] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2015] [Revised: 01/05/2016] [Accepted: 01/08/2016] [Indexed: 12/01/2022]
Abstract
Giant cell arteritis (GCA) is a medium-to-large vessel vasculitis with potentially sight- and life- threatening complications. Our understanding of the pathogenesis, diagnosis, and treatment of GCA has advanced rapidly in recent times. The validity of using the American College of Rheumatology guidelines for diagnosis of GCA in a clinical setting has been robustly challenged. Erythrocyte sedimentation rate, an important marker of inflammation, is lowered by the use of statins and nonsteroidal anti-inflammatory drugs. Conversely, it may be falsely elevated with a low hematocrit. Despite the emergence of new diagnostic modalities, temporal artery biopsy remains the gold standard. Evidence suggests that shorter biopsy lengths and biopsies done weeks to months after initiation of steroid therapy are still useful. New imaging techniques such as positron emission tomography have shown that vascular inflammation in GCA is more widespread than originally thought. GCA, Takayasu arteritis, and polymyalgia rheumatica are no longer thought to exist as distinct entities and are more likely parts of a spectrum of disease. A range of immunosuppressive drugs have been used in conjunction with corticosteroids to treat GCA. In particular, interleukin-6 inhibitors are showing promise as a therapy.
Collapse
Affiliation(s)
- Larry Frohman
- Department of Ophthalmology, Rutgers-New Jersey Medical School, New Jersey, USA; Department of Neurosciences, Rutgers-New Jersey Medical School, New Jersey, USA
| | - Aaron B C Wong
- Department of Ophthalmology, University of Auckland, Auckland, New Zealand
| | - Kaliopy Matheos
- Department of Ophthalmology, University of Auckland, Auckland, New Zealand
| | | | | |
Collapse
|
120
|
Sata M. Cuff-Induced Neointimal Formation in Mouse Models. MOUSE MODELS OF VASCULAR DISEASES 2016. [PMCID: PMC7122099 DOI: 10.1007/978-4-431-55813-2_2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Ischemic heart failure caused by atherosclerosis is a major cause of death worldwide. Although remarkable technological advances have been made in the treatment of coronary heart disease, there is as yet no treatment that can sufficiently suppress the progression of atherosclerosis, including neointimal thickening. Therefore, a precise understanding of the mechanism of neointimal hyperplasia will provide the development of new technologies. Both ApoE-KO and LDLR-KO mice have been employed to generate other relevant mouse models of cardiovascular disease through breeding strategies. Although these mice are effective tools for the investigation of atherosclerosis, development of a progressive atherosclerotic lesion takes a long time, resulting in increase of both the costs and the space needed for the research. Thus, it is necessary to develop simpler tools that would allow easy evaluation of atherosclerosis in mouse models. In this review, we discuss our experience in generating mouse models of cuff-induced injury of the femoral artery and attempt to provide a better understanding of cuff-induced neointimal formation.
Collapse
|
121
|
Wang Y, Song E, Bai B, Vanhoutte PM. Toll-like receptors mediating vascular malfunction: Lessons from receptor subtypes. Pharmacol Ther 2015; 158:91-100. [PMID: 26702901 DOI: 10.1016/j.pharmthera.2015.12.005] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Toll-like receptors (TLR) are a subfamily of pattern recognition receptors (PRR) implicated in a variety of vascular abnormalities. However, the pathophysiological role and the interplay between different TLR-mediated innate and adaptive immune responses during the development of vascular diseases remain largely unspecified. TLR are widely distributed in both immune and nonimmune cells in the blood vessel wall. The expressions and locations of TLR are dynamically regulated in response to distinct molecular patterns derived from pathogens or damaged host cells. As a result, the outcome of TLR signaling is agonist- and cell type-dependent. A better understanding of discrete TLR signaling pathways in the vasculature will provide unprecedented opportunities for the discovery of novel therapies in many inflammatory vascular diseases. The present brief review discusses the role of individual TLR in controlling cellular functions of the vascular system, by focusing on the inflammatory responses within the blood vessel wall which contribute to the development of hypertension and atherosclerosis.
Collapse
Affiliation(s)
- Yu Wang
- State Key Laboratory of Pharmaceutical Biotechnology and Department of Pharmacology and Pharmacy, The University of Hong Kong, Hong Kong, China.
| | - Erfei Song
- State Key Laboratory of Pharmaceutical Biotechnology and Department of Pharmacology and Pharmacy, The University of Hong Kong, Hong Kong, China
| | - Bo Bai
- State Key Laboratory of Pharmaceutical Biotechnology and Department of Pharmacology and Pharmacy, The University of Hong Kong, Hong Kong, China
| | - Paul M Vanhoutte
- State Key Laboratory of Pharmaceutical Biotechnology and Department of Pharmacology and Pharmacy, The University of Hong Kong, Hong Kong, China.
| |
Collapse
|
122
|
Samson M, Bonnotte B. [Pathogenesis of large vessel vasculitis]. Rev Med Interne 2015; 37:264-73. [PMID: 26620872 DOI: 10.1016/j.revmed.2015.10.350] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2015] [Revised: 08/24/2015] [Accepted: 10/27/2015] [Indexed: 01/06/2023]
Abstract
Giant cell arteritis (GCA) and Takayasu's arteritis (TA) are two granulomatous vasculitis affecting large arteries that present specific epidemiological and clinical features. Their pathogenesis is not fully understood but major advances have been obtained during the last years, thus allowing the emergence of new therapeutic strategies. GCA and TA develop on a specific genetic background but share some similarities regarding the immunological pathways involved in their pathogenesis. The trigger of these diseases is not clearly identified but it is thought that an infectious agent could activate and lead to the maturation of dendritic cells that are localized in the adventitia of arteries. Then, the cells of the adaptative immune response are recruited and activated: CD4 T cells that polarize into Th1 and Th17 cells, cytotoxic CD8 T cells and Natural Killer cells. Furthermore, the T regulatory cells (Treg) are decreased both in GCA and TA. Humoral immune response seems also to be involved, especially in TA. Then, the cytokines produced by T lymphocytes (especially IL-17 and IFN-γ) trigger the recruitment and activation of monocytes and their differentiation into macrophages and multinuclear giant cells that produce IL-1β and IL-6 that are responsible for general symptoms of GCA and TA, and cytotoxic mediators and growth factors that trigger the remodeling of the arterial wall leading to aneurysms and ischemic manifestations of GCA an TA.
Collapse
Affiliation(s)
- M Samson
- Service de médecine interne et immunologie clinique, hôpital François Mitterrand, CHU de Dijon, 21000 Dijon, France; Inserm, UMR1098, 25020 Besançon cedex, France; Faculté de médecine, université de Bourgogne, IFR100, Dijon, France
| | - B Bonnotte
- Service de médecine interne et immunologie clinique, hôpital François Mitterrand, CHU de Dijon, 21000 Dijon, France; Inserm, UMR1098, 25020 Besançon cedex, France; Faculté de médecine, université de Bourgogne, IFR100, Dijon, France.
| |
Collapse
|
123
|
Ungprasert P, Thongprayoon C, Warrington KJ. Lower body mass index is associated with a higher risk of giant cell arteritis: a systematic review and meta-analysis. ANNALS OF TRANSLATIONAL MEDICINE 2015; 3:232. [PMID: 26539449 DOI: 10.3978/j.issn.2305-5839.2015.09.31] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
OBJECTIVE To characterize the possible association between body mass index (BMI) and risk of giant cell arteritis (GCA). METHODS We conducted a systematic review of observational studies (case-control or cohort study) that (I) reported BMI of patients with GCA prior to the diagnosis of GCA compared with subjects without GCA and (II) provided relative risk (RR), odds ratio (OR) or hazard ratio (HR) with 95% confidence interval (CI) from its regression analysis. Meta-analysis of the included studies was then performed to estimate the pooled effect using generic variance method of DerSimonian and Laird. RESULTS Three studies encompassing 141 patients with GCA and 85,736 controls met our eligibility criteria and were included in the data analyses. We demonstrated a statistically significant inverse relationship between BMI and risk of subsequent development of GCA as the risk increased by 8% when BMI was reduced by 1.0 kg/m(2) (pooled OR of 0.92/kg/m(2); 95% CI, 0.88-0.96). CONCLUSIONS Our study demonstrated a statistically significant inverse relationship between BMI and risk of subsequent development GCA. The pathophysiologic link behind this negative correlation is not well-characterized and further investigation is required.
Collapse
Affiliation(s)
- Patompong Ungprasert
- 1 Division of Rheumatology, Department of Internal Medicine, Mayo Clinic, Rochester, MN 55905, USA ; 2 Division of Rheumatology, Department of Medicine, Faculty of Medicine Siriraj Hospital, Bangkok, Thailand ; 3 Division of Nephrology and Hypertension, Department of Internal Medicine, Mayo Clinic, Rochester, MN 55905, USA
| | - Charat Thongprayoon
- 1 Division of Rheumatology, Department of Internal Medicine, Mayo Clinic, Rochester, MN 55905, USA ; 2 Division of Rheumatology, Department of Medicine, Faculty of Medicine Siriraj Hospital, Bangkok, Thailand ; 3 Division of Nephrology and Hypertension, Department of Internal Medicine, Mayo Clinic, Rochester, MN 55905, USA
| | - Kenneth J Warrington
- 1 Division of Rheumatology, Department of Internal Medicine, Mayo Clinic, Rochester, MN 55905, USA ; 2 Division of Rheumatology, Department of Medicine, Faculty of Medicine Siriraj Hospital, Bangkok, Thailand ; 3 Division of Nephrology and Hypertension, Department of Internal Medicine, Mayo Clinic, Rochester, MN 55905, USA
| |
Collapse
|
124
|
Balistreri CR. Genetic contribution in sporadic thoracic aortic aneurysm? Emerging evidence of genetic variants related to TLR-4-mediated signaling pathway as risk determinants. Vascul Pharmacol 2015; 74:1-10. [PMID: 26409318 DOI: 10.1016/j.vph.2015.09.006] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2015] [Revised: 08/26/2015] [Accepted: 09/23/2015] [Indexed: 01/16/2023]
Affiliation(s)
- Carmela Rita Balistreri
- Department of Pathobiology and Medical Biotechnologies, University of Palermo, Corso Tukory 211, Palermo 90134, Italy.
| |
Collapse
|
125
|
Khakpour S, Wilhelmsen K, Hellman J. Vascular endothelial cell Toll-like receptor pathways in sepsis. Innate Immun 2015; 21:827-46. [DOI: 10.1177/1753425915606525] [Citation(s) in RCA: 135] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2015] [Accepted: 08/11/2015] [Indexed: 12/20/2022] Open
Abstract
The endothelium forms a vast network that dynamically regulates vascular barrier function, coagulation pathways and vasomotor tone. Microvascular endothelial cells are uniquely situated to play key roles during infection and injury, owing to their widespread distribution throughout the body and their constant interaction with circulating blood. While not viewed as classical immune cells, endothelial cells express innate immune receptors, including the Toll-like receptors (TLRs), which activate intracellular inflammatory pathways mediated through NF-κB and the MAP kinases. TLR agonists, including LPS and bacterial lipopeptides, directly upregulate microvascular endothelial cell expression of inflammatory mediators. Intriguingly, TLR activation also modulates microvascular endothelial cell permeability and the expression of coagulation pathway intermediaries. Microvascular thrombi have been hypothesized to trap microorganisms thereby limiting the spread of infection. However, dysregulated activation of endothelial inflammatory pathways is also believed to lead to coagulopathy and increased vascular permeability, which together promote sepsis-induced organ failure. This article reviews vascular endothelial cell innate immune pathways mediated through the TLRs as they pertain to sepsis, highlighting links between TLRs and coagulation and permeability pathways, and their role in healthy and pathologic responses to infection and sepsis.
Collapse
Affiliation(s)
- Samira Khakpour
- Department of Anesthesia and Perioperative Care, University of California, San Francisco, CA, USA
- Biomedical Sciences and Immunology Programs, University of California, San Francisco, CA, USA
| | - Kevin Wilhelmsen
- Department of Anesthesia and Perioperative Care, University of California, San Francisco, CA, USA
| | - Judith Hellman
- Department of Anesthesia and Perioperative Care, University of California, San Francisco, CA, USA
- Biomedical Sciences and Immunology Programs, University of California, San Francisco, CA, USA
| |
Collapse
|
126
|
Ungprasert P, Upala S, Sanguankeo A, Warrington KJ. Patients with giant cell arteritis have a lower prevalence of diabetes mellitus: A systematic review and meta-analysis. Mod Rheumatol 2015; 26:410-4. [PMID: 26381748 DOI: 10.3109/14397595.2015.1081722] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
OBJECTIVE The aim of this study is to compare the prevalence of diabetes mellitus (DM) in patients who are recently diagnosed with giant cell arteritis (GCA) with age- and sex-matched controls. METHODS We conducted a systematic review and meta-analysis of observational studies that (1) consisted of GCA cohort and non-GCA cohort that was randomly selected from the same population and (2) provided prevalences of DM at the time of diagnosis for patients with GCA and at the index date for controls. Pooled odds ratios and 95% confidence intervals (CIs) were calculated using a random-effect, Mantel-Haenszel analysis. RESULTS Five studies with 903 patients with GCA and 1064 controls were identified and included in our data analysis. We demonstrated a statistically significant lower prevalence of DM among patients with GCA with the pooled OR of 0.74 (95% CI, 0.57-0.97). CONCLUSION At diagnosis, patients with GCA had a lower prevalence of DM. Whether DM could be a protective factor against the development of GCA needs further investigations.
Collapse
Affiliation(s)
- Patompong Ungprasert
- a Department of Internal Medicine , Division of Rheumatology, Mayo Clinic , Rochester , MN , USA .,b Department of Medicine , Faculty of Medicine Siriraj Hospital, Division of Rheumatology , Bangkok , Thailand , and
| | - Sikarin Upala
- c Department of Medicine , Bassett Medical Center , New York , NY , USA
| | - Anawin Sanguankeo
- c Department of Medicine , Bassett Medical Center , New York , NY , USA
| | - Kenneth J Warrington
- a Department of Internal Medicine , Division of Rheumatology, Mayo Clinic , Rochester , MN , USA
| |
Collapse
|
127
|
Abstract
Giant cell arteritis (GCA) is a large-vessel vasculitis predominantly affecting older people, with a peak incidence between 70 and 79 years of age. If untreated, ischaemic complications can be catastrophic for the patient, including blindness. We review the current treatment paradigms for this condition, the mainstay of which is immediate high-dose glucocorticoid therapy with a gradual dose tapering. Adverse events of glucocorticoid therapy are often observed after 12-24 months and corticosteroid-sparing adjuvant therapies are used in severe disease, multiple flares or patients at high risk of prolonged therapy. The current understanding of the pathogenesis of GCA is explored. This has informed the identification of new potential targets and approaches to treatment. Blockade of interleukin (IL)-6 (tocilizumab) and IL-1 (gevokizumab) are being evaluated in phase III clinical trials. It is hoped that improved risk stratification of organ damage and relapses will be developed using imaging and biomarkers, allowing for individualised treatment for patients; however, there remains further work to be done before this becomes a reality.
Collapse
Affiliation(s)
- Lauren Steel
- Rheumatology, Southend University Hospital, Prittlewell Chase, Westcliff-on-sea, Essex, SS0 0RY, UK
| | | | | |
Collapse
|
128
|
Kastbom A, Ärlestig L, Rantapää-Dahlqvist S. Genetic Variants of the NLRP3 Inflammasome Are Associated with Stroke in Patients with Rheumatoid Arthritis. J Rheumatol 2015; 42:1740-5. [PMID: 26178285 DOI: 10.3899/jrheum.141529] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/03/2015] [Indexed: 12/31/2022]
Abstract
OBJECTIVE Inflammasomes are intracellular protein complexes important for the production of pro-inflammatory cytokines. Studies have suggested that the NLRP3 inflammasome influences both the severity of rheumatoid arthritis (RA) and development of atherosclerosis. Therefore, we investigated whether functional genetic variants related to the NLRP3 inflammasome influence the risk of cardiovascular (CV) disease (CVD) in patients with RA. METHODS The incidence of CVD was assessed in 522 patients with established RA by a retrospective survey of medical records in combination with a 6-year prospective followup. NLRP3-Q705K and CARD8-C10X genotypes were analyzed in relation to CVD by logistic regression, adjusting for traditional risk factors, antirheumatic treatment, and age at the onset of RA. RESULTS Carriage of the NLRP3-Q705K minor allele was associated with an increased risk of stroke/transient ischemic attack (TIA; OR 2.01, 95% CI 1.0-4.1, p = 0.05), while CARD8-C10X was not associated with any type of CV event. Patients with ≥ 1 variant allele in both polymorphisms had an increased risk of CVD when compared with patients without variant alleles present in both polymorphisms (adjusted OR 3.05, 95% CI 1.42-6.54, p = 0.004). Stratification showed that this risk was confined to stroke/TIA (adjusted OR 5.09, 95% CI 2.27-11.44, p < 0.0001) and not to myocardial infarction (MI)/angina pectoris (adjusted OR 1.58, 95% CI 0.67-3.73). Risk estimates were consistently higher among female patients. CONCLUSION Genetic variants of the NLRP3 inflammasome influence the risk of stroke/TIA, but not of MI/angina pectoris in Swedish patients with established RA.
Collapse
Affiliation(s)
- Alf Kastbom
- From the Department of Rheumatology, and Department of Clinical and Experimental Medicine, Linköping University, Linköping; the Department of Public Health and Clinical Medicine/Rheumatology, Umeå University, Umeå, Sweden.A. Kastbom, MD, PhD, Department of Rheumatology, and Department of Clinical and Experimental Medicine, Linköping University; L. Ärlestig, PhD; S. Rantapää-Dahlqvist, Department of Public Health and Clinical Medicine/Rheumatology, Umeå University.
| | - Lisbeth Ärlestig
- From the Department of Rheumatology, and Department of Clinical and Experimental Medicine, Linköping University, Linköping; the Department of Public Health and Clinical Medicine/Rheumatology, Umeå University, Umeå, Sweden.A. Kastbom, MD, PhD, Department of Rheumatology, and Department of Clinical and Experimental Medicine, Linköping University; L. Ärlestig, PhD; S. Rantapää-Dahlqvist, Department of Public Health and Clinical Medicine/Rheumatology, Umeå University
| | - Solbritt Rantapää-Dahlqvist
- From the Department of Rheumatology, and Department of Clinical and Experimental Medicine, Linköping University, Linköping; the Department of Public Health and Clinical Medicine/Rheumatology, Umeå University, Umeå, Sweden.A. Kastbom, MD, PhD, Department of Rheumatology, and Department of Clinical and Experimental Medicine, Linköping University; L. Ärlestig, PhD; S. Rantapää-Dahlqvist, Department of Public Health and Clinical Medicine/Rheumatology, Umeå University
| |
Collapse
|
129
|
Sharma A, Maul E. Vasculitis Causing Complete Occlusion of Aorta. J Pediatr 2015; 167:206.e1-2. [PMID: 25937429 DOI: 10.1016/j.jpeds.2015.04.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/19/2015] [Revised: 03/26/2015] [Accepted: 04/06/2015] [Indexed: 10/23/2022]
Affiliation(s)
- Akshay Sharma
- Department of Pediatrics, University of Kentucky College of Medicine, Lexington, Kentucky
| | - Erich Maul
- Department of Pediatrics, University of Kentucky College of Medicine, Lexington, Kentucky
| |
Collapse
|
130
|
Narula NG, Abril A. MDS and GCA: a prognostic dilemma. Clin Rheumatol 2015; 35:807-12. [PMID: 26080752 DOI: 10.1007/s10067-015-2993-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2015] [Revised: 06/03/2015] [Accepted: 06/07/2015] [Indexed: 12/25/2022]
Abstract
A number of autoimmune disorders, such as large vessel vasculitis, have been recognized in patients who have myelodysplastic syndrome (MDS) and myeloproliferative syndrome (MPS). The influence of an autoimmune disorder on the prognosis of patients who have MDS and MPS remains to be determined. We report three cases of myelodysplastic syndrome and large vessel vasculitis seen at our institution and provide a brief literature review of the two disease processes. More studies are needed to determine the pathophysiology of the underlying diseases and whether there truly is a relationship between the disease entities and to discover better markers for disease assessment.
Collapse
Affiliation(s)
- Neha G Narula
- Department of Rheumatology, Mayo Clinic, 4500 San Pablo Road, Jacksonville, FL, 32224, USA.
| | - Andy Abril
- Department of Rheumatology, Mayo Clinic, 4500 San Pablo Road, Jacksonville, FL, 32224, USA
| |
Collapse
|
131
|
Vorkapic E, Lundberg AM, Mäyränpää MI, Eriksson P, Wågsäter D. TRIF adaptor signaling is important in abdominal aortic aneurysm formation. Atherosclerosis 2015; 241:561-8. [PMID: 26100679 DOI: 10.1016/j.atherosclerosis.2015.06.014] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/10/2014] [Revised: 04/27/2015] [Accepted: 06/09/2015] [Indexed: 12/21/2022]
Abstract
OBJECTIVE Abdominal aortic aneurysm (AAA) is characterized by inflammation, loss of smooth muscle cells (SMCs), and degradation of the extracellular matrix in the vessel wall. Innate immune receptors such as Toll-like receptors (TLRs) were recently shown to regulate immunological processes leading to the formation and progression of atherosclerotic plaques as well as to other cardiovascular pathologies. Our aim was to investigate whether blockage of TLR signaling, under the control of TIR domain-containing adaptor protein including IFN-β (TRIF), could inhibit the inflammatory response and AAA development in mice. RESULTS In human AAA, an increased TLR3 and TLR4 expression in association with macrophages and T lymphocytes was demonstrated with immunohistochemical analysis. Angiotensin (Ang) II-induced aneurysm formation was significantly reduced by 30% in ApoE(-/-)Trif(-/-) mice compared to ApoE(-/-) mice. Morphologically, AngII-infused ApoE(-/-)Trif(-/-) mice had a more intact cellular and extracellular matrix while ApoE(-/-) mice infused with AngII displayed an increased medial thickness associated with aortic dissection, thrombus formation, and a more disorganized vessel wall. Gene expression analysis of the abdominal aorta revealed a profound decrease of the inflammatory genes CD68 (P < 0.05), CD11b (P < 0.05), and TNF-α (P < 0.05) and the protease gene MMP-12 (P < 0.01) in ApoE(-/-)Trif(-/-) mice compared to ApoE(-/-) mice infused with AngII. CONCLUSION Our results suggest that signaling through TRIF is important for the inflammatory response of AngII-induced AAA and that blockage of the TRIF pathway reduces vascular inflammation and protects against AAA formation.
Collapse
Affiliation(s)
- Emina Vorkapic
- Division of Drug Research, Department of Medical and Health Sciences, Faculty of Health Sciences, Linköping University, Linköping, Sweden; Atherosclerosis Research Unit, Center for Molecular Medicine, Department of Medicine, Karolinska Institute, Stockholm, Sweden.
| | - Anna M Lundberg
- Center for Molecular Medicine, Department of Medicine, Karolinska University Hospital Solna, Karolinska Institute, Stockholm, Sweden
| | - Mikko I Mäyränpää
- Department of Pathology, Haartman Institute, University of Helsinki, Helsinki, Finland; HUSLAB, Division of Pathology, Meilahti Laboratories of Pathology, Helsinki University Central Hospital, Helsinki Finland
| | - Per Eriksson
- Atherosclerosis Research Unit, Center for Molecular Medicine, Department of Medicine, Karolinska Institute, Stockholm, Sweden
| | - Dick Wågsäter
- Division of Drug Research, Department of Medical and Health Sciences, Faculty of Health Sciences, Linköping University, Linköping, Sweden; Atherosclerosis Research Unit, Center for Molecular Medicine, Department of Medicine, Karolinska Institute, Stockholm, Sweden
| |
Collapse
|
132
|
Coit P, De Lott LB, Nan B, Elner VM, Sawalha AH. DNA methylation analysis of the temporal artery microenvironment in giant cell arteritis. Ann Rheum Dis 2015; 75:1196-202. [PMID: 26038090 DOI: 10.1136/annrheumdis-2014-207116] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2014] [Accepted: 05/02/2015] [Indexed: 01/10/2023]
Abstract
OBJECTIVE To investigate the inflammatory response in giant cell arteritis (GCA) by characterising the DNA methylation pattern within the temporal artery microenvironment. METHODS Twelve patients with non-equivocal histological evidence for GCA and 12 age-matched, sex-matched and ethnicity-matched controls with normal biopsies were studied. DNA was extracted from the affected portions of temporal artery tissue in patients with GCA and from histologically confirmed normal arteries in controls. Genome-wide DNA methylation status was evaluated using the Illumina Infinium HumanMethylation450 BeadChip Array. Differentially methylated loci between affected and unaffected arterial tissues were identified, and subsequent bioinformatic analysis performed. Immunohistochemistry was used to examine tissue expression patterns in temporal artery biopsies. RESULTS We identified 1555 hypomethylated CG sites (853 genes) in affected temporal artery tissue from patients with GCA compared with normal controls. Gene ontology enrichment analysis of hypomethylated genes revealed significant representation in T cell activation and differentiation pathways, including both TH1 and TH17 signatures. Our DNA methylation data suggest a role for increased activity of the calcineurin/nuclear factor of activated T cells (NFAT) signalling pathway in GCA, confirmed by immunohistochemistry showing increased expression and nuclear localisation of NFAT1. NFAT signalling downstream targets such as interleukin (IL)-21/IL-21R and CD40L were overexpressed in GCA-affected arteries. Further, proinflammatory genes including TNF, LTA, LTB, CCR7, RUNX3, CD6, CD40LG, IL2, IL6, NLRP1, IL1B, IL18, IL21, IL23R and IFNG were hypomethylated in the cellular milieu of GCA arteries. CONCLUSIONS We characterised the inflammatory response in GCA-affected arteries using 'epigenetic immunophenotyping' and identified molecules and pathways relevant to disease pathogenesis in GCA.
Collapse
Affiliation(s)
- Patrick Coit
- Division of Rheumatology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Lindsey B De Lott
- Department of Ophthalmology and Visual Sciences, University of Michigan, Ann Arbor, Michigan, USA
| | - Bin Nan
- Department of Biostatistics, School of Public Health, University of Michigan, Ann Arbor, Michigan, USA
| | - Victor M Elner
- Department of Ophthalmology and Visual Sciences, University of Michigan, Ann Arbor, Michigan, USA Department of Pathology, University of Michigan, Ann Arbor, Michigan, USA
| | - Amr H Sawalha
- Division of Rheumatology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA Center for Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, Michigan, USA
| |
Collapse
|
133
|
Kermani TA, Crowson CS, Muratore F, Schmidt J, Matteson EL, Warrington KJ. Extra-cranial giant cell arteritis and Takayasu arteritis: How similar are they? Semin Arthritis Rheum 2015; 44:724-8. [DOI: 10.1016/j.semarthrit.2015.01.005] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2014] [Revised: 01/04/2015] [Accepted: 01/16/2015] [Indexed: 10/24/2022]
|
134
|
Lindeman JHN. The pathophysiologic basis of abdominal aortic aneurysm progression: a critical appraisal. Expert Rev Cardiovasc Ther 2015; 13:839-51. [PMID: 26028299 DOI: 10.1586/14779072.2015.1052408] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
An aneurysm of the abdominal aorta is a common pathology and a major cause of sudden death in the elderly. Currently, abdominal aortic aneurysms (AAAs) can only be treated by surgery and an effective medical therapy is urgently missing. The pathophysiology of AAAs is complex and is believed to be best described as a comprehensive inflammatory response with an accompanying proteolytic imbalance; the latter being held responsible for the progressive weakening of the aortic wall. Remarkably, while interference in inflammatory and/or proteolytic cascades proves highly effective in preclinical studies, emerging clinical studies consistently fail to show a benefit. In fact, some anti-inflammatory interventions appear to adversely influence the disease process. Altogether, recent clinical observations not only challenge the prevailing concepts of AAA progression, but also raise doubt on the translatability of findings from rodent models for growing AAA.
Collapse
Affiliation(s)
- Jan H N Lindeman
- Department Vascular and Transplant Surgery, Leiden University Medical Center, PO Box 9600, 2300 RC Leiden, The Netherlands
| |
Collapse
|
135
|
Schneider S, Koch W, Hoppmann P, Ubrich R, Kemmner S, Steinlechner E, Heemann U, Laugwitz KL, Kastrati A, Baumann M. Association of Toll-like receptor 4 polymorphism with age-dependent systolic blood pressure increase in patients with coronary artery disease. IMMUNITY & AGEING 2015; 12:4. [PMID: 26015800 PMCID: PMC4443624 DOI: 10.1186/s12979-015-0031-2] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 04/01/2015] [Accepted: 05/07/2015] [Indexed: 12/15/2022]
Abstract
Background Systolic blood pressure (SBP) increases steadily with age and bears an independent continuous relationship with the incidence of cardiovascular events. Low-grade inflammation is a suspected pathomechanism causing vascular aging and promote coronary artery disease (CAD). Recent animal studies give evidence that Toll-like receptor 4 (TLR4) modulate inflammation and contribute to age-dependent SBP increase. However, there are no data about TLR4 and age-dependent blood pressure increase in human. Methods and results We therefor investigate a human cohort of 2679 patients with CAD aged between 50–80 years. Genotypes were determined for the TLR4 single nucleotide polymorphism rs4986790 (TLR4 896A/G). Patients were stratified according to tertiles of age and the upper tertile was compared to lower tertiles. In this cohort we show that older patients with the TLR4 896 G allele had significantly lower SBP (TLR4 G allele carriers: 148.2 ± 30.4 mmHg versus A/A allele carrier: 154.9 ± 27.2 mmHg; P < 0.05) and lower pulse pressure (TLR4 G allele carriers: 69.1 ± 29.7 mmHg versus A/A allele carrier: 75.5 ± 26.4 mmHg; P < 0.05) as compared to TLR4 896A/A allele carrier. Conclusion We demonstrate an association between the TLR4 SNP rs4986790 genotype and age-dependant blood pressure increase in patients with coronary artery disease. Electronic supplementary material The online version of this article (doi:10.1186/s12979-015-0031-2) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Simon Schneider
- Medizinische Klinik und Poliklinik, Klinikum rechts der Isar, Technische Universität München, Ismaninger Str.22, 81675 Munich, Germany
| | - Werner Koch
- Deutsches Herzzentrum München, Technische Universität München, Lazarettstr. 36, 80636 Munich, Germany
| | - Petra Hoppmann
- Medizinische Klinik und Poliklinik, Klinikum rechts der Isar, Technische Universität München, Ismaninger Str.22, 81675 Munich, Germany
| | - Romy Ubrich
- Medizinische Klinik und Poliklinik, Klinikum rechts der Isar, Technische Universität München, Ismaninger Str.22, 81675 Munich, Germany
| | - Stephan Kemmner
- Department of Nephrology, Klinikum rechts der Isar, Technische Universität München, Ismaninger Str.22, 81675 Munich, Germany
| | - Eva Steinlechner
- Medizinische Klinik und Poliklinik, Klinikum rechts der Isar, Technische Universität München, Ismaninger Str.22, 81675 Munich, Germany
| | - Uwe Heemann
- Department of Nephrology, Klinikum rechts der Isar, Technische Universität München, Ismaninger Str.22, 81675 Munich, Germany
| | - Karl-Ludwig Laugwitz
- Medizinische Klinik und Poliklinik, Klinikum rechts der Isar, Technische Universität München, Ismaninger Str.22, 81675 Munich, Germany.,German Centre for Cardiovascular Research (DZHK), partner site Munich Heart Alliance, Oudenarder Straße 16, 13347 Munich, Germany
| | - Adnan Kastrati
- Deutsches Herzzentrum München, Technische Universität München, Lazarettstr. 36, 80636 Munich, Germany.,German Centre for Cardiovascular Research (DZHK), partner site Munich Heart Alliance, Oudenarder Straße 16, 13347 Munich, Germany
| | - Marcus Baumann
- Department of Nephrology, Klinikum rechts der Isar, Technische Universität München, Ismaninger Str.22, 81675 Munich, Germany
| |
Collapse
|
136
|
Yan H, Cui B, Zhang X, Fu X, Yan J, Wang X, Lv X, Chen Z, Hu Z. Antagonism of toll-like receptor 2 attenuates the formation and progression of abdominal aortic aneurysm. Acta Pharm Sin B 2015; 5:176-87. [PMID: 26579444 PMCID: PMC4629243 DOI: 10.1016/j.apsb.2015.03.007] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2015] [Accepted: 03/12/2015] [Indexed: 12/17/2022] Open
Abstract
Abdominal aortic aneurysm (AAA) is an inflammatory vascular disorder with high mortality. Accumulating evidence shows that toll-like receptor 2 (TLR2) plays a critical role in the regulation of wound-repairing process after tissue injury. We wondered if TLR2 signaling contributed to the pathogenesis of AAA and that targeting TLR2 would attenuate AAA development and progression. In this study, enhanced expression of TLR2 and its ligands were observed in human AAA tissue. Neutralization of TLR2 protected against AAA development and caused established AAA to regress in mouse models of AAA. In addition, TLR2-deficient mice also failed to develop AAA. The prophylactic and therapeutic effects of blocking TLR2 were accompanied by a significant resolution of inflammation and vascular remodeling, as indicated by the decreased expression or activity of MMP-2/9, α-SMA, inflammatory cytokines, and transcription factors NF-κB, AP-1 and STAT1/3 in AAA tissue. Mechanistically, blocking TLR2 decreased the expression and interaction of TLR2 and several endogenous ligands, which diminished chronic inflammation and vascular remodeling in the vascular tissue of AAA. Our studies indicate that the interactions between TLR2 and its endogenous ligands contribute to the pathogenesis of AAA and that targeting TLR2 offers great potential toward the development of therapeutic agents against AAA.
Collapse
Key Words
- AAA, abdominal aortic aneurysm
- AP-1, activator protein-1
- Abdominal aortic aneurysm
- Ang II, angiotensin II
- DAMP, damage associated molecular pattern
- DAMPs
- DHE, dihydroethidium
- HMGB1, high mobility group B-1
- HSP, heat shock protein
- IOD, integrated optical density
- Immune microenvironment
- MCP-1, monocyte chemoattractant protein-1
- MMP, matrix metalloproteinase
- NF-κB, nuclear factor kappa B
- PAMP, pathogen-associated molecular pattern
- PRRs, pattern recognition receptors
- RAMPs, resolution-associated molecular patterns
- ROS, reactive oxygen species
- STAT1/3, signal transducer and activator of transcription 1/3
- TLR, toll-like receptor
- TLR2
- Th2, type 2 T help
- VVG, Verhoeff van Gieson
- Vascular remodeling
- WT, wide-type
- bip, binding immunoglobulin protein
- α-SMA, α-smooth muscle actin
Collapse
|
137
|
Tang YL, Jiang JH, Wang S, Liu Z, Tang XQ, Peng J, Yang YZ, Gu HF. TLR4/NF-κB signaling contributes to chronic unpredictable mild stress-induced atherosclerosis in ApoE-/- mice. PLoS One 2015; 10:e0123685. [PMID: 25860573 PMCID: PMC4393302 DOI: 10.1371/journal.pone.0123685] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2014] [Accepted: 02/21/2015] [Indexed: 11/18/2022] Open
Abstract
Objective Chronic stress is an important risk factor for atherosclerotic diseases. Our previous studies have shown that chronic unpredictable mild stress (CUMS) accelerates atherosclerosis and up-regulates TLR4/NF-κB expression in apoE-/- mice. However, TLR4/NF-κB signaling whether directly contributes to the development of atherosclerosis in CUMS mice is unclear. We hypothesized that the interference of TLR4/NF-κB can ameliorate CUMS-induced inflammation and atherosclerosis in apoE-/- mice. Methods ApoE-/- mice were exposed to 12 weeks CUMS. Ad-siRNA TLR4 was given by tail vein injection (10 μl/mouse, every 5 days), and PDTC (an inhibitor of NF-κB) was given by intraperitoneal injection (60 mg/kg, once a day). Plasma corticosterone concentrations were determined by solid-phase 125I radioimmunoassay. Atherosclerosis lesions in aortic sinuses were evaluated and quantified by IMAGEPRO PLUS. Western blotting was used to detect the expression of TLR4, NF-κB, and IL-1β in aortas of the mice. Plasma lipid profiles, IL-1β, TNF-α, and MCP-1 were measured by ELISA. Results Our results indicated that CUMS apoE-/- mice treatment with siRNA TLR4 significantly decreased atherosclerosis and down-regulated TLR4, NF-κB, and inflammatory cytokines. PDTC also remarkably reduced atherosclerosis and the levels of IL-1β, TNF-α and MCP-1 in plasma. However, Treatment with siRNA TLR4 or PDTC had no effect on plasma corticosterone levels, and lipid profiles. Conclusions TLR4/NF-κB pathway may participate in CUMS-induced atherosclerosis through activation of proinflammatory cytokines in apoE-/- mice. Our data may provide a new potential therapeutic target for prevention of CUMS -induced atherosclerosis.
Collapse
Affiliation(s)
- Ya Ling Tang
- Key Lab for Arteriosclerology of Hunan Province, Institute of Cardiovascular Disease, University of South China, Hengyang, 421001, People's Republic of China
| | - Jian Hong Jiang
- Chuanshan College, University of South China, Hengyang, 421001, People’s Republic of China
| | - Shuang Wang
- Key Lab for Arteriosclerology of Hunan Province, Institute of Cardiovascular Disease, University of South China, Hengyang, 421001, People's Republic of China
| | - Zhu Liu
- Chuanshan College, University of South China, Hengyang, 421001, People’s Republic of China
| | - Xiao Qing Tang
- Department of Physiology & Institute of Neuroscience, University of South China, Hengyang, 421001, People’s Republic of China
| | - Juan Peng
- Key Lab for Arteriosclerology of Hunan Province, Institute of Cardiovascular Disease, University of South China, Hengyang, 421001, People's Republic of China
| | - Yong-Zong Yang
- Key Lab for Arteriosclerology of Hunan Province, Institute of Cardiovascular Disease, University of South China, Hengyang, 421001, People's Republic of China
| | - Hong-Feng Gu
- Key Lab for Arteriosclerology of Hunan Province, Institute of Cardiovascular Disease, University of South China, Hengyang, 421001, People's Republic of China
- Department of Physiology & Institute of Neuroscience, University of South China, Hengyang, 421001, People’s Republic of China
- * E-mail:
| |
Collapse
|
138
|
Shirai T, Hilhorst M, Harrison DG, Goronzy JJ, Weyand CM. Macrophages in vascular inflammation--From atherosclerosis to vasculitis. Autoimmunity 2015; 48:139-51. [PMID: 25811915 DOI: 10.3109/08916934.2015.1027815] [Citation(s) in RCA: 103] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The spectrum of vascular inflammatory disease ranges from atherosclerosis and hypertension, widespread conditions affecting large proportions of the population, to the vasculitides, rare syndromes leading to fast and irreversible organ failure. Atherosclerosis progresses over decades, inevitably proceeding through multiple phases of disease and causes its major complications when the vessel wall lesion ruptures, giving rise to lumen-occlusive atherothrombosis. Vasculitides of medium and large arteries progress rapidly, causing tissue ischemia through lumen-occlusive intimal hyperplasia. In both disease entities, macrophages play a decisive role in pathogenesis, but function in the context of other immune cells that direct their differentiation and their functional commitments. In atherosclerosis, macrophages are involved in the removal of lipids and tissue debris and make a critical contribution to tissue damage and wall remodeling. In several of the vasculitides, macrophages contribute to granuloma formation, a microstructural platform optimizing macrophage-T-cell interactions, antigen containment and inflammatory amplification. By virtue of their versatility and plasticity, macrophages are able to promote a series of pathogenic functions, ranging from the release of cytokines and enzymes, the production of reactive oxygen species, presentation of antigen and secretion of tissue remodeling factors. However, as short-lived cells that lack memory, macrophages are also amendable to reprogramming, making them promising targets for anti-inflammatory interventions.
Collapse
Affiliation(s)
- Tsuyoshi Shirai
- Division of Immunology and Rheumatology, Department of Medicine, Stanford University School of Medicine , Stanford, CA , USA and
| | | | | | | | | |
Collapse
|
139
|
Abstract
Inflammatory arterial diseases differentially affect the compartments of the vessel wall. The intima and adventitia are commonly involved by the disease process, with luminal and microvascular endothelial cells playing a critical role in the recruitment and activation of leukocytes. In contrast, the avascular media is often spared by immune-mediated disorders. Surprisingly, vascular smooth muscle cells (VSMCs), the predominant and often exclusive cell type of the media, are capable of robust proinflammatory responses to diverse stressors. The multiple cytokines and chemokines produced within the media can profoundly affect macrophage and T cell function, thus amplifying and shaping innate and adaptive immune responses. On the other hand, VSMCs and the extracellular matrix that they produce also display significant anti-inflammatory properties. The balance between the pro- and anti-inflammatory effects of VSMCs and their extracellular matrix versus the strength of the inciting immunologic events determines the pattern of medial pathology. Limitations on the extent of medial infiltration and injury, defined as medial immunoprivilege, are typically seen in arteriosclerotic diseases, such as atherosclerosis and transplant vasculopathy. Conversely, breakdown of medial immunoprivilege that manifests as more intense leukocytic infiltrates, loss of VSMCs, and destruction of the extracellular matrix architecture is a general feature of certain aneurysmal diseases and vasculitides. In this review, we consider the inflammatory and immune functions of VSMCs and how they may lead to medial immunoprivilege or medial inflammation in arterial diseases.
Collapse
Affiliation(s)
- George Tellides
- From the Departments of Surgery (G.T.) and Immunobiology (J.S.P.), Yale University School of Medicine, New Haven, CT; and Veterans Affairs Connecticut Healthcare System, West Haven, CT (G.T.).
| | - Jordan S Pober
- From the Departments of Surgery (G.T.) and Immunobiology (J.S.P.), Yale University School of Medicine, New Haven, CT; and Veterans Affairs Connecticut Healthcare System, West Haven, CT (G.T.)
| |
Collapse
|
140
|
Abstract
All aspects of the pathogenesis of atherosclerosis are critically influenced by the inflammatory response in vascular plaques. Research in the field of innate immunity from the past 2 decades has uncovered many novel mechanisms elucidating how immune cells sense microbes, tissue damage, and metabolic derangements. Here, we summarize which triggers of innate immunity appear during atherogenesis and by which pathways they can contribute to inflammation in atherosclerotic plaques. The increased understanding gained from studies assessing how immune activation is associated with the pathogenesis of atherosclerosis has provided many novel targets for potential therapeutic intervention. Excitingly, the concept that inflammation may be the core of cardiovascular disease is currently being clinically evaluated and will probably encourage further studies in this area.
Collapse
Affiliation(s)
- Sebastian Zimmer
- From the Medizinische Klinik und Poliklinik II (S.Z.) and Institute of Innate Immunity (A.G., E.L.), University Hospitals Bonn, Bonn, Germany; Department of Infectious Diseases and Immunology, University of Massachusetts Medical School, Worcester (E.L.); and German Center of Neurodegenerative Diseases (DZNE), Bonn, Germany (E.L.)
| | - Alena Grebe
- From the Medizinische Klinik und Poliklinik II (S.Z.) and Institute of Innate Immunity (A.G., E.L.), University Hospitals Bonn, Bonn, Germany; Department of Infectious Diseases and Immunology, University of Massachusetts Medical School, Worcester (E.L.); and German Center of Neurodegenerative Diseases (DZNE), Bonn, Germany (E.L.)
| | - Eicke Latz
- From the Medizinische Klinik und Poliklinik II (S.Z.) and Institute of Innate Immunity (A.G., E.L.), University Hospitals Bonn, Bonn, Germany; Department of Infectious Diseases and Immunology, University of Massachusetts Medical School, Worcester (E.L.); and German Center of Neurodegenerative Diseases (DZNE), Bonn, Germany (E.L.).
| |
Collapse
|
141
|
Anders HJ, Baumann M, Tripepi G, Mallamaci F. Immunity in arterial hypertension: associations or causalities? Nephrol Dial Transplant 2015; 30:1959-64. [PMID: 25762356 DOI: 10.1093/ndt/gfv057] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2015] [Accepted: 02/05/2015] [Indexed: 12/20/2022] Open
Abstract
Numerous studies describe associations between markers of inflammation and arterial hypertension (aHT), but does that imply causality? Interventional studies that reduce blood pressure reduced also markers of inflammation, but does immunosuppression improve hypertension? Here, we review the available mechanistic data. Aberrant immunity can trigger endothelial dysfunction but is hardly ever the primary cause of aHT. Innate and adaptive immunity get involved once hypertension has caused vascular wall injury as immunity is a modifier of endothelial dysfunction and vascular wall remodelling. As vascular remodelling progresses, immunity-related mechanisms can become significant cofactors for cardiovascular (CV) disease progression; vice versa, suppressing immunity can improve hypertension and CV outcomes. Innate and adaptive immunity both contribute to vascular wall remodelling. Innate immunity is driven by danger signals that activate Toll-like receptors and other pattern-recognition receptors. Adaptive immunity is based on loss of tolerance against vascular autoantigens and includes autoreactive T-cell immunity as well as non-HLA angiotensin II type 1 receptor-activating autoantibodies. Such processes involve numerous other modulators such as regulatory T cells. Together, immunity is not causal for hypertension but rather an important secondary pathomechanism and a potential therapeutic target in hypertension.
Collapse
Affiliation(s)
- Hans-Joachim Anders
- Nephrologisches Zentrum, Medizinische Klinik und Poliklinik IV, Klinikum der Universität München, Munich, Germany
| | - Marcus Baumann
- Department of Nephrology, Klinikum rechts der Isar der Technischen Universität München, Munich, Germany
| | - Giovanni Tripepi
- Nephrology, Dialysis and Transplantation Unit & CNR-IFC, Clinical Epidemiology and Physiopathology of Renal Disease and Hypertension of Reggio Calabria, Reggio Calabria, Italy
| | - Francesca Mallamaci
- Nephrology, Dialysis and Transplantation Unit & CNR-IFC, Clinical Epidemiology and Physiopathology of Renal Disease and Hypertension of Reggio Calabria, Reggio Calabria, Italy
| |
Collapse
|
142
|
Kölliker Frers R, Bisoendial R, Montoya S, Kerzkerg E, Castilla R, Tak P, Milei J, Capani F. Psoriasis and cardiovascular risk: Immune-mediated crosstalk between metabolic, vascular and autoimmune inflammation. ACTA ACUST UNITED AC 2015. [DOI: 10.1016/j.ijcme.2015.01.005] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
143
|
Chen H, Zheng D, Ambadapadi S, Davids J, Ryden S, Samy H, Bartee M, Sobel E, Dai E, Liu L, Macaulay C, Yachnis A, Weyand C, Thoburn R, Lucas A. Serpin treatment suppresses inflammatory vascular lesions in temporal artery implants (TAI) from patients with giant cell arteritis. PLoS One 2015; 10:e0115482. [PMID: 25658487 PMCID: PMC4319900 DOI: 10.1371/journal.pone.0115482] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2014] [Accepted: 11/24/2014] [Indexed: 01/08/2023] Open
Abstract
Giant cell arteritis (GCA) and Takayasu’s disease are inflammatory vasculitic syndromes (IVS) causing sudden blindness and widespread arterial obstruction and aneurysm formation. Glucocorticoids and aspirin are mainstays of treatment, predominantly targeting T cells. Serp-1, a Myxomavirus-derived serpin, blocks macrophage and T cells in a wide range of animal models. Serp-1 also reduced markers of myocardial injury in a Phase IIa clinical trial for unstable coronary disease. In recent work, we detected improved survival and decreased arterial inflammation in a mouse Herpesvirus model of IVS. Here we examine Serp-1 treatment of human temporal artery (TA) biopsies from patients with suspected TA GCA arteritis after implant (TAI) into the aorta of immunodeficient SCID (severe combined immunodeficiency) mice. TAI positive for arteritis (GCApos) had significantly increased inflammation and plaque when compared to negative TAI (GCAneg). Serp-1 significantly reduced intimal inflammation and CD11b+ cell infiltrates in TAI, with reduced splenocyte Th1, Th17, and Treg. Splenocytes from mice with GCApos grafts had increased gene expression for interleukin-1beta (IL-1β), IL-17, and CD25 and decreased Factor II. Serp-1 decreased IL-1β expression. In conclusion, GCApos TAI xenografts in mice provide a viable disease model and have increased intimal inflammation as expected and Serp-1 significantly reduces vascular inflammatory lesions with reduced IL-1β.
Collapse
Affiliation(s)
- Hao Chen
- Department of Medicine, University of Florida, Gainesville, Florida, United States of America
- Department of Molecular Genetics & Microbiology, University of Florida, Gainesville, Florida, United States of America
| | - Donghang Zheng
- Department of Medicine, University of Florida, Gainesville, Florida, United States of America
- Department of Molecular Genetics & Microbiology, University of Florida, Gainesville, Florida, United States of America
| | - Sriram Ambadapadi
- Department of Medicine, University of Florida, Gainesville, Florida, United States of America
| | - Jennifer Davids
- Department of Medicine, University of Florida, Gainesville, Florida, United States of America
| | - Sally Ryden
- Department of Medicine, University of Florida, Gainesville, Florida, United States of America
| | - Hazem Samy
- Department of Ophthalmology, University of Florida, Gainesville, Florida, United States of America
| | - Mee Bartee
- Department of Medicine, University of Florida, Gainesville, Florida, United States of America
| | - Eric Sobel
- Department of Medicine, University of Florida, Gainesville, Florida, United States of America
| | - Erbin Dai
- Department of Medicine, University of Florida, Gainesville, Florida, United States of America
| | - Liying Liu
- Department of Medicine, University of Florida, Gainesville, Florida, United States of America
| | | | - Anthony Yachnis
- Department of Pathology, University of Florida, Gainesville, Florida, United States of America
| | - Cornelia Weyand
- Division of Immunology and Rheumatology, Stanford University School of Medicine, Stanford, California, United States of America
| | - Robert Thoburn
- Department of Medicine, University of Florida, Gainesville, Florida, United States of America
| | - Alexandra Lucas
- Department of Medicine, University of Florida, Gainesville, Florida, United States of America
- Department of Ophthalmology, University of Florida, Gainesville, Florida, United States of America
- Viron Therapeutics, London, Ontario, Canada
- * E-mail:
| |
Collapse
|
144
|
Ramirez GA, Rovere-Querini P, Sabbadini MG, Manfredi AA. Parietal and intravascular innate mechanisms of vascular inflammation. Arthritis Res Ther 2015; 17:16. [PMID: 25889488 PMCID: PMC4308901 DOI: 10.1186/s13075-015-0528-2] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Sustained inflammation of the vessel walls occurs in a large number of systemic diseases (ranging from atherosclerosis to systemic vasculitides, thrombotic microangiopathies and connective tissue diseases), which are ultimately characterized by ischemia and end-organ failure. Cellular and humoral innate immunity contribute to a common pathogenic background and comprise several potential targets for therapeutic intervention. Here we discuss some recent advances in the effector and regulatory action of neutrophils and in the outcome of their interaction with circulating platelets. In parallel, we discuss novel insights into the role of humoral innate immunity in vascular inflammation. All these topics are discussed in light of potential clinical and therapeutic implications in the near future.
Collapse
Affiliation(s)
- Giuseppe A Ramirez
- IRCCS Ospedale San Raffaele, via Olgettina 60, 20132, Milan, Italy. .,Università Vita Salute San Raffaele, via Olgettina 58, 20132, Milan, Italy.
| | - Patrizia Rovere-Querini
- IRCCS Ospedale San Raffaele, via Olgettina 60, 20132, Milan, Italy. .,Università Vita Salute San Raffaele, via Olgettina 58, 20132, Milan, Italy.
| | - Maria Grazia Sabbadini
- IRCCS Ospedale San Raffaele, via Olgettina 60, 20132, Milan, Italy. .,Università Vita Salute San Raffaele, via Olgettina 58, 20132, Milan, Italy.
| | - Angelo A Manfredi
- IRCCS Ospedale San Raffaele, via Olgettina 60, 20132, Milan, Italy. .,Università Vita Salute San Raffaele, via Olgettina 58, 20132, Milan, Italy.
| |
Collapse
|
145
|
von Rossum A, Laher I, Choy JC. Immune-mediated vascular injury and dysfunction in transplant arteriosclerosis. Front Immunol 2015; 5:684. [PMID: 25628623 PMCID: PMC4290675 DOI: 10.3389/fimmu.2014.00684] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2014] [Accepted: 12/18/2014] [Indexed: 12/16/2022] Open
Abstract
Solid organ transplantation is the only treatment for end-stage organ failure but this life-saving procedure is limited by immune-mediated rejection of most grafts. Blood vessels within transplanted organs are targeted by the immune system and the resultant vascular damage is a main contributor to acute and chronic graft failure. The vasculature is a unique tissue with specific immunological properties. This review discusses the interactions of the immune system with blood vessels in transplanted organs and how these interactions lead to the development of transplant arteriosclerosis, a leading cause of heart transplant failure.
Collapse
Affiliation(s)
- Anna von Rossum
- Department of Molecular Biology and Biochemistry, Simon Fraser University , Burnaby, BC , Canada
| | - Ismail Laher
- Department of Anaesthesiology, Pharmacology and Therapeutics, University of British Columbia , Vancouver, BC , Canada
| | - Jonathan C Choy
- Department of Molecular Biology and Biochemistry, Simon Fraser University , Burnaby, BC , Canada
| |
Collapse
|
146
|
|
147
|
Balistreri CR, Pisano C, Martorana A, Triolo OF, Lio D, Candore G, Ruvolo G. Are the leukocyte telomere length attrition and telomerase activity alteration potential predictor biomarkers for sporadic TAA in aged individuals? AGE (DORDRECHT, NETHERLANDS) 2014; 36:9700. [PMID: 25129574 PMCID: PMC4453932 DOI: 10.1007/s11357-014-9700-x] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/19/2014] [Accepted: 07/30/2014] [Indexed: 06/03/2023]
Abstract
A large variability in occurrence, complications, and age/gender manifestations characterizes individual susceptibility of sporadic thoracic aortic aneurysms (TAA), even in subjects with the same risk factor profiles. The reasons are poorly understood. On the other hand, TAA pathophysiology mechanisms remain unclear than those involved in abdominal aorta aneurysms. However, recent evidence is suggesting a crucial role of biological ageing in inter-individual risk variation of cardiovascular diseases, including sporadic TAA. Biological age rather than chronological age is a better predictor of vascular risk. Relevant assumptions support this concept. In confirming this evidence and our preliminary data, the mean of blood leukocyte telomere length, through use of terminal restriction fragment assay and in blood samples from sporadic TAA patients and controls, was examined. Telomerase activity was also analyzed in two groups. In addition, we verified the weight of genetic inflammatory variants and the major TAA risk factors in telomere/telomerase impairment. Aorta histopathological abnormalities and systemic inflammatory mediators were ultimately correlated with telomere/telomerase impairment. Data obtained demonstrated shorter telomeres and a reduced telomerase activity in TAA patients significantly associated with a genetic inflammatory risk profile, age, gender, smoking, hypertension, a histopathological phenotype, and higher levels of systemic inflammatory mediators than controls. In conclusion, telomere and telomerase activity's detection might be used as predictor biomarkers of sporadic TAA. Their impairment also suggests a strong role of vascular ageing in sporadic TAA, evocated by both environmental and genetic inflammatory factors.
Collapse
Affiliation(s)
- Carmela R Balistreri
- Department of Pathobiology and Medical and Forensic Biotechnologies, University of Palermo, Corso Tukory 211, 90134, Palermo, Italy,
| | | | | | | | | | | | | |
Collapse
|
148
|
Tan Y, Tseng PO, Wang D, Zhang H, Hunter K, Hertzberg J, Stenmark KR, Tan W. Stiffening-induced high pulsatility flow activates endothelial inflammation via a TLR2/NF-κB pathway. PLoS One 2014; 9:e102195. [PMID: 25029271 PMCID: PMC4100881 DOI: 10.1371/journal.pone.0102195] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2013] [Accepted: 06/16/2014] [Indexed: 12/25/2022] Open
Abstract
Stiffening of large arteries is increasingly used as an independent predictor of risk and therapeutic outcome for small artery dysfunction in many diseases including pulmonary hypertension. The molecular mechanisms mediating downstream vascular cell responses to large artery stiffening remain unclear. We hypothesize that high pulsatility flow, induced by large artery stiffening, causes inflammatory responses in downstream pulmonary artery endothelial cells (PAECs) through toll-like receptor (TLR) pathways. To recapitulate the stiffening effect of large pulmonary arteries that occurs in pulmonary hypertension, ultrathin silicone tubes of variable mechanical stiffness were formulated and were placed in a flow circulatory system. These tubes modulated the simulated cardiac output into pulsatile flows with different pulsatility indices, 0.5 (normal) or 1.5 (high). PAECs placed downstream of the tubes were evaluated for their expression of proinflammatory molecules (ICAM-1, VCAM-1, E-selectin and MCP-1), TLR receptors and intracellular NF-κB following flow exposure. Results showed that compared to flow with normal pulsatility, high pulsatility flow induced proinflammatory responses in PAECs, enhanced TLR2 expression but not TLR4, and caused NF-κB activation. Pharmacologic (OxPAPC) and siRNA inhibition of TLR2 attenuated high pulsatility flow-induced pro-inflammatory responses and NF-κB activation in PAECs. We also observed that PAECs isolated from small pulmonary arteries of hypertensive animals exhibiting proximal vascular stiffening demonstrated a durable ex-vivo proinflammatory phenotype (increased TLR2, TLR4 and MCP-1 expression). Intralobar PAECs isolated from vessels of IPAH patients also showed increased TLR2. In conclusion, this study demonstrates for the first time that TLR2/NF-κB signaling mediates endothelial inflammation under high pulsatility flow caused by upstream stiffening, but the role of TLR4 in flow pulsatility-mediated endothelial mechanotransduction remains unclear.
Collapse
Affiliation(s)
- Yan Tan
- Department of Mechanical Engineering, University of Colorado at Boulder, Boulder, Colorado, United States of America
- Department of Pediatrics, University of Colorado at Denver, Aurora, Colorado, United States of America
| | - Pi-Ou Tseng
- Department of Mechanical Engineering, University of Colorado at Boulder, Boulder, Colorado, United States of America
| | - Daren Wang
- Department of Pediatrics, University of Colorado at Denver, Aurora, Colorado, United States of America
| | - Hui Zhang
- Department of Pediatrics, University of Colorado at Denver, Aurora, Colorado, United States of America
| | - Kendall Hunter
- Department of Pediatrics, University of Colorado at Denver, Aurora, Colorado, United States of America
| | - Jean Hertzberg
- Department of Mechanical Engineering, University of Colorado at Boulder, Boulder, Colorado, United States of America
| | - Kurt R. Stenmark
- Department of Pediatrics, University of Colorado at Denver, Aurora, Colorado, United States of America
| | - Wei Tan
- Department of Mechanical Engineering, University of Colorado at Boulder, Boulder, Colorado, United States of America
| |
Collapse
|
149
|
Can the TLR-4-mediated signaling pathway be "a key inflammatory promoter for sporadic TAA"? Mediators Inflamm 2014; 2014:349476. [PMID: 25120286 PMCID: PMC4120489 DOI: 10.1155/2014/349476] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2013] [Accepted: 06/18/2014] [Indexed: 11/18/2022] Open
Abstract
Thoracic aorta shows with advancing age various changes and a progressive deterioration in structure and function. As a result, vascular remodeling (VR) and medial degeneration (MD) occur as pathological entities responsible principally for the sporadic TAA onset. Little is known about their genetic, molecular, and cellular mechanisms. Recent evidence is proposing the strong role of a chronic immune/inflammatory process in their evocation and progression. Thus, we evaluated the potential role of Toll like receptor- (TLR-) 4-mediated signaling pathway and its polymorphisms in sporadic TAA. Genetic, immunohistochemical, and biochemical analyses were assessed. Interestingly, the rs4986790 TLR4 polymorphism confers a higher susceptibility for sporadic TAA (OR = 14.4, P = 0.0008) and it represents, together with rs1799752 ACE, rs3918242 MMP-9, and rs2285053 MMP-2 SNPs, an independent sporadic TAA risk factor. In consistency with these data, a significant association was observed between their combined risk genotype and sporadic TAA. Cases bearing this risk genotype showed higher systemic inflammatory mediator levels, significant inflammatory/immune infiltrate, a typical MD phenotype, lower telomere length, and positive correlations with histopatological abnormalities, hypertension, smoking, and ageing. Thus, TLR4 pathway should seem to have a key role in sporadic TAA. It might represent a potential useful tool for preventing and monitoring sporadic TAA and developing personalized treatments.
Collapse
|
150
|
Abstract
A 79-year-old woman presents with new-onset pain in her neck and both shoulders. She takes 7.5 mg of prednisone per day for giant-cell arteritis. Occipital tenderness and diplopia developed 11 months before presentation. At that time, her erythrocyte sedimentation rate was elevated, at 78 mm per hour, and a temporal-artery biopsy revealed granulomatous arteritis. The diplopia resolved after 6 days of treatment with 60 mg of prednisone daily. Neither headache nor visual symptoms developed when the glucocorticoids were tapered. How should this patient’s care be managed?
Collapse
|