Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 126
Filter
1.
Int J Mol Sci ; 22(19)2021 Sep 30.
Article in English | MEDLINE | ID: mdl-34638942

ABSTRACT

Calcific aortic valve disease (CAVD) is an athero-inflammatory process. Growing evidence supports the inflammation-driven calcification model, mediated by cytokines such as interferons (IFNs) and tumor necrosis factor (TNF)-α. Our goal was investigating IFNs' effects in human aortic valve endothelial cells (VEC) and the potential differences between aortic (aVEC) and ventricular (vVEC) side cells. The endothelial phenotype was analyzed by Western blot, qPCR, ELISA, monocyte adhesion, and migration assays. In mixed VEC populations, IFNs promoted the activation of signal transducers and activators of transcription-1 and nuclear factor-κB, and the subsequent up-regulation of pro-inflammatory molecules. Side-specific VEC were activated with IFN-γ and TNF-α in an orbital shaker flow system. TNF-α, but not IFN-γ, induced hypoxia-inducible factor (HIF)-1α stabilization or endothelial nitric oxide synthase downregulation. Additionally, IFN-γ inhibited TNF-α-induced migration of aVEC. Also, IFN-γ triggered cytokine secretion and adhesion molecule expression in aVEC and vVEC. Finally, aVEC were more prone to cytokine-mediated monocyte adhesion under multiaxial flow conditions as compared with uniaxial flow. In conclusion, IFNs promote inflammation and reduce TNF-α-mediated migration in human VEC. Moreover, monocyte adhesion was higher in inflamed aVEC sheared under multiaxial flow, which may be relevant to understanding the initial stages of CAVD.


Subject(s)
Aortic Valve/metabolism , Endothelial Cells/metabolism , Interferon-alpha/pharmacology , Interferon-gamma/pharmacology , Signal Transduction/drug effects , Stress, Physiological/immunology , Aortic Valve/drug effects , Aortic Valve/immunology , Aortic Valve/pathology , Aortic Valve Stenosis/immunology , Calcinosis/immunology , Cell Adhesion/drug effects , Cell Movement/drug effects , Endothelial Cells/drug effects , Heart Transplantation , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Inflammation/chemically induced , Inflammation/immunology , Monocytes/metabolism , NF-kappa B/metabolism , Phenotype , STAT1 Transcription Factor/metabolism , THP-1 Cells , Transplant Recipients , Tumor Necrosis Factor-alpha/pharmacology
2.
Cardiovasc Res ; 117(13): 2506-2524, 2021 11 22.
Article in English | MEDLINE | ID: mdl-34432007

ABSTRACT

Calcific aortic valve disease (CAVD), and its clinical manifestation that is calcific aortic valve stenosis, is the leading cause for valve disease within the developed world, with no current pharmacological treatment available to delay or halt its progression. Characterized by progressive fibrotic remodelling and subsequent pathogenic mineralization of the valve leaflets, valve disease affects 2.5% of the western population, thus highlighting the need for urgent intervention. Whilst the pathobiology of valve disease is complex, involving genetic factors, lipid infiltration, and oxidative damage, the immune system is now being accepted to play a crucial role in pathogenesis and disease continuation. No longer considered a passive degenerative disease, CAVD is understood to be an active inflammatory process, involving a multitude of pro-inflammatory mechanisms, with both the adaptive and the innate immune system underpinning these complex mechanisms. Within the valve, 15% of cells evolve from haemopoietic origin, and this number greatly expands following inflammation, as macrophages, T lymphocytes, B lymphocytes, and innate immune cells infiltrate the valve, promoting further inflammation. Whether chronic immune infiltration or pathogenic clonal expansion of immune cells within the valve or a combination of the two is responsible for disease progression, it is clear that greater understanding of the immune systems role in valve disease is required to inform future treatment strategies for control of CAVD development.


Subject(s)
Adaptive Immunity , Aortic Valve Stenosis/immunology , Aortic Valve/immunology , Aortic Valve/pathology , Calcinosis/immunology , Hematopoietic System/immunology , Immune System/immunology , Immunity, Innate , Animals , Aortic Valve/metabolism , Aortic Valve/physiopathology , Aortic Valve Stenosis/metabolism , Aortic Valve Stenosis/physiopathology , Calcinosis/metabolism , Calcinosis/physiopathology , Cytokines/metabolism , Hematopoiesis , Hematopoietic System/metabolism , Hematopoietic System/pathology , Humans , Immune System/metabolism , Immune System/physiopathology , Inflammation Mediators/metabolism , Lipid Metabolism , Rheumatic Heart Disease/immunology , Rheumatic Heart Disease/metabolism , Rheumatic Heart Disease/physiopathology , Signal Transduction
3.
Circ Res ; 128(9): 1344-1370, 2021 04 30.
Article in English | MEDLINE | ID: mdl-33914601

ABSTRACT

Calcific aortic valve disease is dramatically increasing in global burden, yet no therapy exists outside of prosthetic replacement. The increasing proportion of younger and more active patients mandates alternative therapies. Studies suggest a window of opportunity for biologically based diagnostics and therapeutics to alleviate or delay calcific aortic valve disease progression. Advancement, however, has been hampered by limited understanding of the complex mechanisms driving calcific aortic valve disease initiation and progression towards clinically relevant interventions.


Subject(s)
Aortic Valve Stenosis/etiology , Aortic Valve/cytology , Aortic Valve/pathology , Calcinosis/etiology , Disease Progression , Endothelial Cells/physiology , Aortic Valve/immunology , Aortic Valve/physiology , Aortic Valve Stenosis/diagnosis , Aortic Valve Stenosis/immunology , Aortic Valve Stenosis/therapy , Calcinosis/diagnosis , Calcinosis/immunology , Calcinosis/therapy , Cell Adhesion Molecules/metabolism , Homeostasis , Humans , Immune System/physiology , Inflammation Mediators/metabolism , Nitric Oxide/biosynthesis , Nitric Oxide Synthase Type III/metabolism , Prognosis , Reactive Oxygen Species , Risk Factors , Vasculitis/etiology
4.
J Biomater Appl ; 35(9): 1218-1232, 2021 04.
Article in English | MEDLINE | ID: mdl-33478311

ABSTRACT

Over one million artificial heart valve transplantations are performed each year due to valvular stenosis or regurgitation. Among them, bioprosthetic heart valves (BHVs) are increasingly being used because of the absence of the need for lifelong anticoagulation. Almost all of the commercial BHVs are treated with Glutaraldehyde (GLUT). As GLUT-treated BHVs are prone to calcification and structural degradation, their durability is greatly reduced with a service life of only 12-15 years. The physiological structure and mechanical properties of the porcine aortic valve (PAV) are closer to that of a human heart valve, so in this study, PAV is used as the model to explore the comprehensive properties of the prepared BHVs by radical polymerization crosslinking method. We found that PAV treated by radical polymerization crosslinking method showed similar ECM stability and biaxial mechanical properties with GLUT-treated PAV. However, radical polymerization crosslinked PAV exhibited better cytocompatibility and endothelialization potential in vitro cell experiment as better anticalcification potential and reduced immune response than GLUT-treated PAV through subcutaneous animal experiments in rats. To conclude, a novel crosslinking method of non-glutaraldehyde fixation of xenogeneic tissues for the preparation of BHVs is expected.


Subject(s)
Aortic Valve/chemistry , Calcification, Physiologic , Heart Valve Prosthesis Implantation , Heart Valve Prosthesis , Animals , Aortic Valve/immunology , Bioprosthesis , Cross-Linking Reagents/chemistry , Glutaral/chemistry , Heart Valve Prosthesis Implantation/adverse effects , Heart Valve Prosthesis Implantation/methods , Human Umbilical Vein Endothelial Cells , Humans , Magnetic Resonance Spectroscopy , Male , Materials Testing , Microscopy, Electron, Scanning , Polymerization , Rats, Sprague-Dawley , Spectroscopy, Fourier Transform Infrared , Swine
5.
J Cardiovasc Transl Res ; 14(3): 503-512, 2021 06.
Article in English | MEDLINE | ID: mdl-33089488

ABSTRACT

We aim to investigate whether structural valve deterioration (SVD) of bioprosthetic xenogenic tissue heart valves (XTHVs) is associated with increased immune cell infiltration and whether co-expression of several chemokines correlates with this increase in immune infiltrate. Explanted XTHVs from patients undergoing redo valve replacement for SVD were obtained. Immunohistochemical, microscopic, and gene expression analysis approaches were used. XTHVs (n = 37) were obtained from 32 patients (mean 67.7 years) after a mean time of 11.6 years post-implantation. Significantly increased immune cellular infiltration was observed in the explanted SVD valves for all immune cell types examined, including T cells, macrophages, B cells, neutrophils, and plasma cells, compared to non-SVD controls. Furthermore, a significantly increased chemokine gradient in explanted SVD valves accompanied immune cell infiltration. These data suggest the development of SVD is associated with a significantly increased burden of immune cellular infiltrate correlated to the induction of a chemokine gradient around the XHTV, representing chronic immune rejection.Graphical abstract Proposed interaction between innate and adaptive immunity leading to the development of structural valve deterioration in xenogenic tissue heart valves.


Subject(s)
Aortic Valve/surgery , Bioprosthesis , Chemokines/metabolism , Heart Valve Prosthesis Implantation/instrumentation , Heart Valve Prosthesis , Mitral Valve/surgery , Prosthesis Failure , Adaptive Immunity , Aged , Aortic Valve/immunology , Aortic Valve/metabolism , Device Removal , Female , Heart Valve Prosthesis Implantation/adverse effects , Heterografts , Humans , Male , Mitral Valve/immunology , Mitral Valve/metabolism , Retrospective Studies , Time Factors , Treatment Outcome
6.
J Thromb Thrombolysis ; 51(3): 821-826, 2021 Apr.
Article in English | MEDLINE | ID: mdl-32749620

ABSTRACT

Infective endocarditis (IE) remains a severe illness with high mortality rate, despite advances in antibiotic therapy and cardiac surgery. If infectious bacteria and platelets are two key players of human IE vegetation developmental process, their interactions and respective roles in fully developed late-stage IE vegetations remain obscure. The objective of this study was to better understand the organization of the different components of the IE vegetation and to provide a detailed description of this vegetation ultrastructure. A late stage Staphylococcal endocarditic vegetation was provided from a 13 years teenager patient. After reception of the surgical piece, we carried out a histological study using routine methods, notably the hematoxylin-eosin-saffron staining. Labeling with the anti-CD 61 antibody was also carried out. In a second step, we used transmission electron microscopy to describe the different regions making up the vegetation. Our ultrastructural study revealed vegetation was clearly composed by three different regions and identified the specific location of the bacteria and platelets in the vegetation tissues. Histological analysis showed that platelets and Staphylococcus aureus were not co-localized. Electron microscopy study confirmed that S. aureus were found at distance from platelets, as well from immune cells, embedded in a biofilm and/or a necrotic area. These results reveal a development of a deep bacteria-only niche in vegetation, raising questions about medication access to these microorganisms. Vegetation composed of three regions: a region rich in bacteria incorporated into the necrotic tissue, the second region composed of fibrin filaments and the third region rich in platelets and free of bacteria.


Subject(s)
Aortic Valve Insufficiency , Aortic Valve , Endocarditis, Bacterial , Heart Valve Prosthesis Implantation/methods , Staphylococcal Infections , Staphylococcus aureus/isolation & purification , Adolescent , Anti-Bacterial Agents/administration & dosage , Aortic Valve/diagnostic imaging , Aortic Valve/immunology , Aortic Valve/microbiology , Aortic Valve/pathology , Aortic Valve Insufficiency/diagnosis , Aortic Valve Insufficiency/etiology , Aortic Valve Insufficiency/physiopathology , Aortic Valve Insufficiency/surgery , Blood Platelets/pathology , Echocardiography/methods , Endocarditis, Bacterial/blood , Endocarditis, Bacterial/microbiology , Endocarditis, Bacterial/pathology , Humans , Immunohistochemistry , Male , Microscopy, Electron, Transmission/methods , Staphylococcal Infections/blood , Staphylococcal Infections/diagnosis , Staphylococcal Infections/physiopathology , Treatment Outcome
7.
Arterioscler Thromb Vasc Biol ; 40(6): e153-e165, 2020 06.
Article in English | MEDLINE | ID: mdl-32295422

ABSTRACT

OBJECTIVE: Macrophages have been described in calcific aortic valve disease, but it is unclear if they promote or counteract calcification. We aimed to determine how macrophages are involved in calcification using the Notch1+/- model of calcific aortic valve disease. Approach and Results: Macrophages in wild-type and Notch1+/- murine aortic valves were characterized by flow cytometry. Macrophages in Notch1+/- aortic valves had increased expression of MHCII (major histocompatibility complex II). We then used bone marrow transplants to test if differences in Notch1+/- macrophages drive disease. Notch1+/- mice had increased valve thickness, macrophage infiltration, and proinflammatory macrophage maturation regardless of transplanted bone marrow genotype. In vitro approaches confirm that Notch1+/- aortic valve cells promote macrophage invasion as quantified by migration index and proinflammatory phenotypes as quantified by Ly6C and CCR2 positivity independent of macrophage genotype. Finally, we found that macrophage interaction with aortic valve cells promotes osteogenic, but not dystrophic, calcification and decreases abundance of the STAT3ß isoform. CONCLUSIONS: This study reveals that Notch1+/- aortic valve disease involves increased macrophage recruitment and maturation driven by altered aortic valve cell secretion, and that increased macrophage recruitment promotes osteogenic calcification and alters STAT3 splicing. Further investigation of STAT3 and macrophage-driven inflammation as therapeutic targets in calcific aortic valve disease is warranted.


Subject(s)
Aortic Valve Stenosis/pathology , Aortic Valve/pathology , Calcinosis/pathology , Macrophages/physiology , STAT3 Transcription Factor/physiology , Animals , Aortic Valve/immunology , Aortic Valve/physiopathology , Aortic Valve Stenosis/immunology , Aortic Valve Stenosis/physiopathology , Bone Marrow Transplantation , Calcinosis/immunology , Calcinosis/physiopathology , Cell Movement , Cyclic S-Oxides/pharmacology , Disease Models, Animal , Gene Expression , Genotype , Humans , Inflammation/pathology , Macrophages/chemistry , Macrophages/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Osteogenesis , Receptor, Notch1/analysis , Receptor, Notch1/genetics , Receptor, Notch1/physiology , STAT3 Transcription Factor/antagonists & inhibitors , STAT3 Transcription Factor/genetics
8.
Am J Physiol Heart Circ Physiol ; 317(1): H141-H155, 2019 07 01.
Article in English | MEDLINE | ID: mdl-31050556

ABSTRACT

Calcific aortic valve disease (CAVD) is highly prevalent and has no pharmaceutical treatment. Surgical replacement of the aortic valve has proved effective in advanced disease but is costly, time limited, and in many cases not optimal for elderly patients. This has driven an increasing interest in noninvasive therapies for patients with CAVD. Adaptive immune cell signaling in the aortic valve has shown potential as a target for such a therapy. Up to 15% of cells in the healthy aortic valve are hematopoietic in origin, and these cells, which include macrophages, T lymphocytes, and B lymphocytes, are increased further in calcified specimens. Additionally, cytokine signaling has been shown to play a causative role in aortic valve calcification both in vitro and in vivo. This review summarizes the physiological presence of hematopoietic cells in the valve, innate and adaptive immune cell infiltration in disease states, and the cytokine signaling pathways that play a significant role in CAVD pathophysiology and may prove to be pharmaceutical targets for this disease in the near future.


Subject(s)
Adaptive Immunity , Aortic Valve Stenosis/immunology , Aortic Valve/immunology , Aortic Valve/pathology , Calcinosis/immunology , Lymphocytes/immunology , Myeloid Cells/immunology , Animals , Aortic Valve/metabolism , Aortic Valve Stenosis/metabolism , Aortic Valve Stenosis/pathology , Calcinosis/metabolism , Calcinosis/pathology , Cytokines/immunology , Cytokines/metabolism , Humans , Immunity, Innate , Lymphocytes/metabolism , Lymphocytes/pathology , Myeloid Cells/metabolism , Myeloid Cells/pathology , Signal Transduction
9.
J Card Surg ; 34(6): 482-494, 2019 Jun.
Article in English | MEDLINE | ID: mdl-31012137

ABSTRACT

Bicuspid aortic valves (BAVs) are the most common congenital cardiac condition and are characterized by a structural abnormality whereby the aortic valve is composed of two leaflets instead of being trileaflet. It is linked to an increased risk for a variety of complications of the aorta, many with an immunological pathogenesis. The aim of this study is to review and analyze the literature regarding immunological processes involving BAVs, associated common pathologies, and their incidence in the population. This study will also examine current trends in surgical and therapeutic approaches to treatment and discuss the future direction of BAV treatment.


Subject(s)
Aortic Valve/abnormalities , Heart Valve Diseases/complications , Heart Valve Diseases/immunology , Immune System Diseases/etiology , Immune System Diseases/immunology , Aortic Valve/immunology , Bicuspid Aortic Valve Disease , Heart Valve Diseases/epidemiology , Heart Valve Diseases/therapy , Humans , Immune System Diseases/epidemiology , Incidence , Risk , Transcatheter Aortic Valve Replacement
10.
J Surg Res ; 230: 101-109, 2018 10.
Article in English | MEDLINE | ID: mdl-30100024

ABSTRACT

BACKGROUND: Calcific aortic stenosis is a chronic inflammatory disease. Proinflammatory stimulation via toll-like receptor 4 (TLR4) causes the aortic valve interstitial cell (AVIC) to undergo phenotypic change. The AVIC first assumes an inflammatory phenotype characterized by the production of inflammatory mediators such as intercellular adhesion molecule-1 (ICAM-1), interleukin-8 (IL-8), and monocyte chemoattractant protein-1 (MCP-1). This change has been linked with an osteogenic phenotypic response. Statins have recently been shown to have anti-inflammatory properties. We therefore hypothesized that statins may have an anti-inflammatory effect on human AVICs by downregulation of TLR4-stimulated inflammatory responses. Our purposes were (1) to determine the effect of simvastatin on TLR4-induced expression of inflammatory mediators in human AVICs and (2) to determine the mechanism(s) through which simvastatin exert this effect. MATERIALS AND METHODS: Human AVICs were isolated from the explanted hearts of four patients undergoing cardiac transplantation. Cells were treated with simvastatin (50 µM) for 1 h before stimulation with TLR4 agonist lipopolysaccharide (LPS, 0.2 µg/mL). Immunoblotting (IB) was used to analyze cell lysates for ICAM-1 expression, and enzyme-linked immunosorbent assay was used to detect IL-8 and MCP-1 in cell culture media. Likewise, lysates were analyzed for TLR4 and nuclear factor-kappa B activation (IB). After simvastatin treatment, lysates were analyzed for TLR4 levels (IB). Statistics were by analysis of variance (P < 0.05). RESULTS: Simvastatin reduced TLR4-induced ICAM-1, IL-8, and MCP-1 expression in AVICs. Simvastatin down-regulated TLR4 levels and suppressed TLR4-induced phosphorylation of nuclear factor-kappa B. CONCLUSIONS: These data demonstrate the potential of a medical therapy (simvastatin) to impact the pathogenesis of aortic stenosis.


Subject(s)
Aortic Valve Stenosis/drug therapy , Aortic Valve/pathology , Calcinosis/drug therapy , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Simvastatin/pharmacology , Toll-Like Receptor 4/immunology , Adult , Aortic Valve/cytology , Aortic Valve/immunology , Aortic Valve Stenosis/immunology , Aortic Valve Stenosis/pathology , Calcinosis/immunology , Calcinosis/pathology , Cardiomyopathy, Dilated/surgery , Cells, Cultured , Drug Evaluation, Preclinical , Heart Transplantation , Humans , Hydroxymethylglutaryl-CoA Reductase Inhibitors/therapeutic use , Male , Middle Aged , Myofibroblasts , Primary Cell Culture , Simvastatin/therapeutic use
11.
Oxid Med Cell Longev ; 2018: 5879281, 2018.
Article in English | MEDLINE | ID: mdl-29854087

ABSTRACT

Bicuspid valve disease is associated with the development of thoracic aortic aneurysm. The molecular mechanisms underlying this association still need to be clarified. Here, we evaluated the circulating levels of T and B lymphocyte subsets associated with the development of vascular diseases in patients with bicuspid aortic valve or tricuspid aortic valve with and without thoracic aortic aneurysm. We unveiled that the circulating levels of the MAIT, CD4+IL-17A+, and NKT T cell subsets were significantly reduced in bicuspid valve disease cases, when compared to tricuspid aortic valve cases in either the presence or the absence of thoracic aortic aneurysm. Among patients with tricuspid aortic valve, these cells were higher in those also affected by thoracic aortic aneurysm. Similar data were obtained by examining CD19+ B cells, naïve B cells (IgD+CD27-), memory unswitched B cells (IgD+CD27+), memory switched B cells (IgD-CD27+), and double-negative B cells (DN) (IgD-CD27-). These cells resulted to be lower in subjects with bicuspid valve disease with respect to patients with tricuspid aortic valve. In whole, our data indicate that patients with bicuspid valve disease show a quantitative reduction of T and B lymphocyte cell subsets. Future studies are encouraged to understand the molecular mechanisms underlying this observation and its pathophysiological significance.


Subject(s)
Aortic Valve/abnormalities , B-Lymphocytes/immunology , Heart Valve Diseases/immunology , T-Lymphocytes/immunology , Aortic Valve/immunology , Bicuspid Aortic Valve Disease , Female , Humans , Male , Middle Aged
12.
Cardiovasc Pathol ; 35: 1-7, 2018.
Article in English | MEDLINE | ID: mdl-29727769

ABSTRACT

INTRODUCTION: Aortic stenosis (AS) is the most common acquired valvular heart disease in adults. Immune system involvement becomes evident during AS development. We sought to investigate the role of different circulating lymphocyte and monocyte subpopulations, with focus on CD4+CD8+ and natural killer T (NKT) cells, in AS. MATERIAL AND METHODS: Blood samples and aortic valves were obtained from patients undergoing elective aortic valve surgery. Valves were dissected and underwent genetic analyses and calcium content assessment. Lymphocytes and monocytes subsets were assessed by flow cytometry. RESULTS: Thirty-eight AS patients were studied. Maximal transvalvular pressure gradient (PGmax) as well as mean transvalvular pressure gradient (PGmean) correlated with the CD4+CD8+ lymphocyte count (r=0.35, P=.03 and r=0.43, P=.006, respectively) and fraction (r=0.43, P=.007 and r=0.48, P=.002, respectively). PGmax and PGmean correlated with CD16+CD56+CD3+ NKT cell count (r=0.39, P=.01 and r=0.43, P=.007, respectively) and fraction (r=0.49, P=.002 and r=0.47, P=.003, respectively). The classical monocyte subpopulation increased after the surgery by 68% (P<.0001). Patients after mini-sternotomy surgery had 47% lower nonclassical monocyte counts than those with full-sternotomy (P=.03). Patients treated with statins had significantly lower postoperative levels of both classical (-25%, P=.04) and nonclassical monocytes (-37%, P=.004) than nontreated individuals. CONCLUSIONS: In patients with severe isolated AS, CD4+CD8+ T cells and CD16+CD56+CD3+ NKT cells are associated with AV pressure gradients. Postoperative monocyte levels are affected by procedure invasiveness and use of statins.


Subject(s)
Aortic Valve Stenosis/immunology , Aortic Valve/immunology , Aortic Valve/pathology , Calcinosis/immunology , Monocytes/immunology , T-Lymphocyte Subsets/immunology , Aged , Aortic Valve/physiopathology , Aortic Valve/surgery , Aortic Valve Stenosis/pathology , Aortic Valve Stenosis/physiopathology , Aortic Valve Stenosis/surgery , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/pathology , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/pathology , Calcinosis/pathology , Calcinosis/physiopathology , Calcinosis/surgery , Cardiac Surgical Procedures , Female , Flow Cytometry , Hemodynamics , Humans , Hydroxymethylglutaryl-CoA Reductase Inhibitors/therapeutic use , Immunophenotyping/methods , Male , Middle Aged , Monocytes/classification , Monocytes/pathology , Natural Killer T-Cells/immunology , Natural Killer T-Cells/pathology , Phenotype , Severity of Illness Index , Sternotomy , T-Lymphocyte Subsets/classification , T-Lymphocyte Subsets/pathology , Treatment Outcome
13.
J Heart Valve Dis ; 26(3): 334-343, 2017 05.
Article in English | MEDLINE | ID: mdl-29092120

ABSTRACT

BACKGROUND AND AIM OF THE STUDY: The aim of this retrospective study was to evaluate the inflammatory response in patients with aortic and/or mitral prostheses, and to correlate the level of inflammatory markers with prosthesis functionality. METHODS: A total of 48 patients with biological or mechanical prostheses was included in the study, in which levels of tumor necrosis factor-alpha (TNFα), interleukin (IL)-1, -4, and -6, interferon-gamma (IFNγ), osteopontin (OPN), intercellular adhesion molecule (ICAM), vascular cell adhesion molecule (VCAM), endothelin-1 and C-reactive protein were analyzed. Functionality of the prosthesis was evaluated using transthoracic echocardiography at three years after surgery. RESULTS: The mean period from the date of surgery was seven years. High levels of IL-1 were found in patients with mechanical prostheses compared to those with bioprostheses (p = 0.04). Patients with aortic bioprostheses and stenosis had higher levels of OPN and endothelin-1, those with aortic mechanical prostheses with stenosis had increased levels of matrix metalloproteinase (MMP)-9, OPN and ICAM, and those with aortic mechanical leakage had increased levels of MMP-1 and endothelin-1. In mitral bioprostheses with leakage of endothelin-1, ICAM and MMP-9 levels were increased, while in mechanical prostheses with leakage there were increases of ICAM and endothelin-1. Tricuspid bioprostheses with double lesions had increased levels of OPN and endothelin-1. CONCLUSIONS: Valvular dysfunction was similar across the types of prosthesis material. IL-1 was increased in subjects with mechanical prostheses independently of dysfunction, while in biological prostheses there were increases in OPN and endothelin-1, and these were related to valvular dysfunction. Given that in the analysis of durability and functionality there were no significant differences between biological and mechanical prostheses, biological prostheses may represent the first treatment option in patients with low economic resources, the elderly, and even young patients.


Subject(s)
Aortic Valve/surgery , Bioprosthesis , Heart Valve Prosthesis Implantation/adverse effects , Heart Valve Prosthesis , Inflammation Mediators/immunology , Inflammation/immunology , Mitral Valve/surgery , Pericardium/transplantation , Aged , Aged, 80 and over , Animals , Aortic Valve/diagnostic imaging , Aortic Valve/immunology , Aortic Valve/physiopathology , Cattle , Echocardiography, Transesophageal , Female , Heterografts , Humans , Inflammation/blood , Inflammation/diagnosis , Inflammation Mediators/blood , Male , Middle Aged , Mitral Valve/diagnostic imaging , Mitral Valve/immunology , Mitral Valve/physiopathology , Pericardium/diagnostic imaging , Pericardium/immunology , Prosthesis Design , Prosthesis Failure , Retrospective Studies , Time Factors , Treatment Outcome
14.
Am J Pathol ; 187(6): 1413-1425, 2017 Jun.
Article in English | MEDLINE | ID: mdl-28431214

ABSTRACT

In calcific aortic valve disease (CAVD), activated T lymphocytes localize with osteoclast regions; however, the functional consequences of this association remain unknown. We hypothesized that CD8+ T cells modulate calcification in CAVD. CAVD valves (n = 52) dissected into noncalcified and calcified portions were subjected to mRNA extraction, real-time quantitative PCR, enzyme-linked immunosorbent assay, and immunohistochemical analyses. Compared with noncalcified portions, calcified regions exhibited elevated transcripts for CD8, interferon (IFN)-γ, CXCL9, Perforin 1, Granzyme B, and heat shock protein 60. Osteoclast-associated receptor activator of NK-κB ligand (RANKL), tartrate-resistant acid phosphatase (TRAP), and osteoclast-associated receptor increased significantly. The stimulation of tissue with phorbol-12-myristate-13-acetate and ionomycin, recapitulating CAVD microenvironment, resulted in IFN-γ release. Real-time quantitative PCR detected mRNAs for CD8+ T-cell activation (Perforin 1, Granzyme B). In stimulated versus unstimulated organoid cultures, elevated IFN-γ reduced the mRNAs encoding for RANKL, TRAP, and Cathepsin K. Molecular imaging showed increased calcium signal intensity in stimulated versus unstimulated parts. CD14+ monocytes treated either with recombinant human IFN-γ or with conditioned media-derived IFN-γ exhibited low levels of Cathepsin K, TRAP, RANK, and tumor necrosis factor receptor-associated factor 6 mRNAs, whereas concentrations of the T-cell co-activators CD80 and CD86 increased in parallel with reduced osteoclast resorptive function, effects abrogated by neutralizing anti-IFN-γ antibodies. CD8+ cell-derived IFN-γ suppresses osteoclast function and may thus favor calcification in CAVD.


Subject(s)
Aortic Valve Stenosis/immunology , Aortic Valve/pathology , CD8-Positive T-Lymphocytes/immunology , Calcinosis/immunology , Calcium/metabolism , Interferon-gamma/immunology , Osteoclasts/metabolism , Aged , Aged, 80 and over , Aortic Valve/immunology , Aortic Valve/metabolism , Aortic Valve/surgery , Aortic Valve Stenosis/metabolism , Aortic Valve Stenosis/surgery , Calcinosis/metabolism , Calcinosis/surgery , Female , Gene Expression Regulation/immunology , Heart Valve Prosthesis Implantation , Humans , Interferon-gamma/biosynthesis , Interferon-gamma/genetics , Lymphocyte Activation/immunology , Male , Middle Aged , Osteoclasts/immunology , Osteoclasts/physiology , RANK Ligand/metabolism , Tissue Culture Techniques/methods
15.
Atherosclerosis ; 260: 1-7, 2017 05.
Article in English | MEDLINE | ID: mdl-28319871

ABSTRACT

BACKGROUND AND AIMS: Elevated levels of lipoprotein(a) [Lp(a)] and oxidized phospholipids on apolipoprotein B-100 (OxPL-apoB) predict the progression of pre-existing mild-to-moderate calcific aortic valve stenosis (CAVS). Whether indirect markers of oxidation-specific epitopes (OSE) are also predictive is not known. The association between IgG and IgM autoantibodies and malondialdehyde-modified low density lipoprotein (MDA-LDL) and IgG and IgM apolipoprotein B immune complexes (apoB-IC), and the hemodynamic progression rate of CAVS was determined in the ASTRONOMER (Aortic Stenosis Progression Observation: Measuring Effects of Rosuvastatin, NCT00800800) trial. METHODS: Plasma levels of IgG and IgM MDA-LDL and apoB-IC were measured in 220 patients with mild-to-moderate CAVS from the ASTRONOMER trial. The endpoint of this study was the progression rate of CAVS, measured by the annualized increase in peak aortic jet velocity (Vpeak) over a median follow-up of 3.5 [2.9-4.5] years. RESULTS: There was no difference in the progression rate of CAVS across tertiles of IgG and IgM MDA-LDL and apoB-IC levels (all p > 0.05). After multivariable analysis, no marker reached significance level to predict faster CAVS progression or need for aortic valve replacement (all p > 0.05). There was no interaction between the OSE antibody titers and plasma levels of Lp(a) or OxPL-apoB, as well as age, with regards to the progression rate of CAVS. CONCLUSIONS: Autoantibody titers to MDA-LDL and apoB-IC, which are an indirect measurement of OSE, unlike direct measurements of OxPL-apoB or their major lipoprotein carrier Lp(a), do not predict the progression of CAVS or need for AVR.


Subject(s)
Antibodies, Anti-Idiotypic/blood , Aortic Valve Stenosis/immunology , Aortic Valve/pathology , Autoantibodies/blood , Calcinosis/complications , Immunoglobulin G/blood , Rosuvastatin Calcium/administration & dosage , Adolescent , Adult , Aged , Aged, 80 and over , Antibodies, Anti-Idiotypic/immunology , Aortic Valve/immunology , Aortic Valve Stenosis/complications , Aortic Valve Stenosis/drug therapy , Aortic Valve Stenosis/etiology , Autoantibodies/immunology , Biomarkers/blood , Calcinosis/drug therapy , Calcinosis/immunology , Disease Progression , Dose-Response Relationship, Drug , Epitopes , Female , Follow-Up Studies , Humans , Immunoglobulin G/immunology , Male , Middle Aged , Oxidation-Reduction , Retrospective Studies , Time Factors , Young Adult
16.
BMC Cardiovasc Disord ; 17(1): 52, 2017 02 03.
Article in English | MEDLINE | ID: mdl-28158996

ABSTRACT

BACKGROUND: The diagnosis of Immunoglobulin G4 (IgG4)-related disease (IgG4-RD), in general, depends on serum IgG4 concentrations and histopathological findings; therefore, diagnosis of IgG4-RD in cardiovascular organs/tissues is often difficult owing to the risk of tissue sampling. METHODS: Prevalence of IgG4-positive lymphoplasmacytic infiltration in 103 consecutive cardiovascular surgical samples from 98 patients with various cardiovascular diseases was analyzed immunohistochemically. RESULTS: The diagnoses of the enrolled patients included aortic aneurysm (abdominal, n = 8; thoracic, n = 9); aortic dissection (n = 20); aortic stenosis (n = 24), aortic regurgitation (n = 10), and mitral stenosis/regurgitation (n = 17). In total, 10 (9.7%) of the 103 specimens showed IgG4-positive cell infiltration with various intensities; five of these were aortic valve specimens from aortic stenosis, and IgG4-positive cell infiltration was present at >10 /HPF in three of them. In one aortic wall sample from an abdominal aortic aneurysm, various histopathological features of IgG4-RD, such as IgG4-positive cell infiltration, obliterating phlebitis, and storiform fibrosis, were observed. CONCLUSIONS: IgG4-positive cell infiltration was observed in 9.7% of the surgical cardiovascular specimens, mainly in the aortic valve from aortic stenosis and in the aortic wall from aortic aneurysm. Whether IgG4-positive cell infiltration has pathophysiological importance in the development or progression of cardiovascular diseases should be investigated in future studies.


Subject(s)
Cardiovascular Diseases/immunology , Chemotaxis, Leukocyte , Immunoglobulin G/analysis , Plasma Cells/immunology , Aged , Aged, 80 and over , Aorta/immunology , Aorta/pathology , Aortic Aneurysm/immunology , Aortic Aneurysm/pathology , Aortic Valve/immunology , Aortic Valve/pathology , Aortic Valve Stenosis/immunology , Aortic Valve Stenosis/pathology , Aortography/methods , Biomarkers/analysis , Biopsy , Cardiovascular Diseases/diagnostic imaging , Cardiovascular Diseases/pathology , Cardiovascular Diseases/surgery , Computed Tomography Angiography , Cross-Sectional Studies , Female , Humans , Immunohistochemistry , Male , Middle Aged , Plasma Cells/pathology
17.
Am J Physiol Cell Physiol ; 312(4): C407-C417, 2017 Apr 01.
Article in English | MEDLINE | ID: mdl-28052863

ABSTRACT

Calcific aortic valve disease is a chronic inflammatory condition, and the inflammatory responses of aortic valve interstitial cells (AVICs) play a critical role in the disease progression. Double-stranded RNA (dsRNA) released from damaged or stressed cells is proinflammatory and may contribute to the mechanism of chronic inflammation observed in diseased aortic valves. The objective of this study is to determine the effect of dsRNA on AVIC inflammatory responses and the underlying mechanism. AVICs from normal human aortic valves were stimulated with polyinosinic-polycytidylic acid [poly(I:C)], a mimic of dsRNA. Poly(I:C) increased the production of IL-6, IL-8, monocyte chemoattractant protein-1, and ICAM-1. Poly(I:C) also induced robust activation of ERK1/2 and NF-κB. Knockdown of Toll-like receptor 3 (TLR3) or Toll-IL-1 receptor domain-containing adapter-inducing IFN-ß (TRIF) suppressed ERK1/2 and NF-κB p65 phosphorylation and reduced inflammatory mediator production induced by poly(I:C). Inhibition of NF-κB, not ERK1/2, reduced inflammatory mediator production in AVICs exposed to poly(I:C). Interestingly, inhibition of NF-κB by prevention of p50 migration failed to suppress inflammatory mediator production. NF-κB p65 intranuclear translocation induced by the TLR4 agonist was reduced by inhibition of p50 migration; however, poly(I:C)-induced p65 translocation was not, although the p65/p50 heterodimer is present in AVICs. Poly(I:C) upregulates the production of multiple inflammatory mediators through the TLR3-TRIF-NF-κB pathway in human AVICs. The NF-κB activated by dsRNA appears not to be the canonical p65/p50 heterodimers.


Subject(s)
Aortic Valve/immunology , Heart Valve Diseases/immunology , Inflammation Mediators/immunology , NF-kappa B/immunology , RNA, Double-Stranded/immunology , Vascular Calcification/immunology , Adaptor Proteins, Vesicular Transport/immunology , Aortic Valve/cytology , Cell Line , Humans , Signal Transduction/immunology , Toll-Like Receptor 3/immunology , Up-Regulation/immunology
18.
Exp Clin Transplant ; 15(3): 329-336, 2017 Jun.
Article in English | MEDLINE | ID: mdl-27210227

ABSTRACT

OBJECTIVES: Endothelial cells harbor many antigenic determinants that may be targets for the immune system. The aim of this study was to determine the immunologic effects of decellularization, using 3 different methods, on xenograft rejection. MATERIALS AND METHODS: In a sterile plate containing phosphate-buffered saline, fresh sheep aortic heart valves were decellularized using 3 different enzymatic methods: with 900 µg/mL of collagenase at 40°C (method A), with 450 µg/mL of collagenase at 4°C (method B), and with 900 µg/mL of collagenase at 4°C (method C). Intact and decellularized valves were implanted subdermally into inbred male albino rabbits and extracted after 21 days (extra valve pieces were also extracted after 60 days, as control samples, for assessing chronic rejection). Valves were histologically analyzed for inflammatory cell infiltration. Subendothelial structure integrity was determined using surface electron microscope. RESULTS: No inflammatory cell infiltration was seen around the decellularized valve with method A, and no subendothelial structure change was observed by surface electron microscope. Infiltration of immune cells involved in rejection was not seen around valves decellularized with method B, although the subendothelial structure was relatively preserved and valve stiffness was increased. With method C, we observed a foreign body-type reaction around the intact valve and the decellularized valve. CONCLUSIONS: Method A is considered the optimal method of decellularization in our study, as this method significantly reduced the immune response to xenograft tissue, while maintaining subendothelial tissue.


Subject(s)
Aortic Valve/transplantation , Bioprosthesis , Heart Valve Prosthesis Implantation/instrumentation , Heart Valve Prosthesis , Tissue Engineering/methods , Animals , Aortic Valve/immunology , Aortic Valve/ultrastructure , Collagenases/metabolism , Endothelial Cells/immunology , Graft Rejection/immunology , Graft Rejection/pathology , Graft Survival , Heart Valve Prosthesis Implantation/adverse effects , Heterografts , Male , Prosthesis Design , Rabbits , Sheep, Domestic , Time Factors
19.
Arthritis Rheumatol ; 68(10): 2476-86, 2016 10.
Article in English | MEDLINE | ID: mdl-27111864

ABSTRACT

OBJECTIVE: The spondyloarthritides (SpA) are a group of rheumatic diseases characterized by ossification and inflammation of entheseal tissue, the region where tendon attaches to bone. Interleukin-23 (IL-23) is involved in the pathogenesis of SpA by acting on IL-23 receptor (IL-23R) expressed on enthesis-resident lymphocytes. Upon IL-23 binding, CD3+CD4-CD8- tissue-resident lymphocytes secrete IL-17A and IL-22, leading to inflammation, bone loss, and ossification. Knowledge about enthesis-resident lymphocytes remains fragmentary, and the contribution of entheseal γ/δ T cells in particular is not clear. This study was undertaken to investigate the presence of γ/δ T cells in the enthesis. METHODS: We used 2-photon microscopy and flow cytometry to analyze entheseal lymphocytes from C57BL/6, Tcrd-H2BeGFP, Rorc-GFP, and IL-23R-eGFP mice. To analyze entheseal γ/δ T cells in IL-23-induced inflammation, Tcrd-H2BeGFP mice were crossed with mice of the susceptible B10.RIII background. Hydrodynamic injection of IL-23 minicircle DNA was performed for overexpression of IL-23 and induction of inflammation. Light-sheet fluorescence microscopy was used to visualize arthritic inflammation. RESULTS: Activated Vγ6+CD27- γ/δ T cells were abundant in uninflamed entheseal tissue and constituted the large majority of retinoic acid receptor-related orphan nuclear receptor γt (RORγt)+IL-23R+ enthesis-resident lymphocytes. Fetal thymus-dependent γ/δ T cells were the main source of IL-17A at the enthesis. Under inflammatory conditions, γ/δ T cells increased in number at the Achilles tendon enthesis, aortic root, and adjacent to the ciliary body. CONCLUSION: Entheseal γ/δ T cells are derived from fetal thymus and are maintained as self-renewing tissue-resident cells. As main IL-17A producers within tissues exposed to mechanical stress including enthesis, γ/δ T cells are key players in the pathogenesis of IL-23-induced local inflammation.


Subject(s)
Achilles Tendon/immunology , Aortic Valve/immunology , Ciliary Body/immunology , Interleukin-23/immunology , Receptors, Antigen, T-Cell, gamma-delta/immunology , Spondylarthropathies/immunology , T-Lymphocyte Subsets/immunology , Achilles Tendon/pathology , Animals , Ankle Joint/immunology , Ankle Joint/pathology , Aortic Valve/pathology , Ciliary Body/pathology , Enthesopathy/immunology , Enthesopathy/pathology , Flow Cytometry , Green Fluorescent Proteins/genetics , Interleukin-17/immunology , Interleukins/immunology , Mice , Mice, Transgenic , Microscopy, Fluorescence , Nuclear Receptor Subfamily 1, Group F, Member 3/immunology , Spondylarthropathies/pathology , T-Lymphocyte Subsets/pathology , X-Ray Microtomography , Interleukin-22
20.
Medicine (Baltimore) ; 95(3): e2564, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26817911

ABSTRACT

To determine the prevalence of antineutrophil cytoplasmic antibodies (ANCA) in patients with infective endocarditis (IE) in internal medicine; and to compare clinical and biochemical features and outcome between patients exhibiting IE with and without ANCA.Fifty consecutive patients with IE underwent ANCA testing. The medical records of these patients were reviewed.Of the 50 patients with IE, 12 exhibited ANCA (24%). ANCA-positive patients with IE exhibited: longer duration between the onset of first symptoms and IE diagnosis (P = 0.02); and more frequently: weight loss (P = 0.017) and renal impairment (P = 0.08), lower levels of C-reactive protein (P = 0.0009) and serum albumin (P = 0.0032), involvement of both aortic and mitral valves (P = 0.009), and longer hospital stay (P = 0.016). Under multivariate analysis, significant factors for ANCA-associated IE were: longer hospital stay (P = 0.004), lower level of serum albumin (P = 0.02), and multiple valve involvement (P = 0.04). Mortality rate was 25% in ANCA patients; death was because of IE complications in all these patients.Our study identifies a high prevalence of ANCA in unselected patients with IE in internal medicine (24%). Our findings further underscore that ANCA may be associated with a subacute form of IE leading to multiple valve involvement and more frequent renal impairment. Because death was due to IE complications in all patients, our data suggest that aggressive therapy may be required to improve such patients' outcome.


Subject(s)
Antibodies, Antineutrophil Cytoplasmic/blood , Endocarditis/immunology , Adult , Aged , Aged, 80 and over , Aortic Valve/immunology , Bicuspid Aortic Valve Disease , C-Reactive Protein/analysis , Endocarditis/blood , Female , Heart Defects, Congenital/blood , Heart Defects, Congenital/immunology , Heart Valve Diseases/blood , Heart Valve Diseases/immunology , Humans , Male , Middle Aged , Mitral Valve , Serum Albumin/analysis , Young Adult
SELECTION OF CITATIONS
SEARCH DETAIL
...