51
|
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: 1.8] [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
|
52
|
Liao M, Liu CL, Lv BJ, Zhang JY, Cheng L, Cheng X, Lindholt JS, Rasmussen LM, Shi GP. Plasma cytokine levels and risks of abdominal aortic aneurysms: A population-based prospective cohort study. Ann Med 2015; 47:245-52. [PMID: 25856542 PMCID: PMC4669056 DOI: 10.3109/07853890.2015.1019916] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
BACKGROUND Abdominal aortic aneurysm (AAA) is characterized by inflammatory cell accumulation in AAA lesions that produce inflammatory cytokines and advance its pathogenesis. Peripheral cytokines may predict the degree or risk of AAA. METHODS AND RESULTS ELISA determined plasma interleukin-6 (IL6), IL10, IL17A, IFN-γ, and C-reactive protein (CRP) from 476 AAA patients and 200 controls. AAA patients had lower IL6, IFN-γ, IL10, IL17A, and higher CRP than controls. IL10 correlated positively with IFN-γ, IL17A, or IL6, but not CRP in control or AAA populations. IL10 associated negatively with systolic blood pressure, whereas CRP associated positively with diastolic blood pressure and body mass index. CRP was an independent AAA risk factor and correlated positively with aortic diameters before and after adjustments for other risk factors. IFN-γ, IL17A, and CRP correlated positively with cross-sectional AAA area after adjustment. IL10 correlated positively with AAA growth rate before and after adjustment. The risk of death doubled in AAA patients with CRP levels above the median. CONCLUSIONS Reduced IFN-γ, IL10, and IL17A in AAA patients, positive correlations of IFN-γ and IL17A with cross-sectional AAA area, IL10 with AAA growth rate, and IL10 with IFN-γ and IL17A suggest combined Th1, Th2, and Th17 immune responses in human AAAs.
Collapse
Affiliation(s)
- Mengyang Liao
- Institute of Cardiology, Union Hospital, Tongji Medical College of Huazhong University of Science and Technology , Wuhan , China
| | | | | | | | | | | | | | | | | |
Collapse
|
53
|
Mao N, Gu T, Shi E, Zhang G, Yu L, Wang C. Phenotypic switching of vascular smooth muscle cells in animal model of rat thoracic aortic aneurysm. Interact Cardiovasc Thorac Surg 2015; 21:62-70. [PMID: 25829166 DOI: 10.1093/icvts/ivv074] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2014] [Accepted: 03/04/2015] [Indexed: 11/13/2022] Open
Abstract
OBJECTIVES To explore if there is phenotypic switching in the vascular smooth muscle cells (vSMCs) of rat thoracic aortic aneurysms and the role it plays in the process of aneurysm formation. METHODS Male SD white rats were assigned randomly to the aneurysm group (AG) and control group (CG). The animal aneurysm model was obtained by soaking the peri-adventitia with porcine pancreatic elastase (PPE). The rats in the CG were given saline to provide contrast. A vascular ultrasound was used to monitor the diameter of the aneurysm. Specimens were stained with haematoxylin and eosin (HE), and α-SMA, SM-MHC, matrix metalloproteinase (MMP)-2 and MMP-9 were detected with immunohistochemistry staining. α-SMA, SM-MHC, MMP-2 and MMP-9 were conducted with western blot. vSMCs taken from the descending aorta of both of the CG and AG were separated and cultured until Passage 3. 3-(4,5-Dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) method were used to analyse cell proliferation. Western blot was used to evaluate MMP-2, MMP-9 expression and flow cytometry was employed to assess cell apoptosis. RESULTS Vascular ultrasound showed obvious dilatation of soaked descending aorta. HE staining showed thickening of thoracic aorta and disarrangement of cells after soaking with PPE. Immunohistochemistry staining showed high expression of MMP-2 and MMP-9 but low expression of SM-MHC and α-SMA in the AG. Tissue western blot analysis of the AG showed that the protein gray value was high in MMP-2 and MMP-9, but low in α-SMA and SM-MHC, which had statistical differences compared with CG with a P-value of <0.05. MTT analysis showed vSMC proliferation activity was higher in the AG than in the CG. Flow cytometry analysis revealed that cell apoptosis between the control and aneurysm groups had significant statistical differences. CONCLUSIONS There is vSMC phenotypic switching in animal models as seen through the rat thoracic aortic aneurysms. This may play an important role in the formation of aneurysms. Our findings are relevant to human aneurysms and may be conducive in the research of aortic aneurysm pathology and treatment.
Collapse
Affiliation(s)
- Naihui Mao
- Department of Cardiac Surgery, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Tianxiang Gu
- Department of Cardiac Surgery, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Enyi Shi
- Department of Cardiac Surgery, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Guangwei Zhang
- Department of Cardiac Surgery, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Lei Yu
- Department of Cardiac Surgery, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Chun Wang
- Department of Cardiac Surgery, The First Affiliated Hospital of China Medical University, Shenyang, China
| |
Collapse
|
54
|
van Dijk RA, Duinisveld AJF, Schaapherder AF, Mulder-Stapel A, Hamming JF, Kuiper J, de Boer OJ, van der Wal AC, Kolodgie FD, Virmani R, Lindeman JHN. A change in inflammatory footprint precedes plaque instability: a systematic evaluation of cellular aspects of the adaptive immune response in human atherosclerosis. J Am Heart Assoc 2015; 4:jah3876. [PMID: 25814626 PMCID: PMC4579929 DOI: 10.1161/jaha.114.001403] [Citation(s) in RCA: 74] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
Background Experimental studies characterize adaptive immune response as a critical factor in the progression and complications of atherosclerosis. Yet, it is unclear whether these observations translate to the human situation. This study systematically evaluates cellular components of the adaptive immune response in a biobank of human aortas covering the full spectrum of atherosclerotic disease. Methods and Results A systematic analysis was performed on 114 well‐characterized perirenal aortic specimens with immunostaining for T‐cell subsets (CD3/4/8/45RA/45RO/FoxP3) and the Th1/non‐Th1/Th17 ratio (CD4+T‐bet+/CD4+T‐bet−/CD4+/interleukin‐17+ double staining). CD20 and CD138 were used to identify B cells and plasma cells, while B‐cell maturation was evaluated by AID/CD21 staining and expression of lymphoid homeostatic CXCL13. Scattered CD4 and CD8 cells with a T memory subtype were found in normal aorta and early, nonprogressive lesions. The total number of T cells increases in progressive atherosclerotic lesions (≈1:5 CD4/CD8 T‐cell ratio). A further increase in medial and adventitial T cells is found upon progression to vulnerable lesions. This critical stage is further hallmarked by de novo formation of adventitial lymphoidlike structures containing B cells and plasma cells, a process accompanied by transient expression of CXCL13. A dramatic reduction of T‐cell subsets, disappearance of lymphoid structures, and loss of CXCL13 expression characterize postruptured lesions. FoxP3 and Th17 T cells were minimally present throughout the atherosclerotic process. Conclusions Transient CXCL13 expression, restricted presence of B cells in human atherosclerosis, along with formation of nonfunctional extranodal lymphoid structures in the phase preceding plaque rupture, indicates a “critical” change in the inflammatory footprint before and during plaque destabilization.
Collapse
Affiliation(s)
- R A van Dijk
- Department of Vascular Surgery, Leiden University Medical Center, Leiden, The Netherlands (D., D., M.S., H., L.)
| | - A J F Duinisveld
- Department of Vascular Surgery, Leiden University Medical Center, Leiden, The Netherlands (D., D., M.S., H., L.)
| | - A F Schaapherder
- Department of Transplantation Surgery, Leiden University Medical Center, Leiden, The Netherlands (S.)
| | - A Mulder-Stapel
- Department of Vascular Surgery, Leiden University Medical Center, Leiden, The Netherlands (D., D., M.S., H., L.)
| | - J F Hamming
- Department of Vascular Surgery, Leiden University Medical Center, Leiden, The Netherlands (D., D., M.S., H., L.)
| | - J Kuiper
- Gorlaeus Laboratories, Division of Biopharmaceutics, Leiden Academic Centre for Drug Research, Leiden, The Netherlands (K.)
| | - O J de Boer
- Department of Pathology, Academic Medical Center, Amsterdam, The Netherlands (B., W.)
| | - A C van der Wal
- Department of Pathology, Academic Medical Center, Amsterdam, The Netherlands (B., W.)
| | - F D Kolodgie
- CVPath Institute Inc., Gaithersburg, MD (K., V.)
| | - R Virmani
- CVPath Institute Inc., Gaithersburg, MD (K., V.)
| | - J H N Lindeman
- Department of Vascular Surgery, Leiden University Medical Center, Leiden, The Netherlands (D., D., M.S., H., L.)
| |
Collapse
|
55
|
Cifani N, Proietta M, Tritapepe L, Di Gioia C, Ferri L, Taurino M, Del Porto F. Stanford-A acute aortic dissection, inflammation, and metalloproteinases: a review. Ann Med 2015; 47:441-6. [PMID: 26339779 DOI: 10.3109/07853890.2015.1073346] [Citation(s) in RCA: 88] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Acute aortic dissection (AAD) is a life-threatening disease with an incidence of about 2.6-3.6 cases per 100,000/year. Depending on the site of rupture, AAD is classified as Stanford-A when the ascending aortic thoracic tract and/or the arch are involved, and Stanford-B when the descending thoracic aorta and/or aortic abdominal tract are targeted. It was recently shown that inflammatory pathways underlie aortic rupture in both type A and type B Stanford AAD. An immune infiltrate has been found within the middle and outer tunics of dissected aortic specimens. It has also been observed that the recall and activation of macrophages inside the middle tunic are key events in the early phases of AAD. Macrophages are able to release metalloproteinases (MMPs) and pro-inflammatory cytokines which, in turn, give rise to matrix degradation and neoangiogenesis. An imbalance between the production of MMPs and MMP tissue inhibitors is pivotal in the extracellular matrix degradation underlying aortic wall remodelling in dissections occurring both in inherited conditions and in atherosclerosis. Among MMPs, MMP-12 is considered a specific marker of aortic wall disease, whatever the genetic predisposition may be. The aim of this review is, therefore, to take a close look at the immune-inflammatory mechanisms underlying Stanford-A AAD.
Collapse
Affiliation(s)
- Noemi Cifani
- a Department of Clinical and Molecular Medicine , Faculty of Medicine and Psychology, Internal Medicine Unit, Sant' Andrea Hospital, Sapienza University of Rome , Rome , Italy.,b Department of Biology and Biotechnology' Charles Darwin' , Sapienza University of Rome , Rome , Italy
| | - Maria Proietta
- a Department of Clinical and Molecular Medicine , Faculty of Medicine and Psychology, Internal Medicine Unit, Sant' Andrea Hospital, Sapienza University of Rome , Rome , Italy
| | - Luigi Tritapepe
- c Department of Anaesthesiology , Critical Medicine and PainTreatment, Faculty of Medicine and Odontology, Policlinico Umberto Primo, Sapienza University of Rome , Rome , Italy
| | - Cira Di Gioia
- d Department of Radiology , Oncology, and Anatomy& Pathology, Faculty of Medicine and Odontology, Policlinico Umberto Primo, Sapienza University of Rome , Rome , Italy
| | - Livia Ferri
- a Department of Clinical and Molecular Medicine , Faculty of Medicine and Psychology, Internal Medicine Unit, Sant' Andrea Hospital, Sapienza University of Rome , Rome , Italy
| | - Maurizio Taurino
- e Department of Clinical and Molecular Medicine,Faculty of Medicine and Psychology , Vascular Surgery Unit, Sant' Andrea Hospital, Sapienza University of Rome , Rome , Italy
| | - Flavia Del Porto
- a Department of Clinical and Molecular Medicine , Faculty of Medicine and Psychology, Internal Medicine Unit, Sant' Andrea Hospital, Sapienza University of Rome , Rome , Italy
| |
Collapse
|
56
|
Kortekaas KE, Meijer CA, Hinnen JW, Dalman RL, Xu B, Hamming JF, Lindeman JH. ACE inhibitors potently reduce vascular inflammation, results of an open proof-of-concept study in the abdominal aortic aneurysm. PLoS One 2014; 9:e111952. [PMID: 25474105 PMCID: PMC4256371 DOI: 10.1371/journal.pone.0111952] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2014] [Accepted: 09/29/2014] [Indexed: 01/11/2023] Open
Abstract
BACKGROUND Independent of their blood pressure lowering effect, ACE inhibitors are thought to reduce vascular inflammation. The clinical relevance of this effect is unclear with the current knowledge. Abdominal aortic aneurysms (AAA) are characterized by a broad, non-specific inflammatory response, and thus provide a clinical platform to evaluate the anti-inflammatory potential of ACE inhibitors. METHODS AND RESULTS Eleven patients scheduled for open AAA repair received ramipril (5 mg/day) during 2-4 weeks preceding surgery. Aortic wall samples were collected during surgery, and compared to matched samples obtained from a biobank. An anti-inflammatory potential was evaluated in a comprehensive analysis that included immunohistochemistry, mRNA and protein analysis. A putative effect of ACE inhibitors on AAA growth was tested separately by comparing 18-month growth rate of patients on ACE inhibitors (n = 82) and those not taking ACE inhibitors (n = 204). Ramipril reduces mRNA expression of multiple pro-inflammatory cytokines such as IL-1β, IL-6, IL-8, TNF -α, Interferon-[Formula: see text], and MCP-1, as well as aortic wall IL-8 and MCP-1 (P = 0.017 and 0.008, respectively) protein content. The is followed by clear effects on cell activation that included a shift towards anti-inflammatory macrophage (M2) subtype. Evaluation of data from the PHAST cohort did not indicate an effect of ACE inhibitors on 18-month aneurysm progression (mean difference at 18 months: -0.24 mm (95% CI: -0.90-0.45, P = NS). CONCLUSIONS ACE inhibition quenches multiple aspects of vascular inflammation in AAA. However, this does not translate into reduced aneurysm growth. TRIAL REGISTRATION Nederlands Trial Register 1345.
Collapse
Affiliation(s)
- Kim E. Kortekaas
- Department of Vascular Surgery, Leiden University Medical Center, Leiden, The Netherlands
- Division of Vascular Surgery, Stanford Medical School, Stanford, California, United States of America
| | - C. Arnoud Meijer
- Department of Vascular Surgery, Leiden University Medical Center, Leiden, The Netherlands
| | - Jan Willem Hinnen
- Department of Vascular Surgery, Leiden University Medical Center, Leiden, The Netherlands
- Department of Vascular Surgery, Jeroen Bosch Ziekenhuis, ‘s Hertogenbosch, The Netherlands
| | - Ronald L. Dalman
- Division of Vascular Surgery, Stanford Medical School, Stanford, California, United States of America
| | - Baohui Xu
- Division of Vascular Surgery, Stanford Medical School, Stanford, California, United States of America
| | - Jaap F. Hamming
- Department of Vascular Surgery, Leiden University Medical Center, Leiden, The Netherlands
| | - Jan H. Lindeman
- Department of Vascular Surgery, Leiden University Medical Center, Leiden, The Netherlands
- * E-mail:
| |
Collapse
|
57
|
Stather PW, Sidloff DA, Dattani N, Gokani VJ, Choke E, Sayers RD, Bown MJ. Meta-analysis and meta-regression analysis of biomarkers for abdominal aortic aneurysm. Br J Surg 2014; 101:1358-72. [DOI: 10.1002/bjs.9593] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2014] [Revised: 03/05/2014] [Accepted: 05/19/2014] [Indexed: 12/30/2022]
Abstract
Abstract
Background
Many studies have investigated the systemic and local expression of biomarkers in patients with abdominal aortic aneurysm (AAA). The natural history of AAA varies between patients, and predictors of the presence and diameter of AAA have not been determined consistently. The aim of this study was to perform a systematic review, meta-analysis and meta-regression of studies comparing biomarkers in patients with and without AAA, with the aim of summarizing the association of identified markers with both AAA presence and size.
Methods and results
Literature review identified 106 studies suitable for inclusion. Meta-analysis demonstrated a significant difference between matrix metalloproteinase (MMP) 9, tissue inhibitor of matrix metalloproteinase 1, interleukin (IL) 6, C-reactive protein (CRP), α1-antitrypsin, triglycerides, lipoprotein(a), apolipoprotein A and high-density lipoprotein in patients with and without AAA. Although meta-analysis was not possible for MMP-2 in aortic tissue, tumour necrosis factor α, osteoprotegerin, osteopontin, interferon γ, intercellular cell adhesion molecule 1 and vascular cell adhesion molecule 1, systematic review suggested an increase in these biomarkers in patients with AAA. Meta-regression analysis identified a significant positive linear correlation between aortic diameter and CRP level.
Conclusion
A wide variety of biomarkers are dysregulated in patients with AAA, but their clinical value is yet to be established. Future research should focus on the most relevant biomarkers of AAA, and how they could be used clinically.
Collapse
Affiliation(s)
- P W Stather
- Department of Cardiovascular Sciences, University of Leicester, Leicester, UK
| | - D A Sidloff
- Department of Cardiovascular Sciences, University of Leicester, Leicester, UK
| | - N Dattani
- Department of Cardiovascular Sciences, University of Leicester, Leicester, UK
| | - V J Gokani
- Department of Cardiovascular Sciences, University of Leicester, Leicester, UK
| | - E Choke
- Department of Cardiovascular Sciences, University of Leicester, Leicester, UK
| | - R D Sayers
- Department of Cardiovascular Sciences, University of Leicester, Leicester, UK
| | - M J Bown
- Department of Cardiovascular Sciences, University of Leicester, Leicester, UK
- National Institute for Health Research Leicester Biomedical Research Unit, University of Leicester, Leicester, UK
| |
Collapse
|
58
|
Suppression of abdominal aortic aneurysm formation by inhibition of prolyl hydroxylase domain protein through attenuation of inflammation and extracellular matrix disruption. Clin Sci (Lond) 2014; 126:671-8. [PMID: 24256203 DOI: 10.1042/cs20130435] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
In the present study we sought to determine the effect of CoCl2, an inhibitor of PHD (prolyl hydroxylase domain protein), on the development of AAA (abdominal aortic aneurysm). AAA was induced in C57BL/6 mice by periaortic application of CaCl2 (AAA group). NaCl (0.9%)-treated mice were used as a sham control (SHAM group). Mice were treated with 0.05% CoCl2 in the drinking water (AAA/CoCl2 group). At 1 and 6 weeks after the operation, aortic tissue was excised for further examination. After 6 weeks of CaCl2 treatment, aortic diameter and macrophage infiltration into the aortic adventitia were increased in the AAA group compared with the SHAM group. Treatment with CoCl2 reduced the aneurysmal size and macrophage infiltration compared with the AAA group. Aortic expression of inflammatory cytokines and MCP-1 (monocyte chemoattractant protein-1) and the activities of MMP-9 (matrix metalloproteinase-9) and MMP-2 were enhanced in the AAA group and attenuated in the AAA/CoCl2 group. Expression of cytokines and the activities of MMPs were already increased after 1 week of CaCl2 treatment, but were suppressed by CoCl2 treatment in association with reduced NF-κB (nuclear factor κB) phosphorylation. Treatment with CoCl2 in mice prevented the development of CaCl2-induced AAA in association with reduced inflammation and ECM (extracellular matrix) disruption. The results of the present study suggest that PHD plays a critical role in the development of AAA and that there is a therapeutic potential for PHD inhibitors in the prevention of AAA development.
Collapse
|
59
|
Proietta M, Tritapepe L, Cifani N, Ferri L, Taurino M, Del Porto F. MMP-12 as a new marker of Stanford-A acute aortic dissection. Ann Med 2014; 46:44-8. [PMID: 24432723 DOI: 10.3109/07853890.2013.876728] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
Abstract
BACKGROUND The study evaluated macrophage cytokines and macrophage metalloprotease (MMP)-12 levels in patients with Stanford-A acute aortic dissection (AAD) and in patients with critical carotid artery stenosis (CAS) compared with patients matched for age, sex, and traditional cardiovascular risk factors (RF). The aim was to identify possible early serum markers of risk for atherosclerotic complications. MATERIALS AND METHODS We selected 65 patients: 23 AAD patients, 21 CAS patients, 21 RF, and 10 healthy subjects (HS). In each patient and control serum, levels of interleukin (IL)-6, IL-8, tumour necrosis factor (TNF)-α, monocyte chemoattractant protein (MCP)-1, vascular endothelial growth factor (VEGF), and MMP-12 were assessed by ELISA. RESULTS A significant increase of MMP-12, IL-6, and IL-8 levels in AAD versus CAS was found. Moreover, MMP-12 was shown to be significantly higher in AAD versus RF, but not in CAS versus RF. A significant increase of IL-6, IL-8, MCP-1, TNF-α, and VEGF levels was observed both in AAD and CAS versus RF. CONCLUSIONS The results suggest that MMP-12 may be considered to be a specific marker of Stanford-A AAD. Furthermore, the study confirmed that in AAD and CAS macrophage cytokines play a key role in the progression of the atherosclerotic disease towards complications.
Collapse
Affiliation(s)
- Maria Proietta
- Dipartimento di Medicina Clinica e Molecolare, Facoltà di Medicina e Psicologia, Ospedale Sant'Andrea, 'Sapienza', Università di Roma , Italia , and UOS Aterosclerosi e Dislipidemia, 'Sapienza', Università di Roma , Italy
| | | | | | | | | | | |
Collapse
|
60
|
Lijkwan MA, Hellingman AA, Bos EJ, van der Bogt KEA, Huang M, Kooreman NG, de Vries MR, Peters HAB, Robbins RC, Hamming JF, Quax PHA, Wu JC. Short hairpin RNA gene silencing of prolyl hydroxylase-2 with a minicircle vector improves neovascularization of hindlimb ischemia. Hum Gene Ther 2014; 25:41-9. [PMID: 24090375 DOI: 10.1089/hum.2013.110] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
In this study, we target the hypoxia inducible factor-1 alpha (HIF-1-alpha) pathway by short hairpin RNA interference therapy targeting prolyl hydroxylase-2 (shPHD2). We use the minicircle (MC) vector technology as an alternative for conventional nonviral plasmid (PL) vectors in order to improve neovascularization after unilateral hindlimb ischemia in a murine model. Gene expression and transfection efficiency of MC and PL, both in vitro and in vivo, were assessed using bioluminescence imaging (BLI) and firefly luciferase (Luc) reporter gene. C57Bl6 mice underwent unilateral electrocoagulation of the femoral artery and gastrocnemic muscle injection with MC-shPHD2, PL-shPHD2, or phosphate-buffered saline (PBS) as control. Blood flow recovery was monitored using laser Doppler perfusion imaging, and collaterals were visualized by immunohistochemistry and angiography. MC-Luc showed a 4.6-fold higher in vitro BLI signal compared with PL-Luc. BLI signals in vivo were 4.3×10(5)±3.3×10(5) (MC-Luc) versus 0.4×10(5)±0.3×10(5) (PL-Luc) at day 28 (p=0.016). Compared with PL-shPHD2 or PBS, MC-shPHD2 significantly improved blood flow recovery, up to 50% from day 3 until day 14 after ischemia induction. MC-shPHD2 significantly increased collateral density and capillary density, as monitored by alpha-smooth muscle actin expression and CD31(+) expression, respectively. Angiography data confirmed the histological findings. Significant downregulation of PHD2 mRNA levels by MC-shPHD2 was confirmed by quantitative polymerase chain reaction. Finally, Western blot analysis confirmed significantly higher levels of HIF-1-alpha protein by MC-shPHD2, compared with PL-shPHD2 and PBS. This study provides initial evidence of a new potential therapeutic approach for peripheral artery disease. The combination of HIF-1-alpha pathway targeting by shPHD2 with the robust nonviral MC plasmid improved postischemic neovascularization, making this approach a promising potential treatment option for critical limb ischemia.
Collapse
Affiliation(s)
- Maarten A Lijkwan
- 1 Department of Medicine and Radiology, Stanford University School of Medicine , Stanford, CA 94305
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
61
|
Harrison SC, Smith AJ, Jones GT, Swerdlow DI, Rampuri R, Bown MJ, on behalf of the Aneurysm Consortium, Folkersen L, Baas AF, de Borst GJ, Blankensteijn JD, Price JF, van der Graaf Y, McLachlan S, Agu O, Hofman A, Uitterlinden AG, Franco-Cereceda A, Ruigrok YM, van't Hof F, Powell JT, van Rij AM, Casas JP, Eriksson P, Holmes MV, Asselbergs FW, Hingorani AD, Humphries SE. Interleukin-6 receptor pathways in abdominal aortic aneurysm. Eur Heart J 2013; 34:3707-16. [PMID: 23111417 PMCID: PMC3869968 DOI: 10.1093/eurheartj/ehs354] [Citation(s) in RCA: 133] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/03/2012] [Revised: 08/24/2012] [Accepted: 09/10/2012] [Indexed: 11/25/2022] Open
Abstract
METHODS We conducted a systematic review and meta-analysis of studies reporting circulating IL-6 in AAA, and new investigations of the association between a common non-synonymous functional variant (Asp358Ala) in the IL-6R gene (IL6R) and AAA, followed the analysis of the variant both in vitro and in vivo. Inflammation may play a role in the development of abdominal aortic aneurysms (AAA). Interleukin-6 (IL-6) signalling through its receptor (IL-6R) is one pathway that could be exploited pharmacologically. We investigated this using a Mendelian randomization approach. RESULTS Up to October 2011, we identified seven studies (869 cases, 851 controls). Meta-analysis demonstrated that AAA cases had higher levels of IL-6 than controls [standardized mean difference (SMD) = 0.46 SD, 95% CI = 0.25-0.66, I(2) = 70%, P = 1.1 × 10-5 random effects]. Meta-analysis of five studies (4524 cases/15 710 controls) demonstrated that rs7529229 (which tags the non-synonymous variant Asp358Ala, rs2228145) was associated with a lower risk of AAA, per Ala358 allele odds ratio 0.84, 95% CI: 0.80-0.89, I(2) = 0%, P = 2.7 × 10-11). In vitro analyses in lymphoblastoid cell lines demonstrated a reduction in the expression of downstream targets (STAT3, MYC and ICAM1) in response to IL-6 stimulation in Ala358 carriers. CONCLUSIONS A Mendelian randomization approach provides robust evidence that signalling via the IL-6R is likely to be a causal pathway in AAA. Drugs that inhibit IL-6R may play a role in AAA management.
Collapse
Affiliation(s)
- Seamus C. Harrison
- Department of Cardiovascular Genetics,Institute of Cardiovascular Science, University College London, Rayne Building, University Street, London WC1E 6JJ, UK
- BHF Laboratories, Centre for Cardiovascular Genetics, Institute of Cardiovascular Science, University College London (UCL), The Rayne Building, 5 University Street, London WC1E 6JF, UK
| | - Andrew J.P. Smith
- Department of Cardiovascular Genetics,Institute of Cardiovascular Science, University College London, Rayne Building, University Street, London WC1E 6JJ, UK
| | | | | | - Riaz Rampuri
- Department of Cardiovascular Genetics,Institute of Cardiovascular Science, University College London, Rayne Building, University Street, London WC1E 6JJ, UK
| | - Matthew J. Bown
- Department of Cardiovascular Sciences, Leicester University, Leicester LE2 7LX, UK
| | | | - Lasse Folkersen
- Atherosclerosis Research Unit, Department of Medicine Solna, Karolinska Institute, Stockholm 171 76, Sweden
| | - Annette F. Baas
- Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht 3584CG, The Netherlands
| | - Gert Jan de Borst
- Vascular Surgery, UMC Utrecht, PO Box 85500, G04.129, Utrecht 3508GA, The Netherlands
| | - Jan D. Blankensteijn
- Department of Surgery, Vascular Surgery, VU Medical Center, PO Box 7057 ZH F 018, Amsterdam 1007, The Netherlands
| | - Jacqueline F. Price
- Wolfson Unit for Prevention of Peripheral Vascular Diseases, University of Edinburgh, Teviot Place, Edinburgh EH8 9AG, UK
| | - Yolanda van der Graaf
- Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht 3584CG, The Netherlands
| | - Stela McLachlan
- Wolfson Unit for Prevention of Peripheral Vascular Diseases, University of Edinburgh, Teviot Place, Edinburgh EH8 9AG, UK
| | - Obi Agu
- Vascular Surgery, University College London Hospital, London NW1 2BU, UK
| | - Albert Hofman
- Department of Epidemiology, Erasmus Medical Center, Rotterdam 3000CA, The Netherlands
| | - Andre G. Uitterlinden
- Department of Epidemiology, Erasmus Medical Center, Rotterdam 3000CA, The Netherlands
| | | | - Ynte M. Ruigrok
- Department of Neurology and Neurosurgery, Rudolf Magnus Institute of Neuroscience, University Medical Center Utrecht, Utrecht 3508 GA, The Netherlands
| | - F.N. van't Hof
- Department of Neurology and Neurosurgery, Rudolf Magnus Institute of Neuroscience, University Medical Center Utrecht, Utrecht 3508 GA, The Netherlands
| | - Janet T. Powell
- Vascular Surgery Research Group, Imperial College Charing Cross Hospital, 4th Floor, Fulham Palace Road, London W6 8RF, UK
| | | | - Juan P. Casas
- London School of Hygiene and Tropical Medicine, Keppel Street, London WC1E 7HT, UK
| | - Per Eriksson
- Department of Cardiovascular Sciences, Leicester University, Leicester LE2 7LX, UK
| | - Michael V. Holmes
- Genetic Epidemiology, University College London, London WC1E 6JJ, UK
| | - Folkert W. Asselbergs
- Division Heart and Lungs, Department of Cardiology, University Medical Center Utrecht, Utrecht, The Netherlands
- Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht, The Netherlands
- Department of Medical Genetics, Biomedical Genetics, University Medical Center, Utrecht, The Netherlands
| | | | - Steve E. Humphries
- Department of Cardiovascular Genetics,Institute of Cardiovascular Science, University College London, Rayne Building, University Street, London WC1E 6JJ, UK
| |
Collapse
|
62
|
Qin Y, Yang Y, Liu R, Cao X, Liu O, Liu J, Wang M, Yang Y, Chen Z, Zhang H, Du J. Combined Cathepsin S and hs-CRP predicting inflammation of Abdominal Aortic Aneurysm. Clin Biochem 2013; 46:1026-1029. [DOI: 10.1016/j.clinbiochem.2013.05.065] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2012] [Revised: 05/18/2013] [Accepted: 05/20/2013] [Indexed: 01/11/2023]
|
63
|
Tan CK, Tan EH, Luo B, Huang CL, Loo JS, Choong C, Tan NS. SMAD3 deficiency promotes inflammatory aortic aneurysms in angiotensin II-infused mice via activation of iNOS. J Am Heart Assoc 2013; 2:e000269. [PMID: 23782924 PMCID: PMC3698794 DOI: 10.1161/jaha.113.000269] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Background Ninety percent of the patients carrying distinct SMAD3 mutations develop aortic aneurysms and dissections, called aneurysms‐osteoarthritis syndrome (AOS). However, the etiology and molecular events downstream of SMAD3 leading to the pathogenesis of aortic aneurysms in these patients still remain elusive. Therefore, we aimed to investigate the vascular phenotypes of SMAD3‐knockout mice. Methods and Results We have shown that angiotensin II–induced vascular inflammation, but not hypertension, leads to aortic aneurysms and dissections, ultimately causing aortic rupture and death in mice. Lipopolysaccharide‐triggered inflammation confirmed that enhanced aortic macrophage recruitment was essential for aneurysm formation in angiotensin II–infused SMAD3‐knockout mice. In contrast, phenylephrine‐triggered hypertension alone was insufficient to induce aortic aneurysms in mice. Using uniaxial tensile and contractility tests, we showed that SMAD3 deficiency resulted in defective aortic biomechanics and physiological functions, which caused weakening of the aortic wall and predisposed the mice to aortic aneurysms. Chromatin immunoprecipitation (ChIP) and re‐ChIP assays revealed that the underlying mechanism involved aberrant upregulation of inducible nitric oxide synthase (iNOS)–derived nitric oxide production and activation of elastolytic matrix metalloproteinases 2 and 9. Administration of clodronate‐liposomes and iNOS inhibitor completely abrogated these aortic conditions, thereby identifying iNOS‐mediated nitric oxide secretion from macrophages as the downstream event of SMAD3 that drives this severe pathology. Conclusions Macrophage depletion and iNOS antagonism represent 2 promising approaches for preventing aortic aneurysms related to SMAD3 mutations and merit further investigation as adjunctive strategies for the life‐threatening manifestations of AOS.
Collapse
Affiliation(s)
- Chek K Tan
- School of Biological Sciences, Nanyang Technological University, Nanyang, Singapore
| | | | | | | | | | | | | |
Collapse
|
64
|
van der Meij E, Koning GG, Vriens PW, Peeters MF, Meijer CA, Kortekaas KE, Dalman RL, van Bockel JH, Hanemaaijer R, Kooistra T, Kleemann R, Lindeman JHN. A clinical evaluation of statin pleiotropy: statins selectively and dose-dependently reduce vascular inflammation. PLoS One 2013; 8:e53882. [PMID: 23349755 PMCID: PMC3551939 DOI: 10.1371/journal.pone.0053882] [Citation(s) in RCA: 87] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2012] [Accepted: 12/04/2012] [Indexed: 11/25/2022] Open
Abstract
Statins are thought to reduce vascular inflammation through lipid independent mechanisms. Evaluation of such an effect in atherosclerotic disease is complicated by simultaneous effects on lipid metabolism. Abdominal aortic aneurysms (AAA) are part of the atherosclerotic spectrum of diseases. Unlike atherosclerotic occlusive disease, AAA is not lipid driven, thus allowing direct evaluation of putative anti-inflammatory effects. The anti-inflammatory potency of increasing doses (0, 20 or 40 mg/day) simvastatin or atorvastatin was evaluated in 63 patients that were at least 6 weeks on statin therapy and who underwent open AAA repair. A comprehensive analysis using immunohistochemistry, mRNA and protein analyses was applied on aortic wall samples collected during surgery. The effect of statins on AAA growth was analyzed in a separate prospective study in incorporating 142 patients. Both statins equally effectively and dose-dependently reduced aortic wall expression of NFκB regulated mediators (i.e. IL-6 (P<0.001) and MCP-1 (P<0.001)); shifted macrophage polarization towards a M2 phenotype (P<0.0003); selectively reduced macrophage-related markers such as cathepsin K and S (P<0.009 and 0.0027 respectively), and ALOX5 (P<0.0009), and reduced vascular wall NFκB activity (40 mg/day group, P<0.016). No effect was found on other cell types. Evaluation of the clinical efficacy of statins to reduce AAA progression did not indicate an effect of statins on aneurysm growth (P<0.337). Hence, in the context of AAA the clinical relevance of statins pleiotropy appears minimal.
Collapse
Affiliation(s)
- Evelien van der Meij
- Department of Vascular Surgery, Leiden University Medical Center, Leiden, The Netherlands
| | - Giel G. Koning
- Department of Surgery, St. Elisabeth Hospital, Tilburg, The Netherlands
| | - Patrick W. Vriens
- Department of Surgery, St. Elisabeth Hospital, Tilburg, The Netherlands
| | - Marcel F. Peeters
- Laboratory for Medical Microbiology, St. Elisabeth Hospital, Tilburg, The Netherlands
| | - C. Arnoud Meijer
- Department of Vascular Surgery, Leiden University Medical Center, Leiden, The Netherlands
| | - Kim E. Kortekaas
- Department of Vascular Surgery, Leiden University Medical Center, Leiden, The Netherlands
- Stanford Medical School, Stanford, California, United States of America
| | - Ronald L. Dalman
- Stanford Medical School, Stanford, California, United States of America
| | - J. Hajo van Bockel
- Department of Vascular Surgery, Leiden University Medical Center, Leiden, The Netherlands
| | - Roeland Hanemaaijer
- Department of Vascular and Metabolic Diseases, TNO (Toegepast Natuurwetenschappelijk Onderzoek) -Quality of Life, Leiden, The Netherlands
| | - Teake Kooistra
- Department of Vascular and Metabolic Diseases, TNO (Toegepast Natuurwetenschappelijk Onderzoek) -Quality of Life, Leiden, The Netherlands
| | - Robert Kleemann
- Department of Vascular and Metabolic Diseases, TNO (Toegepast Natuurwetenschappelijk Onderzoek) -Quality of Life, Leiden, The Netherlands
| | - Jan H. N. Lindeman
- Department of Vascular Surgery, Leiden University Medical Center, Leiden, The Netherlands
- * E-mail:
| |
Collapse
|
65
|
Yamanouchi D, Takei Y, Komori K. Balanced mineralization in the arterial system: possible role of osteoclastogenesis/osteoblastogenesis in abdominal aortic aneurysm and stenotic disease. Circ J 2012; 76:2732-7. [PMID: 23117745 DOI: 10.1253/circj.cj-12-1240] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Arterial calcification is the result of the same highly organized processes as seen in bone, which rely on a delicate balance between osteoblasts and osteoclasts. Although previously understood as passive precipitation, evidence has accumulated to suggest that arterial calcification is the result of organized, regulated processes bearing many similarities to osteogenesis in bone, including the presence of subpopulations of arterial wall cells that retain osteoblastic lineage potential. These cells have the potential to form mineralized nodules and express osteoblast markers, including bone morphogenetic protein-2, osteocalcin, osteopontin, and alkaline phosphatase. By contrast, osteoclast-like cells mediate the catabolic process of mineral resorption. Recent data shows that cells positive for tartrate-resistant acid phosphatase, a major marker for osteoclasts, have been histologically identified in atherosclerotic lesions and are referred to as osteoclast-like cells. Evidence has accumulated to suggest that initial arterial calcification through passive precipitation of calcium phosphate initiates balanced mineralization regulated by osteoclast-like and osteoblast-like cells. Subsequently, various pathogenic conditions may trigger an imbalance between osteoblastogenesis and osteoclastogenesis, leading to either calcification in stenotic/occlusive disease or destruction of the extracellular matrix in aneurysmal disease. Further elucidation of these newly emerging concepts could lead to a novel therapeutic approach to arterial stenotic/occlusive disease and/or abdominal aortic aneurysm.
Collapse
Affiliation(s)
- Dai Yamanouchi
- Division of Vascular Surgery, Department of Surgery, University of Wisconsin School of Medicine and Public Health, Madison, WI 53792, USA.
| | | | | |
Collapse
|
66
|
Meijer C, Kokje V, van Tongeren R, Hamming J, van Bockel J, Möller G, Lindeman J. An Association between Chronic Obstructive Pulmonary Disease and Abdominal Aortic Aneurysm beyond Smoking. Eur J Vasc Endovasc Surg 2012; 44:153-7. [DOI: 10.1016/j.ejvs.2012.05.016] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2012] [Accepted: 05/16/2012] [Indexed: 10/28/2022]
|
67
|
Meijer CA, Le Haen PAA, van Dijk RA, Hira M, Hamming JF, van Bockel JH, Lindeman JH. Activator protein-1 (AP-1) signalling in human atherosclerosis: results of a systematic evaluation and intervention study. Clin Sci (Lond) 2012; 122:421-8. [PMID: 22092038 PMCID: PMC3259695 DOI: 10.1042/cs20110234] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2011] [Revised: 11/07/2011] [Accepted: 11/17/2011] [Indexed: 01/08/2023]
Abstract
Animal studies implicate the AP-1 (activator protein-1) pro-inflammatory pathway as a promising target in the treatment of atherosclerotic disease. It is, however, unclear whether these observations apply to human atherosclerosis. Therefore we evaluated the profile of AP-1 activation through histological analysis and tested the potential benefit of AP-1 inhibition in a clinical trial. AP-1 activation was quantified by phospho-c-Jun nuclear translocation (immunohistochemistry) on a biobank of aortic wall samples from organ donors. The effect of AP-1 inhibition on vascular parameters was tested through a double blind placebo-controlled cross-over study of 28 days doxycycline or placebo in patients with symptomatic peripheral artery disease. Vascular function was assessed by brachial dilation as well as by plasma samples analysed for hs-CRP (high-sensitivity C-reactive protein), IL-6 (interleukin-6), IL-8, ICAM-1 (intercellular adhesion molecule-1), vWF (von Willebrand factor), MCP-1 (monocyte chemoattractant protein-1), PAI-1 (plasminogen activator inhibitor-1) and fibrinogen. Histological evaluation of human atherosclerosis showed minimal AP-1 activation in non-diseased arterial wall (i.e. vessel wall without any signs of atherosclerotic disease). A gradual increase of AP-1 activation was found in non-progressive and progressive phases of atherosclerosis respectively (P<0.044). No significant difference was found between progressive and vulnerable lesions. The expression of phospho-c-Jun diminished as the lesion stabilized (P<0.016) and does not significantly differ from the normal aortic wall (P<0.33). Evaluation of the doxycycline intervention only revealed a borderline-significant reduction of circulating hs-CRP levels (-0.51 μg/ml, P=0.05) and did not affect any of the other markers of systemic inflammation and vascular function. Our studies do not characterize AP-1 as a therapeutic target for progressive human atherosclerotic disease.
Collapse
Key Words
- activator protein 1 (ap-1)
- atherosclerosis
- clinical trial
- doxycycline
- inflammation
- aaa, abdominal aortic aneurysm
- ap-1, activator protein-1
- cvd, cardiovascular disease
- hdl, high-density lipoprotein
- hs-crp, high-sensitivity c-reactive protein
- icam-1, intercellular adhesion molecule-1
- il, interleukin
- mcp-1, monocyte chemoattractant protein-1
- ntg, nitroglycerine
- pai-1, plasminogen activator inhibitor-1
- pad, peripheral arterial disease
- smc, smooth muscle cell
- vwf, von willebrand factor
Collapse
Affiliation(s)
- C. Arnoud Meijer
- Department of Vascular Surgery, Leiden University Medical Center, Leiden, PO Box 9600, 2300 RC Leiden, The Netherlands
| | - Pum A. A. Le Haen
- Department of Vascular Surgery, Leiden University Medical Center, Leiden, PO Box 9600, 2300 RC Leiden, The Netherlands
| | - Rogier A. van Dijk
- Department of Vascular Surgery, Leiden University Medical Center, Leiden, PO Box 9600, 2300 RC Leiden, The Netherlands
| | - Mitsuhisa Hira
- Department of Vascular Surgery, Leiden University Medical Center, Leiden, PO Box 9600, 2300 RC Leiden, The Netherlands
| | - Jaap F. Hamming
- Department of Vascular Surgery, Leiden University Medical Center, Leiden, PO Box 9600, 2300 RC Leiden, The Netherlands
| | - J. Hajo van Bockel
- Department of Vascular Surgery, Leiden University Medical Center, Leiden, PO Box 9600, 2300 RC Leiden, The Netherlands
| | - Jan H. Lindeman
- Department of Vascular Surgery, Leiden University Medical Center, Leiden, PO Box 9600, 2300 RC Leiden, The Netherlands
| |
Collapse
|
68
|
Pera J, Dziedzic T, Adamski M, Jagiella J, Krupa M, Moskala M, Szczudlik A, Slowik A. Interleukin 6-174G>C polymorphism and risk of aneurysmal subarachnoid hemorrhage: case-control study and meta-analysis. Acta Neurol Scand 2012; 125:111-5. [PMID: 21410438 DOI: 10.1111/j.1600-0404.2011.01505.x] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
OBJECTIVES Vascular inflammation contributes to the pathogenesis of aneurysmal subarachnoid hemorrhage (aSAH). Interleukin 6 (IL6) is a proinflammatory cytokine involved in many vascular pathologies. Two studies analyzing an association of the functional IL6 gene -174G>C promoter polymorphism with aSAH provided inconsistent results. The aim of this study was to investigate whether this IL6 polymorphism is associated with aSAH in a Polish population. MATERIAL AND METHODS We genotyped 276 aSAH patients and 581 unrelated control subjects. All were of Caucasian origin. In addition, a meta-analysis combining results of the current and previously published studies was conducted. RESULTS The distribution of IL6 genotypes and alleles did not differ significantly between aSAH (GG - 29.7%, GC - 50.0%, CC - 20.2%, G - 54.7%) and control subjects (GG - 32.0%, GC - 47.3%, CC - 20.7%, G - 44.3%). In the meta-analysis, the IL6-174G>C polymorphism was not associated with aSAH risk either. CONCLUSIONS We failed to find an association between the IL6 -174G>C polymorphism and aSAH in analyzed European populations.
Collapse
Affiliation(s)
- J Pera
- Department of Neurology, Jagiellonian University Medical College, Krakow, Poland.
| | | | | | | | | | | | | | | |
Collapse
|
69
|
Liao M, Xu J, Clair AJ, Ehrman B, Graham LM, Eagleton MJ. Local and systemic alterations in signal transducers and activators of transcription (STAT) associated with human abdominal aortic aneurysms. J Surg Res 2011; 176:321-8. [PMID: 21764069 DOI: 10.1016/j.jss.2011.05.041] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2011] [Revised: 04/21/2011] [Accepted: 05/24/2011] [Indexed: 11/29/2022]
Abstract
BACKGROUND Signal transducers and activators of transcription (STAT) proteins are transcription factors that, when activated by phosphorylation, regulate gene expression and cellular activity. The aim of this study was to evaluate the local and systemic expression and activation of STAT proteins associated with abdominal aortic aneurysms (AAA). METHODS Expression and activation of STAT proteins were assessed in aortic wall samples obtained from patients undergoing repair of AAA (n = 9) and from non-aneurysmal (NA) donors (n = 17). Aortic samples were evaluated for mRNA and protein expression for STAT1, 2, 3, 4, 5a, and 5b using RT-PCR and immunoblot (WB) assays and normalized to ß-actin (expressed as arbitrary units). STAT activation was assessed with WB assays using phosphorylated (p)-STAT-specific antibodies. Alterations in STAT activation were calculated by normalizing pSTAT proteins to corresponding total STAT levels. Immunohistochemistry was performed on AAA and NA samples using the total and pSTAT antibodies. Systemic alterations in STAT activation were assessed by evaluating circulating leukocytes for the presence of pSTAT from patients with AAA (AAA, n = 8), repaired aneurysm (RA, n = 8), or age/gender matched controls with no AAA (CT, n = 8). Flow cytometry was performed to assess for circulating levels of STAT1 (pY701), STAT3 (pY705), and STAT5a (pY694) in monocytes, granulocytes, and lymphocytes. Assessments were made at baseline and in response to in vitro stimulation with IFN-γ (50 ng/mL) or IL-6 (100 ng/mL). Results were analyzed using Student's t-test and are expressed as mean ± SEM. RESULTS In AAA tissue compared with NA, STAT-1 (1.08 ± 0.09 versus 0.62 ± 0.07), -2 (0.98 ± 0.07 versus 0.55 ± 0.08), and -4 (0.89 ± 0.12 versus 0.35 ± 0.11) mRNA levels were elevated (P < 0.01, all). Corresponding increases in STAT protein were only observed for STAT1 (2.77 ± 0.93 versus 0.93 ± 0.08, P < 0.05). Increases in activation were observed in AAA compared with NA in pSTAT2 (0.77 ± 0.1 versus 0.1 ± 0.02, P < 0.01), pSTAT3 (1.6 ± 0.3 versus 0.2 ± 0.06, P < 0.02) and pSTAT5 (0.57 ± 0.03 versus 0.2 ± 0.03, P < 0.05) levels. Phosphorylated STAT1, 2, 3, and 5 were observed in inflammatory cells invading the AAA adventitia. In addition, STAT3 was observed in the media of AAA and NA, but pSTAT3 was only observed in the media of AAA. There were no differences in baseline levels of pSTAT-positive circulating leukocytes. IFN-γ stimulation decreased STAT-5a (pY694)-positive CT lymphocytes to 40% ± 13% of baseline, but had no effect on AAA or RA lymphocytes (116% ± 35%, 102% ± 19%, respectively; P = 0.01). STAT-5a (pY694)-positive CT granulocytes also decreased to 62% ± 18% of baseline compared with AAA or RA granulocytes (122% ± 25%, 126% ± 17%, respectively; P = 0.01). Alterations in STAT1 (pY701) and STAT3 (pY705) were not observed in leukocytes following cytokine stimulation. CONCLUSIONS STAT proteins are important regulators of transcriptional activity and have been linked to cardiovascular disease. The present data suggest that altered levels of phosphorylated STATs are associated with AAA. Understanding their role may provide further insight into the mechanisms of AAA formation and allow for the development of medical treatment options.
Collapse
Affiliation(s)
- Mingfang Liao
- Department of Vascular Surgery, Cleveland Clinic Lerner College of Medicine-CWRU, Cleveland, Ohio 44195, USA
| | | | | | | | | | | |
Collapse
|
70
|
Zhang Q, Huang JH, Xia RP, Duan XH, Jiang YB, Jiang Q, Sun WJ. Suppression of Experimental Abdominal Aortic Aneurysm in a Rat Model by the Phosphodiesterase 3 Inhibitor Cilostazol. J Surg Res 2011; 167:e385-93. [DOI: 10.1016/j.jss.2011.01.017] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2010] [Revised: 12/16/2010] [Accepted: 01/11/2011] [Indexed: 11/26/2022]
|
71
|
del Porto F, Proietta M, Tritapepe L, Miraldi F, Koverech A, Cardelli P, Tabacco F, de Santis V, Vecchione A, Mitterhofer AP, Nofroni I, Amodeo R, Trappolini M, Aliberti G. Inflammation and immune response in acute aortic dissection. Ann Med 2010; 42:622-9. [PMID: 20860543 DOI: 10.3109/07853890.2010.518156] [Citation(s) in RCA: 135] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
OBJECTIVE The aim of our study was to evaluate the lymphocyte subpopulations and the cytokines in the peripheral blood of patients with type-A Stanford acute aortic dissection (AAD group) and to determine whether inflammatory cells are present at the site of aortic dissection. METHODS Thirty-five consecutive patients with type-A Stanford dissection were evaluated for haemochrome and lymphocyte subpopulations (CD3+, CD4+, CD8+, CD19+, CD4+CD25+, CD16+CD56+, CD4+CD28-, CD8+CD28-) by flow cytometry. C-reactive protein (CRP), tumour necrosis factor (TNF)-α, interleukin (IL)-2, IL-4, IL-6, IL-8, IL-10, interferon-gamma (IFN-γ), and monocyte chemoattractant protein (MCP)-1 were measured by ELISA. In addition, immunohistochemical staining with cell type-specific antibodies was performed to study the inflammatory cells detected inside the aortic wall. RESULTS In the AAD group, a significant increase in natural killer (NK) (P = 0.032), B cells (P = 0.022), and CD8+CD28- (P = 0.045) subpopulations was observed, whereas there was a significant decrease in total T lymphocytes (P = 0.004) and T helper fractions (P = 0.005). Moreover, a significant increase in CRP (P < 0.0001), IL-6 (P < 0.0001), IL-8 (P < 0.0001), IL-10 (P < 0.0001), TNF-α (P < 0.0001), and MCP-1 (P < 0.001) was observed; macrophages represented the main population detected inside the media. CONCLUSIONS Our results strongly support the hypothesis of a pivotal role of innate immunity in type-A Stanford AAD.
Collapse
Affiliation(s)
- Flavia del Porto
- Dipartimento di Medicina Clinica e Molecolare, II Facoltà di Medicina e Chirurgia, Università La Sapienza, Ospedale Sant'Andrea, Via di Grottarossa 1035-1039, Rome, Italy.
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
72
|
Tang EHC, Shvartz E, Shimizu K, Rocha VZ, Zheng C, Fukuda D, Shi GP, Sukhova G, Libby P. Deletion of EP4 on bone marrow-derived cells enhances inflammation and angiotensin II-induced abdominal aortic aneurysm formation. Arterioscler Thromb Vasc Biol 2010; 31:261-9. [PMID: 21088251 DOI: 10.1161/atvbaha.110.216580] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
OBJECTIVE To examine whether a lack of prostaglandin E receptor 4 (EP4) on bone marrow-derived cells would increase local inflammation and enhance the formation of abdominal aortic aneurysm (AAA) in vivo. METHODS AND RESULTS Prostaglandin E(2) (PGE(2)) through activation of EP4, can mute inflammation. Hypercholesterolemic low-density lipoprotein receptor knockout (LDLR(-/-)) mice transplanted with either EP4(+/+) (EP4(+/+)/LDLR(-/-)) or EP4(-/-) (EP4(-/-)/LDLR(-/-)) bone marrow received infusions of angiotensin II to induce AAA. Deficiency of EP4 on bone marrow-derived cells increased the incidence (50% of male EP4(+/+)/LDLR(-/-) mice versus 88.9% of male EP4(-/-)/LDLR(-/-) mice developed AAA; and 22% of female EP4(+/+)/LDLR(-/-) mice versus 83.3% of female EP4(-/-)/LDLR(-/-) mice developed AAA) and severity of AAA, increased monocyte chemoattractant protein-1 (2.72-fold in males and 1.64-fold in females), and enhanced infiltration of macrophages (3.8-fold in males and 2.44-fold in females) and T cells (1.88-fold in males and 1.66-fold in females) into AAA lesions. Lack of EP4 on bone marrow-derived cells augmented elastin fragmentation, increased apoptotic markers, and decreased smooth muscle cell accumulation within AAA lesions. CONCLUSIONS Deficiency of EP4 on bone marrow-derived cells boosted inflammation and AAA formation induced by angiotensin II in hyperlipidemic mice. This study affirms the pathophysiologic importance of PGE(2) signaling through EP4 as an endogenous anti-inflammatory pathway involved in experimental aneurysm formation.
Collapse
Affiliation(s)
- Eva H C Tang
- Division of Cardiovascular Medicine, Brigham and Women's Hospital, Harvard Medical School, 77 Ave. Louis Pasteur, Boston, MA 02115, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
73
|
Saratzis A, Abbas AA, Kiskinis D, Melas N, Saratzis N, Kitas GD. Abdominal aortic aneurysm: a review of the genetic basis. Angiology 2010; 62:18-32. [PMID: 20566578 DOI: 10.1177/0003319710373092] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND Abdominal aortic aneurysm (AAA) is a complex disease with a largely unknown pathophysiological background and a strong genetic component. Various studies have tried to link specific genetic variants with AAA. METHODS Systematic review of the literature (1947-2009). RESULTS A total of 249 studies were identified, 89 of which were eventually deemed relevant to this review. Genetic variants (polymorphisms) in a wide variety of genes, most of which encode proteolytic enzymes and inflammatory molecules, have been associated with AAA development and progression. CONCLUSION The genetic basis of AAA remains unknown, and most results from ''candidate-gene'' association studies are contradictory. Further analyses in appropriately powered studies in large, phenotypically well-characterized populations, including genome-wide association studies, are necessary to elucidate the exact genetic contribution to the pathophysiology of AAA.
Collapse
Affiliation(s)
- Athanasios Saratzis
- Russell's Hall Hospital, Dudley Group of Hospitals NHS Foundation Trust, Dudley, West Midlands, UK.
| | | | | | | | | | | |
Collapse
|
74
|
Barillari G, Iovane A, Bonuglia M, Albonici L, Garofano P, Di Campli E, Falchi M, Condò I, Manzari V, Ensoli B. Fibroblast growth factor-2 transiently activates the p53 oncosuppressor protein in human primary vascular smooth muscle cells: Implications for atherogenesis. Atherosclerosis 2010; 210:400-6. [DOI: 10.1016/j.atherosclerosis.2010.01.006] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/06/2009] [Revised: 12/31/2009] [Accepted: 01/06/2010] [Indexed: 10/19/2022]
|
75
|
Abdul-Hussien H, Hanemaaijer R, Kleemann R, Verhaaren BF, van Bockel JH, Lindeman JH. The pathophysiology of abdominal aortic aneurysm growth: Corresponding and discordant inflammatory and proteolytic processes in abdominal aortic and popliteal artery aneurysms. J Vasc Surg 2010; 51:1479-87. [DOI: 10.1016/j.jvs.2010.01.057] [Citation(s) in RCA: 79] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2009] [Revised: 01/12/2010] [Accepted: 01/19/2010] [Indexed: 01/28/2023]
|
76
|
Golledge J, Clancy P, Moran C, Biros E, Rush C, Walker P, Norman P. The novel association of the chemokine CCL22 with abdominal aortic aneurysm. THE AMERICAN JOURNAL OF PATHOLOGY 2010; 176:2098-106. [PMID: 20348247 DOI: 10.2353/ajpath.2010.090416] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
The aim of this study was to examine aortic biopsies with a cytokine array to identify new cytokines associated with abdominal aortic aneurysm (AAA). We assessed the relative expression of 79 cytokines using antibody-based cytokine arrays in a total of 12 AAA and 12 control aortic biopsies. Based on these findings we validated the findings for one cytokine by examining a further 11 AAA and 11 atherothrombosis biopsies and serum from 1028 men, 315 of whom had an AAA. Three cytokines (interleukins 1B and 8, and Chemokine CC motif ligand 22 [CCL22]) were consistently up-regulated in AAA biopsies. Since CCL22 had not previously been associated with aortic dilatation, we confirmed the upregulation of this cytokine in further tissue biopsies and serum using enzyme-linked immunosorbent assay. Median serum concentrations of CCL22 were greater in men with AAA (0.69 ng/ml) than controls (0.56 ng/ml, P < 0.01). Serum CCL22 was independently associated with both small (OR 1.51, 95% CI 1.21-1.88) and large AAA (OR 1.33, 95% CI 1.08-1.62) after adjusting for other risk factors. The association between CCL22 and AAA was also confirmed using immunohistochemistry. The results presented in this study demonstrate a novel association between CCL22 and AAA as well as illustrate how a protein array can be used to identify novel markers of potential pathogenic and diagnostic significance for AAA.
Collapse
Affiliation(s)
- Jonathan Golledge
- Vascular Biology Unit, Department of Surgery, School of Medicine, James Cook University, Townsville, QLD, Australia.
| | | | | | | | | | | | | |
Collapse
|
77
|
Macrophage migration inhibitory factor: critical role in obesity, insulin resistance, and associated comorbidities. Mediators Inflamm 2010; 2010:610479. [PMID: 20169173 PMCID: PMC2821632 DOI: 10.1155/2010/610479] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2009] [Accepted: 01/07/2010] [Indexed: 12/28/2022] Open
Abstract
Obesity is associated with insulin resistance, disturbed glucose homeostasis, low grade inflammation, and comorbidities such as type 2 diabetes and cardiovascular disease. The cytokine macrophage migration inhibitory factor (MIF) is an ubiquitously expressed protein that plays a crucial role in many inflammatory and autoimmune disorders. Increasing evidence suggests that MIF also controls metabolic and inflammatory processes underlying the development of metabolic pathologies associated with obesity. This is a comprehensive summary of our current knowledge on the role of MIF in obesity and obesity-associated comorbidities, based on human clinical data as well as animal models of disease.
Collapse
|
78
|
Grillari J, Grillari-Voglauer R. Novel modulators of senescence, aging, and longevity: Small non-coding RNAs enter the stage. Exp Gerontol 2010; 45:302-11. [PMID: 20080172 DOI: 10.1016/j.exger.2010.01.007] [Citation(s) in RCA: 80] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2009] [Revised: 12/08/2009] [Accepted: 01/08/2010] [Indexed: 02/06/2023]
Abstract
During the last decade evidence has accumulated that the aging process is driven by limited allocation of energy to somatic maintenance resulting in accumulation of stochastic damage. This damage, affecting molecules, cells, and tissues, is counteracted by genetically programmed repair, the efficiency of which thus importantly determines the life and 'health span' of organisms. Therefore, understanding the regulation of gene expression during cellular and organismal aging as well as upon exposure to various damaging events is important to understand the biology of aging and to positively influence the health span. The recent identification of small non-coding RNAs (ncRNAs), has added an additional layer of complexity to the regulation of gene expression with the classes of endogenous small inhibitory RNAs (siRNAs), PIWI-interacting RNAs (piRNAs), QDE1-interacting RNAs (qiRNAs) and microRNAs (miRNAs). Some of these ncRNAs have not yet been identified in mammalian cells and are dependent on RNA-dependent RNA polymerases. The first mammalian enzyme with such activity has only now emerged and surprisingly consists of the catalytic subunit of telomerase (hTERT) together with RMPR, an alternative RNA component. The so far most studied small non-coding RNAs, miRNAs, however, are now increasingly found to operate in the complex network of cellular aging. Recent findings show that (i) miRNAs are regulated during cellular senescence in vitro, (ii) they contribute to tissue regeneration by regulation of stem cell function, and (iii) at least one miRNA modulates the life span of the model organism C. elegans. Additionally, (iv) they act as inhibitors of proteins mediating the insulin/IGF1 and target of rapamycin (TOR) signalling, both of which are conserved modulators of organism life span. Here we will give an overview on the current status of these topics. Since little is so far known on the functions of small ncRNAs in the context of aging and longevity, the entry of the RNA world into the field of biogerontology certainly holds additional surprises and promises. Even more so, as miRNAs are implicated in many age-associated pathologies, and as RNAi and miRNA based therapeutics are on their way to clinics.
Collapse
Affiliation(s)
- Johannes Grillari
- Department of Biotechnology, University of Natural Resources and Applied Life Sciences Vienna, Austria.
| | | |
Collapse
|
79
|
Abstract
OBJECTIVES We try to clear the relationship between high-sensitive C-reactive protein (hsCRP) release and abdominal aortic aneurysm formation. METHODS AND RESULTS A rabbit abdominal aortic aneurysm model was created by elastase perfusion. At days 10, 20, and 30 after elastase perfusion, mean serum hsCRP levels detected by ELISA increased over 200% over their basal level (n = 11, P < 0.05). Serum hsCRP levels were significantly higher in the aneurysm groups than in the sham controls by day 5 (n = 11, P < 0.05) and were positively correlated with percentage vessel diameter changes in the aneurysm group by day 10 (r = 0.8012, n = 33, P < 0.05). In the aneurysm group, increased serum CRP was derived from the liver in early stages, yet from dilated vessels in the later stages, as shown by immunostaining, western blot, and reverse transcriptase-PCR. Similar increased hsCRP levels were also observed in dissected rabbit aortic ring explants from the aneurysm model. Pretreatment with the stretch-activated channel blockers gadolinium or streptomycin, as well as nuclear factor-kappaB inhibitor SN50, blocked hsCRP production in the dilated aortic rings. Stretch-activated channel blockers also inhibited the activation of nuclear factor-kappaB. CONCLUSION During abdominal aortic aneurysm formation, increased serum hsCRP levels derive from aneurysmal arteries with degenerating elastic lamina. This process is mediated by mechanical stretch-activated channel-dependent nuclear factor-kappaB translocation to the nucleus.
Collapse
|
80
|
|
81
|
de Vries DK, Lindeman JHN, Tsikas D, de Heer E, Roos A, de Fijter JW, Baranski AG, van Pelt J, Schaapherder AFM. Early renal ischemia-reperfusion injury in humans is dominated by IL-6 release from the allograft. Am J Transplant 2009; 9:1574-84. [PMID: 19459788 DOI: 10.1111/j.1600-6143.2009.02675.x] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The pathophysiology of ischemia/reperfusion (I/R) injury is complex, and current knowledge of I/R injury in humans is incomplete. In the present study, human living-donor kidney transplantation was used as a highly reproducible model to systematically study various processes potentially involved in early I/R injury. Unique, direct measurements of arteriovenous concentration differences over the kidney revealed massive release of interleukin (IL)-6 in the first 30 minutes of graft reperfusion and a modest release of IL-8. Among the assessed markers of oxidative and nitrosative stress, only 15(S)-8-iso-PGF(2alpha) was released. When assessing cell activation, release of prothrombin factor 1 + 2 indicated thrombocyte activation, whereas there was no release of markers for endothelial activation or neutrophil activation. Common complement activation complex sC5b-9 was not released into the bloodstream, but was released into urine rapidly after reperfusion. To investigate whether IL-6 plays a modulating role in I/R injury, a mouse experiment of renal I/R injury was performed. Neutralizing anti-IL-6 antibody treatment considerably worsened kidney function. In conclusion, this study shows that renal I/R in humans is dominated by local IL-6 release. Neutralization of IL-6 in mice resulted in a significant aggravation of renal I/R injury.
Collapse
Affiliation(s)
- D K de Vries
- Department of Surgery, Leiden University Medical Center, Leiden, The Netherlands
| | | | | | | | | | | | | | | | | |
Collapse
|
82
|
Huang G, Luo C, Gu X, Wu Z, Wang Z, Du Z, Hu C, Tang L. Mechanical strain induces expression of C-reactive protein in human blood vessels. J Pharmacol Exp Ther 2009; 330:206-11. [PMID: 19398609 DOI: 10.1124/jpet.109.150961] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/07/2025] Open
Abstract
C-reactive protein (CRP) is a powerful independent risk factor for cardiovascular diseases. Elevated mechanical strain on vessels induces the local expression of proinflammatory cytokines. We hypothesized that mechanical strain on vessels may induce local CRP expression. Human saphenous vein and internal mammary artery (IMA) rings were stretched in vitro with a mechanical strength of 1, 3, or 5 g. Reverse transcription-polymerase chain reaction and enzyme-linked immunosorbent assay results showed that mechanical stretching significantly induced CRP mRNA and protein expression in the saphenous vein and IMA rings in a strength-dependent manner reaching a maximum at a mechanical strength of 3 g, but CRP expression returned at strengths of >5 g. In vessels, mechanical strain-induced CRP expression was blocked by two stretch-activated ion channel (SAC) blockers: GdCl(3) and streptomycin. Mechanical strain also increased activation of nuclear factor kappaB (NF-kappaB), which was detected with a nonradioactive NF-kappaB p50/p65 EZ-TFA transcription factor assay. Mechanical strain-induced NF-kappaB activation was blocked by SAC blockers and the NF-kappaB inhibitor (SN50, H-Ala-Ala-Val-Ala-Leu-Leu-Pro-Ala-Val-Leu-Leu-Ala-Leu-Leu-Ala-Pro-Val-Gln-Arg-Lys-Arg-Gln-Lys-Leu-Met-Pro-OH). SN50 also blocked mechanical strain-induced CRP expression in vessels. In conclusion, mechanical strain induces CRP expression in IMAs and saphenous veins by activating the SAC-induced NF-kappaB pathway.
Collapse
Affiliation(s)
- Genya Huang
- Division of Cardiology, The First Affiliated Hospital, Sun Yat-sen University, 58 Zhongshan II Road, Guangzhou, Guangdong, People's Republic of China
| | | | | | | | | | | | | | | |
Collapse
|
83
|
Lindeman JHN, Abdul-Hussien H, van Bockel JH, Wolterbeek R, Kleemann R. Clinical trial of doxycycline for matrix metalloproteinase-9 inhibition in patients with an abdominal aneurysm: doxycycline selectively depletes aortic wall neutrophils and cytotoxic T cells. Circulation 2009; 119:2209-16. [PMID: 19364980 DOI: 10.1161/circulationaha.108.806505] [Citation(s) in RCA: 186] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND Doxycycline has been shown to effectively inhibit aneurysm formation in animal models of abdominal aortic aneurysm. Although this effect is ascribed to matrix metalloproteinase-9 inhibition, such an effect is unclear in human studies. We reevaluated the effect of doxycycline on aortic wall protease content in a clinical trial and found that doxycycline selectively reduces neutrophil-derived proteases. We thus hypothesized that doxycycline acts through an effect on vascular inflammation. METHODS AND RESULTS Sixty patients scheduled for elective open aneurysmal repair were randomly assigned to 2 weeks of low-, medium-, or high-dose doxycycline (50, 100, or 300 mg/d, respectively) or no medication (control group). Aortic wall samples were collected at the time of operation, and the effect of doxycycline treatment on vascular inflammation was evaluated. Independently of its dose, doxycycline treatment resulted in a profound but selective suppression of aortic wall inflammation as reflected by a selective 72% reduction of the aortic wall neutrophils and a 95% reduction of the aortic wall cytotoxic T-cell content (median values; P<0.00003). Evaluation of major inflammatory pathways suggested that doxycycline treatment specifically quenched AP-1 and C/EBP proinflammatory transcription pathways (P<0.0158, NS) and reduced vascular interleukin-6 (P<0.00115), interleukin-8 (P<0.00246, NS), interleukin-13 (P<0.0184, NS), and granulocyte colony-stimulating factor (P<0.031, NS) protein levels. Doxycycline was well tolerated; there were no adverse effects. CONCLUSIONS A brief period of doxycycline treatment has a profound but selective effect on vascular inflammation and reduces aortic wall neutrophil and cytotoxic T-cell content. Results of this study are relevant for pharmaceutical stabilization of the abdominal aneurysm and possibly for other inflammatory conditions that involve neutrophils and/or cytotoxic T cells.
Collapse
Affiliation(s)
- Jan H N Lindeman
- Department of Vascular Surgery, K6R, Leiden University Medical Center, PO Box 9600, 2300 RC Leiden, the Netherlands.
| | | | | | | | | |
Collapse
|
84
|
Abdul-Hussien H, Hanemaaijer R, Verheijen JH, van Bockel JH, Geelkerken RH, Lindeman JHN. Doxycycline therapy for abdominal aneurysm: Improved proteolytic balance through reduced neutrophil content. J Vasc Surg 2009; 49:741-9. [PMID: 19268776 DOI: 10.1016/j.jvs.2008.09.055] [Citation(s) in RCA: 86] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2008] [Revised: 09/17/2008] [Accepted: 09/27/2008] [Indexed: 10/21/2022]
Abstract
BACKGROUND Matrix metalloproteinase-9 (MMP-9) is thought to play a central role in abdominal aortic aneurysm (AAA) initiation. Doxycycline, a tetracycline analogue, has direct MMP-9-inhibiting properties in vitro, and it effectively suppresses AAA development in rodents. Observed inhibition of AAA progression, and contradictory findings in human studies evaluating the effect of doxycycline therapy on aortic wall MMP-9, suggest that the effects of doxycycline extend beyond MMP-9 inhibition and that the effect may be dose-dependent. METHODS This clinical trial evaluated the effect of 2 weeks of low- (50 mg/d), medium- (100 mg/d), or high-dose (300 mg/d) doxycycline vs no medication in four groups of 15 patients undergoing elective AAA repair. The effect of doxycycline treatment on MMP and cysteine proteases, and their respective inhibitors, was evaluated by quantitative polymerase chain reaction, Western blot analysis, immunocapture protease activity assays, and immunohistochemistry. RESULTS Doxycycline was well tolerated and no participants dropped out. Doxycycline treatment reduced aortic wall MMP-3 and MMP-25 messenger RNA expression (P < .045 and P < .014, respectively), selectively suppressed neutrophil collagenase and gelatinase (MMP-8 and MMP-9) protein levels (P < .013 and <.004, respectively), and increased protein levels of the protease inhibitors tissue inhibitor of metalloproteinase 1 and cystatin C (P < .029). As for the apparent selective effect on neutrophil-associated proteases, we sought for a reducing effect on aortic wall neutrophil content that was indeed confirmed by immunohistochemical analysis that revealed a 75% reduction in aneurysm wall neutrophil content (P < .001). CONCLUSIONS Independent of its dose, short-term preoperative doxycycline therapy improves the proteolytic balance in AAA, presumably through an effect on aortic wall neutrophil content. This study provides a rationale for doxycycline treatment in patients with an AAA as well as in other (vascular) conditions involving neutrophil influx such as Kawasaki disease and Behçet disease.
Collapse
Affiliation(s)
- Hazem Abdul-Hussien
- Department of Vascular Surgery, Leiden University Medical Center, Leiden, The Netherlands
| | | | | | | | | | | |
Collapse
|
85
|
IL-32-dependent effects of IL-1beta on endothelial cell functions. Proc Natl Acad Sci U S A 2009; 106:3883-8. [PMID: 19228941 DOI: 10.1073/pnas.0813334106] [Citation(s) in RCA: 111] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
Increasing evidence demonstrates that interleukin (IL)-32 is a pro-inflammatory cytokine, inducing IL-1alpha, IL-1beta, IL-6, tumor necrosis factor (TNF)-alpha, and chemokines via nuclear factor (NF)-kappaB, p38 mitogen-activated protein kinase (MAPK), and activating protein (AP)-1 activation. Here we report that IL-32 is expressed and is also functional in human vascular endothelial cells (EC) of various origins. Compared with primary blood monocytes, high levels of IL-32 are constitutively produced in human umbilical vein EC (HUVEC), aortic macrovascular EC, and cardiac as well as pulmonary microvascular EC. At concentrations as low as 0.1 ng/ml, IL-1beta stimulated IL-32 up to 15-fold over constitutive levels, whereas 10 ng/ml of TNFalpha or 100 ng/ml of lipopolysaccharide (LPS) were required to induce similar quantities of IL-32. IL-1beta-induced IL-32 was reduced by inhibition of the IkappaB kinase-beta/NF-kappaB and ERK pathways. In addition to IL-1beta, pro-coagulant concentrations of thrombin or fresh platelets increased IL-32 protein up to 6-fold. IL-1beta and thrombin induced an isoform-switch in steady-state mRNA levels from IL-32alpha/gamma to beta/epsilon. Adult EC responded in a similar fashion. To prove functionality, we silenced endogenous IL-32 with siRNA, decreasing intracellular IL-32 protein levels by 86%. The knockdown of IL-32 resulted in reduction of constitutive as well as IL-1beta-induced intercellular adhesion molecule-1 (ICAM-1) (of 55% and 54%, respectively), IL-1alpha (of 62% and 43%), IL-6 (of 53% and 43%), and IL-8 (of 46% and 42%). In contrast, the anti-inflammatory/anti-coagulant CD141/thrombomodulin increased markedly when IL-32 was silenced. This study introduces IL-32 as a critical regulator of endothelial function, expanding the properties of this cytokine relevant to coagulation, endothelial inflammation, and atherosclerosis.
Collapse
|