1
|
The mechanism and therapy of aortic aneurysms. Signal Transduct Target Ther 2023; 8:55. [PMID: 36737432 PMCID: PMC9898314 DOI: 10.1038/s41392-023-01325-7] [Citation(s) in RCA: 53] [Impact Index Per Article: 26.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2022] [Revised: 12/15/2022] [Accepted: 01/14/2023] [Indexed: 02/05/2023] Open
Abstract
Aortic aneurysm is a chronic aortic disease affected by many factors. Although it is generally asymptomatic, it poses a significant threat to human life due to a high risk of rupture. Because of its strong concealment, it is difficult to diagnose the disease in the early stage. At present, there are no effective drugs for the treatment of aneurysms. Surgical intervention and endovascular treatment are the only therapies. Although current studies have discovered that inflammatory responses as well as the production and activation of various proteases promote aortic aneurysm, the specific mechanisms remain unclear. Researchers are further exploring the pathogenesis of aneurysms to find new targets for diagnosis and treatment. To better understand aortic aneurysm, this review elaborates on the discovery history of aortic aneurysm, main classification and clinical manifestations, related molecular mechanisms, clinical cohort studies and animal models, with the ultimate goal of providing insights into the treatment of this devastating disease. The underlying problem with aneurysm disease is weakening of the aortic wall, leading to progressive dilation. If not treated in time, the aortic aneurysm eventually ruptures. An aortic aneurysm is a local enlargement of an artery caused by a weakening of the aortic wall. The disease is usually asymptomatic but leads to high mortality due to the risk of artery rupture.
Collapse
|
2
|
Wei R, Chang X, Wu Z, Duan C, Xiong J, Guo W. A novel abdominal aortic aneurysm model produced by periarterial application of hydrochloric acid. Exp Anim 2023; 72:38-46. [PMID: 36058844 PMCID: PMC9978129 DOI: 10.1538/expanim.22-0020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Abstract
Previous abdominal aortic aneurysm (AAA) animal modeling methodologies were either expensive or complicated. Here, we developed a novel AAA model which was simple to set up and generated minimal calcification. Twenty-four rats were divided randomly into four groups. Groups 1, 2 and 3 underwent surgery in which 15% hydrochloric acid (HCl) was applied periarterially to the abdominal aorta for 5 min, followed by sacrifice 1 week (group 1), 2 weeks (group 2), and 4 weeks (group 3) after surgery. The maximum aortic diameter (MAD) was measured at surgery and before animal sacrifice. Rats in group 4 were sham-treated. The MADs in group 1, 2 and 3 showed significant dilation compared with group 4, with a mean dilation rate of 33.8% in the first week after surgery. Histopathological examination revealed infiltration of macrophages into the adventitia, obvious apoptosis of smooth muscle cells, and rupture and collapse of the elastic fibers. Furthermore, no calcification was observed in the dilated aorta. The mRNA expression levels of inflammatory factors were at least two-fold higher in group 1 than in group 4, indicating significant inflammatory response in the progression of AAA information. In conclusion, periarterial application of 15% HCl is a convenient and reliable model to create an abdominal aortic aneurysm in rats, and the potential development mechanism may be related to the proinflammatory effects of HCl.
Collapse
Affiliation(s)
- Ren Wei
- Department of Vascular and Endovascular Surgery, General Hospital of People’s Liberation Army, Beijing, Fuxing Road 28, 100853, P.R. China
| | - Xiaojing Chang
- Department of Vascular and Endovascular Surgery, General Hospital of People’s Liberation Army, Beijing, Fuxing Road 28, 100853, P.R. China
| | - Zhongyin Wu
- Department of General Surgery, Affiliated Hospital of Chengde Medical University, Nanyingzi Street 36, Chengde City, Hebei, 067000, P.R. China
| | - Chen Duan
- Department of Vascular Surgery, Affiliated Hospital of Inner Mongolia Medical University, Tongdao Road 1, Hohhot City, Inner Mongolia, 010017, P.R.
China
| | - Jiang Xiong
- Department of Vascular and Endovascular Surgery, General Hospital of People’s Liberation Army, Beijing, Fuxing Road 28, 100853, P.R. China
| | - Wei Guo
- Department of Vascular and Endovascular Surgery, General Hospital of People’s Liberation Army, Beijing, Fuxing Road 28, 100853, P.R. China
| |
Collapse
|
3
|
Edaravone Attenuated Angiotensin II-Induced Atherosclerosis and Abdominal Aortic Aneurysms in Apolipoprotein E-Deficient Mice. Biomolecules 2022; 12:biom12081117. [PMID: 36009011 PMCID: PMC9405883 DOI: 10.3390/biom12081117] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2022] [Revised: 08/09/2022] [Accepted: 08/12/2022] [Indexed: 12/17/2022] Open
Abstract
Background: The aim of the study was to define whether edaravone, a free-radical scavenger, influenced angiotensin II (AngII)-induced atherosclerosis and abdominal aortic aneurysms (AAAs) formation. Methods: Male apolipoprotein E-deficient mice (8–12 weeks old) were fed with a normal diet for 5 weeks. Either edaravone (10 mg/kg/day) or vehicle was injected intraperitoneally for 5 weeks. After 1 week of injections, mice were infused subcutaneously with either AngII (1000 ng/kg/min, n = 16–17 per group) or saline (n = 5 per group) by osmotic minipumps for 4 weeks. Results: AngII increased systolic blood pressure equivalently in mice administered with either edaravone or saline. Edaravone had no effect on plasma total cholesterol concentrations and body weights. AngII infusion significantly increased ex vivo maximal diameters of abdominal aortas and en face atherosclerosis but was significantly attenuated by edaravone administration. Edaravone also reduced the incidence of AngII-induced AAAs. In addition, edaravone diminished AngII-induced aortic MMP-2 activation. Quantitative RT-PCR revealed that edaravone ameliorated mRNA abundance of aortic MCP-1 and IL-1β. Immunostaining demonstrated that edaravone attenuated oxidative stress and macrophage accumulation in the aorta. Furthermore, edaravone administration suppressed thioglycolate-induced mice peritoneal macrophages (MPMs) accumulation and mRNA abundance of MCP-1 in MPMs in male apolipoprotein E-deficient mice. In vitro, edaravone reduced LPS-induced mRNA abundance of MCP-1 in MPMs. Conclusions: Edaravone attenuated AngII-induced AAAs and atherosclerosis in male apolipoprotein E-deficient mice via anti-oxidative action and anti-inflammatory effect.
Collapse
|
4
|
Melin LG, Dall JH, Lindholt JS, Steffensen LB, Beck HC, Elkrog SL, Clausen PD, Rasmussen LM, Stubbe J. Cycloastragenol Inhibits Experimental Abdominal Aortic Aneurysm Progression. Biomedicines 2022; 10:biomedicines10020359. [PMID: 35203568 PMCID: PMC8962318 DOI: 10.3390/biomedicines10020359] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Revised: 01/24/2022] [Accepted: 01/30/2022] [Indexed: 02/04/2023] Open
Abstract
The pathogenesis of abdominal aortic aneurysm involves vascular inflammation and elastin degradation. Astragalusradix contains cycloastragenol, which is known to be anti-inflammatory and to protect against elastin degradation. We hypothesized that cycloastragenol supplementation inhibits abdominal aortic aneurysm progression. Abdominal aortic aneurysm was induced in male rats by intraluminal elastase infusion in the infrarenal aorta and treated daily with cycloastragenol (125 mg/kg/day). Aortic expansion was followed weekly by ultrasound for 28 days. Changes in aneurysmal wall composition were analyzed by mRNA levels, histology, zymography and explorative proteomic analyses. At day 28, mean aneurysm diameter was 37% lower in the cycloastragenol group (p < 0.0001). In aneurysm cross sections, elastin content was insignificantly higher in the cycloastragenol group (10.5% ± 5.9% vs. 19.9% ± 16.8%, p = 0.20), with more preserved elastin lamellae structures (p = 0.0003) and without microcalcifications. Aneurysmal matrix metalloprotease-2 activity was reduced by the treatment (p = 0.022). Messenger RNA levels of inflammatory- and anti-oxidative markers did not differ between groups. Explorative proteomic analysis showed no difference in protein levels when adjusting for multiple testing. Among proteins displaying nominal regulation were fibulin-5 (p = 0.02), aquaporin-1 (p = 0.02) and prostacyclin synthase (p = 0.007). Cycloastragenol inhibits experimental abdominal aortic aneurysm progression. The suggested underlying mechanisms involve decreased matrix metalloprotease-2 activity and preservation of elastin and reduced calcification, thus, cycloastragenol could be considered for trial in abdominal aortic aneurysm patients.
Collapse
Affiliation(s)
- Leander Gaarde Melin
- Centre for Individualized Medicine in Arterial Diseases (CIMA), Odense University Hospital (OUH), 5000 Odense, Denmark; (L.G.M.); (J.H.D.); (J.S.L.); (H.C.B.); (L.M.R.)
- Department of Cardiothoracic and Vascular Surgery, Odense University Hospital, 5000 Odense, Denmark
- Cardiovascular and Renal Research Unit, Institute for Molecular Medicine, University of Southern Denmark, 5000 Odense, Denmark; (L.B.S.); (S.L.E.); (P.D.C.)
| | - Julie Husted Dall
- Centre for Individualized Medicine in Arterial Diseases (CIMA), Odense University Hospital (OUH), 5000 Odense, Denmark; (L.G.M.); (J.H.D.); (J.S.L.); (H.C.B.); (L.M.R.)
- Department of Cardiothoracic and Vascular Surgery, Odense University Hospital, 5000 Odense, Denmark
- Cardiovascular and Renal Research Unit, Institute for Molecular Medicine, University of Southern Denmark, 5000 Odense, Denmark; (L.B.S.); (S.L.E.); (P.D.C.)
| | - Jes S. Lindholt
- Centre for Individualized Medicine in Arterial Diseases (CIMA), Odense University Hospital (OUH), 5000 Odense, Denmark; (L.G.M.); (J.H.D.); (J.S.L.); (H.C.B.); (L.M.R.)
- Department of Cardiothoracic and Vascular Surgery, Odense University Hospital, 5000 Odense, Denmark
| | - Lasse B. Steffensen
- Cardiovascular and Renal Research Unit, Institute for Molecular Medicine, University of Southern Denmark, 5000 Odense, Denmark; (L.B.S.); (S.L.E.); (P.D.C.)
| | - Hans Christian Beck
- Centre for Individualized Medicine in Arterial Diseases (CIMA), Odense University Hospital (OUH), 5000 Odense, Denmark; (L.G.M.); (J.H.D.); (J.S.L.); (H.C.B.); (L.M.R.)
- Department of Clinical Biochemistry and Pharmacology, Odense University Hospital, 5000 Odense, Denmark
| | - Sophie L. Elkrog
- Cardiovascular and Renal Research Unit, Institute for Molecular Medicine, University of Southern Denmark, 5000 Odense, Denmark; (L.B.S.); (S.L.E.); (P.D.C.)
| | - Pernille D. Clausen
- Cardiovascular and Renal Research Unit, Institute for Molecular Medicine, University of Southern Denmark, 5000 Odense, Denmark; (L.B.S.); (S.L.E.); (P.D.C.)
| | - Lars Melholt Rasmussen
- Centre for Individualized Medicine in Arterial Diseases (CIMA), Odense University Hospital (OUH), 5000 Odense, Denmark; (L.G.M.); (J.H.D.); (J.S.L.); (H.C.B.); (L.M.R.)
- Department of Clinical Biochemistry and Pharmacology, Odense University Hospital, 5000 Odense, Denmark
| | - Jane Stubbe
- Centre for Individualized Medicine in Arterial Diseases (CIMA), Odense University Hospital (OUH), 5000 Odense, Denmark; (L.G.M.); (J.H.D.); (J.S.L.); (H.C.B.); (L.M.R.)
- Cardiovascular and Renal Research Unit, Institute for Molecular Medicine, University of Southern Denmark, 5000 Odense, Denmark; (L.B.S.); (S.L.E.); (P.D.C.)
- Correspondence: ; Tel.: +45-6550-3709
| |
Collapse
|
5
|
Reversal of elastase-induced abdominal aortic aneurysm following the delivery of nanoparticle-based pentagalloyl glucose (PGG) is associated with reduced inflammatory and immune markers. Eur J Pharmacol 2021; 910:174487. [PMID: 34516951 DOI: 10.1016/j.ejphar.2021.174487] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Revised: 08/30/2021] [Accepted: 09/02/2021] [Indexed: 11/24/2022]
Abstract
OBJECTIVE An Abdominal aortic aneurysm (AAA), a deadly disease in elderly population, is featured by expansion of aortic diameter, degradation and weakening of vasculature. Its common and significant characteristics are disarray and inflammation in vasculature. We tested the hypothesis that the reversal of abdominal aortic aneurysm by pentagalloyl glucose-loaded nanoparticles (PGG-NPs) therapy that targets degraded elastin suppresses inflammatory and immune markers to ameliorate the pathophysiology of the disease in advance stage aneurysm in a porcine pancreatic elastase (PPE)-induced mouse model of AAA. METHODS AND RESULTS After induction of aneurysm in pathogen-free C57BL/6 male mice by applying PPE peri-adventitially to the abdominal aorta, once a week for two doses of intravenous injections of pentagalloyl glucose-loaded nanoparticles (PGG-NPs) conjugated with elastin targeted antibody were used to reverse the aneurysms. We showed that PGG-NPs therapy could suppress infiltration of macrophages, CD8 and CD4 subsets of T cells, matrix metalloproteinases (MMPs), inflammatory cytokines interferon (IFN-γ) and interleukin (IL)-6 at the local and systemic level. Moreover, such PGG-NPs therapy increases the induction of anti-inflammatory cytokines IL-13, IL-27 and IL-10 at the local and systemic level. The therapy also led to remodeling of elastic lamina at the aneurysm site. CONCLUSION Nanoparticles-loaded pentagalloyl glucose therapy can be an effective treatment option against advanced stage aneurysms to reverse the disease by ameliorating inflammation and restoring arterial homeostasis.
Collapse
|
6
|
Qiu R, Chen S, Hua F, Bian S, Chen J, Li G, Wu X. Betanin Prevents Experimental Abdominal Aortic Aneurysm Progression by Modulating the TLR4/NF-κB and Nrf2/HO-1 Pathways. Biol Pharm Bull 2021; 44:1254-1262. [PMID: 34471054 DOI: 10.1248/bpb.b21-00042] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Betanin, a bioactive ingredient mostly isolated from beetroots, exhibits a protective effect against cardiovascular diseases. However, its effects on abdominal aortic aneurysm (AAA) have not been elucidated. In this study, an AAA model was constructed by infusion of porcine pancreatic elastase in C57BL/6 mice. Mice were then administered with betanin or saline intragastrically once daily for 14 d. Our results showed that treatment with betanin remarkably limited AAA enlargement and mitigated the infiltration of inflammatory cells in the adventitia. The increased expression of proinflammatory cytokines and matrix metalloproteinases (MMPs) was also significantly alleviated following betanin treatment. Furthermore, betanin suppressed the activation of toll-like receptor 4 (TLR4)/nuclear factor-kappaB (NF-κB) signaling in the aortic wall, and downregulated the levels of tissue-reactive oxygen species as well as circulating 8-isoprostane by stimulating the nuclear factor-E2-related factor 2 (Nrf2)/heme oxygenase-1 (HO-1) pathway. Taken together, these data suggest that betanin may attenuate AAA progression and may be used as a therapeutic drug against AAA.
Collapse
Affiliation(s)
- Renfeng Qiu
- Department of Vascular Surgery, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University.,Department of Vascular Surgery, Shouguang People Hospital
| | - Shuxiao Chen
- Department of Vascular Surgery, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University
| | - Fang Hua
- Department of Clinical Laboratory, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University
| | - Shuai Bian
- Department of Vascular Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University
| | - Jianfeng Chen
- Department of Vascular Surgery, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University
| | - Gang Li
- Department of Vascular Surgery, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University.,Department of Vascular Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University
| | - Xuejun Wu
- Department of Vascular Surgery, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University.,Department of Vascular Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University
| |
Collapse
|
7
|
Sassetti E, Clausen MH, Laraia L. Small-Molecule Inhibitors of Reactive Oxygen Species Production. J Med Chem 2021; 64:5252-5275. [PMID: 33856791 DOI: 10.1021/acs.jmedchem.0c01914] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Reactive oxygen species (ROS) are involved in physiological cellular processes including differentiation, proliferation, and apoptosis by acting as signaling molecules or regulators of transcription factors. The maintenance of appropriate cellular ROS levels is termed redox homeostasis, a balance between their production and neutralization. High concentrations of ROS may contribute to severe pathological events including cancer, neurodegenerative, and cardiovascular diseases. In recent years, approaches to target the sources of ROS production directly in order to develop tool compounds or potential therapeutics have been explored. Herein, we briefly outline the major sources of cellular ROS production and comprehensively review the targeting of these by small-molecule inhibitors. We critically assess the value of ROS inhibitors with different mechanisms-of-action, including their potency, mode-of-action, known off-target effects, and clinical or preclinical status, while suggesting future avenues of research in the field.
Collapse
Affiliation(s)
- Elisa Sassetti
- Center for Nanomedicine and Theranostics, Department of Chemistry, Technical University of Denmark, Kemitorvet 207, 2800 Kgs. Lyngby, Denmark
| | - Mads H Clausen
- Center for Nanomedicine and Theranostics, Department of Chemistry, Technical University of Denmark, Kemitorvet 207, 2800 Kgs. Lyngby, Denmark
| | - Luca Laraia
- Center for Nanomedicine and Theranostics, Department of Chemistry, Technical University of Denmark, Kemitorvet 207, 2800 Kgs. Lyngby, Denmark
| |
Collapse
|
8
|
Endotoxin Tolerance in Abdominal Aortic Aneurysm Macrophages, In Vitro: A Case-Control Study. Antioxidants (Basel) 2020; 9:antiox9090896. [PMID: 32967278 PMCID: PMC7554856 DOI: 10.3390/antiox9090896] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2020] [Revised: 09/09/2020] [Accepted: 09/16/2020] [Indexed: 12/21/2022] Open
Abstract
Macrophages are implicated in the pathogenesis of abdominal aortic aneurysm (AAA). This study examined the environmentally conditioned responses of AAA macrophages to inflammatory stimuli. Plasma- and blood-derived monocytes were separated from the whole blood of patients with AAA (30–45 mm diameter; n = 33) and sex-matched control participants (n = 44). Increased concentrations of pro-inflammatory and pro-oxidant biomarkers were detected in the plasma of AAA patients, consistent with systemic inflammation and oxidative stress. However, in monocyte-derived macrophages, a suppressed cytokine response was observed in AAA compared to the control following stimulation with lipopolysaccharide (LPS) (tumor necrosis factor alpha (TNF-α) 26.9 ± 3.3 vs. 15.5 ± 3.2 ng/mL, p < 0.05; IL-6 3.2 ± 0.6 vs. 1.4 ± 0.3 ng/mL, p < 0.01). LPS-stimulated production of 8-isoprostane, a biomarker of oxidative stress, was also markedly lower in AAA compared to control participants. These findings are consistent with developed tolerance in human AAA macrophages. As Toll-like receptor 4 (TLR4) has been implicated in tolerance, macrophages were examined for changes in TLR4 expression and distribution. Although TLR4 mRNA and protein expression were unaltered in AAA, cytosolic internalization of receptors and lipid rafts was found. These findings suggest the inflamed, pro-oxidant AAA microenvironment favors macrophages with an endotoxin-tolerant-like phenotype characterized by a diminished capacity to produce pro-inflammatory mediators that enhance the immune response.
Collapse
|
9
|
Lai CH, Chang JY, Wang KC, Lee FT, Wu HL, Cheng TL. Pharmacological Inhibition of Cathepsin S Suppresses Abdominal Aortic Aneurysm in Mice. Eur J Vasc Endovasc Surg 2020; 59:990-999. [PMID: 32033870 DOI: 10.1016/j.ejvs.2020.01.008] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2019] [Revised: 11/30/2019] [Accepted: 01/09/2020] [Indexed: 12/14/2022]
Abstract
OBJECTIVE Evidence suggests that cathepsin S (CTSS), a potent mammalian elastase, participates in abdominal aortic aneurysm (AAA) formation. This study examines the hypothesis that pharmacological inhibition of CTSS with an α-ketoamide based compound 6r might suppress AAA in mice. METHODS Experimental study of the CaCl2 induced AAA model in B6 mice and angiotensin II (AngII) infused AAA model in ApoE-/- mice. The effects of intraperitoneal administration of 6r (25 mg/kg) and vehicle every three days since one day after AAA induction were evaluated at 28 days using CaCl2 induced (n = 12 per group) and AngII infused (n = 8 per group) models. Additionally, the effects of post-treatment with 6r and vehicle from seven days or 14 days after AAA induction were evaluated at 28 days using the CaCl2 induced model (n = 6 per group). Aortic samples were harvested for histological and biochemical analyses, including cathepsin levels, Verhoeff Van Gieson staining, TUNEL assay, and immunostaining for macrophages. RESULTS In the CaCl2 induced model, treatment with 6r suppressed aortic dilatation observed in vehicle treated controls (median: 0.58 vs. 0.92 mm; p < .001), along with reduced CTSS and cathepsin K (CTSK) levels (both p < .001), preserved elastin integrity (p < .001), fewer medial apoptotic cells (p = .012) and less macrophage infiltration (p = .041). In the AngII infused model, the aortic diameter was smaller in 6r treated mice than in vehicle treated controls (median: 0.95 vs. 1.84 mm; p = .047). The levels of CTSS (p < .001) and CTSK (p = .033) and the numbers of elastin breaks (p < .001), medial apoptotic cells (p < .001) and infiltrating macrophages (p = .030) were attenuated under 6r treatment. Finally, post-treatment with 6r from seven days (p = .046) or 14 days (p = .012) after AAA induction limited CaCl2 induced AAA. CONCLUSION Pharmacological inhibition of CTSS by 6r suppresses AAA formation in mice. Also, post-treatment with 6r retards mouse AAA progression. These findings provide proof of concept validation for CTSS as a potential therapeutic target in AAA.
Collapse
Affiliation(s)
- Chao-Han Lai
- Department of Surgery, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan; Cardiovascular Research Centre, College of Medicine, National Cheng Kung University, Taiwan
| | - Jang-Yang Chang
- Department of Internal Medicine, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan; National Institute of Cancer Research, National Health Research Institutes, Tainan, Taiwan
| | - Kuan-Chieh Wang
- Department of Pharmacy, Chia-Nan University of Pharmacy and Science, Tainan, Taiwan
| | - Fang-Tzu Lee
- Department of Surgery, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan; Cardiovascular Research Centre, College of Medicine, National Cheng Kung University, Taiwan; Department of Biochemistry and Molecular Biology, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Hua-Lin Wu
- Cardiovascular Research Centre, College of Medicine, National Cheng Kung University, Taiwan; Department of Biochemistry and Molecular Biology, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Tsung-Lin Cheng
- Cardiovascular Research Centre, College of Medicine, National Cheng Kung University, Taiwan; Department of Physiology, School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan; Orthopaedic Research Centre, College of Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan; Department of Medical Research, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan.
| |
Collapse
|
10
|
The role of IL-1β in aortic aneurysm. Clin Chim Acta 2020; 504:7-14. [PMID: 31945339 DOI: 10.1016/j.cca.2020.01.007] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2019] [Revised: 01/07/2020] [Accepted: 01/08/2020] [Indexed: 12/12/2022]
Abstract
Interleukin-1β (IL-1β) is a vital cytokine that plays an important role in regulating immune responses to infectious challenges and sterile insults. In addition, two endogenous inhibitors of functional receptor binding, IL-1 receptor antagonist (IL-1Ra), complete the family. To gain biological activity, IL-1β requires processing by the protease caspase-1 and activation of inflammasomes. Numerous clinical association studies and experimental approaches have implicated members of the IL-1 family, their receptors, or components of the processing machinery in the underlying processes of cardiovascular diseases. Here, we summarize the current state of knowledge regarding the pro-inflammatory and disease-modulating role of the IL-1 family in aneurysm. We discuss clinical evidence, signalling pathway, and mechanism of action and last, lend a perspective on currently developing therapeutic strategies involving IL-1β in aneurysm.
Collapse
|
11
|
Yoshimura K, Morikage N, Nishino-Fujimoto S, Furutani A, Shirasawa B, Hamano K. Current Status and Perspectives on Pharmacologic Therapy for Abdominal Aortic Aneurysm. Curr Drug Targets 2019; 19:1265-1275. [PMID: 29284386 PMCID: PMC6182934 DOI: 10.2174/1389450119666171227223331] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2017] [Revised: 12/13/2017] [Accepted: 12/13/2017] [Indexed: 01/16/2023]
Abstract
Background: Abdominal aortic aneurysm (AAA), a common disease involving the segmen-tal expansion and rupture of the aorta, has a high mortality rate. Therapeutic options for AAA are cur-rently limited to surgical repair to prevent catastrophic rupture. Non-surgical approaches, particularly pharmacotherapy, are lacking for the treatment of AAA. Objective: We review both basic and clinical studies and discuss the current challenges to developing medical therapy that reduces AAA progression. Results: Studies using animal models of AAA progression and human AAA explant cultures have identified several potential targets for preventing AAA growth. However, no clinical studies have con-vincingly confirmed the efficacy of any pharmacologic treatment against the growth of AAA. Thus, there is as yet no strong recommendation regarding pharmacotherapy to reduce the risk of AAA pro-gression and rupture. Conclusion: This review identifies concerns that need to be addressed for the field to progress and dis-cusses the challenges that must be overcome in order to develop effective pharmacotherapy to reduce AAA progression in the future.
Collapse
Affiliation(s)
- Koichi Yoshimura
- Department of Surgery and Clinical Science, Yamaguchi University Graduate School of Medicine, Ube, 755-8505, Japan.,Graduate School of Health and Welfare, Yamaguchi Prefectural University, Yamaguchi, 753-8502, Japan
| | - Noriyasu Morikage
- Department of Surgery and Clinical Science, Yamaguchi University Graduate School of Medicine, Ube, 755-8505, Japan
| | - Shizuka Nishino-Fujimoto
- Department of Surgery and Clinical Science, Yamaguchi University Graduate School of Medicine, Ube, 755-8505, Japan
| | - Akira Furutani
- Department of Surgery, Yamaguchi Rosai Hospital, Sanyo-Onoda, 756-0095, Japan
| | - Bungo Shirasawa
- Department of Medical Education, Yamaguchi University Graduate School of Medicine, Ube, 755-8505, Japan
| | - Kimikazu Hamano
- Department of Surgery and Clinical Science, Yamaguchi University Graduate School of Medicine, Ube, 755-8505, Japan
| |
Collapse
|
12
|
Abstract
Current management of aortic aneurysms relies exclusively on prophylactic operative repair of larger aneurysms. Great potential exists for successful medical therapy that halts or reduces aneurysm progression and hence alleviates or postpones the need for surgical repair. Preclinical studies in the context of abdominal aortic aneurysm identified hundreds of candidate strategies for stabilization, and data from preoperative clinical intervention studies show that interventions in the pathways of the activated inflammatory and proteolytic cascades in enlarging abdominal aortic aneurysm are feasible. Similarly, the concept of pharmaceutical aorta stabilization in Marfan syndrome is supported by a wealth of promising studies in the murine models of Marfan syndrome-related aortapathy. Although some clinical studies report successful medical stabilization of growing aortic aneurysms and aortic root stabilization in Marfan syndrome, these claims are not consistently confirmed in larger and controlled studies. Consequently, no medical therapy can be recommended for the stabilization of aortic aneurysms. The discrepancy between preclinical successes and clinical trial failures implies shortcomings in the available models of aneurysm disease and perhaps incomplete understanding of the pathological processes involved in later stages of aortic aneurysm progression. Preclinical models more reflective of human pathophysiology, identification of biomarkers to predict severity of disease progression, and improved design of clinical trials may more rapidly advance the opportunities in this important field.
Collapse
Affiliation(s)
- Jan H. Lindeman
- Dept. Vascular Surgery, Leiden University Medical Center, The Netherlands
| | - Jon S. Matsumura
- Division of Vascular Surgery, School of Medicine and Public Health, University of Wisconsin, Madison, Wisconsin
| |
Collapse
|
13
|
Hu J, Luo J, Wang H, Wang C, Long R, Li A, Zhou Y, Fang Z, Chen Q. The active participation of p22phox-214T/C in the formation of intracranial aneurysm and the suppressive potential of edaravone. Int J Mol Med 2018; 42:2952-2960. [PMID: 30226557 DOI: 10.3892/ijmm.2018.3846] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2018] [Accepted: 08/23/2018] [Indexed: 11/06/2022] Open
Abstract
Oxidative stress reactions play an important role in the pathogenesis of intracranial aneurysm (IA). p22phox is involved in the oxidative stress reaction, and it is a critical subunit of nicotinamide adenine dinucleotide phosphate (NADPH) oxidase. The present study investigated the association of genetic variants within the gene encoding p22phox‑214T/C with IA. The p22phox‑214T/C gene polymorphisms in 192 cases of IA and 112 controls were analyzed by polymerase chain reaction‑restriction fragment length polymorphism (PCR‑RFLP). The mRNA expression of NADPH oxidase was also analyzed by RT‑PCR. The results of RT‑PCR were validated by ELISA. In a rabbit model of elastase‑induced aneurysm, we used edaravone for anti‑oxidative stress treatment to observe the curative effects. In the clinical cases, a significant difference in p22phox‑214T/C allele frequencies in the IA group was observed compared with the control group (P<0.001). The expression level of NADPH oxidase was differed significantly between the IA group and the control group. In the rabbit model of elastase‑induced aneurysm, the success rate of the aneurysmal model in the edaravone group and the wound ulcer rate were lower than those in the control group. In addition, the diameter of the aneurysm was smaller than in the edaravone group than in the control group (3.26±0.13 mm vs. 3.85±0.07 mm), and the expression of matrix metalloproteinase‑9 (MMP‑9) was significantly lower than that in the control group (P<0.0001). Thus, these data suggest the active participation of p22phox‑214T/C in the formation of IA and the suppressive potential of edaravone against IA formation.
Collapse
Affiliation(s)
- Juntao Hu
- Department of Neurosurgery, Remin Hospital of Wuhan University, Wuhan, Hubei 30060, P.R. China
| | - Jie Luo
- Department of Neurosurgery, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei 442000, P.R. China
| | - Hui Wang
- Department of Neurosurgery, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei 442000, P.R. China
| | - Chaojia Wang
- Department of Neurosurgery, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei 442000, P.R. China
| | - Rongpei Long
- Department of English, Hubei University of Medicine, Shiyan, Hubei 442000, P.R. China
| | - Anrong Li
- Department of Neurosurgery, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei 442000, P.R. China
| | - Yi Zhou
- Department of Neurosurgery, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei 442000, P.R. China
| | - Zhicheng Fang
- Department of Neurosurgery, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei 442000, P.R. China
| | - Qianxue Chen
- Department of Neurosurgery, Remin Hospital of Wuhan University, Wuhan, Hubei 30060, P.R. China
| |
Collapse
|
14
|
Dose-dependency of multiple free radical-scavenging activity of edaravone. J Surg Res 2018; 228:147-153. [DOI: 10.1016/j.jss.2018.03.020] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2017] [Revised: 02/16/2018] [Accepted: 03/14/2018] [Indexed: 02/08/2023]
|
15
|
Peng J, He X, Zhang L, Liu P. MicroRNA‑26a protects vascular smooth muscle cells against H2O2‑induced injury through activation of the PTEN/AKT/mTOR pathway. Int J Mol Med 2018; 42:1367-1378. [PMID: 29956734 PMCID: PMC6089772 DOI: 10.3892/ijmm.2018.3746] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2017] [Accepted: 06/20/2018] [Indexed: 01/12/2023] Open
Abstract
Abdominal aortic aneurysm (AAA) is a common disease, which is characterized by the apoptosis of vascular smooth muscle cells (VSMCs). In previous years, microRNAs (miRNAs) have been associated with AAA and functionally implicated in the pathogenesis of this disease. However, the role of miRNAs in the apoptosis of VSMCs remains to be fully elucidated. The present study aimed to elucidate the role and mechanism of miRNAs in protecting against hydrogen peroxide (H2O2)-induced apoptosis in VSMCs. The expression of miRNAs in peripheral blood from patients diagnosed with AAA was analyzed using a microarray and reverse transcription polymerase chain reaction. A VSMC injury model induced by H2O2 was used to determine the potential role of miR-26a against cell injury. Cell viability, cell apoptosis and reactive oxygen species (ROS) generation were determined by a CCK8 assay, flow cytometry and a 2′,7′-DCF diacetate assay, respectively. It was observed that miRNA (miR)-26a (miR-26a-1-5p) was significantly downregulated in peripheral blood samples from patients with AAA. It was revealed that H2O2 treatment dose-dependently inhibited cell viability, enhanced apoptosis and induced the production of ROS, which indicated the success of the model establishment. It was also observed that miR-26a was downregulated in the VSMCs following H2O2 stimulation. The upregulation of miR-26a attenuated H2O2-induced cell injury, as evidenced by the enhancement of cell viability, and inhibition of the activity of caspase-3, apoptosis and ROS production. In addition, phosphatase and tensin homolog (PTEN), a well-known regulator of the AKT/mammalian target of rapamycin (mTOR) pathway, was found to be a direct target of miR-26a in the VSMCs and this was validated using a luciferase reporter assay. Overexpression of PTEN by pcDNA-PTEN plasmids markedly eliminated the protective effects of the overexpression of miR-26a on H2O2-induced cell injury. Finally, it was found that miR-26a mediated its anti-apoptotic action by reactivation of the AKT/mTOR pathway, as demonstrated by the upregulation of phosphorylated (p-)AKT and p-mTOR, and the Akt inhibitor API-2 reversing the protective effects on VSMCs mediated by miR-26a. These results indicated that miR-26a protected VSMCs against H2O2-induced injury through activation of the PTEN/AKT/mTOR pathway, and miR-26a may be considered as a potential prognostic biomarker and therapeutic target in the treatment of AAA.
Collapse
Affiliation(s)
- Junlu Peng
- Department of Vascular Surgery, The First Hospital of Hebei Medical University, Shijiazhuang, Hebei 050000, P.R. China
| | - Xinqi He
- Department of Vascular Surgery, The First Hospital of Hebei Medical University, Shijiazhuang, Hebei 050000, P.R. China
| | - Lei Zhang
- Department of Vascular Surgery, The First Hospital of Hebei Medical University, Shijiazhuang, Hebei 050000, P.R. China
| | - Peng Liu
- Department of Vascular Surgery, The First Hospital of Hebei Medical University, Shijiazhuang, Hebei 050000, P.R. China
| |
Collapse
|
16
|
Tekin G, İsbir S, Şener G, Çevik Ö, Çetinel Ş, Dericioğlu O, Arsan S, Çobanoğlu A. The preventive and curative effects of melatonin against abdominal aortic aneurysm in rats. J Vasc Surg 2017; 67:1546-1555. [PMID: 28478022 DOI: 10.1016/j.jvs.2017.04.028] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2015] [Accepted: 06/05/2016] [Indexed: 12/17/2022]
Abstract
OBJECTIVE Oxygen free radicals are important components involved in the histopathologic tissue alterations observed during abdominal aortic aneurysms (AAAs). This study examined whether melatonin has protective or therapeutic effects against AAAs. METHODS Sprague-Dawley rats were divided into four groups. A CaCl2 model was used to induce AAA. Starting on the operation day (Mel+AAA+Mel group) or 4 weeks after the operation (AAA+Mel group), the rats received intraperitoneal melatonin (10 mg/kg/day) for 6 and 2 weeks, respectively. The control and AAA groups received vehicle for 2 weeks after the sham operation and AAA induction, respectively. Angiographic measurements were recorded at the beginning, week 4, and week 6 of the study. After decapitation, aorta tissues were taken for the measurement of malondialdehyde, 8-hydroxy-2'-deoxyguanosine, glutathione levels, and myeloperoxidase and caspase-3 activity. Matrix metalloproteinase (MMP)-2, MMP-9, tumor necrosis factor-α, and inducible nitric oxide synthase protein expressions were analyzed by Western blot technique. Aortic tissues were also examined by light microscopy. RESULTS CaCl2 caused an inflammatory response and oxidative damage indicated by rises in malondialdehyde and 8-hydroxy-2'-deoxyguanosine levels. Myeloperoxidase and caspase-3 activities were increased, but glutathione levels were reduced. On the one hand, MMP-2, MMP-9, tumor necrosis factor-α, and inducible nitric oxide synthase protein expressions were increased in the vehicle-treated AAA group. On the other hand, melatonin treatment reversed all of these biochemical indices and histopathologic alterations. CONCLUSIONS According to the data, although melatonin tended to reverse the biochemical parameters given on week 4, the preventive effect is more pronounced when given concomitantly with AAA induction because values were closer to the control levels.
Collapse
Affiliation(s)
- Gözde Tekin
- Department of Cardiovascular Surgery, School of Medicine, Marmara University, İstanbul, Turkey.
| | - Selim İsbir
- Department of Cardiovascular Surgery, School of Medicine, Marmara University, İstanbul, Turkey
| | - Göksel Şener
- Department of Pharmacology, School of Pharmacy, Marmara University, İstanbul, Turkey
| | - Özge Çevik
- Department of Biochemistry, School of Pharmacy, Cumhuriyet University, Sivas, Turkey
| | - Şule Çetinel
- Department of Histology and Embryology, School of Medicine, Marmara University, İstanbul, Turkey
| | - Okan Dericioğlu
- Department of Cardiovascular Surgery, School of Medicine, Marmara University, İstanbul, Turkey
| | - Sinan Arsan
- Department of Cardiovascular Surgery, School of Medicine, Marmara University, İstanbul, Turkey
| | - Adnan Çobanoğlu
- Department of Cardiovascular Surgery, School of Medicine, Marmara University, İstanbul, Turkey
| |
Collapse
|
17
|
Yamamura M, Miyamoto Y, Mitsuno M, Tanaka H, Ryomoto M. Pretreatment with the Free Radical Scavenger Edaravone Mitigates Kidney Glycogen Depletion and Neutrophil Infiltration after Leg Ischemia in a Rat Model: A Pilot Study. Ann Vasc Dis 2017. [PMID: 29515706 PMCID: PMC5835430 DOI: 10.3400/avd.oa.17-00019] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Objective: We have previously shown that pretreatment with the free radical scavenger edaravone (Radicut®, Mitsubishi Tanabe Pharma Co., Japan) mitigated skeletal muscle damage due to ischemia reperfusion. In this study, we sought to validate its use in an experimental model of myonephropathic-metabolic syndrome (MNMS). Methods: Either edaravone (3.0 mg/kg; edaravone group; n=4) or saline (saline group; n=6) was intraperitoneally injected into male Lewis rats (508±31 g). Normal kidneys were harvested as control (n=3). MNMS was induced by bilaterally clamping the common femoral arteries for 5 h and declamping 5 h later. Kidney damage was evaluated by quantifying Periodic Acid Schiff (PAS)-positive area (glycogen storage) and esterase-positive cells (neutrophil infiltration). Results: The PAS-positive area in the saline group was significantly lower than that in the normal group (36.9±2.6 vs. 66.9±1.2%, P<0.01); the PAS-positive area in the edaravone group remained comparable to that in the normal group (52.9±0.9%, P<0.01). Esterase-positive cells in the saline group were significantly higher than in normal kidneys (62.4±5.6 vs. 17.5±2.4 cells/mm2, P<0.01), while they were significantly reduced in the edaravone group (32.8±5.7 cells/mm2, P<0.01). Conclusion: Edaravone pretreatment mitigates MNMS-induced kidney damage by reducing both glycogen depletion and neutrophil infiltration.
Collapse
Affiliation(s)
- Mitsuhiro Yamamura
- Department of Cardiovascular Surgery, Hyogo College of Medicine, Nishinomiya, Hyogo, Japan
| | - Yuji Miyamoto
- Department of Cardiovascular Surgery, Hyogo College of Medicine, Nishinomiya, Hyogo, Japan
| | - Masataka Mitsuno
- Department of Cardiovascular Surgery, Hyogo College of Medicine, Nishinomiya, Hyogo, Japan
| | - Hiroe Tanaka
- Department of Cardiovascular Surgery, Hyogo College of Medicine, Nishinomiya, Hyogo, Japan
| | - Masaaki Ryomoto
- Department of Cardiovascular Surgery, Hyogo College of Medicine, Nishinomiya, Hyogo, Japan
| |
Collapse
|
18
|
Lu WW, Jia LX, Ni XQ, Zhao L, Chang JR, Zhang JS, Hou YL, Zhu Y, Guan YF, Yu YR, Du J, Tang CS, Qi YF. Intermedin1-53 Attenuates Abdominal Aortic Aneurysm by Inhibiting Oxidative Stress. Arterioscler Thromb Vasc Biol 2016; 36:2176-2190. [PMID: 27634835 DOI: 10.1161/atvbaha.116.307825] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2015] [Accepted: 08/31/2016] [Indexed: 12/20/2022]
Abstract
OBJECTIVE Oxidative stress plays a critical role in the development of abdominal aortic aneurysm (AAA). Intermedin (IMD) is a regulator of oxidative stress. Here, we investigated whether IMD reduces AAA by inhibiting oxidative stress. APPROACH AND RESULTS In angiotensin II-induced ApoE-/- mouse and CaCl2-induced C57BL/6J mouse model of AAA, IMD1-53 significantly reduced the incidence of AAA and maximal aortic diameter. Ultrasonography, hematoxylin, and eosin staining and Verhoeff-van Gieson staining showed that IMD1-53 significantly decreased the enlarged aortas and elastic lamina degradation induced by angiotensin II or CaCl2. Mechanistically, IMD1-53 attenuated oxidative stress, inflammation, vascular smooth muscle cell apoptosis, and matrix metalloproteinase activation. IMD1-53 inhibited the activation of redox-sensitive signaling pathways, decreased the mRNA and protein expression of nicotinamide adenine dinucleotide phosphate oxidase subunits, and reduced the activity of nicotinamide adenine dinucleotide phosphate oxidase in AAA mice. Expression of Nox4 was upregulated in human AAA segments and in angiotensin II-treated mouse aortas and was markedly decreased by IMD1-53. In vitro, vascular smooth muscle cells with small-interfering RNA knockdown of IMD showed significantly increased angiotensin II-induced reactive oxygen species, and small-interfering RNA knockdown of Nox4 markedly inhibited the reactive oxygen species. IMD knockdown further increased the apoptosis of vascular smooth muscle cells and inflammation, which was reversed by Nox4 knockdown. Preincubation with IMD17-47 and protein kinase A inhibitor H89 inhibited the effect of IMD1-53, reducing Nox4 protein levels. CONCLUSIONS IMD1-53 could have a protective effect on AAA by inhibiting oxidative stress.
Collapse
Affiliation(s)
- Wei-Wei Lu
- From the Key Laboratory of Remodeling-Related Cardiovascular Diseases, Beijing Institute of Heart, Lung, and Blood Vessel Diseases, Beijing An Zhen Hospital, Capital Medical University, Ministry of Education, China (W.-W.L., L.-X.J., X.-Q.N., L.Z., Y.-L.H., J.D., Y.-F.Q.); Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China (W.-W.L., X.-Q.N., L.Z., J.-R.C., J.-S.Z., Y.Z., Y.-F.G., C.-S.T., Y.-F.Q.); and Department of Pathogen Biology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China (W.-W.L., X.-Q.N., J.-S.Z., Y.-L.H., Y.-R.Y., Y.-F.Q.)
| | - Li-Xin Jia
- From the Key Laboratory of Remodeling-Related Cardiovascular Diseases, Beijing Institute of Heart, Lung, and Blood Vessel Diseases, Beijing An Zhen Hospital, Capital Medical University, Ministry of Education, China (W.-W.L., L.-X.J., X.-Q.N., L.Z., Y.-L.H., J.D., Y.-F.Q.); Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China (W.-W.L., X.-Q.N., L.Z., J.-R.C., J.-S.Z., Y.Z., Y.-F.G., C.-S.T., Y.-F.Q.); and Department of Pathogen Biology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China (W.-W.L., X.-Q.N., J.-S.Z., Y.-L.H., Y.-R.Y., Y.-F.Q.)
| | - Xian-Qiang Ni
- From the Key Laboratory of Remodeling-Related Cardiovascular Diseases, Beijing Institute of Heart, Lung, and Blood Vessel Diseases, Beijing An Zhen Hospital, Capital Medical University, Ministry of Education, China (W.-W.L., L.-X.J., X.-Q.N., L.Z., Y.-L.H., J.D., Y.-F.Q.); Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China (W.-W.L., X.-Q.N., L.Z., J.-R.C., J.-S.Z., Y.Z., Y.-F.G., C.-S.T., Y.-F.Q.); and Department of Pathogen Biology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China (W.-W.L., X.-Q.N., J.-S.Z., Y.-L.H., Y.-R.Y., Y.-F.Q.)
| | - Lei Zhao
- From the Key Laboratory of Remodeling-Related Cardiovascular Diseases, Beijing Institute of Heart, Lung, and Blood Vessel Diseases, Beijing An Zhen Hospital, Capital Medical University, Ministry of Education, China (W.-W.L., L.-X.J., X.-Q.N., L.Z., Y.-L.H., J.D., Y.-F.Q.); Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China (W.-W.L., X.-Q.N., L.Z., J.-R.C., J.-S.Z., Y.Z., Y.-F.G., C.-S.T., Y.-F.Q.); and Department of Pathogen Biology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China (W.-W.L., X.-Q.N., J.-S.Z., Y.-L.H., Y.-R.Y., Y.-F.Q.)
| | - Jin-Rui Chang
- From the Key Laboratory of Remodeling-Related Cardiovascular Diseases, Beijing Institute of Heart, Lung, and Blood Vessel Diseases, Beijing An Zhen Hospital, Capital Medical University, Ministry of Education, China (W.-W.L., L.-X.J., X.-Q.N., L.Z., Y.-L.H., J.D., Y.-F.Q.); Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China (W.-W.L., X.-Q.N., L.Z., J.-R.C., J.-S.Z., Y.Z., Y.-F.G., C.-S.T., Y.-F.Q.); and Department of Pathogen Biology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China (W.-W.L., X.-Q.N., J.-S.Z., Y.-L.H., Y.-R.Y., Y.-F.Q.)
| | - Jin-Sheng Zhang
- From the Key Laboratory of Remodeling-Related Cardiovascular Diseases, Beijing Institute of Heart, Lung, and Blood Vessel Diseases, Beijing An Zhen Hospital, Capital Medical University, Ministry of Education, China (W.-W.L., L.-X.J., X.-Q.N., L.Z., Y.-L.H., J.D., Y.-F.Q.); Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China (W.-W.L., X.-Q.N., L.Z., J.-R.C., J.-S.Z., Y.Z., Y.-F.G., C.-S.T., Y.-F.Q.); and Department of Pathogen Biology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China (W.-W.L., X.-Q.N., J.-S.Z., Y.-L.H., Y.-R.Y., Y.-F.Q.)
| | - Yue-Long Hou
- From the Key Laboratory of Remodeling-Related Cardiovascular Diseases, Beijing Institute of Heart, Lung, and Blood Vessel Diseases, Beijing An Zhen Hospital, Capital Medical University, Ministry of Education, China (W.-W.L., L.-X.J., X.-Q.N., L.Z., Y.-L.H., J.D., Y.-F.Q.); Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China (W.-W.L., X.-Q.N., L.Z., J.-R.C., J.-S.Z., Y.Z., Y.-F.G., C.-S.T., Y.-F.Q.); and Department of Pathogen Biology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China (W.-W.L., X.-Q.N., J.-S.Z., Y.-L.H., Y.-R.Y., Y.-F.Q.)
| | - Yi Zhu
- From the Key Laboratory of Remodeling-Related Cardiovascular Diseases, Beijing Institute of Heart, Lung, and Blood Vessel Diseases, Beijing An Zhen Hospital, Capital Medical University, Ministry of Education, China (W.-W.L., L.-X.J., X.-Q.N., L.Z., Y.-L.H., J.D., Y.-F.Q.); Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China (W.-W.L., X.-Q.N., L.Z., J.-R.C., J.-S.Z., Y.Z., Y.-F.G., C.-S.T., Y.-F.Q.); and Department of Pathogen Biology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China (W.-W.L., X.-Q.N., J.-S.Z., Y.-L.H., Y.-R.Y., Y.-F.Q.)
| | - You-Fei Guan
- From the Key Laboratory of Remodeling-Related Cardiovascular Diseases, Beijing Institute of Heart, Lung, and Blood Vessel Diseases, Beijing An Zhen Hospital, Capital Medical University, Ministry of Education, China (W.-W.L., L.-X.J., X.-Q.N., L.Z., Y.-L.H., J.D., Y.-F.Q.); Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China (W.-W.L., X.-Q.N., L.Z., J.-R.C., J.-S.Z., Y.Z., Y.-F.G., C.-S.T., Y.-F.Q.); and Department of Pathogen Biology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China (W.-W.L., X.-Q.N., J.-S.Z., Y.-L.H., Y.-R.Y., Y.-F.Q.)
| | - Yan-Rong Yu
- From the Key Laboratory of Remodeling-Related Cardiovascular Diseases, Beijing Institute of Heart, Lung, and Blood Vessel Diseases, Beijing An Zhen Hospital, Capital Medical University, Ministry of Education, China (W.-W.L., L.-X.J., X.-Q.N., L.Z., Y.-L.H., J.D., Y.-F.Q.); Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China (W.-W.L., X.-Q.N., L.Z., J.-R.C., J.-S.Z., Y.Z., Y.-F.G., C.-S.T., Y.-F.Q.); and Department of Pathogen Biology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China (W.-W.L., X.-Q.N., J.-S.Z., Y.-L.H., Y.-R.Y., Y.-F.Q.)
| | - Jie Du
- From the Key Laboratory of Remodeling-Related Cardiovascular Diseases, Beijing Institute of Heart, Lung, and Blood Vessel Diseases, Beijing An Zhen Hospital, Capital Medical University, Ministry of Education, China (W.-W.L., L.-X.J., X.-Q.N., L.Z., Y.-L.H., J.D., Y.-F.Q.); Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China (W.-W.L., X.-Q.N., L.Z., J.-R.C., J.-S.Z., Y.Z., Y.-F.G., C.-S.T., Y.-F.Q.); and Department of Pathogen Biology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China (W.-W.L., X.-Q.N., J.-S.Z., Y.-L.H., Y.-R.Y., Y.-F.Q.)
| | - Chao-Shu Tang
- From the Key Laboratory of Remodeling-Related Cardiovascular Diseases, Beijing Institute of Heart, Lung, and Blood Vessel Diseases, Beijing An Zhen Hospital, Capital Medical University, Ministry of Education, China (W.-W.L., L.-X.J., X.-Q.N., L.Z., Y.-L.H., J.D., Y.-F.Q.); Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China (W.-W.L., X.-Q.N., L.Z., J.-R.C., J.-S.Z., Y.Z., Y.-F.G., C.-S.T., Y.-F.Q.); and Department of Pathogen Biology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China (W.-W.L., X.-Q.N., J.-S.Z., Y.-L.H., Y.-R.Y., Y.-F.Q.)
| | - Yong-Fen Qi
- From the Key Laboratory of Remodeling-Related Cardiovascular Diseases, Beijing Institute of Heart, Lung, and Blood Vessel Diseases, Beijing An Zhen Hospital, Capital Medical University, Ministry of Education, China (W.-W.L., L.-X.J., X.-Q.N., L.Z., Y.-L.H., J.D., Y.-F.Q.); Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China (W.-W.L., X.-Q.N., L.Z., J.-R.C., J.-S.Z., Y.Z., Y.-F.G., C.-S.T., Y.-F.Q.); and Department of Pathogen Biology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China (W.-W.L., X.-Q.N., J.-S.Z., Y.-L.H., Y.-R.Y., Y.-F.Q.).
| |
Collapse
|
19
|
Prevention of abdominal aortic aneurysm progression by oral administration of green tea polyphenol in a rat model. J Vasc Surg 2016; 65:1803-1812.e2. [PMID: 27473778 DOI: 10.1016/j.jvs.2016.06.003] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2016] [Accepted: 06/05/2016] [Indexed: 01/23/2023]
Abstract
OBJECTIVE Inflammation-mediated elastin destruction in the aortic medial layer is related to progression of abdominal aortic aneurysm (AAA). Epigallocatechin-3-gallate (EGCG), a major component of green tea polyphenols, reportedly increases elastin synthesis in vitro and may possess anti-inflammatory effects. We used a rat model to investigate whether EGCG could prevent AAA progression. METHODS AAA was induced with administration of intraluminal elastase and extraluminal CaCl2 in male rats. Rats were randomly divided into a control group (n = 30) and an EGCG group (n = 30). In the EGCG group, an EGCG solution (20 mg/d) was administered orally to each rat from 2 weeks before AAA induction and continued 4 weeks beyond induction. RESULTS The abdominal aortic diameter was significantly smaller in the EGCG group than in the control group on day 28 (2.9 ± 0.2 vs 2.3 ± 0.1 mm; P < .0001). The medial layer wall thickness and elastin content were significantly greater in the EGCG group than in the control group on day 28 (68.4 ± 13.6 vs 46.7 ± 13.4 μm [P < .001] and 20.3 ± 4.6 vs 9.5 ± 3.6% [P < .0001], respectively). Gene expression levels of tropoelastin and lysyl oxidase were significantly higher in the EGCG group immediately before AAA induction, indicating promoted elastoregeneration by EGCG administration (tropoelastin: 0.59 ± 0.36 control vs 1.24 ± 0.36 EGCG [P < .05], lysyl oxidase: 0.77 ± 0.45 control vs 1.34 ± 0.4 EGCG [P < .05]) (fold increase). Gene expression levels of inflammatory cytokines, including tumor necrosis factor-α and interleukin-1β, were significantly downregulated in the EGCG group (1.82 ± 0.71 vs 0.97 ± 0.59 [P < .05] and 3.91 ± 3.24 vs 0.89 ± 0.59 [P < .05], respectively). On day 7, gene expression levels and gelatinolytic activity of matrix metalloproteinase 9 were significantly lower in the EGCG group (1.41 ± 0.86 vs 0.51 ± 0.42 [P < .05] and 1.00 ± 0.17 vs 0.29 ± 0.12 [P < .0001], respectively), whereas gene expression levels of tissue inhibitors of metalloproteinase-1 were significantly higher in the EGCG group (0.96 ± 0.11 vs 1.14 ± 0.09; P < .05). CONCLUSIONS EGCG attenuated AAA progression in a rat model by preserving the aortic thickness and elastin content of the medial layer through regeneration of elastin, as mediated by anti-inflammatory effects, and subsequent reduction of matrix metalloproteinase activity.
Collapse
|
20
|
Ponedel'kina IY, Gaskarova AR, Khaybrakhmanova EA, Lukina ES, Odinokov VN. Hyaluronic acid based hydroxamate and conjugates with biologically active amines: In vitro effect on matrix metalloproteinase-2. Carbohydr Polym 2016; 144:17-24. [PMID: 27083788 DOI: 10.1016/j.carbpol.2016.02.022] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2015] [Revised: 01/25/2016] [Accepted: 02/07/2016] [Indexed: 10/22/2022]
Abstract
In this study, water soluble hyaluronic acid (HA) based hydroxamate and conjugates with biologically active amines and hydrazides such as p- and o-aminophenols, anthranilic, 4- and 5-aminosalicylic acids, nicotinic, N-benzylnicotinic and isonicotinic hydrazides, p-aminobenzenesulfonamide (Streptocide), p-aminobenzoic acid diethylaminoethyl ester (Procaine), and 4-amino-2,3-dimethyl-1-phenyl-3-pyrazolin-5-one (4-aminoantipyrene) were examined as matrix metalloproteinase-2 inhibitors (MMPIs). In a dose of 0.27-270μM, the most efficient MMPIs were HA conjugates with o-aminophenol=4-aminoantipyrine>4-aminosalicylic acid>5-aminosalicylic acid. Conjugates with Streptocide, Procaine and HA hydroxamate showed 40-50% inhibitory effect at all used concentrations. Conjugates with anthranilic acid and isonicotinic hydrazide (Isoniazid) in a dose of 0.27μM inhibited enzyme activity by ∼70%, but with the concentration increase their inhibitory effect was decreased.
Collapse
Affiliation(s)
- Irina Yu Ponedel'kina
- Institute of Petrochemistry and Catalysis, Russian Academy of Sciences, Ufa 450075, Russian Federation.
| | - Aigul R Gaskarova
- Institute of Petrochemistry and Catalysis, Russian Academy of Sciences, Ufa 450075, Russian Federation
| | - Elvira A Khaybrakhmanova
- Institute of Petrochemistry and Catalysis, Russian Academy of Sciences, Ufa 450075, Russian Federation
| | - Elena S Lukina
- Institute of Petrochemistry and Catalysis, Russian Academy of Sciences, Ufa 450075, Russian Federation
| | - Victor N Odinokov
- Institute of Petrochemistry and Catalysis, Russian Academy of Sciences, Ufa 450075, Russian Federation
| |
Collapse
|
21
|
Tang L, Cong Z, Hao S, Li P, Huang H, Shen Y, Li K, Jing H. Protective effect of melatonin on the development of abdominal aortic aneurysm in a rat model. J Surg Res 2016; 209:266-278.e1. [PMID: 27392820 DOI: 10.1016/j.jss.2016.06.018] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2016] [Revised: 05/23/2016] [Accepted: 06/07/2016] [Indexed: 01/14/2023]
Abstract
BACKGROUND Oxidative injury, inflammation, and apoptosis are involved in the progression of abdominal aortic aneurysm (AAA). Melatonin (MLT) has been reported with an effective antioxidant activity. The objective of the present study was to investigate whether MLT could suppress the development of AAA. METHODS The AAA model was introduced by intraluminal perfusion of elastase in rats. All rats were divided into three groups as follows: (1) sham; (2) AAA + vehicle; and (3) AAA + MLT. Daily administration of MLT (10 mg/kg/d) or vehicle started 3 d before the perfusion and continued for 28 d after perfusion. An ultrasound system was applied to measure the dilation of the aorta. Histologic assays were performed to evaluate the structure, morphology, and apoptotic cells of the aortas; biochemical assays to determine the levels of proteins and lipid peroxide, activities of superoxide dismutase and NADPH oxidases, and cell viability; dihydroethidium fluorescence staining and flow cytometry to detect the presence of reactive oxygen species, and/or cell apoptosis; and electron microscopy to observe the ultrastructure of mitochondria. Cell lines A7R5 and RAW 264.7 were used for in vitro experiments. RESULTS MLT treatment inhibited dilation of the aorta very likely through its antioxidant property; significantly reduced the levels of lipid peroxide, activities of NADPH oxidases, and content of reactive oxygen species; remarkably inhibited NF-κB signaling pathway and activities of matrix metalloproteinases triggered by elastase perfusion. As a result, the mitochondrion-dependent apoptosis was suppressed, cellular energy (ATP) supply was recovered, and mitochondrial morphology remained intact. CONCLUSIONS Our results demonstrate the beneficial effects of MLT on inhibition of AAA formation, suggesting that MLT could be a potential agent for prevention of the development of human AAA.
Collapse
Affiliation(s)
- Li Tang
- Department of Cardiothoracic Surgery, Jinling Hospital, Medical School of Nanjing University, Nanjing, China
| | - Zhuangzhuang Cong
- Department of Cardiothoracic Surgery, Jinling Hospital, Medical School of Nanjing University, Nanjing, China
| | - Shuangying Hao
- Jiangsu Key Laboratory for Molecular Medicine, Medical School of Nanjing University, Nanjing, China
| | - Peng Li
- Department of Cardiothoracic Surgery, Jinling Hospital, Medical School of Nanjing University, Nanjing, China
| | - Hairong Huang
- Department of Cardiothoracic Surgery, Jinling Hospital, Medical School of Nanjing University, Nanjing, China
| | - Yi Shen
- Department of Cardiothoracic Surgery, Jinling Hospital, Medical School of Nanjing University, Nanjing, China.
| | - Kuanyu Li
- Jiangsu Key Laboratory for Molecular Medicine, Medical School of Nanjing University, Nanjing, China.
| | - Hua Jing
- Department of Cardiothoracic Surgery, Jinling Hospital, Medical School of Nanjing University, Nanjing, China.
| |
Collapse
|
22
|
Abstract
Abdominal aortic aneurysm (AAA) is a significant cause of mortality in older adults. A key mechanism implicated in AAA pathogenesis is inflammation and the associated production of reactive oxygen species (ROS) and oxidative stress. These have been suggested to promote degradation of the extracellular matrix (ECM) and vascular smooth muscle apoptosis. Experimental and human association studies suggest that ROS can be favourably modified to limit AAA formation and progression. In the present review, we discuss mechanisms potentially linking ROS to AAA pathogenesis and highlight potential treatment strategies targeting ROS. Currently, none of these strategies has been shown to be effective in clinical practice.
Collapse
|
23
|
Chin K, Wieslander C, Shi H, Balgobin S, Montoya TI, Yanagisawa H, Word RA. Pelvic Organ Support in Animals with Partial Loss of Fibulin-5 in the Vaginal Wall. PLoS One 2016; 11:e0152793. [PMID: 27124299 PMCID: PMC4849714 DOI: 10.1371/journal.pone.0152793] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2015] [Accepted: 03/18/2016] [Indexed: 01/15/2023] Open
Abstract
Compromise of elastic fiber integrity in connective tissues of the pelvic floor is most likely acquired through aging, childbirth-associated injury, and genetic susceptibility. Mouse models of pelvic organ prolapse demonstrate systemic deficiencies in proteins that affect elastogenesis. Prolapse, however, does not occur until several months after birth and is thereby acquired with age or after parturition. To determine the impact of compromised levels of fibulin-5 (Fbln5) during adulthood on pelvic organ support after parturition and elastase-induced injury, tissue-specific conditional knockout (cKO) mice were generated in which doxycycline (dox) treatment results in deletion of Fbln5 in cells that utilize the smooth muscle α actin promoter-driven reverse tetracycline transactivator and tetracycline responsive element-Cre recombinase (i.e., Fbln5f/f/SMA++-rtTA/Cre+, cKO). Fbln5 was decreased significantly in the vagina of cKO mice compared with dox-treated wild type or controls (Fbln5f/f/SMA++-rtTA/Cre-/-). In controls, perineal body length (PBL) and bulge increased significantly after delivery but declined to baseline values within 6-8 weeks. Although overt prolapse did not occur in cKO animals, these transient increases in PBL postpartum were amplified and, unlike controls, parturition-induced increases in PBL (and bulge) did not recover to baseline but remained significantly increased for 12 wks. This lack of recovery from parturition was associated with increased MMP-9 and nondetectable levels of Fbln5 in the postpartum vagina. This predisposition to prolapse was accentuated by injection of elastase into the vaginal wall in which overt prolapse occurred in cKO animals, but rarely in controls. Taken together, our model system in which Fbln5 is conditionally knock-downed in stromal cells of the pelvic floor results in animals that undergo normal elastogenesis during development but lose Fbln5 as adults. The results indicate that vaginal fibulin-5 during development is crucial for baseline pelvic organ support and is also important for protection and recovery from parturition- and elastase-induced prolapse.
Collapse
Affiliation(s)
- Kathleen Chin
- Department of Obstetrics and Gynecology, Division of Female Pelvic Medicine and Reconstructive Surgery, University of Texas Southwestern Medical Center, Dallas, Texas, United States of America
| | - Cecilia Wieslander
- Department of Obstetrics and Gynecology, Division of Female Pelvic Medicine and Reconstructive Surgery, University of Texas Southwestern Medical Center, Dallas, Texas, United States of America
| | - Haolin Shi
- Department of Obstetrics and Gynecology, Division of Female Pelvic Medicine and Reconstructive Surgery, University of Texas Southwestern Medical Center, Dallas, Texas, United States of America
| | - Sunil Balgobin
- Department of Obstetrics and Gynecology, Division of Female Pelvic Medicine and Reconstructive Surgery, University of Texas Southwestern Medical Center, Dallas, Texas, United States of America
| | - T. Ignacio Montoya
- Department of Obstetrics and Gynecology, Division of Female Pelvic Medicine and Reconstructive Surgery, University of Texas Southwestern Medical Center, Dallas, Texas, United States of America
| | - Hiromi Yanagisawa
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, Texas, United States of America
| | - R. Ann Word
- Department of Obstetrics and Gynecology, Division of Female Pelvic Medicine and Reconstructive Surgery, University of Texas Southwestern Medical Center, Dallas, Texas, United States of America
- * E-mail:
| |
Collapse
|
24
|
Glucagon-like peptide-1 prevented abdominal aortic aneurysm development in rats. Surg Today 2015; 46:1099-107. [PMID: 26658813 DOI: 10.1007/s00595-015-1287-z] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2015] [Accepted: 10/28/2015] [Indexed: 02/07/2023]
Abstract
PURPOSE To demonstrate the protective effect of glucagon-like peptide 1 (GLP-1) signaling on the cardiovascular system, we conducted this study to show that the GLP-1 receptor analog (lixisenatide) could inhibit abdominal aortic aneurysm (AAA) development in rats. METHODS Lixisenatide was injected subcutaneously 7 days after aneurysm preparation. We evaluated reactive oxygen species (ROS) expression by dihydroethidium staining and 8-hydroxydeoxyguanosine (8-OHdG; the oxidation product of DNA) by immunohistochemical staining. We also analyzed the effect of GLP-1 signaling on the inflammatory response. Histopathological examination was done on day 28, and the AAA dilatation ratio was calculated. RESULTS On day 14, ROS expression and 8-OHdG-positive cells in the aneurysm walls were seen to have been significantly decreased by lixisenatide treatment. Western blot analysis showed decreased ERK expression. There was significantly reduced tumor necrosis factor-α mRNA expression in the aneurysm walls and CD68-positive cell infiltration in the aneurysm walls. On day 28, it was evident that the lixisenatide had dramatically reduced aneurysm development in the rats. CONCLUSION GLP-1 elevation inhibits AAA development in rats through its anti-oxidant and anti-inflammatory effects. Thus, GLP-1 could be a potent pharmacological target for AAA treatment.
Collapse
|
25
|
Wang KC, Li YH, Shi GY, Tsai HW, Luo CY, Cheng MH, Ma CY, Hsu YY, Cheng TL, Chang BI, Lai CH, Wu HL. Membrane-Bound Thrombomodulin Regulates Macrophage Inflammation in Abdominal Aortic Aneurysm. Arterioscler Thromb Vasc Biol 2015; 35:2412-22. [DOI: 10.1161/atvbaha.115.305529] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2015] [Accepted: 08/18/2015] [Indexed: 01/30/2023]
Abstract
Objective—
Thrombomodulin (TM), a glycoprotein constitutively expressed in the endothelium, is well known for its anticoagulant and anti-inflammatory properties. Paradoxically, we recently found that monocytic membrane-bound TM (ie, endogenous TM expression in monocytes) triggers lipopolysaccharide- and gram-negative bacteria–induced inflammatory responses. However, the significance of membrane-bound TM in chronic sterile vascular inflammation and the development of abdominal aortic aneurysm (AAA) remains undetermined.
Approach and Results—
Implicating a potential role for membrane-bound TM in AAA, we found that TM signals were predominantly localized to macrophages and vascular smooth muscle cells in human aneurysm specimens. Characterization of the CaCl
2
-induced AAA in mice revealed that during aneurysm development, TM expression was mainly localized in infiltrating macrophages and vascular smooth muscle cells. To investigate the function of membrane-bound TM in vivo, transgenic mice with myeloid- (LysMcre/TM
flox/flox
) and vascular smooth muscle cell–specific (SM22-cre
tg
/TM
flox/flox
) TM ablation and their respective wild-type controls (TM
flox/flox
and SM22-cre
tg
/TM
+/+
) were generated. In the mouse CaCl
2
-induced AAA model, deficiency of myeloid TM, but not vascular smooth muscle cell TM, inhibited macrophage accumulation, attenuated proinflammatory cytokine and matrix metalloproteinase-9 production, and finally mitigated elastin destruction and aortic dilatation. In vitro TM-deficient monocytes/macrophages, versus TM wild-type counterparts, exhibited attenuation of proinflammatory mediator expression, adhesion to endothelial cells, and generation of reactive oxygen species. Consistently, myeloid TM–deficient hyperlipidemic mice (ApoE
−/−
/LysMcre/TM
flox/flox
) were resistant to AAA formation induced by angiotensin II infusion, along with reduced macrophage infiltration, suppressed matrix metalloproteinase activities, and diminished oxidative stress.
Conclusions—
Membrane-bound TM in macrophages plays an essential role in the development of AAA by enhancing proinflammatory mediator elaboration, macrophage recruitment, and oxidative stress.
Collapse
Affiliation(s)
- Kuan-Chieh Wang
- From the Department of Biochemistry and Molecular Biology (K.-C.W., G.-Y.S., M.-H.C., C.-Y.M., Y.-Y.H., B.-I.C., H.-L.W.), Institute of Basic Medical Sciences (K.-C.W.), Cardiovascular Research Center (K.-C.W., Y.-H.L., G.-Y.S., C.-Y.L., C.-Y.M., Y.-Y.H., B.-I.C., C.-H.L., H.-L.W.), Department of Internal Medicine (Y.-H.L.), Department of Pathology (H.-W.T.), and Department of Surgery (C.-Y.L., C.-H.L.), National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University,
| | - Yi-Heng Li
- From the Department of Biochemistry and Molecular Biology (K.-C.W., G.-Y.S., M.-H.C., C.-Y.M., Y.-Y.H., B.-I.C., H.-L.W.), Institute of Basic Medical Sciences (K.-C.W.), Cardiovascular Research Center (K.-C.W., Y.-H.L., G.-Y.S., C.-Y.L., C.-Y.M., Y.-Y.H., B.-I.C., C.-H.L., H.-L.W.), Department of Internal Medicine (Y.-H.L.), Department of Pathology (H.-W.T.), and Department of Surgery (C.-Y.L., C.-H.L.), National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University,
| | - Guey-Yueh Shi
- From the Department of Biochemistry and Molecular Biology (K.-C.W., G.-Y.S., M.-H.C., C.-Y.M., Y.-Y.H., B.-I.C., H.-L.W.), Institute of Basic Medical Sciences (K.-C.W.), Cardiovascular Research Center (K.-C.W., Y.-H.L., G.-Y.S., C.-Y.L., C.-Y.M., Y.-Y.H., B.-I.C., C.-H.L., H.-L.W.), Department of Internal Medicine (Y.-H.L.), Department of Pathology (H.-W.T.), and Department of Surgery (C.-Y.L., C.-H.L.), National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University,
| | - Hung-Wen Tsai
- From the Department of Biochemistry and Molecular Biology (K.-C.W., G.-Y.S., M.-H.C., C.-Y.M., Y.-Y.H., B.-I.C., H.-L.W.), Institute of Basic Medical Sciences (K.-C.W.), Cardiovascular Research Center (K.-C.W., Y.-H.L., G.-Y.S., C.-Y.L., C.-Y.M., Y.-Y.H., B.-I.C., C.-H.L., H.-L.W.), Department of Internal Medicine (Y.-H.L.), Department of Pathology (H.-W.T.), and Department of Surgery (C.-Y.L., C.-H.L.), National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University,
| | - Chawn-Yau Luo
- From the Department of Biochemistry and Molecular Biology (K.-C.W., G.-Y.S., M.-H.C., C.-Y.M., Y.-Y.H., B.-I.C., H.-L.W.), Institute of Basic Medical Sciences (K.-C.W.), Cardiovascular Research Center (K.-C.W., Y.-H.L., G.-Y.S., C.-Y.L., C.-Y.M., Y.-Y.H., B.-I.C., C.-H.L., H.-L.W.), Department of Internal Medicine (Y.-H.L.), Department of Pathology (H.-W.T.), and Department of Surgery (C.-Y.L., C.-H.L.), National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University,
| | - Min-Hua Cheng
- From the Department of Biochemistry and Molecular Biology (K.-C.W., G.-Y.S., M.-H.C., C.-Y.M., Y.-Y.H., B.-I.C., H.-L.W.), Institute of Basic Medical Sciences (K.-C.W.), Cardiovascular Research Center (K.-C.W., Y.-H.L., G.-Y.S., C.-Y.L., C.-Y.M., Y.-Y.H., B.-I.C., C.-H.L., H.-L.W.), Department of Internal Medicine (Y.-H.L.), Department of Pathology (H.-W.T.), and Department of Surgery (C.-Y.L., C.-H.L.), National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University,
| | - Chih-Yuan Ma
- From the Department of Biochemistry and Molecular Biology (K.-C.W., G.-Y.S., M.-H.C., C.-Y.M., Y.-Y.H., B.-I.C., H.-L.W.), Institute of Basic Medical Sciences (K.-C.W.), Cardiovascular Research Center (K.-C.W., Y.-H.L., G.-Y.S., C.-Y.L., C.-Y.M., Y.-Y.H., B.-I.C., C.-H.L., H.-L.W.), Department of Internal Medicine (Y.-H.L.), Department of Pathology (H.-W.T.), and Department of Surgery (C.-Y.L., C.-H.L.), National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University,
| | - Yun-Yan Hsu
- From the Department of Biochemistry and Molecular Biology (K.-C.W., G.-Y.S., M.-H.C., C.-Y.M., Y.-Y.H., B.-I.C., H.-L.W.), Institute of Basic Medical Sciences (K.-C.W.), Cardiovascular Research Center (K.-C.W., Y.-H.L., G.-Y.S., C.-Y.L., C.-Y.M., Y.-Y.H., B.-I.C., C.-H.L., H.-L.W.), Department of Internal Medicine (Y.-H.L.), Department of Pathology (H.-W.T.), and Department of Surgery (C.-Y.L., C.-H.L.), National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University,
| | - Tsung-Lin Cheng
- From the Department of Biochemistry and Molecular Biology (K.-C.W., G.-Y.S., M.-H.C., C.-Y.M., Y.-Y.H., B.-I.C., H.-L.W.), Institute of Basic Medical Sciences (K.-C.W.), Cardiovascular Research Center (K.-C.W., Y.-H.L., G.-Y.S., C.-Y.L., C.-Y.M., Y.-Y.H., B.-I.C., C.-H.L., H.-L.W.), Department of Internal Medicine (Y.-H.L.), Department of Pathology (H.-W.T.), and Department of Surgery (C.-Y.L., C.-H.L.), National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University,
| | - Bi-Ing Chang
- From the Department of Biochemistry and Molecular Biology (K.-C.W., G.-Y.S., M.-H.C., C.-Y.M., Y.-Y.H., B.-I.C., H.-L.W.), Institute of Basic Medical Sciences (K.-C.W.), Cardiovascular Research Center (K.-C.W., Y.-H.L., G.-Y.S., C.-Y.L., C.-Y.M., Y.-Y.H., B.-I.C., C.-H.L., H.-L.W.), Department of Internal Medicine (Y.-H.L.), Department of Pathology (H.-W.T.), and Department of Surgery (C.-Y.L., C.-H.L.), National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University,
| | - Chao-Han Lai
- From the Department of Biochemistry and Molecular Biology (K.-C.W., G.-Y.S., M.-H.C., C.-Y.M., Y.-Y.H., B.-I.C., H.-L.W.), Institute of Basic Medical Sciences (K.-C.W.), Cardiovascular Research Center (K.-C.W., Y.-H.L., G.-Y.S., C.-Y.L., C.-Y.M., Y.-Y.H., B.-I.C., C.-H.L., H.-L.W.), Department of Internal Medicine (Y.-H.L.), Department of Pathology (H.-W.T.), and Department of Surgery (C.-Y.L., C.-H.L.), National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University,
| | - Hua-Lin Wu
- From the Department of Biochemistry and Molecular Biology (K.-C.W., G.-Y.S., M.-H.C., C.-Y.M., Y.-Y.H., B.-I.C., H.-L.W.), Institute of Basic Medical Sciences (K.-C.W.), Cardiovascular Research Center (K.-C.W., Y.-H.L., G.-Y.S., C.-Y.L., C.-Y.M., Y.-Y.H., B.-I.C., C.-H.L., H.-L.W.), Department of Internal Medicine (Y.-H.L.), Department of Pathology (H.-W.T.), and Department of Surgery (C.-Y.L., C.-H.L.), National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University,
| |
Collapse
|
26
|
Menteşe U, Turan I, Usta S, Demir S, Koral Ö, Öztaş Menteşe S, Çavuşoğlu IG, Karahan SC, Alver A, Doğan OV, Aykan AÇ. Systemic oxidant/antioxidant balance in human abdominal aortic aneurysm. Perfusion 2015; 31:288-94. [DOI: 10.1177/0267659115598856] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
The aim of this study was to evaluate the oxidant-antioxidant balance in patients with abdominal aortic aneurysms (AAA). Forty-two consecutive patients with AAA and 46 control subjects were included. Total oxidant status (TOS) and total antioxidant status (TAS) levels were measured and the oxidative stress index (OSI) value determined. Serum TOS and OSI values in patients with AAA were higher than those in the controls (p<0.001, p<0.001, respectively). There was a positive correlation between abdominal aortic diameters, serum TOS levels (r=0.592, p<0.001) and OSI values (r=0.598, p<0.001). A cut-off value of 17.68 µmol H2O2 equivalent/L for TOS was associated with 86% sensitivity and 83% specificity and a cut-off value of 1.77 for OSI was associated with 86% sensitivity and 81% specificity for predicting AAA. Systemic oxidative imbalance develops in patients with AAA, particularly as a result of an increase in TOS.
Collapse
Affiliation(s)
- Umit Menteşe
- Department of Cardiovascular Surgery, Ahi Evren Chest and Cardiovascular Surgery Education and Research Hospital, Trabzon, Turkey
| | - Ibrahim Turan
- Department of Genetic and Bioengineering, Gümüşhane University, Faculty of Engineering and Natural Sciences, Gümüşhane, Turkey
| | - Sefer Usta
- Department of Cardiovascular Surgery, Ahi Evren Chest and Cardiovascular Surgery Education and Research Hospital, Trabzon, Turkey
| | - Selim Demir
- Department of Medical Biochemistry, Karadeniz Technical University, Faculty of Medicine, Trabzon, Turkey
| | - Özgür Koral
- Department of Cardiovascular Surgery, Ahi Evren Chest and Cardiovascular Surgery Education and Research Hospital, Trabzon, Turkey
| | - Seda Öztaş Menteşe
- Department of Emergency Medicine, Kanuni Teaching and Research Hospital, Trabzon, Turkey
| | - Ismail Gökhan Çavuşoğlu
- Department of Radiology, Ahi Evren Chest and Cardiovascular Surgery Education and Research Hospital, Trabzon, Turkey
| | - Süleyman Caner Karahan
- Department of Medical Biochemistry, Karadeniz Technical University, Faculty of Medicine, Trabzon, Turkey
| | - Ahmet Alver
- Department of Medical Biochemistry, Karadeniz Technical University, Faculty of Medicine, Trabzon, Turkey
| | - Orhan Veli Doğan
- Department of Cardiovascular Surgery, Sakarya University, Faculty of Medicine, Sakarya, Turkey
| | - Ahmet Çağrı Aykan
- Department of Cardiology, Ahi Evren Chest and Cardiovascular Surgery Education and Research Hospital, Trabzon, Turkey
| |
Collapse
|
27
|
Yu Z, Morimoto K, Yu J, Bao W, Okita Y, Okada K. Endogenous superoxide dismutase activation by oral administration of riboflavin reduces abdominal aortic aneurysm formation in rats. J Vasc Surg 2015; 64:737-45. [PMID: 26070605 DOI: 10.1016/j.jvs.2015.03.045] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2014] [Accepted: 03/01/2015] [Indexed: 12/20/2022]
Abstract
OBJECTIVE Vitamin B2 (riboflavin) reportedly has an antioxidant effect through superoxide dismutase (SOD) activation. However, the effect of riboflavin on abdominal aortic aneurysm (AAA) has never been investigated. In the present study, we examined the hypothesis that riboflavin has a protective effect on AAA formation in an experimental rat model. METHODS The AAA model, which was induced with intraluminal elastase and extraluminal calcium chloride, was created in 36 rats. The 36 rats were divided into a riboflavin group (group R; 25 mg/kg/d), and control group (carboxymethyl cellulose). Riboflavin administration by gastric gavage once per day was started at 3 days before aneurysm preparation. On day 3, SOD activity in aneurysm walls was assayed. On day 7, reactive oxygen species (ROS) levels were semiquantified by dihydroethidium staining, and the oxidation product of DNA produced by ROS, 8-hydroxydeoxyguanosine (8-OHdG), was measured by immunohistochemical staining. Histopathologic examination (hematoxylin/eosin and elastica Van Gieson staining) was performed on day 28, and the AAA dilatation ratio was calculated to evaluate the protective effect of riboflavin. RESULTS On day 3, SOD activity was significantly increased in aneurysm walls by riboflavin administration (370 ± 204 U/mL in normal, 334 ± 86 U/mL in control, 546 ± 143 U/mL in group R; P = .021). On day 7, ROS levels and 8-OHdG-positive cells in aneurysm walls were significantly decreased by riboflavin treatment (ROS levels: 1.0 ± 0.1 in normal, 4.5 ± 0.4 in control, 3.1 ± 0.5 in group R, P < .01; 8-OHdG-positive cells: 30 ± 2 cells in normal, 148 ± 20 cells in control, 109 ± 15 cells in group R, P < .01). Riboflavin treatment significantly reduced matrix metalloproteinase (MMP)-9 messenger RNA expression in aneurysm walls (relative expression: MMP-9: 0.4 ± 0.7 in normal, 2.6 ± 1.3 in control, 0.5 ± 0.3 in group R, P < .01). On day 28, the aortic walls were less dilated and had higher elastin content in group R than in control (dilatation ratio: 194.9% ± 10.9% in control, 158.6% ± 2.5% in group R; P <.01). CONCLUSIONS Riboflavin treatment prevents AAA formation in a rat model through an antioxidant effect and might be a potent pharmacologic agent for AAA treatment in clinical practice.
Collapse
MESH Headings
- 8-Hydroxy-2'-Deoxyguanosine
- Administration, Oral
- Animals
- Antioxidants/administration & dosage
- Aorta, Abdominal/drug effects
- Aorta, Abdominal/enzymology
- Aorta, Abdominal/pathology
- Aortic Aneurysm, Abdominal/chemically induced
- Aortic Aneurysm, Abdominal/enzymology
- Aortic Aneurysm, Abdominal/pathology
- Aortic Aneurysm, Abdominal/prevention & control
- Calcium Chloride
- DNA Damage
- Deoxyguanosine/analogs & derivatives
- Deoxyguanosine/metabolism
- Dilatation, Pathologic
- Disease Models, Animal
- Enzyme Activation
- Enzyme Activators/administration & dosage
- Extracellular Signal-Regulated MAP Kinases/metabolism
- Inflammation Mediators/metabolism
- Male
- Matrix Metalloproteinase 9/genetics
- Matrix Metalloproteinase 9/metabolism
- Oxidative Stress/drug effects
- Pancreatic Elastase
- Rats, Sprague-Dawley
- Reactive Oxygen Species/metabolism
- Riboflavin/administration & dosage
- Superoxide Dismutase/metabolism
- Time Factors
Collapse
Affiliation(s)
- Zhenhai Yu
- Division of Cardiovascular Surgery, Department of Surgery, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Keisuke Morimoto
- Division of Cardiovascular Surgery, Department of Surgery, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Jie Yu
- Division of Cardiovascular Surgery, Department of Surgery, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Wulan Bao
- Division of Cardiovascular Surgery, Department of Surgery, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Yutaka Okita
- Division of Cardiovascular Surgery, Department of Surgery, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Kenji Okada
- Division of Cardiovascular Surgery, Department of Surgery, Kobe University Graduate School of Medicine, Kobe, Japan; Division of Cardiovascular Surgery, Department of Surgery, Shinshu University School of Medicine, Matsumoto, Japan.
| |
Collapse
|
28
|
Tanaka A, Hasegawa T, Morimoto K, Bao W, Yu J, Okita Y, Tabata Y, Okada K. Controlled release of ascorbic acid from gelatin hydrogel attenuates abdominal aortic aneurysm formation in rat experimental abdominal aortic aneurysm model. J Vasc Surg 2014; 60:749-58. [DOI: 10.1016/j.jvs.2013.07.013] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2013] [Revised: 07/01/2013] [Accepted: 07/05/2013] [Indexed: 10/26/2022]
|
29
|
Yamamura M, Miyamoto Y, Mitsuno M, Tanaka H, Ryomoto M. Edaravone injected at the start of reperfusion suppresses myonephropathic metabolic syndrome in rats. Int J Angiol 2014; 23:193-6. [PMID: 25317032 PMCID: PMC4169098 DOI: 10.1055/s-0034-1387825] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022] Open
Abstract
The purpose of this study was to evaluate whether edaravone (Radicut(®), Mitsubishi Tanabe Pharma Co., Osaka, Japan) injected at the start of reperfusion can suppress myonephropathic-metabolic syndrome (MNMS). MNMS models were made by clamping the bilateral common femoral arteries for 5 hours. At de-clamping (at the start of reperfusion), they were intra-peritoneal injected with 9.0 mg/kg of edaravone (the edaravone group, n = 5) or an equal volume of saline (the control group, n = 5). At five hours after de-clamping, the lower extremity muscles were stained with hematoxylin & eosin (H&E) to count the viable cells, and periodic acid- Schiff (PAS) to assess the glycogen storage. The lungs were also stained with H&E to expresse the alveolar wall thickness, and naphthol AS-D chloroacetate esterase to label infiltrating active neutrophils. The viable muscle cells in the edaravone group was significantly greater than that of the control group (593 ± 60 vs. 258 ± 31 cells/mm(2), p < 0.01). The PAS-positive area in the edaravone group was also significantly higher than that in the control group (30.1 ± 6.9 vs. 7.3 ± 2.1%, p < 0.001). The alveolar wall thickness in the edaravone group was significantly lower than that in the control group (63.6 ± 5.6 vs. 17.2 ± 5.2%, p < 0.001). The active neutrophil infiltration in the edaravone group was also significantly lower than that in the control group (249 ± 59 vs. 68 ± 8 cells/mm(2), p < 0.001). We conclude that edaravone injected at the start of reperfusion can suppress not only muscle reperfusion injury but also lung damage.
Collapse
Affiliation(s)
- Mitsuhiro Yamamura
- Department of Cardiovascular Surgery, Hyogo College of Medicine, Nishinomiya, Hyogo, Japan
| | - Yuji Miyamoto
- Department of Cardiovascular Surgery, Hyogo College of Medicine, Nishinomiya, Hyogo, Japan
| | - Masataka Mitsuno
- Department of Cardiovascular Surgery, Hyogo College of Medicine, Nishinomiya, Hyogo, Japan
| | - Hiroe Tanaka
- Department of Cardiovascular Surgery, Hyogo College of Medicine, Nishinomiya, Hyogo, Japan
| | - Masaaki Ryomoto
- Department of Cardiovascular Surgery, Hyogo College of Medicine, Nishinomiya, Hyogo, Japan
| |
Collapse
|
30
|
Shang T, Liu Z, Liu CJ. Antioxidant Vitamin C attenuates experimental abdominal aortic aneurysm development in an elastase-induced rat model. J Surg Res 2014; 188:316-25. [DOI: 10.1016/j.jss.2013.11.1105] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2013] [Revised: 11/18/2013] [Accepted: 11/21/2013] [Indexed: 10/25/2022]
|
31
|
Huyard F, Yzydorczyk C, Castro MM, Cloutier A, Bertagnolli M, Sartelet H, Germain N, Comte B, Schulz R, DeBlois D, Nuyt AM. Remodeling of aorta extracellular matrix as a result of transient high oxygen exposure in newborn rats: implication for arterial rigidity and hypertension risk. PLoS One 2014; 9:e92287. [PMID: 24743169 PMCID: PMC3990546 DOI: 10.1371/journal.pone.0092287] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2013] [Accepted: 02/20/2014] [Indexed: 12/22/2022] Open
Abstract
Neonatal high-oxygen exposure leads to elevated blood pressure, microvascular rarefaction, vascular dysfunction and arterial (aorta) rigidity in adult rats. Whether structural changes are present in the matrix of aorta wall is unknown. Considering that elastin synthesis peaks in late fetal life in humans, and early postnatal life in rodents, we postulated that transient neonatal high-oxygen exposure can trigger premature vascular remodelling. Sprague Dawley rat pups were exposed from days 3 to 10 after birth to 80% oxygen (vs. room air control) and were studied at 4 weeks. Blood pressure and vasomotor response of the aorta to angiotensin II and to the acetylcholine analogue carbachol were not different between groups. Vascular superoxide anion production was similar between groups. There was no difference between groups in aortic cross sectional area, smooth muscle cell number or media/lumen ratio. In oxygen-exposed rats, aorta elastin/collagen content ratio was significantly decreased, the expression of elastinolytic cathepsin S was increased whereas collagenolytic cathepsin K was decreased. By immunofluorescence we observed an increase in MMP-2 and TIMP-1 staining in aortas of oxygen-exposed rats whereas TIMP-2 staining was reduced, indicating a shift in the balance towards degradation of the extra-cellular matrix and increased deposition of collagen. There was no significant difference in MMP-2 activity between groups as determined by gelatin zymography. Overall, these findings indicate that transient neonatal high oxygen exposure leads to vascular wall alterations (decreased elastin/collagen ratio and a shift in the balance towards increased deposition of collagen) which are associated with increased rigidity. Importantly, these changes are present prior to the elevation of blood pressure and vascular dysfunction in this model, and may therefore be contributory.
Collapse
Affiliation(s)
- Fanny Huyard
- Sainte-Justine University Hospital Research Center, Department of Paediatrics, Université de Montréal, Montreal, Québec, Canada
| | - Catherine Yzydorczyk
- Sainte-Justine University Hospital Research Center, Department of Paediatrics, Université de Montréal, Montreal, Québec, Canada
| | - Michele M. Castro
- Departments of Pediatrics & Pharmacology, Cardiovascular Research Centre, University of Alberta, Edmonton, Alberta, Canada
| | - Anik Cloutier
- Sainte-Justine University Hospital Research Center, Department of Paediatrics, Université de Montréal, Montreal, Québec, Canada
| | - Mariane Bertagnolli
- Sainte-Justine University Hospital Research Center, Department of Paediatrics, Université de Montréal, Montreal, Québec, Canada
| | - Hervé Sartelet
- Sainte-Justine University Hospital Research Center, Department of Pathology, Université de Montréal, Montreal, Québec, Canada
| | - Nathalie Germain
- Sainte-Justine University Hospital Research Center, Department of Paediatrics, Université de Montréal, Montreal, Québec, Canada
| | - Blandine Comte
- Unit of Human Nutrition UMR 1019, INRA, Research Centre of Clermont-Ferrand/Theix, Saint-Genès-Champanelle, France
| | - Richard Schulz
- Departments of Pediatrics & Pharmacology, Cardiovascular Research Centre, University of Alberta, Edmonton, Alberta, Canada
| | - Denis DeBlois
- Faculty of Pharmacy, Université de Montréal, Montreal, Quebec, Canada
| | - Anne Monique Nuyt
- Sainte-Justine University Hospital Research Center, Department of Paediatrics, Université de Montréal, Montreal, Québec, Canada
- * E-mail:
| |
Collapse
|
32
|
Delbosc S, Diallo D, Dejouvencel T, Lamiral Z, Louedec L, Martin-Ventura JL, Rossignol P, Leseche G, Michel JB, Meilhac O. Impaired high-density lipoprotein anti-oxidant capacity in human abdominal aortic aneurysm. Cardiovasc Res 2013; 100:307-15. [PMID: 23955602 DOI: 10.1093/cvr/cvt194] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
AIMS Abdominal aortic aneurysm (AAA) is a particular form of atherothrombotic disease characterized by the dilation of the aortic wall and the presence of an intraluminal thrombus (ILT). The objective of the present study was to evaluate the pro-oxidant properties of the ILT and to characterize the anti-oxidant capacity of high-density lipoproteins (HDLs). METHODS AND RESULTS Our results show that ILT, adventitia, and plasma from AAA patients contained high concentrations of lipid and protein oxidation products. Mediators produced within or released by the thrombus and the adventitia were shown to induce reactive oxygen species (ROS) production by cultured aortic smooth muscle cells (AoSMCs) and to trigger the onset of apoptosis (an increase in mitochondrial membrane potential). Iron chelation limited these effects. Both concentration and functionality of HDLs were altered in AAA patients. Plasma levels of Apo A-I were lower, and small HDL subclasses were decreased in AAA patients. Circulating HDLs in AAA patients displayed an impaired capacity to inhibit copper-induced low-density lipoprotein oxidation and AoSMC ROS production. Western blot analyses of HDLs demonstrated that myeloperoxidase is associated with HDL particles in AAA patients. CONCLUSION ILT and adventitia are a source of pro-oxidant products, in particular haemoglobin, which may impact on the wall stability/rupture in AAA. In addition, HDLs from AAA patients exhibit an impaired anti-oxidant activity. In this context, restoring HDL functionality may represent a new therapeutic option in AAA.
Collapse
Affiliation(s)
- Sandrine Delbosc
- INSERM U698, Hemostasis, Bio-engineering and Cardiovascular Remodeling, Hôpital Bichat, 46 Rue Henri Huchard, 75018 Paris, France
| | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Bao W, Morimoto K, Hasegawa T, Sasaki N, Yamashita T, Hirata K, Okita Y, Okada K. Orally administered dipeptidyl peptidase-4 inhibitor (alogliptin) prevents abdominal aortic aneurysm formation through an antioxidant effect in rats. J Vasc Surg 2013; 59:1098-108. [PMID: 23790558 DOI: 10.1016/j.jvs.2013.04.048] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2012] [Revised: 04/17/2013] [Accepted: 04/19/2013] [Indexed: 11/17/2022]
Abstract
OBJECTIVE Dipeptidyl peptidase-4 (DPP-4) inhibitor, a novel antidiabetic drug, has a cardioprotective effect on ischemia-reperfusion injury through an antioxidant effect. However, the effect of DPP-4 inhibitor on aneurysm formation has not been investigated. We aimed to test the hypothesis that the DPP-4 inhibitor, alogliptin, attenuates vascular oxidative stress and thus inhibits abdominal aortic aneurysm (AAA) formation. METHODS AAAs were created with intraluminal elastase and extraluminal calcium chloride in 36 male rats. Rats were divided into three groups: a low dose of alogliptin group (group LD; 1 mg/kg/d), a high-dose group (group HD; 3 mg/kg/d), and a control group (group C, water). Alogliptin was administered by gastric gavage once daily beginning 3 days before surgery. On day 7 after aneurysm preparation, reactive oxygen species (ROS) expression was semiquantified by dihydroethidium staining, and the oxidation product of DNA produced by ROS, 8-hydroxydeoxyguanosine (8-OHdG), was measured by immunohistochemical staining. Blood glucose concentrations were measured. Hematoxylin and eosin and elastica Van Gieson stainings were performed on day 28, and the AAA dilatation ratio was calculated. RESULTS On day 7 (six in each group), dihydroethidium staining of the aneurysm wall showed a reduced level of ROS expression (4.6 ± 0.6 in group C, 2.7 ± 0.3 in group LD, and 1.7 ± 0.5 in group HD; P < .0001) and showed fewer 8-OHdG-positive cells in alogliptin-treated samples (138.1 ± 7.4 cells in group C, 102.5 ± 4.5 cells in group LD, and 66.1 ± 4.5 cells in group HD; P < .0001) The treatment significantly reduced messenger RNA expression of matrix metalloproteinases (MMPs) in aneurysm walls (relative expression: MMP-2: 2.1 ± 0.4 in group C, 1.3 ± 0.3 in group LD, and 0.9 ± 0.2 in group HD; P < .001; MMP-9: 2.0 ± 0.5 in group C, 0.3 ± 0.3 in group LD, and 0.3 ± 0.2 in group HD; P < .001). On day 28 (six in each group), the aortic wall in groups LD and HD was less dilated (dilatation ratio: 199.2% ± 11.8% in group C, 159.6% ± 2.8% in group LD, and 147.1% ± 1.9% in group HD; P < .02 group C vs HD) and had higher elastin content than in group C. The difference in blood glucose levels among the three groups was not significant. CONCLUSIONS The DPP-4 inhibitor, alogliptin, attenuates aneurysm formation and expansion dose-dependently in a rat AAA model via an antioxidative action.
Collapse
Affiliation(s)
- Wulan Bao
- Division of Cardiovascular Surgery, Department of Surgery, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Keisuke Morimoto
- Division of Cardiovascular Surgery, Department of Surgery, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Tomomi Hasegawa
- Division of Cardiovascular Surgery, Department of Surgery, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Naoto Sasaki
- Division of Cardiovascular Medicine, Department of Internal Medicine, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Tomoya Yamashita
- Division of Cardiovascular Medicine, Department of Internal Medicine, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Kenichi Hirata
- Division of Cardiovascular Medicine, Department of Internal Medicine, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Yutaka Okita
- Division of Cardiovascular Surgery, Department of Surgery, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Kenji Okada
- Division of Cardiovascular Surgery, Department of Surgery, Kobe University Graduate School of Medicine, Kobe, Japan.
| |
Collapse
|
34
|
Transforming growth factor-β1 induces matrix metalloproteinase-9 expression in rat vascular smooth muscle cells via ROS-dependent ERK-NF-κB pathways. Mol Cell Biochem 2012; 375:11-21. [PMID: 23275087 DOI: 10.1007/s11010-012-1512-7] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2012] [Accepted: 11/07/2012] [Indexed: 02/07/2023]
Abstract
Both matrix metalloproteinase-9 (MMP9) and transforming growth factors-β1 (TGF-β1) are the important factors in the pathogenesis of the aortic aneurysm (AA) and aortic dissection (AD). Recent studies have shown that inhibition of reactive oxygen species (ROS) production, extracellular signal-regulated kinase 1/2(ERK1/2) or NF-κB pathways is able to suppress aneurysm formation. The median layers of arterial walls are mainly the vascular smooth muscle cells (VSMCs), while the pathogenesis of AA and AD is closely related to the changes in the median layer structure. Thus, we investigated the molecular mechanisms underlying TGF-β1-induced MMP-9 expression in VSMC, the involvement of intracellular ROS and signaling molecules, including ERK1/2 and NF-κB. Rat vascular smooth muscle cells (A7r5) were used. MMP-9 expression was analyzed by gelatin zymography, western blot and RT-PCR. The involvement of intracellular ROS and signaling molecules including ERK1/2 and NF-κB in the responses was investigated using reactive oxygen scavenger N-acetylcysteine (NAC) and pharmacological inhibitors (U0126 and BAY11-7082), determined by ROS testing and western blot testing for their corresponding proteins. TGF-β1 induces MMP-9 expression via ROS-dependent signaling pathway. ROS production leads to activation of ERK1/2 and then activation of the NF-κB transcription factor. Activated NF-κB turns on transcription of the MMP-9 gene. The process in which TGF-β1 induces MMP9 expression involves the ROS-dependent ERK-NF-κB signal pathways in VSMC. This discovery raises a new regulation pathway in the VSMC, and it shows the potential to help to find a new solution to treating aortic aneurysm and aortic dissection.
Collapse
|
35
|
The calcium chloride-induced rodent model of abdominal aortic aneurysm. Atherosclerosis 2012; 226:29-39. [PMID: 23044097 DOI: 10.1016/j.atherosclerosis.2012.09.010] [Citation(s) in RCA: 80] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/27/2012] [Revised: 09/01/2012] [Accepted: 09/14/2012] [Indexed: 11/22/2022]
Abstract
Abdominal aortic aneurysm (AAA) affects ∼5% men aged over 65 years and is an important cause of death in this population. Research into AAA pathogenesis has been fuelled by the need to identify new diagnostic biomarkers and therapeutic targets for this disease. One animal model of AAA involves peri-vascular application of calcium chloride (CaCl(2)) onto the infra-renal aorta of mice and rats to induce extracellular matrix remodelling. Twenty-three studies assessing CaCl(2)-induced AAA and six studies assessing AAA induced by a modified CaCl(2) method were identified. In the current report the preparation and pathological features of this AAA model are discussed. We also compared this animal model to human AAA. CaCl(2)-induced AAA shows the following pathological characteristics typically found in human AAA: calcification, inflammatory cell infiltration, oxidative stress, neovascularisation, elastin degradation and vascular smooth muscle cell apoptosis. A number of mechanisms involved in CaCl(2)-induced AAA have been identified which may be relevant to the pathogenesis of human AAA. Key molecules include c-Jun N-terminal kinase, peroxisome proliferator-activated receptor-γ, chemokine (C-C motif) receptor 2, group x secretory phospholipase A2 and plasminogen. CaCl(2)-induced AAA does not display aortic thrombus, atherosclerosis and rupture which are classical features of human AAA. Advantages of the CaCl(2)-induced AAA technique include (1) it can be applied to wild type mice making assessment of transgenic rodent models more straight forward and rapid; and (2) CaCl(2)-induced AAAs are usually developed in the infra-renal abdominal aorta, which is the most common location of human AAA. Currently findings obtained from the CaCl(2)-induced AAA model or other animal models of AAA have not been translated into the human situation. It is hoped that this deficiency will be corrected over the next decade with a number of clinical trials currently examining novel treatment options for AAA patients.
Collapse
|