1
|
Kim MH, Cho HJ, Ko KJ, Jun KW, Han KD, Hwang JK. Increased incidence of abdominal aortic aneurysm in women with early menopause. J Vasc Surg 2025:S0741-5214(25)00994-2. [PMID: 40306597 DOI: 10.1016/j.jvs.2025.04.034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2025] [Revised: 04/15/2025] [Accepted: 04/19/2025] [Indexed: 05/02/2025]
Abstract
OBJECTIVE In several countries, including the United States and United Kingdom, screening tests are conducted on men 65 years of age or older who smoke to diagnose abdominal aortic aneurysm (AAA) before rupture. In women, however, the incidence of AAA is low, so screening tests are not cost-effective and therefore not recommended. Early detection and treatment of AAA are important for women as they face a four-times-higher risk of rupture and worse surgery outcomes compared with men. Accordingly, we analyzed how differences in the period of exposure to female hormones affect women's AAA risk to find unique risk factors. METHODS We collected data from the National Health Insurance System database for women aged 40 or older who underwent medical checkups and female cancer tests from January to December 2009 (n=3,109,506). Those who were menopausal (n=1,393,271) were included in the study. Participants were tracked until December 2019 to confirm whether AAA was diagnosed. RESULTS There were 3,629 cases diagnosed with AAA. A comparison of the AAA and non-AAA cohorts showed a decreasing trend with increasing age at menopause. Compared with women who reached menopause before the age of 40, those who reached menopause after 55 had a 23% lower risk of AAA. Those who experienced over 40 years of menstruation had a 20% lower risk of AAA compared with fewer than 30 years of menstruation. CONCLUSIONS Women with early menopause face a higher incidence of AAA, so attention should be paid to early diagnosis.
Collapse
Affiliation(s)
- Mi-Hyeong Kim
- Division of Vascular and Transplant Surgery, Department of Surgery, Eunpyeong St. Mary`s Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Hyung-Jin Cho
- Division of Vascular and Transplant Surgery, Department of Surgery, Eunpyeong St. Mary`s Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Kyung-Jai Ko
- Department of Surgery, Kangdong Sacred Heart Hospital, Seoul, Korea
| | - Kang-Woong Jun
- Division of Vascular and Transplant Surgery, Department of Surgery, Bucheon St. Mary`s Hospital, College of Medicine, The Catholic University of Korea, Bucheon, Gyeonggi-do, Korea
| | - Kyung-do Han
- Department of Statistics and Actuarial Science, Soongsil University, Seoul, Korea
| | - Jeong-Kye Hwang
- Division of Vascular and Transplant Surgery, Department of Surgery, Eunpyeong St. Mary`s Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea.
| |
Collapse
|
2
|
Zhang RL, Wang WM, Li JQ, Li RW, Zhang J, Wu Y, Liu Y. The role of miR-155 in cardiovascular diseases: Potential diagnostic and therapeutic targets. INTERNATIONAL JOURNAL OF CARDIOLOGY. CARDIOVASCULAR RISK AND PREVENTION 2025; 24:200355. [PMID: 39760132 PMCID: PMC11699627 DOI: 10.1016/j.ijcrp.2024.200355] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/19/2024] [Revised: 11/21/2024] [Accepted: 12/05/2024] [Indexed: 01/07/2025]
Abstract
Cardiovascular diseases (CVDs), such as atherosclerotic cardiovascular diseases, heart failure (HF), and acute coronary syndrome, represent a significant threat to global health and impose considerable socioeconomic burdens. The intricate pathogenesis of CVD involves various regulatory mechanisms, among which microRNAs (miRNAs) have emerged as critical posttranscriptional regulators. In particular, miR-155 has demonstrated differential expression patterns across a spectrum of CVD and is implicated in the etiology and progression of arterial disorders. This systematic review synthesizes current evidence on the multifaceted roles of miR-155 in the modulation of genes and pathological processes associated with CVD. We delineate the potential of miR-155 as a diagnostic biomarker and therapeutic target, highlighting its significant regulatory influence on conditions such as atherosclerosis, aneurysm, hypertension, HF, myocardial hypertrophy, and oxidative stress. Our analysis underscores the transformative potential of miR-155 as a target for intervention in cardiovascular medicine, warranting further investigation into its clinical applicability.
Collapse
Affiliation(s)
- Rui-Lin Zhang
- Department of Vascular Surgery, The Affiliated Hospital, Southwest Medical University, Luzhou, 646000, China
| | - Wei-Ming Wang
- Department of Vascular Surgery, The Affiliated Hospital, Southwest Medical University, Luzhou, 646000, China
- Metabolic Vascular Disease Key Laboratory of Sichuan Province, The Affiliated Hospital, Southwest Medical University, 646000, Luzhou, China
- Key Laboratory of Medical Electrophysiology, Ministry of Education & Medical Electrophysiological Key Laboratory of Sichuan Province, (Collaborative Innovation Center for Prevention of Cardiovascular Diseases) Institute of Cardiovascular Research, Southwest Medical University, Luzhou, 646000, China
- Department of General Surgery, The Affiliated Hospital, Southwest Medical University, Luzhou, 646000, China
| | - Ji-Qiang Li
- Department of Vascular Surgery, The Affiliated Hospital, Southwest Medical University, Luzhou, 646000, China
| | - Run-Wen Li
- Department of Vascular Surgery, The Affiliated Hospital, Southwest Medical University, Luzhou, 646000, China
| | - Jie Zhang
- Department of Vascular Surgery, The Affiliated Hospital, Southwest Medical University, Luzhou, 646000, China
| | - Ya Wu
- Department of Vascular Surgery, The Affiliated Hospital, Southwest Medical University, Luzhou, 646000, China
- Metabolic Vascular Disease Key Laboratory of Sichuan Province, The Affiliated Hospital, Southwest Medical University, 646000, Luzhou, China
- Key Laboratory of Medical Electrophysiology, Ministry of Education & Medical Electrophysiological Key Laboratory of Sichuan Province, (Collaborative Innovation Center for Prevention of Cardiovascular Diseases) Institute of Cardiovascular Research, Southwest Medical University, Luzhou, 646000, China
- Department of General Surgery, The Affiliated Hospital, Southwest Medical University, Luzhou, 646000, China
| | - Yong Liu
- Department of Vascular Surgery, The Affiliated Hospital, Southwest Medical University, Luzhou, 646000, China
- Metabolic Vascular Disease Key Laboratory of Sichuan Province, The Affiliated Hospital, Southwest Medical University, 646000, Luzhou, China
- Key Laboratory of Medical Electrophysiology, Ministry of Education & Medical Electrophysiological Key Laboratory of Sichuan Province, (Collaborative Innovation Center for Prevention of Cardiovascular Diseases) Institute of Cardiovascular Research, Southwest Medical University, Luzhou, 646000, China
- Department of General Surgery, The Affiliated Hospital, Southwest Medical University, Luzhou, 646000, China
| |
Collapse
|
3
|
Atkinson G, Bianco R, Di Gregoli K, Johnson JL. The contribution of matrix metalloproteinases and their inhibitors to the development, progression, and rupture of abdominal aortic aneurysms. Front Cardiovasc Med 2023; 10:1248561. [PMID: 37799778 PMCID: PMC10549934 DOI: 10.3389/fcvm.2023.1248561] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Accepted: 09/07/2023] [Indexed: 10/07/2023] Open
Abstract
Abdominal aortic aneurysms (AAAs) account for up to 8% of deaths in men aged 65 years and over and 2.2% of women. Patients with AAAs often have atherosclerosis, and intimal atherosclerosis is generally present in AAAs. Accordingly, AAAs are considered a form of atherosclerosis and are frequently referred to as atherosclerotic aneurysms. Pathological observations advocate inflammatory cell infiltration alongside adverse extracellular matrix degradation as key contributing factors to the formation of human atherosclerotic AAAs. Therefore, macrophage production of proteolytic enzymes is deemed responsible for the damaging loss of ECM proteins, especially elastin and fibrillar collagens, which characterise AAA progression and rupture. Matrix metalloproteinases (MMPs) and their regulation by tissue inhibitors metalloproteinases (TIMPs) can orchestrate not only ECM remodelling, but also moderate the proliferation, migration, and apoptosis of resident aortic cells, alongside the recruitment and subsequent behaviour of inflammatory cells. Accordingly, MMPs are thought to play a central regulatory role in the development, progression, and eventual rupture of abdominal aortic aneurysms (AAAs). Together, clinical and animal studies have shed light on the complex and often diverse effects MMPs and TIMPs impart during the development of AAAs. This dichotomy is underlined from evidence utilising broad-spectrum MMP inhibition in animal models and clinical trials which have failed to provide consistent protection from AAA progression, although more encouraging results have been observed through deployment of selective inhibitors. This review provides a summary of the supporting evidence connecting the contribution of individual MMPs to AAA development, progression, and eventual rupture. Topics discussed include structural, functional, and cell-specific diversity of MMP members; evidence from animal models of AAA and comparisons with findings in humans; the dual role of MMPs and the requirement to selectively target individual MMPs; and the advances in identifying aberrant MMP activity. As evidenced, our developing understanding of the multifaceted roles individual MMPs perform during the progression and rupture of AAAs, should motivate clinical trials assessing the therapeutic potential of selective MMP inhibitors, which could restrict AAA-related morbidity and mortality worldwide.
Collapse
Affiliation(s)
| | | | | | - Jason L. Johnson
- Laboratory of Cardiovascular Pathology, Department of Translational Health Sciences, Bristol Medical School, University of Bristol, Bristol, United Kingdom
| |
Collapse
|
4
|
Yang L, Sui HG, Wang MM, Li JY, He XF, Li JY, Wang XZ. MiR-30c-1-3p targets matrix metalloproteinase 9 involved in the rupture of abdominal aortic aneurysms. J Mol Med (Berl) 2022; 100:1209-1221. [PMID: 35840740 PMCID: PMC9329399 DOI: 10.1007/s00109-022-02230-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Revised: 06/23/2022] [Accepted: 06/27/2022] [Indexed: 11/26/2022]
Abstract
Abstract Abdominal aortic aneurysm (AAA) can be fatal if ruptured, but there is no predictive biomarker. Our aim was to evaluate the prognostic potential of microRNAs (miRNAs/miRs) in an AAA mouse model and patients with unruptured AAA (URAAA) and ruptured AAA (RAAA). Among the 64 miRNAs differentially expressed in mice with AAA compared to control, miR-30c-1-3p, miR-432-3p, miR-3154, and miR-379-5p had high homology with human miRNAs. MiR-30c-1-3p plasma levels were significantly lower in patients with RAAA than in those with URAAA or control and tended to negatively correlate with the maximum aortic diameter (r = −0.3153, P = 0.06109). MiR-30c-1-3p targeted matrix metalloproteinase (MMP)-9 mRNA through the coding region and downregulated its expression in vitro. MMP-9 plasma concentrations were significantly higher in the RAAA group than in the URAAA group (P < 0.001) and were negatively associated with miR-30c-1-3p levels (r = −0.3671, P = 0.01981) and positively–with the maximal aortic diameter (r = 0.6251, P < 0.0001). The optimal cutoff values for MMP-9 expression and the maximal aortic diameter were 461.08 ng/ml and 55.95 mm, with areas under the curve of 0.816 and 0.844, respectively. Our results indicate that plasma levels of miR-30c-1-3p and MMP-9 may be candidate biomarkers of AAA progression. Key messages Downregulation of miR-30c-1-3p expression and upregulation of its potential target MMP-9 are predictors of the devastation of AAA. Downregulation of miR-30c-1-3p expression and its downstream impact on MMP-9 have a potential on predicting the development and rupture of AAA.
Supplementary Information The online version contains supplementary material available at 10.1007/s00109-022-02230-2.
Collapse
Affiliation(s)
- Lin Yang
- College of Medicine and Biological Information Engineering, Northeastern University, Shenyang, Liaoning, 110167, China
- Department of Cardiology and Cardiovascular Research Institute, General Hospital of Northern Theater Command, 83 Wenhua Road, Shenyang, 110016, Liaoning, China
| | - Hong-Gang Sui
- Department of Cardiology and Cardiovascular Research Institute, General Hospital of Northern Theater Command, 83 Wenhua Road, Shenyang, 110016, Liaoning, China
| | - Meng-Meng Wang
- Department of Cardiology and Cardiovascular Research Institute, General Hospital of Northern Theater Command, 83 Wenhua Road, Shenyang, 110016, Liaoning, China
| | - Jia-Yin Li
- College of Medicine and Biological Information Engineering, Northeastern University, Shenyang, Liaoning, 110167, China
- Department of Cardiology and Cardiovascular Research Institute, General Hospital of Northern Theater Command, 83 Wenhua Road, Shenyang, 110016, Liaoning, China
| | - Xiao-Feng He
- Department of Cardiology and Cardiovascular Research Institute, General Hospital of Northern Theater Command, 83 Wenhua Road, Shenyang, 110016, Liaoning, China
| | - Jing-Yuan Li
- Department of Cardiology and Cardiovascular Research Institute, General Hospital of Northern Theater Command, 83 Wenhua Road, Shenyang, 110016, Liaoning, China
| | - Xiao-Zeng Wang
- College of Medicine and Biological Information Engineering, Northeastern University, Shenyang, Liaoning, 110167, China.
- Department of Cardiology and Cardiovascular Research Institute, General Hospital of Northern Theater Command, 83 Wenhua Road, Shenyang, 110016, Liaoning, China.
| |
Collapse
|
5
|
Boer GJ, Schröder LBW, Disli MC, Kuijper TM, van de Luijtgaarden KM, Fioole B. A stable aneurysm sac after endovascular aneurysm repair as a predictor for mortality, an in-depth analysis. J Vasc Surg 2022; 76:445-453. [PMID: 35149164 DOI: 10.1016/j.jvs.2022.01.125] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Accepted: 01/21/2022] [Indexed: 11/26/2022]
Abstract
OBJECTIVES The aim of this study was to compare the long-term survival in patients with a stable aneurysmal sac and those with aneurysmal sac regression after endovascular aneurysm repair (EVAR) and to identify independent risk factors for aneurysmal sac regression and mortality after EVAR. METHODS We reviewed all patients who underwent EVAR between 2005 and 2018 with a computed tomography angiography available at 1-year follow-up. Aneurysm sac regression was defined as a diameter decrease of more than 10%. We used a multivariable regression to identify independent risk factors for sac regression. Kaplan-Meier analysis and Cox regression were done to test differences in 5-year mortality between a stable sac and sac regression. RESULTS The inclusion criteria were met by 325 patients, 185 in the sac regression group and 140 in the stable sac group. Multivariable logistic regression analysis showed that treatment for a ruptured aneurysm was an independent risk factor for aneurysmal sac regression (hazard ratio [HR], 0.26; 95% confidence interval [CI], 0.07-0.96). Age (HR, 1.05; 95% CI, 1.01-1.09), ischemic heart disease (HR, 1.94; 95% CI, 1.13-3.31), neck thrombus (HR, 2.72; 95% CI, 1.07-6.95), and a type II endoleak (HR, 19.21; 95% CI, 7.32-50.40) were independent risk factors for a stable aneurysm sac diameter. Multivariable Cox regression showed a significantly increased risk of mortality for patients with a stable aneurysm sac after EVAR (odds ratio, 2.25; 95% CI, 1.36-3.72). There was no significant difference in cause of death between the two groups. CONCLUSION A stable aneurysm sac after EVAR is associated with increased mortality. Age, ischemic heart disease, neck thrombus, and a type II endoleak are independent risk factors for a stable sac. However, a well-founded explanation for this finding is still lacking. Future research should be focussed on aggressive treatment of type II endoleaks and inflammatory processes as potential pathophysiological mechanisms.
Collapse
Affiliation(s)
- Gert Jan Boer
- Department of Vascular Surgery, Maasstad Hospital, Rotterdam, the Netherlands.
| | | | - Maksud C Disli
- Department of Vascular Surgery, Maasstad Hospital, Rotterdam, the Netherlands
| | - Tjallingius M Kuijper
- Clinical epidemiologist and statistician, Maasstad Academy, Maasstad Hospital, Rotterdam, the Netherlands
| | | | - Bram Fioole
- Department of Vascular Surgery, Maasstad Hospital, Rotterdam, the Netherlands
| |
Collapse
|
6
|
Fang L, Ohashi K, Otaka N, Ogawa H, Hiramatsu-Ito M, Kawanishi H, Bando YK, Shibata R, Shimizu Y, Kato K, Takikawa T, Ozaki Y, Takefuji M, Murohara T, Ouchi N. Omentin attenuates angiotensin II-induced abdominal aortic aneurysm formation in apolipoprotein-E knockout mice. Cardiovasc Res 2021; 118:1597-1610. [PMID: 34051089 DOI: 10.1093/cvr/cvab179] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Accepted: 05/28/2021] [Indexed: 01/28/2023] Open
Abstract
AIMS Abdominal aortic aneurysm (AAA) is an increasing and life-threatening disease. Obesity contributes to an increased risk of AAA. Omentin is a circulating adipokine, which is downregulated in obese complications. Here we examined whether omentin could modulate angiotensin (Ang) II-induced AAA formation in apolipoprotein-E knockout (apoE-KO) mice. METHODS AND RESULTS ApoE-KO mice were crossed with transgenic mice expressing the human omentin gene in fat tissue (OMT-Tg mice) to generate ApoE-KO/OMT-Tg mice. ApoE-KO/OMT-Tg and apoE-KO mice were subjected to continuous Ang II infusion by using osmotic mini pumps. ApoE-KO/OMT-Tg mice exhibited a lower incidence of AAA formation and a reduced maximal diameter of AAA compared with apo-E KO mice. ApoE-KO/OMT-Tg mice showed attenuated disruption of medial elastic fibers in response to Ang II compared with apo-E KO mice. ApoE-KO/OMT-Tg mice also displayed reduced expression levels of matrix metalloproteinase (MMP) 9, MMP2 and pro-inflammatory genes in aortic walls compared with apo-E KO mice. Furthermore, systemic administration of omentin also attenuated AAA formation and disruption of medial elastic fibers in response to Ang II in apoE-KO mice. Treatment of human monocyte-derived macrophages with omentin protein attenuated expression of MMP9 and pro-inflammatory mediators, and MMP9 activation after stimulation with lipopolysaccharide (LPS). Treatment of human vascular smooth muscle cells with omentin protein reduced expression and activation of MMP2 after stimulation with tumor necrosis factor α. Omentin treatment increased phosphorylation levels of Akt in human macrophages and vascular smooth muscle cells. The suppressive effects of omentin on MMP9 and MMP2 expression were reversed by inhibition of integrin-αVβ3/PI3-kinase/Akt signaling in macrophages and vascular smooth muscle cells, respectively. CONCLUSION These data suggest that omentin acts as an adipokine that can attenuate Ang II-induced development of AAA through suppression of MMP9 and MMP2 expression and inflammatory response in the vascular wall.
Collapse
Affiliation(s)
- Lixin Fang
- Department of Cardiology, Nagoya University Graduate School of Medicine
| | - Koji Ohashi
- Department of Molecular Medicine and Cardiology, Nagoya University Graduate School of Medicine
| | - Naoya Otaka
- Department of Cardiology, Nagoya University Graduate School of Medicine
| | - Hayato Ogawa
- Department of Cardiology, Nagoya University Graduate School of Medicine
| | | | - Hiroshi Kawanishi
- Department of Cardiology, Nagoya University Graduate School of Medicine
| | - Yasuko K Bando
- Department of Cardiology, Nagoya University Graduate School of Medicine
| | - Rei Shibata
- Department of Advanced Cardiovascular Therapeutics, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Yuuki Shimizu
- Department of Cardiology, Nagoya University Graduate School of Medicine
| | - Katsuhiro Kato
- Department of Cardiology, Nagoya University Graduate School of Medicine
| | - Tomonobu Takikawa
- Department of Cardiology, Nagoya University Graduate School of Medicine
| | - Yuta Ozaki
- Department of Cardiology, Nagoya University Graduate School of Medicine
| | - Mikito Takefuji
- Department of Cardiology, Nagoya University Graduate School of Medicine
| | - Toyoaki Murohara
- Department of Cardiology, Nagoya University Graduate School of Medicine
| | - Noriyuki Ouchi
- Department of Molecular Medicine and Cardiology, Nagoya University Graduate School of Medicine
| |
Collapse
|
7
|
Adams L, Brangsch J, Hamm B, Makowski MR, Keller S. Targeting the Extracellular Matrix in Abdominal Aortic Aneurysms Using Molecular Imaging Insights. Int J Mol Sci 2021; 22:ijms22052685. [PMID: 33799971 PMCID: PMC7962044 DOI: 10.3390/ijms22052685] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Revised: 03/02/2021] [Accepted: 03/04/2021] [Indexed: 12/22/2022] Open
Abstract
This review outlines recent preclinical and clinical advances in molecular imaging of abdominal aortic aneurysms (AAA) with a focus on molecular magnetic resonance imaging (MRI) of the extracellular matrix (ECM). In addition, developments in pharmacologic treatment of AAA targeting the ECM will be discussed and results from animal studies will be contrasted with clinical trials. Abdominal aortic aneurysm (AAA) is an often fatal disease without non-invasive pharmacologic treatment options. The ECM, with collagen type I and elastin as major components, is the key structural component of the aortic wall and is recognized as a target tissue for both initiation and the progression of AAA. Molecular imaging allows in vivo measurement and characterization of biological processes at the cellular and molecular level and sets forth to visualize molecular abnormalities at an early stage of disease, facilitating novel diagnostic and therapeutic pathways. By providing surrogate criteria for the in vivo evaluation of the effects of pharmacological therapies, molecular imaging techniques targeting the ECM can facilitate pharmacological drug development. In addition, molecular targets can also be used in theranostic approaches that have the potential for timely diagnosis and concurrent medical therapy. Recent successes in preclinical studies suggest future opportunities for clinical translation. However, further clinical studies are needed to validate the most promising molecular targets for human application.
Collapse
Affiliation(s)
- Lisa Adams
- Charité—Universitaetsmedizin Berlin Corporate Member of Freie Universität Berlin Humboldt-Universitaet zu Berlin, and Berlin Institute of Health, Charitéplatz 1, 10117 Berlin, Germany; (J.B.); (B.H.); (M.R.M.); (S.K.)
- Berlin Institute of Health (BIH), 10178 Berlin, Germany
- Correspondence: ; Tel.: +49-30-450-627-376
| | - Julia Brangsch
- Charité—Universitaetsmedizin Berlin Corporate Member of Freie Universität Berlin Humboldt-Universitaet zu Berlin, and Berlin Institute of Health, Charitéplatz 1, 10117 Berlin, Germany; (J.B.); (B.H.); (M.R.M.); (S.K.)
| | - Bernd Hamm
- Charité—Universitaetsmedizin Berlin Corporate Member of Freie Universität Berlin Humboldt-Universitaet zu Berlin, and Berlin Institute of Health, Charitéplatz 1, 10117 Berlin, Germany; (J.B.); (B.H.); (M.R.M.); (S.K.)
| | - Marcus R. Makowski
- Charité—Universitaetsmedizin Berlin Corporate Member of Freie Universität Berlin Humboldt-Universitaet zu Berlin, and Berlin Institute of Health, Charitéplatz 1, 10117 Berlin, Germany; (J.B.); (B.H.); (M.R.M.); (S.K.)
- Department of Diagnostic and Interventional Radiology, Technical University of Munich, Ismaninger Str. 22, 81675 Munich, Germany
| | - Sarah Keller
- Charité—Universitaetsmedizin Berlin Corporate Member of Freie Universität Berlin Humboldt-Universitaet zu Berlin, and Berlin Institute of Health, Charitéplatz 1, 10117 Berlin, Germany; (J.B.); (B.H.); (M.R.M.); (S.K.)
| |
Collapse
|
8
|
Up-regulated MCPIP1 in abdominal aortic aneurysm is associated with vascular smooth muscle cell apoptosis and MMPs production. Biosci Rep 2020; 39:220754. [PMID: 31651935 PMCID: PMC6851509 DOI: 10.1042/bsr20191252] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2019] [Revised: 09/24/2019] [Accepted: 10/08/2019] [Indexed: 12/27/2022] Open
Abstract
Abdominal aortic aneurysm (AAA) is often clinically silent before rupture characterized by extensive vascular inflammation and degenerative elasticity of aortic wall. Monocyte chemotactic protein-induced protein-1 (MCPIP1) exhibits anti-infllammatory and pro-apoptotic effects involved in atherogenesis. However, little is known about the expression and the contribution of MCPIP1 in AAA. In the present study, we collected clinical AAA specimens and constructed AAA mice model through Ang-II infusion, and found apparently increased MCPIP1 expression and severe inflammatory infiltration in AAA aortic membrane as evidenced by elevated levels of monocyte chemotactic protein 1 (MCP-1), interleukin 1 β (IL-1β) and NF-κB, as well as HE staining. The elasticity of aortic tunica media was impaired along with multiple apoptosis of vascular smooth muscle cells (VSMCs) in Ang-II-induced aneurysmal mouse. In vitro Ang-II administration of VSMCs induced MCPIP1 expression, accompanied by up-regulation of matrix metalloproteinase (MMP) 2 (MMP-2) and MMP-9, as well as enhancement of VSMCs proliferation and apoptosis, which may cause damage of intima–media elasticity. Silencing MCPIP1 reversed above effects to further restore the balance of proliferation and apoptosis in VSMCs. Overall, our data indicated that up-regulation of MCPIP1 may become a promising candidate for the diagnosis of AAA, and specific knockdown of MCPIP1 in VSMCs could inhibit VSMCs apoptosis and down-regulate MMPs to maintain vascular wall elasticity. Therefore, knockdown of MCPIP1 may serve as a potential target for gene therapy of AAA.
Collapse
|
9
|
Kasashima S, Kawashima A, Kasashima F, Matsumoto Y, Ozaki S. Exacerbation of immunoglobulin G4-related inflammatory abdominal aortic aneurysm after endovascular repair. Pathol Int 2020; 70:812-819. [PMID: 32749024 DOI: 10.1111/pin.12998] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2020] [Revised: 07/13/2020] [Accepted: 07/19/2020] [Indexed: 11/28/2022]
Abstract
A 78-year-old male was admitted to our hospital with lumbar pain and was found to have an abdominal aortic aneurysm (AAA) and femoral artery aneurysm (FAA). Initially, the patient underwent endovascular aneurysm repair (EVAR) for the AAA and aneurysmectomy for the FAA. The FAA was diagnosed by histology as immunoglobulin G4-related disease (IgG4-RD). The preoperative serum IgG4 level was within the normal range, although a slight serum interleukin-6 (IL-6) elevation was observed. Four years later, the AAA-sac diameter had expanded and the serum levels of both IgG4 and IL-6 levels had increased. Six years after the initial EVAR, aneurysmorrhaphy of AAA-sac was performed. The resected specimen revealed adventitial fibrosis and prominent lymphoplasmacytic infiltrate with regulatory T cells, satisfying histological diagnostic criteria for IgG4-RD. Immunoreactive matrix metalloproteinases (MMPs), particularly MMP-2 and MMP-9, and IL-6 were detected within numerous spindle cells in the adventitia of both the FAA and the AAA-sac. Five months after the aneurysmorrhaphy, the residual AAA-sac was again enlarged with a thickened wall that accumulated [18 F] fluoro-2-deoxy-D-glucose (FDG-PET) on positron emission tomography; these findings were paralleled by increased levels of serum IgG4 and IL-6. Therefore, persistent inflammation after EVAR may be attributed to the inflammatory sequelae of IgG4-RD.
Collapse
Affiliation(s)
- Satomi Kasashima
- Department of Clinical Laboratory Science, Kanazawa University, Ishikawa, Japan.,Department of Pathology, National Hospital Organization, Kanazawa Medical Center, Ishikawa, Japan.,Department of Clinical Laboratory, National Hospital Organization, Kanazawa Medical Center, Ishikawa, Japan
| | - Atsuhiro Kawashima
- Department of Pathology, National Hospital Organization, Kanazawa Medical Center, Ishikawa, Japan.,Department of Clinical Laboratory, National Hospital Organization, Kanazawa Medical Center, Ishikawa, Japan
| | - Fuminori Kasashima
- Department of Cardiovascular Surgery, National Hospital Organization, Kanazawa Medical Center, Ishikawa, Japan
| | - Yasushi Matsumoto
- Department of Cardiovascular Surgery, National Hospital Organization, Kanazawa Medical Center, Ishikawa, Japan
| | - Satoru Ozaki
- Department of Clinical Laboratory Science, Kanazawa University, Ishikawa, Japan
| |
Collapse
|
10
|
Li L, Ma W, Pan S, Li Y, Wang H, Wang B, Khalil RA. MiR-126a-5p limits the formation of abdominal aortic aneurysm in mice and decreases ADAMTS-4 expression. J Cell Mol Med 2020; 24:7896-7906. [PMID: 32469162 PMCID: PMC7348185 DOI: 10.1111/jcmm.15422] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2019] [Revised: 04/06/2020] [Accepted: 05/07/2020] [Indexed: 12/14/2022] Open
Abstract
Abdominal aortic aneurysm (AAA) is a serious vascular disease featured by inflammatory infiltration in aortic wall, aortic dilatation and extracellular matrix (ECM) degradation. Dysregulation of microRNAs (miRNAs) is implicated in AAA progress. By profiling miRNA expression in mouse AAA tissues and control aortas, we noted that miR-126a-5p was down-regulated by 18-fold in AAA samples, which was further validated with real-time qPCR. This study was performed to investigate miR-126a-5p's role in AAA formation. In vivo, a 28-d infusion of 1 μg/kg/min Angiotensin (Ang) II was used to induce AAA formation in Apoe-/- mice. MiR-126a-5p (20 mg/kg; MIMAT0000137) or negative control (NC) agomirs were intravenously injected to mice on days 0, 7, 14 and 21 post-Ang II infusion. Our data showed that miR-126a-5p overexpression significantly improved the survival and reduced aortic dilatation in Ang II-infused mice. Elastic fragment and ECM degradation induced by Ang II were also ameliorated by miR-126a-5p. A strong up-regulation of ADAM metallopeptidase with thrombospondin type 1 motif 4 (ADAMTS-4), a secreted proteinase that regulates matrix degradation, was observed in smooth muscle cells (SMCs) of aortic tunica media, which was inhibited by miR-126a-5p. Dual-luciferase results demonstrated ADAMTS-4 as a new and valid target for miR-126a-5p. In vitro, human aortic SMCs (hASMCs) were stimulated by Ang II. Gain- and loss-of-function experiments further confirmed that miR-126-5p prevented Ang II-induced ECM degradation, and reduced ADAMTS-4 expression in hASMCs. In summary, our work demonstrates that miR-126a-5p limits experimental AAA formation and reduces ADAMTS-4 expression in abdominal aortas.
Collapse
Affiliation(s)
- Lei Li
- Department of Vascular SurgeryThe Second Affiliated Hospital of Dalian Medical UniversityDalianChina
- Vascular Surgery Research LaboratoriesDivision of Vascular and Endovascular SurgeryBrigham and Women's HospitalHarvard Medical SchoolBostonMAUSA
| | - Wei Ma
- Department of AnatomyDalian Medical UniversityDalianChina
| | - Shuang Pan
- Department of PhysiologySchool of Basic MedicineJinzhou Medical UniversityJinzhouChina
| | - Yongqi Li
- Graduate School of Comprehensive Human SciencesUniversity of TsukubaTsukubaJapan
| | - Han Wang
- Department of Vascular SurgeryDalian University Affiliated Xinhua HospitalDalianChina
| | - Biao Wang
- Department of Biochemistry and Molecular BiologySchool of Life SciencesChina Medical UniversityShenyangChina
| | - Raouf A. Khalil
- Vascular Surgery Research LaboratoriesDivision of Vascular and Endovascular SurgeryBrigham and Women's HospitalHarvard Medical SchoolBostonMAUSA
| |
Collapse
|
11
|
Wei TH, Hsieh CL. Effect of Acupuncture on the p38 Signaling Pathway in Several Nervous System Diseases: A Systematic Review. Int J Mol Sci 2020; 21:E4693. [PMID: 32630156 PMCID: PMC7370084 DOI: 10.3390/ijms21134693] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Revised: 06/26/2020] [Accepted: 06/28/2020] [Indexed: 12/16/2022] Open
Abstract
Acupuncture is clinically used to treat various diseases and exerts positive local and systemic effects in several nervous system diseases. Advanced molecular and clinical studies have continually attempted to decipher the mechanisms underlying these effects of acupuncture. While a growing understanding of the pathophysiology underlying several nervous system diseases shows it to be related to inflammation and impair cell regeneration after ischemic events, the relationship between the therapeutic mechanism of acupuncture and the p38 MAPK signal pathway has yet to be elucidated. This review discusses the latest advancements in the identification of the effect of acupuncture on the p38 signaling pathway in several nervous system diseases. We electronically searched databases including PubMed, Embase, and the Cochrane Library from their inception to April 2020, using the following keywords alone or in various combinations: "acupuncture", "p38 MAPK pathway", "signaling", "stress response", "inflammation", "immune", "pain", "analgesic", "cerebral ischemic injury", "epilepsy", "Alzheimer's disease", "Parkinson's disease", "dementia", "degenerative", and "homeostasis". Manual acupuncture and electroacupuncture confer positive therapeutic effects by regulating proinflammatory cytokines, ion channels, scaffold proteins, and transcription factors including TRPV1/4, Nav, BDNF, and NADMR1; consequently, p38 regulates various phenomena including cell communication, remodeling, regeneration, and gene expression. In this review article, we found the most common acupoints for the relief of nervous system disorders including GV20, GV14, ST36, ST37, and LI4. Acupuncture exhibits dual regulatory functions of activating or inhibiting different p38 MAPK pathways, contributing to an overall improvement of clinical symptoms and function in several nervous system diseases.
Collapse
Affiliation(s)
- Tzu-Hsuan Wei
- Department of Chinese Medicine, China Medical University Hospital, Taichung 40447, Taiwan;
| | - Ching-Liang Hsieh
- Department of Chinese Medicine, China Medical University Hospital, Taichung 40447, Taiwan;
- Chinese Medicine Research Center, China Medical University, Taichung 40402, Taiwan
- Graduate Institute of Acupuncture Science, College of Chinese Medicine, China Medical University, Taichung 40402, Taiwan
| |
Collapse
|
12
|
Xiao J, Wei Z, Chen X, Chen W, Zhang H, Yang C, Shang Y, Liu J. Experimental abdominal aortic aneurysm growth is inhibited by blocking the JAK2/STAT3 pathway. Int J Cardiol 2020; 312:100-106. [PMID: 32334849 DOI: 10.1016/j.ijcard.2020.03.072] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/23/2019] [Revised: 01/05/2020] [Accepted: 03/27/2020] [Indexed: 12/19/2022]
Abstract
BACKGROUND The JAK/STAT pathway is a vital transcription signaling pathway that regulates gene expression and cellular activity. Our recently published study highlighted the role of IL-17A in abdominal aortic aneurysm (AAA) formation and rupture. IL-17A has been proven to upregulate vascular endothelial growth factor (VEGF) expression in some diseases. However, no study has demonstrated the relationships among JAK2/STAT3, IL-17A and VEGF. Therefore, we hypothesized that IL-17A may up-regulate VEGF expression via the JAK2/STAT3 signaling pathway to amplify the inflammatory response, exacerbate neovascularization, and accelerate AAA progression. METHODS To fully verify our hypothesis, two separate studies were performed: i) a study investigating the influence of JAK2/STAT3 on AAA formation and progression. ii) a study evaluating the relationship among IL-17A, JAK2/STAT3 and VEGF. Human tissues were collected from 7 AAA patients who underwent open surgery and 7 liver transplantation donors. All human aortic tissues were examined by histological and immunohistochemical staining, and Western blotting. Furthermore, mouse aortic tissues were also examined by histological and immunohistochemical staining and Western blotting, and the mouse aortic diameters were assessed by high-resolution Vevo 2100 microimaging system. RESULTS Among human aortic tissues, JAK2/STAT3, IL-17A and VEGF expression levels were higher in AAA tissues than in control tissues. Group treated with WP1066 (a selective JAK2/STAT3 pathway inhibitor), IL-17A, and VEGF groups had AAA incidences of 25%, 40%, and 65%, respectively, while the control group had an incidence of 75%. Histopathological analysis revealed that the IL-17A- and VEGF-related inflammatory responses were attenuated by WP1066. Thus, blocking the JAK2/STAT3 pathway with WP1066 attenuated experimental AAA progression. In addition, in study ii, we found that IL-17A siRNA seemed to attenuate the expression of IL-17A and VEGF in vivo study; treatment with VEGF siRNA decreased the expression of VEGF, while IL-17A expression remained high. In an in vitro study, rhIL-17A treatment increased JAK2/STAT3 and VEGF expression in macrophages in a dose-dependent manner. CONCLUSION Blocking the JAK2/STAT3 pathway with WP1066 (a JAK2/STAT3 specific inhibitor) attenuates experimental AAA progression. During AAA progression, IL-17A may influence the expression of VEGF via the JAK2/STAT3 signaling pathway. This potential mechanism may suggest a novel strategy for nonsurgical AAA treatment.
Collapse
Affiliation(s)
- Jie Xiao
- Department of Cardiovascular Surgery, Central Hospital of Wuhan, Tongji Medical college, Huazhong University of Science and Technology, Wuhan, 430014, Hubei, China
| | - Zhanjie Wei
- Department of Thyroid and Breast Surgery, Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430014, Hubei, China
| | - Xing Chen
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical college, Huazhong University of Science and Technology, Wuhan, 430022, Hubei, China
| | - Weiqiang Chen
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical college, Huazhong University of Science and Technology, Wuhan, 430022, Hubei, China
| | - Hua Zhang
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical college, Huazhong University of Science and Technology, Wuhan, 430022, Hubei, China
| | - Chuanlei Yang
- Department of Cardiovascular Surgery, Central Hospital of Wuhan, Tongji Medical college, Huazhong University of Science and Technology, Wuhan, 430014, Hubei, China
| | - Yuqiang Shang
- Department of Cardiovascular Surgery, Central Hospital of Wuhan, Tongji Medical college, Huazhong University of Science and Technology, Wuhan, 430014, Hubei, China
| | - Jinping Liu
- Department of Cardiovascular Surgery, Zhongnan Hospital, Wuhan University, Wuhan, 430071, Hubei, China.
| |
Collapse
|
13
|
Chumachenko PV, Afanasyev MA, Ivanova AG, Drobkova IP, Kheimets GI, Postnov AY. [Inflammatory infiltrates, vasa vasorum, and endothelial NO synthase in the wall of thoracic aortic aneurysm]. Arkh Patol 2019; 81:45-52. [PMID: 31626204 DOI: 10.17116/patol20198105145] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
OBJECTIVE To elucidate whether there is a relationship between inflammation of the wall of aortic aneurysm and the number of vasa vasorum in it. MATERIAL AND METHODS The investigation material was aortic aneurysm wall segments obtained during surgery. Among the patients, there were 20 men and 5 women. The patients' age ranged from 33 to 69 years. The investigation used monoclonal antibodies to macrophages (CD68), T cells (CD3, CD4, and CD8) and antibodies to von Willebrand factor, endothelial NO synthase, and alpha smooth muscle actin. A morphometric study was conducted. RESULTS Calculation of the number of vasa vasorum (including newly formed vessels) in the adventitia of aortic aneurysm revealed that there was a statistically significant difference between the number of vasa vasorum in patients with an active inflammatory response (Group 1) versus Group 2 patients with a moderate inflammatory process in the aneurysm wall (p≤0.05) and a statistically significant difference between Groups 1 and 3 (without inflammatory infiltrates) (p≤0.05). Endothelial vasa vasorum heterogeneity was found in case of an immune response to NO synthase. At the same time individual vasa vasorium did not contain NO synthase, this enzyme was identified in the endothelium in a number of nearby vessels. CONCLUSION The increase in the number of vasa vasorum in the aneurysm wall in patients with abundant inflammatory infiltrates is due to the fact that some of the inflammatory cytokines of T-cells and macrophages also contribute to angiogenesis.
Collapse
Affiliation(s)
- P V Chumachenko
- National Medical Research Center of Cardiology, Ministry of Health of Russia, Moscow, Russia
| | - M A Afanasyev
- National Medical Research Center of Cardiology, Ministry of Health of Russia, Moscow, Russia
| | - A G Ivanova
- Acad. B.V. Petrovsky Russian Research Center of Surgery, Moscow, Russia
| | - I P Drobkova
- National Medical Research Center of Cardiology, Ministry of Health of Russia, Moscow, Russia
| | - G I Kheimets
- National Medical Research Center of Cardiology, Ministry of Health of Russia, Moscow, Russia
| | - A Yu Postnov
- National Medical Research Center of Cardiology, Ministry of Health of Russia, Moscow, Russia; Research Institute of Human Morphology, Ministry of Science and Higher Education of Russia, Moscow, Russia
| |
Collapse
|
14
|
Spanos K, Nana P, Kouvelos G, Mpatzalexis K, Matsagkas M, Giannoukas AD. Anatomical Differences Between Intact and Ruptured Large Abdominal Aortic Aneurysms. J Endovasc Ther 2019; 27:117-123. [PMID: 31709885 DOI: 10.1177/1526602819886568] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Purpose: To compare different anatomical characteristics between intact and ruptured large abdominal aortic aneurysms (rAAA >80 mm) with the goal of refining the process of estimating rupture risk. Materials and Methods: A retrospective study involving 62 male patients with large (>80 mm) aneurysms matched for age and smoking produced a 31-patient elective group with a mean maximum aneurysm diameter of 92±9.7 mm and a 31-patient rAAA group (mean maximum aneurysm diameter 95.7±12 mm). Preoperative computed tomography angiography scans were analyzed with a dedicated workstation, and anatomical characteristics of the aortic neck, iliac arteries, and aneurysm were compared in multivariable regression analyses; the outcomes are given as the odds ratio (OR) with 95% confidence interval (CI). The prognostic utility of several characteristics as predictors of rupture occurrence was examined with receiver operating characteristic (ROC) curves. Results: Anatomical characteristics differing significantly between elective and ruptured aneurysms were the infrarenal aortic neck diameters at 5 mm, 10 mm and 15 mm; the neck length and calcification; the common iliac artery (CIA) lengths; the iliac artery indexes; the left CIA and external iliac artery diameters; and the total and true lumen aneurysm volumes. Intraluminal thrombus (ILT) volume did not differ (p=0.76), although its distribution in elective vs ruptured cases did [absent: 0% vs 19%, respectively (p=0.025); circumferential: 61% vs 35%, respectively (p=0.04)]. Total aneurysm volume was higher in rAAA (442±140 mL) vs intact AAA (331±143 mL, p=0.014), while the ILT/total aneurysm volume rate was lower in rAAA (55%) vs intact AAA (70%, p=0.02). Multivariate analysis determined that a shorter left CIA (OR 1.07, 95% CI 1.01 to 1.1, p=0.016) and a smaller total aneurysm volume (OR 1.007, CI. 1.001 to 1.014, p=0.016) were associated with intact AAA. After a ROC curve analysis, left CIA length <50 mm demonstrated a lower incidence of rupture (sensitivity 60% and specificity 78%), while total aneurysm volume <380 mL had 60% sensitivity and specificity. Conclusion: Large rAAAs seem to have different anatomical characteristics than similarly sized intact AAAs. Large intact AAAs have lower total aneurysm volumes and shorter left CIAs, with higher ILT/aneurysm volume rates.
Collapse
Affiliation(s)
- Konstantinos Spanos
- Department of Vascular Surgery, University Hospital of Larissa, Faculty of Medicine, School of Health Sciences, University of Thessaly, Larissa, Greece
| | - Petroula Nana
- Department of Vascular Surgery, University Hospital of Larissa, Faculty of Medicine, School of Health Sciences, University of Thessaly, Larissa, Greece
| | - George Kouvelos
- Department of Vascular Surgery, University Hospital of Larissa, Faculty of Medicine, School of Health Sciences, University of Thessaly, Larissa, Greece
| | - Konstantinos Mpatzalexis
- Department of Vascular Surgery, University Hospital of Larissa, Faculty of Medicine, School of Health Sciences, University of Thessaly, Larissa, Greece
| | - Miltiadis Matsagkas
- Department of Vascular Surgery, University Hospital of Larissa, Faculty of Medicine, School of Health Sciences, University of Thessaly, Larissa, Greece
| | - Athanasios D Giannoukas
- Department of Vascular Surgery, University Hospital of Larissa, Faculty of Medicine, School of Health Sciences, University of Thessaly, Larissa, Greece
| |
Collapse
|
15
|
Meekel JP, Mattei G, Costache VS, Balm R, Blankensteijn JD, Yeung KK. A multilayer micromechanical elastic modulus measuring method in ex vivo human aneurysmal abdominal aortas. Acta Biomater 2019; 96:345-353. [PMID: 31306785 DOI: 10.1016/j.actbio.2019.07.019] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2019] [Revised: 06/23/2019] [Accepted: 07/11/2019] [Indexed: 02/07/2023]
Abstract
Abdominal aortic aneurysms (AAA) are common and potentially life-threatening aortic dilatations, due to the effect of hemodynamic changes on the aortic wall. Previous research has shown a potential pathophysiological role for increased macroscopic aneurysmal wall stiffness; however, not investigating micromechanical stiffness. We aimed to compile a new protocol to examine micromechanical live aortic stiffness (elastic moduli), correlated to histological findings with quantitative immunofluorescence (QIF). Live AAA biopsies (n = 7) and non-dilated aortas (controls; n = 3) were sectioned. Local elastic moduli of aortic intima, media and adventitia were analysed in the direction towards the lumen and vice versa with nanoindentation. Smooth muscle cells (SMC), collagen and fibroblasts were examined using QIF. Nanoindentation of AAA vs. controls demonstrated a 4-fold decrease in elastic moduli (p = 0.022) for layers combined and a 26-fold decrease (p = 0.017) for media-to-intima direction. QIF of AAA vs. controls revealed a 4-, 3- and 6-fold decrease of SMC, collagen and fibroblasts, respectively (p = 0.036). Correlations were found between bidirectional intima and media measurements (ρ = 0.661, p = 0.038) and all QIF analyses (ρ = 0.857-0.905, p = 0.002-0.007). We present a novel protocol to analyse microscopic elastic moduli in live aortic tissues using nanoindentation. Hence, our preliminary findings of decreased elastic moduli and altered wall composition warrant further microscopic stiffness investigation to potentially clarify AAA pathophysiology and to explore potential treatment by wall strengthening. STATEMENT OF SIGNIFICANCE: Although extensive research on the pathophysiology of dilated abdominal aortas (aneurysms) has been performed, the exact underlying pathways are still largely unclear. Previously, the macroscopic stiffness of the pathologic and healthy aortic wall has been studied. This study however, for the first time, studied the microscopic stiffness changes in live tissue of dilated and non-dilated abdominal aortas. This new protocol provides a device to analyse the alterations on cellular level within their microenvironment, whereas previous studies studied the aorta as a whole. Outcomes of these measurements might help to better understand the underlying origin of the incidence and progression of aneurysms and other aortic diseases.
Collapse
Affiliation(s)
- Jorn P Meekel
- Department of Vascular Surgery, Amsterdam University Medical Centers, Location VU Medical Center, Amsterdam, The Netherlands; Department of Physiology, Amsterdam University Medical Centers, Location VU Medical Center, Amsterdam Cardiovascular Sciences, Amsterdam, The Netherlands
| | - Giorgio Mattei
- Optics11 B.V., Amsterdam, The Netherlands; Biophotonics & Medical Imaging and LaserLaB, VU University Amsterdam, Amsterdam, The Netherlands; Department of Information Engineering, University of Pisa, Pisa, Italy
| | - Victor S Costache
- Department of Cardiovascular Surgery, Polisano Medlife Hospital, University "L. Blaga" Sibiu, Sibiu, Romania
| | - Ron Balm
- Department of Vascular Surgery, Amsterdam University Medical Centers, Location Amsterdam Medical Center, Amsterdam, the Netherlands
| | - Jan D Blankensteijn
- Department of Vascular Surgery, Amsterdam University Medical Centers, Location VU Medical Center, Amsterdam, The Netherlands
| | - Kak K Yeung
- Department of Vascular Surgery, Amsterdam University Medical Centers, Location VU Medical Center, Amsterdam, The Netherlands; Department of Physiology, Amsterdam University Medical Centers, Location VU Medical Center, Amsterdam Cardiovascular Sciences, Amsterdam, The Netherlands.
| |
Collapse
|
16
|
Georgiadis GS, Antoniou GA, Argyriou C, Schoretsanitis N, Nikolopoulos E, Kapoulas K, Lazarides MK, Tentes I. Correlation of Baseline Plasma and Inguinal Connective Tissue Metalloproteinases and Their Inhibitors With Late High-Pressure Endoleak After Endovascular Aneurysm Repair: Long-term Results. J Endovasc Ther 2019; 26:826-835. [DOI: 10.1177/1526602819871963] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Purpose: To investigate whether plasma and connective tissue matrix metalloproteinases (MMP) and their inhibitors (TIMP) may predict late high-pressure endoleak after endovascular aneurysm repair (EVAR). Materials and Methods: Samples of inguinal fascia and blood were collected in 72 consecutive patients (mean age 73.1 years; 68 men) undergoing primary EVAR with the Endurant stent-graft. Baseline plasma levels of MMP-2, MMP-9, TIMP-1, and TIMP-2 and baseline MMP-2 and MMP-9 activity estimated using gelatin zymography (GZ) were compared between patients who developed late endoleak in follow-up and those who did not. Subgroup analyses were performed between patients with (n=18) and without inguinal hernias and between patients with moderate-diameter (50–59 mm; n=45) or large-diameter (≥60 mm; n=27) abdominal aortic aneurysms (AAA) at primary EVAR. Results: The mean follow-up period was 63.1 months (range 7.5–91.5), during which time 13 (18.1%) patients developed type I (6 Ia and 5 Ib) or 2 type III endoleaks. Only GZ-analyzed proMMP-9 concentrations were higher in the endoleak group than in patients without endoleak (mean difference 8.44, 95% CI −19.653 to −1.087, p=0.03). The patients with primary inguinal hernia at presentation had significantly higher tissue TIMP-2 values (0.8±0.7 vs 0.5±0.4, p=0.018) but lower plasma total (pro- + active) MMP-9 values (11.9±7.8 vs 16.2±7.4, p=0.042) than patients without hernias at the time of EVAR. Patients with AAAs ≥60 mm had significantly higher mean tissue homogenate levels of total (pro- + active) MMP-9 (p=0.025) and total (pro- + active) MMP-2 (p=0.049) as well as higher proMMP-9 (p=0.018) and total (pro- + active) MMP-9 (p=0.021) levels based on GZ compared to patients with moderate-diameter AAAs. Regression analysis revealed a significant association between total (pro- + active) MMP-9 plasma samples and the presence of hernia (OR 0.899, 95% CI 0.817 to 0.989, p=0.029) and between GZ-analyzed proMMP-9 and late endoleak (OR 1.055, 95% CI 1.007 to 1.106, p=0.025). GZ-analyzed proMMP-9 and active MMP-9 were strong predictors of late endoleak in patients with hernia (p=0.012 and p=0.044, respectively) and in patients with AAAs ≥60 mm (p=0.018 and p=0.041 respectively). Conclusion: Inguinal fascial tissue proMMP-9 significantly predicted late endoleak. ProMMP-9 and active MMP-9 biomarkers are significantly associated with late endoleak in hernia patients and in patients with AAAs ≥60 mm. Considering the clinical association between hernia and AAA and the fact that the AAA wall connective tissue environment remains exposed to systemic circulation after EVAR, inguinal fascia extracellular matrix dysregulation and altered MMP activity may reflect similar changes in AAA biology, leading to complications such as endoleak.
Collapse
Affiliation(s)
- George S. Georgiadis
- Department of Vascular Surgery, “Democritus” University of Thrace, University General Hospital of Alexandroupolis, Thrace, Greece
| | - George A. Antoniou
- Department of Vascular Surgery, “Democritus” University of Thrace, University General Hospital of Alexandroupolis, Thrace, Greece
- Department of Vascular and Endovascular Surgery, The Royal Oldham Hospital, Pennine Acute Hospitals NHS Trust, Manchester, UK
| | - Christos Argyriou
- Department of Vascular Surgery, “Democritus” University of Thrace, University General Hospital of Alexandroupolis, Thrace, Greece
| | - Nikolaos Schoretsanitis
- Department of Vascular Surgery, “Democritus” University of Thrace, University General Hospital of Alexandroupolis, Thrace, Greece
| | - Evaggelos Nikolopoulos
- Department of Vascular Surgery, “Democritus” University of Thrace, University General Hospital of Alexandroupolis, Thrace, Greece
| | - Konstantinos Kapoulas
- Department of Vascular Surgery, “Democritus” University of Thrace, University General Hospital of Alexandroupolis, Thrace, Greece
| | - Miltos K. Lazarides
- Department of Vascular Surgery, “Democritus” University of Thrace, University General Hospital of Alexandroupolis, Thrace, Greece
| | - Ioannis Tentes
- Laboratory of Biochemistry, “Democritus” University of Thrace, University General Hospital of Alexandroupolis, Thrace, Greece
| |
Collapse
|
17
|
Tomimori Y, Manno A, Tanaka T, Futamura-Takahashi J, Muto T, Nagahira K. ASB17061, a novel chymase inhibitor, prevented the development of angiotensin II-induced abdominal aortic aneurysm in apolipoprotein E-deficient mice. Eur J Pharmacol 2019; 856:172403. [DOI: 10.1016/j.ejphar.2019.05.032] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2019] [Revised: 04/29/2019] [Accepted: 05/16/2019] [Indexed: 10/26/2022]
|
18
|
Yang X, Peng J, Pang J, Wan W, Chen L. A functional polymorphism in the promoter region of miR-155 predicts the risk of intracranial hemorrhage caused by rupture intracranial aneurysm. J Cell Biochem 2019; 120:18618-18628. [PMID: 31338876 DOI: 10.1002/jcb.28785] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2018] [Accepted: 01/10/2019] [Indexed: 01/28/2023]
Abstract
BACKGROUND This study aimed to study the effect and underlying molecular mechanisms of single-nucleotide polymorphism (SNP) rs767649 during the pathogenesis of intracranial aneurysm (IA) rupture. METHOD Real-time PCR and Western blot analysis were performed to detect the differentiated expression of miR-155 and matrix metalloproteinase-2 (MMP-2) among different sample groups. Computational analysis and luciferase assay were conducted to study the effect of SNP rs767649 on the expression of miR-155 as well as the regulatory relationship between miR-155 and MMP-2. RESULTS In unruptured IA samples, the expression of miR-155 was upregulated while the expression of MMP-2 was downregulated compared with the ruptured IA samples. Similarly, the expression of miR-155 was upregulated while the expression of MMP-2 was downregulated in samples genotyped as AA/AT compared with samples genotyped as TT. In addition, compared with the negative controls, the luciferase activities of cells treated with rs767649A and rs767649T were both elevated with rs767649A-transfected cells expressing the highest luciferase activity. Furthermore, a negative relationship was established between miR-155 and MMP-2 by measuring the luciferase activity of cells cotransfected with miR-155 and the wild-type 3'-untranslated region of MMP-2. CONCLUSION The results of this study showed that the SNP rs767649 in the promoter of miR-155 could reduce the transcription activity of miR-155, while poorly expressed miR-155 could increase the incidence of IA rupture by increasing the expression of MMP-2, especially in subjects carrying the TT genotype of SNP rs767649.
Collapse
Affiliation(s)
- Xiaobo Yang
- Department of Neurosurgery, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Jianhua Peng
- Department of Neurosurgery, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Jinwei Pang
- Department of Neurosurgery, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Weifeng Wan
- Department of Neurosurgery, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Ligang Chen
- Department of Neurosurgery, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| |
Collapse
|
19
|
Seto SW, Chang D, Kiat H, Wang N, Bensoussan A. Chinese Herbal Medicine as a Potential Treatment of Abdominal Aortic Aneurysm. Front Cardiovasc Med 2018; 5:33. [PMID: 29732374 PMCID: PMC5919947 DOI: 10.3389/fcvm.2018.00033] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2017] [Accepted: 03/20/2018] [Indexed: 12/19/2022] Open
Abstract
Abdominal aortic aneurysm (AAA) is an irreversible condition where the abdominal aorta is dilated leading to potentially fatal consequence of aortic rupture. Multiple mechanisms are involved in the development and progression of AAA, including chronic inflammation, oxidative stress, vascular smooth muscle (VSMC) apoptosis, immune cell infiltration and extracellular matrix (ECM) degradation. Currently surgical therapies, including minimally invasive endovascular aneurysm repair (EVAR), are the only viable interventions for AAAs. However, these treatments are not appropriate for the majority of AAAs, which measure <50 mm. Substantial effort has been invested to identify and develop pharmaceutical treatments such as statins and doxycycline for this potentially lethal condition but these interventions failed to offer a cure or to retard the progression of AAA. Chinese herbal medicine (CHM) has been used for the management of cardiovascular diseases for thousands of years in China and other Asian countries. The unique multi-component and multi-target property of CHMs makes it a potentially ideal therapy for multifactorial diseases such as AAA. In this review, we review the current scientific evidence to support the use of CHMs for the treatment of AAA. Mechanisms of action underlying the effects of CHMs on AAA are also discussed.
Collapse
Affiliation(s)
- Sai Wang Seto
- NICM Health Research Institute, Western Sydney University, Penrith, Australia
| | - Dennis Chang
- NICM Health Research Institute, Western Sydney University, Penrith, Australia
| | - Hosen Kiat
- Faculty of Medicine, University of New South Wales, Sydney, Australia.,School of Medicine, Western Sydney University, Penrith, Australia.,Faculty of Medicine and Health Sciences, Macquarie University, Sydney, Australia
| | - Ning Wang
- NICM Health Research Institute, Western Sydney University, Penrith, Australia.,Key Laboratory of Xin'an Medicine, Ministry of Education, Hefei, China.,College of Pharmacy, Anhui University of Chinese Medicine, Hefei, China.,Institute for Pharmacodynamics and Safety Evaluation of Chinese Medicine, Anhui University of Chinese Medicine, Hefei, China
| | - Alan Bensoussan
- NICM Health Research Institute, Western Sydney University, Penrith, Australia
| |
Collapse
|
20
|
Siennicka A, Zuchowski M, Kaczmarczyk M, Cnotliwy M, Clark JS, Jastrzębska M. Tissue factor levels and the fibrinolytic system in thin and thick intraluminal thrombus and underlying walls of abdominal aortic aneurysms. J Vasc Surg 2018; 68:30S-37S. [PMID: 29571624 DOI: 10.1016/j.jvs.2017.12.030] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2017] [Accepted: 12/08/2017] [Indexed: 10/17/2022]
Abstract
BACKGROUND The hemostatic system cooperates with proteolytic degradation in processes allowing abdominal aortic aneurysm (AAA) formation. In previous studies, it has been suggested that aneurysm rupture depends on intraluminal thrombus (ILT) thickness, which varies across each individual aneurysm. We hypothesized that hemostatic components differentially accumulate in AAA tissue in relation to ILT thickness. Thick (A1) and thin (B1) segments of ILTs and aneurysm wall sections A (adjacent to A1) and B (adjacent to B1) from one aneurysm sac were taken from 35 patients undergoing elective repair. METHODS Factor levels were measured using enzyme-linked immunosorbent assay of protein extract. RESULTS Tissue factor (TF) activities were significantly higher in thinner segments of AAA (B1 vs A1, P = .003; B vs A, P < .001; B vs A1, P < .001; B vs B1, P = .001). Significantly higher tissue plasminogen activator was found in thick thrombus-covered wall segments (A) than in B, A1, and B1 (P = .015, P < .001, and P < .001, respectively). Plasminogen concentrations were highest in ILT. Concentrations of α2-antiplasmin in thin ILT adjacent walls (B) were higher compared with wall (A) adjacent to thick ILT (P = .021) and thick ILT (A1; P < .001). Significant correlations between levels of different factors were mostly found in thick ILT (A1). However, no correlations were found at B sites, except for a correlation between plasmin and TF activities (r = 0.55; P = .004). CONCLUSIONS These results suggest that higher TF activities are present in thinner AAA regions. These parameters and local fibrinolysis may be part of the processes leading to destruction of the aneurysm wall.
Collapse
Affiliation(s)
- Aldona Siennicka
- Department of Laboratory Diagnostics, Pomeranian Medical University, Szczecin, Poland.
| | - Marta Zuchowski
- Department of Laboratory Diagnostics, Pomeranian Medical University, Szczecin, Poland
| | - Mariusz Kaczmarczyk
- Department of Clinical and Molecular Biochemistry, Pomeranian Medical University, Szczecin, Poland
| | - Miłosław Cnotliwy
- Department of Vascular Surgery and Angiology, Pomeranian Medical University, Szczecin, Poland
| | - Jeremy Simon Clark
- Department of Clinical and Molecular Biochemistry, Pomeranian Medical University, Szczecin, Poland
| | - Maria Jastrzębska
- Department of Laboratory Diagnostics, Pomeranian Medical University, Szczecin, Poland
| |
Collapse
|
21
|
Kidholm CL, Beck HC, Madsen JB, Palstrøm NB, Lindholt JS, Rasmussen LM. Preliminary analysis of proteome alterations in non-aneurysmal, internal mammary artery tissue from patients with abdominal aortic aneurysms. PLoS One 2018; 13:e0192957. [PMID: 29470511 PMCID: PMC5823374 DOI: 10.1371/journal.pone.0192957] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2017] [Accepted: 02/01/2018] [Indexed: 01/09/2023] Open
Abstract
Objective The pathogenesis of abdominal aortic aneurysms (AAA) involves a disturbed balance of breakdown and buildup of arterial proteins. We envision that individuals with AAA carry generalized arterial protein alterations either because of effects of genetically or environmental AAA risk factors or because of compensatory changes due to signaling molecules released from the affected aneurysmal tissue. Approach Protein extraction and quantitative proteome analysis by LC-MS/MS (liquid chromatography-mass spectrometry) was done on individual samples from the internal mammary artery from 11 individuals with AAA and 33 sex- and age-matched controls without AAA. Samples were selected from a biobank of leftover internal mammary arterial tissue gathered at coronary by-pass operations. Results We identified and quantitated 877 proteins, of which 44 were differentially expressed between the two groups (nominal p-values without correction for multiple testing). Some proteins related to the extracellular matrix displayed altered concentrations in the AAA group, particularly among elastin-related molecules [elastin, microfibrillar-associated protein 4 (MFAP4), lysyl oxidase]. In addition, several histones e.g. (e.g. HIST1H1E, HIST1H2BB) and other vascular cell proteins (e.g. versican, type VI collagen) were altered. Conclusions Our results support the notion that generalized alterations occur in the arterial tree in patients with AAA. Elastin-related proteins and histones seem to be part of such changes, however these preliminary results require replication in an independent set of specimens and validation by functional studies.
Collapse
Affiliation(s)
- Christina Lund Kidholm
- Centre of Individualized Medicine in Arterial Disease (CIMA), Odense University Hospital, Odense, Denmark
- Department of Clinical Biochemistry and Pharmacology, Odense University Hospital, Odense, Denmark
- * E-mail:
| | - Hans Christian Beck
- Department of Clinical Biochemistry and Pharmacology, Odense University Hospital, Odense, Denmark
| | - Julie Bukh Madsen
- Centre of Individualized Medicine in Arterial Disease (CIMA), Odense University Hospital, Odense, Denmark
- Department of Clinical Biochemistry and Pharmacology, Odense University Hospital, Odense, Denmark
| | - Nikolai Bjødstrup Palstrøm
- Centre of Individualized Medicine in Arterial Disease (CIMA), Odense University Hospital, Odense, Denmark
- Department of Clinical Biochemistry and Pharmacology, Odense University Hospital, Odense, Denmark
| | - Jes Sanddal Lindholt
- Centre of Individualized Medicine in Arterial Disease (CIMA), Odense University Hospital, Odense, Denmark
- Department of Vascular Surgery, Odense University Hospital, Odense, Denmark
| | - Lars Melholt Rasmussen
- Centre of Individualized Medicine in Arterial Disease (CIMA), Odense University Hospital, Odense, Denmark
- Department of Clinical Biochemistry and Pharmacology, Odense University Hospital, Odense, Denmark
| |
Collapse
|
22
|
Umebayashi R, Uchida HA, Kakio Y, Subramanian V, Daugherty A, Wada J. Cilostazol Attenuates Angiotensin II-Induced Abdominal Aortic Aneurysms but Not Atherosclerosis in Apolipoprotein E-Deficient Mice. Arterioscler Thromb Vasc Biol 2018; 38:903-912. [PMID: 29437572 DOI: 10.1161/atvbaha.117.309707] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2017] [Accepted: 01/25/2018] [Indexed: 11/16/2022]
Abstract
OBJECTIVE Abdominal aortic aneurysm (AAA) is a permanent dilation of the abdominal aorta associated with rupture, which frequently results in fatal consequences. AAA tissue is commonly characterized by localized structural deterioration accompanied with inflammation and profound accumulation of leukocytes, although the specific function of these cells is unknown. Cilostazol, a phosphodiesterase III inhibitor, is commonly used for patients with peripheral vascular disease or stroke because of its anti-platelet aggregation effect and anti-inflammatory effect, which is vasoprotective effect. In this study, we evaluated the effects of cilostazol on angiotensin II-induced AAA formation. APPROACH AND RESULTS Male apolipoprotein E-deficient mice were fed either normal diet or a diet containing cilostazol (0.1% wt/wt). After 1 week of diet consumption, mice were infused with angiotensin II (1000 ng/kg per minute) for 4 weeks. Angiotensin II infusion increased maximal diameters of abdominal aortas, whereas cilostazol administration significantly attenuated dilatation of abdominal aortas, thereby, reducing AAA incidence. Cilostazol also reduced macrophage accumulation, matrix metalloproteinases activation, and inflammatory gene expression in the aortic media. In cultured vascular endothelial cells, cilostazol reduced expression of inflammatory cytokines and adhesive molecules through activation of the cAMP-PKA (protein kinase A) pathway. CONCLUSIONS Cilostazol attenuated angiotensin II-induced AAA formation by its anti-inflammatory effect through phosphodiesterase III inhibition in the aortic wall. Cilostazol may be a promising new therapeutic option for AAAs.
Collapse
Affiliation(s)
- Ryoko Umebayashi
- From the Department of Nephrology, Rheumatology, Endocrinology and Metabolism (R.U., H.A.U., Y.K., J.W.) and Department of Chronic Kidney Disease and Cardiovascular Disease (H.A.U.), Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Japan; and Saha Cardiovascular Research Center (V.S., A.D.) and Department of Physiology (V.S., A.D.), University of Kentucky, Lexington
| | - Haruhito A Uchida
- From the Department of Nephrology, Rheumatology, Endocrinology and Metabolism (R.U., H.A.U., Y.K., J.W.) and Department of Chronic Kidney Disease and Cardiovascular Disease (H.A.U.), Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Japan; and Saha Cardiovascular Research Center (V.S., A.D.) and Department of Physiology (V.S., A.D.), University of Kentucky, Lexington.
| | - Yuki Kakio
- From the Department of Nephrology, Rheumatology, Endocrinology and Metabolism (R.U., H.A.U., Y.K., J.W.) and Department of Chronic Kidney Disease and Cardiovascular Disease (H.A.U.), Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Japan; and Saha Cardiovascular Research Center (V.S., A.D.) and Department of Physiology (V.S., A.D.), University of Kentucky, Lexington
| | - Venkateswaran Subramanian
- From the Department of Nephrology, Rheumatology, Endocrinology and Metabolism (R.U., H.A.U., Y.K., J.W.) and Department of Chronic Kidney Disease and Cardiovascular Disease (H.A.U.), Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Japan; and Saha Cardiovascular Research Center (V.S., A.D.) and Department of Physiology (V.S., A.D.), University of Kentucky, Lexington
| | - Alan Daugherty
- From the Department of Nephrology, Rheumatology, Endocrinology and Metabolism (R.U., H.A.U., Y.K., J.W.) and Department of Chronic Kidney Disease and Cardiovascular Disease (H.A.U.), Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Japan; and Saha Cardiovascular Research Center (V.S., A.D.) and Department of Physiology (V.S., A.D.), University of Kentucky, Lexington
| | - Jun Wada
- From the Department of Nephrology, Rheumatology, Endocrinology and Metabolism (R.U., H.A.U., Y.K., J.W.) and Department of Chronic Kidney Disease and Cardiovascular Disease (H.A.U.), Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Japan; and Saha Cardiovascular Research Center (V.S., A.D.) and Department of Physiology (V.S., A.D.), University of Kentucky, Lexington
| |
Collapse
|
23
|
Dahal S, Broekelman T, Mecham RP, Ramamurthi A. Maintaining Elastogenicity of Mesenchymal Stem Cell-Derived Smooth Muscle Cells in Two-Dimensional Culture. Tissue Eng Part A 2018; 24:979-989. [PMID: 29264957 DOI: 10.1089/ten.tea.2017.0237] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Abstract
Abdominal aortic aneurysms (AAAs) are localized expansions of the abdominal aorta that grow slowly to rupture. AAA growth is driven by irreversible elastic matrix breakdown in the aorta wall by chronically upregulated matrix metalloproteases (MMPs). Since adult vascular smooth muscle cells (SMCs) poorly regenerate elastic matrix, we previously explored utility of bone marrow mesenchymal stem cells and SMCs derived therefrom (BM-SMCs) for this purpose. One specific differentiated phenotype (cBM-SMCs) generated on a fibronectin substrate in presence of exogenous transforming growth factor-β and platelet-derived growth factor exhibited superior elastogenicity versus other phenotypes, and usefully provided proelastogenic and antiproteolytic stimuli to aneurysmal SMCs. Since in vivo cell therapy demands large cell inoculates, these derived SMCs must be propagated in vitro while maintaining their superior elastogenic, proelastogenic, and antiproteolytic characteristics. In this work, we thus investigated the culture conditions that must be provided to this propagation phase, which ensure that the differentiated SMCs maintain their phenotype and matrix regenerative benefits. Our results indicate that our BM-SMCs retain their phenotype in long-term culture even in the absence of differentiation growth factors and fibronectin substrate, but these conditions must be continued to be provided during postdifferentiation propagation if they are to maintain their superior elastic matrix deposition, crosslinking, and fiber formation properties. Our study, however, showed that cells propagated under these conditions exhibit higher expression of MMP-2, but favorably, no expression of elastolytic MMP-9. Hence, the study outcomes provide crucial guidelines to maintain phenotypic stability of cBM-SMCs during their propagation in two-dimensional culture before their delivery to the AAA wall for therapy.
Collapse
Affiliation(s)
- Shataakshi Dahal
- 1 Department of Biomedical Engineering, Cleveland Clinic , Cleveland, Ohio
| | - Thomas Broekelman
- 2 Department of Cell Biology and Physiology, Washington University at St. Louis , St. Louis, Missouri
| | - Robert P Mecham
- 2 Department of Cell Biology and Physiology, Washington University at St. Louis , St. Louis, Missouri
| | - Anand Ramamurthi
- 1 Department of Biomedical Engineering, Cleveland Clinic , Cleveland, Ohio.,3 Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University , Cleveland, Ohio
| |
Collapse
|
24
|
Boese AC, Chang L, Yin KJ, Chen YE, Lee JP, Hamblin MH. Sex differences in abdominal aortic aneurysms. Am J Physiol Heart Circ Physiol 2018; 314:H1137-H1152. [PMID: 29350999 DOI: 10.1152/ajpheart.00519.2017] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Abdominal aortic aneurysm (AAA) is a vascular disorder with a high case fatality rate in the instance of rupture. AAA is a multifactorial disease, and the etiology is still not fully understood. AAA is more likely to occur in men, but women have a greater risk of rupture and worse prognosis. Women are reportedly protected against AAA possibly by premenopausal levels of estrogen and are, on average, diagnosed at older ages than men. Here, we review the present body of research on AAA pathophysiology in humans, animal models, and cultured cells, with an emphasis on sex differences and sex steroid hormone signaling.
Collapse
Affiliation(s)
- Austin C Boese
- Department of Pharmacology, Tulane University School of Medicine , New Orleans, Louisiana
| | - Lin Chang
- Center for Advanced Models for Translational Sciences and Therapeutics, Department of Internal Medicine, University of Michigan , Ann Arbor, Michigan
| | - Ke-Jie Yin
- Department of Neurology, University of Pittsburgh School of Medicine , Pittsburgh, Pennsylvania
| | - Y Eugene Chen
- Center for Advanced Models for Translational Sciences and Therapeutics, Department of Internal Medicine, University of Michigan , Ann Arbor, Michigan
| | - Jean-Pyo Lee
- Department of Physiology, Tulane University School of Medicine , New Orleans, Louisiana.,Center for Stem Cell Research and Regenerative Medicine , New Orleans, Louisiana
| | - Milton H Hamblin
- Department of Pharmacology, Tulane University School of Medicine , New Orleans, Louisiana
| |
Collapse
|
25
|
A novel reproducible model of aortic aneurysm rupture. Surgery 2017; 163:397-403. [PMID: 29195736 DOI: 10.1016/j.surg.2017.10.003] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2017] [Revised: 10/04/2017] [Accepted: 10/05/2017] [Indexed: 01/14/2023]
Abstract
INTRODUCTION Given the unknown biologic antecedents before aortic aneurysm rupture, the purpose of this study was to establish a reproducible model of aortic aneurysm rupture. METHODS We fed 7-week-old apolipoprotein E deficient mice a high-fat diet for 4 weeks and osmotic infusion pumps containing Angiotensin II were implanted. Angiotensin II was delivered continuously for 4 weeks at either 1,000 ng/kg/min (n = 25) or 2,000 ng/kg/min (n = 29). A third group (n = 14) were given Angiotensin II at 2,000 ng/kg/min and 0.2% β-aminopropionitrile dissolved in drinking water. Surviving mice were killed 28 days after pump placement, aortic diameters were measured, and molecular analyses were performed. RESULTS Survival at 28 days was significantly different among groups with 80% survival in the 1,000 ng/kg/min group, 52% in the 2,000 ng/kg/min group, and only 14% in the Angiotensin II/β-aminopropionitrile group (P = .0001). Concordantly, rupture rates were statistically different among groups (8% versus 38% versus 79%, P < .0001). Rates of abdominal aortic aneurysm were 48%, 55%, and 93%, respectively, with statistically higher rates in the Angiotensin II/β-aminopropionitrile group compared with both the 1,000 ng and 2,000 ng Angiotensin II groups (P = .006 and P = .0165, respectively). Rates of thoracic aortic aneurysm formation were 12%, 52%, and 79% in the 3 groups with a statistically higher rate in the Angiotensin II/β-aminopropionitrile group compared with 1,000 ng group (P < .0001). CONCLUSIONS A reproducible model of aortic aneurysm rupture was developed with a high incidence of abdominal and thoracic aortic aneurysm. This model should enable further studies investigating the pathogenesis of aortic rupture, as well as allow for targeted strategies to prevent human aortic aneurysm rupture.
Collapse
|
26
|
Wang X, Khalil RA. Matrix Metalloproteinases, Vascular Remodeling, and Vascular Disease. ADVANCES IN PHARMACOLOGY (SAN DIEGO, CALIF.) 2017; 81:241-330. [PMID: 29310800 DOI: 10.1016/bs.apha.2017.08.002] [Citation(s) in RCA: 418] [Impact Index Per Article: 52.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Matrix metalloproteinases (MMPs) are a family of zinc-dependent endopeptidases that degrade various proteins in the extracellular matrix (ECM). Typically, MMPs have a propeptide sequence, a catalytic metalloproteinase domain with catalytic zinc, a hinge region or linker peptide, and a hemopexin domain. MMPs are commonly classified on the basis of their substrates and the organization of their structural domains into collagenases, gelatinases, stromelysins, matrilysins, membrane-type (MT)-MMPs, and other MMPs. MMPs are secreted by many cells including fibroblasts, vascular smooth muscle (VSM), and leukocytes. MMPs are regulated at the level of mRNA expression and by activation through removal of the propeptide domain from their latent zymogen form. MMPs are often secreted in an inactive proMMP form, which is cleaved to the active form by various proteinases including other MMPs. MMPs degrade various protein substrates in ECM including collagen and elastin. MMPs could also influence endothelial cell function as well as VSM cell migration, proliferation, Ca2+ signaling, and contraction. MMPs play a role in vascular tissue remodeling during various biological processes such as angiogenesis, embryogenesis, morphogenesis, and wound repair. Alterations in specific MMPs could influence arterial remodeling and lead to various pathological disorders such as hypertension, preeclampsia, atherosclerosis, aneurysm formation, as well as excessive venous dilation and lower extremity venous disease. MMPs are often regulated by endogenous tissue inhibitors of metalloproteinases (TIMPs), and the MMP/TIMP ratio often determines the extent of ECM protein degradation and tissue remodeling. MMPs may serve as biomarkers and potential therapeutic targets for certain vascular disorders.
Collapse
Affiliation(s)
- Xi Wang
- Vascular Surgery Research Laboratories, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, United States
| | - Raouf A Khalil
- Vascular Surgery Research Laboratories, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, United States.
| |
Collapse
|
27
|
Matrix Metalloproteinase Inhibitors as Investigational and Therapeutic Tools in Unrestrained Tissue Remodeling and Pathological Disorders. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2017; 148:355-420. [PMID: 28662828 DOI: 10.1016/bs.pmbts.2017.04.003] [Citation(s) in RCA: 95] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Matrix metalloproteinases (MMPs) are zinc-dependent proteolytic enzymes that degrade various proteins in the extracellular matrix (ECM). MMPs may also regulate the activity of membrane receptors and postreceptor signaling mechanisms and thereby affect cell function. The MMP family includes collagenases, gelatinases, stromelysins, matrilysins, membrane-type MMPs, and other MMPs. Inactive proMMPs are cleaved by other MMPs or proteases into active MMPs, which interact with various protein substrates in ECM and cell surface. MMPs regulate important biological processes such as vascular remodeling and angiogenesis and may be involved in the pathogenesis of cardiovascular disorders such as hypertension, atherosclerosis, and aneurysm. The role of MMPs is often assessed by measuring their mRNA expression, protein levels, and proteolytic activity using gel zymography. MMP inhibitors are also used to assess the role of MMPs in different biological processes and pathological conditions. MMP activity is regulated by endogenous tissue inhibitors of metalloproteinases (TIMPs), and the MMP/TIMP balance could determine the net MMP activity, ECM turnover, and tissue remodeling. Also, several synthetic MMP inhibitors have been developed. Synthetic MMP inhibitors include a large number of zinc-binding globulins (ZBGs), in addition to non-ZBGs and mechanism-based inhibitors. MMP inhibitors have been proposed as potential tools in the management of osteoarthritis, cancer, and cardiovascular disorders. However, most MMP inhibitors have broad-spectrum actions on multiple MMPs and could cause undesirable musculoskeletal side effects. Currently, doxycycline is the only MMP inhibitor approved by the Food and Drug Administration. New generation biological and synthetic MMP inhibitors may show greater MMP specificity and fewer side effects and could be useful in targeting specific MMPs, reducing unrestrained tissue remodeling, and the management of MMP-related pathological disorders.
Collapse
|
28
|
Rabkin SW. The Role Matrix Metalloproteinases in the Production of Aortic Aneurysm. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2017; 147:239-265. [PMID: 28413030 DOI: 10.1016/bs.pmbts.2017.02.002] [Citation(s) in RCA: 132] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Matrix metalloproteinases (MMPs) have been implicated in the pathogenesis of aortic aneurysm because the histology of thoracic aortic aneurysm (TAA) and abdominal aortic aneurysm (AAA) is characterized by the loss of smooth muscle cells in the aortic media and the destruction of extracellular matrix (ECM). Furthermore, AAA have evidence of inflammation and the cellular elements involved in inflammation such as macrophages can produce and/or activate MMPs This chapter focuses on human aortic aneurysm that are not due to specific known genetic causes because this type of aneurysm is the more common type. This chapter will also focus on MMP protein expression rather than on genetic data which may not necessarily translate to increased MMP protein expression. There are supporting data that certain MMPs are increased in the aortic wall. For TAA, it is most notably MMP-1, -9, -12, and -14 and MMP-2 when a bicuspid aortic valve is present. For AAA, it is MMP-1, -2, -3, -9, -12, and -13. The data are weaker or insufficient for the other MMPs. Several studies of gene polymorphisms support MMP-9 for TAA and MMP-3 for AAA as potentially important factors. The signaling pathways in the aorta that can lead to MMP activation include JNK, JAK/stat, osteopontin, and AMP-activated protein kinase alpha2. Substrates in the human vasculature for MMP-3, MMP-9, or MMP-14 include collagen, elastin, ECM glycoprotein, and proteoglycans. Confirmed and potential substrates for MMPs, maintain aortic size and function so that a reduction in their content relative to other components of the aortic wall may produce a failure to maintain aortic size leading to dilatation and aneurysm formation.
Collapse
|
29
|
Chan CYT, Cheuk BLY, Cheng SWK. Abdominal Aortic Aneurysm-Associated MicroRNA-516a-5p Regulates Expressions of Methylenetetrahydrofolate Reductase, Matrix Metalloproteinase-2, and Tissue Inhibitor of Matrix Metalloproteinase-1 in Human Abdominal Aortic Vascular Smooth Muscle Cells. Ann Vasc Surg 2017; 42:263-273. [PMID: 28288890 DOI: 10.1016/j.avsg.2016.10.062] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2016] [Revised: 10/19/2016] [Accepted: 10/20/2016] [Indexed: 12/22/2022]
Abstract
BACKGROUND MicroRNAs (miRNAs or miRs) have been highlighted to be involved in abdominal aortic aneurysm (AAA) with the emergence of recent miRNA microarray profiling studies. miR-516a-5p has been shown to be significantly overexpressed in vascular smooth muscle cells (VSMCs) from human AAA tissues from our previous microarray study, suggesting its crucial association with AAA. In addition, further bioinformatics analysis predicted methylenetetrahydrofolate reductase (MTHFR), which regulates homocysteine (Hcy) metabolism and is proposed to be a risk gene for AAA formation and to be the downregulation target of miR-516a-5p. However, the pathogenic role of miR-516a-5p in VSMCs for AAA formation remains unresolved. This study aims to investigate the role of miR-516a-5p in human VSMCs for AAA pathogenesis. METHODS miR-516a-5p was stably overexpressed and knocked down in VSMCs explant cultured from human abdominal aortic tissues by means of lentiviral system. The MTHFR protein expression was first examined by Western blotting. In addition, the protein expressions of several key components involved in AAA pathogenic features are as follows: matrix metalloproteinase (MMP)-2, MMP-9, tissue inhibitor of matrix metalloproteinase (TIMP)-1 and TIMP-2 for elastin degradation; collagen type 1 alpha 1 for compensatory collagen synthesis; monocyte chemoattractant protein-1 for inflammation, were also evaluated. Apoptotic level of VSMCs was examined by terminal deoxynucleotidyl transferase dUTP nick end labeling assay. RESULTS Results showed that protein expression of MTHFR was significantly downregulated on miR-516a-5p overexpression (P < 0.05) in VSMCs, whereas it was significantly upregulated on miR-516a-5p knockdown (P < 0.05). Of all the AAA key components investigated, only MMP-2 and TIMP-1 protein expressions were found altered. A significant increase in MMP-2 (P < 0.05) and decrease in TIMP-1 (P < 0.05) expressions were observed on miR-516a-5p overexpression in VSMCs. Apoptosis was not promoted on miR-516a-5p overexpression or knockdown in VSMCs. CONCLUSIONS Our findings suggested that miR-516a-5p may regulate MTHFR, MMP-2, and TIMP-1 expressions in human VSMCs, possibly promoting the disruption of Hcy metabolism and proteolytic degradation of elastin for AAA formation.
Collapse
Affiliation(s)
- Crystal Yin Tung Chan
- Division of Vascular Surgery, Department of Surgery, University of Hong Kong Medical Centre, Queen Mary Hospital, Pokfulam, Hong Kong, China.
| | - Bernice Lai Yee Cheuk
- Division of Vascular Surgery, Department of Surgery, University of Hong Kong Medical Centre, Queen Mary Hospital, Pokfulam, Hong Kong, China
| | - Stephen Wing Keung Cheng
- Division of Vascular Surgery, Department of Surgery, University of Hong Kong Medical Centre, Queen Mary Hospital, Pokfulam, Hong Kong, China
| |
Collapse
|
30
|
Vasic N, Glumac S, Pejic S, Amidzic LJ, Tadic Latinovic LJ, Dozic B, Hinic S, Maksimovic Z. Expression of Matrix Metalloproteinases and Endogenous Inhibitors in Abdominal Aortic Aneurysm and Aortoiliac Occlusive Disease (Syndrome Leriche). Folia Biol (Praha) 2017; 63:209-216. [PMID: 29687775 DOI: 10.14712/fb2017063050209] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2024]
Abstract
Matrix metalloproteinases (MMPs) and their tissue inhibitors (TIMPs) play a complex role in the pathogenesis of atherosclerosis. We compared (1) the histopathological findings in patients with abdominal aortic aneurysms (AAA) and aortoiliac occlusive disease (AOD); (2) the expression of MMP-2/MMP-9 and TIMP-1/TIMP-2 in aortic layers, inflammatory cells and smooth muscle cells (SMCs), aiming to identify the common underlying pathogenic mechanisms of the disease development. Samples were obtained from 30 patients with AAA and 30 with AOD. Aortic histology and immunohistochemistry were performed to evaluate inflammatory changes and MMP and TIMP expression. Thrombosis and ulceration were more frequent in AOD than in AAA. The MMP-9 expression was elevated in all aortic layers of AAA patients and in media/adventitia of AOD patients, mainly followed by lower expression of its inhibitor TIMP-1. Higher MMP-9 expression was also found in SMCs and macrophages of both AAA and AOD specimens, while higher TIMP-1/TIMP-2 were predominantly observed in the lymphocytes and macrophages of the aneurysm. These results showed that both conditions exhibited increased MMP-9 expression; however, the MMP expression pattern differed to some degree between the aneurysms and occlusive disease. The variations in molecular mechanisms underlying dilatative/stenosing disease warrant further investigation.
Collapse
Affiliation(s)
- N Vasic
- Department of Vascular Surgery; University Clinical Centre of the Republic of Srpska, Banja Luka, Bosnia and Herzegovina
| | - S Glumac
- Institute of Pathology, School of Medicine, University Clinical Hospital Centre "Bezanijska kosa"; University of Belgrade, Belgrade, Serbia
| | - S Pejic
- Laboratory for Molecular Biology and Endocrinology, "Vinca" Institute of Nuclear Sciences, University Clinical Hospital Centre "Bezanijska kosa"; University of Belgrade, Belgrade, Serbia
| | - L J Amidzic
- Department of Clinical Pathology; University Clinical Centre of the Republic of Srpska, Banja Luka, Bosnia and Herzegovina
| | - L J Tadic Latinovic
- Department of Clinical Pathology; University Clinical Centre of the Republic of Srpska, Banja Luka, Bosnia and Herzegovina
| | - B Dozic
- Institute of Pathology, School of Dental Medicine, University Clinical Hospital Centre "Bezanijska kosa"; University of Belgrade, Belgrade, Serbia
| | - S Hinic
- Department of Cardiology, University Clinical Hospital Centre "Bezanijska kosa"; University of Belgrade, Belgrade, Serbia
| | - Z Maksimovic
- Clinic for Vascular and Endovascular Surgery, Serbian Clinical Centre; University of Belgrade, Belgrade, Serbia
| |
Collapse
|
31
|
Liapis CD, Paraskevas KI. The pivotal role of matrix metalloproteinases in the development of human abdominal aortic aneurysms. Vasc Med 2016; 8:267-71. [PMID: 15125488 DOI: 10.1191/1358863x03vm504ra] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Abdominal aortic aneurysms (AAAs) represent a chronic degenerative condition and impart the risk of a life-threatening episode of rupture. Chronic inflammation and destructive remodeling of the extracellular matrix of the aortic wall constitute trademarks of this entity. Multiple studies have implicated a group of locally produced matrix endopeptidases-the matrix metalloproteinases (MMPs)-as the main culprits of this process. For this reason, extensive research on the Identification of the role of these enzymes, as well as possible alternative pharmacological treatments of AAAs, has taken place during the last few years. The exact role of the several members of the group of metalloproteinases has already been discovered, and conservative therapeutic strategies oriented towards these agents have been suggested, but a Definite treatment plan is still a controversial topic. The possible role of a genetic predisposition to AAAs is another crucial topic that remains to be determined, as it would render the confrontation of this condition much more efficient.
Collapse
Affiliation(s)
- Christos D Liapis
- Department of Vascular Surgery, Athens University Medical School, Athens, Greece
| | | |
Collapse
|
32
|
Matrix Metalloproteinases in Non-Neoplastic Disorders. Int J Mol Sci 2016; 17:ijms17071178. [PMID: 27455234 PMCID: PMC4964549 DOI: 10.3390/ijms17071178] [Citation(s) in RCA: 58] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2016] [Revised: 06/16/2016] [Accepted: 07/04/2016] [Indexed: 12/23/2022] Open
Abstract
The matrix metalloproteinases (MMPs) are zinc-dependent endopeptidases belonging to the metzincin superfamily. There are at least 23 members of MMPs ever reported in human, and they and their substrates are widely expressed in many tissues. Recent growing evidence has established that MMP not only can degrade a variety of components of extracellular matrix, but also can cleave and activate various non-matrix proteins, including cytokines, chemokines and growth factors, contributing to both physiological and pathological processes. In normal conditions, MMP expression and activity are tightly regulated via interactions between their activators and inhibitors. Imbalance among these factors, however, results in dysregulated MMP activity, which causes tissue destruction and functional alteration or local inflammation, leading to the development of diverse diseases, such as cardiovascular disease, arthritis, neurodegenerative disease, as well as cancer. This article focuses on the accumulated evidence supporting a wide range of roles of MMPs in various non-neoplastic diseases and provides an outlook on the therapeutic potential of inhibiting MMP action.
Collapse
|
33
|
Jacob T, Hingorani A, Ascher E. Role of Apoptosis and Proteolysis in the Pathogenesis of Iliac Artery Aneurysms. Vascular 2016; 13:34-42. [PMID: 15895673 DOI: 10.1258/rsmvasc.13.1.34] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
The objective of this study was to investigate the role of inflammation, programmed cell death, its molecular modulators, and proteolysis in the pathogenesis of iliac artery aneurysms (IAAs). Nineteen IAA specimens were obtained from patients undergoing elective surgical repair. All were males with ages ranging from 55 to 85 years (mean 73 years). Controls were iliac arteries ( n = 6) retrieved from surgical patients without aneurysmal disease. Standard histochemical techniques were used to assess elastic lamellae fragmentation and inflammatory infiltrate in aneurysmal and normal tissues. Identification of different types of cells in the aneurysm wall and detection of death-promoting molecules, Fas, p53, perforin, apoptosis-mediating bcl-2 family proteins, apoptotic death substrate, and poly(adenosine diphosphate–ribose) polymerase were performed immunohistochemically. Apoptosis was detected by terminal deoxynucleotidyl transferase–mediated digoxigenin–deoxyuridine triphosphate nick end-labeling (TUNEL) assay and caspase activity. Proteolytic activity was determined by 10% gelatin gel zymography. There is a conspicuous disruption and fragmentation of elastic lamellae in IAAs compared with normal arteries. Increased gelatinolytic activity was observed at 92, 72, and 67 kDa in the aneurysmal tissues. There was a significant loss of vascular smooth muscle cells (VSMCs) in the IAA walls compared with normal arteries ( p < .02). Large numbers of inflammatory cells were observed in the IAA specimens ( p = .01). Only aneurysmal arteries showed CD8+ T cells expressing death-promoting molecules. CD3+, CD8+, CD20+, CD30+, and CD68+ immunoreactive cells were significantly more prominent in the aneurysmal tissues than in the control arteries. There was a significant increase in the number of cells undergoing apoptosis in aneurysmal tissue than in the normal vessels ( p < .02), as well as in the expression of bax, p53, CPP-32, and Fas. Apoptotic cells and proapoptotic molecules predominantly localized to the inflammatory infiltrate. VSMC apoptosis was significant in IAAs. The data confirm the architectural disruption of the IAA wall and illustrate an apparent biologic response involving inflammatory infiltrate, apoptosis, and signaling molecules capable of initiating cell death. In addition to compromising the mechanical integrity of the vessel wall, VSMC loss may contribute to imbalance in the protein profile, accelerating extracellular matrix degradation that could favor IAA development.
Collapse
Affiliation(s)
- Theresa Jacob
- Division of Vascular Surgery, Department of Surgery, Maimonides Medical Center, Brooklyn, NY, USA
| | | | | |
Collapse
|
34
|
Groeneveld ME, van Burink MV, Begieneman MPV, Niessen HWM, Wisselink W, Eringa EC, Yeung KK. Activation of extracellular signal-related kinase in abdominal aortic aneurysm. Eur J Clin Invest 2016; 46:440-7. [PMID: 26988568 DOI: 10.1111/eci.12618] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/26/2015] [Accepted: 03/09/2016] [Indexed: 12/23/2022]
Abstract
BACKGROUND Extracellular matrix degeneration, caused by matrix metalloproteinase-2, facilitates smooth muscle cell migration leading to medial layer decline and, ultimately, abdominal aortic aneurysm. It remains unclear what exactly causes aneurysms to rupture, which leads to death in most patients. The extracellular signal-related kinase may be linked to the latter process. We aimed to clarify the role of extracellular signal-related kinase in aortic aneurysm development and rupture in patients. DESIGN Aortic fragments were harvested during open repair of nonruptured (n = 20) and ruptured (n = 8) aneurysms. As control, nondilated aortas (n = 6) were obtained during autopsy. We determined levels of phosphorylated and total extracellular signal-related kinase by Western blot, matrix metalloproteinase-2 by immunohistochemistry and medial layer thickness by conventional microscopy. RESULTS Nonruptured aneurysms had 1·8 times higher activation of extracellular signal-related kinase (ratio: phosphorylated/total) than controls (P = 0·011). However, the ruptured aneurysms had only 0·9 times the activation of controls (ns). Both nonruptured and ruptured aneurysms showed significantly higher matrix metalloproteinase-2 than controls (3·8 and 4·0-times, respectively; P < 0·005). Of the medial layer thickness in controls, the median was 1·5 mm, in nonruptured 1·0 mm and in ruptured aneurysms 0·7 mm. Activation of extracellular signal-related kinase correlated positively to medial layer thickness (Rs = 0·48; P = 0·014), but not to matrix metalloproteinase-2 (Rs = -0·36; P = 0·10). CONCLUSIONS In this study, nonruptured aneurysms are associated with increased extracellular signal-related kinase activation while ruptured aneurysms are not. Extracellular signal-related kinase was not related to total matrix metalloproteinase-2 expression. We therefore speculate that increased extracellular signal-related kinase, in response to medial layer decline, could be protective against aneurysm rupture.
Collapse
Affiliation(s)
- Menno E Groeneveld
- Department of Vascular Surgery, ICaR-VU, VU University Medical Center (VUmc), Amsterdam, the Netherlands.,Department of Physiology, ICaR-VU, VU University Medical Center (VUmc), Amsterdam, the Netherlands
| | - Max V van Burink
- Department of Vascular Surgery, ICaR-VU, VU University Medical Center (VUmc), Amsterdam, the Netherlands
| | - Mark P V Begieneman
- Department of Pathology and Cardiac Surgery, ICaR-VU, VU University Medical Center (VUmc), Amsterdam, the Netherlands.,Netherlands Forensic Institute, The Hague, the Netherlands
| | - Hans W M Niessen
- Department of Pathology and Cardiac Surgery, ICaR-VU, VU University Medical Center (VUmc), Amsterdam, the Netherlands
| | - Willem Wisselink
- Department of Vascular Surgery, ICaR-VU, VU University Medical Center (VUmc), Amsterdam, the Netherlands
| | - Ed C Eringa
- Department of Physiology, ICaR-VU, VU University Medical Center (VUmc), Amsterdam, the Netherlands
| | - Kak K Yeung
- Department of Vascular Surgery, ICaR-VU, VU University Medical Center (VUmc), Amsterdam, the Netherlands.,Department of Physiology, ICaR-VU, VU University Medical Center (VUmc), Amsterdam, the Netherlands
| |
Collapse
|
35
|
Li G, Yang L, Yuan H, Liu Y, He Y, Wu X, Jin X. Cold-inducible RNA-binding protein plays a central role in the pathogenesis of abdominal aortic aneurysm in a murine experimental model. Surgery 2016; 159:1654-1667. [PMID: 26936526 DOI: 10.1016/j.surg.2016.01.007] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2015] [Revised: 01/13/2016] [Accepted: 01/23/2016] [Indexed: 10/22/2022]
Abstract
BACKGROUND Cold-inducible RNA-binding protein (CIRP) is a recently identified proinflammatory cytokine. We hypothesize that CIRP is involved in the progression of abdominal aortic aneurysms (AAA) and that anti-CIRP treatment could inhibit this progression. METHODS We investigated CIRP expression in the sera and aneurysmal tissues of human AAA patients and elastase-induced AAA rats. To further examine the role of CIRP in the development of AAA, anti-CIRP antibody (1 mg/kg) or nonimmunized control immunoglobulin (Ig)G (1 mg/kg) was injected via the caudal vein in the experimental AAA model. To further investigate the underlying mechanisms, RAW 267.4 cells were stimulated with recombinant murine CIRP (rmCIRP). RESULTS In human AAA tissue, CIRP exhibited a 5.6-fold and 93% increase in mRNA and protein expression, respectively. In a rat AAA model, CIRP was upregulated significantly in a time-dependent manner in the serum and AAA tissue. The anti-CIRP antibody treatment significantly suppressed the dilation of experimental AAA. Simultaneously, inhibition of CIRP significantly attenuated the expression of matrix metalloproteinase (MMP)-2, MMP-9, tumor necrosis factor-α, and monocyte chemoattractant protein-1, and the number of CD68-positive macrophages in the experimental AAA tissue. In vitro, rmCIRP significantly increased MMP-9 messenger RNA expression in a dose-dependent manner by 1.2-fold, 2.9-fold, and 5.5-fold, respectively. Simultaneously, rmCIRP promoted RAW 264.7 cell migration, with an approximately 2.7-fold increase in the number of migrated cells. CONCLUSION Our findings demonstrate that CIRP mediates experimental AAA development by promoting the inflammatory response and inducing MMP-9 expression, demonstrating its potential as a novel target for inhibiting the progression of AAA.
Collapse
Affiliation(s)
- Gang Li
- Department of Vascular Surgery, Shandong Provincial Hospital affiliated to Shandong University, Jinan, China
| | - Le Yang
- Department of Vascular Surgery, Shandong Provincial Hospital affiliated to Shandong University, Jinan, China
| | - Hai Yuan
- Department of Vascular Surgery, Shandong Provincial Hospital affiliated to Shandong University, Jinan, China
| | - Yang Liu
- Department of Vascular Surgery, Shandong Provincial Hospital affiliated to Shandong University, Jinan, China
| | - Yuxiang He
- Department of Vascular Surgery, Shandong Provincial Hospital affiliated to Shandong University, Jinan, China
| | - Xuejun Wu
- Department of Vascular Surgery, Shandong Provincial Hospital affiliated to Shandong University, Jinan, China.
| | - Xing Jin
- Department of Vascular Surgery, Shandong Provincial Hospital affiliated to Shandong University, Jinan, China.
| |
Collapse
|
36
|
Timur UT, van Herwaarden JA, Mihajlovic D, De Jong P, Mali W, Moll FL. (18)F-FDG PET scanning of abdominal aortic aneurysms and correlation with molecular characteristics: a systematic review. EJNMMI Res 2015; 5:76. [PMID: 26695768 PMCID: PMC4688285 DOI: 10.1186/s13550-015-0153-8] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2015] [Accepted: 12/10/2015] [Indexed: 11/21/2022] Open
Abstract
Purpose The purpose of this study is to give an overview of studies investigating the role of fludeoxyglucose F18 (18F-FDG) positron emission tomography (PET) scanning in patients with aortic aneurysms with a focus on molecular characteristics of the aneurysm wall. Methods MEDLINE, EMBASE, and the Cochrane database were searched for relevant articles. After inclusion and exclusion, we selected 18 relevant articles reporting on 18F-FDG PET scanning of aortic aneurysms. Results The sample size of studies is limited, and there are no standardized imaging protocols and quantification methods. 18F-FDG PET scanning was shown to display molecular characteristics of the aortic wall. Different studies showed contradictory findings of aortic 18F-FDG uptake in aneurysm patients compared to controls. Conclusions Non-invasively determining molecular characteristics of aortic wall weakening might lead to better rupture and growth prediction. This might influence the decision of the surgeon between conservative and surgical treatment of aneurysms. To date, there is conflicted evidence regarding the use of 18F-FDG PET scanning to predict aneurysm rupture and growth. The role of 18F-FDG PET scanning in rupture risk prediction needs to be further investigated, and standardized imaging protocols and quantification methods need to be implemented. Electronic supplementary material The online version of this article (doi:10.1186/s13550-015-0153-8) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- U T Timur
- Department of Vascular Surgery, UMC Utrecht, Heidelberglaan 100, Utrecht, 3584 CX, Netherlands.
| | - J A van Herwaarden
- Department of Vascular Surgery, UMC Utrecht, Heidelberglaan 100, Utrecht, 3584 CX, Netherlands
| | - D Mihajlovic
- Department of Vascular Surgery, UMC Utrecht, Heidelberglaan 100, Utrecht, 3584 CX, Netherlands
| | - P De Jong
- Deparment of Radiology, UMC Utrecht, Heidelberglaan 100, Utrecht, 3584 CX, Netherlands
| | - W Mali
- Deparment of Radiology, UMC Utrecht, Heidelberglaan 100, Utrecht, 3584 CX, Netherlands
| | - F L Moll
- Department of Vascular Surgery, UMC Utrecht, Heidelberglaan 100, Utrecht, 3584 CX, Netherlands
| |
Collapse
|
37
|
Ghosh A, Pechota LVTA, Upchurch GR, Eliason JL. Cross-talk between macrophages, smooth muscle cells, and endothelial cells in response to cigarette smoke: the effects on MMP2 and 9. Mol Cell Biochem 2015; 410:75-84. [DOI: 10.1007/s11010-015-2539-3] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2015] [Accepted: 08/18/2015] [Indexed: 01/26/2023]
|
38
|
Cherifi H, Gogly B, Loison-Robert LS, Couty L, Ferré FC, Nassif A, Lafont A, Fournier BP. Comparative study of abdominal and thoracic aortic aneurysms: their pathogenesis and a gingival fibroblasts-based ex vivo treatment. SPRINGERPLUS 2015; 4:231. [PMID: 26110102 PMCID: PMC4475515 DOI: 10.1186/s40064-015-0976-9] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/27/2015] [Accepted: 04/13/2015] [Indexed: 11/10/2022]
Abstract
Aortic aneurysms (AAs) consist of slow proteolysis and loss of both collagen and elastin matrix in the aorta wall, leading to wall dilation, weakening and rupture in well-advanced lesions. This can occur in both abdominal aorta (Abdominal Aortic Aneurysm: AAA) and thoracic aorta (Thoracic Aortic Aneurysm: TAA). To date, no non-surgical therapy has been proposed to slow or stop AA progression. Previously published preclinical studies from our team using an aneurysm rabbit model showed a promising concept for treatment of AAs with gingival fibroblast (GFs) which are readily available cells. In this study, we investigated the possible tissue repair of human AAAs and TAAs using ex vivo models co-cultured with GFs. Histological analysis showed that TAA and AAA are two distinct pathologies. Both lesions presented destruction of the aorta wall, highly evidenced in AAA samples. The results have confirmed the presence of the bacterial Porphyromonas gingivalis (Pg) protein in all AAA samples, but not in TAA samples, indicating the possible role of an infectious factor in the developing and progression of AAA lesions compared to TAA. The co-culture of GFs with AA lesions shows increased expression of TIMP-1, the inhibitor of the aneurysm severity marker MMP-9. Our study indicates that GFs might ameliorate aorta wall reestablishment in both AA types by their regenerative and immunomodulatory capacities. It also demonstrates the possible infectious cause of AAA compared with TAA that may explain their different behavior.
Collapse
Affiliation(s)
- Hafida Cherifi
- Centre de recherche des cordeliers, INSERM UMRS 1138, Team 5, Laboratory of Molecular Oral Pathophysiology, Paris, France ; Paris-Est University, Créteil, France ; Paris-Descartes University, Paris, France ; A. Chenevier/H.Mondor Hospitals, Dental Department, APHP, Créteil, France
| | - Bruno Gogly
- Centre de recherche des cordeliers, INSERM UMRS 1138, Team 5, Laboratory of Molecular Oral Pathophysiology, Paris, France ; Paris-Est University, Créteil, France ; Paris-Descartes University, Paris, France ; A. Chenevier/H.Mondor Hospitals, Dental Department, APHP, Créteil, France
| | - Ludwig-Stanislas Loison-Robert
- Centre de recherche des cordeliers, INSERM UMRS 1138, Team 5, Laboratory of Molecular Oral Pathophysiology, Paris, France ; Paris-Est University, Créteil, France ; Paris-Descartes University, Paris, France ; A. Chenevier/H.Mondor Hospitals, Dental Department, APHP, Créteil, France
| | - Ludovic Couty
- Paris centre de recherche cardiovasculaire, INSERM UMRS 970, Team 11, Paris, France
| | - François Côme Ferré
- Centre de recherche des cordeliers, INSERM UMRS 1138, Team 5, Laboratory of Molecular Oral Pathophysiology, Paris, France ; Paris-Descartes University, Paris, France
| | - Ali Nassif
- Centre de recherche des cordeliers, INSERM UMRS 1138, Team 5, Laboratory of Molecular Oral Pathophysiology, Paris, France ; Paris-Est University, Créteil, France ; A. Chenevier/H.Mondor Hospitals, Dental Department, APHP, Créteil, France
| | - Antoine Lafont
- Paris centre de recherche cardiovasculaire, INSERM UMRS 970, Team 11, Paris, France
| | - Benjamin Pj Fournier
- Centre de recherche des cordeliers, INSERM UMRS 1138, Team 5, Laboratory of Molecular Oral Pathophysiology, Paris, France ; Paris-Diderot University, Paris, France ; Rothschild Hospital, Dental Department, AP-HP, Paris, France
| |
Collapse
|
39
|
Simmers P, Gishto A, Vyavahare N, Kothapalli CR. Nitric oxide stimulates matrix synthesis and deposition by adult human aortic smooth muscle cells within three-dimensional cocultures. Tissue Eng Part A 2015; 21:1455-70. [PMID: 25597545 DOI: 10.1089/ten.tea.2014.0363] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Vascular diseases are characterized by the over-proliferation and migration of aortic smooth muscle cells (SMCs), and degradation of extracellular matrix (ECM) within the vessel wall, leading to compromise in cell-cell and cell-matrix signaling pathways. Tissue engineering approaches to regulate SMC over-proliferation and enhance healthy ECM synthesis showed promise, but resulted in low crosslinking efficiency. Here, we report the benefits of exogenous nitric oxide (NO) cues, delivered from S-Nitrosoglutathione (GSNO), to cell proliferation and matrix deposition by adult human aortic SMCs (HA-SMCs) within three-dimensional (3D) biomimetic cocultures. A coculture platform with two adjacent, permeable 3D culture chambers was developed to enable paracrine signaling between vascular cells. HA-SMCs were cultured in these chambers within collagen hydrogels, either alone or in the presence of human aortic endothelial cells (HA-ECs) cocultures, and exogenously supplemented with varying GSNO dosages (0-100 nM) for 21 days. Results showed that EC cocultures stimulated SMC proliferation within GSNO-free cultures. With increasing GSNO concentration, HA-SMC proliferation decreased in the presence or absence of EC cocultures, while HA-EC proliferation increased. GSNO (100 nM) significantly enhanced the protein amounts synthesized by HA-SMCs, in the presence or absence of EC cocultures, while lower dosages (1-10 nM) offered marginal benefits. Multi-fold increases in the synthesis and deposition of elastin, glycosaminoglycans, hyaluronic acid, and lysyl oxidase crosslinking enzyme (LOX) were noted at higher GSNO dosages, and coculturing with ECs significantly furthered these trends. Similar increases in TIMP-1 and MMP-9 levels were noted within cocultures with increasing GSNO dosages. Such increases in matrix synthesis correlated with NO-stimulated increases in endothelial nitric oxide synthase (eNOS) and inducible nitric oxide synthase (iNOS) expression within EC and SMC cultures, respectively. Results attest to the benefits of delivering NO cues to suppress SMC proliferation and promote robust ECM synthesis and deposition by adult human SMCs, with significant applications in tissue engineering, biomaterial scaffold development, and drug delivery.
Collapse
Affiliation(s)
- Phillip Simmers
- 1 Department of Chemical and Biomedical Engineering, Cleveland State University , Cleveland, Ohio
| | | | | | | |
Collapse
|
40
|
Qi J, Yang P, Yi B, Huo Y, Chen M, Zhang J, Sun J. Heat shock protein 90 inhibition by 17-DMAG attenuates abdominal aortic aneurysm formation in mice. Am J Physiol Heart Circ Physiol 2015; 308:H841-52. [PMID: 25637544 DOI: 10.1152/ajpheart.00470.2014] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/07/2014] [Accepted: 11/23/2014] [Indexed: 01/12/2023]
Abstract
Abdominal aortic aneurysm (AAA) is a common degenerative vascular disease whose pathogenesis is associated with activation of multiple signaling pathways including Jun NH2-terminal kinases (JNK) and NF-κB. It is now well recognized that these pathways are chaperoned by the heat shock protein 90 (Hsp90), suggesting that inhibition of Hsp90 may be a novel strategy for inhibiting AAAs. The aim of this study is to investigate whether inhibition of Hsp90 by 17-DMAG (17-dimethyl-aminothylamino-17-demethoxy-geldanamycin) attenuates ANG II-induced AAA formation in mice, and, if so, to elucidate the mechanisms involved. Apolipoprotein E-null mice were infused with ANG II to induce AAA formation and simultaneously treated by intraperitoneal injection with either vehicle or 17-DMAG for 4 wk. ANG II infusion induced AAA formation in 80% of mice, which was accompanied by increased matrix metalloproteinase (MMP) activity, enhanced tissue inflammation, oxidative stress, and neovascularization. Importantly, these effects were inhibited by 17-DMAG treatment. Mechanistically, we showed that 17-DMAG prevented the formation and progression of AAA through its inhibitory effects on diverse biological pathways including 1) by blocking ANG II-induced phosphorylation of ERK1/2 and JNK that are critically involved in the regulation of MMPs in vascular smooth muscle cells, 2) by inhibiting IκB kinase expression and expression of MCP-1, and 3) by attenuating ANG II-stimulated angiogenic processes critical to AAA formation. Our results demonstrate that inhibition of Hsp90 by 17-DMAG effectively attenuates ANG II-induced AAA formation by simultaneously inhibiting vascular inflammation, extracellular matrix degradation, and angiogenesis, which are critical in the formation and progression of AAAs.
Collapse
Affiliation(s)
- Jia Qi
- Department of Pharmacy, Xinhua Hospital, Shanghai Jiaotong University, Shanghai, China; and Center for Translational Medicine, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Ping Yang
- Department of Pharmacy, Xinhua Hospital, Shanghai Jiaotong University, Shanghai, China; and
| | - Bing Yi
- Center for Translational Medicine, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Yan Huo
- Center for Translational Medicine, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Ming Chen
- Center for Translational Medicine, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Jian Zhang
- Department of Pharmacy, Xinhua Hospital, Shanghai Jiaotong University, Shanghai, China; and
| | - Jianxin Sun
- Center for Translational Medicine, Thomas Jefferson University, Philadelphia, Pennsylvania
| |
Collapse
|
41
|
Ghosh A, Pechota A, Coleman D, Upchurch GR, Eliason JL. Cigarette smoke-induced MMP2 and MMP9 secretion from aortic vascular smooth cells is mediated via the Jak/Stat pathway. Hum Pathol 2014; 46:284-94. [PMID: 25537973 DOI: 10.1016/j.humpath.2014.11.003] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/17/2014] [Revised: 11/10/2014] [Accepted: 11/14/2014] [Indexed: 10/24/2022]
Abstract
It is hypothesized that cigarette smoke may increase MMP2 and MMP9 secretion through Jak/Stat pathway in the aorta, thereby facilitating abdominal aortic aneurysm (AAA) formation/progression in smokers. We observed through zymograms that treatment of male rat aortic vascular smooth muscle cells (RASMC) with an aqueous extract of cigarette smoke (CSE) for 24 hours resulted in a significant increase in pro-MMP9 (P = .005) and a modest increase in pro-MMP2 (P = .055) production. Western blot with protein extracts from CSE-treated RASMC showed up-regulation of pStat3, pJak2, and T-Jak2 and unchanged levels of T-Stat3. Transfection of RASMC with small interfering RNAs for Jak2, Stat3, or both Jak2 and Stat3 significantly reduced pro-MMP9 (P < .005) and pro-MMP2 (P < .05) in medium of CSE-treated RASMC compared with control small interfering RNA-transfected cells. Immunoprecipitation with total Jak2 antibody showed increased pStat3 and T-Stat3 in the cytoplasm and nucleus of CSE-treated RASMC. Immunofluorescence revealed increased presence of pJak2, T-Jak2, pStat3, and T-Stat3 in the cytoplasm and nucleus of the CSE-treated cells. Treatment of control human tissues with CSE resulted in pro-MMP9 secretion and up-regulation of the Jak/Stat proteins. In addition, AAA tissues showed more pJak2 and pStat3 than control human tissues. Therefore, inhibiting the Jak/Stat pathway could be a potential therapeutic approach in the treatment of AAA.
Collapse
Affiliation(s)
- Abhijit Ghosh
- Section of Vascular Surgery, Department of Surgery, Jobst Vascular Research Laboratories, University of Michigan Medical School, Ann Arbor, MI 48109-5867
| | - Angela Pechota
- Section of Vascular Surgery, Department of Surgery, Jobst Vascular Research Laboratories, University of Michigan Medical School, Ann Arbor, MI 48109-5867
| | - Dawn Coleman
- Section of Vascular Surgery, Department of Surgery, Jobst Vascular Research Laboratories, University of Michigan Medical School, Ann Arbor, MI 48109-5867
| | - Gilbert R Upchurch
- University of Virginia, Division of Vascular and Endovascular Surgery, Charlottesville, VA 800679
| | - Jonathan L Eliason
- Section of Vascular Surgery, Department of Surgery, Jobst Vascular Research Laboratories, University of Michigan Medical School, Ann Arbor, MI 48109-5867.
| |
Collapse
|
42
|
Temporal regulation of venous extracellular matrix components during arteriovenous fistula maturation. J Vasc Access 2014; 16:93-106. [PMID: 25262757 DOI: 10.5301/jva.5000290] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/28/2014] [Indexed: 01/08/2023] Open
Abstract
PURPOSE The venous limb of arteriovenous fistulae (AVF) adapts to the arterial environment by dilation and wall thickening; however, the temporal regulation of the expression of extracellular matrix (ECM) components in the venous limb of the maturing AVF has not been well characterized. We used a murine model of AVF maturation that recapitulates human AVF maturation to determine the temporal pattern of expression of these ECM components. METHODS Aortocaval fistulae were created in C57BL/6J mice and the venous limb was analyzed on postoperative days 1, 3, 7, 21, and 42. A gene microarray analysis was performed on day 7; results were confirmed by qPCR, histology, and immunohistochemistry. Proteases, protease inhibitors, collagens, glycoproteins, and other non-collagenous proteins were characterized. RESULTS The maturing AVF has increased expression of many ECM components, including increased collagen and elastin. Matrix metalloproteinases (MMPs) and tissue inhibitor of metalloproteinase 1 (TIMP1) showed increased mRNA and protein expression during the first 7 days of maturation. Increased collagen and elastin expression was also significant at day 7. Expression of structural proteins was increased later during AVF maturation. Osteopontin (OPN) expression was increased at day 1 and sustained during AVF maturation. CONCLUSIONS During AVF maturation, there is significantly increased expression of ECM components, each of which shows distinct temporal patterns during AVF maturation. Increased expression of regulatory proteins such as MMP and TIMP precedes increased expression of structural proteins such as collagen and elastin, potentially mediating a controlled pattern of ECM degradation and vessel remodeling without structural failure.
Collapse
|
43
|
Ghosh A, Lu G, Su G, McEvoy B, Sadiq O, DiMusto PD, Laser A, Futchko JS, Henke PK, Eliason JL, Upchurch GR. Phosphorylation of AKT and abdominal aortic aneurysm formation. THE AMERICAN JOURNAL OF PATHOLOGY 2014; 184:148-58. [PMID: 24332015 DOI: 10.1016/j.ajpath.2013.09.016] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/26/2012] [Revised: 09/05/2013] [Accepted: 09/10/2013] [Indexed: 12/14/2022]
Abstract
It is hypothesized that differential AKT phosphorylation between sexes is important in abdominal aortic aneurysm (AAA) formation. Male C57BL/6 mice undergoing elastase treatment showed a typical AAA phenotype (80% over baseline, P < 0.001) and significantly increased phosphorylated AKT-308 (p308) and total-AKT (T-AKT) at day 14 compared with female mice. Elastase-treated Raw cells produced increased p308 and significant amounts of matrix metalloproteinase 9 (MMP-9), and these effects were suppressed by LY294002 treatment, a known AKT inhibitor. Male and female rat aortic smooth muscle cells treated with elastase for 1, 6, or 24 hours demonstrated that the p308/T-AKT and AKT-Ser-473/T-AKT ratios peaked at 6 hours and were significantly higher in the elastase-treated cells compared with controls. Similarly, male cells had higher phosphorylated AKT/T-AKT levels than female cells. LY294002 also inhibited elastase-induced p308 formation more in female smooth muscle cells than in males, and the corresponding cell media had less pro-MMP-9. AKT siRNA significantly decreased secretion of pro-MMP-9, as well as pro-MMP-2 and active MMP-2 from elastase-treated male rat aortic smooth muscle cells. IHC of male mice AAA aortas showed increased p308, AKT-Ser-473, and T-AKT compared with female mice. Aortas from male AAA patients had a significantly higher p308/T-AKT ratio than female AAA tissues. These data suggest that AKT phosphorylation is important in the upstream regulation of MMP activity, and that differential phosphorylation may be important in sex differences in AAA.
Collapse
Affiliation(s)
- Abhijit Ghosh
- Section of Vascular Surgery, Department of Surgery, Jobst Vascular Research Laboratories, University of Michigan, Ann Arbor, Michigan
| | - Guanyi Lu
- Division of Vascular and Endovascular Surgery, University of Virginia, Charlottesville, Virginia
| | - Gang Su
- Division of Vascular and Endovascular Surgery, University of Virginia, Charlottesville, Virginia
| | - Brendan McEvoy
- Section of Vascular Surgery, Department of Surgery, Jobst Vascular Research Laboratories, University of Michigan, Ann Arbor, Michigan
| | - Omar Sadiq
- Section of Vascular Surgery, Department of Surgery, Jobst Vascular Research Laboratories, University of Michigan, Ann Arbor, Michigan
| | - Paul D DiMusto
- Section of Vascular Surgery, Department of Surgery, Jobst Vascular Research Laboratories, University of Michigan, Ann Arbor, Michigan
| | - Adriana Laser
- Section of Vascular Surgery, Department of Surgery, Jobst Vascular Research Laboratories, University of Michigan, Ann Arbor, Michigan
| | - John S Futchko
- Section of Vascular Surgery, Department of Surgery, Jobst Vascular Research Laboratories, University of Michigan, Ann Arbor, Michigan
| | - Peter K Henke
- Section of Vascular Surgery, Department of Surgery, Jobst Vascular Research Laboratories, University of Michigan, Ann Arbor, Michigan
| | - Jonathan L Eliason
- Section of Vascular Surgery, Department of Surgery, Jobst Vascular Research Laboratories, University of Michigan, Ann Arbor, Michigan
| | - Gilbert R Upchurch
- Division of Vascular and Endovascular Surgery, University of Virginia, Charlottesville, Virginia.
| |
Collapse
|
44
|
Riches K, Angelini TG, Mudhar GS, Kaye J, Clark E, Bailey MA, Sohrabi S, Korossis S, Walker PG, Scott DJA, Porter KE. Exploring smooth muscle phenotype and function in a bioreactor model of abdominal aortic aneurysm. J Transl Med 2013; 11:208. [PMID: 24028184 PMCID: PMC3847145 DOI: 10.1186/1479-5876-11-208] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2013] [Accepted: 09/04/2013] [Indexed: 01/13/2023] Open
Abstract
Background Vascular smooth muscle cells (SMC) are central to arterial structure and function yet their involvement in the progression of abdominal aortic aneurysm (AAA) disease is not well studied. The progressive and silent nature of AAA in man essentially restricts research to the use of “end-stage” tissue recovered during surgical repair. This study aimed to generate an ex vivo model of AAA using protease-treated porcine carotid arteries maintained in a novel bioreactor, and to compare the structural and functional changes in SMC cultured from the recovered vessels with those from human tissue acquired at elective surgical repair. Methods Freshly isolated porcine arteries were pretreated with collagenase and/or elastase before culturing under flow in a bioreactor for 12 days. Human end-stage aneurysmal tissue and saphenous veins from age-matched controls were collected from patients undergoing surgery. SMC were cultured and characterised (immunocytochemistry, measurement of spread cell area) and assessed functionally at the level of proliferation (cell-counting) and matrix-metalloproteinase (MMP) secretion (gelatin zymography). Cellular senescence was investigated using β-galactosidase staining and apoptosis was quantified using a fluorescence-based caspase 3 assay. Results Co-expression of alpha-smooth muscle actin and smooth muscle myosin heavy chain confirmed all cell populations as SMC. Porcine SMC harvested and cultivated after collagenase/elastase pretreatment displayed a prominent “rhomboid” morphology, increased spread area (32%, P < 0.01), impaired proliferation (47% reduction, P < 0.05), increased senescence (52%, P < 0.001), susceptibility to apoptosis and reduced MMP-2 secretion (60% decrease, P < 0.01) compared with SMC from vehicle, collagenase or elastase pre-treated vessels. Notably, these changes were comparable to those observed in human AAA SMC which were 2.4-fold larger than non-aneurysmal SMC (P < 0.001) and exhibited reduced proliferation (39% reduction, P < 0.001), greater apoptosis (4-fold increase, P < 0.001), and increased senescence (61%, P < 0.05). Conclusions Combined collagenase/elastase exposure of porcine artery maintained in a bioreactor under flow conditions induced a SMC phenotype characteristic of those cultured from end-stage AAA specimens. This model has potential and versatility to examine temporal changes in SMC biology and to identify the molecular mechanisms leading to early aberrancies in SMC function. In the longer term this may inform new targets to maintain aortic SMC content and drive cells to a “reparative” phenotype at early stages of the disease.
Collapse
Affiliation(s)
- Kirsten Riches
- Division of Cardiovascular and Diabetes Research, Leeds Institute of Genetics, Health and Therapeutics (LIGHT), University of Leeds, Leeds, UK.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Fu XM, Yamawaki-Ogata A, Oshima H, Ueda Y, Usui A, Narita Y. Intravenous administration of mesenchymal stem cells prevents angiotensin II-induced aortic aneurysm formation in apolipoprotein E-deficient mouse. J Transl Med 2013; 11:175. [PMID: 23875706 PMCID: PMC3726376 DOI: 10.1186/1479-5876-11-175] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2013] [Accepted: 07/17/2013] [Indexed: 12/31/2022] Open
Abstract
Background Mesenchymal stem cells (MSCs) are known to be capable of suppressing inflammatory responses. We previously reported that intra-abdominal implantation of bone marrow-derived MSCs (BM-MSCs) sheet by laparotomy attenuated angiotensin II (AngII)-induced aortic aneurysm (AA) growth in apolipoprotein E-deficient (apoE−/−) mice through anti-inflammation effects. However, cell delivery by laparotomy is invasive; we here demonstrated the effects of multiple intravenous administrations of BM-MSCs on AngII-induced AA formation. Methods BM-MSCs were isolated from femurs and tibiae of male apoE−/− mice. Experimental AA was induced by AngII infusion for 28 days in apoE−/− mice. Mice received weekly intravenous administration of BM-MSCs (n=12) or saline (n=10). After 4 weeks, AA formation incidence, aortic diameter, macrophage accumulation, matrix metalloproteinase (MMP)’ activity, elastin content, and cytokines were evaluated. Results AngII induced AA formation in 100% of the mice in the saline group and 50% in the BM-MSCs treatment group (P < 0.05). A significant decrease of aortic diameter was observed in the BM-MSCs treatment group at ascending and infrarenal levels, which was associated with decreased macrophage infiltration and suppressed activities of MMP-2 and MMP-9 in aortic tissues, as well as a preservation of elastin content of aortic tissues. In addition, interleukin (IL)-1β, IL-6, and monocyte chemotactic protein-1 significantly decreased while insulin-like growth factor-1 and tissue inhibitor of metalloproteinases-2 increased in the aortic tissues of BM-MSCs treatment group. Conclusions Multiple intravenous administrations of BM-MSCs attenuated the development of AngII-induced AA in apoE−/− mice and may become a promising alternative therapeutic strategy for AA progression.
Collapse
Affiliation(s)
- Xian-ming Fu
- Department of Cardiothoracic Surgery, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya Aichi 466-8550, Japan
| | | | | | | | | | | |
Collapse
|
46
|
Qin Y, Cao X, Yang Y, Shi GP. Cysteine protease cathepsins and matrix metalloproteinases in the development of abdominal aortic aneurysms. Future Cardiol 2013; 9:89-103. [PMID: 23259477 DOI: 10.2217/fca.12.71] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Both cysteine protease cathepsins and matrix metalloproteinases are implicated in the pathogenesis of abdominal aortic aneurysms (AAAs) in humans and animals. Blood and aortic tissues from humans or animals with AAAs contain much higher levels of these proteases, and often lower levels of their endogenous inhibitors, than do blood and aortic tissues from healthy subjects. Protease- and protease inhibitor-deficient mice and synthetic protease inhibitors have affirmed that cysteinyl cathepsins and matrix metalloproteinases both participate directly in AAA development in several experimental model systems. Here, we summarize our current understanding of how proteases contribute to the pathogenesis of AAA, and discuss whether proteases or their inhibitors may serve as diagnostic biomarkers or potential therapeutic targets for this common human arterial disease.
Collapse
Affiliation(s)
- Yanwen Qin
- The Key Laboratory of Remodeling-Related Cardiovascular Diseases, Beijing An Zhen Hospital, Capital Medical University, Ministry of Education, Beijing Institute of Heart, Lung & Blood Vessel Diseases, Beijing 100029, China
| | | | | | | |
Collapse
|
47
|
Transforming growth factor-β and abdominal aortic aneurysms. Cardiovasc Pathol 2013; 22:126-32. [DOI: 10.1016/j.carpath.2012.07.005] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/08/2012] [Revised: 07/22/2012] [Accepted: 07/30/2012] [Indexed: 12/22/2022] Open
|
48
|
Abstract
Multiple medical therapies have been proposed to prevent abdominal aortic aneurysm expansion. Use of these medications, hormones, vitamins, and dietary products is based on their ability to alter the pathophysiology of continued aortic wall growth. In this review, the explanation of how these medications can achieve suppression of abdominal aortic aneurysm is explained in relation to their effect on the various aspects of aortic wall inflammation. Despite the large number of animal and observational studies, there remain very few randomized clinical trials to support use of any of these agents. However, there may be sufficient evidence to suggest that statins, doxycycline, vitamin E, cyclooxygenase-2 inhibitors, angiotensin-converting enzyme inhibitors, and angiotensin II receptor blockers can prove beneficial in some individuals.
Collapse
Affiliation(s)
- Russell Samson
- Mote Vascular Foundation, Inc., Florida State University Medical School, Sarasota, FL 34232, USA.
| |
Collapse
|
49
|
Ghosh A, DiMusto PD, Ehrlichman LK, Sadiq O, McEvoy B, Futchko JS, Henke PK, Eliason JL, Upchurch GR. The role of extracellular signal-related kinase during abdominal aortic aneurysm formation. J Am Coll Surg 2012; 215:668-680.e1. [PMID: 22917644 DOI: 10.1016/j.jamcollsurg.2012.06.414] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2012] [Revised: 06/22/2012] [Accepted: 06/27/2012] [Indexed: 12/30/2022]
Abstract
BACKGROUND It is hypothesized that activation of extracellular signal-related kinase (ERK) is critical in activating matrix metalloproteinases (MMPs) during abdominal aortic aneurysm (AAA) formation. STUDY DESIGN C57BL/6 male mice underwent either elastase or heat-inactivated elastase aortic perfusion (n = 9 per group). Mouse aortic smooth muscle cells were transfected with ERK-1 and 2 siRNA along with or without elastase treatment. Mouse and human aortic tissue were analyzed by Western blots, zymograms, and immunohistochemistry, and statistical analysis was done using Graphpad and Image J softwares. RESULTS Western blot and immunohistochemistry documented increased phospho-mitogen-activated protein kinase kinase-1/2 (pMEK-1/2; 153%, p = 0.270 by Western) and pERK (171%, p = 0.004 by Western blot) in the elastase perfused aortas. Male ERK-1(-/-) mice underwent elastase perfusion, and aortic diameter was determined at day 14. ERK-1(-/-) mice failed to develop AAA, and histologic analysis depicted intact collagen and elastin fibers in the aortas. Zymography of aortas of elastase-treated ERK-1(-/-) mice showed lower levels of proMMP2 (p < 0.005) and active MMP2 (p < 0.0001), as well as proMMP9 (p = 0.037) compared with C57BL/6 mice. siRNA transfection of ERK-1 and -2 significantly reduced formation of pro- and active MMP2 (p < 0.01 for both isoforms) in aortic smooth muscle cells treated with elastase in vitro. Human AAA tissue had significantly elevated levels of pMEK-1/2 (150%, p = 0.014) and pERK (159%, p = 0.013) compared with control tissues. CONCLUSIONS The MAPK (mitogen-activated protein kinase)/ERK pathway is an important modulator of MMPs during AAA formation. Targeting the ERK pathway by reagents that inhibit either the expression or phosphorylation of ERK isoforms could be a potential therapy to prevent AAA formation.
Collapse
Affiliation(s)
- Abhijit Ghosh
- Section of Vascular Surgery, Department of Surgery, Jobst Vascular Research Laboratories, University of Michigan, Ann Arbor, MI, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Midline abdominal wall incisional hernia after aortic reconstructive surgery: A prospective study. Surgery 2012; 151:882-8. [DOI: 10.1016/j.surg.2011.12.032] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2011] [Accepted: 12/22/2011] [Indexed: 11/20/2022]
|