1
|
Ding X, Li H, Li L, Yang Y, Chen Z. Potential drug targets for peripheral artery disease identified through Mendelian randomization analysis. Thromb J 2025; 23:52. [PMID: 40394637 PMCID: PMC12093625 DOI: 10.1186/s12959-025-00738-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2024] [Accepted: 05/08/2025] [Indexed: 05/22/2025] Open
Abstract
INTRODUCTION Peripheral Artery Disease (PAD) is a common cardiovascular condition marked by peripheral artery stenosis or occlusion. Despite treatment advancements, patients will still face vascular complications, highlighting the need for innovative therapies. Human proteins play crucial roles in biology and drug research. Mendelian randomization (MR) analysis, a gene-based method, is increasingly used in drug target identification. This study aims to identify PAD-associated plasma proteins through MR analysis for potential therapies. METHODS We first used GWAS data and seven pQTL datasets to identify plasma proteins causally linked to PAD through MR analysis. Then, we performed KEGG pathway enrichment analysis, Bayesian colocalization analysis, and MR-BMA analysis were carried out to investigate mechanisms and prioritize these proteins. Finally, we assessed the druggability of the target proteins using the DrugBank database. RESULTS MR analysis found four plasma proteins causally linked to PAD: MMP3 positively correlated with PAD, while CASS4, ISG15, and MMP1 exhibited negative associations. Bayesian colocalization analysis confirmed these relationships, and the MR-BMA analysis prioritized MMP1 as the main target. KEGG pathway enrichment analysis highlighted lipid metabolism and atherosclerosis pathways as central to these drug targets. The druggability evaluation indicated that drugs targeting these proteins are either in development or already in clinical use. CONCLUSION This study integrates genetic and proteomic data to identify therapeutic targets for PAD and evaluate their potential for drug development. The prioritization of MMP1 and ISG15 as key targets shows promise for PAD treatment, but further validation and clinical exploration of these findings are needed.
Collapse
Affiliation(s)
- Xu Ding
- Department of Vascular Surgery, Chui Yang Liu Hospital affiliated to Tsinghua University, Beijing, China
- Department of Vascular Surgery, Beijing Anzhen Hospital, Capital Medical University, Anzhen Road, Chaoyang District, Beijing, 100029, China
| | - Hui Li
- Department of Intervention, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Lihong Li
- Department of Vascular Surgery, Chui Yang Liu Hospital affiliated to Tsinghua University, Beijing, China
| | - Yongjiu Yang
- Department of Vascular Surgery, Chui Yang Liu Hospital affiliated to Tsinghua University, Beijing, China
| | - Zhong Chen
- Department of Vascular Surgery, Beijing Anzhen Hospital, Capital Medical University, Anzhen Road, Chaoyang District, Beijing, 100029, China.
| |
Collapse
|
2
|
Pan X, Zhang R, Lu B, Chen S, Chen H, Li M, Qin L, Song Z, Yang Y, Wang Z, Yan F, Sun A, Wu F, Zhang L, Wang J, Guzik TJ, Gao P. SM22α-Lineage Perivascular Stromal Cells Contribute to Abdominal Aortic Aneurysm. Circ Res 2025. [PMID: 40371535 DOI: 10.1161/circresaha.124.325750] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/20/2024] [Revised: 04/24/2025] [Accepted: 04/30/2025] [Indexed: 05/16/2025]
Abstract
BACKGROUND Perivascular adipose tissue (PVAT) is a key regulator of vascular dysfunction. Impairment of PVAT phenotypic plasticity with aging may play a role in vascular pathology including abdominal aortic aneurysms (AAAs). Yet, the mechanisms underlying PVAT plasticity in aneurysm pathogenesis remain elusive. METHODS Single-cell RNA sequencing was performed on perivascular stromal cells from young (2- to 3-month-old) and aged (18- to 20-month-old) mice. The expression of PGC-1α (peroxisome proliferator-activated receptor gamma coactivator-1α) was measured in PVAT of aged mice and human aneurysm samples. Loss- and gain-of-function approaches were used to investigate the role of SM22α-lineage perivascular stromal cell-derived PGC-1α in aneurysm development. Molecular mechanisms were explored through transcriptome and functional studies in young and aged mice, SM22αCre; Rosa26RFP/+; PGC1αf/f and SM22αCre; Rosa26RFP/+ mice with Ang II (angiotensin II)-induced and deoxycorticosterone acetate/salt-induced AAA models. RESULTS SM22α+ cells accumulated in PVAT of Ang II-treated aged mice and patients with aortic aneurysms. Single-cell RNA sequencing analysis revealed that aging disrupted the differentiation potential of SM22α-lineage perivascular stromal cells and led to reduced PGC-1α levels. PGC1α downregulation in PVAT was observed in both mouse AAA models and human aneurysm lesions. In mice with SM22α-driven PGC-1α deletion, Ang II-induced AAA formation was accompanied by perivascular stromal cell-to-myofibroblast differentiation. In vitro PGC1α knockdown suppressed nuclear YAP (Yes-associated protein) signaling, reducing adipocyte differentiation, while increasing MMP2 (matrix metalloproteinase 2)-secreting myofibroblasts. Furthermore, PGC-1α overexpression in aged mice or administration of the YAP signaling inhibitor verteporfin in SM22αCre; Rosa26RFP/+; PGC1αf/f mice restored PVAT function and conferred protection against aneurysm formation. Last, we used the radiomics analysis to noninvasively evaluate PVAT in the context of AAA severity in humans. CONCLUSIONS PGC-1α deficiency in SM22α-lineage stromal cells disrupts the balance between adipogenic and myofibrogenic differentiation through regulating YAP signaling, ultimately promoting aneurysm development. Radiomics assessment may present a promising noninvasive approach for PVAT evaluation in aneurysms, offering valuable potential for clinical research.
Collapse
Affiliation(s)
- Xiaoxi Pan
- Department of Cardiovascular Medicine, State Key Laboratory of Medical Genomics, Shanghai Key Laboratory of Hypertension, Department of Hypertension, Ruijin Hospital and Shanghai Institute of Hypertension, Shanghai Jiao Tong University School of Medicine, China. (X.P., R.Z., S.C., H.C., M.L., J.W., P.G.)
| | - Run Zhang
- Department of Cardiovascular Medicine, State Key Laboratory of Medical Genomics, Shanghai Key Laboratory of Hypertension, Department of Hypertension, Ruijin Hospital and Shanghai Institute of Hypertension, Shanghai Jiao Tong University School of Medicine, China. (X.P., R.Z., S.C., H.C., M.L., J.W., P.G.)
| | - Bingling Lu
- School of Biomedical Engineering, Shanghai Jiao Tong University, China (B.L., Z.S., L.Z.)
| | - Siyuan Chen
- Department of Cardiovascular Medicine, State Key Laboratory of Medical Genomics, Shanghai Key Laboratory of Hypertension, Department of Hypertension, Ruijin Hospital and Shanghai Institute of Hypertension, Shanghai Jiao Tong University School of Medicine, China. (X.P., R.Z., S.C., H.C., M.L., J.W., P.G.)
| | - Hongjin Chen
- Department of Cardiovascular Medicine, State Key Laboratory of Medical Genomics, Shanghai Key Laboratory of Hypertension, Department of Hypertension, Ruijin Hospital and Shanghai Institute of Hypertension, Shanghai Jiao Tong University School of Medicine, China. (X.P., R.Z., S.C., H.C., M.L., J.W., P.G.)
| | - Mengyao Li
- Department of Cardiovascular Medicine, State Key Laboratory of Medical Genomics, Shanghai Key Laboratory of Hypertension, Department of Hypertension, Ruijin Hospital and Shanghai Institute of Hypertension, Shanghai Jiao Tong University School of Medicine, China. (X.P., R.Z., S.C., H.C., M.L., J.W., P.G.)
| | - Le Qin
- Department of Radiology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, China. (L.Q., F.Y.)
| | - Zhiyun Song
- School of Biomedical Engineering, Shanghai Jiao Tong University, China (B.L., Z.S., L.Z.)
| | - Yi Yang
- Department of Cardiovascular Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, China. (Y.Y., Z.W.)
| | - Zhe Wang
- Department of Cardiovascular Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, China. (Y.Y., Z.W.)
| | - Fuhua Yan
- Department of Radiology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, China. (L.Q., F.Y.)
| | - Aijun Sun
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai, China (A.S.)
| | - Fang Wu
- Shanghai Clinical Research Center for Aging and Medicine, Department of Geriatrics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, China. (F.W.)
| | - Lichi Zhang
- School of Biomedical Engineering, Shanghai Jiao Tong University, China (B.L., Z.S., L.Z.)
| | - Jiguang Wang
- Department of Cardiovascular Medicine, State Key Laboratory of Medical Genomics, Shanghai Key Laboratory of Hypertension, Department of Hypertension, Ruijin Hospital and Shanghai Institute of Hypertension, Shanghai Jiao Tong University School of Medicine, China. (X.P., R.Z., S.C., H.C., M.L., J.W., P.G.)
| | - Tomasz J Guzik
- Department of Medicine, Jagiellonian University, Colelgium Medicum, Krakow, Poland (T.J.G.)
- Centre for Cardiovascular Sciences, Queen's Medical Research Institute, University of Edinburgh, United Kingdom (T.J.G.)
| | - Pingjin Gao
- Department of Cardiovascular Medicine, State Key Laboratory of Medical Genomics, Shanghai Key Laboratory of Hypertension, Department of Hypertension, Ruijin Hospital and Shanghai Institute of Hypertension, Shanghai Jiao Tong University School of Medicine, China. (X.P., R.Z., S.C., H.C., M.L., J.W., P.G.)
| |
Collapse
|
3
|
Ye W, Yang P, Jin M, Zou J, Zheng Z, Li Y, Zhang D, Ye W, Huang Z, Wang J, Liu Z. Dihydromyricetin mitigates abdominal aortic aneurysm via transcriptional and post-transcriptional regulation of heme oxygenase-1 in vascular smooth muscle cells. Acta Pharm Sin B 2025; 15:1514-1534. [PMID: 40370559 PMCID: PMC12069254 DOI: 10.1016/j.apsb.2025.02.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Revised: 09/15/2024] [Accepted: 11/26/2024] [Indexed: 05/16/2025] Open
Abstract
Abdominal aortic aneurysm (AAA) is a deadly condition of the aorta, carrying a significant risk of death upon rupture. Currently, there is a dearth of efficacious pharmaceutical interventions to impede the advancement of AAA and avert it from rupturing. Here, we investigated dihydromyricetin (DHM), one of the predominant bioactive flavonoids in Ampelopsis grossedentata (A. grossedentata), as a potential agent for inhibiting AAA. DHM effectively blocked the formation of AAA in angiotensin II-infused apolipoprotein E-deficient (ApoE-/-) mice. A combination of network pharmacology and whole transcriptome sequencing analysis revealed that DHM's anti-AAA action is linked to heme oxygenase (HO)-1 (Hmox-1 for the rodent gene) and hypoxia-inducible factor (HIF)-1α in vascular smooth muscle cells (VSMCs). Remarkably, DHM caused a robust rise (∼10-fold) of HO-1 protein expression in VSMCs, thereby suppressing VSMC inflammation and oxidative stress and preserving the VSMC contractile phenotype. Intriguingly, the therapeutic effect of DHM on AAA was largely abrogated by VSMC-specific Hmox1 knockdown in mice. Mechanistically, on one hand, DHM increased the transcription of Hmox-1 by triggering the nuclear translocation and activation of HIF-1α, but not nuclear factor erythroid 2-related factor 2 (NRF2). On the other hand, molecular docking, combined with cellular thermal shift assay (CETSA), isothermal titration calorimetry (ITC), drug affinity responsive target stability (DARTS), co-immunoprecipitation (Co-IP), and site mutant experiments revealed that DHM bonded to HO-1 at Lys243 and prevented its degradation, thereby resulting in considerable HO-1 buildup. In summary, our findings suggest that naturally derived DHM has the capacity to markedly enhance HO-1 expression in VSMCs, which may hold promise as a therapeutic strategy for AAA.
Collapse
Affiliation(s)
- Weile Ye
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research, International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Discovery of Chinese Ministry of Education (MOE), College of Pharmacy, Jinan University, Guangzhou 511436, China
| | - Pinglian Yang
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research, International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Discovery of Chinese Ministry of Education (MOE), College of Pharmacy, Jinan University, Guangzhou 511436, China
| | - Mei Jin
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research, International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Discovery of Chinese Ministry of Education (MOE), College of Pharmacy, Jinan University, Guangzhou 511436, China
| | - Jiami Zou
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research, International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Discovery of Chinese Ministry of Education (MOE), College of Pharmacy, Jinan University, Guangzhou 511436, China
| | - Zhihua Zheng
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research, International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Discovery of Chinese Ministry of Education (MOE), College of Pharmacy, Jinan University, Guangzhou 511436, China
| | - Yuanyuan Li
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research, International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Discovery of Chinese Ministry of Education (MOE), College of Pharmacy, Jinan University, Guangzhou 511436, China
| | - Dongmei Zhang
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research, International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Discovery of Chinese Ministry of Education (MOE), College of Pharmacy, Jinan University, Guangzhou 511436, China
| | - Wencai Ye
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research, International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Discovery of Chinese Ministry of Education (MOE), College of Pharmacy, Jinan University, Guangzhou 511436, China
| | - Zunnan Huang
- Key Laboratory of Big Data Mining and Precision Drug Design of Guangdong Medical University, Key Laboratory of Computer-Aided Drug Design of Dongguan City, Key Laboratory for Research and Development of Natural Drugs of Guangdong Province, School of Pharmacy, Guangdong Medical University, Dongguan 523808, China
| | - Jiaojiao Wang
- Key Laboratory of Big Data Mining and Precision Drug Design of Guangdong Medical University, Key Laboratory of Computer-Aided Drug Design of Dongguan City, Key Laboratory for Research and Development of Natural Drugs of Guangdong Province, School of Pharmacy, Guangdong Medical University, Dongguan 523808, China
| | - Zhiping Liu
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research, International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Discovery of Chinese Ministry of Education (MOE), College of Pharmacy, Jinan University, Guangzhou 511436, China
| |
Collapse
|
4
|
Mitu F, Adam CA, Richter P, Costache AD, Gavril RS, Cojocaru C, Țăruș A, Enache M, Cumpăt CM, Leon MM, Tinică G. Pericardial Fluid Biomarkers as Early Predictors for Postoperative Atrial Fibrillation-A Systematic Review. Diagnostics (Basel) 2025; 15:408. [PMID: 40002559 PMCID: PMC11854266 DOI: 10.3390/diagnostics15040408] [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: 08/30/2024] [Revised: 01/20/2025] [Accepted: 02/05/2025] [Indexed: 02/27/2025] Open
Abstract
(1) Background: Postoperative atrial fibrillation (POAF) is one of the most common complications of cardiac surgery, frequently occurring in the first 2-4 days after surgery. With a variable incidence depending on the type of surgery, research in recent years has focused on identifying predisposing factors with the aim of correcting them and thus decreasing the risk of cardiovascular and total morbidity and mortality. The analysis of pericardial fluid allowed the identification of biomarkers (interleukin-6, mitochondrial DNA, myeloperoxidase or natriuretic peptides) whose presence postoperatively was associated with increased risk of POAF. (2) Materials and Methods: We conducted a search on EMBASE and PubMed and identified 75 articles, of which 10 entered the final analysis. (3) Results: Patients who develop POAF accumulate large amounts of interleukin 6, mitochondrial DNA, myeloperoxidase, or secondary atrial natriuretic peptide as a consequence of the associated inflammatory status, atrial remodeling, or disturbance of homeostasis of various ions. There are also observations that their levels in the pericardium correlate with blood levels, but further studies on larger cohorts of patients are needed to provide new evidence in this regard. (4) Conclusions: Early recognition of patients at risk of developing POAF based on easy-to-dose and easy-to-use biochemical biomarkers, whose association with POAF has been demonstrated so far in small cohorts of patients, has both therapeutic and prognostic implications, which justifies further research on large cohorts of patients.
Collapse
Affiliation(s)
- Florin Mitu
- Department of Medical and Surgical Specialties I, II and III, “Grigore T. Popa” University of Medicine and Pharmacy, University Street No. 16, 700115 Iasi, Romania (A.-D.C.)
- Cardiovascular Rehabilitation Clinic, Clinical Rehabilitation Hospital, Pantelimon Halipa Street No. 14, 700661 Iasi, Romania
- Academy of Medical Sciences, 030167 Bucharest, Romania
- Academy of Romanian Scientists, 700050 Iasi, Romania
| | - Cristina Andreea Adam
- Department of Medical and Surgical Specialties I, II and III, “Grigore T. Popa” University of Medicine and Pharmacy, University Street No. 16, 700115 Iasi, Romania (A.-D.C.)
- Cardiovascular Rehabilitation Clinic, Clinical Rehabilitation Hospital, Pantelimon Halipa Street No. 14, 700661 Iasi, Romania
| | - Patricia Richter
- Department of Medical and Surgical Specialties I, II and III, “Grigore T. Popa” University of Medicine and Pharmacy, University Street No. 16, 700115 Iasi, Romania (A.-D.C.)
- Rheumatology Clinic, Clinical Rehabilitation Hospital, Pantelimon Halipa Street No. 14, 700661 Iasi, Romania
| | - Alexandru-Dan Costache
- Department of Medical and Surgical Specialties I, II and III, “Grigore T. Popa” University of Medicine and Pharmacy, University Street No. 16, 700115 Iasi, Romania (A.-D.C.)
- Cardiovascular Rehabilitation Clinic, Clinical Rehabilitation Hospital, Pantelimon Halipa Street No. 14, 700661 Iasi, Romania
| | - Radu Sebastian Gavril
- Department of Medical and Surgical Specialties I, II and III, “Grigore T. Popa” University of Medicine and Pharmacy, University Street No. 16, 700115 Iasi, Romania (A.-D.C.)
- Cardiovascular Rehabilitation Clinic, Clinical Rehabilitation Hospital, Pantelimon Halipa Street No. 14, 700661 Iasi, Romania
| | - Clementina Cojocaru
- Department of Medical and Surgical Specialties I, II and III, “Grigore T. Popa” University of Medicine and Pharmacy, University Street No. 16, 700115 Iasi, Romania (A.-D.C.)
- Cardiovascular Rehabilitation Clinic, Clinical Rehabilitation Hospital, Pantelimon Halipa Street No. 14, 700661 Iasi, Romania
| | - Andrei Țăruș
- Department of Medical and Surgical Specialties I, II and III, “Grigore T. Popa” University of Medicine and Pharmacy, University Street No. 16, 700115 Iasi, Romania (A.-D.C.)
- Cardiovascular Surgery Clinic, “Prof. Dr. George I. M. Georgescu” Cardiovascular Diseases Institute, 700503 Iasi, Romania
| | - Mihail Enache
- Department of Medical and Surgical Specialties I, II and III, “Grigore T. Popa” University of Medicine and Pharmacy, University Street No. 16, 700115 Iasi, Romania (A.-D.C.)
- Cardiovascular Surgery Clinic, “Prof. Dr. George I. M. Georgescu” Cardiovascular Diseases Institute, 700503 Iasi, Romania
| | - Carmen Marinela Cumpăt
- Department of Medical and Surgical Specialties I, II and III, “Grigore T. Popa” University of Medicine and Pharmacy, University Street No. 16, 700115 Iasi, Romania (A.-D.C.)
- Cardiovascular Rehabilitation Clinic, Clinical Rehabilitation Hospital, Pantelimon Halipa Street No. 14, 700661 Iasi, Romania
| | - Maria Magdalena Leon
- Department of Medical and Surgical Specialties I, II and III, “Grigore T. Popa” University of Medicine and Pharmacy, University Street No. 16, 700115 Iasi, Romania (A.-D.C.)
- Cardiovascular Rehabilitation Clinic, Clinical Rehabilitation Hospital, Pantelimon Halipa Street No. 14, 700661 Iasi, Romania
| | - Grigore Tinică
- Department of Medical and Surgical Specialties I, II and III, “Grigore T. Popa” University of Medicine and Pharmacy, University Street No. 16, 700115 Iasi, Romania (A.-D.C.)
- Academy of Medical Sciences, 030167 Bucharest, Romania
- Academy of Romanian Scientists, 700050 Iasi, Romania
- Cardiovascular Surgery Clinic, “Prof. Dr. George I. M. Georgescu” Cardiovascular Diseases Institute, 700503 Iasi, Romania
| |
Collapse
|
5
|
He H, Jiang T, Ding M, Zhu Y, Xu X, Huang Y, Yu W, Ou H. Nox1/PAK1 is required for angiotensin II-induced vascular inflammation and abdominal aortic aneurysm formation. Redox Biol 2025; 79:103477. [PMID: 39721498 PMCID: PMC11732235 DOI: 10.1016/j.redox.2024.103477] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Revised: 10/27/2024] [Accepted: 12/18/2024] [Indexed: 12/28/2024] Open
Abstract
NADPH oxidase 1 (Nox1) is a major isoform of Nox in vascular smooth muscle cells (VSMCs). VSMC activation and extracellular matrix (ECM) remodelling induce abdominal aortic aneurysm (AAA). In this study, we aim to determine the role of Nox1 in the progression of AAA and explore the underling mechanism. ApoE-/-Nox1SMCko mice in which the Nox1 gene was smooth muscle cell (SMC)-specifically deleted in ApoE-/- background, were infused with angiotensin II (Ang II) for 28 days. We found the Nox1 deficiency reduced AAA formation and increased survival compared with ApoE-/-Nox1y/flox mice. Abdominal aortic ROS and monocyts/macrophages were reduced in the ApoE-/-Nox1SMCko mice after Ang II-infusion. The SMC-specific Nox1 deletion caused less elastin fragments and lower matrix metalloproteinase (MMP) activities in the abdominal aorta. Further, we found the Nox1 protein interacted with p21-activated kinase 1 (PAK1) in Ang II-stimulated VSMCs. The PAK1, controlled by Nox1/ROS, promoted VSMC proliferation, migration and differentiation; this is associated with increased activities of vimentin and cofilin, and cytoskeleton modulation. Moreover, we found that the Nox1/PAK1 activated the downstream MAPKs (ERK1/2, p38 and JNKs) and NF-κB, and upregulated Sp1-mediated MMP2 expression upon Ang II-stimulation. Finally, overexpression of PAK1 in the ApoE-/-Nox1SMCko mice increased vascular elastic fibre degradation, pro-inflammatory cytokine expression and AAA incidence. Therefore, we conclude that Nox1, together with PAK1, facilitates Ang II-induced VSMC activation, vascular inflammation and ECM remodelling, and thus potentiates the AAA formation.
Collapse
MESH Headings
- Animals
- Aortic Aneurysm, Abdominal/metabolism
- Aortic Aneurysm, Abdominal/pathology
- Aortic Aneurysm, Abdominal/etiology
- Aortic Aneurysm, Abdominal/genetics
- Angiotensin II/adverse effects
- NADPH Oxidase 1/metabolism
- NADPH Oxidase 1/genetics
- Mice
- p21-Activated Kinases/metabolism
- p21-Activated Kinases/genetics
- Inflammation/metabolism
- Inflammation/pathology
- Reactive Oxygen Species/metabolism
- Mice, Knockout
- Myocytes, Smooth Muscle/metabolism
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/pathology
- Disease Models, Animal
- Humans
- Male
- Signal Transduction
Collapse
Affiliation(s)
- Hui He
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Guizhou Medical University, Gui'an, 561113, Guizhou, PR China
| | - Tianyu Jiang
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Guizhou Medical University, Gui'an, 561113, Guizhou, PR China
| | - Meng Ding
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Guizhou Medical University, Gui'an, 561113, Guizhou, PR China
| | - Yuan Zhu
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Guizhou Medical University, Gui'an, 561113, Guizhou, PR China
| | - Xiaoting Xu
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Guizhou Medical University, Gui'an, 561113, Guizhou, PR China
| | - Yashuang Huang
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Guizhou Medical University, Gui'an, 561113, Guizhou, PR China
| | - Wenfeng Yu
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Guizhou Medical University, Gui'an, 561113, Guizhou, PR China
| | - Hailong Ou
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Guizhou Medical University, Gui'an, 561113, Guizhou, PR China.
| |
Collapse
|
6
|
Alsabbagh Y, Erben Y, Vandenberg J, Farres H. New Trends of Personalized Medicine in the Management of Abdominal Aortic Aneurysm: A Review. J Pers Med 2024; 14:1148. [PMID: 39728062 DOI: 10.3390/jpm14121148] [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: 10/25/2024] [Revised: 11/30/2024] [Accepted: 12/06/2024] [Indexed: 12/28/2024] Open
Abstract
Abdominal aortic aneurysm (AAA) is a significant vascular condition characterized by the dilation of the abdominal aorta, presenting a substantial risk of rupture and associated high mortality rates. Current management strategies primarily rely on aneurysm diameter and growth rates to predict rupture risk and determine the timing of surgical intervention. However, this approach has limitations, as ruptures can occur in smaller AAAs below surgical thresholds, and many large AAAs remain stable without intervention. This review highlights the need for more precise and individualized assessment tools that integrate biomechanical parameters such as wall stress, wall strength, and hemodynamic factors. Advancements in imaging modalities like ultrasound elastography, computed tomography (CT) angiography, and magnetic resonance imaging (MRI), combined with artificial intelligence, offer enhanced capabilities to assess biomechanical indices and predict rupture risk more accurately. Incorporating these technologies can lead to personalized medicine approaches, improving decision-making regarding the timing of interventions. Additionally, emerging treatments focusing on targeted delivery of therapeutics to weakened areas of the aortic wall, such as nanoparticle-based drug delivery, stem cell therapy, and gene editing techniques like CRISPR-Cas9, show promise in strengthening the aortic wall and halting aneurysm progression. By validating advanced screening modalities and developing targeted treatments, the future management of AAA aims to reduce unnecessary surgeries, prevent ruptures, and significantly improve patient outcomes.
Collapse
Affiliation(s)
- Yaman Alsabbagh
- Division of Vascular and Endovascular Surgery, Mayo Clinic, Jacksonville, FL 32224, USA
| | - Young Erben
- Division of Vascular and Endovascular Surgery, Mayo Clinic, Jacksonville, FL 32224, USA
| | - Jonathan Vandenberg
- Division of Vascular and Endovascular Surgery, Mayo Clinic, Jacksonville, FL 32224, USA
| | - Houssam Farres
- Division of Vascular and Endovascular Surgery, Mayo Clinic, Jacksonville, FL 32224, USA
| |
Collapse
|
7
|
Chu S, Shan D, He L, Yang S, Feng Y, Zhang Y, Yu J. Anemoside B4 attenuates abdominal aortic aneurysm by limiting smooth muscle cell transdifferentiation and its mediated inflammation. Front Immunol 2024; 15:1412022. [PMID: 38881898 PMCID: PMC11176519 DOI: 10.3389/fimmu.2024.1412022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Accepted: 05/17/2024] [Indexed: 06/18/2024] Open
Abstract
Abdominal aortic aneurysm (AAA) is a degenerative disease characterized by local abnormal dilation of the aorta accompanied by vascular smooth muscle cell (VSMC) dysfunction and chronic inflammation. VSMC dedifferentiation, transdifferentiation, and increased expression of matrix metalloproteinases (MMPs) are essential causes of AAA formation. Previous studies from us and others have shown that Anemoside B4 (AB4), a saponin from Pulsatilla chinensis, has anti-inflammatory, anti-tumor, and regulatory effects on VSMC dedifferentiation. The current study aimed to investigate whether AB4 inhibits AAA development and its underlying mechanisms. By using an Ang II induced AAA model in vivo and cholesterol loading mediated VSMC to macrophage transdifferentiation model in vitro, our study demonstrated that AB4 could attenuate AAA pathogenesis, prevent VSMC dedifferentiation and transdifferentiation to macrophage-like cells, decrease vascular inflammation, and suppress MMP expression and activity. Furthermore, KLF4 overexpression attenuated the effects of AB4 on VSMC to macrophage-like cell transition and VSMC inflammation in vitro. In conclusion, AB4 protects against AAA formation in mice by inhibiting KLF4 mediated VSMC transdifferentiation and inflammation. Our study provides the first proof of concept of using AB4 for AAA management.
Collapse
MESH Headings
- Animals
- Aortic Aneurysm, Abdominal/pathology
- Aortic Aneurysm, Abdominal/metabolism
- Aortic Aneurysm, Abdominal/prevention & control
- Aortic Aneurysm, Abdominal/chemically induced
- Cell Transdifferentiation/drug effects
- Kruppel-Like Factor 4/metabolism
- Mice
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/drug effects
- Inflammation/metabolism
- Saponins/pharmacology
- Disease Models, Animal
- Male
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/pathology
- Muscle, Smooth, Vascular/drug effects
- Mice, Inbred C57BL
- Macrophages/metabolism
- Macrophages/drug effects
- Macrophages/immunology
- Angiotensin II/pharmacology
- Humans
Collapse
Affiliation(s)
- Shuhan Chu
- Center for Translational Medicine, Jiangxi University of Chinese Medicine, Nanchang, Jiangxi, China
- Department of Cardiovascular Sciences and Center for Metabolic Disease Research, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, United States
| | - Dan Shan
- Department of Cardiovascular Sciences and Center for Metabolic Disease Research, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, United States
| | - Luling He
- Center for Translational Medicine, Jiangxi University of Chinese Medicine, Nanchang, Jiangxi, China
| | - Shilin Yang
- National Pharmaceutical Engineering Center (NPEC) for Solid Preparation in Chinese Herbal Medicine, Nanchang, Jiangxi, China
| | - Yulin Feng
- National Pharmaceutical Engineering Center (NPEC) for Solid Preparation in Chinese Herbal Medicine, Nanchang, Jiangxi, China
| | - Yifeng Zhang
- Center for Translational Medicine, Jiangxi University of Chinese Medicine, Nanchang, Jiangxi, China
| | - Jun Yu
- Department of Cardiovascular Sciences and Center for Metabolic Disease Research, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, United States
| |
Collapse
|
8
|
Rabia B, Thanigaimani S, Golledge J. The potential involvement of glycocalyx disruption in abdominal aortic aneurysm pathogenesis. Cardiovasc Pathol 2024; 70:107629. [PMID: 38461960 DOI: 10.1016/j.carpath.2024.107629] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/07/2024] [Revised: 03/03/2024] [Accepted: 03/04/2024] [Indexed: 03/12/2024] Open
Abstract
BACKGROUND Abdominal aortic aneurysm is a weakening and expansion of the abdominal aorta. Currently, there is no drug treatment to limit abdominal aortic aneurysm growth. The glycocalyx is the outermost layer of the cell surface, mainly composed of glycosaminoglycans and proteoglycans. OBJECTIVE The aim of this review was to identify a potential relationship between glycocalyx disruption and abdominal aortic aneurysm pathogenesis. METHODS A narrative review of relevant published research was conducted. RESULTS Glycocalyx disruption has been reported to enhance vascular permeability, impair immune responses, dysregulate endothelial function, promote extracellular matrix remodeling and modulate mechanotransduction. All these effects are implicated in abdominal aortic aneurysm pathogenesis. Glycocalyx disruption promotes inflammation through exposure of adhesion molecules and release of proinflammatory mediators. Glycocalyx disruption affects how the endothelium responds to shear stress by reducing nitric oxide availabilty and adversely affecting the storage and release of several antioxidants, growth factors, and antithromotic proteins. These changes exacerbate oxidative stress, stimulate vascular smooth muscle cell dysfunction, and promote thrombosis, all effects implicated in abdominal aortic aneurysm pathogenesis. Deficiency of key component of the glycocalyx, such as syndecan-4, were reported to promote aneurysm formation and rupture in the angiotensin-II and calcium chloride induced mouse models of abdominal aortic aneurysm. CONCLUSION This review provides a summary of past research which suggests that glycocalyx disruption may play a role in abdominal aortic aneurysm pathogenesis. Further research is needed to establish a causal link between glycocalyx disruption and abdominal aortic aneurysm development.
Collapse
Affiliation(s)
- Bibi Rabia
- Queensland Research Centre for Peripheral Vascular Disease, College of Medicine and Dentistry, James Cook University, Townsville, Queensland 4811, Australia; Department of Pharmacy, Hazara University, Mansehra 21300, Pakistan
| | - Shivshankar Thanigaimani
- Queensland Research Centre for Peripheral Vascular Disease, College of Medicine and Dentistry, James Cook University, Townsville, Queensland 4811, Australia; The Australian Institute of Tropical Health and Medicine, James Cook University, Townsville, Queensland 4811, Australia
| | - Jonathan Golledge
- Queensland Research Centre for Peripheral Vascular Disease, College of Medicine and Dentistry, James Cook University, Townsville, Queensland 4811, Australia; The Australian Institute of Tropical Health and Medicine, James Cook University, Townsville, Queensland 4811, Australia; The Department of Vascular and Endovascular Surgery, The Townsville University Hospital, Townsville, Queensland 4810, Australia.
| |
Collapse
|
9
|
Lee JH, Vu HD, Park MH, Huynh PT, Youn SW, Kwon DR. Microcurrent wave alleviates mouse intracranial arterial dolichoectasia development. Sci Rep 2024; 14:7496. [PMID: 38553592 PMCID: PMC10980802 DOI: 10.1038/s41598-024-58333-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Accepted: 03/27/2024] [Indexed: 04/02/2024] Open
Abstract
Intracranial arterial dolichoectasia (IADE) is associated with the interaction of hypertension and inflammation, and microcurrent can be effective in hypertension. Therefore, this study aimed to investigate the therapeutic effect of microcurrent electrical stimulation in a mouse IADE model. This study randomly categorized 20 mice into five groups: group 1-C (healthy control), group 2-D (IADE model), group 3-M + D (microcurrent administration before nephrectomy and until brain surgery), group 4-D + M (microcurrent administration for 4 weeks following brain surgery), and group 5-M (microcurrent administration for 4 weeks). Cerebral artery diameter and thickness and cerebral arterial wall extracellular matrix components were assessed. Among the five groups, group 2-D showed significantly higher cerebral arterial wall diameter (117.79 ± 17.05 µm) and proportion of collagen (42.46 ± 14.12%) and significantly lower arterial wall thickness (9.31 ± 2.26 µm) and proportion of smooth muscle cell (SMC) and elastin in the cerebral arterial wall (SMC: 38.05 ± 10.32%, elastin: 11.11 ± 6.97%). Additionally, group 4-D + M exhibited a non-significantly lower diameter (100.28 ± 25.99 µm) and higher thickness (12.82 ± 5.17 µm). Group 5-M demonstrated no evidence of toxicity in the liver and brain. The pilot study revealed that microcurrent is effective in preventing IADE development, although these beneficial effects warrant further investigation.
Collapse
Affiliation(s)
- Jae Hee Lee
- Department of Rehabilitation Medicine, Daegu Catholic University School of Medicine, Daegu, South Korea
| | - Huy Duc Vu
- Department of Radiology, Kyungpook National University, Daegu, South Korea
| | - Min Hee Park
- Department of Radiology, Kyungpook National University, Daegu, South Korea
| | - Phuong Tu Huynh
- Department of Radiology, Kyungpook National University, Daegu, South Korea
| | - Sung Won Youn
- Department of Radiology, Kyungpook National University, Daegu, South Korea.
| | - Dong Rak Kwon
- Department of Rehabilitation Medicine, Daegu Catholic University School of Medicine, Daegu, South Korea.
| |
Collapse
|
10
|
Menges AL, Nackenhorst M, Müller JR, Engl ML, Hegenloh R, Pelisek J, Geibelt E, Hofmann A, Reeps C, Biro G, Eckstein HH, Zimmermann A, Magee D, Falk M, Sachs N, Busch A. Completing the view - histologic insights from circular AAA specimen including 3D imaging : A methodologic approach towards histologic analysis of circumferential AAA samples. Diagn Pathol 2023; 18:73. [PMID: 37308870 PMCID: PMC10259026 DOI: 10.1186/s13000-023-01359-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2023] [Accepted: 05/23/2023] [Indexed: 06/14/2023] Open
Abstract
Abdominal aortic aneurysm (AAA) is a pathologic enlargement of the infrarenal aorta with an associated risk of rupture. However, the responsible mechanisms are only partially understood. Based on murine and human samples, a heterogeneous distribution of characteristic pathologic features across the aneurysm circumference is expected. Yet, complete histologic workup of the aneurysm sac is scarcely reported. Here, samples from five AAAs covering the complete circumference partially as aortic rings are investigated by histologic means (HE, EvG, immunohistochemistry) and a new method embedding the complete ring. Additionally, two different methods of serial histologic section alignment are applied to create a 3D view. The typical histopathologic features of AAA, elastic fiber degradation, matrix remodeling with collagen deposition, calcification, inflammatory cell infiltration and thrombus coverage were distributed without recognizable pattern across the aneurysm sac in all five patients. Analysis of digitally scanned entire aortic rings facilitates the visualization of these observations. Immunohistochemistry is feasible in such specimen, however, tricky due to tissue disintegration. 3D image stacks were created using open-source and non-generic software correcting for non-rigid warping between consecutive sections. Secondly, 3D image viewers allowed visualization of in-depth changes of the investigated pathologic hallmarks. In conclusion, this exploratory descriptive study demonstrates a heterogeneous histomorphology around the AAA circumference. Warranting an increased sample size, these results might need to be considered in future mechanistic research, especially in reference to intraluminal thrombus coverage. 3D histology of such circular specimen could be a valuable visualization tool for further analysis.
Collapse
Affiliation(s)
- Anna-Leonie Menges
- Department for Vascular Surgery, University Hospital Zurich, Zurich, Switzerland
| | - Maja Nackenhorst
- Department of Pathology, Medical University of Vienna, Vienna, Austria
| | - Johannes R Müller
- DFG Cluster of Excellence "Physics of Life", TU Dresden, Dresden, Germany
| | - Marie-Luise Engl
- Technical University Munich, Department for Vascular and Endovascular Surgery, Klinikum Rechts der Isar, Munich, Germany
| | - Renate Hegenloh
- Technical University Munich, Department for Vascular and Endovascular Surgery, Klinikum Rechts der Isar, Munich, Germany
| | - Jaroslav Pelisek
- Department for Vascular Surgery, University Hospital Zurich, Zurich, Switzerland
| | - Ellen Geibelt
- Light Microscopy Facility, Center for Molecular and Cellular Bioengineering (CMCB), Technische Universität Dresden, Dresden, Germany
| | - Anja Hofmann
- Department for Visceral-, Thoracic and Vascular Surgery, Medical Faculty and University Hospital Carl Gustav Carus, TUD Dresden University of Technology, Fetscherstrasse 74, Dresden, Germany
| | - Christian Reeps
- Department for Visceral-, Thoracic and Vascular Surgery, Medical Faculty and University Hospital Carl Gustav Carus, TUD Dresden University of Technology, Fetscherstrasse 74, Dresden, Germany
| | - Gabor Biro
- Technical University Munich, Department for Vascular and Endovascular Surgery, Klinikum Rechts der Isar, Munich, Germany
| | - Hans-Henning Eckstein
- Technical University Munich, Department for Vascular and Endovascular Surgery, Klinikum Rechts der Isar, Munich, Germany
- German Center for Cardiovascular Research (DZHK), Munich Heart Alliance, Berlin, Germany
| | - Alexander Zimmermann
- Department for Vascular Surgery, University Hospital Zurich, Zurich, Switzerland
| | - Derek Magee
- HeteroGenius Limited, Leeds, UK
- School of Computing, University of Leeds, Leeds, UK
| | - Martin Falk
- Scientific Visualization Group, Department of Science and Technology (ITN), Linköping University, Linköping, Sweden
| | - Nadja Sachs
- Technical University Munich, Department for Vascular and Endovascular Surgery, Klinikum Rechts der Isar, Munich, Germany
- German Center for Cardiovascular Research (DZHK), Munich Heart Alliance, Berlin, Germany
| | - Albert Busch
- Technical University Munich, Department for Vascular and Endovascular Surgery, Klinikum Rechts der Isar, Munich, Germany.
- Department for Visceral-, Thoracic and Vascular Surgery, Medical Faculty and University Hospital Carl Gustav Carus, TUD Dresden University of Technology, Fetscherstrasse 74, Dresden, Germany.
| |
Collapse
|
11
|
Reutersberg B, Metschl S, Salvermoser M, Eckstein HH, Knappich C, Maegdefessel L, Jaroslav P, Busch A. CXCR4 - a possible serum marker for risk stratification of abdominal aortic aneurysms. VASA 2023; 52:124-132. [PMID: 36519232 DOI: 10.1024/0301-1526/a001049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Background: Abdominal aortic aneurysm (AAA) rupture is still associated with a mortality rate of 80-90%. Imaging techniques or molecular fingerprinting for patient-specific risk stratification to identify pending rupture are still lacking. The chemokine (C-X-C motif) receptor (CXCR4) activation by CXCL12 ligand has been identified as a marker of inflammation and atherosclerosis, associated with AAA. Both are highly expressed in the aortic aneurysm wall. However, it is still unclear whether different expression levels of CXCR4 and CXCL12 can distinguish ruptured AAAs (rAAA) from intact AAAs (iAAA). Patients and methods: Abdominal aortic tissue samples (rAAA: n=29; iAAA: n=54) were excised during open aortic repair. Corresponding serum samples from these patients (n=9 from rAAAs; n=47 from iAAA) were drawn pre-surgery. Healthy aortic tissue samples (n=8) obtained from adult kidney donors during transplantation and serum samples from healthy adult volunteers were used as controls (n=5 each). Results: CXCR4 was mainly expressed in the media of the aneurysmatic tissue. Focal positive staining was also observed in areas of inflammatory infiltrates within the adventitia. In tissue lysates, no significant differences between iAAA, rAAA, and healthy controls were observed upon ELISA analysis. In serum samples, the level of CXCR4 was significantly increased in rAAA by 4-fold compared to healthy controls (p=0.011) and 3.0-fold for rAAA compared to iAAA (p<0.001). Furthermore a significant positive correlation between aortic diameter and serum CXCR4 concentration was found for both, iAAA and rAAA (p=0.042). Univariate logistic regression analysis showed that increased CXCR4 serum concentrations were associated with AAA rupture (OR: 4.28, 95% CI: 1.95-12.1, p=0.001). Conclusions: CXCR4 concentration was significantly increased in serum of rAAA patients and showed a significant correlation with an increased aortic diameter. The level of CXCR4 in serum was associated with a more than 4-fold risk increase for rAAA and thus could possibly serve as a biomarker in the future. However, further validation in larger studies is required.
Collapse
Affiliation(s)
- Benedikt Reutersberg
- Department for Vascular and Endovascular Surgery, Munich Vascular Biobank, Munich Aortic Center (MAC), University Hospital Klinikum rechts der Isar, Technical University of Munich, Germany.,Department for Vascular Surgery, University Hospital Zurich, Switzerland
| | - Susanne Metschl
- Department for Vascular and Endovascular Surgery, Munich Vascular Biobank, Munich Aortic Center (MAC), University Hospital Klinikum rechts der Isar, Technical University of Munich, Germany
| | - Michael Salvermoser
- Department for Vascular and Endovascular Surgery, Munich Vascular Biobank, Munich Aortic Center (MAC), University Hospital Klinikum rechts der Isar, Technical University of Munich, Germany.,Department of Pulmonary and Allergy, Dr. von Hauner Children's Hospital, LMU University of Munich, Germany
| | - Hans-Henning Eckstein
- Department for Vascular and Endovascular Surgery, Munich Vascular Biobank, Munich Aortic Center (MAC), University Hospital Klinikum rechts der Isar, Technical University of Munich, Germany
| | - Christoph Knappich
- Department for Vascular and Endovascular Surgery, Munich Vascular Biobank, Munich Aortic Center (MAC), University Hospital Klinikum rechts der Isar, Technical University of Munich, Germany
| | - Lars Maegdefessel
- Department for Vascular and Endovascular Surgery, Munich Vascular Biobank, Munich Aortic Center (MAC), University Hospital Klinikum rechts der Isar, Technical University of Munich, Germany
| | - Pelisek Jaroslav
- Department for Vascular and Endovascular Surgery, Munich Vascular Biobank, Munich Aortic Center (MAC), University Hospital Klinikum rechts der Isar, Technical University of Munich, Germany.,Department for Vascular Surgery, University Hospital Zurich, Switzerland
| | - Albert Busch
- Department for Vascular and Endovascular Surgery, Munich Vascular Biobank, Munich Aortic Center (MAC), University Hospital Klinikum rechts der Isar, Technical University of Munich, Germany.,Division of Vascular and Endovascular Surgery, Department for Visceral-, Thoracic and Vascular Surgery, Medical Faculty Carl Gustav Carus and University Hospital, Technische Universität Dresden, Germany
| |
Collapse
|
12
|
Targeted Inhibition of Matrix Metalloproteinase-8 Prevents Aortic Dissection in a Murine Model. Cells 2022; 11:cells11203218. [PMID: 36291087 PMCID: PMC9600539 DOI: 10.3390/cells11203218] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Revised: 09/30/2022] [Accepted: 10/11/2022] [Indexed: 11/16/2022] Open
Abstract
Aortic dissection (AD) is a lethal aortic pathology without effective medical treatments since the underlying pathological mechanisms responsible for AD remain elusive. Matrix metalloproteinase-8 (MMP8) has been previously identified as a key player in atherosclerosis and arterial remodeling. However, the functional role of MMP8 in AD remains largely unknown. Here, we report that an increased level of MMP8 was observed in 3-aminopropionitrile fumarate (BAPN)-induced murine AD. AD incidence and aortic elastin fragmentation were markedly reduced in MMP8-knockout mice. Importantly, pharmacologic inhibition of MMP8 significantly reduced the AD incidence and aortic elastin fragmentation. We observed less inflammatory cell accumulation, a lower level of aortic inflammation, and decreased smooth muscle cell (SMC) apoptosis in MMP8-knockout mice. In line with our previous observation that MMP8 cleaves Ang I to generate Ang II, BAPN-treated MMP8-knockout mice had increased levels of Ang I, but decreased levels of Ang II and lower blood pressure. Additionally, we observed a decreased expression level of vascular cell adhesion molecule-1 (VCAM1) and a reduced level of reactive oxygen species (ROS) in MMP8-knockout aortas. Mechanistically, our data show that the Ang II/VCAM1 signal axis is responsible for MMP8-mediated inflammatory cell invasion and transendothelial migration, while MMP8-mediated SMC inflammation and apoptosis are attributed to Ang II/ROS signaling. Finally, we observed higher levels of aortic and serum MMP8 in patients with AD. We therefore provide new insights into the molecular mechanisms underlying AD and identify MMP8 as a potential therapeutic target for this life-threatening aortic disease.
Collapse
|
13
|
Fatehi Hassanabad A, Schoettler FI, Kent WD, Adams CA, Holloway DD, Ali IS, Novick RJ, Ahsan MR, McClure RS, Shanmugam G, Kidd WT, Kieser TM, Fedak PW, Deniset JF. Comprehensive characterization of the postoperative pericardial inflammatory response: Potential implications for clinical outcomes. JTCVS OPEN 2022; 12:118-136. [PMID: 36590740 PMCID: PMC9801292 DOI: 10.1016/j.xjon.2022.09.003] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Revised: 08/23/2022] [Accepted: 09/06/2022] [Indexed: 01/04/2023]
Abstract
Objective There is a paucity of data on the inflammatory response that takes place in the pericardial space after cardiac surgery. This study provides a comprehensive assessment of the local postoperative inflammatory response. Methods Forty-three patients underwent cardiotomy, where native pericardial fluid was aspirated and compared with postoperative pericardial effluent collected at 4, 24, and 48 hours' postcardiopulmonary bypass. Flow cytometry was used to define the levels and proportions of specific immune cells. Samples were also probed for concentrations of inflammatory cytokines, matrix metalloproteinases (MMPs), and tissue inhibitors of metalloproteinases (TIMPs). Results Preoperatively, the pericardial space mainly contains macrophages and T cells. However, the postsurgical pericardial space was populated predominately by neutrophils, which constituted almost 80% of immune cells present, and peaked at 24 hours. When surgical approaches were compared, minimally invasive surgery was associated with fewer neutrophils in the pericardial space at 4 hours' postsurgery. Analysis of the intrapericardial concentrations of inflammatory mediators showed interleukin-6, MMP-9, and TIMP-1 to be highest postsurgery. Over time, MMP-9 concentrations decreased significantly, whereas TIMP-1 levels increased, resulting in a significant reduction of the ratio of MMP:TIMP after surgery, suggesting that active inflammatory processes may influence extracellular matrix remodeling. Conclusions These results show that cardiac surgery elicits profound alterations in the immune cell profile in the pericardial space. Defining the cellular and molecular mediators that drive pericardial-specific postoperative inflammatory processes may allow for targeted therapies to reduce immune-mediated complications.
Collapse
Key Words
- AVR, aortic valve replacement
- CABG, coronary artery bypass graft
- CD, cluster of differentiation
- CPB, cardiopulmonary bypass
- DC, dendritic cell
- ECM, extracellular matrix
- FS, full median sternotomy
- IL, interleukin
- IL-1Ra, interleukin-1 receptor antagonist
- Inf DC, inflammatory dendritic cell
- MICS, minimally invasive cardiac surgery
- MMP, matrix metalloproteinase
- MMPtot, total matrix metalloproteinases
- Mφ, macrophage
- NK, natural killer cell
- PAOF, postoperative atrial fibrillation
- PPS, postpericardiotomy syndrome
- RAMT-AVR, right anterior minithoracotomy aortic valve replacement
- SSC, side scatter
- TGFβ, transforming growth factor-beta
- TIMP, tissue inhibitor of metalloproteinases
- TIMPtot, total tissue inhibitors of metalloproteinases
- cDC, classical dendritic cell
- conventional cardiac surgery
- inflammation
- minimally invasive cardiac surgery
- pericardial space
- postoperative pericardial fluid
- sAVR, conventional full median sternotomy surgical aortic valve replacement
Collapse
Affiliation(s)
- Ali Fatehi Hassanabad
- Section of Cardiac Surgery, Department of Cardiac Sciences, Libin Cardiovascular Institute, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Friederike I. Schoettler
- Section of Cardiac Surgery, Department of Cardiac Sciences, Libin Cardiovascular Institute, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
- Department of Cardiac Surgery, Medical Faculty, University Hospital Düsseldorf, Heinrich-Heine-University, Düsseldorf, Germany
| | - William D.T. Kent
- Section of Cardiac Surgery, Department of Cardiac Sciences, Libin Cardiovascular Institute, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Corey A. Adams
- Section of Cardiac Surgery, Department of Cardiac Sciences, Libin Cardiovascular Institute, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Daniel D. Holloway
- Section of Cardiac Surgery, Department of Cardiac Sciences, Libin Cardiovascular Institute, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Imtiaz S. Ali
- Section of Cardiac Surgery, Department of Cardiac Sciences, Libin Cardiovascular Institute, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Richard J. Novick
- Section of Cardiac Surgery, Department of Cardiac Sciences, Libin Cardiovascular Institute, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
- Department of Critical Care Medicine, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Muhammad R. Ahsan
- Section of Cardiac Surgery, Department of Cardiac Sciences, Libin Cardiovascular Institute, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Robert Scott McClure
- Section of Cardiac Surgery, Department of Cardiac Sciences, Libin Cardiovascular Institute, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Ganesh Shanmugam
- Section of Cardiac Surgery, Department of Cardiac Sciences, Libin Cardiovascular Institute, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - William T. Kidd
- Section of Cardiac Surgery, Department of Cardiac Sciences, Libin Cardiovascular Institute, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Teresa M. Kieser
- Section of Cardiac Surgery, Department of Cardiac Sciences, Libin Cardiovascular Institute, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Paul W.M. Fedak
- Section of Cardiac Surgery, Department of Cardiac Sciences, Libin Cardiovascular Institute, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Justin F. Deniset
- Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
- Department of Cardiac Sciences, Libin Cardiovascular Institute, Calgary, Alberta, Canada
- Address for reprints: Justin F. Deniset, PhD, Department of Physiology & Pharmacology, Department of Cardiac Sciences, Libin Cardiovascular Institute Cumming School of Medicine, Health Research Innovation Centre, University of Calgary, 3330 Hospital Dr NW, Room GAC56, Calgary, Alberta, Canada, T2N 4N1.
| |
Collapse
|
14
|
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
|
15
|
Ledford BT, Akerman AW, Sun K, Gillis DC, Weiss JM, Vang J, Willcox S, Clemons TD, Sai H, Qiu R, Karver MR, Griffith JD, Tsihlis ND, Stupp SI, Ikonomidis JS, Kibbe MR. Peptide Amphiphile Supramolecular Nanofibers Designed to Target Abdominal Aortic Aneurysms. ACS NANO 2022; 16:7309-7322. [PMID: 35504018 PMCID: PMC9733406 DOI: 10.1021/acsnano.1c06258] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/26/2023]
Abstract
An abdominal aortic aneurysm (AAA) is a localized dilation of the aorta located in the abdomen that poses a severe risk of death when ruptured. The cause of AAA is not fully understood, but degradation of medial elastin due to elastolytic matrix metalloproteinases is a key step leading to aortic dilation. Current therapeutic interventions are limited to surgical repair to prevent catastrophic rupture. Here, we report the development of injectable supramolecular nanofibers using peptide amphiphile molecules designed to localize to AAA by targeting fragmented elastin, matrix metalloproteinase 2 (MMP-2), and membrane type 1 matrix metalloproteinase. We designed four targeting peptide sequences from X-ray crystallographic data and incorporated them into PA molecules via solid phase peptide synthesis. After coassembling targeted and diluent PAs at different molar ratios, we assessed their ability to form nanofibers using transmission electron microscopy and to localize to AAA in male and female Sprague-Dawley rats using light sheet fluorescence microscopy. We found that three formulations of the PA nanofibers were able to localize to AAA tissue, but the MMP-2 targeting PA substantially outperformed the other nanofibers. Additionally, we demonstrated that the MMP-2 targeting PA nanofibers had an optimal dose of 5 mg (∼12 mg/kg). Our results show that there was not a significant difference in targeting between male and female Sprague-Dawley rats. Given the ability of the MMP-2 targeting PA nanofiber to localize to AAA tissue, future studies will investigate potential diagnostic and targeted drug delivery applications for AAA.
Collapse
Affiliation(s)
- Benjamin T. Ledford
- Department of Surgery, University of Virginia, Charlottesville, VA 22903, USA
- Department of Surgery, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Adam W. Akerman
- Department of Surgery, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Kui Sun
- Department of Surgery, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - David C. Gillis
- Department of Surgery, University of Virginia, Charlottesville, VA 22903, USA
- Department of Surgery, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Jenna M. Weiss
- Department of Surgery, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Johnny Vang
- Department of Surgery, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Smaranda Willcox
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Tristan D. Clemons
- Simpson Querrey Institute for BioNanotechnology, Northwestern University, Chicago, IL 60611, USA
- Department of Chemistry, Northwestern University, Evanston, IL 60208, USA
| | - Hiroaki Sai
- Simpson Querrey Institute for BioNanotechnology, Northwestern University, Chicago, IL 60611, USA
- Department of Materials Science and Engineering, Northwestern University, Evanston, IL 60208, USA
| | - Ruomeng Qiu
- Department of Chemistry, Northwestern University, Evanston, IL 60208, USA
| | - Mark R. Karver
- Simpson Querrey Institute for BioNanotechnology, Northwestern University, Chicago, IL 60611, USA
| | - Jack D. Griffith
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Nick D. Tsihlis
- Department of Surgery, University of Virginia, Charlottesville, VA 22903, USA
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA 22904, USA
- Department of Surgery, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Samuel I. Stupp
- Simpson Querrey Institute for BioNanotechnology, Northwestern University, Chicago, IL 60611, USA
- Department of Chemistry, Northwestern University, Evanston, IL 60208, USA
- Department of Materials Science and Engineering, Northwestern University, Evanston, IL 60208, USA
- Department of Medicine, Northwestern University, Chicago, IL 60611, USA
- Department of Biomedical Engineering, Northwestern University, Evanston, IL 60208, USA
| | - John S. Ikonomidis
- Department of Surgery, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Melina R. Kibbe
- Department of Surgery, University of Virginia, Charlottesville, VA 22903, USA
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA 22904, USA
- Department of Surgery, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Department of Biomedical Engineering, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| |
Collapse
|
16
|
Huang Z, Su H, Zhang T, Li Y. Double-edged sword of diabetes mellitus for abdominal aortic aneurysm. Front Endocrinol (Lausanne) 2022; 13:1095608. [PMID: 36589814 PMCID: PMC9800781 DOI: 10.3389/fendo.2022.1095608] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Accepted: 12/05/2022] [Indexed: 12/23/2022] Open
Abstract
INTRODUCTION Diabetes mellitus (DM) has been proved to contribute to multiple comorbidities that are risk factors for abdominal aortic aneurysm (AAA). Remarkably, evidences from epidemiologic studies have demonstrated a negative association between the two disease states. On the other hand, hyperglycemic state was linked to post-operative morbidities following AAA repair. This review aims to provide a thorough picture on the double-edged nature of DM and major hypoglycemic medications on prevalence, growth rate and rupture of AAA, as well as DM-associated prognosis post AAA repair. METHODS We performed a comprehensive search in electronic databases to look for literatures demonstrating the association between DM and AAA. The primary focus of the literature search was on the impact of DM on the morbidity, enlargement and rupture rate, as well as post-operative complications of AAA. The role of antidiabetic medications was also explored. RESULTS Retrospective epidemiological studies and large database researches associated the presence of DM with decreased prevalence, slower expansion and limited rupture rate of AAA. Major hypoglycemic drugs exert similar protective effect as DM against AAA by targeting pathological hallmarks involved in AAA formation and progression, which were demonstrated predominantly by animal studies. Nevertheless, presence of DM or postoperative hyperglycemia was linked to poorer short-term and long-term prognosis, primarily due to greater risk of infection, longer duration of hospital stays and death. CONCLUSION While DM is a positive factor in the formation and progression of AAA, it is also associated with higher risk of negative outcomes following AAA repair. Concomitant use of antidiabetic medications may contribute to the protective mechanism of DM in AAA, but further studies are still warranted to explore their role following AAA repair.
Collapse
Affiliation(s)
- Zijia Huang
- Department of Pharmacy, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Huiling Su
- Department of Pharmacy, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Tiejun Zhang
- Department of Neurosurgery, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- *Correspondence: Tiejun Zhang, ; Yuwen Li,
| | - Yuwen Li
- Department of Pharmacy, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- *Correspondence: Tiejun Zhang, ; Yuwen Li,
| |
Collapse
|
17
|
Matsushita H, Hayashi H, Nurminsky K, Dunn T, He Y, Pitaktong I, Koda Y, Xu S, Nguyen V, Inoue T, Rodgers D, Nelson K, Johnson J, Hibino N. Novel reinforcement of corrugated nanofiber tissue-engineered vascular graft to prevent aneurysm formation for arteriovenous shunts in an ovine model. JVS Vasc Sci 2022; 3:182-191. [PMID: 35495567 PMCID: PMC9044007 DOI: 10.1016/j.jvssci.2022.01.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2021] [Accepted: 01/25/2022] [Indexed: 11/24/2022] Open
Abstract
Objective Many patients who require hemodialysis treatment will often require a prosthetic graft after multiple surgeries. However, the patency rate of grafts currently available commercially has not been satisfactory. Tissue engineering vascular grafts (TEVGs) are biodegradable scaffolds created to promote autologous cell proliferation and functional neotissue regeneration and, accordingly, have antithrombogenicity. Therefore, TEVGs can be an alternative prosthesis for small diameter grafts. However, owing to the limitations of the graft materials, most TEVGs are rigid and can easily kink when implanted in limited spaces, precluding future clinical application. Previously, we developed a novel corrugated nanofiber graft to prevent graft kinking. Reinforcement of these grafts to ensure their safety is required in a preclinical study. In the present study, three types of reinforcement were applied, and their effectiveness was examined using large animals. Methods In the present study, three different reinforcements for the graft composed of corrugated poly-ε-caprolactone (PCL) blended with poly(L-lactide-co-ε-caprolactone) (PLCL) created with electrospinning were evaluated: 1) a polydioxanone suture, 2) a 2-0 polypropylene suture, 3) a polyethylene terephthalate/polyurethane (PET/PU) outer layer, and PCL/PLCL as the control. These different grafts were then implanted in a U-shape between the carotid artery and jugular vein in seven ovine models for a total of 14 grafts during a 3-month period. In evaluating the different reinforcements, the main factors considered were cell proliferation and a lack of graft dilation, which were evaluated using ultrasound examinations and histologic and mechanical analysis. Results No kinking of the grafts occurred. Overall, re-endothelialization was observed in all the grafts at 3 months after surgery without graft rupture or calcification. The PCL/PLCL grafts and PCL/PLCL grafts with a polydioxanone suture showed high cell infiltration; however, they had become dilated 10 weeks after surgery. In contrast, the PCL/PLCL graft with the 2-0 suture and the PCL/PLCL graft covered with a PET/PU layer did not show any graft expansion. The PCL/PLCL graft covered with a PET/PU layer showed less cell infiltration than that of the PCL/PLCL graft. Conclusions Reinforcement is required to create grafts that can withstand arterial pressure. Reinforcement with suture materials has the potential to maintain cell infiltration into the graft, which could improve the neotissue formation of the graft. In our basic science research study, we investigated tissue engineered vascular grafts for arteriovenous shunts. Our grafts were created with poly-ε-caprolactone and poly(L-lactide-co-ε-caprolactone) and designed with corrugated walls to avoid graft kinking. The grafts were implanted between the carotid artery and external jugular vein in a U-shape using an ovine model. To withstand the high pressure of blood on the arterial system, two types of reinforcement were applied to these tissue engineering vascular grafts. Because reinforcement of the graft could interfere with cell infiltration into the tissue engineering vascular grafts, the methods and material of reinforcement were investigated, in addition to the mechanical properties of the graft.
Collapse
|
18
|
Ducas AA, Kuhn DCS, Bath LC, Lozowy RJ, Boyd AJ. Increased matrix metalloproteinase 9 activity correlates with flow-mediated intraluminal thrombus deposition and wall degeneration in human abdominal aortic aneurysm. JVS Vasc Sci 2021; 1:190-199. [PMID: 34617048 PMCID: PMC8489223 DOI: 10.1016/j.jvssci.2020.09.004] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Accepted: 09/17/2020] [Indexed: 11/15/2022] Open
Abstract
Objective We have previously demonstrated that human abdominal aortic aneurysm (AAA) rupture occurs in zones of low wall shear stress where flow recirculation and intraluminal thrombus (ILT) deposition are increased. Matrix metalloproteinase-9 (MMP-9) is involved in the pathogenesis of AAA via its lytic effect on collagen and elastin. We hypothesize that flow-mediated ILT deposition promotes increased local inflammatory and MMP-9 activity that leads to AAA wall degeneration. The purpose of this study was to examine the correlation between predicted pulsatile flow dynamics and regional differences in MMP-9, elastin, collagen, and ILT deposition in human AAA. Methods Full-thickness aortic tissue samples were collected from 24 patients undergoing open AAA repair. Control infrarenal aortic tissue was obtained from 6 patients undergoing aortobifemoral bypass. Full-thickness aortic tissue and ILT were assessed for MMP-9 levels using a cytokine array assay. Histologic and immunohistochemical assessment of inflammation, collagen and elastin content, and MMP-9 levels were also measured. Three-dimensional AAA geometry was generated from computed tomography angiogram (CTA) images using Mimics software and computational fluid dynamics was used to predict pulsatile aortic blood flow. Results The majority of AAA showed eccentric ILT deposition which was correlated with predicted recirculation blood flow (R2 = –0.17; P < .05). The regions of high ILT were associated with significant increases in inflammation and loss of elastin and collagen compared with regions of low ILT, or with control tissue. MMP-9 was significantly higher in areas of high ILT deposition compared with areas devoid of ILT. Tissue MMP-9 was correlated with the thickness of ILT deposition (R2 = 0.46; P < .05), and was also present in high levels in thick compared with thin ILT. Conclusions We have shown a correlation between flow-mediated ILT deposition with increased tissue levels of MMP-9 activity, increased inflammatory infiltrate, and decreased elastin and collagen content in stereotactically sampled human AAA, suggesting that ILT deposition is associated with local increases in proteolytic activity that may preferentially weaken and promote rupture at selected regions.
Collapse
Affiliation(s)
- Annie A Ducas
- Department of Surgery, University of Manitoba, Manitoba, Canada
| | - David C S Kuhn
- Department of Mechanical Engineering, University of Manitoba, Manitoba, Canada
| | - Lauren C Bath
- Faculty of Medicine, University of Manitoba, Manitoba, Canada
| | - Richard J Lozowy
- Department of Mechanical Engineering, University of Manitoba, Manitoba, Canada
| | - April J Boyd
- Department of Surgery, University of Manitoba, Manitoba, Canada
| |
Collapse
|
19
|
Neutrophils as Regulators and Biomarkers of Cardiovascular Inflammation in the Context of Abdominal Aortic Aneurysms. Biomedicines 2021; 9:biomedicines9091236. [PMID: 34572424 PMCID: PMC8467789 DOI: 10.3390/biomedicines9091236] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2021] [Revised: 09/10/2021] [Accepted: 09/14/2021] [Indexed: 12/22/2022] Open
Abstract
Neutrophils represent up to 70% of circulating leukocytes in healthy humans and combat infection mostly by phagocytosis, degranulation and NETosis. It has been reported that neutrophils are centrally involved in abdominal aortic aneurysm (AAA) pathogenesis. The natural course of AAA is growth and rupture, if left undiagnosed or untreated. The rupture of AAA has a very high mortality and is currently among the leading causes of death worldwide. The use of noninvasive cardiovascular imaging techniques for patient screening, surveillance and postoperative follow-up is well established and recommended by the current guidelines. Neutrophil-derived biomarkers may offer clinical value to the monitoring and prognosis of AAA patients, allowing for potential early therapeutic intervention. Numerous promising biomarkers have been studied. In this review, we discuss neutrophils and neutrophil-derived molecules as regulators and biomarkers of AAA, and our aim was to specifically highlight diagnostic and prognostic markers. Neutrophil-derived biomarkers may potentially, in the future, assist in determining AAA presence, predict size, expansion rate, rupture risk, and postoperative outcome once validated in highly warranted future prospective clinical studies.
Collapse
|
20
|
Lust ST, Shanahan CM, Shipley RJ, Lamata P, Gentleman E. Design considerations for engineering 3D models to study vascular pathologies in vitro. Acta Biomater 2021; 132:114-128. [PMID: 33652164 PMCID: PMC7611653 DOI: 10.1016/j.actbio.2021.02.031] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Revised: 01/28/2021] [Accepted: 02/18/2021] [Indexed: 12/15/2022]
Abstract
Many cardiovascular diseases (CVD) are driven by pathological remodelling of blood vessels, which can lead to aneurysms, myocardial infarction, ischaemia and strokes. Aberrant remodelling is driven by changes in vascular cell behaviours combined with degradation, modification, or abnormal deposition of extracellular matrix (ECM) proteins. The underlying mechanisms that drive the pathological remodelling of blood vessels are multifaceted and disease specific; however, unravelling them may be key to developing therapies. Reductionist models of blood vessels created in vitro that combine cells with biomaterial scaffolds may serve as useful analogues to study vascular disease progression in a controlled environment. This review presents the main considerations for developing such in vitro models. We discuss how the design of blood vessel models impacts experimental readouts, with a particular focus on the maintenance of normal cellular phenotypes, strategies that mimic normal cell-ECM interactions, and approaches that foster intercellular communication between vascular cell types. We also highlight how choice of biomaterials, cellular arrangements and the inclusion of mechanical stimulation using fluidic devices together impact the ability of blood vessel models to mimic in vivo conditions. In the future, by combining advances in materials science, cell biology, fluidics and modelling, it may be possible to create blood vessel models that are patient-specific and can be used to develop and test therapies. STATEMENT OF SIGNIFICANCE: Simplified models of blood vessels created in vitro are powerful tools for studying cardiovascular diseases and understanding the mechanisms driving their progression. Here, we highlight the key structural and cellular components of effective models and discuss how including mechanical stimuli allows researchers to mimic native vessel behaviour in health and disease. We discuss the primary methods used to form blood vessel models and their limitations and conclude with an outlook on how blood vessel models that incorporate patient-specific cells and flows can be used in the future for personalised disease modelling.
Collapse
Affiliation(s)
- Suzette T Lust
- Centre for Craniofacial and Regenerative Biology, King's College London, London SE1 9RT, United Kingdom; School of Biomedical Engineering and Imaging Sciences, King's College London, London SE1 7EH, United Kingdom
| | - Catherine M Shanahan
- School of Cardiovascular Medicine and Sciences, King's College London, London SE5 9NU, United Kingdom
| | - Rebecca J Shipley
- Institute of Healthcare Engineering and Department of Mechanical Engineering, University College London, London WC1E 7JE, United Kingdom
| | - Pablo Lamata
- School of Biomedical Engineering and Imaging Sciences, King's College London, London SE1 7EH, United Kingdom
| | - Eileen Gentleman
- Centre for Craniofacial and Regenerative Biology, King's College London, London SE1 9RT, United Kingdom.
| |
Collapse
|
21
|
Wang W, Zhao T, Geng K, Yuan G, Chen Y, Xu Y. Smoking and the Pathophysiology of Peripheral Artery Disease. Front Cardiovasc Med 2021; 8:704106. [PMID: 34513948 PMCID: PMC8429807 DOI: 10.3389/fcvm.2021.704106] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2021] [Accepted: 07/31/2021] [Indexed: 12/15/2022] Open
Abstract
Smoking is one of the most important preventable factors causing peripheral artery disease (PAD). The purpose of this review is to comprehensively analyze and summarize the pathogenesis and clinical characteristics of smoking in PAD based on existing clinical, in vivo, and in vitro studies. Extensive searches and literature reviews have shown that a large amount of data exists on the pathological process underlying the effects of cigarette smoke and its components on PAD through various mechanisms. Cigarette smoke extracts (CSE) induce endothelial cell dysfunction, smooth muscle cell remodeling and macrophage phenotypic transformation through multiple molecular mechanisms. These pathological changes are the molecular basis for the occurrence and development of peripheral vascular diseases. With few discussions on the topic, we will summarize recent insights into the effect of smoking on regulating PAD through multiple pathways and its possible pathogenic mechanism.
Collapse
Affiliation(s)
- Weiming Wang
- The State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau, China.,Department of General Surgery (Vascular Surgery), The Affiliated Hospital of Southwest Medical University, Luzhou, China.,Key Laboratory of Medical Electrophysiology, Ministry of Education and Medical Electrophysiological Key Laboratory of Sichuan Province, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, China
| | - Tingting Zhao
- The State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau, China
| | - Kang Geng
- The State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau, China
| | - Gang Yuan
- The State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau, China
| | - Yue Chen
- Department of Nuclear Medicine, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Youhua Xu
- The State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau, China
| |
Collapse
|
22
|
Vu HD, Huynh PT, Ryu J, Kang UR, Youn SW, Kim H, Ahn HJ, Park K, Hwang SK, Chang YC, Lee YJ, Lee HJ, Lee J. Melittin-loaded Iron Oxide Nanoparticles Prevent Intracranial Arterial Dolichoectasia Development through Inhibition of Macrophage-mediated Inflammation. Int J Biol Sci 2021; 17:3818-3836. [PMID: 34671201 PMCID: PMC8495379 DOI: 10.7150/ijbs.60588] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Accepted: 08/22/2021] [Indexed: 12/24/2022] Open
Abstract
Rationale: In intracranial arterial dolichoectasia (IADE) development, the feedback loop between inflammatory cytokines and macrophages involves TNF-α and NF-κB signaling pathways and leads to subsequent MMP-9 activation and extracellular matrix (ECM) degeneration. In this proof-of-concept study, melittin-loaded L-arginine-coated iron oxide nanoparticle (MeLioN) was proposed as the protective measure of IADE formation for this macrophage-mediated inflammation and ECM degeneration. Methods: IADE was created in 8-week-old C57BL/6J male mice by inducing hypertension and elastase injection into a basal cistern. Melittin was loaded on the surface of ION as a core-shell structure (hydrodynamic size, 202.4 nm; polydispersity index, 0.158). Treatment of MeLioN (2.5 mg/kg, five doses) started after the IADE induction, and the brain was harvested in the third week. In the healthy control, disease control, and MeLioN-treated group, the morphologic changes of the cerebral arterial wall were measured by diameter, thickness, and ECM composition. The expression level of MMP-9, CD68, MCP-1, TNF-α, and NF-κB was assessed from immunohistochemistry, polymerase chain reaction, and Western blot assay. Results: MeLioN prevented morphologic changes of cerebral arterial wall related to IADE formation by restoring ECM alterations and suppressing MMP-9 expression. MeLioN inhibited MCP-1 expression and reduced CD68-positive macrophage recruitments into cerebral arterial walls. MeLioN blocked TNF-α activation and NF-κB signaling pathway. In the Sylvian cistern, co-localization was found between the CD68-positive macrophage infiltrations and the MeLioN distributions detected on Prussian Blue and T2* gradient-echo MRI, suggesting the role of macrophage harboring MeLioN. Conclusions: The macrophage infiltration into the arterial wall plays a critical role in the MMP-9 secretion. MeLioN, designed for ION-mediated melittin delivery, effectively prevents IADE formation by suppressing macrophage-mediated inflammations and MMP activity. MeLioN can be a promising strategy preventing IADE development in high-risk populations.
Collapse
Affiliation(s)
- Huy Duc Vu
- Department of Radiology, Daegu Catholic University School of Medicine, Daegu, Korea
| | - Phuong Tu Huynh
- Department of Radiology, Daegu Catholic University School of Medicine, Daegu, Korea
| | - Junghwa Ryu
- Department of Radiology, Daegu Catholic University School of Medicine, Daegu, Korea
| | - Ung Rae Kang
- Department of Radiology, Daegu Catholic University School of Medicine, Daegu, Korea
| | - Sung Won Youn
- Department of Radiology, Daegu Catholic University School of Medicine, Daegu, Korea
| | - Hongtae Kim
- Department of Anatomy, Daegu Catholic University School of Medicine, Daegu, Korea
| | - Hyun Jin Ahn
- Department of Pathology, Daegu Catholic University School of Medicine, Daegu, Korea
| | - Kwankyu Park
- Department of Pathology, Daegu Catholic University School of Medicine, Daegu, Korea
| | - Soon-Kyung Hwang
- Department of Molecular Biology, Daegu Catholic University School of Medicine, Daegu, Korea
| | - Young-Chae Chang
- Department of Molecular Biology, Daegu Catholic University School of Medicine, Daegu, Korea
| | - Yong Jig Lee
- Department of Plastic Surgery, Daegu Catholic University School of Medicine, Daegu, Korea
| | - Hui Joong Lee
- Department of Radiology, Kyungpook National University School of Medicine, Daegu, Korea
| | - Jongmin Lee
- Department of Radiology, Kyungpook National University School of Medicine, Daegu, Korea
| |
Collapse
|
23
|
De Leo N, Melillo A, Zhang P, Badach J, Miller H, Lin A, Williamson J, Ghobrial G, Gaughan J, Krishnadoss V, Noshadi I, Brown SA, Carpenter JP. Development of a Model for Abdominal Aortic Aneurysms in Swine. J Surg Res 2021; 268:79-86. [PMID: 34289418 DOI: 10.1016/j.jss.2021.05.047] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Revised: 03/12/2021] [Accepted: 05/26/2021] [Indexed: 01/02/2023]
Abstract
INTRODUCTION Producing a reliable large-animal model of AAA has proven challenging. We sought to create a reproducible swine model of AAA using enzymatic degradation of the aortic wall. METHODS Twelve male Yorkshire swine received periadventitial injections of type 1 collagenase and porcine pancreatic elastase into a 4 cm segment of infrarenal aorta. Nine survived until postoperative day (POD) 21. Aortic growth was monitored at 7 and 14 days using ultrasound. The animals were euthanized on POD 21, and the suprarenal (control) and infrarenal aorta were harvested for analysis, after gross measurement of aortic diameter (AD). Tensile strength was measured and additional segments were collected for histopathological analysis. PCR of matrix metalloproteinases (MMP9) was conducted. Groups were compared with paired t-tests, or ANOVA, where appropriate. RESULTS Average percent growth of AD at POD 21 for treated segments was 27% versus 4.5% for control tissue. The average difference in AD by subject, was 26.7% (P<0.001). Aortic medial thickness was decreased in treated tissue; 235 μm versus 645 μm (P<0.0001). Quantities of both medial elastin fibers, and smooth muscles cells were decreased in treated tissue; 1.8% compared to 9.9% (P<0.0001), and 24% versus 37.4%, respectively. Tensile strength was also decreased in treated tissue; 16.7 MPa versus 29.5 MPa (P=0.0002). A 12-fold increase in expression of MMP9 mRNA was also demonstrated in aneurysmal tissue (P=0.002) CONCLUSION: A reproducible, large-animal model of AAA, with anatomical, histopathological, and biomechanical properties that are clinically translatable, can be achieved with extraluminal enzymatic degradation.
Collapse
Affiliation(s)
- Nicholas De Leo
- Cooper University Hospital/Cooper Medical School of Rowan University, Department of Surgery, Camden, New Jersey.
| | - Atlee Melillo
- Cooper University Hospital/Cooper Medical School of Rowan University, Department of Surgery, Camden, New Jersey
| | - Ping Zhang
- Cooper University Hospital/Cooper Medical School of Rowan University, Department of Surgery, Camden, New Jersey
| | - Jeremy Badach
- Cooper University Hospital/Cooper Medical School of Rowan University, Department of Surgery, Camden, New Jersey
| | - Henry Miller
- Cooper University Hospital/Cooper Medical School of Rowan University, Department of Surgery, Camden, New Jersey
| | - Andrew Lin
- Cooper University Hospital/Cooper Medical School of Rowan University, Department of Surgery, Camden, New Jersey
| | - John Williamson
- Cooper University Hospital/Cooper Medical School of Rowan University, Department of Surgery, Camden, New Jersey
| | - Gaby Ghobrial
- Cooper University Hospital/Cooper Medical School of Rowan University, Department of Surgery, Camden, New Jersey
| | - J Gaughan
- Cooper University Hospital/Cooper Medical School of Rowan University, Department of Surgery, Camden, New Jersey
| | | | - Iman Noshadi
- Rowan University, College of Engineering, Glassboro, New Jersey
| | - Spencer A Brown
- Cooper Research Institute, Education and Research Building, Camden, New Jersey
| | - Jeffrey P Carpenter
- Cooper University Hospital/Cooper Medical School of Rowan University, Department of Surgery, Camden, New Jersey
| |
Collapse
|
24
|
Expression gradient of metalloproteinases and their inhibitors from proximal to distal segments of abdominal aortic aneurysm. J Appl Genet 2021; 62:499-506. [PMID: 34091862 PMCID: PMC8357691 DOI: 10.1007/s13353-021-00642-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Revised: 03/15/2021] [Accepted: 05/26/2021] [Indexed: 11/14/2022]
Abstract
Abdominal aortic aneurysm refers to abnormal, asymmetric distension of the infrarenal aortic wall due to pathological remodelling of the extracellular matrix. The distribution of enzymes remodelling the extracellular matrix and their expression patterns in the affected tissue are largely unknown. The goal of this work was to investigate the expression profiles of 20 selected genes coding for metalloproteinases and their inhibitors in the proximal to the distal direction of the abdominal aortic aneurysm. RNA samples were purified from four lengthwise fragments of aneurysm and border tissue obtained from 29 patients. The quantities of selected mRNAs were determined by real-time PCR to reveal the expression patterns. The genes of interest encode collagenases (MMP1, MMP8, MMP13), gelatinases (MMP2, MMP9), stromelysins (MMP3, MMP7, MMP10, MMP11, MMP12), membrane-type MMPs (MMP14, MMP15, MMP16), tissue inhibitors of metalloproteinases (TIMP1, TIMP2, TIMP3, TIMP4), and ADAMTS proteinases (ADAMTS1, ADAMTS8, and ADAMTS13). It was found that MMP, TIMP, and ADAMTS are expressed in all parts of the aneurysm with different patterns. A developed aneurysm has such a disturbed expression of the main participants in extracellular matrix remodelling that it is difficult to infer the causes of the disorder development. MMP12 secreted by macrophages at the onset of inflammation may initiate extracellular matrix remodelling, which, if not controlled, initiates a feedback loop leading to aneurysm formation.
Collapse
|
25
|
Adventitial recruitment of Lyve-1- macrophages drives aortic aneurysm in an angiotensin-2-based murine model. Clin Sci (Lond) 2021; 135:1295-1309. [PMID: 33978148 DOI: 10.1042/cs20200963] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Revised: 04/30/2021] [Accepted: 05/12/2021] [Indexed: 12/31/2022]
Abstract
OBJECTIVE Aortic macrophage accumulation is characteristic of the pathogenesis of abdominal aortic aneurysm (AAA) but the mechanisms of macrophage accumulation and their phenotype are poorly understood. Lymphatic vessel endothelial receptor-1 (Lyve-1+) resident aortic macrophages independently self-renew and are functionally distinct from monocyte-derived macrophages recruited during inflammation. We hypothesized that Lyve-1+ and Lyve-1- macrophages differentially contribute to aortic aneurysm. Approach and results: Angiotensin-2 and β-aminopropionitrile (AT2/BAPN) were administered to induce AAA in C57BL/6J mice. Using immunohistochemistry (IHC), we demonstrated primarily adventitial accumulation of aortic macrophages, and in association with areas of elastin fragmentation and aortic dissection. Compared with controls, AAA was associated with a relative percent depletion of Lyve-1+ resident aortic macrophages and accumulation of Lyve-1- macrophages. Using CD45.1/CD45.2 parabiosis, we demonstrated aortic macrophage recruitment in AAA. Depletion of aortic macrophages in CCR2-/- mice was associated with reduced aortic dilatation indicating the functional role of recruitment from the bone marrow. Depletion of aortic macrophages using anti-macrophage colony-stimulating factor 1 receptor (MCSF1R)-neutralizing antibody (Ab) reduced the incidence of AAA. Conditional depletion of Lyve-1+ aortic macrophages was achieved by generating Lyve-1wt/cre Csf1rfl/fl mice. Selective depletion of Lyve-1+ aortic macrophages had no protective effects following AT2/BAPN administration and resulted in increased aortic dilatation in the suprarenal aorta. CONCLUSIONS Aortic macrophage accumulation in AAA derives from adventitial recruitment of Lyve-1- macrophages, with relative percent depletion of Lyve-1+ macrophages. Selective targeting of macrophage subtypes represents a potential novel therapeutic avenue for the medical treatment of AAA.
Collapse
|
26
|
Peña R, Valverde S, Alcázar JA, Cebrián P, González-Porras JR, Lozano FS. Abdominal aortic aneurysm and acute appendicitis: a case report and review of the literature. J Med Case Rep 2021; 15:203. [PMID: 33863365 PMCID: PMC8052834 DOI: 10.1186/s13256-021-02703-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2019] [Accepted: 01/27/2021] [Indexed: 11/16/2022] Open
Abstract
Background Abdominal aortic aneurysm and acute appendicitis occur relatively frequently in elderly patients. However, the co-occurrence of the two pathologies is very rare and serious. Case presentation We present the case of an elderly Caucasian patient who was aware of having an abdominal aortic aneurysm but refused treatment and was subsequently admitted to the hospital’s emergency department with acute abdominal symptoms. A computed tomography scan raised the possibility of complication due to the characteristics of the aneurysm. The patient then agreed to emergency surgery. Laparotomy revealed the existence of an acute perforated appendicitis with a significant abscess in the right iliac fossa and an uncomplicated aneurysm. Appendectomy was performed and the abscess drained. The postoperative period passed without complications, and the patient again refused surgery for the aneurysm, which due to its anatomical characteristics was not a candidate for standard endovascular treatment. Conclusions In light of this experience, we review the literature about the relationship between abdominal aortic aneurysm and acute appendicitis.
Collapse
Affiliation(s)
- Rubén Peña
- Servicio de Angiología y Cirugía Vascular, Complejo Asistencial Universitario de Salamanca (CAUSA), Instituto de Investigación Biomédica de Salamanca (IBSAL), Universidad de Salamanca (USAL), Paseo de San Vicente 139, 37007, Salamanca, Spain
| | - Sergio Valverde
- Servicio de Angiología y Cirugía Vascular, Complejo Asistencial Universitario de Salamanca (CAUSA), Instituto de Investigación Biomédica de Salamanca (IBSAL), Universidad de Salamanca (USAL), Paseo de San Vicente 139, 37007, Salamanca, Spain
| | - José A Alcázar
- Servicio de Cirugía General y del Aparato Digestivo, Complejo Asistencial Universitario de Salamanca (CAUSA), Instituto de Investigación Biomédica de Salamanca (IBSAL), Universidad de Salamanca (USAL), Salamanca, Spain
| | - Paloma Cebrián
- Servicio de Radiodiagnóstico, Complejo Asistencial Universitario de Salamanca (CAUSA), Instituto de Investigación Biomédica de Salamanca (IBSAL), Universidad de Salamanca (USAL), Salamanca, Spain
| | - José Ramón González-Porras
- Servicio de Hematología, Complejo Asistencial Universitario de Salamanca (CAUSA), Instituto de Investigación Biomédica de Salamanca (IBSAL), Universidad de Salamanca (USAL), Salamanca, Spain
| | - Francisco S Lozano
- Servicio de Angiología y Cirugía Vascular, Complejo Asistencial Universitario de Salamanca (CAUSA), Instituto de Investigación Biomédica de Salamanca (IBSAL), Universidad de Salamanca (USAL), Paseo de San Vicente 139, 37007, Salamanca, Spain.
| |
Collapse
|
27
|
Andreucci M, Provenzano M, Faga T, Michael A, Patella G, Mastroroberto P, Serraino GF, Bracale UM, Ielapi N, Serra R. Aortic Aneurysms, Chronic Kidney Disease and Metalloproteinases. Biomolecules 2021; 11:194. [PMID: 33573220 PMCID: PMC7912263 DOI: 10.3390/biom11020194] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Revised: 01/21/2021] [Accepted: 01/27/2021] [Indexed: 12/12/2022] Open
Abstract
Metalloproteinases (MPs) are proteolytic enzymes involved in extracellular matrix deposition, regulation of cellular signals of inflammation, proliferation, and apoptosis. Metalloproteinases are classified into three families: Matrix-MPs (MMPs), A-Disintegrin-and-Metalloprotease (ADAMs), and the A-Disintegrin-and-Metalloproteinase-with-Thrombospondin-1-like-Domains (ADAMTS). Previous studies showed that MPs are involved in the development of aortic aneurysms (AA) and, concomitantly, in the onset of chronic kidney disease (CKD). CKD has been, per se, associated with an increased risk for AA. The aim of this review is to examine the pathways that may associate MPs with CKD and AA. Several MMPs, such as MMP-2, -8, -9, and TIMP-1 have been shown to damage the AA wall and to have a toxic effect on renal tubular cells, leading to fibrosis. Similarly, ADAM10 and 17 have been shown to degrade collagen in the AA wall and to worsen kidney function via pro-inflammatory stimuli, the impairment of the Renin-Angiotensin-Aldosterone System, and the degradation of structural proteins. Moreover, MMP-2 and -9 inhibitors reduced aneurysm growth and albuminuria in experimental and human studies. It would be important, in the future, to expand research on MPs from both a prognostic, namely, to refine risk stratification in CKD patients, and a predictive perspective, likely to improve prognosis in response to targeted treatments.
Collapse
Affiliation(s)
- Michele Andreucci
- Department of Health Sciences, “Magna Graecia” University, I-88100 Catanzaro, Italy; (M.A.); (T.F.); (A.M.); (G.P.)
| | - Michele Provenzano
- Department of Medical and Surgical Sciences, University Magna Graecia of Catanzaro, Viale Europa, I-88100 Catanzaro, Italy;
| | - Teresa Faga
- Department of Health Sciences, “Magna Graecia” University, I-88100 Catanzaro, Italy; (M.A.); (T.F.); (A.M.); (G.P.)
| | - Ashour Michael
- Department of Health Sciences, “Magna Graecia” University, I-88100 Catanzaro, Italy; (M.A.); (T.F.); (A.M.); (G.P.)
| | - Gemma Patella
- Department of Health Sciences, “Magna Graecia” University, I-88100 Catanzaro, Italy; (M.A.); (T.F.); (A.M.); (G.P.)
| | - Pasquale Mastroroberto
- Department of Experimental and Clinical Medicine, University of Catanzaro, I-88100 Catanzaro, Italy; (P.M.); (G.F.S.)
| | - Giuseppe Filiberto Serraino
- Department of Experimental and Clinical Medicine, University of Catanzaro, I-88100 Catanzaro, Italy; (P.M.); (G.F.S.)
| | | | - Nicola Ielapi
- Department of Public Health and Infectious Disease, “Sapienza” University of Rome, I-00185 Roma, Italy;
| | - Raffaele Serra
- Department of Medical and Surgical Sciences, University Magna Graecia of Catanzaro, Viale Europa, I-88100 Catanzaro, Italy;
- Interuniversity Center of Phlebolymphology (CIFL), “Magna Graecia” University, I-88100 Catanzaro, Italy
| |
Collapse
|
28
|
Zhang X, Ares WJ, Taussky P, Ducruet AF, Grandhi R. Role of matrix metalloproteinases in the pathogenesis of intracranial aneurysms. Neurosurg Focus 2020; 47:E4. [PMID: 31261127 DOI: 10.3171/2019.4.focus19214] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2019] [Accepted: 04/09/2019] [Indexed: 12/29/2022]
Abstract
Intracranial aneurysms (IAs) are a result of complex interactions between biochemical and mechanical forces and can lead to significant morbidity if they rupture and cause subarachnoid hemorrhage. This review explores the role of matrix metalloproteinases (MMPs) in the pathogenesis and progression of IAs. In addition to providing a review of the normal function of MMPs, it is intended to explore the interaction between inflammation and abnormal blood flow and the resultant pathological vascular remodeling processes seen in the development and rupture of IAs. Also reviewed is the potential for the use of MMPs as a diagnostic tool for assessment of aneurysm development and progression.
Collapse
Affiliation(s)
- Xiaoran Zhang
- 1Department of Neurological Surgery, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania
| | - William J Ares
- 1Department of Neurological Surgery, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania
| | - Philipp Taussky
- 2Department of Neurosurgery, University of Utah School of Medicine, Salt Lake City, Utah; and
| | - Andrew F Ducruet
- 3Department of Neurological Surgery, Barrow Neurological Institute, Phoenix, Arizona
| | - Ramesh Grandhi
- 2Department of Neurosurgery, University of Utah School of Medicine, Salt Lake City, Utah; and
| |
Collapse
|
29
|
Song TJ, Oh SH, Kim J. The impact of statin therapy after surgical or endovascular treatment of cerebral aneurysms. J Neurosurg 2020; 133:182-189. [PMID: 31125972 DOI: 10.3171/2019.3.jns183500] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2018] [Accepted: 03/08/2019] [Indexed: 12/23/2022]
Abstract
OBJECTIVE Cerebral aneurysms represent the most common cause of spontaneous subarachnoid hemorrhage. Statins are lipid-lowering agents that may expert multiple pleiotropic vascular protective effects. The authors hypothesized that statin therapy after coil embolization or surgical clipping of cerebral aneurysms might improve clinical outcomes. METHODS This was a retrospective cohort study using the National Health Insurance Service-National Sample Cohort Database in Korea. Patients who underwent coil embolization or surgical clipping for cerebral aneurysm between 2002 and 2013 were included. Based on prescription claims, the authors calculated the proportion of days covered (PDC) by statins during follow-up as a marker of statin therapy. The primary outcome was a composite of the development of stroke, myocardial infarction, and all-cause death. Multivariate time-dependent Cox regression analyses were performed. RESULTS A total of 1381 patients who underwent coil embolization (n = 542) or surgical clipping (n = 839) of cerebral aneurysms were included in this study. During the mean (± SD) follow-up period of 3.83 ± 3.35 years, 335 (24.3%) patients experienced the primary outcome. Adjustments were performed for sex, age (as a continuous variable), treatment modality, aneurysm rupture status (ruptured or unruptured aneurysm), hypertension, diabetes mellitus, household income level, and prior history of ischemic stroke or intracerebral hemorrhage as time-independent variables and statin therapy during follow-up as a time-dependent variable. Consistent statin therapy (PDC > 80%) was significantly associated with a lower risk of the primary outcome (adjusted hazard ratio 0.34, 95% CI 0.14-0.85). CONCLUSIONS Consistent statin therapy was significantly associated with better prognosis after coil embolization or surgical clipping of cerebral aneurysms.
Collapse
Affiliation(s)
- Tae-Jin Song
- 1Department of Neurology, Mokdong Hospital, Ewha Womans University College of Medicine, Seoul
| | - Seung-Hun Oh
- 2Department of Neurology, CHA Bundang Medical Center, CHA University, Seongnam; and
| | - Jinkwon Kim
- 2Department of Neurology, CHA Bundang Medical Center, CHA University, Seongnam; and
- 3Department of Neurology, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
| |
Collapse
|
30
|
Hosseini V, Mallone A, Mirkhani N, Noir J, Salek M, Pasqualini FS, Schuerle S, Khademhosseini A, Hoerstrup SP, Vogel V. A Pulsatile Flow System to Engineer Aneurysm and Atherosclerosis Mimetic Extracellular Matrix. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2020; 7:2000173. [PMID: 32596117 PMCID: PMC7312268 DOI: 10.1002/advs.202000173] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Indexed: 06/11/2023]
Abstract
Alterations of blood flow patterns strongly correlate with arterial wall diseases such as atherosclerosis and aneurysm. Here, a simple, pumpless, close-loop, easy-to-replicate, and miniaturized flow device is introduced to concurrently expose 3D engineered vascular smooth muscle tissues to high-velocity pulsatile flow versus low-velocity disturbed flow conditions. Two flow regimes are distinguished, one that promotes elastin and impairs collagen I assembly, while the other impairs elastin and promotes collagen assembly. This latter extracellular matrix (ECM) composition shares characteristics with aneurysmal or atherosclerotic tissue phenotypes, thus recapitulating crucial hallmarks of flow-induced tissue morphogenesis in vessel walls. It is shown that the mRNA levels of ECM of collagens and elastin are not affected by the differential flow conditions. Instead, the differential gene expression of matrix metalloproteinase (MMP) and their inhibitors (TIMPs) is flow-dependent, and thus drives the alterations in ECM composition. In further support, treatment with doxycycline, an MMP inhibitor and a clinically used drug to treat vascular diseases, halts the effect of low-velocity flow on the ECM remodeling. This illustrates how the platform can be exploited for drug efficacy studies by providing crucial mechanistic insights into how different therapeutic interventions may affect tissue growth and ECM assembly.
Collapse
Affiliation(s)
- Vahid Hosseini
- Laboratory of Applied MechanobiologyInstitute of Translational MedicineDepartment of Health Sciences and TechnologyETH ZurichZurich8093Switzerland
- Present address:
Department of BioengineeringUniversity of California‐Los AngelesLos AngelesCA90095USA
| | - Anna Mallone
- Institute for Regenerative Medicine (IREM)University of Zurich and Wyss Translational Center ZurichZurich8952Switzerland
| | - Nima Mirkhani
- Responsive Biomedical Systems LabInstitute of Translational MedicineDepartment of Health Sciences and TechnologyETH ZurichZurich8093Switzerland
| | - Jerome Noir
- Institute of GeophysicsDepartment of Earth SciencesETH ZurichZurich8092Switzerland
| | - Mehdi Salek
- Department of Mechanical EngineeringMassachusetts Institute of TechnologyBostonMA02139USA
| | - Francesco Silvio Pasqualini
- Institute for Regenerative Medicine (IREM)University of Zurich and Wyss Translational Center ZurichZurich8952Switzerland
- Synthetic Physiology LaboratoryDepartment of Civil Engineering and ArchitectureUniversity of PaviaPavia27100Italy
| | - Simone Schuerle
- Responsive Biomedical Systems LabInstitute of Translational MedicineDepartment of Health Sciences and TechnologyETH ZurichZurich8093Switzerland
| | - Ali Khademhosseini
- Department of BioengineeringUniversity of California‐Los AngelesLos AngelesCA90095USA
| | - Simon P. Hoerstrup
- Institute for Regenerative Medicine (IREM)University of Zurich and Wyss Translational Center ZurichZurich8952Switzerland
| | - Viola Vogel
- Laboratory of Applied MechanobiologyInstitute of Translational MedicineDepartment of Health Sciences and TechnologyETH ZurichZurich8093Switzerland
| |
Collapse
|
31
|
Ren P, Wu D, Appel R, Zhang L, Zhang C, Luo W, Robertson AAB, Cooper MA, Coselli JS, Milewicz DM, Shen YH, LeMaire SA. Targeting the NLRP3 Inflammasome With Inhibitor MCC950 Prevents Aortic Aneurysms and Dissections in Mice. J Am Heart Assoc 2020; 9:e014044. [PMID: 32223388 PMCID: PMC7428617 DOI: 10.1161/jaha.119.014044] [Citation(s) in RCA: 81] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Background Aortic aneurysms and dissections are highly lethal diseases for which an effective treatment strategy is critically needed to prevent disease progression. The nucleotide‐binding oligomerization domain–like receptor pyrin domain containing 3 (NLRP3)–caspase‐1 inflammasome cascade was recently shown to play an important role in aortic destruction and disease development. In this study, we tested the effects of MCC950, a potent, selective NLRP3 inhibitor, on preventing aortic destruction and aortic aneurysm and dissection formation. Methods and Results In a model of sporadic aortic aneurysm and dissection induced by challenging wild‐type mice with a high‐fat, high‐cholesterol diet and angiotensin II infusion, MCC950 treatment significantly inhibited challenge‐induced aortic dilatation, dissection, and rupture in different thoracic and abdominal aortic segments in both male and female mice. Aortic disease reduction by MCC950 was associated with the prevention of NLRP3–caspase‐1 upregulation, smooth muscle cell contractile protein degradation, aortic cell death, and extracellular matrix destruction. Further investigation revealed that preventing matrix metallopeptidase 9 (MMP‐9) expression and activation in macrophages is an important mechanism underlying MCC950's protective effect. We found that caspase‐1 directly activated MMP‐9 by cleaving its N‐terminal inhibitory domain. Moreover, the genetic knockdown of Nlrp3 or Casp‐1 in mice or treatment of mice with MCC950 diminished the challenge‐induced N‐terminal cleavage of MMP‐9, MMP‐9 activation, and aortic destruction. Conclusions Our findings suggest that the NLRP3–caspase‐1 inflammasome directly activates MMP‐9. Targeting the inflammasome with MCC950 is a promising approach for preventing aortic destruction and aortic aneurysm and dissection development.
Collapse
Affiliation(s)
- Pingping Ren
- Division of Cardiothoracic Surgery Michael E. DeBakey Department of Surgery Baylor College of Medicine Houston TX.,Department of Cardiovascular Surgery Texas Heart Institute Houston TX.,Cardiovascular Research Institute Baylor College of Medicine Houston TX
| | - Darrell Wu
- Division of Cardiothoracic Surgery Michael E. DeBakey Department of Surgery Baylor College of Medicine Houston TX.,Department of Cardiovascular Surgery Texas Heart Institute Houston TX.,Cardiovascular Research Institute Baylor College of Medicine Houston TX
| | - Richard Appel
- Division of Cardiothoracic Surgery Michael E. DeBakey Department of Surgery Baylor College of Medicine Houston TX.,Department of Cardiovascular Surgery Texas Heart Institute Houston TX.,Cardiovascular Research Institute Baylor College of Medicine Houston TX
| | - Lin Zhang
- Division of Cardiothoracic Surgery Michael E. DeBakey Department of Surgery Baylor College of Medicine Houston TX.,Department of Cardiovascular Surgery Texas Heart Institute Houston TX.,Cardiovascular Research Institute Baylor College of Medicine Houston TX
| | - Chen Zhang
- Division of Cardiothoracic Surgery Michael E. DeBakey Department of Surgery Baylor College of Medicine Houston TX.,Department of Cardiovascular Surgery Texas Heart Institute Houston TX.,Cardiovascular Research Institute Baylor College of Medicine Houston TX
| | - Wei Luo
- Division of Cardiothoracic Surgery Michael E. DeBakey Department of Surgery Baylor College of Medicine Houston TX.,Department of Cardiovascular Surgery Texas Heart Institute Houston TX.,Cardiovascular Research Institute Baylor College of Medicine Houston TX
| | - Avril A B Robertson
- Institute for Molecular Bioscience University of Queensland Brisbane Australia
| | - Matthew A Cooper
- Institute for Molecular Bioscience University of Queensland Brisbane Australia
| | - Joseph S Coselli
- Division of Cardiothoracic Surgery Michael E. DeBakey Department of Surgery Baylor College of Medicine Houston TX.,Department of Cardiovascular Surgery Texas Heart Institute Houston TX.,Cardiovascular Research Institute Baylor College of Medicine Houston TX
| | - Dianna M Milewicz
- Division of Medical Genetics Department of Internal Medicine The University of Texas Health Science Center at Houston TX
| | - Ying H Shen
- Division of Cardiothoracic Surgery Michael E. DeBakey Department of Surgery Baylor College of Medicine Houston TX.,Department of Cardiovascular Surgery Texas Heart Institute Houston TX.,Cardiovascular Research Institute Baylor College of Medicine Houston TX
| | - Scott A LeMaire
- Division of Cardiothoracic Surgery Michael E. DeBakey Department of Surgery Baylor College of Medicine Houston TX.,Department of Cardiovascular Surgery Texas Heart Institute Houston TX.,Cardiovascular Research Institute Baylor College of Medicine Houston TX
| |
Collapse
|
32
|
Liu B, Granville DJ, Golledge J, Kassiri Z. Pathogenic mechanisms and the potential of drug therapies for aortic aneurysm. Am J Physiol Heart Circ Physiol 2020; 318:H652-H670. [PMID: 32083977 PMCID: PMC7099451 DOI: 10.1152/ajpheart.00621.2019] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/23/2019] [Revised: 02/13/2020] [Accepted: 02/13/2020] [Indexed: 12/14/2022]
Abstract
Aortic aneurysm is a permanent focal dilation of the aorta. It is usually an asymptomatic disease but can lead to sudden death due to aortic rupture. Aortic aneurysm-related mortalities are estimated at ∼200,000 deaths per year worldwide. Because no pharmacological treatment has been found to be effective so far, surgical repair remains the only treatment for aortic aneurysm. Aortic aneurysm results from changes in the aortic wall structure due to loss of smooth muscle cells and degradation of the extracellular matrix and can form in different regions of the aorta. Research over the past decade has identified novel contributors to aneurysm formation and progression. The present review provides an overview of cellular and noncellular factors as well as enzymes that process extracellular matrix and regulate cellular functions (e.g., matrix metalloproteinases, granzymes, and cathepsins) in the context of aneurysm pathogenesis. An update of clinical trials focusing on therapeutic strategies to slow abdominal aortic aneurysm growth and efforts underway to develop effective pharmacological treatments is also provided.
Collapse
Affiliation(s)
- Bo Liu
- University of Wisconsin, Madison, Department of Surgery, Madison Wisconsin
| | - David J Granville
- International Collaboration on Repair Discoveries Centre and University of British Columbia Centre for Heart Lung Innovation, Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | - Jonathan Golledge
- The Queensland Research Centre for Peripheral Vascular Disease, College of Medicine and Dentistry, James Cook University, Department of Vascular and Endovascular Surgery, Townsville Hospital and Health Services, Townsville, Queensland, Australia
| | - Zamaneh Kassiri
- University of Alberta, Department of Physiology, Cardiovascular Research Center, Faculty of Medicine and Dentistry, Edmonton, Alberta, Canada
| |
Collapse
|
33
|
Cione E, Piegari E, Gallelli G, Caroleo MC, Lamirata E, Curcio F, Colosimo F, Cannataro R, Ielapi N, Colosimo M, de Franciscis S, Gallelli L. Expression of MMP-2, MMP-9, and NGAL in Tissue and Serum of Patients with Vascular Aneurysms and Their Modulation by Statin Treatment: A Pilot Study. Biomolecules 2020; 10:359. [PMID: 32111073 PMCID: PMC7175213 DOI: 10.3390/biom10030359] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2019] [Revised: 02/20/2020] [Accepted: 02/23/2020] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND Matrix metalloproteinases (MMPs) are involved in vascular wall degradation, and drugs able to modulate MMP activity can be used to prevent or treat aneurysmal disease. In this study, we evaluated the effects of statins on MMP-2, MMP-9, and neutrophil gelatinase-associated lipocalin (NGAL) in both plasma and tissue in patients with aneurysmal disease. METHODS We performed a prospective, single-blind, multicenter, control group clinical drug trial on 184 patients of both sexes >18 years old with a diagnosis of arterial aneurysmal disease. Enrolled patients were divided into two groups: Group I under statin treatment and Group II not taking statins. In addition, 122 patients without aneurysmal disease and under statin treatment were enrolled as a control group (Group III). The expression of MMPs and NGAL in plasma was evaluated using ELISA, while their expression in endothelial tissues was evaluated using Western blot. RESULTS The ELISA test revealed greater plasma levels (p < 0.01) of MMPs and NGAL in Groups I and II vs. Group III. Western blot analysis showed higher expression (p < 0.01) of MMPs and NGAL in Group II vs. Group I, and this increase was significantly higher (p < 0.01) in patients treated with low potency statins compared to high potency ones. CONCLUSIONS MMPs and NGAL seem to play a major role in the development of aneurysms, and their modulation by statins suggests that these drugs could be used to prevent arterial aneurysmal disease.
Collapse
Affiliation(s)
- Erika Cione
- Department of Pharmacy, Health and Nutritional Sciences, Department of Excellence 2018-2022, University of Calabria, 87036 Rende, Italy; (E.C.); (M.C.C.); (R.C.)
| | - Elena Piegari
- Department of Experimental Medicine, Section of Pharmacology, University of Campania “Luigi Vanvitelli”, 80138 Napoli, Italy;
| | - Giuseppe Gallelli
- Unit of Vascular Surgery, Department of Surgery, “Pugliese Ciaccio” Hospital, 88100 Catanzaro, Italy;
| | - Maria Cristina Caroleo
- Department of Pharmacy, Health and Nutritional Sciences, Department of Excellence 2018-2022, University of Calabria, 87036 Rende, Italy; (E.C.); (M.C.C.); (R.C.)
| | - Elena Lamirata
- Department of Experimental Medicine, University of Catanzaro, and Vascular Surgery Unit, 88100 Mater Domini Hospital, 88100 Catanzaro, Italy; (E.L.); (F.C.); (S.d.F.)
| | - Francesca Curcio
- Department of Experimental Medicine, University of Catanzaro, and Vascular Surgery Unit, 88100 Mater Domini Hospital, 88100 Catanzaro, Italy; (E.L.); (F.C.); (S.d.F.)
| | - Federica Colosimo
- National Institution of Social Insurance, Department of Medical Law, 88100 Catanzaro, Italy;
| | - Roberto Cannataro
- Department of Pharmacy, Health and Nutritional Sciences, Department of Excellence 2018-2022, University of Calabria, 87036 Rende, Italy; (E.C.); (M.C.C.); (R.C.)
| | - Nicola Ielapi
- Department of Public Health and Infectious Disease, “Sapienza” University of Rome 5, 00185 Roma, Italy;
| | - Manuela Colosimo
- Unit of Microbiology and Virology, “Pugliese Ciaccio” Hospital, 88100 Catanzaro, Italy;
| | - Stefano de Franciscis
- Department of Experimental Medicine, University of Catanzaro, and Vascular Surgery Unit, 88100 Mater Domini Hospital, 88100 Catanzaro, Italy; (E.L.); (F.C.); (S.d.F.)
| | - Luca Gallelli
- Department of Health Sciences, University of Catanzaro, and Clinical Pharmacology and Pharmacovigilance Unit, Mater Domini Hospital, 88100 Catanzaro, Italy
| |
Collapse
|
34
|
Wang Q, Luo Q, Yang Z, Zhao YH, Li J, Wang J, Piao J, Chen X. Weighted gene co-expression network analysis identified six hub genes associated with rupture of intracranial aneurysms. PLoS One 2020; 15:e0229308. [PMID: 32084215 PMCID: PMC7034829 DOI: 10.1371/journal.pone.0229308] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2019] [Accepted: 02/03/2020] [Indexed: 12/19/2022] Open
Abstract
Intracranial aneurysms (IAs) are characterized by localized dilation or ballooning of a cerebral artery. When IAs rupture, blood leaks into the space around the brain to create a subarachnoid hemorrhage. The latter is associated with a higher risk of disability and mortality. The aims of this study were to gain greater insight into the pathogenesis of ruptured IAs, and to clarify whether identified hub genes represent potential biological markers for assessing the likelihood of IA progression and rupture. Briefly, the GSE36791 and GSE73378 datasets from the National Center of Biotechnology Information Gene Expression Omnibus database were reanalyzed and subjected to a weighted gene co-expression network analysis to test the association between gene sets and clinical features. The clinical significance of these genes as potential biomarkers was also examined, with their expression validated by quantitative real-time PCR. A total of 14 co-expression modules and 238 hub genes were identified. In particular, three modules (labeled turquoise, blue, and brown) were found to highly correlate with IA rupture events. Additionally, six potential biomarkers were identified (BASP1, CEBPB, ECHDC2, GZMK, KLHL3, and SLC2A3), which are strongly associated with the progression and rupture of IAs. Taken together, these findings provide novel insights into potential molecular mechanisms responsible for IAs and they highlight the potential for these particular genes to serve as biomarkers for monitoring IA rupture.
Collapse
Affiliation(s)
- Qunhui Wang
- Department of Neurosurgery, The First Hospital of Jilin University, Changchun, Jilin, P. R. China
| | - Qi Luo
- Department of Neurosurgery, The First Hospital of Jilin University, Changchun, Jilin, P. R. China
| | - Zhongxi Yang
- Department of Neurosurgery, The First Hospital of Jilin University, Changchun, Jilin, P. R. China
| | - Yu-Hao Zhao
- Department of Neurosurgery, The First Hospital of Jilin University, Changchun, Jilin, P. R. China
| | - Jiaqi Li
- Department of Neurosurgery, The First Hospital of Jilin University, Changchun, Jilin, P. R. China
| | - Jian Wang
- Department of Neurosurgery, The First Hospital of Jilin University, Changchun, Jilin, P. R. China
| | - Jianmin Piao
- Department of Neurosurgery, The First Hospital of Jilin University, Changchun, Jilin, P. R. China
- * E-mail: (XC); (JP)
| | - Xuan Chen
- Department of Neurosurgery, The First Hospital of Jilin University, Changchun, Jilin, P. R. China
- * E-mail: (XC); (JP)
| |
Collapse
|
35
|
Li Z, Kong W. Cellular signaling in Abdominal Aortic Aneurysm. Cell Signal 2020; 70:109575. [PMID: 32088371 DOI: 10.1016/j.cellsig.2020.109575] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2020] [Revised: 02/19/2020] [Accepted: 02/19/2020] [Indexed: 12/31/2022]
Abstract
Abdominal aortic aneurysms (AAAs) are highly lethal cardiovascular diseases without effective medications. However, the molecular and signaling mechanisms remain unclear. A series of pathological cellular processes have been shown to contribute to AAA formation, including vascular extracellular matrix remodeling, inflammatory and immune responses, oxidative stress, and dysfunction of vascular smooth muscle cells. Each cellular process involves complex cellular signaling, such as NF-κB, MAPK, TGFβ, Notch and inflammasome signaling. In this review, we discuss how cellular signaling networks function in various cellular processes during the pathogenesis and progression of AAA. Understanding the interaction of cellular signaling networks with AAA pathogenesis as well as the crosstalk of different signaling pathways is essential for the development of novel therapeutic approaches to and personalized treatments of AAA diseases.
Collapse
Affiliation(s)
- Zhiqing Li
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing 100191, China
| | - Wei Kong
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing 100191, China.
| |
Collapse
|
36
|
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
|
37
|
Farrell K, Simmers P, Mahajan G, Boytard L, Camardo A, Joshi J, Ramamurthi A, Pinet F, Kothapalli CR. Alterations in phenotype and gene expression of adult human aneurysmal smooth muscle cells by exogenous nitric oxide. Exp Cell Res 2019; 384:111589. [PMID: 31473210 DOI: 10.1016/j.yexcr.2019.111589] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2019] [Revised: 08/24/2019] [Accepted: 08/29/2019] [Indexed: 12/25/2022]
Abstract
Abdominal aortic aneurysms (AAA) are characterized by matrix remodeling, elastin degradation, absence of nitric oxide (NO) signaling, and inflammation, influencing smooth muscle cell (SMC) phenotype and gene expression. Little is known about the biomolecular release and intrinsic biomechanics of human AAA-SMCs. NO delivery could be an attractive therapeutic strategy to restore lost functionality of AAA-SMCs by inhibiting inflammation and cell stiffening. We aim to establish the differences in phenotype and gene expression of adult human AAA-SMCs from healthy SMCs. Based on our previous study which showed benefits of optimal NO dosage delivered via S-Nitrosoglutathione (GSNO) to healthy aortic SMCs, we tested whether such benefits would occur in AAA-SMCs. The mRNA expression of three genes involved in matrix degradation (ACE, ADAMTS5 and ADAMTS8) was significantly downregulated in AAA-SMCs. Total protein and glycosaminoglycans synthesis were higher in AAA-SMCs than healthy-SMCs (p < 0.05 for AAA-vs. healthy- SMC cultures) and was enhanced by GSNO and 3D cultures (p < 0.05 for 3D vs. 2D cultures; p < 0.05 for GSNO vs. non-GSNO cases). Elastin gene expression, synthesis and deposition, desmosine crosslinker levels, and lysyl oxidase (LOX) functional activity were lower, while cell proliferation, iNOS, LOX and fibrillin-1 gene expressions were higher in AAA-SMCs (p < 0.05 between respective cases), with differential benefits from GSNO exposure. GSNO and 3D cultures reduced MMPs -2, -9, and increased TIMP-1 release in AAA-SMC cultures (p < 0.05 for GSNO vs. non-GSNO cultures). AAA-SMCs were inherently stiffer and had smoother surface than healthy SMCs (p < 0.01 in both cases), but GSNO reduced stiffness (~25%; p < 0.01) and increased roughness (p < 0.05) of both cell types. In conclusion, exogenously-delivered NO offers an attractive strategy by providing therapeutic benefits to AAA-SMCs.
Collapse
Affiliation(s)
- Kurt Farrell
- Department of Chemical and Biomedical Engineering, Cleveland State University, Cleveland, OH, 44141, USA
| | - Phillip Simmers
- Department of Chemical and Biomedical Engineering, Cleveland State University, Cleveland, OH, 44141, USA
| | - Gautam Mahajan
- Department of Chemical and Biomedical Engineering, Cleveland State University, Cleveland, OH, 44141, USA
| | - Ludovic Boytard
- University of Lille, Inserm U1167, Institut Pasteur de Lille, France
| | - Andrew Camardo
- Department of Biomedical Engineering, Cleveland Clinic, Cleveland, OH, 44141, USA
| | - Jyotsna Joshi
- Department of Chemical and Biomedical Engineering, Cleveland State University, Cleveland, OH, 44141, USA
| | - Anand Ramamurthi
- Department of Biomedical Engineering, Cleveland Clinic, Cleveland, OH, 44141, USA
| | - Florence Pinet
- University of Lille, Inserm U1167, Institut Pasteur de Lille, France
| | - Chandrasekhar R Kothapalli
- Department of Chemical and Biomedical Engineering, Cleveland State University, Cleveland, OH, 44141, USA.
| |
Collapse
|
38
|
Matrix Metalloproteinase in Abdominal Aortic Aneurysm and Aortic Dissection. Pharmaceuticals (Basel) 2019; 12:ph12030118. [PMID: 31390798 PMCID: PMC6789891 DOI: 10.3390/ph12030118] [Citation(s) in RCA: 119] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2019] [Revised: 07/25/2019] [Accepted: 08/02/2019] [Indexed: 12/12/2022] Open
Abstract
Abdominal Aortic Aneurysm (AAA) affects 4–5% of men over 65, and Aortic Dissection (AD) is a life-threatening aortic pathology associated with high morbidity and mortality. Initiators of AAA and AD include smoking and arterial hypertension, whilst key pathophysiological features of AAA and AD include chronic inflammation, hypoxia, and large modifications to the extra cellular matrix (ECM). As it stands, only surgical methods are available for preventing aortic rupture in patients, which often presents difficulties for recovery. No pharmacological treatment is available, as such researchers are attempting to understand the cellular and molecular pathophysiology of AAA and AD. Upregulation of matrix metalloproteinase (MMPs), particularly MMP-2 and MMP-9, has been identified as a key event occurring during aneurysmal growth. As such, several animal models of AAA and AD have been used to investigate the therapeutic potential of suppressing MMP-2 and MMP-9 activity as well as modulating the activity of other MMPs, and TIMPs involved in the pathology. Whilst several studies have offered promising results, targeted delivery of MMP inhibition still needs to be developed in order to avoid surgery in high risk patients.
Collapse
|
39
|
Yuwen L, Ciqiu Y, Yi S, Ruilei L, Yuanhui L, Bo L, Songqi L, Weiming L, Jie L. A Pilot Study of Protein Microarray for Simultaneous Analysis of 274 Cytokines Between Abdominal Aortic Aneurysm and Normal Aorta. Angiology 2019; 70:830-837. [PMID: 31018647 DOI: 10.1177/0003319719844678] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Cytokines play an important role in the pathogenesis of abdominal aortic aneurysm (AAA). We evaluated the cytokine expression profile of large AAA walls using a 274-cytokine protein array. We hypothesized that AAAs are characterized by an inflammatory, chemotactic cytokine profile. We investigated the cytokine expression profile of 12 patients with AAA and 6 nonaneurysmal controls using an antibody-based protein array. The array generated antibodies against homogenized human aortic tissues to validate the cytokines differentially expressed in AAAs and normal aortas. Data were quantified using fluorescent signal intensities and statistically analyzed by the t test. Fifty-nine cytokines were differentially expressed between the AAA and control samples. Of the 35 selected cytokines that had relative expression >1000, 29 were significantly higher and 6 were lower in AAA samples than in controls. They respectively belonged to CC chemokines, CXC chemokines, pro-inflammatory cytokines, growth factors, proteolytic proteins and inhibitors, and cell adhesion cytokines. Our results show that distinct cytokines are involved in AAAs and suggest that the pathways involving these cytokines may be associated with the pathogenesis and development of AAAs. These findings, if confirmed by larger studies, may suggest treatment targets.
Collapse
Affiliation(s)
- Li Yuwen
- Department of Gastroenterology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong Province, China
| | - Yang Ciqiu
- Department of Breast Cancer, Cancer Center, Guangdong Provincial People’s Hospital & Guangdong Academy of Medical Sciences, Guangzhou, Guangdong Province, China
| | - Shi Yi
- Division of Vascular Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong Province, China
| | - Liu Ruilei
- Department of Breast and Thyroid Surgery, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong Province, China
| | - Lai Yuanhui
- Division of Vascular Surgery, The Eastern Hospital of the First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong Province, China
| | - Lin Bo
- Department of Breast and Thyroid Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong Province, China
| | - Li Songqi
- Department of Breast and Thyroid Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong Province, China
| | - Lv Weiming
- Department of Breast and Thyroid Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong Province, China
| | - Li Jie
- Department of Breast and Thyroid Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong Province, China
| |
Collapse
|
40
|
Phillips EH, Lorch AH, Durkes AC, Goergen CJ. Early pathological characterization of murine dissecting abdominal aortic aneurysms. APL Bioeng 2018; 2:046106. [PMID: 31069328 PMCID: PMC6481730 DOI: 10.1063/1.5053708] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2018] [Accepted: 12/10/2018] [Indexed: 12/11/2022] Open
Abstract
We report here on the early pathology of a well-established murine model of dissecting abdominal aortic aneurysms (AAAs). Continuous infusion of angiotensin II (AngII) into apolipoprotein E-deficient mice induces the formation of aortic dissection and expansion at some point after implantation of miniosmotic pumps containing AngII. While this model has been studied extensively at a chronic stage, we investigated the early pathology of dissecting AAA formation at multiple scales. Using high-frequency ultrasound, we screened 12-week-old male mice daily for initial formation of these aneurysmal lesions between days 3 and 10 post-implantation. We euthanized animals on the day of diagnosis of a dissecting AAA or at day 10 if no aneurysmal lesion developed. Aortic expansion and reduced vessel wall strain occurred in animals regardless of whether a dissecting AAA developed by day 10. The aortas of mice that did not develop dissecting AAAs showed intermediate changes in morphology and biomechanical properties. RNA sequencing and gene expression analysis revealed multiple proinflammatory and matrix remodeling genes to be upregulated in the suprarenal aorta of AngII-infused mice as compared to saline-infused controls. Histology and immunohistochemistry confirmed that extracellular matrix remodeling and inflammatory cell infiltration, notably neutrophils and macrophages, occurred in AngII-infused mice with and without dissecting AAAs but not saline-infused controls. Understanding early disease processes is a critical step forward in translating experimental results in cardiovascular disease research. This work advances our understanding of this well-established murine model with applications for improving early diagnosis and therapy of acute aortic syndrome in humans.
Collapse
Affiliation(s)
- Evan H Phillips
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, Indiana 47907, USA
| | - Adam H Lorch
- Department of Biology, Purdue University, West Lafayette, Indiana 47907, USA
| | - Abigail C Durkes
- Department of Comparative Pathobiology, Purdue University, West Lafayette, Indiana 47907, USA
| | | |
Collapse
|
41
|
Wiernicki I, Parafiniuk M, Kolasa-Wołosiuk A, Gutowska I, Kazimierczak A, Clark J, Baranowska-Bosiacka I, Szumilowicz P, Gutowski P. Relationship between aortic wall oxidative stress/proteolytic enzyme expression and intraluminal thrombus thickness indicates a novel pathomechanism in the progression of human abdominal aortic aneurysm. FASEB J 2018; 33:885-895. [PMID: 30351992 DOI: 10.1096/fj.201800633r] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The possibility that oxidative stress promotes degradation of the extracellular matrix and a relationship between intraluminal thrombus (ILT) thickness and proteolytic activity within the abdominal aortic aneurysm (AAA) wall has been suggested. In the present study, the hypothesis that thin ILT is correlated with an increase in oxidative stress-related enzymes and matrix metalloproteinase-9 (MMP-9) expression within the human AAA wall was investigated. We also studied the antioxidant activity of superoxide dismutases, catalase, glutathione peroxidase, glutathione reductase, and thioredoxin within the full-thickness AAA wall and through fluoroimmunohistochemical staining of catalase and MMP-9 expression within the inner and outer media, in relation to ILT thickness. Reactive oxygen species control the degradation and remodeling of the extracellular matrix by up-regulating proteolytic enzymes, such as MMPs. Results showed that oxidative stress and proteolytic enzyme expression were simultaneously, significantly higher within thin thrombus (≤10 mm)-covered aneurysm wall when compared with the wall covered by thick thrombus (≥25 mm). These findings provide the first demonstration, to our knowledge, of a causative link between oxidative stress instigating proteolytic enzyme expression at the tissue level and human AAA development. Presence of a thin circumferential thrombus should always be considered as a risk factor for the greatest increase in aneurysm growth rate and rupture, giving an indication for surgery timing.-Wiernicki, I., Parafiniuk, M., Kolasa-Wołosiuk, A., Gutowska, I., Kazimierczak, A., Clark, J., Baranowska-Bosiacka, I., Szumilowicz, P., Gutowski, P. Relationship between aortic wall oxidative stress/proteolytic enzyme expression and intraluminal thrombus thickness indicates a novel pathomechanism in the progression of human abdominal aortic aneurysm.
Collapse
Affiliation(s)
- Ireneusz Wiernicki
- Department of Vascular Surgery and Angiology, Pomeranian Medical University, Szczecin, Poland
| | - Miroslaw Parafiniuk
- Department of Forensic Medicine, Pomeranian Medical University, Szczecin, Poland
| | | | - Izabela Gutowska
- Department of Biochemistry and Human Nutrition, Pomeranian Medical University, Szczecin, Poland
| | - Arkadiusz Kazimierczak
- Department of Vascular Surgery and Angiology, Pomeranian Medical University, Szczecin, Poland
| | - Jeremy Clark
- Department of Clinical and Molecular Biochemistry, Faculty of Laboratory Diagnostics and Molecular Medicine, Pomeranian Medical University, Szczecin, Poland
| | - Irena Baranowska-Bosiacka
- Department of Biochemistry and Medical Chemistry, Faculty of Laboratory Diagnostics and Molecular Medicine, Pomeranian Medical University, Szczecin, Poland
| | - Pawel Szumilowicz
- Department of Vascular Surgery and Angiology, Pomeranian Medical University, Szczecin, Poland
| | - Piotr Gutowski
- Department of Vascular Surgery and Angiology, Pomeranian Medical University, Szczecin, Poland
| |
Collapse
|
42
|
Zhou B, Li W, Zhao G, Yu B, Ma B, Liu Z, Xie N, Fu Y, Gong Z, Dai R, Zhang X, Kong W. Rapamycin prevents thoracic aortic aneurysm and dissection in mice. J Vasc Surg 2018; 69:921-932.e3. [PMID: 30253896 DOI: 10.1016/j.jvs.2018.05.246] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2018] [Accepted: 05/25/2018] [Indexed: 11/19/2022]
Abstract
OBJECTIVE The purpose of this study was to investigate whether rapamycin inhibits the development of thoracic aortic aneurysm and dissection (TAAD) in mice. METHODS Three-week-old C57BL/6J male mice were fed a normal diet and randomized into a control group (n = 6), β-aminopropionitrile fumarate (BAPN) group (Gp A; n = 15), BAPN plus rapamycin (5 mg) group (Gp B; n = 8), and BAPN plus rapamycin (10 mg) group (Gp C; n = 8). Gp A, Gp B, and Gp C were administered BAPN (1 g/kg/d) for 4 weeks. One week after BAPN administration, Gp B and Gp C were treated with rapamycin (5 mg/kg/d or 10 mg/kg/d) through gavage for 21 days. Thoracic aortas were harvested for Western blot and immunofluorescence staining at day 14 and for morphologic and histologic analyses at day 28. RESULTS BAPN treatment induced TAAD formation in mice. The incidence of TAAD in control, Gp A, Gp B, and Gp C mice was 0%, 80%, 25%, and 37.5%, respectively. Smaller thoracic aortic diameters (ascending aorta and arch) were observed in Gp B and Gp C mice than in Gp A mice (Gp B vs Gp A: ascending aorta, ex vivo, 1.07 ± 0.21 mm vs 1.80 ± 0.67 mm [P < .05]; aortic arch, ex vivo, 1.51 ± 0.40 mm vs 2.70 ± 1.06 mm [P < .05]; Gp C vs Gp A: ascending aortas, ex vivo, 1.10 ± 0.33 mm vs 1.80 ± 0.67 mm [P < .05]; aortic arch, ex vivo, 1.55 ± 0.56 mm vs 2.70 ± 1.06 mm [P < .05]). TAAD mice exhibited elastin fragmentation, abundant inflammatory cell infiltration, and significantly increased matrix metalloproteinase production in the aorta, and rapamycin treatment alleviated these changes. The protein levels of p-S6K and p-S6 in TAAD aortic tissues increased significantly, whereas they were suppressed by rapamycin. CONCLUSIONS Rapamycin suppressed TAAD formation, probably by inhibition of mechanistic target of rapamycin signaling and reduction of inflammatory cell infiltration and matrix metalloproteinase 9 production. Targeting of the mechanistic target of rapamycin signaling pathway using rapamycin may be a favorable modulation for the clinical treatment of TAAD.
Collapse
MESH Headings
- Aminopropionitrile
- Aortic Dissection/chemically induced
- Aortic Dissection/enzymology
- Aortic Dissection/pathology
- Aortic Dissection/prevention & control
- Animals
- Anti-Inflammatory Agents/pharmacology
- Aorta, Thoracic/drug effects
- Aorta, Thoracic/enzymology
- Aorta, Thoracic/pathology
- Aortic Aneurysm, Thoracic/chemically induced
- Aortic Aneurysm, Thoracic/enzymology
- Aortic Aneurysm, Thoracic/pathology
- Aortic Aneurysm, Thoracic/prevention & control
- Dilatation, Pathologic
- Disease Models, Animal
- Male
- Matrix Metalloproteinase 9/metabolism
- Mice, Inbred C57BL
- Phosphorylation
- Protein Kinase Inhibitors/pharmacology
- Ribosomal Protein S6 Kinases/metabolism
- Signal Transduction/drug effects
- Sirolimus/pharmacology
- TOR Serine-Threonine Kinases/antagonists & inhibitors
- TOR Serine-Threonine Kinases/metabolism
- Vascular Remodeling/drug effects
Collapse
Affiliation(s)
- Biao Zhou
- Department of Vascular Surgery, Peking University People's Hospital, Peking University, Beijing, China
| | - Wei Li
- Department of Vascular Surgery, Peking University People's Hospital, Peking University, Beijing, China
| | - Guizhen Zhao
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, Beijing, China; Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing, China
| | - Bing Yu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, Beijing, China; Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing, China
| | - Baihui Ma
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, Beijing, China; Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing, China
| | - Zhujiang Liu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, Beijing, China; Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing, China
| | - Nan Xie
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, Beijing, China; Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing, China
| | - Yi Fu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, Beijing, China; Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing, China
| | - Ze Gong
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, Beijing, China; Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing, China
| | - Rongbo Dai
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, Beijing, China; Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing, China
| | - Xiaoming Zhang
- Department of Vascular Surgery, Peking University People's Hospital, Peking University, Beijing, China.
| | - Wei Kong
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, Beijing, China; Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing, China
| |
Collapse
|
43
|
Tenorio EJR, Braga AFF, Tirapelli DPDC, Ribeiro MS, Piccinato CE, Joviliano EE. Expression in Whole Blood Samples of miRNA-191 and miRNA-455-3p in Patients with AAA and Their Relationship to Clinical Outcomes after Endovascular Repair. Ann Vasc Surg 2018. [DOI: 10.1016/j.avsg.2018.01.086] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
|
44
|
Interleukin-3 is required for thoracic aneurysm and dissection in a mouse model. Clin Sci (Lond) 2018; 132:1253-1256. [DOI: 10.1042/cs20180185] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2018] [Revised: 05/16/2018] [Accepted: 05/18/2018] [Indexed: 11/17/2022]
Abstract
The pathogenesis of thoracic aortic aneurysm and dissection (TAAD) is complex and incompletely understood. The hallmarks of the disease process are aortic inflammatory cell infiltration and protease mediated elastic fiber disruption. In a study recently published in Clinical Science (2018) 132 (6), 655–668), Liu et al. explore the mechanism through which aortic vascular smooth cells and macrophages participate in TAAD using a mouse model. The authors propose that interleukin-3 (IL-3) released from aortic vascular smooth cells is central to the disease process. IL-3 stimulated matrix metalloproteinase 12 (MMP12) release from macrophages via mitogen activated protein kinase pathways. MMP12 is a protease known to be involved in both aortic aneurysm and dissection. IL-3 knockout mice had significantly reduced aortic wall MMP12, and reduced protease activity. This was associated with protection against TAAD.
Collapse
|
45
|
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
|
46
|
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
|
47
|
Chen YJ, Tsai TH, Wang LY, Hsieh CH. Local Radiotherapy Affects Drug Pharmacokinetics-Exploration of a Neglected but Significant Uncertainty of Cancer Therapy. Technol Cancer Res Treat 2017; 16:705-716. [PMID: 29332468 PMCID: PMC5762083 DOI: 10.1177/1533034617737011] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Purpose: Concurrent chemoradiation therapy is the mainstay of treatment for many types of malignancies. However, concurrent chemoradiation therapy is associated with a greater number of systemic adverse effects than radiotherapy or chemotherapy alone. Summary: Pharmacokinetics is the study of a drug and/or its metabolite kinetics in the body, including absorption, distribution, metabolism, and elimination. The incidences of adverse effects are markedly higher in patients who receive concurrent chemoradiation therapy than in those who receive either radiotherapy or chemotherapy alone. This phenomenon implies that irradiation affects the pharmacokinetics of cytotoxic agents, namely the radiotherapy–pharmacokinetic phenomenon. Experimental animal studies have shown that local irradiation affects the systemic pharmacokinetics of 5-fluorouracil and cisplatin at both low dose (simulating generous dose distributed to normal tissues) and daily practice dose (mimicking therapeutic dose to target volumes). These effects are significant in the circulation of blood and lymphatic system as well as in the hepatobiliary excretion. Furthermore, recent studies have demonstrated that matrix metalloproteinase-8 plays an important role in the radiotherapy–pharmacokinetic phenomenon. Conclusion: In the present review, we provide a general overview of the radiotherapy–pharmacokinetic phenomenon and discuss the possible mechanisms governing the phenomenon.
Collapse
Affiliation(s)
- Yu-Jen Chen
- Institute of Traditional Medicine, School of Medicine, National Yang-Ming University, Taipei, Taiwan.,Department of Radiation Oncology, Mackay Memorial Hospital, Taipei, Taiwan.,Department of Medical Research, Mackay Memorial Hospital, Taipei, Taiwan
| | - Tung-Hu Tsai
- Institute of Traditional Medicine, School of Medicine, National Yang-Ming University, Taipei, Taiwan.,Department of Chemical Engineering, National United University, Miaoli, Taiwan
| | - Li-Ying Wang
- School and Graduate Institute of Physical Therapy, College of Medicine, National Taiwan University, Taipei, Taiwan.,Physical Therapy Center, National Taiwan University Hospital, Taipei, Taiwan
| | - Chen-Hsi Hsieh
- Institute of Traditional Medicine, School of Medicine, National Yang-Ming University, Taipei, Taiwan.,Faculty of Medicine, School of Medicine, National Yang-Ming University, Taipei, Taiwan.,Division of Radiation Oncology, Department of Radiology, Far Eastern Memorial Hospital, Taipei, Taiwan
| |
Collapse
|
48
|
Wang G, Bi L, Wang G, Huang F, Lu M, Zhu K. Microarray analysis to identify the similarities and differences of pathogenesis between aortic occlusive disease and abdominal aortic aneurysm. Vascular 2017; 26:301-314. [PMID: 29087237 DOI: 10.1177/1708538117736695] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Objectives Expression profile of GSE57691 was analyzed to identify the similarities and differences between aortic occlusive disease and abdominal aortic aneurysm. Methods The expression profile of GSE57691 was downloaded from Gene Expression Omnibus database, including 20 small abdominal aortic aneurysm samples, 29 large abdominal aortic aneurysm samples, 9 aortic occlusive disease samples, and 10 control samples. Using the limma package in R, the differentially expressed genes were screened. Followed by enrichment analysis was performed for the differentially expressed genes using database for annotation, visualization, and integrated discovery online tool. Based on string online tool and Cytoscape software, protein-protein interaction network and module analyses were carried out. Moreover, integrated TF platform database and Cytoscape software were used for constructing transcriptional regulatory networks. Results As a result, 1757, 354, and 396 differentially expressed genes separately were identified in aortic occlusive disease, large abdominal aortic aneurysm, and small abdominal aortic aneurysm samples. UBB was significantly enriched in proteolysis related pathways with a high degree in three groups. SPARCL1 was another gene shared by these groups and regulated by NFIA, which had a high degree in transcriptional regulatory network. ACTB, a significant upregulated gene in abdominal aortic aneurysm samples, could be regulated by CLIC4, which was significantly enriched in cell motions. ACLY and NFIB were separately identified in aortic occlusive disease and small abdominal aortic aneurysm samples, and separately enriched in lipid metabolism and negative regulation of cell proliferation. Conclusions The downregulated UBB, NFIA, and SPARCL1 might play key roles in both aortic occlusive disease and abdominal aortic aneurysm, while the upregulated ACTB might only involve in abdominal aortic aneurysm. ACLY and NFIB were specifically involved in aortic occlusive disease and small abdominal aortic aneurysm separately.
Collapse
Affiliation(s)
- Guofu Wang
- Department of Vascular Surgery, Shaoxing Hospital of China Medical University, Shaoxing, China
| | - Lechang Bi
- Department of Vascular Surgery, Shaoxing Hospital of China Medical University, Shaoxing, China
| | - Gaofeng Wang
- Department of Vascular Surgery, Shaoxing Hospital of China Medical University, Shaoxing, China
| | - Feilai Huang
- Department of Vascular Surgery, Shaoxing Hospital of China Medical University, Shaoxing, China
| | - Mingjing Lu
- Department of Vascular Surgery, Shaoxing Hospital of China Medical University, Shaoxing, China
| | - Kai Zhu
- Department of Vascular Surgery, Shaoxing Hospital of China Medical University, Shaoxing, China
| |
Collapse
|
49
|
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: 423] [Impact Index Per Article: 52.9] [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
|
50
|
Qin W, Cao Y, Li L, Chen W, Chen X. Upregulation of ADAMTS‑7 and downregulation of COMP are associated with aortic aneurysm. Mol Med Rep 2017; 16:5459-5463. [PMID: 28849199 PMCID: PMC5647091 DOI: 10.3892/mmr.2017.7293] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2016] [Accepted: 06/29/2017] [Indexed: 01/17/2023] Open
Abstract
Aortic aneurysm (AA) remains a fatal condition with high rates of morbidity and mortality, and the associated underlying mechanism influencing its pathology remains to be elucidated. A disintegrin and metalloproteinase with thrombospondin motifs (ADAMTS)-7 has previously been demonstrated to be involved in the pathogenesis of vascular atherosclerosis via degradation of cartilage oligomeric matrix protein (COMP). The ADAMTS-7/COMP pathway may therefore act as a potential therapeutic target for vascular disorders. To the best of the author's knowledge, the present study aimed to investigate for the first time, the expression of ADAMTS-7 and COMP in human AA. Human aortic aneurysm samples were collected from patients with AA (n=24), and ascending aorta control samples were harvested from dilated cardiomyopathy patients who underwent heart transplantation (n=18). Expression levels of ADAMTS-7 and matrix metalloproteinase-9 were significantly increased in the AA group, as detected by immunohistochemistry (P<0.05). The COMP protein level was markedly decreased in the AA group when compared with the control group, as demonstrated via immunohistochemistry and western blot analysis (P<0.05). The findings suggest that upregulation of ADAMTS-7 and downregulation of COMP are associated with induction of human AA. ADAMTS-7/COMP pathway may provide therefore act as a potential therapeutic target in human AA for efficient, optimal treatment interventions in the future.
Collapse
Affiliation(s)
- Wei Qin
- Department of Cardiothoracic Surgery, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu 210006, P.R. China
| | - Yide Cao
- Department of Cardiothoracic Surgery, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu 210006, P.R. China
| | - Liangpeng Li
- Department of Cardiothoracic Surgery, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu 210006, P.R. China
| | - Wen Chen
- Department of Cardiothoracic Surgery, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu 210006, P.R. China
| | - Xin Chen
- Department of Cardiothoracic Surgery, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu 210006, P.R. China
| |
Collapse
|