1
|
Li H, Hu Q, Zhu D, Wu D. The Role of NAD + Metabolism in Cardiovascular Diseases: Mechanisms and Prospects. Am J Cardiovasc Drugs 2025; 25:307-327. [PMID: 39707143 DOI: 10.1007/s40256-024-00711-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 12/03/2024] [Indexed: 12/23/2024]
Abstract
Nicotinamide adenine dinucleotide (NAD+) is a promising anti-aging molecule that plays a role in cellular energy metabolism and maintains redox homeostasis. Additionally, NAD+ is involved in regulating deacetylases, DNA repair enzymes, inflammation, and epigenetics, making it indispensable in maintaining the basic functions of cells. Research on NAD+ has become a hotspot, particularly regarding its potential in cardiovascular disease (CVD). Many studies have demonstrated that NAD+ plays a crucial role in the occurrence and development of CVD. This review summarizes the biosynthesis and consumption of NAD+, along with its precursors and their effects on raising NAD+ levels. We also discuss new mechanisms of NAD+ regulation in cardiovascular risk factors and its effects of NAD+ on atherosclerosis, aortic aneurysm, heart failure, hypertension, myocardial ischemia-reperfusion injury, diabetic cardiomyopathy, and dilated cardiomyopathy, elucidating different mechanisms and potential treatments. NAD+-centered therapy holds promising advantages and prospects in the field of CVD.
Collapse
Affiliation(s)
- Huimin Li
- Department of Pharmacy, Tongji Hospital, School of Medicine, Tongji University, Shanghai, 200065, China
| | - Qingxun Hu
- Department of Pharmacy, School of Medicine, Shanghai University, Shanghai, 200444, China
| | - Deqiu Zhu
- Department of Pharmacy, Tongji Hospital, School of Medicine, Tongji University, Shanghai, 200065, China.
| | - Dan Wu
- Department of Pharmacy, Tongji Hospital, School of Medicine, Tongji University, Shanghai, 200065, China.
| |
Collapse
|
2
|
Yan J, Xu X, Zhu Y, Wang Y, Duan X. Escin Ia ameliorates DSS-induced chronic colitis in mice by inhibiting inflammation and oxidative stress via the LOXL2/MMP-9 pathway. JOURNAL OF ETHNOPHARMACOLOGY 2025; 345:119623. [PMID: 40090427 DOI: 10.1016/j.jep.2025.119623] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/30/2024] [Revised: 02/16/2025] [Accepted: 03/10/2025] [Indexed: 03/18/2025]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Aesculus wilsonii Rehd.'s dried mature seeds are the source of escin, a significant triterpenoid saponin. Aesculus wilsonii Rehd was first mentioned in the Compendium of Materia Medica, according to the Chinese Pharmacopoeia. It possesses the effectiveness of anti-inflammatory as well as treating gastrointestinal disorders. Escin Ia is the primary active component of escin, exhibiting significant antioxidant and anti-inflammatory properties. An increasing number of studies have demonstrated that escin exhibits a broad spectrum of pharmacological activities beneficial for the protection against gastrointestinal diseases. AIM OF THE STUDY Ulcerative colitis (UC) is a chronic inflammatory bowel disease (IBD) that can be managed through pharmacological treatment; however, it features a high recurrence rate as well as propensity for complications. Therefore, reducing the rate of recurrence and improving the recurrence symptoms should be the primary focus of clinical prevention and treatment. Therefore, this research aims to study the effects of escin Ia on inflammation as well as oxidative stress in mice with chronic UC and to explain the molecular mechanisms underlying its potential to improve recurrent symptoms in UC mice. MATERIALS AND METHODS A mouse model of colitis produced via dextran sodium sulfate (DSS) was developed for in vivo studies. A model of inflammation was created in vitro using caco-2 cells that were generated by lipopolysaccharide (LPS). Through the observation of colitis symptoms and histological morphology in mice, the protective effect of escin Ia against colitis was ascertained. The enzyme-linked immunosorbent assay (ELISA) and biochemical kits were then harnessed to measure the levels of oxidative stress markers as well as inflammatory factors. Additionally, to identify the possible target and molecular mechanism of escin Ia, qRT-PCR and western blotting, immunofluorescence, molecular docking, and molecular dynamics modeling were employed. RESULTS We demonstrated that escin Ia remarkably improved the colitis symptoms as well as histological features of DSS-treated mice, lowered the levels of proinflammatory cytokines as well as oxidative stress biomarkers, and subsequently restored the permeability of the intestinal mucosa. Additionally, high expression of LOXL2 significantly reduced the protective effects of escin Ia in both inflamed mice and Caco-2 cells. Furthermore, escin Ia exhibited a strong binding affinity and notable stability with LOXL2. CONCLUSION Escin Ia inhibits inflammation and oxidative stress through the LOXL2/MMP-9 pathway, thereby restoring intestinal mucosal barrier function. Improved recurrent symptoms in mice with enteritis.
Collapse
Affiliation(s)
- Jing Yan
- School of Pharmacy, Guilin Medical University, Guilin, 541199, China; School of Biomedical Industry, Guilin Medical University, Guilin, 541199, China
| | - Xiaotian Xu
- School of Pharmacy, Guilin Medical University, Guilin, 541199, China
| | - Yizhun Zhu
- School of Pharmacy, Guilin Medical University, Guilin, 541199, China; School of Pharmacy, Faculty of Medicine, Macau University of Science and Technology, Macau SAR, 999078, China
| | - Yuhui Wang
- School of Pharmacy, Guilin Medical University, Guilin, 541199, China; School of Biomedical Industry, Guilin Medical University, Guilin, 541199, China.
| | - Xiaoqun Duan
- School of Pharmacy, Guilin Medical University, Guilin, 541199, China; School of Biomedical Industry, Guilin Medical University, Guilin, 541199, China; School of Pharmacy, Faculty of Medicine, Macau University of Science and Technology, Macau SAR, 999078, China.
| |
Collapse
|
3
|
Ferreira HB, Trindade F, Nogueira-Ferreira R, Leite-Moreira A, Ferreira R, Dias-Neto M, Domingues MR. Lipidomic insights on abdominal aortic aneurysm and peripheral arterial disease. J Mol Med (Berl) 2025; 103:365-380. [PMID: 40011252 PMCID: PMC12003574 DOI: 10.1007/s00109-025-02524-1] [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: 07/04/2024] [Revised: 01/10/2025] [Accepted: 02/18/2025] [Indexed: 02/28/2025]
Abstract
Abdominal aortic aneurysm (AAA) and peripheral arterial disease (PAD) are two cardiovascular diseases associated with considerable morbidity, mortality and quality of life impairment. As they are multifactorial diseases, several factors contribute to their pathogenesis, including oxidative stress and lipid peroxidation, and these may have key roles in the development of these pathologies. Alterations of the lipid metabolism and lipid profile have been reported in cardiovascular diseases but to a lesser extent in AAA and PAD. Modifications in the profile of some molecular lipid species, in particular, native phospholipid and triglyceride species were mainly reported for AAA, while alterations in the fatty acid profile were noticed in the case of PAD. Oxidized phospholipids were also reported for AAA. Although AAA and PAD have a common atherosclerotic root, lipidomics demonstrates the existence of distinct lipid. Lipidomic research regarding AAA and PAD is still scarce and should be set in motion to increase the knowledge on the lipid changes that occur in these diseases, contributing not only to the discovery of new biomarkers for diagnosis and prognosis assessment but also to tailor precision medicine in the clinical field.
Collapse
Affiliation(s)
- Helena Beatriz Ferreira
- Mass Spectrometry Center, LAQV-REQUIMTE, Department of Chemistry, University of Aveiro, Campus Universitário de Santiago, 3810-193, Aveiro, Portugal.
| | - Fábio Trindade
- RISE-Health, Department of Surgery and Physiology, Faculty of Medicine, University of Porto, 4200-319, Porto, Portugal
| | - Rita Nogueira-Ferreira
- RISE-Health, Department of Surgery and Physiology, Faculty of Medicine, University of Porto, 4200-319, Porto, Portugal
| | - Adelino Leite-Moreira
- RISE-Health, Department of Surgery and Physiology, Faculty of Medicine, University of Porto, 4200-319, Porto, Portugal
- Department of Cardiothoracic Surgery, Centro Hospitalar Universitário São João, 4200-319, Porto, Portugal
| | - Rita Ferreira
- Mass Spectrometry Center, LAQV-REQUIMTE, Department of Chemistry, University of Aveiro, Campus Universitário de Santiago, 3810-193, Aveiro, Portugal
| | - Marina Dias-Neto
- RISE-Health, Department of Surgery and Physiology, Faculty of Medicine, University of Porto, 4200-319, Porto, Portugal
- Department of Angiology and Vascular Surgery, Unidade Local de Saúde São João, Porto, Portugal
| | - M Rosário Domingues
- Mass Spectrometry Center, LAQV-REQUIMTE, Department of Chemistry, University of Aveiro, Campus Universitário de Santiago, 3810-193, Aveiro, Portugal
- CESAM - Centre for Environmental and Marine Studies, Department of Chemistry, University of Aveiro, Campus Universitário de Santiago, 3810-193, Aveiro, Portugal
| |
Collapse
|
4
|
He H, Chen L, Peng J, Guo J, Xiao X, Dou C, Chen H, Zhan S, Han X, Yao W. ROS-responsive nanoparticles with selenomethionine for ferroptosis modulation in abdominal aortic aneurysm. iScience 2025; 28:111880. [PMID: 40104069 PMCID: PMC11914196 DOI: 10.1016/j.isci.2025.111880] [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: 09/03/2024] [Revised: 12/02/2024] [Accepted: 01/21/2025] [Indexed: 03/20/2025] Open
Abstract
Oxidative stress, particularly ROS accumulation, plays a key role in the development of abdominal aortic aneurysm (AAA). Surgical treatments and current drugs for AAA have limitations, including lack of specificity and significant side effects. This study constructed ROS-responsive nanoparticles using phenylthio-modified dendritic polylysine (PDP) loaded with selenomethionine (PDPs-Se) for AAA treatment, and elucidated its mechanism of action. In-vitro studies revealed that PDPs-Se enhanced the clearance of ROS by increasing the levels of superoxide dismutase (SOD) and glutathione (GSH) while reducing malondialdehyde (MDA) levels. Furthermore, PDPs-Se upregulated the expression levels of GPX4, SLC7A11, and FTH1 to suppress ferroptosis and modulate the differentiation of vascular smooth muscle cells (VSMCs) from a synthetic to a contractile phenotype. In-vivo experiments revealed that PDPs-Se attenuated the progression of AAA by inhibiting oxidative stress responses and improving the aortic wall thickness, indicating its potential as an approach for AAA therapy.
Collapse
Affiliation(s)
- Haipeng He
- Department of Vascular Surgery, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Lei Chen
- Department of Vascular Surgery, Zhejiang Provincial People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Jiaxin Peng
- Department of Vascular Surgery, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Jinyan Guo
- Department of Anesthesia, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Xue Xiao
- Department of Anesthesia, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Chaoxun Dou
- Department of Anesthesia, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Huining Chen
- Department of Vascular Surgery, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Songbiao Zhan
- Department of Vascular Surgery, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Xue Han
- Department of Anesthesia, Sun Yat-sen Memorial Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Weifeng Yao
- Department of Anesthesia, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| |
Collapse
|
5
|
Roslik M, Zharikov Y, Vovkogon A, Zharova N, Pontes-Silva A, Zharikova T. Aortic aneurysm: Correlations with phenotypes associated with connective tissue dysplasia. Microvasc Res 2025; 157:104754. [PMID: 39401669 DOI: 10.1016/j.mvr.2024.104754] [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: 06/11/2024] [Revised: 08/29/2024] [Accepted: 10/09/2024] [Indexed: 10/20/2024]
Abstract
An aortic aneurysm is a localized enlargement that exceeds the normal diameter of the vessel by 50 %, posing a risk due to the likelihood of rupture. The cause of aortic aneurysm, especially in young people, is connective tissue dysplasia, a condition characterized by defects in the assembly of collagen and elastin proteins, leading to changes in elastic properties and disruption of the formation of organs and their systems. The article presents data confirming the relationship between many morphological manifestations of connective tissue dysplasia (e.g., funnel-shaped deformation of the sternum, scoliosis of the thoracic spine, abdominal hernias, arterial tortuosity, striae of atypical localization) and the risk of aortic aneurysm formation. The literature suggests that the identified combinations of some external manifestations of connective tissue dysplasia deserve special attention and may be constitutional markers for the possible development of aortic aneurysm, which is a promising direction for further research in this area.
Collapse
Affiliation(s)
- Maria Roslik
- I.M. Sechenov First Moscow State Medical University (Sechenov University), Department of Human Anatomy and Histology, Moscow, Russia
| | - Yury Zharikov
- I.M. Sechenov First Moscow State Medical University (Sechenov University), Department of Human Anatomy and Histology, Moscow, Russia
| | - Andzhela Vovkogon
- I.M. Sechenov First Moscow State Medical University (Sechenov University), Department of Human Anatomy and Histology, Moscow, Russia
| | - Nataliya Zharova
- I.M. Sechenov First Moscow State Medical University (Sechenov University), Department of Human Anatomy and Histology, Moscow, Russia
| | - André Pontes-Silva
- Postgraduate Program in Physical Therapy, Department of Physical Therapy, Universidade Federal de São Carlos, São Carlos, São Paulo, Brazil.
| | - Tatiana Zharikova
- I.M. Sechenov First Moscow State Medical University (Sechenov University), Department of Human Anatomy and Histology, Moscow, Russia
| |
Collapse
|
6
|
Burgos-Santamaría A, Rodríguez-Rodríguez P, Arnalich-Montiel A, Arribas SM, Fernández-Riveira C, Barrio-Pérez IM, Río J, Ligero JM, Quintana-Villamandos B. OXY-SCORE and Volatile Anesthetics: A New Perspective of Oxidative Stress in EndoVascular Aneurysm Repair-A Randomized Clinical Trial. Int J Mol Sci 2024; 25:10770. [PMID: 39409100 PMCID: PMC11476523 DOI: 10.3390/ijms251910770] [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: 08/18/2024] [Revised: 09/29/2024] [Accepted: 09/29/2024] [Indexed: 10/20/2024] Open
Abstract
An aortic aneurysm (AA) is a life-threatening condition. Oxidative stress may be a common pathway linking multiple mechanisms of an AA, including vascular inflammation and metalloproteinase activity. Endovascular aneurysm repair (EVAR) is the preferred surgical approach for AA treatment. During surgery, inflammation and ischemia-reperfusion injury occur, and reactive oxygen species (ROS) play a key role in their modulation. Increased perioperative oxidative stress is associated with higher postoperative complications. The use of volatile anesthetics during surgery has been shown to reduce oxidative stress. Individual biomarkers only partially reflect the oxidative status of the patients. A global indicator of oxidative stress (OXY-SCORE) has been validated in various pathologies. This study aimed to compare the effects of the main volatile anesthetics, sevoflurane and desflurane, on oxidative status during EVAR. Eighty consecutive patients undergoing EVAR were randomized into two groups: sevoflurane and desflurane. Plasma biomarkers of oxidative damage (protein carbonylation and malondialdehyde) and antioxidant defense (total thiols, glutathione, nitrates, superoxide dismutase, and catalase activity) were measured before surgery and 24 h after EVAR. The analysis of individual biomarkers showed no significant differences between the groups. However, the OXY-SCORE was positive in the desflurane group (indicating a shift towards antioxidants) and negative in the sevoflurane group (favoring oxidants) (p < 0.044). Compared to sevoflurane, desflurane had a positive effect on oxidative stress during EVAR. The OXY-SCORE could provide a more comprehensive perspective on oxidative stress in this patient population.
Collapse
Affiliation(s)
- Alba Burgos-Santamaría
- Department of Anesthesia and Intensive Care, Gregorio Marañón’s University Hospital, 28007 Madrid, Spain; (A.A.-M.); (C.F.-R.); (I.M.B.-P.); (B.Q.-V.)
| | - Pilar Rodríguez-Rodríguez
- Department of Physiology, Faculty of Medicine, Autónoma University, 28029 Madrid, Spain; (P.R.-R.); (S.M.A.)
| | - Ana Arnalich-Montiel
- Department of Anesthesia and Intensive Care, Gregorio Marañón’s University Hospital, 28007 Madrid, Spain; (A.A.-M.); (C.F.-R.); (I.M.B.-P.); (B.Q.-V.)
| | - Silvia M. Arribas
- Department of Physiology, Faculty of Medicine, Autónoma University, 28029 Madrid, Spain; (P.R.-R.); (S.M.A.)
| | - Carmen Fernández-Riveira
- Department of Anesthesia and Intensive Care, Gregorio Marañón’s University Hospital, 28007 Madrid, Spain; (A.A.-M.); (C.F.-R.); (I.M.B.-P.); (B.Q.-V.)
| | - I. María Barrio-Pérez
- Department of Anesthesia and Intensive Care, Gregorio Marañón’s University Hospital, 28007 Madrid, Spain; (A.A.-M.); (C.F.-R.); (I.M.B.-P.); (B.Q.-V.)
| | - Javier Río
- Department of Angiology and Vascular Surgery, Gregorio Marañón’s University Hospital, 28007 Madrid, Spain; (J.R.); (J.M.L.)
| | - José Manuel Ligero
- Department of Angiology and Vascular Surgery, Gregorio Marañón’s University Hospital, 28007 Madrid, Spain; (J.R.); (J.M.L.)
| | - Begoña Quintana-Villamandos
- Department of Anesthesia and Intensive Care, Gregorio Marañón’s University Hospital, 28007 Madrid, Spain; (A.A.-M.); (C.F.-R.); (I.M.B.-P.); (B.Q.-V.)
- Department of Pharmacology and Toxicology, Faculty of Medicine, Complutense University, 28040 Madrid, Spain
| |
Collapse
|
7
|
Zu HL, Zhuang PP, Peng Y, Peng C, Peng C, Zhu ZJ, Yao Y, Yue J, Wang QS, Zhou WH, Wang HY. Dual-Drug Nanomedicine Assembly with Synergistic Anti-Aneurysmal Effects via Inflammation Suppression and Extracellular Matrix Stabilization. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2402141. [PMID: 38953313 DOI: 10.1002/smll.202402141] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Revised: 06/03/2024] [Indexed: 07/04/2024]
Abstract
Abdominal aortic aneurysm (AAA) represents a critical cardiovascular condition characterized by localized dilation of the abdominal aorta, carrying a significant risk of rupture and mortality. Current treatment options are limited, necessitating novel therapeutic approaches. This study investigates the potential of a pioneering nanodrug delivery system, RAP@PFB, in mitigating AAA progression. RAP@PFB integrates pentagalloyl glucose (PGG) and rapamycin (RAP) within a metal-organic-framework (MOF) structure through a facile assembly process, ensuring remarkable drug loading capacity and colloidal stability. The synergistic effects of PGG, a polyphenolic antioxidant, and RAP, an mTOR inhibitor, collectively regulate key players in AAA pathogenesis, such as macrophages and smooth muscle cells (SMCs). In macrophages, RAP@PFB efficiently scavenges various free radicals, suppresses inflammation, and promotes M1-to-M2 phenotype repolarization. In SMCs, it inhibits apoptosis and calcification, thereby stabilizing the extracellular matrix and reducing the risk of AAA rupture. Administered intravenously, RAP@PFB exhibits effective accumulation at the AAA site, demonstrating robust efficacy in reducing AAA progression through multiple mechanisms. Moreover, RAP@PFB demonstrates favorable biosafety profiles, supporting its potential translation into clinical applications for AAA therapy.
Collapse
Affiliation(s)
- Hong Lin Zu
- Department of Vascular Surgery, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, 510120, China
- The Key Laboratory of Advanced Interdisciplinary Studies, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, 510120, China
| | - Pei Pei Zhuang
- Department of Vascular Surgery, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, 510120, China
| | - Ying Peng
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, Hunan, 410013, China
| | - Chao Peng
- Department of Vascular Surgery, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, 510120, China
| | - Cheng Peng
- Department of Vascular Surgery, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, 510120, China
| | - Zi Jia Zhu
- Department of Vascular Surgery, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, 510120, China
| | - Ye Yao
- Department of Vascular Surgery, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, 510120, China
| | - Jie Yue
- Department of Vascular Surgery, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, 510120, China
| | - Qing Shan Wang
- Department of Vascular Surgery, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, 510120, China
| | - Wen Hu Zhou
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, Hunan, 410013, China
| | - Hai Yang Wang
- Department of Vascular Surgery, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, 510120, China
- The Key Laboratory of Advanced Interdisciplinary Studies, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, 510120, China
| |
Collapse
|
8
|
Liu H, Yang P, Chen S, Wang S, Jiang L, Xiao X, Le S, Chen S, Chen X, Ye P, Xia J. Ncf1 knockout in smooth muscle cells exacerbates angiotensin II-induced aortic aneurysm and dissection by activating the STING pathway. Cardiovasc Res 2024; 120:1081-1096. [PMID: 38639325 PMCID: PMC11288755 DOI: 10.1093/cvr/cvae081] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/22/2023] [Revised: 01/03/2024] [Accepted: 02/05/2024] [Indexed: 04/20/2024] Open
Abstract
AIMS Aortic aneurysm and dissection (AAD) is caused by the progressive loss of aortic smooth muscle cells (SMCs) and is associated with a high mortality rate. Identifying the mechanisms underlying SMC apoptosis is crucial for preventing AAD. Neutrophil cytoplasmic factor 1 (Ncf1) is essential in reactive oxygen species production and SMC apoptosis; Ncf1 absence leads to autoimmune diseases and chronic inflammation. Here, the role of Ncf1 in angiotensin II (Ang II)-induced AAD was investigated. METHODS AND RESULTS Ncf1 expression increased in injured SMCs. Bioinformatic analysis identified Ncf1 as a mediator of AAD-associated SMC damage. Ncf1 expression is positively correlated with DNA replication and repair in SMCs of AAD aortas. AAD incidence increased in Ang II-challenged Sm22CreNcf1fl mice. Transcriptomics showed that Ncf1 knockout activated the stimulator of interferon genes (STING) and cell death pathways. The effects of Ncf1 on SMC death and the STING pathway in vitro were examined. Ncf1 regulated the hydrogen peroxide-mediated activation of the STING pathway and inhibited SMC apoptosis. Mechanistically, Ncf1 knockout promoted the ubiquitination of nuclear factor erythroid 2-related factor 2 (NRF2), thereby inhibiting the negative regulatory effect of NRF2 on the stability of STING mRNA and ultimately promoting STING expression. Additionally, the pharmacological inhibition of STING activation prevented AAD progression. CONCLUSION Ncf1 deficiency in SMCs exacerbated Ang II-induced AAD by promoting NRF2 ubiquitination and degradation and activating the STING pathway. These data suggest that Ncf1 may be a potential therapeutic target for AAD treatment.
Collapse
MESH Headings
- Animals
- Angiotensin II
- Aortic Dissection/metabolism
- Aortic Dissection/pathology
- Aortic Dissection/genetics
- Aortic Dissection/chemically induced
- Aortic Dissection/prevention & control
- Signal Transduction
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/pathology
- Apoptosis
- Muscle, Smooth, Vascular/pathology
- Muscle, Smooth, Vascular/metabolism
- Disease Models, Animal
- Membrane Proteins/metabolism
- Membrane Proteins/genetics
- Mice, Knockout
- Aortic Aneurysm/metabolism
- Aortic Aneurysm/pathology
- Aortic Aneurysm/genetics
- Aortic Aneurysm/chemically induced
- Aortic Aneurysm/prevention & control
- NF-E2-Related Factor 2/metabolism
- NF-E2-Related Factor 2/genetics
- NF-E2-Related Factor 2/deficiency
- Cells, Cultured
- Mice, Inbred C57BL
- Male
- Ubiquitination
- NADPH Oxidases/metabolism
- NADPH Oxidases/genetics
- Humans
- Mice
Collapse
Affiliation(s)
- Hao Liu
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan 430022, China
| | - Peiwen Yang
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan 430022, China
| | - Shu Chen
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan 430022, China
| | - Shilin Wang
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan 430022, China
| | - Lang Jiang
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan 430022, China
| | - Xiaoyue Xiao
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan 430022, China
| | - Sheng Le
- Department of Thoracic Surgery, Zhongnan Hospital, Wuhan University, Wuhan, China
| | - Shanshan Chen
- Key Laboratory for Molecular Diagnosis of Hubei Province, Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Central Laboratory, Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xinzhong Chen
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan 430022, China
| | - Ping Ye
- Department of Cardiology, Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, ShengLi Street 26, Wuhan 430014, China
| | - Jiahong Xia
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan 430022, China
| |
Collapse
|
9
|
Yan H, Hu Y, Lyu Y, Akk A, Hirbe AC, Wickline SA, Pan H, Roberson EDO, Pham CTN. Systemic delivery of murine SOD2 mRNA to experimental abdominal aortic aneurysm mitigates expansion and rupture. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.17.599454. [PMID: 38948794 PMCID: PMC11212962 DOI: 10.1101/2024.06.17.599454] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/02/2024]
Abstract
Background Oxidative stress is implicated in the pathogenesis and progression of abdominal aortic aneurysm (AAA). Antioxidant delivery as a therapeutic for AAA is of substantial interest although clinical translation of antioxidant therapy has met with significant challenges due to limitations in achieving sufficient antioxidant levels at the site of AAA. We posit that nanoparticle-based approaches hold promise to overcome challenges associated with systemic administration of antioxidants. Methods We employed a peptide-based nanoplatform to overexpress a key modulator of oxidative stress, superoxide dismutase 2 (SOD2). The efficacy of systemic delivery of SOD2 mRNA as a nanotherapeutic agent was studied in two different murine AAA models. Unbiased mass spectrometry-enabled proteomics and high-dimensional bioinformatics were used to examine pathways modulated by SOD2 overexpression. Results The murine SOD2 mRNA sequence was mixed with p5RHH, an amphipathic peptide capable of delivering nucleic acids in vivo to form self-assembled nanoparticles of ∼55 nm in diameter. We further demonstrated that the nanoparticle was stable and functional up to four weeks following self-assembly when coated with hyaluronic acid. Delivery of SOD2 mRNA mitigated the expansion of small AAA and largely prevented rupture. Mitigation of AAA was accompanied by enhanced SOD2 protein expression in aortic wall tissue. Concomitant suppression of nitric oxide, inducible nitric oxide synthase expression, and cell death was observed. Proteomic profiling of AAA tissues suggests that SOD2 overexpression augments levels of microRNAs that regulate vascular inflammation and cell apoptosis, inhibits platelet activation/aggregation, and downregulates mitogen-activated protein kinase signaling. Gene set enrichment analysis shows that SOD2 mRNA delivery is associated with activation of oxidative phosphorylation, lipid metabolism, respiratory electron transportation, and tricarboxylic acid cycle pathways. Conclusions These results confirm that SOD2 is key modulator of oxidative stress in AAA. This nanotherapeutic mRNA delivery approach may find translational application in the medical management of small AAA and the prevention of AAA rupture.
Collapse
|
10
|
Wang A, Zhou L. Construction of ferroptosis-related prediction model for pathogenesis, diagnosis and treatment of ruptured abdominal aortic aneurysm. Medicine (Baltimore) 2024; 103:e38134. [PMID: 38728466 PMCID: PMC11081628 DOI: 10.1097/md.0000000000038134] [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: 12/18/2023] [Accepted: 04/12/2024] [Indexed: 05/12/2024] Open
Abstract
Abdominal aortic aneurysm (AAA) is a dangerous cardiovascular disease, which often brings great psychological burden and economic pressure to patients. If AAA rupture occurs, it is a serious threat to patients' lives. Therefore, it is of clinical value to actively explore the pathogenesis of ruptured AAA and prevent its occurrence. Ferroptosis is a new type of cell death dependent on lipid peroxidation, which plays an important role in many cardiovascular diseases. In this study, we used online data and analysis of ferroptosis-related genes to uncover the formation of ruptured AAA and potential therapeutic targets. We obtained ferroptosis-related differentially expressed genes (Fe-DEGs) from GSE98278 dataset and 259 known ferroptosis-related genes from FerrDb website. Enrichment analysis of differentially expressed genes (DEGs) was performed by gene ontology (GO) and Kyoto encyclopedia of genes and genomes (KEGG). Receiver Operating characteristic (ROC) curve was employed to evaluate the diagnostic abilities of Fe-DEGs. Transcription factors and miRNAs of Fe-DEGs were identified through PASTAA and miRDB, miRWalk, TargetScan respectively. Single-sample gene set enrichment analysis (ssGSEA) was used to observe immune infiltration between the stable group and the rupture group. DGIdb database was performed to find potential targeted drugs of DEGs. GO and KEGG enrichment analysis found that DEGs mainly enriched in "cellular divalent inorganic cation homeostasis," "cellular zinc ion homeostasis," "divalent inorganic cation homeostasis," "Mineral absorption," "Cytokine - cytokine receptor interaction," "Coronavirus disease - COVID-19." Two up-regulated Fe-DEGs MT1G and DDIT4 were found to further analysis. Both single and combined applications of MT1G and DDIT4 showed good diagnostic efficacy (AUC = 0.8254, 0.8548, 0.8577, respectively). Transcription factors STAT1 and PU1 of MT1G and ARNT and MAX of DDIT4 were identified. Meanwhile, has_miR-548p-MT1G pairs, has_miR-53-3p/has_miR-181b-5p/ has_miR-664a-3p-DDIT4 pairs were found. B cells, NK cells, Th2 cells were high expression in the rupture group compared with the stable group, while DCs, Th1 cells were low expression in the rupture group. Targeted drugs against immunity, GEMCITABINE and INDOMETHACIN were discovered. We preliminarily explored the clinical significance of Fe-DEGs MT1G and DDIT4 in the diagnosis of ruptured AAA, and proposed possible upstream regulatory transcription factors and miRNAs. In addition, we also analyzed the immune infiltration of stable and rupture groups, and found possible targeted drugs for immunotherapy.
Collapse
Affiliation(s)
- Ailu Wang
- Department of Neonatology, the First Hospital of China Medical University, Shenyang, China
| | - Li Zhou
- Department of Geratology, the First Hospital of China Medical University, Shenyang, China
| |
Collapse
|
11
|
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
|
12
|
Li J, Liu Y, Wei Z, Cheng J, Wu Y. The occurrence and development of abdominal aortic aneurysm may be related to the energy metabolism disorder and local inflammation. Heliyon 2024; 10:e27912. [PMID: 38496900 PMCID: PMC10944252 DOI: 10.1016/j.heliyon.2024.e27912] [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: 10/17/2023] [Revised: 03/01/2024] [Accepted: 03/08/2024] [Indexed: 03/19/2024] Open
Abstract
Background The cellular mechanism of the formation of abdominal aortic aneurysm (AAA) is very complicated. A series of sophisticated events eventually led to significant pathological changes in the anatomical structure and function of the arterial wall and they are still not clear nowadays. Methods We pooled publicly available GEO datasets (GSE57691 and GSE47472) to get a comprehensive comparisons between normal tissues and AAA tissues to try to reveal molecular mechanism underlying the disease. Total 63 AAA samples and 18 normal tissue samples were compared and we fond that there were 784 significantly different gene (DEGs, threshold set as adjusted P < 0.05 and Log FC < 1) were identified. At the same time, we validate the possible signaling factor expression of AAA by comparing the normal tissue of the human body with the AAA tissue. Results In the pathway enrichment, we found that FOXP3 related signaling pathways, inflammation-related cytokine signaling pathways, interleukin-8-CXCR1 related signaling pathways and VEGFA and FGFR1 related signal pathway were significantly enrichmented. In Weighted gene co-expression network analysis (WGCNA), we found that the key hub genes were significantly related to lipid catabolic metabolism, which further verified the possibility that AAA might relate to energy metabolism disorders. Conclusion Based on the comprehensive analysis of previous high-throughput data and the validation of basic experiments, we found that the occurrence of AAA may be related to energy metabolism disorders and local inflammation.
Collapse
Affiliation(s)
- Jun Li
- Department of Endovascular and Vascular Surgery, The Affiliated Hospital of Changchun University of Chinese Medicine, Changchun, China
| | - Yang Liu
- Department of Urology, The Affiliated Hospital of Changchun University of Chinese Medicine, Changchun, China
| | - Zhitao Wei
- Department of Urology, The Affiliated Hospital of Changchun University of Chinese Medicine, Changchun, China
| | - Jie Cheng
- Department of Interventional and Vascular Surgery, Tenth People's Hospital of Tongji University, Shanghai, China
| | - Yongfa Wu
- Department of Interventional and Vascular Surgery, Tenth People's Hospital of Tongji University, Shanghai, China
| |
Collapse
|
13
|
Basnet TB, Khatri B. Oxidative stress-related genetic variation and antioxidant vitamin intake in intact and ruptured abdominal aortic aneurysm: does sex matter? Eur J Prev Cardiol 2024; 31:59-60. [PMID: 37930805 DOI: 10.1093/eurjpc/zwad342] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Accepted: 10/30/2023] [Indexed: 11/07/2023]
Affiliation(s)
- Til Bahadur Basnet
- Division of Epidemiology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
- VA, Tennessee Valley Healthcare System (626), Nashville, TN, USA
| | - Bharat Khatri
- Department of Internal Medicine, B.P. Koirala Institute of Health Sciences, Dharan, Nepal
| |
Collapse
|
14
|
Du P, Hou Y, Su C, Gao J, Yang Y, Zhang J, Cui X, Tang J. The future for the therapeutics of abdominal aortic aneurysm: engineered nanoparticles drug delivery for abdominal aortic aneurysm. Front Bioeng Biotechnol 2024; 11:1324406. [PMID: 38249799 PMCID: PMC10796665 DOI: 10.3389/fbioe.2023.1324406] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Accepted: 12/12/2023] [Indexed: 01/23/2024] Open
Abstract
Abdominal aortic aneurysm (AAA) is a severe cardiovascular disease with a high mortality rate. Several screening and diagnostic methods have been developed for AAA early diagnosis. Open surgery and endovascular aortic repair (EVAR) are clinically available for patients who meet the indications for surgery. However, for non-surgical patients, limited drugs exist to inhibit or reverse the progression of aneurysms due to the complex pathogenesis and biological structure of AAA, failing to accumulate precisely on the lesion to achieve sufficient concentrations. The recently developed nanotechnology offers a new strategy to address this problem by developing drug-carrying nanoparticles with enhanced water solubility and targeting capacity, prolonged duration, and reduced side effects. Despite the rising popularity, limited literature is available to highlight the progression of the field. Herein, in this review, we first discuss the pathogenesis of AAA, the methods of diagnosis and treatment that have been applied clinically, followed by the review of research progressions of constructing different drug-loaded nanoparticles for AAA treatment using engineered nanoparticles. In addition, the feasibility of extracellular vesicles (EVs) and EVs-based nanotechnology for AAA treatment in recent years are highlighted, together with the future perspective. We hope this review will provide a clear picture for the scientists and clinicians to find a new solution for AAA clinical management.
Collapse
Affiliation(s)
- Pengchong Du
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Province Key Laboratory of Cardiac Injury and Repair, Zhengzhou, China
- Henan Province Clinical Research Center for Cardiovascular Diseases, Zhengzhou, China
| | - Yachen Hou
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Province Key Laboratory of Cardiac Injury and Repair, Zhengzhou, China
- Henan Province Clinical Research Center for Cardiovascular Diseases, Zhengzhou, China
| | - Chang Su
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Province Key Laboratory of Cardiac Injury and Repair, Zhengzhou, China
- Henan Province Clinical Research Center for Cardiovascular Diseases, Zhengzhou, China
| | - Jiamin Gao
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Province Key Laboratory of Cardiac Injury and Repair, Zhengzhou, China
- Henan Province Clinical Research Center for Cardiovascular Diseases, Zhengzhou, China
| | - Yu Yang
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Province Key Laboratory of Cardiac Injury and Repair, Zhengzhou, China
- Henan Province Clinical Research Center for Cardiovascular Diseases, Zhengzhou, China
| | - Jinying Zhang
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Province Key Laboratory of Cardiac Injury and Repair, Zhengzhou, China
- Henan Province Clinical Research Center for Cardiovascular Diseases, Zhengzhou, China
| | - Xiaolin Cui
- School of Medicine, The Chinese University of Hong Kong, Shenzhen, China
| | - Junnan Tang
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Province Key Laboratory of Cardiac Injury and Repair, Zhengzhou, China
- Henan Province Clinical Research Center for Cardiovascular Diseases, Zhengzhou, China
| |
Collapse
|
15
|
Pan W, Zhang J, Zhang L, Zhang Y, Song Y, Han L, Tan M, Yin Y, Yang T, Jiang T, Li H. Comprehensive view of macrophage autophagy and its application in cardiovascular diseases. Cell Prolif 2024; 57:e13525. [PMID: 37434325 PMCID: PMC10771119 DOI: 10.1111/cpr.13525] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Revised: 06/17/2023] [Accepted: 06/22/2023] [Indexed: 07/13/2023] Open
Abstract
Cardiovascular diseases (CVDs) are the primary drivers of the growing public health epidemic and the leading cause of premature mortality and economic burden worldwide. With decades of research, CVDs have been proven to be associated with the dysregulation of the inflammatory response, with macrophages playing imperative roles in influencing the prognosis of CVDs. Autophagy is a conserved pathway that maintains cellular functions. Emerging evidence has revealed an intrinsic connection between autophagy and macrophage functions. This review focuses on the role and underlying mechanisms of autophagy-mediated regulation of macrophage plasticity in polarization, inflammasome activation, cytokine secretion, metabolism, phagocytosis, and the number of macrophages. In addition, autophagy has been shown to connect macrophages and heart cells. It is attributed to specific substrate degradation or signalling pathway activation by autophagy-related proteins. Referring to the latest reports, applications targeting macrophage autophagy have been discussed in CVDs, such as atherosclerosis, myocardial infarction, heart failure, and myocarditis. This review describes a novel approach for future CVD therapies.
Collapse
Affiliation(s)
- Wanqian Pan
- Department of CardiologyThe First Affiliated Hospital of Soochow UniversitySuzhouChina
| | - Jun Zhang
- Department of CardiologyThe First Affiliated Hospital of Soochow UniversitySuzhouChina
| | - Lei Zhang
- Department of CardiologyThe First Affiliated Hospital of Soochow UniversitySuzhouChina
| | - Yue Zhang
- Department of CardiologyThe First Affiliated Hospital of Soochow UniversitySuzhouChina
| | - Yiyi Song
- Suzhou Medical College of Soochow UniversitySuzhouChina
| | - Lianhua Han
- Department of CardiologyThe First Affiliated Hospital of Soochow UniversitySuzhouChina
| | - Mingyue Tan
- Department of CardiologyThe First Affiliated Hospital of Soochow UniversitySuzhouChina
| | - Yunfei Yin
- Department of CardiologyThe First Affiliated Hospital of Soochow UniversitySuzhouChina
| | - Tianke Yang
- Department of Ophthalmology, Eye Institute, Eye & ENT HospitalFudan UniversityShanghaiChina
- Department of OphthalmologyThe First Affiliated Hospital of USTC, University of Science and Technology of ChinaHefeiChina
| | - Tingbo Jiang
- Department of CardiologyThe First Affiliated Hospital of Soochow UniversitySuzhouChina
| | - Hongxia Li
- Department of CardiologyThe First Affiliated Hospital of Soochow UniversitySuzhouChina
| |
Collapse
|
16
|
Jadli A, Gomes K, Ballasy N, Wijesuriya T, Belke D, Fedak P, Patel V. Inhibition of smooth muscle cell death by Angiotensin 1-7 protects against abdominal aortic aneurysm. Biosci Rep 2023; 43:BSR20230718. [PMID: 37947205 PMCID: PMC10695742 DOI: 10.1042/bsr20230718] [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: 04/29/2023] [Revised: 10/31/2023] [Accepted: 11/02/2023] [Indexed: 11/12/2023] Open
Abstract
Abdominal aortic aneurysm (AAA) represents a debilitating vascular disease characterized by aortic dilatation and wall rupture if it remains untreated. We aimed to determine the effects of Ang 1-7 in a murine model of AAA and to investigate the molecular mechanisms involved. Eight- to 10-week-old apolipoprotein E-deficient mice (ApoEKO) were infused with Ang II (1.44 mg/kg/day, s.c.) and treated with Ang 1-7 (0.576 mg/kg/day, i.p.). Echocardiographic and histological analyses showed abdominal aortic dilatation and extracellular matrix remodeling in Ang II-infused mice. Treatment with Ang 1-7 led to suppression of Ang II-induced aortic dilatation in the abdominal aorta. The immunofluorescence imaging exhibited reduced smooth muscle cell (SMC) density in the abdominal aorta. The abdominal aortic SMCs from ApoEKO mice exhibited markedly increased apoptosis in response to Ang II. Ang 1-7 attenuated cell death, as evident by increased SMC density in the aorta and reduced annexin V/propidium iodide-positive cells in flow cytometric analysis. Gene expression analysis for contractile and synthetic phenotypes of abdominal SMCs showed preservation of contractile phenotype by Ang 1-7 treatment. Molecular analyses identified increased mitochondrial fission, elevated cellular and mitochondrial reactive oxygen species (ROS) levels, and apoptosis-associated proteins, including cytochrome c, in Ang II-treated aortic SMCs. Ang 1-7 mitigated Ang II-induced mitochondrial fission, ROS generation, and levels of pro-apoptotic proteins, resulting in decreased cell death of aortic SMCs. These results highlight a critical vasculo-protective role of Ang 1-7 in a degenerative aortic disease; increased Ang 1-7 activity may provide a promising therapeutic strategy against the progression of AAA.
Collapse
Affiliation(s)
- Anshul S. Jadli
- Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
- Libin Cardiovascular Institute, University of Calgary, Calgary, AB, Canada
| | - Karina P. Gomes
- Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
- Libin Cardiovascular Institute, University of Calgary, Calgary, AB, Canada
| | - Noura N. Ballasy
- Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
- Libin Cardiovascular Institute, University of Calgary, Calgary, AB, Canada
| | - Tishani Methsala Wijesuriya
- Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
- Libin Cardiovascular Institute, University of Calgary, Calgary, AB, Canada
| | - Darrell Belke
- Libin Cardiovascular Institute, University of Calgary, Calgary, AB, Canada
- Section of Cardiac Surgery, Department of Cardiac Sciences, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Paul W.M. Fedak
- Libin Cardiovascular Institute, University of Calgary, Calgary, AB, Canada
- Section of Cardiac Surgery, Department of Cardiac Sciences, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Vaibhav B. Patel
- Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
- Libin Cardiovascular Institute, University of Calgary, Calgary, AB, Canada
| |
Collapse
|
17
|
Sulistyowati E, Huang SE, Cheng TL, Chao YY, Li CY, Chang CW, Lin MX, Lin MC, Yeh JL. Vasculoprotective Potential of Baicalein in Angiotensin II-Infused Abdominal Aortic Aneurysms through Inhibiting Inflammation and Oxidative Stress. Int J Mol Sci 2023; 24:16004. [PMID: 37958985 PMCID: PMC10647516 DOI: 10.3390/ijms242116004] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Revised: 10/26/2023] [Accepted: 11/03/2023] [Indexed: 11/15/2023] Open
Abstract
Aortic wall inflammation, abnormal oxidative stress and progressive degradation of extracellular matrix proteins are the main characteristics of abdominal aortic aneurysms (AAAs). The nucleotide-binding oligomerization domain-like receptor family pyrin domain containing 3 (NLRP3) inflammasome dysregulation plays a crucial role in aortic damage and disease progression. The first aim of this study was to examine the effect of baicalein (5,6,7-trihydroxy-2-phenyl-4H-1-benzopyran-4-one) on AAA formation in apolipoprotein E-deficient (ApoE-/-) mice. The second aim was to define whether baicalein attenuates aberrant vascular smooth muscle cell (VSMC) proliferation and inflammation in VSMC culture. For male ApoE-/- mice, a clinically relevant AAA model was randomly divided into four groups: saline infusion, baicalein intraperitoneal injection, Angiotensin II (Ang II) infusion and Ang II + baicalein. Twenty-seven days of treatment with baicalein markedly decreased Ang II-infused AAA incidence and aortic diameter, reduced collagen-fiber formation, preserved elastic structure and density and prevented smooth muscle cell contractile protein degradation. Baicalein inhibited rat VSMC proliferation and migration following the stimulation of VSMC cultures with Ang II while blocking the Ang II-inducible cell cycle progression from G0/G1 to the S phase in the synchronized cells. Cal-520 AM staining showed that baicalein decreased cellular calcium in Ang II-induced VSMCs; furthermore, a Western blot assay indicated that baicalein inhibited the expression of PCNA and significantly lowered levels of phospho-Akt and phospho-ERK, along with an increase in baicalein concentration in Ang II-induced VSMCs. Immunofluorescence staining showed that baicalein pretreatment reduced NF-κB nuclear translocation in Ang II-induced VSMCs and furthered the protein expressions of NLRP3 while ASC and caspase-1 were suppressed in a dose-dependent manner. Baicalein pretreatment upregulated Nrf2/HO-1 signaling in Ang II-induced VSMCs. Thus, 2',7'-dichlorodihydrofluorescein diacetate (DCFH-DA) staining showed that its reactive oxygen species (ROS) production decreased, along with the baicalein pretreatment. Our overall results indicate that baicalein could have therapeutic potential in preventing aneurysm development.
Collapse
Affiliation(s)
- Erna Sulistyowati
- Faculty of Medicine, University of Islam Malang, Malang City 65145, Indonesia;
| | - Shang-En Huang
- Department of Pharmacology, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan; (S.-E.H.); (C.-W.C.); (M.-X.L.)
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan;
| | - Tsung-Lin Cheng
- Department of Physiology, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan;
- Regenerative Medicine and Cell Therapy Research Center, Kaohsiung Medical University, Kaohsiung 807, Taiwan
- College of Professional Studies, National Pingtung University of Science and Technology, Pingtung 912, Taiwan
| | - Yu-Ying Chao
- Department of Public Health, College of Health Sciences, Kaohsiung Medical University, Kaohsiung 807, Taiwan;
| | - Chia-Yang Li
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan;
| | - Ching-Wen Chang
- Department of Pharmacology, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan; (S.-E.H.); (C.-W.C.); (M.-X.L.)
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan;
| | - Meng-Xuan Lin
- Department of Pharmacology, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan; (S.-E.H.); (C.-W.C.); (M.-X.L.)
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan;
| | - Ming-Chung Lin
- Department of Anesthesiology, Chi Mei Medical Center, Tainan 710, Taiwan
- Department of Medical Laboratory Science and Biotechnology, Chung Hwa University of Medical Technology, Tainan 717, Taiwan
| | - Jwu-Lai Yeh
- Department of Pharmacology, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan; (S.-E.H.); (C.-W.C.); (M.-X.L.)
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan;
- Department of Medical Research, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan
- Department of Marine Biotechnology and Resources, National Sun Yat-sen University, Kaohsiung 804, Taiwan
| |
Collapse
|
18
|
Hasan M, Al-Thani H, El-Menyar A, Zeidan A, Al-Thani A, Yalcin HC. Disturbed hemodynamics and oxidative stress interaction in endothelial dysfunction and AAA progression: Focus on Nrf2 pathway. Int J Cardiol 2023; 389:131238. [PMID: 37536420 DOI: 10.1016/j.ijcard.2023.131238] [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: 02/03/2023] [Revised: 04/30/2023] [Accepted: 07/31/2023] [Indexed: 08/05/2023]
Abstract
Hemodynamic shear stress is one of the major factors that are involved in the pathogenesis of many cardiovascular diseases including atherosclerosis and abdominal aortic aneurysm (AAA), through its modulatory effect on the endothelial cell's redox homeostasis and mechanosensitive gene expression. Among important mechanisms, oxidative stress, endoplasmic reticulum stress activation, and the subsequent endothelial dysfunction are attributed to disturbed blood flow and low shear stress in the vascular curvature and bifurcations which are considered atheroprone regions and aneurysm occurrence spots. Many pathways were shown to be involved in AAA progression. Of particular interest from recent findings is, the (Nrf2)/Keap-1 pathway, where Nrf2 is a transcription factor that has antioxidant properties and is strongly associated with several CVDs, yet, the exact mechanism by which Nrf2 alleviates CVDs still to be elucidated. Nrf2 expression is closely affected by shear stress and was shown to participate in AAA. In the current review paper, we discussed the link between disturbed hemodynamics and its effect on Nrf2 as a mechanosensitive gene and its role in the development of endothelial dysfunction which is linked to the progression of AAA.
Collapse
Affiliation(s)
- Maram Hasan
- Biomedical Research Center, Qatar University, P.O. Box 2713, Doha, Qatar
| | - Hassan Al-Thani
- Department of Surgery, Trauma and Vascular Surgery, Hamad General Hospital, Hamad Medical Corporation, P.O. Box 3050, Doha, Qatar
| | - Ayman El-Menyar
- Department of Surgery, Trauma and Vascular Surgery, Hamad General Hospital, Hamad Medical Corporation, P.O. Box 3050, Doha, Qatar; Clinical Medicine, Weill Cornell Medical College, Doha, Qatar
| | - Asad Zeidan
- Department of Basic Sciences, College of Medicine, QU health, Qatar University, P.O. Box 2713, Doha, Qatar
| | - Asmaa Al-Thani
- Biomedical Research Center, Qatar University, P.O. Box 2713, Doha, Qatar; Department of Biomedical Science, College of Health Sciences, QU Health, Qatar University, P.O. Box 2713, Doha, Qatar
| | - Huseyin C Yalcin
- Biomedical Research Center, Qatar University, P.O. Box 2713, Doha, Qatar.
| |
Collapse
|
19
|
Hadzikadunic H, Sjælland TB, Lindholt JS, Steffensen LB, Beck HC, Kavaliunaite E, Rasmussen LM, Stubbe J. Nicotine Administration Augments Abdominal Aortic Aneurysm Progression in Rats. Biomedicines 2023; 11:biomedicines11051417. [PMID: 37239088 DOI: 10.3390/biomedicines11051417] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Revised: 04/30/2023] [Accepted: 05/05/2023] [Indexed: 05/28/2023] Open
Abstract
Inflammation and elastin degradation are key hallmarks in the pathogenesis of abdominal aortic aneurysms (AAAs). It has been acknowledged that activation of alpha7 nicotinic acetylcholine receptors (α7nAChRs) attenuates inflammation, termed the cholinergic anti-inflammatory pathway (CAP). Thus, we hypothesize that low-dose nicotine impairs the progression of elastase-induced AAAs in rats by exerting anti-inflammatory and anti-oxidative stress properties. Male Sprague-Dawley rats underwent surgical AAA induction with intraluminal elastase infusion. We compared vehicle rats with rats treated with nicotine (1.25 mg/kg/day), and aneurysm progression was monitored by weekly ultrasound images for 28 days. Nicotine treatment significantly promoted AAA progression (p = 0.031). Additionally, gelatin zymography demonstrated that nicotine significantly reduced pro-matrix metalloproteinase (pro-MMP) 2 (p = 0.029) and MMP9 (p = 0.030) activity in aneurysmal tissue. No significant difference was found in the elastin content or the score of elastin degradation between the groups. Neither infiltrating neutrophils nor macrophages, nor aneurysmal messenger RNA (mRNA) levels of pro- or anti-inflammatory cytokines, differed between the vehicle and nicotine groups. Finally, no difference in mRNA levels of markers for anti-oxidative stress or the vascular smooth muscle cells' contractile phenotype was observed. However, proteomics analyses of non-aneurysmal abdominal aortas revealed that nicotine decreased myristoylated alanine-rich C-kinase substrate and proteins, in ontology terms, inflammatory response and reactive oxygen species, and in contradiction to augmented AAAs. In conclusion, nicotine at a dose of 1.25 mg/kg/day augments AAA expansion in this elastase AAA model. These results do not support the use of low-dose nicotine administration for the prevention of AAA progression.
Collapse
Affiliation(s)
- Hana Hadzikadunic
- Elitary Research Centre of Individualized Treatment for Arterial Disease (CIMA), Odense University Hospital, University of Southern Denmark, 5000 Odense, Denmark
- Cardiovascular and Renal Research Unit, Institute for Molecular Medicine, University of Southern Denmark, 5000 Odense, Denmark
| | - Tea Bøvling Sjælland
- Elitary Research Centre of Individualized Treatment for Arterial Disease (CIMA), Odense University Hospital, University of Southern Denmark, 5000 Odense, Denmark
- Cardiovascular and Renal Research Unit, Institute for Molecular Medicine, University of Southern Denmark, 5000 Odense, Denmark
| | - Jes S Lindholt
- Elitary Research Centre of Individualized Treatment for Arterial Disease (CIMA), Odense University Hospital, University of Southern Denmark, 5000 Odense, Denmark
- Department of Cardiothoracic and Vascular Surgery, Odense University Hospital, 5000 Odense, Denmark
| | - Lasse Bach Steffensen
- Elitary Research Centre of Individualized Treatment for Arterial Disease (CIMA), Odense University Hospital, University of Southern Denmark, 5000 Odense, Denmark
- Cardiovascular and Renal Research Unit, Institute for Molecular Medicine, University of Southern Denmark, 5000 Odense, Denmark
| | - Hans Christian Beck
- Elitary Research Centre of Individualized Treatment for Arterial Disease (CIMA), Odense University Hospital, University of Southern Denmark, 5000 Odense, Denmark
- Department of Clinical Biochemistry, Odense University Hospital, 5000 Odense, Denmark
| | - Egle Kavaliunaite
- Elitary Research Centre of Individualized Treatment for Arterial Disease (CIMA), Odense University Hospital, University of Southern Denmark, 5000 Odense, Denmark
- Cardiovascular and Renal Research Unit, Institute for Molecular Medicine, University of Southern Denmark, 5000 Odense, Denmark
- Department of Cardiothoracic and Vascular Surgery, Odense University Hospital, 5000 Odense, Denmark
| | - Lars Melholt Rasmussen
- Elitary Research Centre of Individualized Treatment for Arterial Disease (CIMA), Odense University Hospital, University of Southern Denmark, 5000 Odense, Denmark
- Department of Clinical Biochemistry, Odense University Hospital, 5000 Odense, Denmark
| | - Jane Stubbe
- Elitary Research Centre of Individualized Treatment for Arterial Disease (CIMA), Odense University Hospital, University of Southern Denmark, 5000 Odense, Denmark
- Cardiovascular and Renal Research Unit, Institute for Molecular Medicine, University of Southern Denmark, 5000 Odense, Denmark
| |
Collapse
|
20
|
Mitochondrial Dysfunction and Increased DNA Damage in Vascular Smooth Muscle Cells of Abdominal Aortic Aneurysm (AAA-SMC). OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2023; 2023:6237960. [PMID: 36743698 PMCID: PMC9891816 DOI: 10.1155/2023/6237960] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Revised: 09/30/2022] [Accepted: 11/24/2022] [Indexed: 01/27/2023]
Abstract
There is increasing evidence for enhanced oxidative stress in the vascular wall of abdominal aortic aneurysms (AAA). Mitochondrial damage and dysfunction are hypothesized to be actors in altered production of reactive oxygen species (ROS) and oxidative stress. However, the role of mitochondria and oxidative stress in vascular remodelling and progression of AAA remains uncertain. We here addressed whether mitochondrial dysfunction is persistently increased in vascular smooth muscle cells (VSMCs) isolated from AAA compared to healthy VSMC. AAA-derived VSMC cultures (AAA-SMC, n = 10) and normal VSMC cultures derived from healthy donors (n = 7) were grown in vitro and analysed for four parameters, indicating mitochondrial dysfunction: (i) mitochondrial content and morphology, (ii) ROS production and antioxidative response, (iii) NADP+/NADPH content and ratio, and (iv) DNA damage, in the presence or absence of angiotensin II (AngII). AAA-SMC displayed increased mitochondrial circularity (rounded shape), reduced mitochondrial area, and reduced perimeter, indicating increased fragmentation and dysfunction compared to healthy controls. This was accompanied by significantly increased O2 - production, reduced NADP+/NADPH levels, a lower antioxidative response (indicated by antioxidative response element- (ARE-) driven luciferase reporter assays), more DNA damage (determined by percentage of γ-H2A.X-positive nuclei), and earlier growth arrest in AAA-SMC. Our data suggest that mitochondrial dysfunction and oxidative stress are persistently increased in AAA-SMC, emphasizing their implication in the pathophysiology of AAA.
Collapse
|
21
|
Wang S, Wang J, Cai D, Li X, Zhong L, He X, Lin Z, Lai Y, Zheng H, Zhou Y, Xiao Z, Liao W, Liao Y, Xiu J, Bin J. Reactive oxygen species-induced long intergenic noncoding RNA p21 accelerates abdominal aortic aneurysm formation by promoting secretary smooth muscle cell phenotypes. J Mol Cell Cardiol 2023; 174:63-76. [PMID: 36436251 DOI: 10.1016/j.yjmcc.2022.11.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Revised: 10/28/2022] [Accepted: 11/10/2022] [Indexed: 11/25/2022]
Abstract
Whether long noncoding RNAs participate in the formation of abdominal aortic aneurysms (AAAs) through the regulation of SMC phenotypic switching is unknown. lincRNA-p21 induced by reactive oxygen species (ROS) is likely functionally associated with SMC phenotypic switching. We thus investigated the role of lincRNA-p21 in SMC phenotypic switching-associated AAA formation and its underlying mechanisms. An analysis of human and mouse abdominal aortic samples revealed that the lincRNA-p21 levels were significantly higher in AAA tissue. Stimulation with hydrogen peroxide upregulated the expression of lincRNA-p21 in a dose-dependent manner and converted SMCs from a contractile phenotype to a synthetic, proteolytic, and proinflammatory phenotype in vitro. Moreover, lincRNA-p21 promoted fracture of elastic fibres, reconstruction of the vascular wall, and AAA formation in vivo by modulating SMC phenotypic switching in two mouse models of AAA induced by angiotensin II or porcine pancreatic elastase (PPE) perfusion. Using a bioinformatics prediction method and luciferase reporter gene assays, we further proved that lincRNA-p21 sponged miR-204-5p to release the transcriptional activity of Mekk3 and promoted the NF-κB pathway and thereby played a role in the SMC phenotypic switch and AAA formation. The ROS levels were positively correlated with the lincRNA-p21 levels in human and mouse AAA tissues. The knockdown of lincRNA-p21 in a PPE-induced mouse AAA model increased the miR-204-5p levels and reduced the expression of Mekk3, whereas lincRNA-p21 overexpression had the opposite effect. Collectively, the results indicated that ROS-induced lincRNA-p21 sponges miR-204-5p to accelerate synthetic and proinflammatory SMC phenotypes through the Mekk3/NF-κB pathway in AAA formation. Thus, lincRNA-p21 may have therapeutic potential for AAA formation.
Collapse
Affiliation(s)
- Shifei Wang
- Department of Cardiology, State Key Laboratory of Organ Failure Research, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Junfen Wang
- Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Donghua Cai
- Department of Cardiology, State Key Laboratory of Organ Failure Research, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Xinzhong Li
- Department of Cardiology, State Key Laboratory of Organ Failure Research, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Lintao Zhong
- Department of Cardiology, Zhuhai People's Hospital (Zhuhai Hospital Affiliated with Jinan University), 519000 Zhuhai, China
| | - Xiang He
- Department of Cardiology, State Key Laboratory of Organ Failure Research, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Zhongqiu Lin
- Department of Cardiology, State Key Laboratory of Organ Failure Research, Nanfang Hospital, Southern Medical University, Guangzhou, China; Department of Geriatrics, General Hospital of Southern Theater Command, PLA, Guangzhou, China
| | - Yanxian Lai
- Department of Cardiology, Guangzhou First People's Hospital, South China University of Technology, Guangzhou, China
| | - Hao Zheng
- Department of Cardiology, State Key Laboratory of Organ Failure Research, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Yilin Zhou
- Department of Cardiology, State Key Laboratory of Organ Failure Research, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Zhiwen Xiao
- Department of Cardiology, State Key Laboratory of Organ Failure Research, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Wangjun Liao
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Yulin Liao
- Department of Cardiology, State Key Laboratory of Organ Failure Research, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Jiancheng Xiu
- Department of Cardiology, State Key Laboratory of Organ Failure Research, Nanfang Hospital, Southern Medical University, Guangzhou, China.
| | - Jianping Bin
- Department of Cardiology, State Key Laboratory of Organ Failure Research, Nanfang Hospital, Southern Medical University, Guangzhou, China.
| |
Collapse
|
22
|
Lin W, Hu K, Li C, Pu W, Yan X, Chen H, Hu H, Deng H, Zhang J. A Multi-Bioactive Nanomicelle-Based "One Stone for Multiple Birds" Strategy for Precision Therapy of Abdominal Aortic Aneurysms. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2022; 34:e2204455. [PMID: 36085560 DOI: 10.1002/adma.202204455] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Abdominal aortic aneurysm (AAA) remains a lethal aortic disease in the elderly. Currently, no effective drugs can be clinically applied to prevent the development of AAA. Herein, a "one stone for multiple birds" strategy for AAA therapy is reported. As a proof of concept, three bioactive conjugates are designed and synthesized, which can assemble into nanomicelles. Cellularly, these nanomicelles significantly inhibit migration and activation of inflammatory cells as well as protect vascular smooth muscle cells (VSMCs) from induced oxidative stress, calcification and apoptosis, with the best effect for nanomicelles (TPTN) derived from a conjugate defined as TPT. After intravenous delivery, TPTN efficiently accumulates in the aneurysmal tissue of AAA rats, showing notable distribution in neutrophils, macrophages and VSMCs, all relevant to AAA pathogenesis. Whereas three examined nanomicelles effectively delay expansion of AAA in rats, TPTN most potently prevents AAA growth by simultaneously normalizing the pro-inflammatory microenvironment and regulating multiple pathological cells. TPTN is effective even at 0.2 mg kg-1 . Besides, TPTN can function as a bioactive nanoplatform for site-specifically delivering and triggerably releasing anti-aneurysmal drugs, affording synergistic therapeutic effects. Consequently, TPTN is a promising multi-bioactive nanotherapy and bioresponsive targeting delivery nanocarrier for effective therapy of AAA and other inflammatory vascular diseases.
Collapse
Affiliation(s)
- Wenjie Lin
- Department of Pharmaceutics, College of Pharmacy, Third Military Medical University (Army Medical University), Chongqing, 400038, China
- Department of Vascular Surgery, Renmin Hospital of Wuhan University, Wuhan University, Wuhan, 430060, China
| | - Kaiyao Hu
- Department of Pharmaceutics, College of Pharmacy, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Chenwen Li
- Department of Pharmaceutics, College of Pharmacy, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Wendan Pu
- Department of Pharmaceutics, College of Pharmacy, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Xinhao Yan
- Department of Pharmaceutics, College of Pharmacy, Third Military Medical University (Army Medical University), Chongqing, 400038, China
- College of Pharmacy and Medical Technology, Hanzhong Vocational and Technical College, Hanzhong, Shaanxi Province, 723000, China
| | - Haiyan Chen
- Department of Pharmaceutics, College of Pharmacy, Third Military Medical University (Army Medical University), Chongqing, 400038, China
- Department of Cardiology, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Houyuan Hu
- Department of Cardiology, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Hongping Deng
- Department of Vascular Surgery, Renmin Hospital of Wuhan University, Wuhan University, Wuhan, 430060, China
| | - Jianxiang Zhang
- Department of Pharmaceutics, College of Pharmacy, Third Military Medical University (Army Medical University), Chongqing, 400038, China
- State Key Lab of Trauma, Burn and Combined Injury, Institute of Combined Injury, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| |
Collapse
|
23
|
Wang K, Song Y, Li H, Song J, Wang S. Identification of differentially expressed ferroptosis-related genes in abdominal aortic aneurysm: Bioinformatics analysis. Front Cardiovasc Med 2022; 9:991613. [PMID: 36247434 PMCID: PMC9558826 DOI: 10.3389/fcvm.2022.991613] [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: 07/11/2022] [Accepted: 09/12/2022] [Indexed: 11/13/2022] Open
Abstract
Purpose Ferroptosis plays a crucial role in the development and progression of abdominal aortic aneurysm (AAA). The aim of this study was to identify differentially expressed genes associated with ferroptosis in AAA through bioinformatics analysis combined with experimental validation. Materials and methods Firstly, the mRNA expression profile datasets GSE57691 and GSE47472 from Gene Expression Omnibus database were screened, and principal component analysis was carried out. Next, the R software (version 4.0.0) was used to analyze potentially differentially expressed genes associated with AAA and ferroptosis. Subsequently, protein–protein interaction analysis, gene ontology enrichment analysis, and Kyoto Encyclopedia of Genes and Genomes pathway enrichment analysis were performed on the selected candidate genes. Finally, quantitative real-time polymerase chain reaction (qRT-PCR) was used to detect the expression levels of the first five selected abnormal ferroptosis-related genes in clinical samples obtained from patients with AAA and healthy controls. Results Based on the information contained in the two datasets, a total of 20 differentially expressed ferroptosis-related genes (three upregulated genes and 17 downregulated genes) were selected. Protein–protein interaction analysis demonstrated interaction between these genes, while gene ontology enrichment analysis of ferroptosis genes with differential expression indicated that some enrichment items were associated with oxidative stress. The qRT-PCR results showed that the expression levels of interleukin-6 (IL-6), peroxiredoxin 1 (PRDX1), and stearoyl-CoA desaturase (SCD) were consistent with the bioinformatics prediction results obtained from the mRNA chip. Conclusion Bioinformatics analysis identified 20 potential ferroptosis-related differentially expressed genes in AAA. Further verification by qRT-PCR showed that IL-6, PRXD1, and SCD might affect the process of AAA by regulating ferroptosis. Our results might assist in further understanding the pathogenesis of AAA and guiding treatment.
Collapse
Affiliation(s)
- Kun Wang
- Department of Cardiovascular Surgery, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Yancheng Song
- Department of Gastrointestinal Surgery, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Hong Li
- Clinical Laboratory, The Affiliated Qingdao Hiser Hospital of Qingdao University, Qingdao, China
| | - Jianshu Song
- Department of Cardiovascular Surgery, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Shizhong Wang
- Department of Cardiovascular Surgery, The Affiliated Hospital of Qingdao University, Qingdao, China
- *Correspondence: Shizhong Wang,
| |
Collapse
|
24
|
Edaravone Attenuated Angiotensin II-Induced Atherosclerosis and Abdominal Aortic Aneurysms in Apolipoprotein E-Deficient Mice. Biomolecules 2022; 12:biom12081117. [PMID: 36009011 PMCID: PMC9405883 DOI: 10.3390/biom12081117] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2022] [Revised: 08/09/2022] [Accepted: 08/12/2022] [Indexed: 12/17/2022] Open
Abstract
Background: The aim of the study was to define whether edaravone, a free-radical scavenger, influenced angiotensin II (AngII)-induced atherosclerosis and abdominal aortic aneurysms (AAAs) formation. Methods: Male apolipoprotein E-deficient mice (8–12 weeks old) were fed with a normal diet for 5 weeks. Either edaravone (10 mg/kg/day) or vehicle was injected intraperitoneally for 5 weeks. After 1 week of injections, mice were infused subcutaneously with either AngII (1000 ng/kg/min, n = 16–17 per group) or saline (n = 5 per group) by osmotic minipumps for 4 weeks. Results: AngII increased systolic blood pressure equivalently in mice administered with either edaravone or saline. Edaravone had no effect on plasma total cholesterol concentrations and body weights. AngII infusion significantly increased ex vivo maximal diameters of abdominal aortas and en face atherosclerosis but was significantly attenuated by edaravone administration. Edaravone also reduced the incidence of AngII-induced AAAs. In addition, edaravone diminished AngII-induced aortic MMP-2 activation. Quantitative RT-PCR revealed that edaravone ameliorated mRNA abundance of aortic MCP-1 and IL-1β. Immunostaining demonstrated that edaravone attenuated oxidative stress and macrophage accumulation in the aorta. Furthermore, edaravone administration suppressed thioglycolate-induced mice peritoneal macrophages (MPMs) accumulation and mRNA abundance of MCP-1 in MPMs in male apolipoprotein E-deficient mice. In vitro, edaravone reduced LPS-induced mRNA abundance of MCP-1 in MPMs. Conclusions: Edaravone attenuated AngII-induced AAAs and atherosclerosis in male apolipoprotein E-deficient mice via anti-oxidative action and anti-inflammatory effect.
Collapse
|
25
|
Ren J, Lv Y, Wu L, Chen S, Lei C, Yang D, Li F, Liu C, Zheng Y. Key ferroptosis-related genes in abdominal aortic aneurysm formation and rupture as determined by combining bioinformatics techniques. Front Cardiovasc Med 2022; 9:875434. [PMID: 36017103 PMCID: PMC9395677 DOI: 10.3389/fcvm.2022.875434] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2022] [Accepted: 07/15/2022] [Indexed: 11/13/2022] Open
Abstract
Objectives Abdominal aortic aneurysm (AAA) is a cardiovascular disease with high mortality and pathogenesis closely related to various cell death types, e.g., autophagy, apoptosis and pyroptosis. However, the association between AAA and ferroptosis is unknown. Methods GSE57691 and GSE98278 dataset were obtained from the Gene Expression Omnibus database, and a ferroptosis-related gene (FRG) set was downloaded from the FerrDb database. These data were normalized, and ferroptosis-related differentially expressed genes (FDEGs, AAA vs. normal samples) were identified using the limma package in R. FRGs expression was analyzed by Gene Set Expression Analysis (GSEA), and FDEGs were analyzed by Gene Ontology (GO) and Kyoto Encyclopedia of Genes (KEGG) pathway enrichment analyses using the clusterProfiler package in R and ClueGO in Cytoscape. Protein–protein interaction networks were assembled using Cytoscape, and crucial FDEGs were identified using CytoHubba. Critical FDEG transcription factors (TFs) were predicted with iRegulon. FDEGs were verified in GSE98278 set, and key FDEGs in AAA (compared with normal samples) and ruptured AAA (RAAA; compared with AAA samples) were identified. Ferroptosis-related immune cell infiltration and correlations with key genes were analyzed by CIBERSORT. Key FEDGs were reverified in Ang II-induced AAA models of ApoE–/– and CD57B/6J mice by immunofluorescence assay. Results In AAA and normal samples, 40 FDEGs were identified, and the expression of suppressive FRGs was significantly downregulated with GSEA. For FDEGs, the GO terms were response to oxidative stress and cellular response to external stimulus, and the KEGG pathways were the TNF and NOD-like receptor signaling pathways. IL6, ALB, CAV1, PTGS2, NOX4, PRDX6, GPX4, HSPA5, HSPB1, and NCF2 were the most enriched genes in the crucial gene cluster. CEBPG, NFAT5, SOX10, GTF2IRD1, STAT1, and RELA were potential TFs affecting these crucial genes. Ferroptosis-related immune cells involved in AAA formation were CD8+ T, naive CD4+ T, and regulatory T cells (Tregs); M0 and M2 macrophages; and eosinophils. Tregs were also involved in RAAA. GPX4, SLC2A1, and PEBP1 expression was downregulated in both the RAAA and AAA samples. GPX4 and PEBP1 were more important in AAA because they influenced ferroptosis-related immune cell infiltration, and SLC2A1 was more important in RAAA. Conclusions This is the first study to show that ferroptosis is crucial to AAA/RAAA formation. The TNF and NOD-like signaling pathways and ferroptosis-related immune cell infiltration play key roles in AAA/RAAA. GPX4 is a key ferroptosis-related gene in AAA. Ferroptosis and related genes might be promising targets in the treatment of AAA/RAAA.
Collapse
Affiliation(s)
- Jinrui Ren
- Department of Vascular Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yanze Lv
- Department of Vascular Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Lianglin Wu
- Department of Vascular Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Siliang Chen
- Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Chuxiang Lei
- Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Dan Yang
- Department of Computational Biology and Bioinformatics, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Fangda Li
- Department of Vascular Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Changzheng Liu
- NHC Key Laboratory of Systems Biology of Pathogens, Institute of Pathogen Biology (IPB), Chinese Academy of Medical Sciences (CAMS) and Peking Union Medical College, Beijing, China
| | - Yuehong Zheng
- State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- State Key Laboratory of Complex Severe and Rare Disease, Department of Vascular Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- *Correspondence: Yuehong Zheng,
| |
Collapse
|
26
|
Li Z, Cong X, Kong W. Matricellular proteins: Potential biomarkers and mechanistic factors in aortic aneurysms. J Mol Cell Cardiol 2022; 169:41-56. [DOI: 10.1016/j.yjmcc.2022.05.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2021] [Revised: 03/30/2022] [Accepted: 05/03/2022] [Indexed: 10/18/2022]
|
27
|
Teng B, Xie C, Zhao Y, Zeng Q, Zhan F, Feng Y, Wang Z. Identification of MEDAG and SERPINE1 Related to Hypoxia in Abdominal Aortic Aneurysm Based on Weighted Gene Coexpression Network Analysis. Front Physiol 2022; 13:926508. [PMID: 35874515 PMCID: PMC9301186 DOI: 10.3389/fphys.2022.926508] [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: 04/22/2022] [Accepted: 06/13/2022] [Indexed: 11/13/2022] Open
Abstract
Purpose: Abdominal aortic aneurysm (AAA) is a severe cardiovascular disease that often results in high mortality due to sudden rupture. This paper aims to explore potential molecular mechanisms and effective targeted therapies to prevent and delay AAA rupture. Methods: We downloaded two microarray datasets (GSE98278 and GSE17901) from the Gene Expression Omnibus (GEO) database. Differential analysis and single-sample gene set enrichment analysis (ssGSEA) of hypoxia scores were performed on 48 AAA patients in GSE98278. We identified hypoxia- and ruptured AAA-related gene modules using weighted gene coexpression network analysis (WGCNA). Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) analyses were performed using the R package clusterProfiler. For candidate genes, validation was conducted on the mouse dataset GSE17901. Finally, we predicted drug candidates associated with the hub genes using the HERB Chinese medicine database. Results: Eighty-two differentially expressed genes were screened in the ruptured and stable groups; 103 differentially expressed genes were identified between the high- and low-hypoxia groups; and WGCNA identified 58 differentially expressed genes. Finally, nine candidate genes were screened, including two hub genes (MEDAG and SERPINE1). We identified pathways such as cytokine-cytokine receptor interaction and T-helper 1-type immune response involved in AAA hypoxia and rupture. We predicted 93 traditional Chinese medicines (TCMs) associated with MEDAG and SERPINE1. Conclusion: We identified the hypoxic molecules MEDAG and SERPINE1 associated with AAA rupture. Our study provides an additional direction for the association between hypoxia and AAA rupture.
Collapse
Affiliation(s)
- Biyun Teng
- Department of Vascular Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Chaozheng Xie
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yu Zhao
- Department of Vascular Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Qiu Zeng
- Department of Vascular Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Fangbiao Zhan
- Department of Orthopedics, Chongqing University Three Gorges Hospital, Chongqing, China
| | - Yangyang Feng
- Department of Vascular Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Zhe Wang
- Department of Vascular Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
28
|
Zhou H, Wang L, Liu S, Wang W. The role of phosphoinositide 3-kinases in immune-inflammatory responses: potential therapeutic targets for abdominal aortic aneurysm. Cell Cycle 2022; 21:2339-2364. [PMID: 35792922 DOI: 10.1080/15384101.2022.2094577] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
The pathogenesis of abdominal aortic aneurysm (AAA) includes inflammatory responses, matrix metalloproteinases (MMPs) degradation, VSMC apoptosis, oxidative stress, and angiogenesis, among which the inflammatory response plays a key role. At present, surgery is the only curing treatment, and no effective drug can delay AAA progression in clinical practice. Therefore, searching for a signaling pathway related to the immune-inflammatory response is an essential direction for developing drugs targeting AAA. Recent studies have confirmed that the PI3K family plays an important role in many inflammatory diseases and is involved in regulating various cellular functions, especially in the immune-inflammatory response. This review focuses on the role of each isoform of PI3K in each stage of AAA immune-inflammatory response, making available explorations for a deeper understanding of the mechanism of inflammation and immune response during the formation and development of AAA.
Collapse
Affiliation(s)
- Haiyang Zhou
- Department of General &vascular Surgery, Xiangya Hospital, Central South University, Changsha, China
| | - Lei Wang
- Department of General &vascular Surgery, Xiangya Hospital, Central South University, Changsha, China
| | - Shuai Liu
- Department of General &vascular Surgery, Xiangya Hospital, Central South University, Changsha, China
| | - Wei Wang
- Department of General &vascular Surgery, Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
29
|
Zhou F, Zheng Z, Zha Z, Xiong T, Pan Y. Nuclear Paraspeckle Assembly Transcript 1 Enhances Hydrogen Peroxide-Induced Human Vascular Smooth Muscle Cell Injury by Regulating miR-30d-5p/A Disintegrin and Metalloprotease 10. Circ J 2022; 86:1007-1018. [PMID: 34880199 DOI: 10.1253/circj.cj-21-0042] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
BACKGROUND Nuclear paraspeckle assembly transcript 1 (NEAT1) has been reported to be involved in the progression of many cancers; however, the role and mechanisms underlying NEAT1 in abdominal aortic aneurysm (AAA) remain unclear. METHODS AND RESULTS The expression of NEAT1, miR-30d-5p and A disintegrin and metalloprotease 10 (ADAM10) was measured by qRT-PCR and western blot. Functional experiments were conducted by using a CCK-8 assay, EDU assay, flow cytometry, western blot, ELISA, and commercial kits. The target relation was confirmed by dual-luciferase reporter assay and the RIP assay. It was then found that NEAT1 was upregulated in peripheral blood of AAA patients ~3.46-fold, smooth muscle cells (SMCs) isolated from AAA tissues ~2.6-fold and in a hydrogen peroxide (H2O2)-induced injury model of human vascular SMC (HVSMCs) ~2.0- and 3.9-fold at 50 µmol/L and 200 µmol/L H2O2treatment, respectively. NEAT1 deletion attenuated H2O2-induced cell proliferation promotion (40.0% vs. 74.3%), apoptosis inhibition (25.0% vs. 13.5%), and reduction of inflammatory response and oxidative stress in HVSMCs. Mechanistically, NEAT1 targeted miR-30d-5p to prevent the degradation of its target, ADAM10, in HVSMCs. Further rescue experiments suggested miR-30d-5p inhibition mitigated the effects of NEAT1 deletion on H2O2-induced HVSMCs. Moreover, ADAM10 overexpression counteracted the inhibitory functions of miR-30d-5p on H2O2-evoked HVSMC injury. CONCLUSIONS NEAT1 promoted H2O2-induced HVSMC injury by inducing cell apoptosis, inflammation and oxidative stress through miR-30d-5p/ADAM10 axis, indicating the possible involvement of NEAT1 in the pathogenesis of AAA.
Collapse
Affiliation(s)
- Fushuo Zhou
- Department of Cardiovascular Surgery, Tongji Hospital, Tongji Medical College of Huazhong University of Science and Technology
| | - Zhi Zheng
- Department of Cardiovascular Surgery, Tongji Hospital, Tongji Medical College of Huazhong University of Science and Technology
| | - Zhengbiao Zha
- Department of Cardiovascular Surgery, Tongji Hospital, Tongji Medical College of Huazhong University of Science and Technology
| | - Tianxin Xiong
- Department of Cardiovascular Surgery, Tongji Hospital, Tongji Medical College of Huazhong University of Science and Technology
| | - Youmin Pan
- Department of Cardiovascular Surgery, Tongji Hospital, Tongji Medical College of Huazhong University of Science and Technology
| |
Collapse
|
30
|
Wang L, Zhou S, Liu Y, Li Y, Sun X. Bibliometric analysis of the inflammatory mechanism in aortic disease. Rev Cardiovasc Med 2022; 23:67. [PMID: 35229558 DOI: 10.31083/j.rcm2302067] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Revised: 10/07/2021] [Accepted: 10/14/2021] [Indexed: 01/02/2025] Open
Abstract
BACKGROUND In view of the key role of inflammation in the pathogenesis of aortic disease, we visually analyzed the research hotspots of inflammatory mechanism in aortic disease in this work through the method of bibliometrics from the Web of Science (WOS) Core database over the past three decades. METHODS A visual bibliometric network of research articles on inflammatory mechanisms in aortic disease was obtained from VOSviewer and Citespace based on the WOS Core Collection. RESULTS A total of 1278 documents from January 1990 to February 2021 were selected for analysis. The United States and China had the highest percentage of articles, comprising 34.01% and 24.92% of articles worldwide, respectively. Harvard University has published the most articles in this field, followed by the University of Michigan and Huazhong University of Science and Technology. The top 3 research hotspots were atherosclerosis, oxidative stress, and macrophages. The journal with the most articles in this area was Arteriosclerosis Thrombosis and Vascular Biology, followed by Atherosclerosis and PLOS One. The research trend on inflammatory mechanisms in the aortic system has 5 distinct directions: (1) atherosclerosis, NF-κB, expression, smooth muscle cell, and oxidative stress; (2) coronary artery disease, C-reactive protein, risk factors, endothelial dysfunction, and aortic stenosis; (3) abdominal aortic aneurysm, matrix metalloproteinases, macrophage, and pathogenesis; (4) cholesterol, metabolism, low-density lipoprotein, gene expression, and a therosclerotic lesions; and (5) calcific aortic valve disease, interstitial cells, calcification, and stenosis. CONCLUSIONS Inflammatory mechanism research has shown a tendency to rise gradually in the aortic field. Numerous studies have explored the role of inflammatory responses in aortic disease, which may increase the risk of endothelial dysfunction (aortic fibrosis and stiffness) and induce plaque formation. Among them, NFκB activation, nitric-oxide synthase expression, and oxidative stress are particularly essential.
Collapse
Affiliation(s)
- Luchen Wang
- Aortic and Vascular Surgery Center, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, 100037 Beijing, China
| | - Sangyu Zhou
- Aortic and Vascular Surgery Center, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, 100037 Beijing, China
| | - Yanxiang Liu
- Aortic and Vascular Surgery Center, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, 100037 Beijing, China
| | - Yunfeng Li
- Aortic and Vascular Surgery Center, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, 100037 Beijing, China
- Shandong University, Qilu Hospital, 250012 Jinan, Shandong, China
| | - Xiaogang Sun
- Aortic and Vascular Surgery Center, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, 100037 Beijing, China
| |
Collapse
|
31
|
Melin LG, Dall JH, Lindholt JS, Steffensen LB, Beck HC, Elkrog SL, Clausen PD, Rasmussen LM, Stubbe J. Cycloastragenol Inhibits Experimental Abdominal Aortic Aneurysm Progression. Biomedicines 2022; 10:biomedicines10020359. [PMID: 35203568 PMCID: PMC8962318 DOI: 10.3390/biomedicines10020359] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Revised: 01/24/2022] [Accepted: 01/30/2022] [Indexed: 02/04/2023] Open
Abstract
The pathogenesis of abdominal aortic aneurysm involves vascular inflammation and elastin degradation. Astragalusradix contains cycloastragenol, which is known to be anti-inflammatory and to protect against elastin degradation. We hypothesized that cycloastragenol supplementation inhibits abdominal aortic aneurysm progression. Abdominal aortic aneurysm was induced in male rats by intraluminal elastase infusion in the infrarenal aorta and treated daily with cycloastragenol (125 mg/kg/day). Aortic expansion was followed weekly by ultrasound for 28 days. Changes in aneurysmal wall composition were analyzed by mRNA levels, histology, zymography and explorative proteomic analyses. At day 28, mean aneurysm diameter was 37% lower in the cycloastragenol group (p < 0.0001). In aneurysm cross sections, elastin content was insignificantly higher in the cycloastragenol group (10.5% ± 5.9% vs. 19.9% ± 16.8%, p = 0.20), with more preserved elastin lamellae structures (p = 0.0003) and without microcalcifications. Aneurysmal matrix metalloprotease-2 activity was reduced by the treatment (p = 0.022). Messenger RNA levels of inflammatory- and anti-oxidative markers did not differ between groups. Explorative proteomic analysis showed no difference in protein levels when adjusting for multiple testing. Among proteins displaying nominal regulation were fibulin-5 (p = 0.02), aquaporin-1 (p = 0.02) and prostacyclin synthase (p = 0.007). Cycloastragenol inhibits experimental abdominal aortic aneurysm progression. The suggested underlying mechanisms involve decreased matrix metalloprotease-2 activity and preservation of elastin and reduced calcification, thus, cycloastragenol could be considered for trial in abdominal aortic aneurysm patients.
Collapse
Affiliation(s)
- Leander Gaarde Melin
- Centre for Individualized Medicine in Arterial Diseases (CIMA), Odense University Hospital (OUH), 5000 Odense, Denmark; (L.G.M.); (J.H.D.); (J.S.L.); (H.C.B.); (L.M.R.)
- Department of Cardiothoracic and Vascular Surgery, Odense University Hospital, 5000 Odense, Denmark
- Cardiovascular and Renal Research Unit, Institute for Molecular Medicine, University of Southern Denmark, 5000 Odense, Denmark; (L.B.S.); (S.L.E.); (P.D.C.)
| | - Julie Husted Dall
- Centre for Individualized Medicine in Arterial Diseases (CIMA), Odense University Hospital (OUH), 5000 Odense, Denmark; (L.G.M.); (J.H.D.); (J.S.L.); (H.C.B.); (L.M.R.)
- Department of Cardiothoracic and Vascular Surgery, Odense University Hospital, 5000 Odense, Denmark
- Cardiovascular and Renal Research Unit, Institute for Molecular Medicine, University of Southern Denmark, 5000 Odense, Denmark; (L.B.S.); (S.L.E.); (P.D.C.)
| | - Jes S. Lindholt
- Centre for Individualized Medicine in Arterial Diseases (CIMA), Odense University Hospital (OUH), 5000 Odense, Denmark; (L.G.M.); (J.H.D.); (J.S.L.); (H.C.B.); (L.M.R.)
- Department of Cardiothoracic and Vascular Surgery, Odense University Hospital, 5000 Odense, Denmark
| | - Lasse B. Steffensen
- Cardiovascular and Renal Research Unit, Institute for Molecular Medicine, University of Southern Denmark, 5000 Odense, Denmark; (L.B.S.); (S.L.E.); (P.D.C.)
| | - Hans Christian Beck
- Centre for Individualized Medicine in Arterial Diseases (CIMA), Odense University Hospital (OUH), 5000 Odense, Denmark; (L.G.M.); (J.H.D.); (J.S.L.); (H.C.B.); (L.M.R.)
- Department of Clinical Biochemistry and Pharmacology, Odense University Hospital, 5000 Odense, Denmark
| | - Sophie L. Elkrog
- Cardiovascular and Renal Research Unit, Institute for Molecular Medicine, University of Southern Denmark, 5000 Odense, Denmark; (L.B.S.); (S.L.E.); (P.D.C.)
| | - Pernille D. Clausen
- Cardiovascular and Renal Research Unit, Institute for Molecular Medicine, University of Southern Denmark, 5000 Odense, Denmark; (L.B.S.); (S.L.E.); (P.D.C.)
| | - Lars Melholt Rasmussen
- Centre for Individualized Medicine in Arterial Diseases (CIMA), Odense University Hospital (OUH), 5000 Odense, Denmark; (L.G.M.); (J.H.D.); (J.S.L.); (H.C.B.); (L.M.R.)
- Department of Clinical Biochemistry and Pharmacology, Odense University Hospital, 5000 Odense, Denmark
| | - Jane Stubbe
- Centre for Individualized Medicine in Arterial Diseases (CIMA), Odense University Hospital (OUH), 5000 Odense, Denmark; (L.G.M.); (J.H.D.); (J.S.L.); (H.C.B.); (L.M.R.)
- Cardiovascular and Renal Research Unit, Institute for Molecular Medicine, University of Southern Denmark, 5000 Odense, Denmark; (L.B.S.); (S.L.E.); (P.D.C.)
- Correspondence: ; Tel.: +45-6550-3709
| |
Collapse
|
32
|
Kessler V, Klopf J, Eilenberg W, Neumayer C, Brostjan C. AAA Revisited: A Comprehensive Review of Risk Factors, Management, and Hallmarks of Pathogenesis. Biomedicines 2022; 10:94. [PMID: 35052774 PMCID: PMC8773452 DOI: 10.3390/biomedicines10010094] [Citation(s) in RCA: 47] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Accepted: 12/30/2021] [Indexed: 01/27/2023] Open
Abstract
Despite declining incidence and mortality rates in many countries, the abdominal aortic aneurysm (AAA) continues to represent a life-threatening cardiovascular condition with an overall prevalence of about 2-3% in the industrialized world. While the risk of AAA development is considerably higher for men of advanced age with a history of smoking, screening programs serve to detect the often asymptomatic condition and prevent aortic rupture with an associated death rate of up to 80%. This review summarizes the current knowledge on identified risk factors, the multifactorial process of pathogenesis, as well as the latest advances in medical treatment and surgical repair to provide a perspective for AAA management.
Collapse
Affiliation(s)
| | | | | | | | - Christine Brostjan
- Department of General Surgery, Division of Vascular Surgery, Medical University of Vienna, Vienna General Hospital, 1090 Vienna, Austria; (V.K.); (J.K.); (W.E.); (C.N.)
| |
Collapse
|
33
|
Hu J, Li Y, Wei Z, Chen H, Sun X, Zhou Q, Zhang Q, Yin Y, Guo M, Chen J, Zhai G, Xu B, Xie J. A reduction in the vascular smooth muscle cell focal adhesion component syndecan-4 is associated with abdominal aortic aneurysm formation. Clin Transl Med 2021; 11:e605. [PMID: 34936241 PMCID: PMC8693440 DOI: 10.1002/ctm2.605] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2021] [Revised: 09/18/2021] [Accepted: 09/23/2021] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Abdominal aortic aneurysm (AAA) is a serious vascular disease for which there is no effective drug treatment. The incidence of AAA increases significantly as a subject ages, and the molecular mechanism of AAA formation remains elusive. In the present study, we investigated the role of syndecan-4 (SDC4), an important component of focal adhesions, in AAA formation and its association with phenotypic changes in vascular smooth muscle cells (VSMCs). METHODS AND RESULTS The protein expression levels of SDC4 were significantly decreased in human AAA tissue and those of an AAA mouse model. Moreover, SDC4 knockout (KO) in mice accelerated the formation and rupture of AAAs induced by angiotensin II (Ang II) and calcium chloride (CaCl2 ) Mechanistically, the decrease in SDC4 led to the transformation of cultured VSMCs from a contractile to a secretory phenotype. The RhoA-F/G-actin-myocardin-related transcription factor-A (MRTF-A) signalling pathway was shown to be involved in SDC4-dependent VSMC alteration. Sphingosine-1-phosphate (S1P), a G-protein-coupled receptor, attenuated the AAA formation in SDC4-KO and wild-type (WT) mice in response to Ang II and CaCl2 stimulation. CONCLUSION We herein demonstrated that silencing SDC4 was associated with increased AAA formation and phenotypic changes in VSMCs via the RhoA-F/G-actin-MRTF-A pathway. These findings indicated that a reduction in SDC4 expression was an important pathological alteration and potential therapeutic target for AAA formation.
Collapse
Affiliation(s)
- Jiaxin Hu
- Department of Cardiology, Nanjing Drum Tower HospitalThe Affiliated Hospital of Nanjing University Medical School, MOE Key Laboratory of Model Animal for Disease Study, Nanjing UniversityNanjingChina
| | - Yuyu Li
- Department of Cardiology, Nanjing Drum Tower HospitalThe Affiliated Hospital of Nanjing University Medical School, MOE Key Laboratory of Model Animal for Disease Study, Nanjing UniversityNanjingChina
| | - Zhonghai Wei
- Department of Cardiology, Nanjing Drum Tower HospitalThe Affiliated Hospital of Nanjing University Medical School, MOE Key Laboratory of Model Animal for Disease Study, Nanjing UniversityNanjingChina
| | - Haiting Chen
- Department of Cardiology, Nanjing Drum Tower HospitalThe Affiliated Hospital of Nanjing University Medical School, MOE Key Laboratory of Model Animal for Disease Study, Nanjing UniversityNanjingChina
| | - Xuan Sun
- Department of Cardiology, Nanjing Drum Tower HospitalThe Affiliated Hospital of Nanjing University Medical School, MOE Key Laboratory of Model Animal for Disease Study, Nanjing UniversityNanjingChina
| | - Qing Zhou
- Department of Cardiac Surgery, Nanjing Drum Tower HospitalThe Affiliated Hospital of Nanjing University Medical SchoolNanjing UniversityNanjingChina
| | - Qi Zhang
- Department of Cardiology, Nanjing Drum Tower HospitalThe Affiliated Hospital of Nanjing University Medical School, MOE Key Laboratory of Model Animal for Disease Study, Nanjing UniversityNanjingChina
| | - Yong Yin
- Department of Cardiology, Nanjing Drum Tower HospitalThe Affiliated Hospital of Nanjing University Medical School, MOE Key Laboratory of Model Animal for Disease Study, Nanjing UniversityNanjingChina
| | - Meng Guo
- Department of Cardiology, Nanjing Drum Tower HospitalThe Affiliated Hospital of Nanjing University Medical School, MOE Key Laboratory of Model Animal for Disease Study, Nanjing UniversityNanjingChina
| | - Jianzhou Chen
- Department of Cardiology, Nanjing Drum Tower HospitalThe Affiliated Hospital of Nanjing University Medical School, MOE Key Laboratory of Model Animal for Disease Study, Nanjing UniversityNanjingChina
| | - Guangyao Zhai
- Department of Cardiology, Beijing Anzhen HospitalCapital Medical UniversityBeijingChina
| | - Biao Xu
- Department of Cardiology, Nanjing Drum Tower HospitalThe Affiliated Hospital of Nanjing University Medical School, MOE Key Laboratory of Model Animal for Disease Study, Nanjing UniversityNanjingChina
| | - Jun Xie
- Department of Cardiology, Nanjing Drum Tower HospitalThe Affiliated Hospital of Nanjing University Medical School, MOE Key Laboratory of Model Animal for Disease Study, Nanjing UniversityNanjingChina
| |
Collapse
|
34
|
Kugo H, Sukketsiri W, Iwamoto K, Suihara S, Moriyama T, Zaima N. Low glucose and serum levels cause an increased inflammatory factor in 3T3-L1 cell through Akt, MAPKs and NF-кB activation. Adipocyte 2021; 10:232-241. [PMID: 33896390 PMCID: PMC8078669 DOI: 10.1080/21623945.2021.1914420] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/04/2022] Open
Abstract
Abdominal aortic aneurysm (AAA) involves the degradation of vascular fibres, and dilation and rupture of the abdominal aorta. Hypoperfusion in the vascular walls due to stenosis of the vasa vasorum is reportedly a cause of AAA onset and involves the induction of adventitial ectopic adipocytes. Recent studies have reported that ectopic adipocytes are associated with AAA rupture in both human and hypoperfusion-induced animal models, highlighting the pathological importance of hypoperfusion and adipocytes in AAA. However, the relationship between hypoperfusion and AAA remains unknown. In this study, we investigated the changes in inflammation-related factors in adipocytes at low glucose and serum levels. Low glucose and serum levels enhanced the production of AAA-related factors in 3T3-L1 cells. Low glucose and serum levels increased the activation of protein kinase B (also known as Akt), extracellular signal-regulated protein kinase 1/2, p38, c-Jun N-terminal kinase, and nuclear factor (NF) кB at the protein level. The inflammatory factors and related signalling pathways were markedly decreased following the return of the cells to normal culture conditions. These data suggest that low glucose and serum levels increase the levels of inflammatory factors through the activation of Akt, mitogen activated protein kinase, and NF-κB signalling pathways.
Collapse
Affiliation(s)
- Hirona Kugo
- Department of Applied Biological Chemistry, Graduate School of Agriculture, Kindai University, Nara City, Japan
| | - Wanida Sukketsiri
- Department of Pharmacology, Division of Health and Applied Sciences, Faculty of Science, Prince of Songkla University, Songkhla, Thailand
| | - Kazuko Iwamoto
- Department of Applied Biological Chemistry, Graduate School of Agriculture, Kindai University, Nara City, Japan
- Department of Health and Nutrition, Faculty of Health Science, Osaka Aoyama University, Minoh City, Japan
| | - Satoki Suihara
- Department of Applied Biological Chemistry, Graduate School of Agriculture, Kindai University, Nara City, Japan
| | - Tatsuya Moriyama
- Department of Applied Biological Chemistry, Graduate School of Agriculture, Kindai University, Nara City, Japan
- Agricultural Technology and Innovation Research Institute, Kindai University, Nara, Japan
| | - Nobuhiro Zaima
- Department of Applied Biological Chemistry, Graduate School of Agriculture, Kindai University, Nara City, Japan
- Agricultural Technology and Innovation Research Institute, Kindai University, Nara, Japan
| |
Collapse
|
35
|
Qing G, Zhiyuan W, Jinge Y, Yuqing M, Zuoguan C, Yongpeng D, Jinfeng Y, Junnan J, Yijia G, Weimin L, Yongjun L. Single-Cell RNA Sequencing Revealed CD14 + Monocytes Increased in Patients With Takayasu's Arteritis Requiring Surgical Management. Front Cell Dev Biol 2021; 9:761300. [PMID: 34671607 PMCID: PMC8521054 DOI: 10.3389/fcell.2021.761300] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2021] [Accepted: 09/13/2021] [Indexed: 12/26/2022] Open
Abstract
Objectives: Takayasu Arteritis (TA) is a highly specific vascular inflammation and poses threat to patients’ health. Although some patients have accepted medical treatment, their culprit lesions require surgical management (TARSM). This study aimed at dissecting the transcriptomes of peripheral blood mononuclear cells (PBMCs) in these patients and to explore potential clinical markers for TA development and progression. Methods: Peripheral blood were collected from four TA patients requiring surgical management and four age-sex matched healthy donors. Single cell RNA sequencing (scRNA-seq) was adopted to explore the transcriptomic diversity and function of their PBMCs. ELISA, qPCR, and FACS were conducted to validate the results of the analysis. Results: A total of 29918 qualified cells were included for downstream analysis. Nine major cell types were confirmed, including CD14+ monocytes, CD8+ T cells, NK cells, CD4+ T cells, B cells, CD16+ monocytes, megakaryocytes, dendritic cells and plasmacytoid dendritic cells. CD14+ monocytes (50.0 vs. 39.3%, p < 0.05) increased in TA patients, as validated by FACS results. TXNIP, AREG, THBS1, and CD163 increased in TA patients. ILs like IL-6, IL-6STP1, IL-6ST, IL-15, and IL-15RA increased in TA group. Conclusion: Transcriptome heterogeneities of PBMCs in TA patients requiring surgical management were revealed in the present study. In the patients with TA, CD14+ monocytes and gene expressions involved in oxidative stress were increased, indicating a new treatment and research direction in this field.
Collapse
Affiliation(s)
- Gao Qing
- Graduate School of Peking Union Medical College, Chinese Academy of Medical Science, Beijing, China.,Department of Vascular Surgery, National Centre of Gerontology, Beijing Hospital, Beijing, China.,National Tuberculosis Clinical Lab of China, Beijing Chest Hospital, Beijing Tuberculosis and Thoracic Tumor Research Institute, Capital Medical University, Beijing, China.,Beijing Key Laboratory in Drug Resistance Tuberculosis Research, Beijing Chest Hospital, Capital Medical University, Beijing, China
| | - Wu Zhiyuan
- Department of Vascular Surgery, National Centre of Gerontology, Beijing Hospital, Beijing, China
| | - Yu Jinge
- Institute of Statistics and Big Data, Renmin University of China, Beijing, China
| | - Miao Yuqing
- Graduate School of Peking Union Medical College, Chinese Academy of Medical Science, Beijing, China.,Department of Vascular Surgery, National Centre of Gerontology, Beijing Hospital, Beijing, China
| | - Chen Zuoguan
- Department of Vascular Surgery, National Centre of Gerontology, Beijing Hospital, Beijing, China
| | - Diao Yongpeng
- Department of Vascular Surgery, National Centre of Gerontology, Beijing Hospital, Beijing, China
| | - Yin Jinfeng
- National Tuberculosis Clinical Lab of China, Beijing Chest Hospital, Beijing Tuberculosis and Thoracic Tumor Research Institute, Capital Medical University, Beijing, China.,Beijing Key Laboratory in Drug Resistance Tuberculosis Research, Beijing Chest Hospital, Capital Medical University, Beijing, China
| | - Jia Junnan
- Beijing Key Laboratory in Drug Resistance Tuberculosis Research, Beijing Chest Hospital, Capital Medical University, Beijing, China
| | - Guo Yijia
- National Tuberculosis Clinical Lab of China, Beijing Chest Hospital, Beijing Tuberculosis and Thoracic Tumor Research Institute, Capital Medical University, Beijing, China.,Beijing Key Laboratory in Drug Resistance Tuberculosis Research, Beijing Chest Hospital, Capital Medical University, Beijing, China
| | - Li Weimin
- National Tuberculosis Clinical Lab of China, Beijing Chest Hospital, Beijing Tuberculosis and Thoracic Tumor Research Institute, Capital Medical University, Beijing, China.,Beijing Key Laboratory in Drug Resistance Tuberculosis Research, Beijing Chest Hospital, Capital Medical University, Beijing, China
| | - Li Yongjun
- Graduate School of Peking Union Medical College, Chinese Academy of Medical Science, Beijing, China.,Department of Vascular Surgery, National Centre of Gerontology, Beijing Hospital, Beijing, China
| |
Collapse
|
36
|
Qiu R, Chen S, Hua F, Bian S, Chen J, Li G, Wu X. Betanin Prevents Experimental Abdominal Aortic Aneurysm Progression by Modulating the TLR4/NF-κB and Nrf2/HO-1 Pathways. Biol Pharm Bull 2021; 44:1254-1262. [PMID: 34471054 DOI: 10.1248/bpb.b21-00042] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Betanin, a bioactive ingredient mostly isolated from beetroots, exhibits a protective effect against cardiovascular diseases. However, its effects on abdominal aortic aneurysm (AAA) have not been elucidated. In this study, an AAA model was constructed by infusion of porcine pancreatic elastase in C57BL/6 mice. Mice were then administered with betanin or saline intragastrically once daily for 14 d. Our results showed that treatment with betanin remarkably limited AAA enlargement and mitigated the infiltration of inflammatory cells in the adventitia. The increased expression of proinflammatory cytokines and matrix metalloproteinases (MMPs) was also significantly alleviated following betanin treatment. Furthermore, betanin suppressed the activation of toll-like receptor 4 (TLR4)/nuclear factor-kappaB (NF-κB) signaling in the aortic wall, and downregulated the levels of tissue-reactive oxygen species as well as circulating 8-isoprostane by stimulating the nuclear factor-E2-related factor 2 (Nrf2)/heme oxygenase-1 (HO-1) pathway. Taken together, these data suggest that betanin may attenuate AAA progression and may be used as a therapeutic drug against AAA.
Collapse
Affiliation(s)
- Renfeng Qiu
- Department of Vascular Surgery, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University.,Department of Vascular Surgery, Shouguang People Hospital
| | - Shuxiao Chen
- Department of Vascular Surgery, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University
| | - Fang Hua
- Department of Clinical Laboratory, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University
| | - Shuai Bian
- Department of Vascular Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University
| | - Jianfeng Chen
- Department of Vascular Surgery, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University
| | - Gang Li
- Department of Vascular Surgery, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University.,Department of Vascular Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University
| | - Xuejun Wu
- Department of Vascular Surgery, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University.,Department of Vascular Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University
| |
Collapse
|
37
|
Circular RNA Expression: Its Potential Regulation and Function in Abdominal Aortic Aneurysms. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:9934951. [PMID: 34306317 PMCID: PMC8263248 DOI: 10.1155/2021/9934951] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Accepted: 05/30/2021] [Indexed: 12/18/2022]
Abstract
Abdominal aortic aneurysms (AAAs) have posed a great threat to human life, and the necessity of its monitoring and treatment is decided by symptomatology and/or the aneurysm size. Accumulating evidence suggests that circular RNAs (circRNAs) contribute a part to the pathogenesis of AAAs. circRNAs are novel single-stranded RNAs with a closed loop structure and high stability, having become the candidate biomarkers for numerous kinds of human disorders. Besides, circRNAs act as molecular "sponge" in organisms, capable of regulating the transcription level. Here, we characterize that the molecular mechanisms underlying the role of circRNAs in AAA development were further elucidated. In the present work, studies on the biosynthesis, bibliometrics, and mechanisms of action of circRNAs were aims comprehensively reviewed, the role of circRNAs in the AAA pathogenic mechanism was illustrated, and their potential in diagnosing AAAs was examined. Moreover, the current evidence about the effects of circRNAs on AAA development through modulating endothelial cells (ECs), macrophages, and vascular smooth muscle cells (VSMCs) was summarized. Through thorough investigation, the molecular mechanisms underlying the role of circRNAs in AAA development were further elucidated. The results demonstrated that circRNAs had the application potential in the diagnosis and prevention of AAAs in clinical practice. The study of circRNA regulatory pathways would be of great assistance to the etiologic research of AAAs.
Collapse
|
38
|
Aberrant Mitochondrial Dynamics: An Emerging Pathogenic Driver of Abdominal Aortic Aneurysm. Cardiovasc Ther 2021; 2021:6615400. [PMID: 34221126 PMCID: PMC8221877 DOI: 10.1155/2021/6615400] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/27/2020] [Revised: 05/13/2021] [Accepted: 06/02/2021] [Indexed: 12/12/2022] Open
Abstract
Abdominal aortic aneurysm (AAA) is defined as a progressive segmental dilation of the abdominal aorta and is associated with high mortality. The characterized features of AAA indicate several underlying mechanisms of AAA formation and progression, including reactive oxygen species production, inflammation, and atherosclerosis. Mitochondrial functions are critical for determining cell fate, and mitochondrial dynamics, especially selective mitochondrial autophagy, which is termed as mitophagy, has emerged as an important player in the pathogenesis of several cardiovascular diseases. The PARKIN/PARIS/PGC1α pathway is associated with AAA formation and has been proposed to play a role in mitochondrial dynamics mediated by the PINK/PARKIN pathway in the pathogenesis underlying AAA. This review is aimed at deepening our understanding of AAA formation and progression, which is vital for the development of potential medical therapies for AAA.
Collapse
|
39
|
Summerhill VI, Sukhorukov VN, Eid AH, Nedosugova LV, Sobenin IA, Orekhov AN. Pathophysiological Aspects of the Development of Abdominal Aortic Aneurysm with a Special Focus on Mitochondrial Dysfunction and Genetic Associations. Biomol Concepts 2021; 12:55-67. [PMID: 34115932 DOI: 10.1515/bmc-2021-0007] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Accepted: 04/28/2021] [Indexed: 01/01/2023] Open
Abstract
Abdominal aortic aneurysm (AAA) is a complex degenerative vascular disease, with considerable morbidity and mortality rates among the elderly population. The mortality of AAA is related to aneurysm expansion (the enlargement of the aortic diameter up to 30 mm and above) and the subsequent rupture. The pathogenesis of AAA involves several biological processes, including aortic mural inflammation, oxidative stress, vascular smooth muscle cell apoptosis, elastin depletion, and degradation of the extracellular matrix. Mitochondrial dysfunction was also found to be associated with AAA formation. The evidence accumulated to date supports a close relationship between environmental and genetic factors in AAA initiation and progression. However, a comprehensive pathophysiological understanding of AAA formation remains incomplete. The open surgical repair of AAA is the only therapeutic option currently available, while a specific pharmacotherapy is still awaited. Therefore, there is a great need to clarify pathophysiological cellular and molecular mechanisms underlying AAA formation that would help to develop effective pharmacological therapies. In this review, pathophysiological aspects of AAA development with a special focus on mitochondrial dysfunction and genetic associations were discussed.
Collapse
Affiliation(s)
- Volha I Summerhill
- Department of Basic Research, Institute for Atherosclerosis Research, Moscow 121609, Russia
| | - Vasily N Sukhorukov
- Laboratory of Cellular and Molecular Pathology of Cardiovascular System, Research Institute of Human Morphology, 3 Tsyurupa Street, Moscow 117418, Russia
| | - Ali H Eid
- Department of Basic Medical Sciences, College of Medicine, QU Health, Qatar University, PO Box 2713, Doha, Qatar.,Biomedical and Pharmaceutical Research Unit, QU Health, Qatar University, PO Box 2713, Doha, Qatar.,Department of Pharmacology and Toxicology, Faculty of Medicine, American University of Beirut, PO Box 11-0236, Beirut-Lebanon
| | - Ludmila V Nedosugova
- I.M. Sechenov First Moscow State Medical University (Sechenov University), 8/2 Trubenskaya Street, Moscow 119991, Russia
| | - Igor A Sobenin
- Laboratory of Cellular and Molecular Pathology of Cardiovascular System, Research Institute of Human Morphology, 3 Tsyurupa Street, Moscow 117418, Russia.,Laboratory of Medical Genetics, National Medical Research Center of Cardiology, 15A 3-rd Cherepkovskaya Street, Moscow 121552, Russia.,Laboratory of Angiopathology, Institute of General Pathology and Pathophysiology, 8 Baltiiskaya Street, Moscow 125315, Russia
| | - Alexander N Orekhov
- Department of Basic Research, Institute for Atherosclerosis Research, Moscow 121609, Russia.,Laboratory of Cellular and Molecular Pathology of Cardiovascular System, Research Institute of Human Morphology, 3 Tsyurupa Street, Moscow 117418, Russia
| |
Collapse
|
40
|
Bilen A, Mercantepe F, Tümkaya L, Yilmaz A, Batcik Ş. The hepatoprotective potential of resveratrol in an experimental model of ruptured abdominal aortic aneurysm via oxidative stress and apoptosis. J Biochem Mol Toxicol 2021; 35:e22836. [PMID: 34075649 DOI: 10.1002/jbt.22836] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2021] [Revised: 05/07/2021] [Accepted: 05/19/2021] [Indexed: 12/19/2022]
Abstract
The mortality rate in ruptured abdominal aortic aneurysms can today be reduced through cardiovascular surgery. However, ischemia and reperfusion-induced tissue damage develop due to aortic cross-clamping applied during surgery. The present study aimed to reduce oxidative stress-induced hepatic damage resulting from ischemia and reperfusion due to aortic cross-clamping during surgery by means of resveratrol administration. Forty male Sprague-Dawley rats were randomly assigned into four groups: control (healthy), glycerol+ischemia/reperfusion (I/R) (sham), I/R, and I/R + Resveratrol. In all groups scheduled for I/R, 60 min of shock was followed by 60 min of ischemia. In the I/R + Resveratrol group, 10 mg/kg of resveratrol was administered 15 min before ischemia and immediately before reperfusion via the intraperitoneal route. In addition, 120 min of reperfusion was applied under anesthesia after ischemia in all groups. Intralobar and interlobar necrosis, vascular congestion, and edematous fields resulting from aortic occlusion were present. Liver tissue malondialdehyde (MDA) levels and cleaved caspase-3 positivity increased, while glutathione (GSH) levels decreased. However, resveratrol administration reduced intralobular and interlobar necrosis, vascular congestion and edematous fields, cleaved caspase-3 positivity, and MDA levels, and increased GSH levels. Our findings suggest that resveratrol is effective against aortic occlusion-induced liver injury by reducing oxidative stress and apoptosis.
Collapse
Affiliation(s)
- Arzu Bilen
- Department of Endocrinology and metabolism diseases, Faculty of Medicine, Ataturk University, Erzurum, Turkey
| | - Filiz Mercantepe
- Department of Endocrinology and metabolism diseases, Faculty of Medicine, Ataturk University, Erzurum, Turkey
| | - Levent Tümkaya
- Department of Histology and Embryology, Faculty of Medicine, Recep Tayyip Erdogan University, Rize, Turkey
| | - Adnan Yilmaz
- Department of Biochemistry, Faculty of Medicine, Recep Tayyip Erdogan University, Rize, Turkey
| | - Şule Batcik
- Department of Anesthesiology and Reanimation, Faculty of Medicine, Recep Tayyip Erdogan University, Rize, Turkey
| |
Collapse
|
41
|
Wu J, Wang W, Chen Z, Xu F, Zheng Y. Proteomics applications in biomarker discovery and pathogenesis for abdominal aortic aneurysm. Expert Rev Proteomics 2021; 18:305-314. [PMID: 33840337 DOI: 10.1080/14789450.2021.1916473] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Introduction: Abdominal aortic aneurysm (AAA) is a common, complex, and life-threatening disease. Currently, the pathogenesis of AAA is not well understood. No biomarkers or specific drugs are available for AAA in clinical applications. Proteomics is a powerful tool in biomarker discovery, exploration of pathogenesis, and drug target identification.Areas covered: We review the application of mass spectrometry-based proteome analysis in AAA patients within the last ten years. Differentially expressed proteins associated with AAA were identified in multiple sample sources, including vascular tissue, intraluminal thrombus, tissue secretome, blood, and cells. Some potential disease biomarkers, pathogenic mechanisms, or therapeutic targets for AAA were discovered using proteome analysis. The challenges and prospects of proteomics applied to AAA are also discussed.Expert opinion: Since most of the previous proteomic studies used relatively small sample sizes, some promising biomarkers need to be validated in multicenter cohorts to accelerate their clinical application. With the rapid development of mass spectrometry technology, modification-specific proteomics and multi-omics research in the future will enhance our understanding of the pathogenesis of AAA and promote biomarker discovery and drug development for clinical translation.
Collapse
Affiliation(s)
- Jianqiang Wu
- Department of Vascular Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.,Medical Research Center, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Wei Wang
- Department of Vascular Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Zhaoran Chen
- Department of Geriatrics, Medical Health Center, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Fang Xu
- Department of Vascular Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yuehong Zheng
- Department of Vascular Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
42
|
Shi J, Guo J, Li Z, Xu B, Miyata M. Importance of NLRP3 Inflammasome in Abdominal Aortic Aneurysms. J Atheroscler Thromb 2021; 28:454-466. [PMID: 33678767 PMCID: PMC8193780 DOI: 10.5551/jat.rv17048] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2021] [Accepted: 01/22/2021] [Indexed: 12/14/2022] Open
Abstract
Abdominal aortic aneurysm (AAA) is a chronic inflammatory degenerative aortic disease, which particularly affects older people. Nucleotide-binding oligomerization domain-like receptor family protein 3 (NLRP3) inflammasome is a multi-protein complex and mediates inflammatory responses by activating caspase 1 for processing premature interleukin (IL)-1β and IL-18. In this review, we first summarize the principle of NLRP3 inflammasome activation and the functionally distinct classes of small molecule NLRP3 inflammasome inhibitors. Next, we provide a comprehensive literature review on the expression of NLRP3 inflammasome effector mediators (IL-1β and IL-18) and components (caspase 1, apoptosis-associated speck-like protein containing a caspase recruitment domain (ASC) and NLRP3) in clinical and experimental AAAs. Finally, we discuss the influence of genetic deficiency or pharmacological inhibition of individual effector mediators and components of NLRP3 inflammasome on experimental AAAs. Accumulating clinical and experimental evidence suggests that NLRP3 inflammasome may be a promise therapeutic target for developing pharmacological strategies for clinical AAA management.
Collapse
Affiliation(s)
- Jinyun Shi
- Center for Hypertension Care, Shanxi Medical University First Hospital, Taiyuan, Shanxi Province, P. R. China
| | - Jia Guo
- Center for Hypertension Care, Shanxi Medical University First Hospital, Taiyuan, Shanxi Province, P. R. China
| | - Zhidong Li
- Department of Pharmacology, Shanxi Medical University, Taiyuan, Shanxi Province, P. R. China
| | - Baohui Xu
- Department of Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Masaaki Miyata
- School of Health Science, Faculty of Medicine, Kagoshima University, Kagoshima University, Kagoshima, Japan
| |
Collapse
|
43
|
Cai Z, Huang J, Yang J, Pan B, Wang W, Ou Y, Wang X, Yang P. LncRNA SENCR suppresses abdominal aortic aneurysm formation by inhibiting smooth muscle cells apoptosis and extracellular matrix degradation. Bosn J Basic Med Sci 2021; 21:323-330. [PMID: 32903173 PMCID: PMC8112567 DOI: 10.17305/bjbms.2020.4994] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Accepted: 08/27/2020] [Indexed: 12/16/2022] Open
Abstract
Abdominal aortic aneurysm (AAA) is a progressive chronic dilatation of the abdominal aorta without effective medical treatment. This study aims to clarify the potential of long non-coding RNA SENCR as a treatment target in AAA. Angiotensin II (Ang-II) was used to establish AAA model in vitro and in vivo. Reverse transcription quantitative PCR and western blot were performed to measure the expression of SENCR and proteins, respectively. Annexin V-FITC/PI double staining was carried out to detect the apoptotic rate in vascular smooth muscle cells (VSMCs), and cell apoptosis in aortic tissues was determined by TUNEL staining. Besides, hematoxylin and eosin and Elastica van Gieson staining were performed for histological analysis of aortic tissues. SENCR was downregulated in AAA tissues and Ang-II-stimulated VSMCs. Overexpression of SENCR could inhibit Ang-II-induced VSMC apoptosis, while inhibition of SENCR facilitated Ang-II-induced VSMC apoptosis. Moreover, the expression of matrix metalloproteinase (MMP)-2 and MMP-9 in Ang-II-induced VSMCs was reduced following SENCR overexpression, while tissue inhibitor of metalloproteinases 1 (TIMP-1) expression was increased. In vivo, overexpression of SENCR improved the pathological change in aortic tissues and the damage in arterial wall elastic fibers induced by Ang-II, as well as suppressed Ang-II-induced cell apoptosis and extracellular matrix degradation. Overall, SENCR was decreased in AAA. Overexpression of SENCR inhibited AAA formation via inhibition of VSMC apoptosis and extracellular matrix degradation. We provided a reliable evidence for SENCR acting as a potential target for AAA treatment.
Collapse
Affiliation(s)
- Zhou Cai
- Department of General Surgery, Xiangya Hospital Central South University, Changsha, China
| | - Jianhua Huang
- Department of General Surgery, Xiangya Hospital Central South University, Changsha, China
| | - Junxiao Yang
- Department of Orthopedics, Xiangya Hospital Central South University, Changsha, China
| | - Baihong Pan
- Department of General Surgery, Xiangya Hospital Central South University, Changsha, China
| | - Wei Wang
- Department of General Surgery, Xiangya Hospital Central South University, Changsha, China
| | - Yangyang Ou
- Department of General Surgery, Xiangya Hospital Central South University, Changsha, China
| | - Xianwei Wang
- Department of General Surgery, Xiangya Hospital Central South University, Changsha, China
| | - Pu Yang
- Department of General Surgery, Xiangya Hospital Central South University, Changsha, China
| |
Collapse
|
44
|
Sánchez-Infantes D, Nus M, Navas-Madroñal M, Fité J, Pérez B, Barros-Membrilla AJ, Soto B, Martínez-González J, Camacho M, Rodriguez C, Mallat Z, Galán M. Oxidative Stress and Inflammatory Markers in Abdominal Aortic Aneurysm. Antioxidants (Basel) 2021; 10:602. [PMID: 33919749 PMCID: PMC8070751 DOI: 10.3390/antiox10040602] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Revised: 04/08/2021] [Accepted: 04/12/2021] [Indexed: 12/18/2022] Open
Abstract
Abdominal aortic aneurysm (AAA) is increasing due to aging of the population and is a major cause of death among the elderly. Ultrasound screening programs are useful in early diagnosis, but aneurysm size is not always a good predictor of rupture. Our aim was to analyze the value of circulating molecules related to oxidative stress and inflammation as new biomarkers to assist the management of AAA. The markers were quantified by ELISA, and their expression in the aneurysmal wall was studied by real-time PCR and by immunostaining. Correlation analysis of the studied markers with aneurysm diameter and peak wall stress (PWS), obtained by finite element analysis, and multivariate regression analysis to assess potential confounding factors were performed. Our study shows an extensive inflammatory infiltration in the aneurysmal wall, mainly composed by T-cells, macrophages and B-cells and altered levels of reactive oxygen species (ROS), IgM, IgG, CD38, GDF15, S100A4 and CD36 in plasma and in the aneurysmal tissue of AAA patients compared with controls. Circulating levels of IgG, CD38 and GDF15 positively correlated with abdominal aortic diameter, and CD38 was correlated with PWS. Our data show that altered levels of IgG, CD38 and GDF15 have potential diagnostic value in the assessment of AAA.
Collapse
Affiliation(s)
- David Sánchez-Infantes
- Department of Basic Sciences of Health, Area of Biochemistry and Molecular Biology, University Rey Juan Carlos, 28922 Alcorcón, Spain;
- Centro de Investigación Biomédica en Red-Fisiopatología de la Obesidad y Nutrición (CIBEROBN), ISCIII, 28029 Madrid, Spain
| | - Meritxell Nus
- Division of Cardiovascular Medicine, University of Cambridge, Cambridge CB2 0QQ, UK; (M.N.); (Z.M.)
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), ISCIII, 28029 Madrid, Spain; (J.M.-G.); (M.C.); (C.R.)
| | - Miquel Navas-Madroñal
- Institut de Recerca del Hospital de la Santa Creu i Sant Pau, 08041 Barcelona, Spain;
- Instituto de Investigación Biomédica Sant Pau (IB Sant Pau), 08025 Barcelona, Spain
| | - Joan Fité
- Servicio de Angiología, Cirugía Vascular y Endovascular, Hospital de la Santa Creu i Sant Pau, 08041 Barcelona, Spain; (J.F.); (B.S.)
| | - Belén Pérez
- Faculty of Medicine, Universidad Autónoma de Barcelona, Bellaterra, 08193 Barcelona, Spain;
| | - Antonio J. Barros-Membrilla
- Unidad Funcional de Patología de la Aorta (UPA), Servicio de Cardiología, Hospital de la Santa Creu i Sant Pau, 08041 Barcelona, Spain;
| | - Begoña Soto
- Servicio de Angiología, Cirugía Vascular y Endovascular, Hospital de la Santa Creu i Sant Pau, 08041 Barcelona, Spain; (J.F.); (B.S.)
| | - José Martínez-González
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), ISCIII, 28029 Madrid, Spain; (J.M.-G.); (M.C.); (C.R.)
- Instituto de Investigación Biomédica Sant Pau (IB Sant Pau), 08025 Barcelona, Spain
- Instituto de Investigaciones Biomédicas de Barcelona-Consejo Superior de Investigaciones Científicas (IIBB-CSIC), 08036 Barcelona, Spain
| | - Mercedes Camacho
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), ISCIII, 28029 Madrid, Spain; (J.M.-G.); (M.C.); (C.R.)
- Institut de Recerca del Hospital de la Santa Creu i Sant Pau, 08041 Barcelona, Spain;
- Instituto de Investigación Biomédica Sant Pau (IB Sant Pau), 08025 Barcelona, Spain
| | - Cristina Rodriguez
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), ISCIII, 28029 Madrid, Spain; (J.M.-G.); (M.C.); (C.R.)
- Institut de Recerca del Hospital de la Santa Creu i Sant Pau, 08041 Barcelona, Spain;
- Instituto de Investigación Biomédica Sant Pau (IB Sant Pau), 08025 Barcelona, Spain
| | - Ziad Mallat
- Division of Cardiovascular Medicine, University of Cambridge, Cambridge CB2 0QQ, UK; (M.N.); (Z.M.)
| | - María Galán
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), ISCIII, 28029 Madrid, Spain; (J.M.-G.); (M.C.); (C.R.)
- Institut de Recerca del Hospital de la Santa Creu i Sant Pau, 08041 Barcelona, Spain;
- Instituto de Investigación Biomédica Sant Pau (IB Sant Pau), 08025 Barcelona, Spain
| |
Collapse
|
45
|
XIST knockdown suppresses vascular smooth muscle cell proliferation and induces apoptosis by regulating miR-1264/WNT5A/β-catenin signaling in aneurysm. Biosci Rep 2021; 41:227680. [PMID: 33501488 PMCID: PMC7960886 DOI: 10.1042/bsr20201810] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2020] [Revised: 12/28/2020] [Accepted: 01/15/2021] [Indexed: 12/30/2022] Open
Abstract
Long non-coding RNAs (lncRNAs) have been ascertained as vital modulators in abdominal aortic aneurysm (AAA) development. In this research, the function and molecular mechanisms of the lncRNA X-inactive specific transcript (XIST) in the evolution of vascular smooth muscle cells (VSMCs) were assessed. Results showed that XIST expression was increased but miR-1264 expression level was reduced in the serum of AAA patients. XIST depletion impeded human aorta VSMCs (HA-VSMCs’) ability to proliferate and stimulate apoptosis, while repressing miR-1264 expression through an unmediated interaction. Additionally, the influence of XIST knockdown on apoptosis and proliferation could be rescued by an miR-1264 inhibitor. Subsequent molecular investigations indicated that WNT5A was miR-1264’s target, and XIST functioned as a competing endogenous RNA (ceRNA) of miR-1264 to raise WNT5A expression. Further, an miR-1264 inhibitor stimulated the proliferation and suppressed the apoptosis of HA-VSMCs through the activation of WNT/β-catenin signaling. Taken together, XIST impeded the apoptosis and stimulated the proliferation of HA-VSMCs via the WNT/β-catenin signaling pathway through miR-1264, demonstrating XIST’s underlying role in AAA.
Collapse
|
46
|
Time-Dependent Pathological Changes in Hypoperfusion-Induced Abdominal Aortic Aneurysm. BIOLOGY 2021; 10:biology10020149. [PMID: 33672844 PMCID: PMC7917844 DOI: 10.3390/biology10020149] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Revised: 02/11/2021] [Accepted: 02/12/2021] [Indexed: 11/19/2022]
Abstract
Simple Summary Abdominal aortic aneurysm (AAA) is a vascular disease that involves gradual dilation of the abdominal aorta and has a high mortality due to rupture. Hypoperfusion due to the obstruction of vasa vasorum, which is a blood supply system in the aortic wall, may be an important factor involved in AAA pathophysiology. A time-dependent analysis is important to understand the pathological cascade following hypoperfusion in the aortic wall. In our study, time-dependent analysis using a hypoperfusion-induced animal model showed that the dynamics of many AAA-related factors might be associated with the increased hypoxia-inducible factor-1α level. Hypoperfusion due to stenosis of the vasa vasorum might be a new drug target for AAA therapeutics. Abstract Hypoperfusion due to vasa vasorum stenosis can cause wall hypoxia and abdominal aortic aneurysm (AAA) development. Even though hypoperfusion is an important contributor toward pathological changes in AAA, the correlation between hypoperfusion and AAA is not fully understood. In this study, a time-dependent semi-quantitative pathological analysis of hypoperfusion-induced aortic wall changes was performed to understand the mechanisms underlying the gradual degradation of the aortic wall leading to AAA formation. AAA-related factors evaluated in this study were grouped according to the timing of dynamic change, and five groups were formed as follows: first group: angiotensin II type 1 receptor, endothelin-1 (ET-1), and malondialdehyde (MDA); second group: matrix metalloproteinase (MMP)-2, -9, -12, M1 macrophages (Mac387+ cells), and monocyte chemotactic protein-1; third group: synthetic smooth muscle cells (SMCs); fourth group: neutrophil elastase, contractile SMCs, and angiotensinogen; and the fifth group: M2 macrophages (CD163+ cells). Hypoxia-inducible factor-1α, ET-1, MDA, and MMP-9 were colocalized with alpha-smooth muscle actin cells in 3 h, suggesting that hypoperfusion-induced hypoxia directly affects the activities of contractile SMCs in the initial stage of AAA. Time-dependent pathological analysis clarified the cascade of AAA-related factors. These findings provide clues for understanding complicated multistage pathologies in AAA.
Collapse
|
47
|
Chen T, Zhang H, Zhang Y, Yang M, Wu J, Yang M, Lin J, Gao W, Tang L, Xu B, Jiang J, Chen X. Association of Circulating and Aortic Zinc and Copper Levels with Clinical Abdominal Aortic Aneurysm: a Meta-analysis. Biol Trace Elem Res 2021; 199:513-526. [PMID: 32557106 DOI: 10.1007/s12011-020-02187-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/04/2020] [Accepted: 05/06/2020] [Indexed: 01/26/2023]
Abstract
It remains obscure whether circulating aortic zinc (Zn) and copper (Cu) levels are associated with the progress of human abdominal aortic aneurysms (AAA). Therefore, we conducted a meta-analysis to explore this relationship. A literature search on circulating and aortic zinc and copper levels and AAA patients was conducted using online databases including PubMed, Embase, and Cochrane up to March 20, 2019. To compare Zn and Cu concentrations in AAA patients with those in aortic occlusive disease (AOD) patients or healthy aorta donors or healthy blood donors, pooled weighted mean difference (WMD) and its 95% confidence interval (CI) were calculated. Subgroup analysis, sensitivity analysis, and meta-regression analysis were applied to explain the heterogeneity and evaluate the robustness of combined results. A total of 10 cross-sectional studies, including 252 cases and 304 controls, were used for meta-analysis. We found that circulating zinc and Zn/Cu ratio in AAA patients were significantly lower [WMD (95%CI): - 2.23 (- 4.10, - 0.36); - 0.18 (- 0.31, - 0.05), respectively] than those in non-AAA patients. Similarly, aneurysmal aorta had significantly lower zinc levels and Zn/Cu ratio [WMD (95%CI): - 9.22 (- 15.37, - 3.07); - 6.46 (- 10.14, - 2.77), respectively] than those in control group. No difference in circulating or aortic copper levels was noted between AAA patients and control group [WMD (95%CI): - 0.24 (- 2.09, 1.61); 0.30 (- 0.01, 0.61) , respectively]. Our meta-analysis suggests that zinc levels and Zn-Cu ratio, but not copper levels, may influence aneurysmal progress of AAA.
Collapse
Affiliation(s)
- Tingting Chen
- Department of Cardiology, Taizhou Hospital Affiliated to Wenzhou Medical University, Linhai, 317000, Zhejiang Province, China
- Laboratory of Cardiovascular Disease, Taizhou Hospital Affiliated to Wenzhou Medical University, Linhai, 317000, Zhejiang Province, China
| | - Hongliang Zhang
- Department of Cardiology, Taizhou Hospital Affiliated to Wenzhou Medical University, Linhai, 317000, Zhejiang Province, China
- Laboratory of Cardiovascular Disease, Taizhou Hospital Affiliated to Wenzhou Medical University, Linhai, 317000, Zhejiang Province, China
| | - Yang Zhang
- Department of Cardiology, Taizhou Hospital Affiliated to Wenzhou Medical University, Linhai, 317000, Zhejiang Province, China
- Laboratory of Cardiovascular Disease, Taizhou Hospital Affiliated to Wenzhou Medical University, Linhai, 317000, Zhejiang Province, China
| | - Mengqi Yang
- Department of Cardiology, Taizhou Hospital Affiliated to Wenzhou Medical University, Linhai, 317000, Zhejiang Province, China
- Laboratory of Cardiovascular Disease, Taizhou Hospital Affiliated to Wenzhou Medical University, Linhai, 317000, Zhejiang Province, China
| | - Juntao Wu
- Department of Cardiology, Taizhou Hospital Affiliated to Wenzhou Medical University, Linhai, 317000, Zhejiang Province, China
- Laboratory of Cardiovascular Disease, Taizhou Hospital Affiliated to Wenzhou Medical University, Linhai, 317000, Zhejiang Province, China
| | - Minjun Yang
- Department of Cardiology, Taizhou Hospital Affiliated to Wenzhou Medical University, Linhai, 317000, Zhejiang Province, China
- Laboratory of Cardiovascular Disease, Taizhou Hospital Affiliated to Wenzhou Medical University, Linhai, 317000, Zhejiang Province, China
| | - Jiangbo Lin
- Department of Cardiology, Taizhou Hospital Affiliated to Wenzhou Medical University, Linhai, 317000, Zhejiang Province, China
- Laboratory of Cardiovascular Disease, Taizhou Hospital Affiliated to Wenzhou Medical University, Linhai, 317000, Zhejiang Province, China
| | - Weixu Gao
- Department of Endocrinology, Taizhou Hospital Affiliated to Wenzhou Medical University, Linhai, 317000, Zhejiang Province, China
| | - Lijiang Tang
- Department of Cardiology, Zhejiang Hospital, Hangzhou, 310013, Zhejiang Province, China
| | - Baohui Xu
- Department of Surgery, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Jianjun Jiang
- Department of Cardiology, Taizhou Hospital Affiliated to Wenzhou Medical University, Linhai, 317000, Zhejiang Province, China
- Laboratory of Cardiovascular Disease, Taizhou Hospital Affiliated to Wenzhou Medical University, Linhai, 317000, Zhejiang Province, China
| | - Xiaofeng Chen
- Department of Cardiology, Taizhou Hospital Affiliated to Wenzhou Medical University, Linhai, 317000, Zhejiang Province, China.
- Laboratory of Cardiovascular Disease, Taizhou Hospital Affiliated to Wenzhou Medical University, Linhai, 317000, Zhejiang Province, China.
| |
Collapse
|
48
|
Piqueras L, Sanz MJ. Angiotensin II and leukocyte trafficking: New insights for an old vascular mediator. Role of redox-signaling pathways. Free Radic Biol Med 2020; 157:38-54. [PMID: 32057992 DOI: 10.1016/j.freeradbiomed.2020.02.002] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/20/2019] [Revised: 01/27/2020] [Accepted: 02/03/2020] [Indexed: 12/20/2022]
Abstract
Inflammation and activation of the immune system are key molecular and cellular events in the pathogenesis of cardiovascular diseases, including atherosclerosis, hypertension-induced target-organ damage, and abdominal aortic aneurysm. Angiotensin II (Ang-II) is the main effector peptide hormone of the renin-angiotensin system. Beyond its role as a potent vasoconstrictor and regulator of blood pressure and fluid homeostasis, Ang-II is intimately involved in the development of vascular lesions in cardiovascular diseases through the activation of different immune cells. The migration of leukocytes from circulation to the arterial subendothelial space is a crucial immune response in lesion development that is mediated through a sequential and coordinated cascade of leukocyte-endothelial cell adhesive interactions involving an array of cell adhesion molecules present on target leukocytes and endothelial cells and the generation and release of chemoattractants that activate and guide leukocytes to sites of emigration. In this review, we outline the key events of Ang-II participation in the leukocyte recruitment cascade, the underlying mechanisms implicated, and the corresponding redox-signaling pathways. We also address the use of inhibitor drugs targeting the effects of Ang-II in the context of leukocyte infiltration in these cardiovascular pathologies, and examine the clinical data supporting the relevance of blocking Ang-II-induced vascular inflammation.
Collapse
Affiliation(s)
- Laura Piqueras
- Department of Pharmacology, Faculty of Medicine, University of Valencia, Valencia, Spain; Institute of Health Research INCLIVA University Clinic Hospital of Valencia, Valencia, Spain; CIBERDEM-Spanish Biomedical Research Center in Diabetes and Associated Metabolic Disorders, Carlos III Health Institute, Spanish Ministry of Health, Madrid, Spain.
| | - Maria-Jesus Sanz
- Department of Pharmacology, Faculty of Medicine, University of Valencia, Valencia, Spain; Institute of Health Research INCLIVA University Clinic Hospital of Valencia, Valencia, Spain; CIBERDEM-Spanish Biomedical Research Center in Diabetes and Associated Metabolic Disorders, Carlos III Health Institute, Spanish Ministry of Health, Madrid, Spain.
| |
Collapse
|
49
|
Tsai SH, Hsu LA, Tsai HY, Yeh YH, Lu CY, Chen PC, Wang JC, Chiu YL, Lin CY, Hsu YJ. Aldehyde dehydrogenase 2 protects against abdominal aortic aneurysm formation by reducing reactive oxygen species, vascular inflammation, and apoptosis of vascular smooth muscle cells. FASEB J 2020; 34:9498-9511. [PMID: 32463165 DOI: 10.1096/fj.201902550rrr] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2019] [Revised: 05/04/2020] [Accepted: 05/07/2020] [Indexed: 12/14/2022]
Abstract
Mitochondrial aldehyde dehydrogenase 2 (ALDH2) is an enzyme that detoxifies aldehydes by converting them to carboxylic acids. ALDH2 deficiency is known to increase oxidative stress. Increased oxidative stress plays a pivotal role in abdominal aortic aneurysm (AAA) pathogenesis. Reactive oxygen species (ROS) promote degradation of the extracellular matrix (ECM) and vascular smooth muscle cell (VSMC) apoptosis. Reducing oxidative stress by an ALDH2 activator could have therapeutic potential for limiting AAA development. We hypothesized that ALDH2 deficiency could increase the risk for AAA by decreasing ROS elimination and that an ALDH2 activator could provide an alternative option for AAA treatment. The National Center for Biotechnology (NCBI) Gene Expression Omnibus (GEO) database was used. Human aortic smooth muscle cells (HASMCs) were used for the in vitro experiments. Gene-targeted ALDH2*2 KI knock-in mice on a C57BL/6J background and apolipoprotein E knockout (ApoE KO) mice were obtained. An animal model of AAA was constructed using osmotic minipumps to deliver 1000 ng/kg/min angiotensin II (AngII) for 28 days. Patients with AAA had significantly lower ALDH2 expression levels than normal subjects. ALDH2*2 KI mice were susceptible to AngII administration, exhibiting significantly increased AAA incidence rates and increased aortic diameters. Alda-1, an ALDH2 activator, reduced AngII-induced ROS production, NF-kB activation, and apoptosis in HASMCs. Alda-1 attenuated AngII-induced aneurysm formation and decreased aortic expansion in ApoE KO mice. We concluded that ALDH2 deficiency is associated with the development of AAAs in humans and a murine disease model. ALDH2 deficiency increases susceptibility to AngII-induced AAA formation by attenuating anti-ROS effects and increasing VSMC apoptosis and vascular inflammation. Alda-1 was shown to attenuate the progression of experimental AAA in a murine model.
Collapse
Affiliation(s)
- Shih-Hung Tsai
- Department of Emergency Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan.,Department of Physiology and Biophysics, Graduate Institute of Physiology, National Defense Medical Center, Taipei, Taiwan
| | - Lung-An Hsu
- Cardiovascular Department, Chang-Gung Memorial Hospital and School of Medicine, Chang-Gung University, Taoyuan, Taiwan
| | - Hsiao-Ya Tsai
- Department of Emergency Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Yung-Hsin Yeh
- Cardiovascular Department, Chang-Gung Memorial Hospital and School of Medicine, Chang-Gung University, Taoyuan, Taiwan
| | - Cheng-Yo Lu
- Department of Emergency Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Po-Chuan Chen
- Department of Emergency Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Jen-Chun Wang
- Department of Emergency Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan.,Institute of Clinical Medicine, National Yang-Ming University, Taipei, Taiwan
| | - Yi-Lin Chiu
- Department of Biochemistry, National Defense Medical Center, Taipei, Taiwan
| | - Chih-Yuan Lin
- Department of Surgery, Division of Cardiovascular surgery, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Yu-Juei Hsu
- Division of Nephrology, Department of Internal Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| |
Collapse
|
50
|
Xiao J, Borné Y, Gottsäter A, Pan J, Acosta S, Engström G. Red Cell Distribution Width is Associated with Future Incidence of Abdominal Aortic Aneurysm in a Population-Based Cohort Study. Sci Rep 2020; 10:7230. [PMID: 32350354 PMCID: PMC7190826 DOI: 10.1038/s41598-020-64331-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2020] [Accepted: 04/15/2020] [Indexed: 12/11/2022] Open
Abstract
Red cell distribution width (RDW) has been suggested to have a predictive potential for several cardiovascular diseases, but its association with abdominal aortic aneurysm (AAA) is unknown. We examined whether RDW is associated with the risk of AAA among 27,260 individuals from the population-based Malmö Diet and Cancer Study cohort. Data of baseline characteristics were collected during 1991–1996. Cox regression was used to estimate hazard ratios (HR) with 95% confidence intervals (CI) for AAA across quartiles of RDW. During a median follow-up of 21.7 years, 491 subjects developed AAA. After adjustment for other confounding factors, participants in the highest quartile of RDW experienced 61% increased risk of AAA as compared to those with the lowest quartile (HR = 1.61, CI = 1.20, 2.12). RDW showed similar relationship with severe (i.e. ruptured or surgically repaired) AAA or non-severe AAA (adjusted HR 1.58 and 1.60, respectively). The observed association between RDW and AAA risk was significant in current smokers (adjusted HR = 1.68, CI = 1.18, 2.38) but not in former smokers (adjusted HR = 1.13, CI = 0.72, 1.79), or never-smokers (adjusted HR = 1.77, CI = 0.74, 4.22). Elevated RDW is associated with increased future incidence of AAA, however the causal and pathophysiological mechanisms remain to be explored.
Collapse
Affiliation(s)
- Jun Xiao
- Department of Cardiovascular Surgery, Fujian Medical University Union Hospital, Fuzhou, China. .,Department of Clinical Sciences in Malmö, Lund University, Malmö, Sweden.
| | - Yan Borné
- Department of Clinical Sciences in Malmö, Lund University, Malmö, Sweden
| | - Anders Gottsäter
- Department of Clinical Sciences in Malmö, Lund University, Malmö, Sweden.,Vascular Centre, Department of Cardiothoracic and Vascular Surgery, Skåne University Hospital, Malmö, Sweden
| | - Jingxue Pan
- Department of Clinical Sciences in Malmö, Lund University, Malmö, Sweden
| | - Stefan Acosta
- Department of Clinical Sciences in Malmö, Lund University, Malmö, Sweden.,Vascular Centre, Department of Cardiothoracic and Vascular Surgery, Skåne University Hospital, Malmö, Sweden
| | - Gunnar Engström
- Department of Clinical Sciences in Malmö, Lund University, Malmö, Sweden.
| |
Collapse
|