1
|
Zhang L, Chen S, Ning M, Guo S, Wen D, Wang H, Sun Y, Yang G, Wang Y, Xue S. Tea Polyphenol-Derived Carbon Dots Alleviate Abdominal Aortic Aneurysm Progression by Mitigating Oxidative Stress and Ferroptosis. ACS APPLIED BIO MATERIALS 2025; 8:688-703. [PMID: 39737545 DOI: 10.1021/acsabm.4c01549] [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] [Indexed: 01/01/2025]
Abstract
Abdominal aortic aneurysm (AAA) is a cardiovascular disease with potentially fatal consequences, yet effective therapies to prevent its progression remain unavailable. Oxidative stress is associated with AAA development. Carbon dots have reactive oxygen species-scavenging activity, while green tea extract exhibits robust antioxidant properties. However, the potential of green tea derived carbon dots in mitigating AAA progression has not been fully elucidated. In this study, tea polyphenol carbon dots (TP-CDs) were synthesized via hydrothermal methods and characterized for their antioxidant properties. The antioxidant effects of TP-CDs were evaluated, and TP-CDs' impact on phenotypic transformation, oxidative stress, apoptosis and ferroptosis was investigated comprehensively in an Ang II-induced AAA model, employing techniques such as Western blotting, flow cytometry, and immunohistochemistry. The results revealed that TP-CDs effectively alleviated oxidative stress induced by Ang II stimulation, thereby inhibiting phenotypic transformation, apoptosis, and ferroptosis in vivo. Furthermore, treatment with TP-CDs significantly attenuated AAA progression in a mouse AAA model. Overall, these findings demonstrate that TP-CDs reduced reactive oxygen species levels in the microenvironment and alleviated the progression of AAA, offering a promising therapeutic strategy for this condition.
Collapse
Affiliation(s)
- Luzheng Zhang
- Department of Cardiovascular Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, P. R. China
| | - Shuyang Chen
- Department of Pulmonary and Critical Care Medicine, Zhongshan Hospital, Fudan University, Shanghai 200032, P. R. China
| | - Mengling Ning
- MOE Key Laboratory of Laser Life Science and SATCM Third Grade Laboratory of Chinese Medicine and Photonics Technology, College of Biophotonics, South China Normal University, Guangzhou 510631, P. R. China
| | - Suxiang Guo
- Department of Cardiovascular Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, P. R. China
| | - Dezhong Wen
- Department of Cardiovascular Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, P. R. China
| | - Heng Wang
- Department of Cardiovascular Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, P. R. China
| | - Yujin Sun
- Department of Cardiovascular Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, P. R. China
| | - Guangdong Yang
- Cardiovascular and Metabolic Research Unit, Laurentian University, Sudbury, OntarioP3B 2R9, Canada
| | - Yuehong Wang
- State Key Laboratory for Oncogenes and Related Genes, Shanghai Cancer Institute Department of Cardiology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, 160 Pujian Road, Shanghai 200127, P. R. China
| | - Song Xue
- Department of Cardiovascular Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, P. R. China
| |
Collapse
|
2
|
Wang L, Zhang Y, Mao C, Li X. Enhancing Exosomal Delivery to Abdominal Aortic Aneurysms using Magnetically Responsive Chemotactic Nanomotors for Elastic Matrix Regenerative Repair. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2405085. [PMID: 39429209 PMCID: PMC11633499 DOI: 10.1002/advs.202405085] [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: 05/11/2024] [Revised: 09/13/2024] [Indexed: 10/22/2024]
Abstract
Abdominal aortic aneurysms (AAAs) involve localized dilation of the abdominal aorta, with the reversal of this condition being significantly limited by the inherently poor and abnormal regenerative repair of the aortic elastic matrix. Mesenchymal stem cell exosomes (MSCEs) are promising regenerative tools; however, achieving precise targeting of AAA with MSCEs is challenging owing to the high blood flow in the arterial system. In this study, an engineered exosomal nanomotor is developed for magnetic and chemical propulsion. The results demonstrate that this nanomotor effectively enhances the delivery of MSCEs to the AAA through magnetic field navigation and catalase-induced chemotaxis. The nanomotor significantly enhances the elastic matrix repair, reduces oxidative stress, and activates the PI3K/Akt pathway, leading to aneurysm shrinkage and reversal. In addition, the nanomotor possesses magnetic resonance imaging capabilities. The use of this nanomotor offers a novel, targeted drug delivery system in a rat model of AAA and holds promise as a potential therapeutic option for this condition.
Collapse
Affiliation(s)
- Lulu Wang
- Department of Vascular SurgeryNanjing Drum Tower HospitalAffiliated Hospital of Medical SchoolNanjing UniversityNanjingJiangsu210008China
| | - Yao Zhang
- National and Local Joint Engineering Research Center of Biomedical Functional MaterialsSchool of Chemistry and Materials ScienceNanjing Normal UniversityNanjingJiangsu210023China
| | - Chun Mao
- Department of Vascular SurgeryNanjing Drum Tower HospitalAffiliated Hospital of Medical SchoolNanjing UniversityNanjingJiangsu210008China
- National and Local Joint Engineering Research Center of Biomedical Functional MaterialsSchool of Chemistry and Materials ScienceNanjing Normal UniversityNanjingJiangsu210023China
| | - Xiaoqiang Li
- Department of Vascular SurgeryNanjing Drum Tower HospitalAffiliated Hospital of Medical SchoolNanjing UniversityNanjingJiangsu210008China
| |
Collapse
|
3
|
Martin-Blazquez A, Martin-Lorenzo M, Santiago-Hernandez A, Heredero A, Donado A, Lopez JA, Anfaiha-Sanchez M, Ruiz-Jimenez R, Esteban V, Vazquez J, Aldamiz-Echevarria G, Alvarez-Llamas G. Analysis of Vascular Smooth Muscle Cells from Thoracic Aortic Aneurysms Reveals DNA Damage and Cell Cycle Arrest as Hallmarks in Bicuspid Aortic Valve Patients. J Proteome Res 2024; 23:3012-3024. [PMID: 38594816 PMCID: PMC11301675 DOI: 10.1021/acs.jproteome.3c00649] [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: 10/04/2023] [Revised: 02/26/2024] [Accepted: 03/24/2024] [Indexed: 04/11/2024]
Abstract
Thoracic aortic aneurysm (TAA) is mainly sporadic and with higher incidence in the presence of a bicuspid aortic valve (BAV) for unknown reasons. The lack of drug therapy to delay TAA progression lies in the limited knowledge of pathophysiology. We aimed to identify the molecular hallmarks that differentiate the aortic dilatation associated with BAV and tricuspid aortic valve (TAV). Aortic vascular smooth muscle cells (VSMCs) isolated from sporadic TAA patients with BAV or TAV were analyzed by mass spectrometry. DNA oxidative damage assay and cell cycle profiling were performed in three independent cohorts supporting proteomics data. The alteration of secreted proteins was confirmed in plasma. Stress phenotype, oxidative stress, and enhanced DNA damage response (increased S-phase arrest and apoptosis) were found in BAV-TAA patients. The increased levels of plasma C1QTNF5, LAMA2, THSB3, and FAP confirm the enhanced stress in BAV-TAA. Plasma FAP and BGN point to an increased inflammatory condition in TAV. The arterial wall of BAV patients shows a limited capacity to counteract drivers of sporadic TAA. The molecular pathways identified support the need of differential molecular diagnosis and therapeutic approaches for BAV and TAV patients, showing specific markers in plasma which may serve to monitor therapy efficacy.
Collapse
Affiliation(s)
- Ariadna Martin-Blazquez
- Immunology
Department, IIS-Fundación Jiménez
Díaz, Fundación Jiménez Díaz Hospital-UAM, 28040 Madrid, Spain
| | - Marta Martin-Lorenzo
- Immunology
Department, IIS-Fundación Jiménez
Díaz, Fundación Jiménez Díaz Hospital-UAM, 28040 Madrid, Spain
| | | | - Angeles Heredero
- Cardiac
Surgery Service, Fundación Jiménez
Díaz Hospital-UAM, 28040 Madrid, Spain
| | - Alicia Donado
- Cardiac
Surgery Service, Fundación Jiménez
Díaz Hospital-UAM, 28040 Madrid, Spain
| | - Juan A Lopez
- Laboratory
of Cardiovascular Proteomics, Centro Nacional
de Investigaciones Cardiovasculares (CNIC), 28029 Madrid, Spain
- CIBER
de Enfermedades Cardiovasculares (CIBERCV), 28029 Madrid, Spain
| | - Miriam Anfaiha-Sanchez
- Immunology
Department, IIS-Fundación Jiménez
Díaz, Fundación Jiménez Díaz Hospital-UAM, 28040 Madrid, Spain
| | - Rocio Ruiz-Jimenez
- Immunology
Department, IIS-Fundación Jiménez
Díaz, Fundación Jiménez Díaz Hospital-UAM, 28040 Madrid, Spain
| | - Vanesa Esteban
- Department
of Allergy and Immunology, IIS-Fundación
Jiménez Díaz, Fundación Jiménez Díaz
Hospital-UAM, 28040 Madrid, Spain
- Faculty
of Medicine and Biomedicine, Alfonso X El
Sabio University, 28691 Madrid, Spain
| | - Jesus Vazquez
- Laboratory
of Cardiovascular Proteomics, Centro Nacional
de Investigaciones Cardiovasculares (CNIC), 28029 Madrid, Spain
- CIBER
de Enfermedades Cardiovasculares (CIBERCV), 28029 Madrid, Spain
| | | | - Gloria Alvarez-Llamas
- Immunology
Department, IIS-Fundación Jiménez
Díaz, Fundación Jiménez Díaz Hospital-UAM, 28040 Madrid, Spain
- RICORS2040, Fundación Jiménez Díaz, 28040 Madrid, Spain
- Department
of Biochemistry and Molecular Biology, Complutense
University, 28040 Madrid, Spain
| |
Collapse
|
4
|
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
|
5
|
Kazaleh M, Gioscia-Ryan R, Ailawadi G, Salmon M. Oxidative Stress and the Pathogenesis of Aortic Aneurysms. Biomedicines 2023; 12:3. [PMID: 38275364 PMCID: PMC10813769 DOI: 10.3390/biomedicines12010003] [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: 11/03/2023] [Revised: 12/13/2023] [Accepted: 12/15/2023] [Indexed: 01/27/2024] Open
Abstract
Aortic aneurysms are responsible for significant morbidity and mortality. Despite their clinical significance, there remain critical knowledge gaps in the pathogenesis of aneurysm disease and the mechanisms involved in aortic rupture. Recent studies have drawn attention to the role of reactive oxygen species (ROS) and their down-stream effectors in chronic cardiovascular diseases and specifically in the pathogenesis of aortic aneurysm formation. This review will discuss current mechanisms of ROS in mediating aortic aneurysms, the failure of endogenous antioxidant systems in chronic vascular diseases, and their relation to the development of aortic aneurysms.
Collapse
Affiliation(s)
- Matthew Kazaleh
- Department of Cardiac Surgery, Michigan Medicine, University of Michigan, Ann Arbor, MI 48109, USA; (M.K.); (G.A.)
| | - Rachel Gioscia-Ryan
- Department of Anesthesiology, Michigan Medicine, University of Michigan, Ann Arbor, MI 48109, USA;
| | - Gorav Ailawadi
- Department of Cardiac Surgery, Michigan Medicine, University of Michigan, Ann Arbor, MI 48109, USA; (M.K.); (G.A.)
- Frankel Cardiovascular Center, Michigan Medicine, University of Michigan, Ann Arbor, MI 48109, USA
| | - Morgan Salmon
- Department of Cardiac Surgery, Michigan Medicine, University of Michigan, Ann Arbor, MI 48109, USA; (M.K.); (G.A.)
- Frankel Cardiovascular Center, Michigan Medicine, University of Michigan, Ann Arbor, MI 48109, USA
| |
Collapse
|
6
|
The mechanism and therapy of aortic aneurysms. Signal Transduct Target Ther 2023; 8:55. [PMID: 36737432 PMCID: PMC9898314 DOI: 10.1038/s41392-023-01325-7] [Citation(s) in RCA: 53] [Impact Index Per Article: 26.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2022] [Revised: 12/15/2022] [Accepted: 01/14/2023] [Indexed: 02/05/2023] Open
Abstract
Aortic aneurysm is a chronic aortic disease affected by many factors. Although it is generally asymptomatic, it poses a significant threat to human life due to a high risk of rupture. Because of its strong concealment, it is difficult to diagnose the disease in the early stage. At present, there are no effective drugs for the treatment of aneurysms. Surgical intervention and endovascular treatment are the only therapies. Although current studies have discovered that inflammatory responses as well as the production and activation of various proteases promote aortic aneurysm, the specific mechanisms remain unclear. Researchers are further exploring the pathogenesis of aneurysms to find new targets for diagnosis and treatment. To better understand aortic aneurysm, this review elaborates on the discovery history of aortic aneurysm, main classification and clinical manifestations, related molecular mechanisms, clinical cohort studies and animal models, with the ultimate goal of providing insights into the treatment of this devastating disease. The underlying problem with aneurysm disease is weakening of the aortic wall, leading to progressive dilation. If not treated in time, the aortic aneurysm eventually ruptures. An aortic aneurysm is a local enlargement of an artery caused by a weakening of the aortic wall. The disease is usually asymptomatic but leads to high mortality due to the risk of artery rupture.
Collapse
|
7
|
Jreije A, Medlej-Hashim M, Hajal J, Saliba Y, Chacar S, Fares N, Khouzami L. Calcitriol Supplementation Protects Against Apoptosis and Alleviates the Severity of Abdominal Aortic Aneurysm Induced by Angiotensin II and Anti-TGFβ. J Cardiovasc Transl Res 2022; 15:1340-1351. [PMID: 35445935 DOI: 10.1007/s12265-022-10254-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Accepted: 04/04/2022] [Indexed: 12/16/2022]
Abstract
The present study aims to assess the effect of vitamin D deficiency (VDD) and its supplementation on the severity of AAA in mice. AAA was induced by AngII and anti-TGF-β administration. Animals were divided into four groups: Sham, mice with AAA, mice with AAA, and VDD, and mice with AAA supplemented with calcitriol. Blood pressure, echocardiography, abdominal aortic tissues, and plasma samples were monitored for all groups. VDD was associated with enhanced activity of cleaved MMP-9 and elastin degradation and positively correlated with the severity of AAA. Calcitriol supplementation decreased the INFγ/IL-10 ratio and enhanced the Nrf2 pathway. Moreover, Cu/Zn-superoxide dismutase expression and catalase and neutral sphingomyelinase activity were exacerbated in AAA and VDD groups. Furthermore, calcitriol supplementation showed a significantly lower protein expression of caspase-8, caspase-3, Bid, and t-Bid, and prevented the apoptosis of VSMCs treated by AngII and anti-TGF-β. Calcitriol supplementation may alleviate AAA severity and could be of great interest in the clinical management of AAA. VDD enhances antioxidant enzymes activity and expression, whereas calcitriol supplementation alleviates AAA severity by re-activating Nrf2 and inhibiting apoptotic pathways.
Collapse
Affiliation(s)
- Afaf Jreije
- Laboratoire de Recherche en Physiologie Et Physiopathologie, Faculté de Médecine, Pôle Technologie Santé, Université Saint Joseph, Beirut, Lebanon
- Cellular and Molecular Physiopathologies (CAMP) Laboratory, Faculty of Sciences II, Lebanese University, Fanar, Lebanon
| | - Myrna Medlej-Hashim
- Cellular and Molecular Physiopathologies (CAMP) Laboratory, Faculty of Sciences II, Lebanese University, Fanar, Lebanon
| | - Joelle Hajal
- Laboratoire de Recherche en Physiologie Et Physiopathologie, Faculté de Médecine, Pôle Technologie Santé, Université Saint Joseph, Beirut, Lebanon
| | - Youakim Saliba
- Laboratoire de Recherche en Physiologie Et Physiopathologie, Faculté de Médecine, Pôle Technologie Santé, Université Saint Joseph, Beirut, Lebanon
| | - Stephanie Chacar
- Laboratoire de Recherche en Physiologie Et Physiopathologie, Faculté de Médecine, Pôle Technologie Santé, Université Saint Joseph, Beirut, Lebanon
| | - Nassim Fares
- Laboratoire de Recherche en Physiologie Et Physiopathologie, Faculté de Médecine, Pôle Technologie Santé, Université Saint Joseph, Beirut, Lebanon.
| | - Lara Khouzami
- Cellular and Molecular Physiopathologies (CAMP) Laboratory, Faculty of Sciences II, Lebanese University, Fanar, Lebanon.
| |
Collapse
|
8
|
NADPH Oxidases in Aortic Aneurysms. Antioxidants (Basel) 2022; 11:antiox11091830. [PMID: 36139902 PMCID: PMC9495752 DOI: 10.3390/antiox11091830] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Revised: 09/08/2022] [Accepted: 09/12/2022] [Indexed: 12/02/2022] Open
Abstract
Abdominal aortic aneurysms (AAAs) are a progressive dilation of the infrarenal aorta and are characterized by inflammatory cell infiltration, smooth muscle cell migration and proliferation, and degradation of the extracellular matrix. Oxidative stress and the production of reactive oxygen species (ROS) have been shown to play roles in inflammatory cell infiltration, and smooth muscle cell migration and apoptosis in AAAs. In this review, we discuss the principles of nicotinamide adenine dinucleotide phosphate oxidase (NADPH oxidase/NOX) signaling and activation. We also discuss the effects of some of the major mediators of NOX signaling in AAAs. Separately, we also discuss the influence of genetic or pharmacologic inhibitors of NADPH oxidases on experimental pre-clinical AAAs. Experimental evidence suggests that NADPH oxidases may be a promising future therapeutic target for developing pharmacologic treatment strategies for halting AAA progression or rupture prevention in the management of clinical AAAs.
Collapse
|
9
|
Role of Oxidative Stress in the Pathogenesis of Atherothrombotic Diseases. Antioxidants (Basel) 2022; 11:antiox11071408. [PMID: 35883899 PMCID: PMC9312358 DOI: 10.3390/antiox11071408] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2022] [Revised: 07/12/2022] [Accepted: 07/19/2022] [Indexed: 12/04/2022] Open
Abstract
Oxidative stress is generated by the imbalance between reactive oxygen species (ROS) formation and antioxidant scavenger system’s activity. Increased ROS, such as superoxide anion, hydrogen peroxide, hydroxyl radical and peroxynitrite, likely contribute to the development and complications of atherosclerotic cardiovascular diseases (ASCVD). In genetically modified mouse models of atherosclerosis, the overexpression of ROS-generating enzymes and uncontrolled ROS formation appear to be associated with accelerated atherosclerosis. Conversely, the overexpression of ROS scavenger systems reduces or stabilizes atherosclerotic lesions, depending on the genetic background of the mouse model. In humans, higher levels of circulating biomarkers derived from the oxidation of lipids (8-epi-prostaglandin F2α, and malondialdehyde), as well as proteins (oxidized low-density lipoprotein, nitrotyrosine, protein carbonyls, advanced glycation end-products), are increased in conditions of high cardiovascular risk or overt ASCVD, and some oxidation biomarkers have been reported as independent predictors of ASCVD in large observational cohorts. In animal models, antioxidant supplementation with melatonin, resveratrol, Vitamin E, stevioside, acacetin and n-polyunsaturated fatty acids reduced ROS and attenuated atherosclerotic lesions. However, in humans, evidence from large, placebo-controlled, randomized trials or prospective studies failed to show any athero-protective effect of antioxidant supplementation with different compounds in different CV settings. However, the chronic consumption of diets known to be rich in antioxidant compounds (e.g., Mediterranean and high-fish diet), has shown to reduce ASCVD over decades. Future studies are needed to fill the gap between the data and targets derived from studies in animals and their pathogenetic and therapeutic significance in human ASCVD.
Collapse
|
10
|
Amioka N, Miyoshi T, Yonezawa T, Kondo M, Akagi S, Yoshida M, Saito Y, Nakamura K, Ito H. Pemafibrate Prevents Rupture of Angiotensin II-Induced Abdominal Aortic Aneurysms. Front Cardiovasc Med 2022; 9:904215. [PMID: 35845076 PMCID: PMC9280056 DOI: 10.3389/fcvm.2022.904215] [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: 03/25/2022] [Accepted: 06/14/2022] [Indexed: 12/26/2022] Open
Abstract
Background Abdominal aortic aneurysm (AAA) is a life-threatening disease that lacks effective preventive therapies. This study aimed to evaluate the effect of pemafibrate, a selective peroxisome proliferator-activated receptor alpha (PPARα) agonist, on AAA formation and rupture. Methods Experimental AAA was induced by subcutaneous angiotensin II (AngII) infusion in ApoE - / - mice for 4 weeks. Pemafibrate (0.1 mg/kg/day) was administered orally. Dihydroethidium staining was used to evaluate the reactive oxygen species (ROS). Results The size of the AngII-induced AAA did not differ between pemafibrate- and vehicle-treated groups. However, a decreased mortality rate due to AAA rupture was observed in pemafibrate-treated mice. Pemafibrate ameliorated AngII-induced ROS and reduced the mRNA expression of interleukin-6 and tumor necrosis factor-α in the aortic wall. Gelatin zymography analysis demonstrated significant inhibition of matrix metalloproteinase-2 activity by pemafibrate. AngII-induced ROS production in human vascular smooth muscle cells was inhibited by pre-treatment with pemafibrate and was accompanied by an increase in catalase activity. Small interfering RNA-mediated knockdown of catalase or PPARα significantly attenuated the anti-oxidative effect of pemafibrate. Conclusion Pemafibrate prevented AAA rupture in a murine model, concomitant with reduced ROS, inflammation, and extracellular matrix degradation in the aortic wall. The protective effect against AAA rupture was partly mediated by the anti-oxidative effect of catalase induced by pemafibrate in the smooth muscle cells.
Collapse
Affiliation(s)
- Naofumi Amioka
- Department of Cardiovascular Medicine, Faculty of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Toru Miyoshi
- Department of Cardiovascular Medicine, Faculty of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Tomoko Yonezawa
- Department of Molecular Biology and Biochemistry, Faculty of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Megumi Kondo
- Department of Cardiovascular Medicine, Faculty of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Satoshi Akagi
- Department of Cardiovascular Medicine, Faculty of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Masashi Yoshida
- Department of Cardiovascular Medicine, Faculty of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Yukihiro Saito
- Department of Cardiovascular Medicine, Faculty of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Kazufumi Nakamura
- Department of Cardiovascular Medicine, Faculty of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Hiroshi Ito
- Department of Cardiovascular Medicine, Faculty of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| |
Collapse
|
11
|
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
|
12
|
Correlation Between Proteolytic Activity and Abdominal Aortic Aneurysm Wall Morphology with Intraluminal Thrombus Volume. Ann Vasc Surg 2022; 87:487-494. [PMID: 35779804 DOI: 10.1016/j.avsg.2022.05.039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2022] [Revised: 05/19/2022] [Accepted: 05/26/2022] [Indexed: 11/24/2022]
Abstract
BACKGROUND The aim of this study was to examine the influence of intraluminal thrombus (ILT) volume on the level of proteolytic activity and the content of abdominal aortic aneurysm (AAA) wall. METHODS The research was designed as a cross-sectional study at the Clinic for Vascular and Endovascular Surgery, Clinical Center of Serbia in the period from April 2017 to February 2018. During this period, a total of 155 patients with asymptomatic AAA underwent open surgical treatment and 50 were included in the study based on inclusion and exclusion criteria. Before surgery, patients included in the study were examined by MRI. During the operation, samples of ILT and AAA wall were taken for biochemical analysis. RESULTS A statistically significant correlation was found between the volume of the ILT and largest AAA diameter (ρ = 0.56; P < 0.001). The correlation of the ILT volume on the anterior wall and the concentration of MMP-9, MMP-2 and NE/ELA in the wall did not find statistical significance. Also, no statistically significant association was found between the volume of ILT and the concentration of ECM proteins (collagen type 3, elastin, proteoglycan) in the corresponding part of the wall. The association of ILT volume with MDA was also of no statistical significance. There was a positive statistical significance found in correlation of volume of ILT and catalase activity in the wall of AAA (ρ = 0.28, P = 0.049). CONCLUSIONS The volume of ILT in the aneurysmal sac seemed not to affect the level of proteolytic activity and the content of the aneurysm wall. However, a positive correlation was found between the ILT and the catalase activity. The effect of ILT on the aneurysm wall and its role in the progression of aneurysmal disease should be examined in future studies.
Collapse
|
13
|
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
|
14
|
Goyal P, Mishra V, Dhamija I, Kumar N, Kumar S. Immobilization of catalase on functionalized magnetic nanoparticles: a statistical approach. 3 Biotech 2022; 12:108. [PMID: 35462953 PMCID: PMC8994807 DOI: 10.1007/s13205-022-03173-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Accepted: 03/19/2022] [Indexed: 11/26/2022] Open
Abstract
Magnetic nanoparticles (MNPs) Fe3O4, by virtue of easily modifiable surface, high surface-to-mass ratio and super-paramagnetic properties, are one of suitable candidates for the enzyme immobilization. Optimization of five important variables viz. concentration of 3-aminopropyl-tri-ethoxy-silane (APTES), glutaraldehyde (GA) and enzyme, time and temperature of loading was carried out using central composite type of experimental design without blocks giving 50 experiments including eight replicates at the central point. Characterization, stability and reusability studies were also carried out with optimized preparation. Results established the correlation between observed and response surface method (RSM) equation envisaged value (R 2 0.99, 0.97 and 0.98 for enzyme's activity, its loading over MNPs and corresponding specific activity, respectively. The predicted values suggested by RSM equation were 64.00 mM of APTES, 10.97 µL of GA, 14.50 mg mL-1 of enzyme for 67 min at 22.6 °C, resulted in activity 32.1 U mg-1 MNPs, while specific activity was 97.7 U mg-1. Transmission electron microscopy (TEM) showed the sizes of MNPs (10.5 ± 1.7 nm), APTES-MNPs (10.23 ± 1.74 nm), GA-APTES-MNPs (11.84 ± 1.49 nm) and Catalase-GA-APTES-MNPs (13.32 ± 2.74 nm) were statistically similar. The enzyme MNPs preparation retained 81.65% activity after 144 h at 4 °C (free enzyme retained 7.87%) and 64.34% activity after 20 reuse cycles. Statistical optimized MNPs-based catalase preparation with high activity and magnetic strength was stable and can be used for further studies related to its application as analytical recyclable enzyme or magnetically oriented delivery in the body. Supplementary Information The online version contains supplementary material available at 10.1007/s13205-022-03173-8.
Collapse
Affiliation(s)
- Pankaj Goyal
- Department of Bioproducts and Biosystems, School of Chemical Engineering, Aalto University, 00076 Helsinki, Finland
- Department of Biotechnology, Dr. B. R. Ambedkar, National Institute of Technology, Jalandhar, Punjab 144011 India
| | - Vartika Mishra
- Department of Biotechnology, Dr. B. R. Ambedkar, National Institute of Technology, Jalandhar, Punjab 144011 India
- Department of Biotechnology Engineering, Chandigarh University, Mohali, Punjab 140413 India
| | - Isha Dhamija
- Department of Pharmaceutical Sciences, Guru Jambheshwar University of Science and Technology, Hisar, Haryana 125001 India
| | - Neeraj Kumar
- Department of Regulatory Affairs, Department of Pharmaceuticals, Ministry of Chemicals and Fertilizers, National Institute of Pharmaceutical Education and Research, Govt. of India, NH 9, Kukatpally, Industrial Estate, Balanagar, Hyderabad, 500037 Telengana India
| | - Sandeep Kumar
- Department of Biotechnology, Dr. B. R. Ambedkar, National Institute of Technology, Jalandhar, Punjab 144011 India
- Department of Regulatory Affairs, Department of Pharmaceuticals, Ministry of Chemicals and Fertilizers, National Institute of Pharmaceutical Education and Research, Govt. of India, NH 9, Kukatpally, Industrial Estate, Balanagar, Hyderabad, 500037 Telengana India
| |
Collapse
|
15
|
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: 37] [Impact Index Per Article: 12.3] [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
|
16
|
Radovanovic J, Banjac K, Obradovic M, Isenovic ER. Antioxidant enzymes and vascular diseases. EXPLORATION OF MEDICINE 2021. [DOI: 10.37349/emed.2021.00070] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Reactive oxygen species (ROS) and reactive nitrogen species (RNS) play a fundamental role in regulating endothelial function and vascular tone in the physiological conditions of a vascular system. However, oxidative stress has detrimental effects on human health, and numerous studies confirmed that high ROS/RNS production contributes to the initiation and progression of cardiovascular diseases. The antioxidant defense has an essential role in the homeostatic functioning of the vascular endothelial system. Endogenous antioxidative defense includes various molecules and enzymes such as superoxide dismutase, catalase, glutathione reductase, and glutathione peroxidase. Together all these antioxidative enzymes are essential for defense against harmful ROS features. ROS are mainly generated from redox-active compounds involved in the mitochondrial respiratory chain. Thus, targeting antioxidative enzymes and mitochondria oxidative balance may be a promising approach for vascular diseases occurrence and treatment. This review summarized the most recent research on the regulation of antioxidative enzymes in vascular diseases.
Collapse
Affiliation(s)
- Jelena Radovanovic
- Department of Radiobiology and Molecular Genetics, “VINČA” Institute of Nuclear Sciences-National Institute of the Republic of Serbia, University of Belgrade, 522 Belgrade, Serbia
| | - Katarina Banjac
- Department of Radiobiology and Molecular Genetics, “VINČA” Institute of Nuclear Sciences-National Institute of the Republic of Serbia, University of Belgrade, 522 Belgrade, Serbia
| | - Milan Obradovic
- Department of Radiobiology and Molecular Genetics, “VINČA” Institute of Nuclear Sciences-National Institute of the Republic of Serbia, University of Belgrade, 522 Belgrade, Serbia
| | - Esma R. Isenovic
- Department of Radiobiology and Molecular Genetics, “VINČA” Institute of Nuclear Sciences-National Institute of the Republic of Serbia, University of Belgrade, 522 Belgrade, Serbia
| |
Collapse
|
17
|
LncRNA GAS5 promotes abdominal aortic aneurysm formation through regulating the miR-185-5p/ADCY7 axis. Anticancer Drugs 2021; 33:225-234. [DOI: 10.1097/cad.0000000000001090] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
18
|
Lu H, Du W, Ren L, Hamblin MH, Becker RC, Chen YE, Fan Y. Vascular Smooth Muscle Cells in Aortic Aneurysm: From Genetics to Mechanisms. J Am Heart Assoc 2021; 10:e023601. [PMID: 34796717 PMCID: PMC9075263 DOI: 10.1161/jaha.121.023601] [Citation(s) in RCA: 81] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Aortic aneurysm, including thoracic aortic aneurysm and abdominal aortic aneurysm, is the second most prevalent aortic disease following atherosclerosis, representing the ninth-leading cause of death globally. Open surgery and endovascular procedures are the major treatments for aortic aneurysm. Typically, thoracic aortic aneurysm has a more robust genetic background than abdominal aortic aneurysm. Abdominal aortic aneurysm shares many features with thoracic aortic aneurysm, including loss of vascular smooth muscle cells (VSMCs), extracellular matrix degradation and inflammation. Although there are limitations to perfectly recapitulating all features of human aortic aneurysm, experimental models provide valuable tools to understand the molecular mechanisms and test novel therapies before human clinical trials. Among the cell types involved in aortic aneurysm development, VSMC dysfunction correlates with loss of aortic wall structural integrity. Here, we discuss the role of VSMCs in aortic aneurysm development. The loss of VSMCs, VSMC phenotypic switching, secretion of inflammatory cytokines, increased matrix metalloproteinase activity, elevated reactive oxygen species, defective autophagy, and increased senescence contribute to aortic aneurysm development. Further studies on aortic aneurysm pathogenesis and elucidation of the underlying signaling pathways are necessary to identify more novel targets for treating this prevalent and clinical impactful disease.
Collapse
Affiliation(s)
- Haocheng Lu
- Department of Internal MedicineCardiovascular CenterUniversity of Michigan Medical CenterAnn ArborMI
| | - Wa Du
- Department of Cancer BiologyUniversity of Cincinnati College of MedicineCincinnatiOH
| | - Lu Ren
- Department of Cancer BiologyUniversity of Cincinnati College of MedicineCincinnatiOH
| | - Milton H. Hamblin
- Department of PharmacologyTulane University School of MedicineNew OrleansLA
| | - Richard C. Becker
- Division of Cardiovascular Health and DiseaseDepartment of Internal MedicineUniversity of Cincinnati College of MedicineCincinnatiOH
| | - Y. Eugene Chen
- Department of Internal MedicineCardiovascular CenterUniversity of Michigan Medical CenterAnn ArborMI
| | - Yanbo Fan
- Department of Cancer BiologyUniversity of Cincinnati College of MedicineCincinnatiOH
- Division of Cardiovascular Health and DiseaseDepartment of Internal MedicineUniversity of Cincinnati College of MedicineCincinnatiOH
| |
Collapse
|
19
|
Tracy EP, Hughes W, Beare JE, Rowe G, Beyer A, LeBlanc AJ. Aging-Induced Impairment of Vascular Function: Mitochondrial Redox Contributions and Physiological/Clinical Implications. Antioxid Redox Signal 2021; 35:974-1015. [PMID: 34314229 PMCID: PMC8905248 DOI: 10.1089/ars.2021.0031] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Significance: The vasculature responds to the respiratory needs of tissue by modulating luminal diameter through smooth muscle constriction or relaxation. Coronary perfusion, diastolic function, and coronary flow reserve are drastically reduced with aging. This loss of blood flow contributes to and exacerbates pathological processes such as angina pectoris, atherosclerosis, and coronary artery and microvascular disease. Recent Advances: Increased attention has recently been given to defining mechanisms behind aging-mediated loss of vascular function and development of therapeutic strategies to restore youthful vascular responsiveness. The ultimate goal aims at providing new avenues for symptom management, reversal of tissue damage, and preventing or delaying of aging-induced vascular damage and dysfunction in the first place. Critical Issues: Our major objective is to describe how aging-associated mitochondrial dysfunction contributes to endothelial and smooth muscle dysfunction via dysregulated reactive oxygen species production, the clinical impact of this phenomenon, and to discuss emerging therapeutic strategies. Pathological changes in regulation of mitochondrial oxidative and nitrosative balance (Section 1) and mitochondrial dynamics of fission/fusion (Section 2) have widespread effects on the mechanisms underlying the ability of the vasculature to relax, leading to hyperconstriction with aging. We will focus on flow-mediated dilation, endothelial hyperpolarizing factors (Sections 3 and 4), and adrenergic receptors (Section 5), as outlined in Figure 1. The clinical implications of these changes on major adverse cardiac events and mortality are described (Section 6). Future Directions: We discuss antioxidative therapeutic strategies currently in development to restore mitochondrial redox homeostasis and subsequently vascular function and evaluate their potential clinical impact (Section 7). Antioxid. Redox Signal. 35, 974-1015.
Collapse
Affiliation(s)
- Evan Paul Tracy
- Department of Physiology, University of Louisville, Louisville, Kentucky, USA
| | - William Hughes
- Department of Medicine and Physiology, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - Jason E Beare
- Cardiovascular Innovation Institute, University of Louisville, Louisville, Kentucky, USA.,Kentucky Spinal Cord Injury Research Center, University of Louisville, Louisville, Kentucky, USA
| | - Gabrielle Rowe
- Department of Physiology, University of Louisville, Louisville, Kentucky, USA
| | - Andreas Beyer
- Department of Medicine and Physiology, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - Amanda Jo LeBlanc
- Department of Physiology, University of Louisville, Louisville, Kentucky, USA.,Cardiovascular Innovation Institute, University of Louisville, Louisville, Kentucky, USA
| |
Collapse
|
20
|
Lopez‐Sanz L, Bernal S, Jimenez‐Castilla L, Prieto I, La Manna S, Gomez‐Lopez S, Blanco‐Colio LM, Egido J, Martin‐Ventura JL, Gomez‐Guerrero C. Fcγ receptor activation mediates vascular inflammation and abdominal aortic aneurysm development. Clin Transl Med 2021; 11:e463. [PMID: 34323424 PMCID: PMC8255062 DOI: 10.1002/ctm2.463] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2021] [Revised: 05/24/2021] [Accepted: 05/30/2021] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND Abdominal aortic aneurysm (AAA), a degenerative vascular pathology characterized by permanent dilation of the aorta, is considered a chronic inflammatory disease involving innate/adaptive immunity. However, the functional role of antibody-dependent immune response against antigens present in the damaged vessel remains unresolved. We hypothesized that engagement of immunoglobulin G (IgG) Fc receptors (FcγR) by immune complexes (IC) in the aortic wall contributes to AAA development. We therefore evaluated FcγR expression in AAA lesions and analysed whether inhibition of FcγR signaling molecules (γ-chain and Syk kinase) influences AAA formation in mice. METHODS FcγR gene/protein expression was assessed in human and mouse AAA tissues. Experimental AAA was induced by aortic elastase perfusion in wild-type (WT) mice and γ-chain knockout (γKO) mice (devoid of activating FcγR) in combination with macrophage adoptive transfer or Syk inhibitor treatment. To verify the mechanisms of FcγR in vitro, vascular smooth muscle cells (VSMC) and macrophages were stimulated with IgG IC. RESULTS FcγR overexpression was detected in adventitia and media layers of human and mouse AAA. Elastase-perfused γKO mice exhibited a decrease in AAA incidence, aortic dilation, elastin degradation, and VSMC loss. This was associated with (1) reduced infiltrating leukocytes and immune deposits in AAA lesions, (2) inflammatory genes and metalloproteinases downregulation, (3) redox balance restoration, and (4) converse phenotype of anti-inflammatory macrophage M2 and contractile VSMC. Adoptive transfer of FcγR-expressing macrophages aggravated aneurysm in γKO mice. In vitro, FcγR deficiency attenuated inflammatory gene expression, oxidative stress, and phenotypic switch triggered by IC. Additionally, Syk inhibition prevented IC-mediated cell responses, reduced inflammation, and mitigated AAA formation. CONCLUSION Our findings provide insight into the role and mechanisms mediating IgG-FcγR-associated inflammation and aortic wall injury in AAA, which might represent therapeutic targets against AAA disease.
Collapse
MESH Headings
- Animals
- Antigen-Antibody Complex/adverse effects
- Aorta, Abdominal/metabolism
- Aorta, Abdominal/pathology
- Aortic Aneurysm, Abdominal/pathology
- Aortic Aneurysm, Abdominal/prevention & control
- Disease Models, Animal
- Humans
- Immunoglobulin gamma-Chains/genetics
- Immunoglobulin gamma-Chains/metabolism
- Inflammation/metabolism
- Inflammation/pathology
- Macrophages/cytology
- Macrophages/immunology
- Macrophages/metabolism
- Male
- Matrix Metalloproteinases/genetics
- Matrix Metalloproteinases/metabolism
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Muscle, Smooth, Vascular/cytology
- Muscle, Smooth, Vascular/metabolism
- Niacinamide/analogs & derivatives
- Niacinamide/therapeutic use
- Oxidative Stress
- Pancreatic Elastase/adverse effects
- Pyrimidines/therapeutic use
- Receptors, IgG/genetics
- Receptors, IgG/metabolism
- Syk Kinase/antagonists & inhibitors
- Syk Kinase/metabolism
Collapse
Affiliation(s)
- Laura Lopez‐Sanz
- Renal, Vascular and Diabetes Research LabIIS‐Fundacion Jimenez Diaz (IIS‐FJD)MadridSpain
- Universidad Autonoma de Madrid (UAM)MadridSpain
- Spanish Biomedical Research Centre in Diabetes and Associated Metabolic Disorders (CIBERDEM)MadridSpain
| | - Susana Bernal
- Renal, Vascular and Diabetes Research LabIIS‐Fundacion Jimenez Diaz (IIS‐FJD)MadridSpain
- Universidad Autonoma de Madrid (UAM)MadridSpain
- Spanish Biomedical Research Centre in Diabetes and Associated Metabolic Disorders (CIBERDEM)MadridSpain
| | - Luna Jimenez‐Castilla
- Renal, Vascular and Diabetes Research LabIIS‐Fundacion Jimenez Diaz (IIS‐FJD)MadridSpain
- Universidad Autonoma de Madrid (UAM)MadridSpain
- Spanish Biomedical Research Centre in Diabetes and Associated Metabolic Disorders (CIBERDEM)MadridSpain
| | - Ignacio Prieto
- Renal, Vascular and Diabetes Research LabIIS‐Fundacion Jimenez Diaz (IIS‐FJD)MadridSpain
- Spanish Biomedical Research Centre in Diabetes and Associated Metabolic Disorders (CIBERDEM)MadridSpain
| | - Sara La Manna
- Renal, Vascular and Diabetes Research LabIIS‐Fundacion Jimenez Diaz (IIS‐FJD)MadridSpain
- Universidad Autonoma de Madrid (UAM)MadridSpain
| | | | - Luis Miguel Blanco‐Colio
- Renal, Vascular and Diabetes Research LabIIS‐Fundacion Jimenez Diaz (IIS‐FJD)MadridSpain
- Spanish Biomedical Research Centre in Cardiovascular Diseases (CIBERCV)MadridSpain
| | - Jesus Egido
- Renal, Vascular and Diabetes Research LabIIS‐Fundacion Jimenez Diaz (IIS‐FJD)MadridSpain
- Universidad Autonoma de Madrid (UAM)MadridSpain
- Spanish Biomedical Research Centre in Diabetes and Associated Metabolic Disorders (CIBERDEM)MadridSpain
| | - Jose Luis Martin‐Ventura
- Renal, Vascular and Diabetes Research LabIIS‐Fundacion Jimenez Diaz (IIS‐FJD)MadridSpain
- Universidad Autonoma de Madrid (UAM)MadridSpain
- Spanish Biomedical Research Centre in Cardiovascular Diseases (CIBERCV)MadridSpain
| | - Carmen Gomez‐Guerrero
- Renal, Vascular and Diabetes Research LabIIS‐Fundacion Jimenez Diaz (IIS‐FJD)MadridSpain
- Universidad Autonoma de Madrid (UAM)MadridSpain
- Spanish Biomedical Research Centre in Diabetes and Associated Metabolic Disorders (CIBERDEM)MadridSpain
| |
Collapse
|
21
|
Portelli SS, Hambly BD, Jeremy RW, Robertson EN. Oxidative stress in genetically triggered thoracic aortic aneurysm: role in pathogenesis and therapeutic opportunities. Redox Rep 2021; 26:45-52. [PMID: 33715602 PMCID: PMC7971305 DOI: 10.1080/13510002.2021.1899473] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Background: The primary objective of this review was to explore the contribution of oxidative stress to the pathogenesis of genetically-triggered thoracic aortic aneurysm (TAA). Genetically-triggered TAAs manifest substantial variability in onset, progression, and risk of aortic dissection, posing a significant clinical management challenge. There is a need for non-invasive biomarkers that predict the natural course of TAA and therapeutics that prevent aneurysm progression. Methods: An online systematic search was conducted within PubMed, MEDLINE, Scopus and ScienceDirect databases using keywords including: oxidative stress, ROS, nitrosative stress, genetically triggered thoracic aortic aneurysm, aortic dilatation, aortic dissection, Marfan syndrome, Bicuspid Aortic Valve, familial TAAD, Loeys Dietz syndrome, and Ehlers Danlos syndrome. Results: There is extensive evidence of oxidative stress and ROS imbalance in genetically triggered TAA. Sources of ROS imbalance are variable but include dysregulation of redox mediators leading to either insufficient ROS removal or increased ROS production. Therapeutic exploitation of redox mediators is being explored in other cardiovascular conditions, with potential application to TAA warranting further investigation. Conclusion: Oxidative stress occurs in genetically triggered TAA, but the precise contribution of ROS to pathogenesis remains incompletely understood. Further research is required to define causative pathological relationships in order to develop therapeutic options.
Collapse
Affiliation(s)
- Stefanie S Portelli
- Discipline of Pathology and Charles Perkins Centre, The University of Sydney, Sydney, Australia
| | - Brett D Hambly
- Discipline of Pathology and Charles Perkins Centre, The University of Sydney, Sydney, Australia
| | - Richmond W Jeremy
- Cardiology Department, Royal Prince Alfred Hospital, Sydney, Australia
| | - Elizabeth N Robertson
- Discipline of Pathology and Charles Perkins Centre, The University of Sydney, Sydney, Australia.,Cardiology Department, Royal Prince Alfred Hospital, Sydney, Australia
| |
Collapse
|
22
|
Lee SE, Park YS. The Emerging Roles of Antioxidant Enzymes by Dietary Phytochemicals in Vascular Diseases. Life (Basel) 2021; 11:life11030199. [PMID: 33806594 PMCID: PMC8001043 DOI: 10.3390/life11030199] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Revised: 02/26/2021] [Accepted: 02/26/2021] [Indexed: 12/21/2022] Open
Abstract
Vascular diseases are major causes of death worldwide, causing pathologies including diabetes, atherosclerosis, and chronic obstructive pulmonary disease (COPD). Exposure of the vascular system to a variety of stressors and inducers has been implicated in the development of various human diseases, including chronic inflammatory diseases. In the vascular wall, antioxidant enzymes form the first line of defense against oxidative stress. Recently, extensive research into the beneficial effects of phytochemicals has been conducted; phytochemicals are found in commonly used spices, fruits, and herbs, and are used to prevent various pathologic conditions, including vascular diseases. The present review aims to highlight the effects of dietary phytochemicals role on antioxidant enzymes in vascular diseases.
Collapse
|
23
|
Cooper HA, Cicalese S, Preston KJ, Kawai T, Okuno K, Choi ET, Kasahara S, Uchida HA, Otaka N, Scalia R, Rizzo V, Eguchi S. Targeting mitochondrial fission as a potential therapeutic for abdominal aortic aneurysm. Cardiovasc Res 2021; 117:971-982. [PMID: 32384150 PMCID: PMC7898955 DOI: 10.1093/cvr/cvaa133] [Citation(s) in RCA: 58] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/27/2019] [Revised: 04/15/2020] [Accepted: 04/30/2020] [Indexed: 11/12/2022] Open
Abstract
AIMS Angiotensin II (AngII) is a potential contributor to the development of abdominal aortic aneurysm (AAA). In aortic vascular smooth muscle cells (VSMCs), exposure to AngII induces mitochondrial fission via dynamin-related protein 1 (Drp1). However, pathophysiological relevance of mitochondrial morphology in AngII-associated AAA remains unexplored. Here, we tested the hypothesis that mitochondrial fission is involved in the development of AAA. METHODS AND RESULTS Immunohistochemistry was performed on human AAA samples and revealed enhanced expression of Drp1. In C57BL6 mice treated with AngII plus β-aminopropionitrile, AAA tissue also showed an increase in Drp1 expression. A mitochondrial fission inhibitor, mdivi1, attenuated AAA size, associated aortic pathology, Drp1 protein induction, and mitochondrial fission but not hypertension in these mice. Moreover, western-blot analysis showed that induction of matrix metalloproteinase-2, which precedes the development of AAA, was blocked by mdivi1. Mdivi1 also reduced the development of AAA in apolipoprotein E-deficient mice infused with AngII. As with mdivi1, Drp1+/- mice treated with AngII plus β-aminopropionitrile showed a decrease in AAA compared to control Drp1+/+ mice. In abdominal aortic VSMCs, AngII induced phosphorylation of Drp1 and mitochondrial fission, the latter of which was attenuated with Drp1 silencing as well as mdivi1. AngII also induced vascular cell adhesion molecule-1 expression and enhanced leucocyte adhesion and mitochondrial oxygen consumption in smooth muscle cells, which were attenuated with mdivi1. CONCLUSION These data indicate that Drp1 and mitochondrial fission play salient roles in AAA development, which likely involves mitochondrial dysfunction and inflammatory activation of VSMCs.
Collapse
MESH Headings
- Aminopropionitrile
- Angiotensin II
- Animals
- Anti-Inflammatory Agents/pharmacology
- Aorta, Abdominal/drug effects
- Aorta, Abdominal/metabolism
- Aorta, Abdominal/pathology
- Aortic Aneurysm, Abdominal/chemically induced
- Aortic Aneurysm, Abdominal/metabolism
- Aortic Aneurysm, Abdominal/pathology
- Aortic Aneurysm, Abdominal/prevention & control
- Case-Control Studies
- Cell Adhesion/drug effects
- Cells, Cultured
- Disease Models, Animal
- Dynamins/genetics
- Dynamins/metabolism
- Humans
- Leukocytes/drug effects
- Leukocytes/metabolism
- Male
- Mice, Inbred C57BL
- Mice, Knockout, ApoE
- Mitochondria, Muscle/drug effects
- Mitochondria, Muscle/genetics
- Mitochondria, Muscle/metabolism
- Mitochondria, Muscle/pathology
- Mitochondrial Dynamics/drug effects
- Muscle, Smooth, Vascular/drug effects
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/pathology
- Myocytes, Smooth Muscle/drug effects
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/pathology
- Oxygen Consumption/drug effects
- Phosphorylation
- Quinazolinones/pharmacology
- Mice
Collapse
Affiliation(s)
- Hannah A Cooper
- Cardiovascular Research Center, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA
| | - Stephanie Cicalese
- Cardiovascular Research Center, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA
| | - Kyle J Preston
- Cardiovascular Research Center, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA
| | - Tatsuo Kawai
- Cardiovascular Research Center, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA
| | - Keisuke Okuno
- Cardiovascular Research Center, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA
| | - Eric T Choi
- Department of Surgery, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA
| | - Shingo Kasahara
- Department of Cardiovascular Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8558, Japan
| | - Haruhito A Uchida
- Department of Chronic Kidney Disease and Cardiovascular Disease, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8558, Japan
| | - Nozomu Otaka
- Department of Nephrology, Rheumatology, Endocrinology and Metabolism, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8558, Japan
| | - Rosario Scalia
- Cardiovascular Research Center, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA
| | - Victor Rizzo
- Cardiovascular Research Center, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA
| | - Satoru Eguchi
- Cardiovascular Research Center, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA
| |
Collapse
|
24
|
Costa TJ, Barros PR, Arce C, Santos JD, da Silva-Neto J, Egea G, Dantas AP, Tostes RC, Jiménez-Altayó F. The homeostatic role of hydrogen peroxide, superoxide anion and nitric oxide in the vasculature. Free Radic Biol Med 2021; 162:615-635. [PMID: 33248264 DOI: 10.1016/j.freeradbiomed.2020.11.021] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Revised: 11/08/2020] [Accepted: 11/19/2020] [Indexed: 02/07/2023]
Abstract
Reactive oxygen and nitrogen species are produced in a wide range of physiological reactions that, at low concentrations, play essential roles in living organisms. There is a delicate equilibrium between formation and degradation of these mediators in a healthy vascular system, which contributes to maintaining these species under non-pathological levels to preserve normal vascular functions. Antioxidants scavenge reactive oxygen and nitrogen species to prevent or reduce damage caused by excessive oxidation. However, an excessive reductive environment induced by exogenous antioxidants may disrupt redox balance and lead to vascular pathology. This review summarizes the main aspects of free radical biochemistry (formation, sources and elimination) and the crucial actions of some of the most biologically relevant and well-characterized reactive oxygen and nitrogen species (hydrogen peroxide, superoxide anion and nitric oxide) in the physiological regulation of vascular function, structure and angiogenesis. Furthermore, current preclinical and clinical evidence is discussed on how excessive removal of these crucial responses by exogenous antioxidants (vitamins and related compounds, polyphenols) may perturb vascular homeostasis. The aim of this review is to provide information of the crucial physiological roles of oxidation in the endothelium, vascular smooth muscle cells and perivascular adipose tissue for developing safer and more effective vascular interventions with antioxidants.
Collapse
Affiliation(s)
- Tiago J Costa
- Pharmacology Department, Ribeirao Preto Medical School, University of São Paulo, Brazil.
| | | | - Cristina Arce
- Department of Biomedical Sciences, University of Barcelona School of Medicine and Health Sciences, Barcelona, Spain; Institut d'Investigacions Biomédiques August Pi i Sunyer (IDIBAPS)-University of Barcelona, Barcelona, Spain; Institut de Nanociencies i Nanotecnologia (IN2UB), University of Barcelona, Barcelona, Spain
| | | | - Júlio da Silva-Neto
- Pharmacology Department, Ribeirao Preto Medical School, University of São Paulo, Brazil
| | - Gustavo Egea
- Department of Biomedical Sciences, University of Barcelona School of Medicine and Health Sciences, Barcelona, Spain; Institut d'Investigacions Biomédiques August Pi i Sunyer (IDIBAPS)-University of Barcelona, Barcelona, Spain; Institut de Nanociencies i Nanotecnologia (IN2UB), University of Barcelona, Barcelona, Spain
| | - Ana Paula Dantas
- Institut Clínic del Tòrax, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Rita C Tostes
- Pharmacology Department, Ribeirao Preto Medical School, University of São Paulo, Brazil
| | - Francesc Jiménez-Altayó
- Department of Pharmacology, Therapeutics and Toxicology, Neuroscience Institute, School of Medicine, Universitat Autònoma de Barcelona, Barcelona, Spain.
| |
Collapse
|
25
|
Oxidant/Antioxidant Profile in the Thoracic Aneurysm of Patients with the Loeys-Dietz Syndrome. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2020; 2020:5392454. [PMID: 32273946 PMCID: PMC7128053 DOI: 10.1155/2020/5392454] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Revised: 02/25/2020] [Accepted: 03/11/2020] [Indexed: 01/10/2023]
Abstract
Patients with the Loeys-Dietz syndrome (LDS) have mutations in the TGF-βR1, TGF-βR2, and SMAD3 genes. However, little is known about the redox homeostasis in the thoracic aortic aneurysms (TAA) they develop. Here, we evaluate the oxidant/antioxidant profile in the TAA tissue from LDS patients and compare it with that in nondamaged aortic tissue from control (C) subjects. We evaluate the enzymatic activities of glutathione peroxidase (GPx), glutathione S-transferase (GST), glutathione reductase (GR), catalase (CAT), superoxide dismutase (SOD) isoforms, and thioredoxin reductase (TrxR). We also analyze some antioxidants from a nonenzymatic system such as selenium (Se), glutathione (GSH), and total antioxidant capacity (TAC). Oxidative stress markers such as lipid peroxidation and carbonylation, as well as xanthine oxidase (ORX) and nuclear factor erythroid 2-related factor 2 (Nrf2) expressions, were also evaluated. TAA from LDS patients showed a decrease in GSH, Se, TAC, GPx, GST, CAT, and TrxR. The SOD activity and ORX expressions were increased, but the Nrf2 expression was decreased. The results suggest that the redox homeostasis is altered in the TAA from LDS patients, favoring ROS overproduction that contributes to the decrease in GSH and TAC and leads to LPO and carbonylation. The decrease in Se and Nrf2 alters the activity and/or expression of some antioxidant enzymes, thus favoring a positive feedback oxidative background that contributes to the TAA formation.
Collapse
|
26
|
Abstract
Aortic aneurysms are a common vascular disease in Western populations that can involve virtually any portion of the aorta. Abdominal aortic aneurysms are much more common than thoracic aortic aneurysms and combined they account for >25 000 deaths in the United States annually. Although thoracic and abdominal aortic aneurysms share some common characteristics, including the gross anatomic appearance, alterations in extracellular matrix, and loss of smooth muscle cells, they are distinct diseases. In recent years, advances in genetic analysis, robust molecular tools, and increased availability of animal models have greatly enhanced our knowledge of the pathophysiology of aortic aneurysms. This review examines the various proposed cellular mechanisms responsible for aortic aneurysm formation and identifies opportunities for future studies.
Collapse
Affiliation(s)
- Raymundo Alain Quintana
- From the Division of Cardiology, Department of Medicine (R.A.Q., W.R.T.), Emory University School of Medicine, Atlanta, GA
| | - W Robert Taylor
- From the Division of Cardiology, Department of Medicine (R.A.Q., W.R.T.), Emory University School of Medicine, Atlanta, GA.,Wallace H. Coulter Department of Biomedical Engineering Georgia Institute of Technology (W.R.T.), Emory University School of Medicine, Atlanta, GA.,Division of Cardiology, Atlanta VA Medical Center, Decatur, GA (W.R.T.)
| |
Collapse
|
27
|
Peroxisomal Hydrogen Peroxide Metabolism and Signaling in Health and Disease. Int J Mol Sci 2019; 20:ijms20153673. [PMID: 31357514 PMCID: PMC6695606 DOI: 10.3390/ijms20153673] [Citation(s) in RCA: 105] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2019] [Revised: 07/24/2019] [Accepted: 07/25/2019] [Indexed: 12/29/2022] Open
Abstract
Hydrogen peroxide (H2O2), a non-radical reactive oxygen species generated during many (patho)physiological conditions, is currently universally recognized as an important mediator of redox-regulated processes. Depending on its spatiotemporal accumulation profile, this molecule may act as a signaling messenger or cause oxidative damage. The focus of this review is to comprehensively evaluate the evidence that peroxisomes, organelles best known for their role in cellular lipid metabolism, also serve as hubs in the H2O2 signaling network. We first briefly introduce the basic concepts of how H2O2 can drive cellular signaling events. Next, we outline the peroxisomal enzyme systems involved in H2O2 metabolism in mammals and reflect on how this oxidant can permeate across the organellar membrane. In addition, we provide an up-to-date overview of molecular targets and biological processes that can be affected by changes in peroxisomal H2O2 metabolism. Where possible, emphasis is placed on the molecular mechanisms and factors involved. From the data presented, it is clear that there are still numerous gaps in our knowledge. Therefore, gaining more insight into how peroxisomes are integrated in the cellular H2O2 signaling network is of key importance to unravel the precise role of peroxisomal H2O2 production and scavenging in normal and pathological conditions.
Collapse
|
28
|
Nanotherapies for Treatment of Cardiovascular Disease: A Case for Antioxidant Targeted Delivery. CURRENT PATHOBIOLOGY REPORTS 2019; 7:47-60. [PMID: 31396435 DOI: 10.1007/s40139-019-00196-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Purpose of Review Cardiovascular disease (CVD) involves a broad range of clinical manifestations resulting from a dysfunctional vascular system. Overproduction of reactive oxygen and nitrogen species are causally implicated in the severity of vascular dysfunction and CVD. Antioxidant therapy is an attractive avenue for treatment of CVD associated pathologies. Implementation of targeted nano-antioxidant therapies has the potential to overcome hurdles associated with systemic delivery of antioxidants. This review examines the currently available options for nanotherapeutic targeting CVD, and explores successful studies showcasing targeted nano-antioxidant therapy. Recent Findings Active targeting strategies in the context of CVD heavily focus on immunotargeting to inflammatory markers like cell adhesion molecules, or to exposed extracellular matrix components. Targeted antioxidant nanotherapies have found success in pre-clinical studies. Summary This review underscores the potential of targeted nanocarriers as means of finding success translating antioxidant therapies to the clinic, all with a focus on CVD.
Collapse
|
29
|
Potential Medication Treatment According to Pathological Mechanisms in Abdominal Aortic Aneurysm. J Cardiovasc Pharmacol 2019; 71:46-57. [PMID: 28953105 DOI: 10.1097/fjc.0000000000000540] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Abdominal aortic aneurysm (AAA) is a vascular disease with high mortality. Because of the lack of effective medications to stop or reverse the progression of AAA, surgical operation has become the most predominant recommendation of treatment for patients. There are many potential mechanisms, including inflammation, smooth muscle cell apoptosis, extracellular matrix degradation, oxidative stress, and so on, involving in AAA pathogenesis. According to those mechanisms, some potential therapeutic drugs have been proposed and tested in animal models and even in clinical trials. This review focuses on recent advances in both pathogenic mechanisms and potential pharmacologic therapies of AAA.
Collapse
|
30
|
Amos D, Cook C, Santanam N. Omega 3 rich diet modulates energy metabolism via GPR120-Nrf2 crosstalk in a novel antioxidant mouse model. Biochim Biophys Acta Mol Cell Biol Lipids 2019; 1864:466-488. [PMID: 30658097 DOI: 10.1016/j.bbalip.2019.01.002] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2018] [Revised: 12/12/2018] [Accepted: 01/07/2019] [Indexed: 02/08/2023]
Abstract
With obesity rates reaching epidemic proportions, more studies concentrated on reducing the risk and treating this epidemic are vital. Redox stress is an important metabolic regulator involved in the pathophysiology of cardiovascular disease, Type 2 diabetes, and obesity. Oxygen and nitrogen-derived free radicals alter glucose and lipid homeostasis in key metabolic tissues, leading to increases in risk of developing metabolic syndrome. Oxidants derived from dietary fat differ in their metabolic regulation, with numerous studies showing benefits from a high omega 3 rich diet compared to the frequently consumed "western diet" rich in saturated fat. Omega 3 (OM3) fatty acids improve lipid profile, lower inflammation, and ameliorate insulin resistance, possibly through maintaining redox homeostasis. This study is based on the hypothesis that altering endogenous antioxidant production and/or increasing OM3 rich diet consumption will improve energy metabolism and maintain insulin sensitivity. We tested the comparative metabolic effects of a diet rich in saturated fat (HFD) and an omega 3-enriched diet (OM3) in the newly developed 'stress-less' mice model that overexpresses the endogenous antioxidant catalase. Eight weeks of dietary intervention showed that mice overexpressing endogenous catalase compared to their wild-type controls when fed an OM3 enriched diet, in contrast to HFD, activated GPR120-Nrf2 cross-talk to maintain balanced energy metabolism, normal circadian rhythm, and insulin sensitivity. These findings suggest that redox regulation of GPR120/FFAR4 might be an important target in reducing risk of metabolic syndrome and associated diseases.
Collapse
Affiliation(s)
- Deborah Amos
- Department of Biomedical Sciences, Joan C. Edwards School of Medicine, Marshall University, 1700 3rd Ave, Huntington, WV 25755-0001, United States
| | - Carla Cook
- Department of Biomedical Sciences, Joan C. Edwards School of Medicine, Marshall University, 1700 3rd Ave, Huntington, WV 25755-0001, United States
| | - Nalini Santanam
- Department of Biomedical Sciences, Joan C. Edwards School of Medicine, Marshall University, 1700 3rd Ave, Huntington, WV 25755-0001, United States.
| |
Collapse
|
31
|
Wiernicki I, Parafiniuk M, Kolasa-Wołosiuk A, Gutowska I, Kazimierczak A, Clark J, Baranowska-Bosiacka I, Szumilowicz P, Gutowski P. Relationship between aortic wall oxidative stress/proteolytic enzyme expression and intraluminal thrombus thickness indicates a novel pathomechanism in the progression of human abdominal aortic aneurysm. FASEB J 2018; 33:885-895. [PMID: 30351992 DOI: 10.1096/fj.201800633r] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The possibility that oxidative stress promotes degradation of the extracellular matrix and a relationship between intraluminal thrombus (ILT) thickness and proteolytic activity within the abdominal aortic aneurysm (AAA) wall has been suggested. In the present study, the hypothesis that thin ILT is correlated with an increase in oxidative stress-related enzymes and matrix metalloproteinase-9 (MMP-9) expression within the human AAA wall was investigated. We also studied the antioxidant activity of superoxide dismutases, catalase, glutathione peroxidase, glutathione reductase, and thioredoxin within the full-thickness AAA wall and through fluoroimmunohistochemical staining of catalase and MMP-9 expression within the inner and outer media, in relation to ILT thickness. Reactive oxygen species control the degradation and remodeling of the extracellular matrix by up-regulating proteolytic enzymes, such as MMPs. Results showed that oxidative stress and proteolytic enzyme expression were simultaneously, significantly higher within thin thrombus (≤10 mm)-covered aneurysm wall when compared with the wall covered by thick thrombus (≥25 mm). These findings provide the first demonstration, to our knowledge, of a causative link between oxidative stress instigating proteolytic enzyme expression at the tissue level and human AAA development. Presence of a thin circumferential thrombus should always be considered as a risk factor for the greatest increase in aneurysm growth rate and rupture, giving an indication for surgery timing.-Wiernicki, I., Parafiniuk, M., Kolasa-Wołosiuk, A., Gutowska, I., Kazimierczak, A., Clark, J., Baranowska-Bosiacka, I., Szumilowicz, P., Gutowski, P. Relationship between aortic wall oxidative stress/proteolytic enzyme expression and intraluminal thrombus thickness indicates a novel pathomechanism in the progression of human abdominal aortic aneurysm.
Collapse
Affiliation(s)
- Ireneusz Wiernicki
- Department of Vascular Surgery and Angiology, Pomeranian Medical University, Szczecin, Poland
| | - Miroslaw Parafiniuk
- Department of Forensic Medicine, Pomeranian Medical University, Szczecin, Poland
| | | | - Izabela Gutowska
- Department of Biochemistry and Human Nutrition, Pomeranian Medical University, Szczecin, Poland
| | - Arkadiusz Kazimierczak
- Department of Vascular Surgery and Angiology, Pomeranian Medical University, Szczecin, Poland
| | - Jeremy Clark
- Department of Clinical and Molecular Biochemistry, Faculty of Laboratory Diagnostics and Molecular Medicine, Pomeranian Medical University, Szczecin, Poland
| | - Irena Baranowska-Bosiacka
- Department of Biochemistry and Medical Chemistry, Faculty of Laboratory Diagnostics and Molecular Medicine, Pomeranian Medical University, Szczecin, Poland
| | - Pawel Szumilowicz
- Department of Vascular Surgery and Angiology, Pomeranian Medical University, Szczecin, Poland
| | - Piotr Gutowski
- Department of Vascular Surgery and Angiology, Pomeranian Medical University, Szczecin, Poland
| |
Collapse
|
32
|
Šćepanović V, Tasić G, Repac N, Nikolić I, Janićijević A, Todorović D, Stojanović M, Šćepanović R, Mitrović D, Šćepanović T, Borozan S, Šćepanović L. The role of oxidative stress as a risk factor for rupture of posterior inferior cerebellar artery aneurysms. Mol Biol Rep 2018; 45:2157-2165. [DOI: 10.1007/s11033-018-4374-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2018] [Accepted: 09/07/2018] [Indexed: 01/01/2023]
|
33
|
Peng H, Zhang K, Liu Z, Xu Q, You B, Li C, Cao J, Zhou H, Li X, Chen J, Cheng G, Shi R, Zhang G. VPO1 Modulates Vascular Smooth Muscle Cell Phenotypic Switch by Activating Extracellular Signal-regulated Kinase 1/2 (ERK 1/2) in Abdominal Aortic Aneurysms. J Am Heart Assoc 2018; 7:e010069. [PMID: 30371171 PMCID: PMC6201418 DOI: 10.1161/jaha.118.010069] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/14/2018] [Accepted: 07/09/2018] [Indexed: 02/04/2023]
Abstract
Background Hydrogen peroxide (H2O2) is a critical molecular signal in the development of abdominal aortic aneurysm ( AAA ) formation. Vascular peroxidase 1 ( VPO 1) catalyzes the production of hypochlorous acid ( HOC l) from H2O2 and significantly enhances oxidative stress. The switch from a contractile phenotype to a synthetic one in vascular smooth muscle cells ( VSMC s) is driven by reactive oxygen species and is recognized as an early and important event in AAA formation. This study aims to determine if VPO 1 plays a critical role in the development of AAA by regulating VSMC phenotypic switch. Methods and Results VPO 1 is upregulated in human and elastase-induced mouse aneurysmal tissues compared with healthy control tissues. Additionally, KLF 4, a nuclear transcriptional factor, is upregulated in aneurysmatic tissues along with a concomitant downregulation of differentiated smooth muscle cell markers and an increase of synthetic phenotypic markers, indicating VSMC phenotypic switch in these diseased tissues. In cultured VSMC s from rat abdominal aorta, H2O2 treatment significantly increases VPO 1 expression and HOC l levels as well as VSMC phenotypic switch. In support of these findings, depletion of VPO 1 significantly attenuates the effects of H2O2 and HOC l treatment. Furthermore, HOC l treatment promotes VSMC phenotypic switch and ERK 1/2 phosphorylation. Pretreatment with U0126 (a specific inhibitor of ERK 1/2) significantly attenuates HOC l-induced VSMC phenotypic switch. Conclusions Our results demonstrate that VPO 1 modulates VSMC phenotypic switch through the H2O2/ VPO 1/ HOC l/ ERK 1/2 signaling pathway and plays a key role in the development of AAA . Our findings also implicate VPO 1 as a novel signaling node that mediates VSMC phenotypic switch and plays a key role in the development of AAA . Clinical Trial Registration URL : www.chictr.org.cn . Unique identifier: Chi CTR 1800016922.
Collapse
MESH Headings
- Aged
- Animals
- Aorta, Abdominal/cytology
- Aortic Aneurysm, Abdominal/metabolism
- Aortic Aneurysm, Abdominal/physiopathology
- Cell Movement
- Cell Proliferation
- Disease Models, Animal
- Female
- Hemeproteins/drug effects
- Hemeproteins/metabolism
- Humans
- Hydrogen Peroxide/pharmacology
- Hypochlorous Acid/pharmacology
- Kruppel-Like Factor 4
- Kruppel-Like Transcription Factors/metabolism
- MAP Kinase Signaling System
- Male
- Matrix Metalloproteinase 2/metabolism
- Mice
- Middle Aged
- Muscle Contraction
- Muscle, Smooth, Vascular/cytology
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/physiopathology
- Myocytes, Smooth Muscle/cytology
- Myocytes, Smooth Muscle/metabolism
- Oxidants/pharmacology
- Peroxidases/drug effects
- Peroxidases/metabolism
- Phenotype
- Reactive Oxygen Species
Collapse
Affiliation(s)
- Huihui Peng
- Department of Cardiovascular MedicineXiangya HospitalCentral South UniversityChangshaChina
| | - Kai Zhang
- Department of Cardiovascular MedicineXiangya HospitalCentral South UniversityChangshaChina
| | - Zhaoya Liu
- Department of Cardiovascular MedicineXiangya HospitalCentral South UniversityChangshaChina
| | - Qian Xu
- Department of Cardiovascular MedicineXiangya HospitalCentral South UniversityChangshaChina
| | - Baiyang You
- Department of Cardiovascular MedicineXiangya HospitalCentral South UniversityChangshaChina
| | - Chan Li
- Department of Cardiovascular MedicineXiangya HospitalCentral South UniversityChangshaChina
| | - Jing Cao
- Department of Cardiovascular MedicineXiangya HospitalCentral South UniversityChangshaChina
| | - Honghua Zhou
- Department of Cardiovascular MedicineXiangya HospitalCentral South UniversityChangshaChina
| | - Xiaohui Li
- Department of PharmacologySchool of Pharmaceutical SciencesCentral South UniversityChangshaChina
| | - Jia Chen
- Department of Humanistic NursingXiangya Nursing SchoolCentral South UniversityChangshaChina
| | - Guangjie Cheng
- Division of Pulmonary, Allergy & Critical Care MedicineDepartment of MedicineUniversity of Alabama at BirminghamAL
| | - Ruizheng Shi
- Department of Cardiovascular MedicineXiangya HospitalCentral South UniversityChangshaChina
| | - Guogang Zhang
- Department of Cardiovascular MedicineXiangya HospitalCentral South UniversityChangshaChina
| |
Collapse
|
34
|
Kim HW, Weintraub NL. Aortic Aneurysm: In Defense of the Vascular Smooth Muscle Cell. Arterioscler Thromb Vasc Biol 2018; 36:2138-2140. [PMID: 27784700 DOI: 10.1161/atvbaha.116.308356] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Affiliation(s)
- Ha Won Kim
- From the Division of Cardiology, Department of Medicine, Vascular Biology Center, Medical College of Georgia at Augusta University
| | - Neal L Weintraub
- From the Division of Cardiology, Department of Medicine, Vascular Biology Center, Medical College of Georgia at Augusta University.
| |
Collapse
|
35
|
Chuaiphichai S, Rashbrook VS, Hale AB, Trelfa L, Patel J, McNeill E, Lygate CA, Channon KM, Douglas G. Endothelial Cell Tetrahydrobiopterin Modulates Sensitivity to Ang (Angiotensin) II-Induced Vascular Remodeling, Blood Pressure, and Abdominal Aortic Aneurysm. Hypertension 2018; 72:128-138. [PMID: 29844152 PMCID: PMC6012043 DOI: 10.1161/hypertensionaha.118.11144] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2018] [Revised: 04/04/2018] [Accepted: 04/10/2018] [Indexed: 12/30/2022]
Abstract
GTPCH (GTP cyclohydrolase 1, encoded by Gch1) is required for the synthesis of tetrahydrobiopterin; a critical regulator of endothelial NO synthase function. We have previously shown that mice with selective loss of Gch1 in endothelial cells have mild vascular dysfunction, but the consequences of endothelial cell tetrahydrobiopterin deficiency in vascular disease pathogenesis are unknown. We investigated the pathological consequence of Ang (angiotensin) II infusion in endothelial cell Gch1 deficient (Gch1fl/fl Tie2cre) mice. Ang II (0.4 mg/kg per day, delivered by osmotic minipump) caused a significant decrease in circulating tetrahydrobiopterin levels in Gch1fl/fl Tie2cre mice and a significant increase in the Nω-nitro-L-arginine methyl ester inhabitable production of H2O2 in the aorta. Chronic treatment with this subpressor dose of Ang II resulted in a significant increase in blood pressure only in Gch1fl/fl Tie2cre mice. This finding was mirrored with acute administration of Ang II, where increased sensitivity to Ang II was observed at both pressor and subpressor doses. Chronic Ang II infusion in Gch1fl/fl Tie2ce mice resulted in vascular dysfunction in resistance mesenteric arteries with an enhanced constrictor and decreased dilator response and medial hypertrophy. Altered vascular remodeling was also observed in the aorta with an increase in the incidence of abdominal aortic aneurysm formation in Gch1fl/fl Tie2ce mice. These findings indicate a specific requirement for endothelial cell tetrahydrobiopterin in modulating the hemodynamic and structural changes induced by Ang II, through modulation of blood pressure, structural changes in resistance vessels, and aneurysm formation in the aorta.
Collapse
Affiliation(s)
- Surawee Chuaiphichai
- From the Division of Cardiovascular Medicine, British Heart Foundation Centre of Research Excellence and Wellcome Trust Centre for Human Genetics, University of Oxford, United Kingdom
| | - Victoria S Rashbrook
- From the Division of Cardiovascular Medicine, British Heart Foundation Centre of Research Excellence and Wellcome Trust Centre for Human Genetics, University of Oxford, United Kingdom
| | - Ashley B Hale
- From the Division of Cardiovascular Medicine, British Heart Foundation Centre of Research Excellence and Wellcome Trust Centre for Human Genetics, University of Oxford, United Kingdom
| | - Lucy Trelfa
- From the Division of Cardiovascular Medicine, British Heart Foundation Centre of Research Excellence and Wellcome Trust Centre for Human Genetics, University of Oxford, United Kingdom
| | - Jyoti Patel
- From the Division of Cardiovascular Medicine, British Heart Foundation Centre of Research Excellence and Wellcome Trust Centre for Human Genetics, University of Oxford, United Kingdom
| | - Eileen McNeill
- From the Division of Cardiovascular Medicine, British Heart Foundation Centre of Research Excellence and Wellcome Trust Centre for Human Genetics, University of Oxford, United Kingdom
| | - Craig A Lygate
- From the Division of Cardiovascular Medicine, British Heart Foundation Centre of Research Excellence and Wellcome Trust Centre for Human Genetics, University of Oxford, United Kingdom
| | - Keith M Channon
- From the Division of Cardiovascular Medicine, British Heart Foundation Centre of Research Excellence and Wellcome Trust Centre for Human Genetics, University of Oxford, United Kingdom.
| | - Gillian Douglas
- From the Division of Cardiovascular Medicine, British Heart Foundation Centre of Research Excellence and Wellcome Trust Centre for Human Genetics, University of Oxford, United Kingdom
| |
Collapse
|
36
|
Lucas ML, Carraro CC, Belló-Klein A, Kalil AN, Aerts NR, Carvalho FB, Fernandes MC, Zettler CG. Oxidative Stress in Aortas of Patients with Advanced Occlusive and Aneurysmal Diseases. Ann Vasc Surg 2018; 52:216-224. [PMID: 29758327 DOI: 10.1016/j.avsg.2018.02.027] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2017] [Revised: 02/05/2018] [Accepted: 02/23/2018] [Indexed: 01/07/2023]
Abstract
BACKGROUND Aortoiliac occlusive disease (AOD) and abdominal aortic aneurysm (AAA) are very important cardiovascular diseases that present different aspects of pathophysiology; however, oxidative stress and inflammatory response seem be relevant in both of them. Our objective was to evaluate oxidative damage and degree of inflammatory infiltrate in aortas of patients surgically treated for AOD and AAA. MATERIALS AND METHODS Levels of reactive oxygen species (ROS), nicotinamide adenine dinucleotide phosphate (NADPH) oxidase activity, and myeloperoxidase (MPO) expression as well as nitrite levels and superoxide dismutase (SOD) and catalase (CAT) activities were evaluated in aortas of patients with AOD (n = 16) or AAA (n = 14), while the control group was formed by cadaveric organ donors (n = 10). We also analyzed the degree of inflammatory infiltrate in these aortas. RESULTS There was an increase in ROS levels and NADPH oxidase activity in patients with AOD and AAA when compared with the control group, and the AOD group demonstrated higher ROS production and NADPH oxidase activity and also nitrite levels when compared with the AAA group (P < 0.001). On the other hand, an increase of SOD activity in the AOD group and CAT activity in the AAA group was observed. Inflammatory infiltrate and MPO expression were higher in the AOD group when compared with the control group (P < 0.05). CONCLUSIONS Oxidative stress is relevant in both AOD and AAA, though AOD presented higher ROS levels and NADPH activity. Increased activities of antioxidant enzymes may be a compensatory phenomenon which occurs in aortas of patients with AOD and AAA. Perhaps, a relationship between oxidative stress and degree of inflammatory infiltrate may exist in the pathophysiology of AOD and AAA.
Collapse
Affiliation(s)
- Márcio L Lucas
- Post-Graduating Course of Medical Sciences, Universidade Federal de Ciências da Saúde de Porto Alegre (UFCSPA), Porto Alegre, Rio Grande do Sul, Brazil; Department of Vascular Surgery, Santa Casa de Porto Alegre, Porto Alegre, Rio Grande do Sul, Brazil.
| | - Cristina C Carraro
- Department of Physiology, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Rio Grande do Sul, Brazil
| | - Adriane Belló-Klein
- Department of Physiology, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Rio Grande do Sul, Brazil
| | - Antônio N Kalil
- Post-Graduating Course of Medical Sciences, Universidade Federal de Ciências da Saúde de Porto Alegre (UFCSPA), Porto Alegre, Rio Grande do Sul, Brazil
| | - Newton R Aerts
- Department of Vascular Surgery, Santa Casa de Porto Alegre, Porto Alegre, Rio Grande do Sul, Brazil
| | - Fabiano B Carvalho
- Post-Graduating Course of Pathology, UFCSPA, Porto Alegre, Rio Grande do Sul, Brazil
| | - Marilda C Fernandes
- Post-Graduating Course of Pathology, UFCSPA, Porto Alegre, Rio Grande do Sul, Brazil
| | - Claudio G Zettler
- Post-Graduating Course of Medical Sciences, Universidade Federal de Ciências da Saúde de Porto Alegre (UFCSPA), Porto Alegre, Rio Grande do Sul, Brazil; Post-Graduating Course of Pathology, UFCSPA, Porto Alegre, Rio Grande do Sul, Brazil
| |
Collapse
|
37
|
Jiménez-Altayó F, Meirelles T, Crosas-Molist E, Sorolla MA, Del Blanco DG, López-Luque J, Mas-Stachurska A, Siegert AM, Bonorino F, Barberà L, García C, Condom E, Sitges M, Rodríguez-Pascual F, Laurindo F, Schröder K, Ros J, Fabregat I, Egea G. Redox stress in Marfan syndrome: Dissecting the role of the NADPH oxidase NOX4 in aortic aneurysm. Free Radic Biol Med 2018; 118:44-58. [PMID: 29471108 DOI: 10.1016/j.freeradbiomed.2018.02.023] [Citation(s) in RCA: 56] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/2017] [Revised: 02/12/2018] [Accepted: 02/15/2018] [Indexed: 11/16/2022]
Abstract
Marfan syndrome (MFS) is characterized by the formation of ascending aortic aneurysms resulting from altered assembly of extracellular matrix fibrillin-containing microfibrils and dysfunction of TGF-β signaling. Here we identify the molecular targets of redox stress in aortic aneurysms from MFS patients, and investigate the role of NOX4, whose expression is strongly induced by TGF-β, in aneurysm formation and progression in a murine model of MFS. Working models included aortae and cultured vascular smooth muscle cells (VSMC) from MFS patients, and a NOX4-deficient Marfan mouse model (Fbn1C1039G/+-Nox4-/-). Increased tyrosine nitration and reactive oxygen species levels were found in the tunica media of human aortic aneurysms and in cultured VSMC. Proteomic analysis identified nitrated and carbonylated proteins, which included smooth muscle α-actin (αSMA) and annexin A2. NOX4 immunostaining increased in the tunica media of human Marfan aorta and was transcriptionally overexpressed in VSMC. Fbn1C1039G/+-Nox4-/- mice aortas showed a reduction of fragmented elastic fibers, which was accompanied by an amelioration in the Marfan-associated enlargement of the aortic root. Increase in the contractile phenotype marker calponin in the tunica media of MFS mice aortas was abrogated in Fbn1C1039G/+-Nox4-/- mice. Endothelial dysfunction evaluated by myography in the Marfan ascending aorta was prevented by the absence of Nox4 or catalase-induced H2O2 decomposition. We conclude that redox stress occurs in MFS, whose targets are actin-based cytoskeleton members and regulators of extracellular matrix homeostasis. Likewise, NOX4 have an impact in the progression of the aortic dilation in MFS and in the structural organization of the aortic tunica media, the VSMC phenotypic modulation, and endothelial function.
Collapse
Affiliation(s)
- Francesc Jiménez-Altayó
- Departament de Farmacologia, Terapèutica i Toxicologia, Institut de Neurociències, Facultat de Medicina, Universitat Autònoma de Barcelona, Bellaterra, Spain
| | - Thayna Meirelles
- Department de Biomedicina, Facultat de Medicina i Ciències de la Salut, Universitat de Barcelona, and IDIBAPS, Barcelona, Spain
| | - Eva Crosas-Molist
- TGF-β and Cancer Group, Oncobell Program, IDIBELL, L'Hospitalet de Llobregat, Barcelona, Spain
| | - M Alba Sorolla
- Institut de Recerca Biomèdica de Lleida (IRB Lleida), Lleida, Spain; Departament de Ciències Mèdiques Bàsiques, Universitat de Lleida, Lleida, Spain
| | - Darya Gorbenko Del Blanco
- Department de Biomedicina, Facultat de Medicina i Ciències de la Salut, Universitat de Barcelona, and IDIBAPS, Barcelona, Spain
| | - Judit López-Luque
- TGF-β and Cancer Group, Oncobell Program, IDIBELL, L'Hospitalet de Llobregat, Barcelona, Spain
| | | | - Ana-Maria Siegert
- Department de Biomedicina, Facultat de Medicina i Ciències de la Salut, Universitat de Barcelona, and IDIBAPS, Barcelona, Spain
| | - Fabio Bonorino
- Department de Biomedicina, Facultat de Medicina i Ciències de la Salut, Universitat de Barcelona, and IDIBAPS, Barcelona, Spain
| | - Laura Barberà
- Department de Biomedicina, Facultat de Medicina i Ciències de la Salut, Universitat de Barcelona, and IDIBAPS, Barcelona, Spain
| | - Carolina García
- Department of Pathology, Hospital de Bellvitge-IDIBELL, L'Hospitalet de Llobregat, Barcelona, and Department of Pathology and Experimental Therapeutics, University of Barcelona, Barcelona, Spain
| | - Enric Condom
- Department of Pathology, Hospital de Bellvitge-IDIBELL, L'Hospitalet de Llobregat, Barcelona, and Department of Pathology and Experimental Therapeutics, University of Barcelona, Barcelona, Spain
| | - Marta Sitges
- Cardiovascular Institute, Hospital Clinic, IDIBAPS-University of Barcelona, Barcelona, Spain
| | | | - Francisco Laurindo
- Heart Institute (InCor), University of São Paulo School of Medicine, São Paulo, Brazil
| | - Katrin Schröder
- German Center of Cardiovascular Research (DZHK), Partner site Rhein Main, Frankfurt am Main, Germany
| | - Joaquim Ros
- Institut de Recerca Biomèdica de Lleida (IRB Lleida), Lleida, Spain; Departament de Ciències Mèdiques Bàsiques, Universitat de Lleida, Lleida, Spain
| | - Isabel Fabregat
- TGF-β and Cancer Group, Oncobell Program, IDIBELL, L'Hospitalet de Llobregat, Barcelona, Spain; Department de Ciències Fisiològiques, Facultat de Medicina i Ciències de la Salut, Universitat de Barcelona, Barcelona, Spain
| | - Gustavo Egea
- Department de Biomedicina, Facultat de Medicina i Ciències de la Salut, Universitat de Barcelona, and IDIBAPS, Barcelona, Spain.
| |
Collapse
|
38
|
Chai H, Tao Z, Chen W, Xu Y, Huang F, Su C, Chen X. Cortistatin attenuates angiotensin II-induced abdominal aortic aneurysm through inactivation of the ERK1/2 signaling pathways. Biochem Biophys Res Commun 2018; 495:1801-1806. [DOI: 10.1016/j.bbrc.2017.12.033] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2017] [Accepted: 12/06/2017] [Indexed: 02/07/2023]
|
39
|
Zhao H, Chen G, Wang H. Gadd153 deficiency attenuates abdominal aortic aneurysm formation in mice. INTERNATIONAL JOURNAL OF CLINICAL AND EXPERIMENTAL PATHOLOGY 2018; 11:169-178. [PMID: 31938098 PMCID: PMC6957950] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 02/04/2015] [Accepted: 03/30/2015] [Indexed: 06/10/2023]
Abstract
Abdominal aortic aneurysms (AAAs) are a chronic inflammatory vascular disease for which pharmacological treatments are not available. Gadd153 is closely associated with the onset of vascular smooth muscle cells (VSMCs) apoptosis. However, a role for Gadd153 in AngII-induced AAA formation is currently unknown. In our study, lentiviral-mediated silencing of Gadd153 through small RNA interference was performed in mice, which was further used for the establishment of mouse experimental AAA induced by infusion of angiotensin II (AngII). We found that Gadd153 deficiency prevented AngII-induced AAA formation in mice 14 days post perfusion compared with wild-type control mice. Moreover, Gadd153 deficiency significantly reduced lesion macrophage and CD4+ T-cell content, T-cell proliferation, SMC apoptosis, and matrix metalloproteinase expression. In vitro studies revealed that Gadd153 deficiency regulated microvessel growth and monocyte migration. In addition, Gadd153 deficiency also affected AAA lesion Mac-3 macrophage accumulation or CD31 microvessel numbers. In conclusion, our study demonstrates that Gadd153 plays an essential role in AngII-induced AAA formation by promoting inflammatory cells proliferation and vascular SMC apoptosis affecting MMPs expression.
Collapse
Affiliation(s)
- Huiying Zhao
- Genetic Diagnosis Center, The First Hospital of Jilin UniversityChangchun 130021, China
| | - Guiying Chen
- Department of Cardiology, The First Affiliated Hospital of Harbin Medical UniversityHarbin 150001, Heilongjiang, China
| | - Haifeng Wang
- Genetic Diagnosis Center, The First Hospital of Jilin UniversityChangchun 130021, China
| |
Collapse
|
40
|
Martin-Ventura JL, Rodrigues-Diez R, Martinez-Lopez D, Salaices M, Blanco-Colio LM, Briones AM. Oxidative Stress in Human Atherothrombosis: Sources, Markers and Therapeutic Targets. Int J Mol Sci 2017; 18:ijms18112315. [PMID: 29099757 PMCID: PMC5713284 DOI: 10.3390/ijms18112315] [Citation(s) in RCA: 36] [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: 10/04/2017] [Revised: 10/27/2017] [Accepted: 10/30/2017] [Indexed: 12/11/2022] Open
Abstract
Atherothrombosis remains one of the main causes of morbidity and mortality worldwide. The underlying pathology is a chronic pathological vascular remodeling of the arterial wall involving several pathways, including oxidative stress. Cellular and animal studies have provided compelling evidence of the direct role of oxidative stress in atherothrombosis, but such a relationship is not clearly established in humans and, to date, clinical trials on the possible beneficial effects of antioxidant therapy have provided equivocal results. Nicotinamide adenine dinucleotide phosphate (NADPH) oxidase is one of the main sources of reactive oxygen species (ROS) in human atherothrombosis. Moreover, leukocyte-derived myeloperoxidase (MPO) and red blood cell-derived iron could be involved in the oxidative modification of lipids/lipoproteins (LDL/HDL) in the arterial wall. Interestingly, oxidized lipoproteins, and antioxidants, have been analyzed as potential markers of oxidative stress in the plasma of patients with atherothrombosis. In this review, we will revise sources of ROS, focusing on NADPH oxidase, but also on MPO and iron. We will also discuss the impact of these oxidative systems on LDL and HDL, as well as the value of these modified lipoproteins as circulating markers of oxidative stress in atherothrombosis. We will finish by reviewing some antioxidant systems and compounds as therapeutic strategies to prevent pathological vascular remodeling.
Collapse
Affiliation(s)
- Jose Luis Martin-Ventura
- Vascular Research Lab, FIIS-Fundación Jiménez Díaz-Autonoma University, 28040 Madrid, Spain.
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), 28029 Madrid, Spain.
| | - Raquel Rodrigues-Diez
- Departamento de Farmacología, Facultad de Medicina, Universidad Autónoma de Madrid, 28029 Madrid, Spain.
- Instituto de Investigación Hospital Universitario La Paz (IdiPAZ), 28046 Madrid, Spain.
| | - Diego Martinez-Lopez
- Vascular Research Lab, FIIS-Fundación Jiménez Díaz-Autonoma University, 28040 Madrid, Spain.
| | - Mercedes Salaices
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), 28029 Madrid, Spain.
- Departamento de Farmacología, Facultad de Medicina, Universidad Autónoma de Madrid, 28029 Madrid, Spain.
- Instituto de Investigación Hospital Universitario La Paz (IdiPAZ), 28046 Madrid, Spain.
| | - Luis Miguel Blanco-Colio
- Vascular Research Lab, FIIS-Fundación Jiménez Díaz-Autonoma University, 28040 Madrid, Spain.
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), 28029 Madrid, Spain.
| | - Ana M Briones
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), 28029 Madrid, Spain.
- Departamento de Farmacología, Facultad de Medicina, Universidad Autónoma de Madrid, 28029 Madrid, Spain.
- Instituto de Investigación Hospital Universitario La Paz (IdiPAZ), 28046 Madrid, Spain.
| |
Collapse
|
41
|
Kawai T, Takayanagi T, Forrester SJ, Preston KJ, Obama T, Tsuji T, Kobayashi T, Boyer MJ, Cooper HA, Kwok HF, Hashimoto T, Scalia R, Rizzo V, Eguchi S. Vascular ADAM17 (a Disintegrin and Metalloproteinase Domain 17) Is Required for Angiotensin II/β-Aminopropionitrile-Induced Abdominal Aortic Aneurysm. Hypertension 2017; 70:959-963. [PMID: 28947615 DOI: 10.1161/hypertensionaha.117.09822] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2017] [Revised: 06/22/2017] [Accepted: 08/31/2017] [Indexed: 01/05/2023]
Abstract
Angiotensin II (AngII)-activated epidermal growth factor receptor has been implicated in abdominal aortic aneurysm (AAA) development. In vascular smooth muscle cells (VSMCs), AngII activates epidermal growth factor receptor via a metalloproteinase, ADAM17 (a disintegrin and metalloproteinase domain 17). We hypothesized that AngII-dependent AAA development would be prevented in mice lacking ADAM17 in VSMCs. To test this concept, control and VSMC ADAM17-deficient mice were cotreated with AngII and a lysyl oxidase inhibitor, β-aminopropionitrile, to induce AAA. We found that 52.4% of control mice did not survive because of aortic rupture. All other surviving control mice developed AAA and demonstrated enhanced expression of ADAM17 in the AAA lesions. In contrast, all AngII and β-aminopropionitrile-treated VSMC ADAM17-deficient mice survived and showed reduction in external/internal diameters (51%/28%, respectively). VSMC ADAM17 deficiency was associated with lack of epidermal growth factor receptor activation, interleukin-6 induction, endoplasmic reticulum/oxidative stress, and matrix deposition in the abdominal aorta of treated mice. However, both VSMC ADAM17-deficient and control mice treated with AngII and β-aminopropionitrile developed comparable levels of hypertension. Treatment of C57Bl/6 mice with an ADAM17 inhibitory antibody but not with control IgG also prevented AAA development. In conclusion, VSMC ADAM17 silencing or systemic ADAM17 inhibition seems to protect mice from AAA formation. The mechanism seems to involve suppression of epidermal growth factor receptor activation.
Collapse
Affiliation(s)
- Tatsuo Kawai
- From the Cardiovascular Research Center, Department of Physiology, Lewis Katz School of Medicine at Temple University, Philadelphia, PA (T. Kawai, T. Takayanagi, S.J.F., K.J.P., T.O., T. Tsuji, T. Kobayashi, M.J.B., H.A.C., R.S., V.R., S.E.); Faculty of Health Sciences, Macau Special Administrative Region, University of Macau, Taipa (H.F.K.); and Department of Anesthesia and Perioperative Care, University of California, San Francisco (T.H.)
| | - Takehiko Takayanagi
- From the Cardiovascular Research Center, Department of Physiology, Lewis Katz School of Medicine at Temple University, Philadelphia, PA (T. Kawai, T. Takayanagi, S.J.F., K.J.P., T.O., T. Tsuji, T. Kobayashi, M.J.B., H.A.C., R.S., V.R., S.E.); Faculty of Health Sciences, Macau Special Administrative Region, University of Macau, Taipa (H.F.K.); and Department of Anesthesia and Perioperative Care, University of California, San Francisco (T.H.)
| | - Steven J Forrester
- From the Cardiovascular Research Center, Department of Physiology, Lewis Katz School of Medicine at Temple University, Philadelphia, PA (T. Kawai, T. Takayanagi, S.J.F., K.J.P., T.O., T. Tsuji, T. Kobayashi, M.J.B., H.A.C., R.S., V.R., S.E.); Faculty of Health Sciences, Macau Special Administrative Region, University of Macau, Taipa (H.F.K.); and Department of Anesthesia and Perioperative Care, University of California, San Francisco (T.H.)
| | - Kyle J Preston
- From the Cardiovascular Research Center, Department of Physiology, Lewis Katz School of Medicine at Temple University, Philadelphia, PA (T. Kawai, T. Takayanagi, S.J.F., K.J.P., T.O., T. Tsuji, T. Kobayashi, M.J.B., H.A.C., R.S., V.R., S.E.); Faculty of Health Sciences, Macau Special Administrative Region, University of Macau, Taipa (H.F.K.); and Department of Anesthesia and Perioperative Care, University of California, San Francisco (T.H.)
| | - Takashi Obama
- From the Cardiovascular Research Center, Department of Physiology, Lewis Katz School of Medicine at Temple University, Philadelphia, PA (T. Kawai, T. Takayanagi, S.J.F., K.J.P., T.O., T. Tsuji, T. Kobayashi, M.J.B., H.A.C., R.S., V.R., S.E.); Faculty of Health Sciences, Macau Special Administrative Region, University of Macau, Taipa (H.F.K.); and Department of Anesthesia and Perioperative Care, University of California, San Francisco (T.H.)
| | - Toshiyuki Tsuji
- From the Cardiovascular Research Center, Department of Physiology, Lewis Katz School of Medicine at Temple University, Philadelphia, PA (T. Kawai, T. Takayanagi, S.J.F., K.J.P., T.O., T. Tsuji, T. Kobayashi, M.J.B., H.A.C., R.S., V.R., S.E.); Faculty of Health Sciences, Macau Special Administrative Region, University of Macau, Taipa (H.F.K.); and Department of Anesthesia and Perioperative Care, University of California, San Francisco (T.H.)
| | - Tomonori Kobayashi
- From the Cardiovascular Research Center, Department of Physiology, Lewis Katz School of Medicine at Temple University, Philadelphia, PA (T. Kawai, T. Takayanagi, S.J.F., K.J.P., T.O., T. Tsuji, T. Kobayashi, M.J.B., H.A.C., R.S., V.R., S.E.); Faculty of Health Sciences, Macau Special Administrative Region, University of Macau, Taipa (H.F.K.); and Department of Anesthesia and Perioperative Care, University of California, San Francisco (T.H.)
| | - Michael J Boyer
- From the Cardiovascular Research Center, Department of Physiology, Lewis Katz School of Medicine at Temple University, Philadelphia, PA (T. Kawai, T. Takayanagi, S.J.F., K.J.P., T.O., T. Tsuji, T. Kobayashi, M.J.B., H.A.C., R.S., V.R., S.E.); Faculty of Health Sciences, Macau Special Administrative Region, University of Macau, Taipa (H.F.K.); and Department of Anesthesia and Perioperative Care, University of California, San Francisco (T.H.)
| | - Hannah A Cooper
- From the Cardiovascular Research Center, Department of Physiology, Lewis Katz School of Medicine at Temple University, Philadelphia, PA (T. Kawai, T. Takayanagi, S.J.F., K.J.P., T.O., T. Tsuji, T. Kobayashi, M.J.B., H.A.C., R.S., V.R., S.E.); Faculty of Health Sciences, Macau Special Administrative Region, University of Macau, Taipa (H.F.K.); and Department of Anesthesia and Perioperative Care, University of California, San Francisco (T.H.)
| | - Hang Fai Kwok
- From the Cardiovascular Research Center, Department of Physiology, Lewis Katz School of Medicine at Temple University, Philadelphia, PA (T. Kawai, T. Takayanagi, S.J.F., K.J.P., T.O., T. Tsuji, T. Kobayashi, M.J.B., H.A.C., R.S., V.R., S.E.); Faculty of Health Sciences, Macau Special Administrative Region, University of Macau, Taipa (H.F.K.); and Department of Anesthesia and Perioperative Care, University of California, San Francisco (T.H.)
| | - Tomoki Hashimoto
- From the Cardiovascular Research Center, Department of Physiology, Lewis Katz School of Medicine at Temple University, Philadelphia, PA (T. Kawai, T. Takayanagi, S.J.F., K.J.P., T.O., T. Tsuji, T. Kobayashi, M.J.B., H.A.C., R.S., V.R., S.E.); Faculty of Health Sciences, Macau Special Administrative Region, University of Macau, Taipa (H.F.K.); and Department of Anesthesia and Perioperative Care, University of California, San Francisco (T.H.)
| | - Rosario Scalia
- From the Cardiovascular Research Center, Department of Physiology, Lewis Katz School of Medicine at Temple University, Philadelphia, PA (T. Kawai, T. Takayanagi, S.J.F., K.J.P., T.O., T. Tsuji, T. Kobayashi, M.J.B., H.A.C., R.S., V.R., S.E.); Faculty of Health Sciences, Macau Special Administrative Region, University of Macau, Taipa (H.F.K.); and Department of Anesthesia and Perioperative Care, University of California, San Francisco (T.H.)
| | - Victor Rizzo
- From the Cardiovascular Research Center, Department of Physiology, Lewis Katz School of Medicine at Temple University, Philadelphia, PA (T. Kawai, T. Takayanagi, S.J.F., K.J.P., T.O., T. Tsuji, T. Kobayashi, M.J.B., H.A.C., R.S., V.R., S.E.); Faculty of Health Sciences, Macau Special Administrative Region, University of Macau, Taipa (H.F.K.); and Department of Anesthesia and Perioperative Care, University of California, San Francisco (T.H.).
| | - Satoru Eguchi
- From the Cardiovascular Research Center, Department of Physiology, Lewis Katz School of Medicine at Temple University, Philadelphia, PA (T. Kawai, T. Takayanagi, S.J.F., K.J.P., T.O., T. Tsuji, T. Kobayashi, M.J.B., H.A.C., R.S., V.R., S.E.); Faculty of Health Sciences, Macau Special Administrative Region, University of Macau, Taipa (H.F.K.); and Department of Anesthesia and Perioperative Care, University of California, San Francisco (T.H.).
| |
Collapse
|
42
|
Mistriotis P, Andreadis ST. Vascular aging: Molecular mechanisms and potential treatments for vascular rejuvenation. Ageing Res Rev 2017; 37:94-116. [PMID: 28579130 DOI: 10.1016/j.arr.2017.05.006] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2017] [Revised: 05/22/2017] [Accepted: 05/25/2017] [Indexed: 12/14/2022]
Abstract
Aging is the main risk factor contributing to vascular dysfunction and the progression of vascular diseases. In this review, we discuss the causes and mechanisms of vascular aging at the tissue and cellular level. We focus on Endothelial Cell (EC) and Smooth Muscle Cell (SMC) aging due to their critical role in mediating the defective vascular phenotype. We elaborate on two categories that contribute to cellular dysfunction: cell extrinsic and intrinsic factors. Extrinsic factors reflect systemic or environmental changes which alter EC and SMC homeostasis compromising vascular function. Intrinsic factors induce EC and SMC transformation resulting in cellular senescence. Replenishing or rejuvenating the aged/dysfunctional vascular cells is critical to the effective repair of the vasculature. As such, this review also elaborates on recent findings which indicate that stem cell and gene therapies may restore the impaired vascular cell function, reverse vascular aging, and prolong lifespan.
Collapse
Affiliation(s)
- Panagiotis Mistriotis
- Bioengineering Laboratory, Department of Chemical and Biological Engineering, University at Buffalo, The State University of New York, Amherst, NY 14260-4200, USA
| | - Stelios T Andreadis
- Bioengineering Laboratory, Department of Chemical and Biological Engineering, University at Buffalo, The State University of New York, Amherst, NY 14260-4200, USA; Department of Biomedical Engineering, University at Buffalo, The State University of New York, Amherst, NY 14260-4200, USA; Center of Excellence in Bioinformatics and Life Sciences, Buffalo, NY 14203, USA.
| |
Collapse
|
43
|
Chen Q, Wang Q, Zhu J, Xiao Q, Zhang L. Reactive oxygen species: key regulators in vascular health and diseases. Br J Pharmacol 2017; 175:1279-1292. [PMID: 28430357 DOI: 10.1111/bph.13828] [Citation(s) in RCA: 207] [Impact Index Per Article: 25.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2017] [Revised: 03/31/2017] [Accepted: 04/11/2017] [Indexed: 12/18/2022] Open
Abstract
ROS are a group of small reactive molecules that play critical roles in the regulation of various cell functions and biological processes. In the vascular system, physiological levels of ROS are essential for normal vascular functions including endothelial homeostasis and smooth muscle cell contraction. In contrast, uncontrolled overproduction of ROS resulting from an imbalance of ROS generation and elimination leads to the development of vascular diseases. Excessive ROS cause vascular cell damage, the recruitment of inflammatory cells, lipid peroxidation, activation of metalloproteinases and deposition of extracellular matrix, collectively leading to vascular remodelling. Evidence from a large number of studies has revealed that ROS and oxidative stress are involved in the initiation and progression of numerous vascular diseases including hypertension, atherosclerosis, restenosis and abdominal aortic aneurysm. Furthermore, considerable research has been implemented to explore antioxidants that can reduce ROS production and oxidative stress in order to ameliorate vascular diseases. In this review, we will discuss the nature and sources of ROS, their roles in vascular homeostasis and specific vascular diseases and various antioxidants as well as some of the pharmacological agents that are capable of reducing ROS and oxidative stress. The aim of this review is to provide information for developing promising clinical strategies targeting ROS to decrease cardiovascular risks. LINKED ARTICLES This article is part of a themed section on Spotlight on Small Molecules in Cardiovascular Diseases. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v175.8/issuetoc.
Collapse
Affiliation(s)
- Qishan Chen
- Department of Cardiology, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Qiwen Wang
- Department of Cardiology, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Jianhua Zhu
- Department of Cardiology, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Qingzhong Xiao
- Centre for Clinical Pharmacology, William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Li Zhang
- Department of Cardiology, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| |
Collapse
|
44
|
Shen YH, LeMaire SA. Molecular pathogenesis of genetic and sporadic aortic aneurysms and dissections. Curr Probl Surg 2017; 54:95-155. [PMID: 28521856 DOI: 10.1067/j.cpsurg.2017.01.001] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2016] [Accepted: 01/16/2017] [Indexed: 12/20/2022]
Affiliation(s)
- Ying H Shen
- Division of Cardiothoracic Surgery, Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, TX; Department of Cardiovascular Surgery, Texas Heart Institute, Houston, TX; Cardiovascular Research Institute, Baylor College of Medicine, Houston, TX.
| | - Scott A LeMaire
- Division of Cardiothoracic Surgery, Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, TX; Department of Cardiovascular Surgery, Texas Heart Institute, Houston, TX; Cardiovascular Research Institute, Baylor College of Medicine, Houston, TX; Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, TX.
| |
Collapse
|
45
|
Abstract
Hypertension has a direct impact on vascular hypertrophy and is a known risk factor for the development of atherosclerosis. Osteopontin (OPN) has emerged as an important protein mediator of inflammation and remodeling of large arteries. However, its role and mechanism of regulation in the setting of hypertension is still unknown. Our objectives for this study were therefore to investigate the role of OPN in hypertension-induced vascular remodeling and inflammation. OPN Knockout (KO) and wild type (WT) mice were made hypertensive with angiotensin II (Ang II) infusion for seven days. We observed that OPN KO aortas were protected against Ang II-induced medial hypertrophy and inflammation, despite comparable increases in systolic blood pressure (SBP) in both groups. OPN expression was increased in WT aortas from hypertensive mice (induced by either Ang II or norepinephrine). OPN expression was increased in aortic smooth muscle cells (SMCs) subjected to cyclic mechanical strain suggesting that mechanical deformation of the aortic wall is responsible in part for the increased OPN expression induced by hypertension. Finally, we utilized hypertensive transgenic smooth muscle cell-specific catalase overexpressing (TgSMC-Cat) mice to determine the role of H2O2 in mediating hypertension-induced increases in OPN expression. We also found that the hypertension-induced increase in OPN expression was inhibited in transgenic smooth muscle cell-specific catalase overexpressing (TgSMC-Cat) mice, suggesting that H2O2, plays a vital role in mediating the hypertension-induced increase in OPN expression. Taken together, these results define a potentially important role for OPN in the pathophysiology of hypertension.
Collapse
|
46
|
Lu WW, Jia LX, Ni XQ, Zhao L, Chang JR, Zhang JS, Hou YL, Zhu Y, Guan YF, Yu YR, Du J, Tang CS, Qi YF. Intermedin1-53 Attenuates Abdominal Aortic Aneurysm by Inhibiting Oxidative Stress. Arterioscler Thromb Vasc Biol 2016; 36:2176-2190. [PMID: 27634835 DOI: 10.1161/atvbaha.116.307825] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2015] [Accepted: 08/31/2016] [Indexed: 12/20/2022]
Abstract
OBJECTIVE Oxidative stress plays a critical role in the development of abdominal aortic aneurysm (AAA). Intermedin (IMD) is a regulator of oxidative stress. Here, we investigated whether IMD reduces AAA by inhibiting oxidative stress. APPROACH AND RESULTS In angiotensin II-induced ApoE-/- mouse and CaCl2-induced C57BL/6J mouse model of AAA, IMD1-53 significantly reduced the incidence of AAA and maximal aortic diameter. Ultrasonography, hematoxylin, and eosin staining and Verhoeff-van Gieson staining showed that IMD1-53 significantly decreased the enlarged aortas and elastic lamina degradation induced by angiotensin II or CaCl2. Mechanistically, IMD1-53 attenuated oxidative stress, inflammation, vascular smooth muscle cell apoptosis, and matrix metalloproteinase activation. IMD1-53 inhibited the activation of redox-sensitive signaling pathways, decreased the mRNA and protein expression of nicotinamide adenine dinucleotide phosphate oxidase subunits, and reduced the activity of nicotinamide adenine dinucleotide phosphate oxidase in AAA mice. Expression of Nox4 was upregulated in human AAA segments and in angiotensin II-treated mouse aortas and was markedly decreased by IMD1-53. In vitro, vascular smooth muscle cells with small-interfering RNA knockdown of IMD showed significantly increased angiotensin II-induced reactive oxygen species, and small-interfering RNA knockdown of Nox4 markedly inhibited the reactive oxygen species. IMD knockdown further increased the apoptosis of vascular smooth muscle cells and inflammation, which was reversed by Nox4 knockdown. Preincubation with IMD17-47 and protein kinase A inhibitor H89 inhibited the effect of IMD1-53, reducing Nox4 protein levels. CONCLUSIONS IMD1-53 could have a protective effect on AAA by inhibiting oxidative stress.
Collapse
Affiliation(s)
- Wei-Wei Lu
- From the Key Laboratory of Remodeling-Related Cardiovascular Diseases, Beijing Institute of Heart, Lung, and Blood Vessel Diseases, Beijing An Zhen Hospital, Capital Medical University, Ministry of Education, China (W.-W.L., L.-X.J., X.-Q.N., L.Z., Y.-L.H., J.D., Y.-F.Q.); Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China (W.-W.L., X.-Q.N., L.Z., J.-R.C., J.-S.Z., Y.Z., Y.-F.G., C.-S.T., Y.-F.Q.); and Department of Pathogen Biology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China (W.-W.L., X.-Q.N., J.-S.Z., Y.-L.H., Y.-R.Y., Y.-F.Q.)
| | - Li-Xin Jia
- From the Key Laboratory of Remodeling-Related Cardiovascular Diseases, Beijing Institute of Heart, Lung, and Blood Vessel Diseases, Beijing An Zhen Hospital, Capital Medical University, Ministry of Education, China (W.-W.L., L.-X.J., X.-Q.N., L.Z., Y.-L.H., J.D., Y.-F.Q.); Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China (W.-W.L., X.-Q.N., L.Z., J.-R.C., J.-S.Z., Y.Z., Y.-F.G., C.-S.T., Y.-F.Q.); and Department of Pathogen Biology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China (W.-W.L., X.-Q.N., J.-S.Z., Y.-L.H., Y.-R.Y., Y.-F.Q.)
| | - Xian-Qiang Ni
- From the Key Laboratory of Remodeling-Related Cardiovascular Diseases, Beijing Institute of Heart, Lung, and Blood Vessel Diseases, Beijing An Zhen Hospital, Capital Medical University, Ministry of Education, China (W.-W.L., L.-X.J., X.-Q.N., L.Z., Y.-L.H., J.D., Y.-F.Q.); Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China (W.-W.L., X.-Q.N., L.Z., J.-R.C., J.-S.Z., Y.Z., Y.-F.G., C.-S.T., Y.-F.Q.); and Department of Pathogen Biology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China (W.-W.L., X.-Q.N., J.-S.Z., Y.-L.H., Y.-R.Y., Y.-F.Q.)
| | - Lei Zhao
- From the Key Laboratory of Remodeling-Related Cardiovascular Diseases, Beijing Institute of Heart, Lung, and Blood Vessel Diseases, Beijing An Zhen Hospital, Capital Medical University, Ministry of Education, China (W.-W.L., L.-X.J., X.-Q.N., L.Z., Y.-L.H., J.D., Y.-F.Q.); Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China (W.-W.L., X.-Q.N., L.Z., J.-R.C., J.-S.Z., Y.Z., Y.-F.G., C.-S.T., Y.-F.Q.); and Department of Pathogen Biology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China (W.-W.L., X.-Q.N., J.-S.Z., Y.-L.H., Y.-R.Y., Y.-F.Q.)
| | - Jin-Rui Chang
- From the Key Laboratory of Remodeling-Related Cardiovascular Diseases, Beijing Institute of Heart, Lung, and Blood Vessel Diseases, Beijing An Zhen Hospital, Capital Medical University, Ministry of Education, China (W.-W.L., L.-X.J., X.-Q.N., L.Z., Y.-L.H., J.D., Y.-F.Q.); Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China (W.-W.L., X.-Q.N., L.Z., J.-R.C., J.-S.Z., Y.Z., Y.-F.G., C.-S.T., Y.-F.Q.); and Department of Pathogen Biology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China (W.-W.L., X.-Q.N., J.-S.Z., Y.-L.H., Y.-R.Y., Y.-F.Q.)
| | - Jin-Sheng Zhang
- From the Key Laboratory of Remodeling-Related Cardiovascular Diseases, Beijing Institute of Heart, Lung, and Blood Vessel Diseases, Beijing An Zhen Hospital, Capital Medical University, Ministry of Education, China (W.-W.L., L.-X.J., X.-Q.N., L.Z., Y.-L.H., J.D., Y.-F.Q.); Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China (W.-W.L., X.-Q.N., L.Z., J.-R.C., J.-S.Z., Y.Z., Y.-F.G., C.-S.T., Y.-F.Q.); and Department of Pathogen Biology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China (W.-W.L., X.-Q.N., J.-S.Z., Y.-L.H., Y.-R.Y., Y.-F.Q.)
| | - Yue-Long Hou
- From the Key Laboratory of Remodeling-Related Cardiovascular Diseases, Beijing Institute of Heart, Lung, and Blood Vessel Diseases, Beijing An Zhen Hospital, Capital Medical University, Ministry of Education, China (W.-W.L., L.-X.J., X.-Q.N., L.Z., Y.-L.H., J.D., Y.-F.Q.); Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China (W.-W.L., X.-Q.N., L.Z., J.-R.C., J.-S.Z., Y.Z., Y.-F.G., C.-S.T., Y.-F.Q.); and Department of Pathogen Biology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China (W.-W.L., X.-Q.N., J.-S.Z., Y.-L.H., Y.-R.Y., Y.-F.Q.)
| | - Yi Zhu
- From the Key Laboratory of Remodeling-Related Cardiovascular Diseases, Beijing Institute of Heart, Lung, and Blood Vessel Diseases, Beijing An Zhen Hospital, Capital Medical University, Ministry of Education, China (W.-W.L., L.-X.J., X.-Q.N., L.Z., Y.-L.H., J.D., Y.-F.Q.); Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China (W.-W.L., X.-Q.N., L.Z., J.-R.C., J.-S.Z., Y.Z., Y.-F.G., C.-S.T., Y.-F.Q.); and Department of Pathogen Biology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China (W.-W.L., X.-Q.N., J.-S.Z., Y.-L.H., Y.-R.Y., Y.-F.Q.)
| | - You-Fei Guan
- From the Key Laboratory of Remodeling-Related Cardiovascular Diseases, Beijing Institute of Heart, Lung, and Blood Vessel Diseases, Beijing An Zhen Hospital, Capital Medical University, Ministry of Education, China (W.-W.L., L.-X.J., X.-Q.N., L.Z., Y.-L.H., J.D., Y.-F.Q.); Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China (W.-W.L., X.-Q.N., L.Z., J.-R.C., J.-S.Z., Y.Z., Y.-F.G., C.-S.T., Y.-F.Q.); and Department of Pathogen Biology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China (W.-W.L., X.-Q.N., J.-S.Z., Y.-L.H., Y.-R.Y., Y.-F.Q.)
| | - Yan-Rong Yu
- From the Key Laboratory of Remodeling-Related Cardiovascular Diseases, Beijing Institute of Heart, Lung, and Blood Vessel Diseases, Beijing An Zhen Hospital, Capital Medical University, Ministry of Education, China (W.-W.L., L.-X.J., X.-Q.N., L.Z., Y.-L.H., J.D., Y.-F.Q.); Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China (W.-W.L., X.-Q.N., L.Z., J.-R.C., J.-S.Z., Y.Z., Y.-F.G., C.-S.T., Y.-F.Q.); and Department of Pathogen Biology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China (W.-W.L., X.-Q.N., J.-S.Z., Y.-L.H., Y.-R.Y., Y.-F.Q.)
| | - Jie Du
- From the Key Laboratory of Remodeling-Related Cardiovascular Diseases, Beijing Institute of Heart, Lung, and Blood Vessel Diseases, Beijing An Zhen Hospital, Capital Medical University, Ministry of Education, China (W.-W.L., L.-X.J., X.-Q.N., L.Z., Y.-L.H., J.D., Y.-F.Q.); Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China (W.-W.L., X.-Q.N., L.Z., J.-R.C., J.-S.Z., Y.Z., Y.-F.G., C.-S.T., Y.-F.Q.); and Department of Pathogen Biology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China (W.-W.L., X.-Q.N., J.-S.Z., Y.-L.H., Y.-R.Y., Y.-F.Q.)
| | - Chao-Shu Tang
- From the Key Laboratory of Remodeling-Related Cardiovascular Diseases, Beijing Institute of Heart, Lung, and Blood Vessel Diseases, Beijing An Zhen Hospital, Capital Medical University, Ministry of Education, China (W.-W.L., L.-X.J., X.-Q.N., L.Z., Y.-L.H., J.D., Y.-F.Q.); Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China (W.-W.L., X.-Q.N., L.Z., J.-R.C., J.-S.Z., Y.Z., Y.-F.G., C.-S.T., Y.-F.Q.); and Department of Pathogen Biology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China (W.-W.L., X.-Q.N., J.-S.Z., Y.-L.H., Y.-R.Y., Y.-F.Q.)
| | - Yong-Fen Qi
- From the Key Laboratory of Remodeling-Related Cardiovascular Diseases, Beijing Institute of Heart, Lung, and Blood Vessel Diseases, Beijing An Zhen Hospital, Capital Medical University, Ministry of Education, China (W.-W.L., L.-X.J., X.-Q.N., L.Z., Y.-L.H., J.D., Y.-F.Q.); Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China (W.-W.L., X.-Q.N., L.Z., J.-R.C., J.-S.Z., Y.Z., Y.-F.G., C.-S.T., Y.-F.Q.); and Department of Pathogen Biology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China (W.-W.L., X.-Q.N., J.-S.Z., Y.-L.H., Y.-R.Y., Y.-F.Q.).
| |
Collapse
|
47
|
Byon CH, Heath JM, Chen Y. Redox signaling in cardiovascular pathophysiology: A focus on hydrogen peroxide and vascular smooth muscle cells. Redox Biol 2016; 9:244-253. [PMID: 27591403 PMCID: PMC5011184 DOI: 10.1016/j.redox.2016.08.015] [Citation(s) in RCA: 115] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2016] [Accepted: 08/23/2016] [Indexed: 02/07/2023] Open
Abstract
Oxidative stress represents excessive intracellular levels of reactive oxygen species (ROS), which plays a major role in the pathogenesis of cardiovascular disease. Besides having a critical impact on the development and progression of vascular pathologies including atherosclerosis and diabetic vasculopathy, oxidative stress also regulates physiological signaling processes. As a cell permeable ROS generated by cellular metabolism involved in intracellular signaling, hydrogen peroxide (H2O2) exerts tremendous impact on cardiovascular pathophysiology. Under pathological conditions, increased oxidase activities and/or impaired antioxidant systems results in uncontrolled production of ROS. In a pro-oxidant environment, vascular smooth muscle cells (VSMC) undergo phenotypic changes which can lead to the development of vascular dysfunction such as vascular inflammation and calcification. Investigations are ongoing to elucidate the mechanisms for cardiovascular disorders induced by oxidative stress. This review mainly focuses on the role of H2O2 in regulating physiological and pathological signals in VSMC.
Collapse
Affiliation(s)
| | - Jack M Heath
- Department of Pathology, Birmingham, AL 35294, USA
| | - Yabing Chen
- Department of Pathology, Birmingham, AL 35294, USA; University of Alabama at Birmingham, and the Birmingham Veterans Affairs Medical Center, Birmingham, AL 35294, USA.
| |
Collapse
|
48
|
Abstract
Abdominal aortic aneurysm (AAA) is a significant cause of mortality in older adults. A key mechanism implicated in AAA pathogenesis is inflammation and the associated production of reactive oxygen species (ROS) and oxidative stress. These have been suggested to promote degradation of the extracellular matrix (ECM) and vascular smooth muscle apoptosis. Experimental and human association studies suggest that ROS can be favourably modified to limit AAA formation and progression. In the present review, we discuss mechanisms potentially linking ROS to AAA pathogenesis and highlight potential treatment strategies targeting ROS. Currently, none of these strategies has been shown to be effective in clinical practice.
Collapse
|
49
|
Onetti Y, Meirelles T, Dantas AP, Schröder K, Vila E, Egea G, Jiménez-Altayó F. NADPH oxidase 4 attenuates cerebral artery changes during the progression of Marfan syndrome. Am J Physiol Heart Circ Physiol 2016; 310:H1081-90. [PMID: 26945079 DOI: 10.1152/ajpheart.00770.2015] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/06/2015] [Accepted: 03/01/2016] [Indexed: 12/11/2022]
Abstract
Marfan syndrome (MFS) is a connective tissue disorder that is often associated with the fibrillin-1 (Fbn1) gene mutation and characterized by cardiovascular alterations, predominantly ascending aortic aneurysms. Although neurovascular complications are uncommon in MFS, the improvement in Marfan patients' life expectancy is revealing other secondary alterations, potentially including neurovascular disorders. However, little is known about small-vessel pathophysiology in MFS. MFS is associated with hyperactivated transforming growth factor (TGF)-β signaling, which among numerous other downstream effectors, induces the NADPH oxidase 4 (Nox4) isoform of NADPH oxidase, a strong enzymatic source of H2O2 We hypothesized that MFS induces middle cerebral artery (MCA) alterations and that Nox4 contributes to them. MCA properties from 3-, 6-, or 9-mo-old Marfan (Fbn1(C1039G/+)) mice were compared with those from age/sex-matched wild-type littermates. At 6 mo, Marfan compared with wild-type mice developed higher MCA wall/lumen (wild-type: 0.081 ± 0.004; Marfan: 0.093 ± 0.002; 60 mmHg; P < 0.05), coupled with increased reactive oxygen species production, TGF-β, and Nox4 expression. However, wall stiffness and myogenic autoregulation did not change. To investigate the influence of Nox4 on cerebrovascular properties, we generated Marfan mice with Nox4 deficiency (Nox4(-/-)). Strikingly, Nox4 deletion in Marfan mice aggravated MCA wall thickening (cross-sectional area; Marfan: 6,660 ± 363 μm(2); Marfan Nox4(-/-): 8,795 ± 824 μm(2); 60 mmHg; P < 0.05), accompanied by decreased TGF-β expression and increased collagen deposition and Nox1 expression. These findings provide the first evidence that Nox4 mitigates cerebral artery structural changes in a murine model of MFS.
Collapse
Affiliation(s)
- Yara Onetti
- Departament de Farmacologia, de Terapèutica i de Toxicologia, Institut de Neurociències, Facultat de Medicina, Universitat Autònoma de Barcelona, Bellaterra, Spain
| | - Thayna Meirelles
- Departament de Biologia Cellular, Immunologia i Neurociències, Facultat de Medicina, Universitat de Barcelona, Barcelona, Spain
| | - Ana P Dantas
- Institut Clínic del Tòrax, Institut d'Investigacions Biomèdiques August Pi i Sunyer, Barcelona, Spain; and
| | - Katrin Schröder
- Institute for Cardiovascular Physiology, Goethe-University Frankfurt, Frankfurt, Germany
| | - Elisabet Vila
- Departament de Farmacologia, de Terapèutica i de Toxicologia, Institut de Neurociències, Facultat de Medicina, Universitat Autònoma de Barcelona, Bellaterra, Spain
| | - Gustavo Egea
- Departament de Biologia Cellular, Immunologia i Neurociències, Facultat de Medicina, Universitat de Barcelona, Barcelona, Spain; Institut Clínic del Tòrax, Institut d'Investigacions Biomèdiques August Pi i Sunyer, Barcelona, Spain; and
| | - Francesc Jiménez-Altayó
- Departament de Farmacologia, de Terapèutica i de Toxicologia, Institut de Neurociències, Facultat de Medicina, Universitat Autònoma de Barcelona, Bellaterra, Spain;
| |
Collapse
|
50
|
Sawada H, Hao H, Naito Y, Oboshi M, Hirotani S, Mitsuno M, Miyamoto Y, Hirota S, Masuyama T. Aortic Iron Overload With Oxidative Stress and Inflammation in Human and Murine Abdominal Aortic Aneurysm. Arterioscler Thromb Vasc Biol 2015; 35:1507-14. [DOI: 10.1161/atvbaha.115.305586] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2014] [Accepted: 03/30/2015] [Indexed: 01/28/2023]
Abstract
Objective—
Although iron is an essential element for maintaining physiological function, excess iron leads to tissue damage caused by oxidative stress and inflammation. Oxidative stress and inflammation play critical roles for the development of abdominal aortic aneurysm (AAA). However, it has not been investigated whether iron plays a role in AAA formation through oxidative stress and inflammation. We, therefore, examined whether iron is involved in the pathophysiology of AAA formation using human AAA walls and murine AAA models.
Approach and Results—
Human aortic walls were collected from 53 patients who underwent cardiovascular surgery (non-AAA=34; AAA=19). Murine AAA was induced by infusion of angiotensin II to apolipoprotein E knockout mice. Iron was accumulated in human and murine AAA walls compared with non-AAA walls. Immunohistochemistry showed that both 8-hydroxy-2′-deoxyguanosine and CD68-positive areas were increased in AAA walls compared with non-AAA walls. The extent of iron accumulated area positively correlated with that of 8-hydroxy-2′-deoxyguanosine expression area and macrophage infiltration area in human and murine AAA walls. We next investigated the effects of dietary iron restriction on AAA formation in mice. Iron restriction reduced the incidence of AAA formation with attenuation of oxidative stress and inflammation. Aortic expression of transferrin receptor 1, intracellular iron transport protein, was increased in human and murine AAA walls, and transferrin receptor 1–positive area was similar to areas where iron accumulated and F4/80 were positive.
Conclusions—
Iron is involved in the pathophysiology of AAA formation with oxidative stress and inflammation. Dietary iron restriction could be a new therapeutic strategy for AAA progression.
Collapse
Affiliation(s)
- Hisashi Sawada
- From the Cardiovascular Division, Department of Internal Medicine (H.S., Y.N., M.O., S. Hirotani, T.M.), Department of Surgical Pathology (H.H., S. Hirota), and Department of Cardiovascular Surgery (M.M., Y.M.), Hyogo College of Medicine, Nishinomiya, Hyogo, Japan
| | - Hiroyuki Hao
- From the Cardiovascular Division, Department of Internal Medicine (H.S., Y.N., M.O., S. Hirotani, T.M.), Department of Surgical Pathology (H.H., S. Hirota), and Department of Cardiovascular Surgery (M.M., Y.M.), Hyogo College of Medicine, Nishinomiya, Hyogo, Japan
| | - Yoshiro Naito
- From the Cardiovascular Division, Department of Internal Medicine (H.S., Y.N., M.O., S. Hirotani, T.M.), Department of Surgical Pathology (H.H., S. Hirota), and Department of Cardiovascular Surgery (M.M., Y.M.), Hyogo College of Medicine, Nishinomiya, Hyogo, Japan
| | - Makiko Oboshi
- From the Cardiovascular Division, Department of Internal Medicine (H.S., Y.N., M.O., S. Hirotani, T.M.), Department of Surgical Pathology (H.H., S. Hirota), and Department of Cardiovascular Surgery (M.M., Y.M.), Hyogo College of Medicine, Nishinomiya, Hyogo, Japan
| | - Shinichi Hirotani
- From the Cardiovascular Division, Department of Internal Medicine (H.S., Y.N., M.O., S. Hirotani, T.M.), Department of Surgical Pathology (H.H., S. Hirota), and Department of Cardiovascular Surgery (M.M., Y.M.), Hyogo College of Medicine, Nishinomiya, Hyogo, Japan
| | - Masataka Mitsuno
- From the Cardiovascular Division, Department of Internal Medicine (H.S., Y.N., M.O., S. Hirotani, T.M.), Department of Surgical Pathology (H.H., S. Hirota), and Department of Cardiovascular Surgery (M.M., Y.M.), Hyogo College of Medicine, Nishinomiya, Hyogo, Japan
| | - Yuji Miyamoto
- From the Cardiovascular Division, Department of Internal Medicine (H.S., Y.N., M.O., S. Hirotani, T.M.), Department of Surgical Pathology (H.H., S. Hirota), and Department of Cardiovascular Surgery (M.M., Y.M.), Hyogo College of Medicine, Nishinomiya, Hyogo, Japan
| | - Seiichi Hirota
- From the Cardiovascular Division, Department of Internal Medicine (H.S., Y.N., M.O., S. Hirotani, T.M.), Department of Surgical Pathology (H.H., S. Hirota), and Department of Cardiovascular Surgery (M.M., Y.M.), Hyogo College of Medicine, Nishinomiya, Hyogo, Japan
| | - Tohru Masuyama
- From the Cardiovascular Division, Department of Internal Medicine (H.S., Y.N., M.O., S. Hirotani, T.M.), Department of Surgical Pathology (H.H., S. Hirota), and Department of Cardiovascular Surgery (M.M., Y.M.), Hyogo College of Medicine, Nishinomiya, Hyogo, Japan
| |
Collapse
|