1
|
Paredes F, Williams HC, Liu X, Holden C, Bogan B, Wang Y, Crotty KM, Yeligar SM, Elorza AA, Lin Z, Rezvan A, San Martin A. The mitochondrial protease ClpP is a druggable target that controls VSMC phenotype by a SIRT1-dependent mechanism. Redox Biol 2024; 73:103203. [PMID: 38823208 PMCID: PMC11169483 DOI: 10.1016/j.redox.2024.103203] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2024] [Revised: 05/12/2024] [Accepted: 05/20/2024] [Indexed: 06/03/2024] Open
Abstract
Vascular smooth muscle cells (VSMCs), known for their remarkable lifelong phenotypic plasticity, play a pivotal role in vascular pathologies through their ability to transition between different phenotypes. Our group discovered that the deficiency of the mitochondrial protein Poldip2 induces VSMC differentiation both in vivo and in vitro. Further comprehensive biochemical investigations revealed Poldip2's specific interaction with the mitochondrial ATPase caseinolytic protease chaperone subunit X (CLPX), which is the regulatory subunit for the caseinolytic protease proteolytic subunit (ClpP) that forms part of the ClpXP complex - a proteasome-like protease evolutionarily conserved from bacteria to humans. This interaction limits the protease's activity, and reduced Poldip2 levels lead to ClpXP complex activation. This finding prompted the hypothesis that ClpXP complex activity within the mitochondria may regulate the VSMC phenotype. Employing gain-of-function and loss-of-function strategies, we demonstrated that ClpXP activity significantly influences the VSMC phenotype. Notably, both genetic and pharmacological activation of ClpXP inhibits VSMC plasticity and fosters a quiescent, differentiated, and anti-inflammatory VSMC phenotype. The pharmacological activation of ClpP using TIC10, currently in phase III clinical trials for cancer, successfully replicates this phenotype both in vitro and in vivo and markedly reduces aneurysm development in a mouse model of elastase-induced aortic aneurysms. Our mechanistic exploration indicates that ClpP activation regulates the VSMC phenotype by modifying the cellular NAD+/NADH ratio and activating Sirtuin 1. Our findings reveal the crucial role of mitochondrial proteostasis in the regulation of the VSMC phenotype and propose the ClpP protease as a novel, actionable target for manipulating the VSMC phenotype.
Collapse
Affiliation(s)
- Felipe Paredes
- Department of Medicine, Division of Cardiology, Emory University, Atlanta, GA, United States
| | - Holly C Williams
- Department of Medicine, Division of Cardiology, Emory University, Atlanta, GA, United States
| | - Xuesong Liu
- Department of Medicine, Division of Cardiology, Emory University, Atlanta, GA, United States; Department of Cardiology, The First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Claire Holden
- Department of Medicine, Division of Cardiology, Emory University, Atlanta, GA, United States
| | - Bethany Bogan
- Department of Medicine, Division of Cardiology, Emory University, Atlanta, GA, United States
| | - Yu Wang
- Department of Medicine, Division of Cardiology, Emory University, Atlanta, GA, United States
| | - Kathryn M Crotty
- Department of Medicine, Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Emory University, Atlanta, GA, United States; Atlanta Veterans Affairs Health Care System, Decatur, GA, United States
| | - Samantha M Yeligar
- Department of Medicine, Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Emory University, Atlanta, GA, United States; Atlanta Veterans Affairs Health Care System, Decatur, GA, United States
| | - Alvaro A Elorza
- Institute of Biomedical Sciences, Faculty of Medicine and Faculty of Life Sciences, Universidad Andres Bello, Santiago, Chile
| | - Zhiyong Lin
- Department of Medicine, Division of Cardiology, Emory University, Atlanta, GA, United States
| | - Amir Rezvan
- Department of Medicine, Division of Cardiology, Emory University, Atlanta, GA, United States
| | - Alejandra San Martin
- Department of Medicine, Division of Cardiology, Emory University, Atlanta, GA, United States; Institute of Biomedical Sciences, Faculty of Medicine and Faculty of Life Sciences, Universidad Andres Bello, Santiago, Chile.
| |
Collapse
|
2
|
Song W, Tu G, Qin L, Wei L, Chen J. Macrophage in Sporadic Thoracic Aortic Aneurysm and Dissection: Potential Therapeutic and Preventing Target. Rev Cardiovasc Med 2023; 24:340. [PMID: 39077089 PMCID: PMC11272886 DOI: 10.31083/j.rcm2412340] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Revised: 09/04/2023] [Accepted: 09/12/2023] [Indexed: 07/31/2024] Open
Abstract
Thoracic aortic aneurysm and dissection (TAAD) is a life-threatening cardiovascular disorder lacking effective clinical pharmacological therapies. The underlying molecular mechanisms of TAAD still remain elusive with participation of versatile cell types and components including endothelial cells (ECs), smooth muscle cells (SMCs), fibroblasts, immune cells, and the extracellular matrix (ECM). The main pathological features of TAAD include SMC dysfunction, phenotypic switching, and ECM degradation, which is closely associated with inflammation and immune cell infiltration. Among various types of immune cells, macrophages are a distinct participator in the formation and progression of TAAD. In this review, we first highlight the important role of inflammation and immune cell infiltration in TAAD. Furthermore, we discuss the role of macrophages in TAAD from the aspects of macrophage origination, classification, and functions. On the basis of experimental and clinical studies, we summarize key regulators of macrophages in TAAD. Finally, we review how targeting macrophages can reduce TAAD in murine models. A better understanding of the molecular and cellular mechanisms of TAAD may provide novel insights into preventing and treating the condition.
Collapse
Affiliation(s)
- Wenyu Song
- Department of Cardiovascular Surgery, Zhongshan Hospital, Fudan University, 200032 Shanghai, China
| | - Guowei Tu
- Cardiac Intensive Care Center, Zhongshan Hospital, Fudan University, 200032 Shanghai, China
| | - Lieyang Qin
- Department of Cardiovascular Surgery, Zhongshan Hospital, Fudan University, 200032 Shanghai, China
| | - Lai Wei
- Department of Cardiovascular Surgery, Zhongshan Hospital, Fudan University, 200032 Shanghai, China
| | - Jinmiao Chen
- Department of Cardiovascular Surgery, Zhongshan Hospital, Fudan University, 200032 Shanghai, China
| |
Collapse
|
3
|
Gong W, Tian Y, Li L. T cells in abdominal aortic aneurysm: immunomodulation and clinical application. Front Immunol 2023; 14:1240132. [PMID: 37662948 PMCID: PMC10471798 DOI: 10.3389/fimmu.2023.1240132] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Accepted: 08/07/2023] [Indexed: 09/05/2023] Open
Abstract
Abdominal aortic aneurysm (AAA) is characterized by inflammatory cell infiltration, extracellular matrix (ECM) degradation, and vascular smooth muscle cell (SMC) dysfunction. The inflammatory cells involved in AAA mainly include immune cells including macrophages, neutrophils, T-lymphocytes and B lymphocytes and endothelial cells. As the blood vessel wall expands, more and more lymphocytes infiltrate into the outer membrane. It was found that more than 50% of lymphocytes in AAA tissues were CD3+ T cells, including CD4+, CD8+T cells, γδ T cells and regulatory T cells (Tregs). Due to the important role of T cells in inflammatory response, an increasing number of researchers have paid attention to the role of T cells in AAA and dug into the relevant mechanism. Therefore, this paper focuses on reviewing the immunoregulatory role of T cells in AAA and their role in immunotherapy, seeking potential targets for immunotherapy and putting forward future research directions.
Collapse
Affiliation(s)
| | | | - Lei Li
- Department of Vascular Surgery, The Second Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
| |
Collapse
|
4
|
Pauli J, Reisenauer T, Winski G, Sachs N, Chernogubova E, Freytag H, Otto C, Reeps C, Eckstein HH, Scholz CJ, Maegdefessel L, Busch A. Apolipoprotein E (ApoE) Rescues the Contractile Smooth Muscle Cell Phenotype in Popliteal Artery Aneurysm Disease. Biomolecules 2023; 13:1074. [PMID: 37509110 PMCID: PMC10377618 DOI: 10.3390/biom13071074] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Revised: 06/22/2023] [Accepted: 06/28/2023] [Indexed: 07/30/2023] Open
Abstract
Popliteal artery aneurysm (PAA) is the most frequent peripheral aneurysm, primarily seen in male smokers with a prevalence below 1%. This exploratory study aims to shed light on cellular mechanisms involved in PAA progression. Sixteen human PAA and eight non-aneurysmatic popliteal artery samples, partially from the same patients, were analyzed by immunohistochemistry, fluorescence imaging, Affymetrix mRNA expression profiling, qPCR and OLink proteomics, and compared to atherosclerotic (n = 6) and abdominal aortic aneurysm (AAA) tissue (n = 19). Additionally, primary cell culture of PAA-derived vascular smooth muscle cells (VSMC) was established for modulation and growth analysis. Compared to non-aneurysmatic popliteal arteries, VSMCs lose the contractile phenotype and the cell proliferation rate increases significantly in PAA. Array analysis identified APOE higher expressed in PAA samples, co-localizing with VSMCs. APOE stimulation of primary human PAA VSMCs significantly reduced cell proliferation. Accordingly, contractile VSMC markers were significantly upregulated. A single case of osseous mechanically induced PAA with a non-diseased VSMC profile emphasizes these findings. Carefully concluded, PAA pathogenesis shows similar features to AAA, yet the mechanisms involved might differ. APOE is specifically higher expressed in PAA tissue and could be involved in VSMC phenotype rescue.
Collapse
Affiliation(s)
- Jessica Pauli
- Department for Vascular and Endovascular Surgery, Klinikum rechts der Isar, Technical University Munich, 81675 Munich, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Munich Heart Alliance, 10785 Berlin, Germany
| | - Tessa Reisenauer
- Department for Vascular and Endovascular Surgery, Klinikum rechts der Isar, Technical University Munich, 81675 Munich, Germany
| | - Greg Winski
- Molecular Vascular Medicine Group, Center for Molecular Medicine, Karolinska Institute, 17177 Stockholm, Sweden
- Perioperative Medicine and Intensive Care, Karolinska University Hospital, 17177 Stockholm, Sweden
| | - Nadja Sachs
- Department for Vascular and Endovascular Surgery, Klinikum rechts der Isar, Technical University Munich, 81675 Munich, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Munich Heart Alliance, 10785 Berlin, Germany
| | - Ekaterina Chernogubova
- Molecular Vascular Medicine Group, Center for Molecular Medicine, Karolinska Institute, 17177 Stockholm, Sweden
| | - Hannah Freytag
- Department for Vascular and Endovascular Surgery, Klinikum rechts der Isar, Technical University Munich, 81675 Munich, Germany
| | - Christoph Otto
- Department of General, Visceral, Transplantation, Vascular & Pediatric Surgery, University Hospital Würzburg, 97080 Würzburg, Germany
| | - Christian Reeps
- Division of Vascular and Endovascular Surgery, Department for Visceral, Thoracic and Vascular Surgery, Medical Faculty Carl Gustav Carus and University Hospital, Technische Universität Dresden, 01307 Dresden, Germany
| | - Hans-Henning Eckstein
- Department for Vascular and Endovascular Surgery, Klinikum rechts der Isar, Technical University Munich, 81675 Munich, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Munich Heart Alliance, 10785 Berlin, Germany
| | | | - Lars Maegdefessel
- Department for Vascular and Endovascular Surgery, Klinikum rechts der Isar, Technical University Munich, 81675 Munich, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Munich Heart Alliance, 10785 Berlin, Germany
- Molecular Vascular Medicine Group, Center for Molecular Medicine, Karolinska Institute, 17177 Stockholm, Sweden
| | - Albert Busch
- Department for Vascular and Endovascular Surgery, Klinikum rechts der Isar, Technical University Munich, 81675 Munich, Germany
- Division of Vascular and Endovascular Surgery, Department for Visceral, Thoracic and Vascular Surgery, Medical Faculty Carl Gustav Carus and University Hospital, Technische Universität Dresden, 01307 Dresden, Germany
| |
Collapse
|
5
|
Zhang J, Guo Y, Zhao X, Pang J, Pan C, Wang J, Wei S, Yu X, Zhang C, Chen Y, Yin H, Xu F. The role of aldehyde dehydrogenase 2 in cardiovascular disease. Nat Rev Cardiol 2023; 20:495-509. [PMID: 36781974 DOI: 10.1038/s41569-023-00839-5] [Citation(s) in RCA: 43] [Impact Index Per Article: 43.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 01/09/2023] [Indexed: 02/15/2023]
Abstract
Aldehyde dehydrogenase 2 (ALDH2) is a mitochondrial enzyme involved in the detoxification of alcohol-derived acetaldehyde and endogenous aldehydes. The inactivating ALDH2 rs671 polymorphism, present in up to 8% of the global population and in up to 50% of the East Asian population, is associated with increased risk of cardiovascular conditions such as coronary artery disease, alcohol-induced cardiac dysfunction, pulmonary arterial hypertension, heart failure and drug-induced cardiotoxicity. Although numerous studies have attributed an accumulation of aldehydes (secondary to alcohol consumption, ischaemia or elevated oxidative stress) to an increased risk of cardiovascular disease (CVD), this accumulation alone does not explain the emerging protective role of ALDH2 rs671 against ageing-related cardiac dysfunction and the development of aortic aneurysm or dissection. ALDH2 can also modulate risk factors associated with atherosclerosis, such as cholesterol biosynthesis and HDL biogenesis in hepatocytes and foam cell formation and efferocytosis in macrophages, via non-enzymatic pathways. In this Review, we summarize the basic biology and the clinical relevance of the enzymatic and non-enzymatic, tissue-specific roles of ALDH2 in CVD, and discuss the future directions in the research and development of therapeutic strategies targeting ALDH2. A thorough understanding of the complex roles of ALDH2 in CVD will improve the diagnosis, management and prognosis of patients with CVD who harbour the ALDH2 rs671 polymorphism.
Collapse
Affiliation(s)
- Jian Zhang
- Department of Emergency Medicine, Chest Pain Center, Shandong Provincial Clinical Research Center for Emergency and Critical Care Medicine, Qilu Hospital of Shandong University, Shandong, China
- Key Laboratory of Emergency and Critical Care Medicine of Shandong Province, Key Laboratory of Cardiopulmonary-Cerebral Resuscitation Research of Shandong Province, Shandong Provincial Engineering Laboratory for Emergency and Critical Care Medicine, Qilu Hospital of Shandong University, Shandong, China
- Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese Ministry of Health and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Qilu Hospital of Shandong University, Shandong, China
| | - Yunyun Guo
- Department of Emergency Medicine, Chest Pain Center, Shandong Provincial Clinical Research Center for Emergency and Critical Care Medicine, Qilu Hospital of Shandong University, Shandong, China
- Key Laboratory of Emergency and Critical Care Medicine of Shandong Province, Key Laboratory of Cardiopulmonary-Cerebral Resuscitation Research of Shandong Province, Shandong Provincial Engineering Laboratory for Emergency and Critical Care Medicine, Qilu Hospital of Shandong University, Shandong, China
- Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese Ministry of Health and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Qilu Hospital of Shandong University, Shandong, China
| | - Xiangkai Zhao
- Department of Emergency Medicine, Chest Pain Center, Shandong Provincial Clinical Research Center for Emergency and Critical Care Medicine, Qilu Hospital of Shandong University, Shandong, China
- Key Laboratory of Emergency and Critical Care Medicine of Shandong Province, Key Laboratory of Cardiopulmonary-Cerebral Resuscitation Research of Shandong Province, Shandong Provincial Engineering Laboratory for Emergency and Critical Care Medicine, Qilu Hospital of Shandong University, Shandong, China
- Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese Ministry of Health and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Qilu Hospital of Shandong University, Shandong, China
| | - Jiaojiao Pang
- Department of Emergency Medicine, Chest Pain Center, Shandong Provincial Clinical Research Center for Emergency and Critical Care Medicine, Qilu Hospital of Shandong University, Shandong, China
- Key Laboratory of Emergency and Critical Care Medicine of Shandong Province, Key Laboratory of Cardiopulmonary-Cerebral Resuscitation Research of Shandong Province, Shandong Provincial Engineering Laboratory for Emergency and Critical Care Medicine, Qilu Hospital of Shandong University, Shandong, China
- Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese Ministry of Health and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Qilu Hospital of Shandong University, Shandong, China
| | - Chang Pan
- Department of Emergency Medicine, Chest Pain Center, Shandong Provincial Clinical Research Center for Emergency and Critical Care Medicine, Qilu Hospital of Shandong University, Shandong, China
- Key Laboratory of Emergency and Critical Care Medicine of Shandong Province, Key Laboratory of Cardiopulmonary-Cerebral Resuscitation Research of Shandong Province, Shandong Provincial Engineering Laboratory for Emergency and Critical Care Medicine, Qilu Hospital of Shandong University, Shandong, China
- Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese Ministry of Health and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Qilu Hospital of Shandong University, Shandong, China
| | - Jiali Wang
- Department of Emergency Medicine, Chest Pain Center, Shandong Provincial Clinical Research Center for Emergency and Critical Care Medicine, Qilu Hospital of Shandong University, Shandong, China
- Key Laboratory of Emergency and Critical Care Medicine of Shandong Province, Key Laboratory of Cardiopulmonary-Cerebral Resuscitation Research of Shandong Province, Shandong Provincial Engineering Laboratory for Emergency and Critical Care Medicine, Qilu Hospital of Shandong University, Shandong, China
- Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese Ministry of Health and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Qilu Hospital of Shandong University, Shandong, China
| | - Shujian Wei
- Department of Emergency Medicine, Chest Pain Center, Shandong Provincial Clinical Research Center for Emergency and Critical Care Medicine, Qilu Hospital of Shandong University, Shandong, China
- Key Laboratory of Emergency and Critical Care Medicine of Shandong Province, Key Laboratory of Cardiopulmonary-Cerebral Resuscitation Research of Shandong Province, Shandong Provincial Engineering Laboratory for Emergency and Critical Care Medicine, Qilu Hospital of Shandong University, Shandong, China
- Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese Ministry of Health and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Qilu Hospital of Shandong University, Shandong, China
| | - Xiao Yu
- Key Laboratory Experimental Teratology of the Ministry of Education, Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Shandong University, Shandong, China
| | - Cheng Zhang
- Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese Ministry of Health and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Qilu Hospital of Shandong University, Shandong, China
- Department of Cardiology, Qilu Hospital of Shandong University, Shandong, China
| | - Yuguo Chen
- Department of Emergency Medicine, Chest Pain Center, Shandong Provincial Clinical Research Center for Emergency and Critical Care Medicine, Qilu Hospital of Shandong University, Shandong, China.
- Key Laboratory of Emergency and Critical Care Medicine of Shandong Province, Key Laboratory of Cardiopulmonary-Cerebral Resuscitation Research of Shandong Province, Shandong Provincial Engineering Laboratory for Emergency and Critical Care Medicine, Qilu Hospital of Shandong University, Shandong, China.
- Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese Ministry of Health and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Qilu Hospital of Shandong University, Shandong, China.
| | - Huiyong Yin
- Chinese Academy of Sciences Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, Innovation Center for Intervention of Chronic Disease and Promotion of Health, Chinese Academy of Sciences, Shanghai, China.
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China.
- Department of Biomedical Sciences, City University of Hong Kong, Hong Kong SAR, China.
| | - Feng Xu
- Department of Emergency Medicine, Chest Pain Center, Shandong Provincial Clinical Research Center for Emergency and Critical Care Medicine, Qilu Hospital of Shandong University, Shandong, China.
- Key Laboratory of Emergency and Critical Care Medicine of Shandong Province, Key Laboratory of Cardiopulmonary-Cerebral Resuscitation Research of Shandong Province, Shandong Provincial Engineering Laboratory for Emergency and Critical Care Medicine, Qilu Hospital of Shandong University, Shandong, China.
- Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese Ministry of Health and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Qilu Hospital of Shandong University, Shandong, China.
| |
Collapse
|
6
|
Guo X, Cai D, Dong K, Li C, Xu Z, Chen SY. DOCK2 Deficiency Attenuates Abdominal Aortic Aneurysm Formation-Brief Report. Arterioscler Thromb Vasc Biol 2023; 43:e210-e217. [PMID: 37021575 PMCID: PMC10212530 DOI: 10.1161/atvbaha.122.318400] [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: 09/03/2021] [Accepted: 03/28/2023] [Indexed: 04/07/2023]
Abstract
BACKGROUND Abdominal aortic aneurysm (AAA) is a potentially lethal disease that lacks pharmacological treatment. Degradation of extracellular matrix proteins, especially elastin laminae, is the hallmark for AAA development. DOCK2 (dedicator of cytokinesis 2) has shown proinflammatory effects in several inflammatory diseases and acts as a novel mediator for vascular remodeling. However, the role of DOCK2 in AAA formation remains unknown. METHODS Ang II (angiotensin II) infusion of ApoE-/- (apolipoprotein E deficient) mouse and topical elastase-induced AAA combined with DOCK2-/- (DOCK2 knockout) mouse models were used to study DOCK2 function in AAA formation/dissection. The relevance of DOCK2 to human AAA was examined using human aneurysm specimens. Elastin fragmentation in AAA lesion was observed by elastin staining. Elastin-degrading enzyme MMP (matrix metalloproteinase) activity was measured by in situ zymography. RESULTS DOCK2 was robustly upregulated in AAA lesion of Ang II-infused ApoE-/- mice, elastase-treated mice, as well as human AAA lesions. DOCK2-/- significantly attenuated the Ang II-induced AAA formation/dissection or rupture in mice along with reduction of MCP-1 (monocyte chemoattractant protein-1) and MMP expression and activity. Accordingly, the elastin fragmentation observed in ApoE-/- mouse aorta infused with Ang II and elastase-treated aorta was significantly attenuated by DOCK2 deficiency. Moreover, DOCK2-/- decreased the prevalence and severity of aneurysm formation, as well as the elastin degradation observed in the topical elastase model. CONCLUSIONS Our results indicate that DOCK2 is a novel regulator for AAA formation. DOCK2 regulates AAA development by promoting MCP-1 and MMP2 expression to incite vascular inflammation and elastin degradation.
Collapse
Affiliation(s)
- Xia Guo
- Department of Cellular and Molecular Biology, The University of Texas Health Science Center at Tyler, Tyler, TX, USA
- Department of Physiology & Pharmacology, University of Georgia, Athens, GA, USA
| | - Dunpeng Cai
- Department of Surgery, School of Medicine, The University of Missouri, Columbia, MO, USA
| | - Kun Dong
- Department of Physiology & Pharmacology, University of Georgia, Athens, GA, USA
| | - Chenxiao Li
- Department of Physiology & Pharmacology, University of Georgia, Athens, GA, USA
| | - Zaiyan Xu
- Department of Physiology & Pharmacology, University of Georgia, Athens, GA, USA
| | - Shi-You Chen
- Department of Surgery, School of Medicine, The University of Missouri, Columbia, MO, USA
- Department of Medical Pharmacology & Physiology, School of Medicine, The University of Missouri, Columbia, MO, USA
- Department of Physiology & Pharmacology, University of Georgia, Athens, GA, USA
| |
Collapse
|
7
|
Yamanouchi D. Unpacking the Complexities of a Silent Killer. Int J Mol Sci 2023; 24:ijms24087125. [PMID: 37108288 PMCID: PMC10139038 DOI: 10.3390/ijms24087125] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Accepted: 03/30/2023] [Indexed: 04/29/2023] Open
Abstract
An abdominal aortic aneurysm (AAA) is a life-threatening condition that affects millions of people worldwide [...].
Collapse
Affiliation(s)
- Dai Yamanouchi
- Division of Vascular Surgery, Department of Surgery, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705, USA
| |
Collapse
|
8
|
Márquez-Sánchez AC, Koltsova EK. Immune and inflammatory mechanisms of abdominal aortic aneurysm. Front Immunol 2022; 13:989933. [PMID: 36275758 PMCID: PMC9583679 DOI: 10.3389/fimmu.2022.989933] [Citation(s) in RCA: 35] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2022] [Accepted: 08/23/2022] [Indexed: 11/13/2022] Open
Abstract
Abdominal aortic aneurysm (AAA) is a life-threatening cardiovascular disease. Immune-mediated infiltration and a destruction of the aortic wall during AAA development plays significant role in the pathogenesis of this disease. While various immune cells had been found in AAA, the mechanisms of their activation and function are still far from being understood. A better understanding of mechanisms regulating the development of aberrant immune cell activation in AAA is essential for the development of novel preventive and therapeutic approaches. In this review we summarize current knowledge about the role of immune cells in AAA and discuss how pathogenic immune cell activation is regulated in this disease.
Collapse
|
9
|
Edaravone Attenuated Angiotensin II-Induced Atherosclerosis and Abdominal Aortic Aneurysms in Apolipoprotein E-Deficient Mice. Biomolecules 2022; 12:biom12081117. [PMID: 36009011 PMCID: PMC9405883 DOI: 10.3390/biom12081117] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2022] [Revised: 08/09/2022] [Accepted: 08/12/2022] [Indexed: 12/17/2022] Open
Abstract
Background: The aim of the study was to define whether edaravone, a free-radical scavenger, influenced angiotensin II (AngII)-induced atherosclerosis and abdominal aortic aneurysms (AAAs) formation. Methods: Male apolipoprotein E-deficient mice (8–12 weeks old) were fed with a normal diet for 5 weeks. Either edaravone (10 mg/kg/day) or vehicle was injected intraperitoneally for 5 weeks. After 1 week of injections, mice were infused subcutaneously with either AngII (1000 ng/kg/min, n = 16–17 per group) or saline (n = 5 per group) by osmotic minipumps for 4 weeks. Results: AngII increased systolic blood pressure equivalently in mice administered with either edaravone or saline. Edaravone had no effect on plasma total cholesterol concentrations and body weights. AngII infusion significantly increased ex vivo maximal diameters of abdominal aortas and en face atherosclerosis but was significantly attenuated by edaravone administration. Edaravone also reduced the incidence of AngII-induced AAAs. In addition, edaravone diminished AngII-induced aortic MMP-2 activation. Quantitative RT-PCR revealed that edaravone ameliorated mRNA abundance of aortic MCP-1 and IL-1β. Immunostaining demonstrated that edaravone attenuated oxidative stress and macrophage accumulation in the aorta. Furthermore, edaravone administration suppressed thioglycolate-induced mice peritoneal macrophages (MPMs) accumulation and mRNA abundance of MCP-1 in MPMs in male apolipoprotein E-deficient mice. In vitro, edaravone reduced LPS-induced mRNA abundance of MCP-1 in MPMs. Conclusions: Edaravone attenuated AngII-induced AAAs and atherosclerosis in male apolipoprotein E-deficient mice via anti-oxidative action and anti-inflammatory effect.
Collapse
|
10
|
Lu S, Wang R, Fu W, Si Y. Applications of Extracellular Vesicles in Abdominal Aortic Aneurysm. Front Cardiovasc Med 2022; 9:927542. [PMID: 35711380 PMCID: PMC9194528 DOI: 10.3389/fcvm.2022.927542] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2022] [Accepted: 05/12/2022] [Indexed: 11/17/2022] Open
Abstract
Abdominal aortic aneurysm (AAA) is a localized expansion of the abdominal aorta which can lead to lethal complication as the rupture of aortic wall. Currently there is still neither competent method to predict the impending rupture of aneurysm, nor effective treatment to arrest the progression of small and asymptomatic aneurysms. Accumulating evidence has confirmed the crucial role of extracellular vesicles (EVs) in the pathological course of AAA, acting as important mediators of intercellular communication. Given the advantages of intrinsic targeting properties, lower toxicity and fair stability, EVs show great potential to serve as biomarkers, therapeutic agents and drug delivery carriers. However, EV therapies still face several major challenges before they can be applied clinically, including off-target effect, low accumulation rate and rapid clearance by mononuclear phagocyte system. In this review, we first illustrate the roles of EV in the pathological process of AAA and evaluate its possible clinical applications. We also identify present challenges for EV applications, highlight different strategies of EV engineering and constructions of EV-like nanoparticles, including EV display technology and membrane hybrid technology. These leading-edge techniques have been recently employed in multiple cardiovascular diseases and their promising application in the field of AAA is discussed.
Collapse
Affiliation(s)
- Shan Lu
- Department of Vascular Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
- Vascular Surgery Institute of Fudan University, Shanghai, China
- National Clinical Research Center for Interventional Medicine, Shanghai, China
| | - Ruihan Wang
- Department of Vascular Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
- Vascular Surgery Institute of Fudan University, Shanghai, China
- National Clinical Research Center for Interventional Medicine, Shanghai, China
| | - Weiguo Fu
- Department of Vascular Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
- Vascular Surgery Institute of Fudan University, Shanghai, China
- National Clinical Research Center for Interventional Medicine, Shanghai, China
- Weiguo Fu
| | - Yi Si
- Department of Vascular Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
- Vascular Surgery Institute of Fudan University, Shanghai, China
- National Clinical Research Center for Interventional Medicine, Shanghai, China
- *Correspondence: Yi Si
| |
Collapse
|
11
|
Rombouts KB, van Merrienboer TAR, Ket JCF, Bogunovic N, van der Velden J, Yeung KK. The role of vascular smooth muscle cells in the development of aortic aneurysms and dissections. Eur J Clin Invest 2022; 52:e13697. [PMID: 34698377 PMCID: PMC9285394 DOI: 10.1111/eci.13697] [Citation(s) in RCA: 89] [Impact Index Per Article: 44.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Revised: 09/12/2021] [Accepted: 10/11/2021] [Indexed: 12/30/2022]
Abstract
BACKGROUND Aortic aneurysms (AA) are pathological dilations of the aorta, associated with an overall mortality rate up to 90% in case of rupture. In addition to dilation, the aortic layers can separate by a tear within the layers, defined as aortic dissections (AD). Vascular smooth muscle cells (vSMC) are the predominant cell type within the aortic wall and dysregulation of vSMC functions contributes to AA and AD development and progression. However, since the exact underlying mechanism is poorly understood, finding potential therapeutic targets for AA and AD is challenging and surgery remains the only treatment option. METHODS In this review, we summarize current knowledge about vSMC functions within the aortic wall and give an overview of how vSMC functions are altered in AA and AD pathogenesis, organized per anatomical location (abdominal or thoracic aorta). RESULTS Important functions of vSMC in healthy or diseased conditions are apoptosis, phenotypic switch, extracellular matrix regeneration and degradation, proliferation and contractility. Stressors within the aortic wall, including inflammatory cell infiltration and (epi)genetic changes, modulate vSMC functions and cause disturbance of processes within vSMC, such as changes in TGF-β signalling and regulatory RNA expression. CONCLUSION This review underscores a central role of vSMC dysfunction in abdominal and thoracic AA and AD development and progression. Further research focused on vSMC dysfunction in the aortic wall is necessary to find potential targets for noninvasive AA and AD treatment options.
Collapse
Affiliation(s)
- Karlijn B Rombouts
- Department of Surgery, Amsterdam University Medical Centers, Amsterdam Cardiovascular Sciences, Location VU Medical Center and AMC, Amsterdam, The Netherlands.,Department of Physiology, Amsterdam University Medical Centers, Amsterdam Cardiovascular Sciences, Location VU Medical Center, Amsterdam, The Netherlands
| | - Tara A R van Merrienboer
- Department of Surgery, Amsterdam University Medical Centers, Amsterdam Cardiovascular Sciences, Location VU Medical Center and AMC, Amsterdam, The Netherlands.,Department of Physiology, Amsterdam University Medical Centers, Amsterdam Cardiovascular Sciences, Location VU Medical Center, Amsterdam, The Netherlands
| | | | - Natalija Bogunovic
- Department of Surgery, Amsterdam University Medical Centers, Amsterdam Cardiovascular Sciences, Location VU Medical Center and AMC, Amsterdam, The Netherlands.,Department of Physiology, Amsterdam University Medical Centers, Amsterdam Cardiovascular Sciences, Location VU Medical Center, Amsterdam, The Netherlands.,Laboratory of Experimental Cardiology, Department of Cardiology, Leiden University Medical Center, Leiden, The Netherlands
| | - Jolanda van der Velden
- Department of Physiology, Amsterdam University Medical Centers, Amsterdam Cardiovascular Sciences, Location VU Medical Center, Amsterdam, The Netherlands
| | - Kak Khee Yeung
- Department of Surgery, Amsterdam University Medical Centers, Amsterdam Cardiovascular Sciences, Location VU Medical Center and AMC, Amsterdam, The Netherlands.,Department of Physiology, Amsterdam University Medical Centers, Amsterdam Cardiovascular Sciences, Location VU Medical Center, Amsterdam, The Netherlands
| |
Collapse
|
12
|
Si K, Lu D, Tian J. Integrated analysis and the identification of a circRNA-miRNA-mRNA network in the progression of abdominal aortic aneurysm. PeerJ 2022; 9:e12682. [PMID: 35036156 PMCID: PMC8711282 DOI: 10.7717/peerj.12682] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Accepted: 12/02/2021] [Indexed: 11/20/2022] Open
Abstract
Background Abdominal aortic aneurysm (AAA) is a disease commonly seen in the elderly. The aneurysm diameter increases yearly, and the larger the AAA the higher the risk of rupture, increasing the risk of death. However, there are no current effective interventions in the early stages of AAA. Methods Four gene expression profiling datasets, including 23 normal artery (NOR) tissue samples and 97 AAA tissue samples, were integrated in order to explore potential molecular biological targets for early intervention. After preprocessing, differentially expressed genes (DEGs) between AAA and NOR were identified using LIMMA package. Gene Ontology and Kyoto Encyclopedia of Genes and Genomes analysis were conducted using the DAVID database. The protein-protein interaction network was constructed and hub genes were identified using the STRING database and plugins in Cytoscape. A circular RNA (circRNA) profile of four NOR tissues versus four AAA tissues was then reanalyzed. A circRNA-miRNA-mRNA interaction network was constructed after predictions were made using the Targetscan and Circinteractome databases. Results A total of 440 DEGs (263 up-regulated and 177 down-regulated) were identified in the AAA group, compared with the NOR group. The majority were associated with the extracellular matrix, tumor necrosis factor-α, and transforming growth factor-β. Ten hub gene-encoded proteins (namely IL6, RPS27A, JUN, UBC, UBA52, FOS, IL1B, MMP9, SPP1 and CCL2) coupled with a higher degree of connectivity hub were identified after protein‐protein interaction network analysis. Our results, in combination with the results of previous studies revealed that miR-635, miR-527, miR-520h, miR-938 and miR-518a-5p may be affected by circ_0005073 and impact the expression of hub genes such as CCL2, SPP1 and UBA52. The miR-1206 may also be affected by circ_0090069 and impact RPS27A expression. Conclusions This circRNA-miRNA-mRNA network may perform critical roles in AAA and may be a novel target for early intervention.
Collapse
Affiliation(s)
- Ke Si
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, People's Republic of China
| | - Da Lu
- Department of Vascular Surgery, Shanghai General Hospital, Shanghai, People's Republic of China
| | - Jianbo Tian
- Institute of Information Engineering, Chinese Academy of Sciences, Beijing, People's Republic of China
| |
Collapse
|
13
|
Chronic Intermittent Hypoxia Regulates CaMKII-Dependent MAPK Signaling to Promote the Initiation of Abdominal Aortic Aneurysm. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2021:2502324. [PMID: 34970414 PMCID: PMC8714336 DOI: 10.1155/2021/2502324] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Revised: 11/15/2021] [Accepted: 11/24/2021] [Indexed: 02/05/2023]
Abstract
Obstructive sleep apnea (OSA) is highly prevalent in patients with abdominal aortic aneurysm (AAA). However, the effects of OSA on AAA initiation in a murine model of sleep apnea have not been completely studied. In this paper, Apoe−/− C57BL/6 mice infused with angiotensin II (Ang II) were placed in chronic intermittent hypoxia (CIH) condition for inducing OSA-related AAA. CIH significantly promoted the incidence of AAA and inhibited the survival of mice. By performing ultrasonography and elastic Van Gieson staining, CIH was found to be effective in promoting aortic dilation and elastin degradation. Immunohistochemical and zymography results show that CIH upregulated the expression and activity of MMP2 and MMP9 and upregulated MCP1 expression while downregulating α-SMA expression. Also, CIH exposure promoted ROS generation, apoptosis, and mitochondria damage in vascular smooth muscle cells (VSMCs), which were measured by ROS assay, TUNEL staining, and transmission electron microscopy. The result of RNA sequencing of mouse aortas displayed that 232 mRNAs were differently expressed between Ang II and Ang II+CIH groups, and CaMKII-dependent p38/Jnk was confirmed as one downstream signaling of CIH. CaMKII-IN-1, an inhibitor of CaMKII, eliminated the effects of CIH on the loss of primary VSMCs. To conclude, a mouse model of OSA-related AAA, which contains the phenotypes of both AAA and OSA, was established in this study. We suggested CIH as a risk factor of AAA initiation through CaMKII-dependent MAPK signaling.
Collapse
|
14
|
Tanaka H, Xu B, Xuan H, Ge Y, Wang Y, Li Y, Wang W, Guo J, Zhao S, Glover KJ, Zheng X, Liu S, Inuzuka K, Fujimura N, Furusho Y, Ikezoe T, Shoji T, Wang L, Fu W, Huang J, Unno N, Dalman RL. Recombinant Interleukin-19 Suppresses the Formation and Progression of Experimental Abdominal Aortic Aneurysms. J Am Heart Assoc 2021; 10:e022207. [PMID: 34459250 PMCID: PMC8649236 DOI: 10.1161/jaha.121.022207] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Background Interleukin-19 is an immunosuppressive cytokine produced by immune and nonimmune cells, but its role in abdominal aortic aneurysm (AAA) pathogenesis is not known. This study aimed to investigate interleukin-19 expression in, and influences on, the formation and progression of experimental AAAs. Methods and Results Human specimens were obtained at aneurysm repair surgery or from transplant donors. Experimental AAAs were created in 10- to 12-week-old male mice via intra-aortic elastase infusion. Influence and potential mechanisms of interleukin-19 treatment on AAAs were assessed via ultrasonography, histopathology, flow cytometry, and gene expression profiling. Immunohistochemistry revealed augmented interleukin-19 expression in both human and experimental AAAs. In mice, interleukin-19 treatment before AAA initiation via elastase infusion suppressed aneurysm formation and progression, with attenuation of medial elastin degradation, smooth-muscle depletion, leukocyte infiltration, neoangiogenesis, and matrix metalloproteinase 2 and 9 expression. Initiation of interleukin-19 treatment after AAA creation limited further aneurysmal degeneration. In additional experiments, interleukin-19 treatment inhibited murine macrophage recruitment following intraperitoneal thioglycolate injection. In classically or alternatively activated macrophages in vitro, interleukin-19 downregulated mRNA expression of inducible nitric oxide synthase, chemokine C-C motif ligand 2, and metalloproteinases 2 and 9 without apparent effect on cytokine-expressing helper or cytotoxic T-cell differentiation, nor regulatory T cellularity, in the aneurysmal aorta or spleen of interleukin-19-treated mice. Interleukin-19 also suppressed AAAs created via angiotensin II infusion in hyperlipidemic mice. Conclusions Based on human evidence and experimental modeling observations, interleukin-19 may influence the development and progression of AAAs.
Collapse
Affiliation(s)
- Hiroki Tanaka
- Divison of Vascular Surgery Department of Surgery Stanford University School of Medicine Stanford CA.,Division of Vascular Surgery Hamamatsu University School of Medicine Hamamatsu Shizuoka Japan
| | - Baohui Xu
- Divison of Vascular Surgery Department of Surgery Stanford University School of Medicine Stanford CA
| | - Haojun Xuan
- Divison of Vascular Surgery Department of Surgery Stanford University School of Medicine Stanford CA
| | - Yingbin Ge
- Department of Physiology Nanjing Medical University Nanjing Jiangsu China
| | - Yan Wang
- Peking University Third HospitalMedical Research Center Haidian Beijing China
| | - Yankui Li
- Divison of Vascular Surgery Department of Surgery Stanford University School of Medicine Stanford CA
| | - Wei Wang
- Department of Surgery Xiangya HospitalSouth Central University School of Medicine Changsha Hunan China
| | - Jia Guo
- Divison of Vascular Surgery Department of Surgery Stanford University School of Medicine Stanford CA
| | - Sihai Zhao
- Divison of Vascular Surgery Department of Surgery Stanford University School of Medicine Stanford CA
| | - Keith J Glover
- Divison of Vascular Surgery Department of Surgery Stanford University School of Medicine Stanford CA
| | - Xiaoya Zheng
- Divison of Vascular Surgery Department of Surgery Stanford University School of Medicine Stanford CA
| | - Shuai Liu
- Department of Surgery Xiangya HospitalSouth Central University School of Medicine Changsha Hunan China
| | - Kazunori Inuzuka
- Division of Vascular Surgery Hamamatsu University School of Medicine Hamamatsu Shizuoka Japan
| | - Naoki Fujimura
- Divison of Vascular Surgery Department of Surgery Stanford University School of Medicine Stanford CA
| | - Yuko Furusho
- Divison of Vascular Surgery Department of Surgery Stanford University School of Medicine Stanford CA
| | - Toru Ikezoe
- Divison of Vascular Surgery Department of Surgery Stanford University School of Medicine Stanford CA
| | - Takahiro Shoji
- Divison of Vascular Surgery Department of Surgery Stanford University School of Medicine Stanford CA
| | - Lixin Wang
- Department of Vascular Surgery Zhongshan HospitalFudan University Shanghai China
| | - Weiguo Fu
- Department of Vascular Surgery Zhongshan HospitalFudan University Shanghai China
| | - Jianhua Huang
- Department of Surgery Xiangya HospitalSouth Central University School of Medicine Changsha Hunan China
| | - Naoki Unno
- Division of Vascular Surgery Hamamatsu University School of Medicine Hamamatsu Shizuoka Japan
| | - Ronald L Dalman
- Divison of Vascular Surgery Department of Surgery Stanford University School of Medicine Stanford CA
| |
Collapse
|
15
|
Abstract
Abdominal aortic aneurysm (AAA) is a common disease associated with significant cardiovascular morbidity and mortality. Up to now, there is still controversy on the choice of treatment method of AAA. Even so, the mechanisms of AAA progression are poorly defined, making targeting new therapies problematic. Current evidence favors an interaction of the hemodynamic microenvironment with local and systemic immune responses. In this review, we aim to provide an update of mechanisms in AAA progression, involving hemodynamics, perivascular adipose tissue, adventitial fibroblasts, vasa vasorum remodeling, intraluminal thrombus, and distribution of macrophage subtypes.
Collapse
Affiliation(s)
- Jiang-Ping Gao
- Department of Vascular Surgery, Medical School of Chinese PLA, Chinese PLA General Hospital, Beijing, China
| | - Wei Guo
- Department of Vascular Surgery, Chinese PLA General Hospital, Beijing, China
| |
Collapse
|
16
|
Chiang MT, Chen IM, Hsu FF, Chen YH, Tsai MS, Hsu YW, Leu HB, Huang PH, Chen JW, Liu FT, Chen YH, Chau LY. Gal-1 (Galectin-1) Upregulation Contributes to Abdominal Aortic Aneurysm Progression by Enhancing Vascular Inflammation. Arterioscler Thromb Vasc Biol 2021; 41:331-345. [PMID: 33147994 DOI: 10.1161/atvbaha.120.315398] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
OBJECTIVE Abdominal aortic aneurysm (AAA) is a vascular degenerative disease causing sudden rupture of aorta and significant mortality in elders. Nevertheless, no prognostic and therapeutic target is available for disease management. Gal-1 (galectin-1) is a β-galactoside-binding lectin constitutively expressed in vasculature with roles in maintaining vascular homeostasis. This study aims to investigate the potential involvement of Gal-1 in AAA progression. Approach and Results: Gal-1 was significantly elevated in circulation and aortic tissues of Ang II (angiotensin II)-infused apoE-deficient mice developing AAA. Gal-1 deficiency reduced incidence and severity of AAA with lower expression of aortic MMPs (matrix metalloproteases) and proinflammatory cytokines. TNFα (tumor necrosis factor alpha) induced Gal-1 expression in cultured vascular smooth muscle cells and adventitial fibroblasts. Gal-1 deletion enhanced TNFα-induced MMP9 expression in fibroblasts but not vascular smooth muscle cells. Cysteinyl-labeling assay demonstrated that aortic Gal-1 exhibited susceptibility to oxidation in vivo. Recombinant oxidized Gal-1 induced expression of MMP9 and inflammatory cytokines to various extents in macrophages, vascular smooth muscle cells, and fibroblasts through activation of MAP (mitogen-activated protein) kinase signaling. Clinically, serum MMP9 level was significantly higher in both patients with AAA and coronary artery disease than in control subjects, whereas serum Gal-1 level was elevated in patients with AAA but not coronary artery disease when compared with controls. CONCLUSIONS Gal-1 is highly induced and contributes to AAA by enhancing matrix degradation activity and inflammatory responses in experimental model. The pathological link between Gal-1 and AAA is also observed in human patients. These findings support the potential of Gal-1 as a disease biomarker and therapeutic target of AAA.
Collapse
MESH Headings
- Adventitia/metabolism
- Adventitia/pathology
- Angiotensin II
- Animals
- Aorta, Abdominal/metabolism
- Aorta, Abdominal/pathology
- Aortic Aneurysm, Abdominal/chemically induced
- Aortic Aneurysm, Abdominal/metabolism
- Aortic Aneurysm, Abdominal/pathology
- Aortitis/chemically induced
- Aortitis/metabolism
- Aortitis/pathology
- Case-Control Studies
- Cells, Cultured
- Cytokines/metabolism
- Disease Models, Animal
- Disease Progression
- Extracellular Matrix/metabolism
- Extracellular Matrix/pathology
- Fibroblasts/metabolism
- Fibroblasts/pathology
- Galectin 1/blood
- Galectin 1/deficiency
- Galectin 1/genetics
- Galectin 1/metabolism
- Humans
- Inflammation Mediators/metabolism
- Macrophages, Peritoneal/metabolism
- Macrophages, Peritoneal/pathology
- Male
- Matrix Metalloproteinase 9/metabolism
- Mice, Inbred C57BL
- Mice, Knockout, ApoE
- Mitogen-Activated Protein Kinases/metabolism
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/pathology
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/pathology
- Signal Transduction
- Up-Regulation
- Vascular Remodeling
- Mice
Collapse
Affiliation(s)
- Ming-Tsai Chiang
- Division of Cardiovascular Research, Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan (M.-T.C., F.-F.H., Yen-Hui Chen, M.-S.T., Y.-W.H., F.-T.L., L.-Y.C.)
| | - I-Ming Chen
- Division of Cardiovascular Surgery, Department of Surgery (I.-M.C.), Taipei Veterans General Hospital, Taiwan
| | - Fu-Fei Hsu
- Division of Cardiovascular Research, Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan (M.-T.C., F.-F.H., Yen-Hui Chen, M.-S.T., Y.-W.H., F.-T.L., L.-Y.C.)
| | - Yen-Hui Chen
- Division of Cardiovascular Research, Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan (M.-T.C., F.-F.H., Yen-Hui Chen, M.-S.T., Y.-W.H., F.-T.L., L.-Y.C.)
| | - Min-Shao Tsai
- Division of Cardiovascular Research, Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan (M.-T.C., F.-F.H., Yen-Hui Chen, M.-S.T., Y.-W.H., F.-T.L., L.-Y.C.)
| | - Yaw-Wen Hsu
- Division of Cardiovascular Research, Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan (M.-T.C., F.-F.H., Yen-Hui Chen, M.-S.T., Y.-W.H., F.-T.L., L.-Y.C.)
| | - Hsin-Bang Leu
- Division of Healthcare and Management, Healthcare Center (H.-B.L.), Taipei Veterans General Hospital, Taiwan
- Department of Medicine, School of Medicine (H.-B.L., Ying-Hwa Chen), National Yang-Ming University, Taipei, Taiwan
| | - Po-Hsun Huang
- Division of Cardiology, Department of Internal Medicine (P.-H.H., J.-W.C., Ying-Hwa Chen), Taipei Veterans General Hospital, Taiwan
- Institute of Clinical Medicine (P.-H.H.), National Yang-Ming University, Taipei, Taiwan
| | - Jaw-Wen Chen
- Division of Cardiology, Department of Internal Medicine (P.-H.H., J.-W.C., Ying-Hwa Chen), Taipei Veterans General Hospital, Taiwan
- Institute of Pharmacology (J.-W.C.), National Yang-Ming University, Taipei, Taiwan
| | - Fu-Tong Liu
- Division of Cardiovascular Research, Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan (M.-T.C., F.-F.H., Yen-Hui Chen, M.-S.T., Y.-W.H., F.-T.L., L.-Y.C.)
| | - Ying-Hwa Chen
- Division of Cardiology, Department of Internal Medicine (P.-H.H., J.-W.C., Ying-Hwa Chen), Taipei Veterans General Hospital, Taiwan
- Department of Medicine, School of Medicine (H.-B.L., Ying-Hwa Chen), National Yang-Ming University, Taipei, Taiwan
| | - Lee-Young Chau
- Division of Cardiovascular Research, Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan (M.-T.C., F.-F.H., Yen-Hui Chen, M.-S.T., Y.-W.H., F.-T.L., L.-Y.C.)
| |
Collapse
|
17
|
Knappich C, Spin JM, Eckstein HH, Tsao PS, Maegdefessel L. Involvement of Myeloid Cells and Noncoding RNA in Abdominal Aortic Aneurysm Disease. Antioxid Redox Signal 2020; 33:602-620. [PMID: 31989839 PMCID: PMC7455479 DOI: 10.1089/ars.2020.8035] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Significance: Abdominal aortic aneurysm (AAA) is a potentially fatal condition, featuring the possibility of high-mortality rupture. To date, prophylactic surgery by means of open surgical repair or endovascular aortic repair at specific thresholds is considered standard therapy. Both surgical options hold different risk profiles of short- and long-term morbidity and mortality. Targeting early stages of AAA development to decelerate disease progression is desirable. Recent Advances: Understanding the pathomechanisms that initiate formation, maintain growth, and promote rupture of AAA is crucial to developing new medical therapeutic options. Inflammatory cells, in particular macrophages, have been investigated for their contribution to AAA disease for decades, whereas evidence on lymphocytes, mast cells, and neutrophils is sparse. Recently, there has been increasing interest in noncoding RNAs (ncRNAs) and their involvement in disease development, including AAA. Critical Issues: The current evidence on myeloid cells and ncRNAs in AAA largely originates from small animal models, making clinical extrapolation difficult. Although it is feasible to collect surgical human AAA samples, these tissues reflect end-stage disease, preventing examination of critical mechanisms behind early AAA formation. Future Directions: Gaining more insight into how myeloid cells and ncRNAs contribute to AAA disease, particularly in early stages, might suggest nonsurgical AAA treatment options. The utilization of large animal models might be helpful in this context to help bridge translational results to humans.
Collapse
Affiliation(s)
- Christoph Knappich
- Department for Vascular and Endovascular Surgery, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Joshua M Spin
- Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, California, USA
| | - Hans-Henning Eckstein
- Department for Vascular and Endovascular Surgery, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Philip S Tsao
- Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, California, USA
| | - Lars Maegdefessel
- Department for Vascular and Endovascular Surgery, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany.,Department of Medicine, Karolinska Institute, Stockholm, Sweden
| |
Collapse
|
18
|
Sharma N, Belenchia AM, Toedebusch R, Pulakat L, Hans CP. AT2R agonist NP-6A4 mitigates aortic stiffness and proteolytic activity in mouse model of aneurysm. J Cell Mol Med 2020; 24:7393-7404. [PMID: 32420690 PMCID: PMC7339180 DOI: 10.1111/jcmm.15342] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2019] [Revised: 03/16/2020] [Accepted: 03/26/2020] [Indexed: 12/12/2022] Open
Abstract
Clinical and experimental studies show that angiotensin II (AngII) promotes vascular pathology via activation of AngII type 1 receptors (AT1Rs). We recently reported that NP-6A4, a selective peptide agonist for AngII type 2 receptor (AT2R), exerts protective effects on human vascular cells subjected to serum starvation or doxorubicin exposure. In this study, we investigated whether NP-6A4-induced AT2R activation could mitigate AngII-induced abdominal aortic aneurism (AAA) using AngII-treated Apoe-/- mice. Male Apoe-/- mice were infused with AngII (1 µg/kg/min) by implanting osmotic pumps subcutaneously for 28 days. A subset of mice was pre-treated subcutaneously with NP-6A4 (2.5 mg/kg/day) or vehicle for 14 days prior to AngII, and treatments were continued for 28 days. NP-6A4 significantly reduced aortic stiffness of the abdominal aorta induced by AngII as determined by ultrasound functional analyses and histochemical analyses. NP-6A4 also increased nitric oxide bioavailability in aortic tissues and suppressed AngII-induced increases in monocyte chemotactic protein-1, osteopontin and proteolytic activity of the aorta. However, NP-6A4 did not affect maximal intraluminal aortic diameter or AAA incidences significantly. These data suggest that the effects of AT2R agonist on vascular pathologies are selective, affecting the aortic stiffness and proteolytic activity without affecting the size of AAA.
Collapse
Affiliation(s)
- Neekun Sharma
- Department of Cardiovascular Medicine, University of Missouri, Columbia, MO, USA.,Dalton Cardiovascular Research Center, University of Missouri, Columbia, MO, USA
| | - Anthony M Belenchia
- Department of Cardiovascular Medicine, University of Missouri, Columbia, MO, USA.,Dalton Cardiovascular Research Center, University of Missouri, Columbia, MO, USA
| | - Ryan Toedebusch
- Department of Cardiovascular Medicine, University of Missouri, Columbia, MO, USA.,Dalton Cardiovascular Research Center, University of Missouri, Columbia, MO, USA
| | - Lakshmi Pulakat
- Department of Cardiovascular Medicine, University of Missouri, Columbia, MO, USA.,Dalton Cardiovascular Research Center, University of Missouri, Columbia, MO, USA.,Medical Pharmacology and Physiology, University of Missouri, Columbia, MO, USA.,Molecular Cardiology Research Institute, Department of Medicine, Tufts Medical Center, Tufts University School of Medicine, Boston, MA, USA
| | - Chetan P Hans
- Department of Cardiovascular Medicine, University of Missouri, Columbia, MO, USA.,Dalton Cardiovascular Research Center, University of Missouri, Columbia, MO, USA.,Medical Pharmacology and Physiology, University of Missouri, Columbia, MO, USA
| |
Collapse
|
19
|
Tumor Necrosis Factor-Like Weak Inducer of Apoptosis (TWEAK)/Fibroblast Growth Factor-Inducible 14 (Fn14) Axis in Cardiovascular Diseases: Progress and Challenges. Cells 2020; 9:cells9020405. [PMID: 32053869 PMCID: PMC7072601 DOI: 10.3390/cells9020405] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Revised: 02/06/2020] [Accepted: 02/07/2020] [Indexed: 12/12/2022] Open
Abstract
Cardiovascular diseases (CVD) are the leading cause of mortality in Western countries. CVD include several pathologies, such as coronary artery disease, stroke, peripheral artery disease, and aortic aneurysm, among others. All of them are characterized by a pathological vascular remodeling in which inflammation plays a key role. Interaction between different members of the tumor necrosis factor superfamily and their cognate receptors induce several biological actions that may participate in CVD. The cytokine tumor necrosis factor-like weak inducer of apoptosis (TWEAK) and its functional receptor, fibroblast growth factor-inducible 14 (Fn14), are abundantly expressed during pathological cardiovascular remodeling. The TWEAK/Fn14 axis controls a variety of cellular functions, such as proliferation, differentiation, and apoptosis, and has several biological functions, such as inflammation and fibrosis that are linked to CVD. It has been demonstrated that persistent TWEAK/Fn14 activation is involved in both vessel and heart remodeling associated with acute and chronic CVD. In this review, we summarized the role of the TWEAK/Fn14 axis during pathological cardiovascular remodeling, highlighting the cellular components and the signaling pathways that are involved in these processes.
Collapse
|
20
|
Smooth muscle-specific Gsα deletion exaggerates angiotensin II-induced abdominal aortic aneurysm formation in mice in vivo. J Mol Cell Cardiol 2019; 132:49-59. [PMID: 31071332 PMCID: PMC7394040 DOI: 10.1016/j.yjmcc.2019.05.002] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/19/2018] [Revised: 04/16/2019] [Accepted: 05/02/2019] [Indexed: 01/12/2023]
Abstract
Objective: Abdominal aortic aneurysm (AAA) is a life-threatening vascular disease without an effective pharmaceutical treatment. Genetic studies have proved the involvement of smooth muscle phenotype switch in the development of AAA. The alpha subunit of the heterotrimeric G stimulatory protein (Gsα) mediates receptor-stimulated production of cyclic adenosine monophosphate (cAMP). However, the role of smooth muscle Gsα in AAA formation remains unknown. Approach and results: In this study, mice with knockout of smooth muscle-specific Gsα (GsαSMKO) were generated by cross-breeding Gsαflox/flox mice with SM22-CreERT2 transgenic mice, induced in adult mice by tamoxifen treatment. Gsα deficiency induced a smooth muscle phenotype switch from a contractile to a synthetic state. Mechanically, Gsα deletion reduced cAMP level and increased the level of human antigen R (HuR), which binds with the adenylate uridylate–rich elements of the 3′ untranslated region of Krüppel-like factor 4 (KLF4) mRNA, thereby increasing the stability of KLF4. Moreover, genetic knockdown of HuR or KLF4 rescued the phenotype switch in Gsα-deficient smooth muscle cells. Furthermore, with acute infusion of angiotensin II, the incidence of AAA was markedly higher in ApoE−/−/GsαSMKO than ApoE−/−/Gsαflox/flox mice and induced increased elastic lamina degradation and aortic expansion. Finally, the levels of Gsα and SM α-actin were significantly lower while those of HuR and KLF4 were higher in human AAA samples than adjacent nonaneurysmal aortic sections. Conclusions: Gsα may play a protective role in AAA formation by regulating the smooth muscle phenotype switch and could be a potential therapeutic target for AAA disease.
Collapse
|
21
|
Salmon M, Spinosa M, Zehner ZE, Upchurch GR, Ailawadi G. Klf4, Klf2, and Zfp148 activate autophagy-related genes in smooth muscle cells during aortic aneurysm formation. Physiol Rep 2019; 7:e14058. [PMID: 31025534 PMCID: PMC6483937 DOI: 10.14814/phy2.14058] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2018] [Revised: 02/21/2019] [Accepted: 02/22/2019] [Indexed: 01/08/2023] Open
Abstract
Abdominal aortic aneurysms (AAAs) are a progressive dilation of the aorta that is characterized by an initial influx of inflammatory cells followed by a pro-inflammatory, migratory, proliferative, and eventually apoptotic smooth muscle cell phenotype. In recent years, the mechanisms related to the initial influx of inflammatory cells have become well-studied; the mechanisms related to chronic aneurysm formation, smooth muscle cell apoptosis and death are less well-characterized. Autophagy is a generally believed to be a protective cellular mechanism that functions to recycle defective proteins and cellular organelles to maintain cellular homeostasis. Our goal with the present study was to investigate the role of autophagy in smooth muscle cells during AAA formation. Levels of the autophagy factors, Beclin, and LC3 were elevated in human and mouse AAA tissue via both qPCR and immunohistochemical analysis. Confocal staining in human and mouse AAA tissue demonstrated Beclin and LC3 were present in smooth muscle cells during AAA formation. Treatment of smooth muscle cells with porcine pancreatic elastase or interleukin (IL)-1β activated autophagy-related genes in vitro while treatment with a siRNA to Kruppel-like transcription factor 4 (Klf4), Kruppel-like transcription factor 2 (Klf2) or Zinc-finger protein 148 (Zfp148) separately inhibited activation of autophagy genes. Chromatin immunoprecipitation assays demonstrated that Klf4, Klf2, and Zfp148 separately bind autophagy genes in smooth muscle cells following elastase treatment. These results demonstrate that autophagy is an important mechanism related to Klfs in smooth muscle cells during AAA formation.
Collapse
Affiliation(s)
- Morgan Salmon
- Department of SurgeryUniversity of Virginia School of MedicineCharlottesvilleVirginiaUSA
| | - Michael Spinosa
- Department of SurgeryUniversity of Virginia School of MedicineCharlottesvilleVirginiaUSA
| | - Zendra E. Zehner
- Department of BiochemistryVirginia Commonwealth University Medical CenterRichmondVirginiaUSA
| | | | - Gorav Ailawadi
- Department of SurgeryUniversity of Virginia School of MedicineCharlottesvilleVirginiaUSA
- The Robert M. Berne Cardiovascular Research CenterUniversity of Virginia School of MedicineCharlottesvilleVirginiaUSA
| |
Collapse
|
22
|
Wu W, Zhang W, Choi M, Zhao J, Gao P, Xue M, Singer HA, Jourd'heuil D, Long X. Vascular smooth muscle-MAPK14 is required for neointimal hyperplasia by suppressing VSMC differentiation and inducing proliferation and inflammation. Redox Biol 2019; 22:101137. [PMID: 30771750 PMCID: PMC6377391 DOI: 10.1016/j.redox.2019.101137] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2019] [Accepted: 02/05/2019] [Indexed: 12/19/2022] Open
Abstract
Injury-induced stenosis is a serious vascular complication. We previously reported that p38α (MAPK14), a redox-regulated p38MAPK family member was a negative regulator of the VSMC contractile phenotype in vitro. Here we evaluated the function of VSMC-MAPK14 in vivo in injury-induced neointima hyperplasia and the underlying mechanism using an inducible SMC-MAPK14 knockout mouse line (iSMC-MAPK14-/-). We show that MAPK14 expression and activity were induced in VSMCs after carotid artery ligation injury in mice and ex vivo cultured human saphenous veins. While the vasculature from iSMC-MAPK14-/- mice was indistinguishable from wildtype littermate controls at baseline, these mice exhibited reduced neointima formation following carotid artery ligation injury. Concomitantly, there was an increased VSMC contractile protein expression in the injured vessels and a decrease in proliferating cells. Blockade of MAPK14 through a selective inhibitor suppressed, while activation of MAPK14 by forced expression of an upstream MAPK14 kinase promoted VSMC proliferation in cultured VSMCs. Genome wide RNA array combined with VSMC lineage tracing studies uncovered that vascular injury evoked robust inflammatory responses including the activation of proinflammatory gene expression and accumulation of CD45 positive inflammatory cells, which were attenuated in iSMC-MAPK14-/- mice. Using multiple pharmacological and molecular approaches to manipulate MAPK14 pathway, we further confirmed the critical role of MAPK14 in activating proinflammatory gene expression in cultured VSMCs, which occurs in a p65/NFkB-dependent pathway. Finally, we found that NOX4 contributes to MAPK14 suppression of the VSMC contractile phenotype. Our results revealed that VSMC-MAPK14 is required for injury-induced neointima formation, likely through suppressing VSMC differentiation and promoting VSMC proliferation and inflammation. Our study will provide mechanistic insights into therapeutic strategies for mitigation of vascular stenosis.
Collapse
Affiliation(s)
- Wen Wu
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY, United States
| | - Wei Zhang
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY, United States
| | - Mihyun Choi
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY, United States
| | - Jinjing Zhao
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY, United States
| | - Ping Gao
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY, United States
| | - Min Xue
- Feed Research Institute, Chinese Academy of Agricultural Sciences, Beijing, PR China
| | - Harold A Singer
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY, United States
| | - David Jourd'heuil
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY, United States
| | - Xiaochun Long
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY, United States.
| |
Collapse
|
23
|
Hibender S, Wanga S, van der Made I, Vos M, Mulder BJM, Balm R, de Vries CJM, de Waard V. Renal cystic disease in the Fbn1C1039G/+ Marfan mouse is associated with enhanced aortic aneurysm formation. Cardiovasc Pathol 2019; 38:1-6. [DOI: 10.1016/j.carpath.2018.10.002] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/12/2018] [Revised: 10/02/2018] [Accepted: 10/03/2018] [Indexed: 12/24/2022] Open
|
24
|
Hadi T, Boytard L, Silvestro M, Alebrahim D, Jacob S, Feinstein J, Barone K, Spiro W, Hutchison S, Simon R, Rateri D, Pinet F, Fenyo D, Adelman M, Moore KJ, Eltzschig HK, Daugherty A, Ramkhelawon B. Macrophage-derived netrin-1 promotes abdominal aortic aneurysm formation by activating MMP3 in vascular smooth muscle cells. Nat Commun 2018; 9:5022. [PMID: 30479344 PMCID: PMC6258757 DOI: 10.1038/s41467-018-07495-1] [Citation(s) in RCA: 96] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2018] [Accepted: 11/06/2018] [Indexed: 12/22/2022] Open
Abstract
Abdominal aortic aneurysms (AAA) are characterized by extensive extracellular matrix (ECM) fragmentation and inflammation. However, the mechanisms by which these events are coupled thereby fueling focal vascular damage are undefined. Here we report through single-cell RNA-sequencing of diseased aorta that the neuronal guidance cue netrin-1 can act at the interface of macrophage-driven injury and ECM degradation. Netrin-1 expression peaks in human and murine aneurysmal macrophages. Targeted deletion of netrin-1 in macrophages protects mice from developing AAA. Through its receptor neogenin-1, netrin-1 induces a robust intracellular calcium flux necessary for the transcriptional regulation and persistent catalytic activation of matrix metalloproteinase-3 (MMP3) by vascular smooth muscle cells. Deficiency in MMP3 reduces ECM damage and the susceptibility of mice to develop AAA. Here, we establish netrin-1 as a major signal that mediates the dynamic crosstalk between inflammation and chronic erosion of the ECM in AAA. Abdominal aortic aneurysms (AAA) are characterized by extensive extracellular matrix degradation. Here Hadi et al. identify a netrin-1/neogenin-based crosstalk between macrophages and vascular smooth muscle cells (VSMCs), leading to the secretion of the matrix metalloproteinase MMP-3 by VSMCs and subsequent matrix degradation in AAA lesions.
Collapse
Affiliation(s)
- Tarik Hadi
- Division of Vascular Surgery, Department of Surgery, New York University Medical Center, New York, NY, 10016, USA
| | - Ludovic Boytard
- Division of Vascular Surgery, Department of Surgery, New York University Medical Center, New York, NY, 10016, USA
| | - Michele Silvestro
- Division of Vascular Surgery, Department of Surgery, New York University Medical Center, New York, NY, 10016, USA
| | - Dornazsadat Alebrahim
- Division of Vascular Surgery, Department of Surgery, New York University Medical Center, New York, NY, 10016, USA
| | - Samson Jacob
- Department of Biochemistry and Molecular Pharmacology, New York University School of Medicine, New York, NY, 10016, USA
| | - Jordyn Feinstein
- Division of Vascular Surgery, Department of Surgery, New York University Medical Center, New York, NY, 10016, USA
| | - Krista Barone
- Division of Vascular Surgery, Department of Surgery, New York University Medical Center, New York, NY, 10016, USA
| | - Wes Spiro
- Leon H. Charney Division of Cardiology, Department of Medicine, New York University School of Medicine, New York, NY, 10016, USA
| | - Susan Hutchison
- Leon H. Charney Division of Cardiology, Department of Medicine, New York University School of Medicine, New York, NY, 10016, USA
| | - Russell Simon
- Division of Vascular Surgery, Department of Surgery, New York University Medical Center, New York, NY, 10016, USA
| | - Debra Rateri
- Department of Physiology and Saha Cardiovascular Research Center, University of Kentucky, Lexington, KY, 40506, USA
| | - Florence Pinet
- University of Lille, Inserm U1167, Institut Pasteur de Lille, 59019, Lille, France
| | - David Fenyo
- Department of Biochemistry and Molecular Pharmacology, New York University School of Medicine, New York, NY, 10016, USA
| | - Mark Adelman
- Division of Vascular Surgery, Department of Surgery, New York University Medical Center, New York, NY, 10016, USA
| | - Kathryn J Moore
- Leon H. Charney Division of Cardiology, Department of Medicine, New York University School of Medicine, New York, NY, 10016, USA
| | - Holger K Eltzschig
- Department of Anesthesiology, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX, 77030, USA
| | - Alan Daugherty
- Department of Physiology and Saha Cardiovascular Research Center, University of Kentucky, Lexington, KY, 40506, USA
| | - Bhama Ramkhelawon
- Division of Vascular Surgery, Department of Surgery, New York University Medical Center, New York, NY, 10016, USA. .,Department of Cell Biology, New York University Medical Center, New York, NY, 10016, USA.
| |
Collapse
|
25
|
van Puijvelde GHM, Foks AC, van Bochove RE, Bot I, Habets KLL, de Jager SC, ter Borg MND, van Osch P, Boon L, Vos M, de Waard V, Kuiper J. CD1d deficiency inhibits the development of abdominal aortic aneurysms in LDL receptor deficient mice. PLoS One 2018; 13:e0190962. [PMID: 29346401 PMCID: PMC5773169 DOI: 10.1371/journal.pone.0190962] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2017] [Accepted: 12/22/2017] [Indexed: 11/19/2022] Open
Abstract
An abdominal aortic aneurysm (AAA) is a dilatation of the abdominal aorta leading to serious complications and mostly to death. AAA development is associated with an accumulation of inflammatory cells in the aorta including NKT cells. An important factor in promoting the recruitment of these inflammatory cells into tissues and thereby contributing to the development of AAA is angiotensin II (Ang II). We demonstrate that a deficiency in CD1d dependent NKT cells under hyperlipidemic conditions (LDLr-/-CD1d-/- mice) results in a strong decline in the severity of angiotensin II induced aneurysm formation when compared with LDLr-/- mice. In addition, we show that Ang II amplifies the activation of NKT cells both in vivo and in vitro. We also provide evidence that type I NKT cells contribute to AAA development by inducing the expression of matrix degrading enzymes in vSMCs and macrophages, and by cytokine dependently decreasing vSMC viability. Altogether, these data prove that CD1d-dependent NKT cells contribute to AAA development in the Ang II-mediated aneurysm model by enhancing aortic degradation, establishing that therapeutic applications which target NKT cells can be a successful way to prevent AAA development.
Collapse
Affiliation(s)
- Gijs H. M. van Puijvelde
- Division of Biopharmaceutics, Leiden Academic Centre for Drug Research, Leiden University, Leiden, The Netherlands
- * E-mail:
| | - Amanda C. Foks
- Division of Biopharmaceutics, Leiden Academic Centre for Drug Research, Leiden University, Leiden, The Netherlands
| | - Rosemarie E. van Bochove
- Division of Biopharmaceutics, Leiden Academic Centre for Drug Research, Leiden University, Leiden, The Netherlands
| | - Ilze Bot
- Division of Biopharmaceutics, Leiden Academic Centre for Drug Research, Leiden University, Leiden, The Netherlands
| | - Kim L. L. Habets
- Division of Biopharmaceutics, Leiden Academic Centre for Drug Research, Leiden University, Leiden, The Netherlands
| | - Saskia C. de Jager
- Division of Biopharmaceutics, Leiden Academic Centre for Drug Research, Leiden University, Leiden, The Netherlands
| | - Mariëtte N. D. ter Borg
- Division of Biopharmaceutics, Leiden Academic Centre for Drug Research, Leiden University, Leiden, The Netherlands
| | - Puck van Osch
- Division of Biopharmaceutics, Leiden Academic Centre for Drug Research, Leiden University, Leiden, The Netherlands
| | | | - Mariska Vos
- Department of Medical Biochemistry, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Vivian de Waard
- Department of Medical Biochemistry, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Johan Kuiper
- Division of Biopharmaceutics, Leiden Academic Centre for Drug Research, Leiden University, Leiden, The Netherlands
| |
Collapse
|
26
|
Stimulation of Alpha7 Nicotinic Acetylcholine Receptor Attenuates Nicotine-Induced Upregulation of MMP, MCP-1, and RANTES through Modulating ERK1/2/AP-1 Signaling Pathway in RAW264.7 and MOVAS Cells. Mediators Inflamm 2017; 2017:2401027. [PMID: 29348704 PMCID: PMC5733626 DOI: 10.1155/2017/2401027] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2017] [Revised: 09/04/2017] [Accepted: 09/27/2017] [Indexed: 12/23/2022] Open
Abstract
Vagus nerve stimulation through alpha7 nicotine acetylcholine receptors (α7-nAChR) signaling had been demonstrated attenuation of inflammation. This study aimed to determine whether PNU-282987, a selective α7-nAChR agonist, affected activities of matrix metalloproteinase (MMP) and inflammatory cytokines in nicotine-treatment RAW264.7 and MOVAS cells and to assess the underlying molecular mechanisms. RAW264.7 and MOVAS cells were treated with nicotine at different concentrations (0, 1, 10, and 100 ng/ml) for 0–120 min. Nicotine markedly stimulated the phosphorylation of extracellular signal-regulated kinase1/2 (ERK1/2) and c-Jun in RAW264.7 cells. Pretreatment with U0126 significantly suppressed phosphorylation of ERK1/2 and further attenuated nicotine-induced activation of c-Jun and upregulation of MMP-2, MMP-9, monocyte chemotactic protein- (MCP-) 1, and regulated upon activation normal T cell expressed and secreted (RANTES). Similarly, nicotine treatment also increased phosphorylation of c-Jun and expressions of MMP-2, MMP-9, MCP-1, and RANTES in MOVAS cells. When cells were pretreated with PNU-282987, nicotine-induced activations of ERK1/2 and c-Jun in RAW264.7 cells and c-Jun in MOVAS cells were effectively inhibited. Furthermore, nicotine-induced secretions of MMP-2, MMP-9, MCP-1, and RANTES were remarkably downregulated. Treatment with α7-nAChR agonist inhibits nicotine-induced upregulation of MMP and inflammatory cytokines through modulating ERK1/2/AP-1 signaling in RAW264.7 cells and AP-1 in MOVAS cells, providing a new therapeutic for abdominal aortic aneurysm.
Collapse
|
27
|
The role of IL-6 in pathogenesis of abdominal aortic aneurysm in mice. PLoS One 2017; 12:e0185923. [PMID: 28982132 PMCID: PMC5628902 DOI: 10.1371/journal.pone.0185923] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2017] [Accepted: 09/21/2017] [Indexed: 11/19/2022] Open
Abstract
Although the pathogenesis of abdominal aortic aneurysm (AAA) remains unclear, evidence is accumulating to support a central role for inflammation. Inflammatory responses are coordinated by various soluble cytokines of which IL-6 is one of the major proinflammatory cytokines. In this study we examined the role of IL-6 in the pathogenesis of experimental AAA induced by a periaortic exposure to CaCl2 in mice. We now report that the administration of MR16-1, a neutralizing monoclonal antibody specific for the mouse IL-6 receptor, mildly suppressed the development of AAA. The inhibition of IL-6 signaling provoked by MR16-1 also resulted in a suppression of Stat3 activity. Conversely, no significant changes in either NFκB activity, Jnk activity or the expression of matrix metalloproteinases (Mmp) -2 and -9 were identified. Transcriptome analyses revealed that MR16-1-sensitive genes encode chemokines and their receptors, as well as factors that regulate vascular permeability and cell migration. Imaging cytometric analyses then consistently demonstrated reduced cellular infiltration for MR16-1-treated AAA. These results suggest that IL-6 plays an important but limited role in AAA pathogenesis, and primarily regulates cell migration and infiltration. These data would also suggest that IL-6 activity may play an important role in scenarios of continuous cellular infiltration, possibly including human AAA.
Collapse
|
28
|
Abstract
Abdominal aortic aneurysm (AAA) is a life-threatening disease associated with high morbidity, and high mortality in the event of aortic rupture. Major advances in open surgical and endovascular repair of AAA have been achieved during the past 2 decades. However, drug-based therapies are still lacking, highlighting a real need for better understanding of the molecular and cellular mechanisms involved in AAA formation and progression. The main pathological features of AAA include extracellular matrix remodelling associated with degeneration and loss of vascular smooth muscle cells and accumulation and activation of inflammatory cells. The inflammatory process has a crucial role in AAA and substantially influences many determinants of aortic wall remodelling. In this Review, we focus specifically on the involvement of monocytes and macrophages, summarizing current knowledge on the roles, origin, and functions of these cells in AAA development and its complications. Furthermore, we show and propose that distinct monocyte and macrophage subsets have critical and differential roles in initiation, progression, and healing of the aneurysmal process. On the basis of experimental and clinical studies, we review potential translational applications to detect, assess, and image macrophage subsets in AAA, and discuss the relevance of these applications for clinical practice.
Collapse
|
29
|
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: 6.6] [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
|
30
|
Ma D, Zheng B, Suzuki T, Zhang R, Jiang C, Bai D, Yin W, Yang Z, Zhang X, Hou L, Zhan H, Wen JK. Inhibition of KLF5-Myo9b-RhoA Pathway-Mediated Podosome Formation in Macrophages Ameliorates Abdominal Aortic Aneurysm. Circ Res 2017; 120:799-815. [PMID: 28115390 DOI: 10.1161/circresaha.116.310367] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/24/2016] [Revised: 01/15/2017] [Accepted: 01/23/2017] [Indexed: 11/16/2022]
Abstract
RATIONALE Abdominal aortic aneurysms (AAAs) are characterized by pathological remodeling of the aortic wall. Although both increased Krüppel-like factor 5 (KLF5) expression and macrophage infiltration have been implicated in vascular remodeling, the role of KLF5 in macrophage infiltration and AAA formation remains unclear. OBJECTIVE To determine the role of KLF5 in AAA formation and macrophage infiltration into AAAs. METHODS AND RESULTS KLF5 expression was significantly increased in human AAA tissues and in 2 mouse models of experimental AAA. Moreover, in myeloid-specific Klf5 knockout mice (myeKlf5-/- mice), macrophage infiltration, medial smooth muscle cell loss, elastin degradation, and AAA formation were markedly decreased. In cell migration and time-lapse imaging analyses, the migration of murine myeKlf5-/- macrophages was impaired, and in luciferase reporter assays, KLF5 activated Myo9b (myosin IXB) transcription by direct binding to the Myo9b promoter. In subsequent coimmunostaining studies, Myo9b was colocalized with filamentous actin, cortactin, vinculin, and Tks5 in the podosomes of phorbol 12,13-dibutyrate-treated macrophages, indicating that Myo9b participates in podosome formation. Gain- and loss-of-function experiments showed that KLF5 promoted podosome formation in macrophages by upregulating Myo9b expression. Furthermore, RhoA-GTP levels increased after KLF5 knockdown in macrophages, suggesting that KLF5 lies upstream of RhoA signaling. Finally, Myo9b expression was increased in human AAA tissues, located in macrophages, and positively correlated with AAA size. CONCLUSIONS These data are the first to indicate that KLF5-dependent regulation of Myo9b/RhoA is required for podosome formation and macrophage migration during AAA formation, warranting consideration of the KLF5-Myo9b-RhoA pathway as a therapeutic target for AAA treatment.
Collapse
Affiliation(s)
- Dong Ma
- From the Department of Biochemistry and Molecular Biology, The Key Laboratory of Neural and Vascular Biology, China Administration of Education, Hebei Medical University, China (D.M., B.Z., R.Z., C.J., D.B., W.Y., Z.Y., X.Z., L.H., J.-k.W.); School of Public Health, North China University of Science and Technology, China (D.M., D.B.); Department of Cardiovascular Sciences, University of Leicester, UK (T.S.); Department of Thoracic Surgery, Tianjin Union Medicine Centre, China (C.J.); Department of Cardiovascular Medicine, Jichi Medical University, Tochigi and Graduate School of Medicine, The University of Tokyo, Japan (H.Z.)
| | - Bin Zheng
- From the Department of Biochemistry and Molecular Biology, The Key Laboratory of Neural and Vascular Biology, China Administration of Education, Hebei Medical University, China (D.M., B.Z., R.Z., C.J., D.B., W.Y., Z.Y., X.Z., L.H., J.-k.W.); School of Public Health, North China University of Science and Technology, China (D.M., D.B.); Department of Cardiovascular Sciences, University of Leicester, UK (T.S.); Department of Thoracic Surgery, Tianjin Union Medicine Centre, China (C.J.); Department of Cardiovascular Medicine, Jichi Medical University, Tochigi and Graduate School of Medicine, The University of Tokyo, Japan (H.Z.)
| | - Toru Suzuki
- From the Department of Biochemistry and Molecular Biology, The Key Laboratory of Neural and Vascular Biology, China Administration of Education, Hebei Medical University, China (D.M., B.Z., R.Z., C.J., D.B., W.Y., Z.Y., X.Z., L.H., J.-k.W.); School of Public Health, North China University of Science and Technology, China (D.M., D.B.); Department of Cardiovascular Sciences, University of Leicester, UK (T.S.); Department of Thoracic Surgery, Tianjin Union Medicine Centre, China (C.J.); Department of Cardiovascular Medicine, Jichi Medical University, Tochigi and Graduate School of Medicine, The University of Tokyo, Japan (H.Z.)
| | - Ruonan Zhang
- From the Department of Biochemistry and Molecular Biology, The Key Laboratory of Neural and Vascular Biology, China Administration of Education, Hebei Medical University, China (D.M., B.Z., R.Z., C.J., D.B., W.Y., Z.Y., X.Z., L.H., J.-k.W.); School of Public Health, North China University of Science and Technology, China (D.M., D.B.); Department of Cardiovascular Sciences, University of Leicester, UK (T.S.); Department of Thoracic Surgery, Tianjin Union Medicine Centre, China (C.J.); Department of Cardiovascular Medicine, Jichi Medical University, Tochigi and Graduate School of Medicine, The University of Tokyo, Japan (H.Z.)
| | - Chunyang Jiang
- From the Department of Biochemistry and Molecular Biology, The Key Laboratory of Neural and Vascular Biology, China Administration of Education, Hebei Medical University, China (D.M., B.Z., R.Z., C.J., D.B., W.Y., Z.Y., X.Z., L.H., J.-k.W.); School of Public Health, North China University of Science and Technology, China (D.M., D.B.); Department of Cardiovascular Sciences, University of Leicester, UK (T.S.); Department of Thoracic Surgery, Tianjin Union Medicine Centre, China (C.J.); Department of Cardiovascular Medicine, Jichi Medical University, Tochigi and Graduate School of Medicine, The University of Tokyo, Japan (H.Z.)
| | - Disi Bai
- From the Department of Biochemistry and Molecular Biology, The Key Laboratory of Neural and Vascular Biology, China Administration of Education, Hebei Medical University, China (D.M., B.Z., R.Z., C.J., D.B., W.Y., Z.Y., X.Z., L.H., J.-k.W.); School of Public Health, North China University of Science and Technology, China (D.M., D.B.); Department of Cardiovascular Sciences, University of Leicester, UK (T.S.); Department of Thoracic Surgery, Tianjin Union Medicine Centre, China (C.J.); Department of Cardiovascular Medicine, Jichi Medical University, Tochigi and Graduate School of Medicine, The University of Tokyo, Japan (H.Z.)
| | - Weina Yin
- From the Department of Biochemistry and Molecular Biology, The Key Laboratory of Neural and Vascular Biology, China Administration of Education, Hebei Medical University, China (D.M., B.Z., R.Z., C.J., D.B., W.Y., Z.Y., X.Z., L.H., J.-k.W.); School of Public Health, North China University of Science and Technology, China (D.M., D.B.); Department of Cardiovascular Sciences, University of Leicester, UK (T.S.); Department of Thoracic Surgery, Tianjin Union Medicine Centre, China (C.J.); Department of Cardiovascular Medicine, Jichi Medical University, Tochigi and Graduate School of Medicine, The University of Tokyo, Japan (H.Z.)
| | - Zhan Yang
- From the Department of Biochemistry and Molecular Biology, The Key Laboratory of Neural and Vascular Biology, China Administration of Education, Hebei Medical University, China (D.M., B.Z., R.Z., C.J., D.B., W.Y., Z.Y., X.Z., L.H., J.-k.W.); School of Public Health, North China University of Science and Technology, China (D.M., D.B.); Department of Cardiovascular Sciences, University of Leicester, UK (T.S.); Department of Thoracic Surgery, Tianjin Union Medicine Centre, China (C.J.); Department of Cardiovascular Medicine, Jichi Medical University, Tochigi and Graduate School of Medicine, The University of Tokyo, Japan (H.Z.)
| | - Xinhua Zhang
- From the Department of Biochemistry and Molecular Biology, The Key Laboratory of Neural and Vascular Biology, China Administration of Education, Hebei Medical University, China (D.M., B.Z., R.Z., C.J., D.B., W.Y., Z.Y., X.Z., L.H., J.-k.W.); School of Public Health, North China University of Science and Technology, China (D.M., D.B.); Department of Cardiovascular Sciences, University of Leicester, UK (T.S.); Department of Thoracic Surgery, Tianjin Union Medicine Centre, China (C.J.); Department of Cardiovascular Medicine, Jichi Medical University, Tochigi and Graduate School of Medicine, The University of Tokyo, Japan (H.Z.)
| | - Lianguo Hou
- From the Department of Biochemistry and Molecular Biology, The Key Laboratory of Neural and Vascular Biology, China Administration of Education, Hebei Medical University, China (D.M., B.Z., R.Z., C.J., D.B., W.Y., Z.Y., X.Z., L.H., J.-k.W.); School of Public Health, North China University of Science and Technology, China (D.M., D.B.); Department of Cardiovascular Sciences, University of Leicester, UK (T.S.); Department of Thoracic Surgery, Tianjin Union Medicine Centre, China (C.J.); Department of Cardiovascular Medicine, Jichi Medical University, Tochigi and Graduate School of Medicine, The University of Tokyo, Japan (H.Z.)
| | - Hong Zhan
- From the Department of Biochemistry and Molecular Biology, The Key Laboratory of Neural and Vascular Biology, China Administration of Education, Hebei Medical University, China (D.M., B.Z., R.Z., C.J., D.B., W.Y., Z.Y., X.Z., L.H., J.-k.W.); School of Public Health, North China University of Science and Technology, China (D.M., D.B.); Department of Cardiovascular Sciences, University of Leicester, UK (T.S.); Department of Thoracic Surgery, Tianjin Union Medicine Centre, China (C.J.); Department of Cardiovascular Medicine, Jichi Medical University, Tochigi and Graduate School of Medicine, The University of Tokyo, Japan (H.Z.)
| | - Jin-Kun Wen
- From the Department of Biochemistry and Molecular Biology, The Key Laboratory of Neural and Vascular Biology, China Administration of Education, Hebei Medical University, China (D.M., B.Z., R.Z., C.J., D.B., W.Y., Z.Y., X.Z., L.H., J.-k.W.); School of Public Health, North China University of Science and Technology, China (D.M., D.B.); Department of Cardiovascular Sciences, University of Leicester, UK (T.S.); Department of Thoracic Surgery, Tianjin Union Medicine Centre, China (C.J.); Department of Cardiovascular Medicine, Jichi Medical University, Tochigi and Graduate School of Medicine, The University of Tokyo, Japan (H.Z.).
| |
Collapse
|
31
|
Yu H, Moran CS, Trollope AF, Woodward L, Kinobe R, Rush CM, Golledge J. Angiopoietin-2 attenuates angiotensin II-induced aortic aneurysm and atherosclerosis in apolipoprotein E-deficient mice. Sci Rep 2016; 6:35190. [PMID: 27767064 PMCID: PMC5073347 DOI: 10.1038/srep35190] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2016] [Accepted: 09/22/2016] [Indexed: 11/09/2022] Open
Abstract
Angiogenesis and inflammation are implicated in aortic aneurysm and atherosclerosis and regulated by angiopoietin-2 (Angpt2). The effect of Angpt2 administration on experimental aortic aneurysm and atherosclerosis was examined. Six-month-old male apolipoprotein E deficient (ApoE-/-) mice were infused with angiotensin II (AngII) and administered subcutaneous human Fc-protein (control) or recombinant Angpt2 (rAngpt2) over 14 days. Administration of rAngpt2 significantly inhibited AngII-induced aortic dilatation and rupture of the suprarenal aorta (SRA), and development of atherosclerosis within the aortic arch. These effects were blood pressure and plasma lipoprotein independent and associated with Tie2 activation and down-regulation of monocyte chemotactic protein-1 (MCP-1) within the SRA. Plasma concentrations of MCP-1 and interleukin-6 were significantly lower in mice receiving rAngpt2. Immunostaining for the monocyte/macrophage marker MOMA-2 and the angiogenesis marker CD31 within the SRA were less in mice receiving rAngpt2 than controls. The percentage of inflammatory (Ly6Chi) monocytes within the bone marrow was increased while that in peripheral blood was decreased by rAngpt2 administration. In conclusion, administration of rAngpt2 attenuated angiotensin II-induced aortic aneurysm and atherosclerosis in ApoE-/- mice associated with reduced aortic inflammation and angiogenesis. Up-regulation of Angpt2 may have potential therapeutic value in patients with aortic aneurysm and atherosclerosis.
Collapse
Affiliation(s)
- Hongyou Yu
- Queensland Research Centre for Peripheral Vascular Disease, College of Medicine and Dentistry, James Cook University, Townsville, 4811, Australia
| | - Corey S Moran
- Queensland Research Centre for Peripheral Vascular Disease, College of Medicine and Dentistry, James Cook University, Townsville, 4811, Australia
| | - Alexandra F Trollope
- Discipline of Anatomy, College of Medicine and Dentistry, James Cook University, Townsville, 4811, Australia
| | - Lynn Woodward
- Queensland Research Centre for Peripheral Vascular Disease, College of Medicine and Dentistry, James Cook University, Townsville, 4811, Australia
| | - Robert Kinobe
- Discipline of Biomedicine, College of Public Health, Medical and Veterinary Sciences, James Cook University, Townsville, 4811, Australia
| | - Catherine M Rush
- Discipline of Biomedicine, College of Public Health, Medical and Veterinary Sciences, James Cook University, Townsville, 4811, Australia
| | - Jonathan Golledge
- Queensland Research Centre for Peripheral Vascular Disease, College of Medicine and Dentistry, James Cook University, Townsville, 4811, Australia.,Department of Vascular and Endovascular Surgery, The Townsville Hospital, Townsville, 4814, Australia
| |
Collapse
|
32
|
Pope NH, Salmon M, Davis JP, Chatterjee A, Su G, Conte MS, Ailawadi G, Upchurch GR. D-series resolvins inhibit murine abdominal aortic aneurysm formation and increase M2 macrophage polarization. FASEB J 2016; 30:4192-4201. [PMID: 27619672 DOI: 10.1096/fj.201600144rr] [Citation(s) in RCA: 88] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2016] [Accepted: 09/01/2016] [Indexed: 12/31/2022]
Abstract
The role of resolvins in abdominal aortic aneurysm (AAA) has not been established. We hypothesized that treatment with D-series resolvins (RvD2 or RvD1) would attenuate murine AAA formation through alterations in macrophage polarization and cytokine expression. Male C57/B6 mice (n = 9 per group) 8 to 12 wk old received RvD2 (100 ng/kg/treatment), RvD1 (100 ng/kg/treatment), or vehicle only every third day beginning 3 d before abdominal aortic perfusion with elastase as prevention. Aortas were collected 14 d after elastase perfusion. Cytokine analysis (n = 5 per group) or confocal microscopy (n = 4 per group) was performed. In a separate experiment, RvD2 was provided to mice with small AAAs 3 d after elastase treatment (n = 8 per group). Additionally, apolipoprotein E knockout mice treated with angiotensin II (1000 ng/kg) were treated with RvD2 or vehicle alone (n = 10 per group) in a nonsurgical model of AAA. To determine the effect of RvD2 on macrophage polarization, confocal staining for macrophages, M1 and M2 macrophage subtypes, α-actin, and DAPI was performed. Mean aortic dilation was 96 ± 13% for vehicle-treated mice, 57 ± 9.7% for RvD2-treated mice, and 61 ± 11% for RvD1-treated mice (P < 0.0001). Proinflammatory cytokines macrophage chemotactic protein 1, C-X-C motif ligand 1, and IL-1β were significantly elevated in control animals compared to RvD2- and RvD1-treated animals (P < 0.05), resulting in a reduction of matrix metalloproteinase 2 and 9 activity in resolvin-treated mice in both elastase and angiotensin II models. Treatment of existing small AAAs with RvD2 demonstrated a 25% reduction in aneurysm size at d 14 compared to vehicle alone (P = 0.018). Confocal histology demonstrated a prevalence of M2 macrophages within the aortic medium in mice treated with RvD2. Resolvin D2 exhibits a potent protective effect against experimental AAA formation. Treatment with RvD2 significantly influences macrophage polarization and decreases several important proinflammatory cytokines. Resolvins and the alteration of macrophage polarization represent potential future targets for prevention of AAA.-Pope, N. H., Salmon, M., Davis, J. P., Chatterjee, A., Su, G., Conte, M. S., Ailawadi, G., Upchurch, G. R., Jr. D-series resolvins inhibit murine abdominal aortic aneurysm formation and increase M2 macrophage polarization.
Collapse
Affiliation(s)
- Nicolas H Pope
- Department of Surgery, University of Virginia, Charlottesville, Virginia, USA; and
| | - Morgan Salmon
- Department of Surgery, University of Virginia, Charlottesville, Virginia, USA; and
| | - John P Davis
- Department of Surgery, University of Virginia, Charlottesville, Virginia, USA; and
| | - Anuran Chatterjee
- Department of Surgery, Division of Vascular and Endovascular Surgery, University of California, San Francisco, San Francisco, California, USA
| | - Gang Su
- Department of Surgery, University of Virginia, Charlottesville, Virginia, USA; and
| | - Michael S Conte
- Department of Surgery, Division of Vascular and Endovascular Surgery, University of California, San Francisco, San Francisco, California, USA
| | - Gorav Ailawadi
- Department of Surgery, University of Virginia, Charlottesville, Virginia, USA; and
| | - Gilbert R Upchurch
- Department of Surgery, University of Virginia, Charlottesville, Virginia, USA; and
| |
Collapse
|
33
|
Chan JK, Glass GE, Ersek A, Freidin A, Williams GA, Gowers K, Espirito Santo AI, Jeffery R, Otto WR, Poulsom R, Feldmann M, Rankin SM, Horwood NJ, Nanchahal J. Low-dose TNF augments fracture healing in normal and osteoporotic bone by up-regulating the innate immune response. EMBO Mol Med 2016; 7:547-61. [PMID: 25770819 PMCID: PMC4492816 DOI: 10.15252/emmm.201404487] [Citation(s) in RCA: 84] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
The mechanism by which trauma initiates healing remains unclear. Precise understanding of these events may define interventions for accelerating healing that could be translated to the clinical arena. We previously reported that addition of low-dose recombinant human TNF (rhTNF) at the fracture site augmented fracture repair in a murine tibial fracture model. Here, we show that local rhTNF treatment is only effective when administered within 24 h of injury, when neutrophils are the major inflammatory cell infiltrate. Systemic administration of anti-TNF impaired fracture healing. Addition of rhTNF enhanced neutrophil recruitment and promoted recruitment of monocytes through CCL2 production. Conversely, depletion of neutrophils or inhibition of the chemokine receptor CCR2 resulted in significantly impaired fracture healing. Fragility, or osteoporotic, fractures represent a major medical problem as they are associated with permanent disability and premature death. Using a murine model of fragility fractures, we found that local rhTNF treatment improved fracture healing during the early phase of repair. If translated clinically, this promotion of fracture healing would reduce the morbidity and mortality associated with delayed patient mobilization.
Collapse
Affiliation(s)
- James K Chan
- Kennedy Institute of Rheumatology, University of Oxford, Oxford, UK
| | - Graeme E Glass
- Kennedy Institute of Rheumatology, University of Oxford, Oxford, UK
| | - Adel Ersek
- Kennedy Institute of Rheumatology, University of Oxford, Oxford, UK
| | - Andrew Freidin
- Kennedy Institute of Rheumatology, University of Oxford, Oxford, UK
| | - Garry A Williams
- Kennedy Institute of Rheumatology, University of Oxford, Oxford, UK
| | - Kate Gowers
- National Heart and Lung Institute, Imperial College London, London, UK
| | | | - Rosemary Jeffery
- Histopathology Laboratory and In Situ Hybridisation Service, Cancer Research UK - London Research Institute, London, UK
| | - William R Otto
- Histopathology Laboratory and In Situ Hybridisation Service, Cancer Research UK - London Research Institute, London, UK
| | - Richard Poulsom
- Histopathology Laboratory and In Situ Hybridisation Service, Cancer Research UK - London Research Institute, London, UK
| | - Marc Feldmann
- Kennedy Institute of Rheumatology, University of Oxford, Oxford, UK
| | - Sara M Rankin
- National Heart and Lung Institute, Imperial College London, London, UK
| | - Nicole J Horwood
- Kennedy Institute of Rheumatology, University of Oxford, Oxford, UK
| | | |
Collapse
|
34
|
Zhang P, Hou S, Chen J, Zhang J, Lin F, Ju R, Cheng X, Ma X, Song Y, Zhang Y, Zhu M, Du J, Lan Y, Yang X. Smad4 Deficiency in Smooth Muscle Cells Initiates the Formation of Aortic Aneurysm. Circ Res 2015; 118:388-99. [PMID: 26699655 DOI: 10.1161/circresaha.115.308040] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/19/2015] [Accepted: 12/21/2015] [Indexed: 12/12/2022]
Abstract
RATIONALE Aortic aneurysm is a life-threatening cardiovascular disorder caused by the predisposition for dissection and rupture. Genetic studies have proved the involvement of the transforming growth factor-β (TGF-β) pathway in aortic aneurysm. Smad4 is the central mediator of the canonical TGF-β signaling pathway. However, the exact role of Smad4 in smooth muscle cells (SMCs) leading to the pathogenesis of aortic aneurysms is largely unknown. OBJECTIVE To determine the role of smooth muscle Smad4 in the pathogenesis of aortic aneurysms. METHODS AND RESULTS Conditional gene knockout strategy combined with histology and expression analysis showed that Smad4 or TGF-β receptor type II deficiency in SMCs led to the occurrence of aortic aneurysms along with an upregulation of cathepsin S and matrix metallopeptidase-12, which are proteases essential for elastin degradation. We further demonstrated a previously unknown downregulation of matrix metallopeptidase-12 by TGF-β in the aortic SMCs, which is largely abrogated in the absence of Smad4. Chemotactic assay and pharmacologic treatment demonstrated that Smad4-deficient SMCs directly triggered aortic wall inflammation via the excessive production of chemokines to recruit macrophages. Monocyte/macrophage depletion or blocking selective chemokine axis largely abrogated the progression of aortic aneurysm caused by Smad4 deficiency in SMCs. CONCLUSIONS The findings reveal that Smad4-dependent TGF-β signaling in SMCs protects against aortic aneurysm formation and dissection. The data also suggest important implications for novel therapeutic strategies to limit the progression of the aneurysm resulting from TGF-β signaling loss-of-function mutations.
Collapse
Affiliation(s)
- Peng Zhang
- From the State Key Laboratory of Proteomics, Collaborative Innovation Center for Cardiovascular Disorders, Genetic Laboratory of Development and Diseases, Institute of Biotechnology, Beijing, PR China (P.Z., S.H., J.C., J.Z., F.L., R.J., X.C., Y.L., X.Y.); Model Organism Division, E-institutes of Shanghai Universities, Shanghai Jiaotong University, Shanghai, PR China (P.Z., J.C., X.Y.); Institute of Vascular Medicine, Peking University Third Hospital and Key Laboratory of Molecular Cardiovascular Sciences, Ministry of Education, Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Ministry of Health, Beijing, PR China (X.M., Y.S., Y.Z.); Model Animal Research Center and MOE Key Laboratory of Model Animal for Disease Study and School of Medicine, Nanjing University, Nanjing, PR China (M.Z.); and Beijing AnZhen Hospital, Affiliated to Capital Medical University, Beijing Institute of Heart, Lung and Blood Vessel Diseases, The Key Laboratory of Remodeling-related Cardiovascular Diseases, Ministry of Education, Beijing, PR China (J.D.)
| | - Siyuan Hou
- From the State Key Laboratory of Proteomics, Collaborative Innovation Center for Cardiovascular Disorders, Genetic Laboratory of Development and Diseases, Institute of Biotechnology, Beijing, PR China (P.Z., S.H., J.C., J.Z., F.L., R.J., X.C., Y.L., X.Y.); Model Organism Division, E-institutes of Shanghai Universities, Shanghai Jiaotong University, Shanghai, PR China (P.Z., J.C., X.Y.); Institute of Vascular Medicine, Peking University Third Hospital and Key Laboratory of Molecular Cardiovascular Sciences, Ministry of Education, Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Ministry of Health, Beijing, PR China (X.M., Y.S., Y.Z.); Model Animal Research Center and MOE Key Laboratory of Model Animal for Disease Study and School of Medicine, Nanjing University, Nanjing, PR China (M.Z.); and Beijing AnZhen Hospital, Affiliated to Capital Medical University, Beijing Institute of Heart, Lung and Blood Vessel Diseases, The Key Laboratory of Remodeling-related Cardiovascular Diseases, Ministry of Education, Beijing, PR China (J.D.)
| | - Jicheng Chen
- From the State Key Laboratory of Proteomics, Collaborative Innovation Center for Cardiovascular Disorders, Genetic Laboratory of Development and Diseases, Institute of Biotechnology, Beijing, PR China (P.Z., S.H., J.C., J.Z., F.L., R.J., X.C., Y.L., X.Y.); Model Organism Division, E-institutes of Shanghai Universities, Shanghai Jiaotong University, Shanghai, PR China (P.Z., J.C., X.Y.); Institute of Vascular Medicine, Peking University Third Hospital and Key Laboratory of Molecular Cardiovascular Sciences, Ministry of Education, Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Ministry of Health, Beijing, PR China (X.M., Y.S., Y.Z.); Model Animal Research Center and MOE Key Laboratory of Model Animal for Disease Study and School of Medicine, Nanjing University, Nanjing, PR China (M.Z.); and Beijing AnZhen Hospital, Affiliated to Capital Medical University, Beijing Institute of Heart, Lung and Blood Vessel Diseases, The Key Laboratory of Remodeling-related Cardiovascular Diseases, Ministry of Education, Beijing, PR China (J.D.)
| | - Jishuai Zhang
- From the State Key Laboratory of Proteomics, Collaborative Innovation Center for Cardiovascular Disorders, Genetic Laboratory of Development and Diseases, Institute of Biotechnology, Beijing, PR China (P.Z., S.H., J.C., J.Z., F.L., R.J., X.C., Y.L., X.Y.); Model Organism Division, E-institutes of Shanghai Universities, Shanghai Jiaotong University, Shanghai, PR China (P.Z., J.C., X.Y.); Institute of Vascular Medicine, Peking University Third Hospital and Key Laboratory of Molecular Cardiovascular Sciences, Ministry of Education, Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Ministry of Health, Beijing, PR China (X.M., Y.S., Y.Z.); Model Animal Research Center and MOE Key Laboratory of Model Animal for Disease Study and School of Medicine, Nanjing University, Nanjing, PR China (M.Z.); and Beijing AnZhen Hospital, Affiliated to Capital Medical University, Beijing Institute of Heart, Lung and Blood Vessel Diseases, The Key Laboratory of Remodeling-related Cardiovascular Diseases, Ministry of Education, Beijing, PR China (J.D.)
| | - Fuyu Lin
- From the State Key Laboratory of Proteomics, Collaborative Innovation Center for Cardiovascular Disorders, Genetic Laboratory of Development and Diseases, Institute of Biotechnology, Beijing, PR China (P.Z., S.H., J.C., J.Z., F.L., R.J., X.C., Y.L., X.Y.); Model Organism Division, E-institutes of Shanghai Universities, Shanghai Jiaotong University, Shanghai, PR China (P.Z., J.C., X.Y.); Institute of Vascular Medicine, Peking University Third Hospital and Key Laboratory of Molecular Cardiovascular Sciences, Ministry of Education, Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Ministry of Health, Beijing, PR China (X.M., Y.S., Y.Z.); Model Animal Research Center and MOE Key Laboratory of Model Animal for Disease Study and School of Medicine, Nanjing University, Nanjing, PR China (M.Z.); and Beijing AnZhen Hospital, Affiliated to Capital Medical University, Beijing Institute of Heart, Lung and Blood Vessel Diseases, The Key Laboratory of Remodeling-related Cardiovascular Diseases, Ministry of Education, Beijing, PR China (J.D.)
| | - Renjie Ju
- From the State Key Laboratory of Proteomics, Collaborative Innovation Center for Cardiovascular Disorders, Genetic Laboratory of Development and Diseases, Institute of Biotechnology, Beijing, PR China (P.Z., S.H., J.C., J.Z., F.L., R.J., X.C., Y.L., X.Y.); Model Organism Division, E-institutes of Shanghai Universities, Shanghai Jiaotong University, Shanghai, PR China (P.Z., J.C., X.Y.); Institute of Vascular Medicine, Peking University Third Hospital and Key Laboratory of Molecular Cardiovascular Sciences, Ministry of Education, Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Ministry of Health, Beijing, PR China (X.M., Y.S., Y.Z.); Model Animal Research Center and MOE Key Laboratory of Model Animal for Disease Study and School of Medicine, Nanjing University, Nanjing, PR China (M.Z.); and Beijing AnZhen Hospital, Affiliated to Capital Medical University, Beijing Institute of Heart, Lung and Blood Vessel Diseases, The Key Laboratory of Remodeling-related Cardiovascular Diseases, Ministry of Education, Beijing, PR China (J.D.)
| | - Xuan Cheng
- From the State Key Laboratory of Proteomics, Collaborative Innovation Center for Cardiovascular Disorders, Genetic Laboratory of Development and Diseases, Institute of Biotechnology, Beijing, PR China (P.Z., S.H., J.C., J.Z., F.L., R.J., X.C., Y.L., X.Y.); Model Organism Division, E-institutes of Shanghai Universities, Shanghai Jiaotong University, Shanghai, PR China (P.Z., J.C., X.Y.); Institute of Vascular Medicine, Peking University Third Hospital and Key Laboratory of Molecular Cardiovascular Sciences, Ministry of Education, Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Ministry of Health, Beijing, PR China (X.M., Y.S., Y.Z.); Model Animal Research Center and MOE Key Laboratory of Model Animal for Disease Study and School of Medicine, Nanjing University, Nanjing, PR China (M.Z.); and Beijing AnZhen Hospital, Affiliated to Capital Medical University, Beijing Institute of Heart, Lung and Blood Vessel Diseases, The Key Laboratory of Remodeling-related Cardiovascular Diseases, Ministry of Education, Beijing, PR China (J.D.)
| | - Xiaowei Ma
- From the State Key Laboratory of Proteomics, Collaborative Innovation Center for Cardiovascular Disorders, Genetic Laboratory of Development and Diseases, Institute of Biotechnology, Beijing, PR China (P.Z., S.H., J.C., J.Z., F.L., R.J., X.C., Y.L., X.Y.); Model Organism Division, E-institutes of Shanghai Universities, Shanghai Jiaotong University, Shanghai, PR China (P.Z., J.C., X.Y.); Institute of Vascular Medicine, Peking University Third Hospital and Key Laboratory of Molecular Cardiovascular Sciences, Ministry of Education, Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Ministry of Health, Beijing, PR China (X.M., Y.S., Y.Z.); Model Animal Research Center and MOE Key Laboratory of Model Animal for Disease Study and School of Medicine, Nanjing University, Nanjing, PR China (M.Z.); and Beijing AnZhen Hospital, Affiliated to Capital Medical University, Beijing Institute of Heart, Lung and Blood Vessel Diseases, The Key Laboratory of Remodeling-related Cardiovascular Diseases, Ministry of Education, Beijing, PR China (J.D.)
| | - Yao Song
- From the State Key Laboratory of Proteomics, Collaborative Innovation Center for Cardiovascular Disorders, Genetic Laboratory of Development and Diseases, Institute of Biotechnology, Beijing, PR China (P.Z., S.H., J.C., J.Z., F.L., R.J., X.C., Y.L., X.Y.); Model Organism Division, E-institutes of Shanghai Universities, Shanghai Jiaotong University, Shanghai, PR China (P.Z., J.C., X.Y.); Institute of Vascular Medicine, Peking University Third Hospital and Key Laboratory of Molecular Cardiovascular Sciences, Ministry of Education, Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Ministry of Health, Beijing, PR China (X.M., Y.S., Y.Z.); Model Animal Research Center and MOE Key Laboratory of Model Animal for Disease Study and School of Medicine, Nanjing University, Nanjing, PR China (M.Z.); and Beijing AnZhen Hospital, Affiliated to Capital Medical University, Beijing Institute of Heart, Lung and Blood Vessel Diseases, The Key Laboratory of Remodeling-related Cardiovascular Diseases, Ministry of Education, Beijing, PR China (J.D.)
| | - Youyi Zhang
- From the State Key Laboratory of Proteomics, Collaborative Innovation Center for Cardiovascular Disorders, Genetic Laboratory of Development and Diseases, Institute of Biotechnology, Beijing, PR China (P.Z., S.H., J.C., J.Z., F.L., R.J., X.C., Y.L., X.Y.); Model Organism Division, E-institutes of Shanghai Universities, Shanghai Jiaotong University, Shanghai, PR China (P.Z., J.C., X.Y.); Institute of Vascular Medicine, Peking University Third Hospital and Key Laboratory of Molecular Cardiovascular Sciences, Ministry of Education, Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Ministry of Health, Beijing, PR China (X.M., Y.S., Y.Z.); Model Animal Research Center and MOE Key Laboratory of Model Animal for Disease Study and School of Medicine, Nanjing University, Nanjing, PR China (M.Z.); and Beijing AnZhen Hospital, Affiliated to Capital Medical University, Beijing Institute of Heart, Lung and Blood Vessel Diseases, The Key Laboratory of Remodeling-related Cardiovascular Diseases, Ministry of Education, Beijing, PR China (J.D.)
| | - Minsheng Zhu
- From the State Key Laboratory of Proteomics, Collaborative Innovation Center for Cardiovascular Disorders, Genetic Laboratory of Development and Diseases, Institute of Biotechnology, Beijing, PR China (P.Z., S.H., J.C., J.Z., F.L., R.J., X.C., Y.L., X.Y.); Model Organism Division, E-institutes of Shanghai Universities, Shanghai Jiaotong University, Shanghai, PR China (P.Z., J.C., X.Y.); Institute of Vascular Medicine, Peking University Third Hospital and Key Laboratory of Molecular Cardiovascular Sciences, Ministry of Education, Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Ministry of Health, Beijing, PR China (X.M., Y.S., Y.Z.); Model Animal Research Center and MOE Key Laboratory of Model Animal for Disease Study and School of Medicine, Nanjing University, Nanjing, PR China (M.Z.); and Beijing AnZhen Hospital, Affiliated to Capital Medical University, Beijing Institute of Heart, Lung and Blood Vessel Diseases, The Key Laboratory of Remodeling-related Cardiovascular Diseases, Ministry of Education, Beijing, PR China (J.D.)
| | - Jie Du
- From the State Key Laboratory of Proteomics, Collaborative Innovation Center for Cardiovascular Disorders, Genetic Laboratory of Development and Diseases, Institute of Biotechnology, Beijing, PR China (P.Z., S.H., J.C., J.Z., F.L., R.J., X.C., Y.L., X.Y.); Model Organism Division, E-institutes of Shanghai Universities, Shanghai Jiaotong University, Shanghai, PR China (P.Z., J.C., X.Y.); Institute of Vascular Medicine, Peking University Third Hospital and Key Laboratory of Molecular Cardiovascular Sciences, Ministry of Education, Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Ministry of Health, Beijing, PR China (X.M., Y.S., Y.Z.); Model Animal Research Center and MOE Key Laboratory of Model Animal for Disease Study and School of Medicine, Nanjing University, Nanjing, PR China (M.Z.); and Beijing AnZhen Hospital, Affiliated to Capital Medical University, Beijing Institute of Heart, Lung and Blood Vessel Diseases, The Key Laboratory of Remodeling-related Cardiovascular Diseases, Ministry of Education, Beijing, PR China (J.D.)
| | - Yu Lan
- From the State Key Laboratory of Proteomics, Collaborative Innovation Center for Cardiovascular Disorders, Genetic Laboratory of Development and Diseases, Institute of Biotechnology, Beijing, PR China (P.Z., S.H., J.C., J.Z., F.L., R.J., X.C., Y.L., X.Y.); Model Organism Division, E-institutes of Shanghai Universities, Shanghai Jiaotong University, Shanghai, PR China (P.Z., J.C., X.Y.); Institute of Vascular Medicine, Peking University Third Hospital and Key Laboratory of Molecular Cardiovascular Sciences, Ministry of Education, Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Ministry of Health, Beijing, PR China (X.M., Y.S., Y.Z.); Model Animal Research Center and MOE Key Laboratory of Model Animal for Disease Study and School of Medicine, Nanjing University, Nanjing, PR China (M.Z.); and Beijing AnZhen Hospital, Affiliated to Capital Medical University, Beijing Institute of Heart, Lung and Blood Vessel Diseases, The Key Laboratory of Remodeling-related Cardiovascular Diseases, Ministry of Education, Beijing, PR China (J.D.).
| | - Xiao Yang
- From the State Key Laboratory of Proteomics, Collaborative Innovation Center for Cardiovascular Disorders, Genetic Laboratory of Development and Diseases, Institute of Biotechnology, Beijing, PR China (P.Z., S.H., J.C., J.Z., F.L., R.J., X.C., Y.L., X.Y.); Model Organism Division, E-institutes of Shanghai Universities, Shanghai Jiaotong University, Shanghai, PR China (P.Z., J.C., X.Y.); Institute of Vascular Medicine, Peking University Third Hospital and Key Laboratory of Molecular Cardiovascular Sciences, Ministry of Education, Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Ministry of Health, Beijing, PR China (X.M., Y.S., Y.Z.); Model Animal Research Center and MOE Key Laboratory of Model Animal for Disease Study and School of Medicine, Nanjing University, Nanjing, PR China (M.Z.); and Beijing AnZhen Hospital, Affiliated to Capital Medical University, Beijing Institute of Heart, Lung and Blood Vessel Diseases, The Key Laboratory of Remodeling-related Cardiovascular Diseases, Ministry of Education, Beijing, PR China (J.D.).
| |
Collapse
|
35
|
Ren J, Liu Z, Wang Q, Giles J, Greenberg J, Sheibani N, Kent KC, Liu B. Andrographolide Ameliorates Abdominal Aortic Aneurysm Progression by Inhibiting Inflammatory Cell Infiltration through Downregulation of Cytokine and Integrin Expression. J Pharmacol Exp Ther 2015; 356:137-47. [PMID: 26483397 DOI: 10.1124/jpet.115.227934] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2015] [Accepted: 10/09/2015] [Indexed: 12/14/2022] Open
Abstract
Abdominal aortic aneurysm (AAA), characterized by exuberant inflammation and tissue deterioration, is a common aortic disease associated with a high mortality rate. There is currently no established pharmacological therapy to treat this progressive disease. Andrographolide (Andro), a major bioactive component of the herbaceous plant Andrographis paniculata, has been found to exhibit potent anti-inflammatory properties by inhibiting nuclear factor κ-light-chain-enhancer of activated B cells (NF-κB) activity in several disease models. In this study, we investigated the ability of Andro to suppress inflammation associated with aneurysms, and whether it may be used to block the progression of AAA. Whereas diseased aortae continued to expand in the solvent-treated group, daily administration of Andro to mice with small aneurysms significantly attenuated aneurysm growth, as measured by the diminished expansion of aortic diameter (165.68 ± 15.85% vs. 90.62 ± 22.91%, P < 0.05). Immunohistochemistry analyses revealed that Andro decreased infiltration of monocytes/macrophages and T cells. Mechanistically, Andro inhibited arterial NF-κB activation and reduced the production of proinflammatory cytokines [CCL2, CXCL10, tumor necrosis factor α, and interferon-γ] in the treated aortae. Furthermore, Andro suppressed α4 integrin expression and attenuated the ability of monocytes/macrophages to adhere to activated endothelial cells. These results indicate that Andro suppresses progression of AAA, likely through inhibition of inflammatory cell infiltration via downregulation of NF-κB-mediated cytokine production and α4 integrin expression. Thus, Andro may offer a pharmacological therapy to slow disease progression in patients with small aneurysms.
Collapse
Affiliation(s)
- Jun Ren
- Division of Vascular Surgery, Department of Surgery (J.R., Z.L., Q.W., J.Gi., J.Gr., K.C.K., B.L.) and Department of Ophthalmology and Visual Sciences (N.S.), University of Wisconsin, Madison, Wisconsin; And Department of Vascular Surgery, 2nd Affiliated Hospital School of Medicine, Zhejiang University, Zhejiang, China (Z.L.)
| | - Zhenjie Liu
- Division of Vascular Surgery, Department of Surgery (J.R., Z.L., Q.W., J.Gi., J.Gr., K.C.K., B.L.) and Department of Ophthalmology and Visual Sciences (N.S.), University of Wisconsin, Madison, Wisconsin; And Department of Vascular Surgery, 2nd Affiliated Hospital School of Medicine, Zhejiang University, Zhejiang, China (Z.L.)
| | - Qiwei Wang
- Division of Vascular Surgery, Department of Surgery (J.R., Z.L., Q.W., J.Gi., J.Gr., K.C.K., B.L.) and Department of Ophthalmology and Visual Sciences (N.S.), University of Wisconsin, Madison, Wisconsin; And Department of Vascular Surgery, 2nd Affiliated Hospital School of Medicine, Zhejiang University, Zhejiang, China (Z.L.)
| | - Jasmine Giles
- Division of Vascular Surgery, Department of Surgery (J.R., Z.L., Q.W., J.Gi., J.Gr., K.C.K., B.L.) and Department of Ophthalmology and Visual Sciences (N.S.), University of Wisconsin, Madison, Wisconsin; And Department of Vascular Surgery, 2nd Affiliated Hospital School of Medicine, Zhejiang University, Zhejiang, China (Z.L.)
| | - Jason Greenberg
- Division of Vascular Surgery, Department of Surgery (J.R., Z.L., Q.W., J.Gi., J.Gr., K.C.K., B.L.) and Department of Ophthalmology and Visual Sciences (N.S.), University of Wisconsin, Madison, Wisconsin; And Department of Vascular Surgery, 2nd Affiliated Hospital School of Medicine, Zhejiang University, Zhejiang, China (Z.L.)
| | - Nader Sheibani
- Division of Vascular Surgery, Department of Surgery (J.R., Z.L., Q.W., J.Gi., J.Gr., K.C.K., B.L.) and Department of Ophthalmology and Visual Sciences (N.S.), University of Wisconsin, Madison, Wisconsin; And Department of Vascular Surgery, 2nd Affiliated Hospital School of Medicine, Zhejiang University, Zhejiang, China (Z.L.)
| | - K Craig Kent
- Division of Vascular Surgery, Department of Surgery (J.R., Z.L., Q.W., J.Gi., J.Gr., K.C.K., B.L.) and Department of Ophthalmology and Visual Sciences (N.S.), University of Wisconsin, Madison, Wisconsin; And Department of Vascular Surgery, 2nd Affiliated Hospital School of Medicine, Zhejiang University, Zhejiang, China (Z.L.)
| | - Bo Liu
- Division of Vascular Surgery, Department of Surgery (J.R., Z.L., Q.W., J.Gi., J.Gr., K.C.K., B.L.) and Department of Ophthalmology and Visual Sciences (N.S.), University of Wisconsin, Madison, Wisconsin; And Department of Vascular Surgery, 2nd Affiliated Hospital School of Medicine, Zhejiang University, Zhejiang, China (Z.L.)
| |
Collapse
|
36
|
Raaz U, Zöllner AM, Schellinger IN, Toh R, Nakagami F, Brandt M, Emrich FC, Kayama Y, Eken S, Adam M, Maegdefessel L, Hertel T, Deng A, Jagger A, Buerke M, Dalman RL, Spin JM, Kuhl E, Tsao PS. Segmental aortic stiffening contributes to experimental abdominal aortic aneurysm development. Circulation 2015; 131:1783-95. [PMID: 25904646 DOI: 10.1161/circulationaha.114.012377] [Citation(s) in RCA: 98] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/03/2014] [Accepted: 03/05/2015] [Indexed: 01/19/2023]
Abstract
BACKGROUND Stiffening of the aortic wall is a phenomenon consistently observed in age and in abdominal aortic aneurysm (AAA). However, its role in AAA pathophysiology is largely undefined. METHODS AND RESULTS Using an established murine elastase-induced AAA model, we demonstrate that segmental aortic stiffening precedes aneurysm growth. Finite-element analysis reveals that early stiffening of the aneurysm-prone aortic segment leads to axial (longitudinal) wall stress generated by cyclic (systolic) tethering of adjacent, more compliant wall segments. Interventional stiffening of AAA-adjacent aortic segments (via external application of surgical adhesive) significantly reduces aneurysm growth. These changes correlate with the reduced segmental stiffness of the AAA-prone aorta (attributable to equalized stiffness in adjacent segments), reduced axial wall stress, decreased production of reactive oxygen species, attenuated elastin breakdown, and decreased expression of inflammatory cytokines and macrophage infiltration, and attenuated apoptosis within the aortic wall, as well. Cyclic pressurization of segmentally stiffened aortic segments ex vivo increases the expression of genes related to inflammation and extracellular matrix remodeling. Finally, human ultrasound studies reveal that aging, a significant AAA risk factor, is accompanied by segmental infrarenal aortic stiffening. CONCLUSIONS The present study introduces the novel concept of segmental aortic stiffening as an early pathomechanism generating aortic wall stress and triggering aneurysmal growth, thereby delineating potential underlying molecular mechanisms and therapeutic targets. In addition, monitoring segmental aortic stiffening may aid the identification of patients at risk for AAA.
Collapse
Affiliation(s)
- Uwe Raaz
- From Division of Cardiovascular Medicine, Stanford University School of Medicine, CA (U.R., I.N.S., R.T., F.N., Y.K., M.A., A.D., A.J., J.MS., P.S.T.); Cardiovascular Institute, Stanford University School of Medicine, CA (U.R., A.M.Z., F.N., M.B., F.C.E., Y.K., M.A., R.L.D., J.M.S., P.S.T.); VA Palo Alto Health Care System, CA (U.R., I.N.S., Y.K., M.A., A.D., A.J., J.M.S., P.S.T.); Department of Mechanical Engineering, Stanford University School of Medicine, CA (A.M.Z., E.K.); Department of Cardiothoracic Surgery, Stanford University School of Medicine, CA (F.C.E., E.K.); Department of Medicine, Karolinska Institute, Stockholm, Sweden (S.E., L.M.); Center for Vascular Medicine, Zwickau, Germany (T.H.); Division of Cardiovascular Medicine and Intensive Care Medicine, Saint Mary's Hospital, Siegen, Germany (M.B.); Division of Vascular Surgery, Stanford University School of Medicine, CA (R.L.D.); and Department of Bioengineering, Stanford University School of Medicine, CA (E.K.)
| | - Alexander M Zöllner
- From Division of Cardiovascular Medicine, Stanford University School of Medicine, CA (U.R., I.N.S., R.T., F.N., Y.K., M.A., A.D., A.J., J.MS., P.S.T.); Cardiovascular Institute, Stanford University School of Medicine, CA (U.R., A.M.Z., F.N., M.B., F.C.E., Y.K., M.A., R.L.D., J.M.S., P.S.T.); VA Palo Alto Health Care System, CA (U.R., I.N.S., Y.K., M.A., A.D., A.J., J.M.S., P.S.T.); Department of Mechanical Engineering, Stanford University School of Medicine, CA (A.M.Z., E.K.); Department of Cardiothoracic Surgery, Stanford University School of Medicine, CA (F.C.E., E.K.); Department of Medicine, Karolinska Institute, Stockholm, Sweden (S.E., L.M.); Center for Vascular Medicine, Zwickau, Germany (T.H.); Division of Cardiovascular Medicine and Intensive Care Medicine, Saint Mary's Hospital, Siegen, Germany (M.B.); Division of Vascular Surgery, Stanford University School of Medicine, CA (R.L.D.); and Department of Bioengineering, Stanford University School of Medicine, CA (E.K.)
| | - Isabel N Schellinger
- From Division of Cardiovascular Medicine, Stanford University School of Medicine, CA (U.R., I.N.S., R.T., F.N., Y.K., M.A., A.D., A.J., J.MS., P.S.T.); Cardiovascular Institute, Stanford University School of Medicine, CA (U.R., A.M.Z., F.N., M.B., F.C.E., Y.K., M.A., R.L.D., J.M.S., P.S.T.); VA Palo Alto Health Care System, CA (U.R., I.N.S., Y.K., M.A., A.D., A.J., J.M.S., P.S.T.); Department of Mechanical Engineering, Stanford University School of Medicine, CA (A.M.Z., E.K.); Department of Cardiothoracic Surgery, Stanford University School of Medicine, CA (F.C.E., E.K.); Department of Medicine, Karolinska Institute, Stockholm, Sweden (S.E., L.M.); Center for Vascular Medicine, Zwickau, Germany (T.H.); Division of Cardiovascular Medicine and Intensive Care Medicine, Saint Mary's Hospital, Siegen, Germany (M.B.); Division of Vascular Surgery, Stanford University School of Medicine, CA (R.L.D.); and Department of Bioengineering, Stanford University School of Medicine, CA (E.K.)
| | - Ryuji Toh
- From Division of Cardiovascular Medicine, Stanford University School of Medicine, CA (U.R., I.N.S., R.T., F.N., Y.K., M.A., A.D., A.J., J.MS., P.S.T.); Cardiovascular Institute, Stanford University School of Medicine, CA (U.R., A.M.Z., F.N., M.B., F.C.E., Y.K., M.A., R.L.D., J.M.S., P.S.T.); VA Palo Alto Health Care System, CA (U.R., I.N.S., Y.K., M.A., A.D., A.J., J.M.S., P.S.T.); Department of Mechanical Engineering, Stanford University School of Medicine, CA (A.M.Z., E.K.); Department of Cardiothoracic Surgery, Stanford University School of Medicine, CA (F.C.E., E.K.); Department of Medicine, Karolinska Institute, Stockholm, Sweden (S.E., L.M.); Center for Vascular Medicine, Zwickau, Germany (T.H.); Division of Cardiovascular Medicine and Intensive Care Medicine, Saint Mary's Hospital, Siegen, Germany (M.B.); Division of Vascular Surgery, Stanford University School of Medicine, CA (R.L.D.); and Department of Bioengineering, Stanford University School of Medicine, CA (E.K.)
| | - Futoshi Nakagami
- From Division of Cardiovascular Medicine, Stanford University School of Medicine, CA (U.R., I.N.S., R.T., F.N., Y.K., M.A., A.D., A.J., J.MS., P.S.T.); Cardiovascular Institute, Stanford University School of Medicine, CA (U.R., A.M.Z., F.N., M.B., F.C.E., Y.K., M.A., R.L.D., J.M.S., P.S.T.); VA Palo Alto Health Care System, CA (U.R., I.N.S., Y.K., M.A., A.D., A.J., J.M.S., P.S.T.); Department of Mechanical Engineering, Stanford University School of Medicine, CA (A.M.Z., E.K.); Department of Cardiothoracic Surgery, Stanford University School of Medicine, CA (F.C.E., E.K.); Department of Medicine, Karolinska Institute, Stockholm, Sweden (S.E., L.M.); Center for Vascular Medicine, Zwickau, Germany (T.H.); Division of Cardiovascular Medicine and Intensive Care Medicine, Saint Mary's Hospital, Siegen, Germany (M.B.); Division of Vascular Surgery, Stanford University School of Medicine, CA (R.L.D.); and Department of Bioengineering, Stanford University School of Medicine, CA (E.K.)
| | - Moritz Brandt
- From Division of Cardiovascular Medicine, Stanford University School of Medicine, CA (U.R., I.N.S., R.T., F.N., Y.K., M.A., A.D., A.J., J.MS., P.S.T.); Cardiovascular Institute, Stanford University School of Medicine, CA (U.R., A.M.Z., F.N., M.B., F.C.E., Y.K., M.A., R.L.D., J.M.S., P.S.T.); VA Palo Alto Health Care System, CA (U.R., I.N.S., Y.K., M.A., A.D., A.J., J.M.S., P.S.T.); Department of Mechanical Engineering, Stanford University School of Medicine, CA (A.M.Z., E.K.); Department of Cardiothoracic Surgery, Stanford University School of Medicine, CA (F.C.E., E.K.); Department of Medicine, Karolinska Institute, Stockholm, Sweden (S.E., L.M.); Center for Vascular Medicine, Zwickau, Germany (T.H.); Division of Cardiovascular Medicine and Intensive Care Medicine, Saint Mary's Hospital, Siegen, Germany (M.B.); Division of Vascular Surgery, Stanford University School of Medicine, CA (R.L.D.); and Department of Bioengineering, Stanford University School of Medicine, CA (E.K.)
| | - Fabian C Emrich
- From Division of Cardiovascular Medicine, Stanford University School of Medicine, CA (U.R., I.N.S., R.T., F.N., Y.K., M.A., A.D., A.J., J.MS., P.S.T.); Cardiovascular Institute, Stanford University School of Medicine, CA (U.R., A.M.Z., F.N., M.B., F.C.E., Y.K., M.A., R.L.D., J.M.S., P.S.T.); VA Palo Alto Health Care System, CA (U.R., I.N.S., Y.K., M.A., A.D., A.J., J.M.S., P.S.T.); Department of Mechanical Engineering, Stanford University School of Medicine, CA (A.M.Z., E.K.); Department of Cardiothoracic Surgery, Stanford University School of Medicine, CA (F.C.E., E.K.); Department of Medicine, Karolinska Institute, Stockholm, Sweden (S.E., L.M.); Center for Vascular Medicine, Zwickau, Germany (T.H.); Division of Cardiovascular Medicine and Intensive Care Medicine, Saint Mary's Hospital, Siegen, Germany (M.B.); Division of Vascular Surgery, Stanford University School of Medicine, CA (R.L.D.); and Department of Bioengineering, Stanford University School of Medicine, CA (E.K.)
| | - Yosuke Kayama
- From Division of Cardiovascular Medicine, Stanford University School of Medicine, CA (U.R., I.N.S., R.T., F.N., Y.K., M.A., A.D., A.J., J.MS., P.S.T.); Cardiovascular Institute, Stanford University School of Medicine, CA (U.R., A.M.Z., F.N., M.B., F.C.E., Y.K., M.A., R.L.D., J.M.S., P.S.T.); VA Palo Alto Health Care System, CA (U.R., I.N.S., Y.K., M.A., A.D., A.J., J.M.S., P.S.T.); Department of Mechanical Engineering, Stanford University School of Medicine, CA (A.M.Z., E.K.); Department of Cardiothoracic Surgery, Stanford University School of Medicine, CA (F.C.E., E.K.); Department of Medicine, Karolinska Institute, Stockholm, Sweden (S.E., L.M.); Center for Vascular Medicine, Zwickau, Germany (T.H.); Division of Cardiovascular Medicine and Intensive Care Medicine, Saint Mary's Hospital, Siegen, Germany (M.B.); Division of Vascular Surgery, Stanford University School of Medicine, CA (R.L.D.); and Department of Bioengineering, Stanford University School of Medicine, CA (E.K.)
| | - Suzanne Eken
- From Division of Cardiovascular Medicine, Stanford University School of Medicine, CA (U.R., I.N.S., R.T., F.N., Y.K., M.A., A.D., A.J., J.MS., P.S.T.); Cardiovascular Institute, Stanford University School of Medicine, CA (U.R., A.M.Z., F.N., M.B., F.C.E., Y.K., M.A., R.L.D., J.M.S., P.S.T.); VA Palo Alto Health Care System, CA (U.R., I.N.S., Y.K., M.A., A.D., A.J., J.M.S., P.S.T.); Department of Mechanical Engineering, Stanford University School of Medicine, CA (A.M.Z., E.K.); Department of Cardiothoracic Surgery, Stanford University School of Medicine, CA (F.C.E., E.K.); Department of Medicine, Karolinska Institute, Stockholm, Sweden (S.E., L.M.); Center for Vascular Medicine, Zwickau, Germany (T.H.); Division of Cardiovascular Medicine and Intensive Care Medicine, Saint Mary's Hospital, Siegen, Germany (M.B.); Division of Vascular Surgery, Stanford University School of Medicine, CA (R.L.D.); and Department of Bioengineering, Stanford University School of Medicine, CA (E.K.)
| | - Matti Adam
- From Division of Cardiovascular Medicine, Stanford University School of Medicine, CA (U.R., I.N.S., R.T., F.N., Y.K., M.A., A.D., A.J., J.MS., P.S.T.); Cardiovascular Institute, Stanford University School of Medicine, CA (U.R., A.M.Z., F.N., M.B., F.C.E., Y.K., M.A., R.L.D., J.M.S., P.S.T.); VA Palo Alto Health Care System, CA (U.R., I.N.S., Y.K., M.A., A.D., A.J., J.M.S., P.S.T.); Department of Mechanical Engineering, Stanford University School of Medicine, CA (A.M.Z., E.K.); Department of Cardiothoracic Surgery, Stanford University School of Medicine, CA (F.C.E., E.K.); Department of Medicine, Karolinska Institute, Stockholm, Sweden (S.E., L.M.); Center for Vascular Medicine, Zwickau, Germany (T.H.); Division of Cardiovascular Medicine and Intensive Care Medicine, Saint Mary's Hospital, Siegen, Germany (M.B.); Division of Vascular Surgery, Stanford University School of Medicine, CA (R.L.D.); and Department of Bioengineering, Stanford University School of Medicine, CA (E.K.)
| | - Lars Maegdefessel
- From Division of Cardiovascular Medicine, Stanford University School of Medicine, CA (U.R., I.N.S., R.T., F.N., Y.K., M.A., A.D., A.J., J.MS., P.S.T.); Cardiovascular Institute, Stanford University School of Medicine, CA (U.R., A.M.Z., F.N., M.B., F.C.E., Y.K., M.A., R.L.D., J.M.S., P.S.T.); VA Palo Alto Health Care System, CA (U.R., I.N.S., Y.K., M.A., A.D., A.J., J.M.S., P.S.T.); Department of Mechanical Engineering, Stanford University School of Medicine, CA (A.M.Z., E.K.); Department of Cardiothoracic Surgery, Stanford University School of Medicine, CA (F.C.E., E.K.); Department of Medicine, Karolinska Institute, Stockholm, Sweden (S.E., L.M.); Center for Vascular Medicine, Zwickau, Germany (T.H.); Division of Cardiovascular Medicine and Intensive Care Medicine, Saint Mary's Hospital, Siegen, Germany (M.B.); Division of Vascular Surgery, Stanford University School of Medicine, CA (R.L.D.); and Department of Bioengineering, Stanford University School of Medicine, CA (E.K.)
| | - Thomas Hertel
- From Division of Cardiovascular Medicine, Stanford University School of Medicine, CA (U.R., I.N.S., R.T., F.N., Y.K., M.A., A.D., A.J., J.MS., P.S.T.); Cardiovascular Institute, Stanford University School of Medicine, CA (U.R., A.M.Z., F.N., M.B., F.C.E., Y.K., M.A., R.L.D., J.M.S., P.S.T.); VA Palo Alto Health Care System, CA (U.R., I.N.S., Y.K., M.A., A.D., A.J., J.M.S., P.S.T.); Department of Mechanical Engineering, Stanford University School of Medicine, CA (A.M.Z., E.K.); Department of Cardiothoracic Surgery, Stanford University School of Medicine, CA (F.C.E., E.K.); Department of Medicine, Karolinska Institute, Stockholm, Sweden (S.E., L.M.); Center for Vascular Medicine, Zwickau, Germany (T.H.); Division of Cardiovascular Medicine and Intensive Care Medicine, Saint Mary's Hospital, Siegen, Germany (M.B.); Division of Vascular Surgery, Stanford University School of Medicine, CA (R.L.D.); and Department of Bioengineering, Stanford University School of Medicine, CA (E.K.)
| | - Alicia Deng
- From Division of Cardiovascular Medicine, Stanford University School of Medicine, CA (U.R., I.N.S., R.T., F.N., Y.K., M.A., A.D., A.J., J.MS., P.S.T.); Cardiovascular Institute, Stanford University School of Medicine, CA (U.R., A.M.Z., F.N., M.B., F.C.E., Y.K., M.A., R.L.D., J.M.S., P.S.T.); VA Palo Alto Health Care System, CA (U.R., I.N.S., Y.K., M.A., A.D., A.J., J.M.S., P.S.T.); Department of Mechanical Engineering, Stanford University School of Medicine, CA (A.M.Z., E.K.); Department of Cardiothoracic Surgery, Stanford University School of Medicine, CA (F.C.E., E.K.); Department of Medicine, Karolinska Institute, Stockholm, Sweden (S.E., L.M.); Center for Vascular Medicine, Zwickau, Germany (T.H.); Division of Cardiovascular Medicine and Intensive Care Medicine, Saint Mary's Hospital, Siegen, Germany (M.B.); Division of Vascular Surgery, Stanford University School of Medicine, CA (R.L.D.); and Department of Bioengineering, Stanford University School of Medicine, CA (E.K.)
| | - Ann Jagger
- From Division of Cardiovascular Medicine, Stanford University School of Medicine, CA (U.R., I.N.S., R.T., F.N., Y.K., M.A., A.D., A.J., J.MS., P.S.T.); Cardiovascular Institute, Stanford University School of Medicine, CA (U.R., A.M.Z., F.N., M.B., F.C.E., Y.K., M.A., R.L.D., J.M.S., P.S.T.); VA Palo Alto Health Care System, CA (U.R., I.N.S., Y.K., M.A., A.D., A.J., J.M.S., P.S.T.); Department of Mechanical Engineering, Stanford University School of Medicine, CA (A.M.Z., E.K.); Department of Cardiothoracic Surgery, Stanford University School of Medicine, CA (F.C.E., E.K.); Department of Medicine, Karolinska Institute, Stockholm, Sweden (S.E., L.M.); Center for Vascular Medicine, Zwickau, Germany (T.H.); Division of Cardiovascular Medicine and Intensive Care Medicine, Saint Mary's Hospital, Siegen, Germany (M.B.); Division of Vascular Surgery, Stanford University School of Medicine, CA (R.L.D.); and Department of Bioengineering, Stanford University School of Medicine, CA (E.K.)
| | - Michael Buerke
- From Division of Cardiovascular Medicine, Stanford University School of Medicine, CA (U.R., I.N.S., R.T., F.N., Y.K., M.A., A.D., A.J., J.MS., P.S.T.); Cardiovascular Institute, Stanford University School of Medicine, CA (U.R., A.M.Z., F.N., M.B., F.C.E., Y.K., M.A., R.L.D., J.M.S., P.S.T.); VA Palo Alto Health Care System, CA (U.R., I.N.S., Y.K., M.A., A.D., A.J., J.M.S., P.S.T.); Department of Mechanical Engineering, Stanford University School of Medicine, CA (A.M.Z., E.K.); Department of Cardiothoracic Surgery, Stanford University School of Medicine, CA (F.C.E., E.K.); Department of Medicine, Karolinska Institute, Stockholm, Sweden (S.E., L.M.); Center for Vascular Medicine, Zwickau, Germany (T.H.); Division of Cardiovascular Medicine and Intensive Care Medicine, Saint Mary's Hospital, Siegen, Germany (M.B.); Division of Vascular Surgery, Stanford University School of Medicine, CA (R.L.D.); and Department of Bioengineering, Stanford University School of Medicine, CA (E.K.)
| | - Ronald L Dalman
- From Division of Cardiovascular Medicine, Stanford University School of Medicine, CA (U.R., I.N.S., R.T., F.N., Y.K., M.A., A.D., A.J., J.MS., P.S.T.); Cardiovascular Institute, Stanford University School of Medicine, CA (U.R., A.M.Z., F.N., M.B., F.C.E., Y.K., M.A., R.L.D., J.M.S., P.S.T.); VA Palo Alto Health Care System, CA (U.R., I.N.S., Y.K., M.A., A.D., A.J., J.M.S., P.S.T.); Department of Mechanical Engineering, Stanford University School of Medicine, CA (A.M.Z., E.K.); Department of Cardiothoracic Surgery, Stanford University School of Medicine, CA (F.C.E., E.K.); Department of Medicine, Karolinska Institute, Stockholm, Sweden (S.E., L.M.); Center for Vascular Medicine, Zwickau, Germany (T.H.); Division of Cardiovascular Medicine and Intensive Care Medicine, Saint Mary's Hospital, Siegen, Germany (M.B.); Division of Vascular Surgery, Stanford University School of Medicine, CA (R.L.D.); and Department of Bioengineering, Stanford University School of Medicine, CA (E.K.)
| | - Joshua M Spin
- From Division of Cardiovascular Medicine, Stanford University School of Medicine, CA (U.R., I.N.S., R.T., F.N., Y.K., M.A., A.D., A.J., J.MS., P.S.T.); Cardiovascular Institute, Stanford University School of Medicine, CA (U.R., A.M.Z., F.N., M.B., F.C.E., Y.K., M.A., R.L.D., J.M.S., P.S.T.); VA Palo Alto Health Care System, CA (U.R., I.N.S., Y.K., M.A., A.D., A.J., J.M.S., P.S.T.); Department of Mechanical Engineering, Stanford University School of Medicine, CA (A.M.Z., E.K.); Department of Cardiothoracic Surgery, Stanford University School of Medicine, CA (F.C.E., E.K.); Department of Medicine, Karolinska Institute, Stockholm, Sweden (S.E., L.M.); Center for Vascular Medicine, Zwickau, Germany (T.H.); Division of Cardiovascular Medicine and Intensive Care Medicine, Saint Mary's Hospital, Siegen, Germany (M.B.); Division of Vascular Surgery, Stanford University School of Medicine, CA (R.L.D.); and Department of Bioengineering, Stanford University School of Medicine, CA (E.K.)
| | - Ellen Kuhl
- From Division of Cardiovascular Medicine, Stanford University School of Medicine, CA (U.R., I.N.S., R.T., F.N., Y.K., M.A., A.D., A.J., J.MS., P.S.T.); Cardiovascular Institute, Stanford University School of Medicine, CA (U.R., A.M.Z., F.N., M.B., F.C.E., Y.K., M.A., R.L.D., J.M.S., P.S.T.); VA Palo Alto Health Care System, CA (U.R., I.N.S., Y.K., M.A., A.D., A.J., J.M.S., P.S.T.); Department of Mechanical Engineering, Stanford University School of Medicine, CA (A.M.Z., E.K.); Department of Cardiothoracic Surgery, Stanford University School of Medicine, CA (F.C.E., E.K.); Department of Medicine, Karolinska Institute, Stockholm, Sweden (S.E., L.M.); Center for Vascular Medicine, Zwickau, Germany (T.H.); Division of Cardiovascular Medicine and Intensive Care Medicine, Saint Mary's Hospital, Siegen, Germany (M.B.); Division of Vascular Surgery, Stanford University School of Medicine, CA (R.L.D.); and Department of Bioengineering, Stanford University School of Medicine, CA (E.K.)
| | - Philip S Tsao
- From Division of Cardiovascular Medicine, Stanford University School of Medicine, CA (U.R., I.N.S., R.T., F.N., Y.K., M.A., A.D., A.J., J.MS., P.S.T.); Cardiovascular Institute, Stanford University School of Medicine, CA (U.R., A.M.Z., F.N., M.B., F.C.E., Y.K., M.A., R.L.D., J.M.S., P.S.T.); VA Palo Alto Health Care System, CA (U.R., I.N.S., Y.K., M.A., A.D., A.J., J.M.S., P.S.T.); Department of Mechanical Engineering, Stanford University School of Medicine, CA (A.M.Z., E.K.); Department of Cardiothoracic Surgery, Stanford University School of Medicine, CA (F.C.E., E.K.); Department of Medicine, Karolinska Institute, Stockholm, Sweden (S.E., L.M.); Center for Vascular Medicine, Zwickau, Germany (T.H.); Division of Cardiovascular Medicine and Intensive Care Medicine, Saint Mary's Hospital, Siegen, Germany (M.B.); Division of Vascular Surgery, Stanford University School of Medicine, CA (R.L.D.); and Department of Bioengineering, Stanford University School of Medicine, CA (E.K.).
| |
Collapse
|
37
|
Wang Q, Shu C, Su J, Li X. A crosstalk triggered by hypoxia and maintained by MCP-1/miR-98/IL-6/p38 regulatory loop between human aortic smooth muscle cells and macrophages leads to aortic smooth muscle cells apoptosis via Stat1 activation. INTERNATIONAL JOURNAL OF CLINICAL AND EXPERIMENTAL PATHOLOGY 2015; 8:2670-2679. [PMID: 26045772 PMCID: PMC4440081] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 12/20/2014] [Accepted: 02/21/2015] [Indexed: 06/04/2023]
Abstract
Hypoxia and inflammation are central characteristics of the abdominal aortic aneurysm (AAA), but the mechanisms for their relationship and actual role remain far from full understood. Here, we showed MCP-1 (monocyte chemotactic protein-1) induced by hypoxia in primary human Aortic Smooth Muscle Cells (hASMCs) increased the chemotaxis of THP-1 macrophages and MCP-1 induced IL-6 expression in THP-1 cells via downregulating miR-98 which directly targets IL-6. In addition, IL-6 positively feedback regulated MCP-1 expression in hASMCs via p38 signal that is independent on hypoxia, and inhibition of p38 signal blocked the effect of IL-6 on MCP-1 expression regulation. Moreover, IL-6 exposure time-dependently induces phASMCs apoptosis via Stat1 activation. Collectively, our data provide compelling evidence on the association between hypoxia and inflammation triggered by hypoxia and then mediated by MCP-1/miR-98/IL-6/p38 regulatory loop, which leads to hASMCs apoptosis via Stat1 activation to contribute to AAA formation and progression.
Collapse
MESH Headings
- Aorta/metabolism
- Aorta/pathology
- Aortic Aneurysm, Abdominal/metabolism
- Aortic Aneurysm, Abdominal/pathology
- Apoptosis/physiology
- Blotting, Western
- Cell Hypoxia
- Cells, Cultured
- Chemokine CCL2/metabolism
- Enzyme-Linked Immunosorbent Assay
- Humans
- Interleukin-6/metabolism
- MAP Kinase Signaling System/physiology
- Macrophages/metabolism
- MicroRNAs/metabolism
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/pathology
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/pathology
- Real-Time Polymerase Chain Reaction
- Receptor Cross-Talk/physiology
- STAT1 Transcription Factor/metabolism
- Transfection
Collapse
Affiliation(s)
- Qing Wang
- Department of Vascular Surgery, The 2 Xiangya Hospital, Central South University139 Renmin Middle Road, Changsha 410011, Hunan, People’s Republic of China
| | - Chang Shu
- Department of Vascular Surgery, The 2 Xiangya Hospital, Central South University139 Renmin Middle Road, Changsha 410011, Hunan, People’s Republic of China
| | - Jing Su
- Hunan Tumor HospitalChangsha, Hunan, People’s Republic of China
| | - Xin Li
- Department of Vascular Surgery, The 2 Xiangya Hospital, Central South University139 Renmin Middle Road, Changsha 410011, Hunan, People’s Republic of China
| |
Collapse
|
38
|
Gao F, Chambon P, Tellides G, Kong W, Zhang X, Li W. Disruption of TGF-β signaling in smooth muscle cell prevents flow-induced vascular remodeling. Biochem Biophys Res Commun 2014; 454:245-50. [PMID: 25451249 DOI: 10.1016/j.bbrc.2014.10.092] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2014] [Accepted: 10/19/2014] [Indexed: 01/09/2023]
Abstract
Transforming growth factor-β (TGF-β) signaling has been prominently implicated in the pathogenesis of vascular remodeling, especially the initiation and progression of flow-induced vascular remodeling. Smooth muscle cells (SMCs) are the principal resident cells in arterial wall and are critical for arterial remodeling. However, the role of TGF-β signaling in SMC for flow-induced vascular remodeling remains unknown. Therefore, the goal of our study was to determine the effect of TGF-β pathway in SMC for vascular remodeling, by using a genetical smooth muscle-specific (SM-specific) TGF-β type II receptor (Tgfbr2) deletion mice model. Mice deficient in the expression of Tgfbr2 (MyhCre.Tgfbr2(f/f)) and their corresponding wild-type background mice (MyhCre.Tgfbr2(WT/WT)) underwent partial ligation of left common carotid artery for 1, 2, or 4 weeks. Then the carotid arteries were harvested and indicated that the disruption of Tgfbr2 in SMC provided prominent inhibition of vascular remodeling. And the thickening of carotid media, proliferation of SMC, infiltration of macrophage, and expression of matrix metalloproteinase (MMP) were all significantly attenuated in Tgfbr2 disruption mice. Our study demonstrated, for the first time, that the TGF-β signaling in SMC plays an essential role in flow-induced vascular remodeling and disruption can prevent this process.
Collapse
Affiliation(s)
- Fu Gao
- Department of Vascular Surgery, Peking University People's Hospital, Beijing, People's Republic of China
| | - Pierre Chambon
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (CNRS UMR7104; INSERM U596; ULP, Collége de France) and Institut Clinique de la Souris, ILLKIRCH, Strasbourg, France
| | - George Tellides
- Department of Surgery, Interdepartmental Program in Vascular Biology and Therapeutics, Yale University School of Medicine, New Haven, CT, USA
| | - Wei Kong
- Department of Physiology and Pathophysiology, Basic Medical College of Peking University, Beijing, People's Republic of China
| | - Xiaoming Zhang
- Department of Vascular Surgery, Peking University People's Hospital, Beijing, People's Republic of China.
| | - Wei Li
- Department of Vascular Surgery, Peking University People's Hospital, Beijing, People's Republic of China
| |
Collapse
|
39
|
Gao F, Chambon P, Offermanns S, Tellides G, Kong W, Zhang X, Li W. Disruption of TGF-β signaling in smooth muscle cell prevents elastase-induced abdominal aortic aneurysm. Biochem Biophys Res Commun 2014; 454:137-43. [PMID: 25450370 DOI: 10.1016/j.bbrc.2014.10.053] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2014] [Accepted: 10/11/2014] [Indexed: 12/21/2022]
Abstract
Transforming growth factor-β (TGF-β) signaling has been significantly implicated in the pathogenesis of aneurysm, prominently the initiation and progression of abdominal aortic aneurysm (AAA). Vascular smooth muscle cell (SMC) is the principal resident cell in aortic wall and is essential for its structure and function. However, the role of TGF-β pathway in SMC for the formation of AAA remains unknown. Therefore, the goal of the present study was to investigate the effect of TGF-β pathway in SMC for AAA pathogenesis, by using a genetical smooth muscle-specific (SM-specific) TGF-β type II receptor (Tgfbr2) disruption animal model. Mice deficient in the expression of Tgfbr2 (MyhCre.Tgfbr2(f/f) and MyhCre.Tgfbr2(WT/f)) and their corresponding wild-type background mice (MyhCre.Tgfbr2(WT/WT)) underwent AAA induction by infrarenal peri-adventitial application of elastase. Fourteen days after elastase treatment, the aortas were analyzed and indicated that disruption of 1 or 2 alleles of Tgfbr2 in SMC provided markedly step-wise protection from AAA formation. And elastin degradation, medial SMC loss, macrophage infiltration, and matrix metalloproteinases (MMP) expression were all significantly reduced in Tgfbr2 deletion mice. Our study demonstrated, for the first time, that the TGF-β signaling pathway in SMC plays a critical role in AAA and disruption can prevent the aneurysm formation.
Collapse
Affiliation(s)
- Fu Gao
- Department of Vascular Surgery, Peking University People's Hospital, Beijing, People's Republic of China
| | - Pierre Chambon
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (CNRS UMR7104; INSERM U596; ULP, Collége de France) and Institut Clinique de la Souris, ILLKIRCH, Strasbourg, France
| | - Stefan Offermanns
- Department of Pharmacology, Max-Planck-Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - George Tellides
- Department of Surgery, Interdepartmental Program in Vascular Biology and Therapeutics, Yale University School of Medicine, New Haven, CT, USA
| | - Wei Kong
- Department of Physiology and Pathophysiology, Basic Medical College of Peking University, Beijing, People's Republic of China
| | - Xiaoming Zhang
- Department of Vascular Surgery, Peking University People's Hospital, Beijing, People's Republic of China.
| | - Wei Li
- Department of Vascular Surgery, Peking University People's Hospital, Beijing, People's Republic of China
| |
Collapse
|
40
|
B2 cells suppress experimental abdominal aortic aneurysms. THE AMERICAN JOURNAL OF PATHOLOGY 2014; 184:3130-41. [PMID: 25194661 DOI: 10.1016/j.ajpath.2014.07.006] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/27/2014] [Revised: 06/16/2014] [Accepted: 07/10/2014] [Indexed: 12/11/2022]
Abstract
Recent reports of rupture in patients with abdominal aortic aneurysm (AAA) receiving B-cell depletion therapy highlight the importance of understanding the role of B cells (B1 and B2 subsets) in the development of AAA. We hypothesized that B2 cells aggravate experimental aneurysm formation. The IHC staining revealed infiltration of B cells in the aorta of wild-type (C57BL/6) mice at day 7 after elastase perfusion and persisted through day 21. Quantification of immune cell types using flow cytometry at day 14 showed significantly greater infiltration of mononuclear cells, including B cells (B2: 93% of total B cells) and T cells in elastase-perfused aortas compared with saline-perfused or normal aortas. muMT (mature B-cell deficient) mice were prone to AAA formation similar to wild-type mice in two different experimental AAA models. Contradicting our hypothesis, adoptive transfer of B2 cells suppressed AAA formation (102.0% ± 7.3% versus 75.2% ± 5.5%; P < 0.05) with concomitant increase in the splenic regulatory T cell (0.24% ± 0.03% versus 0.92% ± 0.23%; P < 0.05) and decrease in aortic infiltration of mononuclear cells. Our data suggest that B2 cells constitute the largest population of B cells in experimental AAA. Furthermore, B2 cells, in the absence of other B-cell subsets, increase splenic regulatory T-cell population and suppress AAA formation.
Collapse
|
41
|
Tarín C, Fernández-Laso V, Sastre C, Madrigal-Matute J, Gómez M, Zaragoza C, Egido J, Burkly LC, Martín-Ventura JL, Blanco-Colio LM. Tumor necrosis factor-like weak inducer of apoptosis or Fn14 deficiency reduce elastase perfusion-induced aortic abdominal aneurysm in mice. J Am Heart Assoc 2014; 3:jah3567. [PMID: 25092786 PMCID: PMC4310358 DOI: 10.1161/jaha.113.000723] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Background Abdominal aortic aneurysm (AAA) involves leukocyte recruitment, inflammatory cytokine production, vascular cell apoptosis, neovascularization, and vascular remodeling, all of which contribute to aortic dilatation. Tumor necrosis factor‐like weak inducer of apoptosis (TWEAK) is a cytokine implicated in proinflammatory responses, angiogenesis, and matrix degradation but its role in AAA formation is currently unknown. Methods and Results Experimental AAA with aortic elastase perfusion in mice was induced in wild‐type (WT), TWEAK deficient (TWEAK KO), or Fn14‐deficient (Fn14 KO) mice. TWEAK or Fn14 KO deficiency reduced aortic expansion, lesion macrophages, CD3+ T cells, neutrophils, CD31+ microvessels, CCL2 and CCL5 chemokines expression, and MMP activity after 14 days postperfusion. TWEAK and Fn14 KO mice also showed a reduced loss of medial vascular smooth muscle cells (VSMC) that was related to a reduced number of apoptotic cells in these animals compared with WT mice. Aortas from WT animals present a higher disruption of the elastic layer and MMP activity than those from TWEAK or Fn14 KO mice, indicating a diminished vascular remodeling in KO animals. In vitro experiments unveiled that TWEAK induces CCL5 secretion and MMP‐9 activation in both VSMC and bone marrow‐derived macrophages, and decrease VSMC viability, effects dependent on Fn14. Conclusions TWEAK/Fn14 axis participates in AAA formation by promoting lesion inflammatory cell accumulation, angiogenesis, matrix‐degrading protease expression, and vascular remodeling. Blocking TWEAK/Fn14 interaction could be a new target for the treatment of AAA.
Collapse
Affiliation(s)
- Carlos Tarín
- Vascular Research Lab, IIS-Fundación Jiménez Díaz, Madrid, Spain (C.T., V.F.L., C.S., J.M.M., J.E., J.L.M.V., L.M.B.C.)
| | - Valvanera Fernández-Laso
- Vascular Research Lab, IIS-Fundación Jiménez Díaz, Madrid, Spain (C.T., V.F.L., C.S., J.M.M., J.E., J.L.M.V., L.M.B.C.)
| | - Cristina Sastre
- Vascular Research Lab, IIS-Fundación Jiménez Díaz, Madrid, Spain (C.T., V.F.L., C.S., J.M.M., J.E., J.L.M.V., L.M.B.C.)
| | - Julio Madrigal-Matute
- Vascular Research Lab, IIS-Fundación Jiménez Díaz, Madrid, Spain (C.T., V.F.L., C.S., J.M.M., J.E., J.L.M.V., L.M.B.C.)
| | - Mónica Gómez
- Spanish National Cardiovascular Research Center, Madrid, Spain (, C.Z.)
| | - Carlos Zaragoza
- Cardiovascular Joint Research Unit, University Hospital Ramón y Cajal Hospital and University Francisco de Vitoria School of Medicine, Madrid, Spain
| | - Jesús Egido
- Vascular Research Lab, IIS-Fundación Jiménez Díaz, Madrid, Spain (C.T., V.F.L., C.S., J.M.M., J.E., J.L.M.V., L.M.B.C.)
| | | | - Jose L Martín-Ventura
- Vascular Research Lab, IIS-Fundación Jiménez Díaz, Madrid, Spain (C.T., V.F.L., C.S., J.M.M., J.E., J.L.M.V., L.M.B.C.)
| | - Luis M Blanco-Colio
- Vascular Research Lab, IIS-Fundación Jiménez Díaz, Madrid, Spain (C.T., V.F.L., C.S., J.M.M., J.E., J.L.M.V., L.M.B.C.)
| |
Collapse
|
42
|
Johnson JL. Emerging regulators of vascular smooth muscle cell function in the development and progression of atherosclerosis. Cardiovasc Res 2014; 103:452-60. [PMID: 25053639 DOI: 10.1093/cvr/cvu171] [Citation(s) in RCA: 108] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
After a period of relative senescence in the field of vascular smooth muscle cell (VSMC) research with particular regards to atherosclerosis, the last few years has witnessed a resurgence, with extensive research re-assessing potential molecular mechanisms and pathways that modulate VSMC behaviour within the atherosclerotic-prone vessel wall and the atherosclerotic plaque itself. Attention has focussed on the pathological contribution of VSMC in plaque calcification; systemic and local mediators such as inflammatory molecules and lipoproteins; autocrine and paracrine regulators which affect cell-cell and cell to matrix contacts alongside cytoskeletal changes. In this brief focused review, recent insights that have been gained into how a myriad of recently identified factors can influence the pathological behaviour of VSMC and their subsequent contribution to atherosclerotic plaque development and progression has been discussed. An overriding theme is the mechanisms involved in the alterations of VSMC function during atherosclerosis.
Collapse
Affiliation(s)
- Jason Lee Johnson
- Laboratory of Cardiovascular Pathology, School of Clinical Sciences, University of Bristol, Research Floor Level Seven, Bristol Royal Infirmary, Bristol BS2 8HW, UK
| |
Collapse
|
43
|
Michineau S, Franck G, Wagner-Ballon O, Dai J, Allaire E, Gervais M. Chemokine (C-X-C motif) receptor 4 blockade by AMD3100 inhibits experimental abdominal aortic aneurysm expansion through anti-inflammatory effects. Arterioscler Thromb Vasc Biol 2014; 34:1747-55. [PMID: 24876351 DOI: 10.1161/atvbaha.114.303913] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
OBJECTIVE Inflammation plays a critical role in the development of abdominal aortic aneurysms (AAAs). Because stromal cell-derived factor 1 (SDF-1) is known for its ability to attract inflammatory cells, we investigated whether SDF-1/chemokine (C-X-C motif) receptor 4 (CXCR4) axis is expressed in aneurysmal aortic wall and plays a role in AAA physiopathology and asked whether its blockade modulates AAA formation and expansion. APPROACH AND RESULTS Quantitative real-time polymerase chain reaction analysis showed that SDF-1α and CXCR4 mRNA levels are increased in both human and CaCl2-induced mouse AAA wall and are positively correlated to the aortic diameter in mice. ELISA quantification and immunostaining demonstrated that, in mice, aortic SDF-1α is rapidly induced during AAA formation, first by apoptotic vascular smooth muscle cells in the injured media and then by adventitial macrophages once AAA is fully established. Using green fluorescent protein-positive (GFP(+/-)) bone marrow transplantation experiments, we demonstrated that aortic SDF-1 overexpression is implicated in the recruitment of bone marrow-derived macrophages within the AAA wall. Furthermore, in mice, blockade of CXCR4 by AMD3100 decreases the infiltration of adventitial macrophages, inhibits AAA formation, and prevents aortic wall destruction. AMD3100 reduces the mRNA levels of MMP-12 and MMP-14 as well as that of inflammatory effectors MCP-1, MIP-1β, MIP-2α, RANTES, IL-1β, IL-6, TNF-α, and E-selectin. Finally, AMD3100 stabilizes the diameter of formed, expanding AAAs in 2 experimental models. CONCLUSIONS SDF-1/CXCR4 axis is upregulated in human and mouse AAAs. Blockade of CXCR4 with AMD3100 suppresses AAA formation and progression in two rodent models. Blockade of SDF-1/CXCR4 axis may represent a new strategy to limit progression of small human AAAs.
Collapse
Affiliation(s)
- Stéphanie Michineau
- From the CNRS EAC 7054, Centre de Recherches Chirurgicales Dominique Chopin, Faculty of Medicine, Paris-Est Créteil University (UPEC), Créteil, France (S.M., G.F., J.D., E.A., M.G.); and Department of Hematology-Immunology, AP-HP, Henri Mondor Hospital, UPEC, Créteil, France (O.W.-B.)
| | - Grégory Franck
- From the CNRS EAC 7054, Centre de Recherches Chirurgicales Dominique Chopin, Faculty of Medicine, Paris-Est Créteil University (UPEC), Créteil, France (S.M., G.F., J.D., E.A., M.G.); and Department of Hematology-Immunology, AP-HP, Henri Mondor Hospital, UPEC, Créteil, France (O.W.-B.)
| | - Orianne Wagner-Ballon
- From the CNRS EAC 7054, Centre de Recherches Chirurgicales Dominique Chopin, Faculty of Medicine, Paris-Est Créteil University (UPEC), Créteil, France (S.M., G.F., J.D., E.A., M.G.); and Department of Hematology-Immunology, AP-HP, Henri Mondor Hospital, UPEC, Créteil, France (O.W.-B.)
| | - Jianping Dai
- From the CNRS EAC 7054, Centre de Recherches Chirurgicales Dominique Chopin, Faculty of Medicine, Paris-Est Créteil University (UPEC), Créteil, France (S.M., G.F., J.D., E.A., M.G.); and Department of Hematology-Immunology, AP-HP, Henri Mondor Hospital, UPEC, Créteil, France (O.W.-B.)
| | - Eric Allaire
- From the CNRS EAC 7054, Centre de Recherches Chirurgicales Dominique Chopin, Faculty of Medicine, Paris-Est Créteil University (UPEC), Créteil, France (S.M., G.F., J.D., E.A., M.G.); and Department of Hematology-Immunology, AP-HP, Henri Mondor Hospital, UPEC, Créteil, France (O.W.-B.)
| | - Marianne Gervais
- From the CNRS EAC 7054, Centre de Recherches Chirurgicales Dominique Chopin, Faculty of Medicine, Paris-Est Créteil University (UPEC), Créteil, France (S.M., G.F., J.D., E.A., M.G.); and Department of Hematology-Immunology, AP-HP, Henri Mondor Hospital, UPEC, Créteil, France (O.W.-B.)
| |
Collapse
|
44
|
Liu M, Chen Y, Yang X, Zhang L, Zhao T, Zhao B, Jia L, Zhu Y, Gao X, Zhang B, Li X, Xiang R, Han J, Duan Y. DanHong Injection inhibits the development of primary abdominal aortic aneurysms in apoE knockout mice. CHINESE SCIENCE BULLETIN-CHINESE 2014. [DOI: 10.1007/s11434-014-0175-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
45
|
Wang Q, Ren J, Morgan S, Liu Z, Dou C, Liu B. Monocyte chemoattractant protein-1 (MCP-1) regulates macrophage cytotoxicity in abdominal aortic aneurysm. PLoS One 2014; 9:e92053. [PMID: 24632850 PMCID: PMC3954911 DOI: 10.1371/journal.pone.0092053] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2013] [Accepted: 02/17/2014] [Indexed: 11/18/2022] Open
Abstract
Aims In abdominal aortic aneurysm (AAA), macrophages are detected in the proximity of aortic smooth muscle cells (SMCs). We have previously demonstrated in a murine model of AAA that apoptotic SMCs attract monocytes and other leukocytes by producing MCP-1. Here we tested whether infiltrating macrophages also directly contribute to SMC apoptosis. Methods and Results Using a SMC/RAW264.7 macrophage co-culture system, we demonstrated that MCP-1-primed RAWs caused a significantly higher level of apoptosis in SMCs as compared to control macrophages. Next, we detected an enhanced Fas ligand (FasL) mRNA level and membrane FasL protein expression in MCP-1-primed RAWs. Neutralizing FasL blocked SMC apoptosis in the co-culture. In situ proximity ligation assay showed that SMCs exposed to primed macrophages contained higher levels of receptor interacting protein-1 (RIP1)/Caspase 8 containing cell death complexes. Silencing RIP1 conferred apoptosis resistance to SMCs. In the mouse elastase injury model of aneurysm, aneurysm induction increased the level of RIP1/Caspase 8 containing complexes in medial SMCs. Moreover, TUNEL-positive SMCs in aneurysmal tissues were frequently surrounded by CD68+/FasL+ macrophages. Conversely, elastase-treated arteries from MCP-1 knockout mice display a reduction of both macrophage infiltration and FasL expression, which was accompanied by diminished apoptosis of SMCs. Conclusion Our data suggest that MCP-1-primed macrophages are more cytotoxic. MCP-1 appears to modulate macrophage cytotoxicity by increasing the level of membrane bound FasL. Thus, we showed that MCP-1-primed macrophages kill SMCs through a FasL/Fas-Caspase8-RIP1 mediated mechanism.
Collapse
Affiliation(s)
- Qiwei Wang
- Division of Vascular Surgery, Department of Surgery, University of Wisconsin-Madison, Wisconsin, United States of America
| | - Jun Ren
- Division of Vascular Surgery, Department of Surgery, University of Wisconsin-Madison, Wisconsin, United States of America
| | - Stephanie Morgan
- Division of Vascular Surgery, Department of Surgery, University of Wisconsin-Madison, Wisconsin, United States of America
| | - Zhenjie Liu
- Division of Vascular Surgery, Department of Surgery, University of Wisconsin-Madison, Wisconsin, United States of America
| | | | - Bo Liu
- Division of Vascular Surgery, Department of Surgery, University of Wisconsin-Madison, Wisconsin, United States of America
- * E-mail:
| |
Collapse
|
46
|
Increased expression of chitinase 3-like 1 in aorta of patients with atherosclerosis and suppression of atherosclerosis in apolipoprotein E-knockout mice by chitinase 3-like 1 gene silencing. Mediators Inflamm 2014; 2014:905463. [PMID: 24729664 PMCID: PMC3960764 DOI: 10.1155/2014/905463] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2013] [Revised: 11/27/2013] [Accepted: 01/23/2014] [Indexed: 11/18/2022] Open
Abstract
INTRODUCTION The purpose of this study was to investigate the changes of chitinase 3-like 1 (CHI3L1) in the aorta of patients with coronary atherosclerosis and to determine whether inhibition of CHI3L1 by lentivirus-mediated RNA interference could stabilize atherosclerotic plaques in apolipoprotein E-knockout (ApoE(-/-)) mice. METHODS We collected discarded aortic specimens from patients undergoing coronary artery bypass graft surgery and renal arterial tissues from kidney donors. A lentivirus carrying small interfering RNA targeting the expression of CHI3L1 was constructed. Fifty ApoE(-/-) mice were divided into control group and CHI3L1 gene silenced group. A constrictive collar was placed around carotid artery to induce plaques formation. Then lentivirus was transfected into carotid plaques. RESULTS We found that CHI3L1 was overexpressed in aorta of patients with atherosclerosis and its expression was correlated with the atherosclerotic risk factors. After lentivirus transduction, mRNA and protein expression of CHI3L1 were attenuated in carotid plaques, leading to reduced plaque content of lipids and macrophages, and increased plaque content of collagen and smooth muscle cells. Moreover, CHI3L1 gene silencing downregulated the expression of local proinflammatory mediators. CONCLUSIONS CHI3L1 is overexpressed in aorta from patients with atherosclerosis and the lentivirus-mediated CHI3L1 gene silencing could represent a new strategy to inhibit plaques progression.
Collapse
|
47
|
Johnston WF, Salmon M, Su G, Lu G, Ailawadi G, Upchurch GR. Aromatase is required for female abdominal aortic aneurysm protection. J Vasc Surg 2014; 61:1565-74.e1-4. [PMID: 24582702 DOI: 10.1016/j.jvs.2014.01.032] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2013] [Revised: 01/09/2014] [Accepted: 01/12/2014] [Indexed: 01/09/2023]
Abstract
OBJECTIVE The protective effects of female gender on the development of abdominal aortic aneurysms (AAAs) have been attributed to anti-inflammatory effects of estrogen. Estrogen synthesis is dependent on the enzyme aromatase, which is located both centrally in the ovaries and peripherally in adipose tissue, bone, and vascular smooth muscle cells. It is hypothesized that deletion of aromatase in both ovarian and peripheral tissues would diminish the protective effect of female gender and would be associated with increased aortic diameter in female mice. METHODS Male and female 8- to 10-week-old mice with aromatase (wild type: WT) and without aromatase (ArKO) underwent elastase aortic perfusion with aortic harvest 14 days following. For the contribution of central and peripheral estrogen conversion to be evaluated, female WT mice were compared with female WT and ArKO mice that had undergone ovariectomy (ovx) at 6 weeks followed by elastase perfusion at 8 to 10 weeks. At aortic harvest, maximal aortic dilation was measured and samples were collected for immunohistochemistry and protein analysis. Serum was collected for serum estradiol concentrations. Groups were compared with analysis of variance. Human and mouse AAA cross sections were analyzed with confocal immunohistochemistry for aromatase, smooth muscle markers, and macrophage markers. RESULTS Female WT mice had significant reduction in aortic dilation compared with male WT mice (F WT, 51.5% ± 15.1% vs M WT, 78.7% ± 14.9%; P < .005). The protective effects of female gender were completely eliminated with deletion of aromatase (F ArKO, 82.6% ± 13.8%; P < .05 vs F WT). Ovariectomy increased aortic dilation in WT mice (F WT ovx, 70.6% ± 11.7%; P < .05 vs F WT). Aromatase deletion with ovariectomy further increased aortic dilation compared with WT ovx mice (F ArKO ovx, 87.3% ± 14.7%, P < .001 vs F WT and P < .05 vs F WT ovx). Accordingly, female ArKO ovx mice had significantly higher levels of the proinflammatory cytokines monocyte chemoattractant protein 1 and interleukin-1β and were associated with increased macrophage staining and decreased elastin staining. Regarding serum hormone levels, decreasing estradiol levels correlated with increasing aortic diameter (R = -0.565; P < .01). By confocal immunohistochemistry, both human and mouse AAA smooth muscle cells (smooth muscle α-actin positive) and macrophages (CD68 positive or Mac-2 positive) expressed aromatase. CONCLUSIONS The protective effect of female gender on AAAs is due to estrogen synthesis and requires the presence of both ovarian and extragonadal/peripheral aromatase. Peripheral estrogen synthesis accounts for roughly half of the protective effect of female gender. If peripheral aromatase could be targeted, high levels of local estrogen could be produced and may avoid the side effects of systemic estrogen.
Collapse
Affiliation(s)
- William F Johnston
- Division of Vascular and Endovascular Surgery, Department of Surgery, University of Virginia, Charlottesville, Va
| | - Morgan Salmon
- Division of Vascular and Endovascular Surgery, Department of Surgery, University of Virginia, Charlottesville, Va
| | - Gang Su
- Division of Vascular and Endovascular Surgery, Department of Surgery, University of Virginia, Charlottesville, Va
| | - Guanyi Lu
- Division of Vascular and Endovascular Surgery, Department of Surgery, University of Virginia, Charlottesville, Va
| | - Gorav Ailawadi
- Division of Cardiothoracic Surgery, Department of Surgery, University of Virginia, Charlottesville, Va; Robert M. Berne Cardiovascular Research Center, University of Virginia, Charlottesville, Va
| | - Gilbert R Upchurch
- Division of Vascular and Endovascular Surgery, Department of Surgery, University of Virginia, Charlottesville, Va; Robert M. Berne Cardiovascular Research Center, University of Virginia, Charlottesville, Va; Department of Molecular Physiology and Biological Physics, University of Virginia, Charlottesville, Va.
| |
Collapse
|
48
|
Lu G, Su G, Zhao Y, Johnston WF, Sherman NE, Rissman EF, Lau C, Ailawadi G, Upchurch GR. Dietary phytoestrogens inhibit experimental aneurysm formation in male mice. J Surg Res 2013; 188:326-38. [PMID: 24388399 DOI: 10.1016/j.jss.2013.11.1108] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2013] [Revised: 11/11/2013] [Accepted: 11/21/2013] [Indexed: 11/16/2022]
Abstract
BACKGROUND The purpose of these experiments was to test the hypothesis that dietary phytoestrogens would diminish experimental aortic aneurysm formation. MATERIALS AND METHODS Six-wk-old C57BL/6 mice were divided into groups, fed either a diet with minimal phytoestrogen content or a regular commercial rodent diet with high phytoestrogen content for 2 wk. At the age of 8 wk, aortic aneurysms were induced by infusing the isolated infrarenal abdominal aorta with 0.4% elastase for 5 min. Mice were recovered and the diameter of the infused aorta was measured at postoperative days 3, 7, and 14. Abdominal aorta samples were collected for histology, cytokine array, and gelatin zymography after aortic diameter measurement. Blood samples were also collected to determine serum phytoestrogens and estradiol levels. Multiple-group comparisons were done using an analysis of variance with post hoc Tukey tests. RESULTS Compared with mice on a minimal phytoestrogen diet, mice on a regular rodent diet had higher levels of serum phytoestrogens (male, 1138 ± 846 ng/dL; female, 310 ± 295 ng/dL). These serum phytoestrogen levels were also much higher than their own endogenous estradiol levels (109-fold higher for males and 35.5-fold higher for females). Although aortic diameters of female mice were unaffected by the phytoestrogen concentration in the diets, male mice on the regular rodent diet (M+ group) developed smaller aortic aneurysms than male mice on the minimal phytoestrogen diet (M- group) on postoperative day 14 (M+ 54.8 ± 8.8% versus M- 109.3 ± 37.6%; P < 0.001). During aneurysm development (postoperative days 3 and 7), there were fewer neutrophils, macrophages, and lymphocytes in the aorta from the M+ group than from the M- group. Concentrations of multiple proinflammatory cytokines (matrix metalloproteinases [MMPs]; interleukin 1β [IL-1β]; IL-6; IL-17; IL-23; monocyte chemoattractant protein-1; regulated on activation, normal T cell expressed and secreted; interferon γ; and tumor necrosis factor α) from aortas of the M+ group were also lower than those from the aortas of the M- group. Zymography also demonstrated that the M+ group had lower levels of aortic MMP-9s than the M- group on postoperative day 14 (P < 0.001 for pro-MMP-9, P < 0.001 for active MMP-9). CONCLUSIONS These results suggest that dietary phytoestrogens inhibit experimental aortic aneurysm formation in male mice via a reduction of the inflammatory response in the aorta wall. The protective effect of dietary phytoestrogens on aneurysm formation warrants further investigation.
Collapse
Affiliation(s)
- Guanyi Lu
- Department of Surgery, University of Virginia Health System, Charlottesville, Virginia
| | - Gang Su
- Department of Surgery, University of Virginia Health System, Charlottesville, Virginia
| | - Yunge Zhao
- Department of Surgery, University of Virginia Health System, Charlottesville, Virginia
| | - William F Johnston
- Department of Surgery, University of Virginia Health System, Charlottesville, Virginia
| | - Nicholas E Sherman
- Department of Microbiology, W.M. Keck Biomedical Mass Spectrometry Laboratory, University of Virginia School of Medicine, Charlottesville, Virginia
| | - Emilie F Rissman
- Department of Biochemistry and Molecular Genetics, University of Virginia School of Medicine, Charlottesville, Virginia
| | - Christine Lau
- Department of Surgery, University of Virginia Health System, Charlottesville, Virginia
| | - Gorav Ailawadi
- Department of Surgery, University of Virginia Health System, Charlottesville, Virginia
| | - Gilbert R Upchurch
- Department of Surgery, University of Virginia Health System, Charlottesville, Virginia.
| |
Collapse
|
49
|
Marinković G, Hibender S, Hoogenboezem M, van Broekhoven A, Girigorie AF, Bleeker N, Hamers AA, Stap J, van Buul JD, de Vries CJ, de Waard V. Immunosuppressive Drug Azathioprine Reduces Aneurysm Progression Through Inhibition of Rac1 and c-Jun-Terminal-N-Kinase in Endothelial Cells. Arterioscler Thromb Vasc Biol 2013; 33:2380-8. [DOI: 10.1161/atvbaha.113.301394] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Affiliation(s)
- Goran Marinković
- From the Department of Medical Biochemistry (G.M., S.H., A.v.B., A.F.G., N.B., A.A.J.H., C.J.M.d.V., V.d.W.), Department of Molecular Cell Biology, Sanquin Research and Landsteiner Laboratory (M.H., J.D.v.B.), and Department of Cell Biology and Histology (J.S.), Academic Medical Center, University of Amsterdam, The Netherlands
| | - Stijntje Hibender
- From the Department of Medical Biochemistry (G.M., S.H., A.v.B., A.F.G., N.B., A.A.J.H., C.J.M.d.V., V.d.W.), Department of Molecular Cell Biology, Sanquin Research and Landsteiner Laboratory (M.H., J.D.v.B.), and Department of Cell Biology and Histology (J.S.), Academic Medical Center, University of Amsterdam, The Netherlands
| | - Mark Hoogenboezem
- From the Department of Medical Biochemistry (G.M., S.H., A.v.B., A.F.G., N.B., A.A.J.H., C.J.M.d.V., V.d.W.), Department of Molecular Cell Biology, Sanquin Research and Landsteiner Laboratory (M.H., J.D.v.B.), and Department of Cell Biology and Histology (J.S.), Academic Medical Center, University of Amsterdam, The Netherlands
| | - Amber van Broekhoven
- From the Department of Medical Biochemistry (G.M., S.H., A.v.B., A.F.G., N.B., A.A.J.H., C.J.M.d.V., V.d.W.), Department of Molecular Cell Biology, Sanquin Research and Landsteiner Laboratory (M.H., J.D.v.B.), and Department of Cell Biology and Histology (J.S.), Academic Medical Center, University of Amsterdam, The Netherlands
| | - Arginell F. Girigorie
- From the Department of Medical Biochemistry (G.M., S.H., A.v.B., A.F.G., N.B., A.A.J.H., C.J.M.d.V., V.d.W.), Department of Molecular Cell Biology, Sanquin Research and Landsteiner Laboratory (M.H., J.D.v.B.), and Department of Cell Biology and Histology (J.S.), Academic Medical Center, University of Amsterdam, The Netherlands
| | - Natascha Bleeker
- From the Department of Medical Biochemistry (G.M., S.H., A.v.B., A.F.G., N.B., A.A.J.H., C.J.M.d.V., V.d.W.), Department of Molecular Cell Biology, Sanquin Research and Landsteiner Laboratory (M.H., J.D.v.B.), and Department of Cell Biology and Histology (J.S.), Academic Medical Center, University of Amsterdam, The Netherlands
| | - Anouk A.J. Hamers
- From the Department of Medical Biochemistry (G.M., S.H., A.v.B., A.F.G., N.B., A.A.J.H., C.J.M.d.V., V.d.W.), Department of Molecular Cell Biology, Sanquin Research and Landsteiner Laboratory (M.H., J.D.v.B.), and Department of Cell Biology and Histology (J.S.), Academic Medical Center, University of Amsterdam, The Netherlands
| | - Jan Stap
- From the Department of Medical Biochemistry (G.M., S.H., A.v.B., A.F.G., N.B., A.A.J.H., C.J.M.d.V., V.d.W.), Department of Molecular Cell Biology, Sanquin Research and Landsteiner Laboratory (M.H., J.D.v.B.), and Department of Cell Biology and Histology (J.S.), Academic Medical Center, University of Amsterdam, The Netherlands
| | - Jaap D. van Buul
- From the Department of Medical Biochemistry (G.M., S.H., A.v.B., A.F.G., N.B., A.A.J.H., C.J.M.d.V., V.d.W.), Department of Molecular Cell Biology, Sanquin Research and Landsteiner Laboratory (M.H., J.D.v.B.), and Department of Cell Biology and Histology (J.S.), Academic Medical Center, University of Amsterdam, The Netherlands
| | - Carlie J.M. de Vries
- From the Department of Medical Biochemistry (G.M., S.H., A.v.B., A.F.G., N.B., A.A.J.H., C.J.M.d.V., V.d.W.), Department of Molecular Cell Biology, Sanquin Research and Landsteiner Laboratory (M.H., J.D.v.B.), and Department of Cell Biology and Histology (J.S.), Academic Medical Center, University of Amsterdam, The Netherlands
| | - Vivian de Waard
- From the Department of Medical Biochemistry (G.M., S.H., A.v.B., A.F.G., N.B., A.A.J.H., C.J.M.d.V., V.d.W.), Department of Molecular Cell Biology, Sanquin Research and Landsteiner Laboratory (M.H., J.D.v.B.), and Department of Cell Biology and Histology (J.S.), Academic Medical Center, University of Amsterdam, The Netherlands
| |
Collapse
|
50
|
Mieth A, Revermann M, Babelova A, Weigert A, Schermuly RT, Brandes RP. L-type calcium channel inhibitor diltiazem prevents aneurysm formation by blood pressure-independent anti-inflammatory effects. Hypertension 2013; 62:1098-104. [PMID: 24082061 DOI: 10.1161/hypertensionaha.113.01986] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Formation of abdominal aortic aneurysms is a progressive inflammatory process that involves infiltration and differentiation of monocytes in the vessel wall, proliferation and migration of smooth muscle cells, and eventually the degradation of the internal elastic lamina, which leads to outward vascular remodeling and distension of the vessel. Because calcium channel blockers exert multiple beneficial effects on the vascular system, we investigated the effect of the benzothiazepine-type calcium channel blocker diltiazem on aneurysm formation in a mouse model. Angiotensin II infusion induced massive suprarenal aortic aneurysm formation in male apolipoprotein E-deficient mice that was blocked by cotreatment with diltiazem even if the blood pressure was controlled by coinfusion of phenylephrine. Diltiazem prevented the angiotensin II-mediated induction of proinflammatory cytokines after 7 days of angiotensin II treatment in the aortic arch attributable to a reduction in the amount of locally infiltrating macrophages. To identify the underlying mechanism, vascular segments and cultured vascular cells as well as monocytes were studied. Diltiazem failed to reduce the angiotensin II-induced expression of proinflammatory chemokines and cytokines in isolated mouse thoracic aortic segments in organ culture. Furthermore, diltiazem did not affect the recruitment of proinflammatory Ly6C(+) monocytes in vivo pointing toward an effect of the compound on gene expression in monocytes/macrophages. Indeed, diltiazem prevented the interleukin-6-induced mRNA expression of interleukin-1β and the monocyte chemoattractant protein CCL12 in peritoneal macrophages and RAW264.7 cells independent of the intracellular calcium concentration. Thus, diltiazem limits aortic aneurysm formation in mice by a blood pressure-independent anti-inflammatory effect on monocytic cells.
Collapse
Affiliation(s)
- Anja Mieth
- Institut für Kardiovaskuläre Physiologie, Fachbereich Medizin der Goethe-Universität, Theodor-Stern-Kai 7, D-60590 Frankfurt am Main, Germany.
| | | | | | | | | | | |
Collapse
|