1
|
Zhang ZB, Cheng YW, Xu L, Li JQ, Pan X, Zhu M, Chen XH, Sun AJ, Lin JR, Gao PJ. Activation of β3-adrenergic receptor by mirabegron prevents aortic dissection/aneurysm by promoting lymphangiogenesis in perivascular adipose tissue. Cardiovasc Res 2024; 120:2307-2319. [PMID: 39288197 DOI: 10.1093/cvr/cvae213] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Revised: 05/06/2024] [Accepted: 07/23/2024] [Indexed: 09/19/2024] Open
Abstract
AIMS β3-Adrenergic receptor (β3-AR) is essential for cardiovascular homoeostasis through regulating adipose tissue function. Perivascular adipose tissue (PVAT) has been implicated in the pathogenesis of aortic dissection and aneurysm (AD/AA). Here, we aim to investigate β3-AR activation-mediated PVAT function in AD/AA. METHODS AND RESULTS Aortas from patients with thoracic aortic dissection (TAD) were collected to detect β3-AR expression in PVAT. ApoE-/- and β-aminopropionitrile monofumarate (BAPN)-treated C57BL/6 mice were induced with Angiotensin II (AngII) to simulate AD/AA and subsequently received either placebo or mirabegron, a β3-AR agonist. The results demonstrated an up-regulation of β3-AR in PVAT of TAD patients and AD/AA mice. Moreover, activation of β3-AR by mirabegron significantly prevented AngII-induced AD/AA formation in mice. RNA-sequencing analysis of adipocytes from PVAT revealed a notable increase of the lymphangiogenic factor, vascular endothelial growth factor C (VEGF-C), in mirabegron-treated mice. Consistently, enhanced lymphangiogenesis was found in PVAT with mirabegron treatment. Mechanistically, the number of CD4+/CD8+ T cells and CD11c+ cells was reduced in PVAT but increased in adjacent draining lymph nodes of mirabegron-treated mice, indicating the improved draining and clearance of inflammatory cells in PVAT by lymphangiogenesis. Importantly, adipocyte-specific VEGF-C knockdown by the adeno-associated virus system restrained lymphangiogenesis and exacerbated inflammatory cell infiltration in PVAT, which ultimately abolished the protection of mirabegron on AD/AA. In addition, the conditional medium derived from mirabegron-treated adipocytes activated the proliferation and tube formation of LECs, which was abrogated by the silencing of VEGF-C in adipocytes. CONCLUSION Our findings illustrated the therapeutic potential of β3-AR activation by mirabegron on AD/AA, which promoted lymphangiogenesis by increasing adipocyte-derived VEGF-C and, therefore, ameliorated PVAT inflammation.
Collapse
MESH Headings
- Animals
- Aortic Dissection/metabolism
- Aortic Dissection/pathology
- Aortic Dissection/prevention & control
- Aortic Dissection/chemically induced
- Aortic Dissection/physiopathology
- Aortic Dissection/genetics
- Lymphangiogenesis/drug effects
- Adrenergic beta-3 Receptor Agonists/pharmacology
- Mice, Inbred C57BL
- Receptors, Adrenergic, beta-3/metabolism
- Receptors, Adrenergic, beta-3/genetics
- Acetanilides/pharmacology
- Vascular Endothelial Growth Factor C/metabolism
- Vascular Endothelial Growth Factor C/genetics
- Humans
- Male
- Disease Models, Animal
- Adipose Tissue/metabolism
- Adipose Tissue/drug effects
- Adipose Tissue/pathology
- Aortic Aneurysm, Thoracic/metabolism
- Aortic Aneurysm, Thoracic/pathology
- Aortic Aneurysm, Thoracic/prevention & control
- Aortic Aneurysm, Thoracic/physiopathology
- Aortic Aneurysm, Thoracic/genetics
- Aortic Aneurysm, Thoracic/chemically induced
- Aortic Aneurysm, Thoracic/drug therapy
- Mice, Knockout, ApoE
- Thiazoles/pharmacology
- Signal Transduction
- Aorta, Thoracic/metabolism
- Aorta, Thoracic/pathology
- Aorta, Thoracic/physiopathology
- Aorta, Thoracic/drug effects
- Angiotensin II
- Adipocytes/metabolism
- Adipocytes/pathology
- Adipocytes/drug effects
- Cells, Cultured
- Mice
- Lymphatic Vessels/metabolism
- Lymphatic Vessels/drug effects
- Lymphatic Vessels/pathology
- Lymphatic Vessels/physiopathology
- Female
- Middle Aged
Collapse
Affiliation(s)
- Ze-Bei Zhang
- Department of Cardiovascular Medicine, State Key Laboratory of Medical Genomics, Shanghai Key Laboratory of Hypertension, Shanghai Institute of Hypertension, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin, 2 Road, Shanghai 200025, China
| | - Yu-Wen Cheng
- Department of Cardiovascular Medicine, State Key Laboratory of Medical Genomics, Shanghai Key Laboratory of Hypertension, Shanghai Institute of Hypertension, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin, 2 Road, Shanghai 200025, China
- Department of Cardiology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Lian Xu
- Department of Cardiovascular Medicine, State Key Laboratory of Medical Genomics, Shanghai Key Laboratory of Hypertension, Shanghai Institute of Hypertension, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin, 2 Road, Shanghai 200025, China
| | - Jia-Qi Li
- Department of Cardiovascular Medicine, State Key Laboratory of Medical Genomics, Shanghai Key Laboratory of Hypertension, Shanghai Institute of Hypertension, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin, 2 Road, Shanghai 200025, China
| | - Xin Pan
- Department of Gerontology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Min Zhu
- Department of Cardiovascular Medicine, State Key Laboratory of Medical Genomics, Shanghai Key Laboratory of Hypertension, Shanghai Institute of Hypertension, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin, 2 Road, Shanghai 200025, China
| | - Xiao-Hui Chen
- Department of Cardiovascular Medicine, State Key Laboratory of Medical Genomics, Shanghai Key Laboratory of Hypertension, Shanghai Institute of Hypertension, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin, 2 Road, Shanghai 200025, China
| | - Ai-Jun Sun
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, Shanghai, China
| | - Jing-Rong Lin
- Department of Cardiovascular Medicine, State Key Laboratory of Medical Genomics, Shanghai Key Laboratory of Hypertension, Shanghai Institute of Hypertension, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin, 2 Road, Shanghai 200025, China
| | - Ping-Jin Gao
- Department of Cardiovascular Medicine, State Key Laboratory of Medical Genomics, Shanghai Key Laboratory of Hypertension, Shanghai Institute of Hypertension, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin, 2 Road, Shanghai 200025, China
| |
Collapse
|
2
|
Stougiannou TM, Christodoulou KC, Karangelis D. Olfactory Receptors and Aortic Aneurysm: Review of Disease Pathways. J Clin Med 2024; 13:7778. [PMID: 39768700 PMCID: PMC11727755 DOI: 10.3390/jcm13247778] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2024] [Revised: 12/10/2024] [Accepted: 12/17/2024] [Indexed: 01/16/2025] Open
Abstract
Aortic aneurysm, the pathological dilatation of the aorta at distinct locations, can be attributed to many different genetic and environmental factors. The resulting pathobiological disturbances generate a complex interplay of processes affecting cells and extracellular molecules of the tunica interna, media and externa. In short, aortic aneurysm can affect processes involving the extracellular matrix, lipid trafficking/atherosclerosis, vascular smooth muscle cells, inflammation, platelets and intraluminal thrombus formation, as well as various endothelial functions. Many of these processes are interconnected, potentiating one another. Newer discoveries, including the involvement of odorant olfactory receptors in these processes, have further shed light on disease initiation and pathology. Olfactory receptors are a varied group of G protein coupled-receptors responsible for the recognition of chemosensory information. Although they comprise many different subgroups, some of which are not well-characterized or identified in humans, odorant olfactory receptors, in particular, are most commonly associated with recognition of olfactory information. They can also be ectopically localized and thus carry out additional functions relevant to the tissue in which they are identified. It is thus the purpose of this narrative review to summarize and present pathobiological processes relevant to the initiation and propagation of aortic aneurysm, while also incorporating evidence associating these ectopically functioning odorant olfactory receptors with the overall pathology.
Collapse
Affiliation(s)
- Theodora M. Stougiannou
- Department of Cardiothoracic Surgery, University General Hospital, Democritus University of Thrace, 68100 Alexandroupolis, Greece; (K.C.C.); (D.K.)
| | | | | |
Collapse
|
3
|
Torimoto K, Vari HR, Nakayama Y, Utsunomiya H, Takeda M, Hashimoto T, Rizzo V, Eguchi S. Glucose transporter 1 in vascular smooth muscle cells is dispensable for abdominal aortic aneurysm induced by angiotensin II. JVS Vasc Sci 2024; 6:100270. [PMID: 39811041 PMCID: PMC11728061 DOI: 10.1016/j.jvssci.2024.100270] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2024] [Accepted: 11/22/2024] [Indexed: 01/16/2025] Open
Abstract
Treatment with an inhibitor of glucose use via glucose transporters (GLUT) has been shown to attenuate experimental abdominal aortic aneurysm (AAA) development in mice. Vascular smooth muscle cell (VSMC) signaling seems to be essential for angiotensin II (Ang II)-induced AAA in mice. Accordingly, we have tested a hypothesis that VSMC silencing of the major GLUT, GLUT1, prevents AAA development and rupture in mice treated with Ang II plus β-aminopropionitrile. A mouse model of inducible VSMC GLUT1 deletion was created and aortic GLUT1 silencing was confirmed. Without Ang II plus β-aminopropionitrile treatment, no difference was observed regarding the external aortic diameter (control 1.06 ± 0.18 mm vs deletion 0.97 ± 0.26 mm) or systolic blood pressure (control 102 ± 9 mm Hg vs deletion 107 ± 11 mm Hg) between control or GLUT1-silenced mice. With treatment, control mice as well as VSMC GLUT1-silenced mice equally developed AAA (control 2.37 ± 0.75 mm vs deletion 2.41 ± 0.93 mm), whereas a tendency toward lower blood pressure was observed in GLUT1 silenced mice (control 150 ± 9 mm Hg vs deletion 135 ± 22 mm Hg). No significant difference was observed regarding the rate of rupture-dependent mortality. We concluded that VSMC GLUT1 is dispensable for AAA development induced by Ang II in mice.
Collapse
Affiliation(s)
- Keiichi Torimoto
- Department of Cardiovascular Science, Lewis Katz School of Medicine at Temple University, Philadelphia, PA
| | - Hymavathi Reddy Vari
- Department of Cardiovascular Science, Lewis Katz School of Medicine at Temple University, Philadelphia, PA
| | - Yuki Nakayama
- Department of Cardiovascular Science, Lewis Katz School of Medicine at Temple University, Philadelphia, PA
- Department of Rehabilitation, Osaka Kawasaki Rehabilitation University, Osaka, Japan
| | - Hirotoshi Utsunomiya
- Department of Rehabilitation, Osaka Kawasaki Rehabilitation University, Osaka, Japan
| | - Masatoshi Takeda
- Department of Rehabilitation, Osaka Kawasaki Rehabilitation University, Osaka, Japan
| | - Tomoki Hashimoto
- Department of Neurosurgery and Neurobiology, Barrow Aneurysm and AVM Research Center, Barrow Neurological Institute Phoenix, Pheonix, AZ
| | - Victor Rizzo
- Department of Cardiovascular Science, Lewis Katz School of Medicine at Temple University, Philadelphia, PA
| | - Satoru Eguchi
- Department of Cardiovascular Science, Lewis Katz School of Medicine at Temple University, Philadelphia, PA
| |
Collapse
|
4
|
Sharma N, Khalyfa A, Cai D, Morales-Quinones M, Soares RN, Higashi Y, Chen S, Gozal D, Padilla J, Manrique-Acevedo C, Chandrasekar B, Martinez-Lemus LA. Chronic intermittent hypoxia facilitates the development of angiotensin II-induced abdominal aortic aneurysm in male mice. J Appl Physiol (1985) 2024; 137:527-539. [PMID: 38867666 PMCID: PMC11424178 DOI: 10.1152/japplphysiol.00842.2023] [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: 11/22/2023] [Revised: 04/11/2024] [Accepted: 06/05/2024] [Indexed: 06/14/2024] Open
Abstract
Obstructive sleep apnea (OSA), characterized by episodes of intermittent hypoxia (IH), is highly prevalent in patients with abdominal aortic aneurysm (AAA). However, whether IH serves as an independent risk factor for AAA development remains to be investigated. Here, we determined the effects of chronic (6 mo) IH on angiotensin (Ang II)-induced AAA development in C57BL/6J male mice and investigated the underlying mechanisms of IH in cultured vascular smooth muscle cells (SMCs). IH increased the susceptibility of mice to develop AAA in response to Ang II infusion by facilitating the augmentation of the abdominal aorta's diameter as assessed by transabdominal ultrasound imaging. Importantly, IH with Ang II augmented aortic elastin degradation and the expression of matrix metalloproteinases (MMPs), mainly MMP8, MMP12, and a disintegrin and metalloproteinase-17 (ADAM17) as measured by histology and immunohistochemistry. Mechanistically, IH increased the activities of MMP2, MMP8, MMP9, MMP12, and ADAM17, while reducing the expression of the MMP regulator reversion-inducing cysteine-rich protein with Kazal motifs (RECK) in cultured SMCs. Aortic samples from human AAA were associated with decreased RECK and increased expression of ADAM17 and MMPs. These data suggest that IH facilitates AAA development when additional stressors are superimposed and that this occurs in association with an increased presence of aortic MMPs and ADAM17, potentially due to IH-induced modulation of RECK expression. These findings support a plausible synergistic link between OSA and AAA and provide a better understanding of the molecular mechanisms underlying the pathogenesis of AAA.NEW & NOTEWORTHY IH facilitates Ang II-induced abdominal aortic diameter expansion and AAA development in C57BL/6J male mice. IH upregulates the expression of specific MMPs such as MMP8, MMP12, and ADAM17. IH directly suppresses RECK expression and increases MMPs activity in SMCs. Human AAA tissues exhibit a downregulation of RECK and an upregulation of ADAM17 and MMPs.
Collapse
MESH Headings
- Aortic Aneurysm, Abdominal/metabolism
- Aortic Aneurysm, Abdominal/chemically induced
- Aortic Aneurysm, Abdominal/pathology
- Animals
- Male
- Angiotensin II
- Mice, Inbred C57BL
- Hypoxia/metabolism
- Hypoxia/complications
- Mice
- ADAM17 Protein/metabolism
- Aorta, Abdominal/metabolism
- Aorta, Abdominal/pathology
- Myocytes, Smooth Muscle/metabolism
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/pathology
- Humans
- Matrix Metalloproteinases/metabolism
- Matrix Metalloproteinase 12/metabolism
- Sleep Apnea, Obstructive/metabolism
- Sleep Apnea, Obstructive/physiopathology
- Sleep Apnea, Obstructive/complications
Collapse
Affiliation(s)
- Neekun Sharma
- NextGen Precision Health, University of Missouri, Columbia, Missouri, United States
- Department of Medicine, Center for Precision Medicine, University of Missouri, Columbia, Missouri, United States
- Division of Endocrinology and Metabolism, Department of Medicine, University of Missouri, Columbia, Missouri, United States
| | - Abdelnaby Khalyfa
- Department of Child Health and the Child Health Research Institute, School of Medicine, University of Missouri, Columbia, Missouri, United States
| | - Dunpeng Cai
- Department of Surgery, University of Missouri, Columbia, Missouri, United States
| | | | - Rogerio N Soares
- NextGen Precision Health, University of Missouri, Columbia, Missouri, United States
| | - Yusuke Higashi
- John W. Deming Department of Medicine, Tulane University, New Orleans, Louisiana, United States
| | - Shiyou Chen
- Department of Surgery, University of Missouri, Columbia, Missouri, United States
- Harry S. Truman Memorial Veterans' Hospital, Columbia, Missouri, United States
| | - David Gozal
- Department of Child Health and the Child Health Research Institute, School of Medicine, University of Missouri, Columbia, Missouri, United States
| | - Jaume Padilla
- NextGen Precision Health, University of Missouri, Columbia, Missouri, United States
- Harry S. Truman Memorial Veterans' Hospital, Columbia, Missouri, United States
- Department of Nutrition and Exercise Physiology, University of Missouri, Columbia, Missouri, United States
| | - Camila Manrique-Acevedo
- NextGen Precision Health, University of Missouri, Columbia, Missouri, United States
- Division of Endocrinology and Metabolism, Department of Medicine, University of Missouri, Columbia, Missouri, United States
- Harry S. Truman Memorial Veterans' Hospital, Columbia, Missouri, United States
| | - Bysani Chandrasekar
- Harry S. Truman Memorial Veterans' Hospital, Columbia, Missouri, United States
- Division of Cadiovascular Medicine, Department of Medicine, University of Missouri, Columbia, Missouri, United States
| | - Luis A Martinez-Lemus
- NextGen Precision Health, University of Missouri, Columbia, Missouri, United States
- Department of Medicine, Center for Precision Medicine, University of Missouri, Columbia, Missouri, United States
- Department of Medical Pharmacology and Physiology, University of Missouri, Columbia, Missouri, United States
| |
Collapse
|
5
|
Guo X, Zhong J, Zhao Y, Fu Y, Sun LY, Yuan A, Liu J, Chen AF, Pu J. LXRα Promotes Abdominal Aortic Aneurysm Formation Through UHRF1 Epigenetic Modification of miR-26b-3p. Circulation 2024; 150:30-46. [PMID: 38557060 PMCID: PMC11219073 DOI: 10.1161/circulationaha.123.065202] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Accepted: 03/11/2024] [Indexed: 04/04/2024]
Abstract
BACKGROUND Abdominal aortic aneurysm (AAA) is a severe aortic disease without effective pharmacological approaches. The nuclear hormone receptor LXRα (liver X receptor α), encoded by the NR1H3 gene, serves as a critical transcriptional mediator linked to several vascular pathologies, but its role in AAA remains elusive. METHODS Through integrated analyses of human and murine AAA gene expression microarray data sets, we identified NR1H3 as a candidate gene regulating AAA formation. To investigate the role of LXRα in AAA formation, we used global Nr1h3-knockout and vascular smooth muscle cell-specific Nr1h3-knockout mice in 2 AAA mouse models induced with angiotensin II (1000 ng·kg·min; 28 days) or calcium chloride (CaCl2; 0.5 mol/L; 42 days). RESULTS Upregulated LXRα was observed in the aortas of patients with AAA and in angiotensin II- or CaCl2-treated mice. Global or vascular smooth muscle cell-specific Nr1h3 knockout inhibited AAA formation in 2 mouse models. Loss of LXRα function prevented extracellular matrix degeneration, inflammation, and vascular smooth muscle cell phenotypic switching. Uhrf1, an epigenetic master regulator, was identified as a direct target gene of LXRα by integrated analysis of transcriptome sequencing and chromatin immunoprecipitation sequencing. Susceptibility to AAA development was consistently enhanced by UHRF1 (ubiquitin-like containing PHD and RING finger domains 1) in both angiotensin II- and CaCl2-induced mouse models. We then determined the CpG methylation status and promoter accessibility of UHRF1-mediated genes using CUT&Tag (cleavage under targets and tagmentation), RRBS (reduced representation bisulfite sequencing), and ATAC-seq (assay for transposase-accessible chromatin with sequencing) in vascular smooth muscle cells, which revealed that the recruitment of UHRF1 to the promoter of miR-26b led to DNA hypermethylation accompanied by relatively closed chromatin states, and caused downregulation of miR-26b expression in AAA. Regarding clinical significance, we found that underexpression of miR-26b-3p correlated with high risk in patients with AAA. Maintaining miR-26b-3p expression prevented AAA progression and alleviated the overall pathological process. CONCLUSIONS Our study reveals a pivotal role of the LXRα/UHRF1/miR-26b-3p axis in AAA and provides potential biomarkers and therapeutic targets for AAA.
Collapse
MESH Headings
- Aortic Aneurysm, Abdominal/genetics
- Aortic Aneurysm, Abdominal/metabolism
- Aortic Aneurysm, Abdominal/pathology
- Aortic Aneurysm, Abdominal/chemically induced
- Animals
- Liver X Receptors/metabolism
- Liver X Receptors/genetics
- Epigenesis, Genetic
- MicroRNAs/genetics
- MicroRNAs/metabolism
- Humans
- CCAAT-Enhancer-Binding Proteins/genetics
- CCAAT-Enhancer-Binding Proteins/metabolism
- Mice
- Mice, Knockout
- Ubiquitin-Protein Ligases/genetics
- Ubiquitin-Protein Ligases/metabolism
- Male
- Disease Models, Animal
- Mice, Inbred C57BL
- DNA Methylation
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/pathology
- Angiotensin II/pharmacology
Collapse
Affiliation(s)
- Xiao Guo
- Department of Cardiology, State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital (X.G., J.Z., Y.Z., Y.F., L.-y.S., A.Y., J.L., J.P.), Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jianmei Zhong
- Department of Cardiology, State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital (X.G., J.Z., Y.Z., Y.F., L.-y.S., A.Y., J.L., J.P.), Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yichao Zhao
- Department of Cardiology, State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital (X.G., J.Z., Y.Z., Y.F., L.-y.S., A.Y., J.L., J.P.), Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yanan Fu
- Department of Cardiology, State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital (X.G., J.Z., Y.Z., Y.F., L.-y.S., A.Y., J.L., J.P.), Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ling-yue Sun
- Department of Cardiology, State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital (X.G., J.Z., Y.Z., Y.F., L.-y.S., A.Y., J.L., J.P.), Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ancai Yuan
- Department of Cardiology, State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital (X.G., J.Z., Y.Z., Y.F., L.-y.S., A.Y., J.L., J.P.), Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Junling Liu
- Department of Cardiology, State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital (X.G., J.Z., Y.Z., Y.F., L.-y.S., A.Y., J.L., J.P.), Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Department of Biochemistry and Molecular Cell Biology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education (J.L.), Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Alex F. Chen
- Institute for Developmental and Regenerative Cardiovascular Medicine, Xinhua Hospital (A.F.C.), Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jun Pu
- Department of Cardiology, State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital (X.G., J.Z., Y.Z., Y.F., L.-y.S., A.Y., J.L., J.P.), Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
6
|
Franklin MK, Sawada H, Ito S, Howatt DA, Amioka N, Liang CL, Zhang N, Graf DB, Moorleghen JJ, Katsumata Y, Lu HS, Daugherty A. β-Aminopropionitrile Induces Distinct Pathologies in the Ascending and Descending Thoracic Aortic Regions of Mice. Arterioscler Thromb Vasc Biol 2024; 44:1555-1569. [PMID: 38779856 PMCID: PMC11209774 DOI: 10.1161/atvbaha.123.320402] [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: 11/07/2023] [Accepted: 05/08/2024] [Indexed: 05/25/2024]
Abstract
BACKGROUND β-aminopropionitrile (BAPN) is a pharmacological inhibitor of LOX (lysyl oxidase) and LOXLs (LOX-like proteins). Administration of BAPN promotes aortopathies, although there is a paucity of data on experimental conditions to generate pathology. The objective of this study was to define experimental parameters and determine whether equivalent or variable aortopathies were generated throughout the aortic tree during BAPN administration in mice. METHODS BAPN was administered in drinking water for a period ranging from 1 to 12 weeks. The impacts of BAPN were first assessed with regard to BAPN dose, and mouse strain, age, and sex. BAPN-induced aortic pathological characterization was conducted using histology and immunostaining. To investigate the mechanistic basis of regional heterogeneity, the ascending and descending thoracic aortas were harvested after 1 week of BAPN administration before the appearance of overt pathology. RESULTS BAPN-induced aortic rupture predominantly occurred or originated in the descending thoracic aorta in young C57BL/6J or N mice. No apparent differences were found between male and female mice. For mice surviving 12 weeks of BAPN administration, profound dilatation was consistently observed in the ascending region, while there were more heterogeneous changes in the descending thoracic region. Pathological features were distinct between the ascending and descending thoracic regions. Aortic pathology in the ascending region was characterized by luminal dilatation and elastic fiber disruption throughout the media. The descending thoracic region frequently had dissections with false lumen formation, collagen deposition, and remodeling of the wall surrounding the false lumen. Cells surrounding the false lumen were predominantly positive for α-SMA (α-smooth muscle actin). One week of BAPN administration compromised contractile properties in both regions equivalently, and RNA sequencing did not show obvious differences between the 2 aortic regions in smooth muscle cell markers, cell proliferation markers, and extracellular components. CONCLUSIONS BAPN-induced pathologies show distinct, heterogeneous features within and between ascending and descending aortic regions in mice.
Collapse
MESH Headings
- Animals
- Aminopropionitrile/toxicity
- Aminopropionitrile/pharmacology
- Aorta, Thoracic/pathology
- Aorta, Thoracic/drug effects
- Aorta, Thoracic/metabolism
- Female
- Male
- Mice, Inbred C57BL
- Disease Models, Animal
- Aortic Rupture/chemically induced
- Aortic Rupture/pathology
- Aortic Rupture/metabolism
- Aortic Rupture/prevention & control
- Mice
- Vascular Remodeling/drug effects
- Dilatation, Pathologic
- Muscle, Smooth, Vascular/pathology
- Muscle, Smooth, Vascular/drug effects
- Muscle, Smooth, Vascular/metabolism
- Age Factors
- Time Factors
- Sex Factors
- Cell Proliferation/drug effects
- Protein-Lysine 6-Oxidase/metabolism
Collapse
Affiliation(s)
| | - Hisashi Sawada
- Saha Cardiovascular Research Center, University of Kentucky, Lexington, KY
- Saha Aortic Center, University of Kentucky, Lexington, KY
- Department of Physiology, University of Kentucky, Lexington, KY
| | - Sohei Ito
- Saha Cardiovascular Research Center, University of Kentucky, Lexington, KY
| | - Deborah A. Howatt
- Saha Cardiovascular Research Center, University of Kentucky, Lexington, KY
| | - Naofumi Amioka
- Saha Cardiovascular Research Center, University of Kentucky, Lexington, KY
| | - Ching-Ling Liang
- Saha Cardiovascular Research Center, University of Kentucky, Lexington, KY
| | - Nancy Zhang
- Saha Cardiovascular Research Center, University of Kentucky, Lexington, KY
| | - David B. Graf
- Saha Cardiovascular Research Center, University of Kentucky, Lexington, KY
| | | | - Yuriko Katsumata
- Department of Biostatistics, College of Public Health, University of Kentucky, Lexington, KY
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY
| | - Hong S. Lu
- Saha Cardiovascular Research Center, University of Kentucky, Lexington, KY
- Saha Aortic Center, University of Kentucky, Lexington, KY
- Department of Physiology, University of Kentucky, Lexington, KY
| | - Alan Daugherty
- Saha Cardiovascular Research Center, University of Kentucky, Lexington, KY
- Saha Aortic Center, University of Kentucky, Lexington, KY
- Department of Physiology, University of Kentucky, Lexington, KY
| |
Collapse
|
7
|
Franklin MK, Sawada H, Ito S, Howatt DA, Amioka N, Liang CL, Zhang N, Graf DB, Moorleghen JJ, Katsumata Y, Lu HS, Daugherty A. β-aminopropionitrile Induces Distinct Pathologies in the Ascending and Descending Thoracic Aortic Regions of Mice. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.10.22.563474. [PMID: 37886537 PMCID: PMC10602045 DOI: 10.1101/2023.10.22.563474] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/28/2023]
Abstract
BACKGROUND β-aminopropionitrile (BAPN) is a pharmacological inhibitor of lysyl oxidase and lysyl oxidase-like proteins. Administration of BAPN promotes aortopathies, although there is a paucity of data on experimental conditions to generate pathology. The objective of this study was to define experimental parameters and determine whether equivalent or variable aortopathies were generated throughout the aortic tree during BAPN administration in mice. METHODS BAPN was administered in drinking water for a period ranging from 1 to 12 weeks. The impacts of BAPN were first assessed with regard to dose, strain, age, and sex. BAPN-induced aortic pathological characterization was conducted using histology and immunostaining. To investigate the mechanistic basis of regional heterogeneity, ascending and descending thoracic aortas were harvested after one week of BAPN administration before the appearance of overt pathology. RESULTS BAPN-induced aortic rupture predominantly occurred or originated in the descending thoracic aorta in young C57BL/6J or N mice. No apparent differences were found between male and female mice. For mice surviving 12 weeks of BAPN administration, profound dilatation was consistently observed in the ascending region, while there were more heterogeneous changes in the descending thoracic region. Pathological features were distinct between the ascending and descending thoracic regions. Aortic pathology in the ascending region was characterized by luminal dilatation and elastic fiber disruption throughout the media. The descending thoracic region frequently had dissections with false lumen formation, collagen deposition, and remodeling of the wall surrounding the false lumen. Cells surrounding the false lumen were predominantly positive for α-smooth muscle actin. One week of BAPN administration compromised contractile properties in both regions equivalently, and RNA sequencing did not show obvious differences between the two aortic regions in smooth muscle cell markers, cell proliferation markers, and extracellular components. CONCLUSIONS BAPN-induced pathologies show distinct, heterogeneous features within and between ascending and descending aortic regions in mice.
Collapse
Affiliation(s)
| | - Hisashi Sawada
- Saha Cardiovascular Research Center, University of Kentucky, Lexington, KY
- Saha Aortic Center, University of Kentucky, Lexington, KY
- Department of Physiology, University of Kentucky, Lexington, KY
| | - Sohei Ito
- Saha Cardiovascular Research Center, University of Kentucky, Lexington, KY
| | - Deborah A. Howatt
- Saha Cardiovascular Research Center, University of Kentucky, Lexington, KY
| | - Naofumi Amioka
- Saha Cardiovascular Research Center, University of Kentucky, Lexington, KY
| | - Ching-Ling Liang
- Saha Cardiovascular Research Center, University of Kentucky, Lexington, KY
| | - Nancy Zhang
- Saha Cardiovascular Research Center, University of Kentucky, Lexington, KY
| | - David B. Graf
- Saha Cardiovascular Research Center, University of Kentucky, Lexington, KY
| | | | - Yuriko Katsumata
- Department of Biostatistics, College of Public Health, University of Kentucky, Lexington, KY
- Sanders-Brown Center on Aging University of Kentucky, Lexington, KY
| | - Hong S. Lu
- Saha Cardiovascular Research Center, University of Kentucky, Lexington, KY
- Saha Aortic Center, University of Kentucky, Lexington, KY
- Department of Physiology, University of Kentucky, Lexington, KY
| | - Alan Daugherty
- Saha Cardiovascular Research Center, University of Kentucky, Lexington, KY
- Saha Aortic Center, University of Kentucky, Lexington, KY
- Department of Physiology, University of Kentucky, Lexington, KY
| |
Collapse
|
8
|
Eguchi S, Sparks MA, Sawada H, Lu HS, Daugherty A, Zhuo JL. Recent Advances in Understanding the Molecular Pathophysiology of Angiotensin II Receptors: Lessons From Cell-Selective Receptor Deletion in Mice. Can J Cardiol 2023; 39:1795-1807. [PMID: 37394059 DOI: 10.1016/j.cjca.2023.06.421] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Revised: 06/08/2023] [Accepted: 06/20/2023] [Indexed: 07/04/2023] Open
Abstract
The renin-angiotensin system (RAS) is an essential hormonal system involved in water and sodium reabsorption, renal blood flow regulation, and arterial constriction. Systemic stimulation of the RAS with infusion of the main peptide angiotensin II (Ang II) in animals as well as pathological elevation of renin (ie, renovascular hypertension) to increase circulatory Ang II in humans ultimately lead to hypertension and end organ damage. In addition to hypertension, accumulating evidence supports that the Ang II type 1 receptor exerts a critical role in cardiovascular and kidney diseases independent of blood pressure elevation. In the past 2 decades, the identification of an increased number of peptides and receptors has facilitated the concept that the RAS has detrimental and beneficial effects on the cardiovascular system depending on which RAS components are activated. For example, angiotensin 1-7 and Ang II type 2 receptors act as a counter-regulatory system against the classical RAS by mediating vasodilation. Although the RAS as an endocrine system for regulation of blood pressure is well established, there remain many unanswered questions and controversial findings regarding blood pressure regulation and pathophysiological regulation of cardiovascular diseases at the tissue level. This review article includes the latest knowledge gleaned from cell type-selective gene deleted mice regarding cell type-specific roles of Ang II receptors and their significance in health and diseases are discussed. In particular, we focus on the roles of these receptors expressed in vascular, cardiac, and kidney epithelial cells.
Collapse
Affiliation(s)
- Satoru Eguchi
- Sol Sherry Thrombosis Research Center, Lewis Katz School of Medicine at Temple University, Philadelphia, Pennsylvania, USA.
| | - Matthew A Sparks
- Division of Nephrology, Department of Medicine, Duke University School of Medicine, Durham, North Carolina, USA; Renal Section, Durham VA Medical Center, Durham, North Carolina, USA
| | - Hisashi Sawada
- Department of Physiology, Saha Cardiovascular Center, and Saha Aortic Center, University of Kentucky College of Medicine, Lexington, Kentucky, USA
| | - Hong S Lu
- Department of Physiology, Saha Cardiovascular Center, and Saha Aortic Center, University of Kentucky College of Medicine, Lexington, Kentucky, USA
| | - Alan Daugherty
- Department of Physiology, Saha Cardiovascular Center, and Saha Aortic Center, University of Kentucky College of Medicine, Lexington, Kentucky, USA
| | - Jia L Zhuo
- Tulane Hypertension and Renal Center of Excellence and Department of Physiology, Tulane University School of Medicine, New Orleans, Louisiana, USA
| |
Collapse
|
9
|
Okuno K, Torimoto K, Cicalese SM, Hashimoto T, Sparks MA, Rizzo V, Eguchi S. Smooth muscle angiotensin II type 1A receptor is required for abdominal aortic aneurysm formation induced by angiotensin II plus β-aminopropionitrile. J Mol Cell Cardiol 2023; 176:55-57. [PMID: 36736688 DOI: 10.1016/j.yjmcc.2023.01.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/03/2023] [Revised: 01/26/2023] [Accepted: 01/27/2023] [Indexed: 02/05/2023]
Affiliation(s)
- Keisuke Okuno
- Cardiovascular Research Center, Lewis Katz School of Medicine at Temple University, 3500 N. Broad Street, Philadelphia, PA 19140, United States of America
| | - Keiichi Torimoto
- Cardiovascular Research Center, Lewis Katz School of Medicine at Temple University, 3500 N. Broad Street, Philadelphia, PA 19140, United States of America
| | - Stephanie M Cicalese
- Cardiovascular Research Center, Lewis Katz School of Medicine at Temple University, 3500 N. Broad Street, Philadelphia, PA 19140, United States of America
| | - Tomoki Hashimoto
- Barrow Aneurysm and AVM Research Center, Departments of Neurosurgery and Neurobiology, Barrow Neurological Institute, 350 W. Thomas Rd, Phoenix, AZ 85013, United States of America
| | - Matthew A Sparks
- Department of Medicine, Division of Nephrology, Duke University Medical Center, 2 Genome Ct, Durham, NC 27710, United States of America
| | - Victor Rizzo
- Cardiovascular Research Center, Lewis Katz School of Medicine at Temple University, 3500 N. Broad Street, Philadelphia, PA 19140, United States of America.
| | - Satoru Eguchi
- Cardiovascular Research Center, Lewis Katz School of Medicine at Temple University, 3500 N. Broad Street, Philadelphia, PA 19140, United States of America.
| |
Collapse
|
10
|
Ming Y, Zhou X, Liu G, Abudupataer M, Zhu S, Xiang B, Yin X, Lai H, Sun Y, Wang C, Li J, Zhu K. PM2.5 exposure exacerbates mice thoracic aortic aneurysm and dissection by inducing smooth muscle cell apoptosis via the MAPK pathway. CHEMOSPHERE 2023; 313:137500. [PMID: 36495979 DOI: 10.1016/j.chemosphere.2022.137500] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Revised: 11/18/2022] [Accepted: 12/05/2022] [Indexed: 06/17/2023]
Abstract
Air pollution is a major public health concern worldwide. Exposure to fine particulate matter (PM2.5) is closely associated with cardiovascular diseases. However, the effect of PM2.5 exposure on thoracic aortic aneurysm and dissection (TAAD) has not been fully elucidated. Diesel exhaust particulate (DEP) is an important component of PM2.5, which causes health effects and is closely related to the incidence of cardiovascular disease. In the current study, we found that DEP exposure increased the incidence of aortic dissection (AD) in β-aminopropionitrile (BAPN)-induced thoracic aortic aneurysm (TAA). In addition, exposure to PM2.5 increased the diameter of the thoracic aorta in mice models. The number of apoptotic cells increased in the aortic wall of PM2.5-treated mice, as did the protein expression level of BAX/Bcl2 and cleaved caspase3/caspase3. Using a rhythmically stretching aortic mechanical simulation model, fluorescent staining indicated that PM2.5 administration could induce mitochondrial dysfunction and increase reactive oxygen species (ROS) levels in human aortic smooth muscle cells (HASMCs). Furthermore, ERK1/2 mitogen-activated protein kinase (MAPK) signaling pathways participated in the apoptosis of HASMCs after PM2.5 exposure. Therefore, we concluded that PM2.5 exposure could exacerbate the progression of TAAD, which could be induced by the increased apoptosis in HASMCs through the ERK1/2 MAPK signaling pathway.
Collapse
Affiliation(s)
- Yang Ming
- Department of Cardiac Surgery, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, 200032, China; Shanghai Institute of Cardiovascular Diseases, 180 Fenglin Road, Shanghai, 200032, China
| | - Xiaonan Zhou
- Department of Cardiac Surgery, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, 200032, China; Shanghai Institute of Cardiovascular Diseases, 180 Fenglin Road, Shanghai, 200032, China
| | - Gang Liu
- Department of Cardiac Surgery, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, 200032, China; Shanghai Institute of Cardiovascular Diseases, 180 Fenglin Road, Shanghai, 200032, China
| | - Mieradilijiang Abudupataer
- Department of Cardiac Surgery, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, 200032, China; Shanghai Institute of Cardiovascular Diseases, 180 Fenglin Road, Shanghai, 200032, China
| | - Shichao Zhu
- Department of Cardiac Surgery, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, 200032, China; Shanghai Institute of Cardiovascular Diseases, 180 Fenglin Road, Shanghai, 200032, China
| | - Bitao Xiang
- Department of Cardiac Surgery, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, 200032, China; Shanghai Institute of Cardiovascular Diseases, 180 Fenglin Road, Shanghai, 200032, China
| | - Xiujie Yin
- Department of Cardiac Surgery, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, 200032, China; Shanghai Institute of Cardiovascular Diseases, 180 Fenglin Road, Shanghai, 200032, China
| | - Hao Lai
- Department of Cardiac Surgery, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, 200032, China; Shanghai Institute of Cardiovascular Diseases, 180 Fenglin Road, Shanghai, 200032, China
| | - Yongxin Sun
- Department of Cardiac Surgery, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, 200032, China; Shanghai Institute of Cardiovascular Diseases, 180 Fenglin Road, Shanghai, 200032, China
| | - Chunsheng Wang
- Department of Cardiac Surgery, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, 200032, China; Shanghai Institute of Cardiovascular Diseases, 180 Fenglin Road, Shanghai, 200032, China.
| | - Jun Li
- Department of Cardiac Surgery, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, 200032, China; Shanghai Institute of Cardiovascular Diseases, 180 Fenglin Road, Shanghai, 200032, China.
| | - Kai Zhu
- Department of Cardiac Surgery, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, 200032, China; Shanghai Institute of Cardiovascular Diseases, 180 Fenglin Road, Shanghai, 200032, China.
| |
Collapse
|
11
|
Jreije A, Medlej-Hashim M, Hajal J, Saliba Y, Chacar S, Fares N, Khouzami L. Calcitriol Supplementation Protects Against Apoptosis and Alleviates the Severity of Abdominal Aortic Aneurysm Induced by Angiotensin II and Anti-TGFβ. J Cardiovasc Transl Res 2022; 15:1340-1351. [PMID: 35445935 DOI: 10.1007/s12265-022-10254-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Accepted: 04/04/2022] [Indexed: 12/16/2022]
Abstract
The present study aims to assess the effect of vitamin D deficiency (VDD) and its supplementation on the severity of AAA in mice. AAA was induced by AngII and anti-TGF-β administration. Animals were divided into four groups: Sham, mice with AAA, mice with AAA, and VDD, and mice with AAA supplemented with calcitriol. Blood pressure, echocardiography, abdominal aortic tissues, and plasma samples were monitored for all groups. VDD was associated with enhanced activity of cleaved MMP-9 and elastin degradation and positively correlated with the severity of AAA. Calcitriol supplementation decreased the INFγ/IL-10 ratio and enhanced the Nrf2 pathway. Moreover, Cu/Zn-superoxide dismutase expression and catalase and neutral sphingomyelinase activity were exacerbated in AAA and VDD groups. Furthermore, calcitriol supplementation showed a significantly lower protein expression of caspase-8, caspase-3, Bid, and t-Bid, and prevented the apoptosis of VSMCs treated by AngII and anti-TGF-β. Calcitriol supplementation may alleviate AAA severity and could be of great interest in the clinical management of AAA. VDD enhances antioxidant enzymes activity and expression, whereas calcitriol supplementation alleviates AAA severity by re-activating Nrf2 and inhibiting apoptotic pathways.
Collapse
Affiliation(s)
- Afaf Jreije
- Laboratoire de Recherche en Physiologie Et Physiopathologie, Faculté de Médecine, Pôle Technologie Santé, Université Saint Joseph, Beirut, Lebanon
- Cellular and Molecular Physiopathologies (CAMP) Laboratory, Faculty of Sciences II, Lebanese University, Fanar, Lebanon
| | - Myrna Medlej-Hashim
- Cellular and Molecular Physiopathologies (CAMP) Laboratory, Faculty of Sciences II, Lebanese University, Fanar, Lebanon
| | - Joelle Hajal
- Laboratoire de Recherche en Physiologie Et Physiopathologie, Faculté de Médecine, Pôle Technologie Santé, Université Saint Joseph, Beirut, Lebanon
| | - Youakim Saliba
- Laboratoire de Recherche en Physiologie Et Physiopathologie, Faculté de Médecine, Pôle Technologie Santé, Université Saint Joseph, Beirut, Lebanon
| | - Stephanie Chacar
- Laboratoire de Recherche en Physiologie Et Physiopathologie, Faculté de Médecine, Pôle Technologie Santé, Université Saint Joseph, Beirut, Lebanon
| | - Nassim Fares
- Laboratoire de Recherche en Physiologie Et Physiopathologie, Faculté de Médecine, Pôle Technologie Santé, Université Saint Joseph, Beirut, Lebanon.
| | - Lara Khouzami
- Cellular and Molecular Physiopathologies (CAMP) Laboratory, Faculty of Sciences II, Lebanese University, Fanar, Lebanon.
| |
Collapse
|
12
|
Yin L, Kent EW, Wang B. Progress in murine models of ruptured abdominal aortic aneurysm. Front Cardiovasc Med 2022; 9:950018. [PMID: 36035911 PMCID: PMC9411998 DOI: 10.3389/fcvm.2022.950018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2022] [Accepted: 07/27/2022] [Indexed: 02/03/2023] Open
Abstract
Abdominal aortic aneurysm (AAA) is a focal dilation of the aorta that is prevalent in aged populations. The progressive and unpredictable expansion of AAA could result in aneurysmal rupture, which is associated with ~80% mortality. Due to the expanded screening efforts and progress in diagnostic tools, an ever-increasing amount of asymptomatic AAA patients are being identified yet without a cure to stop the rampant aortic expansion. A key barrier that hinders the development of effective AAA treatment is our incomplete understanding of the cellular and molecular basis of its pathogenesis and progression into rupture. Animal models provide invaluable mechanistic insights into AAA pathophysiology. However, there is no single experimental model that completely recapitulate the complex biology behind AAA, and different AAA-inducing methodologies are associated with distinct disease course and rupture rate. In this review article, we summarize the established murine models of ruptured AAA and discuss their respective strengths and utilities.
Collapse
Affiliation(s)
| | | | - Bowen Wang
- Department of Surgery, School of Medicine, University of Virginia, Charlottesville, VA, United States
| |
Collapse
|
13
|
Hu M, Hiroyasu S, Granville DJ, Kassiri Z. Implications of SM22α-Cre expression in keratinocytes and un-anticipated inflammatory skin lesions in a model of atherosclerosis. Am J Physiol Heart Circ Physiol 2022; 323:H528-H534. [PMID: 35930445 DOI: 10.1152/ajpheart.00325.2022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Genetically modified mice are widely used to recapitulate human diseases. Atherosclerosis can be induced in mice with low density lipoprotein receptor (Ldlr)-deficiency and high fat diet (HFD). Disintegrin and metalloproteinase-17 (ADAM17) in the smooth muscle cell (SMC) contributes to vascular pathologies, and hence its role in atherosclerosis was investigated. ADAM17 deletion in SMCs by Sm22α-Cre driver (Ldlr-/-/Adam17Sm22Cre) and HFD resulted in severe skin lesions in >70% of mice, associated with skin inflammation, which were not observed in Ldlr-/--HFD, nor in mice with SMC-deficiency of ADAM17 by a different Cre-driver (Ldlr-/-/Adam17Myh11Cre). We found that Sm22α is highly expressed in keratinocytes (compared to SMCs), which could underlie the observed skin lesion in Ldlr-/-/Adam17Sm22Cre-HFD. Although expression of Sm22α in non-SMC cells has been reported, this is the first study demonstrating a severe side-effect resulting from the off-target expression of Sm22α-Cre, resulting in ADAM17 loss in keratinocytes that led to a moribund state.
Collapse
Affiliation(s)
- Mei Hu
- Department of Physiology, Cardiovascular Research Center, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| | - Sho Hiroyasu
- Department of Dermatology, Osaka Metropolitan University Graduate School of Medicine, Osaka, Japan.,ICORD Centre and Department of Pathology and Laboratory Medicine, Vancouver Coastal Health Research Institute, University of British Columbia, Vancouver, BC, Canada
| | - David J Granville
- ICORD Centre and Department of Pathology and Laboratory Medicine, Vancouver Coastal Health Research Institute, University of British Columbia, Vancouver, BC, Canada
| | - Zamaneh Kassiri
- Department of Physiology, Cardiovascular Research Center, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| |
Collapse
|
14
|
Li Z, Cong X, Kong W. Matricellular proteins: Potential biomarkers and mechanistic factors in aortic aneurysms. J Mol Cell Cardiol 2022; 169:41-56. [DOI: 10.1016/j.yjmcc.2022.05.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2021] [Revised: 03/30/2022] [Accepted: 05/03/2022] [Indexed: 10/18/2022]
|
15
|
Effect of Anti-Hypertensive Medication on Plasma Concentrations of Lysyl Oxidase: Evidence for Aldosterone-IL-6-Dependent Regulation of Lysyl Oxidase Blood Concentration. Biomedicines 2022; 10:biomedicines10071748. [PMID: 35885053 PMCID: PMC9313098 DOI: 10.3390/biomedicines10071748] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Revised: 07/18/2022] [Accepted: 07/19/2022] [Indexed: 11/30/2022] Open
Abstract
Lysyl oxidase (LOX) is a secretory protein that catalyzes elastin and collagen cross-linking. Lowering LOX expression and activity in endothelial cells is associated with a high risk of aneurysms and vascular malformation. Interleukin-6 (IL-6), elevated in hypertension, is known to suppress LOX expression. The influence of anti-hypertensive medication on the plasma LOX concentration is currently unknown. In a cohort of 34 patients diagnosed with resistant hypertension and treated with up to nine different drugs, blood concentration of LOX was analyzed to identify drugs that have an impact on plasma LOX concentration. Key findings were confirmed in a second independent patient cohort of 37 patients diagnosed with dilated cardiomyopathy. Blood concentrations of aldosterone and IL-6 were analyzed. In vitro, the effect of IL-6 on LOX expression was analyzed in endothelial cells. Patients receiving aldosterone antagonists had the highest plasma LOX concentration in both cohorts. This effect was independent of sex, age, blood pressure, body mass index, and co-medication. Blood aldosterone concentration correlates with plasma IL-6 concentration. In vitro, IL-6 decreased the expression of LOX in endothelial cells but not fibroblasts. Aldosterone was identified as a factor that affects blood concentration of LOX in an IL-6-dependent manner.
Collapse
|
16
|
Geng N, Chen T, Chen L, Zhang H, Sun L, Lyu Y, Che X, Xiao Q, Tao Z, Shao Q. Nuclear receptor Nur77 protects against oxidative stress by maintaining mitochondrial homeostasis via regulating mitochondrial fission and mitophagy in smooth muscle cell. J Mol Cell Cardiol 2022; 170:22-33. [PMID: 35661620 DOI: 10.1016/j.yjmcc.2022.05.007] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Revised: 04/25/2022] [Accepted: 05/12/2022] [Indexed: 02/03/2023]
Abstract
Angiotensin II (AngII) induces disruption of mitochondrial homeostasis and oxidative stress. Nuclear receptor NR4A1 (Nur77) plays an important role in vascular smooth muscle cells (VSMCs) function. However, the role of Nur77 in AngII-induced mitochondrial dynamics and oxidative stress in VSMCs remains unknown. In an in vitro model of AngII-treated cells, we discovered that Nur77 knockout aggravated AngII-induced oxidative stress in VSMCs, whereas activation of Nur77 by celastrol diminished them. Concomitantly, disturbance of mitochondrial dynamics induced by AngII was further exacerbated in Nur77 deficient VSMCs compared to wild-type (WT) VSMCs. Interestingly, Nur77 deletion increased mitochondrial fission but not fusion as evidenced by upregulated fission related genes (Fis1 and Drp1) but not fusion (Opa1 and Mfn2) under AngII stimulation in VSMCs. Mechanically, Nur77 could directly bind to the promoter regions of Fis1 and Drp1 and repress their transcription. Furthermore, we observed that Nur77 additionally promoted mitochondrial homeostasis by increasing mitophagic flux in a transcription-independent manner upon AngII challenge. By using an in vivo model of AngII-induced abdominal aortic aneurysm (AAA), we finally validated the protective role of Nur77 involved in the mitochondrial fission process and mitophagic flux in aortas, which was correlated with the occurrence and development of AAA in AngII-infused mice. Our data defines an essential role of Nur77 in regulating oxidative stress by maintaining mitochondrial homeostasis in VSMCs via both transcription-dependent and transcription-independent manner, supporting the therapeutic potential of Nur77 targeting in vascular diseases.
Collapse
Affiliation(s)
- Na Geng
- Department of Cardiology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Taiwei Chen
- Department of Cardiology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Long Chen
- Department of Cardiology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Hengyuan Zhang
- Department of Cardiology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Lingyue Sun
- Department of Cardiology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yuyan Lyu
- Department of Cardiology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xinyu Che
- Department of Cardiology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Qingqing Xiao
- Department of Cardiology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhenyu Tao
- Department of Cardiology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Qin Shao
- Department of Cardiology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, 241 West Huaihai Road, Shanghai 200030, China.
| |
Collapse
|
17
|
Kilic T, Okuno K, Eguchi S, Kassiri Z. Disintegrin and Metalloproteinases (ADAMs [A Disintegrin and Metalloproteinase] and ADAMTSs [ADAMs With a Thrombospondin Motif]) in Aortic Aneurysm. Hypertension 2022; 79:1327-1338. [PMID: 35543145 DOI: 10.1161/hypertensionaha.122.17963] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Aortic aneurysm is a complex pathology that can be lethal if not detected in time. Although several molecular mechanisms and pathways have been identified to be involved in aortic aneurysm development and growth, the current lack of an effective pharmacological treatment highlights the need for a more thorough understanding of the factors that regulate the remodeling of the aortic wall in response to triggers that lead to aneurysm formation. This task is further complicated by the regional heterogeneity of the aorta and that thoracic and abdominal aortic aneurysm are distinct pathologies with different risk factors and distinct course of progression. ADAMs (a disintegrin and metalloproteinases) and ADAMTS (ADAMs with a thrombospondin motif) are proteinases that share similarities with other proteinases but possess unique and diverse properties that place them in a category of their own. In this review, we discuss what is known on how ADAMs and ADAMTSs are altered in abdominal aortic aneurysm and thoracic aortic aneurysm in patients, in different animal models, and their role in regulating the function of different vascular and inflammatory cell types. A full understanding of the role of ADAMs and ADAMTSs in aortic aneurysm will help reveal a more complete understanding of the underlying mechanism driving aneurysm formation, which will help towards developing an effective treatment in preventing or limiting the growth of aortic aneurysm.
Collapse
Affiliation(s)
- Tolga Kilic
- Department of Physiology, Cardiovascular Research Center, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Canada (T.K., Z.K.)
| | - Keisuke Okuno
- Cardiovascular Research Center and Department of Cardiovascular Science, Lewis Katz School of Medicine at Temple University, Philadelphia, PA (K.O., S.E.)
| | - Satoru Eguchi
- Cardiovascular Research Center and Department of Cardiovascular Science, Lewis Katz School of Medicine at Temple University, Philadelphia, PA (K.O., S.E.)
| | - Zamaneh Kassiri
- Department of Physiology, Cardiovascular Research Center, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Canada (T.K., Z.K.)
| |
Collapse
|
18
|
ADAM17: A novel treatment target for aneurysms. Biomed Pharmacother 2022; 148:112712. [DOI: 10.1016/j.biopha.2022.112712] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Revised: 02/02/2022] [Accepted: 02/08/2022] [Indexed: 12/20/2022] Open
|
19
|
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: 103] [Impact Index Per Article: 34.3] [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
|
20
|
ADAM and ADAMTS disintegrin and metalloproteinases as major factors and molecular targets in vascular malfunction and disease. ADVANCES IN PHARMACOLOGY (SAN DIEGO, CALIF.) 2022; 94:255-363. [PMID: 35659374 PMCID: PMC9231755 DOI: 10.1016/bs.apha.2021.11.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
A Disintegrin and Metalloproteinase (ADAM) and A Disintegrin and Metalloproteinase with Thrombospondin Motifs (ADAMTS) are two closely related families of proteolytic enzymes. ADAMs are largely membrane-bound enzymes that act as molecular scissors or sheddases of membrane-bound proteins, growth factors, cytokines, receptors and ligands, whereas ADAMTS are mainly secreted enzymes. ADAMs have a pro-domain, and a metalloproteinase, disintegrin, cysteine-rich and transmembrane domain. Similarly, ADAMTS family members have a pro-domain, and a metalloproteinase, disintegrin, and cysteine-rich domain, but instead of a transmembrane domain they have thrombospondin motifs. Most ADAMs and ADAMTS are activated by pro-protein convertases, and can be regulated by G-protein coupled receptor agonists, Ca2+ ionophores and protein kinase C. Activated ADAMs and ADAMTS participate in numerous vascular processes including angiogenesis, vascular smooth muscle cell proliferation and migration, vascular cell apoptosis, cell survival, tissue repair, and wound healing. ADAMs and ADAMTS also play a role in vascular malfunction and cardiovascular diseases such as hypertension, atherosclerosis, coronary artery disease, myocardial infarction, heart failure, peripheral artery disease, and vascular aneurysm. Decreased ADAMTS13 is involved in thrombotic thrombocytopenic purpura and microangiopathies. The activity of ADAMs and ADAMTS can be regulated by endogenous tissue inhibitors of metalloproteinases and other synthetic small molecule inhibitors. ADAMs and ADAMTS can be used as diagnostic biomarkers and molecular targets in cardiovascular disease, and modulators of ADAMs and ADAMTS activity may provide potential new approaches for the management of cardiovascular disorders.
Collapse
|
21
|
Singh B, Singh D, Verma V, Yadav R, Kumar R. Angiotensin-converting enzyme 2 as a potential therapeutic target for COVID-19: A review. J Pharm Anal 2021; 12:215-220. [PMID: 34934510 PMCID: PMC8677424 DOI: 10.1016/j.jpha.2021.12.003] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2021] [Revised: 12/04/2021] [Accepted: 12/14/2021] [Indexed: 02/06/2023] Open
Abstract
As of August 16, 2021, there have been 207,173,086 confirmed cases and 4,361,996 deaths due to the coronavirus disease (COVID-19), and the pandemic remains a global challenge. To date, no effective and approved drugs are available for the treatment of COVID-19. Angiotensin-converting enzyme 2 (ACE2) plays a crucial role in the invasion into host cells by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), the etiological agent of COVID-19. Notably, ACE2 density is influenced by medical conditions, such as hypertension, or by drugs, including angiotensin-converting enzyme inhibitors (ACEIs) and angiotensin receptor blockers (ARBs), which can change the fate of SARS-CoV-2 infectivity. ACE2 is a target for these drugs and can be manipulated to limit the viral entry and replication within the cells. Different strategies aimed at blocking ACE2 with small molecules, peptides, and antibodies, or by neutralizing the virus through its competitive binding with human recombinant soluble ACE2 (hrsACE2) are currently under investigation. In this article, we review the current state of knowledge that emphasizes the need to find effective therapeutic agents against COVID-19 by exploiting ACE2 as a potential target. The increased soluble ACE2 levels and the application of hrsACE2 in patients with COVID-19 can be implemented to control the disease. It has not yet been established whether hypertension and other comorbidities, independent of age, have a direct role in COVID-19. Therefore, the use of renin-angiotensin system inhibitors, ACEIs and ARBs, should not be discontinued during COVID-19 treatment. Blockage of the interaction between the SARS-CoV-2 S protein and ACE2 as a strategy to treat COVID-19 is underway. ACE2 upregulation leads to the increased release of soluble ACE2. Increasing the levels of soluble ACE2 and hrsACE2 has the potential to prevent SARS-CoV-2 infection and reverse lung injury.
Collapse
Affiliation(s)
- Bhagat Singh
- Department of Medical Laboratory Technology, Faculty of Paramedical Sciences, Uttar Pradesh University of Medical Sciences, Etawah, 206130, India
| | - Dheer Singh
- Department of Anaesthesiology and Critical Care, Uttar Pradesh University of Medical Sciences, Etawah, 206130, India
| | - Vinod Verma
- Stem Cell Research Centre, Department of Hematology, Sanjay Gandhi Post Graduate Institute of Medical Sciences, Lucknow, 226014, India
| | - Ramakant Yadav
- Department of Neurology, Uttar Pradesh University of Medical Sciences, Etawah, 206130, India
| | - Raj Kumar
- Department of Neurosurgery, Uttar Pradesh University of Medical Sciences, Etawah, 206130, India
| |
Collapse
|
22
|
Wei M, Wang X, Song Y, Zhu D, Qi D, Jiao S, Xie G, Liu Y, Yu B, Du J, Wang Y, Qu A. Inhibition of Peptidyl Arginine Deiminase 4-Dependent Neutrophil Extracellular Trap Formation Reduces Angiotensin II-Induced Abdominal Aortic Aneurysm Rupture in Mice. Front Cardiovasc Med 2021; 8:676612. [PMID: 34395553 PMCID: PMC8360833 DOI: 10.3389/fcvm.2021.676612] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Accepted: 06/30/2021] [Indexed: 01/08/2023] Open
Abstract
Objective: Neutrophil infiltration plays an important role in the initiation and development of abdominal aortic aneurysm (AAA). Recent studies suggested that neutrophils could release neutrophil extracellular traps (NETs), leading to tissue injury in cardiovascular diseases. However, the role of NETs in AAA is elusive. This study aimed to investigate the role and underlying mechanism of NETs in AAA development. Methods and Results: An angiotensin II (Ang II) infusion-induced AAA model was established to investigate the role of NETs during AAA development. Immunofluorescence staining showed that citrullinated histone 3 (citH3), myeloperoxidase (MPO), and neutrophil elastase (NE) (NET marker) expressions were significantly increased in Ang II-infused ApoE−/− mice. The circulating double-stranded DNA (dsDNA) level was also elevated, indicating the increased NET formation during AAA. PAD4 inhibitor YW3-56 inhibited Ang II-induced NET formation. Disruption of NET formation by YW3-56 markedly reduced Ang II-induced AAA rupture, as revealed by decreased aortic diameter, vascular smooth muscle cell (VSMC) apoptosis, and elastin degradation. Apoptosis of VSMC was evaluated by TUNEL staining and Annexin V-FITC/PI staining through flow cytometry. Western blot and inhibition experiments revealed that NETs induced VSMC apoptosis via p38/JNK pathway, indicating that PAD4-dependent NET formation played an important role in AAA. Conclusions: This study suggests that PAD4-dependent NET formation is critical for AAA rupture, which provides a novel potential therapeutic strategy for AAA disease.
Collapse
Affiliation(s)
- Ming Wei
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing, China.,Key Laboratory of Remodeling-Related Cardiovascular Diseases, Ministry of Education, Beijing, China
| | - Xia Wang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing, China.,Key Laboratory of Remodeling-Related Cardiovascular Diseases, Ministry of Education, Beijing, China
| | - Yanting Song
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing, China.,Key Laboratory of Remodeling-Related Cardiovascular Diseases, Ministry of Education, Beijing, China
| | - Di Zhu
- School of Pharmaceutical Sciences, Capital Medical University, Beijing, China
| | - Dan Qi
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing, China.,Key Laboratory of Remodeling-Related Cardiovascular Diseases, Ministry of Education, Beijing, China
| | - Shiyu Jiao
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing, China.,Key Laboratory of Remodeling-Related Cardiovascular Diseases, Ministry of Education, Beijing, China
| | - Guomin Xie
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing, China.,Key Laboratory of Remodeling-Related Cardiovascular Diseases, Ministry of Education, Beijing, China
| | - Ye Liu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing, China.,Key Laboratory of Remodeling-Related Cardiovascular Diseases, Ministry of Education, Beijing, China
| | - Baoqi Yu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing, China.,Key Laboratory of Remodeling-Related Cardiovascular Diseases, Ministry of Education, Beijing, China
| | - Jie Du
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing, China.,Beijing Institute of Heart, Lung and Blood Vessel Diseases, Beijing, China.,Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Yuji Wang
- School of Pharmaceutical Sciences, Capital Medical University, Beijing, China
| | - Aijuan Qu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing, China.,Key Laboratory of Remodeling-Related Cardiovascular Diseases, Ministry of Education, Beijing, China
| |
Collapse
|
23
|
Behl T, Shah S, Kaur I, Yadav S, Kanwar R, Seth S, Wig N, Sharma KK, Yadav HN. Role of ACE 2 and Vitamin D: The Two Players in Global Fight against COVID-19 Pandemic. ANNALS OF THE NATIONAL ACADEMY OF MEDICAL SCIENCES (INDIA) 2021. [DOI: 10.1055/s-0041-1729781] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
AbstractThe global pandemic of coronavirus disease 2019 (COVID-19) has spread across the borders, gaining attention from both health care professional and researchers to understand the mode of entry and actions induced by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), its causative agent in the human body. The role of angiotensin-converting enzyme–2 (ACE2) in facilitating the entry of the virus in the host cell by binding to it is similar to SARS-CoV-1, the causative agent for severe acute respiratory syndrome (SARS) which emerged in 2003. Besides the role of ACE2 as a molecular target for the virus, the review displays the potential benefits of ACE2 enzyme and various agents that modify its activity in curbing the effects of the deadly virus, thus unfolding a dual character of ACE2 in the current pandemic. As evident by the differences in the susceptibility toward viral infection in children and geriatric population, it must be noted that the older population has limited ACE2 levels and greater infection risk, whereas the situation is reversed in the case of the pediatric population, demonstrating the defensive character of ACE2 in the latter, despite acting as receptor target for SARS-CoV-2. Also, the upregulation of ACE2 levels by estrogen has indicated greater resistance to infection in females than in the male human population. ACE2 is a carboxypeptidase, which degrades angiotensin II and counteracts its actions to protect against cardiovascular risks associated with the virus. Another contribution of this enzyme is supported by the role of circulating soluble ACE2, which acts as a receptor to bind the virus but does not mediate its actions, therefore blocking its interaction to membrane-bound ACE2 receptors. The review also shares the enhanced risks of developing COVID-19 infection by using ACE inhibitors and ARBs. However, both these agents have been reported to upregulate ACE2 levels; yet, adequate evidence regarding their role is quite inconsistent in human studies. Furthermore, the role of vitamin D has been highlighted in regulating the immune system of the body through renin-angiotensin-aldosterone system (RAAS) inhibition, by downregulating host cell receptor expression to prevent virus attachment. Besides, vitamin D also acts through several other mechanisms like upregulating antimicrobial peptides, fighting against the proinflammatory milieu created by the invading virus, and interfering with the viral replication cycle as well as calcitriol-mediated blockage of CREB protein. Hypovitaminosis D is attributed to elevated risks of acute respiratory distress syndrome (ARDS), lung damage, and cardiovascular disorders, further increasing the severity of COVID-19 infection.
Collapse
Affiliation(s)
- Tapan Behl
- Department of Pharmacology, Chitkara University, Punjab, India
| | - Sadia Shah
- Department of Pharmacology, All India Institute of Medical Sciences, New Delhi, India
| | - Ishnoor Kaur
- Department of Pharmacology, Chitkara University, Punjab, India
| | - Sushma Yadav
- Department of Obstetrics and Gynaecology, Shaheed Hasan Khan Mewati Government Medical College, Haryana, India
| | - Raj Kanwar
- Department of Nephrology, All India Institute of Medical Sciences, New Delhi, India
| | - Sandeep Seth
- Department of Cardiology, All India Institute of Medical Sciences, New Delhi, India
| | - Naveet Wig
- Department of Medicine, All India Institute of Medical Sciences, New Delhi, India
| | - K. K. Sharma
- National Academy of Medical Sciences (India), New Delhi, India
| | | |
Collapse
|
24
|
Kawai T, Elliott KJ, Scalia R, Eguchi S. Contribution of ADAM17 and related ADAMs in cardiovascular diseases. Cell Mol Life Sci 2021; 78:4161-4187. [PMID: 33575814 PMCID: PMC9301870 DOI: 10.1007/s00018-021-03779-w] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Revised: 12/23/2020] [Accepted: 01/27/2021] [Indexed: 02/06/2023]
Abstract
A disintegrin and metalloproteases (ADAMs) are key mediators of cell signaling by ectodomain shedding of various growth factors, cytokines, receptors and adhesion molecules at the cellular membrane. ADAMs regulate cell proliferation, cell growth, inflammation, and other regular cellular processes. ADAM17, the most extensively studied ADAM family member, is also known as tumor necrosis factor (TNF)-α converting enzyme (TACE). ADAMs-mediated shedding of cytokines such as TNF-α orchestrates immune system or inflammatory cascades and ADAMs-mediated shedding of growth factors causes cell growth or proliferation by transactivation of the growth factor receptors including epidermal growth factor receptor. Therefore, increased ADAMs-mediated shedding can induce inflammation, tissue remodeling and dysfunction associated with various cardiovascular diseases such as hypertension and atherosclerosis, and ADAMs can be a potential therapeutic target in these diseases. In this review, we focus on the role of ADAMs in cardiovascular pathophysiology and cardiovascular diseases. The main aim of this review is to stimulate new interest in this area by highlighting remarkable evidence.
Collapse
Affiliation(s)
- Tatsuo Kawai
- Cardiovascular Research Center, Lewis Katz School of Medicine At Temple University, Philadelphia, PA, USA
| | - Katherine J Elliott
- Cardiovascular Research Center, Lewis Katz School of Medicine At Temple University, Philadelphia, PA, USA
| | - Rosario Scalia
- Cardiovascular Research Center, Lewis Katz School of Medicine At Temple University, Philadelphia, PA, USA
| | - Satoru Eguchi
- Cardiovascular Research Center, Lewis Katz School of Medicine At Temple University, Philadelphia, PA, USA.
| |
Collapse
|
25
|
Adams L, Brangsch J, Hamm B, Makowski MR, Keller S. Targeting the Extracellular Matrix in Abdominal Aortic Aneurysms Using Molecular Imaging Insights. Int J Mol Sci 2021; 22:ijms22052685. [PMID: 33799971 PMCID: PMC7962044 DOI: 10.3390/ijms22052685] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Revised: 03/02/2021] [Accepted: 03/04/2021] [Indexed: 12/22/2022] Open
Abstract
This review outlines recent preclinical and clinical advances in molecular imaging of abdominal aortic aneurysms (AAA) with a focus on molecular magnetic resonance imaging (MRI) of the extracellular matrix (ECM). In addition, developments in pharmacologic treatment of AAA targeting the ECM will be discussed and results from animal studies will be contrasted with clinical trials. Abdominal aortic aneurysm (AAA) is an often fatal disease without non-invasive pharmacologic treatment options. The ECM, with collagen type I and elastin as major components, is the key structural component of the aortic wall and is recognized as a target tissue for both initiation and the progression of AAA. Molecular imaging allows in vivo measurement and characterization of biological processes at the cellular and molecular level and sets forth to visualize molecular abnormalities at an early stage of disease, facilitating novel diagnostic and therapeutic pathways. By providing surrogate criteria for the in vivo evaluation of the effects of pharmacological therapies, molecular imaging techniques targeting the ECM can facilitate pharmacological drug development. In addition, molecular targets can also be used in theranostic approaches that have the potential for timely diagnosis and concurrent medical therapy. Recent successes in preclinical studies suggest future opportunities for clinical translation. However, further clinical studies are needed to validate the most promising molecular targets for human application.
Collapse
Affiliation(s)
- Lisa Adams
- Charité—Universitaetsmedizin Berlin Corporate Member of Freie Universität Berlin Humboldt-Universitaet zu Berlin, and Berlin Institute of Health, Charitéplatz 1, 10117 Berlin, Germany; (J.B.); (B.H.); (M.R.M.); (S.K.)
- Berlin Institute of Health (BIH), 10178 Berlin, Germany
- Correspondence: ; Tel.: +49-30-450-627-376
| | - Julia Brangsch
- Charité—Universitaetsmedizin Berlin Corporate Member of Freie Universität Berlin Humboldt-Universitaet zu Berlin, and Berlin Institute of Health, Charitéplatz 1, 10117 Berlin, Germany; (J.B.); (B.H.); (M.R.M.); (S.K.)
| | - Bernd Hamm
- Charité—Universitaetsmedizin Berlin Corporate Member of Freie Universität Berlin Humboldt-Universitaet zu Berlin, and Berlin Institute of Health, Charitéplatz 1, 10117 Berlin, Germany; (J.B.); (B.H.); (M.R.M.); (S.K.)
| | - Marcus R. Makowski
- Charité—Universitaetsmedizin Berlin Corporate Member of Freie Universität Berlin Humboldt-Universitaet zu Berlin, and Berlin Institute of Health, Charitéplatz 1, 10117 Berlin, Germany; (J.B.); (B.H.); (M.R.M.); (S.K.)
- Department of Diagnostic and Interventional Radiology, Technical University of Munich, Ismaninger Str. 22, 81675 Munich, Germany
| | - Sarah Keller
- Charité—Universitaetsmedizin Berlin Corporate Member of Freie Universität Berlin Humboldt-Universitaet zu Berlin, and Berlin Institute of Health, Charitéplatz 1, 10117 Berlin, Germany; (J.B.); (B.H.); (M.R.M.); (S.K.)
| |
Collapse
|
26
|
Cooper HA, Cicalese S, Preston KJ, Kawai T, Okuno K, Choi ET, Kasahara S, Uchida HA, Otaka N, Scalia R, Rizzo V, Eguchi S. Targeting mitochondrial fission as a potential therapeutic for abdominal aortic aneurysm. Cardiovasc Res 2021; 117:971-982. [PMID: 32384150 PMCID: PMC7898955 DOI: 10.1093/cvr/cvaa133] [Citation(s) in RCA: 71] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/27/2019] [Revised: 04/15/2020] [Accepted: 04/30/2020] [Indexed: 11/12/2022] Open
Abstract
AIMS Angiotensin II (AngII) is a potential contributor to the development of abdominal aortic aneurysm (AAA). In aortic vascular smooth muscle cells (VSMCs), exposure to AngII induces mitochondrial fission via dynamin-related protein 1 (Drp1). However, pathophysiological relevance of mitochondrial morphology in AngII-associated AAA remains unexplored. Here, we tested the hypothesis that mitochondrial fission is involved in the development of AAA. METHODS AND RESULTS Immunohistochemistry was performed on human AAA samples and revealed enhanced expression of Drp1. In C57BL6 mice treated with AngII plus β-aminopropionitrile, AAA tissue also showed an increase in Drp1 expression. A mitochondrial fission inhibitor, mdivi1, attenuated AAA size, associated aortic pathology, Drp1 protein induction, and mitochondrial fission but not hypertension in these mice. Moreover, western-blot analysis showed that induction of matrix metalloproteinase-2, which precedes the development of AAA, was blocked by mdivi1. Mdivi1 also reduced the development of AAA in apolipoprotein E-deficient mice infused with AngII. As with mdivi1, Drp1+/- mice treated with AngII plus β-aminopropionitrile showed a decrease in AAA compared to control Drp1+/+ mice. In abdominal aortic VSMCs, AngII induced phosphorylation of Drp1 and mitochondrial fission, the latter of which was attenuated with Drp1 silencing as well as mdivi1. AngII also induced vascular cell adhesion molecule-1 expression and enhanced leucocyte adhesion and mitochondrial oxygen consumption in smooth muscle cells, which were attenuated with mdivi1. CONCLUSION These data indicate that Drp1 and mitochondrial fission play salient roles in AAA development, which likely involves mitochondrial dysfunction and inflammatory activation of VSMCs.
Collapse
MESH Headings
- Aminopropionitrile
- Angiotensin II
- Animals
- Anti-Inflammatory Agents/pharmacology
- Aorta, Abdominal/drug effects
- Aorta, Abdominal/metabolism
- Aorta, Abdominal/pathology
- Aortic Aneurysm, Abdominal/chemically induced
- Aortic Aneurysm, Abdominal/metabolism
- Aortic Aneurysm, Abdominal/pathology
- Aortic Aneurysm, Abdominal/prevention & control
- Case-Control Studies
- Cell Adhesion/drug effects
- Cells, Cultured
- Disease Models, Animal
- Dynamins/genetics
- Dynamins/metabolism
- Humans
- Leukocytes/drug effects
- Leukocytes/metabolism
- Male
- Mice, Inbred C57BL
- Mice, Knockout, ApoE
- Mitochondria, Muscle/drug effects
- Mitochondria, Muscle/genetics
- Mitochondria, Muscle/metabolism
- Mitochondria, Muscle/pathology
- Mitochondrial Dynamics/drug effects
- Muscle, Smooth, Vascular/drug effects
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/pathology
- Myocytes, Smooth Muscle/drug effects
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/pathology
- Oxygen Consumption/drug effects
- Phosphorylation
- Quinazolinones/pharmacology
- Mice
Collapse
Affiliation(s)
- Hannah A Cooper
- Cardiovascular Research Center, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA
| | - Stephanie Cicalese
- Cardiovascular Research Center, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA
| | - Kyle J Preston
- Cardiovascular Research Center, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA
| | - Tatsuo Kawai
- Cardiovascular Research Center, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA
| | - Keisuke Okuno
- Cardiovascular Research Center, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA
| | - Eric T Choi
- Department of Surgery, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA
| | - Shingo Kasahara
- Department of Cardiovascular Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8558, Japan
| | - Haruhito A Uchida
- Department of Chronic Kidney Disease and Cardiovascular Disease, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8558, Japan
| | - Nozomu Otaka
- Department of Nephrology, Rheumatology, Endocrinology and Metabolism, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8558, Japan
| | - Rosario Scalia
- Cardiovascular Research Center, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA
| | - Victor Rizzo
- Cardiovascular Research Center, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA
| | - Satoru Eguchi
- Cardiovascular Research Center, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA
| |
Collapse
|
27
|
Okuyama M, Jiang W, Javidan A, Chen JZ, Howatt DA, Yang L, Hamaguchi M, Yasugi T, Aono J, Vazquez-Padron RI, Subramanian V. Lysyl Oxidase Inhibition Ablates Sexual Dimorphism of Abdominal Aortic Aneurysm Formation in Mice. Circulation 2020; 142:1993-1995. [PMID: 33196308 DOI: 10.1161/circulationaha.119.044986] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Affiliation(s)
- Michihiro Okuyama
- Saha Cardiovascular Research Center (M.O., W.J., A.J., J.Z.C., D.A.H., L.Y., V.S.), University of Kentucky, Lexington
| | - Weihua Jiang
- Saha Cardiovascular Research Center (M.O., W.J., A.J., J.Z.C., D.A.H., L.Y., V.S.), University of Kentucky, Lexington
| | - Aida Javidan
- Saha Cardiovascular Research Center (M.O., W.J., A.J., J.Z.C., D.A.H., L.Y., V.S.), University of Kentucky, Lexington
| | - Jeff Zheying Chen
- Saha Cardiovascular Research Center (M.O., W.J., A.J., J.Z.C., D.A.H., L.Y., V.S.), University of Kentucky, Lexington.,Department of Physiology (J.Z.C., V.S.), University of Kentucky, Lexington
| | - Deborah A Howatt
- Saha Cardiovascular Research Center (M.O., W.J., A.J., J.Z.C., D.A.H., L.Y., V.S.), University of Kentucky, Lexington
| | - Lihua Yang
- Saha Cardiovascular Research Center (M.O., W.J., A.J., J.Z.C., D.A.H., L.Y., V.S.), University of Kentucky, Lexington
| | - Mika Hamaguchi
- Department of Cardiology, Pulmonology, Hypertension, and Nephrology (M.H., J.A.), Ehime University Graduate School of Medicine, Toon, Japan
| | - Takumi Yasugi
- Department of Cardiovascular and Thoracic Surgery (T.Y.), Ehime University Graduate School of Medicine, Toon, Japan
| | - Jun Aono
- Department of Cardiology, Pulmonology, Hypertension, and Nephrology (M.H., J.A.), Ehime University Graduate School of Medicine, Toon, Japan
| | | | - Venkateswaran Subramanian
- Saha Cardiovascular Research Center (M.O., W.J., A.J., J.Z.C., D.A.H., L.Y., V.S.), University of Kentucky, Lexington.,Department of Physiology (J.Z.C., V.S.), University of Kentucky, Lexington
| |
Collapse
|
28
|
Factor XII blockade inhibits aortic dilatation in angiotensin II-infused apolipoprotein E-deficient mice. Clin Sci (Lond) 2020; 134:1049-1061. [PMID: 32309850 DOI: 10.1042/cs20191020] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Revised: 04/05/2020] [Accepted: 04/20/2020] [Indexed: 12/23/2022]
Abstract
Abdominal aortic aneurysm (AAA) is an important cause of mortality in older adults. Chronic inflammation and excessive matrix remodelling are considered important in AAA pathogenesis. Kinins are bioactive peptides important in regulating inflammation. Stimulation of the kinin B2 receptor has been previously reported to promote AAA development and rupture in a mouse model. The endogenous B2 receptor agonist, bradykinin, is generated from the kallikrein-kinin system following activation of plasma kallikrein by Factor XII (FXII). In the current study whole-body FXII deletion, or neutralisation of activated FXII (FXIIa), inhibited expansion of the suprarenal aorta (SRA) of apolipoprotein E-deficient mice in response to angiotensin II (AngII) infusion. FXII deficiency or FXIIa neutralisation led to decreased aortic tumor necrosis factor-α-converting enzyme (TACE/a disintegrin and metalloproteinase-17 (aka tumor necrosis factor-α-converting enzyme) (ADAM-17)) activity, plasma kallikrein concentration, and epithelial growth factor receptor (EGFR) phosphorylation compared with controls. FXII deficiency or neutralisation also reduced Akt1 and Erk1/2 phosphorylation and decreased expression and levels of active matrix metalloproteinase (Mmp)-2 and Mmp-9. The findings suggest that FXII, kallikrein, ADAM-17, and EGFR are important molecular mediators by which AngII induces aneurysm in apolipoprotein E-deficient mice. This could be a novel pathway to target in the design of drugs to limit AAA progression.
Collapse
|
29
|
Behl T, Kaur I, Bungau S, Kumar A, Uddin MS, Kumar C, Pal G, Sahil, Shrivastava K, Zengin G, Arora S. The dual impact of ACE2 in COVID-19 and ironical actions in geriatrics and pediatrics with possible therapeutic solutions. Life Sci 2020; 257:118075. [PMID: 32653522 PMCID: PMC7347488 DOI: 10.1016/j.lfs.2020.118075] [Citation(s) in RCA: 77] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2020] [Revised: 07/05/2020] [Accepted: 07/06/2020] [Indexed: 01/08/2023]
Abstract
The novel corona virus disease has shaken the entire world with its deadly effects and rapid transmission rates, posing a significant challenge to the healthcare authorities to develop suitable therapeutic solution to save lives on earth. The review aims to grab the attention of the researchers all over the globe, towards the role of ACE2 in COVID-19 disease. ACE2 serves as a molecular target for the SARS-CoV-2, to enter the target cell, by interacting with the viral glycoprotein spikes. However, the complexity began when numerous studies identified the protective response of ACE2 in abbreviating the harmful effects of vasoconstrictor, anti-inflammatory peptide, angiotensin 2, by mediating its conversion to angiotensin-(1-7), which exercised antagonistic actions to angiotensin 2. Furthermore, certain investigations revealed greater resistance among children as compared to the geriatrics, towards COVID-19 infection, despite the elevated expression of ACE2 in pediatric population. Based upon such evidences, the review demonstrated possible therapeutic interventions, targeting both the protective and deleterious effects of ACE2 in COVID-19 disease, primarily inhibiting ACE2-virus interactions or administering soluble ACE2. Thus, the authors aim to provide an opportunity for the researchers to consider RAAS system to be a significant element in development of suitable treatment regime for COVID-19 pandemic.
Collapse
Affiliation(s)
- Tapan Behl
- Chitkara College of Pharmacy, Chitkara University, 140401, Punjab, India.
| | - Ishnoor Kaur
- Chitkara College of Pharmacy, Chitkara University, 140401, Punjab, India
| | - Simona Bungau
- Department of Pharmacy, Faculty of Medicine and Pharmacy, University of Oradea, Romania.
| | - Arun Kumar
- Chitkara College of Pharmacy, Chitkara University, 140401, Punjab, India
| | - Md Sahab Uddin
- Department of Pharmacy, Southeast University, 1213 Dhaka, Bangladesh; Pharmakon Neuroscience Research Network, Dhaka, Bangladesh
| | - Chanchal Kumar
- Department of Medical Microbiology, Vallabhbhai Patel Chest Institute, University of Delhi, Delhi, India
| | - Giridhari Pal
- Vallabhbhai Patel Chest Institute, University of Delhi, New Delhi, 110007 Delhi, India
| | - Sahil
- Department of Computer Science, Panjab University, Chandigarh, India
| | - Kamal Shrivastava
- Department of Medical Microbiology, Vallabhbhai Patel Chest Institute, University of Delhi, Delhi, India
| | | | - Sandeep Arora
- Chitkara College of Pharmacy, Chitkara University, 140401, Punjab, India
| |
Collapse
|
30
|
Zheng HQ, Rong JB, Ye FM, Xu YC, Lu HS, Wang JA. Induction of thoracic aortic dissection: a mini-review of β-aminopropionitrile-related mouse models. J Zhejiang Univ Sci B 2020; 21:603-610. [PMID: 32748576 PMCID: PMC7445087 DOI: 10.1631/jzus.b2000022] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2020] [Accepted: 04/20/2020] [Indexed: 02/06/2023]
Abstract
Thoracic aortic dissection (TAD) is one of the most lethal aortic diseases due to its acute onset, rapid progress, and high rate of aortic rupture. The pathogenesis of TAD is not completely understood. In this mini-review, we introduce three emerging experimental mouse TAD models using β-aminopropionitrile (BAPN) alone, BAPN for a prolonged duration (four weeks) and then with added infusion of angiotensin II (AngII), or co-administration of BAPN and AngII chronically. We aim to provide insights into appropriate application of these three mouse models, thereby enhancing the understanding of the molecular mechanisms of TAD.
Collapse
Affiliation(s)
- Hai-qiong Zheng
- Department of Cardiology, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310009, China
- Cardiovascular Key Laboratory of Zhejiang Province, Hangzhou 310009, China
| | - Jia-bing Rong
- Department of Cardiology, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310009, China
- Cardiovascular Key Laboratory of Zhejiang Province, Hangzhou 310009, China
| | - Fei-ming Ye
- Department of Cardiology, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310009, China
- Cardiovascular Key Laboratory of Zhejiang Province, Hangzhou 310009, China
| | - Yin-chuan Xu
- Department of Cardiology, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310009, China
- Cardiovascular Key Laboratory of Zhejiang Province, Hangzhou 310009, China
| | - Hong S. Lu
- Saha Cardiovascular Research Center, University of Kentucky, Lexington, KY 40536, USA
- Department of Physiology, College of Medicine, University of Kentucky, Lexington, KY 40536, USA
| | - Jian-an Wang
- Department of Cardiology, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310009, China
- Cardiovascular Key Laboratory of Zhejiang Province, Hangzhou 310009, China
| |
Collapse
|
31
|
Gutsaeva DR, Shalaby L, Powell FL, Thounaojam MC, Abouhish H, Wetzstein SA, Jadeja RN, Kwok HF, Martin PM, Bartoli M. Inactivation of Endothelial ADAM17 Reduces Retinal Ischemia-Reperfusion Induced Neuronal and Vascular Damage. Int J Mol Sci 2020; 21:E5379. [PMID: 32751103 PMCID: PMC7432237 DOI: 10.3390/ijms21155379] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2020] [Revised: 07/24/2020] [Accepted: 07/27/2020] [Indexed: 12/12/2022] Open
Abstract
Retinal ischemia contributes to visual impairment in ischemic retinopathies. A disintegrin and metalloproteinase ADAM17 is implicated in multiple vascular pathologies through its ability to regulate inflammatory signaling via ectodomain shedding. We investigated the role of endothelial ADAM17 in neuronal and vascular degeneration associated with retinal ischemia reperfusion (IR) injury using mice with conditional inactivation of ADAM17 in vascular endothelium. ADAM17Cre-flox and control ADAM17flox mice were subjected to 40 min of pressure-induced retinal ischemia, with the contralateral eye serving as control. Albumin extravasation and retinal leukostasis were evaluated 48 h after reperfusion. Retinal morphometric analysis was conducted 7 days after reperfusion. Degenerate capillaries were assessed by elastase digest and visual function was evaluated by optokinetic test 14 and 7 days following ischemia, respectively. Lack of ADAM17 decreased vascular leakage and reduced retinal thinning and ganglion cell loss in ADAM17Cre-flox mice. Further, ADAM17Cre-flox mice exhibited a remarkable reduction in capillary degeneration following IR. Decrease in neurovascular degeneration in ADAM17Cre-flox mice correlated with decreased activation of caspase-3 and was associated with reduction in oxidative stress and retinal leukostasis. In addition, knockdown of ADAM17 resulted in decreased cleavage of p75NTR, the process known to be associated with retinal cell apoptosis. A decline in visual acuity evidenced by decrease in spatial frequency threshold observed in ADAM17flox mice was partially restored in ADAM17-endothelial deficient mice. The obtained results provide evidence that endothelial ADAM17 is an important contributor to IR-induced neurovascular damage in the retina and suggest that interventions directed at regulating ADAM17 activity can be beneficial for alleviating the consequences of retinal ischemia.
Collapse
Affiliation(s)
- Diana R Gutsaeva
- Department of Ophthalmology, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA; (L.S.); (M.C.T.); (H.A.); (M.B.)
| | - Lamiaa Shalaby
- Department of Ophthalmology, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA; (L.S.); (M.C.T.); (H.A.); (M.B.)
| | - Folami L Powell
- Department of Biochemistry and Molecular Biology, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA; (F.L.P.); (R.N.J.); (P.M.M.)
| | - Menaka C Thounaojam
- Department of Ophthalmology, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA; (L.S.); (M.C.T.); (H.A.); (M.B.)
| | - Hossameldin Abouhish
- Department of Ophthalmology, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA; (L.S.); (M.C.T.); (H.A.); (M.B.)
- Department of Clinical Pharmacology, Faculty of Medicine, Mansoura University, 35516 Mansoura, Egypt
| | | | - Ravirajsinh N Jadeja
- Department of Biochemistry and Molecular Biology, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA; (F.L.P.); (R.N.J.); (P.M.M.)
| | - Hang Fai Kwok
- Institute of Translational Medicine, Faculty of Health Sciences, University of Macau, Avenida de Universidade, Macau 999078;
| | - Pamela M Martin
- Department of Biochemistry and Molecular Biology, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA; (F.L.P.); (R.N.J.); (P.M.M.)
| | - Manuela Bartoli
- Department of Ophthalmology, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA; (L.S.); (M.C.T.); (H.A.); (M.B.)
| |
Collapse
|
32
|
Laczko R, Csiszar K. Lysyl Oxidase (LOX): Functional Contributions to Signaling Pathways. Biomolecules 2020; 10:biom10081093. [PMID: 32708046 PMCID: PMC7465975 DOI: 10.3390/biom10081093] [Citation(s) in RCA: 61] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Revised: 07/12/2020] [Accepted: 07/13/2020] [Indexed: 12/12/2022] Open
Abstract
Cu-dependent lysyl oxidase (LOX) plays a catalytic activity-related, primary role in the assembly of the extracellular matrix (ECM), a dynamic structural and regulatory framework which is essential for cell fate, differentiation and communication during development, tissue maintenance and repair. LOX, additionally, plays both activity-dependent and independent extracellular, intracellular and nuclear roles that fulfill significant functions in normal tissues, and contribute to vascular, cardiac, pulmonary, dermal, placenta, diaphragm, kidney and pelvic floor disorders. LOX activities have also been recognized in glioblastoma, diabetic neovascularization, osteogenic differentiation, bone matrix formation, ligament remodeling, polycystic ovary syndrome, fetal membrane rupture and tumor progression and metastasis. In an inflammatory context, LOX plays a role in diminishing pluripotent mesenchymal cell pools which are relevant to the pathology of diabetes, osteoporosis and rheumatoid arthritis. Most of these conditions involve mechanisms with complex cell and tissue type-specific interactions of LOX with signaling pathways, not only as a regulatory target, but also as an active player, including LOX-mediated alterations of cell surface receptor functions and mutual regulatory activities within signaling loops. In this review, we aim to provide insight into the diverse ways in which LOX participates in signaling events, and explore the mechanistic details and functional significance of the regulatory and cross-regulatory interactions of LOX with the EGFR, PDGF, VEGF, TGF-β, mechano-transduction, inflammatory and steroid signaling pathways.
Collapse
|
33
|
Roshanravan N, Ghaffari S, Hedayati M. Angiotensin converting enzyme-2 as therapeutic target in COVID-19. Diabetes Metab Syndr 2020; 14:637-639. [PMID: 32428864 PMCID: PMC7214324 DOI: 10.1016/j.dsx.2020.05.022] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/02/2020] [Revised: 05/09/2020] [Accepted: 05/09/2020] [Indexed: 01/01/2023]
Abstract
The pandemic of coronavirus disease 2019 (COVID-19) is a global health emergency that poses a significant threat to world people's health. This outbreak causes major challenges to healthcare systems. Given the lack of effective treatments or vaccine for it, the identification of novel and safe drugs against COVID-19 infection is an urgent need. Angiotensin-converting enzyme 2 (ACE2) is not only an entry receptor of the SARS-CoV-2 virus, the virus that causes COVID-19, but also can protect from lung injury. In this view, we highlighted potential approaches to address ACE2-mediated SARS-CoV-2 virus, including 1) delivering an excessive soluble form of ACE2 (recombinant human ACE2: rhACE2) and 2) inhibition of the interaction between SARS-CoV-2 virus and ACE2 by some compounds with competitive effects (morphine and codeine). Further clinical trials in this regard can reveal a more definite conclusion against the COVID-19 disaster.
Collapse
Affiliation(s)
- Neda Roshanravan
- Cardiovascular Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Samad Ghaffari
- Cardiovascular Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Mehdi Hedayati
- Cellular and Molecular Endocrine Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
34
|
Jana S, Chute M, Hu M, Winkelaar G, Owen CA, Oudit GY, Kassiri Z. ADAM (a Disintegrin and Metalloproteinase) 15 Deficiency Exacerbates Ang II (Angiotensin II)-Induced Aortic Remodeling Leading to Abdominal Aortic Aneurysm. Arterioscler Thromb Vasc Biol 2020; 40:1918-1934. [PMID: 32522006 PMCID: PMC7370975 DOI: 10.1161/atvbaha.120.314600] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Supplemental Digital Content is available in the text. Objective: ADAM (a disintegrin and metalloproteinase) 15—a membrane-bound metalloprotease from the ADAM (disintegrin and metalloproteinase) family—has been linked to endothelial permeability, inflammation, and metastasis. However, its function in aortic aneurysm has not been explored. We aimed to determine the function of ADAM15 in the pathogenesis of aortic remodeling and aneurysm formation. Approach and Results: Male Adam15-deficient and WT (wild type) mice (10 weeks old), on standard laboratory diet, received Ang II (angiotensin II; 1.5 mg/kg per day) or saline (Alzet pump) for 2 or 4 weeks. Ang II increased ADAM15 in WT aorta, while Adam15-deficiency resulted in abdominal aortic aneurysm characterized by loss of medial smooth muscle cells (SMCs), elastin fragmentation, inflammation, but unaltered Ang II–mediated hypertension. In the abdominal aortic tissue and primary aortic SMCs culture, Adam15 deficiency decreased SMC proliferation, increased apoptosis, and reduced contractile properties along with F-actin depolymerization to G-actin. Ang II triggered a markedly greater increase in THBS (thrombospondin) 1 in Adam15-deficient aorta, primarily the medial layer in vivo, and in aortic SMC in vitro; increased SSH1 (slingshot homolog 1) phosphatase activity and cofilin dephosphorylation that promoted F-actin depolymerization and G-actin accumulation. rhTHBS1 (recombinant THBS1) alone was sufficient to activate the cofilin pathway, increase G-actin, and induce apoptosis of aortic SMCs, confirming the key role of THBS1 in this process. Further, in human abdominal aortic aneurysm specimens, decreased ADAM15 was associated with increased THBS1 levels and loss of medial SMCs. Conclusions: This study is the first to demonstrate a key role for ADAM15 in abdominal aortic aneurysm through regulating the SMC function, thereby placing ADAM15 in a critical position as a potential therapeutic target for abdominal aortic aneurysm.
Collapse
Affiliation(s)
- Sayantan Jana
- From the Department of Physiology, Cardiovascular Research Center (S.J., M.C., M.H., G.Y.O., Z.K.), University of Alberta, Edmonton, Canada
| | - Michael Chute
- From the Department of Physiology, Cardiovascular Research Center (S.J., M.C., M.H., G.Y.O., Z.K.), University of Alberta, Edmonton, Canada
| | - Mei Hu
- From the Department of Physiology, Cardiovascular Research Center (S.J., M.C., M.H., G.Y.O., Z.K.), University of Alberta, Edmonton, Canada
| | - Gerrit Winkelaar
- Division of Vascular Surgery, University of Alberta and Northern Alberta Vascular Center, Grey Nuns Hospital, Edmonton, Canada (G.W.)
| | - Caroline A Owen
- Brigham and Women's Hospital/Harvard Medical School, Boston, MA (C.A.O.)
| | - Gavin Y Oudit
- From the Department of Physiology, Cardiovascular Research Center (S.J., M.C., M.H., G.Y.O., Z.K.), University of Alberta, Edmonton, Canada.,Department of Medicine, Division of Cardiology, Mazankowski Alberta Heart Institute (G.Y.O.), University of Alberta, Edmonton, Canada
| | - Zamaneh Kassiri
- From the Department of Physiology, Cardiovascular Research Center (S.J., M.C., M.H., G.Y.O., Z.K.), University of Alberta, Edmonton, Canada
| |
Collapse
|
35
|
Endoplasmic reticulum stress and mitochondrial biogenesis are potential therapeutic targets for abdominal aortic aneurysm. Clin Sci (Lond) 2020; 133:2023-2028. [PMID: 31654572 DOI: 10.1042/cs20190648] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2019] [Revised: 09/25/2019] [Accepted: 09/27/2019] [Indexed: 01/08/2023]
Abstract
Endoplasmic reticulum (ER) and mitochondria are crucial organelles for cell homeostasis and alterations of these organelles have been implicated in cardiovascular disease. However, their roles in abdominal aortic aneurysm (AAA) pathogenesis remain largely unknown. In a recent issue of Clinical Science, Navas-Madronal et al. ((2019), 133(13), 1421-1438) reported that enhanced ER stress and dysregulation of mitochondrial biogenesis are associated with AAA pathogenesis in humans. The authors also proposed that disruption in oxysterols network such as an elevated concentration of 7-ketocholestyerol in plasma is a causative factor for AAA progression. Their findings highlight new insights into the underlying mechanism of AAA progression through ER stress and dysregulation of mitochondrial biogenesis. Here, we will discuss the background, significance of the study, and future directions.
Collapse
|
36
|
Hu Z, Li B, Wang Z, Hu X, Zhang M, Chen R, Wu Q, Jia F. The sympathetic transmitter norepinephrine inhibits VSMC proliferation induced by TGFβ by suppressing the expression of the TGFβ receptor ALK5 in aorta remodeling. Mol Med Rep 2020; 22:387-397. [PMID: 32319652 PMCID: PMC7248509 DOI: 10.3892/mmr.2020.11088] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2019] [Accepted: 03/18/2020] [Indexed: 12/12/2022] Open
Abstract
The sympathetic system is involved in the arterial diseases, but its mechanism remains poorly understood. The present study aimed to explore the impact of the sympathetic neurotransmitter norepinephrine (NE) on transforming growth factor (TGF) β signaling and the role of NE in aortic remodeling. Guanethidine was used to induce a regional chemical sympathetic denervation (CSD) in angiotensin II (AngII) and β-aminopropionitrile (BAPN)-induced aortic aneurysm models. The diameter of the aorta was measured, and elastic fiber staining was performed. TGFβ type I receptor kinase (ALK5) expression in rat aortic NE-treated vascular smooth muscle cells (VSMCs) was detected by reverse transcription-quantitative PCR and western blotting. The effects of NE and ALK5 overexpression on migration, proliferation, apoptosis and TGFβ signaling were also evaluated. Furthermore, adrenergic receptor blockers were used to determine which receptor was involved in the modulation on TGFβ signaling by NE. The results of the present study demonstrated that CSD protected rats from AngII+BAPN-induced aortic remodeling and aneurysm formation. Compared with the control group, NE inhibited VSMC proliferation and migration, but promoted apoptosis by suppressing ALK5 expression, reversing the effects of TGFβ signaling through the suppression of the SMAD-dependent canonical pathway and promotion of the non-canonical pathway. These effects were prevented by ALK5 overexpression. The inhibition of α- or β-adrenergic receptors alleviated the NE-mediated suppression of ALK5 expression. In conclusion, regional CSD protected rats from aortic aneurysm. NE inhibited SMAD2/3-dependent TGFβ signaling by suppressing ALK5 expression, which may serve an important role in VSMC biological functions. Both α- and β-adrenergic receptors were involved in the regulation of ALK5 expression by NE. Abnormal sympathetic innervation of the aorta may be used as a therapeutic target in aortic diseases.
Collapse
Affiliation(s)
- Zhipeng Hu
- Department of Cardiovascular Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Bowen Li
- Department of Cardiovascular Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Zhiwei Wang
- Department of Cardiovascular Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Xiaoping Hu
- Department of Cardiovascular Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Min Zhang
- Department of Cardiovascular Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Ruoshi Chen
- Department of Cardiovascular Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Qi Wu
- Department of Cardiovascular Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Fangyuan Jia
- Department of Cardiovascular Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| |
Collapse
|
37
|
Li Y, Wang W, Li L, Khalil RA. MMPs and ADAMs/ADAMTS inhibition therapy of abdominal aortic aneurysm. Life Sci 2020; 253:117659. [PMID: 32283055 DOI: 10.1016/j.lfs.2020.117659] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2020] [Revised: 04/05/2020] [Accepted: 04/07/2020] [Indexed: 12/12/2022]
Abstract
Abdominal aortic aneurysm (AAA) is a chronic vascular degenerative disease featured by progressive dilation and remodeling of the vascular wall, which may lead to aortic rupture and high mortality. The occurrence and development of AAA involve multiple mechanisms, including extracellular matrix degradation, chronic inflammation, oxidative stress, apoptosis of vascular smooth muscle cells and innate immunity. Extracellular matrix degradation is considered as the most important mechanism causing AAA. Matrix metalloproteinases (MMPs) are key factors in this process, contributing greatly to the occurrence and development of AAA. But whether the zinc-dependent endopeptidases (ADAM/ADAMTS) are involved in this process is very little known. This study is a review about the role of MMPs and ADAM/ADAMT as well as the existing MMP inhibitors in abdominal aortic aneurysm, with the purpose of providing reference for the clinical treatment of abdominal aortic aneurysm.
Collapse
Affiliation(s)
- Yongqi Li
- Department of Vascular Surgery, The Second Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China; Graduate School of Comprehensive Human Sciences, University of Tsukuba, Japan
| | - Weicheng Wang
- Emergency Center, The Second Hospital of Dalian Medical University, Dalian, Liaoning, China
| | - Lei Li
- Department of Vascular Surgery, The Second Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China; Vascular Surgery Research Laboratories, Division of Vascular and Endovascular Surgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.
| | - Raouf A Khalil
- Vascular Surgery Research Laboratories, Division of Vascular and Endovascular Surgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
38
|
Liu B, Granville DJ, Golledge J, Kassiri Z. Pathogenic mechanisms and the potential of drug therapies for aortic aneurysm. Am J Physiol Heart Circ Physiol 2020; 318:H652-H670. [PMID: 32083977 PMCID: PMC7099451 DOI: 10.1152/ajpheart.00621.2019] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/23/2019] [Revised: 02/13/2020] [Accepted: 02/13/2020] [Indexed: 12/14/2022]
Abstract
Aortic aneurysm is a permanent focal dilation of the aorta. It is usually an asymptomatic disease but can lead to sudden death due to aortic rupture. Aortic aneurysm-related mortalities are estimated at ∼200,000 deaths per year worldwide. Because no pharmacological treatment has been found to be effective so far, surgical repair remains the only treatment for aortic aneurysm. Aortic aneurysm results from changes in the aortic wall structure due to loss of smooth muscle cells and degradation of the extracellular matrix and can form in different regions of the aorta. Research over the past decade has identified novel contributors to aneurysm formation and progression. The present review provides an overview of cellular and noncellular factors as well as enzymes that process extracellular matrix and regulate cellular functions (e.g., matrix metalloproteinases, granzymes, and cathepsins) in the context of aneurysm pathogenesis. An update of clinical trials focusing on therapeutic strategies to slow abdominal aortic aneurysm growth and efforts underway to develop effective pharmacological treatments is also provided.
Collapse
Affiliation(s)
- Bo Liu
- University of Wisconsin, Madison, Department of Surgery, Madison Wisconsin
| | - David J Granville
- International Collaboration on Repair Discoveries Centre and University of British Columbia Centre for Heart Lung Innovation, Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | - Jonathan Golledge
- The Queensland Research Centre for Peripheral Vascular Disease, College of Medicine and Dentistry, James Cook University, Department of Vascular and Endovascular Surgery, Townsville Hospital and Health Services, Townsville, Queensland, Australia
| | - Zamaneh Kassiri
- University of Alberta, Department of Physiology, Cardiovascular Research Center, Faculty of Medicine and Dentistry, Edmonton, Alberta, Canada
| |
Collapse
|
39
|
Role of Endothelial ADAM17 in Early Vascular Changes Associated with Diabetic Retinopathy. J Clin Med 2020; 9:jcm9020400. [PMID: 32024241 PMCID: PMC7073770 DOI: 10.3390/jcm9020400] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2019] [Revised: 01/25/2020] [Accepted: 01/30/2020] [Indexed: 02/06/2023] Open
Abstract
ADAM17, a disintegrin and metalloproteinase 17, is a transmembrane metalloproteinase that regulates bioavailability of multiple membrane-bound proteins via ectodomain shedding. ADAM17 activity was shown to contribute to a number of vascular pathologies, but its role in the context of diabetic retinopathy (DR) is not determined. We found that expression and enzymatic activity of ADAM17 are upregulated in human diabetic postmortem retinas and a mouse model of streptozotocin-induced diabetes. To further investigate the contribution of ADAM17 to vascular alterations associated with DR, we used human retinal endothelial cells (HREC) treated with ADAM17 neutralizing antibodies and exposed to glucidic stress and streptozotocin-induced endothelial ADAM17 knockout mice. Evaluation of vascular permeability, vascular inflammation, and oxidative stress was performed. Loss of ADAM17 in endothelial cells markedly reduced oxidative stress evidenced by decreased levels of superoxide, 3-nitrotyrosine, and 4-hydroxynonenal and decreased leukocyte-endothelium adhesive interactions in vivo and in vitro. Reduced leukostasis was associated with decreased vascular permeability and was accompanied by downregulation of intercellular adhesion molecule-1 expression. Reduction in oxidative stress in HREC was associated with downregulation of NAD(P)H oxidase 4 (Nox4) expression. Our data suggest a role for endothelial ADAM17 in DR pathogenesis and identify ADAM17 as a potential new therapeutic target for DR.
Collapse
|
40
|
Liu Y, Wang X, Wang H, Hu T. Identification of key genes and pathways in abdominal aortic aneurysm by integrated bioinformatics analysis. J Int Med Res 2019; 48:300060519894437. [PMID: 31885343 PMCID: PMC7783286 DOI: 10.1177/0300060519894437] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Objectives To identify key genes associated with abdominal aortic aneurysm (AAA) by
integrating a microarray profile and a single-cell RNA-seq dataset. Methods The microarray profile of GSE7084 and the single-cell RNA-seq dataset were
obtained from the Gene Express Omnibus database. Differentially expressed
genes (DEGs) were chosen using the R package and annotated by Gene Ontology
and Kyoto Encyclopedia of Genes and Genomics analysis. The hub genes were
identified based on their degrees of interaction in the protein-protein
interaction (PPI) network. Expression of hub genes was determined using
single-cell RNA-seq analysis. Results In total, 507 upregulated and 842 downregulated DEGs were identified and
associated with AAA. The upregulated DEGs were enriched into 9 biological
processes and 10 biological pathways, which were closely involved in the
pathogenesis and progression of AAA. Based on the PPI network, we focused on
six hub genes, four of which were novel target genes compared with the known
aneurysm gene database. Using single-cell RNA-seq analysis, we explored the
four genes expressed in vascular cells of AAA: CANX,
CD44, DAXX, and
STAT1. Conclusions We identified key genes that may provide insight into the mechanism of AAA
pathogenesis and progression and that have potential to be therapeutic
targets.
Collapse
Affiliation(s)
- Yihai Liu
- Department of Cardiology, The
Affiliated Huaian No. 1 People’s Hospital of
Nanjing
Medical University, Huaian, China
| | - Xixi Wang
- Department of Neurology, Affiliated
Shanghai First People’s Hospital of
Nanjing
Medical University, Nanjing, China
| | - Hongye Wang
- Department of Cardiology, The
Affiliated Huaian No. 1 People’s Hospital of
Nanjing
Medical University, Huaian, China
| | - Tingting Hu
- Department of Cardiology, The
Affiliated Huaian No. 1 People’s Hospital of
Nanjing
Medical University, Huaian, China
- Tingting Hu, Department of Cardiology, the
Affiliated Huaian No. 1 People’s Hospital of Nanjing Medical University, Beijing
West Road 6, Huaian 223001, China.
| |
Collapse
|
41
|
Jana S, Hu M, Shen M, Kassiri Z. Extracellular matrix, regional heterogeneity of the aorta, and aortic aneurysm. Exp Mol Med 2019; 51:1-15. [PMID: 31857579 PMCID: PMC6923362 DOI: 10.1038/s12276-019-0286-3] [Citation(s) in RCA: 133] [Impact Index Per Article: 22.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2019] [Accepted: 06/17/2019] [Indexed: 12/12/2022] Open
Abstract
Aortic aneurysm is an asymptomatic disease with dire outcomes if undiagnosed. Aortic aneurysm rupture is a significant cause of death worldwide. To date, surgical repair or endovascular repair (EVAR) is the only effective treatment for aortic aneurysm, as no pharmacological treatment has been found effective. Aortic aneurysm, a focal dilation of the aorta, can be formed in the thoracic (TAA) or the abdominal (AAA) region; however, our understanding as to what determines the site of aneurysm formation remains quite limited. The extracellular matrix (ECM) is the noncellular component of the aortic wall, that in addition to providing structural support, regulates bioavailability of an array of growth factors and cytokines, thereby influencing cell function and behavior that ultimately determine physiological or pathological remodeling of the aortic wall. Here, we provide an overview of the ECM proteins that have been reported to be involved in aortic aneurysm formation in humans or animal models, and the experimental models for TAA and AAA and the link to ECM manipulations. We also provide a comparative analysis, where data available, between TAA and AAA, and how aberrant ECM proteolysis versus disrupted synthesis may determine the site of aneurysm formation. A review of aneurysm formation, swelling in blood vessel, in the aorta, examines distinctions between two forms of the condition and the role of proteins in the extracellular matrix which surrounds cells of the arterial wall. Rupture of aneurysms in the aorta, the body’s main artery, is a major cause of death. Researchers led by Zamaneh Kassiri at the University of Alberta, Edmonton, Canada, emphasize that aneurysms in the thoracic and abdominal regions of the aorta are distinct conditions with crucial differences in their causes. Disrupted production and assembly of the extracellular matrix and its proteins may underlie thoracic aneurysm formation. Factors triggering the degradation of extracellular matrix proteins may be more significant in abdominal aneurysms. Understanding the differing molecular mechanisms involved could help address the current lack of effective drug treatments for these dangerous conditions.
Collapse
Affiliation(s)
- Sayantan Jana
- Department of Physiology, Cardiovascular Research Center, University of Alberta, Edmonton, AB, Canada
| | - Mei Hu
- Department of Physiology, Cardiovascular Research Center, University of Alberta, Edmonton, AB, Canada
| | - Mengcheng Shen
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA, USA
| | - Zamaneh Kassiri
- Department of Physiology, Cardiovascular Research Center, University of Alberta, Edmonton, AB, Canada.
| |
Collapse
|
42
|
Deletion of interleukin-18 attenuates abdominal aortic aneurysm formation. Atherosclerosis 2019; 289:14-20. [PMID: 31445353 DOI: 10.1016/j.atherosclerosis.2019.08.003] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/15/2018] [Revised: 07/04/2019] [Accepted: 08/16/2019] [Indexed: 12/19/2022]
Abstract
BACKGROUND AND AIMS Abdominal aortic aneurysm (AAA) is a common disease; however, its exact pathogenesis remains unknown, and no specific medical therapies are available. Interleukin (IL)-18 plays a crucial role in atherosclerotic plaque destabilization and is a strong predictor of cardiovascular death. Here, we investigated the role of IL-18 in AAA pathogenesis using an experimental mouse model. METHODS AND RESULTS After infusion of angiotensin II (Ang II) for 4 weeks and β-aminopropionitrile (BAPN) for 2 weeks, 58% of C57/6J wild-type (WT) mice developed AAA associated with enhanced expression of IL-18; however, disease incidence was significantly lower in IL-18-/- mice than in WT mice (p < 0.01), although no significant difference was found in systolic blood pressure between WT mice and IL-18-/- mice in this model. Additionally, IL-18 deletion significantly attenuated Ang II/BAPN-induced macrophage infiltration, macrophage polarization into inflammatory M1 phenotype, and matrix metalloproteinase (MMP) activation in abdominal aortas, which is associated with reduced expression of osteopontin (OPN). CONCLUSIONS These findings indicate that IL-18 plays an important role in the development of AAA by enhancing OPN expression, macrophage recruitment, and MMP activation. Moreover, IL-18 represents a previously unrecognized therapeutic target for the prevention of AAA formation.
Collapse
|
43
|
Zhong S, Khalil RA. A Disintegrin and Metalloproteinase (ADAM) and ADAM with thrombospondin motifs (ADAMTS) family in vascular biology and disease. Biochem Pharmacol 2019; 164:188-204. [PMID: 30905657 DOI: 10.1016/j.bcp.2019.03.033] [Citation(s) in RCA: 59] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2019] [Accepted: 03/20/2019] [Indexed: 12/11/2022]
Abstract
A Disintegrin and Metalloproteinase (ADAM) is a family of proteolytic enzymes that possess sheddase function and regulate shedding of membrane-bound proteins, growth factors, cytokines, ligands and receptors. Typically, ADAMs have a pro-domain, and a metalloproteinase, disintegrin, cysteine-rich and a characteristic transmembrane domain. Most ADAMs are activated by proprotein convertases, but can also be regulated by G-protein coupled receptor agonists, Ca2+ ionophores and protein kinase C activators. A Disintegrin and Metalloproteinase with Thrombospondin Motifs (ADAMTS) is a family of secreted enzymes closely related to ADAMs. Like ADAMs, ADAMTS members have a pro-domain, and a metalloproteinase, disintegrin, and cysteine-rich domain, but they lack a transmembrane domain and instead have characteristic thrombospondin motifs. Activated ADAMs perform several functions and participate in multiple cardiovascular processes including vascular smooth muscle cell proliferation and migration, angiogenesis, vascular cell apoptosis, cell survival, tissue repair, and wound healing. ADAMs may also be involved in pathological conditions and cardiovascular diseases such as atherosclerosis, hypertension, aneurysm, coronary artery disease, myocardial infarction and heart failure. Like ADAMs, ADAMTS have a wide-spectrum role in vascular biology and cardiovascular pathophysiology. ADAMs and ADAMTS activity is naturally controlled by endogenous inhibitors such as tissue inhibitors of metalloproteinases (TIMPs), and their activity can also be suppressed by synthetic small molecule inhibitors. ADAMs and ADAMTS can serve as important diagnostic biomarkers and potential therapeutic targets for cardiovascular disorders. Natural and synthetic inhibitors of ADAMs and ADAMTS could be potential therapeutic tools for the management of cardiovascular diseases.
Collapse
Affiliation(s)
- Sheng Zhong
- Vascular Surgery Research Laboratories, Division of Vascular and Endovascular Surgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Raouf A Khalil
- Vascular Surgery Research Laboratories, Division of Vascular and Endovascular Surgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
44
|
Forrester SJ, Booz GW, Sigmund CD, Coffman TM, Kawai T, Rizzo V, Scalia R, Eguchi S. Angiotensin II Signal Transduction: An Update on Mechanisms of Physiology and Pathophysiology. Physiol Rev 2018; 98:1627-1738. [PMID: 29873596 DOI: 10.1152/physrev.00038.2017] [Citation(s) in RCA: 718] [Impact Index Per Article: 102.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
The renin-angiotensin-aldosterone system plays crucial roles in cardiovascular physiology and pathophysiology. However, many of the signaling mechanisms have been unclear. The angiotensin II (ANG II) type 1 receptor (AT1R) is believed to mediate most functions of ANG II in the system. AT1R utilizes various signal transduction cascades causing hypertension, cardiovascular remodeling, and end organ damage. Moreover, functional cross-talk between AT1R signaling pathways and other signaling pathways have been recognized. Accumulating evidence reveals the complexity of ANG II signal transduction in pathophysiology of the vasculature, heart, kidney, and brain, as well as several pathophysiological features, including inflammation, metabolic dysfunction, and aging. In this review, we provide a comprehensive update of the ANG II receptor signaling events and their functional significances for potential translation into therapeutic strategies. AT1R remains central to the system in mediating physiological and pathophysiological functions of ANG II, and participation of specific signaling pathways becomes much clearer. There are still certain limitations and many controversies, and several noteworthy new concepts require further support. However, it is expected that rigorous translational research of the ANG II signaling pathways including those in large animals and humans will contribute to establishing effective new therapies against various diseases.
Collapse
Affiliation(s)
- Steven J Forrester
- Cardiovascular Research Center, Lewis Katz School of Medicine at Temple University , Philadelphia, Pennsylvania ; Department of Pharmacology and Toxicology, School of Medicine, University of Mississippi Medical Center , Jackson, Mississippi ; Department of Pharmacology, Center for Hypertension Research, Roy J. and Lucille A. Carver College of Medicine, University of Iowa , Iowa City, Iowa ; and Duke-NUS, Singapore and Department of Medicine, Duke University Medical Center , Durham, North Carolina
| | - George W Booz
- Cardiovascular Research Center, Lewis Katz School of Medicine at Temple University , Philadelphia, Pennsylvania ; Department of Pharmacology and Toxicology, School of Medicine, University of Mississippi Medical Center , Jackson, Mississippi ; Department of Pharmacology, Center for Hypertension Research, Roy J. and Lucille A. Carver College of Medicine, University of Iowa , Iowa City, Iowa ; and Duke-NUS, Singapore and Department of Medicine, Duke University Medical Center , Durham, North Carolina
| | - Curt D Sigmund
- Cardiovascular Research Center, Lewis Katz School of Medicine at Temple University , Philadelphia, Pennsylvania ; Department of Pharmacology and Toxicology, School of Medicine, University of Mississippi Medical Center , Jackson, Mississippi ; Department of Pharmacology, Center for Hypertension Research, Roy J. and Lucille A. Carver College of Medicine, University of Iowa , Iowa City, Iowa ; and Duke-NUS, Singapore and Department of Medicine, Duke University Medical Center , Durham, North Carolina
| | - Thomas M Coffman
- Cardiovascular Research Center, Lewis Katz School of Medicine at Temple University , Philadelphia, Pennsylvania ; Department of Pharmacology and Toxicology, School of Medicine, University of Mississippi Medical Center , Jackson, Mississippi ; Department of Pharmacology, Center for Hypertension Research, Roy J. and Lucille A. Carver College of Medicine, University of Iowa , Iowa City, Iowa ; and Duke-NUS, Singapore and Department of Medicine, Duke University Medical Center , Durham, North Carolina
| | - Tatsuo Kawai
- Cardiovascular Research Center, Lewis Katz School of Medicine at Temple University , Philadelphia, Pennsylvania ; Department of Pharmacology and Toxicology, School of Medicine, University of Mississippi Medical Center , Jackson, Mississippi ; Department of Pharmacology, Center for Hypertension Research, Roy J. and Lucille A. Carver College of Medicine, University of Iowa , Iowa City, Iowa ; and Duke-NUS, Singapore and Department of Medicine, Duke University Medical Center , Durham, North Carolina
| | - Victor Rizzo
- Cardiovascular Research Center, Lewis Katz School of Medicine at Temple University , Philadelphia, Pennsylvania ; Department of Pharmacology and Toxicology, School of Medicine, University of Mississippi Medical Center , Jackson, Mississippi ; Department of Pharmacology, Center for Hypertension Research, Roy J. and Lucille A. Carver College of Medicine, University of Iowa , Iowa City, Iowa ; and Duke-NUS, Singapore and Department of Medicine, Duke University Medical Center , Durham, North Carolina
| | - Rosario Scalia
- Cardiovascular Research Center, Lewis Katz School of Medicine at Temple University , Philadelphia, Pennsylvania ; Department of Pharmacology and Toxicology, School of Medicine, University of Mississippi Medical Center , Jackson, Mississippi ; Department of Pharmacology, Center for Hypertension Research, Roy J. and Lucille A. Carver College of Medicine, University of Iowa , Iowa City, Iowa ; and Duke-NUS, Singapore and Department of Medicine, Duke University Medical Center , Durham, North Carolina
| | - Satoru Eguchi
- Cardiovascular Research Center, Lewis Katz School of Medicine at Temple University , Philadelphia, Pennsylvania ; Department of Pharmacology and Toxicology, School of Medicine, University of Mississippi Medical Center , Jackson, Mississippi ; Department of Pharmacology, Center for Hypertension Research, Roy J. and Lucille A. Carver College of Medicine, University of Iowa , Iowa City, Iowa ; and Duke-NUS, Singapore and Department of Medicine, Duke University Medical Center , Durham, North Carolina
| |
Collapse
|
45
|
Shen M, Hu M, Fedak PWM, Oudit GY, Kassiri Z. Cell-Specific Functions of ADAM17 Regulate the Progression of Thoracic Aortic Aneurysm. Circ Res 2018; 123:372-388. [PMID: 29930147 DOI: 10.1161/circresaha.118.313181] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/05/2018] [Revised: 06/03/2018] [Accepted: 06/20/2018] [Indexed: 12/31/2022]
Abstract
RATIONALE ADAM17 (a disintegrin and metalloproteinase-17) is a membrane-bound enzyme that regulates bioavailability of multiple transmembrane proteins by proteolytic processing. ADAM17 has been linked to several pathologies, but its role in thoracic aortic aneurysm (TAA) has not been determined. OBJECTIVE The objective of this study was to explore the cell-specific functions of vascular ADAM17 in the pathogenesis and progression of TAA. METHODS AND RESULTS In aneurysmal thoracic aorta from patients, ADAM17 was increased in tunica media and intima. To determine the function of ADAM17 in the major cells types within these regions, we generated mice lacking ADAM17 in smooth muscle cells (SMC; Adam17f/f/Sm22Cre/+ ) or endothelial cells (Adam17f/f/Tie2Cre/+ ). ADAM17 deficiency in either cell type was sufficient to suppress TAA dilation markedly and adverse remodeling in males and females (in vivo) although through different mechanisms. ADAM17 deficiency in SMCs prevented the contractile-to-synthetic phenotypic switching in these cells after TAA induction, preventing perivascular fibrosis, inflammation, and adverse aortic remodeling. Loss of ADAM17 in endothelial cells protected the integrity of the intimal barrier by preserving the adherens junction (vascular endothelial-cadherin) and tight junctions (junctional adhesion molecule-A and claudin). In vitro studies on primary mouse thoracic SMCs and human primary aortic SMCs and endothelial cells (±ADAM17 small interfering RNA) confirmed the cell-specific functions of ADAM17 and demonstrated the cross-species validity of these findings. To determine the impact of ADAM17 inhibition in treating TAA, we used an ADAM17-selective inhibitor (PF-548) before or 3 days after TAA induction. In both cases, ADAM17 inhibition prevented progression of aneurysmal growth. CONCLUSIONS We have identified distinct cell-specific functions of ADAM17 in TAA progression, promoting pathological remodeling of SMC and impairing integrity of the intimal endothelial cell barrier. The dual impact of ADAM17 deficiency (or inhibition) in protecting 2 major cell types in the aortic wall highlights the unique position of this proteinase as a critical treatment target for TAA.
Collapse
Affiliation(s)
- Mengcheng Shen
- From the Department of Physiology (M.S., M.H., Z.K.).,Faculty of Medicine and Dentistry (M.S., M.H., G.Y.O., Z.K.)
| | - Mei Hu
- From the Department of Physiology (M.S., M.H., Z.K.).,Faculty of Medicine and Dentistry (M.S., M.H., G.Y.O., Z.K.)
| | - Paul W M Fedak
- Cardiovascular Research Centre, Mazankowski Alberta Heart Institute, University of Alberta, Edmonton, Canada; Section of Cardiac Surgery, Department of Cardiac Sciences, University of Calgary, Libin Cardiovascular Institute of Alberta, Canada (P.W.M.F.).,Division of Cardiac Surgery, Bluhm Cardiovascular Institute, Northwestern Memorial Hospital, Chicago, IL (P.W.M.F.)
| | - Gavin Y Oudit
- Department of Medicine (G.Y.O.).,Faculty of Medicine and Dentistry (M.S., M.H., G.Y.O., Z.K.)
| | - Zamaneh Kassiri
- From the Department of Physiology (M.S., M.H., Z.K.) .,Faculty of Medicine and Dentistry (M.S., M.H., G.Y.O., Z.K.)
| |
Collapse
|
46
|
Eguchi S, Kawai T, Scalia R, Rizzo V. Understanding Angiotensin II Type 1 Receptor Signaling in Vascular Pathophysiology. Hypertension 2018; 71:804-810. [PMID: 29581215 DOI: 10.1161/hypertensionaha.118.10266] [Citation(s) in RCA: 138] [Impact Index Per Article: 19.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Affiliation(s)
- Satoru Eguchi
- From the Cardiovascular Research Center, Lewis Katz School of Medicine, Temple University, Philadelphia, PA.
| | - Tatsuo Kawai
- From the Cardiovascular Research Center, Lewis Katz School of Medicine, Temple University, Philadelphia, PA
| | - Rosario Scalia
- From the Cardiovascular Research Center, Lewis Katz School of Medicine, Temple University, Philadelphia, PA
| | - Victor Rizzo
- From the Cardiovascular Research Center, Lewis Katz School of Medicine, Temple University, Philadelphia, PA
| |
Collapse
|
47
|
Mathieu S, El Khoury N, Rivard K, Paradis P, Nemer M, Fiset C. Angiotensin II Overstimulation Leads to an Increased Susceptibility to Dilated Cardiomyopathy and Higher Mortality in Female Mice. Sci Rep 2018; 8:952. [PMID: 29343862 PMCID: PMC5772611 DOI: 10.1038/s41598-018-19436-5] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2017] [Accepted: 11/15/2017] [Indexed: 11/09/2022] Open
Abstract
Heart failure (HF) is associated with high mortality and affects men and women differently. The underlying mechanisms for these sex-related differences remain largely unexplored. Accordingly, using mice with cardiac-specific overexpression of the angiotensin II (ANGII) type 1 receptor (AT1R), we explored male-female differences in the manifestations of hypertrophy and HF. AT1R mice of both sexes feature electrical and Ca2+ handling alterations, systolic dysfunction, hypertrophy and develop HF. However, females had much higher mortality (21.0%) rate than males (5.5%). In females, AT1R stimulation leads to more pronounced eccentric hypertrophy (larger increase in LV mass/body weight ratio [+31%], in cell length [+27%], in LV internal end-diastolic [LVIDd, +34%] and systolic [LVIDs, +67%] diameter) and dilation (larger decrease in LV posterior wall thickness, +17%) than males. In addition, in female AT1R mice the cytosolic Ca2+ extrusion mechanisms were more severely compromised and were associated with a specific increased in Ca2+ sparks (by 187%) and evidence of SR Ca2+ leak. Altogether, these results suggest that female AT1R mice have more severe eccentric hypertrophy, dysfunction and compromised Ca2+ dynamics. These findings indicate that females are more susceptible to the adverse effects of AT1R stimulation than males favouring the development of HF and increased mortality.
Collapse
Affiliation(s)
- Sophie Mathieu
- Research Center, Montreal Heart Institute, 5000 Bélanger, Montréal, Québec, Canada.,Faculty of Pharmacy, Université de Montréal, Montréal, Québec, Canada
| | - Nabil El Khoury
- Research Center, Montreal Heart Institute, 5000 Bélanger, Montréal, Québec, Canada.,Department of Pharmacology and Physiology, Faculty of Medicine, Université de Montréal, Montréal, Québec, Canada
| | - Katy Rivard
- Research Center, Montreal Heart Institute, 5000 Bélanger, Montréal, Québec, Canada.,Faculty of Pharmacy, Université de Montréal, Montréal, Québec, Canada
| | - Pierre Paradis
- Lady Davis Institute, McGill University, Montreal, Québec, Canada
| | - Mona Nemer
- Ottawa University, Ottawa, Ontario, Canada
| | - Céline Fiset
- Research Center, Montreal Heart Institute, 5000 Bélanger, Montréal, Québec, Canada. .,Faculty of Pharmacy, Université de Montréal, Montréal, Québec, Canada.
| |
Collapse
|