1
|
Daugherty A, Milewicz DM, Dichek DA, Ghaghada KB, Humphrey JD, LeMaire SA, Li Y, Mallat Z, Saeys Y, Sawada H, Shen YH, Suzuki T, Zhou (周桢) Z. Recommendations for Design, Execution, and Reporting of Studies on Experimental Thoracic Aortopathy in Preclinical Models. Arterioscler Thromb Vasc Biol 2025; 45:609-631. [PMID: 40079138 PMCID: PMC12018150 DOI: 10.1161/atvbaha.124.320259] [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] [Indexed: 03/14/2025]
Abstract
There is a recent dramatic increase in research on thoracic aortic diseases that includes aneurysms, dissections, and rupture. Experimental studies predominantly use mice in which aortopathy is induced by chemical interventions, genetic manipulations, or both. Many parameters should be deliberated in experimental design in concert with multiple considerations when providing dimensional data and characterization of aortic tissues. The purpose of this review is to provide recommendations on guidance in (1) the selection of a mouse model and experimental conditions for the study, (2) parameters for standardizing detection and measurements of aortic diseases, (3) meaningful interpretation of characteristics of diseased aortic tissue, and (4) reporting standards that include rigor and transparency.
Collapse
Affiliation(s)
- Alan Daugherty
- Saha Cardiovascular Research Center, Saha Aortic Center, Department of Physiology, University of Kentucky, KY, USA
| | - Dianna M. Milewicz
- Division of Medical Genetics, Department of Internal Medicine, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - David A. Dichek
- Division of Cardiology, Department of Medicine, University of Washington, Seattle, WA, USA
| | - Ketan B. Ghaghada
- Department of Radiology, Texas Children’s Hospital, and Department of Radiology, Baylor College of Medicine Houston, TX, USA
| | - Jay D. Humphrey
- Department of Biomedical Engineering, Yale University, New Haven, CT, USA
| | - Scott A. LeMaire
- Heart & Vascular Institute, Geisinger Health System, Danville, PA, USA
- Division of Cardiothoracic Surgery, Michael E. DeBakey Department of Surgery and Cardiovascular Research Institute, Baylor College of Medicine, Houston, TX, USA
| | - Yanming Li
- Division of Cardiothoracic Surgery, Michael E. DeBakey Department of Surgery and Cardiovascular Research Institute, Baylor College of Medicine, Houston, TX, USA
| | - Ziad Mallat
- Division of Cardiorespiratory Medicine, Department of Medicine, Victor Phillip Dahdaleh Heart and Lung Research Institute, University of Cambridge, Cambridge, UK; Unversité de Paris, Inserm U970, Paris Cardiovascular Research Centre, Paris, France
| | - Yvan Saeys
- Data Mining and Modelling for Biomedicine, VIB Center for Inflammation Research, Department of Applied Mathematics, Computer Science and Statistics, Ghent University Ghent, Belgium
| | - Hisashi Sawada
- Saha Cardiovascular Research Center, Saha Aortic Center, Department of Physiology, University of Kentucky, KY, USA
| | - Ying H. Shen
- Division of Cardiothoracic Surgery, Michael E. DeBakey Department of Surgery and Cardiovascular Research Institute, Baylor College of Medicine, Houston, TX, USA
| | - Toru Suzuki
- Department of Cardiovascular Sciences, University of Leicester and NIHR Leicester Biomedical Research Centre, Leicester, UK and Institute of Medical Science, University of Tokyo, Tokyo, Japan
| | - Zhen Zhou (周桢)
- Division of Medical Genetics, Department of Internal Medicine, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, USA
| |
Collapse
|
2
|
Yamamoto K, Scilabra SD, Bonelli S, Jensen A, Scavenius C, Enghild JJ, Strickland DK. Novel insights into the multifaceted and tissue-specific roles of the endocytic receptor LRP1. J Biol Chem 2024; 300:107521. [PMID: 38950861 PMCID: PMC11325810 DOI: 10.1016/j.jbc.2024.107521] [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: 03/13/2024] [Revised: 06/22/2024] [Accepted: 06/26/2024] [Indexed: 07/03/2024] Open
Abstract
Receptor-mediated endocytosis provides a mechanism for the selective uptake of specific molecules thereby controlling the composition of the extracellular environment and biological processes. The low-density lipoprotein receptor-related protein 1 (LRP1) is a widely expressed endocytic receptor that regulates cellular events by modulating the levels of numerous extracellular molecules via rapid endocytic removal. LRP1 also participates in signalling pathways through this modulation as well as in the interaction with membrane receptors and cytoplasmic adaptor proteins. LRP1 SNPs are associated with several diseases and conditions such as migraines, aortic aneurysms, cardiopulmonary dysfunction, corneal clouding, and bone dysmorphology and mineral density. Studies using Lrp1 KO mice revealed a critical, nonredundant and tissue-specific role of LRP1 in regulating various physiological events. However, exactly how LRP1 functions to regulate so many distinct and specific processes is still not fully clear. Our recent proteomics studies have identified more than 300 secreted proteins that either directly interact with LRP1 or are modulated by LRP1 in various tissues. This review will highlight the remarkable ability of this receptor to regulate secreted molecules in a tissue-specific manner and discuss potential mechanisms underpinning such specificity. Uncovering the depth of these "hidden" specific interactions modulated by LRP1 will provide novel insights into a dynamic and complex extracellular environment that is involved in diverse biological and pathological processes.
Collapse
Affiliation(s)
- Kazuhiro Yamamoto
- Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool, United Kingdom.
| | - Simone D Scilabra
- Proteomics Group of Ri.MED Foundation, Research Department IRCCS ISMETT, Palermo, Italy
| | - Simone Bonelli
- Proteomics Group of Ri.MED Foundation, Research Department IRCCS ISMETT, Palermo, Italy; Department of Biological, Chemical and Pharmaceutical Sciences and Technologies, University of Palermo, Palermo, Italy
| | - Anders Jensen
- Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool, United Kingdom
| | - Carsten Scavenius
- Department of Molecular Biology and Genetics, Aarhus University, Aarhus, Denmark
| | - Jan J Enghild
- Department of Molecular Biology and Genetics, Aarhus University, Aarhus, Denmark
| | - Dudley K Strickland
- Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
3
|
Daugherty A, Sawada H, Sheppard MB, Lu HS. Angiotensinogen as a Therapeutic Target for Cardiovascular and Metabolic Diseases. Arterioscler Thromb Vasc Biol 2024; 44:1021-1030. [PMID: 38572647 PMCID: PMC11225801 DOI: 10.1161/atvbaha.124.318374] [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] [Indexed: 04/05/2024]
Abstract
AGT (angiotensinogen) is the unique precursor for the generation of all the peptides of the renin-angiotensin system, but it has received relatively scant attention compared to many other renin-angiotensin system components. Focus on AGT has increased recently, particularly with the evolution of drugs to target the synthesis of the protein. AGT is a noninhibitory serpin that has several conserved domains in addition to the angiotensin II sequences at the N terminus. Increased study is needed on the structure-function relationship to resolve many unknowns regarding AGT metabolism. Constitutive whole-body genetic deletion of Agt in mice leads to multiple developmental defects creating a challenge to use these mice for mechanistic studies. This has been overcome by creating Agt-floxed mice to enable the development of cell-specific deficiencies that have provided considerable insight into a range of cardiovascular and associated diseases. This has been augmented by the recent development of pharmacological approaches targeting hepatocytes in humans to promote protracted inhibition of AGT synthesis. Genetic deletion or pharmacological inhibition of Agt has been demonstrated to be beneficial in a spectrum of diseases experimentally, including hypertension, atherosclerosis, aortic and superior mesenteric artery aneurysms, myocardial dysfunction, and hepatic steatosis. This review summarizes the findings of recent studies utilizing AGT manipulation as a therapeutic approach.
Collapse
Affiliation(s)
- 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
| | - 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
| | - Mary B. Sheppard
- 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
- Department of Family and Community Medicine, University of Kentucky, Lexington, KY
- Department of Surgery, 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
| |
Collapse
|
4
|
Pedroza AJ, Cheng P, Dalal AR, Baeumler K, Kino A, Tognozzi E, Shad R, Yokoyama N, Nakamura K, Mitchel O, Hiesinger W, MacFarlane EG, Fleischmann D, Woo YJ, Quertermous T, Fischbein MP. Early clinical outcomes and molecular smooth muscle cell phenotyping using a prophylactic aortic arch replacement strategy in Loeys-Dietz syndrome. J Thorac Cardiovasc Surg 2023; 166:e332-e376. [PMID: 37500053 PMCID: PMC11888900 DOI: 10.1016/j.jtcvs.2023.07.023] [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: 03/23/2023] [Revised: 06/12/2023] [Accepted: 07/11/2023] [Indexed: 07/29/2023]
Abstract
OBJECTIVES Patients with Loeys-Dietz syndrome demonstrate a heightened risk of distal thoracic aortic events after valve-sparing aortic root replacement. This study assesses the clinical risks and hemodynamic consequences of a prophylactic aortic arch replacement strategy in Loeys-Dietz syndrome and characterizes smooth muscle cell phenotype in Loeys-Dietz syndrome aneurysmal and normal-sized downstream aorta. METHODS Patients with genetically confirmed Loeys-Dietz syndrome (n = 8) underwent prophylactic aortic arch replacement during valve-sparing aortic root replacement. Four-dimensional flow magnetic resonance imaging studies were performed in 4 patients with Loeys-Dietz syndrome (valve-sparing aortic root replacement + arch) and compared with patients with contemporary Marfan syndrome (valve-sparing aortic root replacement only, n = 5) and control patients (without aortopathy, n = 5). Aortic tissues from 4 patients with Loeys-Dietz syndrome and 2 organ donors were processed for anatomically segmented single-cell RNA sequencing and histologic assessment. RESULTS Patients with Loeys-Dietz syndrome valve-sparing aortic root replacement + arch had no deaths, major morbidity, or aortic events in a median of 2 years follow-up. Four-dimensional magnetic resonance imaging demonstrated altered flow parameters in patients with postoperative aortopathy relative to controls, but no clear deleterious changes due to arch replacement. Integrated analysis of aortic single-cell RNA sequencing data (>49,000 cells) identified a continuum of abnormal smooth muscle cell phenotypic modulation in Loeys-Dietz syndrome defined by reduced contractility and enriched extracellular matrix synthesis, adhesion receptors, and transforming growth factor-beta signaling. These modulated smooth muscle cells populated the Loeys-Dietz syndrome tunica media with gradually reduced density from the overtly aneurysmal root to the nondilated arch. CONCLUSIONS Patients with Loeys-Dietz syndrome demonstrated excellent surgical outcomes without overt downstream flow or shear stress disturbances after concomitant valve-sparing aortic root replacement + arch operations. Abnormal smooth muscle cell-mediated aortic remodeling occurs within the normal diameter, clinically at-risk Loeys-Dietz syndrome arch segment. These initial clinical and pathophysiologic findings support concomitant arch replacement in Loeys-Dietz syndrome.
Collapse
Affiliation(s)
- Albert J Pedroza
- Department of Cardiothoracic Surgery, Stanford University School of Medicine, Stanford, Calif
| | - Paul Cheng
- Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, Calif
| | - Alex R Dalal
- Department of Cardiothoracic Surgery, Stanford University School of Medicine, Stanford, Calif
| | - Kathrin Baeumler
- Department of Radiology, Stanford University School of Medicine, Stanford, Calif
| | - Aya Kino
- Department of Radiology, Stanford University School of Medicine, Stanford, Calif
| | - Emily Tognozzi
- Department of Cardiothoracic Surgery, Stanford University School of Medicine, Stanford, Calif
| | - Rohan Shad
- Department of Cardiothoracic Surgery, Stanford University School of Medicine, Stanford, Calif
| | - Nobu Yokoyama
- Department of Cardiothoracic Surgery, Stanford University School of Medicine, Stanford, Calif
| | - Ken Nakamura
- Department of Cardiothoracic Surgery, Stanford University School of Medicine, Stanford, Calif
| | - Olivia Mitchel
- Department of Cardiothoracic Surgery, Stanford University School of Medicine, Stanford, Calif
| | - William Hiesinger
- Department of Cardiothoracic Surgery, Stanford University School of Medicine, Stanford, Calif
| | - Elena Gallo MacFarlane
- Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, Md
| | - Dominik Fleischmann
- Department of Radiology, Stanford University School of Medicine, Stanford, Calif
| | - Y Joseph Woo
- Department of Cardiothoracic Surgery, Stanford University School of Medicine, Stanford, Calif
| | - Thomas Quertermous
- Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, Calif
| | - Michael P Fischbein
- Department of Cardiothoracic Surgery, Stanford University School of Medicine, Stanford, Calif.
| |
Collapse
|
5
|
Klarin D, Devineni P, Sendamarai AK, Angueira AR, Graham SE, Shen YH, Levin MG, Pirruccello JP, Surakka I, Karnam PR, Roychowdhury T, Li Y, Wang M, Aragam KG, Paruchuri K, Zuber V, Shakt GE, Tsao NL, Judy RL, Vy HMT, Verma SS, Rader DJ, Do R, Bavaria JE, Nadkarni GN, Ritchie MD, Burgess S, Guo DC, Ellinor PT, LeMaire SA, Milewicz DM, Willer CJ, Natarajan P, Tsao PS, Pyarajan S, Damrauer SM. Genome-wide association study of thoracic aortic aneurysm and dissection in the Million Veteran Program. Nat Genet 2023; 55:1106-1115. [PMID: 37308786 PMCID: PMC10335930 DOI: 10.1038/s41588-023-01420-z] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Accepted: 05/05/2023] [Indexed: 06/14/2023]
Abstract
The current understanding of the genetic determinants of thoracic aortic aneurysms and dissections (TAAD) has largely been informed through studies of rare, Mendelian forms of disease. Here, we conducted a genome-wide association study (GWAS) of TAAD, testing ~25 million DNA sequence variants in 8,626 participants with and 453,043 participants without TAAD in the Million Veteran Program, with replication in an independent sample of 4,459 individuals with and 512,463 without TAAD from six cohorts. We identified 21 TAAD risk loci, 17 of which have not been previously reported. We leverage multiple downstream analytic methods to identify causal TAAD risk genes and cell types and provide human genetic evidence that TAAD is a non-atherosclerotic aortic disorder distinct from other forms of vascular disease. Our results demonstrate that the genetic architecture of TAAD mirrors that of other complex traits and that it is not solely inherited through protein-altering variants of large effect size.
Collapse
Affiliation(s)
- Derek Klarin
- Veterans Affairs (VA) Palo Alto Healthcare System, Palo Alto, CA, USA.
- Department of Surgery, Stanford University School of Medicine, Palo Alto, CA, USA.
| | - Poornima Devineni
- Center for Data and Computational Sciences, VA Boston Healthcare System, Boston, MA, USA
| | - Anoop K Sendamarai
- Center for Data and Computational Sciences, VA Boston Healthcare System, Boston, MA, USA
- Carbone Cancer Center, University of Wisconsin-Madison, Madison, WI, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Anthony R Angueira
- Institute for Diabetes, Obesity and Metabolism, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Sarah E Graham
- Department of Internal Medicine, Division of Cardiology, University of Michigan, Ann Arbor, MI, USA
| | - Ying H Shen
- Division of Cardiothoracic Surgery, Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, TX, USA
- Department of Cardiovascular Surgery, Texas Heart Institute, Houston, TX, USA
| | - Michael G Levin
- Division of Cardiovascular Medicine, Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Department of Medicine, Corporal Michael J. Crescenz VA Medical Center, Philadelphia, PA, USA
| | - James P Pirruccello
- Division of Cardiology, University of California San Francisco, San Francisco, CA, USA
- Institute for Human Genetics, University of California San Francisco, San Francisco, CA, USA
| | - Ida Surakka
- Department of Internal Medicine, Division of Cardiology, University of Michigan, Ann Arbor, MI, USA
| | - Purushotham R Karnam
- Center for Data and Computational Sciences, VA Boston Healthcare System, Boston, MA, USA
| | - Tanmoy Roychowdhury
- Department of Internal Medicine, Division of Cardiology, University of Michigan, Ann Arbor, MI, USA
- Department of Genetics, Yale School of Medicine, New Haven, CT, USA
| | - Yanming Li
- Division of Cardiothoracic Surgery, Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, TX, USA
| | - Minxian Wang
- CAS Key Laboratory of Genome Sciences and Information, Beijing Institute of Genomics, Chinese Academy of Sciences and China National Center for Bioinformation, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Krishna G Aragam
- Department of Medicine, Harvard Medical School, Boston, MA, USA
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA, USA
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Kaavya Paruchuri
- Department of Medicine, Harvard Medical School, Boston, MA, USA
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA, USA
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Verena Zuber
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, London, UK
- MRC Centre for Environment and Health, School of Public Health, Imperial College London, London, UK
- UK Dementia Research Institute at Imperial College, Imperial College London, London, UK
| | - Gabrielle E Shakt
- Division of Cardiovascular Medicine, Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Noah L Tsao
- Division of Cardiovascular Medicine, Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Renae L Judy
- Department of Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, USA
| | - Ha My T Vy
- Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Shefali S Verma
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Daniel J Rader
- Division of Translational Medicine and Human Genetics, Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
- Department of Genetics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Ron Do
- Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Joseph E Bavaria
- Division of Cardiovascular Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | | | - Marylyn D Ritchie
- Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Institute for Biomedical Informatics, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, USA
| | - Stephen Burgess
- Medical Research Council Biostatistics Unit, University of Cambridge, Cambridge, UK
- Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
| | - Dong-Chuan Guo
- Division of Medical Genetics, Department of Internal Medicine, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Patrick T Ellinor
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA, USA
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Cardiology Division, Massachusetts General Hospital, Boston, MA, USA
| | - Scott A LeMaire
- Division of Cardiothoracic Surgery, Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, TX, USA
- Department of Cardiovascular Surgery, Texas Heart Institute, Houston, TX, USA
- Cardiovascular Research Institute, Baylor College of Medicine, Houston, TX, USA
| | - Dianna M Milewicz
- Division of Medical Genetics, Department of Internal Medicine, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Cristen J Willer
- Department of Internal Medicine, Division of Cardiology, University of Michigan, Ann Arbor, MI, USA
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI, USA
- Department of Human Genetics, University of Michigan, Ann Arbor, MI, USA
| | - Pradeep Natarajan
- Department of Medicine, Harvard Medical School, Boston, MA, USA
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA, USA
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Philip S Tsao
- Veterans Affairs (VA) Palo Alto Healthcare System, Palo Alto, CA, USA
- Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA
- Stanford Cardiovascular Institute, Stanford, CA, USA
| | - Saiju Pyarajan
- Center for Data and Computational Sciences, VA Boston Healthcare System, Boston, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Scott M Damrauer
- Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
- Corporal Michael J. Crescenz VA Medical Center, Philadelphia, PA, USA.
- Department of Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
6
|
Munshaw S, Redpath AN, Pike BT, Smart N. Thymosin β4 preserves vascular smooth muscle phenotype in atherosclerosis via regulation of low density lipoprotein related protein 1 (LRP1). Int Immunopharmacol 2023; 115:109702. [PMID: 37724952 PMCID: PMC10666903 DOI: 10.1016/j.intimp.2023.109702] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Accepted: 12/29/2022] [Indexed: 01/21/2023]
Abstract
Atherosclerosis is a progressive, degenerative vascular disease and a leading cause of morbidity and mortality. In response to endothelial damage, platelet derived growth factor (PDGF)-BB induced phenotypic modulation of medial smooth muscle cells (VSMCs) promotes atherosclerotic lesion formation and destabilisation of the vessel wall. VSMC sensitivity to PDGF-BB is determined by endocytosis of Low density lipoprotein receptor related protein 1 (LRP1)-PDGFR β complexes to balance receptor recycling with lysosomal degradation. Consequently, LRP1 is implicated in various arterial diseases. Having identified Tβ4 as a regulator of LRP1-mediated endocytosis to protect against aortic aneurysm, we sought to determine whether Tβ4 may additionally function to protect against atherosclerosis, by regulating LRP1-mediated growth factor signalling. By single cell transcriptomic analysis, Tmsb4x, encoding Tβ4, strongly correlated with contractile gene expression and was significantly down-regulated in cells that adopted a modulated phenotype in atherosclerosis. We assessed susceptibility to atherosclerosis of global Tβ4 knockout mice using the ApoE-/- hypercholesterolaemia model. Inflammation, elastin integrity, VSMC phenotype and signalling were analysed in the aortic root and descending aorta. Tβ4KO; ApoE-/- mice develop larger atherosclerotic plaques than control mice, with medial layer degeneration characterised by accelerated VSMC phenotypic modulation. Defects in Tβ4KO; ApoE-/- mice phenocopied those in VSMC-specific LRP1 nulls and, moreover, were underpinned by hyperactivated LRP1-PDGFRβ signalling. We identify an atheroprotective role for endogenous Tβ4 in maintaining differentiated VSMC phenotype via LRP1-mediated PDGFRβ signalling.
Collapse
Affiliation(s)
- Sonali Munshaw
- Burdon Sanderson Cardiac Science Centre, Department of Physiology, Anatomy & Genetics, University of Oxford, Sherrington Building, South Parks Road, Oxford OX1 3PT, UK
| | - Andia N Redpath
- Burdon Sanderson Cardiac Science Centre, Department of Physiology, Anatomy & Genetics, University of Oxford, Sherrington Building, South Parks Road, Oxford OX1 3PT, UK; Institute of Developmental and Regenerative Medicine, University of Oxford, IMS-Tetsuya Nakamura Building, Old Road Campus, Oxford OX3 7TY, UK
| | - Benjamin T Pike
- Burdon Sanderson Cardiac Science Centre, Department of Physiology, Anatomy & Genetics, University of Oxford, Sherrington Building, South Parks Road, Oxford OX1 3PT, UK
| | - Nicola Smart
- Burdon Sanderson Cardiac Science Centre, Department of Physiology, Anatomy & Genetics, University of Oxford, Sherrington Building, South Parks Road, Oxford OX1 3PT, UK; Institute of Developmental and Regenerative Medicine, University of Oxford, IMS-Tetsuya Nakamura Building, Old Road Campus, Oxford OX3 7TY, UK.
| |
Collapse
|
7
|
Zhang JM, Au DT, Sawada H, Franklin MK, Moorleghen JJ, Howatt DA, Wang P, Aicher BO, Hampton B, Migliorini M, Ni F, Mullick AE, Wani MM, Ucuzian AA, Lu HS, Muratoglu SC, Daugherty A, Strickland DK. LRP1 protects against excessive superior mesenteric artery remodeling by modulating angiotensin II-mediated signaling. JCI Insight 2023; 8:e164751. [PMID: 36472907 PMCID: PMC9977308 DOI: 10.1172/jci.insight.164751] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Accepted: 11/30/2022] [Indexed: 12/12/2022] Open
Abstract
Vascular smooth muscle cells (vSMCs) exert a critical role in sensing and maintaining vascular integrity. These cells abundantly express the low-density lipoprotein receptor-related protein 1 (LRP1), a large endocytic signaling receptor that recognizes numerous ligands, including apolipoprotein E-rich lipoproteins, proteases, and protease-inhibitor complexes. We observed the spontaneous formation of aneurysms in the superior mesenteric artery (SMA) of both male and female mice in which LRP1 was genetically deleted in vSMCs (smLRP1-/- mice). Quantitative proteomics revealed elevated abundance of several proteins in smLRP1-/- mice that are known to be induced by angiotensin II-mediated (AngII-mediated) signaling, suggesting that this pathway was dysregulated. Administration of losartan, an AngII type I receptor antagonist, or an angiotensinogen antisense oligonucleotide to reduce plasma angiotensinogen concentrations restored the normal SMA phenotype in smLRP1-/- mice and prevented aneurysm formation. Additionally, using a vascular injury model, we noted excessive vascular remodeling and neointima formation in smLRP1-/- mice that was restored by losartan administration. Together, these findings reveal that LRP1 regulates vascular integrity and remodeling of the SMA by attenuating excessive AngII-mediated signaling.
Collapse
Affiliation(s)
- Jackie M Zhang
- Center for Vascular and Inflammatory Diseases and
- Department of Surgery, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Dianaly T Au
- Center for Vascular and Inflammatory Diseases and
| | - Hisashi Sawada
- Saha Cardiovascular Research Center and Saha Aortic Center and
- Department of Physiology, University of Kentucky, Lexington, Kentucky, USA
| | | | | | | | - Pengjun Wang
- Saha Cardiovascular Research Center and Saha Aortic Center and
| | - Brittany O Aicher
- Center for Vascular and Inflammatory Diseases and
- Department of Surgery, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | | | | | - Fenge Ni
- Center for Vascular and Inflammatory Diseases and
| | | | | | - Areck A Ucuzian
- Center for Vascular and Inflammatory Diseases and
- Department of Surgery, University of Maryland School of Medicine, Baltimore, Maryland, USA
- Vascular Services, Baltimore Veterans Affairs Medical Center, Baltimore, Maryland, USA
| | - Hong S Lu
- Saha Cardiovascular Research Center and Saha Aortic Center and
- Department of Physiology, University of Kentucky, Lexington, Kentucky, USA
| | | | - Alan Daugherty
- Saha Cardiovascular Research Center and Saha Aortic Center and
- Department of Physiology, University of Kentucky, Lexington, Kentucky, USA
| | - Dudley K Strickland
- Center for Vascular and Inflammatory Diseases and
- Department of Surgery, University of Maryland School of Medicine, Baltimore, Maryland, USA
- Department of Physiology, University of Maryland School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
8
|
Dong CX, Malecki C, Robertson E, Hambly B, Jeremy R. Molecular Mechanisms in Genetic Aortopathy-Signaling Pathways and Potential Interventions. Int J Mol Sci 2023; 24:ijms24021795. [PMID: 36675309 PMCID: PMC9865322 DOI: 10.3390/ijms24021795] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 01/02/2023] [Accepted: 01/11/2023] [Indexed: 01/18/2023] Open
Abstract
Thoracic aortic disease affects people of all ages and the majority of those aged <60 years have an underlying genetic cause. There is presently no effective medical therapy for thoracic aneurysm and surgery remains the principal intervention. Unlike abdominal aortic aneurysm, for which the inflammatory/atherosclerotic pathogenesis is well established, the mechanism of thoracic aneurysm is less understood. This paper examines the key cell signaling systems responsible for the growth and development of the aorta, homeostasis of endothelial and vascular smooth muscle cells and interactions between pathways. The evidence supporting a role for individual signaling pathways in pathogenesis of thoracic aortic aneurysm is examined and potential novel therapeutic approaches are reviewed. Several key signaling pathways, notably TGF-β, WNT, NOTCH, PI3K/AKT and ANGII contribute to growth, proliferation, cell phenotype and survival for both vascular smooth muscle and endothelial cells. There is crosstalk between pathways, and between vascular smooth muscle and endothelial cells, with both synergistic and antagonistic interactions. A common feature of the activation of each is response to injury or abnormal cell stress. Considerable experimental evidence supports a contribution of each of these pathways to aneurysm formation. Although human information is less, there is sufficient data to implicate each pathway in the pathogenesis of human thoracic aneurysm. As some pathways i.e., WNT and NOTCH, play key roles in tissue growth and organogenesis in early life, it is possible that dysregulation of these pathways results in an abnormal aortic architecture even in infancy, thereby setting the stage for aneurysm development in later life. Given the fine tuning of these signaling systems, functional polymorphisms in key signaling elements may set up a future risk of thoracic aneurysm. Multiple novel therapeutic agents have been developed, targeting cell signaling pathways, predominantly in cancer medicine. Future investigations addressing cell specific targeting, reduced toxicity and also less intense treatment effects may hold promise for effective new medical treatments of thoracic aortic aneurysm.
Collapse
Affiliation(s)
- Charlotte Xue Dong
- Faculty of Health and Medical Sciences, University of Sydney, Sydney, NSW 2006, Australia
| | - Cassandra Malecki
- Faculty of Health and Medical Sciences, University of Sydney, Sydney, NSW 2006, Australia
- The Baird Institute, Camperdown, NSW 2042, Australia
| | - Elizabeth Robertson
- Faculty of Health and Medical Sciences, University of Sydney, Sydney, NSW 2006, Australia
| | - Brett Hambly
- Faculty of Health and Medical Sciences, University of Sydney, Sydney, NSW 2006, Australia
| | - Richmond Jeremy
- Faculty of Health and Medical Sciences, University of Sydney, Sydney, NSW 2006, Australia
- The Baird Institute, Camperdown, NSW 2042, Australia
- Correspondence:
| |
Collapse
|
9
|
Ito S, Lu HS, Daugherty A, Sawada H. Embryonic Heterogeneity of Smooth Muscle Cells in the Complex Mechanisms of Thoracic Aortic Aneurysms. Genes (Basel) 2022; 13:genes13091618. [PMID: 36140786 PMCID: PMC9498804 DOI: 10.3390/genes13091618] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Revised: 09/02/2022] [Accepted: 09/05/2022] [Indexed: 11/24/2022] Open
Abstract
Smooth muscle cells (SMCs) are the major cell type of the aortic wall and play a pivotal role in the pathophysiology of thoracic aortic aneurysms (TAAs). TAAs occur in a region-specific manner with the proximal region being a common location. In this region, SMCs are derived embryonically from either the cardiac neural crest or the second heart field. These cells of distinct origins reside in specific locations and exhibit different biological behaviors in the complex mechanism of TAAs. The purpose of this review is to enhance understanding of the embryonic heterogeneity of SMCs in the proximal thoracic aorta and their functions in TAAs.
Collapse
Affiliation(s)
- Sohei Ito
- Saha Cardiovascular Research Center, College of Medicine, University of Kentucky, Lexington, KY 40536, USA
- Saha Aortic Center, College of Medicine, University of Kentucky, Lexington, KY 40536, USA
| | - Hong S. Lu
- Saha Cardiovascular Research Center, College of Medicine, University of Kentucky, Lexington, KY 40536, USA
- Saha Aortic Center, College of Medicine, University of Kentucky, Lexington, KY 40536, USA
- Department of Physiology, College of Medicine, University of Kentucky, Lexington, KY 40536, USA
| | - Alan Daugherty
- Saha Cardiovascular Research Center, College of Medicine, University of Kentucky, Lexington, KY 40536, USA
- Saha Aortic Center, College of Medicine, University of Kentucky, Lexington, KY 40536, USA
- Department of Physiology, College of Medicine, University of Kentucky, Lexington, KY 40536, USA
| | - Hisashi Sawada
- Saha Cardiovascular Research Center, College of Medicine, University of Kentucky, Lexington, KY 40536, USA
- Saha Aortic Center, College of Medicine, University of Kentucky, Lexington, KY 40536, USA
- Department of Physiology, College of Medicine, University of Kentucky, Lexington, KY 40536, USA
- Correspondence: ; Tel.: +1-(859)-218-1705
| |
Collapse
|
10
|
Liu Z, Andraska E, Akinbode D, Mars W, Alvidrez RIM. LRP1 in the Vascular Wall. CURRENT PATHOBIOLOGY REPORTS 2022. [DOI: 10.1007/s40139-022-00231-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
11
|
Ye D, Wu C, Chen H, Liang CL, Howatt DA, Franklin MK, Moorleghen JJ, Tyagi SC, Uijl E, Danser AHJ, Sawada H, Daugherty A, Lu HS. Fludrocortisone Induces Aortic Pathologies in Mice. Biomolecules 2022; 12:825. [PMID: 35740952 PMCID: PMC9220881 DOI: 10.3390/biom12060825] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Revised: 06/07/2022] [Accepted: 06/08/2022] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND AND OBJECTIVE In an experiment designed to explore the mechanisms of fludrocortisone-induced high blood pressure, we serendipitously observed aortic aneurysms in mice infused with fludrocortisone. The purpose of this study was to investigate whether fludrocortisone induces aortic pathologies in both normocholesterolemic and hypercholesterolemic mice. METHODS AND RESULTS Male adult C57BL/6J mice were infused with either vehicle (85% polyethylene glycol 400 (PEG-400) and 15% dimethyl sulfoxide (DMSO); n = 5) or fludrocortisone (12 mg/kg/day dissolved in 85% PEG-400 and 15% DMSO; n = 15) for 28 days. Fludrocortisone-infused mice had higher systolic blood pressure, compared to mice infused with vehicle. Fludrocortisone induced aortic pathologies in 4 of 15 mice with 3 having pathologies in the ascending and aortic arch regions and 1 having pathology in both the ascending and descending thoracic aorta. No pathologies were noted in abdominal aortas. Subsequently, we infused either vehicle (n = 5/group) or fludrocortisone (n = 15/group) into male ApoE -/- mice fed a normal laboratory diet or LDL receptor -/- mice fed either normal or Western diet. Fludrocortisone increased systolic blood pressure, irrespective of mouse strain or diet. In ApoE -/- mice infused with fludrocortisone, 2 of 15 mice had ascending aortic pathologies, but no mice had abdominal aortic pathologies. In LDL receptor -/- mice fed normal diet, 5 had ascending/arch pathologies and 1 had pathologies in the ascending, arch, and suprarenal aortic regions. In LDL receptor -/- mice fed Western diet, 2 died of aortic rupture in either the descending thoracic or abdominal region, and 2 of the 13 survived mice had ascending/arch aortic pathologies. Aortic pathologies included hemorrhage, wall thickening or thinning, or dilation. Only ascending aortic diameter in LDLR -/- mice fed Western diet reached statistical significance, compared to their vehicle. CONCLUSION Fludrocortisone induces aortic pathologies independent of hypercholesterolemia. As indicated by the findings in mouse studies, people who are taking or have taken fludrocortisone might have an increased risk of aortic pathologies.
Collapse
Affiliation(s)
- Dien Ye
- Saha Cardiovascular Research Center, Lexington, KY 40536, USA; (D.Y.); (C.W.); (H.C.); (C.-L.L.); (D.A.H.); (M.K.F.); (J.J.M.); (S.C.T.); (H.S.); (A.D.)
- Division of Pharmacology and Vascular Medicine, Department of Internal Medicine, Erasmus University Medical Center, 3015 CN Rotterdam, The Netherlands; (E.U.); (A.H.J.D.)
| | - Congqing Wu
- Saha Cardiovascular Research Center, Lexington, KY 40536, USA; (D.Y.); (C.W.); (H.C.); (C.-L.L.); (D.A.H.); (M.K.F.); (J.J.M.); (S.C.T.); (H.S.); (A.D.)
- Saha Cardiovascular Research Center, Department of Surgery, College of Medicine, University of Kentucky, Lexington, KY 40536, USA
| | - Hui Chen
- Saha Cardiovascular Research Center, Lexington, KY 40536, USA; (D.Y.); (C.W.); (H.C.); (C.-L.L.); (D.A.H.); (M.K.F.); (J.J.M.); (S.C.T.); (H.S.); (A.D.)
| | - Ching-Ling Liang
- Saha Cardiovascular Research Center, Lexington, KY 40536, USA; (D.Y.); (C.W.); (H.C.); (C.-L.L.); (D.A.H.); (M.K.F.); (J.J.M.); (S.C.T.); (H.S.); (A.D.)
| | - Deborah A. Howatt
- Saha Cardiovascular Research Center, Lexington, KY 40536, USA; (D.Y.); (C.W.); (H.C.); (C.-L.L.); (D.A.H.); (M.K.F.); (J.J.M.); (S.C.T.); (H.S.); (A.D.)
| | - Michael K. Franklin
- Saha Cardiovascular Research Center, Lexington, KY 40536, USA; (D.Y.); (C.W.); (H.C.); (C.-L.L.); (D.A.H.); (M.K.F.); (J.J.M.); (S.C.T.); (H.S.); (A.D.)
| | - Jessica J. Moorleghen
- Saha Cardiovascular Research Center, Lexington, KY 40536, USA; (D.Y.); (C.W.); (H.C.); (C.-L.L.); (D.A.H.); (M.K.F.); (J.J.M.); (S.C.T.); (H.S.); (A.D.)
| | - Samuel C. Tyagi
- Saha Cardiovascular Research Center, Lexington, KY 40536, USA; (D.Y.); (C.W.); (H.C.); (C.-L.L.); (D.A.H.); (M.K.F.); (J.J.M.); (S.C.T.); (H.S.); (A.D.)
- Saha Cardiovascular Research Center, Department of Surgery, College of Medicine, University of Kentucky, Lexington, KY 40536, USA
- Department of Physiology, University of Kentucky, Lexington, KY 40536, USA
- Saha Aortic Center, University of Kentucky, Lexington, KY 40536, USA
| | - Estrellita Uijl
- Division of Pharmacology and Vascular Medicine, Department of Internal Medicine, Erasmus University Medical Center, 3015 CN Rotterdam, The Netherlands; (E.U.); (A.H.J.D.)
| | - A. H. Jan Danser
- Division of Pharmacology and Vascular Medicine, Department of Internal Medicine, Erasmus University Medical Center, 3015 CN Rotterdam, The Netherlands; (E.U.); (A.H.J.D.)
| | - Hisashi Sawada
- Saha Cardiovascular Research Center, Lexington, KY 40536, USA; (D.Y.); (C.W.); (H.C.); (C.-L.L.); (D.A.H.); (M.K.F.); (J.J.M.); (S.C.T.); (H.S.); (A.D.)
- Department of Physiology, University of Kentucky, Lexington, KY 40536, USA
- Saha Aortic Center, University of Kentucky, Lexington, KY 40536, USA
| | - Alan Daugherty
- Saha Cardiovascular Research Center, Lexington, KY 40536, USA; (D.Y.); (C.W.); (H.C.); (C.-L.L.); (D.A.H.); (M.K.F.); (J.J.M.); (S.C.T.); (H.S.); (A.D.)
- Department of Physiology, University of Kentucky, Lexington, KY 40536, USA
- Saha Aortic Center, University of Kentucky, Lexington, KY 40536, USA
| | - Hong S. Lu
- Saha Cardiovascular Research Center, Lexington, KY 40536, USA; (D.Y.); (C.W.); (H.C.); (C.-L.L.); (D.A.H.); (M.K.F.); (J.J.M.); (S.C.T.); (H.S.); (A.D.)
- Department of Physiology, University of Kentucky, Lexington, KY 40536, USA
- Saha Aortic Center, University of Kentucky, Lexington, KY 40536, USA
| |
Collapse
|
12
|
Sawada H, Lu HS, Cassis LA, Daugherty A. Twenty Years of Studying AngII (Angiotensin II)-Induced Abdominal Aortic Pathologies in Mice: Continuing Questions and Challenges to Provide Insight Into the Human Disease. Arterioscler Thromb Vasc Biol 2022; 42:277-288. [PMID: 35045728 PMCID: PMC8866209 DOI: 10.1161/atvbaha.121.317058] [Citation(s) in RCA: 43] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
AngII (angiotensin II) infusion in mice has been used to provide mechanistic insight into human abdominal aortic aneurysms for over 2 decades. This is a technically facile animal model that recapitulates multiple facets of the human disease. Although numerous publications have reported abdominal aortic aneurysms with AngII infusion in mice, there remain many fundamental unanswered questions such as uniformity of describing the pathological characteristics and which cell type is stimulated by AngII to promote abdominal aortic aneurysms. Extrapolation of the findings to provide insight into the human disease has been hindered by the preponderance of studies designed to determine the effects on initiation of abdominal aortic aneurysms, rather than a more clinically relevant scenario of determining efficacy on the established disease. The purpose of this review is to enhance understanding of AngII-induced abdominal aortic pathologies in mice, thereby providing greater insight into the human disease.
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
| | - 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
| | - Lisa A. Cassis
- Department of Pharmacology and Nutritional Sciences, 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
|
13
|
Imaging Techniques for Aortic Aneurysms and Dissections in Mice: Comparisons of Ex Vivo, In Situ, and Ultrasound Approaches. Biomolecules 2022; 12:biom12020339. [PMID: 35204838 PMCID: PMC8869425 DOI: 10.3390/biom12020339] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Revised: 02/14/2022] [Accepted: 02/16/2022] [Indexed: 01/04/2023] Open
Abstract
Aortic aneurysms and dissections are life-threatening conditions that have a high risk for lethal bleeding and organ malperfusion. Many studies have investigated the molecular basis of these diseases using mouse models. In mice, ex vivo, in situ, and ultrasound imaging are major approaches to evaluate aortic diameters, a common parameter to determine the severity of aortic aneurysms. However, accurate evaluations of aortic dimensions by these imaging approaches could be challenging due to pathological features of aortic aneurysms. Currently, there is no standardized mode to assess aortic dissections in mice. It is important to understand the characteristics of each approach for reliable evaluation of aortic dilatations. In this review, we summarize imaging techniques used for aortic visualization in recent mouse studies and discuss their pros and cons. We also provide suggestions to facilitate the visualization of mouse aortas.
Collapse
|
14
|
Sawada H, Katsumata Y, Higashi H, Zhang C, Li Y, Morgan S, Lee LH, Singh SA, Chen JZ, Franklin MK, Moorleghen JJ, Howatt DA, Rateri DL, Shen YH, LeMaire SA, Aikawa M, Majesky MW, Lu HS, Daugherty A. Second Heart Field-derived Cells Contribute to Angiotensin II-mediated Ascending Aortopathies. Circulation 2022; 145:987-1001. [PMID: 35143327 PMCID: PMC9008740 DOI: 10.1161/circulationaha.121.058173] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Background: The ascending aorta is a common location for aneurysm and dissection. This aortic region is populated by a mosaic of medial and adventitial cells that are embryonically derived from either the second heart field (SHF) or the cardiac neural crest. SHF-derived cells populate areas that coincide with the spatial specificity of thoracic aortopathies. The purpose of this study was to determine whether and how SHF-derived cells contribute to ascending aortopathies. Methods: Ascending aortic pathologies were examined in patients with sporadic thoracic aortopathies and angiotensin II (AngII)-infused mice. Ascending aortas without overt pathology from AngII-infused mice were subjected to mass spectrometry assisted proteomics, and molecular features of SHF-derived cells were determined by single cell transcriptomic analyses. Genetic deletion of either low-density lipoprotein receptor-related protein 1 (Lrp1) or transforming growth factor-β receptor 2 (Tgfbr2) in SHF-derived cells was conducted to examine the impact of SHF-derived cells on vascular integrity. Results: Pathologies in human ascending aortic aneurysmal tissues were predominant in outer medial layers and adventitia. This gradient was mimicked in mouse aortas following AngII infusion that was coincident with the distribution of SHF-derived cells. Proteomics indicated that brief AngII infusion, prior to overt pathology, evoked downregulation of SMC proteins and differential expression of extracellular matrix proteins, including several LRP1 ligands. LRP1 deletion in SHF-derived cells augmented AngII-induced ascending aortic aneurysm and rupture. Single cell transcriptomic analysis revealed that brief AngII infusion decreased Lrp1 and Tgfbr2 mRNA abundance in SHF-derived cells and induced a unique fibroblast population with low abundance of Tgfbr2 mRNA. SHF-specific Tgfbr2 deletion led to embryonic lethality at E12.5 with dilatation of the outflow tract and retroperitoneal hemorrhage. Integration of proteomic and single cell transcriptomics results identified plasminogen activator inhibitor 1 (PAI1) as the most increased protein in SHF-derived SMCs and fibroblasts during AngII infusion. Immunostaining revealed a transmural gradient of PAI1 in both ascending aortas of AngII-infused mice and human ascending aneurysmal aortas that mimicked the gradient of medial and adventitial pathologies. Conclusions: SHF-derived cells exert a critical role in maintaining vascular integrity through LRP1 and TGF-β signaling associated with increases of aortic PAI1.
Collapse
Affiliation(s)
- Hisashi Sawada
- Saha Cardiovascular Research Center, College of Medicine, University of Kentucky, Lexington, KY; Saha Aortic Center, College of Medicine, University of Kentucky, Lexington, KY; Department of Physiology, College of Medicine, 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
| | - Hideyuki Higashi
- Center for Interdisciplinary Cardiovascular Sciences, Cardiovascular Division, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Chen Zhang
- Division of Cardiothoracic Surgery, Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, TX; Department of Cardiovascular Surgery, Texas Heart Institute, Houston, TX
| | - Yanming Li
- Division of Cardiothoracic Surgery, Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, TX; Department of Cardiovascular Surgery, Texas Heart Institute, Houston, TX
| | - Stephanie Morgan
- Center for Interdisciplinary Cardiovascular Sciences, Cardiovascular Division, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Lang H Lee
- Center for Interdisciplinary Cardiovascular Sciences, Cardiovascular Division, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Sasha A Singh
- Center for Interdisciplinary Cardiovascular Sciences, Cardiovascular Division, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Jeff Z Chen
- Saha Cardiovascular Research Center, College of Medicine, University of Kentucky, Lexington, KY; Department of Physiology, College of Medicine, University of Kentucky, Lexington, KY
| | - Michael K Franklin
- Saha Cardiovascular Research Center, College of Medicine, University of Kentucky, Lexington, KY
| | - Jessica J Moorleghen
- Saha Cardiovascular Research Center, College of Medicine, University of Kentucky, Lexington, KY
| | - Deborah A Howatt
- Saha Cardiovascular Research Center, College of Medicine, University of Kentucky, Lexington, KY
| | - Debra L Rateri
- Saha Cardiovascular Research Center, College of Medicine, University of Kentucky, Lexington, KY
| | - Ying H Shen
- Division of Cardiothoracic Surgery, Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, TX; Department of Cardiovascular Surgery, Texas Heart Institute, Houston, TX
| | - Scott A LeMaire
- Division of Cardiothoracic Surgery, Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, TX; Department of Cardiovascular Surgery, Texas Heart Institute, Houston, TX
| | - Masanori Aikawa
- Center for Interdisciplinary Cardiovascular Sciences, Cardiovascular Division, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Mark W Majesky
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA; Department of Pediatrics, University of Washington, Seattle, WA; Center for Developmental Biology and Regenerative Medicine, Seattle Children's Research Institute, Seattle, WA
| | - Hong S Lu
- Saha Cardiovascular Research Center, College of Medicine, University of Kentucky, Lexington, KY; Saha Aortic Center, College of Medicine, University of Kentucky, Lexington, KY; Department of Physiology, College of Medicine, University of Kentucky, Lexington, KY
| | - Alan Daugherty
- Saha Cardiovascular Research Center, College of Medicine, University of Kentucky, Lexington, KY; Saha Aortic Center, College of Medicine, University of Kentucky, Lexington, KY; Department of Physiology, College of Medicine, University of Kentucky, Lexington, KY
| |
Collapse
|
15
|
Erhart P, Körfer D, Grond-Ginsbach C, Qiao JL, Bischoff MS, Hempel M, Schaaf CP, Grau A, Böckler D. Genetic Variation in LRP1 Associates with Stanford Type B Aortic Dissection Risk and Clinical Outcome. J Cardiovasc Dev Dis 2022; 9:jcdd9010014. [PMID: 35050224 PMCID: PMC8780592 DOI: 10.3390/jcdd9010014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2021] [Revised: 12/29/2021] [Accepted: 01/04/2022] [Indexed: 02/04/2023] Open
Abstract
Genetic variation in LRP1 (low-density lipoprotein receptor-related protein 1) was reported to be associated with thoracic aortic dissections and aneurysms. The aims of this study were to confirm this association in a prospective single-center patient cohort of patients with acute Stanford type B aortic dissections (STBAD) and to assess the impact of LRP1 variation on clinical outcome. The single nucleotide variation (SNV) rs11172113 within the LRP1 gene was genotyped in 113 STBAD patients and 768 healthy control subjects from the same population. The T-allele of rs11172113 was more common in STBAD patients as compared to the reference group (72.6% vs. 59.6%) and confirmed to be an independent risk factor for STBAD (p = 0.002) after sex and age adjustment in a logistic regression model analyzing diabetes, smoking and hypertension as additional risk factors. Analysis of clinical follow-up (median follow-up 2.0 years) revealed that patients with the T-allele were more likely to suffer aorta-related complications (T-allele 75.6% vs. 63.8%; p = 0.022). In this study sample of STBAD patients, variation in LRP1 was an independent risk factor for STBAD and affected clinical outcome.
Collapse
Affiliation(s)
- Philipp Erhart
- Department of Vascular and Endovascular Surgery, Heidelberg University Hospital, 69120 Heidelberg, Germany; (D.K.); (C.G.-G.); (J.-L.Q.); (M.S.B.); (D.B.)
- Correspondence: ; Tel.: +49-6221-56-6249
| | - Daniel Körfer
- Department of Vascular and Endovascular Surgery, Heidelberg University Hospital, 69120 Heidelberg, Germany; (D.K.); (C.G.-G.); (J.-L.Q.); (M.S.B.); (D.B.)
| | - Caspar Grond-Ginsbach
- Department of Vascular and Endovascular Surgery, Heidelberg University Hospital, 69120 Heidelberg, Germany; (D.K.); (C.G.-G.); (J.-L.Q.); (M.S.B.); (D.B.)
| | - Jia-Lu Qiao
- Department of Vascular and Endovascular Surgery, Heidelberg University Hospital, 69120 Heidelberg, Germany; (D.K.); (C.G.-G.); (J.-L.Q.); (M.S.B.); (D.B.)
| | - Moritz S. Bischoff
- Department of Vascular and Endovascular Surgery, Heidelberg University Hospital, 69120 Heidelberg, Germany; (D.K.); (C.G.-G.); (J.-L.Q.); (M.S.B.); (D.B.)
| | - Maja Hempel
- Institute of Human Genetics, Heidelberg University, 69120 Heidelberg, Germany; (M.H.); (C.P.S.)
| | - Christian P. Schaaf
- Institute of Human Genetics, Heidelberg University, 69120 Heidelberg, Germany; (M.H.); (C.P.S.)
| | - Armin Grau
- Department of Neurology, Community Hospital Klinikum der Stadt Ludwigshafen am Rhein, 67063 Ludwigshafen, Germany;
| | - Dittmar Böckler
- Department of Vascular and Endovascular Surgery, Heidelberg University Hospital, 69120 Heidelberg, Germany; (D.K.); (C.G.-G.); (J.-L.Q.); (M.S.B.); (D.B.)
| |
Collapse
|
16
|
Mougin Z, Huguet Herrero J, Boileau C, Le Goff C. ADAMTS Proteins and Vascular Remodeling in Aortic Aneurysms. Biomolecules 2021; 12:12. [PMID: 35053160 PMCID: PMC8773774 DOI: 10.3390/biom12010012] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Revised: 12/16/2021] [Accepted: 12/20/2021] [Indexed: 12/11/2022] Open
Abstract
Extracellular matrix (ECM) in the vascular wall is a highly dynamic structure composed of a set of different molecules such as elastins, collagens, fibronectin (Fn), laminins, proteoglycans, and polysaccharides. ECM undergoes remodeling processes to regulate vascular smooth muscle and endothelial cells' proliferation, differentiation, and adhesion. Abnormalities affecting the ECM can lead to alteration in cellular behavior and from this, this can conduce to the development of pathologies. Metalloproteases play a key role in maintaining the homeostasis of ECM by mediating the cleavage of different ECM components. There are different types of metalloproteases: matrix metalloproteinases (MMPs), disintegrin and metalloproteinases (ADAMs), and ADAMs with thrombospondin motifs (ADAMTSs). ADAMTSs have been found to participate in cardiovascular physiology and diseases and specifically in aortic aneurysms. This review aims to decipher the potential role of ADAMTS proteins in the physiopathologic development of Thoracic Aortic Aneurysms (TAA) and Abdominal Aortic Aneurysms (AAA). This review will focus on what is known on the ADAMTS family involved in human aneurysms from human tissues to mouse models. The recent findings on THSD4 (encoding ADAMTSL6) mutations in TAA give a new insight on the involvement of the ADAMTS family in TAA.
Collapse
Affiliation(s)
- Zakaria Mougin
- INSERM U1148, Laboratory of Vascular Translational Science, Université de Paris, Hôpital Bichat, F-75018 Paris, France; (Z.M.); (J.H.H.); (C.B.)
| | - Julia Huguet Herrero
- INSERM U1148, Laboratory of Vascular Translational Science, Université de Paris, Hôpital Bichat, F-75018 Paris, France; (Z.M.); (J.H.H.); (C.B.)
| | - Catherine Boileau
- INSERM U1148, Laboratory of Vascular Translational Science, Université de Paris, Hôpital Bichat, F-75018 Paris, France; (Z.M.); (J.H.H.); (C.B.)
- Département de Génétique, AP-HP, Hôpital Bichat, F-75018 Paris, France
| | - Carine Le Goff
- INSERM U1148, Laboratory of Vascular Translational Science, Université de Paris, Hôpital Bichat, F-75018 Paris, France; (Z.M.); (J.H.H.); (C.B.)
| |
Collapse
|
17
|
Guo Y, Tang Z, Yan B, Yin H, Tai S, Peng J, Cui Y, Gui Y, Belke D, Zhou S, Zheng XL. PCSK9 (Proprotein Convertase Subtilisin/Kexin Type 9) Triggers Vascular Smooth Muscle Cell Senescence and Apoptosis: Implication of Its Direct Role in Degenerative Vascular Disease. Arterioscler Thromb Vasc Biol 2021; 42:67-86. [PMID: 34809446 DOI: 10.1161/atvbaha.121.316902] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
OBJECTIVE PCSK9 (proprotein convertase subtilisin/kexin type 9) plays a critical role in cholesterol metabolism via the PCSK9-LDLR (low-density lipoprotein receptor) axis in the liver; however, evidence indicates that PCSK9 directly contributes to the pathogenesis of various diseases through mechanisms independent of its LDL-cholesterol regulation. The objective of this study was to determine how PCSK9 directly acts on vascular smooth muscle cells (SMCs), contributing to degenerative vascular disease. Approach and Results: We first examined the effects of PCSK9 on cultured human aortic SMCs. Overexpression of PCSK9 downregulated the expression of ApoER2 (apolipoprotein E receptor 2), a known target of PCSK9. Treatment with soluble recombinant human ApoER2 or the DNA synthesis inhibitor, hydroxyurea, inhibited PCSK9-induced polyploidization and other cellular responses of human SMCs. Treatment with antibodies against ApoER2 resulted in similar effects to those observed with PCSK9 overexpression. Inducible, SMC-specific knockout of Pcsk9 accelerated neointima formation in mouse carotid arteries and reduced age-related arterial stiffness. PCSK9 was expressed in SMCs of human atherosclerotic lesions and abundant in the "shoulder" regions of vulnerable atherosclerotic plaques. PCSK9 was also expressed in SMCs of abdominal aortic aneurysm, which was inversely related to the expression of smooth muscle α-actin. CONCLUSIONS Our findings demonstrate that PCSK9 inhibits proliferation and induces polyploidization, senescence, and apoptosis, which may be relevant to various degenerative vascular diseases.
Collapse
Affiliation(s)
- Yanan Guo
- Departments of Biochemistry and Molecular Biology and Physiology and Pharmacology (Y. Guo, Z.T., B.Y., H.Y., Y. Gui, X.-L. Zheng).,Department of Cardiology, the Second Xiangya Hospital of Central South University, Changsha, China (Y. Guo, S.T., S.Z.)
| | - Zhihan Tang
- Departments of Biochemistry and Molecular Biology and Physiology and Pharmacology (Y. Guo, Z.T., B.Y., H.Y., Y. Gui, X.-L. Zheng).,Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerology of Hunan Province, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, School of Basic Medical Sciences, Hengyang Medical School, University of South China, Hengyang, Hunan (Z.T., B.Y., J.P., Y.C.)
| | - Binjie Yan
- Departments of Biochemistry and Molecular Biology and Physiology and Pharmacology (Y. Guo, Z.T., B.Y., H.Y., Y. Gui, X.-L. Zheng).,Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerology of Hunan Province, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, School of Basic Medical Sciences, Hengyang Medical School, University of South China, Hengyang, Hunan (Z.T., B.Y., J.P., Y.C.)
| | - Hao Yin
- Departments of Biochemistry and Molecular Biology and Physiology and Pharmacology (Y. Guo, Z.T., B.Y., H.Y., Y. Gui, X.-L. Zheng).,Now with Robarts Research Institute, Schulich School of Medicine and Dentistry, Western University, London, Canada (H.Y.)
| | - Shi Tai
- Department of Cardiology, the Second Xiangya Hospital of Central South University, Changsha, China (Y. Guo, S.T., S.Z.)
| | - Juan Peng
- Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerology of Hunan Province, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, School of Basic Medical Sciences, Hengyang Medical School, University of South China, Hengyang, Hunan (Z.T., B.Y., J.P., Y.C.)
| | - Yuting Cui
- Departments of Biochemistry and Molecular Biology and Physiology and Pharmacology (Y. Guo, Z.T., B.Y., H.Y., Y. Gui, X.-L. Zheng).,Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerology of Hunan Province, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, School of Basic Medical Sciences, Hengyang Medical School, University of South China, Hengyang, Hunan (Z.T., B.Y., J.P., Y.C.)
| | - Yu Gui
- Departments of Biochemistry and Molecular Biology and Physiology and Pharmacology (Y. Guo, Z.T., B.Y., H.Y., Y. Gui, X.-L. Zheng)
| | - Darrell Belke
- Departments of Biochemistry and Molecular Biology and Physiology and Pharmacology (Y. Guo, Z.T., B.Y., H.Y., Y. Gui, X.-L. Zheng)
| | - Shenghua Zhou
- Department of Cardiology, the Second Xiangya Hospital of Central South University, Changsha, China (Y. Guo, S.T., S.Z.)
| | - Xi-Long Zheng
- Departments of Biochemistry and Molecular Biology and Physiology and Pharmacology (Y. Guo, Z.T., B.Y., H.Y., Y. Gui, X.-L. Zheng)
| |
Collapse
|
18
|
Weininger G, Ostberg N, Shang M, Zafar M, Ziganshin BA, Liu S, Erben Y, Elefteriades JA. Lipid profiles help to explain protection from systemic atherosclerosis in patients with ascending aortic aneurysm. J Thorac Cardiovasc Surg 2021; 163:e129-e132. [PMID: 34635315 DOI: 10.1016/j.jtcvs.2021.09.031] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Revised: 08/23/2021] [Accepted: 09/14/2021] [Indexed: 11/28/2022]
Affiliation(s)
- Gabe Weininger
- Aortic Institute at Yale-New Haven Hospital, Yale University School of Medicine, New Haven, Conn
| | - Nicolai Ostberg
- Aortic Institute at Yale-New Haven Hospital, Yale University School of Medicine, New Haven, Conn; New York University Grossman School of Medicine, New York, NY
| | - Michael Shang
- Aortic Institute at Yale-New Haven Hospital, Yale University School of Medicine, New Haven, Conn
| | - Mohammad Zafar
- Aortic Institute at Yale-New Haven Hospital, Yale University School of Medicine, New Haven, Conn
| | - Bulat A Ziganshin
- Aortic Institute at Yale-New Haven Hospital, Yale University School of Medicine, New Haven, Conn; Department of Cardiovascular and Endovascular Surgery, Kazan State Medical University, Kazan, Russia
| | - Shirley Liu
- Aortic Institute at Yale-New Haven Hospital, Yale University School of Medicine, New Haven, Conn
| | - Young Erben
- Aortic Institute at Yale-New Haven Hospital, Yale University School of Medicine, New Haven, Conn; Division of Vascular and Endovascular Surgery, Mayo Clinic, Jacksonville, Fla
| | - John A Elefteriades
- Aortic Institute at Yale-New Haven Hospital, Yale University School of Medicine, New Haven, Conn.
| |
Collapse
|
19
|
Kim ESH, Saw J, Kadian-Dodov D, Wood M, Ganesh SK. FMD and SCAD: Sex-Biased Arterial Diseases With Clinical and Genetic Pleiotropy. Circ Res 2021; 128:1958-1972. [PMID: 34110898 DOI: 10.1161/circresaha.121.318300] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Multifocal fibromuscular dysplasia (FMD) and spontaneous coronary artery dissection are both sex-biased diseases disproportionately affecting women over men in a 9:1 ratio. Traditionally known in the context of renovascular hypertension, recent advances in knowledge about FMD have demonstrated that FMD is a systemic arteriopathy presenting as arterial stenosis, aneurysm, and dissection in virtually any arterial bed. FMD is also characterized by major cardiovascular presentations including hypertension, stroke, and myocardial infarction. Similar to FMD, spontaneous coronary artery dissection is associated with a high prevalence of extracoronary vascular abnormalities, including FMD, aneurysm, and extracoronary dissection, and recent studies have also found genetic associations between the two diseases. This review will summarize the relationship between FMD and spontaneous coronary artery dissection with a focus on common clinical associations, histopathologic mechanisms, genetic susceptibilities, and the biology of these diseases. The current status of disease models and critical future research directions will also be addressed.
Collapse
Affiliation(s)
- Esther S H Kim
- Division of Cardiovascular Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN (E.S.H.K.)
| | - Jacqueline Saw
- Division of Cardiology, Vancouver General Hospital, University of British Columbia Canada (J.S.)
| | - Daniella Kadian-Dodov
- Zena and Michael A. Wiener Cardiovascular Institute, Marie-Joseé and Henry R. Kravis Center for Cardiovascular Health, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY (D.K.-D.)
| | - Malissa Wood
- Division of Cardiology, Harvard Medical School, Massachusetts General Hospital, Boston (M.W.)
| | - Santhi K Ganesh
- Division of Cardiovascular Medicine, Department of Internal Medicine (S.K.G.), University of Michigan Medical School, Ann Arbor.,Department of Human Genetics (S.K.G.), University of Michigan Medical School, Ann Arbor
| |
Collapse
|
20
|
Munshaw S, Bruche S, Redpath AN, Jones A, Patel J, Dubé KN, Lee R, Hester SS, Davies R, Neal G, Handa A, Sattler M, Fischer R, Channon KM, Smart N. Thymosin β4 protects against aortic aneurysm via endocytic regulation of growth factor signaling. J Clin Invest 2021; 131:127884. [PMID: 33784254 PMCID: PMC8121525 DOI: 10.1172/jci127884] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2019] [Accepted: 03/23/2021] [Indexed: 01/06/2023] Open
Abstract
Vascular stability and tone are maintained by contractile smooth muscle cells (VSMCs). However, injury-induced growth factors stimulate a contractile-synthetic phenotypic modulation which increases susceptibility to abdominal aortic aneurysm (AAA). As a regulator of embryonic VSMC differentiation, we hypothesized that Thymosin β4 (Tβ4) may function to maintain healthy vasculature throughout postnatal life. This was supported by the identification of an interaction with low density lipoprotein receptor related protein 1 (LRP1), an endocytic regulator of platelet-derived growth factor BB (PDGF-BB) signaling and VSMC proliferation. LRP1 variants have been implicated by genome-wide association studies with risk of AAA and other arterial diseases. Tβ4-null mice displayed aortic VSMC and elastin defects that phenocopy those of LRP1 mutants, and their compromised vascular integrity predisposed them to Angiotensin II-induced aneurysm formation. Aneurysmal vessels were characterized by enhanced VSMC phenotypic modulation and augmented PDGFR-β signaling. In vitro, enhanced sensitivity to PDGF-BB upon loss of Tβ4 was associated with dysregulated endocytosis, with increased recycling and reduced lysosomal targeting of LRP1-PDGFR-β. Accordingly, the exacerbated aneurysmal phenotype in Tβ4-null mice was rescued upon treatment with the PDGFR-β antagonist Imatinib. Our study identifies Tβ4 as a key regulator of LRP1 for maintaining vascular health, and provides insights into the mechanisms of growth factor-controlled VSMC phenotypic modulation underlying aortic disease progression.
Collapse
MESH Headings
- Angiotensin II/adverse effects
- Angiotensin II/pharmacology
- Animals
- Aortic Aneurysm, Abdominal/chemically induced
- Aortic Aneurysm, Abdominal/genetics
- Aortic Aneurysm, Abdominal/metabolism
- Aortic Aneurysm, Abdominal/prevention & control
- Becaplermin/genetics
- Becaplermin/metabolism
- Low Density Lipoprotein Receptor-Related Protein-1/genetics
- Low Density Lipoprotein Receptor-Related Protein-1/metabolism
- Male
- Mice
- Mice, Knockout
- Muscle, Smooth, Vascular/metabolism
- Myocytes, Smooth Muscle/metabolism
- Receptor, Platelet-Derived Growth Factor beta/genetics
- Receptor, Platelet-Derived Growth Factor beta/metabolism
- Signal Transduction/drug effects
- Signal Transduction/genetics
- Thymosin/genetics
- Thymosin/metabolism
- Thymosin/pharmacology
Collapse
Affiliation(s)
- Sonali Munshaw
- Burdon Sanderson Cardiac Science Centre, Department of Physiology, Anatomy & Genetics, University of Oxford, Sherrington Building, Oxford, United Kingdom
| | - Susann Bruche
- Burdon Sanderson Cardiac Science Centre, Department of Physiology, Anatomy & Genetics, University of Oxford, Sherrington Building, Oxford, United Kingdom
| | - Andia N. Redpath
- Burdon Sanderson Cardiac Science Centre, Department of Physiology, Anatomy & Genetics, University of Oxford, Sherrington Building, Oxford, United Kingdom
| | - Alisha Jones
- Institute of Structural Biology, Helmholtz Zentrum München, Neuherberg, Munich, Germany
- Biomolecular NMR and Center for Integrated Protein Science Munich at Chemistry Department, Technical University of Munich, Garching, Munich, Germany
| | - Jyoti Patel
- BHF Centre of Research Excellence, Division of Cardiovascular Medicine, John Radcliffe Hospital, University of Oxford, Oxford, United Kingdom
| | | | - Regent Lee
- Nuffield Department of Surgical Sciences, University of Oxford, Oxford, United Kingdom
| | - Svenja S. Hester
- Nuffield Department of Medicine, Target Discovery Institute, University of Oxford, Oxford, United Kingdom
| | - Rachel Davies
- Burdon Sanderson Cardiac Science Centre, Department of Physiology, Anatomy & Genetics, University of Oxford, Sherrington Building, Oxford, United Kingdom
| | - Giles Neal
- Burdon Sanderson Cardiac Science Centre, Department of Physiology, Anatomy & Genetics, University of Oxford, Sherrington Building, Oxford, United Kingdom
| | - Ashok Handa
- Nuffield Department of Surgical Sciences, University of Oxford, Oxford, United Kingdom
| | - Michael Sattler
- Institute of Structural Biology, Helmholtz Zentrum München, Neuherberg, Munich, Germany
- Biomolecular NMR and Center for Integrated Protein Science Munich at Chemistry Department, Technical University of Munich, Garching, Munich, Germany
| | - Roman Fischer
- Nuffield Department of Medicine, Target Discovery Institute, University of Oxford, Oxford, United Kingdom
| | - Keith M. Channon
- BHF Centre of Research Excellence, Division of Cardiovascular Medicine, John Radcliffe Hospital, University of Oxford, Oxford, United Kingdom
| | - Nicola Smart
- Burdon Sanderson Cardiac Science Centre, Department of Physiology, Anatomy & Genetics, University of Oxford, Sherrington Building, Oxford, United Kingdom
| |
Collapse
|
21
|
He Z, Wang G, Wu J, Tang Z, Luo M. The molecular mechanism of LRP1 in physiological vascular homeostasis and signal transduction pathways. Biomed Pharmacother 2021; 139:111667. [PMID: 34243608 DOI: 10.1016/j.biopha.2021.111667] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2021] [Revised: 04/07/2021] [Accepted: 04/23/2021] [Indexed: 01/10/2023] Open
Abstract
Interactions between vascular smooth muscle cells (VSMCs), endothelial cells (ECs), pericytes (PCs) and macrophages (MФ), the major components of blood vessels, play a crucial role in maintaining vascular structural and functional homeostasis. Low-density lipoprotein (LDL) receptor-related protein-1 (LRP1), a transmembrane receptor protein belonging to the LDL receptor family, plays multifunctional roles in maintaining endocytosis, homeostasis, and signal transduction. Accumulating evidence suggests that LRP1 modulates vascular homeostasis mainly by regulating vasoactive substances and specific intracellular signaling pathways, including the plasminogen activator inhibitor 1 (PAI-1) signaling pathway, platelet-derived growth factor (PDGF) signaling pathway, transforming growth factor-β (TGF-β) signaling pathway and vascular endothelial growth factor (VEGF) signaling pathway. The aim of the present review is to focus on recent advances in the discovery and mechanism of vascular homeostasis regulated by LRP1-dependent signaling pathways. These recent discoveries expand our understanding of the mechanisms controlling LRP1 as a target for studies on vascular complications.
Collapse
Affiliation(s)
- Zhaohui He
- Collaborative Innovation Center for Prevention and Treatment of Cardiovascular Disease of Sichuan Province, Drug Discovery Reseach Center, Southwest Medical University, 319 Zhongshan Road, Luzhou, Sichuan 646000, China; Department of Clinical Medicine, the Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Gang Wang
- Collaborative Innovation Center for Prevention and Treatment of Cardiovascular Disease of Sichuan Province, Drug Discovery Reseach Center, Southwest Medical University, 319 Zhongshan Road, Luzhou, Sichuan 646000, China; Laboratory for Cardiovascular Pharmacology of Department of Pharmacology, the School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China
| | - Jianbo Wu
- Collaborative Innovation Center for Prevention and Treatment of Cardiovascular Disease of Sichuan Province, Drug Discovery Reseach Center, Southwest Medical University, 319 Zhongshan Road, Luzhou, Sichuan 646000, China; Laboratory for Cardiovascular Pharmacology of Department of Pharmacology, the School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China; Dalton Cardiovascular Research Center, University of Missouri, Columbia, MO, United States
| | - Zonghao Tang
- Collaborative Innovation Center for Prevention and Treatment of Cardiovascular Disease of Sichuan Province, Drug Discovery Reseach Center, Southwest Medical University, 319 Zhongshan Road, Luzhou, Sichuan 646000, China; Laboratory for Cardiovascular Pharmacology of Department of Pharmacology, the School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China.
| | - Mao Luo
- Collaborative Innovation Center for Prevention and Treatment of Cardiovascular Disease of Sichuan Province, Drug Discovery Reseach Center, Southwest Medical University, 319 Zhongshan Road, Luzhou, Sichuan 646000, China; Laboratory for Cardiovascular Pharmacology of Department of Pharmacology, the School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China.
| |
Collapse
|
22
|
Sharma N, Hans CP. Interleukin 12p40 Deficiency Promotes Abdominal Aortic Aneurysm by Activating CCN2/MMP2 Pathways. J Am Heart Assoc 2021; 10:e017633. [PMID: 33470127 PMCID: PMC7955443 DOI: 10.1161/jaha.120.017633] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Accepted: 11/16/2020] [Indexed: 12/14/2022]
Abstract
Background Development of abdominal aortic aneurysm (AAA) is associated with proinflammatory cytokines including interleukin-12 (IL12). Deficiency of interleukin 12p40 (IL12p40) increases localized fibrotic events by promoting TGFβ2 (transforming growth factor β)-dependent anti-inflammatory response. Here, we determined whether IL12p40 deficiency in apolipoprotein E-/- mice attenuates the development of AAA by antagonizing proinflammatory response. Methods and Results Double knockout (DKO) mice were generated by crossbreeding IL12p40-/- mice with apolipoprotein E-/- mice (n=12). Aneurysmal studies were performed using angiotensin II (1 µg/kg/min; subcutaneous). Surprisingly, DKO mice did not prevent the development of AAA with angiotensin II infusion. Immunohistological analysis, however, showed distinct pathological features between apolipoprotein E-/- and DKO mice. Polymerase chain reaction (7 day) and cytokine arrays (28 day) of the aortic tissues from DKO mice showed significantly increased expression of cytokines related to anti-inflammatory response (interleukin 5 and interleukin 13), synthetic vascular smooth muscle cell phenotype (Activin receptor-like kinase-1 (ALK-1), artemin, and betacellulin) and T helper 17-associated response (4-1BB, interleukin-17e (Il17e) and Cd40 ligand (Cd-40L)). Indeed, DKO mice exhibited increased expression of the fibro-proteolytic pathway in the medial layer of aortae induced by cellular communication network factor 2 (CCN2) and Cd3+IL17+ cells compared with apolipoprotein E-/- mice. Laser capture microdissection showed predominant expression of CCN2/TGFβ2 in the medial layer of human AAA. Finally, Ccn2 haploinsufficiency in the mice showed decreased AAA incidence in response to elastase infusion, associated with decreased matrix metalloproteinase-2 expression. Conclusions Our study reveals novel roles for IL12p40 deficiency in inducing fibro-proteolytic activities in the aneurysmal mouse model. Mechanistically, these effects of IL12p40 deficiency are mediated by CCN2/matrix metalloproteinase-2 crosstalk in the medial layer of aneurysmal aortae.
Collapse
MESH Headings
- Aged
- Animals
- Aorta, Abdominal/diagnostic imaging
- Aorta, Abdominal/metabolism
- Aorta, Abdominal/physiopathology
- Aortic Aneurysm, Abdominal/etiology
- Aortic Aneurysm, Abdominal/genetics
- Aortic Aneurysm, Abdominal/metabolism
- Blotting, Western
- Cells, Cultured
- Connective Tissue Growth Factor/biosynthesis
- Connective Tissue Growth Factor/genetics
- Disease Models, Animal
- Electrocardiography
- Female
- Gene Expression Regulation
- Humans
- Interleukin-12 Subunit p40/blood
- Interleukin-12 Subunit p40/deficiency
- Male
- Matrix Metalloproteinase 2/biosynthesis
- Matrix Metalloproteinase 2/genetics
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Middle Aged
- RNA/genetics
- T-Lymphocytes/metabolism
- T-Lymphocytes/pathology
- Ultrasonography
- Vascular Stiffness/physiology
Collapse
Affiliation(s)
- Neekun Sharma
- Department of Cardiovascular MedicineUniversity of MissouriColumbiaMO
- Dalton Cardiovascular Research CenterUniversity of MissouriColumbiaMO
| | - Chetan P. Hans
- Department of Cardiovascular MedicineUniversity of MissouriColumbiaMO
- Dalton Cardiovascular Research CenterUniversity of MissouriColumbiaMO
- Department of Medical Pharmacology and PhysiologyUniversity of MissouriColumbiaMO
| |
Collapse
|
23
|
Abstract
The a disintegrin-like and metalloproteinase with thrombospondin motif (ADAMTS) family comprises 19 proteases that regulate the structure and function of extracellular proteins in the extracellular matrix and blood. The best characterized cardiovascular role is that of ADAMTS-13 in blood. Moderately low ADAMTS-13 levels increase the risk of ischeamic stroke and very low levels (less than 10%) can cause thrombotic thrombocytopenic purpura (TTP). Recombinant ADAMTS-13 is currently in clinical trials for treatment of TTP. Recently, new cardiovascular roles for ADAMTS proteases have been discovered. Several ADAMTS family members are important in the development of blood vessels and the heart, especially the valves. A number of studies have also investigated the potential role of ADAMTS-1, -4 and -5 in cardiovascular disease. They cleave proteoglycans such as versican, which represent major structural components of the arteries. ADAMTS-7 and -8 are attracting considerable interest owing to their implication in atherosclerosis and pulmonary arterial hypertension, respectively. Mutations in the ADAMTS19 gene cause progressive heart valve disease and missense variants in ADAMTS6 are associated with cardiac conduction. In this review, we discuss in detail the evidence for these and other cardiovascular roles of ADAMTS family members, their proteolytic substrates and the potential molecular mechanisms involved.
Collapse
Affiliation(s)
- Salvatore Santamaria
- Centre for Haematology, Imperial College London, Du Cane Road, London W12 0NN, UK
| | - Rens de Groot
- Centre for Haematology, Imperial College London, Du Cane Road, London W12 0NN, UK.,Institute of Cardiovascular Science, University College London, 51 Chenies Mews, London WC1E 6HX, UK
| |
Collapse
|
24
|
Legaki E, Siasos G, Klonaris C, Athanasiadis D, Patelis N, Sioziou A, Oikonomou E, Liakakos T, Gazouli M, Tousoulis D. Mir-335-5p as a potential regulator of LRP1 expression in abdominal aortic aneurysm. Hellenic J Cardiol 2020; 61:430-432. [PMID: 32088330 DOI: 10.1016/j.hjc.2020.01.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2019] [Revised: 12/21/2019] [Accepted: 01/18/2020] [Indexed: 11/27/2022] Open
Affiliation(s)
- Evangelia Legaki
- Department of Basic Medical Sciences, Laboratory of Biology, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Gerasimos Siasos
- First Department of Cardiology, National and Kapodistrian University of Athens Hippokration General Hospital, Athens, Greece.
| | - Christos Klonaris
- First Department of Surgery, Vascular Unit, National and Kapodistrian University of Athens, Laiko General Hospital, Athens, Greece
| | - Dimitrios Athanasiadis
- First Department of Surgery, Vascular Unit, National and Kapodistrian University of Athens, Laiko General Hospital, Athens, Greece
| | - Nikolaos Patelis
- First Department of Surgery, Vascular Unit, National and Kapodistrian University of Athens, Laiko General Hospital, Athens, Greece
| | - Anna Sioziou
- Department of Basic Medical Sciences, Laboratory of Biology, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Evangelos Oikonomou
- First Department of Cardiology, National and Kapodistrian University of Athens Hippokration General Hospital, Athens, Greece
| | - Theodoros Liakakos
- First Department of Surgery, Vascular Unit, National and Kapodistrian University of Athens, Laiko General Hospital, Athens, Greece
| | - Maria Gazouli
- Department of Basic Medical Sciences, Laboratory of Biology, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Dimitrios Tousoulis
- First Department of Cardiology, National and Kapodistrian University of Athens Hippokration General Hospital, Athens, Greece
| |
Collapse
|
25
|
Sawada H, Franklin MK, Moorleghen JJ, Howatt DA, Kukida M, Lu HS, Daugherty A. Ultrasound Monitoring of Descending Aortic Aneurysms and Dissections in Mice. Arterioscler Thromb Vasc Biol 2020; 40:2557-2559. [PMID: 32847392 DOI: 10.1161/atvbaha.120.314799] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Affiliation(s)
- Hisashi Sawada
- Saha Cardiovascular Research Center (H.S., M.K.F., J.J.M., D.A.H., M.K., H.S.L., A.D.), University of Kentucky, Lexington
| | - Michael K Franklin
- Saha Cardiovascular Research Center (H.S., M.K.F., J.J.M., D.A.H., M.K., H.S.L., A.D.), University of Kentucky, Lexington
| | - Jessica J Moorleghen
- Saha Cardiovascular Research Center (H.S., M.K.F., J.J.M., D.A.H., M.K., H.S.L., A.D.), University of Kentucky, Lexington
| | - Deborah A Howatt
- Saha Cardiovascular Research Center (H.S., M.K.F., J.J.M., D.A.H., M.K., H.S.L., A.D.), University of Kentucky, Lexington
| | - Masayoshi Kukida
- Saha Cardiovascular Research Center (H.S., M.K.F., J.J.M., D.A.H., M.K., H.S.L., A.D.), University of Kentucky, Lexington
| | - Hong S Lu
- Saha Cardiovascular Research Center (H.S., M.K.F., J.J.M., D.A.H., M.K., H.S.L., A.D.), University of Kentucky, Lexington.,Department of Physiology (H.S.L., A.D.), University of Kentucky, Lexington
| | - Alan Daugherty
- Saha Cardiovascular Research Center (H.S., M.K.F., J.J.M., D.A.H., M.K., H.S.L., A.D.), University of Kentucky, Lexington.,Department of Physiology (H.S.L., A.D.), University of Kentucky, Lexington
| |
Collapse
|
26
|
Hiromi T, Yokoyama U, Kurotaki D, Mamun A, Ishiwata R, Ichikawa Y, Nishihara H, Umemura M, Fujita T, Yasuda S, Minami T, Goda M, Uchida K, Suzuki S, Takeuchi I, Masuda M, Breyer RM, Tamura T, Ishikawa Y. Excessive EP4 Signaling in Smooth Muscle Cells Induces Abdominal Aortic Aneurysm by Amplifying Inflammation. Arterioscler Thromb Vasc Biol 2020; 40:1559-1573. [PMID: 32321307 PMCID: PMC7253191 DOI: 10.1161/atvbaha.120.314297] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
OBJECTIVE Excessive prostaglandin E2 production is a hallmark of abdominal aortic aneurysm (AAA). Enhanced expression of prostaglandin E2 receptor EP4 (prostaglandin E receptor 4) in vascular smooth muscle cells (VSMCs) has been demonstrated in human AAAs. Although moderate expression of EP4 contributes to vascular homeostasis, the roles of excessive EP4 in vascular pathology remain uncertain. We aimed to investigate whether EP4 overexpression in VSMCs exacerbates AAAs. Approach and Results: We constructed mice with EP4 overexpressed selectively in VSMCs under an SM22α promoter (EP4-Tg). Most EP4-Tg mice died within 2 weeks of Ang II (angiotensin II) infusion due to AAA, while nontransgenic mice given Ang II displayed no overt phenotype. EP4-Tg developed much larger AAAs than nontransgenic mice after periaortic CaCl2 application. In contrast, EP4fl/+;SM22-Cre;ApoE-/- and EP4fl/+;SM22-Cre mice, which are EP4 heterozygous knockout in VSMCs, rarely exhibited AAA after Ang II or CaCl2 treatment, respectively. In Ang II-infused EP4-Tg aorta, Ly6Chi inflammatory monocyte/macrophage infiltration and MMP-9 (matrix metalloprotease-9) activation were enhanced. An unbiased analysis revealed that EP4 stimulation positively regulated the genes binding cytokine receptors in VSMCs, in which IL (interleukin)-6 was the most strongly upregulated. In VSMCs of EP4-Tg and human AAAs, EP4 stimulation caused marked IL-6 production via TAK1 (transforming growth factor-β-activated kinase 1), NF-κB (nuclear factor-kappa B), JNK (c-Jun N-terminal kinase), and p38. Inhibition of IL-6 prevented Ang II-induced AAA formation in EP4-Tg. In addition, EP4 stimulation decreased elastin/collagen cross-linking protein LOX (lysyl oxidase) in both human and mouse VSMCs. CONCLUSIONS Dysregulated EP4 overexpression in VSMCs promotes inflammatory monocyte/macrophage infiltration and attenuates elastin/collagen fiber formation, leading to AAA exacerbation.
Collapse
Affiliation(s)
- Taro Hiromi
- From the Cardiovascular Research Institute (T.H., U.Y., A.M., R.I., Y.I., M.U., T.F., Y.I.), Yokohama City University, Japan.,Department of Emergency Medicine (T.H., I.T.), Yokohama City University Graduate School of Medicine, Japan
| | - Utako Yokoyama
- From the Cardiovascular Research Institute (T.H., U.Y., A.M., R.I., Y.I., M.U., T.F., Y.I.), Yokohama City University, Japan.,Department of Physiology, Tokyo Medical University, Japan (U.Y.)
| | - Daisuke Kurotaki
- Department of Immunology (D.K., T.T.), Yokohama City University Graduate School of Medicine, Japan
| | - Al Mamun
- From the Cardiovascular Research Institute (T.H., U.Y., A.M., R.I., Y.I., M.U., T.F., Y.I.), Yokohama City University, Japan
| | - Ryo Ishiwata
- From the Cardiovascular Research Institute (T.H., U.Y., A.M., R.I., Y.I., M.U., T.F., Y.I.), Yokohama City University, Japan
| | - Yasuhiro Ichikawa
- From the Cardiovascular Research Institute (T.H., U.Y., A.M., R.I., Y.I., M.U., T.F., Y.I.), Yokohama City University, Japan
| | - Hiroshi Nishihara
- Keio Cancer Center, Keio University School of Medicine, Tokyo, Japan (H.N.)
| | - Masanari Umemura
- From the Cardiovascular Research Institute (T.H., U.Y., A.M., R.I., Y.I., M.U., T.F., Y.I.), Yokohama City University, Japan
| | - Takayuki Fujita
- From the Cardiovascular Research Institute (T.H., U.Y., A.M., R.I., Y.I., M.U., T.F., Y.I.), Yokohama City University, Japan
| | - Shota Yasuda
- Department of Surgery (S.Y., M.G., S.S., M.M.), Yokohama City University, Japan
| | - Tomoyuki Minami
- Cardiovascular Center, Yokohama City University Medical Center, Japan (T.M., K.U.)
| | - Motohiko Goda
- Department of Surgery (S.Y., M.G., S.S., M.M.), Yokohama City University, Japan
| | - Keiji Uchida
- Cardiovascular Center, Yokohama City University Medical Center, Japan (T.M., K.U.)
| | - Shinichi Suzuki
- Department of Surgery (S.Y., M.G., S.S., M.M.), Yokohama City University, Japan
| | - Ichiro Takeuchi
- Department of Emergency Medicine (T.H., I.T.), Yokohama City University Graduate School of Medicine, Japan
| | - Munetaka Masuda
- Department of Surgery (S.Y., M.G., S.S., M.M.), Yokohama City University, Japan
| | - Richard M Breyer
- Department of Medicine, Vanderbilt University, Nashville, TN (R.M.B.)
| | - Tomohiko Tamura
- Department of Immunology (D.K., T.T.), Yokohama City University Graduate School of Medicine, Japan
| | - Yoshihiro Ishikawa
- From the Cardiovascular Research Institute (T.H., U.Y., A.M., R.I., Y.I., M.U., T.F., Y.I.), Yokohama City University, Japan
| |
Collapse
|
27
|
Shen YH, LeMaire SA, Webb NR, Cassis LA, Daugherty A, Lu HS. Aortic Aneurysms and Dissections Series. Arterioscler Thromb Vasc Biol 2020; 40:e37-e46. [PMID: 32101472 DOI: 10.1161/atvbaha.120.313991] [Citation(s) in RCA: 72] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The aortic wall is composed of highly dynamic cell populations and extracellular matrix. In response to changes in the biomechanical environment, aortic cells and extracellular matrix modulate their structure and functions to increase aortic wall strength and meet the hemodynamic demand. Compromise in the structural and functional integrity of aortic components leads to aortic degeneration, biomechanical failure, and the development of aortic aneurysms and dissections (AAD). A better understanding of the molecular pathogenesis of AAD will facilitate the development of effective medications to treat these conditions. Here, we summarize recent findings on AAD published in ATVB. In this issue, we focus on the dynamics of aortic cells and extracellular matrix in AAD; in the next issue, we will focus on the role of signaling pathways in AAD.
Collapse
Affiliation(s)
- Ying H Shen
- From the Division of Cardiothoracic Surgery, Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, TX (Y.H.S., S.A.L.).,Department of Cardiovascular Surgery, Texas Heart Institute, Houston (Y.H.S., S.A.L.)
| | - Scott A LeMaire
- From the Division of Cardiothoracic Surgery, Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, TX (Y.H.S., S.A.L.).,Department of Cardiovascular Surgery, Texas Heart Institute, Houston (Y.H.S., S.A.L.)
| | - Nancy R Webb
- Department of Pharmacology and Nutritional Sciences (N.R.W., L.A.C.), University of Kentucky, Lexington
| | - Lisa A Cassis
- Department of Pharmacology and Nutritional Sciences (N.R.W., L.A.C.), University of Kentucky, Lexington
| | - Alan Daugherty
- Department of Physiology and Saha Cardiovascular Research Center (A.D., H.S.L.), University of Kentucky, Lexington
| | - Hong S Lu
- Department of Physiology and Saha Cardiovascular Research Center (A.D., H.S.L.), University of Kentucky, Lexington
| |
Collapse
|
28
|
Davis FM, Daugherty A, Lu HS. Updates of Recent Aortic Aneurysm Research. Arterioscler Thromb Vasc Biol 2020; 39:e83-e90. [PMID: 30811252 DOI: 10.1161/atvbaha.119.312000] [Citation(s) in RCA: 73] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Affiliation(s)
- Frank M Davis
- From the Department of Surgery, University of Michigan, Ann Arbor (F.M.D.)
| | - Alan Daugherty
- Saha Cardiovascular Research Center (A.D., H.S.L.), University of Kentucky, Lexington.,Department of Physiology (A.D., H.S.L.), University of Kentucky, Lexington
| | - Hong S Lu
- Saha Cardiovascular Research Center (A.D., H.S.L.), University of Kentucky, Lexington.,Department of Physiology (A.D., H.S.L.), University of Kentucky, Lexington
| |
Collapse
|
29
|
Au DT, Arai AL, Fondrie WE, Muratoglu SC, Strickland DK. Role of the LDL Receptor-Related Protein 1 in Regulating Protease Activity and Signaling Pathways in the Vasculature. Curr Drug Targets 2019; 19:1276-1288. [PMID: 29749311 DOI: 10.2174/1389450119666180511162048] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2017] [Revised: 04/24/2018] [Accepted: 04/24/2018] [Indexed: 12/22/2022]
Abstract
Aortic aneurysms represent a significant clinical problem as they largely go undetected until a rupture occurs. Currently, an understanding of mechanisms leading to aneurysm formation is limited. Numerous studies clearly indicate that vascular smooth muscle cells play a major role in the development and response of the vasculature to hemodynamic changes and defects in these responses can lead to aneurysm formation. The LDL receptor-related protein 1 (LRP1) is major smooth muscle cell receptor that has the capacity to mediate the endocytosis of numerous ligands and to initiate and regulate signaling pathways. Genetic evidence in humans and mouse models reveal a critical role for LRP1 in maintaining the integrity of the vasculature. Understanding the mechanisms by which this is accomplished represents an important area of research, and likely involves LRP1's ability to regulate levels of proteases known to degrade the extracellular matrix as well as its ability to modulate signaling events.
Collapse
Affiliation(s)
- Dianaly T Au
- Center for Vascular and Inflammatory Diseases, Biopark I, R213, 800 W. Baltimore Street, Baltimore, Maryland 21201, MD, United States
| | - Allison L Arai
- Center for Vascular and Inflammatory Diseases, Biopark I, R213, 800 W. Baltimore Street, Baltimore, Maryland 21201, MD, United States
| | - William E Fondrie
- Center for Vascular and Inflammatory Diseases, Biopark I, R213, 800 W. Baltimore Street, Baltimore, Maryland 21201, MD, United States
| | - Selen C Muratoglu
- Center for Vascular and Inflammatory Diseases, Biopark I, R213, 800 W. Baltimore Street, Baltimore, Maryland 21201, MD, United States.,Department of Physiology, University of Maryland School of Medicine, Baltimore, Maryland 21201, MD, United States
| | - Dudley K Strickland
- Center for Vascular and Inflammatory Diseases, Biopark I, R213, 800 W. Baltimore Street, Baltimore, Maryland 21201, MD, United States.,Department of Physiology, University of Maryland School of Medicine, Baltimore, Maryland 21201, MD, United States.,Department of Surgery, University of Maryland School of Medicine, Baltimore, Maryland 21201, MD, United States
| |
Collapse
|
30
|
Sawada H, Chen JZ, Wright BC, Moorleghen JJ, Lu HS, Daugherty A. Ultrasound Imaging of the Thoracic and Abdominal Aorta in Mice to Determine Aneurysm Dimensions. J Vis Exp 2019. [PMID: 30907888 DOI: 10.3791/59013] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
Contemporary high-resolution ultrasound instruments have sufficient resolution to facilitate the measurement of mouse aortas. These instruments have been widely used to measure aortic dimensions in mouse models of aortic aneurysms. Aortic aneurysms are defined as permanent dilations of the aorta, which occur most frequently in the ascending and abdominal regions. Sequential measurements of aortic dimensions by ultrasound are the principal approach for assessing the development and progression of aortic aneurysms in vivo. Although many reported studies used ultrasound imaging to measure aortic diameters as a primary endpoint, there are confounding factors, such as probe position and cardiac cycle, that may impact the accuracy of data acquisition, analysis, and interpretation. The purpose of this protocol is to provide a practical guide on the use of ultrasound to measure the aortic diameter in a reliable and reproducible manner. This protocol introduces the preparation of mice and instruments, the acquisition of appropriate ultrasound images, and data analysis.
Collapse
Affiliation(s)
- Hisashi Sawada
- Saha Cardiovascular Research Center, University of Kentucky
| | - Jeff Z Chen
- Department of Physiology, University of Kentucky
| | | | | | - Hong S Lu
- Saha Cardiovascular Research Center, University of Kentucky; Department of Physiology, University of Kentucky
| | - Alan Daugherty
- Saha Cardiovascular Research Center, University of Kentucky; Department of Physiology, University of Kentucky;
| |
Collapse
|
31
|
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
|
32
|
Au DT, Ying Z, Hernández-Ochoa EO, Fondrie WE, Hampton B, Migliorini M, Galisteo R, Schneider MF, Daugherty A, Rateri DL, Strickland DK, Muratoglu SC. LRP1 (Low-Density Lipoprotein Receptor-Related Protein 1) Regulates Smooth Muscle Contractility by Modulating Ca 2+ Signaling and Expression of Cytoskeleton-Related Proteins. Arterioscler Thromb Vasc Biol 2018; 38:2651-2664. [PMID: 30354243 PMCID: PMC6214382 DOI: 10.1161/atvbaha.118.311197] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2017] [Accepted: 09/12/2018] [Indexed: 01/12/2023]
Abstract
Objective- Mutations affecting contractile-related proteins in the ECM (extracellular matrix), microfibrils, or vascular smooth muscle cells can predispose the aorta to aneurysms. We reported previously that the LRP1 (low-density lipoprotein receptor-related protein 1) maintains vessel wall integrity, and smLRP1-/- mice exhibited aortic dilatation. The current study focused on defining the mechanisms by which LRP1 regulates vessel wall function and integrity. Approach and Results- Isometric contraction assays demonstrated that vasoreactivity of LRP1-deficient aortic rings was significantly attenuated when stimulated with vasoconstrictors, including phenylephrine, thromboxane receptor agonist U-46619, increased potassium, and L-type Ca2+ channel ligand FPL-64176. Quantitative proteomics revealed proteins involved in actin polymerization and contraction were significantly downregulated in aortas of smLRP1-/- mice. However, studies with calyculin A indicated that although aortic muscle from smLRP1-/- mice can contract in response to calyculin A, a role for LRP1 in regulating the contractile machinery is not revealed. Furthermore, intracellular calcium imaging experiments identified defects in calcium release in response to a RyR (ryanodine receptor) agonist in smLRP1-/- aortic rings and cultured vascular smooth muscle cells. Conclusions- These results identify a critical role for LRP1 in modulating vascular smooth muscle cell contraction by regulating calcium signaling events that potentially protect against aneurysm development.
Collapse
MESH Headings
- Actin Cytoskeleton/drug effects
- Actin Cytoskeleton/genetics
- Actin Cytoskeleton/metabolism
- Actin Cytoskeleton/ultrastructure
- Animals
- Aorta/metabolism
- Calcium Channels/genetics
- Calcium Channels/metabolism
- Calcium Signaling/drug effects
- Cytoskeletal Proteins/genetics
- Cytoskeletal Proteins/metabolism
- Female
- Gene Expression Regulation
- Low Density Lipoprotein Receptor-Related Protein-1
- Male
- Mice, Knockout
- Muscle, Smooth, Vascular/drug effects
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/ultrastructure
- Receptors, LDL/deficiency
- Receptors, LDL/genetics
- Receptors, LDL/metabolism
- Ryanodine Receptor Calcium Release Channel/genetics
- Ryanodine Receptor Calcium Release Channel/metabolism
- Tissue Culture Techniques
- Tumor Suppressor Proteins/deficiency
- Tumor Suppressor Proteins/genetics
- Tumor Suppressor Proteins/metabolism
- Vasoconstriction/drug effects
- Vasoconstrictor Agents/pharmacology
Collapse
Affiliation(s)
- Dianaly T. Au
- Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Zhekang Ying
- Department of Medicine Cardiology Division, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Erick O. Hernández-Ochoa
- Department of Biochemistry and Molecular Biology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - William E. Fondrie
- Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Brian Hampton
- Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Mary Migliorini
- Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, MD 21201, USA
- Saha Cardiovascular Research Center and Department of Physiology, University of Kentucky, Lexington, KY 40536, USA
| | - Rebeca Galisteo
- Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Martin F. Schneider
- Department of Biochemistry and Molecular Biology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Alan Daugherty
- Saha Cardiovascular Research Center and Department of Physiology, University of Kentucky, Lexington, KY 40536, USA
| | - Debra L. Rateri
- Saha Cardiovascular Research Center and Department of Physiology, University of Kentucky, Lexington, KY 40536, USA
| | - Dudley K. Strickland
- Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, MD 21201, USA
- Department of Surgery, University of Maryland School of Medicine, Baltimore, MD 21201, USA
- Department of Physiology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Selen C. Muratoglu
- Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, MD 21201, USA
- Department of Physiology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| |
Collapse
|
33
|
Munshaw S, Bruche S, Redpath AN, Patel J, Dubé KN, Channon KM, Smart N. O4 THYMOSIN β4 MEDIATES VASCULAR PROTECTION VIA INTERACTION WITH LOW DENSITY LIPOPROTEIN RECEPTOR RELATED PROTEIN 1 (LRP1). Cardiovasc Res 2018. [DOI: 10.1093/cvr/cvy216.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Affiliation(s)
- S Munshaw
- Department of Physiology, Anatomy & Genetics, University of Oxford, Sherrington Building, South Parks Road, Oxford, UK
| | - S Bruche
- Department of Physiology, Anatomy & Genetics, University of Oxford, Sherrington Building, South Parks Road, Oxford, UK
| | - A N Redpath
- Department of Physiology, Anatomy & Genetics, University of Oxford, Sherrington Building, South Parks Road, Oxford, UK
| | - J Patel
- Division of Cardiovascular Medicine, University of Oxford, John Radcliffe Hospital, Oxford, UK
| | - K N Dubé
- Department of Physiology, Anatomy & Genetics, University of Oxford, Sherrington Building, South Parks Road, Oxford, UK
| | - K M Channon
- Division of Cardiovascular Medicine, University of Oxford, John Radcliffe Hospital, Oxford, UK
| | - N Smart
- Department of Physiology, Anatomy & Genetics, University of Oxford, Sherrington Building, South Parks Road, Oxford, UK
| |
Collapse
|
34
|
Fava M, Barallobre-Barreiro J, Mayr U, Lu R, Didangelos A, Baig F, Lynch M, Catibog N, Joshi A, Barwari T, Yin X, Jahangiri M, Mayr M. Role of ADAMTS-5 in Aortic Dilatation and Extracellular Matrix Remodeling. Arterioscler Thromb Vasc Biol 2018; 38:1537-1548. [PMID: 29622560 PMCID: PMC6026471 DOI: 10.1161/atvbaha.117.310562] [Citation(s) in RCA: 65] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2017] [Accepted: 03/19/2018] [Indexed: 11/16/2022]
Abstract
Supplemental Digital Content is available in the text. Objective— Thoracic aortic aneurysm (TAA), a degenerative disease of the aortic wall, is accompanied by changes in the structure and composition of the aortic ECM (extracellular matrix). The ADAMTS (a disintegrin and metalloproteinase with thrombospondin motifs) family of proteases has recently been implicated in TAA formation. This study aimed to investigate the contribution of ADAMTS-5 to TAA development. Approach and Results— A model of aortic dilatation by AngII (angiotensin II) infusion was adopted in mice lacking the catalytic domain of ADAMTS-5 (Adamts5Δcat). Adamts5Δcat mice showed an attenuated rise in blood pressure while displaying increased dilatation of the ascending aorta (AsAo). Interestingly, a proteomic comparison of the aortic ECM from AngII-treated wild-type and Adamts5Δcat mice revealed versican as the most upregulated ECM protein in Adamts5Δcat mice. This was accompanied by a marked reduction of ADAMTS-specific versican cleavage products (versikine) and a decrease of LRP1 (low-density lipoprotein-related protein 1). Silencing LRP1 expression in human aortic smooth muscle cells reduced the expression of ADAMTS5, attenuated the generation of versikine, but increased soluble ADAMTS-1. A similar increase in ADAMTS-1 was observed in aortas of AngII-treated Adamts5Δcat mice but was not sufficient to maintain versican processing and prevent aortic dilatation. Conclusions— Our results support the emerging role of ADAMTS proteases in TAA. ADAMTS-5 rather than ADAMTS-1 is the key protease for versican regulation in murine aortas. Further studies are needed to define the ECM substrates of the different ADAMTS proteases and their contribution to TAA formation.
Collapse
MESH Headings
- ADAMTS1 Protein/metabolism
- ADAMTS5 Protein/deficiency
- ADAMTS5 Protein/genetics
- ADAMTS5 Protein/metabolism
- Angiotensin II
- Animals
- Aorta, Thoracic/enzymology
- Aorta, Thoracic/pathology
- Aortic Aneurysm, Thoracic/chemically induced
- Aortic Aneurysm, Thoracic/enzymology
- Aortic Aneurysm, Thoracic/genetics
- Aortic Aneurysm, Thoracic/pathology
- Cells, Cultured
- Dilatation, Pathologic
- Disease Models, Animal
- Extracellular Matrix/enzymology
- Extracellular Matrix/pathology
- Humans
- Low Density Lipoprotein Receptor-Related Protein-1/genetics
- Low Density Lipoprotein Receptor-Related Protein-1/metabolism
- Male
- Mice, Knockout
- Muscle, Smooth, Vascular/enzymology
- Myocytes, Smooth Muscle
- Receptors, LDL/metabolism
- Tumor Suppressor Proteins/metabolism
- Vascular Remodeling
- Versicans/metabolism
Collapse
Affiliation(s)
- Marika Fava
- From the King's British Heart Foundation Centre, King's College London, United Kingdom (M.F., J.B.-B., U.M., R.L., A.D., F.B., M.L., N.C., A.J., T.B., X.Y., M.M.)
- St George's University of London, NHS Trust, United Kingdom (M.F., M.J.)
- Cardiovascular Institute, Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York (M.F., M.M.)
| | - Javier Barallobre-Barreiro
- From the King's British Heart Foundation Centre, King's College London, United Kingdom (M.F., J.B.-B., U.M., R.L., A.D., F.B., M.L., N.C., A.J., T.B., X.Y., M.M.)
| | - Ursula Mayr
- From the King's British Heart Foundation Centre, King's College London, United Kingdom (M.F., J.B.-B., U.M., R.L., A.D., F.B., M.L., N.C., A.J., T.B., X.Y., M.M.)
| | - Ruifang Lu
- From the King's British Heart Foundation Centre, King's College London, United Kingdom (M.F., J.B.-B., U.M., R.L., A.D., F.B., M.L., N.C., A.J., T.B., X.Y., M.M.)
| | - Athanasios Didangelos
- From the King's British Heart Foundation Centre, King's College London, United Kingdom (M.F., J.B.-B., U.M., R.L., A.D., F.B., M.L., N.C., A.J., T.B., X.Y., M.M.)
| | - Ferheen Baig
- From the King's British Heart Foundation Centre, King's College London, United Kingdom (M.F., J.B.-B., U.M., R.L., A.D., F.B., M.L., N.C., A.J., T.B., X.Y., M.M.)
| | - Marc Lynch
- From the King's British Heart Foundation Centre, King's College London, United Kingdom (M.F., J.B.-B., U.M., R.L., A.D., F.B., M.L., N.C., A.J., T.B., X.Y., M.M.)
| | - Norman Catibog
- From the King's British Heart Foundation Centre, King's College London, United Kingdom (M.F., J.B.-B., U.M., R.L., A.D., F.B., M.L., N.C., A.J., T.B., X.Y., M.M.)
| | - Abhishek Joshi
- From the King's British Heart Foundation Centre, King's College London, United Kingdom (M.F., J.B.-B., U.M., R.L., A.D., F.B., M.L., N.C., A.J., T.B., X.Y., M.M.)
| | - Temo Barwari
- From the King's British Heart Foundation Centre, King's College London, United Kingdom (M.F., J.B.-B., U.M., R.L., A.D., F.B., M.L., N.C., A.J., T.B., X.Y., M.M.)
| | - Xiaoke Yin
- From the King's British Heart Foundation Centre, King's College London, United Kingdom (M.F., J.B.-B., U.M., R.L., A.D., F.B., M.L., N.C., A.J., T.B., X.Y., M.M.)
| | - Marjan Jahangiri
- St George's University of London, NHS Trust, United Kingdom (M.F., M.J.)
| | - Manuel Mayr
- From the King's British Heart Foundation Centre, King's College London, United Kingdom (M.F., J.B.-B., U.M., R.L., A.D., F.B., M.L., N.C., A.J., T.B., X.Y., M.M.)
- Cardiovascular Institute, Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York (M.F., M.M.)
| |
Collapse
|
35
|
Boukais K, Borges LF, Venisse L, Touat Z, François D, Arocas V, Jondeau G, Declerck P, Bouton MC, Michel JB. Clearance of plasmin-PN-1 complexes by vascular smooth muscle cells in human aneurysm of the ascending aorta. Cardiovasc Pathol 2017; 32:15-25. [PMID: 29149696 DOI: 10.1016/j.carpath.2017.10.002] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/03/2017] [Revised: 09/06/2017] [Accepted: 10/18/2017] [Indexed: 12/20/2022] Open
Abstract
Plasminogen is a circulating zymogen which enters the arterial wall by radial, transmural hydraulic conductance, where it is converted to plasmin by tissue plasminogen activator t-PA on an activation platform involving S100A4 on the vascular smooth muscle cell (vSMC) membrane. Plasmin is involved in the progression of human thoracic aneurysm of the ascending aorta (TAA). vSMCs protect the TAA wall from plasmin-induced proteolytic injury by expressing high levels of antiproteases. Protease nexin-1 (PN-1) is a tissue antiprotease belonging to the serpin superfamily, expressed in the vascular wall, and is able to form a covalent complex with plasmin. LDL receptor-related protein-1 (LRP-1) is a scavenger receptor implicated in protease-antiprotease complex internalization. In this study, we investigated whether PN-1 and LRP-1 are involved in the inhibition and clearance of plasminogen by the SMCs of human TAA. We demonstrated an overexpression of S100A4, PN-1, and LRP-1 in the medial layer of human TAA. Plasminogen activation taking place in the media of TAA was revealed by immunohistochemical staining and plasmin activity analyses. We showed by cell biology studies that plasmin-PN-1 complexes are internalized via LRP-1 in vSMCs from healthy and TAA media. Thus, two complementary mechanisms are involved in the protective role of PN-1 in human TAA: one involving plasmin inhibition and the other involving tissue clearance of plasmin-PN1 complexes via the scavenger receptor LRP-1.
Collapse
Affiliation(s)
- Kamel Boukais
- UMR 1148, Laboratory for Vascular Translational Science, Inserm; Paris7 Denis Diderot University
| | - Luciano F Borges
- Heart Institute (InCor), Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil; Department of Biological science, Federal University of São Paulo, São Paulo, Brazil
| | - Laurence Venisse
- UMR 1148, Laboratory for Vascular Translational Science, Inserm; Paris7 Denis Diderot University
| | - Ziad Touat
- UMR 1148, Laboratory for Vascular Translational Science, Inserm
| | - Déborah François
- UMR 1148, Laboratory for Vascular Translational Science, Inserm; Paris7 Denis Diderot University
| | - Véronique Arocas
- UMR 1148, Laboratory for Vascular Translational Science, Inserm; Paris7 Denis Diderot University
| | - Guillaume Jondeau
- UMR 1148, Laboratory for Vascular Translational Science, Inserm; Centre national de Référence pour le Syndrome de Marfan et apparentés, Hôpital Xavier Bichat
| | - Paul Declerck
- Laboratory for Therapeutic and Diagnostic Antibodies, Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, Leuven, Belgium
| | - Marie-Christine Bouton
- UMR 1148, Laboratory for Vascular Translational Science, Inserm; Paris7 Denis Diderot University
| | - Jean-Baptiste Michel
- UMR 1148, Laboratory for Vascular Translational Science, Inserm; Paris7 Denis Diderot University.
| |
Collapse
|
36
|
MESH Headings
- Animals
- Aorta, Abdominal/metabolism
- Aorta, Abdominal/pathology
- Aorta, Abdominal/physiopathology
- Aorta, Thoracic/metabolism
- Aorta, Thoracic/pathology
- Aorta, Thoracic/physiopathology
- Aortic Aneurysm, Abdominal/epidemiology
- Aortic Aneurysm, Abdominal/metabolism
- Aortic Aneurysm, Abdominal/pathology
- Aortic Aneurysm, Abdominal/physiopathology
- Aortic Aneurysm, Thoracic/epidemiology
- Aortic Aneurysm, Thoracic/metabolism
- Aortic Aneurysm, Thoracic/pathology
- Aortic Aneurysm, Thoracic/physiopathology
- Disease Models, Animal
- Humans
- Risk Factors
- Signal Transduction
- Vascular Remodeling
Collapse
Affiliation(s)
- Hong Lu
- From the Department of Physiology, Saha Cardiovascular Research Center, University of Kentucky, Lexington.
| | - Alan Daugherty
- From the Department of Physiology, Saha Cardiovascular Research Center, University of Kentucky, Lexington
| |
Collapse
|
37
|
Cavanaugh NB, Qian L, Westergaard NM, Kutschke WJ, Born EJ, Turek JW. A Novel Murine Model of Marfan Syndrome Accelerates Aortopathy and Cardiomyopathy. Ann Thorac Surg 2017; 104:657-665. [PMID: 28347539 DOI: 10.1016/j.athoracsur.2016.10.077] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/25/2016] [Revised: 10/09/2016] [Accepted: 10/17/2016] [Indexed: 01/04/2023]
Abstract
BACKGROUND Marfan syndrome (MFS) represents a genetic disorder with variable phenotypic expression. The main cardiovascular sequelae of MFS include aortic aneurysm/dissection and cardiomyopathy. Although significant advances in the understanding of transforming growth factor beta signaling have led to promising therapeutic targets for the treatment of aortopathy, clinical studies have tempered this optimism. In particular, these studies suggest additional signaling pathways that play a significant role in disease progression. To date, studies aimed at elucidating molecular mechanisms involved in MFS-induced disease progression have been hampered by the lack of an accelerated disease model. METHODS Wild-type B6.129 mice and MFS Fbn1C1039G/+ mice underwent subcutaneous, cervical osmotic minipump installation with sodium chloride (wild-type mice, n = 39; MFS mice, n = 12) or angiotensin II, 4.5 mg/kg daily (wild-type mice, n = 11; MFS mice; n = 35) for as long as 28 days. Hemodynamic measurements were obtained throughout the experiment. Aortas and hearts were analyzed by transthoracic echocardiography and histopathology study. RESULTS This accelerated murine MFS model replicates increased mortality from MFS-related maladies (20.0%, 39.3%, and 52.9% at 10, 14, and 28 days, respectively). Aortic diameters in accelerated MFS mice were significantly enlarged at 10 days after minipump implantation and correlated with a higher degree of elastin fragmentation. Accelerated MFS mice also demonstrated dilated cardiomyopathy at 14 days, even without aortic insufficiency, suggesting an intrinsic etiology. CONCLUSIONS A novel in vivo model consisting of subcutaneously delivered angiotensin II in MFS mice reproducibly causes accelerated aortic aneurysm formation and cardiomyopathy. This model allows for better investigation of MFS sequelae by rapid experimental processes.
Collapse
Affiliation(s)
- Nicholas B Cavanaugh
- Department of Cardiothoracic Surgery, Francois M. Abboud Cardiovascular Research Center, University of Iowa Carver College of Medicine, Iowa City, Iowa
| | - Lan Qian
- Department of Cardiothoracic Surgery, Francois M. Abboud Cardiovascular Research Center, University of Iowa Carver College of Medicine, Iowa City, Iowa
| | - Nicole M Westergaard
- Department of Cardiothoracic Surgery, Francois M. Abboud Cardiovascular Research Center, University of Iowa Carver College of Medicine, Iowa City, Iowa
| | - William J Kutschke
- Department of Internal Medicine, Francois M. Abboud Cardiovascular Research Center, University of Iowa Carver College of Medicine, Iowa City, Iowa
| | - Ella J Born
- Department of Cardiothoracic Surgery, Francois M. Abboud Cardiovascular Research Center, University of Iowa Carver College of Medicine, Iowa City, Iowa
| | - Joseph W Turek
- Department of Cardiothoracic Surgery, Francois M. Abboud Cardiovascular Research Center, University of Iowa Carver College of Medicine, Iowa City, Iowa.
| |
Collapse
|
38
|
Shen YH, LeMaire SA. Molecular pathogenesis of genetic and sporadic aortic aneurysms and dissections. Curr Probl Surg 2017; 54:95-155. [PMID: 28521856 DOI: 10.1067/j.cpsurg.2017.01.001] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2016] [Accepted: 01/16/2017] [Indexed: 12/20/2022]
Affiliation(s)
- Ying H Shen
- Division of Cardiothoracic Surgery, Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, TX; Department of Cardiovascular Surgery, Texas Heart Institute, Houston, TX; Cardiovascular Research Institute, Baylor College of Medicine, Houston, TX.
| | - Scott A LeMaire
- Division of Cardiothoracic Surgery, Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, TX; Department of Cardiovascular Surgery, Texas Heart Institute, Houston, TX; Cardiovascular Research Institute, Baylor College of Medicine, Houston, TX; Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, TX.
| |
Collapse
|
39
|
Wu D, Ren P, Zheng Y, Zhang L, Xu G, Xie W, Lloyd EE, Zhang S, Zhang Q, Curci JA, Coselli JS, Milewicz DM, Shen YH, LeMaire SA. NLRP3 (Nucleotide Oligomerization Domain-Like Receptor Family, Pyrin Domain Containing 3)-Caspase-1 Inflammasome Degrades Contractile Proteins: Implications for Aortic Biomechanical Dysfunction and Aneurysm and Dissection Formation. Arterioscler Thromb Vasc Biol 2017; 37:694-706. [PMID: 28153878 DOI: 10.1161/atvbaha.116.307648] [Citation(s) in RCA: 84] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2016] [Accepted: 01/24/2017] [Indexed: 01/15/2023]
Abstract
OBJECTIVE Increasing evidence suggests that contractile dysfunction in smooth muscle cells (SMCs) plays a critical role in aortic biomechanical dysfunction and aortic aneurysm and dissection (AAD) development. However, the mechanisms underlying SMC contractile dysfunction in sporadic AAD are poorly understood. In this study, we examined the role of the NLRP3 (nucleotide oligomerization domain-like receptor family, pyrin domain containing 3)-caspase-1 inflammasome, a key inflammatory cascade, in SMC contractile dysfunction in AAD. APPROACH AND RESULTS We observed significant SMC contractile protein degradation in aortas from patients with sporadic thoracic AAD. The contractile protein degradation was associated with activation of the NLRP3-caspase-1 inflammasome cascade. In SMCs, caspase-1 bound and directly cleaved and degraded contractile proteins, leading to contractile dysfunction. Furthermore, Nlrp3 or caspase-1 deficiency in mice significantly reduced angiotensin II-induced contractile protein degradation, biomechanical dysfunction, and AAD formation in both thoracic and abdominal aortas. Finally, blocking this cascade with the inflammasome inhibitor, glyburide (an antidiabetic medication), reduced angiotensin II-induced AAD formation. CONCLUSIONS Inflammasome-caspase-1-mediated degradation of SMC contractile proteins may contribute to aortic biomechanical dysfunction and AAD development. This cascade may be a therapeutic target in AAD formation. In addition, glyburide may have protective effects against AAD development.
Collapse
Affiliation(s)
- Darrell Wu
- From the Division of Cardiothoracic Surgery, Michael E. DeBakey Department of Surgery (D.W., P. R., Y.Z., L.Z., G.X., W.X., J.S.C., Q.Z., Y.H.S., S.A.L.), Cardiovascular Research Institute (J.S.C., Y.H.S., S.A.L.), and Department of Molecular Physiology and Biophysics (D.W., E.E.L., S.A.L.), Baylor College of Medicine, Houston, TX; Department of Cardiovascular Surgery, Texas Heart Institute, Houston (D.W., P.R., G.X., L.Z., W.X., J.S.C., Y.H.S., S.A.L.); Cardiology MD Anderson Cancer Center, Houston, Texas (S.Z.); Division of Vascular Surgery, Vanderbilt University School of Medicine, Nashville, TN (J.A.C.); and Division of Medical Genetics, Department of Internal Medicine, The University of Texas Health Science Center at Houston (D.M.M)
| | - Pingping Ren
- From the Division of Cardiothoracic Surgery, Michael E. DeBakey Department of Surgery (D.W., P. R., Y.Z., L.Z., G.X., W.X., J.S.C., Q.Z., Y.H.S., S.A.L.), Cardiovascular Research Institute (J.S.C., Y.H.S., S.A.L.), and Department of Molecular Physiology and Biophysics (D.W., E.E.L., S.A.L.), Baylor College of Medicine, Houston, TX; Department of Cardiovascular Surgery, Texas Heart Institute, Houston (D.W., P.R., G.X., L.Z., W.X., J.S.C., Y.H.S., S.A.L.); Cardiology MD Anderson Cancer Center, Houston, Texas (S.Z.); Division of Vascular Surgery, Vanderbilt University School of Medicine, Nashville, TN (J.A.C.); and Division of Medical Genetics, Department of Internal Medicine, The University of Texas Health Science Center at Houston (D.M.M)
| | - Yanqiu Zheng
- From the Division of Cardiothoracic Surgery, Michael E. DeBakey Department of Surgery (D.W., P. R., Y.Z., L.Z., G.X., W.X., J.S.C., Q.Z., Y.H.S., S.A.L.), Cardiovascular Research Institute (J.S.C., Y.H.S., S.A.L.), and Department of Molecular Physiology and Biophysics (D.W., E.E.L., S.A.L.), Baylor College of Medicine, Houston, TX; Department of Cardiovascular Surgery, Texas Heart Institute, Houston (D.W., P.R., G.X., L.Z., W.X., J.S.C., Y.H.S., S.A.L.); Cardiology MD Anderson Cancer Center, Houston, Texas (S.Z.); Division of Vascular Surgery, Vanderbilt University School of Medicine, Nashville, TN (J.A.C.); and Division of Medical Genetics, Department of Internal Medicine, The University of Texas Health Science Center at Houston (D.M.M)
| | - Lin Zhang
- From the Division of Cardiothoracic Surgery, Michael E. DeBakey Department of Surgery (D.W., P. R., Y.Z., L.Z., G.X., W.X., J.S.C., Q.Z., Y.H.S., S.A.L.), Cardiovascular Research Institute (J.S.C., Y.H.S., S.A.L.), and Department of Molecular Physiology and Biophysics (D.W., E.E.L., S.A.L.), Baylor College of Medicine, Houston, TX; Department of Cardiovascular Surgery, Texas Heart Institute, Houston (D.W., P.R., G.X., L.Z., W.X., J.S.C., Y.H.S., S.A.L.); Cardiology MD Anderson Cancer Center, Houston, Texas (S.Z.); Division of Vascular Surgery, Vanderbilt University School of Medicine, Nashville, TN (J.A.C.); and Division of Medical Genetics, Department of Internal Medicine, The University of Texas Health Science Center at Houston (D.M.M)
| | - Gaiping Xu
- From the Division of Cardiothoracic Surgery, Michael E. DeBakey Department of Surgery (D.W., P. R., Y.Z., L.Z., G.X., W.X., J.S.C., Q.Z., Y.H.S., S.A.L.), Cardiovascular Research Institute (J.S.C., Y.H.S., S.A.L.), and Department of Molecular Physiology and Biophysics (D.W., E.E.L., S.A.L.), Baylor College of Medicine, Houston, TX; Department of Cardiovascular Surgery, Texas Heart Institute, Houston (D.W., P.R., G.X., L.Z., W.X., J.S.C., Y.H.S., S.A.L.); Cardiology MD Anderson Cancer Center, Houston, Texas (S.Z.); Division of Vascular Surgery, Vanderbilt University School of Medicine, Nashville, TN (J.A.C.); and Division of Medical Genetics, Department of Internal Medicine, The University of Texas Health Science Center at Houston (D.M.M)
| | - Wanmu Xie
- From the Division of Cardiothoracic Surgery, Michael E. DeBakey Department of Surgery (D.W., P. R., Y.Z., L.Z., G.X., W.X., J.S.C., Q.Z., Y.H.S., S.A.L.), Cardiovascular Research Institute (J.S.C., Y.H.S., S.A.L.), and Department of Molecular Physiology and Biophysics (D.W., E.E.L., S.A.L.), Baylor College of Medicine, Houston, TX; Department of Cardiovascular Surgery, Texas Heart Institute, Houston (D.W., P.R., G.X., L.Z., W.X., J.S.C., Y.H.S., S.A.L.); Cardiology MD Anderson Cancer Center, Houston, Texas (S.Z.); Division of Vascular Surgery, Vanderbilt University School of Medicine, Nashville, TN (J.A.C.); and Division of Medical Genetics, Department of Internal Medicine, The University of Texas Health Science Center at Houston (D.M.M)
| | - Eric E Lloyd
- From the Division of Cardiothoracic Surgery, Michael E. DeBakey Department of Surgery (D.W., P. R., Y.Z., L.Z., G.X., W.X., J.S.C., Q.Z., Y.H.S., S.A.L.), Cardiovascular Research Institute (J.S.C., Y.H.S., S.A.L.), and Department of Molecular Physiology and Biophysics (D.W., E.E.L., S.A.L.), Baylor College of Medicine, Houston, TX; Department of Cardiovascular Surgery, Texas Heart Institute, Houston (D.W., P.R., G.X., L.Z., W.X., J.S.C., Y.H.S., S.A.L.); Cardiology MD Anderson Cancer Center, Houston, Texas (S.Z.); Division of Vascular Surgery, Vanderbilt University School of Medicine, Nashville, TN (J.A.C.); and Division of Medical Genetics, Department of Internal Medicine, The University of Texas Health Science Center at Houston (D.M.M)
| | - Sui Zhang
- From the Division of Cardiothoracic Surgery, Michael E. DeBakey Department of Surgery (D.W., P. R., Y.Z., L.Z., G.X., W.X., J.S.C., Q.Z., Y.H.S., S.A.L.), Cardiovascular Research Institute (J.S.C., Y.H.S., S.A.L.), and Department of Molecular Physiology and Biophysics (D.W., E.E.L., S.A.L.), Baylor College of Medicine, Houston, TX; Department of Cardiovascular Surgery, Texas Heart Institute, Houston (D.W., P.R., G.X., L.Z., W.X., J.S.C., Y.H.S., S.A.L.); Cardiology MD Anderson Cancer Center, Houston, Texas (S.Z.); Division of Vascular Surgery, Vanderbilt University School of Medicine, Nashville, TN (J.A.C.); and Division of Medical Genetics, Department of Internal Medicine, The University of Texas Health Science Center at Houston (D.M.M)
| | - Qianzi Zhang
- From the Division of Cardiothoracic Surgery, Michael E. DeBakey Department of Surgery (D.W., P. R., Y.Z., L.Z., G.X., W.X., J.S.C., Q.Z., Y.H.S., S.A.L.), Cardiovascular Research Institute (J.S.C., Y.H.S., S.A.L.), and Department of Molecular Physiology and Biophysics (D.W., E.E.L., S.A.L.), Baylor College of Medicine, Houston, TX; Department of Cardiovascular Surgery, Texas Heart Institute, Houston (D.W., P.R., G.X., L.Z., W.X., J.S.C., Y.H.S., S.A.L.); Cardiology MD Anderson Cancer Center, Houston, Texas (S.Z.); Division of Vascular Surgery, Vanderbilt University School of Medicine, Nashville, TN (J.A.C.); and Division of Medical Genetics, Department of Internal Medicine, The University of Texas Health Science Center at Houston (D.M.M)
| | - John A Curci
- From the Division of Cardiothoracic Surgery, Michael E. DeBakey Department of Surgery (D.W., P. R., Y.Z., L.Z., G.X., W.X., J.S.C., Q.Z., Y.H.S., S.A.L.), Cardiovascular Research Institute (J.S.C., Y.H.S., S.A.L.), and Department of Molecular Physiology and Biophysics (D.W., E.E.L., S.A.L.), Baylor College of Medicine, Houston, TX; Department of Cardiovascular Surgery, Texas Heart Institute, Houston (D.W., P.R., G.X., L.Z., W.X., J.S.C., Y.H.S., S.A.L.); Cardiology MD Anderson Cancer Center, Houston, Texas (S.Z.); Division of Vascular Surgery, Vanderbilt University School of Medicine, Nashville, TN (J.A.C.); and Division of Medical Genetics, Department of Internal Medicine, The University of Texas Health Science Center at Houston (D.M.M)
| | - Joseph S Coselli
- From the Division of Cardiothoracic Surgery, Michael E. DeBakey Department of Surgery (D.W., P. R., Y.Z., L.Z., G.X., W.X., J.S.C., Q.Z., Y.H.S., S.A.L.), Cardiovascular Research Institute (J.S.C., Y.H.S., S.A.L.), and Department of Molecular Physiology and Biophysics (D.W., E.E.L., S.A.L.), Baylor College of Medicine, Houston, TX; Department of Cardiovascular Surgery, Texas Heart Institute, Houston (D.W., P.R., G.X., L.Z., W.X., J.S.C., Y.H.S., S.A.L.); Cardiology MD Anderson Cancer Center, Houston, Texas (S.Z.); Division of Vascular Surgery, Vanderbilt University School of Medicine, Nashville, TN (J.A.C.); and Division of Medical Genetics, Department of Internal Medicine, The University of Texas Health Science Center at Houston (D.M.M)
| | - Dianna M Milewicz
- From the Division of Cardiothoracic Surgery, Michael E. DeBakey Department of Surgery (D.W., P. R., Y.Z., L.Z., G.X., W.X., J.S.C., Q.Z., Y.H.S., S.A.L.), Cardiovascular Research Institute (J.S.C., Y.H.S., S.A.L.), and Department of Molecular Physiology and Biophysics (D.W., E.E.L., S.A.L.), Baylor College of Medicine, Houston, TX; Department of Cardiovascular Surgery, Texas Heart Institute, Houston (D.W., P.R., G.X., L.Z., W.X., J.S.C., Y.H.S., S.A.L.); Cardiology MD Anderson Cancer Center, Houston, Texas (S.Z.); Division of Vascular Surgery, Vanderbilt University School of Medicine, Nashville, TN (J.A.C.); and Division of Medical Genetics, Department of Internal Medicine, The University of Texas Health Science Center at Houston (D.M.M)
| | - Ying H Shen
- From the Division of Cardiothoracic Surgery, Michael E. DeBakey Department of Surgery (D.W., P. R., Y.Z., L.Z., G.X., W.X., J.S.C., Q.Z., Y.H.S., S.A.L.), Cardiovascular Research Institute (J.S.C., Y.H.S., S.A.L.), and Department of Molecular Physiology and Biophysics (D.W., E.E.L., S.A.L.), Baylor College of Medicine, Houston, TX; Department of Cardiovascular Surgery, Texas Heart Institute, Houston (D.W., P.R., G.X., L.Z., W.X., J.S.C., Y.H.S., S.A.L.); Cardiology MD Anderson Cancer Center, Houston, Texas (S.Z.); Division of Vascular Surgery, Vanderbilt University School of Medicine, Nashville, TN (J.A.C.); and Division of Medical Genetics, Department of Internal Medicine, The University of Texas Health Science Center at Houston (D.M.M)
| | - Scott A LeMaire
- From the Division of Cardiothoracic Surgery, Michael E. DeBakey Department of Surgery (D.W., P. R., Y.Z., L.Z., G.X., W.X., J.S.C., Q.Z., Y.H.S., S.A.L.), Cardiovascular Research Institute (J.S.C., Y.H.S., S.A.L.), and Department of Molecular Physiology and Biophysics (D.W., E.E.L., S.A.L.), Baylor College of Medicine, Houston, TX; Department of Cardiovascular Surgery, Texas Heart Institute, Houston (D.W., P.R., G.X., L.Z., W.X., J.S.C., Y.H.S., S.A.L.); Cardiology MD Anderson Cancer Center, Houston, Texas (S.Z.); Division of Vascular Surgery, Vanderbilt University School of Medicine, Nashville, TN (J.A.C.); and Division of Medical Genetics, Department of Internal Medicine, The University of Texas Health Science Center at Houston (D.M.M).
| |
Collapse
|
40
|
Chan CYT, Chan YC, Cheuk BLY, Cheng SWK. Clearance of matrix metalloproteinase-9 is dependent on low-density lipoprotein receptor-related protein-1 expression downregulated by microRNA-205 in human abdominal aortic aneurysm. J Vasc Surg 2017; 65:509-520. [DOI: 10.1016/j.jvs.2015.10.065] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2015] [Accepted: 10/03/2015] [Indexed: 01/07/2023]
|
41
|
TRPM8 downregulation by angiotensin II in vascular smooth muscle cells is involved in hypertension. Mol Med Rep 2017; 15:1900-1908. [PMID: 28138709 DOI: 10.3892/mmr.2017.6158] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2015] [Accepted: 12/08/2016] [Indexed: 11/05/2022] Open
Abstract
Angiotensin II (Ang II)-induced injury of vascular smooth muscle cells (VSMCs) serves an important role in hypertension and other cardiovascular disorders. Transient receptor potential melastatin 8 (TRPM8) is a thermally‑regulated Ca2+‑permeable channel that is activated by reduced body temperature. Although several recent studies have revealed the regulatory effect of TRPM8 in vascular tone and hypertension, the precise role of TRPM8 in dysfunction of vascular smooth muscle cells (VSMCs) induced by Ang II remains elusive. In the present study, the possible function of TRPM8 in Ang II‑induced VSMCs malfunction in vivo and in vitro was investigated. In the aortae from rats that had undergone a two‑kidney one‑clip operation, which is a widely‑used renovascular hypertension model, the mRNA and protein levels of TRPM8 were reduced. In addition, exogenous Ang II treatment decreased TRPM8 mRNA and protein expression levels in primary cultures of rat VSMCs. TRPM8 activation by menthol, a pharmacological agonist, in VSMCs, significantly attenuated the Ang II‑induced increase in reactive oxygen species and H2O2 production. In addition, TRPM8 activation reduced the Ang II‑induced upregulation of NADPH oxidase (NOX) 1 and NOX4 in VSMCs. Furthermore, TRPM8 activation relieved the Ang II‑induced activation of ras homolog gene family, member A‑rho associated protein kinase 2 and janus kinase 2 signaling pathways in VSMCs. In conclusion, the results presented in the current study indicated that TRPM8 downregulation by Ang II in VSMCs may be involved in hypertension.
Collapse
|
42
|
Krishna SM, Seto SW, Jose RJ, Li J, Morton SK, Biros E, Wang Y, Nsengiyumva V, Lindeman JHN, Loots GG, Rush CM, Craig JM, Golledge J. Wnt Signaling Pathway Inhibitor Sclerostin Inhibits Angiotensin II-Induced Aortic Aneurysm and Atherosclerosis. Arterioscler Thromb Vasc Biol 2016; 37:553-566. [PMID: 28062506 DOI: 10.1161/atvbaha.116.308723] [Citation(s) in RCA: 117] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2016] [Accepted: 12/07/2016] [Indexed: 01/28/2023]
Abstract
OBJECTIVE Sclerostin (SOST) has been identified as an important regulator of bone formation; however, it has not been previously implicated in arterial disease. The aim of this study was to assess the role of SOST in aortic aneurysm (AA) and atherosclerosis using human samples, a mouse model, and in vitro investigations. APPROACH AND RESULTS SOST protein was downregulated in human and mouse AA samples compared with controls. Transgenic introduction of human SOST in apolipoprotein E-deficient (ApoE-/-) mice (SOSTTg .ApoE-/-) and administration of recombinant mouse Sost inhibited angiotensin II-induced AA and atherosclerosis. Serum concentrations of several proinflammatory cytokines were significantly reduced in SOSTTg .ApoE-/- mice. Compared with controls, the aortas of mice receiving recombinant mouse Sost and SOSTTg .ApoE-/- mice showed reduced matrix degradation, reduced elastin breaks, and preserved collagen. Decreased inflammatory cell infiltration and a reduction in the expression of wingless-type mouse mammary virus integration site/β-catenin responsive genes, including matrix metalloproteinase-9, osteoprotegerin, and osteopontin, were observed in the aortas of SOSTTg .ApoE-/- mice. SOST expression was downregulated and the wingless-type mouse mammary virus integration site/β-catenin pathway was activated in human AA samples. The cytosine-phosphate-guanine islands in the SOST gene promoter showed significantly higher methylation in human AA samples compared with controls. Incubation of vascular smooth muscle cells with the demethylating agent 5-azacytidine resulted in upregulation of SOST, suggesting that SOST is epigenetically regulated. CONCLUSIONS This study identifies that SOST is expressed in the aorta and downregulated in human AA possibly because of epigenetic silencing. Upregulating SOST inhibits AA and atherosclerosis development, with potential important implications for treating these vascular diseases.
Collapse
Affiliation(s)
- Smriti Murali Krishna
- From the Vascular Biology Unit, Queensland Research Centre for Peripheral Vascular Disease, College of Medicine and Dentistry, James Cook University, Townsville, Australia (S.M.K., S.-W.S., R.J.J., J.L., S.K.M., E.B., Y.W., V.N., J.G.); National Institute of Complementary Medicine (NICM), School of Science and Health, Western Sydney University, Campbelltown, NSW, Australia (S.-W.S.); School of Applied and Biomedical Sciences, Faculty of Science and Technology, Federation University Australia (Y.W.); Department of Vascular and Transplant Surgery, Leiden University Medical Center, The Netherlands (J.H.N.L.); Physical and Life Sciences Division, Lawrence Livermore National Laboratory, CA (G.G.L.); Discipline of Biomedicine, College of Public Health, Medical and Veterinary Sciences, James Cook University, Townsville, Australia (C.M.R.); Murdoch Childrens Research Institute, Royal Children's Hospital and Department of Paediatrics, University of Melbourne, Parkville, VIC, Australia (J.M.C.); and Department of Vascular and Endovascular Surgery, The Townsville Hospital, Queensland, Australia (J.G.)
| | - Sai-Wang Seto
- From the Vascular Biology Unit, Queensland Research Centre for Peripheral Vascular Disease, College of Medicine and Dentistry, James Cook University, Townsville, Australia (S.M.K., S.-W.S., R.J.J., J.L., S.K.M., E.B., Y.W., V.N., J.G.); National Institute of Complementary Medicine (NICM), School of Science and Health, Western Sydney University, Campbelltown, NSW, Australia (S.-W.S.); School of Applied and Biomedical Sciences, Faculty of Science and Technology, Federation University Australia (Y.W.); Department of Vascular and Transplant Surgery, Leiden University Medical Center, The Netherlands (J.H.N.L.); Physical and Life Sciences Division, Lawrence Livermore National Laboratory, CA (G.G.L.); Discipline of Biomedicine, College of Public Health, Medical and Veterinary Sciences, James Cook University, Townsville, Australia (C.M.R.); Murdoch Childrens Research Institute, Royal Children's Hospital and Department of Paediatrics, University of Melbourne, Parkville, VIC, Australia (J.M.C.); and Department of Vascular and Endovascular Surgery, The Townsville Hospital, Queensland, Australia (J.G.)
| | - Roby J Jose
- From the Vascular Biology Unit, Queensland Research Centre for Peripheral Vascular Disease, College of Medicine and Dentistry, James Cook University, Townsville, Australia (S.M.K., S.-W.S., R.J.J., J.L., S.K.M., E.B., Y.W., V.N., J.G.); National Institute of Complementary Medicine (NICM), School of Science and Health, Western Sydney University, Campbelltown, NSW, Australia (S.-W.S.); School of Applied and Biomedical Sciences, Faculty of Science and Technology, Federation University Australia (Y.W.); Department of Vascular and Transplant Surgery, Leiden University Medical Center, The Netherlands (J.H.N.L.); Physical and Life Sciences Division, Lawrence Livermore National Laboratory, CA (G.G.L.); Discipline of Biomedicine, College of Public Health, Medical and Veterinary Sciences, James Cook University, Townsville, Australia (C.M.R.); Murdoch Childrens Research Institute, Royal Children's Hospital and Department of Paediatrics, University of Melbourne, Parkville, VIC, Australia (J.M.C.); and Department of Vascular and Endovascular Surgery, The Townsville Hospital, Queensland, Australia (J.G.)
| | - Jiaze Li
- From the Vascular Biology Unit, Queensland Research Centre for Peripheral Vascular Disease, College of Medicine and Dentistry, James Cook University, Townsville, Australia (S.M.K., S.-W.S., R.J.J., J.L., S.K.M., E.B., Y.W., V.N., J.G.); National Institute of Complementary Medicine (NICM), School of Science and Health, Western Sydney University, Campbelltown, NSW, Australia (S.-W.S.); School of Applied and Biomedical Sciences, Faculty of Science and Technology, Federation University Australia (Y.W.); Department of Vascular and Transplant Surgery, Leiden University Medical Center, The Netherlands (J.H.N.L.); Physical and Life Sciences Division, Lawrence Livermore National Laboratory, CA (G.G.L.); Discipline of Biomedicine, College of Public Health, Medical and Veterinary Sciences, James Cook University, Townsville, Australia (C.M.R.); Murdoch Childrens Research Institute, Royal Children's Hospital and Department of Paediatrics, University of Melbourne, Parkville, VIC, Australia (J.M.C.); and Department of Vascular and Endovascular Surgery, The Townsville Hospital, Queensland, Australia (J.G.)
| | - Susan K Morton
- From the Vascular Biology Unit, Queensland Research Centre for Peripheral Vascular Disease, College of Medicine and Dentistry, James Cook University, Townsville, Australia (S.M.K., S.-W.S., R.J.J., J.L., S.K.M., E.B., Y.W., V.N., J.G.); National Institute of Complementary Medicine (NICM), School of Science and Health, Western Sydney University, Campbelltown, NSW, Australia (S.-W.S.); School of Applied and Biomedical Sciences, Faculty of Science and Technology, Federation University Australia (Y.W.); Department of Vascular and Transplant Surgery, Leiden University Medical Center, The Netherlands (J.H.N.L.); Physical and Life Sciences Division, Lawrence Livermore National Laboratory, CA (G.G.L.); Discipline of Biomedicine, College of Public Health, Medical and Veterinary Sciences, James Cook University, Townsville, Australia (C.M.R.); Murdoch Childrens Research Institute, Royal Children's Hospital and Department of Paediatrics, University of Melbourne, Parkville, VIC, Australia (J.M.C.); and Department of Vascular and Endovascular Surgery, The Townsville Hospital, Queensland, Australia (J.G.)
| | - Erik Biros
- From the Vascular Biology Unit, Queensland Research Centre for Peripheral Vascular Disease, College of Medicine and Dentistry, James Cook University, Townsville, Australia (S.M.K., S.-W.S., R.J.J., J.L., S.K.M., E.B., Y.W., V.N., J.G.); National Institute of Complementary Medicine (NICM), School of Science and Health, Western Sydney University, Campbelltown, NSW, Australia (S.-W.S.); School of Applied and Biomedical Sciences, Faculty of Science and Technology, Federation University Australia (Y.W.); Department of Vascular and Transplant Surgery, Leiden University Medical Center, The Netherlands (J.H.N.L.); Physical and Life Sciences Division, Lawrence Livermore National Laboratory, CA (G.G.L.); Discipline of Biomedicine, College of Public Health, Medical and Veterinary Sciences, James Cook University, Townsville, Australia (C.M.R.); Murdoch Childrens Research Institute, Royal Children's Hospital and Department of Paediatrics, University of Melbourne, Parkville, VIC, Australia (J.M.C.); and Department of Vascular and Endovascular Surgery, The Townsville Hospital, Queensland, Australia (J.G.)
| | - Yutang Wang
- From the Vascular Biology Unit, Queensland Research Centre for Peripheral Vascular Disease, College of Medicine and Dentistry, James Cook University, Townsville, Australia (S.M.K., S.-W.S., R.J.J., J.L., S.K.M., E.B., Y.W., V.N., J.G.); National Institute of Complementary Medicine (NICM), School of Science and Health, Western Sydney University, Campbelltown, NSW, Australia (S.-W.S.); School of Applied and Biomedical Sciences, Faculty of Science and Technology, Federation University Australia (Y.W.); Department of Vascular and Transplant Surgery, Leiden University Medical Center, The Netherlands (J.H.N.L.); Physical and Life Sciences Division, Lawrence Livermore National Laboratory, CA (G.G.L.); Discipline of Biomedicine, College of Public Health, Medical and Veterinary Sciences, James Cook University, Townsville, Australia (C.M.R.); Murdoch Childrens Research Institute, Royal Children's Hospital and Department of Paediatrics, University of Melbourne, Parkville, VIC, Australia (J.M.C.); and Department of Vascular and Endovascular Surgery, The Townsville Hospital, Queensland, Australia (J.G.)
| | - Vianne Nsengiyumva
- From the Vascular Biology Unit, Queensland Research Centre for Peripheral Vascular Disease, College of Medicine and Dentistry, James Cook University, Townsville, Australia (S.M.K., S.-W.S., R.J.J., J.L., S.K.M., E.B., Y.W., V.N., J.G.); National Institute of Complementary Medicine (NICM), School of Science and Health, Western Sydney University, Campbelltown, NSW, Australia (S.-W.S.); School of Applied and Biomedical Sciences, Faculty of Science and Technology, Federation University Australia (Y.W.); Department of Vascular and Transplant Surgery, Leiden University Medical Center, The Netherlands (J.H.N.L.); Physical and Life Sciences Division, Lawrence Livermore National Laboratory, CA (G.G.L.); Discipline of Biomedicine, College of Public Health, Medical and Veterinary Sciences, James Cook University, Townsville, Australia (C.M.R.); Murdoch Childrens Research Institute, Royal Children's Hospital and Department of Paediatrics, University of Melbourne, Parkville, VIC, Australia (J.M.C.); and Department of Vascular and Endovascular Surgery, The Townsville Hospital, Queensland, Australia (J.G.)
| | - Jan H N Lindeman
- From the Vascular Biology Unit, Queensland Research Centre for Peripheral Vascular Disease, College of Medicine and Dentistry, James Cook University, Townsville, Australia (S.M.K., S.-W.S., R.J.J., J.L., S.K.M., E.B., Y.W., V.N., J.G.); National Institute of Complementary Medicine (NICM), School of Science and Health, Western Sydney University, Campbelltown, NSW, Australia (S.-W.S.); School of Applied and Biomedical Sciences, Faculty of Science and Technology, Federation University Australia (Y.W.); Department of Vascular and Transplant Surgery, Leiden University Medical Center, The Netherlands (J.H.N.L.); Physical and Life Sciences Division, Lawrence Livermore National Laboratory, CA (G.G.L.); Discipline of Biomedicine, College of Public Health, Medical and Veterinary Sciences, James Cook University, Townsville, Australia (C.M.R.); Murdoch Childrens Research Institute, Royal Children's Hospital and Department of Paediatrics, University of Melbourne, Parkville, VIC, Australia (J.M.C.); and Department of Vascular and Endovascular Surgery, The Townsville Hospital, Queensland, Australia (J.G.)
| | - Gabriela G Loots
- From the Vascular Biology Unit, Queensland Research Centre for Peripheral Vascular Disease, College of Medicine and Dentistry, James Cook University, Townsville, Australia (S.M.K., S.-W.S., R.J.J., J.L., S.K.M., E.B., Y.W., V.N., J.G.); National Institute of Complementary Medicine (NICM), School of Science and Health, Western Sydney University, Campbelltown, NSW, Australia (S.-W.S.); School of Applied and Biomedical Sciences, Faculty of Science and Technology, Federation University Australia (Y.W.); Department of Vascular and Transplant Surgery, Leiden University Medical Center, The Netherlands (J.H.N.L.); Physical and Life Sciences Division, Lawrence Livermore National Laboratory, CA (G.G.L.); Discipline of Biomedicine, College of Public Health, Medical and Veterinary Sciences, James Cook University, Townsville, Australia (C.M.R.); Murdoch Childrens Research Institute, Royal Children's Hospital and Department of Paediatrics, University of Melbourne, Parkville, VIC, Australia (J.M.C.); and Department of Vascular and Endovascular Surgery, The Townsville Hospital, Queensland, Australia (J.G.)
| | - Catherine M Rush
- From the Vascular Biology Unit, Queensland Research Centre for Peripheral Vascular Disease, College of Medicine and Dentistry, James Cook University, Townsville, Australia (S.M.K., S.-W.S., R.J.J., J.L., S.K.M., E.B., Y.W., V.N., J.G.); National Institute of Complementary Medicine (NICM), School of Science and Health, Western Sydney University, Campbelltown, NSW, Australia (S.-W.S.); School of Applied and Biomedical Sciences, Faculty of Science and Technology, Federation University Australia (Y.W.); Department of Vascular and Transplant Surgery, Leiden University Medical Center, The Netherlands (J.H.N.L.); Physical and Life Sciences Division, Lawrence Livermore National Laboratory, CA (G.G.L.); Discipline of Biomedicine, College of Public Health, Medical and Veterinary Sciences, James Cook University, Townsville, Australia (C.M.R.); Murdoch Childrens Research Institute, Royal Children's Hospital and Department of Paediatrics, University of Melbourne, Parkville, VIC, Australia (J.M.C.); and Department of Vascular and Endovascular Surgery, The Townsville Hospital, Queensland, Australia (J.G.)
| | - Jeffrey M Craig
- From the Vascular Biology Unit, Queensland Research Centre for Peripheral Vascular Disease, College of Medicine and Dentistry, James Cook University, Townsville, Australia (S.M.K., S.-W.S., R.J.J., J.L., S.K.M., E.B., Y.W., V.N., J.G.); National Institute of Complementary Medicine (NICM), School of Science and Health, Western Sydney University, Campbelltown, NSW, Australia (S.-W.S.); School of Applied and Biomedical Sciences, Faculty of Science and Technology, Federation University Australia (Y.W.); Department of Vascular and Transplant Surgery, Leiden University Medical Center, The Netherlands (J.H.N.L.); Physical and Life Sciences Division, Lawrence Livermore National Laboratory, CA (G.G.L.); Discipline of Biomedicine, College of Public Health, Medical and Veterinary Sciences, James Cook University, Townsville, Australia (C.M.R.); Murdoch Childrens Research Institute, Royal Children's Hospital and Department of Paediatrics, University of Melbourne, Parkville, VIC, Australia (J.M.C.); and Department of Vascular and Endovascular Surgery, The Townsville Hospital, Queensland, Australia (J.G.)
| | - Jonathan Golledge
- From the Vascular Biology Unit, Queensland Research Centre for Peripheral Vascular Disease, College of Medicine and Dentistry, James Cook University, Townsville, Australia (S.M.K., S.-W.S., R.J.J., J.L., S.K.M., E.B., Y.W., V.N., J.G.); National Institute of Complementary Medicine (NICM), School of Science and Health, Western Sydney University, Campbelltown, NSW, Australia (S.-W.S.); School of Applied and Biomedical Sciences, Faculty of Science and Technology, Federation University Australia (Y.W.); Department of Vascular and Transplant Surgery, Leiden University Medical Center, The Netherlands (J.H.N.L.); Physical and Life Sciences Division, Lawrence Livermore National Laboratory, CA (G.G.L.); Discipline of Biomedicine, College of Public Health, Medical and Veterinary Sciences, James Cook University, Townsville, Australia (C.M.R.); Murdoch Childrens Research Institute, Royal Children's Hospital and Department of Paediatrics, University of Melbourne, Parkville, VIC, Australia (J.M.C.); and Department of Vascular and Endovascular Surgery, The Townsville Hospital, Queensland, Australia (J.G.).
| |
Collapse
|
43
|
Meffert P, Tscheuschler A, Beyersdorf F, Heilmann C, Kocher N, Uffelmann X, Discher P, Rylski B, Siepe M, Kari FA. Characterization of serum matrix metalloproteinase 2/9 levels in patients with ascending aortic aneurysms. Interact Cardiovasc Thorac Surg 2016; 24:20-26. [DOI: 10.1093/icvts/ivw309] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2016] [Revised: 06/29/2016] [Accepted: 07/11/2016] [Indexed: 01/05/2023] Open
|
44
|
Abstract
PURPOSE OF REVIEW Abdominal aortic aneurysm (AAA) is a pathological condition of permanent dilation that portends the potentially fatal consequence of aortic rupture. This review emphasizes recent advances in mechanistic insight into aneurysm pathogenesis and potential pharmacologic therapies that are on the horizon for AAAs. RECENT FINDINGS An increasing body of evidence demonstrates that genetic factors, including 3p12.3, DAB2IP, LDLr, LRP1, matrix metalloproteinase (MMP)-3, TGFBR2, and SORT1 loci, are associated with AAA development. Current human studies and animal models have shown that many leukocytes and inflammatory mediators, such as IL-1, IL-17, TGF-β, and angiotensin II, are involved in the pathogenesis of AAAs. Leukocytic infiltration into aortic media leads to smooth muscle cell depletion, generation of reactive oxygen species, and extracellular matrix fragmentation. Preclinical investigations into pharmacological therapies for AAAs have provided intriguing insight into the roles of microRNAs in regulating many pathological pathways in AAA development. Several large clinical trials are ongoing, seeking to translate preclinical findings into therapeutic options. SUMMARY Recent studies have identified many potential mechanisms involved in AAA pathogenesis that provide insight into the development of a medical treatment for this disease.
Collapse
|
45
|
Zhao Y, Li D, Zhao J, Song J, Zhao Y. The role of the low-density lipoprotein receptor–related protein 1 (LRP-1) in regulating blood-brain barrier integrity. Rev Neurosci 2016; 27:623-34. [PMID: 27206317 DOI: 10.1515/revneuro-2015-0069] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2015] [Accepted: 03/25/2016] [Indexed: 12/18/2022]
Abstract
AbstractThe blood-brain barrier (BBB) is a protective structure that helps maintaining the homeostasis in cerebral microenvironment by limiting the passage of molecules into the brain. BBB is formed by closely conjugated endothelial cells, with astrocytic endfeet surrounded and extracellular matrix (ECM) consolidated. Numerous neurological diseases can cause disturbance of BBB, leading to brain edema and neurological dysfunctions. The low-density lipoprotein (LDL) receptor–related protein 1 (LRP-1), a member of the LDL receptor gene family, is involved in a lot of important processes in the brain under both physiological and pathological conditions. As a membrane receptor, LRP-1 interacts with a variety of ligands and mediates the internalization of several important substances. LRP-1 is found responsible for inducing the opening of BBB following ischemic attack. It has also been reported that LRP-1 regulates several tight junction proteins and mediates the clearance of major ECM-degrading proteinases. In this review, we briefly discussed the role of LRP-1 in regulating BBB integrity by modulating tight junction proteins, endothelial cells and the remodeling of ECM.
Collapse
Affiliation(s)
- Yahui Zhao
- 1Department of Neurosurgery, the First Affiliated Hospital of Xi’, and Jiaotong University College of Medicine, Xi’an 710061, Shaanxi Province, China
| | - Dandong Li
- 2Department of Neurosurgery, the Second Affiliated Hospital of Wenzhou Medical University, Wenzhou 325027, Zhejiang Province, China
| | - Junjie Zhao
- 1Department of Neurosurgery, the First Affiliated Hospital of Xi’, and Jiaotong University College of Medicine, Xi’an 710061, Shaanxi Province, China
| | - Jinning Song
- 1Department of Neurosurgery, the First Affiliated Hospital of Xi’, and Jiaotong University College of Medicine, Xi’an 710061, Shaanxi Province, China
| | - Yonglin Zhao
- 1Department of Neurosurgery, the First Affiliated Hospital of Xi’, and Jiaotong University College of Medicine, Xi’an 710061, Shaanxi Province, China
| |
Collapse
|
46
|
Tai HC, Tsai PJ, Chen JY, Lai CH, Wang KC, Teng SH, Lin SC, Chang AYW, Jiang MJ, Li YH, Wu HL, Maeda N, Tsai YS. Peroxisome Proliferator-Activated Receptor γ Level Contributes to Structural Integrity and Component Production of Elastic Fibers in the Aorta. Hypertension 2016; 67:1298-308. [PMID: 27045031 DOI: 10.1161/hypertensionaha.116.07367] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2016] [Accepted: 03/09/2016] [Indexed: 01/24/2023]
Abstract
Loss of integrity and massive disruption of elastic fibers are key features of abdominal aortic aneurysm (AAA). Peroxisome proliferator-activated receptor γ (PPARγ) has been shown to attenuate AAA through inhibition of inflammation and proteolytic degradation. However, its involvement in elastogenesis during AAA remains unclear. PPARγ was highly expressed in human AAA within all vascular cells, including inflammatory cells and fibroblasts. In the aortas of transgenic mice expressing PPARγ at 25% normal levels (Pparg(C) (/-) mice), we observed the fragmentation of elastic fibers and reduced expression of vital elastic fiber components of elastin and fibulin-5. These were not observed in mice with 50% normal PPARγ expression (Pparg(+/-) mice). Infusion of a moderate dose of angiotensin II (500 ng/kg per minute) did not induce AAA but Pparg(+/-) aorta developed flattened elastic lamellae, whereas Pparg(C/-) aorta showed severe destruction of elastic fibers. After infusion of angiotensin II at 1000 ng/kg per minute, 73% of Pparg(C/-) mice developed atypical suprarenal aortic aneurysms: superior mesenteric arteries were dilated with extensive collagen deposition in adventitia and infiltrations of inflammatory cells. Although matrix metalloproteinase inhibition by doxycycline somewhat attenuated the dilation of aneurysm, it did not reduce the incidence nor elastic lamella deterioration in angiotensin II-infused Pparg(C/-) mice. Furthermore, PPARγ antagonism downregulated elastin and fibulin-5 in fibroblasts, but not in vascular smooth muscle cells. Chromatin immunoprecipitation assay demonstrated PPARγ binding in the genomic sequence of fibulin-5 in fibroblasts. Our results underscore the importance of PPARγ in AAA development though orchestrating proper elastogenesis and preserving elastic fiber integrity.
Collapse
Affiliation(s)
- Haw-Chih Tai
- From the Institute of Clinical Medicine (H.-C.T., J.-Y.C., C.-H.L., Y.-S.T.), Cardiovascular Research Center (H.-C.T., J.-Y.C., C.-H.L., K.-C.W., M.-J.J., Y.-H.L., H.-L.W., Y.-S.T.), Departments of Medical Laboratory Science and Biotechnology (P.-J.T.), Biochemistry and Molecular Biology (K.-C.W., H.-L.W.), Physiology (S.-C.L., A.Y.W.C.), Cell Biology and Anatomy (M.-J.J.), National Cheng Kung University, Tainan, Taiwan, Republic of China; Departments of Internal Medicine (J.-Y.C., Y.-H.L.), Surgery (C.-H.L.), and Research Center of Clinical Medicine (Y.-S.T.), National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan, Republic of China; Graduate Institute of Biomedical Sciences, Chang Gung University, Taoyuan, Taiwan, Republic of China (S.-H.T.); and Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill (N.M.)
| | - Pei-Jane Tsai
- From the Institute of Clinical Medicine (H.-C.T., J.-Y.C., C.-H.L., Y.-S.T.), Cardiovascular Research Center (H.-C.T., J.-Y.C., C.-H.L., K.-C.W., M.-J.J., Y.-H.L., H.-L.W., Y.-S.T.), Departments of Medical Laboratory Science and Biotechnology (P.-J.T.), Biochemistry and Molecular Biology (K.-C.W., H.-L.W.), Physiology (S.-C.L., A.Y.W.C.), Cell Biology and Anatomy (M.-J.J.), National Cheng Kung University, Tainan, Taiwan, Republic of China; Departments of Internal Medicine (J.-Y.C., Y.-H.L.), Surgery (C.-H.L.), and Research Center of Clinical Medicine (Y.-S.T.), National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan, Republic of China; Graduate Institute of Biomedical Sciences, Chang Gung University, Taoyuan, Taiwan, Republic of China (S.-H.T.); and Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill (N.M.)
| | - Ju-Yi Chen
- From the Institute of Clinical Medicine (H.-C.T., J.-Y.C., C.-H.L., Y.-S.T.), Cardiovascular Research Center (H.-C.T., J.-Y.C., C.-H.L., K.-C.W., M.-J.J., Y.-H.L., H.-L.W., Y.-S.T.), Departments of Medical Laboratory Science and Biotechnology (P.-J.T.), Biochemistry and Molecular Biology (K.-C.W., H.-L.W.), Physiology (S.-C.L., A.Y.W.C.), Cell Biology and Anatomy (M.-J.J.), National Cheng Kung University, Tainan, Taiwan, Republic of China; Departments of Internal Medicine (J.-Y.C., Y.-H.L.), Surgery (C.-H.L.), and Research Center of Clinical Medicine (Y.-S.T.), National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan, Republic of China; Graduate Institute of Biomedical Sciences, Chang Gung University, Taoyuan, Taiwan, Republic of China (S.-H.T.); and Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill (N.M.)
| | - Chao-Han Lai
- From the Institute of Clinical Medicine (H.-C.T., J.-Y.C., C.-H.L., Y.-S.T.), Cardiovascular Research Center (H.-C.T., J.-Y.C., C.-H.L., K.-C.W., M.-J.J., Y.-H.L., H.-L.W., Y.-S.T.), Departments of Medical Laboratory Science and Biotechnology (P.-J.T.), Biochemistry and Molecular Biology (K.-C.W., H.-L.W.), Physiology (S.-C.L., A.Y.W.C.), Cell Biology and Anatomy (M.-J.J.), National Cheng Kung University, Tainan, Taiwan, Republic of China; Departments of Internal Medicine (J.-Y.C., Y.-H.L.), Surgery (C.-H.L.), and Research Center of Clinical Medicine (Y.-S.T.), National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan, Republic of China; Graduate Institute of Biomedical Sciences, Chang Gung University, Taoyuan, Taiwan, Republic of China (S.-H.T.); and Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill (N.M.)
| | - Kuan-Chieh Wang
- From the Institute of Clinical Medicine (H.-C.T., J.-Y.C., C.-H.L., Y.-S.T.), Cardiovascular Research Center (H.-C.T., J.-Y.C., C.-H.L., K.-C.W., M.-J.J., Y.-H.L., H.-L.W., Y.-S.T.), Departments of Medical Laboratory Science and Biotechnology (P.-J.T.), Biochemistry and Molecular Biology (K.-C.W., H.-L.W.), Physiology (S.-C.L., A.Y.W.C.), Cell Biology and Anatomy (M.-J.J.), National Cheng Kung University, Tainan, Taiwan, Republic of China; Departments of Internal Medicine (J.-Y.C., Y.-H.L.), Surgery (C.-H.L.), and Research Center of Clinical Medicine (Y.-S.T.), National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan, Republic of China; Graduate Institute of Biomedical Sciences, Chang Gung University, Taoyuan, Taiwan, Republic of China (S.-H.T.); and Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill (N.M.)
| | - Shih-Hua Teng
- From the Institute of Clinical Medicine (H.-C.T., J.-Y.C., C.-H.L., Y.-S.T.), Cardiovascular Research Center (H.-C.T., J.-Y.C., C.-H.L., K.-C.W., M.-J.J., Y.-H.L., H.-L.W., Y.-S.T.), Departments of Medical Laboratory Science and Biotechnology (P.-J.T.), Biochemistry and Molecular Biology (K.-C.W., H.-L.W.), Physiology (S.-C.L., A.Y.W.C.), Cell Biology and Anatomy (M.-J.J.), National Cheng Kung University, Tainan, Taiwan, Republic of China; Departments of Internal Medicine (J.-Y.C., Y.-H.L.), Surgery (C.-H.L.), and Research Center of Clinical Medicine (Y.-S.T.), National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan, Republic of China; Graduate Institute of Biomedical Sciences, Chang Gung University, Taoyuan, Taiwan, Republic of China (S.-H.T.); and Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill (N.M.)
| | - Shih-Chieh Lin
- From the Institute of Clinical Medicine (H.-C.T., J.-Y.C., C.-H.L., Y.-S.T.), Cardiovascular Research Center (H.-C.T., J.-Y.C., C.-H.L., K.-C.W., M.-J.J., Y.-H.L., H.-L.W., Y.-S.T.), Departments of Medical Laboratory Science and Biotechnology (P.-J.T.), Biochemistry and Molecular Biology (K.-C.W., H.-L.W.), Physiology (S.-C.L., A.Y.W.C.), Cell Biology and Anatomy (M.-J.J.), National Cheng Kung University, Tainan, Taiwan, Republic of China; Departments of Internal Medicine (J.-Y.C., Y.-H.L.), Surgery (C.-H.L.), and Research Center of Clinical Medicine (Y.-S.T.), National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan, Republic of China; Graduate Institute of Biomedical Sciences, Chang Gung University, Taoyuan, Taiwan, Republic of China (S.-H.T.); and Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill (N.M.)
| | - Alice Y W Chang
- From the Institute of Clinical Medicine (H.-C.T., J.-Y.C., C.-H.L., Y.-S.T.), Cardiovascular Research Center (H.-C.T., J.-Y.C., C.-H.L., K.-C.W., M.-J.J., Y.-H.L., H.-L.W., Y.-S.T.), Departments of Medical Laboratory Science and Biotechnology (P.-J.T.), Biochemistry and Molecular Biology (K.-C.W., H.-L.W.), Physiology (S.-C.L., A.Y.W.C.), Cell Biology and Anatomy (M.-J.J.), National Cheng Kung University, Tainan, Taiwan, Republic of China; Departments of Internal Medicine (J.-Y.C., Y.-H.L.), Surgery (C.-H.L.), and Research Center of Clinical Medicine (Y.-S.T.), National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan, Republic of China; Graduate Institute of Biomedical Sciences, Chang Gung University, Taoyuan, Taiwan, Republic of China (S.-H.T.); and Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill (N.M.)
| | - Meei-Jyh Jiang
- From the Institute of Clinical Medicine (H.-C.T., J.-Y.C., C.-H.L., Y.-S.T.), Cardiovascular Research Center (H.-C.T., J.-Y.C., C.-H.L., K.-C.W., M.-J.J., Y.-H.L., H.-L.W., Y.-S.T.), Departments of Medical Laboratory Science and Biotechnology (P.-J.T.), Biochemistry and Molecular Biology (K.-C.W., H.-L.W.), Physiology (S.-C.L., A.Y.W.C.), Cell Biology and Anatomy (M.-J.J.), National Cheng Kung University, Tainan, Taiwan, Republic of China; Departments of Internal Medicine (J.-Y.C., Y.-H.L.), Surgery (C.-H.L.), and Research Center of Clinical Medicine (Y.-S.T.), National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan, Republic of China; Graduate Institute of Biomedical Sciences, Chang Gung University, Taoyuan, Taiwan, Republic of China (S.-H.T.); and Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill (N.M.)
| | - Yi-Heng Li
- From the Institute of Clinical Medicine (H.-C.T., J.-Y.C., C.-H.L., Y.-S.T.), Cardiovascular Research Center (H.-C.T., J.-Y.C., C.-H.L., K.-C.W., M.-J.J., Y.-H.L., H.-L.W., Y.-S.T.), Departments of Medical Laboratory Science and Biotechnology (P.-J.T.), Biochemistry and Molecular Biology (K.-C.W., H.-L.W.), Physiology (S.-C.L., A.Y.W.C.), Cell Biology and Anatomy (M.-J.J.), National Cheng Kung University, Tainan, Taiwan, Republic of China; Departments of Internal Medicine (J.-Y.C., Y.-H.L.), Surgery (C.-H.L.), and Research Center of Clinical Medicine (Y.-S.T.), National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan, Republic of China; Graduate Institute of Biomedical Sciences, Chang Gung University, Taoyuan, Taiwan, Republic of China (S.-H.T.); and Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill (N.M.)
| | - Hua-Lin Wu
- From the Institute of Clinical Medicine (H.-C.T., J.-Y.C., C.-H.L., Y.-S.T.), Cardiovascular Research Center (H.-C.T., J.-Y.C., C.-H.L., K.-C.W., M.-J.J., Y.-H.L., H.-L.W., Y.-S.T.), Departments of Medical Laboratory Science and Biotechnology (P.-J.T.), Biochemistry and Molecular Biology (K.-C.W., H.-L.W.), Physiology (S.-C.L., A.Y.W.C.), Cell Biology and Anatomy (M.-J.J.), National Cheng Kung University, Tainan, Taiwan, Republic of China; Departments of Internal Medicine (J.-Y.C., Y.-H.L.), Surgery (C.-H.L.), and Research Center of Clinical Medicine (Y.-S.T.), National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan, Republic of China; Graduate Institute of Biomedical Sciences, Chang Gung University, Taoyuan, Taiwan, Republic of China (S.-H.T.); and Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill (N.M.)
| | - Nobuyo Maeda
- From the Institute of Clinical Medicine (H.-C.T., J.-Y.C., C.-H.L., Y.-S.T.), Cardiovascular Research Center (H.-C.T., J.-Y.C., C.-H.L., K.-C.W., M.-J.J., Y.-H.L., H.-L.W., Y.-S.T.), Departments of Medical Laboratory Science and Biotechnology (P.-J.T.), Biochemistry and Molecular Biology (K.-C.W., H.-L.W.), Physiology (S.-C.L., A.Y.W.C.), Cell Biology and Anatomy (M.-J.J.), National Cheng Kung University, Tainan, Taiwan, Republic of China; Departments of Internal Medicine (J.-Y.C., Y.-H.L.), Surgery (C.-H.L.), and Research Center of Clinical Medicine (Y.-S.T.), National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan, Republic of China; Graduate Institute of Biomedical Sciences, Chang Gung University, Taoyuan, Taiwan, Republic of China (S.-H.T.); and Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill (N.M.)
| | - Yau-Sheng Tsai
- From the Institute of Clinical Medicine (H.-C.T., J.-Y.C., C.-H.L., Y.-S.T.), Cardiovascular Research Center (H.-C.T., J.-Y.C., C.-H.L., K.-C.W., M.-J.J., Y.-H.L., H.-L.W., Y.-S.T.), Departments of Medical Laboratory Science and Biotechnology (P.-J.T.), Biochemistry and Molecular Biology (K.-C.W., H.-L.W.), Physiology (S.-C.L., A.Y.W.C.), Cell Biology and Anatomy (M.-J.J.), National Cheng Kung University, Tainan, Taiwan, Republic of China; Departments of Internal Medicine (J.-Y.C., Y.-H.L.), Surgery (C.-H.L.), and Research Center of Clinical Medicine (Y.-S.T.), National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan, Republic of China; Graduate Institute of Biomedical Sciences, Chang Gung University, Taoyuan, Taiwan, Republic of China (S.-H.T.); and Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill (N.M.).
| |
Collapse
|
47
|
Kuivaniemi H, Ryer EJ, Elmore JR, Tromp G. Understanding the pathogenesis of abdominal aortic aneurysms. Expert Rev Cardiovasc Ther 2016; 13:975-87. [PMID: 26308600 DOI: 10.1586/14779072.2015.1074861] [Citation(s) in RCA: 256] [Impact Index Per Article: 28.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
An aortic aneurysm is a dilatation in which the aortic diameter is ≥3.0 cm. If left untreated, the aortic wall continues to weaken and becomes unable to withstand the forces of the luminal blood pressure resulting in progressive dilatation and rupture, a catastrophic event associated with a mortality of 50-80%. Smoking and positive family history are important risk factors for the development of abdominal aortic aneurysms (AAA). Several genetic risk factors have also been identified. On the histological level, visible hallmarks of AAA pathogenesis include inflammation, smooth muscle cell apoptosis, extracellular matrix degradation and oxidative stress. We expect that large genetic, genomic, epigenetic, proteomic and metabolomic studies will be undertaken by international consortia to identify additional risk factors and biomarkers, and to enhance our understanding of the pathobiology of AAA. Collaboration between different research groups will be important in overcoming the challenges to develop pharmacological treatments for AAA.
Collapse
Affiliation(s)
- Helena Kuivaniemi
- a 1 Sigfried and Janet Weis Center for Research, Geisinger Health System, Danville, PA 17822, USA
| | | | | | | |
Collapse
|
48
|
Lu H, Howatt DA, Balakrishnan A, Moorleghen JJ, Rateri DL, Cassis LA, Daugherty A. Subcutaneous Angiotensin II Infusion using Osmotic Pumps Induces Aortic Aneurysms in Mice. J Vis Exp 2015. [PMID: 26436287 PMCID: PMC4692630 DOI: 10.3791/53191] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Osmotic pumps continuously deliver compounds at a constant rate into small animals. This article introduces a standard protocol used to induce aortic aneurysms via subcutaneous infusion of angiotensin II (AngII) from implanted osmotic pumps. This protocol includes calculation of AngII amount and dissolution, osmotic pump filling, implantation of osmotic pumps subcutaneously, observation after pump implantation, and harvest of aortas to visualize aortic aneurysms in mice. Subcutaneous infusion of AngII through osmotic pumps following this protocol is a reliable and reproducible technique to induce both abdominal and thoracic aortic aneurysms in mice. Infusion durations range from a few days to several months based on the purpose of the study. AngII 1,000 ng/kg/min is sufficient to provide maximal effects on abdominal aortic aneurysmal formation in male hypercholesterolemic mouse models such as apolipoprotein E deficient or low-density lipoprotein receptor deficient mice. Incidence of abdominal aortic aneurysms induced by AngII infusion via osmotic pumps is 5-10 times lower in female hypercholesterolemic mice and also lower in both genders of normocholesterolemic mice. In contrast, AngII-induced thoracic aortic aneurysms in mice are not hypercholesterolemia or gender-dependent. Importantly, multiple features of this mouse model recapitulate those of human aortic aneurysms.
Collapse
Affiliation(s)
| | | | | | | | - Debra L Rateri
- Saha Cardiovascular Research Center, University of Kentucky
| | - Lisa A Cassis
- Department of Pharmacology and Nutritional Sciences, University of Kentucky
| | - Alan Daugherty
- Saha Cardiovascular Research Center, University of Kentucky;
| |
Collapse
|
49
|
Moxon JV, Behl-Gilhotra R, Morton SK, Krishna SM, Seto SW, Biros E, Nataatmadja M, West M, Walker PJ, Norman PE, Golledge J. Plasma Low-density Lipoprotein Receptor-related Protein 1 Concentration is not Associated with Human Abdominal Aortic Aneurysm Presence. Eur J Vasc Endovasc Surg 2015; 50:466-73. [PMID: 26188720 DOI: 10.1016/j.ejvs.2015.06.023] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2015] [Accepted: 06/06/2015] [Indexed: 12/29/2022]
Abstract
OBJECTIVE/BACKGROUND Recent genetic data suggest that a polymorphism of LRP1 is an independent risk factor for abdominal aortic aneurysm (AAA). The aims of this study were to assess whether plasma and aortic concentrations of low-density lipoprotein receptor-related protein 1 (LRP1) are associated with AAA, and to investigate the possible relevance of LRP1 to AAA pathophysiology. METHODS Three analyses were conducted. First, plasma LRP1 concentrations were measured in community-dwelling men with and without AAA (n = 189 and n = 309, respectively) using enzyme-linked immunosorbent assay. Second, Western blotting analyses were employed to compare the expression of LRP1 protein in aortic biopsies collected from patients with AAA and nonaneurysmal postmortem donors (n = 6/group). Finally, the effect of in vitro LRP1 blockade on matrix metalloprotease 9 (MMP9) clearance by vascular smooth muscle cells was assessed by zymography. RESULTS Plasma LRP1 concentrations did not differ between groups of men with and without AAA (median concentration 4.56 μg/mL [interquartile range {IQR} (3.39-5.96)] and 4.43 μg/mL [IQR 3.44-5.84], respectively; p = .48), and were not associated with AAA after adjusting for other risk factors (odds ratio 1.10 [95% confidence interval: 0.91-1.32]; p = 0.35). In contrast, LRP1 expression was approximately 3.4-fold lower in aortic biopsies recovered from patients with AAA compared with controls (median [IQR] expression 1.72 [0.94-3.14] and 5.91 [4.63-6.94] relative density units, respectively; p < .01). In vitro LRP1 blockade significantly reduced the ability of vascular smooth muscle cells to internalize extracellular MMP9. CONCLUSIONS These data suggest that aortic but not circulating LRP1 is downregulated in patients with AAA and indicates a possible role for this protein in clearing an aneurysm-relevant ligand.
Collapse
Affiliation(s)
- J V Moxon
- Queensland Research Centre for Peripheral Vascular Disease, College of Medicine and Dentistry, James Cook University, Townsville, QLD 4811, Australia
| | - R Behl-Gilhotra
- Queensland Research Centre for Peripheral Vascular Disease, College of Medicine and Dentistry, James Cook University, Townsville, QLD 4811, Australia
| | - S K Morton
- Queensland Research Centre for Peripheral Vascular Disease, College of Medicine and Dentistry, James Cook University, Townsville, QLD 4811, Australia
| | - S M Krishna
- Queensland Research Centre for Peripheral Vascular Disease, College of Medicine and Dentistry, James Cook University, Townsville, QLD 4811, Australia
| | - S W Seto
- Queensland Research Centre for Peripheral Vascular Disease, College of Medicine and Dentistry, James Cook University, Townsville, QLD 4811, Australia; National Institute of Complementary Medicine, University of Western Sydney, Campbelltown, NSW 2560, Australia
| | - E Biros
- Queensland Research Centre for Peripheral Vascular Disease, College of Medicine and Dentistry, James Cook University, Townsville, QLD 4811, Australia
| | - M Nataatmadja
- The Cardiovascular Research Group, Department of Medicine, University of Queensland, The Prince Charles Hospital, Brisbane, QLD 4032, Australia
| | - M West
- The Cardiovascular Research Group, Department of Medicine, University of Queensland, The Prince Charles Hospital, Brisbane, QLD 4032, Australia
| | - P J Walker
- School of Medicine, Discipline of Surgery and Centre for Clinical Research, University of Queensland, Herston, QLD 4072, Australia
| | - P E Norman
- School of Surgery, University of Western Australia, Perth, WA 6009, Australia
| | - J Golledge
- Queensland Research Centre for Peripheral Vascular Disease, College of Medicine and Dentistry, James Cook University, Townsville, QLD 4811, Australia; School of Medicine, Discipline of Surgery and Centre for Clinical Research, University of Queensland, Herston, QLD 4072, Australia; Department of Vascular and Endovascular Surgery, The Townsville Hospital, Townsville, QLD 4814, Australia.
| |
Collapse
|
50
|
Haggerty CM, Mattingly AC, Gong MC, Su W, Daugherty A, Fornwalt BK. Telemetric Blood Pressure Assessment in Angiotensin II-Infused ApoE-/- Mice: 28 Day Natural History and Comparison to Tail-Cuff Measurements. PLoS One 2015; 10:e0130723. [PMID: 26086817 PMCID: PMC4472759 DOI: 10.1371/journal.pone.0130723] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2015] [Accepted: 05/22/2015] [Indexed: 12/03/2022] Open
Abstract
Abdominal aortic aneurysm (AAA) is a disease of the aortic wall, which can progress to catastrophic rupture. Assessment of mechanical characteristics of AAA, such as aortic distensibility, may provide important insights to help identify at-risk patients and understand disease progression. While the majority of studies on this topic have focused on retrospective patient data, recent studies have used mouse models of AAA to prospectively evaluate the evolution of aortic mechanics. Quantification of aortic distensibility requires accurate measurement of arterial blood pressure, particularly pulse pressure, which is challenging to perform accurately in murine models. We hypothesized that volume/pressure tail-cuff measurements of arterial pulse pressure in anesthetized mice would have sufficient accuracy to enable calculations of aortic distensibility with minimal error. Telemetry devices and osmotic mini-pumps filled with saline or angiotensin-II were surgically implanted in male apolipoprotein-E deficient (ApoE-/-) mice. Blood pressure in the aortic arch was measured continuously via telemetry. In addition, simultaneous blood pressure measurements with a volume/pressure tail-cuff system were performed under anesthesia at specific intervals to assess agreement between techniques. Compared to controls, mice infused with angiotensin-II had an overall statistically significant increase in systolic pressure, with no overall difference in pulse pressure; however, pulse pressure did increase significantly with time. Systolic measurements agreed well between telemetry and tail-cuff (coefficient of variation = 10%), but agreement of pulse pressure was weak (20%). In fact, group-averaged pulse pressure from telemetry was a better predictor of a subject’s pulse pressure on a given day than a simultaneous tail-cuff measurement. Furthermore, these approximations introduced acceptable errors (15.1 ± 12.8%) into the calculation of aortic distensibility. Contrary to our hypothesis, we conclude that tail-cuff measures of arterial pulse pressure have limited accuracy. Future studies of aneurysm mechanics using the ApoE-/-/angiotensin-II model would be better in assuming pulse pressure profiles consistent with our telemetry findings instead of attempting to measure pulse pressure in individual mice.
Collapse
Affiliation(s)
- Christopher M. Haggerty
- Saha Cardiovascular Research Center, University of Kentucky, Lexington, Kentucky, United States of America
| | - Andrea C. Mattingly
- Saha Cardiovascular Research Center, University of Kentucky, Lexington, Kentucky, United States of America
| | - Ming C. Gong
- Department of Physiology, University of Kentucky, Lexington, Kentucky, United States of America
| | - Wen Su
- Saha Cardiovascular Research Center, University of Kentucky, Lexington, Kentucky, United States of America
| | - Alan Daugherty
- Saha Cardiovascular Research Center, University of Kentucky, Lexington, Kentucky, United States of America
| | - Brandon K. Fornwalt
- Saha Cardiovascular Research Center, University of Kentucky, Lexington, Kentucky, United States of America
- Department of Physiology, University of Kentucky, Lexington, Kentucky, United States of America
- Department of Biomedical Engineering, University of Kentucky, Lexington, Kentucky, United States of America
- Department of Pediatrics, University of Kentucky, Lexington, Kentucky, United States of America
- * E-mail:
| |
Collapse
|