1
|
Curry-Koski T, Curtin L, Esfandiarei M, Currier TT. Cerebral Microvascular Density, Permeability of the Blood-Brain Barrier, and Neuroinflammatory Responses Indicate Early Aging Characteristics in a Marfan Syndrome Mouse Model. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.30.601409. [PMID: 39005441 PMCID: PMC11244932 DOI: 10.1101/2024.06.30.601409] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/16/2024]
Abstract
Marfan Syndrome (MFS) is a connective tissue disorder due to mutations in fibrillin-1 ( Fbn1 ), where a Fbn1 missense mutation ( Fbn1 C1039G/+ ) can result in systemic increases in the bioavailability and signaling of transforming growth factor-β (TGF-β). In a well-established mouse model of MFS ( Fbn1 C1041G/+ ), pre-mature aging of the aortic wall and the progression of aortic root aneurysm are observed by 6-months-of-age. TGF-β signaling has been implicated in cerebrovascular dysfunction, loss of blood-brain barrier (BBB) integrity, and age-related neuroinflammation. We have reported that pre-mature vascular aging in MFS mice could extend to cerebrovasculature, where peak blood flow velocity in the posterior cerebral artery (PCA) of 6-month-old (6M) MFS mice was reduced, similarly to 12-month-old (12M) control mice. Case studies of MFS patients have documented neurovascular manifestations, including intracranial aneurysms, stroke, arterial tortuosity, as well as headaches and migraines, with reported incidence of pain and chronic fatigue. Despite these significant clinical observations, investigation into cerebrovascular dysfunction and neuropathology in MFS remains limited. Using 6M-control ( C57BL/6 ) and 6M-MFS ( Fbn1 C1041G/+ ) and healthy 12M-control male and female mice, we test the hypothesis that abnormal Fbn1 protein expression is associated with altered cerebral microvascular density, BBB permeability, and neuroinflammation in the PCA-perfused hippocampus, all indicative of a pre-mature aging brain phenotype. Using Glut1 staining, 6M-MFS mice and 12M-CTRL similarly present decreased microvascular density in the dentate gyrus (DG), cornu ammonis 1 (CA1), and cornu ammonis 3 (CA3) regions of the hippocampus. 6M-MFS mice exhibit increased BBB permeability in the DG, CA1, and CA3 as evident by Immunoglobulin G (IgG) staining, which was more comparable to 12M-CTRL mice. 6M-MFS mice show a higher number of microglia in the hippocampus compared to age-matched control mice, a pattern resembling that of 12M-CTRL mice. This study represents the first known investigation into neuropathology in a mouse model of MFS and indicates that the pathophysiology underlying MFS leads to a systemic pre-mature aging phenotype. This study is crucial for identifying and understanding MFS-associated neurovascular and neurological abnormalities, underscoring the need for research aimed at improving the quality of life and managing pre-mature aging symptoms in MFS and related connective tissue disorders.
Collapse
|
2
|
Curry T, Barrameda ME, Thomas TC, Esfandiarei M. In vivo phenotypic vascular dysfunction extends beyond the aorta in a mouse model for fibrillin-1 (Fbn1) mutation. Sci Rep 2024; 14:5779. [PMID: 38461168 PMCID: PMC10924961 DOI: 10.1038/s41598-024-56438-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2023] [Accepted: 03/06/2024] [Indexed: 03/11/2024] Open
Abstract
In individuals with Marfan Syndrome (MFS), fibrillin-1 gene (FBN1) mutations can lead to vascular wall weakening and dysfunction. The experimental mouse model of MFS (Fbn1C1041G/+) has been advantageous in investigating MFS-associated life-threatening aortic aneurysms. It is well established that the MFS mouse model exhibits an accelerated-aging phenotype in elastic organs like the aorta, lung, and skin. However, the impact of Fbn1 mutations on the in vivo function and structure of various artery types with the consideration of sex and age, has not been adequately explored in real-time and a clinically relevant context. In this study, we investigate if Fbn1 mutation contributes to sex-dependent alterations in central and cerebral vascular function similar to phenotypic changes associated with normal aging in healthy control mice. In vivo ultrasound imaging of central and cerebral vasculature was performed in 6-month-old male and female MFS and C57BL/6 mice and sex-matched 12-month-old (middle-aged) healthy control mice. Our findings confirm aortic enlargement (aneurysm) and wall stiffness in MFS mice, but with exacerbation in male diameters. Coronary artery blood flow velocity (BFV) in diastole was not different but left pulmonary artery BFV was decreased in MFS and 12-month-old control mice regardless of sex. At 6 months of age, MFS male mice show decreased posterior cerebral artery BFV as compared to age-matched control males, with no difference observed between female cohorts. Reduced mitral valve early-filling velocities were indicated in MFS mice regardless of sex. Male MFS mice also demonstrated left ventricular hypertrophy. Overall, these results underscore the significance of biological sex in vascular function and structure in MFS mice, while highlighting a trend of pre-mature vascular aging phenotype in MFS mice that is comparable to phenotypes observed in older healthy controls. Furthermore, this research is a vital step in understanding MFS's broader implications and sets the stage for more in-depth future analyses, while providing data-driven preclinical justification for re-evaluating diagnostic approaches and therapeutic efficacy.
Collapse
Affiliation(s)
- T Curry
- College of Medicine-Phoenix, University of Arizona, Phoenix, AZ, USA
- Barrow Neurological Institute at Phoenix Children's Hospital, Phoenix, AZ, USA
| | - M E Barrameda
- Biomedical Sciences Program, College of Graduate Studies, Midwestern University, 19555 N 59th Ave., Glendale, AZ, 85308, USA
| | - T Currier Thomas
- College of Medicine-Phoenix, University of Arizona, Phoenix, AZ, USA.
- Barrow Neurological Institute at Phoenix Children's Hospital, Phoenix, AZ, USA.
- Biomedical Sciences Program, College of Graduate Studies, Midwestern University, 19555 N 59th Ave., Glendale, AZ, 85308, USA.
- Arizona State University, Tempe, AZ, USA.
- Phoenix VA Health Care System, Phoenix, AZ, USA.
| | - M Esfandiarei
- College of Medicine-Phoenix, University of Arizona, Phoenix, AZ, USA.
- Biomedical Sciences Program, College of Graduate Studies, Midwestern University, 19555 N 59th Ave., Glendale, AZ, 85308, USA.
- Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada.
| |
Collapse
|
3
|
Fameli N, van Breemen C, Groschner K. Nanojunctions: Specificity of Ca 2+ signaling requires nano-scale architecture of intracellular membrane contact sites. Cell Calcium 2024; 117:102837. [PMID: 38011822 DOI: 10.1016/j.ceca.2023.102837] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Revised: 10/31/2023] [Accepted: 11/17/2023] [Indexed: 11/29/2023]
Abstract
Spatio-temporal definition of Ca2+ signals involves the assembly of signaling complexes within the nano-architecture of contact sites between the sarco/endoplasmic reticulum (SR/ER) and the plasma membrane (PM). While the requirement of precise spatial assembly and positioning of the junctional signaling elements is well documented, the role of the nano-scale membrane architecture itself, as an ion-reflecting confinement of the signalling unit, remains as yet elusive. Utilizing the Na+/Ca2+ Exchanger-1 / SR/ER Ca2+ ATPase-2-mediated ER Ca2+ refilling process as a junctional signalling paradigm, we provide here the first evidence for an indispensable cellular function of the junctional membrane architecture. Our stochastic modeling approach demonstrates that junctional ER Ca2+ refilling operates exclusively at nano-scale membrane spacing, with a strong inverse relationship between junctional width and signaling efficiency. Our model predicts a breakdown of junctional Ca2+ signaling with loss of reflecting membrane confinement. In addition we consider interactions between Ca2+ and the phospholipid membrane surface, which may support interfacial Ca2+ transport and promote receptor targeting. Alterations in the molecular and nano-scale membrane organization at organelle-PM contacts are suggested as a new concept in pathophysiology.
Collapse
Affiliation(s)
| | - Cornelis van Breemen
- Department of Anesthesiology, Pharmacology, and Therapeutics, The University of British Columbia, Vancouver, British Columbia, Canada
| | - Klaus Groschner
- Gottfried Schatz Research Center for Cell Signaling, Metabolism and Aging, Medical University of Graz, Graz, Austria.
| |
Collapse
|
4
|
Curry T, Barrameda ME, Currier Thomas T, Esfandiarei M. In Vivo Phenotypic Vascular Dysfunction Extends Beyond the Aorta in a Mouse Model for Fibrillin-1 ( FBN1 ) Mutation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.11.18.567641. [PMID: 38014144 PMCID: PMC10680800 DOI: 10.1101/2023.11.18.567641] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2023]
Abstract
In individuals with Marfan Syndrome (MFS), fibrillin-1 gene ( FBN1 ) mutations can lead to vascular wall weakening and dysfunction. The experimental mouse model of MFS ( FBN1 C1041G/+ ) has been advantageous in investigating MFS-associated life-threatening aortic aneurysms. Although the MFS mouse model presents an accelerated-aging phenotype in elastic organs (e.g., lung, skin), the impact of FBN1 mutations on other central and peripheral arteries function and structure with the consideration of the impact of sex remains underexplored. In this study, we investigate if FBN1 mutation contributes to sex-dependent alterations in central and cerebral vascular function similar to phenotypic changes associated with normal aging in healthy control mice. In vivo ultrasound imaging of central and cerebral vasculature was performed in 6-month-old male and female MFS and C57BL/6 mice and sex-matched 12-month-old (middle-aged) healthy control mice. Our findings confirm aortic enlargement (aneurysm) and wall stiffness in MFS mice, but with exacerbation in male diameters. Coronary artery blood flow velocity (BFV) in diastole was not different but left pulmonary artery BFV was decreased in MFS and 12-month-old control mice regardless of sex. At 6 months of age, MFS male mice show decreased posterior cerebral artery BFV as compared to age-matched control males, with no difference observed between female cohorts. Reduced mitral valve early-filling velocities were indicated in MFS mice regardless of sex. Male MFS mice also demonstrated left ventricular hypertrophy. Overall, these results underscore the significance of biological sex in vascular function and structure in MFS mice, while highlighting a trend of pre-mature vascular aging phenotype in MFS mice that is comparable to phenotypes observed in older healthy controls.
Collapse
|
5
|
Fibrillin-1 Regulates Arteriole Integrity in the Retina. Biomolecules 2022; 12:biom12101330. [PMID: 36291539 PMCID: PMC9599515 DOI: 10.3390/biom12101330] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Revised: 09/16/2022] [Accepted: 09/17/2022] [Indexed: 11/26/2022] Open
Abstract
Fibrillin-1 is an extracellular matrix protein that assembles into microfibrils that provide critical functions in large blood vessels and other tissues. Mutations in the fibrillin-1 gene are associated with cardiovascular, ocular, and skeletal abnormalities in Marfan syndrome. Fibrillin-1 is a component of the wall of large arteries but has been poorly described in other vessels. We examined the microvasculature in the retina using wild type mice and two models of Marfan syndrome, Fbn1C1041G/+ and Fbn1mgR/mgR. In the mouse retina, fibrillin-1 was detected around arterioles, in close contact with the basement membrane, where it colocalized with MAGP1. Both a mutation in fibrillin-1 or fibrillin-1 underexpression characteristically altered the microvasculature. In Fbn1C1041G/+ and Fbn1mgR/mgR mice, arterioles were enlarged with reduced MAGP1 deposition and focal loss of smooth muscle cell coverage. Losartan, which prevents aortic enlargement in Fbn1C1041G/+ mice, prevented smooth muscle cell loss and vessel leakiness when administrated in a preventive mode. Moreover, losartan also partially rescued the defects in a curative mode. Thus, fibrillin-1/MAGP1 performs essential functions in arteriolar integrity and mutant fibrillin-1-induced defects can be prevented or partially rescued pharmacologically. These new findings could have implications for people with Marfan syndrome.
Collapse
|
6
|
Weismann CG, Hlebowicz J, Åkesson A, Liuba P, Hanseus K. Comprehensive Characterization of Arterial and Cardiac Function in Marfan Syndrome-Can Biomarkers Help Improve Outcome? Front Physiol 2022; 13:873373. [PMID: 35547588 PMCID: PMC9081671 DOI: 10.3389/fphys.2022.873373] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Accepted: 03/14/2022] [Indexed: 11/17/2022] Open
Abstract
Background: Marfan Syndrome (MFS) has been associated with increased aortic stiffness and left ventricular dysfunction. The latter may be due to the underlying genotype and/or secondary to aortic stiffening (vascular-ventricular interaction). The aim of this study was to characterize arterial and cardiac function in MFS using a multimodal approach. Methods: Prospective observational study of MFS patients and healthy controls. Methods included echocardiography, ascending aortic distensibility, common carotid intima media thickness [cIMT], parameters of wave reflection, carotid-femoral pulse wave velocity [cfPWV]), reactive hyperemia index [RHI], and biomarker analysis (Olink, CVII panel). Results: We included 20 patients with MFS and 67 controls. Ascending aortic distensibility, cIMT and RHI were decreased, while all parameters of arterial wave reflection, stiffness and BNP levels were increased in the MFS group. Both systolic and diastolic function were impaired relative to controls. Within the MFS group, no significant correlation between arterial and cardiac function was identified. However, cfPWV correlated significantly with indexed left ventricular mass and volume in MFS. Bran natriuretic peptide (BNP) was the only biomarker significantly elevated in MFS following correction for age and sex. Conclusions: MFS patients have generally increased aortic stiffness, endothelial dysfunction and BNP levels while cIMT is decreased, supporting that the mechanism of general stiffening is different from acquired vascular disease. CfPWV is associated with cardiac size, blood pressure and BNP in MFS patients. These may be early markers of disease progression that are suitable for monitoring pharmacological treatment effects in MFS patients.
Collapse
Affiliation(s)
- Constance G Weismann
- Department of Clinical Sciences, Lund University, Lund, Sweden.,Department of Pediatric Cardiology, Pediatric Heart Center, Skåne University Hospital, Lund, Sweden.,Department of Pediatric Cardiology and Pediatric Intensive Care Medicine, Ludwig-Maximilian University Hospital, Munich, Germany
| | - Joanna Hlebowicz
- Department of Clinical Sciences, Lund University, Lund, Sweden.,Department of Cardiology, Skåne University Hospital, Lund University, Lund, Sweden
| | - Anna Åkesson
- Clinical Studies Sweden - Froum South, Skåne University Hospital, Lund, Sweden
| | - Petru Liuba
- Department of Clinical Sciences, Lund University, Lund, Sweden.,Department of Pediatric Cardiology, Pediatric Heart Center, Skåne University Hospital, Lund, Sweden
| | - Katarina Hanseus
- Department of Clinical Sciences, Lund University, Lund, Sweden.,Department of Pediatric Cardiology, Pediatric Heart Center, Skåne University Hospital, Lund, Sweden
| |
Collapse
|
7
|
Camarda JA, Dholakia RJ, Wang H, Samyn MM, Cava JR, LaDisa JF. A Pilot Study Characterizing Flow Patterns in the Thoracic Aorta of Patients With Connective Tissue Disease: Comparison to Age- and Gender-Matched Controls via Fluid Structure Interaction. Front Pediatr 2022; 10:772142. [PMID: 35601426 PMCID: PMC9114664 DOI: 10.3389/fped.2022.772142] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Accepted: 03/25/2022] [Indexed: 12/02/2022] Open
Abstract
Prior computational and imaging studies described changes in flow patterns for patients with Marfan syndrome, but studies are lacking for related populations. This pilot study addresses this void by characterizing wall shear stress (WSS) indices for patients with Loeys-Dietz and undifferentiated connective tissue diseases. Using aortic valve-based velocity profiles from magnetic resonance imaging as input to patient-specific fluid structure interaction (FSI) models, we determined local flow patterns throughout the aorta for four patients with various connective tissue diseases (Loeys-Dietz with the native aorta, connective tissue disease of unclear etiology with native aorta in female and male patients, and an untreated patient with Marfan syndrome, as well as twin patients with Marfan syndrome who underwent valve-sparing root replacement). FSI simulations used physiological boundary conditions and material properties to replicate available measurements. Time-averaged WSS (TAWSS) and oscillatory shear index (OSI) results are presented with localized comparison to age- and gender-matched control participants. Ascending aortic dimensions were greater in almost all patients with connective tissue diseases relative to their respective control. Differences in TAWSS and OSI were driven by local morphological differences and cardiac output. For example, the model for one twin had a more pronounced proximal descending aorta in the vicinity of the ductus ligamentum that impacted WSS indices relative to the other. We are optimistic that the results of this study can serve as a foundation for larger future studies on the connective tissue disorders presented in this article.
Collapse
Affiliation(s)
- Joseph A Camarda
- Department of Pediatrics, Division of Cardiology, Herma Heart Institute, Children's Wisconsin and the Medical College of Wisconsin, Milwaukee, WI, United States
| | - Ronak J Dholakia
- Department of Biomedical Engineering, Marquette University the Medical College of Wisconsin, Milwaukee, WI, United States
| | - Hongfeng Wang
- Department of Biomedical Engineering, Marquette University the Medical College of Wisconsin, Milwaukee, WI, United States
| | - Margaret M Samyn
- Department of Pediatrics, Division of Cardiology, Herma Heart Institute, Children's Wisconsin and the Medical College of Wisconsin, Milwaukee, WI, United States.,Department of Biomedical Engineering, Marquette University the Medical College of Wisconsin, Milwaukee, WI, United States
| | - Joseph R Cava
- Department of Pediatrics, Division of Cardiology, Herma Heart Institute, Children's Wisconsin and the Medical College of Wisconsin, Milwaukee, WI, United States
| | - John F LaDisa
- Department of Pediatrics, Division of Cardiology, Herma Heart Institute, Children's Wisconsin and the Medical College of Wisconsin, Milwaukee, WI, United States.,Department of Biomedical Engineering, Marquette University the Medical College of Wisconsin, Milwaukee, WI, United States.,Departments of Medicine, Division of Cardiovascular Medicine and Physiology, Medical College of Wisconsin, Milwaukee, WI, United States
| |
Collapse
|
8
|
Tehrani AY, Ciufolini MA, Bernatchez P. Nitric oxide in the Marfan vasculature: Friend or foe? Nitric Oxide 2021; 116:27-34. [PMID: 34478846 DOI: 10.1016/j.niox.2021.08.006] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2021] [Revised: 08/13/2021] [Accepted: 08/25/2021] [Indexed: 10/20/2022]
Abstract
Marfan syndrome (MFS) is a connective tissue disorder caused by mutations in the FBN1 gene, which encodes fibrillin-1, a protein essential for the formation and stabilization of elastic fibers as well as signaling homeostasis. Progressive aortic root widening is the most serious manifestation of MFS as it can lead to aortic dissection, aneurysm formation and rupture. However, despite their ability to decrease the hemodynamic stress the aorta is subjected to, anti-hypertensive medications often lead to underwhelming reductions in the rate of aortic root dilation, which illustrates how fragmental our understanding of MFS-associated aortic remodeling is. This manuscript summarizes recent evidence that document nitric oxide (NO) synthase (NOS)-related changes to the vasculature during the pathogenesis of MFS and how they result in a unique state of vascular dysfunction that likely plays a causal role in the aortic root widening process. We also review how clinic-approved and experimental therapies as well lifestyle approaches may promote aortic root stability by correcting NO homeostasis, which if properly optimized may improve outcomes in this population afflicted by a notoriously refractory type of aortopathy.
Collapse
Affiliation(s)
- Arash Y Tehrani
- Centre for Heart + Lung Innovation, St. Paul's Hospital, Department of Anesthesiology, Pharmacology & Therapeutics, Faculty of Medicine, University of British Columbia (UBC), Vancouver, Canada
| | | | - Pascal Bernatchez
- Centre for Heart + Lung Innovation, St. Paul's Hospital, Department of Anesthesiology, Pharmacology & Therapeutics, Faculty of Medicine, University of British Columbia (UBC), Vancouver, Canada.
| |
Collapse
|
9
|
Tehrani AY, White Z, Milad N, Esfandiarei M, Seidman MA, Bernatchez P. Blood pressure-independent inhibition of Marfan aortic root widening by the angiotensin II receptor blocker valsartan. Physiol Rep 2021; 9:e14877. [PMID: 34042309 PMCID: PMC8157789 DOI: 10.14814/phy2.14877] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2021] [Revised: 04/11/2021] [Accepted: 04/20/2021] [Indexed: 12/11/2022] Open
Abstract
Marfan syndrome (MFS) is a genetic disorder that results in accelerated aortic root widening and aneurysm. However, management of MFS patients with blood pressure (BP)-lowering medications, such as angiotensin II (AngII) receptor blocker (ARB) losartan, continues to pose challenges due to their questionable efficacy at attenuating the rate of aortic root widening in patients. Herein we investigate the anti-aortic root widening effects of a sub-BP-lowering dose valsartan, an ARB previously linked to non-BP lowering anti-remodeling effects. Despite absence of BP-lowering effects, valsartan attenuated MFS aortic root widening by 75.9%, which was similar to a hypotensive dose of losartan (79.4%) when assessed by ultrasound echocardiography. Medial thickening, elastic fiber fragmentation, and phospho-ERK signaling were also inhibited to a similar degree with both treatments. Valsartan and losartan decreased vascular contractility ex vivo between 60% and 80%, in a nitric oxide (NO)-sensitive fashion. Valsartan increased acetylcholine (Ach)-induced vessel relaxation and phospho-eNOS levels in the aortic vessel supporting BP-independent activation of protective endothelial function, which is critical to ARB-mediated aortic root stability. This study supports the concept of achieving aortic root stability with valsartan in absence of BP-lowering effects, which may help address efficacy and compliance issues with losartan-based MFS patient management.
Collapse
Affiliation(s)
- Arash Y. Tehrani
- Centre for Heart Lung InnovationUniversity of British ColumbiaVancouverBCCanada
- Department of AnesthesiologyPharmacology & TherapeuticsUniversity of British ColumbiaVancouverBCCanada
| | - Zoe White
- Centre for Heart Lung InnovationUniversity of British ColumbiaVancouverBCCanada
- Department of AnesthesiologyPharmacology & TherapeuticsUniversity of British ColumbiaVancouverBCCanada
| | - Nadia Milad
- Centre for Heart Lung InnovationUniversity of British ColumbiaVancouverBCCanada
- Department of AnesthesiologyPharmacology & TherapeuticsUniversity of British ColumbiaVancouverBCCanada
| | - Mitra Esfandiarei
- Department of AnesthesiologyPharmacology & TherapeuticsUniversity of British ColumbiaVancouverBCCanada
- Department of Biomedical SciencesCollege of Graduate StudiesMidwestern UniversityGlendaleArizonaUSA
| | - Michael A. Seidman
- Centre for Heart Lung InnovationDepartment of Pathology and Laboratory MedicineUniversity of British ColumbiaVancouverBCCanada
| | - Pascal Bernatchez
- Centre for Heart Lung InnovationUniversity of British ColumbiaVancouverBCCanada
- Department of AnesthesiologyPharmacology & TherapeuticsUniversity of British ColumbiaVancouverBCCanada
| |
Collapse
|
10
|
Cui JZ, Harris KC, Raedschelders K, Hollander Z, Potts JE, De Souza A, Kiess M, McManus BM, Bernatchez P, Raffin LA, Paine H, van Breemen C, Sandor GGS, Esfandiarei M. Aortic Dimensions, Biophysical Properties, and Plasma Biomarkers in Children and Adults with Marfan or Loeys-Dietz Syndrome. CJC Open 2020; 3:585-594. [PMID: 34027363 PMCID: PMC8134910 DOI: 10.1016/j.cjco.2020.12.018] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Accepted: 12/20/2020] [Indexed: 12/13/2022] Open
Abstract
Background Aortic dilation, stiffening, and dissection are common and potentially lethal complications of Marfan syndrome (MFS) and Loeys-Dietz syndrome (LDS), which involve abnormal transforming growth factor beta (TGF-β) signalling. The relation of aortic dimensions, stiffness, and biomarker levels is unknown. The objective of this study was to measure aortic dimensions, stiffness, TGF-β and matrix metalloproteinase (MMP) levels, and endothelial function in patients with MFS, and to compare TGF-β levels in patients with MFS receiving different therapeutic regimens. Methods This was a cohort study of 40 MFS and 4 LDS patients and 87 control participants. Aortic dimension and stiffness indexes, including pulse wave velocity (PWV), were measured using echocardiography and Doppler. Total and free TGF-β and MMP blood levels were measured using Quantikine (R&D Systems, Inc, Minneapolis, MN) and Quanterix (Billerica, MA) kits. Endothelial function was measured using brachial artery flow-mediated dilation. Results PWV was increased in patients with MFS. There were increased MMP-2 levels in those with MFS but no increase in free or total TGF-β or MMP-9 levels compared with control participants. There was no difference in TGF-β levels between MFS patients receiving no medications, angiotensin receptor blockers, and β-blockers. PWV correlated most strongly with age. Endothelial function showed premature gradual decline in patients with MFS. Conclusions Despite the increased PWV, monitoring aortic stiffness or TGF-β levels would not be helpful in patients with MFS. TGF-β levels were not increased and the increased MMP-2 levels suggest consideration of a different therapeutic target.
Collapse
Affiliation(s)
- Jason Z Cui
- Department of Anesthesiology, Pharmacology and Therapeutics, British Columbia Children's Hospital Child and Family Research Institute, University of British Columbia, Vancouver, British Columbia, Canada.,Department of Cardiothoracic Surgery, School of Medicine, Stanford University, Palo Alto, California, USA
| | - Kevin C Harris
- Children's Heart Centre, British Columbia Children's Hospital, University of British Columbia, Vancouver, British Columbia, Canada
| | - Koen Raedschelders
- Advanced Clinical Biosystems Research Institute at Smidt Heart Institute, Los Angeles, California, USA
| | - Zsuzsanna Hollander
- UBC James Hogg Research Centre, University of British Columbia, Vancouver, British Columbia, Canada
| | - James E Potts
- Children's Heart Centre, British Columbia Children's Hospital, University of British Columbia, Vancouver, British Columbia, Canada
| | - Astrid De Souza
- Children's Heart Centre, British Columbia Children's Hospital, University of British Columbia, Vancouver, British Columbia, Canada
| | - Marla Kiess
- Division of Cardiology, St Paul's Hospital, Vancouver, British Columbia, Canada
| | - Bruce M McManus
- UBC James Hogg Research Centre, University of British Columbia, Vancouver, British Columbia, Canada.,Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | - Pascal Bernatchez
- Department of Anesthesiology, Pharmacology and Therapeutics, Centre for Heart and Lung Innovation, St Paul's Hospital, University of British Columbia, Vancouver, British Columbia, Canada
| | - Leslie A Raffin
- Children's Heart Centre, British Columbia Children's Hospital, University of British Columbia, Vancouver, British Columbia, Canada
| | - Heidi Paine
- Children's Heart Centre, British Columbia Children's Hospital, University of British Columbia, Vancouver, British Columbia, Canada
| | - Cornelis van Breemen
- Department of Anesthesiology, Pharmacology and Therapeutics, British Columbia Children's Hospital Child and Family Research Institute, University of British Columbia, Vancouver, British Columbia, Canada
| | - George G S Sandor
- Children's Heart Centre, British Columbia Children's Hospital, University of British Columbia, Vancouver, British Columbia, Canada
| | - Mitra Esfandiarei
- Department of Anesthesiology, Pharmacology and Therapeutics, British Columbia Children's Hospital Child and Family Research Institute, University of British Columbia, Vancouver, British Columbia, Canada.,Department of Biomedical Sciences, College of Graduate Studies, Midwestern University, Glendale, Arizona, USA
| |
Collapse
|
11
|
Stenosis coexists with compromised α1-adrenergic contractions in the ascending aorta of a mouse model of Williams-Beuren syndrome. Sci Rep 2020; 10:889. [PMID: 31965005 PMCID: PMC6972706 DOI: 10.1038/s41598-020-57803-3] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2019] [Accepted: 01/07/2020] [Indexed: 11/08/2022] Open
Abstract
Williams-Beuren syndrome (WBS) is a rare disorder caused by a heterozygous deletion of 26–28 contiguous genes that affects the brain and cardiovascular system. Here, we investigated whether WBS affects aortic structure and function in the complete deletion (CD) mouse model harbouring the most common deletion found in WBS patients. Thoracic aortas from 3–4 months-old male CD mice and wild-type littermates were mounted in wire myographs or were processed for histomorphometrical analysis. Nitric oxide synthase (NOS) isoforms and oxidative stress levels were assessed. Ascending aortas from young adult CD mice showed moderate (50%) luminal stenosis, whereas endothelial function and oxidative stress were comparable to wild-type. CD mice showed greater contractions to KCl. However, α1-adrenergic contractions to phenylephrine, but not with a thromboxane analogue, were compromised. Decreased phenylephrine responses were not affected by selective inducible NOS blockade with 1400 W, but were prevented by the non-selective NOS inhibitor L-NAME and the selective neuronal NOS inhibitor SMTC. Consistently, CD mice showed increased neuronal NOS expression in aortas. Overall, aortic stenosis in CD mice coexists with excessive nNOS-derived NO signaling that compromises ascending aorta α1-adrenergic contractions. We suggest that increased neuronal NOS signaling may act as a physiological ‘brake’ against the detrimental effects of stenosis.
Collapse
|
12
|
Csala A, Zwinderman AH, Hof MH. Multiset sparse partial least squares path modeling for high dimensional omics data analysis. BMC Bioinformatics 2020; 21:9. [PMID: 31918677 PMCID: PMC6953292 DOI: 10.1186/s12859-019-3286-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2019] [Accepted: 11/20/2019] [Indexed: 12/29/2022] Open
Abstract
BACKGROUND Recent technological developments have enabled the measurement of a plethora of biomolecular data from various omics domains, and research is ongoing on statistical methods to leverage these omics data to better model and understand biological pathways and genetic architectures of complex phenotypes. Current reviews report that the simultaneous analysis of multiple (i.e. three or more) high dimensional omics data sources is still challenging and suitable statistical methods are unavailable. Often mentioned challenges are the lack of accounting for the hierarchical structure between omics domains and the difficulty of interpretation of genomewide results. This study is motivated to address these challenges. We propose multiset sparse Partial Least Squares path modeling (msPLS), a generalized penalized form of Partial Least Squares path modeling, for the simultaneous modeling of biological pathways across multiple omics domains. msPLS simultaneously models the effect of multiple molecular markers, from multiple omics domains, on the variation of multiple phenotypic variables, while accounting for the relationships between data sources, and provides sparse results. The sparsity in the model helps to provide interpretable results from analyses of hundreds of thousands of biomolecular variables. RESULTS With simulation studies, we quantified the ability of msPLS to discover associated variables among high dimensional data sources. Furthermore, we analysed high dimensional omics datasets to explore biological pathways associated with Marfan syndrome and with Chronic Lymphocytic Leukaemia. Additionally, we compared the results of msPLS to the results of Multi-Omics Factor Analysis (MOFA), which is an alternative method to analyse this type of data. CONCLUSIONS msPLS is an multiset multivariate method for the integrative analysis of multiple high dimensional omics data sources. It accounts for the relationship between multiple high dimensional data sources while it provides interpretable results through its sparse solutions. The biomarkers found by msPLS in the omics datasets can be interpreted in terms of biological pathways associated with the pathophysiology of Marfan syndrome and of Chronic Lymphocytic Leukaemia. Additionally, msPLS outperforms MOFA in terms of variation explained in the chronic lymphocytic leukaemia dataset while it identifies the two most important clinical markers for Chronic Lymphocytic Leukaemia AVAILABILITY: http://uva.csala.me/mspls.https://github.com/acsala/2018_msPLS.
Collapse
Affiliation(s)
- Attila Csala
- Department of Clinical Epidemiology, Biostatistics and Bioinformatics, University of Amsterdam, Amsterdam, 1105 AZ The Netherlands
| | - Aeilko H. Zwinderman
- Department of Clinical Epidemiology, Biostatistics and Bioinformatics, University of Amsterdam, Amsterdam, 1105 AZ The Netherlands
| | - Michel H. Hof
- Department of Clinical Epidemiology, Biostatistics and Bioinformatics, University of Amsterdam, Amsterdam, 1105 AZ The Netherlands
| |
Collapse
|
13
|
Hao H, Tian W, Pan C, Jiao Y, Deng X, Fan J, Han J, Han S, Wang M, Li P. Marsdenia tenacissima extract dilated small mesenteric arteries via stimulating endothelial nitric oxide synthase and inhibiting calcium influx. JOURNAL OF ETHNOPHARMACOLOGY 2019; 238:111847. [PMID: 30946966 DOI: 10.1016/j.jep.2019.111847] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/20/2018] [Revised: 03/19/2019] [Accepted: 03/30/2019] [Indexed: 06/09/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Marsdenia tenacissima is a traditional Chinese medicine that is known to be effective in combating cancer as well as reducing blood pressure. The efficacy and mechanisms of Marsdenia tenacissima in treating cancer have been well described. However, the potential vasoactivities of Marsdenia tenacissima remain poorly known. AIM OF THE STUDY To determine the vasoactive effects of the water-soluble part of marsdenia tenacissima in mesenteric resistance arteries of the mice, and to explore the underlying mechanisms. MATERIALS AND METHODS Isometric vessel tension study was used to examine the effects of marsdenia tenacissima extract (MTE) on vasodilation of the mesenteric arteries of mice. KCl, phenylephrine (PE) and 9,11-Dideoxy-11α,9α-epoxymethanoprostaglandin F2α (U46619) were used as vasoconstrictors. Y27632, Nitro-L-arginine methyl ester hydrochloride (L-NAME) and indomethacin were used to explore the underlying mechanisms for the vasoactivities of MTE. Western blot and nitric oxide (NO) assay were used to evaluate the effects of MTE on the activities of endothelial nitric oxide synthase (eNOS). RESULTS MTE (5-50 mg/mL), but not vehicle, dose-dependently relaxed the mesenteric arteries constricted with KCl, PE or U46619, in which relaxations to KCl were more pronounced than that to PE or U46619. Pre-incubation of the vessels with MTE (40 mg/mL) reduced the vasoconstrictions caused by calcium influx. Decreasing calcium sensitivity by inhibition of Rho kinase (ROCK) significantly augmented the vasorelaxation of MTE. While, inhibition of endothelial cells by pre-incubation with L-NAME (300 μM) and indomethacin (10 μM) or denudating endothelial cells attenuated vasorelaxations of MTE to KCl, and with a larger potency, to U46619. In both human umbilical vein endothelial cells (HUVECs) and human heart microvascular endothelial cells (HMECs), the phosphorylations of eNOS and the production of NO were significantly enhanced after treatment of MTE for 2, 5, 10, 30 min. CONCLUSIONS MTE, the water-soluble part of marsdenia tenacissima, was effective in relaxing mesenteric resistance arteries via inhibiting calcium influx and stimulating eNOS activities.
Collapse
Affiliation(s)
- Huifeng Hao
- Department of Integration of Chinese and Western Medicine, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education, Beijing), Peking University Cancer Hospital & Institute, Beijing, 100142, PR China
| | - Wenjia Tian
- Department of Gastroenterology, Peking University International Hospital, Beijing, 102206, PR China
| | - Chunshui Pan
- Tasly Microcirculation Research Center, Peking University Health Science Center, Beijing, 100191, PR China
| | - Yanna Jiao
- Department of Integration of Chinese and Western Medicine, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education, Beijing), Peking University Cancer Hospital & Institute, Beijing, 100142, PR China
| | - Xinxin Deng
- Ningxia Medical University Pharmacy College, Key Laboratory of Hui Ethnic Medicine Modernization, Ministry of Education, Ningxia Research Center of Modern Hui Medicine Engineering and Technology, Yinchuan, 750004, PR China
| | - Jingyu Fan
- Tasly Microcirculation Research Center, Peking University Health Science Center, Beijing, 100191, PR China
| | - Jingyan Han
- Tasly Microcirculation Research Center, Peking University Health Science Center, Beijing, 100191, PR China; Department of Integration of Chinese and Western Medicine, School of Basic Medical Sciences, Peking University, Beijing, 100191, PR China
| | - Shuyan Han
- Department of Integration of Chinese and Western Medicine, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education, Beijing), Peking University Cancer Hospital & Institute, Beijing, 100142, PR China.
| | - Miao Wang
- State Key Laboratory of Cardiovascular Disease, and Clinical Pharmacology Center, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100037, China.
| | - Pingping Li
- Department of Integration of Chinese and Western Medicine, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education, Beijing), Peking University Cancer Hospital & Institute, Beijing, 100142, PR China.
| |
Collapse
|
14
|
Sellers SL, Milad N, Chan R, Mielnik M, Jermilova U, Huang PL, de Crom R, Hirota JA, Hogg JC, Sandor GG, Van Breemen C, Esfandiarei M, Seidman MA, Bernatchez P. Inhibition of Marfan Syndrome Aortic Root Dilation by Losartan: Role of Angiotensin II Receptor Type 1-Independent Activation of Endothelial Function. THE AMERICAN JOURNAL OF PATHOLOGY 2019; 188:574-585. [PMID: 29433732 DOI: 10.1016/j.ajpath.2017.11.006] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/23/2017] [Revised: 10/18/2017] [Accepted: 11/16/2017] [Indexed: 01/13/2023]
Abstract
Marfan syndrome (MFS) is a genetic disorder that frequently leads to aortic root dissection and aneurysm. Despite promising preclinical and pilot clinical data, a recent large-scale study using antihypertensive angiotensin II (AngII) receptor type 1 (ATR1) blocker losartan has failed to meet expectations at preventing MFS-associated aortic root dilation, casting doubts about optimal therapy. To study the deleterious role of normal ATR1 signaling in aortic root widening, we generated MFS mice lacking ATR1a expression in an attempt to preserve protective ATR2 signaling. Despite being hypotensive and resistant to AngII vasopressor effects, MFS/ATR1a-null mice showed unabated aortic root enlargement and remained fully responsive to losartan, confirming that blood pressure lowering is of minor therapeutic value in MFS and that losartan's antiremodeling properties may be ATR1 independent. Having shown that MFS causes endothelial dysfunction and that losartan can activate endothelial function in mice and patients, we found that nitric oxide synthase (NOS) inhibition renders losartan therapeutically inactive, whereas multiple transgenic and pharmacologic models of endothelial NOS activation block aortic root dilation by correcting extracellular signal-regulated kinase signaling. In vitro, losartan can increase endothelial NO release in the absence of AngII and correct MFS NO levels in vivo. Our data suggest that increased protective endothelial function, rather than ATR1 inhibition or blood pressure lowering, might be of therapeutic significance in preventing aortic root disease in MFS.
Collapse
Affiliation(s)
- Stephanie L Sellers
- Department of Anesthesiology, Pharmacology and Therapeutics, University of British Columbia (UBC), Vancouver, British Columbia, Canada; UBC Centre for Heart Lung Innovation and St. Paul's Hospital, University of British Columbia (UBC), Vancouver, British Columbia, Canada
| | - Nadia Milad
- Department of Anesthesiology, Pharmacology and Therapeutics, University of British Columbia (UBC), Vancouver, British Columbia, Canada; UBC Centre for Heart Lung Innovation and St. Paul's Hospital, University of British Columbia (UBC), Vancouver, British Columbia, Canada
| | - Rayleigh Chan
- Department of Anesthesiology, Pharmacology and Therapeutics, University of British Columbia (UBC), Vancouver, British Columbia, Canada; UBC Centre for Heart Lung Innovation and St. Paul's Hospital, University of British Columbia (UBC), Vancouver, British Columbia, Canada
| | - Michael Mielnik
- Department of Anesthesiology, Pharmacology and Therapeutics, University of British Columbia (UBC), Vancouver, British Columbia, Canada; UBC Centre for Heart Lung Innovation and St. Paul's Hospital, University of British Columbia (UBC), Vancouver, British Columbia, Canada
| | - Una Jermilova
- Department of Anesthesiology, Pharmacology and Therapeutics, University of British Columbia (UBC), Vancouver, British Columbia, Canada; UBC Centre for Heart Lung Innovation and St. Paul's Hospital, University of British Columbia (UBC), Vancouver, British Columbia, Canada
| | - Paul L Huang
- Cardiovascular Research Centre, Massachusetts General Hospital, Harvard University, Charlestown, Massachusetts
| | - Rini de Crom
- Department of Cell Biology and Genetics, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Jeremy A Hirota
- UBC Centre for Heart Lung Innovation and St. Paul's Hospital, University of British Columbia (UBC), Vancouver, British Columbia, Canada; Division of Respiratory Medicine, Department of Medicine, Chan-Yeung Centre for Occupational and Environmental Respiratory Disease, University of British Columbia (UBC), Vancouver, British Columbia, Canada
| | - James C Hogg
- UBC Centre for Heart Lung Innovation and St. Paul's Hospital, University of British Columbia (UBC), Vancouver, British Columbia, Canada; Department of Pathology and Laboratory Medicine, University of British Columbia (UBC), Vancouver, British Columbia, Canada
| | - George G Sandor
- Providence Health Care, and the Child and Family Research Institute, University of British Columbia (UBC), Vancouver, British Columbia, Canada
| | - Casey Van Breemen
- Providence Health Care, and the Child and Family Research Institute, University of British Columbia (UBC), Vancouver, British Columbia, Canada
| | - Mitra Esfandiarei
- Department of Biomedical Sciences, Midwestern University, Glendale, Arizona
| | - Michael A Seidman
- UBC Centre for Heart Lung Innovation and St. Paul's Hospital, University of British Columbia (UBC), Vancouver, British Columbia, Canada; Department of Pathology and Laboratory Medicine, University of British Columbia (UBC), Vancouver, British Columbia, Canada
| | - Pascal Bernatchez
- Department of Anesthesiology, Pharmacology and Therapeutics, University of British Columbia (UBC), Vancouver, British Columbia, Canada; UBC Centre for Heart Lung Innovation and St. Paul's Hospital, University of British Columbia (UBC), Vancouver, British Columbia, Canada.
| |
Collapse
|
15
|
Li M, Qian M, Kyler K, Xu J. Endothelial-Vascular Smooth Muscle Cells Interactions in Atherosclerosis. Front Cardiovasc Med 2018; 5:151. [PMID: 30406116 PMCID: PMC6207093 DOI: 10.3389/fcvm.2018.00151] [Citation(s) in RCA: 137] [Impact Index Per Article: 22.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2018] [Accepted: 10/04/2018] [Indexed: 12/20/2022] Open
Abstract
Atherosclerosis is a chronic progressive inflammatory process that can eventually lead to cardiovascular disease (CVD). Despite available treatment, the prevalence of atherosclerotic CVD, which has become the leading cause of death worldwide, persists. Identification of new mechanisms of atherogenesis are highly needed in order to develop an effective therapeutic treatment. The blood vessels contain two primary major cell types: endothelial cells (EC) and vascular smooth muscle cells (VSMC). Each of these performs an essential function in sustaining vascular homeostasis. EC-VSMC communication is essential not only to development, but also to the homeostasis of mature blood vessels. Aberrant EC-VSMC interaction could promote atherogenesis. Identification of the mode of EC-VSMC crosstalk that regulates vascular functionality and sustains homeostasis may offer strategic insights for prevention and treatment of atherosclerotic CVD. Here we will review the molecular mechanisms underlying the interplay between EC and VSMC that could contribute to atherosclerosis. We also highlight open questions for future research directions.
Collapse
Affiliation(s)
- Manna Li
- Department of Medicine, Harold Hamm Diabetes Center, University of Oklahoma Health Sciences Center, Oklahoma, OK, United States
| | - Ming Qian
- Department of Medicine, Harold Hamm Diabetes Center, University of Oklahoma Health Sciences Center, Oklahoma, OK, United States
| | - Kathy Kyler
- Office of Research Administration, University of Oklahoma Health Sciences Center, Oklahoma, OK, United States
| | - Jian Xu
- Department of Medicine, Harold Hamm Diabetes Center, University of Oklahoma Health Sciences Center, Oklahoma, OK, United States
| |
Collapse
|
16
|
Lomelí O, Pérez-Torres I, Márquez R, Críales S, Mejía AM, Chiney C, Hernández-Lemus E, Soto ME. The Evaluation of Flow-Mediated Vasodilation in the Brachial Artery Correlates With Endothelial Dysfunction Evaluated by Nitric Oxide Synthase Metabolites in Marfan Syndrome Patients. Front Physiol 2018; 9:965. [PMID: 30246778 PMCID: PMC6110856 DOI: 10.3389/fphys.2018.00965] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2017] [Accepted: 07/02/2018] [Indexed: 01/22/2023] Open
Abstract
Marfan syndrome (MS) is of the most common connective tissue disorders. Although most patients have mutations in the fibrillin-1 gene (FBN1) and more than 1,700 mutations have been described, there are no mutations in less than 10% of patients. Aortic dilation is the most important complication; it involves chronic inflammatory processes and endothelial dysfunction. Prospective study from March 2015 to January 2017, in a cohort of 32 patients of MS confirmed by Ghent criteria and 35 controls of both genders, with a median age of 26 years (18-56). Patients had no comorbidities such as diabetes, hypertension, and/or neoplasms. They were not being treated with statin, NSAIDs, calcium antagonists, oral nitrates, and/or beta-blockers during 7 days prior to the study and patients with smoking history in the last 4 years. Controls were matched by age and gender. We analyzed endothelial dysfunction by flow-mediated vasodilation in the brachial artery, determining the maximum peak flow in the reactive hyperemia phase with a Philips Envisor device with Doppler capability. Its correlation with serum levels of biological markers that could participate in endothelial dysfunction pathways such as NO3-/NO2- ratio, NO2- , citrulline, TNFα, IL-1, IL-6, IL-10, IL-8, osteopontin, ICAM, VCAM, and NO3-/NO2- was determined. Endothelial dysfunction was found in 21 MS patients (65%). The aortic annulus (AAo) was of 27 mm (22-40) and 24 mm (22-30) (p = 0.04) in MS patients with and without dysfunction. The level of NO3-/NO2- ratio, was of 108.95 ± 12.05 nM/ml in controls vs. 170.04 ± 18.76 nM/ml in MS (p = 0.002), NO2- was of 33.78 ± 3.41 vs. 43.95 ± 2.59 nM/ml (p = 0.03), citrulline 62.65 ± 3.46 vs. 72.81 ± 4.35 μMol/ml (p = 0.06). VCAM median was 39 pg/ml (0-86) vs. 32 pg/ml (11-66) (p = 0.03), respectively. The correlation of VCAM with triglycerides (TG) was of 0.62 (p = 0.005). There were no differences in TNFα, IL-1, IL-6, IL-8, IL-10, and osteopontin. MS endothelial dysfunction is related to aortic diameters, and increased levels of VCAM, L-citrulline and NO3-/NO2- ratio, NO2- . VCAM-1 has a significant correlation with TG and could play a significant role in endothelial dysfunction.
Collapse
Affiliation(s)
- Oscar Lomelí
- Department of Echocardiography, National Institute of Cardiology "Ignacio Chávez", Mexico City, Mexico
| | - Israel Pérez-Torres
- Department of Pathology, National Institute of Cardiology "Ignacio Chávez", Mexico City, Mexico
| | - Ricardo Márquez
- Department of Immunology, National Institute of Cardiology "Ignacio Chávez", Mexico City, Mexico
| | - Sergio Críales
- Department of Computed Tomography, National Institute of Cardiology "Ignacio Chávez", Mexico City, Mexico
| | - Ana M Mejía
- Blood Bank, National Institute of Cardiology "Ignacio Chávez", Mexico City, Mexico
| | - Claudia Chiney
- Central Laboratory, National Institute of Cardiology "Ignacio Chávez", Mexico City, Mexico
| | - Enrique Hernández-Lemus
- Computational Genomics Division, National Institute of Genomic Medicine, Mexico City, Mexico
| | - Maria E Soto
- Department of Immunology, National Institute of Cardiology "Ignacio Chávez", Mexico City, Mexico
| |
Collapse
|
17
|
Mas-Stachurska A, Siegert AM, Batlle M, Gorbenko Del Blanco D, Meirelles T, Rubies C, Bonorino F, Serra-Peinado C, Bijnens B, Baudin J, Sitges M, Mont L, Guasch E, Egea G. Cardiovascular Benefits of Moderate Exercise Training in Marfan Syndrome: Insights From an Animal Model. J Am Heart Assoc 2017; 6:JAHA.117.006438. [PMID: 28947563 PMCID: PMC5634291 DOI: 10.1161/jaha.117.006438] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND Marfan syndrome (MF) leads to aortic root dilatation and a predisposition to aortic dissection, mitral valve prolapse, and primary and secondary cardiomyopathy. Overall, regular physical exercise is recommended for a healthy lifestyle, but dynamic sports are strongly discouraged in MF patients. Nonetheless, evidence supporting this recommendation is lacking. Therefore, we studied the role of long-term dynamic exercise of moderate intensity on the MF cardiovascular phenotype. METHODS AND RESULTS In a transgenic mouse model of MF (Fbn1C1039G/+), 4-month-old wild-type and MF mice were subjected to training on a treadmill for 5 months; sedentary littermates served as controls for each group. Aortic and cardiac remodeling was assessed by echocardiography and histology. The 4-month-old MF mice showed aortic root dilatation, elastic lamina rupture, and tunica media fibrosis, as well as cardiac hypertrophy, left ventricular fibrosis, and intramyocardial vessel remodeling. Over the 5-month experimental period, aortic root dilation rate was significantly greater in the sedentary MF group, compared with the wild-type group (∆mm, 0.27±0.07 versus 0.13±0.02, respectively). Exercise significantly blunted the aortic root dilation rate in MF mice compared with sedentary MF littermates (∆mm, 0.10±0.04 versus 0.27±0.07, respectively). However, these 2 groups were indistinguishable by aortic root stiffness, tunica media fibrosis, and elastic lamina ruptures. In MF mice, exercise also produced cardiac hypertrophy regression without changes in left ventricular fibrosis. CONCLUSIONS Our results in a transgenic mouse model of MF indicate that moderate dynamic exercise mitigates the progression of the MF cardiovascular phenotype.
Collapse
Affiliation(s)
| | - Anna-Maria Siegert
- Departament de Biomedicina, Facultat de Medicina, Universitat de Barcelona, Spain
| | - Monsterrat Batlle
- Institut d'Investigacions Biomédiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain.,CIBERCV, Barcelona, Spain
| | | | - Thayna Meirelles
- Departament de Biomedicina, Facultat de Medicina, Universitat de Barcelona, Spain
| | - Cira Rubies
- Institut d'Investigacions Biomédiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Fabio Bonorino
- Departament de Biomedicina, Facultat de Medicina, Universitat de Barcelona, Spain
| | - Carla Serra-Peinado
- Departament de Biomedicina, Facultat de Medicina, Universitat de Barcelona, Spain
| | - Bart Bijnens
- ICREA, Barcelona, Spain.,Universitat Pompeu Fabra, Barcelona, Spain
| | - Julio Baudin
- Institut d'Investigacions Biomédiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Marta Sitges
- Institut Cardiovascular, Hospital Clínic de Barcelona Universitat de Barcelona, Spain.,Institut d'Investigacions Biomédiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain.,CIBERCV, Barcelona, Spain
| | - Lluís Mont
- Institut Cardiovascular, Hospital Clínic de Barcelona Universitat de Barcelona, Spain.,Institut d'Investigacions Biomédiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain.,CIBERCV, Barcelona, Spain
| | - Eduard Guasch
- Institut Cardiovascular, Hospital Clínic de Barcelona Universitat de Barcelona, Spain .,Institut d'Investigacions Biomédiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain.,CIBERCV, Barcelona, Spain
| | - Gustavo Egea
- Departament de Biomedicina, Facultat de Medicina, Universitat de Barcelona, Spain .,Institut de Nanociències i Nanotecnologia (IN2UB), Universitat de Barcelona, Spain.,Institut d'Investigacions Biomédiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| |
Collapse
|
18
|
Arif R, Zaradzki M, Remes A, Seppelt P, Kunze R, Schröder H, Schwill S, Ensminger SM, Robinson PN, Karck M, Müller OJ, Hecker M, Wagner AH, Kallenbach K. AP-1 Oligodeoxynucleotides Reduce Aortic Elastolysis in a Murine Model of Marfan Syndrome. MOLECULAR THERAPY. NUCLEIC ACIDS 2017; 9:69-79. [PMID: 29246325 PMCID: PMC5608502 DOI: 10.1016/j.omtn.2017.08.014] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/30/2016] [Revised: 08/28/2017] [Accepted: 08/29/2017] [Indexed: 11/28/2022]
Abstract
Marfan syndrome is characterized by high expression of matrix metalloproteinases (MMPs) in aortic smooth muscle cells (AoSMCs) associated with medial elastolysis and aortic root aneurysm. We aimed to reduce aortic elastolysis through decrease of MMP expression with decoy oligodeoxynucleotides (dODNs) neutralizing the transcription factor activating factor-1 (AP-1). AP-1 abundance in nuclear extracts as well as MMP-2 and MMP-9 expression were significantly increased in isolated mAoSMC of mgR/mgR Marfan mice compared to wild-type cells. Exposure to AP-1 neutralizing dODNs resulted in a significant reduction of basal and interleukin-1β-stimulated MMP expression and activity in mAoSMCs. Moreover, increased migration and formation of superoxide radical anions was substantially decreased in mAoSMCs by AP-1 dODN treatment. Aortic grafts from donor Marfan mice were treated with AP-1- dODN ex vivo and implanted as infrarenal aortic interposition grafts in mgR/mgR mice. Pretreatment of aortic grafts with AP-1 dODN led to reduced elastolysis, macrophage infiltration, and MMP activity. Permeability of the endothelial monolayer was increased for dODN in mgR/mgR aortae with observed loss of tight junction proteins ZO-1 and occludin, enabling dODN to reach the tunica media. Targeting AP-1 activity offers a new potential strategy to treat the vascular phenotype associated with Marfan syndrome.
Collapse
Affiliation(s)
- Rawa Arif
- Department of Cardiac Surgery, University Hospital Heidelberg, Heidelberg, Germany.
| | - Marcin Zaradzki
- Department of Cardiac Surgery, University Hospital Heidelberg, Heidelberg, Germany
| | - Anca Remes
- Institute of Physiology and Pathophysiology, Heidelberg University, Heidelberg, Germany
| | - Philipp Seppelt
- Department of Cardiac Surgery, University Hospital Heidelberg, Heidelberg, Germany
| | - Reiner Kunze
- Institute of Physiology and Pathophysiology, Heidelberg University, Heidelberg, Germany
| | - Hannes Schröder
- Institute of Physiology and Pathophysiology, Heidelberg University, Heidelberg, Germany
| | - Simon Schwill
- Department of Cardiac Surgery, University Hospital Heidelberg, Heidelberg, Germany
| | - Stephan M Ensminger
- Department of Cardiovascular Surgery, Heart and Cardiovascular Centre North Rhine-Westphalia, Ruhr University, Bochum, Germany
| | - Peter N Robinson
- Institute for Medical Genetics, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Matthias Karck
- Department of Cardiac Surgery, University Hospital Heidelberg, Heidelberg, Germany
| | - Oliver J Müller
- Department of Internal Medicine III, University Hospital Heidelberg and DZHK (German Center for Cardiovascular Research), partner site Heidelberg/Mannheim, Heidelberg, Germany
| | - Markus Hecker
- Institute of Physiology and Pathophysiology, Heidelberg University, Heidelberg, Germany
| | - Andreas H Wagner
- Institute of Physiology and Pathophysiology, Heidelberg University, Heidelberg, Germany
| | - Klaus Kallenbach
- Department of Cardiac Surgery, University Hospital Heidelberg, Heidelberg, Germany; INCCI HaerzZenter, Department of Cardiac Surgery, Luxembourg, Luxembourg
| |
Collapse
|
19
|
Gibson C, Nielsen C, Alex R, Cooper K, Farney M, Gaufin D, Cui JZ, van Breemen C, Broderick TL, Vallejo-Elias J, Esfandiarei M. Mild aerobic exercise blocks elastin fiber fragmentation and aortic dilatation in a mouse model of Marfan syndrome associated aortic aneurysm. J Appl Physiol (1985) 2017; 123:147-160. [PMID: 28385916 DOI: 10.1152/japplphysiol.00132.2017] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2017] [Revised: 03/29/2017] [Accepted: 04/03/2017] [Indexed: 11/22/2022] Open
Abstract
Regular low-impact physical activity is generally allowed in patients with Marfan syndrome, a connective tissue disorder caused by heterozygous mutations in the fibrillin-1 gene. However, being above average in height encourages young adults with this syndrome to engage in high-intensity contact sports, which unfortunately increases the risk for aortic aneurysm and rupture, the leading cause of death in Marfan syndrome. In this study, we investigated the effects of voluntary (cage-wheel) or forced (treadmill) aerobic exercise at different intensities on aortic function and structure in a mouse model of Marfan syndrome. Four-week-old Marfan and wild-type mice were subjected to voluntary and forced exercise regimens or sedentary lifestyle for 5 mo. Thoracic aortic tissue was isolated and subjected to structural and functional studies. Our data showed that exercise improved aortic wall structure and function in Marfan mice and that the beneficial effect was biphasic, with an optimum at low intensity exercise (55-65% V̇o2max) and tapering off at a higher intensity of exercise (85% V̇o2max). The mechanism underlying the reduced elastin fragmentation in Marfan mice involved reduction of the expression of matrix metalloproteinases 2 and 9 within the aortic wall. These findings present the first evidence of potential beneficial effects of mild exercise on the structural integrity of the aortic wall in Marfan syndrome associated aneurysm. Our finding that moderate, but not strenuous, exercise protects aortic structure and function in a mouse model of Marfan syndrome could have important implications for the medical care of young Marfan patients.NEW & NOTEWORTHY The present study provides conclusive scientific evidence that daily exercise can improve aortic health in a mouse model of Marfan syndrome associated aortic aneurysm, and it establishes the threshold for the exercise intensity beyond which exercise may not be as protective. These findings establish a platform for a new focus on promoting regular exercise in Marfan patients at an optimum intensity and create a paradigm shift in clinical care of Marfan patients suffering from aortic aneurysm complications.
Collapse
Affiliation(s)
| | - Cory Nielsen
- Department of Biomedical Sciences, College of Health Sciences
| | - Ramona Alex
- Department of Biomedical Sciences, College of Health Sciences
| | - Kimbal Cooper
- Department of Biomedical Sciences, College of Health Sciences
| | - Michael Farney
- Department of Biomedical Sciences, College of Health Sciences
| | - Douglas Gaufin
- Department of Biomedical Sciences, College of Health Sciences
| | - Jason Z Cui
- Department of Anesthesiology, Pharmacology, and Therapeutics, University of British Columbia, Vancouver, British Columbia, Canada
| | - Cornelis van Breemen
- Department of Anesthesiology, Pharmacology, and Therapeutics, University of British Columbia, Vancouver, British Columbia, Canada
| | - Tom L Broderick
- Department of Physiology, Arizona College of Osteopathic Medicine, Midwestern University, Glendale, Arizona; and
| | - Johana Vallejo-Elias
- Department of Physiology, Arizona College of Osteopathic Medicine, Midwestern University, Glendale, Arizona; and
| | - Mitra Esfandiarei
- Department of Biomedical Sciences, College of Health Sciences; .,Department of Anesthesiology, Pharmacology, and Therapeutics, University of British Columbia, Vancouver, British Columbia, Canada
| |
Collapse
|
20
|
De Moudt S, Leloup A, Van Hove C, De Meyer G, Fransen P. Isometric Stretch Alters Vascular Reactivity of Mouse Aortic Segments. Front Physiol 2017; 8:157. [PMID: 28360864 PMCID: PMC5352655 DOI: 10.3389/fphys.2017.00157] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2017] [Accepted: 02/28/2017] [Indexed: 02/02/2023] Open
Abstract
Most vaso-reactive studies in mouse aortic segments are performed in isometric conditions and at an optimal preload, which is the preload corresponding to a maximal contraction by non-receptor or receptor-mediated stimulation. In general, this optimal preload ranges from about 1.2 to 8.0 mN/mm, which according to Laplace's law roughly correlates with transmural pressures of 10-65 mmHg. For physiologic transmural pressures around 100 mmHg, preloads of 15.0 mN/mm should be implemented. The present study aimed to compare vascular reactivity of 2 mm mouse (C57Bl6) aortic segments preloaded at optimal (8.0 mN/mm) vs. (patho) physiological (10.0-32.5 mN/mm) preload. Voltage-dependent contractions of aortic segments, induced by increasing extracellular K+, and contractions by α1-adrenergic stimulation with phenylephrine (PE) were studied at these preloads in the absence and presence of L-NAME to inhibit basal release of NO from endothelial cells (EC). In the absence of basal NO release and with higher than optimal preload, contractions evoked by depolarization or PE were attenuated, whereas in the presence of basal release of NO PE-, but not depolarization-induced contractions were preload-independent. Phasic contractions by PE, as measured in the absence of external Ca2+, were decreased at higher than optimal preload suggestive for a lower contractile SR Ca2+ content at physiological preload. Further, in the presence of external Ca2+, contractions by Ca2+ influx via voltage-dependent Ca2+ channels were preload-independent, whereas non-selective cation channel-mediated contractions were increased. The latter contractions were very sensitive to the basal release of NO, which itself seemed to be preload-independent. Relaxation by endogenous NO (acetylcholine) of aortic segments pre-contracted with PE was preload-independent, whereas relaxation by exogenous NO (diethylamine NONOate) displayed higher sensitivity at high preload. Results indicated that stretching aortic segments to higher than optimal preload depolarizes the SMC and causes Ca2+ unloading of the contractile SR, making them extremely sensitive to small changes in the basal release of NO from EC as can occur in hypertension or arterial stiffening.
Collapse
Affiliation(s)
- Sofie De Moudt
- Laboratory of Physiopharmacology, Department of Pharmaceutical Sciences, University of Antwerp Antwerp, Belgium
| | - Arthur Leloup
- Laboratory of Physiopharmacology, Department of Pharmaceutical Sciences, University of Antwerp Antwerp, Belgium
| | - Cor Van Hove
- Laboratory of Pharmacology, Faculty of Medicine and Health Sciences, University of Antwerp Antwerp, Belgium
| | - Guido De Meyer
- Laboratory of Physiopharmacology, Department of Pharmaceutical Sciences, University of Antwerp Antwerp, Belgium
| | - Paul Fransen
- Laboratory of Physiopharmacology, Department of Pharmaceutical Sciences, University of Antwerp Antwerp, Belgium
| |
Collapse
|
21
|
Perrucci GL, Rurali E, Gowran A, Pini A, Antona C, Chiesa R, Pompilio G, Nigro P. Vascular smooth muscle cells in Marfan syndrome aneurysm: the broken bricks in the aortic wall. Cell Mol Life Sci 2017; 74:267-277. [PMID: 27535662 PMCID: PMC11107581 DOI: 10.1007/s00018-016-2324-9] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2016] [Revised: 07/14/2016] [Accepted: 08/02/2016] [Indexed: 01/22/2023]
Abstract
Marfan syndrome (MFS) is a connective tissue disorder with multiple organ manifestations. The genetic cause of this syndrome is the mutation of the FBN1 gene, encoding the extracellular matrix (ECM) protein fibrillin-1. This genetic alteration leads to the degeneration of microfibril structures and ECM integrity in the tunica media of the aorta. Indeed, thoracic aortic aneurysm and dissection represent the leading cause of death in MFS patients. To date, the most effective treatment option for this pathology is the surgical substitution of the damaged aorta. To highlight novel therapeutic targets, we review the molecular mechanisms related to MFS etiology in vascular smooth muscle cells, the foremost cellular type involved in MFS pathogenesis.
Collapse
Affiliation(s)
- Gianluca L Perrucci
- Department of Clinical Sciences and Community Health, University of Milan, Via Festa del Perdono 7, 20122, Milan, Italy
- Unit of Vascular Biology and Regenerative Medicine, Centro Cardiologico Monzino-IRCCS, Via C. Parea 4, 20138, Milan, Italy
| | - Erica Rurali
- Unit of Vascular Biology and Regenerative Medicine, Centro Cardiologico Monzino-IRCCS, Via C. Parea 4, 20138, Milan, Italy
| | - Aoife Gowran
- Unit of Vascular Biology and Regenerative Medicine, Centro Cardiologico Monzino-IRCCS, Via C. Parea 4, 20138, Milan, Italy
| | - Alessandro Pini
- Department of Cardiology, Marfan Clinic®, "Luigi Sacco" University of Milan, Via G.B. Grassi 74, 20157, Milan, Italy
| | - Carlo Antona
- Cardiovascular Surgery Department, "Luigi Sacco" University of Milan, Via G.B. Grassi 74, 20157, Milan, Italy
- FoRCardioLab, "Luigi Sacco" University of Milan, Via G.B. Grassi 74, 20157, Milan, Italy
| | - Roberto Chiesa
- Department of Vascular Surgery, San Raffaele Scientific Institute Hospital, Vita-Salute University, Milan, Italy
| | - Giulio Pompilio
- Department of Clinical Sciences and Community Health, University of Milan, Via Festa del Perdono 7, 20122, Milan, Italy.
- Unit of Vascular Biology and Regenerative Medicine, Centro Cardiologico Monzino-IRCCS, Via C. Parea 4, 20138, Milan, Italy.
- Department of Cardiovascular Surgery, Centro Cardiologico Monzino-IRCCS, Via C. Parea 4, 20138, Milan, Italy.
| | - Patrizia Nigro
- Unit of Vascular Biology and Regenerative Medicine, Centro Cardiologico Monzino-IRCCS, Via C. Parea 4, 20138, Milan, Italy.
| |
Collapse
|
22
|
An iPSC-derived vascular model of Marfan syndrome identifies key mediators of smooth muscle cell death. Nat Genet 2016; 49:97-109. [PMID: 27893734 DOI: 10.1038/ng.3723] [Citation(s) in RCA: 132] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2016] [Accepted: 10/19/2016] [Indexed: 12/14/2022]
Abstract
Marfan syndrome (MFS) is a heritable connective tissue disorder caused by mutations in FBN1, which encodes the extracellular matrix protein fibrillin-1. To investigate the pathogenesis of aortic aneurysms in MFS, we generated a vascular model derived from human induced pluripotent stem cells (MFS-hiPSCs). Our MFS-hiPSC-derived smooth muscle cells (SMCs) recapitulated the pathology seen in Marfan aortas, including defects in fibrillin-1 accumulation, extracellular matrix degradation, transforming growth factor-β (TGF-β) signaling, contraction and apoptosis; abnormalities were corrected by CRISPR-based editing of the FBN1 mutation. TGF-β inhibition rescued abnormalities in fibrillin-1 accumulation and matrix metalloproteinase expression. However, only the noncanonical p38 pathway regulated SMC apoptosis, a pathological mechanism also governed by Krüppel-like factor 4 (KLF4). This model has enabled us to dissect the molecular mechanisms of MFS, identify novel targets for treatment (such as p38 and KLF4) and provided an innovative human platform for the testing of new drugs.
Collapse
|
23
|
Lee L, Cui JZ, Cua M, Esfandiarei M, Sheng X, Chui WA, Xu MH, Sarunic MV, Beg MF, van Breemen C, Sandor GGS, Tibbits GF. Aortic and Cardiac Structure and Function Using High-Resolution Echocardiography and Optical Coherence Tomography in a Mouse Model of Marfan Syndrome. PLoS One 2016; 11:e0164778. [PMID: 27824871 PMCID: PMC5100915 DOI: 10.1371/journal.pone.0164778] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2015] [Accepted: 10/02/2016] [Indexed: 12/23/2022] Open
Abstract
Marfan syndrome (MFS) is an autosomal-dominant disorder of connective tissue caused by mutations in the fibrillin-1 (FBN1) gene. Mortality is often due to aortic dissection and rupture. We investigated the structural and functional properties of the heart and aorta in a [Fbn1C1039G/+] MFS mouse using high-resolution ultrasound (echo) and optical coherence tomography (OCT). Echo was performed on 6- and 12-month old wild type (WT) and MFS mice (n = 8). In vivo pulse wave velocity (PWV), aortic root diameter, ejection fraction, stroke volume, left ventricular (LV) wall thickness, LV mass and mitral valve early and atrial velocities (E/A) ratio were measured by high resolution echocardiography. OCT was performed on 12-month old WT and MFS fixed mouse hearts to measure ventricular volume and mass. The PWV was significantly increased in 6-mo MFS vs. WT (366.6 ± 19.9 vs. 205.2 ± 18.1 cm/s; p = 0.003) and 12-mo MFS vs. WT (459.5 ± 42.3 vs. 205.3 ± 30.3 cm/s; p< 0.0001). PWV increased with age in MFS mice only. We also found a significantly enlarged aortic root and decreased E/A ratio in MFS mice compared with WT for both age groups. The [Fbn1C1039G/+] mouse model of MFS replicates many of the anomalies of Marfan patients including significant aortic dilation, central aortic stiffness, LV systolic and diastolic dysfunction. This is the first demonstration of the direct measurement in vivo of pulse wave velocity non-invasively in the aortic arch of MFS mice, a robust measure of aortic stiffness and a critical clinical parameter for the assessment of pathology in the Marfan syndrome.
Collapse
Affiliation(s)
- Ling Lee
- Child and Family Research Institute, Department of Cardiovascular Sciences, Vancouver, BC, Canada
- Department of Biomedical Physiology and Kinesiology, Simon Fraser University, Burnaby, BC, Canada
| | - Jason Z. Cui
- Child and Family Research Institute, Department of Cardiovascular Sciences, Vancouver, BC, Canada
- Division of Cardiology, Department of Pediatrics, UBC, Vancouver, BC, Canada
| | - Michelle Cua
- School of Engineering Science, Simon Fraser University, Burnaby, BC, Canada
| | - Mitra Esfandiarei
- Child and Family Research Institute, Department of Cardiovascular Sciences, Vancouver, BC, Canada
- Anesthesiology, Pharmacology and Therapeutics, Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Xiaoye Sheng
- Child and Family Research Institute, Department of Cardiovascular Sciences, Vancouver, BC, Canada
- Department of Biomedical Physiology and Kinesiology, Simon Fraser University, Burnaby, BC, Canada
| | - Winsey Audrey Chui
- School of Engineering Science, Simon Fraser University, Burnaby, BC, Canada
| | - Michael Haoying Xu
- Child and Family Research Institute, Department of Cardiovascular Sciences, Vancouver, BC, Canada
| | - Marinko V. Sarunic
- School of Engineering Science, Simon Fraser University, Burnaby, BC, Canada
| | - Mirza Faisal Beg
- School of Engineering Science, Simon Fraser University, Burnaby, BC, Canada
| | - Cornelius van Breemen
- Child and Family Research Institute, Department of Cardiovascular Sciences, Vancouver, BC, Canada
- Anesthesiology, Pharmacology and Therapeutics, Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada
| | - George G. S. Sandor
- Child and Family Research Institute, Department of Cardiovascular Sciences, Vancouver, BC, Canada
- Division of Cardiology, Department of Pediatrics, UBC, Vancouver, BC, Canada
| | - Glen F. Tibbits
- Child and Family Research Institute, Department of Cardiovascular Sciences, Vancouver, BC, Canada
- Department of Biomedical Physiology and Kinesiology, Simon Fraser University, Burnaby, BC, Canada
- * E-mail:
| |
Collapse
|
24
|
Lee JJ, Galatioto J, Rao S, Ramirez F, Costa KD. Losartan Attenuates Degradation of Aorta and Lung Tissue Micromechanics in a Mouse Model of Severe Marfan Syndrome. Ann Biomed Eng 2016; 44:2994-3006. [PMID: 27090893 PMCID: PMC5880286 DOI: 10.1007/s10439-016-1616-4] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2015] [Accepted: 04/07/2016] [Indexed: 01/24/2023]
Abstract
Marfan syndrome (MFS) is an autosomal dominant disease of the connective tissue due to mutations in the fibrillin-1 gene (FBN1). This study aimed at characterizing microelastic properties of the ascending aortic wall and lung parenchyma tissues from wild type (WT) and age-matched Fbn1 hypomorphic mice (Fbn1(mgR/mgR) mice) to identify tissue-specific biomechanical effects of aging and disease in MFS. Atomic force microscopy was used to indent lung parenchyma and aortic wall tissues, using Hybrid Eshelby Decomposition analysis to extract layer-specific properties of the intima and media. The intima stiffened with age and was not different between WT and Fbn1(mgR/mgR) tissues, whereas the media layer of MFS aortas showed progressive structural and mechanical degradation with a modulus that was 50% softer than WT by 3.5 months of age. Similarly, MFS mice displayed progressive structural and mechanical deterioration of lung tissue, which was over 85% softer than WT by 3.5 months of age. Chronic treatment with the angiotensin type I receptor antagonist, losartan, attenuated the aorta and lung tissue degradation, resulting in structural and mechanical properties not significantly different from age-matched WT controls. By revealing micromechanical softening of elastin-rich aorta and lung tissues with disease progression in fibrillin-1 deficient mice, our findings support the use of losartan as a prophylactic treatment that may abrogate the life-threatening symptoms of MFS.
Collapse
Affiliation(s)
- Jia-Jye Lee
- Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY, 10029, USA
- Department of Biomedical Engineering, The City College of New York, 160 Convent Ave, New York, NY, 10031, USA
| | - Josephine Galatioto
- Department of Pharmacology and Systems Therapeutics, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY, 10029, USA
| | - Satish Rao
- Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY, 10029, USA
| | - Francesco Ramirez
- Department of Pharmacology and Systems Therapeutics, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY, 10029, USA
| | - Kevin D Costa
- Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY, 10029, USA.
| |
Collapse
|
25
|
Leloup AJA, Van Hove CE, Kurdi A, De Moudt S, Martinet W, De Meyer GRY, Schrijvers DM, De Keulenaer GW, Fransen P. A novel set-up for the ex vivo analysis of mechanical properties of mouse aortic segments stretched at physiological pressure and frequency. J Physiol 2016; 594:6105-6115. [PMID: 27256450 DOI: 10.1113/jp272623] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2016] [Accepted: 05/31/2016] [Indexed: 01/20/2023] Open
Abstract
KEY POINTS Cyclic stretch is known to alter intracellular pathways involved in vessel tone regulation. We developed a novel set-up that allows straightforward characterization of the biomechanical properties of the mouse aorta while stretched at a physiological heart rate (600 beats min-1 ). Active vessel tone was shown to have surprisingly large effects on isobaric stiffness. The effect of structural vessel wall alterations was confirmed using a genetic mouse model. This set-up will contribute to a better understanding of how active vessel wall components and mechanical stimuli such as stretch frequency and amplitude regulate aortic mechanics. ABSTRACT Cyclic stretch is a major contributor to vascular function. However, isolated mouse aortas are frequently studied at low stretch frequency or even in isometric conditions. Pacing experiments in rodents and humans show that arterial compliance is stretch frequency dependent. The Rodent Oscillatory Tension Set-up to study Arterial Compliance is an in-house developed organ bath set-up that clamps aortic segments to imposed preloads at physiological rates up to 600 beats min-1 . The technique enables us to derive pressure-diameter loops and assess biomechanical properties of the segment. To validate the applicability of this set-up we aimed to confirm the effects of distension pressure and vascular smooth muscle tone on arterial stiffness. At physiological stretch frequency (10 Hz), the Peterson modulus (EP ; 293 (10) mmHg) for wild-type mouse aorta increased 22% upon a rise in pressure from 80-120 mmHg to 100-140 mmHg, while, at normal pressure, EP increased 80% upon maximal contraction of the vascular smooth muscle cells. We further validated the method using a mouse model with a mutation in the fibrillin-1 gene and an endothelial nitric oxide synthase knock-out model. Both models are known to have increased arterial stiffness, and this was confirmed using the set-up. To our knowledge, this is the first set-up that facilitates the study of biomechanical properties of mouse aortic segments at physiological stretch frequency and pressure. We believe that this set-up can contribute to a better understanding of how cyclic stretch frequency, amplitude and active vessel wall components influence arterial stiffening.
Collapse
Affiliation(s)
- Arthur J A Leloup
- University of Antwerp, Department of Pharmaceutical Sciences, Laboratory of Physiopharmacology, Campus Drie Eiken, Universiteitsplein 1, B-2610, Antwerp, Belgium.
| | - Cor E Van Hove
- University of Antwerp, Faculty of Medicine and Health Sciences, Laboratory of Pharmacology, Campus Drie Eiken, Universiteitsplein 1, B-2610, Antwerp, Belgium
| | - Ammar Kurdi
- University of Antwerp, Department of Pharmaceutical Sciences, Laboratory of Physiopharmacology, Campus Drie Eiken, Universiteitsplein 1, B-2610, Antwerp, Belgium
| | - Sofie De Moudt
- University of Antwerp, Department of Pharmaceutical Sciences, Laboratory of Physiopharmacology, Campus Drie Eiken, Universiteitsplein 1, B-2610, Antwerp, Belgium
| | - Wim Martinet
- University of Antwerp, Department of Pharmaceutical Sciences, Laboratory of Physiopharmacology, Campus Drie Eiken, Universiteitsplein 1, B-2610, Antwerp, Belgium
| | - Guido R Y De Meyer
- University of Antwerp, Department of Pharmaceutical Sciences, Laboratory of Physiopharmacology, Campus Drie Eiken, Universiteitsplein 1, B-2610, Antwerp, Belgium
| | - Dorien M Schrijvers
- University of Antwerp, Department of Pharmaceutical Sciences, Laboratory of Physiopharmacology, Campus Drie Eiken, Universiteitsplein 1, B-2610, Antwerp, Belgium
| | - Gilles W De Keulenaer
- University of Antwerp, Department of Pharmaceutical Sciences, Laboratory of Physiopharmacology, Campus Drie Eiken, Universiteitsplein 1, B-2610, Antwerp, Belgium
| | - Paul Fransen
- University of Antwerp, Department of Pharmaceutical Sciences, Laboratory of Physiopharmacology, Campus Drie Eiken, Universiteitsplein 1, B-2610, Antwerp, Belgium
| |
Collapse
|
26
|
Infusion of Hibiscus sabdariffa L. Modulates Oxidative Stress in Patients with Marfan Syndrome. Mediators Inflamm 2016; 2016:8625203. [PMID: 27413258 PMCID: PMC4927999 DOI: 10.1155/2016/8625203] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2016] [Revised: 05/12/2016] [Accepted: 05/26/2016] [Indexed: 01/16/2023] Open
Abstract
Marfan syndrome (MFS) is associated with progressive aortic dilatation, endothelial dysfunction, and oxidative stress that contribute to the early acute dissection of the vessel and can end up in rupture of the aorta and sudden death. Many studies have described that the organic acids from Hibiscus sabdariffa Linne (HSL) calyces increase cellular antioxidant capacity and decrease oxidative stress. Here we evaluate if the antioxidant properties of HSL infusion improve oxidative stress in MFS patients. Activities of extra cellular super oxide dismutase (ECSOD), glutathione peroxidase (GPx), glutathione-S-transferase (GST), glutathione reductase (GSSG-R), glutathione (GSH), lipid peroxidation (LPO) index, total antioxidant capacity (TAC), and ascorbic acid were determined in plasma from MFS patients. Values before and after 3 months of the treatment with 2% HSL infusion were compared in control and MFS subjects. After treatment, there was a significant decrease in ECSOD (p = 0.03), EGPx (p = 0.04), GST (p = 0.03), GSH (p = 0.01), and TAC and ascorbic acid (p = 0.02) but GSSG-R activity (p = 0.04) and LPO (p = 0.02) were increased in MFS patients in comparison to patients receiving the HSL treatment and C subjects. Therefore, the infusion of HSL calyces has antioxidant properties that allow an increase in antioxidant capacity of both the enzymatic and nonenzymatic systems, in the plasma of the MSF patients.
Collapse
|
27
|
Soto ME, Iturriaga Hernández AV, Guarner Lans V, Zuñiga-Muñoz A, Aranda Fraustro A, Velázquez Espejel R, Pérez-Torres I. Participation of oleic acid in the formation of the aortic aneurysm in Marfan syndrome patients. Prostaglandins Other Lipid Mediat 2016; 123:46-55. [PMID: 27163200 DOI: 10.1016/j.prostaglandins.2016.05.001] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2016] [Revised: 05/04/2016] [Accepted: 05/05/2016] [Indexed: 02/07/2023]
Abstract
Marfan syndrome (MFS) is associated with progressive aortic dilatation and endothelial dysfunction that lead to early acute dissection and rupture of the aorta and sudden death. Alteration in fatty acid (FA) metabolism can stimulate nitric oxide (NO) overproduction which increases the activity of the inducible form of NO synthase (iNOS) that is involved in endothelial dysfunction. We evaluated the participation of FA in the formation of thoracic aneurysms in MFS and its relation to the iNOS. Oleic acid (OA), iNOS, citrulline, nitrates and nitrites, TGF-β1, TNF-α, monounsaturated FA and NO synthase activity were significantly increased (p<0.05) in tissue from the aortas of MFS. Saturated FA, eNOS and HDL were significantly decreased (p<0.05). Arachidonic acid, delta-9 desaturase tended to increase and histological examination showed an increase in cystic necrosis, elastic fibers and collagen in MFS. The increase in OA contributes to the altered pathway of iNOS, which favors endothelial dysfunction and formation of the aortic aneurysms in MFS.
Collapse
Affiliation(s)
- María Elena Soto
- Department of Immunology, Instituto Nacional de Cardiología "Ignacio Chávez", Juan Badiano 1, Sección XVI, Tlalpan, 14080 México City, DF, Mexico
| | - Alejandra Valeria Iturriaga Hernández
- Department of Cardiothoracic Surgery, Instituto Nacional de Cardiología "Ignacio Chávez", Juan Badiano 1, Sección XVI, Tlalpan, 14080 México City, DF, Mexico
| | - Verónica Guarner Lans
- Department of Physiology, Instituto Nacional de Cardiología "Ignacio Chávez", Juan Badiano 1, Sección XVI, Tlalpan, 14080 México City, DF, Mexico
| | - Alejandra Zuñiga-Muñoz
- Department of Pathology, Instituto Nacional de Cardiología "Ignacio Chávez", Juan Badiano 1, Sección XVI, Tlalpan, 14080 México City, DF, Mexico
| | - Alberto Aranda Fraustro
- Department of Pathology, Instituto Nacional de Cardiología "Ignacio Chávez", Juan Badiano 1, Sección XVI, Tlalpan, 14080 México City, DF, Mexico
| | - Rodrigo Velázquez Espejel
- Department of Pathology, Instituto Nacional de Cardiología "Ignacio Chávez", Juan Badiano 1, Sección XVI, Tlalpan, 14080 México City, DF, Mexico
| | - Israel Pérez-Torres
- Department of Pathology, Instituto Nacional de Cardiología "Ignacio Chávez", Juan Badiano 1, Sección XVI, Tlalpan, 14080 México City, DF, Mexico.
| |
Collapse
|
28
|
Onetti Y, Meirelles T, Dantas AP, Schröder K, Vila E, Egea G, Jiménez-Altayó F. NADPH oxidase 4 attenuates cerebral artery changes during the progression of Marfan syndrome. Am J Physiol Heart Circ Physiol 2016; 310:H1081-90. [PMID: 26945079 DOI: 10.1152/ajpheart.00770.2015] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/06/2015] [Accepted: 03/01/2016] [Indexed: 12/11/2022]
Abstract
Marfan syndrome (MFS) is a connective tissue disorder that is often associated with the fibrillin-1 (Fbn1) gene mutation and characterized by cardiovascular alterations, predominantly ascending aortic aneurysms. Although neurovascular complications are uncommon in MFS, the improvement in Marfan patients' life expectancy is revealing other secondary alterations, potentially including neurovascular disorders. However, little is known about small-vessel pathophysiology in MFS. MFS is associated with hyperactivated transforming growth factor (TGF)-β signaling, which among numerous other downstream effectors, induces the NADPH oxidase 4 (Nox4) isoform of NADPH oxidase, a strong enzymatic source of H2O2 We hypothesized that MFS induces middle cerebral artery (MCA) alterations and that Nox4 contributes to them. MCA properties from 3-, 6-, or 9-mo-old Marfan (Fbn1(C1039G/+)) mice were compared with those from age/sex-matched wild-type littermates. At 6 mo, Marfan compared with wild-type mice developed higher MCA wall/lumen (wild-type: 0.081 ± 0.004; Marfan: 0.093 ± 0.002; 60 mmHg; P < 0.05), coupled with increased reactive oxygen species production, TGF-β, and Nox4 expression. However, wall stiffness and myogenic autoregulation did not change. To investigate the influence of Nox4 on cerebrovascular properties, we generated Marfan mice with Nox4 deficiency (Nox4(-/-)). Strikingly, Nox4 deletion in Marfan mice aggravated MCA wall thickening (cross-sectional area; Marfan: 6,660 ± 363 μm(2); Marfan Nox4(-/-): 8,795 ± 824 μm(2); 60 mmHg; P < 0.05), accompanied by decreased TGF-β expression and increased collagen deposition and Nox1 expression. These findings provide the first evidence that Nox4 mitigates cerebral artery structural changes in a murine model of MFS.
Collapse
Affiliation(s)
- Yara Onetti
- Departament de Farmacologia, de Terapèutica i de Toxicologia, Institut de Neurociències, Facultat de Medicina, Universitat Autònoma de Barcelona, Bellaterra, Spain
| | - Thayna Meirelles
- Departament de Biologia Cellular, Immunologia i Neurociències, Facultat de Medicina, Universitat de Barcelona, Barcelona, Spain
| | - Ana P Dantas
- Institut Clínic del Tòrax, Institut d'Investigacions Biomèdiques August Pi i Sunyer, Barcelona, Spain; and
| | - Katrin Schröder
- Institute for Cardiovascular Physiology, Goethe-University Frankfurt, Frankfurt, Germany
| | - Elisabet Vila
- Departament de Farmacologia, de Terapèutica i de Toxicologia, Institut de Neurociències, Facultat de Medicina, Universitat Autònoma de Barcelona, Bellaterra, Spain
| | - Gustavo Egea
- Departament de Biologia Cellular, Immunologia i Neurociències, Facultat de Medicina, Universitat de Barcelona, Barcelona, Spain; Institut Clínic del Tòrax, Institut d'Investigacions Biomèdiques August Pi i Sunyer, Barcelona, Spain; and
| | - Francesc Jiménez-Altayó
- Departament de Farmacologia, de Terapèutica i de Toxicologia, Institut de Neurociències, Facultat de Medicina, Universitat Autònoma de Barcelona, Bellaterra, Spain;
| |
Collapse
|
29
|
Akazawa Y, Inaba Y, Hachiya A, Motoki N, Matsuzaki S, Minatoya K, Morisaki T, Morisaki H, Kosaki K, Kosho T, Koike K. Reversible cerebral vasoconstriction syndrome and posterior reversible encephalopathy syndrome in a boy with Loeys-Dietz syndrome. Am J Med Genet A 2015; 167A:2435-9. [DOI: 10.1002/ajmg.a.37202] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2015] [Accepted: 05/25/2015] [Indexed: 11/09/2022]
Affiliation(s)
- Yohei Akazawa
- Department of Pediatrics; Shinshu University School of Medicine; Matsumoto Japan
| | - Yuji Inaba
- Department of Pediatrics; Shinshu University School of Medicine; Matsumoto Japan
| | - Akira Hachiya
- Department of Pediatrics; Shinshu University School of Medicine; Matsumoto Japan
| | - Noriko Motoki
- Department of Pediatrics; Shinshu University School of Medicine; Matsumoto Japan
| | - Satoshi Matsuzaki
- Department of Pediatrics; Shinshu University School of Medicine; Matsumoto Japan
| | - Kenji Minatoya
- Department of Vascular Surgery; National Cerebral and Cardiovascular Center; Suita Japan
| | - Takayuki Morisaki
- Department of Bioscience and Genetics; National Cerebral and Cardiovascular Center Research Institute; Suita Japan
| | - Hiroko Morisaki
- Department of Bioscience and Genetics; National Cerebral and Cardiovascular Center Research Institute; Suita Japan
| | - Kenjiro Kosaki
- Department of Pediatrics; Keio University School of Medicine; Tokyo Japan
| | - Tomoki Kosho
- Department Medical Genetics; Shinshu University School of Medicine; Matsumoto Japan
| | - Kenichi Koike
- Department of Pediatrics; Shinshu University School of Medicine; Matsumoto Japan
| |
Collapse
|
30
|
Lilly B. We have contact: endothelial cell-smooth muscle cell interactions. Physiology (Bethesda) 2015; 29:234-41. [PMID: 24985327 DOI: 10.1152/physiol.00047.2013] [Citation(s) in RCA: 82] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Blood vessels are composed of two primary cell types, endothelial cells and smooth muscle cells, each providing a unique contribution to vessel function. Signaling between these two cell types is essential for maintaining tone in mature vessels, and their communication is critical during development, and for repair and remodeling associated with blood vessel growth. This review will highlight the pathways that endothelial cells and smooth muscle cells utilize to communicate during vessel formation and discuss how disruptions in these pathways contribute to disease.
Collapse
Affiliation(s)
- Brenda Lilly
- Department of Pediatrics, Nationwide Children's Hospital, The Heart Center, The Ohio State University, Columbus, Ohio
| |
Collapse
|
31
|
Vascular dysfunctions in the isolated aorta of double-transgenic hypertensive mice developing aortic aneurysm. Pflugers Arch 2014; 467:1945-63. [PMID: 25385304 DOI: 10.1007/s00424-014-1644-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2014] [Revised: 10/29/2014] [Accepted: 10/31/2014] [Indexed: 01/09/2023]
Abstract
Angiotensin-II and oxidative stress are involved in the genesis of aortic aneurysms, a phenomenon exacerbated by endothelial nitric oxide synthase (eNOS) deletion or uncoupling. The purpose of this work was to study the endothelial function in wild-type C57BL/6 (BL) and transgenic mice expressing the h-angiotensinogen and h-renin genes (AR) subjected to either a control, or a high-salt diet plus a treatment with a NO-synthase inhibitor, N-ω-nitro-L-arginine-methyl-ester (L-NAME; BLSL and ARSL). BLSL showed a moderate increase in blood pressure, while ARSL became severely hypertensive. Seventy-five percent of ARSL developed aortic aneurysms, characterized by major histo-morphological changes and associated with an increase in NADP(H) oxidase-2 (NOX2) expression. Contractile responses (KCl, norepinephrine, U-46619) were similar in the four groups of mice, and relaxations were not affected in BLSL and AR. However, in ARSL, endothelium-dependent relaxations (acetylcholine, UK-14304) were significantly reduced, and this dysfunction was similar in aortae without or with aneurysms. The endothelial impairment was unaffected by catalase, superoxide-dismutase mimetic, radical scavengers, cyclooxygenase inhibition, or TP-receptor blockade and could not be attributed to sGC oxidation. Thus, ARSL is a severe hypertension model developing aortic aneurysm. A vascular dysfunction, involving both endothelial (reduced role of NO) and smooth muscle cells, precedes aneurysms formation and, paradoxically, does not appear to involve oxidative stress.
Collapse
|
32
|
Soto ME, Soria-Castro E, Lans VG, Ontiveros EM, Mejía BIH, Hernandez HJM, García RB, Herrera V, Pérez-Torres I. Analysis of oxidative stress enzymes and structural and functional proteins on human aortic tissue from different aortopathies. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2014; 2014:760694. [PMID: 25101153 PMCID: PMC4102031 DOI: 10.1155/2014/760694] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/04/2014] [Revised: 05/28/2014] [Accepted: 05/28/2014] [Indexed: 01/15/2023]
Abstract
The role of oxidative stress in different aortopathies is evaluated. Thirty-two tissue samples from 18 men and 14 women were divided into: 4 control (C) subjects, 11 patients with systemic arterial hypertension (SAH), 4 with variants of Marfan's syndrome (MV), 9 with Marfan's syndrome (M), 2 with Turner's syndrome, and 2 with Takayasu's arteritis (TA). Aorta fragments were homogenized. Lipoperoxidation (LPO), copper-zinc and manganese superoxide dismutase (Mn and Cu-Zn-SOD), catalase (CAT), glutathione peroxidase (GPx), glutathione S-transferase (GST), endothelial nitric oxide synthase (eNOS), nitrates and nitrites (NO3(-)/NO2(-)), and type IV collagen, and laminin were evaluated. There was an increase in Mn- and Cu-Zn-SOD activity in SAH, MV, M, and Turner's syndrome. There was also an increase in CAT activity in M and Turner' syndrome. GPx and GST activity decreased and LPO increased in all groups. eNOS was decreased in SAH, MV, and M and NO3 (-)/NO2 (-) were increased in SAH and TA. Type IV collagen was decreased in Turner's syndrome and TA. Laminin γ-1 was decreased in MV and increased in M. In conclusion, similarities and differences in oxidative stress in the different aortopathies studied including pathologies with aneurysms were found with alterations in SOD, CAT, GPx, GST, and eNOS activity that modify subendothelial basement membrane proteins.
Collapse
Affiliation(s)
- María Elena Soto
- Immunology Department, National Institute of Cardiology "Ignacio Chavez", Juan Badiano 1, Sección XVI, Tlalpan, 14080 Mexico City, DF, Mexico
| | - Elizabeth Soria-Castro
- Pathology Department, National Institute of Cardiology "Ignacio Chavez", Juan Badiano 1, Sección XVI, Tlalpan, 14080 Mexico City, DF, Mexico
| | - Verónica Guarner Lans
- Physiology Department, National Institute of Cardiology "Ignacio Chavez", Juan Badiano 1, Sección XVI, Tlalpan, 14080 Mexico City, DF, Mexico
| | - Eleazar Muruato Ontiveros
- Cardiovascular Surgery Department, National Institute of Cardiology "Ignacio Chavez", Juan Badiano 1, Sección XVI, Tlalpan, 14080 Mexico City, DF, Mexico
| | - Benjamín Iván Hernández Mejía
- Cardiovascular Surgery Department, National Institute of Cardiology "Ignacio Chavez", Juan Badiano 1, Sección XVI, Tlalpan, 14080 Mexico City, DF, Mexico
| | - Humberto Jorge Martínez Hernandez
- Cardiovascular Surgery Department, National Institute of Cardiology "Ignacio Chavez", Juan Badiano 1, Sección XVI, Tlalpan, 14080 Mexico City, DF, Mexico
| | - Rodolfo Barragán García
- Cardiovascular Surgery Department, National Institute of Cardiology "Ignacio Chavez", Juan Badiano 1, Sección XVI, Tlalpan, 14080 Mexico City, DF, Mexico
| | - Valentín Herrera
- Cardiovascular Surgery Department, National Institute of Cardiology "Ignacio Chavez", Juan Badiano 1, Sección XVI, Tlalpan, 14080 Mexico City, DF, Mexico
| | - Israel Pérez-Torres
- Pathology Department, National Institute of Cardiology "Ignacio Chavez", Juan Badiano 1, Sección XVI, Tlalpan, 14080 Mexico City, DF, Mexico
| |
Collapse
|
33
|
Jiménez-Altayó F, Onetti Y, Heras M, Dantas AP, Vila E. Western-style diet modulates contractile responses to phenylephrine differently in mesenteric arteries from senescence-accelerated prone (SAMP8) and resistant (SAMR1) mice. AGE (DORDRECHT, NETHERLANDS) 2013; 35:1219-1234. [PMID: 22777652 PMCID: PMC3705122 DOI: 10.1007/s11357-012-9450-6] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/14/2012] [Accepted: 06/24/2012] [Indexed: 06/01/2023]
Abstract
The influence of two known cardiovascular risk factors, aging and consumption of a high-fat diet, on vascular mesenteric artery reactivity was examined in a mouse model of accelerated senescence (SAM). Five-month-old SAM prone (SAMP8) and resistant (SAMR1) female mice were fed a Western-type high-fat diet (WD; 8 weeks). Mesenteric arteries were dissected, and vascular reactivity, protein and messenger RNA expression, superoxide anion (O 2 (·-) ) and hydrogen peroxide formation were evaluated by wire myography, immunofluorescence, RT-qPCR, ethidium fluorescence and ferric-xylenol orange, respectively. Contraction to KCl and relaxation to acetylcholine remained unchanged irrespective of senescence and diet. Although similar contractions to phenylephrine were observed in SAMR1 and SAMP8, accelerated senescence was associated with decreased eNOS and nNOS and increased O 2 (·-) synthesis. Senescence-related alterations were compensated, at least partly, by the contribution of NO derived from iNOS and the enhanced endogenous antioxidant capacity of superoxide dismutase 1 to maintain vasoconstriction. Administration of a WD induced qualitatively different alterations in phenylephrine contractions of mesenteric arteries from SAMR1 and SAMP8. SAMR1 showed increased contractions partly as a result of decreased NO availability generated by decreased eNOS and nNOS and enhanced O 2 (·-) formation. In contrast, WD feeding in SAMP8 resulted in reduced contractions due to, at least in part, the increased functional participation of iNOS-derived NO. In conclusion, senescence-dependent intrinsic alterations during early stages of vascular senescence may promote vascular adaptation and predispose to further changes in response to high-fat intake, which may lead to the progression of aging-related cardiovascular disease, whereas young subjects lack the capacity for this adaptation.
Collapse
Affiliation(s)
- Francesc Jiménez-Altayó
- Departament de Farmacologia, Terapèutica i Toxicologia, Institut de Neurociències, Facultat de Medicina, Universitat Autònoma de Barcelona (UAB), 08193, Bellaterra, Cerdanyola del Vallès, Spain.
| | | | | | | | | |
Collapse
|
34
|
Cook JR, Nistala H, Ramirez F. Drug-based therapies for vascular disease in Marfan syndrome: from mouse models to human patients. ACTA ACUST UNITED AC 2011; 77:366-73. [PMID: 20687182 DOI: 10.1002/msj.20200] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Marfan syndrome is a congenital disorder of the connective tissue with a long history of clinical and basic science breakthroughs that have forged our understanding of vascular-disease pathogenesis. The biomedical importance of Marfan syndrome was recently underscored by the discovery that the underlying genetic lesion impairs both tissue integrity and transforming growth factor-beta regulation of cell behavior. This discovery has led to the successful implementation of the first pharmacological intervention in a connective-tissue disorder otherwise incurable by either gene-based or stem cell-based therapeutic strategies. More generally, information gathered from the study of Marfan syndrome pathogenesis has the potential to improve the clinical management of common acquired disorders of connective-tissue degeneration.
Collapse
Affiliation(s)
- Jason R Cook
- Mount Sinai School of Medicine, New York, NY, USA
| | | | | |
Collapse
|