1
|
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
|
2
|
Abdalla N, Ortiz-Romero P, Rodriguez-Rovira I, Pérez-Jurado LA, Egea G, Campuzano V. The Combined Treatment of Curcumin with Verapamil Ameliorates the Cardiovascular Pathology in a Williams-Beuren Syndrome Mouse Model. Int J Mol Sci 2023; 24:ijms24043261. [PMID: 36834670 PMCID: PMC9961051 DOI: 10.3390/ijms24043261] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Revised: 02/03/2023] [Accepted: 02/03/2023] [Indexed: 02/11/2023] Open
Abstract
Williams-Beuren syndrome (WBS) is a rare disorder caused by a recurrent microdeletion with hallmarks of cardiovascular manifestations, mainly supra-valvular aortic stenosis (SVAS). Unfortunately, there is currently no efficient treatment. We investigated the effect of chronic oral treatment with curcumin and verapamil on the cardiovascular phenotype of a murine model of WBS harbouring a similar deletion, CD (complete deletion) mice. We analysed systolic blood pressure in vivo and the histopathology of the ascending aorta and the left ventricular myocardium to determine the effects of treatments and their underlying mechanism. Molecular analysis showed significantly upregulated xanthine oxidoreductase (XOR) expression in the aorta and left ventricular myocardium of CD mice. This overexpression is concomitant with increased levels of nitrated proteins as a result of byproduct-mediated oxidative stress damage, indicating that XOR-generated oxidative stress impacts the pathophysiology of cardiovascular manifestations in WBS. Only the combined therapy of curcumin and verapamil resulted in a significant improvement of cardiovascular parameters via activation of the nuclear factor erythroid 2 (NRF2) and reduction of XOR and nitrated protein levels. Our data suggested that the inhibition of XOR and oxidative stress damage could help prevent the severe cardiovascular injuries of this disorder.
Collapse
Affiliation(s)
- Noura Abdalla
- Department of Biomedical Sciences, School of Medicine and Health Sciences, University of Barcelona, 08036 Barcelona, Spain
| | - Paula Ortiz-Romero
- Department of Biomedical Sciences, School of Medicine and Health Sciences, University of Barcelona, 08036 Barcelona, Spain
| | - Isaac Rodriguez-Rovira
- Department of Biomedical Sciences, School of Medicine and Health Sciences, University of Barcelona, 08036 Barcelona, Spain
| | - Luis A. Pérez-Jurado
- Department of Medicine and Life Sciences, University Pompeu Fabra, 08003 Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), ISCIII, 28029 Madrid, Spain
- Genetics Service, Hospital del Mar & Hospital del Mar Research Institute (IMIM), 08003 Barcelona, Spain
| | - Gustavo Egea
- Department of Biomedical Sciences, School of Medicine and Health Sciences, University of Barcelona, 08036 Barcelona, Spain
| | - Victoria Campuzano
- Department of Biomedical Sciences, School of Medicine and Health Sciences, University of Barcelona, 08036 Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), ISCIII, 28029 Madrid, Spain
- Correspondence:
| |
Collapse
|
3
|
Mieremet A, van der Stoel M, Li S, Coskun E, van Krimpen T, Huveneers S, de Waard V. Endothelial dysfunction in Marfan syndrome mice is restored by resveratrol. Sci Rep 2022; 12:22504. [PMID: 36577770 PMCID: PMC9797556 DOI: 10.1038/s41598-022-26662-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2022] [Accepted: 12/19/2022] [Indexed: 12/29/2022] Open
Abstract
Patients with Marfan syndrome (MFS) develop thoracic aortic aneurysms as the aorta presents excessive elastin breaks, fibrosis, and vascular smooth muscle cell (vSMC) death due to mutations in the FBN1 gene. Despite elaborate vSMC to aortic endothelial cell (EC) signaling, the contribution of ECs to the development of aortic pathology remains largely unresolved. The aim of this study is to investigate the EC properties in Fbn1C1041G/+ MFS mice. Using en face immunofluorescence confocal microscopy, we showed that EC alignment with blood flow was reduced, EC roundness was increased, individual EC surface area was larger, and EC junctional linearity was decreased in aortae of Fbn1C1041G/+ MFS mice. This modified EC phenotype was most prominent in the ascending aorta and occurred before aortic dilatation. To reverse EC morphology, we performed treatment with resveratrol. This restored EC blood flow alignment, junctional linearity, phospho-eNOS expression, and improved the structural integrity of the internal elastic lamina of Fbn1C1041G/+ mice. In conclusion, these experiments identify the involvement of ECs and underlying internal elastic lamina in MFS aortic pathology, which could act as potential target for future MFS pharmacotherapies.
Collapse
Affiliation(s)
- Arnout Mieremet
- Department of Medical Biochemistry, Amsterdam UMC Location University of Amsterdam, Meibergdreef 9, Amsterdam, The Netherlands
- Amsterdam Cardiovascular Sciences, Atherosclerosis and Ischemic Syndromes, Amsterdam, The Netherlands
| | - Miesje van der Stoel
- Department of Medical Biochemistry, Amsterdam UMC Location University of Amsterdam, Meibergdreef 9, Amsterdam, The Netherlands
- Amsterdam Cardiovascular Sciences, Atherosclerosis and Ischemic Syndromes, Amsterdam, The Netherlands
- Amsterdam Cardiovascular Sciences, Microcirculation, Amsterdam, The Netherlands
| | - Siyu Li
- Department of Medical Biochemistry, Amsterdam UMC Location University of Amsterdam, Meibergdreef 9, Amsterdam, The Netherlands
- Amsterdam Cardiovascular Sciences, Atherosclerosis and Ischemic Syndromes, Amsterdam, The Netherlands
| | - Evrim Coskun
- Department of Medical Biochemistry, Amsterdam UMC Location University of Amsterdam, Meibergdreef 9, Amsterdam, The Netherlands
- Amsterdam Cardiovascular Sciences, Atherosclerosis and Ischemic Syndromes, Amsterdam, The Netherlands
| | - Tsveta van Krimpen
- Department of Medical Biochemistry, Amsterdam UMC Location University of Amsterdam, Meibergdreef 9, Amsterdam, The Netherlands
- Amsterdam Cardiovascular Sciences, Atherosclerosis and Ischemic Syndromes, Amsterdam, The Netherlands
- Amsterdam Cardiovascular Sciences, Microcirculation, Amsterdam, The Netherlands
| | - Stephan Huveneers
- Department of Medical Biochemistry, Amsterdam UMC Location University of Amsterdam, Meibergdreef 9, Amsterdam, The Netherlands
- Amsterdam Cardiovascular Sciences, Atherosclerosis and Ischemic Syndromes, Amsterdam, The Netherlands
- Amsterdam Cardiovascular Sciences, Microcirculation, Amsterdam, The Netherlands
| | - Vivian de Waard
- Department of Medical Biochemistry, Amsterdam UMC Location University of Amsterdam, Meibergdreef 9, Amsterdam, The Netherlands.
- Amsterdam Cardiovascular Sciences, Atherosclerosis and Ischemic Syndromes, Amsterdam, The Netherlands.
| |
Collapse
|
4
|
Rodríguez-Rovira I, Arce C, De Rycke K, Pérez B, Carretero A, Arbonés M, Teixidò-Turà G, Gómez-Cabrera MC, Campuzano V, Jiménez-Altayó F, Egea G. Allopurinol blocks aortic aneurysm in a mouse model of Marfan syndrome via reducing aortic oxidative stress. Free Radic Biol Med 2022; 193:538-550. [PMID: 36347404 DOI: 10.1016/j.freeradbiomed.2022.11.001] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Revised: 10/28/2022] [Accepted: 11/02/2022] [Indexed: 11/06/2022]
Abstract
BACKGROUND Increasing evidence indicates that redox stress participates in MFS aortopathy, though its mechanistic contribution is little known. We reported elevated reactive oxygen species (ROS) formation and NADPH oxidase NOX4 upregulation in MFS patients and mouse aortae. Here we address the contribution of xanthine oxidoreductase (XOR), which catabolizes purines into uric acid and ROS in MFS aortopathy. METHODS AND RESULTS In aortic samples from MFS patients, XOR protein expression, revealed by immunohistochemistry, increased in both the tunicae intima and media of the dilated zone. In MFS mice (Fbn1C1041G/+), aortic XOR mRNA transcripts and enzymatic activity of the oxidase form (XO) were augmented in the aorta of 3-month-old mice but not in older animals. The administration of the XOR inhibitor allopurinol (ALO) halted the progression of aortic root aneurysm in MFS mice. ALO administrated before the onset of the aneurysm prevented its subsequent development. ALO also inhibited MFS-associated endothelial dysfunction as well as elastic fiber fragmentation, nuclear translocation of pNRF2 and increased 3'-nitrotyrosine levels, and collagen maturation remodeling, all occurring in the tunica media. ALO reduced the MFS-associated large aortic production of H2O2, and NOX4 and MMP2 transcriptional overexpression. CONCLUSIONS Allopurinol interferes in aortic aneurysm progression acting as a potent antioxidant. This study strengthens the concept that redox stress is an important determinant of aortic aneurysm formation and progression in MFS and warrants the evaluation of ALO therapy in MFS patients.
Collapse
Affiliation(s)
- Isaac Rodríguez-Rovira
- Department of Biomedical Sciences, University of Barcelona School of Medicine and Health Sciences, 08036, Barcelona, Spain
| | - Cristina Arce
- Department of Biomedical Sciences, University of Barcelona School of Medicine and Health Sciences, 08036, Barcelona, Spain
| | - Karo De Rycke
- Department of Biomedical Sciences, University of Barcelona School of Medicine and Health Sciences, 08036, Barcelona, Spain
| | - Belén Pérez
- Department of Pharmacology, Toxicology and Therapeutics, Neuroscience Institute, School of Medicine, Autonomous University of Barcelona, 08193, Cerdanyola del Vallès, Spain
| | - Aitor Carretero
- Department of Physiology, Faculty of Medicine, University of Valencia, CIBERFES, Fundación Investigación Hospital Clínico Universitario/INCLIVA, Valencia, Spain
| | - Marc Arbonés
- Department of Biomedical Sciences, University of Barcelona School of Medicine and Health Sciences, 08036, Barcelona, Spain
| | - Gisela Teixidò-Turà
- Department of Cardiology, Hospital Universitari Vall d'Hebron, Barcelona, Spain; CIBER-CV, Vall d'Hebrón Institut de Recerca (VHIR), Barcelona, Spain
| | - Mari Carmen Gómez-Cabrera
- Department of Physiology, Faculty of Medicine, University of Valencia, CIBERFES, Fundación Investigación Hospital Clínico Universitario/INCLIVA, Valencia, Spain
| | - Victoria Campuzano
- Department of Biomedical Sciences, University of Barcelona School of Medicine and Health Sciences, 08036, Barcelona, Spain; Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), ISCIII, Spain
| | - Francesc Jiménez-Altayó
- Department of Pharmacology, Toxicology and Therapeutics, Neuroscience Institute, School of Medicine, Autonomous University of Barcelona, 08193, Cerdanyola del Vallès, Spain
| | - Gustavo Egea
- Department of Biomedical Sciences, University of Barcelona School of Medicine and Health Sciences, 08036, Barcelona, Spain.
| |
Collapse
|
5
|
Hibender S, Li S, Postma AV, Hoogeland ME, Klaver D, Pouw RB, Niessen HW, Driessen AHG, Koolbergen DR, de Vries CJM, Baars MJH, Houweling AC, Krijnen PA, de Waard V. No prominent role for complement C1-esterase inhibitor in Marfan syndrome mice. VASCULAR BIOLOGY (BRISTOL, ENGLAND) 2022; 4:40-49. [PMID: 36279189 PMCID: PMC9782404 DOI: 10.1530/vb-22-0016] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Accepted: 10/24/2022] [Indexed: 12/05/2022]
Abstract
Marfan syndrome (MFS) is a connective tissue disorder causing aortic aneurysm formation. Currently, only prophylactic aortic surgery and blood pressure-lowering drugs are available to reduce the risk of aortic rupture. Upon whole genome sequencing of a Marfan family, we identified a complement gene C1R variant (p.Ser152Leu), which is associated with severe aortic patients. Therefore, we assessed the role of complement activation in MFS aortic tissue. Expression of various complement genes and proteins was detected in human and murine MFS aneurysm tissue, which prompted us to study complement inhibition in MFS mice. Treatment of the Fbn1C1041G/+ MFS mice with human plasma-derived C1-esterase inhibitor Cetor® resulted in reduced complement deposition, decreased macrophage influx in the aorta, and lower circulating TNFα levels. However, in line with previous anti-inflammatory treatments, complement inhibition did not change the aortic dilatation rate in this MFS mouse model. Thus, while complement factors/component 3 activation were detected in human/murine MFS aorta, Cetor® had no effect on aortic dilatation in MFS mice, indicating that complement inhibition is not a suitable treatment strategy in MFS.
Collapse
Affiliation(s)
- Stijntje Hibender
- Amsterdam UMC Location University of Amsterdam, Department of Medical Biochemistry, Meibergdreef, Amsterdam, The Netherlands
- Amsterdam Cardiovascular Sciences, Atherosclerosis & Ischemic Syndromes, Amsterdam, The Netherlands
| | - Siyu Li
- Amsterdam UMC Location University of Amsterdam, Department of Medical Biochemistry, Meibergdreef, Amsterdam, The Netherlands
- Amsterdam Cardiovascular Sciences, Atherosclerosis & Ischemic Syndromes, Amsterdam, The Netherlands
| | - Alex V Postma
- Amsterdam Cardiovascular Sciences, Atherosclerosis & Ischemic Syndromes, Amsterdam, The Netherlands
- Amsterdam UMC Location University of Amsterdam, Department of Medical Biology, Meibergdreef, Amsterdam, The Netherlands
- Amsterdam UMC Location University of Amsterdam, Department of Human Genetics, Meibergdreef, Amsterdam, The Netherlands
| | - Myrthe E Hoogeland
- Amsterdam UMC Location University of Amsterdam, Department of Medical Biochemistry, Meibergdreef, Amsterdam, The Netherlands
| | - Denise Klaver
- Amsterdam UMC Location University of Amsterdam, Department of Medical Biochemistry, Meibergdreef, Amsterdam, The Netherlands
| | - Richard B Pouw
- Sanquin Research, Department of Immunopathology, Plesmanlaan, Amsterdam, The Netherlands
- Amsterdam UMC Location University of Amsterdam, Landsteiner Laboratory, Meibergdreef, Amsterdam, The Netherlands
| | - Hans W Niessen
- Amsterdam Cardiovascular Sciences, Atherosclerosis & Ischemic Syndromes, Amsterdam, The Netherlands
- Amsterdam UMC Location University of Amsterdam, Department of Pathology, Meibergdreef, Amsterdam, The Netherlands
| | - Antoine HG Driessen
- Amsterdam UMC Location University of Amsterdam, Heart Center, Department of Cardiothoracic Surgery, Meibergdreef, Amsterdam, The Netherlands
| | - David R Koolbergen
- Amsterdam UMC Location University of Amsterdam, Heart Center, Department of Cardiothoracic Surgery, Meibergdreef, Amsterdam, The Netherlands
| | - Carlie JM de Vries
- Amsterdam UMC Location University of Amsterdam, Department of Medical Biochemistry, Meibergdreef, Amsterdam, The Netherlands
- Amsterdam Cardiovascular Sciences, Atherosclerosis & Ischemic Syndromes, Amsterdam, The Netherlands
| | - Marieke JH Baars
- Amsterdam UMC Location University of Amsterdam, Department of Human Genetics, Meibergdreef, Amsterdam, The Netherlands
| | - Arjan C Houweling
- Amsterdam Cardiovascular Sciences, Atherosclerosis & Ischemic Syndromes, Amsterdam, The Netherlands
- Amsterdam UMC Location University of Amsterdam, Department of Human Genetics, Meibergdreef, Amsterdam, The Netherlands
| | - Paul A Krijnen
- Amsterdam Cardiovascular Sciences, Atherosclerosis & Ischemic Syndromes, Amsterdam, The Netherlands
- Amsterdam UMC Location University of Amsterdam, Department of Pathology, Meibergdreef, Amsterdam, The Netherlands
| | - Vivian de Waard
- Amsterdam UMC Location University of Amsterdam, Department of Medical Biochemistry, Meibergdreef, Amsterdam, The Netherlands
- Amsterdam Cardiovascular Sciences, Atherosclerosis & Ischemic Syndromes, Amsterdam, The Netherlands
| |
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
|
Bollati M, Lopez C, Bioletto F, Ponzetto F, Ghigo E, Maccario M, Parasiliti-Caprino M. Atrial Fibrillation and Aortic Ectasia as Complications of Primary Aldosteronism: Focus on Pathophysiological Aspects. Int J Mol Sci 2022; 23:2111. [PMID: 35216224 PMCID: PMC8875197 DOI: 10.3390/ijms23042111] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Revised: 02/07/2022] [Accepted: 02/12/2022] [Indexed: 11/22/2022] Open
Abstract
Primary aldosteronism (PA) is the most common cause of secondary hypertension. A growing body of evidence has suggested that, beyond its well-known effects on blood pressure and electrolyte balance, aldosterone excess can exert pro-inflammatory, pro-oxidant and pro-fibrotic effects on the kidney, blood vessels and heart, leading to potentially harmful pathophysiological consequences. In clinical studies, PA has been associated with an increased risk of cardiovascular, cerebrovascular, renal and metabolic complication compared to essential hypertension, including atrial fibrillation (AF) and aortic ectasia. An increased prevalence of AF in patients with PA has been demonstrated in several clinical studies. Aldosterone excess seems to be involved in the pathogenesis of AF by inducing cardiac structural and electrical remodeling that in turn predisposes to arrhythmogenicity. The association between PA and aortic ectasia is less established, but several studies have demonstrated an effect of aldosterone on aortic stiffness, vascular smooth muscle cells and media composition that, in turn, might lead to an increased risk of aortic dilation and dissection. In this review, we focus on the current evidence regarding the potential role of aldosterone excess in the pathogenesis of AF and aortic ectasia.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Mirko Parasiliti-Caprino
- Endocrinology, Diabetes and Metabolism, City of Health and Science University Hospital, Department of Medical Sciences, University of Turin, 10126 Turin, Italy; (M.B.); (C.L.); (F.B.); (F.P.); (E.G.); (M.M.)
| |
Collapse
|
8
|
Pathophysiology and Therapeutics of Thoracic Aortic Aneurysm in Marfan Syndrome. Biomolecules 2022; 12:biom12010128. [PMID: 35053276 PMCID: PMC8773516 DOI: 10.3390/biom12010128] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 01/04/2022] [Accepted: 01/06/2022] [Indexed: 12/16/2022] Open
Abstract
About 20% of individuals afflicted with thoracic aortic disease have single-gene mutations that predispose the vessel to aneurysm formation and/or acute aortic dissection often without associated syndromic features. One widely studied exception is Marfan syndrome (MFS) in which mutations in the extracellular protein fibrillin-1 cause additional abnormalities in the heart, eyes, and skeleton. Mouse models of MFS have been instrumental in delineating major cellular and molecular determinants of thoracic aortic disease. In spite of research efforts, translating experimental findings from MFS mice into effective drug therapies for MFS patients remains an unfulfilled promise. Here, we describe a series of studies that have implicated endothelial dysfunction and improper angiotensin II and TGFβ signaling in driving thoracic aortic disease in MFS mice. We also discuss how these investigations have influenced the way we conceptualized possible new therapies to slow down or even halt aneurysm progression in this relatively common connective tissue disorder.
Collapse
|
9
|
Chen H, Ng KY, Li S, Jin G, Cao Q, Lian Z, Luo X, Ding X, Zheng D. CHARACTERISTICS OF THE FOVEAL MICROVASCULATURE IN CHILDREN WITH MARFAN SYNDROME: An Optical Coherence Tomography Angiography Study. Retina 2022; 42:138-151. [PMID: 34935768 PMCID: PMC8687615 DOI: 10.1097/iae.0000000000003272] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
PURPOSE To investigate the characteristics of foveal microvasculature in children with Marfan syndrome (MFS). METHODS Ninety eyes from 45 MFS patients and 76 eyes from 38 healthy individuals of age-matched, sex-matched, and axial length-matched were enrolled. Characteristics of the superficial capillary plexus including the vessel density, perfusion density, and foveal avascular zone were analyzed by optical coherence tomography angiography. RESULTS The vessel density and the circularity index of the foveal avascular zone were significantly decreased in the MFS group compared with the controls (P = 0.017 and P = 0.004 respectively). In MFS group, the central vessel density (P = 0.003) and perfusion density (P = 0.001) were negatively correlated with the best-corrected visual acuity. The foveal avascular zone area was correlated with the aortic diameters (P = 0.001) and the paratemporal perfusion density was correlated with the ejection fraction (P = 0.003). Moreover, the paratemporal perfusion density and the circularity index of foveal avascular zone were found to be correlated with the aortic Z-score (P < 0.001 and P = 0.003 respectively). CONCLUSION Retinal microvascular decrease and its correlation with best-corrected visual acuity and cardiac functions were observed in the MFS group. The optical coherence tomography angiography may help to characterize the underlying pathophysiology features of MFS and enable early detection and prevention of vascular changes in MFS.
Collapse
Affiliation(s)
- Hui Chen
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China.
| | - Kit Yee Ng
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China.
| | - Songshan Li
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China.
| | - Guangming Jin
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China.
| | - Qianzhong Cao
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China.
| | - Zhangkai Lian
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China.
| | - Xiaoling Luo
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China.
| | - Xiaoyan Ding
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China.
| | - Danying Zheng
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China.
| |
Collapse
|
10
|
van Andel MM, Groenink M, van den Berg MP, Timmermans J, Scholte AJHA, Mulder BJM, Zwinderman AH, de Waard V. Genome-wide methylation patterns in Marfan syndrome. Clin Epigenetics 2021; 13:217. [PMID: 34895303 PMCID: PMC8665617 DOI: 10.1186/s13148-021-01204-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2021] [Accepted: 11/27/2021] [Indexed: 12/29/2022] Open
Abstract
Background Marfan syndrome (MFS) is a connective tissue disorder caused by mutations in the Fibrillin-1 gene (FBN1). Here, we undertook the first epigenome-wide association study (EWAS) in patients with MFS aiming at identifying DNA methylation loci associated with MFS phenotypes that may shed light on the disease process. Methods The Illumina 450 k DNA-methylation array was used on stored peripheral whole-blood samples of 190 patients with MFS originally included in the COMPARE trial. An unbiased genome-wide approach was used, and methylation of CpG-sites across the entire genome was evaluated. Additionally, we investigated CpG-sites across the FBN1-locus (15q21.1) more closely, since this is the gene defective in MFS. Differentially Methylated Positions (DMPs) and Differentially Methylated Regions (DMRs) were identified through regression analysis. Associations between methylation levels and aortic diameters and presence or absence of 21 clinical features of MFS at baseline were analyzed. Moreover, associations between aortic diameter change, and the occurrence of clinical events (death any cause, type-A or -B dissection/rupture, or aortic surgery) and methylation levels were analyzed. Results We identified 28 DMPs that are significantly associated with aortic diameters in patients with MFS. Seven of these DMPs (25%) could be allocated to a gene that was previously associated with cardiovascular diseases (HDAC4, IGF2BP3, CASZ1, SDK1, PCDHGA1, DIO3, PTPRN2). Moreover, we identified seven DMPs that were significantly associated with aortic diameter change and five DMP’s that associated with clinical events. No significant associations at p < 10–8 or p < 10–6 were found with any of the non-cardiovascular phenotypic MFS features. Investigating DMRs, clusters were seen mostly on X- and Y, and chromosome 18–22. The remaining DMRs indicated involvement of a large family of protocadherins on chromosome 5, which were not reported in MFS before. Conclusion This EWAS in patients with MFS has identified a number of methylation loci significantly associated with aortic diameters, aortic dilatation rate and aortic events. Our findings add to the slowly growing literature on the regulation of gene expression in MFS patients. Supplementary Information The online version contains supplementary material available at 10.1186/s13148-021-01204-4.
Collapse
Affiliation(s)
- Mitzi M van Andel
- Department of Cardiology, Amsterdam UMC, University of Amsterdam, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands.
| | - Maarten Groenink
- Department of Cardiology, Amsterdam UMC, University of Amsterdam, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands.,Department of Radiology, Amsterdam UMC, Amsterdam, The Netherlands
| | - Maarten P van den Berg
- Department of Cardiology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Janneke Timmermans
- Department of Cardiology, Radboud University Hospital, Nijmegen, The Netherlands
| | - Arthur J H A Scholte
- Department of Cardiology, Leiden University Medical Center, Leiden, The Netherlands
| | - Barbara J M Mulder
- Department of Cardiology, Amsterdam UMC, University of Amsterdam, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands
| | - Aeilko H Zwinderman
- Department of Clinical Epidemiology, Biostatistics and Bioinformatics, Amsterdam UMC, Amsterdam, The Netherlands
| | - Vivian de Waard
- Department of Medical Biochemistry, Amsterdam UMC, Amsterdam Cardiovascular Sciences, Amsterdam, The Netherlands
| |
Collapse
|
11
|
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
|
12
|
Rysz J, Gluba-Brzózka A, Rokicki R, Franczyk B. Oxidative Stress-Related Susceptibility to Aneurysm in Marfan's Syndrome. Biomedicines 2021; 9:biomedicines9091171. [PMID: 34572356 PMCID: PMC8467736 DOI: 10.3390/biomedicines9091171] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Revised: 08/23/2021] [Accepted: 09/01/2021] [Indexed: 01/01/2023] Open
Abstract
The involvement of highly reactive oxygen-derived free radicals (ROS) in the genesis and progression of various cardiovascular diseases, including arrhythmias, aortic dilatation, aortic dissection, left ventricular hypertrophy, coronary arterial disease and congestive heart failure, is well-established. It has also been suggested that ROS may play a role in aortic aneurysm formation in patients with Marfan's syndrome (MFS). This syndrome is a multisystem disorder with manifestations including cardiovascular, skeletal, pulmonary and ocular systems, however, aortic aneurysm and dissection are still the most life-threatening manifestations of MFS. In this review, we will concentrate on the impact of oxidative stress on aneurysm formation in patients with MFS as well as on possible beneficial effects of some agents with antioxidant properties. Mechanisms responsible for oxidative stress in the MFS model involve a decreased expression of superoxide dismutase (SOD) as well as enhanced expression of NAD(P)H oxidase, inducible nitric oxide synthase (iNOS) and xanthine oxidase. The results of studies have indicated that reactive oxygen species may be involved in smooth muscle cell phenotype switching and apoptosis as well as matrix metalloproteinase activation, resulting in extracellular matrix (ECM) remodeling. The progression of the thoracic aortic aneurysm was suggested to be associated with markedly impaired aortic contractile function and decreased nitric oxide-mediated endothelial-dependent relaxation.
Collapse
Affiliation(s)
- Jacek Rysz
- Department of Nephrology, Hypertension and Family Medicine, Medical University of Lodz, 90-549 Lodz, Poland; (J.R.); (B.F.)
| | - Anna Gluba-Brzózka
- Department of Nephrology, Hypertension and Family Medicine, Medical University of Lodz, 90-549 Lodz, Poland; (J.R.); (B.F.)
- Correspondence: or ; Tel.: +48-42-639-3750
| | - Robert Rokicki
- Clinic of Hand Surgery, Medical University of Lodz, 90-549 Lodz, Poland;
| | - Beata Franczyk
- Department of Nephrology, Hypertension and Family Medicine, Medical University of Lodz, 90-549 Lodz, Poland; (J.R.); (B.F.)
| |
Collapse
|
13
|
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
|
14
|
Balint B, Federspiel JM, Schwab T, Ehrlich T, Ramsthaler F, Schäfers HJ. Aortic Regurgitation Is Associated With Ascending Aortic Remodeling in the Nondilated Aorta. Arterioscler Thromb Vasc Biol 2021; 41:1179-1190. [PMID: 33441026 DOI: 10.1161/atvbaha.120.315739] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
OBJECTIVE The probability of aortic complications in patients with bicuspid aortic valve is higher in association with aortic regurgitation (AR) compared with aortic stenosis (AS) or normally functioning valves. The objective of this study was to determine whether this is related to the specific characteristics of aneurysmatic dilatation that includes AR or whether AR itself has a negative impact on the aortic wall, independent of aneurysmatic dilatation. Approach and Results: Nondilated aortic specimens were harvested intraoperatively from individuals with tricuspid aortic valves and either AS (n=10) or AR (n=16). For controls, nondilated aortas were harvested during autopsies from individuals with tricuspid aortic valves and no evidence of aortic valve disease (n=10). Histological and immunohistochemical analyses revealed that compared with control aortas, overall medial degeneration was more severe in AR-aortas (P=0.005) but not AS-aortas (P=0.23). This pathological remodeling included mucoid extracellular matrix accumulation (P=0.005), elastin loss (P=0.003), elastin fragmentation (P=0.008), and decreased expression of fibrillin (P=0.003) and collagen (P=0.008). Furthermore, eNOS (endothelial nitric oxide synthase) expression was decreased in the intima (P=0.0008) and in vasa vasorum (P=0.004) of AR-aortas but not AS-aortas (all P>0.05). Likewise, subendothelial apoptosis was increased in AR-aortas (P=0.03) but not AS-aortas (P=0.50). CONCLUSIONS AR has a negative effect on the nondilated ascending aortic wall. Accordingly, our results support the need for more detailed studies of the aortic wall in relation to aortic valve disease and may ultimately lead to more aggressive clinical monitoring and/or surgical criteria for patients with relevant AR. Graphic Abstract: A graphic abstract is available for this article.
Collapse
Affiliation(s)
- Brittany Balint
- Department of Thoracic- and Cardiovascular Surgery (B.B., J.M.F., T.S., T.E., H.-J.S.), Saarland University Medical Center, Homburg, Saar, Germany
| | - Jan M Federspiel
- Department of Thoracic- and Cardiovascular Surgery (B.B., J.M.F., T.S., T.E., H.-J.S.), Saarland University Medical Center, Homburg, Saar, Germany
| | - Tanja Schwab
- Department of Thoracic- and Cardiovascular Surgery (B.B., J.M.F., T.S., T.E., H.-J.S.), Saarland University Medical Center, Homburg, Saar, Germany
| | - Tristan Ehrlich
- Department of Thoracic- and Cardiovascular Surgery (B.B., J.M.F., T.S., T.E., H.-J.S.), Saarland University Medical Center, Homburg, Saar, Germany
| | - Frank Ramsthaler
- Institute of Forensic Medicine (F.R.), Saarland University Medical Center, Homburg, Saar, Germany
| | - Hans-Joachim Schäfers
- Department of Thoracic- and Cardiovascular Surgery (B.B., J.M.F., T.S., T.E., H.-J.S.), Saarland University Medical Center, Homburg, Saar, Germany
| |
Collapse
|
15
|
van Andel MM, Groenink M, Zwinderman AH, Mulder BJM, de Waard V. The Potential Beneficial Effects of Resveratrol on Cardiovascular Complications in Marfan Syndrome Patients⁻Insights from Rodent-Based Animal Studies. Int J Mol Sci 2019; 20:E1122. [PMID: 30841577 PMCID: PMC6429290 DOI: 10.3390/ijms20051122] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2019] [Revised: 02/26/2019] [Accepted: 03/01/2019] [Indexed: 12/12/2022] Open
Abstract
Marfan syndrome (MFS) patients are at risk for cardiovascular disease. In particular, for aortic aneurysm formation, which ultimately can result in a life-threatening aortic dissection or rupture. Over the years, research into a sufficient pharmacological treatment option against aortopathy has expanded, mostly due to the development of rodent disease models for aneurysm formation and dissections. Unfortunately, no optimal treatment strategy has yet been identified for MFS. The biologically-potent polyphenol resveratrol (RES), that occurs in nuts, plants, and the skin of grapes, was shown to have a positive effect on aortic repair in various rodent aneurysm models. RES demonstrated to affect aortic integrity and aortic dilatation. The beneficial processes relevant for MFS included the improvement of endothelial dysfunction, extracellular matrix degradation, and smooth muscle cell death. For the wide range of beneficial effects on these mechanisms, evidence was found for the following involved pathways; alleviating oxidative stress (change in eNOS/iNOS balance and decrease in NOX4), reducing protease activity to preserve the extracellular matrix (decrease in MMP2), and improving smooth muscle cell survival affecting aortic aging (changing the miR21/miR29 balance). Besides aortic features, MFS patients may also suffer from manifestations concerning the heart, such as mitral valve prolapse and left ventricular impairment, where evidence from rodent models shows that RES may aid in promoting cardiomyocyte survival directly (SIRT1 activation) or by reducing oxidative stress (increasing superoxide dismutase) and increasing autophagy (AMPK activation). This overview discusses recent RES studies in animal models of aortic aneurysm formation and heart failure, where different advantageous effects have been reported that may collectively improve the aortic and cardiac pathology in patients with MFS. Therefore, a clinical study with RES in MFS patients seems justified, to validate RES effectiveness, and to judge its suitability as potential new treatment strategy.
Collapse
Affiliation(s)
- Mitzi M van Andel
- Department of Cardiology, Amsterdam UMC, Academic Medical Center, Amsterdam Cardiovascular Sciences, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands.
| | - Maarten Groenink
- Department of Cardiology, Amsterdam UMC, Academic Medical Center, Amsterdam Cardiovascular Sciences, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands.
- Department of Radiology, Amsterdam UMC, Academic Medical Center, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands.
| | - Aeilko H Zwinderman
- Department of Clinical Epidemiology, Amsterdam UMC, Academic Medical Center, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands.
| | - Barbara J M Mulder
- Department of Cardiology, Amsterdam UMC, Academic Medical Center, Amsterdam Cardiovascular Sciences, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands.
| | - Vivian de Waard
- Department of Medical Biochemistry, Amsterdam UMC, Academic Medical Center, Amsterdam Cardiovascular Sciences, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands.
| |
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
|
Takeda N, Hara H, Fujiwara T, Kanaya T, Maemura S, Komuro I. TGF-β Signaling-Related Genes and Thoracic Aortic Aneurysms and Dissections. Int J Mol Sci 2018; 19:ijms19072125. [PMID: 30037098 PMCID: PMC6073540 DOI: 10.3390/ijms19072125] [Citation(s) in RCA: 86] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2018] [Revised: 07/13/2018] [Accepted: 07/16/2018] [Indexed: 12/29/2022] Open
Abstract
Transforming growth factor-β (TGF)-β signaling plays a crucial role in the development and maintenance of various organs, including the vasculature. Accordingly, the mutations in TGF-β signaling pathway-related genes cause heritable disorders of the connective tissue, such as Marfan syndrome (MFS), Loeys-Dietz syndrome (LDS), and Shprintzen-Goldberg syndrome (SGS), and these syndromes may affect skeletal, ocular, pulmonary, and cardiovascular systems. Aortic root aneurysms are common problems that can result in aortic dissection or rupture, which is the leading cause of sudden death in the natural history of MFS and LDS, and recent improvements in surgical treatment have improved life expectancy. However, there is currently no genotype-specific medical treatment. Accumulating evidence suggest that not only structural weakness of connective tissue but also increased TGF-β signaling contributes to the complicated pathogenesis of aortic aneurysm formation, but a comprehensive understanding of governing molecular mechanisms remains lacking. Inhibition of angiotensin II receptor signaling and endothelial dysfunction have gained attention as a possible MFS treatment strategy, but interactions with TGF-β signaling remain elusive. Heterozygous loss-of-function mutations in TGF-β receptors 1 and 2 (TGFBR1 and TGFBR2) cause LDS, but TGF-β signaling is activated in the aorta (referred to as the TGF-β paradox) by mechanisms yet to be elucidated. In this review, we present and discuss the current understanding of molecular mechanisms responsible for aortopathies of MFS and related disorders.
Collapse
Affiliation(s)
- Norifumi Takeda
- Department of Cardiovascular Medicine, The University of Tokyo Hospital, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8655, Japan.
| | - Hironori Hara
- Department of Cardiovascular Medicine, The University of Tokyo Hospital, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8655, Japan.
| | - Takayuki Fujiwara
- Department of Cardiovascular Medicine, The University of Tokyo Hospital, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8655, Japan.
| | - Tsubasa Kanaya
- Department of Cardiovascular Medicine, The University of Tokyo Hospital, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8655, Japan.
| | - Sonoko Maemura
- Department of Cardiovascular Medicine, The University of Tokyo Hospital, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8655, Japan.
| | - Issei Komuro
- Department of Cardiovascular Medicine, The University of Tokyo Hospital, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8655, Japan.
| |
Collapse
|
18
|
|
19
|
Yamashiro Y, Yanagisawa H. Crossing Bridges between Extra- and Intra-Cellular Events in Thoracic Aortic Aneurysms. J Atheroscler Thromb 2017; 25:99-110. [PMID: 28943527 PMCID: PMC5827090 DOI: 10.5551/jat.rv17015] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Thoracic aortic aneurysms (TAAs) are common, life-threatening diseases and are a major cause of mortality and morbidity. Over the past decade, genetic approaches have revealed that 1) activation of the transforming growth factor beta (TGF-β) signaling, 2) alterations in the contractile apparatus of vascular smooth muscle cells (SMCs), and 3) defects in the extracellular matrix (ECM) were responsible for development of TAAs. Most recently, a fourth mechanism has been proposed in that dysfunction of mechanosensing in the aortic wall in response to hemodynamic stress may be a key driver of TAAs. Interestingly, the elastin-contractile unit, which is an anatomical and functional unit connecting extracellular elastic laminae to the intracellular SMC contractile filaments, via cell surface receptors, has been shown to play a critical role in the mechanosensing of SMCs, and many genes identified in TAAs encode for proteins along this continuum. However, it is still debated whether these four pathways converge into a common pathway. Currently, an effective therapeutic strategy based on the underlying mechanism of each type of TAAs has not been established. In this review, we will update the present knowledge on the molecular mechanism of TAAs with a focus on the signaling pathways potentially involved in the initiation of TAAs. Finally, we will evaluate current therapeutic strategies for TAAs and propose new directions for future treatment of TAAs.
Collapse
Affiliation(s)
- Yoshito Yamashiro
- Life Science Center of Tsukuba Advanced Research Alliance, University of Tsukuba
| | - Hiromi Yanagisawa
- Life Science Center of Tsukuba Advanced Research Alliance, University of Tsukuba
| |
Collapse
|
20
|
Jekell A, Kalani M, Kahan T. The effects of alpha 1-adrenoceptor blockade and angiotensin converting enzyme inhibition on central and brachial blood pressure and vascular reactivity: the doxazosin-ramipril study. Heart Vessels 2016; 32:674-684. [PMID: 27885499 PMCID: PMC5446849 DOI: 10.1007/s00380-016-0924-9] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/06/2016] [Accepted: 11/18/2016] [Indexed: 12/24/2022]
Abstract
We aimed to study whether inhibition of the renin–angiotensin–aldosterone system has effects on vascular structure and function beyond the effects on blood pressure reduction alone. Patients with mild-to-moderate hypertension (n = 61, age 54 ± 12 years, 34% women) received the angiotensin converting enzyme inhibitor ramipril 10 mg or the alpha 1-adrenoceptor blocker doxazosin 8 mg double-blind for 12 weeks. Aortic blood pressure, pulse wave velocity, and augmentation index were assessed by applanation tonometry. Endothelial function was studied by forearm post-ischemic flow mediated vasodilatation and by pulse wave analysis with beta 2-adrenoceptor agonist stimulation. Skin microvascular reactivity was assessed by laser Doppler fluxmetry and iontophoresis. Treatment with doxazosin or ramipril reduced aortic and brachial blood pressures (all P < 0.001), with greater reductions in aortic than brachial systolic blood pressures (P = 0.021) and aortic/brachial pulse pressure ratio (P = 0.005). Compared to doxazosin, ramipril reduced carotid-femoral and carotid-radial pulse wave velocity (both P < 0.05). Forearm endothelial dependent and independent vasodilatation, assessed by post-ischemic flow mediated vasodilatation and glyceryl trinitrate, and by pulse wave analysis remained unchanged by both doxazosin and ramipril. In addition, skin microvascular endothelial dependent (acetylcholine) and independent vasodilatation (sodium nitroprusside) remained unchanged. In conclusion, ramipril reduced indices of aortic stiffness, suggesting that angiotensin converting enzyme inhibitor therapy may have effects beyond blood pressure reduction. However, treatment did not appear to influence endothelial function. Evidence of endothelial dysfunction and its possible improvement by antihypertensive treatment might require more advanced hypertension. This study is registered at ClinicalTrials.gov (NCT02901977) and at EudraCT (# 2007-000631-25).
Collapse
Affiliation(s)
- Andreas Jekell
- Division of Cardiovascular Medicine, Department of Clinical Sciences, Danderyd Hospital, Karolinska Institutet, Stockholm, Sweden.,Department of Cardiology, Danderyd University Hospital Corp, Stockholm, Sweden
| | - Majid Kalani
- Division of Cardiovascular Medicine, Department of Clinical Sciences, Danderyd Hospital, Karolinska Institutet, Stockholm, Sweden
| | - Thomas Kahan
- Division of Cardiovascular Medicine, Department of Clinical Sciences, Danderyd Hospital, Karolinska Institutet, Stockholm, Sweden. .,Department of Cardiology, Danderyd University Hospital Corp, Stockholm, Sweden.
| |
Collapse
|
21
|
Hibender S, Franken R, van Roomen C, Ter Braake A, van der Made I, Schermer EE, Gunst Q, van den Hoff MJ, Lutgens E, Pinto YM, Groenink M, Zwinderman AH, Mulder BJM, de Vries CJM, de Waard V. Resveratrol Inhibits Aortic Root Dilatation in the Fbn1C1039G/+ Marfan Mouse Model. Arterioscler Thromb Vasc Biol 2016; 36:1618-26. [PMID: 27283746 PMCID: PMC4961273 DOI: 10.1161/atvbaha.116.307841] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2016] [Accepted: 05/25/2016] [Indexed: 02/07/2023]
Abstract
Supplemental Digital Content is available in the text. Objective— Marfan syndrome (MFS) is a connective tissue disorder caused by mutations in the fibrillin-1 gene. Patients with MFS are at risk of aortic aneurysm formation and dissection. Usually, blood pressure–lowering drugs are used to reduce aortic events; however, this is not sufficient for most patients. In the aorta of smooth muscle cell–specific sirtuin-1–deficient mice, spontaneous aneurysm formation and senescence are observed. Resveratrol is known to enhance sirtuin-1 activity and to reduce senescence, which prompted us to investigate the effectiveness of resveratrol in inhibition of aortic dilatation in the Fbn1C1039G/+ MFS mouse model. Approach and Results— Aortic senescence strongly correlates with aortic root dilatation rate in MFS mice. However, although resveratrol inhibits aortic dilatation, it only shows a trend toward reduced aortic senescence. Resveratrol enhances nuclear localization of sirtuin-1 in the vessel wall and, in contrast to losartan, does not affect leukocyte infiltration nor activation of SMAD2 and extracellular signal–regulated kinases 1/2 (ERK1/2). Interestingly, specific sirtuin-1 activation (SRT1720) or inhibition (sirtinol) in MFS mice does not affect aortic root dilatation rate, although senescence is changed. Resveratrol reduces aortic elastin breaks and decreases micro-RNA-29b expression coinciding with enhanced antiapoptotic Bcl-2 expression and decreased number of terminal apoptotic cells. In cultured smooth muscle cells, the resveratrol effect on micro-RNA-29b downregulation is endothelial cell and nuclear factor κB-dependent. Conclusions— Resveratrol inhibits aortic root dilatation in MFS mice by promoting elastin integrity and smooth muscle cell survival, involving downregulation of the aneurysm-related micro-RNA-29b in the aorta. On the basis of these data, resveratrol holds promise as a novel intervention strategy for patients with MFS.
Collapse
Affiliation(s)
- Stijntje Hibender
- From the Department of Medical Biochemistry (S.H., C.v.R., A.t.B., E.E.S., E.L., C.J.M.d.V., V.d.W.), Department of Cardiology (R.F., M.G., B.J.M.M.), Department of Experimental Cardiology (I.v.d.M., Y.M.P.), Heart Failure Research Center (Q.G., M.J.v.d.H.), Department of Radiology (M.G.), Department of Clinical Epidemiology and Biostatistics (A.H.Z.), Academic Medical Center, Amsterdam, The Netherlands; and Institute for Cardiovascular Prevention (IPEK) and Ludwig Maximilians University, Munich, Germany (E.L.)
| | - Romy Franken
- From the Department of Medical Biochemistry (S.H., C.v.R., A.t.B., E.E.S., E.L., C.J.M.d.V., V.d.W.), Department of Cardiology (R.F., M.G., B.J.M.M.), Department of Experimental Cardiology (I.v.d.M., Y.M.P.), Heart Failure Research Center (Q.G., M.J.v.d.H.), Department of Radiology (M.G.), Department of Clinical Epidemiology and Biostatistics (A.H.Z.), Academic Medical Center, Amsterdam, The Netherlands; and Institute for Cardiovascular Prevention (IPEK) and Ludwig Maximilians University, Munich, Germany (E.L.)
| | - Cindy van Roomen
- From the Department of Medical Biochemistry (S.H., C.v.R., A.t.B., E.E.S., E.L., C.J.M.d.V., V.d.W.), Department of Cardiology (R.F., M.G., B.J.M.M.), Department of Experimental Cardiology (I.v.d.M., Y.M.P.), Heart Failure Research Center (Q.G., M.J.v.d.H.), Department of Radiology (M.G.), Department of Clinical Epidemiology and Biostatistics (A.H.Z.), Academic Medical Center, Amsterdam, The Netherlands; and Institute for Cardiovascular Prevention (IPEK) and Ludwig Maximilians University, Munich, Germany (E.L.)
| | - Anique Ter Braake
- From the Department of Medical Biochemistry (S.H., C.v.R., A.t.B., E.E.S., E.L., C.J.M.d.V., V.d.W.), Department of Cardiology (R.F., M.G., B.J.M.M.), Department of Experimental Cardiology (I.v.d.M., Y.M.P.), Heart Failure Research Center (Q.G., M.J.v.d.H.), Department of Radiology (M.G.), Department of Clinical Epidemiology and Biostatistics (A.H.Z.), Academic Medical Center, Amsterdam, The Netherlands; and Institute for Cardiovascular Prevention (IPEK) and Ludwig Maximilians University, Munich, Germany (E.L.)
| | - Ingeborg van der Made
- From the Department of Medical Biochemistry (S.H., C.v.R., A.t.B., E.E.S., E.L., C.J.M.d.V., V.d.W.), Department of Cardiology (R.F., M.G., B.J.M.M.), Department of Experimental Cardiology (I.v.d.M., Y.M.P.), Heart Failure Research Center (Q.G., M.J.v.d.H.), Department of Radiology (M.G.), Department of Clinical Epidemiology and Biostatistics (A.H.Z.), Academic Medical Center, Amsterdam, The Netherlands; and Institute for Cardiovascular Prevention (IPEK) and Ludwig Maximilians University, Munich, Germany (E.L.)
| | - Edith E Schermer
- From the Department of Medical Biochemistry (S.H., C.v.R., A.t.B., E.E.S., E.L., C.J.M.d.V., V.d.W.), Department of Cardiology (R.F., M.G., B.J.M.M.), Department of Experimental Cardiology (I.v.d.M., Y.M.P.), Heart Failure Research Center (Q.G., M.J.v.d.H.), Department of Radiology (M.G.), Department of Clinical Epidemiology and Biostatistics (A.H.Z.), Academic Medical Center, Amsterdam, The Netherlands; and Institute for Cardiovascular Prevention (IPEK) and Ludwig Maximilians University, Munich, Germany (E.L.)
| | - Quinn Gunst
- From the Department of Medical Biochemistry (S.H., C.v.R., A.t.B., E.E.S., E.L., C.J.M.d.V., V.d.W.), Department of Cardiology (R.F., M.G., B.J.M.M.), Department of Experimental Cardiology (I.v.d.M., Y.M.P.), Heart Failure Research Center (Q.G., M.J.v.d.H.), Department of Radiology (M.G.), Department of Clinical Epidemiology and Biostatistics (A.H.Z.), Academic Medical Center, Amsterdam, The Netherlands; and Institute for Cardiovascular Prevention (IPEK) and Ludwig Maximilians University, Munich, Germany (E.L.)
| | - Maurice J van den Hoff
- From the Department of Medical Biochemistry (S.H., C.v.R., A.t.B., E.E.S., E.L., C.J.M.d.V., V.d.W.), Department of Cardiology (R.F., M.G., B.J.M.M.), Department of Experimental Cardiology (I.v.d.M., Y.M.P.), Heart Failure Research Center (Q.G., M.J.v.d.H.), Department of Radiology (M.G.), Department of Clinical Epidemiology and Biostatistics (A.H.Z.), Academic Medical Center, Amsterdam, The Netherlands; and Institute for Cardiovascular Prevention (IPEK) and Ludwig Maximilians University, Munich, Germany (E.L.)
| | - Esther Lutgens
- From the Department of Medical Biochemistry (S.H., C.v.R., A.t.B., E.E.S., E.L., C.J.M.d.V., V.d.W.), Department of Cardiology (R.F., M.G., B.J.M.M.), Department of Experimental Cardiology (I.v.d.M., Y.M.P.), Heart Failure Research Center (Q.G., M.J.v.d.H.), Department of Radiology (M.G.), Department of Clinical Epidemiology and Biostatistics (A.H.Z.), Academic Medical Center, Amsterdam, The Netherlands; and Institute for Cardiovascular Prevention (IPEK) and Ludwig Maximilians University, Munich, Germany (E.L.)
| | - Yigal M Pinto
- From the Department of Medical Biochemistry (S.H., C.v.R., A.t.B., E.E.S., E.L., C.J.M.d.V., V.d.W.), Department of Cardiology (R.F., M.G., B.J.M.M.), Department of Experimental Cardiology (I.v.d.M., Y.M.P.), Heart Failure Research Center (Q.G., M.J.v.d.H.), Department of Radiology (M.G.), Department of Clinical Epidemiology and Biostatistics (A.H.Z.), Academic Medical Center, Amsterdam, The Netherlands; and Institute for Cardiovascular Prevention (IPEK) and Ludwig Maximilians University, Munich, Germany (E.L.)
| | - Maarten Groenink
- From the Department of Medical Biochemistry (S.H., C.v.R., A.t.B., E.E.S., E.L., C.J.M.d.V., V.d.W.), Department of Cardiology (R.F., M.G., B.J.M.M.), Department of Experimental Cardiology (I.v.d.M., Y.M.P.), Heart Failure Research Center (Q.G., M.J.v.d.H.), Department of Radiology (M.G.), Department of Clinical Epidemiology and Biostatistics (A.H.Z.), Academic Medical Center, Amsterdam, The Netherlands; and Institute for Cardiovascular Prevention (IPEK) and Ludwig Maximilians University, Munich, Germany (E.L.)
| | - Aeilko H Zwinderman
- From the Department of Medical Biochemistry (S.H., C.v.R., A.t.B., E.E.S., E.L., C.J.M.d.V., V.d.W.), Department of Cardiology (R.F., M.G., B.J.M.M.), Department of Experimental Cardiology (I.v.d.M., Y.M.P.), Heart Failure Research Center (Q.G., M.J.v.d.H.), Department of Radiology (M.G.), Department of Clinical Epidemiology and Biostatistics (A.H.Z.), Academic Medical Center, Amsterdam, The Netherlands; and Institute for Cardiovascular Prevention (IPEK) and Ludwig Maximilians University, Munich, Germany (E.L.)
| | - Barbara J M Mulder
- From the Department of Medical Biochemistry (S.H., C.v.R., A.t.B., E.E.S., E.L., C.J.M.d.V., V.d.W.), Department of Cardiology (R.F., M.G., B.J.M.M.), Department of Experimental Cardiology (I.v.d.M., Y.M.P.), Heart Failure Research Center (Q.G., M.J.v.d.H.), Department of Radiology (M.G.), Department of Clinical Epidemiology and Biostatistics (A.H.Z.), Academic Medical Center, Amsterdam, The Netherlands; and Institute for Cardiovascular Prevention (IPEK) and Ludwig Maximilians University, Munich, Germany (E.L.)
| | - Carlie J M de Vries
- From the Department of Medical Biochemistry (S.H., C.v.R., A.t.B., E.E.S., E.L., C.J.M.d.V., V.d.W.), Department of Cardiology (R.F., M.G., B.J.M.M.), Department of Experimental Cardiology (I.v.d.M., Y.M.P.), Heart Failure Research Center (Q.G., M.J.v.d.H.), Department of Radiology (M.G.), Department of Clinical Epidemiology and Biostatistics (A.H.Z.), Academic Medical Center, Amsterdam, The Netherlands; and Institute for Cardiovascular Prevention (IPEK) and Ludwig Maximilians University, Munich, Germany (E.L.)
| | - Vivian de Waard
- From the Department of Medical Biochemistry (S.H., C.v.R., A.t.B., E.E.S., E.L., C.J.M.d.V., V.d.W.), Department of Cardiology (R.F., M.G., B.J.M.M.), Department of Experimental Cardiology (I.v.d.M., Y.M.P.), Heart Failure Research Center (Q.G., M.J.v.d.H.), Department of Radiology (M.G.), Department of Clinical Epidemiology and Biostatistics (A.H.Z.), Academic Medical Center, Amsterdam, The Netherlands; and Institute for Cardiovascular Prevention (IPEK) and Ludwig Maximilians University, Munich, Germany (E.L.).
| |
Collapse
|
22
|
Maeda J, Kosaki K, Shiono J, Kouno K, Aeba R, Yamagishi H. Variable severity of cardiovascular phenotypes in patients with an early-onset form of Marfan syndrome harboring FBN1 mutations in exons 24-32. Heart Vessels 2016; 31:1717-23. [PMID: 26796135 DOI: 10.1007/s00380-016-0793-2] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/29/2015] [Accepted: 01/08/2016] [Indexed: 11/29/2022]
Abstract
A subgroup of patients with Marfan syndrome (MFS) who have mutations in exons 24-32 of the FBN1 gene manifests severe atrioventricular valve insufficiency and skeletal problems as early as the neonatal period. These patients usually die in the first 2 years of life, thus a region between exons 24 and 32 of FBN1 is recognized as a critical region for this neonatal form of MFS (nMFS). Here, we report five consecutive patients who manifested a cardiovascular phenotype until infancy with mutations in the critical region for nMFS. Although three of these patients showed severe mitral regurgitation and died before reaching 1 year of age, the remaining two patients survived for over 5 years under medical and/or surgical interventions. Two splicing mutations and one missense mutation were identified in the three deceased patients, whereas two missense mutations were found in the two survivors. Currently, the clinical severity of patients with early-onset MFS harboring mutations in the critical region for nMFS seem to be more variable than ever thought, and intensive treatments are recommended even in this subgroup of patients with MFS.
Collapse
Affiliation(s)
- Jun Maeda
- Department of Pediatrics, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582, Japan.
| | - Kenjiro Kosaki
- Center for Medical Genetics, Keio University School of Medicine, Tokyo, Japan
| | - Junko Shiono
- Department of Pediatric Cardiology, Ibaraki Children's Hospital, Ibaraki, Japan
| | - Kazuki Kouno
- Department of Pediatrics, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582, Japan
| | - Ryo Aeba
- Division of Cardiovascular Surgery, Keio University School of Medicine, Tokyo, Japan
| | - Hiroyuki Yamagishi
- Department of Pediatrics, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582, Japan
| |
Collapse
|
23
|
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
|
24
|
Comparison of pulse wave velocity assessed by three different techniques: Arteriograph, Complior, and Echo-tracking. Heart Vessels 2015; 31:568-77. [PMID: 25633054 DOI: 10.1007/s00380-015-0632-x] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/06/2014] [Accepted: 01/16/2015] [Indexed: 10/24/2022]
Abstract
Arterial stiffness estimated by pulse wave velocity (PWV) is an independent predictor of cardiovascular morbidity and mortality. Although recommended by the current guidelines, clinical applicability of this parameter is difficult, due to differences between the various techniques used to measure it and to biological variability. Our aim was to compare PWV assessed by 3 different commercially available systems. 100 subjects (51 ± 16 years, 45 men) were evaluated using the 3 methods: an oscillometric technique (Arteriograph, PWV-A); a piezo-electric method (Complior, PWV-C); and an high-resolution ultrasound technique implemented with an Echo-tracking system (Aloka, PWV-E). Conventional biological markers were measured. Correlations of PWV measured by the 3 methods were poor (r = 0.39, r = 0.39, and r = 0.31 for PWV-A vs. PWV-C, PWV-A vs. PWV-E, and PWV-C vs. PWV-E, respectively, all p < 0.05). By Bland-Altman analysis, mean difference (±SD) of PWV-A vs. PWV-C was -1.9 ± 2.0 m/s, of PWV-A vs. PWV-E -3.6 ± 1.9 m/s, and of PWV-C vs. PWV-E -2.7 ± 1.9 m/s, with a wide coefficient of variation (22.3, 25.7, and 25.7 %, respectively). As expected, PWV-A, PWV-C, and PWV-E correlated with other arterial stiffness parameters, such as intima-media thickness (r = 0.22, r = 0.22, and r = 0.36, respectively), E p (r = 0.37, r = 0.26, and r = 0.94, respectively), and augmentation index measured by Arteriograph method (r = 0.66, r = 0.35, and r = 0.26, respectively); all p < 0.05. Assessment of PWV is markedly dependent on the technique used to measure it, related to various methods for measuring traveled distance of the arterial wave. Our results suggest the urgent need to establish reference values of PWV for each of these techniques, separately, to be used in routine clinical practice.
Collapse
|
25
|
A randomized, double blind pilot study to assess the effects of losartan vs. atenolol on the biophysical properties of the aorta in patients with Marfan and Loeys–Dietz syndromes. Int J Cardiol 2015; 179:470-5. [DOI: 10.1016/j.ijcard.2014.11.082] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/30/2014] [Revised: 10/23/2014] [Accepted: 11/06/2014] [Indexed: 11/22/2022]
|
26
|
Fujita D, Takeda N, Imai Y, Inuzuka R, Komuro I, Hirata Y. Pathophysiology and Japanese clinical characteristics in Marfan syndrome. Pediatr Int 2014; 56:484-91. [PMID: 24931486 DOI: 10.1111/ped.12423] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/25/2014] [Revised: 05/22/2014] [Accepted: 05/28/2014] [Indexed: 11/27/2022]
Abstract
Marfan syndrome is an autosomal dominant heritable disorder of the connective tissue, caused by mutations of the gene FBN1, which encodes fibrillin-1, a major component of the microfibrils of the extracellular matrix. Fibrillin-1 interacts with transforming growth factor-β (TGF-β), and dysregulated TGF-β signaling plays a major role in the development of connective tissue disease and familial aortic aneurysm and dissection, including Marfan syndrome. Losartan, an angiotensin II blocker, has the potential to reduce TGF-β signaling and is expected to be an additional therapeutic option. Clinical diagnosis is made using the Ghent nosology, which requires comprehensive patient assessment and has been proven to work well, but evaluation of some of the diagnostic criteria by a single physician is difficult and time-consuming. A Marfan clinic was established at the University of Tokyo Hospital in 2005, together with cardiologists, cardiac surgeons, pediatricians, orthopedists, and ophthalmologists in one place, for the purpose of speedy and accurate evaluation and diagnosis of Marfan syndrome. In this review, we discuss the recent progress in diagnosis and treatment of Marfan syndrome, and the characteristics of Japanese patients with Marfan syndrome.
Collapse
Affiliation(s)
- Daishi Fujita
- Department of Cardiovascular Medicine, University of Tokyo Hospital, Tokyo, Japan
| | | | | | | | | | | |
Collapse
|
27
|
Suzuki JI, Imai Y, Aoki M, Fujita D, Aoyama N, Tada Y, Wakayama K, Akazawa H, Izumi Y, Isobe M, Komuro I, Nagai R, Hirata Y. Periodontitis in cardiovascular disease patients with or without Marfan syndrome--a possible role of Prevotella intermedia. PLoS One 2014; 9:e95521. [PMID: 24748407 PMCID: PMC3991676 DOI: 10.1371/journal.pone.0095521] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2014] [Accepted: 03/27/2014] [Indexed: 02/06/2023] Open
Abstract
Background Although periodontitis is a risk factor for cardiovascular disease (CVD), the influence of periodontitis on Marfan syndrome (MFS) with CVD is unclear. The aim of this study was to assess the relationship between periodontal bacterial burden and MSF with CVD. Methods and Results The subjects were patients with MFS with CVD (n = 47); age and gender matched non-MFS CVD patients (n = 48) were employed as controls. Full-mouth clinical measurements, including number of teeth, probing of pocket depth (PD), bleeding on probing (BOP) and community periodontal index (CPI) were recorded. We also evaluated the existence of three periodontal pathogens, Porphyromonas gingivalis, Aggregatibacter actinomycetemcomitans, and Prevotella intermedia using polymerase chain reaction assays. Serum antibody titers against the pathogens were also measured. We revealed that MFS with CVD patients had periodontitis more frequently than the age and gender matched non-MFS CVD control subjects. MFS with CVD patients had significantly severer periodontitis, fewer remaining teeth and deeper PD compared to the non-MFS CVD controls. Furthermore, the serum antibody titer level against Prevotella intermedia was significantly lower in MFS plus CVD patients compared to the non-MFS CVD patients. Conclusion Periodontitis may influence the pathophysiology of cardiovascular complications in MFS patients. A specific periodontal pathogen might be a crucial therapeutic target to prevent CVD development.
Collapse
Affiliation(s)
- Jun-ichi Suzuki
- Department of Advanced Clinical Science and Therapeutics, The University of Tokyo, Bunkyo, Tokyo, Japan
- * E-mail:
| | - Yasushi Imai
- Jichi Medical University, Shimotsuke, Tochigi, Japan
| | - Mieko Aoki
- Department of Cardiovascular Medicine, The University of Tokyo, Bunkyo, Tokyo, Japan
| | - Daishi Fujita
- Department of Cardiovascular Medicine, The University of Tokyo, Bunkyo, Tokyo, Japan
| | - Norio Aoyama
- Department of Periodontology, Tokyo Medical and Dental University, Bunkyo, Tokyo, Japan
| | - Yuko Tada
- Department of Advanced Clinical Science and Therapeutics, The University of Tokyo, Bunkyo, Tokyo, Japan
| | - Kouji Wakayama
- Department of Advanced Clinical Science and Therapeutics, The University of Tokyo, Bunkyo, Tokyo, Japan
| | - Hiroshi Akazawa
- Department of Advanced Clinical Science and Therapeutics, The University of Tokyo, Bunkyo, Tokyo, Japan
| | - Yuichi Izumi
- Department of Periodontology, Tokyo Medical and Dental University, Bunkyo, Tokyo, Japan
| | - Mitsuaki Isobe
- Department of Cardiovascular Medicine, Tokyo Medical and Dental University, Bunkyo, Tokyo, Japan
| | - Issei Komuro
- Department of Cardiovascular Medicine, The University of Tokyo, Bunkyo, Tokyo, Japan
| | - Ryozo Nagai
- Jichi Medical University, Shimotsuke, Tochigi, Japan
| | | |
Collapse
|
28
|
Suzuki JI, Imai Y, Aoki M, Fujita D, Aoyama N, Tada Y, Akazawa H, Izumi Y, Isobe M, Komuro I, Nagai R, Hirata Y. High incidence and severity of periodontitis in patients with Marfan syndrome in Japan. Heart Vessels 2013; 30:692-5. [PMID: 24202189 DOI: 10.1007/s00380-013-0434-y] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/02/2013] [Accepted: 10/25/2013] [Indexed: 11/30/2022]
Abstract
Marfan syndrome (MFS) is a systemic connective tissue disorder caused by mutations in the extracellular matrix protein fibrillin-1. While it is known that patients with MFS are at high risk of dental disorders and cardiovascular diseases, little information has been provided to date. To clarify the prevalence of periodontitis in patients with MFS, their oral condition and cardiovascular complications were evaluated. The subjects were patients with MFS (n = 40) who attended the University of Tokyo hospital; age- and gender-matched healthy individuals (n = 14) constituted a control group. Cardiovascular complications and full-mouth clinical measurements, including number of teeth, probing of pocket depth (PD), bleeding on probing (BOP), and community periodontal index (CPI) were recorded. MFS patients had more frequent cardiovascular complications (95 %) compared with the controls (0 %). MFS patients had periodontitis (CPI 3 and 4) more frequently (87.5 %) than the age- and gender-matched control subjects (35.7 %). Furthermore, MFS patients had significantly more severe periodontitis (CPI 2.90 ± 0.12 vs 1.64 ± 0.32) and fewer remaining teeth (26.7 ± 0.4 vs 28.4 ± 0.4) compared with the controls. However, PD and BOP were comparable between MFS patients and the control group. A high incidence of periodontitis and cardiovascular complications was observed in Japanese MFS patients.
Collapse
Affiliation(s)
- Jun-Ichi Suzuki
- Department of Advanced Clinical Science and Therapeutics, University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan.
| | - Yasushi Imai
- Department of Cardiovascular Medicine, University of Tokyo, Tokyo, Japan
| | - Mieko Aoki
- Department of Cardiovascular Medicine, University of Tokyo, Tokyo, Japan
| | - Daishi Fujita
- Department of Cardiovascular Medicine, University of Tokyo, Tokyo, Japan
| | - Norio Aoyama
- Section of Periodontology, Department of Hard Tissue Engineering, Tokyo Medical and Dental University, Tokyo, Japan
| | - Yuko Tada
- Department of Advanced Clinical Science and Therapeutics, University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan
| | - Hiroshi Akazawa
- Department of Advanced Clinical Science and Therapeutics, University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan
| | - Yuichi Izumi
- Section of Periodontology, Department of Hard Tissue Engineering, Tokyo Medical and Dental University, Tokyo, Japan
| | - Mitsuaki Isobe
- Department of Cardiovascular Medicine, Tokyo Medical and Dental University, Tokyo, Japan
| | - Issei Komuro
- Department of Cardiovascular Medicine, University of Tokyo, Tokyo, Japan
| | | | | |
Collapse
|