1
|
Zaradzki M, Rehberg F, Zwaans V, Hecker M, Karck M, Arif R, Soethoff JP, Wagner AH. Stabilisation of extracellular matrix is crucial to rapamycin-mediated life span increase in Marfan mgR/mgR mice. Biochem Pharmacol 2025; 235:116830. [PMID: 40021021 DOI: 10.1016/j.bcp.2025.116830] [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: 08/18/2024] [Revised: 12/13/2024] [Accepted: 02/25/2025] [Indexed: 03/03/2025]
Abstract
Marfan syndrome is a hereditary connective tissue disorder caused by heterozygous mutations in the fibrillin-1 gene (FBN1) and altered TGF-β signalling. Life-threatening complications involve thoracic aortic aneurysms (TAA) and dissections due to the disruption of microfibrillar assembly in the aortic wall. We previously demonstrated that Rapamycin, a typical mTOR pathway inhibitor, limits the ascending aorta elastolysis and expansion, significantly increasing lifespan in an established murine model of Marfan syndrome (Zaradzki et al., Biochem Pharmacol 2022). This study aimed to investigate how mTOR inhibition stabilises the aorta in fibrillin-1 hypomorphic mgR/mgR mice and previously observed increased life expectancy. We used antibody microarrays to detect protein expression in the proximal thoracic aorta of sham or rapamycin-treated male and female mgR/mgR mice immediately after the two-week treatment. Immunofluorescence staining was performed to visualize and quantify protein expression in the ascending thoracic aorta and arch four weeks after the short-term rapamycin treatment was completed. We showed that rapamycin significantly increased the abundance of extracellular matrix (ECM) proteins like cytokeratin-18 and betaglycan, also known as the TGF-β type 3 receptor (TGFBR3). In addition, it raises the abundance of aggrecanase-2 (ADAMTS5) and xylosyltransferase-1 proteins, enzymes involved in ECM remodelling and homeostasis. In conclusion, rapamycin affects the composition and organization of key ECM components, which determine the structure-function relationships in the aorta, thereby maintaining the balance critical for the increase in life expectancy. Using mTOR modulators for targeted therapy may help to prevent aortic complications of MFS and improve clinical outcomes.
Collapse
Affiliation(s)
- Marcin Zaradzki
- Department of Cardiac Surgery, University Hospital Heidelberg, Im Neuenheimer Feld 420, 69120 Heidelberg, Germany
| | - Franziska Rehberg
- Department of Cardiovascular Physiology, Heidelberg University, Im Neuenheimer Feld 326, 69120 Heidelberg, Germany
| | - Vanessa Zwaans
- Department of Cardiovascular Physiology, Heidelberg University, Im Neuenheimer Feld 326, 69120 Heidelberg, Germany
| | - Markus Hecker
- Department of Cardiovascular Physiology, Heidelberg University, Im Neuenheimer Feld 326, 69120 Heidelberg, Germany
| | - Matthias Karck
- Department of Cardiac Surgery, University Hospital Heidelberg, Im Neuenheimer Feld 420, 69120 Heidelberg, Germany
| | - Rawa Arif
- Department of Cardiac Surgery, University Hospital Heidelberg, Im Neuenheimer Feld 420, 69120 Heidelberg, Germany
| | - Jasmin P Soethoff
- Department of Cardiac Surgery, University Hospital Heidelberg, Im Neuenheimer Feld 420, 69120 Heidelberg, Germany.
| | - Andreas H Wagner
- Department of Cardiovascular Physiology, Heidelberg University, Im Neuenheimer Feld 326, 69120 Heidelberg, Germany
| |
Collapse
|
2
|
Dubacher N, Sugiyama K, Smith JD, Nussbaumer V, Csonka M, Ferenczi S, Kovács KJ, Caspar SM, Lamberti L, Meienberg J, Yanagisawa H, Sheppard MB, Matyas G. Novel Insights into the Aortic Mechanical Properties of Mice Modeling Hereditary Aortic Diseases. Thromb Haemost 2025; 125:142-152. [PMID: 38950604 PMCID: PMC11737803 DOI: 10.1055/s-0044-1787957] [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: 01/03/2024] [Accepted: 05/23/2024] [Indexed: 07/03/2024]
Abstract
OBJECTIVE Hereditary aortic diseases (hADs) increase the risk of aortic dissections and ruptures. Recently, we have established an objective approach to measure the rupture force of the murine aorta, thereby explaining the outcomes of clinical studies and assessing the added value of approved drugs in vascular Ehlers-Danlos syndrome (vEDS). Here, we applied our approach to six additional mouse hAD models. MATERIAL AND METHODS We used two mouse models (Fbn1C1041G and Fbn1mgR ) of Marfan syndrome (MFS) as well as one smooth-muscle-cell-specific knockout (SMKO) of Efemp2 and three CRISPR/Cas9-engineered knock-in models (Ltbp1, Mfap4, and Timp1). One of the two MFS models was subjected to 4-week-long losartan treatment. Per mouse, three rings of the thoracic aorta were prepared, mounted on a tissue puller, and uniaxially stretched until rupture. RESULTS The aortic rupture force of the SMKO and both MFS models was significantly lower compared with wild-type mice but in both MFS models higher than in mice modeling vEDS. In contrast, the Ltbp1, Mfap4, and Timp1 knock-in models presented no impaired aortic integrity. As expected, losartan treatment reduced aneurysm formation but surprisingly had no impact on the aortic rupture force of our MFS mice. CONCLUSION Our read-out system can characterize the aortic biomechanical integrity of mice modeling not only vEDS but also related hADs, allowing the aortic-rupture-force-focused comparison of mouse models. Furthermore, aneurysm progression alone may not be a sufficient read-out for aortic rupture, as antihypertensive drugs reducing aortic dilatation might not strengthen the weakened aortic wall. Our results may enable identification of improved medical therapies of hADs.
Collapse
Affiliation(s)
- Nicolo Dubacher
- Center for Cardiovascular Genetics and Gene Diagnostics, Swiss Foundation for People with Rare Diseases, Schlieren-Zurich, Switzerland
- Translational Cardiovascular Technologies, Department of Health Sciences, ETH Zurich, Zurich, Switzerland
| | - Kaori Sugiyama
- Institute for Advanced Research of Biosystem Dynamics, Research Institute for Science and Engineering, Waseda University, Tokyo, Japan
- Life Science Center for Survival Dynamics, Tsukuba Advanced Research Alliance, University of Tsukuba, Tsukuba, Japan
| | - Jeffrey D. Smith
- Saha Cardiovascular Research Center, University of Kentucky, Lexington, Kentucky, United States
| | - Vanessa Nussbaumer
- Center for Cardiovascular Genetics and Gene Diagnostics, Swiss Foundation for People with Rare Diseases, Schlieren-Zurich, Switzerland
| | - Máté Csonka
- Center for Cardiovascular Genetics and Gene Diagnostics, Swiss Foundation for People with Rare Diseases, Schlieren-Zurich, Switzerland
- Heart and Vascular Center, Semmelweis University, Budapest, Hungary
| | - Szilamér Ferenczi
- Laboratory of Molecular Neuroendocrinology, Institute of Experimental Medicine, Budapest, Hungary
| | - Krisztina J. Kovács
- Laboratory of Molecular Neuroendocrinology, Institute of Experimental Medicine, Budapest, Hungary
| | - Sylvan M. Caspar
- Center for Cardiovascular Genetics and Gene Diagnostics, Swiss Foundation for People with Rare Diseases, Schlieren-Zurich, Switzerland
| | - Lisa Lamberti
- Center for Cardiovascular Genetics and Gene Diagnostics, Swiss Foundation for People with Rare Diseases, Schlieren-Zurich, Switzerland
| | - Janine Meienberg
- Center for Cardiovascular Genetics and Gene Diagnostics, Swiss Foundation for People with Rare Diseases, Schlieren-Zurich, Switzerland
| | - Hiromi Yanagisawa
- Life Science Center for Survival Dynamics, Tsukuba Advanced Research Alliance, University of Tsukuba, Tsukuba, Japan
| | - Mary B. Sheppard
- Department of Family and Community Medicine, University of Kentucky, Lexington, Kentucky, United States
- Saha Aortic Center, University of Kentucky, Lexington, Kentucky, United States
| | - Gabor Matyas
- Center for Cardiovascular Genetics and Gene Diagnostics, Swiss Foundation for People with Rare Diseases, Schlieren-Zurich, Switzerland
- Zurich Center for Integrative Human Physiology, University of Zurich, Zurich, Switzerland
| |
Collapse
|
3
|
Udugampolage NS, Frolova S, Taurino J, Pini A, Martelli F, Voellenkle C. Coding and Non-Coding Transcriptomic Landscape of Aortic Complications in Marfan Syndrome. Int J Mol Sci 2024; 25:7367. [PMID: 39000474 PMCID: PMC11242319 DOI: 10.3390/ijms25137367] [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: 05/20/2024] [Revised: 06/21/2024] [Accepted: 06/27/2024] [Indexed: 07/16/2024] Open
Abstract
Marfan syndrome (MFS) is a rare congenital disorder of the connective tissue, leading to thoracic aortic aneurysms (TAA) and dissection, among other complications. Currently, the most efficient strategy to prevent life-threatening dissection is preventive surgery. Periodic imaging applying complex techniques is required to monitor TAA progression and to guide the timing of surgical intervention. Thus, there is an acute demand for non-invasive biomarkers for diagnosis and prognosis, as well as for innovative therapeutic targets of MFS. Unraveling the intricate pathomolecular mechanisms underlying the syndrome is vital to address these needs. High-throughput platforms are particularly well-suited for this purpose, as they enable the integration of different datasets, such as transcriptomic and epigenetic profiles. In this narrative review, we summarize relevant studies investigating changes in both the coding and non-coding transcriptome and epigenome in MFS-induced TAA. The collective findings highlight the implicated pathways, such as TGF-β signaling, extracellular matrix structure, inflammation, and mitochondrial dysfunction. Potential candidates as biomarkers, such as miR-200c, as well as therapeutic targets emerged, like Tfam, associated with mitochondrial respiration, or miR-632, stimulating endothelial-to-mesenchymal transition. While these discoveries are promising, rigorous and extensive validation in large patient cohorts is indispensable to confirm their clinical relevance and therapeutic potential.
Collapse
Affiliation(s)
| | - Svetlana Frolova
- Molecular Cardiology Laboratory, IRCCS Policlinico San Donato, 20097 Milan, Italy; (S.F.); (C.V.)
- Department of Biosciences, University of Milan, 20122 Milan, Italy
| | - Jacopo Taurino
- Cardiovascular-Genetic Center, IRCCS Policlinico San Donato, 20097 Milan, Italy; (N.S.U.); (J.T.); (A.P.)
| | - Alessandro Pini
- Cardiovascular-Genetic Center, IRCCS Policlinico San Donato, 20097 Milan, Italy; (N.S.U.); (J.T.); (A.P.)
| | - Fabio Martelli
- Molecular Cardiology Laboratory, IRCCS Policlinico San Donato, 20097 Milan, Italy; (S.F.); (C.V.)
| | - Christine Voellenkle
- Molecular Cardiology Laboratory, IRCCS Policlinico San Donato, 20097 Milan, Italy; (S.F.); (C.V.)
| |
Collapse
|
4
|
Vandersteen AM, Weerakkody RA, Parry DA, Kanonidou C, Toddie-Moore DJ, Vandrovcova J, Darlay R, Santoyo-Lopez J, Meynert A, Kazkaz H, Grahame R, Cummings C, Bartlett M, Ghali N, Brady AF, Pope FM, van Dijk FS, Cordell HJ, Aitman TJ. Genetic complexity of diagnostically unresolved Ehlers-Danlos syndrome. J Med Genet 2024; 61:232-238. [PMID: 37813462 DOI: 10.1136/jmg-2023-109329] [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: 04/10/2023] [Accepted: 09/18/2023] [Indexed: 10/17/2023]
Abstract
BACKGROUND The Ehlers-Danlos syndromes (EDS) are heritable disorders of connective tissue (HDCT), reclassified in the 2017 nosology into 13 subtypes. The genetic basis for hypermobile Ehlers-Danlos syndrome (hEDS) remains unknown. METHODS Whole exome sequencing (WES) was undertaken on 174 EDS patients recruited from a national diagnostic service for complex EDS and a specialist clinic for hEDS. Patients had already undergone expert phenotyping, laboratory investigation and gene sequencing, but were without a genetic diagnosis. Filtered WES data were reviewed for genes underlying Mendelian disorders and loci reported in EDS linkage, transcriptome and genome-wide association studies (GWAS). A genetic burden analysis (Minor Allele Frequency (MAF) <0.05) incorporating 248 Avon Longitudinal Study of Parents and Children (ALSPAC) controls sequenced as part of the UK10K study was undertaken using TASER methodology. RESULTS Heterozygous pathogenic (P) or likely pathogenic (LP) variants were identified in known EDS and Loeys-Dietz (LDS) genes. Multiple variants of uncertain significance where segregation and functional analysis may enable reclassification were found in genes associated with EDS, LDS, heritable thoracic aortic disease (HTAD), Mendelian disorders with EDS symptomatology and syndromes with EDS-like features. Genetic burden analysis revealed a number of novel loci, although none reached the threshold for genome-wide significance. Variants with biological plausibility were found in genes and pathways not currently associated with EDS or HTAD. CONCLUSIONS We demonstrate the clinical utility of large panel-based sequencing and WES for patients with complex EDS in distinguishing rare EDS subtypes, LDS and related syndromes. Although many of the P and LP variants reported in this cohort would be identified with current panel testing, they were not at the time of this study, highlighting the use of extended panels and WES as a clinical tool for complex EDS. Our results are consistent with the complex genetic architecture of EDS and suggest a number of novel hEDS and HTAD candidate genes and pathways.
Collapse
Affiliation(s)
- Anthony M Vandersteen
- Maritime Medical Genetics Service, IWK Health Centre, Halifax, Nova Scotia, Canada
- Faculty of Medicine, Department of Pediatrics, Dalhousie University, Halifax, Nova Scotia, Canada
- Centre for Genomic and Experimental Medicine, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, UK
| | - Ruwan A Weerakkody
- Centre for Genomic and Experimental Medicine, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, UK
- Institute of Clinical Sciences, Imperial College London, London, UK
- Department of Vascular Surgery, Royal Free Hospital, London, UK
| | - David A Parry
- Centre for Genomic and Experimental Medicine, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, UK
| | - Christina Kanonidou
- Department of Clinical Biochemistry, Queen Elizabeth University Hospital, NHS Greater Glasgow and Clyde, Glasgow, UK
| | - Daniel J Toddie-Moore
- Centre for Genomic and Experimental Medicine, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, UK
| | - Jana Vandrovcova
- Department of Neuromuscular Diseases, UCL Queen Street Institute of Neurology, University College London, London, UK
| | - Rebecca Darlay
- Population Health Sciences Institute, Newcastle University, Newcastle upon Tyne, UK
| | | | - Alison Meynert
- MRC Human Genetics Unit, University of Edinburgh, Edinburgh, UK
| | - Hanadi Kazkaz
- Department of Rheumatology, University College London Hospitals NHS Foundation Trust, London, UK
| | - Rodney Grahame
- Department of Rheumatology, University College London Hospitals NHS Foundation Trust, London, UK
| | - Carole Cummings
- Ehlers-Danlos Syndrome National Diagnostic Service, London North West University Healthcare NHS Trust, Northwick Park Hospital, Harrow, UK
- Department of Metabolism, Digestion and Reproduction Section of Genetics and Genomics, Imperial College London, London, UK
| | - Marion Bartlett
- Ehlers-Danlos Syndrome National Diagnostic Service, London North West University Healthcare NHS Trust, Northwick Park Hospital, Harrow, UK
- Department of Metabolism, Digestion and Reproduction Section of Genetics and Genomics, Imperial College London, London, UK
| | - Neeti Ghali
- Ehlers-Danlos Syndrome National Diagnostic Service, London North West University Healthcare NHS Trust, Northwick Park Hospital, Harrow, UK
- Department of Metabolism, Digestion and Reproduction Section of Genetics and Genomics, Imperial College London, London, UK
| | - Angela F Brady
- Ehlers-Danlos Syndrome National Diagnostic Service, London North West University Healthcare NHS Trust, Northwick Park Hospital, Harrow, UK
- Department of Metabolism, Digestion and Reproduction Section of Genetics and Genomics, Imperial College London, London, UK
| | - F Michael Pope
- Ehlers-Danlos Syndrome National Diagnostic Service, London North West University Healthcare NHS Trust, Northwick Park Hospital, Harrow, UK
- Department of Metabolism, Digestion and Reproduction Section of Genetics and Genomics, Imperial College London, London, UK
| | - Fleur S van Dijk
- Ehlers-Danlos Syndrome National Diagnostic Service, London North West University Healthcare NHS Trust, Northwick Park Hospital, Harrow, UK
- Department of Metabolism, Digestion and Reproduction Section of Genetics and Genomics, Imperial College London, London, UK
| | - Heather J Cordell
- Population Health Sciences Institute, Newcastle University, Newcastle upon Tyne, UK
| | - Timothy J Aitman
- Centre for Genomic and Experimental Medicine, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, UK
| |
Collapse
|
5
|
Zaradzki M, Mohr F, Lont S, Soethoff J, Remes A, Arif R, Müller OJ, Karck M, Hecker M, Wagner AH. Short-term rapamycin treatment increases life span and attenuates aortic aneurysm in a murine model of Marfan-Syndrome. Biochem Pharmacol 2022; 205:115280. [PMID: 36198355 DOI: 10.1016/j.bcp.2022.115280] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Revised: 09/27/2022] [Accepted: 09/27/2022] [Indexed: 12/01/2022]
Abstract
BACKGROUND Marfan syndrome (MFS) is a genetic disorder leading to medial aortic degeneration and life-limiting dissections. To date, there is no causal prevention or therapy. Rapamycin is a potent and selective inhibitor of the mechanistic target of rapamycin (mTOR) protein kinase, regulating cell growth and metabolism. The mgR/mgR mice represent an accepted MFS model for studying aortic pathologies to understand the underlying molecular pathomechanisms. This study investigated whether rapamycin inhibits the development of thoracic aortic aneurysms and dissections in mgR/mgR mice. METHODS Isolated primary aortic smooth muscle cells (mAoSMCs) from mgR/mgR mice were used for in vitro studies. Two mg kg/BW rapamycin was injected intraperitoneally daily for two weeks, beginning at 7-8 weeks of age. Mice were sacrificed 30 days post-treatment. Histopathological and immunofluorescence analyses were performed using adequate tissue specimens and techniques. Animal survival was evaluated accompanied by periodic echocardiographic examinations of the aorta. RESULTS The protein level of the phosphorylated ribosomal protein S6 (p-RPS6), a downstream target of mTOR, was significantly increased in the aortic tissue of mgR/mgR mice. In mAoSMCs isolated from these animals, expression of mTOR, p-RPS6, tumour necrosis factor α, matrix metalloproteinase-2 and -9 was significantly suppressed by rapamycin, demonstrating its anti-inflammatory capacity. Short-term rapamycin treatment of Marfan mice was associated with delayed aneurysm formation, medial aortic elastolysis and improved survival. CONCLUSIONS Short-term rapamycin-mediated mTOR inhibition significantly reduces aortic aneurysm formation and thus increases survival in mgR/mgR mice. Our results may offer the first causal treatment option to prevent aortic complications in MFS patients.
Collapse
Affiliation(s)
- M Zaradzki
- Department of Cardiac Surgery, University Hospital Heidelberg, Heidelberg, Germany
| | - F Mohr
- Department of Cardiovascular Physiology, Heidelberg University, Heidelberg, Germany
| | - S Lont
- Department of Cardiovascular Physiology, Heidelberg University, Heidelberg, Germany
| | - J Soethoff
- Department of Cardiac Surgery, University Hospital Heidelberg, Heidelberg, Germany
| | - A Remes
- Department of Internal Medicine III, University of Kiel and University Hospital Schleswig-Holstein, Kiel; German Centre for Cardiovascular Research, Partner Site Hamburg/Kiel/Lübeck, Germany
| | - R Arif
- Department of Cardiac Surgery, University Hospital Heidelberg, Heidelberg, Germany
| | - O J Müller
- Department of Internal Medicine III, University of Kiel and University Hospital Schleswig-Holstein, Kiel; German Centre for Cardiovascular Research, Partner Site Hamburg/Kiel/Lübeck, Germany
| | - M Karck
- Department of Cardiac Surgery, University Hospital Heidelberg, Heidelberg, Germany
| | - M Hecker
- Department of Cardiovascular Physiology, Heidelberg University, Heidelberg, Germany
| | - A H Wagner
- Department of Cardiovascular Physiology, Heidelberg University, Heidelberg, Germany.
| |
Collapse
|
6
|
Xu C, Liu X, Fang X, Yu L, Lau HC, Li D, Liu X, Li H, Ren J, Xu B, Jiang J, Tang L, Chen X. Single-Cell RNA Sequencing Reveals Smooth Muscle Cells Heterogeneity in Experimental Aortic Dissection. Front Genet 2022; 13:836593. [PMID: 36035191 PMCID: PMC9403608 DOI: 10.3389/fgene.2022.836593] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Accepted: 06/16/2022] [Indexed: 11/13/2022] Open
Abstract
Purpose: This study aims to illustrate the cellular landscape in the aorta of experimental aortic dissection (AD) and elaborate on the smooth muscle cells (SMCs) heterogeneity and functions among various cell types.Methods: Male Apolipoprotein deficient (ApoE−/−) mice at 28 weeks of age were infused with Ang II (2,500 ng/kg/min) to induce AD. Aortas from euthanized mice were harvested after 7 days for 10×Genomics single-cell RNA sequencing (scRNA-seq), followed by the identification of cell types and differentially expressed genes (DEGs). Gene Ontology (GO) enrichment and Kyoto Encyclopedia of Genes and Genomes (KEGG) analysis was conducted.Results: AD was successfully induced in ApoE−/− mice. scRNA-seq identified 15 cell clusters and nine cell types, including non-immune cells (endothelials, fibroblasts, and SMCs) and immune cells (B cells, natural killer T cell, macrophages, dendritic cells, neutrophils, and mast cells). The relative numbers of SMCs were remarkably changed, and seven core DEGs (ACTA2,IL6,CTGF,BGN,ITGA8,THBS1, and CDH5) were identified in SMCs. Moreover, we found SMCs can differentiate into 8 different subtypes through single-cell trajectory analysis.Conclusion: scRNA-seq technology can successfully identify unique cell composition in experimental AD. To our knowledge, this is the first study that provided the complete cellular landscape in AD tissues from mice, seven core DEGs and eight subtypes of SMCs were identified, and the SMCs have evolution from matrix type to inflammatory type.
Collapse
Affiliation(s)
- Cheng Xu
- Department of Cardiology, Taizhou Hospital Affiliated to Wenzhou Medical University, Taizhou, China
| | - Xiaowei Liu
- Department of Cardiology, Zhejiang Hospital, Hangzhou, China
| | - Xiaoxin Fang
- Department of Cardiology, Taizhou Hospital Affiliated to Wenzhou Medical University, Taizhou, China
| | - Lei Yu
- Department of Cardiology, Taizhou Hospital Affiliated to Wenzhou Medical University, Taizhou, China
| | - Hui Chong Lau
- Department of Medicine, Crozer-Chester Medical Center, Upland, PA, United States
| | - Danlei Li
- Department of Cardiology, Taizhou Hospital Affiliated to Wenzhou Medical University, Taizhou, China
| | - Xiaoman Liu
- Department of Cardiology, Taizhou Hospital Affiliated to Wenzhou Medical University, Taizhou, China
| | - Haili Li
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Justin Ren
- Department of Cardiology, Taizhou Hospital Affiliated to Wenzhou Medical University, Taizhou, China
| | - Baohui Xu
- Department of Surgery, Stanford University School of Medicine, Stanford, CA, United States
| | - Jianjun Jiang
- Department of Cardiology, Taizhou Hospital Affiliated to Wenzhou Medical University, Taizhou, China
| | - Lijiang Tang
- Department of Cardiology, Zhejiang Hospital, Hangzhou, China
- *Correspondence: Lijiang Tang, ; Xiaofeng Chen,
| | - Xiaofeng Chen
- Department of Cardiology, Taizhou Hospital Affiliated to Wenzhou Medical University, Taizhou, China
- Department of Radiation Oncology, Indiana University School of Medicine, Indianapolis, IN, United States
- *Correspondence: Lijiang Tang, ; Xiaofeng Chen,
| |
Collapse
|
7
|
Kanaan R, Medlej-Hashim M, Jounblat R, Pilecki B, Sorensen GL. Microfibrillar-associated protein 4 in health and disease. Matrix Biol 2022; 111:1-25. [DOI: 10.1016/j.matbio.2022.05.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Revised: 05/04/2022] [Accepted: 05/24/2022] [Indexed: 10/18/2022]
|
8
|
Wang XP, Li QL, Li W, Zhang T, Li XY, Jiao Y, Zhang XM, Jiang JJ, Zhang X, Zhang XM. Dexamethasone attenuated thoracic aortic aneurysm and dissection in vascular smooth muscle cell Tgfbr2 disrupted mice with CCL8 suppression. Exp Physiol 2022; 107:631-645. [PMID: 35344629 DOI: 10.1113/ep090190] [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: 11/01/2021] [Accepted: 03/23/2022] [Indexed: 11/08/2022]
Abstract
NEW FINDINGS What is the central question of this study? The aim of this study was to investigate the relationship of CCL8 and the thoracic aortic aneurysm and dissection (TAAD) formation in postnatal mice with vascular smooth muscle cell (VSMC) Tgfbr2 disruption and whether dexamethasone could be a potential treatment. What is the main finding and its importance? CCL8 was associated with the formation of TAAD in VSMC Tgfbr2 disrupted mice. Dexamethasone reduced TAAD formation and inhibited MAPK (p-p38) and NF-κB (p-p65) signaling pathways. CCL8 might be an important promoter in aortic inflammation. DEX provided potential therapeutic effects in TAAD treatment. ABSTRACT Aortic inflammation plays a vital role in initiation and progression of thoracic aortic aneurysm and dissection (TAAD). The disturbance of transforming growth factor-β (TGF-β) signaling pathway is believed to be one of the pathogenic mechanisms of TAAD. Initially, Myh11-CreERT2 .Tgfbr2f/f male mice were used to build TAAD mice model. And bioinformatics analyses revealed the enriched inflammatory signal pathways and upregulated chemokine CCL8. So we hypothesized that vascular smooth muscle cell (VSMC) Tgfbr2 disruption in postnatal mice resulted in aortic inflammation associated with CCL8 secretion. Then real-time quantitative PCR and serum ELISA results confirmed that CCL8 expression began to increase after VSMC Tgfbr2 disruption. Next, we cultured mouse thoracic aortas ex vivo, and observed that the protein expressions of CCL8 in culture supernatants were increased by ELISA. Subsequently, the co-localization of CCL8 with α-smooth muscle actin (α-SMA) orCD68 was found significantly increased by immunofluorescence. Then, dexamethasone (DEX) was used to treat TAAD in VSMC Tgfbr2 disrupted mice The results of histochemical, immunofluorescence and immunohistochemical staining indicated that DEX therapy reduced CCL8 secretion, inflammatory cell recruitment, aortic medial thickening, elastic fiber fragmentating, extracellular matrix degradation, contractile apparatus impairment, thereby ameliorated TAAD formation. Western blot showed that MAPK and NF-κB signaling pathways in aorta were overactivated after VSMC Tgfbr2 disruption, but inhibited by DEX therapy. Altogether, CCL8 might be an important promoter in TAAD formation of VSMC Tgfbr2 disrupted mice. And DEX provided potential therapeutic effects in TAAD treatment. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Xi-Peng Wang
- Department of Vascular Surgery, Peking University People's Hospital, Beijing, People's Republic of China
| | - Qing-Le Li
- Department of Vascular Surgery, Peking University People's Hospital, Beijing, People's Republic of China
| | - Wei Li
- Department of Vascular Surgery, Peking University People's Hospital, Beijing, People's Republic of China
| | - Tao Zhang
- Department of Vascular Surgery, Peking University People's Hospital, Beijing, People's Republic of China
| | - Xiao-Yan Li
- Beijing Anzhen Hospital, Capital Medical University, Beijing Institute of Heart, Lung and Blood Vessel Disease, Beijing, People's Republic of China
| | - Yang Jiao
- Department of Vascular Surgery, Peking University People's Hospital, Beijing, People's Republic of China
| | - Xue-Min Zhang
- Department of Vascular Surgery, Peking University People's Hospital, Beijing, People's Republic of China
| | - Jing-Jun Jiang
- Department of Vascular Surgery, Peking University People's Hospital, Beijing, People's Republic of China
| | - Xiaoping Zhang
- Beijing Anzhen Hospital, Capital Medical University, Beijing Institute of Heart, Lung and Blood Vessel Disease, Beijing, People's Republic of China
| | - Xiao-Ming Zhang
- Department of Vascular Surgery, Peking University People's Hospital, Beijing, People's Republic of China
| |
Collapse
|
9
|
Sex-Related Effects on Cardiac Development and Disease. J Cardiovasc Dev Dis 2022; 9:jcdd9030090. [PMID: 35323638 PMCID: PMC8949052 DOI: 10.3390/jcdd9030090] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Accepted: 03/16/2022] [Indexed: 02/06/2023] Open
Abstract
Cardiovascular diseases (CVD) are the leading cause of morbidity and mortality. Interestingly, male and female patients with CVD exhibit distinct epidemiological and pathophysiological characteristics, implying a potentially important role for primary and secondary sex determination factors in heart development, aging, disease and therapeutic responses. Here, we provide a concise review of the field and discuss current gaps in knowledge as a step towards elucidating the “sex determination–heart axis”. We specifically focus on cardiovascular manifestations of abnormal sex determination in humans, such as in Turner and Klinefelter syndromes, as well as on the differences in cardiac regenerative potential between species with plastic and non-plastic sexual phenotypes. Sex-biased cardiac repair mechanisms are also discussed with a focus on the role of the steroid hormone 17β-estradiol. Understanding the “sex determination–heart axis” may offer new therapeutic possibilities for enhanced cardiac regeneration and/or repair post-injury.
Collapse
|
10
|
JACK N, MUTO T, IEMITSU K, WATANABE T, UMEYAMA K, OHGANE J, NAGASHIMA H. Genetically engineered animal models for Marfan syndrome: challenges associated with the generation of pig models for diseases caused by haploinsufficiency. J Reprod Dev 2022; 68:233-237. [PMID: 35598970 PMCID: PMC9334321 DOI: 10.1262/jrd.2022-027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Recent developments in reproductive biology have enabled the generation of genetically engineered pigs as models for inherited human diseases. Although a variety of such models for
monogenic diseases are currently available, reproduction of human diseases caused by haploinsufficiency remains a major challenge. The present study compares the phenotypes of mouse and pig
models of Marfan syndrome (MFS), with a special focus on the expressivity and penetrance of associated symptoms. Furthermore, investigation of the gene regulation mechanisms associated with
haploinsufficiency will be of immense utility in developing faithful MFS pig models.
Collapse
Affiliation(s)
- Naomi JACK
- Meiji University International Institute for Bio-Resource Research, Kawasaki 214-7824, Japan
| | - Tomoyuki MUTO
- Laboratory of Medical Bioengineering, Department of Life Sciences, School of Agriculture, Meiji University, Kawasaki 214-8571, Japan
| | - Keigo IEMITSU
- Laboratory of Medical Bioengineering, Department of Life Sciences, School of Agriculture, Meiji University, Kawasaki 214-8571, Japan
| | - Tamaki WATANABE
- Laboratory of Medical Bioengineering, Department of Life Sciences, School of Agriculture, Meiji University, Kawasaki 214-8571, Japan
| | - Kazuhiro UMEYAMA
- Meiji University International Institute for Bio-Resource Research, Kawasaki 214-7824, Japan
| | - Jun OHGANE
- Meiji University International Institute for Bio-Resource Research, Kawasaki 214-7824, Japan
| | - Hiroshi NAGASHIMA
- Meiji University International Institute for Bio-Resource Research, Kawasaki 214-7824, Japan
| |
Collapse
|
11
|
Chen JZ, Sawada H, Ye D, Katsumata Y, Kukida M, Ohno-Urabe S, Moorleghen JJ, Franklin MK, Howatt DA, Sheppard MB, Mullick AE, Lu HS, Daugherty A. Deletion of AT1a (Angiotensin II Type 1a) Receptor or Inhibition of Angiotensinogen Synthesis Attenuates Thoracic Aortopathies in Fibrillin1 C1041G/+ Mice. Arterioscler Thromb Vasc Biol 2021; 41:2538-2550. [PMID: 34407634 PMCID: PMC8458261 DOI: 10.1161/atvbaha.121.315715] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Objective: A cardinal feature of Marfan syndrome is thoracic aortic aneurysm. The contribution of the renin-angiotensin system via AT1aR (Ang II [angiotensin II] receptor type 1a) to thoracic aortic aneurysm progression remains controversial because the beneficial effects of angiotensin receptor blockers have been ascribed to off-target effects. This study used genetic and pharmacological modes of attenuating angiotensin receptor and ligand, respectively, to determine their roles on thoracic aortic aneurysm in mice with fibrillin-1 haploinsufficiency (Fbn1C1041G/+). Approach and Results: Thoracic aortic aneurysm in Fbn1C1041G/+ mice was found to be strikingly sexual dimorphic. Males displayed aortic dilation over 12 months while aortic dilation in Fbn1C1041G/+ females did not differ significantly from wild-type mice. To determine the role of AT1aR, Fbn1C1041G/+ mice that were either +/+ or -/- for AT1aR were generated. AT1aR deletion reduced expansion of ascending aorta and aortic root diameter from 1 to 12 months of age in males. Medial thickening and elastin fragmentation were attenuated. An antisense oligonucleotide against angiotensinogen was administered to male Fbn1C1041G/+ mice to determine the effects of Ang II depletion. Antisense oligonucleotide against angiotensinogen administration attenuated dilation of the ascending aorta and aortic root and reduced extracellular remodeling. Aortic transcriptome analyses identified potential targets by which inhibition of the renin-angiotensin system reduced aortic dilation in Fbn1C1041G/+ mice. Conclusions: Deletion of AT1aR or inhibition of Ang II production exerted similar effects in attenuating pathologies in the proximal thoracic aorta of male Fbn1C1041G/+ mice. Inhibition of the renin-angiotensin system attenuated dysregulation of genes within the aorta related to pathology of Fbn1C1041G/+ mice.
Collapse
MESH Headings
- Angiotensinogen/genetics
- Angiotensinogen/metabolism
- Animals
- Aorta, Thoracic/metabolism
- Aorta, Thoracic/pathology
- Aortic Aneurysm, Thoracic/genetics
- Aortic Aneurysm, Thoracic/metabolism
- Aortic Aneurysm, Thoracic/pathology
- Aortic Aneurysm, Thoracic/prevention & control
- Disease Models, Animal
- Female
- Fibrillin-1/genetics
- Fibrillin-1/metabolism
- Gene Deletion
- Genetic Predisposition to Disease
- Haploinsufficiency
- Male
- Marfan Syndrome/genetics
- Marfan Syndrome/metabolism
- Marfan Syndrome/pathology
- Mice, Inbred C57BL
- Mice, Knockout
- Oligonucleotides, Antisense/genetics
- Oligonucleotides, Antisense/metabolism
- Phenotype
- Receptor, Angiotensin, Type 1/deficiency
- Receptor, Angiotensin, Type 1/genetics
- Renin-Angiotensin System/genetics
- Sex Characteristics
- Sex Factors
- Transcriptome
- Mice
Collapse
Affiliation(s)
- Jeff Z. Chen
- Saha Cardiovascular Research Center, University of Kentucky, Lexington, KY
- Saha Aortic Center, University of Kentucky, Lexington, KY
- Department of Physiology, University of Kentucky, Lexington, KY
| | - Hisashi Sawada
- Saha Cardiovascular Research Center, University of Kentucky, Lexington, KY
- Saha Aortic Center, University of Kentucky, Lexington, KY
- Department of Physiology, University of Kentucky, Lexington, KY
| | - Dien Ye
- Saha Cardiovascular Research Center, University of Kentucky, Lexington, KY
- Saha Aortic Center, University of Kentucky, Lexington, KY
| | - Yuriko Katsumata
- Department Biostatistics, University of Kentucky, Lexington, KY
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY
| | - Masayoshi Kukida
- Saha Cardiovascular Research Center, University of Kentucky, Lexington, KY
- Saha Aortic Center, University of Kentucky, Lexington, KY
| | - Satoko Ohno-Urabe
- Saha Cardiovascular Research Center, University of Kentucky, Lexington, KY
- Saha Aortic Center, University of Kentucky, Lexington, KY
| | - Jessica J. Moorleghen
- Saha Cardiovascular Research Center, University of Kentucky, Lexington, KY
- Saha Aortic Center, University of Kentucky, Lexington, KY
| | - Michael K. Franklin
- Saha Cardiovascular Research Center, University of Kentucky, Lexington, KY
- Saha Aortic Center, University of Kentucky, Lexington, KY
| | - Deborah A. Howatt
- Saha Cardiovascular Research Center, University of Kentucky, Lexington, KY
- Saha Aortic Center, University of Kentucky, Lexington, KY
| | - Mary B. Sheppard
- Saha Cardiovascular Research Center, University of Kentucky, Lexington, KY
- Saha Aortic Center, University of Kentucky, Lexington, KY
- Department of Physiology, University of Kentucky, Lexington, KY
- Department of Family and Community Medicine, University of Kentucky, Lexington, KY
- Department of Surgery, University of Kentucky, Lexington, KY
| | | | - Hong S. Lu
- Saha Cardiovascular Research Center, University of Kentucky, Lexington, KY
- Saha Aortic Center, University of Kentucky, Lexington, KY
- Department of Physiology, University of Kentucky, Lexington, KY
| | - Alan Daugherty
- Saha Cardiovascular Research Center, University of Kentucky, Lexington, KY
- Saha Aortic Center, University of Kentucky, Lexington, KY
- Department of Physiology, University of Kentucky, Lexington, KY
| |
Collapse
|
12
|
Rare Causes of Arterial Hypertension and Thoracic Aortic Aneurysms-A Case-Based Review. Diagnostics (Basel) 2021; 11:diagnostics11030446. [PMID: 33807627 PMCID: PMC8001303 DOI: 10.3390/diagnostics11030446] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2021] [Revised: 02/28/2021] [Accepted: 03/01/2021] [Indexed: 11/16/2022] Open
Abstract
Thoracic aortic aneurysms may result in dissection with fatal consequences if undetected. A young male patient with no relevant familial history, after having been investigated for hypertension, was diagnosed with an ascending aortic aneurysm involving the aortic root and the proximal tubular segment, associated with a septal atrial defect. The patient underwent a Bentall surgery protocol without complications. Clinical examination revealed dorso-lumbar scoliosis and no other signs of underlying connective tissue disease. Microscopic examination revealed strikingly severe medial degeneration of the aorta, with areas of deep disorganization of the medial musculo-elastic structural units and mucoid material deposition. Genetic testing found a variant of unknown significance the PRKG1 gene encoding the protein kinase cGMP-dependent 1, which is important in blood pressure regulation. There may be genetic links between high blood pressure and thoracic aortic aneurysm determinants. Hypertension was found in FBN1 gene mutations encoding fibrillin and in PRKG1 mutations. Possible mechanisms involving the renin-angiotensin system, the role of oxidative stress, osteopontin, epigenetic modifications and other genes are reviewed. Close follow-up and strict hypertension control are required to reduce the risk of dissection. Hypertension, scoliosis and other extra-aortic signs suggesting a connective tissue disease are possible clues for diagnosis.
Collapse
|
13
|
Latorre M, Humphrey JD. Numerical knockouts-In silico assessment of factors predisposing to thoracic aortic aneurysms. PLoS Comput Biol 2020; 16:e1008273. [PMID: 33079926 PMCID: PMC7598929 DOI: 10.1371/journal.pcbi.1008273] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2020] [Revised: 10/30/2020] [Accepted: 08/19/2020] [Indexed: 02/06/2023] Open
Abstract
Myriad risk factors–including uncontrolled hypertension, aging, and diverse genetic mutations–contribute to the development and enlargement of thoracic aortic aneurysms. Detailed analyses of clinical data and longitudinal studies of murine models continue to provide insight into the natural history of these potentially lethal conditions. Yet, because of the co-existence of multiple risk factors in most cases, it has been difficult to isolate individual effects of the many different factors or to understand how they act in combination. In this paper, we use a data-informed computational model of the initiation and progression of thoracic aortic aneurysms to contrast key predisposing risk factors both in isolation and in combination; these factors include localized losses of elastic fiber integrity, aberrant collagen remodeling, reduced smooth muscle contractility, and dysfunctional mechanosensing or mechanoregulation of extracellular matrix along with superimposed hypertension and aortic aging. In most cases, mild-to-severe localized losses in cellular function or matrix integrity give rise to varying degrees of local dilatations of the thoracic aorta, with enlargement typically exacerbated in cases wherein predisposing risk factors co-exist. The simulations suggest, for the first time, that effects of compromised smooth muscle contractility are more important in terms of dysfunctional mechanosensing and mechanoregulation of matrix than in vessel-level control of diameter and, furthermore, that dysfunctional mechanobiological control can yield lesions comparable to those in cases of compromised elastic fiber integrity. Particularly concerning, therefore, is that loss of constituents such as fibrillin-1, as in Marfan syndrome, can compromise both elastic fiber integrity and mechanosensing. Aneurysms are local dilatations of the arterial wall that are responsible for significant disability and death. Detailed analyses of clinical data continue to provide insight into the natural history of these potentially lethal conditions, with myriad risk factors–including uncontrolled hypertension, aging, and diverse genetic mutations–contributing to their development and enlargement. Yet, because of the co-existence of these risk factors in most cases, it has been difficult to isolate individual effects or to understand how they act in combination. In this paper, we use a computational model of the initiation and progression of thoracic aortic aneurysms to contrast key predisposing factors both in isolation and in combination as well as with superimposed hypertension and aging. The present study recovers many findings from mouse models but with new and important observations that promise to guide in vivo and ex vivo studies as we seek to understand and eventually better treat these complex, multi-factorial lesions, with data-informed patient-specific computations eventually the way forward.
Collapse
Affiliation(s)
- M. Latorre
- Department of Biomedical Engineering, Yale University, New Haven, CT, United States of America
| | - J. D. Humphrey
- Department of Biomedical Engineering, Yale University, New Haven, CT, United States of America
- * E-mail:
| |
Collapse
|
14
|
Effects of fibrillin mutations on the behavior of heart muscle cells in Marfan syndrome. Sci Rep 2020; 10:16756. [PMID: 33028885 PMCID: PMC7542175 DOI: 10.1038/s41598-020-73802-w] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Accepted: 09/21/2020] [Indexed: 12/29/2022] Open
Abstract
Marfan syndrome (MFS) is a systemic disorder of connective tissue caused by pathogenic variants in the fibrillin-1 (FBN1) gene. Myocardial dysfunction has been demonstrated in MFS patients and mouse models, but little is known about the intrinsic effect on the cardiomyocytes (CMs). In this study, both induced pluripotent stem cells derived from a MFS-patient and the line with the corrected FBN1 mutation were differentiated to CMs. Several functional analyses are performed on this model to study MFS related cardiomyopathy. Atomic force microscopy revealed that MFS CMs are stiffer compared to corrected CMs. The contraction amplitude of MFS CMs is decreased compared to corrected CMs. Under normal culture conditions, MFS CMs show a lower beat-to-beat variability compared to corrected CMs using multi electrode array. Isoproterenol-induced stress or cyclic strain demonstrates lack of support from the matrix in MFS CMs. This study reports the first cardiac cell culture model for MFS, revealing abnormalities in the behavior of MFS CMs that are related to matrix defects. Based on these results, we postulate that impaired support from the extracellular environment plays a key role in the improper functioning of CMs in MFS.
Collapse
|
15
|
Sabbatini AR, Kararigas G. Estrogen-related mechanisms in sex differences of hypertension and target organ damage. Biol Sex Differ 2020; 11:31. [PMID: 32487164 PMCID: PMC7268741 DOI: 10.1186/s13293-020-00306-7] [Citation(s) in RCA: 89] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Accepted: 05/04/2020] [Indexed: 12/13/2022] Open
Abstract
Hypertension (HTN) is a primary risk factor for cardiovascular (CV) events, target organ damage (TOD), premature death and disability worldwide. The pathophysiology of HTN is complex and influenced by many factors including biological sex. Studies show that the prevalence of HTN is higher among adults aged 60 and over, highlighting the increase of HTN after menopause in women. Estrogen (E2) plays an important role in the development of systemic HTN and TOD, exerting several modulatory effects. The influence of E2 leads to alterations in mechanisms regulating the sympathetic nervous system, renin-angiotensin-aldosterone system, body mass, oxidative stress, endothelial function and salt sensitivity; all associated with a crucial inflammatory state and influenced by genetic factors, ultimately resulting in cardiac, vascular and renal damage in HTN. In the present article, we discuss the role of E2 in mechanisms accounting for the development of HTN and TOD in a sex-specific manner. The identification of targets with therapeutic potential would contribute to the development of more efficient treatments according to individual needs.
Collapse
Affiliation(s)
| | - Georgios Kararigas
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany.
- DZHK (German Centre for Cardiovascular Research), partner site Berlin, Berlin, Germany.
| |
Collapse
|
16
|
Altinbas L, Bormann N, Lehmann D, Jeuthe S, Wulsten D, Kornak U, Robinson PN, Wildemann B, Kararigas G. Assessment of Bones Deficient in Fibrillin-1 Microfibrils Reveals Pronounced Sex Differences. Int J Mol Sci 2019; 20:ijms20236059. [PMID: 31805661 PMCID: PMC6928642 DOI: 10.3390/ijms20236059] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2019] [Revised: 11/25/2019] [Accepted: 11/28/2019] [Indexed: 12/13/2022] Open
Abstract
Defects in the extracellular matrix protein fibrillin-1 that perturb transforming growth factor beta (TGFβ) bioavailability lead to Marfan syndrome (MFS). MFS is an autosomal-dominant disorder, which is associated with connective tissue and skeletal defects, among others. To date, it is unclear how biological sex impacts the structural and functional properties of bone in MFS. The aim of this study was to investigate the effects of sex on bone microarchitecture and mechanical properties in mice with deficient fibrillin-1, a model of human MFS. Bones of 11-week-old male and female Fbn1mgR/mgR mice were investigated. Three-dimensional micro-computed tomography of femora and vertebrae revealed a lower ratio of trabecular bone volume to tissue volume, reduced trabecular number and thickness, and greater trabecular separation in females vs. males. Three-point bending of femora revealed significantly lower post-yield displacement and work-to-fracture in females vs. males. Mechanistically, we found higher Smad2 and ERK1/2 phosphorylation in females vs. males, demonstrating a greater activation of TGFβ signaling in females. In summary, the present findings show pronounced sex differences in the matrix and function of bones deficient in fibrillin-1 microfibrils. Consequently, sex-specific analysis of bone characteristics in patients with MFS may prove useful in improving the clinical management and life quality of these patients, through the development of sex-specific therapeutic approaches.
Collapse
Affiliation(s)
- Lukas Altinbas
- BIH Center for Regenerative Therapies, Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, 13353 Berlin, Germany
| | - Nicole Bormann
- BIH Center for Regenerative Therapies, Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, 13353 Berlin, Germany
- Julius Wolff Institute for Biomechanics and Musculoskeletal Regeneration, Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, 13353 Berlin, Germany
| | - Daniel Lehmann
- BIH Center for Regenerative Therapies, Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, 13353 Berlin, Germany
| | - Sarah Jeuthe
- Max-Delbrück-Center for Molecular Medicine, 13125 Berlin, Germany
| | - Dag Wulsten
- BIH Center for Regenerative Therapies, Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, 13353 Berlin, Germany
- Julius Wolff Institute for Biomechanics and Musculoskeletal Regeneration, Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, 13353 Berlin, Germany
| | - Uwe Kornak
- BIH Center for Regenerative Therapies, Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, 13353 Berlin, Germany
- Institute for Medical Genetics and Human Genetics, Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, 13353 Berlin, Germany
| | - Peter N. Robinson
- The Jackson Laboratory for Genomic Medicine, Farmington, CT 06032, USA
| | - Britt Wildemann
- BIH Center for Regenerative Therapies, Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, 13353 Berlin, Germany
- Julius Wolff Institute for Biomechanics and Musculoskeletal Regeneration, Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, 13353 Berlin, Germany
- Experimental Trauma Surgery, University Hospital Jena, 07743 Jena, Germany
| | - Georgios Kararigas
- Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, 10117 Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, Germany
- Correspondence: ; Tel.: +49-30-450-525355
| |
Collapse
|
17
|
Wagner AH, Zaradzki M, Arif R, Remes A, Müller OJ, Kallenbach K. Marfan syndrome: A therapeutic challenge for long-term care. Biochem Pharmacol 2019; 164:53-63. [PMID: 30926475 DOI: 10.1016/j.bcp.2019.03.034] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2019] [Accepted: 03/25/2019] [Indexed: 12/14/2022]
Abstract
Marfan syndrome (MFS) is an autosomal dominant genetic disorder caused by mutations in the fibrillin-1 gene. Acute aortic dissection is the leading cause of death in patients suffering from MFS and consequence of medial degeneration and aneurysm formation. In addition to its structural function in the formation of elastic fibers, fibrillin has a major role in keeping maintaining transforming growth factor β (TGF-β) in an inactive form. Dysfunctional fibrillin increases TGF-β bioavailability and concentration in the extracellular matrix, leading to activation of proinflammatory transcription factors. In turn, these events cause increased expression of matrix metalloproteinases and cytokines that control the migration and infiltration of inflammatory cells into the aorta. Moreover, TGF-β causes accumulation of reactive oxygen species leading to further degradation of elastin fibers. All these processes result in medial elastolysis, which increases the risk of vascular complications. Although MFS is a hereditary disease, symptoms and traits are usually not noticeable at birth. During childhood or adolescence affected individuals present with severe tissue weaknesses, especially in the aorta, heart, eyes, and skeleton. Considering this, even young patients should avoid activities that exert additional stress and pressure on the aorta and the cardiovascular system. Thus, if the diagnosis is made and prophylactic treatment is initiated in a timely fashion, MFS and its preliminary pathophysiologic vascular remodeling can be successfully ameliorated reducing the risk of life-threatening complications. This commentary focuses on new research opportunities and molecular findings on MFS, discusses future challenges and possible long-term therapies.
Collapse
Affiliation(s)
- A H Wagner
- Institute of Physiology and Pathophysiology, Division of Cardiovascular Physiology, Heidelberg University, Germany.
| | - M Zaradzki
- Department of Cardiac Surgery, University Hospital Heidelberg, Heidelberg, Germany
| | - R Arif
- Department of Cardiac Surgery, University Hospital Heidelberg, Heidelberg, Germany
| | - A Remes
- Department of Internal Medicine III, University Hospital Kiel, Kiel, Germany
| | - O J Müller
- Department of Internal Medicine III, University Hospital Kiel, Kiel, Germany
| | - K Kallenbach
- INCCI HaerzZenter, Department of Cardiac Surgery, Luxembourg, Luxembourg
| |
Collapse
|
18
|
Bhushan R, Altinbas L, Jäger M, Zaradzki M, Lehmann D, Timmermann B, Clayton NP, Zhu Y, Kallenbach K, Kararigas G, Robinson PN. An integrative systems approach identifies novel candidates in Marfan syndrome-related pathophysiology. J Cell Mol Med 2019; 23:2526-2535. [PMID: 30677223 PMCID: PMC6433740 DOI: 10.1111/jcmm.14137] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2018] [Revised: 12/11/2018] [Accepted: 12/13/2018] [Indexed: 12/30/2022] Open
Abstract
Marfan syndrome (MFS) is an autosomal dominant genetic disorder caused by mutations in the FBN1 gene. Although many peripheral tissues are affected, aortic complications, such as dilation, dissection and rupture, are the leading causes of MFS‐related mortality. Aberrant TGF‐beta signalling plays a major role in the pathophysiology of MFS. However, the contributing mechanisms are still poorly understood. Here, we aimed at identifying novel aorta‐specific pathways involved in the pathophysiology of MFS. For this purpose, we employed the Fbn1 under‐expressing mgR/mgR mouse model of MFS. We performed RNA‐sequencing of aortic tissues of 9‐week‐old mgR/mgR mice compared with wild‐type (WT) mice. With a false discovery rate <5%, our analysis revealed 248 genes to be differentially regulated including 20 genes previously unrelated with MFS‐related pathology. Among these, we identified Igfbp2, Ccl8, Spp1, Mylk2, Mfap4, Dsp and H19. We confirmed the expression of regulated genes by quantitative real‐time PCR. Pathway classification revealed transcript signatures involved in chemokine signalling, cardiac muscle contraction, dilated and hypertrophic cardiomyopathy. Furthermore, our immunoblot analysis of aortic tissues revealed altered regulation of pSmad2 signalling, Perk1/2, Igfbp2, Mfap4, Ccl8 and Mylk2 protein levels in mgR/mgR vs WT mice. Together, our integrative systems approach identified several novel factors associated with MFS‐aortic‐specific pathophysiology that might offer potential novel therapeutic targets for MFS.
Collapse
Affiliation(s)
- Raghu Bhushan
- Charité University Hospital, Berlin, Germany.,Yenepoya Research Centre, Yenepoya (Deemed to be University), Deralakatte, Mangalore, India
| | | | - Marten Jäger
- Charité University Hospital, Berlin, Germany.,Berlin Institute of Health (BIH) Core Genomics Facility, Charité, University Medical Center, Berlin, Germany
| | - Marcin Zaradzki
- Department of Cardiac Surgery, University Hospital Heidelberg, Heidelberg, Germany
| | | | | | | | | | - Klaus Kallenbach
- Department of Cardiac Surgery, University Hospital Heidelberg, Heidelberg, Germany.,Department of Cardiac Surgery, INCCI HaerzZenter, Luxembourg, Luxembourg
| | - Georgios Kararigas
- Charité University Hospital, Berlin, Germany.,DZHK (German Centre for Cardiovascular Research), Berlin, Germany
| | - Peter N Robinson
- Charité University Hospital, Berlin, Germany.,Max Planck Institute for Molecular Genetics, Berlin, Germany.,The Jackson Laboratory for Genomic Medicine, Farmington, Connecticut
| |
Collapse
|