1
|
Ganjibakhsh M, Tkachenko Y, Knutsen RH, Kozel BA. Toward a rational therapeutic for elastin related disease: Key considerations for elastin based regenerative medicine strategies. Matrix Biol 2025; 138:8-21. [PMID: 40158781 DOI: 10.1016/j.matbio.2025.03.003] [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: 01/04/2025] [Revised: 03/07/2025] [Accepted: 03/23/2025] [Indexed: 04/02/2025]
Abstract
Elastin is a connective tissue protein, produced from the ELN gene, that provides elasticity and recoil to tissues that stretch, such as the large arteries of the body, lung parenchyma, skin, ligaments and elastic cartilages. It is produced as a soluble monomer, tropoelastin, that when cross-linked in the extracellular space generates a polymer that is extraordinarily stable, with a predicted half-life of >70 years. Although data suggest ongoing elastin transcription, it is rare to see new elastin deposited outside of its tight developmental window. Consequently, elastin-related disease comes about primarily in one of three scenarios: (1) inadequate elastin deposition, (2) production of poor-quality elastic fibers, or (3) increased destruction of previously deposited elastin. By understanding the pathways controlling elastin production and maintenance, we can design new therapeutics to thwart those abnormal processes. In this review, we will summarize the diseases arising from genetic and environmental alteration of elastin (Williams syndrome, supravalvar aortic stenosis, autosomal dominant cutis laxa, and ELN-related vascular and connective tissue dysfunction) and then describe the mechanisms controlling elastin production and maintenance that might be manipulated to generate novel therapeutics aimed at these conditions. We will end by summarizing existing therapeutic strategies targeting these disease mechanisms before outlining future approaches that may better solve the challenges associated with elastin based regenerative medicine.
Collapse
Affiliation(s)
- Meysam Ganjibakhsh
- Institute of Genomic Medicine, Abigail Wexler Research Institute, Nationwide Children's Hospital, OH 43205, USA
| | - Yanina Tkachenko
- Institute of Genomic Medicine, Abigail Wexler Research Institute, Nationwide Children's Hospital, OH 43205, USA
| | - Russell H Knutsen
- Institute of Genomic Medicine, Abigail Wexler Research Institute, Nationwide Children's Hospital, OH 43205, USA
| | - Beth A Kozel
- Institute of Genomic Medicine, Abigail Wexler Research Institute, Nationwide Children's Hospital, OH 43205, USA; Department of Pediatrics, The Ohio State University, OH 43210, USA.
| |
Collapse
|
2
|
van Deel ED, Snelders M, van Vliet N, Te Riet L, van den Bosch TPP, Fiedler LR, van Spreeuwel ACC, Bax NAM, Boontje N, Halabi CM, Sasaki T, Reinhardt DP, van der Velden J, Bouten CVC, von der Thüsen JH, Danser AHJ, Duncker DJ, Schneider MD, van der Pluijm I, Essers J. Induction of cardiac fibulin-4 protects against pressure overload-induced cardiac hypertrophy and heart failure. Commun Biol 2025; 8:661. [PMID: 40274989 PMCID: PMC12022050 DOI: 10.1038/s42003-025-08087-8] [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: 05/13/2024] [Accepted: 04/14/2025] [Indexed: 04/26/2025] Open
Abstract
The prevailing view of fibulin-4 deficient mice is that the cardiac phenotype is the result of aortic and/or valvular disease. In the present study, we have tested whether the cardiac phenotype is, at least in part, the consequence of primary cardiac effects of fibulin-4. We have found fibulin-4 expression to be activated throughout the myocardium in wildtype (fibulin-4+/+) C57Bl/6J;129 Sv mice subjected to transverse aortic constriction (TAC). In contrast, haploinsufficient fibulin-4+/R mice exposed to severe TAC do not show this increase in myocardial fibulin-4 expression, but display altered physical properties of myocardial tissue. Moreover, TAC-induced cardiac fibrosis, pulmonary congestion, and mortality are aggravated in fibulin-4+/R mice. In vitro investigations of myocardial tissue show that fibulin-4 deficiency results in cardiomyocyte hypertrophy, and a decreased beating frequency and contractile force. In conclusion, we demonstrate functions for fibulin-4 in cardiac homeostasis and show that reduced fibulin-4 expression drives myocardial disease in response to cardiac pressure overload, independent of aortic valvular pathology.
Collapse
Affiliation(s)
- E D van Deel
- Department of Molecular Genetics, Erasmus University Medical Center, Rotterdam, the Netherlands
- Division of Experimental Cardiology, Department of Cardiology, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - M Snelders
- Department of Molecular Genetics, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - N van Vliet
- Department of Molecular Genetics, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - L Te Riet
- Division of Pharmacology, Department of Internal Medicine, Erasmus University Medical Center, Rotterdam, the Netherlands
- Department of Vascular Surgery, Cardiovascular Institute, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - T P P van den Bosch
- Department of Pathology and Clinical Bioinformatics, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - L R Fiedler
- British Heart Foundation Centre of Research Excellence, National Heart and Lung Institute, Imperial College London, London, UK
| | - A C C van Spreeuwel
- Department of Biomedical Engineering, and Institute for Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, the Netherlands
| | - N A M Bax
- Department of Biomedical Engineering, and Institute for Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, the Netherlands
| | - N Boontje
- Department of Physiology, VU University Medical Center, Institute for Cardiovascular Research (ICaR-VU), Amsterdam, the Netherlands
| | - C M Halabi
- Division of Nephrology, Department of Pediatrics, Washington University School of Medicine, St. Louis, MO, USA
| | - T Sasaki
- Department of Pharmacology, Faculty of Medicine, Oita University, Oita, Japan
| | - D P Reinhardt
- Faculty of Medicine and Health Sciences, McGill University, Montreal, QC, Canada
- Faculty of Dentistry and Oral Health Sciences, McGill University, Montreal, QC, Canada
| | - J van der Velden
- Department of Physiology, VU University Medical Center, Institute for Cardiovascular Research (ICaR-VU), Amsterdam, the Netherlands
| | - C V C Bouten
- Department of Biomedical Engineering, and Institute for Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, the Netherlands
| | - J H von der Thüsen
- Department of Pathology and Clinical Bioinformatics, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - A H J Danser
- Division of Pharmacology, Department of Internal Medicine, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - D J Duncker
- Division of Experimental Cardiology, Department of Cardiology, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - M D Schneider
- British Heart Foundation Centre of Research Excellence, National Heart and Lung Institute, Imperial College London, London, UK
| | - I van der Pluijm
- Department of Molecular Genetics, Erasmus University Medical Center, Rotterdam, the Netherlands
- Department of Vascular Surgery, Cardiovascular Institute, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - J Essers
- Department of Molecular Genetics, Erasmus University Medical Center, Rotterdam, the Netherlands.
- Department of Vascular Surgery, Cardiovascular Institute, Erasmus University Medical Center, Rotterdam, the Netherlands.
- Department of Radiotherapy, Erasmus University Medical Center, Rotterdam, the Netherlands.
| |
Collapse
|
3
|
Kailash KA, Akanda SR, Davis AL, Crandall CL, Castro LA, Setton LA, Wagenseil JE. A multiphasic model for determination of mouse ascending thoracic aorta mass transport properties with and without aneurysm. Biomech Model Mechanobiol 2025; 24:93-105. [PMID: 39470949 DOI: 10.1007/s10237-024-01897-5] [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: 05/08/2024] [Accepted: 10/10/2024] [Indexed: 11/01/2024]
Abstract
Thoracic aortic aneurysms (TAAs) are associated with aortic wall remodeling that affects transmural transport or the movement of fluid and solute across the wall. In previous work, we used a Fbln4E57K/E57K (MU) mouse model to investigate transmural transport changes as a function of aneurysm severity. We compared wild-type (WT), MU with no aneurysm (MU-NA), MU with aneurysm (MU-A), and MU with an additional genetic mutation that led to increased aneurysm penetrance (MU-XA). We found that all aneurysmal aortas (MU-A and MU-XA) had lower fluid flux compared to WT. Non-aneurysmal aortas (MU-NA) had higher 4 kDa FITC-dextran solute flux than WT, but aneurysmal MU-A and MU-XA aortas had solute fluxes similar to WT. Our experimental results could not isolate competing factors, such as changes in aortic geometry and solid material properties among these mouse models, to determine how intrinsic transport properties change with aneurysm severity. The objective of this study is to use biphasic and multiphasic models to identify changes in transport material properties. Our biphasic model indicates that hydraulic permeability is significantly decreased in the severe aneurysm model (MU-XA) compared to non-aneurysmal aortas (MU-NA). Our multiphasic model shows that effective solute diffusivity is increased in MU-NA aortas compared to all others. Our findings reveal changes in intrinsic transport properties that depend on aneurysm severity and are important for understanding the movement of fluids and solutes that may play a role in the diagnosis, progression, or treatment of TAA.
Collapse
Affiliation(s)
- Keshav A Kailash
- Department of Biomedical Engineering, Washington University, St. Louis, MO, USA
| | - Shamimur R Akanda
- Department of Biomedical Engineering, Washington University, St. Louis, MO, USA
| | - Alexandra L Davis
- Department of Biomedical Engineering, Washington University, St. Louis, MO, USA
| | - Christie L Crandall
- Department of Mechanical Engineering and Materials Science, Washington University, One Brookings Dr., CB 1185, St. Louis, MO, 63130, USA
| | - Luis A Castro
- Department of Mechanical Engineering and Materials Science, Washington University, One Brookings Dr., CB 1185, St. Louis, MO, 63130, USA
| | - Lori A Setton
- Department of Biomedical Engineering, Washington University, St. Louis, MO, USA
- Department of Orthopedic Surgery, Washington University, St. Louis, MO, USA
- Department of Mechanical Engineering and Materials Science, Washington University, One Brookings Dr., CB 1185, St. Louis, MO, 63130, USA
| | - Jessica E Wagenseil
- Department of Mechanical Engineering and Materials Science, Washington University, One Brookings Dr., CB 1185, St. Louis, MO, 63130, USA.
| |
Collapse
|
4
|
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
|
5
|
Yao G, Zheng X, Hu Y, Zhao Y, Kong B, Ti Y, Bu PL. FBLN7 mediates vascular smooth muscle cell phenotype switching and vascular remodeling in hypertension. Theranostics 2024; 14:7569-7588. [PMID: 39659565 PMCID: PMC11626941 DOI: 10.7150/thno.102593] [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: 08/20/2024] [Accepted: 10/26/2024] [Indexed: 12/12/2024] Open
Abstract
Rationale: Arterial remodeling serves as a pivotal mechanism underlying the development of diseases such as hypertension. Fibulin-7 (FBLN7), an adhesion protein, remains enigmatic regarding its role in these pathological processes. This study aims to explore whether FBLN7 influences vascular remodeling and its underlying mechanisms. Methods: We generated FBLN7 knockout mice and smooth muscle-specific FBLN7 overexpression mice. Vascular remodeling models were established by administering angiotensin II (Ang II) for 28 days. RNA sequencing, western blot, and immunofluorescence assays were employed to investigate the biological function of FBLN7 in vascular smooth muscle cells (VSMCs). The interaction mechanism between FBLN7 and cell membrane receptors was explored through mass spectrometry analysis, co-immunoprecipitation techniques and molecular dynamics simulations. Results: Bioinformatics analysis revealed an upregulation of FBLN7 expression in the vascular remodeling model, with FBLN7 predominantly localized in VSMCs. Subsequent in vivo validation demonstrated that FBLN7 knockout attenuated Ang II-induced vascular remodeling, reducing aortic wall thickness and collagen formation. Conversely, VSMC-specific overexpression of FBLN7 via AAV vectors exacerbating the remodeling phenotype. Functionally speaking, FBLN7 potentiates Ang II-mediated phenotypic transformation. Mechanistically, FBLN7 interacts with the extracellular and transmembrane domains of syndecan-4 (SDC4) via its C-terminal region, affecting SDC4 signaling and dimer formation. This interaction inhibits SDC4-mediated activation of the Rho-associated protein kinase pathway, subsequently reducing nuclear translocation of myocardin-related transcription factor A, leading to decreased transcription of genes associated with the contractile VSMCs phenotype. Conclusions: These findings reveal FBLN7 promotes the transition of VSMCs from a contractile to a synthetic phenotype, thereby aggravating vascular remodeling. This provides further insights into the pathogenesis of vascular remodeling and potential therapeutic strategies.
Collapse
Affiliation(s)
| | | | | | | | | | - Yun Ti
- State Key Laboratory for Innovation and Transformation of Luobing Theory; Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences; Department of Cardiology, Qilu Hospital of Shandong University, Jinan, 250012, China
| | - Pei li Bu
- State Key Laboratory for Innovation and Transformation of Luobing Theory; Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences; Department of Cardiology, Qilu Hospital of Shandong University, Jinan, 250012, China
| |
Collapse
|
6
|
Kabir AU, Zeng C, Subramanian M, Wu J, Kim M, Krchma K, Wang X, Halabi CM, Pan H, Wickline SA, Fremont DH, Artyomov MN, Choi K. ZBTB46 coordinates angiogenesis and immunity to control tumor outcome. Nat Immunol 2024; 25:1546-1554. [PMID: 39134750 DOI: 10.1038/s41590-024-01936-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2023] [Accepted: 07/16/2024] [Indexed: 09/01/2024]
Abstract
Tumor angiogenesis and immunity show an inverse correlation in cancer progression and outcome1. Here, we report that ZBTB46, a repressive transcription factor and a widely accepted marker for classical dendritic cells (DCs)2,3, controls both tumor angiogenesis and immunity. Zbtb46 was downregulated in both DCs and endothelial cells by tumor-derived factors to facilitate robust tumor growth. Zbtb46 downregulation led to a hallmark pro-tumor microenvironment (TME), including dysfunctional vasculature and immunosuppressive conditions. Analysis of human cancer data revealed a similar association of low ZBTB46 expression with an immunosuppressive TME and a worse prognosis. In contrast, enforced Zbtb46 expression led to TME changes to restrict tumor growth. Mechanistically, Zbtb46-deficient endothelial cells were highly angiogenic, and Zbtb46-deficient bone marrow progenitors upregulated Cebpb and diverted the DC program to immunosuppressive myeloid lineage output, potentially explaining the myeloid lineage skewing phenomenon in cancer4. Conversely, enforced Zbtb46 expression normalized tumor vessels and, by suppressing Cebpb, skewed bone marrow precursors toward immunostimulatory myeloid lineage output, leading to an immune-hot TME. Remarkably, Zbtb46 mRNA treatment synergized with anti-PD1 immunotherapy to improve tumor management in preclinical models. These findings identify ZBTB46 as a critical factor for angiogenesis and for myeloid lineage skewing in cancer and suggest that maintaining its expression could have therapeutic benefits.
Collapse
Affiliation(s)
- Ashraf Ul Kabir
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, USA
| | - Carisa Zeng
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, USA
| | - Madhav Subramanian
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, USA
| | - Jun Wu
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, USA
| | - Minseo Kim
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, USA
| | - Karen Krchma
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, USA
| | - Xiaoli Wang
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, USA
| | - Carmen M Halabi
- Department of Pediatrics, Washington University School of Medicine, St. Louis, MO, USA
| | - Hua Pan
- Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Samuel A Wickline
- Health Heart Institute, Morsani College of Medicine, University of South Florida, Tampa, FL, USA
| | - Daved H Fremont
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, USA
| | - Maxim N Artyomov
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, USA
| | - Kyunghee Choi
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, USA.
| |
Collapse
|
7
|
Used-Gavín A, Larrañaga-Moreira JM, Lago-Cascudo R, Mosquera-Rodríguez VX, Barriales-Villa R. Giant ascending aortic aneurysm with impending rupture as presentation of cutis laxa 1B: a case report. Eur Heart J Case Rep 2023; 7:ytad530. [PMID: 38025136 PMCID: PMC10660401 DOI: 10.1093/ehjcr/ytad530] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Revised: 10/15/2023] [Accepted: 10/25/2023] [Indexed: 12/01/2023]
Abstract
Background Thoracic aortic aneurysms are rarely symptomatic but can result in acute aortic syndromes, associated with a high mortality rate. While most cases may be acquired, a genetic basis is evident in approximately 20-25% of the cases, especially among patients under 50 years of age, and those exhibiting syndromic features or family history. Although autosomal dominant inheritance is predominant in familial aortopathies, exceptions exist, such as cutis laxa 1B (CL1B)-related aortic disease, caused by variants in EFEMP2 gene, that follows an autosomal recessive inheritance pattern. Case summary We present the case of a 26-year-old male with a giant ascending aorta aneurysm and massive pericardial effusion, which was ultimately diagnosed of CL1B due to the p.Ser137Cys variant in the EFEMP2 gene in homozygosis. The patient underwent successful ascending aorta replacement (Bentall´s procedure). There were not complications or further events after 2 years of follow-up. Discussion This case underscores the importance of genetic testing in young patients presenting with aortopathies, syndromic features, or atypical presentations, irrespective of family history.
Collapse
Affiliation(s)
- Alejandro Used-Gavín
- Inherited Cardiovascular Diseases Unit, Hospital Universitario A Coruña (HUAC), As Xubias 84, 8th floor, 15006 A Coruña, Spain
- Instituto de Investigación Biomédica de A Coruña (INIBIC), Universidade da Coruña (UDC), Servizo Galego de Saúde (SERGAS), As Xubias 84, 15006 A Coruña, Spain
| | - José María Larrañaga-Moreira
- Inherited Cardiovascular Diseases Unit, Hospital Universitario A Coruña (HUAC), As Xubias 84, 8th floor, 15006 A Coruña, Spain
- Instituto de Investigación Biomédica de A Coruña (INIBIC), Universidade da Coruña (UDC), Servizo Galego de Saúde (SERGAS), As Xubias 84, 15006 A Coruña, Spain
| | - Rafael Lago-Cascudo
- Instituto de Investigación Biomédica de A Coruña (INIBIC), Universidade da Coruña (UDC), Servizo Galego de Saúde (SERGAS), As Xubias 84, 15006 A Coruña, Spain
- Pathology Department, Hospital Universitario A Coruña (HUAC), A Coruña, Spain
| | - Víctor X Mosquera-Rodríguez
- Instituto de Investigación Biomédica de A Coruña (INIBIC), Universidade da Coruña (UDC), Servizo Galego de Saúde (SERGAS), As Xubias 84, 15006 A Coruña, Spain
- Cardiac Surgery Department, Hospital Universitario A Coruña (HUAC), As Xubias 84, 8th floor, 15006 A Coruña, Spain
| | - Roberto Barriales-Villa
- Inherited Cardiovascular Diseases Unit, Hospital Universitario A Coruña (HUAC), As Xubias 84, 8th floor, 15006 A Coruña, Spain
- Instituto de Investigación Biomédica de A Coruña (INIBIC), Universidade da Coruña (UDC), Servizo Galego de Saúde (SERGAS), As Xubias 84, 15006 A Coruña, Spain
- Centro de Investigación Biomédica en Red (CIBERCV), Av. Monforte de Lemos, 3-5, 28029 Madrid, España
| |
Collapse
|
8
|
Tanino R, Tsubata Y, Hotta T, Okimoto T, Amano Y, Takechi M, Tanaka T, Akita T, Nagase M, Yamashita C, Wada K, Isobe T. Characterization of a spontaneous mouse model of mild, accelerated aging via ECM degradation in emphysematous lungs. Sci Rep 2023; 13:10740. [PMID: 37400563 DOI: 10.1038/s41598-023-37638-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Accepted: 06/25/2023] [Indexed: 07/05/2023] Open
Abstract
Emphysema limits airflow and causes irreversible progression of chronic obstructive pulmonary disease (COPD). Strain differences must be considered when selecting mouse models of COPD, owing to disease complexity. We previously reported that a novel C57BL/6JJcl substrain, the Mayumi-Emphysema (ME) mouse, exhibits spontaneous emphysema; however, the other characteristics remain unknown. We aimed to characterize the lungs of ME mice and determine their experimental availability as a model. ME mice had a lower body weight than the control C57BL/6JJcl mice, with a median survival time of ~80 weeks. ME mice developed diffused emphysema with respiratory dysfunction from 8 to 26 weeks of age, but did not develop bronchial wall thickening. Proteomic analyses revealed five extracellular matrix-related clusters in downregulated lung proteins in ME mice. Moreover, EFEMP2/fibulin-4, an essential extracellular matrix protein, was the most downregulated protein in the lungs of ME mice. Murine and human EFEMP2 were detected in the pulmonary artery. Furthermore, patients with mild COPD showed decreased EFEMP2 levels in the pulmonary artery when compared to those without COPD. The ME mouse is a model of mild, accelerated aging with low-inflammatory emphysema and respiratory dysfunction that progresses with age and pulmonary EFEMP2 decrease, similar to that observed in patients with mild COPD.
Collapse
Affiliation(s)
- Ryosuke Tanino
- Department of Internal Medicine, Division of Respiratory Medicine and Medical Oncology, Faculty of Medicine, Shimane University, 89-1 Enya, Izumo, Shimane, 693-8501, Japan
| | - Yukari Tsubata
- Department of Internal Medicine, Division of Respiratory Medicine and Medical Oncology, Faculty of Medicine, Shimane University, 89-1 Enya, Izumo, Shimane, 693-8501, Japan.
| | - Takamasa Hotta
- Department of Internal Medicine, Division of Respiratory Medicine and Medical Oncology, Faculty of Medicine, Shimane University, 89-1 Enya, Izumo, Shimane, 693-8501, Japan
| | - Tamio Okimoto
- Department of Internal Medicine, Division of Respiratory Medicine and Medical Oncology, Faculty of Medicine, Shimane University, 89-1 Enya, Izumo, Shimane, 693-8501, Japan
| | - Yoshihiro Amano
- Department of Internal Medicine, Division of Respiratory Medicine and Medical Oncology, Faculty of Medicine, Shimane University, 89-1 Enya, Izumo, Shimane, 693-8501, Japan
| | - Mayumi Takechi
- Department of Experimental Animals, Interdisciplinary Center for Science Research, Organization for Research and Academic Information, Shimane University, Izumo, Japan
| | - Tetsuya Tanaka
- Department of Human Nutrition, Faculty of Contemporary Life Science, Chugoku Gakuen University, Okayama, Japan
| | - Tomomi Akita
- Department of Pharmaceutics and Drug Delivery, Faculty of Pharmaceutical Sciences, Tokyo University of Science, Noda, Japan
| | - Mamiko Nagase
- Department of Organ Pathology, Faculty of Medicine, Shimane University, Izumo, Japan
| | - Chikamasa Yamashita
- Department of Pharmaceutics and Drug Delivery, Faculty of Pharmaceutical Sciences, Tokyo University of Science, Noda, Japan
| | - Koichiro Wada
- Department of Pharmacology, Faculty of Medicine, Shimane University, Izumo, Japan
| | - Takeshi Isobe
- Department of Internal Medicine, Division of Respiratory Medicine and Medical Oncology, Faculty of Medicine, Shimane University, 89-1 Enya, Izumo, Shimane, 693-8501, Japan
| |
Collapse
|
9
|
Crandall CL, Wu Y, Kailash KA, Bersi MR, Halabi CM, Wagenseil JE. Changes in transmural mass transport correlate with ascending thoracic aortic aneurysm diameter in a fibulin-4 E57K knockin mouse model. Am J Physiol Heart Circ Physiol 2023; 325:H113-H124. [PMID: 37267118 PMCID: PMC10292979 DOI: 10.1152/ajpheart.00036.2023] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Revised: 05/22/2023] [Accepted: 05/25/2023] [Indexed: 06/04/2023]
Abstract
Thoracic aortic aneurysm is characterized by dilation of the aortic diameter by greater than 50%, which can lead to dissection or rupture. Common histopathology includes extracellular matrix remodeling that may affect transmural mass transport, defined as the movement of fluids and solutes across the wall. We measured in vitro ascending thoracic aorta mass transport in a mouse model with partial aneurysm phenotype penetration due to a mutation in the extracellular matrix protein fibulin-4 [Fbln4E57K/E57K, referred to as MU-A (aneurysm) or MU-NA (no aneurysm)]. To push the aneurysm phenotype, we also included MU mice with reduced levels of lysyl oxidase [Fbln4E57K/E57K;Lox+/-, referred to as MU-XA (extreme aneurysm)] and compared all groups to wild-type (WT) littermates. The phenotype variation allows investigation of how aneurysm severity correlates with mass transport parameters and extracellular matrix organization. We found that MU-NA ascending thoracic aortae have similar hydraulic conductance (Lp) to WT, but 397% higher solute permeability (ω) for 4 kDa FITC-dextran. In contrast, MU-A and MU-XA ascending thoracic aortae have 44-68% lower Lp and similar ω to WT. The results suggest that ascending thoracic aortic aneurysm progression involves an initial increase in ω, followed by a decrease in Lp after the aneurysm has formed. All MU ascending thoracic aortae are longer and have increased elastic fiber fragmentation in the extracellular matrix. There is a negative correlation between diameter and Lp or ω in MU ascending thoracic aortae. Changes in mass transport due to elastic fiber fragmentation could contribute to aneurysm progression or be leveraged for treatment.NEW & NOTEWORTHY Transmural mass transport is quantified in the ascending thoracic aorta of mice with a mutation in fibulin-4 that is associated with thoracic aortic aneurysms. Fluid and solute transport depend on aneurysm severity, correlate with elastic fiber fragmentation, and may be affected by proteoglycan deposition. Transport properties of the ascending thoracic aorta are provided and can be used in computational models. The changes in mass transport may contribute to aneurysm progression or be leveraged for aneurysm treatment.
Collapse
Affiliation(s)
- Christie L Crandall
- Department of Mechanical Engineering and Materials Science, Washington University, St. Louis, Missouri, United States
| | - Yufan Wu
- Department of Mechanical Engineering and Materials Science, Washington University, St. Louis, Missouri, United States
| | - Keshav A Kailash
- Department of Biomedical Engineering, Washington University, St. Louis, Missouri, United States
| | - Mathew R Bersi
- Department of Mechanical Engineering and Materials Science, Washington University, St. Louis, Missouri, United States
| | - Carmen M Halabi
- Pediatric Nephrology, Washington University School of Medicine, St. Louis, Missouri, United States
| | - Jessica E Wagenseil
- Department of Mechanical Engineering and Materials Science, Washington University, St. Louis, Missouri, United States
| |
Collapse
|
10
|
Brengle BM, Lin M, Roth RA, Jones KD, Wagenseil JE, Mecham RP, Halabi CM. A new mouse model of elastin haploinsufficiency highlights the importance of elastin to vascular development and blood pressure regulation. Matrix Biol 2023; 117:1-14. [PMID: 36773748 PMCID: PMC12074569 DOI: 10.1016/j.matbio.2023.02.003] [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: 10/11/2022] [Revised: 01/09/2023] [Accepted: 02/07/2023] [Indexed: 02/11/2023]
Abstract
Supravalvular aortic stenosis (SVAS) is an autosomal dominant disease resulting from elastin (ELN) haploinsufficiency. Individuals with SVAS typically develop a thickened arterial media with an increased number of elastic lamellae and smooth muscle cell (SMC) layers and stenosis superior to the aortic valve. A mouse model of SVAS (Eln+/-) was generated that recapitulates many aspects of the human disease, including increased medial SMC layers and elastic lamellae, large artery stiffness, and hypertension. The vascular changes in these mice were thought to be responsible for the hypertension phenotype. However, a renin gene (Ren) duplication in the original 129/Sv genetic background and carried through numerous strain backcrosses raised the possibility of renin-mediated effects on blood pressure. To exclude excess renin activity as a disease modifier, we utilized the Cre-LoxP system to rederive Eln hemizygous mice on a pure C57BL/6 background (Sox2-Cre;Elnf/f). Here we show that Sox2-Cre;Eln+/f mice, with a single Ren1 gene and normal renin levels, phenocopy the original global knockout line. Characteristic traits include an increased number of elastic lamellae and SMC layers, stiff elastic arteries, and systolic hypertension with widened pulse pressure. Importantly, small resistance arteries of Sox2-Cre;Eln+/f mice exhibit a significant change in endothelial cell function and hypercontractility to angiotensin II, findings that point to pathway-specific alterations in resistance arteries that contribute to the hypertensive phenotype. These data confirm that the cardiovascular changes, particularly systolic hypertension, seen in Eln+/- mice are due to Eln hemizygosity rather than Ren duplication.
Collapse
Affiliation(s)
- Bridget M Brengle
- Department of Pediatrics, Division of Nephrology, Washington University School of Medicine, St. Louis, MO, USA
| | - Michelle Lin
- Department of Pediatrics, Division of Nephrology, Washington University School of Medicine, St. Louis, MO, USA
| | - Robyn A Roth
- Department of Pediatrics, Division of Nephrology, Washington University School of Medicine, St. Louis, MO, USA
| | - Kara D Jones
- Department of Pediatrics, Division of Nephrology, Washington University School of Medicine, St. Louis, MO, USA
| | - Jessica E Wagenseil
- Department of Mechanical Engineering and Materials Science, Washington University in St. Louis, St. Louis, MO, USA
| | - Robert P Mecham
- Department of Cell Biology and Physiology, Washington University School of Medicine, St. Louis, MO, U.S.A
| | - Carmen M Halabi
- Department of Pediatrics, Division of Nephrology, Washington University School of Medicine, St. Louis, MO, USA.
| |
Collapse
|
11
|
Salarian M, Ghim M, Toczek J, Han J, Weiss D, Spronck B, Ramachandra AB, Jung JJ, Kukreja G, Zhang J, Lakheram D, Kim SK, Humphrey JD, Sadeghi MM. Homeostatic, Non-Canonical Role of Macrophage Elastase in Vascular Integrity. Circ Res 2023; 132:432-448. [PMID: 36691905 PMCID: PMC9930896 DOI: 10.1161/circresaha.122.322096] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
BACKGROUND Matrix metalloproteinase (MMP)-12 is highly expressed in abdominal aortic aneurysms and its elastolytic function has been implicated in the pathogenesis. This concept is challenged, however, by conflicting data. Here, we sought to revisit the role of MMP-12 in abdominal aortic aneurysm. METHODS Apoe-/- and Mmp12-/-/Apoe-/- mice were infused with Ang II (angiotensin). Expression of neutrophil extracellular traps (NETs) markers and complement component 3 (C3) levels were evaluated by immunostaining in aortas of surviving animals. Plasma complement components were analyzed by immunoassay. The effects of a complement inhibitor, IgG-FH1-5 (factor H-immunoglobulin G), and macrophage-specific MMP-12 deficiency on adverse aortic remodeling and death from rupture in Ang II-infused mice were determined. RESULTS Unexpectedly, death from aortic rupture was significantly higher in Mmp12-/-/Apoe-/- mice. This associated with more neutrophils, citrullinated histone H3 and neutrophil elastase, markers of NETs, and C3 levels in Mmp12-/- aortas. These findings were recapitulated in additional models of abdominal aortic aneurysm. MMP-12 deficiency also led to more pronounced elastic laminae degradation and reduced collagen integrity. Higher plasma C5a in Mmp12-/- mice pointed to complement overactivation. Treatment with IgG-FH1-5 decreased aortic wall NETosis and reduced adverse aortic remodeling and death from rupture in Ang II-infused Mmp12-/- mice. Finally, macrophage-specific MMP-12 deficiency recapitulated the effects of global MMP-12 deficiency on complement deposition and NETosis, as well as adverse aortic remodeling and death from rupture in Ang II-infused mice. CONCLUSIONS An MMP-12 deficiency/complement activation/NETosis pathway compromises aortic integrity, which predisposes to adverse vascular remodeling and abdominal aortic aneurysm rupture. Considering these new findings, the role of macrophage MMP-12 in vascular homeostasis demands re-evaluation of MMP-12 function in diverse settings.
Collapse
Affiliation(s)
- Mani Salarian
- Section of Cardiovascular Medicine and Cardiovascular Research Center, Yale School of Medicine, New Haven, CT (M.S., M.G., J.T., J.H., J.-J.J., G.K., J.Z., M.M.S.)
- VA Connecticut Healthcare System, West Haven, CT (M.S., M.G., J.T., J.H., J.-J.J., G.K., J.Z., M.M.S.)
| | - Mean Ghim
- Section of Cardiovascular Medicine and Cardiovascular Research Center, Yale School of Medicine, New Haven, CT (M.S., M.G., J.T., J.H., J.-J.J., G.K., J.Z., M.M.S.)
- VA Connecticut Healthcare System, West Haven, CT (M.S., M.G., J.T., J.H., J.-J.J., G.K., J.Z., M.M.S.)
| | - Jakub Toczek
- Section of Cardiovascular Medicine and Cardiovascular Research Center, Yale School of Medicine, New Haven, CT (M.S., M.G., J.T., J.H., J.-J.J., G.K., J.Z., M.M.S.)
- VA Connecticut Healthcare System, West Haven, CT (M.S., M.G., J.T., J.H., J.-J.J., G.K., J.Z., M.M.S.)
| | - Jinah Han
- Section of Cardiovascular Medicine and Cardiovascular Research Center, Yale School of Medicine, New Haven, CT (M.S., M.G., J.T., J.H., J.-J.J., G.K., J.Z., M.M.S.)
- VA Connecticut Healthcare System, West Haven, CT (M.S., M.G., J.T., J.H., J.-J.J., G.K., J.Z., M.M.S.)
| | - Dar Weiss
- Department of Biomedical Engineering, Yale University, New Haven, CT (D.W., B.S., A.B.R., J.D.H.)
| | - Bart Spronck
- Department of Biomedical Engineering, Yale University, New Haven, CT (D.W., B.S., A.B.R., J.D.H.)
| | - Abhay B. Ramachandra
- Department of Biomedical Engineering, Yale University, New Haven, CT (D.W., B.S., A.B.R., J.D.H.)
| | - Jae-Joon Jung
- Section of Cardiovascular Medicine and Cardiovascular Research Center, Yale School of Medicine, New Haven, CT (M.S., M.G., J.T., J.H., J.-J.J., G.K., J.Z., M.M.S.)
- VA Connecticut Healthcare System, West Haven, CT (M.S., M.G., J.T., J.H., J.-J.J., G.K., J.Z., M.M.S.)
| | - Gunjan Kukreja
- Section of Cardiovascular Medicine and Cardiovascular Research Center, Yale School of Medicine, New Haven, CT (M.S., M.G., J.T., J.H., J.-J.J., G.K., J.Z., M.M.S.)
- VA Connecticut Healthcare System, West Haven, CT (M.S., M.G., J.T., J.H., J.-J.J., G.K., J.Z., M.M.S.)
| | - Jiasheng Zhang
- Section of Cardiovascular Medicine and Cardiovascular Research Center, Yale School of Medicine, New Haven, CT (M.S., M.G., J.T., J.H., J.-J.J., G.K., J.Z., M.M.S.)
- VA Connecticut Healthcare System, West Haven, CT (M.S., M.G., J.T., J.H., J.-J.J., G.K., J.Z., M.M.S.)
| | | | - Sung-Kwon Kim
- Alexion Pharmaceuticals, New Haven, CT (D.L., S.-K.K.)
| | - Jay D. Humphrey
- Department of Biomedical Engineering, Yale University, New Haven, CT (D.W., B.S., A.B.R., J.D.H.)
- Vascular Biology and Therapeutics Program, Yale School of Medicine, New Haven, CT (J.D.H.)
| | - Mehran M. Sadeghi
- Section of Cardiovascular Medicine and Cardiovascular Research Center, Yale School of Medicine, New Haven, CT (M.S., M.G., J.T., J.H., J.-J.J., G.K., J.Z., M.M.S.)
- VA Connecticut Healthcare System, West Haven, CT (M.S., M.G., J.T., J.H., J.-J.J., G.K., J.Z., M.M.S.)
| |
Collapse
|
12
|
Nguyen TAV, Lino CA, Hang HT, Alves JV, Thang BQ, Shin SJ, Sugiyama K, Matsunaga H, Takeyama H, Yamashiro Y, Yanagisawa H. Protective Role of Endothelial Fibulin-4 in Valvulo-Arterial Integrity. J Am Heart Assoc 2022; 12:e026942. [PMID: 36565192 PMCID: PMC9973605 DOI: 10.1161/jaha.122.026942] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Background Homeostasis of the vessel wall is cooperatively maintained by endothelial cells (ECs), smooth muscle cells, and adventitial fibroblasts. The genetic deletion of fibulin-4 (Fbln4) in smooth muscle cells (SMKO) leads to the formation of thoracic aortic aneurysms with the disruption of elastic fibers. Although Fbln4 is expressed in the entire vessel wall, its function in ECs and relevance to the maintenance of valvulo-arterial integrity are not fully understood. Methods and Results Gene silencing of FBLN4 was conducted on human aortic ECs to evaluate morphological changes and gene expression profile. Fbln4 double knockout (DKO) mice in ECs and smooth muscle cells were generated and subjected to histological analysis, echocardiography, Western blotting, RNA sequencing, and immunostaining. An evaluation of the thoracic aortic aneurysm phenotype and screening of altered signaling pathways were performed. Knockdown of FBLN4 in human aortic ECs induced mesenchymal cell-like changes with the upregulation of mesenchymal genes, including TAGLN and MYL9. DKO mice showed the exacerbation of thoracic aortic aneurysms when compared with those of SMKO and upregulated Thbs1, a mechanical stress-responsive molecule, throughout the aorta. DKO mice also showed progressive aortic valve thickening with collagen deposition from postnatal day 14, as well as turbulent flow in the ascending aorta. Furthermore, RNA sequencing and immunostaining of the aortic valve revealed the upregulation of genes involved in endothelial-to-mesenchymal transition, inflammatory response, and tissue fibrosis in DKO valves and the presence of activated valve interstitial cells. Conclusions The current study uncovers the pivotal role of endothelial fibulin-4 in the maintenance of valvulo-arterial integrity, which influences thoracic aortic aneurysm progression.
Collapse
Affiliation(s)
- Tram Anh Vu Nguyen
- Life Science Center for Survival Dynamics, Tsukuba Advanced Research AllianceUniversity of TsukubaIbarakiJapan,Ph.D. Program in Human Biology, School of Integrative and Global MajorsUniversity of TsukubaIbarakiJapan
| | - Caroline Antunes Lino
- Department of AnatomyUniversity of Sao Paulo, Institute of Biomedical SciencesSao PauloBrazil
| | - Huynh Thuy Hang
- Life Science Center for Survival Dynamics, Tsukuba Advanced Research AllianceUniversity of TsukubaIbarakiJapan,Graduate School of Comprehensive Human SciencesUniversity of TsukubaIbarakiJapan
| | - Juliano Vilela Alves
- Department of PharmacologyUniversity of Sao Paulo, Ribeirao Preto Medical SchoolRibeirao PretoBrazil
| | - Bui Quoc Thang
- Life Science Center for Survival Dynamics, Tsukuba Advanced Research AllianceUniversity of TsukubaIbarakiJapan,Deputy Head of Scientific Research Department‐ Training center, Cho Ray hospitalHo Chi Minh CityVietnam
| | - Seung Jae Shin
- Life Science Center for Survival Dynamics, Tsukuba Advanced Research AllianceUniversity of TsukubaIbarakiJapan,Graduate School of Life and Environmental SciencesUniversity of TsukubaIbarakiJapan
| | - Kaori Sugiyama
- Life Science Center for Survival Dynamics, Tsukuba Advanced Research AllianceUniversity of TsukubaIbarakiJapan,Institute for Advanced Research of Biosystem Dynamics, Waseda Research Institute for Science and Engineering, Waseda UniversityTokyoJapan
| | - Hiroko Matsunaga
- Research organization for Nano and Life InnovationWaseda UniversityTokyoJapan
| | - Haruko Takeyama
- Institute for Advanced Research of Biosystem Dynamics, Waseda Research Institute for Science and Engineering, Waseda UniversityTokyoJapan,Research organization for Nano and Life InnovationWaseda UniversityTokyoJapan,Department of Life Science and Medical BioscienceWaseda UniversityTokyoJapan,Computational Bio Big‐Data Open Innovation LaboratoryAIST‐Waseda UniversityTokyoJapan
| | - Yoshito Yamashiro
- Life Science Center for Survival Dynamics, Tsukuba Advanced Research AllianceUniversity of TsukubaIbarakiJapan,Present address:
Department of Advanced Medical TechnologiesNational Cerebral and Cardiovascular Center Research InstituteOsaka564‐8565Japan
| | - Hiromi Yanagisawa
- Life Science Center for Survival Dynamics, Tsukuba Advanced Research AllianceUniversity of TsukubaIbarakiJapan,Faculty of MedicineUniversity of TsukubaIbarakiJapan
| |
Collapse
|
13
|
Rastogi V, Stefens SJM, Houwaart J, Verhagen HJM, de Bruin JL, van der Pluijm I, Essers J. Molecular Imaging of Aortic Aneurysm and Its Translational Power for Clinical Risk Assessment. Front Med (Lausanne) 2022; 9:814123. [PMID: 35492343 PMCID: PMC9051391 DOI: 10.3389/fmed.2022.814123] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Accepted: 03/21/2022] [Indexed: 01/03/2023] Open
Abstract
Aortic aneurysms (AAs) are dilations of the aorta, that are often fatal upon rupture. Diagnostic radiological techniques such as ultrasound (US), magnetic resonance imaging (MRI), and computed tomography (CT) are currently used in clinical practice for early diagnosis as well as clinical follow-up for preemptive surgery of AA and prevention of rupture. However, the contemporary imaging-based risk prediction of aneurysm enlargement or life-threatening aneurysm-rupture remains limited as these are restricted to visual parameters which fail to provide a personalized risk assessment. Therefore, new insights into early diagnostic approaches to detect AA and therefore to prevent aneurysm-rupture are crucial. Multiple new techniques are developed to obtain a more accurate understanding of the biological processes and pathological alterations at a (micro)structural and molecular level of aortic degeneration. Advanced anatomical imaging combined with molecular imaging, such as molecular MRI, or positron emission tomography (PET)/CT provides novel diagnostic approaches for in vivo visualization of targeted biomarkers. This will aid in the understanding of aortic aneurysm disease pathogenesis and insight into the pathways involved, and will thus facilitate early diagnostic analysis of aneurysmal disease. In this study, we reviewed these molecular imaging modalities and their association with aneurysm growth and/or rupture risk and their limitations. Furthermore, we outline recent pre-clinical and clinical developments in molecular imaging of AA and provide future perspectives based on the advancements made within the field. Within the vastness of pre-clinical markers that have been studied in mice, molecular imaging targets such as elastin/collagen, albumin, matrix metalloproteinases and immune cells demonstrate promising results regarding rupture risk assessment within the pre-clinical setting. Subsequently, these markers hold potential as a future diagnosticum of clinical AA assessment. However currently, clinical translation of molecular imaging is still at the onset. Future human trials are required to assess the effectivity of potentially viable molecular markers with various imaging modalities for clinical rupture risk assessment.
Collapse
Affiliation(s)
- Vinamr Rastogi
- Department of Vascular Surgery, Erasmus University Medical Center, Rotterdam, Netherlands
| | - Sanne J. M. Stefens
- Department of Molecular Genetics, Erasmus University Medical Center, Rotterdam, Netherlands
| | - Judith Houwaart
- Department of Molecular Genetics, Erasmus University Medical Center, Rotterdam, Netherlands
| | - Hence J. M. Verhagen
- Department of Vascular Surgery, Erasmus University Medical Center, Rotterdam, Netherlands
| | - Jorg L. de Bruin
- Department of Vascular Surgery, Erasmus University Medical Center, Rotterdam, Netherlands
| | - Ingrid van der Pluijm
- Department of Vascular Surgery, Erasmus University Medical Center, Rotterdam, Netherlands
- Department of Molecular Genetics, Erasmus University Medical Center, Rotterdam, Netherlands
| | - Jeroen Essers
- Department of Vascular Surgery, Erasmus University Medical Center, Rotterdam, Netherlands
- Department of Molecular Genetics, Erasmus University Medical Center, Rotterdam, Netherlands
- Department of Radiation Oncology, Erasmus University Medical Center, Rotterdam, Netherlands
- *Correspondence: Jeroen Essers
| |
Collapse
|
14
|
Siddiqui HB, Dogru S, Lashkarinia SS, Pekkan K. Soft-Tissue Material Properties and Mechanogenetics during Cardiovascular Development. J Cardiovasc Dev Dis 2022; 9:jcdd9020064. [PMID: 35200717 PMCID: PMC8876703 DOI: 10.3390/jcdd9020064] [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: 11/03/2021] [Revised: 01/22/2022] [Accepted: 01/28/2022] [Indexed: 12/17/2022] Open
Abstract
During embryonic development, changes in the cardiovascular microstructure and material properties are essential for an integrated biomechanical understanding. This knowledge also enables realistic predictive computational tools, specifically targeting the formation of congenital heart defects. Material characterization of cardiovascular embryonic tissue at consequent embryonic stages is critical to understand growth, remodeling, and hemodynamic functions. Two biomechanical loading modes, which are wall shear stress and blood pressure, are associated with distinct molecular pathways and govern vascular morphology through microstructural remodeling. Dynamic embryonic tissues have complex signaling networks integrated with mechanical factors such as stress, strain, and stiffness. While the multiscale interplay between the mechanical loading modes and microstructural changes has been studied in animal models, mechanical characterization of early embryonic cardiovascular tissue is challenging due to the miniature sample sizes and active/passive vascular components. Accordingly, this comparative review focuses on the embryonic material characterization of developing cardiovascular systems and attempts to classify it for different species and embryonic timepoints. Key cardiovascular components including the great vessels, ventricles, heart valves, and the umbilical cord arteries are covered. A state-of-the-art review of experimental techniques for embryonic material characterization is provided along with the two novel methods developed to measure the residual and von Mises stress distributions in avian embryonic vessels noninvasively, for the first time in the literature. As attempted in this review, the compilation of embryonic mechanical properties will also contribute to our understanding of the mature cardiovascular system and possibly lead to new microstructural and genetic interventions to correct abnormal development.
Collapse
Affiliation(s)
- Hummaira Banu Siddiqui
- Department of Mechanical Engineering, Koc University, Istanbul 34450, Turkey; (H.B.S.); (S.D.); (S.S.L.)
| | - Sedat Dogru
- Department of Mechanical Engineering, Koc University, Istanbul 34450, Turkey; (H.B.S.); (S.D.); (S.S.L.)
- Department of Biomedical Engineering, Boston University, Boston, MA 02215, USA
| | - Seyedeh Samaneh Lashkarinia
- Department of Mechanical Engineering, Koc University, Istanbul 34450, Turkey; (H.B.S.); (S.D.); (S.S.L.)
- Department of Bioengineering, Imperial College London, London SW7 2BX, UK
| | - Kerem Pekkan
- Department of Mechanical Engineering, Koc University, Istanbul 34450, Turkey; (H.B.S.); (S.D.); (S.S.L.)
- Correspondence: ; Tel.: +90-(533)-356-3595
| |
Collapse
|
15
|
Rabaglino MB, Wakabayashi M, Pearson JT, Jensen LJ. Effect of age on the vascular proteome in middle cerebral arteries and mesenteric resistance arteries in mice. Mech Ageing Dev 2021; 200:111594. [PMID: 34756926 DOI: 10.1016/j.mad.2021.111594] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Revised: 10/11/2021] [Accepted: 10/26/2021] [Indexed: 10/20/2022]
Abstract
Aging is associated with hypertension and brain blood flow dysregulation, which are major risk factors for cardiovascular and neurodegenerative diseases. Structural remodeling, endothelial dysfunction, or hypercontractility of resistance vessels may cause increased total peripheral resistance and hypertension. Recent studies showed that G protein- and RhoA/Rho-kinase pathways are involved in increased mean arterial pressure (MAP) and arterial tone in middle-aged mice. We aimed to characterize the age-dependent changes in the vascular proteome in normal laboratory mice using mass spectrometry and bioinformatics analyses on middle cerebral arteries and mesenteric resistance arteries from young (3 months) vs. middle-aged (14 months) mice. In total, 31 proteins were significantly affected by age whereas 172 proteins were differentially expressed by vessel type. Hierarchical clustering revealed that 207 proteins were significantly changed or clustered by age. Vitamin B6 pathway, Biosynthesis of antibiotics, Regulation of actin cytoskeleton and Endocytosis were the top enriched KEGG pathways by age. Several proteins in the RhoA/Rho-kinase pathway changed in a manner consistent with hypertension and dysregulation of cerebral perfusion. Although aging had a less profound effect than vessel type on the resistance artery proteome, regulation of actin cytoskeleton, including the RhoA/Rho-kinase pathway, is an important target for age-dependent hypertension.
Collapse
Affiliation(s)
- Maria Belen Rabaglino
- Dept. of Applied Mathematics and Computer Science, Danish Technical University, Denmark
| | - Masaki Wakabayashi
- Omics Research Center, National Cerebral and Cardiovascular Center, Osaka, Japan
| | - James Todd Pearson
- Dept. of Cardiac Physiology, National Cerebral and Cardiovascular Center Research Institute, Osaka, Japan; Monash Biomedicine Discovery Institute and Department of Physiology, Monash University, Clayton, Australia
| | - Lars Jørn Jensen
- Dept. of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Denmark.
| |
Collapse
|
16
|
Kabir AU, Subramanian M, Lee DH, Wang X, Krchma K, Wu J, Naismith T, Halabi CM, Kim JY, Pulous FE, Petrich BG, Kim S, Park HC, Hanson PI, Pan H, Wickline SA, Fremont DH, Park C, Choi K. Dual role of endothelial Myct1 in tumor angiogenesis and tumor immunity. Sci Transl Med 2021; 13:13/583/eabb6731. [PMID: 33658356 DOI: 10.1126/scitranslmed.abb6731] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Revised: 10/01/2020] [Accepted: 02/11/2021] [Indexed: 12/11/2022]
Abstract
The cross-talk between angiogenesis and immunity within the tumor microenvironment (TME) is critical for tumor prognosis. While pro-angiogenic and immunosuppressive TME promote tumor growth, anti-angiogenic and immune stimulatory TME inhibit tumor progression. Therefore, there is a great interest in achieving vascular normalization to improve drug delivery and enhance antitumor immunity. However, anti-vascular endothelial growth factor (VEGF) mechanisms to normalize tumor vessels have offered limited therapeutic efficacies for patients with cancer. Here, we report that Myct1, a direct target of ETV2, was nearly exclusively expressed in endothelial cells. In preclinical mouse tumor models, Myct1 deficiency reduced angiogenesis, enhanced high endothelial venule formation, and promoted antitumor immunity, leading to restricted tumor progression. Analysis of The Cancer Genome Atlas (TCGA) datasets revealed a significant (P < 0.05) correlation between MYCT1 expression, angiogenesis, and antitumor immunity in human cancers, as suggested by decreased FOXP3 expression and increased antitumor macrophages in patients with low MYCT1 expression. Mechanistically, MYCT1 interacted with tight junction protein Zona Occludens 1 and regulated Rho GTPase-mediated actin cytoskeleton dynamics, thereby promoting endothelial motility in the angiogenic environment. Myct1-deficient endothelial cells facilitated trans-endothelial migration of cytotoxic T lymphocytes and polarization of M1 macrophages. Myct1 targeting combined with anti-PD1 treatment significantly (P < 0.05) increased complete tumor regression and long-term survival in anti-PD1-responsive and -refractory tumor models in mice. Our data collectively support a critical role for Myct1 in controlling tumor angiogenesis and reprogramming tumor immunity. Myct1-targeted vascular control, in combination with immunotherapy, may become an exciting therapeutic strategy.
Collapse
Affiliation(s)
- Ashraf Ul Kabir
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110-1093, USA.,Molecular and Cell Biology Program, Washington University School of Medicine, St. Louis, MO 63110-1093, USA
| | - Madhav Subramanian
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110-1093, USA
| | - Dong Hun Lee
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Xiaoli Wang
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110-1093, USA
| | - Karen Krchma
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110-1093, USA
| | - Jun Wu
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110-1093, USA
| | - Teri Naismith
- Department of Cell Biology and Physiology, Washington University School of Medicine, St. Louis, MO 63110-1093, USA
| | - Carmen M Halabi
- Department of Pediatrics, Washington University School of Medicine, St. Louis, MO 63110-1093, USA
| | - Ju Young Kim
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Fadi E Pulous
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Brian G Petrich
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Suhyun Kim
- Department of Biomedical Sciences, College of Medicine, Korea University, Ansan 15335, Republic of Korea
| | - Hae-Chul Park
- Department of Biomedical Sciences, College of Medicine, Korea University, Ansan 15335, Republic of Korea
| | - Phyllis I Hanson
- Department of Biological Chemistry, University of Michigan Medical School, Ann Arbor, MI 48109-5624, USA
| | - Hua Pan
- Health Heart Institute, Morsani College of Medicine, University of South Florida, Tampa, FL 33612, USA
| | - Samuel A Wickline
- Health Heart Institute, Morsani College of Medicine, University of South Florida, Tampa, FL 33612, USA
| | - Daved H Fremont
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110-1093, USA
| | - Changwon Park
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA 30322, USA. .,Department of Molecular and Cellular Physiology, Louisiana State University Health Science Center, Shreveport, LA 71103, USA
| | - Kyunghee Choi
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110-1093, USA. .,Molecular and Cell Biology Program, Washington University School of Medicine, St. Louis, MO 63110-1093, USA.,Graduate School of Biotechnology, Kyung Hee University, Yong In 17104, Republic of Korea
| |
Collapse
|
17
|
Role of Fibulins in Embryonic Stage Development and Their Involvement in Various Diseases. Biomolecules 2021; 11:biom11050685. [PMID: 34063320 PMCID: PMC8147605 DOI: 10.3390/biom11050685] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Revised: 04/29/2021] [Accepted: 04/30/2021] [Indexed: 12/24/2022] Open
Abstract
The extracellular matrix (ECM) plays an important role in the evolution of early metazoans, as it provides structural and biochemical support to the surrounding cells through the cell–cell and cell–matrix interactions. In multi-cellular organisms, ECM plays a pivotal role in the differentiation of tissues and in the development of organs. Fibulins are ECM glycoproteins, found in a variety of tissues associated with basement membranes, elastic fibers, proteoglycan aggregates, and fibronectin microfibrils. The expression profile of fibulins reveals their role in various developmental processes such as elastogenesis, development of organs during the embryonic stage, tissue remodeling, maintenance of the structural integrity of basement membrane, and elastic fibers, as well as other cellular processes. Apart from this, fibulins are also involved in the progression of human diseases such as cancer, cardiac diseases, congenital disorders, and chronic fibrotic disorders. Different isoforms of fibulins show a dual role of tumor-suppressive and tumor-promoting activities, depending on the cell type and cellular microenvironment in the body. Knockout animal models have provided deep insight into their role in development and diseases. The present review covers details of the structural and expression patterns, along with the role of fibulins in embryonic development and disease progression, with more emphasis on their involvement in the modulation of cancer diseases.
Collapse
|
18
|
Abstract
Significance: The vascular extracellular matrix (ECM) not only provides mechanical stability but also manipulates vascular cell behaviors, which are crucial for vascular function and homeostasis. ECM remodeling, which alters vascular wall mechanical properties and exposes vascular cells to bioactive molecules, is involved in the development and progression of hypertension. Recent Advances: This brief review summarized the dynamic changes in ECM components and their modification and degradation during hypertension and after antihypertensive treatment. We also discussed how alterations in the ECM amount, assembly, mechanical properties, and degradation fragment generation provide input into the pathological process of hypertension. Critical Issues: Although the relevance between ECM remodeling and hypertension has been recognized, the underlying mechanism by which ECM remodeling initiates the development of hypertension remains unclear. Therefore, the modulation of ECM remodeling on arterial stiffness and hypertension in genetically modified rodent models is summarized in this review. The circulating biomarkers based on ECM metabolism and therapeutic strategies targeting ECM disorders in hypertension are also introduced. Future Directions: Further research will provide more comprehensive understanding of ECM remodeling in hypertension by the application of matridomic and degradomic approaches. The better understanding of mechanisms underlying vascular ECM remodeling may provide novel potential therapeutic strategies for preventing and treating hypertension. Antioxid. Redox Signal. 34, 765-783.
Collapse
Affiliation(s)
- Zeyu Cai
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, Beijing, China.,Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing, China
| | - Ze Gong
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, Beijing, China.,Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing, China
| | - Zhiqing Li
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, Beijing, China.,Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing, China
| | - Li Li
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, Beijing, China.,Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing, China
| | - Wei Kong
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, Beijing, China.,Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing, China
| |
Collapse
|
19
|
Creamer TJ, Bramel EE, MacFarlane EG. Insights on the Pathogenesis of Aneurysm through the Study of Hereditary Aortopathies. Genes (Basel) 2021; 12:183. [PMID: 33514025 PMCID: PMC7912671 DOI: 10.3390/genes12020183] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Revised: 01/20/2021] [Accepted: 01/22/2021] [Indexed: 12/15/2022] Open
Abstract
Thoracic aortic aneurysms (TAA) are permanent and localized dilations of the aorta that predispose patients to a life-threatening risk of aortic dissection or rupture. The identification of pathogenic variants that cause hereditary forms of TAA has delineated fundamental molecular processes required to maintain aortic homeostasis. Vascular smooth muscle cells (VSMCs) elaborate and remodel the extracellular matrix (ECM) in response to mechanical and biochemical cues from their environment. Causal variants for hereditary forms of aneurysm compromise the function of gene products involved in the transmission or interpretation of these signals, initiating processes that eventually lead to degeneration and mechanical failure of the vessel. These include mutations that interfere with transduction of stimuli from the matrix to the actin-myosin cytoskeleton through integrins, and those that impair signaling pathways activated by transforming growth factor-β (TGF-β). In this review, we summarize the features of the healthy aortic wall, the major pathways involved in the modulation of VSMC phenotypes, and the basic molecular functions impaired by TAA-associated mutations. We also discuss how the heterogeneity and balance of adaptive and maladaptive responses to the initial genetic insult might contribute to disease.
Collapse
Affiliation(s)
- Tyler J. Creamer
- McKusick-Nathans Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; (T.J.C.); (E.E.B.)
- Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Emily E. Bramel
- McKusick-Nathans Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; (T.J.C.); (E.E.B.)
- Predoctoral Training in Human Genetics and Molecular Biology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Elena Gallo MacFarlane
- McKusick-Nathans Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; (T.J.C.); (E.E.B.)
- Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| |
Collapse
|
20
|
Abstract
PURPOSE OF REVIEW Elastin has historically been described as an amorphous protein that functions to provide recoil to tissues that stretch. However, evidence is growing that elastin's role may not be limited to biomechanics. In this minireview, we will summarize current knowledge regarding vascular elastic fibers, focusing on structural differences along the arterial tree and how those differences may influence the behavior of affiliated cells. RECENT FINDINGS Regional heterogeneity, including differences in elastic lamellar number, density and cell developmental origin, plays an important role in vessel health and function. These differences impact cell-cell communication, proliferation and movement. Perturbations of normal cell-matrix interactions are correlated with human diseases including aneurysm, atherosclerosis and hypertension. SUMMARY Although classically described as a structural protein, recent data suggest that differences in elastin deposition along the arterial tree have important effects on heterotypic cell interactions and human disease.
Collapse
|
21
|
Song R, Zhang L. Cardiac ECM: Its Epigenetic Regulation and Role in Heart Development and Repair. Int J Mol Sci 2020; 21:ijms21228610. [PMID: 33203135 PMCID: PMC7698074 DOI: 10.3390/ijms21228610] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2020] [Revised: 11/07/2020] [Accepted: 11/13/2020] [Indexed: 12/14/2022] Open
Abstract
The extracellular matrix (ECM) is the non-cellular component in the cardiac microenvironment, and serves essential structural and regulatory roles in establishing and maintaining tissue architecture and cellular function. The patterns of molecular and biochemical ECM alterations in developing and adult hearts depend on the underlying injury type. In addition to exploring how the ECM regulates heart structure and function in heart development and repair, this review conducts an inclusive discussion of recent developments in the role, function, and epigenetic guidelines of the ECM. Moreover, it contributes to the development of new therapeutics for cardiovascular disease.
Collapse
Affiliation(s)
- Rui Song
- Correspondence: (R.S.); (L.Z.); Tel.: +1-909-558-4325 (R.S. & L.Z.)
| | - Lubo Zhang
- Correspondence: (R.S.); (L.Z.); Tel.: +1-909-558-4325 (R.S. & L.Z.)
| |
Collapse
|
22
|
McClenaghan C, Huang Y, Matkovich SJ, Kovacs A, Weinheimer CJ, Perez R, Broekelmann TJ, Harter TM, Lee JM, Remedi MS, Nichols CG. The Mechanism of High-Output Cardiac Hypertrophy Arising From Potassium Channel Gain-of-Function in Cantú Syndrome. FUNCTION 2020; 1:zqaa004. [PMID: 32865539 PMCID: PMC7446247 DOI: 10.1093/function/zqaa004] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2020] [Revised: 06/08/2020] [Accepted: 06/10/2020] [Indexed: 01/06/2023] Open
Abstract
Dramatic cardiomegaly arising from gain-of-function (GoF) mutations in the ATP-sensitive potassium (KATP) channels genes, ABCC9 and KCNJ8, is a characteristic feature of Cantú syndrome (CS). How potassium channel over-activity results in cardiac hypertrophy, as well as the long-term consequences of cardiovascular remodeling in CS, is unknown. Using genome-edited mouse models of CS, we therefore sought to dissect the pathophysiological mechanisms linking KATP channel GoF to cardiac remodeling. We demonstrate that chronic reduction of systemic vascular resistance in CS is accompanied by elevated renin-angiotensin signaling, which drives cardiac enlargement and blood volume expansion. Cardiac enlargement in CS results in elevation of basal cardiac output, which is preserved in aging. However, the cardiac remodeling includes altered gene expression patterns that are associated with pathological hypertrophy and are accompanied by decreased exercise tolerance, suggestive of reduced cardiac reserve. Our results identify a high-output cardiac hypertrophy phenotype in CS which is etiologically and mechanistically distinct from other myocardial hypertrophies, and which exhibits key features of high-output heart failure (HOHF). We propose that CS is a genetically-defined HOHF disorder and that decreased vascular smooth muscle excitability is a novel mechanism for HOHF pathogenesis.
Collapse
Affiliation(s)
- Conor McClenaghan
- Center for the Investigation of Membrane Excitability Diseases, Washington University School of Medicine, St. Louis, MO 63110, USA
- Departments of Cell Biology and Physiology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Yan Huang
- Center for the Investigation of Membrane Excitability Diseases, Washington University School of Medicine, St. Louis, MO 63110, USA
- Departments of Cell Biology and Physiology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Scot J Matkovich
- Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Attila Kovacs
- Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Carla J Weinheimer
- Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Ron Perez
- Neurology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Thomas J Broekelmann
- Departments of Cell Biology and Physiology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Theresa M Harter
- Center for the Investigation of Membrane Excitability Diseases, Washington University School of Medicine, St. Louis, MO 63110, USA
- Departments of Cell Biology and Physiology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Jin-Moo Lee
- Neurology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Maria S Remedi
- Center for the Investigation of Membrane Excitability Diseases, Washington University School of Medicine, St. Louis, MO 63110, USA
- Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Colin G Nichols
- Center for the Investigation of Membrane Excitability Diseases, Washington University School of Medicine, St. Louis, MO 63110, USA
- Departments of Cell Biology and Physiology, Washington University School of Medicine, St. Louis, MO 63110, USA
| |
Collapse
|
23
|
Gabriela Espinosa M, Catalin Staiculescu M, Kim J, Marin E, Wagenseil JE. Elastic Fibers and Large Artery Mechanics in Animal Models of Development and Disease. J Biomech Eng 2019; 140:2666245. [PMID: 29222533 DOI: 10.1115/1.4038704] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2017] [Indexed: 12/21/2022]
Abstract
Development of a closed circulatory system requires that large arteries adapt to the mechanical demands of high, pulsatile pressure. Elastin and collagen uniquely address these design criteria in the low and high stress regimes, resulting in a nonlinear mechanical response. Elastin is the core component of elastic fibers, which provide the artery wall with energy storage and recoil. The integrity of the elastic fiber network is affected by component insufficiency or disorganization, leading to an array of vascular pathologies and compromised mechanical behavior. In this review, we discuss how elastic fibers are formed and how they adapt in development and disease. We discuss elastic fiber contributions to arterial mechanical behavior and remodeling. We primarily present data from mouse models with elastic fiber deficiencies, but suggest that alternate small animal models may have unique experimental advantages and the potential to provide new insights. Advanced ultrastructural and biomechanical data are constantly being used to update computational models of arterial mechanics. We discuss the progression from early phenomenological models to microstructurally motivated strain energy functions for both collagen and elastic fiber networks. Although many current models individually account for arterial adaptation, complex geometries, and fluid-solid interactions (FSIs), future models will need to include an even greater number of factors and interactions in the complex system. Among these factors, we identify the need to revisit the role of time dependence and axial growth and remodeling in large artery mechanics, especially in cardiovascular diseases that affect the mechanical integrity of the elastic fibers.
Collapse
Affiliation(s)
| | | | - Jungsil Kim
- Department of Mechanical Engineering and Materials Science, Washington University, St. Louis, MO 63130
| | - Eric Marin
- Department of Biomedical Engineering, Saint Louis University, St. Louis, MO 63103
| | - Jessica E Wagenseil
- Department of Mechanical Engineering and Materials Science, Washington University, , St. Louis, MO 63130 e-mail:
| |
Collapse
|
24
|
Elastic fibers and biomechanics of the aorta: Insights from mouse studies. Matrix Biol 2019; 85-86:160-172. [PMID: 30880160 DOI: 10.1016/j.matbio.2019.03.001] [Citation(s) in RCA: 58] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2019] [Revised: 02/28/2019] [Accepted: 03/04/2019] [Indexed: 12/14/2022]
Abstract
Elastic fibers are major components of the extracellular matrix (ECM) in the aorta and support a life-long cycling of stretch and recoil. Elastic fibers are formed from mid-gestation throughout early postnatal development and the synthesis is regulated at multiple steps, including coacervation, deposition, cross-linking, and assembly of insoluble elastin onto microfibril scaffolds. To date, more than 30 molecules have been shown to associate with elastic fibers and some of them play a critical role in the formation and maintenance of elastic fibers in vivo. Because the aorta is subjected to high pressure from the left ventricle, elasticity of the aorta provides the Windkessel effect and maintains stable blood flow to distal organs throughout the cardiac cycle. Disruption of elastic fibers due to congenital defects, inflammation, or aging dramatically reduces aortic elasticity and affects overall vessel mechanics. Another important component in the aorta is the vascular smooth muscle cells (SMCs). Elastic fibers and SMCs alternate to create a highly organized medial layer within the aortic wall. The physical connections between elastic fibers and SMCs form the elastin-contractile units and maintain cytoskeletal organization and proper responses of SMCs to mechanical strain. In this review, we revisit the components of elastic fibers and their roles in elastogenesis and how a loss of each component affects biomechanics of the aorta. Finally, we discuss the significance of elastin-contractile units in the maintenance of SMC function based on knowledge obtained from mouse models of human disease.
Collapse
|
25
|
Humphrey JD, Tellides G. Central artery stiffness and thoracic aortopathy. Am J Physiol Heart Circ Physiol 2019; 316:H169-H182. [PMID: 30412443 PMCID: PMC6880196 DOI: 10.1152/ajpheart.00205.2018] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/26/2018] [Revised: 10/22/2018] [Accepted: 10/31/2018] [Indexed: 12/20/2022]
Abstract
Thoracic aortopathy, especially aneurysm, dissection, and rupture, is responsible for significant morbidity and mortality. Uncontrolled hypertension and aging are primary risk factors for such conditions, and they contribute to an increase in the mechanical stress on the wall and an increase in its structural vulnerability, respectively. Select genetic mutations also predispose to these lethal conditions, and the collection of known mutations suggests that dysfunctional mechanosensing and mechanoregulation of the extracellular matrix may contribute to pathogenesis and disease progression. In the absence of a well-accepted pharmacotherapy, nonsurgical treatments tend to focus on reducing the mechanical loading on the aorta, particularly via the use of antihypertensive medications and recommendations to avoid strenuous exercises such as weight lifting. In this brief review, we discuss the important effects of central artery stiffening on global hemodynamics and, in particular, on the increase in pulse pressure that acts on the proximal thoracic aorta. We consider Marfan syndrome as an illustrative aortopathy but discuss other conditions leading to thoracic aortic aneurysm and dissection. We highlight the importance of phenotyping the aorta biomechanically, not just clinically, and emphasize the utility of mouse models in elucidating molecular and mechanical mechanisms of disease. Notwithstanding the widely recognized role of central artery stiffening in driving end-organ disease, we suggest that there is similarly a need to consider its key role in thoracic aortopathy.
Collapse
Affiliation(s)
- J. D. Humphrey
- Department of Biomedical Engineering, Yale University, New Haven, Connecticut
- Vascular Biology and Therapeutics Program, Yale University, New Haven, Connecticut
| | - G. Tellides
- Department of Surgery, Yale University, New Haven, Connecticut
- Vascular Biology and Therapeutics Program, Yale University, New Haven, Connecticut
| |
Collapse
|
26
|
Roles of short fibulins, a family of matricellular proteins, in lung matrix assembly and disease. Matrix Biol 2018; 73:21-33. [DOI: 10.1016/j.matbio.2018.02.003] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2017] [Revised: 12/26/2017] [Accepted: 02/01/2018] [Indexed: 12/19/2022]
|
27
|
Extracellular Interactions between Fibulins and Transforming Growth Factor (TGF)-β in Physiological and Pathological Conditions. Int J Mol Sci 2018; 19:ijms19092787. [PMID: 30227601 PMCID: PMC6163299 DOI: 10.3390/ijms19092787] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2018] [Revised: 09/11/2018] [Accepted: 09/12/2018] [Indexed: 12/25/2022] Open
Abstract
Transforming growth factor (TGF)-β is a multifunctional peptide growth factor that has a vital role in the regulation of cell growth, differentiation, inflammation, and repair in a variety of tissues, and its dysregulation mediates a number of pathological conditions including fibrotic disorders, chronic inflammation, cardiovascular diseases, and cancer progression. Regulation of TGF-β signaling is multifold, but one critical site of regulation is via interaction with certain extracellular matrix (ECM) microenvironments, as TGF-β is primarily secreted as a biologically inactive form sequestrated into ECM. Several ECM proteins are known to modulate TGF-β signaling via cell–matrix interactions, including thrombospondins, SPARC (Secreted Protein Acidic and Rich in Cystein), tenascins, osteopontin, periostin, and fibulins. Fibulin family members consist of eight ECM glycoproteins characterized by a tandem array of calcium-binding epidermal growth factor-like modules and a common C-terminal domain. Fibulins not only participate in structural integrity of basement membrane and elastic fibers, but also serve as mediators for cellular processes and tissue remodeling as they are highly upregulated during embryonic development and certain disease processes, especially at the sites of epithelial–mesenchymal transition (EMT). Emerging studies have indicated a close relationship between fibulins and TGF-β signaling, but each fibulin plays a different role in a context-dependent manner. In this review, regulatory interactions between fibulins and TGF-β signaling are discussed. Understanding biological roles of fibulins in TGF-β regulation may introduce new insights into the pathogenesis of some human diseases.
Collapse
|
28
|
Fibrillin microfibrils and elastic fibre proteins: Functional interactions and extracellular regulation of growth factors. Semin Cell Dev Biol 2018; 89:109-117. [PMID: 30016650 PMCID: PMC6461133 DOI: 10.1016/j.semcdb.2018.07.016] [Citation(s) in RCA: 102] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2018] [Revised: 07/04/2018] [Accepted: 07/13/2018] [Indexed: 02/02/2023]
Abstract
Fibrillin microfibrils are extensible polymers that endow connective tissues with long-range elasticity and have widespread distributions in both elastic and non-elastic tissues. They act as a template for elastin deposition during elastic fibre formation and are essential for maintaining the integrity of tissues such as blood vessels, lung, skin and ocular ligaments. A reduction in fibrillin is seen in tissues in vascular ageing, chronic obstructive pulmonary disease, skin ageing and UV induced skin damage, and age-related vision deterioration. Most mutations in fibrillin cause Marfan syndrome, a genetic disease characterised by overgrowth of the long bones and other skeletal abnormalities with cardiovascular and eye defects. However, mutations in fibrillin and fibrillin-binding proteins can also cause short-stature pathologies. All of these diseases have been linked to dysregulated growth factor signalling which forms a major functional role for fibrillin.
Collapse
|
29
|
Kabir AU, Lee TJ, Pan H, Berry JC, Krchma K, Wu J, Liu F, Kang HK, Hinman K, Yang L, Hamilton S, Zhou Q, Veis DJ, Mecham RP, Wickline SA, Miller MJ, Choi K. Requisite endothelial reactivation and effective siRNA nanoparticle targeting of Etv2/Er71 in tumor angiogenesis. JCI Insight 2018; 3:97349. [PMID: 29669933 DOI: 10.1172/jci.insight.97349] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2017] [Accepted: 03/20/2018] [Indexed: 01/24/2023] Open
Abstract
Angiogenesis, new blood vessel formation from preexisting vessels, is critical for solid tumor growth. As such, there have been efforts to inhibit angiogenesis as a means to obstruct tumor growth. However, antiangiogenic therapy faces major challenges to the selective targeting of tumor-associated-vessels, as current antiangiogenic targets also disrupt steady-state vessels. Here, we demonstrate that the developmentally critical transcription factor Etv2 is selectively upregulated in both human and mouse tumor-associated endothelial cells (TAECs) and is required for tumor angiogenesis. Two-photon imaging revealed that Etv2-deficient tumor-associated vasculature remained similar to that of steady-state vessels. Etv2-deficient TAECs displayed decreased Flk1 (also known as Vegfr2) expression, FLK1 activation, and proliferation. Endothelial tube formation, proliferation, and sprouting response to VEGF, but not to FGF2, was reduced in Etv2-deficient ECs. ROS activated Etv2 expression in ECs, and ROS blockade inhibited Etv2 expression in TAECs in vivo. Systemic administration of Etv2 siRNA nanoparticles potently inhibited tumor growth and angiogenesis without cardiovascular side effects. These studies highlight a link among vascular oxidative stress, Etv2 expression, and VEGF response that is critical for tumor angiogenesis. Targeting the ETV2 pathway might offer a unique opportunity for more selective antiangiogenic therapies.
Collapse
Affiliation(s)
- Ashraf Ul Kabir
- Department of Pathology and Immunology and.,Molecular and Cell Biology Program, Washington University School of Medicine, St. Louis, Missouri, USA
| | | | - Hua Pan
- Health Heart Institute, Morsani College of Medicine, University of South Florida, Tampa, Florida, USA
| | - Jeffrey C Berry
- Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, USA
| | | | - Jun Wu
- Department of Pathology and Immunology and
| | - Fang Liu
- Department of Pathology and Immunology and
| | - Hee-Kyoung Kang
- Department of Pharmacology, School of Medicine, Jeju National University, Jeju, South Korea
| | - Kristina Hinman
- Department of Cell Biology and Physiology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Lihua Yang
- Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Samantha Hamilton
- Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Qingyu Zhou
- Department of Pharmaceutical Science, College of Pharmacy, University of South Florida, Tampa, Florida, USA
| | - Deborah J Veis
- Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Robert P Mecham
- Department of Cell Biology and Physiology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Samuel A Wickline
- Health Heart Institute, Morsani College of Medicine, University of South Florida, Tampa, Florida, USA
| | - Mark J Miller
- Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Kyunghee Choi
- Department of Pathology and Immunology and.,Molecular and Cell Biology Program, Washington University School of Medicine, St. Louis, Missouri, USA.,Graduate School of Biotechnology, Kyung Hee University, Yongin, South Korea
| |
Collapse
|