1
|
Dai Y, Zhu W, Flores Banuelos AG, Li J, Mukherjee S, Algaze C, Wu JC. Generation of two induced pluripotent stem cell lines from patients with Williams syndrome. Stem Cell Res 2024; 78:103460. [PMID: 38861775 DOI: 10.1016/j.scr.2024.103460] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Revised: 05/17/2024] [Accepted: 06/02/2024] [Indexed: 06/13/2024] Open
Abstract
Williams syndrome (WS) is a relatively rare genetic disorder. It arises from a microdeletion in chromosome 7q11.23, resulting in the loss of one copy of more than 20 genes. Disorders in multiple systems, including cardiovascular and nervous systems, occur in patients with WS. Here, we generated two human induced pluripotent stem cell (iPSC) lines from WS patients. Both lines expressed pluripotency markers at gene and protein levels. They possessed normal karyotypes and the potential to differentiate into three germ layers. They serve as a useful tool to study disease mechanism, test drugs, and identify promising therapeutics for patients with WS.
Collapse
Affiliation(s)
- Yuanyuan Dai
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA 94305, USA; Department of Medicine, Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Wenjuan Zhu
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA 94305, USA; Department of Medicine, Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Amira G Flores Banuelos
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA 94305, USA; Department of Medicine, Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Juana Li
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA 94305, USA; Department of Medicine, Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Souhrid Mukherjee
- Greenstone Biosciences, 3160 Porter Dr, Suite 140, Palo Alto, CA 94304, USA
| | - Claudia Algaze
- Department of Pediatrics, Division of Pediatric Cardiology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Joseph C Wu
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA 94305, USA; Department of Medicine, Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA.
| |
Collapse
|
2
|
Saito J, Dave JM, Lau FD, Greif DM. Presenilin-1 in smooth muscle cells facilitates hypermuscularization in elastin aortopathy. iScience 2024; 27:108636. [PMID: 38226162 PMCID: PMC10788461 DOI: 10.1016/j.isci.2023.108636] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Revised: 11/16/2023] [Accepted: 12/01/2023] [Indexed: 01/17/2024] Open
Abstract
Smooth muscle cell (SMC) accumulation is central to the pathogenesis of elastin-defective arterial diseases, including supravalvular aortic stenosis (SVAS). We previously demonstrated that elastin insufficiency activates Notch signaling in aortic SMCs. Activation of Notch is catalyzed by the enzyme gamma-secretase, but the role of catalytic subunits presenilin (PSEN)-1 or PSEN-2 in elastin aortopathy is not defined. Genetic approaches reveal that endothelial cell-specific Psen1 deletion does not improve elastin aortopathy whereas the deletion of either Psen1 in SMCs or Psen2 globally attenuates Notch pathway and SMC proliferation, mitigating aortic disease. With SMC-specific Psen1 deletion in elastin nulls, these rescue effects are more robust and in fact, survival is increased. SMC deletion of Psen1 also attenuates hypermuscularization in newborns heterozygous for the elastin null gene, which genetically mimics SVAS. Similarly, the pharmacological inhibition of PSEN-1 mitigates SMC accumulation in elastin aortopathy. These findings put forth SMC PSEN-1 as a potential therapeutic target in SVAS.
Collapse
Affiliation(s)
- Junichi Saito
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine, Yale University, New Haven, CT 06511, USA
- Department of Genetics, Yale University, New Haven, CT 06511, USA
- Stem Cell Center, Yale University, New Haven, CT 06511, USA
| | - Jui M. Dave
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine, Yale University, New Haven, CT 06511, USA
- Department of Genetics, Yale University, New Haven, CT 06511, USA
- Stem Cell Center, Yale University, New Haven, CT 06511, USA
| | - Freddy Duarte Lau
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine, Yale University, New Haven, CT 06511, USA
- Department of Genetics, Yale University, New Haven, CT 06511, USA
| | - Daniel M. Greif
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine, Yale University, New Haven, CT 06511, USA
- Department of Genetics, Yale University, New Haven, CT 06511, USA
- Stem Cell Center, Yale University, New Haven, CT 06511, USA
| |
Collapse
|
3
|
Dixon AJ, Osei-Owusu P. Elastin haploinsufficiency accelerates age-related structural and functional changes in the renal microvasculature and impairment of renal hemodynamics in female mice. Front Physiol 2023; 14:1141094. [PMID: 37179824 PMCID: PMC10167050 DOI: 10.3389/fphys.2023.1141094] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Accepted: 04/11/2023] [Indexed: 05/15/2023] Open
Abstract
Age-related decline in functional elastin is associated with increased arterial stiffness, a known risk factor for developing cardiovascular disease. While the contribution of elastin insufficiency to the stiffening of conduit arteries is well described, little is known about the impact on the structure and function of the resistance vasculature, which contributes to total peripheral resistance and the regulation of organ perfusion. In this study, we determined how elastin insufficiency impinges on age-related changes in the structure and biomechanical properties of the renal microvasculature, altering renal hemodynamics and the response of the renal vascular bed to changes in renal perfusion pressure (RPP) in female mice. Using Doppler ultrasonography, we found that resistive index and pulsatility index were elevated in young Eln +/- and aged mice. Histological examination showed thinner internal and external elastic laminae, accompanied by increased elastin fragmentation in the medial layer without any calcium deposits in the small intrarenal arteries of kidneys from young Eln +/- and aged mice. Pressure myography of interlobar arteries showed that vessels from young Eln +/- and aged mice had a slight decrease in distensibility during pressure loading but a substantial decline in vascular recoil efficiency upon pressure unloading. To examine whether structural changes in the renal microvasculature influenced renal hemodynamics, we clamped neurohumoral input and increased renal perfusion pressure by simultaneously occluding the superior mesenteric and celiac arteries. Increased renal perfusion pressure caused robust changes in blood pressure in all groups; however, changes in renal vascular resistance and renal blood flow (RBF) were blunted in young Eln +/- and aged mice, accompanied by decreased autoregulatory index, indicating greater impairment of renal autoregulation. Finally, increased pulse pressure in aged Eln +/- mice positively correlated with high renal blood flow. Together, our data show that the loss of elastin negatively affects the structural and functional integrity of the renal microvasculature, ultimately worsening age-related decline in kidney function.
Collapse
Affiliation(s)
- Alethia J Dixon
- Physiology and Biophysics, School of Medicine, Case Western Reserve University, Cleveland, OH, United States
| | - Patrick Osei-Owusu
- Physiology and Biophysics, School of Medicine, Case Western Reserve University, Cleveland, OH, United States
| |
Collapse
|
4
|
Zhang E, Spronck B, Humphrey JD, Karniadakis GE. G2Φnet: Relating genotype and biomechanical phenotype of tissues with deep learning. PLoS Comput Biol 2022; 18:e1010660. [PMID: 36315608 PMCID: PMC9668200 DOI: 10.1371/journal.pcbi.1010660] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Revised: 11/16/2022] [Accepted: 10/17/2022] [Indexed: 11/05/2022] Open
Abstract
Many genetic mutations adversely affect the structure and function of load-bearing soft tissues, with clinical sequelae often responsible for disability or death. Parallel advances in genetics and histomechanical characterization provide significant insight into these conditions, but there remains a pressing need to integrate such information. We present a novel genotype-to-biomechanical phenotype neural network (G2Φnet) for characterizing and classifying biomechanical properties of soft tissues, which serve as important functional readouts of tissue health or disease. We illustrate the utility of our approach by inferring the nonlinear, genotype-dependent constitutive behavior of the aorta for four mouse models involving defects or deficiencies in extracellular constituents. We show that G2Φnet can infer the biomechanical response while simultaneously ascribing the associated genotype by utilizing limited, noisy, and unstructured experimental data. More broadly, G2Φnet provides a powerful method and a paradigm shift for correlating genotype and biomechanical phenotype quantitatively, promising a better understanding of their interplay in biological tissues.
Collapse
Affiliation(s)
- Enrui Zhang
- Division of Applied Mathematics, Brown University, Providence, Rhode Island, United States of America
| | - Bart Spronck
- Department of Biomedical Engineering, Maastricht University, Maastricht, the Netherlands
| | - Jay D. Humphrey
- Department of Biomedical Engineering, Yale University, New Haven, Connecticut, United States of America
| | - George Em Karniadakis
- Division of Applied Mathematics, Brown University, Providence, Rhode Island, United States of America
- School of Engineering, Brown University, Providence, Rhode Island, United States of America
| |
Collapse
|
5
|
Lin CJ, Cocciolone AJ, Wagenseil JE. Elastin, arterial mechanics, and stenosis. Am J Physiol Cell Physiol 2022; 322:C875-C886. [PMID: 35196168 PMCID: PMC9037699 DOI: 10.1152/ajpcell.00448.2021] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Elastin is a long-lived extracellular matrix protein that is organized into elastic fibers that provide elasticity to the arterial wall, allowing stretch and recoil with each cardiac cycle. By forming lamellar units with smooth muscle cells, elastic fibers transduce tissue-level mechanics to cell-level changes through mechanobiological signaling. Altered amounts or assembly of elastic fibers leads to changes in arterial structure and mechanical behavior that compromise cardiovascular function. In particular, genetic mutations in the elastin gene (ELN) that reduce elastin protein levels are associated with focal arterial stenosis, or narrowing of the arterial lumen, such as that seen in supravalvular aortic stenosis and Williams-Beuren syndrome. Global reduction of Eln levels in mice allows investigation of the tissue- and cell-level arterial mechanical changes and associated alterations in smooth muscle cell phenotype that may contribute to stenosis formation. A loxP-floxed Eln allele in mice highlights cell type- and developmental origin-specific mechanobiological effects of reduced elastin amounts. Eln production is required in distinct cell types for elastic layer formation in different parts of the mouse vasculature. Eln deletion in smooth muscle cells from different developmental origins in the ascending aorta leads to characteristic patterns of vascular stenosis and neointima. Dissecting the mechanobiological signaling associated with local Eln depletion and subsequent smooth muscle cell response may help develop new therapeutic interventions for elastin-related diseases.
Collapse
Affiliation(s)
- Chien-Jung Lin
- 1Department of Cell Biology and Physiology, Washington University, St. Louis, Missouri,2Cardiovascular Division, Department of Medicine, Washington University, St. Louis, Missouri
| | - Austin J. Cocciolone
- 3Department of Biomedical Engineering, Washington University, St. Louis, Missouri
| | - Jessica E. Wagenseil
- 4Department of Mechanical Engineering and Materials Science, Washington University, St. Louis, Missouri
| |
Collapse
|
6
|
Ellis MW, Riaz M, Huang Y, Anderson CW, Luo J, Park J, Lopez CA, Batty LD, Gibson KH, Qyang Y. Epigallocatechin gallate facilitates extracellular elastin fiber formation in induced pluripotent stem cell derived vascular smooth muscle cells for tissue engineering. J Mol Cell Cardiol 2022; 163:167-174. [PMID: 34979103 PMCID: PMC8920537 DOI: 10.1016/j.yjmcc.2021.12.014] [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: 08/16/2021] [Revised: 12/22/2021] [Accepted: 12/28/2021] [Indexed: 02/03/2023]
Abstract
Tissue engineered vascular grafts possess several advantages over synthetic or autologous grafts, including increased availability and reduced rates of infection and thrombosis. Engineered grafts constructed from human induced pluripotent stem cell derivatives further offer enhanced reproducibility in graft production. One notable obstacle to clinical application of these grafts is the lack of elastin in the vessel wall, which would serve to endow compliance in addition to mechanical strength. This study establishes the ability of the polyphenol compound epigallocatechin gallate, a principal component of green tea, to facilitate the extracellular formation of elastin fibers in vascular smooth muscle cells derived from human induced pluripotent stem cells. Further, this study describes the creation of a doxycycline-inducible elastin expression system to uncouple elastin production from vascular smooth muscle cell proliferative capacity to permit fiber formation in conditions conducive to robust tissue engineering.
Collapse
Affiliation(s)
- Matthew W Ellis
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine, Yale School of Medicine, New Haven, CT 06511, USA; Yale Stem Cell Center, New Haven, CT 06520, USA; Department of Cellular and Molecular Physiology, Yale University, New Haven, CT 06519, USA
| | - Muhammad Riaz
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine, Yale School of Medicine, New Haven, CT 06511, USA; Yale Stem Cell Center, New Haven, CT 06520, USA
| | - Yan Huang
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine, Yale School of Medicine, New Haven, CT 06511, USA; Yale Stem Cell Center, New Haven, CT 06520, USA
| | - Christopher W Anderson
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine, Yale School of Medicine, New Haven, CT 06511, USA; Yale Stem Cell Center, New Haven, CT 06520, USA; Department of Pathology, Yale University, New Haven, CT 06520, USA
| | - Jiesi Luo
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine, Yale School of Medicine, New Haven, CT 06511, USA; Yale Stem Cell Center, New Haven, CT 06520, USA
| | - Jinkyu Park
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine, Yale School of Medicine, New Haven, CT 06511, USA; Yale Stem Cell Center, New Haven, CT 06520, USA
| | - Colleen A Lopez
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine, Yale School of Medicine, New Haven, CT 06511, USA; Yale Stem Cell Center, New Haven, CT 06520, USA
| | - Luke D Batty
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine, Yale School of Medicine, New Haven, CT 06511, USA; Yale Stem Cell Center, New Haven, CT 06520, USA; Department of Pathology, Yale University, New Haven, CT 06520, USA
| | - Kimberley H Gibson
- Center for Cellular and Molecular Imaging: Electron Microscopy, Department of Cell Biology, Yale School of Medicine, New Haven, CT 06520, USA
| | - Yibing Qyang
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine, Yale School of Medicine, New Haven, CT 06511, USA; Yale Stem Cell Center, New Haven, CT 06520, USA; Department of Pathology, Yale University, New Haven, CT 06520, USA; Vascular Biology and Therapeutics Program, Yale University School of Medicine, New Haven, CT 06520, USA.
| |
Collapse
|
7
|
Lin M, Roth RA, Kozel BA, Mecham RP, Halabi CM. Loss of Angiotensin II Type 2 Receptor Improves Blood Pressure in Elastin Insufficiency. Front Cardiovasc Med 2021; 8:782138. [PMID: 34790711 PMCID: PMC8591102 DOI: 10.3389/fcvm.2021.782138] [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] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Accepted: 10/11/2021] [Indexed: 11/13/2022] Open
Abstract
There is ample evidence supporting a role for angiotensin II type 2 receptor (AT2R) in counterbalancing the effects of angiotensin II (ang II) through the angiotensin II type 1 receptor by promoting vasodilation and having anti-inflammatory effects. Elastin insufficiency in both humans and mice results in large artery stiffness and systolic hypertension. Unexpectedly, mesenteric arteries from elastin insufficient (Eln+/−) mice were shown to have significant vasoconstriction to AT2R agonism in vitro suggesting that AT2R may have vasoconstrictor effects in elastin insufficiency. Given the potential promise for the use of AT2R agonists clinically, the goal of this study was to determine whether AT2R has vasoconstrictive effects in elastin insufficiency in vivo. To avoid off-target effects of agonists and antagonists, mice lacking AT2R (Agtr2−/Y) were bred to Eln+/− mice and cardiovascular parameters were assessed in wild-type (WT), Agtr2−/Y, Eln+/−, and Agtr2−/Y;Eln+/− littermates. As previously published, Agtr2−/Y mice were normotensive at baseline and had no large artery stiffness, while Eln+/− mice exhibited systolic hypertension and large artery stiffness. Loss of AT2R in Eln+/− mice did not affect large artery stiffness or arterial structure but resulted in significant reduction of both systolic and diastolic blood pressure. These data support a potential vasocontractile role for AT2R in elastin insufficiency. Careful consideration and investigation are necessary to determine the patient population that might benefit from the use of AT2R agonists.
Collapse
Affiliation(s)
- Michelle Lin
- Division of Nephrology, Department of Pediatrics, Washington University School of Medicine, Saint Louis, MO, United States
| | - Robyn A Roth
- Department of Cell Biology and Physiology, Washington University School of Medicine, Saint Louis, MO, United States
| | - Beth A Kozel
- National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, MD, United States
| | - Robert P Mecham
- Department of Cell Biology and Physiology, Washington University School of Medicine, Saint Louis, MO, United States
| | - Carmen M Halabi
- Division of Nephrology, Department of Pediatrics, Washington University School of Medicine, Saint Louis, MO, United States
| |
Collapse
|
8
|
Mohabeer AL, Kroetsch JT, McFadden M, Khosraviani N, Broekelmann TJ, Hou G, Zhang H, Zhou YQ, Wang M, Gramolini AO, Mecham RP, Heximer SP, Bolz SS, Bendeck MP. Deletion of type VIII collagen reduces blood pressure, increases carotid artery functional distensibility and promotes elastin deposition. Matrix Biol Plus 2021; 12:100085. [PMID: 34693248 PMCID: PMC8517381 DOI: 10.1016/j.mbplus.2021.100085] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2021] [Revised: 09/14/2021] [Accepted: 09/21/2021] [Indexed: 11/19/2022] Open
Abstract
Arterial stiffening is a significant predictor of cardiovascular disease development and mortality. In elastic arteries, stiffening refers to the loss and fragmentation of elastic fibers, with a progressive increase in collagen fibers. Type VIII collagen (Col-8) is highly expressed developmentally, and then once again dramatically upregulated in aged and diseased vessels characterized by arterial stiffening. Yet its biophysical impact on the vessel wall remains unknown. The purpose of this study was to test the hypothesis that Col-8 functions as a matrix scaffold to maintain vessel integrity during extracellular matrix (ECM) development. These changes are predicted to persist into the adult vasculature, and we have tested this in our investigation. Through our in vivo and in vitro studies, we have determined a novel interaction between Col-8 and elastin. Mice deficient in Col-8 (Col8-/-) had reduced baseline blood pressure and increased arterial compliance, indicating an enhanced Windkessel effect in conducting arteries. Differences in both the ECM composition and VSMC activity resulted in Col8-/- carotid arteries that displayed increased crosslinked elastin and functional distensibility, but enhanced catecholamine-induced VSMC contractility. In vitro studies revealed that the absence of Col-8 dramatically increased tropoelastin mRNA and elastic fiber deposition in the ECM, which was decreased with exogenous Col-8 treatment. These findings suggest a causative role for Col-8 in reducing mRNA levels of tropoelastin and the presence of elastic fibers in the matrix. Moreover, we also found that Col-8 and elastin have opposing effects on VSMC phenotype, the former promoting a synthetic phenotype, whereas the latter confers quiescence. These studies further our understanding of Col-8 function and open a promising new area of investigation related to elastin biology.
Collapse
Affiliation(s)
- Amanda L. Mohabeer
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
- Translational Biology and Engineering Program, Ted Rogers Centre for Heart Research, University of Toronto, Toronto, Ontario, Canada
| | - Jeffrey T. Kroetsch
- Department of Physiology, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
- Translational Biology and Engineering Program, Ted Rogers Centre for Heart Research, University of Toronto, Toronto, Ontario, Canada
- Toronto Centre for Microvascular Medicine at TBEP, University of Toronto, Toronto, Ontario, Canada
| | - Meghan McFadden
- Institute of Biomedical Engineering, University of Toronto, Toronto, Ontario, Canada
- Translational Biology and Engineering Program, Ted Rogers Centre for Heart Research, University of Toronto, Toronto, Ontario, Canada
| | - Negin Khosraviani
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
- Translational Biology and Engineering Program, Ted Rogers Centre for Heart Research, University of Toronto, Toronto, Ontario, Canada
| | - Thomas J. Broekelmann
- Department of Cell Biology and Physiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Guangpei Hou
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
- Translational Biology and Engineering Program, Ted Rogers Centre for Heart Research, University of Toronto, Toronto, Ontario, Canada
| | - Hangjun Zhang
- Department of Physiology, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
- Translational Biology and Engineering Program, Ted Rogers Centre for Heart Research, University of Toronto, Toronto, Ontario, Canada
| | - Yu-Qing Zhou
- Institute of Biomedical Engineering, University of Toronto, Toronto, Ontario, Canada
- Translational Biology and Engineering Program, Ted Rogers Centre for Heart Research, University of Toronto, Toronto, Ontario, Canada
| | - Minyao Wang
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
- Translational Biology and Engineering Program, Ted Rogers Centre for Heart Research, University of Toronto, Toronto, Ontario, Canada
| | - Anthony O. Gramolini
- Department of Physiology, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
- Translational Biology and Engineering Program, Ted Rogers Centre for Heart Research, University of Toronto, Toronto, Ontario, Canada
| | - Robert P. Mecham
- Department of Cell Biology and Physiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Scott P. Heximer
- Department of Physiology, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
- Translational Biology and Engineering Program, Ted Rogers Centre for Heart Research, University of Toronto, Toronto, Ontario, Canada
| | - Steffen-Sebastian Bolz
- Department of Physiology, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
- Translational Biology and Engineering Program, Ted Rogers Centre for Heart Research, University of Toronto, Toronto, Ontario, Canada
- Toronto Centre for Microvascular Medicine at TBEP, University of Toronto, Toronto, Ontario, Canada
| | - Michelle P. Bendeck
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
- Translational Biology and Engineering Program, Ted Rogers Centre for Heart Research, University of Toronto, Toronto, Ontario, Canada
- Corresponding author at: TBEP, University of Toronto, 661 University Ave, Rm. 1432, Toronto, ON M5G 1M1, Canada.
| |
Collapse
|
9
|
Kozel BA, Barak B, Ae Kim C, Mervis CB, Osborne LR, Porter M, Pober BR. Williams syndrome. Nat Rev Dis Primers 2021; 7:42. [PMID: 34140529 PMCID: PMC9437774 DOI: 10.1038/s41572-021-00276-z] [Citation(s) in RCA: 102] [Impact Index Per Article: 34.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 05/13/2021] [Indexed: 11/09/2022]
Abstract
Williams syndrome (WS) is a relatively rare microdeletion disorder that occurs in as many as 1:7,500 individuals. WS arises due to the mispairing of low-copy DNA repetitive elements at meiosis. The deletion size is similar across most individuals with WS and leads to the loss of one copy of 25-27 genes on chromosome 7q11.23. The resulting unique disorder affects multiple systems, with cardinal features including but not limited to cardiovascular disease (characteristically stenosis of the great arteries and most notably supravalvar aortic stenosis), a distinctive craniofacial appearance, and a specific cognitive and behavioural profile that includes intellectual disability and hypersociability. Genotype-phenotype evidence is strongest for ELN, the gene encoding elastin, which is responsible for the vascular and connective tissue features of WS, and for the transcription factor genes GTF2I and GTF2IRD1, which are known to affect intellectual ability, social functioning and anxiety. Mounting evidence also ascribes phenotypic consequences to the deletion of BAZ1B, LIMK1, STX1A and MLXIPL, but more work is needed to understand the mechanism by which these deletions contribute to clinical outcomes. The age of diagnosis has fallen in regions of the world where technological advances, such as chromosomal microarray, enable clinicians to make the diagnosis of WS without formally suspecting it, allowing earlier intervention by medical and developmental specialists. Phenotypic variability is considerable for all cardinal features of WS but the specific sources of this variability remain unknown. Further investigation to identify the factors responsible for these differences may lead to mechanism-based rather than symptom-based therapies and should therefore be a high research priority.
Collapse
Affiliation(s)
- Beth A. Kozel
- Translational Vascular Medicine Branch, National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, USA
| | - Boaz Barak
- The Sagol School of Neuroscience and The School of Psychological Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Chong Ae Kim
- Department of Pediatrics, Universidade de São Paulo, São Paulo, Brazil
| | - Carolyn B. Mervis
- Department of Psychological and Brain Sciences, University of Louisville, Louisville, USA
| | - Lucy R. Osborne
- Department of Medicine, University of Toronto, Ontario, Canada
| | - Melanie Porter
- Department of Psychology, Macquarie University, Sydney, Australia
| | - Barbara R. Pober
- Department of Pediatrics, Massachusetts General Hospital and Harvard Medical School, Boston, USA
| |
Collapse
|
10
|
Kim SH, Monticone RE, McGraw KR, Wang M. Age-associated proinflammatory elastic fiber remodeling in large arteries. Mech Ageing Dev 2021; 196:111490. [PMID: 33839189 PMCID: PMC8154723 DOI: 10.1016/j.mad.2021.111490] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2021] [Revised: 03/24/2021] [Accepted: 04/05/2021] [Indexed: 12/12/2022]
Abstract
Elastic fibers are the main components of the extracellular matrix of the large arterial wall. Elastic fiber remodeling is an intricate process of synthesis and degradation of the core elastin protein and microfibrils accompanied by the assembly and disassembly of accessory proteins. Age-related morphological, structural, and functional proinflammatory remodeling within the elastic fiber has a profound effect upon the integrity, elasticity, calcification, amyloidosis, and stiffness of the large arterial wall. An age-associated increase in arterial stiffness is a major risk factor for the pathogenesis of diseases of the large arteries such as hypertensive and atherosclerotic vasculopathy. This mini review is an update on the key molecular, cellular, functional, and structural mechanisms of elastic fiber proinflammatory remodeling in large arteries with aging. Targeting structural and functional integrity of the elastic fiber may be an effective approach to impede proinflammatory arterial remodeling with advancing age.
Collapse
Affiliation(s)
- Soo Hyuk Kim
- Laboratory of Cardiovascular Science, Intramural Research Program, National Institution on Aging, National Institutes of Health, Biomedical Research Center (BRC), 251 Bayview Boulevard, Baltimore, MD, 21224, USA
| | - Robert E Monticone
- Laboratory of Cardiovascular Science, Intramural Research Program, National Institution on Aging, National Institutes of Health, Biomedical Research Center (BRC), 251 Bayview Boulevard, Baltimore, MD, 21224, USA
| | - Kimberly R McGraw
- Laboratory of Cardiovascular Science, Intramural Research Program, National Institution on Aging, National Institutes of Health, Biomedical Research Center (BRC), 251 Bayview Boulevard, Baltimore, MD, 21224, USA
| | - Mingyi Wang
- Laboratory of Cardiovascular Science, Intramural Research Program, National Institution on Aging, National Institutes of Health, Biomedical Research Center (BRC), 251 Bayview Boulevard, Baltimore, MD, 21224, USA.
| |
Collapse
|
11
|
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
|
12
|
Lerch S, Zuber R, Gehring N, Wang Y, Eckel B, Klass KD, Lehmann FO, Moussian B. Resilin matrix distribution, variability and function in Drosophila. BMC Biol 2020; 18:195. [PMID: 33317537 PMCID: PMC7737337 DOI: 10.1186/s12915-020-00902-4] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2020] [Accepted: 10/19/2020] [Indexed: 11/23/2022] Open
Abstract
Background Elasticity prevents fatigue of tissues that are extensively and repeatedly deformed. Resilin is a resilient and elastic extracellular protein matrix in joints and hinges of insects. For its mechanical properties, Resilin is extensively analysed and applied in biomaterial and biomedical sciences. However, there is only indirect evidence for Resilin distribution and function in an insect. Commonly, the presence of dityrosines that covalently link Resilin protein monomers (Pro-Resilin), which are responsible for its mechanical properties and fluoresce upon UV excitation, has been considered to reflect Resilin incidence. Results Using a GFP-tagged Resilin version, we directly identify Resilin in pliable regions of the Drosophila body, some of which were not described before. Interestingly, the amounts of dityrosines are not proportional to the amounts of Resilin in different areas of the fly body, arguing that the mechanical properties of Resilin matrices vary according to their need. For a functional analysis of Resilin matrices, applying the RNA interference and Crispr/Cas9 techniques, we generated flies with reduced or eliminated Resilin function, respectively. We find that these flies are flightless but capable of locomotion and viable suggesting that other proteins may partially compensate for Resilin function. Indeed, localizations of the potentially elastic protein Cpr56F and Resilin occasionally coincide. Conclusions Thus, Resilin-matrices are composite in the way that varying amounts of different elastic proteins and dityrosinylation define material properties. Understanding the biology of Resilin will have an impact on Resilin-based biomaterial and biomedical sciences.
Collapse
Affiliation(s)
- Steven Lerch
- Applied Zoology, Technical University of Dresden, Dresden, Germany.,Animal Genetics, Interfaculty Institute of Cell Biology, University of Tübingen, Tübingen, Germany.,Senckenberg Natural History Collections, Dresden, Germany
| | - Renata Zuber
- Applied Zoology, Technical University of Dresden, Dresden, Germany
| | - Nicole Gehring
- Animal Genetics, Interfaculty Institute of Cell Biology, University of Tübingen, Tübingen, Germany
| | - Yiwen Wang
- Animal Genetics, Interfaculty Institute of Cell Biology, University of Tübingen, Tübingen, Germany
| | - Barbara Eckel
- Applied Zoology, Technical University of Dresden, Dresden, Germany
| | | | | | - Bernard Moussian
- Applied Zoology, Technical University of Dresden, Dresden, Germany. .,Animal Genetics, Interfaculty Institute of Cell Biology, University of Tübingen, Tübingen, Germany. .,CNRS, Inserm Institute of Biology Valrose, Université Côte d'Azur, Nice, France.
| |
Collapse
|
13
|
Fedintsev A, Moskalev A. Stochastic non-enzymatic modification of long-lived macromolecules - A missing hallmark of aging. Ageing Res Rev 2020; 62:101097. [PMID: 32540391 DOI: 10.1016/j.arr.2020.101097] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Revised: 05/05/2020] [Accepted: 06/04/2020] [Indexed: 12/12/2022]
Abstract
Damage accumulation in long-living macromolecules (especially extracellular matrix (ECM) proteins, nuclear pore complex (NPC) proteins, and histones) is a missing hallmark of aging. Stochastic non-enzymatic modifications of ECM trigger cellular senescence as well as many other hallmarks of aging affect organ barriers integrity and drive tissue fibrosis. The importance of it for aging makes it a key target for interventions. The most promising of them can be AGE inhibitors (chelators, O-acetyl group or transglycating activity compounds, amadorins and amadoriases), glucosepane breakers, stimulators of elastogenesis, and RAGE antagonists.
Collapse
Affiliation(s)
- Alexander Fedintsev
- Institute of Biology of FRC of Komi Scientific Center, Ural Branch of Russian Academy of Sciences, Syktyvkar, Russia
| | - Alexey Moskalev
- Institute of Biology of FRC of Komi Scientific Center, Ural Branch of Russian Academy of Sciences, Syktyvkar, Russia.
| |
Collapse
|
14
|
Kinnear C, Agrawal R, Loo C, Pahnke A, Rodrigues DC, Thompson T, Akinrinade O, Ahadian S, Keeley F, Radisic M, Mital S, Ellis J. Everolimus Rescues the Phenotype of Elastin Insufficiency in Patient Induced Pluripotent Stem Cell-Derived Vascular Smooth Muscle Cells. Arterioscler Thromb Vasc Biol 2020; 40:1325-1339. [PMID: 32212852 PMCID: PMC7176340 DOI: 10.1161/atvbaha.119.313936] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Supplemental Digital Content is available in the text. Objective: Elastin gene deletion or mutation leads to arterial stenoses due to vascular smooth muscle cell (SMC) proliferation. Human induced pluripotent stem cells–derived SMCs can model the elastin insufficiency phenotype in vitro but show only partial rescue with rapamycin. Our objective was to identify drug candidates with superior efficacy in rescuing the SMC phenotype in elastin insufficiency patients. Approach and Results: SMCs generated from induced pluripotent stem cells from 5 elastin insufficiency patients with severe recurrent vascular stenoses (3 Williams syndrome and 2 elastin mutations) were phenotypically immature, hyperproliferative, poorly responsive to endothelin, and exerted reduced tension in 3-dimensional smooth muscle biowires. Elastin mRNA and protein were reduced in SMCs from patients compared to healthy control SMCs. Fourteen drug candidates were tested on patient SMCs. Of the mammalian target of rapamycin inhibitors studied, everolimus restored differentiation, rescued proliferation, and improved endothelin-induced calcium flux in all patient SMCs except one Williams syndrome. Of the calcium channel blockers, verapamil increased SMC differentiation and reduced proliferation in Williams syndrome patient cells but not in elastin mutation patients and had no effect on endothelin response. Combination treatment with everolimus and verapamil was not superior to everolimus alone. Other drug candidates had limited efficacy. Conclusions: Everolimus caused the most consistent improvement in SMC differentiation, proliferation and in SMC function in patients with both syndromic and nonsyndromic elastin insufficiency, and offers the best candidate for drug repurposing for treatment of elastin insufficiency associated vasculopathy.
Collapse
Affiliation(s)
- Caroline Kinnear
- From the Program in Genetics and Genome Biology, The Hospital for Sick Children, Toronto, Ontario, Canada (C.K., R.A., O.A., S.M.)
| | - Rahul Agrawal
- From the Program in Genetics and Genome Biology, The Hospital for Sick Children, Toronto, Ontario, Canada (C.K., R.A., O.A., S.M.)
| | - Caitlin Loo
- Program in Developmental and Stem Cell Biology, The Hospital for Sick Children, Toronto, Ontario, Canada (C.L., D.C.R., T.T., J.E.).,Department of Molecular Genetics (C.L., J.E.), University of Toronto, Ontario, Canada
| | - Aric Pahnke
- Institute of Biomaterials and Biomedical Engineering (A.P., S.A., M.R.), University of Toronto, Ontario, Canada.,Department of Chemical Engineering and Applied Chemistry (A.P., S.A., M.R.), University of Toronto, Ontario, Canada
| | - Deivid Carvalho Rodrigues
- Program in Developmental and Stem Cell Biology, The Hospital for Sick Children, Toronto, Ontario, Canada (C.L., D.C.R., T.T., J.E.)
| | - Tadeo Thompson
- Program in Developmental and Stem Cell Biology, The Hospital for Sick Children, Toronto, Ontario, Canada (C.L., D.C.R., T.T., J.E.)
| | - Oyediran Akinrinade
- From the Program in Genetics and Genome Biology, The Hospital for Sick Children, Toronto, Ontario, Canada (C.K., R.A., O.A., S.M.)
| | - Samad Ahadian
- Institute of Biomaterials and Biomedical Engineering (A.P., S.A., M.R.), University of Toronto, Ontario, Canada.,Department of Chemical Engineering and Applied Chemistry (A.P., S.A., M.R.), University of Toronto, Ontario, Canada
| | - Fred Keeley
- Department of Biochemistry (F.K.), University of Toronto, Ontario, Canada.,Program in Molecular Medicine, The Hospital for Sick Children, Toronto, Ontario, Canada (F.K.)
| | - Milica Radisic
- Institute of Biomaterials and Biomedical Engineering (A.P., S.A., M.R.), University of Toronto, Ontario, Canada.,Department of Chemical Engineering and Applied Chemistry (A.P., S.A., M.R.), University of Toronto, Ontario, Canada
| | - Seema Mital
- From the Program in Genetics and Genome Biology, The Hospital for Sick Children, Toronto, Ontario, Canada (C.K., R.A., O.A., S.M.).,Department of Pediatrics, The Hospital for Sick Children (S.M.), University of Toronto, Ontario, Canada
| | - James Ellis
- Program in Developmental and Stem Cell Biology, The Hospital for Sick Children, Toronto, Ontario, Canada (C.L., D.C.R., T.T., J.E.).,Department of Molecular Genetics (C.L., J.E.), University of Toronto, Ontario, Canada
| |
Collapse
|
15
|
Stenosis coexists with compromised α1-adrenergic contractions in the ascending aorta of a mouse model of Williams-Beuren syndrome. Sci Rep 2020; 10:889. [PMID: 31965005 PMCID: PMC6972706 DOI: 10.1038/s41598-020-57803-3] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2019] [Accepted: 01/07/2020] [Indexed: 11/08/2022] Open
Abstract
Williams-Beuren syndrome (WBS) is a rare disorder caused by a heterozygous deletion of 26–28 contiguous genes that affects the brain and cardiovascular system. Here, we investigated whether WBS affects aortic structure and function in the complete deletion (CD) mouse model harbouring the most common deletion found in WBS patients. Thoracic aortas from 3–4 months-old male CD mice and wild-type littermates were mounted in wire myographs or were processed for histomorphometrical analysis. Nitric oxide synthase (NOS) isoforms and oxidative stress levels were assessed. Ascending aortas from young adult CD mice showed moderate (50%) luminal stenosis, whereas endothelial function and oxidative stress were comparable to wild-type. CD mice showed greater contractions to KCl. However, α1-adrenergic contractions to phenylephrine, but not with a thromboxane analogue, were compromised. Decreased phenylephrine responses were not affected by selective inducible NOS blockade with 1400 W, but were prevented by the non-selective NOS inhibitor L-NAME and the selective neuronal NOS inhibitor SMTC. Consistently, CD mice showed increased neuronal NOS expression in aortas. Overall, aortic stenosis in CD mice coexists with excessive nNOS-derived NO signaling that compromises ascending aorta α1-adrenergic contractions. We suggest that increased neuronal NOS signaling may act as a physiological ‘brake’ against the detrimental effects of stenosis.
Collapse
|
16
|
Vindin H, Mithieux SM, Weiss AS. Elastin architecture. Matrix Biol 2019; 84:4-16. [DOI: 10.1016/j.matbio.2019.07.005] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2019] [Revised: 07/08/2019] [Accepted: 07/08/2019] [Indexed: 11/15/2022]
|
17
|
Lin CJ, Staiculescu MC, Hawes JZ, Cocciolone AJ, Hunkins BM, Roth RA, Lin CY, Mecham RP, Wagenseil JE. Heterogeneous Cellular Contributions to Elastic Laminae Formation in Arterial Wall Development. Circ Res 2019; 125:1006-1018. [PMID: 31590613 DOI: 10.1161/circresaha.119.315348] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
RATIONALE Elastin is an important ECM (extracellular matrix) protein in large and small arteries. Vascular smooth muscle cells (SMCs) produce the layered elastic laminae found in elastic arteries but synthesize little elastin in muscular arteries. However, muscular arteries have a well-defined internal elastic lamina (IEL) that separates endothelial cells (ECs) from SMCs. The extent to which ECs contribute elastin to the IEL is unknown. OBJECTIVE To use targeted elastin (Eln) deletion in mice to explore the relative contributions of SMCs and ECs to elastic laminae formation in different arteries. METHODS AND RESULTS We used SMC- and EC-specific Cre recombinase transgenes with a novel floxed Eln allele to focus gene inactivation in mice. Inactivation of Eln in SMCs using Sm22aCre resulted in depletion of elastic laminae in the arterial wall with the exception of the IEL and SMC clusters in the outer media near the adventitia. Inactivation of elastin in ECs using Tie2Cre or Cdh5Cre resulted in normal medial elastin and a typical IEL in elastic arteries. In contrast, the IEL was absent or severely disrupted in muscular arteries. Interruptions in the IEL resulted in neointimal formation in the ascending aorta but not in muscular arteries. CONCLUSIONS Combined with lineage-specific fate mapping systems, our knockout results document an unexpected heterogeneity in vascular cells that produce the elastic laminae. SMCs and ECs can independently form an IEL in most elastic arteries, whereas ECs are the major source of elastin for the IEL in muscular and resistance arteries. Neointimal formation at IEL disruptions in the ascending aorta confirms that the IEL is a critical physical barrier between SMCs and ECs in the large elastic arteries. Our studies provide new information about how SMCs and ECs contribute elastin to the arterial wall and how local elastic laminae defects may contribute to cardiovascular disease.
Collapse
Affiliation(s)
- Chien-Jung Lin
- From the Department of Cell Biology and Physiology (C.-J.L., B.M.H., R.A.R., R.P.M.).,Department of Internal Medicine, Cardiovascular Division (C.-J.L.)
| | - Marius C Staiculescu
- Department of Mechanical Engineering and Materials Science (M.C.S., J.Z.H., J.E.W.)
| | - Jie Z Hawes
- Department of Mechanical Engineering and Materials Science (M.C.S., J.Z.H., J.E.W.)
| | - Austin J Cocciolone
- Departments of Biomedical Engineering (A.J.C.), Washington University, St. Louis, MO
| | - Bridget M Hunkins
- From the Department of Cell Biology and Physiology (C.-J.L., B.M.H., R.A.R., R.P.M.)
| | - Robyn A Roth
- From the Department of Cell Biology and Physiology (C.-J.L., B.M.H., R.A.R., R.P.M.)
| | - Chieh-Yu Lin
- Pathology and Immunology (C.-Y.L.), Washington University, St. Louis, MO
| | - Robert P Mecham
- From the Department of Cell Biology and Physiology (C.-J.L., B.M.H., R.A.R., R.P.M.)
| | - Jessica E Wagenseil
- Department of Mechanical Engineering and Materials Science (M.C.S., J.Z.H., J.E.W.)
| |
Collapse
|
18
|
Henninger HB, Ellis BJ, Scott SA, Weiss JA. Contributions of elastic fibers, collagen, and extracellular matrix to the multiaxial mechanics of ligament. J Mech Behav Biomed Mater 2019; 99:118-126. [PMID: 31351401 DOI: 10.1016/j.jmbbm.2019.07.018] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2018] [Revised: 05/29/2019] [Accepted: 07/19/2019] [Indexed: 12/21/2022]
Abstract
Elastin is a biopolymer known to provide resilience to extensible biologic tissues through elastic recoil of its highly crosslinked molecular network. Recent studies have demonstrated that elastic fibers in ligament provide significant resistance to tensile and especially shear stress. We hypothesized that the biomechanics of elastic fibers in ligament could be described as transversely isotropic with both fiber and matrix components in a multi-material mixture. Similarly, we hypothesized that material coefficients derived using the experimental tensile response could be used to predict the experimental shear response. Experimental data for uniaxial and transverse tensile testing of control tissues, and those enzymatically digested to disrupt elastin, were used as inputs to a material coefficient optimization algorithm. An additive decomposition of the strain energy was used to model the total stress as the sum of contributions from collagen fibers, elastic fibers, elastic matrix, and ground substance matrix. Matrices were modeled as isotropic Veronda-Westmann hyperelastic materials, whereas fiber families were modeled as piecewise exponential-linear hyperelastic materials. Optimizations provided excellent fits to the tensile experimental data for each treatment case and material model. Given the disparity in magnitude of stresses between longitudinal and transverse/shear tests and agreement between models and experiments, the hypothesized transversely isotropic material of elastin symmetry was supported. In addition, the coefficients derived from uniaxial and transverse tensile experiments provided reasonable predictions of the experimental behavior during shear deformation. The magnitudes of coefficients representing stress, nonlinearity, and stiffness supported the experimental evidence that elastic fibers dominate the low strain tensile and shear response of ligament. These findings demonstrate that the additive decomposition modeling strategy can represent each discrete fiber and matrix constituent and their relative contribution to the material response of the tissue. These experimental data and the validated constitutive model provide essential inputs and a framework to refine existing computational models of ligament and tendon mechanics by explicitly representing the mechanical contributions of elastic fibers.
Collapse
Affiliation(s)
- Heath B Henninger
- Department of Biomedical Engineering, University of Utah, Salt Lake City, UT, USA; Scientific Computing and Imaging Institute, University of Utah, Salt Lake City, UT, USA; Department of Orthopaedics, University of Utah, Salt Lake City, UT, USA
| | - Benjamin J Ellis
- Department of Biomedical Engineering, University of Utah, Salt Lake City, UT, USA; Scientific Computing and Imaging Institute, University of Utah, Salt Lake City, UT, USA
| | - Sara A Scott
- Department of Biomedical Engineering, University of Utah, Salt Lake City, UT, USA
| | - Jeffrey A Weiss
- Department of Biomedical Engineering, University of Utah, Salt Lake City, UT, USA; Scientific Computing and Imaging Institute, University of Utah, Salt Lake City, UT, USA; Department of Orthopaedics, University of Utah, Salt Lake City, UT, USA.
| |
Collapse
|
19
|
Angelov SN, Zhu J, Hu JH, Dichek DA. What's the Skinny on Elastin Deficiency and Supravalvular Aortic Stenosis? Arterioscler Thromb Vasc Biol 2019; 37:740-742. [PMID: 28446468 DOI: 10.1161/atvbaha.117.309257] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Affiliation(s)
- Stoyan N Angelov
- From the Department of Medicine, University of Washington School of Medicine, Seattle
| | - Jay Zhu
- From the Department of Medicine, University of Washington School of Medicine, Seattle
| | - Jie Hong Hu
- From the Department of Medicine, University of Washington School of Medicine, Seattle
| | - David A Dichek
- From the Department of Medicine, University of Washington School of Medicine, Seattle.
| |
Collapse
|
20
|
Chang CJ, Hsu HC, Ho WJ, Chang GJ, Pang JHS, Chen WJ, Huang CC, Lai YJ. Cathepsin S promotes the development of pulmonary arterial hypertension. Am J Physiol Lung Cell Mol Physiol 2019; 317:L1-L13. [PMID: 31017016 DOI: 10.1152/ajplung.00530.2018] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Cysteine cathepsin proteases play critical roles in cardiovascular disease progression and are implicated in extracellular matrix (ECM) degradation. Patients with pulmonary arterial hypertension (PAH) exhibit increased elastase production by pulmonary arterial smooth muscle cells (PASMCs), which is related to the degradation of elastic fibers and pulmonary vascular remodeling. However, the mechanism by which cathepsins regulate the ECM and PASMC proliferation in PAH remains unclear. We hypothesized that cathepsin proteases in PASMCs promote the development of PAH. Here, we show overexpression of cathepsin S (Cat S) and degradation of elastic laminae in the lungs of patients with idiopathic PAH and in the PASMCs of monocrotaline-induced PAH model (MCT-PAH) rats. In addition, pulmonary hypertension can be treated in MCT-PAH rats by administering a selective Cat S inhibitor, Millipore-219393, which stimulates peroxisome proliferator-activated receptor-γ (PPARγ) to inhibit the expression of Cat S, thus suppressing the proliferation and migration of MCT-PAH PASMCs. We then reduced Cat S or PPARγ expression by using small interfering RNA in human PASMCs to demonstrate a mechanistic link between Cat S signaling and PPARγ protein, and the results suggest that PPARγ is upstream of Cat S signaling. In conclusion, the activity of Cat S in pulmonary vascular remodeling and degradation of elastin fibers through the disruption of PPARγ is pathophysiologically significant in PAH.
Collapse
Affiliation(s)
- Chi-Jen Chang
- Cardiovascular Division, Chang Gung Memorial Hospital , Taoyuan , Taiwan
| | - Hsiu-Chi Hsu
- Department of Respiratory Therapy, Chang Gung University College of Medicine , Taoyuan , Taiwan.,Graduate Institute of Clinical Medical Sciences, Chang Gung University College of Medicine , Taoyuan , Taiwan
| | - Wan-Jing Ho
- Cardiovascular Division, Chang Gung Memorial Hospital , Taoyuan , Taiwan
| | - Gwo-Jyh Chang
- Graduate Institute of Clinical Medical Sciences, Chang Gung University College of Medicine , Taoyuan , Taiwan
| | - Jong-Hwei S Pang
- Graduate Institute of Clinical Medical Sciences, Chang Gung University College of Medicine , Taoyuan , Taiwan.,Department of Physical Medicine and Rehabilitation, Chang Gung Memorial Hospital , Taoyuan , Taiwan
| | - Wei-Jan Chen
- Cardiovascular Division, Chang Gung Memorial Hospital , Taoyuan , Taiwan
| | - Chung-Chi Huang
- Department of Respiratory Therapy, Chang Gung University College of Medicine , Taoyuan , Taiwan.,Division of Thoracic Medicine, Chang Gung Memorial Hospital , Taoyuan , Taiwan
| | - Ying-Ju Lai
- Cardiovascular Division, Chang Gung Memorial Hospital , Taoyuan , Taiwan.,Department of Respiratory Therapy, Chang Gung University College of Medicine , Taoyuan , Taiwan.,Department of Respiratory Care, Chang Gung University of Science and Technology, Chiayi, Taiwan
| |
Collapse
|
21
|
Jiang Y, Luo Y, Tang Y, Moats R, Warburton D, Zhou S, Lou J, Pryhuber GS, Shi W, Wang LL. Alteration of cystic airway mesenchyme in congenital pulmonary airway malformation. Sci Rep 2019; 9:5296. [PMID: 30923323 PMCID: PMC6439218 DOI: 10.1038/s41598-019-41777-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2017] [Accepted: 03/08/2019] [Indexed: 12/21/2022] Open
Abstract
Congenital pulmonary airway malformation (CPAM) is the most common congenital lesion detected in the neonatal lung, which may lead to respiratory distress, infection, and pneumothorax. CPAM is thought to result from abnormal branching morphogenesis during fetal lung development, arising from different locations within the developing respiratory tract. However, the pathogenic mechanisms are unknown, and previous studies have focused on abnormalities in airway epithelial cells. We have analyzed 13 excised lung specimens from infants (age < 1 year) with a confirmed diagnosis of type 2 CPAM, which is supposed to be derived from abnormal growth of intrapulmonary distal airways. By examining the mesenchymal components including smooth muscle cells, laminin, and elastin in airway and cystic walls using immunofluorescence staining, we found that the thickness and area of the smooth muscle layer underlining the airway cysts in these CPAM tissue sections were significantly decreased compared with those in bronchiolar walls of normal controls. Extracellular elastin fibers were also visually reduced or absent in airway cystic walls. In particular, a layer of elastin fibers seen in normal lung between airway epithelia and underlying smooth muscle cells was missing in type 2 CPAM samples. Thus, our data demonstrate for the first time that airway cystic lesions in type 2 CPAM occur not only in airway epithelial cells, but also in adjacent mesenchymal tissues, including airway smooth muscle cells and their extracellular protein products. This provides a new direction to study the molecular and cellular mechanisms of CPAM pathogenesis in human.
Collapse
Affiliation(s)
- Yi Jiang
- Department of Pathology, the Second Xiangya Hospital of Central South University, Changsha, China.,Developmental Biology and Regenerative Medicine Program, Children's Hospital Los Angeles, Los Angeles, CA, 90027, USA
| | - Yongfeng Luo
- Developmental Biology and Regenerative Medicine Program, Children's Hospital Los Angeles, Los Angeles, CA, 90027, USA
| | - Yang Tang
- Developmental Biology and Regenerative Medicine Program, Children's Hospital Los Angeles, Los Angeles, CA, 90027, USA
| | - Rex Moats
- Developmental Biology and Regenerative Medicine Program, Children's Hospital Los Angeles, Los Angeles, CA, 90027, USA
| | - David Warburton
- Developmental Biology and Regenerative Medicine Program, Children's Hospital Los Angeles, Los Angeles, CA, 90027, USA
| | - Shengmei Zhou
- Department of Pathology and Laboratory Medicine, Children's Hospital Los Angeles, Los Angeles, CA, 90027, USA
| | - Jianlin Lou
- Institute of Occupational Diseases, Zhejiang Academy of Medical Sciences, Hangzhou, China
| | - Gloria S Pryhuber
- Department of Pediatrics, University of Rochester School of Medicine and Dentistry, Rochester, NY, 14642, USA
| | - Wei Shi
- Developmental Biology and Regenerative Medicine Program, Children's Hospital Los Angeles, Los Angeles, CA, 90027, USA.
| | - Larry L Wang
- Department of Pathology and Laboratory Medicine, Children's Hospital Los Angeles, Los Angeles, CA, 90027, USA.
| |
Collapse
|
22
|
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: 41] [Impact Index Per Article: 8.2] [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
|
23
|
Majesky MW. Vascular Development. Arterioscler Thromb Vasc Biol 2019; 38:e17-e24. [PMID: 29467221 DOI: 10.1161/atvbaha.118.310223] [Citation(s) in RCA: 56] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2018] [Accepted: 01/22/2018] [Indexed: 12/13/2022]
Abstract
The vascular system forms as a branching network of endothelial cells that acquire identity as arterial, venous, hemogenic, or lymphatic. Endothelial specification depends on gene targets transcribed by Ets domain-containing factors, including Ets variant gene 2 (Etv2), together with the activity of chromatin-remodeling complexes containing Brahma-related gene-1 (Brg1). Once specified and assembled into vessels, mechanisms regulating lumen diameter and axial growth ensure that the structure of the branching vascular network matches the need for perfusion of target tissues. In addition, blood vessels provide important morphogenic cues that guide or direct the development of organs forming around them. As the embryo grows and lumen diameters increase, smooth muscle cells wrap around the nascent vessel walls to provide mechanical strength and vasomotor control of the circulation. Increasing mechanical stretch and wall strain promote smooth muscle cell differentiation via coupling of actin cytoskeletal remodeling to myocardin and serum response factor-dependent transcription. Remodeling of artery walls by developmental signaling pathways reappears in postnatal blood vessels during physiological and pathological adaptation to vessel wall injury, inflammation, or chronic hypoxia. Recent reports providing insights into major steps in vascular development are reviewed here with a particular emphasis on studies that have been recently published in Arteriosclerosis, Thrombosis, and Vascular Biology.
Collapse
Affiliation(s)
- Mark W Majesky
- From the Center for Developmental Biology and Regenerative Medicine, Seattle Children's Research Institute, WA; and Departments of Pediatrics and Pathology, University of Washington, Seattle.
| |
Collapse
|
24
|
Spatial and temporal changes in extracellular elastin and laminin distribution during lung alveolar development. Sci Rep 2018; 8:8334. [PMID: 29844468 PMCID: PMC5974327 DOI: 10.1038/s41598-018-26673-1] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2017] [Accepted: 05/17/2018] [Indexed: 12/14/2022] Open
Abstract
Lung alveolarization requires precise coordination of cell growth with extracellular matrix (ECM) synthesis and deposition. The role of extracellular matrices in alveogenesis is not fully understood, because prior knowledge is largely extrapolated from two-dimensional structural analysis. Herein, we studied temporospatial changes of two important ECM proteins, laminin and elastin that are tightly associated with alveolar capillary growth and lung elastic recoil respectively, during both mouse and human lung alveolarization. By combining protein immunofluorescence staining with two- and three-dimensional imaging, we found that the laminin network was simplified along with the thinning of septal walls during alveogenesis, and more tightly associated with alveolar endothelial cells in matured lung. In contrast, elastin fibers were initially localized to the saccular openings of nascent alveoli, forming a ring-like structure. Then, throughout alveolar growth, the number of such alveolar mouth ring-like structures increased, while the relative ring size decreased. These rings were interconnected via additional elastin fibers. The apparent patches and dots of elastin at the tips of alveolar septae found in two-dimensional images were cross sections of elastin ring fibers in the three-dimension. Thus, the previous concept that deposition of elastin at alveolar tips drives septal inward growth may potentially be conceptually challenged by our data.
Collapse
|
25
|
Cocciolone AJ, Hawes JZ, Staiculescu MC, Johnson EO, Murshed M, Wagenseil JE. Elastin, arterial mechanics, and cardiovascular disease. Am J Physiol Heart Circ Physiol 2018; 315:H189-H205. [PMID: 29631368 DOI: 10.1152/ajpheart.00087.2018] [Citation(s) in RCA: 167] [Impact Index Per Article: 27.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Large, elastic arteries are composed of cells and a specialized extracellular matrix that provides reversible elasticity and strength. Elastin is the matrix protein responsible for this reversible elasticity that reduces the workload on the heart and dampens pulsatile flow in distal arteries. Here, we summarize the elastin protein biochemistry, self-association behavior, cross-linking process, and multistep elastic fiber assembly that provide large arteries with their unique mechanical properties. We present measures of passive arterial mechanics that depend on elastic fiber amounts and integrity such as the Windkessel effect, structural and material stiffness, and energy storage. We discuss supravalvular aortic stenosis and autosomal dominant cutis laxa-1, which are genetic disorders caused by mutations in the elastin gene. We present mouse models of supravalvular aortic stenosis, autosomal dominant cutis laxa-1, and graded elastin amounts that have been invaluable for understanding the role of elastin in arterial mechanics and cardiovascular disease. We summarize acquired diseases associated with elastic fiber defects, including hypertension and arterial stiffness, diabetes, obesity, atherosclerosis, calcification, and aneurysms and dissections. We mention animal models that have helped delineate the role of elastic fiber defects in these acquired diseases. We briefly summarize challenges and recent advances in generating functional elastic fibers in tissue-engineered arteries. We conclude with suggestions for future research and opportunities for therapeutic intervention in genetic and acquired elastinopathies.
Collapse
Affiliation(s)
- Austin J Cocciolone
- Department of Biomedical Engineering, Washington University , St. Louis, Missouri
| | - Jie Z Hawes
- Department of Mechanical Engineering and Materials Science, Washington University , St. Louis, Missouri
| | - Marius C Staiculescu
- Department of Mechanical Engineering and Materials Science, Washington University , St. Louis, Missouri
| | - Elizabeth O Johnson
- Department of Mechanical Engineering and Materials Science, Washington University , St. Louis, Missouri
| | - Monzur Murshed
- Faculty of Dentistry, Department of Medicine, and Shriners Hospital for Children, McGill University , Montreal, Quebec , Canada
| | - Jessica E Wagenseil
- Department of Mechanical Engineering and Materials Science, Washington University , St. Louis, Missouri
| |
Collapse
|
26
|
Yeo J, Jung G, Tarakanova A, Martín-Martínez FJ, Qin Z, Cheng Y, Zhang YW, Buehler MJ. Multiscale modeling of keratin, collagen, elastin and related human diseases: Perspectives from atomistic to coarse-grained molecular dynamics simulations. EXTREME MECHANICS LETTERS 2018; 20:112-124. [PMID: 33344740 PMCID: PMC7745951 DOI: 10.1016/j.eml.2018.01.009] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
Scleroproteins are an important category of proteins within the human body that adopt filamentous, elongated conformations in contrast with typical globular proteins. These include keratin, collagen, and elastin, which often serve a common mechanical function in structural support of cells and tissues. Genetic mutations alter these proteins, disrupting their functions and causing diseases. Computational characterization of these mutations has proven to be extremely valuable in identifying the intricate structure-function relationships of scleroproteins from the molecular scale up, especially if combined with multiscale experimental analysis and the synthesis of model proteins to test specific structure-function relationships. In this work, we review numerous critical diseases that are related to keratin, collagen, and elastin, and through several case studies, we propose ways of extensively utilizing multiscale modeling, from atomistic to coarse-grained molecular dynamics simulations, to uncover the molecular origins for some of these diseases and to aid in the development of novel cures and therapies. As case studies, we examine the effects of the genetic disease Epidermolytic Hyperkeratosis (EHK) on the structure and aggregation of keratins 1 and 10; we propose models to understand the diseases of Osteogenesis Imperfecta (OI) and Alport syndrome (AS) that affect the mechanical and aggregation properties of collagen; and we develop atomistic molecular dynamics and elastic network models of elastin to determine the role of mutations in diseases such as Cutis Laxa and Supravalvular Aortic Stenosis on elastin's structure and molecular conformational motions and implications for assembly.
Collapse
Affiliation(s)
- Jingjie Yeo
- Laboratory for Atomistic and Molecular Mechanics, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
- Institute of High Performance Computing, Agency for Science, Technology and Research (A*STAR), Singapore 138632
| | - GangSeob Jung
- Laboratory for Atomistic and Molecular Mechanics, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Anna Tarakanova
- Laboratory for Atomistic and Molecular Mechanics, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Francisco J. Martín-Martínez
- Laboratory for Atomistic and Molecular Mechanics, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Zhao Qin
- Laboratory for Atomistic and Molecular Mechanics, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Yuan Cheng
- Institute of High Performance Computing, Agency for Science, Technology and Research (A*STAR), Singapore 138632
| | - Yong-Wei Zhang
- Institute of High Performance Computing, Agency for Science, Technology and Research (A*STAR), Singapore 138632
| | - Markus J. Buehler
- Laboratory for Atomistic and Molecular Mechanics, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| |
Collapse
|
27
|
Duque Lasio ML, Kozel BA. Elastin-driven genetic diseases. Matrix Biol 2018; 71-72:144-160. [PMID: 29501665 DOI: 10.1016/j.matbio.2018.02.021] [Citation(s) in RCA: 60] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2017] [Revised: 02/23/2018] [Accepted: 02/23/2018] [Indexed: 02/08/2023]
Abstract
Elastic fibers provide recoil to tissues that undergo repeated deformation, such as blood vessels, lungs and skin. Composed of elastin and its accessory proteins, the fibers are produced within a restricted developmental window and are stable for decades. Their eventual breakdown is associated with a loss of tissue resiliency and aging. Rare alteration of the elastin (ELN) gene produces disease by impacting protein dosage (supravalvar aortic stenosis, Williams Beuren syndrome and Williams Beuren region duplication syndrome) and protein function (autosomal dominant cutis laxa). This review highlights aspects of the elastin molecule and its assembly process that contribute to human disease and also discusses potential therapies aimed at treating diseases of elastin insufficiency.
Collapse
Affiliation(s)
| | - Beth A Kozel
- National Institutes of Health, National Heart Lung and Blood Institute, Bethesda, MD, USA.
| |
Collapse
|
28
|
Mecham RP. Elastin in lung development and disease pathogenesis. Matrix Biol 2018; 73:6-20. [PMID: 29331337 DOI: 10.1016/j.matbio.2018.01.005] [Citation(s) in RCA: 113] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2017] [Revised: 12/30/2017] [Accepted: 01/07/2018] [Indexed: 12/24/2022]
Abstract
Elastin is expressed in most tissues that require elastic recoil. The protein first appeared coincident with the closed circulatory system, and was critical for the evolutionary success of the vertebrate lineage. Elastin is expressed by multiple cell types in the lung, including mesothelial cells in the pleura, smooth muscle cells in airways and blood vessels, endothelial cells, and interstitial fibroblasts. This highly crosslinked protein associates with fibrillin-containing microfibrils to form the elastic fiber, which is the physiological structure that functions in the extracellular matrix. Elastic fibers can be woven into many different shapes depending on the mechanical needs of the tissue. In large pulmonary vessels, for example, elastin forms continuous sheets, or lamellae, that separate smooth muscle layers. Outside of the vasculature, elastic fibers form an extensive fiber network that originates in the central bronchi and inserts into the distal airspaces and visceral pleura. The fibrous cables form a looping system that encircle the alveolar ducts and terminal air spaces and ensures that applied force is transmitted equally to all parts of the lung. Normal lung function depends on proper secretion and assembly of elastin, and either inhibition of elastin fiber assembly or degradation of existing elastin results in lung dysfunction and disease.
Collapse
Affiliation(s)
- Robert P Mecham
- Department of Cell Biology & Physiology, Washington University School of Medicine, St. Louis, MO, USA.
| |
Collapse
|
29
|
Martínez-Revelles S, García-Redondo AB, Avendaño MS, Varona S, Palao T, Orriols M, Roque FR, Fortuño A, Touyz RM, Martínez-González J, Salaices M, Rodríguez C, Briones AM. Lysyl Oxidase Induces Vascular Oxidative Stress and Contributes to Arterial Stiffness and Abnormal Elastin Structure in Hypertension: Role of p38MAPK. Antioxid Redox Signal 2017; 27:379-397. [PMID: 28010122 PMCID: PMC5563924 DOI: 10.1089/ars.2016.6642] [Citation(s) in RCA: 84] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/29/2016] [Revised: 12/22/2016] [Accepted: 12/22/2016] [Indexed: 12/19/2022]
Abstract
AIMS Vascular stiffness, structural elastin abnormalities, and increased oxidative stress are hallmarks of hypertension. Lysyl oxidase (LOX) is an elastin crosslinking enzyme that produces H2O2 as a by-product. We addressed the interplay between LOX, oxidative stress, vessel stiffness, and elastin. RESULTS Angiotensin II (Ang II)-infused hypertensive mice and spontaneously hypertensive rats (SHR) showed increased vascular LOX expression and stiffness and an abnormal elastin structure. Mice over-expressing LOX in vascular smooth muscle cells (TgLOX) exhibited similar mechanical and elastin alterations to those of hypertensive models. LOX inhibition with β-aminopropionitrile (BAPN) attenuated mechanical and elastin alterations in TgLOX mice, Ang II-infused mice, and SHR. Arteries from TgLOX mice, Ang II-infused mice, and/or SHR exhibited increased vascular H2O2 and O2.- levels, NADPH oxidase activity, and/or mitochondrial dysfunction. BAPN prevented the higher oxidative stress in hypertensive models. Treatment of TgLOX and Ang II-infused mice and SHR with the mitochondrial-targeted superoxide dismutase mimetic mito-TEMPO, the antioxidant apocynin, or the H2O2 scavenger polyethylene glycol-conjugated catalase (PEG-catalase) reduced oxidative stress, vascular stiffness, and elastin alterations. Vascular p38 mitogen-activated protein kinase (p38MAPK) activation was increased in Ang II-infused and TgLOX mice and this effect was prevented by BAPN, mito-TEMPO, or PEG-catalase. SB203580, the p38MAPK inhibitor, normalized vessel stiffness and elastin structure in TgLOX mice. INNOVATION We identify LOX as a novel source of vascular reactive oxygen species and a new pathway involved in vascular stiffness and elastin remodeling in hypertension. CONCLUSION LOX up-regulation is associated with enhanced oxidative stress that promotes p38MAPK activation, elastin structural alterations, and vascular stiffness. This pathway contributes to vascular abnormalities in hypertension. Antioxid. Redox Signal. 27, 379-397.
Collapse
Affiliation(s)
- Sonia Martínez-Revelles
- Departamento de Farmacología, Universidad Autónoma de Madrid (UAM), Instituto de Investigación Hospital Universitario La Paz (IdiPAZ), Madrid, Spain
- CIBER de Enfermedades Cardiovasculares, Spain
| | - Ana B. García-Redondo
- Departamento de Farmacología, Universidad Autónoma de Madrid (UAM), Instituto de Investigación Hospital Universitario La Paz (IdiPAZ), Madrid, Spain
- CIBER de Enfermedades Cardiovasculares, Spain
| | - María S. Avendaño
- Departamento de Farmacología, Universidad Autónoma de Madrid (UAM), Instituto de Investigación Hospital Universitario La Paz (IdiPAZ), Madrid, Spain
| | - Saray Varona
- CIBER de Enfermedades Cardiovasculares, Spain
- Centro de Investigación Cardiovascular (CSIC-ICCC), IIB-Sant Pau, Barcelona, Spain
| | - Teresa Palao
- Departamento de Farmacología, Universidad Autónoma de Madrid (UAM), Instituto de Investigación Hospital Universitario La Paz (IdiPAZ), Madrid, Spain
| | - Mar Orriols
- CIBER de Enfermedades Cardiovasculares, Spain
- Centro de Investigación Cardiovascular (CSIC-ICCC), IIB-Sant Pau, Barcelona, Spain
| | - Fernanda R. Roque
- Departamento de Farmacología, Universidad Autónoma de Madrid (UAM), Instituto de Investigación Hospital Universitario La Paz (IdiPAZ), Madrid, Spain
| | - Ana Fortuño
- Program of Cardiovascular Diseases, Centre for Applied Medical Research, University of Navarra, Pamplona, Spain
| | - Rhian M. Touyz
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, United Kingdom
| | - Jose Martínez-González
- CIBER de Enfermedades Cardiovasculares, Spain
- Centro de Investigación Cardiovascular (CSIC-ICCC), IIB-Sant Pau, Barcelona, Spain
| | - Mercedes Salaices
- Departamento de Farmacología, Universidad Autónoma de Madrid (UAM), Instituto de Investigación Hospital Universitario La Paz (IdiPAZ), Madrid, Spain
- CIBER de Enfermedades Cardiovasculares, Spain
| | - Cristina Rodríguez
- CIBER de Enfermedades Cardiovasculares, Spain
- Centro de Investigación Cardiovascular (CSIC-ICCC), IIB-Sant Pau, Barcelona, Spain
| | - Ana M. Briones
- Departamento de Farmacología, Universidad Autónoma de Madrid (UAM), Instituto de Investigación Hospital Universitario La Paz (IdiPAZ), Madrid, Spain
- CIBER de Enfermedades Cardiovasculares, Spain
| |
Collapse
|
30
|
Jiao Y, Li G, Li Q, Ali R, Qin L, Li W, Qyang Y, Greif DM, Geirsson A, Humphrey JD, Tellides G. mTOR (Mechanistic Target of Rapamycin) Inhibition Decreases Mechanosignaling, Collagen Accumulation, and Stiffening of the Thoracic Aorta in Elastin-Deficient Mice. Arterioscler Thromb Vasc Biol 2017; 37:1657-1666. [PMID: 28751568 DOI: 10.1161/atvbaha.117.309653] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2016] [Accepted: 07/17/2017] [Indexed: 12/20/2022]
Abstract
OBJECTIVE Elastin deficiency because of heterozygous loss of an ELN allele in Williams syndrome causes obstructive aortopathy characterized by medial thickening and fibrosis and consequent aortic stiffening. Previous work in Eln-null mice with a severe arterial phenotype showed that inhibition of mTOR (mechanistic target of rapamycin), a key regulator of cell growth, lessened the aortic obstruction but did not prevent early postnatal death. We investigated the effects of mTOR inhibition in Eln-null mice partially rescued by human ELN that manifest a less severe arterial phenotype and survive long term. APPROACH AND RESULTS Thoracic aortas of neonatal and juvenile mice with graded elastin deficiency exhibited increased signaling through both mTOR complex 1 and 2. Despite lower predicted wall stress, there was increased phosphorylation of focal adhesion kinase, suggestive of greater integrin activation, and increased transforming growth factor-β-signaling mediators, associated with increased collagen expression. Pharmacological blockade of mTOR by rapalogs did not improve luminal stenosis but reduced mechanosignaling (in delayed fashion after mTOR complex 1 inhibition), medial collagen accumulation, and stiffening of the aorta. Rapalog administration also retarded somatic growth, however, and precipitated neonatal deaths. Complementary, less-toxic strategies to inhibit mTOR via altered growth factor and nutrient responses were not effective. CONCLUSIONS In addition to previously demonstrated therapeutic benefits of rapalogs decreasing smooth muscle cell proliferation in the absence of elastin, we find that rapalogs also prevent aortic fibrosis and stiffening attributable to partial elastin deficiency. Our findings suggest that mTOR-sensitive perturbation of smooth muscle cell mechanosensing contributes to elastin aortopathy.
Collapse
Affiliation(s)
- Yang Jiao
- From the Department of Surgery (Y.J., G.L., Q.L., R.A., L.Q., W.L., A.G., G.T.), Department of Internal Medicine (Y.Q., D.M.G.), and Vascular Biology and Therapeutics Program (Y.Q., D.M.G., A.G., J.D.H., G.T.), Yale University School of Medicine, New Haven, CT; Department of Vascular Surgery, Peking University People's Hospital, Beijing, P. R. China (Y.J., Q.L., W.L.); Department of Biomedical Engineering, Yale University, New Haven, CT (J.D.H.); and Veterans Affairs Connecticut Healthcare System, West Haven (G.T.)
| | - Guangxin Li
- From the Department of Surgery (Y.J., G.L., Q.L., R.A., L.Q., W.L., A.G., G.T.), Department of Internal Medicine (Y.Q., D.M.G.), and Vascular Biology and Therapeutics Program (Y.Q., D.M.G., A.G., J.D.H., G.T.), Yale University School of Medicine, New Haven, CT; Department of Vascular Surgery, Peking University People's Hospital, Beijing, P. R. China (Y.J., Q.L., W.L.); Department of Biomedical Engineering, Yale University, New Haven, CT (J.D.H.); and Veterans Affairs Connecticut Healthcare System, West Haven (G.T.)
| | - Qingle Li
- From the Department of Surgery (Y.J., G.L., Q.L., R.A., L.Q., W.L., A.G., G.T.), Department of Internal Medicine (Y.Q., D.M.G.), and Vascular Biology and Therapeutics Program (Y.Q., D.M.G., A.G., J.D.H., G.T.), Yale University School of Medicine, New Haven, CT; Department of Vascular Surgery, Peking University People's Hospital, Beijing, P. R. China (Y.J., Q.L., W.L.); Department of Biomedical Engineering, Yale University, New Haven, CT (J.D.H.); and Veterans Affairs Connecticut Healthcare System, West Haven (G.T.)
| | - Rahmat Ali
- From the Department of Surgery (Y.J., G.L., Q.L., R.A., L.Q., W.L., A.G., G.T.), Department of Internal Medicine (Y.Q., D.M.G.), and Vascular Biology and Therapeutics Program (Y.Q., D.M.G., A.G., J.D.H., G.T.), Yale University School of Medicine, New Haven, CT; Department of Vascular Surgery, Peking University People's Hospital, Beijing, P. R. China (Y.J., Q.L., W.L.); Department of Biomedical Engineering, Yale University, New Haven, CT (J.D.H.); and Veterans Affairs Connecticut Healthcare System, West Haven (G.T.)
| | - Lingfeng Qin
- From the Department of Surgery (Y.J., G.L., Q.L., R.A., L.Q., W.L., A.G., G.T.), Department of Internal Medicine (Y.Q., D.M.G.), and Vascular Biology and Therapeutics Program (Y.Q., D.M.G., A.G., J.D.H., G.T.), Yale University School of Medicine, New Haven, CT; Department of Vascular Surgery, Peking University People's Hospital, Beijing, P. R. China (Y.J., Q.L., W.L.); Department of Biomedical Engineering, Yale University, New Haven, CT (J.D.H.); and Veterans Affairs Connecticut Healthcare System, West Haven (G.T.)
| | - Wei Li
- From the Department of Surgery (Y.J., G.L., Q.L., R.A., L.Q., W.L., A.G., G.T.), Department of Internal Medicine (Y.Q., D.M.G.), and Vascular Biology and Therapeutics Program (Y.Q., D.M.G., A.G., J.D.H., G.T.), Yale University School of Medicine, New Haven, CT; Department of Vascular Surgery, Peking University People's Hospital, Beijing, P. R. China (Y.J., Q.L., W.L.); Department of Biomedical Engineering, Yale University, New Haven, CT (J.D.H.); and Veterans Affairs Connecticut Healthcare System, West Haven (G.T.)
| | - Yibing Qyang
- From the Department of Surgery (Y.J., G.L., Q.L., R.A., L.Q., W.L., A.G., G.T.), Department of Internal Medicine (Y.Q., D.M.G.), and Vascular Biology and Therapeutics Program (Y.Q., D.M.G., A.G., J.D.H., G.T.), Yale University School of Medicine, New Haven, CT; Department of Vascular Surgery, Peking University People's Hospital, Beijing, P. R. China (Y.J., Q.L., W.L.); Department of Biomedical Engineering, Yale University, New Haven, CT (J.D.H.); and Veterans Affairs Connecticut Healthcare System, West Haven (G.T.)
| | - Daniel M Greif
- From the Department of Surgery (Y.J., G.L., Q.L., R.A., L.Q., W.L., A.G., G.T.), Department of Internal Medicine (Y.Q., D.M.G.), and Vascular Biology and Therapeutics Program (Y.Q., D.M.G., A.G., J.D.H., G.T.), Yale University School of Medicine, New Haven, CT; Department of Vascular Surgery, Peking University People's Hospital, Beijing, P. R. China (Y.J., Q.L., W.L.); Department of Biomedical Engineering, Yale University, New Haven, CT (J.D.H.); and Veterans Affairs Connecticut Healthcare System, West Haven (G.T.)
| | - Arnar Geirsson
- From the Department of Surgery (Y.J., G.L., Q.L., R.A., L.Q., W.L., A.G., G.T.), Department of Internal Medicine (Y.Q., D.M.G.), and Vascular Biology and Therapeutics Program (Y.Q., D.M.G., A.G., J.D.H., G.T.), Yale University School of Medicine, New Haven, CT; Department of Vascular Surgery, Peking University People's Hospital, Beijing, P. R. China (Y.J., Q.L., W.L.); Department of Biomedical Engineering, Yale University, New Haven, CT (J.D.H.); and Veterans Affairs Connecticut Healthcare System, West Haven (G.T.)
| | - Jay D Humphrey
- From the Department of Surgery (Y.J., G.L., Q.L., R.A., L.Q., W.L., A.G., G.T.), Department of Internal Medicine (Y.Q., D.M.G.), and Vascular Biology and Therapeutics Program (Y.Q., D.M.G., A.G., J.D.H., G.T.), Yale University School of Medicine, New Haven, CT; Department of Vascular Surgery, Peking University People's Hospital, Beijing, P. R. China (Y.J., Q.L., W.L.); Department of Biomedical Engineering, Yale University, New Haven, CT (J.D.H.); and Veterans Affairs Connecticut Healthcare System, West Haven (G.T.)
| | - George Tellides
- From the Department of Surgery (Y.J., G.L., Q.L., R.A., L.Q., W.L., A.G., G.T.), Department of Internal Medicine (Y.Q., D.M.G.), and Vascular Biology and Therapeutics Program (Y.Q., D.M.G., A.G., J.D.H., G.T.), Yale University School of Medicine, New Haven, CT; Department of Vascular Surgery, Peking University People's Hospital, Beijing, P. R. China (Y.J., Q.L., W.L.); Department of Biomedical Engineering, Yale University, New Haven, CT (J.D.H.); and Veterans Affairs Connecticut Healthcare System, West Haven (G.T.).
| |
Collapse
|
31
|
Jiao Y, Li G, Korneva A, Caulk AW, Qin L, Bersi MR, Li Q, Li W, Mecham RP, Humphrey JD, Tellides G. Deficient Circumferential Growth Is the Primary Determinant of Aortic Obstruction Attributable to Partial Elastin Deficiency. Arterioscler Thromb Vasc Biol 2017; 37:930-941. [PMID: 28254817 DOI: 10.1161/atvbaha.117.309079] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2016] [Accepted: 02/17/2017] [Indexed: 12/13/2022]
Abstract
OBJECTIVE Williams syndrome is characterized by obstructive aortopathy attributable to heterozygous loss of ELN, the gene encoding elastin. Lesions are thought to result primarily from excessive smooth muscle cell (SMC) proliferation and consequent medial expansion, although an initially smaller caliber and increased stiffness of the aorta may contribute to luminal narrowing. The relative contributions of such abnormalities to the obstructive phenotype had not been defined. APPROACH AND RESULTS We quantified determinants of luminal stenosis in thoracic aortas of Eln-/- mice incompletely rescued by human ELN. Moderate obstruction was largely because of deficient circumferential growth, most prominently of ascending segments, despite increased axial growth. Medial thickening was evident in these smaller diameter elastin-deficient aortas, with medial area similar to that of larger diameter control aortas. There was no difference in cross-sectional SMC number between mutant and wild-type genotypes at multiple stages of postnatal development. Decreased elastin content was associated with medial fibrosis and reduced aortic distensibility because of increased structural stiffness but preserved material stiffness. Elastin-deficient SMCs exhibited greater contractile-to-proliferative phenotypic modulation in vitro than in vivo. We confirmed increased medial collagen without evidence of increased medial area or SMC number in a small ascending aorta with thickened media of a Williams syndrome subject. CONCLUSIONS Deficient circumferential growth is the predominant mechanism for moderate obstructive aortic disease resulting from partial elastin deficiency. Our findings suggest that diverse aortic manifestations in Williams syndrome result from graded elastin content, and SMC hyperplasia causing medial expansion requires additional elastin loss superimposed on ELN haploinsufficiency.
Collapse
Affiliation(s)
- Yang Jiao
- From the Department of Surgery, Yale University School of Medicine, New Haven, CT (Y.J., G.L., L.Q., Q.L., W.L., G.T.); Department of Vascular Surgery, Peking University People's Hospital, Beijing, People's Republic of China (Y.J., Q.L., W.L.); Department of Vascular Surgery, The First Hospital of China Medical University, Shenyang, People's Republic of China (G.L.); Department of Biomedical Engineering, Yale University, New Haven, CT (A.K., A.W.C., M.R.B., J.D.H.); Department of Cell Biology, Washington University School of Medicine, St Louis, MO (R.P.M.); Interdepartmental Program in Vascular Biology and Therapeutics, Yale University School of Medicine (J.D.H., G.T.); and Veterans Affairs Connecticut Healthcare System, West Haven (G.T.)
| | - Guangxin Li
- From the Department of Surgery, Yale University School of Medicine, New Haven, CT (Y.J., G.L., L.Q., Q.L., W.L., G.T.); Department of Vascular Surgery, Peking University People's Hospital, Beijing, People's Republic of China (Y.J., Q.L., W.L.); Department of Vascular Surgery, The First Hospital of China Medical University, Shenyang, People's Republic of China (G.L.); Department of Biomedical Engineering, Yale University, New Haven, CT (A.K., A.W.C., M.R.B., J.D.H.); Department of Cell Biology, Washington University School of Medicine, St Louis, MO (R.P.M.); Interdepartmental Program in Vascular Biology and Therapeutics, Yale University School of Medicine (J.D.H., G.T.); and Veterans Affairs Connecticut Healthcare System, West Haven (G.T.)
| | - Arina Korneva
- From the Department of Surgery, Yale University School of Medicine, New Haven, CT (Y.J., G.L., L.Q., Q.L., W.L., G.T.); Department of Vascular Surgery, Peking University People's Hospital, Beijing, People's Republic of China (Y.J., Q.L., W.L.); Department of Vascular Surgery, The First Hospital of China Medical University, Shenyang, People's Republic of China (G.L.); Department of Biomedical Engineering, Yale University, New Haven, CT (A.K., A.W.C., M.R.B., J.D.H.); Department of Cell Biology, Washington University School of Medicine, St Louis, MO (R.P.M.); Interdepartmental Program in Vascular Biology and Therapeutics, Yale University School of Medicine (J.D.H., G.T.); and Veterans Affairs Connecticut Healthcare System, West Haven (G.T.)
| | - Alexander W Caulk
- From the Department of Surgery, Yale University School of Medicine, New Haven, CT (Y.J., G.L., L.Q., Q.L., W.L., G.T.); Department of Vascular Surgery, Peking University People's Hospital, Beijing, People's Republic of China (Y.J., Q.L., W.L.); Department of Vascular Surgery, The First Hospital of China Medical University, Shenyang, People's Republic of China (G.L.); Department of Biomedical Engineering, Yale University, New Haven, CT (A.K., A.W.C., M.R.B., J.D.H.); Department of Cell Biology, Washington University School of Medicine, St Louis, MO (R.P.M.); Interdepartmental Program in Vascular Biology and Therapeutics, Yale University School of Medicine (J.D.H., G.T.); and Veterans Affairs Connecticut Healthcare System, West Haven (G.T.)
| | - Lingfeng Qin
- From the Department of Surgery, Yale University School of Medicine, New Haven, CT (Y.J., G.L., L.Q., Q.L., W.L., G.T.); Department of Vascular Surgery, Peking University People's Hospital, Beijing, People's Republic of China (Y.J., Q.L., W.L.); Department of Vascular Surgery, The First Hospital of China Medical University, Shenyang, People's Republic of China (G.L.); Department of Biomedical Engineering, Yale University, New Haven, CT (A.K., A.W.C., M.R.B., J.D.H.); Department of Cell Biology, Washington University School of Medicine, St Louis, MO (R.P.M.); Interdepartmental Program in Vascular Biology and Therapeutics, Yale University School of Medicine (J.D.H., G.T.); and Veterans Affairs Connecticut Healthcare System, West Haven (G.T.)
| | - Matthew R Bersi
- From the Department of Surgery, Yale University School of Medicine, New Haven, CT (Y.J., G.L., L.Q., Q.L., W.L., G.T.); Department of Vascular Surgery, Peking University People's Hospital, Beijing, People's Republic of China (Y.J., Q.L., W.L.); Department of Vascular Surgery, The First Hospital of China Medical University, Shenyang, People's Republic of China (G.L.); Department of Biomedical Engineering, Yale University, New Haven, CT (A.K., A.W.C., M.R.B., J.D.H.); Department of Cell Biology, Washington University School of Medicine, St Louis, MO (R.P.M.); Interdepartmental Program in Vascular Biology and Therapeutics, Yale University School of Medicine (J.D.H., G.T.); and Veterans Affairs Connecticut Healthcare System, West Haven (G.T.)
| | - Qingle Li
- From the Department of Surgery, Yale University School of Medicine, New Haven, CT (Y.J., G.L., L.Q., Q.L., W.L., G.T.); Department of Vascular Surgery, Peking University People's Hospital, Beijing, People's Republic of China (Y.J., Q.L., W.L.); Department of Vascular Surgery, The First Hospital of China Medical University, Shenyang, People's Republic of China (G.L.); Department of Biomedical Engineering, Yale University, New Haven, CT (A.K., A.W.C., M.R.B., J.D.H.); Department of Cell Biology, Washington University School of Medicine, St Louis, MO (R.P.M.); Interdepartmental Program in Vascular Biology and Therapeutics, Yale University School of Medicine (J.D.H., G.T.); and Veterans Affairs Connecticut Healthcare System, West Haven (G.T.)
| | - Wei Li
- From the Department of Surgery, Yale University School of Medicine, New Haven, CT (Y.J., G.L., L.Q., Q.L., W.L., G.T.); Department of Vascular Surgery, Peking University People's Hospital, Beijing, People's Republic of China (Y.J., Q.L., W.L.); Department of Vascular Surgery, The First Hospital of China Medical University, Shenyang, People's Republic of China (G.L.); Department of Biomedical Engineering, Yale University, New Haven, CT (A.K., A.W.C., M.R.B., J.D.H.); Department of Cell Biology, Washington University School of Medicine, St Louis, MO (R.P.M.); Interdepartmental Program in Vascular Biology and Therapeutics, Yale University School of Medicine (J.D.H., G.T.); and Veterans Affairs Connecticut Healthcare System, West Haven (G.T.)
| | - Robert P Mecham
- From the Department of Surgery, Yale University School of Medicine, New Haven, CT (Y.J., G.L., L.Q., Q.L., W.L., G.T.); Department of Vascular Surgery, Peking University People's Hospital, Beijing, People's Republic of China (Y.J., Q.L., W.L.); Department of Vascular Surgery, The First Hospital of China Medical University, Shenyang, People's Republic of China (G.L.); Department of Biomedical Engineering, Yale University, New Haven, CT (A.K., A.W.C., M.R.B., J.D.H.); Department of Cell Biology, Washington University School of Medicine, St Louis, MO (R.P.M.); Interdepartmental Program in Vascular Biology and Therapeutics, Yale University School of Medicine (J.D.H., G.T.); and Veterans Affairs Connecticut Healthcare System, West Haven (G.T.)
| | - Jay D Humphrey
- From the Department of Surgery, Yale University School of Medicine, New Haven, CT (Y.J., G.L., L.Q., Q.L., W.L., G.T.); Department of Vascular Surgery, Peking University People's Hospital, Beijing, People's Republic of China (Y.J., Q.L., W.L.); Department of Vascular Surgery, The First Hospital of China Medical University, Shenyang, People's Republic of China (G.L.); Department of Biomedical Engineering, Yale University, New Haven, CT (A.K., A.W.C., M.R.B., J.D.H.); Department of Cell Biology, Washington University School of Medicine, St Louis, MO (R.P.M.); Interdepartmental Program in Vascular Biology and Therapeutics, Yale University School of Medicine (J.D.H., G.T.); and Veterans Affairs Connecticut Healthcare System, West Haven (G.T.)
| | - George Tellides
- From the Department of Surgery, Yale University School of Medicine, New Haven, CT (Y.J., G.L., L.Q., Q.L., W.L., G.T.); Department of Vascular Surgery, Peking University People's Hospital, Beijing, People's Republic of China (Y.J., Q.L., W.L.); Department of Vascular Surgery, The First Hospital of China Medical University, Shenyang, People's Republic of China (G.L.); Department of Biomedical Engineering, Yale University, New Haven, CT (A.K., A.W.C., M.R.B., J.D.H.); Department of Cell Biology, Washington University School of Medicine, St Louis, MO (R.P.M.); Interdepartmental Program in Vascular Biology and Therapeutics, Yale University School of Medicine (J.D.H., G.T.); and Veterans Affairs Connecticut Healthcare System, West Haven (G.T.).
| |
Collapse
|
32
|
Fang F, Lake SP. Multiscale mechanical integrity of human supraspinatus tendon in shear after elastin depletion. J Mech Behav Biomed Mater 2016; 63:443-455. [PMID: 27472764 DOI: 10.1016/j.jmbbm.2016.06.032] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2016] [Revised: 06/27/2016] [Accepted: 06/30/2016] [Indexed: 01/28/2023]
Abstract
Human supraspinatus tendon (SST) exhibits region-specific nonlinear mechanical properties under tension, which have been attributed to its complex multiaxial physiological loading environment. However, the mechanical response and underlying multiscale mechanism regulating SST behavior under other loading scenarios are poorly understood. Furthermore, little is known about the contribution of elastin to tendon mechanics. We hypothesized that (1) SST exhibits region-specific shear mechanical properties, (2) fiber sliding is the predominant mode of local matrix deformation in SST in shear, and (3) elastin helps maintain SST mechanical integrity by facilitating force transfer among collagen fibers. Through the use of biomechanical testing and multiphoton microscopy, we measured the multiscale mechanical behavior of human SST in shear before and after elastase treatment. Three distinct SST regions showed similar stresses and microscale deformation. Collagen fiber reorganization and sliding were physical mechanisms observed as the SST response to shear loading. Measures of microscale deformation were highly variable, likely due to a high degree of extracellular matrix heterogeneity. After elastase treatment, tendon exhibited significantly decreased stresses under shear loading, particularly at low strains. These results show that elastin contributes to tendon mechanics in shear, further complementing our understanding of multiscale tendon structure-function relationships.
Collapse
Affiliation(s)
- Fei Fang
- Department of Mechanical Engineering & Materials Science, Washington University in St. Louis, United States
| | - Spencer P Lake
- Department of Mechanical Engineering & Materials Science, Washington University in St. Louis, United States; Department of Biomedical Engineering, Washington University in St. Louis, United States; Department of Orthopaedic Surgery, Washington University in St. Louis, United States.
| |
Collapse
|
33
|
Duca L, Blaise S, Romier B, Laffargue M, Gayral S, El Btaouri H, Kawecki C, Guillot A, Martiny L, Debelle L, Maurice P. Matrix ageing and vascular impacts: focus on elastin fragmentation. Cardiovasc Res 2016; 110:298-308. [DOI: 10.1093/cvr/cvw061] [Citation(s) in RCA: 140] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/27/2015] [Accepted: 02/07/2016] [Indexed: 12/17/2022] Open
|
34
|
Yeo GC, Tarakanova A, Baldock C, Wise SG, Buehler MJ, Weiss AS. Subtle balance of tropoelastin molecular shape and flexibility regulates dynamics and hierarchical assembly. SCIENCE ADVANCES 2016; 2:e1501145. [PMID: 26998516 PMCID: PMC4795673 DOI: 10.1126/sciadv.1501145] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/23/2015] [Accepted: 11/20/2015] [Indexed: 05/02/2023]
Abstract
The assembly of the tropoelastin monomer into elastin is vital for conferring elasticity on blood vessels, skin, and lungs. Tropoelastin has dual needs for flexibility and structure in self-assembly. We explore the structure-dynamics-function interplay, consider the duality of molecular order and disorder, and identify equally significant functional contributions by local and global structures. To study these organizational stratifications, we perturb a key hinge region by expressing an exon that is universally spliced out in human tropoelastins. We find a herniated nanostructure with a displaced C terminus and explain by molecular modeling that flexible helices are replaced with substantial β sheets. We see atypical higher-order cross-linking and inefficient assembly into discontinuous, thick elastic fibers. We explain this dysfunction by correlating local and global structural effects with changes in the molecule's assembly dynamics. This work has general implications for our understanding of elastomeric proteins, which balance disordered regions with defined structural modules at multiple scales for functional assembly.
Collapse
Affiliation(s)
- Giselle C. Yeo
- Charles Perkins Centre, The University of Sydney, Camperdown, New South Wales 2006, Australia
- School of Molecular Bioscience, The University of Sydney, Sydney, New South Wales 2006, Australia
| | - Anna Tarakanova
- Laboratory for Atomistic and Molecular Mechanics, Department of Civil and Environmental Engineering, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA 02139, USA
| | - Clair Baldock
- Wellcome Trust Centre for Cell-Matrix Research, Faculty of Life Sciences, University of Manchester, Manchester M13 9PT, UK
| | - Steven G. Wise
- The Heart Research Institute, Newtown, New South Wales 2050, Australia
| | - Markus J. Buehler
- Laboratory for Atomistic and Molecular Mechanics, Department of Civil and Environmental Engineering, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA 02139, USA
| | - Anthony S. Weiss
- Charles Perkins Centre, The University of Sydney, Camperdown, New South Wales 2006, Australia
- School of Molecular Bioscience, The University of Sydney, Sydney, New South Wales 2006, Australia
- Bosch Institute, The University of Sydney, Sydney, New South Wales 2006, Australia
| |
Collapse
|
35
|
Weiss AS. Perspectives on the Molecular and Biological Implications of Tropoelastin in Human Tissue Elasticity. Aust J Chem 2016. [DOI: 10.1071/ch16452] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
The elasticity of a range of vertebrate and particularly human tissues depends on the dynamic and persistent protein elastin. This elasticity is diverse, and comprises skin, blood vessels, and lung, and is essential for tissue viability. Elastin is predominantly made by assembling tropoelastin, which is an asymmetric 20-nm-long protein molecule. This overview considers tropoelastin’s molecular features and biological interactions in the context of its value in tissue repair.
Collapse
|
36
|
Cuberos H, Vallée B, Vourc'h P, Tastet J, Andres CR, Bénédetti H. Roles of LIM kinases in central nervous system function and dysfunction. FEBS Lett 2015; 589:3795-806. [PMID: 26545494 DOI: 10.1016/j.febslet.2015.10.032] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2015] [Revised: 10/21/2015] [Accepted: 10/28/2015] [Indexed: 12/30/2022]
Abstract
LIM kinase 1 (LIMK1) and LIM kinase 2 (LIMK2) regulate actin dynamics by phosphorylating cofilin. In this review, we outline studies that have shown an involvement of LIMKs in neuronal function and we detail some of the pathways and molecular mechanisms involving LIMKs in neurodevelopment and synaptic plasticity. We also review the involvement of LIMKs in neuronal diseases and emphasize the differences in the regulation of LIMKs expression and mode of action. We finally present the existence of a cofilin-independent pathway also involved in neuronal function. A better understanding of the differences between both LIMKs and of the precise molecular mechanisms involved in their mode of action and regulation is now required to improve our understanding of the physiopathology of the neuronal diseases associated with LIMKs.
Collapse
Affiliation(s)
- H Cuberos
- CNRS UPR 4301, CBM, Orléans, France; UMR INSERM U930, Université François-Rabelais, Tours, France
| | - B Vallée
- CNRS UPR 4301, CBM, Orléans, France
| | - P Vourc'h
- UMR INSERM U930, Université François-Rabelais, Tours, France; CHRU de Tours, Service de Biochimie et de Biologie Moléculaire, Tours, France
| | - J Tastet
- University Medical Center Utrecht, Brain Center Rudolf Magnus, Utrecht, Netherlands
| | - C R Andres
- UMR INSERM U930, Université François-Rabelais, Tours, France; CHRU de Tours, Service de Biochimie et de Biologie Moléculaire, Tours, France
| | | |
Collapse
|
37
|
Henninger HB, Valdez WR, Scott SA, Weiss JA. Elastin governs the mechanical response of medial collateral ligament under shear and transverse tensile loading. Acta Biomater 2015; 25:304-12. [PMID: 26162584 DOI: 10.1016/j.actbio.2015.07.011] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2014] [Revised: 06/25/2015] [Accepted: 07/06/2015] [Indexed: 10/23/2022]
Abstract
Elastin is a highly extensible structural protein network that provides near-elastic resistance to deformation in biological tissues. In ligament, elastin is localized between and along the collagen fibers and fascicles. When ligament is stretched along the primary collagen axis, elastin supports a relatively high percentage of load. We hypothesized that elastin may also provide significant load support under elongation transverse to the primary collagen axis and shear along the collagen axis. Quasi-static transverse tensile and shear material tests were performed to quantify the mechanical contributions of elastin during deformation of porcine medial collateral ligament. Dose response studies were conducted to determine the level of elastase enzymatic degradation required to produce a maximal change in the mechanical response. Maximal changes in peak stress occurred after 3h of treatment with 2U/ml porcine pancreatic elastase. Elastin degradation resulted in a 60-70% reduction in peak stress and a 2-3× reduction in modulus for both test protocols. These results demonstrate that elastin provides significant resistance to elongation transverse to the collagen axis and shear along the collagen axis while only constituting 4% of the tissue dry weight. The magnitudes of the elastin contribution to peak transverse and shear stress were approximately 0.03 MPa, as compared to 2 MPa for axial tensile tests, suggesting that elastin provides a highly anisotropic contribution to the mechanical response of ligament and is the dominant structural protein resisting transverse and shear deformation of the tissue.
Collapse
|
38
|
Le VP, Cheng JK, Kim J, Staiculescu MC, Ficker SW, Sheth SC, Bhayani SA, Mecham RP, Yanagisawa H, Wagenseil JE. Mechanical factors direct mouse aortic remodelling during early maturation. J R Soc Interface 2015; 12:20141350. [PMID: 25652465 DOI: 10.1098/rsif.2014.1350] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
Numerous diseases have been linked to genetic mutations that lead to reduced amounts or disorganization of arterial elastic fibres. Previous work has shown that mice with reduced amounts of elastin (Eln+/-) are able to live a normal lifespan through cardiovascular adaptations, including changes in haemodynamic stresses, arterial geometry and arterial wall mechanics. It is not known if the timeline and presence of these adaptations are consistent in other mouse models of elastic fibre disease, such as those caused by the absence of fibulin-5 expression (Fbln5-/-). Adult Fbln5-/- mice have disorganized elastic fibres, decreased arterial compliance and high blood pressure. We examined mechanical behaviour of the aorta in Fbln5-/- mice through early maturation when the elastic fibres are being assembled. We found that the physiologic circumferential stretch, stress and modulus of Fbln5-/- aorta are maintained near wild-type levels. Constitutive modelling suggests that elastin contributions to the total stress are decreased, whereas collagen contributions are increased. Understanding how collagen fibre structure and mechanics compensate for defective elastic fibres to meet the mechanical requirements of the maturing aorta may help to better understand arterial remodelling in human elastinopathies.
Collapse
Affiliation(s)
- Victoria P Le
- Department of Biomedical Engineering, Saint Louis University, St Louis, MO, USA
| | - Jeffrey K Cheng
- Department of Biomedical Engineering, Washington University, St Louis, MO, USA
| | - Jungsil Kim
- Department of Mechanical Engineering and Materials Science, Washington University, St Louis, MO, USA
| | - Marius C Staiculescu
- Department of Mechanical Engineering and Materials Science, Washington University, St Louis, MO, USA
| | - Shawn W Ficker
- Department of Biomedical Engineering, Saint Louis University, St Louis, MO, USA
| | - Saahil C Sheth
- Department of Biomedical Engineering, Saint Louis University, St Louis, MO, USA
| | - Siddharth A Bhayani
- Department of Biomedical Engineering, Saint Louis University, St Louis, MO, USA
| | - Robert P Mecham
- Department of Cell Biology and Physiology, Washington University Medical School, St Louis, MO, USA
| | - Hiromi Yanagisawa
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Jessica E Wagenseil
- Department of Mechanical Engineering and Materials Science, Washington University, St Louis, MO, USA
| |
Collapse
|
39
|
Halabi CM, Broekelmann TJ, Knutsen RH, Ye L, Mecham RP, Kozel BA. Chronic antihypertensive treatment improves pulse pressure but not large artery mechanics in a mouse model of congenital vascular stiffness. Am J Physiol Heart Circ Physiol 2015; 309:H1008-16. [PMID: 26232234 DOI: 10.1152/ajpheart.00288.2015] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/22/2015] [Accepted: 07/27/2015] [Indexed: 01/08/2023]
Abstract
Increased arterial stiffness is a common characteristic of humans with Williams-Beuren syndrome and mouse models of elastin insufficiency. Arterial stiffness is associated with multiple negative cardiovascular outcomes, including myocardial infarction, stroke, and sudden death. Therefore, identifying therapeutic interventions that improve arterial stiffness in response to changes in elastin levels is of vital importance. The goal of this study was to determine the effect of chronic pharmacologic therapy with different classes of antihypertensive medications on arterial stiffness in elastin insufficiency. Elastin-insufficient mice 4-6 wk of age and wild-type littermates were subcutaneously implanted with osmotic micropumps delivering a continuous dose of one of the following: vehicle, losartan, nicardipine, or propranolol for 8 wk. At the end of treatment period, arterial blood pressure and large artery compliance and remodeling were assessed. Our results show that losartan and nicardipine treatment lowered blood pressure and pulse pressure in elastin-insufficient mice. Elastin and collagen content of abdominal aortas as well as ascending aorta and carotid artery biomechanics were not affected by any of the drug treatments in either genotype. By reducing pulse pressure and shifting the working pressure range of an artery to a more compliant region of the pressure-diameter curve, antihypertensive medications may mitigate the consequences of arterial stiffness, an effect that is drug class independent. These data emphasize the importance of early recognition and long-term management of hypertension in Williams-Beuren syndrome and elastin insufficiency.
Collapse
Affiliation(s)
- Carmen M Halabi
- Departments of Pediatrics Washington University School of Medicine, St. Louis, Missouri; and
| | - Thomas J Broekelmann
- Cell Biology and Physiology, Washington University School of Medicine, St. Louis, Missouri
| | - Russell H Knutsen
- Cell Biology and Physiology, Washington University School of Medicine, St. Louis, Missouri
| | - Li Ye
- Departments of Pediatrics Washington University School of Medicine, St. Louis, Missouri; and
| | - Robert P Mecham
- Cell Biology and Physiology, Washington University School of Medicine, St. Louis, Missouri
| | - Beth A Kozel
- Departments of Pediatrics Washington University School of Medicine, St. Louis, Missouri; and
| |
Collapse
|
40
|
|
41
|
Lannoy M, Slove S, Louedec L, Choqueux C, Journé C, Michel JB, Jacob MP. Inhibition of ERK1/2 Phosphorylation: A New Strategy to Stimulate Elastogenesis in the Aorta. Hypertension 2014; 64:423-30. [DOI: 10.1161/hypertensionaha.114.03352] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Affiliation(s)
- Morgane Lannoy
- From INSERM, U1148, Hôpital Bichat, Paris F-75018, France (M.L., S.S., L.L., C.C., C.J., J.-B.M., M.-P.J.); Univ Paris Diderot, Sorbonne Paris Cité, Paris F-75018, France (M.L., S.S., C.C., C.J., J.-B.M.); and Fédération de Recherche en Imagerie Multimodalité, Paris F-75018, France (C.J.)
| | - Séverin Slove
- From INSERM, U1148, Hôpital Bichat, Paris F-75018, France (M.L., S.S., L.L., C.C., C.J., J.-B.M., M.-P.J.); Univ Paris Diderot, Sorbonne Paris Cité, Paris F-75018, France (M.L., S.S., C.C., C.J., J.-B.M.); and Fédération de Recherche en Imagerie Multimodalité, Paris F-75018, France (C.J.)
| | - Liliane Louedec
- From INSERM, U1148, Hôpital Bichat, Paris F-75018, France (M.L., S.S., L.L., C.C., C.J., J.-B.M., M.-P.J.); Univ Paris Diderot, Sorbonne Paris Cité, Paris F-75018, France (M.L., S.S., C.C., C.J., J.-B.M.); and Fédération de Recherche en Imagerie Multimodalité, Paris F-75018, France (C.J.)
| | - Christine Choqueux
- From INSERM, U1148, Hôpital Bichat, Paris F-75018, France (M.L., S.S., L.L., C.C., C.J., J.-B.M., M.-P.J.); Univ Paris Diderot, Sorbonne Paris Cité, Paris F-75018, France (M.L., S.S., C.C., C.J., J.-B.M.); and Fédération de Recherche en Imagerie Multimodalité, Paris F-75018, France (C.J.)
| | - Clément Journé
- From INSERM, U1148, Hôpital Bichat, Paris F-75018, France (M.L., S.S., L.L., C.C., C.J., J.-B.M., M.-P.J.); Univ Paris Diderot, Sorbonne Paris Cité, Paris F-75018, France (M.L., S.S., C.C., C.J., J.-B.M.); and Fédération de Recherche en Imagerie Multimodalité, Paris F-75018, France (C.J.)
| | - Jean-Baptiste Michel
- From INSERM, U1148, Hôpital Bichat, Paris F-75018, France (M.L., S.S., L.L., C.C., C.J., J.-B.M., M.-P.J.); Univ Paris Diderot, Sorbonne Paris Cité, Paris F-75018, France (M.L., S.S., C.C., C.J., J.-B.M.); and Fédération de Recherche en Imagerie Multimodalité, Paris F-75018, France (C.J.)
| | - Marie-Paule Jacob
- From INSERM, U1148, Hôpital Bichat, Paris F-75018, France (M.L., S.S., L.L., C.C., C.J., J.-B.M., M.-P.J.); Univ Paris Diderot, Sorbonne Paris Cité, Paris F-75018, France (M.L., S.S., C.C., C.J., J.-B.M.); and Fédération de Recherche en Imagerie Multimodalité, Paris F-75018, France (C.J.)
| |
Collapse
|
42
|
Xu J, Shi GP. Vascular wall extracellular matrix proteins and vascular diseases. Biochim Biophys Acta Mol Basis Dis 2014; 1842:2106-2119. [PMID: 25045854 DOI: 10.1016/j.bbadis.2014.07.008] [Citation(s) in RCA: 239] [Impact Index Per Article: 23.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2014] [Revised: 07/07/2014] [Accepted: 07/14/2014] [Indexed: 01/08/2023]
Abstract
Extracellular matrix proteins form the basic structure of blood vessels. Along with providing basic structural support to blood vessels, matrix proteins interact with different sets of vascular cells via cell surface integrin or non-integrin receptors. Such interactions induce vascular cell de novo synthesis of new matrix proteins during blood vessel development or remodeling. Under pathological conditions, vascular matrix proteins undergo proteolytic processing, yielding bioactive fragments to influence vascular wall matrix remodeling. Vascular cells also produce alternatively spliced variants that induce vascular cell production of different matrix proteins to interrupt matrix homeostasis, leading to increased blood vessel stiffness; vascular cell migration, proliferation, or death; or vascular wall leakage and rupture. Destruction of vascular matrix proteins leads to vascular cell or blood-borne leukocyte accumulation, proliferation, and neointima formation within the vascular wall; blood vessels prone to uncontrolled enlargement during blood flow diastole; tortuous vein development; and neovascularization from existing pathological tissue microvessels. Here we summarize discoveries related to blood vessel matrix proteins within the past decade from basic and clinical studies in humans and animals - from expression to cross-linking, assembly, and degradation under physiological and vascular pathological conditions, including atherosclerosis, aortic aneurysms, varicose veins, and hypertension.
Collapse
Affiliation(s)
- Junyan Xu
- Department of Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Guo-Ping Shi
- Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
43
|
Affiliation(s)
- R. Thomas Collins
- From the Arkansas Children’s Hospital and University of Arkansas for Medical Sciences, Little Rock, AR
| |
Collapse
|
44
|
Zhang P, Huang A, Morales-Ruiz M, Starcher BC, Huang Y, Sessa WC, Niklason LE, Giordano FJ. Engineered zinc-finger proteins can compensate genetic haploinsufficiency by transcriptional activation of the wild-type allele: application to Willams-Beuren syndrome and supravalvular aortic stenosis. Hum Gene Ther 2013; 23:1186-99. [PMID: 22891920 DOI: 10.1089/hum.2011.201] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Williams-Beuren syndrome (WBS) and supravalvular aortic stenosis (SVAS) are genetic syndromes marked by the propensity to develop severe vascular stenoses. Vascular lesions in both syndromes are caused by haploinsufficiency of the elastin gene. We used these distinct genetic syndromes as models to evaluate the feasibility of using engineered zinc-finger protein transcription factors (ZFPs) to achieve compensatory expression of haploinsufficient genes by inducing augmented expression from the remaining wild-type allele. For complex genes with multiple splice variants, this approach could have distinct advantages over cDNA-based gene replacement strategies. Targeting the elastin gene, we show that transcriptional activation by engineered ZFPs can induce compensatory expression from the wild-type allele in the setting of classic WBS and SVAS genetic mutations, increase elastin expression in wild-type cells, induce expression of the major elastin splice variants, and recapitulate their natural stoichiometry. Further, we establish that transcriptional activation of the mutant allele in SVAS does not overcome nonsense-mediated decay, and thus ZFP-mediated transcriptional activation is not likely to induce production of a mutant protein, a crucial consideration. Finally, we show in bioengineered blood vessels that ZFP-mediated induction of elastin expression is capable of stimulating functional elastogenesis. Haploinsufficiency is a common mechanism of genetic disease. These findings have significant implications for WBS and SVAS, and establish that haploinsufficiency can be overcome by targeted transcriptional activation without inducing protein expression from the mutant allele.
Collapse
Affiliation(s)
- Pei Zhang
- Vascular Biology and Therapeutics Program, Yale University School of Medicine, New Haven, CT 06520, USA
| | | | | | | | | | | | | | | |
Collapse
|
45
|
Li W, Li Q, Qin L, Ali R, Qyang Y, Tassabehji M, Pober BR, Sessa WC, Giordano FJ, Tellides G. Rapamycin inhibits smooth muscle cell proliferation and obstructive arteriopathy attributable to elastin deficiency. Arterioscler Thromb Vasc Biol 2013; 33:1028-35. [PMID: 23493289 DOI: 10.1161/atvbaha.112.300407] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
OBJECTIVE Patients with elastin deficiency attributable to gene mutation (supravalvular aortic stenosis) or chromosomal microdeletion (Williams syndrome) are characterized by obstructive arteriopathy resulting from excessive smooth muscle cell (SMC) proliferation, mural expansion, and inadequate vessel size. We investigated whether rapamycin, an inhibitor of the cell growth regulator mammalian target of rapamycin (mTOR) and effective against other SMC proliferative disorders, is of therapeutic benefit in experimental models of elastin deficiency. APPROACH AND RESULTS As previously reported, Eln(-/-) mice demonstrated SMC hyperplasia and severe stenosis of the aorta, whereas Eln(+/-) mice exhibited a smaller diameter aorta with more numerous but thinner elastic lamellae. Increased mTOR signaling was detected in elastin-deficient aortas of newborn pups that was inhibited by maternal administration of rapamycin. mTOR inhibition reduced SMC proliferation and aortic obstruction in Eln(-/-) pups and prevented medial hyperlamellation in Eln(+/-) weanlings without compromising aortic size. However, rapamycin did not prolong the survival of Eln(-/-) pups, and it retarded the somatic growth of juvenile Eln(+/-) and Eln(+/+) mice. In cell cultures, rapamycin inhibited prolonged mTOR activation and enhanced proliferation of SMC derived from patients with supravalvular aortic stenosis and with Williams syndrome. CONCLUSIONS mTOR inhibition may represent a pharmacological strategy to treat diffuse arteriopathy resulting from elastin deficiency.
Collapse
Affiliation(s)
- Wei Li
- Department of Surgery, Interdepartmental Program in Vascular Biology and Therapeutics, Yale University School of Medicine, New Haven, CT, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Kinnear C, Chang WY, Khattak S, Hinek A, Thompson T, de Carvalho Rodrigues D, Kennedy K, Mahmut N, Pasceri P, Stanford WL, Ellis J, Mital S. Modeling and rescue of the vascular phenotype of Williams-Beuren syndrome in patient induced pluripotent stem cells. Stem Cells Transl Med 2012; 2:2-15. [PMID: 23283491 DOI: 10.5966/sctm.2012-0054] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Elastin haploinsufficiency in Williams-Beuren syndrome (WBS) leads to increased vascular smooth muscle cell (SMC) proliferation and stenoses. Our objective was to generate a human induced pluripotent stem (hiPS) cell model for in vitro assessment of the WBS phenotype and to test the ability of candidate agents to rescue the phenotype. hiPS cells were reprogrammed from skin fibroblasts of a WBS patient with aortic and pulmonary stenosis and healthy control BJ fibroblasts using four-factor retrovirus reprogramming and were differentiated into SMCs. Differentiated SMCs were treated with synthetic elastin-binding protein ligand 2 (EBPL2) (20 μg/ml) or the antiproliferative drug rapamycin (100 nM) for 5 days. We generated four WBS induced pluripotent stem (iPS) cell lines that expressed pluripotency genes and differentiated into all three germ layers. Directed differentiation of BJ iPS cells yielded an 85%-92% pure SMC population that expressed differentiated SMC markers, were functionally contractile, and formed tube-like structures on three-dimensional gel assay. Unlike BJ iPS cells, WBS iPS cells generated immature SMCs that were highly proliferative, showed lower expression of differentiated SMC markers, reduced response to the vasoactive agonists, carbachol and endothelin-1, impaired vascular tube formation, and reduced calcium flux. EBPL2 partially rescued and rapamycin fully rescued the abnormal SMC phenotype by decreasing the smooth muscle proliferation rate and enhancing differentiation and tube formation. WBS iPS cell-derived SMCs demonstrate an immature proliferative phenotype with reduced functional and contractile properties, thereby recapitulating the human disease phenotype. The ability of rapamycin to rescue the phenotype provides an attractive therapeutic candidate for patients with WBS and vascular stenoses.
Collapse
Affiliation(s)
- Caroline Kinnear
- Department of Pediatrics, University of Toronto, Toronto, Ontario, Canada
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
|
48
|
Morimoto M, Yu Z, Stenzel P, Clewing JM, Najafian B, Mayfield C, Hendson G, Weinkauf JG, Gormley AK, Parham DM, Ponniah U, André JL, Asakura Y, Basiratnia M, Bogdanović R, Bokenkamp A, Bonneau D, Buck A, Charrow J, Cochat P, Cordeiro I, Deschenes G, Fenkçi MS, Frange P, Fründ S, Fryssira H, Guillen-Navarro E, Keller K, Kirmani S, Kobelka C, Lamfers P, Levtchenko E, Lewis DB, Massella L, McLeod DR, Milford DV, Nobili F, Saraiva JM, Semerci CN, Shoemaker L, Stajić N, Stein A, Taha D, Wand D, Zonana J, Lücke T, Boerkoel CF. Reduced elastogenesis: a clue to the arteriosclerosis and emphysematous changes in Schimke immuno-osseous dysplasia? Orphanet J Rare Dis 2012; 7:70. [PMID: 22998683 PMCID: PMC3568709 DOI: 10.1186/1750-1172-7-70] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2012] [Accepted: 09/14/2012] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Arteriosclerosis and emphysema develop in individuals with Schimke immuno-osseous dysplasia (SIOD), a multisystem disorder caused by biallelic mutations in SMARCAL1 (SWI/SNF-related, matrix-associated, actin-dependent regulator of chromatin, subfamily a-like 1). However, the mechanism by which the vascular and pulmonary disease arises in SIOD remains unknown. METHODS We reviewed the records of 65 patients with SMARCAL1 mutations. Molecular and immunohistochemical analyses were conducted on autopsy tissue from 4 SIOD patients. RESULTS Thirty-two of 63 patients had signs of arteriosclerosis and 3 of 51 had signs of emphysema. The arteriosclerosis was characterized by intimal and medial hyperplasia, smooth muscle cell hyperplasia and fragmented and disorganized elastin fibers, and the pulmonary disease was characterized by panlobular enlargement of air spaces. Consistent with a cell autonomous disorder, SMARCAL1 was expressed in arterial and lung tissue, and both the aorta and lung of SIOD patients had reduced expression of elastin and alterations in the expression of regulators of elastin gene expression. CONCLUSIONS This first comprehensive study of the vascular and pulmonary complications of SIOD shows that these commonly cause morbidity and mortality and might arise from impaired elastogenesis. Additionally, the effect of SMARCAL1 deficiency on elastin expression provides a model for understanding other features of SIOD.
Collapse
Affiliation(s)
- Marie Morimoto
- Provincial Medical Genetics Program, Department of Medical Genetics, Children's and Women's Health Centre of BC, 4500 Oak Street, Room C234, Vancouver, BC, V6H 3N1, Canada
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Le VP, Wagenseil JE. Echocardiographic Characterization of Postnatal Development in Mice with Reduced Arterial Elasticity. Cardiovasc Eng Technol 2012; 3:424-438. [PMID: 23646094 DOI: 10.1007/s13239-012-0108-4] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
PURPOSE Decreased expression of elastin results in smaller, less compliant arteries and high blood pressure. In mice, these differences become more significant with postnatal development. It is known that arterial size and compliance directly affect cardiac function, but the temporal changes in cardiac function have not been investigated in elastin insufficient mice. The aim of this study is to correlate changes in arterial size and compliance with cardiac function in wildtype (WT) and elastin haploinsufficient (Eln+/- ) mice from birth to adulthood. METHODS Ultrasound scans were performed at the ages of 3, 7, 14, 21, 30, 60, and 90 days on male and female WT and Eln+/- mice. 2-D ultrasound and pulse wave Doppler images were used to measure the dimensions and function of the left ventricle (LV), ascending aorta and carotid arteries. RESULTS Eln+/- arteries are smaller and less compliant at most ages, with significant differences from WT as early as 3 days old. Surprisingly, there are no correlations (R2 < 0.2) between arterial size and compliance with measures of LV hypertrophy or systolic function. There are weak correlations (0.2 < R2 < 0.5) between arterial size and compliance with measures of LV diastolic function. CONCLUSIONS Eln+/- mice have similar cardiac function to WT throughout postnatal development, demonstrating the remarkable ability of the developing cardiovascular system to adapt to mechanical and hemodynamic changes. Correlations between arterial size and compliance with diastolic function show that these measures may be useful indicators of early diastolic dysfunction.
Collapse
Affiliation(s)
- Victoria P Le
- Department of Biomedical Engineering, Saint Louis University, St. Louis, MO
| | | |
Collapse
|
50
|
Ge X, Ren Y, Bartulos O, Lee MY, Yue Z, Kim KY, Li W, Amos PJ, Bozkulak EC, Iyer A, Zheng W, Zhao H, Martin KA, Kotton DN, Tellides G, Park IH, Yue L, Qyang Y. Modeling supravalvular aortic stenosis syndrome with human induced pluripotent stem cells. Circulation 2012; 126:1695-704. [PMID: 22914687 DOI: 10.1161/circulationaha.112.116996] [Citation(s) in RCA: 90] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
BACKGROUND Supravalvular aortic stenosis (SVAS) is caused by mutations in the elastin (ELN) gene and is characterized by abnormal proliferation of vascular smooth muscle cells (SMCs) that can lead to narrowing or blockage of the ascending aorta and other arterial vessels. Having patient-specific SMCs available may facilitate the study of disease mechanisms and development of novel therapeutic interventions. METHODS AND RESULTS Here, we report the development of a human induced pluripotent stem cell (iPSC) line from a patient with SVAS caused by the premature termination in exon 10 of the ELN gene resulting from an exon 9 four-nucleotide insertion. We showed that SVAS iPSC-derived SMCs (iPSC-SMCs) had significantly fewer organized networks of smooth muscle α-actin filament bundles, a hallmark of mature contractile SMCs, compared with control iPSC-SMCs. The addition of elastin recombinant protein or enhancement of small GTPase RhoA signaling was able to rescue the formation of smooth muscle α-actin filament bundles in SVAS iPSC-SMCs. Cell counts and BrdU analysis revealed a significantly higher proliferation rate in SVAS iPSC-SMCs than control iPSC-SMCs. Furthermore, SVAS iPSC-SMCs migrated at a markedly higher rate to the chemotactic agent platelet-derived growth factor compared with the control iPSC-SMCs. We also provided evidence that elevated activity of extracellular signal-regulated kinase 1/2 is required for hyperproliferation of SVAS iPSC-SMCs. The phenotype was confirmed in iPSC-SMCs generated from a patient with deletion of elastin owing to Williams-Beuren syndrome. CONCLUSIONS SVAS iPSC-SMCs recapitulate key pathological features of patients with SVAS and may provide a promising strategy to study disease mechanisms and to develop novel therapies.
Collapse
Affiliation(s)
- Xin Ge
- YCVRC Section of Cardiovascular Medicine, Department of Internal Medicine, Yale School of Medicine, Yale Stem Cell Center, Ste 773A, 300 George St, New Haven, CT 06510, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|