1
|
Lin C, Mazzuca MQ, Khalil RA. Increased uterine arterial tone, stiffness and remodeling with augmented matrix metalloproteinase-1 and -7 in uteroplacental ischemia-induced hypertensive pregnancy. Biochem Pharmacol 2024; 228:116227. [PMID: 38643908 PMCID: PMC11410528 DOI: 10.1016/j.bcp.2024.116227] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Revised: 03/29/2024] [Accepted: 04/18/2024] [Indexed: 04/23/2024]
Abstract
Preeclampsia is a pregnancy-related disorder manifested as hypertensive pregnancy (HTN-Preg) and often fetal growth restriction (FGR), but the mechanisms involved are unclear. We have reported enhanced reactivity of systemic vessels in HTN-Preg rats, but the critical changes in the uterine circulation are less clear. We tested whether HTN-Preg involves localized aberrations in uterine arterial tone, stiffness and remodeling by matrix metalloproteinases (MMPs). Blood pressure (BP) and litter size were recorded in normal pregnant (Preg) rats and Preg rats with reduced uteroplacental perfusion pressure (RUPP). Isolated uterine arteries were placed in a pressure myograph for measuring intrinsic and extrinsic tone and arterial stiffness. Arteries were bathed in normal Krebs solution (2.5 mM Ca2+), Ca2+-free (2 mM EGTA) Krebs, treated with sodium nitroprusside (SNP), or endothelium denuded, then pressurized at 10 mmHg steps from 10 to 110 mmHg, and the % change in diameter was analyzed to measure total (active + passive), active Ca2+-dependent myogenic, passive, and endothelium-dependent tone, respectively. BP was higher and the litter size and pup weight were reduced in RUPP vs Preg rats. In normal Krebs, increasing intraluminal pressure caused smaller increments in diameter in arteries of RUPP vs Preg rats, suggesting greater total vascular tone. Arterial incubation in Ca2+-free Krebs, treatment with SNP or endothelium-removal abolished the differences in vascular tone, and subtraction of each of these components from total vascular tone revealed significant active Ca2+-dependent myogenic, passive, and endothelium-dependent tone, respectively, in RUPP vs Preg rats. The total and passive strain-stress curves were shifted leftward in arteries of RUPP vs Preg rats, indicating increased uterine arterial stiffness. Arterial sections showed decreased lumen/total and increased wall/total area, and immunohistochemistry revealed greater MMP-1 and MMP-7 staining particularly in the media, suggesting uterine arterial remodeling by MMPs in RUPP vs Preg rats. The increased uterine arterial active myogenic, passive, and endothelium-dependent tone, arterial stiffness and remodeling by MMPs would further reduce uterine blood flow and exacerbate uteroplacental ischemia, FGR and HTN-Preg.
Collapse
Affiliation(s)
- Chen Lin
- Vascular Surgery Research Laboratories, Division of Vascular and Endovascular Surgery, Brigham and Women's Hospital, and Harvard Medical School, Boston, MA, United States
| | - Marc Q Mazzuca
- Vascular Surgery Research Laboratories, Division of Vascular and Endovascular Surgery, Brigham and Women's Hospital, and Harvard Medical School, Boston, MA, United States
| | - Raouf A Khalil
- Vascular Surgery Research Laboratories, Division of Vascular and Endovascular Surgery, Brigham and Women's Hospital, and Harvard Medical School, Boston, MA, United States.
| |
Collapse
|
2
|
Gao J, Martin L, Loffet EA, Durel JF, Oikonomou P, Nerurkar NL. MATERIAL PROPERTIES OF THE EMBRYONIC SMALL INTESTINE DURING BUCKLING MORPHOGENESIS. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.07.606927. [PMID: 39149332 PMCID: PMC11326276 DOI: 10.1101/2024.08.07.606927] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 08/17/2024]
Abstract
During embryonic development, tissues undergo dramatic deformations as functional morphologies are stereotypically sculpted from simple rudiments. Formation of healthy, functional organs therefore requires tight control over the material properties of embryonic tissues during development, yet the biological basis of embryonic tissue mechanics is poorly understood. The present study investigates the mechanics of the embryonic small intestine, a tissue that is compactly organized in the body cavity by a mechanical instability during development, wherein differential elongation rates between the intestinal tube and its attached mesentery create compressive forces that buckle the tube into loops with wavelength and curvature that are tightly conserved for a given species. Focusing on the intestinal tube, we combined micromechanical testing with histologic analyses and enzymatic degradation experiments to conclude that elastic fibers closely associated with intestinal smooth muscle layers are responsible for the bending stiffness of the tube, and for establishing its pronounced mechanical anisotropy. These findings provide insights into the developmental role of elastic fibers in controlling tissue stiffness, and raise new questions on the physiologic function of elastic fibers in the intestine during adulthood.
Collapse
Affiliation(s)
- Jenny Gao
- Department of Biomedical Engineering, Columbia University, New York NY 10027
| | - Lucia Martin
- Department of Biomedical Engineering, Columbia University, New York NY 10027
| | - Elise A. Loffet
- Department of Biomedical Engineering, Columbia University, New York NY 10027
| | - John F. Durel
- Department of Biomedical Engineering, Columbia University, New York NY 10027
| | | | - Nandan L. Nerurkar
- Department of Biomedical Engineering, Columbia University, New York NY 10027
| |
Collapse
|
3
|
Ma Y, Ma Z, Zhang Y, Luo C, Huang P, Tong J, Ding H, Liu H. Apigenin and baicalein ameliorate thoracic aortic structural deterioration and cognitive deficit via inhibiting AGEs/RAGE/NF-κB pathway in D-galactose-induced aging rats. Eur J Pharmacol 2024; 976:176660. [PMID: 38795756 DOI: 10.1016/j.ejphar.2024.176660] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Revised: 05/04/2024] [Accepted: 05/16/2024] [Indexed: 05/28/2024]
Abstract
Apigenin and baicalein are structurally related flavonoids that have been reported to have multiple pharmacological activities. The aim of this study was to investigate the protective effects and potential mechanisms of apigenin and baicalein in D-galactose-induced aging rats. First, apigenin and baicalein showed remarkable antioxidant activity and anti-glycation activity in vitro. Secondly, the protective effects of apigenin and baicalein on aging rats were investigated. We found that apigenin and baicalein supplementation significantly ameliorated aging-related changes such as declines in the spatial learning and memory and histopathological damage of the hippocampus and thoracic aorta. In addition, our data showed that apigenin and baicalein alleviated oxidative stress as illustrated by decreasing MDA level, increasing SOD activity and GSH level. Further data showed that they significantly reduced the accumulation of advanced glycation end products (AGEs), inhibited the expression of RAGE, down-regulated phosphorylated nuclear factor (p-NF-κB (p65)). Our results suggested that the protective effects of apigenin and baicalein on aging rats were at least partially related to the inhibition of AGEs/RAGE/NF-κB pathway and the improvement of oxidative damage. Overall, apigenin and baicalein showed almost equal anti-aging efficacy. Our results provided an experimental basis for the application of apigenin and baicalein to delay the aging process.
Collapse
Affiliation(s)
- Yufang Ma
- Key Laboratory of Combinatorial Biosynthesis and Drug Discovery, Wuhan University, Wuhan, Hubei, 430072, China
| | - Zhenming Ma
- College of Software Engineering, Chengdu University of Information Technology, Chengdu, Sichuan, 610200, China
| | - Yiyuan Zhang
- Key Laboratory of Combinatorial Biosynthesis and Drug Discovery, Wuhan University, Wuhan, Hubei, 430072, China
| | - Chunyun Luo
- Key Laboratory of Combinatorial Biosynthesis and Drug Discovery, Wuhan University, Wuhan, Hubei, 430072, China
| | - Puxin Huang
- Key Laboratory of Combinatorial Biosynthesis and Drug Discovery, Wuhan University, Wuhan, Hubei, 430072, China
| | - Jing Tong
- Key Laboratory of Combinatorial Biosynthesis and Drug Discovery, Wuhan University, Wuhan, Hubei, 430072, China.
| | - Hong Ding
- Key Laboratory of Combinatorial Biosynthesis and Drug Discovery, Wuhan University, Wuhan, Hubei, 430072, China.
| | - Honghui Liu
- Key Laboratory of Combinatorial Biosynthesis and Drug Discovery, Wuhan University, Wuhan, Hubei, 430072, China.
| |
Collapse
|
4
|
Wang X, Zou Z, Li K, Ren C, Yu X, Zhang Y, Zhao P, Yan S, Li Q. Design and fabrication of dual-layer PCL nanofibrous scaffolds with inductive influence on vascular cell responses. Colloids Surf B Biointerfaces 2024; 240:113988. [PMID: 38810467 DOI: 10.1016/j.colsurfb.2024.113988] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Revised: 05/03/2024] [Accepted: 05/22/2024] [Indexed: 05/31/2024]
Abstract
Confronted with the profound threat of cardiovascular diseases to health, vascular tissue engineering presents potential beyond the limitations of autologous and allogeneic grafts, offering a promising solution. This study undertakes an initial exploration into the impact of a natural active protein, elastin, on vascular cell behavior, by incorporating with polycaprolactone to prepare fibrous tissue engineering scaffold. The results reveal that elastin serves to foster endothelial cell adhesion and proliferation, suppress smooth muscle cell proliferation, and induce macrophage polarization. Furthermore, the incorporation of elastin contributes to heightened scaffold strength, compliance, and elongation, concomitantly lowering the elastic modulus. Subsequently, a bilayer oriented polycaprolactone (PCL) scaffold infused with elastin is proposed. This design draws inspiration from the cellular arrangement of native blood vessels, leveraging oriented fibers to guide cell orientation. The resulting fiber scaffold exhibits commendable mechanical properties and cell infiltration capacity, imparting valuable insights for the rapid endothelialization of vascular scaffolds.
Collapse
Affiliation(s)
- Xiaofeng Wang
- School of Mechanics and Safety Engineering, Zhengzhou University, Zhengzhou 450001, China; The State Key Laboratory of Fluid Power and Mechatronic Systems, Zhejiang University, Hangzhou 310027, China; National Center for International Research of Micro-Nano Molding Technology, Zhengzhou University, Zhengzhou 450001, China
| | - Zifan Zou
- School of Mechanics and Safety Engineering, Zhengzhou University, Zhengzhou 450001, China; National Center for International Research of Micro-Nano Molding Technology, Zhengzhou University, Zhengzhou 450001, China
| | - Kecheng Li
- School of Mechanics and Safety Engineering, Zhengzhou University, Zhengzhou 450001, China; National Center for International Research of Micro-Nano Molding Technology, Zhengzhou University, Zhengzhou 450001, China
| | - Cuihong Ren
- School of Mechanics and Safety Engineering, Zhengzhou University, Zhengzhou 450001, China; National Center for International Research of Micro-Nano Molding Technology, Zhengzhou University, Zhengzhou 450001, China
| | - Xiaorong Yu
- School of Mechanics and Safety Engineering, Zhengzhou University, Zhengzhou 450001, China; National Center for International Research of Micro-Nano Molding Technology, Zhengzhou University, Zhengzhou 450001, China.
| | - Yang Zhang
- National Center for International Research of Micro-Nano Molding Technology, Zhengzhou University, Zhengzhou 450001, China
| | - Peng Zhao
- The State Key Laboratory of Fluid Power and Mechatronic Systems, Zhejiang University, Hangzhou 310027, China
| | - Shujie Yan
- School of Mechanics and Safety Engineering, Zhengzhou University, Zhengzhou 450001, China; National Center for International Research of Micro-Nano Molding Technology, Zhengzhou University, Zhengzhou 450001, China.
| | - Qian Li
- School of Mechanics and Safety Engineering, Zhengzhou University, Zhengzhou 450001, China; National Center for International Research of Micro-Nano Molding Technology, Zhengzhou University, Zhengzhou 450001, China
| |
Collapse
|
5
|
Bywaters BC, Trache A, Rivera GM. Modulation of arterial intima stiffness by disturbed blood flow. Exp Biol Med (Maywood) 2024; 249:10090. [PMID: 39143955 PMCID: PMC11323813 DOI: 10.3389/ebm.2024.10090] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Accepted: 07/09/2024] [Indexed: 08/16/2024] Open
Abstract
The intima, comprising the endothelium and the subendothelial matrix, plays a crucial role in atherosclerosis pathogenesis. The mechanical stress arising from disturbed blood flow (d-flow) and the stiffening of the arterial wall contributes to endothelial dysfunction. However, the specific impacts of these physical forces on the mechanical environment of the intima remain undetermined. Here, we investigated whether inhibiting collagen crosslinking could ameliorate the detrimental effects of persistent d-flow on the mechanical properties of the intima. Partial ligation of the left carotid artery (LCA) was performed in C57BL/6J mice, inducing d-flow. The right carotid artery (RCA) served as an internal control. Carotids were collected 2 days and 2 weeks after surgery to study acute and chronic effects of d-flow on the mechanical phenotype of the intima. The chronic effects of d-flow were decoupled from the ensuing arterial wall stiffening by administration of β-aminopropionitrile (BAPN), an inhibitor of collagen crosslinking by lysyl oxidase (LOX) enzymes. Atomic force microscopy (AFM) was used to determine stiffness of the endothelium and the denuded subendothelial matrix in en face carotid preparations. The stiffness of human aortic endothelial cells (HAEC) cultured on soft and stiff hydrogels was also determined. Acute exposure to d-flow caused a slight decrease in endothelial stiffness in male mice but had no effect on the stiffness of the subendothelial matrix in either sex. Regardless of sex, the intact endothelium was softer than the subendothelial matrix. In contrast, exposure to chronic d-flow led to a substantial increase in the endothelial and subendothelial stiffness in both sexes. The effects of chronic d-flow were largely prevented by concurrent BAPN administration. In addition, HAEC displayed reduced stiffness when cultured on soft vs. stiff hydrogels. We conclude that chronic d-flow results in marked stiffening of the arterial intima, which can be effectively prevented by inhibition of collagen crosslinking.
Collapse
Affiliation(s)
- Briana C. Bywaters
- Department of Veterinary Pathobiology, Texas A&M University, College Station, TX, United States
| | - Andreea Trache
- Department of Medical Physiology, Texas A&M Health Science Center, Bryan, TX, United States
- Department of Biomedical Engineering, Texas A&M University, College Station, TX, United States
| | - Gonzalo M. Rivera
- Department of Veterinary Pathobiology, Texas A&M University, College Station, TX, United States
| |
Collapse
|
6
|
DeFreitas MJ, Shelton EL, Schmidt AF, Ballengee S, Tian R, Chen P, Sharma M, Levine A, Katz ED, Rojas C, Abitbol CL, Hunter J, Kulandavelu S, Wu S, Young KC, Benny M. Neonatal hyperoxia exposure leads to developmental programming of cardiovascular and renal disease in adult rats. Sci Rep 2024; 14:16742. [PMID: 39033222 PMCID: PMC11271593 DOI: 10.1038/s41598-024-65844-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Accepted: 06/25/2024] [Indexed: 07/23/2024] Open
Abstract
Premature infants are often exposed to hyperoxia. However, there is limited data regarding the mechanistic underpinnings linking neonatal hyperoxia exposure and its contribution to cardio-renal dysfunction in adults born preterm. Our objective was to determine whether neonatal hyperoxia induces systemic vascular stiffness and cardio-renal dysfunction in adulthood. Newborn rats were randomly assigned to room air (RA) or hyperoxia (85% O2) from postnatal day 1 to 14, then recovered in RA until 1 year of life. Arterial stiffness, cardio-renal histomorphometry, and fibrosis in the aorta, heart, and kidney were assessed. RNA-sequencing (RNA-seq) of the aorta and kidney was also done. Adult rats exposed to neonatal hyperoxia had increased aortic and mesenteric artery stiffness as demonstrated by wire and pressure myography. They also had cardiomyocyte hypertrophy, glomerulomegaly, and tubular injury. Hyperoxia exposure altered the transcriptome profile associated with fibrosis and matrix remodeling in the aorta and kidney. There was also increased TGF-β1 levels and fibrosis in the aorta, left ventricle, and kidney. In conclusion, neonatal hyperoxia exposure was associated with systemic vascular and cardio-renal alterations in 1-year-old rats. Further studies to determine how targeted therapies could reprogram cardio-renal injury after neonatal hyperoxia exposure are indicated.
Collapse
Affiliation(s)
- Marissa J DeFreitas
- Department of Pediatrics/Division of Nephrology, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Elaine L Shelton
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Augusto F Schmidt
- Department of Pediatrics/Division of Neonatology, Batchelor Children's Research Institute, Miller School of Medicine, University of Miami, P.O. Box 016960 (R-131), Miami, FL, 33101, USA
| | - Sydne Ballengee
- Department of Pediatrics/Division of Neonatology, Batchelor Children's Research Institute, Miller School of Medicine, University of Miami, P.O. Box 016960 (R-131), Miami, FL, 33101, USA
| | - Runxia Tian
- Department of Pediatrics/Division of Neonatology, Batchelor Children's Research Institute, Miller School of Medicine, University of Miami, P.O. Box 016960 (R-131), Miami, FL, 33101, USA
| | - PingPing Chen
- Department of Pediatrics/Division of Neonatology, Batchelor Children's Research Institute, Miller School of Medicine, University of Miami, P.O. Box 016960 (R-131), Miami, FL, 33101, USA
| | - Mayank Sharma
- Department of Pediatrics/Division of Neonatology, Batchelor Children's Research Institute, Miller School of Medicine, University of Miami, P.O. Box 016960 (R-131), Miami, FL, 33101, USA
| | - Amanda Levine
- Department of Pediatrics/Division of Neonatology, Batchelor Children's Research Institute, Miller School of Medicine, University of Miami, P.O. Box 016960 (R-131), Miami, FL, 33101, USA
| | - Emily Davidovic Katz
- Department of Pediatrics/Division of Neonatology, Batchelor Children's Research Institute, Miller School of Medicine, University of Miami, P.O. Box 016960 (R-131), Miami, FL, 33101, USA
| | - Claudia Rojas
- Department of Pathology, Memorial Healthcare Systems, Hollywood, FL, USA
| | - Carolyn L Abitbol
- Department of Pediatrics/Division of Nephrology, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Juanita Hunter
- Department of Pediatrics/Division of Cardiology, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Shathiyah Kulandavelu
- Department of Pediatrics/Division of Nephrology, University of Miami Miller School of Medicine, Miami, FL, USA
- Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Shu Wu
- Department of Pediatrics/Division of Neonatology, Batchelor Children's Research Institute, Miller School of Medicine, University of Miami, P.O. Box 016960 (R-131), Miami, FL, 33101, USA
| | - Karen C Young
- Department of Pediatrics/Division of Neonatology, Batchelor Children's Research Institute, Miller School of Medicine, University of Miami, P.O. Box 016960 (R-131), Miami, FL, 33101, USA
| | - Merline Benny
- Department of Pediatrics/Division of Neonatology, Batchelor Children's Research Institute, Miller School of Medicine, University of Miami, P.O. Box 016960 (R-131), Miami, FL, 33101, USA.
| |
Collapse
|
7
|
Olea E, Valverde-Pérez E, Docio I, Prieto-Lloret J, Aaronson PI, Rocher A. Pulmonary Vascular Responses to Chronic Intermittent Hypoxia in a Guinea Pig Model of Obstructive Sleep Apnea. Int J Mol Sci 2024; 25:7484. [PMID: 39000591 PMCID: PMC11242077 DOI: 10.3390/ijms25137484] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Revised: 07/02/2024] [Accepted: 07/05/2024] [Indexed: 07/16/2024] Open
Abstract
Experimental evidence suggests that chronic intermittent hypoxia (CIH), a major hallmark of obstructive sleep apnea (OSA), boosts carotid body (CB) responsiveness, thereby causing increased sympathetic activity, arterial and pulmonary hypertension, and cardiovascular disease. An enhanced circulatory chemoreflex, oxidative stress, and NO signaling appear to play important roles in these responses to CIH in rodents. Since the guinea pig has a hypofunctional CB (i.e., it is a natural CB knockout), in this study we used it as a model to investigate the CB dependence of the effects of CIH on pulmonary vascular responses, including those mediated by NO, by comparing them with those previously described in the rat. We have analyzed pulmonary artery pressure (PAP), the hypoxic pulmonary vasoconstriction (HPV) response, endothelial function both in vivo and in vitro, and vascular remodeling (intima-media thickness, collagen fiber content, and vessel lumen area). We demonstrate that 30 days of the exposure of guinea pigs to CIH (FiO2, 5% for 40 s, 30 cycles/h) induces pulmonary artery remodeling but does not alter endothelial function or the contractile response to phenylephrine (PE) in these arteries. In contrast, CIH exposure increased the systemic arterial pressure and enhanced the contractile response to PE while decreasing endothelium-dependent vasorelaxation to carbachol in the aorta without causing its remodeling. We conclude that since all of these effects are independent of CB sensitization, there must be other oxygen sensors, beyond the CB, with the capacity to alter the autonomic control of the heart and vascular function and structure in CIH.
Collapse
Affiliation(s)
- Elena Olea
- Departamento de Enfermería, Facultad de Enfermería Universidad de Valladolid, 47005 Valladolid, Spain
- Unidad de Excelencia Instituto de Biomedicina y Genética Molecular (IBGM), Universidad de Valladolid-CSIC, 47005 Valladolid, Spain
| | - Esther Valverde-Pérez
- Unidad de Excelencia Instituto de Biomedicina y Genética Molecular (IBGM), Universidad de Valladolid-CSIC, 47005 Valladolid, Spain
- Departamento de Bioquímica y Biología Molecular y Fisiología, Facultad de Medicina, Universidad de Valladolid, 47005 Valladolid, Spain
| | - Inmaculada Docio
- Departamento de Bioquímica y Biología Molecular y Fisiología, Facultad de Medicina, Universidad de Valladolid, 47005 Valladolid, Spain
| | - Jesus Prieto-Lloret
- Unidad de Excelencia Instituto de Biomedicina y Genética Molecular (IBGM), Universidad de Valladolid-CSIC, 47005 Valladolid, Spain
- Departamento de Bioquímica y Biología Molecular y Fisiología, Facultad de Medicina, Universidad de Valladolid, 47005 Valladolid, Spain
| | - Philip I Aaronson
- Department of Inflammation Biology, School of Immunology and Microbial Sciences, Faculty of Life Sciences and Medicine, King's College London, London SE1 9RT, UK
| | - Asunción Rocher
- Unidad de Excelencia Instituto de Biomedicina y Genética Molecular (IBGM), Universidad de Valladolid-CSIC, 47005 Valladolid, Spain
- Departamento de Bioquímica y Biología Molecular y Fisiología, Facultad de Medicina, Universidad de Valladolid, 47005 Valladolid, Spain
| |
Collapse
|
8
|
Chandra Sekar N, Khoshmanesh K, Baratchi S. Bioengineered models of cardiovascular diseases. Atherosclerosis 2024; 393:117565. [PMID: 38714426 DOI: 10.1016/j.atherosclerosis.2024.117565] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/25/2023] [Revised: 04/15/2024] [Accepted: 04/25/2024] [Indexed: 05/09/2024]
Abstract
Age-associated cardiovascular diseases (CVDs), predominantly resulting from artery-related disorders such as atherosclerosis, stand as a leading cause of morbidity and mortality among the elderly population. Consequently, there is a growing interest in the development of clinically relevant bioengineered models of CVDs. Recent developments in bioengineering and material sciences have paved the way for the creation of intricate models that closely mimic the structure and surroundings of native cardiac tissues and blood vessels. These models can be utilized for basic research purposes and for identifying pharmaceutical interventions and facilitating drug discovery. The advancement of vessel-on-a-chip technologies and the development of bioengineered and humanized in vitro models of the cardiovascular system have the potential to revolutionize CVD disease modelling. These technologies offer pathophysiologically relevant models at a fraction of the cost and time required for traditional experimentation required in vivo. This progress signifies a significant advancement in the field, transitioning from conventional 2D cell culture models to advanced 3D organoid and vessel-on-a-chip models. These innovative models are specifically designed to explore the complexities of vascular aging and stiffening, crucial factors in the development of cardiovascular diseases. This review summarizes the recent progress of various bioengineered in vitro platforms developed for investigating the pathophysiology of human cardiovascular system with more focus on advanced 3D vascular platforms.
Collapse
Affiliation(s)
- Nadia Chandra Sekar
- School of Health & Biomedical Sciences, RMIT University, Bundoora, Victoria, 3082, Australia; Baker Heart and Diabetes Institute, Melbourne, Victoria, 3004, Australia
| | - Khashayar Khoshmanesh
- Baker Heart and Diabetes Institute, Melbourne, Victoria, 3004, Australia; School of Engineering, RMIT University, Melbourne, Victoria, 3000, Australia
| | - Sara Baratchi
- School of Health & Biomedical Sciences, RMIT University, Bundoora, Victoria, 3082, Australia; Baker Heart and Diabetes Institute, Melbourne, Victoria, 3004, Australia; Department of Cardiometabolic Health, The University of Melbourne, Parkville, Victoria, 3010, Australia.
| |
Collapse
|
9
|
Dong H, Leach JR, Kao E, Zhou A, Chitiboi T, Zhu C, Ballweber M, Jiang F, Lee YJ, Iannuzzi J, Gasper W, Saloner D, Hope MD, Mitsouras D. Measurement of Abdominal Aortic Aneurysm Strain Using MR Deformable Image Registration: Accuracy and Relationship to Recent Aneurysm Progression. Invest Radiol 2024; 59:425-432. [PMID: 37855728 PMCID: PMC11026303 DOI: 10.1097/rli.0000000000001035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2023]
Abstract
BACKGROUND Management of asymptomatic abdominal aortic aneurysm (AAA) based on maximum aneurysm diameter and growth rate fails to preempt many ruptures. Assessment of aortic wall biomechanical properties may improve assessment of progression and rupture risk. This study aimed to assess the accuracy of AAA wall strain measured by cine magnetic resonance imaging (MRI) deformable image registration (MR strain) and investigate its relationship with recent AAA progression. METHODS The MR strain accuracy was evaluated in silico against ground truth strain in 54 synthetic MRIs generated from a finite element model simulation of an AAA patient's abdomen for different aortic pulse pressures, tissue motions, signal intensity variations, and image noise. Evaluation included bias with 95% confidence interval (CI) and correlation analysis. Association of MR strain with AAA growth rate was assessed in 25 consecutive patients with >6 months of prior surveillance, for whom cine balanced steady-state free-precession imaging was acquired at the level of the AAA as well as the proximal, normal-caliber aorta. Univariate and multivariate regressions were used to associate growth rate with clinical variables, maximum AAA diameter (D max ), and peak circumferential MR strain through the cardiac cycle. The MR strain interoperator variability was assessed using bias with 95% CI, intraclass correlation coefficient, and coefficient of variation. RESULTS In silico experiments revealed an MR strain bias of 0.48% ± 0.42% and a slope of correlation to ground truth strain of 0.963. In vivo, AAA MR strain (1.2% ± 0.6%) was highly reproducible (bias ± 95% CI, 0.03% ± 0.31%; intraclass correlation coefficient, 97.8%; coefficient of variation, 7.14%) and was lower than in the nonaneurysmal aorta (2.4% ± 1.7%). D max ( β = 0.087) and MR strain ( β = -1.563) were both associated with AAA growth rate. The MR strain remained an independent factor associated with growth rate ( β = -0.904) after controlling for D max . CONCLUSIONS Deformable image registration analysis can accurately measure the circumferential strain of the AAA wall from standard cine MRI and may offer patient-specific insight regarding AAA progression.
Collapse
Affiliation(s)
- Huiming Dong
- From the Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, CA (H.D., J.L., E.K., A.Z., C.Z., M.B., Y.J.L., D.S., M.H., D.M.); Vascular Imaging Research Center, San Francisco Veteran Affairs Medical Center, San Francisco, CA (H.D., J.L., E.K., A.Z., C.Z., M.B., D.S., M.H., D.M.); Siemens Healthineers (T.C.); Department of Radiology, University of Washington, Seattle, WA (C.Z.); Department of Epidemiology and Biostatistics, University of California, San Francisco, San Francisco, CA (F.J.); Department of Surgery, University of California, San Francisco, San Francisco, CA (J.I., W. G.); and Department of Vascular Surgery, San Francisco Veteran Affairs Medical Center, San Francisco, CA (J.I., W.G.)
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
10
|
Wu J, Steward RL. Disturbed fluid flow reinforces endothelial tractions and intercellular stresses. J Biomech 2024; 169:112156. [PMID: 38761747 DOI: 10.1016/j.jbiomech.2024.112156] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Revised: 04/25/2024] [Accepted: 05/14/2024] [Indexed: 05/20/2024]
Abstract
Disturbed fluid flow is well understood to have significant ramifications on endothelial function, but the impact disturbed flow has on endothelial biomechanics is not well understood. In this study, we measured tractions, intercellular stresses, and cell velocity of endothelial cells exposed to disturbed flow using a custom-fabricated flow chamber. Our flow chamber exposed cells to disturbed fluid flow within the following spatial zones: zone 1 (inlet; length 0.676-2.027 cm): 0.0037 ± 0.0001 Pa; zone 2 (middle; length 2.027-3.716 cm): 0.0059 ± 0.0005 Pa; and zone 3 (outlet; length 3.716-5.405 cm): 0.0051 ± 0.0025 Pa. Tractions and intercellular stresses were observed to be highest in the middle of the chamber (zone 2) and lowest at the chamber outlet (zone 3), while cell velocity was highest near the chamber inlet (zone 1), and lowest near the middle of the chamber (zone 2). Our findings suggest endothelial biomechanical response to disturbed fluid flow to be dependent on not only shear stress magnitude, but the spatial shear stress gradient as well. We believe our results will be useful to a host of fields including endothelial cell biology, the cardiovascular field, and cellular biomechanics in general.
Collapse
Affiliation(s)
- Jingwen Wu
- Department of Mechanical and Aerospace Engineering, University of Central Florida, Orlando, FL, United States
| | - R L Steward
- Department of Mechanical and Aerospace Engineering, University of Central Florida, Orlando, FL, United States.
| |
Collapse
|
11
|
Lee HJ, Choi JW. Association between waist circumference change after smoking cessation and incidence of hypertension in Korean adults. Public Health 2024; 229:73-79. [PMID: 38402666 DOI: 10.1016/j.puhe.2024.01.028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Revised: 12/14/2023] [Accepted: 01/26/2024] [Indexed: 02/27/2024]
Abstract
OBJECTIVES This study investigates the association between smoking cessation and hypertension incidence, as well as the association between waist circumference change after smoking cessation and hypertension incidence. STUDY DESIGN This was a nationwide population-based cohort study. METHODS We used the Korean Health Screening Cohort data and included 158,505 participants who had undergone two or more health examinations between 2008 and 2011, with follow-ups throughout 2019. Smoking cessation and waist changes were captured based on difference between first and follow-up screening dates. Hazard ratio (HR) and 95% confidence interval (CI) for hypertension risk were estimated using multivariable Cox proportional hazard regression models. RESULTS There were 31,270 cases of hypertension during a median follow-up of 8.50 years. After adjusting for potential confounding factors, HR for hypertension were 1.01 (95% CI: 0.97-1.05), 0.91 (95% CI: 0.87-0.95), and 0.88 (95% CI: 0.85-0.91) for recent quitters, long-term quitters, and non-smokers, respectively, compared with current smokers. HR for hypertension, compared with current smokers, were 0.89 (95% CI: 0.84-0.94), 0.91 (95% CI: 0.85-0.97), and 0.99 (95% CI: 0.91-1.08) for long-term quitters with no waist gain, long-term quitters with waist gain of 0.1-5.0 cm, and long-term quitters with waist gain of ≥5.0 cm, respectively. CONCLUSIONS Long-term smoking cessation was significantly associated with decreased risk of hypertension, and long-term smoking cessation with no waist gain or less than 5.0 cm of waist gain was significantly associated with decreased risk of hypertension. However, more than 5.0 cm of waist gain can attenuate the effect of long-term smoking cessation on lowering the risk of hypertension.
Collapse
Affiliation(s)
- H J Lee
- Department of Statistics and Data Science, Yonsei University, Seoul, Republic of Korea
| | - J W Choi
- Health Insurance Research Institute, National Health Insurance Service, Wonju, Republic of Korea.
| |
Collapse
|
12
|
Sapey E, Crowley LE, Edgar RG, Griffiths D, Samanta S, Crisford H, Bolton CE, Hurst JR, Stockley RA. Cardiovascular disease in Alpha 1 antitrypsin deficiency: an observational study assessing the role of neutrophil proteinase activity and the suitability of validated screening tools. Orphanet J Rare Dis 2024; 19:130. [PMID: 38515138 PMCID: PMC10956254 DOI: 10.1186/s13023-024-03124-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Accepted: 03/03/2024] [Indexed: 03/23/2024] Open
Abstract
BACKGROUND Alpha 1 Antitrypsin Deficiency (AATD) is a rare, inherited lung disease which shares features with Chronic Obstructive Pulmonary Disease (COPD) but has a greater burden of proteinase related tissue damage. These proteinases are associated with cardiovascular disease (CVD) in the general population. It is unclear whether patients with AATD have a greater risk of CVD compared to usual COPD, how best to screen for this, and whether neutrophil proteinases are implicated in AATD-associated CVD. This study had three aims. To compare CVD risk in never-augmented AATD patients to non-AATD COPD and healthy controls (HC). To assess relationships between CVD risk and lung physiology. To determine if neutrophil proteinase activity was associated with CVD risk in AATD. Cardiovascular risk was assessed by QRISK2® score and aortic stiffness measurements using carotid-femoral (aortic) pulse wave velocity (aPWV). Medical history, computed tomography scans and post-bronchodilator lung function parameters were reviewed. Systemic proteinase 3 activity was measured. Patients were followed for 4 years, to assess CVD development. RESULTS 228 patients with AATD, 50 with non-AATD COPD and 51 healthy controls were recruited. In all COPD and HC participants, QRISK2® and aPWV gave concordant results (with both measures either high or in the normal range). This was not the case in AATD. Once aPWV was adjusted for age and smoking history, aPWV was highest and QRISK2® lowest in AATD patients compared to the COPD or HC participants. Higher aPWV was associated with impairments in lung physiology, the presence of emphysema on CT scan and proteinase 3 activity following adjustment for age, smoking status and traditional CVD risk factors (using QRISK2® scores) in AATD. There were no such relationships with QRISK2® in AATD. AATD patients with confirmed CVD at four-year follow up had a higher aPWV but not QRISK2® at baseline assessment. CONCLUSION aPWV measured CVD risk is elevated in AATD. This risk is not captured by QRISK2®. There is a relationship between aPWV, lung disease and proteinase-3 activity. Proteinase-driven breakdown of elastin fibres in large arteries and lungs is a putative mechanism and forms a potential therapeutic target for CVD in AATD.
Collapse
Affiliation(s)
- E Sapey
- Institute of Inflammation and Ageing, University of Birmingham, Birmingham, B15 2GW, UK
- University Hospitals Birmingham NHS Foundation Trust, Edgbaston, Birmingham, West Midlands, UK
| | - L E Crowley
- Institute of Inflammation and Ageing, University of Birmingham, Birmingham, B15 2GW, UK.
- University Hospitals Birmingham NHS Foundation Trust, Edgbaston, Birmingham, West Midlands, UK.
| | - R G Edgar
- Institute of Applied Health, University of Birmingham, Birmingham, West Midlands, UK
| | - D Griffiths
- University Hospitals Birmingham NHS Foundation Trust, Edgbaston, Birmingham, West Midlands, UK
| | - S Samanta
- UCL Respiratory, University College London, London, UK
| | - H Crisford
- Institute of Inflammation and Ageing, University of Birmingham, Birmingham, B15 2GW, UK
| | - C E Bolton
- NIHR Nottingham BRC Respiratory Theme, School of Medicine, University of Nottingham, City Hospital NUH Trust, Nottingham, UK
| | - J R Hurst
- UCL Respiratory, University College London, London, UK
| | - R A Stockley
- University Hospitals Birmingham NHS Foundation Trust, Edgbaston, Birmingham, West Midlands, UK
| |
Collapse
|
13
|
Melton HJ, Zhang Z, Wu C. SUMMIT-FA: a new resource for improved transcriptome imputation using functional annotations. Hum Mol Genet 2024; 33:624-635. [PMID: 38129112 PMCID: PMC10954367 DOI: 10.1093/hmg/ddad205] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Revised: 10/24/2023] [Accepted: 11/30/2023] [Indexed: 12/23/2023] Open
Abstract
Transcriptome-wide association studies (TWAS) integrate gene expression prediction models and genome-wide association studies (GWAS) to identify gene-trait associations. The power of TWAS is determined by the sample size of GWAS and the accuracy of the expression prediction model. Here, we present a new method, the Summary-level Unified Method for Modeling Integrated Transcriptome using Functional Annotations (SUMMIT-FA), which improves gene expression prediction accuracy by leveraging functional annotation resources and a large expression quantitative trait loci (eQTL) summary-level dataset. We build gene expression prediction models in whole blood using SUMMIT-FA with the comprehensive functional database MACIE and eQTL summary-level data from the eQTLGen consortium. We apply these models to GWAS for 24 complex traits and show that SUMMIT-FA identifies significantly more gene-trait associations and improves predictive power for identifying "silver standard" genes compared to several benchmark methods. We further conduct a simulation study to demonstrate the effectiveness of SUMMIT-FA.
Collapse
Affiliation(s)
- Hunter J Melton
- Department of Statistics, Florida State University, 214 Rogers Building, 117 N. Woodward Avenue, Tallahassee, FL 32306, United States
| | - Zichen Zhang
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, 7007 Bertner Avenue, Unit 1689, Houston, TX 77030, United States
| | - Chong Wu
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, 7007 Bertner Avenue, Unit 1689, Houston, TX 77030, United States
| |
Collapse
|
14
|
Depenveiller C, Baud S, Belloy N, Bochicchio B, Dandurand J, Dauchez M, Pepe A, Pomès R, Samouillan V, Debelle L. Structural and physical basis for the elasticity of elastin. Q Rev Biophys 2024; 57:e3. [PMID: 38501287 DOI: 10.1017/s0033583524000040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/20/2024]
Abstract
Elastin function is to endow vertebrate tissues with elasticity so that they can adapt to local mechanical constraints. The hydrophobicity and insolubility of the mature elastin polymer have hampered studies of its molecular organisation and structure-elasticity relationships. Nevertheless, a growing number of studies from a broad range of disciplines have provided invaluable insights, and several structural models of elastin have been proposed. However, many questions remain regarding how the primary sequence of elastin (and the soluble precursor tropoelastin) governs the molecular structure, its organisation into a polymeric network, and the mechanical properties of the resulting material. The elasticity of elastin is known to be largely entropic in origin, a property that is understood to arise from both its disordered molecular structure and its hydrophobic character. Despite a high degree of hydrophobicity, elastin does not form compact, water-excluding domains and remains highly disordered. However, elastin contains both stable and labile secondary structure elements. Current models of elastin structure and function are drawn from data collected on tropoelastin and on elastin-like peptides (ELPs) but at the tissue level, elasticity is only achieved after polymerisation of the mature elastin. In tissues, the reticulation of tropoelastin chains in water defines the polymer elastin that bears elasticity. Similarly, ELPs require polymerisation to become elastic. There is considerable interest in elastin especially in the biomaterials and cosmetic fields where ELPs are widely used. This review aims to provide an up-to-date survey of/perspective on current knowledge about the interplay between elastin structure, solvation, and entropic elasticity.
Collapse
Affiliation(s)
- Camille Depenveiller
- UMR URCA/CNRS 7369, Matrice Extracellulaire et Dynamique Cellulaire (MEDyC), UFR Sciences Exactes et Naturelles, SFR CAP Santé, Université de Reims Champagne-Ardenne, Reims, France
- Unité de Génie Enzymatique et Cellulaire UMR 7025 CNRS, Université de Picardie Jules Verne, Amiens, France
| | - Stéphanie Baud
- UMR URCA/CNRS 7369, Matrice Extracellulaire et Dynamique Cellulaire (MEDyC), UFR Sciences Exactes et Naturelles, SFR CAP Santé, Université de Reims Champagne-Ardenne, Reims, France
| | - Nicolas Belloy
- UMR URCA/CNRS 7369, Matrice Extracellulaire et Dynamique Cellulaire (MEDyC), UFR Sciences Exactes et Naturelles, SFR CAP Santé, Université de Reims Champagne-Ardenne, Reims, France
| | - Brigida Bochicchio
- Laboratory of Bioinspired Materials, Department of Science, University of Basilicata, Potenza, Italy
| | - Jany Dandurand
- CIRIMAT UMR 5085, Université Paul Sabatier, Université de Toulouse, Toulouse, France
| | - Manuel Dauchez
- UMR URCA/CNRS 7369, Matrice Extracellulaire et Dynamique Cellulaire (MEDyC), UFR Sciences Exactes et Naturelles, SFR CAP Santé, Université de Reims Champagne-Ardenne, Reims, France
| | - Antonietta Pepe
- Laboratory of Bioinspired Materials, Department of Science, University of Basilicata, Potenza, Italy
| | - Régis Pomès
- Molecular Medicine, Hospital for Sick Children, Toronto, ON, Canada
- Department of Biochemistry, University of Toronto, Toronto, ON, Canada
| | - Valérie Samouillan
- CIRIMAT UMR 5085, Université Paul Sabatier, Université de Toulouse, Toulouse, France
| | - Laurent Debelle
- UMR URCA/CNRS 7369, Matrice Extracellulaire et Dynamique Cellulaire (MEDyC), UFR Sciences Exactes et Naturelles, SFR CAP Santé, Université de Reims Champagne-Ardenne, Reims, France
| |
Collapse
|
15
|
Ibarrola J, Xiang RR, Sun Z, Lu Q, Hill MA, Jaffe IZ. Inhibition of the histone methyltransferase EZH2 induces vascular stiffness. Clin Sci (Lond) 2024; 138:251-268. [PMID: 38362910 DOI: 10.1042/cs20231478] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2023] [Revised: 02/14/2024] [Accepted: 02/16/2024] [Indexed: 02/17/2024]
Abstract
Vascular stiffness increases with aging, obesity and hypertension and predicts cardiovascular risk. The levels of histone H3-lysine-27 methylation (H3K27me) and the histone methyltransferase EZH2 both decrease in aging vessels, driving vascular stiffness. The impact of EZH2 inhibitors on vascular stiffness is unknown. We tested the hypothesis that the EZH2 inhibitor GSK126, currently in development for cancer treatment, increases vascular stiffness and explored underlying molecular mechanisms. Young (3 month) and middle-aged (12 month) male mice were treated with GSK126 for 1-2 months and primary human aortic smooth muscle cells (HASMCs) from young male and female donors were treated with GSK126 for 24-48 h. Stiffness was measured in vivo by pulse wave velocity and in vitro by atomic force microscopy (AFM) and vascular structure was quantified histologically. Extracellular matrix proteins were studied by qRT-PCR, immunoblotting, zymography and chromatin immunoprecipitation. GSK126 treatment decreased H3K27 methylation (H3K27me) and increased acetylation (H3K27ac) in mouse vessels and in HASMCs. In GSK126-treated mice, aortic stiffness increased without changes in vascular fibrosis. EZH2 inhibition enhanced elastin fiber degradation and matrix metalloprotease-2 (MMP2) expression. In HASMCs, GSK126 treatment increased synthetic phenotype markers and intrinsic HASMCs stiffness by AFM with altered cytoskeletal structure and increased nuclear actin staining. GSK126 also increased MMP2 protein expression, activity and enrichment of H3K27ac at the MMP2 promoter in HASMCs. GSK126 causes vascular stiffening, inducing MMP2 activity, elastin degradation, and modulation of SMC phenotype and cytoskeletal stiffness. These findings suggest that EZH2 inhibitors used to treat cancer could negatively impact the vasculature by enhancing stiffness and merits examination in human trials.
Collapse
Affiliation(s)
- Jaime Ibarrola
- Molecular Cardiology Research Institute, Tufts Medical Center, Boston, MA 02111, U.S.A
| | - Rachel R Xiang
- Molecular Cardiology Research Institute, Tufts Medical Center, Boston, MA 02111, U.S.A
| | - Zhe Sun
- Department of Medical Pharmacology and Physiology, Dalton Cardiovascular Research Center, University of Missouri, Columbia, MO 65203, U.S.A
| | - Qing Lu
- Molecular Cardiology Research Institute, Tufts Medical Center, Boston, MA 02111, U.S.A
| | - Michael A Hill
- Department of Medical Pharmacology and Physiology, Dalton Cardiovascular Research Center, University of Missouri, Columbia, MO 65203, U.S.A
| | - Iris Z Jaffe
- Molecular Cardiology Research Institute, Tufts Medical Center, Boston, MA 02111, U.S.A
| |
Collapse
|
16
|
Kailash KA, Hawes JZ, Cocciolone AJ, Bersi MR, Mecham RP, Wagenseil JE. Constitutive Modeling of Mouse Arteries Suggests Changes in Directional Coupling and Extracellular Matrix Remodeling That Depend on Artery Type, Age, Sex, and Elastin Amounts. J Biomech Eng 2024; 146:051001. [PMID: 37646627 DOI: 10.1115/1.4063272] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Accepted: 08/21/2023] [Indexed: 09/01/2023]
Abstract
Arterial stiffening occurs during natural aging, is associated with an increased risk of adverse cardiovascular events, and can follow different timelines in males and females. One mechanism of arterial stiffening includes remodeling of the extracellular matrix (ECM), which alters the wall material properties. We used elastin haploinsufficient (Eln+/-) and wildtype (Eln+/+) mice to investigate how material properties of two different arteries (ascending aorta and carotid artery) change with age, sex, and ECM composition. We used a constitutive model by Dong and Sun that is based on the Holzapfel-Gasser-Ogden (HGO) type, but does not require a discrete number of fibrous ECM families and allows varied deformation coupling. We find that the amount of deformation coupling for the best fit model depends on the artery type. We also find that remodeling to maintain homeostatic (i.e., young, wildtype) values of biomechanical parameters with age, sex, and ECM composition depends on the artery type, with ascending aorta being more adaptable than carotid artery. Fitted material constants indicate sex-dependent remodeling that may be important for determining the time course of arterial stiffening in males and females. We correlated fitted material constants with ECM composition measured by biochemical (ascending aorta) or histological (carotid artery) methods. We show significant correlations between ECM composition and material parameters for the mean values for each group, with biochemical measurements correlating more strongly than histological measurements. Understanding how arterial stiffening depends on age, sex, ECM composition, and artery type may help design effective, personalized clinical treatment strategies.
Collapse
Affiliation(s)
- Keshav A Kailash
- Biomedical Engineering, Washington University, St. Louis, MO 63130
| | - Jie Z Hawes
- Mechanical Engineering and Materials Science, Washington University, St. Louis, MO 63130
| | - Austin J Cocciolone
- Mechanical Engineering and Materials Science, Washington University, St. Louis, MO 63130
| | - Matthew R Bersi
- Mechanical Engineering and Materials Science, Washington University, St. Louis, MO 63130
| | - Robert P Mecham
- Cell Biology and Physiology, Washington University, St. Louis, MO 63130
| | - Jessica E Wagenseil
- Mechanical Engineering and Materials Science, Washington University, One Brookings Dr., MSC 1185-208-125, St. Louis, MO 63130
| |
Collapse
|
17
|
Beachley V, Kuo J, Kasyanov V, Mironov V, Wen X. Biomimetic crimped/aligned microstructure to optimize the mechanics of fibrous hybrid materials for compliant vascular grafts. J Mech Behav Biomed Mater 2024; 150:106301. [PMID: 38141364 DOI: 10.1016/j.jmbbm.2023.106301] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Revised: 10/28/2023] [Accepted: 12/02/2023] [Indexed: 12/25/2023]
Abstract
The precise mechanical properties of many tissues are highly dependent on both the composition and arrangement of the nanofibrous extracellular matrix. It is well established that collagen nanofibers exhibit a crimped microstructure in several tissues such as blood vessel, tendon, and heart valve. This collagen fiber arrangement results in the classic non-linear 'J-shaped' stress strain curve characteristic of these tissues. Synthetic biomimetic fibrous materials with a crimped microstructure similar to natural collagen demonstrate similar mechanical properties to natural tissues. The following work describes a nanofabrication method based on electrospinning used to fabricate two component hybrid electrospun fibrous materials that mimic the microstructure and mechanical properties of vascular tissue. The properties of these samples can be precisely and predictably optimized by modifying fabrication parameters. Tubular grafts with biomimetic microstructure were constructed to demonstrate the potential of this fabrication method in vascular graft replacement applications. It was possible to closely match both the overall geometry and the compliance of specific blood vessels by optimizing graft microstructure.
Collapse
Affiliation(s)
- Vince Beachley
- Department of Biomedical Engineering, Rowan University, Glassboro, NJ, 08028, USA.
| | - Jonathan Kuo
- Department of Bioengineering, Clemson University, Clemson, SC, USA
| | | | - Vladimir Mironov
- Center for Biomedical Engineering, National University of Science and Technology (MISIS), Moscow, Russia
| | - Xuejun Wen
- Institute for Engineering and Medicine, Virgina Commonwealth University, Richmond, VA, USA
| |
Collapse
|
18
|
Parikh M, Pierce GN. Considerations for choosing an optimal animal model of cardiovascular disease. Can J Physiol Pharmacol 2024; 102:75-85. [PMID: 37748198 DOI: 10.1139/cjpp-2023-0206] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/27/2023]
Abstract
The decision to use the optimal animal model to mimic the various types of cardiovascular disease is a critical one for a basic scientist. Clinical cardiovascular disease can be complex and presents itself as atherosclerosis, hypertension, ischemia/reperfusion injury, myocardial infarcts, and cardiomyopathies, amongst others. This may be further complicated by the simultaneous presence of two or more cardiovascular lesions (for example, atherosclerosis and hypertension) and co-morbidities (i.e., diabetes, infectious disease, obesity, etc). This variety and merging of disease states creates an unusually difficult situation for the researcher who needs to identify the optimal animal model that is available to best represent all of the characteristics of the clinical cardiovascular disease. The present manuscript reviews the characteristics of the various animal models of cardiovascular disease available today, their advantages and disadvantages, with the goal to allow the reader access to the most recent data available for optimal choices prior to the initiation of the study. The animal species that can be chosen, the methods of generating these models of cardiovascular disease, as well as the specific cardiovascular lesions involved in each of these models are reviewed. A particular focus on the JCR:LA-cp rat as a model of cardiovascular disease is discussed.
Collapse
Affiliation(s)
- Mihir Parikh
- Department of Physiology and Pathophysiology, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB, Canada
- Institute of Cardiovascular Sciences, Albrechtsen Research Centre, St. Boniface Hospital, Winnipeg, MB, Canada
| | - Grant N Pierce
- Department of Physiology and Pathophysiology, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB, Canada
- Institute of Cardiovascular Sciences, Albrechtsen Research Centre, St. Boniface Hospital, Winnipeg, MB, Canada
| |
Collapse
|
19
|
Seeburun S, Wu S, Hemani D, Pham L, Ju D, Xie Y, Kata P, Li L. Insights into elastic fiber fragmentation: Mechanisms and treatment of aortic aneurysm in Marfan syndrome. Vascul Pharmacol 2023; 153:107215. [PMID: 37640090 PMCID: PMC10872825 DOI: 10.1016/j.vph.2023.107215] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2023] [Accepted: 08/25/2023] [Indexed: 08/31/2023]
Abstract
Marfan syndrome (MFS) is an autosomal dominant connective tissue disorder caused by mutations in fibrillin 1 (FBN1) gene. These mutations result in defects in the skeletal, ocular, and cardiovascular systems. Aortic aneurysm is the leading cause of premature mortality in untreated MFS patients. Elastic fiber fragmentation in the aortic vessel wall is a hallmark of MFS-associated aortic aneurysms. FBN1 mutations result in FBN1 fragments that also contribute to elastic fiber fragmentation. Although recent research has advanced our understanding of MFS, the contribution of elastic fiber fragmentation to the pathogenesis of aneurysm formation remains poorly understood. This review provides a comprehensive overview of the molecular mechanisms of elastic fiber fragmentation and its role in the pathogenesis of aortic aneurysm progression. Increased comprehension of elastic fragmentation has significant clinical implications for developing targeted interventions to block aneurysm progression, which would benefit not only individuals with Marfan syndrome but also other patients with aneurysms. Moreover, this review highlights an overlooked connection between inhibiting aneurysm and the restoration of elastic fibers in the vessel wall with various aneurysm inhibitors, including drugs and chemicals. Investigating the underlying molecular mechanisms could uncover innovative therapeutic strategies to inhibit elastin fragmentation and prevent the progression of aneurysms.
Collapse
Affiliation(s)
- Sheilabi Seeburun
- Department of Internal Medicine, Wayne State University, Detroit MI, USA
| | - Shichao Wu
- Department of Internal Medicine, Wayne State University, Detroit MI, USA
| | - Darshi Hemani
- Center for Molecular Medicine and Genetics, Wayne State University, Detroit MI, USA
| | - Lucynda Pham
- Department of Internal Medicine, Wayne State University, Detroit MI, USA
- Center for Molecular Medicine and Genetics, Wayne State University, Detroit MI, USA
| | - Donghong Ju
- Department of Internal Medicine, Wayne State University, Detroit MI, USA
- Department of Oncology, Wayne State University, Detroit MI, USA
| | - Youming Xie
- Department of Oncology, Wayne State University, Detroit MI, USA
| | - Priyaranjan Kata
- Department of Internal Medicine, Wayne State University, Detroit MI, USA
| | - Li Li
- Department of Internal Medicine, Wayne State University, Detroit MI, USA
- Center for Molecular Medicine and Genetics, Wayne State University, Detroit MI, USA
| |
Collapse
|
20
|
Hu X, Logan JG, Kwon Y, Lima JAC, Jacobs DR, Duprez D, Brumback L, Taylor KD, Durda P, Johnson WC, Cornell E, Guo X, Liu Y, Tracy RP, Blackwell TW, Papanicolaou G, Mitchell GF, Rich SS, Rotter JI, Van Den Berg DJ, Chirinos JA, Hughes TM, Garrett-Bakelman FE, Manichaikul A. Multi-ancestry epigenome-wide analyses identify methylated sites associated with aortic augmentation index in TOPMed MESA. Sci Rep 2023; 13:17680. [PMID: 37848499 PMCID: PMC10582077 DOI: 10.1038/s41598-023-44806-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Accepted: 10/12/2023] [Indexed: 10/19/2023] Open
Abstract
Despite the prognostic value of arterial stiffness (AS) and pulsatile hemodynamics (PH) for cardiovascular morbidity and mortality, epigenetic modifications that contribute to AS/PH remain unknown. To gain a better understanding of the link between epigenetics (DNA methylation) and AS/PH, we examined the relationship of eight measures of AS/PH with CpG sites and co-methylated regions using multi-ancestry participants from Trans-Omics for Precision Medicine (TOPMed) Multi-Ethnic Study of Atherosclerosis (MESA) with sample sizes ranging from 438 to 874. Epigenome-wide association analysis identified one genome-wide significant CpG (cg20711926-CYP1B1) associated with aortic augmentation index (AIx). Follow-up analyses, including gene set enrichment analysis, expression quantitative trait methylation analysis, and functional enrichment analysis on differentially methylated positions and regions, further prioritized three CpGs and their annotated genes (cg23800023-ETS1, cg08426368-TGFB3, and cg17350632-HLA-DPB1) for AIx. Among these, ETS1 and TGFB3 have been previously prioritized as candidate genes. Furthermore, both ETS1 and HLA-DPB1 have significant tissue correlations between Whole Blood and Aorta in GTEx, which suggests ETS1 and HLA-DPB1 could be potential biomarkers in understanding pathophysiology of AS/PH. Overall, our findings support the possible role of epigenetic regulation via DNA methylation of specific genes associated with AIx as well as identifying potential targets for regulation of AS/PH.
Collapse
Affiliation(s)
- Xiaowei Hu
- Center for Public Health Genomics, University of Virginia, Charlottesville, VA, 22908, USA
| | - Jeongok G Logan
- School of Nursing, University of Virginia, Charlottesville, VA, USA
| | - Younghoon Kwon
- Department of Medicine, University of Washington, Seattle, WA, USA
| | - Joao A C Lima
- Department of Internal Medicine, School of Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - David R Jacobs
- Division of Epidemiology, School of Public Health, University of Minnesota, Minneapolis, MN, USA
| | - Daniel Duprez
- Cardiovascular Division, University of Minnesota, Minneapolis, MN, USA
| | - Lyndia Brumback
- Department of Biostatistics, University of Washington, Seattle, WA, USA
| | - Kent D Taylor
- The Institute for Translational Genomics and Population Sciences, Department of Pediatrics, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA, USA
| | - Peter Durda
- Laboratory for Clinical Biochemistry Research, University of Vermont, Burlington, VT, USA
| | - W Craig Johnson
- Department of Biostatistics, University of Washington, Seattle, WA, USA
| | - Elaine Cornell
- Laboratory for Clinical Biochemistry Research, University of Vermont, Burlington, VT, USA
| | - Xiuqing Guo
- The Institute for Translational Genomics and Population Sciences, Department of Pediatrics, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA, USA
| | - Yongmei Liu
- Division of Cardiology, Department of Medicine, Duke University, Durham, NC, USA
| | - Russell P Tracy
- Laboratory for Clinical Biochemistry Research, University of Vermont, Burlington, VT, USA
| | - Thomas W Blackwell
- Department of Biostatistics, School of Public Health, University of Michigan, Ann Arbor, MI, USA
| | - George Papanicolaou
- Epidemiology Branch, National Heart, Lung and Blood Institute, Bethesda, MD, USA
| | | | - Stephen S Rich
- Center for Public Health Genomics, University of Virginia, Charlottesville, VA, 22908, USA
| | - Jerome I Rotter
- The Institute for Translational Genomics and Population Sciences, Department of Pediatrics, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA, USA
| | - David J Van Den Berg
- Department of Preventive Medicine and Center for Genetic Epidemiology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Julio A Chirinos
- Division of Cardiovascular Medicine, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Timothy M Hughes
- Department of Internal Medicine - Section of Gerontology and Geriatric Medicine, and Department of Epidemiology and Prevention, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Francine E Garrett-Bakelman
- Department of Biochemistry and Molecular Genetics, Department of Medicine, University of Virginia, 1340 Jefferson Park Ave., Pinn hall 6054, Charlottesville, VA, 22908, USA.
| | - Ani Manichaikul
- Center for Public Health Genomics, University of Virginia, Charlottesville, VA, 22908, USA.
| |
Collapse
|
21
|
Huryn LA, Flaherty T, Nolen R, Prasov L, Zein WM, Cukras CA, Osgood S, Raja N, Levin MD, Vitale S, Brooks BP, Hufnagel RB, Kozel BA. Novel ophthalmic findings and deep phenotyping in Williams-Beuren syndrome. Br J Ophthalmol 2023; 107:1554-1559. [PMID: 35760456 PMCID: PMC10074447 DOI: 10.1136/bjophthalmol-2022-321103] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Accepted: 05/26/2022] [Indexed: 11/03/2022]
Abstract
BACKGROUND/AIMS To characterise the ocular manifestations of Williams-Beuren syndrome (WBS) and compare these to patients with isolated elastin mediated supravalvular aortic stenosis (SVAS). METHODS Fifty-seven patients with a diagnosis of WBS and five with SVAS underwent comprehensive ophthalmic evaluation at the National Institutes of Health from 2017 to 2020, including best-corrected visual acuity, slit-lamp biomicroscopy, optical biometry, dilated fundus examination, optical coherence tomography and colour fundus imaging. RESULTS Mean age of the 57 WBS patients was 20.3 years (range 3-60 years). Best-corrected visual acuity ranged from 20/20 to 20/400 with mean spherical equivalent near plano OU. Twenty-four eyes (21.8%) had an axial length (AL) less than 20.5 mm and 38 eyes (34.5%) had an AL measuring 20.5-22.0 mm. Stellate iris and retinal arteriolar tortuosity were noted in 30 (52.6%) and 51 (89.5%) WBS patients, respectively. Novel retinal findings in WBS included small hypopigmented retinal deposits (OD 29/57, OS 27/57) and broad foveal pit contour (OD 44/55, OS 42/51). Of the five patients with SVAS, none had stellate iris or broad foveal pit contour while 2/5 had retinal arteriolar tortuosity. CONCLUSION WBS is a complex multisystem genetic disorder with diverse ophthalmic findings that differ from those seen in isolated elastin mediated SVAS. These results suggest other genes within the WBS critical region, aside from ELN, may be involved in observed ocular phenotypes and perhaps broader ocular development. Furthermore, retinal arteriolar tortuosity may provide future insight into systemic vascular findings in WBS.
Collapse
Affiliation(s)
- Laryssa A Huryn
- Ophthalmic Genetics and Visual Function Branch, National Eye Institute, Bethesda, Maryland, USA
| | - Taylor Flaherty
- Ophthalmic Genetics and Visual Function Branch, National Eye Institute, Bethesda, Maryland, USA
| | - Rosalie Nolen
- Ophthalmic Genetics and Visual Function Branch, National Eye Institute, Bethesda, Maryland, USA
| | - Lev Prasov
- Ophthalmic Genetics and Visual Function Branch, National Eye Institute, Bethesda, Maryland, USA
- Department of Ophthalmology and Visual Sciences, W K Kellogg Eye Center, Ann Arbor, Michigan, USA
- Department of Human Genetics, University of Michigan, Ann Arbor, Michigan, USA
| | - Wadih M Zein
- Ophthalmic Genetics and Visual Function Branch, National Eye Institute, Bethesda, Maryland, USA
| | - Catherine A Cukras
- Division of Epidemiology and Clinical Applications, National Eye Institute, NIH, Bethesda, Maryland, USA
| | - Sharon Osgood
- Translational Vascular Medicine Branch, National Heart Lung and Blood Institute, Bethesda, Maryland, USA
| | - Neelam Raja
- Translational Vascular Medicine Branch, National Heart Lung and Blood Institute, Bethesda, Maryland, USA
| | - Mark D Levin
- Translational Vascular Medicine Branch, National Heart Lung and Blood Institute, Bethesda, Maryland, USA
| | - Susan Vitale
- Division of Epidemiology and Clinical Applications, National Eye Institute, NIH, Bethesda, Maryland, USA
| | - Brian P Brooks
- Ophthalmic Genetics and Visual Function Branch, National Eye Institute, Bethesda, Maryland, USA
| | - Robert B Hufnagel
- Ophthalmic Genetics and Visual Function Branch, National Eye Institute, Bethesda, Maryland, USA
| | - Beth A Kozel
- Translational Vascular Medicine Branch, National Heart Lung and Blood Institute, Bethesda, Maryland, USA
| |
Collapse
|
22
|
Neutel CHG, Wesley CD, Van Praet M, Civati C, Roth L, De Meyer GRY, Martinet W, Guns PJ. Empagliflozin decreases ageing-associated arterial stiffening and vascular fibrosis under normoglycemic conditions. Vascul Pharmacol 2023; 152:107212. [PMID: 37619798 DOI: 10.1016/j.vph.2023.107212] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Revised: 08/14/2023] [Accepted: 08/21/2023] [Indexed: 08/26/2023]
Abstract
Arterial stiffness is a hallmark of vascular ageing and results in increased blood flow pulsatility to the periphery, damaging end-organs such as the heart, kidneys and brain. Treating or "reversing" arterial stiffness has therefore become a central target in the field of vascular ageing. SGLT2 inhibitors, initially developed in the context of type 2 diabetes mellitus, have become a cornerstone of heart failure treatment. Additionally, effects on the vasculature have been reported. Here, we demonstrate that treatment with the SGLT2 inhibitor empagliflozin (7 weeks, 15 mg/kg/day) decreased ageing-induced arterial stiffness of the aorta in old mice with normal blood glucose levels. However, no universal mechanism was identified. While empagliflozin reduced the ageing-associated increase in collagen type I in the medial layer of the abdominal infrarenal aorta and decreased medial TGF-β deposition, this was not observed in the thoracic descending aorta. Moreover, empagliflozin was not able to prevent elastin fragmentation. In conclusion, empagliflozin decreased arterial stiffness in aged mice, indicating that SGLT2 inhibition could be a valuable strategy in mitigating vascular ageing. Further research is warranted to unravel the underlying, possibly region-specific, mechanisms.
Collapse
Affiliation(s)
- Cédric H G Neutel
- Laboratory of Physiopharmacology, University of Antwerp, Campus Drie Eiken, Antwerp, Belgium.
| | - Callan D Wesley
- Laboratory of Physiopharmacology, University of Antwerp, Campus Drie Eiken, Antwerp, Belgium
| | - Melissa Van Praet
- Laboratory of Physiopharmacology, University of Antwerp, Campus Drie Eiken, Antwerp, Belgium
| | - Celine Civati
- Laboratory of Physiopharmacology, University of Antwerp, Campus Drie Eiken, Antwerp, Belgium
| | - Lynn Roth
- Laboratory of Physiopharmacology, University of Antwerp, Campus Drie Eiken, Antwerp, Belgium
| | - Guido R Y De Meyer
- Laboratory of Physiopharmacology, University of Antwerp, Campus Drie Eiken, Antwerp, Belgium
| | - Wim Martinet
- Laboratory of Physiopharmacology, University of Antwerp, Campus Drie Eiken, Antwerp, Belgium
| | - Pieter-Jan Guns
- Laboratory of Physiopharmacology, University of Antwerp, Campus Drie Eiken, Antwerp, Belgium
| |
Collapse
|
23
|
Atkinson G, Bianco R, Di Gregoli K, Johnson JL. The contribution of matrix metalloproteinases and their inhibitors to the development, progression, and rupture of abdominal aortic aneurysms. Front Cardiovasc Med 2023; 10:1248561. [PMID: 37799778 PMCID: PMC10549934 DOI: 10.3389/fcvm.2023.1248561] [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: 06/27/2023] [Accepted: 09/07/2023] [Indexed: 10/07/2023] Open
Abstract
Abdominal aortic aneurysms (AAAs) account for up to 8% of deaths in men aged 65 years and over and 2.2% of women. Patients with AAAs often have atherosclerosis, and intimal atherosclerosis is generally present in AAAs. Accordingly, AAAs are considered a form of atherosclerosis and are frequently referred to as atherosclerotic aneurysms. Pathological observations advocate inflammatory cell infiltration alongside adverse extracellular matrix degradation as key contributing factors to the formation of human atherosclerotic AAAs. Therefore, macrophage production of proteolytic enzymes is deemed responsible for the damaging loss of ECM proteins, especially elastin and fibrillar collagens, which characterise AAA progression and rupture. Matrix metalloproteinases (MMPs) and their regulation by tissue inhibitors metalloproteinases (TIMPs) can orchestrate not only ECM remodelling, but also moderate the proliferation, migration, and apoptosis of resident aortic cells, alongside the recruitment and subsequent behaviour of inflammatory cells. Accordingly, MMPs are thought to play a central regulatory role in the development, progression, and eventual rupture of abdominal aortic aneurysms (AAAs). Together, clinical and animal studies have shed light on the complex and often diverse effects MMPs and TIMPs impart during the development of AAAs. This dichotomy is underlined from evidence utilising broad-spectrum MMP inhibition in animal models and clinical trials which have failed to provide consistent protection from AAA progression, although more encouraging results have been observed through deployment of selective inhibitors. This review provides a summary of the supporting evidence connecting the contribution of individual MMPs to AAA development, progression, and eventual rupture. Topics discussed include structural, functional, and cell-specific diversity of MMP members; evidence from animal models of AAA and comparisons with findings in humans; the dual role of MMPs and the requirement to selectively target individual MMPs; and the advances in identifying aberrant MMP activity. As evidenced, our developing understanding of the multifaceted roles individual MMPs perform during the progression and rupture of AAAs, should motivate clinical trials assessing the therapeutic potential of selective MMP inhibitors, which could restrict AAA-related morbidity and mortality worldwide.
Collapse
Affiliation(s)
| | | | | | - Jason L. Johnson
- Laboratory of Cardiovascular Pathology, Department of Translational Health Sciences, Bristol Medical School, University of Bristol, Bristol, United Kingdom
| |
Collapse
|
24
|
An DW, Hansen TW, Aparicio LS, Chori B, Huang QF, Wei FF, Cheng YB, Yu YL, Sheng CS, Gilis-Malinowska N, Boggia J, Wojciechowska W, Niiranen TJ, Tikhonoff V, Casiglia E, Narkiewicz K, Stolarz-Skrzypek K, Kawecka-Jaszcz K, Jula AM, Yang WY, Woodiwiss AJ, Filipovský J, Wang JG, Rajzer MW, Verhamme P, Nawrot TS, Staessen JA, Li Y. Derivation of an Outcome-Driven Threshold for Aortic Pulse Wave Velocity: An Individual-Participant Meta-Analysis. Hypertension 2023; 80:1949-1959. [PMID: 37470187 PMCID: PMC10424824 DOI: 10.1161/hypertensionaha.123.21318] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Accepted: 06/08/2023] [Indexed: 07/21/2023]
Abstract
BACKGROUND Aortic pulse wave velocity (PWV) predicts cardiovascular events (CVEs) and total mortality (TM), but previous studies proposing actionable PWV thresholds have limited generalizability. This individual-participant meta-analysis is aimed at defining, testing calibration, and validating an outcome-driven threshold for PWV, using 2 populations studies, respectively, for derivation IDCARS (International Database of Central Arterial Properties for Risk Stratification) and replication MONICA (Monitoring of Trends and Determinants in Cardiovascular Disease Health Survey - Copenhagen). METHODS A risk-carrying PWV threshold for CVE and TM was defined by multivariable Cox regression, using stepwise increasing PWV thresholds and by determining the threshold yielding a 5-year risk equivalent with systolic blood pressure of 140 mm Hg. The predictive performance of the PWV threshold was assessed by computing the integrated discrimination improvement and the net reclassification improvement. RESULTS In well-calibrated models in IDCARS, the risk-carrying PWV thresholds converged at 9 m/s (10 m/s considering the anatomic pulse wave travel distance). With full adjustments applied, the threshold predicted CVE (hazard ratio [CI]: 1.68 [1.15-2.45]) and TM (1.61 [1.01-2.55]) in IDCARS and in MONICA (1.40 [1.09-1.79] and 1.55 [1.23-1.95]). In IDCARS and MONICA, the predictive accuracy of the threshold for both end points was ≈0.75. Integrated discrimination improvement was significant for TM in IDCARS and for both TM and CVE in MONICA, whereas net reclassification improvement was not for any outcome. CONCLUSIONS PWV integrates multiple risk factors into a single variable and might replace a large panel of traditional risk factors. Exceeding the outcome-driven PWV threshold should motivate clinicians to stringent management of risk factors, in particular hypertension, which over a person's lifetime causes stiffening of the elastic arteries as waypoint to CVE and death.
Collapse
Affiliation(s)
- De-Wei An
- Department of Cardiovascular Medicine, Shanghai Key Laboratory of Hypertension, Shanghai Institute of Hypertension, State Key Laboratory of Medical Genomics, National Research Centre for Translational Medicine, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, China (D.-W.A., Q.-F.H., Y. B.C., C. S.S., J. G.W., Y.L.)
- Non-Profit Research Association Alliance for the Promotion of Preventive Medicine, Belgium (D.-W.A, T.W.H, B.C., Y.-L.Y., J.A.S.)
- Research Unit Environment and Health, Department of Public Health and Primary Care, University of Leuven, Belgium (D.-W.A, Y.-L.Y., T.S.N.)
| | - Tine W. Hansen
- Non-Profit Research Association Alliance for the Promotion of Preventive Medicine, Belgium (D.-W.A, T.W.H, B.C., Y.-L.Y., J.A.S.)
- The Steno Diabetes Center Copenhagen, Herlev, and Center for Health, Capital Region of Denmark, Copenhagen (T.W.H.)
| | - Lucas S. Aparicio
- Servicio de Clínica Médica, Sección Hipertensión Arterial, Hospital Italiano de Buenos Aires, Argentina (L.S.A.)
| | - Babangida Chori
- Non-Profit Research Association Alliance for the Promotion of Preventive Medicine, Belgium (D.-W.A, T.W.H, B.C., Y.-L.Y., J.A.S.)
- Centre for Environmental Sciences, Hasselt University, Belgium (B.C., T.S.N.)
- Department of Internal Medicine, Faculty of Clinical Sciences, College of Health Sciences, University of Abuja, Nigeria (B.C.)
| | - Qi-Fang Huang
- Department of Cardiovascular Medicine, Shanghai Key Laboratory of Hypertension, Shanghai Institute of Hypertension, State Key Laboratory of Medical Genomics, National Research Centre for Translational Medicine, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, China (D.-W.A., Q.-F.H., Y. B.C., C. S.S., J. G.W., Y.L.)
| | - Fang-Fei Wei
- Department of Cardiology, the First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China (F.-F.W.)
| | - Yi-Bang Cheng
- Department of Cardiovascular Medicine, Shanghai Key Laboratory of Hypertension, Shanghai Institute of Hypertension, State Key Laboratory of Medical Genomics, National Research Centre for Translational Medicine, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, China (D.-W.A., Q.-F.H., Y. B.C., C. S.S., J. G.W., Y.L.)
| | - Yu-Ling Yu
- Non-Profit Research Association Alliance for the Promotion of Preventive Medicine, Belgium (D.-W.A, T.W.H, B.C., Y.-L.Y., J.A.S.)
- Research Unit Environment and Health, Department of Public Health and Primary Care, University of Leuven, Belgium (D.-W.A, Y.-L.Y., T.S.N.)
| | - Chang-Sheng Sheng
- Department of Cardiovascular Medicine, Shanghai Key Laboratory of Hypertension, Shanghai Institute of Hypertension, State Key Laboratory of Medical Genomics, National Research Centre for Translational Medicine, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, China (D.-W.A., Q.-F.H., Y. B.C., C. S.S., J. G.W., Y.L.)
| | - Natasza Gilis-Malinowska
- Hypertension Unit, Department of Hypertension and Diabetology, Medical University of Gdańsk, Poland (N.G.-M., K.N.)
| | - José Boggia
- Centro de Nefrología and Departamento de Fisiopatología, Hospital de Clínicas, Universidad de la República, Montevideo, Uruguay (J.B.)
| | - Wiktoria Wojciechowska
- First Department of Cardiology, Interventional Electrocardiology and Hypertension, Jagiellonian University Medical College, Kraków, Poland (W.W., K.S.-S., M.R., K.K.-J)
| | - Teemu J. Niiranen
- Department of Chronic Disease Prevention, Finnish Institute for Health and Welfare, Turku, Finland (T.J.N., A.M.J.)
- Department of Medicine, Turku University Hospital and University of Turku, Finland (T.J.N., A.M.J.)
| | | | - Edoardo Casiglia
- Department of Medicine, University of Padova, Italy (V.T., E.C.)
| | - Krzysztof Narkiewicz
- Hypertension Unit, Department of Hypertension and Diabetology, Medical University of Gdańsk, Poland (N.G.-M., K.N.)
| | - Katarzyna Stolarz-Skrzypek
- First Department of Cardiology, Interventional Electrocardiology and Hypertension, Jagiellonian University Medical College, Kraków, Poland (W.W., K.S.-S., M.R., K.K.-J)
| | - Kalina Kawecka-Jaszcz
- First Department of Cardiology, Interventional Electrocardiology and Hypertension, Jagiellonian University Medical College, Kraków, Poland (W.W., K.S.-S., M.R., K.K.-J)
| | - Antti M. Jula
- Department of Chronic Disease Prevention, Finnish Institute for Health and Welfare, Turku, Finland (T.J.N., A.M.J.)
- Department of Medicine, Turku University Hospital and University of Turku, Finland (T.J.N., A.M.J.)
| | - Wen-Yi Yang
- Department of Cardiology, Shanghai General Hospital, China (W.-Y.Y.)
| | - Angela J. Woodiwiss
- School of Physiology, University of the Witwatersrand, Johannesburg, South Africa (A.W.)
| | - Jan Filipovský
- Faculty of Medicine, Charles University, Pilsen, Czech Republic (J.F.)
| | - Ji-Guang Wang
- Department of Cardiovascular Medicine, Shanghai Key Laboratory of Hypertension, Shanghai Institute of Hypertension, State Key Laboratory of Medical Genomics, National Research Centre for Translational Medicine, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, China (D.-W.A., Q.-F.H., Y. B.C., C. S.S., J. G.W., Y.L.)
| | - Marek W. Rajzer
- First Department of Cardiology, Interventional Electrocardiology and Hypertension, Jagiellonian University Medical College, Kraków, Poland (W.W., K.S.-S., M.R., K.K.-J)
| | - Peter Verhamme
- Center for Molecular and Vascular Biology, KU Leuven Department of Cardiovascular Sciences, University of Leuven, Belgium (P.V.)
| | - Tim S. Nawrot
- Research Unit Environment and Health, Department of Public Health and Primary Care, University of Leuven, Belgium (D.-W.A, Y.-L.Y., T.S.N.)
- Centre for Environmental Sciences, Hasselt University, Belgium (B.C., T.S.N.)
| | - Jan A. Staessen
- Non-Profit Research Association Alliance for the Promotion of Preventive Medicine, Belgium (D.-W.A, T.W.H, B.C., Y.-L.Y., J.A.S.)
- Biomedical Science Group, Faculty of Medicine, University of Leuven, Belgium (J.A.S.)
| | - Yan Li
- Department of Cardiovascular Medicine, Shanghai Key Laboratory of Hypertension, Shanghai Institute of Hypertension, State Key Laboratory of Medical Genomics, National Research Centre for Translational Medicine, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, China (D.-W.A., Q.-F.H., Y. B.C., C. S.S., J. G.W., Y.L.)
| |
Collapse
|
25
|
Chen Y, Yuan Y, Chen Y, Jiang X, Hua X, Chen Z, Wang J, Liu H, Zhou Q, Yu Y, Yang Z, Yu Y, Wang Y, Wang Q, Li Y, Chen J, Wang Y. Novel signaling axis of FHOD1-RNF213-Col1α/Col3α in the pathogenesis of hypertension-induced tunica media thickening. J Mol Cell Cardiol 2023; 182:57-72. [PMID: 37482037 DOI: 10.1016/j.yjmcc.2023.07.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Revised: 07/05/2023] [Accepted: 07/15/2023] [Indexed: 07/25/2023]
Abstract
Hypertension-induced tunica media thickening (TMT) is the most important fundamental for the subsequent complications like stroke and cardiovascular diseases. Pathogenically, TMT originates from both vascular smooth muscle cells (VSMCs) hypertrophy due to synthesizing more amount of intracellular contractile proteins and excess secretion of extracellular matrix. However, what key molecules are involved in the pathogenesis of TMT is unknown. We hypothesize that formin homology 2 domain-containing protein 1 (FHOD1), an amply expressed mediator for assembly of thin actin filament in VSMCs, is a key regulator for the pathogenesis of TMT. In this study, we found that FHOD1 expression and its phosphorylation/activation were both upregulated in the arteries of three kinds of hypertensive rats. Ang-II induced actin filament formation and hypertrophy through activation and upregulation of FHOD1 in VSMCs. Active FHOD1-mediated actin filament assembly and secretions of collagen-1α/collagen-3α played crucial roles in Ang-II-induced VSMCs hypertrophy in vitro and hypertensive TMT in vivo. Proteomics demonstrated that activated FL-FHOD1 or its C-terminal diaphanous-autoregulatory domain significantly upregulated RNF213 (ring finger protein 213), a 591-kDa cytosolic E3 ubiquitin ligase with its loss-of-functional mutations being a susceptibility gene for Moyamoya disease which has prominent tunica media thinning in both intracranial and systemic arteries. Mechanistically, activated FHOD1 upregulated its downstream effector RNF213 independently of its classical pathway of decreasing G-actin/F-actin ratio, transcription, and translation, but dependently on its C-terminus-mediated stabilization of RNF213 protein. FHOD1-RNF213 signaling dramatically promoted collagen-1α/collagen-3α syntheses in VSMCs. Our results discovered a novel signaling axis of FHOD1-RNF213-collagen-1α/collagen-3α and its key role in the pathogenesis of hypertensive TMT.
Collapse
Affiliation(s)
- Yuanyuan Chen
- Molecular Cardiology Research Laboratory, Department of Cardiology, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200092, China
| | - Yuchan Yuan
- Molecular Cardiology Research Laboratory, Department of Cardiology, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200092, China
| | - Yuhan Chen
- Molecular Cardiology Research Laboratory, Department of Cardiology, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200092, China
| | - Xueze Jiang
- Molecular Cardiology Research Laboratory, Department of Cardiology, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200092, China
| | - Xuesheng Hua
- Molecular Cardiology Research Laboratory, Department of Cardiology, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200092, China
| | - Zhiyong Chen
- Molecular Cardiology Research Laboratory, Department of Cardiology, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200092, China
| | - Julie Wang
- Department of Computer Science, Brown University, Providence, RI 02912, USA
| | - Hua Liu
- Department of Intensive Care Med, Zhongshan Hospital of Fudan University, Shanghai 200032, China
| | - Qing Zhou
- Molecular Cardiology Research Laboratory, Department of Cardiology, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200092, China
| | - Ying Yu
- Molecular Cardiology Research Laboratory, Department of Cardiology, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200092, China
| | - Zhenwei Yang
- Molecular Cardiology Research Laboratory, Department of Cardiology, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200092, China
| | - Yi Yu
- Molecular Cardiology Research Laboratory, Department of Cardiology, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200092, China
| | - Yongqin Wang
- Division of Rheumatology and Immunology, University of Toledo Medical center, 3120 Glendale Avenue, Toledo, OH 43614, USA
| | - Qunshan Wang
- Molecular Cardiology Research Laboratory, Department of Cardiology, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200092, China
| | - Yigang Li
- Molecular Cardiology Research Laboratory, Department of Cardiology, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200092, China
| | - Jie Chen
- Molecular Cardiology Research Laboratory, Department of Cardiology, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200092, China.
| | - Yuepeng Wang
- Molecular Cardiology Research Laboratory, Department of Cardiology, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200092, China.
| |
Collapse
|
26
|
Sakr HF, Sirasanagandla SR, Das S, Bima AI, Elsamanoudy AZ. Insulin Resistance and Hypertension: Mechanisms Involved and Modifying Factors for Effective Glucose Control. Biomedicines 2023; 11:2271. [PMID: 37626767 PMCID: PMC10452601 DOI: 10.3390/biomedicines11082271] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2023] [Revised: 07/18/2023] [Accepted: 08/01/2023] [Indexed: 08/27/2023] Open
Abstract
Factors such as aging, an unhealthy lifestyle with decreased physical activity, snacking, a standard Western diet, and smoking contribute to raising blood pressure to a dangerous level, increasing the risk of coronary artery disease and heart failure. Atherosclerosis, or aging of the blood vessels, is a physiological process that has accelerated in the last decades by the overconsumption of carbohydrates as the primary sources of caloric intake, resulting in increased triglycerides and VLDL-cholesterol and insulin spikes. Classically, medications ranging from beta blockers to angiotensin II blockers and even calcium channel blockers were used alone or in combination with lifestyle modifications as management tools in modern medicine to control arterial blood pressure. However, it is not easy to control blood pressure or the associated complications. A low-carbohydrate, high-fat (LCHF) diet can reduce glucose and insulin spikes, improve insulin sensitivity, and lessen atherosclerosis risk factors. We reviewed articles describing the etiology of insulin resistance (IR) and its impact on arterial blood pressure from databases including PubMed, PubMed Central, and Google Scholar. We discuss how the LCHF diet is beneficial to maintaining arterial blood pressure at normal levels, slowing down the progression of atherosclerosis, and reducing the use of antihypertensive medications. The mechanisms involved in IR associated with hypertension are also highlighted.
Collapse
Affiliation(s)
- Hussein F. Sakr
- Department of Physiology, College of Medicine and Health Sciences, Sultan Qaboos University, Muscat 123, Oman
| | - Srinivasa Rao Sirasanagandla
- Department of Human and Clinical Anatomy, College of Medicine and Health Sciences, Sultan Qaboos University, Muscat 123, Oman; (S.R.S.); (S.D.)
| | - Srijit Das
- Department of Human and Clinical Anatomy, College of Medicine and Health Sciences, Sultan Qaboos University, Muscat 123, Oman; (S.R.S.); (S.D.)
| | - Abdulhadi I. Bima
- Department of Clinical Biochemistry, Faculty of Medicine, King Abdulaziz University, Jeddah 21465, Saudi Arabia; (A.I.B.); (A.Z.E.)
| | - Ayman Z. Elsamanoudy
- Department of Clinical Biochemistry, Faculty of Medicine, King Abdulaziz University, Jeddah 21465, Saudi Arabia; (A.I.B.); (A.Z.E.)
| |
Collapse
|
27
|
Wang D, Brady T, Santhanam L, Gerecht S. The extracellular matrix mechanics in the vasculature. NATURE CARDIOVASCULAR RESEARCH 2023; 2:718-732. [PMID: 39195965 DOI: 10.1038/s44161-023-00311-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/13/2022] [Accepted: 06/20/2023] [Indexed: 08/29/2024]
Abstract
Mechanical stimuli from the extracellular matrix (ECM) modulate vascular differentiation, morphogenesis and dysfunction of the vasculature. With innovation in measurements, we can better characterize vascular microenvironment mechanics in health and disease. Recent advances in material sciences and stem cell biology enable us to accurately recapitulate the complex and dynamic ECM mechanical microenvironment for in vitro studies. These biomimetic approaches help us understand the signaling pathways in disease pathologies, identify therapeutic targets, build tissue replacement and activate tissue regeneration. This Review analyzes how ECM mechanics regulate vascular homeostasis and dysfunction. We highlight approaches to examine ECM mechanics at tissue and cellular levels, focusing on how mechanical interactions between cells and the ECM regulate vascular phenotype, especially under certain pathological conditions. Finally, we explore the development of biomaterials to emulate, measure and alter the physical microenvironment of pathological ECM to understand cell-ECM mechanical interactions toward the development of therapeutics.
Collapse
Affiliation(s)
- Dafu Wang
- Department of Biomedical Engineering, Duke University, Durham, NC, USA
| | - Travis Brady
- Department of Anesthesiology and Critical Care Medicine and Department of Biomedical Engineering, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Lakshmi Santhanam
- Department of Anesthesiology and Critical Care Medicine and Department of Biomedical Engineering, Johns Hopkins School of Medicine, Baltimore, MD, USA
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, USA
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Sharon Gerecht
- Department of Biomedical Engineering, Duke University, Durham, NC, USA.
| |
Collapse
|
28
|
Karageorgos GM, Liang P, Mobadersany N, Gami P, Konofagou EE. Unsupervised deep learning-based displacement estimation for vascular elasticity imaging applications. Phys Med Biol 2023; 68:10.1088/1361-6560/ace0f0. [PMID: 37348487 PMCID: PMC10528442 DOI: 10.1088/1361-6560/ace0f0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2023] [Accepted: 06/22/2023] [Indexed: 06/24/2023]
Abstract
Objective. Arterial wall stiffness can provide valuable information on the proper function of the cardiovascular system. Ultrasound elasticity imaging techniques have shown great promise as a low-cost and non-invasive tool to enable localized maps of arterial wall stiffness. Such techniques rely upon motion detection algorithms that provide arterial wall displacement estimation.Approach. In this study, we propose an unsupervised deep learning-based approach, originally proposed for image registration, in order to enable improved quality arterial wall displacement estimation at high temporal and spatial resolutions. The performance of the proposed network was assessed through phantom experiments, where various models were trained by using ultrasound RF signals, or B-mode images, as well as different loss functions.Main results. Using the mean square error (MSE) for the training process provided the highest signal-to-noise ratio when training on the B-modes images (30.36 ± 1.14 dB) and highest contrast-to-noise ratio when training on the RF signals (32.84 ± 1.89 dB). In addition, training the model on RF signals demonstrated the capability of providing accurate localized pulse wave velocity (PWV) maps, with a mean relative error (MREPWV) of 3.32 ± 1.80% and anR2 of 0.97 ± 0.03. Finally, the developed model was tested in human common carotid arteriesin vivo, providing accurate tracking of the distension pulse wave propagation, with an MREPWV= 3.86 ± 2.69% andR2 = 0.95 ± 0.03.Significance. In conclusion, a novel displacement estimation approach was presented, showing promise in improving vascular elasticity imaging techniques.
Collapse
Affiliation(s)
- Grigorios M Karageorgos
- Biomedical Engineering Department, Columbia University, New York, NY, United States of America
| | - Pengcheng Liang
- Biomedical Engineering Department, Columbia University, New York, NY, United States of America
| | - Nima Mobadersany
- Department of Radiology, Columbia University, New York, NY, United States of America
| | - Parth Gami
- Biomedical Engineering Department, Columbia University, New York, NY, United States of America
| | - Elisa E Konofagou
- Biomedical Engineering Department, Columbia University, New York, NY, United States of America
- Department of Radiology, Columbia University, New York, NY, United States of America
| |
Collapse
|
29
|
Kucherenko MM, Sang P, Yao J, Gransar T, Dhital S, Grune J, Simmons S, Michalick L, Wulsten D, Thiele M, Shomroni O, Hennig F, Yeter R, Solowjowa N, Salinas G, Duda GN, Falk V, Vyavahare NR, Kuebler WM, Knosalla C. Elastin stabilization prevents impaired biomechanics in human pulmonary arteries and pulmonary hypertension in rats with left heart disease. Nat Commun 2023; 14:4416. [PMID: 37479718 PMCID: PMC10362055 DOI: 10.1038/s41467-023-39934-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Accepted: 07/04/2023] [Indexed: 07/23/2023] Open
Abstract
Pulmonary hypertension worsens outcome in left heart disease. Stiffening of the pulmonary artery may drive this pathology by increasing right ventricular dysfunction and lung vascular remodeling. Here we show increased stiffness of pulmonary arteries from patients with left heart disease that correlates with impaired pulmonary hemodynamics. Extracellular matrix remodeling in the pulmonary arterial wall, manifested by dysregulated genes implicated in elastin degradation, precedes the onset of pulmonary hypertension. The resulting degradation of elastic fibers is paralleled by an accumulation of fibrillar collagens. Pentagalloyl glucose preserves arterial elastic fibers from elastolysis, reduces inflammation and collagen accumulation, improves pulmonary artery biomechanics, and normalizes right ventricular and pulmonary hemodynamics in a rat model of pulmonary hypertension due to left heart disease. Thus, targeting extracellular matrix remodeling may present a therapeutic approach for pulmonary hypertension due to left heart disease.
Collapse
Affiliation(s)
- Mariya M Kucherenko
- Department of Cardiothoracic and Vascular Surgery, Deutsches Herzzentrum der Charité (DHZC), Augustenburger Platz 1, 13353, Berlin, Germany
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Germany, Charitéplatz 1, 10117, Berlin, Germany
- Institute of Physiology, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Charitéplatz 1, 10117, Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), partner site Berlin, Germany
| | - Pengchao Sang
- Department of Cardiothoracic and Vascular Surgery, Deutsches Herzzentrum der Charité (DHZC), Augustenburger Platz 1, 13353, Berlin, Germany
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Germany, Charitéplatz 1, 10117, Berlin, Germany
- Institute of Physiology, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Charitéplatz 1, 10117, Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), partner site Berlin, Germany
| | - Juquan Yao
- Department of Cardiothoracic and Vascular Surgery, Deutsches Herzzentrum der Charité (DHZC), Augustenburger Platz 1, 13353, Berlin, Germany
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Germany, Charitéplatz 1, 10117, Berlin, Germany
- Institute of Physiology, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Charitéplatz 1, 10117, Berlin, Germany
| | - Tara Gransar
- Department of Cardiothoracic and Vascular Surgery, Deutsches Herzzentrum der Charité (DHZC), Augustenburger Platz 1, 13353, Berlin, Germany
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Germany, Charitéplatz 1, 10117, Berlin, Germany
- Institute of Physiology, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Charitéplatz 1, 10117, Berlin, Germany
| | - Saphala Dhital
- Department of Bioengineering, Clemson University, 29634, Clemson, SC, USA
| | - Jana Grune
- Department of Cardiothoracic and Vascular Surgery, Deutsches Herzzentrum der Charité (DHZC), Augustenburger Platz 1, 13353, Berlin, Germany
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Germany, Charitéplatz 1, 10117, Berlin, Germany
- Institute of Physiology, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Charitéplatz 1, 10117, Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), partner site Berlin, Germany
| | - Szandor Simmons
- Institute of Physiology, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Charitéplatz 1, 10117, Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), partner site Berlin, Germany
| | - Laura Michalick
- Institute of Physiology, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Charitéplatz 1, 10117, Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), partner site Berlin, Germany
| | - Dag Wulsten
- Julius Wolff Institute for Biomechanics and Musculoskeletal Regeneration, Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Augustenburger Platz 1, 13353, Berlin, Germany
| | - Mario Thiele
- Julius Wolff Institute for Biomechanics and Musculoskeletal Regeneration, Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Augustenburger Platz 1, 13353, Berlin, Germany
| | - Orr Shomroni
- NGS Integrative Genomics (NIG), Justus-von-Liebig-Weg 11, 37077, Göttingen, Germany
| | - Felix Hennig
- Department of Cardiothoracic and Vascular Surgery, Deutsches Herzzentrum der Charité (DHZC), Augustenburger Platz 1, 13353, Berlin, Germany
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Germany, Charitéplatz 1, 10117, Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), partner site Berlin, Germany
| | - Ruhi Yeter
- Department of Cardiothoracic and Vascular Surgery, Deutsches Herzzentrum der Charité (DHZC), Augustenburger Platz 1, 13353, Berlin, Germany
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Germany, Charitéplatz 1, 10117, Berlin, Germany
| | - Natalia Solowjowa
- Department of Cardiothoracic and Vascular Surgery, Deutsches Herzzentrum der Charité (DHZC), Augustenburger Platz 1, 13353, Berlin, Germany
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Germany, Charitéplatz 1, 10117, Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), partner site Berlin, Germany
| | - Gabriela Salinas
- NGS Integrative Genomics (NIG), Justus-von-Liebig-Weg 11, 37077, Göttingen, Germany
| | - Georg N Duda
- Julius Wolff Institute for Biomechanics and Musculoskeletal Regeneration, Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Augustenburger Platz 1, 13353, Berlin, Germany
- Berlin Institute of Health Center for Regenerative Therapies, Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Augustenburger Platz 1, 13353, Berlin, Germany
| | - Volkmar Falk
- Department of Cardiothoracic and Vascular Surgery, Deutsches Herzzentrum der Charité (DHZC), Augustenburger Platz 1, 13353, Berlin, Germany
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Germany, Charitéplatz 1, 10117, Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), partner site Berlin, Germany
- Department of Health Science and Technology, Translational Cardiovascular Technology, LFW C 13.2, ETH Zurich, Universitätstrasse 2, 8092, Zürich, Switzerland
| | - Naren R Vyavahare
- Department of Bioengineering, Clemson University, 29634, Clemson, SC, USA
| | - Wolfgang M Kuebler
- Institute of Physiology, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Charitéplatz 1, 10117, Berlin, Germany.
- DZHK (German Centre for Cardiovascular Research), partner site Berlin, Germany.
- Departments of Physiology and Surgery, University of Toronto, 1 King´s College Circle, Toronto, ON M5S 1A8, Canada.
| | - Christoph Knosalla
- Department of Cardiothoracic and Vascular Surgery, Deutsches Herzzentrum der Charité (DHZC), Augustenburger Platz 1, 13353, Berlin, Germany.
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Germany, Charitéplatz 1, 10117, Berlin, Germany.
- DZHK (German Centre for Cardiovascular Research), partner site Berlin, Germany.
| |
Collapse
|
30
|
Manichaikul A, Hu X, Logan J, Kwon Y, Lima J, Jacobs D, Duprez D, Brumback L, Taylor K, Durda P, Johnson C, Cornell E, Guo X, Liu Y, Tracy R, Blackwell T, Papanicolaou G, Mitchell G, Rich S, Rotter J, Van Den Berg D, Chirinos J, Hughes T, Garrett-Bakelman F. Multi-ancestry epigenome-wide analyses identify methylated sites associated with aortic augmentation index in TOPMed MESA. RESEARCH SQUARE 2023:rs.3.rs-3125948. [PMID: 37502922 PMCID: PMC10371087 DOI: 10.21203/rs.3.rs-3125948/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/29/2023]
Abstract
Despite the prognostic value of arterial stiffness (AS) and pulsatile hemodynamics (PH) for cardiovascular morbidity and mortality, epigenetic modifications that contribute to AS/PH remain unknown. To gain a better understanding of the link between epigenetics (DNA methylation) and AS/PH, we examined the relationship of eight measures of AS/PH with CpG sites and co-methylated regions using multi-ancestry participants from Trans-Omics for Precision Medicine (TOPMed) Multi-Ethnic Study of Atherosclerosis (MESA) with sample sizes ranging from 438 to 874. Epigenome-wide association analysis identified one genome-wide significant CpG (cg20711926-CYP1B1) associated with aortic augmentation index (AIx). Follow-up analyses, including gene set enrichment analysis, expression quantitative trait methylation analysis, and functional enrichment analysis on differentially methylated positions and regions, further prioritized three CpGs and their annotated genes (cg23800023-ETS1, cg08426368-TGFB3, and cg17350632-HLA-DPB1) for AIx. Among these, ETS1 and TGFB3 have been previously prioritized as candidate genes. Furthermore, both ETS1 and HLA-DPB1 have significant tissue correlations between Whole Blood and Aorta in GTEx, which suggests ETS1 and HLA-DPB1 could be potential biomarkers in understanding pathophysiology of AS/PH. Overall, our findings support the possible role of epigenetic regulation via DNA methylation of specific genes associated with AIx as well as identifying potential targets for regulation of AS/PH.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Kent Taylor
- The Institute for Translational Genomics and Population Sciences
| | | | | | | | | | | | | | | | | | | | - Stephen Rich
- Center for Public Health Genomics, Department of Public Health Sciences, University of Virginia
| | - Jerome Rotter
- The Institute for Translational Genomics and Population Sciences, Department of Pediatrics, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center
| | | | | | | | | |
Collapse
|
31
|
Brown RB. Salt-Sensitive Hypertension: Mediation by Salt-Induced Hypervolemia and Phosphate-Induced Vascular Calcification. CLINICAL MEDICINE INSIGHTS-CARDIOLOGY 2023; 17:11795468231158206. [PMID: 37434790 PMCID: PMC10331233 DOI: 10.1177/11795468231158206] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Accepted: 02/01/2023] [Indexed: 07/13/2023]
Abstract
Preventing hypertension by restricting dietary salt intake, sodium chloride, is well established in public health policy, but a pathophysiological mechanism has yet to explain the controversial clinical finding that some individuals have a greater risk of hypertension from exposure to salt intake, termed salt-sensitive hypertension. The present perspective paper synthesizes interdisciplinary findings from the research literature and offers novel insights proposing that the pathogenesis of salt-sensitive hypertension is mediated by interaction of salt-induced hypervolemia and phosphate-induced vascular calcification. Arterial stiffness and blood pressure increase as calcification in the vascular media layer reduces arterial elasticity, preventing arteries from expanding to accommodate extracellular fluid overload in hypervolemia related to salt intake. Furthermore, phosphate has been found to be a direct inducer of vascular calcification. Reduction of dietary phosphate may help reduce salt-sensitive hypertension by lowering the prevalence and progression of vascular calcification. Further research should investigate the correlation of vascular calcification with salt-sensitive hypertension, and public health recommendations to prevent hypertension should encourage reductions of both sodium-induced hypervolemia and phosphate-induced vascular calcification.
Collapse
Affiliation(s)
- Ronald B Brown
- School of Public Health Sciences, University of Waterloo, Waterloo, ON, Canada
| |
Collapse
|
32
|
Subramanian Balachandar VA, Steward RL. Extracellular matrix composition alters endothelial force transmission. Am J Physiol Cell Physiol 2023; 325:C314-C323. [PMID: 37335028 PMCID: PMC10393341 DOI: 10.1152/ajpcell.00106.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Revised: 06/02/2023] [Accepted: 06/09/2023] [Indexed: 06/21/2023]
Abstract
Extracellular matrix (ECM) composition is important in a host of pathophysiological processes such as angiogenesis, atherosclerosis, and diabetes, and during each of these processes ECM composition has been reported to change over time. However, the impact ECM composition has on the ability of endothelium to respond mechanically is currently unknown. Therefore, in this study, we seeded human umbilical vein endothelial cells (HUVECs) onto soft hydrogels coated with an ECM concentration of 0.1 mg/mL at the following collagen I (Col-I) and fibronectin (FN) ratios: 100% Col-I, 75% Col-I-25% FN, 50% Col-I-50% FN, 25% Col-I-75% FN, and 100% FN. We subsequently measured tractions, intercellular stresses, strain energy, cell morphology, and cell velocity. Our results revealed that tractions and strain energy are maximal at 50% Col-I-50% FN and minimal at 100% Col-I and 100% FN. Intercellular stress response was maximal on 50% Col-I-50% FN and minimal on 25% Col-I-75% FN. Cell area and cell circularity displayed a divergent relationship for different Col-I and FN ratios. We believe that these results will be of great importance to the cardiovascular field, biomedical field, and cell mechanics.NEW & NOTEWORTHY The endothelium constitutes the innermost layer of all blood vessels and plays an important role in vascular physiology and pathology. During certain vascular diseases, the extracellular matrix has been suggested to transition from a collagen-rich matrix to a fibronectin-rich matrix. In this study, we demonstrate the impact various collagen and fibronectin ratios have on endothelial biomechanical and morphological response.
Collapse
Affiliation(s)
- Vignesh Aravind Subramanian Balachandar
- Department of Mechanical and Aerospace Engineering, College of Engineering and Computer Science, University of Central Florida, Orlando, Florida, United States
- Department of Cell Biology, University of Virginia, Charlottesville, Virginia, United States
| | - Robert L Steward
- Department of Mechanical and Aerospace Engineering, College of Engineering and Computer Science, University of Central Florida, Orlando, Florida, United States
- Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, Florida, United States
| |
Collapse
|
33
|
Rovas G, Bikia V, Stergiopulos N. Design and computational optimization of compliance-matching aortic grafts. Front Bioeng Biotechnol 2023; 11:1179174. [PMID: 37456727 PMCID: PMC10341153 DOI: 10.3389/fbioe.2023.1179174] [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: 03/03/2023] [Accepted: 06/19/2023] [Indexed: 07/18/2023] Open
Abstract
Introduction: Synthetic vascular grafts have been widely used in clinical practice for aortic replacement surgery. Despite their high rates of surgical success, they remain significantly less compliant than the native aorta, resulting in a phenomenon called compliance mismatch. This incompatibility of elastic properties may cause serious post-operative complications, including hypertension and myocardial hypertrophy. Methods: To mitigate the risk for these complications, we designed a multi-layer compliance-matching stent-graft, that we optimized computationally using finite element analysis, and subsequently evaluated in vitro. Results: We found that our compliance-matching grafts attained the distensibility of healthy human aortas, including those of young adults, thereby significantly exceeding the distensibility of gold-standard grafts. The compliant grafts maintained their properties in a wide range of conditions that are expected after the implantation. Furthermore, the computational model predicted the graft radius with enough accuracy to allow computational optimization to be performed effectively. Conclusion: Compliance-matching grafts may offer a valuable improvement over existing prostheses and they could potentially mitigate the risk for post-operative complications attributed to excessive graft stiffness.
Collapse
|
34
|
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
|
35
|
Sigaeva T, Zhang Y. A novel constitutive model considering the role of elastic lamellae' structural heterogeneity in homogenizing transmural stress distribution in arteries. J R Soc Interface 2023; 20:20220837. [PMID: 37042193 PMCID: PMC10091191 DOI: 10.1098/rsif.2022.0837] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2022] [Accepted: 03/21/2023] [Indexed: 04/13/2023] Open
Abstract
Understanding how the homeostatic stress state can be reached in arterial tissues can provide new insights into vascular physiology. Even though the function of maintaining homeostasis is often linked to the concentric layers of medial elastic lamellae, how the lamellae are capable of evenly distributing the stress transmurally remains to be understood. The recent microstructural study by Yu et al. (2018 J. R. Soc. Interface 15, 20180492) revealed that, circumferentially, lamellar layers closer to the lumen are wavier than the ones further away from it and, thus, experience more unfolding when subjected to blood pressure. Motivated by this peculiar finding, the current study, for the first time, proposes a novel approach to model elastic lamellae and such structural heterogeneity using the extensible worm-like chain model. When implemented into the material description of the conventional two-layer artery model, in which adventitial collagen is modelled using the inextensible worm-like chain model, it is demonstrated that structural heterogeneity in elastic lamellae plays an important role in dictating transmural stress distribution and, therefore, the homeostasis of the arterial wall.
Collapse
Affiliation(s)
- Taisiya Sigaeva
- Department of Systems Design Engineering, University of Waterloo, Waterloo, Ontario, Canada N2L 3G1
| | - Yanhang Zhang
- Department of Mechanical Engineering, Boston University, Boston, MA 02215, USA
- Department of Biomedical Engineering, Boston University, Boston, MA 02215, USA
| |
Collapse
|
36
|
Smith KA, Lin AH, Stevens AH, Yu SM, Weiss JA, Timmins LH. Collagen Molecular Damage is a Hallmark of Early Atherosclerosis Development. J Cardiovasc Transl Res 2023; 16:463-472. [PMID: 36097314 DOI: 10.1007/s12265-022-10316-y] [Citation(s) in RCA: 3] [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: 05/04/2022] [Accepted: 08/30/2022] [Indexed: 10/14/2022]
Abstract
Remodeling of extracellular matrix proteins underlies the development of cardiovascular disease. Herein, we utilized a novel molecular probe, collagen hybridizing peptide (CHP), to target collagen molecular damage during atherogenesis. The thoracic aorta was dissected from ApoE-/- mice that had been on a high-fat diet for 0-18 weeks. Using an optimized protocol, tissues were stained with Cy3-CHP and digested to quantify CHP with a microplate assay. Results demonstrated collagen molecular damage, inferred from Cy3-CHP fluorescence, was a function of location and time on the high-fat diet. Tissue from the aortic arch showed a significant increase in collagen molecular damage after 18 weeks, while no change was observed in tissue from the descending aorta. No spatial differences in fluorescence were observed between the superior and inferior arch tissue. Our results provide insight into the early changes in collagen during atherogenesis and present a new opportunity in the subclinical diagnosis of atherosclerosis.
Collapse
Affiliation(s)
- Kelly A Smith
- Department of Biomedical Engineering, University of Utah, Salt Lake City, UT, 84112, USA
| | - Allen H Lin
- Department of Biomedical Engineering, University of Utah, Salt Lake City, UT, 84112, USA
- Scientific Computing and Imaging Institute, University of Utah, Salt Lake City, UT, 84112, USA
| | - Alexander H Stevens
- Department of Biomedical Engineering, University of Utah, Salt Lake City, UT, 84112, USA
| | - S Michael Yu
- Department of Biomedical Engineering, University of Utah, Salt Lake City, UT, 84112, USA
- Department of Molecular Pharmaceutics, University of Utah, Salt Lake City, UT, 84112, USA
| | - Jeffrey A Weiss
- Department of Biomedical Engineering, University of Utah, Salt Lake City, UT, 84112, USA
- Scientific Computing and Imaging Institute, University of Utah, Salt Lake City, UT, 84112, USA
- Department of Orthopaedics, University of Utah, Salt Lake City, UT, 84112, USA
| | - Lucas H Timmins
- Department of Biomedical Engineering, University of Utah, Salt Lake City, UT, 84112, USA.
- Scientific Computing and Imaging Institute, University of Utah, Salt Lake City, UT, 84112, USA.
| |
Collapse
|
37
|
Gueldner PH, Marini AX, Li B, Darvish CJ, Chung TK, Weinbaum JS, Curci JA, Vorp DA. Mechanical and matrix effects of short and long-duration exposure to beta-aminopropionitrile in elastase-induced model abdominal aortic aneurysm in mice. JVS Vasc Sci 2023; 4:100098. [PMID: 37152846 PMCID: PMC10160690 DOI: 10.1016/j.jvssci.2023.100098] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Accepted: 12/23/2022] [Indexed: 02/19/2023] Open
Abstract
Objective Evaluate the mechanical and matrix effects on abdominal aortic aneurysms (AAA) during the initial aortic dilation and after prolonged exposure to beta-aminopropionitrile (BAPN) in a topical elastase AAA model. Methods Abdominal aortae of C57/BL6 mice were exposed to topical elastase with or without BAPN in the drinking water starting 4 days before elastase exposure. For the standard AAA model, animals were harvested at 2 weeks after active elastase (STD2) or heat-inactivated elastase (SHAM2). For the enhanced elastase model, BAPN treatment continued for either 4 days (ENH2b) or until harvest (ENH2) at 2 weeks; BAPN was continued until harvest at 8 weeks in one group (ENH8). Each group underwent assessment of aortic diameter, mechanical testing (tangent modulus and ultimate tensile strength [UTS]), and quantification of insoluble elastin and bulk collagen in both the elastase exposed aorta as well as the descending thoracic aorta. Results BAPN treatment did not increase aortic dilation compared with the standard model after 2 weeks (ENH2, 1.65 ± 0.23 mm; ENH2b, 1.49 ± 0.39 mm; STD2, 1.67 ± 0.29 mm; and SHAM2, 0.73 ± 0.10 mm), but did result in increased dilation after 8 weeks (4.3 ± 2.0 mm; P = .005). After 2 weeks, compared with the standard model, continuous therapy with BAPN did not have an effect on UTS (24.84 ± 7.62 N/cm2; 18.05 ± 4.95 N/cm2), tangent modulus (32.60 ± 9.83 N/cm2; 26.13 ± 9.10 N/cm2), elastin (7.41 ± 2.43%; 7.37 ± 4.00%), or collagen (4.25 ± 0.79%; 5.86 ± 1.19%) content. The brief treatment, EHN2b, resulted in increased aortic collagen content compared with STD2 (7.55 ± 2.48%; P = .006) and an increase in UTS compared with ENH2 (35.18 ± 18.60 N/cm2; P = .03). The ENH8 group had the lowest tangent modulus (3.71 ± 3.10 N/cm2; P = .005) compared with all aortas harvested at 2 weeks and a lower UTS (2.18 ± 2.18 N/cm2) compared with both the STD2 (24.84 ± 7.62 N/cm2; P = .008) and ENH2b (35.18 ± 18.60 N/cm2; P = .001) groups. No differences in the mechanical properties or matrix protein concentrations were associated with abdominal elastase exposure or BAPN treatment for the thoracic aorta. The tangent modulus was higher in the STD2 group (32.60 ± 9.83 N/cm2; P = .0456) vs the SHAM2 group (17.99 ± 5.76 N/cm2), and the UTS was lower in the ENH2 group (18.05 ± 4.95 N/cm2; P = .0292) compared with the ENH2b group (35.18 ± 18.60 N/cm2). The ENH8 group had the lowest tangent modulus (3.71 ± 3.10 N/cm2; P = .005) compared with all aortas harvested at 2 weeks and a lower UTS (2.18 ± 2.18 N/cm2) compared with both the STD2 (24.84 ± 7.62 N/cm2; P = .008) and ENH2b (35.18 ± 18.60 N/cm2; P = .001) groups. Abdominal aortic elastin in the STD2 group (7.41 ± 2.43%; P = .035) was lower compared with the SHAM2 group (15.29 ± 7.66%). Aortic collagen was lower in the STD2 group (4.25 ± 0.79%; P = .007) compared with the SHAM2 group (12.44 ± 6.02%) and higher for the ENH2b (7.55 ± 2.48%; P = .006) compared with the STD2 group. Conclusions Enhancing an elastase AAA model with BAPN does not affect the initial (2-week) dilation phase substantially, either mechanically or by altering the matrix content. Late mechanical and matrix effects of prolonged BAPN treatment are limited to the elastase-exposed segment of the aorta. Clinical Relevance This paper explores the use of short- and long-term exposure to beta-aminopropionitrile to create an enhanced topical elastase abdominal aortic aneurysm model in mice. Readouts of aneurysm severity included loss of mechanical stability and vascular extracellular matrix composition reminiscent of what is seen in the course of human disease. Additionally, we show that the thoracic aorta, unlike the findings below the renal arteries, is not damaged in our animal model.
Collapse
Affiliation(s)
- Pete H. Gueldner
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA
| | - Ande X. Marini
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA
| | - Bo Li
- Department of Vascular Surgery, Vanderbilt University, Nashville, TN
| | - Cyrus J. Darvish
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA
| | - Timothy K. Chung
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA
| | - Justin S. Weinbaum
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA
- Department of Pathology, University of Pittsburgh, Pittsburgh, PA
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA
| | - John A. Curci
- Department of Vascular Surgery, Vanderbilt University, Nashville, TN
| | - David A. Vorp
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA
- Department of Surgery, University of Pittsburgh, Pittsburgh, PA
- Department of Chemical and Petroleum Engineering, University of Pittsburgh, Pittsburgh, PA
- Department of Mechanical Engineering and Materials Science, University of Pittsburgh, Pittsburgh, PA
- Department of Cardiothoracic Surgery, University of Pittsburgh, Pittsburgh, PA
- Clinical & Translational Sciences Institute, University of Pittsburgh, Pittsburgh, PA
- Center for Vascular Remodeling and Regeneration, University of Pittsburgh, Pittsburgh, PA
| |
Collapse
|
38
|
What did we learn from the International Databases on Ambulatory and Home Blood Pressure in Relation to Cardiovascular Outcome? Hypertens Res 2023; 46:934-949. [PMID: 36737461 PMCID: PMC10073019 DOI: 10.1038/s41440-023-01191-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Revised: 01/08/2023] [Accepted: 01/09/2023] [Indexed: 02/05/2023]
Abstract
To assess in individual-person meta-analyses how out-of-office blood pressure (BP) contributes to risk stratification and the management of hypertension, an international consortium set up the International Databases on Ambulatory (IDACO) and Home (IDHOCO) Blood Pressure in Relation to Cardiovascular Outcome. This review summarizes key findings of recent IDACO/IDHOCO articles. Among various BP indexes derived from office and ambulatory BP recordings, the 24-h and nighttime BP level were the best predictors of adverse health outcomes. Second, using the 10-year cardiovascular risk associated with guideline-endorsed office BP thresholds as reference, corresponding thresholds were derived for home and ambulatory BP. Stratified by the underlying cardiovascular risk, the rate of cardiovascular events in white-coat hypertensive patients and matched normotensive controls were not substantially different. The observation that masked hypertension carries a high cardiovascular risk was replicated in Nigerian Blacks, using home BP monitoring. The thresholds for 24-h mean arterial pressure, i.e., the BP component measured by oscillometric devices, delineating normotension, elevated BP and hypertension were <90, 90 to 92 and ≥92 mmHg. At young age, the absolute risk associated with out-of-office BP was low, but the relative risk was high, whereas with advancing age, the relative risk decreased and the absolute risk increased. Using pulse pressure as an exemplary case, the relative risks of death, cardiovascular endpoints and stroke decreased over 3-fold from 55 to 75 years of age, whereas in contrast absolute risk rose 3-fold. In conclusion, IDACO/IDHOCO forcefully support the notion that the pressing need to curb the hypertension pandemic cannot be met without out-of-the-office BP monitoring.
Collapse
|
39
|
Mazzuca MQ, Buyukcelebi K, Lin C, Khalil RA. Increased Ca 2+-dependent intrinsic tone and arterial stiffness in mesenteric microvessels of hypertensive pregnant rats. Biochem Pharmacol 2023; 208:115353. [PMID: 36435203 PMCID: PMC9877182 DOI: 10.1016/j.bcp.2022.115353] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Revised: 11/17/2022] [Accepted: 11/18/2022] [Indexed: 11/26/2022]
Abstract
Preeclampsia is a pregnancy-related hypertensive disorder (HTN-Preg) with unclear mechanisms. We have shown increased vascular reactivity to extrinsic vasoconstrictors in HTN-Preg rats. Here, we test whether microvascular intrinsic tone and arterial stiffness could contribute to HTN-Preg, and examined the underlying cellular mechanisms. On gestational day 19, BP was recorded in normal pregnant (Preg) rats and Preg rats with reduced uterine perfusion pressure (RUPP), and mesenteric microvessels were mounted on a pressure myograph for measurement of intrinsic tone, simultaneous changes in [Ca2+]i (fura-2 340/380 ratio), and arterial stiffness. Arteries were incubated in Ca2+-containing and 0 Ca2+ (2 mM EGTA) Krebs, pressurized at 10 to 110 mmHg in 10 mmHg increments, and the % change in vessel diameter from initial diameter at 10 mmHg was analyzed for measurement of total (active + passive) intrinsic tone and passive intrinsic response, respectively. The passive response was then subtracted from the total intrinsic tone to determine the active myogenic tone. The strain-stress relationship was also constructed as a measure of arterial stiffness. BP was higher in RUPP vs Preg rats. In Ca2+-containing Krebs, increases in intraluminal pressure caused smaller increases in diameter and greater increases in [Ca2+]i in microvessels of RUPP vs Preg rats, suggesting increased Ca2+-dependent myogenic tone. In 0 Ca2+ Krebs, increases in pressure also caused less increases in diameter in microvessels of RUPP vs Preg rats, but with no changes in [Ca2+]i, suggesting changes in the structure and mechanics of the arterial wall. The total and passive strain-stress relationship was shifted to the left in microvessels of RUPP vs Preg rats, suggesting increased arterial wall stiffness. Histology and immunohistochemistry showed greater vascular wall thickness and collagen-I staining in RUPP vs Preg rats, supporting changes in the wall architecture and structural proteins. The increased active myogenic tone and underlying increases in Ca2+ signaling as well as the increased passive intrinsic response, arterial stiffness and collagen-I in the mesenteric microvessels could play a role in the regulation of blood flow to the splanchnic region and the increased vascular resistance and BP in HTN-Preg.
Collapse
Affiliation(s)
- Marc Q Mazzuca
- Vascular Surgery Research Laboratories, Division of Vascular and Endovascular Surgery, Brigham and Women's Hospital, and Harvard Medical School, Boston, MA, United States
| | - Kadir Buyukcelebi
- Vascular Surgery Research Laboratories, Division of Vascular and Endovascular Surgery, Brigham and Women's Hospital, and Harvard Medical School, Boston, MA, United States
| | - Chen Lin
- Vascular Surgery Research Laboratories, Division of Vascular and Endovascular Surgery, Brigham and Women's Hospital, and Harvard Medical School, Boston, MA, United States
| | - Raouf A Khalil
- Vascular Surgery Research Laboratories, Division of Vascular and Endovascular Surgery, Brigham and Women's Hospital, and Harvard Medical School, Boston, MA, United States.
| |
Collapse
|
40
|
Cuevas RA, Wong R, Joolharzadeh P, Moorhead WJ, Chu CC, Callahan J, Crane A, Boufford CK, Parise AM, Parwal A, Behzadi P, St Hilaire C. Ecto-5'-nucleotidase (Nt5e/CD73)-mediated adenosine signaling attenuates TGFβ-2 induced elastin and cellular contraction. Am J Physiol Cell Physiol 2023; 324:C327-C338. [PMID: 36503240 PMCID: PMC9902218 DOI: 10.1152/ajpcell.00054.2022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Revised: 11/21/2022] [Accepted: 12/05/2022] [Indexed: 12/15/2022]
Abstract
Arterial calcification due to deficiency of CD73 (ACDC) is a rare genetic disease caused by a loss-of-function mutation in the NT5E gene encoding the ecto-5'-nucleotidase (cluster of differentiation 73, CD73) enzyme. Patients with ACDC develop vessel arteriomegaly, tortuosity, and vascular calcification in their lower extremity arteries. Histological analysis shows that patients with ACDC vessels exhibit fragmented elastin fibers similar to that seen in aneurysmal-like pathologies. It is known that alterations in transforming growth factor β (TGFβ) pathway signaling contribute to this elastin phenotype in several connective tissue diseases, as TGFβ regulates extracellular matrix (ECM) remodeling. Our study investigates whether CD73-derived adenosine modifies TGFβ signaling in vascular smooth muscle cells (SMCs). We show that Nt5e-/- SMCs have elevated contractile markers and elastin gene expression compared with Nt5e+/+ SMCs. Ecto-5'-nucleotidase (Nt5e)-deficient SMCs exhibit increased TGFβ-2 and activation of small mothers against decapentaplegic (SMAD) signaling, elevated elastin transcript and protein, and potentiate SMC contraction. These effects were diminished when the A2b adenosine receptor was activated. Our results identify a novel link between adenosine and TGFβ signaling, where adenosine signaling via the A2b adenosine receptor attenuates TGFβ signaling to regulate SMC homeostasis. We discuss how disruption in adenosine signaling is implicated in ACDC vessel tortuosity and could potentially contribute to other aneurysmal pathogenesis.
Collapse
Affiliation(s)
- Rolando A Cuevas
- Division of Cardiology, Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
- Pittsburgh Heart, Lung, and Blood Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Ryan Wong
- Division of Cardiology, Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
- Pittsburgh Heart, Lung, and Blood Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Pouya Joolharzadeh
- Division of Cardiology, Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
- Pittsburgh Heart, Lung, and Blood Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - William J Moorhead
- Division of Cardiology, Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
- Pittsburgh Heart, Lung, and Blood Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Claire C Chu
- Division of Cardiology, Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
- Pittsburgh Heart, Lung, and Blood Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Jack Callahan
- Division of Cardiology, Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
- Pittsburgh Heart, Lung, and Blood Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Alex Crane
- Division of Cardiology, Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
- Pittsburgh Heart, Lung, and Blood Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Camille K Boufford
- Division of Cardiology, Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
- Pittsburgh Heart, Lung, and Blood Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Angelina M Parise
- Division of Cardiology, Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
- Pittsburgh Heart, Lung, and Blood Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Aneesha Parwal
- Division of Cardiology, Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
- Pittsburgh Heart, Lung, and Blood Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Parya Behzadi
- Division of Cardiology, Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
- Pittsburgh Heart, Lung, and Blood Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Cynthia St Hilaire
- Division of Cardiology, Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
- Pittsburgh Heart, Lung, and Blood Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, Pennsylvania
- Department of Bioengineering, Swanson School of Engineering, University of Pittsburgh, Pittsburgh, Pennsylvania
| |
Collapse
|
41
|
Coleman-Belin J, Harris A, Chen B, Zhou J, Ciulla T, Verticchio A, Antman G, Chang M, Siesky B. Aging Effects on Optic Nerve Neurodegeneration. Int J Mol Sci 2023; 24:2573. [PMID: 36768896 PMCID: PMC9917079 DOI: 10.3390/ijms24032573] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Revised: 01/19/2023] [Accepted: 01/23/2023] [Indexed: 02/03/2023] Open
Abstract
Common risk factors for many ocular pathologies involve non-pathologic, age-related damage to the optic nerve. Understanding the mechanisms of age-related changes can facilitate targeted treatments for ocular pathologies that arise at any point in life. In this review, we examine these age-related, neurodegenerative changes in the optic nerve, contextualize these changes from the anatomic to the molecular level, and appreciate their relationship with ocular pathophysiology. From simple structural and mechanical changes at the optic nerve head (ONH), to epigenetic and biochemical alterations of tissue and the environment, multiple age-dependent mechanisms drive extracellular matrix (ECM) remodeling, retinal ganglion cell (RGC) loss, and lowered regenerative ability of respective axons. In conjunction, aging decreases the ability of myelin to preserve maximal conductivity, even with "successfully" regenerated axons. Glial cells, however, regeneratively overcompensate and result in a microenvironment that promotes RGC axonal death. Better elucidating optic nerve neurodegeneration remains of interest, specifically investigating human ECM, RGCs, axons, oligodendrocytes, and astrocytes; clarifying the exact processes of aged ocular connective tissue alterations and their ultrastructural impacts; and developing novel technologies and pharmacotherapies that target known genetic, biochemical, matrisome, and neuroinflammatory markers. Management models should account for age-related changes when addressing glaucoma, diabetic retinopathy, and other blinding diseases.
Collapse
Affiliation(s)
- Janet Coleman-Belin
- Department of Ophthalmology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Alon Harris
- Department of Ophthalmology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Bo Chen
- Department of Ophthalmology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Jing Zhou
- Department of Ophthalmology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Thomas Ciulla
- Vitreoretinal Medicine and Surgery, Midwest Eye Institute, Indianapolis, IN 46290, USA
| | - Alice Verticchio
- Department of Ophthalmology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Gal Antman
- Department of Ophthalmology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Department of Ophthalmology, Rabin Medical Center, Petah Tikva 4941492, Israel
| | - Michael Chang
- Department of Ophthalmology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Brent Siesky
- Department of Ophthalmology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| |
Collapse
|
42
|
SubramanianBalachandar V, Steward RL. Extracellular Matrix Composition Alters Endothelial Force Transmission. RESEARCH SQUARE 2023:rs.3.rs-2499973. [PMID: 36747754 PMCID: PMC9900979 DOI: 10.21203/rs.3.rs-2499973/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
ECM composition is important in a host of pathophysiological processes such as angiogenesis, atherosclerosis, and diabetes, for example and during each of these processes ECM composition has been reported to change over time. However, the impact ECM composition has on the endothelium’s ability to respond mechanically is currently unknown. Therefore, in this study we seeded human umbilical vein endothelial cells (HUVECs) onto soft hydrogels coated with an ECM concentration of 0.1 mg/mL at the following collagen I (Col-I) and fibronectin (FN) ratios: 100%Col-I, 75%Col-I-25%FN, 50%Col-I-50%FN, 25%Col-I-75%FN, and 100%FN. We subsequently measured tractions, intercellular stresses, strain energy, cell morphology, and cell velocity. Our results revealed huvecs seeded on gels coated with 50% Col-I - 50% FN to have the highest intercellular stresses, tractions, strain energies, but the lowest velocities and cell circularity. Huvecs seeded on 100% Col-I had the lowest tractions, cell area while havingthe highest velocities and cell circularity. In addition, cells cultured on 25% Col-I and 75% FN had the lowest intercellular stresses, but the highest cell area. Huvecs cultured on 100% FN yielded the lowest strain energies. We believe these results will be of great importance to the cardiovascular field, biomedical field, and cell mechanics. Summary: Study the influence of different Col-I - FN ECM compositions on endothelial cell mechanics and morphology.
Collapse
|
43
|
Hume RD, Kanagalingam S, Deshmukh T, Chen S, Mithieux SM, Rashid FN, Roohani I, Lu J, Doan T, Graham D, Clayton ZE, Slaughter E, Kizana E, Stempien-Otero AS, Brown P, Thomas L, Weiss AS, Chong JJ. Tropoelastin Improves Post-Infarct Cardiac Function. Circ Res 2023; 132:72-86. [PMID: 36453283 PMCID: PMC9829044 DOI: 10.1161/circresaha.122.321123] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/03/2022]
Abstract
BACKGROUND Myocardial infarction (MI) is among the leading causes of death worldwide. Following MI, necrotic cardiomyocytes are replaced by a stiff collagen-rich scar. Compared to collagen, the extracellular matrix protein elastin has high elasticity and may have more favorable properties within the cardiac scar. We sought to improve post-MI healing by introducing tropoelastin, the soluble subunit of elastin, to alter scar mechanics early after MI. METHODS AND RESULTS We developed an ultrasound-guided direct intramyocardial injection method to administer tropoelastin directly into the left ventricular anterior wall of rats subjected to induced MI. Experimental groups included shams and infarcted rats injected with either PBS vehicle control or tropoelastin. Compared to vehicle treated controls, echocardiography assessments showed tropoelastin significantly improved left ventricular ejection fraction (64.7±4.4% versus 46.0±3.1% control) and reduced left ventricular dyssynchrony (11.4±3.5 ms versus 31.1±5.8 ms control) 28 days post-MI. Additionally, tropoelastin reduced post-MI scar size (8.9±1.5% versus 20.9±2.7% control) and increased scar elastin (22±5.8% versus 6.2±1.5% control) as determined by histological assessments. RNA sequencing (RNAseq) analyses of rat infarcts showed that tropoelastin injection increased genes associated with elastic fiber formation 7 days post-MI and reduced genes associated with immune response 11 days post-MI. To show translational relevance, we performed immunohistochemical analyses on human ischemic heart disease cardiac samples and showed an increase in tropoelastin within fibrotic areas. Using RNA-seq we also demonstrated the tropoelastin gene ELN is upregulated in human ischemic heart disease and during human cardiac fibroblast-myofibroblast differentiation. Furthermore, we showed by immunocytochemistry that human cardiac fibroblast synthesize increased elastin in direct response to tropoelastin treatment. CONCLUSIONS We demonstrate for the first time that purified human tropoelastin can significantly repair the infarcted heart in a rodent model of MI and that human cardiac fibroblast synthesize elastin. Since human cardiac fibroblasts are primarily responsible for post-MI scar synthesis, our findings suggest exciting future clinical translation options designed to therapeutically manipulate this synthesis.
Collapse
Affiliation(s)
- Robert D. Hume
- Centre for Heart Research, Westmead Institute for Medical Research, NSW, Australia (R.D.H., S.K., T.D., S.C., F.N.R., J.L., Z.E.C., E.K., J.J.H.C.).,Sydney Medical School, University of Sydney, NSW, Australia (R.D.H., T.D., F.R., Z.E.C., E.K., J.J.H.C.)
| | - Shaan Kanagalingam
- Centre for Heart Research, Westmead Institute for Medical Research, NSW, Australia (R.D.H., S.K., T.D., S.C., F.N.R., J.L., Z.E.C., E.K., J.J.H.C.)
| | - Tejas Deshmukh
- Centre for Heart Research, Westmead Institute for Medical Research, NSW, Australia (R.D.H., S.K., T.D., S.C., F.N.R., J.L., Z.E.C., E.K., J.J.H.C.).,Department of Cardiology, Westmead Hospital, NSW, Australia (T.D., J.L., E.K., P.B., L.T., J.J.H.C.).,Sydney Medical School, University of Sydney, NSW, Australia (R.D.H., T.D., F.R., Z.E.C., E.K., J.J.H.C.)
| | - Siqi Chen
- Centre for Heart Research, Westmead Institute for Medical Research, NSW, Australia (R.D.H., S.K., T.D., S.C., F.N.R., J.L., Z.E.C., E.K., J.J.H.C.)
| | - Suzanne M. Mithieux
- Charles Perkins Centre, University of Sydney, NSW, Australia (S.M.M., A.S.W.).,School of Life and Environmental Sciences, University of Sydney, NSW, Australia (S.M.M., A.S.W.)
| | - Fairooj N. Rashid
- Centre for Heart Research, Westmead Institute for Medical Research, NSW, Australia (R.D.H., S.K., T.D., S.C., F.N.R., J.L., Z.E.C., E.K., J.J.H.C.)
| | - Iman Roohani
- School of Biomedical Engineering, University of Sydney, NSW, Australia (I.R.).,School of Chemistry, University of New South Wales, Australia (I.R.)
| | - Juntang Lu
- Centre for Heart Research, Westmead Institute for Medical Research, NSW, Australia (R.D.H., S.K., T.D., S.C., F.N.R., J.L., Z.E.C., E.K., J.J.H.C.).,Department of Cardiology, Westmead Hospital, NSW, Australia (T.D., J.L., E.K., P.B., L.T., J.J.H.C.)
| | - Tram Doan
- Centre for Cancer Research, Westmead Institute for Medical Research, NSW, Australia (T.D.‚ D.G.)
| | - Dinny Graham
- Centre for Cancer Research, Westmead Institute for Medical Research, NSW, Australia (T.D.‚ D.G.).,Westmead Breast Cancer Institute, NSW, Australia (D.G.).,Westmead Clinical School, University of Sydney, NSW, Australia (D.G., L.T.)
| | - Zoe E. Clayton
- Centre for Heart Research, Westmead Institute for Medical Research, NSW, Australia (R.D.H., S.K., T.D., S.C., F.N.R., J.L., Z.E.C., E.K., J.J.H.C.).,Sydney Medical School, University of Sydney, NSW, Australia (R.D.H., T.D., F.R., Z.E.C., E.K., J.J.H.C.)
| | | | - Eddy Kizana
- Centre for Heart Research, Westmead Institute for Medical Research, NSW, Australia (R.D.H., S.K., T.D., S.C., F.N.R., J.L., Z.E.C., E.K., J.J.H.C.).,Department of Cardiology, Westmead Hospital, NSW, Australia (T.D., J.L., E.K., P.B., L.T., J.J.H.C.).,Sydney Medical School, University of Sydney, NSW, Australia (R.D.H., T.D., F.R., Z.E.C., E.K., J.J.H.C.)
| | - April S. Stempien-Otero
- Department of Medicine, Division of Cardiology, University of Washington School of Medicine, Seattle, WA (A.S.S.-O.)
| | - Paula Brown
- Department of Cardiology, Westmead Hospital, NSW, Australia (T.D., J.L., E.K., P.B., L.T., J.J.H.C.)
| | - Liza Thomas
- Department of Cardiology, Westmead Hospital, NSW, Australia (T.D., J.L., E.K., P.B., L.T., J.J.H.C.).,Westmead Clinical School, University of Sydney, NSW, Australia (D.G., L.T.)
| | | | - James J.H. Chong
- Centre for Heart Research, Westmead Institute for Medical Research, NSW, Australia (R.D.H., S.K., T.D., S.C., F.N.R., J.L., Z.E.C., E.K., J.J.H.C.).,Department of Cardiology, Westmead Hospital, NSW, Australia (T.D., J.L., E.K., P.B., L.T., J.J.H.C.).,Sydney Medical School, University of Sydney, NSW, Australia (R.D.H., T.D., F.R., Z.E.C., E.K., J.J.H.C.)
| |
Collapse
|
44
|
Wang K, Wen D, Xu X, Zhao R, Jiang F, Yuan S, Zhang Y, Gao Y, Li Q. Extracellular matrix stiffness-The central cue for skin fibrosis. Front Mol Biosci 2023; 10:1132353. [PMID: 36968277 PMCID: PMC10031116 DOI: 10.3389/fmolb.2023.1132353] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2022] [Accepted: 02/20/2023] [Indexed: 03/29/2023] Open
Abstract
Skin fibrosis is a physiopathological process featuring the excessive deposition of extracellular matrix (ECM), which is the main architecture that provides structural support and constitutes the microenvironment for various cellular behaviors. Recently, increasing interest has been drawn to the relationship between the mechanical properties of the ECM and the initiation and modulation of skin fibrosis, with the engagement of a complex network of signaling pathways, the activation of mechanosensitive proteins, and changes in immunoregulation and metabolism. Simultaneous with the progression of skin fibrosis, the stiffness of ECM increases, which in turn perturbs mechanical and humoral homeostasis to drive cell fate toward an outcome that maintains and enhances the fibrosis process, thus forming a pro-fibrotic "positive feedback loop". In this review, we highlighted the central role of the ECM and its dynamic changes at both the molecular and cellular levels in skin fibrosis. We paid special attention to signaling pathways regulated by mechanical cues in ECM remodeling. We also systematically summarized antifibrotic interventions targeting the ECM, hopefully enlightening new strategies for fibrotic diseases.
Collapse
Affiliation(s)
- Kang Wang
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Department of Burn and Plastic Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Dongsheng Wen
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xuewen Xu
- Department of Burn and Plastic Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Rui Zhao
- West China School of Medicine, Sichuan University, Chengdu, Sichuan, China
| | - Feipeng Jiang
- West China School of Medicine, Sichuan University, Chengdu, Sichuan, China
| | - Shengqin Yuan
- School of Public Administration, Sichuan University, Chengdu, Sichuan, China
| | - Yifan Zhang
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- *Correspondence: Yifan Zhang, ; Ya Gao, ; Qingfeng Li,
| | - Ya Gao
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- *Correspondence: Yifan Zhang, ; Ya Gao, ; Qingfeng Li,
| | - Qingfeng Li
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- *Correspondence: Yifan Zhang, ; Ya Gao, ; Qingfeng Li,
| |
Collapse
|
45
|
Wang X, Chan V, Corridon PR. Acellular Tissue-Engineered Vascular Grafts from Polymers: Methods, Achievements, Characterization, and Challenges. Polymers (Basel) 2022; 14:4825. [PMID: 36432950 PMCID: PMC9695055 DOI: 10.3390/polym14224825] [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: 09/28/2022] [Revised: 11/03/2022] [Accepted: 11/03/2022] [Indexed: 11/11/2022] Open
Abstract
Extensive and permanent damage to the vasculature leading to different pathogenesis calls for developing innovative therapeutics, including drugs, medical devices, and cell therapies. Innovative strategies to engineer bioartificial/biomimetic vessels have been extensively exploited as an effective replacement for vessels that have seriously malfunctioned. However, further studies in polymer chemistry, additive manufacturing, and rapid prototyping are required to generate highly engineered vascular segments that can be effectively integrated into the existing vasculature of patients. One recently developed approach involves designing and fabricating acellular vessel equivalents from novel polymeric materials. This review aims to assess the design criteria, engineering factors, and innovative approaches for the fabrication and characterization of biomimetic macro- and micro-scale vessels. At the same time, the engineering correlation between the physical properties of the polymer and biological functionalities of multiscale acellular vascular segments are thoroughly elucidated. Moreover, several emerging characterization techniques for probing the mechanical properties of tissue-engineered vascular grafts are revealed. Finally, significant challenges to the clinical transformation of the highly promising engineered vessels derived from polymers are identified, and unique perspectives on future research directions are presented.
Collapse
Affiliation(s)
- Xinyu Wang
- Department of Biomedical Engineering and Healthcare Engineering Innovation Center, Khalifa University, Abu Dhabi P.O. Box 127788, United Arab Emirates
- Department of Immunology and Physiology, College of Medicine and Health Sciences, Khalifa University, Abu Dhabi P.O. Box 127788, United Arab Emirates
| | - Vincent Chan
- Department of Biomedical Engineering and Healthcare Engineering Innovation Center, Khalifa University, Abu Dhabi P.O. Box 127788, United Arab Emirates
| | - Peter R. Corridon
- Department of Biomedical Engineering and Healthcare Engineering Innovation Center, Khalifa University, Abu Dhabi P.O. Box 127788, United Arab Emirates
- Department of Immunology and Physiology, College of Medicine and Health Sciences, Khalifa University, Abu Dhabi P.O. Box 127788, United Arab Emirates
- Center for Biotechnology, Khalifa University, Abu Dhabi P.O. Box 127788, United Arab Emirates
| |
Collapse
|
46
|
Gavish B, Bursztyn M, Thijs L, Wei DM, Melgarejo JD, Zhang ZY, Boggia J, Hansen TW, Asayama K, Ohkubo T, Kikuya M, Yang WY, Stolarz-Skrzypek K, Malyutina S, Casiglia E, Lind L, Li Y, Kawecka-Jaszcz K, Filipovský J, Tikhonoff V, Gilis-Malinowska N, Dolan E, Sandoya E, Narkiewicz K, Wang JG, Imai Y, Maestre GE, O’Brien E, Staessen JA. Predictive power of 24-h ambulatory pulse pressure and its components for mortality and cardiovascular outcomes in 11 848 participants recruited from 13 populations. J Hypertens 2022; 40:2245-2255. [PMID: 35950994 PMCID: PMC10366954 DOI: 10.1097/hjh.0000000000003258] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
BACKGROUND The role of pulse pressure (PP) 'widening' at older and younger age as a cardiovascular risk factor is still controversial. Mean PP, as determined from repeated blood pressure (BP) readings, can be expressed as a sum of two components: 'elastic PP' (elPP) and 'stiffening PP' (stPP) associated, respectively, with stiffness at the diastole and its relative change during the systole. We investigated the association of 24-h ambulatory PP, elPP, and stPP ('PP variables') with mortality and composite cardiovascular events in different age classes. METHOD Longitudinal population-based cohort study of adults with baseline observations that included 24-h ambulatory BP. Age classes were age 40 or less, 40-50, 50-60, 60-70, and over 70 years. Co-primary endpoints were total mortality and composite cardiovascular events. The relative risk expressed by hazard ratio per 1SD increase for each of the PP variables was calculated from multivariable-adjusted Cox regression models. RESULTS The 11 848 participants from 13 cohorts (age 53 ± 16 years, 50% men) were followed for up for 13.7 ± 6.7 years. A total of 2946 participants died (18.1 per 1000 person-years) and 2093 experienced a fatal or nonfatal cardiovascular event (12.9 per 1000 person-years). Mean PP, elPP, and stPP were, respectively, 49.7, 43.5, and 6.2 mmHg, and elPP and stPP were uncorrelated ( r = -0.07). At age 50-60 years, all PP variables displayed association with risk for almost all outcomes. From age over 60 years to age over 70 years, hazard ratios of of PP and elPP were similar and decreased gradually but differently for pulse rate lower than or higher than 70 bpm, whereas stPP lacked predictive power in most cases. For age 40 years or less, elPP showed protective power for coronary events, whereas stPP and PP predicted stroke events. Adjusted and unadjusted hazard ratio variations were similar over the entire age range. CONCLUSION This study provides a new basis for associating PP components with outcome and arterial properties in different age groups and at different pulse rates for both old and young age. The similarity between adjusted and unadjusted hazard ratios supports the clinical usefulness of PP components but further studies are needed to assess the prognostic significance of the PP components, especially at the young age.
Collapse
Affiliation(s)
| | - Michael Bursztyn
- Faculty of Medicine Hebrew University, Jerusalem, Hypertension Clinic Hadassah Medical Center Mount-Scopus, Jerusalem and Department of Medicine D, Beilinson Hospital, Petach-Tikva, Israel
| | - Lutgarde Thijs
- Research Unit Hypertension and Cardiovascular Epidemiology, KU Leuven Department of Cardiovascular Sciences, University of Leuven, Leuven, Belgium
| | - Dong-Mei Wei
- Research Unit Hypertension and Cardiovascular Epidemiology, KU Leuven Department of Cardiovascular Sciences, University of Leuven, Leuven, Belgium
| | - Jesus D. Melgarejo
- Research Unit Hypertension and Cardiovascular Epidemiology, KU Leuven Department of Cardiovascular Sciences, University of Leuven, Leuven, Belgium
- Laboratory of Neurosciences, Faculty of Medicine, University of Zulia, Maracaibo, Zulia, Venezuela
| | - Zhen-Yu Zhang
- Research Unit Hypertension and Cardiovascular Epidemiology, KU Leuven Department of Cardiovascular Sciences, University of Leuven, Leuven, Belgium
| | - Jose Boggia
- Centro de Nefrología and Departamento de Fisiopatología, Hospital de Clínicas, Universidad de la República, Montevideo, Uruguay
| | - Tine W. Hansen
- Steno Diabetes Center Copenhagen, Gentofte and Research Centre for Prevention and Health, Capital Region of Denmark, Denmark
| | - Kei Asayama
- Research Unit Hypertension and Cardiovascular Epidemiology, KU Leuven Department of Cardiovascular Sciences, University of Leuven, Leuven, Belgium
- Department of Hygiene and Public Health, Teikyo University School of Medicine, Tokyo
- Tohoku Institute for Management of Blood Pressure, Sendai, Japan
| | - Takayoshi Ohkubo
- Department of Hygiene and Public Health, Teikyo University School of Medicine, Tokyo
- Tohoku Institute for Management of Blood Pressure, Sendai, Japan
| | - Masahiro Kikuya
- Department of Hygiene and Public Health, Teikyo University School of Medicine, Tokyo
| | - Wen-Yi Yang
- Department of Cardiology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Katarzyna Stolarz-Skrzypek
- First Department of Cardiology, Interventional Electrocardiology and Hypertension, Jagiellonian University Medical College, Kraków, Poland
| | - Sofia Malyutina
- Institute of Internal and Preventive Medicine, Internal and Preventive Medicine - Branch of the Institute of Cytology and Genetics, Siberian Branch of the Russian Academy of Science, Novosibirsk, Russian Federation
| | | | - Lars Lind
- Section of Geriatrics, Department of Public Health and Caring Sciences, Uppsala University, Uppsala, Sweden
| | - Yan Li
- Department of Cardiovascular Medicine, Shanghai Institute of Hypertension, Shanghai Key Laboratory of Hypertension, China
| | - Kalina Kawecka-Jaszcz
- First Department of Cardiology, Interventional Electrocardiology and Hypertension, Jagiellonian University Medical College, Kraków, Poland
| | - Jan Filipovský
- Faculty of Medicine, Charles University, Pilsen, Czech Republic
| | | | | | - Eamon Dolan
- Conway Institute, University College Dublin, Dublin, Ireland
- Stroke and Hypertension Unit, Blanchardstown, Dublin, Ireland
| | - Edgardo Sandoya
- Asociación Española Primera de Socorros Mutuos, Montevideo, Uruguay
| | | | - Ji-Guang Wang
- Department of Cardiovascular Medicine, Shanghai Institute of Hypertension, Shanghai Key Laboratory of Hypertension, China
| | - Yutaka Imai
- Tohoku Institute for Management of Blood Pressure, Sendai, Japan
| | - Gladys E. Maestre
- Department of Neurosciences and Department of Human Genetics, University of Texas Rio Grande Valley School of Medicine, Brownsville, Texas, USA
| | - Eoin O’Brien
- Conway Institute, University College Dublin, Dublin, Ireland
| | - Jan A. Staessen
- Research Institute Alliance for the Promotion of Preventive Medicine, Mechelen
- Biomedical Science Group, University of Leuven, Leuven, Belgium
| | | |
Collapse
|
47
|
Increased Prevalence of Blood Pressure Instability Over Twenty-Four Hours in Chronic Spinal Cord Injury. Neurotrauma Rep 2022; 3:522-533. [DOI: 10.1089/neur.2022.0007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
|
48
|
Liu SF, Nambiar Veetil N, Li Q, Kucherenko MM, Knosalla C, Kuebler WM. Pulmonary hypertension: Linking inflammation and pulmonary arterial stiffening. Front Immunol 2022; 13:959209. [PMID: 36275740 PMCID: PMC9579293 DOI: 10.3389/fimmu.2022.959209] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Accepted: 09/14/2022] [Indexed: 11/13/2022] Open
Abstract
Pulmonary hypertension (PH) is a progressive disease that arises from multiple etiologies and ultimately leads to right heart failure as the predominant cause of morbidity and mortality. In patients, distinct inflammatory responses are a prominent feature in different types of PH, and various immunomodulatory interventions have been shown to modulate disease development and progression in animal models. Specifically, PH-associated inflammation comprises infiltration of both innate and adaptive immune cells into the vascular wall of the pulmonary vasculature—specifically in pulmonary vascular lesions—as well as increased levels of cytokines and chemokines in circulating blood and in the perivascular tissue of pulmonary arteries (PAs). Previous studies suggest that altered hemodynamic forces cause lung endothelial dysfunction and, in turn, adherence of immune cells and release of inflammatory mediators, while the resulting perivascular inflammation, in turn, promotes vascular remodeling and the progression of PH. As such, a vicious cycle of endothelial activation, inflammation, and vascular remodeling may develop and drive the disease process. PA stiffening constitutes an emerging research area in PH, with relevance in PH diagnostics, prognostics, and as a therapeutic target. With respect to its prognostic value, PA stiffness rivals the well-established measurement of pulmonary vascular resistance as a predictor of disease outcome. Vascular remodeling of the arterial extracellular matrix (ECM) as well as vascular calcification, smooth muscle cell stiffening, vascular wall thickening, and tissue fibrosis contribute to PA stiffening. While associations between inflammation and vascular stiffening are well-established in systemic vascular diseases such as atherosclerosis or the vascular manifestations of systemic sclerosis, a similar connection between inflammatory processes and PA stiffening has so far not been addressed in the context of PH. In this review, we discuss potential links between inflammation and PA stiffening with a specific focus on vascular calcification and ECM remodeling in PH.
Collapse
Affiliation(s)
- Shao-Fei Liu
- Institute of Physiology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- German Centre for Cardiovascular Research (DZHK), Berlin, Germany
| | - Netra Nambiar Veetil
- Institute of Physiology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- German Centre for Cardiovascular Research (DZHK), Berlin, Germany
- Department of Cardiothoracic and Vascular Surgery, German Heart Center, Berlin, Germany
| | - Qiuhua Li
- Institute of Physiology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- German Centre for Cardiovascular Research (DZHK), Berlin, Germany
| | - Mariya M. Kucherenko
- Institute of Physiology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- German Centre for Cardiovascular Research (DZHK), Berlin, Germany
- Department of Cardiothoracic and Vascular Surgery, German Heart Center, Berlin, Germany
- *Correspondence: Mariya M. Kucherenko,
| | - Christoph Knosalla
- German Centre for Cardiovascular Research (DZHK), Berlin, Germany
- Department of Cardiothoracic and Vascular Surgery, German Heart Center, Berlin, Germany
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Wolfgang M. Kuebler
- Institute of Physiology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- German Centre for Cardiovascular Research (DZHK), Berlin, Germany
- German Center for Lung Research (DZL), Gießen, Germany
- The Keenan Research Centre for Biomedical Science, St. Michael’s Hospital, Toronto, ON, Canada
- Department of Surgery and Physiology, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
49
|
Sawada H, Ohno-Urabe S, Ye D, Franklin MK, Moorleghen JJ, Howatt DA, Mullick AE, Daugherty A, Lu HS. Inhibition of the Renin-Angiotensin System Fails to Suppress β-Aminopropionitrile-Induced Thoracic Aortopathy in Mice-Brief Report. Arterioscler Thromb Vasc Biol 2022; 42:1254-1261. [PMID: 36004642 PMCID: PMC9492637 DOI: 10.1161/atvbaha.122.317712] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND Cross-linking of lysine residues in elastic and collagen fibers is a vital process in aortic development. Inhibition of lysyl oxidase by BAPN (β-aminopropionitrile) leads to thoracic aortopathies in mice. Although the renin-angiotensin system contributes to several types of thoracic aortopathies, it remains unclear whether inhibition of the renin-angiotensin system protects against aortopathy caused by the impairment of elastic fiber/collagen crosslinking. METHODS BAPN (0.5% wt/vol) was started in drinking water to induce aortopathies in male C57BL/6J mice at 4 weeks of age for 4 weeks. Five approaches were used to investigate the impact of the renin-angiotensin system. Bulk RNA sequencing was performed to explore potential molecular mechanisms of BAPN-induced thoracic aortopathies. RESULTS Losartan increased plasma renin concentrations significantly, compared with vehicle-infused mice, indicating effective angiotensin II type 1 receptor inhibition. However, losartan did not suppress BAPN-induced aortic rupture and dilatation. Since losartan is a surmountable inhibitor of the renin-angiotensin system, irbesartan, an insurmountable inhibitor, was also tested. Although increased plasma renin concentrations indicated effective inhibition, irbesartan did not ameliorate aortic rupture and dilatation in BAPN-administered mice. Thus, BAPN-induced thoracic aortopathies were refractory to angiotensin II type 1 receptor blockade. Next, we inhibited angiotensin II production by pharmacological or genetic depletion of AGT (angiotensinogen), the unique precursor of angiotensin II. However, neither suppressed BAPN-induced thoracic aortic rupture and dilatation. Aortic RNA sequencing revealed molecular changes during BAPN administration that were distinct from other types of aortopathies in which angiotensin II type 1 receptor inhibition protects against aneurysm formation. CONCLUSIONS Inhibition of either angiotensin II action or production of the renin-angiotensin system does not attenuate BAPN-induced thoracic aortopathies in mice.
Collapse
Affiliation(s)
- Hisashi Sawada
- Saha Cardiovascular Research Center, University of Kentucky, Lexington, KY
- Saha Aortic Center, University of Kentucky, Lexington, KY
- Department of Physiology, University of Kentucky, Lexington, KY
| | - Satoko Ohno-Urabe
- Saha Cardiovascular Research Center, University of Kentucky, Lexington, KY
| | - Dien Ye
- Saha Cardiovascular Research Center, University of Kentucky, Lexington, KY
| | - Michael K. Franklin
- Saha Cardiovascular Research Center, University of Kentucky, Lexington, KY
- Saha Aortic Center, University of Kentucky, Lexington, KY
| | - Jessica J. Moorleghen
- Saha Cardiovascular Research Center, University of Kentucky, Lexington, KY
- Saha Aortic Center, University of Kentucky, Lexington, KY
| | - Deborah A. Howatt
- Saha Cardiovascular Research Center, University of Kentucky, Lexington, KY
- Saha Aortic Center, University of Kentucky, Lexington, KY
| | | | - Alan Daugherty
- Saha Cardiovascular Research Center, University of Kentucky, Lexington, KY
- Saha Aortic Center, University of Kentucky, Lexington, KY
- Department of Physiology, University of Kentucky, Lexington, KY
| | - Hong S. Lu
- Saha Cardiovascular Research Center, University of Kentucky, Lexington, KY
- Saha Aortic Center, University of Kentucky, Lexington, KY
- Department of Physiology, University of Kentucky, Lexington, KY
| |
Collapse
|
50
|
Fooladi S, Faramarz S, Dabiri S, Kajbafzadeh A, Nematollahi MH, Mehrabani M. An efficient strategy to recellularization of a rat aorta scaffold: an optimized decellularization, detergent removal, and Apelin-13 immobilization. Biomater Res 2022; 26:46. [PMID: 36138491 PMCID: PMC9502639 DOI: 10.1186/s40824-022-00295-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Accepted: 09/07/2022] [Indexed: 11/21/2022] Open
Abstract
Background Tissue engineering of native vessels is an alternative approach for patients with vascular disease who lack sufficient saphenous vein or other suitable conduits for autologous vascular graft. Moreover, the harvest of vessels prolongs the surgical procedure and it may lead to the morbidity of donor site in elder patients: therefore, it seems that the use of tissue-engineered vessels would be an attractive and less invasive substitute for autologous vascular grafts. Apelin-13 plays a pivotal role in cell proliferation, survival, and attachment; therefore, covalent attachment of apelin-13 to the acellular scaffolds might be a favorable approach for improving recellularization efficacy. Methods In the present study, the decellularization process was performed using various detergents. Afterward, the efficacy of decellularization procedure was evaluated using multiple approaches including assessment of DNA, hydroxyproline, and GAG content as well as Masson’s trichrome and orcein staining used for collagen and elastin determination. Subsequently, the scaffold was bioconjugated with apelin-13 using the EDC-NHS linker and acellular scaffolds were recellularized using fibroblasts, endothelial cells, and smooth muscle cells. SEM images and characterization methods were also used to evaluate the effect of apelin-13 attachment to the acellular scaffold on tissue recellularization. We also developed a novel strategy to eliminate the remnant detergents from the scaffold and increase cell viability by incubating acellular scaffolds with Bio-Beads SM-2 resin. Testometric tensile testing machine was also used for the assessment of mechanical properties and uniaxial tensile strength of decellularized and recellularized vessels compared to that of native tissues. Results Our results proposed 16-h perfusion of 0.25% sodium dodecyl sulfate (SDS) + 0.5% Triton X-100 combination to the vessel as an optimal decellularization protocol in terms of cell elimination as well as extracellular matrix preservation. Furthermore, the results demonstrated considerable elevation of cell adhesion and proliferation in scaffolds bioconjugated with apelin-13. The immunohistochemical (IHC) staining of CD31, α-SMA, and vimentin markers suggested placement of seeded cells in the suitable sites and considerable elevation of cell attachment within the scaffolds bioconjugated with apelin-13 compared to the non-bioconjugated, and decellularized groups. Moreover, the quantitative analysis of IHC staining of CD31, α-SMA, and vimentin markers suggested considerable elevation in the number of endothelial, smooth muscle, and fibroblast cells in the recellularized scaffolds bioconjugated with apelin-13 group (1.4% ± 0.02, 6.66% ± 0.23, and 9.87% ± 0.13%, respectively) compared to the non-bioconjugated scaffolds (0.03% ± 0.01, 0.28% ± 0.01, and 1.2% ± 0.09%, respectively) and decellularized groups (0.03% ± 0.007, 0.05% ± 0.01, and 0.13% ±0.005%, respectively). Although the maximum strain to the rupture was reduced in tissues decellularized using 0.5% SDS and CHAPS compared to that of native ones (116% ± 6.79, 139.1% ± 3.24, and 164% ± 8.54%, respectively), ultimate stress was decreased in all decellularized and recellularized groups. Besides, our results indicated that cell viability on the 1st, 3rd, and 7th day was 100.79% ± 0.7, 100.34% ± 0.08, and 111.24% ± 1.7% for the decellularized rat aorta conjugated with apelin-13, which was incubated for 48-h with Bio-Beads SM-2, and 73.37% ± 7.99, 47.6% ± 11.69, and 27.3% ± 7.89% for decellularized rat aorta scaffolds conjugated with apelin-13 and washed 48-h by PBS, respectively. These findings reveal that the incubation of the scaffold with Bio-Beads SM-2 is a novel and promising approach for increasing cell viability and growth within the scaffold. Conclusions In conclusion, our results provide a platform in which xenograft vessels are decellularized properly in a short time, and the recellularization process is significantly improved after the bioconjugation of the acellular scaffold with apelin-13 in terms of cell adhesion and viability within the scaffold. Graphical Abstract ![]()
Collapse
Affiliation(s)
- Saba Fooladi
- Student Research Committee, Kerman University of Medical Sciences, Kerman, Iran
| | - Sanaz Faramarz
- Student Research Committee, Kerman University of Medical Sciences, Kerman, Iran
| | - Shahriar Dabiri
- Department of Pathology, Pathology and Stem Cells Research Center, Afzalipour Medical School, Kerman University of Medical Sciences, Kerman, Iran
| | - Abdolmohammad Kajbafzadeh
- Pediatric Urology and Regenerative Medicine Research Center, Gene, Cell and Tissue Research Institute, Children Hospital Medical Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohammad Hadi Nematollahi
- Physiology Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran. .,Department of Clinical Biochemistry, Kerman University of Medical Sciences, Kerman, Iran.
| | - Mehrnaz Mehrabani
- Physiology Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran.
| |
Collapse
|