1
|
Chen Y, Zhu Y, Ren X, Ding L, Xu Y, Zhou M, Dong R, Jin P, Chen X, Fan X, Li M, Gong Y, Wang Y. Endothelial Cell Senescence in Marfan Syndrome: Pathogenesis and Therapeutic Potential of TGF-β Pathway Inhibition. J Am Heart Assoc 2025; 14:e037826. [PMID: 40240926 DOI: 10.1161/jaha.124.037826] [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: 07/21/2024] [Accepted: 12/18/2024] [Indexed: 04/18/2025]
Abstract
BACKGROUND Marfan syndrome (MFS) is a heritable connective tissue disorder caused by mutations in the Fibrillin-1 gene, which encodes the extracellular matrix protein fibrillin-1. Patients with MFS are predisposed to aortic aneurysms and dissections, significantly contributing to mortality. Emerging evidence suggests that endothelial cell (EC) senescence plays a critical role in the pathogenesis of aortic aneurysms in MFS. This study aims to elucidate the role of EC senescence in the development of aortic aneurysms in MFS using a vascular model derived from human induced pluripotent stem cells. METHODS AND RESULTS We generated human induced pluripotent stem cells lines from 2 patients with MFS carrying specific Fibrillin-1 mutations and differentiated these into ECs. These MFS-hiPSC-derived ECs were characterized using immunofluorescence, reverse transcription-quantitative polymerase chain reaction, and Western blotting. Functional assays including cell proliferation, scratch wound, tube formation, NO content detection, and senescence-associated β-galactosidase staining were conducted. RNA sequencing was performed to elucidate underlying signaling pathways, and pharmacological inhibition of the transforming growth factor-beta pathway was assessed for its therapeutic potential. MFS-hiPSC-derived ECs recapitulated the pathological features observed in Marfan aortas, particularly pronounced cellular senescence, decreased cell proliferation, and abnormal transforming growth factor-beta and NF-κB signaling. These senescent ECs exhibited diminished proliferative and migratory capacities, reduced NO signaling, increased production of inflammatory cytokines, and attenuated responses to inflammatory stimuli. Importantly, senescence and dysfunction in MFS-hiPSCderived ECs were ameliorated by transforming growth factor-beta signaling pathway inhibitor, SB-431542, suggesting a potential therapeutic strategy. CONCLUSIONS This study highlights the pivotal role of endothelial cell senescence in the pathogenesis of aortic aneurysms in MFS. Our human induced pluripotent stem cells-based disease model provides new insights into the disease mechanisms and underscores the potential of targeting the transforming growth factor-beta pathway to mitigate endothelial dysfunction and senescence, offering a promising therapeutic avenue for MFS.
Collapse
Affiliation(s)
- Yuhao Chen
- Department of Cell Biology, Institute of Hypoxia Medicine, School of Basic Medical Sciences Wenzhou Medical University Zhejiang China
| | - Yuankang Zhu
- Department of Gerontology Xinhua Hospital, Shanghai Jiao Tong University School of Medicine Shanghai China
| | - Xiaoli Ren
- Department of Cell Biology, Institute of Hypoxia Medicine, School of Basic Medical Sciences Wenzhou Medical University Zhejiang China
| | - Lu Ding
- Department of Cell Biology, Institute of Hypoxia Medicine, School of Basic Medical Sciences Wenzhou Medical University Zhejiang China
| | - Yubin Xu
- Department of Cell Biology, Institute of Hypoxia Medicine, School of Basic Medical Sciences Wenzhou Medical University Zhejiang China
| | - Miqi Zhou
- Department of Cell Biology, Institute of Hypoxia Medicine, School of Basic Medical Sciences Wenzhou Medical University Zhejiang China
| | - Runze Dong
- Department of Cell Biology, Institute of Hypoxia Medicine, School of Basic Medical Sciences Wenzhou Medical University Zhejiang China
| | - Peifeng Jin
- Department of Cardiac Surgery The First Affiliated Hospital of Wenzhou Medical University Zhejiang China
| | - Xiufang Chen
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences Wenzhou Medical University Zhejiang China
| | - Xiaofang Fan
- Department of Cell Biology, Institute of Hypoxia Medicine, School of Basic Medical Sciences Wenzhou Medical University Zhejiang China
| | - Ming Li
- Cardiac Regeneration Research Institute, School of Basic Medical Sciences Wenzhou Medical University Zhejiang China
| | - Yongsheng Gong
- Department of Cell Biology, Institute of Hypoxia Medicine, School of Basic Medical Sciences Wenzhou Medical University Zhejiang China
| | - Yongyu Wang
- Department of Cell Biology, Institute of Hypoxia Medicine, School of Basic Medical Sciences Wenzhou Medical University Zhejiang China
- Cardiac Regeneration Research Institute, School of Basic Medical Sciences Wenzhou Medical University Zhejiang China
| |
Collapse
|
2
|
Dalal AR, Pedroza AJ, Kim J, Gilles C, Gu W, Kusadokoro S, Shad R, Mitchel O, Jackson W, Hiesinger W, Berry G, MacFarlane EG, Quertermous T, Cheng P, Fischbein MP. Chemokine (C-C Motif) Ligand 2 Expressing Adventitial Fibroblast Expansion During Loeys-Dietz Syndrome Aortic Aneurysm Formation. Arterioscler Thromb Vasc Biol 2025; 45:722-742. [PMID: 40109260 PMCID: PMC12018128 DOI: 10.1161/atvbaha.124.322069] [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: 11/25/2024] [Accepted: 03/03/2025] [Indexed: 03/22/2025]
Abstract
BACKGROUND Loeys-Dietz syndrome (LDS), caused by mutations in the TGF-β (transforming growth factor-β) signaling cascade, leads to aggressive thoracic aneurysms. While vascular smooth muscle cell (SMC) phenotype modulation has been implicated in thoracic aneurysm formation, we sought to characterize the role of cell state transitions in LDS aneurysm pathogenesis. METHODS We performed single-cell transcriptomic characterization of aortic root/ascending aorta from a murine LDS model (Tgfbr2G357W/+ versus littermate WT [wild-type] control) at 8 weeks, 24 weeks, and aortic root/ascending aortic samples from human LDS surgical specimens (n=5 LDS [TGFBR1/2] and n=2 donor control) to understand cell state transitions and transcriptomic alterations in LDS. Select cell markers were spatially localized with RNA in situ hybridization, immunofluorescence, and immunohistochemistry. Single-cell RNA sequencing of murine and human LDS samples (>30 000 cells) revealed unique SMC, fibroblast, and macrophage transcriptomic profiles in LDS. RESULTS Instead of SMC phenotypic modulation seen in Marfan syndrome, transcriptomic alterations observed in LDS are most prominent in the adventitial fibroblast in the Tgfbr2G357W/+ mouse model. While a distinct modulated SMC cluster does not appear in Tgfbr2G357W/+, SMCs transcriptomically differ from WT counterparts. Adventitial fibroblasts were activated into a proinflammatory state associated with increased macrophage recruitment (Ccl2, Il6, Ccl7, and Cxcl2) and fibrotic response genes (Col1a1, Col1a2, and Col3a1), with a 6-fold increase in aortic wall macrophage content in Tgfbr2G357W/+ compared with WT. Similar findings were also observed in human LDS aortic samples with increased proinflammatory adventitial fibroblast transcriptomic program in parallel with heightened macrophage recruitment. CONCLUSIONS Despite phenotypic similarities in aneurysm formation, the dominant cellular and molecular mechanism of Marfan syndrome and LDS aneurysms are distinct. LDS mouse and human adventitial fibroblasts transcriptomically modulate into a proinflammatory state. Adventitial fibroblasts, in addition to SMCs, are another important pathological cell population during LDS aneurysm formation to consider for targeted therapy to potentially impede LDS aneurysm formation.
Collapse
MESH Headings
- Animals
- Loeys-Dietz Syndrome/genetics
- Loeys-Dietz Syndrome/pathology
- Loeys-Dietz Syndrome/metabolism
- Loeys-Dietz Syndrome/complications
- Humans
- Adventitia/pathology
- Adventitia/metabolism
- Receptor, Transforming Growth Factor-beta Type II/genetics
- Receptor, Transforming Growth Factor-beta Type II/metabolism
- Disease Models, Animal
- Fibroblasts/metabolism
- Fibroblasts/pathology
- Male
- Mice
- Mice, Inbred C57BL
- Aortic Aneurysm, Thoracic/pathology
- Aortic Aneurysm, Thoracic/genetics
- Aortic Aneurysm, Thoracic/metabolism
- Female
- Transcriptome
- Phenotype
- Macrophages/metabolism
- Macrophages/pathology
- Aortic Aneurysm/pathology
- Aortic Aneurysm/metabolism
- Aortic Aneurysm/genetics
- Chemokine CCL2/metabolism
- Chemokine CCL2/genetics
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/pathology
- Aorta/pathology
- Aorta/metabolism
- Single-Cell Analysis
- Muscle, Smooth, Vascular/pathology
- Muscle, Smooth, Vascular/metabolism
Collapse
Affiliation(s)
- Alex R. Dalal
- Department of Cardiothoracic Surgery, Stanford University School of Medicine. Stanford CA, USA
| | - Albert J. Pedroza
- Department of Cardiothoracic Surgery, Stanford University School of Medicine. Stanford CA, USA
| | - Jennifer Kim
- Department of Cardiothoracic Surgery, Stanford University School of Medicine. Stanford CA, USA
| | - Casey Gilles
- Department of Cardiothoracic Surgery, Stanford University School of Medicine. Stanford CA, USA
| | - Wenduo Gu
- Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford CA USA
| | - Sho Kusadokoro
- Department of Cardiothoracic Surgery, Stanford University School of Medicine. Stanford CA, USA
| | - Rohan Shad
- Department of Cardiothoracic Surgery, Stanford University School of Medicine. Stanford CA, USA
| | - Olivia Mitchel
- Department of Cardiothoracic Surgery, Stanford University School of Medicine. Stanford CA, USA
| | - William Jackson
- Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford CA USA
| | - William Hiesinger
- Department of Cardiothoracic Surgery, Stanford University School of Medicine. Stanford CA, USA
| | - Gerald Berry
- Department of Pathology, Stanford University School of Medicine, Stanford CA USA
| | - Elena Gallo MacFarlane
- Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Thomas Quertermous
- Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford CA USA
| | - Paul Cheng
- Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford CA USA
| | - Michael P. Fischbein
- Department of Cardiothoracic Surgery, Stanford University School of Medicine. Stanford CA, USA
| |
Collapse
|
3
|
Jiang B, Ren P, He C, Wang M, Murtada SI, Ruiz-Rodríguez MJ, Chen Y, Ramachandra AB, Li G, Qin L, Assi R, Schwartz MA, Humphrey JD, Tellides G. Short-term disruption of TGF-β signaling in adult mice renders the aorta vulnerable to hypertension-induced dissection. JCI Insight 2025; 10:e182629. [PMID: 39932797 PMCID: PMC11949005 DOI: 10.1172/jci.insight.182629] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Accepted: 02/05/2025] [Indexed: 02/13/2025] Open
Abstract
Hypertension and transient increases in blood pressure from extreme exertion are risk factors for aortic dissection in patients with age-related vascular degeneration or inherited connective tissue disorders. Yet, a common experimental model of angiotensin II-induced aortopathy in mice appears independent of high blood pressure, as lesions do not occur in response to an alternative vasoconstrictor, norepinephrine, and are not prevented by cotreatment with a vasodilator, hydralazine. We investigated vasoconstrictor administration to adult mice following 1 week of disrupted TGF-β signaling in smooth muscle cells (SMCs). Norepinephrine increased blood pressure and induced aortic dissection by 7 days and even within 30 minutes (as did angiotensin II) that was prevented by hydralazine. Initial medial injury manifested as blood extravasation among SMCs and fibrillar matrix, progressive delamination from accumulation of blood, and stretched or ruptured SMCs with persistent attachments to elastic fibers. Altered regulatory contractile molecule expression was not of pathological importance. Rather, reduced synthesis of extracellular matrix yielded a vulnerable aortic phenotype by decreasing medial collagen, most dynamically basement membrane-associated multiplexin collagen, and impairing cell-matrix adhesion. We conclude that transient and sustained increases in blood pressure can cause dissection in aortas rendered vulnerable by inhibition of TGF-β-driven extracellular matrix production by SMCs.
Collapse
Affiliation(s)
- Bo Jiang
- Department of Surgery (Cardiac), Yale School of Medicine, New Haven, Connecticut, USA
| | - Pengwei Ren
- Department of Surgery (Cardiac), Yale School of Medicine, New Haven, Connecticut, USA
| | - Changshun He
- Department of Surgery (Cardiac), Yale School of Medicine, New Haven, Connecticut, USA
| | - Mo Wang
- Department of Surgery (Cardiac), Yale School of Medicine, New Haven, Connecticut, USA
| | - Sae-Il Murtada
- Department of Biomedical Engineering, Yale School of Engineering and Applied Science, New Haven, Connecticut, USA
| | | | - Yu Chen
- Department of Surgery (Cardiac), Yale School of Medicine, New Haven, Connecticut, USA
| | - Abhay B. Ramachandra
- Department of Biomedical Engineering, Yale School of Engineering and Applied Science, New Haven, Connecticut, USA
| | - Guangxin Li
- Department of Surgery (Cardiac), Yale School of Medicine, New Haven, Connecticut, USA
| | - Lingfeng Qin
- Department of Surgery (Cardiac), Yale School of Medicine, New Haven, Connecticut, USA
| | - Roland Assi
- Department of Surgery (Cardiac), Yale School of Medicine, New Haven, Connecticut, USA
- Program in Vascular Biology and Therapeutics, Yale School of Medicine, New Haven, Connecticut, USA
- Veterans Affairs Connecticut Healthcare System, West Haven, Connecticut, USA
| | - Martin A. Schwartz
- Department of Biomedical Engineering, Yale School of Engineering and Applied Science, New Haven, Connecticut, USA
- Department of Medicine (Cardiology)
- Department of Cell Biology, and
- Yale Cardiovascular Research Center, Yale School of Medicine, New Haven, Connecticut, USA
| | - Jay D. Humphrey
- Department of Biomedical Engineering, Yale School of Engineering and Applied Science, New Haven, Connecticut, USA
- Program in Vascular Biology and Therapeutics, Yale School of Medicine, New Haven, Connecticut, USA
| | - George Tellides
- Department of Surgery (Cardiac), Yale School of Medicine, New Haven, Connecticut, USA
- Program in Vascular Biology and Therapeutics, Yale School of Medicine, New Haven, Connecticut, USA
- Veterans Affairs Connecticut Healthcare System, West Haven, Connecticut, USA
| |
Collapse
|
4
|
Cantalupo A, Asano K, Dikalov S, Gordon D, Ramirez F. Fibrillin-1 Deficiency Perturbs Aortic Cholinergic Relaxation and Adrenergic Contraction in a Mouse Model of Early Onset Progressively Severe Marfan Syndrome. J Vasc Res 2025; 62:96-108. [PMID: 39827863 PMCID: PMC11961321 DOI: 10.1159/000542481] [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: 05/28/2024] [Accepted: 11/03/2024] [Indexed: 01/22/2025] Open
Abstract
INTRODUCTION The pathogenic role of nitric oxide (NO) signaling during development of thoracic aortic aneurysm (TAA) in Marfan syndrome (MFS) is currently unclear. We characterized vasomotor function and its relationship to the activity of the NO-generating enzymes in mice with early onset progressively severe MFS. METHODS Wire myography, immunoblotting, measurements of aortic NO, and superoxide levels were used to compare vasomotor function, contractile protein levels, and the activity of endothelial and inducible NO synthase (eNOS and iNOS, respectively) in ascending thoracic aortas of Fbn1mgR/mgR mice relative to wild-type littermates. RESULTS Isometric force measurements of aortic rings from 16-day-old male Fbn1mgR/mgR mice revealed a significant reduction in acetylcholine-induced relaxation and increased phenylephrine (PE)-promoted contractility, associated with abnormally low eNOSSer1177 phosphorylation, decreased NO production, and augmented superoxide levels. Greater aortic contractility was associated with α1-adrenoceptor upregulation and normal levels of contractile proteins. While iNOS inhibition had no effect on vasomotor functions, mutant aortic rings preincubated with a nonspecific NOS inhibitor yielded a greater PE response, implying a significant contribution of endothelial dysfunction to aortic hypercontractility. CONCLUSION Impaired eNOS signaling disrupts aortic cholinergic relaxation and adrenergic contraction in MFS mice with dissecting TAA. INTRODUCTION The pathogenic role of nitric oxide (NO) signaling during development of thoracic aortic aneurysm (TAA) in Marfan syndrome (MFS) is currently unclear. We characterized vasomotor function and its relationship to the activity of the NO-generating enzymes in mice with early onset progressively severe MFS. METHODS Wire myography, immunoblotting, measurements of aortic NO, and superoxide levels were used to compare vasomotor function, contractile protein levels, and the activity of endothelial and inducible NO synthase (eNOS and iNOS, respectively) in ascending thoracic aortas of Fbn1mgR/mgR mice relative to wild-type littermates. RESULTS Isometric force measurements of aortic rings from 16-day-old male Fbn1mgR/mgR mice revealed a significant reduction in acetylcholine-induced relaxation and increased phenylephrine (PE)-promoted contractility, associated with abnormally low eNOSSer1177 phosphorylation, decreased NO production, and augmented superoxide levels. Greater aortic contractility was associated with α1-adrenoceptor upregulation and normal levels of contractile proteins. While iNOS inhibition had no effect on vasomotor functions, mutant aortic rings preincubated with a nonspecific NOS inhibitor yielded a greater PE response, implying a significant contribution of endothelial dysfunction to aortic hypercontractility. CONCLUSION Impaired eNOS signaling disrupts aortic cholinergic relaxation and adrenergic contraction in MFS mice with dissecting TAA.
Collapse
MESH Headings
- Animals
- Marfan Syndrome/physiopathology
- Marfan Syndrome/genetics
- Marfan Syndrome/metabolism
- Marfan Syndrome/complications
- Fibrillin-1/deficiency
- Fibrillin-1/genetics
- Male
- Disease Models, Animal
- Nitric Oxide Synthase Type III/metabolism
- Aorta, Thoracic/physiopathology
- Aorta, Thoracic/metabolism
- Aorta, Thoracic/drug effects
- Nitric Oxide/metabolism
- Vasodilation/drug effects
- Vasoconstriction/drug effects
- Nitric Oxide Synthase Type II/metabolism
- Aortic Aneurysm, Thoracic/physiopathology
- Aortic Aneurysm, Thoracic/metabolism
- Aortic Aneurysm, Thoracic/genetics
- Aortic Aneurysm, Thoracic/etiology
- Acetylcholine/pharmacology
- Superoxides/metabolism
- Receptors, Adrenergic, alpha-1/metabolism
- Receptors, Adrenergic, alpha-1/drug effects
- Signal Transduction
- Phosphorylation
- Mice, Inbred C57BL
- Adrenergic alpha-1 Receptor Agonists/pharmacology
- Severity of Illness Index
- Phenylephrine/pharmacology
- Mice
- Vasodilator Agents/pharmacology
- Disease Progression
- Adipokines
Collapse
Affiliation(s)
- Anna Cantalupo
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Pediatrics, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Keiichi Asano
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Sergey Dikalov
- Division of Clinical Pharmacology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Dylan Gordon
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Francesco Ramirez
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| |
Collapse
|
5
|
Liang G, Lv XF, Huang W, Jin YJ, Roquid KA, Kawase H, Offermanns S. Loss of Smooth Muscle Tenascin-X Inhibits Vascular Remodeling Through Increased TGF-β Signaling. Arterioscler Thromb Vasc Biol 2024; 44:1748-1763. [PMID: 38934115 DOI: 10.1161/atvbaha.123.321067] [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: 04/08/2024] [Accepted: 06/10/2024] [Indexed: 06/28/2024]
Abstract
BACKGROUND Vascular smooth muscle cells (VSMCs) are highly plastic. Vessel injury induces a phenotypic transformation from differentiated to dedifferentiated VSMCs, which involves reduced expression of contractile proteins and increased production of extracellular matrix and inflammatory cytokines. This transition plays an important role in several cardiovascular diseases such as atherosclerosis, hypertension, and aortic aneurysm. TGF-β (transforming growth factor-β) is critical for VSMC differentiation and to counterbalance the effect of dedifferentiating factors. However, the mechanisms controlling TGF-β activity and VSMC phenotypic regulation under in vivo conditions are poorly understood. The extracellular matrix protein TN-X (tenascin-X) has recently been shown to bind TGF-β and to prevent it from activating its receptor. METHODS We studied the role of TN-X in VSMCs in various murine disease models using tamoxifen-inducible SMC-specific knockout and adeno-associated virus-mediated knockdown. RESULTS In hypertensive and high-fat diet-fed mice, after carotid artery ligation as well as in human aneurysmal aortae, expression of Tnxb, the gene encoding TN-X, was increased in VSMCs. Mice with smooth muscle cell-specific loss of TN-X (SMC-Tnxb-KO) showed increased TGF-β signaling in VSMCs, as well as upregulated expression of VSMC differentiation marker genes during vascular remodeling compared with controls. SMC-specific TN-X deficiency decreased neointima formation after carotid artery ligation and reduced vessel wall thickening during Ang II (angiotensin II)-induced hypertension. SMC-Tnxb-KO mice lacking ApoE showed reduced atherosclerosis and Ang II-induced aneurysm formation under high-fat diet. Adeno-associated virus-mediated SMC-specific expression of short hairpin RNA against Tnxb showed similar beneficial effects. Treatment with an anti-TGF-β antibody or additional SMC-specific loss of the TGF-β receptor reverted the effects of SMC-specific TN-X deficiency. CONCLUSIONS In summary, TN-X critically regulates VSMC plasticity during vascular injury by inhibiting TGF-β signaling. Our data indicate that inhibition of vascular smooth muscle TN-X may represent a strategy to prevent and treat pathological vascular remodeling.
Collapse
MESH Headings
- Animals
- Humans
- Male
- Mice
- Angiotensin II
- Aortic Aneurysm/metabolism
- Aortic Aneurysm/pathology
- Aortic Aneurysm/genetics
- Aortic Aneurysm/prevention & control
- Carotid Artery Injuries/pathology
- Carotid Artery Injuries/metabolism
- Carotid Artery Injuries/genetics
- Cells, Cultured
- Diet, High-Fat
- Disease Models, Animal
- Hypertension/metabolism
- Hypertension/pathology
- Hypertension/physiopathology
- Hypertension/genetics
- Mice, Inbred C57BL
- Mice, Knockout
- Mice, Knockout, ApoE
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/pathology
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/pathology
- Neointima
- Phenotype
- Signal Transduction
- Tenascin/metabolism
- Tenascin/genetics
- Tenascin/deficiency
- Transforming Growth Factor beta/metabolism
- Vascular Remodeling
Collapse
Affiliation(s)
- Guozheng Liang
- Department of Pharmacology, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany (G.L., X.-F.L., W.H., Y.-J.J., K.A.R., H.K., S.O.)
| | - Xiao-Fei Lv
- Department of Pharmacology, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany (G.L., X.-F.L., W.H., Y.-J.J., K.A.R., H.K., S.O.)
- Department of Pharmacology, Cardiac and Cerebral Vascular Research Center, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, China (X.-F.L.)
| | - Wei Huang
- Department of Pharmacology, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany (G.L., X.-F.L., W.H., Y.-J.J., K.A.R., H.K., S.O.)
| | - Young-June Jin
- Department of Pharmacology, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany (G.L., X.-F.L., W.H., Y.-J.J., K.A.R., H.K., S.O.)
| | - Kenneth Anthony Roquid
- Department of Pharmacology, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany (G.L., X.-F.L., W.H., Y.-J.J., K.A.R., H.K., S.O.)
| | - Haruya Kawase
- Department of Pharmacology, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany (G.L., X.-F.L., W.H., Y.-J.J., K.A.R., H.K., S.O.)
| | - Stefan Offermanns
- Department of Pharmacology, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany (G.L., X.-F.L., W.H., Y.-J.J., K.A.R., H.K., S.O.)
- Center for Molecular Medicine, Goethe University Frankfurt, Germany (S.O.)
- Cardiopulmonary Institute, Bad Nauheim, Germany (S.O.)
- German Center for Cardiovascular Research, Bad Nauheim, Germany (S.O.)
| |
Collapse
|
6
|
Dhawan U, Williams JA, Windmill JFC, Childs P, Gonzalez-Garcia C, Dalby MJ, Salmeron-Sanchez M. Engineered Surfaces That Promote Capture of Latent Proteins to Facilitate Integrin-Mediated Mechanical Activation of Growth Factors. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2310789. [PMID: 38253339 DOI: 10.1002/adma.202310789] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Revised: 01/13/2024] [Indexed: 01/24/2024]
Abstract
Conventional osteogenic platforms utilize active growth factors to repair bone defects that are extensive in size, but they can adversely affect patient health. Here, an unconventional osteogenic platform is reported that functions by promoting capture of inactive osteogenic growth factor molecules to the site of cell growth for subsequent integrin-mediated activation, using a recombinant fragment of latent transforming growth factor beta-binding protein-1 (rLTBP1). It is shown that rLTBP1 binds to the growth-factor- and integrin-binding domains of fibronectin on poly(ethyl acrylate) surfaces, which immobilizes rLTBP1 and promotes the binding of latency associated peptide (LAP), within which inactive transforming growth factor beta 1 (TGF-β1) is bound. rLTBP1 facilitates the interaction of LAP with integrin β1 and the subsequent mechanically driven release of TGF-β1 to stimulate canonical TGF-β1 signaling, activating osteogenic marker expression in vitro and complete regeneration of a critical-sized bone defect in vivo.
Collapse
Affiliation(s)
- Udesh Dhawan
- Centre for the Cellular Microenvironment, Mazumdar-Shaw Advanced Research Centre, University of Glasgow, Glasgow, G116EW, UK
| | - Jonathan A Williams
- Department of Biomedical Engineering, Wolfson Building, University of Strathclyde, Glasgow, G4 0NW, UK
| | - James F C Windmill
- Centre for Ultrasonic Engineering, Department of Electronic and Electrical Engineering, University of Strathclyde, Glasgow, G11XW, UK
| | - Peter Childs
- Department of Biomedical Engineering, Wolfson Building, University of Strathclyde, Glasgow, G4 0NW, UK
| | - Cristina Gonzalez-Garcia
- Centre for the Cellular Microenvironment, Mazumdar-Shaw Advanced Research Centre, University of Glasgow, Glasgow, G116EW, UK
| | - Matthew J Dalby
- Centre for the Cellular Microenvironment, Mazumdar-Shaw Advanced Research Centre, University of Glasgow, Glasgow, G116EW, UK
| | - Manuel Salmeron-Sanchez
- Centre for the Cellular Microenvironment, Mazumdar-Shaw Advanced Research Centre, University of Glasgow, Glasgow, G116EW, UK
- Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute for Science and Technology (BIST), Barcelona, 08028, Spain
- Institució Catalana de Recerca i Estudis Avançats (ICREA), Barcelona, 08010, Spain
| |
Collapse
|
7
|
Jiang B, Ren P, He C, Wang M, Murtada SI, Chen Y, Ramachandra AB, Li G, Qin L, Assi R, Schwartz MA, Humphrey JD, Tellides G. Short-Term Disruption of TGFβ Signaling in Adult Mice Renders the Aorta Vulnerable to Hypertension-Induced Dissection. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.22.590484. [PMID: 38712205 PMCID: PMC11071440 DOI: 10.1101/2024.04.22.590484] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/08/2024]
Abstract
Hypertension and transient increases in blood pressure from extreme exertion are risk factors for aortic dissection in patients with age-related vascular degeneration or inherited connective tissue disorders. Yet, the common experimental model of angiotensin II-induced aortopathy in mice appears independent of high blood pressure as lesions do not occur in response to an alternative vasoconstrictor, norepinephrine, and are not prevented by co-treatment with a vasodilator, hydralazine. We investigated vasoconstrictor administration to adult mice 1 week after disruption of TGFβ signaling in smooth muscle cells. Norepinephrine increased blood pressure and induced aortic dissection by 7 days and even within 30 minutes that was rescued by hydralazine; results were similar with angiotensin II. Changes in regulatory contractile molecule expression were not of pathological significance. Rather, reduced synthesis of extracellular matrix yielded a vulnerable aortic phenotype by decreasing medial collagen, most dynamically type XVIII, and impairing cell-matrix adhesion. We conclude that transient and sustained increases in blood pressure cause dissection in aortas rendered vulnerable by inhibition of TGFβ-driven extracellular matrix production by smooth muscle cells. A corollary is that medial fibrosis, a frequent feature of medial degeneration, may afford some protection against aortic dissection.
Collapse
|
8
|
Singh AA, Shetty DK, Jacob AG, Bayraktar S, Sinha S. Understanding genomic medicine for thoracic aortic disease through the lens of induced pluripotent stem cells. Front Cardiovasc Med 2024; 11:1349548. [PMID: 38440211 PMCID: PMC10910110 DOI: 10.3389/fcvm.2024.1349548] [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: 12/04/2023] [Accepted: 01/31/2024] [Indexed: 03/06/2024] Open
Abstract
Thoracic aortic disease (TAD) is often silent until a life-threatening complication occurs. However, genetic information can inform both identification and treatment at an early stage. Indeed, a diagnosis is important for personalised surveillance and intervention plans, as well as cascade screening of family members. Currently, only 20% of heritable TAD patients have a causative mutation identified and, consequently, further advances in genetic coverage are required to define the remaining molecular landscape. The rapid expansion of next generation sequencing technologies is providing a huge resource of genetic data, but a critical issue remains in functionally validating these findings. Induced pluripotent stem cells (iPSCs) are patient-derived, reprogrammed cell lines which allow mechanistic insights, complex modelling of genetic disease and a platform to study aortic genetic variants. This review will address the need for iPSCs as a frontline diagnostic tool to evaluate variants identified by genomic discovery studies and explore their evolving role in biological insight through to drug discovery.
Collapse
Affiliation(s)
| | | | | | | | - Sanjay Sinha
- Wellcome-MRC Cambridge Stem Cell Institute, Jeffrey Cheah Biomedical Centre, Cambridge, United Kingdom
| |
Collapse
|
9
|
Lei C, Kan H, Xian X, Chen W, Xiang W, Song X, Wu J, Yang D, Zheng Y. FAM3A reshapes VSMC fate specification in abdominal aortic aneurysm by regulating KLF4 ubiquitination. Nat Commun 2023; 14:5360. [PMID: 37660071 PMCID: PMC10475135 DOI: 10.1038/s41467-023-41177-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: 11/05/2022] [Accepted: 08/24/2023] [Indexed: 09/04/2023] Open
Abstract
Reprogramming of vascular smooth muscle cell (VSMC) differentiation plays an essential role in abdominal aortic aneurysm (AAA). However, the underlying mechanisms are still unclear. We explore the expression of FAM3A, a newly identified metabolic cytokine, and whether and how FAM3A regulates VSMC differentiation in AAA. We discover that FAM3A is decreased in the aortas and plasma in AAA patients and murine models. Overexpression or supplementation of FAM3A significantly attenuate the AAA formation, manifested by maintenance of the well-differentiated VSMC status and inhibition of VSMC transformation toward macrophage-, chondrocyte-, osteogenic-, mesenchymal-, and fibroblast-like cell subpopulations. Importantly, FAM3A induces KLF4 ubiquitination and reduces its phosphorylation and nuclear localization. Here, we report FAM3A as a VSMC fate-shaping regulator in AAA and reveal the underlying mechanism associated with KLF4 ubiquitination and stability, which may lead to the development of strategies based on FAM3A to restore VSMC homeostasis in AAA.
Collapse
Affiliation(s)
- Chuxiang Lei
- Department of Vascular Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Dongcheng District, Beijing, 100730, China
| | - Haoxuan Kan
- Department of Vascular Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Dongcheng District, Beijing, 100730, China
| | - Xiangyu Xian
- Department of Vascular Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Dongcheng District, Beijing, 100730, China
| | - Wenlin Chen
- Department of Vascular Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Dongcheng District, Beijing, 100730, China
| | - Wenxuan Xiang
- Department of Vascular Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Dongcheng District, Beijing, 100730, China
| | - Xiaohong Song
- Department of Vascular Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Dongcheng District, Beijing, 100730, China
- State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Dongcheng District, Beijing, 100730, China
| | - Jianqiang Wu
- Department of Vascular Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Dongcheng District, Beijing, 100730, China
- State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Dongcheng District, Beijing, 100730, China
| | - Dan Yang
- Department of Computational Biology and Bioinformatics, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences and Peking Union Medical College, Haidian District, Beijing, 100193, China.
| | - Yuehong Zheng
- Department of Vascular Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Dongcheng District, Beijing, 100730, China.
| |
Collapse
|
10
|
Hu Y, Cai Z, He B. Smooth Muscle Heterogeneity and Plasticity in Health and Aortic Aneurysmal Disease. Int J Mol Sci 2023; 24:11701. [PMID: 37511460 PMCID: PMC10380637 DOI: 10.3390/ijms241411701] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2023] [Revised: 07/16/2023] [Accepted: 07/18/2023] [Indexed: 07/30/2023] Open
Abstract
Vascular smooth muscle cells (VSMCs) are the predominant cell type in the medial layer of the aorta, which plays a critical role in the maintenance of aortic wall integrity. VSMCs have been suggested to have contractile and synthetic phenotypes and undergo phenotypic switching to contribute to the deteriorating aortic wall structure. Recently, the unprecedented heterogeneity and diversity of VSMCs and their complex relationship to aortic aneurysms (AAs) have been revealed by high-resolution research methods, such as lineage tracing and single-cell RNA sequencing. The aortic wall consists of VSMCs from different embryonic origins that respond unevenly to genetic defects that directly or indirectly regulate VSMC contractile phenotype. This difference predisposes to hereditary AAs in the aortic root and ascending aorta. Several VSMC phenotypes with different functions, for example, secreting VSMCs, proliferative VSMCs, mesenchymal stem cell-like VSMCs, immune-related VSMCs, proinflammatory VSMCs, senescent VSMCs, and stressed VSMCs are identified in non-hereditary AAs. The transformation of VSMCs into different phenotypes is an adaptive response to deleterious stimuli but can also trigger pathological remodeling that exacerbates the pathogenesis and development of AAs. This review is intended to contribute to the understanding of VSMC diversity in health and aneurysmal diseases. Papers that give an update on VSMC phenotype diversity in health and aneurysmal disease are summarized and recent insights on the role of VSMCs in AAs are discussed.
Collapse
Affiliation(s)
- Yunwen Hu
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200030, China
| | - Zhaohua Cai
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200030, China
| | - Ben He
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200030, China
| |
Collapse
|
11
|
Jara ZP, Harford T, Singh KD, Desnoyer R, Kumar A, Srinivasan D, Karnik SS. Distinct Mechanisms of β-Arrestin-Biased Agonist and Blocker of AT1R in Preventing Aortic Aneurysm and Associated Mortality. Hypertension 2023; 80:385-402. [PMID: 36440576 PMCID: PMC9852074 DOI: 10.1161/hypertensionaha.122.19232] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2022] [Accepted: 11/04/2022] [Indexed: 11/29/2022]
Abstract
BACKGROUND Aortic aneurysm (AA) is a "silent killer" human disease with no effective treatment. Although the therapeutic potential of various pharmacological agents have been evaluated, there are no reports of β-arrestin-biased AT1R (angiotensin-II type-1 receptor) agonist (TRV027) used to prevent the progression of AA. METHODS We tested the hypothesis that TRV027 infusion in AngII (angiotensin II)-induced mouse model of AA prevents AA. High-fat-diet-fed ApoE (apolipoprotein E gene)-null mice were infused with AngII to induce AA and co-infused with TRV027 and a clinically used AT1R blocker Olmesartan to prevent AA. Aortas explanted from different ligand infusion groups were compared with assess different grades of AA or lack of AA. RESULTS AngII produced AA in ≈67% male mice with significant mortality associated with AA rupture. We observed ≈13% mortality due to aortic arch dissection without aneurysm in male mice. AngII-induced AA and mortality was prevented by co-infusion of TRV027 or Olmesartan, but through different mechanisms. In TRV027 co-infused mice aortic wall thickness, elastin content, new DNA, and protein synthesis were higher than untreated and Olmesartan co-infused mice. Co-infusion with both TRV027 and Olmesartan prevented endoplasmic reticulum stress, fibrosis, and vasomotor hyper responsiveness. CONCLUSIONS TRV027-engaged AT1R prevented AA and associated mortality by distinct molecular mechanisms compared with the AT1R blocker, Olmesartan. Developing novel β-arrestin-biased AT1R ligands may yield promising drugs to combat AA.
Collapse
Affiliation(s)
- Zaira Palomino Jara
- Cardiovascular and Metabolic Sciences Department, Lerner Research Institute, Cleveland Clinic
| | - Terri Harford
- Cardiovascular and Metabolic Sciences Department, Lerner Research Institute, Cleveland Clinic
| | | | - Russell Desnoyer
- Cardiovascular and Metabolic Sciences Department, Lerner Research Institute, Cleveland Clinic
| | - Avinash Kumar
- Pathobiology Department, Lerner Research Institute, Cleveland Clinic
| | | | - Sadashiva S. Karnik
- Cardiovascular and Metabolic Sciences Department, Lerner Research Institute, Cleveland Clinic
| |
Collapse
|
12
|
Verstraeten A, Fedoryshchenko I, Loeys B. The emerging role of endothelial cells in the pathogenesis of thoracic aortic aneurysm and dissection. Eur Heart J 2023; 44:1262-1264. [PMID: 36650899 PMCID: PMC10079389 DOI: 10.1093/eurheartj/ehac771] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Affiliation(s)
- Aline Verstraeten
- Cardiogenomics and Functional Genomics, Center for Medical Genetics, University of Antwerp/Antwerp University Hospital, Prins Boudewijnlaan 43, 2650 Antwerp, Belgium
| | - Ivanna Fedoryshchenko
- Cardiogenomics and Functional Genomics, Center for Medical Genetics, University of Antwerp/Antwerp University Hospital, Prins Boudewijnlaan 43, 2650 Antwerp, Belgium
| | - Bart Loeys
- Cardiogenomics and Functional Genomics, Center for Medical Genetics, University of Antwerp/Antwerp University Hospital, Prins Boudewijnlaan 43, 2650 Antwerp, Belgium
| |
Collapse
|
13
|
Dong CX, Malecki C, Robertson E, Hambly B, Jeremy R. Molecular Mechanisms in Genetic Aortopathy-Signaling Pathways and Potential Interventions. Int J Mol Sci 2023; 24:ijms24021795. [PMID: 36675309 PMCID: PMC9865322 DOI: 10.3390/ijms24021795] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 01/02/2023] [Accepted: 01/11/2023] [Indexed: 01/18/2023] Open
Abstract
Thoracic aortic disease affects people of all ages and the majority of those aged <60 years have an underlying genetic cause. There is presently no effective medical therapy for thoracic aneurysm and surgery remains the principal intervention. Unlike abdominal aortic aneurysm, for which the inflammatory/atherosclerotic pathogenesis is well established, the mechanism of thoracic aneurysm is less understood. This paper examines the key cell signaling systems responsible for the growth and development of the aorta, homeostasis of endothelial and vascular smooth muscle cells and interactions between pathways. The evidence supporting a role for individual signaling pathways in pathogenesis of thoracic aortic aneurysm is examined and potential novel therapeutic approaches are reviewed. Several key signaling pathways, notably TGF-β, WNT, NOTCH, PI3K/AKT and ANGII contribute to growth, proliferation, cell phenotype and survival for both vascular smooth muscle and endothelial cells. There is crosstalk between pathways, and between vascular smooth muscle and endothelial cells, with both synergistic and antagonistic interactions. A common feature of the activation of each is response to injury or abnormal cell stress. Considerable experimental evidence supports a contribution of each of these pathways to aneurysm formation. Although human information is less, there is sufficient data to implicate each pathway in the pathogenesis of human thoracic aneurysm. As some pathways i.e., WNT and NOTCH, play key roles in tissue growth and organogenesis in early life, it is possible that dysregulation of these pathways results in an abnormal aortic architecture even in infancy, thereby setting the stage for aneurysm development in later life. Given the fine tuning of these signaling systems, functional polymorphisms in key signaling elements may set up a future risk of thoracic aneurysm. Multiple novel therapeutic agents have been developed, targeting cell signaling pathways, predominantly in cancer medicine. Future investigations addressing cell specific targeting, reduced toxicity and also less intense treatment effects may hold promise for effective new medical treatments of thoracic aortic aneurysm.
Collapse
Affiliation(s)
- Charlotte Xue Dong
- Faculty of Health and Medical Sciences, University of Sydney, Sydney, NSW 2006, Australia
| | - Cassandra Malecki
- Faculty of Health and Medical Sciences, University of Sydney, Sydney, NSW 2006, Australia
- The Baird Institute, Camperdown, NSW 2042, Australia
| | - Elizabeth Robertson
- Faculty of Health and Medical Sciences, University of Sydney, Sydney, NSW 2006, Australia
| | - Brett Hambly
- Faculty of Health and Medical Sciences, University of Sydney, Sydney, NSW 2006, Australia
| | - Richmond Jeremy
- Faculty of Health and Medical Sciences, University of Sydney, Sydney, NSW 2006, Australia
- The Baird Institute, Camperdown, NSW 2042, Australia
- Correspondence:
| |
Collapse
|
14
|
Mieremet A, van der Stoel M, Li S, Coskun E, van Krimpen T, Huveneers S, de Waard V. Endothelial dysfunction in Marfan syndrome mice is restored by resveratrol. Sci Rep 2022; 12:22504. [PMID: 36577770 PMCID: PMC9797556 DOI: 10.1038/s41598-022-26662-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2022] [Accepted: 12/19/2022] [Indexed: 12/29/2022] Open
Abstract
Patients with Marfan syndrome (MFS) develop thoracic aortic aneurysms as the aorta presents excessive elastin breaks, fibrosis, and vascular smooth muscle cell (vSMC) death due to mutations in the FBN1 gene. Despite elaborate vSMC to aortic endothelial cell (EC) signaling, the contribution of ECs to the development of aortic pathology remains largely unresolved. The aim of this study is to investigate the EC properties in Fbn1C1041G/+ MFS mice. Using en face immunofluorescence confocal microscopy, we showed that EC alignment with blood flow was reduced, EC roundness was increased, individual EC surface area was larger, and EC junctional linearity was decreased in aortae of Fbn1C1041G/+ MFS mice. This modified EC phenotype was most prominent in the ascending aorta and occurred before aortic dilatation. To reverse EC morphology, we performed treatment with resveratrol. This restored EC blood flow alignment, junctional linearity, phospho-eNOS expression, and improved the structural integrity of the internal elastic lamina of Fbn1C1041G/+ mice. In conclusion, these experiments identify the involvement of ECs and underlying internal elastic lamina in MFS aortic pathology, which could act as potential target for future MFS pharmacotherapies.
Collapse
Affiliation(s)
- Arnout Mieremet
- Department of Medical Biochemistry, Amsterdam UMC Location University of Amsterdam, Meibergdreef 9, Amsterdam, The Netherlands
- Amsterdam Cardiovascular Sciences, Atherosclerosis and Ischemic Syndromes, Amsterdam, The Netherlands
| | - Miesje van der Stoel
- Department of Medical Biochemistry, Amsterdam UMC Location University of Amsterdam, Meibergdreef 9, Amsterdam, The Netherlands
- Amsterdam Cardiovascular Sciences, Atherosclerosis and Ischemic Syndromes, Amsterdam, The Netherlands
- Amsterdam Cardiovascular Sciences, Microcirculation, Amsterdam, The Netherlands
| | - Siyu Li
- Department of Medical Biochemistry, Amsterdam UMC Location University of Amsterdam, Meibergdreef 9, Amsterdam, The Netherlands
- Amsterdam Cardiovascular Sciences, Atherosclerosis and Ischemic Syndromes, Amsterdam, The Netherlands
| | - Evrim Coskun
- Department of Medical Biochemistry, Amsterdam UMC Location University of Amsterdam, Meibergdreef 9, Amsterdam, The Netherlands
- Amsterdam Cardiovascular Sciences, Atherosclerosis and Ischemic Syndromes, Amsterdam, The Netherlands
| | - Tsveta van Krimpen
- Department of Medical Biochemistry, Amsterdam UMC Location University of Amsterdam, Meibergdreef 9, Amsterdam, The Netherlands
- Amsterdam Cardiovascular Sciences, Atherosclerosis and Ischemic Syndromes, Amsterdam, The Netherlands
- Amsterdam Cardiovascular Sciences, Microcirculation, Amsterdam, The Netherlands
| | - Stephan Huveneers
- Department of Medical Biochemistry, Amsterdam UMC Location University of Amsterdam, Meibergdreef 9, Amsterdam, The Netherlands
- Amsterdam Cardiovascular Sciences, Atherosclerosis and Ischemic Syndromes, Amsterdam, The Netherlands
- Amsterdam Cardiovascular Sciences, Microcirculation, Amsterdam, The Netherlands
| | - Vivian de Waard
- Department of Medical Biochemistry, Amsterdam UMC Location University of Amsterdam, Meibergdreef 9, Amsterdam, The Netherlands.
- Amsterdam Cardiovascular Sciences, Atherosclerosis and Ischemic Syndromes, Amsterdam, The Netherlands.
| |
Collapse
|
15
|
Taniguchi R, Ohashi Y, Lee JS, Hu H, Gonzalez L, Zhang W, Langford J, Matsubara Y, Yatsula B, Tellides G, Fahmy TM, Hoshina K, Dardik A. Endothelial Cell TGF-β (Transforming Growth Factor-Beta) Signaling Regulates Venous Adaptive Remodeling to Improve Arteriovenous Fistula Patency. Arterioscler Thromb Vasc Biol 2022; 42:868-883. [PMID: 35510552 DOI: 10.1161/atvbaha.122.317676] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
BACKGROUND Arteriovenous fistulae (AVF) are the gold standard for vascular access for hemodialysis. Although the vein must thicken and dilate for successful hemodialysis, excessive wall thickness leads to stenosis causing AVF failure. Since TGF-β (transforming growth factor-beta) regulates ECM (extracellular matrix) deposition and smooth muscle cell (SMC) proliferation-critical components of wall thickness-we hypothesized that disruption of TGF-β signaling prevents excessive wall thickening during venous remodeling. METHODS A mouse aortocaval fistula model was used. SB431542-an inhibitor of TGF-β receptor I-was encapsulated in nanoparticles and applied to the AVF adventitia in C57BL/6J mice. Alternatively, AVFs were created in mice with conditional disruption of TGF-β receptors in either SMCs or endothelial cells. Doppler ultrasound was performed serially to confirm patency and to measure vessel diameters. AVFs were harvested at predetermined time points for histological and immunofluorescence analyses. RESULTS Inhibition of TGF-β signaling with SB431542-containing nanoparticles significantly reduced p-Smad2-positive cells in the AVF wall during the early maturation phase (days 7-21) and was associated with decreased AVF wall thickness that showed both decreased collagen density and decreased SMC proliferation. SMC-specific TGF-β signaling disruption decreased collagen density but not SMC proliferation or wall thickness. Endothelial cell-specific TGF-β signaling disruption decreased both collagen density and SMC proliferation in the AVF wall and was associated with reduced wall thickness, increased outward remodeling, and improved AVF patency. CONCLUSIONS Endothelial cell-targeted TGF-β inhibition may be a translational strategy to improve AVF patency.
Collapse
Affiliation(s)
- Ryosuke Taniguchi
- Vascular Biology and Therapeutics Program (R.T., Y.O., H.H., L.G., W.Z., J.L., Y.M., B.Y., G.T., A.D.), Yale School of Medicine, New Haven, CT.,Division of Vascular Surgery, The University of Tokyo, Japan (R.T., Y.O., K.H.)
| | - Yuichi Ohashi
- Vascular Biology and Therapeutics Program (R.T., Y.O., H.H., L.G., W.Z., J.L., Y.M., B.Y., G.T., A.D.), Yale School of Medicine, New Haven, CT.,Division of Vascular Surgery, The University of Tokyo, Japan (R.T., Y.O., K.H.)
| | - Jung Seok Lee
- Department of Biomedical Engineering, Yale University, New Haven, CT (J.S.L., T.M.F.)
| | - Haidi Hu
- Vascular Biology and Therapeutics Program (R.T., Y.O., H.H., L.G., W.Z., J.L., Y.M., B.Y., G.T., A.D.), Yale School of Medicine, New Haven, CT.,Department of Vascular and Thyroid Surgery, The First Hospital of China Medical University, Shenyang (H.H.)
| | - Luis Gonzalez
- Vascular Biology and Therapeutics Program (R.T., Y.O., H.H., L.G., W.Z., J.L., Y.M., B.Y., G.T., A.D.), Yale School of Medicine, New Haven, CT
| | - Weichang Zhang
- Vascular Biology and Therapeutics Program (R.T., Y.O., H.H., L.G., W.Z., J.L., Y.M., B.Y., G.T., A.D.), Yale School of Medicine, New Haven, CT
| | - John Langford
- Vascular Biology and Therapeutics Program (R.T., Y.O., H.H., L.G., W.Z., J.L., Y.M., B.Y., G.T., A.D.), Yale School of Medicine, New Haven, CT
| | - Yutaka Matsubara
- Vascular Biology and Therapeutics Program (R.T., Y.O., H.H., L.G., W.Z., J.L., Y.M., B.Y., G.T., A.D.), Yale School of Medicine, New Haven, CT.,Department of Surgery and Sciences, Kyushu University, Fukuoka, Japan (Y.M.)
| | - Bogdan Yatsula
- Vascular Biology and Therapeutics Program (R.T., Y.O., H.H., L.G., W.Z., J.L., Y.M., B.Y., G.T., A.D.), Yale School of Medicine, New Haven, CT
| | - George Tellides
- Vascular Biology and Therapeutics Program (R.T., Y.O., H.H., L.G., W.Z., J.L., Y.M., B.Y., G.T., A.D.), Yale School of Medicine, New Haven, CT.,Division of Cardiac Surgery, Department of Surgery (G.T.), Yale School of Medicine, New Haven, CT.,Department of Surgery, VA Connecticut Healthcare Systems, West Haven, CT (G.T., A.D.)
| | - Tarek M Fahmy
- Department of Biomedical Engineering, Yale University, New Haven, CT (J.S.L., T.M.F.)
| | - Katsuyuki Hoshina
- Division of Vascular Surgery, The University of Tokyo, Japan (R.T., Y.O., K.H.)
| | - Alan Dardik
- Vascular Biology and Therapeutics Program (R.T., Y.O., H.H., L.G., W.Z., J.L., Y.M., B.Y., G.T., A.D.), Yale School of Medicine, New Haven, CT.,Division of Vascular and Endovascular Surgery, Department of Surgery (A.D.), Yale School of Medicine, New Haven, CT.,Department of Surgery, VA Connecticut Healthcare Systems, West Haven, CT (G.T., A.D.)
| |
Collapse
|
16
|
Jauhiainen S, Kiema M, Hedman M, Laakkonen JP. Large Vessel Cell Heterogeneity and Plasticity: Focus in Aortic Aneurysms. Arterioscler Thromb Vasc Biol 2022; 42:811-818. [PMID: 35587695 DOI: 10.1161/atvbaha.121.316237] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Smooth muscle cells and endothelial cells have a remarkable level of plasticity in vascular pathologies. In thoracic and abdominal aortic aneurysms, smooth muscle cells have been suggested to undergo phenotypic switching and to contribute to degradation of the aortic wall structure in response to, for example, inflammatory mediators, dysregulation of growth factor signaling or oxidative stress. Recently, endothelial-to-mesenchymal transition, and a clonal expansion of degradative smooth muscle cells and immune cells, as well as mesenchymal stem-like cells have been suggested to contribute to the progression of aortic aneurysms. What are the factors driving the aortic cell phenotype changes and how vascular flow, known to affect aortic wall structure and to be altered in aortic aneurysms, could affect aortic cell remodeling? In this review, we summarize the current literature on aortic cell heterogeneity and phenotypic switching in relation to changes in vascular flow and aortic wall structure in aortic aneurysms in clinical samples with special focus on smooth muscle and endothelial cells. The differences between thoracic and abdominal aortic aneurysms are discussed.
Collapse
Affiliation(s)
- Suvi Jauhiainen
- A.I. Virtanen Institute for Molecular Sciences (S.J., M.K., J.P.L.), University of Eastern Finland, Kuopio
| | - Miika Kiema
- A.I. Virtanen Institute for Molecular Sciences (S.J., M.K., J.P.L.), University of Eastern Finland, Kuopio
| | - Marja Hedman
- Institute of Clinical Medicine (M.H.), University of Eastern Finland, Kuopio
- Department of Clinical Radiology, Kuopio University Hospital, Finland (M.H.)
- Department of Heart and Thoracic Surgery, Kuopio University Hospital, Heart Center, Kuopio, Finland (M.H.)
| | - Johanna P Laakkonen
- A.I. Virtanen Institute for Molecular Sciences (S.J., M.K., J.P.L.), University of Eastern Finland, Kuopio
| |
Collapse
|
17
|
Lee CY, Angelov SN, Zhu J, Bi L, Sanford N, Alp Yildirim I, Dichek DA. Blockade of TGF-β (Transforming Growth Factor Beta) Signaling by Deletion of Tgfbr2 in Smooth Muscle Cells of 11-Month-Old Mice Alters Aortic Structure and Causes Vasomotor Dysfunction-Brief Report. Arterioscler Thromb Vasc Biol 2022; 42:764-771. [PMID: 35443795 DOI: 10.1161/atvbaha.122.317603] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND To test the hypothesis that smooth muscle cell (SMC) TGF-β (transforming growth factor beta) signaling contributes to maintenance of aortic structure and function beyond the early postnatal period. METHODS We deleted the TBR2 (type 2 TGF-β receptor) in SMC of 11-month-old mice (genotype Acta2-CreERT2+/0 Tgfbr2f/f, termed TBR2SMΔ) and compared their ascending aorta structure and vasomotor function to controls (Acta2-CreERT20/0 Tgfbr2f/f, termed TBR2f/f). RESULTS We confirmed loss of aortic SMC TBR2 by immunoblotting. Four weeks after SMC TBR2 loss, TBR2SMΔ mice did not have aortic rupture, ulceration, dissection, dilation, or evidence of medial hemorrhage. However, aortic medial area of TBR2SMΔ mice was increased by 27% (0.14±0.01 versus 0.11±0.01 mm2; P=0.01) and medial thickness was increased by 23% (40±1.9 versus 33±1.3 μm; P=0.004) compared with littermate controls. Wire myography performed on ascending aortic rings showed hypercontractility of TBR2SMΔ aortas to phenylephrine (Emax, 15.9±1.2 versus 10.8±0.7 mN; P=0.0003) and reduced relaxation and sensitivity to acetylcholine (Emax, 64±14% versus 96±2%; P=0.001; -logEC50, 6.9±0.1 versus 7.7±0.1; P=0.0001). Neither maximal relaxation nor sensitivity to sodium nitroprusside differed (Emax, 102±0.3% versus 101±0.3%; -logEC50, 8.0±0.04 versus 7.9±0.08; P>0.4 for both). CONCLUSIONS Loss of TGF-β signaling in aortic SMC of 1-year-old mice does not cause early severe aortopathy or death; however, it causes mild structural and substantial physiological abnormalities. SMC TGF-β signaling plays an important role in maintaining aortic homeostasis in older mice. This role should be considered in the design of clinical studies that aim to prevent aortopathy by blocking SMC TGF-β signaling.
Collapse
Affiliation(s)
- Chloe Y Lee
- Department of Medicine (C.Y.L., S.N.A., L.B., N.S., I.A.Y., D.A.D.), University of Washington School of Medicine, Seattle
| | - Stoyan N Angelov
- Department of Medicine (C.Y.L., S.N.A., L.B., N.S., I.A.Y., D.A.D.), University of Washington School of Medicine, Seattle
| | - Jay Zhu
- Department of Surgery (J.Z.), University of Washington School of Medicine, Seattle
| | - Lianxiang Bi
- Department of Medicine (C.Y.L., S.N.A., L.B., N.S., I.A.Y., D.A.D.), University of Washington School of Medicine, Seattle
| | - Nicole Sanford
- Department of Medicine (C.Y.L., S.N.A., L.B., N.S., I.A.Y., D.A.D.), University of Washington School of Medicine, Seattle
| | - Ilkay Alp Yildirim
- Department of Medicine (C.Y.L., S.N.A., L.B., N.S., I.A.Y., D.A.D.), University of Washington School of Medicine, Seattle
| | - David A Dichek
- Department of Medicine (C.Y.L., S.N.A., L.B., N.S., I.A.Y., D.A.D.), University of Washington School of Medicine, Seattle.,Institute for Stem Cell and Regenerative Medicine' Department of Laboratory Medicine and Pathology (D.A.D.), University of Washington School of Medicine, Seattle
| |
Collapse
|
18
|
Zhang F, King MW. Immunomodulation Strategies for the Successful Regeneration of a Tissue-Engineered Vascular Graft. Adv Healthc Mater 2022; 11:e2200045. [PMID: 35286778 PMCID: PMC11468936 DOI: 10.1002/adhm.202200045] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Revised: 02/18/2022] [Indexed: 01/02/2023]
Abstract
Cardiovascular disease leads to the highest morbidity worldwide. There is an urgent need to solve the lack of a viable arterial graft for patients requiring coronary artery bypass surgery. The current gold standard is to use the patient's own blood vessel, such as a saphenous vein graft. However, some patients do not have appropriate vessels to use because of systemic disease or secondary surgery. On the other hand, there is no commercially available synthetic vascular graft available on the market for small diameter (<6 mm) blood vessels like coronary, carotid, and peripheral popliteal arteries. Tissue-engineered vascular grafts (TEVGs) are studied in recent decades as a promising alternative to synthetic arterial prostheses. Yet only a few studies have proceeded to a clinical trial. Recent studies have uncovered that the host immune response can be directed toward increasing the success of a TEVG by shedding light on ways to modulate the macrophage response and improve the tissue regeneration outcome. In this review, the basic concepts of vascular tissue engineering and immunoengineering are considered. The state-of-art of TEVGs is summarized and the role of macrophages in TEVG regeneration is analyzed. Current immunomodulatory strategies based on biomaterials are also discussed.
Collapse
Affiliation(s)
- Fan Zhang
- Wilson College of TextilesNorth Carolina State UniversityRaleighNC27606USA
| | - Martin W. King
- Wilson College of TextilesNorth Carolina State UniversityRaleighNC27606USA
| |
Collapse
|
19
|
Pathophysiology and Therapeutics of Thoracic Aortic Aneurysm in Marfan Syndrome. Biomolecules 2022; 12:biom12010128. [PMID: 35053276 PMCID: PMC8773516 DOI: 10.3390/biom12010128] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 01/04/2022] [Accepted: 01/06/2022] [Indexed: 12/16/2022] Open
Abstract
About 20% of individuals afflicted with thoracic aortic disease have single-gene mutations that predispose the vessel to aneurysm formation and/or acute aortic dissection often without associated syndromic features. One widely studied exception is Marfan syndrome (MFS) in which mutations in the extracellular protein fibrillin-1 cause additional abnormalities in the heart, eyes, and skeleton. Mouse models of MFS have been instrumental in delineating major cellular and molecular determinants of thoracic aortic disease. In spite of research efforts, translating experimental findings from MFS mice into effective drug therapies for MFS patients remains an unfulfilled promise. Here, we describe a series of studies that have implicated endothelial dysfunction and improper angiotensin II and TGFβ signaling in driving thoracic aortic disease in MFS mice. We also discuss how these investigations have influenced the way we conceptualized possible new therapies to slow down or even halt aneurysm progression in this relatively common connective tissue disorder.
Collapse
|