1
|
DeMaria WG, Figueroa-Milla AE, Kaija A, Harrington AE, Tero B, Ryzhova L, Liaw L, Rolle MW. Endothelial Cells Increase Mesenchymal Stem Cell Differentiation in Scaffold-Free 3D Vascular Tissue. Tissue Eng Part A 2024. [PMID: 39109944 DOI: 10.1089/ten.tea.2024.0122] [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: 09/13/2024] Open
Abstract
In this study, we present a versatile, scaffold-free approach to create ring-shaped engineered vascular tissue segments using human mesenchymal stem cell-derived smooth muscle cells (hMSC-SMCs) and endothelial cells (ECs). We hypothesized that incorporation of ECs would increase hMSC-SMC differentiation without compromising tissue ring strength or fusion to form tissue tubes. Undifferentiated hMSCs and ECs were co-seeded into custom ring-shaped agarose wells using four different concentrations of ECs: 0%, 10%, 20%, and 30%. Co-seeded EC and hMSC rings were cultured in SMC differentiation medium for a total of 22 days. Tissue rings were then harvested for histology, Western blotting, wire myography, and uniaxial tensile testing to examine their structural and functional properties. Differentiated hMSC tissue rings comprising 20% and 30% ECs exhibited significantly greater SMC contractile protein expression, endothelin-1 (ET-1)-meditated contraction, and force at failure compared with the 0% EC rings. On average, the 0%, 10%, 20%, and 30% EC rings exhibited a contractile force of 0.745 ± 0.117, 0.830 ± 0.358, 1.31 ± 0.353, and 1.67 ± 0.351 mN (mean ± standard deviation [SD]) in response to ET-1, respectively. Additionally, the mean maximum force at failure for the 0%, 10%, 20%, and 30% EC rings was 88.5 ± 36. , 121 ± 59.1, 147 ± 43.1, and 206 ± 0.8 mN (mean ± SD), respectively. Based on these results, 30% EC rings were fused together to form tissue-engineered blood vessels (TEBVs) and compared with 0% EC TEBV controls. The addition of 30% ECs in TEBVs did not affect ring fusion but did result in significantly greater SMC protein expression (calponin and smoothelin). In summary, co-seeding hMSCs with ECs to form tissue rings resulted in greater contraction, strength, and hMSC-SMC differentiation compared with hMSCs alone and indicates a method to create a functional 3D human vascular cell coculture model.
Collapse
Affiliation(s)
- William G DeMaria
- Department of Biomedical Engineering, Worcester Polytechnic Institute, Worcester, Massachusetts, USA
| | - Andre E Figueroa-Milla
- Department of Biomedical Engineering, Worcester Polytechnic Institute, Worcester, Massachusetts, USA
| | - Abigail Kaija
- MaineHealth Institute for Research, Scarborough, Maine, USA
| | | | - Benjamin Tero
- MaineHealth Institute for Research, Scarborough, Maine, USA
- The Roux Institute, Northeastern University, Portland, Maine, USA
| | - Larisa Ryzhova
- MaineHealth Institute for Research, Scarborough, Maine, USA
| | - Lucy Liaw
- MaineHealth Institute for Research, Scarborough, Maine, USA
| | - Marsha W Rolle
- Department of Biomedical Engineering, Worcester Polytechnic Institute, Worcester, Massachusetts, USA
- The Roux Institute, Northeastern University, Portland, Maine, USA
- Department of Chemical Engineering, Northeastern University, Boston, Massachusetts, USA
| |
Collapse
|
2
|
Shores KL, Truskey GA. Mechanotransduction of the vasculature in Hutchinson-Gilford Progeria Syndrome. Front Physiol 2024; 15:1464678. [PMID: 39239311 PMCID: PMC11374724 DOI: 10.3389/fphys.2024.1464678] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2024] [Accepted: 08/13/2024] [Indexed: 09/07/2024] Open
Abstract
Hutchinson-Gilford Progeria Syndrome (HGPS) is a premature aging disorder that causes severe cardiovascular disease, resulting in the death of patients in their teenage years. The disease pathology is caused by the accumulation of progerin, a mutated form of the nuclear lamina protein, lamin A. Progerin binds to the inner nuclear membrane, disrupting nuclear integrity, and causes severe nuclear abnormalities and changes in gene expression. This results in increased cellular inflammation, senescence, and overall dysfunction. The molecular mechanisms by which progerin induces the disease pathology are not fully understood. Progerin's detrimental impact on nuclear mechanics and the role of the nucleus as a mechanosensor suggests dysfunctional mechanotransduction could play a role in HGPS. This is especially relevant in cells exposed to dynamic, continuous mechanical stimuli, like those of the vasculature. The endothelial (ECs) and smooth muscle cells (SMCs) within arteries rely on physical forces produced by blood flow to maintain function and homeostasis. Certain regions within arteries produce disturbed flow, leading to an impaired transduction of mechanical signals, and a reduction in cellular function, which also occurs in HGPS. In this review, we discuss the mechanics of nuclear mechanotransduction, how this is disrupted in HGPS, and what effect this has on cell health and function. We also address healthy responses of ECs and SMCs to physiological mechanical stimuli and how these responses are impaired by progerin accumulation.
Collapse
Affiliation(s)
- Kevin L Shores
- Department of Biomedical Engineering, Duke University, Durham, NC, United States
| | - George A Truskey
- Department of Biomedical Engineering, Duke University, Durham, NC, United States
| |
Collapse
|
3
|
Gheorghe SR, Crăciun AM, Ilyés T, Tisa IB, Sur L, Lupan I, Samasca G, Silaghi CN. Converging Mechanisms of Vascular and Cartilaginous Calcification. BIOLOGY 2024; 13:565. [PMID: 39194503 DOI: 10.3390/biology13080565] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/29/2024] [Revised: 07/19/2024] [Accepted: 07/24/2024] [Indexed: 08/29/2024]
Abstract
Physiological calcification occurs in bones and epiphyseal cartilage as they grow, whereas ectopic calcification occurs in blood vessels, cartilage, and soft tissues. Although it was formerly thought to be a passive and degenerative process associated with aging, ectopic calcification has been identified as an active cell-mediated process resembling osteogenesis, and an increasing number of studies have provided evidence for this paradigm shift. A significant association between vascular calcification and cardiovascular risk has been demonstrated by various studies, which have shown that arterial calcification has predictive value for future coronary events. With respect to cartilaginous calcification, calcium phosphate or hydroxyapatite crystals can form asymptomatic deposits in joints or periarticular tissues, contributing to the pathophysiology of osteoarthritis, rheumatoid arthritis, ankylosing spondylitis, tendinitis, and bursitis. The risk factors and sequence of events that initiate ectopic calcification, as well as the mechanisms that prevent the development of this pathology, are still topics of debate. Consequently, in this review, we focus on the nexus of the mechanisms underlying vascular and cartilaginous calcifications, trying to circumscribe the similarities and disparities between them to provide more clarity in this regard.
Collapse
Affiliation(s)
- Simona R Gheorghe
- Department of Medical Biochemistry, Iuliu Hatieganu University of Medicine and Pharmacy, 400349 Cluj-Napoca, Romania
| | - Alexandra M Crăciun
- Department of Medical Biochemistry, Iuliu Hatieganu University of Medicine and Pharmacy, 400349 Cluj-Napoca, Romania
| | - Tamás Ilyés
- Department of Medical Biochemistry, Iuliu Hatieganu University of Medicine and Pharmacy, 400349 Cluj-Napoca, Romania
| | - Ioana Badiu Tisa
- Department of Pediatrics III, Iuliu Hatieganu University of Medicine and Pharmacy, 400217 Cluj-Napoca, Romania
| | - Lucia Sur
- Department of Pediatrics I, Iuliu Hatieganu University of Medicine and Pharmacy, 400370 Cluj-Napoca, Romania
| | - Iulia Lupan
- Department of Molecular Biology, Babes-Bolyai University, 400084 Cluj-Napoca, Romania
| | - Gabriel Samasca
- Department of Immunology, Iuliu Hatieganu University of Medicine and Pharmacy, 400162 Cluj-Napoca, Romania
| | - Ciprian N Silaghi
- Department of Medical Biochemistry, Iuliu Hatieganu University of Medicine and Pharmacy, 400349 Cluj-Napoca, Romania
| |
Collapse
|
4
|
Karkhaneh Yousefi AA, Petit C, Ben Hassine A, Avril S. Stiffness sensing by smooth muscle cells: Continuum mechanics modeling of the acto-myosin role. J Mech Behav Biomed Mater 2023; 144:105990. [PMID: 37385127 DOI: 10.1016/j.jmbbm.2023.105990] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Revised: 05/30/2023] [Accepted: 06/23/2023] [Indexed: 07/01/2023]
Abstract
Aortic smooth muscle cells (SMCs) play a vital role in maintaining homeostasis in the aorta by sensing and responding to mechanical stimuli. However, the mechanisms that underlie the ability of SMCs to sense and respond to stiffness change in their environment are still partially unclear. In this study, we focus on the role of acto-myosin contractility in stiffness sensing and introduce a novel continuum mechanics approach based on the principles of thermal strains. Each stress fiber satisfies a universal stress-strain relationship driven by a Young's modulus, a contraction coefficient scaling the fictitious thermal strain, a maximum contraction stress and a softening parameter describing the sliding effects between actin and myosin filaments. To account for the inherent variability of cellular responses, large populations of SMCs are modeled with the finite-element method, each cell having a random number and a random arrangement of stress fibers. Moreover, the level of myosin activation in each stress fiber satisfies a Weibull probability density function. Model predictions are compared to traction force measurements on different SMC lineages. It is demonstrated that the model not only predicts well the effects of substrate stiffness on cellular traction, but it can also successfully approximate the statistical variations of cellular tractions induced by intercellular variability. Finally, stresses in the nuclear envelope and in the nucleus are computed with the model, showing that the variations of cytoskeletal forces induced by substrate stiffness directly induce deformations of the nucleus which can potentially alter gene expression. The predictability of the model combined to its relative simplicity are promising assets for further investigation of stiffness sensing in 3D environments. Eventually, this could contribute to decipher the effects of mechanosensitivity impairment, which are known to be at the root of aortic aneurysms.
Collapse
Affiliation(s)
| | - Claudie Petit
- Mines Saint-Etienne, Université Jean Monnet, INSERM, U1059 SAINBIOSE, 42023, Saint-Etienne, France
| | - Amira Ben Hassine
- Mines Saint-Etienne, Université Jean Monnet, INSERM, U1059 SAINBIOSE, 42023, Saint-Etienne, France
| | - Stéphane Avril
- Mines Saint-Etienne, Université Jean Monnet, INSERM, U1059 SAINBIOSE, 42023, Saint-Etienne, France.
| |
Collapse
|
5
|
Salmasi MY, Alwis S, Cyclewala S, Jarral OA, Mohamed H, Mozalbat D, Nienaber CA, Athanasiou T, Morris-Rosendahl D. The genetic basis of thoracic aortic disease: The future of aneurysm classification? Hellenic J Cardiol 2023; 69:41-50. [PMID: 36202327 DOI: 10.1016/j.hjc.2022.09.009] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Revised: 09/24/2022] [Accepted: 09/25/2022] [Indexed: 11/04/2022] Open
Abstract
The expansion in the repertoire of genes linked to thoracic aortic aneurysms (TAA) has revolutionised our understanding of the disease process. The clinical benefits of such progress are numerous, particularly helping our understanding of non-syndromic hereditary causes of TAA (HTAAD) and further refinement in the subclassification of disease. Furthermore, the understanding of aortic biomechanics and mechanical homeostasis has been significantly informed by the discovery of deleterious mutations and their effect on aortic phenotype. The drawbacks in genetic testing in TAA lie with the inability to translate genotype to accurate prognostication in the risk of thoracic aortic dissection (TAD), which is a life-threatening condition. Under current guidelines, there are no metrics by which those at risk for dissection with normal aortic diameters may undergo preventive surgery. Future research lies with more advanced genetic diagnosis of HTAAD and investigation of the diverse pathways involved in its pathophysiology, which will i) serve to improve our understanding of the underlying mechanisms, ii) improve guidelines for treatment and iii) prevent complications for HTAAD and sporadic aortopathies.
Collapse
Affiliation(s)
| | | | | | - Omar A Jarral
- Department of Surgery and Cancer, Imperial College London, UK
| | - Heba Mohamed
- Royal Brompton and Harefield Foundation Trust, UK
| | | | | | | | | |
Collapse
|
6
|
Antonova L, Kutikhin A, Sevostianova V, Lobov A, Repkin E, Krivkina E, Velikanova E, Mironov A, Mukhamadiyarov R, Senokosova E, Khanova M, Shishkova D, Markova V, Barbarash L. Controlled and Synchronised Vascular Regeneration upon the Implantation of Iloprost- and Cationic Amphiphilic Drugs-Conjugated Tissue-Engineered Vascular Grafts into the Ovine Carotid Artery: A Proteomics-Empowered Study. Polymers (Basel) 2022; 14:polym14235149. [PMID: 36501545 PMCID: PMC9736446 DOI: 10.3390/polym14235149] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2022] [Revised: 11/17/2022] [Accepted: 11/24/2022] [Indexed: 11/30/2022] Open
Abstract
Implementation of small-diameter tissue-engineered vascular grafts (TEVGs) into clinical practice is still delayed due to the frequent complications, including thrombosis, aneurysms, neointimal hyperplasia, calcification, atherosclerosis, and infection. Here, we conjugated a vasodilator/platelet inhibitor, iloprost, and an antimicrobial cationic amphiphilic drug, 1,5-bis-(4-tetradecyl-1,4-diazoniabicyclo [2.2.2]octan-1-yl) pentane tetrabromide, to the luminal surface of electrospun poly(ε-caprolactone) (PCL) TEVGs for preventing thrombosis and infection, additionally enveloped such TEVGs into the PCL sheath to preclude aneurysms, and implanted PCLIlo/CAD TEVGs into the ovine carotid artery (n = 12) for 6 months. The primary patency was 50% (6/12 animals). TEVGs were completely replaced with the vascular tissue, free from aneurysms, calcification, atherosclerosis and infection, completely endothelialised, and had clearly distinguishable medial and adventitial layers. Comparative proteomic profiling of TEVGs and contralateral carotid arteries found that TEVGs lacked contractile vascular smooth muscle cell markers, basement membrane components, and proteins mediating antioxidant defense, concurrently showing the protein signatures of upregulated protein synthesis, folding and assembly, enhanced energy metabolism, and macrophage-driven inflammation. Collectively, these results suggested a synchronised replacement of PCL with a newly formed vascular tissue but insufficient compliance of PCLIlo/CAD TEVGs, demanding their testing in the muscular artery position or stimulation of vascular smooth muscle cell specification after the implantation.
Collapse
Affiliation(s)
- Larisa Antonova
- Department of Experimental Medicine, Research Institute for Complex Issues of Cardiovascular Diseases, 6 Sosnovy Boulevard, Kemerovo 650002, Russia
| | - Anton Kutikhin
- Department of Experimental Medicine, Research Institute for Complex Issues of Cardiovascular Diseases, 6 Sosnovy Boulevard, Kemerovo 650002, Russia
- Correspondence: ; Tel.: +7-9609077067
| | - Viktoriia Sevostianova
- Department of Experimental Medicine, Research Institute for Complex Issues of Cardiovascular Diseases, 6 Sosnovy Boulevard, Kemerovo 650002, Russia
| | - Arseniy Lobov
- Department of Regenerative Biomedicine, Research Institute of Cytology, 4 Tikhoretskiy Prospekt, Saint Petersburg 194064, Russia
| | - Egor Repkin
- Centre for Molecular and Cell Technologies, Saint Petersburg State University, Universitetskaya Embankment, 7/9, Saint Petersburg 199034, Russia
| | - Evgenia Krivkina
- Department of Experimental Medicine, Research Institute for Complex Issues of Cardiovascular Diseases, 6 Sosnovy Boulevard, Kemerovo 650002, Russia
| | - Elena Velikanova
- Department of Experimental Medicine, Research Institute for Complex Issues of Cardiovascular Diseases, 6 Sosnovy Boulevard, Kemerovo 650002, Russia
| | - Andrey Mironov
- Department of Experimental Medicine, Research Institute for Complex Issues of Cardiovascular Diseases, 6 Sosnovy Boulevard, Kemerovo 650002, Russia
| | - Rinat Mukhamadiyarov
- Department of Experimental Medicine, Research Institute for Complex Issues of Cardiovascular Diseases, 6 Sosnovy Boulevard, Kemerovo 650002, Russia
| | - Evgenia Senokosova
- Department of Experimental Medicine, Research Institute for Complex Issues of Cardiovascular Diseases, 6 Sosnovy Boulevard, Kemerovo 650002, Russia
| | - Mariam Khanova
- Department of Experimental Medicine, Research Institute for Complex Issues of Cardiovascular Diseases, 6 Sosnovy Boulevard, Kemerovo 650002, Russia
| | - Daria Shishkova
- Department of Experimental Medicine, Research Institute for Complex Issues of Cardiovascular Diseases, 6 Sosnovy Boulevard, Kemerovo 650002, Russia
| | - Victoria Markova
- Department of Experimental Medicine, Research Institute for Complex Issues of Cardiovascular Diseases, 6 Sosnovy Boulevard, Kemerovo 650002, Russia
| | - Leonid Barbarash
- Department of Experimental Medicine, Research Institute for Complex Issues of Cardiovascular Diseases, 6 Sosnovy Boulevard, Kemerovo 650002, Russia
| |
Collapse
|
7
|
Karakaya C, van Turnhout MC, Visser VL, Ristori T, Bouten CVC, Sahlgren CM, Loerakker S. Notch signaling regulates strain-mediated phenotypic switching of vascular smooth muscle cells. Front Cell Dev Biol 2022; 10:910503. [PMID: 36036000 PMCID: PMC9412035 DOI: 10.3389/fcell.2022.910503] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Accepted: 07/11/2022] [Indexed: 11/27/2022] Open
Abstract
Mechanical stimuli experienced by vascular smooth muscle cells (VSMCs) and mechanosensitive Notch signaling are important regulators of vascular growth and remodeling. However, the interplay between mechanical cues and Notch signaling, and its contribution to regulate the VSMC phenotype are still unclear. Here, we investigated the role of Notch signaling in regulating strain-mediated changes in VSMC phenotype. Synthetic and contractile VSMCs were cyclically stretched for 48 h to determine the temporal changes in phenotypic features. Different magnitudes of strain were applied to investigate its effect on Notch mechanosensitivity and the phenotypic regulation of VSMCs. In addition, Notch signaling was inhibited via DAPT treatment and activated with immobilized Jagged1 ligands to understand the role of Notch on strain-mediated phenotypic changes of VSMCs. Our data demonstrate that cyclic strain induces a decrease in Notch signaling along with a loss of VSMC contractile features. Accordingly, the activation of Notch signaling during cyclic stretching partially rescued the contractile features of VSMCs. These findings demonstrate that Notch signaling has an important role in regulating strain-mediated phenotypic switching of VSMCs.
Collapse
Affiliation(s)
- Cansu Karakaya
- Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, Netherlands
- Institute for Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, Netherlands
| | - Mark C. van Turnhout
- Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, Netherlands
| | - Valery L. Visser
- Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, Netherlands
| | - Tommaso Ristori
- Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, Netherlands
- Institute for Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, Netherlands
| | - Carlijn V. C. Bouten
- Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, Netherlands
- Institute for Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, Netherlands
| | - Cecilia M. Sahlgren
- Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, Netherlands
- Institute for Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, Netherlands
- Faculty of Science and Engineering, Åbo Akademi University, Turku, Finland
| | - Sandra Loerakker
- Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, Netherlands
- Institute for Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, Netherlands
- *Correspondence: Sandra Loerakker,
| |
Collapse
|
8
|
Karakaya C, van Asten JGM, Ristori T, Sahlgren CM, Loerakker S. Mechano-regulated cell-cell signaling in the context of cardiovascular tissue engineering. Biomech Model Mechanobiol 2022; 21:5-54. [PMID: 34613528 PMCID: PMC8807458 DOI: 10.1007/s10237-021-01521-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Accepted: 09/15/2021] [Indexed: 01/18/2023]
Abstract
Cardiovascular tissue engineering (CVTE) aims to create living tissues, with the ability to grow and remodel, as replacements for diseased blood vessels and heart valves. Despite promising results, the (long-term) functionality of these engineered tissues still needs improvement to reach broad clinical application. The functionality of native tissues is ensured by their specific mechanical properties directly arising from tissue organization. We therefore hypothesize that establishing a native-like tissue organization is vital to overcome the limitations of current CVTE approaches. To achieve this aim, a better understanding of the growth and remodeling (G&R) mechanisms of cardiovascular tissues is necessary. Cells are the main mediators of tissue G&R, and their behavior is strongly influenced by both mechanical stimuli and cell-cell signaling. An increasing number of signaling pathways has also been identified as mechanosensitive. As such, they may have a key underlying role in regulating the G&R of tissues in response to mechanical stimuli. A more detailed understanding of mechano-regulated cell-cell signaling may thus be crucial to advance CVTE, as it could inspire new methods to control tissue G&R and improve the organization and functionality of engineered tissues, thereby accelerating clinical translation. In this review, we discuss the organization and biomechanics of native cardiovascular tissues; recent CVTE studies emphasizing the obtained engineered tissue organization; and the interplay between mechanical stimuli, cell behavior, and cell-cell signaling. In addition, we review past contributions of computational models in understanding and predicting mechano-regulated tissue G&R and cell-cell signaling to highlight their potential role in future CVTE strategies.
Collapse
Affiliation(s)
- Cansu Karakaya
- Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, the Netherlands
- Institute for Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, the Netherlands
| | - Jordy G M van Asten
- Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, the Netherlands
- Institute for Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, the Netherlands
| | - Tommaso Ristori
- Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, the Netherlands
- Institute for Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, the Netherlands
- Department of Biomedical Engineering, Boston University, Boston, MA, USA
| | - Cecilia M Sahlgren
- Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, the Netherlands
- Institute for Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, the Netherlands
- Faculty of Science and Engineering, Biosciences, Åbo Akademi, Turku, Finland
| | - Sandra Loerakker
- Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, the Netherlands.
- Institute for Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, the Netherlands.
| |
Collapse
|
9
|
Regulation of SMC traction forces in human aortic thoracic aneurysms. Biomech Model Mechanobiol 2021; 20:717-731. [PMID: 33449277 PMCID: PMC7979631 DOI: 10.1007/s10237-020-01412-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2019] [Accepted: 12/12/2020] [Indexed: 01/03/2023]
Abstract
Smooth muscle cells (SMCs) usually express a contractile phenotype in the healthy aorta. However, aortic SMCs have the ability to undergo profound changes in phenotype in response to changes in their extracellular environment, as occurs in ascending thoracic aortic aneurysms (ATAA). Accordingly, there is a pressing need to quantify the mechanobiological effects of these changes at single cell level. To address this need, we applied Traction Force Microscopy (TFM) on 759 cells coming from three primary healthy (AoPrim) human SMC lineages and three primary aneurysmal (AnevPrim) human SMC lineages, from age and gender matched donors. We measured the basal traction forces applied by each of these cells onto compliant hydrogels of different stiffness (4, 8, 12, 25 kPa). Although the range of force generation by SMCs suggested some heterogeneity, we observed that: 1. the traction forces were significantly larger on substrates of larger stiffness; 2. traction forces in AnevPrim were significantly higher than in AoPrim cells. We modelled computationally the dynamic force generation process in SMCs using the motor-clutch model and found that it accounts well for the stiffness-dependent traction forces. The existence of larger traction forces in the AnevPrim SMCs were related to the larger size of cells in these lineages. We conclude that phenotype changes occurring in ATAA, which were previously known to reduce the expression of elongated and contractile SMCs (rendering SMCs less responsive to vasoactive agents), tend also to induce stronger SMCs. Future work aims at understanding the causes of this alteration process in aortic aneurysms.
Collapse
|
10
|
Laurent S, Boutouyrie P. Arterial Stiffness and Hypertension in the Elderly. Front Cardiovasc Med 2020; 7:544302. [PMID: 33330638 PMCID: PMC7673379 DOI: 10.3389/fcvm.2020.544302] [Citation(s) in RCA: 81] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2020] [Accepted: 09/17/2020] [Indexed: 12/21/2022] Open
Abstract
Hypertension prevalence increases with age. Age and high blood pressure are the two main determinants of arterial stiffness. In elderly hypertensives, large arteries stiffen and systolic and pulse pressures increase, due to wave reflections. A major reason for measuring arterial stiffness in clinical practice in elderly hypertensive patients comes from the repeated demonstration that arterial stiffness and wave reflections have a predictive value for CV events. A large body of evidence has been published during the last two decades, concerning the epidemiology, pathophysiology, and pharmacology of large arteries in hypertension in various settings of age. Particularly, two expert consensus documents have reviewed the methodological agreements for measuring arterial stiffness. The concepts of Early Vascular Aging (EVA) and Supernormal Vascular Aging (SUPERNOVA) help to better understand on which determinants of arterial stiffness it is possible to act, in order to limit target organ damage and cardiovascular complications. This review will address the issues of the cellular and molecular mechanisms of arterial stiffening in elderly hypertensives, the consequences of arterial stiffening on central systolic and pulse (systolic minus diastolic, PP) pressures and target organs, the methodology for measuring arterial stiffness, central pulse pressure and wave reflection, the epidemiological determinants of arterial stiffening in elderly hypertensives, the pharmacology of arterial destiffening, and how the concepts of EVA and SUPERNOVA apply to the detection of organ damage and prevention of CV complications.
Collapse
Affiliation(s)
- Stéphane Laurent
- Assistance-Publique Hôpitaux de Paris, Université de Paris, Paris, France
- PARCC-INSERM U970, Paris, France
- Department of Pharmacology and Hôpital Européen Georges Pompidou, Paris, France
| | - Pierre Boutouyrie
- Assistance-Publique Hôpitaux de Paris, Université de Paris, Paris, France
- PARCC-INSERM U970, Paris, France
- Department of Pharmacology and Hôpital Européen Georges Pompidou, Paris, France
| |
Collapse
|
11
|
Ristori T, Stassen OMJA, Sahlgren CM, Loerakker S. Lateral induction limits the impact of cell connectivity on Notch signaling in arterial walls. INTERNATIONAL JOURNAL FOR NUMERICAL METHODS IN BIOMEDICAL ENGINEERING 2020; 36:e3323. [PMID: 32058657 PMCID: PMC7217017 DOI: 10.1002/cnm.3323] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/20/2019] [Revised: 12/17/2019] [Accepted: 02/02/2020] [Indexed: 06/10/2023]
Abstract
It is well known that arteries grow and remodel in response to mechanical stimuli. Vascular smooth muscle cells are the main mediators of this process, as they can switch phenotype from contractile to synthetic, and vice-versa, based on the surrounding bio-chemo-mechanical stimuli. A correct regulation of this phenotypic switch is fundamental to obtain and maintain arterial homeostasis. Notch, a mechanosensitive signaling pathway, is one of the main regulators of the vascular smooth muscle cell phenotype. Therefore, understanding Notch dynamics is key to elucidate arterial growth, remodeling, and mechanobiology. We have recently developed a one-dimensional agent-based model to investigate Notch signaling in arteries. However, due to its one-dimensional formulation, the model cannot be adopted to study complex nonsymmetrical geometries and, importantly, it cannot capture the realistic "cell connectivity" in arteries, here defined as the number of cell neighbors. Notch functions via direct cell-cell contact; thus, the number of cell neighbors could be an essential feature of Notch dynamics. Here, we extended the agent-based model to a two-dimensional formulation, to investigate the effects of cell connectivity on Notch dynamics and cell phenotypes in arteries. The computational results, supported by a sensitivity analysis, indicate that cell connectivity has marginal effects when Notch dynamics is dominated by the process of lateral induction, which induces all cells to have a uniform phenotype. When lateral induction is weaker, cells exhibit a nonuniform phenotype distribution and the percentage of synthetic cells within an artery depends on the number of neighbors.
Collapse
Affiliation(s)
- Tommaso Ristori
- Department of Biomedical EngineeringEindhoven University of TechnologyEindhovenThe Netherlands
- Faculty of Science and Engineering, BiosciencesÅbo Academi UniversityTurkuFinland
- Institute for Complex Molecular SystemsEindhoven University of TechnologyEindhovenThe Netherlands
| | - Oscar M. J. A. Stassen
- Department of Biomedical EngineeringEindhoven University of TechnologyEindhovenThe Netherlands
- Faculty of Science and Engineering, BiosciencesÅbo Academi UniversityTurkuFinland
| | - Cecilia M. Sahlgren
- Department of Biomedical EngineeringEindhoven University of TechnologyEindhovenThe Netherlands
- Faculty of Science and Engineering, BiosciencesÅbo Academi UniversityTurkuFinland
- Institute for Complex Molecular SystemsEindhoven University of TechnologyEindhovenThe Netherlands
| | - Sandra Loerakker
- Department of Biomedical EngineeringEindhoven University of TechnologyEindhovenThe Netherlands
- Institute for Complex Molecular SystemsEindhoven University of TechnologyEindhovenThe Netherlands
| |
Collapse
|
12
|
Srivastava R, Rolyan H, Xie Y, Li N, Bhat N, Hong L, Esteghamat F, Adeniran A, Geirsson A, Zhang J, Ge G, Nobrega M, Martin KA, Mani A. TCF7L2 (Transcription Factor 7-Like 2) Regulation of GATA6 (GATA-Binding Protein 6)-Dependent and -Independent Vascular Smooth Muscle Cell Plasticity and Intimal Hyperplasia. Arterioscler Thromb Vasc Biol 2019; 39:250-262. [PMID: 30567484 PMCID: PMC6365015 DOI: 10.1161/atvbaha.118.311830] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Supplemental Digital Content is available in the text. Objective— TCF7L2 (transcription factor 7-like 2) is a Wnt-regulated transcription factor that maintains stemness and promotes proliferation in embryonic tissues and adult stem cells. Mice with a coronary artery disease–linked mutation in Wnt-coreceptor LRP6 (LDL receptor-related protein 6) exhibit vascular smooth muscle cell dedifferentiation and obstructive coronary artery disease, which are paradoxically associated with reduced TCF7L2 expression. We conducted a comprehensive study to explore the role of TCF7L2 in vascular smooth muscle cell differentiation and protection against intimal hyperplasia. Approach and Results— Using multiple mouse models, we demonstrate here that TCF7L2 promotes differentiation and inhibits proliferation of vascular smooth muscle cells. TCF7L2 accomplishes these effects by stabilization of GATA6 (GATA-binding protein 6) and upregulation of SM-MHC (smooth muscle cell myosin heavy chain) and cell cycle inhibitors. Accordingly, TCF7L2 haploinsufficient mice exhibited increased susceptibility to injury-induced hyperplasia, while mice overexpressing TCF7L2 were protected against injury-induced intimal hyperplasia compared with wild-type littermates. Consequently, the overexpression of TCF7L2 in LRP6 mutant mice rescued the injury-induced intimal hyperplasia. Conclusions— Our novel findings imply cell type-specific functional role of TCF7L2 and provide critical insight into mechanisms underlying the pathogenesis of intimal hyperplasia.
Collapse
Affiliation(s)
- Roshni Srivastava
- From the Yale Cardiovascular Research Center (R.S., H.R., Y.X., N.L., N,B., L.H., F.E., J.Z., G.G., K.A.M., A.M.), Yale School of Medicine, New Haven, CT
| | - Harshvardhan Rolyan
- From the Yale Cardiovascular Research Center (R.S., H.R., Y.X., N.L., N,B., L.H., F.E., J.Z., G.G., K.A.M., A.M.), Yale School of Medicine, New Haven, CT
| | - Yi Xie
- From the Yale Cardiovascular Research Center (R.S., H.R., Y.X., N.L., N,B., L.H., F.E., J.Z., G.G., K.A.M., A.M.), Yale School of Medicine, New Haven, CT
| | - Na Li
- From the Yale Cardiovascular Research Center (R.S., H.R., Y.X., N.L., N,B., L.H., F.E., J.Z., G.G., K.A.M., A.M.), Yale School of Medicine, New Haven, CT
| | - Neha Bhat
- From the Yale Cardiovascular Research Center (R.S., H.R., Y.X., N.L., N,B., L.H., F.E., J.Z., G.G., K.A.M., A.M.), Yale School of Medicine, New Haven, CT
| | - Lingjuan Hong
- From the Yale Cardiovascular Research Center (R.S., H.R., Y.X., N.L., N,B., L.H., F.E., J.Z., G.G., K.A.M., A.M.), Yale School of Medicine, New Haven, CT
| | - Fatemehsadat Esteghamat
- From the Yale Cardiovascular Research Center (R.S., H.R., Y.X., N.L., N,B., L.H., F.E., J.Z., G.G., K.A.M., A.M.), Yale School of Medicine, New Haven, CT
| | | | - Arnar Geirsson
- Department of Surgery (A.G.), Yale School of Medicine, New Haven, CT
| | - Jiasheng Zhang
- From the Yale Cardiovascular Research Center (R.S., H.R., Y.X., N.L., N,B., L.H., F.E., J.Z., G.G., K.A.M., A.M.), Yale School of Medicine, New Haven, CT
| | - Guanghao Ge
- From the Yale Cardiovascular Research Center (R.S., H.R., Y.X., N.L., N,B., L.H., F.E., J.Z., G.G., K.A.M., A.M.), Yale School of Medicine, New Haven, CT
| | - Marcelo Nobrega
- Department of Human Genetics, University of Chicago, IL (M.N.)
| | - Kathleen A Martin
- From the Yale Cardiovascular Research Center (R.S., H.R., Y.X., N.L., N,B., L.H., F.E., J.Z., G.G., K.A.M., A.M.), Yale School of Medicine, New Haven, CT
| | - Arya Mani
- From the Yale Cardiovascular Research Center (R.S., H.R., Y.X., N.L., N,B., L.H., F.E., J.Z., G.G., K.A.M., A.M.), Yale School of Medicine, New Haven, CT.,Department of Genetics (A.M.), Yale School of Medicine, New Haven, CT
| |
Collapse
|
13
|
Brown IAM, Diederich L, Good ME, DeLalio LJ, Murphy SA, Cortese-Krott MM, Hall JL, Le TH, Isakson BE. Vascular Smooth Muscle Remodeling in Conductive and Resistance Arteries in Hypertension. Arterioscler Thromb Vasc Biol 2019; 38:1969-1985. [PMID: 30354262 DOI: 10.1161/atvbaha.118.311229] [Citation(s) in RCA: 147] [Impact Index Per Article: 29.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Cardiovascular disease is a leading cause of death worldwide and accounts for >17.3 million deaths per year, with an estimated increase in incidence to 23.6 million by 2030. 1 Cardiovascular death represents 31% of all global deaths 2 -with stroke, heart attack, and ruptured aneurysms predominantly contributing to these high mortality rates. A key risk factor for cardiovascular disease is hypertension. Although treatment or reduction in hypertension can prevent the onset of cardiovascular events, existing therapies are only partially effective. A key pathological hallmark of hypertension is increased peripheral vascular resistance because of structural and functional changes in large (conductive) and small (resistance) arteries. In this review, we discuss the clinical implications of vascular remodeling, compare the differences between vascular smooth muscle cell remodeling in conductive and resistance arteries, discuss the genetic factors associated with vascular smooth muscle cell function in hypertensive patients, and provide a prospective assessment of current and future research and pharmacological targets for the treatment of hypertension.
Collapse
Affiliation(s)
- Isola A M Brown
- From the Robert M. Berne Cardiovascular Research Center (I.A.M.B., M.E.G., L.J.D., S.A.M., B.E.I.)
| | - Lukas Diederich
- Cardiovascular Research Laboratory, Division of Cardiology, Pneumology and Angiology, Medical Faculty, Heinrich Heine University, Dusseldorf, Germany (L.D., M.M.C.-K.)
| | - Miranda E Good
- From the Robert M. Berne Cardiovascular Research Center (I.A.M.B., M.E.G., L.J.D., S.A.M., B.E.I.)
| | - Leon J DeLalio
- From the Robert M. Berne Cardiovascular Research Center (I.A.M.B., M.E.G., L.J.D., S.A.M., B.E.I.).,Department of Pharmacology (L.J.D.)
| | - Sara A Murphy
- From the Robert M. Berne Cardiovascular Research Center (I.A.M.B., M.E.G., L.J.D., S.A.M., B.E.I.)
| | - Miriam M Cortese-Krott
- Cardiovascular Research Laboratory, Division of Cardiology, Pneumology and Angiology, Medical Faculty, Heinrich Heine University, Dusseldorf, Germany (L.D., M.M.C.-K.)
| | - Jennifer L Hall
- Lillehei Heart Institute (J.L.H.).,Division of Cardiology, Department of Medicine (J.L.H.), University of Minnesota, Minneapolis.,American Heart Association, Dallas, TX (J.L.H.)
| | - Thu H Le
- Division of Nephrology, Department of Medicine (T.H.L.)
| | - Brant E Isakson
- From the Robert M. Berne Cardiovascular Research Center (I.A.M.B., M.E.G., L.J.D., S.A.M., B.E.I.).,Department of Molecular Physiology and Biophysics (B.E.I.), University of Virginia School of Medicine, Charlottesville
| |
Collapse
|
14
|
Wang L, Nakamura F. Identification of Filamin A Mechanobinding Partner I: Smoothelin Specifically Interacts with the Filamin A Mechanosensitive Domain 21. Biochemistry 2019; 58:4726-4736. [PMID: 30990690 DOI: 10.1021/acs.biochem.9b00100] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Filamin A (FLNA) is a ubiquitously expressed actin cross-linking protein and a scaffold of numerous binding partners to regulate cell proliferation, migration, and survival. FLNA is a homodimer, and each subunit has an N-terminal actin-binding domain followed by 24 immunoglobulin-like repeats (R). FLNA mediates mechanotransduction by force-induced conformational changes of its cryptic integrin-binding site on R21. Here, we identified two novel FLNA-binding partners, smoothelins (SMTN A and B) and leucine zipper protein 1 (LUZP1), using stable isotope labeling by amino acids in cell culture (SILAC)-based proteomics followed by an in silico screening for proteins having a consensus FLNA-binding domain. We found that, although SMTN does not interact with full-length FLNA, it binds to FLNA variant 1 (FLNAvar-1) that exposes the cryptic CD cleft of R21. Point mutations on the C strand that disrupt the integrin binding also block the SMTN interaction. We identified FLNA-binding domains on SMTN using mutagenesis and used the mutant SMTN to investigate the role of the FLNA-SMTN interaction on the dynamics and localization of SMTN in living cells. Fluorescence recovery after photobleaching (FRAP) of GFP-labeled SMTN in living cells demonstrated that the non-FLNA-binding mutant SMTN diffuses faster than wild-type SMTN. Moreover, inhibition of Rho-kinase using Y27632 also increases the diffusion. These data demonstrated that SMTN specifically interacts with FLNAvar-1 and mechanically activated FLNA in cells. The companion report (Wang and Nakamura, 2019) describes the interactions of FLNA with the transcript of the LUZP1 gene.
Collapse
Affiliation(s)
- Lina Wang
- School of Pharmaceutical Science and Technology, Life Science Platform , Tianjin University , 92 Weijin Road , Nankai District, Tianjin 300072 , China
| | - Fumihiko Nakamura
- School of Pharmaceutical Science and Technology, Life Science Platform , Tianjin University , 92 Weijin Road , Nankai District, Tianjin 300072 , China
| |
Collapse
|
15
|
Keeley TP, Mann GE. Defining Physiological Normoxia for Improved Translation of Cell Physiology to Animal Models and Humans. Physiol Rev 2019; 99:161-234. [PMID: 30354965 DOI: 10.1152/physrev.00041.2017] [Citation(s) in RCA: 183] [Impact Index Per Article: 36.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
The extensive oxygen gradient between the air we breathe (Po2 ~21 kPa) and its ultimate distribution within mitochondria (as low as ~0.5-1 kPa) is testament to the efforts expended in limiting its inherent toxicity. It has long been recognized that cell culture undertaken under room air conditions falls short of replicating this protection in vitro. Despite this, difficulty in accurately determining the appropriate O2 levels in which to culture cells, coupled with a lack of the technology to replicate and maintain a physiological O2 environment in vitro, has hindered addressing this issue thus far. In this review, we aim to address the current understanding of tissue Po2 distribution in vivo and summarize the attempts made to replicate these conditions in vitro. The state-of-the-art techniques employed to accurately determine O2 levels, as well as the issues associated with reproducing physiological O2 levels in vitro, are also critically reviewed. We aim to provide the framework for researchers to undertake cell culture under O2 levels relevant to specific tissues and organs. We envisage that this review will facilitate a paradigm shift, enabling translation of findings under physiological conditions in vitro to disease pathology and the design of novel therapeutics.
Collapse
Affiliation(s)
- Thomas P Keeley
- King's British Heart Foundation Centre of Research Excellence, School of Cardiovascular Medicine and Sciences, Faculty of Life Sciences and Medicine, King's College London , London , United Kingdom
| | - Giovanni E Mann
- King's British Heart Foundation Centre of Research Excellence, School of Cardiovascular Medicine and Sciences, Faculty of Life Sciences and Medicine, King's College London , London , United Kingdom
| |
Collapse
|
16
|
Garoffolo G, Madonna R, de Caterina R, Pesce M. Cell based mechanosensing in vascular patho-biology: More than a simple go-with the flow. Vascul Pharmacol 2018; 111:7-14. [DOI: 10.1016/j.vph.2018.06.013] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2017] [Revised: 02/10/2018] [Accepted: 06/16/2018] [Indexed: 12/12/2022]
|
17
|
Katchunga P, Kaishusha D, Mwambusa R, Mubalama E. [Prevalence of the proximal aortic root dilatation and it relationship with left ventricular remodelling among South-Kivu Congolese patients: A cross sectional study]. Ann Cardiol Angeiol (Paris) 2018; 67:250-255. [PMID: 29909952 DOI: 10.1016/j.ancard.2018.03.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2017] [Accepted: 03/01/2018] [Indexed: 06/08/2023]
Abstract
AIM Studies of the influence of aortic root remodelling on the left ventricle are scarce in sub Saharan Africa even though this region has got high prevalence of arterial hypertension which is positively related to sinuses of Valsalva diameter. The present work aims to determine the frequency of the proximal aortic root enlargement and to evaluate its association with the left ventricle structure and function respectively among Congolese patients. METHOD Four hundreds and three (403) patients who realised transthoracic cardiac echography were recruited. The association between the proximal aortic root diameter and the left ventricular echocardiographic parameters was modelised in a multiple linear regression using the Stepwise method. RESULTS Among the 403 patients, 69.4% were hypertensive. A multivariate linear regression analysis showed correlations between the proximal aortic root diameter and the left ventricular mass (β=1.12; P=0.01), the left ventricular diastolic diameter (β=0.26, P=0.001) and the E/A ratio (β=-0.02; P<0.0001) respectively. The independent predictor of the proximal aortic root diameter were age (β=0.06, P=0.0006), duration of arterial hypertension (β=0.11, P=0.002) and diastolic blood pressure (β=0.03; P=0.02). The frequency of the proximal aortic root dilatation was 3.5%. CONCLUSION These results infancies the importance of including the proximal aortic root dilatation as specific infra clinic risk factor among Congolese.
Collapse
Affiliation(s)
- P Katchunga
- Clinique Saint-Luc-de-Bukavu, B.P 02, avenue des Martyrs, Bukavu, République démocratique du Congo; Observatoire des maladies non transmissibles, faculté de médecine, université Catholique de Bukavu, B.P 285, Bukavu, République démocratique du Congo.
| | - D Kaishusha
- Observatoire des maladies non transmissibles, faculté de médecine, université Catholique de Bukavu, B.P 285, Bukavu, République démocratique du Congo
| | - R Mwambusa
- Clinique Saint-Luc-de-Bukavu, B.P 02, avenue des Martyrs, Bukavu, République démocratique du Congo
| | - E Mubalama
- Clinique Saint-Luc-de-Bukavu, B.P 02, avenue des Martyrs, Bukavu, République démocratique du Congo
| |
Collapse
|
18
|
Cyclic Mechanical Stretch Up-regulates Hepatoma-Derived Growth Factor Expression in Cultured Rat Aortic Smooth Muscle Cells. Biosci Rep 2018; 38:BSR20171398. [PMID: 29467272 PMCID: PMC5857908 DOI: 10.1042/bsr20171398] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2017] [Revised: 02/14/2018] [Accepted: 02/21/2018] [Indexed: 11/19/2022] Open
Abstract
Hepatoma-derived growth factor (HDGF) is a potent mitogen for vascular smooth muscle cells (SMCs) during embryogenesis and injury repair of vessel walls. Whether mechanical stimuli modulate HDGF expression remains unknown. The present study aimed at investigating whether cyclic mechanical stretch plays a regulatory role in HDGF expression and regenerative cytokine production in aortic SMCs. A SMC cell line was grown on a silicone-based elastomer chamber with extracellular matrix coatings (either type I collagen or fibronectin) and received cyclic and uniaxial mechanical stretches with 10% deformation at frequency 1 Hz. Morphological observation showed that fibronectin coating provided better cell adhesion and spreading and that consecutive 6 h of cyclic mechanical stretch remarkably induced reorientation and realignment of SMCs. Western blotting detection demonstrated that continuous mechanical stimuli elicited up-regulation of HDGF and proliferative cell nuclear antigen, a cell proliferative marker. Signal kinetic profiling study indicated that cyclic mechanical stretch induced signaling activity in RhoA/ROCK and PI3K/Akt cascades. Kinase inhibition study further showed that blockade of PI3K activity suppressed the stretch-induced tumor necrosis factor-α (TNF-α), whereas RhoA/ROCK inhibition significantly blunted the interleukin-6 (IL-6) production and HDGF overexpression. Moreover, siRNA-mediated HDGF gene silencing significantly suppressed constitutive expression of IL-6, but not TNF-α, in SMCs. These findings support the role of HDGF in maintaining vascular expression of IL-6, which has been regarded a crucial regenerative factor for acute vascular injury. In conclusion, cyclic mechanical stretch may maintain constitutive expression of HDGF in vascular walls and be regarded an important biophysical regulator in vascular regeneration.
Collapse
|
19
|
Lacolley P, Regnault V, Segers P, Laurent S. Vascular Smooth Muscle Cells and Arterial Stiffening: Relevance in Development, Aging, and Disease. Physiol Rev 2017; 97:1555-1617. [DOI: 10.1152/physrev.00003.2017] [Citation(s) in RCA: 332] [Impact Index Per Article: 47.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2017] [Revised: 05/15/2017] [Accepted: 05/26/2017] [Indexed: 12/18/2022] Open
Abstract
The cushioning function of large arteries encompasses distension during systole and recoil during diastole which transforms pulsatile flow into a steady flow in the microcirculation. Arterial stiffness, the inverse of distensibility, has been implicated in various etiologies of chronic common and monogenic cardiovascular diseases and is a major cause of morbidity and mortality globally. The first components that contribute to arterial stiffening are extracellular matrix (ECM) proteins that support the mechanical load, while the second important components are vascular smooth muscle cells (VSMCs), which not only regulate actomyosin interactions for contraction but mediate also mechanotransduction in cell-ECM homeostasis. Eventually, VSMC plasticity and signaling in both conductance and resistance arteries are highly relevant to the physiology of normal and early vascular aging. This review summarizes current concepts of central pressure and tensile pulsatile circumferential stress as key mechanical determinants of arterial wall remodeling, cell-ECM interactions depending mainly on the architecture of cytoskeletal proteins and focal adhesion, the large/small arteries cross-talk that gives rise to target organ damage, and inflammatory pathways leading to calcification or atherosclerosis. We further speculate on the contribution of cellular stiffness along the arterial tree to vascular wall stiffness. In addition, this review provides the latest advances in the identification of gene variants affecting arterial stiffening. Now that important hemodynamic and molecular mechanisms of arterial stiffness have been elucidated, and the complex interplay between ECM, cells, and sensors identified, further research should study their potential to halt or to reverse the development of arterial stiffness.
Collapse
Affiliation(s)
- Patrick Lacolley
- INSERM, U1116, Vandœuvre-lès-Nancy, France; Université de Lorraine, Nancy, France; IBiTech-bioMMeda, Department of Electronics and Information Systems, Ghent University, Gent, Belgium; Department of Pharmacology, European Georges Pompidou Hospital, Assistance Publique Hôpitaux de Paris, France; PARCC INSERM, UMR 970, Paris, France; and University Paris Descartes, Paris, France
| | - Véronique Regnault
- INSERM, U1116, Vandœuvre-lès-Nancy, France; Université de Lorraine, Nancy, France; IBiTech-bioMMeda, Department of Electronics and Information Systems, Ghent University, Gent, Belgium; Department of Pharmacology, European Georges Pompidou Hospital, Assistance Publique Hôpitaux de Paris, France; PARCC INSERM, UMR 970, Paris, France; and University Paris Descartes, Paris, France
| | - Patrick Segers
- INSERM, U1116, Vandœuvre-lès-Nancy, France; Université de Lorraine, Nancy, France; IBiTech-bioMMeda, Department of Electronics and Information Systems, Ghent University, Gent, Belgium; Department of Pharmacology, European Georges Pompidou Hospital, Assistance Publique Hôpitaux de Paris, France; PARCC INSERM, UMR 970, Paris, France; and University Paris Descartes, Paris, France
| | - Stéphane Laurent
- INSERM, U1116, Vandœuvre-lès-Nancy, France; Université de Lorraine, Nancy, France; IBiTech-bioMMeda, Department of Electronics and Information Systems, Ghent University, Gent, Belgium; Department of Pharmacology, European Georges Pompidou Hospital, Assistance Publique Hôpitaux de Paris, France; PARCC INSERM, UMR 970, Paris, France; and University Paris Descartes, Paris, France
| |
Collapse
|
20
|
Jiang ZQ, Zhou YL, Chen X, Li LY, Liang SY, Lin S, Shu MQ. Different effects of neuropeptide Y on proliferation of vascular smooth muscle cells via regulation of Geminin. Mol Cell Biochem 2017; 433:205-211. [PMID: 28386846 DOI: 10.1007/s11010-017-3028-7] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2017] [Accepted: 04/01/2017] [Indexed: 12/19/2022]
Abstract
The proliferation-promoting effect of neuropeptide Y (NPY) always functions in low-serum-cultured vascular smooth muscle cells (VSMCs), and the phenotypic switch of VSMCs is regulated by concentrations of serum. Whether the property of the NPY proliferative effect in VSMCs relies on phenotype of VSMCs is unclear. We aimed to explore the role of NPY on proliferation of different VSMC phenotypes in the pathogenesis of atherosclerosis. By stimulating A10 cells with 200 nM NPY in 0.5 or 10% serum, 3H-thymidine and 5-ethynyl-2'-deoxyuridine (EdU) and CCK8 measurements were used to detect VSMC proliferation. RT-PCR and Flow cytometry were performed to detect the factors involved in different properties of the NPY proliferative effect in VSMCs. Instead of facilitating proliferation, NPY had no significant effect on the growth of VSMCs when cultured in 10% serum (VSMCs stayed at synthetic states). The underlying mechanism may be involved in down-regulation of Y1 receptor (P < 0.05 vs. Vehicle) and up-regulation of Geminin (P < 0.05 vs. Vehicle) in 10% serum-cultured VSMCs co-incubated with 200 nM NPY. Besides, modulation of Geminin was effectively blocked by the Y1 receptor antagonist. The stimulation of NPY on proliferation of VSMCs could be a double-edged sword in the development of atherosclerosis and thus provides new knowledge for therapy of atherosclerosis.
Collapse
Affiliation(s)
- Zhou-Qin Jiang
- Department of Cardiology, Southwest Hospital, Third Military Medical University, No. 30 Gaotanyan, Shapingba, Chongqing, 400038, China
| | - You-Li Zhou
- Department of Cardiology, Southwest Hospital, Third Military Medical University, No. 30 Gaotanyan, Shapingba, Chongqing, 400038, China
| | - Xia Chen
- Department of Cardiology, Southwest Hospital, Third Military Medical University, No. 30 Gaotanyan, Shapingba, Chongqing, 400038, China
| | - Lin-Yu Li
- Department of Cardiology, Southwest Hospital, Third Military Medical University, No. 30 Gaotanyan, Shapingba, Chongqing, 400038, China
| | - Shi-Yu Liang
- Department of Cardiology, Southwest Hospital, Third Military Medical University, No. 30 Gaotanyan, Shapingba, Chongqing, 400038, China
| | - Shu Lin
- Department of Cardiology, Southwest Hospital, Third Military Medical University, No. 30 Gaotanyan, Shapingba, Chongqing, 400038, China. .,School of Health Science, IIIawarra Health and Medical Research Institute, University of Wollongong, Wollongong, NSW, 2522, Australia.
| | - Mao-Qin Shu
- Department of Cardiology, Southwest Hospital, Third Military Medical University, No. 30 Gaotanyan, Shapingba, Chongqing, 400038, China.
| |
Collapse
|
21
|
Bredov DV, Evstifeeva AU. Role of mechano-dependent cell movements in the establishment of spatial organization of axial rudiments in Xenopus laevis embryos. Russ J Dev Biol 2017. [DOI: 10.1134/s1062360417010039] [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]
|
22
|
Lindesay G, Ragonnet C, Chimenti S, Villeneuve N, Vayssettes-Courchay C. Age and hypertension strongly induce aortic stiffening in rats at basal and matched blood pressure levels. Physiol Rep 2016; 4:4/10/e12805. [PMID: 27233301 PMCID: PMC4886171 DOI: 10.14814/phy2.12805] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2016] [Accepted: 04/30/2016] [Indexed: 12/31/2022] Open
Abstract
Age and hypertension are major causes of large artery remodeling and stiffening, a cardiovascular risk factor for heart and kidney damage. The aged spontaneously hypertensive rat (SHR) model is recognized for human cardiovascular pathology, but discrepancies appeared in studies of arterial stiffness. We performed experiments using a robust analysis via echo tracking in 20-week adult (n = 8) and 80-week-old SHR (n = 7), with age-matched normotensive Wistar Kyoto rats (WKY, n = 6;6) at basal and matched levels of blood pressure (BP). After anesthesia with pentobarbital, abdominal aortic diameter and pressure were recorded and BP was decreased by clonidine i.v. At basal BP, aortic pulse distension, compliance, and distensibility (AD) were reduced and stiffness index increased with age and hypertension and further altered with age + hypertension. When BP was adjusted in SHR to that of normotensive rats (130 mmHg), there was no difference between 20-week-old SHR and WKY Importantly, the age effect was maintained in both WKY and SHR and accentuated by hypertension in old rats. At 130 mmHg, with similar pulse pressure in the four groups, AD (kPa(-3)) = 24.2 ± 1 in 20 weeks WKY, 19.7 ± 1.4 in 20 weeks SHR, 12.4 ± 1.3 in 80 weeks WKY and 6.6 ± 0.6 in 80 weeks SHR; distension = 7.6 ± 0.4%, 6.7 ± 0.6%, 3.7 ± 0.3%, and 1.8 ± 0.2% in the same groups. In conclusion, reduced distensibility, that is, stiffening due to age is clearly shown here in both WKY and SHR as well as a synergistic effect of age and hypertension. This technique will allow new studies on the mechanisms responsible and drug intervention.
Collapse
Affiliation(s)
- George Lindesay
- Cardiovascular Discovery Research Unit, Servier Research Institute, Suresnes, France
| | - Christophe Ragonnet
- Cardiovascular Discovery Research Unit, Servier Research Institute, Suresnes, France
| | - Stefano Chimenti
- Cardiovascular Discovery Research Unit, Servier Research Institute, Suresnes, France
| | - Nicole Villeneuve
- Cardiovascular Discovery Research Unit, Servier Research Institute, Suresnes, France
| | | |
Collapse
|
23
|
Ferri N, Marchianò S, Tibolla G, Baetta R, Dhyani A, Ruscica M, Uboldi P, Catapano AL, Corsini A. PCSK9 knock-out mice are protected from neointimal formation in response to perivascular carotid collar placement. Atherosclerosis 2016; 253:214-224. [DOI: 10.1016/j.atherosclerosis.2016.07.910] [Citation(s) in RCA: 61] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/15/2016] [Revised: 07/15/2016] [Accepted: 07/20/2016] [Indexed: 12/25/2022]
|
24
|
Bono N, Pezzoli D, Levesque L, Loy C, Candiani G, Fiore GB, Mantovani D. Unraveling the role of mechanical stimulation on smooth muscle cells: A comparative study between 2D and 3D models. Biotechnol Bioeng 2016; 113:2254-63. [PMID: 26987444 DOI: 10.1002/bit.25979] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2015] [Revised: 03/01/2016] [Accepted: 03/09/2016] [Indexed: 01/08/2023]
Abstract
A thorough understanding of cell response to combined culture configuration and mechanical cues is of paramount importance in vascular tissue engineering applications. Herein, we investigated and compared the response of vascular smooth muscle cells (vSMCs) cultured in different culture environments (2D cell monolayers and 3D cellularized collagen-based gels) in combination with mechanical stimulation (7% uniaxial cyclic strain, 1 Hz) for 2 and 5 days. When cyclic strain was applied, two different responses, in terms of cell orientation and expression of contractile-phenotype proteins, were observed in 2D and 3D models. Specifically, in 2D configuration, cyclic strain caused ∼50% of cell population to align nearly perpendicular (80-90 degrees) to the strain direction, while not influencing the contractile-phenotype protein expression, as compared to the 2D static controls. Conversely, the application of uniaxial strain to 3D constructs induced a ∼60% cell alignment almost parallel (0-10 degrees) to the strain direction. Moreover, 3D mechanical stimulation applied for 5 days induced a twofold increase of SM α-actin level and a 14-fold increase of calponin expression as compared to 3D static controls. Altogether these findings provide a new insight into the potential to drive cell behavior by modulating the extracellular matrix and the biomechanical environment. Biotechnol. Bioeng. 2016;113: 2254-2263. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- N Bono
- μBS Lab, Department of Electronics, Information and Bioengineering, Politecnico di Milano, Milan, Italy.,Laboratory for Biomaterials and Bioengineering, Department of Min-Met-Materials and CHU de Québec Research Center, Laval University, Quebec City, QC, Canada
| | - D Pezzoli
- Laboratory for Biomaterials and Bioengineering, Department of Min-Met-Materials and CHU de Québec Research Center, Laval University, Quebec City, QC, Canada
| | - L Levesque
- Laboratory for Biomaterials and Bioengineering, Department of Min-Met-Materials and CHU de Québec Research Center, Laval University, Quebec City, QC, Canada
| | - C Loy
- Laboratory for Biomaterials and Bioengineering, Department of Min-Met-Materials and CHU de Québec Research Center, Laval University, Quebec City, QC, Canada
| | - G Candiani
- BioCell, Department of Chemistry, Materials and Chemical Engineering, Politecnico di Milano, Milan, Italy
| | - G B Fiore
- μBS Lab, Department of Electronics, Information and Bioengineering, Politecnico di Milano, Milan, Italy
| | - D Mantovani
- Laboratory for Biomaterials and Bioengineering, Department of Min-Met-Materials and CHU de Québec Research Center, Laval University, Quebec City, QC, Canada.
| |
Collapse
|
25
|
Abstract
Pathophysiological studies have extensively investigated the structural factor in hypertension, including large and small artery remodeling and functional changes. Here, we review the recent literature on the alterations in small and large arteries in hypertension. We discuss the possible mechanisms underlying these abnormalities and we explain how they accompany and often precede hypertension. Finally, we propose an integrated pathophysiological approach to better understand how the cross-talk between large and small artery changes interacts in pressure wave transmission, exaggerates cardiac, brain and kidney damage, and lead to cardiovascular and renal complications. We focus on patients with essential hypertension because this is the most prevalent form of hypertension, and describe other forms of hypertension only for contrasting their characteristics with those of uncomplicated essential hypertension.
Collapse
Affiliation(s)
- Stéphane Laurent
- From the Department of Pharmacology, Assistance Publique-Hôpitaux de Paris, Hôpital Européen Georges Pompidou, Paris, France (S.L., P.B.); Université Paris-Descartes, Paris, France (S.L., P.B.); and INSERM U 970, Paris, France (S.L., P.B.).
| | - Pierre Boutouyrie
- From the Department of Pharmacology, Assistance Publique-Hôpitaux de Paris, Hôpital Européen Georges Pompidou, Paris, France (S.L., P.B.); Université Paris-Descartes, Paris, France (S.L., P.B.); and INSERM U 970, Paris, France (S.L., P.B.)
| |
Collapse
|
26
|
Pires PW, Jackson WF, Dorrance AM. Regulation of myogenic tone and structure of parenchymal arterioles by hypertension and the mineralocorticoid receptor. Am J Physiol Heart Circ Physiol 2015; 309:H127-36. [PMID: 25910805 DOI: 10.1152/ajpheart.00168.2015] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/09/2015] [Accepted: 04/23/2015] [Indexed: 11/22/2022]
Abstract
Proper perfusion is vital for maintenance of neuronal homeostasis and brain function. Changes in the function and structure of cerebral parenchymal arterioles (PAs) could impair blood flow regulation and increase the risk of cerebrovascular diseases, including dementia and stroke. Hypertension alters the structure and function of large cerebral arteries, but its effects on PAs remain unknown. We hypothesized that hypertension increases myogenic tone and induces inward remodeling in PAs; we further proposed that antihypertensive therapy or mineralocorticoid receptor (MR) blockade would reverse the effects of hypertension. PAs from 18-wk-old stroke-prone spontaneously hypertensive rats (SHRSP) were isolated and cannulated in a pressure myograph. At 50-mmHg intraluminal pressure, PAs from SHRSP showed higher myogenic tone (%tone: 39.1 ± 1.9 vs. 28.7 ± 2.5%, P < 0.01) and smaller resting luminal diameter (34.7 ± 1.9 vs. 46.2 ± 2.4 μm, P < 0.01) than those from normotensive Wistar-Kyoto rats, through a mechanism that seems to require Ca(2+) influx through L-type voltage-gated Ca(2+) channels. PAs from SHRSP showed inward remodeling (luminal diameter at 60 mmHg: 55.2 ± 1.4 vs. 75.7 ± 5.1 μm, P < 0.01) and a paradoxical increase in distensibility and compliance. Treatment of SHRSP for 6 wk with antihypertensive therapy reduced PAs' myogenic tone, increased their resting luminal diameter, and prevented inward remodeling. In contrast, treatment of SHRSP for 6 wk with an MR antagonist did not reduce blood pressure or myogenic tone, but prevented inward remodeling. Thus, while hypertensive remodeling of PAs may involve the MR, myogenic tone seems to be independent of MR activity.
Collapse
Affiliation(s)
- Paulo W Pires
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, Michigan; and Department of Pharmacology, University of Nevada School of Medicine, Reno, Nevada
| | - William F Jackson
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, Michigan; and
| | - Anne M Dorrance
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, Michigan; and
| |
Collapse
|
27
|
Humphrey JD, Schwartz MA, Tellides G, Milewicz DM. Role of mechanotransduction in vascular biology: focus on thoracic aortic aneurysms and dissections. Circ Res 2015; 116:1448-61. [PMID: 25858068 PMCID: PMC4420625 DOI: 10.1161/circresaha.114.304936] [Citation(s) in RCA: 259] [Impact Index Per Article: 28.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Thoracic aortic diseases that involve progressive enlargement, acute dissection, or rupture are influenced by the hemodynamic loads and mechanical properties of the wall. We have only limited understanding, however, of the mechanobiological processes that lead to these potentially lethal conditions. Homeostasis requires that intramural cells sense their local chemomechanical environment and establish, maintain, remodel, or repair the extracellular matrix to provide suitable compliance and yet sufficient strength. Proper sensing, in turn, necessitates both receptors that connect the extracellular matrix to intracellular actomyosin filaments and signaling molecules that transmit the related information to the nucleus. Thoracic aortic aneurysms and dissections are associated with poorly controlled hypertension and mutations in genes for extracellular matrix constituents, membrane receptors, contractile proteins, and associated signaling molecules. This grouping of factors suggests that these thoracic diseases result, in part, from dysfunctional mechanosensing and mechanoregulation of the extracellular matrix by the intramural cells, which leads to a compromised structural integrity of the wall. Thus, improved understanding of the mechanobiology of aortic cells could lead to new therapeutic strategies for thoracic aortic aneurysms and dissections.
Collapse
MESH Headings
- Aortic Dissection/genetics
- Aortic Dissection/metabolism
- Aortic Dissection/pathology
- Aortic Dissection/physiopathology
- Aortic Dissection/therapy
- Animals
- Aorta, Thoracic/metabolism
- Aorta, Thoracic/pathology
- Aorta, Thoracic/physiopathology
- Aortic Aneurysm, Thoracic/genetics
- Aortic Aneurysm, Thoracic/metabolism
- Aortic Aneurysm, Thoracic/pathology
- Aortic Aneurysm, Thoracic/physiopathology
- Aortic Aneurysm, Thoracic/therapy
- Aortic Rupture/genetics
- Aortic Rupture/metabolism
- Aortic Rupture/pathology
- Aortic Rupture/physiopathology
- Aortic Rupture/therapy
- Biomechanical Phenomena
- Disease Progression
- Extracellular Matrix Proteins/genetics
- Extracellular Matrix Proteins/metabolism
- Genetic Predisposition to Disease
- Hemodynamics
- Humans
- Mechanotransduction, Cellular
- Phenotype
- Stress, Mechanical
Collapse
Affiliation(s)
- Jay D Humphrey
- From the Departments of Biomedical Engineering (J.D.H., M.A.S.), Medicine (Cardiology) (M.A.S.), Cell Biology (M.A.S.), and Surgery (G.T.), Yale University, New Haven, CT; and Department of Internal Medicine, University of Texas Health Science Center, Houston (D.M.M.)
| | - Martin A Schwartz
- From the Departments of Biomedical Engineering (J.D.H., M.A.S.), Medicine (Cardiology) (M.A.S.), Cell Biology (M.A.S.), and Surgery (G.T.), Yale University, New Haven, CT; and Department of Internal Medicine, University of Texas Health Science Center, Houston (D.M.M.)
| | - George Tellides
- From the Departments of Biomedical Engineering (J.D.H., M.A.S.), Medicine (Cardiology) (M.A.S.), Cell Biology (M.A.S.), and Surgery (G.T.), Yale University, New Haven, CT; and Department of Internal Medicine, University of Texas Health Science Center, Houston (D.M.M.)
| | - Dianna M Milewicz
- From the Departments of Biomedical Engineering (J.D.H., M.A.S.), Medicine (Cardiology) (M.A.S.), Cell Biology (M.A.S.), and Surgery (G.T.), Yale University, New Haven, CT; and Department of Internal Medicine, University of Texas Health Science Center, Houston (D.M.M.).
| |
Collapse
|
28
|
Huang AH, Niklason LE. Engineering of arteries in vitro. Cell Mol Life Sci 2014; 71:2103-18. [PMID: 24399290 PMCID: PMC4024341 DOI: 10.1007/s00018-013-1546-3] [Citation(s) in RCA: 77] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2013] [Revised: 12/17/2013] [Accepted: 12/18/2013] [Indexed: 12/20/2022]
Abstract
This review will focus on two elements that are essential for functional arterial regeneration in vitro: the mechanical environment and the bioreactors used for tissue growth. The importance of the mechanical environment to embryological development, vascular functionality, and vascular graft regeneration will be discussed. Bioreactors generate mechanical stimuli to simulate biomechanical environment of arterial system. This system has been used to reconstruct arterial grafts with appropriate mechanical strength for implantation by controlling the chemical and mechanical environments in which the grafts are grown. Bioreactors are powerful tools to study the effect of mechanical stimuli on extracellular matrix architecture and mechanical properties of engineered vessels. Hence, biomimetic systems enable us to optimize chemo-biomechanical culture conditions to regenerate engineered vessels with physiological properties similar to those of native arteries. In addition, this article reviews various bioreactors designed especially to apply axial loading to engineered arteries. This review will also introduce and examine different approaches and techniques that have been used to engineer biologically based vascular grafts, including collagen-based grafts, fibrin-gel grafts, cell sheet engineering, biodegradable polymers, and decellularization of native vessels.
Collapse
Affiliation(s)
- Angela H Huang
- Department of Biomedical Engineering, Yale University, New Haven, CT, 06511, USA,
| | | |
Collapse
|
29
|
Abstract
To date, over 65 mutations in the gene encoding Cx43 (connexin43) have been linked to the autosomal-dominant disease ODDD (oculodentodigital dysplasia). A subset of these patients experience bladder incontinence which could be due to underlying neurogenic deterioration or aberrant myogenic regulation. BSMCs (bladder smooth muscle cells) from wild-type and two Cx43 mutant lines (Cx43(G60S) and Cx43(I130T)) that mimic ODDD exhibit a significant reduction in total Cx43. Dye transfer studies revealed that the G60S mutant was a potent dominant-negative inhibitor of co-expressed Cx43, a property not equally shared by the I130T mutant. BSMCs from both mutant mouse strains were defective in their ability to contract, which is indicative of phenotype changes due to harbouring the Cx43 mutants. Upon stretching, Cx43 levels were significantly elevated in controls and mutants containing BSMCs, but the non-muscle myosin heavy chain A levels were only reduced in cells from control mice. Although the Cx43(G60S) mutant mice showed no difference in voided urine volume or frequency, the Cx43(I130T) mice voided less frequently. Thus, similar to the diversity of morbidities seen in ODDD patients, genetically modified mice also display mutation-specific changes in bladder function. Furthermore, although mutant mice have compromised smooth muscle contraction and response to stretch, overriding bladder defects in Cx43(I130T) mice are likely to be complemented by neurogenic changes.
Collapse
|
30
|
Diamantouros SE, Hurtado-Aguilar LG, Schmitz-Rode T, Mela P, Jockenhoevel S. Pulsatile perfusion bioreactor system for durability testing and compliance estimation of tissue engineered vascular grafts. Ann Biomed Eng 2013; 41:1979-89. [PMID: 23681651 DOI: 10.1007/s10439-013-0823-5] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2013] [Accepted: 05/02/2013] [Indexed: 10/26/2022]
Abstract
The aim of the study was to design, construct, and test a bioreactor for the conditioning of tissue-engineered vascular grafts under physiological pressure, flow, and environmental conditions and up to supra-physiological pulse frequencies (5 Hz) as the first step towards durability testing. The system also allows the calculation of the compliance of vascular grafts as an indicator of tissue development. The system relies on the combination of a pulse-free pump and a linear magnetic actuator applying pressure pulses with controllable profile and frequency. The compliance estimation is based on the accurate measurement of the vessel's diameter by means of an optical micrometre. Software-based interface enables the control of a magnetic actuator and data acquisition to monitor the conditions of the system. Porcine carotid arteries were tested in the bioreactor for up to 4 weeks at different pulse frequencies. The tissue was analysed by means of histology and immunohistochemistry. Physiological pressures (~80 and 120 mmHg for diastolic and systolic phase, respectively) were generated in the system at frequencies between 1 and 5 Hz. The environmental conditions within the bioreactor were monitored and online determination of the compliance of the arteries was achieved under sterile conditions. Conditioning of the grafts resulted in the abundant production of ECM proteins. In conclusion, we developed a bioreactor for the conditioning of tissue engineered vascular grafts under controlled pressure conditions. The system is suitable to perform durability tests at supra-physiological pulse rates and physiological pressure levels under continuous monitoring of environmental variables (pH, pO2, pCO2, and temperature) and compliance.
Collapse
Affiliation(s)
- Stefanos E Diamantouros
- Department of Tissue Engineering & Textile Implants, AME-Helmholtz Institute for Biomedical Engineering, Aachen University, Pauwelsstrasse 20, 52074, Aachen, Germany
| | | | | | | | | |
Collapse
|
31
|
Abstract
Vascular smooth muscle cells have attracted considerable interest as a model for a flexible program of gene expression. This cell type arises throughout the embryo body plan via poorly understood signaling cascades that direct the expression of transcription factors and microRNAs which, in turn, orchestrate the activation of contractile genes collectively defining this cell lineage. The discovery of myocardin and its close association with serum response factor has represented a major break-through for the molecular understanding of vascular smooth muscle cell differentiation. Retinoids have been shown to improve the outcome of vessel wall remodeling following injury and have provided further insights into the molecular circuitry that defines the vascular smooth muscle cell phenotype. This review summarizes the progress to date in each of these areas of vascular smooth muscle cell biology.
Collapse
|
32
|
Downregulation of miR-223 and miR-153 mediates mechanical stretch-stimulated proliferation of venous smooth muscle cells via activation of the insulin-like growth factor-1 receptor. Arch Biochem Biophys 2012; 528:204-11. [PMID: 23046980 DOI: 10.1016/j.abb.2012.08.015] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2012] [Revised: 08/15/2012] [Accepted: 08/30/2012] [Indexed: 11/21/2022]
Abstract
Autologous venous grafts, used to circumvent occluded coronary arteries during coronary artery bypass, often develop thrombosis and neointimal hyperplasia. During neointimal hyperplasia, vascular smooth muscle cells (VSMCs), exposed to substantially higher pressure and hemodynamic forces, proliferate and extracellular matrix accumulate causing narrowing of the vessel lumen. Activation of insulin-like growth factor-1 receptor (IGF-1R) has been confirmed to be critically involved in mechanical stretch-stimulated VSMC proliferation. However, the comprehensive mechanisms responsible for activation of IGF-1R in VSMCs by mechanical stretch remain unclear. This study found that miR-223 and miR-153, targeted to IGF-1R, were down-regulated in VSMCs under stretch stress by miRNA microarray analysis in conjunction with Target Scan analysis. Overexpression of miR-223 or miR-153 down-regulated IGF-1R expression and activity in VSMCs under stretch stress. Specifically, overexpression of miR-223 and miR-153 inhibited stretch stress-enhanced VSMC proliferation and the activity of PI3K-AKT signaling. In conclusion, our study indicates that miR-153 and miR-223 are reduced in VSMCs by stretch stress, contributing to IGF-1R activation and resultant VSMC proliferation. Thus, miR-153 and miR-223 may be viable therapeutic targets for mechanical stretch-induced neointimal hyperplasia in vein grafts.
Collapse
|
33
|
Menon A, Eddinger TJ, Wang H, Wendell DC, Toth JM, LaDisa JF. Altered hemodynamics, endothelial function, and protein expression occur with aortic coarctation and persist after repair. Am J Physiol Heart Circ Physiol 2012; 303:H1304-18. [PMID: 23023871 DOI: 10.1152/ajpheart.00420.2012] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Coarctation of the aorta (CoA) is associated with substantial morbidity despite treatment. Mechanically induced structural and functional vascular changes are implicated; however, their relationship with smooth muscle (SM) phenotypic expression is not fully understood. Using a clinically representative rabbit model of CoA and correction, we quantified mechanical alterations from a 20-mmHg blood pressure (BP) gradient in the thoracic aorta and related the expression of key SM contractile and focal adhesion proteins with remodeling, relaxation, and stiffness. Systolic and mean BP were elevated for CoA rabbits compared with controls leading to remodeling, stiffening, an altered force response, and endothelial dysfunction both proximally and distally. The proximal changes persisted for corrected rabbits despite >12 wk of normal BP (~4 human years). Computational fluid dynamic simulations revealed reduced wall shear stress (WSS) proximally in CoA compared with control and corrected rabbits. Distally, WSS was markedly increased in CoA rabbits due to a stenotic velocity jet, which has persistent effects as WSS was significantly reduced in corrected rabbits. Immunohistochemistry revealed significantly increased nonmuscle myosin and reduced SM myosin heavy chain expression in the proximal arteries of CoA and corrected rabbits but no differences in SM α-actin, talin, or fibronectin. These findings indicate that CoA can cause alterations in the SM phenotype contributing to structural and functional changes in the proximal arteries that accompany the mechanical stimuli of elevated BP and altered WSS. Importantly, these changes are not reversed upon BP correction and may serve as markers of disease severity, which explains the persistent morbidity observed in CoA patients.
Collapse
Affiliation(s)
- Arjun Menon
- Department of Biomedical Engineering, Marquette University, Milwaukee, WI, USA
| | | | | | | | | | | |
Collapse
|
34
|
Turczyńska KM, Sadegh MK, Hellstrand P, Swärd K, Albinsson S. MicroRNAs are essential for stretch-induced vascular smooth muscle contractile differentiation via microRNA (miR)-145-dependent expression of L-type calcium channels. J Biol Chem 2012; 287:19199-206. [PMID: 22474293 PMCID: PMC3365952 DOI: 10.1074/jbc.m112.341073] [Citation(s) in RCA: 55] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2012] [Revised: 03/26/2012] [Indexed: 11/06/2022] Open
Abstract
Stretch of the vascular wall is an important stimulus to maintain smooth muscle contractile differentiation that is known to depend on L-type calcium influx, Rho-activation, and actin polymerization. The role of microRNAs in this response was investigated using tamoxifen-inducible and smooth muscle-specific Dicer KO mice. In the absence of Dicer, which is required for microRNA maturation, smooth muscle microRNAs were completely ablated. Stretch-induced contractile differentiation and Rho-dependent cofilin-2 phosphorylation were dramatically reduced in Dicer KO vessels. On the other hand, acute stretch-sensitive growth signaling, which is independent of influx through L-type calcium channels, was not affected by Dicer KO. Contractile differentiation induced by the actin polymerizing agent jasplakinolide was not altered by deletion of Dicer, suggesting an effect upstream of actin polymerization. Basal and stretch-induced L-type calcium channel expressions were both decreased in Dicer KO portal veins, and inhibition of L-type channels in control vessels mimicked the effects of Dicer deletion. Furthermore, inhibition of miR-145, a highly expressed microRNA in smooth muscle, resulted in a similar reduction of L-type calcium channel expression. This was abolished by the Ca(2+)/calmodulin-dependent protein kinase II inhibitor KN93, suggesting that Ca(2+)/calmodulin-dependent protein kinase IIδ, a target of miR-145 and up-regulated in Dicer KO, plays a role in the regulation of L-type channel expression. These results show that microRNAs play a crucial role in stretch-induced contractile differentiation in the vascular wall in part via miR-145-dependent regulation of L-type calcium channels.
Collapse
MESH Headings
- Animals
- Calcium Channels, L-Type/biosynthesis
- Calcium Channels, L-Type/genetics
- Calcium Signaling/drug effects
- Calcium Signaling/physiology
- Calcium-Calmodulin-Dependent Protein Kinase Type 2/genetics
- Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism
- Cells, Cultured
- DEAD-box RNA Helicases/genetics
- DEAD-box RNA Helicases/metabolism
- Male
- Mice
- Mice, Knockout
- MicroRNAs/genetics
- MicroRNAs/metabolism
- Muscle Proteins/genetics
- Muscle Proteins/metabolism
- Muscle, Smooth, Vascular/cytology
- Muscle, Smooth, Vascular/metabolism
- Myocytes, Smooth Muscle/cytology
- Myocytes, Smooth Muscle/metabolism
- Protein Kinase Inhibitors/pharmacology
- Ribonuclease III/genetics
- Ribonuclease III/metabolism
- Up-Regulation/drug effects
- Up-Regulation/physiology
Collapse
Affiliation(s)
- Karolina M. Turczyńska
- From the Department of Experimental Medical Science, Lund University, 221 84 Lund, Sweden
| | | | - Per Hellstrand
- From the Department of Experimental Medical Science, Lund University, 221 84 Lund, Sweden
| | - Karl Swärd
- From the Department of Experimental Medical Science, Lund University, 221 84 Lund, Sweden
| | - Sebastian Albinsson
- From the Department of Experimental Medical Science, Lund University, 221 84 Lund, Sweden
| |
Collapse
|
35
|
Karam JP, Muscari C, Montero-Menei CN. Combining adult stem cells and polymeric devices for tissue engineering in infarcted myocardium. Biomaterials 2012; 33:5683-95. [PMID: 22594970 DOI: 10.1016/j.biomaterials.2012.04.028] [Citation(s) in RCA: 81] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2012] [Accepted: 04/08/2012] [Indexed: 12/18/2022]
Abstract
An increasing number of studies in cardiac cell therapy have provided encouraging results for cardiac repair. Adult stem cells may overcome ethical and availability concerns, with the additional advantages, in some cases, to allow autologous grafts to be performed. However, the major problems of cell survival, cell fate determination and engraftment after transplantation, still remain. Tissue-engineering strategies combining scaffolds and cells have been developed and have to be adapted for each type of application to enhance stem cell function. Scaffold properties required for cardiac cell therapy are here discussed. New tissue engineering advances that may be implemented in combination with adult stem cells for myocardial infarction therapy are also presented. Biomaterials not only provide a 3D support for the cells but may also mimic the structural architecture of the heart. Using hydrogels or particulate systems, the biophysical and biochemical microenvironments of transplanted cells can also be controlled. Advances in biomaterial engineering have permitted the development of sophisticated drug-releasing materials with a biomimetic 3D support that allow a better control of the microenvironment of transplanted cells.
Collapse
|
36
|
Li X, Chu JS, Yang L, Li S. Anisotropic effects of mechanical strain on neural crest stem cells. Ann Biomed Eng 2012; 40:598-605. [PMID: 22109803 DOI: 10.1007/s10439-011-0403-5] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2011] [Accepted: 09/09/2011] [Indexed: 10/15/2022]
Abstract
Neural crest stem cells (NCSCs) are multipotent and play an important role during the development and tissue regeneration. However, the anisotropic effects of mechanical strain on NCSCs are not known. To investigate the anisotropic mechanosensing by NCSCs, NCSCs derived from induced pluripotent stem cells were cultured on micropatterned membranes, and subjected to cyclic uniaxial strain in the direction parallel or perpendicular to the microgrooves. Cell and nuclear shape were both regulated by micropatterning and mechanical strain. Among the unpatterned, parallel-patterned and perpendicular-patterned groups, mechanical strain caused an increase in histone deacetylase activity in the parallel-patterned group, accompanied by the increase of cell proliferation. In addition, mechanical strain increased the expression of contractile marker calponin-1 but not other differentiation markers in the unpatterned and parallel-patterned groups. These results demonstrated that NCSCs responded differently to the anisotropic mechanical environment. Understanding the mechanical regulation of NCSCs will reveal the role of mechanical factors in NCSC differentiation during development, and provide a basis for using NCSCs for tissue engineering.
Collapse
Affiliation(s)
- Xian Li
- 111 Project Laboratory of Biomechanics and Tissue Repair, Bioengineering College, Chongqing University, Chongqing 400044, China.
| | | | | | | |
Collapse
|
37
|
The primary cilium as a dual sensor of mechanochemical signals in chondrocytes. Cell Mol Life Sci 2012; 69:2101-7. [PMID: 22241332 PMCID: PMC3375420 DOI: 10.1007/s00018-011-0911-3] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2011] [Revised: 12/18/2011] [Accepted: 12/21/2011] [Indexed: 01/17/2023]
Abstract
The primary cilium is an immotile, solitary, and microtubule-based structure that projects from cell surfaces into the extracellular environment. The primary cilium functions as a dual sensor, as mechanosensors and chemosensors. The primary cilia coordinate several essential cell signaling pathways that are mainly involved in cell division and differentiation. A primary cilium malfunction can result in several human diseases. Mechanical loading is sense by mechanosensitive cells in nearly all tissues and organs. With this sensation, the mechanical signal is further transduced into biochemical signals involving pathways such as Akt, PKA, FAK, ERK, and MAPK. In this review, we focus on the fundamental functional and structural features of primary cilia in chondrocytes and chondrogenic cells.
Collapse
|
38
|
Takemoto K, Mizutani T, Tamura K, Takeda K, Haga H, Kawabata K. The Number of Cyclic Stretch Regulates Cellular Elasticity in C2C12 Myoblasts. Cell 2012. [DOI: 10.4236/cellbio.2012.11001] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
39
|
Li X, Chu J, Wang A, Zhu Y, Chu WK, Yang L, Li S. Uniaxial mechanical strain modulates the differentiation of neural crest stem cells into smooth muscle lineage on micropatterned surfaces. PLoS One 2011; 6:e26029. [PMID: 22016804 PMCID: PMC3189240 DOI: 10.1371/journal.pone.0026029] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2011] [Accepted: 09/15/2011] [Indexed: 11/19/2022] Open
Abstract
Neural crest stem cells (NCSCs) play an important role in the development and represent a valuable cell source for tissue engineering. However, how mechanical factors in vivo regulate NCSC differentiation is not understood. Here NCSCs were derived from induced pluripotent stem cells and used as a model to determine whether vascular mechanical strain modulates the differentiation of NCSCs into smooth muscle (SM) lineage. NCSCs were cultured on micropatterned membranes to mimic the organization of smooth muscle cells (SMCs), and subjected to cyclic uniaxial strain. Mechanical strain enhanced NCSC proliferation and ERK2 phosphorylation. In addition, mechanical strain induced contractile marker calponin-1 within 2 days and slightly induced SM myosin within 5 days. On the other hand, mechanical strain suppressed the differentiation of NCSCs into Schwann cells. The induction of calponin-1 by mechanical strain was inhibited by neural induction medium but further enhanced by TGF-β. For NCSCs pre-treated with TGF-β, mechanical strain induced the gene expression of both calponin-1 and SM myosin. Our results demonstrated that mechanical strain regulates the differentiation of NCSCs in a manner dependent on biochemical factors and the differentiation stage of NCSCs. Understanding the mechanical regulation of NCSC differentiation will shed light on the development and remodeling of vascular tissues, and how transplanted NCSCs respond to mechanical factors.
Collapse
Affiliation(s)
- Xian Li
- Bioengineering College, Chongqing University, Chongqing, China
- Department of Bioengineering, University of California, Berkeley, California, United States of America
| | - Julia Chu
- Department of Bioengineering, University of California, Berkeley, California, United States of America
| | - Aijun Wang
- Department of Bioengineering, University of California, Berkeley, California, United States of America
| | - Yiqian Zhu
- Department of Bioengineering, University of California, Berkeley, California, United States of America
| | - Wai Keung Chu
- Department of Bioengineering, University of California San Diego, La Jolla, California, United States of America
| | - Li Yang
- Bioengineering College, Chongqing University, Chongqing, China
| | - Song Li
- Department of Bioengineering, University of California, Berkeley, California, United States of America
| |
Collapse
|
40
|
Rensen S, Doevendans P, van Eys G. Regulation and characteristics of vascular smooth muscle cell phenotypic diversity. Neth Heart J 2011; 15:100-8. [PMID: 17612668 PMCID: PMC1847757 DOI: 10.1007/bf03085963] [Citation(s) in RCA: 648] [Impact Index Per Article: 49.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Vascular smooth muscle cells can perform both contractile and synthetic functions, which are associated with and characterised by changes in morphology, proliferation and migration rates, and the expression of different marker proteins. The resulting phenotypic diversity of smooth muscle cells appears to be a function of innate genetic programmes and environmental cues, which include biochemical factors, extracellular matrix components, and physical factors such as stretch and shear stress. Because of the diversity among smooth muscle cells, blood vessels attain the flexibility that is necessary to perform efficiently under different physiological and pathological conditions. In this review, we discuss recent literature demonstrating the extent and nature of smooth muscle cell diversity in the vascular wall and address the factors that affect smooth muscle cell phenotype. (Neth Heart J 2007;15:100-8.).
Collapse
Affiliation(s)
- S.S.M. Rensen
- Department of Genetics and Cell Biology, Cardiovascular Research Institute Maastricht, University of Maastricht, the Netherlands
| | - P.A.F.M. Doevendans
- Department of Cardiology, Heart Lung Centre Utrecht, Interuniversity Cardiology Institute, the Netherlands
| | - G.J.J.M. van Eys
- Department of Genetics and Cell Biology, Cardiovascular Research Institute Maastricht, University of Maastricht, the Netherlands
| |
Collapse
|
41
|
Sharifpoor S, Simmons CA, Labow RS, Paul Santerre J. Functional characterization of human coronary artery smooth muscle cells under cyclic mechanical strain in a degradable polyurethane scaffold. Biomaterials 2011; 32:4816-29. [PMID: 21463894 DOI: 10.1016/j.biomaterials.2011.03.034] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2011] [Accepted: 03/12/2011] [Indexed: 10/18/2022]
Abstract
There are few synthetic elastomeric biomaterials that simultaneously provide the required biological conditioning and the ability to translate biomechanical stimuli to vascular smooth muscle cells (VSMCs). Biomechanical stresses are important physiological elements that regulate VSMC function, and polyurethane elastomers are a class of materials capable of facilitating the translation of stress induced biomechanics. In this study, human coronary artery smooth muscle cells (hCASMCs), which were seeded into a porous degradable polar/hydrophobic/ionic (D-PHI) polyurethane scaffold, were subjected to uniaxial cyclic mechanical strain (CMS) over a span of four weeks using a customized bioreactor. The distribution, proliferation and contractile protein expression of hCASMCs in the scaffold were then analyzed and compared to those grown under static conditions. Four weeks of CMS, applied to the elastomeric scaffold, resulted in statistically greater DNA mass, more cell area coverage and a better distribution of cells deeper within the scaffold construct. Furthermore, CMS samples demonstrated improved tensile mechanical properties following four weeks of culture, suggesting the generation of more extracellular matrix within the polyurethane constructs. The expression of smooth muscle α-actin, calponin and smooth muscle myosin heavy chain and the absence of Ki-67+ cells in both static and CMS cultures, throughout the 4 weeks, suggest that hCASMCs retained their contractile character on these biomaterials. The study highlights the importance of implementing physiologically-relevant biomechanical stimuli in the development of synthetic elastomeric tissue engineering scaffolds.
Collapse
Affiliation(s)
- Soroor Sharifpoor
- University of Toronto, Institute of Biomaterials and Biomedical Engineering, Faculty of Dentistry, 124 Edward Street, Toronto, Ontario, Canada M5G1G6
| | | | | | | |
Collapse
|
42
|
Desai LP, Wu Y, Tepper RS, Gunst SJ. Mechanical stimuli and IL-13 interact at integrin adhesion complexes to regulate expression of smooth muscle myosin heavy chain in airway smooth muscle tissue. Am J Physiol Lung Cell Mol Physiol 2011; 301:L275-84. [PMID: 21642449 DOI: 10.1152/ajplung.00043.2011] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Airway smooth muscle phenotype may be modulated in response to external stimuli under physiological and pathophysiological conditions. The effect of mechanical forces on airway smooth muscle phenotype were evaluated in vitro by suspending weights of 0.5 or 1 g from the ends of canine tracheal smooth muscle tissues, incubating the weighted tissues for 6 h, and then measuring the expression of the phenotypic marker protein, smooth muscle myosin heavy chain (SmMHC). Incubation of the tissues at a high load significantly increased expression of SmMHC compared with incubation at low load. Incubation of the tissues at a high load also decreased activation of PKB/Akt, as indicated by its phosphorylation at Ser 473. Inhibition of Akt or phosphatidylinositol-3,4,5 triphosphate-kinase increased SmMHC expression in tissues at low load but did not affect SmMHC expression at high load. IL-13 induced a significant increase in Akt activation and suppressed the expression of SmMHC protein at both low and high loads. The role of integrin signaling in mechanotransduction was evaluated by expressing a PINCH (LIM1-2) fragment in the muscle tissues that prevents the membrane localization of the integrin-binding IPP complex (ILK/PINCH/α-parvin), and also by expressing an inactive integrin-linked kinase mutant (ILK S343A) that inhibits endogenous ILK activity. Both mutants inhibited Akt activation and increased expression of SmMHC protein at low load but had no effect at high load. These results suggest that mechanical stress and IL-13 both act through an integrin-mediated signaling pathway to oppositely regulate the expression of phenotypic marker proteins in intact airway smooth muscle tissues. The stimulatory effects of mechanical stress on contractile protein expression oppose the suppression of contractile protein expression mediated by IL-13; thus the imposition of mechanical strain may inhibit changes in airway smooth muscle phenotype induced by inflammatory mediators.
Collapse
Affiliation(s)
- Leena P Desai
- Dept. of Cellular & Integrative Physiology, Indiana Univ. School of Medicine, Indianapolis, IN 46202, USA
| | | | | | | |
Collapse
|
43
|
Rapid Engineered Small Diameter Vascular Grafts from Smooth Muscle Cells. Cardiovasc Eng Technol 2011. [DOI: 10.1007/s13239-011-0044-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
|
44
|
Schutte SC, Chen Z, Brockbank KGM, Nerem RM. Cyclic strain improves strength and function of a collagen-based tissue-engineered vascular media. Tissue Eng Part A 2011; 16:3149-57. [PMID: 20504073 DOI: 10.1089/ten.tea.2010.0009] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Tissue-engineered blood vessels may provide a solution to the lack of suitable blood vessels for coronary and peripheral vessel bypass grafting. Cyclic strain can be used to provide a more physiological environment that may result in tissue that more closely resembles native artery. In this study, cyclic strain is applied to a collagen-based, tissue-engineered vascular medium. An increased culture time was used to allow the tissue to adhere to the silastic sleeve and to eliminate longitudinal compaction. Cyclic strain improved tissue strength through increased collagen content as well as some radial tissue compaction. Mechanical stimulation promoted a more contractile phenotype and led to a greater contractile response to the vasoconstrictor endothelin-1.
Collapse
Affiliation(s)
- Stacey C Schutte
- Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, Georgia 30332, USA
| | | | | | | |
Collapse
|
45
|
Beamish JA, He P, Kottke-Marchant K, Marchant RE. Molecular regulation of contractile smooth muscle cell phenotype: implications for vascular tissue engineering. TISSUE ENGINEERING PART B-REVIEWS 2011; 16:467-91. [PMID: 20334504 DOI: 10.1089/ten.teb.2009.0630] [Citation(s) in RCA: 287] [Impact Index Per Article: 22.1] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The molecular regulation of smooth muscle cell (SMC) behavior is reviewed, with particular emphasis on stimuli that promote the contractile phenotype. SMCs can shift reversibly along a continuum from a quiescent, contractile phenotype to a synthetic phenotype, which is characterized by proliferation and extracellular matrix (ECM) synthesis. This phenotypic plasticity can be harnessed for tissue engineering. Cultured synthetic SMCs have been used to engineer smooth muscle tissues with organized ECM and cell populations. However, returning SMCs to a contractile phenotype remains a key challenge. This review will integrate recent work on how soluble signaling factors, ECM, mechanical stimulation, and other cells contribute to the regulation of contractile SMC phenotype. The signal transduction pathways and mechanisms of gene expression induced by these stimuli are beginning to be elucidated and provide useful information for the quantitative analysis of SMC phenotype in engineered tissues. Progress in the development of tissue-engineered scaffold systems that implement biochemical, mechanical, or novel polymer fabrication approaches to promote contractile phenotype will also be reviewed. The application of an improved molecular understanding of SMC biology will facilitate the design of more potent cell-instructive scaffold systems to regulate SMC behavior.
Collapse
Affiliation(s)
- Jeffrey A Beamish
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, Ohio 44106-7207, USA
| | | | | | | |
Collapse
|
46
|
Cao Y, Poon YF, Feng J, Rayatpisheh S, Chan V, Chan-Park MB. Regulating orientation and phenotype of primary vascular smooth muscle cells by biodegradable films patterned with arrays of microchannels and discontinuous microwalls. Biomaterials 2010; 31:6228-38. [PMID: 20537704 DOI: 10.1016/j.biomaterials.2010.04.059] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2010] [Accepted: 04/29/2010] [Indexed: 01/30/2023]
Abstract
Vascular smooth muscle cells (vSMCs) cultured in vitro are known to exhibit phenotype hyperplasticity. This plasticity is potentially very useful in tissue engineering of blood vessels. The synthetic phenotype is necessary for cell proliferation on the tissue scaffold but the cells must ultimately assume a quiescent, contractile phenotype for normal vascular function. In vitro control of vSMC phenotype has been challenging. This study shows that microchannel scaffolds with discontinuous walls can support primary vSMC proliferation and, when the cells reach confluence inside the channels, transform the cell phenotype towards greater contractility and promote cell alignment. A thorough time-resolved study was undertaken to characterize the expression of the contractile proteins alpha-actin, calponin, myosin heavy chain (MHC) and smoothelin as a function of time and initial cell density on microchannel scaffolds. The results consistently indicate that primary vSMCs cultured on the microchannel substrate substantially align parallel to the microwalls, become more elongated and significantly increase their expression of contractile proteins only when the cells reach confluence. MHC immunostaining was visible in the micropatterned cells after confluence but not in flat substrate cells or non-confluent micropatterned cells, which further verifies the increased contractility of the confluent channel-constrained vSMCs. The higher total amount of deposited elastin and collagen in confluent flat cultures than in confluent micropatterned cultures also provides confirmation of the higher contractility of the channel-constrained cells. These results establish that our microchanneled film can trigger the switch of primary vSMCs from a proliferative state to a more contractile phenotype at confluence.
Collapse
Affiliation(s)
- Ye Cao
- School of Chemical and Biomedical Engineering, Nanyang Technological University, 62 Nanyang Drive, Singapore 637459
| | | | | | | | | | | |
Collapse
|
47
|
Allahverdian S, Francis GA. Cholesterol Homeostasis and High-Density Lipoprotein Formation in Arterial Smooth Muscle Cells. Trends Cardiovasc Med 2010; 20:96-102. [DOI: 10.1016/j.tcm.2010.09.002] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 08/31/2010] [Indexed: 01/24/2023]
|
48
|
Lim YM, Gwon HJ, Choi JH, Shin J, Nho YC, Jeong SI, Chong MS, Lee YM, Kwon IK, Kim SE. Preparation and biocompatibility study of gelatin/kappa-carrageenan scaffolds. Macromol Res 2010. [DOI: 10.1007/s13233-009-0068-1] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
|
49
|
Hirayama Y, Sumpio BE. Role of Ligand-Specific Integrins in Endothelial Cell Alignment and Elongation Induced by Cyclic Strain. ACTA ACUST UNITED AC 2009; 14:275-83. [DOI: 10.1080/10623320701746248] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
|
50
|
Bulick AS, Muñoz-Pinto DJ, Qu X, Mani M, Cristancho D, Urban M, Hahn MS. Impact of endothelial cells and mechanical conditioning on smooth muscle cell extracellular matrix production and differentiation. Tissue Eng Part A 2009; 15:815-25. [PMID: 19108675 DOI: 10.1089/ten.tea.2008.0179] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
The aim of the current study was to explore the separate and coupled effects of endothelial cell (EC) presence and mechanical conditioning on smooth muscle cell (SMC) responses by combining bilayered poly(ethylene glycol) diacrylate (PEGDA) hydrogels with a pulsatile flow bioreactor. Each construct was composed of an outer PEGDA layer containing SMC and an inner PEGDA layer, either with or without EC. After an initial 3 days of static culture, EC(+) and EC(-) constructs were each further divided into two subgroups, half of which received mechanical conditioning mimetic of late gestation (mean pressures of approximately 50 mmHg and peak-to-trough pressure differentials of approximately 20 mmHg at approximately 140-180 beats/min) and half of which were cultured statically. After 18 additional days of culture, the SMC-containing layer of each construct was harvested, and western blots and quantitative histology were conducted to compare collagen type I, collagen type III, and elastin levels among treatment groups. SMC differentiation was evaluated by focusing on SMC marker calponin h1 and direct regulators of its gene expression-the transcription factor serum response factor (SRF) and two of its binding partners, myocardin and Elk-1. Combined EC and pulsatile flow conditioning increased elastin production, but decreased collagen type I deposition. Further, combined EC presence and mechanical stimulation increased SRF levels and the ratio of myocardin to active, phosphorylated Elk-1. This modulation of SRF and its binding partners appeared to result in a net increase in SMC differentiation, as evidenced by an associated increase in calponin h1 levels.
Collapse
Affiliation(s)
- Allen S Bulick
- Department of Chemical Engineering, Texas A&M University , College Station, Texas
| | | | | | | | | | | | | |
Collapse
|