1
|
Paukner D, Jennings IR, Cyron CJ, Humphrey JD. Dynamic biaxial loading of vascular smooth muscle cell seeded tissue equivalents. J Mech Behav Biomed Mater 2024; 157:106639. [PMID: 38970943 DOI: 10.1016/j.jmbbm.2024.106639] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Revised: 06/18/2024] [Accepted: 06/18/2024] [Indexed: 07/08/2024]
Abstract
An intricate reciprocal relationship exists between adherent synthetic cells and their extracellular matrix (ECM). These cells deposit, organize, and degrade the ECM, which in turn influences cell phenotype via responses that include sensitivity to changes in the mechanical state that arises from changes in external loading. Collagen-based tissue equivalents are commonly used as simple but revealing model systems to study cell-matrix interactions. Nevertheless, few quantitative studies report changes in the forces that the cells establish and maintain in such gels under dynamic loading. Moreover, most prior studies have been limited to uniaxial experiments despite many soft tissues, including arteries, experiencing multiaxial loading in vivo. To begin to close this gap, we use a custom biaxial bioreactor to subject collagen gels seeded with primary aortic smooth muscle cells to different biaxial loading conditions. These conditions include cyclic loading with different amplitudes as well as different mechanical constraints at the boundaries of a cruciform sample. Irrespective of loading amplitude and boundary condition, similar mean steady-state biaxial forces emerged across all tests. Additionally, stiffness-force relationships assessed via intermittent equibiaxial force-extension tests showed remarkable similarity for ranges of forces to which the cells adapted during periods of cyclic loading. Taken together, these findings are consistent with a load-mediated homeostatic response by vascular smooth muscle cells.
Collapse
Affiliation(s)
- Daniel Paukner
- Institute for Continuum and Material Mechanics, Hamburg University of Technology, Hamburg, Germany; Institute of Material Systems Modeling, Helmholtz-Zentrum Hereon, Geesthacht, Germany; Department of Biomedical Engineering, Yale University, New Haven, CT, USA.
| | | | - Christian J Cyron
- Institute for Continuum and Material Mechanics, Hamburg University of Technology, Hamburg, Germany; Institute of Material Systems Modeling, Helmholtz-Zentrum Hereon, Geesthacht, Germany
| | - Jay D Humphrey
- Department of Biomedical Engineering, Yale University, New Haven, CT, USA.
| |
Collapse
|
2
|
Zhao J, Yoshizumi M. A Comprehensive Retrospective Study on the Mechanisms of Cyclic Mechanical Stretch-Induced Vascular Smooth Muscle Cell Death Underlying Aortic Dissection and Potential Therapeutics for Preventing Acute Aortic Aneurysm and Associated Ruptures. Int J Mol Sci 2024; 25:2544. [PMID: 38473793 DOI: 10.3390/ijms25052544] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Revised: 02/19/2024] [Accepted: 02/20/2024] [Indexed: 03/14/2024] Open
Abstract
Acute aortic dissection (AAD) and associated ruptures are the leading causes of death in cardiovascular diseases (CVDs). Hypertension is a prime risk factor for AAD. However, the molecular mechanisms underlying AAD remain poorly understood. We previously reported that cyclic mechanical stretch (CMS) leads to the death of rat aortic smooth muscle cells (RASMCs). This review focuses on the mechanisms of CMS-induced vascular smooth muscle cell (VSMC) death. Moreover, we have also discussed the potential therapeutics for preventing AAD and aneurysm ruptures.
Collapse
Affiliation(s)
- Jing Zhao
- Department of Pharmacology, Nara Medical University School of Medicine, 840 Shijo-Cho, Kashihara 634-8521, Japan
| | - Masanori Yoshizumi
- Department of Pharmacology, Nara Medical University School of Medicine, 840 Shijo-Cho, Kashihara 634-8521, Japan
| |
Collapse
|
3
|
Fu S, Li H, Wu Y, Wang J. Nano-/micro-scaled hydroxyapatite ceramic construction and the regulation of immune-associated osteogenic differentiation. J Biomed Mater Res A 2024; 112:193-209. [PMID: 37680167 DOI: 10.1002/jbm.a.37606] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2023] [Revised: 08/04/2023] [Accepted: 08/24/2023] [Indexed: 09/09/2023]
Abstract
Hydroxyapatite (HA) bioceramic is a promising substitute for bone defects, and the surface properties are major factors that influence bioactivity and osteoinductivity. In this study, two kinds of HA bioceramics with nanoscale (n-HA) and microscale (m-HA) surface topography were designed to mimic the natural bone, thus enhancing the stimulation of osteogenic differentiation and revealing the potential mechanism. Compared to m-HA, n-HA owned a larger surface roughness, a stronger wettability, and reduced hardness and indentation modulus. Based on these properties, n-HA could maintain the conformation of vitronectin better than m-HA, which may contribute to higher cellular activities and a stronger promotion of osteogenic differentiation of mesenchymal stem cells (MSCs). Further RNA sequencing analysis compared the molecular expression between n-HA and m-HA. Six hundred twenty-seven differentially expressed genes were identified in MSCs, and 17 upregulated genes and 610 downregulated genes were included when n-HA compared to m-HA. The GO cluster analysis and enriched Kyoto encyclopedia of genes and genome signaling pathways revealed a close correlation with the immune process in both upregulated (chemokine signaling pathway and cytokine-cytokine receptor interaction) and downregulated pathways (osteoclasts differentiation). It suggested that the nanoscale surface topography of HA enhanced the osteoinductivity of MSCs and could not be separated from its regulation of immune function and the retention of adsorbed protein conformation.
Collapse
Affiliation(s)
- Shijia Fu
- School of Life Sciences, Northwestern Polytechnical University, Xi'an, China
| | - Huishan Li
- School of Life Sciences, Northwestern Polytechnical University, Xi'an, China
| | - Yue Wu
- School of Life Sciences, Northwestern Polytechnical University, Xi'an, China
| | - Jing Wang
- School of Life Sciences, Northwestern Polytechnical University, Xi'an, China
| |
Collapse
|
4
|
Wu T, Li N, Zhang Q, Liu R, Zhao H, Fan Z, Zhuo L, Yang Y, Xu Y. MKL1 fuels ROS-induced proliferation of vascular smooth muscle cells by modulating FOXM1 transcription. Redox Biol 2022; 59:102586. [PMID: 36587486 PMCID: PMC9823229 DOI: 10.1016/j.redox.2022.102586] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2022] [Accepted: 12/22/2022] [Indexed: 12/31/2022] Open
Abstract
Reactive oxygen species (ROS) promotes vascular injury and neointima formation in part by stimulating proliferation of vascular smooth muscle cells (VSMC). The underlying transcriptional mechanism, however, is not completely understood. Here we report that VSMC-specific deletion of MKL1 in mice suppressed neointima formation in a classic model of vascular injury. Likewise, pharmaceutical inhibition of MKL1 activity by CCG-1423 similarly mollified neointima formation in mice. Over-expression of a constitutively active MKL1 in vascular smooth muscle cells enhanced proliferation in a ROS-dependent manner. On the contrary, MKL1 depletion or inhibition attenuated VSMC proliferation. PCR array based screening identified forkhead box protein M1 (FOXM1) as a direct target for MKL1. MKL1 interacted with E2F1 to activate FOXM1 expression. Concordantly, FOXM1 depletion ameliorated MKL1-dependent VSMC proliferation. Of interest, ROS-induced MKL1 phosphorylation through MK2 was essential for its interaction with E2F1 and consequently FOXM1 trans-activation. Importantly, a positive correlation between FOXM1 expression and VSMC proliferation was identified in arterial specimens from patients with restenosis. Taken together, our data suggest that a redox-sensitive phosphorylation-switch of MKL1 activates FOXM1 transcription and mediates ROS fueled vascular smooth muscle proliferation. Targeting the MK-2/MKL1/FOXM1 axis may be considered as a reasonable approach for treatment of restenosis.
Collapse
Affiliation(s)
- Teng Wu
- Key Laboratory of Targeted Intervention of Cardiovascular Disease and Collaborative Innovation Center for Cardiovascular Translational Medicine, Department of Pathophysiology, Nanjing Medical University, Nanjing, China
| | - Nan Li
- Department of Human Anatomy, Nanjing Medical University, Nanjing, China
| | - Qiumei Zhang
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing, China
| | - Ruiqi Liu
- Key Laboratory of Targeted Intervention of Cardiovascular Disease and Collaborative Innovation Center for Cardiovascular Translational Medicine, Department of Pathophysiology, Nanjing Medical University, Nanjing, China
| | - Hongwei Zhao
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing, China
| | - Zhiwen Fan
- Department of Pathology, Nanjing Drum Tower Hospital Affiliated with Nanjing University School of Medicine, Nanjing, China
| | - Lili Zhuo
- Department of Geriatrics, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, China.
| | - Yuyu Yang
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing, China; Institute of Biomedical Research and College of Life Sciences, Liaocheng University, Liaocheng, China.
| | - Yong Xu
- Key Laboratory of Targeted Intervention of Cardiovascular Disease and Collaborative Innovation Center for Cardiovascular Translational Medicine, Department of Pathophysiology, Nanjing Medical University, Nanjing, China; Institute of Biomedical Research and College of Life Sciences, Liaocheng University, Liaocheng, China.
| |
Collapse
|
5
|
Zieleniewska NA, Kazberuk M, Chlabicz M, Eljaszewicz A, Kamiński K. Trained Immunity as a Trigger for Atherosclerotic Cardiovascular Disease-A Literature Review. J Clin Med 2022; 11:jcm11123369. [PMID: 35743439 PMCID: PMC9224533 DOI: 10.3390/jcm11123369] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Revised: 06/05/2022] [Accepted: 06/09/2022] [Indexed: 11/17/2022] Open
Abstract
Atherosclerosis remains the leading cause of cardiovascular diseases and represents a primary public health challenge. This chronic state may lead to a number of life-threatening conditions, such as myocardial infarction and stroke. Lipid metabolism alterations and inflammation remain at the forefront of the pathogenesis of atherosclerotic cardiovascular disease, but the overall mechanism is not yet fully understood. Recently, significant effects of trained immunity on atherosclerotic plaque formation and development have been reported. An increased reaction to restimulation with the same stimulator is a hallmark of the trained innate immune response. The impact of trained immunity is a prominent factor in both acute and chronic coronary syndrome, which we outline in this review.
Collapse
Affiliation(s)
- Natalia Anna Zieleniewska
- Department of Population Medicine and Lifestyle Diseases Prevention, Medical University of Białystok, 15-259 Bialystok, Poland; (N.A.Z.); (M.C.)
- Department of Cardiology, Teaching University Hospital of Białystok, 15-259 Bialystok, Poland
| | - Małgorzata Kazberuk
- Scientific Group of Department of Population Medicine and Lifestyle Diseases Prevention, Medical University of Białystok, 15-259 Bialystok, Poland;
| | - Małgorzata Chlabicz
- Department of Population Medicine and Lifestyle Diseases Prevention, Medical University of Białystok, 15-259 Bialystok, Poland; (N.A.Z.); (M.C.)
- Department of Invasive Cardiology, Teaching University Hospital of Białystok, 15-259 Bialystok, Poland
| | - Andrzej Eljaszewicz
- Department of Regenerative Medicine and Immune Regulation, Medical University of Białystok, 15-259 Bialystok, Poland;
| | - Karol Kamiński
- Department of Population Medicine and Lifestyle Diseases Prevention, Medical University of Białystok, 15-259 Bialystok, Poland; (N.A.Z.); (M.C.)
- Department of Cardiology, Teaching University Hospital of Białystok, 15-259 Bialystok, Poland
- Correspondence:
| |
Collapse
|
6
|
Mechanical stretching of cells and lipid nanoparticles for nucleic acid delivery. J Control Release 2021; 339:208-219. [PMID: 34563590 DOI: 10.1016/j.jconrel.2021.09.021] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Accepted: 09/17/2021] [Indexed: 11/23/2022]
Abstract
Gene therapy has gained popularity in the treatment of incurable diseases. However, cell components, such as surface membrane, cytoskeleton protein, and nuclear envelope, retard the transport of nucleic acids, lowering the transfection efficiency. We developed a physical-chemical hybrid platform (S-RCLs) involving cationic lipid nanoparticles (RCLs) exposed to cyclic stretch. The transfection efficiency and delivery mechanisms of S-RCLs for siRNAs and pDNAs (plasmid DNAs encoding luciferase) were investigated. S-RCLs effectively delivered both siRNAs and pDNAs by overcoming the cell barriers. Mechanistically, S-RCLs promote the cellular uptake mediated by CD44, EH-domain containing 2 (EHD2), and caveolin-1 (CAV-1); intracellular transport via MAP6 Domain Containing 1 (Map6d1) and F-actin; and DNA transcription regulated by LSM3 and Hist1h3e in the nucleus. Thus, S-RCLs are a promising hybrid platform with excellent efficiency and biocompatibility for gene delivery both in vitro and in vivo.
Collapse
|
7
|
Abstract
Vascular smooth muscle cells (VSMC) are now considered important contributors to the pathophysiological and biophysical mechanisms underlying arterial stiffening in aging. Here, we review mechanisms whereby VSMC stiffening alters vascular function and contributes to the changes in vascular stiffening observed in aging and cardiovascular disease. Vascular stiffening in arterial aging was historically associated with changes in the extracellular matrix; however, new evidence suggests that endothelial and vascular smooth muscle cell stiffness also contribute to overall blood vessel stiffness. Furthermore, VSMC play an integral role in regulating matrix deposition and vessel wall contractility via interaction between the actomyosin contractile unit and adhesion structures that anchor the cell within the extracellular matrix. Aged-induce phenotypic modulation of VSMC from a contractile to a synthetic phenotype is associated with decreased cellular contractility and increased cell stiffness. Aged VSMC also display reduced mechanosensitivity and adaptation to mechanical signals from their microenvironment due to impaired intracellular signaling. Finally, evidence for decreased contractility in arteries from aged animals demonstrate that changes at the cellular level result in decreased functional properties at the tissue level.
Collapse
|
8
|
Role of Vascular Smooth Muscle Cell Phenotype Switching in Arteriogenesis. Int J Mol Sci 2021; 22:ijms221910585. [PMID: 34638923 PMCID: PMC8508942 DOI: 10.3390/ijms221910585] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2021] [Revised: 09/26/2021] [Accepted: 09/27/2021] [Indexed: 12/12/2022] Open
Abstract
Arteriogenesis is one of the primary physiological means by which the circulatory collateral system restores blood flow after significant arterial occlusion in peripheral arterial disease patients. Vascular smooth muscle cells (VSMCs) are the predominant cell type in collateral arteries and respond to altered blood flow and inflammatory conditions after an arterial occlusion by switching their phenotype between quiescent contractile and proliferative synthetic states. Maintaining the contractile state of VSMC is required for collateral vascular function to regulate blood vessel tone and blood flow during arteriogenesis, whereas synthetic SMCs are crucial in the growth and remodeling of the collateral media layer to establish more stable conduit arteries. Timely VSMC phenotype switching requires a set of coordinated actions of molecular and cellular mediators to result in an expansive remodeling of collaterals that restores the blood flow effectively into downstream ischemic tissues. This review overviews the role of VSMC phenotypic switching in the physiological arteriogenesis process and how the VSMC phenotype is affected by the primary triggers of arteriogenesis such as blood flow hemodynamic forces and inflammation. Better understanding the role of VSMC phenotype switching during arteriogenesis can identify novel therapeutic strategies to enhance revascularization in peripheral arterial disease.
Collapse
|
9
|
Dey K, Roca E, Ramorino G, Sartore L. Progress in the mechanical modulation of cell functions in tissue engineering. Biomater Sci 2021; 8:7033-7081. [PMID: 33150878 DOI: 10.1039/d0bm01255f] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
In mammals, mechanics at multiple stages-nucleus to cell to ECM-underlie multiple physiological and pathological functions from its development to reproduction to death. Under this inspiration, substantial research has established the role of multiple aspects of mechanics in regulating fundamental cellular processes, including spreading, migration, growth, proliferation, and differentiation. However, our understanding of how these mechanical mechanisms are orchestrated or tuned at different stages to maintain or restore the healthy environment at the tissue or organ level remains largely a mystery. Over the past few decades, research in the mechanical manipulation of the surrounding environment-known as substrate or matrix or scaffold on which, or within which, cells are seeded-has been exceptionally enriched in the field of tissue engineering and regenerative medicine. To do so, traditional tissue engineering aims at recapitulating key mechanical milestones of native ECM into a substrate for guiding the cell fate and functions towards specific tissue regeneration. Despite tremendous progress, a big puzzle that remains is how the cells compute a host of mechanical cues, such as stiffness (elasticity), viscoelasticity, plasticity, non-linear elasticity, anisotropy, mechanical forces, and mechanical memory, into many biological functions in a cooperative, controlled, and safe manner. High throughput understanding of key cellular decisions as well as associated mechanosensitive downstream signaling pathway(s) for executing these decisions in response to mechanical cues, solo or combined, is essential to address this issue. While many reports have been made towards the progress and understanding of mechanical cues-particularly, substrate bulk stiffness and viscoelasticity-in regulating the cellular responses, a complete picture of mechanical cues is lacking. This review highlights a comprehensive view on the mechanical cues that are linked to modulate many cellular functions and consequent tissue functionality. For a very basic understanding, a brief discussion of the key mechanical players of ECM and the principle of mechanotransduction process is outlined. In addition, this review gathers together the most important data on the stiffness of various cells and ECM components as well as various tissues/organs and proposes an associated link from the mechanical perspective that is not yet reported. Finally, beyond addressing the challenges involved in tuning the interplaying mechanical cues in an independent manner, emerging advances in designing biomaterials for tissue engineering are also explored.
Collapse
Affiliation(s)
- Kamol Dey
- Department of Applied Chemistry and Chemical Engineering, Faculty of Science, University of Chittagong, Bangladesh
| | | | | | | |
Collapse
|
10
|
Maxey AP, McCain ML. Tools, techniques, and future opportunities for characterizing the mechanobiology of uterine myometrium. Exp Biol Med (Maywood) 2021; 246:1025-1035. [PMID: 33554648 DOI: 10.1177/1535370221989259] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
The myometrium is the smooth muscle layer of the uterus that generates the contractions that drive processes such as menstruation and childbirth. Aberrant contractions of the myometrium can result in preterm birth, insufficient progression of labor, or other difficulties that can lead to maternal or fetal complications or even death. To investigate the underlying mechanisms of these conditions, the most common model systems have conventionally been animal models and human tissue strips, which have limitations mostly related to relevance and scalability, respectively. Myometrial smooth muscle cells have also been isolated from patient biopsies and cultured in vitro as a more controlled experimental system. However, in vitro approaches have focused primarily on measuring the effects of biochemical stimuli and neglected biomechanical stimuli, despite the extensive evidence indicating that remodeling of tissue rigidity or excessive strain is associated with uterine disorders. In this review, we first describe the existing approaches for modeling human myometrium with animal models and human tissue strips and compare their advantages and disadvantages. Next, we introduce existing in vitro techniques and assays for assessing contractility and summarize their applications in elucidating the role of biochemical or biomechanical stimuli on human myometrium. Finally, we conclude by proposing the translation of "organ on chip" approaches to myometrial smooth muscle cells as new paradigms for establishing their fundamental mechanobiology and to serve as next-generation platforms for drug development.
Collapse
Affiliation(s)
- Antonina P Maxey
- Laboratory for Living Systems Engineering, Department of Biomedical Engineering, USC Viterbi School of Engineering, University of Southern California, Los Angeles, CA 90089, USA
| | - Megan L McCain
- Laboratory for Living Systems Engineering, Department of Biomedical Engineering, USC Viterbi School of Engineering, University of Southern California, Los Angeles, CA 90089, USA.,Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine of USC, University of Southern California, Los Angeles, CA 90033, USA
| |
Collapse
|
11
|
Rai N, Shihan M, Seeger W, Schermuly RT, Novoyatleva T. Genetic Delivery and Gene Therapy in Pulmonary Hypertension. Int J Mol Sci 2021; 22:ijms22031179. [PMID: 33503992 PMCID: PMC7865388 DOI: 10.3390/ijms22031179] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2020] [Revised: 01/19/2021] [Accepted: 01/20/2021] [Indexed: 02/06/2023] Open
Abstract
Pulmonary hypertension (PH) is a progressive complex fatal disease of multiple etiologies. Hyperproliferation and resistance to apoptosis of vascular cells of intimal, medial, and adventitial layers of pulmonary vessels trigger excessive pulmonary vascular remodeling and vasoconstriction in the course of pulmonary arterial hypertension (PAH), a subgroup of PH. Multiple gene mutation/s or dysregulated gene expression contribute to the pathogenesis of PAH by endorsing the proliferation and promoting the resistance to apoptosis of pulmonary vascular cells. Given the vital role of these cells in PAH progression, the development of safe and efficient-gene therapeutic approaches that lead to restoration or down-regulation of gene expression, generally involved in the etiology of the disease is the need of the hour. Currently, none of the FDA-approved drugs provides a cure against PH, hence innovative tools may offer a novel treatment paradigm for this progressive and lethal disorder by silencing pathological genes, expressing therapeutic proteins, or through gene-editing applications. Here, we review the effectiveness and limitations of the presently available gene therapy approaches for PH. We provide a brief survey of commonly existing and currently applicable gene transfer methods for pulmonary vascular cells in vitro and describe some more recent developments for gene delivery existing in the field of PH in vivo.
Collapse
Affiliation(s)
- Nabham Rai
- Excellence Cluster Cardio-Pulmonary Institute (CPI), Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Justus Liebig University of Giessen, Aulweg 130, 35392 Giessen, Germany; (N.R.); (M.S.); (W.S.); (R.T.S.)
| | - Mazen Shihan
- Excellence Cluster Cardio-Pulmonary Institute (CPI), Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Justus Liebig University of Giessen, Aulweg 130, 35392 Giessen, Germany; (N.R.); (M.S.); (W.S.); (R.T.S.)
| | - Werner Seeger
- Excellence Cluster Cardio-Pulmonary Institute (CPI), Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Justus Liebig University of Giessen, Aulweg 130, 35392 Giessen, Germany; (N.R.); (M.S.); (W.S.); (R.T.S.)
- Max Planck Institute for Heart and Lung Research, 61231 Bad Nauheim, Germany
- Institute for Lung Health (ILH), 35392 Giessen, Germany
| | - Ralph T. Schermuly
- Excellence Cluster Cardio-Pulmonary Institute (CPI), Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Justus Liebig University of Giessen, Aulweg 130, 35392 Giessen, Germany; (N.R.); (M.S.); (W.S.); (R.T.S.)
| | - Tatyana Novoyatleva
- Excellence Cluster Cardio-Pulmonary Institute (CPI), Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Justus Liebig University of Giessen, Aulweg 130, 35392 Giessen, Germany; (N.R.); (M.S.); (W.S.); (R.T.S.)
- Correspondence:
| |
Collapse
|
12
|
Zhao J, Nakahira K, Kimura A, Kyotani Y, Yoshizumi M. Upregulation of iNOS Protects Cyclic Mechanical Stretch-Induced Cell Death in Rat Aorta Smooth Muscle Cells. Int J Mol Sci 2020; 21:E8660. [PMID: 33212839 PMCID: PMC7698365 DOI: 10.3390/ijms21228660] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Revised: 11/06/2020] [Accepted: 11/14/2020] [Indexed: 01/09/2023] Open
Abstract
Aortic dissection and aneurysm are associated with abnormal hemodynamic loads originating from hypertension. Our previous study demonstrated that cyclic mechanical stretch (CMS, mimicked hypertension) caused the death of rat aortic smooth muscle cells (RASMCs) in a mitogen activated-protein kinases (MAPKs)-dependent manner. The current study investigated the effects of inducible nitric oxide synthase (iNOS) on CMS-induced RASMC death. cDNA microarrays for CMS-treated RASMCs showed that iNOS expression levels were increased in response to CMS. Real-time polymerase chain reaction (PCR) analysis demonstrated that this increase was p38 MAPK (p38)-dependent. NO production was also increased. This increase could be inhibited by p38 and iNOS inhibitors. Thus, CMS-induced iNOS synthesized NO. CMS-induced cell death in RASMCs was increased by the iNOS inhibitor but abrogated by the long-acting NO donor DETA-NONOate. Increased iNOS expression was confirmed in the abdominal aortic constriction mouse model. Signal transducers and activators of transcription 1 (STAT1) was activated in stretched RASMCs, and iNOS expression and NO production were inhibited by the STAT1 inhibitor nifuroxazide. Our findings suggest that RASMCs were protected by iNOS from CMS-stimulated cell death through the STAT1 and p38 signal pathways independently.
Collapse
MESH Headings
- Animals
- Aorta/cytology
- Aorta/enzymology
- Gene Expression Regulation, Enzymologic
- Male
- Mechanotransduction, Cellular
- Muscle, Smooth, Vascular/cytology
- Muscle, Smooth, Vascular/enzymology
- Myocytes, Smooth Muscle/cytology
- Myocytes, Smooth Muscle/enzymology
- Nitric Oxide Synthase Type II/biosynthesis
- Rats
- Rats, Sprague-Dawley
- Stress, Mechanical
- Up-Regulation
Collapse
Affiliation(s)
- Jing Zhao
- Department of Pharmacology, Nara Medical University School of Medicine, 840 Shijo-Cho, Kashihara 634-8521, Japan; (K.N.); (Y.K.); (M.Y.)
| | - Kiichi Nakahira
- Department of Pharmacology, Nara Medical University School of Medicine, 840 Shijo-Cho, Kashihara 634-8521, Japan; (K.N.); (Y.K.); (M.Y.)
| | - Akihiko Kimura
- Department of Forensic Medicine, Wakayama Medical University, 811-1 Kimiidera, Wakayama 641-8509, Japan;
| | - Yoji Kyotani
- Department of Pharmacology, Nara Medical University School of Medicine, 840 Shijo-Cho, Kashihara 634-8521, Japan; (K.N.); (Y.K.); (M.Y.)
| | - Masanori Yoshizumi
- Department of Pharmacology, Nara Medical University School of Medicine, 840 Shijo-Cho, Kashihara 634-8521, Japan; (K.N.); (Y.K.); (M.Y.)
| |
Collapse
|
13
|
Wang L, Deng L, Lin N, Shi Y, Chen J, Zhou Y, Chen D, Liu S, Li C. Berberine inhibits proliferation and apoptosis of vascular smooth muscle cells induced by mechanical stretch via the PDI/ERS and MAPK pathways. Life Sci 2020; 259:118253. [PMID: 32795536 DOI: 10.1016/j.lfs.2020.118253] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2020] [Revised: 08/03/2020] [Accepted: 08/08/2020] [Indexed: 12/13/2022]
Abstract
AIMS We recently demonstrated that mechanical stretch increases the proliferation and apoptosis of vascular smooth muscle cells (VSMCs) by activating the protein disulfide isomerase (PDI) redox system, thus accelerating atherosclerotic lesion formation in the transplanted vein. At present, there are no efficient intervention measures to prevent this phenomenon. Berberine inhibits pathological vascular remodeling caused by hypertension, but the underlying mechanism is controversial. Herein, we investigate the role of berberine and the underlying mechanism of its effects on mechanical stretch-induced VSMC proliferation and apoptosis. MAIN METHODS Mouse VSMCs cultivated on flexible membranes were pretreated for 1 h with one of the following substances: berberine, PDI inhibitor bacitracin, MAPK inhibitors, or ERS inhibitor 4-PBA. VSMCs were then subjected to mechanical stretch. Immunofluorescence and western blot were used to detect proliferation and apoptosis, as well as to analyze signaling pathways in VSMCs. KEY FINDINGS Our results showed that berberine inhibits the PDI-endoplasmic reticulum stress system, thereby attenuating the simultaneous increase of VSMC proliferation and apoptosis in response to mechanical stretch. Interestingly, MAPK inhibitors PD98059, SP600125, and SB202190 significantly reduced the activation of ERS signaling cascades, and their combination with berberine had additive effects. The ERS inhibitor 4-PBA reduced PDI activation and ERS signaling, but not MAPK phosphorylation. Moreover, caspase-3 and caspase-12 were downregulated by berberine. SIGNIFICANCE These results illustrate a novel mechanism of action of berberine that has practical implications. Our data provide important insights for the prevention and treatment of vascular remodeling and diseases caused by mechanical stretching during hypertension.
Collapse
Affiliation(s)
- Linli Wang
- Department of Histology and Embryology, Zhongshan School of Medicine, Sun Yat-sen University, China
| | - Lie Deng
- Department of Histology and Embryology, Zhongshan School of Medicine, Sun Yat-sen University, China
| | - Ning Lin
- Department of Histology and Embryology, Zhongshan School of Medicine, Sun Yat-sen University, China
| | - Yi Shi
- Division of Vascular Surgery, The First Affiliated Hospital of Sun Yat-sen University, China
| | - Jingbo Chen
- Department of Histology and Embryology, Zhongshan School of Medicine, Sun Yat-sen University, China
| | - Yan Zhou
- Department of Histology and Embryology, Zhongshan School of Medicine, Sun Yat-sen University, China
| | - Dadi Chen
- Experimental Center for Basic Medical Teaching, Zhongshan School of Medicine, Sun Yat-sen University, China
| | - Shuying Liu
- Department of Histology and Embryology, Zhongshan School of Medicine, Sun Yat-sen University, China.
| | - Chaohong Li
- Department of Histology and Embryology, Zhongshan School of Medicine, Sun Yat-sen University, China.
| |
Collapse
|
14
|
Kim BG, Sung JS, Jang Y, Cha YJ, Kang S, Han HH, Lee JH, Cho NH. Compression-induced expression of glycolysis genes in CAFs correlates with EMT and angiogenesis gene expression in breast cancer. Commun Biol 2019; 2:313. [PMID: 31428701 PMCID: PMC6694123 DOI: 10.1038/s42003-019-0553-9] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2019] [Accepted: 07/17/2019] [Indexed: 12/12/2022] Open
Abstract
Tumor growth increases compressive stress within a tissue, which is associated with solid tumor progression. However, very little is known about how compressive stress contributes to tumor progression. Here, we show that compressive stress induces glycolysis in human breast cancer associated fibroblast (CAF) cells and thereby contributes to the expression of epithelial to mesenchymal (EMT)- and angiogenesis-related genes in breast cancer cells. Lactate production was increased in compressed CAF cells, in a manner dependent on the expression of metabolic genes ENO2, HK2, and PFKFB3. Conditioned medium from compressed CAFs promoted the proliferation of breast cancer cells and the expression of EMT and/or angiogenesis-related genes. In patient tissues with high compressive stress, the expression of compression-induced metabolic genes was significantly and positively correlated with EMT and/or angiogenesis-related gene expression and metastasis size. These findings illustrate a mechanotransduction pathway involving stromal glycolysis that may be relevant also for other solid tumours.
Collapse
Affiliation(s)
- Baek Gil Kim
- Department of Pathology, Yonsei University College of Medicine, Seoul, South Korea
- Brain Korea 21 Plus Project for Medical Science, Yonsei University College of Medicine, Seoul, South Korea
| | - Jin Sol Sung
- Brain Korea 21 Plus Project for Medical Science, Yonsei University College of Medicine, Seoul, South Korea
| | - Yeonsue Jang
- Department of Pathology, Yonsei University College of Medicine, Seoul, South Korea
| | - Yoon Jin Cha
- Department of Pathology, Yonsei University College of Medicine, Seoul, South Korea
| | - Suki Kang
- Department of Pathology, Yonsei University College of Medicine, Seoul, South Korea
- Severance Biomedical Science Institute (SBSI), Yonsei University College of Medicine, Seoul, South Korea
| | - Hyun Ho Han
- Brain Korea 21 Plus Project for Medical Science, Yonsei University College of Medicine, Seoul, South Korea
| | - Joo Hyun Lee
- Brain Korea 21 Plus Project for Medical Science, Yonsei University College of Medicine, Seoul, South Korea
| | - Nam Hoon Cho
- Department of Pathology, Yonsei University College of Medicine, Seoul, South Korea
- Brain Korea 21 Plus Project for Medical Science, Yonsei University College of Medicine, Seoul, South Korea
- Severance Biomedical Science Institute (SBSI), Yonsei University College of Medicine, Seoul, South Korea
- Global 5-5-10 System Biology, Yonsei University, Seoul, South Korea
| |
Collapse
|
15
|
Biophysical implications of Maxwell stress in electric field stimulated cellular microenvironment on biomaterial substrates. Biomaterials 2019; 209:54-66. [DOI: 10.1016/j.biomaterials.2019.04.010] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2018] [Revised: 04/05/2019] [Accepted: 04/06/2019] [Indexed: 01/09/2023]
|
16
|
Arnold C, Feldner A, Zappe M, Komljenovic D, De La Torre C, Ruzicka P, Hecker M, Neuhofer W, Korff T. Genetic ablation of NFAT5/TonEBP in smooth muscle cells impairs flow- and pressure-induced arterial remodeling in mice. FASEB J 2018; 33:3364-3377. [PMID: 30383452 DOI: 10.1096/fj.201801594r] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
The arterial wall adapts to alterations in blood flow and pressure by remodeling the cellular and extracellular architecture. Biomechanical stress of vascular smooth muscle cells (VSMCs) in the media is thought to precede this process and promote their activation and subsequent proliferation. However, molecular determinants orchestrating the transcriptional phenotype under these conditions have been insufficiently studied. We identified the transcription factor, nuclear factor of activated T cells 5 (NFAT5; or tonicity enhancer-binding protein) as a crucial regulatory element of mechanical stress responses of VSMCs. Here, the relevance of NFAT5 for arterial growth and thickening is investigated in mice upon inducible smooth muscle cell (SMC)-specific genetic ablation of Nfat5. In cultured mouse VSMCs, loss of Nfat5 inhibits the expression of gene sets involved in the control of the cell cycle and the interaction with the extracellular matrix and cytoskeletal dynamics. In vivo, SMC-specific knockout of Nfat5 did not affect the general vascular architecture and blood pressure levels under baseline conditions. However, proliferation of VSMCs and the thickening of the arterial wall were inhibited during both flow-induced collateral remodeling and hypertension-mediated arterial hypertrophy. Whereas originally described as a hypertonicity-responsive transcription factor, these findings identify NFAT5 as a novel molecular determinant of biomechanically induced phenotype changes of VSMCs and wall stress-induced arterial remodeling processes.-Arnold, C., Feldner, A., Zappe, M., Komljenovic, D., De La Torre, C., Ruzicka, P., Hecker, M., Neuhofer, W., Korff, T. Genetic ablation of NFAT5/TonEBP in smooth muscle cells impairs flow- and pressure-induced arterial remodeling in mice.
Collapse
Affiliation(s)
- Caroline Arnold
- Department of Cardiovascular Physiology, Institute of Physiology and Pathophysiology, Heidelberg University, Heidelberg, Germany
| | - Anja Feldner
- Department of Cardiovascular Physiology, Institute of Physiology and Pathophysiology, Heidelberg University, Heidelberg, Germany
| | - Maren Zappe
- Department of Cardiovascular Physiology, Institute of Physiology and Pathophysiology, Heidelberg University, Heidelberg, Germany
| | - Dorde Komljenovic
- Division of Medical Physics in Radiology, German Cancer Research Center, Heidelberg, Germany
| | - Carolina De La Torre
- Center of Medical Research, Medical Faculty Mannheim, Heidelberg University, Heidelberg, Germany
| | - Philipp Ruzicka
- Department of Cardiovascular Physiology, Institute of Physiology and Pathophysiology, Heidelberg University, Heidelberg, Germany
| | - Markus Hecker
- Department of Cardiovascular Physiology, Institute of Physiology and Pathophysiology, Heidelberg University, Heidelberg, Germany
| | - Wolfgang Neuhofer
- Medical Clinic V, University Hospital Mannheim, Heidelberg University, Heidelberg, Germany
| | - Thomas Korff
- Department of Cardiovascular Physiology, Institute of Physiology and Pathophysiology, Heidelberg University, Heidelberg, Germany.,European Center for Angioscience, Medical Faculty Mannheim, Heidelberg University, Heidelberg, Germany
| |
Collapse
|
17
|
Arai M, Sera T, Hasegawa T, Kudo S. Spatial and temporal translocation of PKCα in single endothelial cell in response to mechanical stimulus. Exp Cell Res 2018; 367:205-215. [DOI: 10.1016/j.yexcr.2018.03.038] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2017] [Revised: 03/28/2018] [Accepted: 03/29/2018] [Indexed: 12/14/2022]
|
18
|
Guo F, Yang L, Luo J, Quan H, Wang Z, Peng H, Hong C, Li J, Jiang Z, Zhang L, Qin X. Involvement of CGRP-RCP in the caveolin-1/ERK1/2 signal pathway in the static pressure-induced proliferation of vascular smooth muscle cells. J Cell Physiol 2018; 233:6910-6920. [PMID: 29741760 DOI: 10.1002/jcp.26582] [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: 07/18/2017] [Accepted: 03/08/2018] [Indexed: 11/09/2022]
Abstract
Previous study suggested that the receptor component protein (RCP), one of the components of calcitonin gene-related peptide (CGRP) receptor, plays a multiple role in the cellular signal transduction. The study was designed to investigate whether or not the RCP involved in the regulation of caveolin-1/extracellular signal-regulated kinases-1 and -2 (ERK1/2) signal pathway in the vascular smooth muscle cells (VSMCs) proliferation induced by static pressure. Mouse-derived VSMCs line A10 (A10 VSMCs) was served as project in this experiment. Results showed that the A10 VSMCs viability and proliferating cell nuclear antigen (PCNA) expression which were increased by static pressure were inhibited by pretreatment of CGRP. In like manner, the expressions of the decreased-caveolin-1 and the increased-phosphorylated ERK1/2 (p-ERK1/2) induced by static pressure were significantly reversed by pretreatment of CGRP, respectively. Meanwhile, the expression of RCP was up-regulated by the static pressure. Silence of RCP gene with the small interrupt RNA (siRNA) not only significantly increased A10 VSMC proliferation but also increased the expression of p-ERK1/2 in response to static pressure. When treatment of A10 VSMCs with 120-mmHg static pressure for different time, however, the protein band of caveolin-1 and RCP was the least at time point of 10 min, but the p-ERK1/2 expression was the most maximum. In conclusion, RCP maybe involved in the static pressure-induced A10 VSMCs proliferation by regulation of caveolin-1/ERK1/2 signal pathway.
Collapse
Affiliation(s)
- Feng Guo
- Institute of Pharmacy and Pharmacology, Key Laboratory for Arteriosclerology of Hunan Province, University of South China, Hengyang, China
| | - Li Yang
- Institute of Pharmacy and Pharmacology, Key Laboratory for Arteriosclerology of Hunan Province, University of South China, Hengyang, China
| | - Jingfei Luo
- Institute of Pharmacy and Pharmacology, Key Laboratory for Arteriosclerology of Hunan Province, University of South China, Hengyang, China
| | - Haiyan Quan
- Institute of Pharmacy and Pharmacology, Key Laboratory for Arteriosclerology of Hunan Province, University of South China, Hengyang, China
| | - Zhen Wang
- Institute of Pharmacy and Pharmacology, Key Laboratory for Arteriosclerology of Hunan Province, University of South China, Hengyang, China
| | - Hongyan Peng
- Institute of Pharmacy and Pharmacology, Key Laboratory for Arteriosclerology of Hunan Province, University of South China, Hengyang, China
| | - Chenliang Hong
- Institute of Pharmacy and Pharmacology, Key Laboratory for Arteriosclerology of Hunan Province, University of South China, Hengyang, China
| | - Jie Li
- Institute of Pharmacy and Pharmacology, Key Laboratory for Arteriosclerology of Hunan Province, University of South China, Hengyang, China
| | - Zhisheng Jiang
- Institute of Pharmacy and Pharmacology, Key Laboratory for Arteriosclerology of Hunan Province, University of South China, Hengyang, China
| | - Liang Zhang
- Palmer Laboratory of Cell and Molecular Biology, Palmer College of Chiropractic, Port Orange, Florida
| | - Xuping Qin
- Institute of Pharmacy and Pharmacology, Key Laboratory for Arteriosclerology of Hunan Province, University of South China, Hengyang, China
| |
Collapse
|
19
|
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
|
20
|
Henderson K, Sligar AD, Le VP, Lee J, Baker AB. Biomechanical Regulation of Mesenchymal Stem Cells for Cardiovascular Tissue Engineering. Adv Healthc Mater 2017; 6. [PMID: 28945009 DOI: 10.1002/adhm.201700556] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2017] [Revised: 06/22/2017] [Indexed: 12/15/2022]
Abstract
Mesenchymal stem cells (MSCs) are an appealing potential therapy for vascular diseases; however, many challenges remain in their clinical translation. While the use of biochemical, pharmacological, and substrate-mediated treatments to condition MSCs has been subjected to intense investigation, there has been far less exploration of using these treatments in combination with applied mechanical force for conditioning MSCs toward vascular phenotypes. This review summarizes the current understanding of the use of applied mechanical forces to differentiate MSCs into vascular cells and enhance their therapeutic potential for cardiovascular disease. First recent work on the use of material-based mechanical cues for differentiation of MSCs into vascular and cardiovascular phenotypes is examined. Then a summary of the studies using mechanical stretch or shear stress in combination with biochemical treatments to enhance vascular phenotypes in MSCs is presented.
Collapse
Affiliation(s)
- Kayla Henderson
- Department of Biomedical Engineering; University of Texas at Austin; Austin 78712 TX USA
| | - Andrew D. Sligar
- Department of Biomedical Engineering; University of Texas at Austin; Austin 78712 TX USA
| | - Victoria P. Le
- Department of Biomedical Engineering; University of Texas at Austin; Austin 78712 TX USA
| | - Jason Lee
- Department of Biomedical Engineering; University of Texas at Austin; Austin 78712 TX USA
| | - Aaron B. Baker
- Department of Biomedical Engineering; University of Texas at Austin; Austin 78712 TX USA
- Institute for Cellular and Molecular Biology; University of Texas at Austin; Austin 78712 TX USA
- The Institute for Computational Engineering and Sciences; University of Texas at Austin; Austin 78712 TX USA
- Institute for Biomaterials; Drug Delivery and Regenerative Medicine; University of Texas at Austin; Austin 78712 TX USA
| |
Collapse
|
21
|
Ping S, Liu S, Zhou Y, Li Z, Li Y, Liu K, Bardeesi AS, Wang L, Chen J, Deng L, Wang J, Wang H, Chen D, Zhang Z, Sheng P, Li C. Protein disulfide isomerase-mediated apoptosis and proliferation of vascular smooth muscle cells induced by mechanical stress and advanced glycosylation end products result in diabetic mouse vein graft atherosclerosis. Cell Death Dis 2017; 8:e2818. [PMID: 28542133 PMCID: PMC5520728 DOI: 10.1038/cddis.2017.213] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2016] [Revised: 03/12/2017] [Accepted: 04/05/2017] [Indexed: 01/08/2023]
Abstract
Protein disulfide isomerase (PDI) involves cell survival and death. Whether PDI mediates mechanical stretch stress (SS) and/or advanced glycosylation end products (AGEs) -triggered simultaneous increases in proliferation and apoptosis of vascular smooth muscle cells (VSMCs) is unknown. Here, we hypothesized that different expression levels of PDI trigger completely opposite cell fates among the different VSMC subtypes. Mouse veins were grafted into carotid arteries of non-diabetic and diabetic mice for 8 weeks; the grafted veins underwent simultaneous increases in proliferation and apoptosis, which triggered vein graft arterializations in non-diabetic or atherosclerosis in diabetic mice. A higher rate of proliferation and apoptosis was seen in the diabetic group. SS and/or AGEs stimulated the quiescent cultured VSMCs, resulting in simultaneous increases in proliferation and apoptosis; they could induce increased PDI activation and expression. Both in vivo and in vitro, the proliferating VSMCs indicated weak co-expression of PDI and SM-α-actin while apoptotic or dead cells showed strong co-expression of both. Either SS or AGEs rapidly upregulated the expression of PDI, NOX1 and ROS, and their combination had synergistic effects. Inhibiting PDI simultaneously suppressed the proliferation and apoptosis of VSMCs, while inhibition of SM-α-actin with cytochalasin D led to increased apoptosis and cleaved caspases-3 but had no effect on proliferation. In conclusion, different expression levels of PDI in VSMCs induced by SS and/or AGEs triggered a simultaneous increase in proliferation and apoptosis, accelerated vein graft arterializations or atherosclerosis, leading us to propose PDI as a novel target for the treatment of vascular remodeling and diseases.
Collapse
Affiliation(s)
- Suning Ping
- Department of Histology and Embryology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Shuying Liu
- Department of Histology and Embryology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Yuhuan Zhou
- Department of Histology and Embryology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Ziqing Li
- Department of Joint Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Yuhuang Li
- Department of Histology and Embryology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Kefeng Liu
- Department of Histology and Embryology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Adham Sa Bardeesi
- Department of Histology and Embryology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Linli Wang
- Department of Histology and Embryology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Jingbo Chen
- Department of Histology and Embryology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Lie Deng
- Department of Histology and Embryology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Jingjing Wang
- Department of Histology and Embryology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Hong Wang
- Department of Histology and Embryology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Dadi Chen
- Experimental Center for Basic Medical Teaching, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Zhengyu Zhang
- Department of Histology and Embryology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, China.,Department of Histology and Embryology, School of Basic Medicine, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Puyi Sheng
- Department of Joint Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Chaohong Li
- Department of Histology and Embryology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, China
| |
Collapse
|
22
|
Baish JW, Kunert C, Padera TP, Munn LL. Synchronization and Random Triggering of Lymphatic Vessel Contractions. PLoS Comput Biol 2016; 12:e1005231. [PMID: 27935958 PMCID: PMC5147819 DOI: 10.1371/journal.pcbi.1005231] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2016] [Accepted: 10/14/2016] [Indexed: 11/21/2022] Open
Abstract
The lymphatic system is responsible for transporting interstitial fluid back to the bloodstream, but unlike the cardiovascular system, lacks a centralized pump-the heart–to drive flow. Instead, each collecting lymphatic vessel can individually contract and dilate producing unidirectional flow enforced by intraluminal check valves. Due to the large number and spatial distribution of such pumps, high-level coordination would be unwieldy. This leads to the question of how each segment of lymphatic vessel responds to local signals that can contribute to the coordination of pumping on a network basis. Beginning with elementary fluid mechanics and known cellular behaviors, we show that two complementary oscillators emerge from i) mechanical stretch with calcium ion transport and ii) fluid shear stress induced nitric oxide production (NO). Using numerical simulation and linear stability analysis we show that the newly identified shear-NO oscillator shares similarities with the well-known Van der Pol oscillator, but has unique characteristics. Depending on the operating conditions, the shear-NO process may i) be inherently stable, ii) oscillate spontaneously in response to random disturbances or iii) synchronize with weak periodic stimuli. When the complementary shear-driven and stretch-driven oscillators interact, either may dominate, producing a rich family of behaviors similar to those observed in vivo. For decades, cardiovascular physiology has been an area of intense research, and we have a fundamental understanding of the mechanisms the heart uses to drive blood flow through the distributed network of vessels in the body. The lymphatic system is now receiving similar attention as more is learned about its functional role in disease processes. The importance of the lymphatic system in collecting excess fluid from tissues and returning it to the blood is well known, but how the lymph flow is regulated without a central pump is poorly understood. Each segment of collecting lymphatic vessel can independently contract yielding a network of distributed pump/conduits. This paper shows how the lymphatic muscle cells that squeeze fluid along the lymphatic vessels can be effectively regulated using only chemical and mechanical signals that they receive from their immediate microenvironment. Using stability theory and the tools of nonlinear dynamics we identify two complementary oscillators that respond to stretch of the vessel wall and shear of fluid flowing over the vessel wall. Numerical simulations of the combined oscillators show that they have characteristics well suited to the regulation of distributed systems in general and may have application in other biological and physical contexts.
Collapse
Affiliation(s)
- James W. Baish
- Department of Biomedical Engineering, Bucknell University, Lewisburg, Pennsylvania, United States of America
- * E-mail:
| | - Christian Kunert
- Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, United States of America
- AMGEN, Cambridge, Massachusetts, United States of America
| | - Timothy P. Padera
- Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, United States of America
| | - Lance L. Munn
- Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, United States of America
| |
Collapse
|
23
|
Tu AY, Cheung MC, Zhu X, Knopp RH, Albers JJ. Low-Density Lipoprotein Inhibits Secretion of Phospholipid Transfer Protein in Human Trophoblastic BeWo Cells. Exp Biol Med (Maywood) 2016; 229:1046-52. [PMID: 15522841 DOI: 10.1177/153537020422901009] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Human plasma phospholipid transfer protein (PLTP) plays an important role in lipoprotein metabolism. In this study, we investigated the effects of lipoproteins on the secretion of PLTP in cultured BeWo choriocarcinoma cells. Low-density lipoproteins (LDLs) decreased PLTP secretion in a dose- and time-dependent manner, whereas very low density lipoproteins and high-density lipoproteins (HDLs) had little effect. LDL suppression of PLTP secretion was not altered by the inhibition of both LDL receptor and LDL receptor–related protein with receptor-associated protein. Mitogen-activated protein kinase (MAPK) kinase (MEK) inhibitor, U0126, could abolish the LDL-mediated inhibition of PLTP secretion. Furthermore, LDL, but not HDL, could stimulate the expression of MAPK phosphatase-1 (MKP-1) in BeWo cells that resulted in the inactivation of p44/p42 extracellular signal-regulated kinase (ERK) 1 and 2, the family members of MAPKs. These results support the conclusion that LDL-mediated suppression of PLTP secretion in BeWo cells is through a LDL receptor-independent MAPK signaling pathway.
Collapse
Affiliation(s)
- An-Yue Tu
- Department of Medicine, Northwest Lipid Research Laboratories, Northwest Lipid Research Clinic, Division of Metabolism, Endocrinology, and Nutrition, University of Washington, 2121 N 35th Street, Seattle, WA 98103, USA.
| | | | | | | | | |
Collapse
|
24
|
Wang J, Liu K, Wang H, Li Z, Li Y, Ping S, Bardeesi ASA, Guo Y, Zhou Y, Pei T, Deng L, Sheng P, Liu S, Li C. Role of nifedipine and hydrochlorothiazide in MAPK activation and vascular smooth muscle cell proliferation and apoptosis. Herz 2016; 42:573-584. [DOI: 10.1007/s00059-016-4489-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2016] [Revised: 08/28/2016] [Accepted: 09/25/2016] [Indexed: 10/20/2022]
|
25
|
Ayoub S, Ferrari G, Gorman RC, Gorman JH, Schoen FJ, Sacks MS. Heart Valve Biomechanics and Underlying Mechanobiology. Compr Physiol 2016; 6:1743-1780. [PMID: 27783858 PMCID: PMC5537387 DOI: 10.1002/cphy.c150048] [Citation(s) in RCA: 56] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Heart valves control unidirectional blood flow within the heart during the cardiac cycle. They have a remarkable ability to withstand the demanding mechanical environment of the heart, achieving lifetime durability by processes involving the ongoing remodeling of the extracellular matrix. The focus of this review is on heart valve functional physiology, with insights into the link between disease-induced alterations in valve geometry, tissue stress, and the subsequent cell mechanobiological responses and tissue remodeling. We begin with an overview of the fundamentals of heart valve physiology and the characteristics and functions of valve interstitial cells (VICs). We then provide an overview of current experimental and computational approaches that connect VIC mechanobiological response to organ- and tissue-level deformations and improve our understanding of the underlying functional physiology of heart valves. We conclude with a summary of future trends and offer an outlook for the future of heart valve mechanobiology, specifically, multiscale modeling approaches, and the potential directions and possible challenges of research development. © 2016 American Physiological Society. Compr Physiol 6:1743-1780, 2016.
Collapse
Affiliation(s)
- Salma Ayoub
- Center for Cardiovascular Simulation, Institute for Computational Engineering and Sciences, Department of Biomedical Engineering, The University of Texas at Austin, Austin, USA
| | - Giovanni Ferrari
- Gorman Cardiovascular Research Group, University of Pennsylvania, Philadelphia, USA
| | - Robert C. Gorman
- Gorman Cardiovascular Research Group, University of Pennsylvania, Philadelphia, USA
| | - Joseph H. Gorman
- Gorman Cardiovascular Research Group, University of Pennsylvania, Philadelphia, USA
| | - Frederick J. Schoen
- Department of Pathology, Brigham and Women’s Hospital, Boston, Massachusetts, USA
| | - Michael S. Sacks
- Center for Cardiovascular Simulation, Institute for Computational Engineering and Sciences, Department of Biomedical Engineering, The University of Texas at Austin, Austin, USA
| |
Collapse
|
26
|
Murshid SA. The role of osteocytes during experimental orthodontic tooth movement: A review. Arch Oral Biol 2016; 73:25-33. [PMID: 27653146 DOI: 10.1016/j.archoralbio.2016.09.001] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2015] [Revised: 09/05/2016] [Accepted: 09/06/2016] [Indexed: 12/17/2022]
Abstract
OBJECTIVE To explore the types of orthodontic force-induced mechanical stimuli that regulate osteocyte function. DESIGN In orthodontics, a tooth can be moved through the alveolar bone when an appropriate orthodontic force is applied. These mechanical loads stimulate cells within the bone tissue around the tooth. These cellular responses lead to bone resorption on the side of the tooth where the pressure has been applied and bone deposition on the side of the tooth experiencing tension. Recently, osteocytes were identified to function as mechano-sensory cells in bone tissue that direct bone resorption and bone formation. Based on recent literature, the proposed function of osteocytes during orthodontic tooth movement is explored with better understanding. RESULTS Several stimuli regulating osteocyte function have been highlighted, and their potential roles in events initiating osteocyte sensing of orthodontic force have been explored in detail. The most popular hypotheses for osteocyte response include stress-induced bone matrix deformation/microcrack formation and fluid-flow shear stress. CONCLUSIONS Understanding osteocyte function under mechanical stress may have profound implications in future orthodontic treatments.
Collapse
Affiliation(s)
- Sakhr A Murshid
- Department of Pedodontics, Orthodontics and Preventive Dentistry, Faculty of Dentistry, Thamar University, Thamar City, Yemen.
| |
Collapse
|
27
|
Shi M, Wei J, Meng WY, Wang N, Wang T, Wang YG. Effects of phased joint intervention on Rho/ROCK expression levels in patients with portal hypertension. Exp Ther Med 2016; 12:1618-1624. [PMID: 27602079 DOI: 10.3892/etm.2016.3454] [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: 03/29/2015] [Accepted: 05/11/2016] [Indexed: 11/06/2022] Open
Abstract
The current study investigated the effects of phased joint intervention on clinical efficacy and Rho/Rho-associated coil protein kinase (ROCK) expression in patients with portal hypertension complicated by esophageal variceal bleeding (EVB) and hypersplenism. Patients with portal hypertension (n=53) caused by liver cirrhosis complicated by EVB and hypersplenism treated with phased joint intervention were assessed, and portal hemodynamics, blood, liver function, complications, and rebleeding incidence were analyzed. Reverse transcription-quantitative polymerase chain reaction was used to measure Rho, ROCK1 and ROCK2 mRNA expression levels in peripheral blood mononuclear cells prior to and following phased joint intervention, and western blotting was employed to determine the protein expression levels of Rho, ROCK1, ROCK2, phosphorylated (p) myosin phosphatase target subunit 1 (MYPT1) and total-MYPT1. All patients underwent an emergency assessment of hemostasis with a 100% success rate. Varicose veins were alleviated, and portal hemodynamics and liver function improved following intervention. Furthermore, preoperative and postoperative expression levels of Rho, ROCK1 and ROCK2 mRNA were higher compared with the control group. Notably, the mRNA expression levels of Rho, ROCK1 and ROCK2 in the postoperative group were significantly lower when compared with the preoperative group. Protein expression levels of Rho, ROCK1, ROCK2 and pMYPT1 in the postoperative group were lower, as compared with the preoperative group. Concentration levels of transforming growth factor-β1, connective tissue growth factor and platelet-derived growth factor in peripheral blood were significantly reduced following phased joint intervention. Therefore, the present findings demonstrated that phased joint intervention is able to effectively treat EVB and hypersplenism, and improve liver function. The efficacy of phased joint intervention may be associated with its role in the regulation of the Rho-ROCK signaling pathway.
Collapse
Affiliation(s)
- Min Shi
- Department of Gastroenterology, Shanghai Tongren Hospital, Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200336, P.R. China
| | - Jue Wei
- Department of Gastroenterology, Shanghai Tongren Hospital, Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200336, P.R. China
| | - Wen-Ying Meng
- Department of Gastroenterology, Shanghai Tongren Hospital, Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200336, P.R. China
| | - Na Wang
- Department of Gastroenterology, Shanghai Tongren Hospital, Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200336, P.R. China
| | - Ting Wang
- Department of Gastroenterology, Shanghai Tongren Hospital, Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200336, P.R. China
| | - Yu-Gang Wang
- Department of Gastroenterology, Shanghai Tongren Hospital, Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200336, P.R. China
| |
Collapse
|
28
|
Huang CH, Ciou JS, Chen ST, Kok VC, Chung Y, Tsai JJP, Kurubanjerdjit N, Huang CYF, Ng KL. Identify potential drugs for cardiovascular diseases caused by stress-induced genes in vascular smooth muscle cells. PeerJ 2016; 4:e2478. [PMID: 27703845 PMCID: PMC5045879 DOI: 10.7717/peerj.2478] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2016] [Accepted: 08/23/2016] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND Abnormal proliferation of vascular smooth muscle cells (VSMC) is a major cause of cardiovascular diseases (CVDs). Many studies suggest that vascular injury triggers VSMC dedifferentiation, which results in VSMC changes from a contractile to a synthetic phenotype; however, the underlying molecular mechanisms are still unclear. METHODS In this study, we examined how VSMC responds under mechanical stress by using time-course microarray data. A three-phase study was proposed to investigate the stress-induced differentially expressed genes (DEGs) in VSMC. First, DEGs were identified by using the moderated t-statistics test. Second, more DEGs were inferred by using the Gaussian Graphical Model (GGM). Finally, the topological parameters-based method and cluster analysis approach were employed to predict the last batch of DEGs. To identify the potential drugs for vascular diseases involve VSMC proliferation, the drug-gene interaction database, Connectivity Map (cMap) was employed. Success of the predictions were determined using in-vitro data, i.e. MTT and clonogenic assay. RESULTS Based on the differential expression calculation, at least 23 DEGs were found, and the findings were qualified by previous studies on VSMC. The results of gene set enrichment analysis indicated that the most often found enriched biological processes are cell-cycle-related processes. Furthermore, more stress-induced genes, well supported by literature, were found by applying graph theory to the gene association network (GAN). Finally, we showed that by processing the cMap input queries with a cluster algorithm, we achieved a substantial increase in the number of potential drugs with experimental IC50 measurements. With this novel approach, we have not only successfully identified the DEGs, but also improved the DEGs prediction by performing the topological and cluster analysis. Moreover, the findings are remarkably validated and in line with the literature. Furthermore, the cMap and DrugBank resources were used to identify potential drugs and targeted genes for vascular diseases involve VSMC proliferation. Our findings are supported by in-vitro experimental IC50, binding activity data and clinical trials. CONCLUSION This study provides a systematic strategy to discover potential drugs and target genes, by which we hope to shed light on the treatments of VSMC proliferation associated diseases.
Collapse
Affiliation(s)
- Chien-Hung Huang
- Department of Computer Science and Information Engineering, National Formosa University, Yun-Lin, Taiwan
| | - Jin-Shuei Ciou
- Department of Bioinformatics and Medical Engineering, Asia University, Taichung, Taiwan
| | - Shun-Tsung Chen
- Department of Bioinformatics and Medical Engineering, Asia University, Taichung, Taiwan
| | - Victor C. Kok
- Department of Bioinformatics and Medical Engineering, Asia University, Taichung, Taiwan
- Division of Medical Oncology, Kuang Tien General Hospital Cancer Center, Taichung, Taiwan
| | - Yi Chung
- Department of Bioinformatics and Medical Engineering, Asia University, Taichung, Taiwan
| | - Jeffrey J. P. Tsai
- Department of Bioinformatics and Medical Engineering, Asia University, Taichung, Taiwan
| | | | - Chi-Ying F. Huang
- Institute of Biopharmaceutical Sciences, National Yang-Ming University, Taipei, Taiwan
| | - Ka-Lok Ng
- Department of Bioinformatics and Medical Engineering, Asia University, Taichung, Taiwan
- Department of Medical Research, China Medical University Hospital, China Medical University, Taichung, Taiwan
| |
Collapse
|
29
|
Sehgel NL, Vatner SF, Meininger GA. "Smooth Muscle Cell Stiffness Syndrome"-Revisiting the Structural Basis of Arterial Stiffness. Front Physiol 2015; 6:335. [PMID: 26635621 PMCID: PMC4649054 DOI: 10.3389/fphys.2015.00335] [Citation(s) in RCA: 92] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2015] [Accepted: 11/02/2015] [Indexed: 02/05/2023] Open
Abstract
In recent decades, the pervasiveness of increased arterial stiffness in patients with cardiovascular disease has become increasingly apparent. Though, this phenomenon has been well documented in humans and animal models of disease for well over a century, there has been surprisingly limited development in a deeper mechanistic understanding of arterial stiffness. Much of the historical literature has focused on changes in extracellular matrix proteins—collagen and elastin. However, extracellular matrix changes alone appear insufficient to consistently account for observed changes in vascular stiffness, which we observed in our studies of aortic stiffness in aging monkeys. This led us to examine novel mechanisms operating at the level of the vascular smooth muscle cell (VSMC)—that include increased cell stiffness and adhesion to extracellular matrix—which that may be interrelated with other mechanisms contributing to arterial stiffness. We introduce these observations as a new concept—the Smooth Muscle Cell Stiffness Syndrome (SMCSS)—within the field of arterial stiffness and posit that stiffening of vascular cells impairs vascular function and may contribute stiffening to the vasculature with aging and cardiovascular disease. Importantly, this review article revisits the structural basis of arterial stiffness in light of these novel findings. Such classification of SMCSS and its contextualization into our current understanding of vascular mechanics may be useful in the development of strategic therapeutics to directly target arterial stiffness.
Collapse
Affiliation(s)
- Nancy L Sehgel
- Department of Cell Biology and Molecular Medicine, New Jersey Medical School, Rutgers University - Biomedical and Health Sciences Newark, NJ, USA ; Department of Biomedical Engineering, New Jersey Institute of Technology Newark, NJ, USA
| | - Stephen F Vatner
- Department of Biomedical Engineering, New Jersey Institute of Technology Newark, NJ, USA
| | - Gerald A Meininger
- Dalton Cardiovascular Research Center, Department of Medical Pharmacology and Physiology, University of Missouri Columbia, MO, USA
| |
Collapse
|
30
|
Lee CH, Carruthers CA, Ayoub S, Gorman RC, Gorman JH, Sacks MS. Quantification and simulation of layer-specific mitral valve interstitial cells deformation under physiological loading. J Theor Biol 2015; 373:26-39. [PMID: 25791285 PMCID: PMC4404233 DOI: 10.1016/j.jtbi.2015.03.004] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2014] [Revised: 02/12/2015] [Accepted: 03/03/2015] [Indexed: 01/15/2023]
Abstract
Within each of the four layers of mitral valve (MV) leaflet tissues there resides a heterogeneous population of interstitial cells that maintain the structural integrity of the MV tissue via protein biosynthesis and enzymatic degradation. There is increasing evidence that tissue stress-induced MV interstitial cell (MVIC) deformations can have deleterious effects on their biosynthetic states that are potentially related to the reduction of tissue-level maintenance and to subsequent organ-level failure. To better understand the interrelationships between tissue-level loading and cellular responses, we developed the following integrated experimental-computational approach. Since in vivo cellular deformations are not directly measurable, we quantified the in-situ layer-specific MVIC deformations for each of the four layers under a controlled biaxial tension loading device coupled to multi-photon microscopy. Next, we explored the interrelationship between the MVIC stiffness and deformation to layer-specific tissue mechanical and structural properties using a macro-micro finite element computational model. Experimental results indicated that the MVICs in the fibrosa and ventricularis layers deformed significantly more than those in the atrialis and spongiosa layers, reaching a nucleus aspect ratio of 3.3 under an estimated maximum physiological tension of 150N/m. The simulated MVIC moduli for the four layers were found to be all within a narrow range of 4.71-5.35kPa, suggesting that MVIC deformation is primarily controlled by each tissue layer's respective structure and mechanical behavior rather than the intrinsic MVIC stiffness. This novel result further suggests that while the MVICs may be phenotypically and biomechanically similar throughout the leaflet, they experience layer-specific mechanical stimulatory inputs due to distinct extracellular matrix architecture and mechanical behaviors of the four MV leaflet tissue layers. This also suggests that MVICs may behave in a layer-specific manner in response to mechanical stimuli in both normal and surgically modified MVs.
Collapse
Affiliation(s)
- Chung-Hao Lee
- Center for Cardiovascular Simulation, Institute for Computational Engineering and Sciences (ICES), Department of Biomedical Engineering, The University of Texas at Austin, 201 East 24th Street, POB 5.236, 1 University Station C0200, Austin, TX 78712, USA
| | - Christopher A Carruthers
- Cardiac Rhythm Disease Management (CRDM) Clinical Specialist, Medtronic, Minneapolis, MN 55432, USA
| | - Salma Ayoub
- Center for Cardiovascular Simulation, Institute for Computational Engineering and Sciences (ICES), Department of Biomedical Engineering, The University of Texas at Austin, 201 East 24th Street, POB 5.236, 1 University Station C0200, Austin, TX 78712, USA
| | - Robert C Gorman
- Gorman Cardiovascular Research Group, University of Pennsylvania, 3400 Civic Center Blvd, Philadelphia, PA 19104, USA
| | - Joseph H Gorman
- Gorman Cardiovascular Research Group, University of Pennsylvania, 3400 Civic Center Blvd, Philadelphia, PA 19104, USA
| | - Michael S Sacks
- Center for Cardiovascular Simulation, Institute for Computational Engineering and Sciences (ICES), Department of Biomedical Engineering, The University of Texas at Austin, 201 East 24th Street, POB 5.236, 1 University Station C0200, Austin, TX 78712, USA.
| |
Collapse
|
31
|
Lee KP, Sudjarwo GW, Jung SH, Lee D, Lee DY, Lee GB, Baek S, Kim DY, Lee HM, Kim B, Kwon SC, Won KJ. Carvacrol inhibits atherosclerotic neointima formation by downregulating reactive oxygen species production in vascular smooth muscle cells. Atherosclerosis 2015; 240:367-73. [PMID: 25875388 DOI: 10.1016/j.atherosclerosis.2015.03.038] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/10/2014] [Revised: 03/01/2015] [Accepted: 03/21/2015] [Indexed: 10/23/2022]
Abstract
OBJECTIVE Carvacrol (2-methyl-5-(1-methylethyl) phenol), a cyclic monoterpene, exerts protective activities in a variety of pathological states including tumor growth, inflammation, and oxidative stress. However, it is unknown whether carvacrol affects events in vascular cells during the development of atherosclerotic neointima. We investigated the effects of carvacrol on the migration and proliferation of rat aortic smooth muscle cells (RASMCs) and on vascular neointima formation. METHODS AND RESULTS Carvacrol significantly inhibited platelet-derived growth factor (PDGF)-BB-stimulated RASMC migration and proliferation in a concentration-dependent manner. Cell viability was not affected by treatment with carvacrol. Carvacrol attenuated the expression of NADPH oxidase (NOX) 1 and the phosphorylation of p38 mitogen-activated protein kinase (MAPK) and extracellular signal-regulated kinase 1/2 in response to PDGF-BB. Moreover, carvacrol suppressed the PDGF-BB-stimulated generation of H2O2 and inhibited the activity of NOX in RASMCs. Treatment with carvacrol inhibited PDGF-BB-induced aortic sprout outgrowth, balloon injury-evoked vascular neointima formation, and expression of proliferating cell nuclear antigen in the neointima. CONCLUSION These findings indicate that carvacrol inhibits migration and proliferation of RASMCs by suppressing the reactive oxygen species-mediated MAPK signaling pathway in these cells, thereby attenuating vascular neointimal formation. Carvacrol may be a promising agent for preventing vascular restenosis or atherosclerosis.
Collapse
Affiliation(s)
- Kang Pa Lee
- Department of Physiology, School of Medicine, Konkuk University, 1 Hwayang-dong, Gwangjin-gu, Seoul 143-701, South Korea
| | - Giftania W Sudjarwo
- Department of Physiology, School of Medicine, Konkuk University, 1 Hwayang-dong, Gwangjin-gu, Seoul 143-701, South Korea
| | - Seung Hyo Jung
- Department of Physiology, School of Medicine, Konkuk University, 1 Hwayang-dong, Gwangjin-gu, Seoul 143-701, South Korea
| | - Donghyen Lee
- Department of Physiology, School of Medicine, Konkuk University, 1 Hwayang-dong, Gwangjin-gu, Seoul 143-701, South Korea
| | - Dong-Youb Lee
- Department of Physiology, School of Medicine, Konkuk University, 1 Hwayang-dong, Gwangjin-gu, Seoul 143-701, South Korea
| | - Gyoung Beom Lee
- Department of Physiology, School of Medicine, Konkuk University, 1 Hwayang-dong, Gwangjin-gu, Seoul 143-701, South Korea
| | - Suji Baek
- Department of Physiology, School of Medicine, Konkuk University, 1 Hwayang-dong, Gwangjin-gu, Seoul 143-701, South Korea
| | - Do-Yoon Kim
- Department of Cosmetic Science, College of Natural Science, Hoseo University, Asan 336-795, South Korea
| | - Hwan Myung Lee
- Department of Cosmetic Science, College of Natural Science, Hoseo University, Asan 336-795, South Korea
| | - Bokyung Kim
- Department of Physiology, School of Medicine, Konkuk University, 1 Hwayang-dong, Gwangjin-gu, Seoul 143-701, South Korea
| | - Seong-Chun Kwon
- Department of Physiology, Catholic Kwandong University College of Medicine, Kangneung 201-701, South Korea.
| | - Kyung Jong Won
- Department of Physiology, School of Medicine, Konkuk University, 1 Hwayang-dong, Gwangjin-gu, Seoul 143-701, South Korea.
| |
Collapse
|
32
|
Complement C1q-induced activation of β-catenin signalling causes hypertensive arterial remodelling. Nat Commun 2015; 6:6241. [PMID: 25716000 PMCID: PMC4351572 DOI: 10.1038/ncomms7241] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2013] [Accepted: 01/08/2015] [Indexed: 12/21/2022] Open
Abstract
Hypertension induces structural remodelling of arteries, which leads to arteriosclerosis and end-organ damage. Hyperplasia of vascular smooth muscle cells (VSMCs) and infiltration of immune cells are the hallmark of hypertensive arterial remodelling. However, the precise molecular mechanisms of arterial remodelling remain elusive. We have recently reported that complement C1q activates β-catenin signalling independent of Wnts. Here, we show a critical role of complement C1-induced activation of β-catenin signalling in hypertensive arterial remodelling. Activation of β-catenin and proliferation of VSMCs were observed after blood-pressure elevation, which were prevented by genetic and chemical inhibition of β-catenin signalling. Macrophage depletion and C1qa gene deletion attenuated the hypertension-induced β-catenin signalling, proliferation of VSMCs and pathological arterial remodelling. Our findings unveil the link between complement C1 and arterial remodelling and suggest that C1-induced activation of β-catenin signalling becomes a novel therapeutic target to prevent arteriosclerosis in patients with hypertension. The role of macrophages in hypertension-induced arterial remodeling is poorly understood. Here, Sumida et al. show that high blood pressure drives the alternatively activated macrophages to secrete complement C1q protein, which in turn elicits proliferative β-catenin signalling in the arterial smooth muscle cells.
Collapse
|
33
|
Sen S, Diercke K, Zingler S, Lux CJ, Erber R. Compression induces Ephrin-A2 in PDL fibroblasts via c-fos. J Dent Res 2015; 94:464-72. [PMID: 25604255 DOI: 10.1177/0022034514567197] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Abstract
Ephrin-A2-EphA2 and ephrin-B2-EphB4 interactions have been implicated in the regulation of bone remodeling. We previously demonstrated a potential role for members of the Eph-ephrin family of receptor tyrosine kinases for bone remodeling during orthodontic tooth movement: compression-dependent upregulation of ephrin-A2 in fibroblasts of the periodontal ligament (PDL) attenuated osteogenesis in osteoblasts of the alveolar bone. However, factors affecting the regulation of ephrin-A2 expression upon the application of compressive forces remained unclear. Here, we report a mechano-dependent pathway of ephrin-A2 induction in PDL fibroblasts (PDLFs) involving extracellular signal-regulated kinases (ERK) 1/2 and c-fos. PDLF subjected to compressive forces (30.3 g/cm(2)) upregulated c-fos and ephrin-A2 mRNA and protein expression and displayed increased ERK1/2 phosphorylation. Inhibition of the MAP kinase kinase (MEK)/ERK1/2 pathway using the specific MEK inhibitor U0126 significantly reduced ephrin-A2 messenger RNA upregulation upon compression. Silencing of c-fos using a small interfering RNA approach led to a significant inhibition of ephrin-A2 induction upon the application of compressive forces. Interestingly, ephrin-A2 stimulation of PDLF induced c-fos expression and led also to the induction of ephrin-A2 expression. Using a reporter gene construct in murine 3T3 cells, we found that ephrin-A2 was able to stimulate serum response element (SRE)-dependent luciferase activity. As the regulation of c-fos is SRE dependent, ephrin-A2 might induce c-fos via SRE activation. Taken together, we provide evidence for an ERK1/2- and c-fos-dependent regulation of ephrin-A2 in compressed PDLF and suggest a novel pathway for ephrin-A2 induction emanating from ephrin-A2 itself. We showed previously that ephrin-A2 at compression sites might contribute to tooth movement by inhibiting osteogenic differentiation. The regulatory pathway of ephrin-A2 induction during tooth movement identified in this study might be accessible for pharmacological interventions.
Collapse
Affiliation(s)
- S Sen
- Department of Orthodontics and Dentofacial Orthopaedics, Dental School, University of Heidelberg, Heidelberg, Germany
| | - K Diercke
- Department of Orthodontics and Dentofacial Orthopaedics, Dental School, University of Heidelberg, Heidelberg, Germany
| | - S Zingler
- Department of Orthodontics and Dentofacial Orthopaedics, Dental School, University of Heidelberg, Heidelberg, Germany
| | - C J Lux
- Department of Orthodontics and Dentofacial Orthopaedics, Dental School, University of Heidelberg, Heidelberg, Germany
| | - R Erber
- Department of Orthodontics and Dentofacial Orthopaedics, Dental School, University of Heidelberg, Heidelberg, Germany
| |
Collapse
|
34
|
Variation in Cardiac Pulse Frequencies Modulates vSMC Phenotype Switching During Vascular Remodeling. Cardiovasc Eng Technol 2014; 6:59-70. [PMID: 26577103 DOI: 10.1007/s13239-014-0204-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/01/2014] [Accepted: 11/07/2014] [Indexed: 01/22/2023]
Abstract
In vitro perfusion systems have exposed vascular constructs to mechanical conditions that emulate physiological pulse pressure and found significant improvements in graft development. However, current models maintain constant, or set pulse/shear mechanics that do not account for the natural temporal variation in frequency. With an aim to develop clinically relevant small diameter vascular grafts, these investigations detail a perfusion culture model that incorporates temporal pulse pressure variation. Our objective was to test the hypothesis that short-term variation in heart rate, such as changes in respiratory activity, plays a significant role in vascular remodeling and graft development. The pulse rate of a healthy volunteer was logged to model the effect of daily activities on heart rate. Vascular bioreactors were used to deliver perfusion conditions based on modeled frequencies of temporal pulse variability, termed Physiologically Modeled Pulse Dynamics (PMPD). Acellular scaffolds derived from the human umbilical vein were seeded with human vascular smooth muscle cells and perfused under defined pulsatile conditions. vSMC exposed to constant pulse frequencies expressed a contractile phenotype, while exposure to PMPD drove cells to a synthetic state with continued cell proliferation, increased tensile strength and stiffness as well as diminished vasoactivity. Results show the temporal variation associated with normal heart physiology to have a profound effect on vascular remodeling and vasoactive function. While these models are representative of vascular regeneration further investigation is required to understanding these and other key regulators in vSMC phenotype switching in non-pathological or wound healing states. This understanding has important clinical implications that may lead to improved treatments that enhance vessel regeneration.
Collapse
|
35
|
Otten M, Ott W, Jobst MA, Milles LF, Verdorfer T, Pippig DA, Nash MA, Gaub HE. From genes to protein mechanics on a chip. Nat Methods 2014; 11:1127-1130. [PMID: 25194847 PMCID: PMC4216144 DOI: 10.1038/nmeth.3099] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2014] [Accepted: 07/29/2014] [Indexed: 01/28/2023]
Abstract
Single-molecule force spectroscopy enables mechanical testing of individual proteins, but low experimental throughput limits the ability to screen constructs in parallel. We describe a microfluidic platform for on-chip expression, covalent surface attachment and measurement of single-molecule protein mechanical properties. A dockerin tag on each protein molecule allowed us to perform thousands of pulling cycles using a single cohesin-modified cantilever. The ability to synthesize and mechanically probe protein libraries enables high-throughput mechanical phenotyping.
Collapse
Affiliation(s)
- Marcus Otten
- Lehrstuhl für Angewandte Physik, Ludwig-Maximilians-Universität, 80799 Munich, Germany
- Center for Nanoscience (CeNS), Ludwig-Maximilians-Universität, 80799 Munich, Germany
| | - Wolfgang Ott
- Lehrstuhl für Angewandte Physik, Ludwig-Maximilians-Universität, 80799 Munich, Germany
- Center for Nanoscience (CeNS), Ludwig-Maximilians-Universität, 80799 Munich, Germany
| | - Markus A Jobst
- Lehrstuhl für Angewandte Physik, Ludwig-Maximilians-Universität, 80799 Munich, Germany
- Center for Nanoscience (CeNS), Ludwig-Maximilians-Universität, 80799 Munich, Germany
| | - Lukas F Milles
- Lehrstuhl für Angewandte Physik, Ludwig-Maximilians-Universität, 80799 Munich, Germany
- Center for Nanoscience (CeNS), Ludwig-Maximilians-Universität, 80799 Munich, Germany
| | - Tobias Verdorfer
- Lehrstuhl für Angewandte Physik, Ludwig-Maximilians-Universität, 80799 Munich, Germany
- Center for Nanoscience (CeNS), Ludwig-Maximilians-Universität, 80799 Munich, Germany
| | - Diana A Pippig
- Lehrstuhl für Angewandte Physik, Ludwig-Maximilians-Universität, 80799 Munich, Germany
- Center for Nanoscience (CeNS), Ludwig-Maximilians-Universität, 80799 Munich, Germany
- Center for Integrated Protein Science Munich (CIPSM), Ludwig-Maximilians-Universität, 80799 Munich, Germany
| | - Michael A Nash
- Lehrstuhl für Angewandte Physik, Ludwig-Maximilians-Universität, 80799 Munich, Germany
- Center for Nanoscience (CeNS), Ludwig-Maximilians-Universität, 80799 Munich, Germany
| | - Hermann E Gaub
- Lehrstuhl für Angewandte Physik, Ludwig-Maximilians-Universität, 80799 Munich, Germany
- Center for Nanoscience (CeNS), Ludwig-Maximilians-Universität, 80799 Munich, Germany
| |
Collapse
|
36
|
Sreenivasappa H, Chaki SP, Lim SM, Trzeciakowski JP, Davidson MW, Rivera GM, Trache A. Selective regulation of cytoskeletal tension and cell–matrix adhesion by RhoA and Src. Integr Biol (Camb) 2014; 6:743-54. [DOI: 10.1039/c4ib00019f] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
|
37
|
Takano-Yamamoto T. Osteocyte function under compressive mechanical force. JAPANESE DENTAL SCIENCE REVIEW 2014. [DOI: 10.1016/j.jdsr.2013.10.004] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
|
38
|
Thromboxane A(2) receptor stimulation promotes closure of the rat ductus arteriosus through enhancing neointima formation. PLoS One 2014; 9:e94895. [PMID: 24736499 PMCID: PMC3988076 DOI: 10.1371/journal.pone.0094895] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2014] [Accepted: 03/20/2014] [Indexed: 12/02/2022] Open
Abstract
Ductus arteriosus (DA) closure follows constriction and remodeling of the entire vessel wall. Patent ductus arteriosus occurs when the DA does not close after birth, and this condition is currently treated using cyclooxygenase inhibitors. However, the efficacy of cyclooxygenase inhibitors is often limited. Our previous study demonstrated that low-dose thromboxane A2 receptor (TP) stimulation constricted the DA with minimal adverse effects in rat neonates. However, its effect on DA remodeling remains unknown. In this study, we focused on the impact of the exogenous TP stimulation on the DA remodeling, especially intimal thickening. Using DA explants from rat fetuses at embryonic day 19 as a ex vivo model and primary cultured rat DA smooth muscle cells from embryonic day 21 as a in vitro model, we evaluated the effect of TP stimulation on the DA remodeling. The selective TP agonists U46619 and I-BOP promoted neointima formation in the ex vivo DA explants, and TP stimulation increased DA SMC migration in a dose-dependent manner. Both effects were inhibited by the selective TP antagonist SQ29548 or the siRNA against TP. TP stimulation also increased DA SMC proliferation in the presence of 10% fetal bovine serum. LC/MS/MS analysis revealed that TP stimulation increased secretion of several extracellular matrix proteins that may contribute to an increase in neointima formation. In conclusion, we uncovered that exogenous administration of TP agonist promotes neointima formation through the induction of migration and proliferation of DA SMC, which could contribute to DA closure and also to its vasoconstrictive action.
Collapse
|
39
|
Heger M, van Golen RF, Broekgaarden M, van den Bos RR, Neumann HAM, van Gulik TM, van Gemert MJC. Endovascular laser–tissue interactions and biological responses in relation to endovenous laser therapy. Lasers Med Sci 2013; 29:405-22. [DOI: 10.1007/s10103-013-1490-3] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2013] [Accepted: 11/03/2013] [Indexed: 01/11/2023]
|
40
|
Chen M, Zhang W, Lu X, Hoggatt AM, Gunst SJ, Kassab GS, Tune JD, Herring BP. Regulation of 130-kDa smooth muscle myosin light chain kinase expression by an intronic CArG element. J Biol Chem 2013; 288:34647-57. [PMID: 24151072 DOI: 10.1074/jbc.m113.510362] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023] Open
Abstract
The mylk1 gene encodes a 220-kDa nonmuscle myosin light chain kinase (MLCK), a 130-kDa smooth muscle MLCK (smMLCK), as well as the non-catalytic product telokin. Together, these proteins play critical roles in regulating smooth muscle contractility. Changes in their expression are associated with many pathological conditions; thus, it is important to understand the mechanisms regulating expression of mylk1 gene transcripts. Previously, we reported a highly conserved CArG box, which binds serum response factor, in intron 15 of mylk1. Because this CArG element is near the promoter that drives transcription of the 130-kDa smMLCK, we examined its role in regulating expression of this transcript. Results show that deletion of the intronic CArG region from a β-galactosidase reporter gene abolished transgene expression in mice in vivo. Deletion of the CArG region from the endogenous mylk1 gene, specifically in smooth muscle cells, decreased expression of the 130-kDa smMLCK by 40% without affecting expression of the 220-kDa MLCK or telokin. This reduction in 130-kDa smMLCK expression resulted in decreased phosphorylation of myosin light chains, attenuated smooth muscle contractility, and a 24% decrease in small intestine length that was associated with a significant reduction of Ki67-positive smooth muscle cells. Overall, these data show that the CArG element in intron 15 of the mylk1 gene is necessary for maximal expression of the 130-kDa smMLCK and that the 130-kDa smMLCK isoform is specifically required to regulate smooth muscle contractility and small intestine smooth muscle cell proliferation.
Collapse
Affiliation(s)
- Meng Chen
- From the Department of Cellular and Integrative Physiology, Indiana University School of Medicine, Indianapolis, Indiana 46202
| | | | | | | | | | | | | | | |
Collapse
|
41
|
Hatami J, Tafazzoli-Shadpour M, Haghighipour N, Shokrgozar MA, Janmaleki M. Influence of Cyclic Stretch on Mechanical Properties of Endothelial Cells. EXPERIMENTAL MECHANICS 2013; 53:1291-1298. [DOI: 10.1007/s11340-013-9744-3] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/06/2024]
|
42
|
Seo KW, Lee SJ, Kim YH, Bae JU, Park SY, Bae SS, Kim CD. Mechanical stretch increases MMP-2 production in vascular smooth muscle cells via activation of PDGFR-β/Akt signaling pathway. PLoS One 2013; 8:e70437. [PMID: 23950935 PMCID: PMC3737227 DOI: 10.1371/journal.pone.0070437] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2013] [Accepted: 06/18/2013] [Indexed: 12/21/2022] Open
Abstract
Increased blood pressure, leading to mechanical stress on vascular smooth muscle cells (VSMC), is a known risk factor for vascular remodeling via increased activity of matrix metalloproteinase (MMP) within the vascular wall. This study aimed to identify cell surface mechanoreceptors and intracellular signaling pathways that influence VSMC to produce MMP in response to mechanical stretch (MS). When VSMC was stimulated with MS (0–10% strain, 60 cycles/min), both production and gelatinolytic activity of MMP-2, but not MMP-9, were increased in a force-dependent manner. MS-enhanced MMP-2 expression and activity were inhibited by molecular inhibition of Akt using Akt siRNA as well as by PI3K/Akt inhibitors, LY293002 and AI, but not by MAPK inhibitors such as PD98059, SP600125 and SB203580. MS also increased Akt phosphorylation in VSMC, which was attenuated by AG1295, a PDGF receptor (PDGFR) inhibitor, but not by inhibitors for other receptor tyrosine kinase including EGF, IGF, and FGF receptors. Although MS activated PDGFR-α as well as PDGFR-β in VSMC, MS-induced Akt phosphorylation was inhibited by molecular deletion of PDGFR-β using siRNA, but not by inhibition of PDGFR-α. Collectively, our data indicate that MS induces MMP-2 production in VSMC via activation of Akt pathway, that is mediated by activation of PDGFR-β signaling pathways.
Collapse
MESH Headings
- Animals
- Cells, Cultured
- Gene Expression
- Immunoblotting
- Immunohistochemistry
- Matrix Metalloproteinase 2/genetics
- Matrix Metalloproteinase 2/metabolism
- Microscopy, Confocal
- Muscle, Smooth, Vascular/cytology
- Muscle, Smooth, Vascular/metabolism
- Myocytes, Smooth Muscle/cytology
- Myocytes, Smooth Muscle/metabolism
- Phosphorylation
- Proto-Oncogene Proteins c-akt/antagonists & inhibitors
- Proto-Oncogene Proteins c-akt/genetics
- Proto-Oncogene Proteins c-akt/metabolism
- RNA Interference
- Rats
- Rats, Sprague-Dawley
- Reactive Oxygen Species/metabolism
- Receptor, Platelet-Derived Growth Factor beta/antagonists & inhibitors
- Receptor, Platelet-Derived Growth Factor beta/genetics
- Receptor, Platelet-Derived Growth Factor beta/metabolism
- Reverse Transcriptase Polymerase Chain Reaction
- Signal Transduction
- Stress, Mechanical
- Tyrphostins/pharmacology
Collapse
Affiliation(s)
- Kyo Won Seo
- Department of Pharmacology, School of Medicine, and Medical Research Center for Ischemic Tissue Regeneration, Pusan National University, Yangsan, Gyeongnam, Republic of Korea
| | - Seung Jin Lee
- Department of Pharmacology, School of Medicine, and Medical Research Center for Ischemic Tissue Regeneration, Pusan National University, Yangsan, Gyeongnam, Republic of Korea
| | - Yun Hak Kim
- Department of Pharmacology, School of Medicine, and Medical Research Center for Ischemic Tissue Regeneration, Pusan National University, Yangsan, Gyeongnam, Republic of Korea
| | - Jin Ung Bae
- Department of Pharmacology, School of Medicine, and Medical Research Center for Ischemic Tissue Regeneration, Pusan National University, Yangsan, Gyeongnam, Republic of Korea
| | - So Youn Park
- Department of Pharmacology, School of Medicine, and Medical Research Center for Ischemic Tissue Regeneration, Pusan National University, Yangsan, Gyeongnam, Republic of Korea
| | - Sun Sik Bae
- Department of Pharmacology, School of Medicine, and Medical Research Center for Ischemic Tissue Regeneration, Pusan National University, Yangsan, Gyeongnam, Republic of Korea
| | - Chi Dae Kim
- Department of Pharmacology, School of Medicine, and Medical Research Center for Ischemic Tissue Regeneration, Pusan National University, Yangsan, Gyeongnam, Republic of Korea
- * E-mail:
| |
Collapse
|
43
|
Abstract
It is well known that the altered blood flow is related to vascular diseases, including atherosclerosis, restenosis, and arteriosclerosis, which preferentially located at areas with the disturbed blood flow, suggesting that altered biomechanical stress may exert their effect on the vascular disease. Recent evidence indicated the presence of abundant stem/progenitor cells in the vessel wall, in which laminar shear stress can stimulate these cells to differentiate towards endothelial lineage, while cyclic strain results in smooth muscle differentiation. In line with this, it was evidenced that altered biomechanical stress in stented vessels may lead to 'wrong' direction of vascular stem cell differentiation resulting in restenosis. However, the underlying mechanisms are not well understood. In this article, we will give an overview of the effect of the local flow pattern on stem/progenitor cell differentiation and the possible mechanism on how the blood flow influences stem cell behaviours in the development of vascular diseases.
Collapse
Affiliation(s)
- Cheng Zhang
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Health, Shandong University Qilu Hospital, Jinan, China
| | | | | | | |
Collapse
|
44
|
Chen LJ, Wei SY, Chiu JJ. Mechanical regulation of epigenetics in vascular biology and pathobiology. J Cell Mol Med 2013; 17:437-48. [PMID: 23551392 PMCID: PMC3822644 DOI: 10.1111/jcmm.12031] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2012] [Accepted: 01/09/2013] [Indexed: 12/16/2022] Open
Abstract
Vascular endothelial cells (ECs) and smooth muscle cells (VSMCs) are constantly exposed to haemodynamic forces, including blood flow-induced fluid shear stress and cyclic stretch from blood pressure. These forces modulate vascular cell gene expression and function and, therefore, influence vascular physiology and pathophysiology in health and disease. Epigenetics, including DNA methylation, histone modification/chromatin remodelling and RNA-based machinery, refers to the study of heritable changes in gene expression that occur without changes in the DNA sequence. The role of haemodynamic force-induced epigenetic modifications in the regulation of vascular gene expression and function has recently been elucidated. This review provides an introduction to the epigenetic concepts that relate to vascular physiology and pathophysiology. Through the studies of gene expression, cell proliferation, angiogenesis, migration and pathophysiological states, we present a conceptual framework for understanding how mechanical force-induced epigenetic modifications work to control vascular gene expression and function and, hence, the development of vascular disorders. This research contributes to our knowledge of how the mechanical environment impacts the chromatin state of ECs and VSMCs and the consequent cellular behaviours.
Collapse
Affiliation(s)
- Li-Jing Chen
- Institute of Cellular and System Medicine, National Health Research Institutes, Miaoli 350, Taiwan
| | | | | |
Collapse
|
45
|
Associated inflammation or increased flow-mediated shear stress, but not pressure alone, disrupts endothelial caveolin-1 in infants with pulmonary hypertension. Pulm Circ 2013. [PMID: 23372934 DOI: 10.4103/2045-8932] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Endothelial caveolin-1 loss is an important feature of pulmonary hypertension (PH); the rescue of caveolin-1 abrogates experimental PH. Recent studies in human PH suggest that the endothelial caveolin-1 loss is followed by an enhanced expression of caveolin-1 in smooth muscle cells (SMC) with subsequent neointima formation. In order to evaluate caveolin-1 expression in infants with PH, we examined the available clinical histories, hemodynamic data, and the expression of caveolin-1, PECAM-1, vWF, and smooth muscle α-actin in the lung biopsy/autopsy specimens obtained from infants with congenital heart disease (CHD, n = 8) and lung disease (n = 9). In CHD group, PH associated with increased pulmonary blood flow exhibited loss of endothelial caveolin-1 and PECAM-1 in pulmonary arteries; additional vWF loss was associated with enhanced expression of caveolin-1 in SMC. In the absence of PH, increased or decreased pulmonary blood flow did not disrupt endothelial caveolin-1, PECAM-1, or vWF; nor was there any enhanced expression of caveolin-1 in SMC. In Lung Disease + PH group, caveolin-1, PECAM-1, and vWF were well preserved in seven infants, and importantly, SMC in these arteries did not exhibit enhanced caveolin-1 expression. Two infants with associated inflammatory disease exhibited loss of endothelial caveolin-1 and PECAM-1; additional loss of vWF was accompanied by enhanced expression of caveolin-1 in SMC. Thus, associated flow-induced shear stress or inflammation, but not elevated pulmonary artery pressure alone, disrupts endothelial caveolin-1. Subsequent vWF loss, indicative of extensive endothelial damage is associated with enhanced expression of caveolin-1 in SMC, which may worsen the disease.
Collapse
|
46
|
Dereddy N, Huang J, Erb M, Guzel S, Wolk JH, Sett SS, Gewitz MH, Mathew R. Associated inflammation or increased flow-mediated shear stress, but not pressure alone, disrupts endothelial caveolin-1 in infants with pulmonary hypertension. Pulm Circ 2013; 2:492-500. [PMID: 23372934 PMCID: PMC3555420 DOI: 10.4103/2045-8932.105038] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Endothelial caveolin-1 loss is an important feature of pulmonary hypertension (PH); the rescue of caveolin-1 abrogates experimental PH. Recent studies in human PH suggest that the endothelial caveolin-1 loss is followed by an enhanced expression of caveolin-1 in smooth muscle cells (SMC) with subsequent neointima formation. In order to evaluate caveolin-1 expression in infants with PH, we examined the available clinical histories, hemodynamic data, and the expression of caveolin-1, PECAM-1, vWF, and smooth muscle α-actin in the lung biopsy/autopsy specimens obtained from infants with congenital heart disease (CHD, n = 8) and lung disease (n = 9). In CHD group, PH associated with increased pulmonary blood flow exhibited loss of endothelial caveolin-1 and PECAM-1 in pulmonary arteries; additional vWF loss was associated with enhanced expression of caveolin-1 in SMC. In the absence of PH, increased or decreased pulmonary blood flow did not disrupt endothelial caveolin-1, PECAM-1, or vWF; nor was there any enhanced expression of caveolin-1 in SMC. In Lung Disease + PH group, caveolin-1, PECAM-1, and vWF were well preserved in seven infants, and importantly, SMC in these arteries did not exhibit enhanced caveolin-1 expression. Two infants with associated inflammatory disease exhibited loss of endothelial caveolin-1 and PECAM-1; additional loss of vWF was accompanied by enhanced expression of caveolin-1 in SMC. Thus, associated flow-induced shear stress or inflammation, but not elevated pulmonary artery pressure alone, disrupts endothelial caveolin-1. Subsequent vWF loss, indicative of extensive endothelial damage is associated with enhanced expression of caveolin-1 in SMC, which may worsen the disease.
Collapse
Affiliation(s)
- Narendra Dereddy
- Section of Pediatric Cardiology, Maria Fareri Children's Hospital, New York Medical College, Valhalla, New York, USA
| | | | | | | | | | | | | | | |
Collapse
|
47
|
Chun J, Joo EJ, Kang M, Kim YS. Platycodin D induces anoikis and caspase-mediated apoptosis via p38 MAPK in AGS human gastric cancer cells. J Cell Biochem 2012; 114:456-70. [DOI: 10.1002/jcb.24386] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2012] [Accepted: 08/30/2012] [Indexed: 01/13/2023]
|
48
|
Chu J, Pham NT, Olate N, Kislitsyna K, Day MC, LeTourneau PA, Kots A, Stewart RH, Laine GA, Cox CS, Uray K. Biphasic regulation of myosin light chain phosphorylation by p21-activated kinase modulates intestinal smooth muscle contractility. J Biol Chem 2012; 288:1200-13. [PMID: 23161543 DOI: 10.1074/jbc.m112.370718] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Supraphysiological mechanical stretching in smooth muscle results in decreased contractile activity. However, the mechanism is unclear. Previous studies indicated that intestinal motility dysfunction after edema development is associated with increased smooth muscle stress and decreased myosin light chain (MLC) phosphorylation in vivo, providing an ideal model for studying mechanical stress-mediated decrease in smooth muscle contraction. Primary human intestinal smooth muscle cells (hISMCs) were subjected to either control cyclical stretch (CCS) or edema (increasing) cyclical stretch (ECS), mimicking the biophysical forces in non-edematous and edematous intestinal smooth muscle in vivo. ECS induced significant decreases in phosphorylation of MLC and MLC phosphatase targeting subunit (MYPT1) and a significant increase in p21-activated kinase (PAK) activity compared with CCS. PAK regulated MLC phosphorylation in an activity-dependent biphasic manner. PAK activation increased MLC and MYPT1 phosphorylation in CCS but decreased MLC and MYPT1 phosphorylation in hISMCs subjected to ECS. PAK inhibition had the opposite results. siRNA studies showed that PAK1 plays a critical role in regulating MLC phosphorylation in hISMCs. PAK1 enhanced MLC phosphorylation via phosphorylating MYPT1 on Thr-696, whereas PAK1 inhibited MLC phosphorylation via decreasing MYPT1 on both Thr-696 and Thr-853. Importantly, in vivo data indicated that PAK activity increased in edematous tissue, and inhibition of PAK in edematous intestine improved intestinal motility. We conclude that PAK1 positively regulates MLC phosphorylation in intestinal smooth muscle through increasing inhibitory phosphorylation of MYPT1 under physiologic conditions, whereas PAK1 negatively regulates MLC phosphorylation via inhibiting MYPT1 phosphorylation when PAK activity is increased under pathologic conditions.
Collapse
Affiliation(s)
- Ji Chu
- Department of Pediatric Surgery, University of Texas Medical School, Houston, Texas 77030, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Zhang Z, Zhang M, Li Y, Liu S, Ping S, Wang J, Ning F, Xie F, Li C. Simvastatin inhibits the additive activation of ERK1/2 and proliferation of rat vascular smooth muscle cells induced by combined mechanical stress and oxLDL through LOX-1 pathway. Cell Signal 2012; 25:332-40. [PMID: 23072789 DOI: 10.1016/j.cellsig.2012.10.006] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2012] [Accepted: 10/10/2012] [Indexed: 01/11/2023]
Abstract
Vein grafts interposed into arteries are susceptible to the development of atherosclerosis due to rapid increases in blood pressure. This process is accelerated in patients with hyperlipidemia. The molecular mechanism underlying this process is unknown. In this study, quiescent rat vascular smooth muscle cells (VSMCs) were treated in vitro with mechanical stretch stress (10% elongation) with and without oxLDL (25 μg/ml) in the presence and absence of simvastatin (2.5 μmol/L). The results demonstrate that stretch stress and oxLDL can each induce activation of ERK1/2 and Ki-67 expression in VSMCs, but the peak levels of ERK activation and Ki-67 expression were observed in groups subjected to both stretch stress and oxLDL. Simvastatin was found to inhibit increased ERK activation and Ki-67 expression in VSMCs subjected to stretch stress with or without oxLDL. Mechanically, simvastatin was also found to inhibit increased expression of LOX-1 (a receptor of oxLDL) in VSMCs subjected to stretch stress with or without oxLDL. Knockdown of LOX-1 via small interfering RNAs (siRNA-LOX-1) resulted in obvious inhibition of ERK activation in VSMCs subjected to stretch stress with and without oxLDL. These results suggest that combined stretch stress and oxLDL can additively promote the activation of ERK1/2 leading to accelerated proliferation of VSMCs (e.g. increased Ki-67 expression) via LOX-1 signal pathway. This was found to be partially inhibited by simvastatin. These results may provide important data for the treatment and prevention of hypertension with or without hyperlipidemia.
Collapse
Affiliation(s)
- Zhengyu Zhang
- Department of Histology and Embryology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Effect of HSP65 on the expression of adhesion molecules in mice heart endothelial cells. Inflammation 2012; 35:1049-57. [PMID: 22160869 DOI: 10.1007/s10753-011-9410-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/14/2022]
Abstract
This study aims to research the effect of HSP65 on the expression of adhesion molecules in activated mice heart endothelial cells (MHECs), which were from myocardial tissue of newborn animals. We used different concentrations of LPS as potent inducers to stimulate MHECs, adhesion molecule expression in vitro, including intercellular adhesion molecule-1 (ICAM-1), vascular adhesion molecule-1 (VCAM-1), E-, and P-selectins, then compared the mRNA and protein levels of adhesion molecules expression with or without HSP65 treatment at different levels. The optimal concentration of LPS to induce MHECs adhesion molecule expression is 100 ng/ml; HSP65 treatment significantly reduced the mRNA and protein levels of MHECs' ICAM-1, VCAM-1, E-, and P-selectins expression (p < 0.05), and the optimal concentration of HSP65 in inhibiting MHECs activation is 0.8 ng. HSP65 has the inhibitory effect on adhesion molecules expression in activated MHECs.
Collapse
|