1
|
Yun K, Seo Y, Oh Y, Kim S, Maeng HS, Cho JG, Son YB, Son DJ, Kwon K, Kim S, Jo H, Park H. Lysyl-tRNA synthetase 1 promotes atherogenesis via autophagy-related secretion and inflammation. Sci Rep 2025; 15:11099. [PMID: 40169806 PMCID: PMC11961584 DOI: 10.1038/s41598-025-96046-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2024] [Accepted: 03/25/2025] [Indexed: 04/03/2025] Open
Abstract
Lysyl-tRNA synthetase 1 (KARS1), an aminoacyl-tRNA synthetase, was recently identified as a secreted pro-inflammatory agent. However, the vascular secretion and functions of KARS1 have not been characterized. This study investigated the secretion mechanisms of KARS1 and explored its functional roles in vascular biology. We found that KARS1 expression was upregulated by oscillatory shear stress, an atherogenic factor, suggesting the presence of free KARS1 dissociated from aminoacyl-tRNA synthetase complexes. Moreover, in the presence of Ca2+, serum starvation triggered free cytosolic KARS1 release from endothelial cells via secretory autophagy. Both phosphatidylinositol 3-phosphate kinase and caveolin-1 were either supplementary or essential for KARS1 secretion. Secreted KARS1 co-localized in the exosome fraction of post-culture media and was externally exposed. Further, secreted KARS1 inhibited shear-induced activation of various signaling molecules, including extracellular signal-regulated kinase, protein kinase B, and endothelial nitric oxide synthetase. Secreted KARS1 in atherosclerotic plaques also acted as an atherogenic or proinflammatory autocrine/paracrine molecule. Additionally, KARS1 participated in vessel alteration. Collectively, these findings describe novel vascular features of KARS1 in response to shear stress, providing insights into shear stress-controlling mechanisms of the vascular system.
Collapse
Affiliation(s)
- Kangmin Yun
- Department of Molecular Biology, Institute of Nanosensor and Biotechnology, Dankook University, 119 Dandae-Ro, Dongnam-Gu, Cheonan-Si, Chungnam, 31116, South Korea
| | - Youngsik Seo
- Department of Molecular Biology, Institute of Nanosensor and Biotechnology, Dankook University, 119 Dandae-Ro, Dongnam-Gu, Cheonan-Si, Chungnam, 31116, South Korea
| | - Yerim Oh
- Department of Molecular Biology, Institute of Nanosensor and Biotechnology, Dankook University, 119 Dandae-Ro, Dongnam-Gu, Cheonan-Si, Chungnam, 31116, South Korea
| | - Sunghyen Kim
- Department of Molecular Biology, Institute of Nanosensor and Biotechnology, Dankook University, 119 Dandae-Ro, Dongnam-Gu, Cheonan-Si, Chungnam, 31116, South Korea
| | - Hye Sun Maeng
- Department of Molecular Biology, Institute of Nanosensor and Biotechnology, Dankook University, 119 Dandae-Ro, Dongnam-Gu, Cheonan-Si, Chungnam, 31116, South Korea
| | - Jin Gu Cho
- Department of Molecular Biology, Institute of Nanosensor and Biotechnology, Dankook University, 119 Dandae-Ro, Dongnam-Gu, Cheonan-Si, Chungnam, 31116, South Korea
| | - Young Bae Son
- College of Pharmacy, Chungbuk National University, 194-21 Osongsaengmyong 1-Ro, Osong-Eup, Heungduk-Gu, Cheongju, 28160, Chungbuk, Republic of Korea
| | - Dong Ju Son
- College of Pharmacy, Chungbuk National University, 194-21 Osongsaengmyong 1-Ro, Osong-Eup, Heungduk-Gu, Cheongju, 28160, Chungbuk, Republic of Korea
| | - Kihwan Kwon
- Division of Cardiology, Department of Internal Medicine, Ewha Womans University College of Medicine, Ewha Womans University Mokdong Hospital, Seoul, Republic of Korea
| | - Sunghoon Kim
- Department of Integrative Biotechnology, Interdisciplinary Graduate Program, Medicinal Bioconvergence Research Center, Institute for Artificial Intelligence and Biomedical Research, College of Pharmacy, Yonsei University, 85 Songdogwahak-Ro, Yeonsu-Gu, Incheon, 21983, Korea
| | - Hanjoong Jo
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA
| | - Heonyong Park
- Department of Molecular Biology, Institute of Nanosensor and Biotechnology, Dankook University, 119 Dandae-Ro, Dongnam-Gu, Cheonan-Si, Chungnam, 31116, South Korea.
| |
Collapse
|
2
|
Mendez J, Toker A. Minimizing Shear Stress in Cell Signaling Studies. Curr Protoc 2024; 4:e1019. [PMID: 38506433 DOI: 10.1002/cpz1.1019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/21/2024]
Abstract
Cellular signal transduction comprises a complex series of biochemical reactions by which extracellular signals such as growth factors, hormones, cytokines, and neurotransmitters are translated into specific intracellular responses. Signal transduction is mediated by protein kinase phosphorylation cascades that culminate in the regulation of numerous cellular responses, including division, differentiation, migration, and survival. Importantly, signal relay pathways are dysregulated in human diseases, making the study of signal transduction important for both uncovering basic biology and understanding pathophysiology. Established laboratory cell culture models are useful for studying signal transduction mechanisms, but differences in sample handling procedures can introduce unwanted variability in experimental outcomes and conclusions. One such potential source of experimental variability is the introduction of fluid shear stress upon handling of tissue culture cells. Fluid shear stress triggers a wide range of cellular responses in adherent cell culture, including stimulating the production of cyclic AMP, potentiating the activation of extracellular signal-regulated kinases 1 and 2 (ERK1/2), and ultimately inducing changes in the gene expression of growth and remodeling factors. Further, mechanical stress on cells is physiologically relevant to the development of many pathologies. Here, we describe a detailed protocol for cell lysis and protein extraction that minimizes shear stress induced by classical cell harvest protocols. We also highlight the impact of fluid shear stress by using immunoblotting to assess ERK pathway activation as a readout for this protocol. © 2024 Wiley Periodicals LLC. Basic Protocol 1: Gentle cell lysis and protein extraction Basic Protocol 2: Immunoblotting for cell signaling readouts by SDS-PAGE.
Collapse
Affiliation(s)
- Josefina Mendez
- Department of Pathology and Cancer Center, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts
| | - Alex Toker
- Department of Pathology and Cancer Center, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
3
|
Crozet F, Levayer R. Emerging roles and mechanisms of ERK pathway mechanosensing. Cell Mol Life Sci 2023; 80:355. [PMID: 37947896 PMCID: PMC10638131 DOI: 10.1007/s00018-023-05007-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Revised: 10/11/2023] [Accepted: 10/16/2023] [Indexed: 11/12/2023]
Abstract
The coupling between mechanical forces and modulation of cell signalling pathways is essential for tissue plasticity and their adaptation to changing environments. Whilst the number of physiological and pathological relevant roles of mechanotransduction has been rapidly expanding over the last decade, studies have been mostly focussing on a limited number of mechanosensitive pathways, which include for instance Hippo/YAP/TAZ pathway, Wnt/β-catenin or the stretch-activated channel Piezo. However, the recent development and spreading of new live sensors has provided new insights into the contribution of ERK pathway in mechanosensing in various systems, which emerges now as a fast and modular mechanosensitive pathway. In this review, we will document key in vivo and in vitro examples that have established a clear link between cell deformation, mechanical stress and modulation of ERK signalling, comparing the relevant timescale and mechanical stress. We will then discuss different molecular mechanisms that have been proposed so far, focussing on the epistatic link between mechanics and ERK and discussing the relevant cellular parameters affecting ERK signalling. We will finish by discussing the physiological and the pathological consequences of the link between ERK and mechanics, outlining how this interplay is instrumental for self-organisation and long-range cell-cell coordination.
Collapse
Affiliation(s)
- Flora Crozet
- Department of Developmental and Stem Cell Biology, Institut Pasteur, Université de Paris Cité, CNRS UMR 3738, 25 Rue du Dr. Roux, 75015, Paris, France
| | - Romain Levayer
- Department of Developmental and Stem Cell Biology, Institut Pasteur, Université de Paris Cité, CNRS UMR 3738, 25 Rue du Dr. Roux, 75015, Paris, France.
| |
Collapse
|
4
|
Tamargo IA, Baek KI, Kim Y, Park C, Jo H. Flow-induced reprogramming of endothelial cells in atherosclerosis. Nat Rev Cardiol 2023; 20:738-753. [PMID: 37225873 PMCID: PMC10206587 DOI: 10.1038/s41569-023-00883-1] [Citation(s) in RCA: 77] [Impact Index Per Article: 38.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 04/25/2023] [Indexed: 05/26/2023]
Abstract
Atherosclerotic diseases such as myocardial infarction, ischaemic stroke and peripheral artery disease continue to be leading causes of death worldwide despite the success of treatments with cholesterol-lowering drugs and drug-eluting stents, raising the need to identify additional therapeutic targets. Interestingly, atherosclerosis preferentially develops in curved and branching arterial regions, where endothelial cells are exposed to disturbed blood flow with characteristic low-magnitude oscillatory shear stress. By contrast, straight arterial regions exposed to stable flow, which is associated with high-magnitude, unidirectional shear stress, are relatively well protected from the disease through shear-dependent, atheroprotective endothelial cell responses. Flow potently regulates structural, functional, transcriptomic, epigenomic and metabolic changes in endothelial cells through mechanosensors and mechanosignal transduction pathways. A study using single-cell RNA sequencing and chromatin accessibility analysis in a mouse model of flow-induced atherosclerosis demonstrated that disturbed flow reprogrammes arterial endothelial cells in situ from healthy phenotypes to diseased ones characterized by endothelial inflammation, endothelial-to-mesenchymal transition, endothelial-to-immune cell-like transition and metabolic changes. In this Review, we discuss this emerging concept of disturbed-flow-induced reprogramming of endothelial cells (FIRE) as a potential pro-atherogenic mechanism. Defining the flow-induced mechanisms through which endothelial cells are reprogrammed to promote atherosclerosis is a crucial area of research that could lead to the identification of novel therapeutic targets to combat the high prevalence of atherosclerotic disease.
Collapse
Affiliation(s)
- Ian A Tamargo
- Wallace H. Coulter Department of Biomedical Engineering, Emory University and Georgia Institute of Technology, Atlanta, GA, USA
- Molecular and Systems Pharmacology Program, Emory University, Atlanta, GA, USA
| | - Kyung In Baek
- Wallace H. Coulter Department of Biomedical Engineering, Emory University and Georgia Institute of Technology, Atlanta, GA, USA
| | - Yerin Kim
- Wallace H. Coulter Department of Biomedical Engineering, Emory University and Georgia Institute of Technology, Atlanta, GA, USA
| | - Christian Park
- Wallace H. Coulter Department of Biomedical Engineering, Emory University and Georgia Institute of Technology, Atlanta, GA, USA
| | - Hanjoong Jo
- Wallace H. Coulter Department of Biomedical Engineering, Emory University and Georgia Institute of Technology, Atlanta, GA, USA.
- Molecular and Systems Pharmacology Program, Emory University, Atlanta, GA, USA.
- Department of Medicine, Emory University School, Atlanta, GA, USA.
| |
Collapse
|
5
|
Davis MJ, Earley S, Li YS, Chien S. Vascular mechanotransduction. Physiol Rev 2023; 103:1247-1421. [PMID: 36603156 PMCID: PMC9942936 DOI: 10.1152/physrev.00053.2021] [Citation(s) in RCA: 90] [Impact Index Per Article: 45.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Revised: 09/26/2022] [Accepted: 10/04/2022] [Indexed: 01/07/2023] Open
Abstract
This review aims to survey the current state of mechanotransduction in vascular smooth muscle cells (VSMCs) and endothelial cells (ECs), including their sensing of mechanical stimuli and transduction of mechanical signals that result in the acute functional modulation and longer-term transcriptomic and epigenetic regulation of blood vessels. The mechanosensors discussed include ion channels, plasma membrane-associated structures and receptors, and junction proteins. The mechanosignaling pathways presented include the cytoskeleton, integrins, extracellular matrix, and intracellular signaling molecules. These are followed by discussions on mechanical regulation of transcriptome and epigenetics, relevance of mechanotransduction to health and disease, and interactions between VSMCs and ECs. Throughout this review, we offer suggestions for specific topics that require further understanding. In the closing section on conclusions and perspectives, we summarize what is known and point out the need to treat the vasculature as a system, including not only VSMCs and ECs but also the extracellular matrix and other types of cells such as resident macrophages and pericytes, so that we can fully understand the physiology and pathophysiology of the blood vessel as a whole, thus enhancing the comprehension, diagnosis, treatment, and prevention of vascular diseases.
Collapse
Affiliation(s)
- Michael J Davis
- Department of Medical Pharmacology and Physiology, University of Missouri, Columbia, Missouri
| | - Scott Earley
- Department of Pharmacology, University of Nevada, Reno, Nevada
| | - Yi-Shuan Li
- Department of Bioengineering, University of California, San Diego, California
- Institute of Engineering in Medicine, University of California, San Diego, California
| | - Shu Chien
- Department of Bioengineering, University of California, San Diego, California
- Institute of Engineering in Medicine, University of California, San Diego, California
- Department of Medicine, University of California, San Diego, California
| |
Collapse
|
6
|
Tauchi M, Oshita K, Urschel K, Furtmair R, Kühn C, Stumpfe FM, Botos B, Achenbach S, Dietel B. The Involvement of Cx43 in JNK1/2-Mediated Endothelial Mechanotransduction and Human Plaque Progression. Int J Mol Sci 2023; 24:ijms24021174. [PMID: 36674690 PMCID: PMC9863493 DOI: 10.3390/ijms24021174] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Revised: 12/23/2022] [Accepted: 12/31/2022] [Indexed: 01/11/2023] Open
Abstract
Atherosclerotic lesions preferentially develop at bifurcations, characterized by non-uniform shear stress (SS). The aim of this study was to investigate SS-induced endothelial activation, focusing on stress-regulated mitogen-activated protein kinases (MAPK) and downstream signaling, and its relation to gap junction proteins, Connexins (Cxs). Human umbilical vein endothelial cells were exposed to flow ("mechanical stimulation") and stimulated with TNF-α ("inflammatory stimulation"). Phosphorylated levels of MAPKs (c-Jun N-terminal kinase (JNK1/2), extracellular signal-regulated kinase (ERK), and p38 kinase (p38K)) were quantified by flow cytometry, showing the activation of JNK1/2 and ERK. THP-1 cell adhesion under non-uniform SS was suppressed by the inhibition of JNK1/2, not of ERK. Immunofluorescence staining and quantitative real-time PCR demonstrated an induction of c-Jun and c-Fos and of Cx43 in endothelial cells by non-uniform SS, and the latter was abolished by JNK1/2 inhibition. Furthermore, plaque inflammation was analyzed in human carotid plaques (n = 40) using immunohistochemistry and quanti-gene RNA-assays, revealing elevated Cx43+ cell counts in vulnerable compared to stable plaques. Cx43+ cell burden in the plaque shoulder correlated with intraplaque neovascularization and lipid core size, while an inverse correlation was observed with fibrous cap thickness. Our results constitute the first report that JNK1/2 mediates Cx43 mechanoinduction in endothelial cells by atheroprone shear stress and that Cx43 is expressed in human carotid plaques. The correlation of Cx43+ cell counts with markers of plaque vulnerability implies its contribution to plaque progression.
Collapse
Affiliation(s)
- Miyuki Tauchi
- Department of Cardiology and Angiology, Erlangen University Hospital, Friedrich-Alexander University Erlangen-Nürnberg (FAU), 91054 Erlangen, Germany
- Cognitive and Molecular Research Institute of Brain Diseases, Kurume University, Kurume 830-0011, Japan
| | - Kensuke Oshita
- Department of Cardiology and Angiology, Erlangen University Hospital, Friedrich-Alexander University Erlangen-Nürnberg (FAU), 91054 Erlangen, Germany
- Department of Anesthesiology, School of Medicine, Kurume University, Kurume 830-0011, Japan
| | - Katharina Urschel
- Department of Cardiology and Angiology, Erlangen University Hospital, Friedrich-Alexander University Erlangen-Nürnberg (FAU), 91054 Erlangen, Germany
| | - Roman Furtmair
- Department of Cardiology and Angiology, Erlangen University Hospital, Friedrich-Alexander University Erlangen-Nürnberg (FAU), 91054 Erlangen, Germany
| | - Constanze Kühn
- Department of Cardiology and Angiology, Erlangen University Hospital, Friedrich-Alexander University Erlangen-Nürnberg (FAU), 91054 Erlangen, Germany
| | - Florian M. Stumpfe
- Department of Cardiology and Angiology, Erlangen University Hospital, Friedrich-Alexander University Erlangen-Nürnberg (FAU), 91054 Erlangen, Germany
| | - Balazs Botos
- Department of Vascular Surgery, Hospital of Nürnberg-Süd, 90471 Nürnberg, Germany
| | - Stephan Achenbach
- Department of Cardiology and Angiology, Erlangen University Hospital, Friedrich-Alexander University Erlangen-Nürnberg (FAU), 91054 Erlangen, Germany
| | - Barbara Dietel
- Department of Cardiology and Angiology, Erlangen University Hospital, Friedrich-Alexander University Erlangen-Nürnberg (FAU), 91054 Erlangen, Germany
- Correspondence:
| |
Collapse
|
7
|
Du R, Li D, Zhu M, Zheng L, Ren K, Han D, Li L, Ji J, Fan Y. Cell senescence alters responses of porcine trabecular meshwork cells to shear stress. Front Cell Dev Biol 2022; 10:1083130. [PMID: 36478743 PMCID: PMC9721263 DOI: 10.3389/fcell.2022.1083130] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Accepted: 11/04/2022] [Indexed: 10/05/2024] Open
Abstract
Mechanical microenvironment and cellular senescence of trabecular meshwork cells (TMCs) are suspected to play a vital role in primary open-angle glaucoma pathogenesis. However, central questions remain about the effect of shear stress on TMCs and how aging affects this process. We have investigated the effect of shear stress on the biomechanical properties and extracellular matrix regulation of normal and senescent TMCs. We found a more significant promotion of Fctin formation, a more obvious realignment of F-actin fibers, and a more remarkable increase in the stiffness of normal cells in response to the shear stress, in comparison with that of senescent cells. Further, as compared to normal cells, senescent cells show a reduced extracellular matrix turnover after shear stress stimulation, which might be attributed to the different phosphorylation levels of the extracellular signal-regulated kinase. Our results suggest that TMCs are able to sense and respond to the shear stress and cellular senescence undermines the mechanobiological response, which may lead to progressive failure of cellular TM function with age.
Collapse
Affiliation(s)
- Ruotian Du
- Key Laboratory of Biomechanics and Mechanobiology of Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing, China
| | - Dongyan Li
- Key Laboratory of Biomechanics and Mechanobiology of Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing, China
| | - Meng Zhu
- Key Laboratory of Biomechanics and Mechanobiology of Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing, China
| | - Lisha Zheng
- Key Laboratory of Biomechanics and Mechanobiology of Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing, China
| | - Keli Ren
- Lab for Biological Imaging and Nanomedicine, National Center for Nanoscience and Technology, Beijing, China
| | - Dong Han
- Lab for Biological Imaging and Nanomedicine, National Center for Nanoscience and Technology, Beijing, China
| | - Long Li
- State Key Laboratory of Nonlinear Mechanics and Beijing Key Laboratory of Engineered Construction and Mechanobiology, Institute of Mechanics, Chinese Academy of Sciences, Beijing, China
| | - Jing Ji
- Key Laboratory of Biomechanics and Mechanobiology of Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing, China
| | - Yubo Fan
- Key Laboratory of Biomechanics and Mechanobiology of Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing, China
| |
Collapse
|
8
|
Fang JS, Burt JM. Connexin37 Regulates Cell Cycle in the Vasculature. J Vasc Res 2022; 60:73-86. [PMID: 36067749 DOI: 10.1159/000525619] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Accepted: 06/08/2022] [Indexed: 11/19/2022] Open
Abstract
Control of vascular cell growth responses is critical for development and maintenance of a healthy vasculature. Connexins - the proteins comprising gap junction channels - are key regulators of cell growth in diseases such as cancer, but their involvement in controlling cell growth in the vasculature is less well appreciated. Connexin37 (Cx37) is one of four connexin isotypes expressed in the vessel wall. Its primary role in blood vessels relies on its unique ability to transduce flow-sensitive signals into changes in cell cycle status of endothelial (and perhaps, mural) cells. Here, we review available evidence for Cx37's role in the regulation of vascular growth, vessel organization, and vascular tone in healthy and diseased vasculature. We propose a novel mechanism whereby Cx37 accomplishes this with a phosphorylation-dependent transition between closed (growth-suppressive) and multiple open (growth-permissive) channel conformations that result from interactions of the C-terminus with cell-cycle regulators to limit or support cell cycle progression. Lastly, we discuss Cx37 and its downstream signaling as a novel potential target in the treatment of cardiovascular disease, and we address outstanding research questions that still challenge the development of such therapies.
Collapse
Affiliation(s)
- Jennifer S Fang
- Department of Cell and Molecular Biology, Tulane University, New Orleans, Louisiana, USA
| | - Janis M Burt
- Department of Physiology, University of Arizona, Tucson, Arizona, USA
| |
Collapse
|
9
|
Suwittayarak R, Klincumhom N, Ngaokrajang U, Namangkalakul W, Ferreira JN, Pavasant P, Osathanon T. Shear Stress Enhances the Paracrine-Mediated Immunoregulatory Function of Human Periodontal Ligament Stem Cells via the ERK Signalling Pathway. Int J Mol Sci 2022; 23:ijms23137119. [PMID: 35806124 PMCID: PMC9266779 DOI: 10.3390/ijms23137119] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Revised: 06/16/2022] [Accepted: 06/23/2022] [Indexed: 11/25/2022] Open
Abstract
Relevant immunomodulatory effects have been proposed following allogeneic cell-based therapy with human periodontal ligament stem cells (hPDLSCs). This study aimed to examine the influence of shear stress on the immunosuppressive capacity of hPDLSCs. Cells were subjected to shear stress at different magnitudes (0.5, 5 and 10 dyn/cm2). The expression of immunosuppressive markers was evaluated in shear stress-induced hPDLSCs using qRT-PCR, western blot, enzyme activity and enzyme-linked immunosorbent assays. The effects of a shear stress-derived condition medium (SS-CM) on T cell proliferation were examined using a resazurin assay. Treg differentiation was investigated using qRT-PCR and flow cytometry analysis. Our results revealed that shear stress increased mRNA expression of IDO and COX2 but not TGF-β1 and IFN-γ. IDO activity, kynurenine and active TGF-β1 increased in SS-CM when compared to the non-shear stress-derived conditioned medium (CTL-CM). The amount of kynurenine in SS-CM was reduced in the presence of cycloheximide and ERK inhibitor. Subsequently, T cell proliferation decreased in SS-CM compared to CTL-CM. Treg differentiation was promoted in SS-CM, indicated by FOXP3, IL-10 expression and CD4+CD25hiCD127lo/− subpopulation. In conclusion, shear stress promotes kynurenine production through ERK signalling in hPDLSC, leading to the inhibition of T cell proliferation and the promotion of Treg cell differentiation.
Collapse
Affiliation(s)
- Ravipha Suwittayarak
- Center of Excellence for Regenerative Dentistry, Department of Anatomy, Faculty of Dentistry, Chulalongkorn University, Bangkok 10330, Thailand; (R.S.); (U.N.); (P.P.)
| | - Nuttha Klincumhom
- Center of Excellence for Regenerative Dentistry, Department of Anatomy, Faculty of Dentistry, Chulalongkorn University, Bangkok 10330, Thailand; (R.S.); (U.N.); (P.P.)
- Correspondence:
| | - Utapin Ngaokrajang
- Center of Excellence for Regenerative Dentistry, Department of Anatomy, Faculty of Dentistry, Chulalongkorn University, Bangkok 10330, Thailand; (R.S.); (U.N.); (P.P.)
| | - Worachat Namangkalakul
- Dental Stem Cell Biology Research Unit, Department of Anatomy, Faculty of Dentistry, Chulalongkorn University, Bangkok 10330, Thailand; (W.N.); (T.O.)
| | - João N. Ferreira
- Avatar Biotechnologies for Oral Health and Healthy Longevity Research Unit, Faculty of Dentistry, Chulalongkorn University, Bangkok 10330, Thailand;
| | - Prasit Pavasant
- Center of Excellence for Regenerative Dentistry, Department of Anatomy, Faculty of Dentistry, Chulalongkorn University, Bangkok 10330, Thailand; (R.S.); (U.N.); (P.P.)
| | - Thanaphum Osathanon
- Dental Stem Cell Biology Research Unit, Department of Anatomy, Faculty of Dentistry, Chulalongkorn University, Bangkok 10330, Thailand; (W.N.); (T.O.)
| |
Collapse
|
10
|
He G, Kan S, Xu S, Sun X, Li R, Shu W, Chen M. LXN deficiency regulates cytoskeleton remodelling by promoting proteolytic cleavage of Filamin A in vascular endothelial cells. J Cell Mol Med 2021; 25:6815-6827. [PMID: 34085389 PMCID: PMC8278077 DOI: 10.1111/jcmm.16685] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Revised: 04/25/2021] [Accepted: 05/12/2021] [Indexed: 12/19/2022] Open
Abstract
Endothelial cells (ECs) respond to blood shear stress by changing their morphology is important for maintaining vascular homeostasis. Studies have documented a relationship between endothelial cell shape and the stress flow, and however, the mechanism underlying this cytoskeletal rearrangement due to shear stress remains uncertain. In this paper, we demonstrate that laminar shear stress (LSS) significantly reduces latexin (LXN) expression in ECs. By using siRNA and cell imaging, we demonstrated that LXN knockdown results in the morphologic change and F‐actin remodelling just like what LSS does in ECs. We further demonstrate that LXN interacts with Filamin A (FLNA) and regulates FLNA proteolytic cleavage and nuclei translocation. By constructing LXN‐/‐ mice and ApoE‐/‐LXN‐/‐ double knockout mice, we evaluated the effect of LXN knockout on aortic endothelium damage in mice. We found that LXN deficiency significantly improves vascular permeability, vasodilation and atherosclerosis in mice. Our findings provide confident evidence, for the first time, that LXN is a novel regulator for morphological maintenance of ECs, and LXN deficiency has a protective effect on vascular homeostasis. This provides new strategies and drug targets for the treatment of vascular diseases.
Collapse
Affiliation(s)
- Guozhang He
- State Key Laboratory for the Chemistry and Molecular Engineering of Medicinal Resources, School of Chemistry and Pharmacy, Guangxi Normal University, Guilin, China.,Collaborative Innovation Center for Guangxi Ethnic Medicine, Guangxi Normal University, Guilin, China
| | - Shuang Kan
- State Key Laboratory for the Chemistry and Molecular Engineering of Medicinal Resources, School of Chemistry and Pharmacy, Guangxi Normal University, Guilin, China.,Collaborative Innovation Center for Guangxi Ethnic Medicine, Guangxi Normal University, Guilin, China
| | - Shaohua Xu
- State Key Laboratory for the Chemistry and Molecular Engineering of Medicinal Resources, School of Chemistry and Pharmacy, Guangxi Normal University, Guilin, China.,Collaborative Innovation Center for Guangxi Ethnic Medicine, Guangxi Normal University, Guilin, China
| | - Xuchen Sun
- State Key Laboratory for the Chemistry and Molecular Engineering of Medicinal Resources, School of Chemistry and Pharmacy, Guangxi Normal University, Guilin, China.,Collaborative Innovation Center for Guangxi Ethnic Medicine, Guangxi Normal University, Guilin, China
| | - Rong Li
- State Key Laboratory for the Chemistry and Molecular Engineering of Medicinal Resources, School of Chemistry and Pharmacy, Guangxi Normal University, Guilin, China.,Collaborative Innovation Center for Guangxi Ethnic Medicine, Guangxi Normal University, Guilin, China
| | - Wei Shu
- College of Biotechnology, Guilin Medical University, Guilin, China
| | - Ming Chen
- State Key Laboratory for the Chemistry and Molecular Engineering of Medicinal Resources, School of Chemistry and Pharmacy, Guangxi Normal University, Guilin, China.,Collaborative Innovation Center for Guangxi Ethnic Medicine, Guangxi Normal University, Guilin, China
| |
Collapse
|
11
|
Van Guilder GP, Preston CC, Munce TA, Faustino RS. Impacts of circulating microRNAs in exercise-induced vascular remodeling. Am J Physiol Heart Circ Physiol 2021; 320:H2401-H2415. [PMID: 33989080 DOI: 10.1152/ajpheart.00894.2020] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Cardiovascular adaptation underlies all athletic training modalities, with a variety of factors contributing to overall response during exercise-induced stimulation. In this regard the role of circulating biomarkers is a well-established and invaluable tool for monitoring cardiovascular function. Specifically, novel biomarkers such as circulating cell free DNA and RNA are now becoming attractive tools for monitoring cardiovascular function with the advent of next generation technologies that can provide unprecedented precision and resolution of these molecular signatures, paving the way for novel diagnostic and prognostic avenues to better understand physiological remodeling that occurs in trained versus untrained states. In particular, microRNAs are a species of regulatory RNAs with pleiotropic effects on multiple pathways in tissue-specific manners. Furthermore, the identification of cell free microRNAs within peripheral circulation represents a distal signaling mechanism that is just beginning to be explored via a diversity of molecular and bioinformatic approaches. This article provides an overview of the emerging field of sports/performance genomics with a focus on the role of microRNAs as novel functional diagnostic and prognostic tools, and discusses present knowledge in the context of athletic vascular remodeling. This review concludes with current advantages and limitations, touching upon future directions and implications for applying contemporary systems biology knowledge of exercise-induced physiology to better understand how disruption can lead to pathology.
Collapse
Affiliation(s)
- Gary P Van Guilder
- Vascular Protection Research Laboratory, Exercise & Sport Science Department, Western Colorado University, Gunnison, Colorado
| | - Claudia C Preston
- Genetics and Genomics Group, Sanford Research, Sioux Falls, South Dakota
| | - Thayne A Munce
- Environmental Influences on Health & Disease Group, Sanford Research, Sioux Falls, South Dakota.,Department of Pediatrics, Sanford School of Medicine, University of South Dakota, Sioux Falls, South Dakota
| | - Randolph S Faustino
- Genetics and Genomics Group, Sanford Research, Sioux Falls, South Dakota.,Department of Pediatrics, Sanford School of Medicine, University of South Dakota, Sioux Falls, South Dakota
| |
Collapse
|
12
|
Hu Y, Chen M, Wang M, Li X. Flow-mediated vasodilation through mechanosensitive G protein-coupled receptors in endothelial cells. Trends Cardiovasc Med 2021; 32:61-70. [PMID: 33406458 DOI: 10.1016/j.tcm.2020.12.010] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Revised: 12/15/2020] [Accepted: 12/29/2020] [Indexed: 12/11/2022]
Abstract
Currently, endothelium-dependent vasodilatation involves three main mechanisms: production of nitric oxide (NO) by endothelial nitric oxide synthase (eNOS), synthesis of prostanoids by cyclooxygenase, and/or opening of calcium-sensitive potassium channels. Researchers have proposed multiple mechanosensors that may be involved in flow-mediated vasodilation (FMD), including G protein-coupled receptors (GPCRs), ion channels, and intercellular junction proteins, among others. However, GPCRs are considered the major mechanosensors that play a pivotal role in shear stress signal transduction. Among mechanosensitive GPCRs, G protein-coupled receptor 68, histamine H1 receptors, sphingosine-1-phosphate receptor 1, and bradykinin B2 receptors have been identified as endothelial sensors of flow shear stress regulating flow-mediated vasodilation. Thus, this review aims to expound on the mechanism whereby flow shear stress promotes vasodilation through the proposed mechanosensitive GPCRs in ECs.
Collapse
Affiliation(s)
- Yong Hu
- Department of Hand and Foot Surgery, The Second Hospital, Cheeloo College of Medicine, Shandong University, No.247, Beiyuan Street, Jinan, Shandong Province, 250031, China.
| | - Miao Chen
- Department of Hand and Foot Surgery, The First Hospital of Jilin University, No.71, Xinmin Street, Changchun, Jilin Province, 130021, China.
| | - Meili Wang
- Department of Obstetrics, Maternal and Child Health Care Hospital of Shandong Province, Shandong University, NO.238, Jingshi East Road, Jinan, Shandong, 250012, China.
| | - Xiucun Li
- Department of Hand and Foot Surgery, The Second Hospital, Cheeloo College of Medicine, Shandong University, No.247, Beiyuan Street, Jinan, Shandong Province, 250031, China; Department of Anatomy and Histoembryology, School of Basic Medical Science, Cheeloo College of Medicine, Shandong University, NO.44, Wenhua West Road, Jinan, Shandong, 250012, China.
| |
Collapse
|
13
|
Brugger MS, Baumgartner K, Mauritz SCF, Gerlach SC, Röder F, Schlosser C, Fluhrer R, Wixforth A, Westerhausen C. Vibration enhanced cell growth induced by surface acoustic waves as in vitro wound-healing model. Proc Natl Acad Sci U S A 2020; 117:31603-31613. [PMID: 33257581 PMCID: PMC7749343 DOI: 10.1073/pnas.2005203117] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
We report on in vitro wound-healing and cell-growth studies under the influence of radio-frequency (rf) cell stimuli. These stimuli are supplied either by piezoactive surface acoustic waves (SAWs) or by microelectrode-generated electric fields, both at frequencies around 100 MHz. Employing live-cell imaging, we studied the time- and power-dependent healing of artificial wounds on a piezoelectric chip for different cell lines. If the cell stimulation is mediated by piezomechanical SAWs, we observe a pronounced, significant maximum of the cell-growth rate at a specific SAW amplitude, resulting in an increase of the wound-healing speed of up to 135 ± 85% as compared to an internal reference. In contrast, cells being stimulated only by electrical fields of the same magnitude as the ones exposed to SAWs exhibit no significant effect. In this study, we investigate this effect for different wavelengths, amplitude modulation of the applied electrical rf signal, and different wave modes. Furthermore, to obtain insight into the biological response to the stimulus, we also determined both the cell-proliferation rate and the cellular stress levels. While the proliferation rate is significantly increased for a wide power range, cell stress remains low and within the normal range. Our findings demonstrate that SAW-based vibrational cell stimulation bears the potential for an alternative method to conventional ultrasound treatment, overcoming some of its limitations.
Collapse
Affiliation(s)
- Manuel S Brugger
- Experimental Physics I, Institute of Physics, University of Augsburg, 86159 Augsburg, Germany
- Stiftung der Deutschen Wirtschaft gGmbH, 10178 Berlin, Germany
| | - Kathrin Baumgartner
- Experimental Physics I, Institute of Physics, University of Augsburg, 86159 Augsburg, Germany
- Studienstiftung des deutschen Volkes e.V., 53175 Bonn, Germany
| | - Sophie C F Mauritz
- Experimental Physics I, Institute of Physics, University of Augsburg, 86159 Augsburg, Germany
| | - Stefan C Gerlach
- Experimental Physics I, Institute of Physics, University of Augsburg, 86159 Augsburg, Germany
- Hans-Seidel-Stiftung e.V., 80636 Munich, Germany
| | - Florian Röder
- Experimental Physics I, Institute of Physics, University of Augsburg, 86159 Augsburg, Germany
| | - Christine Schlosser
- Biochemistry and Molecular Biology, Institute of Theoretical Medicine, University of Augsburg, 86159 Augsburg, Germany
| | - Regina Fluhrer
- Biochemistry and Molecular Biology, Institute of Theoretical Medicine, University of Augsburg, 86159 Augsburg, Germany
- German Center for Neurodegenerative Diseases (DZNE) e.V., 81377 Munich, Germany
| | - Achim Wixforth
- Experimental Physics I, Institute of Physics, University of Augsburg, 86159 Augsburg, Germany
- Center for NanoScience, 80799 Munich, Germany
| | - Christoph Westerhausen
- Experimental Physics I, Institute of Physics, University of Augsburg, 86159 Augsburg, Germany;
- Center for NanoScience, 80799 Munich, Germany
- Physiology, Institute of Theoretical Medicine, University of Augsburg, 86159 Augsburg, Germany
| |
Collapse
|
14
|
Lutz A, Jung D, Diem K, Fauler M, Port F, Gottschalk K, Felder E. Acute effects of cell stretch on keratin filaments in A549 lung cells. FASEB J 2020; 34:11227-11242. [PMID: 32632966 DOI: 10.1096/fj.201903160rr] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Revised: 05/29/2020] [Accepted: 06/18/2020] [Indexed: 12/31/2022]
Abstract
Keratin filaments (KFs) comprise the intermediate filaments of epithelial cells and are well known for their cytoprotective properties and their mechanical resilience. Although, several studies have demonstrated KFs' remarkable tensile properties relatively little is known about acute implications of mechanical stretch on KFs in living cells. This includes structural effects on the KFs and their higher level assembly structures as well as posttranslational response mechanisms to possibly modify KF's properties. We subjected simple epithelial A549 lung cells to 30% unidirectional stretch and already after 10 seconds we observed morphological changes of the KF-network as well as structural effects on their desmosomal anchor sites-both apparently caused by the tensile strain. Interestingly, the effect on the desmosomes was attenuated after 30 seconds of cell stretch with a concomitant increase in phosphorylation of keratin8-S432, keratin18-S53, and keratin18-S34 without an apparent increase in keratin solubility. When mimicking the phosphorylation of keratin18-S34 the stretch-induced effect on the desmosomes could be diminished and probing the cell surface with atomic force microscopy showed a lowered elastic modulus. We conclude that the stretch-induced KF phosphorylation affects KF's tensile properties, probably to lower the mechanical load on strained desmosomal cell-cell contacts, and hence, preserve epithelial integrity.
Collapse
Affiliation(s)
- Anngrit Lutz
- Department of General Physiology, Ulm University, Ulm, Germany
| | - Dominik Jung
- Department of General Physiology, Ulm University, Ulm, Germany
| | - Kathrin Diem
- Department of General Physiology, Ulm University, Ulm, Germany
| | - Michael Fauler
- Department of General Physiology, Ulm University, Ulm, Germany
| | - Fabian Port
- Department of Experimental Physics, Ulm University, Ulm, Germany
| | - Kay Gottschalk
- Department of Experimental Physics, Ulm University, Ulm, Germany
| | - Edward Felder
- Department of General Physiology, Ulm University, Ulm, Germany
| |
Collapse
|
15
|
Epidermal Growth Factor Receptor and Abl2 Kinase Regulate Distinct Steps of Human Papillomavirus 16 Endocytosis. J Virol 2020; 94:JVI.02143-19. [PMID: 32188731 DOI: 10.1128/jvi.02143-19] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2020] [Accepted: 03/11/2020] [Indexed: 02/07/2023] Open
Abstract
Human papillomavirus 16 (HPV16), the leading cause of cervical cancer, exploits a novel endocytic pathway during host cell entry. This mechanism shares many requirements with macropinocytosis but differs in the mode of vesicle formation. Previous work indicated a role of the epidermal growth factor receptor (EGFR) in HPV16 endocytosis. However, the functional outcome of EGFR signaling and its downstream targets during HPV16 uptake are not well characterized. Here, we analyzed the functional importance of signal transduction via EGFR and its downstream effectors for endocytosis of HPV16. Our findings indicate two phases of EGFR signaling as follows: a-likely dispensable-transient activation with or shortly after cell binding and signaling required throughout the process of asynchronous internalization of HPV16. Interestingly, EGFR inhibition interfered with virus internalization and strongly reduced the number of endocytic pits, suggesting a role for EGFR signaling in the induction of HPV16 endocytosis. Moreover, we identified the Src-related kinase Abl2 as a novel regulator of virus uptake. Inhibition of Abl2 resulted in an accumulation of misshaped endocytic pits, indicating Abl2's importance for endocytic vesicle maturation. Since Abl2 rather than Src, a regulator of membrane ruffling during macropinocytosis, mediated downstream signaling of EGFR, we propose that the selective effector targeting downstream of EGFR determines whether HPV16 endocytosis or macropinocytosis is induced.IMPORTANCE Human papillomaviruses are small, nonenveloped DNA viruses that infect skin and mucosa. The so-called high-risk HPVs (e.g., HPV16, HPV18, HPV31) have transforming potential and are associated with various anogenital and oropharyngeal tumors. These viruses enter host cells by a novel endocytic pathway with unknown cellular function. To date, it is unclear how endocytic vesicle formation occurs mechanistically. Here, we addressed the role of epidermal growth factor receptor signaling, which has previously been implicated in HPV16 endocytosis and identified the kinase Abl2 as a novel regulator of virus uptake. Since other viruses, such as influenza A virus and lymphocytic choriomeningitis virus, possibly make use of related mechanisms, our findings shed light on fundamental strategies of virus entry and may in turn help to develop new host cell-targeted antiviral strategies.
Collapse
|
16
|
Nath S, Pigula M, Khan AP, Hanna W, Ruhi MK, Dehkordy FM, Pushpavanam K, Rege K, Moore K, Tsujita Y, Conrad C, Inci F, del Carmen MG, Franco W, Celli JP, Demirci U, Hasan T, Huang HC, Rizvi I. Flow-induced Shear Stress Confers Resistance to Carboplatin in an Adherent Three-Dimensional Model for Ovarian Cancer: A Role for EGFR-Targeted Photoimmunotherapy Informed by Physical Stress. J Clin Med 2020; 9:jcm9040924. [PMID: 32231055 PMCID: PMC7230263 DOI: 10.3390/jcm9040924] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2020] [Revised: 03/20/2020] [Accepted: 03/23/2020] [Indexed: 02/06/2023] Open
Abstract
A key reason for the persistently grim statistics associated with metastatic ovarian cancer is resistance to conventional agents, including platinum-based chemotherapies. A major source of treatment failure is the high degree of genetic and molecular heterogeneity, which results from significant underlying genomic instability, as well as stromal and physical cues in the microenvironment. Ovarian cancer commonly disseminates via transcoelomic routes to distant sites, which is associated with the frequent production of malignant ascites, as well as the poorest prognosis. In addition to providing a cell and protein-rich environment for cancer growth and progression, ascitic fluid also confers physical stress on tumors. An understudied area in ovarian cancer research is the impact of fluid shear stress on treatment failure. Here, we investigate the effect of fluid shear stress on response to platinum-based chemotherapy and the modulation of molecular pathways associated with aggressive disease in a perfusion model for adherent 3D ovarian cancer nodules. Resistance to carboplatin is observed under flow with a concomitant increase in the expression and activation of the epidermal growth factor receptor (EGFR) as well as downstream signaling members mitogen-activated protein kinase/extracellular signal-regulated kinase (MEK) and extracellular signal-regulated kinase (ERK). The uptake of platinum by the 3D ovarian cancer nodules was significantly higher in flow cultures compared to static cultures. A downregulation of phospho-focal adhesion kinase (p-FAK), vinculin, and phospho-paxillin was observed following carboplatin treatment in both flow and static cultures. Interestingly, low-dose anti-EGFR photoimmunotherapy (PIT), a targeted photochemical modality, was found to be equally effective in ovarian tumors grown under flow and static conditions. These findings highlight the need to further develop PIT-based combinations that target the EGFR, and sensitize ovarian cancers to chemotherapy in the context of flow-induced shear stress.
Collapse
Affiliation(s)
- Shubhankar Nath
- Wellman Center for Photomedicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA; (S.N.); (M.P.); (A.P.K.); (M.K.R.); (F.M.D.); (K.M.); (Y.T.); (W.F.); (T.H.)
| | - Michael Pigula
- Wellman Center for Photomedicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA; (S.N.); (M.P.); (A.P.K.); (M.K.R.); (F.M.D.); (K.M.); (Y.T.); (W.F.); (T.H.)
| | - Amjad P. Khan
- Wellman Center for Photomedicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA; (S.N.); (M.P.); (A.P.K.); (M.K.R.); (F.M.D.); (K.M.); (Y.T.); (W.F.); (T.H.)
| | - William Hanna
- Department of Physics, College of Science and Mathematics, University of Massachusetts at Boston, Boston, MA 02125, USA; (W.H.); (J.P.C.)
| | - Mustafa Kemal Ruhi
- Wellman Center for Photomedicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA; (S.N.); (M.P.); (A.P.K.); (M.K.R.); (F.M.D.); (K.M.); (Y.T.); (W.F.); (T.H.)
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill, Chapel Hill, NC and North Carolina State University, Raleigh, NC 27599, USA
| | - Farzaneh Mahmoodpoor Dehkordy
- Wellman Center for Photomedicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA; (S.N.); (M.P.); (A.P.K.); (M.K.R.); (F.M.D.); (K.M.); (Y.T.); (W.F.); (T.H.)
| | - Karthik Pushpavanam
- School for Engineering of Matter, Transport and Energy, Ira A. Fulton Schools of Engineering, Arizona State University, Tempe, AZ 85287, USA; (K.P.); (K.R.)
| | - Kaushal Rege
- School for Engineering of Matter, Transport and Energy, Ira A. Fulton Schools of Engineering, Arizona State University, Tempe, AZ 85287, USA; (K.P.); (K.R.)
| | - Kaitlin Moore
- Wellman Center for Photomedicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA; (S.N.); (M.P.); (A.P.K.); (M.K.R.); (F.M.D.); (K.M.); (Y.T.); (W.F.); (T.H.)
| | - Yujiro Tsujita
- Wellman Center for Photomedicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA; (S.N.); (M.P.); (A.P.K.); (M.K.R.); (F.M.D.); (K.M.); (Y.T.); (W.F.); (T.H.)
- Department of Urology, National Defense Medical College, Tokorozawa, Saitama 359-8513, Japan
| | - Christina Conrad
- Fischell Department of Bioengineering, University of Maryland, College Park, MD 20742, USA; (C.C.); (H.-C.H.)
| | - Fatih Inci
- Bio-Acoustic MEMS in Medicine (BAMM) Laboratory, Canary Center at Stanford for Cancer Early Detection, Department of Radiology School of Medicine Stanford University, Palo Alto, CA 94304, USA; (F.I.); (U.D.)
| | - Marcela G. del Carmen
- Division of Gynecologic Oncology, Vincent Obstetrics and Gynecology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA;
| | - Walfre Franco
- Wellman Center for Photomedicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA; (S.N.); (M.P.); (A.P.K.); (M.K.R.); (F.M.D.); (K.M.); (Y.T.); (W.F.); (T.H.)
| | - Jonathan P. Celli
- Department of Physics, College of Science and Mathematics, University of Massachusetts at Boston, Boston, MA 02125, USA; (W.H.); (J.P.C.)
| | - Utkan Demirci
- Bio-Acoustic MEMS in Medicine (BAMM) Laboratory, Canary Center at Stanford for Cancer Early Detection, Department of Radiology School of Medicine Stanford University, Palo Alto, CA 94304, USA; (F.I.); (U.D.)
| | - Tayyaba Hasan
- Wellman Center for Photomedicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA; (S.N.); (M.P.); (A.P.K.); (M.K.R.); (F.M.D.); (K.M.); (Y.T.); (W.F.); (T.H.)
| | - Huang-Chiao Huang
- Fischell Department of Bioengineering, University of Maryland, College Park, MD 20742, USA; (C.C.); (H.-C.H.)
- Marlene and Stewart Greenebaum Cancer Center, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Imran Rizvi
- Wellman Center for Photomedicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA; (S.N.); (M.P.); (A.P.K.); (M.K.R.); (F.M.D.); (K.M.); (Y.T.); (W.F.); (T.H.)
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill, Chapel Hill, NC and North Carolina State University, Raleigh, NC 27599, USA
- Lineberger Comprehensive Cancer Center, University of North Carolina School of Medicine, Chapel Hill, NC 27599, USA
- Correspondence:
| |
Collapse
|
17
|
Wang F, Diesendruck CE. Effect of disulphide loop length on mechanochemical structural stability of macromolecules. Chem Commun (Camb) 2020; 56:2143-2146. [DOI: 10.1039/c9cc07439b] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
Polymer chains folded with a single disulphide loop are shown to present distinct rates of mechanochemical fragmentation.
Collapse
Affiliation(s)
- Feng Wang
- Schulich Faculty of Chemistry and Russell-Berrie Nanotechnology Institute
- Technion – Israel Institute of Technology
- Haifa
- Israel
- School of Chemical Engineering
| | - Charles E. Diesendruck
- Schulich Faculty of Chemistry and Russell-Berrie Nanotechnology Institute
- Technion – Israel Institute of Technology
- Haifa
- Israel
| |
Collapse
|
18
|
Bailey KA, Moreno E, Haj FG, Simon SI, Passerini AG. Mechanoregulation of p38 activity enhances endoplasmic reticulum stress-mediated inflammation by arterial endothelium. FASEB J 2019; 33:12888-12899. [PMID: 31499005 DOI: 10.1096/fj.201900236r] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
Endothelial up-regulation of VCAM-1 at susceptible sites in arteries modulates the recruitment efficiency of inflammatory monocytes that initiates atherosclerotic lesion formation. We reported that hydrodynamic shear stress (SS) mechanoregulates inflammation in human aortic endothelial cells through endoplasmic reticulum (ER) stress via activation of the transcription factor x-box binding protein 1 (XBP1). Here, a microfluidic flow channel that produces a linear gradient of SS along a continuous monolayer of endothelium was used to delve the mechanisms underlying transcriptional regulation of TNF-α-stimulated VCAM-1 expression. High-resolution immunofluorescence imaging enabled continuous detection of platelet endothelial cell adhesion molecule 1 (PECAM-1)-dependent, outside-in signaling as a function of SS magnitude. Differential expression of VCAM-1 and intercellular adhesion molecule 1 (ICAM-1) was regulated by the spatiotemporal activation of MAPKs, ER stress markers, and transcription factors, which was dependent on the mechanosensing of SS through PECAM-1 and PI3K. Inhibition of p38 specifically abrogated the rise to peak VCAM-1 at low SS (2 dyn/cm2), whereas inhibition of ERK1/2 attenuated peak ICAM-1 at high SS (12 dyn/cm2). A shear stress-regulated temporal rise in p38 phosphorylation activated the nuclear translocation of XBP1, which together with the transcription factor IFN regulatory factor 1, promoted maximum VCAM-1 expression. These data reveal a mechanism by which SS sensitizes the endothelium to a cytokine-induced ER stress response to spatially regulate inflammation promoting atherosclerosis.-Bailey, K. A., Moreno, E., Haj, F. G., Simon, S. I., Passerini, A. G. Mechanoregulation of p38 activity enhances endoplasmic reticulum stress-mediated inflammation by arterial endothelium.
Collapse
Affiliation(s)
- Keith A Bailey
- Department of Biomedical Engineering, University of California-Davis, Davis, California, USA
| | - Emily Moreno
- Department of Biomedical Engineering, University of California-Davis, Davis, California, USA
| | - Fawaz G Haj
- Department of Nutrition, University of California-Davis, Davis, California, USA.,Department of Internal Medicine, University of California-Davis, Davis, California, USA
| | - Scott I Simon
- Department of Biomedical Engineering, University of California-Davis, Davis, California, USA
| | - Anthony G Passerini
- Department of Biomedical Engineering, University of California-Davis, Davis, California, USA
| |
Collapse
|
19
|
Meza D, Musmacker B, Steadman E, Stransky T, Rubenstein DA, Yin W. Endothelial Cell Biomechanical Responses are Dependent on Both Fluid Shear Stress and Tensile Strain. Cell Mol Bioeng 2019; 12:311-325. [PMID: 31719917 DOI: 10.1007/s12195-019-00585-0] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2018] [Accepted: 06/28/2019] [Indexed: 12/15/2022] Open
Abstract
Introduction The goal of this study was to investigate how concurrent shear stress and tensile strain affect endothelial cell biomechanical responses. Methods Human coronary artery endothelial cells were exposed to concurrent pulsatile shear stress and cyclic tensile strain in a programmable shearing and stretching device. Three shear stress-tensile strain conditions were used: (1) pulsatile shear stress at 1 Pa and cyclic tensile strain at 7%, simulating normal stress/strain conditions in a healthy coronary artery; (2) shear stress at 3.7 Pa and tensile strain at 3%, simulating pathological stress/strain conditions near a stenosis; (3) shear stress at 0.7 Pa and tensile strain at 5%, simulating pathological stress/strain conditions in a recirculation zone. Cell morphology was quantified using immunofluorescence microscopy. Cell surface PECAM-1 phosphorylation, ICAM-1 expression, ERK1/2 and NF-κB activation were measured using ELISA or Western blot. Results Simultaneous stimulation from pulsatile shear stress and cyclic tensile strain induced a significant increase in cell area, compared to that induced by shear stress or tensile strain alone. The combined stimulation caused significant increases in PECAM-1 phosphorylation. The combined stimulation also significantly enhanced EC surface ICAM-1 expression (compared to that under shear stress alone) and transcriptional factor NF-κB activation (compared to that under control conditions). Conclusion Pulsatile shear stress and cyclic tensile strain could induce increased but not synergistic effect on endothelial cell morphology or activation. The combined mechanical stimulation can be relayed from cell membrane to nucleus. Therefore, to better understand how mechanical conditions affect endothelial cell mechanotransduction and cardiovascular disease development, both shear stress and tensile strain need to be considered.
Collapse
Affiliation(s)
- Daphne Meza
- Department of Biomedical Engineering, Stony Brook University, Stony Brook, NY 11794 USA
| | - Bryan Musmacker
- Department of Biomedical Engineering, Stony Brook University, Stony Brook, NY 11794 USA
| | - Elisabeth Steadman
- Department of Biomedical Engineering, Stony Brook University, Stony Brook, NY 11794 USA
| | - Thomas Stransky
- Department of Biomedical Engineering, Stony Brook University, Stony Brook, NY 11794 USA
| | - David A Rubenstein
- Department of Biomedical Engineering, Stony Brook University, Stony Brook, NY 11794 USA
| | - Wei Yin
- Department of Biomedical Engineering, Stony Brook University, Stony Brook, NY 11794 USA
- Stony Brook University, Bioengineering Building, Room 109, Stony Brook, NY 11794 USA
| |
Collapse
|
20
|
Tobin SW, Alibhai FJ, Lee MM, Yeganeh A, Wu J, Li SH, Guo J, Tsang K, Tumiati L, Rocha R, Butany J, Yau TM, Ouzounian M, David TE, Weisel RD, Li RK. Novel mediators of aneurysm progression in bicuspid aortic valve disease. J Mol Cell Cardiol 2019; 132:71-83. [PMID: 31047984 DOI: 10.1016/j.yjmcc.2019.04.022] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2018] [Revised: 04/06/2019] [Accepted: 04/20/2019] [Indexed: 12/28/2022]
Abstract
Bicuspid aortic valve (BAV) disease is a congenital abnormality that is associated with ascending aortic aneurysm yet many of the molecular mechanisms remain unknown. To identify novel molecular mechanisms of aneurysm formation we completed microarray analysis of the proximal (severely dilated) and distal (less dilated) regions of the ascending aorta from five patients with BAV. We identified 180 differentially expressed genes, 40 of which were validated by RT-qPCR. Most genes had roles in inflammation and endothelial cell function including cytokines and growth factors, cell surface receptors and the Activator Protein 1 (AP-1) transcription factor family (FOS, FOSB and JUN) which was chosen for further study. AP-1 was differentially expressed within paired BAV aneurysmal samples (n = 8) but not Marfan patients (n = 5). FOS protein was significantly enriched in BAV aortas compared to normal aortas but unexpectedly, ERK1/2 activity, an upstream regulator of FOS was reduced. ERK1/2 activity was restored when BAV smooth muscle cells were cultured in vitro. An mRNA-miRNA network within paired patient samples identified AP-1 as a central hub of miRNA regulation. FOS knockdown in BAV SMCs increased expression of miR-27a, a stretch responsive miRNA. AP-1 and miR-27a were also dysregulated in a mouse model of aortic constriction. In summary, this study identified a central role for AP-1 signaling in BAV aortic dilatation by using paired mRNA-miRNA patient sample. Upstream analysis of AP-1 regulation showed that the ERK1/2 signaling pathway is dysregulated and thus represents a novel chain of mediators of aortic dilatation in BAV which should be considered in future studies.
Collapse
Affiliation(s)
- Stephanie W Tobin
- Division of Cardiovascular Surgery, Toronto General Hospital Research Institute and Peter Munk Cardiac Centre, University Health Network, Toronto, ON, Canada
| | - Faisal J Alibhai
- Division of Cardiovascular Surgery, Toronto General Hospital Research Institute and Peter Munk Cardiac Centre, University Health Network, Toronto, ON, Canada
| | - Myunghyun M Lee
- Division of Cardiovascular Surgery, Toronto General Hospital Research Institute and Peter Munk Cardiac Centre, University Health Network, Toronto, ON, Canada; Department of Surgery, Division of Cardiac Surgery, University of Toronto, Toronto, ON, Canada
| | - Azadeh Yeganeh
- Division of Cardiovascular Surgery, Toronto General Hospital Research Institute and Peter Munk Cardiac Centre, University Health Network, Toronto, ON, Canada
| | - Jie Wu
- Division of Cardiovascular Surgery, Toronto General Hospital Research Institute and Peter Munk Cardiac Centre, University Health Network, Toronto, ON, Canada; Laboratory of Medical Genetics, Harbin Medical University, Harbin, China
| | - Shu-Hong Li
- Division of Cardiovascular Surgery, Toronto General Hospital Research Institute and Peter Munk Cardiac Centre, University Health Network, Toronto, ON, Canada
| | - Jian Guo
- Division of Cardiovascular Surgery, Toronto General Hospital Research Institute and Peter Munk Cardiac Centre, University Health Network, Toronto, ON, Canada
| | - Katherine Tsang
- Division of Cardiovascular Surgery, Toronto General Hospital Research Institute and Peter Munk Cardiac Centre, University Health Network, Toronto, ON, Canada; Department of Surgery, Division of Cardiac Surgery, University of Toronto, Toronto, ON, Canada
| | - Laura Tumiati
- Division of Cardiovascular Surgery, Toronto General Hospital Research Institute and Peter Munk Cardiac Centre, University Health Network, Toronto, ON, Canada; Department of Surgery, Division of Cardiac Surgery, University of Toronto, Toronto, ON, Canada
| | - Rodolfo Rocha
- Division of Cardiovascular Surgery, Toronto General Hospital Research Institute and Peter Munk Cardiac Centre, University Health Network, Toronto, ON, Canada; Department of Surgery, Division of Cardiac Surgery, University of Toronto, Toronto, ON, Canada
| | - Jagdish Butany
- Department of Pathology, University Health Network, Toronto, ON, Canada
| | - Terrence M Yau
- Division of Cardiovascular Surgery, Toronto General Hospital Research Institute and Peter Munk Cardiac Centre, University Health Network, Toronto, ON, Canada; Department of Surgery, Division of Cardiac Surgery, University of Toronto, Toronto, ON, Canada
| | - Maral Ouzounian
- Division of Cardiovascular Surgery, Toronto General Hospital Research Institute and Peter Munk Cardiac Centre, University Health Network, Toronto, ON, Canada; Department of Surgery, Division of Cardiac Surgery, University of Toronto, Toronto, ON, Canada
| | - Tirone E David
- Division of Cardiovascular Surgery, Toronto General Hospital Research Institute and Peter Munk Cardiac Centre, University Health Network, Toronto, ON, Canada; Department of Surgery, Division of Cardiac Surgery, University of Toronto, Toronto, ON, Canada
| | - Richard D Weisel
- Division of Cardiovascular Surgery, Toronto General Hospital Research Institute and Peter Munk Cardiac Centre, University Health Network, Toronto, ON, Canada; Department of Surgery, Division of Cardiac Surgery, University of Toronto, Toronto, ON, Canada
| | - Ren-Ke Li
- Division of Cardiovascular Surgery, Toronto General Hospital Research Institute and Peter Munk Cardiac Centre, University Health Network, Toronto, ON, Canada; Department of Surgery, Division of Cardiac Surgery, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
21
|
Fang Y, Wu D, Birukov KG. Mechanosensing and Mechanoregulation of Endothelial Cell Functions. Compr Physiol 2019; 9:873-904. [PMID: 30873580 PMCID: PMC6697421 DOI: 10.1002/cphy.c180020] [Citation(s) in RCA: 125] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Vascular endothelial cells (ECs) form a semiselective barrier for macromolecules and cell elements regulated by dynamic interactions between cytoskeletal elements and cell adhesion complexes. ECs also participate in many other vital processes including innate immune reactions, vascular repair, secretion, and metabolism of bioactive molecules. Moreover, vascular ECs represent a unique cell type exposed to continuous, time-dependent mechanical forces: different patterns of shear stress imposed by blood flow in macrovasculature and by rolling blood cells in the microvasculature; circumferential cyclic stretch experienced by the arterial vascular bed caused by heart propulsions; mechanical stretch of lung microvascular endothelium at different magnitudes due to spontaneous respiration or mechanical ventilation in critically ill patients. Accumulating evidence suggests that vascular ECs contain mechanosensory complexes, which rapidly react to changes in mechanical loading, process the signal, and develop context-specific adaptive responses to rebalance the cell homeostatic state. The significance of the interactions between specific mechanical forces in the EC microenvironment together with circulating bioactive molecules in the progression and resolution of vascular pathologies including vascular injury, atherosclerosis, pulmonary edema, and acute respiratory distress syndrome has been only recently recognized. This review will summarize the current understanding of EC mechanosensory mechanisms, modulation of EC responses to humoral factors by surrounding mechanical forces (particularly the cyclic stretch), and discuss recent findings of magnitude-specific regulation of EC functions by transcriptional, posttranscriptional and epigenetic mechanisms using -omics approaches. We also discuss ongoing challenges and future opportunities in developing new therapies targeting dysregulated mechanosensing mechanisms to treat vascular diseases. © 2019 American Physiological Society. Compr Physiol 9:873-904, 2019.
Collapse
Affiliation(s)
- Yun Fang
- Department of Medicine, University of Chicago, Chicago, Illinois, USA,Correspondence to
| | - David Wu
- Department of Medicine, University of Chicago, Chicago, Illinois, USA
| | - Konstantin G. Birukov
- Department of Anesthesiology, University of Maryland Baltimore School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
22
|
PAR-1 is a novel mechano-sensor transducing laminar flow-mediated endothelial signaling. Sci Rep 2018; 8:15172. [PMID: 30310081 PMCID: PMC6181929 DOI: 10.1038/s41598-018-33222-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2018] [Accepted: 09/18/2018] [Indexed: 01/01/2023] Open
Abstract
Recent studies have indicated that protease-activated receptor-1 (PAR-1) is involved in cytoprotective and anti-inflammatory responses in endothelial cells (ECs). However, the role of PAR-1 in laminar flow-mediated atheroprotective responses remains unknown. Herein, we investigated whether PAR-1 regulates laminar flow-mediated mechanotransduction in ECs. Confocal analysis showed that PAR-1 was internalized into early endosomes in response to laminar flow. In addition, flow cytometry analysis showed that cell surface expression of PAR-1 was reduced by laminar flow, suggesting that PAR-1 was activated in response to laminar flow. Depletion of PAR-1 using human PAR-1 siRNA inhibited unidirectional laminar flow-mediated actin stress fiber formation and cellular alignment as well as atheroprotective gene expressions in HUVECs. Moreover, PAR-1 knockdown inhibited laminar flow-stimulated eNOS phosphorylation, and inhibited the phosphorylations of Src, AMPK, ERK5 and HDAC5. Furthermore, PAR-1 depletion inhibited laminar flow-mediated anti-inflammatory responses as demonstrated by reduced TNFα-induced VCAM-1 expression and by monocyte adhesion to HUVECs, and prevented laminar flow-mediated anti-apoptotic response. An investigation of the role of PAR-1 in vasomotor modulation using mouse aortic rings revealed that acetylcholine-induced vasorelaxation was diminished in PAR-1 deficient mice compared to littermate controls. Taken together, these findings suggest that PAR-1 be viewed as a novel pharmacologic target for the treatment of vascular diseases, including atherosclerosis.
Collapse
|
23
|
Shear-Induced Nitric Oxide Production by Endothelial Cells. Biophys J 2017; 111:208-21. [PMID: 27410748 DOI: 10.1016/j.bpj.2016.05.034] [Citation(s) in RCA: 85] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2015] [Revised: 04/30/2016] [Accepted: 05/23/2016] [Indexed: 02/06/2023] Open
Abstract
We present a biochemical model of the wall shear stress-induced activation of endothelial nitric oxide synthase (eNOS) in an endothelial cell. The model includes three key mechanotransducers: mechanosensing ion channels, integrins, and G protein-coupled receptors. The reaction cascade consists of two interconnected parts. The first is rapid activation of calcium, which results in formation of calcium-calmodulin complexes, followed by recruitment of eNOS from caveolae. The second is phosphorylation of eNOS by protein kinases PKC and AKT. The model also includes a negative feedback loop due to inhibition of calcium influx into the cell by cyclic guanosine monophosphate (cGMP). In this feedback, increased nitric oxide (NO) levels cause an increase in cGMP levels, so that cGMP inhibition of calcium influx can limit NO production. The model was used to predict the dynamics of NO production by an endothelial cell subjected to a step increase of wall shear stress from zero to a finite physiologically relevant value. Among several experimentally observed features, the model predicts a highly nonlinear, biphasic transient behavior of eNOS activation and NO production: a rapid initial activation due to the very rapid influx of calcium into the cytosol (occurring within 1-5 min) is followed by a sustained period of activation due to protein kinases.
Collapse
|
24
|
Barr LA, Lambert JP, Shimizu Y, Barouch LA, Naqvi N, Calvert JW. Exercise training provides cardioprotection by activating and coupling endothelial nitric oxide synthase via a β 3-adrenergic receptor-AMP-activated protein kinase signaling pathway. Med Gas Res 2017; 7:1-8. [PMID: 28480026 PMCID: PMC5402342 DOI: 10.4103/2045-9912.202904] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/02/2022] Open
Abstract
Exercise training confers sustainable protection against ischemia/reperfusion injury. However, the mechanism by which this process occurs is not fully understood. Previously, it was shown that β3-adrenergic receptors (β3-ARs) play a critical role in regulating the activation of endothelial nitric oxide synthase (eNOS) in response to exercise and play a critical role in exercise-mediated cardioprotection. Intriguingly, a deficiency in β3-ARs led to increased myocardial injury following exercise training. The purpose of the current study was to determine mechanisms by which β3-ARs are linked to eNOS activation and to determine the mechanism responsible for the exacerbated ischemia/reperfusion injury displayed by β3-AR deficient (β3-AR KO) mice after exercise training. Wild-type (n = 37) and β3-AR KO (n = 40) mice were subjected to voluntary wheel running for 4 weeks. Western blot analysis revealed that neither protein kinase B nor protein kinase A linked β3-ARs to eNOS following exercise training. However, analysis revealed a role for AMP-activated protein kinase (AMPK). Specifically, exercise training increased the phosphorylation of AMPK in the hearts of wild-type mice, but failed to do so in the hearts of β3-AR KO mice. Additional studies revealed that exercise training rendered eNOS less coupled and increased NOS-dependent superoxide levels in β3-AR KO mice. Finally, supplementing β3-AR KO mice with the eNOS coupler, tetrahydrobiopterin, during the final week of exercise training reduced myocardial infarction. These findings provide important information that exercise training protects the heart in the setting of myocardial ischemia/reperfusion injury by activating and coupling eNOS via the stimulation of a β3-AR-AMPK signaling pathway.
Collapse
Affiliation(s)
- Larry A Barr
- Department of Surgery, Division of Cardiothoracic Surgery, Carlyle Fraser Heart Center, Emory University School of Medicine, Atlanta, GA, USA
| | - Jonathan P Lambert
- Department of Surgery, Division of Cardiothoracic Surgery, Carlyle Fraser Heart Center, Emory University School of Medicine, Atlanta, GA, USA
| | - Yuuki Shimizu
- Department of Surgery, Division of Cardiothoracic Surgery, Carlyle Fraser Heart Center, Emory University School of Medicine, Atlanta, GA, USA
| | - Lili A Barouch
- Department of Medicine, Division of Cardiology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Nawazish Naqvi
- Department of Medicine, Division of Cardiology, Emory University School of Medicine, Atlanta, GA, USA
| | - John W Calvert
- Department of Surgery, Division of Cardiothoracic Surgery, Carlyle Fraser Heart Center, Emory University School of Medicine, Atlanta, GA, USA
| |
Collapse
|
25
|
Lee HJ, Diaz MF, Price KM, Ozuna JA, Zhang S, Sevick-Muraca EM, Hagan JP, Wenzel PL. Fluid shear stress activates YAP1 to promote cancer cell motility. Nat Commun 2017; 8:14122. [PMID: 28098159 PMCID: PMC5253685 DOI: 10.1038/ncomms14122] [Citation(s) in RCA: 176] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2016] [Accepted: 11/04/2016] [Indexed: 12/28/2022] Open
Abstract
Mechanical stress is pervasive in egress routes of malignancy, yet the intrinsic effects of force on tumour cells remain poorly understood. Here, we demonstrate that frictional force characteristic of flow in the lymphatics stimulates YAP1 to drive cancer cell migration; whereas intensities of fluid wall shear stress (WSS) typical of venous or arterial flow inhibit taxis. YAP1, but not TAZ, is strictly required for WSS-enhanced cell movement, as blockade of YAP1, TEAD1-4 or the YAP1–TEAD interaction reduces cellular velocity to levels observed without flow. Silencing of TEAD phenocopies loss of YAP1, implicating transcriptional transactivation function in mediating force-enhanced cell migration. WSS dictates expression of a network of YAP1 effectors with executive roles in invasion, chemotaxis and adhesion downstream of the ROCK–LIMK–cofilin signalling axis. Altogether, these data implicate YAP1 as a fluid mechanosensor that functions to regulate genes that promote metastasis. Fluid frictional forces around cancer cells influence chemokine production and delivery of chemotherapeutic drugs but it is unclear if they directly impact tumour biology through biomechanical effects. Here, the authors show that wall shear stress stimulates cancer cell migration through a ROCK–LIMK–YAP axis.
Collapse
Affiliation(s)
- Hyun Jung Lee
- Children's Regenerative Medicine Program, Department of Pediatric Surgery, University of Texas Health Science Center at Houston, Houston, Texas 77030, USA.,Center for Stem Cell and Regenerative Medicine, The Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center at Houston, Houston, Texas 77030, USA
| | - Miguel F Diaz
- Children's Regenerative Medicine Program, Department of Pediatric Surgery, University of Texas Health Science Center at Houston, Houston, Texas 77030, USA.,Center for Stem Cell and Regenerative Medicine, The Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center at Houston, Houston, Texas 77030, USA
| | - Katherine M Price
- Department of BioSciences, Rice University, Houston, Texas 77030, USA
| | - Joyce A Ozuna
- Department of BioSciences, Rice University, Houston, Texas 77030, USA
| | - Songlin Zhang
- Department of Pathology, The University of Texas Medical School, Houston, Texas 77030, USA
| | - Eva M Sevick-Muraca
- Center for Molecular Imaging, The Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center at Houston, Houston, Texas 77030, USA
| | - John P Hagan
- Vivian L. Smith Department of Neurosurgery, University of Texas Health Science Center at Houston, Houston, Texas 77030, USA
| | - Pamela L Wenzel
- Children's Regenerative Medicine Program, Department of Pediatric Surgery, University of Texas Health Science Center at Houston, Houston, Texas 77030, USA.,Center for Stem Cell and Regenerative Medicine, The Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center at Houston, Houston, Texas 77030, USA
| |
Collapse
|
26
|
Meza D, Shanmugavelayudam SK, Mendoza A, Sanchez C, Rubenstein DA, Yin W. Platelets modulate endothelial cell response to dynamic shear stress through PECAM-1. Thromb Res 2016; 150:44-50. [PMID: 28013181 DOI: 10.1016/j.thromres.2016.12.003] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2016] [Revised: 11/07/2016] [Accepted: 12/04/2016] [Indexed: 01/13/2023]
Abstract
INTRODUCTION Both vascular endothelial cells and platelets are sensitive to blood flow induced shear stress. We have recently reported that platelet-endothelial cell interaction could greatly affect platelet activation under flow. In the present study, we aimed to investigate how platelet-endothelial cell interaction affected endothelial cell inflammatory responses under flow. MATERIALS AND METHODS Human coronary artery endothelial cells were exposed to normal or low pulsatile shear stress with or without the presence of platelets. Following shear exposure, endothelial cell ICAM-1 expression was measured using ELISA, Western blot and PCR; cell surface PECAM-1 expression/phosphorylation was measured using ELISA. Platelet adhesion to endothelial cells was quantified using immunofluorescence microscopy. To determine the role of PECAM-1 in platelet-endothelial cell interaction, endothelial cell PECAM-1 expression was suppressed using siRNA. RESULTS Pathological low shear stress induced a significant increase in endothelial cell ICAM-1 expression, at both protein and mRNA levels. Platelet adhesion to endothelial cells increased significantly under low shear stress, co-localizing with PECAM-1 at endothelial cell junctions. The presence of platelets inhibited low shear stress-induced ICAM-1 upregulation. When endothelial cell PECAM-1 expression was suppressed, platelet adhesion to endothelial cells under low shear stress decreased significantly; endothelial cell ICAM-1 expression was not affected by shear stress, with or without platelets. CONCLUSIONS These results suggested that PECAM-1 could mediate platelet adhesion to endothelial cells under shear stress. Platelets binding to endothelial cells interfered with endothelial cell mechanotransduction through PECAM-1, affecting endothelial cell inflammatory responses towards pathological shear flow.
Collapse
Affiliation(s)
- Daphne Meza
- Department of Biomedical Engineering, Stony Brook University, Stony Brook, NY 11794, United States
| | - Saravan K Shanmugavelayudam
- School of Mechanical and Aerospace Engineering, Oklahoma State University, Stillwater, OK 74074, United States
| | - Arielys Mendoza
- Department of Biomedical Engineering, Stony Brook University, Stony Brook, NY 11794, United States
| | - Coralys Sanchez
- Department of Biomedical Engineering, Stony Brook University, Stony Brook, NY 11794, United States
| | - David A Rubenstein
- Department of Biomedical Engineering, Stony Brook University, Stony Brook, NY 11794, United States
| | - Wei Yin
- Department of Biomedical Engineering, Stony Brook University, Stony Brook, NY 11794, United States; School of Mechanical and Aerospace Engineering, Oklahoma State University, Stillwater, OK 74074, United States.
| |
Collapse
|
27
|
Wang YI, Abaci HE, Shuler ML. Microfluidic blood-brain barrier model provides in vivo-like barrier properties for drug permeability screening. Biotechnol Bioeng 2016; 114:184-194. [PMID: 27399645 DOI: 10.1002/bit.26045] [Citation(s) in RCA: 379] [Impact Index Per Article: 42.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2016] [Revised: 05/19/2016] [Accepted: 07/04/2016] [Indexed: 12/23/2022]
Abstract
Efficient delivery of therapeutics across the neuroprotective blood-brain barrier (BBB) remains a formidable challenge for central nervous system drug development. High-fidelity in vitro models of the BBB could facilitate effective early screening of drug candidates targeting the brain. In this study, we developed a microfluidic BBB model that is capable of mimicking in vivo BBB characteristics for a prolonged period and allows for reliable in vitro drug permeability studies under recirculating perfusion. We derived brain microvascular endothelial cells (BMECs) from human induced pluripotent stem cells (hiPSCs) and cocultured them with rat primary astrocytes on the two sides of a porous membrane on a pumpless microfluidic platform for up to 10 days. The microfluidic system was designed based on the blood residence time in human brain tissues, allowing for medium recirculation at physiologically relevant perfusion rates with no pumps or external tubing, meanwhile minimizing wall shear stress to test whether shear stress is required for in vivo-like barrier properties in a microfluidic BBB model. This BBB-on-a-chip model achieved significant barrier integrity as evident by continuous tight junction formation and in vivo-like values of trans-endothelial electrical resistance (TEER). The TEER levels peaked above 4000 Ω · cm2 on day 3 on chip and were sustained above 2000 Ω · cm2 up to 10 days, which are the highest sustained TEER values reported in a microfluidic model. We evaluated the capacity of our microfluidic BBB model to be used for drug permeability studies using large molecules (FITC-dextrans) and model drugs (caffeine, cimetidine, and doxorubicin). Our analyses demonstrated that the permeability coefficients measured using our model were comparable to in vivo values. Our BBB-on-a-chip model closely mimics physiological BBB barrier functions and will be a valuable tool for screening of drug candidates. The residence time-based design of a microfluidic platform will enable integration with other organ modules to simulate multi-organ interactions on drug response. Biotechnol. Bioeng. 2017;114: 184-194. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Ying I Wang
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, 381 Kimball Hall, Ithaca, New York, 14853-7202
| | - Hasan Erbil Abaci
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, 381 Kimball Hall, Ithaca, New York, 14853-7202
| | - Michael L Shuler
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, 381 Kimball Hall, Ithaca, New York, 14853-7202
| |
Collapse
|
28
|
Luff SA, Papoutsakis ET. Megakaryocytic Maturation in Response to Shear Flow Is Mediated by the Activator Protein 1 (AP-1) Transcription Factor via Mitogen-activated Protein Kinase (MAPK) Mechanotransduction. J Biol Chem 2016; 291:7831-43. [PMID: 26814129 DOI: 10.1074/jbc.m115.707174] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2015] [Indexed: 12/26/2022] Open
Abstract
Megakaryocytes (MKs) are exposed to shear flow as they migrate from the bone marrow hematopoietic compartment into circulation to release pro/preplatelets into circulating blood. Shear forces promote DNA synthesis, polyploidization, and maturation in MKs, and platelet biogenesis. To investigate mechanisms underlying these MK responses to shear, we carried out transcriptional analysis on immature and mature stem cell-derived MKs exposed to physiological shear. In immature (day (d)9) MKs, shear exposure up-regulated genes related to growth and MK maturation, whereas in mature (d12) MKs, it up-regulated genes involved in apoptosis and intracellular transport. Following shear-flow exposure, six activator protein 1 (AP-1) transcripts (ATF4,JUNB,JUN,FOSB,FOS, andJUND) were up-regulated at d9 and two AP-1 proteins (JunD and c-Fos) were up-regulated both at d9 and d12. We show that mitogen-activated protein kinase (MAPK) signaling is linked to both the shear stress response and AP-1 up-regulation. c-Jun N-terminal kinase (JNK) phosphorylation increased significantly following shear stimulation, whereas JNK inhibition reduced shear-induced JunD expression. Although p38 phosphorylation did not increase following shear flow, its inhibition reduced shear-induced JunD and c-Fos expression. JNK inhibition reduced fibrinogen binding and P-selectin expression of d12 platelet-like particles (PLPs), whereas p38 inhibition reduced fibrinogen binding of d12 PLPs. AP-1 expression correlated with increased MK DNA synthesis and polyploidization, which might explain the observed impact of shear on MKs. To summarize, we show that MK exposure to shear forces results in JNK activation, AP-1 up-regulation, and downstream transcriptional changes that promote maturation of immature MKs and platelet biogenesis in mature MKs.
Collapse
Affiliation(s)
- Stephanie A Luff
- From the Department of Biological Sciences, Delaware Biotechnology Institute, and
| | - Eleftherios T Papoutsakis
- From the Department of Biological Sciences, Delaware Biotechnology Institute, and Department of Chemical and Biomolecular Engineering, University of Delaware, Newark, Delaware 19711
| |
Collapse
|
29
|
Kohn JC, Zhou DW, Bordeleau F, Zhou AL, Mason BN, Mitchell MJ, King MR, Reinhart-King CA. Cooperative effects of matrix stiffness and fluid shear stress on endothelial cell behavior. Biophys J 2015; 108:471-8. [PMID: 25650915 DOI: 10.1016/j.bpj.2014.12.023] [Citation(s) in RCA: 118] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2014] [Revised: 12/02/2014] [Accepted: 12/15/2014] [Indexed: 11/28/2022] Open
Abstract
Arterial hemodynamic shear stress and blood vessel stiffening both significantly influence the arterial endothelial cell (EC) phenotype and atherosclerosis progression, and both have been shown to signal through cell-matrix adhesions. However, the cooperative effects of fluid shear stress and matrix stiffness on ECs remain unknown. To investigate these cooperative effects, we cultured bovine aortic ECs on hydrogels matching the elasticity of the intima of compliant, young, or stiff, aging arteries. The cells were then exposed to laminar fluid shear stress of 12 dyn/cm(2). Cells grown on more compliant matrices displayed increased elongation and tighter EC-cell junctions. Notably, cells cultured on more compliant substrates also showed decreased RhoA activation under laminar shear stress. Additionally, endothelial nitric oxide synthase and extracellular signal-regulated kinase phosphorylation in response to fluid shear stress occurred more rapidly in ECs cultured on more compliant substrates, and nitric oxide production was enhanced. Together, our results demonstrate that a signaling cross talk between stiffness and fluid shear stress exists within the vascular microenvironment, and, importantly, matrices mimicking young and healthy blood vessels can promote and augment the atheroprotective signals induced by fluid shear stress. These data suggest that targeting intimal stiffening and/or the EC response to intima stiffening clinically may improve vascular health.
Collapse
Affiliation(s)
- Julie C Kohn
- Department of Biomedical Engineering, Cornell University, Ithaca, New York
| | - Dennis W Zhou
- Department of Biomedical Engineering, Cornell University, Ithaca, New York
| | - François Bordeleau
- Department of Biomedical Engineering, Cornell University, Ithaca, New York
| | - Allen L Zhou
- Department of Biomedical Engineering, Cornell University, Ithaca, New York
| | - Brooke N Mason
- Department of Biomedical Engineering, Cornell University, Ithaca, New York
| | - Michael J Mitchell
- Department of Biomedical Engineering, Cornell University, Ithaca, New York
| | - Michael R King
- Department of Biomedical Engineering, Cornell University, Ithaca, New York
| | | |
Collapse
|
30
|
Wang S, Iring A, Strilic B, Albarrán Juárez J, Kaur H, Troidl K, Tonack S, Burbiel JC, Müller CE, Fleming I, Lundberg JO, Wettschureck N, Offermanns S. P2Y₂ and Gq/G₁₁ control blood pressure by mediating endothelial mechanotransduction. J Clin Invest 2015; 125:3077-86. [PMID: 26168216 DOI: 10.1172/jci81067] [Citation(s) in RCA: 157] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2015] [Accepted: 05/28/2015] [Indexed: 12/17/2022] Open
Abstract
Elevated blood pressure is a key risk factor for developing cardiovascular diseases. Blood pressure is largely determined by vasodilatory mediators, such as nitric oxide (NO), that are released from the endothelium in response to fluid shear stress exerted by the flowing blood. Previous work has identified several mechanotransduction signaling processes that are involved in fluid shear stress-induced endothelial effects, but how fluid shear stress initiates the response is poorly understood. Here, we evaluated human and bovine endothelial cells and found that the purinergic receptor P2Y2 and the G proteins Gq/G11 mediate fluid shear stress-induced endothelial responses, including [Ca2+]i transients, activation of the endothelial NO synthase (eNOS), phosphorylation of PECAM-1 and VEGFR-2, as well as activation of SRC and AKT. In response to fluid shear stress, endothelial cells released ATP, which activates the purinergic P2Y2 receptor. Mice with induced endothelium-specific P2Y2 or Gq/G11 deficiency lacked flow-induced vasodilation and developed hypertension that was accompanied by reduced eNOS activation. Together, our data identify P2Y2 and Gq/G11 as a critical endothelial mechanosignaling pathway that is upstream of previously described mechanotransduction processes and demonstrate that P2Y2 and Gq/G11 are required for basal endothelial NO formation, vascular tone, and blood pressure.
Collapse
|
31
|
Vilahur G, Cubedo J, Padró T, Casaní L, Mendieta G, González A, Badimon L. Intake of cooked tomato sauce preserves coronary endothelial function and improves apolipoprotein A-I and apolipoprotein J protein profile in high-density lipoproteins. Transl Res 2015; 166:44-56. [PMID: 25514506 DOI: 10.1016/j.trsl.2014.11.004] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/28/2014] [Revised: 11/11/2014] [Accepted: 11/13/2014] [Indexed: 10/24/2022]
Abstract
Intake of tomatoes has been linked with healthy diets (eg, Mediterranean diet). However, it remains unknown whether tomato intake exerts protective effects on the vasculature. The aim of this study was to determine whether medium-term supplementation with cooked tomato sauce (CTS) Mediterranean style (sofrito) attenuates diet-induced coronary endothelial dysfunction in an animal model with clinical impact and explore the mechanisms behind the effects. Pigs (N = 18) were fed a 10-day hypercholesterolemic diet. Half of the animals were given a supplement of 100 g/d of CTS (21.5 mg lycopene per day). Coronary responses to escalating doses of vasoactive drugs (acetylcholine, calcium ionophore, and sodium nitroprusside) and L-NG-monomethylarginine (endothelial nitric oxide synthase [eNOS] inhibitor) were measured using flow Doppler. In the coronary arteries, we investigated eNOS gene expression and activation, monocyte chemoattractant protein 1 (MCP-1) expression, and oxidative DNA damage. In the circulation, we investigated lipoprotein resistance to oxidation and the differential proteomic protein profile. In dyslipidemic animals, CTS intake prevented diet-induced impairment of receptor-operated and nonreceptor-operated endothelial-dependent coronary vasodilation. These beneficial effects were associated with enhanced eNOS transcription and activation and diminished DNA damage in the coronary arteries. CTS-fed animals showed lower lipid peroxidation, higher high-density lipoprotein (HDL) antioxidant potential and plasma lycopene levels of 0.16 mg/L. Interestingly, improved HDL functionality was associated with protein profile changes in apolipoprotein A-I and apolipoprotein J. Lipids levels and MCP-1 expression were not affected by CTS. We report that CTS intake protects against low-density lipoprotein-induced coronary endothelial dysfunction by reducing oxidative damage, enhancing eNOS expression and activity, and improving HDL functionality.
Collapse
Affiliation(s)
- Gemma Vilahur
- Cardiovascular Research Center, CSIC-ICCC, Hospital de la Santa Creu i Sant Pau, IIB-Sant Pau, Barcelona, Spain
| | - Judit Cubedo
- Cardiovascular Research Center, CSIC-ICCC, Hospital de la Santa Creu i Sant Pau, IIB-Sant Pau, Barcelona, Spain
| | - Teresa Padró
- Cardiovascular Research Center, CSIC-ICCC, Hospital de la Santa Creu i Sant Pau, IIB-Sant Pau, Barcelona, Spain
| | - Laura Casaní
- Cardiovascular Research Center, CSIC-ICCC, Hospital de la Santa Creu i Sant Pau, IIB-Sant Pau, Barcelona, Spain
| | - Guiomar Mendieta
- Cardiovascular Research Center, CSIC-ICCC, Hospital de la Santa Creu i Sant Pau, IIB-Sant Pau, Barcelona, Spain
| | - Alicia González
- Nutrition and Health Unit, Gallina Blanca Star, Barcelona, Spain
| | - Lina Badimon
- Cardiovascular Research Center, CSIC-ICCC, Hospital de la Santa Creu i Sant Pau, IIB-Sant Pau, Barcelona, Spain; Cardiovascular Research Chair, UAB, Barcelona, Spain.
| |
Collapse
|
32
|
Digital microfluidic immunocytochemistry in single cells. Nat Commun 2015; 6:7513. [PMID: 26104298 PMCID: PMC4491823 DOI: 10.1038/ncomms8513] [Citation(s) in RCA: 83] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2015] [Accepted: 05/14/2015] [Indexed: 01/06/2023] Open
Abstract
We report a new technique called Digital microfluidic Immunocytochemistry in Single Cells (DISC). DISC automates protocols for cell culture, stimulation and immunocytochemistry, enabling the interrogation of protein phosphorylation on pulsing with stimulus for as little as 3 s. DISC was used to probe the phosphorylation states of platelet-derived growth factor receptor (PDGFR) and the downstream signalling protein, Akt, to evaluate concentration- and time-dependent effects of stimulation. The high time resolution of the technique allowed for surprising new observations-for example, a 10 s pulse stimulus of a low concentration of PDGF is sufficient to cause >30% of adherent fibroblasts to commit to Akt activation. With the ability to quantitatively probe signalling events with high time resolution at the single-cell level, we propose that DISC may be an important new technique for a wide range of applications, especially for screening signalling responses of a heterogeneous cell population.
Collapse
|
33
|
Dunn J, Simmons R, Thabet S, Jo H. The role of epigenetics in the endothelial cell shear stress response and atherosclerosis. Int J Biochem Cell Biol 2015; 67:167-76. [PMID: 25979369 DOI: 10.1016/j.biocel.2015.05.001] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2015] [Revised: 04/30/2015] [Accepted: 05/02/2015] [Indexed: 12/15/2022]
Abstract
Currently in the field of vascular biology, the role of epigenetics in endothelial cell biology and vascular disease has attracted more in-depth study. Using both in vitro and in vivo models of blood flow, investigators have recently begun to reveal the underlying epigenetic regulation of endothelial gene expression. Recently, our group, along with two other independent groups, have demonstrated that blood flow controls endothelial gene expression by DNA methyltransferases (DNMT1 and 3A). Disturbed flow (d-flow), characterized by low and oscillating shear stress (OS), is pro-atherogenic and induces expression of DNMT1 both in vivo and in vitro. D-flow regulates genome-wide DNA methylation patterns in a DNMT-dependent manner. The DNMT inhibitor 5-Aza-2'deoxycytidine (5Aza) or DNMT1 siRNA reduces OS-induced endothelial inflammation. Moreover, 5Aza inhibits the development of atherosclerosis in ApoE(-/-) mice. Through a systems biological analysis of genome-wide DNA methylation patterns and gene expression data, we found 11 mechanosensitive genes which were suppressed by d-flow in vivo, experienced hypermethylation in their promoter region in response to d-flow, and were rescued by 5Aza treatment. Interestingly, among these mechanosensitive genes, the two transcription factors HoxA5 and Klf3 contain cAMP-response-elements (CRE), which may indicate that methylation of CRE sites could serve as a mechanosensitive master switch in gene expression. These findings provide new insight into the mechanism by which flow controls epigenetic DNA methylation patterns, which in turn alters endothelial gene expression, regulates vascular biology, and induces atherosclerosis. These novel findings have broad implications for understanding the biochemical mechanisms of atherogenesis and provide a basis for identifying potential therapeutic targets for atherosclerosis. This article is part of a Directed Issue entitled: Epigenetics dynamics in development and disease.
Collapse
Affiliation(s)
- Jessilyn Dunn
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, USA
| | - Rachel Simmons
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, USA
| | - Salim Thabet
- Division of Cardiology, Georgia Institute of Technology and Emory University, USA
| | - Hanjoong Jo
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, USA; Division of Cardiology, Georgia Institute of Technology and Emory University, USA.
| |
Collapse
|
34
|
Huveneers S, Daemen MJAP, Hordijk PL. Between Rho(k) and a hard place: the relation between vessel wall stiffness, endothelial contractility, and cardiovascular disease. Circ Res 2015; 116:895-908. [PMID: 25722443 DOI: 10.1161/circresaha.116.305720] [Citation(s) in RCA: 145] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Vascular stiffness is a mechanical property of the vessel wall that affects blood pressure, permeability, and inflammation. As a result, vascular stiffness is a key driver of (chronic) human disorders, including pulmonary arterial hypertension, kidney disease, and atherosclerosis. Responses of the endothelium to stiffening involve integration of mechanical cues from various sources, including the extracellular matrix, smooth muscle cells, and the forces that derive from shear stress of blood. This response in turn affects endothelial cell contractility, which is an important property that regulates endothelial stiffness, permeability, and leukocyte-vessel wall interactions. Moreover, endothelial stiffening reduces nitric oxide production, which promotes smooth muscle cell contraction and vasoconstriction. In fact, vessel wall stiffening, and microcirculatory endothelial dysfunction, precedes hypertension and thus underlies the development of vascular disease. Here, we review the cross talk among vessel wall stiffening, endothelial contractility, and vascular disease, which is controlled by Rho-driven actomyosin contractility and cellular mechanotransduction. In addition to discussing the various inputs and relevant molecular events in the endothelium, we address which actomyosin-regulated changes at cell adhesion complexes are genetically associated with human cardiovascular disease. Finally, we discuss recent findings that broaden therapeutic options for targeting this important mechanical signaling pathway in vascular pathogenesis.
Collapse
Affiliation(s)
- Stephan Huveneers
- From the Department of Molecular Cell Biology, Sanquin Research and Landsteiner Laboratory, Swammerdam Institute for Life Sciences (S.H., P.L.H.) and Department of Pathology (M.J.A.P.D.), Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands.
| | - Mat J A P Daemen
- From the Department of Molecular Cell Biology, Sanquin Research and Landsteiner Laboratory, Swammerdam Institute for Life Sciences (S.H., P.L.H.) and Department of Pathology (M.J.A.P.D.), Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Peter L Hordijk
- From the Department of Molecular Cell Biology, Sanquin Research and Landsteiner Laboratory, Swammerdam Institute for Life Sciences (S.H., P.L.H.) and Department of Pathology (M.J.A.P.D.), Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| |
Collapse
|
35
|
Chatterjee S, Fujiwara K, Pérez NG, Ushio-Fukai M, Fisher AB. Mechanosignaling in the vasculature: emerging concepts in sensing, transduction and physiological responses. Am J Physiol Heart Circ Physiol 2015; 308:H1451-62. [PMID: 25862828 DOI: 10.1152/ajpheart.00105.2015] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/19/2015] [Accepted: 03/23/2015] [Indexed: 01/08/2023]
Abstract
Cells are constantly exposed to mechanical forces that play a role in modulating cellular structure and function. The cardiovascular system experiences physical forces in the form of shear stress and stretch associated with blood flow and contraction, respectively. These forces are sensed by endothelial cells and cardiomyocytes and lead to responses that control vascular and cardiac homeostasis. This was highlighted at the Pan American Physiological Society meeting at Iguassu Falls, Brazil, in a symposium titled "Mechanosignaling in the Vasculature." This symposium presented recent research that showed the existence of a vital link between mechanosensing and downstream redox sensitive signaling cascades. This link helps to transduce and transmit the physical force into an observable physiological response. The speakers showcased how mechanosensors such as ion channels, membrane receptor kinases, adhesion molecules, and other cellular components transduce the force via redox signals (such as reactive oxygen species and nitric oxide) to receptors (transcription factors, growth factors, etc.). Receptor activated pathways then lead to cellular responses including cellular proliferation, contraction, and remodeling. These responses have major relevance to the physiology and pathophysiology of various cardiovascular diseases. Thus an understanding of the complex series of events, from the initial sensing through the final response, is essential for progress in this field. Overall, this symposium addressed some important emerging concepts in the field of mechanosignaling and the eventual pathophysiological responses.
Collapse
Affiliation(s)
- Shampa Chatterjee
- Institute for Environmental Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania;
| | - Keigi Fujiwara
- Department of Cardiology University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Néstor Gustavo Pérez
- Centro de Investigaciones Cardiovasculares, Universidad Nacional de La Plata, La Plata, Argentina; and
| | - Masuko Ushio-Fukai
- Department of Pharmacology, Center for Lung and Vascular Biology, University of Illinois at Chicago, Chicago, Illinois
| | - Aron B Fisher
- Institute for Environmental Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania
| |
Collapse
|
36
|
Lu S, Wang Y. Single-cell imaging of mechanotransduction in endothelial cells. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2014; 126:25-51. [PMID: 25081613 DOI: 10.1016/b978-0-12-394624-9.00002-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Endothelial cells (ECs) are constantly exposed to chemical and mechanical microenvironment in vivo. In mechanotransduction, cells can sense and translate the extracellular mechanical cues into intracellular biochemical signals, to regulate cellular processes. This regulation is crucial for many physiological functions, such as cell adhesion, migration, proliferation, and survival, as well as the progression of disease such as atherosclerosis. Here, we overview the current molecular understanding of mechanotransduction in ECs associated with atherosclerosis, especially those in response to physiological shear stress. The enabling technology of live-cell imaging has allowed the study of spatiotemporal molecular events and unprecedented understanding of intracellular signaling responses in mechanotransduction. Hence, we also introduce recent studies on mechanotransduction using single-cell imaging technologies.
Collapse
Affiliation(s)
- Shaoying Lu
- Department of Bioengineering, Institute of Engineering in Medicine, University of California, San Diego, La Jolla, California, USA
| | - Yingxiao Wang
- Department of Bioengineering, Institute of Engineering in Medicine, University of California, San Diego, La Jolla, California, USA
| |
Collapse
|
37
|
Mechanosensitive properties in the endothelium and their roles in the regulation of endothelial function. J Cardiovasc Pharmacol 2013; 61:461-70. [PMID: 23429585 DOI: 10.1097/fjc.0b013e31828c0933] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
: Vascular endothelial cells (ECs) line the luminal surface of blood vessels, which are exposed constantly to mechanical stimuli, such as fluid shear stress, cyclic strain, and blood pressure. In recent years, more and more evidence indicates that ECs sense these mechanical stimuli and subsequently convert mechanical stimuli into intracellular signals. The properties of ECs that sense the mechanical stimuli are defined as mechanosensors. There are a variety of mechanosensors that have been identified in ECs. These mechanosensors play an important role in regulating the function of the endothelium and vascular function, including blood pressure. This review focuses on the mechanosensors that have been identified in ECs and on the roles that mechanosensors play in the regulation of endothelium function, and in the regulation of vascular function.
Collapse
|
38
|
Lee HJ, Li N, Evans SM, Diaz MF, Wenzel PL. Biomechanical force in blood development: extrinsic physical cues drive pro-hematopoietic signaling. Differentiation 2013; 86:92-103. [PMID: 23850217 DOI: 10.1016/j.diff.2013.06.004] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2013] [Revised: 06/17/2013] [Accepted: 06/19/2013] [Indexed: 02/07/2023]
Abstract
The hematopoietic system is dynamic during development and in adulthood, undergoing countless spatial and temporal transitions during the course of one's life. Microenvironmental cues in the many unique hematopoietic niches differ, characterized by distinct soluble molecules, membrane-bound factors, and biophysical features that meet the changing needs of the blood system. Research from the last decade has revealed the importance of substrate elasticity and biomechanical force in determination of stem cell fate. Our understanding of the role of these factors in hematopoiesis is still relatively poor; however, the developmental origin of blood cells from the endothelium provides a model for comparison. Many endothelial mechanical sensors and second messenger systems may also determine hematopoietic stem cell fate, self renewal, and homing behaviors. Further, the intimate contact of hematopoietic cells with mechanosensitive cell types, including osteoblasts, endothelial cells, mesenchymal stem cells, and pericytes, places them in close proximity to paracrine signaling downstream of mechanical signals. The objective of this review is to present an overview of the sensors and intracellular signaling pathways activated by mechanical cues and highlight the role of mechanotransductive pathways in hematopoiesis.
Collapse
Affiliation(s)
- Hyun Jung Lee
- Children's Regenerative Medicine Program, Department of Pediatric Surgery, University of Texas Medical School at Houston, Houston, TX 77030, USA; Center for Stem Cell and Regenerative Medicine, The Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | | | | | | | | |
Collapse
|
39
|
Abstract
Blood vessels alter their morphology and function in response to changes in blood flow, and their responses are based on blood flow detection by the vascular endothelium. Endothelial cells (ECs) covering the inner surface of blood vessels sense shear stress generated by flowing blood and transmit the signal into the interior of the cell, which evokes a cellular response. The EC response to shear stress is closely linked to the regulation of vascular tone, blood coagulation and fibrinolysis, angiogenesis, and vascular remodelling, and it plays an important role in maintaining the homoeostasis of the circulatory system. Impairment of the EC response to shear stress leads to the development of vascular diseases such as hypertension, thrombosis, aneurysms, and atherosclerosis. Rapid progress has been made in elucidating shear stress mechanotransduction by using in vitro methods that apply controlled levels of shear stress to cultured ECs in fluid-dynamically designed flow-loading devices. The results have revealed that shear stress is converted into intracellular biochemical signals that are mediated by a variety of membrane molecules and microdomains, including ion channels, receptors, G-proteins, adhesion molecules, the cytoskeleton, caveolae, the glycocalyx, and primary cilia, and that multiple downstream signalling pathways become activated almost simultaneously. Nevertheless, neither the shear-stress-sensing mechanisms nor the sensor molecules that initially sense shear stress are yet known. Their identification would contribute to a better understanding of the pathophysiology of the vascular diseases that occur in a blood flow-dependent manner and to the development of new treatments for them.
Collapse
Affiliation(s)
- Joji Ando
- Laboratory of Biomedical Engineering, School of Medicine, Dokkyo Medical University, 880 Kita-kobayashi, Mibu, Tochigi 321-0293, Japan.
| | | |
Collapse
|
40
|
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: 5.4] [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
|
41
|
Morino-Koga S, Yano S, Kondo T, Shimauchi Y, Matsuyama S, Okamoto Y, Suico MA, Koga T, Sato T, Shuto T, Arima H, Wada I, Araki E, Kai H. Insulin receptor activation through its accumulation in lipid rafts by mild electrical stress. J Cell Physiol 2013; 228:439-46. [PMID: 22740366 DOI: 10.1002/jcp.24149] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Insulin resistance is due to the reduced cellular response to insulin in peripheral tissues. The interaction of insulin with its receptor is the first step in insulin action and thus the identified target of insulin resistance. It has been well established that defects or mutations in the insulin receptor (IR) cause insulin resistance. Therefore, an IR activator might be a novel therapeutic approach for insulin resistance. Our previous report showed that mild electrical stress (MES) enhanced the insulin-induced signaling pathway. However, the molecular mechanism of the effect of MES remains unclear. We assessed the effect of MES, which is characterized by low-intensity direct current, on insulin signaling in vitro and in vivo. Here, we showed that MES activated the insulin signaling in an insulin-independent manner and improved insulin resistance in peripheral tissues of high fat-fed mice. Moreover, we found that MES increased the localization of IR in lipid rafts and enhanced the level of phosphorylated Akt in insulin-resistant hepatic cells. Ablation of lipid rafts disrupted the effect of MES on Akt activation. Our findings indicate that MES has potential as an activator of IR in an insulin-independent manner, and might be beneficial for insulin resistance in type 2 diabetes.
Collapse
Affiliation(s)
- Saori Morino-Koga
- Department of Molecular Medicine, Global COE Cell Fate Regulation Research and Education Unit, Kumamoto University, Kumamoto, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Nakamizo H, Suzuki H, Miura S, Mogami S, Kishikawa H, Yoshida H, Matsui H, Hibi T. Transmural pressure loading enhances gastric mucosal cell proliferation. Dig Dis Sci 2012; 57:2545-2554. [PMID: 22644739 DOI: 10.1007/s10620-012-2208-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/06/2012] [Accepted: 04/25/2012] [Indexed: 01/25/2023]
Abstract
AIM Although increased intraluminal pressure in the stomach due to gastric outlet obstruction or functional gastric motor dysfunction, including gastroparesis, may affect gastric mucosal integrity, the direct effect of mechanical pressure on gastric mucosal cells has not yet been fully investigated. The aims of this study were to determine whether exposure to transmural pressure would affect the proliferation of gastric mucosal cells and to elucidate the intracellular signaling pathways involved. METHODS Cellular proliferation and DNA synthesis were evaluated in rat gastric epithelial cells exposed to high transmural pressures. The levels of activation of 3 MAP kinases, ERK, JNK, and p38, were assessed, and the induction of immediate early gene expression was examined. The activation of nuclear factor activator protein-1 (AP-1) was evaluated by an electrophoretic mobility shift assay. RESULTS Exposure to high transmural pressure significantly increased DNA synthesis within 24 h, with the most marked increase observed after exposure to a pressure of 80 mmHg, and this increase was inhibited by the MEK1 inhibitor PD98059. Early activation of ERK kinase, but not of JNK or p38 kinase, was detected after pressure loading. Early induction of the c-fos and c-myc genes and activation of the AP-1 transcription factor were also demonstrated within 3 h of exposure to 80 mmHg of pressure. CONCLUSION Gastric mucosal cell proliferation induced by exposure to high transmural pressure may be related to early activation of ERK, the induction of c-fos and c-myc, and the activation of AP-1.
Collapse
Affiliation(s)
- Hiromasa Nakamizo
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan
| | | | | | | | | | | | | | | |
Collapse
|
43
|
Jeong E, Lee HR, Pyee J, Park H. Pinosylvin Induces Cell Survival, Migration and Anti-Adhesiveness of Endothelial Cells via Nitric Oxide Production. Phytother Res 2012; 27:610-7. [DOI: 10.1002/ptr.4770] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2012] [Revised: 05/28/2012] [Accepted: 06/05/2012] [Indexed: 11/10/2022]
|
44
|
Yamamoto K, Ando J. New molecular mechanisms for cardiovascular disease:blood flow sensing mechanism in vascular endothelial cells. J Pharmacol Sci 2011; 116:323-31. [PMID: 21757846 DOI: 10.1254/jphs.10r29fm] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022] Open
Abstract
Endothelial cells (ECs) lining blood vessels have a variety of functions and play a critical role in the homeostasis of the circulatory system. It has become clear that biomechanical forces generated by blood flow regulate EC functions. ECs are in direct contact with blood flow and exposed to shear stress, a frictional force generated by flowing blood. A number of recent studies have revealed that ECs recognize changes in shear stress and transmit signals to the interior of the cell, which leads to cell responses that involve changes in cell morphology, cell function, and gene expression. These EC responses to shear stress are thought to play important roles in blood flow-dependent phenomena such as vascular tone control, angiogenesis, vascular remodeling, and atherogenesis. Much research has been done on shear stress sensing and signal transduction, and their molecular mechanisms are gradually becoming understood. However, much remains uncertain, and many candidates have been proposed for shear stress sensors. More extensive studies of vascular mechanobiology should increase our understanding of the molecular basis of the blood flow-mediated control of vascular functions.
Collapse
Affiliation(s)
- Kimiko Yamamoto
- Laboratory of System Physiology, Department of Biomedical Engineering, Graduate School of Medicine, University of Tokyo, Japan.
| | | |
Collapse
|
45
|
Wenger KH, El-Awady AR, Messer RLW, Sharawy MM, White G, Lapp CA. Pneumatic pressure bioreactor for cyclic hydrostatic stress application: mechanobiology effects on periodontal ligament cells. J Appl Physiol (1985) 2011; 111:1072-9. [PMID: 21757574 DOI: 10.1152/japplphysiol.01175.2010] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
A bioreactor system was developed to provide high-amplitude cyclic hydrostatic compressive stress (cHSC) using compressed air mixed commercially as needed to create partial pressures of oxygen and carbon dioxide appropriate for the cells under investigation. Operating pressures as high as 300 psi are achievable in this system at cyclic speeds of up to 0.2 Hz. In this study, ligamentous fibroblasts from human periodontal ligaments (n = 6) were compressed on two consecutive days at 150 psi for 3 h each day, and the mRNA for families of extracellular matrix protein and protease isoforms was evaluated by real-time PCR array. Several integrins were significantly upregulated, most notably alpha-3 (6.4-fold), as was SPG7 (12.1-fold). Among the collagens, Col8a1 was highly upregulated at 53.5-fold, with Col6a1, Col6a2, and Col7a1 also significantly upregulated 4.4- to 8.5-fold. MMP-1 was the most affected at 122.9-fold upregulation. MMP-14 likewise increased 17.8-fold with slight reductions for the gelatinases and a significant increase of TIMP-2 at 5.8-fold. The development of this bioreactor system and its utility in characterizing periodontal ligament fibroblast mechanobiology in intermediate-term testing hold promise for better simulating the conditions of the musculoskeletal system and the large cyclic compressive stresses joints may experience in gait, exertion, and mastication.
Collapse
Affiliation(s)
- Karl H Wenger
- Department of Orthopaedic Surgery, Georgia Health Sciences University, Augusta, GA 30912, USA.
| | | | | | | | | | | |
Collapse
|
46
|
Knox R, Chen E, Wang XJ, Jeffries B, Yamaguchi M, Kim S, Sumpio B. 17β-Estradiol exposure leads to activation of ERK and p38 but not JNK in vascular endothelial cells. Int J Angiol 2011. [DOI: 10.1007/s00547-004-1068-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022] Open
|
47
|
Goral V, Jin Y, Sun H, Ferrie AM, Wu Q, Fang Y. Agonist-directed desensitization of the β2-adrenergic receptor. PLoS One 2011; 6:e19282. [PMID: 21541288 PMCID: PMC3082563 DOI: 10.1371/journal.pone.0019282] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2011] [Accepted: 03/25/2011] [Indexed: 01/22/2023] Open
Abstract
The β(2)-adrenergic receptor (β(2)AR) agonists with reduced tachyphylaxis may offer new therapeutic agents with improved tolerance profile. However, receptor desensitization assays are often inferred at the single signaling molecule level, thus ligand-directed desensitization is poorly understood. Here we report a label-free biosensor whole cell assay with microfluidics to determine ligand-directed desensitization of the β(2)AR. Together with mechanistic deconvolution using small molecule inhibitors, the receptor desensitization and resensitization patterns under the short-term agonist exposure manifested the long-acting agonism of salmeterol, and differentiated the mechanisms of agonist-directed desensitization between a full agonist epinephrine and a partial agonist pindolol. This study reveals the cellular mechanisms of agonist-selective β(2)AR desensitization at the whole cell level.
Collapse
Affiliation(s)
- Vasiliy Goral
- Biochemical Technologies, Science and Technology Division, Corning Inc., Corning, New York, United States of America
| | - Yan Jin
- Biochemical Technologies, Science and Technology Division, Corning Inc., Corning, New York, United States of America
| | - Haiyan Sun
- Biochemical Technologies, Science and Technology Division, Corning Inc., Corning, New York, United States of America
| | - Ann M. Ferrie
- Biochemical Technologies, Science and Technology Division, Corning Inc., Corning, New York, United States of America
| | - Qi Wu
- Biochemical Technologies, Science and Technology Division, Corning Inc., Corning, New York, United States of America
| | - Ye Fang
- Biochemical Technologies, Science and Technology Division, Corning Inc., Corning, New York, United States of America
| |
Collapse
|
48
|
Goral V, Wu Q, Sun H, Fang Y. Label-free optical biosensor with microfluidics for sensing ligand-directed functional selectivity on trafficking of thrombin receptor. FEBS Lett 2011; 585:1054-60. [PMID: 21382370 DOI: 10.1016/j.febslet.2011.03.003] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2010] [Revised: 02/13/2011] [Accepted: 03/01/2011] [Indexed: 01/08/2023]
|
49
|
Jung WS, Cho J, In K, Kim J, Cho KH, Park JM, Moon SK, Kim KW, Park SU, Pyee J, Park SG, Jeong Y, Park H, Ko CN. Chunghyul-dan acts as an anti-inflammatory agent in endothelial cells by regulating gene expression. Anim Cells Syst (Seoul) 2010. [DOI: 10.1080/19768354.2010.525824] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022] Open
|
50
|
Flores D, Battini L, Gusella GL, Rohatgi R. Fluid shear stress induces renal epithelial gene expression through polycystin-2-dependent trafficking of extracellular regulated kinase. Nephron Clin Pract 2010; 117:p27-36. [PMID: 21109758 DOI: 10.1159/000321640] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2009] [Accepted: 09/20/2010] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND The cilium and cilial proteins have emerged as principal mechanosensors of renal epithelial cells responsible for translating mechanical forces into intracellular signals. Polycystin-2 (PC-2), a cilial protein, regulates flow/shear-induced changes in intracellular Ca(2+) ([Ca(2+)](i)) and recently has been implicated in the regulation of mitogen-activated protein (MAP) kinases. We hypothesize that fluid shear stress (FSS) activates PC-2 which regulates MAP kinase and, in turn, induces MAP kinase-dependent gene expression, specifically, monocyte chemoattractant protein-1 (MCP-1). METHODS To test this, PC-2 expression was constitutively reduced in a murine inner medullary collecting duct (IMCD3) cell line, and the expression of FSS-induced MCP-1 expression and MAP kinase signaling compared between the parental (PC-2-expressing) and PC-2-deficient IMCD3 cells. RESULTS FSS induces MAP kinase signaling and downstream MCP-1 mRNA expression in wild-type IMCD3 cells, while inhibitors of MAP kinase prevented the FSS-induced MCP-1 mRNA response. In contradistinction, FSS did not induce MCP-1 mRNA expression in PC-2-deficient cells, but did increase activation of the upstream MAP kinases. Wild-type cells exposed to FSS augmented the nuclear abundance of activated MAP kinase while PC-2-deficient cells did not. CONCLUSIONS PC-2 regulates FSS-induced MAP kinase trafficking into the nucleus of CD cells.
Collapse
Affiliation(s)
- Daniel Flores
- Department of Medicine, The Mount Sinai School of Medicine, New York, NY 10000209, USA
| | | | | | | |
Collapse
|