51
|
The critical role of SENP1-mediated GATA2 deSUMOylation in promoting endothelial activation in graft arteriosclerosis. Nat Commun 2017; 8:15426. [PMID: 28569748 PMCID: PMC5461500 DOI: 10.1038/ncomms15426] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2016] [Accepted: 03/29/2017] [Indexed: 02/07/2023] Open
Abstract
Data from clinical research and our previous study have suggested the potential involvement of SENP1, the major protease of post-translational SUMOylation, in cardiovascular disorders. Here, we investigate the role of SENP1-mediated SUMOylation in graft arteriosclerosis (GA), the major cause of allograft failure. We observe an endothelial-specific induction of SENP1 and GATA2 in clinical graft rejection specimens that show endothelial activation-mediated vascular remodelling. In mouse aorta transplantation GA models, endothelial-specific SENP1 knockout grafts demonstrate limited neointima formation with attenuated leukocyte recruitment, resulting from diminished induction of adhesion molecules in the graft endothelium due to increased GATA2 SUMOylation. Mechanistically, inflammation-induced SENP1 promotes the deSUMOylation of GATA2 and IκBα in endothelial cells, resulting in increased GATA2 stability, promoter-binding capability and NF-κB activity, which leads to augmented endothelial activation and inflammation. Therefore, upon inflammation, endothelial SENP1-mediated SUMOylation drives GA by regulating the synergistic effect of GATA2 and NF-κB and consequent endothelial dysfunction.
Collapse
|
52
|
The arterial microenvironment: the where and why of atherosclerosis. Biochem J 2017; 473:1281-95. [PMID: 27208212 DOI: 10.1042/bj20150844] [Citation(s) in RCA: 120] [Impact Index Per Article: 17.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2015] [Accepted: 02/15/2016] [Indexed: 12/11/2022]
Abstract
The formation of atherosclerotic plaques in the large and medium sized arteries is classically driven by systemic factors, such as elevated cholesterol and blood pressure. However, work over the past several decades has established that atherosclerotic plaque development involves a complex coordination of both systemic and local cues that ultimately determine where plaques form and how plaques progress. Although current therapeutics for atherosclerotic cardiovascular disease primarily target the systemic risk factors, a large array of studies suggest that the local microenvironment, including arterial mechanics, matrix remodelling and lipid deposition, plays a vital role in regulating the local susceptibility to plaque development through the regulation of vascular cell function. Additionally, these microenvironmental stimuli are capable of tuning other aspects of the microenvironment through collective adaptation. In this review, we will discuss the components of the arterial microenvironment, how these components cross-talk to shape the local microenvironment, and the effect of microenvironmental stimuli on vascular cell function during atherosclerotic plaque formation.
Collapse
|
53
|
Franck G, Mawson T, Sausen G, Salinas M, Masson GS, Cole A, Beltrami-Moreira M, Chatzizisis Y, Quillard T, Tesmenitsky Y, Shvartz E, Sukhova GK, Swirski FK, Nahrendorf M, Aikawa E, Croce KJ, Libby P. Flow Perturbation Mediates Neutrophil Recruitment and Potentiates Endothelial Injury via TLR2 in Mice: Implications for Superficial Erosion. Circ Res 2017; 121:31-42. [PMID: 28428204 DOI: 10.1161/circresaha.117.310694] [Citation(s) in RCA: 132] [Impact Index Per Article: 18.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/24/2017] [Revised: 04/10/2017] [Accepted: 04/20/2017] [Indexed: 01/25/2023]
Abstract
RATIONALE Superficial erosion currently causes up to a third of acute coronary syndromes; yet, we lack understanding of its mechanisms. Thrombi because of superficial intimal erosion characteristically complicate matrix-rich atheromata in regions of flow perturbation. OBJECTIVE This study tested in vivo the involvement of disturbed flow and of neutrophils, hyaluronan, and Toll-like receptor 2 ligation in superficial intimal injury, a process implicated in superficial erosion. METHODS AND RESULTS In mouse carotid arteries with established intimal lesions tailored to resemble the substrate of human eroded plaques, acute flow perturbation promoted downstream endothelial cell activation, neutrophil accumulation, endothelial cell death and desquamation, and mural thrombosis. Neutrophil loss-of-function limited these findings. Toll-like receptor 2 agonism activated luminal endothelial cells, and deficiency of this innate immune receptor decreased intimal neutrophil adherence in regions of local flow disturbance, reducing endothelial cell injury and local thrombosis (P<0.05). CONCLUSIONS These results implicate flow disturbance, neutrophils, and Toll-like receptor 2 signaling as mechanisms that contribute to superficial erosion, a cause of acute coronary syndrome of likely growing importance in the statin era.
Collapse
Affiliation(s)
- Grégory Franck
- From the Department of Cardiovascular Medicine (G.F., T.M., G.S., M.S., A.C., M.B.-M., Y.C., T.Q., Y.T., E.S., G.K.S., E.A., K.J.C., P.L.), and Center for Interdisciplinary Cardiovascular Sciences (E.A.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA; Center for Systems Biology, Massachusetts General Hospital, Harvard Medical School, Boston (G.S.M., F.K.S., M.N.); and Department of Engineering and Technology, College of Engineering and Computing, Nova Southeastern University, Fort Lauderdale, FL (M.S.)
| | - Thomas Mawson
- From the Department of Cardiovascular Medicine (G.F., T.M., G.S., M.S., A.C., M.B.-M., Y.C., T.Q., Y.T., E.S., G.K.S., E.A., K.J.C., P.L.), and Center for Interdisciplinary Cardiovascular Sciences (E.A.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA; Center for Systems Biology, Massachusetts General Hospital, Harvard Medical School, Boston (G.S.M., F.K.S., M.N.); and Department of Engineering and Technology, College of Engineering and Computing, Nova Southeastern University, Fort Lauderdale, FL (M.S.)
| | - Grasiele Sausen
- From the Department of Cardiovascular Medicine (G.F., T.M., G.S., M.S., A.C., M.B.-M., Y.C., T.Q., Y.T., E.S., G.K.S., E.A., K.J.C., P.L.), and Center for Interdisciplinary Cardiovascular Sciences (E.A.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA; Center for Systems Biology, Massachusetts General Hospital, Harvard Medical School, Boston (G.S.M., F.K.S., M.N.); and Department of Engineering and Technology, College of Engineering and Computing, Nova Southeastern University, Fort Lauderdale, FL (M.S.)
| | - Manuel Salinas
- From the Department of Cardiovascular Medicine (G.F., T.M., G.S., M.S., A.C., M.B.-M., Y.C., T.Q., Y.T., E.S., G.K.S., E.A., K.J.C., P.L.), and Center for Interdisciplinary Cardiovascular Sciences (E.A.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA; Center for Systems Biology, Massachusetts General Hospital, Harvard Medical School, Boston (G.S.M., F.K.S., M.N.); and Department of Engineering and Technology, College of Engineering and Computing, Nova Southeastern University, Fort Lauderdale, FL (M.S.)
| | - Gustavo Santos Masson
- From the Department of Cardiovascular Medicine (G.F., T.M., G.S., M.S., A.C., M.B.-M., Y.C., T.Q., Y.T., E.S., G.K.S., E.A., K.J.C., P.L.), and Center for Interdisciplinary Cardiovascular Sciences (E.A.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA; Center for Systems Biology, Massachusetts General Hospital, Harvard Medical School, Boston (G.S.M., F.K.S., M.N.); and Department of Engineering and Technology, College of Engineering and Computing, Nova Southeastern University, Fort Lauderdale, FL (M.S.)
| | - Andrew Cole
- From the Department of Cardiovascular Medicine (G.F., T.M., G.S., M.S., A.C., M.B.-M., Y.C., T.Q., Y.T., E.S., G.K.S., E.A., K.J.C., P.L.), and Center for Interdisciplinary Cardiovascular Sciences (E.A.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA; Center for Systems Biology, Massachusetts General Hospital, Harvard Medical School, Boston (G.S.M., F.K.S., M.N.); and Department of Engineering and Technology, College of Engineering and Computing, Nova Southeastern University, Fort Lauderdale, FL (M.S.)
| | - Marina Beltrami-Moreira
- From the Department of Cardiovascular Medicine (G.F., T.M., G.S., M.S., A.C., M.B.-M., Y.C., T.Q., Y.T., E.S., G.K.S., E.A., K.J.C., P.L.), and Center for Interdisciplinary Cardiovascular Sciences (E.A.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA; Center for Systems Biology, Massachusetts General Hospital, Harvard Medical School, Boston (G.S.M., F.K.S., M.N.); and Department of Engineering and Technology, College of Engineering and Computing, Nova Southeastern University, Fort Lauderdale, FL (M.S.)
| | - Yiannis Chatzizisis
- From the Department of Cardiovascular Medicine (G.F., T.M., G.S., M.S., A.C., M.B.-M., Y.C., T.Q., Y.T., E.S., G.K.S., E.A., K.J.C., P.L.), and Center for Interdisciplinary Cardiovascular Sciences (E.A.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA; Center for Systems Biology, Massachusetts General Hospital, Harvard Medical School, Boston (G.S.M., F.K.S., M.N.); and Department of Engineering and Technology, College of Engineering and Computing, Nova Southeastern University, Fort Lauderdale, FL (M.S.)
| | - Thibault Quillard
- From the Department of Cardiovascular Medicine (G.F., T.M., G.S., M.S., A.C., M.B.-M., Y.C., T.Q., Y.T., E.S., G.K.S., E.A., K.J.C., P.L.), and Center for Interdisciplinary Cardiovascular Sciences (E.A.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA; Center for Systems Biology, Massachusetts General Hospital, Harvard Medical School, Boston (G.S.M., F.K.S., M.N.); and Department of Engineering and Technology, College of Engineering and Computing, Nova Southeastern University, Fort Lauderdale, FL (M.S.)
| | - Yevgenia Tesmenitsky
- From the Department of Cardiovascular Medicine (G.F., T.M., G.S., M.S., A.C., M.B.-M., Y.C., T.Q., Y.T., E.S., G.K.S., E.A., K.J.C., P.L.), and Center for Interdisciplinary Cardiovascular Sciences (E.A.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA; Center for Systems Biology, Massachusetts General Hospital, Harvard Medical School, Boston (G.S.M., F.K.S., M.N.); and Department of Engineering and Technology, College of Engineering and Computing, Nova Southeastern University, Fort Lauderdale, FL (M.S.)
| | - Eugenia Shvartz
- From the Department of Cardiovascular Medicine (G.F., T.M., G.S., M.S., A.C., M.B.-M., Y.C., T.Q., Y.T., E.S., G.K.S., E.A., K.J.C., P.L.), and Center for Interdisciplinary Cardiovascular Sciences (E.A.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA; Center for Systems Biology, Massachusetts General Hospital, Harvard Medical School, Boston (G.S.M., F.K.S., M.N.); and Department of Engineering and Technology, College of Engineering and Computing, Nova Southeastern University, Fort Lauderdale, FL (M.S.)
| | - Galina K Sukhova
- From the Department of Cardiovascular Medicine (G.F., T.M., G.S., M.S., A.C., M.B.-M., Y.C., T.Q., Y.T., E.S., G.K.S., E.A., K.J.C., P.L.), and Center for Interdisciplinary Cardiovascular Sciences (E.A.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA; Center for Systems Biology, Massachusetts General Hospital, Harvard Medical School, Boston (G.S.M., F.K.S., M.N.); and Department of Engineering and Technology, College of Engineering and Computing, Nova Southeastern University, Fort Lauderdale, FL (M.S.)
| | - Filip K Swirski
- From the Department of Cardiovascular Medicine (G.F., T.M., G.S., M.S., A.C., M.B.-M., Y.C., T.Q., Y.T., E.S., G.K.S., E.A., K.J.C., P.L.), and Center for Interdisciplinary Cardiovascular Sciences (E.A.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA; Center for Systems Biology, Massachusetts General Hospital, Harvard Medical School, Boston (G.S.M., F.K.S., M.N.); and Department of Engineering and Technology, College of Engineering and Computing, Nova Southeastern University, Fort Lauderdale, FL (M.S.)
| | - Matthias Nahrendorf
- From the Department of Cardiovascular Medicine (G.F., T.M., G.S., M.S., A.C., M.B.-M., Y.C., T.Q., Y.T., E.S., G.K.S., E.A., K.J.C., P.L.), and Center for Interdisciplinary Cardiovascular Sciences (E.A.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA; Center for Systems Biology, Massachusetts General Hospital, Harvard Medical School, Boston (G.S.M., F.K.S., M.N.); and Department of Engineering and Technology, College of Engineering and Computing, Nova Southeastern University, Fort Lauderdale, FL (M.S.)
| | - Elena Aikawa
- From the Department of Cardiovascular Medicine (G.F., T.M., G.S., M.S., A.C., M.B.-M., Y.C., T.Q., Y.T., E.S., G.K.S., E.A., K.J.C., P.L.), and Center for Interdisciplinary Cardiovascular Sciences (E.A.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA; Center for Systems Biology, Massachusetts General Hospital, Harvard Medical School, Boston (G.S.M., F.K.S., M.N.); and Department of Engineering and Technology, College of Engineering and Computing, Nova Southeastern University, Fort Lauderdale, FL (M.S.)
| | - Kevin J Croce
- From the Department of Cardiovascular Medicine (G.F., T.M., G.S., M.S., A.C., M.B.-M., Y.C., T.Q., Y.T., E.S., G.K.S., E.A., K.J.C., P.L.), and Center for Interdisciplinary Cardiovascular Sciences (E.A.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA; Center for Systems Biology, Massachusetts General Hospital, Harvard Medical School, Boston (G.S.M., F.K.S., M.N.); and Department of Engineering and Technology, College of Engineering and Computing, Nova Southeastern University, Fort Lauderdale, FL (M.S.)
| | - Peter Libby
- From the Department of Cardiovascular Medicine (G.F., T.M., G.S., M.S., A.C., M.B.-M., Y.C., T.Q., Y.T., E.S., G.K.S., E.A., K.J.C., P.L.), and Center for Interdisciplinary Cardiovascular Sciences (E.A.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA; Center for Systems Biology, Massachusetts General Hospital, Harvard Medical School, Boston (G.S.M., F.K.S., M.N.); and Department of Engineering and Technology, College of Engineering and Computing, Nova Southeastern University, Fort Lauderdale, FL (M.S.).
| |
Collapse
|
54
|
Chistiakov DA, Orekhov AN, Bobryshev YV. Effects of shear stress on endothelial cells: go with the flow. Acta Physiol (Oxf) 2017; 219:382-408. [PMID: 27246807 DOI: 10.1111/apha.12725] [Citation(s) in RCA: 270] [Impact Index Per Article: 38.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2016] [Revised: 02/17/2016] [Accepted: 05/30/2016] [Indexed: 12/11/2022]
Abstract
Haemodynamic forces influence the functional properties of vascular endothelium. Endothelial cells (ECs) have a variety of receptors, which sense flow and transmit mechanical signals through mechanosensitive signalling pathways to recipient molecules that lead to phenotypic and functional changes. Arterial architecture varies greatly exhibiting bifurcations, branch points and curved regions, which are exposed to various flow patterns. Clinical studies showed that atherosclerotic plaques develop preferentially at arterial branches and curvatures, that is in the regions exposed to disturbed flow and shear stress. In the atheroprone regions, the endothelium has a proinflammatory phenotype associated with low nitric oxide production, reduced barrier function and increased proadhesive, procoagulant and proproliferative properties. Atheroresistant regions are exposed to laminar flow and high shear stress that induce prosurvival antioxidant signals and maintain the quiescent phenotype in ECs. Indeed, various flow patterns contribute to phenotypic and functional heterogeneity of arterial endothelium whose response to proatherogenic stimuli is differentiated. This may explain the preferential development of endothelial dysfunction in arterial sites with disturbed flow.
Collapse
Affiliation(s)
- D. A. Chistiakov
- Department of Medical Nanobiotechnology; Pirogov Russian State Medical University; Moscow Russia
| | - A. N. Orekhov
- Institute of General Pathology and Pathophysiology; Russian Academy of Medical Sciences; Moscow Russia
- Institute for Atherosclerosis Research; Skolkovo Innovative Center; Moscow Russia
- Department of Biophysics; Biological Faculty; Moscow State University; Moscow Russia
| | - Y. V. Bobryshev
- Institute of General Pathology and Pathophysiology; Russian Academy of Medical Sciences; Moscow Russia
- Faculty of Medicine and St Vincent's Centre for Applied Medical Research; University of New South Wales; Sydney NSW Australia
- School of Medicine; University of Western Sydney; Campbelltown NSW Australia
| |
Collapse
|
55
|
Le NT, Martin JF, Fujiwara K, Abe JI. Sub-cellular localization specific SUMOylation in the heart. Biochim Biophys Acta Mol Basis Dis 2017; 1863:2041-2055. [PMID: 28130202 DOI: 10.1016/j.bbadis.2017.01.018] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2016] [Revised: 11/21/2016] [Accepted: 01/09/2017] [Indexed: 12/27/2022]
Abstract
Although the majority of SUMO substrates are localized in the nucleus, SUMOylation is not limited to nuclear proteins and can be also detected in extra-nuclear proteins. In this review, we will highlight and discuss how SUMOylation in different cellular compartments regulate biological processes. First, we will discuss the key role of SUMOylation of proteins in the extra-nuclear compartment in cardiomyocytes, which is overwhelmingly cardio-protective. On the other hand, SUMOylation of nuclear proteins is generally detrimental to the cardiac function mainly because of the trans-repressive nature of SUMOylation on many transcription factors. We will also discuss the potential role of SUMOylation in epigenetic regulation. In this review, we will propose a new concept that shuttling of SUMO proteases between the nuclear and extra-nuclear compartments without changing their enzymatic activity regulates the extent of SUMOylation in these compartments and determines the response and fate of cardiomyocytes after cardiac insults. Approaches focused specifically to inhibit this shuttling in cardiomyocytes will be necessary to understand the whole picture of SUMOylation and its pathophysiological consequences in the heart, especially after cardiac insults. This article is part of a Special Issue entitled: Genetic and epigenetic control of heart failure - edited by Jun Ren & Megan Yingmei Zhang.
Collapse
Affiliation(s)
- Nhat-Tu Le
- Department of Cardiology - Research, Division of Internal Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - James F Martin
- Department of Molecular Physiology and Biophysics, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| | - Keigi Fujiwara
- Department of Cardiology - Research, Division of Internal Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Jun-Ichi Abe
- Department of Cardiology - Research, Division of Internal Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| |
Collapse
|
56
|
Abe JI, Sandhu UG, Hoang NM, Thangam M, Quintana-Quezada RA, Fujiwara K, Le NT. Coordination of Cellular Localization-Dependent Effects of Sumoylation in Regulating Cardiovascular and Neurological Diseases. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 963:337-358. [PMID: 28197922 PMCID: PMC5716632 DOI: 10.1007/978-3-319-50044-7_20] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Sumoylation, a reversible post-transcriptional modification process, of proteins are involved in cellular differentiation, growth, and even motility by regulating various protein functions. Sumoylation is not limited to cytosolic proteins as recent evidence shows that nuclear proteins, those associated with membranes, and mitochondrial proteins are also sumoylated. Moreover, it is now known that sumoylation plays an important role in the process of major human ailments such as malignant, cardiovascular and neurological diseases. In this chapter, we will highlight and discuss how the localization of SUMO protease and SUMO E3 ligase in different compartments within a cell regulates biological processes that depend on sumoylation. First, we will discuss the key role of sumoylation in the nucleus, which leads to the development of endothelial dysfunction and atherosclerosis . We will then discuss how sumoylation of plasma membrane potassium channel proteins are involved in epilepsy and arrhythmia. Mitochondrial proteins are known to be also sumoylated, and the importance of dynamic-related protein 1 (DRP1) sumoylation on mitochondrial function will be discussed. As we will emphasize throughout this review, sumoylation plays crucial roles in different cellular compartments, which is coordinately regulated by the translocation of various SUMO proteases and SUMO E3 ligase. Comprehensive approach will be necessary to understand the molecular mechanism for efficiently moving around various enzymes that regulate sumoylation within cells.
Collapse
Affiliation(s)
- Jun-Ichi Abe
- Department of Cardiology - Research, Division of Internal Medicine, The University of Texas MD Anderson Cancer Center, 2121 W. Holcombe Blvd, Unit Number: 1101, Room Number: IBT8.803E, Houston, TX, 77030, USA.
| | - Uday G Sandhu
- Department of Cardiology - Research, Division of Internal Medicine, The University of Texas MD Anderson Cancer Center, 2121 W. Holcombe Blvd, Unit Number: 1101, Room Number: IBT8.803E, Houston, TX, 77030, USA
| | - Nguyet Minh Hoang
- Department of Cardiology - Research, Division of Internal Medicine, The University of Texas MD Anderson Cancer Center, 2121 W. Holcombe Blvd, Unit Number: 1101, Room Number: IBT8.803E, Houston, TX, 77030, USA
| | - Manoj Thangam
- Department of Cardiology - Research, Division of Internal Medicine, The University of Texas MD Anderson Cancer Center, 2121 W. Holcombe Blvd, Unit Number: 1101, Room Number: IBT8.803E, Houston, TX, 77030, USA
| | - Raymundo A Quintana-Quezada
- Department of Cardiology - Research, Division of Internal Medicine, The University of Texas MD Anderson Cancer Center, 2121 W. Holcombe Blvd, Unit Number: 1101, Room Number: IBT8.803E, Houston, TX, 77030, USA
| | - Keigi Fujiwara
- Department of Cardiology - Research, Division of Internal Medicine, The University of Texas MD Anderson Cancer Center, 2121 W. Holcombe Blvd, Unit Number: 1101, Room Number: IBT8.803E, Houston, TX, 77030, USA
| | - Nhat Tu Le
- Department of Cardiology - Research, Division of Internal Medicine, The University of Texas MD Anderson Cancer Center, 2121 W. Holcombe Blvd, Unit Number: 1101, Room Number: IBT8.803E, Houston, TX, 77030, USA
| |
Collapse
|
57
|
Flow signaling and atherosclerosis. Cell Mol Life Sci 2016; 74:1835-1858. [PMID: 28039525 PMCID: PMC5391278 DOI: 10.1007/s00018-016-2442-4] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2016] [Revised: 12/12/2016] [Accepted: 12/15/2016] [Indexed: 12/26/2022]
Abstract
Atherosclerosis rarely develops in the region of arteries exposed to undisturbed flow (u-flow, unidirectional flow). Instead, atherogenesis occurs in the area exposed to disturbed flow (d-flow, multidirectional flow). Based on these general pathohistological observations, u-flow is considered to be athero-protective, while d-flow is atherogenic. The fact that u-flow and d-flow induce such clearly different biological responses in the wall of large arteries indicates that these two types of flow activate each distinct intracellular signaling cascade in vascular endothelial cells (ECs), which are directly exposed to blood flow. The ability of ECs to differentially respond to the two types of flow provides an opportunity to identify molecular events that lead to endothelial dysfunction and atherosclerosis. In this review, we will focus on various molecular events, which are differentially regulated by these two flow types. We will discuss how various kinases, ER stress, inflammasome, SUMOylation, and DNA methylation play roles in the differential flow response, endothelial dysfunction, and atherosclerosis. We will also discuss the interplay among the molecular events and how they coordinately regulate flow-dependent signaling and cellular responses. It is hoped that clear understanding of the way how the two flow types beget each unique phenotype in ECs will lead us to possible points of intervention against endothelial dysfunction and cardiovascular diseases.
Collapse
|
58
|
Sekar TV, Foygel K, Devulapally R, Kumar V, Malhotra S, Massoud TF, Paulmurugan R. Molecular Imaging Biosensor Monitors p53 Sumoylation in Cells and Living Mice. Anal Chem 2016; 88:11420-11428. [PMID: 27934110 DOI: 10.1021/acs.analchem.6b02048] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Small molecule mediated stabilization of p53 tumor suppressor protein through sumoylation is a promising new strategy for improving cancer chemotherapy. A molecular tool that monitors p53 sumoylation status and expedites screening for drugs that enhance p53 sumoylation would be beneficial. We report a molecularly engineered reporter fragment complementation biosensor based on optical imaging of Firefly luciferase (FLuc), to quantitatively image p53 sumoylation and desumoylation in cells and living mice. We initially characterized this biosensor by successfully imaging sumoylation of several target proteins, achieving significant FLuc complementation for ERα (p < 0.01), p53 (p < 0.005), FKBP12 (p < 0.03), ID (p < 0.03), and HDAC1 (p < 0.002). We then rigorously tested the sensitivity and specificity of the biosensor using several variants of p53 and SUMO1, including deletion mutants, and those with modified sequences containing the SUMO-acceptor site of target proteins. Next we evaluated the performance of the biosensor in HepG2 cells by treatment with ginkgolic acid, a drug that reduces p53 sumoylation, as well as trichostatin A, a potential inducer of p53 sumoylation by enhancement of its nuclear export. Lastly, we demonstrated the in vivo utility of this biosensor in monitoring and quantifying the effects of these drugs on p53 sumoylation in living mice using bioluminescence imaging. Adoption of this biosensor in future high throughput drug screening has the important potential to help identify new and repurposed small molecules that alter p53 sumoylation, and to preclinically evaluate candidate anticancer drugs in living animals.
Collapse
Affiliation(s)
- Thillai V Sekar
- Molecular Imaging Program at Stanford, Bio-X Program, Stanford University School of Medicine , Palo Alto, California 94304, United States
| | - Kira Foygel
- Molecular Imaging Program at Stanford, Bio-X Program, Stanford University School of Medicine , Palo Alto, California 94304, United States
| | - Rammohan Devulapally
- Molecular Imaging Program at Stanford, Bio-X Program, Stanford University School of Medicine , Palo Alto, California 94304, United States
| | - Vineet Kumar
- Radiation Oncology, Stanford University School of Medicine , Stanford, California 94305, United States
| | - Sanjay Malhotra
- Radiation Oncology, Stanford University School of Medicine , Stanford, California 94305, United States
| | - Tarik F Massoud
- Molecular Imaging Program at Stanford, Bio-X Program, Stanford University School of Medicine , Palo Alto, California 94304, United States
| | - Ramasamy Paulmurugan
- Molecular Imaging Program at Stanford, Bio-X Program, Stanford University School of Medicine , Palo Alto, California 94304, United States
| |
Collapse
|
59
|
Serbanovic-Canic J, de Luca A, Warboys C, Ferreira PF, Luong LA, Hsiao S, Gauci I, Mahmoud M, Feng S, Souilhol C, Bowden N, Ashton JP, Walczak H, Firmin D, Krams R, Mason JC, Haskard DO, Sherwin S, Ridger V, Chico TJA, Evans PC. Zebrafish Model for Functional Screening of Flow-Responsive Genes. Arterioscler Thromb Vasc Biol 2016; 37:130-143. [PMID: 27834691 PMCID: PMC5172514 DOI: 10.1161/atvbaha.116.308502] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2016] [Accepted: 10/23/2016] [Indexed: 12/22/2022]
Abstract
Supplemental Digital Content is available in the text. Objective— Atherosclerosis is initiated at branches and bends of arteries exposed to disturbed blood flow that generates low shear stress. This mechanical environment promotes lesions by inducing endothelial cell (EC) apoptosis and dysfunction via mechanisms that are incompletely understood. Although transcriptome-based studies have identified multiple shear-responsive genes, most of them have an unknown function. To address this, we investigated whether zebrafish embryos can be used for functional screening of mechanosensitive genes that regulate EC apoptosis in mammalian arteries. Approach and Results— First, we demonstrated that flow regulates EC apoptosis in developing zebrafish vasculature. Specifically, suppression of blood flow in zebrafish embryos (by targeting cardiac troponin) enhanced that rate of EC apoptosis (≈10%) compared with controls exposed to flow (≈1%). A panel of candidate regulators of apoptosis were identified by transcriptome profiling of ECs from high and low shear stress regions of the porcine aorta. Genes that displayed the greatest differential expression and possessed 1 to 2 zebrafish orthologues were screened for the regulation of apoptosis in zebrafish vasculature exposed to flow or no-flow conditions using a knockdown approach. A phenotypic change was observed in 4 genes; p53-related protein (PERP) and programmed cell death 2–like protein functioned as positive regulators of apoptosis, whereas angiopoietin-like 4 and cadherin 13 were negative regulators. The regulation of perp, cdh13, angptl4, and pdcd2l by shear stress and the effects of perp and cdh13 on EC apoptosis were confirmed by studies of cultured EC exposed to flow. Conclusions— We conclude that a zebrafish model of flow manipulation coupled to gene knockdown can be used for functional screening of mechanosensitive genes in vascular ECs, thus providing potential therapeutic targets to prevent or treat endothelial injury at atheroprone sites.
Collapse
Affiliation(s)
- Jovana Serbanovic-Canic
- From the Department of Infection, Immunity and Cardiovascular Disease (J.S.-C., L.A.L., S.H., I.G., M.M., S.F., C.S., N.B., J.-P.A., V.R., T.J.A.C., P.C.E.), INSIGNEO Institute for In Silico Medicine (J.S.-C., V.R., T.J.A.C., P.C.E.), and the Bateson Centre (J.S.-C., J.-P.A., T.J.A.C., P.C.E.), University of Sheffield, United Kingdom; and Departments of Cardiovascular Science (A.d.L., C.W., J.C.M., D.O.H.), Imaging (P.F.F., D.F.), Bioengineering (R.K.), and Aeronautics (S.S.) Imperial College London, United Kingdom; and Cancer Institute, Faculty of Medical Sciences (H.W.), University College London, United Kingdom
| | - Amalia de Luca
- From the Department of Infection, Immunity and Cardiovascular Disease (J.S.-C., L.A.L., S.H., I.G., M.M., S.F., C.S., N.B., J.-P.A., V.R., T.J.A.C., P.C.E.), INSIGNEO Institute for In Silico Medicine (J.S.-C., V.R., T.J.A.C., P.C.E.), and the Bateson Centre (J.S.-C., J.-P.A., T.J.A.C., P.C.E.), University of Sheffield, United Kingdom; and Departments of Cardiovascular Science (A.d.L., C.W., J.C.M., D.O.H.), Imaging (P.F.F., D.F.), Bioengineering (R.K.), and Aeronautics (S.S.) Imperial College London, United Kingdom; and Cancer Institute, Faculty of Medical Sciences (H.W.), University College London, United Kingdom
| | - Christina Warboys
- From the Department of Infection, Immunity and Cardiovascular Disease (J.S.-C., L.A.L., S.H., I.G., M.M., S.F., C.S., N.B., J.-P.A., V.R., T.J.A.C., P.C.E.), INSIGNEO Institute for In Silico Medicine (J.S.-C., V.R., T.J.A.C., P.C.E.), and the Bateson Centre (J.S.-C., J.-P.A., T.J.A.C., P.C.E.), University of Sheffield, United Kingdom; and Departments of Cardiovascular Science (A.d.L., C.W., J.C.M., D.O.H.), Imaging (P.F.F., D.F.), Bioengineering (R.K.), and Aeronautics (S.S.) Imperial College London, United Kingdom; and Cancer Institute, Faculty of Medical Sciences (H.W.), University College London, United Kingdom
| | - Pedro F Ferreira
- From the Department of Infection, Immunity and Cardiovascular Disease (J.S.-C., L.A.L., S.H., I.G., M.M., S.F., C.S., N.B., J.-P.A., V.R., T.J.A.C., P.C.E.), INSIGNEO Institute for In Silico Medicine (J.S.-C., V.R., T.J.A.C., P.C.E.), and the Bateson Centre (J.S.-C., J.-P.A., T.J.A.C., P.C.E.), University of Sheffield, United Kingdom; and Departments of Cardiovascular Science (A.d.L., C.W., J.C.M., D.O.H.), Imaging (P.F.F., D.F.), Bioengineering (R.K.), and Aeronautics (S.S.) Imperial College London, United Kingdom; and Cancer Institute, Faculty of Medical Sciences (H.W.), University College London, United Kingdom
| | - Le A Luong
- From the Department of Infection, Immunity and Cardiovascular Disease (J.S.-C., L.A.L., S.H., I.G., M.M., S.F., C.S., N.B., J.-P.A., V.R., T.J.A.C., P.C.E.), INSIGNEO Institute for In Silico Medicine (J.S.-C., V.R., T.J.A.C., P.C.E.), and the Bateson Centre (J.S.-C., J.-P.A., T.J.A.C., P.C.E.), University of Sheffield, United Kingdom; and Departments of Cardiovascular Science (A.d.L., C.W., J.C.M., D.O.H.), Imaging (P.F.F., D.F.), Bioengineering (R.K.), and Aeronautics (S.S.) Imperial College London, United Kingdom; and Cancer Institute, Faculty of Medical Sciences (H.W.), University College London, United Kingdom
| | - Sarah Hsiao
- From the Department of Infection, Immunity and Cardiovascular Disease (J.S.-C., L.A.L., S.H., I.G., M.M., S.F., C.S., N.B., J.-P.A., V.R., T.J.A.C., P.C.E.), INSIGNEO Institute for In Silico Medicine (J.S.-C., V.R., T.J.A.C., P.C.E.), and the Bateson Centre (J.S.-C., J.-P.A., T.J.A.C., P.C.E.), University of Sheffield, United Kingdom; and Departments of Cardiovascular Science (A.d.L., C.W., J.C.M., D.O.H.), Imaging (P.F.F., D.F.), Bioengineering (R.K.), and Aeronautics (S.S.) Imperial College London, United Kingdom; and Cancer Institute, Faculty of Medical Sciences (H.W.), University College London, United Kingdom
| | - Ismael Gauci
- From the Department of Infection, Immunity and Cardiovascular Disease (J.S.-C., L.A.L., S.H., I.G., M.M., S.F., C.S., N.B., J.-P.A., V.R., T.J.A.C., P.C.E.), INSIGNEO Institute for In Silico Medicine (J.S.-C., V.R., T.J.A.C., P.C.E.), and the Bateson Centre (J.S.-C., J.-P.A., T.J.A.C., P.C.E.), University of Sheffield, United Kingdom; and Departments of Cardiovascular Science (A.d.L., C.W., J.C.M., D.O.H.), Imaging (P.F.F., D.F.), Bioengineering (R.K.), and Aeronautics (S.S.) Imperial College London, United Kingdom; and Cancer Institute, Faculty of Medical Sciences (H.W.), University College London, United Kingdom
| | - Marwa Mahmoud
- From the Department of Infection, Immunity and Cardiovascular Disease (J.S.-C., L.A.L., S.H., I.G., M.M., S.F., C.S., N.B., J.-P.A., V.R., T.J.A.C., P.C.E.), INSIGNEO Institute for In Silico Medicine (J.S.-C., V.R., T.J.A.C., P.C.E.), and the Bateson Centre (J.S.-C., J.-P.A., T.J.A.C., P.C.E.), University of Sheffield, United Kingdom; and Departments of Cardiovascular Science (A.d.L., C.W., J.C.M., D.O.H.), Imaging (P.F.F., D.F.), Bioengineering (R.K.), and Aeronautics (S.S.) Imperial College London, United Kingdom; and Cancer Institute, Faculty of Medical Sciences (H.W.), University College London, United Kingdom
| | - Shuang Feng
- From the Department of Infection, Immunity and Cardiovascular Disease (J.S.-C., L.A.L., S.H., I.G., M.M., S.F., C.S., N.B., J.-P.A., V.R., T.J.A.C., P.C.E.), INSIGNEO Institute for In Silico Medicine (J.S.-C., V.R., T.J.A.C., P.C.E.), and the Bateson Centre (J.S.-C., J.-P.A., T.J.A.C., P.C.E.), University of Sheffield, United Kingdom; and Departments of Cardiovascular Science (A.d.L., C.W., J.C.M., D.O.H.), Imaging (P.F.F., D.F.), Bioengineering (R.K.), and Aeronautics (S.S.) Imperial College London, United Kingdom; and Cancer Institute, Faculty of Medical Sciences (H.W.), University College London, United Kingdom
| | - Celine Souilhol
- From the Department of Infection, Immunity and Cardiovascular Disease (J.S.-C., L.A.L., S.H., I.G., M.M., S.F., C.S., N.B., J.-P.A., V.R., T.J.A.C., P.C.E.), INSIGNEO Institute for In Silico Medicine (J.S.-C., V.R., T.J.A.C., P.C.E.), and the Bateson Centre (J.S.-C., J.-P.A., T.J.A.C., P.C.E.), University of Sheffield, United Kingdom; and Departments of Cardiovascular Science (A.d.L., C.W., J.C.M., D.O.H.), Imaging (P.F.F., D.F.), Bioengineering (R.K.), and Aeronautics (S.S.) Imperial College London, United Kingdom; and Cancer Institute, Faculty of Medical Sciences (H.W.), University College London, United Kingdom
| | - Neil Bowden
- From the Department of Infection, Immunity and Cardiovascular Disease (J.S.-C., L.A.L., S.H., I.G., M.M., S.F., C.S., N.B., J.-P.A., V.R., T.J.A.C., P.C.E.), INSIGNEO Institute for In Silico Medicine (J.S.-C., V.R., T.J.A.C., P.C.E.), and the Bateson Centre (J.S.-C., J.-P.A., T.J.A.C., P.C.E.), University of Sheffield, United Kingdom; and Departments of Cardiovascular Science (A.d.L., C.W., J.C.M., D.O.H.), Imaging (P.F.F., D.F.), Bioengineering (R.K.), and Aeronautics (S.S.) Imperial College London, United Kingdom; and Cancer Institute, Faculty of Medical Sciences (H.W.), University College London, United Kingdom
| | - John-Paul Ashton
- From the Department of Infection, Immunity and Cardiovascular Disease (J.S.-C., L.A.L., S.H., I.G., M.M., S.F., C.S., N.B., J.-P.A., V.R., T.J.A.C., P.C.E.), INSIGNEO Institute for In Silico Medicine (J.S.-C., V.R., T.J.A.C., P.C.E.), and the Bateson Centre (J.S.-C., J.-P.A., T.J.A.C., P.C.E.), University of Sheffield, United Kingdom; and Departments of Cardiovascular Science (A.d.L., C.W., J.C.M., D.O.H.), Imaging (P.F.F., D.F.), Bioengineering (R.K.), and Aeronautics (S.S.) Imperial College London, United Kingdom; and Cancer Institute, Faculty of Medical Sciences (H.W.), University College London, United Kingdom
| | - Henning Walczak
- From the Department of Infection, Immunity and Cardiovascular Disease (J.S.-C., L.A.L., S.H., I.G., M.M., S.F., C.S., N.B., J.-P.A., V.R., T.J.A.C., P.C.E.), INSIGNEO Institute for In Silico Medicine (J.S.-C., V.R., T.J.A.C., P.C.E.), and the Bateson Centre (J.S.-C., J.-P.A., T.J.A.C., P.C.E.), University of Sheffield, United Kingdom; and Departments of Cardiovascular Science (A.d.L., C.W., J.C.M., D.O.H.), Imaging (P.F.F., D.F.), Bioengineering (R.K.), and Aeronautics (S.S.) Imperial College London, United Kingdom; and Cancer Institute, Faculty of Medical Sciences (H.W.), University College London, United Kingdom
| | - David Firmin
- From the Department of Infection, Immunity and Cardiovascular Disease (J.S.-C., L.A.L., S.H., I.G., M.M., S.F., C.S., N.B., J.-P.A., V.R., T.J.A.C., P.C.E.), INSIGNEO Institute for In Silico Medicine (J.S.-C., V.R., T.J.A.C., P.C.E.), and the Bateson Centre (J.S.-C., J.-P.A., T.J.A.C., P.C.E.), University of Sheffield, United Kingdom; and Departments of Cardiovascular Science (A.d.L., C.W., J.C.M., D.O.H.), Imaging (P.F.F., D.F.), Bioengineering (R.K.), and Aeronautics (S.S.) Imperial College London, United Kingdom; and Cancer Institute, Faculty of Medical Sciences (H.W.), University College London, United Kingdom
| | - Rob Krams
- From the Department of Infection, Immunity and Cardiovascular Disease (J.S.-C., L.A.L., S.H., I.G., M.M., S.F., C.S., N.B., J.-P.A., V.R., T.J.A.C., P.C.E.), INSIGNEO Institute for In Silico Medicine (J.S.-C., V.R., T.J.A.C., P.C.E.), and the Bateson Centre (J.S.-C., J.-P.A., T.J.A.C., P.C.E.), University of Sheffield, United Kingdom; and Departments of Cardiovascular Science (A.d.L., C.W., J.C.M., D.O.H.), Imaging (P.F.F., D.F.), Bioengineering (R.K.), and Aeronautics (S.S.) Imperial College London, United Kingdom; and Cancer Institute, Faculty of Medical Sciences (H.W.), University College London, United Kingdom
| | - Justin C Mason
- From the Department of Infection, Immunity and Cardiovascular Disease (J.S.-C., L.A.L., S.H., I.G., M.M., S.F., C.S., N.B., J.-P.A., V.R., T.J.A.C., P.C.E.), INSIGNEO Institute for In Silico Medicine (J.S.-C., V.R., T.J.A.C., P.C.E.), and the Bateson Centre (J.S.-C., J.-P.A., T.J.A.C., P.C.E.), University of Sheffield, United Kingdom; and Departments of Cardiovascular Science (A.d.L., C.W., J.C.M., D.O.H.), Imaging (P.F.F., D.F.), Bioengineering (R.K.), and Aeronautics (S.S.) Imperial College London, United Kingdom; and Cancer Institute, Faculty of Medical Sciences (H.W.), University College London, United Kingdom
| | - Dorian O Haskard
- From the Department of Infection, Immunity and Cardiovascular Disease (J.S.-C., L.A.L., S.H., I.G., M.M., S.F., C.S., N.B., J.-P.A., V.R., T.J.A.C., P.C.E.), INSIGNEO Institute for In Silico Medicine (J.S.-C., V.R., T.J.A.C., P.C.E.), and the Bateson Centre (J.S.-C., J.-P.A., T.J.A.C., P.C.E.), University of Sheffield, United Kingdom; and Departments of Cardiovascular Science (A.d.L., C.W., J.C.M., D.O.H.), Imaging (P.F.F., D.F.), Bioengineering (R.K.), and Aeronautics (S.S.) Imperial College London, United Kingdom; and Cancer Institute, Faculty of Medical Sciences (H.W.), University College London, United Kingdom
| | - Spencer Sherwin
- From the Department of Infection, Immunity and Cardiovascular Disease (J.S.-C., L.A.L., S.H., I.G., M.M., S.F., C.S., N.B., J.-P.A., V.R., T.J.A.C., P.C.E.), INSIGNEO Institute for In Silico Medicine (J.S.-C., V.R., T.J.A.C., P.C.E.), and the Bateson Centre (J.S.-C., J.-P.A., T.J.A.C., P.C.E.), University of Sheffield, United Kingdom; and Departments of Cardiovascular Science (A.d.L., C.W., J.C.M., D.O.H.), Imaging (P.F.F., D.F.), Bioengineering (R.K.), and Aeronautics (S.S.) Imperial College London, United Kingdom; and Cancer Institute, Faculty of Medical Sciences (H.W.), University College London, United Kingdom
| | - Victoria Ridger
- From the Department of Infection, Immunity and Cardiovascular Disease (J.S.-C., L.A.L., S.H., I.G., M.M., S.F., C.S., N.B., J.-P.A., V.R., T.J.A.C., P.C.E.), INSIGNEO Institute for In Silico Medicine (J.S.-C., V.R., T.J.A.C., P.C.E.), and the Bateson Centre (J.S.-C., J.-P.A., T.J.A.C., P.C.E.), University of Sheffield, United Kingdom; and Departments of Cardiovascular Science (A.d.L., C.W., J.C.M., D.O.H.), Imaging (P.F.F., D.F.), Bioengineering (R.K.), and Aeronautics (S.S.) Imperial College London, United Kingdom; and Cancer Institute, Faculty of Medical Sciences (H.W.), University College London, United Kingdom
| | - Timothy J A Chico
- From the Department of Infection, Immunity and Cardiovascular Disease (J.S.-C., L.A.L., S.H., I.G., M.M., S.F., C.S., N.B., J.-P.A., V.R., T.J.A.C., P.C.E.), INSIGNEO Institute for In Silico Medicine (J.S.-C., V.R., T.J.A.C., P.C.E.), and the Bateson Centre (J.S.-C., J.-P.A., T.J.A.C., P.C.E.), University of Sheffield, United Kingdom; and Departments of Cardiovascular Science (A.d.L., C.W., J.C.M., D.O.H.), Imaging (P.F.F., D.F.), Bioengineering (R.K.), and Aeronautics (S.S.) Imperial College London, United Kingdom; and Cancer Institute, Faculty of Medical Sciences (H.W.), University College London, United Kingdom
| | - Paul C Evans
- From the Department of Infection, Immunity and Cardiovascular Disease (J.S.-C., L.A.L., S.H., I.G., M.M., S.F., C.S., N.B., J.-P.A., V.R., T.J.A.C., P.C.E.), INSIGNEO Institute for In Silico Medicine (J.S.-C., V.R., T.J.A.C., P.C.E.), and the Bateson Centre (J.S.-C., J.-P.A., T.J.A.C., P.C.E.), University of Sheffield, United Kingdom; and Departments of Cardiovascular Science (A.d.L., C.W., J.C.M., D.O.H.), Imaging (P.F.F., D.F.), Bioengineering (R.K.), and Aeronautics (S.S.) Imperial College London, United Kingdom; and Cancer Institute, Faculty of Medical Sciences (H.W.), University College London, United Kingdom.
| |
Collapse
|
60
|
Xu S, Koroleva M, Yin M, Jin ZG. Atheroprotective laminar flow inhibits Hippo pathway effector YAP in endothelial cells. Transl Res 2016; 176:18-28.e2. [PMID: 27295628 PMCID: PMC5116386 DOI: 10.1016/j.trsl.2016.05.003] [Citation(s) in RCA: 57] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/26/2015] [Revised: 04/25/2016] [Accepted: 05/18/2016] [Indexed: 12/22/2022]
Abstract
Atherosclerosis is a mechanobiology-related disease that preferentially develops in the aortic arch and arterial branches, which are exposed to disturbed/turbulent blood flow but less in thoracic aorta where the flow pattern is steady laminar flow (LF). Increasing evidence supports that steady LF with high shear stress is protective against atherosclerosis. However, the molecular mechanisms of LF-mediated atheroprotection remain incompletely understood. Hippo/YAP (yes-associated protein) pathway senses and effects mechanical cues and has been reported to be a master regulator of cell proliferation, differentiation, and tissue homeostasis. Here, we show that LF inhibits YAP activity in endothelial cells (ECs). We observed that YAP is highly expressed in mouse EC-enriched tissues (lung and aorta) and in human ECs. Furthermore, we found in apolipoprotein E deficient (ApoE(-/-)) mice and human ECs, LF decreased the level of nuclear YAP protein and YAP target gene expression (connective tissue growth factor and cysteine-rich protein 61) through promoting Hippo kinases LATS1/2-dependent YAP (Serine 127) phosphorylation. Functionally, we revealed that YAP depletion in ECs phenocopying LF responses, reduced the expression of cell cycle gene cyclin A1 (CCNA1) and proinflammatory gene CCL2 (MCP-1). Taken together, we demonstrate that atheroprotective LF inhibits endothelial YAP activation, which may contribute to LF-mediated ECs quiescence and anti-inflammation.
Collapse
Affiliation(s)
- Suowen Xu
- Aab Cardiovascular Research Institute, Department of Medicine, University of Rochester School of Medicine and Dentistry, Rochester, NY
| | - Marina Koroleva
- Aab Cardiovascular Research Institute, Department of Medicine, University of Rochester School of Medicine and Dentistry, Rochester, NY
| | - Meimei Yin
- Aab Cardiovascular Research Institute, Department of Medicine, University of Rochester School of Medicine and Dentistry, Rochester, NY
| | - Zheng Gen Jin
- Aab Cardiovascular Research Institute, Department of Medicine, University of Rochester School of Medicine and Dentistry, Rochester, NY.
| |
Collapse
|
61
|
Heo KS, Berk BC, Abe JI. Disturbed Flow-Induced Endothelial Proatherogenic Signaling Via Regulating Post-Translational Modifications and Epigenetic Events. Antioxid Redox Signal 2016; 25:435-50. [PMID: 26714841 PMCID: PMC5076483 DOI: 10.1089/ars.2015.6556] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/27/2015] [Revised: 12/02/2015] [Accepted: 12/23/2015] [Indexed: 12/21/2022]
Abstract
SIGNIFICANCE Hemodynamic shear stress, the frictional force exerted onto the vascular endothelial cell (EC) surface, influences vascular EC functions. Atherosclerotic plaque formation in the endothelium is known to be site specific: disturbed blood flow (d-flow) formed at the lesser curvature of the aortic arch and branch points promotes plaque formation, and steady laminar flow (s-flow) at the greater curvature is atheroprotective. RECENT ADVANCES Post-translational modifications (PTMs), including phosphorylation and SUMOylation, and epigenetic events, including DNA methylation and histone modifications, provide a new perspective on the pathogenesis of atherosclerosis, elucidating how gene expression is altered by d-flow. Activation of PKCζ and p90RSK, SUMOylation of ERK5 and p53, and DNA hypermethylation are uniquely induced by d-flow, but not by s-flow. CRITICAL ISSUES Extensive cross talk has been observed among the phosphorylation, SUMOylation, acetylation, and methylation PTMs, as well as among epigenetic events along the cascade of d-flow-induced signaling, from the top (mechanosensory systems) to the bottom (epigenetic events). In addition, PKCζ activation plays a role in regulating SUMOylation-related enzymes of PIAS4, p90RSK activation plays a role in regulating SUMOylation-related enzymes of Sentrin/SUMO-specific protease (SENP)2, and DNA methyltransferase SUMOylation may play a role in d-flow signaling. FUTURE DIRECTIONS Although possible contributions of DNA events such as histone modification and the epigenetic and cytosolic events of PTMs in d-flow signaling have become clearer, determining the interplay of each PTM and epigenetic event will provide a new paradigm to elucidate the difference between d-flow and s-flow and lead to novel therapeutic interventions to inhibit plaque formation. Antioxid. Redox Signal. 25, 435-450.
Collapse
Affiliation(s)
- Kyung-Sun Heo
- Department of Cardiology, Division of Internal Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Bradford C. Berk
- Department of Medicine, Aab Cardiovascular Research Institute, University of Rochester Medical Center, Rochester, New York
| | - Jun-ichi Abe
- Department of Cardiology, Division of Internal Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas
| |
Collapse
|
62
|
Bowden N, Bryan MT, Duckles H, Feng S, Hsiao S, Kim HR, Mahmoud M, Moers B, Serbanovic-Canic J, Xanthis I, Ridger VC, Evans PC. Experimental Approaches to Study Endothelial Responses to Shear Stress. Antioxid Redox Signal 2016; 25:389-400. [PMID: 26772071 DOI: 10.1089/ars.2015.6553] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
SIGNIFICANCE Shear stress controls multiple physiological processes in endothelial cells (ECs). RECENT ADVANCES The response of ECs to shear has been studied using a range of in vitro and in vivo models. CRITICAL ISSUES This article describes some of the experimental techniques that can be used to study endothelial responses to shear stress. It includes an appraisal of large animal, rodent, and zebrafish models of vascular mechanoresponsiveness. It also describes several bioreactors to apply flow to cells and physical methods to separate mechanoresponses from mass transport mechanisms. FUTURE DIRECTIONS We conclude that combining in vitro and in vivo approaches can provide a detailed mechanistic view of vascular responses to force and that high-throughput systems are required for unbiased assessment of the function of shear-induced molecules. Antioxid. Redox Signal. 25, 389-400.
Collapse
Affiliation(s)
- Neil Bowden
- 1 Department of Infection, Immunity and Cardiovascular Disease and INSIGNEO Institute of in silico Medicine, Sheffield, United Kingdom
| | - Matthew T Bryan
- 1 Department of Infection, Immunity and Cardiovascular Disease and INSIGNEO Institute of in silico Medicine, Sheffield, United Kingdom
| | - Hayley Duckles
- 1 Department of Infection, Immunity and Cardiovascular Disease and INSIGNEO Institute of in silico Medicine, Sheffield, United Kingdom
| | - Shuang Feng
- 1 Department of Infection, Immunity and Cardiovascular Disease and INSIGNEO Institute of in silico Medicine, Sheffield, United Kingdom
| | - Sarah Hsiao
- 1 Department of Infection, Immunity and Cardiovascular Disease and INSIGNEO Institute of in silico Medicine, Sheffield, United Kingdom
| | - Hyejeong Rosemary Kim
- 1 Department of Infection, Immunity and Cardiovascular Disease and INSIGNEO Institute of in silico Medicine, Sheffield, United Kingdom .,2 The Bateson Centre, University of Sheffield , Sheffield, United Kingdom
| | - Marwa Mahmoud
- 1 Department of Infection, Immunity and Cardiovascular Disease and INSIGNEO Institute of in silico Medicine, Sheffield, United Kingdom
| | - Britta Moers
- 1 Department of Infection, Immunity and Cardiovascular Disease and INSIGNEO Institute of in silico Medicine, Sheffield, United Kingdom
| | - Jovana Serbanovic-Canic
- 1 Department of Infection, Immunity and Cardiovascular Disease and INSIGNEO Institute of in silico Medicine, Sheffield, United Kingdom .,2 The Bateson Centre, University of Sheffield , Sheffield, United Kingdom
| | - Ioannis Xanthis
- 1 Department of Infection, Immunity and Cardiovascular Disease and INSIGNEO Institute of in silico Medicine, Sheffield, United Kingdom
| | - Victoria C Ridger
- 1 Department of Infection, Immunity and Cardiovascular Disease and INSIGNEO Institute of in silico Medicine, Sheffield, United Kingdom
| | - Paul C Evans
- 1 Department of Infection, Immunity and Cardiovascular Disease and INSIGNEO Institute of in silico Medicine, Sheffield, United Kingdom .,2 The Bateson Centre, University of Sheffield , Sheffield, United Kingdom
| |
Collapse
|
63
|
Abe JI. Multiple Functions of Protein Inhibitor of Activated STAT1 in Regulating Endothelial Cell Proliferation and Inflammation. Arterioscler Thromb Vasc Biol 2016; 36:1717-9. [PMID: 27559144 DOI: 10.1161/atvbaha.116.308131] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Affiliation(s)
- Jun-Ichi Abe
- From the Department of Cardiology, University of Texas MD Anderson Cancer Center, Houston.
| |
Collapse
|
64
|
Mason JC. Cytoprotective pathways in the vascular endothelium. Do they represent a viable therapeutic target? Vascul Pharmacol 2016; 86:41-52. [PMID: 27520362 DOI: 10.1016/j.vph.2016.08.002] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2016] [Accepted: 08/08/2016] [Indexed: 12/28/2022]
Abstract
The vascular endothelium is a critical interface, which separates the organs from the blood and its contents. The endothelium has a wide variety of functions and maintenance of endothelial homeostasis is a multi-dimensional active process, disruption of which has potentially deleterious consequences if not reversed. Vascular injury predisposes to endothelial apoptosis, dysfunction and development of atherosclerosis. Endothelial dysfunction is an end-point, a central feature of which is increased ROS generation, a reduction in endothelial nitric oxide synthase and increased nitric oxide consumption. A dysfunctional endothelium is a common feature of diseases including rheumatoid arthritis, systemic lupus erythematosus, diabetes mellitus and chronic renal impairment. The endothelium is endowed with a variety of constitutive and inducible mechanisms that act to minimise injury and facilitate repair. Endothelial cytoprotection can be enhanced by exogenous factors such as vascular endothelial growth factor, prostacyclin and laminar shear stress. Target genes include endothelial nitric oxide synthase, heme oxygenase-1, A20 and anti-apoptotic members of the B cell lymphoma protein-2 family. In light of the importance of endothelial function, and the link between its disruption and the risk of atherothrombosis, interest has focused on therapeutic conditioning and reversal of endothelial dysfunction. A detailed understanding of cytoprotective signalling pathways, their regulation and target genes is now required to identify novel therapeutic targets. The ultimate aim is to add vasculoprotection to current therapeutic strategies for systemic inflammatory diseases, in an attempt to reduce vascular injury and prevent or retard atherogenesis.
Collapse
Affiliation(s)
- Justin C Mason
- Vascular Science, Imperial Centre for Translational and Experimental Medicine, National Heart and Lung Institute, Imperial College London, Hammersmith Hospital, London, UK.
| |
Collapse
|
65
|
Balaguru UM, Sundaresan L, Manivannan J, Majunathan R, Mani K, Swaminathan A, Venkatesan S, Kasiviswanathan D, Chatterjee S. Disturbed flow mediated modulation of shear forces on endothelial plane: A proposed model for studying endothelium around atherosclerotic plaques. Sci Rep 2016; 6:27304. [PMID: 27255968 PMCID: PMC4891674 DOI: 10.1038/srep27304] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2015] [Accepted: 05/16/2016] [Indexed: 02/07/2023] Open
Abstract
Disturbed fluid flow or modulated shear stress is associated with vascular conditions such as atherosclerosis, thrombosis, and aneurysm. In vitro simulation of the fluid flow around the plaque micro-environment remains a challenging approach. Currently available models have limitations such as complications in protocols, high cost, incompetence of co-culture and not being suitable for massive expression studies. Hence, the present study aimed to develop a simple, versatile model based on Computational Fluid Dynamics (CFD) simulation. Current observations of CFD have shown the regions of modulated shear stress by the disturbed fluid flow. To execute and validate the model in real sense, cell morphology, cytoskeletal arrangement, cell death, reactive oxygen species (ROS) profile, nitric oxide production and disturbed flow markers under the above condition were assessed. Endothelium at disturbed flow region which had been exposed to low shear stress and swirling flow pattern showed morphological and expression similarities with the pathological disturbed flow environment reported previously. Altogether, the proposed model can serve as a platform to simulate the real time micro-environment of disturbed flow associated with eccentric plaque shapes and the possibilities of studying its downstream events.
Collapse
Affiliation(s)
- Uma Maheswari Balaguru
- Vascular Biology Lab, AU-KBC Research Centre, MIT campus of Anna University, Chennai, India
| | | | - Jeganathan Manivannan
- Vascular Biology Lab, AU-KBC Research Centre, MIT campus of Anna University, Chennai, India
| | - Reji Majunathan
- Vascular Biology Lab, AU-KBC Research Centre, MIT campus of Anna University, Chennai, India
| | - Krishnapriya Mani
- Vascular Biology Lab, AU-KBC Research Centre, MIT campus of Anna University, Chennai, India
| | - Akila Swaminathan
- Vascular Biology Lab, AU-KBC Research Centre, MIT campus of Anna University, Chennai, India
| | | | | | - Suvro Chatterjee
- Vascular Biology Lab, AU-KBC Research Centre, MIT campus of Anna University, Chennai, India.,Centre for Biotechnology, Anna University, Chennai, India
| |
Collapse
|
66
|
Scott DW, Tolbert CE, Burridge K. Tension on JAM-A activates RhoA via GEF-H1 and p115 RhoGEF. Mol Biol Cell 2016; 27:1420-30. [PMID: 26985018 PMCID: PMC4850030 DOI: 10.1091/mbc.e15-12-0833] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2015] [Accepted: 03/10/2016] [Indexed: 12/20/2022] Open
Abstract
Forces on JAM-A activate RhoA to increase cell stiffness. Activation of RhoA requires GEF-H1 and p115 RhoGEF activation downstream of FAK/ERK and Src family kinases, respectively. Junctional adhesion molecule A (JAM-A) is a broadly expressed adhesion molecule that regulates cell–cell contacts and facilitates leukocyte transendothelial migration. The latter occurs through interactions with the integrin LFA-1. Although we understand much about JAM-A, little is known regarding the protein’s role in mechanotransduction or as a modulator of RhoA signaling. We found that tension imposed on JAM-A activates RhoA, which leads to increased cell stiffness. Activation of RhoA in this system depends on PI3K-mediated activation of GEF-H1 and p115 RhoGEF. These two GEFs are further regulated by FAK/ERK and Src family kinases, respectively. Finally, we show that phosphorylation of JAM-A at Ser-284 is required for RhoA activation in response to tension. These data demonstrate a direct role of JAM-A in mechanosignaling and control of RhoA and implicate Src family kinases in the regulation of p115 RhoGEF.
Collapse
Affiliation(s)
- David W Scott
- Department of Cell Biology and Physiology and Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599
| | - Caitlin E Tolbert
- Department of Cell Biology and Physiology and Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599
| | - Keith Burridge
- Department of Cell Biology and Physiology and Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599 McAllister Heart Institute, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599
| |
Collapse
|
67
|
Abstract
Atherosclerosis remains a major cause of morbidity and mortality worldwide, and a thorough understanding of the underlying pathophysiological mechanisms is crucial for the development of new therapeutic strategies. Although atherosclerosis is a systemic inflammatory disease, coronary atherosclerotic plaques are not uniformly distributed in the vascular tree. Experimental and clinical data highlight that biomechanical forces, including wall shear stress (WSS) and plaque structural stress (PSS), have an important role in the natural history of coronary atherosclerosis. Endothelial cell function is heavily influenced by changes in WSS, and longitudinal animal and human studies have shown that coronary regions with low WSS undergo increased plaque growth compared with high WSS regions. Local alterations in WSS might also promote transformation of stable to unstable plaque subtypes. Plaque rupture is determined by the balance between PSS and material strength, with plaque composition having a profound effect on PSS. Prospective clinical studies are required to ascertain whether integrating mechanical parameters with medical imaging can improve our ability to identify patients at highest risk of rapid disease progression or sudden cardiac events.
Collapse
|
68
|
Abe JI, Le NT, Heo KS. Role for SUMOylation in disturbed flow-induced atherosclerotic plaque formation. Biomed Eng Lett 2015. [DOI: 10.1007/s13534-015-0199-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
|
69
|
Kim GY, Kim H, Lim HJ, Park HY. Coronin 1A depletion protects endothelial cells from TNFα-induced apoptosis by modulating p38β expression and activation. Cell Signal 2015; 27:1688-93. [DOI: 10.1016/j.cellsig.2015.04.012] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2015] [Revised: 04/23/2015] [Accepted: 04/25/2015] [Indexed: 11/15/2022]
|
70
|
Wu C, Huang RT, Kuo CH, Kumar S, Kim CW, Lin YC, Chen YJ, Birukova A, Birukov KG, Dulin NO, Civelek M, Lusis AJ, Loyer X, Tedgui A, Dai G, Jo H, Fang Y. Mechanosensitive PPAP2B Regulates Endothelial Responses to Atherorelevant Hemodynamic Forces. Circ Res 2015; 117:e41-e53. [PMID: 26034042 DOI: 10.1161/circresaha.117.306457] [Citation(s) in RCA: 67] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/23/2015] [Accepted: 06/01/2015] [Indexed: 02/07/2023]
Abstract
RATIONALE PhosPhatidic Acid Phosphatase type 2B (PPAP2B), an integral membrane protein known as lipid phosphate phosphatase (LPP3) that inactivates lysophosphatidic acid, was implicated in coronary artery disease (CAD) by genome-wide association studies. However, it is unclear whether genome-wide association studies-identified coronary artery disease genes, including PPAP2B, participate in mechanotransduction mechanisms by which vascular endothelia respond to local atherorelevant hemodynamics that contribute to the regional nature of atherosclerosis. OBJECTIVE To establish the critical role of PPAP2B in endothelial responses to hemodynamics. METHODS AND RESULTS Reduced PPAP2B was detected in vivo in mouse and swine aortic arch (AA) endothelia exposed to chronic disturbed flow, and in mouse carotid artery endothelia subjected to surgically induced acute disturbed flow. In humans, PPAP2B was reduced in the downstream part of carotid plaques where low shear stress prevails. In culture, reduced PPAP2B was measured in human aortic endothelial cells under atherosusceptible waveform mimicking flow in human carotid sinus. Flow-sensitive microRNA-92a and transcription factor KLF2 were identified as upstream inhibitor and activator of endothelial PPAP2B, respectively. PPAP2B suppression abrogated atheroprotection of unidirectional flow; inhibition of lysophosphatidic acid receptor 1 restored the flow-dependent, anti-inflammatory phenotype in PPAP2B-deficient cells. PPAP2B inhibition resulted in myosin light-chain phosphorylation and intercellular gaps, which were abolished by lysophosphatidic acid receptor 1/2 inhibition. Expression quantitative trait locus mapping demonstrated PPAP2B coronary artery disease risk allele is not linked to PPAP2B expression in various human tissues but significantly associated with reduced PPAP2B in human aortic endothelial cells. CONCLUSIONS Atherorelevant flows dynamically modulate endothelial PPAP2B expression through miR-92a and KLF2. Mechanosensitive PPAP2B plays a critical role in promoting anti-inflammatory phenotype and maintaining vascular integrity of endothelial monolayer under atheroprotective flow.
Collapse
Affiliation(s)
- Congqing Wu
- Department of Medicine, University of Chicago, Los Angeles
| | - Ru-Ting Huang
- Department of Medicine, University of Chicago, Los Angeles
| | | | - Sandeep Kumar
- Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Los Angeles
| | - Chan Woo Kim
- Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Los Angeles
| | - Yen-Chen Lin
- Department of Medicine, University of Chicago, Los Angeles
| | - Yen-Ju Chen
- Department of Medicine, University of Chicago, Los Angeles
| | - Anna Birukova
- Department of Medicine, University of Chicago, Los Angeles
| | | | | | - Mete Civelek
- Department of Medicine, University of California, Los Angeles
| | - Aldons J Lusis
- Department of Medicine, University of California, Los Angeles
| | - Xavier Loyer
- Paris-Cardiovascular Research Center, University Paris Descartes
| | - Alain Tedgui
- Paris-Cardiovascular Research Center, University Paris Descartes
| | - Guohao Dai
- Department of Biomedical Engineering, Rensselaer Polytechnic Institute
| | - Hanjoong Jo
- Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Los Angeles
| | - Yun Fang
- Department of Medicine, University of Chicago, Los Angeles
| |
Collapse
|
71
|
Diebold I, Hennigs JK, Miyagawa K, Li CG, Nickel NP, Kaschwich M, Cao A, Wang L, Reddy S, Chen PI, Nakahira K, Alcazar MAA, Hopper RK, Ji L, Feldman BJ, Rabinovitch M. BMPR2 preserves mitochondrial function and DNA during reoxygenation to promote endothelial cell survival and reverse pulmonary hypertension. Cell Metab 2015; 21:596-608. [PMID: 25863249 PMCID: PMC4394191 DOI: 10.1016/j.cmet.2015.03.010] [Citation(s) in RCA: 163] [Impact Index Per Article: 18.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/04/2014] [Revised: 12/19/2014] [Accepted: 03/19/2015] [Indexed: 01/17/2023]
Abstract
Mitochondrial dysfunction, inflammation, and mutant bone morphogenetic protein receptor 2 (BMPR2) are associated with pulmonary arterial hypertension (PAH), an incurable disease characterized by pulmonary arterial (PA) endothelial cell (EC) apoptosis, decreased microvessels, and occlusive vascular remodeling. We hypothesized that reduced BMPR2 induces PAEC mitochondrial dysfunction, promoting a pro-inflammatory or pro-apoptotic state. Mice with EC deletion of BMPR2 develop hypoxia-induced pulmonary hypertension that, in contrast to non-transgenic littermates, does not reverse upon reoxygenation and is associated with reduced PA microvessels and lung EC p53, PGC1α and TFAM, regulators of mitochondrial biogenesis, and mitochondrial DNA. Decreasing PAEC BMPR2 by siRNA during reoxygenation represses p53, PGC1α, NRF2, TFAM, mitochondrial membrane potential, and ATP and induces mitochondrial DNA deletion and apoptosis. Reducing PAEC BMPR2 in normoxia increases p53, PGC1α, TFAM, mitochondrial membrane potential, ATP production, and glycolysis, and induces mitochondrial fission and a pro-inflammatory state. These features are recapitulated in PAECs from PAH patients with mutant BMPR2.
Collapse
Affiliation(s)
- Isabel Diebold
- Department of Pediatrics and Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA 94305, USA; The Vera Moulton Wall Center for Pulmonary Vascular Disease, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Jan K Hennigs
- Department of Pediatrics and Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA 94305, USA; The Vera Moulton Wall Center for Pulmonary Vascular Disease, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Kazuya Miyagawa
- Department of Pediatrics and Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA 94305, USA; The Vera Moulton Wall Center for Pulmonary Vascular Disease, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Caiyun G Li
- Department of Pediatrics and Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA 94305, USA; The Vera Moulton Wall Center for Pulmonary Vascular Disease, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Nils P Nickel
- Department of Pediatrics and Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA 94305, USA; The Vera Moulton Wall Center for Pulmonary Vascular Disease, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Mark Kaschwich
- Department of Pediatrics and Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA 94305, USA; The Vera Moulton Wall Center for Pulmonary Vascular Disease, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Aiqin Cao
- Department of Pediatrics and Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA 94305, USA; The Vera Moulton Wall Center for Pulmonary Vascular Disease, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Lingli Wang
- Department of Pediatrics and Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA 94305, USA; The Vera Moulton Wall Center for Pulmonary Vascular Disease, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Sushma Reddy
- Department of Pediatrics and Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Pin-I Chen
- Department of Pediatrics and Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA 94305, USA; The Vera Moulton Wall Center for Pulmonary Vascular Disease, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Kiichi Nakahira
- Department of Medicine, Weill Cornell Medical College, New York, NY 10065, USA
| | - Miguel A Alejandre Alcazar
- Department of Pediatrics and Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA 94305, USA; The Vera Moulton Wall Center for Pulmonary Vascular Disease, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Rachel K Hopper
- Department of Pediatrics and Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA 94305, USA; The Vera Moulton Wall Center for Pulmonary Vascular Disease, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Lijuan Ji
- Department of Pediatrics and Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Brian J Feldman
- Department of Pediatrics and Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Marlene Rabinovitch
- Department of Pediatrics and Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA 94305, USA; The Vera Moulton Wall Center for Pulmonary Vascular Disease, Stanford University School of Medicine, Stanford, CA 94305, USA.
| |
Collapse
|
72
|
Heo KS, Le NT, Cushman HJ, Giancursio CJ, Chang E, Woo CH, Sullivan MA, Taunton J, Yeh ETH, Fujiwara K, Abe JI. Disturbed flow-activated p90RSK kinase accelerates atherosclerosis by inhibiting SENP2 function. J Clin Invest 2015; 125:1299-310. [PMID: 25689261 DOI: 10.1172/jci76453] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2014] [Accepted: 01/06/2015] [Indexed: 01/31/2023] Open
Abstract
Disturbed blood flow (d-flow) causes endothelial cell (EC) dysfunction, leading to atherosclerotic plaque formation. We have previously shown that d-flow increases SUMOylation of p53 and ERK5 through downregulation of sentrin/SUMO-specific protease 2 (SENP2) function; however, it is not known how SENP2 itself is regulated by d-flow. Here, we determined that d-flow activated the serine/threonine kinase p90RSK, which subsequently phosphorylated threonine 368 (T368) of SENP2. T368 phosphorylation promoted nuclear export of SENP2, leading to downregulation of eNOS expression and upregulation of proinflammatory adhesion molecule expression and apoptosis. In an LDLR-deficient murine model of atherosclerosis, EC-specific overexpression of p90RSK increased EC dysfunction and lipid accumulation in the aorta compared with control animals; however, these pathologic changes were not observed in atherosclerotic mice overexpressing dominant negative p90RSK (DN-p90RSK). Moreover, depletion of SENP2 in these mice abolished the protective effect of DN-p90RSK overexpression. We propose that p90RSK-mediated SENP2-T368 phosphorylation is a master switch in d-flow-induced signaling, leading to EC dysfunction and atherosclerosis.
Collapse
|
73
|
Loss of One Copy of Zfp148 Reduces Lesional Macrophage Proliferation and Atherosclerosis in Mice by Activating p53. Circ Res 2014; 115:781-9. [DOI: 10.1161/circresaha.115.304992] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Rationale:
Cell proliferation and cell cycle control mechanisms are thought to play central roles in the pathogenesis of atherosclerosis. The transcription factor Zinc finger protein 148 (Zfp148) was shown recently to maintain cell proliferation under oxidative conditions by suppressing p53, a checkpoint protein that arrests proliferation in response to various stressors. It is established that inactivation of p53 accelerates atherosclerosis, but whether increased p53 activation confers protection against the disease remains to be determined.
Objective:
We aimed to test the hypothesis that
Zfp148
deficiency reduces atherosclerosis by unleashing p53 activity.
Methods and Results:
Mice harboring a gene-trap mutation in the
Zfp148
locus (
Zfp148
gt/+
) were bred onto the apolipoprotein E (
Apoe
)
–/–
genetic background and fed a high-fat or chow diet. Loss of 1 copy of
Zfp148
markedly reduced atherosclerosis without affecting lipid metabolism. Bone marrow transplantation experiments revealed that the effector cell is of hematopoietic origin. Peritoneal macrophages and atherosclerotic lesions from
Zfp148
gt/+
Apoe
–/–
mice showed increased levels of phosphorylated p53 compared with controls, and atherosclerotic lesions contained fewer proliferating macrophages.
Zfp148
gt/+
Apoe
–/–
mice were further crossed with p53-null mice (
Trp53
–/–
[the gene encoding p53]). There was no difference in atherosclerosis between
Zfp148
gt/+
Apoe
–/–
mice and controls on a
Trp53
+/–
genetic background, and there was no difference in levels of phosphorylated p53 or cell proliferation.
Conclusions:
Zfp148
deficiency increases p53 activity and protects against atherosclerosis by causing proliferation arrest of lesional macrophages, suggesting that drugs targeting macrophage proliferation may be useful in the treatment of atherosclerosis.
Collapse
|
74
|
Abstract
Atherosclerosis is a focal disease that develops preferentially where nonlaminar, disturbed blood flow occurs, such as branches, bifurcations, and curvatures of large arteries. Endothelial cells sense and respond differently to disturbed flow compared with steady laminar flow. Disturbed flow that occurs in so-called atheroprone areas activates proinflammatory and apoptotic signaling, and this results in endothelial dysfunction and leads to subsequent development of atherosclerosis. In contrast, steady laminar flow as atheroprotective flow promotes expression of many anti-inflammatory genes, such as Kruppel-like factor 2 and endothelial nitric oxide synthase and inhibits endothelial inflammation and athrogenesis. Here we will discuss that disturbed flow and steady laminar flow induce pro- and antiatherogenic events via flow type-specific mechanotransduction pathways. We will focus on 5 mechanosensitive pathways: mitogen-activated protein kinases/extracellular signal-regulated kinase 5/Kruppel-like factor 2 signaling, extracellular signal-regulated kinase/peroxisome proliferator-activated receptor signaling, and mechanosignaling pathways involving SUMOylation, protein kinase C-ζ, and p90 ribosomal S6 kinase. We think that clarifying regulation mechanisms between these 2 flow types will provide new insights into therapeutic approaches for the prevention and treatment of atherosclerosis.
Collapse
Affiliation(s)
- Jun-ichi Abe
- From the Aab Cardiovascular Research Institute, University of Rochester, NY.
| | - Bradford C Berk
- From the Aab Cardiovascular Research Institute, University of Rochester, NY.
| |
Collapse
|
75
|
Kwak BR, Bäck M, Bochaton-Piallat ML, Caligiuri G, Daemen MJAP, Davies PF, Hoefer IE, Holvoet P, Jo H, Krams R, Lehoux S, Monaco C, Steffens S, Virmani R, Weber C, Wentzel JJ, Evans PC. Biomechanical factors in atherosclerosis: mechanisms and clinical implications. Eur Heart J 2014; 35:3013-20, 3020a-3020d. [PMID: 25230814 DOI: 10.1093/eurheartj/ehu353] [Citation(s) in RCA: 313] [Impact Index Per Article: 31.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Blood vessels are exposed to multiple mechanical forces that are exerted on the vessel wall (radial, circumferential and longitudinal forces) or on the endothelial surface (shear stress). The stresses and strains experienced by arteries influence the initiation of atherosclerotic lesions, which develop at regions of arteries that are exposed to complex blood flow. In addition, plaque progression and eventually plaque rupture is influenced by a complex interaction between biological and mechanical factors-mechanical forces regulate the cellular and molecular composition of plaques and, conversely, the composition of plaques determines their ability to withstand mechanical load. A deeper understanding of these interactions is essential for designing new therapeutic strategies to prevent lesion development and promote plaque stabilization. Moreover, integrating clinical imaging techniques with finite element modelling techniques allows for detailed examination of local morphological and biomechanical characteristics of atherosclerotic lesions that may be of help in prediction of future events. In this ESC Position Paper on biomechanical factors in atherosclerosis, we summarize the current 'state of the art' on the interface between mechanical forces and atherosclerotic plaque biology and identify potential clinical applications and key questions for future research.
Collapse
Affiliation(s)
- Brenda R Kwak
- Department of Pathology and Immunology, University of Geneva, CMU, Rue Michel-Servet 1, CH-1211 Geneva, Switzerland
| | | | | | | | | | | | - Imo E Hoefer
- University Medical Center Urecht, Utrecht, The Netherlands
| | | | | | | | | | | | | | | | | | | | - Paul C Evans
- Department of Cardiovascular Science, Medical School, University of Sheffield, Beech Hill Road, Sheffield S10 2RX, UK
| |
Collapse
|
76
|
PTEN phosphatase-independent maintenance of glandular morphology in a predictive colorectal cancer model system. Neoplasia 2014; 15:1218-30. [PMID: 24348097 DOI: 10.1593/neo.121516] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2012] [Revised: 10/08/2013] [Accepted: 10/11/2013] [Indexed: 02/06/2023] Open
Abstract
Organotypic models may provide mechanistic insight into colorectal cancer (CRC) morphology. Three-dimensional (3D) colorectal gland formation is regulated by phosphatase and tensin homologue deleted on chromosome 10 (PTEN) coupling of cell division cycle 42 (cdc42) to atypical protein kinase C (aPKC). This study investigated PTEN phosphatase-dependent and phosphatase-independent morphogenic functions in 3D models and assessed translational relevance in human studies. Isogenic PTEN-expressing or PTEN-deficient 3D colorectal cultures were used. In translational studies, apical aPKC activity readout was assessed against apical membrane (AM) orientation and gland morphology in 3D models and human CRC. We found that catalytically active or inactive PTEN constructs containing an intact C2 domain enhanced cdc42 activity, whereas mutants of the C2 domain calcium binding region 3 membrane-binding loop (M-CBR3) were ineffective. The isolated PTEN C2 domain (C2) accumulated in membrane fractions, but C2 M-CBR3 remained in cytosol. Transfection of C2 but not C2 M-CBR3 rescued defective AM orientation and 3D morphogenesis of PTEN-deficient Caco-2 cultures. The signal intensity of apical phospho-aPKC correlated with that of Na(+)/H(+) exchanger regulatory factor-1 (NHERF-1) in the 3D model. Apical NHERF-1 intensity thus provided readout of apical aPKC activity and associated with glandular morphology in the model system and human colon. Low apical NHERF-1 intensity in CRC associated with disruption of glandular architecture, high cancer grade, and metastatic dissemination. We conclude that the membrane-binding function of the catalytically inert PTEN C2 domain influences cdc42/aPKC-dependent AM dynamics and gland formation in a highly relevant 3D CRC morphogenesis model system.
Collapse
|
77
|
Abstract
Hemodynamic shear stress, the frictional force acting on vascular endothelial cells, is crucial for endothelial homeostasis under normal physiological conditions. When discussing blood flow effects on various forms of endothelial (dys)function, one considers two flow patterns: steady laminar flow and disturbed flow because endothelial cells respond differently to these flow types both in vivo and in vitro. Laminar flow which exerts steady laminar shear stress is atheroprotective while disturbed flow creates an atheroprone environment. Emerging evidence has provided new insights into the cellular mechanisms of flow-dependent regulation of vascular function that leads to cardiovascular events such as atherosclerosis, atherothrombosis, and myocardial infarction. In order to study effects of shear stress and different types of flow, various models have been used. In this review, we will summarize our current views on how disturbed flow-mediated signaling pathways are involved in the development of atherosclerosis.
Collapse
Affiliation(s)
- Kyung-Sun Heo
- Aab Cardiovascular Research Institute, University of Rochester, Rochester, NY 14642,
USA
| | - Keigi Fujiwara
- Aab Cardiovascular Research Institute, University of Rochester, Rochester, NY 14642,
USA
| | - Jun-ichi Abe
- Aab Cardiovascular Research Institute, University of Rochester, Rochester, NY 14642,
USA
| |
Collapse
|
78
|
Shao B, Bayraktutan U. Hyperglycaemia promotes human brain microvascular endothelial cell apoptosis via induction of protein kinase C-ßI and prooxidant enzyme NADPH oxidase. Redox Biol 2014; 2:694-701. [PMID: 24936444 PMCID: PMC4052534 DOI: 10.1016/j.redox.2014.05.005] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2014] [Revised: 05/21/2014] [Accepted: 05/23/2014] [Indexed: 01/13/2023] Open
Abstract
Blood-brain barrier disruption represents a key feature in hyperglycaemia-aggravated cerebral damage after an ischaemic stroke. Although the underlying mechanisms remain largely unknown, activation of protein kinase C (PKC) is thought to play a critical role. This study examined whether apoptosis of human brain microvascular endothelial cells (HBMEC) might contribute to hyperglycaemia-evoked barrier damage and assessed the specific role of PKC in this phenomenon. Treatments with hyperglycaemia (25 mM) or phorbol myristate acetate (PMA, a protein kinase C activator, 100 nM) significantly increased NADPH oxidase activity, O2 (•-) generation, proapoptotic protein Bax expression, TUNEL-positive staining and caspase-3/7 activities. Pharmacological inhibition of NADPH oxidase, PKC-a, PKC-ß or PKC-ßI via their specific inhibitors and neutralisation of O2 (•-) by a cell-permeable superoxide dismutase mimetic, MnTBAP normalised all the aforementioned increases induced by hyperglycaemia. Suppression of these PKC isoforms also negated the stimulatory effects of hyperglycaemia on the protein expression of NADPH oxidase membrane-bound components, Nox2 and p22-phox which determine the overall enzymatic activity. Silencing of PKC-ßI gene through use of specific siRNAs abolished the effects of both hyperglycaemia and PMA on endothelial cell NADPH oxidase activity, O2 (•-) production and apoptosis and consequently improved the integrity and function of an in vitro model of human cerebral barrier comprising HBMEC, astrocytes and pericytes. Hyperglycaemia-mediated apoptosis of HBMEC contributes to cerebral barrier dysfunction and is modulated by sequential activations of PKC-ßI and NADPH oxidase.
Collapse
Affiliation(s)
- Beili Shao
- Stroke, Division of Clinical Neuroscience, School of Medicine, University of Nottingham, UK
| | - Ulvi Bayraktutan
- Stroke, Division of Clinical Neuroscience, School of Medicine, University of Nottingham, UK
| |
Collapse
|
79
|
Heo KS, Cushman HJ, Akaike M, Woo CH, Wang X, Qiu X, Fujiwara K, Abe JI. ERK5 activation in macrophages promotes efferocytosis and inhibits atherosclerosis. Circulation 2014; 130:180-91. [PMID: 25001623 DOI: 10.1161/circulationaha.113.005991] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
BACKGROUND Efferocytosis is a process by which dead and dying cells are removed by phagocytic cells. Efferocytosis by macrophages is thought to curb the progression of atherosclerosis, but the mechanistic insight of this process is lacking. METHODS AND RESULTS When macrophages were fed apoptotic cells or treated with pitavastatin in vitro, efferocytosis-related signaling and phagocytic capacity were upregulated in an ERK5 activity-dependent manner. Macrophages isolated from macrophage-specific ERK5-null mice exhibited reduced efferocytosis and levels of gene and protein expression of efferocytosis-related molecules. When these mice were crossed with low-density lipoprotein receptor(-/-) mice and fed a high-cholesterol diet, atherosclerotic plaque formation was accelerated, and the plaques had more advanced and vulnerable morphology. CONCLUSIONS Our results demonstrate that ERK5, which is robustly activated by statins, is a hub molecule that upregulates macrophage efferocytosis, thereby suppressing atherosclerotic plaque formation. Molecules that upregulate ERK5 and its signaling in macrophages may be good drug targets for suppressing cardiovascular diseases.
Collapse
Affiliation(s)
- Kyung-Sun Heo
- From the Aab Cardiovascular Research Institute and Department of Medicine, University of Rochester School of Medicine and Dentistry, Rochester, NY (K.H., H.J.C., C.W., K.F., J.A.); Department of Medical Education, Institute of Health Biosciences, University of Tokushima Graduate School, Kuramoto-cho, Tokushima, Japan (M.A.); Faculty of Life Sciences, University of Manchester, Manchester, United Kingdom (X.W.); and Biostatistics and Computational Biology, University of Rochester School of Medicine and Dentistry, Rochester, NY (X.Q.).
| | - Hannah J Cushman
- From the Aab Cardiovascular Research Institute and Department of Medicine, University of Rochester School of Medicine and Dentistry, Rochester, NY (K.H., H.J.C., C.W., K.F., J.A.); Department of Medical Education, Institute of Health Biosciences, University of Tokushima Graduate School, Kuramoto-cho, Tokushima, Japan (M.A.); Faculty of Life Sciences, University of Manchester, Manchester, United Kingdom (X.W.); and Biostatistics and Computational Biology, University of Rochester School of Medicine and Dentistry, Rochester, NY (X.Q.)
| | - Masashi Akaike
- From the Aab Cardiovascular Research Institute and Department of Medicine, University of Rochester School of Medicine and Dentistry, Rochester, NY (K.H., H.J.C., C.W., K.F., J.A.); Department of Medical Education, Institute of Health Biosciences, University of Tokushima Graduate School, Kuramoto-cho, Tokushima, Japan (M.A.); Faculty of Life Sciences, University of Manchester, Manchester, United Kingdom (X.W.); and Biostatistics and Computational Biology, University of Rochester School of Medicine and Dentistry, Rochester, NY (X.Q.)
| | - Chang-Hoon Woo
- From the Aab Cardiovascular Research Institute and Department of Medicine, University of Rochester School of Medicine and Dentistry, Rochester, NY (K.H., H.J.C., C.W., K.F., J.A.); Department of Medical Education, Institute of Health Biosciences, University of Tokushima Graduate School, Kuramoto-cho, Tokushima, Japan (M.A.); Faculty of Life Sciences, University of Manchester, Manchester, United Kingdom (X.W.); and Biostatistics and Computational Biology, University of Rochester School of Medicine and Dentistry, Rochester, NY (X.Q.)
| | - Xin Wang
- From the Aab Cardiovascular Research Institute and Department of Medicine, University of Rochester School of Medicine and Dentistry, Rochester, NY (K.H., H.J.C., C.W., K.F., J.A.); Department of Medical Education, Institute of Health Biosciences, University of Tokushima Graduate School, Kuramoto-cho, Tokushima, Japan (M.A.); Faculty of Life Sciences, University of Manchester, Manchester, United Kingdom (X.W.); and Biostatistics and Computational Biology, University of Rochester School of Medicine and Dentistry, Rochester, NY (X.Q.)
| | - Xing Qiu
- From the Aab Cardiovascular Research Institute and Department of Medicine, University of Rochester School of Medicine and Dentistry, Rochester, NY (K.H., H.J.C., C.W., K.F., J.A.); Department of Medical Education, Institute of Health Biosciences, University of Tokushima Graduate School, Kuramoto-cho, Tokushima, Japan (M.A.); Faculty of Life Sciences, University of Manchester, Manchester, United Kingdom (X.W.); and Biostatistics and Computational Biology, University of Rochester School of Medicine and Dentistry, Rochester, NY (X.Q.)
| | - Keigi Fujiwara
- From the Aab Cardiovascular Research Institute and Department of Medicine, University of Rochester School of Medicine and Dentistry, Rochester, NY (K.H., H.J.C., C.W., K.F., J.A.); Department of Medical Education, Institute of Health Biosciences, University of Tokushima Graduate School, Kuramoto-cho, Tokushima, Japan (M.A.); Faculty of Life Sciences, University of Manchester, Manchester, United Kingdom (X.W.); and Biostatistics and Computational Biology, University of Rochester School of Medicine and Dentistry, Rochester, NY (X.Q.)
| | - Jun-ichi Abe
- From the Aab Cardiovascular Research Institute and Department of Medicine, University of Rochester School of Medicine and Dentistry, Rochester, NY (K.H., H.J.C., C.W., K.F., J.A.); Department of Medical Education, Institute of Health Biosciences, University of Tokushima Graduate School, Kuramoto-cho, Tokushima, Japan (M.A.); Faculty of Life Sciences, University of Manchester, Manchester, United Kingdom (X.W.); and Biostatistics and Computational Biology, University of Rochester School of Medicine and Dentistry, Rochester, NY (X.Q.).
| |
Collapse
|
80
|
Warboys CM, de Luca A, Amini N, Luong L, Duckles H, Hsiao S, White A, Biswas S, Khamis R, Chong CK, Cheung WM, Sherwin SJ, Bennett MR, Gil J, Mason JC, Haskard DO, Evans PC. Disturbed flow promotes endothelial senescence via a p53-dependent pathway. Arterioscler Thromb Vasc Biol 2014; 34:985-95. [PMID: 24651677 DOI: 10.1161/atvbaha.114.303415] [Citation(s) in RCA: 144] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
OBJECTIVE Although atherosclerosis is associated with systemic risk factors such as age, high cholesterol, and obesity, plaque formation occurs predominately at branches and bends that are exposed to disturbed patterns of blood flow. The molecular mechanisms that link disturbed flow-generated mechanical forces with arterial injury are uncertain. To illuminate them, we investigated the effects of flow on endothelial cell (EC) senescence. APPROACH AND RESULTS LDLR(-/-) (low-density lipoprotein receptor(-/-)) mice were exposed to a high-fat diet for 2 to 12 weeks (or to a normal chow diet as a control) before the assessment of cellular senescence in aortic ECs. En face staining revealed that senescence-associated β-galactosidase activity and p53 expression were elevated in ECs at sites of disturbed flow in response to a high-fat diet. By contrast, ECs exposed to undisturbed flow did not express senescence-associated β-galactosidase or p53. Studies of aortae from healthy pigs (aged 6 months) also revealed enhanced senescence-associated β-galactosidase staining at sites of disturbed flow. These data suggest that senescent ECs accumulate at disturbed flow sites during atherogenesis. We used in vitro flow systems to examine whether a causal relationship exists between flow and EC senescence. Exposure of cultured ECs to flow (using either an orbital shaker or a syringe-pump flow bioreactor) revealed that disturbed flow promoted EC senescence compared with static conditions, whereas undisturbed flow reduced senescence. Gene silencing studies demonstrated that disturbed flow induced EC senescence via a p53-p21 signaling pathway. Disturbed flow-induced senescent ECs exhibited reduced migration compared with nonsenescent ECs in a scratch wound closure assay, and thus may be defective for arterial repair. However, pharmacological activation of sirtuin 1 (using resveratrol or SRT1720) protected ECs from disturbed flow-induced senescence. CONCLUSIONS Disturbed flow promotes endothelial senescence via a p53-p21-dependent pathway which can be inhibited by activation of sirtuin 1. These observations support the principle that pharmacological activation of sirtuin 1 may promote cardiovascular health by suppressing EC senescence at atheroprone sites.
Collapse
Affiliation(s)
- Christina M Warboys
- From the British Heart Foundation Cardiovascular Science Unit, National Heart and Lung Institute (C.M.W., A.d.L., NA., R.K., W.-M.C., J.C.M., D.O.H.), Department of Aeronautics (S.J.S.), and MRC Clinical Sciences Centre (J.G.), Imperial College London, London, United Kingdom; Departments of Cardiovascular Science (L.L., H.D., S.H., S.B., P.C.E.) and Materials Science and Engineering (A.W., C.K.C.) and Insigneo Institute of In Silico Medicine (P.C.E.), University of Sheffield, Sheffield, United Kingdom; and Division of Cardiovascular Medicine, University of Cambridge, Cambridge, United Kingdom (M.R.B.)
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
81
|
Abstract
At least 468 individual genes have been manipulated by molecular methods to study their effects on the initiation, promotion, and progression of atherosclerosis. Most clinicians and many investigators, even in related disciplines, find many of these genes and the related pathways entirely foreign. Medical schools generally do not attempt to incorporate the relevant molecular biology into their curriculum. A number of key signaling pathways are highly relevant to atherogenesis and are presented to provide a context for the gene manipulations summarized herein. The pathways include the following: the insulin receptor (and other receptor tyrosine kinases); Ras and MAPK activation; TNF-α and related family members leading to activation of NF-κB; effects of reactive oxygen species (ROS) on signaling; endothelial adaptations to flow including G protein-coupled receptor (GPCR) and integrin-related signaling; activation of endothelial and other cells by modified lipoproteins; purinergic signaling; control of leukocyte adhesion to endothelium, migration, and further activation; foam cell formation; and macrophage and vascular smooth muscle cell signaling related to proliferation, efferocytosis, and apoptosis. This review is intended primarily as an introduction to these key signaling pathways. They have become the focus of modern atherosclerosis research and will undoubtedly provide a rich resource for future innovation toward intervention and prevention of the number one cause of death in the modern world.
Collapse
Affiliation(s)
- Paul N Hopkins
- Cardiovascular Genetics, Department of Internal Medicine, University of Utah, Salt Lake City, Utah, USA.
| |
Collapse
|
82
|
Santiago A, Li D, Zhao LY, Godsey A, Liao D. p53 SUMOylation promotes its nuclear export by facilitating its release from the nuclear export receptor CRM1. Mol Biol Cell 2013; 24:2739-52. [PMID: 23825024 PMCID: PMC3756925 DOI: 10.1091/mbc.e12-10-0771] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2012] [Revised: 06/24/2013] [Accepted: 06/24/2013] [Indexed: 12/11/2022] Open
Abstract
Chromosomal region maintenance 1 (CRM1) mediates p53 nuclear export. Although p53 SUMOylation promotes its nuclear export, the underlying mechanism is unclear. Here we show that tethering of a small, ubiquitin-like modifier (SUMO) moiety to p53 markedly increases its cytoplasmic localization. SUMO attachment to p53 does not affect its oligomerization, suggesting that subunit dissociation required for exposing p53's nuclear export signal (NES) is unnecessary for p53 nuclear export. Surprisingly, SUMO-mediated p53 nuclear export depends on the SUMO-interacting motif (SIM)-binding pocket of SUMO-1. The CRM1 C-terminal domain lacking the NES-binding groove interacts with tetrameric p53, and the proper folding of the p53 core domain, rather than the presence of the N- or C-terminal tails, appears to be important for p53-CRM1 interaction. The CRM1 Huntington, EF3, a subunit of PP2A, and TOR1 9 (HEAT9) loop, which regulates GTP-binding nuclear protein Ran binding and cargo release, contains a prototypical SIM. Remarkably, disruption of this SIM in conjunction with a mutated SIM-binding groove of SUMO-1 markedly enhances the binding of CRM1 to p53-SUMO-1 and their accumulation in the nuclear pore complexes (NPCs), as well as their persistent association in the cytoplasm. We propose that SUMOylation of a CRM1 cargo such as p53 at the NPCs unlocks the HEAT9 loop of CRM1 to facilitate the disassembly of the transporting complex and cargo release to the cytoplasm.
Collapse
Affiliation(s)
- Aleixo Santiago
- Department of Anatomy and Cell Biology, UF Health Cancer Center, and UF Genetics Institute, University of Florida College of Medicine, Gainesville, FL 32610
| | - Dawei Li
- Department of Anatomy and Cell Biology, UF Health Cancer Center, and UF Genetics Institute, University of Florida College of Medicine, Gainesville, FL 32610
- Department of Urology, Qilu Hospital, Shandong University, Jinan 250012, Shandong, China
| | - Lisa Y. Zhao
- Department of Anatomy and Cell Biology, UF Health Cancer Center, and UF Genetics Institute, University of Florida College of Medicine, Gainesville, FL 32610
| | - Adam Godsey
- Department of Anatomy and Cell Biology, UF Health Cancer Center, and UF Genetics Institute, University of Florida College of Medicine, Gainesville, FL 32610
| | - Daiqing Liao
- Department of Anatomy and Cell Biology, UF Health Cancer Center, and UF Genetics Institute, University of Florida College of Medicine, Gainesville, FL 32610
| |
Collapse
|
83
|
Heo KS, Chang E, Le NT, Cushman H, Yeh ETH, Fujiwara K, Abe JI. De-SUMOylation enzyme of sentrin/SUMO-specific protease 2 regulates disturbed flow-induced SUMOylation of ERK5 and p53 that leads to endothelial dysfunction and atherosclerosis. Circ Res 2013; 112:911-23. [PMID: 23381569 DOI: 10.1161/circresaha.111.300179] [Citation(s) in RCA: 83] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
RATIONALE Disturbed flow induces proinflammatory and apoptotic responses in endothelial cells, causing them to become dysfunctional and subsequently proatherogenic. OBJECTIVE Although a possible link between SUMOylation of p53 and ERK5 detected during endothelial apoptosis and inflammation has been suggested, the mechanistic insights, especially under the proatherogenic flow condition, remain largely unknown. METHODS AND RESULTS SUMOylation of p53 and ERK5 was induced by disturbed flow but not by steady laminar flow. To examine the role of the disturbed flow-induced p53 and ERK5 SUMOylation, we used de-SUMOylation enzyme of sentrin/Small Ubiquitin-like MOdifier (SUMO)-specific protease 2 deficiency (Senp2(+/-)) mice and observed a significant increase in endothelial apoptosis and adhesion molecule expression both in vitro and in vivo. These increases, however, were significantly inhibited in endothelial cells overexpressing p53 and ERK5 SUMOylation site mutants. Senp2(+/-) mice exhibited increased leukocyte rolling along the endothelium, and accelerated formation of atherosclerotic lesions was observed in Senp2(+/-)/Ldlr(-/-), but not in Senp2(+/+)/Ldlr(-/-), mice fed a high-cholesterol diet. Notably, the extent of lesion size in the aortic arch of Senp2(+/-)/Ldlr(-/-) mice was much larger than that in the descending aorta, also suggesting a crucial role of the disturbed flow-induced SUMOylation of proteins, including p53 and ERK5 in atherosclerosis formation. CONCLUSIONS These data show the unique role of sentrin/SUMO-specific protease 2 on endothelial function under disturbed flow and suggest that SUMOylation of p53 and ERK5 by disturbed flow contributes to the atherosclerotic plaque formation. Molecules involved in this newly discovered signaling will be useful targets for controlling endothelial cells dysfunction and consequently atherosclerosis formation.
Collapse
Affiliation(s)
- Kyung-Sun Heo
- Aab Cardiovascular Research Institute and Department of Medicine, University of Rochester School of Medicine and Dentistry, 601 Elmwood Ave, Rochester, NY 14642, USA
| | | | | | | | | | | | | |
Collapse
|
84
|
Heo KS, Chang E, Takei Y, Le NT, Woo CH, Sullivan MA, Morrell C, Fujiwara K, Abe JI. Phosphorylation of protein inhibitor of activated STAT1 (PIAS1) by MAPK-activated protein kinase-2 inhibits endothelial inflammation via increasing both PIAS1 transrepression and SUMO E3 ligase activity. Arterioscler Thromb Vasc Biol 2012. [PMID: 23202365 DOI: 10.1161/atvbaha.112.300619] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
OBJECTIVE Protein inhibitor of activated signal transducer and activator of transcription-1 (PIAS1) is known to function as small ubiquitin-like modifier (SUMO) E3 ligase as well as transrepressor. The aim of the study is to elucidate the regulatory mechanisms for these 2 different functions, especially with respect to endothelial inflammation. METHODS AND RESULTS The mitogen-activated protein kinase (MAPK)-activated protein kinase-2 is a proinflammatory kinase and phosphorylates PIAS1 at the Ser522 residue. Activation of MAPK-activated protein kinase-2 enhances p53-SUMOylation, but a PIAS1 phosphorylation mutant, PIAS1-S522A, abolished this p53-SUMOylation, suggesting a critical role for PIAS1-S522 phosphorylation in its SUMO ligase activity. Because nuclear p53 can inhibit Kruppel-like factor 2 promoter activity, we investigated the roles for PIAS1 phosphorylation and p53-SUMOylation in the Kruppel-like factor 2 and endothelial NO synthase expression. Both MAPK-activated protein kinase-2 and PIAS1 overexpression increased Kruppel-like factor 2 promoter activity and endothelial NO synthase expression, which were inhibited by expressing a p53-SUMOylation defective mutant, p53-K386R, and PIAS1-S522A. PIAS1-S522A also abolished the anti-inflammatory effect of wild-type PIAS1 in vitro and also in vivo, which was examined by leukocyte rolling in microvessels of skin grafts transduced by adenovirus encoding PIAS1-WT or - S522A mutant. CONCLUSIONS Our study has identified a novel negative feedback regulatory pathway through which MAPK-activated protein kinase-2 limits endothelial inflammation via the PIAS1 S522 phosphorylation-mediated increase in PIAS1 transrepression and SUMO ligase activity.
Collapse
Affiliation(s)
- Kyung-Sun Heo
- Aab Cardiovascular Research Institute and Department of Medicine, University of Rochester School of Medicine and Dentistry, Rochester, NY 14642, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
85
|
Kim GY, Nigro P, Fujiwara K, Abe JI, Berk BC. p62 binding to protein kinase C ζ regulates tumor necrosis factor α-induced apoptotic pathway in endothelial cells. Arterioscler Thromb Vasc Biol 2012; 32:2974-80. [PMID: 23023376 DOI: 10.1161/atvbaha.112.300054] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
OBJECTIVE Protein kinase C (PKC) ζ is a key pathological mediator of endothelial cell apoptosis. p62 is a scaffold protein that regulates several cell signaling pathways by binding to target proteins. Because PKCζ and p62 contain Phox/Bem1p (PB1) modules that mediate protein-protein interactions, we hypothesized that an interaction between p62 and PKCζ is required for tumor necrosis factor α-induced PKCζ signaling in endothelial cells. METHODS AND RESULTS In human umbilical vein endothelial cell, tumor necrosis factor α (10 ng/mL) enhanced the interaction between p62 and PKCζ. Transfection with p62 small interfering RNA reduced the activation of both PKCζ and its downstream targets JNK and caspase 3, suggesting that p62 is necessary for PKCζ signaling. Overexpression of only the PB1 domain of p62 inhibited p62-PKCζ interaction, showing that binding of these 2 proteins is mediated by their PB1 domains. Furthermore, overexpression of the p62 PB1 domain suppressed tumor necrosis factor α-induced PKCζ activation and subsequent activation of JNK and caspase 3. Finally, transfection of either p62 small interfering RNA or the PB1 domain of p62 inhibited human umbilical vein endothelial cell apoptosis. CONCLUSIONS Our results suggest a novel function of p62 that regulates the activity of PKCζ by binding to PKCζ, thereby activating the PKCζ-JNK-caspase 3 apoptotic pathway in endothelial cells.
Collapse
Affiliation(s)
- Geun-Young Kim
- Department of Medicine, Aab Cardiovascular Research Institute, University of Rochester School of Medicine and Dentistry, Rochester, NY 14642, USA
| | | | | | | | | |
Collapse
|
86
|
Reactive Oxygen Species, SUMOylation, and Endothelial Inflammation. Int J Inflam 2012; 2012:678190. [PMID: 22991685 PMCID: PMC3443607 DOI: 10.1155/2012/678190] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2012] [Accepted: 06/26/2012] [Indexed: 12/14/2022] Open
Abstract
Although the exact mechanism through which NADPH oxidases (Nox's) generate reactive oxygen species (ROS) is still not completely understood, it is widely considered that ROS accumulation is the cause of oxidative stress in endothelial cells. Increasing pieces of evidence strongly indicate the role for ROS in endothelial inflammation and dysfunction and subsequent development of atherosclerotic plaques, which are causes of various pathological cardiac events. An overview for a causative relationship between ROS and endothelial inflammation will be provided in this review. Particularly, a crucial role for specific protein SUMOylation in endothelial inflammation will be presented. Given that SUMOylation of specific proteins leads to increased endothelial inflammation, targeting specific SUMOylated proteins may be an elegant, effective strategy to control inflammation. In addition, the involvement of ROS production in increasing the risk of recurrent coronary events in a sub-group of non-diabetic, post-infarction patients with elevated levels of HDL-cholesterol will be presented with the emphasis that elevated HDL-cholesterol under certain inflammatory conditions can lead to increased incidence of cardiovascular events.
Collapse
|
87
|
Wang XQ, Nigro P, World C, Fujiwara K, Yan C, Berk BC. Thioredoxin interacting protein promotes endothelial cell inflammation in response to disturbed flow by increasing leukocyte adhesion and repressing Kruppel-like factor 2. Circ Res 2012; 110:560-8. [PMID: 22267843 DOI: 10.1161/circresaha.111.256362] [Citation(s) in RCA: 72] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
RATIONALE Endothelial cells (EC) at regions exposed to disturbed flow (d-flow) are predisposed to inflammation and the subsequent development of atherosclerosis. We previously showed that thioredoxin interacting protein (TXNIP) was required for tumor necrosis factor-mediated expression of vascular cell adhesion molecule-1. OBJECTIVE We sought to investigate the role of TXNIP in d-flow-induced cell adhesion molecule expression and leukocyte interaction with vessels, and the mechanisms by which TXNIP suppresses athero-protective gene expression. METHODS AND RESULTS Using en face staining of mouse aorta, we found a dramatic increase of TXNIP in EC at sites exposed to d-flow as compared to steady flow. EC-specific TXNIP (EC-TXNIP) knockout mice showed significant decreases in vascular cell adhesion molecule-1 and intercellular adhesion molecule-1 mRNA expression in the d-flow regions of mouse aorta. Intravital microscopy of mesenteric venules showed that leukocyte rolling time was decreased, whereas rolling velocity was increased significantly in EC-TXNIP knockout mice. In vitro experiments using a cutout flow chamber to generate varying flow patterns showed that increased TXNIP was required for d-flow-induced EC-monocyte adhesion. Furthermore, we found that the expression of Kruppel-like factor 2, a key anti-inflammatory transcription factor in EC, was inhibited by TXNIP. Luciferase and chromatin immunoprecipitation assays showed that TXNIP was present within a repressing complex on the Kruppel-like factor 2 promoter. CONCLUSIONS These data demonstrate the essential role for TXNIP in mediating EC-leukocyte adhesion under d-flow, as well as define a novel mechanism by which TXNIP acts as a transcriptional corepressor to regulate Kruppel-like factor 2-dependent gene expression.
Collapse
Affiliation(s)
- Xiao-Qun Wang
- Aab Cardiovascular Research Institute, 601 Elmwood Avenue, University of Rochester School of Medicine and Dentistry, Rochester, NY 14642, USA
| | | | | | | | | | | |
Collapse
|
88
|
Lim JH, Woo CH. Laminar flow activation of ERK5 leads to cytoprotective effect via CHIP-mediated p53 ubiquitination in endothelial cells. Anat Cell Biol 2011; 44:265-73. [PMID: 22254155 PMCID: PMC3254880 DOI: 10.5115/acb.2011.44.4.265] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2011] [Revised: 11/21/2011] [Accepted: 11/23/2011] [Indexed: 12/04/2022] Open
Abstract
Atherosclerosis is readily observed in areas where disturbed flow is formed, while the atheroprotective region is found in areas with steady laminar flow (L-flow). It has been established that L-flow protects endothelial cells against endothelial dysfunction, including apoptosis and inflammation. It has also been reported that extracellular signal-regulated kinase 5 (ERK5) regulated endothelial integrity and protected endothelial cells from vascular dysfunction and disease under L-flow. However, the molecular mechanism by which L-flow-induced ERK5 activation inhibits endothelial apoptosis has not yet been determined. Transcription factor p53 is a major pro-apoptotic factor which contributes to apoptosis in various cell types. In this study, we found that 15-deoxy-Δ(12,14)-prostaglandin J2 induced p53 expression and that endothelial apoptosis was reduced under the L-flow condition. This anti-apoptotic response was reversed by the biochemical inhibition of ERK5 activation. It was also found that activation of ERK5 protected endothelial apoptosis in a C terminus of Hsc70-interacting protein (CHIP) ubiquitin ligase-dependent manner. Moreover, molecular interaction between ERK5-CHIP and p53 ubiquitination were addressed with a CHIP ubiquitin ligase activity assay. Taken together, our data suggest that the ERK5-CHIP signal module elicited by L-flow plays an important role in the anti-apoptotic mechanism in endothelial cells.
Collapse
Affiliation(s)
- Jae Hyang Lim
- Department of Biology and Center for Inflammation, Immunity and Infection, Georgia State University, Atlanta, GA, USA
| | | |
Collapse
|
89
|
Heo KS, Fujiwara K, Abe JI. Disturbed-flow-mediated vascular reactive oxygen species induce endothelial dysfunction. Circ J 2011; 75:2722-30. [PMID: 22076424 DOI: 10.1253/circj.cj-11-1124] [Citation(s) in RCA: 69] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Emerging evidence is revealing the different roles of steady laminar flow (s-flow) and disturbed flow (d-flow) in the regulation of the vascular endothelium. s-flow is atheroprotective while d-flow creates an atheroprone environment. Most recently, we found unique atheroprone signals, which involve protein kinase C (PKC)ζ activation, elicited by d-flow. We and others have defined a novel role for PKCζ as a shared mediator for tumor necrosis factor alpha (TNF alpha) and d-flow, which cause pro-inflammatory and pro-apoptotic events in endothelial cells (ECs) in the atheroprone environment. Under such conditions, ONOO(-) formation is increased in a d-flow-mediated PKCζ-dependent manner. Here, we propose a new signaling pathway involving d-flow-induced EC inflammation via PKCζ-ERK5 interaction-mediated downregulation of KLF2/eNOS stability, which leads to PKCζ-mediated p53-SUMOylation and EC apoptosis. In addition, we highlight several mechanisms contributing to endothelial dysfunction, focusing on the relations between flow patterns and activation of reactive oxygen species generating enzymes.
Collapse
Affiliation(s)
- Kyung-Sun Heo
- Aab Cardiovascular Research Institute, University of Rochester, NY, USA
| | | | | |
Collapse
|
90
|
Stindt MH, Carter S, Vigneron AM, Ryan KM, Vousden KH. MDM2 promotes SUMO-2/3 modification of p53 to modulate transcriptional activity. Cell Cycle 2011; 10:3176-88. [PMID: 21900752 PMCID: PMC3218624 DOI: 10.4161/cc.10.18.17436] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2011] [Revised: 07/28/2011] [Accepted: 07/28/2011] [Indexed: 01/02/2023] Open
Abstract
The tumor suppressor p53 is extensively regulated by post-translational modification, including modification by the small ubiquitin-related modifier SUMO. We show here that MDM2, previously shown to promote ubiquitin, Nedd8 and SUMO-1 modification of p53, can also enhance conjugation of endogenous SUMO-2/3 to p53. Sumoylation activity requires p53-MDM2 binding but does not depend on an intact RING finger. Both ARF and L11 can promote SUMO-2/3 conjugation of p53. However, unlike the previously described SUMO-1 conjugation of p53 by an MDM2-ARF complex, this activity does not depend on the ability of MDM2 to relocalize to the nucleolus. Interestingly, the SUMO consensus is not conserved in mouse p53, which is therefore not modified by SUMO-2/3. Finally, we show that conjugation of SUMO-2/3 to p53 correlates with a reduction of both activation and repression of a subset of p53-target genes.
Collapse
|
91
|
Takabe W, Alberts-Grill N, Jo H. Disturbed flow: p53 SUMOylation in the turnover of endothelial cells. ACTA ACUST UNITED AC 2011; 193:805-7. [PMID: 21624951 PMCID: PMC3105545 DOI: 10.1083/jcb.201104140] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Disturbed blood flow induces apoptosis of vascular endothelial cells, which causes atherosclerosis. In this issue, Heo et al. (2011. J. Cell Biol. doi:10.1083/jcb.201010051) sheds light on p53’s role in this phenomenon. Disturbed flow induces peroxynitrite production, which activates protein kinase C ζ and it’s binding to the E3 SUMO (small ubiquitin-like modifier) ligase PIASy (protein inhibitor of activated STATy). This leads to p53 SUMOylation and its export to the cytosol, where it binds to the antiapoptotic protein Bcl-2 to induce apoptosis.
Collapse
Affiliation(s)
- Wakako Takabe
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA 30322, USA
| | | | | |
Collapse
|