1
|
Jin YJ, Liang G, Li R, Wang S, Alnouri MW, Bentsen M, Kuenne C, Günther S, Yan Y, Li Y, Wettschureck N, Offermanns S. Phosphorylation of endothelial histone H3.3 serine 31 by PKN1 links flow-induced signaling to proatherogenic gene expression. NATURE CARDIOVASCULAR RESEARCH 2025; 4:180-196. [PMID: 39779823 PMCID: PMC11825370 DOI: 10.1038/s44161-024-00593-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Accepted: 11/29/2024] [Indexed: 01/11/2025]
Abstract
Atherosclerotic lesions develop preferentially in arterial regions exposed to disturbed blood flow, where endothelial cells acquire an inflammatory phenotype. How disturbed flow induces endothelial cell inflammation is incompletely understood. Here we show that histone H3.3 phosphorylation at serine 31 (H3.3S31) regulates disturbed-flow-induced endothelial inflammation by allowing rapid induction of FOS and FOSB, required for inflammatory gene expression. We identified protein kinase N1 (PKN1) as the kinase responsible for disturbed-flow-induced H3.3S31 phosphorylation. Disturbed flow activates PKN1 in an integrin α5β1-dependent manner and induces its translocation into the nucleus, and PKN1 is also involved in the phosphorylation of the AP-1 transcription factor JUN. Mice with endothelium-specific PKN1 loss or endothelial expression of S31 phosphorylation-deficient H.3.3 mutants show reduced endothelial inflammation and disturbed-flow-induced vascular remodeling in vitro and in vivo. Together, we identified a pathway whereby disturbed flow through PKN1-mediated histone phosphorylation and FOS/FOSB induction promotes inflammatory gene expression and vascular inflammation.
Collapse
Affiliation(s)
- Young-June Jin
- Department of Pharmacology, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany.
| | - Guozheng Liang
- Department of Pharmacology, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Rui Li
- Department of Pharmacology, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - ShengPeng Wang
- Department of Pharmacology, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
- Department of Cardiovascular Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Mohamad Wessam Alnouri
- Department of Pharmacology, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Mette Bentsen
- Bioinformatics and Deep Sequencing Platform, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Carsten Kuenne
- Bioinformatics and Deep Sequencing Platform, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Stefan Günther
- Bioinformatics and Deep Sequencing Platform, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Yang Yan
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Yongxin Li
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Nina Wettschureck
- Department of Pharmacology, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
- Centre for Molecular Medicine, Medical Faculty, JW Goethe University Frankfurt, Frankfurt, Germany
- Cardiopulmonary Institute (CPI), Frankfurt, Germany
- German Center for Cardiovascular Research (DZHK), Rhine-Main site, Frankfurt and Bad Nauheim, Germany
| | - Stefan Offermanns
- Department of Pharmacology, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany.
- Centre for Molecular Medicine, Medical Faculty, JW Goethe University Frankfurt, Frankfurt, Germany.
- Cardiopulmonary Institute (CPI), Frankfurt, Germany.
- German Center for Cardiovascular Research (DZHK), Rhine-Main site, Frankfurt and Bad Nauheim, Germany.
| |
Collapse
|
2
|
Mierke CT. Mechanosensory entities and functionality of endothelial cells. Front Cell Dev Biol 2024; 12:1446452. [PMID: 39507419 PMCID: PMC11538060 DOI: 10.3389/fcell.2024.1446452] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2024] [Accepted: 10/04/2024] [Indexed: 11/08/2024] Open
Abstract
The endothelial cells of the blood circulation are exposed to hemodynamic forces, such as cyclic strain, hydrostatic forces, and shear stress caused by the blood fluid's frictional force. Endothelial cells perceive mechanical forces via mechanosensors and thus elicit physiological reactions such as alterations in vessel width. The mechanosensors considered comprise ion channels, structures linked to the plasma membrane, cytoskeletal spectrin scaffold, mechanoreceptors, and junctional proteins. This review focuses on endothelial mechanosensors and how they alter the vascular functions of endothelial cells. The current state of knowledge on the dysregulation of endothelial mechanosensitivity in disease is briefly presented. The interplay in mechanical perception between endothelial cells and vascular smooth muscle cells is briefly outlined. Finally, future research avenues are highlighted, which are necessary to overcome existing limitations.
Collapse
|
3
|
Fish A, Kulkarni A. Flow-Induced Shear Stress Primes NLRP3 Inflammasome Activation in Macrophages via Piezo1. ACS APPLIED MATERIALS & INTERFACES 2024; 16:4505-4518. [PMID: 38240257 DOI: 10.1021/acsami.3c18645] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/01/2024]
Abstract
The NLRP3 inflammasome is a crucial component of the innate immune system, playing a pivotal role in initiating and regulating the body's inflammatory response to various pathogens and cellular damage. Environmental stimuli, such as temperature, pH level, and nutrient availability, can influence the behavior and functions of innate immune cells, including immune cell activity, proliferation, and cytokine production. However, there is limited understanding regarding how mechanical forces, like shear stress, govern the intrinsic inflammatory reaction, particularly the activation of the NLRP3 inflammasome, and how shear stress impacts NLRP3 inflammasome activation through its capacity to induce alterations in gene expression and cytokine secretion. Here, we investigated how shear stress can act as a priming signal in NLRP3 inflammasome activation by exposing immortalized bone marrow-derived macrophages (iBMDMs) to numerous physiologically relevant magnitudes of shear stress before chemically inducing inflammasome activation. We demonstrated that shear stress of large magnitudes was able to prime iBMDMs more effectively for inflammasome activation compared to lower shear stress magnitudes, as quantified by the percentage of cells where ASC-CFP specks formed and IL-1β secretion, the hallmarks of inflammasome activation. Testing this in NLRP3 and caspase-1 knockout iBMDMs showed that the NLRP3 inflammasome was primarily primed for activation due to shear stress exposure. Quantitative polymerase chain reaction (qPCR) and a small-molecule inhibitor study mechanistically determined that shear stress regulates the NLRP3 inflammasome by upregulating Piezo1, IKKβ, and NLRP3. These findings offer insights into the mechanistic relationship among physiological shear stresses, inflammasome activation, and their impact on the progression of inflammatory diseases and their interconnected pathogenesis.
Collapse
Affiliation(s)
- Adam Fish
- Department of Chemical Engineering, University of Massachusetts, Amherst, Massachusetts 01003, United States
| | - Ashish Kulkarni
- Department of Chemical Engineering, University of Massachusetts, Amherst, Massachusetts 01003, United States
- Center for Bioactive Delivery, Institute for Applied Life Sciences, University of Massachusetts, Amherst, Massachusetts 01003, United States
| |
Collapse
|
4
|
Kuramoto K, Liang H, Hong JH, He C. Exercise-activated hepatic autophagy via the FN1-α5β1 integrin pathway drives metabolic benefits of exercise. Cell Metab 2023; 35:620-632.e5. [PMID: 36812915 PMCID: PMC10079584 DOI: 10.1016/j.cmet.2023.01.011] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/29/2022] [Revised: 12/01/2022] [Accepted: 01/26/2023] [Indexed: 02/24/2023]
Abstract
How exercise elicits systemic metabolic benefits in both muscles and non-contractile tissues is unclear. Autophagy is a stress-induced lysosomal degradation pathway that mediates protein and organelle turnover and metabolic adaptation. Exercise activates autophagy in not only contracting muscles but also non-contractile tissues including the liver. However, the role and mechanism of exercise-activated autophagy in non-contractile tissues remain mysterious. Here, we show that hepatic autophagy activation is essential for exercise-induced metabolic benefits. Plasma or serum from exercised mice is sufficient to activate autophagy in cells. By proteomic studies, we identify fibronectin (FN1), which was previously considered as an extracellular matrix protein, as an exercise-induced, muscle-secreted, autophagy-inducing circulating factor. Muscle-secreted FN1 mediates exercise-induced hepatic autophagy and systemic insulin sensitization via the hepatic receptor α5β1 integrin and the downstream IKKα/β-JNK1-BECN1 pathway. Thus, we demonstrate that hepatic autophagy activation drives exercise-induced metabolic benefits against diabetes via muscle-secreted soluble FN1 and hepatic α5β1 integrin signaling.
Collapse
Affiliation(s)
- Kenta Kuramoto
- Department of Cell and Developmental Biology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Huijia Liang
- Department of Cell and Developmental Biology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Jung-Hwa Hong
- Department of Cell and Developmental Biology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Congcong He
- Department of Cell and Developmental Biology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA.
| |
Collapse
|
5
|
Davis MJ, Earley S, Li YS, Chien S. Vascular mechanotransduction. Physiol Rev 2023; 103:1247-1421. [PMID: 36603156 PMCID: PMC9942936 DOI: 10.1152/physrev.00053.2021] [Citation(s) in RCA: 90] [Impact Index Per Article: 45.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Revised: 09/26/2022] [Accepted: 10/04/2022] [Indexed: 01/07/2023] Open
Abstract
This review aims to survey the current state of mechanotransduction in vascular smooth muscle cells (VSMCs) and endothelial cells (ECs), including their sensing of mechanical stimuli and transduction of mechanical signals that result in the acute functional modulation and longer-term transcriptomic and epigenetic regulation of blood vessels. The mechanosensors discussed include ion channels, plasma membrane-associated structures and receptors, and junction proteins. The mechanosignaling pathways presented include the cytoskeleton, integrins, extracellular matrix, and intracellular signaling molecules. These are followed by discussions on mechanical regulation of transcriptome and epigenetics, relevance of mechanotransduction to health and disease, and interactions between VSMCs and ECs. Throughout this review, we offer suggestions for specific topics that require further understanding. In the closing section on conclusions and perspectives, we summarize what is known and point out the need to treat the vasculature as a system, including not only VSMCs and ECs but also the extracellular matrix and other types of cells such as resident macrophages and pericytes, so that we can fully understand the physiology and pathophysiology of the blood vessel as a whole, thus enhancing the comprehension, diagnosis, treatment, and prevention of vascular diseases.
Collapse
Affiliation(s)
- Michael J Davis
- Department of Medical Pharmacology and Physiology, University of Missouri, Columbia, Missouri
| | - Scott Earley
- Department of Pharmacology, University of Nevada, Reno, Nevada
| | - Yi-Shuan Li
- Department of Bioengineering, University of California, San Diego, California
- Institute of Engineering in Medicine, University of California, San Diego, California
| | - Shu Chien
- Department of Bioengineering, University of California, San Diego, California
- Institute of Engineering in Medicine, University of California, San Diego, California
- Department of Medicine, University of California, San Diego, California
| |
Collapse
|
6
|
Pang X, He X, Qiu Z, Zhang H, Xie R, Liu Z, Gu Y, Zhao N, Xiang Q, Cui Y. Targeting integrin pathways: mechanisms and advances in therapy. Signal Transduct Target Ther 2023; 8:1. [PMID: 36588107 PMCID: PMC9805914 DOI: 10.1038/s41392-022-01259-6] [Citation(s) in RCA: 433] [Impact Index Per Article: 216.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Revised: 11/14/2022] [Accepted: 11/21/2022] [Indexed: 01/03/2023] Open
Abstract
Integrins are considered the main cell-adhesion transmembrane receptors that play multifaceted roles as extracellular matrix (ECM)-cytoskeletal linkers and transducers in biochemical and mechanical signals between cells and their environment in a wide range of states in health and diseases. Integrin functions are dependable on a delicate balance between active and inactive status via multiple mechanisms, including protein-protein interactions, conformational changes, and trafficking. Due to their exposure on the cell surface and sensitivity to the molecular blockade, integrins have been investigated as pharmacological targets for nearly 40 years, but given the complexity of integrins and sometimes opposite characteristics, targeting integrin therapeutics has been a challenge. To date, only seven drugs targeting integrins have been successfully marketed, including abciximab, eptifibatide, tirofiban, natalizumab, vedolizumab, lifitegrast, and carotegrast. Currently, there are approximately 90 kinds of integrin-based therapeutic drugs or imaging agents in clinical studies, including small molecules, antibodies, synthetic mimic peptides, antibody-drug conjugates (ADCs), chimeric antigen receptor (CAR) T-cell therapy, imaging agents, etc. A serious lesson from past integrin drug discovery and research efforts is that successes rely on both a deep understanding of integrin-regulatory mechanisms and unmet clinical needs. Herein, we provide a systematic and complete review of all integrin family members and integrin-mediated downstream signal transduction to highlight ongoing efforts to develop new therapies/diagnoses from bench to clinic. In addition, we further discuss the trend of drug development, how to improve the success rate of clinical trials targeting integrin therapies, and the key points for clinical research, basic research, and translational research.
Collapse
Affiliation(s)
- Xiaocong Pang
- grid.411472.50000 0004 1764 1621Department of Pharmacy, Peking University First Hospital, Xishiku Street, Xicheng District, 100034 Beijing, China ,grid.411472.50000 0004 1764 1621Institute of Clinical Pharmacology, Peking University First Hospital, Xueyuan Road 38, Haidian District, 100191 Beijing, China
| | - Xu He
- grid.411472.50000 0004 1764 1621Department of Pharmacy, Peking University First Hospital, Xishiku Street, Xicheng District, 100034 Beijing, China ,grid.411472.50000 0004 1764 1621Institute of Clinical Pharmacology, Peking University First Hospital, Xueyuan Road 38, Haidian District, 100191 Beijing, China
| | - Zhiwei Qiu
- grid.411472.50000 0004 1764 1621Department of Pharmacy, Peking University First Hospital, Xishiku Street, Xicheng District, 100034 Beijing, China ,grid.411472.50000 0004 1764 1621Institute of Clinical Pharmacology, Peking University First Hospital, Xueyuan Road 38, Haidian District, 100191 Beijing, China
| | - Hanxu Zhang
- grid.411472.50000 0004 1764 1621Department of Pharmacy, Peking University First Hospital, Xishiku Street, Xicheng District, 100034 Beijing, China ,grid.411472.50000 0004 1764 1621Institute of Clinical Pharmacology, Peking University First Hospital, Xueyuan Road 38, Haidian District, 100191 Beijing, China
| | - Ran Xie
- grid.411472.50000 0004 1764 1621Department of Pharmacy, Peking University First Hospital, Xishiku Street, Xicheng District, 100034 Beijing, China ,grid.411472.50000 0004 1764 1621Institute of Clinical Pharmacology, Peking University First Hospital, Xueyuan Road 38, Haidian District, 100191 Beijing, China
| | - Zhiyan Liu
- grid.411472.50000 0004 1764 1621Department of Pharmacy, Peking University First Hospital, Xishiku Street, Xicheng District, 100034 Beijing, China ,grid.411472.50000 0004 1764 1621Institute of Clinical Pharmacology, Peking University First Hospital, Xueyuan Road 38, Haidian District, 100191 Beijing, China
| | - Yanlun Gu
- grid.411472.50000 0004 1764 1621Department of Pharmacy, Peking University First Hospital, Xishiku Street, Xicheng District, 100034 Beijing, China ,grid.411472.50000 0004 1764 1621Institute of Clinical Pharmacology, Peking University First Hospital, Xueyuan Road 38, Haidian District, 100191 Beijing, China
| | - Nan Zhao
- grid.411472.50000 0004 1764 1621Department of Pharmacy, Peking University First Hospital, Xishiku Street, Xicheng District, 100034 Beijing, China ,grid.411472.50000 0004 1764 1621Institute of Clinical Pharmacology, Peking University First Hospital, Xueyuan Road 38, Haidian District, 100191 Beijing, China
| | - Qian Xiang
- Department of Pharmacy, Peking University First Hospital, Xishiku Street, Xicheng District, 100034, Beijing, China. .,Institute of Clinical Pharmacology, Peking University First Hospital, Xueyuan Road 38, Haidian District, 100191, Beijing, China.
| | - Yimin Cui
- Department of Pharmacy, Peking University First Hospital, Xishiku Street, Xicheng District, 100034, Beijing, China. .,Institute of Clinical Pharmacology, Peking University First Hospital, Xueyuan Road 38, Haidian District, 100191, Beijing, China.
| |
Collapse
|
7
|
He L, Zhang CL, Chen Q, Wang L, Huang Y. Endothelial shear stress signal transduction and atherogenesis: From mechanisms to therapeutics. Pharmacol Ther 2022; 235:108152. [PMID: 35122834 DOI: 10.1016/j.pharmthera.2022.108152] [Citation(s) in RCA: 70] [Impact Index Per Article: 23.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2021] [Revised: 01/13/2022] [Accepted: 01/27/2022] [Indexed: 10/19/2022]
Abstract
Atherosclerotic vascular disease and its complications are among the top causes of mortality worldwide. In the vascular lumen, atherosclerotic plaques are not randomly distributed. Instead, they are preferentially localized at the curvature and bifurcations along the arterial tree, where shear stress is low or disturbed. Numerous studies demonstrate that endothelial cell phenotypic change (e.g., inflammation, oxidative stress, endoplasmic reticulum stress, apoptosis, autophagy, endothelial-mesenchymal transition, endothelial permeability, epigenetic regulation, and endothelial metabolic adaptation) induced by oscillatory shear force play a fundamental role in the initiation and progression of atherosclerosis. Mechano-sensors, adaptor proteins, kinases, and transcriptional factors work closely at different layers to transduce the shear stress force from the plasma membrane to the nucleus in endothelial cells, thereby controlling the expression of genes that determine cell fate and phenotype. An in-depth understanding of these mechano-sensitive signaling cascades shall provide new translational strategies for therapeutic intervention of atherosclerotic vascular disease. This review updates the recent advances in endothelial mechano-transduction and its role in the pathogenesis of atherosclerosis, and highlights the perspective of new anti-atherosclerosis therapies through targeting these mechano-regulated signaling molecules.
Collapse
Affiliation(s)
- Lei He
- School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Cheng-Lin Zhang
- Department of Pathophysiology, School of Basic Medical Sciences, Shenzhen University Health Science Center, Shenzhen 518060, China; Shenzhen Research Institute, The Chinese University of Hong Kong, Hong Kong, China
| | - Qinghua Chen
- Department of Biomedical Sciences, City University of Hong Kong, Hong Kong, China
| | - Li Wang
- Department of Biomedical Sciences, City University of Hong Kong, Hong Kong, China
| | - Yu Huang
- Department of Biomedical Sciences, City University of Hong Kong, Hong Kong, China.
| |
Collapse
|
8
|
Liu M, Iosef C, Rao S, Domingo-Gonzalez R, Fu S, Snider P, Conway SJ, Umbach GS, Heilshorn SC, Dewi RE, Dahl MJ, Null DM, Albertine KH, Alvira CM. Transforming Growth Factor-induced Protein Promotes NF-κB-mediated Angiogenesis during Postnatal Lung Development. Am J Respir Cell Mol Biol 2021; 64:318-330. [PMID: 33264084 PMCID: PMC7909333 DOI: 10.1165/rcmb.2020-0153oc] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Accepted: 10/29/2020] [Indexed: 12/23/2022] Open
Abstract
Pulmonary angiogenesis is a key driver of alveolarization. Our prior studies showed that NF-κB promotes pulmonary angiogenesis during early alveolarization. However, the mechanisms regulating temporal-specific NF-κB activation in the pulmonary vasculature are unknown. To identify mechanisms that activate proangiogenic NF-κB signaling in the developing pulmonary vasculature, proteomic analysis of the lung secretome was performed using two-dimensional difference gel electrophoresis. NF-κB activation and angiogenic function was assessed in primary pulmonary endothelial cells (PECs) and TGFBI (transforming growth factor-β-induced protein)-regulated genes identified using RNA sequencing. Alveolarization and pulmonary angiogenesis was assessed in wild-type and Tgfbi null mice exposed to normoxia or hyperoxia. Lung TGFBI expression was determined in premature lambs supported by invasive and noninvasive respiratory support. Secreted factors from the early alveolar, but not the late alveolar or adult lung, promoted proliferation and migration in quiescent, adult PECs. Proteomic analysis identified TGFBI as one protein highly expressed by the early alveolar lung that promoted PEC migration by activating NF-κB via αvβ3 integrins. RNA sequencing identified Csf3 as a TGFBI-regulated gene that enhances nitric oxide production in PECs. Loss of TGFBI in mice exaggerated the impaired pulmonary angiogenesis induced by chronic hyperoxia, and TGFBI expression was disrupted in premature lambs with impaired alveolarization. Our studies identify TGFBI as a developmentally regulated protein that promotes NF-κB-mediated angiogenesis during early alveolarization by enhancing nitric oxide production. We speculate that dysregulation of TGFBI expression may contribute to diseases marked by impaired alveolar and vascular growth.
Collapse
Affiliation(s)
- Min Liu
- Department of Pediatrics, Center for Excellence in Pulmonary Biology, and
| | - Cristiana Iosef
- Department of Pediatrics, Center for Excellence in Pulmonary Biology, and
| | - Shailaja Rao
- Department of Pediatrics, Center for Excellence in Pulmonary Biology, and
| | | | - Sha Fu
- Department of Pediatrics, Center for Excellence in Pulmonary Biology, and
- Liuyang People’s Hospital, Hunan, China
| | - Paige Snider
- Herman B Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, Indiana
| | - Simon J. Conway
- Herman B Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, Indiana
| | - Gray S. Umbach
- Department of Pediatrics, Center for Excellence in Pulmonary Biology, and
- University of Texas Southwestern Medical School, Dallas, Texas; and
| | - Sarah C. Heilshorn
- Department of Materials Science and Engineering, Stanford University, Stanford, California
| | - Ruby E. Dewi
- Department of Materials Science and Engineering, Stanford University, Stanford, California
| | - Mar J. Dahl
- Department of Pediatrics, University of Utah School of Medicine, Salt Lake City, Utah
| | - Donald M. Null
- Department of Pediatrics, University of Utah School of Medicine, Salt Lake City, Utah
| | - Kurt H. Albertine
- Department of Pediatrics, University of Utah School of Medicine, Salt Lake City, Utah
| | - Cristina M. Alvira
- Department of Pediatrics, Center for Excellence in Pulmonary Biology, and
| |
Collapse
|
9
|
Wolpe AG, Ruddiman CA, Hall PJ, Isakson BE. Polarized Proteins in Endothelium and Their Contribution to Function. J Vasc Res 2021; 58:65-91. [PMID: 33503620 DOI: 10.1159/000512618] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Accepted: 10/27/2020] [Indexed: 12/11/2022] Open
Abstract
Protein localization in endothelial cells is tightly regulated to create distinct signaling domains within their tight spatial restrictions including luminal membranes, abluminal membranes, and interendothelial junctions, as well as caveolae and calcium signaling domains. Protein localization in endothelial cells is also determined in part by the vascular bed, with differences between arteries and veins and between large and small arteries. Specific protein polarity and localization is essential for endothelial cells in responding to various extracellular stimuli. In this review, we examine protein localization in the endothelium of resistance arteries, with occasional references to other vessels for contrast, and how that polarization contributes to endothelial function and ultimately whole organism physiology. We highlight the protein localization on the luminal surface, discussing important physiological receptors and the glycocalyx. The protein polarization to the abluminal membrane is especially unique in small resistance arteries with the presence of the myoendothelial junction, a signaling microdomain that regulates vasodilation, feedback to smooth muscle cells, and ultimately total peripheral resistance. We also discuss the interendothelial junction, where tight junctions, adherens junctions, and gap junctions all convene and regulate endothelial function. Finally, we address planar cell polarity, or axial polarity, and how this is regulated by mechanosensory signals like blood flow.
Collapse
Affiliation(s)
- Abigail G Wolpe
- Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, Virginia, USA.,Department of Cell Biology, University of Virginia School of Medicine, Charlottesville, Virginia, USA
| | - Claire A Ruddiman
- Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, Virginia, USA.,Department of Pharmacology, University of Virginia School of Medicine, Charlottesville, Virginia, USA
| | - Phillip J Hall
- Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, Virginia, USA
| | - Brant E Isakson
- Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, Virginia, USA, .,Department of Molecular Physiology and Biophysics, University of Virginia School of Medicine, Charlottesville, Virginia, USA,
| |
Collapse
|
10
|
Nakayama A, Albarrán-Juárez J, Liang G, Roquid KA, Iring A, Tonack S, Chen M, Müller OJ, Weinstein LS, Offermanns S. Disturbed flow-induced Gs-mediated signaling protects against endothelial inflammation and atherosclerosis. JCI Insight 2020; 5:140485. [PMID: 33268595 PMCID: PMC7714404 DOI: 10.1172/jci.insight.140485] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Accepted: 10/28/2020] [Indexed: 01/05/2023] Open
Abstract
Atherosclerosis develops preferentially in areas of the arterial system, in which blood flow is disturbed. Exposure of endothelial cells to disturbed flow has been shown to induce inflammatory signaling, including NF-κB activation, which leads to the expression of leukocyte adhesion molecules and chemokines. Here, we show that disturbed flow promotes the release of adrenomedullin from endothelial cells, which in turn activates its Gs-coupled receptor calcitonin receptor–like receptor (CALCRL). This induces antiinflammatory signaling through cAMP and PKA, and it results in reduced endothelial inflammation in vitro and in vivo. Suppression of endothelial expression of Gαs, the α subunit of the G-protein Gs; CALCRL; or adrenomedullin leads to increased disturbed flow–induced inflammatory signaling in vitro and in vivo. Furthermore, mice with induced endothelial-specific deficiency of Gαs, CALCRL, or adrenomedullin show increased atherosclerotic lesions. Our data identify an antiinflammatory signaling pathway in endothelial cells stimulated by disturbed flow and suggest activation of the endothelial adrenomedullin/CALCRL/Gs system as a promising approach to inhibit progression of atherosclerosis. Disturbed flow promotes the release of adrenomedullin from endothelial cells and activates Gs-mediated signaling, which reduces endothelial inflammation in vitro and in vivo.
Collapse
Affiliation(s)
- Akiko Nakayama
- Max Planck Institute for Heart and Lung Research, Department of Pharmacology, Bad Nauheim, Germany
| | - Julián Albarrán-Juárez
- Max Planck Institute for Heart and Lung Research, Department of Pharmacology, Bad Nauheim, Germany
| | - Guozheng Liang
- Max Planck Institute for Heart and Lung Research, Department of Pharmacology, Bad Nauheim, Germany
| | - Kenneth Anthony Roquid
- Max Planck Institute for Heart and Lung Research, Department of Pharmacology, Bad Nauheim, Germany
| | - András Iring
- Max Planck Institute for Heart and Lung Research, Department of Pharmacology, Bad Nauheim, Germany
| | - Sarah Tonack
- Max Planck Institute for Heart and Lung Research, Department of Pharmacology, Bad Nauheim, Germany
| | - Min Chen
- Metabolic Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases, NIH, Bethesda, Maryland, USA
| | - Oliver J Müller
- Department of Internal Medicine III, University of Kiel, Kiel, and German Center for Cardiovascular Research (DZHK), Partner site Hamburg/Kiel/Lübeck, Germany
| | - Lee S Weinstein
- Metabolic Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases, NIH, Bethesda, Maryland, USA
| | - Stefan Offermanns
- Max Planck Institute for Heart and Lung Research, Department of Pharmacology, Bad Nauheim, Germany.,Centre for Molecular Medicine, Medical Faculty, J.W. Goethe University Frankfurt, Frankfurt, Germany.,DZHK RheinMain, Germany
| |
Collapse
|
11
|
Affiliation(s)
- Catherine Demos
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology (C.D., D.W., H.J.), Emory University, Atlanta
| | - Darian Williams
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology (C.D., D.W., H.J.), Emory University, Atlanta
| | - Hanjoong Jo
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology (C.D., D.W., H.J.), Emory University, Atlanta
- Department of Medicine (H.J.), Emory University, Atlanta
| |
Collapse
|
12
|
Ward AO, Angelini GD, Caputo M, Evans PC, Johnson JL, Suleiman MS, Tulloh RM, George SJ, Zakkar M. NF-κB inhibition prevents acute shear stress-induced inflammation in the saphenous vein graft endothelium. Sci Rep 2020; 10:15133. [PMID: 32934266 PMCID: PMC7492228 DOI: 10.1038/s41598-020-71781-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2019] [Accepted: 08/14/2020] [Indexed: 12/17/2022] Open
Abstract
The long saphenous vein (LSV) is commonly used as a conduit in coronary artery bypass grafting. However, long term patency remains limited by the development of vascular inflammation, intimal hyperplasia and accelerated atherosclerosis. The impact of acute exposure of venous endothelial cells (ECs) to acute arterial wall shear stress (WSS) in the arterial circulation, and the subsequent activation of inflammatory pathways, remain poorly defined. Here, we tested the hypothesis that acute exposure of venous ECs to high shear stress is associated with inflammatory responses that are regulated by NF-κB both in-vitro and ex-vivo. Analysis of the LSV endothelium revealed that activation of NF-κB occurred within 30 min after exposure to arterial rates of shear stress. Activation of NF-κB was associated with increased levels of CCL2 production and enhanced binding of monocytes in LSVECs exposed to 6 h acute arterial WSS. Consistent with this, ex vivo exposure of LSVs to acute arterial WSS promoted monocyte interactions with the vessel lumen. Inhibition of the NF-κB pathway prevented acute arterial WSS-induced CCL2 production and reduced monocyte adhesion, both in vitro and in human LSV ex vivo, demonstrating that this pathway is necessary for the induction of the acute arterial WSS-induced pro-inflammatory response. We have identified NF-κB as a critical regulator of acute endothelial inflammation in saphenous vein in response to acute arterial WSS. Localised endothelial-specific inhibition of the NF-κB pathway may be beneficial to prevent vein graft inflammation and consequent failure.
Collapse
Affiliation(s)
- Alexander O Ward
- Bristol Medical School, University of Bristol, Research Floor Level 7, Queens' Building, Bristol Royal Infirmary, Upper Maudlin Street, Bristol, BS2 8HW, UK
| | - Gianni D Angelini
- Bristol Medical School, University of Bristol, Research Floor Level 7, Queens' Building, Bristol Royal Infirmary, Upper Maudlin Street, Bristol, BS2 8HW, UK
| | - Massimo Caputo
- Bristol Medical School, University of Bristol, Research Floor Level 7, Queens' Building, Bristol Royal Infirmary, Upper Maudlin Street, Bristol, BS2 8HW, UK
| | - Paul C Evans
- Department of Infection, Immunity and Cardiovascular Diseases, University of Sheffield, Sheffield, S10 2TN, UK
| | - Jason L Johnson
- Bristol Medical School, University of Bristol, Research Floor Level 7, Queens' Building, Bristol Royal Infirmary, Upper Maudlin Street, Bristol, BS2 8HW, UK
| | - M Saadeh Suleiman
- Bristol Medical School, University of Bristol, Research Floor Level 7, Queens' Building, Bristol Royal Infirmary, Upper Maudlin Street, Bristol, BS2 8HW, UK
| | - Robert M Tulloh
- Bristol Medical School, University of Bristol, Research Floor Level 7, Queens' Building, Bristol Royal Infirmary, Upper Maudlin Street, Bristol, BS2 8HW, UK
| | - Sarah J George
- Bristol Medical School, University of Bristol, Research Floor Level 7, Queens' Building, Bristol Royal Infirmary, Upper Maudlin Street, Bristol, BS2 8HW, UK
| | - Mustafa Zakkar
- Bristol Medical School, University of Bristol, Research Floor Level 7, Queens' Building, Bristol Royal Infirmary, Upper Maudlin Street, Bristol, BS2 8HW, UK.
| |
Collapse
|
13
|
Recruitment and maturation of the coronary collateral circulation: Current understanding and perspectives in arteriogenesis. Microvasc Res 2020; 132:104058. [PMID: 32798552 DOI: 10.1016/j.mvr.2020.104058] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2020] [Revised: 06/09/2020] [Accepted: 08/11/2020] [Indexed: 12/13/2022]
Abstract
The coronary collateral circulation is a rich anastomotic network of primitive vessels which have the ability to augment in size and function through the process of arteriogenesis. In this review, we evaluate the current understandings of the molecular and cellular mechanisms by which this process occurs, specifically focussing on elevated fluid shear stress (FSS), inflammation, the redox state and gene expression along with the integrative, parallel and simultaneous process by which this occurs. The initiating step of arteriogenesis occurs following occlusion of an epicardial coronary artery, with an increase in FSS detected by mechanoreceptors within the endothelium. This must occur within a 'redox window' where an equilibrium of oxidative and reductive factors are present. These factors initially result in an inflammatory milieu, mediated by neutrophils as well as lymphocytes, with resultant activation of a number of downstream molecular pathways resulting in increased expression of proteins involved in monocyte attraction and adherence; namely vascular cell adhesion molecule 1 (VCAM-1), monocyte chemoattractant protein 1 (MCP-1) and transforming growth factor beta (TGF-β). Once monocytes and other inflammatory cells adhere to the endothelium they enter the extracellular matrix and differentiate into macrophages in an effort to create a favourable environment for vessel growth and development. Activated macrophages secrete inflammatory cytokines such as tumour necrosis factor-α (TNF-α), growth factors such as fibroblast growth factor-2 (FGF-2) and matrix metalloproteinases. Finally, vascular smooth muscle cells proliferate and switch to a contractile phenotype, resulting in an increased diameter and functionality of the collateral vessel, thereby allowing improved perfusion of the distal myocardium subtended by the occluded vessel. This simultaneously reduces FSS within the collateral vessel, inhibiting further vessel growth.
Collapse
|
14
|
Zhang C, Zhou T, Chen Z, Yan M, Li B, Lv H, Wang C, Xiang S, Shi L, Zhu Y, Ai D. Coupling of Integrin α5 to Annexin A2 by Flow Drives Endothelial Activation. Circ Res 2020; 127:1074-1090. [PMID: 32673515 DOI: 10.1161/circresaha.120.316857] [Citation(s) in RCA: 74] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
RATIONALE Atherosclerosis preferentially occurs at specific sites of the vasculature where endothelial cells (ECs) are exposed to disturbed blood flow. Translocation of integrin α5 to lipid rafts promotes integrin activation and ligation, which is critical for oscillatory shear stress (OSS)-induced EC activation. However, the underlying mechanism of OSS promoted integrin α5 lipid raft translocation has remained largely unknown. OBJECTIVE The objective of this study was to specify the mechanotransduction mechanism of OSS-induced integrin α5 translocation and subsequent EC activation. METHODS AND RESULTS Mass spectrometry studies identified endothelial ANXA2 (annexin A2) as a potential carrier allowing integrin α5β1 to traffic in response to OSS. Interference by siRNA of AnxA2 in ECs greatly decreased OSS-induced integrin α5β1 translocation to lipid rafts, EC activation, and monocyte adhesion. Pharmacological and genetic inhibition of PTP1B (protein tyrosine phosphatase 1B) blunted OSS-induced integrin α5β1 activation, which is dependent on Piezo1-mediated calcium influx in ECs. Furthermore, ANXA2 was identified as a direct substrate of activated PTP1B by mass spectrometry. Using bioluminescence resonance energy transfer assay, PTP1B-dephosphorylated ANXA2 at Y24 was found to lead to conformational freedom of the C-terminal core domain from the N-terminal domain of ANXA2. Immunoprecipitation assays showed that this unmasked ANXA2-C-terminal core domain specifically binds to an integrin α5 nonconserved cytoplasmic domain but not β1. Importantly, ectopic lentiviral overexpression of an ANXA2Y24F mutant increased and shRNA against Ptp1B decreased integrin α5β1 ligation, inflammatory signaling, and progression of plaques at atheroprone sites in apolipoprotein E (ApoE)-/- mice. However, the antiatherosclerotic effect of Ptp1B shRNA was abolished in AnxA2-/-ApoE-/- mice. CONCLUSIONS Our data elucidate a novel endothelial mechanotransduction molecular mechanism linking atheroprone flow and activation of integrin α5β1, thereby identifying a class of potential therapeutic targets for atherosclerosis. Graphic Abstract: An graphic abstract is available for this article.
Collapse
Affiliation(s)
- Chenghu Zhang
- State Key Laboratory of Experimental Hematology, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education) and Department of Physiology and Pathophysiology, Tianjin Key Laboratory of Metabolic Diseases (C.Z., T.Z., Z.C., M.Y., B.L., H.L., C.W., Y.Z., D.A.), Tianjin Medical University, China
| | - Ting Zhou
- State Key Laboratory of Experimental Hematology, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education) and Department of Physiology and Pathophysiology, Tianjin Key Laboratory of Metabolic Diseases (C.Z., T.Z., Z.C., M.Y., B.L., H.L., C.W., Y.Z., D.A.), Tianjin Medical University, China
| | - Zhipeng Chen
- State Key Laboratory of Experimental Hematology, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education) and Department of Physiology and Pathophysiology, Tianjin Key Laboratory of Metabolic Diseases (C.Z., T.Z., Z.C., M.Y., B.L., H.L., C.W., Y.Z., D.A.), Tianjin Medical University, China
| | - Meng Yan
- State Key Laboratory of Experimental Hematology, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education) and Department of Physiology and Pathophysiology, Tianjin Key Laboratory of Metabolic Diseases (C.Z., T.Z., Z.C., M.Y., B.L., H.L., C.W., Y.Z., D.A.), Tianjin Medical University, China
| | - Bochuan Li
- State Key Laboratory of Experimental Hematology, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education) and Department of Physiology and Pathophysiology, Tianjin Key Laboratory of Metabolic Diseases (C.Z., T.Z., Z.C., M.Y., B.L., H.L., C.W., Y.Z., D.A.), Tianjin Medical University, China
| | - Huizhen Lv
- State Key Laboratory of Experimental Hematology, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education) and Department of Physiology and Pathophysiology, Tianjin Key Laboratory of Metabolic Diseases (C.Z., T.Z., Z.C., M.Y., B.L., H.L., C.W., Y.Z., D.A.), Tianjin Medical University, China.,National Clinical Research Center for Blood Diseases; Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China (H.L., L.S., D.A.)
| | - Chunjiong Wang
- State Key Laboratory of Experimental Hematology, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education) and Department of Physiology and Pathophysiology, Tianjin Key Laboratory of Metabolic Diseases (C.Z., T.Z., Z.C., M.Y., B.L., H.L., C.W., Y.Z., D.A.), Tianjin Medical University, China
| | - Song Xiang
- Department of Biochemistry and Molecular Biology (S.X., L.S.), Tianjin Medical University, China
| | - Lei Shi
- Department of Biochemistry and Molecular Biology (S.X., L.S.), Tianjin Medical University, China.,National Clinical Research Center for Blood Diseases; Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China (H.L., L.S., D.A.)
| | - Yi Zhu
- State Key Laboratory of Experimental Hematology, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education) and Department of Physiology and Pathophysiology, Tianjin Key Laboratory of Metabolic Diseases (C.Z., T.Z., Z.C., M.Y., B.L., H.L., C.W., Y.Z., D.A.), Tianjin Medical University, China
| | - Ding Ai
- State Key Laboratory of Experimental Hematology, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education) and Department of Physiology and Pathophysiology, Tianjin Key Laboratory of Metabolic Diseases (C.Z., T.Z., Z.C., M.Y., B.L., H.L., C.W., Y.Z., D.A.), Tianjin Medical University, China.,National Clinical Research Center for Blood Diseases; Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China (H.L., L.S., D.A.)
| |
Collapse
|
15
|
He M, Martin M, Marin T, Chen Z, Gongol B. Endothelial mechanobiology. APL Bioeng 2020; 4:010904. [PMID: 32095737 PMCID: PMC7032971 DOI: 10.1063/1.5129563] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2019] [Accepted: 01/19/2020] [Indexed: 12/11/2022] Open
Abstract
Lining the luminal surface of the vasculature, endothelial cells (ECs) are in direct
contact with and differentially respond to hemodynamic forces depending on their anatomic
location. Pulsatile shear stress (PS) is defined by laminar flow and is predominantly
located in straight vascular regions, while disturbed or oscillatory shear stress (OS) is
localized to branch points and bifurcations. Such flow patterns have become a central
focus of vascular diseases, such as atherosclerosis, because the focal distribution of
endothelial dysfunction corresponds to regions exposed to OS, whereas endothelial
homeostasis is maintained in regions defined by PS. Deciphering the mechanotransduction
events that occur in ECs in response to differential flow patterns has required the
innovation of multidisciplinary approaches in both in vitro and
in vivo systems. The results from these studies have identified a
multitude of shear stress-regulated molecular networks in the endothelium that are
implicated in health and disease. This review outlines the significance of scientific
findings generated in collaboration with Dr. Shu Chien.
Collapse
Affiliation(s)
- Ming He
- Department of Medicine, University of California, San Diego, California 92093, USA
| | - Marcy Martin
- Department of Pediatrics, Stanford University School of Medicine, Stanford, California 94305, USA
| | - Traci Marin
- Department of Health Sciences, Victor Valley College, Victorville, California 92395, USA
| | - Zhen Chen
- Department of Diabetes Complications and Metabolism, Beckman Research Institute, City of Hope, California 91010, USA
| | - Brendan Gongol
- Department of Medicine, University of California, San Diego, California 92093, USA
| |
Collapse
|
16
|
Wettschureck N, Strilic B, Offermanns S. Passing the Vascular Barrier: Endothelial Signaling Processes Controlling Extravasation. Physiol Rev 2019; 99:1467-1525. [PMID: 31140373 DOI: 10.1152/physrev.00037.2018] [Citation(s) in RCA: 195] [Impact Index Per Article: 32.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
A central function of the vascular endothelium is to serve as a barrier between the blood and the surrounding tissue of the body. At the same time, solutes and cells have to pass the endothelium to leave or to enter the bloodstream to maintain homeostasis. Under pathological conditions, for example, inflammation, permeability for fluid and cells is largely increased in the affected area, thereby facilitating host defense. To appropriately function as a regulated permeability filter, the endothelium uses various mechanisms to allow solutes and cells to pass the endothelial layer. These include transcellular and paracellular pathways of which the latter requires remodeling of intercellular junctions for its regulation. This review provides an overview on endothelial barrier regulation and focuses on the endothelial signaling mechanisms controlling the opening and closing of paracellular pathways for solutes and cells such as leukocytes and metastasizing tumor cells.
Collapse
Affiliation(s)
- Nina Wettschureck
- Department of Pharmacology, Max Planck Institute for Heart and Lung Research , Bad Nauheim , Germany ; and Centre for Molecular Medicine, Medical Faculty, J.W. Goethe University Frankfurt , Frankfurt , Germany
| | - Boris Strilic
- Department of Pharmacology, Max Planck Institute for Heart and Lung Research , Bad Nauheim , Germany ; and Centre for Molecular Medicine, Medical Faculty, J.W. Goethe University Frankfurt , Frankfurt , Germany
| | - Stefan Offermanns
- Department of Pharmacology, Max Planck Institute for Heart and Lung Research , Bad Nauheim , Germany ; and Centre for Molecular Medicine, Medical Faculty, J.W. Goethe University Frankfurt , Frankfurt , Germany
| |
Collapse
|
17
|
Ikedo T, Kataoka H, Minami M, Hayashi K, Miyata T, Nagata M, Fujikawa R, Yokode M, Imai H, Matsuda T, Miyamoto S. Sequential Inward Bending of Arterial Bifurcations is Associated with Intracranial Aneurysm Formation. World Neurosurg 2019; 129:e361-e366. [PMID: 31176059 DOI: 10.1016/j.wneu.2019.05.153] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2019] [Revised: 05/15/2019] [Accepted: 05/16/2019] [Indexed: 10/26/2022]
Abstract
OBJECTIVE To investigate the association between vascular morphology and the development of intracranial aneurysms (IAs), the morphological changes of intracranial arteries after IA induction were examined using a rodent model. METHODS The vascular morphology of the circle of Willis in rats was visualized at 1 week and at 3 months after IA induction using 7-T magnetic resonance imaging. The following 2 angle parameters were defined: the angle between the parent artery and the daughter arteries (PD angle), and the widening of the daughter arteries (DD angle). The correlations of the angle parameters with IA size and with the number of macrophages infiltrated in the IA wall by immunohistochemistry were examined. RESULTS Magnetic resonance imaging showed bending of the arteries over time around the predilection site for IAs. The PD angle increased significantly 1 week after IA induction (P < 0.05) and correlated with IA size (P < 0.01). The DD angle did not increase after 1 week, but increased 3 months after IA induction (P < 0.01). The PD angle 1 week after surgery also correlated with the number of infiltrated macrophages in aneurysmal walls (P = 0.01). CONCLUSIONS Sequential inward bending of arterial bifurcations occurred after IA induction in the rat model. The degree of arterial bending correlated with IA development and inflammation in the IA wall, suggesting that the vascular morphology may be strongly associated with IA development through a proinflammatory mechanism.
Collapse
Affiliation(s)
- Taichi Ikedo
- Department of Neurosurgery, Kyoto University Graduate School of Medicine, Kyoto, Japan; Department of Clinical Innovative Medicine, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Hiroharu Kataoka
- Department of Neurosurgery, Kyoto University Graduate School of Medicine, Kyoto, Japan.
| | - Manabu Minami
- Department of Clinical Innovative Medicine, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Kosuke Hayashi
- Department of Neurosurgery, Kyoto University Graduate School of Medicine, Kyoto, Japan; Department of Clinical Innovative Medicine, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Takeshi Miyata
- Department of Neurosurgery, Kyoto University Graduate School of Medicine, Kyoto, Japan; Department of Clinical Innovative Medicine, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Manabu Nagata
- Department of Neurosurgery, Kyoto University Graduate School of Medicine, Kyoto, Japan; Department of Clinical Innovative Medicine, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Risako Fujikawa
- Department of Clinical Innovative Medicine, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Masayuki Yokode
- Department of Clinical Innovative Medicine, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Hirohiko Imai
- Department of Systems Science, Graduate School of Informatics, Kyoto University, Kyoto, Japan
| | - Tetsuya Matsuda
- Department of Systems Science, Graduate School of Informatics, Kyoto University, Kyoto, Japan
| | - Susumu Miyamoto
- Department of Neurosurgery, Kyoto University Graduate School of Medicine, Kyoto, Japan
| |
Collapse
|
18
|
Xanthis I, Souilhol C, Serbanovic-Canic J, Roddie H, Kalli AC, Fragiadaki M, Wong R, Shah DR, Askari JA, Canham L, Akhtar N, Feng S, Ridger V, Waltho J, Pinteaux E, Humphries MJ, Bryan MT, Evans PC. β1 integrin is a sensor of blood flow direction. J Cell Sci 2019; 132:jcs.229542. [PMID: 31076511 PMCID: PMC6589088 DOI: 10.1242/jcs.229542] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2019] [Accepted: 04/13/2019] [Indexed: 12/26/2022] Open
Abstract
Endothelial cell (EC) sensing of fluid shear stress direction is a critical determinant of vascular health and disease. Unidirectional flow induces EC alignment and vascular homeostasis, whereas bidirectional flow has pathophysiological effects. ECs express several mechanoreceptors that respond to flow, but the mechanism for sensing shear stress direction is poorly understood. We determined, by using in vitro flow systems and magnetic tweezers, that β1 integrin is a key sensor of force direction because it is activated by unidirectional, but not bidirectional, shearing forces. β1 integrin activation by unidirectional force was amplified in ECs that were pre-sheared in the same direction, indicating that alignment and β1 integrin activity has a feedforward interaction, which is a hallmark of system stability. En face staining and EC-specific genetic deletion studies in the murine aorta revealed that β1 integrin is activated and is essential for EC alignment at sites of unidirectional flow but is not activated at sites of bidirectional flow. In summary, β1 integrin sensing of unidirectional force is a key mechanism for decoding blood flow mechanics to promote vascular homeostasis.This article has an associated First Person interview with the first author of the paper.
Collapse
Affiliation(s)
- Ioannis Xanthis
- Department of Infection, Immunity and Cardiovascular Disease, INSIGNEO Institute for In Silico Medicine, and the Bateson Centre, University of Sheffield, Sheffield S10 2TN, UK
| | - Celine Souilhol
- Department of Infection, Immunity and Cardiovascular Disease, INSIGNEO Institute for In Silico Medicine, and the Bateson Centre, University of Sheffield, Sheffield S10 2TN, UK
| | - Jovana Serbanovic-Canic
- Department of Infection, Immunity and Cardiovascular Disease, INSIGNEO Institute for In Silico Medicine, and the Bateson Centre, University of Sheffield, Sheffield S10 2TN, UK
| | - Hannah Roddie
- Department of Infection, Immunity and Cardiovascular Disease, INSIGNEO Institute for In Silico Medicine, and the Bateson Centre, University of Sheffield, Sheffield S10 2TN, UK
| | - Antreas C Kalli
- Leeds Institute of Medical Research at St James's and Astbury Centre for Structural Molecular Biology, University of Leeds, Leeds LS2 9JT, UK
| | - Maria Fragiadaki
- Department of Infection, Immunity and Cardiovascular Disease, INSIGNEO Institute for In Silico Medicine, and the Bateson Centre, University of Sheffield, Sheffield S10 2TN, UK
| | - Raymond Wong
- Faculty of Biology, Medicine & Health, University of Manchester, Manchester M13 9PL, UK
| | - Dhruv R Shah
- Department of Infection, Immunity and Cardiovascular Disease, INSIGNEO Institute for In Silico Medicine, and the Bateson Centre, University of Sheffield, Sheffield S10 2TN, UK
| | - Janet A Askari
- Wellcome Trust Centre for Cell-Matrix Research, Faculty of Biology, Medicine & Health, University of Manchester, Manchester M13 9PL, UK
| | - Lindsay Canham
- Department of Infection, Immunity and Cardiovascular Disease, INSIGNEO Institute for In Silico Medicine, and the Bateson Centre, University of Sheffield, Sheffield S10 2TN, UK
| | - Nasreen Akhtar
- Department of Oncology and Metabolism, University of Sheffield, Sheffield S10 2TN, UK
| | - Shuang Feng
- Department of Infection, Immunity and Cardiovascular Disease, INSIGNEO Institute for In Silico Medicine, and the Bateson Centre, University of Sheffield, Sheffield S10 2TN, UK
| | - Victoria Ridger
- Department of Infection, Immunity and Cardiovascular Disease, INSIGNEO Institute for In Silico Medicine, and the Bateson Centre, University of Sheffield, Sheffield S10 2TN, UK
| | - Jonathan Waltho
- Department of Molecular Biology and Biotechnology, University of Sheffield, Sheffield S10 2TN, UK
| | - Emmanuel Pinteaux
- Faculty of Biology, Medicine & Health, University of Manchester, Manchester M13 9PL, UK
| | - Martin J Humphries
- Wellcome Trust Centre for Cell-Matrix Research, Faculty of Biology, Medicine & Health, University of Manchester, Manchester M13 9PL, UK
| | - Matthew T Bryan
- Department of Infection, Immunity and Cardiovascular Disease, INSIGNEO Institute for In Silico Medicine, and the Bateson Centre, University of Sheffield, Sheffield S10 2TN, UK
| | - Paul C Evans
- Department of Infection, Immunity and Cardiovascular Disease, INSIGNEO Institute for In Silico Medicine, and the Bateson Centre, University of Sheffield, Sheffield S10 2TN, UK
| |
Collapse
|
19
|
Albarrán-Juárez J, Iring A, Wang S, Joseph S, Grimm M, Strilic B, Wettschureck N, Althoff TF, Offermanns S. Piezo1 and G q/G 11 promote endothelial inflammation depending on flow pattern and integrin activation. J Exp Med 2018; 215:2655-2672. [PMID: 30194266 PMCID: PMC6170174 DOI: 10.1084/jem.20180483] [Citation(s) in RCA: 213] [Impact Index Per Article: 30.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2018] [Revised: 06/22/2018] [Accepted: 08/01/2018] [Indexed: 12/18/2022] Open
Abstract
Atherosclerosis preferentially develops in areas of disturbed flow. Albarrán-Juárez et al. provide evidence that this depends on at least two different endothelial mechanosignaling pathways, a flow direction-independent pathway involving Piezo1 and Gq/G11, as well as integrin signaling, which is only initiated in response to disturbed flow. The vascular endothelium is constantly exposed to mechanical forces, including fluid shear stress exerted by the flowing blood. Endothelial cells can sense different flow patterns and convert the mechanical signal of laminar flow into atheroprotective signals, including eNOS activation, whereas disturbed flow in atheroprone areas induces inflammatory signaling, including NF-κB activation. How endothelial cells distinguish different flow patterns is poorly understood. Here we show that both laminar and disturbed flow activate the same initial pathway involving the mechanosensitive cation channel Piezo1, the purinergic P2Y2 receptor, and Gq/G11-mediated signaling. However, only disturbed flow leads to Piezo1- and Gq/G11-mediated integrin activation resulting in focal adhesion kinase-dependent NF-κB activation. Mice with induced endothelium-specific deficiency of Piezo1 or Gαq/Gα11 show reduced integrin activation, inflammatory signaling, and progression of atherosclerosis in atheroprone areas. Our data identify critical steps in endothelial mechanotransduction, which distinguish flow pattern-dependent activation of atheroprotective and atherogenic endothelial signaling and suggest novel therapeutic strategies to treat inflammatory vascular disorders such as atherosclerosis.
Collapse
Affiliation(s)
- Julián Albarrán-Juárez
- Max Planck Institute for Heart and Lung Research, Department of Pharmacology, Bad Nauheim, Germany
| | - Andras Iring
- Max Planck Institute for Heart and Lung Research, Department of Pharmacology, Bad Nauheim, Germany
| | - ShengPeng Wang
- Max Planck Institute for Heart and Lung Research, Department of Pharmacology, Bad Nauheim, Germany
| | - Sayali Joseph
- Max Planck Institute for Heart and Lung Research, Department of Pharmacology, Bad Nauheim, Germany
| | - Myriam Grimm
- Max Planck Institute for Heart and Lung Research, Department of Pharmacology, Bad Nauheim, Germany
| | - Boris Strilic
- Max Planck Institute for Heart and Lung Research, Department of Pharmacology, Bad Nauheim, Germany
| | - Nina Wettschureck
- Max Planck Institute for Heart and Lung Research, Department of Pharmacology, Bad Nauheim, Germany.,Center for Molecular Medicine, Medical Faculty, J.W. Goethe University Frankfurt, Frankfurt, Germany.,German Center for Cardiovascular Research (DZHK)
| | - Till F Althoff
- Max Planck Institute for Heart and Lung Research, Department of Pharmacology, Bad Nauheim, Germany.,Charité - Universitätsmedizin Berlin, Department of Cardiology and Angiology, Campus Mitte, Berlin, Germany.,German Center for Cardiovascular Research (DZHK)
| | - Stefan Offermanns
- Max Planck Institute for Heart and Lung Research, Department of Pharmacology, Bad Nauheim, Germany .,Center for Molecular Medicine, Medical Faculty, J.W. Goethe University Frankfurt, Frankfurt, Germany.,German Center for Cardiovascular Research (DZHK)
| |
Collapse
|
20
|
Signorelli F, Sela S, Gesualdo L, Chevrel S, Tollet F, Pailler-Mattei C, Tacconi L, Turjman F, Vacca A, Schul DB. Hemodynamic Stress, Inflammation, and Intracranial Aneurysm Development and Rupture: A Systematic Review. World Neurosurg 2018; 115:234-244. [DOI: 10.1016/j.wneu.2018.04.143] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2018] [Revised: 04/18/2018] [Accepted: 04/19/2018] [Indexed: 10/17/2022]
|
21
|
Urner S, Kelly-Goss M, Peirce SM, Lammert E. Mechanotransduction in Blood and Lymphatic Vascular Development and Disease. ADVANCES IN PHARMACOLOGY (SAN DIEGO, CALIF.) 2017; 81:155-208. [PMID: 29310798 DOI: 10.1016/bs.apha.2017.08.009] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The blood and lymphatic vasculatures are hierarchical networks of vessels, which constantly transport fluids and, therefore, are exposed to a variety of mechanical forces. Considering the role of mechanotransduction is key for fully understanding how these vascular systems develop, function, and how vascular pathologies evolve. During embryonic development, for example, initiation of blood flow is essential for early vascular remodeling, and increased interstitial fluid pressure as well as initiation of lymph flow is needed for proper development and maturation of the lymphatic vasculature. In this review, we introduce specific mechanical forces that affect both the blood and lymphatic vasculatures, including longitudinal and circumferential stretch, as well as shear stress. In addition, we provide an overview of the role of mechanotransduction during atherosclerosis and secondary lymphedema, which both trigger tissue fibrosis.
Collapse
Affiliation(s)
- Sofia Urner
- Institute of Metabolic Physiology, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Molly Kelly-Goss
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA, United States
| | - Shayn M Peirce
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA, United States
| | - Eckhard Lammert
- Institute of Metabolic Physiology, Heinrich Heine University Düsseldorf, Düsseldorf, Germany; Institute for Beta Cell Biology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University, Düsseldorf, Germany.
| |
Collapse
|
22
|
Activation and inflammation of the venous endothelium in vein graft disease. Atherosclerosis 2017; 265:266-274. [PMID: 28865843 DOI: 10.1016/j.atherosclerosis.2017.08.023] [Citation(s) in RCA: 59] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/09/2017] [Revised: 07/19/2017] [Accepted: 08/23/2017] [Indexed: 01/13/2023]
Abstract
The long saphenous vein is the most commonly used conduit in coronary artery bypass graft (CABG) surgery when bypassing multiple diseased arteries; however, its use is complicated by the development of vascular inflammation, intimal hyperplasia and accelerated atherosclerosis leading to compromised graft efficacy. Despite refinement of surgical techniques to improve graft patency, late vein graft failure remains a significant problem. Moreover, there is a lack of pharmacological interventions proven to be effective in the treatment of late vein graft failure. A greater understanding of the molecular nature of the disease and the interactions between endothelial and smooth muscle cells as a result of alterations in local haemodynamics may assist with designing future beneficial pharmacological interventions. Venous endothelial cells (ECs) are physiologically adapted to chronic low shear stress; however, once the graft is implanted into the arterial circulation, they become suddenly exposed to acute high levels of shear stress. A small number of in vitro and ex vivo studies have demonstrated that acute high shear stress is associated with the activation of a pro-inflammatory profile in saphenous vein ECs, which may be mediated by mitogen-activated protein kinase (MAPK) and nuclear factor-κB (NF-κB) signalling pathways. The impact of acute changes in shear stress on venous ECs and the role of ECs in the development of intimal hyperplasia remains incomplete and is the subject of this review.
Collapse
|
23
|
Nakajima H, Mochizuki N. Flow pattern-dependent endothelial cell responses through transcriptional regulation. Cell Cycle 2017; 16:1893-1901. [PMID: 28820314 DOI: 10.1080/15384101.2017.1364324] [Citation(s) in RCA: 70] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022] Open
Abstract
Blood flow provides endothelial cells (ECs) lining the inside of blood vessels with mechanical stimuli as well as humoral stimuli. Fluid shear stress, the frictional force between flowing blood and ECs, is recognized as an essential mechanical cue for vascular growth, remodeling, and homeostasis. ECs differentially respond to distinct flow patterns. High laminar shear flow leads to inhibition of cell cycle progression and stabilizes vessels, whereas low shear flow or disturbed flow leads to increased turnover of ECs and inflammatory responses of ECs prone to atherogenic. These differences of EC responses dependent on flow pattern are mainly ascribed to distinct patterns of gene expression. In this review, we highlight flow pattern-dependent transcriptional regulation in ECs by focusing on KLF2 and NFκB, major transcription factors responding to laminar flow and disturbed flow, respectively. Moreover, we introduce roles of a new flow-responsive transcriptional co-regulator, YAP, in blood vessel maintenance and discuss how these transcriptional regulators are spatiotemporally regulated by flow and then regulate EC functions in normal and pathological conditions.
Collapse
Affiliation(s)
- Hiroyuki Nakajima
- a Department of Cell Biology, National Cerebral and Cardiovascular Center Research Institute , Suita , Osaka , Japan
| | - Naoki Mochizuki
- a Department of Cell Biology, National Cerebral and Cardiovascular Center Research Institute , Suita , Osaka , Japan.,b AMED-CREST. National Cerebral and Cardiovascular Center , Suita , Osaka , Japan
| |
Collapse
|
24
|
Abstract
Living cells and tissues experience physical forces and chemical stimuli in a human body. The process of converting mechanical forces into biochemical activities and gene expression is mechanochemical transduction or mechanotransduction. Significant advances have been made in understanding mechanotransduction at cellular and molecular levels over the last two decades. However, major challenges remain in elucidating how a living cell integrates signals from mechanotransduction with chemical signals to regulate gene expression and to generate coherent biological responses in living tissues in physiological conditions and diseases.
Collapse
Affiliation(s)
- Ning Wang
- School of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, Hubei 430074, China; Department of Mechanical Science and Engineering, College of Engineering, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, USA
| |
Collapse
|
25
|
Integrin signaling in atherosclerosis. Cell Mol Life Sci 2017; 74:2263-2282. [PMID: 28246700 DOI: 10.1007/s00018-017-2490-4] [Citation(s) in RCA: 97] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2016] [Revised: 01/24/2017] [Accepted: 02/15/2017] [Indexed: 02/07/2023]
Abstract
Atherosclerosis, a chronic lipid-driven inflammatory disease affecting large arteries, represents the primary cause of cardiovascular disease in the world. The local remodeling of the vessel intima during atherosclerosis involves the modulation of vascular cell phenotype, alteration of cell migration and proliferation, and propagation of local extracellular matrix remodeling. All of these responses represent targets of the integrin family of cell adhesion receptors. As such, alterations in integrin signaling affect multiple aspects of atherosclerosis, from the earliest induction of inflammation to the development of advanced fibrotic plaques. Integrin signaling has been shown to regulate endothelial phenotype, facilitate leukocyte homing, affect leukocyte function, and drive smooth muscle fibroproliferative remodeling. In addition, integrin signaling in platelets contributes to the thrombotic complications that typically drive the clinical manifestation of cardiovascular disease. In this review, we examine the current literature on integrin regulation of atherosclerotic plaque development and the suitability of integrins as potential therapeutic targets to limit cardiovascular disease and its complications.
Collapse
|
26
|
Yurdagul A, Orr AW. Blood Brothers: Hemodynamics and Cell-Matrix Interactions in Endothelial Function. Antioxid Redox Signal 2016; 25:415-34. [PMID: 26715135 PMCID: PMC5011636 DOI: 10.1089/ars.2015.6525] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/08/2015] [Revised: 11/25/2015] [Accepted: 12/23/2015] [Indexed: 12/29/2022]
Abstract
SIGNIFICANCE Alterations in endothelial function contribute to a variety of vascular diseases. In pathological conditions, the endothelium shows a reduced ability to regulate vasodilation (endothelial dysfunction) and a conversion toward a proinflammatory and leaky phenotype (endothelial activation). At the interface between the vessel wall and blood, the endothelium exists in a complex microenvironment and must translate changes in these environmental signals to alterations in vessel function. Mechanical stimulation and endothelial cell interactions with the vascular matrix, as well as a host of soluble factors, coordinately contribute to this dynamic regulation. RECENT ADVANCES Blood hemodynamics play an established role in the regulation of endothelial function. However, a growing body of work suggests that subendothelial matrix composition similarly and coordinately regulates endothelial cell phenotype such that blood flow affects matrix remodeling, which affects the endothelial response to flow. CRITICAL ISSUES Hemodynamics and soluble factors likely affect endothelial matrix remodeling through multiple mechanisms, including transforming growth factor β signaling and alterations in cell-matrix receptors, such as the integrins. Likewise, differential integrin signaling following matrix remodeling appears to regulate several key flow-induced responses, including nitric oxide production, regulation of oxidant stress, and activation of proinflammatory signaling and gene expression. Microvascular remodeling responses, such as angiogenesis and arteriogenesis, may also show coordinated regulation by flow and matrix. FUTURE DIRECTIONS Identifying the mechanisms regulating the dynamic interplay between hemodynamics and matrix remodeling and their contribution to the pathogenesis of cardiovascular disease remains an important research area with therapeutic implications across a variety of conditions. Antioxid. Redox Signal. 25, 415-434.
Collapse
Affiliation(s)
- Arif Yurdagul
- Department of Cell Biology and Anatomy, Louisiana State University Health Sciences Center–Shreveport, Shreveport, Louisiana
| | - A. Wayne Orr
- Department of Cell Biology and Anatomy, Louisiana State University Health Sciences Center–Shreveport, Shreveport, Louisiana
- Department of Pathology and Translational Pathobiology, Louisiana State University Health Sciences Center–Shreveport, Shreveport, Louisiana
| |
Collapse
|
27
|
Minami T, Satoh K, Nogi M, Kudo S, Miyata S, Tanaka SI, Shimokawa H. Statins up-regulate SmgGDS through β1-integrin/Akt1 pathway in endothelial cells. Cardiovasc Res 2015; 109:151-61. [PMID: 26598509 DOI: 10.1093/cvr/cvv253] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/15/2015] [Accepted: 11/10/2015] [Indexed: 12/26/2022] Open
Abstract
AIMS The pleiotropic effects of HMG-CoA reductase inhibitors (statins) independent of cholesterol-lowering effects have attracted much attention. We have recently demonstrated that the pleiotropic effects of statins are partly mediated through up-regulation of small GTP-binding protein dissociation stimulator (SmgGDS) with a resultant Rac1 degradation and reduced oxidative stress. However, it remains to be elucidated what molecular mechanisms are involved. METHODS AND RESULTS To first determine in what tissue statins up-regulate SmgGDS expression, we examined the effects of two statins (atorvastatin 10 mg/kg per day and pravastatin 50 mg/kg per day for 1 week) on SmgGDS expression in mice in vivo. The two statins increased SmgGDS expression especially in the aorta. Atorvastatin also increased SmgGDS expression in cultured human umbilical venous endothelial cells (HUVEC) and human aortic endothelial cells, but not in human aortic vascular smooth muscle cells. Furthermore, Akt phosphorylation was transiently enhanced only in HUVEC in response to atorvastatin. Then, to examine whether Akt is involved for up-regulation of SmgGDS by statins, we knocked out Akt1 by its siRNA in HUVEC, which abolished the effects by atorvastatin to up-regulate SmgGDS. Furthermore, when we knocked down β1-integrin to elucidate the upstream molecule of Akt1, the effect of atorvastatin to up-regulate SmgGDS was abolished. Finally, we confirmed that Akt activator, SC79, significantly up-regulate SmgGDS in HUVEC. CONCLUSION These results indicate that statins selectively up-regulate SmgGDS in endothelial cells, for which the β1-integrin/Akt1 pathway may be involved, demonstrating the novel aspects of the pleiotropic effects of statins.
Collapse
Affiliation(s)
- Tatsuro Minami
- Department of Cardiovascular Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan Laboratory for Pharmacology, Pharmaceuticals Research Center, Asahi Kasei Pharma Corporation, Izunokuni, Japan
| | - Kimio Satoh
- Department of Cardiovascular Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Masamichi Nogi
- Department of Cardiovascular Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Shun Kudo
- Department of Cardiovascular Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Satoshi Miyata
- Department of Cardiovascular Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Shin-ichi Tanaka
- Department of Cardiovascular Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan Laboratory for Pharmacology, Pharmaceuticals Research Center, Asahi Kasei Pharma Corporation, Izunokuni, Japan
| | - Hiroaki Shimokawa
- Department of Cardiovascular Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan
| |
Collapse
|
28
|
Chen J, Green J, Yurdagul A, Albert P, McInnis MC, Orr AW. αvβ3 Integrins Mediate Flow-Induced NF-κB Activation, Proinflammatory Gene Expression, and Early Atherogenic Inflammation. THE AMERICAN JOURNAL OF PATHOLOGY 2015. [PMID: 26212910 DOI: 10.1016/j.ajpath.2015.05.013] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Endothelial cell interactions with transitional matrix proteins, such as fibronectin, occur early during atherogenesis and regulate shear stress-induced endothelial cell activation. Multiple endothelial cell integrins bind transitional matrix proteins, including α5β1, αvβ3, and αvβ5. However, the role these integrins play in mediating shear stress-induced endothelial cell activation remains unclear. Therefore, we sought to elucidate which integrin heterodimers mediate shear stress-induced endothelial cell activation and early atherogenesis. We now show that inhibiting αvβ3 integrins (S247, siRNA), but not α5β1 or αvβ5, blunts shear stress-induced proinflammatory signaling (NF-κB, p21-activated kinase) and gene expression (ICAM1, VCAM1). Importantly, inhibiting αvβ3 did not affect cytokine-induced proinflammatory responses or inhibit all shear stress-induced signaling, because Akt, endothelial nitric oxide synthase, and extracellular regulated kinase activation remained intact. Furthermore, inhibiting αv integrins (S247), but not α5 (ATN-161), in atherosclerosis-prone apolipoprotein E knockout mice significantly reduced vascular remodeling after acute induction of disturbed flow. S247 treatment similarly reduced early diet-induced atherosclerotic plaque formation associated with both diminished inflammation (expression of vascular cell adhesion molecule 1, plaque macrophage content) and reduced smooth muscle incorporation. Inducible, endothelial cell-specific αv integrin deletion similarly blunted inflammation in models of disturbed flow and diet-induced atherogenesis but did not affect smooth muscle incorporation. Our studies identify αvβ3 as the primary integrin heterodimer mediating shear stress-induced proinflammatory responses and as a key contributor to early atherogenic inflammation.
Collapse
Affiliation(s)
- Jie Chen
- Department of Pathology, Louisiana State University Health Sciences Center, Shreveport, Louisiana
| | - Jonette Green
- Department of Pathology, Louisiana State University Health Sciences Center, Shreveport, Louisiana
| | - Arif Yurdagul
- Department of Pathology, Louisiana State University Health Sciences Center, Shreveport, Louisiana; Department of Cell Biology and Anatomy, Louisiana State University Health Sciences Center, Shreveport, Louisiana
| | - Patrick Albert
- Department of Pathology, Louisiana State University Health Sciences Center, Shreveport, Louisiana
| | - Marshall C McInnis
- Department of Pathology, Louisiana State University Health Sciences Center, Shreveport, Louisiana
| | - A Wayne Orr
- Department of Pathology, Louisiana State University Health Sciences Center, Shreveport, Louisiana.
| |
Collapse
|
29
|
Inflammatory mediators in vascular disease: identifying promising targets for intracranial aneurysm research. Mediators Inflamm 2015; 2015:896283. [PMID: 25922566 PMCID: PMC4397479 DOI: 10.1155/2015/896283] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2015] [Revised: 03/18/2015] [Accepted: 03/19/2015] [Indexed: 12/21/2022] Open
Abstract
Inflammatory processes are implicated in many diseases of the vasculature and have been shown to play a key role in the formation of intracranial aneurysms (IAs). Although the specific mechanisms underlying these processes have been thoroughly investigated in related pathologies, such as atherosclerosis, there remains a paucity of information regarding the immunopathology of IA. Cells such as macrophages and lymphocytes and their effector molecules have been suggested to be players in IA, but their specific interactions and the role of other components of the inflammatory response have yet to be determined. Drawing parallels between the pathogenesis of IA and other vascular disorders could provide a roadmap for developing a mechanistic understanding of the immunopathology of IA and uncovering useful targets for therapeutic intervention. Future research should address the presence and function of leukocyte subsets, mechanisms of leukocyte recruitment and activation, and the role of damage-associated molecular patterns in IA.
Collapse
|
30
|
Kataoka H. Molecular mechanisms of the formation and progression of intracranial aneurysms. Neurol Med Chir (Tokyo) 2015; 55:214-29. [PMID: 25761423 PMCID: PMC4533330 DOI: 10.2176/nmc.ra.2014-0337] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Until recently, only a little was understood about molecular mechanisms of the development of an intracranial aneurysm (IA). Recent advancements over the last decade in the field of genetics and molecular biology have provided us a wide variety of evidences supporting the notion that chronic inflammation is closely associated with the pathogenesis of IA development. In the field of genetics, large-scale Genome-wide association studies (GWAS) has identified some IA susceptible loci and genes related to cell cycle and endothelial function. Researches in molecular biology using human samples and animal models have revealed the common pathway of the initiation, progression, and rupture of IAs. IA formation begins with endothelial dysfunction followed by pathological remodeling with degenerative changes of vascular walls. Medical treatments inhibiting inflammatory cascades in IA development are likely to prevent IA progression and rupture. Statins and aspirin are expected to suppress IA progression by their anti-inflammatory effects. Decoy oligodeoxynucleotides (ODNs) inhibiting inflammatory transcription factors such as nuclear factor kappa-B (NF-κB) and Ets-1 are the other promising choice of the prevention of IA development. Further clarification of molecular mechanisms of the formation and progression of IAs will shed light to the pathogenesis of IA development and provide insight into novel diagnostic and therapeutic strategies for IAs.
Collapse
Affiliation(s)
- Hiroharu Kataoka
- Department of Neurosurgery, National Cerebral and Cardiovascular Center
| |
Collapse
|
31
|
Yokoi T, Isono T, Saitoh M, Yoshimura Y, Nozaki K. Suppression of cerebral aneurysm formation in rats by a tumor necrosis factor-α inhibitor. J Neurosurg 2014; 120:1193-200. [PMID: 24628611 DOI: 10.3171/2014.1.jns13818] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
OBJECT Although cerebral aneurysmal subarachnoid hemorrhage is a devastating disease for humans, effective medical treatments have not yet been established. Recent reports have shown that regression of some inflammatory-related mediators has protective effects in experimental cerebral aneurysm models. This study corroborated the effectiveness of tumor necrosis factor-α (TNF-α) inhibitor for experimentally induced cerebral aneurysms in rats. METHODS Five-week-old male rats were prepared for induction of cerebral aneurysms and divided into 3 groups, 2 groups administered different concentrations of a TNF-α inhibitor (etanercept), and 1 control group. One month after aneurysm induction, 7-T MRI was performed. The TNF-α inhibitor groups received subcutaneous injection of 25 μg or 2.5 μg of etanercept, and the control group received subcutaneous injection of normal saline every week. The TNF-α inhibitor administrations were started at 1 month after aneurysm induction to evaluate its suppressive effects on preexisting cerebral aneurysms. Arterial circles of Willis were obtained and evaluated 3 months after aneurysm induction. RESULTS Rats administered a TNF-α inhibitor experienced significant increases in media thickness and reductions in aneurysmal size compared with the control group. Immunohistochemical staining showed that treatment with a TNF-α inhibitor suppressed matrix metalloproteinase (MMP)-9 and inducible nitric oxide synthase (iNOS) expression through the luminal surface of the endothelial cell layer, the media and the adventitia at the site of aneurysmal formation, and the anterior cerebral artery-olfactory artery bifurcation. Quantitative polymerase chain reaction also showed suppression of MMP-9 and iNOS by TNF-α inhibitor administration. CONCLUSIONS Therapeutic administration of a TNF-α inhibitor significantly reduced the formation of aneurysms in rats. These data also suggest that TNF-α suppression reduced some inflammatory-related mediators that are in the downstream pathway of nuclear factor-κB.
Collapse
|
32
|
Raaz U, Toh R, Maegdefessel L, Adam M, Nakagami F, Emrich FC, Spin JM, Tsao PS. Hemodynamic regulation of reactive oxygen species: implications for vascular diseases. Antioxid Redox Signal 2014; 20:914-28. [PMID: 23879326 PMCID: PMC3924901 DOI: 10.1089/ars.2013.5507] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
SIGNIFICANCE Arterial blood vessels functionally and structurally adapt to altering hemodynamic forces in order to accommodate changing needs and to provide stress homeostasis. This ability is achieved at the cellular level by converting mechanical stimulation into biochemical signals (i.e., mechanotransduction). Physiological mechanical stress helps maintain vascular structure and function, whereas pathologic or aberrant stress may impair cellular mechano-signaling, and initiate or augment cellular processes that drive disease. RECENT ADVANCES Reactive oxygen species (ROS) may represent an intriguing class of mechanically regulated second messengers. Chronically enhanced ROS generation may be induced by adverse mechanical stresses, and is associated with a multitude of vascular diseases. Although a causal relationship has clearly been demonstrated in large numbers of animal studies, an effective ROS-modulating therapy still remains to be established by clinical studies. CRITICAL ISSUES AND FUTURE DIRECTIONS This review article focuses on the role of various mechanical forces (in the form of laminar shear stress, oscillatory shear stress, or cyclic stretch) as modulators of ROS-driven signaling, and their subsequent effects on vascular biology and homeostasis, as well as on specific diseases such as arteriosclerosis, hypertension, and abdominal aortic aneurysms. Specifically, it highlights the significance of the various NADPH oxidase (NOX) isoforms as critical ROS generators in the vasculature. Directed targeting of defined components in the complex network of ROS (mechano-)signaling may represent a key for successful translation of experimental findings into clinical practice.
Collapse
Affiliation(s)
- Uwe Raaz
- 1 Division of Cardiovascular Medicine, Stanford University School of Medicine , Stanford, California
| | | | | | | | | | | | | | | |
Collapse
|
33
|
Abstract
This review looks at potential developments in medical devices which may be based upon nanofeaturing implant and tissue engineering scaffolds, and describes the basic science upon which such expectations are based.
Collapse
Affiliation(s)
- Adam Curtis
- Centre for Cell Engineering, University of Glasgow, Glasgow G12 8QQ, Scotland, UK.
| |
Collapse
|
34
|
Kataoka H, Aoki T. Molecular basis for the development of intracranial aneurysm. Expert Rev Neurother 2014. [DOI: 10.1586/ern.09.155] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
35
|
DeVerse JS, Sandhu AS, Mendoza N, Edwards CM, Sun C, Simon SI, Passerini AG. Shear stress modulates VCAM-1 expression in response to TNF-α and dietary lipids via interferon regulatory factor-1 in cultured endothelium. Am J Physiol Heart Circ Physiol 2013; 305:H1149-57. [PMID: 23934855 DOI: 10.1152/ajpheart.00311.2013] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Dyslipidemia is a primary risk factor for cardiovascular disease, but the specific mechanisms that determine the localization of atherosclerotic plaques in arteries are not well defined. Triglyceride-rich lipoproteins (TGRL) isolated from human plasma after a high-fat meal modulate TNF-α-induced VCAM-1 expression in cultured human aortic endothelial cells (HAECs) via an interferon regulatory factor (IRF)-1-dependent transcriptional mechanism. We examined whether fluid shear stress acts as a mediator of IRF-1-dependent VCAM-1 expression in response to cytokine and dietary lipids. IRF-1 and VCAM-1 were examined by immunofluorescence in TNF-α-stimulated HAEC monolayers exposed to TGRL and a linear gradient of shear stress ranging from 0 to 16 dyn/cm(2) in a microfluidic device. Shear stress alone modulated TNF-α-induced VCAM-1 expression, eliciting a 150% increase at low shear stress (2 dyn/cm(2)) and a 70% decrease at high shear stress (12 dyn/cm(2)) relative to static. These differences correlated with a 60% increase in IRF-1 expression under low shear stress and a 40% decrease under high shear stress. The addition of TGRL along with cytokine activated a fourfold increase in VCAM-1 expression and a twofold increase in IRF-1 expression. The combined effect of shear stress and TGRL on the upregulation of membrane VCAM-1 was abolished by transfection of HAECs with IRF-1-specific small interfering RNA. In a healthy swine model, elevated levels of endothelial IRF-1 were also observed within atherosusceptible regions of the aorta by Western blot analysis and immunohistochemistry, implicating arterial hemodynamics in the regulation of IRF-1 expression. These data demonstrate direct roles for fluid shear stress and postprandial TGRL from human serum in the regulation of IRF-1 expression and downstream inflammatory responses in HAECs.
Collapse
Affiliation(s)
- J Sherrod DeVerse
- Department of Biomedical Engineering, University of California, Davis, CA
| | | | | | | | | | | | | |
Collapse
|
36
|
Laux DW, Young S, Donovan JP, Mansfield CJ, Upton PD, Roman BL. Circulating Bmp10 acts through endothelial Alk1 to mediate flow-dependent arterial quiescence. Development 2013; 140:3403-12. [PMID: 23863480 DOI: 10.1242/dev.095307] [Citation(s) in RCA: 78] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Blood flow plays crucial roles in vascular development, remodeling and homeostasis, but the molecular pathways required for transducing flow signals are not well understood. In zebrafish embryos, arterial expression of activin receptor-like kinase 1 (alk1), which encodes a TGFβ family type I receptor, is dependent on blood flow, and loss of alk1 mimics lack of blood flow in terms of dysregulation of a subset of flow-responsive arterial genes and increased arterial endothelial cell number. These data suggest that blood flow activates Alk1 signaling to promote a flow-responsive gene expression program that limits nascent arterial caliber. Here, we demonstrate that restoration of endothelial alk1 expression to flow-deprived arteries fails to rescue Alk1 activity or normalize arterial endothelial cell gene expression or number, implying that blood flow may play an additional role in Alk1 signaling independent of alk1 induction. To this end, we define cardiac-derived Bmp10 as the crucial ligand for endothelial Alk1 in embryonic vascular development, and provide evidence that circulating Bmp10 acts through endothelial Alk1 to limit endothelial cell number in and thereby stabilize the caliber of nascent arteries. Thus, blood flow promotes Alk1 activity by concomitantly inducing alk1 expression and distributing Bmp10, thereby reinforcing this signaling pathway, which functions to limit arterial caliber at the onset of flow. Because mutations in ALK1 cause arteriovenous malformations (AVMs), our findings suggest that an impaired flow response initiates AVM development.
Collapse
Affiliation(s)
- Derek W Laux
- Department of Biological Sciences, University of Pittsburgh, Pittsburgh, PA 15260, USA
| | | | | | | | | | | |
Collapse
|
37
|
Shear Stress Activates eNOS at the Endothelial Apical Surface Through β1 Containing Integrins and Caveolae. Cell Mol Bioeng 2013; 6:346-354. [PMID: 23956799 DOI: 10.1007/s12195-013-0276-9] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
There is now a large body of evidence demonstrating that fluid mechanical forces generated by blood flowing through the vasculature play a direct role in regulating endothelial cell structure and function. Integrin receptors that localize to the basal surface of the endothelium participate in both outside-in and inside-out signaling events that influence endothelial gene expression and morphology in response to flow. Our analyses of apical plasma membranes derived from cultured bovine aortic endothelial cells revealed that integrins are also expressed on this cell surface. Here, we tested whether these integrins participate in mechanotransduction events that are known to occur on the endothelial cell luminal/apical membrane. We found that apically expressed β1 integrins are rapidly activated in response to acute shear stress. Blockade of β1 integrin activation attenuated a shear-induced signaling cascade involving Src-family kinase, PI3-kinase, Akt and eNOS on this cell surface. In addition, β1 integrin activation and associated signaling events were dependent on the structural integrity of caveolae but not the actin cytoskeleton. Taken together, these data indicate that endothelial responses to shear stress are mediated by spatially distinct pools of integrins.
Collapse
|
38
|
Vascular Endothelium. TISSUE FUNCTIONING AND REMODELING IN THE CIRCULATORY AND VENTILATORY SYSTEMS 2013. [DOI: 10.1007/978-1-4614-5966-8_9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
|
39
|
Mechanotransduction in embryonic vascular development. Biomech Model Mechanobiol 2012; 11:1149-68. [PMID: 22744845 DOI: 10.1007/s10237-012-0412-9] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2012] [Accepted: 06/09/2012] [Indexed: 12/25/2022]
Abstract
A plethora of biochemical signals provides spatial and temporal cues that carefully orchestrate the complex process of vertebrate embryonic development. The embryonic vasculature develops not only in the context of these biochemical cues, but also in the context of the biomechanical forces imparted by blood flow. In the mature vasculature, different blood flow regimes induce distinct genetic programs, and significant progress has been made toward understanding how these forces are perceived by endothelial cells and transduced into biochemical signals. However, it cannot be assumed that paradigms that govern the mature vasculature are pertinent to the developing embryonic vasculature. The embryonic vasculature can respond to the mechanical forces of blood flow, and these responses are critical in vascular remodeling, certain aspects of sprouting angiogenesis, and maintenance of arterial-venous identity. Here, we review data regarding mechanistic aspects of endothelial cell mechanotransduction, with a focus on the response to shear stress, and elaborate upon the multifarious effects of shear stress on the embryonic vasculature. In addition, we discuss emerging predictive vascular growth models and highlight the prospect of combining signaling pathway information with computational modeling. We assert that correlation of precise measurements of hemodynamic parameters with effects on endothelial cell gene expression and cell behavior is required for fully understanding how blood flow-induced loading governs normal vascular development and shapes congenital cardiovascular abnormalities.
Collapse
|
40
|
Reichenberg Y, Lanir Y. Duration of microbead seeding on endothelial cells significantly affects their response to magnetic excitation. PHYSICAL REVIEW. E, STATISTICAL, NONLINEAR, AND SOFT MATTER PHYSICS 2012; 85:041915. [PMID: 22680506 DOI: 10.1103/physreve.85.041915] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/27/2011] [Revised: 03/10/2012] [Indexed: 06/01/2023]
Abstract
Our investigation of endothelial cell rheology using optical magnetic twisting cytometry revealed that with time following incubation of ferromagnetic beads on the cells, beads were sinking into the cells and an increasing number of beads demonstrated apparent absurd negative rheological properties. In parallel, the beads' average rheological response changed considerably over time, both in magnitude and in distribution. It was hypothesized that the apparent negative rheological response was related to the above sinking process of seeded beads into the cells, resulting in an elevation of the beads' rotation axis, thus causing a reversal of the beads' lateral movement direction in response to twisting external magnetic excitation. The results suggest that microbead-based rheological characterization of cells should be interpreted with caution, while considering the time of data acquisition.
Collapse
Affiliation(s)
- Yaniv Reichenberg
- Faculty of Biomedical Engineering, Technion-Israel Institute of Technology, Haifa 32000, Israel.
| | | |
Collapse
|
41
|
Urschel K, Garlichs CD, Daniel WG, Cicha I. VEGFR2 signalling contributes to increased endothelial susceptibility to TNF-α under chronic non-uniform shear stress. Atherosclerosis 2011; 219:499-509. [PMID: 22019447 DOI: 10.1016/j.atherosclerosis.2011.09.045] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/19/2011] [Revised: 09/04/2011] [Accepted: 09/27/2011] [Indexed: 01/19/2023]
Abstract
OBJECTIVES Vascular endothelial growth factor receptor 2 (VEGFR2), a tyrosine kinase receptor activated by VEGF and shear stress, is critically involved in endothelial mechanotransduction. We investigated the role of VEGFR2 in non-uniform shear stress-induced endothelial susceptibility to inflammatory stimuli. METHODS Endothelial cells (ECs) were exposed to non-uniform shear stress, followed by stimulation with TNF-α. ECs were transfected with siRNAs against VEGFR2. Alternatively, ECs were treated with blocking antibody against VEGFR2, or with inhibitors of VEGFR2 (ZM 323881), PI3K (LY 294002), or Src-kinase (PP2). THP-1 monocytes were used for dynamic adhesion assays. Endothelial protein expression was determined by immunofluorescence. RESULTS siRNA against VEGFR2 decreased VEGFR2 protein expression by 40% as determined by Western blotting. In endothelial cells exposed to non-uniform shear stress, VEGFR2 knockdown inhibited TNF-α-induced NF-κB translocation to the nucleus, and the upregulation of VCAM-1 and E-selectin. Consequently, monocytic cell recruitment to endothelium under non-uniform shear stress conditions was reduced. Similar effects were observed by blocking VEGFR2 activity using a specific inhibitor ZM 323881, or an antibody against VEGFR2 before TNF-α stimulation. Inhibition of PI3K with LY 294002 significantly reduced non-uniform shear stress-induced endothelial susceptibility to TNF-α, whereas blocking Src-kinase with PP2 was ineffective. CONCLUSION VEGFR2 is critically involved in adhesion molecule induction and monocytic cell recruitment to endothelium in response to non-uniform shear stress and TNF-α. Targeting the mechanosensory cascade can prevent endothelial activation in atherosclerosis-prone regions.
Collapse
Affiliation(s)
- Katharina Urschel
- Department of Cardiology and Angiology, University of Erlangen-Nuremberg, Erlangen, Germany
| | | | | | | |
Collapse
|
42
|
|
43
|
MicroRNA-10a regulation of proinflammatory phenotype in athero-susceptible endothelium in vivo and in vitro. Proc Natl Acad Sci U S A 2010; 107:13450-5. [PMID: 20624982 DOI: 10.1073/pnas.1002120107] [Citation(s) in RCA: 358] [Impact Index Per Article: 23.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
A chronic proinflammatory state precedes pathological change in arterial endothelial cells located within regions of susceptibility to atherosclerosis. The potential contributions of regulatory microRNAs to this disequilibrium were investigated by artery site-specific profiling in normal adult swine. Expression of endothelial microRNA10a (miR-10a) was lower in the athero-susceptible regions of the inner aortic arch and aorto-renal branches than elsewhere. Expression of Homeobox A1 (HOXA1), a known miR-10a target, was up-regulated in the same locations. Endothelial transcriptome microarray analysis of miR-10a knockdown in cultured human aortic endothelial cells (HAEC) identified IkappaB/NF-kappaB-mediated inflammation as the top category of up-regulated biological processes. Phosphorylation of IkappaBalpha, a prerequisite for IkappaBalpha proteolysis and NF-kappaB activation, was significantly up-regulated in miR-10a knockdown HAEC and was accompanied by increased nuclear expression of NF-kappaB p65. The inflammatory biomarkers monocyte chemotactic protein 1 (MCP-1), IL-6, IL-8, vascular cell adhesion molecule 1 (VCAM-1), and E-selectin were elevated following miR-10a knockdown. Conversely, knockin of miR-10a (a conservative 25-fold increase) inhibited the basal expression of VCAM-1 and E-selectin in HAEC. Two key regulators of IkappaBalpha degradation--mitogen-activated kinase kinase kinase 7 (MAP3K7; TAK1) and beta-transducin repeat-containing gene (betaTRC)--contain a highly conserved miR-10a binding site in the 3' UTR. Both molecules were up-regulated by miR-10a knockdown and suppressed by miR-10a knockin, and evidence of direct miR-10a binding to the 3' UTR was demonstrated by luciferase assay. Comparative expression studies of endothelium located in athero-susceptible aortic arch and athero-protected descending thoracic aorta identified significantly up-regulated MAP3K7, betaTRC, phopho-IkappaBalpha, and nuclear p65 expression suggesting that the differential expression of miR-10a contributes to the regulation of proinflammatory endothelial phenotypes in athero-susceptible regions in vivo.
Collapse
|
44
|
Young SRL, Gerard-O'Riley R, Harrington M, Pavalko FM. Activation of NF-kappaB by fluid shear stress, but not TNF-alpha, requires focal adhesion kinase in osteoblasts. Bone 2010; 47:74-82. [PMID: 20353839 PMCID: PMC2891440 DOI: 10.1016/j.bone.2010.03.014] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/09/2009] [Revised: 02/24/2010] [Accepted: 03/22/2010] [Indexed: 10/19/2022]
Abstract
When bone is mechanically loaded fluid shear stress (FSS) is generated as a result of the movement of interstitial fluid across the membranes of osteoblasts and osteocytes. This external mechanical loading stimulates changes in the activity of cytoplasmic signaling molecules and alters gene expression in bone cells. This process, referred to as mechanotransduction, is vital for maintaining bone health in vivo by regulating the balance between bone formation and bone resorption. This current study focuses on the role of focal adhesions, sites of integrin-mediated cellular attachment to the extracellular matrix, and their proposed function as mechanosensors in bone cells. We examined the role of a key component of focal adhesions and of mechanotransduction, focal adhesion kinase (FAK) in regulation of FSS- and tumor necrosis factor-alpha (TNF-alpha)-induced activation of nuclear factor-kappa B (NF-kappaB) signaling in osteoblasts. Immortalized FAK(+/+) and FAK(-)(/)(-) osteoblasts were exposed to periods of oscillatory fluid shear stress (OFF) and NF-kappaB activation was analyzed. We determined that FAK is required for OFF-induced nuclear translocation and activation of NF-kappaB in osteoblasts. In addition we found that OFF-induced phosphorylation of the IkappaB kinases (IKKalpha/beta) in both FAK(+/+) and FAK(-/-) osteoblasts, but only FAK(+/+) osteoblasts demonstrated the resulting degradation of NF-kappaB inhibitors IkappaBalpha and IkappaBbeta. OFF did not induce the degradation of IkappaBepsilon or the processing of p105 in either FAK(+/+) and FAK(-/-) osteoblasts. To compare the role of FAK in mediating OFF-induced mechanotransduction to the well characterized activation of NF-kappaB by inflammatory cytokines, we exposed FAK(+/+) and FAK(-/-) osteoblasts to TNF-alpha. Interestingly, FAK was not required for TNF-alpha induced NF-kappaB activation in osteoblasts. In addition we determined that TNF-alpha treatment did not induce the degradation of IkappaBbeta as did OFF. These data indicate a novel relationship between FAK and NF-kappaB activation in osteoblast mechanotransduction and demonstrates that the mechanism of FSS-induced NF-kappaB activation in osteoblasts differs from the well characterized TNF-alpha-induced activation.
Collapse
Affiliation(s)
- Suzanne R L Young
- Department of Cellular and Integrative Physiology, Indiana University School of Medicine, Indianapolis, IN 46202, USA.
| | | | | | | |
Collapse
|
45
|
Shear Stress Regulates the Flk-1/Cbl/PI3K/NF-κB Pathway Via Actin and Tyrosine Kinases. Cell Mol Bioeng 2009; 2:341-350. [PMID: 20011623 DOI: 10.1007/s12195-009-0069-3] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022] Open
Abstract
Vascular endothelial cells (ECs) are continuously exposed to mechanical stimuli (e.g., shear stress). Our previous study has shown that the shear-induced nuclear factor-κB (NF-κB) activation is mediated by integrins [Bhullar, I. S., Y. S. Li, H. Miao, E. Zandi, M. Kim, et al. J. Biol. Chem. 273:30544-30549, 1998]. The shear-activated integrins can also transactivate Flk-1 (a receptor for vascular endothelial growth factor (VEGF)) [Wang, Y., H. Miao, S. Li, K. D. Chen, Y. S. Li, et al. Am. J. Physiol. Cell Physiol. 283:C1540-C1547, 2002], which subsequently recruits Casitas B-lineage lymphoma (Cbl) to regulate inhibitor of κB protein kinase (IKK) [Wang, Y., J. Chang, Y. C. Li, Y. S. Li, J. Y. Shyy, and S. Chien. Am. J. Physiol. Heart Circ. Physiol. 286:H685-H692, 2004], an upstream molecule of NF-κB. Therefore, shear stress may likely utilize the Flk-1/Cbl pathway in regulating NF-κB. In this paper, we confirmed that the inhibition of Flk-1 by its specific inhibitor SU1498 blocked the shear-induced NF-κB translocation. The inhibition of Cbl (an adaptor protein which binds to Flk-1 upon shear) by using a negative mutant (Cbl(nm)) also blocked the promoter activity of NF-κB, and the inhibition of the Cbl-downstream molecule phosphatidylinositol-3-kinase (PI3K) abolished the NF-κB translocation. Further experiments revealed that the disruption of actin cytoskeleton inhibited the Flk-1 and Cbl interaction and NF-κB translocation. The inhibition of focal adhesion kinase (FAK) and Src family kinases, which are involved in the integrin-mediated focal adhesion complex, also blocked the shear-induced NF-κB translocation. Together with our previous findings that integrins mediate the shear-induced activation of Flk-1 and NF-κB [Bhullar, I. S., Y. S. Li, H. Miao, E. Zandi, M. Kim, et al. J. Biol. Chem. 273:30544-30549, 1998; Wang, Y., H. Miao, S. Li, K. D. Chen, Y. S. Li, et al. Am. J. Physiol. Cell Physiol. 283:C1540-C1547, 2002], the present results suggest that Flk-1, Cbl, and PI3K act upstream to NF-κB in response to shear stress. This Flk-1/Cbl/PI3K/NF-κB signaling pathway may be originated from integrins and transmitted by key tyrosine kinases and actin cytoskeleton. These results shed new lights on the molecular mechanism by which mechanical shear stress activates the NF-κB signaling pathway, which is critical for vascular inflammatory responses and atherosclerosis.
Collapse
|
46
|
Petzold T, Orr AW, Hahn C, Jhaveri KA, Parsons JT, Schwartz MA. Focal adhesion kinase modulates activation of NF-kappaB by flow in endothelial cells. Am J Physiol Cell Physiol 2009; 297:C814-22. [PMID: 19587216 DOI: 10.1152/ajpcell.00226.2009] [Citation(s) in RCA: 64] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Atherogenesis involves activation of NF-kappaB in endothelial cells by fluid shear stress. Because this pathway involves integrins, we investigated the involvement of focal adhesion kinase (FAK). We found that FAK was not required for flow-stimulated translocation of the p65 NF-kappaB subunit to the nucleus but was essential for phosphorylation of p65 on serine 536 and induction of ICAM-1, an NF-kappaB-dependent gene. NF-kappaB activation by TNF-alpha or hydrogen peroxide was FAK independent. Events upstream of NF-kappaB, including integrin activation, Rac activation, reactive oxygen production, and degradation of IkappaB, were FAK independent. FAK therefore regulates NF-kappaB phosphorylation and transcriptional activity in response to flow by a novel mechanism.
Collapse
Affiliation(s)
- Tobias Petzold
- Robert M. Berne Cardiovascular Research Center, University of Virginia, Charlottesville, Virginia 22908, USA
| | | | | | | | | | | |
Collapse
|
47
|
Castier Y, Ramkhelawon B, Riou S, Tedgui A, Lehoux S. Role of NF-kappaB in flow-induced vascular remodeling. Antioxid Redox Signal 2009; 11:1641-9. [PMID: 19320561 DOI: 10.1089/ars.2008.2393] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Vascular remodeling associated with increased blood flow involves reactive oxygen species (ROS)-dependent activation of matrix metalloproteinases (MMPs). To investigate the potential role of NF-kappaB in this process, human umbilical vein endothelial cells were subjected to different flow conditions during a 24-h period. Normal (15 dynes/cm(2)) and high (30 dynes/cm(2)) shear stress induced IkappaBalpha degradation and NF-kappaB p65 phosphorylation, and activated MMP-2 and MMP-9. These effects were blunted in cells incubated with the NF-kappaB inhibitor pyrrolidine dithio-carbamate (PDTC). In mice, creation of a carotid artery-jugular vein arteriovenous fistula (AVF) increased carotid blood flow sixfold, triggering the increase in carotid diameter from 459 +/- 8 microm (before AVF) to 531 +/- 13 and 669 +/- 21 microm (7 and 21 days after AVF). ROS production and NF-kappaB activity were enhanced in fistulated carotids, but only the latter was blocked by PDTC, although PDTC blocked ROS production in vitro. In PDTC-treated mice, changes in carotid caliber and shear stress matched controls at 7 days, but carotids enlarged only marginally thereafter, reaching only 578 +/- 8 microm at 21 days (p < 0.01 vs. untreated). Similarly, both MMP-9 expression and activity were abrogated by PDTC at 3 weeks. Hence, induction of NF-kappaB by shear stress contributes to MMP induction and allows long-term flow-induced vascular enlargement.
Collapse
Affiliation(s)
- Yves Castier
- Parts Cardiovascular Research Center, Inserm U970, HEGP, Paris, France
| | | | | | | | | |
Collapse
|
48
|
Abstract
Forces that are associated with blood flow are major determinants of vascular morphogenesis and physiology. Blood flow is crucial for blood vessel development during embryogenesis and for regulation of vessel diameter in adult life. It is also a key factor in atherosclerosis, which, despite the systemic nature of major risk factors, occurs mainly in regions of arteries that experience disturbances in fluid flow. Recent data have highlighted the potential endothelial mechanotransducers that might mediate responses to blood flow, the effects of atheroprotective rather than atherogenic flow, the mechanisms that contribute to the progression of the disease and how systemic factors interact with flow patterns to cause atherosclerosis.
Collapse
Affiliation(s)
| | - Martin A. Schwartz
- Department of Microbiology, University of Virginia
- Robert M. Berne Cardiovascular Research Center, Mellon Prostate Cancer Research Center and Departments of Cell Biology and Biomedical Engineering, University of Virginia. Charlottesville VA 22908
| |
Collapse
|
49
|
Lowering caveolin-1 expression in human vascular endothelial cells inhibits signal transduction in response to shear stress. Int J Cell Biol 2008; 2009:532432. [PMID: 20111626 PMCID: PMC2809413 DOI: 10.1155/2009/532432] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2008] [Accepted: 10/19/2008] [Indexed: 11/17/2022] Open
Abstract
Vascular endothelial cells have an extensive response to physiological levels of shear stress. There is evidence that the protein caveolin-1 is involved in the early phase of this response. In this study, caveolin-1 was downregulated in human endothelial cells by RNAi. When these cells were subjected to a shear stress of 15 dyn/cm(2) for 10 minutes, activation of Akt and ERK1/2 was significantly lower than in control cells. Moreover, activation of Akt and ERK1/2 in response to vascular endothelial growth factor was significantly lower in cells with low levels of caveolin-1. However, activation of integrin-mediated signaling during cell adhesion onto fibronectin was not hampered by lowered caveolin-1 levels. In conclusion, caveolin-1 is an essential component in the response of endothelial cells to shear stress. Furthermore, the results suggest that the role of caveolin-1 in this process lies in facilitating efficient VEGFR2-mediated signaling.
Collapse
|
50
|
Birukova AA, Rios A, Birukov KG. Long-term cyclic stretch controls pulmonary endothelial permeability at translational and post-translational levels. Exp Cell Res 2008; 314:3466-77. [PMID: 18824167 PMCID: PMC2638120 DOI: 10.1016/j.yexcr.2008.09.003] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2008] [Revised: 08/22/2008] [Accepted: 09/10/2008] [Indexed: 11/17/2022]
Abstract
We have previously described differential effects of physiologic (5%) and pathologic (18%) cyclic stretch (CS) on agonist-induced pulmonary endothelial permeability. This study examined acute and chronic effects of CS on agonist-induced intracellular signaling and cell morphology in the human lung macro- and microvascular endothelial cell (EC) monolayers. Endothelial permeability was assessed by analysis of morphological changes, parameters of cell contraction and measurements of transendothelial electrical resistance. Exposure of both microvascular and macrovascular EC to 18% CS for 2-96 h increased thrombin-induced permeability and monolayer disruption. Interestingly, the ability to promote thrombin responses was present in EC cultures exposed to 48-96 h of CS even after replating onto non-elastic substrates. In turn, physiologic CS preconditioning (72 h) attenuated thrombin-induced paracellular gap formation and MLC phosphorylation in replated EC cultures. Long-term preconditioning at 18% CS (72 h) increased the content of signaling and contractile proteins including Rho GTPase, MLC, MLC kinase, ZIP kinase, PAR1, caldesmon and HSP27 in the pulmonary microvascular and macrovascular cells. We conclude that short term CS regulates EC permeability via modulation of agonist-induced signaling, whereas long-term CS controls endothelial barrier at both post-translational level and via magnitude-dependent regulation of pulmonary EC phenotype, signaling and contractile protein expression.
Collapse
Affiliation(s)
- Anna A Birukova
- Department of Medicine, University of Chicago, Chicago, IL 60637, USA
| | | | | |
Collapse
|