1
|
Santana Nunez D, Malik AB, Lee Q, Ahn SJ, Coctecon-Murillo A, Lazarko D, Levitan I, Mehta D, Komarova YA. Piezo1 induces endothelial responses to shear stress via soluble adenylyl Cyclase-IP 3R2 circuit. iScience 2023; 26:106661. [PMID: 37168565 PMCID: PMC10164902 DOI: 10.1016/j.isci.2023.106661] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Revised: 01/30/2023] [Accepted: 04/07/2023] [Indexed: 05/13/2023] Open
Abstract
Endothelial cells (ECs) continuously sense and adapt to changes in shear stress generated by blood flow. Here, we show that the activation of the mechanosensitive channel Piezo1 by defined shear forces induces Ca2+ entry into the endoplasmic reticulum (ER) via the ER Ca2+ ATPase pump. This entry is followed by inositol trisphosphate receptor 2 (IP3R2)-elicited ER Ca2+ release into the cytosol. The mechanism of ER Ca2+ release involves the generation of cAMP by soluble adenylyl cyclase (sAC), leading to IP3R2-evoked Ca2+ gating. Depleting sAC or IP3R2 prevents ER Ca2+ release and blocks EC alignment in the direction of flow. Overexpression of constitutively active Akt1 restores the shear-induced alignment of ECs lacking Piezo1 or IP3R2, as well as the flow-induced vasodilation in endothelial restricted Piezo1 knockout mice. These studies describe an unknown Piezo1-cAMP-IP3R2 circuit as an essential mechanism activating Akt signaling and inducing adaptive changes in ECs to laminar flow.
Collapse
Affiliation(s)
- Dianicha Santana Nunez
- Department of Pharmacology and Regenerative Medicine, the Center for Lung and Vascular Biology, University of Illinois College of Medicine, Chicago, IL, USA
| | - Asrar B. Malik
- Department of Pharmacology and Regenerative Medicine, the Center for Lung and Vascular Biology, University of Illinois College of Medicine, Chicago, IL, USA
| | - Quinn Lee
- Department of Pharmacology and Regenerative Medicine, the Center for Lung and Vascular Biology, University of Illinois College of Medicine, Chicago, IL, USA
| | - Sang Joon Ahn
- Department of Medicine, University of Illinois College of Medicine, Chicago, IL, USA
| | - Arnold Coctecon-Murillo
- Department of Pharmacology and Regenerative Medicine, the Center for Lung and Vascular Biology, University of Illinois College of Medicine, Chicago, IL, USA
| | - Dana Lazarko
- Department of Medicine, University of Illinois College of Medicine, Chicago, IL, USA
| | - Irena Levitan
- Department of Medicine, University of Illinois College of Medicine, Chicago, IL, USA
| | - Dolly Mehta
- Department of Pharmacology and Regenerative Medicine, the Center for Lung and Vascular Biology, University of Illinois College of Medicine, Chicago, IL, USA
| | - Yulia A. Komarova
- Department of Pharmacology and Regenerative Medicine, the Center for Lung and Vascular Biology, University of Illinois College of Medicine, Chicago, IL, USA
| |
Collapse
|
2
|
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: 53] [Impact Index Per Article: 53.0] [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
|
3
|
Mathematical modeling of intracellular calcium in presence of receptor: a homeostatic model for endothelial cell. Biomech Model Mechanobiol 2023; 22:217-232. [PMID: 36219362 DOI: 10.1007/s10237-022-01643-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2022] [Accepted: 09/20/2022] [Indexed: 11/02/2022]
Abstract
Calcium is a ubiquitous molecule and second messenger that regulates many cellular functions ranging from exocytosis to cell proliferation at different time scales. In the vasculature, a constant adenosine triphosphate (ATP) concentration is maintained because of ATP released by red blood cells (RBCs). These ATP molecules continuously react with purinergic receptors on the surface of endothelial cells (ECs). Consequently, a cascade of chemical reactions are triggered that result in a transient cytoplasmic calcium (Ca[Formula: see text]), followed by return to its basal concentration. The mathematical models proposed in the literature are able to reproduce the transient peak. However, the trailing concentration is always higher than the basal cytoplasmic Ca[Formula: see text] concentrations, and the Ca[Formula: see text] concentration in endoplasmic reticulum (ER) remains lower than its initial concentration. This means that the intracellular homeostasis is not recovered. We propose, herein, a minimal model of calcium kinetics. We find that the desensitization of EC surface receptors due to phosphorylation and recycling plays a vital role in maintaining calcium homeostasis in the presence of a constant stimulus (ATP). The model is able to capture several experimental observations such as refilling of Ca[Formula: see text] in the ER, variation of cytoplasmic Ca[Formula: see text] transient peak in ECs, the resting cytoplasmic Ca[Formula: see text] concentration, the effect of removing ATP from the plasma on Ca[Formula: see text] homeostasis, and the saturation of cytoplasmic Ca[Formula: see text] transient peak with increase in ATP concentration. Direct confrontation with several experimental results is conducted. This work paves the way for systematic studies on coupling between blood flow and chemical signaling, and should contribute to a better understanding of the relation between (patho)physiological conditions and Ca[Formula: see text] kinetics.
Collapse
|
4
|
Santamaría R, González-Álvarez M, Delgado R, Esteban S, Arroyo AG. Remodeling of the Microvasculature: May the Blood Flow Be With You. Front Physiol 2020; 11:586852. [PMID: 33178049 PMCID: PMC7593767 DOI: 10.3389/fphys.2020.586852] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Accepted: 09/09/2020] [Indexed: 12/12/2022] Open
Abstract
The vasculature ensures optimal delivery of nutrients and oxygen throughout the body, and to achieve this function it must continually adapt to varying tissue demands. Newly formed vascular plexuses during development are immature and require dynamic remodeling to generate well-patterned functional networks. This is achieved by remodeling of the capillaries preserving those which are functional and eliminating other ones. A balanced and dynamically regulated capillary remodeling will therefore ensure optimal distribution of blood and nutrients to the tissues. This is particularly important in pathological contexts in which deficient or excessive vascular remodeling may worsen tissue perfusion and hamper tissue repair. Blood flow is a major determinant of microvascular reshaping since capillaries are pruned when relatively less perfused and they split when exposed to high flow in order to shape the microvascular network for optimal tissue perfusion and oxygenation. The molecular machinery underlying blood flow sensing by endothelial cells is being deciphered, but much less is known about how this translates into endothelial cell responses as alignment, polarization and directed migration to drive capillary remodeling, particularly in vivo. Part of this knowledge is theoretical from computational models since blood flow hemodynamics are not easily recapitulated by in vitro or ex vivo approaches. Moreover, these events are difficult to visualize in vivo due to their infrequency and briefness. Studies had been limited to postnatal mouse retina and vascular beds in zebrafish but new tools as advanced microscopy and image analysis are strengthening our understanding of capillary remodeling. In this review we introduce the concept of remodeling of the microvasculature and its relevance in physiology and pathology. We summarize the current knowledge on the mechanisms contributing to capillary regression and to capillary splitting highlighting the key role of blood flow to orchestrate these processes. Finally, we comment the potential and possibilities that microfluidics offers to this field. Since capillary remodeling mechanisms are often reactivated in prevalent pathologies as cancer and cardiovascular disease, all this knowledge could be eventually used to improve the functionality of capillary networks in diseased tissues and promote their repair.
Collapse
Affiliation(s)
- Ricardo Santamaría
- Department of Vascular Pathophysiology, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - María González-Álvarez
- Department of Molecular Biomedicine, Centro de Investigaciones Biológicas Margarita Salas (CIB-CSIC), Madrid, Spain
| | - Raquel Delgado
- Department of Vascular Pathophysiology, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Sergio Esteban
- Department of Vascular Pathophysiology, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Alicia G. Arroyo
- Department of Vascular Pathophysiology, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
- Department of Molecular Biomedicine, Centro de Investigaciones Biológicas Margarita Salas (CIB-CSIC), Madrid, Spain
| |
Collapse
|
5
|
Wei R, Lunn SE, Tam R, Gust SL, Classen B, Kerr PM, Plane F. Vasoconstrictor stimulus determines the functional contribution of myoendothelial feedback to mesenteric arterial tone. J Physiol 2018; 596:1181-1197. [PMID: 29411383 DOI: 10.1113/jp274797] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2017] [Accepted: 01/17/2018] [Indexed: 12/16/2022] Open
Abstract
KEY POINTS In isolated resistance arteries, endothelial modulation of vasoconstrictor responses to α1 -adrenoceptor agonists occurs via a process termed myoendothelial feedback: localized inositol trisphosphate (InsP3 )-dependent Ca2+ transients activate intermediate conductance Ca2+ -activated K+ (IKCa ) channels, hyperpolarizing the endothelial membrane potential to limit further reductions in vessel diameter. We demonstrate that IKCa channel-mediated myoendothelial feedback limits responses of isolated mesenteric arteries to noradrenaline and nerve stimulation, but not to the thromboxane A2 mimetic U46619 or to increases in intravascular pressure. In contrast, in the intact mesenteric bed, although responses to exogenous noradrenaline were limited by IKCa channel-mediated myoendothelial feedback, release of NO and activation of endothelial small conductance Ca2+ -activated K+ (SKCa ) channels in response to increases in shear stress appeared to be the primary mediators of endothelial modulation of vasoconstriction. We propose that (1) the functional contribution of myoendothelial feedback to arterial tone is determined by the nature of the vasoconstrictor stimulus, and (2) although IKCa channel-mediated myoendothelial feedback may contribute to local control of arterial diameter, in the intact vascular bed, increases in shear stress may be the major stimulus for engagement of the endothelium during vasoconstriction. ABSTRACT Constriction of isolated resistance arteries in response to α1 -adrenoceptor agonists is limited by reciprocal engagement of inhibitory endothelial mechanisms via myoendothelial feedback. In the current model of feedback, agonist stimulation of smooth muscle cells results in localized InsP3 -dependent Ca2+ transients that activate endothelial IKCa channels. The subsequent hyperpolarization of the endothelial membrane potential then feeds back to the smooth muscle to limit further reductions in vessel diameter. We hypothesized that the functional contribution of InsP3 -IKCa channel-mediated myoendothelial feedback to limiting arterial diameter may be influenced by the nature of the vasoconstrictor stimulus. To test this hypothesis, we investigated the functional role of myoendothelial feedback in modulating responses of rat mesenteric resistance arteries to the adrenoceptor agonist noradrenaline, the thromboxane A2 mimetic U46619, increases in intravascular pressure and stimulation of perivascular sympathetic nerves. In isolated arteries, responses to noradrenaline and stimulation of sympathetic nerves, but not to U46619 and increases in intravascular pressure, were modulated by IKCa channel-dependent myoendothelial feedback. In the intact mesenteric bed perfused under conditions of constant flow, responses to exogenous noradrenaline were modulated by myoendothelial feedback, but shear stress-induced release of NO and activation of endothelial SKCa channels appeared to be the primary mediators of endothelial modulation of vasoconstriction to agonists and nerve stimulation. Thus, we propose that myoendothelial feedback may contribute to local control of diameter within arterial segments, but at the level of the intact vascular bed, increases in shear stress may be the major stimulus for engagement of the endothelium during vasoconstriction.
Collapse
Affiliation(s)
- R Wei
- Department of Pharmacology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, T6G 2H7, Canada
| | - S E Lunn
- Department of Pharmacology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, T6G 2H7, Canada
| | - R Tam
- Department of Pharmacology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, T6G 2H7, Canada
| | - S L Gust
- Department of Pharmacology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, T6G 2H7, Canada
| | - B Classen
- Department of Pharmacology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, T6G 2H7, Canada
| | - P M Kerr
- Department of Nursing Science, Faculty of Nursing, MacEwan University, Edmonton, Alberta, T5J 4S2, Canada
| | - F Plane
- Department of Pharmacology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, T6G 2H7, Canada.,Cardiovascular Research Centre, University of Alberta, Edmonton, Alberta, T6G 2H7, Canada
| |
Collapse
|
6
|
Thomas A, Wang S, Sohrabi S, Orr C, He R, Shi W, Liu Y. Characterization of vascular permeability using a biomimetic microfluidic blood vessel model. BIOMICROFLUIDICS 2017; 11:024102. [PMID: 28344727 PMCID: PMC5336476 DOI: 10.1063/1.4977584] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/30/2016] [Accepted: 02/08/2017] [Indexed: 05/18/2023]
Abstract
The inflammatory response in endothelial cells (ECs) leads to an increase in vascular permeability through the formation of gaps. However, the dynamic nature of vascular permeability and external factors involved is still elusive. In this work, we use a biomimetic blood vessel (BBV) microfluidic model to measure in real-time the change in permeability of the EC layer under culture in physiologically relevant flow conditions. This platform studies the dynamics and characterizes vascular permeability when the EC layer is triggered with an inflammatory agent using tracer molecules of three different sizes, and the results are compared to a transwell insert study. We also apply an analytical model to compare the permeability data from the different tracer molecules to understand the physiological and bio-transport significance of endothelial permeability based on the molecule of interest. A computational model of the BBV model is also built to understand the factors influencing transport of molecules of different sizes under flow. The endothelial monolayer cultured under flow in the BBV model was treated with thrombin, a serine protease that induces a rapid and reversible increase in endothelium permeability. On analysis of permeability data, it is found that the transport characteristics for fluorescein isothiocyanate (FITC) dye and FITC Dextran 4k Da molecules are similar in both BBV and transwell models, but FITC Dextran 70k Da molecules show increased permeability in the BBV model as convection flow (Peclet number > 1) influences the molecule transport in the BBV model. We also calculated from permeability data the relative increase in intercellular gap area during thrombin treatment for ECs in the BBV and transwell insert models to be between 12% and 15%. This relative increase was found to be within range of what we quantified from F-actin stained EC layer images. The work highlights the importance of incorporating flow in in vitro vascular models, especially in studies involving transport of large size objects such as antibodies, proteins, nano/micro particles, and cells.
Collapse
Affiliation(s)
- Antony Thomas
- Bioengineering Program, Lehigh University , Bethlehem, Pennsylvania 18015, USA
| | - Shunqiang Wang
- Department of Mechanical Engineering and Mechanics, Lehigh University , Bethlehem, Pennsylvania 18015, USA
| | - Salman Sohrabi
- Department of Mechanical Engineering and Mechanics, Lehigh University , Bethlehem, Pennsylvania 18015, USA
| | - Colin Orr
- Bioengineering Program, Lehigh University , Bethlehem, Pennsylvania 18015, USA
| | - Ran He
- Department of Mechanical Engineering and Mechanics, Lehigh University , Bethlehem, Pennsylvania 18015, USA
| | - Wentao Shi
- Bioengineering Program, Lehigh University , Bethlehem, Pennsylvania 18015, USA
| | | |
Collapse
|
7
|
Abstract
Fluid shear stress is an important environmental cue that governs vascular physiology and pathology, but the molecular mechanisms that mediate endothelial responses to flow are only partially understood. Gating of ion channels by flow is one mechanism that may underlie many of the known responses. Here, we review the literature on endothelial ion channels whose activity is modulated by flow with an eye toward identifying important questions for future research.
Collapse
Affiliation(s)
- Kristin A Gerhold
- Department of Internal Medicine (Cardiology), Yale Cardiovascular Research Center, Yale University, New Haven, Connecticut; and
| | - Martin A Schwartz
- Department of Internal Medicine (Cardiology), Yale Cardiovascular Research Center, Yale University, New Haven, Connecticut; and Departments of Cell Biology and Biomedical Engineering, Yale University, New Haven, Connecticut
| |
Collapse
|
8
|
Scheitlin CG, Julian JA, Shanmughapriya S, Madesh M, Tsoukias NM, Alevriadou BR. Endothelial mitochondria regulate the intracellular Ca2+ response to fluid shear stress. Am J Physiol Cell Physiol 2016; 310:C479-90. [PMID: 26739489 DOI: 10.1152/ajpcell.00171.2015] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2015] [Accepted: 01/04/2016] [Indexed: 02/04/2023]
Abstract
Shear stress is known to stimulate an intracellular free calcium concentration ([Ca(2+)]i) response in vascular endothelial cells (ECs). [Ca(2+)]i is a key second messenger for signaling that leads to vasodilation and EC survival. Although it is accepted that the shear-induced [Ca(2+)]i response is, in part, due to Ca(2+) release from the endoplasmic reticulum (ER), the role of mitochondria (second largest Ca(2+) store) is unknown. We hypothesized that the mitochondria play a role in regulating [Ca(2+)]i in sheared ECs. Cultured ECs, loaded with a Ca(2+)-sensitive fluorophore, were exposed to physiological levels of shear stress. Shear stress elicited [Ca(2+)]i transients in a percentage of cells with a fraction of them displaying oscillations. Peak magnitudes, percentage of oscillating ECs, and oscillation frequencies depended on the shear level. [Ca(2+)]i transients/oscillations were present when experiments were conducted in Ca(2+)-free solution (plus lanthanum) but absent when ECs were treated with a phospholipase C inhibitor, suggesting that the ER inositol 1,4,5-trisphosphate receptor is responsible for the [Ca(2+)]i response. Either a mitochondrial uncoupler or an electron transport chain inhibitor, but not a mitochondrial ATP synthase inhibitor, prevented the occurrence of transients and especially inhibited the oscillations. Knockdown of the mitochondrial Ca(2+) uniporter also inhibited the shear-induced [Ca(2+)]i transients/oscillations compared with controls. Hence, EC mitochondria, through Ca(2+) uptake/release, regulate the temporal profile of shear-induced ER Ca(2+) release. [Ca(2+)]i oscillation frequencies detected were within the range for activation of mechanoresponsive kinases and transcription factors, suggesting that dysfunctional EC mitochondria may contribute to cardiovascular disease by deregulating the shear-induced [Ca(2+)]i response.
Collapse
Affiliation(s)
- Christopher G Scheitlin
- Departments of Biomedical Engineering and Internal Medicine, Division of Cardiovascular Medicine, and Davis Heart and Lung Research Institute, The Ohio State University, Columbus, Ohio
| | - Justin A Julian
- Departments of Biomedical Engineering and Internal Medicine, Division of Cardiovascular Medicine, and Davis Heart and Lung Research Institute, The Ohio State University, Columbus, Ohio
| | - Santhanam Shanmughapriya
- Department of Medical Genetics and Molecular Biochemistry and Center for Translational Medicine, Temple University, Philadelphia, Pennsylvania; and
| | - Muniswamy Madesh
- Department of Medical Genetics and Molecular Biochemistry and Center for Translational Medicine, Temple University, Philadelphia, Pennsylvania; and
| | - Nikolaos M Tsoukias
- Department of Biomedical Engineering, Florida International University, Miami, Florida
| | - B Rita Alevriadou
- Departments of Biomedical Engineering and Internal Medicine, Division of Cardiovascular Medicine, and Davis Heart and Lung Research Institute, The Ohio State University, Columbus, Ohio;
| |
Collapse
|
9
|
Tatsumi H, Toyota M, Furuichi T, Sokabe M. Calcium mobilizations in response to changes in the gravity vector in Arabidopsis seedlings: possible cellular mechanisms. PLANT SIGNALING & BEHAVIOR 2014; 9:e29099. [PMID: 25763612 PMCID: PMC4203510 DOI: 10.4161/psb.29099] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/20/2014] [Revised: 05/02/2014] [Accepted: 05/02/2014] [Indexed: 05/11/2023]
Abstract
Gravity influences the growth direction of higher plants. Changes in the gravity vector (gravistimulation) immediately promote the increase in the cytoplasmic free calcium ion concentration ([Ca(2+)]c) in Arabidopsis (Arabidopsis thaliana) seedlings. When the seedlings are gravistimulated by reorientation at 180°, a transient two peaked (biphasic) [Ca(2+)]c-increase arises in their hypocotyl and petioles. Parabolic flights (PFs) can generate a variety of gravity-stimuli, and enables us to measure gravity-induced [Ca(2+)]c-increases without specimen rotation, which demonstrate that Arabidopsis seedlings possess a rapid gravity-sensing mechanism linearly transducing a wide range of gravitational changes into Ca(2+) signals on a sub-second timescale. Hypergravity by centrifugation (20 g or 300 g) also induces similar transient [Ca(2+)]c-increases. In this review, we propose models for possible cellular processes of the garavi-stimulus-induced [Ca(2+)]c-increase, and evaluate those by examining whether the model fits well with the kinetic parameters derived from the [Ca(2+)]c-increases obtained by applying gravistimulus with different amplitudes and time sequences.
Collapse
Affiliation(s)
- Hitoshi Tatsumi
- Nagoya University Graduate School of Medicine; Nagoya, Japan
| | - Masatsugu Toyota
- Department of Botany; University of Wisconsin; Madison, WI USA
- Precursory Research for Embryonic Science and Technology (PRESTO); Japan Science and Technology Agency (JST); Kawaguchi, Saitama, Japan
| | - Takuya Furuichi
- Department of Health and Nutrition; Gifu Women’s University; Gifu, Japan
| | - Masahiro Sokabe
- Nagoya University Graduate School of Medicine; Nagoya, Japan
| |
Collapse
|
10
|
Melchior B, Frangos JA. Gαq/11-mediated intracellular calcium responses to retrograde flow in endothelial cells. Am J Physiol Cell Physiol 2012; 303:C467-73. [PMID: 22700794 DOI: 10.1152/ajpcell.00117.2012] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Disturbed flow patterns, including reversal in flow direction, are key factors in the development of dysfunctional endothelial cells (ECs) and atherosclerotic lesions. An almost immediate response of ECs to fluid shear stress is the increase in cytosolic calcium concentration ([Ca(2+)](i)). Whether the source of [Ca(2+)](i) is extracellular, released from Ca(2+) intracellular stores, or both is still undefined, though it is likely dependent on the nature of forces involved. We have previously shown that a change in flow direction (retrograde flow) on a flow-adapted endothelial monolayer induces the remodeling of the cell-cell junction along with a dramatic [Ca(2+)](i) burst compared with cells exposed to unidirectional or orthograde flow. The heterotrimeric G protein-α q and 11 subunit (Gα(q/11)) is a likely candidate in effecting shear-induced increases in [Ca(2+)](i) since its expression is enriched at the junction and has been previously shown to be activated within seconds after onset of flow. In flow-adapted human ECs, we have investigated to what extent the Gα(q/11) pathway mediates calcium dynamics after reversal in flow direction. We observed that the elapsed time to peak [Ca(2+)](i) response to a 10 dyn/cm(2) retrograde shear stress was increased by 11 s in cells silenced with small interfering RNA directed against Gα(q/11). A similar lag in [Ca(2+)](i) transient was observed after cells were treated with the phospholipase C (PLC)-βγ inhibitor, U-73122, or the phosphatidylinositol-specific PLC inhibitor, edelfosine, compared with controls. Lower levels of inositol 1,4,5-trisphosphate accumulation seconds after the onset of flow correlated with the increased lag in [Ca(2+)](i) responses observed with the different treatments. In addition, inhibition of the inositol 1,4,5-trisphosphate receptor entirely abrogated flow-induced [Ca(2+)](i). Taken together, our results identify the Gα(q/11)-PLC pathway as the initial trigger for retrograde flow-induced endoplasmic reticulum calcium store release, thereby offering a novel approach to regulating EC dysfunctions in regions subjected to the reversal of blood flow.
Collapse
Affiliation(s)
- Benoît Melchior
- La Jolla Bioengineering Institute, San Diego, California, USA
| | | |
Collapse
|
11
|
Flow-dependent mass transfer may trigger endothelial signaling cascades. PLoS One 2012; 7:e35260. [PMID: 22558132 PMCID: PMC3338739 DOI: 10.1371/journal.pone.0035260] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2011] [Accepted: 03/14/2012] [Indexed: 01/08/2023] Open
Abstract
It is well known that fluid mechanical forces directly impact endothelial signaling pathways. But while this general observation is clear, less apparent are the underlying mechanisms that initiate these critical signaling processes. This is because fluid mechanical forces can offer a direct mechanical input to possible mechanotransducers as well as alter critical mass transport characteristics (i.e., concentration gradients) of a host of chemical stimuli present in the blood stream. However, it has recently been accepted that mechanotransduction (direct mechanical force input), and not mass transfer, is the fundamental mechanism for many hemodynamic force-modulated endothelial signaling pathways and their downstream gene products. This conclusion has been largely based, indirectly, on accepted criteria that correlate signaling behavior and shear rate and shear stress, relative to changes in viscosity. However, in this work, we investigate the negative control for these criteria. Here we computationally and experimentally subject mass-transfer limited systems, independent of mechanotransduction, to the purported criteria. The results showed that the negative control (mass-transfer limited system) produced the same trends that have been used to identify mechanotransduction-dominant systems. Thus, the widely used viscosity-related shear stress and shear rate criteria are insufficient in determining mechanotransduction-dominant systems. Thus, research should continue to consider the importance of mass transfer in triggering signaling cascades.
Collapse
|
12
|
Sobolewski P, Kandel J, Klinger AL, Eckmann DM. Air bubble contact with endothelial cells in vitro induces calcium influx and IP3-dependent release of calcium stores. Am J Physiol Cell Physiol 2011; 301:C679-86. [PMID: 21633077 PMCID: PMC3273994 DOI: 10.1152/ajpcell.00046.2011] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2011] [Accepted: 05/31/2011] [Indexed: 01/05/2023]
Abstract
Gas embolism is a serious complication of decompression events and clinical procedures, but the mechanism of resulting injury remains unclear. Previous work has demonstrated that contact between air microbubbles and endothelial cells causes a rapid intracellular calcium transient and can lead to cell death. Here we examined the mechanism responsible for the calcium rise. Single air microbubbles (50-150 μm), trapped at the tip of a micropipette, were micromanipulated into contact with individual human umbilical vein endothelial cells (HUVECs) loaded with Fluo-4 (a fluorescent calcium indicator). Changes in intracellular calcium were then recorded via epifluorescence microscopy. First, we confirmed that HUVECs rapidly respond to air bubble contact with a calcium transient. Next, we examined the involvement of extracellular calcium influx by conducting experiments in low calcium buffer, which markedly attenuated the response, or by pretreating cells with stretch-activated channel blockers (gadolinium chloride or ruthenium red), which abolished the response. Finally, we tested the role of intracellular calcium release by pretreating cells with an inositol 1,4,5-trisphosphate (IP3) receptor blocker (xestospongin C) or phospholipase C inhibitor (neomycin sulfate), which eliminated the response in 64% and 67% of cases, respectively. Collectively, our results lead us to conclude that air bubble contact with endothelial cells causes an influx of calcium through a stretch-activated channel, such as a transient receptor potential vanilloid family member, triggering the release of calcium from intracellular stores via the IP3 pathway.
Collapse
Affiliation(s)
- Peter Sobolewski
- Dept. of Anesthesiology and Critical Care, Univ. of Pennsylvania, Philadelphia, 19104-4283, USA
| | | | | | | |
Collapse
|
13
|
Patrick CW, McIntire LV. Fluid Shear Stress Effects on Endothelial Cell Cytosolic pH. ACTA ACUST UNITED AC 2011; 1:53-70. [PMID: 19877915 DOI: 10.1089/ten.1995.1.53] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Fluid flow can modulate endothelial cell intracellular pH (pH(i)). Venous and arterial shear stresses of 1.4 and 14 dyn/cm2, respectively, induced intracellular acidification. The kinetics of the process and magnitude of acidification were dependent on the level of shear stress. Endothelial cells exposed to a venous shear stress were able to recover from the acidification, whereas cells exposed to an arterial shear stress remained acidic. Addition of SITS (1 mM), a HCO(3) (-)/CI(-) exchange inhibitor, greatly reduced the shear stress induced acidification, suggesting that the HCO(3) (-)/C1(-) exchanger is activated by shear stress. Shear stress may activate the exchanger by lowering the [HCO(3) (-)] at the cell surface via convective mass transfer. Altering the HCO(3) (-) gradient across the cell membrane activates the exchanger and, as a consequence, results in intracellular acidification. Perfusion with media containing ATP (10 microM) altered the kinetics of flow-induced acidification observed at both shear stress levels. ATP modulation of pH(i) may be coupled to the rise in [Ca(2+)](j) known to occur with ATP stimulation. To summarize, media perfusion induces intracellular acidification in endothelial cells, and there is evidence to suggest that pH(i) may serve as a second messenger to modulate flow associated changes in endothelial cell metabolism.
Collapse
Affiliation(s)
- C W Patrick
- Cox Laboratory for Biomedical Engineering, Institute of Biosciences and Bioengineering, Rice University, Houston, Texas 77005-1892
| | | |
Collapse
|
14
|
Abstract
The physiological mechanisms that regulate adaptive plasticity of clonal organisms are key to their success in changing environments. Here, we review the mechanisms that regulate morphological plasticity of colonial hydrozoans. There is a heritable, genetic basis to colony form, but environmentally-induced plasticity and self-reinforcing developmental physiology explain much of total phenotypic variance. Morphological development of colonial hydrozoans emerges from interactions among (1) behaviors which drive gastrovascular transport, (2) architecture of the gastrovascular system that determines hydrodynamic characteristics of vascular flow, and, (3) gene products that vary in response to physiological signals provided by gastrovascular transport. Several morphogenetic signaling mechanisms have been identified, including, reactive oxygen species and nutrient concentrations in the hydroplasm, and hydromechanical forces associated with gastrovascular transport. We present a conceptual model of the interacting forces that drive hydrozoan morphological development. Several avenues for future research are suggested by the synthesis of information from prior studies of hydrozoans. Elucidating the morphogenetic signaling pathways responsive to metabolites or hydromechanical forces and the epigenetic effect of vascular architecture on colony form may give new insight into the self-maintenance of indeterminately growing and continuously developing vascular systems.
Collapse
Affiliation(s)
- S R Dudgeon
- Department of Biology, California State University, Northridge, CA, USA
| | | |
Collapse
|
15
|
Vinci B, Murphy E, Iori E, Marescotti MC, Avogaro A, Ahluwalia A. Flow-regulated glucose and lipid metabolism in adipose tissue, endothelial cell and hepatocyte cultures in a modular bioreactor. Biotechnol J 2010; 5:618-26. [PMID: 20518065 DOI: 10.1002/biot.201000009] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
Static cell culture has serious limitations in its ability to represent cellular behaviour within a live organism. In vivo, cells are constantly exposed to the flow of bodily fluids and contact with other cell types. Bioreactors provide the opportunity to study cells in an environment that more closely resembles the in vivo setting because cell cultures can be exposed to dynamic flow in contact with or in proximity to other cell types. In this study we compared the metabolic profile of a dynamic cell culture system to that of a static cell culture in three different cellular phenotypes: adipocytes, endothelial cells and hepatocytes. Albumin, glucose, free fatty acids, glycerol, and lactate were measured over 48 h. We show that all three cell types have increased glucose uptake in the presence of flow; lactate release was also significantly affected. We provide robust evidence that the presence of flow significantly modifies cellular metabolism. While flow provides a more uniform nutrient distribution and increases metabolite turnover, our results indicate that different cell types have specific metabolic responses to flow, suggesting cell-specific flow-regulated activation of metabolite signalling pathways.
Collapse
Affiliation(s)
- Bruna Vinci
- Centro Interdipartimentale di Ricerca E. Piaggio, Faculty of Engineering, University of Pisa, Pisa, Italy
| | | | | | | | | | | |
Collapse
|
16
|
Koller A, Kaley G. Shear Stress Dependent Regulation of Vascular Resistance in Health and Disease: Role of Endothelium. ACTA ACUST UNITED AC 2009. [DOI: 10.3109/10623329609024701] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
17
|
Van Ijzendoorn SC, Heemskerk JW, Reutelingsperger CP. Interactions between Endothelial Cells and Blood Platelets. ACTA ACUST UNITED AC 2009. [DOI: 10.3109/10623329509053385] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
|
18
|
Brailoiu E, Jiang X, Brailoiu GC, Yang J, Chang JK, Wang H, Dun NJ. State-dependent calcium mobilization by urotensin-II in cultured human endothelial cells. Peptides 2008; 29:721-6. [PMID: 18314227 PMCID: PMC2387077 DOI: 10.1016/j.peptides.2007.12.019] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/04/2007] [Revised: 12/11/2007] [Accepted: 12/14/2007] [Indexed: 02/07/2023]
Abstract
Human endothelial cells express urotensin-II (U-II) as well as its receptor GPR14. Using microfluorimetric techniques, the effect of human U-II on cytosolic Ca2+ concentrations [Ca2+]i in cultured human aortic endothelial cells (HAECs) loaded with Fura-2 was evaluated in static or flow conditions. Under the static state, U-II (100 nM) abolished spontaneous Ca2+ oscillations, which occurred in a population of cultured HAEC. Similarly, U-II reduced thrombin-, but not ATP-induced calcium responses, suggesting that the peptide does not alter the Gq/11/IP3 pathway; rather, it modifies the coupling between protease-activated receptors and Gq/11/IP3. Under the flow condition, U-II (1, 10 and 100 nM) produced a dose-dependent increase in [Ca2+]i, which was subjected to desensitization. The result demonstrates a state-dependent effect of U-II in cultured HAEC, which may explain the variable responses to U-II under different experimental conditions.
Collapse
Affiliation(s)
- Eugen Brailoiu
- Department of Pharmacology, 3420 N. Broad Street, Temple University School of Medicine, Philadelphia, PA 19140, USA.
| | | | | | | | | | | | | |
Collapse
|
19
|
Kang HG, Shim EB, Chang KS. A new multiphysics model for the physiological responses of vascular endothelial cells to fluid shear stress. J Physiol Sci 2007; 57:299-309. [PMID: 17963593 DOI: 10.2170/physiolsci.rp005407] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2007] [Accepted: 10/28/2007] [Indexed: 11/05/2022]
Abstract
Vascular endothelial cell (VEC) responds to wall shear stress that has not only spatial variation, but also temporal gradient. To simplify the problem, we first studied how the calcium dynamics of VEC responded to the steady wall shear stress of varying magnitude in a stenosed artery. We then studied how the VEC responded to the periodic shear stress that had temporal variation, as in the pulsatile blood flow. To investigate the multiphysics model of VEC in vitro, we used a mathematical model for intracellular calcium dynamics and a computational fluid dynamics (CFD) method for arterial wall shear stress, either steady or periodic. The CFD results showed that for the steady stenotic flow, the wall shear stress in the recirculating flow was lower than the threshold value, 4 dyne/cm(2), at two particular points: flow separation and flow reattachment. For these subthreshold shear stresses, the peak value of the transient calcium response did not hit the normal saturated level, but reached a reduced magnitude. We investigated the effect of severity of stenosis (SOS) of the stenosed artery. For the pulsatile flow, the so-called shear stress slew rate or the temporal gradient of the first upsurge of the periodic flow was an important factor for the VEC calcium dynamics. The calcium response had a finite range of parameter for SOS and shear stress slew rate in which the calcium response was more sensitive than elsewhere, showing a sigmoid pattern.
Collapse
Affiliation(s)
- Hyun Goo Kang
- School of Mechanical, Aerospace and Systems Engineering, Korea Advanced Institute of Science and Technology, Daejeon 305-701, Korea
| | | | | |
Collapse
|
20
|
Miyazaki T, Honda K, Ohata H. Modulation of Ca2+ transients in cultured endothelial cells in response to fluid flow through alphav integrin. Life Sci 2007; 81:1421-30. [PMID: 17931663 DOI: 10.1016/j.lfs.2007.08.041] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2007] [Revised: 08/28/2007] [Accepted: 08/31/2007] [Indexed: 11/30/2022]
Abstract
In order to determine whether integrin dynamics is associated with intracellular Ca(2+) concentration ([Ca(2+)](i)) mobilization in ECs in response to hemodynamic forces, changes in [Ca(2+)](i) in fluo-4-loaded cultured bovine aortic endothelial cells (BAECs) under fluid flow conditions were visualized employing laser scanning confocal microscopy. Following the onset of flow stimulus, transient increases in [Ca(2+)](i) occurred several times in individual BAECs during the 30-min observation period. The frequency of these [Ca(2+)](i) transients was clearly reduced by the application of an integrin antagonist (GRGDSP peptide). Furthermore, treatment of cells with an integrin activator (Mn(2+)) resulted in reduction of peak [Ca(2+)](i) levels and elevated frequency, which was markedly rescued upon GRGDSP administration. In contrast, an actin de-polymerizing agent (cytochalasin D) exerted no inhibitory effects; rather, cytochalasin D more likely facilitated [Ca(2+)](i) transients. Moreover, [Ca(2+)](i) transients, which were suppressed by short interference RNA-induced silencing of alphav integrin, exhibited greater frequently in cells cultured on vitronectin substratum in comparison with those cultured on fibronectin or collagen substratum. Either removal of extracellular Ca(2+), application of an inhibitor of endoplasmic reticulum Ca(2+)-ATPase (thapsigargin) or non-selective cation channel blocker (La(3+)) inhibited the [Ca(2+)](i) transients. Additionally, [Ca(2+)](i) transients were attenuated by extracellular signal-regulated kinase (ERK) kinase inhibitor (U0126); in contrast, [Ca(2+)](i) transients were unaffected by tyrosine kinase inhibitor (genistein) or phosphatidylinositol 3-kinase (PI3K) inhibitor (LY294002). Therefore, our findings revealed that alphav integrin dynamics modulates the frequency of flow-induced [Ca(2+)](i) transients in BAECs in an ERK-dependent fashion.
Collapse
Affiliation(s)
- Takuro Miyazaki
- Department of Pharmacology, School of Pharmaceutical Sciences, Showa University, Hatanodai, Shinagawa-ku, Tokyo 142-8555, Japan.
| | | | | |
Collapse
|
21
|
Miyazaki T, Honda K, Ohata H. Requirement of Ca2+ influx- and phosphatidylinositol 3-kinase-mediated m-calpain activity for shear stress-induced endothelial cell polarity. Am J Physiol Cell Physiol 2007; 293:C1216-25. [PMID: 17596297 DOI: 10.1152/ajpcell.00083.2007] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Proteolytic activity in sheared human umbilical vein endothelial cells (HUVECs) was measured using a fluorogenic substrate and laser scanning confocal microscopy to clarify the key role of an intracellular Ca(2+)-sensitive protease, calpain, in these cells in response to shear stress. Within physiological shear range, activity in the cells was enhanced in shear-dependent fashion. Short interfering RNA-induced silencing of m-calpain, but not of micro-calpain, suppressed the activity. Either removal of extracellular Ca(2+) or application of an intracellular Ca(2+) chelator (BAPTA/AM) or nonselective cation channel blocker (Gd(3+)) reduced proteolytic activity. Furthermore, activity was suppressed by phosphatidylinositol bisphosphate (PIP(2)) chelator (neomycin) or phosphatidylinositol 3-kinase (PI3K) inhibitor (LY294002); in contrast, activity, which was partially inhibited by ERK kinase inhibitor (U0126, PD98059), was unaffected by PLC inhibitor (U73122). Moreover, Akt phosphorylation downstream of PI3K, which was elicited by shear, was attenuated by neomycin but not by calpain inhibitor (calpeptin). Following assessment of shear stress-induced focal adhesion (FA) and cytoskeletal dynamics using interference reflection/green fluorescence protein-actin microscopy, we found that either calpain or PI3K inhibition impaired shear stress-induced polarization of FAs via stabilization of FA structures. Additionally, HUVEC alignment and cytoskeletal remodeling, which was accompanied by calpain-mediated cleavage of vinculin and talin, were also elicited by prolonged application of shear and impaired by m-calpain knockdown. Thus, these results revealed that physiological shear stress elicits Ca(2+) influx-sensitive activation of m-calpain in HUVECs. This activity is facilitated primarily through the PI3K pathway; furthermore, it is essential for subsequent FA reorganization and cell alignment under shear conditions.
Collapse
Affiliation(s)
- Takuro Miyazaki
- Department of Pharmacology, School of Pharmaceutical Sciences, Showa University, Tokyo, Japan.
| | | | | |
Collapse
|
22
|
Köhler R, Hoyer J. Role of TRPV4 in the Mechanotransduction of Shear Stress in Endothelial Cells. TRP ION CHANNEL FUNCTION IN SENSORY TRANSDUCTION AND CELLULAR SIGNALING CASCADES 2006. [DOI: 10.1201/9781420005844.ch27] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
|
23
|
Hong D, Jaron D, Buerk DG, Barbee KA. Heterogeneous response of microvascular endothelial cells to shear stress. Am J Physiol Heart Circ Physiol 2006; 290:H2498-508. [PMID: 16415079 DOI: 10.1152/ajpheart.00828.2005] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
We investigated changes in calcium concentration in cultured bovine aortic endothelial cells (BAECs) and rat adrenomedulary endothelial cells (RAMECs, microvascular) in response to different levels of shear stress. In BAECs, the onset of shear stress elicited a transient increase in intracellular calcium concentration that was spatially uniform, synchronous, and dose dependent. In contrast, the response of RAMECs was heterogeneous in time and space. Shear stress induced calcium waves that originated from one or several cells and propagated to neighboring cells. The number and size of the responding groups of cells did not depend on the magnitude of shear stress or the magnitude of the calcium change in the responding cells. The initiation and the propagation of calcium waves in RAMECs were significantly suppressed under conditions in which either purinergic receptors were blocked by suramin or extracellular ATP was degraded by apyrase. Exogenously applied ATP produced similarly heterogeneous responses. The number of responding cells was dependent on ATP concentration, but the magnitude of the calcium change was not. Our data suggest that shear stress stimulates RAMECs to release ATP, causing the increase in intracellular calcium concentration via purinergic receptors in cells that are heterogeneously sensitive to ATP. The propagation of the calcium signal is also mediated by ATP, and the spatial pattern suggests a locally elevated ATP concentration in the vicinity of the initially responding cells.
Collapse
Affiliation(s)
- D Hong
- School of Biomedical Engineering, Science, and Health Systems, Drexel University, Philadelphia, PA 19104, USA
| | | | | | | |
Collapse
|
24
|
Shin J, Jo H, Park H. Caveolin-1 is transiently dephosphorylated by shear stress-activated protein tyrosine phosphatase mu. Biochem Biophys Res Commun 2006; 339:737-41. [PMID: 16325778 DOI: 10.1016/j.bbrc.2005.11.077] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2005] [Accepted: 11/14/2005] [Indexed: 02/07/2023]
Abstract
Endothelial cells are subjected to hemodynamic shear stress, which regulates multiple vascular functions partially by the caveolin-1-dependent mechanisms. Caveolin-1 is a principal protein in the plasma membrane microdomains called caveolae and interacts with various signaling molecules. Recently, caveolin-1 was elucidated to be phosphorylated on tyrosine 14. However, it is not known how phosphorylation of caveolin-1 is controlled in endothelium. In this study, we found that caveolin-1 is phosphorylated by p38 mitogen-activated protein kinase (MAPK) under a static condition. When endothelial cells were exposed to shear stress, caveolin-1 was transiently dephosphorylated. Since the activity of p38 MAPK was not affected by shear stress, the shear-dependent dephosphorylation of caveolin-1 was not mediated by p38 MAPK. Of interest, sodium orthovanadate, an inhibitor for phosphatases, blocked the shear-dependent dephosphorylation of caveolin-1. We also observed that protein tyrosine phosphatase mu was transiently activated by shear stress, suggesting its role in the dephosphorylation of caveolin-1.
Collapse
Affiliation(s)
- Jaeyoung Shin
- Department of Molecular Biology and Institute of Nanosensor and Biotechnology, Dankook University, San 8, Hannam-dong, Yongsan-ku, Seoul 140-714, Republic of Korea
| | | | | |
Collapse
|
25
|
Abstract
The microvascular endothelial cell monolayer localized at the critical interface between the blood and vessel wall has the vital functions of regulating tissue fluid balance and supplying the essential nutrients needed for the survival of the organism. The endothelial cell is an exquisite “sensor” that responds to diverse signals generated in the blood, subendothelium, and interacting cells. The endothelial cell is able to dynamically regulate its paracellular and transcellular pathways for transport of plasma proteins, solutes, and liquid. The semipermeable characteristic of the endothelium (which distinguishes it from the epithelium) is crucial for establishing the transendothelial protein gradient (the colloid osmotic gradient) required for tissue fluid homeostasis. Interendothelial junctions comprise a complex array of proteins in series with the extracellular matrix constituents and serve to limit the transport of albumin and other plasma proteins by the paracellular pathway. This pathway is highly regulated by the activation of specific extrinsic and intrinsic signaling pathways. Recent evidence has also highlighted the importance of the heretofore enigmatic transcellular pathway in mediating albumin transport via transcytosis. Caveolae, the vesicular carriers filled with receptor-bound and unbound free solutes, have been shown to shuttle between the vascular and extravascular spaces depositing their contents outside the cell. This review summarizes and analyzes the recent data from genetic, physiological, cellular, and morphological studies that have addressed the signaling mechanisms involved in the regulation of both the paracellular and transcellular transport pathways.
Collapse
Affiliation(s)
- Dolly Mehta
- Center of Lung and Vascular Biology, Dept. of Pharmacology (M/C 868), University of Illinois, 835 S. Wolcott Avenue, Chicago, IL 60612, USA
| | | |
Collapse
|
26
|
Cuvelier SL, Paul S, Shariat N, Colarusso P, Patel KD. Eosinophil adhesion under flow conditions activates mechanosensitive signaling pathways in human endothelial cells. ACTA ACUST UNITED AC 2005; 202:865-76. [PMID: 16172263 PMCID: PMC2212932 DOI: 10.1084/jem.20041315] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Leukocyte transmigration can be affected by shear stress; however, the mechanisms by which shear stress modulates transmigration are unknown. We found that adhesion of eosinophils or an eosinophilic cell line to intereukin 4–stimulated endothelial cells led to a shear-dependent increase in endothelial cell intracellular calcium and increased phosphorylation of extracellular signal-regulated kinase (ERK) 2, but not c-Jun NH2-terminal kinase or p38 mitogen-activated protein kinase. Latex beads coated with antibodies were used to characterize the role of specific endothelial cell surface molecules in initiating signaling under shear conditions. We found that ligation of either vascular cell adhesion molecule–1 or E-selectin, but not major histocompatibility complex class I, induced a shear-dependent increase in ERK2 phosphorylation in cytokine-stimulated endothelial cells. Disassembly of the actin cytoskeleton with latrunculin A prevented ERK2 phosphorylation after adhesion under flow conditions, supporting a role for the cytoskeleton in mechanosensing. Rapid phosphorylation of focal adhesion kinase and paxillin occurred under identical conditions, suggesting that focal adhesions were also involved in mechanotransduction. Finally, we found that Rho-associated protein kinase and calpain were both critical in the subsequent transendothelial migration of eosinophils under flow conditions. These data suggest that ligation of leukocyte adhesion molecules under flow conditions leads to mechanotransduction in endothelial cells, which can regulate subsequent leukocyte trafficking.
Collapse
Affiliation(s)
- Susan L Cuvelier
- Department of Physiology and Biophysics, Immunology Research Group, University of Calgary, Calgary, Alberta, Canada T2N 4N1
| | | | | | | | | |
Collapse
|
27
|
Ahmmed GU, Malik AB. Functional role of TRPC channels in the regulation of endothelial permeability. Pflugers Arch 2005; 451:131-42. [PMID: 15988589 DOI: 10.1007/s00424-005-1461-z] [Citation(s) in RCA: 75] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2005] [Accepted: 04/28/2005] [Indexed: 12/25/2022]
Abstract
The endothelial cells (ECs) form a semipermeable barrier between the blood and the tissue. An important function of the endothelium is to maintain the integrity of the barrier function of the vessel wall. Ca(2+) signaling in ECs plays a key role in maintaining the barrier integrity. Transient receptor potential canonical (TRPC) channels are mammalian homologs of Drosophila TRP Ca(2+)-permeable channels expressed in EC. TRPC channels are thought to function as a Ca(2+) entry channel operated by store-depletion as well as receptor-activated channels in a variety of cell types, including ECs. Inflammatory mediators such as thrombin, histamine, bradykinin, and others increase endothelial permeability by actin polymerization-dependent EC rounding and formation of inter-endothelial gaps, a process critically dependent on the increase in EC cytosolic [Ca(2+)] ([Ca(2+)](i)). Increase in endothelial permeability depends on both intracellular Ca(2+) release and extracellular Ca(2+) entry through TRPC channels. This review summarizes recent findings on the role of TRPC channels in the mechanism of Ca(2+) entry in ECs, and, in particular, the role of TRPC channels in regulating endothelial barrier function.
Collapse
Affiliation(s)
- Gias U Ahmmed
- Department of Pharmacology and the Center for Lung and Vascular Biology, The University of Illinois, College of Medicine, 835 S Wolcott Avenue, Chicago, IL 60612, USA
| | | |
Collapse
|
28
|
Ridge KM, Linz L, Flitney FW, Kuczmarski ER, Chou YH, Omary MB, Sznajder JI, Goldman RD. Keratin 8 phosphorylation by protein kinase C delta regulates shear stress-mediated disassembly of keratin intermediate filaments in alveolar epithelial cells. J Biol Chem 2005; 280:30400-5. [PMID: 15972820 DOI: 10.1074/jbc.m504239200] [Citation(s) in RCA: 106] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Phosphorylation of keratin intermediate filaments (IF) is known to affect their assembly state and organization; however, little is known about the mechanisms regulating keratin phosphorylation. In this study, we demonstrate that shear stress, but not stretch, causes disassembly of keratin IF in lung alveolar epithelial cells (AEC) and that this disassembly is regulated by protein kinase C delta-mediated phosphorylation of keratin 8 (K8) Ser-73. Specifically, in AEC subjected to shear stress, keratin IF are disassembled, as reflected by their increased solubility. In contrast, AEC subjected to stretch showed no changes in the state of assembly of IF. Pretreatment with the protein kinase C (PKC) inhibitor, bisindolymaleimide, prevents the increase in solubility of either K8 or its assembly partner K18 in shear-stressed AEC. Phosphoserine-specific antibodies demonstrate that K8 Ser-73 is phosphorylated in a time-dependent manner in shear-stressed AEC. Furthermore, we showed that shear stress activates PKC delta and that the PKC delta peptide antagonist, delta V1-1, significantly attenuates the shear stress-induced increase in keratin phosphorylation and solubility. These data suggested that shear stress mediates the phosphorylation of serine residues in K8, leading to the disassembly of IF in alveolar epithelial cells. Importantly, these data provided clues regarding a molecular link between mechanically induced signal transduction and alterations in cytoskeletal IF.
Collapse
Affiliation(s)
- Karen M Ridge
- Division of Pulmonary and Critical Care Medicine, Northwestern University Medical School, Chicago, Illinois 60611, USA.
| | | | | | | | | | | | | | | |
Collapse
|
29
|
Sasamoto A, Nagino M, Kobayashi S, Naruse K, Nimura Y, Sokabe M. Mechanotransduction by integrin is essential for IL-6 secretion from endothelial cells in response to uniaxial continuous stretch. Am J Physiol Cell Physiol 2004; 288:C1012-22. [PMID: 15613495 DOI: 10.1152/ajpcell.00314.2004] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
We previously reported that uniaxial continuous stretch in human umbilical vein endothelial cells (HUVECs) induced interleukin-6 (IL-6) secretion via IkappaB kinase (IKK)/nuclear factor-kappaB (NF-kappaB) activation. The aim of the present study was to clarify the upstream signaling mechanism responsible for this phenomenon. Stretch-induced IKK activation and IL-6 secretion were inhibited by application of alpha(5)beta(1) integrin-inhibitory peptide (GRGDNP), phosphatidylinositol 3-kinase inhibitor (LY-294002), phospholipase C-gamma inhibitor (U-73122), or protein kinase C inhibitor (H7). Although depletion of intra- or extracellular Ca(2+) pool using thapsigargin (TG) or EGTA, respectively, showed little effect, a TG-EGTA mixture significantly inhibited stretch-induced IKK activation and IL-6 secretion. An increase in the intracellular Ca(2+) concentration ([Ca(2+)](i)) upon continuous stretch was observed even in the presence of TG, EGTA, or GRGDNP, but not in a solution containing the TG-EGTA mixture, indicating that both integrin activation and [Ca(2+)](i) rise are crucial factors for stretch-induced IKK activation and after IL-6 secretion in HUVECs. Furthermore, while PKC activity was inhibited by the TG-EGTA mixture, GRGDNP, LY-294002, or U-73122, PLC-gamma activity was retarded by GRGDNP or LY-294002. These results indicate that continuous stretch-induced IL-6 secretion in HUVECs depends on outside-in signaling via integrins followed by a PI3-K-PLC-gamma-PKC-IKK-NF-kappaB signaling cascade. Another crucial factor, [Ca(2+)](i) increase, may at least be required to activate PKC needed for NF-kappaB activation.
Collapse
Affiliation(s)
- Akitoshi Sasamoto
- Department of Physiology, Nagoya Univ. Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya 466-8550, Japan.
| | | | | | | | | | | |
Collapse
|
30
|
Sérée E, Villard PH, Pascussi JM, Pineau T, Maurel P, Nguyen QB, Fallone F, Martin PM, Champion S, Lacarelle B, Savouret JF, Barra Y. Evidence for a new human CYP1A1 regulation pathway involving PPAR-alpha and 2 PPRE sites. Gastroenterology 2004; 127:1436-45. [PMID: 15521013 DOI: 10.1053/j.gastro.2004.08.023] [Citation(s) in RCA: 75] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/02/2022]
Abstract
BACKGROUND AND AIMS Cytochrome P450 1A1 catalyzes the degradation of endobiotics (estradiol, fatty acids, and so on) and the bioactivation of numerous environmental procarcinogens, such as arylamines and polycyclic aromatic hydrocarbons, that are found in food. Several peroxisome proliferators and arachidonic acid derivatives enhance cytochrome P450 1A1 activity, but the mechanisms involved remain unknown. The aim of this work was to study the role of peroxisome proliferator-activated receptors in cytochrome P450 1A1 gene induction. METHODS The role of peroxisome proliferator-activated receptor transcription factors in cytochrome P450 1A1 induction was assessed by means of enzymatic activities, quantitative real-time polymerase chain reaction, gene reporter assays, mutagenesis, and electrophoretic mobility shift assay. RESULTS We show that peroxisome proliferator-activated receptor-alpha agonists (WY-14643, bezafibrate, clofibrate, and phthalate) induce human cytochrome P450 1A1 gene expression, whereas 2,4-thiazolidinedione, a specific peroxisome proliferator-activated receptor-gamma agonist, represses it. The induction of cytochrome P450 1A1 transcripts by WY-14643 was associated with a marked increase of ethoxyresorufin O -deethylase activity (10-fold at 200 mumol/L). Transfection of peroxisome proliferator-activated receptor-alpha complementary DNA enhanced cytochrome P450 1A1 messenger RNA induction by WY-14643, although WY-14643 failed to activate xenobiotic responsive element sequences. Two peroxisome proliferator response element sites were located at positions -931/-919 and -531/-519 of the cytochrome P450 1A1 promoter. Their inactivation by directed mutagenesis suppressed the inductive effect of WY-14643 on cytochrome P450 1A1 promoter activation. Electrophoretic mobility shift assay and chromatin immunoprecipitation assay experiments showed that the 2 cytochrome P450 1A1 peroxisome proliferator response element sites bind the peroxisome proliferator-activated receptor-alpha/retinoid X receptor-alpha heterodimer. CONCLUSIONS We describe here a new cytochrome P450 1A1 induction pathway involving peroxisome proliferator-activated receptor-alpha and 2 peroxisome proliferator response element sites, indicating that peroxisome proliferator-activated receptor-alpha ligands, which are common environmental compounds, may be involved in carcinogenesis.
Collapse
Affiliation(s)
- E Sérée
- FRE Centre National de la Recherche Scientifique, Faculté de Pharmacie, Marseille, France.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Miyakawa AA, de Lourdes Junqueira M, Krieger JE. Identification of two novel shear stress responsive elements in rat angiotensin I converting enzyme promoter. Physiol Genomics 2004; 17:107-13. [PMID: 14872008 DOI: 10.1152/physiolgenomics.00169.2003] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Mechanical forces contribute to maintenance of cardiovascular homeostasis via the control of release and production of vasoactive substances. We demonstrated previously that shear stress decreases rat ACE activity and expression. Using a reporter gene approach and mutagenesis, we show now that the classic shear stress responsive element or SSRE (GAGACC) contained within 1,274 bp of this promoter is not functional in response to shear stress (15 dyn/cm2, 18 h) [for the wild-type ACE promoter (WLuc), static control (C) = 107 ± 6.5%, shear stress (SS) = 65.9 ± 9.4%, n = 8; for the promoter with the classic SSRE mutated (WSS-mut), C = 100 ± 8.2%, SS = 60.2 ± 5.2%, n = 10, respectively]. Analysis of progressive deletion mutants unraveled a 57-bp fragment, position −251 to −195, from the transcription start site, containing functional SSRE (for WLuc, C = 107 ± 6.5%, SS = 65.9 ± 9.4%, n = 8; for 378, C = 100 ± 6.4%, SS = 60.4 ± 4.3%, n = 11; for 251, C = 99.7 ± 2.6%, SS = 63.2 ± 5.5%, n = 7; for 194, C = 104.6 ± 8.1%, SS = 92.4 ± 6.9%, n = 9). This fragment responded to shear stress even in the context of a heterologous promoter. Finally, functional analysis of mutated candidate regulatory elements identified by gel shift, DNase I footprint, and conservation of aligned sequences revealed that only the double mutant (Barbie/GAGA-mut) but not isolated disruption of the Barbie (WBarbie-mut) or the GAGA (WGAGA-mut) prevented the shear-stress-induced response (for Barbie/GAGA-mut, C = 97.9 ± 5%, SS = 99.4 ± 7.2%, n = 6; for WBarbie-mut, C = 106.1 ± 8.6%, SS = 65.9 ± 9.4%, n = 6; for WGAGA-mut, C = 100.1 ± 2.9%, SS = 66.7 ± 1.6, n = 6;). Taken together, these data provide direct evidence for the new role of Barbie and GAGA boxes in mediating the shear-stress-induced downregulation of rat ACE expression and demonstrate that the classic SSRE (GAGACC) is not functional under the experimental conditions tested.
Collapse
Affiliation(s)
- Ayumi Aurea Miyakawa
- Laboratory of Genetic and Molecular Cardiology, Heart Institute (InCor) and Internal Medicine Department/LIM13, University of São Paulo Medical School, 05403-901 São Paulo SP, Brazil
| | | | | |
Collapse
|
32
|
Park H, Shin J, Lee JW, Jo H. Fibronectin‐dependent cell adhesion is required for shear‐dependent ERK activation. ACTA ACUST UNITED AC 2004. [DOI: 10.1080/12265071.2004.9647730] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
|
33
|
Gear ARL, Camerini D. Platelet chemokines and chemokine receptors: linking hemostasis, inflammation, and host defense. Microcirculation 2003; 10:335-50. [PMID: 12851650 DOI: 10.1038/sj.mn.7800198] [Citation(s) in RCA: 124] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2002] [Accepted: 02/24/2003] [Indexed: 12/28/2022]
Abstract
Blood platelets play critical roles in hemostasis, providing rapid essential protection against bleeding and catalyzing the important slower formation of stable blood clots via the coagulation cascade. They are also involved in protection from infection by phagocytosis of pathogens and by secreting chemokines that attract leukocytes. Platelet function usually is activated by primary agonists such as adenosine diphosphate (ADP), thrombin, and collagen, whereas secondary agonists like adrenalin do not induce aggregation on their own but become highly effective in the presence of low levels of primary agonists. Current research has revealed that chemokines represent an important additional class of agonists capable of causing significant activation of platelet function. Early work on platelet alpha-granule proteins suggested that platelet factor 4, now known as CXCL4, modulated aggregation and secretion induced by low agonist levels. Subsequent reports revealed the presence in platelets of messenger RNA for several additional chemokines and chemokine receptors. Three chemokines in particular, CXCL12 (SDF-1), CCL17 (TARC), and CCL22 (MDC), recently have been shown to be strong and rapid activators of platelet aggregation and adhesion after their binding to platelet CXCR4 or CCR4, when acting in combination with low levels of primary agonists. CXCL12 can be secreted by endothelial cells and is present in atherosclerotic plaques, whereas CCL17 and CCL22 are secreted by monocytes and macrophages. Platelet activation leads to the release of alpha-granule chemokines, including CCL3 (MIP-1alpha), CCL5 (RANTES), CCL7 (MCP-3), CCL17, CXCL1 (growth-regulated oncogene-alpha), CXCL5 (ENA-78), and CXCL8 (IL-8), which attract leukocytes and further activate other platelets. These findings help to provide a direct linkage between hemostasis, infection, and inflammation and the development of atherosclerosis.
Collapse
Affiliation(s)
- Adrian R L Gear
- Department of Biochemistry and Molecular Genetics, University of Virginia Health Sciences Center, Charlottesville, VA 22908, USA.
| | | |
Collapse
|
34
|
Clark CB, McKnight NL, Frangos JA. Strain and strain rate activation of G proteins in human endothelial cells. Biochem Biophys Res Commun 2002; 299:258-62. [PMID: 12437979 DOI: 10.1016/s0006-291x(02)02628-1] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
The endothelium is known to sense and respond to its physical environment, but the underlying mechanisms and early events of endothelial cell mechanotransduction are not well understood. The present study measured G protein activation by mechanical strain in human umbilical vein endothelial cells (HUVEC) directly by photoincorporation of a hydrolysis resistant, radiolabeled GTP analog. Ten percent uniaxial strain at a strain rate of 20% s(-1) over 1min activated a 38kDa Galpha subunit 167+/-17% relative to controls, while 2% cyclic strain failed to significantly activate the protein (117+/-19%). A single cycle of 10% strain at 20% s(-1) strain rate activated the Galpha subunit 152+/-25%, while activation at the same strain but lower strain rate (0.3% s(-1)) was not significantly different from controls (116+/-12%). Western blot analysis identified the 38kDa protein as Galpha(q/11). These results demonstrate the rapid activation of G proteins in HUVEC by cyclic uniaxial strain in a strain- and strain rate-dependent manner.
Collapse
Affiliation(s)
- Craig B Clark
- Department of Bioengineering, University of California San Diego, 92093-0142, La Jolla, CA, USA
| | | | | |
Collapse
|
35
|
Mazzucato M, Pradella P, Cozzi MR, De Marco L, Ruggeri ZM. Sequential cytoplasmic calcium signals in a 2-stage platelet activation process induced by the glycoprotein Ibalpha mechanoreceptor. Blood 2002; 100:2793-800. [PMID: 12351387 DOI: 10.1182/blood-2002-02-0514] [Citation(s) in RCA: 136] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
We found that the interaction of platelets with immobilized von Willebrand factor (VWF) under flow induces distinct elevations of cytosolic Ca(++) concentration ([Ca(++)](i)) that are associated with sequential stages of integrin alpha(IIb)beta(3) activation. Fluid-dynamic conditions that are compatible with the existence of tensile stress on the bonds between glycoprotein Ibalpha (GPIbalpha) and the VWF A1 domain led to Ca(++) release from intracellular stores (type alpha/beta peaks), which preceded stationary platelet adhesion. Raised levels of cyclic adenosine monophosphate (cAMP) and cyclic guanosine monophosphate, as well as membrane-permeable calcium chelators, inhibited these [Ca(++)](i) oscillations and prevented stable adhesion without affecting the dynamic characteristics of the typical platelet translocation on VWF mediated by GPIbalpha. Once adhesion was established through the integrin alpha(IIb)beta(3), new [Ca(++)](i) oscillations (type gamma) of greater amplitude and duration, and involving a transmembrane ion flux, developed in association with the recruitment of additional platelets into aggregates. Degradation of released adenosine diphosphate (ADP) to AMP or inhibition of phosphatidylinositol 3-kinase (PI3-K) prevented this response without affecting stationary adhesion and blocked aggregation. These findings indicate that an initial signal induced by stressed GPIbalpha-VWF bonds leads to alpha(IIb)beta(3) activation sufficient to support localized platelet adhesion. Then, additional signals from ADP receptors and possibly ligand-occupied alpha(IIb)beta(3), with the contribution of a pathway involving PI3-K, amplify platelet activation to the level required for aggregation. Our conclusions modify those proposed by others regarding the mechanisms that regulate signaling between GPIbalpha and alpha(IIb)beta(3) and lead to platelet adhesion and aggregation on immobilized VWF.
Collapse
Affiliation(s)
- Mario Mazzucato
- Servizio Immunotrasfusionale e Analisi Cliniche, Centro di Riferimento Oncologico, Aviano, Italy
| | | | | | | | | |
Collapse
|
36
|
Ho PC, Melbin J, Nesto RW. Scholarly review of geometry and compliance: biomechanical perspectives on vascular injury and healing. ASAIO J 2002; 48:337-45. [PMID: 12141460 DOI: 10.1097/00002480-200207000-00002] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
Mechanical stress and strain upon cardiovascular tissue are important factors that influence the ultimate configuration of clinically observed disease entities. Although mechanical forces can stimulate cellular changes and response, structural or geometric alterations introduced by disease processes can, in turn, influence local hemodynamic conditions. Dynamic interactions of structural parameters, such as arterial compliance and geometry, can further contribute to the final determination of the mechanical conditions and outcome of the vessel. Manipulation of vascular compliance and geometry may, therefore, have desirable effects. In this article, fundamental vascular biomechanical forces are defined and their association with cellular response and clinical disease processes are introduced. The interplay between vascular geometry and compliance is emphasized, and the potential for mechanical solutions to vascular diseases are explored.
Collapse
Affiliation(s)
- Paul C Ho
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | | | | |
Collapse
|
37
|
Abstract
OBJECTIVE The endothelium is normally subjected to mechanical deformation resulting from shear stress and from strain associated with stretch of the vessel wall. These stimuli are detected by a mechanosensor that initiates a variety of signaling systems responsible for triggering the functional responses. The identity of the mechanosensor has not been established. This article discusses the different mechanisms of mechanosensing that have been proposed and reviews the literature with respect to signaling systems that are activated in response to stress and strain in endothelium. DATA SOURCES Published literature related to mechanotransduction, signal transduction pathways initiated by strain in endothelium, and pathophysiologic effects of abnormal shear forces in diseases. DATA EXTRACTION AND SYNTHESIS Proposed mechanisms of mechanosensing include stretch-sensitive ion channels, protein kinases associated with the cytoskeleton, integrin-cytoskeletal interactions, cytoskeletal-nuclear interactions, and oxidase systems capable of generating reactive oxygen species. However, the molecular identity of the mechanosensor is not known, nor is it clear whether multiple sensing mechanisms exist. CONCLUSIONS Many responses are initiated in cells subjected to mechanical deformation, including alterations in ion channel conductance, activation of signal transduction pathways, and altered expression of specific genes. Future progress in this field will require a critical distinction between cell systems that become activated during mechanical strain and the identity of the cellular mechanosensor that triggers subsequent responses.
Collapse
Affiliation(s)
- Mir H Ali
- University of Chicago, Pulmonary and Critical Care Medicine, 5841 South Maryland Avenue, Chicago, IL, USA
| | | |
Collapse
|
38
|
Carvalho R, Bumann A, Schaffer J, Gerstenfeld L. Predominant integrin ligands expressed by osteoblasts show preferential regulation in response to both cell adhesion and mechanical perturbation. J Cell Biochem 2002. [DOI: 10.1002/jcb.10031] [Citation(s) in RCA: 38] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
|
39
|
Kim M, Javed NH, Yu JG, Christofi F, Cooke HJ. Mechanical stimulation activates Galphaq signaling pathways and 5-hydroxytryptamine release from human carcinoid BON cells. J Clin Invest 2001; 108:1051-9. [PMID: 11581306 PMCID: PMC200950 DOI: 10.1172/jci12467] [Citation(s) in RCA: 67] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
5-Hydroxytryptamine (5-HT) released from enterochromaffin cells activates secretory and peristaltic reflexes necessary for lubrication and propulsion of intestinal luminal contents. The aim of this study was to identify mechanosensitive intracellular signaling pathways that regulate 5-HT release. Human carcinoid BON cells displayed 5-HT immunoreactivity associated with granules dispersed throughout the cells or at the borders. Mechanical stimulation by rotational shaking released 5-HT from BON cells or from guinea pig jejunum during neural blockade with tetrodotoxin. In streptolysin O-permeabilized cells, guanosine 5'-O- (2-thiodiphosphate) (GDP-beta-S) and a synthetic peptide derived from the COOH terminus of Galphaq abolished mechanically evoked 5-HT release, while the NH(2)-terminal peptide did not. An antisense phosphorothioated oligonucleotide targeted to a unique sequence of Galphaq abolished mechanically evoked 5-HT release and reduced Galphaq protein levels without affecting the expression of Galpha(11). Depletion and chelation of extracellular calcium did not alter mechanically evoked 5-HT release, whereas depletion of intracellular calcium stores by thapsigargin and chelation of intracellular calcium by 1,2-bis (o-Aminophenoxy) ethane-N,N,N',N'-tetraacetic acid tetra (acetoxymethyl) ester (BAPTA-AM) reduced 5-HT release. Mechanically evoked 5-HT release was inhibited by somatostatin-14 in a concentration-dependent manner. The results suggest that mechanical stimulation of enterochromaffin-derived BON cells directly or indirectly stimulates a G protein-coupled receptor that activates Galphaq, mobilizes intracellular calcium, and causes 5-HT release.
Collapse
Affiliation(s)
- M Kim
- Department of Pharmacology, The Ohio State University, College of Medicine, Columbus, Ohio 43210, USA
| | | | | | | | | |
Collapse
|
40
|
Qiu W, Kass DA, Hu Q, Ziegelstein RC. Determinants of shear stress-stimulated endothelial nitric oxide production assessed in real-time by 4,5-diaminofluorescein fluorescence. Biochem Biophys Res Commun 2001; 286:328-35. [PMID: 11500041 DOI: 10.1006/bbrc.2001.5401] [Citation(s) in RCA: 42] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The extremely short biological half-life of endothelial-derived nitric oxide (NO) has impeded real-time measurements of NO synthesis. We used the membrane-permeable fluorescent probe 4,5-diaminofluorescein diacetate (DAF-2 DA) to study determinants of NO synthesis in bovine aortic endothelial cells (BAECs). A step increase in shear stress (SS) from 0.3 to 3.4 dyne/cm(2) triggered an increase in DAF-2 fluorescence starting 3.0 +/- 0.5 min after the flow rise and peaking at 44.7 +/- 7.2 min. This was abolished by intracellular Ca(2+) chelation, but was unaffected by blocking extracellular Ca(2+) influx or by inhibiting SS-related changes in intracellular pH. The increase in DAF-2 fluorescence occurred significantly earlier in BAECs transfected with either superoxide dismutase (SOD) or catalase (CAT), indicating concomitant reactive oxygen species (ROS) generation by SS and "competition" between ROS- and DAF-2-NO interactions. These data provide novel insights into several NO signaling determinants and reveal that DAF-2 can assess real-time SS-stimulated NO synthesis in endothelial cells. This should facilitate the analysis of NO-signaling pathways.
Collapse
Affiliation(s)
- W Qiu
- Department of Medicine, Johns Hopkins Hospital, Baltimore, Maryland 21224, USA
| | | | | | | |
Collapse
|
41
|
Ungvari Z, Sun D, Huang A, Kaley G, Koller A. Role of endothelial [Ca2+]i in activation of eNOS in pressurized arterioles by agonists and wall shear stress. Am J Physiol Heart Circ Physiol 2001; 281:H606-12. [PMID: 11454563 DOI: 10.1152/ajpheart.2001.281.2.h606] [Citation(s) in RCA: 39] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
In cultured endothelial cells, Ca2+-dependent and -independent activation of nitric oxide (NO) synthesis to agonists and flow/wall shear stress (WSS) has been demonstrated. However, the presence and function of these pathways are less well known in microvessels that can be exposed to a high level of WSS. We hypothesized that the role of changes in endothelial intracellular calcium concentration ([Ca2+]i) is different in agonist- and WSS-induced release of NO. Thus changes in endothelial [Ca2+]i and diameter of intact pressurized (approximately 100 microm at 80 mmHg) gracilis skeletal muscle arterioles of rats were measured by fluorescent videomicroscopy. Acetylcholine (ACh) and increases in WSS (by increasing intraluminal flow) elicited dilations (maximum 91 +/- 2% and 34 +/- 4%) that could be inhibited by N(omega)-nitro-L-arginine methyl ester (L-NAME), a NO synthase blocker. In diameter-clamped arterioles, ACh caused substantial increases in the endothelial calcium fluorescence ratio (ER(Ca), maximum 43 +/- 5%), which was significantly greater than changes in ER(Ca) (maximum approximately 10%) to increases in WSS. The Ca(2+) ionophore A-23187 also substantially increased ER(Ca) (maximum 38 +/- 5%) and elicited significant L-NAME-sensitive arteriolar dilations (maximum 45 +/- 7%). Intraluminal administration of the tyrosine kinase inhibitor genistein had no effect on dilations induced by ACh or the NO donor sodium nitroprusside, whereas it eliminated WSS-induced dilations. Collectively, our data suggest that, in endothelium of skeletal muscle arterioles, NO synthesis is activated by shear stress without a substantial increase in [Ca2+]i, most likely by activation of tyrosine kinase pathways, whereas NO release by ACh and A-23187 is associated with substantial increases in [Ca2+]i.
Collapse
Affiliation(s)
- Z Ungvari
- Department of Pathophysiology, Semmelweis University of Medicine, H-1445 Budapest, Hungary
| | | | | | | | | |
Collapse
|
42
|
Osanai T, Akutsu N, Fujita N, Nakano T, Takahashi K, Guan W, Okumura K. Cross talk between prostacyclin and nitric oxide under shear in smooth muscle cell: role in monocyte adhesion. Am J Physiol Heart Circ Physiol 2001; 281:H177-82. [PMID: 11406483 DOI: 10.1152/ajpheart.2001.281.1.h177] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
We tested the hypothesis that at sites of vascular damage, vessel homeostasis is maintained through the cross talk of shear-induced production of prostacyclin and nitric oxide (NO) in vascular smooth muscle cells (VSMC). Confluent A7r5 cells derived from rat aortic VSMC and mesenteric VSMC were exposed to shear stress at 15 dyn/cm(2) for 90 min with the use of a cone-plate device, and productions of prostacyclin and NO were examined. Shear stress increased cumulative production of prostacyclin by 3- to 3.5-fold and that of NO by 6- to 7.5-fold. Western blot analysis showed that inducible NO synthase protein was expressed after shear stress in both types of VSMC. Inhibition of NO synthase enhanced the shear-induced production of prostacyclin from 40 to 60%. Shear-induced production of NO was suppressed by 70% after treatment with 10(-4) M of indomethacin. A7r5 cells adhesiveness for monocytes was suppressed by 50% after shear stress. This suppression was abolished by pretreatment with 10(-4) M of indomethacin, whereas inhibition of NO synthase only minimally inhibited it. We conclude that there is a cross talk of shear-induced production of prostacyclin and NO in VSMC. At sites of vascular damage, prostacyclin synthesis may prevent monocyte adhesiveness for VSMC through the concomitant enhancement of NO production.
Collapse
Affiliation(s)
- T Osanai
- Second Department of Internal Medicine, Hirosaki University School of Medicine, Hirosaki, 036-8562 Japan.
| | | | | | | | | | | | | |
Collapse
|
43
|
Ways DK, Sheetz MJ. The role of protein kinase C in the development of the complications of diabetes. VITAMINS AND HORMONES 2001; 60:149-93. [PMID: 11037624 DOI: 10.1016/s0083-6729(00)60019-5] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Diabetes mellitus produces a state of chronic hyperglycemia which in turn leads to the development of severe complications including retinopathy, nephropathy, neuropathy, and atherosclerosis. Many different mechanisms have been put forward to attempt to explain how glucose elevations can damage these various organ systems. Protein kinase C activation is one of the sequelae of hyperglycemia and is thought to play a role in the development of diabetic complications. There are multiple mechanisms for its activation in the diabetic state and multiple downstream effects attributable to that activation. The role of protein kinase C activation in the development of the above-mentioned complications of diabetes is discussed in this chapter. In addition, the potential use of isoform-specific inhibitors of protein kinase C for the treatment of diabetic complications is proposed.
Collapse
Affiliation(s)
- D K Ways
- Lilly Research Labs, Eli Lilly and Company Indianapolis, Indiana 46285, USA
| | | |
Collapse
|
44
|
Ariyoshi H, Yoshikawa N, Aono Y, Tsuji Y, Ueda A, Tokunaga M, Sakon M, Monden M. Localized activation of m-calpain in migrating human umbilical vein endothelial cells stimulated by shear stress. J Cell Biochem 2001. [DOI: 10.1002/1097-4644(20010401)81:1<184::aid-jcb1034>3.0.co;2-g] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
|
45
|
Blackman BR, Thibault LE, Barbee KA. Selective modulation of endothelial cell [Ca2+]i response to flow by the onset rate of shear stress. J Biomech Eng 2000; 122:274-82. [PMID: 10923296 DOI: 10.1115/1.429660] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
The response of endothelial cells (ECs) to their hemodynamic environment strongly influences normal vascular physiology and the pathogenesis of atherosclerosis. Unique responses to the complex flow patterns in lesion-prone regions imply that the temporal and spatial features of the mechanical stimuli modulate the cellular response to flow. We report the first systematic study of the effects of temporal gradients of shear stress on ECs. Flow was applied to cultured ECs using a novel cone-and-plate device allowing precise and independent control of the shear stress magnitude and the onset rate. Intracellular free calcium concentration ([Ca2+]i) increased rapidly following the onset of flow, and the characteristics of the transient were modulated by both the shear stress magnitude and onset rate. ECs were most sensitive to shear stress applied at physiological onset rates. Furthermore, the relative contribution of extracellular calcium and IP3-mediated release were dependent upon the specific flow regime.
Collapse
Affiliation(s)
- B R Blackman
- Department of Bioengineering, University of Pennsylvania, Philadelphia 19104, USA
| | | | | |
Collapse
|
46
|
Taba Y, Sasaguri T, Miyagi M, Abumiya T, Miwa Y, Ikeda T, Mitsumata M. Fluid shear stress induces lipocalin-type prostaglandin D(2) synthase expression in vascular endothelial cells. Circ Res 2000; 86:967-73. [PMID: 10807869 DOI: 10.1161/01.res.86.9.967] [Citation(s) in RCA: 101] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Ligands for peroxisome proliferator-activated receptor gamma, such as the thiazolidinedione class of antidiabetic drugs and 15-deoxy-Delta(12,14)-prostaglandin J(2) (15d-PGJ(2)), modulate various processes in atherogenesis. In search of cells that generate prostaglandin D(2) (PGD(2)), the metabolic precursor of 15d-PGJ(2), we identified PGD(2) from culture medium of endothelial cells. To study how PGD(2) production is regulated in endothelial cells, we investigated the role of fluid shear stress in the metabolism of PGD(2). Endothelial cells expressed the mRNA for the lipocalin-type PGD(2) synthase (L-PGDS) both in vitro and in vivo. Loading laminar shear stress using a parallel-plate flow chamber markedly enhanced the gene expression of L-PGDS, with the maximal effect being obtained at 15 to 30 dyne/cm(2). The expression began to increase within 6 hours after loading shear stress and reached the maximal level at 18 to 24 hours. In contrast, shear stress did not alter the expression levels of PGI(2) synthase and thromboxane A(2) synthase. In parallel with the increase in the expression level of L-PGDS, endothelial cells released PGD(2) and 15d-PGJ(2) into culture medium. These results demonstrate that shear stress promotes PGD(2) production by stimulating L-PGDS expression and suggest the possibility that a peroxisome proliferator-activated receptor gamma ligand is produced in vascular wall in response to blood flow.
Collapse
Affiliation(s)
- Y Taba
- Department of Bioscience, National Cardiovascular Center Research Institute, Osaka, Japan
| | | | | | | | | | | | | |
Collapse
|
47
|
Haidekker MA, L'Heureux N, Frangos JA. Fluid shear stress increases membrane fluidity in endothelial cells: a study with DCVJ fluorescence. Am J Physiol Heart Circ Physiol 2000; 278:H1401-6. [PMID: 10749738 DOI: 10.1152/ajpheart.2000.278.4.h1401] [Citation(s) in RCA: 134] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Fluid shear stress (FSS) has been shown to be an ubiquitous stimulator of mammalian cell metabolism. Although many of the intracellular signal transduction pathways have been characterized, the primary mechanoreceptor for FSS remains unknown. One hypothesis is that the cytoplasmic membrane acts as the receptor for FSS, leading to increased membrane fluidity, which in turn leads to the activation of heterotrimetric G proteins (13). 9-(Dicyanovinyl)-julolidine (DCVJ) is a fluorescent probe that integrates into the cell membrane and changes its quantum yield with the viscosity of the environment. In a parallel-plate flow chamber, confluent layers of DCVJ-labeled human endothelial cells were exposed to different levels of FSS. With increased FSS, a reduced fluorescence intensity was observed, indicating an increase of membrane fluidity. Step changes of FSS caused an approximately linear drop of fluorescence within 5 s, showing fast and almost full recovery after shear cessation. A linear dose-response relationship between shear stress and membrane fluidity changes was observed. The average fluidity increase over the entire cell monolayer was 22% at 26 dyn/cm(2). This study provides evidence for a link between FSS and membrane fluidity, and suggests that the membrane is an important flow mechanosensor of the cell.
Collapse
Affiliation(s)
- M A Haidekker
- Department of Bioengineering, University of California, San Diego, La Jolla, California 92093-0412, USA
| | | | | |
Collapse
|
48
|
Ranjan V, Waterbury R, Xiao Z, Diamond SL. Fluid shear stress induction of the transcriptional activator c-fos in human and bovine endothelial cells, HeLa, and Chinese hamster ovary cells. Biotechnol Bioeng 2000; 49:383-90. [DOI: 10.1002/(sici)1097-0290(19960220)49:4<383::aid-bit4>3.0.co;2-l] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
|
49
|
Hochleitner BW, Hochleitner EO, Obrist P, Eberl T, Amberger A, Xu Q, Margreiter R, Wick G. Fluid shear stress induces heat shock protein 60 expression in endothelial cells in vitro and in vivo. Arterioscler Thromb Vasc Biol 2000; 20:617-23. [PMID: 10712382 DOI: 10.1161/01.atv.20.3.617] [Citation(s) in RCA: 78] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Recent investigations indicate that the initial event in the pathogenesis of atherosclerosis involves an (auto)immunologic injury to the vessel wall. Heat shock proteins (hsps), which are expressed on the endothelial cell surface, constitute possible autoantigens. After being exposed to shear stress of 30 dyne/cm(2) in vitro by means of a rotational viscometer, human umbilical vein endothelial cells were immunohistochemically stained for hsp 60 by the monoclonal antibody ML-30; static control cells were negative. Maximal hsp 60 induction was observed after 12 hours of hemodynamic stress. In Northern blots, the level of hsp 60 mRNA was markedly increased after only 1 hour of shear stress in human umbilical vein endothelial cells compared with static control cells. In vivo investigations in Lewis rats confirmed these in vitro findings: the intima and media of frozen sections of the right common carotid artery exposed to increased wall shear stress (after ligation of the left common carotid artery) were stained for hsp 60. The vessel wall of the left low-shear-stress-exposed side was negative. These findings demonstrate that shear stress results in hsp 60 induction in endothelial cells in vivo and in vitro, providing the prerequisite for humoral and cellular reactions to endothelial hsp in the earliest stages of atherosclerosis.
Collapse
MESH Headings
- Animals
- Arteriosclerosis/genetics
- Arteriosclerosis/immunology
- Autoantigens/genetics
- Blood Pressure
- Blotting, Northern
- Carotid Artery, Common/cytology
- Carotid Artery, Common/physiopathology
- Cells, Cultured
- Chaperonin 60/analysis
- Chaperonin 60/genetics
- Chaperonin 60/immunology
- Culture Media/pharmacology
- Endothelium, Vascular/chemistry
- Endothelium, Vascular/cytology
- Endothelium, Vascular/immunology
- Female
- Gene Expression/immunology
- Humans
- Ligation
- Perfusion
- RNA, Messenger/analysis
- Rats
- Rats, Inbred Lew
- Staining and Labeling
- Stress, Mechanical
- Umbilical Veins/cytology
- Viscosity
Collapse
Affiliation(s)
- B W Hochleitner
- Institute of Biomedical Aging Research, Austrian Academy of Sciences, Innsbruck, Austria.
| | | | | | | | | | | | | | | |
Collapse
|
50
|
|