1
|
Nayak AK, Das SL, Misbah C. Endothelial calcium dynamics elicited by ATP release from red blood cells. Sci Rep 2024; 14:13550. [PMID: 38866785 DOI: 10.1038/s41598-024-63306-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2023] [Accepted: 05/27/2024] [Indexed: 06/14/2024] Open
Abstract
Red blood cells (RBCs) exhibit an interesting response to hydrodynamic flow, releasing adenosine triphosphate (ATP). Subsequently, these liberated ATP molecules initiate a crucial interaction with endothelial cells (ECs), thereby setting off a cascade involving the release of calcium ions (Ca2 + ). Ca2 + exerts control over a plethora of cellular functions, and acts as a mediator for dilation and contraction of blood vessel walls. This study focuses on the relationship between RBC dynamics and Ca2 + dynamics, based on numerical simulations under Poiseuille flow within a linear two-dimensional channel. It is found that the concentration of ATP depends upon a variety of factors, including RBC density, channel width, and the vigor of the flow. The results of our investigation reveals several features. Firstly, the peak amplitude of Ca2 + per EC escalates in direct proportion to the augmentation of RBC concentration. Secondly, increasing the flow strength induces a reduction in the time taken to reach the peak of Ca2 + concentration, under the condition of a constant channel width. Additionally, when flow strength remains constant, an increase in channel width corresponds to an elevation in calcium peak amplitude, coupled with a decrease in peak time. This implies that Ca2 + signals should transition from relatively unconstrained channels to more confined pathways within real vascular networks. This notion gains support from our examination of calcium propagation in a linear channel. In this scenario, the localized Ca2 + release initiates a propagating wave that gradually encompasses the entire channel. Notably, our computed propagation speed agrees with observations.
Collapse
Affiliation(s)
| | - Sovan Lal Das
- Physical and Chemical Biology Laboratory, and Department of Mechanical Engineering, Indian Institute of Technology Palakkad, Palakkad, 678623, India
| | - Chaouqi Misbah
- CNRS, LIPhy, Université Grenoble Alpes, 38000, Grenoble, France.
| |
Collapse
|
2
|
Gorobets O, Gorobets S, Polyakova T, Zablotskii V. Modulation of calcium signaling and metabolic pathways in endothelial cells with magnetic fields. NANOSCALE ADVANCES 2024; 6:1163-1182. [PMID: 38356636 PMCID: PMC10863714 DOI: 10.1039/d3na01065a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Accepted: 01/21/2024] [Indexed: 02/16/2024]
Abstract
Calcium signaling plays a crucial role in various physiological processes, including muscle contraction, cell division, and neurotransmitter release. Dysregulation of calcium levels and signaling has been linked to a range of pathological conditions such as neurodegenerative disorders, cardiovascular disease, and cancer. Here, we propose a theoretical model that predicts the modulation of calcium ion channel activity and calcium signaling in the endothelium through the application of either a time-varying or static gradient magnetic field (MF). This modulation is achieved by exerting magnetic forces or torques on either biogenic or non-biogenic magnetic nanoparticles that are bound to endothelial cell membranes. Since calcium signaling in endothelial cells induces neuromodulation and influences blood flow control, treatment with a magnetic field shows promise for regulating neurovascular coupling and treating vascular dysfunctions associated with aging and neurodegenerative disorders. Furthermore, magnetic treatment can enable control over the decoding of Ca signals, ultimately impacting protein synthesis. The ability to modulate calcium wave frequencies using MFs and the MF-controlled decoding of Ca signaling present promising avenues for treating diseases characterized by calcium dysregulation.
Collapse
Affiliation(s)
- Oksana Gorobets
- National Technical University of Ukraine, "Igor Sikorsky Kyiv Polytechnic Institute" Ukraine
| | - Svitlana Gorobets
- National Technical University of Ukraine, "Igor Sikorsky Kyiv Polytechnic Institute" Ukraine
| | - Tatyana Polyakova
- Institute of Physics of the Czech Academy of Sciences Prague Czech Republic
| | - Vitalii Zablotskii
- Institute of Physics of the Czech Academy of Sciences Prague Czech Republic
- International Magnetobiology Frontier Research Center (iMFRC), Science Island Hefei China
| |
Collapse
|
3
|
Banecki KMRM, Dora KA. Endothelin-1 in Health and Disease. Int J Mol Sci 2023; 24:11295. [PMID: 37511055 PMCID: PMC10379484 DOI: 10.3390/ijms241411295] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Revised: 07/04/2023] [Accepted: 07/05/2023] [Indexed: 07/30/2023] Open
Abstract
Discovered almost 40 years ago, the potent vasoconstrictor peptide endothelin-1 (ET-1) has a wide range of roles both physiologically and pathologically. In recent years, there has been a focus on the contribution of ET-1 to disease. This has led to the development of various ET receptor antagonists, some of which are approved for the treatment of pulmonary arterial hypertension, while clinical trials for other diseases have been numerous yet, for the most part, unsuccessful. However, given the vast physiological impact of ET-1, it is both surprising and disappointing that therapeutics targeting the ET-1 pathway remain limited. Strategies aimed at the pathways influencing the synthesis and release of ET-1 could provide new therapeutic avenues, yet research using cultured cells in vitro has had little follow up in intact ex vivo and in vivo preparations. This article summarises what is currently known about the synthesis, storage and release of ET-1 as well as the role of ET-1 in several diseases including cardiovascular diseases, COVID-19 and chronic pain. Unravelling the ET-1 pathway and identifying therapeutic targets has the potential to treat many diseases whether through disease prevention, slowing disease progression or reversing pathology.
Collapse
Affiliation(s)
| | - Kim A Dora
- Department of Pharmacology, University of Oxford, Mansfield Road, Oxford OX1 3QT, UK
| |
Collapse
|
4
|
Gou Z, Zhang H, Misbah C. Heterogeneous ATP patterns in microvascular networks. J R Soc Interface 2023; 20:20230186. [PMID: 37464803 PMCID: PMC10354495 DOI: 10.1098/rsif.2023.0186] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Accepted: 06/21/2023] [Indexed: 07/20/2023] Open
Abstract
ATP is not only an energy carrier but also serves as an important signalling molecule in many physiological processes. Abnormal ATP level in blood vessel is known to be related to several pathologies, such as inflammation, hypoxia and atherosclerosis. Using advanced numerical methods, we analysed ATP released by red blood cells (RBCs) and its degradation by endothelial cells (ECs) in a cat mesentery-inspired vascular network, accounting for RBC mutual interaction and interactions with vascular walls. Our analysis revealed a heterogeneous ATP distribution in the network, with higher concentrations in the cell-free layer, concentration peaks around bifurcations and heterogeneity among vessels of the same level. These patterns arise from the spatio-temporal organization of RBCs induced by the network geometry. It is further shown that an alteration of hematocrit and flow strength significantly affects ATP level as well as heterogeneity in the network. These findings constitute a first building block to elucidate the intricate nature of ATP patterns in vascular networks and the far reaching consequences for other biochemical signalling, such as calcium, by ECs.
Collapse
Affiliation(s)
- Zhe Gou
- CNRS, LIPhy, Université Grenoble Alpes, 38000 Grenoble, France
| | - Hengdi Zhang
- CNRS, LIPhy, Université Grenoble Alpes, 38000 Grenoble, France
- Shenzhen Sibionics Co. Ltd, Shenzhen, People’s Republic of China
| | - Chaouqi Misbah
- CNRS, LIPhy, Université Grenoble Alpes, 38000 Grenoble, France
| |
Collapse
|
5
|
Hong SG, Ashby JW, Kennelly JP, Wu M, Chattopadhyay E, Foreman R, Tontonoz P, Turowski P, Gallagher-Jones M, Mack JJ. Polarized Mechanosensitive Signaling Domains Protect Arterial Endothelial Cells Against Inflammation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.05.26.542500. [PMID: 37292837 PMCID: PMC10246006 DOI: 10.1101/2023.05.26.542500] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Endothelial cells (ECs) in the descending aorta are exposed to high laminar shear stress, which supports an anti-inflammatory phenotype that protects them from atherosclerosis. High laminar shear stress also supports flow-aligned cell elongation and front-rear polarity, but whether this is required for athero-protective signaling is unclear. Here, we show that Caveolin-1-rich microdomains become polarized at the downstream end of ECs exposed to continuous high laminar flow. These microdomains are characterized by higher membrane rigidity, filamentous actin (F-actin) and lipid accumulation. Transient receptor potential vanilloid-type 4 (Trpv4) ion channels, while ubiquitously expressed, mediate localized Ca 2+ entry at these microdomains where they physically interact with clustered Caveolin-1. The resultant focal bursts in Ca 2+ activate the anti-inflammatory factor endothelial nitric oxide synthase (eNOS) within the confines of these domains. Importantly, we find that signaling at these domains requires both cell body elongation and sustained flow. Finally, Trpv4 signaling at these domains is necessary and sufficient to suppress inflammatory gene expression. Our work reveals a novel polarized mechanosensitive signaling hub that induces an anti-inflammatory response in arterial ECs exposed to high laminar shear stress.
Collapse
|
6
|
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
|
7
|
The spectrin cytoskeleton integrates endothelial mechanoresponses. Nat Cell Biol 2022; 24:1226-1238. [PMID: 35817960 DOI: 10.1038/s41556-022-00953-5] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Accepted: 06/01/2022] [Indexed: 12/13/2022]
Abstract
Physiological blood flow induces the secretion of vasoactive compounds, notably nitric oxide, and promotes endothelial cell elongation and reorientation parallel to the direction of applied shear. How shear is sensed and relayed to intracellular effectors is incompletely understood. Here, we demonstrate that an apical spectrin network is essential to convey the force imposed by shear to endothelial mechanosensors. By anchoring CD44, spectrins modulate the cell surface density of hyaluronan and sense and translate shear into changes in plasma membrane tension. Spectrins also regulate the stability of apical caveolae, where the mechanosensitive PIEZO1 channels are thought to reside. Accordingly, shear-induced PIEZO1 activation and the associated calcium influx were absent in spectrin-deficient cells. As a result, cell realignment and flow-induced endothelial nitric oxide synthase stimulation were similarly dependent on spectrin. We conclude that the apical spectrin network is not only required for shear sensing but also transmits and distributes the resulting tensile forces to mechanosensors that elicit protective and vasoactive responses.
Collapse
|
8
|
Selahi A, Chakraborty S, Muthuchamy M, Zawieja DC, Jain A. Intracellular calcium dynamics of lymphatic endothelial and muscle cells co-cultured in a Lymphangion-Chip under pulsatile flow. Analyst 2022; 147:2953-2965. [DOI: 10.1039/d2an00396a] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
A Lymphangion-Chip consisting an endothelial lumen co-cultured with muscle cells was exposed to step or pulsatile flow. The real-time analyses of intracellular calcium dynamics reveal the coupling of signaling between these cells under complex flows.
Collapse
Affiliation(s)
- Amirali Selahi
- Department of Biomedical Engineering, College of Engineering, Texas A&M University, College Station, TX, USA
| | - Sanjukta Chakraborty
- Department of Medical Physiology, College of Medicine, Texas A&M Health Science Center, Bryan, TX, USA
| | - Mariappan Muthuchamy
- Department of Medical Physiology, College of Medicine, Texas A&M Health Science Center, Bryan, TX, USA
| | - David C. Zawieja
- Department of Medical Physiology, College of Medicine, Texas A&M Health Science Center, Bryan, TX, USA
| | - Abhishek Jain
- Department of Biomedical Engineering, College of Engineering, Texas A&M University, College Station, TX, USA
- Department of Medical Physiology, College of Medicine, Texas A&M Health Science Center, Bryan, TX, USA
- Department of Cardiovascular Sciences, Houston Methodist Academic Institute, Houston, TX, USA
| |
Collapse
|
9
|
Choi SJ, Lillicrap D. A sticky proposition: The endothelial glycocalyx and von Willebrand factor. J Thromb Haemost 2020; 18:781-785. [PMID: 31984614 PMCID: PMC7594466 DOI: 10.1111/jth.14743] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2019] [Revised: 01/10/2020] [Accepted: 01/14/2020] [Indexed: 01/19/2023]
Abstract
Von Willebrand factor (VWF) is a critical component of the hemostatic system. Basal secretion of VWF from endothelial cells is the principal determinant of an individual's baseline plasma VWF levels, while endothelial VWF release can also be induced by several biochemical agonists and biomechanical forces such as increased shear stress. However, the mechanotransduction machinery responsible for this latter response is unclear. Here we propose that the endothelial glycocalyx (EGC), a dynamic layer of proteins and carbohydrates that covers the surface of the vascular endothelium, may play a key role in mediating this response. The EGC has previously been implicated in mediating the mechanotransduction of shear stress in other shear-responsive endothelial processes, such as nitric oxide production and stem cell differentiation. Here, we hypothesize that a similar mechanism may be responsible for the basal secretion of endothelial VWF, whereby the EGC mediates the mechanotransduction of physiological shear stress generated by flowing blood, that in turn contributes to the maintenance of physiological plasma VWF levels.
Collapse
Affiliation(s)
- Seon Jae Choi
- Department of Pathology and Molecular Medicine, Queen's University, Kingston, ON, Canada
| | - David Lillicrap
- Department of Pathology and Molecular Medicine, Queen's University, Kingston, ON, Canada
| |
Collapse
|
10
|
Meza CA, La Favor JD, Kim DH, Hickner RC. Endothelial Dysfunction: Is There a Hyperglycemia-Induced Imbalance of NOX and NOS? Int J Mol Sci 2019; 20:ijms20153775. [PMID: 31382355 PMCID: PMC6696313 DOI: 10.3390/ijms20153775] [Citation(s) in RCA: 184] [Impact Index Per Article: 36.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2019] [Revised: 07/29/2019] [Accepted: 07/31/2019] [Indexed: 02/07/2023] Open
Abstract
NADPH oxidases (NOX) are enzyme complexes that have received much attention as key molecules in the development of vascular dysfunction. NOX have the primary function of generating reactive oxygen species (ROS), and are considered the main source of ROS production in endothelial cells. The endothelium is a thin monolayer that lines the inner surface of blood vessels, acting as a secretory organ to maintain homeostasis of blood flow. The enzymatic production of nitric oxide (NO) by endothelial NO synthase (eNOS) is critical in mediating endothelial function, and oxidative stress can cause dysregulation of eNOS and endothelial dysfunction. Insulin is a stimulus for increases in blood flow and endothelium-dependent vasodilation. However, cardiovascular disease and type 2 diabetes are characterized by poor control of the endothelial cell redox environment, with a shift toward overproduction of ROS by NOX. Studies in models of type 2 diabetes demonstrate that aberrant NOX activation contributes to uncoupling of eNOS and endothelial dysfunction. It is well-established that endothelial dysfunction precedes the onset of cardiovascular disease, therefore NOX are important molecular links between type 2 diabetes and vascular complications. The aim of the current review is to describe the normal, healthy physiological mechanisms involved in endothelial function, and highlight the central role of NOX in mediating endothelial dysfunction when glucose homeostasis is impaired.
Collapse
Affiliation(s)
- Cesar A Meza
- Department of Nutrition, Food & Exercise Sciences, Florida State University, Tallahassee, FL 32306, USA
| | - Justin D La Favor
- Department of Nutrition, Food & Exercise Sciences, Florida State University, Tallahassee, FL 32306, USA
| | - Do-Houn Kim
- Department of Nutrition, Food & Exercise Sciences, Florida State University, Tallahassee, FL 32306, USA
| | - Robert C Hickner
- Department of Nutrition, Food & Exercise Sciences, Florida State University, Tallahassee, FL 32306, USA.
- Institute of Sports Sciences and Medicine, College of Human Sciences, Florida State University, Tallahassee, FL 32306, USA.
- Department of Biokinetics, Exercise and Leisure Sciences, School of Health Sciences, University of KwaZulu-Natal, Westville 4041, South Africa.
| |
Collapse
|
11
|
Surya VN, Michalaki E, Fuller GG, Dunn AR. Lymphatic endothelial cell calcium pulses are sensitive to spatial gradients in wall shear stress. Mol Biol Cell 2019; 30:923-931. [PMID: 30811261 PMCID: PMC6589782 DOI: 10.1091/mbc.e18-10-0618] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Cytosolic calcium (Ca2+) is a ubiquitous second messenger that influences numerous aspects of cellular function. In many cell types, cytosolic Ca2+ concentrations are characterized by periodic pulses, whose dynamics can influence downstream signal transduction. Here, we examine the general question of how cells use Ca2+ pulses to encode input stimuli in the context of the response of lymphatic endothelial cells (LECs) to fluid flow. Previous work shows that fluid flow regulates Ca2+ dynamics in LECs and that Ca2+-dependent signaling plays a key role in regulating lymphatic valve formation during embryonic development. However, how fluid flow might influence the Ca2+ pulse dynamics of individual LECs has remained, to our knowledge, little explored. We used live-cell imaging to characterize Ca2+ pulse dynamics in LECs exposed to fluid flow in an in vitro flow device that generates spatial gradients in wall shear stress (WSS), such as are found at sites of valve formation. We found that the frequency of Ca2+ pulses was sensitive to the magnitude of WSS, while the duration of individual Ca2+ pulses increased in the presence of spatial gradients in WSS. These observations provide an example of how cells can separately modulate Ca2+ pulse frequency and duration to encode distinct forms of information, a phenomenon that could extend to other cell types.
Collapse
Affiliation(s)
- Vinay N Surya
- Department of Chemical Engineering, Stanford University, Stanford, CA 94305
| | | | - Gerald G Fuller
- Department of Chemical Engineering, Stanford University, Stanford, CA 94305
| | - Alexander R Dunn
- Department of Chemical Engineering, Stanford University, Stanford, CA 94305.,Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA 94305
| |
Collapse
|
12
|
Lin Q, Zhao L, Jing R, Trexler C, Wang H, Li Y, Tang H, Huang F, Zhang F, Fang X, Liu J, Jia N, Chen J, Ouyang K. Inositol 1,4,5-Trisphosphate Receptors in Endothelial Cells Play an Essential Role in Vasodilation and Blood Pressure Regulation. J Am Heart Assoc 2019; 8:e011704. [PMID: 30755057 PMCID: PMC6405661 DOI: 10.1161/jaha.118.011704] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/07/2018] [Accepted: 01/17/2019] [Indexed: 01/06/2023]
Abstract
Background Endothelial NO synthase plays a central role in regulating vasodilation and blood pressure. Intracellular Ca2+ mobilization is a critical modulator of endothelial NO synthase function, and increased cytosolic Ca2+ concentration in endothelial cells is able to induce endothelial NO synthase phosphorylation. Ca2+ release mediated by 3 subtypes of inositol 1,4,5-trisphosphate receptors ( IP 3Rs) from the endoplasmic reticulum and subsequent Ca2+ entry after endoplasmic reticulum Ca2+ store depletion has been proposed to be the major pathway to mobilize Ca2+ in endothelial cells. However, the physiological role of IP 3Rs in regulating blood pressure remains largely unclear. Methods and Results To investigate the role of endothelial IP 3Rs in blood pressure regulation, we first generated an inducible endothelial cell-specific IP 3R1 knockout mouse model and found that deletion of IP 3R1 in adult endothelial cells did not affect vasodilation and blood pressure. Considering all 3 subtypes of IP 3Rs are expressed in mouse endothelial cells, we further generated inducible endothelial cell-specific IP 3R triple knockout mice and found that deletion of all 3 IP 3R subtypes decreased plasma NO concentration and increased basal blood pressure. Furthermore, IP 3R deficiency reduced acetylcholine-induced vasodilation and endothelial NO synthase phosphorylation at Ser1177. Conclusions Our results reveal that IP 3R-mediated Ca2+ release in vascular endothelial cells plays an important role in regulating vasodilation and physiological blood pressure.
Collapse
MESH Headings
- Animals
- Aorta, Thoracic/metabolism
- Aorta, Thoracic/pathology
- Aorta, Thoracic/physiopathology
- Blood Pressure/physiology
- Calcium/metabolism
- Disease Models, Animal
- Endothelial Cells/metabolism
- Endothelial Cells/pathology
- Endothelium, Vascular/metabolism
- Endothelium, Vascular/pathology
- Endothelium, Vascular/physiopathology
- Hypertension/metabolism
- Hypertension/pathology
- Hypertension/physiopathology
- Immunoblotting
- Inositol 1,4,5-Trisphosphate Receptors/physiology
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Myography
- Vasodilation/physiology
Collapse
Affiliation(s)
- Qingsong Lin
- Drug Discovery CenterState Key Laboratory of Chemical OncogenomicsSchool of Chemical Biology and BiotechnologyPeking University Shenzhen Graduate SchoolShenzhenChina
| | - Lingyun Zhao
- Drug Discovery CenterState Key Laboratory of Chemical OncogenomicsSchool of Chemical Biology and BiotechnologyPeking University Shenzhen Graduate SchoolShenzhenChina
| | - Ran Jing
- Department of CardiologyThe Second Xiangya HospitalCentral South UniversityChangshaHunanChina
| | - Christa Trexler
- Department of MedicineSchool of MedicineUniversity of California San DiegoLa JollaCA
| | - Hong Wang
- Drug Discovery CenterState Key Laboratory of Chemical OncogenomicsSchool of Chemical Biology and BiotechnologyPeking University Shenzhen Graduate SchoolShenzhenChina
| | - Yali Li
- Drug Discovery CenterState Key Laboratory of Chemical OncogenomicsSchool of Chemical Biology and BiotechnologyPeking University Shenzhen Graduate SchoolShenzhenChina
| | - Huayuan Tang
- Drug Discovery CenterState Key Laboratory of Chemical OncogenomicsSchool of Chemical Biology and BiotechnologyPeking University Shenzhen Graduate SchoolShenzhenChina
| | - Fang Huang
- Drug Discovery CenterState Key Laboratory of Chemical OncogenomicsSchool of Chemical Biology and BiotechnologyPeking University Shenzhen Graduate SchoolShenzhenChina
| | - Fei Zhang
- Drug Discovery CenterState Key Laboratory of Chemical OncogenomicsSchool of Chemical Biology and BiotechnologyPeking University Shenzhen Graduate SchoolShenzhenChina
| | - Xi Fang
- Department of MedicineSchool of MedicineUniversity of California San DiegoLa JollaCA
| | - Jie Liu
- Department of PathophysiologySchool of MedicineShenzhen UniversityShenzhenChina
| | - Nan Jia
- Department of CardiologyThe Eighth Affiliated HospitalSun Yat‐sen UniversityShenzhenChina
| | - Ju Chen
- Department of MedicineSchool of MedicineUniversity of California San DiegoLa JollaCA
| | - Kunfu Ouyang
- Drug Discovery CenterState Key Laboratory of Chemical OncogenomicsSchool of Chemical Biology and BiotechnologyPeking University Shenzhen Graduate SchoolShenzhenChina
| |
Collapse
|
13
|
Barbee KA, Parikh JB, Liu Y, Buerk DG, Jaron D. Effect of spatial heterogeneity and colocalization of eNOS and capacitative calcium entry channels on shear stress-induced NO production by endothelial cells: A modeling approach. Cell Mol Bioeng 2018; 11:143-155. [PMID: 30288177 DOI: 10.1007/s12195-018-0520-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Introduction Colocalization of endothelial nitric oxide synthase (eNOS) and capacitative Ca2+ entry (CCE) channels in microdomains such as cavaeolae in endothelial cells (ECs) has been shown to significantly affect intracellular Ca2+ dynamics and NO production, but the effect has not been well quantified. Methods We developed a two-dimensional continuum model of an EC integrating shear stress-mediated ATP production, intracellular Ca2+ mobilization, and eNOS activation to investigate the effects of spatial colocalization of plasma membrane eNOS and CCE channels on Ca2+ dynamics and NO production in response to flow-induced shear stress. Our model examines the hypothesis that subcellular colocalization of cellular components can be critical for optimal coupling of NO production to blood flow. Results Our simulations predict that heterogeneity of CCE can result in formation of microdomains with significantly higher Ca2+ compared to the average cytosolic Ca2+. Ca2+ buffers with lower or no mobility further enhanced Ca2+ gradients relative to mobile buffers. Colocalization of eNOS to CCE channels significantly increased NO production. Conclusions Our results provide quantitative understanding for the role of spatial heterogeneity and the compartmentalization of signals in regulation of shear stress-induced NO production.
Collapse
Affiliation(s)
- Kenneth A Barbee
- School of Biomedical Engineering, Science and Health Systems, Drexel University, 3140 Market St., Bossone 704, Philadelphia, PA 19104 USA
| | - Jaimit B Parikh
- IBM Thomas J. Watson Research Center, 1101 Kitchawan Rd., Yorktown Heights, NY USA 10598
| | - Yien Liu
- School of Biomedical Engineering, Science and Health Systems, Drexel University, 3140 Market St., Bossone 704, Philadelphia, PA 19104 USA
| | - Donald G Buerk
- School of Biomedical Engineering, Science and Health Systems, Drexel University, 3140 Market St., Bossone 704, Philadelphia, PA 19104 USA
| | - Dov Jaron
- School of Biomedical Engineering, Science and Health Systems, Drexel University, 3140 Market St., Bossone 704, Philadelphia, PA 19104 USA
| |
Collapse
|
14
|
Suresh K, Shimoda LA. Endothelial Cell Reactive Oxygen Species and Ca 2+ Signaling in Pulmonary Hypertension. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 967:299-314. [PMID: 29047094 DOI: 10.1007/978-3-319-63245-2_18] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Pulmonary hypertension (PH) refers to a disorder characterized by elevated pulmonary arterial pressure, leading to right ventricular overload and eventually right ventricular failure, which results in high morbidity and mortality. PH is associated with heterogeneous etiologies and distinct molecular mechanisms, including abnormal migration and proliferation of endothelial and smooth muscle cells. Although the exact details are not fully elucidated, reactive oxygen species (ROS) have been shown to play a key role in promoting abnormal function in pulmonary arterial smooth muscle and endothelial cells in PH. In endothelial cells, ROS can be generated from sources such as NADPH oxidase and mitochondria, which in turn can serve as signaling molecules in a wide variety of processes including posttranslational modification of proteins involved in Ca2+ homeostasis. In this chapter, we discuss the role of ROS in promoting abnormal vasoreactivity and endothelial migration and proliferation in various models of PH. Furthermore, we draw particular attention to the role of ROS-induced increases in intracellular Ca2+ concentration in the pathobiology of PH.
Collapse
Affiliation(s)
- Karthik Suresh
- Division of Pulmonary and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, 21224, USA. .,Johns Hopkins Asthma and Allergy Center, 5501 Hopkins Bayview Circle, Baltimore, MD, 21224, USA.
| | - Larissa A Shimoda
- Division of Pulmonary and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, 21224, USA
| |
Collapse
|
15
|
Electrophysiological experiments in microgravity: lessons learned and future challenges. NPJ Microgravity 2018; 4:7. [PMID: 29619409 PMCID: PMC5876337 DOI: 10.1038/s41526-018-0042-3] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2017] [Revised: 03/07/2018] [Accepted: 03/07/2018] [Indexed: 02/08/2023] Open
Abstract
Advances in electrophysiological experiments have led to the discovery of mechanosensitive ion channels (MSCs) and the identification of the physiological function of specific MSCs. They are believed to play important roles in mechanosensitive pathways by allowing for cells to sense their mechanical environment. However, the physiological function of many MSCs has not been conclusively identified. Therefore, experiments have been developed that expose cells to various mechanical loads, such as shear flow, membrane indentation, osmotic challenges and hydrostatic pressure. In line with these experiments, mechanical unloading, as experienced in microgravity, represents an interesting alternative condition, since exposure to microgravity leads to a series of physiological adaption processes. As outlined in this review, electrophysiological experiments performed in microgravity have shown an influence of gravity on biological functions depending on ion channels at all hierarchical levels, from the cellular level to organs. In this context, calcium signaling represents an interesting cellular pathway, as it involves the direct action of calcium-permeable ion channels, and specific gravitatic cells have linked graviperception to this pathway. Multiple key proteins in the graviperception pathways have been identified. However, measurements on vertebrae cells have revealed controversial results. In conclusion, electrophysiological experiments in microgravity have shown that ion-channel-dependent physiological processes are altered in mechanically unloaded conditions. Future experiments may provide a better understanding of the underlying mechanisms.
Collapse
|
16
|
Alevriadou BR, Shanmughapriya S, Patel A, Stathopulos PB, Madesh M. Mitochondrial Ca 2+ transport in the endothelium: regulation by ions, redox signalling and mechanical forces. J R Soc Interface 2017; 14:rsif.2017.0672. [PMID: 29237825 DOI: 10.1098/rsif.2017.0672] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2017] [Accepted: 11/16/2017] [Indexed: 02/07/2023] Open
Abstract
Calcium (Ca2+) transport by mitochondria is an important component of the cell Ca2+ homeostasis machinery in metazoans. Ca2+ uptake by mitochondria is a major determinant of bioenergetics and cell fate. Mitochondrial Ca2+ uptake occurs via the mitochondrial Ca2+ uniporter (MCU) complex, an inner mitochondrial membrane protein assembly consisting of the MCU Ca2+ channel, as its core component, and the MCU complex regulatory/auxiliary proteins. In this review, we summarize the current knowledge on the molecular nature of the MCU complex and its regulation by intra- and extramitochondrial levels of divalent ions and reactive oxygen species (ROS). Intracellular Ca2+ concentration ([Ca2+]i), mitochondrial Ca2+ concentration ([Ca2+]m) and mitochondrial ROS (mROS) are intricately coupled in regulating MCU activity. Here, we highlight the contribution of MCU activity to vascular endothelial cell (EC) function. Besides the ionic and oxidant regulation, ECs are continuously exposed to haemodynamic forces (either pulsatile or oscillatory fluid mechanical shear stresses, depending on the precise EC location within the arteries). Thus, we also propose an EC mechanotransduction-mediated regulation of MCU activity in the context of vascular physiology and atherosclerotic vascular disease.
Collapse
Affiliation(s)
- B Rita Alevriadou
- Department of Biomedical Engineering, The Ohio State University, Columbus, OH 43210, USA .,Department of Internal Medicine, Division of Cardiovascular Medicine, The Ohio State University, Columbus, OH 43210, USA.,Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH 43210, USA
| | - Santhanam Shanmughapriya
- Department of Medical Genetics and Molecular Biochemistry, Temple University, Philadelphia, PA 19140, USA.,Center for Translational Medicine, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA
| | - Akshar Patel
- Department of Biomedical Engineering, The Ohio State University, Columbus, OH 43210, USA.,Department of Internal Medicine, Division of Cardiovascular Medicine, The Ohio State University, Columbus, OH 43210, USA.,Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH 43210, USA
| | - Peter B Stathopulos
- Department of Physiology and Pharmacology, Western University, London, Ontario, Canada N6A 5C1
| | - Muniswamy Madesh
- Department of Medical Genetics and Molecular Biochemistry, Temple University, Philadelphia, PA 19140, USA .,Center for Translational Medicine, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA
| |
Collapse
|
17
|
Mack JJ, Mosqueiro TS, Archer BJ, Jones WM, Sunshine H, Faas GC, Briot A, Aragón RL, Su T, Romay MC, McDonald AI, Kuo CH, Lizama CO, Lane TF, Zovein AC, Fang Y, Tarling EJ, de Aguiar Vallim TQ, Navab M, Fogelman AM, Bouchard LS, Iruela-Arispe ML. NOTCH1 is a mechanosensor in adult arteries. Nat Commun 2017; 8:1620. [PMID: 29158473 PMCID: PMC5696341 DOI: 10.1038/s41467-017-01741-8] [Citation(s) in RCA: 196] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2016] [Accepted: 10/13/2017] [Indexed: 02/06/2023] Open
Abstract
Endothelial cells transduce mechanical forces from blood flow into intracellular signals required for vascular homeostasis. Here we show that endothelial NOTCH1 is responsive to shear stress, and is necessary for the maintenance of junctional integrity, cell elongation, and suppression of proliferation, phenotypes induced by laminar shear stress. NOTCH1 receptor localizes downstream of flow and canonical NOTCH signaling scales with the magnitude of fluid shear stress. Reduction of NOTCH1 destabilizes cellular junctions and triggers endothelial proliferation. NOTCH1 suppression results in changes in expression of genes involved in the regulation of intracellular calcium and proliferation, and preventing the increase of calcium signaling rescues the cell-cell junctional defects. Furthermore, loss of Notch1 in adult endothelium increases hypercholesterolemia-induced atherosclerosis in the descending aorta. We propose that NOTCH1 is atheroprotective and acts as a mechanosensor in adult arteries, where it integrates responses to laminar shear stress and regulates junctional integrity through modulation of calcium signaling.
Collapse
Affiliation(s)
- Julia J Mack
- Department of Molecular, Cell & Developmental Biology, University of California, Los Angeles, CA, 90095, USA
| | - Thiago S Mosqueiro
- Institute for Quantitative and Computational Biology, University of California, Los Angeles, CA, 90095, USA
| | - Brian J Archer
- Department of Bioengineering, University of California, Los Angeles, CA, 90095, USA
| | - William M Jones
- Department of Molecular, Cell & Developmental Biology, University of California, Los Angeles, CA, 90095, USA
| | - Hannah Sunshine
- Interdepartmental Graduate Program in Molecular, Cellular and Integrative Physiology, University of California, Los Angeles, CA, 90095, USA
| | - Guido C Faas
- Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, CA, 90095, USA
| | - Anais Briot
- Department of Molecular, Cell & Developmental Biology, University of California, Los Angeles, CA, 90095, USA
| | - Raquel L Aragón
- Molecular Biology Interdisciplinary Graduate Program, Molecular Biology Institute, University of California, Los Angeles, CA, 90095, USA
| | - Trent Su
- Department of Biological Chemistry, University of California, Los Angeles, CA, 90095, USA
| | - Milagros C Romay
- Department of Molecular, Cell & Developmental Biology, University of California, Los Angeles, CA, 90095, USA
| | - Austin I McDonald
- Molecular Biology Interdisciplinary Graduate Program, Molecular Biology Institute, University of California, Los Angeles, CA, 90095, USA
| | - Cheng-Hsiang Kuo
- Department of Medicine, University of Chicago, Chicago, IL, 60637, USA
| | - Carlos O Lizama
- Cardiovascular Research Institute, University of California, San Francisco, CA, 94158, USA
| | - Timothy F Lane
- Department of Biological Chemistry, University of California, Los Angeles, CA, 90095, USA
- Department of Ob-Gyn, David Geffen School of Medicine, University of California, Los Angeles, CA, 90095, USA
- Molecular Biology Institute, University of California, Los Angeles, CA, 90095, USA
| | - Ann C Zovein
- Cardiovascular Research Institute, University of California, San Francisco, CA, 94158, USA
| | - Yun Fang
- Department of Medicine, University of Chicago, Chicago, IL, 60637, USA
| | - Elizabeth J Tarling
- Department of Biological Chemistry, University of California, Los Angeles, CA, 90095, USA
- Molecular Biology Institute, University of California, Los Angeles, CA, 90095, USA
- Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, CA, 90095, USA
| | - Thomas Q de Aguiar Vallim
- Department of Biological Chemistry, University of California, Los Angeles, CA, 90095, USA
- Molecular Biology Institute, University of California, Los Angeles, CA, 90095, USA
- Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, CA, 90095, USA
| | - Mohamad Navab
- Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, CA, 90095, USA
| | - Alan M Fogelman
- Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, CA, 90095, USA
| | - Louis S Bouchard
- Department of Bioengineering, University of California, Los Angeles, CA, 90095, USA
- Molecular Biology Institute, University of California, Los Angeles, CA, 90095, USA
- Department of Chemistry and Biochemistry, University of California, Los Angeles, CA, 90095, USA
| | - M Luisa Iruela-Arispe
- Department of Molecular, Cell & Developmental Biology, University of California, Los Angeles, CA, 90095, USA.
- Molecular Biology Institute, University of California, Los Angeles, CA, 90095, USA.
| |
Collapse
|
18
|
Xu S, Li X, LaPenna KB, Yokota SD, Huke S, He P. New insights into shear stress-induced endothelial signalling and barrier function: cell-free fluid versus blood flow. Cardiovasc Res 2017; 113:508-518. [PMID: 28158679 DOI: 10.1093/cvr/cvx021] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/14/2016] [Accepted: 01/27/2017] [Indexed: 02/05/2023] Open
Abstract
Aims Fluid shear stress (SS) is known to regulate endothelial cell (EC) function. Most of the studies, however, focused on the effects of cell-free fluid-generated wall SS on ECs. The objective of this study was to investigate how changes in blood flow altered EC signalling and endothelial function directly through wall SS and indirectly through SS effects on red blood cells (RBCs). Methods and results Experiments were conducted in individually perfused rat venules. We experimentally induced changes in SS that were quantified by measured flow velocity and fluid viscosity. The concomitant changes in EC [Ca2+]i and nitric oxide (NO) were measured with fluorescent markers, and EC barrier function was assessed by fluorescent microsphere accumulation at EC junctions using confocal imaging. EC eNOS activation was evaluated by immunostaining. In response to changes in SS, increases in EC [Ca2+]i and gap formation occurred only in blood or RBC solution perfused vessels, whereas SS-dependent NO production and eNOS-Ser1177 phosphorylation occurred in both plasma and blood perfused vessels. A bioluminescent assay detected SS-dependent ATP release from RBCs. Pharmacological inhibition and genetic modification of pannexin-1 channels on RBCs abolished SS-dependent ATP release and SS-induced increases in EC [Ca2+]i and gap formation. Conclusions SS-induced EC NO production occurs in both cell free fluid and blood perfused vessels, whereas SS-induced increases in EC [Ca2+]i and EC gap formation require the presence of RBCs, attributing to SS-induced pannexin-1 channel dependent release of ATP from RBCs. Thus, changes in blood flow alter vascular EC function through both wall SS and SS exerted on RBCs, and RBC released ATP contributes to SS-induced changes in EC barrier function.
Collapse
Affiliation(s)
- Sulei Xu
- Department of Cellular and Molecular Physiology, College of Medicine, Pennsylvania University, 500 University Drive, Hershey, PA 17033, USA
| | - Xiang Li
- Department of Cellular and Molecular Physiology, College of Medicine, Pennsylvania University, 500 University Drive, Hershey, PA 17033, USA
| | - Kyle Brian LaPenna
- Department of Cellular and Molecular Physiology, College of Medicine, Pennsylvania University, 500 University Drive, Hershey, PA 17033, USA
| | - Stanley David Yokota
- Department of Physiology and Pharmacology, School of Medicine, West Virginia University, One Medical Center Drive, Morgantown, WV 26506, USA
| | - Sabine Huke
- Department of Medicine, University of Alabama at Birmingham, 901 19th street South. Birmingham, AL 35294, USA
| | - Pingnian He
- Department of Cellular and Molecular Physiology, College of Medicine, Pennsylvania University, 500 University Drive, Hershey, PA 17033, USA
| |
Collapse
|
19
|
Shen Y, Cheng Y, Uyeda TQP, Plaza GR. Cell Mechanosensors and the Possibilities of Using Magnetic Nanoparticles to Study Them and to Modify Cell Fate. Ann Biomed Eng 2017; 45:2475-2486. [PMID: 28744841 DOI: 10.1007/s10439-017-1884-7] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2017] [Accepted: 07/07/2017] [Indexed: 12/13/2022]
Abstract
The use of magnetic nanoparticles (MNPs) is a promising technique for future advances in biomedical applications. This idea is supported by the availability of MNPs that can target specific cell components, the variety of shapes of MNPs and the possibility of finely controlling the applied magnetic forces. To examine this opportunity, here we review the current developments in the use of MNPs to mechanically stimulate cells and, specifically, the cell mechanotransduction systems. We analyze the cell components that may act as mechanosensors and their effect on cell fate and we focus on the promising possibilities of controlling stem-cell differentiation, inducing cancer-cell death and treating nervous-system diseases.
Collapse
Affiliation(s)
- Yajing Shen
- The Institute for Translational Nanomedicine, Shanghai East Hospital, The Institute for Biomedical Engineering & Nano Science, Tongji University School of Medicine, Shanghai, 200120, China
| | - Yu Cheng
- The Institute for Translational Nanomedicine, Shanghai East Hospital, The Institute for Biomedical Engineering & Nano Science, Tongji University School of Medicine, Shanghai, 200120, China.
| | - Taro Q P Uyeda
- The Institute for Translational Nanomedicine, Shanghai East Hospital, The Institute for Biomedical Engineering & Nano Science, Tongji University School of Medicine, Shanghai, 200120, China.,Department of Physics, Faculty of Science and Engineering, Waseda University, Tokyo, 169-8555, Japan
| | - Gustavo R Plaza
- The Institute for Translational Nanomedicine, Shanghai East Hospital, The Institute for Biomedical Engineering & Nano Science, Tongji University School of Medicine, Shanghai, 200120, China. .,Center for Biomedical Technology, Universidad Politécnica de Madrid, 28223, Pozuelo de Alarcón, Spain.
| |
Collapse
|
20
|
Wuest SL, Stern P, Casartelli E, Egli M. Fluid Dynamics Appearing during Simulated Microgravity Using Random Positioning Machines. PLoS One 2017; 12:e0170826. [PMID: 28135286 PMCID: PMC5279744 DOI: 10.1371/journal.pone.0170826] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2016] [Accepted: 01/11/2017] [Indexed: 01/13/2023] Open
Abstract
Random Positioning Machines (RPMs) are widely used as tools to simulate microgravity on ground. They consist of two gimbal mounted frames, which constantly rotate biological samples around two perpendicular axes and thus distribute the Earth's gravity vector in all directions over time. In recent years, the RPM is increasingly becoming appreciated as a laboratory instrument also in non-space-related research. For instance, it can be applied for the formation of scaffold-free spheroid cell clusters. The kinematic rotation of the RPM, however, does not only distribute the gravity vector in such a way that it averages to zero, but it also introduces local forces to the cell culture. These forces can be described by rigid body analysis. Although RPMs are commonly used in laboratories, the fluid motion in the cell culture flasks on the RPM and the possible effects of such on cells have not been examined until today; thus, such aspects have been widely neglected. In this study, we used a numerical approach to describe the fluid dynamic characteristic occurring inside a cell culture flask turning on an operating RPM. The simulations showed that the fluid motion within the cell culture flask never reached a steady state or neared a steady state condition. The fluid velocity depends on the rotational velocity of the RPM and is in the order of a few centimeters per second. The highest shear stresses are found along the flask walls; depending of the rotational velocity, they can reach up to a few 100 mPa. The shear stresses in the "bulk volume," however, are always smaller, and their magnitude is in the order of 10 mPa. In conclusion, RPMs are highly appreciated as reliable tools in microgravity research. They have even started to become useful instruments in new research fields of mechanobiology. Depending on the experiment, the fluid dynamic on the RPM cannot be neglected and needs to be taken into consideration. The results presented in this study elucidate the fluid motion and provide insight into the convection and shear stresses that occur inside a cell culture flask during RPM experiments.
Collapse
Affiliation(s)
- Simon L. Wuest
- Lucerne University of Applied Sciences and Arts, School of Engineering and Architecture, CC Aerospace Biomedical Science and Technology, Space Biology Group, Hergiswil, Switzerland
| | - Philip Stern
- Lucerne University of Applied Sciences and Arts, School of Engineering and Architecture, CC Fluid Mechanics and Hydraulic Machines, Horw, Switzerland
| | - Ernesto Casartelli
- Lucerne University of Applied Sciences and Arts, School of Engineering and Architecture, CC Fluid Mechanics and Hydraulic Machines, Horw, Switzerland
| | - Marcel Egli
- Lucerne University of Applied Sciences and Arts, School of Engineering and Architecture, CC Aerospace Biomedical Science and Technology, Space Biology Group, Hergiswil, Switzerland
- * E-mail:
| |
Collapse
|
21
|
Abstract
Microfluidics is an interdisciplinary field intersecting many areas in engineering. Utilizing a combination of physics, chemistry, biology, and biotechnology, along with practical applications for designing devices that use low volumes of fluids to achieve high-throughput screening, is a major goal in microfluidics. Microfluidic approaches allow the study of cells growth and differentiation using a variety of conditions including control of fluid flow that generates shear stress. Recently, Piezo1 channels were shown to respond to fluid shear stress and are crucial for vascular development. This channel is ideal for studying fluid shear stress applied to cells using microfluidic devices. We have developed an approach that allows us to analyze the role of Piezo channels on any given cell and serves as a high-throughput screen for drug discovery. We show that this approach can provide detailed information about the inhibitors of Piezo channels.
Collapse
|
22
|
Wilson C, Lee MD, McCarron JG. Acetylcholine released by endothelial cells facilitates flow-mediated dilatation. J Physiol 2016; 594:7267-7307. [PMID: 27730645 PMCID: PMC5157078 DOI: 10.1113/jp272927] [Citation(s) in RCA: 84] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2016] [Accepted: 10/03/2016] [Indexed: 01/24/2023] Open
Abstract
KEY POINTS The endothelium plays a pivotal role in the vascular response to chemical and mechanical stimuli. The endothelium is exquisitely sensitive to ACh, although the physiological significance of ACh-induced activation of the endothelium is unknown. In the present study, we investigated the mechanisms of flow-mediated endothelial calcium signalling. Our data establish that flow-mediated endothelial calcium responses arise from the autocrine action of non-neuronal ACh released by the endothelium. ABSTRACT Circulating blood generates frictional forces (shear stress) on the walls of blood vessels. These frictional forces critically regulate vascular function. The endothelium senses these frictional forces and, in response, releases various vasodilators that relax smooth muscle cells in a process termed flow-mediated dilatation. Although some elements of the signalling mechanisms have been identified, precisely how flow is sensed and transduced to cause the release of relaxing factors is poorly understood. By imaging signalling in large areas of the endothelium of intact arteries, we show that the endothelium responds to flow by releasing ACh. Once liberated, ACh acts to trigger calcium release from the internal store in endothelial cells, nitric oxide production and artery relaxation. Flow-activated release of ACh from the endothelium is non-vesicular and occurs via organic cation transporters. ACh is generated following mitochondrial production of acetylCoA. Thus, we show ACh is an autocrine signalling molecule released from endothelial cells, and identify a new role for the classical neurotransmitter in endothelial mechanotransduction.
Collapse
Affiliation(s)
- Calum Wilson
- Strathclyde Institute of Pharmacy and Biomedical SciencesUniversity of StrathclydeSIPBS BuildingGlasgowUK
| | - Matthew D. Lee
- Strathclyde Institute of Pharmacy and Biomedical SciencesUniversity of StrathclydeSIPBS BuildingGlasgowUK
| | - John G. McCarron
- Strathclyde Institute of Pharmacy and Biomedical SciencesUniversity of StrathclydeSIPBS BuildingGlasgowUK
| |
Collapse
|
23
|
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
|
24
|
Heemskerk N, Asimuddin M, Oort C, van Rijssel J, van Buul JD. Annexin A2 Limits Neutrophil Transendothelial Migration by Organizing the Spatial Distribution of ICAM-1. THE JOURNAL OF IMMUNOLOGY 2016; 196:2767-78. [DOI: 10.4049/jimmunol.1501322] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/16/2015] [Accepted: 01/05/2016] [Indexed: 01/13/2023]
|
25
|
Kirby PL, Buerk DG, Parikh J, Barbee KA, Jaron D. Mathematical model for shear stress dependent NO and adenine nucleotide production from endothelial cells. Nitric Oxide 2016; 52:1-15. [PMID: 26529478 PMCID: PMC4703509 DOI: 10.1016/j.niox.2015.10.004] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2015] [Revised: 10/21/2015] [Accepted: 10/27/2015] [Indexed: 11/19/2022]
Abstract
We developed a mass transport model for a parallel-plate flow chamber apparatus to predict the concentrations of nitric oxide (NO) and adenine nucleotides (ATP, ADP) produced by cultured endothelial cells (ECs) and investigated how the net rates of production, degradation, and mass transport for these three chemical species vary with changes in wall shear stress (τw). These simulations provide an improved understanding of experimental results obtained with parallel-plate flow chambers and allows quantitative analysis of the relationship between τw, adenine nucleotide concentrations, and NO produced by ECs. Experimental data obtained after altering ATP and ADP concentrations with apyrase were analyzed to quantify changes in the rate of NO production (RNO). The effects of different isoforms of apyrase on ATP and ADP concentrations and nucleotide-dependent changes in RNO could be predicted with the model. A decrease in ATP was predicted with apyrase, but an increase in ADP was simulated due to degradation of ATP. We found that a simple proportional relationship relating a component of RNO to the sum of ATP and ADP provided a close match to the fitted curve for experimentally measured changes in RNO with apyrase. Estimates for the proportionality constant ranged from 0.0067 to 0.0321 μM/s increase in RNO per nM nucleotide concentration, depending on which isoform of apyrase was modeled, with the largest effect of nucleotides on RNO at low τw (<6 dyn/cm(2)).
Collapse
Affiliation(s)
- Patrick L Kirby
- School of Biomedical Engineering, Science and Health Systems, Drexel University, Philadelphia, PA 19104, USA
| | - Donald G Buerk
- School of Biomedical Engineering, Science and Health Systems, Drexel University, Philadelphia, PA 19104, USA
| | - Jaimit Parikh
- School of Biomedical Engineering, Science and Health Systems, Drexel University, Philadelphia, PA 19104, USA
| | - Kenneth A Barbee
- School of Biomedical Engineering, Science and Health Systems, Drexel University, Philadelphia, PA 19104, USA
| | - Dov Jaron
- School of Biomedical Engineering, Science and Health Systems, Drexel University, Philadelphia, PA 19104, USA.
| |
Collapse
|
26
|
Velasco V, Gruenthal M, Zusstone E, Thomas JMD, Berson RE, Keynton RS, Williams SJ. An orbital shear platform for real-time, in vitro endothelium characterization. Biotechnol Bioeng 2016; 113:1336-44. [PMID: 26615057 DOI: 10.1002/bit.25893] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2015] [Revised: 10/29/2015] [Accepted: 11/24/2015] [Indexed: 01/13/2023]
Abstract
Electrical impedance techniques have been used to characterize endothelium morphology, permeability, and motility in vitro. However, these impedance platforms have been limited to either static endothelium studies and/or induced laminar fluid flow at a constant, single shear stress value. In this work, we present a microfabricated impedance sensor for real-time, in vitro characterization of human umbilical vein endothelial cells (HUVECs) undergoing oscillatory hydrodynamic shear. Oscillatory shear was applied with an orbital shaker and the electrical impedance was measured by a microfabricated impedance chip with discrete electrodes positioned at radial locations of 0, 2.5, 5.0, 7.5, 10.0, and 12.5 mm from the center of the chip. Depending on their radial position within the circular orbital platform, HUVECs were exposed to shear values ranging between 0.6 and 6.71 dyne/cm(2) (according to numerical simulations) for 22 h. Impedance spectra were fit to an equivalent circuit model and the trans-endothelial resistance and monolayer's capacitance were extracted. Results demonstrated that, compared to measurements acquired before the onset of shear, cells at the center of the platform that experienced low steady shear stress (∼2.2 dyne/cm(2) ) had an average change in trans-endothelial resistance of 6.99 ± 4.06% and 1.78 ± 2.40% change in cell capacitance after 22 hours of shear exposure; cells near the periphery of the well (r = 12.5 mm) experienced transient shears (2.5-6.7 dyne/cm(2) ) and exhibited a greater change in trans-endothelial resistance (24.2 ± 10.8%) and cell capacitance (4.57 ± 5.39%). This study, demonstrates that the orbital shear platform provides a simple system that can capture and quantify the real-time cellular morphology as a result of induced shear stress. The orbital shear platform presented in this work, compared to traditional laminar platforms, subjects cells to more physiologically relevant oscillatory shear as well as exposes the sample to several shear values simultaneously. Biotechnol. Bioeng. 2016;113: 1336-1344. © 2015 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Vanessa Velasco
- Department of Mechanical Engineering, University of Louisville, Louisville, 40292, Kentucky
| | - Mark Gruenthal
- Department of Mechanical Engineering, University of Louisville, Louisville, 40292, Kentucky
| | - Esther Zusstone
- School of Nursing, University of Louisville, Louisville, Kentucky
| | - Jonathan M D Thomas
- Department of Chemical Engineering, University of Louisville, Louisville, Kentucky
| | - R Eric Berson
- Department of Chemical Engineering, University of Louisville, Louisville, Kentucky
| | - Robert S Keynton
- Department of Bioengineering, University of Louisville, Louisville, Kentucky
| | - Stuart J Williams
- Department of Mechanical Engineering, University of Louisville, Louisville, 40292, Kentucky.
| |
Collapse
|
27
|
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
|
28
|
Kurth F, Franco-Obregón A, Casarosa M, Küster SK, Wuertz-Kozak K, Dittrich PS. Transient receptor potential vanilloid 2-mediated shear-stress responses in C2C12 myoblasts are regulated by serum and extracellular matrix. FASEB J 2015. [PMID: 26207028 DOI: 10.1096/fj.15-275396] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
The developmental sensitivity of skeletal muscle to mechanical forces is unparalleled in other tissues. Calcium entry via reputedly mechanosensitive transient receptor potential (TRP) channel classes has been shown to play an essential role in both the early proliferative stage and subsequent differentiation of skeletal muscle myoblasts, particularly TRP canonical (TRPC) 1 and TRP vanilloid (TRPV) 2. Here we show that C2C12 murine myoblasts respond to fluid flow-induced shear stress with increments in cytosolic calcium that are largely initiated by the mechanosensitive opening of TRPV2 channels. Response to fluid flow was augmented by growth in low extracellular serum concentration (5 vs. 20% fetal bovine serum) by greater than 9-fold and at 18 h in culture, coincident with the greatest TRPV2 channel expression under identical conditions (P < 0.02). Fluid flow responses were also enhanced by substrate functionalization with laminin, rather than with fibronectin, agreeing with previous findings that the gating of TRPV2 is facilitated by laminin. Fluid flow-induced calcium increments were blocked by ruthenium red (27%) and SKF-96365 (38%), whereas they were unaltered by 2-aminoethoxydiphenyl borate, further corroborating that TRPV2 channels play a predominant role in fluid flow mechanosensitivity over that of TRPC1 and TRP melastatin (TRPM) 7.
Collapse
Affiliation(s)
- Felix Kurth
- *Department of Biosystems and Science Engineering and Institute for Biomechanics, Eidgenössische Technische Hochschule Zürich, Switzerland; Department of Surgery, Yong Loo Lin School of Medicine, and Department of Physiology, National University of Singapore, Singapore; and National University Hospital Sports Centre, Singapore
| | - Alfredo Franco-Obregón
- *Department of Biosystems and Science Engineering and Institute for Biomechanics, Eidgenössische Technische Hochschule Zürich, Switzerland; Department of Surgery, Yong Loo Lin School of Medicine, and Department of Physiology, National University of Singapore, Singapore; and National University Hospital Sports Centre, Singapore
| | - Marco Casarosa
- *Department of Biosystems and Science Engineering and Institute for Biomechanics, Eidgenössische Technische Hochschule Zürich, Switzerland; Department of Surgery, Yong Loo Lin School of Medicine, and Department of Physiology, National University of Singapore, Singapore; and National University Hospital Sports Centre, Singapore
| | - Simon K Küster
- *Department of Biosystems and Science Engineering and Institute for Biomechanics, Eidgenössische Technische Hochschule Zürich, Switzerland; Department of Surgery, Yong Loo Lin School of Medicine, and Department of Physiology, National University of Singapore, Singapore; and National University Hospital Sports Centre, Singapore
| | - Karin Wuertz-Kozak
- *Department of Biosystems and Science Engineering and Institute for Biomechanics, Eidgenössische Technische Hochschule Zürich, Switzerland; Department of Surgery, Yong Loo Lin School of Medicine, and Department of Physiology, National University of Singapore, Singapore; and National University Hospital Sports Centre, Singapore
| | - Petra S Dittrich
- *Department of Biosystems and Science Engineering and Institute for Biomechanics, Eidgenössische Technische Hochschule Zürich, Switzerland; Department of Surgery, Yong Loo Lin School of Medicine, and Department of Physiology, National University of Singapore, Singapore; and National University Hospital Sports Centre, Singapore
| |
Collapse
|
29
|
Jafarnejad M, Cromer WE, Kaunas RR, Zhang SL, Zawieja DC, Moore JE. Measurement of shear stress-mediated intracellular calcium dynamics in human dermal lymphatic endothelial cells. Am J Physiol Heart Circ Physiol 2015; 308:H697-706. [PMID: 25617358 PMCID: PMC4385995 DOI: 10.1152/ajpheart.00744.2014] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/16/2014] [Accepted: 01/19/2015] [Indexed: 11/22/2022]
Abstract
The shear stress applied to lymphatic endothelial cells (LEC) by lymph flow changes dramatically under normal conditions as well as in response to disease conditions and immune reactions. In general, LEC are known to regulate the contraction frequency and strength of lymphatic pumping in response to shear stress. Intracellular calcium concentration ([Ca(2+)]i) is an important factor that regulates lymphatic contraction characteristics. In this study, we measured changes in the [Ca(2+)]i under different shear stress levels and determined the source of this calcium signal. Briefly, human dermal LEC were cultured in custom-made microchannels for 3 days before loading with 2 µM fura-2 AM, a ratiometric calcium dye to measure [Ca(2+)]i. Step changes in shear stress resulted in a rapid increase in [Ca(2+)]i followed by a gradual return to the basal level and sometimes below the initial baseline (45.2 ± 2.2 nM). The [Ca(2+)]i reached a peak at 126.2 ± 5.6 nM for 10 dyn/cm(2) stimulus, whereas the peak was only 71.8 ± 5.4 nM for 1 dyn/cm(2) stimulus, indicating that the calcium signal depends on the magnitude of shear stress. Removal of the extracellular calcium from the buffer or pharmocological blockade of calcium release-activated calcium (CRAC) channels significantly reduced the peak [Ca(2+)]i, demonstrating a role of extracellular calcium entry. Inhibition of endoplasmic reticulum (ER) calcium pumps showed the importance of intracellular calcium stores in the initiation of this signal. In conclusion, we demonstrated that the shear-mediated calcium signal is dependent on the magnitude of the shear and involves ER store calcium release and extracellular calcium entry.
Collapse
Affiliation(s)
- M Jafarnejad
- Department of Bioengineering, Imperial College, London, England
| | - W E Cromer
- Department of Medical Physiology, Texas A&M Health Science Center, Temple, Texas; and
| | - R R Kaunas
- Department of Biomedical Engineering, Texas A&M University, College Station, Texas
| | - S L Zhang
- Department of Medical Physiology, Texas A&M Health Science Center, Temple, Texas; and
| | - D C Zawieja
- Department of Medical Physiology, Texas A&M Health Science Center, Temple, Texas; and
| | - J E Moore
- Department of Bioengineering, Imperial College, London, England;
| |
Collapse
|
30
|
Son GY, Yang YM, Park WS, Chang I, Shin DM. Hypotonic stress induces RANKL via transient receptor potential melastatin 3 (TRPM3) and vaniloid 4 (TRPV4) in human PDL cells. J Dent Res 2015; 94:473-81. [PMID: 25595364 DOI: 10.1177/0022034514567196] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Bone remodeling occurs in response to various types of mechanical stress. The periodontal ligament (PDL) plays an important role in mechanical stress-mediated alveolar bone remodeling. However, the underlying mechanism at the cellular level has not been extensively studied. In this study, we investigated the effect of shear stress on the expression of bone remodeling factors, including receptor activator of nuclear factor-kappa B (NF-κB) ligand (RANKL) and osteoprotegerin (OPG), as well as its upstream signaling pathway in primary human PDL cells. We applied hypotonic stress to reproduce shear stress to PDL cells. Hypotonic stress induced the messenger RNA (mRNA) and protein expression of RANKL but not OPG. It also increased intracellular Ca(2+) concentration ([Ca(2+)]i). Extracellular Ca(2+) depletion and nonspecific plasma membrane Ca(2+) channel blockers completely inhibited the increase in both [Ca(2+)]i and RANKL mRNA expression. We identified the expression and activation of transient receptor potential melastatin 3 (TRPM3) and vaniloid 4 (TRPV4) channels in PDL cells. Pregnenolone sulfate (PS) and 4α-phorbol 12, 13-didecanoate (4α-PDD), which are agonists of TRPM3 and TRPV4, augmented Ca(2+) influx and RANKL mRNA expression. Both pharmacological (2-aminoethoxydiphenyl borate [2-APB], ruthenium red [RR], ononetin [Ono], and HC 067047 [HC]) and genetic (small interfering RNA [siRNA]) inhibitors of TRPM3 and TRPV4 reduced the hypotonic stress-mediated increase in [Ca(2+)]i and RANKL mRNA expression. Our study shows that hypotonic stress induced RANKL mRNA expression via TRPM3- and TRPV4-mediated extracellular Ca(2+) influx and RANKL expression. This signaling pathway in PDL cells may play a critical role in mechanical stress-mediated alveolar bone remodeling.
Collapse
Affiliation(s)
- G Y Son
- Department of Oral Biology, Yonsei University College of Dentistry, Seoul, Korea BK21 PLUS Project, Yonsei University College of Dentistry, Seoul, Korea
| | - Y M Yang
- Department of Oral Biology, Yonsei University College of Dentistry, Seoul, Korea
| | - W S Park
- Department of Advanced General Dentistry, Yonsei University College of Dentistry, Seoul, Korea
| | - I Chang
- Department of Oral Biology, Yonsei University College of Dentistry, Seoul, Korea
| | - D M Shin
- Department of Oral Biology, Yonsei University College of Dentistry, Seoul, Korea BK21 PLUS Project, Yonsei University College of Dentistry, Seoul, Korea
| |
Collapse
|
31
|
Modeling of TRPV₄-C₁ -mediated calcium signaling in vascular endothelial cells induced by fluid shear stress and ATP. Biomech Model Mechanobiol 2015; 14:979-93. [PMID: 25577546 DOI: 10.1007/s10237-015-0647-3] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2014] [Accepted: 01/02/2015] [Indexed: 10/24/2022]
Abstract
The calcium signaling plays a vital role in flow-dependent vascular endothelial cell (VEC) physiology. Variations in fluid shear stress and ATP concentration in blood vessels can activate dynamic responses of cytosolic-free [Formula: see text] through various calcium channels on the plasma membrane. In this paper, a novel dynamic model has been proposed for transient receptor potential vanilloid 4 [Formula: see text]-mediated intracellular calcium dynamics in VECs induced by fluid shear stress and ATP. Our model includes [Formula: see text] signaling pathways through P2Y receptors and [Formula: see text] channels (indirect mechanism) and captures the roles of the [Formula: see text] compound channels in VEC [Formula: see text] signaling in response to fluid shear stress (direct mechanism). In particular, it takes into account that the [Formula: see text] compound channels are regulated by intracellular [Formula: see text] and [Formula: see text] concentrations. The simulation studies have demonstrated that the dynamic responses of calcium concentration produced by the proposed model correlate well with the existing experimental observations. We also conclude from the simulation studies that endogenously released ATP may play an insignificant role in the process of intracellular [Formula: see text] response to shear stress.
Collapse
|
32
|
Praetorius HA. The primary cilium as sensor of fluid flow: new building blocks to the model. A review in the theme: cell signaling: proteins, pathways and mechanisms. Am J Physiol Cell Physiol 2014; 308:C198-208. [PMID: 25428884 DOI: 10.1152/ajpcell.00336.2014] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
The primary cilium is an extraordinary organelle. For many years, it had the full attention of only a few dedicated scientists fascinated by its uniqueness. Unexpectedly, after decades of obscurity, it has moved very quickly into the limelight with the increasing evidence of its central role in the many genetic variations that lead to what are now known as ciliopathies. These studies implicated unique biological functions of the primary cilium, which are not completely straightforward. In parallel, and initially completely unrelated to the ciliopathies, the primary cilium was characterized functionally as an organelle that makes cells more susceptible to changes in fluid flow. Thus the primary cilium was suggested to function as a flow-sensing device. This characterization has been substantiated for many epithelial cell types over the years. Nevertheless, part of the central mechanism of signal transduction has not been explained, largely because of the substantial technical challenges of working with this delicate organelle. The current review considers the recent advances that allow us to fill some of the holes in the model of signal transduction in cilium-mediated responses to fluid flow and to pursue the physiological implications of this peculiar organelle.
Collapse
Affiliation(s)
- Helle A Praetorius
- Department of Biomedicine-Physiology, Aarhus University, Aarhus, Denmark
| |
Collapse
|
33
|
Abstract
Endothelial cell dysfunction is the hallmark of every cardiovascular disease/condition, including atherosclerosis and ischemia/reperfusion injury. Fluid shear stress acting on the vascular endothelium is known to regulate cell homeostasis. Altered hemodynamics is thought to play a causative role in endothelial dysfunction. The dysfunction is associated with/preceded by mitochondrial oxidative stress. Studies by our group and others have shown that the form and/or function of the mitochondrial network are affected when endothelial cells are exposed to shear stress in the absence or presence of additional physicochemical stimuli. The present review will summarize the current knowledge on the interconnections among intracellular Ca2+ - nitric oxide - mitochondrial reactive oxygen species, mitochondrial fusion/fission, autophagy/mitophagy, and cell apoptosis vs. survival. More specifically, it will list the evidence on potential regulation of the above intracellular species and processes by the fluid shear stress acting on the endothelium under either physiological flow conditions or during reperfusion (following a period of ischemia). Understanding how the local hemodynamics affects mitochondrial physiology and the cell redox state may lead to development of novel therapeutic strategies for prevention or treatment of the endothelial dysfunction and, hence, of cardiovascular disease.
Collapse
|
34
|
Zhang X, Huk DJ, Wang Q, Lincoln J, Zhao Y. A microfluidic shear device that accommodates parallel high and low stress zones within the same culturing chamber. BIOMICROFLUIDICS 2014; 8:054106. [PMID: 25332743 PMCID: PMC4189595 DOI: 10.1063/1.4894783] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/27/2014] [Accepted: 08/26/2014] [Indexed: 05/02/2023]
Abstract
Fluid shear stress (FSS) plays a critical role in regulating endothelium function and maintaining vascular homeostasis. Current microfluidic devices for studying FSS effects on cells either separate high shear stress zone and low shear stress zone into different culturing chambers, or arranging the zones serially along the flow direction, which complicates subsequent data interpretation. In this paper, we report a diamond shaped microfluidic shear device where the high shear stress zone and the low shear stress zone are arranged in parallel within one culturing chamber. Since the zones with different shear stress magnitudes are aligned normal to the flow direction, the cells in one stress group are not substantially affected by the flow-induced cytokine/chemokine releases by cells in the other group. Cell loading experiments using human umbilical vein endothelial cells show that the device is able to reveal stress magnitude-dependent and loading duration-dependent cell responses. The co-existence of shear stress zones with varied magnitudes within the same culturing chamber not only ensures that all the cells are subject to the identical culturing conditions, but also allows the resemblance of the differential shear stress pattern in natural arterial conditions. The device is expected to provide a new solution for studying the effects of heterogeneous hemodynamic patterns in the onset and progression of various vascular diseases.
Collapse
Affiliation(s)
- X Zhang
- Laboratory for Biomedical Microsystems, Department of Biomedical Engineering, The Ohio State University , Columbus, Ohio 43210, USA
| | - D J Huk
- The Heart Center and Nationwide Children's Hospital Research Institute , Columbus, Ohio 43205, USA
| | - Q Wang
- Laboratory for Biomedical Microsystems, Department of Biomedical Engineering, The Ohio State University , Columbus, Ohio 43210, USA
| | | | - Y Zhao
- Laboratory for Biomedical Microsystems, Department of Biomedical Engineering, The Ohio State University , Columbus, Ohio 43210, USA
| |
Collapse
|
35
|
Andrews AM, Jaron D, Buerk DG, Barbee KA. Shear stress-induced NO production is dependent on ATP autocrine signaling and capacitative calcium entry. Cell Mol Bioeng 2014; 7:510-520. [PMID: 25386222 DOI: 10.1007/s12195-014-0351-x] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022] Open
Abstract
Flow-induced production of nitric oxide (NO) by endothelial cells plays a fundamental role in vascular homeostasis. However, the mechanisms by which shear stress activates NO production remain unclear due in part to limitations in measuring NO, especially under flow conditions. Shear stress elicits the release of ATP, but the relative contribution of autocrine stimulation by ATP to flow-induced NO production has not been established. Furthermore, the importance of calcium in shear stress-induced NO production remains controversial, and in particular the role of capacitive calcium entry (CCE) has yet to be determined. We have utilized our unique NO measurement device to investigate the role of ATP autocrine signaling and CCE in shear stress-induced NO production. We found that endogenously released ATP and downstream activation of purinergic receptors and CCE plays a significant role in shear stress-induced NO production. ATP-induced eNOS phophorylation under static conditions is also dependent on CCE. Inhibition of protein kinase C significantly inhibited eNOS phosphorylation and the calcium response. To our knowledge, we are the first to report on the role of CCE in the mechanism of acute shear stress-induced NO response. In addition, our work highlights the importance of ATP autocrine signaling in shear stress-induced NO production.
Collapse
Affiliation(s)
- Allison M Andrews
- School of Biomedical Engineering, Science, and Health Systems, Drexel University, 3141 Market St. Philadelphia, PA 19104, USA
| | - Dov Jaron
- School of Biomedical Engineering, Science, and Health Systems, Drexel University, 3141 Market St. Philadelphia, PA 19104, USA
| | - Donald G Buerk
- School of Biomedical Engineering, Science, and Health Systems, Drexel University, 3141 Market St. Philadelphia, PA 19104, USA
| | - Kenneth A Barbee
- School of Biomedical Engineering, Science, and Health Systems, Drexel University, 3141 Market St. Philadelphia, PA 19104, USA
| |
Collapse
|
36
|
Madden RMJ, Han SK, Herzog W. The effect of compressive loading magnitude on in situ chondrocyte calcium signaling. Biomech Model Mechanobiol 2014; 14:135-42. [PMID: 24853775 PMCID: PMC4282695 DOI: 10.1007/s10237-014-0594-4] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2013] [Accepted: 05/06/2014] [Indexed: 12/19/2022]
Abstract
Chondrocyte metabolism is stimulated by deformation and is associated with structural changes in the cartilage extracellular matrix (ECM), suggesting that these cells are involved in maintaining tissue health and integrity. Calcium signaling is an initial step in chondrocyte mechanotransduction that has been linked to many cellular processes. Previous studies using isolated chondrocytes proposed loading magnitude as an important factor regulating this response. However, calcium signaling in the intact cartilage differs compared to isolated cells. The purpose of this study was to investigate the effect of loading magnitude on chondrocyte calcium signaling in intact cartilage. We hypothesized that the percentage of cells exhibiting at least one calcium signal increases with increasing load. Fully intact rabbit femoral condyle and patellar bone/cartilage samples were incubated in calcium-sensitive dyes and imaged continuously under compressive loads of 10-40 % strain. Calcium signaling was primarily associated with the dynamic loading phase and greatly increased beyond a threshold deformation of about 10 % nominal tissue strain. There was a trend toward more cells exhibiting calcium signaling as loading magnitude increased (p = 0.133). These results provide novel information toward identifying mechanisms underlying calcium-dependent signaling pathways related to cartilage homeostasis and possibly the onset and progression of osteoarthritis.
Collapse
Affiliation(s)
- Ryan M J Madden
- Human Performance Laboratory, University of Calgary, 2500 University Dr. NW, Calgary, AB, T2N 1N4, Canada,
| | | | | |
Collapse
|
37
|
Devika NT, Jaffar Ali BM. Analysing calcium dependent and independent regulation of eNOS in endothelium triggered by extracellular signalling events. MOLECULAR BIOSYSTEMS 2014; 9:2653-64. [PMID: 23954998 DOI: 10.1039/c3mb70258h] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
The vascular endothelium, the intima of blood vessels, coordinately interacts with several biochemical factors expressing endothelial nitric oxide synthase (eNOS) to produce nitric oxide (NO), a potent endogenous vasodilator. The present study investigated the regulation of eNOS by multiple molecular signal transduction pathways, namely vascular endothelial growth factor (VEGF-A) and shear stress which are implicated in the process of angiogenesis and vascular remodelling respectively. In response to signal transduction upstream by VEGF-A and shear stress, different signalling pathways mediated by kinases and intracellular calcium potentiates eNOS activation leading to nitric oxide release. Our study revealed a distinct pattern of eNOS activation driven by VEGF-A and shear stress, maintaining the signalling specificity of the respective pathways. A transient response to eNOS activation was observed under VEGF-A and shear stress stimulus when mediated by calcium dependent cascades, whereas a sustained response was produced by calcium independent vascular signalling kinases. Furthermore, we found that the basal arterial shear stress enhanced eNOS activity when stimulated synergistically even at low VEGF-A levels which might be utilized to facilitate specific endothelial cell functions. Moreover, our study revealed that the presence of PI3K imparted transient behaviour to PLCγ1 supporting the hypothesis that regression and formation of tube structures are mediated by PLCγ1 and PI3K respectively in endothelial cells. This fact is corroborated by the absence of transient behaviour when PI3K is inhibited. We therefore obtained subtle insights into the control mechanism governing the role of specific signalling proteins which are obligate for the regulation of endothelial cell function and the consequent modulation of the nitric oxide release pattern.
Collapse
Affiliation(s)
- N T Devika
- AU-KBC Research Centre, Madras Institute of Technology, Anna University, Chrompet, Chennai - 600 044, Tamil Nadu, India.
| | | |
Collapse
|
38
|
Tarbell JM, Shi ZD, Dunn J, Jo H. Fluid Mechanics, Arterial Disease, and Gene Expression. ANNUAL REVIEW OF FLUID MECHANICS 2014; 46:591-614. [PMID: 25360054 PMCID: PMC4211638 DOI: 10.1146/annurev-fluid-010313-141309] [Citation(s) in RCA: 93] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/08/2023]
Abstract
This review places modern research developments in vascular mechanobiology in the context of hemodynamic phenomena in the cardiovascular system and the discrete localization of vascular disease. The modern origins of this field are traced, beginning in the 1960s when associations between flow characteristics, particularly blood flow-induced wall shear stress, and the localization of atherosclerotic plaques were uncovered, and continuing to fluid shear stress effects on the vascular lining endothelial) cells (ECs), including their effects on EC morphology, biochemical production, and gene expression. The earliest single-gene studies and genome-wide analyses are considered. The final section moves from the ECs lining the vessel wall to the smooth muscle cells and fibroblasts within the wall that are fluid me chanically activated by interstitial flow that imposes shear stresses on their surfaces comparable with those of flowing blood on EC surfaces. Interstitial flow stimulates biochemical production and gene expression, much like blood flow on ECs.
Collapse
Affiliation(s)
- John M Tarbell
- Department of Biomedical Engineering, The City College of New York, New York, NY 10031
| | - Zhong-Dong Shi
- Developmental Biology Program, Sloan-Kettering Institute, New York, NY 10065
| | - Jessilyn Dunn
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, Georgia 30322
| | - Hanjoong Jo
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, Georgia 30322
| |
Collapse
|
39
|
Abstract
Myocilin is a broadly expressed protein that when mutated uniquely causes glaucoma. While no function has been ascribed to explain focal disease, some properties of myocilin are known. Myocilin is a cytoplasmic protein that also localizes to vesicles specifically as part of a large membrane-associated complex with properties similar to the SNARE machinery that function in vesicle fusion. Its role in vesicle dynamics has not been detailed, however myocilin intersects with the endocytic compartment at the level of the multivesicular body. Since internalized GPCRs are sorted in the multivesicular body, we investigated whether myocilin functions in ligand-dependent GPR143 endocytosis. Using recombinant systems we found that the kinetics of myocilin recruitment to biotinylated membrane proteins was similar to that of arrestin-3. We also co-localized myocilin with GPR143 and Arrestin-2 by confocal microscopy. However, wild-type myocilin differed significantly in its association kinetics and co-localization with internalized proteins from mutant myocilin (P370L or T377M). Moreover, we found that myocilin bound to the cytoplasmic tail of GPR143, an interaction mediated by its amino terminal helix-turn-helix domain. Hydrodynamic analyses show that the myocilin-GPR143 protein complex is >158 kD and stable in 500 mM KCl, but not 0.1% SDS. Collectively, data indicate that myocilin is recruited to the membrane compartment, interacting with GPCR proteins during ligand-mediated endocytosis and that GPCR signaling underlies pathology in myocilin glaucoma.
Collapse
|
40
|
Lee HJ, Li N, Evans SM, Diaz MF, Wenzel PL. Biomechanical force in blood development: extrinsic physical cues drive pro-hematopoietic signaling. Differentiation 2013; 86:92-103. [PMID: 23850217 DOI: 10.1016/j.diff.2013.06.004] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2013] [Revised: 06/17/2013] [Accepted: 06/19/2013] [Indexed: 02/07/2023]
Abstract
The hematopoietic system is dynamic during development and in adulthood, undergoing countless spatial and temporal transitions during the course of one's life. Microenvironmental cues in the many unique hematopoietic niches differ, characterized by distinct soluble molecules, membrane-bound factors, and biophysical features that meet the changing needs of the blood system. Research from the last decade has revealed the importance of substrate elasticity and biomechanical force in determination of stem cell fate. Our understanding of the role of these factors in hematopoiesis is still relatively poor; however, the developmental origin of blood cells from the endothelium provides a model for comparison. Many endothelial mechanical sensors and second messenger systems may also determine hematopoietic stem cell fate, self renewal, and homing behaviors. Further, the intimate contact of hematopoietic cells with mechanosensitive cell types, including osteoblasts, endothelial cells, mesenchymal stem cells, and pericytes, places them in close proximity to paracrine signaling downstream of mechanical signals. The objective of this review is to present an overview of the sensors and intracellular signaling pathways activated by mechanical cues and highlight the role of mechanotransductive pathways in hematopoiesis.
Collapse
Affiliation(s)
- Hyun Jung Lee
- Children's Regenerative Medicine Program, Department of Pediatric Surgery, University of Texas Medical School at Houston, Houston, TX 77030, USA; Center for Stem Cell and Regenerative Medicine, The Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | | | | | | | | |
Collapse
|
41
|
Vascular Endothelium. TISSUE FUNCTIONING AND REMODELING IN THE CIRCULATORY AND VENTILATORY SYSTEMS 2013. [DOI: 10.1007/978-1-4614-5966-8_9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
|
42
|
Abstract
Extracellular adenine nucleotides ATP and ADP on vascular endothelial cells may play a role in the localization of atherogenesis by regulating the release of nitric oxide from endothelial cells and modulating intracellular calcium levels. To quantitatively investigate the concentration distribution of nucleotides on the luminal surface of the human thoracic aorta, we numerically simulated the transport of nucleotides using an aorta model constructed based on MRI images and analyzed the effects of different factors on nucleotide transport, such as ATP release rate (S(ATP)), pulsatile flow and the absence of ATP in the main blood stream. The numerical results revealed that the combined concentration of ATP and ADP (c(w-ATP+ADP)) on the aortic surface varied from place to place, being relatively low in disturbed flow regions. In addition, c(w-ATP+ADP) was significantly affected by S(ATP). For relatively slow S(ATP), such as the moderate sigmoidal release model, c(w-ATP+ADP) was very low in certain flow regions with low wall shear stress. However, for very rapid S(ATP), such as the rapid linear release model, c(w-ATP+ADP) was relatively high in these same regions. The results also demonstrated that for relatively slow S(ATP), pulsatile blood flow enhanced c(w-ATP+ADP). However, for very rapid S(ATP), pulsatile blood flow would reduce c(w-ATP+ADP). Moreover, the absence of ATP within the main blood stream would not influence the distribution of c(w-ATP+ADP). In conclusion, the concentration distribution of nucleotides along the aortic wall was quite uneven and determined by both the ATP release rate and the blood flow pattern in the aorta.
Collapse
Affiliation(s)
- Xiao Liu
- Key Laboratory for Biomechanics and Mechanobiology of the Ministry of Education, School of Biological Science and Medical Engineering, Beihang University, Beijing, China
| | | | | | | |
Collapse
|
43
|
Moccia F, Berra-Romani R, Tanzi F. Update on vascular endothelial Ca 2+ signalling: A tale of ion channels, pumps and transporters. World J Biol Chem 2012; 3:127-58. [PMID: 22905291 PMCID: PMC3421132 DOI: 10.4331/wjbc.v3.i7.127] [Citation(s) in RCA: 86] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/17/2012] [Revised: 07/04/2012] [Accepted: 07/11/2012] [Indexed: 02/05/2023] Open
Abstract
A monolayer of endothelial cells (ECs) lines the lumen of blood vessels and forms a multifunctional transducing organ that mediates a plethora of cardiovascular processes. The activation of ECs from as state of quiescence is, therefore, regarded among the early events leading to the onset and progression of potentially lethal diseases, such as hypertension, myocardial infarction, brain stroke, and tumor. Intracellular Ca2+ signals have long been know to play a central role in the complex network of signaling pathways regulating the endothelial functions. Notably, recent work has outlined how any change in the pattern of expression of endothelial channels, transporters and pumps involved in the modulation of intracellular Ca2+ levels may dramatically affect whole body homeostasis. Vascular ECs may react to both mechanical and chemical stimuli by generating a variety of intracellular Ca2+ signals, ranging from brief, localized Ca2+ pulses to prolonged Ca2+ oscillations engulfing the whole cytoplasm. The well-defined spatiotemporal profile of the subcellular Ca2+ signals elicited in ECs by specific extracellular inputs depends on the interaction between Ca2+ releasing channels, which are located both on the plasma membrane and in a number of intracellular organelles, and Ca2+ removing systems. The present article aims to summarize both the past and recent literature in the field to provide a clear-cut picture of our current knowledge on the molecular nature and the role played by the components of the Ca2+ machinery in vascular ECs under both physiological and pathological conditions.
Collapse
Affiliation(s)
- Francesco Moccia
- Francesco Moccia, Franco Tanzi, Department of Biology and Biotechnologies "Lazzaro Spallanzani", Laboratory of Physiology, University of Pavia, Via Forlanini 6, 27100 Pavia, Italy
| | | | | |
Collapse
|
44
|
Matsui TS, Kaunas R, Kanzaki M, Sato M, Deguchi S. Non-muscle myosin II induces disassembly of actin stress fibres independently of myosin light chain dephosphorylation. Interface Focus 2011; 1:754-66. [PMID: 23050080 DOI: 10.1098/rsfs.2011.0031] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2011] [Accepted: 07/07/2011] [Indexed: 01/11/2023] Open
Abstract
Dynamic remodelling of actin stress fibres (SFs) allows non-muscle cells to adapt to applied forces such as uniaxial cell shortening. However, the mechanism underlying rapid and selective disassembly of SFs oriented in the direction of shortening remains to be elucidated. Here, we investigated how myosin crossbridge cycling induced by MgATP is associated with SF disassembly. Moderate concentrations of MgATP, or [MgATP], induced SF contraction. Meanwhile, at [MgATP] slightly higher than the physiological level, periodic actin patterns emerged along the length of SFs and dispersed within seconds. The actin fragments were diverse in length, but comparable to those in characteristic sarcomeric units of SFs. These results suggest that MgATP-bound non-muscle myosin II dissociates from the individual actin filaments that constitute the sarcomeric units, resulting in unbundling-induced disassembly rather than end-to-end actin depolymerization. This rapid SF disassembly occurred independent of dephosphorylation of myosin light chain. In terms of effects on actin-myosin interactions, a rise in [MgATP] is functionally equivalent to a temporal decrease in the total number of actin-myosin crossbridges. Actin-myosin crossbridges are known to be reduced by an assisting load on myosin. Thus, the present study suggests that reducing the number of actin-myosin crossbridges promotes rapid and orientation-dependent disassembly of SFs after cell shortening.
Collapse
Affiliation(s)
- Tsubasa S Matsui
- Department of Biomedical Engineering , Tohoku University , Sendai 980-8579 , Japan
| | | | | | | | | |
Collapse
|
45
|
Dixit N, Yamayoshi I, Nazarian A, Simon SI. Migrational guidance of neutrophils is mechanotransduced via high-affinity LFA-1 and calcium flux. THE JOURNAL OF IMMUNOLOGY 2011; 187:472-81. [PMID: 21632714 DOI: 10.4049/jimmunol.1004197] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Acute inflammation triggers the innate immune response of neutrophils that efficiently traffic from the bloodstream to concentrate at high numbers at the site of tissue infection or wounding. A gatekeeper in this process is activation of β(2) integrins, which form bond clusters with ICAM-1 on the endothelial surface. These bond clusters serve dual functions of providing adhesive strength to anchor neutrophils under the shear forces of blood flow and directional guidance for cell polarization and subsequent transmigration on inflamed endothelium. We hypothesized that shear forces transmitted through high-affinity LFA-1 facilitates the cooperation with the calcium release-activated channel Orai1 in directing localized cytoskeletal activation and directed migration. By using vascular mimetic microfluidic channels, we observed neutrophil arrest on a substrate of either ICAM-1 or allosteric Abs that stabilize a high- or low-affinity conformation of LFA-1. Neutrophils captured via low-affinity LFA-1 did not exhibit intracellular calcium flux, F-actin polymerization, cell polarization, or directional migration under shear flow. In contrast, high-affinity LFA-1 provided orientation along a uropod-pseudopod axis that required calcium flux through Orai1. We demonstrate how the shear stress of blood flow can transduce distinct outside-in signals at focal sites of high-affinity LFA-1 that provide contact-mediated guidance for neutrophil emigration.
Collapse
Affiliation(s)
- Neha Dixit
- Department of Biomedical Engineering, University of California, Davis, Davis, CA 95616, USA
| | | | | | | |
Collapse
|
46
|
Mazzag B, Barakat AI. The effect of noisy flow on endothelial cell mechanotransduction: a computational study. Ann Biomed Eng 2010; 39:911-21. [PMID: 20963495 PMCID: PMC3033522 DOI: 10.1007/s10439-010-0181-5] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2010] [Accepted: 10/06/2010] [Indexed: 11/24/2022]
Abstract
Flow in the arterial system is mostly laminar, but turbulence occurs in vivo under both normal and pathological conditions. Turbulent and laminar flow elicit significantly different responses in endothelial cells (ECs), but the mechanisms allowing ECs to distinguish between these different flow regimes remain unknown. The authors present a computational model that describes the effect of turbulence on mechanical force transmission within ECs. Because turbulent flow is inherently "noisy" with random fluctuations in pressure and velocity, our model focuses on the effect of signal noise (a stochastically changing force) on the deformation of intracellular transduction sites including the nucleus, cell-cell adhesion proteins (CCAPs), and focal adhesion sites (FAS). The authors represent these components of the mechanical signaling pathway as linear viscoelastic structures (Kelvin bodies) connected to the cell surface via cytoskeletal elements. The authors demonstrate that FAS are more sensitive to signal noise than the nucleus or CCAP. The relative sensitivity of these various structures to noise is affected by the nature of the cytoskeletal connections within the cell. Finally, changes in the compliance of the nucleus dramatically affect nuclear sensitivity to noise, suggesting that pathologies that alter nuclear mechanical properties will be associated with abnormal EC responsiveness to turbulent flow.
Collapse
Affiliation(s)
- Bori Mazzag
- Department of Mathematics, Humboldt State University, 1 Harpst Street, Arcata, CA 95521, USA.
| | | |
Collapse
|
47
|
Mendoza SA, Fang J, Gutterman DD, Wilcox DA, Bubolz AH, Li R, Suzuki M, Zhang DX. TRPV4-mediated endothelial Ca2+ influx and vasodilation in response to shear stress. Am J Physiol Heart Circ Physiol 2009; 298:H466-76. [PMID: 19966050 DOI: 10.1152/ajpheart.00854.2009] [Citation(s) in RCA: 244] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The transient receptor potential vallinoid type 4 (TRPV4) channel has been implicated in the endothelial shear response and flow-mediated dilation, although the precise functions of this channel remain poorly understood. In the present study, we investigated the role of TRPV4 in shear stress-induced endothelial Ca(2+) entry and the potential link between this signaling response and relaxation of small resistance arteries. Using immunohistochemical analysis and RT-PCR, we detected strong expression of TRPV4 protein and mRNA in the endothelium in situ and endothelial cells freshly isolated from mouse small mesenteric arteries. The selective TRPV4 agonist GSK1016790A increased endothelial Ca(2+) and induced potent relaxation of small mesenteric arteries from wild-type (WT) but not TRPV4(-/-) mice. Luminal flow elicited endothelium-dependent relaxations that involved both nitric oxide and EDHFs. Both nitric oxide and EDHF components of flow-mediated relaxation were markedly reduced in TRPV4(-/-) mice compared with WT controls. Using a fura-2/Mn(2+) quenching assay, shear was observed to produce rapid Ca(2+) influx in endothelial cells, which was markedly inhibited by the TRPV4 channel blocker ruthenium red and TRPV4-specific short interfering RNA. Flow elicited a similar TRPV4-mediated Ca(2+) entry in HEK-293 cells transfected with TRPV4 channels but not in nontransfected cells. Collectively, these data indicate that TRPV4 may be a potential candidate of mechanosensitive channels in endothelial cells through which the shear stimulus is transduced into Ca(2+) signaling, leading to the release of endothelial relaxing factors and flow-mediated dilation of small resistance arteries.
Collapse
Affiliation(s)
- Suelhem A Mendoza
- Department of Medicine, Medical College of Wisconsin, 8701 Watertown Plank Rd., Milwaukee, WI 53226, USA
| | | | | | | | | | | | | | | |
Collapse
|
48
|
Choi HW, Barakat AI. Modulation of ATP/ADP concentration at the endothelial cell surface by flow: effect of cell topography. Ann Biomed Eng 2009; 37:2459-68. [PMID: 19763828 PMCID: PMC2778776 DOI: 10.1007/s10439-009-9793-z] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2009] [Accepted: 08/31/2009] [Indexed: 11/30/2022]
Abstract
Determining how flow affects the concentration of the adenine nucleotides ATP and ADP at the vascular endothelial cell (EC) surface is essential for understanding flow-induced mobilization of intracellular calcium. Previously, mathematical models were formulated to describe the ATP/ADP concentration at the EC surface; however, all previous models assumed the endothelium to be flat. In the present study we investigate the effect of surface undulations on ATP/ADP concentration at the EC surface. The results demonstrate that under certain geometric and flow conditions, the ATP + ADP concentration at the EC surface is considerably lower for a wavy cell surface than for a flat surface. Because ECs in regions of disturbed arterial flow are expected to have larger undulations than cells in non-disturbed flow zones, our findings suggest that ECs in regions of flow disturbance would exhibit lower ATP + ADP concentrations at their surfaces, which may lead to impaired calcium signaling. If validated experimentally, the present results may contribute to our understanding of endothelial cell dysfunction observed in regions of disturbed flow.
Collapse
Affiliation(s)
- Hyo Won Choi
- Department of Mechanical and Aeronautical Engineering, University of California, Davis, One Shields Avenue, Davis, CA 95616 USA
| | - Abdul I. Barakat
- Department of Mechanical and Aeronautical Engineering, University of California, Davis, One Shields Avenue, Davis, CA 95616 USA
| |
Collapse
|
49
|
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]
|
50
|
|