Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
Lab Chip ; 24(8): 2347-2357, 2024 Apr 16.
Article in English | MEDLINE | ID: mdl-38576401

ABSTRACT

Atrial fibrillation (AF) is the most common type of cardiac arrhythmia and an important contributor to morbidity and mortality. Endothelial dysfunction has been postulated to be an important contributing factor in cardiovascular events in patients with AF. However, how vascular endothelial cells respond to arrhythmic flow is not fully understood, mainly due to the limitation of current in vitro systems to mimic arrhythmic flow conditions. To address this limitation, we developed a microfluidic system to study the effect of arrhythmic flow on the mechanobiology of human aortic endothelial cells (HAECs). The system utilises a computer-controlled piezoelectric pump for generating arrhythmic flow with a unique ability to control the variability in both the frequency and amplitude of pulse waves. The flow rate is modulated to reflect physiological or pathophysiological shear stress levels on endothelial cells. This enabled us to systematically dissect the importance of variability in the frequency and amplitude of pulses and shear stress level on endothelial cell mechanobiology. Our results indicated that arrhythmic flow at physiological shear stress level promotes endothelial cell spreading and reduces the plasma membrane-to-cytoplasmic distribution of ß-catenin. In contrast, arrhythmic flow at low and atherogenic shear stress levels does not promote endothelial cell spreading or redistribution of ß-catenin. Interestingly, under both shear stress levels, arrhythmic flow induces inflammation by promoting monocyte adhesion via an increase in ICAM-1 expression. Collectively, our microfluidic system provides opportunities to study the effect of arrhythmic flows on vascular endothelial mechanobiology in a systematic and reproducible manner.


Subject(s)
Endothelial Cells , beta Catenin , Humans , beta Catenin/metabolism , Microfluidics , Aorta , Inflammation/metabolism , Stress, Mechanical , Cells, Cultured
2.
ACS Appl Mater Interfaces ; 15(51): 59103-59116, 2023 Dec 27.
Article in English | MEDLINE | ID: mdl-38073418

ABSTRACT

How endothelial cells sense and respond to dynamic changes in their biophysical surroundings as we age is not fully understood. Vascular stiffness is clearly a contributing factor not only in several cardiovascular diseases but also in physiological processes such as aging and vascular dementia. To address this gap, we utilized a microfluidic model to explore how substrate stiffness in the presence of shear stress affects endothelial morphology, senescence, proliferation, and inflammation. We also studied the role of mechanosensitive ion channel Piezo1 in endothelial responses under the combined effect of shear stress and substrate stiffness. To do so, we cultured endothelial cells inside microfluidic channels covered with fibronectin-coated elastomer with elastic moduli of 40 and 200 kPa, respectively, mimicking the stiffness of the vessel walls in young and aged arteries. The endothelial cells were exposed to atheroprotective and atherogenic shear stress levels of 10 and 2 dyn/cm2, respectively. Our findings show that substrate stiffness affects senescence under atheroprotective flow conditions and cytoskeleton remodeling, senescence, and inflammation under atherogenic flow conditions. Additionally, we found that the expression of Piezo1 plays a crucial role in endothelial adaptation to flow and regulation of inflammation under both atheroprotective and atherogenic shear stress levels. However, Piezo1 contribution to endothelial senescence was limited to the soft substrate and atheroprotective shear stress level. Overall, our study characterizes the response of endothelial cells to the combined effect of shear stress and substrate stiffness and reveals a previously unidentified role of Piezo1 in endothelial response to vessel stiffening, which potentially can be therapeutically targeted to alleviate endothelial dysfunction in aging adults.


Subject(s)
Atherosclerosis , Vascular Stiffness , Humans , Aged , Biomechanical Phenomena , Endothelial Cells/metabolism , Mechanotransduction, Cellular/physiology , Atherosclerosis/metabolism , Inflammation/metabolism , Stress, Mechanical
3.
Curr Allergy Asthma Rep ; 23(8): 435-442, 2023 08.
Article in English | MEDLINE | ID: mdl-37233850

ABSTRACT

PURPOSE OF REVIEW: Atopic dermatitis (AD) remains a dermatological disease that imposes a significant burden on society. Air pollution has previously been linked to both the onset and severity of atopic dermatitis. As air pollution remains a critical environmental factor impacting human health, this review seeks to provide an overview of the relationship between different air pollutants and AD. RECENT FINDINGS: AD can develop from multiple causes that can be broadly grouped into epidermal barrier dysfunction and immune dysregulation. Air pollution imposes significant health risks and includes a wide variety of pollutant types. AD has been linked to outdoor air pollutants such as particulate matter (PM), volatile organic compounds (VOC), gaseous compounds, and heavy metals. Exposure to indoor pollutants such as tobacco smoke and fungal molds has also been associated with an increased incidence of AD. While different pollutants impact distinct molecular pathways in the cell, they mostly converge on ROS product, DNA damage, and dysregulated T-cell activity and cytokine production. The presented review suggests a strengthening tie between air pollution and AD. It points to opportunities for further studies to clarify, as well as potential therapeutic opportunities that leverage the mechanistic relationships between air pollution and AD.


Subject(s)
Air Pollutants , Air Pollution , Dermatitis, Atopic , Environmental Pollutants , Humans , Dermatitis, Atopic/epidemiology , Dermatitis, Atopic/etiology , Air Pollution/adverse effects , Air Pollutants/adverse effects , Particulate Matter/adverse effects
4.
ACS Appl Mater Interfaces ; 15(4): 4863-4872, 2023 Feb 01.
Article in English | MEDLINE | ID: mdl-36652631

ABSTRACT

Endothelial cells lining blood vessels are continuously exposed to biophysical cues that regulate their function in health and disease. As we age, blood vessels lose their elasticity and become stiffer. Vessel stiffness alters the mechanical forces that endothelial cells experience. Despite ample evidence on the contribution of endothelial cells to vessel stiffness, less is known about how vessel stiffness affects endothelial cells. In this study, we developed a versatile model to study the cooperative effect of substrate stiffness and cyclic stretch on human aortic endothelial cells. We cultured endothelial cells on elastomeric wells covered with fibronectin-coated polyacrylamide gel. Varying the concentrations of acrylamide and bis-acrylamide enabled us to produce soft and stiff substrates with elastic modules of 40 and 200 kPa, respectively. Using a customized three-dimensional (3D) printed cam-driven system, the cells were exposed to 5 and 10% cyclic stretch levels. This enabled us to mimic the stiffness and stretch levels that endothelial cells experience in young and aged arteries. Using this model, we found that endothelial cells cultured on a soft substrate had minimal cytoskeletal alignment to the direction of the stretch compared to the ones cultured on the stiff substrate. We also observed an increase in the cellular area and aspect ratio in cells cultured on the stiff substrate, both of which are positively regulated by cyclic stretch. However, neither cyclic stretch nor substrate stiffness significantly affected the nuclear circularity. Additionally, we found that the accumulation of NF-κB in the nucleus, endothelial proliferation, tube formation, and expression of IL1ß depends on the stretch level and substrate stiffness. Our model can be further used to investigate the complex signaling pathways associated with vessel stiffening that govern the endothelial responses to mechanical forces.


Subject(s)
Cell Culture Techniques , Endothelial Cells , Humans , Aged , Endothelial Cells/metabolism , Elasticity , Mechanical Phenomena , Cells, Cultured , Acrylamides/metabolism
5.
ACS Appl Mater Interfaces ; 14(36): 40559-40568, 2022 Sep 14.
Article in English | MEDLINE | ID: mdl-36047858

ABSTRACT

Piezo1 is a recently discovered Ca2+ permeable ion channel that has emerged as an integral sensor of hemodynamic forces within the cardiovascular system, contributing to vascular development and blood pressure regulation. However, how the composition of the extracellular matrix (ECM) affects the mechanosensitivity of Piezo1 in response to hemodynamic forces remains poorly understood. Using a combination of microfluidics and calcium imaging techniques, we probe the shear stress sensitivity of single HEK293T cells engineered to stably express Piezo1 in the presence of different ECM proteins. Our experiments show that Piezo1 sensitivity to shear stress is not dependent on the presence of ECM proteins. However, different ECM proteins regulate the sensitivity of Piezo1 depending on the shear stress level. Under high shear stress, fibronectin sensitizes Piezo1 response to shear, while under low shear stress, Piezo1 mechanosensitivity is improved in the presence of collagen types I and IV and laminin. Moreover, we report that α5ß1 and αvß3 integrins are involved in Piezo1 sensitivity at high shear, while αvß3 and αvß5 integrins are involved in regulating the Piezo1 response at low shear stress. These results demonstrate that the ECM/integrin interactions influence Piezo1 mechanosensitivity and could represent a mechanism whereby extracellular forces are transmitted to Piezo1 channels, providing new insights into the mechanism by which Piezo1 senses shear stress.


Subject(s)
Ion Channels , Mechanotransduction, Cellular , Extracellular Matrix/metabolism , Extracellular Matrix Proteins/metabolism , HEK293 Cells , Humans , Integrins/metabolism , Mechanotransduction, Cellular/physiology
6.
Neurocrit Care ; 36(1): 240-247, 2022 02.
Article in English | MEDLINE | ID: mdl-34296399

ABSTRACT

BACKGROUND: Clazosentan, an endothelin-1 receptor antagonist, has been shown to prevent the development of large vessel angiographic vasospasm after aneurysmal subarachnoid hemorrhage. We hypothesized that clazosentan can improve cerebral perfusion for territories affected by angiographically confirmed vasospasm. METHODS: The REVERSE study (REversal of Vasospasm with clazosEntan post-aneuRysmal Subarachnoid hEmorrhage) was a prospective multicenter open-label pilot study of adult patients with aneurysmal subarachnoid hemorrhage who received intravenous clazosentan after developing moderate to severe angiographic vasospasm. Using the radiographic data from the REVERSE study and additional retrospective radiographic data from our tertiary medical center, we compared the impact of intravenous clazosentan with intraarterial vasodilator therapy (medical standard of care) on vasospasm reversal using time to peak perfusion (TTPP; the time interval between the peak opacification of contrast dye in the main artery supplying an anatomically defined territory and the parenchymal phase when the dye is diffusely present in the brain parenchyma). RESULTS: Both intravenous clazosentan (n = 7 vessels) and intraarterial vasodilator therapy (n = 11 vessels) resulted in a statistically significant improvement in TTPP at 24 h post intervention, when compared with the TTPP just prior to intervention for territories with angiographically confirmed severe vasospasm in the proximal arteries at baseline (linear mixed-effect model, p = 0.02). The clazosentan and intraarterial vasodilator therapy groups exhibited no statistically significant interaction term [time x treatment group (medical standard of care vs. clazosentan)] in our model (p = 0.71), suggesting similar temporal course of two therapies. CONCLUSIONS: In our small pilot study, intravenous clazosentan administered for at least 24 h had an effect comparable with that of intraarterial vasodilator therapy in reversing angiographically confirmed severe vasospasm. Our results may indicate that clazosentan, in an appropriately selected patient cohort, could offer a noninvasive approach for alleviating vasospasm.


Subject(s)
Subarachnoid Hemorrhage , Vasospasm, Intracranial , Adult , Dioxanes , Humans , Perfusion , Pilot Projects , Prospective Studies , Pyridines , Pyrimidines , Retrospective Studies , Subarachnoid Hemorrhage/complications , Subarachnoid Hemorrhage/drug therapy , Sulfonamides , Tetrazoles , Treatment Outcome , Vasospasm, Intracranial/diagnostic imaging , Vasospasm, Intracranial/drug therapy , Vasospasm, Intracranial/etiology
7.
Biol Rev Camb Philos Soc ; 97(2): 604-614, 2022 04.
Article in English | MEDLINE | ID: mdl-34781417

ABSTRACT

Piezo1 is a mechanosensitive ion channel with essential roles in cardiovascular, lung, urinary, and immune functions. Piezo1 is widely distributed in different tissues in the human body and its specific roles have been identified following a decade of research; however, not all are well understood. Many structural and functional characteristics of Piezo1 have been discovered and are known to differ greatly from the characteristics of other mechanosensitive ion channels. Understanding the mechanisms by which this ion channel functions may be useful in determining its physiological roles in various organ systems. This review provides insight into the signalling pathways activated by mechanical stimulation of Piezo1 in various organ systems and cell types. We discuss downstream targets of Piezo1 and the overall effects resulting from Piezo1 activation, which may provide insights into potential treatment targets for diseases involving this ion channel.


Subject(s)
Cardiovascular System , Ion Channels , Mechanotransduction, Cellular , Humans , Ion Channels/metabolism , Mechanotransduction, Cellular/physiology , Signal Transduction
8.
Lab Chip ; 22(2): 262-271, 2022 01 18.
Article in English | MEDLINE | ID: mdl-34931212

ABSTRACT

Microfluidic systems are widely used for studying the mechanotransduction of flow-induced shear stress in mechanosensitive cells. However, these studies are generally performed under constant flow rates, mainly, due to the deficiency of existing pumps for generating transient flows. To address this limitation, we have developed a unique dynamic gravity pump to generate transient flows in microfluidics. The pump utilises a motorised 3D-printed cam-lever mechanism to change the inlet pressure of the system in repeated cycles. 3D printing technology facilitates the rapid and low-cost prototyping of the pump. Customised transient flow patterns can be generated by modulating the profile, size, and rotational speed of the cam, location of the hinge along the lever, and the height of the syringe. Using this unique dynamic gravity pump, we investigated the mechanotransduction of shear stress in HEK293 cells stably expressing Piezo1 mechanosensitive ion channel under transient flows. The controllable, simple, low-cost, compact, and modular design of the pump makes it suitable for studying the mechanobiology of shear sensitive cells under transient flows.


Subject(s)
Ion Channels , Mechanotransduction, Cellular , HEK293 Cells , Humans , Mechanotransduction, Cellular/physiology , Printing, Three-Dimensional , Stress, Mechanical
9.
Front Bioeng Biotechnol ; 9: 791116, 2021.
Article in English | MEDLINE | ID: mdl-34957080

ABSTRACT

Here, we describe a motorized cam-driven system for the cyclic stretch of aortic endothelial cells. Our modular design allows for generating customized spatiotemporal stretch profiles by varying the profile and size of 3D printed cam and follower elements. The system is controllable, compact, inexpensive, and amenable for parallelization and long-term experiments. Experiments using human aortic endothelial cells show significant changes in the cytoskeletal structure and morphology of cells following exposure to 5 and 10% cyclic stretch over 9 and 16 h. The system provides upportunities for exploring the complex molecular and cellular processes governing the response of mechanosensitive cells under cyclic stretch.

10.
J Cell Physiol ; 236(4): 2976-2987, 2021 04.
Article in English | MEDLINE | ID: mdl-32959903

ABSTRACT

Mechanosensitive ion channels mediate endothelial responses to blood flow and orchestrate their physiological function in response to hemodynamic forces. In this study, we utilized microfluidic technologies to study the shear-induced sensitization of endothelial Piezo-1 to its selective agonist, Yoda-1. We demonstrated that shear stress-induced sensitization is brief and can be impaired when exposing aortic endothelial cells to low and proatherogenic levels of shear stress. Our results suggest that shear stress-induced sensitization of Piezo-1 to Yoda-1 is independent of cell-cell adhesion and is mediated by the PI3K-AKT signaling pathway. We also found that shear stress increases the membrane density of Piezo-1 channels in endothelial cells. To further confirm our findings, we performed experiments using a carotid artery ligation mouse model and demonstrated that transient changes in blood-flow pattern, resulting from a high-degree ligation of the mouse carotid artery alters the distribution of Piezo-1 channels across the endothelial layer. These results suggest that shear stress influences the function of Piezo-1 channels via changes in membrane density, providing a new model of shear-stress sensitivity for Piezo-1 ion channel.


Subject(s)
Aorta/cytology , Endothelial Cells/metabolism , Ion Channels/metabolism , Mechanotransduction, Cellular , Stress, Mechanical , Calcium/metabolism , Cell Adhesion , Cytoskeleton/metabolism , Dynamins/metabolism , HEK293 Cells , Humans , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Pyrazines/metabolism , Rheology , Signal Transduction , Thiadiazoles/metabolism
11.
iScience ; 23(5): 101132, 2020 May 22.
Article in English | MEDLINE | ID: mdl-32434143

ABSTRACT

Maintaining a balance between protein degradation and protein synthesis is necessary for neurodevelopment. Although the E3 ubiquitin ligase anaphase promoting complex and its regulatory subunit Cdh1 (Cdh1-APC) has been shown to regulate learning and memory, the underlying mechanisms are unclear. Here, we have identified a role of Cdh1-APC as a regulator of protein synthesis in neurons. Proteomic profiling revealed that Cdh1-APC interacts with known regulators of translation, including stress granule proteins. Inhibition of Cdh1-APC activity caused an increase in stress granule formation that is dependent on fragile X mental retardation protein (FMRP). We propose a model in which Cdh1-APC targets stress granule proteins, such as FMRP, and inhibits the formation of stress granules, leading to protein synthesis. Elucidation of a role for Cdh1-APC in regulation of stress granules and protein synthesis in neurons has implications for how Cdh1-APC can regulate protein-synthesis-dependent synaptic plasticity underlying learning and memory.

12.
Traffic ; 21(7): 454-462, 2020 07.
Article in English | MEDLINE | ID: mdl-32374065

ABSTRACT

RNA granule formation, which can be regulated by RNA-binding proteins (RBPs) such as fragile X mental retardation protein (FMRP), acts as a mechanism to control both the repression and subcellular localization of translation. Dysregulated assembly of RNA granules has been implicated in multiple neurological disorders, such as amyotrophic lateral sclerosis. Thus, it is crucial to understand the cellular pathways impinging upon granule assembly or disassembly. The goal of this review is to summarize recent advances in our understanding of the role of the RBP, FMRP, in translational repression underlying RNA granule dynamics, mRNA transport and localized. We summarize the known mechanisms of translational regulation by FMRP, the role of FMRP in RNA transport granules, fragile X granules and stress granules. Focusing on the emerging link between FMRP and stress granules, we propose a model for how hyperassembly and hypoassembly of RNA granules may contribute to neurological diseases.


Subject(s)
Amyotrophic Lateral Sclerosis , Fragile X Mental Retardation Protein , Amyotrophic Lateral Sclerosis/genetics , Cytoplasmic Granules , Fragile X Mental Retardation Protein/genetics , Humans , RNA
13.
Front Pharmacol ; 10: 6, 2019.
Article in English | MEDLINE | ID: mdl-30728775

ABSTRACT

TRPV4 is a non-selective cation channel that tunes the function of different tissues including the vascular endothelium, lung, chondrocytes, and neurons. GSK1016790A is the selective and potent agonist of TRPV4 and a pharmacological tool that is used to study the TRPV4 physiological function in vitro and in vivo. It remains unknown how the sensitivity of TRPV4 to this agonist is regulated. The spatial and temporal dynamics of receptors are the major determinants of cellular responses to stimuli. Membrane translocation has been shown to control the response of several members of the transient receptor potential (TRP) family of ion channels to different stimuli. Here, we show that TRPV4 stimulation with GSK1016790A caused an increase in [Ca2+]i that is stable for a few minutes. Single molecule analysis of TRPV4 channels showed that the density of TRPV4 at the plasma membrane is controlled through two modes of membrane trafficking, complete, and partial vesicular fusion. Further, we show that the density of TRPV4 at the plasma membrane decreased within 20 min, as they translocate to the recycling endosomes and that the surface density is dependent on the release of calcium from the intracellular stores and is controlled via a PI3K, PKC, and RhoA signaling pathway.

SELECTION OF CITATIONS
SEARCH DETAIL
...