Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 65
1.
Diabetes ; 2024 Jun 21.
Article En | MEDLINE | ID: mdl-38905153

Despite advances in the treatment of atherosclerotic cardiovascular disease, it remains the leading cause of death in patients with diabetes. Even when risk factors are mitigated, the disease progresses, and thus newer targets need to be identified that directly inhibit the underlying pathobiology of atherosclerosis in diabetes. A single cell sequencing approach was utilised to distinguish the proatherogenic transcriptional profile in aortic cells in diabetes using a streptozotocin induced-diabetic Apoe-/- mouse model. Human carotid endarterectomy specimens from individuals with and without diabetes were also evaluated via immunohistochemical analysis. Further mechanistic studies were performed in human aortic endothelial cells and human THP-1 derived macrophages. We then performed a preclinical study using an AP-1 inhibitor in a diabetic Apoe-/- mouse model. Single cell RNA sequencing analysis identified the AP-1 complex as a novel target in diabetes-associated atherosclerosis. AP-1 levels were elevated in carotid endarterectomy specimens from diabetic when compared to non-diabetic individuals. AP-1 was validated as a mechanosensitive transcription factor via immunofluorescence staining for regional heterogeneity of endothelial cells of the aortic region exposed to turbulent blood flow and by performing microfluidics experiments in HAECs. AP-1 inhibition with T-5224 blunted endothelial cell activation as assessed by a monocyte adhesion assay and expression of genes relevant to endothelial function. Furthermore, AP-1 inhibition attenuated foam cell formation. Critically, treatment with T-5224 attenuated atherosclerosis development in diabetic Apoe-/- mice. This study has identified the AP-1 complex as a novel target, inhibition of which treats the underlying pathobiology of atherosclerosis in diabetes.

2.
Atherosclerosis ; 393: 117565, 2024 Jun.
Article En | MEDLINE | ID: mdl-38714426

Age-associated cardiovascular diseases (CVDs), predominantly resulting from artery-related disorders such as atherosclerosis, stand as a leading cause of morbidity and mortality among the elderly population. Consequently, there is a growing interest in the development of clinically relevant bioengineered models of CVDs. Recent developments in bioengineering and material sciences have paved the way for the creation of intricate models that closely mimic the structure and surroundings of native cardiac tissues and blood vessels. These models can be utilized for basic research purposes and for identifying pharmaceutical interventions and facilitating drug discovery. The advancement of vessel-on-a-chip technologies and the development of bioengineered and humanized in vitro models of the cardiovascular system have the potential to revolutionize CVD disease modelling. These technologies offer pathophysiologically relevant models at a fraction of the cost and time required for traditional experimentation required in vivo. This progress signifies a significant advancement in the field, transitioning from conventional 2D cell culture models to advanced 3D organoid and vessel-on-a-chip models. These innovative models are specifically designed to explore the complexities of vascular aging and stiffening, crucial factors in the development of cardiovascular diseases. This review summarizes the recent progress of various bioengineered in vitro platforms developed for investigating the pathophysiology of human cardiovascular system with more focus on advanced 3D vascular platforms.


Bioengineering , Cardiovascular Diseases , Lab-On-A-Chip Devices , Humans , Cardiovascular Diseases/physiopathology , Animals , Tissue Engineering/methods , Models, Cardiovascular , Organoids , Cell Culture Techniques
3.
Lab Chip ; 24(8): 2347-2357, 2024 Apr 16.
Article En | MEDLINE | ID: mdl-38576401

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.


Endothelial Cells , beta Catenin , Humans , beta Catenin/metabolism , Microfluidics , Aorta , Inflammation/metabolism , Stress, Mechanical , Cells, Cultured
4.
ACS Appl Mater Interfaces ; 15(51): 59103-59116, 2023 Dec 27.
Article En | MEDLINE | ID: mdl-38073418

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.


Atherosclerosis , Vascular Stiffness , Humans , Aged , Biomechanical Phenomena , Endothelial Cells/metabolism , Mechanotransduction, Cellular/physiology , Atherosclerosis/metabolism , Inflammation/metabolism , Stress, Mechanical
5.
ACS Biomater Sci Eng ; 9(12): 6947-6955, 2023 Dec 11.
Article En | MEDLINE | ID: mdl-38018792

Foam cell formation is a complex blood vessel pathology, which is characterized by a series of events, including endothelium dysfunction, inflammation, and accumulation of immune cells underneath the blood vessel walls. Novel bioengineered models capable of recapitulating these events are required to better understand the complex pathological processes underlying the development of foam cell formation and, consequently, advanced bioengineered platforms for screening drugs. Here, we generated a microfluidic blood vessel model, incorporating a three-dimensional (3D) extracellular matrix coated with an endothelial layer. This system enables us to perform experiments under a dynamic microenvironment that recapitulates the complexities of the native vascular regions. Using this model, we studied the effectors that regulate monocyte adhesion and migration, as well as foam cell formation inside vessel walls. We found that monocyte adhesion and migration are regulated by both the endothelium and monocytes themselves. Monocytes migrated into the extracellular matrix only when endothelial cells were cultured in the vessel model. In addition, the exposure of an endothelial layer to tumor necrosis factor α (TNF-α) and low shear stress both increased monocyte migration into the subendothelial space toward the matrix. Furthermore, we demonstrated the process of foam cell formation, 3 days after transmigration of peripheral blood mononuclear cells (PBMCs) into the vessel wall. We showed that pre-exposure of PBMCs to high shear rates increases their adhesion and migration through the TNF-α-treated endothelium but does not affect their capacity to form foam cells. The versatility of our model allows for mechanistic studies on foam cell formation under customized pathological conditions.


Endothelial Cells , Foam Cells , Foam Cells/metabolism , Foam Cells/pathology , Leukocytes, Mononuclear , Tumor Necrosis Factor-alpha/pharmacology , Tumor Necrosis Factor-alpha/metabolism , Monocytes/metabolism
7.
Biophys Rev ; 15(1): 19-33, 2023 Feb.
Article En | MEDLINE | ID: mdl-36909958

Cardiovascular diseases are the leading cause of mortality, morbidity, and hospitalization around the world. Recent technological advances have facilitated analyzing, visualizing, and monitoring cardiovascular diseases using emerging computational fluid dynamics, blood flow imaging, and wearable sensing technologies. Yet, computational cost, limited spatiotemporal resolution, and obstacles for thorough data analysis have hindered the utility of such techniques to curb cardiovascular diseases. We herein discuss how leveraging machine learning techniques, and in particular deep learning methods, could overcome these limitations and offer promise for translation. We discuss the remarkable capacity of recently developed machine learning techniques to accelerate flow modeling, enhance the resolution while reduce the noise and scanning time of current blood flow imaging techniques, and accurate detection of cardiovascular diseases using a plethora of data collected by wearable sensors.

8.
Sci Rep ; 13(1): 5202, 2023 03 30.
Article En | MEDLINE | ID: mdl-36997576

Macrophages are heterogeneous innate immune cells that are functionally shaped by their surrounding microenvironment. Diverse macrophage populations have multifaceted differences related to their morphology, metabolism, expressed markers, and functions, where the identification of the different phenotypes is of an utmost importance in modelling immune response. While expressed markers are the most used signature to classify phenotypes, multiple reports indicate that macrophage morphology and autofluorescence are also valuable clues that can be used in the identification process. In this work, we investigated macrophage autofluorescence as a distinct feature for classifying six different macrophage phenotypes, namely: M0, M1, M2a, M2b, M2c, and M2d. The identification was based on extracted signals from multi-channel/multi-wavelength flow cytometer. To achieve the identification, we constructed a dataset containing 152,438 cell events each having a response vector of 45 optical signals fingerprint. Based on this dataset, we applied different supervised machine learning methods to detect phenotype specific fingerprint from the response vector, where the fully connected neural network architecture provided the highest classification accuracy of 75.8% for the six phenotypes compared simultaneously. Furthermore, by restricting the number of phenotypes in the experiment, the proposed framework produces higher classification accuracies, averaging 92.0%, 91.9%, 84.2%, and 80.4% for a pool of two, three, four, five phenotypes, respectively. These results indicate the potential of the intrinsic autofluorescence for classifying macrophage phenotypes, with the proposed method being quick, simple, and cost-effective way to accelerate the discovery of macrophage phenotypical diversity.


Machine Learning , Macrophages , Macrophages/metabolism , Phenotype
9.
Anal Chem ; 95(5): 3089-3097, 2023 02 07.
Article En | MEDLINE | ID: mdl-36692453

Here, we describe the generation of dynamic vortices in micro-scale cavities at low flow rates. The system utilizes a computer-controlled audio speaker to axially oscillate the inlet tube of the microfluidic system at desired frequencies and amplitudes. The oscillation of the tube induces transiently high flow rates in the system, which facilitates the generation of dynamic vortices inside the cavity. The size of the vortices can be modulated by varying the tube oscillation frequency or amplitude. The vortices can be generated in single or serial cavities and in a wide range of cavity sizes. We demonstrate the suitability of the tube oscillation mechanism for the pulsed injection of water-based solutions or whole blood into the cavity. The injection rate can be controlled by the oscillation characteristics of the tube, enabling the injection of liquids at ultralow flow rates. The dynamic vortices facilitate the rapid mixing of the injected liquid with the main flow. The controllability and versatility of this technology allow for the development of programmable inertial microfluidic systems for performing multistep biological assays.


Blood Chemical Analysis , Microfluidics , Microfluidics/methods , Blood Chemical Analysis/methods
10.
ACS Appl Mater Interfaces ; 15(4): 4863-4872, 2023 Feb 01.
Article En | MEDLINE | ID: mdl-36652631

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.


Cell Culture Techniques , Endothelial Cells , Humans , Aged , Endothelial Cells/metabolism , Elasticity , Mechanical Phenomena , Cells, Cultured , Acrylamides/metabolism
11.
EMBO Mol Med ; 15(1): e16236, 2023 01 11.
Article En | MEDLINE | ID: mdl-36468184

C-reactive protein (CRP) is an early-stage acute phase protein and highly upregulated in response to inflammatory reactions. We recently identified a novel mechanism that leads to a conformational change from the native, functionally relatively inert, pentameric CRP (pCRP) structure to a pentameric CRP intermediate (pCRP*) and ultimately to the monomeric CRP (mCRP) form, both exhibiting highly pro-inflammatory effects. This transition in the inflammatory profile of CRP is mediated by binding of pCRP to activated/damaged cell membranes via exposed phosphocholine lipid head groups. We designed a tool compound as a low molecular weight CRP inhibitor using the structure of phosphocholine as a template. X-ray crystallography revealed specific binding to the phosphocholine binding pockets of pCRP. We provide in vitro and in vivo proof-of-concept data demonstrating that the low molecular weight tool compound inhibits CRP-driven exacerbation of local inflammatory responses, while potentially preserving pathogen-defense functions of CRP. The inhibition of the conformational change generating pro-inflammatory CRP isoforms via phosphocholine-mimicking compounds represents a promising, potentially broadly applicable anti-inflammatory therapy.


C-Reactive Protein , Phosphorylcholine , Humans , Phosphorylcholine/pharmacology , Inflammation/drug therapy , Inflammation/metabolism , Cell Membrane/metabolism , Anti-Inflammatory Agents
12.
Small Methods ; : e2301427, 2023 Dec 31.
Article En | MEDLINE | ID: mdl-38161266

Here, the generation of dynamic vortices across microscale barriers using the tube oscillation mechanism is demonstrated. Using a combination of high-speed imaging and computational flow dynamics, the cyclic formation, expansion, and collapse of vortices are studied. The dynamics of vortices across circular , triangular, and blade-shape barriers are investigated at different tube oscillation frequencies. The formation of an array of synchronous vortices across parallel blade-shaped barriers is demonstrated. The transient flows caused by these dynamic vortex arrays are harnessed for the rapid and efficient mixing of blood samples . A circular barrier scribed with a narrow orifice on its shoulder is used to facilitate the injection of liquid into the microfluidic channel, and its rapid mixing with the main flow through the dynamic vortices generated across the barrier. This approach facilitates the generation of vortices with desirable configurations, sizes, and dynamics in a highly controllable, programmable, and predictable manner while operating at low static flow rates.

13.
Sci Rep ; 12(1): 18907, 2022 11 07.
Article En | MEDLINE | ID: mdl-36344673

Photobiomodulation (PBM) refers to the use of light to modulate cellular processes, and has demonstrated utility in improving wound healing outcomes, and reducing pain and inflammation. Despite the potential benefits of PBM, the precise molecular mechanisms through which it influences cell behavior are not yet well understood. Inconsistent reporting of key light parameters has created uncertainty around optimal exposure profiles. In addition, very low intensities of light, < 0.1 J/cm2, have not been thoroughly examined for their use in PBM. Here, we present a custom-made compact, and modular LED-based exposure system for studying the effects of very low-intensity visible light (cell proliferation, migration, ROS production, and mitochondrial membrane potential) of three different wavelengths in a parallel manner. The device allows for six repeats of three different exposure conditions plus a non-irradiated control on a single 24-well plate. The immortalised human keratinocyte cell line, HaCaT, was selected as a major cellular component of the skin epidermal barrier. Furthermore, an in vitro wound model was developed by allowing the HaCaT to form a confluent monolayer, then scratching the cells with a pipette tip to form a wound. Cells were exposed to yellow (585 nm, 0.09 mW, ~ 3.7 mJ/cm2), orange (610 nm, 0.8 mW, ~ 31 mJ/cm2), and red (660 nm, 0.8 mW, ~ 31 mJ/cm2) light for 10 min. 48 h post-irradiation, immunohistochemistry was performed to evaluate cell viability, proliferation, ROS production, and mitochondrial membrane potential. The results demonstrate increased proliferation and decreased scratch area for all exposure conditions, however only red light increased the mitochondrial activity. Oxidative stress levels did not increase for any of the exposures. The present exposure system provides opportunities to better understand the complex cellular mechanisms driven by the irradiation of skin cells with visible light.


Low-Level Light Therapy , Humans , Low-Level Light Therapy/methods , Reactive Oxygen Species/metabolism , Keratinocytes/metabolism , Wound Healing/radiation effects , Cell Proliferation/radiation effects , Light
14.
ACS Appl Mater Interfaces ; 14(36): 40559-40568, 2022 Sep 14.
Article En | MEDLINE | ID: mdl-36047858

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.


Ion Channels , Mechanotransduction, Cellular , Extracellular Matrix/metabolism , Extracellular Matrix Proteins/metabolism , HEK293 Cells , Humans , Integrins/metabolism , Mechanotransduction, Cellular/physiology
15.
Lab Chip ; 22(10): 1917-1928, 2022 05 17.
Article En | MEDLINE | ID: mdl-35420623

Microfluidic systems incorporating sudden expansions are widely used for generation of vortex flow patterns. However, the formation of vortices requires high flow rates to induce inertial effects. Here, we introduce a new method for generating dynamic vortices in microfluidics at low static flow rates. Human blood is driven through a microfluidic channel incorporating a semi-circular stenosis barrier. The inlet tube of the channel is axially oscillated using a computer-controlled audio-speaker. The tube oscillation induces high transient flow rates in the channel, which generates dynamic vortices across the stenosis barrier. The size of the vortices can be modulated by varying the frequency and amplitude of tube oscillation. Various vortex flow patterns can be generated by varying the flow rate. The formation and size of the vortices can be predicted using the Reynolds number of the oscillating tube. We demonstrate the potential application of the system for investigating the adhesion and phagocytosis of circulating immune cells under pathologically high shear rates induced at the stenosis. This approach facilitates the development of versatile and controllable inertial microfluidic systems for performing various cellular assays while operating at low static flow rates and low sample volumes.


Microfluidics , Constriction, Pathologic , Humans , Microfluidics/methods
16.
Front Cardiovasc Med ; 9: 783543, 2022.
Article En | MEDLINE | ID: mdl-35355968

Calcific aortic valve disease (CAVD) is a common acquired valvulopathy, which carries a high burden of mortality. Chronic inflammation has been postulated as the predominant pathophysiological process underlying CAVD. So far, no effective medical therapies exist to halt the progression of CAVD. This review aims to outline the known pathways of inflammation and calcification in CAVD, focussing on the critical roles of mechanical stress and mechanosensing in the perpetuation of valvular inflammation. Following initiation of valvular inflammation, dysregulation of proinflammatory and osteoregulatory signalling pathways stimulates endothelial-mesenchymal transition of valvular endothelial cells (VECs) and differentiation of valvular interstitial cells (VICs) into active myofibroblastic and osteoblastic phenotypes, which in turn mediate valvular extracellular matrix remodelling and calcification. Mechanosensitive signalling pathways convert mechanical forces experienced by valve leaflets and circulating cells into biochemical signals and may provide the positive feedback loop that promotes acceleration of disease progression in the advanced stages of CAVD. Mechanosensing is implicated in multiple aspects of CAVD pathophysiology. The mechanosensitive RhoA/ROCK and YAP/TAZ systems are implicated in aortic valve leaflet mineralisation in response to increased substrate stiffness. Exposure of aortic valve leaflets, endothelial cells and platelets to high shear stress results in increased expression of mediators of VIC differentiation. Upregulation of the Piezo1 mechanoreceptor has been demonstrated to promote inflammation in CAVD, which normalises following transcatheter valve replacement. Genetic variants and inhibition of Notch signalling accentuate VIC responses to altered mechanical stresses. The study of mechanosensing pathways has revealed promising insights into the mechanisms that perpetuate inflammation and calcification in CAVD. Mechanotransduction of altered mechanical stresses may provide the sought-after coupling link that drives a vicious cycle of chronic inflammation in CAVD. Mechanosensing pathways may yield promising targets for therapeutic interventions and prognostic biomarkers with the potential to improve the management of CAVD.

17.
Biol Rev Camb Philos Soc ; 97(2): 604-614, 2022 04.
Article En | MEDLINE | ID: mdl-34781417

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.


Cardiovascular System , Ion Channels , Mechanotransduction, Cellular , Humans , Ion Channels/metabolism , Mechanotransduction, Cellular/physiology , Signal Transduction
18.
Biosens Bioelectron ; 198: 113814, 2022 Feb 15.
Article En | MEDLINE | ID: mdl-34823964

The detection of cancer cells at the single-cell level enables many novel functionalities such as next-generation cancer prognosis and accurate cellular analysis. While surface-enhanced Raman spectroscopy (SERS) has been widely considered as an effective tool in a low-cost and label-free manner, however, it is challenging to discriminate single cancer cells with an accuracy above 90% mainly due to the poor biocompatibility of the noble-metal-based SERS agents. Here, we report a dual-functional nanoprobe based on dopant-driven plasmonic oxides, demonstrating a maximum accuracy above 90% in distinguishing single THP-1 cell from peripheral blood mononuclear cell (PBMC) and human embryonic kidney (HEK) 293 from human macrophage cell line U937 based on their SERS patterns. Furthermore, this nanoprobe can be triggered by the bio-redox response from individual cells towards stimuli, empowering another complementary colorimetric cell detection, approximately achieving the unity discrimination accuracy at a single-cell level. Our strategy could potentially enable the future accurate and low-cost detection of cancer cells from mixed cell samples.


Biosensing Techniques , Metal Nanoparticles , Neoplasms , HEK293 Cells , Humans , Leukocytes, Mononuclear , Neoplasms/diagnosis , Oxides , Spectrum Analysis, Raman
19.
Lab Chip ; 22(2): 262-271, 2022 01 18.
Article En | MEDLINE | ID: mdl-34931212

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.


Ion Channels , Mechanotransduction, Cellular , HEK293 Cells , Humans , Mechanotransduction, Cellular/physiology , Printing, Three-Dimensional , Stress, Mechanical
20.
Front Bioeng Biotechnol ; 9: 791116, 2021.
Article En | MEDLINE | ID: mdl-34957080

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.

...