1
|
Kosuru R, Romito O, Sharma GP, Ferraresso F, Ghadrdoost Nakhchi B, Yang K, Mammoto T, Mammoto A, Kastrup CJ, Zhang DX, Goldspink PH, Trebak M, Chrzanowska M. Rap1A Modulates Store-Operated Calcium Entry in the Lung Endothelium: A Novel Mechanism Controlling NFAT-Mediated Vascular Inflammation and Permeability. Arterioscler Thromb Vasc Biol 2024; 44:2271-2287. [PMID: 39324266 PMCID: PMC11495542 DOI: 10.1161/atvbaha.124.321458] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Accepted: 09/05/2024] [Indexed: 09/27/2024]
Abstract
BACKGROUND Store-operated calcium entry mediated by STIM (stromal interaction molecule)-1-Orai1 (calcium release-activated calcium modulator 1) is essential in endothelial cell (EC) functions, affecting signaling, NFAT (nuclear factor for activated T cells)-induced transcription, and metabolic programs. While the small GTPase Rap1 (Ras-proximate-1) isoforms, including the predominant Rap1B, are known for their role in cadherin-mediated adhesion, EC deletion of Rap1A after birth uniquely disrupts lung endothelial barrier function. Here, we elucidate the specific mechanisms by which Rap1A modulates lung vascular integrity and inflammation. METHODS The role of EC Rap1A in lung inflammation and permeability was examined using in vitro and in vivo approaches. RESULTS We explored Ca2+ signaling in human ECs following siRNA-mediated knockdown of Rap1A or Rap1B. Rap1A knockdown, unlike Rap1B, significantly increased store-operated calcium entry in response to a GPCR (G-protein-coupled receptor) agonist, ATP (500 µmol/L), or thapsigargin (250 nmol/L). This enhancement was attenuated by Orai1 channel blockers 10 μmol/L BTP2 (N-[4-[3,5-bis(trifluoromethyl)-1H-pyrazol-1-yl]phenyl]-4-methyl-1,2,3-thiadiazole-5-carboxamide), 10 μmol/L GSK-7975A, and 5 μmol/L Gd3+. Whole-cell patch clamp measurements revealed enhanced Ca2+ release-activated Ca2+ current density in siRap1A ECs. Rap1A depletion in ECs led to increased NFAT1 nuclear translocation and activity and elevated levels of proinflammatory cytokines (CXCL1 [C-X-C motif chemokine ligand 1], CXCL11 [C-X-C motif chemokine 11], CCL5 [chemokine (C-C motif) ligand 5], and IL-6 [interleukin-6]). Notably, reducing Orai1 expression in siRap1A ECs normalized store-operated calcium entry, NFAT activity, and endothelial hyperpermeability in vitro. EC-specific Rap1A knockout (Rap1AiΔEC) mice displayed an inflammatory lung phenotype with increased lung permeability and inflammation markers, along with higher Orai1 expression. Delivery of siRNA against Orai1 to lung endothelium using lipid nanoparticles effectively normalized Orai1 levels in lung ECs, consequently reducing hyperpermeability and inflammation in Rap1AiΔEC mice. CONCLUSIONS Our findings uncover a novel role of Rap1A in regulating Orai1-mediated Ca2+ entry and expression, crucial for NFAT-mediated transcription and endothelial inflammation. This study distinguishes the unique function of Rap1A from that of the predominant Rap1B isoform and highlights the importance of normalizing Orai1 expression in maintaining lung vascular integrity and modulating endothelial functions.
Collapse
Affiliation(s)
- Ramoji Kosuru
- Versiti Blood Research Institute, Milwaukee, WI (R.K., G.P.S., F.F., B.G.N., C.J.K., M.C.)
| | - Olivier Romito
- Department of Pharmacology and Chemical Biology (O.R., M.T.), University of Pittsburgh School of Medicine, PA
| | - Guru Prasad Sharma
- Versiti Blood Research Institute, Milwaukee, WI (R.K., G.P.S., F.F., B.G.N., C.J.K., M.C.)
| | - Francesca Ferraresso
- Versiti Blood Research Institute, Milwaukee, WI (R.K., G.P.S., F.F., B.G.N., C.J.K., M.C.)
| | | | - Kai Yang
- Data Science Institute (K.Y.), Medical College of Wisconsin, Milwaukee
| | - Tadanori Mammoto
- Department of Pediatrics (T.M., A.M.), Medical College of Wisconsin, Milwaukee
| | - Akiko Mammoto
- Department of Pediatrics (T.M., A.M.), Medical College of Wisconsin, Milwaukee
| | - Christian J. Kastrup
- Versiti Blood Research Institute, Milwaukee, WI (R.K., G.P.S., F.F., B.G.N., C.J.K., M.C.)
| | - David X. Zhang
- Department of Medicine (D.X.Z.), Medical College of Wisconsin, Milwaukee
| | - Paul H. Goldspink
- Department of Physiology and Biophysics, University of Illinois Chicago (P.H.G.)
| | - Mohamed Trebak
- Department of Pharmacology and Chemical Biology (O.R., M.T.), University of Pittsburgh School of Medicine, PA
- Vascular Medicine Institute (M.T.), University of Pittsburgh School of Medicine, PA
- UPMC Hillman Cancer Center (M.T.), University of Pittsburgh School of Medicine, PA
| | - Magdalena Chrzanowska
- Versiti Blood Research Institute, Milwaukee, WI (R.K., G.P.S., F.F., B.G.N., C.J.K., M.C.)
- Department of Pharmacology and Toxicology (M.C.), Medical College of Wisconsin, Milwaukee
- Cardiovascular Center (M.C.), Medical College of Wisconsin, Milwaukee
| |
Collapse
|
2
|
Schaller L, Gudermann T, Dietrich A. TRPV4 Mediates Alveolar Epithelial Barrier Integrity and Induces ADAM10-Driven E-Cadherin Shedding. Cells 2024; 13:1717. [PMID: 39451235 PMCID: PMC11506556 DOI: 10.3390/cells13201717] [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: 09/23/2024] [Revised: 10/11/2024] [Accepted: 10/15/2024] [Indexed: 10/26/2024] Open
Abstract
Transient receptor potential vanilloid 4 (TRPV4) channels have been associated with numerous pulmonary pathologies, including hypertension, asthma, and acute lung injury. However, their role in the alveolar epithelium remains unclear. We performed impedance-based resistance measurements in primary differentiated alveolar epithelial type I (AT1) cells from wild-type (WT) and TRPV4-deficient (TRPV4-/-) C57/BL6J mice to detect changes in AT1 barrier integrity upon TRPV4 activation. Both pharmacological (GSK1016790A) and a low pH-driven activation of TRPV4 were quantified, and the downstream effects on adherens junctions were assessed through the Western blotting of epithelial cadherin (E-cadherin) protein levels. Importantly, a drop in pH caused a rapid decrease in AT1 barrier resistance and increased the formation of a ~35 kDa E-cadherin C-terminal fragment, with both effects significantly reduced in TRPV4-/- AT1 cells. Similarly, the pharmacological activation of TRPV4 in AT1 cells triggered an immediate transient loss of barrier resistance and the formation of the same E-cadherin fragment, which was again diminished by TRPV4 deficiency. Moreover, TRPV4-mediated E-cadherin cleavage was significantly reduced by GI254023X, an antagonist of a disintegrin and metalloprotease 10 (ADAM10). Our results confirm the role of TRPV4 in regulating alveolar epithelial barrier permeability and provide insight into a novel signaling pathway by which TRPV4-induced Ca2+ influx stimulates metalloprotease-driven ectodomain shedding.
Collapse
Affiliation(s)
| | | | - Alexander Dietrich
- Walther Straub Institute for Pharmacology and Toxicology, Member of the German Center for Lung Research (DZL), Medical Faculty, LMU-Munich, Nussbaumstrasse 26, 80336 Munich, Germany; (L.S.); (T.G.)
| |
Collapse
|
3
|
Ugodnikov A, Chebotarev O, Persson H, Simmons CA. Sensitivity and Validation of Porous Membrane Electrical Cell Substrate Impedance Spectroscopy (PM-ECIS) for Measuring Endothelial Barrier Properties. ACS Biomater Sci Eng 2024; 10:5327-5335. [PMID: 38943620 DOI: 10.1021/acsbiomaterials.3c01898] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/01/2024]
Abstract
Measurement of endothelial and epithelial barrier integrity is important for a variety of in vitro models, including Transwell assays, cocultures, and organ-on-chip platforms. Barrier resistance is typically measured by trans-endothelial electrical resistance (TEER), but TEER is invasive and cannot accurately measure isolated monolayer resistance in coculture or most organ-on-chip devices. These limitations are addressed by porous membrane electrical cell-substrate impedance sensing (PM-ECIS), which measures barrier integrity in cell monolayers grown directly on permeable membranes patterned with electrodes. Here, we advanced the design and utility of PM-ECIS by investigating its sensitivity to working electrode size and correlation with TEER. Gold electrodes were fabricated on porous membrane inserts using hot embossing and UV lithography, with working electrode diameters of 250, 500, and 750 μm within the same insert. Sensitivity to resistance changes (4 kHz) during endothelial barrier formation was inversely proportional to electrode size, with the smallest being the most sensitive (p < 0.001). Similarly, smaller electrodes were most sensitive to changes in impedance (40 kHz) corresponding to cell spreading and proliferation (p < 0.001). Barrier disruption with both EGTA and thrombin was detectable by all electrode sizes. Resistances measured by PM-ECIS vs TEER for sodium chloride solutions were positively and significantly correlated for all electrode sizes (r > 0.9; p < 0.0001), but only with 750 μm electrodes for endothelial monolayers (r = 0.71; p = 0.058). These data inform the design and selection of PM-ECIS electrodes for specific applications and support PM-ECIS as a promising alternative to conventional TEER for direct, noninvasive, real-time assessment of cells cultured on porous membranes in conventional and organ-on-chip barrier models.
Collapse
Affiliation(s)
- Alisa Ugodnikov
- Translational Biology & Engineering Program, Ted Rogers Centre for Heart Research, Toronto, ON M5G 1M1, Canada
- Institute of Biomedical Engineering, University of Toronto, Toronto , ON M5S 3G9, Canada
| | - Oleg Chebotarev
- Translational Biology & Engineering Program, Ted Rogers Centre for Heart Research, Toronto, ON M5G 1M1, Canada
| | - Henrik Persson
- Translational Biology & Engineering Program, Ted Rogers Centre for Heart Research, Toronto, ON M5G 1M1, Canada
| | - Craig A Simmons
- Translational Biology & Engineering Program, Ted Rogers Centre for Heart Research, Toronto, ON M5G 1M1, Canada
- Institute of Biomedical Engineering, University of Toronto, Toronto , ON M5S 3G9, Canada
- Department of Mechanical & Industrial Engineering, University of Toronto, Toronto, ON M5S 3G8, Canada
| |
Collapse
|
4
|
Chiu WC, Chen WL, Lai YT, Hung YH, Lo CM. Cell-Electrode Models for Impedance Analysis of Epithelial and Endothelial Monolayers Cultured on Microelectrodes. SENSORS (BASEL, SWITZERLAND) 2024; 24:4214. [PMID: 39000992 PMCID: PMC11244256 DOI: 10.3390/s24134214] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Revised: 06/21/2024] [Accepted: 06/27/2024] [Indexed: 07/16/2024]
Abstract
Electric cell-substrate impedance sensing has been used to measure transepithelial and transendothelial impedances of cultured cell layers and extract cell parameters such as junctional resistance, cell-substrate separation, and membrane capacitance. Previously, a three-path cell-electrode model comprising two transcellular pathways and one paracellular pathway was developed for the impedance analysis of MDCK cells. By ignoring the resistances of the lateral intercellular spaces, we develop a simplified three-path model for the impedance analysis of epithelial cells and solve the model equations in a closed form. The calculated impedance values obtained from this simplified cell-electrode model at frequencies ranging from 31.25 Hz to 100 kHz agree well with the experimental data obtained from MDCK and OVCA429 cells. We also describe how the change in each model-fitting parameter influences the electrical impedance spectra of MDCK cell layers. By assuming that the junctional resistance is much smaller than the specific impedance through the lateral cell membrane, the simplified three-path model reduces to a two-path model, which can be used for the impedance analysis of endothelial cells and other disk-shaped cells with low junctional resistances. The measured impedance spectra of HUVEC and HaCaT cell monolayers nearly coincide with the impedance data calculated from the two-path model.
Collapse
Affiliation(s)
- Wei-Chih Chiu
- Department of Biomedical Engineering, National Yang Ming Chiao Tung University, Taipei 11221, Taiwan; (W.-C.C.); (W.-L.C.)
| | - Wei-Ling Chen
- Department of Biomedical Engineering, National Yang Ming Chiao Tung University, Taipei 11221, Taiwan; (W.-C.C.); (W.-L.C.)
| | - Yi-Ting Lai
- Research Center for Applied Sciences, Academia Sinica, Taipei 11529, Taiwan;
| | - Yu-Han Hung
- Department of Neurology, University of California, Irvine, CA 92697, USA
| | - Chun-Min Lo
- Department of Biomedical Engineering, National Yang Ming Chiao Tung University, Taipei 11221, Taiwan; (W.-C.C.); (W.-L.C.)
| |
Collapse
|
5
|
Ugodnikov A, Persson H, Simmons CA. Bridging barriers: advances and challenges in modeling biological barriers and measuring barrier integrity in organ-on-chip systems. LAB ON A CHIP 2024; 24:3199-3225. [PMID: 38689569 DOI: 10.1039/d3lc01027a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/02/2024]
Abstract
Biological barriers such as the blood-brain barrier, skin, and intestinal mucosal barrier play key roles in homeostasis, disease physiology, and drug delivery - as such, it is important to create representative in vitro models to improve understanding of barrier biology and serve as tools for therapeutic development. Microfluidic cell culture and organ-on-a-chip (OOC) systems enable barrier modelling with greater physiological fidelity than conventional platforms by mimicking key environmental aspects such as fluid shear, accurate microscale dimensions, mechanical cues, extracellular matrix, and geometrically defined co-culture. As the prevalence of barrier-on-chip models increases, so does the importance of tools that can accurately assess barrier integrity and function without disturbing the carefully engineered microenvironment. In this review, we first provide a background on biological barriers and the physiological features that are emulated through in vitro barrier models. Then, we outline molecular permeability and electrical sensing barrier integrity assessment methods, and the related challenges specific to barrier-on-chip implementation. Finally, we discuss future directions in the field, as well important priorities to consider such as fabrication costs, standardization, and bridging gaps between disciplines and stakeholders.
Collapse
Affiliation(s)
- Alisa Ugodnikov
- Translational Biology & Engineering Program, Ted Rogers Centre for Heart Research, Toronto, ON M5G 1M1, Canada.
- Institute of Biomedical Engineering, University of Toronto, Toronto, ON M5S 3G9, Canada
| | - Henrik Persson
- Translational Biology & Engineering Program, Ted Rogers Centre for Heart Research, Toronto, ON M5G 1M1, Canada.
| | - Craig A Simmons
- Translational Biology & Engineering Program, Ted Rogers Centre for Heart Research, Toronto, ON M5G 1M1, Canada.
- Institute of Biomedical Engineering, University of Toronto, Toronto, ON M5S 3G9, Canada
- Department of Mechanical & Industrial Engineering, University of Toronto, Toronto, ON M5S 3G8, Canada
| |
Collapse
|
6
|
Brailoiu E, Barr JL, Wittorf HN, Inan S, Unterwald EM, Brailoiu GC. Modulation of the Blood-Brain Barrier by Sigma-1R Activation. Int J Mol Sci 2024; 25:5147. [PMID: 38791182 PMCID: PMC11121402 DOI: 10.3390/ijms25105147] [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: 03/11/2024] [Revised: 05/05/2024] [Accepted: 05/07/2024] [Indexed: 05/26/2024] Open
Abstract
Sigma non-opioid intracellular receptor 1 (Sigma-1R) is an intracellular chaperone protein residing on the endoplasmic reticulum at the mitochondrial-associated membrane (MAM) region. Sigma-1R is abundant in the brain and is involved in several physiological processes as well as in various disease states. The role of Sigma-1R at the blood-brain barrier (BBB) is incompletely characterized. In this study, the effect of Sigma-1R activation was investigated in vitro on rat brain microvascular endothelial cells (RBMVEC), an important component of the blood-brain barrier (BBB), and in vivo on BBB permeability in rats. The Sigma-1R agonist PRE-084 produced a dose-dependent increase in mitochondrial calcium, and mitochondrial and cytosolic reactive oxygen species (ROS) in RBMVEC. PRE-084 decreased the electrical resistance of the RBMVEC monolayer, measured with the electric cell-substrate impedance sensing (ECIS) method, indicating barrier disruption. These effects were reduced by pretreatment with Sigma-1R antagonists, BD 1047 and NE 100. In vivo assessment of BBB permeability in rats indicates that PRE-084 produced a dose-dependent increase in brain extravasation of Evans Blue and sodium fluorescein brain; the effect was reduced by the Sigma-1R antagonists. Immunocytochemistry studies indicate that PRE-084 produced a disruption of tight and adherens junctions and actin cytoskeleton. The brain microcirculation was directly visualized in vivo in the prefrontal cortex of awake rats with a miniature integrated fluorescence microscope (aka, miniscope; Doric Lenses Inc.). Miniscope studies indicate that PRE-084 increased sodium fluorescein extravasation in vivo. Taken together, these results indicate that Sigma-1R activation promoted oxidative stress and increased BBB permeability.
Collapse
Affiliation(s)
- Eugen Brailoiu
- Center for Substance Abuse Research, Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, USA; (E.B.); (J.L.B.); (S.I.)
- Department of Neural Sciences, Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, USA
| | - Jeffrey L. Barr
- Center for Substance Abuse Research, Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, USA; (E.B.); (J.L.B.); (S.I.)
| | - Hailey N. Wittorf
- Department of Pharmaceutical Sciences, Jefferson College of Pharmacy, Thomas Jefferson University, Philadelphia, PA 19107, USA;
| | - Saadet Inan
- Center for Substance Abuse Research, Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, USA; (E.B.); (J.L.B.); (S.I.)
| | - Ellen M. Unterwald
- Center for Substance Abuse Research, Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, USA; (E.B.); (J.L.B.); (S.I.)
- Department of Neural Sciences, Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, USA
| | - Gabriela Cristina Brailoiu
- Department of Pharmaceutical Sciences, Jefferson College of Pharmacy, Thomas Jefferson University, Philadelphia, PA 19107, USA;
| |
Collapse
|
7
|
Chebotarev O, Ugodnikov A, Simmons CA. Porous Membrane Electrical Cell-Substrate Impedance Spectroscopy for Versatile Assessment of Biological Barriers In Vitro. ACS APPLIED BIO MATERIALS 2024; 7:2000-2011. [PMID: 38447196 DOI: 10.1021/acsabm.4c00114] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/08/2024]
Abstract
Cell culture models of endothelial and epithelial barriers typically use porous membrane inserts (e.g., Transwell inserts) as a permeable substrate on which barrier cells are grown, often in coculture with other cell types on the opposite side of the membrane. Current methods to characterize barrier function in porous membrane inserts can disrupt the barrier or provide bulk measurements that cannot isolate barrier cell resistance alone. Electrical cell-substrate impedance sensing (ECIS) addresses these limitations, but its implementation on porous membrane inserts has been limited by costly manufacturing, low sensitivity, and lack of validation for barrier assessment. Here, we present porous membrane ECIS (PM-ECIS), a cost-effective method to adapt ECIS technology to porous substrate-based in vitro models. We demonstrate high fidelity patterning of electrodes on porous membranes that can be incorporated into well plates of a variety of sizes with excellent cell biocompatibility with mono- and coculture set ups. PM-ECIS provided sensitive, real-time measurement of isolated changes in endothelial cell barrier impedance with cell growth and barrier disruption. Barrier function characterized by PM-ECIS resistance correlated well with permeability coefficients obtained from simultaneous molecular tracer permeability assays performed on the same cultures, validating the device. Integration of ECIS into conventional porous cell culture inserts provides a versatile, sensitive, and automated alternative to current methods to measure barrier function in vitro, including molecular tracer assays and transepithelial/endothelial electrical resistance.
Collapse
Affiliation(s)
- Oleg Chebotarev
- Translational Biology & Engineering Program, Ted Rogers Centre for Heart Research, Toronto, ON M5G 1M1, Canada
- Department of Mechanical & Industrial Engineering, University of Toronto, Toronto, ON M5S 3G8, Canada
| | - Alisa Ugodnikov
- Translational Biology & Engineering Program, Ted Rogers Centre for Heart Research, Toronto, ON M5G 1M1, Canada
- Institute of Biomedical Engineering, University of Toronto, Toronto, ON M5S 3G9, Canada
| | - Craig A Simmons
- Translational Biology & Engineering Program, Ted Rogers Centre for Heart Research, Toronto, ON M5G 1M1, Canada
- Department of Mechanical & Industrial Engineering, University of Toronto, Toronto, ON M5S 3G8, Canada
- Institute of Biomedical Engineering, University of Toronto, Toronto, ON M5S 3G9, Canada
| |
Collapse
|
8
|
Chung DD, Chen AC, Choo CH, Zhang W, Williams D, Griffis CG, Bonezzi P, Jatavallabhula K, Sampath AP, Aldave AJ. Investigation of the functional impact of CHED- and FECD4-associated SLC4A11 mutations in human corneal endothelial cells. PLoS One 2024; 19:e0296928. [PMID: 38252645 PMCID: PMC10802951 DOI: 10.1371/journal.pone.0296928] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Accepted: 12/22/2023] [Indexed: 01/24/2024] Open
Abstract
Mutations in the solute linked carrier family 4 member 11 (SLC4A11) gene are associated with congenital hereditary endothelial dystrophy (CHED) and Fuchs corneal endothelial dystrophy type 4 (FECD4), both characterized by corneal endothelial cell (CEnC) dysfunction and/or cell loss leading to corneal edema and visual impairment. In this study, we characterize the impact of CHED-/FECD4-associated SLC4A11 mutations on CEnC function and SLC4A11 protein localization by generating and comparing human CEnC (hCEnC) lines expressing wild type SLC4A11 (SLC4A11WT) or mutant SLC4A11 harboring CHED-/FECD4-associated SLC4A11 mutations (SLC4A11MU). SLC4A11WT and SLC4A11MU hCEnC lines were generated to express either SLC4A11 variant 2 (V2WT and V2MU) or variant 3 (V3WT and V3MU), the two major variants expressed in ex vivo hCEnC. Functional assays were performed to assess cell barrier, proliferation, viability, migration, and NH3-induced membrane conductance. We demonstrate SLC4A11-/- and SLC4A11MU hCEnC lines exhibited increased migration rates, altered proliferation and decreased cell viability compared to SLC4A11WT hCEnC. Additionally, SLC4A11-/- hCEnC demonstrated decreased cell-substrate adhesion and membrane capacitances compared to SLC4A11WT hCEnC. Induction with 10mM NH4Cl led SLC4A11WT hCEnC to depolarize; conversely, SLC4A11-/- hCEnC hyperpolarized and the majority of SLC4A11MU hCEnC either hyperpolarized or had minimal membrane potential changes following NH4Cl induction. Immunostaining of primary hCEnC and SLC4A11WT hCEnC lines for SLC4A11 demonstrated predominately plasma membrane staining with poor or partial colocalization with mitochondrial marker COX4 within a subset of punctate subcellular structures. Overall, our findings suggest CHED-associated SLC4A11 mutations likely lead to hCEnC dysfunction, and ultimately CHED, by interfering with cell migration, proliferation, viability, membrane conductance, barrier function, and/or cell surface localization of the SLC4A11 protein in hCEnC. Additionally, based on their similar subcellular localization and exhibiting similar cell functional profiles, protein isoforms encoded by SLC4A11 variant 2 and variant 3 likely have highly overlapping functional roles in hCEnC.
Collapse
Affiliation(s)
- Doug D. Chung
- Department of Ophthalmology, Stein Eye Institute at UCLA, Los Angeles, California, United States of America
| | - Angela C. Chen
- Department of Ophthalmology, Stein Eye Institute at UCLA, Los Angeles, California, United States of America
| | - Charlene H. Choo
- Department of Ophthalmology, Stein Eye Institute at UCLA, Los Angeles, California, United States of America
| | - Wenlin Zhang
- Department of Ophthalmology, Stein Eye Institute at UCLA, Los Angeles, California, United States of America
| | - Dominic Williams
- Department of Ophthalmology, Stein Eye Institute at UCLA, Los Angeles, California, United States of America
| | - Christopher G. Griffis
- Department of Ophthalmology, Stein Eye Institute at UCLA, Los Angeles, California, United States of America
| | - Paul Bonezzi
- Department of Ophthalmology, Stein Eye Institute at UCLA, Los Angeles, California, United States of America
| | - Kavya Jatavallabhula
- Department of Ophthalmology, Stein Eye Institute at UCLA, Los Angeles, California, United States of America
| | - Alapakkam P. Sampath
- Department of Ophthalmology, Stein Eye Institute at UCLA, Los Angeles, California, United States of America
| | - Anthony J. Aldave
- Department of Ophthalmology, Stein Eye Institute at UCLA, Los Angeles, California, United States of America
| |
Collapse
|
9
|
Arman S, Tilley RD, Gooding JJ. A review of electrochemical impedance as a tool for examining cell biology and subcellular mechanisms: merits, limits, and future prospects. Analyst 2024; 149:269-289. [PMID: 38015145 DOI: 10.1039/d3an01423a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2023]
Abstract
Herein the development of cellular impedance biosensors, electrochemical impedance spectroscopy, and the general principles and terms associated with the cell-electrode interface is reviewed. This family of techniques provides quantitative and sensitive information into cell responses to stimuli in real-time with high temporal resolution. The applications of cell-based impedance biosensors as a readout in cell biology is illustrated with a diverse range of examples. The current state of the field, its limitations, the possible available solutions, and the potential benefits of developing biosensors are discussed.
Collapse
Affiliation(s)
- Seyedyousef Arman
- School of Chemistry, The University of New South Wales, Sydney, New South Wales 2052, Australia.
- Australia Centre for Nanomedicine, The University of New South Wales, Sydney, New South Wales 2052, Australia
| | - Richard D Tilley
- School of Chemistry, The University of New South Wales, Sydney, New South Wales 2052, Australia.
- Electron Microscope Unit, Mark Wainwright Analytical Centre, The University of New South Wales, Sydney, New South Wales 2052, Australia
| | - J Justin Gooding
- School of Chemistry, The University of New South Wales, Sydney, New South Wales 2052, Australia.
- Australia Centre for Nanomedicine, The University of New South Wales, Sydney, New South Wales 2052, Australia
| |
Collapse
|
10
|
Moztarzadeh S, Sepic S, Hamad I, Waschke J, Radeva MY, García-Ponce A. Cortactin is in a complex with VE-cadherin and is required for endothelial adherens junction stability through Rap1/Rac1 activation. Sci Rep 2024; 14:1218. [PMID: 38216638 PMCID: PMC10786853 DOI: 10.1038/s41598-024-51269-3] [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: 06/30/2023] [Accepted: 01/03/2024] [Indexed: 01/14/2024] Open
Abstract
Vascular permeability is mediated by Cortactin (Cttn) and regulated by several molecules including cyclic-adenosine-monophosphate, small Rho family GTPases and the actin cytoskeleton. However, it is unclear whether Cttn directly interacts with any of the junctional components or if Cttn intervenes with signaling pathways affecting the intercellular contacts and the cytoskeleton. To address these questions, we employed immortalized microvascular myocardial endothelial cells derived from wild-type and Cttn-knock-out mice. We found that lack of Cttn compromised barrier integrity due to fragmented membrane distribution of different junctional proteins. Moreover, immunoprecipitations revealed that Cttn is within the VE-cadherin-based adherens junction complex. In addition, lack of Cttn slowed-down barrier recovery after Ca2+ repletion. The role of Cttn for cAMP-mediated endothelial barrier regulation was analyzed using Forskolin/Rolipram. In contrast to Cttn-KO, WT cells reacted with increased transendothelial electrical resistance. Absence of Cttn disturbed Rap1 and Rac1 activation in Cttn-depleted cells. Surprisingly, despite the absence of Cttn, direct activation of Rac1/Cdc42/RhoA by CN04 increased barrier resistance and induced well-defined cortical actin and intracellular actin bundles. In summary, our data show that Cttn is required for basal barrier integrity by allowing proper membrane distribution of junctional proteins and for cAMP-mediated activation of the Rap1/Rac1 signaling pathway.
Collapse
Affiliation(s)
- Sina Moztarzadeh
- Chair of Vegetative Anatomy, Faculty of Medicine, Ludwig-Maximilians-University (LMU) Munich, Pettenkoferstraße 11, 80336, Munich, Germany
| | - Sara Sepic
- Chair of Vegetative Anatomy, Faculty of Medicine, Ludwig-Maximilians-University (LMU) Munich, Pettenkoferstraße 11, 80336, Munich, Germany
| | - Ibrahim Hamad
- Chair of Vegetative Anatomy, Faculty of Medicine, Ludwig-Maximilians-University (LMU) Munich, Pettenkoferstraße 11, 80336, Munich, Germany
| | - Jens Waschke
- Chair of Vegetative Anatomy, Faculty of Medicine, Ludwig-Maximilians-University (LMU) Munich, Pettenkoferstraße 11, 80336, Munich, Germany
| | - Mariya Y Radeva
- Chair of Vegetative Anatomy, Faculty of Medicine, Ludwig-Maximilians-University (LMU) Munich, Pettenkoferstraße 11, 80336, Munich, Germany
| | - Alexander García-Ponce
- Chair of Vegetative Anatomy, Faculty of Medicine, Ludwig-Maximilians-University (LMU) Munich, Pettenkoferstraße 11, 80336, Munich, Germany.
| |
Collapse
|
11
|
Choi JS, Doo HM, Kim B, Lee SH, Sung S, Go G, Suarez A, Kim Y, Weon BM, Choi B, Kim HJ, Kim D. NanoIEA: A Nanopatterned Interdigitated Electrode Array-Based Impedance Assay for Real-Time Measurement of Aligned Endothelial Cell Barrier Functions. Adv Healthc Mater 2024; 13:e2301124. [PMID: 37820720 PMCID: PMC10841753 DOI: 10.1002/adhm.202301124] [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: 07/04/2023] [Revised: 09/18/2023] [Indexed: 10/13/2023]
Abstract
A nanopatterned interdigitated electrode array (nanoIEA)-based impedance assay is developed for quantitative real-time measurement of aligned endothelial cell (EC) barrier functions in vitro. A bioinspired poly(3,4-dihydroxy-L-phenylalanine) (poly (l-DOPA)) coating is applied to improve the human brain EC adhesion onto the Nafion nanopatterned surfaces. It is found that a poly (l-DOPA)-coated Nafion grooved nanopattern makes the human brain ECs orient along the nanopattern direction. Aligned human brain ECs on Nafion nanopatterns exhibit increased expression of genes encoding tight and adherens junction proteins. Aligned human brain ECs also have enhanced impedance and resistance versus unaligned ones. Treatment with a glycogen synthase kinase-3 inhibitor (GSK3i) further increases impedance and resistance, suggesting synergistic effects occur on the cell-cell tightness of in vitro human brain ECs via a combination of anisotropic matrix nanotopography and GSK3i treatment. It is found that this enhanced cell-cell tightness of the combined approach is accompanied by increased expression of claudin protein. These data demonstrate that the proposed nanoIEA assay integrated with poly (l-DOPA)-coated Nafion nanopatterns and interdigitated electrode arrays can make not only biomimetic aligned ECs, but also enable real-time measurement of the enhanced barrier functions of aligned ECs via tighter cell-cell junctions.
Collapse
Affiliation(s)
- Jong Seob Choi
- Department of Biomedical Engineering, Center for Microphysiological SystemsJohns Hopkins UniversityBaltimoreMD21205USA
- Division of Advanced Materials EngineeringKongju National UniversityCheonanChungnam31080South Korea
| | - Hyun Myung Doo
- Department of Health Sciences and TechnologySAIHSTSungkyunkwan UniversitySeoul06351South Korea
- Department of Biomedical Research CenterKorea University Guro HospitalSeoul08308South Korea
- Division of Medical Oncology, Department of Internal MedicineKorea University Guro Hospital, Korea University College of MedicineSeoul08308South Korea
| | - Byunggik Kim
- Department of Mechanical EngineeringJohns Hopkins UniversityBaltimoreMD21218USA
| | - Su Han Lee
- Digital Health Care Research CenterGumi Electronics and Information Technology Research Institute (GERI)GumiGyeongbuk39253South Korea
| | - Sang‐keun Sung
- Digital Health Care Research CenterGumi Electronics and Information Technology Research Institute (GERI)GumiGyeongbuk39253South Korea
| | - Gwangjun Go
- Department of Biomedical Engineering, Center for Microphysiological SystemsJohns Hopkins UniversityBaltimoreMD21205USA
- Department of Mechanical EngineeringChosun UniversityGwangju61452South Korea
| | - Allister Suarez
- Department of Biomedical Engineering, Center for Microphysiological SystemsJohns Hopkins UniversityBaltimoreMD21205USA
| | - Yeseul Kim
- SKKU Advanced Institute of Nanotechnology (SAINT)School of Advanced Materials Science and EngineeringSungkyunkwan UniversitySuwon16419South Korea
| | - Byung Mook Weon
- SKKU Advanced Institute of Nanotechnology (SAINT)School of Advanced Materials Science and EngineeringSungkyunkwan UniversitySuwon16419South Korea
| | - Byung‐Ok Choi
- Department of Health Sciences and TechnologySAIHSTSungkyunkwan UniversitySeoul06351South Korea
- Department of NeurologySamsung Medical CenterSungkyunkwan University School of MedicineSeoul06351South Korea
| | - Hyung Jin Kim
- School of Electrical and Electronic EngineeringUlsan CollegeUlsan44610South Korea
| | - Deok‐Ho Kim
- Department of Biomedical Engineering, Center for Microphysiological SystemsJohns Hopkins UniversityBaltimoreMD21205USA
- Department of Mechanical EngineeringJohns Hopkins UniversityBaltimoreMD21218USA
- Department of MedicineJohns Hopkins University School of MedicineBaltimoreMD21205USA
- Institute for NanobiotechnologyJohns Hopkins UniversityBaltimoreMD21218USA
| |
Collapse
|
12
|
Huang X, Liang F, Huang B, Luo H, Shi J, Wang L, Peng J, Chen Y. On-chip real-time impedance monitoring of hiPSC-derived and artificial basement membrane-supported endothelium. Biosens Bioelectron 2023; 235:115324. [PMID: 37201240 DOI: 10.1016/j.bios.2023.115324] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2023] [Revised: 04/11/2023] [Accepted: 04/14/2023] [Indexed: 05/20/2023]
Abstract
Recent advances have shown the high sensibility of electrochemical impedance spectroscopy in real-time monitoring of cell barriers on a chip. Here, we applied this method to the investigation of human induced pluripotent stem cell (hiPSC) derived and artificial basement membrane (ABM) supported endothelial barrier. The ABM was obtained by self-assembling type IV collagen and laminin with a monolayer of crosslinked gelatin nanofibers. The hiPSCs were differentiated into brain microvascular endothelial cells (BMECs) and then plated on the ABM. After incubation for two days, the ABM-BMEC assembly was placed as a tissue insert into a microfluidic device for culture and real-time impedance monitoring over days. We found a significantly enhanced stability of the BMEC barrier in a serum-free and bromodeoxyuridine (BrdU) containing culture medium compared to the conventional culture due to the restricted cell proliferation. We also found that the BMEC barrier was sensitive to stimuli such as thrombin and that the change of the barrier impedance was mainly due to the change of the cell layer resistance. We can thus advocate this method to investigate the integrity of the cell barrier and the barrier-based assays.
Collapse
Affiliation(s)
- Xiaochen Huang
- École Normale Supérieure-PSL Research University, Département de Chimie, Sorbonne Universités-UPMC Univ Paris 06, CNRS UMR 8640, PASTEUR, 24, Rue Lhomond, 75005, Paris, France
| | - Feng Liang
- École Normale Supérieure-PSL Research University, Département de Chimie, Sorbonne Universités-UPMC Univ Paris 06, CNRS UMR 8640, PASTEUR, 24, Rue Lhomond, 75005, Paris, France
| | - Boxin Huang
- École Normale Supérieure-PSL Research University, Département de Chimie, Sorbonne Universités-UPMC Univ Paris 06, CNRS UMR 8640, PASTEUR, 24, Rue Lhomond, 75005, Paris, France
| | - Haoyue Luo
- École Normale Supérieure-PSL Research University, Département de Chimie, Sorbonne Universités-UPMC Univ Paris 06, CNRS UMR 8640, PASTEUR, 24, Rue Lhomond, 75005, Paris, France
| | - Jian Shi
- MesoBioTech, 231 Rue Saint-Honoré, 75001, Paris, France
| | - Li Wang
- MesoBioTech, 231 Rue Saint-Honoré, 75001, Paris, France
| | - Juan Peng
- École Normale Supérieure-PSL Research University, Département de Chimie, Sorbonne Universités-UPMC Univ Paris 06, CNRS UMR 8640, PASTEUR, 24, Rue Lhomond, 75005, Paris, France.
| | - Yong Chen
- École Normale Supérieure-PSL Research University, Département de Chimie, Sorbonne Universités-UPMC Univ Paris 06, CNRS UMR 8640, PASTEUR, 24, Rue Lhomond, 75005, Paris, France.
| |
Collapse
|
13
|
Koda A, Ishii Y, Kashiwagi A, Fujikawa M, Kikuchi K, Hashimoto R, Ueda Y, Doi T. The Effects of Mucopolysaccharide Polysulfate on Steroid-Induced Tight Junction Barrier Dysfunction in Human Epidermal Keratinocytes and a 3D Skin Model. Skin Pharmacol Physiol 2023; 36:186-194. [PMID: 36966539 PMCID: PMC10652656 DOI: 10.1159/000529962] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Accepted: 02/24/2023] [Indexed: 11/15/2023]
Abstract
INTRODUCTION The long-term use of topical corticosteroids (TCS) is associated with side effects such as skin atrophy and barrier deterioration. Moisturizers, such as mucopolysaccharide polysulfate (MPS), have been reported to prevent relapses in atopic dermatitis (AD) when used in combination with TCS. However, the mechanisms underlying the positive effects of MPS in combination with TCS in AD are poorly understood. In the present study, we investigated the effects of MPS in combination with clobetasol 17-propionate (CP) on tight junction (TJ) barrier function in human epidermal keratinocytes (HEKa) and 3D skin models. METHODS The expression of claudin-1, which is crucial for TJ barrier function in keratinocytes, and transepithelial electrical resistance (TEER) was measured in CP-treated human keratinocytes incubated with and without MPS. A TJ permeability assay, using Sulfo-NHS-Biotin as a tracer, was also conducted in a 3D skin model. RESULTS CP reduced claudin-1 expression and TEER in human keratinocytes, whereas MPS inhibited these CP-induced effects. Moreover, MPS inhibited the increase in CP-induced TJ permeability in a 3D skin model. CONCLUSION The present study demonstrated that MPS improved TJ barrier impairment induced by CP. The improvement of TJ barrier function may partially be responsible for the delayed relapse of AD induced by the combination of MPS and TCS.
Collapse
Affiliation(s)
- Akira Koda
- Drug Development Laboratories, Kyoto R&D Center, Drug Development Research Laboratories, Maruho Co., Ltd., Kyoto, Japan
| | - Yuko Ishii
- Drug Development Laboratories, Kyoto R&D Center, Drug Development Research Laboratories, Maruho Co., Ltd., Kyoto, Japan
| | - Ayu Kashiwagi
- Drug Development Laboratories, Kyoto R&D Center, Drug Development Research Laboratories, Maruho Co., Ltd., Kyoto, Japan
| | - Mika Fujikawa
- Drug Development Laboratories, Kyoto R&D Center, Drug Development Research Laboratories, Maruho Co., Ltd., Kyoto, Japan
| | - Keisuke Kikuchi
- Drug Development Laboratories, Kyoto R&D Center, Drug Development Research Laboratories, Maruho Co., Ltd., Kyoto, Japan
| | - Ryota Hashimoto
- Drug Development Laboratories, Kyoto R&D Center, Drug Development Research Laboratories, Maruho Co., Ltd., Kyoto, Japan
| | - Yuhki Ueda
- Drug Development Laboratories, Kyoto R&D Center, Drug Development Research Laboratories, Maruho Co., Ltd., Kyoto, Japan
| | - Takaaki Doi
- Drug Development Laboratories, Kyoto R&D Center, Drug Development Research Laboratories, Maruho Co., Ltd., Kyoto, Japan
| |
Collapse
|
14
|
Curtis TM, Nilon AM, Greenberg AJ, Besner M, Scibek JJ, Nichols JA, Huie JL. Odorant Binding Causes Cytoskeletal Rearrangement, Leading to Detectable Changes in Endothelial and Epithelial Barrier Function and Micromotion. BIOSENSORS 2023; 13:329. [PMID: 36979541 PMCID: PMC10046532 DOI: 10.3390/bios13030329] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Revised: 02/23/2023] [Accepted: 02/27/2023] [Indexed: 06/18/2023]
Abstract
Non-olfactory cells have excellent biosensor potential because they express functional olfactory receptors (ORs) and are non-neuronal cells that are easy to culture. ORs are G-protein coupled receptors (GPCRs), and there is a well-established link between different classes of G-proteins and cytoskeletal structure changes affecting cellular morphology that has been unexplored for odorant sensing. Thus, the present study was conducted to determine if odorant binding in non-olfactory cells causes cytoskeletal changes that will lead to cell changes detectable by electric cell-substrate impedance sensing (ECIS). To this end, we used the human umbilical vein endothelial cells (HUVECs), which express OR10J5, and the human keratinocyte (HaCaT) cells, which express OR2AT4. Using these two different cell barriers, we showed that odorant addition, lyral and Sandalore, respectively, caused an increase in cAMP, changes in the organization of the cytoskeleton, and a decrease in the integrity of the junctions between the cells, causing a decrease in cellular electrical resistance. In addition, the random cellular movement of the monolayers (micromotion) was significantly decreased after odorant exposure. Collectively, these data demonstrate a new physiological role of olfactory receptor signaling in endothelial and epithelial cell barriers and represent a new label-free method to detect odorant binding.
Collapse
Affiliation(s)
- Theresa M. Curtis
- Department of Biological Sciences, SUNY Cortland, Cortland, NY 13045, USA
| | - Annabella M. Nilon
- Department of Biological Sciences, SUNY Cortland, Cortland, NY 13045, USA
| | | | - Matthew Besner
- Department of Biological Sciences, SUNY Cortland, Cortland, NY 13045, USA
| | - Jacob J. Scibek
- Department of Biological Sciences, SUNY Cortland, Cortland, NY 13045, USA
| | | | | |
Collapse
|
15
|
Combining Electrostimulation with Impedance Sensing to Promote and Track Osteogenesis within a Titanium Implant. Biomedicines 2023; 11:biomedicines11030697. [PMID: 36979676 PMCID: PMC10045247 DOI: 10.3390/biomedicines11030697] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Revised: 02/03/2023] [Accepted: 02/17/2023] [Indexed: 03/02/2023] Open
Abstract
(1) Background: Electrical stimulation is a promising alternative to promote bone fracture healing but with the limitation of tracking the osteogenesis progress in vivo. To overcome this issue, we present an opportunity to combine the electrical stimulation of a commercial titanium implant, which promotes osteogenesis within the fracture, with a real-time readout of the osteogenic progress by impedance sensing. This makes it possible to adjust the electrical stimulation modalities to the individual patient’s fracture healing process. (2) Methods: In detail, osteogenic differentiation of several cell types was monitored under continuous or pulsatile electrical stimulation at 0.7 V AC/20 Hz for at least seven days on a titanium implant by electric cell-substrate impedance sensing (ECIS). For control, chemical induction of osteogenic differentiation was induced. (3) Results: The most significant challenge was to discriminate impedance changes caused by proliferation events from those initiated by osteogenic differentiation. This discrimination was achieved by remodeling the impedance parameter Alpha (α), which increases over time for pulsatile electrically stimulated stem cells. Boosted α-values were accompanied by an increased formation of actin stress fibers and a reduced expression of the focal adhesion kinase in the cell periphery; morphological alterations known to occur during osteogenesis. (4) Conclusions: This work provided the basis for developing an effective fracture therapy device, which can induce osteogenesis on the one hand, and would allow us to monitor the induction process on the other hand.
Collapse
|
16
|
Miller JL, Kanke M, Rauner G, Bakhle KM, Sethupathy P, Van de Walle GR. Comparative Analysis of microRNAs that Stratify in vitro Mammary stem and Progenitor Activity Reveals Functionality of Human miR-92b-3p. J Mammary Gland Biol Neoplasia 2022; 27:253-269. [PMID: 36190643 DOI: 10.1007/s10911-022-09525-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Accepted: 09/07/2022] [Indexed: 01/19/2023] Open
Abstract
Mammary stem/progenitor cells are fundamental for mammary gland development and function. However, much remains to be elucidated regarding their function in mammals beyond the traditionally studied rodents, human, and to a lesser extent, ruminants. Due to the growing appreciation for microRNAs (miRNAs) as regulators of stem cells and their progenitors, we compared miRNA expression in mammary stem/progenitor cells from mammals with varying mammary stem/progenitor activity in vitro, in order to identify miRNA candidates that regulate stem/progenitor self-renewal and function. Mammosphere-derived epithelial cells (MDECs), which are primary cell lines enriched in mammary stem and progenitor cells, were generated from six mammalian species (i.e., cow, human, pig, horse, dog, and rat) and small RNA sequencing was performed. We identified 9 miRNAs that were significantly differentially expressed in MDEC cultures with a low versus high mammary stem/progenitor activity. miR-92b-3p was selected for functional follow-up studies, as this miRNA is understudied in primary mammary cells but has well-described gene targets that are known to regulate mammary stem/progenitor activity. Altering the expression of miR-92b-3p in MDECs from species with low stem/progenitor activity (human and cow) and those with high stem/progenitor activity (dog and rat) via inhibition and overexpression, respectively, resulted in significantly decreased mammosphere formation of human MDECs, but showed no significant effects in cow, dog, or rat MDECs. This study is the first to perform small RNA sequencing in MDECs from various mammals and highlights that conserved miRNAs can have different functions in mammary stem/progenitor cells across species.
Collapse
Affiliation(s)
- James L Miller
- Baker Institute for Animal Health, Ithaca , United States
| | - Matt Kanke
- Department of Biomedical Sciences, College of Veterinary Medicine, Cornell University, Ithaca, NY, USA
| | - Gat Rauner
- Baker Institute for Animal Health, Ithaca , United States
| | | | - Praveen Sethupathy
- Department of Biomedical Sciences, College of Veterinary Medicine, Cornell University, Ithaca, NY, USA
| | - Gerlinde R Van de Walle
- Baker Institute for Animal Health, Ithaca , United States.
- Baker Institute for Animal Health, College of Veterinary Medicine, Cornell University, 14850, Ithaca, NY, USA.
| |
Collapse
|
17
|
Ebrahim AS, Ebrahim T, Kani H, Ibrahim AS, Carion TW, Berger EA. Functional optimization of electric cell-substrate impedance sensing (ECIS) using human corneal epithelial cells. Sci Rep 2022; 12:14126. [PMID: 35986158 PMCID: PMC9391335 DOI: 10.1038/s41598-022-18182-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Accepted: 08/08/2022] [Indexed: 12/26/2022] Open
Abstract
An intact epithelium is key to maintaining corneal integrity and barrier function which can lead to impaired ocular defense and sight-threatening opacity when compromised. Electrical cell-substrate impedance sensing or ECIS is a non-invasive method to measure real-time cellular behaviors including barrier function and cell migration. The current study uses ECIS technology to assess and optimize human telomerase-immortalized corneal epithelial cells to generate quantifiable measurements that accurately reflect changes in cell behavior in vitro. Five cell densities were assessed in two different media to determine the optimal conditions for monitoring of cellular behavior over time. Parameters of evaluation included: overall impedance (Z), barrier resistance (R), cell capacitance (C), and mathematical modeling of the R data to further generate Rb (the electrical resistance between HUCLs), α (the resistance between the HUCLs and the substrate), and Cm (the capacitance of the cell membrane) measurements. All parameters of assessment strongly indicated DMEM/F12 at 60,000 cells as the optimal condition for ECIS assessment of HUCLs. Furthermore, this work highlights the ability of the sensitive ECIS biosensor technology to comprehensively and quantitatively assess corneal epithelial cell structure and function and the importance of optimizing not only cell density, but choice of media used for in vitro culturing.
Collapse
|
18
|
Mekala N, Gheewala N, Rom S, Sriram U, Persidsky Y. Blocking of P2X7r Reduces Mitochondrial Stress Induced by Alcohol and Electronic Cigarette Exposure in Brain Microvascular Endothelial Cells. Antioxidants (Basel) 2022; 11:1328. [PMID: 35883819 PMCID: PMC9311929 DOI: 10.3390/antiox11071328] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Revised: 06/27/2022] [Accepted: 07/03/2022] [Indexed: 12/15/2022] Open
Abstract
Studies in both humans and animal models demonstrated that chronic alcohol/e-cigarette (e-Cig) exposure affects mitochondrial function and impairs barrier function in brain microvascular endothelial cells (BMVECs). Identification of the signaling pathways by which chronic alcohol/e-Cig exposure induces mitochondrial damage in BMVEC is vital for protection of the blood-brain barrier (BBB). To address the issue, we treated human BMVEC [hBMVECs (D3 cell-line)] with ethanol (ETH) [100 mM], acetaldehyde (ALD) [100 μM], or e-cigarette (e-Cig) [35 ng/mL of 1.8% or 0% nicotine] conditioned medium and showed reduced mitochondrial oxidative phosphorylation (OXPHOS) measured by a Seahorse analyzer. Seahorse data were further complemented with the expression of mitochondrial OXPHOS proteins detected by Western blots. We also observed cytosolic escape of ATP and its extracellular release due to the disruption of mitochondrial membrane potential caused by ETH, ALD, or 1.8% e-Cig exposure. Moreover ETH, ALD, or 1.8% e-Cig treatment resulted in elevated purinergic P2X7r and TRPV1 channel gene expression, measured using qPCR. We also demonstrated the protective role of P2X7r antagonist A804598 (10 μM) in restoring mitochondrial oxidative phosphorylation levels and preventing extracellular ATP release. In a BBB functional assay using trans-endothelial electrical resistance, we showed that blocking the P2X7r channel enhanced barrier function. In summary, we identified the potential common pathways of mitochondrial injury caused by ETH, ALD, and 1.8% e-Cig which allow new protective interventions. We are further investigating the potential link between P2X7 regulatory pathways and mitochondrial health.
Collapse
Affiliation(s)
| | | | | | | | - Yuri Persidsky
- Department of Pathology and Laboratory Medicine, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA; (N.M.); (N.G.); (S.R.); (U.S.)
| |
Collapse
|
19
|
Yamaguchi E, Yao J, Aymond A, Chrisey DB, Nieman GF, Bates JHT, Gaver DP. Electric Cell-Substrate Impedance Sensing (ECIS) as a Platform for Evaluating Barrier-Function Susceptibility and Damage from Pulmonary Atelectrauma. BIOSENSORS 2022; 12:390. [PMID: 35735538 PMCID: PMC9221382 DOI: 10.3390/bios12060390] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Revised: 05/27/2022] [Accepted: 06/02/2022] [Indexed: 11/16/2022]
Abstract
Biophysical insults that either reduce barrier function (COVID-19, smoke inhalation, aspiration, and inflammation) or increase mechanical stress (surfactant dysfunction) make the lung more susceptible to atelectrauma. We investigate the susceptibility and time-dependent disruption of barrier function associated with pulmonary atelectrauma of epithelial cells that occurs in acute respiratory distress syndrome (ARDS) and ventilator-induced lung injury (VILI). This in vitro study was performed using Electric Cell-substrate Impedance Sensing (ECIS) as a noninvasive evaluating technique for repetitive stress stimulus/response on monolayers of the human lung epithelial cell line NCI-H441. Atelectrauma was mimicked through recruitment/derecruitment (RD) of a semi-infinite air bubble to the fluid-occluded micro-channel. We show that a confluent monolayer with a high level of barrier function is nearly impervious to atelectrauma for hundreds of RD events. Nevertheless, barrier function is eventually diminished, and after a critical number of RD insults, the monolayer disintegrates exponentially. Confluent layers with lower initial barrier function are less resilient. These results indicate that the first line of defense from atelectrauma resides with intercellular binding. After disruption, the epithelial layer community protection is diminished and atelectrauma ensues. ECIS may provide a platform for identifying damaging stimuli, ventilation scenarios, or pharmaceuticals that can reduce susceptibility or enhance barrier-function recovery.
Collapse
Affiliation(s)
- Eiichiro Yamaguchi
- Department of Biomedical Engineering, Tulane University, New Orleans, LA 70118, USA; (J.Y.); (A.A.)
| | - Joshua Yao
- Department of Biomedical Engineering, Tulane University, New Orleans, LA 70118, USA; (J.Y.); (A.A.)
| | - Allison Aymond
- Department of Biomedical Engineering, Tulane University, New Orleans, LA 70118, USA; (J.Y.); (A.A.)
| | - Douglas B. Chrisey
- Department of Physics and Engineering Physics, Tulane University, New Orleans, LA 70118, USA;
| | - Gary F. Nieman
- Department of Surgery, Upstate Medical University, Syracuse, NY 13210, USA;
| | - Jason H. T. Bates
- Department of Medicine, University of Vermont, Burlington, VT 05405, USA;
| | - Donald P. Gaver
- Department of Biomedical Engineering, Tulane University, New Orleans, LA 70118, USA; (J.Y.); (A.A.)
| |
Collapse
|
20
|
Hung YH, Chiu WC, Fuh SR, Lai YT, Tung TH, Huang CC, Lo CM. ECIS Based Electric Fence Method for Measurement of Human Keratinocyte Migration on Different Substrates. BIOSENSORS 2022; 12:bios12050293. [PMID: 35624596 PMCID: PMC9138674 DOI: 10.3390/bios12050293] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Revised: 04/28/2022] [Accepted: 05/01/2022] [Indexed: 05/28/2023]
Abstract
Electric Cell-substrate Impedance Sensing (ECIS) is an impedance-based, real-time, and label-free measuring system for monitoring cellular activities in tissue culture. Previously, ECIS wound healing assay has been used to wound cells with high electric current and monitor the subsequent cell migration. In this study, we applied ECIS electric fence (EF) method, an alternative to electrical wounding, to assess the effects of different surface coatings on human keratinocyte (HaCaT) migration. The EF prevents inoculated cells from attaching or migrating to the fenced electrode surface while maintaining the integrity of the surface coating. After the EF is turned off, cells migrate into the cell-free area, and the increase in measured impedance is monitored. We cultured HaCaT cells on gold electrodes without coating or coated with poly-L-lysin (PLL), poly-D-lysine (PDL), or type-I collagen. We quantified migration rates according to the different slopes in the impedance time series. It was observed that either poly-L-lysine (PLL) or poly-D-lysine (PDL) limits cell adhesion and migration rates. Furthermore, the surface charge of the coated substrate in the culture condition positively correlates with the cell adhesion and migration process. Our results indicate that the EF method is useful for determining cell migration rates on specific surface coatings.
Collapse
Affiliation(s)
- Yu-Han Hung
- Department of Biomedical Engineering, National Yang Ming Chiao Tung University, Taipei 112, Taiwan; (Y.-H.H.); (W.-C.C.); (S.-R.F.); (Y.-T.L.); (T.-H.T.)
| | - Wei-Chih Chiu
- Department of Biomedical Engineering, National Yang Ming Chiao Tung University, Taipei 112, Taiwan; (Y.-H.H.); (W.-C.C.); (S.-R.F.); (Y.-T.L.); (T.-H.T.)
| | - Shyh-Rong Fuh
- Department of Biomedical Engineering, National Yang Ming Chiao Tung University, Taipei 112, Taiwan; (Y.-H.H.); (W.-C.C.); (S.-R.F.); (Y.-T.L.); (T.-H.T.)
- Department of Aesthetic Medicine, Chen Hsin General Hospital, Taipei 112, Taiwan
| | - Yi-Ting Lai
- Department of Biomedical Engineering, National Yang Ming Chiao Tung University, Taipei 112, Taiwan; (Y.-H.H.); (W.-C.C.); (S.-R.F.); (Y.-T.L.); (T.-H.T.)
| | - Tse-Hua Tung
- Department of Biomedical Engineering, National Yang Ming Chiao Tung University, Taipei 112, Taiwan; (Y.-H.H.); (W.-C.C.); (S.-R.F.); (Y.-T.L.); (T.-H.T.)
| | - Chun-Chung Huang
- Department of Biomedical Engineering, National Yang Ming Chiao Tung University, Taipei 112, Taiwan; (Y.-H.H.); (W.-C.C.); (S.-R.F.); (Y.-T.L.); (T.-H.T.)
| | - Chun-Min Lo
- Department of Biomedical Engineering, National Yang Ming Chiao Tung University, Taipei 112, Taiwan; (Y.-H.H.); (W.-C.C.); (S.-R.F.); (Y.-T.L.); (T.-H.T.)
| |
Collapse
|
21
|
Rapier CE, Jagadeesan S, Vatine G, Ben-Yoav H. Microfluidic channel sensory system for electro-addressing cell location, determining confluency, and quantifying a general number of cells. Sci Rep 2022; 12:3248. [PMID: 35228609 PMCID: PMC8885753 DOI: 10.1038/s41598-022-07194-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2021] [Accepted: 02/14/2022] [Indexed: 01/01/2023] Open
Abstract
Microfluidics is a highly useful platform for culturing, monitoring, and testing biological cells. The integration of electrodes into microfluidic channels extends the functionality, sensing, and testing capabilities of microfluidic systems. By employing an electrochemical impedance spectroscopy (EIS) technique, the non-invasive, label-free detection of the activities of cells in real-time can be achieved. To address the movement toward spatially resolving cells in cell culture, we developed a sensory system capable of electro-addressing cell location within a microfluidic channel. This simple system allows for real-time cell location, integrity monitoring (of barrier producing cells), and confluency sensing without the need for frequent optical evaluation-saving time. EIS results demonstrate that cells within microfluidic channels can be located between various pairs of electrodes at different positions along the length of the device. Impedance spectra clearly differentiates between empty, sparse, and confluent microfluidic channels. The system also senses the level of cell confluence between electrode pairs-allowing for the relative quantification of cells in different areas of the microfluidic channel. The system's electrode layout can easily be incorporated into other devices. Namely, organ-on-a-chip devices, that require the monitoring of precise cell location and confluency levels for understanding tissue function, modeling diseases, and for testing therapeutics.
Collapse
Affiliation(s)
- Crystal E Rapier
- Nanobioelectronics Laboratory (NBEL), Department of Biomedical Engineering, Ben-Gurion University of the Negev, Beer Sheva, Israel
| | - Srikanth Jagadeesan
- The Department of Physiology and Cell Biology, Faculty of Health Sciences, The Regenerative Medicine and Stem Cell (RMSC) Research Center, The Zlotowski Center for Neuroscience, Ben-Gurion University of the Negev, Beer Sheva, Israel
| | - Gad Vatine
- The Department of Physiology and Cell Biology, Faculty of Health Sciences, The Regenerative Medicine and Stem Cell (RMSC) Research Center, The Zlotowski Center for Neuroscience, Ben-Gurion University of the Negev, Beer Sheva, Israel.
| | - Hadar Ben-Yoav
- Nanobioelectronics Laboratory (NBEL), Department of Biomedical Engineering, Ben-Gurion University of the Negev, Beer Sheva, Israel.
| |
Collapse
|
22
|
Blood-Brain Barrier Disruption Mediated by FFA1 Receptor-Evidence Using Miniscope. Int J Mol Sci 2022; 23:ijms23042258. [PMID: 35216375 PMCID: PMC8875452 DOI: 10.3390/ijms23042258] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Revised: 02/13/2022] [Accepted: 02/16/2022] [Indexed: 02/05/2023] Open
Abstract
Omega-3 polyunsaturated fatty acids (n-3 PUFAs), obtained from diet and dietary supplements, have been tested in clinical trials for the prevention or treatment of several diseases. n-3 PUFAs exert their effects by activation of free fatty acid (FFA) receptors. FFA1 receptor, expressed in the pancreas and brain, is activated by medium- to long-chain fatty acids. Despite some beneficial effects on cognition, the effects of n-3 PUFAs on the blood-brain barrier (BBB) are not clearly understood. We examined the effects of FFA1 activation on BBB permeability in vitro, using rat brain microvascular endothelial cells (RBMVEC), and in vivo, by assessing Evans Blue extravasation and by performing live imaging of brain microcirculation in adult rats. AMG837, a synthetic FFA1 agonist, produced a dose-dependent decrease in RBMVEC monolayer resistance assessed with Electric Cell-Substrate Impedance Sensing (ECIS); the effect was attenuated by the FFA1 antagonist, GW1100. Immunofluorescence studies revealed that AMG837 produced a disruption in tight and adherens junction proteins. AMG837 increased Evans Blue content in the rat brain in a dose-dependent manner. Live imaging studies of rat brain microcirculation with miniaturized fluorescence microscopy (miniscope) showed that AMG837 increased extravasation of sodium fluorescein. Taken together, our results demonstrate that FFA1 receptor activation reduced RBMVEC barrier function and produced a transient increase in BBB permeability.
Collapse
|
23
|
Fikatas A, Dehairs J, Noppen S, Doijen J, Vanderhoydonc F, Meyen E, Swinnen JV, Pannecouque C, Schols D. Deciphering the Role of Extracellular Vesicles Derived from ZIKV-Infected hcMEC/D3 Cells on the Blood-Brain Barrier System. Viruses 2021; 13:v13122363. [PMID: 34960632 PMCID: PMC8708812 DOI: 10.3390/v13122363] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Revised: 11/12/2021] [Accepted: 11/22/2021] [Indexed: 12/12/2022] Open
Abstract
To date, no vaccines or antivirals are available against Zika virus (ZIKV). In addition, the mechanisms underlying ZIKV-associated pathogenesis of the central nervous system (CNS) are largely unexplored. Getting more insight into the cellular pathways that ZIKV recruits to facilitate infection of susceptible cells will be crucial for establishing an effective treatment strategy. In general, cells secrete a number of vesicles, known as extracellular vesicles (EVs), in response to viral infections. These EVs serve as intercellular communicators. Here, we investigated the role of EVs derived from ZIKV-infected human brain microvascular endothelial cells on the blood–brain barrier (BBB) system. We demonstrated that ZIKV-infected EVs (IEVs) can incorporate viral components, including ZIKV RNA, NS1, and E-protein, and further transfer them to several types of CNS cells. Using label-free impedance-based biosensing, we observed that ZIKV and IEVs can temporally disturb the monolayer integrity of BBB-mimicking cells, possibly by inducing structural rearrangements of the adherent protein VE-cadherin (immunofluorescence staining). Finally, differences in the lipidomic profile between EVs and their parental cells possibly suggest a preferential sorting mechanism of specific lipid species into the vesicles. To conclude, these data suggest that IEVs could be postulated as vehicles (Trojan horse) for ZIKV transmission via the BBB.
Collapse
Affiliation(s)
- Antonios Fikatas
- Laboratory of Virology and Chemotherapy, Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, Faculty of Medicine, KU Leuven, 3000 Leuven, Belgium; (A.F.); (S.N.); (J.D.); (E.M.); (C.P.)
| | - Jonas Dehairs
- Laboratory of Lipid Metabolism and Cancer, Department of Oncology, Faculty of Medicine, KU Leuven, 3000 Leuven, Belgium; (J.D.); (F.V.); (J.V.S.)
| | - Sam Noppen
- Laboratory of Virology and Chemotherapy, Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, Faculty of Medicine, KU Leuven, 3000 Leuven, Belgium; (A.F.); (S.N.); (J.D.); (E.M.); (C.P.)
| | - Jordi Doijen
- Laboratory of Virology and Chemotherapy, Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, Faculty of Medicine, KU Leuven, 3000 Leuven, Belgium; (A.F.); (S.N.); (J.D.); (E.M.); (C.P.)
| | - Frank Vanderhoydonc
- Laboratory of Lipid Metabolism and Cancer, Department of Oncology, Faculty of Medicine, KU Leuven, 3000 Leuven, Belgium; (J.D.); (F.V.); (J.V.S.)
| | - Eef Meyen
- Laboratory of Virology and Chemotherapy, Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, Faculty of Medicine, KU Leuven, 3000 Leuven, Belgium; (A.F.); (S.N.); (J.D.); (E.M.); (C.P.)
| | - Johannes V. Swinnen
- Laboratory of Lipid Metabolism and Cancer, Department of Oncology, Faculty of Medicine, KU Leuven, 3000 Leuven, Belgium; (J.D.); (F.V.); (J.V.S.)
| | - Christophe Pannecouque
- Laboratory of Virology and Chemotherapy, Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, Faculty of Medicine, KU Leuven, 3000 Leuven, Belgium; (A.F.); (S.N.); (J.D.); (E.M.); (C.P.)
| | - Dominique Schols
- Laboratory of Virology and Chemotherapy, Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, Faculty of Medicine, KU Leuven, 3000 Leuven, Belgium; (A.F.); (S.N.); (J.D.); (E.M.); (C.P.)
- Correspondence: ; Tel.: +32-16-32-19-98
| |
Collapse
|
24
|
Modelling and Differential Quantification of Electric Cell-Substrate Impedance Sensing Growth Curves. SENSORS 2021; 21:s21165286. [PMID: 34450726 PMCID: PMC8401457 DOI: 10.3390/s21165286] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 07/26/2021] [Accepted: 08/02/2021] [Indexed: 11/29/2022]
Abstract
Measurement of cell surface coverage has become a common technique for the assessment of growth behavior of cells. As an indirect measurement method, this can be accomplished by monitoring changes in electrode impedance, which constitutes the basis of electric cell-substrate impedance sensing (ECIS). ECIS typically yields growth curves where impedance is plotted against time, and changes in single cell growth behavior or cell proliferation can be displayed without significantly impacting cell physiology. To provide better comparability of ECIS curves in different experimental settings, we developed a large toolset of R scripts for their transformation and quantification. They allow importing growth curves generated by ECIS systems, edit, transform, graph and analyze them while delivering quantitative data extracted from reference points on the curve. Quantification is implemented through three different curve fit algorithms (smoothing spline, logistic model, segmented regression). From the obtained models, curve reference points such as the first derivative maximum, segmentation knots and area under the curve are then extracted. The scripts were tested for general applicability in real-life cell culture experiments on partly anonymized cell lines, a calibration setup with a cell dilution series of impedance versus seeded cell number and finally IPEC-J2 cells treated with 1% and 5% ethanol.
Collapse
|
25
|
Marottoli FM, Trevino TN, Geng X, Arbieva Z, Kanabar P, Maienschein-Cline M, Lee JC, Lutz SE, Tai LM. Autocrine Effects of Brain Endothelial Cell-Produced Human Apolipoprotein E on Metabolism and Inflammation in vitro. Front Cell Dev Biol 2021; 9:668296. [PMID: 34178992 PMCID: PMC8225247 DOI: 10.3389/fcell.2021.668296] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Accepted: 05/11/2021] [Indexed: 11/13/2022] Open
Abstract
Reports of APOE4-associated neurovascular dysfunction during aging and in neurodegenerative disorders has led to ongoing research to identify underlying mechanisms. In this study, we focused on whether the APOE genotype of brain endothelial cells modulates their own phenotype. We utilized a modified primary mouse brain endothelial cell isolation protocol that enabled us to perform experiments without subculture. Through initial characterization we found, that compared to APOE3, APOE4 brain endothelial cells produce less apolipoprotein E (apoE) and have altered metabolic and inflammatory gene expression profiles. Further analysis revealed APOE4 brain endothelial cultures have higher preference for oxidative phosphorylation over glycolysis and, accordingly, higher markers of mitochondrial activity. Mitochondrial activity generates reactive oxygen species, and, with APOE4, there were higher mitochondrial superoxide levels, lower levels of antioxidants related to heme and glutathione and higher markers/outcomes of oxidative damage to proteins and lipids. In parallel, or resulting from reactive oxygen species, there was greater inflammation in APOE4 brain endothelial cells including higher chemokine levels and immune cell adhesion under basal conditions and after low-dose lipopolysaccharide (LPS) treatment. In addition, paracellular permeability was higher in APOE4 brain endothelial cells in basal conditions and after high-dose LPS treatment. Finally, we found that a nuclear receptor Rev-Erb agonist, SR9009, improved functional metabolic markers, lowered inflammation and modulated paracellular permeability at baseline and following LPS treatment in APOE4 brain endothelial cells. Together, our data suggest that autocrine signaling of apoE in brain endothelial cells represents a novel cellular mechanism for how APOE regulates neurovascular function.
Collapse
Affiliation(s)
- Felecia M Marottoli
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, IL, United States
| | - Troy N Trevino
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, IL, United States
| | - Xue Geng
- Department of Bioengineering, University of Illinois at Chicago, Chicago, IL, United States
| | - Zarema Arbieva
- Genome Research Core, Research Resources Center, University of Illinois at Chicago, Chicago, IL, United States
| | - Pinal Kanabar
- Research Informatics Core, Research Resources Center, University of Illinois at Chicago, Chicago, IL, United States
| | - Mark Maienschein-Cline
- Research Informatics Core, Research Resources Center, University of Illinois at Chicago, Chicago, IL, United States
| | - James C Lee
- Department of Bioengineering, University of Illinois at Chicago, Chicago, IL, United States
| | - Sarah E Lutz
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, IL, United States
| | - Leon M Tai
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, IL, United States
| |
Collapse
|
26
|
Mucopolysaccharide polysulfate promotes microvascular stabilization and barrier integrity of dermal microvascular endothelial cells via activation of the angiopoietin-1/Tie2 pathway. J Dermatol Sci 2021; 103:25-32. [PMID: 34148739 DOI: 10.1016/j.jdermsci.2021.05.008] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Revised: 04/28/2021] [Accepted: 05/31/2021] [Indexed: 01/01/2023]
Abstract
BACKGROUND Mucopolysaccharide polysulfate (MPS) is a heparinoid and MPS-containing formulations are widely used as moisturizers for dry skin and to treat peripheral vascular insufficiency. Although MPS has therapeutic effects in skin diseases with microvascular abnormalities, the effects of MPS on microvascular function remain incompletely understood. OBJECTIVE The aim of this study was to evaluate the functional activities of MPS on human pericytes (HPC) and human dermal microvascular endothelial cells (HDMEC) in vitro, and on microvascular permeability of the skin. METHODS The protein expression of angiopoietin (Ang)-1 in HPC, and platelet-derived growth factor-BB (PDGF-BB) and phosphorylated tyrosine-protein kinase receptor 2 (Tie2) in HDMEC were measured in the presence or absence of MPS. The vascular barrier was evaluated by the expressions of claudin-5 and vascular endothelial (VE)-cadherin, and transendothelial electrical resistance (TEER). RESULTS In HPC, MPS dose-dependently enhanced Ang-1 secretion, which activated Tie2 in HDMEC. In HDMEC, MPS significantly increased the production of PDGF-BB, which is important for the recruitment of HPC to the vascular endothelium, and significantly increased the phosphorylation of Tie2, which results in the activation of the Ang-1/Tie2 signaling . MPS significantly increased the expression of tight junction protein claudin-5 and TEER in the HDMEC. Moreover, the intradermal injection of MPS prevented vascular endothelial growth factor-induced increase in vascular permeability in mouse skin. CONCLUSION We found that MPS promoted microvascular stabilization and barrier integrity in HDMEC via Ang-1/Tie2 activation. These results suggest that MPS might improve microvascular abnormalities in various diseases accompanied by disturbances in Ang-1/Tie2 signaling.
Collapse
|
27
|
Wang SH, Tung TH, Chiu SP, Chou HY, Hung YH, Lai YT, Lee YW, Lee SP, Lo CM. Detecting Effects of Low Levels of FCCP on Stem Cell Micromotion and Wound-Healing Migration by Time-Series Capacitance Measurement. SENSORS 2021; 21:s21093017. [PMID: 33923058 PMCID: PMC8123359 DOI: 10.3390/s21093017] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Revised: 04/23/2021] [Accepted: 04/23/2021] [Indexed: 01/17/2023]
Abstract
Electric cell–substrate impedance sensing (ECIS) has been used as a real-time impedance-based method to quantify cell behavior in tissue culture. The method is capable of measuring both the resistance and capacitance of a cell-covered microelectrode at various AC frequencies. In this study, we demonstrate the application of high-frequency capacitance measurement (f = 40 or 64 kHz) for the sensitive detection of both the micromotion and wound-healing migration of human mesenchymal stem cells (hMSCs). Impedance measurements of cell-covered electrodes upon the challenge of various concentrations of carbonyl cyanide 4-(trifluoromethoxy)phenylhydrazone (FCCP), from 0.1 to 30 μM, were conducted using ECIS. FCCP is an uncoupler of mitochondrial oxidative phosphorylation (OXPHOS), thereby reducing mitochondrial ATP production. By numerically analyzing the time-series capacitance data, a dose-dependent decrease in hMSC micromotion and wound-healing migration was observed, and the effect was significantly detected at levels as low as 0.1 μM. While most reported works with ECIS use the resistance/impedance time series, our results suggest the potential use of high-frequency capacitance time series for assessing migratory cell behavior such as micromotion and wound-healing migration.
Collapse
Affiliation(s)
- Si-Han Wang
- Department of Biomedical Engineering, National Yang-Ming University, Taipei 11221, Taiwan; (S.-H.W.); (T.-H.T.); (H.-Y.C.); (Y.-H.H.); (Y.-T.L.); (Y.-W.L.)
| | - Tse-Hua Tung
- Department of Biomedical Engineering, National Yang-Ming University, Taipei 11221, Taiwan; (S.-H.W.); (T.-H.T.); (H.-Y.C.); (Y.-H.H.); (Y.-T.L.); (Y.-W.L.)
| | - Sheng-Po Chiu
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Tri-Service General Hospital Songshan Branch, National Defense Medical Center, Taipei 11490, Taiwan;
| | - Hsin-Yi Chou
- Department of Biomedical Engineering, National Yang-Ming University, Taipei 11221, Taiwan; (S.-H.W.); (T.-H.T.); (H.-Y.C.); (Y.-H.H.); (Y.-T.L.); (Y.-W.L.)
| | - Yu-Han Hung
- Department of Biomedical Engineering, National Yang-Ming University, Taipei 11221, Taiwan; (S.-H.W.); (T.-H.T.); (H.-Y.C.); (Y.-H.H.); (Y.-T.L.); (Y.-W.L.)
| | - Yi-Ting Lai
- Department of Biomedical Engineering, National Yang-Ming University, Taipei 11221, Taiwan; (S.-H.W.); (T.-H.T.); (H.-Y.C.); (Y.-H.H.); (Y.-T.L.); (Y.-W.L.)
| | - Yu-Wei Lee
- Department of Biomedical Engineering, National Yang-Ming University, Taipei 11221, Taiwan; (S.-H.W.); (T.-H.T.); (H.-Y.C.); (Y.-H.H.); (Y.-T.L.); (Y.-W.L.)
| | - Shiao-Pieng Lee
- Division of Oral and Maxillofacial Surgery, Department of Dentistry, Tri-Service General Hospital, Taipei 11490, Taiwan
- School of Dentistry, National Defense Medical Center, Taipei 11490, Taiwan
- Department of Biomedical Engineering, National Defense Medical Center, Taipei 11490, Taiwan
- Correspondence: (S.-P.L.); (C.-M.L.)
| | - Chun-Min Lo
- Department of Biomedical Engineering, National Yang-Ming University, Taipei 11221, Taiwan; (S.-H.W.); (T.-H.T.); (H.-Y.C.); (Y.-H.H.); (Y.-T.L.); (Y.-W.L.)
- Correspondence: (S.-P.L.); (C.-M.L.)
| |
Collapse
|
28
|
Callesen KT, Yuste-Montalvo A, Poulsen LK, Jensen BM, Esteban V. In Vitro Investigation of Vascular Permeability in Endothelial Cells from Human Artery, Vein and Lung Microvessels at Steady-State and Anaphylactic Conditions. Biomedicines 2021; 9:biomedicines9040439. [PMID: 33921871 PMCID: PMC8072631 DOI: 10.3390/biomedicines9040439] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2021] [Revised: 03/27/2021] [Accepted: 04/14/2021] [Indexed: 12/30/2022] Open
Abstract
Human anaphylactic reactions largely involve an increase in vascular permeability, which is mainly controlled by endothelial cells (ECs). Due to the acute and serious nature of human anaphylaxis, in vivo studies of blood vessels must be replaced or supplemented with in vitro models. Therefore, we used a macromolecular tracer assay (MMTA) to investigate the EC permeability of three phenotypes of human ECs: artery (HAECs), vein (HSVECs) and microvessels from lung (HMLECs). ECs were stimulated with two fast-acting anaphylactic mediators (histamine and platelet-activating factor (PAF)) and one longer-lasting mediator (thrombin). At steady-state conditions, HSVEC monolayers were the most permeable and HMLEC the least (15.8% and 8.3% after 60 min, respectively). No response was found in ECs from artery or vein to any stimuli. ECs from microvessels reacted to stimulation with thrombin and also demonstrated a tendency of increased permeability for PAF. There was no reaction for histamine. This was not caused by missing receptor expression, as all three EC phenotypes expressed receptors for both PAF and histamine. The scarce response to fast-acting mediators illustrates that the MMTA is not suitable for investigating EC permeability to anaphylactic mediators.
Collapse
Affiliation(s)
- Katrine T. Callesen
- Laboratory of Medical Allergology, Copenhagen University Hospital at Gentofte, DK-2900 Hellerup, Denmark; (K.T.C.); (L.K.P.); (B.M.J.)
| | - Alma Yuste-Montalvo
- Department of Allergy and Immunology, IIS-Fundación Jiménez Díaz, UAM, 28040 Madrid, Spain;
| | - Lars K. Poulsen
- Laboratory of Medical Allergology, Copenhagen University Hospital at Gentofte, DK-2900 Hellerup, Denmark; (K.T.C.); (L.K.P.); (B.M.J.)
| | - Bettina M. Jensen
- Laboratory of Medical Allergology, Copenhagen University Hospital at Gentofte, DK-2900 Hellerup, Denmark; (K.T.C.); (L.K.P.); (B.M.J.)
| | - Vanesa Esteban
- Department of Allergy and Immunology, IIS-Fundación Jiménez Díaz, UAM, 28040 Madrid, Spain;
- Faculty of Medicine and Biomedicine, Alfonso X El Sabio University, 28691 Madrid, Spain
- Red de Asma, Reacciones Adversas y Alérgicas (ARADyAL), Instituto de Salud Carlos III, 28029 Madrid, Spain
- Correspondence:
| |
Collapse
|
29
|
Moysidou C, Pitsalidis C, Al‐Sharabi M, Withers AM, Zeitler JA, Owens RM. 3D Bioelectronic Model of the Human Intestine. Adv Biol (Weinh) 2021. [DOI: 10.1002/adbi.202000306] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Affiliation(s)
- Chrysanthi‐Maria Moysidou
- Department of Chemical Engineering and Biotechnology University of Cambridge Philippa Fawcett Drive Cambridge CB3 0AS UK
| | - Charalampos Pitsalidis
- Department of Chemical Engineering and Biotechnology University of Cambridge Philippa Fawcett Drive Cambridge CB3 0AS UK
| | - Mohammed Al‐Sharabi
- Department of Chemical Engineering and Biotechnology University of Cambridge Philippa Fawcett Drive Cambridge CB3 0AS UK
| | - Aimee M. Withers
- Department of Chemical Engineering and Biotechnology University of Cambridge Philippa Fawcett Drive Cambridge CB3 0AS UK
| | - J. Axel Zeitler
- Department of Chemical Engineering and Biotechnology University of Cambridge Philippa Fawcett Drive Cambridge CB3 0AS UK
| | - Róisín M. Owens
- Department of Chemical Engineering and Biotechnology University of Cambridge Philippa Fawcett Drive Cambridge CB3 0AS UK
| |
Collapse
|
30
|
Santaterra VAG, Fiusa MML, Hounkpe BW, Chenou F, Tonasse WV, da Costa LNG, Garcia-Weber D, Domingos IDF, de Lima F, Borba-Junior IT, Araújo ADS, Lucena-Araújo AR, Bezerra MAC, Dos Santos MNN, Costa FF, Millán J, De Paula EV. Endothelial Barrier Integrity Is Disrupted In Vitro by Heme and by Serum From Sickle Cell Disease Patients. Front Immunol 2020; 11:535147. [PMID: 33381108 PMCID: PMC7767881 DOI: 10.3389/fimmu.2020.535147] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2020] [Accepted: 11/04/2020] [Indexed: 12/14/2022] Open
Abstract
Free extracellular heme has been shown to activate several compartments of innate immunity, acting as a danger-associated molecular pattern (DAMP) in hemolytic diseases. Although localized endothelial barrier (EB) disruption is an important part of inflammation that allows circulating leukocytes to reach inflamed tissues, non-localized/deregulated disruption of the EB can lead to widespread microvascular hyperpermeability and secondary tissue damage. In mouse models of sickle cell disease (SCD), EB disruption has been associated with the development of a form of acute lung injury that closely resembles acute chest syndrome (ACS), and that can be elicited by acute heme infusion. Here we explored the effect of heme on EB integrity using human endothelial cell monolayers, in experimental conditions that include elements that more closely resemble in vivo conditions. EB integrity was assessed by electric cell-substrate impedance sensing in the presence of varying concentrations of heme and sera from SCD patients or healthy volunteers. Heme caused a dose-dependent decrease of the electrical resistance of cell monolayers, consistent with EB disruption, which was confirmed by staining of junction protein VE-cadherin. In addition, sera from SCD patients, but not from healthy volunteers, were also capable to induce EB disruption. Interestingly, these effects were not associated with total heme levels in serum. However, when heme was added to sera from SCD patients, but not from healthy volunteers, EB disruption could be elicited, and this effect was associated with hemopexin serum levels. Together our in vitro studies provide additional support to the concept of heme as a DAMP in hemolytic conditions.
Collapse
Affiliation(s)
| | | | | | - Francine Chenou
- School of Medical Sciences, University of Campinas, Campinas, Brazil
| | | | - Loredana Nilkenes Gomes da Costa
- School of Medical Sciences, University of Campinas, Campinas, Brazil.,Department of Biomedicine, Federal University of Piaui, Parnaiba, Brazil
| | - Diego Garcia-Weber
- Centro de Biologia Molecular Severo Ochoa, Consejo Superior de Investigaciones Cientificas, Universidad Autonoma de Madrid, Madrid, Spain
| | - Igor de Farias Domingos
- Genetics Postgraduate Program, Federal University of Pernambuco, Recife, Brazil.,Department of Clinical and Toxicological Analysis, Federal University of Rio Grande do Norte, Natal, Brazil
| | - Franciele de Lima
- School of Medical Sciences, University of Campinas, Campinas, Brazil
| | | | - Aderson da Silva Araújo
- Department of Internal Medicine, Hematology and Hemotherapy Foundation of Pernambuco (HEMOPE), Recife, Brazil
| | | | | | | | - Fernando Ferreira Costa
- School of Medical Sciences, University of Campinas, Campinas, Brazil.,Hematology and Hemotherapy Center, University of Campinas, Campinas, Brazil
| | - Jaime Millán
- Centro de Biologia Molecular Severo Ochoa, Consejo Superior de Investigaciones Cientificas, Universidad Autonoma de Madrid, Madrid, Spain
| | - Erich Vinicius De Paula
- School of Medical Sciences, University of Campinas, Campinas, Brazil.,Hematology and Hemotherapy Center, University of Campinas, Campinas, Brazil
| |
Collapse
|
31
|
Abayzeed SA. Plasmonic-based impedance microspectroscopy of optically heterogeneous samples. BIOMEDICAL OPTICS EXPRESS 2020; 11:6168-6180. [PMID: 33282482 PMCID: PMC7687972 DOI: 10.1364/boe.395474] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Revised: 06/19/2020] [Accepted: 06/22/2020] [Indexed: 05/20/2023]
Abstract
A robust impedance microscopy technique is presented. This optical tool enables high resolution imaging of electrical properties with promising biophysical applications. The underlying principle is that surface plasmon resonance (SPR) sensors are able to measure perturbations of surface charge density and therefore can be used to compute the impedance of surface-adhered cells. However, the ability to perform reliable quantitative impedance imaging is affected by the optical heterogeneity of the cell-sensor interface. To address this issue, a novel method for quantitative time-resolved resonance angle tracking is developed and applied to correct for the effect of the optical properties. To demonstrate the capability of this technique, impedance microspectroscopy of bovine serum albumin (BSA) patterns was performed enabling measurements of capacitance with submicroscopic resolution. The work presented offers an impedance microspectroscopy method that will create new avenues in studying the electrical properties of single cells and biomolecules as well as bio-electrical currents.
Collapse
|
32
|
Abasi S, Bhat A, Guiseppi‐Elie A. Electrode Selection for Electrostimulation and TEER Using ECSARA. ELECTROANAL 2020. [DOI: 10.1002/elan.202060313] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Affiliation(s)
- Sara Abasi
- Center for Bioelectronics Biosensors and Biochips (C3B®) Department of Biomedical Engineering and Department of Electrical and Computer Engineering Texas A&M University College Station TX 77843 USA
| | - Ankita Bhat
- Center for Bioelectronics Biosensors and Biochips (C3B®) Department of Biomedical Engineering and Department of Electrical and Computer Engineering Texas A&M University College Station TX 77843 USA
| | - Anthony Guiseppi‐Elie
- Center for Bioelectronics Biosensors and Biochips (C3B®) Department of Biomedical Engineering and Department of Electrical and Computer Engineering Texas A&M University College Station TX 77843 USA
- Houston Methodist Institute for Academic Medicine and Houston Methodist Research Institute 6670 Bertner Ave. Houston TX 77030 USA
- Department of Electrical and Computer Engineering Texas A&M University College Station TX 77843 USA
- ABTECH Scientific, Inc. Biotechnology Research Park 800 East Leigh Street Richmond VA 23219 USA
| |
Collapse
|
33
|
Bussooa A. Characterising Vascular Cell Monolayers Using Electrochemical Impedance Spectroscopy and a Novel Electroanalytical Plot. Nanotechnol Sci Appl 2020; 13:89-101. [PMID: 33061321 PMCID: PMC7520662 DOI: 10.2147/nsa.s266663] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Accepted: 08/27/2020] [Indexed: 11/23/2022] Open
Abstract
INTRODUCTION Biological research relies on the culture of mammalian cells, which are prone to changes in phenotype during experiments involving several passages of cells. In regenerative medicine, specifically, there is an increasing need to expand the characterisation landscape for stem cells by identifying novel stable markers. This paper reports on a novel electric cell-substrate impedance sensing-based electroanalytical diagram which can be used for the "electrical characterisation" of cell monolayers consisting of smooth muscle cells, endothelial cells or co-culture. MATERIALS AND METHODS Interdigitated electrodes were microfabricated using standard cleanroom procedures and integrated into cell chambers. Electrochemical impedance spectroscopy data were acquired for 2 vascular cell types after they formed monolayers on the electrodes. RESULTS AND DISCUSSION A Mean impedance per unit area vs Mean phase plots provided a reproducible, visually obvious and statistically significant method of characterising cell monolayers. This electroanalytic diagram has never been used in previous papers, but it confirms findings by other research groups using similar approaches that the complex impedance spectra of different cell type are different. Further work is required to determine whether this method could be extended to other cell types, and if this is the case, a library of "signature spectra" could be generated for "electrical characterisation" of cells.
Collapse
Affiliation(s)
- Anubhav Bussooa
- BHF Cardiovascular Research Centre, University of Glasgow, Glasgow G12 8TA, UK
| |
Collapse
|
34
|
Anwer S, Szaszi K. Measuring Cell Growth and Junction Development in Epithelial Cells Using Electric Cell-Substrate Impedance Sensing (ECIS). Bio Protoc 2020; 10:e3729. [PMID: 33659390 DOI: 10.21769/bioprotoc.3729] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2020] [Revised: 06/18/2020] [Accepted: 07/14/2020] [Indexed: 12/11/2022] Open
Abstract
Electric Cell-substrate Impedance Sensing (ECIS) is an automated method that can be used to quantify processes such as cell attachment, growth, migration and barrier functions (i.e., the properties of tight junctions). The method provides simultaneous information on cell number and tight junction function by detecting electric parameters of cells grown on electrodes. Samples are probed with small alternating current (AC) over a range of frequencies, and changes in capacitance and impedance are measured over time. Capacitance reflects the degree of electrode coverage by cells, that correlates with cell number, and can be used to assess cell proliferation or migration. Impedance values inform about barrier function. Obtaining real-time simultaneous information on these parameters is unique to this system and is of great value for addressing fundamental questions such as the role of tight junction proteins in cell growth and migration. This protocol describes the use of ECIS to follow cell growth and tight junction-dependent barrier generation in tubular epithelial cells. We used this method to explore how depleting claudin-2, a tight junction protein affects tubular cell growth and barrier function. During the process, cells are transfected with control or claudin-2-specific siRNA, and 24h later plated on electrodes. ECIS automatically collects information on cell growth and barrier as the monolayer develops. The data are initially analyzed using the ECIS software and exported into a graph software for further processing.
Collapse
Affiliation(s)
- Shaista Anwer
- Keenan Research Centre for Biomedical Science of the St. Michael's Hospital, Toronto, Canada
| | - Katalin Szaszi
- Keenan Research Centre for Biomedical Science of the St. Michael's Hospital, Toronto, Canada.,Dept. of Surgery, University of Toronto, ON, Canada
| |
Collapse
|
35
|
Correlation of host inflammatory cytokines and immune-related metabolites, but not viral NS1 protein, with disease severity of dengue virus infection. PLoS One 2020; 15:e0237141. [PMID: 32764789 PMCID: PMC7413495 DOI: 10.1371/journal.pone.0237141] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2019] [Accepted: 07/21/2020] [Indexed: 12/13/2022] Open
Abstract
Severe dengue can be lethal caused by manifestations such as severe bleeding, fluid accumulation and organ impairment. This study aimed to investigate the role of dengue non-structural 1 (NS1) protein and host factors contributing to severe dengue. Electrical cell-substrate impedance sensing system was used to investigate the changes in barrier function of microvascular endothelial cells treated NS1 protein and serum samples from patients with different disease severity. Cytokines and metabolites profiles were assessed using a multiplex cytokine assay and liquid chromatography mass spectrometry respectively. The findings showed that NS1 was able to induce the loss of barrier function in microvascular endothelium in a dose dependent manner, however, the level of NS1 in serum samples did not correlate with the extent of vascular leakage induced. Further assessment of host factors revealed that cytokines such as CCL2, CCL5, CCL20 and CXCL1, as well as adhesion molecule ICAM-1, that are involved in leukocytes infiltration were expressed higher in dengue patients in comparison to healthy individuals. In addition, metabolomics study revealed the presence of deregulated metabolites involved in the phospholipid metabolism pathway in patients with severe manifestations. In conclusion, disease severity in dengue virus infection did not correlate directly with NS1 level, but instead with host factors that are involved in the regulation of junctional integrity and phospholipid metabolism. However, as the studied population was relatively small in this study, these exploratory findings should be confirmed by expanding the sample size using an independent cohort to further establish the significance of this study.
Collapse
|
36
|
Bolla BS, Erdei L, Urbán E, Burián K, Kemény L, Szabó K. Cutibacterium acnes regulates the epidermal barrier properties of HPV-KER human immortalized keratinocyte cultures. Sci Rep 2020; 10:12815. [PMID: 32733073 PMCID: PMC7393503 DOI: 10.1038/s41598-020-69677-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2020] [Accepted: 07/13/2020] [Indexed: 12/20/2022] Open
Abstract
Our skin provides a physical barrier to separate the internal part of our body from the environment. Maintenance of complex barrier functions is achieved through anatomical structures in the skin, the stratified squamous epithelium specialized junctional organelles, called tight junctions (TJs). Several members of our microbial communities are known to affect the differentiation state and function of the colonized organ. Whether and how interactions between skin cells and cutaneous microbes, including Cutibacterium acnes (C. acnes), modify the structure and/or function of our skin is currently only partly understood. Thus, in our studies, we investigated whether C. acnes may affect the epidermal barrier using in vitro model systems. Real-time cellular analysis showed that depending on the keratinocyte differentiation state, the applied C. acnes strains and their dose, the measured impedance values change, together with the expression of selected TJ proteins. These may reflect barrier alterations, which can be partially restored upon antibiotic–antimycotic treatment. Our findings suggest that C. acnes can actively modify the barrier properties of cultured keratinocytes, possibly through alteration of tight cell-to-cell contacts. Similar events may play important roles in our skin, in the maintenance of cutaneous homeostasis.
Collapse
Affiliation(s)
- Beáta Szilvia Bolla
- Department of Dermatology and Allergology, University of Szeged, Szeged, Hungary.,HCEMM-SZTE Skin Research Group, Szeged, Hungary
| | - Lilla Erdei
- Department of Dermatology and Allergology, University of Szeged, Szeged, Hungary.,HCEMM-SZTE Skin Research Group, Szeged, Hungary
| | - Edit Urbán
- Department of Public Health, University of Szeged, Szeged, Hungary
| | - Katalin Burián
- Institute of Clinical Microbiology, University of Szeged, Szeged, Hungary
| | - Lajos Kemény
- Department of Dermatology and Allergology, University of Szeged, Szeged, Hungary.,HCEMM-SZTE Skin Research Group, Szeged, Hungary.,MTA-SZTE Dermatological Research Group, Szeged, Hungary
| | - Kornélia Szabó
- Department of Dermatology and Allergology, University of Szeged, Szeged, Hungary. .,MTA-SZTE Dermatological Research Group, Szeged, Hungary.
| |
Collapse
|
37
|
Tung TH, Wang SH, Huang CC, Su TY, Lo CM. Use of Discrete Wavelet Transform to Assess Impedance Fluctuations Obtained from Cellular Micromotion. SENSORS 2020; 20:s20113250. [PMID: 32517325 PMCID: PMC7309086 DOI: 10.3390/s20113250] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Revised: 06/05/2020] [Accepted: 06/05/2020] [Indexed: 12/21/2022]
Abstract
Electric cell–substrate impedance sensing (ECIS) is an attractive method for monitoring cell behaviors in tissue culture in real time. The time series impedance fluctuations of the cell-covered electrodes measured by ECIS are the phenomena accompanying cellular micromotion as cells continually rearrange their cell–cell and cell–substrate adhesion sites. Accurate assessment of these fluctuations to extract useful information from raw data is important for both scientific and practical purposes. In this study, we apply discrete wavelet transform (DWT) to analyze the concentration-dependent effect of cytochalasin B on human umbilical vein endothelial cells (HUVECs). The sampling rate of the impedance time series is 1 Hz and each data set consists of 2048 points. Our results demonstrate that, in the Daubechies (db) wavelet family, db1 is the optimal mother wavelet function for DWT-based analysis to assess the effect of cytochalasin B on HUVEC micromotion. By calculating the energy, standard deviation, variance, and signal magnitude area of DWT detail coefficients at level 1, we are able to significantly distinguish cytotoxic concentrations of cytochalasin B as low as 0.1 μM, and in a concentration-dependent manner. Furthermore, DWT-based analysis indicates the possibility to decrease the sampling rate of the micromotion measurement from 1 Hz to 1/16 Hz without decreasing the discerning power. The statistical measures of DWT detail coefficients are effective methods for determining both the sampling rate and the number of individual samples for ECIS-based micromotion assays.
Collapse
Affiliation(s)
- Tse-Hua Tung
- Department of Biomedical Engineering, National Yang-Ming University, Taipei 11221, Taiwan; (T.-H.T.); (S.-H.W.); (C.-C.H.)
| | - Si-Han Wang
- Department of Biomedical Engineering, National Yang-Ming University, Taipei 11221, Taiwan; (T.-H.T.); (S.-H.W.); (C.-C.H.)
| | - Chun-Chung Huang
- Department of Biomedical Engineering, National Yang-Ming University, Taipei 11221, Taiwan; (T.-H.T.); (S.-H.W.); (C.-C.H.)
| | - Tai-Yuan Su
- Department of Electrical Engineering, Yuan-Ze University, Chung-Li 32003, Taiwan
- Correspondence: (T.-Y.S.); (C.-M.L.)
| | - Chun-Min Lo
- Department of Biomedical Engineering, National Yang-Ming University, Taipei 11221, Taiwan; (T.-H.T.); (S.-H.W.); (C.-C.H.)
- Correspondence: (T.-Y.S.); (C.-M.L.)
| |
Collapse
|
38
|
Phenotypic and functional characterization of corneal endothelial cells during in vitro expansion. Sci Rep 2020; 10:7402. [PMID: 32366916 PMCID: PMC7198491 DOI: 10.1038/s41598-020-64311-x] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2019] [Accepted: 02/14/2020] [Indexed: 02/08/2023] Open
Abstract
The advent of cell culture-based methods for the establishment and expansion of human corneal endothelial cells (CEnC) has provided a source of transplantable corneal endothelium, with a significant potential to challenge the one donor-one recipient paradigm. However, concerns over cell identity remain, and a comprehensive characterization of the cultured CEnC across serial passages has not been performed. To this end, we compared two established CEnC culture methods by assessing the transcriptomic changes that occur during in vitro expansion. In confluent monolayers, low mitogenic culture conditions preserved corneal endothelial cell state identity better than culture in high mitogenic conditions. Expansion by continuous passaging induced replicative cell senescence. Transcriptomic analysis of the senescent phenotype identified a cell senescence signature distinct for CEnC. We identified activation of both classic and new cell signaling pathways that may be targeted to prevent senescence, a significant barrier to realizing the potential clinical utility of in vitro expansion.
Collapse
|
39
|
Alfano DN, Klei LR, Klei HB, Trotta M, Gough PJ, Foley KP, Bertin J, Sumpter TL, Lucas PC, McAllister-Lucas LM. MALT1 Protease Plays a Dual Role in the Allergic Response by Acting in Both Mast Cells and Endothelial Cells. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2020; 204:2337-2348. [PMID: 32213560 DOI: 10.4049/jimmunol.1900281] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/08/2019] [Accepted: 02/21/2020] [Indexed: 01/26/2023]
Abstract
The signaling protein MALT1 plays a key role in promoting NF-κB activation in Ag-stimulated lymphocytes. In this capacity, MALT1 has two functions, acting as a scaffolding protein and as a substrate-specific protease. MALT1 is also required for NF-κB-dependent induction of proinflammatory cytokines after FcεR1 stimulation in mast cells, implicating a role in allergy. Because MALT1 remains understudied in this context, we sought to investigate how MALT1 proteolytic activity contributes to the overall allergic response. We compared bone marrow-derived mast cells from MALT1 knockout (MALT1-/-) and MALT1 protease-deficient (MALTPD/PD) mice to wild-type cells. We found that MALT1-/- and MALT1PD/PD mast cells are equally impaired in cytokine production following FcεRI stimulation, indicating that MALT1 scaffolding activity is insufficient to drive the cytokine response and that MALT1 protease activity is essential. In addition to cytokine production, acute mast cell degranulation is a critical component of allergic response. Intriguingly, whereas degranulation is MALT1-independent, MALT1PD/PD mice are protected from vascular edema induced by either passive cutaneous anaphylaxis or direct challenge with histamine, a major granule component. This suggests a role for MALT1 protease activity in endothelial cells targeted by mast cell-derived vasoactive substances. Indeed, we find that in human endothelial cells, MALT1 protease is activated following histamine treatment and is required for histamine-induced permeability. We thus propose a dual role for MALT1 protease in allergic response, mediating 1) IgE-dependent mast cell cytokine production, and 2) histamine-induced endothelial permeability. This dual role indicates that therapeutic inhibitors of MALT1 protease could work synergistically to control IgE-mediated allergic disease.
Collapse
Affiliation(s)
- Danielle N Alfano
- Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, PA 15224
| | - Linda R Klei
- Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, PA 15224
| | - Hanna B Klei
- Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, PA 15224
| | - Matthew Trotta
- Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, PA 15224
| | - Peter J Gough
- Pattern Recognition Receptor Discovery Performance Unit, GlaxoSmithKline, Collegeville, PA 19406
| | - Kevin P Foley
- Pattern Recognition Receptor Discovery Performance Unit, GlaxoSmithKline, Collegeville, PA 19406
| | - John Bertin
- Pattern Recognition Receptor Discovery Performance Unit, GlaxoSmithKline, Collegeville, PA 19406
| | - Tina L Sumpter
- Department of Dermatology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15224
| | - Peter C Lucas
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15224; and .,Vascular Medicine Institute, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261
| | - Linda M McAllister-Lucas
- Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, PA 15224; .,Vascular Medicine Institute, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261
| |
Collapse
|
40
|
Shivappagowdar A, Pati S, Narayana C, Ayana R, Kaushik H, Sah R, Garg S, Khanna A, Kumari J, Garg L, Sagar R, Singh S. A small bioactive glycoside inhibits epsilon toxin and prevents cell death. Dis Model Mech 2019; 12:dmm.040410. [PMID: 31492678 PMCID: PMC6826021 DOI: 10.1242/dmm.040410] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2019] [Accepted: 08/23/2019] [Indexed: 12/24/2022] Open
Abstract
Clostridium perfringens epsilon toxin (Etx) is categorized as the third most lethal bioterrorism agent by the Centers for Disease Control and Prevention (CDC), with no therapeutic counter measures available for humans. Here, we have developed a high-affinity inhibitory compound by synthesizing and evaluating the structure activity relationship (SAR) of a library of diverse glycosides (numbered 1-12). SAR of glycoside-Etx heptamers revealed exceptionally strong H-bond interactions of glycoside-4 with a druggable pocket in the oligomerization and β-hairpin region of Etx. Analysis of its structure suggested that glycoside-4 might self-aggregate to form a robust micelle-like supra-molecular complex due to its linear side-chain architecture, which was authenticated by fluorescence spectroscopy. Further, this micelle hinders the Etx monomer-monomer interaction required for oligomerization, validated by both surface plasmon resonance (SPR) and immunoblotting. This phenomenon in turn leads to blockage of pore formation. Downstream evaluation revealed that glycoside-4 effectively blocked cell death of Etx-treated cultured primary cells and maintained cellular homeostasis via disrupting oligomerization, blocking pore formation, restoring calcium homeostasis, stabilizing the mitochondrial membrane and impairing high mobility group box 1 (HMGB1) translocation from nucleus to cytoplasm. Furthermore, a single dosage of glycoside-4 protected the Etx-challenged mice and restored normal function to multiple organs. This work reports for the first time a potent, nontoxic glycoside with strong ability to occlude toxin lethality, representing it as a bio-arm therapeutic against Etx-based biological threat.
Collapse
Affiliation(s)
- Abhishek Shivappagowdar
- Department of Life Sciences, School of Natural Sciences, Shiv Nadar University, Greater Noida, Uttar Pradesh 201314, India
| | - Soumya Pati
- Department of Life Sciences, School of Natural Sciences, Shiv Nadar University, Greater Noida, Uttar Pradesh 201314, India
| | - Chintam Narayana
- Department of Chemistry, School of Natural Sciences, Shiv Nadar University, Greater Noida, Uttar Pradesh 201314, India
| | - Rajagopal Ayana
- Department of Life Sciences, School of Natural Sciences, Shiv Nadar University, Greater Noida, Uttar Pradesh 201314, India
| | - Himani Kaushik
- Gene Regulation Laboratory, National Institute of Immunology, New Delhi 110067, India
| | - Raj Sah
- Special Centre for Molecular Medicine, Jawaharlal Nehru University, New Delhi 110067, India
| | - Swati Garg
- Department of Life Sciences, School of Natural Sciences, Shiv Nadar University, Greater Noida, Uttar Pradesh 201314, India
| | - Ashish Khanna
- Department of Chemistry, Institute of Science, Banaras Hindu University, Varanasi 221005, India
| | - Jyoti Kumari
- Department of Life Sciences, School of Natural Sciences, Shiv Nadar University, Greater Noida, Uttar Pradesh 201314, India
| | - Lalit Garg
- Gene Regulation Laboratory, National Institute of Immunology, New Delhi 110067, India
| | - Ram Sagar
- Department of Chemistry, Institute of Science, Banaras Hindu University, Varanasi 221005, India
| | - Shailja Singh
- Special Centre for Molecular Medicine, Jawaharlal Nehru University, New Delhi 110067, India
| |
Collapse
|
41
|
GPR55-mediated effects on brain microvascular endothelial cells and the blood-brain barrier. Neuroscience 2019; 414:88-98. [PMID: 31279825 DOI: 10.1016/j.neuroscience.2019.06.039] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2019] [Revised: 06/26/2019] [Accepted: 06/27/2019] [Indexed: 12/31/2022]
Abstract
GPR55, an atypical cannabinoid receptor activated by lysophosphatidylinositol (LPI) has been involved in various physiological and pathological processes. We examined the effect of GPR55 activation on rat brain microvascular endothelial cells (RBMVEC), an essential component of the blood-brain barrier (BBB). GPR55 was detected in RBMVEC by western blot and immunocytochemistry. Treatment of RBMVEC with LPI increased cytosolic Ca2+ concentration, [Ca2+]i, in a concentration-dependent manner; the effect was abolished by the GPR55 antagonist, ML-193. Repetitive application of LPI induced tachyphylaxis. LPI-induced increase in [Ca2+]i was not sensitive to U-73122, a phospholipase C inhibitor, but was abolished by the blockade of voltage-gated Ca2+ channels or in Ca2+-free saline, indicating that Ca2+ influx was involved in this response. LPI induced a biphasic change in RBMVEC membrane potential: a fast depolarization followed by a long-lasting hyperpolarization. The hyperpolarization phase was prevented by apamin and charibdotoxin, inhibitors of small- and intermediate-conductance Ca2+-activated K+ channels (KCa). Immunofluorescence studies indicate that LPI produced transient changes in tight and adherens junctions proteins and F-actin stress fibers. LPI decreased the electrical resistance of RBMVEC monolayer assessed with Electric Cell-Substrate Impedance Sensing (ECIS) in a dose-dependent manner. In vivo studies indicate that systemic administration of LPI increased the permeability of the BBB, assessed with Evans Blue method. Taken together, our results indicate that GPR55 activation modulates the function of endothelial cells of brain microvessels, produces a transient reduction in endothelial barrier function and increases BBB permeability.
Collapse
|
42
|
Hisano Y, Kono M, Cartier A, Engelbrecht E, Kano K, Kawakami K, Xiong Y, Piao W, Galvani S, Yanagida K, Kuo A, Ono Y, Ishida S, Aoki J, Proia RL, Bromberg JS, Inoue A, Hla T. Lysolipid receptor cross-talk regulates lymphatic endothelial junctions in lymph nodes. J Exp Med 2019; 216:1582-1598. [PMID: 31147448 PMCID: PMC6605750 DOI: 10.1084/jem.20181895] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2018] [Revised: 03/29/2019] [Accepted: 05/06/2019] [Indexed: 12/16/2022] Open
Abstract
Sphingosine 1-phosphate (S1P) and lysophosphatidic acid (LPA) activate G protein-coupled receptors (GPCRs) to regulate biological processes. Using a genome-wide CRISPR/dCas9-based GPCR signaling screen, LPAR1 was identified as an inducer of S1PR1/β-arrestin coupling while suppressing Gαi signaling. S1pr1 and Lpar1-positive lymphatic endothelial cells (LECs) of lymph nodes exhibit constitutive S1PR1/β-arrestin signaling, which was suppressed by LPAR1 antagonism. Pharmacological inhibition or genetic loss of function of Lpar1 reduced the frequency of punctate junctions at sinus-lining LECs. Ligand activation of transfected LPAR1 in endothelial cells remodeled junctions from continuous to punctate structures and increased transendothelial permeability. In addition, LPAR1 antagonism in mice increased lymph node retention of adoptively transferred lymphocytes. These data suggest that cross-talk between LPAR1 and S1PR1 promotes the porous junctional architecture of sinus-lining LECs, which enables efficient lymphocyte trafficking. Heterotypic inter-GPCR coupling may regulate complex cellular phenotypes in physiological milieu containing many GPCR ligands.
Collapse
Affiliation(s)
- Yu Hisano
- Vascular Biology Program, Boston Children's Hospital, Department of Surgery, Harvard Medical School, Boston, MA
| | - Mari Kono
- Genetics of Development and Disease Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD
| | - Andreane Cartier
- Vascular Biology Program, Boston Children's Hospital, Department of Surgery, Harvard Medical School, Boston, MA
| | - Eric Engelbrecht
- Vascular Biology Program, Boston Children's Hospital, Department of Surgery, Harvard Medical School, Boston, MA
| | - Kuniyuki Kano
- Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Japan
| | - Kouki Kawakami
- Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Japan
| | - Yanbao Xiong
- Department of Surgery, University of Maryland School of Medicine, Baltimore, MD
| | - Wenji Piao
- Department of Surgery, University of Maryland School of Medicine, Baltimore, MD
| | - Sylvain Galvani
- Vascular Biology Program, Boston Children's Hospital, Department of Surgery, Harvard Medical School, Boston, MA
| | - Keisuke Yanagida
- Vascular Biology Program, Boston Children's Hospital, Department of Surgery, Harvard Medical School, Boston, MA
| | - Andrew Kuo
- Vascular Biology Program, Boston Children's Hospital, Department of Surgery, Harvard Medical School, Boston, MA
| | - Yuki Ono
- Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Japan
| | - Satoru Ishida
- Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Japan
| | - Junken Aoki
- Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Japan
| | - Richard L Proia
- Genetics of Development and Disease Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD
| | - Jonathan S Bromberg
- Department of Surgery, University of Maryland School of Medicine, Baltimore, MD
| | - Asuka Inoue
- Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Japan
| | - Timothy Hla
- Vascular Biology Program, Boston Children's Hospital, Department of Surgery, Harvard Medical School, Boston, MA
| |
Collapse
|
43
|
ZEB1 insufficiency causes corneal endothelial cell state transition and altered cellular processing. PLoS One 2019; 14:e0218279. [PMID: 31194824 PMCID: PMC6564028 DOI: 10.1371/journal.pone.0218279] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2019] [Accepted: 05/29/2019] [Indexed: 12/13/2022] Open
Abstract
The zinc finger e-box binding homeobox 1 (ZEB1) transcription factor is a master regulator of the epithelial to mesenchymal transition (EMT), and of the reverse mesenchymal to epithelial transition (MET) processes. ZEB1 plays an integral role in mediating cell state transitions during cell lineage specification, wound healing and disease. EMT/MET are characterized by distinct changes in molecular and cellular phenotype that are generally context-independent. Posterior polymorphous corneal dystrophy (PPCD), associated with ZEB1 insufficiency, provides a new biological context in which to understand and evaluate the classic EMT/MET paradigm. PPCD is characterized by a cadherin-switch and transition to an epithelial-like transcriptomic and cellular phenotype, which we study in a cell-based model of PPCD generated using CRISPR-Cas9-mediated ZEB1 knockout in corneal endothelial cells (CEnCs). Transcriptomic and functional studies support the hypothesis that CEnC undergo a MET-like transition in PPCD, termed endothelial to epithelial transition (EnET), and lead to the conclusion that EnET may be considered a corollary to the classic EMT/MET paradigm.
Collapse
|
44
|
Advantages and shortcomings of cell-based electrical impedance measurements as a GPCR drug discovery tool. Biosens Bioelectron 2019; 137:33-44. [PMID: 31077988 DOI: 10.1016/j.bios.2019.04.041] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2019] [Revised: 04/05/2019] [Accepted: 04/20/2019] [Indexed: 12/13/2022]
Abstract
G Protein-Coupled Receptors (GPCRs) transduce extracellular signals and activate intracellular pathways, usually through activating associated G proteins. Due to their involvement in many human diseases, they are recognized worldwide as valuable drug targets. Many experimental approaches help identify small molecules that target GPCRs, including in vitro cell-based reporter assays and binding studies. Most cell-based assays use one signaling pathway or reporter as an assay readout. Moreover, they often require cell labeling or the integration of reporter systems. Over the last decades, cell-based electrical impedance biosensors have been explored for drug discovery. This label-free method holds many advantages over other cellular assays in GPCR research. The technology requires no cell manipulation and offers real-time kinetic measurements of receptor-mediated cellular changes. Instead of measuring the activity of a single reporter, the impedance readout includes information on multiple signaling events. This is beneficial when screening for ligands targeting orphan GPCRs since the signaling cascade(s) of the majority of these receptors are unknown. Due to its sensitivity, the method also applies to cellular models more relevant to disease, including patient-derived cell cultures. Despite its advantages, remaining issues regarding data comparability and interpretability has limited implementation of cell-based electrical impedance (CEI) in drug discovery. Future optimization must include both full exploitation of CEI response data using various ways of analysis as well as further exploration of its potential to detect biased activities early on in drug discovery. Here, we review the contribution of CEI technology to GPCR research, discuss its comparative benefits, and provide recommendations.
Collapse
|
45
|
Detection of apoptotic and live pre-osteoblast cell line using impedance-based biosensors with variable electrode design. Biosens Bioelectron 2019; 128:37-44. [PMID: 30616216 DOI: 10.1016/j.bios.2018.11.057] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2018] [Revised: 11/22/2018] [Accepted: 11/23/2018] [Indexed: 01/03/2023]
Abstract
Electrical impedance-based sensing of cell activity has become a powerful analytical tool that allows the monitoring of several relevant biological processes associated with cell evolution and morphology. In these types of biosensors, the electrode design has a direct impact on the sensitivity because it defines the capability of the biosensor to measure small changes in the impedance resulting from cell activities. Herein, impedance-based biosensors arrays with several configurations were successfully developed and used to study the impact of the electrode layout on the dynamics of cultured pre-osteoblast cells. The biosensor design was initially validated by measuring the effect of electrode design on the capacitance of a dielectric polymer (parylene) that mimics the dielectric characteristics of cell populations, results are shown in the Supplementary information section. Results from in vitro cell growth indicate that the optimized design of single electrodes with a diameter of 50 µm, are the most sensitive to cell motion whereas increasing the number of electrodes allows clear differentiation between living and dead cells after 3 h of inducing apoptosis. Apoptosis death was induced with Staurosporine, a chemical mediator of apoptosis in osteoblasts. These impedance results have been validated with optical imaging and flow cytometry analysis that were performed on parallel cultures. Frequency and electrolyte concentration effects are also discussed.
Collapse
|
46
|
Stolwijk JA, Skiba M, Kade C, Bernhardt G, Buschauer A, Hübner H, Gmeiner P, Wegener J. Increasing the throughput of label-free cell assays to study the activation of G-protein-coupled receptors by using a serial agonist exposure protocol. Integr Biol (Camb) 2019; 11:99-108. [PMID: 31083709 DOI: 10.1093/intbio/zyz010] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2019] [Revised: 03/12/2019] [Accepted: 05/03/2019] [Indexed: 12/25/2022]
Abstract
Label-free, holistic assays, monitoring, for example, the impedance of cells on electrodes, are gaining increasing popularity in the evaluation of G-protein-coupled receptor (GPCR) ligands. It is the strength of these approaches to provide the integrated cellular response non-invasively, highly automated and with a device-dependent time resolution down to several milliseconds. With an increasing number of samples to be studied in parallel, the available time resolution is, however, reduced and the cost for the disposable sensor arrays may become limiting. Inspired by protocols from organ pharmacology, we investigated a simple serial agonist addition assay that circumvents these limitations in impedance-based cellular assays. Using a serial addition of increasing concentrations of a GPCR agonist while continuously monitoring the sample's impedance, we were able to establish a full concentration-response curve for the endogenous agonist histamine on a single layer of U-373 MG cells endogenously expressing the histamine 1 receptor (H1R). This approach is validated with respect to conventional, parallel agonist addition protocols and studies using H1R antagonists such as mepyramine. Applicability of the serial agonist addition assay was shown for other GPCRs known for their signaling via one of the canonical G-protein pathways, Gq, Gi/0 or Gs as well. The serial agonist addition protocol has the potential to further strengthen the output of label-free analysis of GPCR activation.
Collapse
Affiliation(s)
- J A Stolwijk
- Institute of Analytical Chemistry, Chemo- and Biosensors, University of Regensburg, Regensburg, Germany
| | - M Skiba
- Institute of Analytical Chemistry, Chemo- and Biosensors, University of Regensburg, Regensburg, Germany
| | - C Kade
- Institute of Analytical Chemistry, Chemo- and Biosensors, University of Regensburg, Regensburg, Germany
| | - G Bernhardt
- Institute of Pharmacy, University of Regensburg, Regensburg, Germany
| | - A Buschauer
- Institute of Pharmacy, University of Regensburg, Regensburg, Germany
| | - H Hübner
- Department of Chemistry and Pharmacy, Friedrich-Alexander University Erlangen-Nürnberg FAU, Erlangen, Germany
| | - P Gmeiner
- Department of Chemistry and Pharmacy, Friedrich-Alexander University Erlangen-Nürnberg FAU, Erlangen, Germany
| | - J Wegener
- Institute of Analytical Chemistry, Chemo- and Biosensors, University of Regensburg, Regensburg, Germany
- Fraunhofer Research Institution for Microsystems and Solid State Technologies EMFT, Munich, Germany
| |
Collapse
|
47
|
Grimsey NJ, Lin Y, Narala R, Rada CC, Mejia-Pena H, Trejo J. G protein-coupled receptors activate p38 MAPK via a non-canonical TAB1-TAB2- and TAB1-TAB3-dependent pathway in endothelial cells. J Biol Chem 2019; 294:5867-5878. [PMID: 30760523 DOI: 10.1074/jbc.ra119.007495] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2019] [Revised: 02/07/2019] [Indexed: 01/03/2023] Open
Abstract
Endothelial dysfunction is induced by inflammatory mediators including multiple G protein-coupled receptor (GPCR) agonists. However, the GPCR signaling pathways that promote endothelial dysfunction are incompletely understood. We previously showed that thrombin promotes endothelial barrier disruption through autophosphorylation and activation of p38 mitogen-activated protein kinase (MAPK) via a non-canonical transforming growth factor-β-activated protein kinase-1-binding protein-1 (TAB1) and TAB2-dependent pathway rather than the canonical three-tiered kinase cascade. Here, we sought to determine whether other GPCR agonists stimulate p38 MAPK activation via this non-canonical pathway in human endothelial cells derived from different vascular beds. Using primary human umbilical vein endothelial cells (HUVECs), HUVEC-derived EA.hy926 cells, and human dermal microvascular endothelial cells (HDMECs), we found that both non-canonical and canonical p38 activation pathways components are expressed in these various endothelial cell types, including TAB3, a structurally-related TAB2 homolog. Moreover, multiple GPCRs agonists, including thrombin, histamine, prostaglandin E2, and ADP, stimulated robust p38 autophosphorylation, whereas phosphorylation of the upstream MAPKs MAP kinase kinase 3 (MKK3) and MKK6, was virtually undetectable, indicating that non-canonical p38 activation may exist for other GPCRs. Indeed, in EA.hy926 cells, thrombin- and histamine-stimulated p38 activation depended on TAB1-TAB2, whereas in primary HUVECs, both TAB1-TAB2 and TAB1-TAB3 were required for p38 activation. In HDMECs, thrombin-induced p38 activation depended on TAB1-TAB3, but histamine-induced p38 activation required TAB1-TAB2. Moreover, thrombin- and histamine-stimulated interleukin-6 production required both TAB1-TAB2 and TAB1-TAB3 in HUVEC. We conclude that multiple GPCR agonists utilize non-canonical TAB1-TAB2 and TAB1-TAB3-dependent p38 activation to promote endothelial inflammatory responses.
Collapse
Affiliation(s)
- Neil J Grimsey
- From the Department of Pharmacology, University of California, San Diego, La Jolla, California 92093
| | - Ying Lin
- From the Department of Pharmacology, University of California, San Diego, La Jolla, California 92093
| | - Rachan Narala
- From the Department of Pharmacology, University of California, San Diego, La Jolla, California 92093
| | - Cara C Rada
- From the Department of Pharmacology, University of California, San Diego, La Jolla, California 92093; Biomedical Sciences Graduate Program, School of Medicine, University of California, San Diego, La Jolla, California 92093
| | - Hilda Mejia-Pena
- From the Department of Pharmacology, University of California, San Diego, La Jolla, California 92093
| | - JoAnn Trejo
- From the Department of Pharmacology, University of California, San Diego, La Jolla, California 92093.
| |
Collapse
|
48
|
Investigating the Effects of Stove Emissions on Ocular and Cancer Cells. Sci Rep 2019; 9:1870. [PMID: 30755694 PMCID: PMC6372759 DOI: 10.1038/s41598-019-38803-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2018] [Accepted: 12/20/2018] [Indexed: 12/19/2022] Open
Abstract
More than a third of the world’s population relies on solid fuels for cooking and heating, with major health consequences. Although solid fuel combustion emissions are known to increase the prevalence of illnesses such as chronic obstructive pulmonary disease and lung cancer, however, their effect on the eyes is underexplored. This study assesses the acute toxicity of solid fuel combustion emissions on healthy ocular cells and a cancer cell line. Three healthy ocular cell lines (corneal, lens, and retinal epithelial cells) and a cancer cell line (Chinese hamster ovary cells) were exposed to liquid and gas phase emissions from applewood and coal combustion. Following the exposure, real-time cell attachment behavior was monitored for at least 120 hours with electrical cell impedance spectroscopy. The viability of the cells, amount of apoptotic cells, and generation of reactive oxygen species (ROS) were quantified with MTT, ApoTox-Glo, and ROS-Glo H2O2 assays, respectively. The results showed that coal emissions compromised the viability of ocular cells more than applewood emissions. Interestingly, the cancer cells, although their viability was not compromised, generated 1.7 to 2.7 times more ROS than healthy cells. This acute exposure study provides compelling proof that biomass combustion emissions compromise the viability of ocular cells and increase ROS generation. The increased ROS generation was fatal for ocular cells, but it promoted the growth of cancer cells.
Collapse
|
49
|
Characterization of Host and Bacterial Contributions to Lung Barrier Dysfunction Following Co-infection with 2009 Pandemic Influenza and Methicillin Resistant Staphylococcus aureus. Viruses 2019; 11:v11020116. [PMID: 30699912 PMCID: PMC6409999 DOI: 10.3390/v11020116] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2019] [Accepted: 01/26/2019] [Indexed: 12/12/2022] Open
Abstract
Influenza viruses are a threat to global public health resulting in ~500,000 deaths each year. Despite an intensive vaccination program, influenza infections remain a recurrent, yet unsolved public health problem. Secondary bacterial infections frequently complicate influenza infections during seasonal outbreaks and pandemics, resulting in increased morbidity and mortality. Staphylococcus aureus, including methicillin-resistant S. aureus (MRSA), is frequently associated with these co-infections, including the 2009 influenza pandemic. Damage to alveolar epithelium is a major contributor to severe influenza-bacterial co-infections and can result in gas exchange abnormalities, fluid leakage, and respiratory insufficiency. These deleterious manifestations likely involve both pathogen- and host-mediated mechanisms. However, there is a paucity of information regarding the mechanisms (pathogen- and/or host-mediated) underlying influenza-bacterial co-infection pathogenesis. To address this, we characterized the contributions of viral-, bacterial-, and host-mediated factors to the altered structure and function of alveolar epithelial cells during co-infection with a focus on the 2009 pandemic influenza (pdm2009) and MRSA. Here, we characterized pdm2009 and MRSA replication kinetics, temporal host kinome responses, modulation of MRSA virulence factors, and disruption of alveolar barrier integrity in response to pdm2009-MRSA co-infection. Our results suggest that alveolar barrier disruption during co-infection is mediated primarily through host response dysregulation, resulting in loss of alveolar barrier integrity.
Collapse
|
50
|
Zhou W, Graham K, Lucendo-Villarin B, Flint O, Hay DC, Bagnaninchi P. Combining stem cell-derived hepatocytes with impedance sensing to better predict human drug toxicity. Expert Opin Drug Metab Toxicol 2018; 15:77-83. [PMID: 30572740 DOI: 10.1080/17425255.2019.1558208] [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: 01/09/2023]
Abstract
Background: The liver plays a central role in human drug metabolism. To model drug metabolism, the major cell type of the liver, the hepatocyte, is commonly used. Hepatocytes can be derived from human and animal sources, including pluripotent stem cells. Cell-based models have shown promise in modeling human drug exposure. The assays used in those studies are normally 'snap-shot' in nature, and do not provide the complete picture of human drug exposure. Research design and methods: In this study, we employ stem cell-derived hepatocytes and impedance sensing to model human drug toxicity. This impedance-based stem cell assay reports hepatotoxicity in real time after treatment with compounds provided by industry. Results: Using electric cell-substrate impedance Sensing (ECIS), we were able to accurately measure drug toxicity post-drug exposure in real time and more quickly than gold standard biochemical assays. Conclusions: ECIS is robust and non-destructive methodology capable of monitoring human drug exposure with superior performance to current gold standard 'snapshot' assays. We believe that the methodology presented within this article could prove valuable in the quest to better predict off-target effects of drugs in humans.
Collapse
Affiliation(s)
- Wenli Zhou
- a Department of Medical Oncology , Changzheng Hospital, Navy medical University , Shanghai , China
| | - Karen Graham
- b MRC Centre for Regenerative Medicine, 5 Little France Drive , University of Edinburgh , Edinburgh , UK
| | - Baltasar Lucendo-Villarin
- b MRC Centre for Regenerative Medicine, 5 Little France Drive , University of Edinburgh , Edinburgh , UK
| | - Oliver Flint
- b MRC Centre for Regenerative Medicine, 5 Little France Drive , University of Edinburgh , Edinburgh , UK
| | - David C Hay
- b MRC Centre for Regenerative Medicine, 5 Little France Drive , University of Edinburgh , Edinburgh , UK
| | - Pierre Bagnaninchi
- b MRC Centre for Regenerative Medicine, 5 Little France Drive , University of Edinburgh , Edinburgh , UK
| |
Collapse
|