1
|
Lino M, Persson H, Paknahad M, Ugodnikov A, Farhang Ghahremani M, Takeuchi LE, Chebotarev O, Horst C, Simmons CA. A pumpless microfluidic co-culture system to model the effects of shear flow on biological barriers. LAB ON A CHIP 2025; 25:1489-1501. [PMID: 39925127 DOI: 10.1039/d4lc00835a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/11/2025]
Abstract
Biological barriers formed by the endothelium and epithelium regulate nutrient exchange, disease development, and drug delivery. Organ-on-chip (OOC) systems effectively model these barriers by incorporating key biophysical cues like microscale dimensions, co-culture, and fluid flow-induced shear stress. The majority of microfluidic OOC platforms, however, require syringe and pump systems which are hindered by several limitations, including large footprints, elaborate designs, long setup times, and a high rate of failure (contamination, leakage, etc.). Here we describe VitroFlo, a pump-free microfluidic device designed for in vitro biological barrier modeling with 12 independent co-culture modules that can be simultaneously subjected to tunable, unidirectional flow with physiological shear stresses ranging from 0.01-10 dyn/cm2. We demonstrate application of the device to model vascular endothelial, blood-brain, and intestinal epithelial barriers, and confirm shear stress-dependent cell alignment, tight junction protein expression, barrier maturation, permeability, and paracrine signaling between co-cultured cells. The VitroFlo platform enables scalable and cost-effective modeling of physiological barriers to facilitate the translation of findings from in vitro systems to preclinical models.
Collapse
Affiliation(s)
- Marsel Lino
- Department of Mechanical and Industrial Engineering, University of Toronto, 5 King's College Road, Toronto, ON, Canada
- Translational Biology and Engineering Program, Ted Rogers Centre for Heart Research, University of Toronto, 661 University Avenue, Toronto, ON, Canada.
| | - Henrik Persson
- Department of Mechanical and Industrial Engineering, University of Toronto, 5 King's College Road, Toronto, ON, Canada
- Translational Biology and Engineering Program, Ted Rogers Centre for Heart Research, University of Toronto, 661 University Avenue, Toronto, ON, Canada.
- Institute of Biomedical Engineering, University of Toronto, 164 College Street, Toronto, ON, Canada
| | - Mohammad Paknahad
- Department of Mechanical and Industrial Engineering, University of Toronto, 5 King's College Road, Toronto, ON, Canada
- Translational Biology and Engineering Program, Ted Rogers Centre for Heart Research, University of Toronto, 661 University Avenue, Toronto, ON, Canada.
| | - Alisa Ugodnikov
- Translational Biology and Engineering Program, Ted Rogers Centre for Heart Research, University of Toronto, 661 University Avenue, Toronto, ON, Canada.
- Institute of Biomedical Engineering, University of Toronto, 164 College Street, Toronto, ON, Canada
| | - Morvarid Farhang Ghahremani
- Department of Mechanical and Industrial Engineering, University of Toronto, 5 King's College Road, Toronto, ON, Canada
- Translational Biology and Engineering Program, Ted Rogers Centre for Heart Research, University of Toronto, 661 University Avenue, Toronto, ON, Canada.
| | - Lily E Takeuchi
- Translational Biology and Engineering Program, Ted Rogers Centre for Heart Research, University of Toronto, 661 University Avenue, Toronto, ON, Canada.
- Institute of Biomedical Engineering, University of Toronto, 164 College Street, Toronto, ON, Canada
| | - Oleg Chebotarev
- Department of Mechanical and Industrial Engineering, University of Toronto, 5 King's College Road, Toronto, ON, Canada
- Translational Biology and Engineering Program, Ted Rogers Centre for Heart Research, University of Toronto, 661 University Avenue, Toronto, ON, Canada.
| | - Caleb Horst
- CellScale Biomaterials Testing, Waterloo, ON, Canada
| | - Craig A Simmons
- Department of Mechanical and Industrial Engineering, University of Toronto, 5 King's College Road, Toronto, ON, Canada
- Translational Biology and Engineering Program, Ted Rogers Centre for Heart Research, University of Toronto, 661 University Avenue, Toronto, ON, Canada.
- Institute of Biomedical Engineering, University of Toronto, 164 College Street, Toronto, ON, Canada
| |
Collapse
|
2
|
Werner S, Pamies D, Zurich MG, Suter-Dick L. Hepatic and extra-hepatic metabolism of propylene glycol ethers in the context of central nervous system toxicity. Toxicology 2025; 512:154081. [PMID: 39929342 DOI: 10.1016/j.tox.2025.154081] [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: 01/09/2025] [Revised: 02/05/2025] [Accepted: 02/06/2025] [Indexed: 02/14/2025]
Abstract
Propylene glycol ethers (PGEs) are mixtures of an α-isomer and a β-isomer (β-PGE) that is oxidized via alcohol dehydrogenase (ADH) and aldehyde dehydrogenase (ALDH) to potentially neurotoxic alkoxy propionic acids (β-metabolites). While the liver is the primary organ for ADH- and ALDH-mediated metabolism, the contribution to the metabolism of β-PGEs by the blood-brain barrier (BBB) and the brain remains unknown. Here, we aimed to assess the neurotoxic potential of PGEs after systemic exposure by (1) comparing 3D HepaRG and human liver subcellular fraction (S9) for the in vitro determination of the kinetics of hepatic metabolism for β-PGEs, (2) evaluating the BBB-permeability of PGEs and β-metabolites, (3) determining the presence of ADH1 and ALDH2 and the extent of metabolization of β-PGEs in the BBB and brain. The results show that 3D HepaRG and S9 served as competent systems to estimate the enzymatic kinetic (clearance) for β-metabolite formation. We observed that PGEs and the β-metabolites could cross the BBB, based on their permeance across a cellular barrier consisting of the hCMEC/D3 cell line. Metabolic enzymes were not exclusive to the liver, as expression of ADH1 and ALDH2 was demonstrated using RT-qPCR, Western blot, and immunostainings in the BBB in vitro models and in BrainSpheres. Furthermore, LC-MS/MS quantification of the β-metabolites in all in vitro models revealed that 3D HepaRG had a similar metabolic capacity to primary human hepatocytes and that the amount of β-metabolite formed per protein in the BBB was approximately 10-30 % of that in the liver. We also demonstrated active metabolism in the BrainSpheres. In conclusion, the hepatic in vitro models provided data that will help to refine toxicokinetic models and predict internal exposures, thereby supporting the risk assessment of PGEs. In addition, the high permeance of the PGEs and the β-metabolites across the BBB increases the plausibility of neurotoxicity upon systemic exposure. This is further supported by the presence of active ADH1 and ALDH2 enzymes in the BBB in vitro systems and in BrainSpheres, suggesting metabolite formation in the central nervous system. Hence, we suggest that BBB-permeance and extra-hepatic metabolism of the β-PGEs may contribute to the neurotoxicity of PGEs.
Collapse
Affiliation(s)
- Sophie Werner
- University of Applied Sciences and Arts Northwestern Switzerland, School of Life Sciences, Muttenz, Switzerland; Department of Pharmaceutical Sciences, University of Basel, Basel, Switzerland; Swiss Centre for Applied Human Toxicology (SCAHT), Basel, Switzerland.
| | - David Pamies
- Swiss Centre for Applied Human Toxicology (SCAHT), Basel, Switzerland; Department of Biomedical Science, University of Lausanne, Lausanne, Switzerland.
| | - Marie-Gabrielle Zurich
- Swiss Centre for Applied Human Toxicology (SCAHT), Basel, Switzerland; Department of Biomedical Science, University of Lausanne, Lausanne, Switzerland.
| | - Laura Suter-Dick
- University of Applied Sciences and Arts Northwestern Switzerland, School of Life Sciences, Muttenz, Switzerland; Swiss Centre for Applied Human Toxicology (SCAHT), Basel, Switzerland.
| |
Collapse
|
3
|
Chavarria D, Georges KA, O’Grady BJ, Hassan KK, Lippmann ES. Modular cone-and-plate device for mechanofluidic assays in Transwell inserts. Front Bioeng Biotechnol 2025; 13:1494553. [PMID: 39931136 PMCID: PMC11807968 DOI: 10.3389/fbioe.2025.1494553] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Accepted: 01/06/2025] [Indexed: 02/13/2025] Open
Abstract
In this work, we present a cost effective and open-source modular cone-and-plate (MoCAP) device that incorporates shear stress in the popular Transwell® insert system. This system acts as a lid that incorporates flow into 24-well Transwell® inserts while preserving the ability to conduct molecular profiling assays. Moreover, the MoCAP device can be rapidly reconfigured to test multiple shear stress profiles within a single device. To demonstrate the utility of the MoCAP, we conducted select assays on several different brain microvascular endothelial cell (BMEC) lines that comprise models of the blood-brain barrier (BBB), since shear stress can play an important role in BBB function. Our results characterize how shear stress modulates passive barrier function and GLUT1 expression across the different BMEC lines. Overall, we anticipate this low cost mechanofluidic device will be useful to the mechanobiology community.
Collapse
Affiliation(s)
- Daniel Chavarria
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN, United States
| | - Kissamy A. Georges
- Department of Bioengineering, University of Massachusetts Dartmouth, Dartmouth, MA, United States
| | - Brian J. O’Grady
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN, United States
| | - Khalid K. Hassan
- School for Science and Math at Vanderbilt, Vanderbilt University, Nashville, TN, United States
| | - Ethan S. Lippmann
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN, United States
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, United States
- Vanderbilt Brain Institute, Vanderbilt University, Nashville, TN, United States
- Department of Neurology, Vanderbilt University Medical Center, Nashville, TN, United States
- Interdisciplinary Materials Science Program, Vanderbilt University, Nashville, TN, United States
- Vanderbilt Memory and Alzheimer’s Center, Vanderbilt University Medical Center, Nashville, TN, United States
| |
Collapse
|
4
|
Sun J, Song S. Advances in modeling permeability and selectivity of the blood-brain barrier using microfluidics. MICROFLUIDICS AND NANOFLUIDICS 2024; 28:44. [PMID: 39781566 PMCID: PMC11709447 DOI: 10.1007/s10404-024-02741-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Accepted: 06/15/2024] [Indexed: 01/12/2025]
Abstract
The blood-brain barrier (BBB) protects the brain by actively allowing the entry of ions and nutrients while limiting the passage of from toxins and pathogens. A healthy BBB has low permeability and high selectivity to maintain normal brain functions. Increased BBB permeability can result from neurological diseases and traumatic injuries. Modern engineering technologies such as microfluidics and fabrication techniques have advanced the development of BBB models to simulate the basic functions of BBB. However, the intrinsic BBB properties are difficult to replicate. Existing in vitro BBB models demonstrate inconsistent BBB permeability and selectivity due to variations in microfluidic design, cell types and arrangement, expression of tight junction (TJ) proteins, and use of shear stress. Specifically, microfluidic designs have flow channels of different sizes, complexity, topology, and modular structure. Different cell types are selected to mimic various physiological conditions. These factors make it challenging to compare results obtained using different experimental setups. This paper highlights key factors that play important roles in influencing microfluidic models and discusses how these factors contribute to permeability and selectivity of the BBB models.
Collapse
Affiliation(s)
- Jindi Sun
- Department of Biomedical Engineering, The University of Arizona, 1200 E University Blvd, Tucson 85721, Arizona, USA
| | - Shang Song
- Department of Biomedical Engineering, The University of Arizona, 1200 E University Blvd, Tucson 85721, Arizona, USA
- Departments of Neuroscience GIDP, Materials Science and Engineering, and BIO5 Institute, The University of Arizona, 1200 E University Blvd, Tucson 85721, Arizona, USA
| |
Collapse
|
5
|
Mantecón-Oria M, Rivero MJ, Diban N, Urtiaga A. On the quest of reliable 3D dynamic in vitro blood-brain barrier models using polymer hollow fiber membranes: Pitfalls, progress, and future perspectives. Front Bioeng Biotechnol 2022; 10:1056162. [PMID: 36483778 PMCID: PMC9723404 DOI: 10.3389/fbioe.2022.1056162] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Accepted: 11/07/2022] [Indexed: 09/10/2024] Open
Abstract
With the increasing concern of neurodegenerative diseases, the development of new therapies and effective pharmaceuticals targeted to central nervous system (CNS) illnesses is crucial for ensuring social and economic sustainability in an ageing world. Unfortunately, many promising treatments at the initial stages of the pharmaceutical development process, that is at the in vitro screening stages, do not finally show the expected results at the clinical level due to their inability to cross the human blood-brain barrier (BBB), highlighting the inefficiency of in vitro BBB models to recapitulate the real functionality of the human BBB. In the last decades research has focused on the development of in vitro BBB models from basic 2D monolayer cultures to 3D cell co-cultures employing different system configurations. Particularly, the use of polymeric hollow fiber membranes (HFs) as scaffolds plays a key role in perfusing 3D dynamic in vitro BBB (DIV-BBB) models. Their incorporation into a perfusion bioreactor system may potentially enhance the vascularization and oxygenation of 3D cell cultures improving cell communication and the exchange of nutrients and metabolites through the microporous membranes. The quest for developing a benchmark 3D dynamic in vitro blood brain barrier model requires the critical assessment of the different aspects that limits the technology. This article will focus on identifying the advantages and main limitations of the HFs in terms of polymer materials, microscopic porous morphology, and other practical issues that play an important role to adequately mimic the physiological environment and recapitulate BBB architecture. Based on this study, we consider that future strategic advances of this technology to become fully implemented as a gold standard DIV-BBB model will require the exploration of novel polymers and/or composite materials, and the optimization of the morphology of the membranes towards thinner HFs (<50 μm) with higher porosities and surface pore sizes of 1-2 µm to facilitate the intercommunication via regulatory factors between the cell co-culture models of the BBB.
Collapse
Affiliation(s)
- Marián Mantecón-Oria
- Departamento de Ingenierias Química y Biomolecular, Universidad de Cantabria, Santander, Spain
- Instituto Marqués de Valdecilla (IDIVAL), Santander, Spain
| | - María J. Rivero
- Departamento de Ingenierias Química y Biomolecular, Universidad de Cantabria, Santander, Spain
| | - Nazely Diban
- Departamento de Ingenierias Química y Biomolecular, Universidad de Cantabria, Santander, Spain
- Instituto Marqués de Valdecilla (IDIVAL), Santander, Spain
| | - Ane Urtiaga
- Departamento de Ingenierias Química y Biomolecular, Universidad de Cantabria, Santander, Spain
- Instituto Marqués de Valdecilla (IDIVAL), Santander, Spain
| |
Collapse
|
6
|
Harding IC, O'Hare NR, Vigliotti M, Caraballo A, Lee CI, Millican K, Herman IM, Ebong EE. Developing a transwell millifluidic device for studying blood-brain barrier endothelium. LAB ON A CHIP 2022; 22:4603-4620. [PMID: 36326069 PMCID: PMC11416711 DOI: 10.1039/d2lc00657j] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
Blood-brain barrier (BBB) endothelial cell (EC) function depends on flow conditions and on supportive cells, like pericytes and astrocytes, which have been shown to be both beneficial and detrimental for brain EC function. Most studies investigating BBB EC function lack physiological relevance, using sub-physiological shear stress magnitudes and/or omitting pericytes and astrocytes. In this study, we developed a millifluidic device compatible with standard transwell inserts to investigate BBB function. In contrast to standard polydimethylsiloxane (PDMS) microfluidic devices, this model allows for easy, reproducible shear stress exposure without common limitations of PDMS devices such as inadequate nutrient diffusion and air bubble formation. In no-flow conditions, we first used the device to examine the impact of primary human pericytes and astrocytes on human brain microvascular EC (HBMEC) barrier integrity. Astrocytes, pericytes, and a 1-to-1 ratio of both cell types increased HBMEC barrier integrity via reduced 3 and 40 kDa fluorescent dextran permeability and increased claudin-5 expression. There were differing levels of low 3 kDa permeability in HBMEC-pericyte, HBMEC-astrocyte, and HBMEC-astrocyte-pericyte co-cultures, while levels of low 40 kDa permeability were consistent across co-cultures. The 3 kDa findings suggest that pericytes provide more barrier support to the BBB model compared to astrocytes, although both supportive cell types are permeability reducers. Incorporation of 24-hour 12 dynes per cm2 flow significantly reduced dextran permeability in HBMEC monolayers, but not in the tri-culture model. These results indicate that tri-culture may exert more pronounced impact on overall BBB permeability than flow exposure. In both cases, monolayer and tri-culture, flow exposure interestingly reduced HBMEC expression of both claudin-5 and occludin. ZO-1 expression, and localization at cell-cell junctions increased in the tri-culture but exhibited no apparent change in the HBMEC monolayer. Under flow conditions, we also observed HBMEC alignment in the tri-culture but not in HBMEC monolayers, indicating supportive cells and flow are both essential to observe brain EC alignment in vitro. Collectively, these results support the necessity of physiologically relevant, multicellular BBB models when investigating BBB EC function. Consideration of the roles of shear stress and supportive cells within the BBB is critical for elucidating the physiology of the neurovascular unit.
Collapse
Affiliation(s)
- Ian C Harding
- Department of Bioengineering, Northeastern University, Boston, MA, USA
| | - Nicholas R O'Hare
- Department of Chemical Engineering, Northeastern University, 360 Huntington Avenue, 129 Interdisciplinary Science and Engineering Complex, Boston, MA, 02115, USA.
| | - Mark Vigliotti
- Department of Chemical Engineering, Northeastern University, 360 Huntington Avenue, 129 Interdisciplinary Science and Engineering Complex, Boston, MA, 02115, USA.
| | - Alex Caraballo
- Department of Chemical Engineering, Northeastern University, 360 Huntington Avenue, 129 Interdisciplinary Science and Engineering Complex, Boston, MA, 02115, USA.
| | - Claire I Lee
- Department of Bioengineering, Northeastern University, Boston, MA, USA
| | - Karina Millican
- Department of Bioengineering, Northeastern University, Boston, MA, USA
| | - Ira M Herman
- Department of Developmental, Molecular, and Chemical Biology, Tufts School of Graduate Biomedical Sciences, Boston, MA, USA
- Center for Innovations in Wound Healing Research, Tufts University School of Medicine, Boston, MA, USA
| | - Eno E Ebong
- Department of Bioengineering, Northeastern University, Boston, MA, USA
- Department of Chemical Engineering, Northeastern University, 360 Huntington Avenue, 129 Interdisciplinary Science and Engineering Complex, Boston, MA, 02115, USA.
- Department of Neuroscience, Albert Einstein College of Medicine, New York, NY, USA
| |
Collapse
|
7
|
Schreiner TG, Creangă-Murariu I, Tamba BI, Lucanu N, Popescu BO. In Vitro Modeling of the Blood–Brain Barrier for the Study of Physiological Conditions and Alzheimer’s Disease. Biomolecules 2022; 12:biom12081136. [PMID: 36009030 PMCID: PMC9405874 DOI: 10.3390/biom12081136] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Revised: 08/09/2022] [Accepted: 08/16/2022] [Indexed: 11/16/2022] Open
Abstract
The blood–brain barrier (BBB) is an essential structure for the maintenance of brain homeostasis. Alterations to the BBB are linked with a myriad of pathological conditions and play a significant role in the onset and evolution of neurodegenerative diseases, including Alzheimer’s disease. Thus, a deeper understanding of the BBB’s structure and function is mandatory for a better knowledge of neurodegenerative disorders and the development of effective therapies. Because studying the BBB in vivo imposes overwhelming difficulties, the in vitro approach remains the main possible way of research. With many in vitro BBB models having been developed over the last years, the main aim of this review is to systematically present the most relevant designs used in neurological research. In the first part of the article, the physiological and structural–functional parameters of the human BBB are detailed. Subsequently, available BBB models are presented in a comparative approach, highlighting their advantages and limitations. Finally, the new perspectives related to the study of Alzheimer’s disease with the help of novel devices that mimic the in vivo human BBB milieu gives the paper significant originality.
Collapse
Affiliation(s)
- Thomas Gabriel Schreiner
- Faculty of Medicine, “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania
- Department of Neurology, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania
- Department of Electrical Measurements and Materials, Faculty of Electrical Engineering and Information Technology, Gheorghe Asachi Technical University of Iasi, 21-23 Professor Dimitrie Mangeron Blvd., 700050 Iasi, Romania
- Correspondence:
| | - Ioana Creangă-Murariu
- Advanced Research and Development Center for Experimental Medicine (CEMEX), “Grigore T. Popa” University of Medicine and Pharmacy, Universitatii Str., No. 16, 700155 Iasi, Romania
| | - Bogdan Ionel Tamba
- Advanced Research and Development Center for Experimental Medicine (CEMEX), “Grigore T. Popa” University of Medicine and Pharmacy, Universitatii Str., No. 16, 700155 Iasi, Romania
| | - Nicolae Lucanu
- Department of Applied Electronics and Intelligent Systems, Faculty of Electronics, Telecommunications and Information Technology, Gheorghe Asachi Technical University of Iasi, 21-23 Professor Dimitrie Mangeron Blvd., 700050 Iasi, Romania
| | - Bogdan Ovidiu Popescu
- Faculty of Medicine, “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania
- Neurology Department, Colentina Clinical Hospital, 020125 Bucharest, Romania
- Laboratory of Cell Biology, Neurosciences and Experimental Myology, “Victor Babes” National Institute of Pathology, 050096 Bucharest, Romania
| |
Collapse
|
8
|
Choublier N, Taghi M, Menet MC, Le Gall M, Bruce J, Chafey P, Guillonneau F, Moreau A, Denizot C, Parmentier Y, Nakib S, Borderie D, Bouzinba-Segard H, Couraud PO, Bourdoulous S, Declèves X. Exposure of human cerebral microvascular endothelial cells hCMEC/D3 to laminar shear stress induces vascular protective responses. Fluids Barriers CNS 2022; 19:41. [PMID: 35658915 PMCID: PMC9164338 DOI: 10.1186/s12987-022-00344-w] [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: 12/20/2021] [Accepted: 05/23/2022] [Indexed: 01/01/2023] Open
Abstract
Endothelial cells (ECs) are constantly submitted in vivo to hemodynamical forces derived from the blood circulation, including shear stress (SS). ECs are able to detect SS and consequently adapt their phenotype, thus affecting many endothelial functions. If a plethora of shear stress-regulated molecular networks have been described in peripheral ECs, less is known about the molecular responses of microvascular brain ECs which constitute the blood-brain barrier (BBB). In this work, we investigated the response of human cerebral microvascular ECs to laminar physiological shear stress using the well characterized hCMEC/D3 cell line. Interestingly, we showed that hCMEC/D3 cells responded to shear stress by aligning perpendicularly to the flow direction, contrary to peripheral endothelial cells which aligned in the flow direction. Whole proteomic profiles were compared between hCMEC/D3 cells cultured either in static condition or under 5 or 10 dyn.cm-2 SS for 3 days. 3592 proteins were identified and expression levels were significantly affected for 3% of them upon both SS conditions. Pathway analyses were performed which revealed that most proteins overexpressed by SS refer to the antioxidant defense, probably mediated by activation of the NRF2 transcriptional factor. Regarding down-regulated proteins, most of them participate to the pro-inflammatory response, cell motility and proliferation. These findings confirm the induction of EC quiescence by laminar physiological SS and reveal a strong protective effect of SS on hCMEC/D3 cells, suggesting a similar effect on the BBB. Our results also showed that SS did not significantly increase expression levels nor did it affect the localization of junctional proteins and did not afect either the functional activity of several ABC transporters (P-glycoprotein and MRPs). This work provides new insights on the response of microvascular brain ECs to SS and on the importance of SS for optimizing in vitro BBB models.
Collapse
Affiliation(s)
- Nina Choublier
- INSERM, Optimisation Thérapeutique en Neuropsychopharmacologie, Université Paris Cité, 75006, Paris, France.
| | - Meryam Taghi
- INSERM, Optimisation Thérapeutique en Neuropsychopharmacologie, Université Paris Cité, 75006, Paris, France
| | - Marie-Claude Menet
- Institut de Chimie Physique, CNRS, Université Paris-Saclay, 91405, Orsay, France
| | - Morgane Le Gall
- 3P5 Proteom'IC Facility, Institut Cochin, INSERM, CNRS, Université de Paris, F-75014, Paris, France
| | - Johanna Bruce
- 3P5 Proteom'IC Facility, Institut Cochin, INSERM, CNRS, Université de Paris, F-75014, Paris, France
| | - Philippe Chafey
- 3P5 Proteom'IC Facility, Institut Cochin, INSERM, CNRS, Université de Paris, F-75014, Paris, France
| | - François Guillonneau
- 3P5 Proteom'IC Facility, Institut Cochin, INSERM, CNRS, Université de Paris, F-75014, Paris, France
| | | | | | | | - Samir Nakib
- Service de Biochimie, Hôpital Cochin, Assistance Publique-Hôpitaux de Paris, 75014, Paris, France
| | - Didier Borderie
- Service de Biochimie, Hôpital Cochin, Assistance Publique-Hôpitaux de Paris, 75014, Paris, France
| | - Haniaa Bouzinba-Segard
- CNRS, INSERM, Institut Cochin, Inserm, CNRS, Université Paris Cité, 75014, Paris, France
| | - Pierre-Olivier Couraud
- CNRS, INSERM, Institut Cochin, Inserm, CNRS, Université Paris Cité, 75014, Paris, France
| | - Sandrine Bourdoulous
- CNRS, INSERM, Institut Cochin, Inserm, CNRS, Université Paris Cité, 75014, Paris, France
| | - Xavier Declèves
- INSERM, Optimisation Thérapeutique en Neuropsychopharmacologie, Université Paris Cité, 75006, Paris, France.
- Biologie du Médicament Et Toxicologie, AP-HP, Hôpital Cochin, 75014, Paris, France.
| |
Collapse
|
9
|
De Stefano P, Bianchi E, Dubini G. The impact of microfluidics in high-throughput drug-screening applications. BIOMICROFLUIDICS 2022; 16:031501. [PMID: 35646223 PMCID: PMC9142169 DOI: 10.1063/5.0087294] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Accepted: 05/02/2022] [Indexed: 05/05/2023]
Abstract
Drug discovery is an expensive and lengthy process. Among the different phases, drug discovery and preclinical trials play an important role as only 5-10 of all drugs that begin preclinical tests proceed to clinical trials. Indeed, current high-throughput screening technologies are very expensive, as they are unable to dispense small liquid volumes in an accurate and quick way. Moreover, despite being simple and fast, drug screening assays are usually performed under static conditions, thus failing to recapitulate tissue-specific architecture and biomechanical cues present in vivo even in the case of 3D models. On the contrary, microfluidics might offer a more rapid and cost-effective alternative. Although considered incompatible with high-throughput systems for years, technological advancements have demonstrated how this gap is rapidly reducing. In this Review, we want to further outline the role of microfluidics in high-throughput drug screening applications by looking at the multiple strategies for cell seeding, compartmentalization, continuous flow, stimuli administration (e.g., drug gradients or shear stresses), and single-cell analyses.
Collapse
Affiliation(s)
- Paola De Stefano
- Laboratory of Biological Structure Mechanics, Department of Chemistry, Materials and Chemical Engineering “G. Natta,” Politecnico di Milano, Italy
| | - Elena Bianchi
- Laboratory of Biological Structure Mechanics, Department of Chemistry, Materials and Chemical Engineering “G. Natta,” Politecnico di Milano, Italy
| | - Gabriele Dubini
- Laboratory of Biological Structure Mechanics, Department of Chemistry, Materials and Chemical Engineering “G. Natta,” Politecnico di Milano, Italy
| |
Collapse
|