1
|
Qiu B, Pompe S, Xenaki KT, Di Maggio A, Moreno CB, van Bergen En Henegouwen PMP, Mastrobattista E, Oliveira S, Caiazzo M. Receptor-mediated transcytosis of nanobodies targeting the heparin-binding EGF-like growth factor in human blood-brain barrier models. J Control Release 2025; 383:113852. [PMID: 40393531 DOI: 10.1016/j.jconrel.2025.113852] [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: 12/12/2024] [Revised: 03/20/2025] [Accepted: 05/14/2025] [Indexed: 05/22/2025]
Abstract
Transport of molecules into the brain is regulated by the blood-brain barrier (BBB). Receptor-mediated transcytosis (RMT) is a targeted vesicular transport mechanism of brain endothelial cells that can be employed to specifically transport large therapeutic molecules into the brain. ProHB-EGF is the transmembrane precursor of the heparin-binding EGF-like growth factor (HB-EGF) present on the intraluminal side of the brain endothelial cells. This molecule is characterized as an internalizing transport receptor with so far no discovery of endogenous ligands. In this study, we describe the selection and characterization of two nanobodies (named F12 and H7) with high binding affinity for proHB-EGF and their BBB transcytosis potential were tested in vitro. For the human BBB model, we found that a polarized co-culture environment was crucial for the expression and cell surface display of proHB-EGF. The ability of F12 and H7 to pass the BBB via RMT was demonstrated in both a primary human brain microvascular endothelial cell-based BBB model and a human induced pluripotent stem cell (hiPSC)-derived iBBB model. Our studies demonstrate that the proHB-EGF targeting Nbs are promising BBB shuttle molecules for delivery of therapeutic molecules into the brain.
Collapse
Affiliation(s)
- Boning Qiu
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences (UIPS), Utrecht University, 3584 CG Utrecht, the Netherlands.
| | - Sara Pompe
- Cell Biology, Neurobiology and Biophysics, Department of Biology, Science Faculty, Utrecht University, 3584 CH Utrecht, the Netherlands
| | - Katerina T Xenaki
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences (UIPS), Utrecht University, 3584 CG Utrecht, the Netherlands; Cell Biology, Neurobiology and Biophysics, Department of Biology, Science Faculty, Utrecht University, 3584 CH Utrecht, the Netherlands.
| | - Alessia Di Maggio
- Cell Biology, Neurobiology and Biophysics, Department of Biology, Science Faculty, Utrecht University, 3584 CH Utrecht, the Netherlands.
| | - Clara Belinchón Moreno
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences (UIPS), Utrecht University, 3584 CG Utrecht, the Netherlands; Cell Biology, Neurobiology and Biophysics, Department of Biology, Science Faculty, Utrecht University, 3584 CH Utrecht, the Netherlands
| | - Paul M P van Bergen En Henegouwen
- Cell Biology, Neurobiology and Biophysics, Department of Biology, Science Faculty, Utrecht University, 3584 CH Utrecht, the Netherlands.
| | - Enrico Mastrobattista
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences (UIPS), Utrecht University, 3584 CG Utrecht, the Netherlands.
| | - Sabrina Oliveira
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences (UIPS), Utrecht University, 3584 CG Utrecht, the Netherlands; Cell Biology, Neurobiology and Biophysics, Department of Biology, Science Faculty, Utrecht University, 3584 CH Utrecht, the Netherlands.
| | - Massimiliano Caiazzo
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences (UIPS), Utrecht University, 3584 CG Utrecht, the Netherlands; Department of Molecular Medicine and Medical Biotechnology, University of Naples "Federico II", Via Pansini 5, 80131 Naples, Italy.
| |
Collapse
|
2
|
Gutleb AC, Murugadoss S, Stępnik M, SenGupta T, El Yamani N, Longhin EM, Olsen AKH, Wyrzykowska E, Jagiello K, Judzinska B, Cambier S, Honza T, McFadden E, Shaposhnikov S, Puzyn T, Serchi T, Weber P, Arnesdotter E, Skakalova V, Jirsova K, Grudzinski IP, Collins A, Rundén-Pran E, Dusinska M. New Approach Methods (NAMs) for genotoxicity assessment of nano- and advanced materials; Advantages and challenges. MUTATION RESEARCH. GENETIC TOXICOLOGY AND ENVIRONMENTAL MUTAGENESIS 2025; 904:503867. [PMID: 40382189 DOI: 10.1016/j.mrgentox.2025.503867] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Revised: 03/07/2025] [Accepted: 03/26/2025] [Indexed: 05/20/2025]
Abstract
Genotoxicity assessment is essential for ensuring chemical safety and mitigating risks to human health and the environment. Traditional methods, reliant on animal models, are time-consuming, costly, and raise ethical concerns. New Approach Methods (NAMs) offer innovative, cost-effective, and ethical alternatives, playing a pivotal role in both traditional and next-generation risk assessment (NGRA) by minimizing the need for animal testing, particularly in genotoxicity evaluations. However, the development of NAMs often overlooks the particular physicochemical properties of nanomaterials (NMs), which significantly influence their toxicological behaviour and can interfere with genotoxicity evaluation. This underscores an urgent need for the standardization and adaptation of NAMs to address nano- and advanced material-specific genotoxicity challenges. In this review, we summarize the challenges associated with genotoxicity testing of NMs and highlight the suitability of existing in vitro and in silico NAMs for NMs and advanced materials, enabling genotoxicity testing across various exposure routes and organ systems. Despite considerable progress, regulatory validation remains constrained by the absence of approved test guidelines and standardized protocols. To achieve regulatory acceptance, it is crucial to adapt NAMs to NM-specific exposure scenarios, refine test systems to better mimic human biology, develop tailored in vitro protocols, and ensure thorough characterisation of NMs both in pristine form and dispersed in culture medium. Collaborative efforts among scientists, regulators, industry, and advocacy groups are vital to improving the reliability and regulatory acceptance of NAMs. By addressing these challenges, NAMs have the potential to revolutionize genotoxicity risk assessment, advancing it towards a more sustainable, efficient and ethical framework.
Collapse
Affiliation(s)
- Arno C Gutleb
- Environmental Sustainability Assessment and Circularity (SUSTAIN) Unit, Luxembourg Institute of Science and Technology (LIST), Luxembourg; INVITROLIZE sarl, Wellenstein, Luxembourg.
| | - Sivakumar Murugadoss
- Health Effects Laboratory, Department of Environmental Chemistry and Health Effects, NILU The Climate and Environmental Research Institute, Norway.
| | - Maciej Stępnik
- QSAR Lab Ltd, Trzy Lipy 3, Gdańsk, Poland; Department of Toxicology, Faculty of Pharmacy, Medical University of Łódź, Poland.
| | - Tanima SenGupta
- Health Effects Laboratory, Department of Environmental Chemistry and Health Effects, NILU The Climate and Environmental Research Institute, Norway.
| | - Naouale El Yamani
- Health Effects Laboratory, Department of Environmental Chemistry and Health Effects, NILU The Climate and Environmental Research Institute, Norway.
| | - Eleonora Marta Longhin
- Health Effects Laboratory, Department of Environmental Chemistry and Health Effects, NILU The Climate and Environmental Research Institute, Norway.
| | - Ann-Karin Hardie Olsen
- Health Effects Laboratory, Department of Environmental Chemistry and Health Effects, NILU The Climate and Environmental Research Institute, Norway.
| | | | - Karolina Jagiello
- QSAR Lab Ltd, Trzy Lipy 3, Gdańsk, Poland; University of Gdańsk, Faculty of Chemistry, Laboratory of Environmental Chemoinformatics, Wita Stwosza, Gdańsk 63, 80-308, Poland.
| | - Beata Judzinska
- QSAR Lab Ltd, Trzy Lipy 3, Gdańsk, Poland; University of Gdańsk, Faculty of Chemistry, Laboratory of Environmental Chemoinformatics, Wita Stwosza, Gdańsk 63, 80-308, Poland.
| | - Sebastien Cambier
- Environmental Sustainability Assessment and Circularity (SUSTAIN) Unit, Luxembourg Institute of Science and Technology (LIST), Luxembourg.
| | - Tatiana Honza
- Health Effects Laboratory, Department of Environmental Chemistry and Health Effects, NILU The Climate and Environmental Research Institute, Norway.
| | - Erin McFadden
- Health Effects Laboratory, Department of Environmental Chemistry and Health Effects, NILU The Climate and Environmental Research Institute, Norway.
| | | | - Tomasz Puzyn
- QSAR Lab Ltd, Trzy Lipy 3, Gdańsk, Poland; University of Gdańsk, Faculty of Chemistry, Laboratory of Environmental Chemoinformatics, Wita Stwosza, Gdańsk 63, 80-308, Poland.
| | - Tommaso Serchi
- Environmental Sustainability Assessment and Circularity (SUSTAIN) Unit, Luxembourg Institute of Science and Technology (LIST), Luxembourg.
| | - Pamina Weber
- Environmental Sustainability Assessment and Circularity (SUSTAIN) Unit, Luxembourg Institute of Science and Technology (LIST), Luxembourg
| | - Emma Arnesdotter
- Environmental Sustainability Assessment and Circularity (SUSTAIN) Unit, Luxembourg Institute of Science and Technology (LIST), Luxembourg
| | | | - Katerina Jirsova
- Laboratory of Biology and Pathology of the Eye, Institute of Biology and Medical Genetics, 1st Faculty of Medicine and General Teaching Hospital, Charles University, Prague, Czech Republic.
| | - Ireneusz P Grudzinski
- Department of Toxicology and Food Science, Faculty of Pharmacy, Medical University of Warsaw, Banacha Str. 1, Warsaw 02-097, Poland.
| | - Andrew Collins
- NorGenoTech, Norway; Department of Nutrition, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway.
| | - Elise Rundén-Pran
- Health Effects Laboratory, Department of Environmental Chemistry and Health Effects, NILU The Climate and Environmental Research Institute, Norway.
| | - Maria Dusinska
- Health Effects Laboratory, Department of Environmental Chemistry and Health Effects, NILU The Climate and Environmental Research Institute, Norway.
| |
Collapse
|
3
|
Vetter J, Palagi I, Waisman A, Blaeser A. Recent advances in blood-brain barrier-on-a-chip models. Acta Biomater 2025; 197:1-28. [PMID: 40127880 DOI: 10.1016/j.actbio.2025.03.041] [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: 11/20/2024] [Revised: 03/19/2025] [Accepted: 03/21/2025] [Indexed: 03/26/2025]
Abstract
The blood-brain barrier is a physiological barrier between the vascular system and the nervous system. Under healthy conditions, it restricts the passage of most biomolecules into the brain, making drug development exceedingly challenging. Conventional cell-based in vitro models provide valuable insights into certain features of the BBB. Nevertheless, these models often lack the three-dimensional structure and dynamic interactions of the surrounding microenvironment, which greatly influence cell functionality. Consequently, considerable efforts have been made to enhance in vitro models for drug development and disease research. Recently, microfluidic organ-on-a-chip systems have emerged as promising candidates to better mimic the dynamic nature of the BBB. This review provides a comprehensive overview of recent BBB-on-chip devices. The typical building blocks, chip designs, the perfusion infrastructure, and readouts used to characterize and evaluate BBB formation are presented, analyzed, and discussed in detail. STATEMENT OF SIGNIFICANCE: The blood-brain barrier (BBB) is a highly selective barrier that controls what can enter the brain. While it protects the brain from harmful substances, it also hinders the delivery of treatments for neurological diseases such as Alzheimer's and Parkinson's. Due to its complexity, studying the BBB in living organisms remains difficult. However, recent advances in "organ-on-a-chip" technology have allowed scientists to create small, engineered models that replicate the BBB. These models provide a powerful platform to study diseases and test potential drugs with greater accuracy than traditional methods. Organ-on-a-chip devices are designed to mimic the behavior of organs or tissues in the human body, offering a more realistic and controlled environment for research. This review highlights recent breakthroughs in BBB-on-a-chip technology, showing how these models enhance current research and have the potential to transform the way we study brain diseases and develop new drugs. By integrating biology and engineering, BBB-on-a-chip technology has the potential to transform neuroscience research, improve drug development, and enhance our understanding of brain disorders.
Collapse
Affiliation(s)
- Johanna Vetter
- Institute for BioMedical Printing Technology, Technical University of Darmstadt, Darmstadt, Germany
| | - Ilaria Palagi
- Institute for Molecular Medicine, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Ari Waisman
- Institute for Molecular Medicine, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany; Research Center for Immunotherapy (FZI), University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Andreas Blaeser
- Institute for BioMedical Printing Technology, Technical University of Darmstadt, Darmstadt, Germany; Centre for Synthetic Biology, Technical University of Darmstadt, Darmstadt, Germany.
| |
Collapse
|
4
|
Mathew‐Schmitt S, Oerter S, Reitenbach E, Gätzner S, Höchner A, Jahnke H, Piontek J, Neuhaus W, Brachner A, Metzger M, Appelt‐Menzel A. Generation of Advanced Blood-Brain Barrier Spheroids Using Human-Induced Pluripotent Stem Cell-Derived Brain Capillary Endothelial-Like Cells. Adv Biol (Weinh) 2025; 9:e2400442. [PMID: 39912766 PMCID: PMC12001013 DOI: 10.1002/adbi.202400442] [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: 08/13/2024] [Revised: 11/12/2024] [Indexed: 02/07/2025]
Abstract
Extensively studied blood-brain barrier (BBB) in-vitro models are established on 2D cell culture inserts. However, they do not accurately represent 3D in-vivo microenvironments due to lack of direct neurovascular unit cellular contacts. Here, the establishment and characterization of a self-assembled 3D BBB spheroid model using human-induced pluripotent stem cell (hiPSC)-derived brain capillary endothelial-like cells (iBCECs) in combination with primary human astrocytes (ACs) and pericytes (PCs) are reported. This investigation compares 3D spheroids with 2D mono-cultured iBCECs derived from two different hiPSC lines and two differentiation strategies. It is observed that spheroid properties vary depending on the differentiation strategy or type of hiPSC line applied for model generation. However, spheroids demonstrate in-vivo like tight junction ultrastructure and, in comparison to 2D models, higher transcript expression of BBB specific genes. Furthermore, they possess characteristic barrier integrity, barrier functionality, and protein expression. It is inferred that hiPSC-derived BBB spheroids hold a strong potential as a reliable future BBB in-vitro test system.
Collapse
Affiliation(s)
- Sanjana Mathew‐Schmitt
- Chair Tissue Engineering and Regenerative Medicine (TERM)University Hospital Würzburg97070WürzburgGermany
| | - Sabrina Oerter
- Chair Tissue Engineering and Regenerative Medicine (TERM)University Hospital Würzburg97070WürzburgGermany
- Fraunhofer Institute for Silicate Research ISCTranslational Centre Regenerative Therapies (TLC‐RT)97070WürzburgGermany
| | - Evelin Reitenbach
- Fraunhofer Institute for Silicate Research ISCTranslational Centre Regenerative Therapies (TLC‐RT)97070WürzburgGermany
| | - Sabine Gätzner
- Chair Tissue Engineering and Regenerative Medicine (TERM)University Hospital Würzburg97070WürzburgGermany
| | - Alevtina Höchner
- Fraunhofer Institute for Silicate Research ISCTranslational Centre Regenerative Therapies (TLC‐RT)97070WürzburgGermany
| | - Heinz‐Georg Jahnke
- Biotechnological‐Biomedical Center (BBZ)University of Leipzig04103LeipzigGermany
| | - Jörg Piontek
- Clinical Physiology/Nutritional MedicineDepartment of GastroenterologyRheumatology and Infectious DiseasesCharité–Universitätsmedizin Berlin12203BerlinGermany
| | - Winfried Neuhaus
- AIT Austrian Institute of Technology GmbHCentre Health and Bioresources, Competence Unit Molecular DiagnosticsVienna1210Austria
- Department of MedicineFaculty Dentistry and MedicinePrivate Danube UniversityKrems3500Austria
| | - Andreas Brachner
- AIT Austrian Institute of Technology GmbHCentre Health and Bioresources, Competence Unit Molecular DiagnosticsVienna1210Austria
| | - Marco Metzger
- Fraunhofer Institute for Silicate Research ISCTranslational Centre Regenerative Therapies (TLC‐RT)97070WürzburgGermany
| | - Antje Appelt‐Menzel
- Chair Tissue Engineering and Regenerative Medicine (TERM)University Hospital Würzburg97070WürzburgGermany
- Fraunhofer Institute for Silicate Research ISCTranslational Centre Regenerative Therapies (TLC‐RT)97070WürzburgGermany
| |
Collapse
|
5
|
Shamul JG, Wang Z, Gong H, Ou W, White AM, Moniz-Garcia DP, Gu S, Clyne AM, Quiñones-Hinojosa A, He X. Meta-analysis of the make-up and properties of in vitro models of the healthy and diseased blood-brain barrier. Nat Biomed Eng 2025; 9:566-598. [PMID: 39304761 PMCID: PMC11922799 DOI: 10.1038/s41551-024-01250-2] [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: 07/06/2023] [Accepted: 08/08/2024] [Indexed: 09/22/2024]
Abstract
In vitro models of the human blood-brain barrier (BBB) are increasingly used to develop therapeutics that can cross the BBB for treating diseases of the central nervous system. Here we report a meta-analysis of the make-up and properties of transwell and microfluidic models of the healthy BBB and of BBBs in glioblastoma, Alzheimer's disease, Parkinson's disease and inflammatory diseases. We found that the type of model, the culture method (static or dynamic), the cell types and cell ratios, and the biomaterials employed as extracellular matrix are all crucial to recapitulate the low permeability and high expression of tight-junction proteins of the BBB, and to obtain high trans-endothelial electrical resistance. Specifically, for models of the healthy BBB, the inclusion of endothelial cells and pericytes as well as physiological shear stresses (~10-20 dyne cm-2) are necessary, and when astrocytes are added, astrocytes or pericytes should outnumber endothelial cells. We expect this meta-analysis to facilitate the design of increasingly physiological models of the BBB.
Collapse
Affiliation(s)
- James G Shamul
- Fischell Department of Bioengineering, University of Maryland, College Park, MD, USA
- RNA Mediated Gene Regulation Section, RNA Biology Laboratory, Center for Cancer Research, National Cancer Institute, Frederick, MD, USA
| | - Zhiyuan Wang
- Fischell Department of Bioengineering, University of Maryland, College Park, MD, USA
| | - Hyeyeon Gong
- Fischell Department of Bioengineering, University of Maryland, College Park, MD, USA
| | - Wenquan Ou
- Fischell Department of Bioengineering, University of Maryland, College Park, MD, USA
| | - Alisa M White
- Fischell Department of Bioengineering, University of Maryland, College Park, MD, USA
| | | | - Shuo Gu
- RNA Mediated Gene Regulation Section, RNA Biology Laboratory, Center for Cancer Research, National Cancer Institute, Frederick, MD, USA
| | - Alisa Morss Clyne
- Fischell Department of Bioengineering, University of Maryland, College Park, MD, USA
- Robert E. Fischell Institute for Biomedical Devices, University of Maryland, College Park, MD, USA
- Brain and Behavior Institute, University of Maryland, College Park, MD, USA
| | | | - Xiaoming He
- Fischell Department of Bioengineering, University of Maryland, College Park, MD, USA.
- Robert E. Fischell Institute for Biomedical Devices, University of Maryland, College Park, MD, USA.
- Brain and Behavior Institute, University of Maryland, College Park, MD, USA.
- Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland, Baltimore, MD, USA.
| |
Collapse
|
6
|
Soliman Y, Al-Khodor J, Yildirim Köken G, Mustafaoglu N. A guide for blood-brain barrier models. FEBS Lett 2025; 599:599-644. [PMID: 39533665 DOI: 10.1002/1873-3468.15053] [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: 05/13/2024] [Revised: 10/18/2024] [Accepted: 10/20/2024] [Indexed: 11/16/2024]
Abstract
Understanding the intricate mechanisms underlying brain-related diseases hinges on unraveling the pivotal role of the blood-brain barrier (BBB), an essential dynamic interface crucial for maintaining brain equilibrium. This review offers a comprehensive analysis of BBB physiology, delving into its cellular and molecular components while exploring a wide range of in vivo and in vitro BBB models. Notably, recent advancements in 3D cell culture techniques are explicitly discussed, as they have significantly improved the fidelity of BBB modeling by enabling the replication of physiologically relevant environments under flow conditions. Special attention is given to the cellular aspects of in vitro BBB models, alongside discussions on advances in stem cell technologies, providing valuable insights into generating robust cellular systems for BBB modeling. The diverse array of cell types used in BBB modeling, depending on their sources, is meticulously examined in this comprehensive review, scrutinizing their respective derivation protocols and implications. By synthesizing diverse approaches, this review sheds light on the improvements of BBB models to capture physiological conditions, aiding in understanding BBB interactions in health and disease conditions to foster clinical developments.
Collapse
Affiliation(s)
- Yomna Soliman
- Faculty of Engineering and Natural Sciences, Sabancı University, Istanbul, Turkey
- Faculty of Pharmacy, Mansoura University, Egypt
| | - Jana Al-Khodor
- Faculty of Engineering and Natural Sciences, Sabancı University, Istanbul, Turkey
| | | | - Nur Mustafaoglu
- Faculty of Engineering and Natural Sciences, Sabancı University, Istanbul, Turkey
- Sabancı University Nanotechnology Research and Application Center, Istanbul, Turkey
| |
Collapse
|
7
|
Maboko LM, Theron A, Panayides J, Cordier W, Fisher D, Steenkamp V. Evaluating Blood-Brain Barrier Permeability, Cytotoxicity, and Activity of Potential Acetylcholinesterase Inhibitors: In Vitro and In Silico Study. Pharmacol Res Perspect 2024; 12:e70043. [PMID: 39651604 PMCID: PMC11841676 DOI: 10.1002/prp2.70043] [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: 07/01/2024] [Revised: 11/08/2024] [Accepted: 11/14/2024] [Indexed: 12/11/2024] Open
Abstract
Acetylcholinesterase inhibitors (AChEIs) remain the first-line treatment for Alzheimer's disease. However, these drugs are largely symptomatic and often associated with adverse effects. This study aimed to evaluate novel pharmacophores for their in vitro AChEI activity, blood-brain barrier (BBB) permeability, and cytotoxic potential, hypothesizing that a combination of AChEIs could enhance symptom management while minimizing toxicity. A library of 1453 synthetic pharmacophores was assessed using in vitro and in silico methods to determine their feasibility as an inhibitor of the AChE enzyme. An in-house miniaturized Ellman's assay determined acellular AChEI activities, while pharmacokinetic properties were evaluated using the SwissADME web tool. The combinational effects of in silico BBB-permeable pharmacophores and donepezil were examined using a checkerboard AChEI assay. Cytotoxicity of active compounds and their synergistic combinations was assessed in SH-SY5Y neuroblastoma and bEnd.5 cells using the sulforhodamine B assay. Cellular AChEI activity of active in silico BBB-permeable predicted compounds was determined using an SH-SY5Y AChE-based assay. An in vitro BBB model was used to assess the effect of compounds on the integrity of the bEnd.5 monolayer. Out of the screened compounds, 12 demonstrated 60% AChEI activity at 5 μM, with compound A51 showing the lowest IC50 (0.20 μM). Five compounds were identified as BBB-permeable, with the donepezil-C53 combination at ¼IC50 exhibiting the strongest synergy (CI = 0.82). Compounds A136 and C129, either alone or with donepezil, showed cytotoxicity. Notably, compound C53, both alone and in combination with donepezil, demonstrated high AChEI activity and promising BBB permeability, warranting further investigation.
Collapse
Affiliation(s)
- L. M. Maboko
- Department of Pharmacology, School of Medicine, Faculty of Health SciencesUniversity of PretoriaPretoriaSouth Africa
| | - A. Theron
- Future Production: Chemicals, Council for Scientific and Industrial ResearchPretoriaSouth Africa
| | - J.‐L. Panayides
- Future Production: Chemicals, Council for Scientific and Industrial ResearchPretoriaSouth Africa
| | - W. Cordier
- Department of Pharmacology, School of Medicine, Faculty of Health SciencesUniversity of PretoriaPretoriaSouth Africa
| | - D. Fisher
- Department of Medical BioSciences, Faculty of Natural Sciences, Neurobiology Research GroupUniversity of Western CapeCape TownSouth Africa
| | - V. Steenkamp
- Department of Pharmacology, School of Medicine, Faculty of Health SciencesUniversity of PretoriaPretoriaSouth Africa
| |
Collapse
|
8
|
Königer L, Malkmus C, Mahdy D, Däullary T, Götz S, Schwarz T, Gensler M, Pallmann N, Cheufou D, Rosenwald A, Möllmann M, Groneberg D, Popp C, Groeber‐Becker F, Steinke M, Hansmann J. ReBiA-Robotic Enabled Biological Automation: 3D Epithelial Tissue Production. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2406608. [PMID: 39324843 PMCID: PMC11615785 DOI: 10.1002/advs.202406608] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Revised: 08/08/2024] [Indexed: 09/27/2024]
Abstract
The Food and Drug Administration's recent decision to eliminate mandatory animal testing for drug approval marks a significant shift to alternative methods. Similarly, the European Parliament is advocating for a faster transition, reflecting public preference for animal-free research practices. In vitro tissue models are increasingly recognized as valuable tools for regulatory assessments before clinical trials, in line with the 3R principles (Replace, Reduce, Refine). Despite their potential, barriers such as the need for standardization, availability, and cost hinder their widespread adoption. To address these challenges, the Robotic Enabled Biological Automation (ReBiA) system is developed. This system uses a dual-arm robot capable of standardizing laboratory processes within a closed automated environment, translating manual processes into automated ones. This reduces the need for process-specific developments, making in vitro tissue models more consistent and cost-effective. ReBiA's performance is demonstrated through producing human reconstructed epidermis, human airway epithelial models, and human intestinal organoids. Analyses confirm that these models match the morphology and protein expression of manually prepared and native tissues, with similar cell viability. These successes highlight ReBiA's potential to lower barriers to broader adoption of in vitro tissue models, supporting a shift toward more ethical and advanced research methods.
Collapse
Affiliation(s)
- Lukas Königer
- Translational Center Regenerative TherapiesFraunhofer Institute for Silicate Research ISC97070WürzburgGermany
| | - Christoph Malkmus
- Translational Center Regenerative TherapiesFraunhofer Institute for Silicate Research ISC97070WürzburgGermany
- Institute of Medical Engineering SchweinfurtTechnical University of Applied Sciences Würzburg‐Schweinfurt97421SchweinfurtGermany
| | - Dalia Mahdy
- Chair of Tissue Engineering and Regenerative MedicineUniversity Hospital Würzburg97070WürzburgGermany
| | - Thomas Däullary
- Chair of Tissue Engineering and Regenerative MedicineUniversity Hospital Würzburg97070WürzburgGermany
- Chair of Cellular ImmunotherapyUniversity Hospital Würzburg97080WürzburgGermany
| | - Susanna Götz
- Faculty of Design WürzburgTechnical University of Applied Sciences Würzburg‐Schweinfurt97070WürzburgGermany
| | - Thomas Schwarz
- Translational Center Regenerative TherapiesFraunhofer Institute for Silicate Research ISC97070WürzburgGermany
| | - Marius Gensler
- Chair of Tissue Engineering and Regenerative MedicineUniversity Hospital Würzburg97070WürzburgGermany
| | - Niklas Pallmann
- Chair of Tissue Engineering and Regenerative MedicineUniversity Hospital Würzburg97070WürzburgGermany
| | - Danjouma Cheufou
- Department of Thoracic SurgeryKlinikum Würzburg Mitte97070WürzburgGermany
| | | | - Marc Möllmann
- Translational Center Regenerative TherapiesFraunhofer Institute for Silicate Research ISC97070WürzburgGermany
| | - Dieter Groneberg
- Translational Center Regenerative TherapiesFraunhofer Institute for Silicate Research ISC97070WürzburgGermany
| | - Christina Popp
- Translational Center Regenerative TherapiesFraunhofer Institute for Silicate Research ISC97070WürzburgGermany
| | - Florian Groeber‐Becker
- Translational Center Regenerative TherapiesFraunhofer Institute for Silicate Research ISC97070WürzburgGermany
- Department of OphthalmologyUniversity Clinic Düsseldorf40225DüsseldorfGermany
| | - Maria Steinke
- Translational Center Regenerative TherapiesFraunhofer Institute for Silicate Research ISC97070WürzburgGermany
- Department of Oto‐Rhino‐LaryngologyPlasticAesthetic and Reconstructive Head and Neck SurgeryUniversity Hospital Würzburg97080WürzburgGermany
| | - Jan Hansmann
- Translational Center Regenerative TherapiesFraunhofer Institute for Silicate Research ISC97070WürzburgGermany
- Institute of Medical Engineering SchweinfurtTechnical University of Applied Sciences Würzburg‐Schweinfurt97421SchweinfurtGermany
| |
Collapse
|
9
|
Pang B, Wu L, Peng Y. In vitro modelling of the neurovascular unit for ischemic stroke research: Emphasis on human cell applications and 3D model design. Exp Neurol 2024; 381:114942. [PMID: 39222766 DOI: 10.1016/j.expneurol.2024.114942] [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: 03/15/2024] [Revised: 07/16/2024] [Accepted: 08/29/2024] [Indexed: 09/04/2024]
Abstract
Ischemic stroke has garnered global medical attention as one of the most serious cerebrovascular diseases. The mechanisms involved in both the development and recovery phases of ischemic stroke are complex, involving intricate interactions among different types of cells, each with its own unique functions. To better understand the possible pathogenesis, neurovascular unit (NVU), a concept comprising neurons, endothelial cells, mural cells, glial cells, and extracellular matrix components, has been used in analysing various brain diseases, particularly in ischemic stroke, aiming to depict the interactions between cerebral vasculature and neural cells. While in vivo models often face limitations in terms of reproducibility and the ability to precisely mimic human pathophysiology, it is now important to establish in vitro NVU models for ischemic stroke research. In order to accurately portray the pathological processes occurring within the brain, a diverse array of NVU 2D and 3D in vitro models, each possessing unique characteristics and advantages, have been meticulously developed. This review presents a comprehensive overview of recent advancements in in vitro models specifically tailored for investigating ischemic stroke. Through a systematic categorization of these developments, we elucidate the intricate links between NVU components and the pathogenesis of ischemic stroke. Furthermore, we explore the distinct advantages offered by innovative NVU models, notably 3D models, which closely emulate in vivo conditions. Additionally, an examination of current therapeutic modalities for ischemic stroke developed utilizing in vitro NVU models is provided. Serving as a valuable reference, this review aids in the design and implementation of effective in vitro models for ischemic stroke research.
Collapse
Affiliation(s)
- Bo Pang
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
| | - Lei Wu
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
| | - Ying Peng
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China.
| |
Collapse
|
10
|
Kanjanasirirat P, Saengsawang W, Ketsawatsomkron P, Asavapanumas N, Borwornpinyo S, Soodvilai S, Hongeng S, Charoensutthivarakul S. GDNF and cAMP significantly enhance in vitro blood-brain barrier integrity in a humanized tricellular transwell model. Heliyon 2024; 10:e39343. [PMID: 39492921 PMCID: PMC11530796 DOI: 10.1016/j.heliyon.2024.e39343] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Revised: 10/11/2024] [Accepted: 10/11/2024] [Indexed: 11/05/2024] Open
Abstract
Blood-brain barrier (BBB) is a crucial membrane safeguarding neural tissue by controlling the molecular exchange between blood and the brain. However, assessing BBB permeability presents challenges for central nervous system (CNS) drug development. In vitro studies of BBB-permeable agents before animal testing are essential to mitigate failures. Improved in vitro models are needed to mimic physiologically relevant BBB integrity. Here, we established an in vitro human-derived triculture BBB model, coculturing hCMEC/D3 with primary astrocytes and pericytes in a transwell format. This study found that the triculture BBB model exhibited significantly higher paracellular tightness (TEER 147.6 ± 6.5 Ω × cm2) than its monoculture counterpart (106.3 ± 1.0 Ω × cm2). Additionally, BBB permeability in the triculture model was significantly lower. While GDNF and cAMP have been shown to promote BBB integrity in monoculture models, their effect in our model was previously unreported. Our study demonstrates that both GDNF and cAMP increased TEER values (around 200 Ω × cm2 for each; 237.6 ± 17.7 Ω × cm2 for co-treatment) compared to untreated control, and decreased BBB permeability, mediated by increased claudin-5 expression. In summary, this humanized triculture BBB model, enhanced by GDNF and cAMP, offers an alternative for exploring in vitro drug penetration into the human brain.
Collapse
Affiliation(s)
- Phongthon Kanjanasirirat
- School of Bioinnovation and Bio-Based Product Intelligence, Faculty of Science, Mahidol University, Bangkok, 10400, Thailand
- Excellent Center for Drug Discovery (ECDD), Faculty of Science, Mahidol University, Bangkok, 10400, Thailand
- Department of Pathobiology, Faculty of Science, Mahidol University, Bangkok, 10400, Thailand
| | - Witchuda Saengsawang
- Department of Basic Biomedical Sciences, Dr. William M. Scholl College of Podiatric Medicine, Rosalind Franklin University of Medicine and Science, North Chicago, IL, 60064, USA
| | - Pimonrat Ketsawatsomkron
- Chakri Naruebodindra Medical Institute, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Samut Prakarn, 10540, Thailand
| | - Nithi Asavapanumas
- Chakri Naruebodindra Medical Institute, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Samut Prakarn, 10540, Thailand
| | - Suparerk Borwornpinyo
- Excellent Center for Drug Discovery (ECDD), Faculty of Science, Mahidol University, Bangkok, 10400, Thailand
- Department of Biotechnology, Faculty of Science, Mahidol University, Bangkok, 10400, Thailand
| | - Sunhapas Soodvilai
- Excellent Center for Drug Discovery (ECDD), Faculty of Science, Mahidol University, Bangkok, 10400, Thailand
- Department of Physiology, Faculty of Science, Mahidol University, Bangkok, 10400, Thailand
| | - Suradej Hongeng
- Excellent Center for Drug Discovery (ECDD), Faculty of Science, Mahidol University, Bangkok, 10400, Thailand
- Department of Pediatrics, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok, 10400, Thailand
| | - Sitthivut Charoensutthivarakul
- School of Bioinnovation and Bio-Based Product Intelligence, Faculty of Science, Mahidol University, Bangkok, 10400, Thailand
- Excellent Center for Drug Discovery (ECDD), Faculty of Science, Mahidol University, Bangkok, 10400, Thailand
- Center for Neuroscience, Faculty of Science, Mahidol University, Bangkok, 10400, Thailand
| |
Collapse
|
11
|
Mathew-Schmitt S, Peindl M, Neundorf P, Dandekar G, Metzger M, Nickl V, Appelt-Menzel A. Blood-tumor barrier in focus - investigation of glioblastoma-induced effects on the blood-brain barrier. J Neurooncol 2024; 170:67-77. [PMID: 39196480 PMCID: PMC11446994 DOI: 10.1007/s11060-024-04760-w] [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: 05/08/2024] [Accepted: 06/26/2024] [Indexed: 08/29/2024]
Abstract
PURPOSE Glioblastoma (GBM) is the most prevalent, malignant, primary brain tumor in adults, characterized by limited treatment options, frequent relapse, and short survival after diagnosis. Until now, none of the existing therapy and treatment approaches have proven to be an effective cure. The availability of predictive human blood-tumor barrier (BTB) test systems that can mimic in-vivo pathophysiology of GBM would be of great interest in preclinical research. Here, we present the establishment of a new BTB in-vitro test system combining GBM spheroids and BBB models derived from human induced pluripotent stem cells (hiPSCs). METHODS We co-cultured hiPSC-derived brain capillary endothelial-like cells (iBCECs) with GBM spheroids derived from U87-MG and U373-MG cell lines in a cell culture insert-based format. Spheroids were monitored over 168 hours (h) of culture, characterized for GBM-specific marker expression and treated with standard chemotherapeutics to distinguish inhibitory effects between 2D mono-culture and 3D spheroids. GBM-induced changes on iBCECs barrier integrity were verified via measurement of transendothelial electrical resistance (TEER), immunocytochemical staining of tight junction (TJ) proteins claudin-5 and occludin as well as the glucose transporter-1 (Glut-1). GBM-induced secretion of vascular endothelial growth factor (VEGF) was additionally quantified. RESULTS Our hypothesis was validated by reduced expression of TJ proteins, occludin and claudin-5 together with significant barrier breakdown in iBCECs after only 24 h of co-culture, demonstrated by reduction in TEER from 1313 ± 265 Ω*cm2 to 712 ± 299 Ω*cm2 (iBCECs + U87-MG) and 762 ± 316 Ω*cm2 (iBCECs + U373-MG). Furthermore, 3D spheroids show more resistance to standard GBM chemotherapeutics in-vitro compared to 2D cultures. CONCLUSIONS We demonstrate the establishment of a simplified, robust in-vitro BTB test system, with potential application in preclinical therapeutic screening and in studying GBM-induced pathological changes at the BBB.
Collapse
Affiliation(s)
- Sanjana Mathew-Schmitt
- Chair Tissue Engineering and Regenerative Medicine, University Hospital Würzburg, Röntgenring 11, 97070, Würzburg, Germany
| | - Matthias Peindl
- Chair Tissue Engineering and Regenerative Medicine, University Hospital Würzburg, Röntgenring 11, 97070, Würzburg, Germany
| | - Philipp Neundorf
- Chair Tissue Engineering and Regenerative Medicine, University Hospital Würzburg, Röntgenring 11, 97070, Würzburg, Germany
| | - Gudrun Dandekar
- Chair Tissue Engineering and Regenerative Medicine, University Hospital Würzburg, Röntgenring 11, 97070, Würzburg, Germany
| | - Marco Metzger
- Chair Tissue Engineering and Regenerative Medicine, University Hospital Würzburg, Röntgenring 11, 97070, Würzburg, Germany
- Translational Centre Regenerative Therapies TLC-RT, Fraunhofer Institute for Silicate Research ISC, Röntgenring 11, 97070, Würzburg, Germany
| | - Vera Nickl
- Section Experimental Neurosurgery, Department of Neurosurgery, University Hospital Würzburg, Josef-Schneider-Straße 11, 97080, Würzburg, Germany
| | - Antje Appelt-Menzel
- Chair Tissue Engineering and Regenerative Medicine, University Hospital Würzburg, Röntgenring 11, 97070, Würzburg, Germany.
- Translational Centre Regenerative Therapies TLC-RT, Fraunhofer Institute for Silicate Research ISC, Röntgenring 11, 97070, Würzburg, Germany.
| |
Collapse
|
12
|
Sampedro-Viana A, Fernández-Rodicio S, Castillo J, Hervella P, Alonso-Alonso ML, Iglesias-Rey R. Assessment of Mannitol-Induced Chronic Blood-Brain Barrier Dysfunction In Vivo Using Magnetic Resonance. Int J Mol Sci 2024; 25:9792. [PMID: 39337280 PMCID: PMC11431755 DOI: 10.3390/ijms25189792] [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: 08/02/2024] [Revised: 09/05/2024] [Accepted: 09/06/2024] [Indexed: 09/30/2024] Open
Abstract
The blood-brain barrier (BBB) is essential for protection and plays a crucial role in chronic neurological disorders like small-vessel disease and Alzheimer's disease. Its complexity poses significant challenges for effective diagnostics and treatments, highlighting the need for novel animal models and comprehensive BBB dysfunction studies. This study investigates chronic BBB dysfunction induction using osmotic disruption via mannitol in healthy adult male Sprague Dawley rats over 12 weeks. Group 1 received 1 bolus/week (2.0 g/kg), Group 2 received 3 boluses/week (1.5 g/kg), and Group 3 received 3 boluses/week (2.5 g/kg). BBB dysfunction was assessed using gadolinium (Gd) infusion and MRI to evaluate location, severity, evolution, and persistence. MR spectroscopy (MRS) examined the brain metabolism changes due to intravenous mannitol, with T2-weighted MRI assessing brain lesions. Biomarkers of neuroinflammation were analyzed in the highest mannitol dose group. Our data show chronic BBB dysfunction primarily in the cortex, hippocampus, and striatum, but not in the corpus callosum of rats under periodic mannitol dosing in groups 1 and 2. MRS identified a distinctive metabolite signature, including changes in alanine, choline, and N-acetyl aspartate in the striatum of Group 1. No significant differences were found in the serum levels of all pro- and anti-inflammatory cytokines analyzed in the high-dose Group 3. This study underscores the feasibility and implications of using osmotic disruption to model chronic BBB dysfunction, offering insights for future neuroprotection and therapeutic strategies research.
Collapse
Affiliation(s)
- Ana Sampedro-Viana
- Neuroimaging and Biotechnology Laboratory (NOBEL), Clinical Neurosciences Research Laboratory (LINC), Health Research Institute of Santiago de Compostela (IDIS), Hospital Clínico Universitario, Rúa Travesa da Choupana s/n, 15706 Santiago de Compostela, Spain
| | - Sabela Fernández-Rodicio
- Neuroimaging and Biotechnology Laboratory (NOBEL), Clinical Neurosciences Research Laboratory (LINC), Health Research Institute of Santiago de Compostela (IDIS), Hospital Clínico Universitario, Rúa Travesa da Choupana s/n, 15706 Santiago de Compostela, Spain
| | - José Castillo
- Neuroimaging and Biotechnology Laboratory (NOBEL), Clinical Neurosciences Research Laboratory (LINC), Health Research Institute of Santiago de Compostela (IDIS), Hospital Clínico Universitario, Rúa Travesa da Choupana s/n, 15706 Santiago de Compostela, Spain
| | - Pablo Hervella
- Neuroimaging and Biotechnology Laboratory (NOBEL), Clinical Neurosciences Research Laboratory (LINC), Health Research Institute of Santiago de Compostela (IDIS), Hospital Clínico Universitario, Rúa Travesa da Choupana s/n, 15706 Santiago de Compostela, Spain
| | - María Luz Alonso-Alonso
- Neuroimaging and Biotechnology Laboratory (NOBEL), Clinical Neurosciences Research Laboratory (LINC), Health Research Institute of Santiago de Compostela (IDIS), Hospital Clínico Universitario, Rúa Travesa da Choupana s/n, 15706 Santiago de Compostela, Spain
| | - Ramón Iglesias-Rey
- Neuroimaging and Biotechnology Laboratory (NOBEL), Clinical Neurosciences Research Laboratory (LINC), Health Research Institute of Santiago de Compostela (IDIS), Hospital Clínico Universitario, Rúa Travesa da Choupana s/n, 15706 Santiago de Compostela, Spain
| |
Collapse
|
13
|
Spathopoulou A, Podlesnic M, De Gaetano L, Kirsch EM, Tisch M, Finotello F, Aigner L, Günther K, Edenhofer F. Single-cell Profiling of Reprogrammed Human Neural Stem Cells Unveils High Similarity to Neural Progenitors in the Developing Central Nervous System. Stem Cell Rev Rep 2024; 20:1325-1339. [PMID: 38519702 PMCID: PMC11222274 DOI: 10.1007/s12015-024-10698-3] [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] [Accepted: 02/14/2024] [Indexed: 03/25/2024]
Abstract
BACKGROUND Similar to induced pluripotent cells (iPSCs), induced neural stem cells (iNSCs) can be directly converted from human somatic cells such as dermal fibroblasts and peripheral blood monocytes. While previous studies have demonstrated the resemblance of iNSCs to neural stem cells derived from primary sources and embryonic stem cells, respectively, a comprehensive analysis of the correlation between iNSCs and their physiological counterparts remained to be investigated. METHODS Nowadays, single-cell sequencing technologies provide unique opportunities for in-depth cellular benchmarking of complex cell populations. Our study involves the comprehensive profiling of converted human iNSCs at a single-cell transcriptomic level, alongside conventional methods, like flow cytometry and immunofluorescence stainings. RESULTS Our results show that the iNSC conversion yields a homogeneous cell population expressing bona fide neural stem cell markers. Extracting transcriptomic signatures from published single cell transcriptomic atlas data and comparison to the iNSC transcriptome reveals resemblance to embryonic neuroepithelial cells of early neurodevelopmental stages observed in vivo at 5 weeks of development. CONCLUSION Our data underscore the physiological relevance of directly converted iNSCs, making them a valuable in vitro system for modeling human central nervous system development and establishing translational applications in cell therapy and compound screening.
Collapse
Affiliation(s)
- Angeliki Spathopoulou
- Department of Molecular Biology & CMBI, Genomics, Stem Cell & Regenerative Medicine Group, University of Innsbruck, Technikerstraße 25, 6020, Innsbruck, Austria
| | - Martina Podlesnic
- Department of Molecular Biology & CMBI, Genomics, Stem Cell & Regenerative Medicine Group, University of Innsbruck, Technikerstraße 25, 6020, Innsbruck, Austria
| | - Laura De Gaetano
- Department of Molecular Biology & CMBI, Genomics, Stem Cell & Regenerative Medicine Group, University of Innsbruck, Technikerstraße 25, 6020, Innsbruck, Austria
| | - Elena Marie Kirsch
- Institute of Molecular Regenerative Medicine, Paracelsus Medical University, Salzburg, Austria
- Center for Stroke Research, Charité - Universitätsmedizin Berlin, Berlin, Germany
- Department of Experimental Neurology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Marcel Tisch
- Department of Molecular Biology & CMBI, Genomics, Stem Cell & Regenerative Medicine Group, University of Innsbruck, Technikerstraße 25, 6020, Innsbruck, Austria
| | - Francesca Finotello
- Department of Molecular Biology, Digital Science Center (DiSC), University of Innsbruck, Innsbruck, Austria
| | - Ludwig Aigner
- Institute of Molecular Regenerative Medicine, Paracelsus Medical University, Salzburg, Austria
| | - Katharina Günther
- Department of Molecular Biology & CMBI, Genomics, Stem Cell & Regenerative Medicine Group, University of Innsbruck, Technikerstraße 25, 6020, Innsbruck, Austria
- Institute of Molecular Regenerative Medicine, Paracelsus Medical University, Salzburg, Austria
| | - Frank Edenhofer
- Department of Molecular Biology & CMBI, Genomics, Stem Cell & Regenerative Medicine Group, University of Innsbruck, Technikerstraße 25, 6020, Innsbruck, Austria.
| |
Collapse
|
14
|
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
|
15
|
Malik JR, Modebelu UO, Fletcher CV, Podany AT, Scarsi KK, Byrareddy SN, Anand RK, Buch S, Sil S, Le J, Bradley JS, Brown AN, Sutar D, Avedissian SN. Establishment of a Four-Cell In Vitro Blood-Brain Barrier Model With Human Primary Brain Cells. Curr Protoc 2024; 4:e1067. [PMID: 38857108 PMCID: PMC11783448 DOI: 10.1002/cpz1.1067] [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] [Indexed: 06/11/2024]
Abstract
The blood-brain barrier (BBB) constitutes a crucial protective anatomical layer with a microenvironment that tightly controls material transit. Constructing an in vitro BBB model to replicate in vivo features requires the sequential layering of constituent cell types. Maintaining heightened integrity in the observed tight junctions during both the establishment and post-experiment phases is crucial to the success of these models. We have developed an in vitro BBB model that replicates the cellular composition and spatial orientation of in vivo BBB observed in humans. The experiment includes comprehensive procedures and steps aimed at enhancing the integration of the four-cell model. Departing from conventional in vitro BBB models, our methodology eliminates the necessity for pre-coated plates to facilitate cell adhesion, thereby improving cell visualization throughout the procedure. An in-house coating strategy and a simple yet effective approach significantly reduce costs and provides superior imaging of cells and corresponding tight junction protein expression. Also, our BBB model includes all four primary cell types that are structural parts of the human BBB. With its innovative and user-friendly features, our in-house optimized in vitro four-cell-based BBB model showcases novel methodology and provides a promising experimental platform for drug screening processes. © 2024 The Authors. Current Protocols published by Wiley Periodicals LLC. Basic Protocol 1: Coating and culture system Basic Protocol 2: Cell seeding and Transwell insert handling Basic Protocol 3: Assessment of model functionality.
Collapse
Affiliation(s)
- Johid R. Malik
- Antiviral Pharmacology Laboratory, College of Pharmacy, University of Nebraska Medical Center, Omaha, Nebraska
- These authors contributed equally to this work
| | - Ukamaka O. Modebelu
- Antiviral Pharmacology Laboratory, College of Pharmacy, University of Nebraska Medical Center, Omaha, Nebraska
- These authors contributed equally to this work
| | - Courtney V. Fletcher
- Antiviral Pharmacology Laboratory, College of Pharmacy, University of Nebraska Medical Center, Omaha, Nebraska
- Division of Infectious Diseases, Department of Medicine, University of Nebraska Medical Center, Omaha, Nebraska
| | - Anthony T. Podany
- Antiviral Pharmacology Laboratory, College of Pharmacy, University of Nebraska Medical Center, Omaha, Nebraska
| | - Kimberly K. Scarsi
- Antiviral Pharmacology Laboratory, College of Pharmacy, University of Nebraska Medical Center, Omaha, Nebraska
- Division of Infectious Diseases, Department of Medicine, University of Nebraska Medical Center, Omaha, Nebraska
| | - Siddappa N. Byrareddy
- Department of Pharmacology & Experimental Neurosciences, University of Nebraska Medical Center, Omaha, Nebraska
| | | | - Shilpa Buch
- Department of Pharmacology & Experimental Neurosciences, University of Nebraska Medical Center, Omaha, Nebraska
| | - Susmia Sil
- Department of Pharmacology & Experimental Neurosciences, University of Nebraska Medical Center, Omaha, Nebraska
| | - Jennifer Le
- University of California San Diego, Skaggs School of Pharmacy and Pharmaceutical Science, La Jolla, California
| | - John S. Bradley
- University of California San Diego School of Medicine, Department of Pediatrics, San Diego, California
| | - Ashley N. Brown
- Institute for Therapeutic Innovation, College of Medicine, University of Florida, Orlando, Florida
| | - Debapriya Sutar
- Department of Pharmacology & Experimental Neurosciences, University of Nebraska Medical Center, Omaha, Nebraska
| | - Sean N. Avedissian
- Antiviral Pharmacology Laboratory, College of Pharmacy, University of Nebraska Medical Center, Omaha, Nebraska
| |
Collapse
|
16
|
Porkoláb G, Mészáros M, Szecskó A, Vigh JP, Walter FR, Figueiredo R, Kálomista I, Hoyk Z, Vizsnyiczai G, Gróf I, Jan JS, Gosselet F, Pirity MK, Vastag M, Hudson N, Campbell M, Veszelka S, Deli MA. Synergistic induction of blood-brain barrier properties. Proc Natl Acad Sci U S A 2024; 121:e2316006121. [PMID: 38748577 PMCID: PMC11126970 DOI: 10.1073/pnas.2316006121] [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: 09/14/2023] [Accepted: 04/05/2024] [Indexed: 05/27/2024] Open
Abstract
Blood-brain barrier (BBB) models derived from human stem cells are powerful tools to improve our understanding of cerebrovascular diseases and to facilitate drug development for the human brain. Yet providing stem cell-derived endothelial cells with the right signaling cues to acquire BBB characteristics while also retaining their vascular identity remains challenging. Here, we show that the simultaneous activation of cyclic AMP and Wnt/β-catenin signaling and inhibition of the TGF-β pathway in endothelial cells robustly induce BBB properties in vitro. To target this interaction, we present a small-molecule cocktail named cARLA, which synergistically enhances barrier tightness in a range of BBB models across species. Mechanistically, we reveal that the three pathways converge on Wnt/β-catenin signaling to mediate the effect of cARLA via the tight junction protein claudin-5. We demonstrate that cARLA shifts the gene expressional profile of human stem cell-derived endothelial cells toward the in vivo brain endothelial signature, with a higher glycocalyx density and efflux pump activity, lower rates of endocytosis, and a characteristic endothelial response to proinflammatory cytokines. Finally, we illustrate how cARLA can improve the predictive value of human BBB models regarding the brain penetration of drugs and targeted nanoparticles. Due to its synergistic effect, high reproducibility, and ease of use, cARLA has the potential to advance drug development for the human brain by improving BBB models across laboratories.
Collapse
Affiliation(s)
- Gergő Porkoláb
- Institute of Biophysics, Biological Research Centre, Hungarian Research Network, SzegedH-6726, Hungary
- Doctoral School of Biology, University of Szeged, SzegedH-6720, Hungary
| | - Mária Mészáros
- Institute of Biophysics, Biological Research Centre, Hungarian Research Network, SzegedH-6726, Hungary
| | - Anikó Szecskó
- Institute of Biophysics, Biological Research Centre, Hungarian Research Network, SzegedH-6726, Hungary
- Doctoral School of Biology, University of Szeged, SzegedH-6720, Hungary
| | - Judit P. Vigh
- Institute of Biophysics, Biological Research Centre, Hungarian Research Network, SzegedH-6726, Hungary
- Doctoral School of Biology, University of Szeged, SzegedH-6720, Hungary
| | - Fruzsina R. Walter
- Institute of Biophysics, Biological Research Centre, Hungarian Research Network, SzegedH-6726, Hungary
| | | | - Ildikó Kálomista
- In Vitro Metabolism Laboratory, Gedeon Richter, BudapestH-1103, Hungary
| | - Zsófia Hoyk
- Institute of Biophysics, Biological Research Centre, Hungarian Research Network, SzegedH-6726, Hungary
| | - Gaszton Vizsnyiczai
- Institute of Biophysics, Biological Research Centre, Hungarian Research Network, SzegedH-6726, Hungary
| | - Ilona Gróf
- Institute of Biophysics, Biological Research Centre, Hungarian Research Network, SzegedH-6726, Hungary
| | - Jeng-Shiung Jan
- Department of Chemical Engineering, National Cheng Kung University, Tainan70101, Taiwan
| | - Fabien Gosselet
- Laboratoire de la Barriére Hémato-Encéphalique, Université d’Artois, Lens62307, France
| | - Melinda K. Pirity
- Institute of Genetics, Biological Research Centre, Hungarian Research Network, SzegedH-6726, Hungary
| | - Monika Vastag
- In Vitro Metabolism Laboratory, Gedeon Richter, BudapestH-1103, Hungary
| | - Natalie Hudson
- Smurfit Institute of Genetics, Trinity College Dublin, DublinD02 VF25, Ireland
| | - Matthew Campbell
- Smurfit Institute of Genetics, Trinity College Dublin, DublinD02 VF25, Ireland
| | - Szilvia Veszelka
- Institute of Biophysics, Biological Research Centre, Hungarian Research Network, SzegedH-6726, Hungary
| | - Mária A. Deli
- Institute of Biophysics, Biological Research Centre, Hungarian Research Network, SzegedH-6726, Hungary
| |
Collapse
|
17
|
Tal T, Myhre O, Fritsche E, Rüegg J, Craenen K, Aiello-Holden K, Agrillo C, Babin PJ, Escher BI, Dirven H, Hellsten K, Dolva K, Hessel E, Heusinkveld HJ, Hadzhiev Y, Hurem S, Jagiello K, Judzinska B, Klüver N, Knoll-Gellida A, Kühne BA, Leist M, Lislien M, Lyche JL, Müller F, Colbourne JK, Neuhaus W, Pallocca G, Seeger B, Scharkin I, Scholz S, Spjuth O, Torres-Ruiz M, Bartmann K. New approach methods to assess developmental and adult neurotoxicity for regulatory use: a PARC work package 5 project. FRONTIERS IN TOXICOLOGY 2024; 6:1359507. [PMID: 38742231 PMCID: PMC11089904 DOI: 10.3389/ftox.2024.1359507] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Accepted: 03/18/2024] [Indexed: 05/16/2024] Open
Abstract
In the European regulatory context, rodent in vivo studies are the predominant source of neurotoxicity information. Although they form a cornerstone of neurotoxicological assessments, they are costly and the topic of ethical debate. While the public expects chemicals and products to be safe for the developing and mature nervous systems, considerable numbers of chemicals in commerce have not, or only to a limited extent, been assessed for their potential to cause neurotoxicity. As such, there is a societal push toward the replacement of animal models with in vitro or alternative methods. New approach methods (NAMs) can contribute to the regulatory knowledge base, increase chemical safety, and modernize chemical hazard and risk assessment. Provided they reach an acceptable level of regulatory relevance and reliability, NAMs may be considered as replacements for specific in vivo studies. The European Partnership for the Assessment of Risks from Chemicals (PARC) addresses challenges to the development and implementation of NAMs in chemical risk assessment. In collaboration with regulatory agencies, Project 5.2.1e (Neurotoxicity) aims to develop and evaluate NAMs for developmental neurotoxicity (DNT) and adult neurotoxicity (ANT) and to understand the applicability domain of specific NAMs for the detection of endocrine disruption and epigenetic perturbation. To speed up assay time and reduce costs, we identify early indicators of later-onset effects. Ultimately, we will assemble second-generation developmental neurotoxicity and first-generation adult neurotoxicity test batteries, both of which aim to provide regulatory hazard and risk assessors and industry stakeholders with robust, speedy, lower-cost, and informative next-generation hazard and risk assessment tools.
Collapse
Affiliation(s)
- Tamara Tal
- Helmholtz Centre for Environmental Research – UFZ, Chemicals in the Environment Research Section, Leipzig, Germany
- University of Leipzig, Medical Faculty, Leipzig, Germany
| | - Oddvar Myhre
- Norwegian Institute of Public Health – NIPH, Department of Chemical Toxicology, Oslo, Norway
| | - Ellen Fritsche
- IUF – Leibniz Research Institute for Environmental Medicine, Düsseldorf, Germany
- DNTOX GmbH, Düsseldorf, Germany
- Swiss Centre for Applied Human Toxicology, University of Basel, Basel, Switzerland
| | - Joëlle Rüegg
- Uppsala University, Department of Organismal Biology, Uppsala, Sweden
| | - Kai Craenen
- European Chemicals Agency (ECHA), Helsinki, Finland
| | | | - Caroline Agrillo
- Uppsala University, Department of Organismal Biology, Uppsala, Sweden
| | - Patrick J. Babin
- Université de Bordeaux, Institut National de la Santé et de la Recherche Médicale (INSERM), Maladies Rares: Génétique et Métabolisme (MRGM), Pessac, France
| | - Beate I. Escher
- Helmholtz Centre for Environmental Research – UFZ, Chemicals in the Environment Research Section, Leipzig, Germany
| | - Hubert Dirven
- Norwegian Institute of Public Health – NIPH, Department of Chemical Toxicology, Oslo, Norway
| | | | - Kristine Dolva
- University of Oslo, Section for Pharmacology and Pharmaceutical Biosciences, Department of Pharmacy, Olso, Norway
| | - Ellen Hessel
- Dutch Nation Institute for Public Health and the Environment (RIVM), Centre for Health Protection, Bilthoven, Netherlands
| | - Harm J. Heusinkveld
- Dutch Nation Institute for Public Health and the Environment (RIVM), Centre for Health Protection, Bilthoven, Netherlands
| | - Yavor Hadzhiev
- University of Birmingham, Centre for Environmental Research and Justice, Birmingham, UK
| | - Selma Hurem
- Norwegian University of Life Sciences (NMBU), Faculty of Veterinary Medicine, Ås, Norway
| | - Karolina Jagiello
- University of Gdansk, Laboratory of Environmental Chemoinformatics, Gdansk, Poland
| | - Beata Judzinska
- University of Gdansk, Laboratory of Environmental Chemoinformatics, Gdansk, Poland
| | - Nils Klüver
- Helmholtz Centre for Environmental Research – UFZ, Chemicals in the Environment Research Section, Leipzig, Germany
| | - Anja Knoll-Gellida
- Université de Bordeaux, Institut National de la Santé et de la Recherche Médicale (INSERM), Maladies Rares: Génétique et Métabolisme (MRGM), Pessac, France
| | - Britta A. Kühne
- University of Veterinary Medicine Hannover, Foundation, Institute for Food Quality and Food Safety, Hannover, Germany
| | - Marcel Leist
- University of Konstanz, In Vitro Toxicology and Biomedicine/CAAT-Europe, Konstanz, Germany
| | - Malene Lislien
- Norwegian Institute of Public Health – NIPH, Department of Chemical Toxicology, Oslo, Norway
| | - Jan L. Lyche
- Norwegian University of Life Sciences (NMBU), Faculty of Veterinary Medicine, Ås, Norway
| | - Ferenc Müller
- University of Birmingham, Centre for Environmental Research and Justice, Birmingham, UK
| | - John K. Colbourne
- University of Birmingham, Centre for Environmental Research and Justice, Birmingham, UK
| | - Winfried Neuhaus
- AIT Austrian Institute of Technology GmbH, Competence Unit Molecular Diagnostics, Center Health and Bioresources, Vienna, Austria
- Danube Private University, Faculty of Dentistry and Medicine, Department of Medicine, Krems, Austria
| | - Giorgia Pallocca
- University of Konstanz, In Vitro Toxicology and Biomedicine/CAAT-Europe, Konstanz, Germany
| | - Bettina Seeger
- University of Veterinary Medicine Hannover, Foundation, Institute for Food Quality and Food Safety, Hannover, Germany
| | - Ilka Scharkin
- IUF – Leibniz Research Institute for Environmental Medicine, Düsseldorf, Germany
| | - Stefan Scholz
- Helmholtz Centre for Environmental Research – UFZ, Chemicals in the Environment Research Section, Leipzig, Germany
| | - Ola Spjuth
- Uppsala University and Science for Life Laboratory, Department of Pharmaceutical Biosciences, Uppsala, Sweden
| | - Monica Torres-Ruiz
- Instituto de Salud Carlos III (ISCIII), Centro Nacional de Sanidad Ambiental (CNSA), Environmental Toxicology Unit, Majadahonda, Spain
| | - Kristina Bartmann
- IUF – Leibniz Research Institute for Environmental Medicine, Düsseldorf, Germany
- DNTOX GmbH, Düsseldorf, Germany
| |
Collapse
|
18
|
Barnett AM, Mullaney JA, McNabb WC, Roy NC. Culture media and format alter cellular composition and barrier integrity of porcine colonoid-derived monolayers. Tissue Barriers 2024; 12:2222632. [PMID: 37340938 DOI: 10.1080/21688370.2023.2222632] [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: 03/01/2023] [Accepted: 06/04/2023] [Indexed: 06/22/2023] Open
Abstract
Intestinal organoid technology has revolutionized our approach to in vitro cell culture due in part to their three-dimensional structures being more like the native tissue from which they were derived with respect to cellular composition and architecture. For this reason, organoids are becoming the new gold standard for undertaking intestinal epithelial cell research. Unfortunately, their otherwise advantageous three-dimensional geometry prevents easy access to the apical epithelium, which is a major limitation when studying interactions between dietary or microbial components and host tissues. To overcome this problem, we developed porcine colonoid-derived monolayers cultured on both permeable Transwell inserts and tissue culture treated polystyrene plates. We found that seeding density and culture format altered the expression of genes encoding markers of specific cell types (stem cells, colonocytes, goblets, and enteroendocrine cells), and barrier maturation (tight junctions). Additionally, we found that changes to the formulation of the culture medium altered the cellular composition of colonoids and of monolayers derived from them, resulting in cultures with an increasingly differentiated phenotype that was similar to that of their tissue of origin.
Collapse
Affiliation(s)
- Alicia M Barnett
- AgResearch Ltd, Grasslands Research Centre, Palmerston North, New Zealand
- Riddet Institute, Massey University, Palmerston North, New Zealand
| | - Jane A Mullaney
- AgResearch Ltd, Grasslands Research Centre, Palmerston North, New Zealand
- Riddet Institute, Massey University, Palmerston North, New Zealand
- Liggins Institute, The High-Value Nutrition National Science Challenge, Auckland, New Zealand
| | - Warren C McNabb
- Riddet Institute, Massey University, Palmerston North, New Zealand
- Liggins Institute, The High-Value Nutrition National Science Challenge, Auckland, New Zealand
| | - Nicole C Roy
- Riddet Institute, Massey University, Palmerston North, New Zealand
- Liggins Institute, The High-Value Nutrition National Science Challenge, Auckland, New Zealand
- Department of Nutrition, The University of Otago, Dunedin, New Zealand
| |
Collapse
|
19
|
Watson BE, Miles JA, Moss MA. Human in vitro blood barrier models: architectures and applications. Tissue Barriers 2024; 12:2222628. [PMID: 37339009 PMCID: PMC11042067 DOI: 10.1080/21688370.2023.2222628] [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: 03/01/2023] [Revised: 05/28/2023] [Accepted: 06/04/2023] [Indexed: 06/22/2023] Open
Abstract
Blood barriers serve as key points of transport for essential molecules as well as lines of defense to protect against toxins. In vitro modeling of these barriers is common practice in the study of their physiology and related diseases. This review describes a common method of using an adaptable, low cost, semipermeable, suspended membrane to experimentally model three blood barriers in the human body: the blood-brain barrier (BBB), the gut-blood barrier (GBB), and the air-blood barrier (ABB). The GBB and ABB both protect from the outside environment, while the BBB protects the central nervous system from potential neurotoxic agents in the blood. These barriers share several commonalities, including the formation of tight junctions, polarized cellular monolayers, and circulatory system contact. Cell architectures used to mimic barrier anatomy as well as applications to study function, dysfunction, and response provide an overview of the versatility enabled by these cultural systems.
Collapse
Affiliation(s)
| | - Julia A. Miles
- Biomedical Engineering Program, Univ of South Carolina, Columbia, SCUSA
| | - Melissa A. Moss
- Biomedical Engineering Program, Univ of South Carolina, Columbia, SCUSA
- Department of Chemical Engineering, Univ of South Carolina, Columbia, SCUSA
| |
Collapse
|
20
|
Xavier G, Navarrete Santos A, Hartmann C, Santoro ML, Flegel N, Reinsch J, Majer A, Ehrhardt T, Pfeifer J, Simm A, Hollemann T, Belangero SI, Rujescu D, Jung M. Comparison of Extracellular Vesicles from Induced Pluripotent Stem Cell-Derived Brain Cells. Int J Mol Sci 2024; 25:3575. [PMID: 38612385 PMCID: PMC11011287 DOI: 10.3390/ijms25073575] [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: 02/13/2024] [Revised: 03/08/2024] [Accepted: 03/18/2024] [Indexed: 04/14/2024] Open
Abstract
The pathophysiology of many neuropsychiatric disorders is still poorly understood. Identification of biomarkers for these diseases could benefit patients due to better classification and stratification. Exosomes excreted into the circulatory system can cross the blood-brain barrier and carry a cell type-specific set of molecules. Thus, exosomes are a source of potential biomarkers for many diseases, including neuropsychiatric disorders. Here, we investigated exosomal proteins produced from human-induced pluripotent stem cells (iPSCs) and iPSC-derived neural stem cells, neural progenitors, neurons, astrocytes, microglia-like cells, and brain capillary endothelial cells. Of the 31 exosome surface markers analyzed, a subset of biomarkers were significantly enriched in astrocytes (CD29, CD44, and CD49e), microglia-like cells (CD44), and neural stem cells (SSEA4). To identify molecular fingerprints associated with disease, circulating exosomes derived from healthy control (HC) individuals were compared against schizophrenia (SCZ) patients and late-onset Alzheimer's disease (LOAD) patients. A significant epitope pattern was identified for LOAD (CD1c and CD2) but not for SCZ compared to HC. Thus, analysis of cell type- and disease-specific exosome signatures of iPSC-derived cell cultures may provide a valuable model system to explore proteomic biomarkers for the identification of novel disease profiles.
Collapse
Affiliation(s)
- Gabriela Xavier
- LiNC—Laboratory of Integrative Neuroscience, Universidade Federal de São Paulo (UNIFESP), São Paulo CEP 05039-032, Brazil
- Genetics Division, Department of Morphology and Genetics, Universidade Federal de São Paulo (UNIFESP), São Paulo CEP 04023-900, Brazil
| | - Alexander Navarrete Santos
- Centre for Medical Basic Research, Medical Faculty, Martin Luther University Halle-Wittenberg, 06120 Halle (Saale), Germany
| | - Carla Hartmann
- Institute for Physiological Chemistry, Medical Faculty, Martin Luther University Halle-Wittenberg, 06114 Halle (Saale), Germany; (C.H.)
| | - Marcos L. Santoro
- LiNC—Laboratory of Integrative Neuroscience, Universidade Federal de São Paulo (UNIFESP), São Paulo CEP 05039-032, Brazil
- Genetics Division, Department of Morphology and Genetics, Universidade Federal de São Paulo (UNIFESP), São Paulo CEP 04023-900, Brazil
| | - Nicole Flegel
- Institute for Biochemistry, Friedrich Alexander University Erlangen-Nürnberg, 91054 Erlangen, Germany;
| | - Jessica Reinsch
- Institute for Physiological Chemistry, Medical Faculty, Martin Luther University Halle-Wittenberg, 06114 Halle (Saale), Germany; (C.H.)
| | - Annika Majer
- Institute for Physiological Chemistry, Medical Faculty, Martin Luther University Halle-Wittenberg, 06114 Halle (Saale), Germany; (C.H.)
| | - Toni Ehrhardt
- Institute for Physiological Chemistry, Medical Faculty, Martin Luther University Halle-Wittenberg, 06114 Halle (Saale), Germany; (C.H.)
| | - Jenny Pfeifer
- Institute for Physiological Chemistry, Medical Faculty, Martin Luther University Halle-Wittenberg, 06114 Halle (Saale), Germany; (C.H.)
| | - Andreas Simm
- Clinic for Cardiac and Thoracic Surgery, Martin Luther University Halle-Wittenberg, 06120 Halle (Saale), Germany
| | - Thomas Hollemann
- Institute for Physiological Chemistry, Medical Faculty, Martin Luther University Halle-Wittenberg, 06114 Halle (Saale), Germany; (C.H.)
| | - Sintia I. Belangero
- LiNC—Laboratory of Integrative Neuroscience, Universidade Federal de São Paulo (UNIFESP), São Paulo CEP 05039-032, Brazil
- Genetics Division, Department of Morphology and Genetics, Universidade Federal de São Paulo (UNIFESP), São Paulo CEP 04023-900, Brazil
| | - Dan Rujescu
- Department of Psychiatry and Psychotherapy, Medical University of Vienna, 1090 Vienna, Austria
| | - Matthias Jung
- Institute for Physiological Chemistry, Medical Faculty, Martin Luther University Halle-Wittenberg, 06114 Halle (Saale), Germany; (C.H.)
| |
Collapse
|
21
|
Ko J, Song J, Choi N, Kim HN. Patient-Derived Microphysiological Systems for Precision Medicine. Adv Healthc Mater 2024; 13:e2303161. [PMID: 38010253 PMCID: PMC11469251 DOI: 10.1002/adhm.202303161] [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: 11/06/2023] [Indexed: 11/29/2023]
Abstract
Patient-derived microphysiological systems (P-MPS) have emerged as powerful tools in precision medicine that provide valuable insight into individual patient characteristics. This review discusses the development of P-MPS as an integration of patient-derived samples, including patient-derived cells, organoids, and induced pluripotent stem cells, into well-defined MPSs. Emphasizing the necessity of P-MPS development, its significance as a nonclinical assessment approach that bridges the gap between traditional in vitro models and clinical outcomes is highlighted. Additionally, guidance is provided for engineering approaches to develop microfluidic devices and high-content analysis for P-MPSs, enabling high biological relevance and high-throughput experimentation. The practical implications of the P-MPS are further examined by exploring the clinically relevant outcomes obtained from various types of patient-derived samples. The construction and analysis of these diverse samples within the P-MPS have resulted in physiologically relevant data, paving the way for the development of personalized treatment strategies. This study describes the significance of the P-MPS in precision medicine, as well as its unique capacity to offer valuable insights into individual patient characteristics.
Collapse
Affiliation(s)
- Jihoon Ko
- Department of BioNano TechnologyGachon UniversitySeongnam‐siGyeonggi‐do13120Republic of Korea
| | - Jiyoung Song
- Brain Science InstituteKorea Institute of Science and Technology (KIST)Seoul02792Republic of Korea
| | - Nakwon Choi
- Brain Science InstituteKorea Institute of Science and Technology (KIST)Seoul02792Republic of Korea
- Division of Bio‐Medical Science & TechnologyKIST SchoolSeoul02792Republic of Korea
- KU‐KIST Graduate School of Converging Science and TechnologyKorea UniversitySeoul02841Republic of Korea
| | - Hong Nam Kim
- Brain Science InstituteKorea Institute of Science and Technology (KIST)Seoul02792Republic of Korea
- Division of Bio‐Medical Science & TechnologyKIST SchoolSeoul02792Republic of Korea
- School of Mechanical EngineeringYonsei UniversitySeoul03722Republic of Korea
- Yonsei‐KIST Convergence Research InstituteYonsei UniversitySeoul03722Republic of Korea
| |
Collapse
|
22
|
Deli MA, Porkoláb G, Kincses A, Mészáros M, Szecskó A, Kocsis AE, Vigh JP, Valkai S, Veszelka S, Walter FR, Dér A. Lab-on-a-chip models of the blood-brain barrier: evolution, problems, perspectives. LAB ON A CHIP 2024; 24:1030-1063. [PMID: 38353254 DOI: 10.1039/d3lc00996c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/28/2024]
Abstract
A great progress has been made in the development and use of lab-on-a-chip devices to model and study the blood-brain barrier (BBB) in the last decade. We present the main types of BBB-on-chip models and their use for the investigation of BBB physiology, drug and nanoparticle transport, toxicology and pathology. The selection of the appropriate cell types to be integrated into BBB-on-chip devices is discussed, as this greatly impacts the physiological relevance and translatability of findings. We identify knowledge gaps, neglected engineering and cell biological aspects and point out problems and contradictions in the literature of BBB-on-chip models, and suggest areas for further studies to progress this highly interdisciplinary field. BBB-on-chip models have an exceptional potential as predictive tools and alternatives of animal experiments in basic and preclinical research. To exploit the full potential of this technique expertise from materials science, bioengineering as well as stem cell and vascular/BBB biology is necessary. There is a need for better integration of these diverse disciplines that can only be achieved by setting clear parameters for characterizing both the chip and the BBB model parts technically and functionally.
Collapse
Affiliation(s)
- Mária A Deli
- HUN-REN Biological Research Centre, Institute of Biophysics, Szeged, Hungary.
| | - Gergő Porkoláb
- HUN-REN Biological Research Centre, Institute of Biophysics, Szeged, Hungary.
- Doctoral School of Biology, University of Szeged, Hungary
| | - András Kincses
- HUN-REN Biological Research Centre, Institute of Biophysics, Szeged, Hungary.
| | - Mária Mészáros
- HUN-REN Biological Research Centre, Institute of Biophysics, Szeged, Hungary.
| | - Anikó Szecskó
- HUN-REN Biological Research Centre, Institute of Biophysics, Szeged, Hungary.
- Doctoral School of Biology, University of Szeged, Hungary
| | - Anna E Kocsis
- HUN-REN Biological Research Centre, Institute of Biophysics, Szeged, Hungary.
| | - Judit P Vigh
- HUN-REN Biological Research Centre, Institute of Biophysics, Szeged, Hungary.
- Doctoral School of Biology, University of Szeged, Hungary
| | - Sándor Valkai
- HUN-REN Biological Research Centre, Institute of Biophysics, Szeged, Hungary.
| | - Szilvia Veszelka
- HUN-REN Biological Research Centre, Institute of Biophysics, Szeged, Hungary.
| | - Fruzsina R Walter
- HUN-REN Biological Research Centre, Institute of Biophysics, Szeged, Hungary.
| | - András Dér
- HUN-REN Biological Research Centre, Institute of Biophysics, Szeged, Hungary.
| |
Collapse
|
23
|
Körtge A, Breitrück A, Doß S, Hofrichter J, Nelz SC, Krüsemann H, Wasserkort R, Fitzner B, Hecker M, Mitzner S, Zettl UK. The Utility of Miniaturized Adsorbers in Exploring the Cellular and Molecular Effects of Blood Purification: A Pilot Study with a Focus on Immunoadsorption in Multiple Sclerosis. Int J Mol Sci 2024; 25:2590. [PMID: 38473836 DOI: 10.3390/ijms25052590] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Revised: 02/02/2024] [Accepted: 02/09/2024] [Indexed: 03/14/2024] Open
Abstract
Immunoadsorption (IA) has proven to be clinically effective in the treatment of steroid-refractory multiple sclerosis (MS) relapses, but its mechanism of action remains unclear. We used miniaturized adsorber devices with a tryptophan-immobilized polyvinyl alcohol (PVA) gel sorbent to mimic the IA treatment of patients with MS in vitro. The plasma was screened before and after adsorption with regard to disease-specific mediators, and the effect of the IA treatment on the migration of neutrophils and the integrity of the endothelial cell barrier was tested in cell-based models. The in vitro IA treatment with miniaturized adsorbers resulted in reduced plasma levels of cytokines and chemokines. We also found a reduced migration of neutrophils towards patient plasma treated with the adsorbers. Furthermore, the IA-treated plasma had a positive effect on the endothelial cell barrier's integrity in the cell culture model. Our findings suggest that IA results in a reduced infiltration of cells into the central nervous system by reducing leukocyte transmigration and preventing blood-brain barrier breakdown. This novel approach of performing in vitro blood purification therapies on actual patient samples with miniaturized adsorbers and testing their effects in cell-based assays that investigate specific hypotheses of the pathophysiology provides a promising platform for elucidating the mechanisms of action of those therapies in various diseases.
Collapse
Affiliation(s)
- Andreas Körtge
- Department of Extracorporeal Therapy Systems, Fraunhofer Institute for Cell Therapy and Immunology IZI, 18057 Rostock, Germany
- Division of Nephrology, Center for Internal Medicine, Rostock University Medical Center, 18057 Rostock, Germany
| | - Anne Breitrück
- Department of Extracorporeal Therapy Systems, Fraunhofer Institute for Cell Therapy and Immunology IZI, 18057 Rostock, Germany
- Division of Nephrology, Center for Internal Medicine, Rostock University Medical Center, 18057 Rostock, Germany
| | - Sandra Doß
- Department of Extracorporeal Therapy Systems, Fraunhofer Institute for Cell Therapy and Immunology IZI, 18057 Rostock, Germany
| | - Jacqueline Hofrichter
- Department of Extracorporeal Therapy Systems, Fraunhofer Institute for Cell Therapy and Immunology IZI, 18057 Rostock, Germany
- Division of Nephrology, Center for Internal Medicine, Rostock University Medical Center, 18057 Rostock, Germany
| | - Sophie-Charlotte Nelz
- Division of Nephrology, Center for Internal Medicine, Rostock University Medical Center, 18057 Rostock, Germany
| | - Horst Krüsemann
- Department of Extracorporeal Therapy Systems, Fraunhofer Institute for Cell Therapy and Immunology IZI, 18057 Rostock, Germany
| | - Reinhold Wasserkort
- Department of Extracorporeal Therapy Systems, Fraunhofer Institute for Cell Therapy and Immunology IZI, 18057 Rostock, Germany
- Division of Nephrology, Center for Internal Medicine, Rostock University Medical Center, 18057 Rostock, Germany
| | - Brit Fitzner
- Division of Neuroimmunology, Department of Neurology, Rostock University Medical Center, 18147 Rostock, Germany
| | - Michael Hecker
- Division of Neuroimmunology, Department of Neurology, Rostock University Medical Center, 18147 Rostock, Germany
| | - Steffen Mitzner
- Department of Extracorporeal Therapy Systems, Fraunhofer Institute for Cell Therapy and Immunology IZI, 18057 Rostock, Germany
- Division of Nephrology, Center for Internal Medicine, Rostock University Medical Center, 18057 Rostock, Germany
| | - Uwe Klaus Zettl
- Division of Neuroimmunology, Department of Neurology, Rostock University Medical Center, 18147 Rostock, Germany
| |
Collapse
|
24
|
Vollmuth N, Sin J, Kim BJ. Host-microbe interactions at the blood-brain barrier through the lens of induced pluripotent stem cell-derived brain-like endothelial cells. mBio 2024; 15:e0286223. [PMID: 38193670 PMCID: PMC10865987 DOI: 10.1128/mbio.02862-23] [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] [Indexed: 01/10/2024] Open
Abstract
Microbe-induced meningoencephalitis/meningitis is a life-threatening infection of the central nervous system (CNS) that occurs when pathogens are able to cross the blood-brain barrier (BBB) and gain access to the CNS. The BBB consists of highly specialized brain endothelial cells that exhibit specific properties to allow tight regulation of CNS homeostasis and prevent pathogen crossing. However, during meningoencephalitis/meningitis, the BBB fails to protect the CNS. Modeling the BBB remains a challenge due to the specialized characteristics of these cells. In this review, we cover the induced pluripotent stem cell-derived, brain-like endothelial cell model during host-pathogen interaction, highlighting the strengths and recent work on various pathogens known to interact with the BBB. As stem cell technologies are becoming more prominent, the stem cell-derived, brain-like endothelial cell model has been able to reveal new insights in vitro, which remain challenging with other in vitro cell-based models consisting of primary human brain endothelial cells and immortalized human brain endothelial cell lines.
Collapse
Affiliation(s)
- Nadine Vollmuth
- Department of Biological Sciences, University of Alabama, Tuscaloosa, Alabama, USA
| | - Jon Sin
- Department of Biological Sciences, University of Alabama, Tuscaloosa, Alabama, USA
| | - Brandon J. Kim
- Department of Biological Sciences, University of Alabama, Tuscaloosa, Alabama, USA
- Department of Microbiology, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
- Center for Convergent Biosciences and Medicine, University of Alabama, Tuscaloosa, Alabama, USA
- Alabama Life Research Institute, University of Alabama, Tuscaloosa, Alabama, USA
| |
Collapse
|
25
|
Heo C, Kwak HJ, Ngo LH, Woo RS, Lee SJ. Implementation of the neuro-glia-vascular unit through co-culture of adult neural stem cells and vascular cells and transcriptomic analysis of diverse Aβ assembly types. J Neurosci Methods 2024; 402:110029. [PMID: 38042304 DOI: 10.1016/j.jneumeth.2023.110029] [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: 08/10/2023] [Revised: 11/05/2023] [Accepted: 11/28/2023] [Indexed: 12/04/2023]
Abstract
BACKGROUND The blood-brain barrier (BBB) is a specialized layer between blood vessels and tissue in the brain, which is comprised of a neuro-glia-vascular (NGV) unit, thus play a vital role in various brain diseases. NEW METHOD We developed the in vitro NGV units by co-culturing brain microvascular endothelial cells (BMECs; bEnd.3) and primary neural stem cells extracted from subventricular zone of adult mice. This approach was designed to mimic the RNA profile conditions found in the microvessels of a mouse brain and confirmed through various comparative transcriptome analyses. RESULTS Optimal NGV unit development was achieved by adjusting cell density-dependent co-culture ratios. Specifically, the morphogenic development and neuronal association of astrocyte endfeet were well observed in the contact region with BMECs in the NGV unit. Through transcriptome analysis, we compared co-cultured bEnd.3/NSCs with monocultured bEnd.3 or NSCs and additionally compared them with previously reported mouse brain vascular tissue to show that this NGV unit model is a suitable in vitro model for neurological disease such as Alzheimer's disease (AD). COMPARISON WITH EXISTING METHOD(S) This in vitro NGV unit was formed from neural stem cells and vascular cells in the brain of adult mice, not embryos. It is very useful for studying brain disease mechanisms by identifying proteins and genes associated with diseases progress. CONCLUSIONS We suggest that this simple in vitro NGV model is appropriate to investigate the relationship between BBB changes and pathological factors in the fields of neurovascular biology and cerebrovascular diseases including AD.
Collapse
Affiliation(s)
- Chaejeong Heo
- Center for Integrated Nanostructure Physics (CINAP), Institute for Basic Science (IBS), Suwon 16419, South Korea; Institute for Quantum Biophysics (IQB), Department of Biophysics, Sungkyunkwan University, Suwon 16419, South Korea
| | - Hee-Jin Kwak
- Center for Integrated Nanostructure Physics (CINAP), Institute for Basic Science (IBS), Suwon 16419, South Korea
| | - Long Hoang Ngo
- Department of Bioactive Material Sciences and Research Center of Bioactive Materials, Jeonbuk National University, Jeonju, Jeollabuk-do 54896, South Korea
| | - Ran-Sook Woo
- Department of Anatomy and Neuroscience, College of Medicine, Eulji University, Daejeon 34824, South Korea.
| | - Sook-Jeong Lee
- Department of Bioactive Material Sciences and Research Center of Bioactive Materials, Jeonbuk National University, Jeonju, Jeollabuk-do 54896, South Korea.
| |
Collapse
|
26
|
Ozgür B, Puris E, Brachner A, Appelt-Menzel A, Oerter S, Balzer V, Holst MR, Christiansen RF, Hyldig K, Buckley ST, Kristensen M, Auriola S, Jensen A, Fricker G, Nielsen MS, Neuhaus W, Brodin B. Characterization of an iPSC-based barrier model for blood-brain barrier investigations using the SBAD0201 stem cell line. Fluids Barriers CNS 2023; 20:96. [PMID: 38115090 PMCID: PMC10731806 DOI: 10.1186/s12987-023-00501-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Accepted: 12/07/2023] [Indexed: 12/21/2023] Open
Abstract
BACKGROUND Blood-brain barrier (BBB) models based on primary murine, bovine, and porcine brain capillary endothelial cell cultures have long been regarded as robust models with appropriate properties to examine the functional transport of small molecules. However, species differences sometimes complicate translating results from these models to human settings. During the last decade, brain capillary endothelial-like cells (BCECs) have been generated from stem cell sources to model the human BBB in vitro. The aim of the present study was to establish and characterize a human BBB model using human induced pluripotent stem cell (hiPSC)-derived BCECs from the hIPSC line SBAD0201. METHODS The model was evaluated using transcriptomics, proteomics, immunocytochemistry, transendothelial electrical resistance (TEER) measurements, and, finally, transport assays to assess the functionality of selected transporters and receptor (GLUT-1, LAT-1, P-gp and LRP-1). RESULTS The resulting BBB model displayed an average TEER of 5474 ± 167 Ω·cm2 and cell monolayer formation with claudin-5, ZO-1, and occludin expression in the tight junction zones. The cell monolayers expressed the typical BBB markers VE-cadherin, VWF, and PECAM-1. Transcriptomics and quantitative targeted absolute proteomics analyses revealed that solute carrier (SLC) transporters were found in high abundance, while the expression of efflux transporters was relatively low. Transport assays using GLUT-1, LAT-1, and LRP-1 substrates and inhibitors confirmed the functional activities of these transporters and receptors in the model. A transport assay suggested that P-gp was not functionally expressed in the model, albeit antibody staining revealed that P-gp was localized at the luminal membrane. CONCLUSIONS In conclusion, the novel SBAD0201-derived BBB model formed tight monolayers and was proven useful for studies investigating GLUT-1, LAT-1, and LRP-1 mediated transport across the BBB. However, the model did not express functional P-gp and thus is not suitable for the performance of drug efflux P-gp reletated studies.
Collapse
Affiliation(s)
- Burak Ozgür
- Department of Pharmacy, University of Copenhagen, Universitetsparken 2, Copenhagen, DK-2100, Denmark
- Biotherapeutic Discovery, H. Lundbeck A/S, Valby, DK-2500, Denmark
| | - Elena Puris
- Institute of Pharmacy and Molecular Biotechnology, Ruprecht-Karls-University, Heidelberg, Germany
| | - Andreas Brachner
- AIT - Austrian Institute of Technology GmbH, Vienna, 1210, Austria
| | - Antje Appelt-Menzel
- Chair Tissue Engineering and Regenerative Medicine (TERM), University Hospital Würzburg, 97070, Würzburg, Germany
- Fraunhofer Institute for Silicate Research ISC, Translational Center Regenerative Therapies (TLC-RT) Röntgenring 11, 97070, Würzburg, Germany
| | - Sabrina Oerter
- Chair Tissue Engineering and Regenerative Medicine (TERM), University Hospital Würzburg, 97070, Würzburg, Germany
- Fraunhofer Institute for Silicate Research ISC, Translational Center Regenerative Therapies (TLC-RT) Röntgenring 11, 97070, Würzburg, Germany
| | - Viktor Balzer
- Institute of Pharmacy and Molecular Biotechnology, Ruprecht-Karls-University, Heidelberg, Germany
| | | | | | - Kathrine Hyldig
- Biotherapeutic Discovery, H. Lundbeck A/S, Valby, DK-2500, Denmark
- Department of Biomedicine, Aarhus University, Aarhus, DK-8000, Denmark
| | - Stephen T Buckley
- Global Research Technologies, Novo Nordisk A/S, Måløv, DK-2760, Denmark
| | - Mie Kristensen
- Department of Pharmacy, University of Copenhagen, Universitetsparken 2, Copenhagen, DK-2100, Denmark
| | - Seppo Auriola
- School of Pharmacy, University of Eastern Finland, Kuopio, Finland
| | - Allan Jensen
- Biotherapeutic Discovery, H. Lundbeck A/S, Valby, DK-2500, Denmark
| | - Gert Fricker
- Institute of Pharmacy and Molecular Biotechnology, Ruprecht-Karls-University, Heidelberg, Germany
| | | | - Winfried Neuhaus
- AIT - Austrian Institute of Technology GmbH, Vienna, 1210, Austria
- Department of Medicine, Faculty of Medicine and Dentistry, Danube Private University, Krems, 3500, Austria
| | - Birger Brodin
- Department of Pharmacy, University of Copenhagen, Universitetsparken 2, Copenhagen, DK-2100, Denmark.
| |
Collapse
|
27
|
Khalil A, Barras A, Boukherroub R, Tseng CL, Devos D, Burnouf T, Neuhaus W, Szunerits S. Enhancing paracellular and transcellular permeability using nanotechnological approaches for the treatment of brain and retinal diseases. NANOSCALE HORIZONS 2023; 9:14-43. [PMID: 37853828 DOI: 10.1039/d3nh00306j] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/20/2023]
Abstract
Paracellular permeability across epithelial and endothelial cells is, in large part, regulated by apical intercellular junctions also referred to as tight junctions (TJs). These junctions contribute to the spatial definition of different tissue compartments within organisms, separating them from the outside world as well as from inner compartments, with their primary physiological role of maintaining tissue homeostasis. TJs restrict the free, passive diffusion of ions and hydrophilic small molecules through paracellular clefts and are important for appropriate cell polarization and transporter protein localisation, supporting the controlled transcellular diffusion of smaller and larger hydrophilic as well as hydrophobic substances. This traditional diffusion barrier concept of TJs has been challenged lately, owing to a better understanding of the components that are associated with TJs. It is now well-established that mutations in TJ proteins are associated with a range of human diseases and that a change in the membrane fluidity of neighbouring cells can open possibilities for therapeutics to cross intercellular junctions. Nanotechnological approaches, exploiting ultrasound or hyperosmotic agents and permeation enhancers, are the paradigm for achieving enhanced paracellular diffusion. The other widely used transport route of drugs is via transcellular transport, allowing the passage of a variety of pro-drugs and nanoparticle-encapsulated drugs via different mechanisms based on receptors and others. For a long time, there was an expectation that lipidic nanocarriers and polymeric nanostructures could revolutionize the field for the delivery of RNA and protein-based therapeutics across different biological barriers equipped with TJs (e.g., blood-brain barrier (BBB), retina-blood barrier (RBB), corneal TJs, etc.). However, only a limited increase in therapeutic efficiency has been reported for most systems until now. The purpose of this review is to explore the reasons behind the current failures and to examine the emergence of synthetic and cell-derived nanomaterials and nanotechnological approaches as potential game-changers in enhancing drug delivery to target locations both at and across TJs using innovative concepts. Specifically, we will focus on recent advancements in various nanotechnological strategies enabling the bypassing or temporally opening of TJs to the brain and to the retina, and discuss their advantages and limitations.
Collapse
Affiliation(s)
- Asmaa Khalil
- Univ. Lille, CNRS, Univ. Polytechnique Hauts-de-France, UMR 8520 - IEMN, F-59000 Lille, France.
| | - Alexandre Barras
- Univ. Lille, CNRS, Univ. Polytechnique Hauts-de-France, UMR 8520 - IEMN, F-59000 Lille, France.
| | - Rabah Boukherroub
- Univ. Lille, CNRS, Univ. Polytechnique Hauts-de-France, UMR 8520 - IEMN, F-59000 Lille, France.
| | - Ching-Li Tseng
- Taipei Medical University, Graduate Institute of Biomedical Materials and Tissue Engineering (GIBMTE), New Taipei City 235603, Taiwan
- Taipei Medical University, International PhD Program in Biomedical Engineering (IPBME), New Taipei City 235603, Taiwan
| | - David Devos
- University Lille, CHU-Lille, Inserm, U1172, Lille Neuroscience & Cognition, LICEND, Lille, France
| | - Thierry Burnouf
- Taipei Medical University, Graduate Institute of Biomedical Materials and Tissue Engineering (GIBMTE), New Taipei City 235603, Taiwan
- Taipei Medical University, International PhD Program in Biomedical Engineering (IPBME), New Taipei City 235603, Taiwan
| | - Winfried Neuhaus
- AIT - Austrian Institute of Technology GmbH, Center Health and Bioresources, Competence Unit Molecular Diagnostics, 1210 Vienna, Austria
- Laboratory for Life Sciences and Technology (LiST), Faculty of Medicine and Dentistry, Danube Private University, 3500 Krems, Austria
| | - Sabine Szunerits
- Univ. Lille, CNRS, Univ. Polytechnique Hauts-de-France, UMR 8520 - IEMN, F-59000 Lille, France.
| |
Collapse
|
28
|
Burgio F, Gaiser C, Brady K, Gatta V, Class R, Schrage R, Suter-Dick L. A Perfused In Vitro Human iPSC-Derived Blood-Brain Barrier Faithfully Mimics Transferrin Receptor-Mediated Transcytosis of Therapeutic Antibodies. Cell Mol Neurobiol 2023; 43:4173-4187. [PMID: 37698826 PMCID: PMC10661771 DOI: 10.1007/s10571-023-01404-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Accepted: 08/22/2023] [Indexed: 09/13/2023]
Abstract
Delivering biologics to elicit a therapeutic response in the central nervous system (CNS) remains challenging due to the presence of the blood-brain barrier (BBB). Receptor-mediated transcytosis is a strategy to improve brain exposure after systemic drug administration. The availability of a clinically relevant in vitro BBB model is crucial to investigate transcytosis pathways and to predict the penetration of biologics into the CNS. We created a perfused human in vitro BBB model made of induced pluripotent stem cells (iPSC)-derived brain microvascular endothelial cells (BMEC) for studying transferrin receptor-mediated transcytosis. iPSC-derived BMEC were seeded in the top channel of a three-lane microfluidic device (OrganoPlate®). After 2 days in culture, the established cell model exhibited relevant BBB features, including physiological transendothelial electrical resistance in a transwell setting (1500 Ω*cm2), reduced apparent permeability (Papp) to the fluorescence tracer Lucifer yellow (20-fold less than cell-free chips), expression of key BBB markers such as tight junctions proteins, transporters, receptors and functional P-gp efflux pump. Moreover, the model exhibited functional transferrin receptor-mediated uptake and transcytosis. To assess selective transferrin receptor-mediated transcytosis, a mixture of anti-human transferrin receptor (MEM-189) and control (sheep IgG anti-bovine serum albumin) antibodies was perfused in the top channel for 2 h. The Papp of MEM-189 was 11-fold higher than that of the control antibody, demonstrating facilitated receptor-mediated transcytosis. Compared to published work reporting a 2-fold ratio, this result is remarkable and establishes the suitability of our model for exploring receptor-mediated transcytosis and screening of antibodies for putative brain shuttle application. A perfused in vitro human model made of iPSC-derived BMEC with the chief characteristics (barrier tightness, functionality) of the human BBB can be applied to study transferrin receptor (TfR)-mediated transcytosis of therapeutic antibodies. This may bring critical advances in drug shuttle technology. Graphical abstract generated with biorender.com.
Collapse
Affiliation(s)
- Floriana Burgio
- University of Applied Sciences and Arts Northwestern Switzerland (FHNW), Muttenz, Switzerland
| | - Carine Gaiser
- University of Applied Sciences and Arts Northwestern Switzerland (FHNW), Muttenz, Switzerland
| | - Kevin Brady
- Development Sciences, UCB Biopharma SRL, Braine L'Alleud, Belgium
| | - Viviana Gatta
- Neuroscience Therapeutic Area, UCB Biopharma SRL, Braine L'Alleud, Belgium
| | - Reiner Class
- Development Sciences, UCB Biopharma SRL, Braine L'Alleud, Belgium
| | - Ramona Schrage
- Neuroscience Therapeutic Area, UCB Biopharma SRL, Braine L'Alleud, Belgium
| | - Laura Suter-Dick
- University of Applied Sciences and Arts Northwestern Switzerland (FHNW), Muttenz, Switzerland.
| |
Collapse
|
29
|
Haferkamp U, Hartmann C, Abid CL, Brachner A, Höchner A, Gerhartl A, Harwardt B, Leckzik S, Leu J, Metzger M, Nastainczyk-Wulf M, Neuhaus W, Oerter S, Pless O, Rujescu D, Jung M, Appelt-Menzel A. Human isogenic cells of the neurovascular unit exert transcriptomic cell type-specific effects on a blood-brain barrier in vitro model of late-onset Alzheimer disease. Fluids Barriers CNS 2023; 20:78. [PMID: 37907966 PMCID: PMC10617216 DOI: 10.1186/s12987-023-00471-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Accepted: 10/01/2023] [Indexed: 11/02/2023] Open
Abstract
BACKGROUND The function of the blood-brain barrier (BBB) is impaired in late-onset Alzheimer disease (LOAD), but the associated molecular mechanisms, particularly with respect to the high-risk APOE4/4 genotype, are not well understood. For this purpose, we developed a multicellular isogenic model of the neurovascular unit (NVU) based on human induced pluripotent stem cells. METHODS The human NVU was modeled in vitro using isogenic co-cultures of astrocytes, brain capillary endothelial-like cells (BCECs), microglia-like cells, neural stem cells (NSCs), and pericytes. Physiological and pathophysiological properties were investigated as well as the influence of each single cell type on the characteristics and function of BCECs. The barriers established by BCECs were analyzed for specific gene transcription using high-throughput quantitative PCR. RESULTS Co-cultures were found to tighten the barrier of BCECs and alter its transcriptomic profile under both healthy and disease conditions. In vitro differentiation of brain cell types that constitute the NVU was not affected by the LOAD background. The supportive effect of NSCs on the barrier established by BCECs was diminished under LOAD conditions. Transcriptomes of LOAD BCECs were modulated by different brain cell types. NSCs were found to have the strongest effect on BCEC gene regulation and maintenance of the BBB. Co-cultures showed cell type-specific functional contributions to BBB integrity under healthy and LOAD conditions. CONCLUSIONS Cell type-dependent transcriptional effects on LOAD BCECs were identified. Our study suggests that different brain cell types of the NVU have unique roles in maintaining barrier integrity that vary under healthy and LOAD conditions. .
Collapse
Affiliation(s)
- Undine Haferkamp
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Discovery Research ScreeningPort, 22525, Hamburg, Germany
| | - Carla Hartmann
- Institute for Physiological Chemistry, Medical Faculty of the Martin, Luther University Halle-Wittenberg, Hollystrasse 1, 06114, Halle (Saale), Germany
| | - Chaudhry Luqman Abid
- Institute for Physiological Chemistry, Medical Faculty of the Martin, Luther University Halle-Wittenberg, Hollystrasse 1, 06114, Halle (Saale), Germany
| | - Andreas Brachner
- Center Health and Bioresources, Competence Unit Molecular Diagnostics, AIT Austrian Institute of Technology GmbH, Vienna, 1210, Austria
| | - Alevtina Höchner
- Fraunhofer Institute for Silicate Research ISC, Translational Center Regenerative Therapies (TLC-RT), 97070, Würzburg, Germany
| | - Anna Gerhartl
- Center Health and Bioresources, Competence Unit Molecular Diagnostics, AIT Austrian Institute of Technology GmbH, Vienna, 1210, Austria
| | - Bernadette Harwardt
- Institute for Physiological Chemistry, Medical Faculty of the Martin, Luther University Halle-Wittenberg, Hollystrasse 1, 06114, Halle (Saale), Germany
| | - Selin Leckzik
- Institute for Physiological Chemistry, Medical Faculty of the Martin, Luther University Halle-Wittenberg, Hollystrasse 1, 06114, Halle (Saale), Germany
| | - Jennifer Leu
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Discovery Research ScreeningPort, 22525, Hamburg, Germany
| | - Marco Metzger
- Fraunhofer Institute for Silicate Research ISC, Translational Center Regenerative Therapies (TLC-RT), 97070, Würzburg, Germany
- Chair Tissue Engineering and Regenerative Medicine (TERM), University Hospital Würzburg, 97070, Würzburg, Germany
| | | | - Winfried Neuhaus
- Center Health and Bioresources, Competence Unit Molecular Diagnostics, AIT Austrian Institute of Technology GmbH, Vienna, 1210, Austria
- Department of Medicine, Faculty of Medicine and Dentistry, Danube Private University, Krems, 3500, Austria
| | - Sabrina Oerter
- Fraunhofer Institute for Silicate Research ISC, Translational Center Regenerative Therapies (TLC-RT), 97070, Würzburg, Germany
- Chair Tissue Engineering and Regenerative Medicine (TERM), University Hospital Würzburg, 97070, Würzburg, Germany
| | - Ole Pless
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Discovery Research ScreeningPort, 22525, Hamburg, Germany
| | - Dan Rujescu
- Department of Psychiatry and Psychotherapy, Division of General Psychiatry, Medical University of Vienna, Vienna, 1090, Austria
| | - Matthias Jung
- Institute for Physiological Chemistry, Medical Faculty of the Martin, Luther University Halle-Wittenberg, Hollystrasse 1, 06114, Halle (Saale), Germany.
| | - Antje Appelt-Menzel
- Fraunhofer Institute for Silicate Research ISC, Translational Center Regenerative Therapies (TLC-RT), 97070, Würzburg, Germany.
- Chair Tissue Engineering and Regenerative Medicine (TERM), University Hospital Würzburg, 97070, Würzburg, Germany.
| |
Collapse
|
30
|
Graybill PM, Jacobs EJ, Jana A, Agashe A, Nain AS, Davalos RV. Ultra-thin and ultra-porous nanofiber networks as a basement-membrane mimic. LAB ON A CHIP 2023; 23:4565-4578. [PMID: 37772328 PMCID: PMC10623910 DOI: 10.1039/d3lc00304c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/30/2023]
Abstract
Current basement membrane (BM) mimics used for modeling endothelial and epithelial barriers in vitro do not faithfully recapitulate key in vivo physiological properties such as BM thickness, porosity, stiffness, and fibrous composition. Here, we use networks of precisely arranged nanofibers to form ultra-thin (∼3 μm thick) and ultra-porous (∼90%) BM mimics for blood-brain barrier modeling. We show that these nanofiber networks enable close contact between endothelial monolayers and pericytes across the membrane, which are known to regulate barrier tightness. Cytoskeletal staining and transendothelial electrical resistance (TEER) measurements reveal barrier formation on nanofiber membranes integrated within microfluidic devices and transwell inserts. Further, significantly higher TEER values indicate a biological benefit for co-cultures formed on the ultra-thin nanofiber membranes. Our BM mimic overcomes critical technological challenges in forming co-cultures that are in proximity and facilitate cell-cell contact, while still being constrained to their respective sides. We anticipate that our nanofiber networks will find applications in drug discovery, cell migration, and barrier dysfunction studies.
Collapse
Affiliation(s)
- Philip M Graybill
- Bioelectromechanical Systems Lab, Department of Biomedical Engineering and Mechanics, Virginia Tech, Blacksburg, VA, USA.
| | - Edward J Jacobs
- Bioelectromechanical Systems Lab, Department of Biomedical Engineering and Mechanics, Virginia Tech, Blacksburg, VA, USA.
| | - Aniket Jana
- Spinneret-Based Tunable Engineering Parameters (STEP) Lab, Department of Mechanical Engineering, Virginia Tech, Blacksburg, VA, USA.
| | - Atharva Agashe
- Spinneret-Based Tunable Engineering Parameters (STEP) Lab, Department of Mechanical Engineering, Virginia Tech, Blacksburg, VA, USA.
| | - Amrinder S Nain
- Spinneret-Based Tunable Engineering Parameters (STEP) Lab, Department of Mechanical Engineering, Virginia Tech, Blacksburg, VA, USA.
| | - Rafael V Davalos
- Bioelectromechanical Systems Lab, Department of Biomedical Engineering and Mechanics, Virginia Tech, Blacksburg, VA, USA.
| |
Collapse
|
31
|
Walther J, Kirsch EM, Hellwig L, Schmerbeck SS, Holloway PM, Buchan AM, Mergenthaler P. Reinventing the Penumbra - the Emerging Clockwork of a Multi-modal Mechanistic Paradigm. Transl Stroke Res 2023; 14:643-666. [PMID: 36219377 PMCID: PMC10444697 DOI: 10.1007/s12975-022-01090-9] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Revised: 09/16/2022] [Accepted: 09/21/2022] [Indexed: 11/25/2022]
Abstract
The concept of the ischemic penumbra was originally defined as the area around a necrotic stroke core and seen as the tissue at imminent risk of further damage. Today, the penumbra is generally considered as time-sensitive hypoperfused brain tissue with decreased oxygen and glucose availability, salvageable tissue as treated by intervention, and the potential target for neuroprotection in focal stroke. The original concept entailed electrical failure and potassium release but one short of neuronal cell death and was based on experimental stroke models, later confirmed in clinical imaging studies. However, even though the basic mechanisms have translated well, conferring brain protection, and improving neurological outcome after stroke based on the pathophysiological mechanisms in the penumbra has yet to be achieved. Recent findings shape the modern understanding of the penumbra revealing a plethora of molecular and cellular pathophysiological mechanisms. We now propose a new model of the penumbra, one which we hope will lay the foundation for future translational success. We focus on the availability of glucose, the brain's central source of energy, and bioenergetic failure as core pathophysiological concepts. We discuss the relation of mitochondrial function in different cell types to bioenergetics and apoptotic cell death mechanisms, autophagy, and neuroinflammation, to glucose metabolism in what is a dynamic ischemic penumbra.
Collapse
Affiliation(s)
- Jakob Walther
- Charité - Universitätsmedizin Berlin, Department of Neurology with Experimental Neurology, Charitéplatz 1, 10117, Berlin, Germany
- Charité - Universitätsmedizin Berlin, Center for Stroke Research Berlin, Charitéplatz 1, 10117, Berlin, Germany
| | - Elena Marie Kirsch
- Charité - Universitätsmedizin Berlin, Department of Neurology with Experimental Neurology, Charitéplatz 1, 10117, Berlin, Germany
- Charité - Universitätsmedizin Berlin, Center for Stroke Research Berlin, Charitéplatz 1, 10117, Berlin, Germany
| | - Lina Hellwig
- Charité - Universitätsmedizin Berlin, Department of Neurology with Experimental Neurology, Charitéplatz 1, 10117, Berlin, Germany
- Charité - Universitätsmedizin Berlin, Center for Stroke Research Berlin, Charitéplatz 1, 10117, Berlin, Germany
| | - Sarah S Schmerbeck
- Charité - Universitätsmedizin Berlin, Department of Neurology with Experimental Neurology, Charitéplatz 1, 10117, Berlin, Germany
- Charité - Universitätsmedizin Berlin, Center for Stroke Research Berlin, Charitéplatz 1, 10117, Berlin, Germany
| | - Paul M Holloway
- Acute Stroke Programme, Radcliffe Department of Medicine, University of Oxford, Oxford, OX3 9DU, UK
| | - Alastair M Buchan
- Charité - Universitätsmedizin Berlin, Center for Stroke Research Berlin, Charitéplatz 1, 10117, Berlin, Germany.
- Acute Stroke Programme, Radcliffe Department of Medicine, University of Oxford, Oxford, OX3 9DU, UK.
| | - Philipp Mergenthaler
- Charité - Universitätsmedizin Berlin, Department of Neurology with Experimental Neurology, Charitéplatz 1, 10117, Berlin, Germany.
- Charité - Universitätsmedizin Berlin, Center for Stroke Research Berlin, Charitéplatz 1, 10117, Berlin, Germany.
- Charité - Universitätsmedizin Berlin, NeuroCure Clinical Research Center, Charitéplatz 1, 10117, Berlin, Germany.
- Acute Stroke Programme, Radcliffe Department of Medicine, University of Oxford, Oxford, OX3 9DU, UK.
| |
Collapse
|
32
|
Bolden CT, Skibber MA, Olson SD, Zamorano Rojas M, Milewicz S, Gill BS, Cox CS. Validation and characterization of a novel blood-brain barrier platform for investigating traumatic brain injury. Sci Rep 2023; 13:16150. [PMID: 37752338 PMCID: PMC10522590 DOI: 10.1038/s41598-023-43214-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Accepted: 09/21/2023] [Indexed: 09/28/2023] Open
Abstract
The Blood-Brain Barrier (BBB) is a highly-selective physiologic barrier responsible for maintaining cerebral homeostasis. Innovative in vitro models of the BBB are needed to provide useful insights into BBB function with CNS disorders like traumatic brain injury (TBI). TBI is a multidimensional and highly complex pathophysiological condition that requires intrinsic models to elucidate its mechanisms. Current models either lack fluidic shear stress, or neglect hemodynamic parameters important in recapitulating the human in vivo BBB phenotype. To address these limitations in the field, we developed a fluid dynamic novel platform which closely mimics these parameters. To validate our platform, Matrigel-coated Transwells were seeded with brain microvascular endothelial cells, both with and without co-cultured primary human astrocytes and bone-marrow mesenchymal stem cells. In this article we characterized BBB functional properties such as TEER and paracellular permeability. Our platform demonstrated physiologic relevant decreases in TEER in response to an ischemic environment, while directly measuring barrier fluid fluctuation. These recordings were followed with recovery, implying stability of the model. We also demonstrate that our dynamic platform is responsive to inflammatory and metabolic cues with resultant permeability coefficients. These results indicate that this novel dynamic platform will be a valuable tool for evaluating the recapitulating BBB function in vitro, screening potential novel therapeutics, and establishing a relevant paradigm to evaluate the pathophysiology of TBI.
Collapse
Affiliation(s)
- Christopher T Bolden
- Department of Pediatric Surgery, McGovern Medical School, The University of Texas Health Science Center at Houston (UTHealth), Houston, TX, USA.
- Center for Translational Injury Research, The University of Texas Health Science Center at Houston (UTHealth), Houston, TX, USA.
| | - Max A Skibber
- Department of Pediatric Surgery, McGovern Medical School, The University of Texas Health Science Center at Houston (UTHealth), Houston, TX, USA
| | - Scott D Olson
- Department of Pediatric Surgery, McGovern Medical School, The University of Texas Health Science Center at Houston (UTHealth), Houston, TX, USA
| | - Miriam Zamorano Rojas
- Department of Pediatric Surgery, McGovern Medical School, The University of Texas Health Science Center at Houston (UTHealth), Houston, TX, USA
| | - Samantha Milewicz
- Department of Pediatric Surgery, McGovern Medical School, The University of Texas Health Science Center at Houston (UTHealth), Houston, TX, USA
| | - Brijesh S Gill
- Department of Surgery, McGovern Medical School, The University of Texas Health Science Center at Houston (UTHealth), Houston, TX, USA
| | - Charles S Cox
- Department of Pediatric Surgery, McGovern Medical School, The University of Texas Health Science Center at Houston (UTHealth), Houston, TX, USA.
- Center for Translational Injury Research, The University of Texas Health Science Center at Houston (UTHealth), Houston, TX, USA.
| |
Collapse
|
33
|
Brandl S, Reindl M. Blood-Brain Barrier Breakdown in Neuroinflammation: Current In Vitro Models. Int J Mol Sci 2023; 24:12699. [PMID: 37628879 PMCID: PMC10454051 DOI: 10.3390/ijms241612699] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Revised: 08/07/2023] [Accepted: 08/08/2023] [Indexed: 08/27/2023] Open
Abstract
The blood-brain barrier, which is formed by tightly interconnected microvascular endothelial cells, separates the brain from the peripheral circulation. Together with other central nervous system-resident cell types, including pericytes and astrocytes, the blood-brain barrier forms the neurovascular unit. Upon neuroinflammation, this barrier becomes leaky, allowing molecules and cells to enter the brain and to potentially harm the tissue of the central nervous system. Despite the significance of animal models in research, they may not always adequately reflect human pathophysiology. Therefore, human models are needed. This review will provide an overview of the blood-brain barrier in terms of both health and disease. It will describe all key elements of the in vitro models and will explore how different compositions can be utilized to effectively model a variety of neuroinflammatory conditions. Furthermore, it will explore the existing types of models that are used in basic research to study the respective pathologies thus far.
Collapse
Affiliation(s)
| | - Markus Reindl
- Clinical Department of Neurology, Medical University of Innsbruck, 6020 Innsbruck, Austria;
| |
Collapse
|
34
|
Hartl N, Gabold B, Adams F, Uhl P, Oerter S, Gätzner S, Metzger M, König AC, Hauck SM, Appelt-Menzel A, Mier W, Fricker G, Merkel OM. Overcoming the blood-brain barrier? - prediction of blood-brain permeability of hydrophobically modified polyethylenimine polyplexes for siRNA delivery into the brain with in vitro and in vivo models. J Control Release 2023; 360:613-629. [PMID: 37437848 DOI: 10.1016/j.jconrel.2023.07.019] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Revised: 06/23/2023] [Accepted: 07/08/2023] [Indexed: 07/14/2023]
Abstract
The blood-brain barrier (BBB) is a highly selective biological barrier that represents a major bottleneck in the treatment of all types of central nervous system (CNS) disorders. Small interfering RNA (siRNA) offers in principle a promising therapeutic approach, e.g., for brain tumors, by downregulating brain tumor-related genes and inhibiting tumor growth via RNA interference. In an effort to develop efficient siRNA nanocarriers for crossing the BBB, we utilized polyethyleneimine (PEI) polymers hydrophobically modified with either stearic-acid (SA) or dodecylacrylamide (DAA) subunits and evaluated their suitability for delivering siRNA across the BBB in in vitro and in vivo BBB models depending on their structure. Physicochemical characteristics of siRNA-polymer complexes (polyplexes (PXs)), e.g., particle size and surface charge, were measured by dynamic light scattering and laser Doppler anemometry, whereas siRNA condensation ability of polymers and polyplex stability was evaluated by spectrophotometric methods. The composition of the biomolecule corona that absorbs on polyplexes upon encountering physiological fluids was investigated by sodium dodecyl sulfate polyacrylamide gel electrophoresis (SDS-PAGE) and by a liquid chromatography-tandem mass spectrometry (LC-MS-MS) method. Cellular internalization abilities of PXs into brain endothelial cells (hCMEC/D3) was confirmed, and a BBB permeation assay using a human induced pluripotent stem cell (hiPSC)-derived BBB model revealed similar abilities to cross the BBB for all formulations under physiological conditions. However, biodistribution studies of radiolabeled PXs in mice were inconsistent with in vitro results as the detected amount of radiolabeled siRNA in the brain delivered with PEI PXs was higher compared to PEI-SA PXs. Taken together, PEI PXs were shown to be a suitable nanocarrier to deliver small amounts of siRNA across the BBB into the brain but more sophisticated human BBB models that better represent physiological conditions and biodistribution are required to provide highly predictive in vitro data for human CNS drug development in the future.
Collapse
Affiliation(s)
- Natascha Hartl
- Ludwig-Maximilians-University, Pharmaceutical Technology and Biopharmaceutics, Butenandtstr. 5-13, 81377, Munich, Germany
| | - Bettina Gabold
- Ludwig-Maximilians-University, Pharmaceutical Technology and Biopharmaceutics, Butenandtstr. 5-13, 81377, Munich, Germany
| | - Friederike Adams
- University of Stuttgart, Institute of Polymer Chemistry, Macromolecular Materials and Fiber Chemistry, Pfaffenwaldring 55, 70569 Stuttgart, Germany
| | - Philipp Uhl
- University Hospital Heidelberg, Department of Nuclear Medicine, Im Neuenheimer Feld 400, 69120 Heidelberg, Germany
| | - Sabrina Oerter
- Fraunhofer Institute for Silicate Research (ISC), Translational Center Regenerative Therapies (TLC-RT), 97070 Würzburg, Germany; University Hospital Würzburg, Chair of Tissue Engineering and Regenerative Medicine (TERM), 97070 Würzburg, Germany
| | - Sabine Gätzner
- Fraunhofer Institute for Silicate Research (ISC), Translational Center Regenerative Therapies (TLC-RT), 97070 Würzburg, Germany
| | - Marco Metzger
- Fraunhofer Institute for Silicate Research (ISC), Translational Center Regenerative Therapies (TLC-RT), 97070 Würzburg, Germany; University Hospital Würzburg, Chair of Tissue Engineering and Regenerative Medicine (TERM), 97070 Würzburg, Germany
| | - Ann-Christine König
- Helmholtz Centrum Munich - German Research Center for Environmental Health, Research Unit Protein Science, Heidemannsstr. 1, 80939, Munich, Germany
| | - Stefanie M Hauck
- Helmholtz Centrum Munich - German Research Center for Environmental Health, Research Unit Protein Science, Heidemannsstr. 1, 80939, Munich, Germany
| | - Antje Appelt-Menzel
- Fraunhofer Institute for Silicate Research (ISC), Translational Center Regenerative Therapies (TLC-RT), 97070 Würzburg, Germany; University Hospital Würzburg, Chair of Tissue Engineering and Regenerative Medicine (TERM), 97070 Würzburg, Germany
| | - Walter Mier
- University Hospital Heidelberg, Department of Nuclear Medicine, Im Neuenheimer Feld 400, 69120 Heidelberg, Germany
| | - Gert Fricker
- University of Heidelberg, Institute for Pharmacy & Molekular Biotechnology, Im Neuenheimer Feld 329, 69120 Heidelberg, Germany
| | - Olivia M Merkel
- Ludwig-Maximilians-University, Pharmaceutical Technology and Biopharmaceutics, Butenandtstr. 5-13, 81377, Munich, Germany.
| |
Collapse
|
35
|
Singh S, Agrawal M, Vashist R, Patel RK, Sangave SD, Alexander A. Recent advancements on in vitro blood-brain barrier model: A reliable and efficient screening approach for preclinical and clinical investigation. Expert Opin Drug Deliv 2023; 20:1839-1857. [PMID: 38100459 DOI: 10.1080/17425247.2023.2295940] [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: 09/06/2023] [Accepted: 12/13/2023] [Indexed: 12/17/2023]
Abstract
INTRODUCTION The efficiency of brain therapeutics is greatly hindered by the blood-brain barrier (BBB). BBB's protective function, selective permeability, and dynamic functionality maintain the harmony between the brain and peripheral region. Thus, the design of any novel drug carrier system requires the complete study and investigation of BBB permeability, efflux transport, and the effect of associated cellular and non-vascular unit trafficking on BBB penetrability. The in vitro BBB models offer a most promising, and reliable mode of initial investigation of BBB permeability and associated factors as strong evidence for further preclinical and clinical investigation. AREA COVERED This review work covers the structure and functions of BBB components and different types of in vitro BBB models along with factors affecting BBB model development and model selection criteria. EXPERT OPINION In vivo models assume to reciprocate the physiological environment to the maximum extent. However, the interspecies variability, NVUs trafficking, dynamic behavior of BBB, etc., lead to non-reproducible results. The in vitro models are comparatively less complex, and flexible, as per the study design, could generate substantial evidence and help identify suitable in vivo animal model selection.
Collapse
Affiliation(s)
- Snigdha Singh
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research, Guwahati, India
| | - Mukta Agrawal
- School of Pharmacy and Technology Management, Narsee Monjee Institute of Management Studies, Mahbubnagar, India
| | - Rajat Vashist
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research, Guwahati, India
| | - Rohit K Patel
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research, Guwahati, India
| | | | - Amit Alexander
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research, Guwahati, India
| |
Collapse
|
36
|
Alves ADH, Nucci MP, Ennes do Valle NM, Missina JM, Mamani JB, Rego GNA, Dias OFM, Garrigós MM, de Oliveira FA, Gamarra LF. Current overview of induced pluripotent stem cell-based blood-brain barrier-on-a-chip. World J Stem Cells 2023; 15:632-653. [PMID: 37424947 PMCID: PMC10324508 DOI: 10.4252/wjsc.v15.i6.632] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Revised: 04/10/2023] [Accepted: 05/08/2023] [Indexed: 06/26/2023] Open
Abstract
BACKGROUND Induced pluripotent stem cells (iPSCs) show great ability to differentiate into any tissue, making them attractive candidates for pathophysiological investigations. The rise of organ-on-a-chip technology in the past century has introduced a novel way to make in vitro cell cultures that more closely resemble their in vivo environments, both structural and functionally. The literature still lacks consensus on the best conditions to mimic the blood-brain barrier (BBB) for drug screening and other personalized therapies. The development of models based on BBB-on-a-chip using iPSCs is promising and is a potential alternative to the use of animals in research.
AIM To analyze the literature for BBB models on-a-chip involving iPSCs, describe the microdevices, the BBB in vitro construction, and applications.
METHODS We searched for original articles indexed in PubMed and Scopus that used iPSCs to mimic the BBB and its microenvironment in microfluidic devices. Thirty articles were identified, wherein only 14 articles were finally selected according to the inclusion and exclusion criteria. Data compiled from the selected articles were organized into four topics: (1) Microfluidic devices design and fabrication; (2) characteristics of the iPSCs used in the BBB model and their differentiation conditions; (3) BBB-on-a-chip reconstruction process; and (4) applications of BBB microfluidic three-dimensional models using iPSCs.
RESULTS This study showed that BBB models with iPSCs in microdevices are quite novel in scientific research. Important technological advances in this area regarding the use of commercial BBB-on-a-chip were identified in the most recent articles by different research groups. Conventional polydimethylsiloxane was the most used material to fabricate in-house chips (57%), whereas few studies (14.3%) adopted polymethylmethacrylate. Half the models were constructed using a porous membrane made of diverse materials to separate the channels. iPSC sources were divergent among the studies, but the main line used was IMR90-C4 from human fetal lung fibroblast (41.2%). The cells were differentiated through diverse and complex processes either to endothelial or neural cells, wherein only one study promoted differentiation inside the chip. The construction process of the BBB-on-a-chip involved previous coating mostly with fibronectin/collagen IV (39.3%), followed by cell seeding in single cultures (36%) or co-cultures (64%) under controlled conditions, aimed at developing an in vitro BBB that mimics the human BBB for future applications.
CONCLUSION This review evidenced technological advances in the construction of BBB models using iPSCs. Nonetheless, a definitive BBB-on-a-chip has not yet been achieved, hindering the applicability of the models.
Collapse
Affiliation(s)
| | - Mariana Penteado Nucci
- Hospital Israelita Albert Einstein, São Paulo 05529-060, São Paulo, Brazil
- Hospital das Clínicas HCFMUSP, Faculdade de Medicina, Universidade de São Paulo, São Paulo 05403-010, São Paulo, Brazil
| | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Schalla MA, Oerter S, Cubukova A, Metzger M, Appelt-Menzel A, Stengel A. Locked Out: Phoenixin-14 Does Not Cross a Stem-Cell-Derived Blood-Brain Barrier Model. Brain Sci 2023; 13:980. [PMID: 37508911 PMCID: PMC10377091 DOI: 10.3390/brainsci13070980] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Revised: 06/19/2023] [Accepted: 06/20/2023] [Indexed: 07/30/2023] Open
Abstract
Phoenixin-14 is a recently discovered peptide regulating appetite. Interestingly, it is expressed in the gastrointestinal tract; however, its supposed receptor, GPR173, is predominantly found in hypothalamic areas. To date, it is unknown how peripherally secreted phoenixin-14 is able to reach its centrally located receptor. To investigate whether phoenixin is able to pass the blood-brain barrier, we used an in vitro mono-culture blood-brain barrier (BBB) model consisting of brain capillary-like endothelial cells derived from human induced-pluripotent stem cells (hiPSC-BCECs). The passage of 1 nMol and 10 nMol of phoenixin-14 via the mono-culture was measured after 30, 60, 90, 120, 150, 180, 210, and 240 min using a commercial ELISA kit. The permeability coefficients (PC) of 1 nMol and 10 nMol phoenixin-14 were 0.021 ± 0.003 and 0.044 ± 0.013 µm/min, respectively. In comparison with the PC of solutes known to cross the BBB in vivo, those of phoenixin-14 in both concentrations are very low. Here, we show that phoenixin-14 alone is not able to cross the BBB, suggesting that the effects of peripherally secreted phoenixin-14 depend on a co-transport mechanism at the BBB in vivo. The mechanisms responsible for phoenixin-14's orexigenic property along the gut-brain axis warrant further research.
Collapse
Affiliation(s)
- Martha A Schalla
- Charité Center for Internal Medicine and Dermatology, Department for Psychosomatic Medicine; Charite-Universitätsmedizin BerlinCorporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health, 12203 Berlin, Germany
- Department of Gynecology and Obstetrics, HELIOS Kliniken GmbH, 78628 Rottweil, Germany
- Department of Psychosomatic Medicine and Psychotherapy, University Hospital Tübingen, Osianderstr. 5, 72076 Tübingen, Germany
| | - Sabrina Oerter
- Fraunhofer Institute for Silicate Research ISC, Translational Center Regenerative Therapies (TLC-RT), 97070 Würzburg, Germany
- Chair Tissue Engineering and Regenerative Medicine (TERM), University Hospital Würzburg, 97070 Würzburg, Germany
| | - Alevtina Cubukova
- Chair Tissue Engineering and Regenerative Medicine (TERM), University Hospital Würzburg, 97070 Würzburg, Germany
| | - Marco Metzger
- Fraunhofer Institute for Silicate Research ISC, Translational Center Regenerative Therapies (TLC-RT), 97070 Würzburg, Germany
- Chair Tissue Engineering and Regenerative Medicine (TERM), University Hospital Würzburg, 97070 Würzburg, Germany
| | - Antje Appelt-Menzel
- Fraunhofer Institute for Silicate Research ISC, Translational Center Regenerative Therapies (TLC-RT), 97070 Würzburg, Germany
- Chair Tissue Engineering and Regenerative Medicine (TERM), University Hospital Würzburg, 97070 Würzburg, Germany
| | - Andreas Stengel
- Charité Center for Internal Medicine and Dermatology, Department for Psychosomatic Medicine; Charite-Universitätsmedizin BerlinCorporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health, 12203 Berlin, Germany
- Department of Psychosomatic Medicine and Psychotherapy, University Hospital Tübingen, Osianderstr. 5, 72076 Tübingen, Germany
| |
Collapse
|
38
|
Malik JR, Fletcher CV, Podany AT, Dyavar SR, Scarsi KK, Pais GM, Scheetz MH, Avedissian SN. A novel 4-cell in-vitro blood-brain barrier model and its characterization by confocal microscopy and TEER measurement. J Neurosci Methods 2023; 392:109867. [PMID: 37116621 PMCID: PMC10275325 DOI: 10.1016/j.jneumeth.2023.109867] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 04/18/2023] [Accepted: 04/25/2023] [Indexed: 04/30/2023]
Abstract
The blood-brain barrier (BBB) is a protective cellular anatomical layer with a dynamic micro-environment, tightly regulating the transport of materials across it. To achieve in-vivo characteristics, an in-vitro BBB model requires the constituent cell types to be layered in an appropriate order. A cost-effective in-vitro BBB model is desired to facilitate central nervous system (CNS) drug penetration studies. Enhanced integrity of tight junctions observed during the in-vitro BBB establishment and post-experiment is essential in these models. We successfully developed an in-vitro BBB model mimicking the in-vivo cell composition and a distinct order of seeding primary human brain cells. Unlike other in-vitro BBB models, our work avoids the need for pre-coated plates for cell adhesion and provides better cell visualization during the procedure. We found that using bovine collagen-I coating, followed by bovine fibronectin coating and poly-L-lysine coating, yields better adhesion and layering of cells on the transwell membrane compared to earlier reported use of collagen and poly-L-lysine only. Our results indicated better cell visibility and imaging with the polyester transwell membrane as well as point to a higher and more stable Trans Endothelial Electrical Resistance values in this plate. In addition, we found that the addition of zinc induced higher claudin 5 expressions in neuronal cells. Dolutegravir, a drug used in the treatment of HIV, is known to appear in moderate concentrations in the CNS. Thus, dolutegravir was used to assess the functionality of the final model and cells. Using primary cells and an in-house coating strategy substantially reduces costs and provides superior imaging of cells and their tight junction protein expression. Our 4-cell-based BBB model is a suitable experimental model for the drug screening process.
Collapse
Affiliation(s)
- Johid R Malik
- Antiviral Pharmacology Laboratory, College of Pharmacy, University of Nebraska Medical Center, Omaha, NE, USA
| | - Courtney V Fletcher
- Antiviral Pharmacology Laboratory, College of Pharmacy, University of Nebraska Medical Center, Omaha, NE, USA; Division of Infectious Diseases, Department of Medicine, University of Nebraska Medical Center, Omaha, NE, USA
| | - Anthony T Podany
- Antiviral Pharmacology Laboratory, College of Pharmacy, University of Nebraska Medical Center, Omaha, NE, USA
| | | | - Kimberly K Scarsi
- Antiviral Pharmacology Laboratory, College of Pharmacy, University of Nebraska Medical Center, Omaha, NE, USA; Division of Infectious Diseases, Department of Medicine, University of Nebraska Medical Center, Omaha, NE, USA
| | - Gwendolyn M Pais
- Department of Pharmacy Practice, Chicago College of Pharmacy, Midwestern University, Downers Grove, IL, USA; Midwestern University, College of Pharmacy Center of Pharmacometric Excellence, Downers Grove, IL, USA
| | - Marc H Scheetz
- Department of Pharmacy Practice, Chicago College of Pharmacy, Midwestern University, Downers Grove, IL, USA; Midwestern University, College of Pharmacy Center of Pharmacometric Excellence, Downers Grove, IL, USA
| | - Sean N Avedissian
- Antiviral Pharmacology Laboratory, College of Pharmacy, University of Nebraska Medical Center, Omaha, NE, USA.
| |
Collapse
|
39
|
Kaya S, Callan B, Hawthorne S. Non-Invasive, Targeted Nanoparticle-Mediated Drug Delivery across a Novel Human BBB Model. Pharmaceutics 2023; 15:pharmaceutics15051382. [PMID: 37242623 DOI: 10.3390/pharmaceutics15051382] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Revised: 04/20/2023] [Accepted: 04/28/2023] [Indexed: 05/28/2023] Open
Abstract
The blood-brain barrier (BBB) is a highly sophisticated system with the ability to regulate compounds transporting through the barrier and reaching the central nervous system (CNS). The BBB protects the CNS from toxins and pathogens but can cause major issues when developing novel therapeutics to treat neurological disorders. PLGA nanoparticles have been developed to successfully encapsulate large hydrophilic compounds for drug delivery. Within this paper, we discuss the encapsulation of a model compound Fitc-dextran, a large molecular weight (70 kDa), hydrophilic compound, with over 60% encapsulation efficiency (EE) within a PLGA nanoparticle (NP). The NP surface was chemically modified with DAS peptide, a ligand that we designed which has an affinity for nicotinic receptors, specifically alpha 7 nicotinic receptors, found on the surface of brain endothelial cells. The attachment of DAS transports the NP across the BBB by receptor-mediated transcytosis (RMT). Assessment of the delivery efficacy of the DAS-conjugated Fitc-dextran-loaded PLGA NP was studied in vitro using our optimal triculture in vitro BBB model, which successfully replicates the in vivo BBB environment, producing high TEER (≥230 ) and high expression of ZO1 protein. Utilising our optimal BBB model, we successfully transported fourteen times the concentration of DAS-Fitc-dextran-PLGA NP compared to non-conjugated Fitc-dextran-PLGA NP. Our novel in vitro model is a viable method of high-throughput screening of potential therapeutic delivery systems to the CNS, such as our receptor-targeted DAS ligand-conjugated NP, whereby only lead therapeutic compounds will progress to in vivo studies.
Collapse
Affiliation(s)
- Shona Kaya
- School of Pharmacy and Pharmaceutical Sciences, Ulster University, Coleraine, N. Ireland BT52 1SA, UK
| | - Bridgeen Callan
- School of Pharmacy and Pharmaceutical Sciences, Ulster University, Coleraine, N. Ireland BT52 1SA, UK
| | - Susan Hawthorne
- School of Pharmacy and Pharmaceutical Sciences, Ulster University, Coleraine, N. Ireland BT52 1SA, UK
| |
Collapse
|
40
|
Rodgers TM, Muzzio N, Valero A, Ahmad I, Lüdtke TU, Moya SE, Romero G. Poly (β-amino Ester) Nanoparticles Modified with a Rabies Virus-derived peptide for the Delivery of ASCL1 Across a 3D In Vitro Model of the Blood Brain Barrier. ACS APPLIED NANO MATERIALS 2023; 6:6299-6311. [PMID: 37274933 PMCID: PMC10234607 DOI: 10.1021/acsanm.3c00651] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
Gene editing has emerged as a therapeutic approach to manipulate the genome for killing cancer cells, protecting healthy tissues, and improving immune response to a tumor. The gene editing tool achaete-scute family bHLH transcription factor 1 CRISPR guide RNA (ASCL1-gRNA) is known to restore neuronal lineage potential, promote terminal differentiation, and attenuate tumorigenicity in glioblastoma tumors. Here, we fabricated a polymeric nonviral carrier to encapsulate ASCL1-gRNA by electrostatic interactions and deliver it into glioblastoma cells across a 3D in vitro model of the blood-brain barrier (BBB). To mimic rabies virus (RV) neurotropism, gene-loaded poly (β-amino ester) nanoparticles are surface functionalized with a peptide derivative of rabies virus glycoprotein (RVG29). The capability of the obtained NPs, hereinafter referred to as RV-like NPs, to travel across the BBB, internalize into glioblastoma cells and deliver ASCL1-gRNA are investigated in a 3D BBB in vitro model through flow cytometry and CLSM microscopy. The formation of nicotinic acetylcholine receptors in the 3D BBB in vitro model is confirmed by immunochemistry. These receptors are known to bind to RVG29. Unlike Lipofectamine that primarily internalizes and transfects endothelial cells, RV-like NPs are capable to travel across the BBB, preferentially internalize glioblastoma cells and deliver ASCL1-gRNA at an efficiency of 10 % causing non-cytotoxic effects.
Collapse
Affiliation(s)
- Tina M Rodgers
- Department of Biomedical Engineering and Chemical Engineering, The University of Texas at San Antonio, San Antonio, Texas, 78249, USA
| | - Nicolas Muzzio
- Department of Biomedical Engineering and Chemical Engineering, The University of Texas at San Antonio, San Antonio, Texas, 78249, USA
| | - Andrea Valero
- Department of Biomedical Engineering and Chemical Engineering, The University of Texas at San Antonio, San Antonio, Texas, 78249, USA
| | - Ikram Ahmad
- Department of Biomedical Engineering and Chemical Engineering, The University of Texas at San Antonio, San Antonio, Texas, 78249, USA
| | - Tanja Ursula Lüdtke
- Soft Matter Nanotechnology, CIC biomaGUNE, Basque Research and Technology Alliance (BRTA), Paseo Miramon 182, Donostia/San Sebastian, Gipuzkoa, 20014 Spain
| | - Sergio E Moya
- Soft Matter Nanotechnology, CIC biomaGUNE, Basque Research and Technology Alliance (BRTA), Paseo Miramon 182, Donostia/San Sebastian, Gipuzkoa, 20014 Spain
| | - Gabriela Romero
- Department of Biomedical Engineering and Chemical Engineering, The University of Texas at San Antonio, San Antonio, Texas, 78249, USA
| |
Collapse
|
41
|
Hudecz D, McCloskey MC, Vergo S, Christensen S, McGrath JL, Nielsen MS. Modelling a Human Blood-Brain Barrier Co-Culture Using an Ultrathin Silicon Nitride Membrane-Based Microfluidic Device. Int J Mol Sci 2023; 24:5624. [PMID: 36982697 PMCID: PMC10058651 DOI: 10.3390/ijms24065624] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Revised: 03/07/2023] [Accepted: 03/09/2023] [Indexed: 03/18/2023] Open
Abstract
Understanding the vesicular trafficking of receptors and receptor ligands in the brain capillary endothelium is essential for the development of the next generations of biologics targeting neurodegenerative diseases. Such complex biological questions are often approached by in vitro models in combination with various techniques. Here, we present the development of a stem cell-based human in vitro blood-brain barrier model composed of induced brain microvascular endothelial cells (iBMECs) on the modular µSiM (a microdevice featuring a silicon nitride membrane) platform. The µSiM was equipped with a 100 nm thick nanoporous silicon nitride membrane with glass-like imaging quality that allowed the use of high-resolution in situ imaging to study the intracellular trafficking. As a proof-of-concept experiment, we investigated the trafficking of two monoclonal antibodies (mAb): an anti-human transferrin receptor mAb (15G11) and an anti-basigin mAb (#52) using the µSiM-iBMEC-human astrocyte model. Our results demonstrated effective endothelial uptake of the selected antibodies; however, no significant transcytosis was observed when the barrier was tight. In contrast, when the iBMECs did not form a confluent barrier on the µSiM, the antibodies accumulated inside both the iBMECs and astrocytes, demonstrating that the cells have an active endocytic and subcellular sorting machinery and that the µSiM itself does not hinder antibody transport. In conclusion, our µSiM-iBMEC-human astrocyte model provides a tight barrier with endothelial-like cells, which can be used for high-resolution in situ imaging and for studying receptor-mediated transport and transcytosis in a physiological barrier.
Collapse
Affiliation(s)
- Diana Hudecz
- Department of Biomedicine, Aarhus University, 8000 Aarhus, Denmark
| | - Molly C. McCloskey
- Department of Biomedical Engineering, University of Rochester, Rochester, NY 14627, USA
| | - Sandra Vergo
- Biotherapeutic Discovery, H. Lundbeck A/S, Valby, 2500 Copenhagen, Denmark
| | - Søren Christensen
- Biotherapeutic Discovery, H. Lundbeck A/S, Valby, 2500 Copenhagen, Denmark
| | - James L. McGrath
- Department of Biomedical Engineering, University of Rochester, Rochester, NY 14627, USA
| | | |
Collapse
|
42
|
Bayir E. Development of a three-dimensional in vitro blood-brain barrier using the chitosan-alginate polyelectrolyte complex as the extracellular matrix. J BIOACT COMPAT POL 2023. [DOI: 10.1177/08839115231157096] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/18/2023]
Abstract
Polyelectrolyte complexes (PECs) consist of a spontaneous assembly of oppositely charged polysaccharides. PECs can be used to obtain a hydrogel tissue scaffold in tissue culture. In this study, it is aimed to use PEC as a blood-brain barrier (BBB) model scaffold. By mixing polycationic chitosan and polyanionic alginate solutions at a certain ratio it was obtained a 3D hydrogel scaffold and mimicked in vivo environment of the tissue. The PEC hydrogel scaffold’s chemical, physical, and mechanical characterizations were performed with FTIR, DSC, DMA, and Micro-CT analyses. In order to develop an in vitro BBB model, the human neuroblastoma cell line (SH-SY5Y) and mouse astrocyte cell line (C8-D1A) were mixed into a hydrogel, which is the abluminal side of the BBB. Human microvascular endothelial cells (HBEC-5i) were seeded on the hydrogel, and it was aimed to mimic the luminal side of the BBB. The characterization of the BBB model was determined by measuring the TEER, observation of the cell morphology with SEM, performing the permeability of Lucifer Yellow, and observation of tight junction proteins with immunofluorescence staining. As a result, HBEC-5i cells expressed tight junction proteins (ZO-1 and Claudin-5), showed TEER of 340 ± 22 Ω.cm2, and the Lucifer Yellow permeability of 7.4 × 10−7 ± 2.7 × 10−7 cm/s, which was suitable for use as an in vitro BBB model. Using a hydrogel PEC composed of chitosan and alginate as an extracellular matrix increased the direct interaction of endothelial cells, astrocytes, and neurons with each other and thus obtained a much less permeable model compared to other standard transwell models. Graphical abstract [Formula: see text]
Collapse
Affiliation(s)
- Ece Bayir
- Ege University Central Research Test and Analysis Laboratory Application and Research Center (EGE-MATAL), Ege University, Izmir, Turkey
| |
Collapse
|
43
|
Nakayama-Kitamura K, Shigemoto-Mogami Y, Toyoda H, Mihara I, Moriguchi H, Naraoka H, Furihata T, Ishida S, Sato K. Usefulness of a humanized tricellular static transwell blood-brain barrier model as a microphysiological system for drug development applications. - A case study based on the benchmark evaluations of blood-brain barrier microphysiological system. Regen Ther 2023; 22:192-202. [PMID: 36891355 PMCID: PMC9988422 DOI: 10.1016/j.reth.2023.02.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Revised: 01/21/2023] [Accepted: 02/08/2023] [Indexed: 03/06/2023] Open
Abstract
Microphysiological system (MPS), a new technology for in vitro testing platforms, have been acknowledged as a strong tool for drug development. In the central nervous system (CNS), the blood‒brain barrier (BBB) limits the permeation of circulating substances from the blood vessels to the brain, thereby protecting the CNS from circulating xenobiotic compounds. At the same time, the BBB hinders drug development by introducing challenges at various stages, such as pharmacokinetics/pharmacodynamics (PK/PD), safety assessment, and efficacy assessment. To solve these problems, efforts are being made to develop a BBB MPS, particularly of a humanized type. In this study, we suggested minimal essential benchmark items to establish the BBB-likeness of a BBB MPS; these criteria support end users in determining the appropriate range of applications for a candidate BBB MPS. Furthermore, we examined these benchmark items in a two-dimensional (2D) humanized tricellular static transwell BBB MPS, the most conventional design of BBB MPS with human cell lines. Among the benchmark items, the efflux ratios of P-gp and BCRP showed high reproducibility in two independent facilities, while the directional transports meditated through Glut1 or TfR were not confirmed. We have organized the protocols of the experiments described above as standard operating procedures (SOPs). We here provide the SOPs with the flow chart including entire procedure and how to apply each SOP. Our study is important developmental step of BBB MPS towards the social acceptance, which enable end users to check and compare the performance the BBB MPSs.
Collapse
Key Words
- BBB, blood-brain barrier
- BCRP
- BCRP, Breast cancer resistance protein
- Blood‒brain barrier (BBB)
- CNS, central nervous system
- Glut1, Glucose transporter 1
- HASTR, Human astrocytes
- HBMEC, Human brain microvascular endothelial cells
- HBPC, Human brain pericyte
- LC-MS/MS, Liquid chromatography with tandem mass spectrometry
- LY, Lucifer yellow
- MPS, Microphysiological system
- Microphysiological system (MPS)
- P-gp
- P-gp, P-glycoprotein
- TEER, Trans-endothelial electrical resistance
- TfR, Transferrin receptor
Collapse
Affiliation(s)
- Kimiko Nakayama-Kitamura
- Laboratory of Neuropharmacology, Division of Pharmacology, National Institute of Health Science, 3-25-26 Tonomachi, Kawasaki-ku, Kawasaki City, Kanagawa, Japan
| | - Yukari Shigemoto-Mogami
- Laboratory of Neuropharmacology, Division of Pharmacology, National Institute of Health Science, 3-25-26 Tonomachi, Kawasaki-ku, Kawasaki City, Kanagawa, Japan
| | - Hiroko Toyoda
- Stem Cell Evaluation Technology Research Association, Grande Building 8F, 2-26-9 Hatchobori, Chuo-ku, Tokyo 104-0032, Japan
| | - Ikue Mihara
- Stem Cell Evaluation Technology Research Association, Grande Building 8F, 2-26-9 Hatchobori, Chuo-ku, Tokyo 104-0032, Japan
| | - Hiroyuki Moriguchi
- Stem Cell Evaluation Technology Research Association, Grande Building 8F, 2-26-9 Hatchobori, Chuo-ku, Tokyo 104-0032, Japan
| | - Hitoshi Naraoka
- Stem Cell Evaluation Technology Research Association, Grande Building 8F, 2-26-9 Hatchobori, Chuo-ku, Tokyo 104-0032, Japan
| | - Tomomi Furihata
- School of Pharmacy, Tokyo University of Pharmacy and Life Sciences, 1432-1 Horinouchi, Hachioji, Tokyo 192-0392 Japan
| | - Seiichi Ishida
- Laboratory of Neuropharmacology, Division of Pharmacology, National Institute of Health Science, 3-25-26 Tonomachi, Kawasaki-ku, Kawasaki City, Kanagawa, Japan.,Division of Applied Life Science, Graduate School of Engineering, Sojo University, 4-22-1 Ikeda, Nishi-ku, Kumamoto City, Kumamoto, Japan
| | - Kaoru Sato
- Laboratory of Neuropharmacology, Division of Pharmacology, National Institute of Health Science, 3-25-26 Tonomachi, Kawasaki-ku, Kawasaki City, Kanagawa, Japan
| |
Collapse
|
44
|
Khamis ZI, Sarker DB, Xue Y, Al-Akkary N, James VD, Zeng C, Li Y, Sang QXA. Modeling Human Brain Tumors and the Microenvironment Using Induced Pluripotent Stem Cells. Cancers (Basel) 2023; 15:cancers15041253. [PMID: 36831595 PMCID: PMC9954701 DOI: 10.3390/cancers15041253] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Revised: 02/14/2023] [Accepted: 02/14/2023] [Indexed: 02/18/2023] Open
Abstract
Brain cancer is a group of diverse and rapidly growing malignancies that originate in the central nervous system (CNS) and have a poor prognosis. The complexity of brain structure and function makes brain cancer modeling extremely difficult, limiting pathological studies and therapeutic developments. Advancements in human pluripotent stem cell technology have opened a window of opportunity for brain cancer modeling, providing a wealth of customizable methods to simulate the disease in vitro. This is achieved with the advent of genome editing and genetic engineering technologies that can simulate germline and somatic mutations found in human brain tumors. This review investigates induced pluripotent stem cell (iPSC)-based approaches to model human brain cancer. The applications of iPSCs as renewable sources of individual brain cell types, brain organoids, blood-brain barrier (BBB), and brain tumor models are discussed. The brain tumor models reviewed are glioblastoma and medulloblastoma. The iPSC-derived isogenic cells and three-dimensional (3D) brain cancer organoids combined with patient-derived xenografts will enhance future compound screening and drug development for these deadly human brain cancers.
Collapse
Affiliation(s)
- Zahraa I. Khamis
- Department of Chemistry and Biochemistry, Florida State University, Tallahassee, FL 32306, USA
- Department of Industrial and Manufacturing Engineering, FAMU-FSU College of Engineering, Florida State University, Tallahassee, FL 32310, USA
- High-Performance Materials Institute, Florida State University, Tallahassee, FL 32310, USA
- Laboratory of Cancer Biology and Molecular Immunology, Department of Biochemistry, Faculty of Sciences-I, Lebanese University, Beirut, Lebanon
| | - Drishty B. Sarker
- Department of Chemistry and Biochemistry, Florida State University, Tallahassee, FL 32306, USA
| | - Yu Xue
- Department of Chemistry and Biochemistry, Florida State University, Tallahassee, FL 32306, USA
| | - Nancy Al-Akkary
- Laboratory of Cancer Biology and Molecular Immunology, Department of Biochemistry, Faculty of Sciences-I, Lebanese University, Beirut, Lebanon
| | - Viviana D. James
- Department of Chemistry and Biochemistry, Florida State University, Tallahassee, FL 32306, USA
| | - Changchun Zeng
- Department of Industrial and Manufacturing Engineering, FAMU-FSU College of Engineering, Florida State University, Tallahassee, FL 32310, USA
- High-Performance Materials Institute, Florida State University, Tallahassee, FL 32310, USA
| | - Yan Li
- Department of Chemical and Biomedical Engineering, FAMU-FSU College of Engineering, Florida State University, Tallahassee, FL 32306, USA
- Institute of Molecular Biophysics, Florida State University, Tallahassee, FL 32306, USA
| | - Qing-Xiang Amy Sang
- Department of Chemistry and Biochemistry, Florida State University, Tallahassee, FL 32306, USA
- Institute of Molecular Biophysics, Florida State University, Tallahassee, FL 32306, USA
- Correspondence: ; Tel.: +1-850-644-8683; Fax: +1-850-644-8281
| |
Collapse
|
45
|
Li GS, Chen GH, Wang KH, Wang XX, Hu XS, Wei B, Hu Y. Neurovascular Unit Compensation from Adjacent Level May Contribute to Spontaneous Functional Recovery in Experimental Cervical Spondylotic Myelopathy. Int J Mol Sci 2023; 24:ijms24043408. [PMID: 36834841 PMCID: PMC9962900 DOI: 10.3390/ijms24043408] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2022] [Revised: 12/23/2022] [Accepted: 02/06/2023] [Indexed: 02/11/2023] Open
Abstract
The progression and remission of cervical spondylotic myelopathy (CSM) are quite unpredictable due to the ambiguous pathomechanisms. Spontaneous functional recovery (SFR) has been commonly implicated in the natural course of incomplete acute spinal cord injury (SCI), while the evidence and underlying pathomechanisms of neurovascular unit (NVU) compensation involved in SFR remains poorly understood in CSM. In this study, we investigate whether compensatory change of NVU, in particular in the adjacent level of the compressive epicenter, is involved in the natural course of SFR, using an established experimental CSM model. Chronic compression was created by an expandable water-absorbing polyurethane polymer at C5 level. Neurological function was dynamically assessed by BBB scoring and somatosensory evoked potential (SEP) up to 2 months. (Ultra)pathological features of NVUs were presented by histopathological and TEM examination. Quantitative analysis of regional vascular profile area/number (RVPA/RVPN) and neuroglial cells numbers were based on the specific EBA immunoreactivity and neuroglial biomarkers, respectively. Functional integrity of blood spinal cord barrier (BSCB) was detected by Evan blue extravasation test. Although destruction of the NVU, including disruption of the BSCB, neuronal degeneration and axon demyelination, as well as dramatic neuroglia reaction, were found in the compressive epicenter and spontaneous locomotor and sensory function recovery were verified in the modeling rats. In particular, restoration of BSCB permeability and an evident increase in RVPA with wrapping proliferated astrocytic endfeet in gray matter and neuron survival and synaptic plasticity were confirmed in the adjacent level. TEM findings also proved ultrastructural restoration of the NVU. Thus, NVU compensation changes in the adjacent level may be one of the essential pathomechanisms of SFR in CSM, which could be a promising endogenous target for neurorestoration.
Collapse
Affiliation(s)
- Guang-Sheng Li
- Spinal Division of Orthopedic and Traumatology Center, The Affiliated Hospital of Guangdong Medical University, Zhanjiang 524002, China
- Department of Orthopaedics and Traumatology, The University of Hong Kong, Hong Kong, China
| | - Guang-Hua Chen
- Spinal Division of Orthopedic and Traumatology Center, The Affiliated Hospital of Guangdong Medical University, Zhanjiang 524002, China
- Correspondence: (G.-H.C.); (Y.H.)
| | - Kang-Heng Wang
- Spinal Division of Orthopedic and Traumatology Center, The Affiliated Hospital of Guangdong Medical University, Zhanjiang 524002, China
| | - Xu-Xiang Wang
- Spinal Division of Orthopedic and Traumatology Center, The Affiliated Hospital of Guangdong Medical University, Zhanjiang 524002, China
| | - Xiao-Song Hu
- Spinal Division of Orthopedic and Traumatology Center, The Affiliated Hospital of Guangdong Medical University, Zhanjiang 524002, China
| | - Bo Wei
- Spinal Division of Orthopedic and Traumatology Center, The Affiliated Hospital of Guangdong Medical University, Zhanjiang 524002, China
| | - Yong Hu
- Spinal Division of Orthopedic and Traumatology Center, The Affiliated Hospital of Guangdong Medical University, Zhanjiang 524002, China
- Department of Orthopaedics and Traumatology, The University of Hong Kong, Hong Kong, China
- Correspondence: (G.-H.C.); (Y.H.)
| |
Collapse
|
46
|
Girard SD, Julien-Gau I, Molino Y, Combes BF, Greetham L, Khrestchatisky M, Nivet E. High and low permeability of human pluripotent stem cell-derived blood-brain barrier models depend on epithelial or endothelial features. FASEB J 2023; 37:e22770. [PMID: 36688807 DOI: 10.1096/fj.202201422r] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 12/21/2022] [Accepted: 12/28/2022] [Indexed: 01/24/2023]
Abstract
The search for reliable human blood-brain barrier (BBB) models represents a challenge for the development/testing of strategies aiming to enhance brain delivery of drugs. Human-induced pluripotent stem cells (hiPSCs) have raised hopes in the development of predictive BBB models. Differentiating strategies are thus required to generate endothelial cells (ECs), a major component of the BBB. Several hiPSC-based protocols have reported the generation of in vitro models with significant differences in barrier properties. We studied in depth the properties of iPSCs byproducts from two protocols that have been established to yield these in vitro barrier models. Our analysis/study reveals that iPSCs derivatives endowed with EC features yield high permeability models while the cells that exhibit outstanding barrier properties show principally epithelial cell-like (EpC) features. We found that models containing EpC-like cells express tight junction proteins, transporters/efflux pumps and display a high functional tightness with very low permeability, which are features commonly shared between BBB and epithelial barriers. Our study demonstrates that hiPSC-based BBB models need extensive characterization beforehand and that a reliable human BBB model containing EC-like cells and displaying low permeability is still needed.
Collapse
Affiliation(s)
- Stéphane D Girard
- Institute of NeuroPhysiopathology, INP, CNRS, Aix-Marseille University, Marseille, France
- Faculty of Medicine, VECT-HORUS SAS, Marseille, France
| | | | - Yves Molino
- Faculty of Medicine, VECT-HORUS SAS, Marseille, France
| | | | - Louise Greetham
- Institute of NeuroPhysiopathology, INP, CNRS, Aix-Marseille University, Marseille, France
| | - Michel Khrestchatisky
- Institute of NeuroPhysiopathology, INP, CNRS, Aix-Marseille University, Marseille, France
| | - Emmanuel Nivet
- Institute of NeuroPhysiopathology, INP, CNRS, Aix-Marseille University, Marseille, France
| |
Collapse
|
47
|
Experimental Models of In Vitro Blood-Brain Barrier for CNS Drug Delivery: An Evolutionary Perspective. Int J Mol Sci 2023; 24:ijms24032710. [PMID: 36769032 PMCID: PMC9916529 DOI: 10.3390/ijms24032710] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Revised: 01/24/2023] [Accepted: 01/27/2023] [Indexed: 02/04/2023] Open
Abstract
Central nervous system (CNS) disorders represent one of the leading causes of global health burden. Nonetheless, new therapies approved against these disorders are among the lowest compared to their counterparts. The absence of reliable and efficient in vitro blood-brain barrier (BBB) models resembling in vivo barrier properties stands out as a significant roadblock in developing successful therapy for CNS disorders. Therefore, advancement in the creation of robust and sensitive in vitro BBB models for drug screening might allow us to expedite neurological drug development. This review discusses the major in vitro BBB models developed as of now for exploring the barrier properties of the cerebral vasculature. Our main focus is describing existing in vitro models, including the 2D transwell models covering both single-layer and co-culture models, 3D organoid models, and microfluidic models with their construction, permeability measurement, applications, and limitations. Although microfluidic models are better at recapitulating the in vivo properties of BBB than other models, significant gaps still exist for their use in predicting the performance of neurotherapeutics. However, this comprehensive account of in vitro BBB models can be useful for researchers to create improved models in the future.
Collapse
|
48
|
Mamana J, Humber GM, Espinal ER, Seo S, Vollmuth N, Sin J, Kim BJ. Coxsackievirus B3 infects and disrupts human induced-pluripotent stem cell derived brain-like endothelial cells. Front Cell Infect Microbiol 2023; 13:1171275. [PMID: 37139492 PMCID: PMC10149843 DOI: 10.3389/fcimb.2023.1171275] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Accepted: 03/30/2023] [Indexed: 05/05/2023] Open
Abstract
Coxsackievirus B3 (CVB3) is a significant human pathogen that is commonly found worldwide. CVB3 among other enteroviruses, are the leading causes of aseptic meningo-encephalitis which can be fatal especially in young children. How the virus gains access to the brain is poorly-understood, and the host-virus interactions that occur at the blood-brain barrier (BBB) is even less-characterized. The BBB is a highly specialized biological barrier consisting primarily of brain endothelial cells which possess unique barrier properties and facilitate the passage of nutrients into the brain while restricting access to toxins and pathogens including viruses. To determine the effects of CVB3 infection on the BBB, we utilized a model of human induced-pluripotent stem cell-derived brain-like endothelial cells (iBECs) to ascertain if CVB3 infection may alter barrier cell function and overall survival. In this study, we determined that these iBECs indeed are susceptible to CVB3 infection and release high titers of extracellular virus. We also determined that infected iBECs maintain high transendothelial electrical resistance (TEER) during early infection despite possessing high viral load. TEER progressively declines at later stages of infection. Interestingly, despite the high viral burden and TEER disruptions at later timepoints, infected iBEC monolayers remain intact, indicating a low degree of late-stage virally-mediated cell death, which may contribute to prolonged viral shedding. We had previously reported that CVB3 infections rely on the activation of transient receptor vanilloid potential 1 (TRPV1) and found that inhibiting TRPV1 activity with SB-366791 significantly limited CVB3 infection of HeLa cervical cancer cells. Similarly in this study, we observed that treating iBECs with SB-366791 significantly reduced CVB3 infection, which suggests that not only can this drug potentially limit viral entry into the brain, but also demonstrates that this infection model could be a valuable platform for testing antiviral treatments of neurotropic viruses. In all, our findings elucidate the unique effects of CVB3 infection on the BBB and shed light on potential mechanisms by which the virus can initiate infections in the brain.
Collapse
Affiliation(s)
- Julia Mamana
- Department of Biological Sciences, University of Alabama, Tuscaloosa, AL, United States
| | - Gabrielle M. Humber
- Department of Biological Sciences, University of Alabama, Tuscaloosa, AL, United States
| | - Eric R. Espinal
- Department of Biological Sciences, University of Alabama, Tuscaloosa, AL, United States
| | - Soojung Seo
- Department of Biological Sciences, University of Alabama, Tuscaloosa, AL, United States
| | - Nadine Vollmuth
- Department of Biological Sciences, University of Alabama, Tuscaloosa, AL, United States
| | - Jon Sin
- Department of Biological Sciences, University of Alabama, Tuscaloosa, AL, United States
- *Correspondence: Jon Sin, ; Brandon J. Kim,
| | - Brandon J. Kim
- Department of Biological Sciences, University of Alabama, Tuscaloosa, AL, United States
- Department of Microbiology, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL, United States
- Center for Convergent Biosciences and Medicine, University of Alabama, Tuscaloosa, AL, United States
- Alabama Life Research Institute, University of Alabama, Tuscaloosa, AL, United States
- *Correspondence: Jon Sin, ; Brandon J. Kim,
| |
Collapse
|
49
|
Blood brain barrier-on-a-chip to model neurological diseases. J Drug Deliv Sci Technol 2023. [DOI: 10.1016/j.jddst.2023.104174] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
|
50
|
Human mini-blood-brain barrier models for biomedical neuroscience research: a review. Biomater Res 2022; 26:82. [PMID: 36527159 PMCID: PMC9756735 DOI: 10.1186/s40824-022-00332-z] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Accepted: 12/01/2022] [Indexed: 12/23/2022] Open
Abstract
The human blood-brain barrier (BBB) is a unique multicellular structure that is in critical demand for fundamental neuroscience studies and therapeutic evaluation. Despite substantial achievements in creating in vitro human BBB platforms, challenges in generating specifics of physiopathological relevance are viewed as impediments to the establishment of in vitro models. In this review, we provide insight into the development and deployment of in vitro BBB models that allow investigation of the physiology and pathology of neurological therapeutic avenues. First, we highlight the critical components, including cell sources, biomaterial glue collections, and engineering techniques to reconstruct a miniaturized human BBB. Second, we describe recent breakthroughs in human mini-BBBs for investigating biological mechanisms in neurology. Finally, we discuss the application of human mini-BBBs to medical approaches. This review provides strategies for understanding neurological diseases, a validation model for drug discovery, and a potential approach for generating personalized medicine.
Collapse
|