1
|
Salamah M, Sipos B, Schelz Z, Zupkó I, Kiricsi Á, Szalenkó-Tőkés Á, Rovó L, Katona G, Balogh GT, Csóka I. Development, in vitro and ex vivo characterization of lamotrigine-loaded bovine serum albumin nanoparticles using QbD approach. Drug Deliv 2025; 32:2460693. [PMID: 39901331 PMCID: PMC11795762 DOI: 10.1080/10717544.2025.2460693] [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: 09/04/2024] [Revised: 11/26/2024] [Accepted: 01/24/2025] [Indexed: 02/05/2025] Open
Abstract
The present study aimed to prepare and optimize lamotrigine-loaded bovine serum albumin nanoparticles (LAM-NP) using the Quality by Design (QbD) approach and to investigate both the in vitro and ex vivo effects of different cross-linking agents glutaraldehyde (GLUT), glucose (GLUC) and 1-(3-dimethylaminutesopropyl)-3-ethylcarbodiimide hydrochloride (EDC) on intranasal applicability. Cross-linked LAM-NP from EDC (NP-EDC-1) showed the lowest Z-average value (163.7 ± 1.9 nm) and drug encapsulation efficacy (EE%) of 97.31 ± 0.17%. The drug release of GLUC cross-linked LAM-NP (NP-GLUC-9), glutaraldehyde cross-linked LAM-NP (NP-GLUT-2), and NP-EDC-1 at blood circulation conditions was higher than the initial LAM. The results of the blood-brain barrier parallel artificial membrane permeability assay (BBB-PAMPA) showed an increase in the permeability of LAM through the BBB with NP-GLUC-9 and an increase in flux with all selected formulations. The ex vivo study showed that LAM diffusion from the selected formulations through the human nasal mucosa was higher than in case of initial LAM. The cytotoxicity study indicated that BSA-NP reduced LAM toxicity, and GLUC 9 mM and EDC 1 mg could be alternative cross-linking agents to avoid GLUT 2% v/v toxicity. Furthermore, permeability through Caco-2 cells showed that nasal epithelial transport/absorption of LAM was improved by using BSA-NPs. The use of BSA-NP may be a promising approach to enhance the solubility, permeability through BBB and decrease the frequency of dosing and adverse effects of LAM.
Collapse
Affiliation(s)
- Maryana Salamah
- Institute of Pharmaceutical Technology and Regulatory Affairs, Faculty of Pharmacy, University of Szeged, Szeged, Hungary
- Institute of Pharmacodynamics and Biopharmacy, Faculty of Pharmacy, University of Szeged, Szeged, Hungary
| | - Bence Sipos
- Institute of Pharmaceutical Technology and Regulatory Affairs, Faculty of Pharmacy, University of Szeged, Szeged, Hungary
| | - Zsuzsanna Schelz
- Institute of Pharmacodynamics and Biopharmacy, Faculty of Pharmacy, University of Szeged, Szeged, Hungary
| | - István Zupkó
- Institute of Pharmacodynamics and Biopharmacy, Faculty of Pharmacy, University of Szeged, Szeged, Hungary
| | - Ágnes Kiricsi
- Department of Oto-Rhino-Laryngology and Head-Neck Surgery, University of Szeged, Szeged, Hungary
| | - Ágnes Szalenkó-Tőkés
- Department of Oto-Rhino-Laryngology and Head-Neck Surgery, University of Szeged, Szeged, Hungary
| | - László Rovó
- Department of Oto-Rhino-Laryngology and Head-Neck Surgery, University of Szeged, Szeged, Hungary
| | - Gábor Katona
- Institute of Pharmaceutical Technology and Regulatory Affairs, Faculty of Pharmacy, University of Szeged, Szeged, Hungary
| | - György Tibor Balogh
- Department of Pharmaceutical Chemistry, Semmelweis University, Budapest, Hungary
- Center for Pharmacology and Drug Research & Development, Semmelweis University, Budapest, Hungary
| | - Ildikó Csóka
- Institute of Pharmaceutical Technology and Regulatory Affairs, Faculty of Pharmacy, University of Szeged, Szeged, Hungary
| |
Collapse
|
2
|
Zandona A, Szecskó A, Žunec S, Jovanović IN, Bušić V, Sokač DG, Deli MA, Katalinić M, Veszelka S. Nicotinamide derivatives protect the blood-brain barrier against oxidative stress. Biomed Pharmacother 2025; 186:118018. [PMID: 40174541 DOI: 10.1016/j.biopha.2025.118018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2025] [Revised: 03/24/2025] [Accepted: 03/27/2025] [Indexed: 04/04/2025] Open
Abstract
Nicotinamides play a crucial role in energy metabolism and maintenance of the redox homeostasis counteracting oxidative stress and elevated reactive oxidative species (ROS) in human cells. The levels of nicotinamides decline with age and are associated with various pathologies, including ones linked with the blood-brain barrier disorder. Therefore, the investigation of the bioactivity of synthetic nicotinamide derivates (NAs) and evaluation of their potential to protect the blood-brain barrier (BBB) from oxidative stress is emerging as an important new strategy. In the current study, we tested different NAs as potential exogenous substitutes for such biological processes. All tested derivatives were non-toxic and attenuated elevation of ROS production in brain endothelial cells induced by tert-butyl hydroperoxide (tBHP), but one specifically was protective on the cell-cultured model of the BBB. The most promising NA was a derivative containing methoxy moiety (NA-4OCH3), which not only increased cell impedance, but had a protective effect on brain endothelial cells barrier against tBHP-induced oxidative stress on several levels: reducing the ROS level and restoring the activity of glutathione, mitochondrial membrane potential, superoxide dismutase enzymes activity to the basal level. In addition, NA-4OCH3 increased the integrity of both human and rat cell-based BBB model after tBHP-treatment seen by the elevated transendothelial electrical resistance, tight junction protein claudin-5 level as well as the decreased permeability of markers across the barrier. This study highlights novel approach to protect the BBB from oxidative stress-induced dysfunction, positioning NA-4OCH3 as potential neuroprotective agent for ROS-mediated disease interventions, with implications for neurodegeneration and BBB.
Collapse
Affiliation(s)
- Antonio Zandona
- Division of Toxicology, Institute for Medical Research and Occupational Health, Ksaverska cesta 2, Zagreb HR-10001, Croatia
| | - Anikó Szecskó
- Institute of Biophysics, HUN-REN Biological Research Centre, Temesvári krt. 62, Szeged 6726, Hungary; Doctoral School of Biology, University of Szeged, Szeged, Hungary
| | - Suzana Žunec
- Division of Toxicology, Institute for Medical Research and Occupational Health, Ksaverska cesta 2, Zagreb HR-10001, Croatia
| | - Ivana Novak Jovanović
- Division of Toxicology, Institute for Medical Research and Occupational Health, Ksaverska cesta 2, Zagreb HR-10001, Croatia
| | - Valentina Bušić
- Faculty of Food Technology Osijek, Josip Juraj Strossmayer University of Osijek, Kuhačeva 20, Osijek HR-31000, Croatia
| | - Dajana Gašo Sokač
- Faculty of Food Technology Osijek, Josip Juraj Strossmayer University of Osijek, Kuhačeva 20, Osijek HR-31000, Croatia
| | - Mária A Deli
- Institute of Biophysics, HUN-REN Biological Research Centre, Temesvári krt. 62, Szeged 6726, Hungary
| | - Maja Katalinić
- Division of Toxicology, Institute for Medical Research and Occupational Health, Ksaverska cesta 2, Zagreb HR-10001, Croatia.
| | - Szilvia Veszelka
- Institute of Biophysics, HUN-REN Biological Research Centre, Temesvári krt. 62, Szeged 6726, Hungary.
| |
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
|
Szecskó A, Mészáros M, Simões B, Cavaco M, Chaparro C, Porkoláb G, Castanho MARB, Deli MA, Neves V, Veszelka S. PepH3-modified nanocarriers for delivery of therapeutics across the blood-brain barrier. Fluids Barriers CNS 2025; 22:31. [PMID: 40170024 PMCID: PMC11959756 DOI: 10.1186/s12987-025-00641-0] [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: 08/30/2024] [Accepted: 03/14/2025] [Indexed: 04/03/2025] Open
Abstract
BACKGROUND Nanocarriers targeting the blood-brain barrier (BBB) are promising drug delivery systems to enhance the penetration of therapeutic molecules into the brain. Immunotherapy, particularly monoclonal antibodies designed to bind amyloid-beta peptides have become a promising strategy for Alzheimer's disease, but ensuring efficacy and safety is challenging and crucial for these therapies. Our aim was to develop an innovative nanocarriers conjugated with PepH3, a cationic peptide derived from Dengue virus type-2 capsid protein that crosses the BBB and acts as a shuttle peptide for the encapsulated single domain antibody (sdAb) recognizing Aβ oligomers. RESULTS PepH3 peptide enhanced the uptake of the nanoparticles (NPs) into brain endothelial cells, and transcytosis of sdAb, as a potential therapeutic molecule, across both rat and human BBB culture models. The cargo uptake was a temperature dependent active process that was reduced by metabolic and endocytosis inhibitors. The cellular uptake of the cationic PepH3-tagged NPs decreased when the negative surface charge of brain endothelial cells became more positive after treatments with a cationic lipid or with neuraminidase by digesting the glycocalyx. The NPs colocalized mostly with endoplasmic reticulum and Golgi apparatus and not with lysosomes, indicating the cargo may avoid cellular degradation. CONCLUSIONS Our results support that combination of NPs with a potential brain shuttle peptide such as PepH3 peptide can improve the delivery of antibody fragments across the BBB.
Collapse
Affiliation(s)
- Anikó Szecskó
- Biological Barriers Research Group, Institute of Biophysics, HUN-REN Biological Research Centre, Szeged, Hungary
- Doctoral School of Biology, University of Szeged, Szeged, Hungary
| | - Mária Mészáros
- Biological Barriers Research Group, Institute of Biophysics, HUN-REN Biological Research Centre, Szeged, Hungary
- One Health Institute, Faculty of Health Sciences, University of Debrecen, Nagyerdei krt. 98, Debrecen, H-4032, Hungary
| | - Beatriz Simões
- Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
| | - Marco Cavaco
- Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
| | - Catarina Chaparro
- Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
| | - Gergő Porkoláb
- Biological Barriers Research Group, Institute of Biophysics, HUN-REN Biological Research Centre, Szeged, Hungary
| | - Miguel A R B Castanho
- Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
| | - Mária A Deli
- Biological Barriers Research Group, Institute of Biophysics, HUN-REN Biological Research Centre, Szeged, Hungary
| | - Vera Neves
- Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal.
| | - Szilvia Veszelka
- Biological Barriers Research Group, Institute of Biophysics, HUN-REN Biological Research Centre, Szeged, Hungary.
| |
Collapse
|
5
|
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
|
6
|
Koru I, Atasever-Arslan B. Protective effects of sesamol against cigarette smoke toxicity on the blood-brain barrier. BMC Complement Med Ther 2025; 25:68. [PMID: 39987119 PMCID: PMC11846467 DOI: 10.1186/s12906-025-04796-z] [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: 10/04/2024] [Accepted: 01/29/2025] [Indexed: 02/24/2025] Open
Abstract
Cigarette smoke comprises nicotine, reactive oxygen species (ROS), and carcinogens, which can induce oxidative stress and inflammation, leading to disruption of the blood-brain barrier. This study utilized cigarette smoke extract (CSE) in an in vitro model of the blood-brain barrier (BBB).Sesamol is a phenolic compound derived from Sesamum indicum L. Its potential to reduce inflammation and provide protection was also examined. As a result of the study, it was found that CSE significantly increases permeability by degrading the BBB, whereas a protective effect was observed in the sesamol-incubated group within the BBB model. While the Sesamol + CSE group does not entirely prevent the damage induced by CSE in the barrier, it does exhibit a mitigating effect on the damage.In HUVEC cells, a significant decrease in IL-8 levels was observed in sesamol and sesamol + CSE groups. In T98G cells, IL-8 levels were elevated in the CSE group, while a reduction was observed in the sesamol and sesamol + CSE groups. TNF-α levels went up in the CSE group but down in the sesamol and sesamol + CSE groups in T98G cells. Furthermore, the IL-6 levels were significantly increased in both the sesamol and sesamol + CSE groups in HUVEC cells, while a decrease was noted in T98G cells in sesamol treatment. The increase in IL-8 and TNF-α levels in T98G cells due to CSE indicates an inflammatory response. It can contribute to the enhanced BBB permeability. As a result, sesamol reduced inflammation caused by CSE by controlling IL-8, IL-6, and TNF-α. This molecule may serve a therapeutic role by diminishing inflammation and protecting the blood-brain barrier from damage.
Collapse
Affiliation(s)
- Ildem Koru
- Department of Biotechnology, Graduate School of Science, Uskudar University, Istanbul, Turkey
| | - Belkıs Atasever-Arslan
- Department of Molecular Biology and Genetics, Faculty of Engineering and Natural Sciences, Uskudar University, Istanbul, Turkey.
| |
Collapse
|
7
|
Kocsis AE, Kucsápszky N, Santa-Maria AR, Hunyadi A, Deli MA, Walter FR. Much More than Nutrients: The Protective Effects of Nutraceuticals on the Blood-Brain Barrier in Diseases. Nutrients 2025; 17:766. [PMID: 40077636 PMCID: PMC11901837 DOI: 10.3390/nu17050766] [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: 01/22/2025] [Revised: 02/14/2025] [Accepted: 02/19/2025] [Indexed: 03/14/2025] Open
Abstract
The dysfunction of the blood-brain barrier (BBB) is well described in several diseases, and is considered a pathological factor in many neurological disorders. This review summarizes the most important groups of natural compounds, including alkaloids, flavonoids, anthocyanidines, carotenoids, lipids, and vitamins that were investigated for their potential protective effects on brain endothelium. The brain penetration of these compounds and their interaction with BBB efflux transporters and solute carriers are discussed. The cerebrovascular endothelium is considered a therapeutic target for natural compounds in diseases. In preclinical studies modeling systemic and central nervous system diseases, nutraceuticals exerted beneficial effects on the BBB. In vivo, they decreased BBB permeability, brain edema, astrocyte swelling, and morphological changes in the vessel structure and basal lamina. At the level of brain endothelial cells, nutraceuticals increased cell survival and decreased apoptosis. From the general endothelial functions, decreased angiogenesis and increased levels of vasodilating agents were demonstrated. From the BBB functions, elevated barrier integrity by tightened intercellular junctions, and increased expression and activity of BBB transporters, such as efflux pumps, solute carriers, and metabolic enzymes, were shown. Nutraceuticals enhanced the antioxidative defense and exerted anti-inflammatory effects at the BBB. The most important signaling changes mediating the increased cell survival and BBB stability were the activation of the WNT, PI3K-AKT, and NRF2 pathways, and inhibition of the MAPK, JNK, ERK, and NF-κB pathways. Nutraceuticals represent a valuable source of new potentially therapeutic molecules to treat brain diseases by protecting the BBB.
Collapse
Affiliation(s)
- Anna E. Kocsis
- Biological Barriers Research Group, Institute of Biophysics, HUN-REN Biological Research Centre, H-6726 Szeged, Hungary; (A.E.K.); (N.K.)
| | - Nóra Kucsápszky
- Biological Barriers Research Group, Institute of Biophysics, HUN-REN Biological Research Centre, H-6726 Szeged, Hungary; (A.E.K.); (N.K.)
| | - Ana Raquel Santa-Maria
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02115, USA
| | - Attila Hunyadi
- Institute of Pharmacognosy, University of Szeged, Eötvös u. 6, H-6720 Szeged, Hungary;
- Interdisciplinary Centre of Natural Products, University of Szeged, Eötvös u. 6, H-6720 Szeged, Hungary
- HUN-REN-SZTE Biologically Active Natural Products Research Group, Eötvös u. 6, H-6720 Szeged, Hungary
- Graduate Institute of Natural Products, Kaohsiung Medical University, Shih-Chuan 1st Rd. 100, Kaohsiung 807, Taiwan
| | - Mária A. Deli
- Biological Barriers Research Group, Institute of Biophysics, HUN-REN Biological Research Centre, H-6726 Szeged, Hungary; (A.E.K.); (N.K.)
| | - Fruzsina R. Walter
- Biological Barriers Research Group, Institute of Biophysics, HUN-REN Biological Research Centre, H-6726 Szeged, Hungary; (A.E.K.); (N.K.)
| |
Collapse
|
8
|
Asibor YE, Oyebamiji AK, Latona DF, Semire B. Computational screening of phytochemicals present in some Nigerian medicinal plants against sickle cell disease. Sci Rep 2024; 14:26368. [PMID: 39487201 PMCID: PMC11530684 DOI: 10.1038/s41598-024-75078-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: 07/05/2024] [Accepted: 10/01/2024] [Indexed: 11/04/2024] Open
Abstract
Four hundred Phytochemical (bio-active) compounds having predictive activity for treating Sickle Cell Anemia were screened, using PASS online computational resource. Twenty-six compounds out of the four hundred compounds which showed high probability for treating sickle were further screened for pharmacokinetics profiles (ADMET properties) using SwissAdmet, AdmetSAR 2 and Pro-tox II online resources. Only thirteen compounds that displayed good ADMET properties from the twenty-six were further used for DFT calculations and molecular docking against carbonmonoxy sickle hemoglobin (PDB ID: 5E6E). Molecular docking analysis reinforced by DFT calculations showed that two compounds, phenanthrene-5,6-dione (A9) and 2-(3,4-dihydroxyphenyl)-5,7-dihydroxychromen-4-one (A13, Luteolin) had the best binding affinity of - 8.3 and - 8.9 kcal/mol, respectively, compared to voxelotor (GBT-440), a drug use in treating sickle cell disease. Molecular dynamic simulations showed that 2-(3,4-dihydroxyphenyl)-5,7-dihydroxychromen-4-one (A13, Luteolin) is highly stable with the protein than voxelotor.
Collapse
Affiliation(s)
| | - Abel Kolawole Oyebamiji
- Department of Chemistry and Industrial Chemistry, Bowen University, Iwo, Osun State, Nigeria
| | - Dayo Felix Latona
- Department of Pure and Applied Chemistry, Osun State University, Osogbo, Nigeria
| | - Banjo Semire
- Computational Chemistry Laboratory, Department of Pure and Applied Chemistry, Ladoke Akintola University of Technology, Ogbomoso, Oyo State, Nigeria
| |
Collapse
|
9
|
Pires CL, Moreno MJ. Improving the Accuracy of Permeability Data to Gain Predictive Power: Assessing Sources of Variability in Assays Using Cell Monolayers. MEMBRANES 2024; 14:157. [PMID: 39057665 PMCID: PMC11278619 DOI: 10.3390/membranes14070157] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Revised: 07/05/2024] [Accepted: 07/11/2024] [Indexed: 07/28/2024]
Abstract
The ability to predict the rate of permeation of new compounds across biological membranes is of high importance for their success as drugs, as it determines their efficacy, pharmacokinetics, and safety profile. In vitro permeability assays using Caco-2 monolayers are commonly employed to assess permeability across the intestinal epithelium, with an extensive number of apparent permeability coefficient (Papp) values available in the literature and a significant fraction collected in databases. The compilation of these Papp values for large datasets allows for the application of artificial intelligence tools for establishing quantitative structure-permeability relationships (QSPRs) to predict the permeability of new compounds from their structural properties. One of the main challenges that hinders the development of accurate predictions is the existence of multiple Papp values for the same compound, mostly caused by differences in the experimental protocols employed. This review addresses the magnitude of the variability within and between laboratories to interpret its impact on QSPR modelling, systematically and quantitatively assessing the most common sources of variability. This review emphasizes the importance of compiling consistent Papp data and suggests strategies that may be used to obtain such data, contributing to the establishment of robust QSPRs with enhanced predictive power.
Collapse
Affiliation(s)
- Cristiana L. Pires
- Coimbra Chemistry Center—Institute of Molecular Sciences (CQC-IMS), University of Coimbra, 3004-535 Coimbra, Portugal
- Chemistry Department, Faculty of Science and Technology, University of Coimbra, 3004-535 Coimbra, Portugal
| | - Maria João Moreno
- Coimbra Chemistry Center—Institute of Molecular Sciences (CQC-IMS), University of Coimbra, 3004-535 Coimbra, Portugal
- Chemistry Department, Faculty of Science and Technology, University of Coimbra, 3004-535 Coimbra, Portugal
| |
Collapse
|
10
|
Bhatt M, Lazzarin E, Alberto-Silva AS, Domingo G, Zerlotti R, Gradisch R, Bazzone A, Sitte HH, Stockner T, Bossi E. Unveiling the crucial role of betaine: modulation of GABA homeostasis via SLC6A1 transporter (GAT1). Cell Mol Life Sci 2024; 81:269. [PMID: 38884791 PMCID: PMC11335192 DOI: 10.1007/s00018-024-05309-w] [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: 04/12/2024] [Revised: 06/04/2024] [Accepted: 06/06/2024] [Indexed: 06/18/2024]
Abstract
Betaine is an endogenous osmolyte that exhibits therapeutic potential by mitigating various neurological disorders. However, the underlying cellular and molecular mechanisms responsible for its neuroprotective effects remain puzzling.In this study, we describe a possible mechanism behind the positive impact of betaine in preserving neurons from excitotoxicity. Here we demonstrate that betaine at low concentration modulates the GABA uptake by GAT1 (slc6a1), the predominant GABA transporter in the central nervous system. This modulation occurs through the temporal inhibition of the transporter, wherein prolonged occupancy by betaine impedes the swift transition of the transporter to the inward conformation. Importantly, the modulatory effect of betaine on GAT1 is reversible, as the blocking of GAT1 disappears with increased extracellular GABA. Using electrophysiology, mass spectroscopy, radiolabelled cellular assay, and molecular dynamics simulation we demonstrate that betaine has a dual role in GAT1: at mM concentration acts as a slow substrate, and at µM as a temporal blocker of GABA, when it is below its K0.5. Given this unique modulatory characteristic and lack of any harmful side effects, betaine emerges as a promising neuromodulator of the inhibitory pathways improving GABA homeostasis via GAT1, thereby conferring neuroprotection against excitotoxicity.
Collapse
Affiliation(s)
- Manan Bhatt
- Department of Biotechnology and Life Science, Laboratory of Cellular and Molecular Physiology, University of Insubria, Via J. H. Dunant 3, 21100, Varese, Italy
| | - Erika Lazzarin
- Center for Physiology and Pharmacology, Institute of Pharmacology, Medical University of Vienna, 1090, Vienna, Austria
| | - Ana Sofia Alberto-Silva
- Center for Physiology and Pharmacology, Institute of Pharmacology, Medical University of Vienna, 1090, Vienna, Austria
| | - Guido Domingo
- Department of Biotechnology and Life Science, Laboratory of Cellular and Molecular Physiology, University of Insubria, Via J. H. Dunant 3, 21100, Varese, Italy
| | - Rocco Zerlotti
- Nanion Technologies GmbH, Ganghoferstr. 70a, 80339, Munich, Germany
| | - Ralph Gradisch
- Institute of Pharmacology and Toxicology, University of Zurich, Zurich, Switzerland
| | - Andre Bazzone
- Nanion Technologies GmbH, Ganghoferstr. 70a, 80339, Munich, Germany
| | - Harald H Sitte
- Center for Physiology and Pharmacology, Institute of Pharmacology, Medical University of Vienna, 1090, Vienna, Austria
- Hourani Center for Applied Scientific Research, Al-Ahliyya Amman University, Amman, 19328, Jordan
- Center for Addiction Research and Science, Medical University of Vienna, 1090, Vienna, Austria
| | - Thomas Stockner
- Center for Physiology and Pharmacology, Institute of Pharmacology, Medical University of Vienna, 1090, Vienna, Austria
| | - Elena Bossi
- Department of Biotechnology and Life Science, Laboratory of Cellular and Molecular Physiology, University of Insubria, Via J. H. Dunant 3, 21100, Varese, Italy.
- Centre for Neuroscience, University of Insubria, 21100, Varese, Italy.
| |
Collapse
|
11
|
Eltanameli B, Piñeiro-Llanes J, Cristofoletti R. Recent advances in cell-based in vitro models for predicting drug permeability across brain, intestinal, and pulmonary barriers. Expert Opin Drug Metab Toxicol 2024; 20:439-458. [PMID: 38850058 DOI: 10.1080/17425255.2024.2366390] [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: 02/26/2024] [Accepted: 06/06/2024] [Indexed: 06/09/2024]
Abstract
INTRODUCTION Recent years have witnessed remarkable progress in the development of cell-based in vitro models aimed at predicting drug permeability, particularly focusing on replicating the barrier properties of the blood-brain barrier (BBB), intestinal epithelium, and lung epithelium. AREA COVERED This review provides an overview of 2D in vitro platforms, including monocultures and co-culture systems, highlighting their respective advantages and limitations. Additionally, it discusses tools and techniques utilized to overcome these limitations, paving the way for more accurate predictions of drug permeability. Furthermore, this review delves into emerging technologies, particularly microphysiological systems (MPS), encompassing static platforms such as organoids and dynamic platforms like microfluidic devices. Literature searches were performed using PubMed and Google Scholar. We focus on key terms such as in vitro permeability models, MPS, organoids, intestine, BBB, and lungs. EXPERT OPINION The potential of these MPS to mimic physiological conditions more closely offers promising avenues for drug permeability assessment. However, transitioning these advanced models from bench to industry requires rigorous validation against regulatory standards. Thus, there is a pressing need to validate MPS to industry and regulatory agency standards to exploit their potential in drug permeability prediction fully. This review underscores the importance of such validation processes to facilitate the translation of these innovative technologies into routine pharmaceutical practice.
Collapse
Affiliation(s)
- Bassma Eltanameli
- Center for Pharmacometrics & Systems Pharmacology, Department of Pharmaceutics, College of Pharmacy, University of Florida, Orlando, FL, USA
- Department of Pharmaceutics, Faculty of Pharmacy, Mansoura University, Mansoura, Egypt
| | - Janny Piñeiro-Llanes
- Center for Pharmacometrics & Systems Pharmacology, Department of Pharmaceutics, College of Pharmacy, University of Florida, Orlando, FL, USA
| | - Rodrigo Cristofoletti
- Center for Pharmacometrics & Systems Pharmacology, Department of Pharmaceutics, College of Pharmacy, University of Florida, Orlando, FL, USA
| |
Collapse
|
12
|
Wasielewska JM, Szostak K, McInnes LE, Quek H, Chaves JCS, Liddell JR, Koistinaho J, Oikari LE, Donnelly PS, White AR. Patient-Derived Blood-Brain Barrier Model for Screening Copper Bis(thiosemicarbazone) Complexes as Potential Therapeutics in Alzheimer's Disease. ACS Chem Neurosci 2024; 15:1432-1455. [PMID: 38477556 DOI: 10.1021/acschemneuro.3c00743] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/14/2024] Open
Abstract
Alzheimer's disease (AD) is the most prevalent cause of dementia characterized by a progressive cognitive decline. Addressing neuroinflammation represents a promising therapeutic avenue to treat AD; however, the development of effective antineuroinflammatory compounds is often hindered by their limited blood-brain barrier (BBB) permeability. Consequently, there is an urgent need for accurate, preclinical AD patient-specific BBB models to facilitate the early identification of immunomodulatory drugs capable of efficiently crossing the human AD BBB. This study presents a unique approach to BBB drug permeability screening as it utilizes the familial AD patient-derived induced brain endothelial-like cell (iBEC)-based model, which exhibits increased disease relevance and serves as an improved BBB drug permeability assessment tool when compared to traditionally employed in vitro models. To demonstrate its utility as a small molecule drug candidate screening platform, we investigated the effects of diacetylbis(N(4)-methylthiosemicarbazonato)copper(II) (CuII(atsm)) and a library of metal bis(thiosemicarbazone) complexes─a class of compounds exhibiting antineuroinflammatory therapeutic potential in neurodegenerative disorders. By evaluating the toxicity, cellular accumulation, and permeability of those compounds in the AD patient-derived iBEC, we have identified 3,4-hexanedione bis(N(4)-methylthiosemicarbazonato)copper(II) (CuII(dtsm)) as a candidate with good transport across the AD BBB. Furthermore, we have developed a multiplex approach where AD patient-derived iBEC were combined with immune modulators TNFα and IFNγ to establish an in vitro model representing the characteristic neuroinflammatory phenotype at the patient's BBB. Here, we observed that treatment with CuII(dtsm) not only reduced the expression of proinflammatory cytokine genes but also reversed the detrimental effects of TNFα and IFNγ on the integrity and function of the AD iBEC monolayer. This suggests a novel pathway through which copper bis(thiosemicarbazone) complexes may exert neurotherapeutic effects on AD by mitigating BBB neuroinflammation and related BBB integrity impairment. Together, the presented model provides an effective and easily scalable in vitro BBB platform for screening AD drug candidates. Its improved translational potential makes it a valuable tool for advancing the development of metal-based compounds aimed at modulating neuroinflammation in AD.
Collapse
Affiliation(s)
- Joanna M Wasielewska
- Mental Health and Neuroscience Program, QIMR Berghofer Medical Research Institute, Herston, QLD 4006, Australia
- Faculty of Medicine, University of Queensland, Herston, QLD 4006, Australia
| | - Kathryn Szostak
- School of Chemistry, Bio21 Institute for Molecular Science and Biotechnology, The University of Melbourne, Parkville, VIC 3010, Australia
| | - Lachlan E McInnes
- School of Chemistry, Bio21 Institute for Molecular Science and Biotechnology, The University of Melbourne, Parkville, VIC 3010, Australia
| | - Hazel Quek
- Mental Health and Neuroscience Program, QIMR Berghofer Medical Research Institute, Herston, QLD 4006, Australia
- School of Biomedical Science, University of Queensland, St. Lucia, QLD 4067, Australia
| | - Juliana C S Chaves
- Mental Health and Neuroscience Program, QIMR Berghofer Medical Research Institute, Herston, QLD 4006, Australia
- School of Biomedical Sciences, Faculty of Health, Queensland University of Technology, Brisbane, QLD 4000, Australia
| | - Jeffrey R Liddell
- Department of Anatomy and Physiology, The University of Melbourne, Parkville, VIC 3010, Australia
| | - Jari Koistinaho
- Drug Research Program, Division of Pharmacology and Pharmacotherapy, University of Helsinki, Helsinki 00014,Finland
- Neuroscience Centre, Helsinki Institute of Life Science, University of Helsinki, Helsinki 00014, Finland
| | - Lotta E Oikari
- Mental Health and Neuroscience Program, QIMR Berghofer Medical Research Institute, Herston, QLD 4006, Australia
| | - Paul S Donnelly
- School of Chemistry, Bio21 Institute for Molecular Science and Biotechnology, The University of Melbourne, Parkville, VIC 3010, Australia
| | - Anthony R White
- Mental Health and Neuroscience Program, QIMR Berghofer Medical Research Institute, Herston, QLD 4006, Australia
- School of Biomedical Science, University of Queensland, St. Lucia, QLD 4067, Australia
| |
Collapse
|
13
|
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
|
14
|
Vágvölgyi M, Laczkó D, Santa-Maria AR, Vigh JP, Walter FR, Berkecz R, Deli MA, Tóth G, Hunyadi A. 17-Oxime ethers of oxidized ecdysteroid derivatives modulate oxidative stress in human brain endothelial cells and dose-dependently might protect or damage the blood-brain barrier. PLoS One 2024; 19:e0290526. [PMID: 38386637 PMCID: PMC10883584 DOI: 10.1371/journal.pone.0290526] [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: 08/09/2023] [Accepted: 01/07/2024] [Indexed: 02/24/2024] Open
Abstract
20-Hydroxyecdysone and several of its oxidized derivatives exert cytoprotective effect in mammals including humans. Inspired by this bioactivity of ecdysteroids, in the current study it was our aim to prepare a set of sidechain-modified derivatives and to evaluate their potential to protect the blood-brain barrier (BBB) from oxidative stress. Six novel ecdysteroids, including an oxime and five oxime ethers, were obtained through regioselective synthesis from a sidechain-cleaved calonysterone derivative 2 and fully characterized by comprehensive NMR techniques revealing their complete 1H and 13C signal assignments. Surprisingly, several compounds sensitized hCMEC/D3 brain microvascular endothelial cells to tert-butyl hydroperoxide (tBHP)-induced oxidative damage as recorded by impedance measurements. Compound 8, containing a benzyloxime ether moiety in its sidechain, was the only one that exerted a protective effect at a higher, 10 μM concentration, while at lower (10 nM- 1 μM) concentrations it promoted tBHP-induced cellular damage. Brain endothelial cells were protected from tBHP-induced barrier integrity decrease by treatment with 10 μM of compound 8, which also mitigated the intracellular reactive oxygen species production elevated by tBHP. Based on our results, 17-oxime ethers of oxidized ecdysteroids modulate oxidative stress of the BBB in a way that may point towards unexpected toxicity. Further studies are needed to evaluate any possible risk connected to dietary ecdysteroid consumption and CNS pathologies in which BBB damage plays an important role.
Collapse
Affiliation(s)
- Máté Vágvölgyi
- Institute of Pharmacognosy, University of Szeged, Szeged, Hungary
| | - Dávid Laczkó
- Institute of Pharmacognosy, University of Szeged, Szeged, Hungary
| | - Ana Raquel Santa-Maria
- Institute of Biophysics, HUN-REN Biological Research Centre, Szeged, Hungary
- Wyss Institute for Biologically Inspired Engineering at Harvard University, Boston, MA, United States of America
| | - Judit P. Vigh
- Institute of Biophysics, HUN-REN Biological Research Centre, Szeged, Hungary
- Doctoral School of Biology, University of Szeged, Szeged, Hungary
| | - Fruzsina R. Walter
- Institute of Biophysics, HUN-REN Biological Research Centre, Szeged, Hungary
| | - Róbert Berkecz
- Institute of Pharmaceutical Analysis, University of Szeged, Szeged, Hungary
| | - Mária A. Deli
- Institute of Biophysics, HUN-REN Biological Research Centre, Szeged, Hungary
| | - Gábor Tóth
- NMR Group, Department of Inorganic and Analytical Chemistry, Budapest University of Technology and Economics, Budapest, Hungary
| | - Attila Hunyadi
- Institute of Pharmacognosy, University of Szeged, Szeged, Hungary
- Interdisciplinary Centre of Natural Products, University of Szeged, Szeged, Hungary
- HUN-REN-SZTE Biologically Active Natural Products Research Group, Szeged, Hungary
| |
Collapse
|
15
|
Mehta P, Soliman A, Rodriguez-Vera L, Schmidt S, Muniz P, Rodriguez M, Forcadell M, Gonzalez-Perez E, Vozmediano V. Interspecies Brain PBPK Modeling Platform to Predict Passive Transport through the Blood-Brain Barrier and Assess Target Site Disposition. Pharmaceutics 2024; 16:226. [PMID: 38399280 PMCID: PMC10892872 DOI: 10.3390/pharmaceutics16020226] [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: 12/16/2023] [Revised: 01/30/2024] [Accepted: 02/01/2024] [Indexed: 02/25/2024] Open
Abstract
The high failure rate of central nervous system (CNS) drugs is partly associated with an insufficient understanding of target site exposure. Blood-brain barrier (BBB) permeability evaluation tools are needed to explore drugs' ability to access the CNS. An outstanding aspect of physiologically based pharmacokinetic (PBPK) models is the integration of knowledge on drug-specific and system-specific characteristics, allowing the identification of the relevant factors involved in target site distribution. We aimed to qualify a PBPK platform model to be used as a tool to predict CNS concentrations when significant transporter activity is absent and human data are sparse or unavailable. Data from the literature on the plasma and CNS of rats and humans regarding acetaminophen, oxycodone, lacosamide, ibuprofen, and levetiracetam were collected. Human BBB permeability values were extrapolated from rats using inter-species differences in BBB surface area. The percentage of predicted AUC and Cmax within the 1.25-fold criterion was 85% and 100% for rats and humans, respectively, with an overall GMFE of <1.25 in all cases. This work demonstrated the successful application of the PBPK platform for predicting human CNS concentrations of drugs passively crossing the BBB. Future applications include the selection of promising CNS drug candidates and the evaluation of new posologies for existing drugs.
Collapse
Affiliation(s)
- Parsshava Mehta
- Center for Pharmacometrics and Systems Pharmacology, Department of Pharmaceutics, College of Pharmacy, University of Florida, Orlando, FL 32827, USA; (P.M.); (A.S.); (S.S.)
| | - Amira Soliman
- Center for Pharmacometrics and Systems Pharmacology, Department of Pharmaceutics, College of Pharmacy, University of Florida, Orlando, FL 32827, USA; (P.M.); (A.S.); (S.S.)
- Department of Pharmacy Practice, Faculty of Pharmacy, Helwan University, Helwan 11795, Egypt
| | - Leyanis Rodriguez-Vera
- Model Informed Development, CTI Laboratories, Covington, KY 41011, USA; (L.R.-V.); (P.M.); (M.R.)
| | - Stephan Schmidt
- Center for Pharmacometrics and Systems Pharmacology, Department of Pharmaceutics, College of Pharmacy, University of Florida, Orlando, FL 32827, USA; (P.M.); (A.S.); (S.S.)
| | - Paula Muniz
- Model Informed Development, CTI Laboratories, Covington, KY 41011, USA; (L.R.-V.); (P.M.); (M.R.)
| | - Monica Rodriguez
- Model Informed Development, CTI Laboratories, Covington, KY 41011, USA; (L.R.-V.); (P.M.); (M.R.)
| | - Marta Forcadell
- Neuraxpharm Pharmaceuticals SL, Clinical Research and Evidence-Generation Science, 08970 Barcelona, Spain; (M.F.); (E.G.-P.)
| | - Emili Gonzalez-Perez
- Neuraxpharm Pharmaceuticals SL, Clinical Research and Evidence-Generation Science, 08970 Barcelona, Spain; (M.F.); (E.G.-P.)
| | - Valvanera Vozmediano
- Center for Pharmacometrics and Systems Pharmacology, Department of Pharmaceutics, College of Pharmacy, University of Florida, Orlando, FL 32827, USA; (P.M.); (A.S.); (S.S.)
- Model Informed Development, CTI Laboratories, Covington, KY 41011, USA; (L.R.-V.); (P.M.); (M.R.)
| |
Collapse
|
16
|
Badawi AH, Mohamad NA, Stanslas J, Kirby BP, Neela VK, Ramasamy R, Basri H. In Vitro Blood-Brain Barrier Models for Neuroinfectious Diseases: A Narrative Review. Curr Neuropharmacol 2024; 22:1344-1373. [PMID: 38073104 PMCID: PMC11092920 DOI: 10.2174/1570159x22666231207114346] [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: 09/12/2022] [Revised: 11/04/2022] [Accepted: 11/25/2022] [Indexed: 05/16/2024] Open
Abstract
The blood-brain barrier (BBB) is a complex, dynamic, and adaptable barrier between the peripheral blood system and the central nervous system. While this barrier protects the brain and spinal cord from inflammation and infection, it prevents most drugs from reaching the brain tissue. With the expanding interest in the pathophysiology of BBB, the development of in vitro BBB models has dramatically evolved. However, due to the lack of a standard model, a range of experimental protocols, BBB-phenotype markers, and permeability flux markers was utilized to construct in vitro BBB models. Several neuroinfectious diseases are associated with BBB dysfunction. To conduct neuroinfectious disease research effectively, there stems a need to design representative in vitro human BBB models that mimic the BBB's functional and molecular properties. The highest necessity is for an in vitro standardised BBB model that accurately represents all the complexities of an intact brain barrier. Thus, this in-depth review aims to describe the optimization and validation parameters for building BBB models and to discuss previous research on neuroinfectious diseases that have utilized in vitro BBB models. The findings in this review may serve as a basis for more efficient optimisation, validation, and maintenance of a structurally- and functionally intact BBB model, particularly for future studies on neuroinfectious diseases.
Collapse
Affiliation(s)
- Ahmad Hussein Badawi
- Department of Neurology, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, 43400, Serdang, Selangor, Malaysia
| | - Nur Afiqah Mohamad
- Department of Neurology, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, 43400, Serdang, Selangor, Malaysia
- Centre for Foundation Studies, Lincoln University College, 47301, Petaling Jaya, Selangor, Malaysia
| | - Johnson Stanslas
- Department of Medicine, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, 43400, Serdang, Selangor, Malaysia
| | - Brian Patrick Kirby
- School of Pharmacy and Biomolecular Sciences, RCSI University of Medicine and Health Sciences, Dublin, Ireland
| | - Vasantha Kumari Neela
- Department of Medical Microbiology and Parasitology, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, 43400, Serdang, Selangor, Malaysia
| | - Rajesh Ramasamy
- Department of Pathology, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, 43400, Serdang, Selangor, Malaysia
| | - Hamidon Basri
- Department of Neurology, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, 43400, Serdang, Selangor, Malaysia
| |
Collapse
|
17
|
Ronaldson PT, Davis TP. Blood-brain barrier transporters: a translational consideration for CNS delivery of neurotherapeutics. Expert Opin Drug Deliv 2024; 21:71-89. [PMID: 38217410 PMCID: PMC10842757 DOI: 10.1080/17425247.2024.2306138] [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: 10/02/2023] [Accepted: 01/12/2024] [Indexed: 01/15/2024]
Abstract
INTRODUCTION Successful neuropharmacology requires optimization of CNS drug delivery and, by extension, free drug concentrations at brain molecular targets. Detailed assessment of blood-brain barrier (BBB) physiological characteristics is necessary to achieve this goal. The 'next frontier' in CNS drug delivery is targeting BBB uptake transporters, an approach that requires evaluation of brain endothelial cell transport processes so that effective drug accumulation and improved therapeutic efficacy can occur. AREAS COVERED BBB permeability of drugs is governed by tight junction protein complexes (i.e., physical barrier) and transporters/enzymes (i.e., biochemical barrier). For most therapeutics, a component of blood-to-brain transport involves passive transcellular diffusion. Small molecule drugs that do not possess acceptable physicochemical characteristics for passive permeability may utilize putative membrane transporters for CNS uptake. While both uptake and efflux transport mechanisms are expressed at the brain microvascular endothelium, uptake transporters can be targeted for optimization of brain drug delivery and improved treatment of neurological disease states. EXPERT OPINION Uptake transporters represent a unique opportunity to optimize brain drug delivery by leveraging the endogenous biology of the BBB. A rigorous understanding of these transporters is required to improve translation from the bench to clinical trials and stimulate the development of new treatment paradigms for neurological diseases.
Collapse
Affiliation(s)
| | - Thomas P. Davis
- Department of Pharmacology, University of Arizona College of Medicine
| |
Collapse
|
18
|
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: 0] [Impact Index Per Article: 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
|
19
|
Ahmadighadykolaei H, Lambert JA, Raeeszadeh-Sarmazdeh M. TIMP-1 Protects Tight Junctions of Brain Endothelial Cells From MMP-Mediated Degradation. Pharm Res 2023; 40:2121-2131. [PMID: 37700105 PMCID: PMC10878538 DOI: 10.1007/s11095-023-03593-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Accepted: 08/18/2023] [Indexed: 09/14/2023]
Abstract
OBJECTIVE The blood-brain barrier (BBB) plays a critical role in central nervous system homeostasis, and the integrity of BBB is disrupted in many neurodegenerative diseases. Matrix metalloproteinases (MMPs) degrade the tight junctions (TJs) of endothelial cells and basement membrane components essential to BBB integrity, which leads to increased BBB permeability and allows inflammatory cells and neurotoxic substances to enter the brain. Tissue inhibitors of metalloproteinases (TIMPs), endogenous inhibitors of MMPs, regulate MMP activity, thereby maintaining BBB integrity. METHODS The disruptive impacts of MMP-3 and MMP-9 on BBB and protective effect of TIMP-1 were investigated in a simplified in vitro model of the BBB, which was generated using rat brain microvascular endothelial cells (RBMEC). The main features of BBB formation, including permeability and the trans-endothelial electrical resistance (TEER), were monitored over time after the addition of MMP-3 and MMP-9 and their complexes with TIMP-1 inhibitor. RESULTS Our results indicated that MMP-3 and MMP-9 caused a dose-dependent disruption of the BBB, with 1.5 µM MMPs resulting in an over threefold increase in permeability, while TIMP-1 inhibition protected the integrity of the BBB model and recovered TEER and permeability of RBMECs. The disruption and recovery of tight junction proteins of RBMECs after MMP and TIMP treatment were also detected using fluorescent microscopy. CONCLUSION MMP-9 and MMP-3 disrupt the BBB by degrading tight junctions in endothelial cells, and TIMP-1 could inhibit the disruptive effect of MMP-3 and MMP-9 by showing potential as therapeutic protein against MMP-related diseases where BBB disruption plays a role.
Collapse
Affiliation(s)
- Hannaneh Ahmadighadykolaei
- Department of Chemical and Materials Engineering, University of Nevada, 1664 N. Virginia St, Reno, NV, 89557, USA
| | - Janet A Lambert
- Department of Chemical and Materials Engineering, University of Nevada, 1664 N. Virginia St, Reno, NV, 89557, USA
- Department of Pharmacology, University of Nevada, Reno School of Medicine, Reno, NV, USA
| | - Maryam Raeeszadeh-Sarmazdeh
- Department of Chemical and Materials Engineering, University of Nevada, 1664 N. Virginia St, Reno, NV, 89557, USA.
| |
Collapse
|
20
|
Sánchez-Dengra B, García-Montoya E, González-Álvarez I, Bermejo M, González-Álvarez M. Establishment and Validation of a New Co-Culture for the Evaluation of the Permeability through the Blood-Brain Barrier in Patients with Glioblastoma. Pharmaceutics 2023; 15:pharmaceutics15051431. [PMID: 37242673 DOI: 10.3390/pharmaceutics15051431] [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: 03/02/2023] [Revised: 04/18/2023] [Accepted: 04/30/2023] [Indexed: 05/28/2023] Open
Abstract
Currently, the mechanisms involved in drug access to the central nervous system (CNS) are not completely elucidated, and research efforts to understand the behaviour of the therapeutic agents to access the blood-brain barrier continue with the utmost importance. The aim of this work was the creation and validation of a new in vitro model capable of predicting the in vivo permeability across the blood-brain barrier in the presence of glioblastoma. The selected in vitro method was a cell co-culture model of epithelial cell lines (MDCK and MDCK-MDR1) with a glioblastoma cell line (U87-MG). Several drugs were tested (letrozole, gemcitabine, methotrexate and ganciclovir). Comparison of the proposed in vitro model, MDCK and MDCK-MDR1 co-cultured with U87-MG, and in vivo studies showed a great predictability for each cell line, with R2 values of 0.8917 and 0.8296, respectively. Therefore, both cells lines (MDCK and MDCK-MDR1) are valid for predicting the access of drugs to the CNS in the presence of glioblastoma.
Collapse
Affiliation(s)
- Bárbara Sánchez-Dengra
- Engineering: Pharmacokinetics and Pharmaceutical Technology Area, Miguel Hernandez University, San Juan de Alicante, 03550 Alicante, Spain
| | - Elena García-Montoya
- Engineering: Pharmacokinetics and Pharmaceutical Technology Area, Miguel Hernandez University, San Juan de Alicante, 03550 Alicante, Spain
| | - Isabel González-Álvarez
- Engineering: Pharmacokinetics and Pharmaceutical Technology Area, Miguel Hernandez University, San Juan de Alicante, 03550 Alicante, Spain
| | - Marival Bermejo
- Engineering: Pharmacokinetics and Pharmaceutical Technology Area, Miguel Hernandez University, San Juan de Alicante, 03550 Alicante, Spain
| | - Marta González-Álvarez
- Engineering: Pharmacokinetics and Pharmaceutical Technology Area, Miguel Hernandez University, San Juan de Alicante, 03550 Alicante, Spain
| |
Collapse
|
21
|
Tóth G, Santa-Maria AR, Herke I, Gáti T, Galvis-Montes D, Walter FR, Deli MA, Hunyadi A. Highly Oxidized Ecdysteroids from a Commercial Cyanotis arachnoidea Root Extract as Potent Blood-Brain Barrier Protective Agents. JOURNAL OF NATURAL PRODUCTS 2023; 86:1074-1080. [PMID: 36825873 PMCID: PMC10152481 DOI: 10.1021/acs.jnatprod.2c00948] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Indexed: 05/04/2023]
Abstract
Ecdysteroid-containing herbal extracts, commonly prepared from the roots of Cyanotis arachnoidea, are marketed worldwide as a "green" anabolic food supplement. Herein are reported the isolation and complete 1H and 13C NMR signal assignments of three new minor ecdysteroids (compounds 2-4) from this extract. Compound 4 was identified as a possible artifact that gradually forms through the autoxidation of calonysterone. The compounds tested demonstrated a significant protective effect on the blood-brain barrier endothelial cells against oxidative stress or inflammation at a concentration of 1 μM. Based on these results, minor ecdysteroids present in food supplements may offer health benefits in various neurodegenerative disease states.
Collapse
Affiliation(s)
- Gábor Tóth
- Department
of Inorganic and Analytical Chemistry, NMR Group, Budapest University of Technology and Economics, H-1111 Budapest, Hungary
| | - Ana R. Santa-Maria
- Institute
of Biophysics, Biological Research Centre, Szeged H-6726, Hungary
- Wyss
Institute for Biologically Inspired Engineering at Harvard University, Boston, Massachusetts 02115, United States
| | - Ibolya Herke
- Department
of Inorganic and Analytical Chemistry, NMR Group, Budapest University of Technology and Economics, H-1111 Budapest, Hungary
| | - Tamás Gáti
- Servier
Research Institute of Medicinal Chemistry (SRIMC), H-1031 Budapest, Hungary
| | | | - Fruzsina R. Walter
- Institute
of Biophysics, Biological Research Centre, Szeged H-6726, Hungary
| | - Mária A. Deli
- Institute
of Biophysics, Biological Research Centre, Szeged H-6726, Hungary
| | - Attila Hunyadi
- Institute of Pharmacognosy, and Interdisciplinary
Centre of Natural Products, University of
Szeged, H-6720 Szeged, Hungary
| |
Collapse
|
22
|
Wei W, Cardes F, Hierlemann A, Modena MM. 3D In Vitro Blood-Brain-Barrier Model for Investigating Barrier Insults. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2205752. [PMID: 36782313 PMCID: PMC10104638 DOI: 10.1002/advs.202205752] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Revised: 01/18/2023] [Indexed: 06/18/2023]
Abstract
Blood-brain-barrier (BBB) disruption has been associated with a variety of central-nervous-system diseases. In vitro BBB models enable to investigate how the barrier reacts to external injury events, commonly referred to as insults. Here, a human-cell-based BBB platform with integrated, transparent electrodes to monitor barrier tightness in real time at high resolution is presented. The BBB model includes human cerebral endothelial cells and primary pericytes and astrocytes in a 3D arrangement within a pump-free, open-microfluidic platform. With this platform, this study demonstrates that oxygen-glucose deprivation (OGD), which mimics the characteristics of an ischemic insult, induces a rapid remodeling of the cellular actin structures and subsequent morphological changes in the endothelial cells. High-resolution live imaging shows the formation of large actin stress-fiber bundles in the endothelial layer during OGD application, which ultimately leads to cell shrinkage and barrier breakage. Simultaneous electrical measurements evidence a rapid decrease of the barrier electrical resistance before the appearance of stress fibers, which indicates that the barrier function is compromised already before the appearance of drastic morphological changes. The results demonstrate that the BBB platform recapitulates the main barrier functions in vitro and can be used to investigate rapid reorganization of the BBB upon application of external stimuli.
Collapse
Affiliation(s)
- Wei Wei
- ETH ZürichDepartment of Biosystems Science and EngineeringBio Engineering LaboratoryBasel4058Switzerland
| | - Fernando Cardes
- ETH ZürichDepartment of Biosystems Science and EngineeringBio Engineering LaboratoryBasel4058Switzerland
| | - Andreas Hierlemann
- ETH ZürichDepartment of Biosystems Science and EngineeringBio Engineering LaboratoryBasel4058Switzerland
| | - Mario M. Modena
- ETH ZürichDepartment of Biosystems Science and EngineeringBio Engineering LaboratoryBasel4058Switzerland
| |
Collapse
|
23
|
Kincses A, Vigh JP, Petrovszki D, Valkai S, Kocsis AE, Walter FR, Lin HY, Jan JS, Deli MA, Dér A. The Use of Sensors in Blood-Brain Barrier-on-a-Chip Devices: Current Practice and Future Directions. BIOSENSORS 2023; 13:bios13030357. [PMID: 36979569 PMCID: PMC10046513 DOI: 10.3390/bios13030357] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Revised: 02/27/2023] [Accepted: 03/02/2023] [Indexed: 06/01/2023]
Abstract
The application of lab-on-a-chip technologies in in vitro cell culturing swiftly resulted in improved models of human organs compared to static culture insert-based ones. These chip devices provide controlled cell culture environments to mimic physiological functions and properties. Models of the blood-brain barrier (BBB) especially profited from this advanced technological approach. The BBB represents the tightest endothelial barrier within the vasculature with high electric resistance and low passive permeability, providing a controlled interface between the circulation and the brain. The multi-cell type dynamic BBB-on-chip models are in demand in several fields as alternatives to expensive animal studies or static culture inserts methods. Their combination with integrated biosensors provides real-time and noninvasive monitoring of the integrity of the BBB and of the presence and concentration of agents contributing to the physiological and metabolic functions and pathologies. In this review, we describe built-in sensors to characterize BBB models via quasi-direct current and electrical impedance measurements, as well as the different types of biosensors for the detection of metabolites, drugs, or toxic agents. We also give an outlook on the future of the field, with potential combinations of existing methods and possible improvements of current techniques.
Collapse
Affiliation(s)
- András Kincses
- Institute of Biophysics, Biological Research Centre, H-6726 Szeged, Hungary; (A.K.); (J.P.V.); (D.P.); (S.V.); (A.E.K.); (F.R.W.)
| | - Judit P. Vigh
- Institute of Biophysics, Biological Research Centre, H-6726 Szeged, Hungary; (A.K.); (J.P.V.); (D.P.); (S.V.); (A.E.K.); (F.R.W.)
- Doctoral School of Biology, University of Szeged, H-6720 Szeged, Hungary
| | - Dániel Petrovszki
- Institute of Biophysics, Biological Research Centre, H-6726 Szeged, Hungary; (A.K.); (J.P.V.); (D.P.); (S.V.); (A.E.K.); (F.R.W.)
- Doctoral School of Multidisciplinary Medical Sciences, University of Szeged, H-6720 Szeged, Hungary
| | - Sándor Valkai
- Institute of Biophysics, Biological Research Centre, H-6726 Szeged, Hungary; (A.K.); (J.P.V.); (D.P.); (S.V.); (A.E.K.); (F.R.W.)
| | - Anna E. Kocsis
- Institute of Biophysics, Biological Research Centre, H-6726 Szeged, Hungary; (A.K.); (J.P.V.); (D.P.); (S.V.); (A.E.K.); (F.R.W.)
| | - Fruzsina R. Walter
- Institute of Biophysics, Biological Research Centre, H-6726 Szeged, Hungary; (A.K.); (J.P.V.); (D.P.); (S.V.); (A.E.K.); (F.R.W.)
| | - Hung-Yin Lin
- Department of Chemical and Materials Engineering, National University of Kaohsiung, Kaohsiung 81148, Taiwan;
| | - Jeng-Shiung Jan
- Department of Chemical Engineering, National Cheng Kung University, Tainan 70101, Taiwan;
| | - Mária A. Deli
- Institute of Biophysics, Biological Research Centre, H-6726 Szeged, Hungary; (A.K.); (J.P.V.); (D.P.); (S.V.); (A.E.K.); (F.R.W.)
| | - András Dér
- Institute of Biophysics, Biological Research Centre, H-6726 Szeged, Hungary; (A.K.); (J.P.V.); (D.P.); (S.V.); (A.E.K.); (F.R.W.)
| |
Collapse
|
24
|
Medina-Flores F, Hurtado-Alvarado G, Deli MA, Gómez-González B. The Active Role of Pericytes During Neuroinflammation in the Adult Brain. Cell Mol Neurobiol 2023; 43:525-541. [PMID: 35195811 PMCID: PMC11415175 DOI: 10.1007/s10571-022-01208-5] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Accepted: 02/13/2022] [Indexed: 12/11/2022]
Abstract
Microvessels in the central nervous system (CNS) have one of the highest populations of pericytes, indicating their crucial role in maintaining homeostasis. Pericytes are heterogeneous cells located around brain microvessels; they present three different morphologies along the CNS vascular tree: ensheathing, mesh, and thin-strand pericytes. At the arteriole-capillary transition ensheathing pericytes are found, while mesh and thin-strand pericytes are located at capillary beds. Brain pericytes are essential for the establishment and maintenance of the blood-brain barrier, which restricts the passage of soluble and potentially toxic molecules from the circulatory system to the brain parenchyma. Pericytes play a key role in regulating local inflammation at the CNS. Pericytes can respond differentially, depending on the degree of inflammation, by secreting a set of neurotrophic factors to promote cell survival and regeneration, or by potentiating inflammation through the release of inflammatory mediators (e.g., cytokines and chemokines), and the overexpression of cell adhesion molecules. Under inflammatory conditions, pericytes may regulate immune cell trafficking to the CNS and play a role in perpetuating local inflammation. In this review, we describe pericyte responses during acute and chronic neuroinflammation.
Collapse
Affiliation(s)
- Fernanda Medina-Flores
- Area of Neurosciences, Department Biology of Reproduction, CBS, Universidad Autónoma Metropolitana, Unidad Iztapalapa, Av. San Rafael Atlixco No. 186, Col. Vicentina, Deleg. Iztapalapa, 09340, Mexico City, Mexico
- Posgrado en Biología Experimental, Universidad Autónoma Metropolitana, Unidad Iztapalapa, Mexico City, Mexico
| | - Gabriela Hurtado-Alvarado
- Departamento de Biología Celular Y Fisiología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Maria A Deli
- Institute of Biophysics, Biological Research Centre, Szeged, Hungary
| | - Beatriz Gómez-González
- Area of Neurosciences, Department Biology of Reproduction, CBS, Universidad Autónoma Metropolitana, Unidad Iztapalapa, Av. San Rafael Atlixco No. 186, Col. Vicentina, Deleg. Iztapalapa, 09340, Mexico City, Mexico.
| |
Collapse
|
25
|
Targeting Human Endothelial Cells with Glutathione and Alanine Increases the Crossing of a Polypeptide Nanocarrier through a Blood-Brain Barrier Model and Entry to Human Brain Organoids. Cells 2023; 12:cells12030503. [PMID: 36766845 PMCID: PMC9914642 DOI: 10.3390/cells12030503] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2022] [Revised: 01/31/2023] [Accepted: 02/01/2023] [Indexed: 02/05/2023] Open
Abstract
Nanoparticles (NPs) are the focus of research efforts that aim to develop successful drug delivery systems for the brain. Polypeptide nanocarriers are versatile platforms and combine high functionality with good biocompatibility and biodegradability. The key to the efficient brain delivery of NPs is the specific targeting of cerebral endothelial cells that form the blood-brain barrier (BBB). We have previously discovered that the combination of two different ligands of BBB nutrient transporters, alanine and glutathione, increases the permeability of vesicular NPs across the BBB. Our aim here was to investigate whether the combination of these molecules can also promote the efficient transfer of 3-armed poly(l-glutamic acid) NPs across a human endothelial cell and brain pericyte BBB co-culture model. Alanine and glutathione dual-targeted polypeptide NPs showed good cytocompatibility and elevated cellular uptake in a time-dependent and active manner. Targeted NPs had a higher permeability across the BBB model and could subsequently enter midbrain-like organoids derived from healthy and Parkinson's disease patient-specific stem cells. These results indicate that poly(l-glutamic acid) NPs can be used as nanocarriers for nervous system application and that the right combination of molecules that target cerebral endothelial cells, in this case alanine and glutathione, can facilitate drug delivery to the brain.
Collapse
|
26
|
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: 3] [Impact Index Per Article: 1.5] [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
|
27
|
Harding IC, O'Hare NR, Vigliotti M, Caraballo A, Lee CI, Millican K, Herman IM, Ebong EE. Developing a transwell millifluidic device for studying blood-brain barrier endothelium. LAB ON A CHIP 2022; 22:4603-4620. [PMID: 36326069 PMCID: PMC11416711 DOI: 10.1039/d2lc00657j] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
Blood-brain barrier (BBB) endothelial cell (EC) function depends on flow conditions and on supportive cells, like pericytes and astrocytes, which have been shown to be both beneficial and detrimental for brain EC function. Most studies investigating BBB EC function lack physiological relevance, using sub-physiological shear stress magnitudes and/or omitting pericytes and astrocytes. In this study, we developed a millifluidic device compatible with standard transwell inserts to investigate BBB function. In contrast to standard polydimethylsiloxane (PDMS) microfluidic devices, this model allows for easy, reproducible shear stress exposure without common limitations of PDMS devices such as inadequate nutrient diffusion and air bubble formation. In no-flow conditions, we first used the device to examine the impact of primary human pericytes and astrocytes on human brain microvascular EC (HBMEC) barrier integrity. Astrocytes, pericytes, and a 1-to-1 ratio of both cell types increased HBMEC barrier integrity via reduced 3 and 40 kDa fluorescent dextran permeability and increased claudin-5 expression. There were differing levels of low 3 kDa permeability in HBMEC-pericyte, HBMEC-astrocyte, and HBMEC-astrocyte-pericyte co-cultures, while levels of low 40 kDa permeability were consistent across co-cultures. The 3 kDa findings suggest that pericytes provide more barrier support to the BBB model compared to astrocytes, although both supportive cell types are permeability reducers. Incorporation of 24-hour 12 dynes per cm2 flow significantly reduced dextran permeability in HBMEC monolayers, but not in the tri-culture model. These results indicate that tri-culture may exert more pronounced impact on overall BBB permeability than flow exposure. In both cases, monolayer and tri-culture, flow exposure interestingly reduced HBMEC expression of both claudin-5 and occludin. ZO-1 expression, and localization at cell-cell junctions increased in the tri-culture but exhibited no apparent change in the HBMEC monolayer. Under flow conditions, we also observed HBMEC alignment in the tri-culture but not in HBMEC monolayers, indicating supportive cells and flow are both essential to observe brain EC alignment in vitro. Collectively, these results support the necessity of physiologically relevant, multicellular BBB models when investigating BBB EC function. Consideration of the roles of shear stress and supportive cells within the BBB is critical for elucidating the physiology of the neurovascular unit.
Collapse
Affiliation(s)
- Ian C Harding
- Department of Bioengineering, Northeastern University, Boston, MA, USA
| | - Nicholas R O'Hare
- Department of Chemical Engineering, Northeastern University, 360 Huntington Avenue, 129 Interdisciplinary Science and Engineering Complex, Boston, MA, 02115, USA.
| | - Mark Vigliotti
- Department of Chemical Engineering, Northeastern University, 360 Huntington Avenue, 129 Interdisciplinary Science and Engineering Complex, Boston, MA, 02115, USA.
| | - Alex Caraballo
- Department of Chemical Engineering, Northeastern University, 360 Huntington Avenue, 129 Interdisciplinary Science and Engineering Complex, Boston, MA, 02115, USA.
| | - Claire I Lee
- Department of Bioengineering, Northeastern University, Boston, MA, USA
| | - Karina Millican
- Department of Bioengineering, Northeastern University, Boston, MA, USA
| | - Ira M Herman
- Department of Developmental, Molecular, and Chemical Biology, Tufts School of Graduate Biomedical Sciences, Boston, MA, USA
- Center for Innovations in Wound Healing Research, Tufts University School of Medicine, Boston, MA, USA
| | - Eno E Ebong
- Department of Bioengineering, Northeastern University, Boston, MA, USA
- Department of Chemical Engineering, Northeastern University, 360 Huntington Avenue, 129 Interdisciplinary Science and Engineering Complex, Boston, MA, 02115, USA.
- Department of Neuroscience, Albert Einstein College of Medicine, New York, NY, USA
| |
Collapse
|
28
|
Effects of Hydroxypropyl-Beta-Cyclodextrin on Cultured Brain Endothelial Cells. Molecules 2022; 27:molecules27227738. [PMID: 36431844 PMCID: PMC9694004 DOI: 10.3390/molecules27227738] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Revised: 11/05/2022] [Accepted: 11/07/2022] [Indexed: 11/12/2022] Open
Abstract
The application of 2-hydroxypropyl-beta-cyclodextrin (HPBCD) in the treatment of the rare cholesterol and lipid storage disorder Niemann-Pick disease type C opened new perspectives in the development of an efficient therapy. Even if the systemic administration of HPBCD was found to be effective, its low permeability across the blood-brain barrier (BBB) limited the positive neurological effects. Nevertheless, the cellular interactions of HPBCD with brain capillary endothelial cells have not been investigated in detail. In this study, the cytotoxicity, permeability, and cellular internalization of HPBCD on primary rat and immortalized human (hCMEC/D3) brain capillary endothelial cells were investigated. HPBCD shows no cytotoxicity on endothelial cells up to 100 µM, measured by impedance kinetics. Using a fluorescent derivative of HPBCD (FITC-HPBCD) the permeability measurements reveal that on an in vitro triple co-culture BBB model, FITC-HPBCD has low permeability, 0.50 × 10-6 cm/s, while on hCMEC/D3 cell layers, the permeability is higher, 1.86 × 10-5 cm/s. FITC-HPBCD enters brain capillary endothelial cells, is detected in cytoplasmic vesicles and rarely localized in lysosomes. The cellular internalization of HPBCD at the BBB can help to develop new strategies for improved HPBCD effects after systemic administration.
Collapse
|
29
|
Lee MH, Jan JS, Thomas JL, Shih YP, Li JA, Lin CY, Ooya T, Barna L, Mészáros M, Harazin A, Porkoláb G, Veszelka S, Deli MA, Lin HY. Cellular Therapy Using Epitope-Imprinted Composite Nanoparticles to Remove α-Synuclein from an In Vitro Model. Cells 2022; 11:cells11162584. [PMID: 36010659 PMCID: PMC9406856 DOI: 10.3390/cells11162584] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Revised: 08/09/2022] [Accepted: 08/18/2022] [Indexed: 11/16/2022] Open
Abstract
Several degenerative disorders of the central nervous system, including Parkinson’s disease (PD), are related to the pathological aggregation of proteins. Antibodies against toxic disease proteins, such as α-synuclein (SNCA), are therefore being developed as possible therapeutics. In this work, one peptide (YVGSKTKEGVVHGVA) from SNCA was used as the epitope to construct magnetic molecularly imprinted composite nanoparticles (MMIPs). These composite nanoparticles were characterized by dynamic light scattering (DLS), high-performance liquid chromatography (HPLC), isothermal titration calorimetry (ITC), Brunauer–Emmett–Teller (BET) analysis, and superconducting quantum interference device (SQUID) analysis. Finally, the viability of brain endothelial cells that were treated with MMIPs was measured, and the extraction of SNCA from CRISPR/dCas9a-activated HEK293T cells from the in vitro model system was demonstrated for the therapeutic application of MMIPs.
Collapse
Affiliation(s)
- Mei-Hwa Lee
- Department of Materials Science and Engineering, I-Shou University, Kaohsiung 84001, Taiwan
| | - Jeng-Shiung Jan
- Department of Chemical Engineering, National Cheng Kung University, Tainan 70101, Taiwan
| | - James L. Thomas
- Department of Physics and Astronomy, University of New Mexico, Albuquerque, NM 87131, USA
| | - Yuan-Pin Shih
- Department of Chemical and Materials Engineering, National University of Kaohsiung, Kaohsiung 81148, Taiwan
| | - Jin-An Li
- Department of Chemical and Materials Engineering, National University of Kaohsiung, Kaohsiung 81148, Taiwan
| | - Chien-Yu Lin
- Department of Chemical and Materials Engineering, National University of Kaohsiung, Kaohsiung 81148, Taiwan
| | - Tooru Ooya
- Graduate School of Engineering, Department of Chemical Science and Engineering, Kobe University, Kobe 657-8501, Japan
- Center for Advanced Medical Engineering Research & Development (CAMED), Kobe University, Kobe 657-8501, Japan
| | - Lilla Barna
- Institute of Biophysics, Biological Research Centre, H-6726 Szeged, Hungary
- Doctoral School in Biology, University of Szeged, H-6720 Szeged, Hungary
| | - Mária Mészáros
- Institute of Biophysics, Biological Research Centre, H-6726 Szeged, Hungary
| | - András Harazin
- Institute of Biophysics, Biological Research Centre, H-6726 Szeged, Hungary
| | - Gergő Porkoláb
- Institute of Biophysics, Biological Research Centre, H-6726 Szeged, Hungary
- Doctoral School in Biology, University of Szeged, H-6720 Szeged, Hungary
| | - Szilvia Veszelka
- Institute of Biophysics, Biological Research Centre, H-6726 Szeged, Hungary
| | - Maria A. Deli
- Institute of Biophysics, Biological Research Centre, H-6726 Szeged, Hungary
- Correspondence: (M.A.D.); (H.-Y.L.)
| | - Hung-Yin Lin
- Department of Chemical and Materials Engineering, National University of Kaohsiung, Kaohsiung 81148, Taiwan
- Correspondence: (M.A.D.); (H.-Y.L.)
| |
Collapse
|
30
|
Branca JJV, Carrino D, Paternostro F, Morucci G, Fiorillo C, Nicoletti C, Gulisano M, Ghelardini C, Di Cesare Mannelli L, Becatti M, Pacini A. The Protection of Zinc against Acute Cadmium Exposure: A Morphological and Molecular Study on a BBB In Vitro Model. Cells 2022; 11:cells11101646. [PMID: 35626683 PMCID: PMC9140137 DOI: 10.3390/cells11101646] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Revised: 05/09/2022] [Accepted: 05/13/2022] [Indexed: 02/04/2023] Open
Abstract
Cadmium (Cd) is a well-known occupational and environmental pollutant worldwide, and its toxicity is widely recognised. Cd is reported to increase the permeability of the blood–brain barrier (BBB) and to penetrate and accumulate in the brain. Although many lines of evidence show that Cd toxicity is induced by different mechanisms, one of the best known is the Cd-dependent production of reactive oxygen species (ROS). Zinc is a trace element known as coenzyme and cofactor for many antioxidant proteins, such as metallothioneins and superoxide dismutase enzymes. To date, very little is known about the role of Zn in preventing Cd-induced blood–brain barrier (BBB) alterations. The goal of this study was to test the Zn antioxidant capacity against Cd-dependent alterations in a rat brain endothelial cell line (RBE4), as an in vitro model for BBB. In order to mimic acute Cd poisoning, RBE4 cells were treated with CdCl2 30 µM for 24 h. The protective role of ZnCl2 (50 µM) was revealed by evaluating the cell viability, reactive oxygen species (ROS) quantification, cytochrome C distribution, and the superoxide dismutase (SOD) protein activity. Additionally, the effectiveness of Zn in counteracting the Cd-induced damage was investigated by evaluating the expression levels of proteins already known to be involved in the Cd signalling pathway, such as GRP78 (an endoplasmic reticulum (ER) stress protein), caspase3 pro- and cleaved forms, and BAX. Finally, we evaluated if Zn was able to attenuate the alterations of zonula occludens-1 (ZO-1), one of the tight-junction (TJ) proteins involved in the formation of the BBB. Our data clearly demonstrate that Zn, by protecting from the SOD activity impairment induced by Cd, is able to prevent the triggering of the Cd-dependent signalling pathway that leads to ZO-1 dislocation and downregulation, and BBB damage.
Collapse
Affiliation(s)
- Jacopo J. V. Branca
- Department of Experimental and Clinical Medicine, Histology and Anatomy Section, University of Firenze, 50134 Firenze, Italy; (J.J.V.B.); (D.C.); (F.P.); (C.N.); (M.G.)
| | - Donatello Carrino
- Department of Experimental and Clinical Medicine, Histology and Anatomy Section, University of Firenze, 50134 Firenze, Italy; (J.J.V.B.); (D.C.); (F.P.); (C.N.); (M.G.)
| | - Ferdinando Paternostro
- Department of Experimental and Clinical Medicine, Histology and Anatomy Section, University of Firenze, 50134 Firenze, Italy; (J.J.V.B.); (D.C.); (F.P.); (C.N.); (M.G.)
| | - Gabriele Morucci
- Department of Translational Research and New Technology in Medicine and Surgery, University of Pisa, 56126 Pisa, Italy;
| | - Claudia Fiorillo
- Department of Experimental and Clinical Biomedical Sciences ‘Mario Serio’, University of Firenze, 50134 Firenze, Italy; (C.F.); (M.B.)
| | - Claudio Nicoletti
- Department of Experimental and Clinical Medicine, Histology and Anatomy Section, University of Firenze, 50134 Firenze, Italy; (J.J.V.B.); (D.C.); (F.P.); (C.N.); (M.G.)
| | - Massimo Gulisano
- Department of Experimental and Clinical Medicine, Histology and Anatomy Section, University of Firenze, 50134 Firenze, Italy; (J.J.V.B.); (D.C.); (F.P.); (C.N.); (M.G.)
| | - Carla Ghelardini
- Department of Neuroscience, Psychology, Drug Research and Child Health (NEUROFARBA), Pharmacology and Toxicology Section, University of Firenze, 50139 Firenze, Italy; (C.G.); (L.D.C.M.)
| | - Lorenzo Di Cesare Mannelli
- Department of Neuroscience, Psychology, Drug Research and Child Health (NEUROFARBA), Pharmacology and Toxicology Section, University of Firenze, 50139 Firenze, Italy; (C.G.); (L.D.C.M.)
| | - Matteo Becatti
- Department of Experimental and Clinical Biomedical Sciences ‘Mario Serio’, University of Firenze, 50134 Firenze, Italy; (C.F.); (M.B.)
| | - Alessandra Pacini
- Department of Experimental and Clinical Medicine, Histology and Anatomy Section, University of Firenze, 50134 Firenze, Italy; (J.J.V.B.); (D.C.); (F.P.); (C.N.); (M.G.)
- Correspondence: ; Tel.: +39-055-2758067
| |
Collapse
|
31
|
Huber I, Pandur E, Sipos K, Barna L, Harazin A, Deli MA, Tyukodi L, Gulyás-Fekete G, Kulcsár G, Rozmer Z. Novel cyclic C 5-curcuminoids penetrating the blood-brain barrier: Design, synthesis and antiproliferative activity against astrocytoma and neuroblastoma cells. Eur J Pharm Sci 2022; 173:106184. [PMID: 35413433 DOI: 10.1016/j.ejps.2022.106184] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Revised: 03/26/2022] [Accepted: 04/04/2022] [Indexed: 11/25/2022]
Abstract
Novel series of cyclic C5-curcuminoids 17a-j and 19-22 were prepared as cytotoxic agents and evaluated against human neuroblastoma (SH-SY5Y) or human grade IV astrocytoma (CCF-STTG1) cell lines in low (∼0.1 nM - 10 nM) concentrations. Among the tested 21 derivatives, 16 displayed potent antiproliferative activity with IC50 values in the low nanomolar to picomolar range (IC50 = 7.483-0.139 nM). Highly active compounds like N-monocarboxylic derivative 19b with IC50 = 0.139 nM value against neuroblastoma and N-alkyl substituted 11 with IC50 = 0.257 nM against astrocytoma proved some degree of selectivity toward non-cancerous astrocytes and kidney cells. This potent anticancer activity did not show a strong correlation with experimental logPTLC values, but the most potent antiproliferative molecules 11-13 and 19-22 are belonging to discrete subgroups of the cyclic C5-curcuminoids. Compounds 12, 17c and 19b were subjected to blood-brain barrier (BBB) penetration studies, too. The BBB was revealed to be permeable for all of them but, as the apparent permeability coefficient (Papp) values mirrored, in different ratios. Lower toxicity of 12, 17c and 19b was observed toward primary rat brain endothelial cells of the BBB model, which means they remained undamaged under 10 µM concentrations. Penetration depends, at least in part, on albumin binding of 12, 17c and 19b and the presence of monocarboxylic acid transporters in the case of 19b. Permeation through the BBB and albumin binding, we described here, is the first example of cyclic C5-curcuminoids as to our knowledge.
Collapse
Affiliation(s)
- Imre Huber
- Department of Pharmaceutical Chemistry, University of Pécs, Pécs, Hungary.
| | - Edina Pandur
- Department of Pharmaceutical Biology, University of Pécs, Pécs, Hungary
| | - Katalin Sipos
- Department of Pharmaceutical Biology, University of Pécs, Pécs, Hungary
| | - Lilla Barna
- Biological Barriers Research Group, Institute of Biophysics, Biological Research Centre, Hungarian Academy of Sciences, Szeged, Hungary
| | - András Harazin
- Biological Barriers Research Group, Institute of Biophysics, Biological Research Centre, Hungarian Academy of Sciences, Szeged, Hungary
| | - Mária A Deli
- Biological Barriers Research Group, Institute of Biophysics, Biological Research Centre, Hungarian Academy of Sciences, Szeged, Hungary
| | - Levente Tyukodi
- Department of Pharmaceutical Chemistry, University of Pécs, Pécs, Hungary
| | | | - Győző Kulcsár
- Department of Pharmaceutical Chemistry, University of Pécs, Pécs, Hungary
| | - Zsuzsanna Rozmer
- Department of Pharmaceutical Chemistry, University of Pécs, Pécs, Hungary
| |
Collapse
|
32
|
de Lange ECM, Hammarlund Udenaes M. Understanding the Blood-Brain Barrier and Beyond: Challenges and Opportunities for Novel CNS Therapeutics. Clin Pharmacol Ther 2022; 111:758-773. [PMID: 35220577 PMCID: PMC9305478 DOI: 10.1002/cpt.2545] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Accepted: 01/27/2022] [Indexed: 11/11/2022]
Abstract
This review addresses questions on how to accomplish successful central nervous system (CNS) drug delivery (i.e., having the right concentration at the right CNS site, at the right time), by understanding the rate and extent of blood‐brain barrier (BBB) transport and intra‐CNS distribution in relation to CNS target site(s) exposure. To this end, we need to obtain and integrate quantitative and connected data on BBB using the Combinatory Mapping Approach that includes in vivo and ex vivo animal measurements, and the physiologically based comprehensive LEICNSPK3.0 mathematical model that can translate from animals to humans. For small molecules, slow diffusional BBB transport and active influx and efflux BBB transport determine the differences between plasma and CNS pharmacokinetics. Obviously, active efflux is important for limiting CNS drug delivery. Furthermore, liposomal formulations of small molecules may to a certain extent circumvent active influx and efflux at the BBB. Interestingly, for CNS pathologies, despite all reported disease associated BBB and CNS functional changes in animals and humans, integrative studies typically show a lack of changes on CNS drug delivery for the small molecules. In contrast, the understanding of the complex vesicle‐based BBB transport modes that are important for CNS delivery of large molecules is in progress, and their BBB transport seems to be significantly affected by CNS diseases. In conclusion, today, CNS drug delivery of small drugs can be well assessed and understood by integrative approaches, although there is still quite a long way to go to understand CNS drug delivery of large molecules.
Collapse
Affiliation(s)
- Elizabeth C M de Lange
- Predictive Pharmacology Group, Systems Pharmacology and Pharmacy, LACDR, Leiden University, Leiden, The Netherlands
| | | |
Collapse
|
33
|
Experimental Comparison of Primary and hiPS-Based In Vitro Blood–Brain Barrier Models for Pharmacological Research. Pharmaceutics 2022; 14:pharmaceutics14040737. [PMID: 35456571 PMCID: PMC9031459 DOI: 10.3390/pharmaceutics14040737] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Revised: 03/25/2022] [Accepted: 03/28/2022] [Indexed: 01/27/2023] Open
Abstract
In vitro model systems of the blood–brain barrier (BBB) play an essential role in pharmacological research, specifically during the development and preclinical evaluation of new drug candidates. Within the past decade, the trend in research and further development has moved away from models based on primary cells of animal origin towards differentiated models derived from human induced pluripotent stem cells (hiPSs). However, this logical progression towards human model systems from renewable cell sources opens up questions about the transferability of results generated in the primary cell models. In this study, we have evaluated both models with identical experimental parameters and achieved a directly comparable characterisation showing no significant differences in protein expression or permeability even though the achieved transendothelial electrical resistance (TEER) values showed significant differences. In the course of this investigation, we also determined a significant deviation of both model systems from the in vivo BBB circumstances, specifically concerning the presence or absence of serum proteins in the culture media. Thus, we have further evaluated both systems when confronted with an in vivo-like distribution of serum and found a notable improvement in the differential permeability of hydrophilic and lipophilic compounds in the hiPS-derived BBB model. We then transferred this model into a microfluidic setup while maintaining the differential serum distribution and evaluated the permeability coefficients, which showed good comparability with values in the literature. Therefore, we have developed a microfluidic hiPS-based BBB model with characteristics comparable to the established primary cell-based model.
Collapse
|
34
|
In Vitro-In Vivo Correlation of Blood-Brain Barrier Permeability of Drugs: A Feasibility Study Towards Development of Prediction Methods for Brain Drug Concentration in Humans. Pharm Res 2022; 39:1575-1586. [PMID: 35288803 DOI: 10.1007/s11095-022-03189-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Accepted: 02/03/2022] [Indexed: 10/18/2022]
Abstract
PURPOSE In vitro human blood-brain barrier (BBB) models in combination with central nervous system-physiologically based pharmacokinetic (CNS-PBPK) modeling, hereafter referred to as the "BBB/PBPK" method, are expected to contribute to prediction of brain drug concentration profiles in humans. As part of our ongoing effort to develop a BBB/PBPK method, we tried to clarify the relationship of in vivo BBB permeability data to those in vitro obtained from a human immortalized cell-based tri-culture BBB model (hiBBB), which we have recently created. METHODS The hiBBB models were developed and functionally characterized as previously described. The in vitro BBB permeabilities (Pe, × 10-6 cm/s) of seventeen compounds were determined by permeability assays, and in vivo BBB permeabilities (QECF) for eight drugs were estimated by CNS-PBPK modeling. The correlation of the Pe values with the QECF values was analyzed by linear regression analysis. RESULTS The hiBBB models showed intercellular barrier properties and several BBB transporter functions, which were enough to provide a wide dynamic range of Pe values from 5.7 ± 0.7 (rhodamine 123) to 2580.4 ± 781.9 (rivastigmine). Furthermore, the in vitro Pe values of the eight drugs showed a good correlation (R2 = 0.96) with their in vivo QECF values estimated from human clinical data. CONCLUSION We show that in vitro human BBB models provide clinically relevant BBB permeability that can be used as input for CNS-PBPK modeling. Therefore, our findings will encourage the development of a BBB/PBPK method as a promising approach for predicting brain drug concentration profiles in humans.
Collapse
|
35
|
Walter FR, Harazin A, Tóth AE, Veszelka S, Santa-Maria AR, Barna L, Kincses A, Biczó G, Balla Z, Kui B, Maléth J, Cervenak L, Tubak V, Kittel Á, Rakonczay Z, Deli MA. Blood-brain barrier dysfunction in L-ornithine induced acute pancreatitis in rats and the direct effect of L-ornithine on cultured brain endothelial cells. Fluids Barriers CNS 2022; 19:16. [PMID: 35177109 PMCID: PMC8851707 DOI: 10.1186/s12987-022-00308-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Accepted: 01/18/2022] [Indexed: 02/01/2023] Open
Abstract
BACKGROUND In severe acute pancreatitis (AP) the CNS is affected manifesting in neurological symptoms. Earlier research from our laboratory showed blood-brain barrier (BBB) permeability elevation in a taurocholate-induced AP model. Here we aimed to further explore BBB changes in AP using a different, non-invasive in vivo model induced by L-ornithine. Our goal was also to identify whether L-ornithine, a cationic amino acid, has a direct effect on brain endothelial cells in vitro contributing to the observed BBB changes. METHODS AP was induced in rats by the intraperitoneal administration of L-ornithine-HCl. Vessel permeability and the gene expression of the primary transporter of L-ornithine, cationic amino acid transporter-1 (Cat-1) in the brain cortex, pancreas, liver and lung were determined. Ultrastructural changes were followed by transmission electron microscopy. The direct effect of L-ornithine was tested on primary rat brain endothelial cells and a triple co-culture model of the BBB. Viability and barrier integrity, including permeability and TEER, nitrogen monoxide (NO) and reactive oxygen species (ROS) production and NF-κB translocation were measured. Fluorescent staining for claudin-5, occludin, ZO-1, β-catenin, cell adhesion molecules Icam-1 and Vcam-1 and mitochondria was performed. Cell surface charge was measured by laser Doppler velocimetry. RESULTS In the L-ornithine-induced AP model vessel permeability for fluorescein and Cat-1 expression levels were elevated in the brain cortex and pancreas. On the ultrastructural level surface glycocalyx and mitochondrial damage, tight junction and basal membrane alterations, and glial edema were observed. L-ornithine decreased cell impedance and elevated the BBB model permeability in vitro. Discontinuity in the surface glycocalyx labeling and immunostaining of junctional proteins, cytoplasmic redistribution of ZO-1 and β-catenin, and elevation of Vcam-1 expression were measured. ROS production was increased and mitochondrial network was damaged without NF-κB, NO production or mitochondrial membrane potential alterations. Similar ultrastructural changes were seen in L-ornithine treated brain endothelial cells as in vivo. The basal negative zeta potential of brain endothelial cells became more positive after L-ornithine treatment. CONCLUSION We demonstrated BBB damage in the L-ornithine-induced rat AP model suggesting a general, AP model independent effect. L-ornithine induced oxidative stress, decreased barrier integrity and altered BBB morphology in a culture BBB model. These data suggest a direct effect of the cationic L-ornithine on brain endothelium. Endothelial surface glycocalyx injury was revealed both in vivo and in vitro, as an additional novel component of the BBB-related pathological changes in AP.
Collapse
Affiliation(s)
- Fruzsina R Walter
- Institute of Biophysics, Biological Research Centre, Temesvári krt. 62, Szeged, 6726, Hungary
| | - András Harazin
- Institute of Biophysics, Biological Research Centre, Temesvári krt. 62, Szeged, 6726, Hungary
- Department of Biomedicine, Faculty of Health, Aarhus University, Høegh-Guldbergs Gade 10, 8000, Aarhus C, Denmark
| | - Andrea E Tóth
- Institute of Biophysics, Biological Research Centre, Temesvári krt. 62, Szeged, 6726, Hungary
- Department of Biomedicine, Faculty of Health, Aarhus University, Høegh-Guldbergs Gade 10, 8000, Aarhus C, Denmark
| | - Szilvia Veszelka
- Institute of Biophysics, Biological Research Centre, Temesvári krt. 62, Szeged, 6726, Hungary
| | - Ana R Santa-Maria
- Institute of Biophysics, Biological Research Centre, Temesvári krt. 62, Szeged, 6726, Hungary
- Wyss Institute for Biologically Inspired Engineering at Harvard University, 3 Blackfan Circle, Boston, MA, 02115, USA
| | - Lilla Barna
- Institute of Biophysics, Biological Research Centre, Temesvári krt. 62, Szeged, 6726, Hungary
| | - András Kincses
- Institute of Biophysics, Biological Research Centre, Temesvári krt. 62, Szeged, 6726, Hungary
| | - György Biczó
- Department of Medicine, University of Szeged, Kálvária sgt 57, Szeged, 6725, Hungary
| | - Zsolt Balla
- Department of Medicine, University of Szeged, Kálvária sgt 57, Szeged, 6725, Hungary
- Institute of Applied Sciences, Department of Environmental Biology and Education, Juhász Gyula Faculty of Education, University of Szeged, Boldogasszony sgt. 6, Szeged, 6725, Hungary
| | - Balázs Kui
- Department of Medicine, University of Szeged, Kálvária sgt 57, Szeged, 6725, Hungary
| | - József Maléth
- Department of Medicine, University of Szeged, Kálvária sgt 57, Szeged, 6725, Hungary
- HAS-USZ Momentum Epithelial Cell Signaling and Secretion Research Group, University of Szeged, Dóm sqr. 10, Szeged, 6720, Hungary
- HCEMM-SZTE Molecular Gastroenterology Research Group, University of Szeged, Dóm sqr. 10, Szeged, 6720, Hungary
| | - László Cervenak
- Department of Internal Medicine and Hematology, Research Laboratory, Semmelweis University, Üllői út 26, Budapest, 1085, Hungary
| | - Vilmos Tubak
- Creative Laboratory Ltd, Temesvári krt. 62, Szeged, 6726, Hungary
| | - Ágnes Kittel
- Institute of Experimental Medicine, Eötvös Loránd Research Network, Szigony u. 43, Budapest, 1083, Hungary
| | - Zoltán Rakonczay
- Department of Medicine, University of Szeged, Kálvária sgt 57, Szeged, 6725, Hungary
- Department of Pathophysiology, University of Szeged, Semmelweis u. 1, Szeged, 6701, Hungary
| | - Mária A Deli
- Institute of Biophysics, Biological Research Centre, Temesvári krt. 62, Szeged, 6726, Hungary.
| |
Collapse
|
36
|
Tezuka K, Suzuki M, Sato R, Kawarada S, Terasaki T, Uchida Y. Activation of Annexin
A2
signaling at the blood‐brain barrier in a mouse model of multiple sclerosis. J Neurochem 2022; 160:662-674. [DOI: 10.1111/jnc.15578] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2021] [Revised: 01/18/2022] [Accepted: 01/18/2022] [Indexed: 11/26/2022]
Affiliation(s)
- Kenta Tezuka
- Graduate School of Pharmaceutical Sciences Tohoku University Japan
| | - Masayoshi Suzuki
- Graduate School of Pharmaceutical Sciences Tohoku University Japan
| | - Risa Sato
- Graduate School of Pharmaceutical Sciences Tohoku University Japan
| | - Shohei Kawarada
- Graduate School of Pharmaceutical Sciences Tohoku University Japan
| | - Tetsuya Terasaki
- Graduate School of Pharmaceutical Sciences Tohoku University Japan
| | - Yasuo Uchida
- Graduate School of Pharmaceutical Sciences Tohoku University Japan
| |
Collapse
|
37
|
Veszelka S, Mészáros M, Porkoláb G, Szecskó A, Kondor N, Ferenc G, Polgár TF, Katona G, Kóta Z, Kelemen L, Páli T, Vigh JP, Walter FR, Bolognin S, Schwamborn JC, Jan JS, Deli MA. A Triple Combination of Targeting Ligands Increases the Penetration of Nanoparticles across a Blood-Brain Barrier Culture Model. Pharmaceutics 2021; 14:pharmaceutics14010086. [PMID: 35056983 PMCID: PMC8778049 DOI: 10.3390/pharmaceutics14010086] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Revised: 12/17/2021] [Accepted: 12/21/2021] [Indexed: 12/30/2022] Open
Abstract
Nanosized drug delivery systems targeting transporters of the blood-brain barrier (BBB) are promising carriers to enhance the penetration of therapeutics into the brain. The expression of solute carriers (SLC) is high and shows a specific pattern at the BBB. Here we show that targeting ligands ascorbic acid, leucine and glutathione on nanoparticles elevated the uptake of albumin cargo in cultured primary rat brain endothelial cells. Moreover, we demonstrated the ability of the triple-targeted nanovesicles to deliver their cargo into midbrain organoids after crossing the BBB model. The cellular uptake was temperature- and energy-dependent based on metabolic inhibition. The process was decreased by filipin and cytochalasin D, indicating that the cellular uptake of nanoparticles was partially mediated by endocytosis. The uptake of the cargo encapsulated in triple-targeted nanoparticles increased after modification of the negative zeta potential of endothelial cells by treatment with a cationic lipid or after cleaving the glycocalyx with an enzyme. We revealed that targeted nanoparticles elevated plasma membrane fluidity, indicating the fusion of nanovesicles with endothelial cell membranes. Our data indicate that labeling nanoparticles with three different ligands of multiple transporters of brain endothelial cells can promote the transfer and delivery of molecules across the BBB.
Collapse
Affiliation(s)
- Szilvia Veszelka
- Biological Research Centre, Institute of Biophysics, Eötvös Loránd Research Network, Temesvári krt. 62, H-6726 Szeged, Hungary; (M.M.); (G.P.); (A.S.); (N.K.); (T.F.P.); (Z.K.); (L.K.); (T.P.); (J.P.V.); (F.R.W.)
- Correspondence: (S.V.); (M.A.D.)
| | - Mária Mészáros
- Biological Research Centre, Institute of Biophysics, Eötvös Loránd Research Network, Temesvári krt. 62, H-6726 Szeged, Hungary; (M.M.); (G.P.); (A.S.); (N.K.); (T.F.P.); (Z.K.); (L.K.); (T.P.); (J.P.V.); (F.R.W.)
| | - Gergő Porkoláb
- Biological Research Centre, Institute of Biophysics, Eötvös Loránd Research Network, Temesvári krt. 62, H-6726 Szeged, Hungary; (M.M.); (G.P.); (A.S.); (N.K.); (T.F.P.); (Z.K.); (L.K.); (T.P.); (J.P.V.); (F.R.W.)
- Doctoral School of Biology, University of Szeged, Dugonics tér 13, H-6720 Szeged, Hungary
| | - Anikó Szecskó
- Biological Research Centre, Institute of Biophysics, Eötvös Loránd Research Network, Temesvári krt. 62, H-6726 Szeged, Hungary; (M.M.); (G.P.); (A.S.); (N.K.); (T.F.P.); (Z.K.); (L.K.); (T.P.); (J.P.V.); (F.R.W.)
| | - Nóra Kondor
- Biological Research Centre, Institute of Biophysics, Eötvös Loránd Research Network, Temesvári krt. 62, H-6726 Szeged, Hungary; (M.M.); (G.P.); (A.S.); (N.K.); (T.F.P.); (Z.K.); (L.K.); (T.P.); (J.P.V.); (F.R.W.)
| | - Györgyi Ferenc
- Biological Research Centre, Institute of Plant Biology, Eötvös Loránd Research Network, Temesvári krt. 62, H-6726 Szeged, Hungary;
| | - Tamás F. Polgár
- Biological Research Centre, Institute of Biophysics, Eötvös Loránd Research Network, Temesvári krt. 62, H-6726 Szeged, Hungary; (M.M.); (G.P.); (A.S.); (N.K.); (T.F.P.); (Z.K.); (L.K.); (T.P.); (J.P.V.); (F.R.W.)
| | - Gábor Katona
- Faculty of Pharmacy, Institute of Pharmaceutical Technology and Regulatory Affairs, University of Szeged, Eötvös u. 6, H-6720 Szeged, Hungary;
| | - Zoltán Kóta
- Biological Research Centre, Institute of Biophysics, Eötvös Loránd Research Network, Temesvári krt. 62, H-6726 Szeged, Hungary; (M.M.); (G.P.); (A.S.); (N.K.); (T.F.P.); (Z.K.); (L.K.); (T.P.); (J.P.V.); (F.R.W.)
| | - Lóránd Kelemen
- Biological Research Centre, Institute of Biophysics, Eötvös Loránd Research Network, Temesvári krt. 62, H-6726 Szeged, Hungary; (M.M.); (G.P.); (A.S.); (N.K.); (T.F.P.); (Z.K.); (L.K.); (T.P.); (J.P.V.); (F.R.W.)
| | - Tibor Páli
- Biological Research Centre, Institute of Biophysics, Eötvös Loránd Research Network, Temesvári krt. 62, H-6726 Szeged, Hungary; (M.M.); (G.P.); (A.S.); (N.K.); (T.F.P.); (Z.K.); (L.K.); (T.P.); (J.P.V.); (F.R.W.)
| | - Judit P. Vigh
- Biological Research Centre, Institute of Biophysics, Eötvös Loránd Research Network, Temesvári krt. 62, H-6726 Szeged, Hungary; (M.M.); (G.P.); (A.S.); (N.K.); (T.F.P.); (Z.K.); (L.K.); (T.P.); (J.P.V.); (F.R.W.)
- Doctoral School of Biology, University of Szeged, Dugonics tér 13, H-6720 Szeged, Hungary
| | - Fruzsina R. Walter
- Biological Research Centre, Institute of Biophysics, Eötvös Loránd Research Network, Temesvári krt. 62, H-6726 Szeged, Hungary; (M.M.); (G.P.); (A.S.); (N.K.); (T.F.P.); (Z.K.); (L.K.); (T.P.); (J.P.V.); (F.R.W.)
| | - Silvia Bolognin
- Luxembourg Centre for Systems Biomedicine (LCSB), Developmental and Cellular Biology, University of Luxembourg, 4365 Belvaux, Luxembourg; (S.B.); (J.C.S.)
| | - Jens C. Schwamborn
- Luxembourg Centre for Systems Biomedicine (LCSB), Developmental and Cellular Biology, University of Luxembourg, 4365 Belvaux, Luxembourg; (S.B.); (J.C.S.)
| | - Jeng-Shiung Jan
- Department of Chemical Engineering, National Cheng Kung University, Tainan 70101, Taiwan;
| | - Mária A. Deli
- Biological Research Centre, Institute of Biophysics, Eötvös Loránd Research Network, Temesvári krt. 62, H-6726 Szeged, Hungary; (M.M.); (G.P.); (A.S.); (N.K.); (T.F.P.); (Z.K.); (L.K.); (T.P.); (J.P.V.); (F.R.W.)
- Correspondence: (S.V.); (M.A.D.)
| |
Collapse
|
38
|
New In Vitro Methodology for Kinetics Distribution Prediction in the Brain. An Additional Step towards an Animal-Free Approach. Animals (Basel) 2021; 11:ani11123521. [PMID: 34944295 PMCID: PMC8697921 DOI: 10.3390/ani11123521] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Revised: 11/30/2021] [Accepted: 12/02/2021] [Indexed: 12/02/2022] Open
Abstract
Simple Summary The prevalence of neurological disorders in humans is rising year after year. This fact necessitates the development of new drugs for treating these pathologies. Traditionally, drugs have been tested in animals prior to use in human experiments; however, the use of animals in experimentation must be controlled and as low as possible. Because of that, here we proposed a new in vitro approach with which the access and distribution of drugs into the brain can be evaluated without using/killing any animals. Abstract The development of new drugs or formulations for central nervous system (CNS) diseases is a complex pharmacologic and pharmacokinetic process; it is important to evaluate their access to the CNS through the blood–brain barrier (BBB) and their distribution once they have acceded to the brain. The gold standard tool for obtaining this information is the animal microdialysis technique; however, according to 3Rs principles, it would be better to have an “animal-free” alternative technique. Because of that, the purpose of this work was to develop a new formulation to substitute the brain homogenate in the in vitro tests used for the prediction of a drug’s distribution in the brain. Fresh eggs have been used to prepare an emulsion with the same proportion in proteins and lipids as a human brain; this emulsion has proved to be able to predict both the unbound fraction of drug in the brain (fu,brain) and the apparent volume of distribution in the brain (Vu,brain) when tested in in vitro permeability tests. The new formulation could be used as a screening tool; only the drugs with a proper in vitro distribution would pass to microdialysis studies, contributing to the refinement, reduction and replacement of animals in research.
Collapse
|
39
|
Akel H, Csóka I, Ambrus R, Bocsik A, Gróf I, Mészáros M, Szecskó A, Kozma G, Veszelka S, Deli MA, Kónya Z, Katona G. In Vitro Comparative Study of Solid Lipid and PLGA Nanoparticles Designed to Facilitate Nose-to-Brain Delivery of Insulin. Int J Mol Sci 2021; 22:13258. [PMID: 34948054 PMCID: PMC8703723 DOI: 10.3390/ijms222413258] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Revised: 12/03/2021] [Accepted: 12/06/2021] [Indexed: 01/25/2023] Open
Abstract
The brain insulin metabolism alteration has been addressed as a pathophysiological factor underlying Alzheimer's disease (AD). Insulin can be beneficial in AD, but its macro-polypeptide nature negatively influences the chances of reaching the brain. The intranasal (IN) administration of therapeutics in AD suggests improved brain-targeting. Solid lipid nanoparticles (SLNs) and poly(lactic-co-glycolic acid) nanoparticles (PLGA NPs) are promising carriers to deliver the IN-administered insulin to the brain due to the enhancement of the drug permeability, which can even be improved by chitosan-coating. In the present study, uncoated and chitosan-coated insulin-loaded SLNs and PLGA NPs were formulated and characterized. The obtained NPs showed desirable physicochemical properties supporting IN applicability. The in vitro investigations revealed increased mucoadhesion, nasal diffusion, and drug release rate of both insulin-loaded nanocarriers over native insulin with the superiority of chitosan-coated SLNs. Cell-line studies on human nasal epithelial and brain endothelial cells proved the safety IN applicability of nanoparticles. Insulin-loaded nanoparticles showed improved insulin permeability through the nasal mucosa, which was promoted by chitosan-coating. However, native insulin exceeded the blood-brain barrier (BBB) permeation compared with nanoparticulate formulations. Encapsulating insulin into chitosan-coated NPs can be beneficial for ensuring structural stability, enhancing nasal absorption, followed by sustained drug release.
Collapse
Affiliation(s)
- Hussein Akel
- Faculty of Pharmacy, Institute of Pharmaceutical Technology and Regulatory Affairs, University of Szeged, Eötvös Str. 6, H-6720 Szeged, Hungary; (H.A.); (I.C.); (R.A.)
| | - Ildikó Csóka
- Faculty of Pharmacy, Institute of Pharmaceutical Technology and Regulatory Affairs, University of Szeged, Eötvös Str. 6, H-6720 Szeged, Hungary; (H.A.); (I.C.); (R.A.)
| | - Rita Ambrus
- Faculty of Pharmacy, Institute of Pharmaceutical Technology and Regulatory Affairs, University of Szeged, Eötvös Str. 6, H-6720 Szeged, Hungary; (H.A.); (I.C.); (R.A.)
| | - Alexandra Bocsik
- Biological Research Centre, Institute of Biophysics, Temesvári Blvd. 62, H-6726 Szeged, Hungary; (A.B.); (I.G.); (M.M.); (A.S.); (S.V.); (M.A.D.)
| | - Ilona Gróf
- Biological Research Centre, Institute of Biophysics, Temesvári Blvd. 62, H-6726 Szeged, Hungary; (A.B.); (I.G.); (M.M.); (A.S.); (S.V.); (M.A.D.)
| | - Mária Mészáros
- Biological Research Centre, Institute of Biophysics, Temesvári Blvd. 62, H-6726 Szeged, Hungary; (A.B.); (I.G.); (M.M.); (A.S.); (S.V.); (M.A.D.)
| | - Anikó Szecskó
- Biological Research Centre, Institute of Biophysics, Temesvári Blvd. 62, H-6726 Szeged, Hungary; (A.B.); (I.G.); (M.M.); (A.S.); (S.V.); (M.A.D.)
| | - Gábor Kozma
- Department of Applied & Environmental Chemistry, Faculty of Science and Informatics, Rerrich Béla Sqr. 1, H-6720 Szeged, Hungary; (G.K.); (Z.K.)
| | - Szilvia Veszelka
- Biological Research Centre, Institute of Biophysics, Temesvári Blvd. 62, H-6726 Szeged, Hungary; (A.B.); (I.G.); (M.M.); (A.S.); (S.V.); (M.A.D.)
| | - Mária A. Deli
- Biological Research Centre, Institute of Biophysics, Temesvári Blvd. 62, H-6726 Szeged, Hungary; (A.B.); (I.G.); (M.M.); (A.S.); (S.V.); (M.A.D.)
| | - Zoltán Kónya
- Department of Applied & Environmental Chemistry, Faculty of Science and Informatics, Rerrich Béla Sqr. 1, H-6720 Szeged, Hungary; (G.K.); (Z.K.)
| | - Gábor Katona
- Faculty of Pharmacy, Institute of Pharmaceutical Technology and Regulatory Affairs, University of Szeged, Eötvös Str. 6, H-6720 Szeged, Hungary; (H.A.); (I.C.); (R.A.)
| |
Collapse
|
40
|
Neumaier F, Zlatopolskiy BD, Neumaier B. Drug Penetration into the Central Nervous System: Pharmacokinetic Concepts and In Vitro Model Systems. Pharmaceutics 2021; 13:1542. [PMID: 34683835 PMCID: PMC8538549 DOI: 10.3390/pharmaceutics13101542] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2021] [Revised: 09/17/2021] [Accepted: 09/20/2021] [Indexed: 12/22/2022] Open
Abstract
Delivery of most drugs into the central nervous system (CNS) is restricted by the blood-brain barrier (BBB), which remains a significant bottleneck for development of novel CNS-targeted therapeutics or molecular tracers for neuroimaging. Consistent failure to reliably predict drug efficiency based on single measures for the rate or extent of brain penetration has led to the emergence of a more holistic framework that integrates data from various in vivo, in situ and in vitro assays to obtain a comprehensive description of drug delivery to and distribution within the brain. Coupled with ongoing development of suitable in vitro BBB models, this integrated approach promises to reduce the incidence of costly late-stage failures in CNS drug development, and could help to overcome some of the technical, economic and ethical issues associated with in vivo studies in animal models. Here, we provide an overview of BBB structure and function in vivo, and a summary of the pharmacokinetic parameters that can be used to determine and predict the rate and extent of drug penetration into the brain. We also review different in vitro models with regard to their inherent shortcomings and potential usefulness for development of fast-acting drugs or neurotracers labeled with short-lived radionuclides. In this regard, a special focus has been set on those systems that are sufficiently well established to be used in laboratories without significant bioengineering expertise.
Collapse
Affiliation(s)
- Felix Neumaier
- Institute of Radiochemistry and Experimental Molecular Imaging, Faculty of Medicine and University Hospital Cologne, University of Cologne, Kerpener Str. 62, 50937 Cologne, Germany; (B.D.Z.); (B.N.)
- Forschungszentrum Jülich GmbH, Institute of Neuroscience and Medicine, Nuclear Chemistry (INM-5), Wilhelm-Johnen-Str., 52428 Jülich, Germany
| | - Boris D. Zlatopolskiy
- Institute of Radiochemistry and Experimental Molecular Imaging, Faculty of Medicine and University Hospital Cologne, University of Cologne, Kerpener Str. 62, 50937 Cologne, Germany; (B.D.Z.); (B.N.)
- Forschungszentrum Jülich GmbH, Institute of Neuroscience and Medicine, Nuclear Chemistry (INM-5), Wilhelm-Johnen-Str., 52428 Jülich, Germany
| | - Bernd Neumaier
- Institute of Radiochemistry and Experimental Molecular Imaging, Faculty of Medicine and University Hospital Cologne, University of Cologne, Kerpener Str. 62, 50937 Cologne, Germany; (B.D.Z.); (B.N.)
- Forschungszentrum Jülich GmbH, Institute of Neuroscience and Medicine, Nuclear Chemistry (INM-5), Wilhelm-Johnen-Str., 52428 Jülich, Germany
| |
Collapse
|
41
|
Xiong B, Wang Y, Chen Y, Xing S, Liao Q, Chen Y, Li Q, Li W, Sun H. Strategies for Structural Modification of Small Molecules to Improve Blood-Brain Barrier Penetration: A Recent Perspective. J Med Chem 2021; 64:13152-13173. [PMID: 34505508 DOI: 10.1021/acs.jmedchem.1c00910] [Citation(s) in RCA: 104] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
In the development of central nervous system (CNS) drugs, the blood-brain barrier (BBB) restricts many drugs from entering the brain to exert therapeutic effects. Although many novel delivery methods of large molecule drugs have been designed to assist transport, small molecule drugs account for the vast majority of the CNS drugs used clinically. From this perspective, we review studies from the past five years that have sought to modify small molecules to increase brain exposure. Medicinal chemists make it easier for small molecules to cross the BBB by improving diffusion, reducing efflux, and activating carrier transporters. On the basis of their excellent work, we summarize strategies for structural modification of small molecules to improve BBB penetration. These strategies are expected to provide a reference for the future development of small molecule CNS drugs.
Collapse
Affiliation(s)
- Baichen Xiong
- School of Pharmacy, China Pharmaceutical University, Nanjing 211198, People's Republic of China
| | - Yuanyuan Wang
- School of Pharmacy, China Pharmaceutical University, Nanjing 211198, People's Republic of China
| | - Ying Chen
- Department of Natural Medicinal Chemistry, China Pharmaceutical University, Nanjing 211198, People's Republic of China
| | - Shuaishuai Xing
- School of Pharmacy, China Pharmaceutical University, Nanjing 211198, People's Republic of China
| | - Qinghong Liao
- Department of Natural Medicinal Chemistry, China Pharmaceutical University, Nanjing 211198, People's Republic of China
| | - Yao Chen
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, People's Republic of China
| | - Qi Li
- School of Pharmacy, China Pharmaceutical University, Nanjing 211198, People's Republic of China.,School of Basic Medicine, Qingdao University, Qingdao 266071, People's Republic of China
| | - Wei Li
- School of Pharmacy, China Pharmaceutical University, Nanjing 211198, People's Republic of China
| | - Haopeng Sun
- School of Pharmacy, China Pharmaceutical University, Nanjing 211198, People's Republic of China
| |
Collapse
|
42
|
Watanabe D, Nakagawa S, Morofuji Y, Tóth AE, Vastag M, Aruga J, Niwa M, Deli MA. Characterization of a Primate Blood-Brain Barrier Co-Culture Model Prepared from Primary Brain Endothelial Cells, Pericytes and Astrocytes. Pharmaceutics 2021; 13:pharmaceutics13091484. [PMID: 34575559 PMCID: PMC8470770 DOI: 10.3390/pharmaceutics13091484] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Revised: 09/09/2021] [Accepted: 09/13/2021] [Indexed: 01/25/2023] Open
Abstract
Culture models of the blood-brain barrier (BBB) are important research tools. Their role in the preclinical phase of drug development to estimate the permeability for potential neuropharmaceuticals is especially relevant. Since species differences in BBB transport systems exist, primate models are considered as predictive for drug transport to brain in humans. Based on our previous expertise we have developed and characterized a non-human primate co-culture BBB model using primary cultures of monkey brain endothelial cells, rat brain pericytes, and rat astrocytes. Monkey brain endothelial cells in the presence of both pericytes and astrocytes (EPA model) expressed enhanced barrier properties and increased levels of tight junction proteins occludin, claudin-5, and ZO-1. Co-culture conditions also elevated the expression of key BBB influx and efflux transporters, including glucose transporter-1, MFSD2A, ABCB1, and ABCG2. The correlation between the endothelial permeability coefficients of 10 well known drugs was higher (R2 = 0.8788) when the monkey and rat BBB culture models were compared than when the monkey culture model was compared to mouse in vivo data (R2 = 0.6619), hinting at transporter differences. The applicability of the new non-human primate model in drug discovery has been proven in several studies.
Collapse
Affiliation(s)
- Daisuke Watanabe
- Department of Medical Pharmacology, Nagasaki University Graduate School of Biomedical Sciences, 1-12-4 Sakamoto, Nagasaki 852-8523, Japan; (D.W.); (J.A.)
- BBB Laboratory, PharmaCo-Cell Co., Ltd., Nagasaki 852-8135, Japan;
| | - Shinsuke Nakagawa
- Department of Pharmaceutical Care and Health Sciences, Faculty of Pharmaceutical Sciences, Fukuoka University, 8-19-1 Nanakuma, Jonan-ku, Fukuoka 814-0180, Japan;
| | - Yoichi Morofuji
- Department of Neurosurgery, Nagasaki University Graduate School of Biomedical Sciences, 1-7-1 Sakamoto, Nagasaki 852-8501, Japan;
| | - Andrea E. Tóth
- Institute of Biophysics, Biological Research Centre, Temesvári krt. 62, H-6726 Szeged, Hungary;
| | - Monika Vastag
- In Vitro Metabolism Research, Division of Pharmacology and Drug Safety Research, Gedeon Richter Plc., Gyömrői út 19-21, H-1103 Budapest, Hungary;
| | - Jun Aruga
- Department of Medical Pharmacology, Nagasaki University Graduate School of Biomedical Sciences, 1-12-4 Sakamoto, Nagasaki 852-8523, Japan; (D.W.); (J.A.)
| | - Masami Niwa
- BBB Laboratory, PharmaCo-Cell Co., Ltd., Nagasaki 852-8135, Japan;
| | - Mária A. Deli
- Institute of Biophysics, Biological Research Centre, Temesvári krt. 62, H-6726 Szeged, Hungary;
- Correspondence:
| |
Collapse
|
43
|
Physiologically Based Pharmacokinetic (PBPK) Modeling for Predicting Brain Levels of Drug in Rat. Pharmaceutics 2021; 13:pharmaceutics13091402. [PMID: 34575476 PMCID: PMC8471455 DOI: 10.3390/pharmaceutics13091402] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Revised: 08/20/2021] [Accepted: 08/30/2021] [Indexed: 11/17/2022] Open
Abstract
One of the main obstacles in neurological disease treatment is the presence of the blood-brain barrier. New predictive high-throughput screening tools are essential to avoid costly failures in the advanced phases of development and to contribute to the 3 Rs policy. The objective of this work was to jointly develop a new in vitro system coupled with a physiological-based pharmacokinetic (PBPK) model able to predict brain concentration levels of different drugs in rats. Data from in vitro tests with three different cells lines (MDCK, MDCK-MDR1 and hCMEC/D3) were used together with PK parameters and three scaling factors for adjusting the model predictions to the brain and plasma profiles of six model drugs. Later, preliminary quantitative structure-property relationships (QSPRs) were constructed between the scaling factors and the lipophilicity of drugs. The predictability of the model was evaluated by internal validation. It was concluded that the PBPK model, incorporating the barrier resistance to transport, the disposition within the brain and the drug-brain binding combined with MDCK data, provided the best predictions for passive diffusion and carrier-mediated transported drugs, while in the other cell lines, active transport influence can bias predictions.
Collapse
|
44
|
Santa-Maria AR, Walter FR, Figueiredo R, Kincses A, Vigh JP, Heymans M, Culot M, Winter P, Gosselet F, Dér A, Deli MA. Flow induces barrier and glycocalyx-related genes and negative surface charge in a lab-on-a-chip human blood-brain barrier model. J Cereb Blood Flow Metab 2021; 41:2201-2215. [PMID: 33563079 PMCID: PMC8393308 DOI: 10.1177/0271678x21992638] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Microfluidic lab-on-a-chip (LOC) devices allow the study of blood-brain barrier (BBB) properties in dynamic conditions. We studied a BBB model, consisting of human endothelial cells derived from hematopoietic stem cells in co-culture with brain pericytes, in an LOC device to study fluid flow in the regulation of endothelial, BBB and glycocalyx-related genes and surface charge. The highly negatively charged endothelial surface glycocalyx functions as mechano-sensor detecting shear forces generated by blood flow on the luminal side of brain endothelial cells and contributes to the physical barrier of the BBB. Despite the importance of glycocalyx in the regulation of BBB permeability in physiological conditions and in diseases, the underlying mechanisms remained unclear. The MACE-seq gene expression profiling analysis showed differentially expressed endothelial, BBB and glycocalyx core protein genes after fluid flow, as well as enriched pathways for the extracellular matrix molecules. We observed increased barrier properties, a higher intensity glycocalyx staining and a more negative surface charge of human brain-like endothelial cells (BLECs) in dynamic conditions. Our work is the first study to provide data on BBB properties and glycocalyx of BLECs in an LOC device under dynamic conditions and confirms the importance of fluid flow for BBB culture models.
Collapse
Affiliation(s)
- Ana R Santa-Maria
- Institute of Biophysics, Biological Research Centre, Szeged, Hungary.,Doctoral School of Biology, University of Szeged, Szeged, Hungary.,Department of Biotechnology, University of Szeged, Szeged, Hungary
| | - Fruzsina R Walter
- Institute of Biophysics, Biological Research Centre, Szeged, Hungary.,Department of Biotechnology, University of Szeged, Szeged, Hungary
| | - Ricardo Figueiredo
- GenXPro GmbH, Frankfurt-Am-Main, Germany.,Johann Wolfgang Goethe University, Frankfurt, Frankfurt-Am-Main, Germany
| | - András Kincses
- Institute of Biophysics, Biological Research Centre, Szeged, Hungary.,Doctoral School of Multidisciplinary Medical Sciences, University of Szeged, Szeged, Hungary
| | - Judit P Vigh
- Institute of Biophysics, Biological Research Centre, Szeged, Hungary.,Doctoral School of Biology, University of Szeged, Szeged, Hungary
| | - Marjolein Heymans
- Université d'Artois, Laboratoire de la Barrière Hémato-Encéphalique (LBHE), Lens, France
| | - Maxime Culot
- Université d'Artois, Laboratoire de la Barrière Hémato-Encéphalique (LBHE), Lens, France
| | | | - Fabien Gosselet
- Université d'Artois, Laboratoire de la Barrière Hémato-Encéphalique (LBHE), Lens, France
| | - András Dér
- Institute of Biophysics, Biological Research Centre, Szeged, Hungary
| | - Mária A Deli
- Institute of Biophysics, Biological Research Centre, Szeged, Hungary
| |
Collapse
|
45
|
NMDA receptors elicit flux-independent intracellular Ca 2+ signals via metabotropic glutamate receptors and flux-dependent nitric oxide release in human brain microvascular endothelial cells. Cell Calcium 2021; 99:102454. [PMID: 34454368 DOI: 10.1016/j.ceca.2021.102454] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 07/28/2021] [Accepted: 08/12/2021] [Indexed: 12/20/2022]
Abstract
The excitatory neurotransmitter glutamate gates post-synaptic N-methyl-d-aspartate (NMDA) receptors (NMDARs) to mediate extracellular Ca2+ entry and stimulate neuronal nitric oxide (NO) synthase to release NO and trigger neurovascular coupling (NVC). Neuronal and glial NMDARs may also operate in a flux-independent manner, although it is unclear whether their non-ionotropic mode of action is involved in NVC. Recently, endothelial NMDARs were found to trigger Ca2+-dependent NO production and induce NVC, but the underlying mode of signaling remains elusive. Herein, we report that GluN1 protein, as well as GluN2C and GluN3B transcripts and proteins, were expressed and that NMDA did not elicit inward currents, but induced a dose-dependent increase in intracellular Ca2+ concentration ([Ca2+]i) in the human brain microvascular endothelial cell line, hCMEC/D3. A multidisciplinary approach, including live cell imaging, whole-cell patch-clamp recordings, pharmacological manipulation and gene targeting, revealed that NMDARs increase the [Ca2+]i in a flux-independent manner in hCMEC/D3 cells. The Ca2+ response to NMDA was triggered by endogenous Ca2+ release from the endoplasmic reticulum and the lysosomal Ca2+ stores and sustained by store-operated Ca2+ entry. Unexpectedly, pharmacological and genetic blockade of mGluR1 and mGluR5 dramatically impaired NMDARs-mediated Ca2+ signals. These findings indicate that NMDARs may increase the endothelial [Ca2+]i in a flux-independent manner via group 1 mGluRs. However, imaging of DAF-FM fluorescence revealed that NMDARs may also induce Ca2+-dependent NO release by signaling in a flux-dependent manner. These findings, therefore, shed novel light on the mechanisms whereby brain microvascular endothelium decodes glutamatergic signaling and regulates NVC.
Collapse
|
46
|
Fekete T, Mészáros M, Szegletes Z, Vizsnyiczai G, Zimányi L, Deli MA, Veszelka S, Kelemen L. Optically Manipulated Microtools to Measure Adhesion of the Nanoparticle-Targeting Ligand Glutathione to Brain Endothelial Cells. ACS APPLIED MATERIALS & INTERFACES 2021; 13:39018-39029. [PMID: 34397215 DOI: 10.1021/acsami.1c08454] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Targeting nanoparticles as drug delivery platforms is crucial to facilitate their cellular entry. Docking of nanoparticles by targeting ligands on cell membranes is the first step for the initiation of cellular uptake. As a model system, we studied brain microvascular endothelial cells, which form the anatomical basis of the blood-brain barrier, and the tripeptide glutathione, one of the most effective targeting ligands of nanoparticles to cross the blood-brain barrier. To investigate this initial docking step between glutathione and the membrane of living brain endothelial cells, we applied our recently developed innovative optical method. We present a microtool, with a task-specific geometry used as a probe, actuated by multifocus optical tweezers to characterize the adhesion probability and strength of glutathione-coated surfaces to the cell membrane of endothelial cells. The binding probability of the glutathione-coated surface and the adhesion force between the microtool and cell membrane was measured in a novel arrangement: cells were cultured on a vertical polymer wall and the mechanical forces were generated laterally and at the same time, perpendicularly to the plasma membrane. The adhesion force values were also determined with more conventional atomic force microscopy (AFM) measurements using functionalized colloidal probes. The optical trapping-based method was found to be suitable to measure very low adhesion forces (≤ 20 pN) without a high level of noise, which is characteristic for AFM measurements in this range. The holographic optical tweezers-directed functionalized microtools may help characterize the adhesion step of nanoparticles initiating transcytosis and select ligands to target nanoparticles.
Collapse
Affiliation(s)
- Tamás Fekete
- Institute of Biophysics, Biological Research Centre, Eötvös Loránd Research Network (ELKH), Szeged 6726, Hungary
- Doctoral School in Multidisciplinary Medicine, University of Szeged, Szeged 6720, Hungary
| | - Mária Mészáros
- Institute of Biophysics, Biological Research Centre, Eötvös Loránd Research Network (ELKH), Szeged 6726, Hungary
| | - Zsolt Szegletes
- Institute of Biophysics, Biological Research Centre, Eötvös Loránd Research Network (ELKH), Szeged 6726, Hungary
| | - Gaszton Vizsnyiczai
- Institute of Biophysics, Biological Research Centre, Eötvös Loránd Research Network (ELKH), Szeged 6726, Hungary
| | - László Zimányi
- Institute of Biophysics, Biological Research Centre, Eötvös Loránd Research Network (ELKH), Szeged 6726, Hungary
| | - Mária A Deli
- Institute of Biophysics, Biological Research Centre, Eötvös Loránd Research Network (ELKH), Szeged 6726, Hungary
| | - Szilvia Veszelka
- Institute of Biophysics, Biological Research Centre, Eötvös Loránd Research Network (ELKH), Szeged 6726, Hungary
| | - Lóránd Kelemen
- Institute of Biophysics, Biological Research Centre, Eötvös Loránd Research Network (ELKH), Szeged 6726, Hungary
| |
Collapse
|
47
|
Matsunaga Y, Nakagawa S, Morofuji Y, Dohgu S, Watanabe D, Horie N, Izumo T, Niwa M, Walter FR, Santa-Maria AR, Deli MA, Matsuo T. MAP Kinase Pathways in Brain Endothelial Cells and Crosstalk with Pericytes and Astrocytes Mediate Contrast-Induced Blood-Brain Barrier Disruption. Pharmaceutics 2021; 13:1272. [PMID: 34452232 PMCID: PMC8400240 DOI: 10.3390/pharmaceutics13081272] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Revised: 08/10/2021] [Accepted: 08/13/2021] [Indexed: 11/16/2022] Open
Abstract
Neurointervention with contrast media (CM) has rapidly increased, but the impact of CM extravasation and the related side effects remain controversial. This study investigated the effect of CM on blood-brain barrier (BBB) integrity. We established in vitro BBB models using primary cultures of rat BBB-related cells. To assess the effects of CM on BBB functions, we evaluated transendothelial electrical resistance, permeability, and tight junction (TJ) protein expression using immunohistochemistry (IHC) and Western blotting. To investigate the mechanism of iopamidol-induced barrier dysfunction, the role of mitogen-activated protein (MAP) kinases in brain endothelial cells was examined. We assessed the effect of conditioned medium derived from astrocytes and pericytes under iopamidol treatment. Short-term iopamidol exposure on the luminal side induced transient, while on the abluminal side caused persistent BBB dysfunction. IHC and immunoblotting revealed CM decreased the expression of TJ proteins. Iopamidol-induced barrier dysfunction was improved via the regulation of MAP kinase pathways. Conditioned medium from CM-exposed pericytes or astrocytes lacks the ability to enhance barrier function. CM may cause BBB dysfunction. MAP kinase pathways in brain endothelial cells and the interactions of astrocytes and pericytes mediate iopamidol-induced barrier dysfunction. CM extravasation may have negative effects on clinical outcomes in patients.
Collapse
Affiliation(s)
- Yuki Matsunaga
- Department of Neurosurgery, Graduate School of Biomedical Sciences, Nagasaki University, 1-7-1 Sakamoto, Nagasaki 852-8501, Japan; (Y.M.); (N.H.); (T.I.); (T.M.)
| | - Shinsuke Nakagawa
- Department of Pharmaceutical Care and Health Sciences, Faculty of Pharmaceutical Sciences, Fukuoka University, 8-19-1 Nanakuma, Jonan-ku, Fukuoka 814-0180, Japan; (S.N.); (S.D.)
| | - Yoichi Morofuji
- Department of Neurosurgery, Graduate School of Biomedical Sciences, Nagasaki University, 1-7-1 Sakamoto, Nagasaki 852-8501, Japan; (Y.M.); (N.H.); (T.I.); (T.M.)
| | - Shinya Dohgu
- Department of Pharmaceutical Care and Health Sciences, Faculty of Pharmaceutical Sciences, Fukuoka University, 8-19-1 Nanakuma, Jonan-ku, Fukuoka 814-0180, Japan; (S.N.); (S.D.)
| | - Daisuke Watanabe
- BBB Laboratory, PharmaCo-Cell Company Ltd., Dai-ichi-senshu bldg. 2nd Floor, 6-19 Chitose-machi, Nagasaki 852-8135, Japan; (D.W.); (M.N.)
| | - Nobutaka Horie
- Department of Neurosurgery, Graduate School of Biomedical Sciences, Nagasaki University, 1-7-1 Sakamoto, Nagasaki 852-8501, Japan; (Y.M.); (N.H.); (T.I.); (T.M.)
| | - Tsuyoshi Izumo
- Department of Neurosurgery, Graduate School of Biomedical Sciences, Nagasaki University, 1-7-1 Sakamoto, Nagasaki 852-8501, Japan; (Y.M.); (N.H.); (T.I.); (T.M.)
| | - Masami Niwa
- BBB Laboratory, PharmaCo-Cell Company Ltd., Dai-ichi-senshu bldg. 2nd Floor, 6-19 Chitose-machi, Nagasaki 852-8135, Japan; (D.W.); (M.N.)
| | - Fruzsina R. Walter
- Biological Barriers Research Group, Institute of Biophysics, Biological Research Centre, 6726 Szeged, Hungary; (F.R.W.); (A.R.S.-M.); (M.A.D.)
| | - Ana Raquel Santa-Maria
- Biological Barriers Research Group, Institute of Biophysics, Biological Research Centre, 6726 Szeged, Hungary; (F.R.W.); (A.R.S.-M.); (M.A.D.)
| | - Maria A. Deli
- Biological Barriers Research Group, Institute of Biophysics, Biological Research Centre, 6726 Szeged, Hungary; (F.R.W.); (A.R.S.-M.); (M.A.D.)
| | - Takayuki Matsuo
- Department of Neurosurgery, Graduate School of Biomedical Sciences, Nagasaki University, 1-7-1 Sakamoto, Nagasaki 852-8501, Japan; (Y.M.); (N.H.); (T.I.); (T.M.)
| |
Collapse
|
48
|
Gericke B, Borsdorf S, Wienböker I, Noack A, Noack S, Löscher W. Similarities and differences in the localization, trafficking, and function of P-glycoprotein in MDR1-EGFP-transduced rat versus human brain capillary endothelial cell lines. Fluids Barriers CNS 2021; 18:36. [PMID: 34344390 PMCID: PMC8330100 DOI: 10.1186/s12987-021-00266-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Accepted: 06/28/2021] [Indexed: 12/24/2022] Open
Abstract
Background In vitro models based on brain capillary endothelial cells (BCECs) are among the most versatile tools in blood–brain barrier research for testing drug penetration into the brain and how this is affected by efflux transporters such as P-glycoprotein (Pgp). However, compared to freshly isolated brain capillaries or primary BCECs, the expression of Pgp in immortalized BCEC lines is markedly lower, which prompted us previously to transduce the widely used human BCEC line hCMEC/D3 with a doxycycline-inducible MDR1-EGFP fusion plasmid. The EGFP-labeled Pgp in these cells allows studying the localization and trafficking of the transporter and how these processes are affected by drug exposure. Here we used this strategy for the rat BCEC line RBE4 and performed a face-to-face comparison of RBE4 and hCMEC/D3 wild-type (WT) and MDR1-EGFP transduced cells. Methods MDR1-EGFP-transduced variants were derived from WT cells by lentiviral transduction, using an MDR1-linker-EGFP vector. Localization, trafficking, and function of Pgp were compared in WT and MDR1-EGFP transduced cell lines. Primary cultures of rat BCECs and freshly isolated rat brain capillaries were used for comparison. Results All cells exhibited typical BCEC morphology. However, significant differences were observed in the localization of Pgp in that RBE4-MDR1-EGFP cells expressed Pgp primarily at the plasma membrane, whereas in hCMEC/D3 cells, the Pgp-EGFP fusion protein was visible both at the plasma membrane and in endolysosomal vesicles. Exposure to doxorubicin increased the number of Pgp-EGFP-positive endolysosomes, indicating a lysosomotropic effect. Furthermore, lysosomal trapping of doxorubicin was observed, likely contributing to the protection of the cell nucleus from damage. In cocultures of WT and MDR1-EGFP transduced cells, intercellular Pgp-EGFP trafficking was observed in RBE4 cells as previously reported for hCMEC/D3 cells. Compared to WT cells, the MDR1-EGFP transduced cells exhibited a significantly higher expression and function of Pgp. However, the junctional tightness of WT and MDR1-EGFP transduced RBE4 and hCMEC/D3 cells was markedly lower than that of primary BCECs, excluding the use of the cell lines for studying vectorial drug transport. Conclusions The present data indicate that MDR1-EGFP transduced RBE4 cells are an interesting tool to study the biogenesis of lysosomes and Pgp-mediated lysosomal drug trapping in response to chemotherapeutic agents and other compounds at the level of the blood–brain barrier. Supplementary Information The online version contains supplementary material available at 10.1186/s12987-021-00266-z.
Collapse
Affiliation(s)
- Birthe Gericke
- Department of Pharmacology, Toxicology, and Pharmacy, University of Veterinary Medicine Hannover, Bünteweg 17, 30559, Hannover, Germany.,Center for Systems Neuroscience, Hannover, Germany
| | - Saskia Borsdorf
- Department of Pharmacology, Toxicology, and Pharmacy, University of Veterinary Medicine Hannover, Bünteweg 17, 30559, Hannover, Germany
| | - Inka Wienböker
- Department of Pharmacology, Toxicology, and Pharmacy, University of Veterinary Medicine Hannover, Bünteweg 17, 30559, Hannover, Germany.,Center for Systems Neuroscience, Hannover, Germany
| | - Andreas Noack
- Department of Pharmacology, Toxicology, and Pharmacy, University of Veterinary Medicine Hannover, Bünteweg 17, 30559, Hannover, Germany
| | - Sandra Noack
- Department of Trauma Surgery, Hannover Medical School, Hannover, Germany
| | - Wolfgang Löscher
- Department of Pharmacology, Toxicology, and Pharmacy, University of Veterinary Medicine Hannover, Bünteweg 17, 30559, Hannover, Germany. .,Center for Systems Neuroscience, Hannover, Germany.
| |
Collapse
|
49
|
Augustine R, Aqel AH, Kalva SN, Joshy KS, Nayeem A, Hasan A. Bioengineered microfluidic blood-brain barrier models in oncology research. Transl Oncol 2021; 14:101087. [PMID: 33865030 PMCID: PMC8066424 DOI: 10.1016/j.tranon.2021.101087] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Revised: 02/25/2021] [Accepted: 03/23/2021] [Indexed: 12/15/2022] Open
Abstract
Metastasis is the major reason for most brain tumors with up to a 50% chance of occurrence in patients with other types of malignancies. Brain metastasis occurs if cancer cells succeed to cross the 'blood-brain barrier' (BBB). Moreover, changes in the structure and function of BBB can lead to the onset and progression of diseases including neurological disorders and brain-metastases. Generating BBB models with structural and functional features of intact BBB is highly important to better understand the molecular mechanism of such ailments and finding novel therapeutic agents targeting them. Hence, researchers are developing novel in vitro BBB platforms that can recapitulate the structural and functional characteristics of BBB. Brain endothelial cells-based in vitro BBB models have thus been developed to investigate the mechanism of brain metastasis through BBB and facilitate the testing of brain targeted anticancer drugs. Bioengineered constructs integrated with microfluidic platforms are vital tools for recapitulating the features of BBB in vitro closely as possible. In this review, we outline the fundamentals of BBB biology, recent developments in the microfluidic BBB platforms, and provide a concise discussion of diverse types of bioengineered BBB models with an emphasis on the application of them in brain metastasis and cancer research in general. We also provide insights into the challenges and prospects of the current bioengineered microfluidic platforms in cancer research.
Collapse
Affiliation(s)
- Robin Augustine
- Department of Mechanical and Industrial Engineering, College of Engineering, Qatar University, 2713 Doha, Qatar; Biomedical Research Center (BRC), Qatar University, PO Box 2713 Doha, Qatar.
| | - Ahmad H Aqel
- Department of Mechanical and Industrial Engineering, College of Engineering, Qatar University, 2713 Doha, Qatar; Biomedical Research Center (BRC), Qatar University, PO Box 2713 Doha, Qatar
| | - Sumama Nuthana Kalva
- Department of Mechanical and Industrial Engineering, College of Engineering, Qatar University, 2713 Doha, Qatar; Biomedical Research Center (BRC), Qatar University, PO Box 2713 Doha, Qatar
| | - K S Joshy
- Department of Mechanical and Industrial Engineering, College of Engineering, Qatar University, 2713 Doha, Qatar; Biomedical Research Center (BRC), Qatar University, PO Box 2713 Doha, Qatar
| | - Ajisha Nayeem
- Department of Biotechnology, St. Mary's College, Thrissur 680020, Kerala, India
| | - Anwarul Hasan
- Department of Mechanical and Industrial Engineering, College of Engineering, Qatar University, 2713 Doha, Qatar; Biomedical Research Center (BRC), Qatar University, PO Box 2713 Doha, Qatar.
| |
Collapse
|
50
|
Vigh JP, Kincses A, Ozgür B, Walter FR, Santa-Maria AR, Valkai S, Vastag M, Neuhaus W, Brodin B, Dér A, Deli MA. Transendothelial Electrical Resistance Measurement across the Blood-Brain Barrier: A Critical Review of Methods. MICROMACHINES 2021; 12:mi12060685. [PMID: 34208338 PMCID: PMC8231150 DOI: 10.3390/mi12060685] [Citation(s) in RCA: 58] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/13/2021] [Revised: 06/04/2021] [Accepted: 06/08/2021] [Indexed: 01/25/2023]
Abstract
The blood–brain barrier (BBB) represents the tightest endothelial barrier within the cardiovascular system characterized by very low ionic permeability. Our aim was to describe the setups, electrodes, and instruments to measure electrical resistance across brain microvessels and culture models of the BBB, as well as critically assess the influence of often neglected physical and technical parameters such as temperature, viscosity, current density generated by different electrode types, surface size, circumference, and porosity of the culture insert membrane. We demonstrate that these physical and technical parameters greatly influence the measurement of transendothelial electrical resistance/resistivity (TEER) across BBB culture models resulting in severalfold differences in TEER values of the same biological model, especially in the low-TEER range. We show that elevated culture medium viscosity significantly increases, while higher membrane porosity decreases TEER values. TEER data measured by chopstick electrodes can be threefold higher than values measured by chamber electrodes due to different electrode size and geometry, resulting in current distribution inhomogeneity. An additional shunt resistance at the circumference of culture inserts results in lower TEER values. A detailed description of setups and technical parameters is crucial for the correct interpretation and comparison of TEER values of BBB models.
Collapse
Affiliation(s)
- Judit P. Vigh
- Institute of Biophysics, Biological Research Centre, 6726 Szeged, Hungary; (J.P.V.); (A.K.); (F.R.W.); (A.R.S.-M.); (S.V.)
- Doctoral School of Biology, University of Szeged, 6720 Szeged, Hungary
| | - András Kincses
- Institute of Biophysics, Biological Research Centre, 6726 Szeged, Hungary; (J.P.V.); (A.K.); (F.R.W.); (A.R.S.-M.); (S.V.)
| | - Burak Ozgür
- Department of Pharmacy, University of Copenhagen, 2100 Copenhagen, Denmark; (B.O.); (B.B.)
| | - Fruzsina R. Walter
- Institute of Biophysics, Biological Research Centre, 6726 Szeged, Hungary; (J.P.V.); (A.K.); (F.R.W.); (A.R.S.-M.); (S.V.)
| | - Ana Raquel Santa-Maria
- Institute of Biophysics, Biological Research Centre, 6726 Szeged, Hungary; (J.P.V.); (A.K.); (F.R.W.); (A.R.S.-M.); (S.V.)
| | - Sándor Valkai
- Institute of Biophysics, Biological Research Centre, 6726 Szeged, Hungary; (J.P.V.); (A.K.); (F.R.W.); (A.R.S.-M.); (S.V.)
| | - Mónika Vastag
- In Vitro Metabolism Research, Division of Pharmacology and Drug Safety, Gedeon Richter Plc., 1103 Budapest, Hungary;
| | - Winfried Neuhaus
- Center for Health and Bioresources, Competence Unit Molecular Diagnostics, AIT—Austrian Institute of Technology GmbH, 1210 Vienna, Austria;
| | - Birger Brodin
- Department of Pharmacy, University of Copenhagen, 2100 Copenhagen, Denmark; (B.O.); (B.B.)
| | - András Dér
- Institute of Biophysics, Biological Research Centre, 6726 Szeged, Hungary; (J.P.V.); (A.K.); (F.R.W.); (A.R.S.-M.); (S.V.)
- Correspondence: (A.D.); (M.A.D.)
| | - Mária A. Deli
- Institute of Biophysics, Biological Research Centre, 6726 Szeged, Hungary; (J.P.V.); (A.K.); (F.R.W.); (A.R.S.-M.); (S.V.)
- Correspondence: (A.D.); (M.A.D.)
| |
Collapse
|