1
|
Al-Majdoub ZM, Cheong J, Mizuno K, Hogan J, De Bruyn T, Kanta A, Guo J, Hop CECA, Zientek M, Galetin A, Ogungbenro K, Rostami-Hodjegan A, Barber J. Transporter expressions as part of required scaling factor to support in vitro in vivo extrapolation for blood-brain barrier drug permeability. Eur J Pharm Sci 2025; 209:107022. [PMID: 39826620 DOI: 10.1016/j.ejps.2025.107022] [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: 08/14/2024] [Revised: 01/13/2025] [Accepted: 01/16/2025] [Indexed: 01/22/2025]
Abstract
Access of drugs to the central nervous system is limited by the blood-brain barrier, and this in turn affects drug efficacy/toxicity. To date, most drug discovery optimization paradigms have relied heavily on in vitro transporter assays and preclinical species pharmacokinetic evaluation to provide a qualitative assessment of human brain penetration. Because of the lack of human brain pharmacokinetic data, mechanistic models for preclinical species, combined with in vitro and in silico data, are useful for translation to human. These models require transporter expression data to be measured in both in vitro and in vivo systems. The purpose of this work was to quantify transporter expression and generate scaling factors (SFs) to enable in vitro in vivo extrapolation (IVIVE) of transporter-mediated processes and to support the development of PBPK model of the brain in rats. SF represents the ratio of abundance of the relevant transporter in the tissue relative to transporter expressing cells. Using quantitative proteomics with QconCAT technology, the expression of human and rat P-gp (ABCB1/Abcb1) and BCRP/Bcrp (ABCG2/Abcg2) was measured in rat brain microvessels, mock and transfected cell lines including, Madin-Darby Canine Kidney I (MDCK I), Madin-Darby Canine Kidney II (MDCK II) and Porcine Kidney epithelial cells (LLC-PK1). P-gp expression ranged from 32 to 71 pmol/mg in rat brain microvessels, exceeding literature values of 14.1-25.2 pmol/mg protein. Conversely, Bcrp expression ranged between 0.02-0.27 pmol/mg protein lower than the literature range (2-6.2 pmol/mg protein). P-gp expression in MDCK I and LLC-PK1 cells transfected with rat Mdr1a was similar (within 1.5-fold) as was human P-gp expression in MDR1 transfected LLC-PK1 and MDCK II cells. The generated SFs were 34.4 and 50.4 for brain P-gp (depending on the cell line used) and 0.53 for brain Bcrp. Endogenous P-gp transporter was detected in MDCK II cell lines when protein expression was measured using a surrogate peptide that was shared across species. The current work provides a framework for proteomics-informed translation of in vitro P-gp and BCRP-related kinetics of drugs and supports the development of PBPK models to predict drug disposition in the brain.
Collapse
Affiliation(s)
| | | | | | | | | | - Anne Kanta
- Takeda Pharmaceuticals Limited, San Diego, CA, USA
| | | | | | - Mike Zientek
- Takeda Pharmaceuticals Limited, San Diego, CA, USA
| | - Aleksandra Galetin
- Centre for Applied Pharmacokinetic Research, University of Manchester, UK
| | - Kayode Ogungbenro
- Centre for Applied Pharmacokinetic Research, University of Manchester, UK
| | - Amin Rostami-Hodjegan
- Centre for Applied Pharmacokinetic Research, University of Manchester, UK; Certara UK Limited, Simcyp Division, Sheffield, UK
| | - Jill Barber
- Centre for Applied Pharmacokinetic Research, University of Manchester, UK
| |
Collapse
|
2
|
Vinh T, Le PH, Nguyen BP, Nguyen-Vo TH. Masked graph transformer for blood-brain barrier permeability prediction. J Mol Biol 2025; 437:168983. [PMID: 39929372 DOI: 10.1016/j.jmb.2025.168983] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Revised: 01/17/2025] [Accepted: 02/02/2025] [Indexed: 02/22/2025]
Abstract
The blood-brain barrier (BBB) is a highly protective structure that strictly regulates the passage of molecules, ensuring the central nervous system remains free from harmful chemicals and maintains brain homeostasis. Since most compounds cannot easily cross the BBB, assessing the blood-brain barrier permeability (BBBP) of drug candidates is critical in drug discovery. While several computational methods have been developed to screen BBBP with promising results, these approaches have limitations that affect their predictive power. In this study, we constructed classification models for screening the BBBP of molecules. Our models were trained with chemical data featurized by a Masked Graph Transformer-based Pretrained (MGTP) encoder. The molecular encoder was designed to generate molecular features for various downstream tasks. The training of the MGTP encoder was guided by masked attention-based learning, improving the model's generalization in encoding molecular structures. The results showed that classification models developed using MGTP features had outperformed those using other representations in 6 out of 8 cases, demonstrating the effectiveness of the proposed encoder. Also, chemical diversity analysis confirmed the encoder's ability to effectively distinguish between different classes of molecules.
Collapse
Affiliation(s)
- Tuan Vinh
- Department of Chemistry, Emory University, 201 Dowman Drive, Atlanta, GA 30322-1007, United States.
| | - Phuc H Le
- Faculty of Information Technology, Ho Chi Minh City Open University, 97 Vo Van Tan, District 3, Ho Chi Minh City 70000, Viet Nam.
| | - Binh P Nguyen
- Faculty of Information Technology, Ho Chi Minh City Open University, 97 Vo Van Tan, District 3, Ho Chi Minh City 70000, Viet Nam; Victoria University of Wellington, Kelburn Parade, Wellington 6012, New Zealand.
| | - Thanh-Hoang Nguyen-Vo
- Faculty of Information Technology, Ho Chi Minh City Open University, 97 Vo Van Tan, District 3, Ho Chi Minh City 70000, Viet Nam.
| |
Collapse
|
3
|
Charernsriwilaiwat N, Thaitrong R, Plianwong S, Opanasopit P, Songprakhon P, Subongkot T. Development and Transportation Pathway Evaluation of Liposomes with Bile Acids for Enhancing the Blood-Brain Barrier Penetration of Methotrexate. Pharmaceutics 2025; 17:269. [PMID: 40006636 PMCID: PMC11859986 DOI: 10.3390/pharmaceutics17020269] [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: 01/16/2025] [Revised: 01/31/2025] [Accepted: 02/12/2025] [Indexed: 02/27/2025] Open
Abstract
Background/Objectives: The purpose of this study was to create bile acid-containing liposomes to improve methotrexate blood-brain barrier penetration and to assess the liposome transportation mechanism across the blood-brain barrier. Methods: The improvement of liposome penetration was investigated utilizing human brain microvascular endothelial cells in an in vitro blood-brain barrier model. Using confocal laser scanning microscopy (CLSM) and flow cytometry, liposomes were labeled with fluorescent phospholipids to facilitate their passage across the blood-brain barrier. Results: The produced liposomes with bile acid exhibited a negative surface charge and an average particle size of between 30 and 148 nm. According to an in vitro blood-brain barrier penetration study, the methotrexate penetration was increased by liposomes containing 1% glycocholic acid but not by liposomes containing taurocholic acid. For transport pathway evaluation across the blood-brain barrier of these liposomes, CLSM revealed that fluorescent liposomes were present inside cells treated with specific endocytosis inhibitors, indicating that the cellular internalization of the particles was not involved in endocytosis. Conclusions: Liposomes supplemented with 1% glycocholic acid could enhance the penetration of methotrexate across the blood-brain barrier, while taurocholic acid could not. The transport of liposomes with 1% glycocholic acid across the blood-brain barrier occurs via the transcellular pathway through which it penetrates cells. In contrast, the paracellular pathway was a minor pathway.
Collapse
Affiliation(s)
- Natthan Charernsriwilaiwat
- Department of Pharmaceutical Technology, Faculty of Pharmaceutical Sciences, Burapha University, Chonburi 20131, Thailand; (N.C.); (R.T.)
| | - Rattanan Thaitrong
- Department of Pharmaceutical Technology, Faculty of Pharmaceutical Sciences, Burapha University, Chonburi 20131, Thailand; (N.C.); (R.T.)
| | - Samarwadee Plianwong
- Department of Pharmaceutical Chemistry, Faculty of Pharmaceutical Sciences, Burapha University, Chonburi 20131, Thailand;
| | - Praneet Opanasopit
- Department of Industrial Pharmacy, Faculty of Pharmacy, Silpakorn University, Nakhon Pathom 73000, Thailand;
| | - Pucharee Songprakhon
- Division of Molecular Medicine, Research Department, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand;
| | - Thirapit Subongkot
- Department of Pharmaceutical Technology, Faculty of Pharmaceutical Sciences, Burapha University, Chonburi 20131, Thailand; (N.C.); (R.T.)
| |
Collapse
|
4
|
Nogueira Pinto H, Zarekiani P, de Vries HE. Neuroglia and the blood-brain barrier. HANDBOOK OF CLINICAL NEUROLOGY 2025; 209:127-141. [PMID: 40122621 DOI: 10.1016/b978-0-443-19104-6.00014-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/25/2025]
Abstract
The blood-brain barrier (BBB) is a highly dynamic and complex structure, present throughout the brain vasculature, that safeguards the brain against blood-borne insults. Neuroglial cells play a major role in its development, function, and homeostasis of the BBB by establishing intricate interactions via direct cell-cell contacts and paracrine signaling. Astrocytes, pericytes, oligodendrocytes, and microglia, alongside specialized brain endothelial cells, orchestrate key events in the brain in health and disease, which can be partially recapitulated by in vitro and in vivo models for biomedical research. This chapter presents a detailed description of the main cellular and molecular mechanisms that govern the neuroglia-BBB crosstalk and the available models for its investigation, emphasizing the importance of each cell population and the synergistic roles they play in the brain.
Collapse
Affiliation(s)
- Henrique Nogueira Pinto
- Amsterdam UMC location Vrije Universiteit Amsterdam, Department of Molecular Cell Biology and Immunology, Amsterdam, The Netherlands; Amsterdam Neuroscience, Neuroinfection & Inflammation, Amsterdam, The Netherlands
| | - Parand Zarekiani
- Amsterdam UMC location Vrije Universiteit Amsterdam, Department of Molecular Cell Biology and Immunology, Amsterdam, The Netherlands; Amsterdam Neuroscience, Neuroinfection & Inflammation, Amsterdam, The Netherlands; Amsterdam UMC location Vrije Universiteit Amsterdam, Department of Pathology, Amsterdam, The Netherlands; Amsterdam UMC location Vrije Universiteit Amsterdam, Department of Child Neurology, Amsterdam Leukodystrophy Center, Emma Children's Hospital, Amsterdam, The Netherlands
| | - Helga E de Vries
- Amsterdam UMC location Vrije Universiteit Amsterdam, Department of Molecular Cell Biology and Immunology, Amsterdam, The Netherlands; Amsterdam Neuroscience, Neuroinfection & Inflammation, Amsterdam, The Netherlands; MS Center Amsterdam, Amsterdam, The Netherlands.
| |
Collapse
|
5
|
Karcz T, Szczepańska K, Mogilski S, Moroz A, Olejarz-Maciej A, Humphrys LJ, Pockes S, Siwek A, Dubiel K, Staszewski M, Calmels T, Waczyński K, Kieć-Kononowicz K. Guanidine Derivative ADS1017, a Potent Histamine H 3 Receptor Antagonist with Promising Analgesic Activity and Satisfactory Safety Profile. ACS Chem Neurosci 2024; 15:4441-4457. [PMID: 39652796 DOI: 10.1021/acschemneuro.4c00480] [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: 12/19/2024] Open
Abstract
In this study, we selected 12 guanidine derivatives from the previously described ligand library and determined their affinity at histamine H3 and H4 receptors (H3R and H4R, respectively). Moreover, we also checked their intrinsic activity toward H3R and muscarinic M1, M2, and M4 receptors (M1R, M2R, and M4R, respectively). Since ADS1017 has been proved to be the most selective and highly potent H3 antagonist in our series, we chose it as the lead structure for further biological evaluation. To extend the study of its in vivo efficacy, we proposed an alternative synthetic route that resulted in an increased yield. Interestingly, ADS1017 showed a broad spectrum of analgesic activity in both nociceptive and neuropathic pain models. Finally, as a result of comprehensive analysis of its off-target activity and ADMETox parameters, we confirmed the moderate selectivity of ADS1017 and its promising drug-like properties.
Collapse
Affiliation(s)
- Tadeusz Karcz
- Department of Technology and Biotechnology of Drugs, Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna 9, Kraków 30-688, Poland
| | - Katarzyna Szczepańska
- Department of Medicinal Chemistry, Maj Institute of Pharmacology, Polish Academy of Sciences, Smętna 12, Kraków 31-343, Poland
| | - Szczepan Mogilski
- Department of Pharmacodynamics, Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna 9, Kraków 30-688, Poland
| | - Aleksandra Moroz
- R&D Centre, Celon Pharma S.A., Marymoncka 15, Kazuń Nowy 05-152, Poland
| | - Agnieszka Olejarz-Maciej
- Department of Technology and Biotechnology of Drugs, Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna 9, Kraków 30-688, Poland
| | - Laura J Humphrys
- Institute of Pharmacy, Faculty of Chemistry and Pharmacy, University of Regensburg, Universitätsstraße 31, Regensburg D-93053, Germany
| | - Steffen Pockes
- Institute of Pharmacy, Faculty of Chemistry and Pharmacy, University of Regensburg, Universitätsstraße 31, Regensburg D-93053, Germany
| | - Agata Siwek
- Department of Pharmacobiology, Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna 9, Kraków 30-688, Poland
| | - Krzysztof Dubiel
- R&D Centre, Celon Pharma S.A., Marymoncka 15, Kazuń Nowy 05-152, Poland
| | - Marek Staszewski
- Department of Synthesis and Technology of Drugs, Medical University of Lodz, Muszyńskiego 1, Łódź 90-151, Poland
| | - Thierry Calmels
- Bioprojet-Biotech, 4rue du Chesnay Beauregard, Saint-Gregoire Cedex 35762, France
| | - Krzysztof Waczyński
- Department of Synthesis and Technology of Drugs, Medical University of Lodz, Muszyńskiego 1, Łódź 90-151, Poland
| | - Katarzyna Kieć-Kononowicz
- Department of Technology and Biotechnology of Drugs, Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna 9, Kraków 30-688, Poland
| |
Collapse
|
6
|
Ruczyński J, Prochera K, Kaźmierczak N, Kosznik-Kwaśnicka K, Piechowicz L, Mucha P, Rekowski P. New Conjugates of Vancomycin with Cell-Penetrating Peptides-Synthesis, Antimicrobial Activity, Cytotoxicity, and BBB Permeability Studies. Molecules 2024; 29:5519. [PMID: 39683678 DOI: 10.3390/molecules29235519] [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: 10/06/2024] [Revised: 11/17/2024] [Accepted: 11/20/2024] [Indexed: 12/18/2024] Open
Abstract
Vancomycin (Van) is a glycopeptide antibiotic commonly used as a last resort for treating life-threatening infections caused by multidrug-resistant bacterial strains, such as Staphylococcus aureus and Enterococcus spp. However, its effectiveness is currently limited due to the rapidly increasing number of drug-resistant clinical strains and its inherent cytotoxicity and poor penetration into cells and specific regions of the body, such as the brain. One of the most promising strategies to enhance its efficacy appears to be the covalent attachment of cell-penetrating peptides (CPPs) to the Van structure. In this study, a series of vancomycin conjugates with CPPs-such as TP10, Tat (47-57), PTD4, and Arg9-were designed and synthesized. These conjugates were tested for antimicrobial activity against four reference strains (Staphylococcus aureus, Enterococcus faecalis, Escherichia coli, and Pseudomonas aeruginosa) and two clinical drug-resistant strains: methicillin-resistant S. aureus and vancomycin-resistant E. faecium. In addition, cytotoxicity tests (using a human fibroblast cell line) and blood-brain barrier (BBB) permeability tests (using a parallel artificial membrane permeability assay-PAMPA-BBB assay) were conducted for selected compounds. Our research demonstrated that conjugation of Van with CPPs, particularly with Tat (47-57), Arg9, or TP10, significantly enhances its antimicrobial activity against Gram-positive bacteria such as S. aureus and Enterococcus spp., reduces its cytotoxicity, and improves its access to brain tissues. We conclude that these findings provide a strong foundation for the design of novel antimicrobial agents effective in treating infections caused by drug-resistant staphylococcal and enterococcal strains, while also being capable of crossing the BBB.
Collapse
Affiliation(s)
- Jarosław Ruczyński
- Laboratory of Chemistry of Biologically Active Compounds, Faculty of Chemistry, University of Gdańsk, Wita Stwosza 63, 80-308 Gdańsk, Poland
| | - Katarzyna Prochera
- Laboratory of Chemistry of Biologically Active Compounds, Faculty of Chemistry, University of Gdańsk, Wita Stwosza 63, 80-308 Gdańsk, Poland
| | - Natalia Kaźmierczak
- Department of Medical Microbiology, Faculty of Medicine, Medical University of Gdańsk, Dębowa 25, 80-204 Gdańsk, Poland
| | - Katarzyna Kosznik-Kwaśnicka
- Department of Medical Microbiology, Faculty of Medicine, Medical University of Gdańsk, Dębowa 25, 80-204 Gdańsk, Poland
| | - Lidia Piechowicz
- Department of Medical Microbiology, Faculty of Medicine, Medical University of Gdańsk, Dębowa 25, 80-204 Gdańsk, Poland
| | - Piotr Mucha
- Laboratory of Chemistry of Biologically Active Compounds, Faculty of Chemistry, University of Gdańsk, Wita Stwosza 63, 80-308 Gdańsk, Poland
| | - Piotr Rekowski
- Laboratory of Chemistry of Biologically Active Compounds, Faculty of Chemistry, University of Gdańsk, Wita Stwosza 63, 80-308 Gdańsk, Poland
| |
Collapse
|
7
|
Virameteekul S, Lees AJ, Bhidayasiri R. Small Particles, Big Potential: Polymeric Nanoparticles for Drug Delivery in Parkinson's Disease. Mov Disord 2024; 39:1922-1937. [PMID: 39077831 DOI: 10.1002/mds.29939] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Revised: 07/01/2024] [Accepted: 07/03/2024] [Indexed: 07/31/2024] Open
Abstract
Despite the availability of a number of efficacious treatments for Parkinson's disease, their limitations and drawbacks, particularly related to low brain bioavailability and associated side effects, emphasize the need for alternative and more effective therapeutic approaches. Nanomedicine, the application of nanotechnology in medicine, has received considerable interest in recent years as a method of effectively delivering potentially therapeutic molecules to the brain. In particular, polymeric nanoparticles, constructed from biodegradable polymer, have shown great promise in enhancing therapeutic efficacy, reducing toxicity, and ensuring targeted delivery. However, their clinical translation remains a considerable challenge. This article reviews recent in vitro and in vivo studies using polymeric nanoparticles as drug and gene delivery systems for Parkinson's disease with their challenges and future directions. We are also particularly interested in the technical properties, mechanism, drugs release patterns, and delivery strategies to overcome the blood-brain barrier. © 2024 International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- Sasivimol Virameteekul
- Department of Biochemistry, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
- Reta Lila Weston Institute of Neurological Studies, UCL Queen Square Institute of Neurology, London, UK
- Chulalongkorn Centre of Excellence for Parkinson's Disease and Related Disorders, Department of Medicine, Faculty of Medicine, Chulalongkorn University and King Chulalongkorn Memorial Hospital, Thai Red Cross Society, Bangkok, Thailand
| | - Andrew J Lees
- Reta Lila Weston Institute of Neurological Studies, UCL Queen Square Institute of Neurology, London, UK
| | - Roongroj Bhidayasiri
- Chulalongkorn Centre of Excellence for Parkinson's Disease and Related Disorders, Department of Medicine, Faculty of Medicine, Chulalongkorn University and King Chulalongkorn Memorial Hospital, Thai Red Cross Society, Bangkok, Thailand
- The Academy of Science, The Royal Society of Thailand, Bangkok, Thailand
| |
Collapse
|
8
|
Nakayama-Kitamura K, Shigemoto-Mogami Y, Piantino M, Naka Y, Yamada A, Kitano S, Furihata T, Matsusaki M, Sato K. Collagen I Microfiber Promotes Brain Capillary Network Formation in Three-Dimensional Blood-Brain Barrier Microphysiological Systems. Biomedicines 2024; 12:2500. [PMID: 39595066 PMCID: PMC11591679 DOI: 10.3390/biomedicines12112500] [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: 09/25/2024] [Revised: 10/20/2024] [Accepted: 10/24/2024] [Indexed: 11/28/2024] Open
Abstract
BACKGROUND The blood-brain barrier (BBB) strictly regulates the penetration of substances into the brain, which, although important for maintaining brain homeostasis, may delay drug development because of the difficulties in predicting pharmacokinetics/pharmacodynamics (PKPD), toxicokinetics/toxicodynamics (TKTD), toxicity, safety, and efficacy in the central nervous system (CNS). Moreover, BBB functional proteins show species differences; therefore, humanized in vitro BBB models are urgently needed to improve the predictability of preclinical studies. Recently, international trends in the 3Rs in animal experiments and the approval of the FDA Modernization Act 2.0 have accelerated the application of microphysiological systems (MPSs) in preclinical studies, and in vitro BBB models have become synonymous with BBB-MPSs. Recently, we developed an industrialized humanized BBB-MPS, BBB-NET. In our previous report, we reproduced transferrin receptor (TfR)-mediated transcytosis with high efficiency and robustness, using hydrogels including fibrin and collagen I microfibers (CMFs). METHODS We investigated how adding CMFs to the fibrin gel benefits BBB-NETs. RESULTS We showed that CMFs accelerate capillary network formation and maturation by promoting astrocyte (AC) survival, and clarified that integrin β1 is involved in the mechanism of CMFs. CONCLUSIONS Our data suggest that the quality control (QC) of CMFs is important for ensuring the stable production of BBB-NETs.
Collapse
Affiliation(s)
- Kimiko Nakayama-Kitamura
- Laboratory of Neuropharmacology, Division of Pharmacology, National Institute of Health Sciences, 3-25-26, Tonomachi, Kawasaki-ku, Kawasaki City 210-9501, Kanagawa, Japan; (K.N.-K.); (Y.S.-M.)
| | - Yukari Shigemoto-Mogami
- Laboratory of Neuropharmacology, Division of Pharmacology, National Institute of Health Sciences, 3-25-26, Tonomachi, Kawasaki-ku, Kawasaki City 210-9501, Kanagawa, Japan; (K.N.-K.); (Y.S.-M.)
| | - Marie Piantino
- Joint Research Laboratory for Social Implementation of Cultured Meat, Osaka University, 2-1 Yamadaoka, Suita 565-0871, Osaka, Japan; (M.P.); (M.M.)
| | - Yasuhiro Naka
- Department of Applied Chemistry, Graduate School of Engineering, Osaka University, 2-1 Yamadaoka, Suita 565-0871, Osaka, Japan;
| | - Asuka Yamada
- TOPPAN Holdings Inc., TOPPAN Technical Research Institute, 4-2 Takanodaiminami, Sugitomachi, Saitama 345-8508, Saitama, Japan; (A.Y.); (S.K.)
- Joint Research Laboratory (TOPPAN) for Advanced Cell Regulatory Chemistry, Graduate School of Engineering, Osaka University, Yamadaoka, Suita 565-0871, Osaka, Japan
| | - Shiro Kitano
- TOPPAN Holdings Inc., TOPPAN Technical Research Institute, 4-2 Takanodaiminami, Sugitomachi, Saitama 345-8508, Saitama, Japan; (A.Y.); (S.K.)
- Joint Research Laboratory (TOPPAN) for Advanced Cell Regulatory Chemistry, Graduate School of Engineering, Osaka University, Yamadaoka, Suita 565-0871, Osaka, Japan
| | - Tomomi Furihata
- Laboratory of Advanced Drug Developmen Sciences, School of Pharmacy, Tokyo University of Pharmacy and Life Sciences, 1432-1 Horinouchi, Hachioji 192-0392, Tokyo, Japan;
| | - Michiya Matsusaki
- Joint Research Laboratory for Social Implementation of Cultured Meat, Osaka University, 2-1 Yamadaoka, Suita 565-0871, Osaka, Japan; (M.P.); (M.M.)
- Department of Applied Chemistry, Graduate School of Engineering, Osaka University, 2-1 Yamadaoka, Suita 565-0871, Osaka, Japan;
| | - Kaoru Sato
- Laboratory of Neuropharmacology, Division of Pharmacology, National Institute of Health Sciences, 3-25-26, Tonomachi, Kawasaki-ku, Kawasaki City 210-9501, Kanagawa, Japan; (K.N.-K.); (Y.S.-M.)
| |
Collapse
|
9
|
Wang F, Qi L, Zhang Z, Duan H, Wang Y, Zhang K, Li J. The Mechanism and Latest Research Progress of Blood-Brain Barrier Breakthrough. Biomedicines 2024; 12:2302. [PMID: 39457617 PMCID: PMC11504064 DOI: 10.3390/biomedicines12102302] [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: 08/23/2024] [Revised: 09/19/2024] [Accepted: 10/01/2024] [Indexed: 10/28/2024] Open
Abstract
The bloodstream and the central nervous system (CNS) are separated by the blood-brain barrier (BBB), an intricate network of blood vessels. Its main role is to regulate the environment within the brain. The primary obstacle for drugs to enter the CNS is the low permeability of the BBB, presenting a significant hurdle in treating brain disorders. In recent years, significant advancements have been made in researching methods to breach the BBB. However, understanding how to penetrate the BBB is essential for researching drug delivery techniques. Therefore, this article reviews the methods and mechanisms for breaking through the BBB, as well as the current research progress on this mechanism.
Collapse
Affiliation(s)
- Fei Wang
- School of Materials Science and Engineering, Zhengzhou University, Zhengzhou 450001, China; (F.W.); (L.Q.); (Z.Z.); (H.D.); (Y.W.)
| | - Liujie Qi
- School of Materials Science and Engineering, Zhengzhou University, Zhengzhou 450001, China; (F.W.); (L.Q.); (Z.Z.); (H.D.); (Y.W.)
| | - Zhongna Zhang
- School of Materials Science and Engineering, Zhengzhou University, Zhengzhou 450001, China; (F.W.); (L.Q.); (Z.Z.); (H.D.); (Y.W.)
| | - Huimin Duan
- School of Materials Science and Engineering, Zhengzhou University, Zhengzhou 450001, China; (F.W.); (L.Q.); (Z.Z.); (H.D.); (Y.W.)
| | - Yanchao Wang
- School of Materials Science and Engineering, Zhengzhou University, Zhengzhou 450001, China; (F.W.); (L.Q.); (Z.Z.); (H.D.); (Y.W.)
| | - Kun Zhang
- School of Life Science, Zhengzhou University, Zhengzhou 450001, China
| | - Jingan Li
- School of Materials Science and Engineering, Zhengzhou University, Zhengzhou 450001, China; (F.W.); (L.Q.); (Z.Z.); (H.D.); (Y.W.)
| |
Collapse
|
10
|
Song Q, Li J, Li T, Li H. Nanomaterials that Aid in the Diagnosis and Treatment of Alzheimer's Disease, Resolving Blood-Brain Barrier Crossing Ability. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2403473. [PMID: 39101248 PMCID: PMC11481234 DOI: 10.1002/advs.202403473] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Revised: 07/04/2024] [Indexed: 08/06/2024]
Abstract
As a form of dementia, Alzheimer's disease (AD) suffers from no efficacious cure, yet AD treatment is still imperative, as it ameliorates the symptoms or prevents it from deteriorating or maintains the current status to the longest extent. The human brain is the most sensitive and complex organ in the body, which is protected by the blood-brain barrier (BBB). This yet induces the difficulty in curing AD as the drugs or nanomaterials that are much inhibited from reaching the lesion site. Thus, BBB crossing capability of drug delivery system remains a significant challenge in the development of neurological therapeutics. Fortunately, nano-enabled delivery systems possess promising potential to achieve multifunctional diagnostics/therapeutics against various targets of AD owing to their intriguing advantages of nanocarriers, including easy multifunctionalization on surfaces, high surface-to-volume ratio with large payloads, and potential ability to cross the BBB, making them capable of conquering the limitations of conventional drug candidates. This review, which focuses on the BBB crossing ability of the multifunctional nanomaterials in AD diagnosis and treatment, will provide an insightful vision that is conducive to the development of AD-related nanomaterials.
Collapse
Affiliation(s)
- Qingting Song
- Department of ChemistryThe Chinese University of Hong KongHong KongChina
| | - Junyou Li
- Department of ChemistryThe Chinese University of Hong KongHong KongChina
| | - Ting Li
- Department of ChemistryThe Chinese University of Hong KongHong KongChina
| | - Hung‐Wing Li
- Department of ChemistryThe Chinese University of Hong KongHong KongChina
| |
Collapse
|
11
|
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
|
12
|
Danz K, Fleddermann J, Koch M, Fecioru E, Maahs L, Kinsinger N, Krämer J, Kraegeloh A, Wagner S. Evaluation of the Transport and Binding of Dopamine-Loaded PLGA Nanoparticles for the Treatment of Parkinson's Disease Using In Vitro Model Systems. Pharmaceutics 2024; 16:571. [PMID: 38794233 PMCID: PMC11125169 DOI: 10.3390/pharmaceutics16050571] [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: 02/21/2024] [Revised: 04/08/2024] [Accepted: 04/12/2024] [Indexed: 05/26/2024] Open
Abstract
The treatment of Parkinson's disease has been moving into the focus of pharmaceutical development. Yet, the necessity for reliable model systems in the development phase has made research challenging and in vivo models necessary. We have established reliable, reproducible in vitro model systems to evaluate the binding and transport of dopamine-loaded PLGA nanoparticles for the treatment of Parkinson's disease and put the results in context with comparable in vivo results. The in vitro models have provided similar results concerning the usability of the investigated nanoparticles as the previously used in vivo models and thus provide a good alternative in line with the 3R principles in pharmaceutical research.
Collapse
Affiliation(s)
- Karin Danz
- Fraunhofer Institute for Biomedical Engineering IBMT, Joseph-von-Fraunhofer-Weg 1, 66280 Sulzbach, Germany
| | - Jana Fleddermann
- INM—Leibniz Institute for New Materials, Campus D2 2, 66123 Saarbrücken, Germany (M.K.); (A.K.)
| | - Marcus Koch
- INM—Leibniz Institute for New Materials, Campus D2 2, 66123 Saarbrücken, Germany (M.K.); (A.K.)
| | - Elena Fecioru
- Eurofins PHAST Development GmbH & Co. KG, Byk-Gulden-Str. 2, 78467 Konstanz, Germany
| | - Lorenz Maahs
- Pharmacelsus GmbH, Science Park 2, 66123 Saarbrücken, Germany; (L.M.); (N.K.)
| | - Nicole Kinsinger
- Pharmacelsus GmbH, Science Park 2, 66123 Saarbrücken, Germany; (L.M.); (N.K.)
| | | | - Annette Kraegeloh
- INM—Leibniz Institute for New Materials, Campus D2 2, 66123 Saarbrücken, Germany (M.K.); (A.K.)
| | - Sylvia Wagner
- Fraunhofer Institute for Biomedical Engineering IBMT, Joseph-von-Fraunhofer-Weg 1, 66280 Sulzbach, Germany
| |
Collapse
|
13
|
Jovanović M, Radan M, Čarapić M, Filipović N, Nikolic K, Crevar M. Application of parallel artificial membrane permeability assay technique and chemometric modeling for blood-brain barrier permeability prediction of protein kinase inhibitors. Future Med Chem 2024; 16:873-885. [PMID: 38639375 PMCID: PMC11373572 DOI: 10.4155/fmc-2023-0390] [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: 12/18/2023] [Accepted: 03/26/2024] [Indexed: 04/20/2024] Open
Abstract
Aim: This study aims to investigate the passive diffusion of protein kinase inhibitors through the blood-brain barrier (BBB) and to develop a model for their permeability prediction. Materials & methods: We used the parallel artificial membrane permeability assay to obtain logPe values of each of 34 compounds and calculated descriptors for these structures to perform quantitative structure-property relationship modeling, creating different regression models. Results: The logPe values have been calculated for all 34 compounds. Support vector machine regression was considered the most reliable, and CATS2D_09_DA, CATS2D_04_AA, B04[N-S] and F07[C-N] descriptors were identified as the most influential to passive BBB permeability. Conclusion: The quantitative structure-property relationship-support vector machine regression model that has been generated can serve as an efficient method for preliminary screening of BBB permeability of new analogs.
Collapse
Affiliation(s)
- Milan Jovanović
- University of Belgrade – Faculty of Pharmacy, Department of Pharmaceutical Chemistry, Vojvode Stepe 450, P.O.Box 146, 11221, Belgrade, Serbia
- University of Belgrade – “VINCA” Institute of Nuclear Sciences – National Institute of the Republic of Serbia, Department of Molecular Biology & Endocrinology, Mike Petrovica Alasa 12-14, Vinca, 11351, Belgrade, Serbia
| | - Milica Radan
- Institute for Medicinal Plant Research “Dr. Josif Pančić”, Tadeuša Košćuška 1, Belgrade, 11000, Serbia
| | - Marija Čarapić
- Medicines & Medical Devices Agency of Serbia, Vojvode Stepe 458, 11000, Belgrade, Serbia
| | - Nenad Filipović
- University of Belgrade – Faculty of Agriculture, Nemanjina 6, 11000, Belgrade, Serbia
| | - Katarina Nikolic
- University of Belgrade – Faculty of Pharmacy, Department of Pharmaceutical Chemistry, Vojvode Stepe 450, P.O.Box 146, 11221, Belgrade, Serbia
| | - Milkica Crevar
- University of Belgrade – Faculty of Pharmacy, Department of Pharmaceutical Chemistry, Vojvode Stepe 450, P.O.Box 146, 11221, Belgrade, Serbia
| |
Collapse
|
14
|
Lin Y, Gan L, Ren L, Ma C, Dai M, Qian K, Ye Q, Lin X. Acupuncture with specific mode electro-stimulation effectively and transiently opens the BBB through Shh signaling pathway. Neuroreport 2023; 34:873-886. [PMID: 37942738 DOI: 10.1097/wnr.0000000000001970] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2023]
Abstract
To explore a new method that patients with brain diseases such as stroke sequelae are hindered by blood-brain barrier (BBB) in clinical treatment. Research preliminarily found that acupuncture with specific mode electro-stimulation (EA) to open BBB-assisted drug delivery may be is an effective means to improve the clinical efficacy of brain disease patients. So here we further explore the features and mechanism. Middle cerebral artery occlusion/R recovery rats were employed as the animal model. Laser Doppler monitoring cerebral blood flow decreased to 45 ± 10% of the baseline value as modeling criteria and TTC staining observed infarcted areas of brain tissue. The permeability of FITC-Dextran and EB in the frontal lobe of rats was observed by microscope. After that, Western blot and Immunofluorescence staining for the detection of the shh and Gli1 signal molecule, Claudin-5 Occludin ZO-1 tight junction (TJ) proteins. EA can open the BBB stably and effectively, and has the characteristics of starting to close soon after the end of EA; EA inhibits the Shh-Gli1 signaling pathway, and downregulates Occludin ZO-1 TJ proteins. These results suggest that EA is safe and reversible in opening the BBB, and its mechanism is related to the inhibition of Shh signaling pathway to down-regulate the expression of TJ proteins.
Collapse
Affiliation(s)
- Yubo Lin
- Department of Neurobiology and Acupuncture Research, The Third Clinical Medical College, Zhejiang Chinese Medical University, Key Laboratory of Acupuncture and Neurology of Zhejiang Province, Hangzhou, China
| | | | | | | | | | | | | | | |
Collapse
|
15
|
Zhao Y, Ma J, Cui Y, Lin H. A method to isolate human dermal microvascular pericytes without the use of magnetic beads sorting in vitro. Tissue Cell 2023; 84:102171. [PMID: 37480631 DOI: 10.1016/j.tice.2023.102171] [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: 04/17/2023] [Revised: 07/08/2023] [Accepted: 07/18/2023] [Indexed: 07/24/2023]
Abstract
Human dermal microvascular pericytes (HDMPCs) are a critical component of the skin flap microvasculature and play a role in regulating flap blood flow and integrity. Pericytes were isolated mostly via magnetic bead sorting in the published literature. In this study, we discuss in detail how to separate and concentrate pericytes from human facial flaps using enzyme digestion and differential adherence instead of magnetic bead sorting. Cultured HDMPCs were seen to have well-spread irregular edges, with most cells having two longitudinal pericytic processes. The phalloidin staining revealed that HDMPCs had prominent stress fibers, and the nucleus deviated to the side that interacted with the neighboring pericytic processes. Flow cytometry analysis showed that the positive rates of NG2 in the first and second passages were 91.2% ± 0.7% and 98.2% ± 0.1% separately. And the immunofluorescence and western blot results demonstrated a positive expression of α smooth muscle actin (αSMA), platelet-derived growth factor receptor β (PDGFRβ), and NG-2, while the endothelial cell marker CD31 was negatively expressed. In summary, we established a straightforward methodology for selectively isolating and identifying HDMPCs as well as generating high-purity cell cultures in vitro without the use of magnetic bead sorting.
Collapse
Affiliation(s)
- Yinhua Zhao
- Plastic and Reconstructive Surgery, Beijing Anzhen Hospital, Capital Medical University, 2 Anzhen road, Chaoyang district, Beijing 100029, China
| | - Jiaxing Ma
- Plastic and Reconstructive Surgery, Beijing Anzhen Hospital, Capital Medical University, 2 Anzhen road, Chaoyang district, Beijing 100029, China
| | - Yue Cui
- Plastic and Reconstructive Surgery, Beijing Anzhen Hospital, Capital Medical University, 2 Anzhen road, Chaoyang district, Beijing 100029, China
| | - Huang Lin
- Plastic and Reconstructive Surgery, Beijing Anzhen Hospital, Capital Medical University, 2 Anzhen road, Chaoyang district, Beijing 100029, China.
| |
Collapse
|
16
|
Ding Z, Fan X, Zhang Y, Yao M, Wang G, Dong Y, Liu J, Song W. The glymphatic system: a new perspective on brain diseases. Front Aging Neurosci 2023; 15:1179988. [PMID: 37396658 PMCID: PMC10308198 DOI: 10.3389/fnagi.2023.1179988] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2023] [Accepted: 05/29/2023] [Indexed: 07/04/2023] Open
Abstract
The glymphatic system is a brain-wide perivascular pathway driven by aquaporin-4 on the endfeet of astrocytes, which can deliver nutrients and active substances to the brain parenchyma through periarterial cerebrospinal fluid (CSF) influx pathway and remove metabolic wastes through perivenous clearance routes. This paper summarizes the composition, overall fluid flow, solute transport, related diseases, affecting factors, and preclinical research methods of the glymphatic system. In doing so, we aim to provide direction and reference for more relevant researchers in the future.
Collapse
|
17
|
Ledwig V, Reichl S. Isolation and Cultivation of Porcine Endothelial Cells, Pericytes and Astrocytes to Develop an In Vitro Blood-Brain Barrier Model for Drug Permeation Testing. Pharmaceutics 2023; 15:1688. [PMID: 37376136 DOI: 10.3390/pharmaceutics15061688] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Revised: 05/25/2023] [Accepted: 06/02/2023] [Indexed: 06/29/2023] Open
Abstract
The blood-brain barrier (BBB) is the bottleneck in the development of new drugs to reach the brain. Due to the BBB, toxic substances cannot enter the brain, but promising drug candidates also pass the BBB poorly. Suitable in vitro BBB models are therefore of particular importance during the preclinical development process, as they can not only reduce animal testing but also enable new drugs to be developed more quickly. The aim of this study was to isolate cerebral endothelial cells, pericytes, and astrocytes from the porcine brain to produce a primary model of the BBB. Additionally, as primary cells are well suited by their properties but the isolation is complex and better reproducibility with immortalized cells must be ensured, there is a high demand for immortalized cells with suitable properties for use as a BBB model. Thus, isolated primary cells can also serve as the basis for a suitable immortalization technique to generate new cell lines. In this work, cerebral endothelial cells, pericytes, and astrocytes were successfully isolated and expanded using a mechanical/enzymatic method. Furthermore, in a triple coculture model, the cells showed a significant increase in barrier integrity compared with endothelial cell monoculture, as determined by transendothelial electrical resistance measurement and permeation studies using sodium fluorescein. The results demonstrate the opportunity to obtain all three cell types significantly involved in BBB formation from one species, thus providing a suitable tool for testing the permeation properties of new drug candidates. In addition, the protocols are a promising starting point to generate new cell lines of BBB-forming cells as a novel approach for BBB in vitro models.
Collapse
Affiliation(s)
- Verena Ledwig
- Center of Pharmaceutical Engineering (PVZ), Technische Universität Braunschweig, Franz-Liszt-Straße 35A, 38106 Braunschweig, Germany
- Institute of Pharmaceutical Technology and Biopharmaceutics, Technische Universität Braunschweig, Mendelssohnstraße 1, 38106 Braunschweig, Germany
| | - Stephan Reichl
- Center of Pharmaceutical Engineering (PVZ), Technische Universität Braunschweig, Franz-Liszt-Straße 35A, 38106 Braunschweig, Germany
- Institute of Pharmaceutical Technology and Biopharmaceutics, Technische Universität Braunschweig, Mendelssohnstraße 1, 38106 Braunschweig, Germany
| |
Collapse
|
18
|
Li Q, Wong HL, Ip YL, Chu WY, Li MS, Saha C, Shih KC, Chan YK. Current microfluidic platforms for reverse engineering of cornea. Mater Today Bio 2023; 20:100634. [PMID: 37139464 PMCID: PMC10149412 DOI: 10.1016/j.mtbio.2023.100634] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Revised: 04/04/2023] [Accepted: 04/10/2023] [Indexed: 05/05/2023] Open
Abstract
According to the World Health Organization, corneal blindness constitutes 5.1% of global blindness population. Surgical outcomes have been improved significantly in the treatment of corneal blindness. However, corneal transplantation is limited by global shortage of donor tissue, prompting researchers to explore alternative therapies such as novel ocular pharmaceutics to delay corneal disease progression. Animal models are commonly adopted for investigating pharmacokinetics of ocular drugs. However, this approach is limited by physiological differences in the eye between animals and human, ethical issues and poor bench-to-bedside translatability. Cornea-on-a-chip (CoC) microfluidic platforms have gained great attention as one of the advanced in vitro strategies for constructing physiologically representative corneal models. With significant improvements in tissue engineering technology, CoC integrates corneal cells with microfluidics to recapitulate human corneal microenvironment for the study of corneal pathophysiological changes and evaluation of ocular drugs. Such model, in complement to animal studies, can potentially accelerate translational research, in particular the pre-clinical screening of ophthalmic medication, driving clinical treatment advancement for corneal diseases. This review provides an overview of engineered CoC platforms with respect to their merits, applications, and technical challenges. Emerging directions in CoC technology are also proposed for further investigations, to accentuate preclinical obstacles in corneal research.
Collapse
Affiliation(s)
- Qinyu Li
- Department of Ophthalmology, LKS Faculty of Medicine, The University of Hong Kong, 999077, Hong Kong, China
| | - Ho Lam Wong
- Department of Ophthalmology, LKS Faculty of Medicine, The University of Hong Kong, 999077, Hong Kong, China
| | - Yan Lam Ip
- Department of Ophthalmology, LKS Faculty of Medicine, The University of Hong Kong, 999077, Hong Kong, China
| | - Wang Yee Chu
- Department of Ophthalmology, LKS Faculty of Medicine, The University of Hong Kong, 999077, Hong Kong, China
| | - Man Shek Li
- Department of Ophthalmology, LKS Faculty of Medicine, The University of Hong Kong, 999077, Hong Kong, China
| | - Chinmoy Saha
- Department of Ophthalmology, LKS Faculty of Medicine, The University of Hong Kong, 999077, Hong Kong, China
| | - Kendrick Co Shih
- Department of Ophthalmology, LKS Faculty of Medicine, The University of Hong Kong, 999077, Hong Kong, China
| | - Yau Kei Chan
- Department of Ophthalmology, LKS Faculty of Medicine, The University of Hong Kong, 999077, Hong Kong, China
| |
Collapse
|
19
|
Nakayama-Kitamura K, Shigemoto-Mogami Y, Toyoda H, Mihara I, Moriguchi H, Naraoka H, Furihata T, Ishida S, Sato K. Usefulness of a humanized tricellular static transwell blood-brain barrier model as a microphysiological system for drug development applications. - A case study based on the benchmark evaluations of blood-brain barrier microphysiological system. Regen Ther 2023; 22:192-202. [PMID: 36891355 PMCID: PMC9988422 DOI: 10.1016/j.reth.2023.02.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Revised: 01/21/2023] [Accepted: 02/08/2023] [Indexed: 03/06/2023] Open
Abstract
Microphysiological system (MPS), a new technology for in vitro testing platforms, have been acknowledged as a strong tool for drug development. In the central nervous system (CNS), the blood‒brain barrier (BBB) limits the permeation of circulating substances from the blood vessels to the brain, thereby protecting the CNS from circulating xenobiotic compounds. At the same time, the BBB hinders drug development by introducing challenges at various stages, such as pharmacokinetics/pharmacodynamics (PK/PD), safety assessment, and efficacy assessment. To solve these problems, efforts are being made to develop a BBB MPS, particularly of a humanized type. In this study, we suggested minimal essential benchmark items to establish the BBB-likeness of a BBB MPS; these criteria support end users in determining the appropriate range of applications for a candidate BBB MPS. Furthermore, we examined these benchmark items in a two-dimensional (2D) humanized tricellular static transwell BBB MPS, the most conventional design of BBB MPS with human cell lines. Among the benchmark items, the efflux ratios of P-gp and BCRP showed high reproducibility in two independent facilities, while the directional transports meditated through Glut1 or TfR were not confirmed. We have organized the protocols of the experiments described above as standard operating procedures (SOPs). We here provide the SOPs with the flow chart including entire procedure and how to apply each SOP. Our study is important developmental step of BBB MPS towards the social acceptance, which enable end users to check and compare the performance the BBB MPSs.
Collapse
Key Words
- BBB, blood-brain barrier
- BCRP
- BCRP, Breast cancer resistance protein
- Blood‒brain barrier (BBB)
- CNS, central nervous system
- Glut1, Glucose transporter 1
- HASTR, Human astrocytes
- HBMEC, Human brain microvascular endothelial cells
- HBPC, Human brain pericyte
- LC-MS/MS, Liquid chromatography with tandem mass spectrometry
- LY, Lucifer yellow
- MPS, Microphysiological system
- Microphysiological system (MPS)
- P-gp
- P-gp, P-glycoprotein
- TEER, Trans-endothelial electrical resistance
- TfR, Transferrin receptor
Collapse
Affiliation(s)
- Kimiko Nakayama-Kitamura
- Laboratory of Neuropharmacology, Division of Pharmacology, National Institute of Health Science, 3-25-26 Tonomachi, Kawasaki-ku, Kawasaki City, Kanagawa, Japan
| | - Yukari Shigemoto-Mogami
- Laboratory of Neuropharmacology, Division of Pharmacology, National Institute of Health Science, 3-25-26 Tonomachi, Kawasaki-ku, Kawasaki City, Kanagawa, Japan
| | - Hiroko Toyoda
- Stem Cell Evaluation Technology Research Association, Grande Building 8F, 2-26-9 Hatchobori, Chuo-ku, Tokyo 104-0032, Japan
| | - Ikue Mihara
- Stem Cell Evaluation Technology Research Association, Grande Building 8F, 2-26-9 Hatchobori, Chuo-ku, Tokyo 104-0032, Japan
| | - Hiroyuki Moriguchi
- Stem Cell Evaluation Technology Research Association, Grande Building 8F, 2-26-9 Hatchobori, Chuo-ku, Tokyo 104-0032, Japan
| | - Hitoshi Naraoka
- Stem Cell Evaluation Technology Research Association, Grande Building 8F, 2-26-9 Hatchobori, Chuo-ku, Tokyo 104-0032, Japan
| | - Tomomi Furihata
- School of Pharmacy, Tokyo University of Pharmacy and Life Sciences, 1432-1 Horinouchi, Hachioji, Tokyo 192-0392 Japan
| | - Seiichi Ishida
- Laboratory of Neuropharmacology, Division of Pharmacology, National Institute of Health Science, 3-25-26 Tonomachi, Kawasaki-ku, Kawasaki City, Kanagawa, Japan.,Division of Applied Life Science, Graduate School of Engineering, Sojo University, 4-22-1 Ikeda, Nishi-ku, Kumamoto City, Kumamoto, Japan
| | - Kaoru Sato
- Laboratory of Neuropharmacology, Division of Pharmacology, National Institute of Health Science, 3-25-26 Tonomachi, Kawasaki-ku, Kawasaki City, Kanagawa, Japan
| |
Collapse
|
20
|
Zhu YN, Gu XL, Wang LY, Guan N, Li CG. All-Trans Retinoic Acid Promotes M2 Macrophage Polarization in Vitro by Activating the p38MAPK/STAT6 Signaling Pathway. Immunol Invest 2023; 52:298-318. [PMID: 36731128 DOI: 10.1080/08820139.2023.2173077] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
BACKGROUND M2-type macrophages are inflammation-suppressing cells that are differentiated after induction by cytokines such as IL-4 or IL-13, which play an important regulatory role in inflammation and influence the regression of inflammation-related diseases. All-trans retinoic acid (ATRA) has an important role in suppressing immune-mediated inflammatory responses but the effect and underlying mechanism of ATRA on the polarization of M2 macrophages remains unclear. METHODS Macrophages were isolated from peritoneal wash fluid, and IL-4 (20 ng/mL) was used to construct a m2-type macrophage polarization model. The model was incubated with different concentrations of ATRA (15 µg/ml, 30 µg/ml, 45 µg/ml) for 24 h, and pretreated macrophages with p38MAPKα inhibitor SB202190 (20 μM). MTT, Trypan blue staining, Annexin V-PE/7-AAD staining, flow cytometry, real-time PCR and western blotting were used to investigate the effect and mechanism of ATRA on the polarization of M2 macrophages. RESULTS Compared with the IL-4 group, the proportion of F4/80+CD206+ M2-type macrophages was significantly higher in the ATRA group (P < 0.01). mRNA and protein expression levels of Arg-1, IL-10 and TGF-β1 were as significantly higher (P < 0.01) in the ATRA group as phosphorylation levels of STAT6 and p38MAPK (P < 0.01). After pretreatment with the addition of the inhibitor SB202190, M2-type macrophages proportion and their associated factors expression were significantly (P < 0.01) reduced, as compared with those in the ATRA group, but they were comparable (P > 0.05) with the IL-4 group. CONCLUSION The combination of ATRA and IL-4 activated the p38MAPK/STAT6-signaling pathway to promote polarization of M2 macrophages.
Collapse
Affiliation(s)
- Ya-Nan Zhu
- Department of Periodontics and Mucasa, The Second Affiliated Hospital of Jinzhou Medical University, Jinzhou, Liaoning, P.R. China
| | - Xiao-Li Gu
- Department of Periodontics and Mucasa, The Second Affiliated Hospital of Jinzhou Medical University, Jinzhou, Liaoning, P.R. China
| | - Lin-Yuan Wang
- Department of Periodontics and Mucasa, The Second Affiliated Hospital of Jinzhou Medical University, Jinzhou, Liaoning, P.R. China
| | - Ning Guan
- Key Laboratory of Brain and Spinal Cord Injury Research, First Affiliated Hospital of Jinzhou Medical University, Jinzhou, Liaoning, P.R. China
| | - Chen-Guang Li
- Key Laboratory of Brain and Spinal Cord Injury Research, First Affiliated Hospital of Jinzhou Medical University, Jinzhou, Liaoning, P.R. China
| |
Collapse
|
21
|
Mota IFL, de Lima LS, Santana BDM, Gobbo GDAM, Bicca JVML, Azevedo JRM, Veras LG, Taveira RDAA, Pinheiro GB, Mortari MR. Alzheimer's Disease: Innovative Therapeutic Approaches Based on Peptides and Nanoparticles. Neuroscientist 2023; 29:78-96. [PMID: 34018874 DOI: 10.1177/10738584211016409] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
Alzheimer's disease (AD) is the main cause of dementia in the world and its etiology is not yet fully understood. The pathology of AD is primarily characterized by intracellular neurofibrillary tangles and extracellular amyloid-β plaques. Unfortunately, few treatment options are available, and most treat symptoms, as is the case of acetylcholinesterase inhibitors (IAChE) and N-methyl-d-aspartate receptor antagonists. For more than 20 years pharmaceutical research has targeted the "amyloid cascade hypothesis," but this has not produced meaningful results, leading researchers to focus now on other characteristics of the disease and on multitarget approaches. This review aims to evaluate some new treatments that are being developed and studied. Among these are new treatments based on peptides, which have high selectivity and low toxicity; however, these compounds have a short half-life and encounter challenges when crossing the blood-brain barrier. The present review discusses up-and-coming peptides tested as treatments and explores some nanotechnological strategies to overcome the downsides. These compounds are promising, as they not only act on the symptoms but also aim to prevent progressive neuronal loss.
Collapse
Affiliation(s)
- Isabela F L Mota
- Laboratory of Neuropharmacology, Department of Physiological Sciences, Institute of Biological Sciences, University of Brasília, Brasília, Brazil
| | - Larissa S de Lima
- Laboratory of Neuropharmacology, Department of Physiological Sciences, Institute of Biological Sciences, University of Brasília, Brasília, Brazil
| | - Bruna de M Santana
- Laboratory of Neuropharmacology, Department of Physiological Sciences, Institute of Biological Sciences, University of Brasília, Brasília, Brazil
| | - Giovanna de A M Gobbo
- Laboratory of Neuropharmacology, Department of Physiological Sciences, Institute of Biological Sciences, University of Brasília, Brasília, Brazil
| | - João V M L Bicca
- Laboratory of Neuropharmacology, Department of Physiological Sciences, Institute of Biological Sciences, University of Brasília, Brasília, Brazil
| | - Juliana R M Azevedo
- Laboratory of Neuropharmacology, Department of Physiological Sciences, Institute of Biological Sciences, University of Brasília, Brasília, Brazil
| | - Letícia G Veras
- Laboratory of Neuropharmacology, Department of Physiological Sciences, Institute of Biological Sciences, University of Brasília, Brasília, Brazil
| | - Rodrigo de A A Taveira
- Laboratory of Neuropharmacology, Department of Physiological Sciences, Institute of Biological Sciences, University of Brasília, Brasília, Brazil
| | - Gabriela B Pinheiro
- Laboratory of Neuropharmacology, Department of Physiological Sciences, Institute of Biological Sciences, University of Brasília, Brasília, Brazil
| | - Márcia R Mortari
- Laboratory of Neuropharmacology, Department of Physiological Sciences, Institute of Biological Sciences, University of Brasília, Brasília, Brazil
| |
Collapse
|
22
|
Sawant-Basak A, Chen L, Lockwood P, Boyden T, Doran AC, Mancuso J, Zasadny K, McCarthy T, Morris ED, Carson RE, Esterlis I, Huang Y, Nabulsi N, Planeta B, Fullerton T. Investigating CNS distribution of PF-05212377, a P-glycoprotein substrate, by translation of 5-HT 6 receptor occupancy from non-human primates to humans. Biopharm Drug Dispos 2023; 44:48-59. [PMID: 36825693 DOI: 10.1002/bdd.2351] [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: 10/27/2022] [Revised: 02/10/2023] [Accepted: 02/14/2023] [Indexed: 02/25/2023]
Abstract
PF-05212377 (SAM760) is a potent and selective 5-HT6 antagonist, previously under development for the treatment of Alzheimer's disease. In vitro, PF-05212377 was determined to be a P-gp/non-BCRP human transporter substrate. Species differences were observed in the in vivo brain penetration of PF-05212377 with a ratio of the unbound concentration in brain/unbound concentration in plasma (Cbu /Cpu ) of 0.05 in rat and 0.64 in non-human primates (NHP). Based on pre-clinical evidence, brain penetration and target engagement of PF-05212377 was confirmed in NHP using positron emission tomography (PET) measured 5-HT6 receptor occupancy (%RO). The NHP Cpu EC50 of PF-05212377 was 0.31 nM (consistent with the in vitro human 5HT6 Ki : 0.32 nM). P-gp has been reported to be expressed in higher abundance at the rat BBB and in similar abundance at the BBB of non-human primates and human; brain penetration of PF-05212377 in humans was postulated to be similar to that in non-human primates. In humans, PF-05212377 demonstrated dose and concentration dependent increases in 5-HT6 RO; maximal 5-HT6 RO of ∼80% was measured in humans at doses of ≥15 mg with an estimated unbound plasma EC50 of 0.37 nM (which was similar to the in vitro human 5HT6 binding Ki 0.32 nM). In conclusion, cumulative evidence from NHP and human PET RO assessments confirmed that NHP is more appropriate than the rat for the prediction of human brain penetration of PF-05212377, a P-gp/non-BCRP substrate. Clinical trial number: NCT01258751.
Collapse
Affiliation(s)
- Aarti Sawant-Basak
- Clinical Pharmacology, Early Clinical Development, Worldwide Research, Development and Medical, Pfizer Inc, Cambridge, Massachusetts, USA
| | - Laigao Chen
- Digital Sciences and Translational Imaging, Early Clinical Development, Worldwide Research, Development and Medical, Pfizer Inc, Cambridge, Massachusetts, USA
| | - Peter Lockwood
- Clinical Pharmacology, Early Clinical Development, Worldwide Research, Development and Medical, Pfizer Inc, Cambridge, Massachusetts, USA
| | - Tracey Boyden
- Pharmacokinetics, Dynamics, and Metabolism, Medicine Design, Worldwide Research, Development, and Medical, Pfizer Inc., Groton, Connecticut, USA
| | - Angela C Doran
- Pharmacokinetics, Dynamics, and Metabolism, Medicine Design, Worldwide Research, Development, and Medical, Pfizer Inc., Groton, Connecticut, USA
| | - Jessica Mancuso
- Biostatistics, Early Clinical Development, Worldwide Research, Development and Medical, Pfizer Inc, Cambridge, Massachusetts, USA
| | - Kenneth Zasadny
- Digital Sciences and Translational Imaging, Early Clinical Development, Worldwide Research, Development and Medical, Pfizer Inc, Cambridge, Massachusetts, USA
| | - Timothy McCarthy
- Digital Sciences and Translational Imaging, Early Clinical Development, Worldwide Research, Development and Medical, Pfizer Inc, Cambridge, Massachusetts, USA
| | - Evan D Morris
- Department of Radiology and Biomedical Imaging, Yale University, New Haven, Connecticut, USA
| | - Richard E Carson
- Department of Radiology and Biomedical Imaging, Yale University, New Haven, Connecticut, USA
| | - Irina Esterlis
- Department of Radiology and Biomedical Imaging, Yale University, New Haven, Connecticut, USA
| | - Yiyun Huang
- Department of Radiology and Biomedical Imaging, Yale University, New Haven, Connecticut, USA
| | - Nabeel Nabulsi
- Department of Radiology and Biomedical Imaging, Yale University, New Haven, Connecticut, USA
| | - Beata Planeta
- Department of Radiology and Biomedical Imaging, Yale University, New Haven, Connecticut, USA
| | - Terence Fullerton
- Internal Medicine, Global Product Development, Pfizer Inc, Groton, CT, USA
| |
Collapse
|
23
|
Alkahtani S, AL-Johani NS, Alarifi S. Mechanistic Insights, Treatment Paradigms, and Clinical Progress in Neurological Disorders: Current and Future Prospects. Int J Mol Sci 2023; 24:1340. [PMID: 36674852 PMCID: PMC9865061 DOI: 10.3390/ijms24021340] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2022] [Revised: 12/18/2022] [Accepted: 12/19/2022] [Indexed: 01/12/2023] Open
Abstract
Neurodegenerative diseases (NDs) are a major cause of disability and are related to brain development. The neurological signs of brain lesions can vary from mild clinical shortfalls to more delicate and severe neurological/behavioral symptoms and learning disabilities, which are progressive. In this paper, we have tried to summarize a collective view of various NDs and their possible therapeutic outcomes. These diseases often occur as a consequence of the misfolding of proteins post-translation, as well as the dysfunctional trafficking of proteins. In the treatment of neurological disorders, a challenging hurdle to cross regarding drug delivery is the blood-brain barrier (BBB). The BBB plays a unique role in maintaining the homeostasis of the central nervous system (CNS) by exchanging components between the circulations and shielding the brain from neurotoxic pathogens and detrimental compounds. Here, we outline the current knowledge about BBB deterioration in the evolving brain, its origin, and therapeutic interventions. Additionally, we summarize the physiological scenarios of the BBB and its role in various cerebrovascular diseases. Overall, this information provides a detailed account of BBB functioning and the development of relevant treatments for neurological disorders. This paper will definitely help readers working in the field of neurological scientific communities.
Collapse
Affiliation(s)
- Saad Alkahtani
- Department of Zoology, College of Science, King Saud University, P.O. Box 2455, Riyadh 11451, Saudi Arabia
| | | | | |
Collapse
|
24
|
Sánchez-Martínez JD, Garcia AR, Alvarez-Rivera G, Valdés A, Brito MA, Cifuentes A. In Vitro Study of the Blood-Brain Barrier Transport of Natural Compounds Recovered from Agrifood By-Products and Microalgae. Int J Mol Sci 2022; 24:ijms24010533. [PMID: 36613976 PMCID: PMC9820279 DOI: 10.3390/ijms24010533] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2022] [Revised: 12/19/2022] [Accepted: 12/24/2022] [Indexed: 12/31/2022] Open
Abstract
Agrifood by-products and microalgae represent a low-cost and valuable source of bioactive compounds with neuroprotective properties. However, the neuroprotective effectiveness of therapeutic molecules can be limited by their capacity to cross the blood-brain barrier (BBB) and reach the brain. In this research, various green extracts from Robinia pseudoacacia (ASFE), Cyphomandra betacea (T33), Coffea arabica (PPC1), Olea europaea L., (OL-SS), Citrus sinensis (PLE100) by-products and from the microalgae Dunaliella salina (DS) that have demonstrated in vitro neuroprotective potential were submitted to an in vitro BBB permeability and transport assay based on an immortalized human brain microvascular endothelial cells (HBMEC) model. Toxicity and BBB integrity tests were performed, and the transport of target bioactive molecules across the BBB were evaluated after 2 and 4 h of incubation using gas and liquid chromatography coupled to quadrupole-time-of-flight mass spectrometry (GC/LC-Q-TOF-MS). The HBMEC-BBB transport assay revealed a high permeability of representative neuroprotective compounds, such as mono- and sesquiterpenoids, phytosterols and some phenolic compounds. The obtained results from the proposed in vitro BBB cellular model provide further evidence of the neuroprotective potential of the target natural extracts, which represent a promising source of functional ingredients to be transferred into food supplements, food additives, or nutraceuticals with scientifically supported neuroprotective claims.
Collapse
Affiliation(s)
- José David Sánchez-Martínez
- Laboratory of Foodomics, Institute of Food Science Research, CIAL, UAM-CSIC, Nicolás Cabrera 9, 28049 Madrid, Spain
| | - Ana Rita Garcia
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisboa, Portugal
- Department of Pharmaceutical Sciences and Medicines, Faculty of Pharmacy, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisboa, Portugal
| | - Gerardo Alvarez-Rivera
- Laboratory of Foodomics, Institute of Food Science Research, CIAL, UAM-CSIC, Nicolás Cabrera 9, 28049 Madrid, Spain
| | - Alberto Valdés
- Laboratory of Foodomics, Institute of Food Science Research, CIAL, UAM-CSIC, Nicolás Cabrera 9, 28049 Madrid, Spain
| | - Maria Alexandra Brito
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisboa, Portugal
- Department of Pharmaceutical Sciences and Medicines, Faculty of Pharmacy, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisboa, Portugal
- Correspondence: (M.A.B.); (A.C.); Tel.: +351-217946449 (M.A.B.); Tel.: +34-910017955 (A.C.)
| | - Alejandro Cifuentes
- Laboratory of Foodomics, Institute of Food Science Research, CIAL, UAM-CSIC, Nicolás Cabrera 9, 28049 Madrid, Spain
- Correspondence: (M.A.B.); (A.C.); Tel.: +351-217946449 (M.A.B.); Tel.: +34-910017955 (A.C.)
| |
Collapse
|
25
|
Di Scala C, Armstrong N, Chahinian H, Chabrière E, Fantini J, Yahi N. AmyP53, a Therapeutic Peptide Candidate for the Treatment of Alzheimer’s and Parkinson’s Disease: Safety, Stability, Pharmacokinetics Parameters and Nose-to Brain Delivery. Int J Mol Sci 2022; 23:ijms232113383. [PMID: 36362170 PMCID: PMC9654333 DOI: 10.3390/ijms232113383] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2022] [Revised: 10/26/2022] [Accepted: 10/30/2022] [Indexed: 11/06/2022] Open
Abstract
Neurodegenerative disorders are a major public health issue. Despite decades of research efforts, we are still seeking an efficient cure for these pathologies. The initial paradigm of large aggregates of amyloid proteins (amyloid plaques, Lewis bodies) as the root cause of Alzheimer’s and Parkinson’s diseases has been mostly dismissed. Instead, membrane-bound oligomers forming Ca2+-permeable amyloid pores are now considered appropriate targets for these diseases. Over the last 20 years, our group deciphered the molecular mechanisms of amyloid pore formation, which appeared to involve a common pathway for all amyloid proteins, including Aβ (Alzheimer) and α-synuclein (Parkinson). We then designed a short peptide (AmyP53), which prevents amyloid pore formation by targeting gangliosides, the plasma membrane receptors of amyloid proteins. Herein, we show that aqueous solutions of AmyP53 are remarkably stable upon storage at temperatures up to 45 °C for several months. AmyP53 appeared to be more stable in whole blood than in plasma. Pharmacokinetics studies in rats demonstrated that the peptide can rapidly and safely reach the brain after intranasal administration. The data suggest both the direct transport of AmyP53 via the olfactory bulb (and/or the trigeminal nerve) and an indirect transport via the circulation and the blood–brain barrier. In vitro experiments confirmed that AmyP53 is as active as cargo peptides in crossing the blood–brain barrier, consistent with its amino acid sequence specificities and physicochemical properties. Overall, these data open a route for the use of a nasal spray formulation of AmyP53 for the prevention and/or treatment of Alzheimer’s and Parkinson’s diseases in future clinical trials in humans.
Collapse
Affiliation(s)
- Coralie Di Scala
- Neuroscience Center—HiLIFE, Helsinki Institute of Life Science, University of Helsinki, 00014 Helsinki, Finland
| | - Nicholas Armstrong
- IRD, APHM, MEPHI, IHU Méditerranée Infection, Aix Marseille University, 13005 Marseille, France
| | - Henri Chahinian
- INSERM UMR_S 1072, Aix Marseille University, 13015 Marseille, France
| | - Eric Chabrière
- IRD, APHM, MEPHI, IHU Méditerranée Infection, Aix Marseille University, 13005 Marseille, France
| | - Jacques Fantini
- INSERM UMR_S 1072, Aix Marseille University, 13015 Marseille, France
| | - Nouara Yahi
- INSERM UMR_S 1072, Aix Marseille University, 13015 Marseille, France
- Correspondence:
| |
Collapse
|
26
|
Affiliation(s)
- Peter W. Kenny
- Berwick-on-Sea, North Coast Road, Blanchisseuse, Saint George, Trinidad and Tobago
| |
Collapse
|
27
|
Cytoskeleton Elements Contribute to Prion Peptide-Induced Endothelial Barrier Breakdown in a Blood–Brain Barrier In Vitro System. Int J Mol Sci 2022; 23:ijms232012126. [PMID: 36293002 PMCID: PMC9603506 DOI: 10.3390/ijms232012126] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Revised: 10/02/2022] [Accepted: 10/07/2022] [Indexed: 11/17/2022] Open
Abstract
The mechanisms involved in the interaction of PrP 106-126, a peptide corresponding to the prion protein amyloidogenic region, with the blood–brain barrier (BBB) were studied. PrP 106-126 treatment that was previously shown to impair BBB function, reduced cAMP levels in cultured brain endothelial cells, increased nitric oxide (NO) levels, and changed the activation mode of the small GTPases Rac1 (inactivation) and RhoA (activation). The latter are well established regulators of endothelial barrier properties that act via cytoskeletal elements. Indeed, liquid chromatography-mass spectrometry (LC-MS)-based proteomic profiling study revealed extensive changes in expression of cytoskeleton-related proteins. These results shed light on the nature of the interaction between the prion peptide PrP 106-126 and the BBB and emphasize the importance of the cytoskeleton in endothelium response to prion- induced stress.
Collapse
|
28
|
Zhang S, Gan L, Cao F, Wang H, Gong P, Ma C, Ren L, Lin Y, Lin X. The barrier and interface mechanisms of the brain barrier, and brain drug delivery. Brain Res Bull 2022; 190:69-83. [PMID: 36162603 DOI: 10.1016/j.brainresbull.2022.09.017] [Citation(s) in RCA: 52] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2022] [Revised: 08/25/2022] [Accepted: 09/20/2022] [Indexed: 11/26/2022]
Abstract
Three different barriers are formed between the cerebrovascular and the brain parenchyma: the blood-brain barrier (BBB), the blood-cerebrospinal fluid barrier (BCSFB), and the cerebrospinal fluid-brain barrier (CBB). The BBB is the main regulator of blood and central nervous system (CNS) material exchange. The semipermeable nature of the BBB limits the passage of larger molecules and hydrophilic small molecules, Food and Drug Administration (FDA)-approved drugs for the CNS have been generally limited to lipid-soluble small molecules. Although the complexity of the BBB affects CNS drug delivery, understanding the composition and function of the BBB can provide a platform for the development of new methods for CNS drug delivery. This review summarizes the classification of the brain barrier, the composition and role of the basic structures of the BBB, and the transport, barrier, and destruction mechanisms of the BBB; discusses the advantages and disadvantages of different drug delivery methods and prospects for future drug delivery strategies.
Collapse
Affiliation(s)
- Shanshan Zhang
- The Second Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou 310005, Zhejiang Province, China
| | - Lin Gan
- Department of Neurobiology and Acupuncture Research, The Third Clinical Medical College, Zhejiang Chinese Medical University, Key Laboratory of Acupuncture and Neurology of Zhejiang Province, Hangzhou 310053, China
| | - Fengye Cao
- Yiyang The First Hospital of Traditional Chinese Medicine, Yiyang, Hunan Province, 413000, China
| | - Hao Wang
- Department of Neurobiology and Acupuncture Research, The Third Clinical Medical College, Zhejiang Chinese Medical University, Key Laboratory of Acupuncture and Neurology of Zhejiang Province, Hangzhou 310053, China
| | - Peng Gong
- Department of Neurobiology and Acupuncture Research, The Third Clinical Medical College, Zhejiang Chinese Medical University, Key Laboratory of Acupuncture and Neurology of Zhejiang Province, Hangzhou 310053, China
| | - Congcong Ma
- Department of Neurobiology and Acupuncture Research, The Third Clinical Medical College, Zhejiang Chinese Medical University, Key Laboratory of Acupuncture and Neurology of Zhejiang Province, Hangzhou 310053, China
| | - Li Ren
- Department of Neurobiology and Acupuncture Research, The Third Clinical Medical College, Zhejiang Chinese Medical University, Key Laboratory of Acupuncture and Neurology of Zhejiang Province, Hangzhou 310053, China
| | - Yubo Lin
- Department of Neurobiology and Acupuncture Research, The Third Clinical Medical College, Zhejiang Chinese Medical University, Key Laboratory of Acupuncture and Neurology of Zhejiang Province, Hangzhou 310053, China
| | - Xianming Lin
- Department of Neurobiology and Acupuncture Research, The Third Clinical Medical College, Zhejiang Chinese Medical University, Key Laboratory of Acupuncture and Neurology of Zhejiang Province, Hangzhou 310053, China.
| |
Collapse
|
29
|
Fang Y, Zhang G, Bai Z, Yan Y, Song X, Zhao X, Yang P, Zhang Z. Low-intensity ultrasound: A novel technique for adjuvant treatment of gliomas. Biomed Pharmacother 2022; 153:113394. [DOI: 10.1016/j.biopha.2022.113394] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2022] [Revised: 07/03/2022] [Accepted: 07/07/2022] [Indexed: 11/02/2022] Open
|
30
|
Chronic Low Dose Morphine Does Not Alter Two In Vitro BBB Models. Brain Sci 2022; 12:brainsci12070888. [PMID: 35884695 PMCID: PMC9312884 DOI: 10.3390/brainsci12070888] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2022] [Revised: 06/16/2022] [Accepted: 07/01/2022] [Indexed: 12/07/2022] Open
Abstract
The blood–brain barrier (BBB) mediates cellular and molecular passage between the central nervous system (CNS) and peripheral circulation. Compromised BBB integrity has been linked to neurocognitive deficits in multiple diseases and various infections, including those associated with HIV-1 infection. Understanding the impact of exposure to pharmaceuticals, such as those utilized for pain management by patients suffering from CNS disease, on BBB regulation and function is clinically important. In this study, we modelled two different BBB systems; a primary human co-culture and a cell line monoculture. These systems were both exposed to three daily repeat doses of morphine and examined for alterations to BBB integrity via permeability, PBMC transmigration, and chemokine gradient changes. We did not find any significant changes to either BBB system with repeat morphine dosing, suggesting that repeat morphine exposure may not play a significant role in BBB changes.
Collapse
|
31
|
Electrospun Scaffolds as Cell Culture Substrates for the Cultivation of an In Vitro Blood-Brain Barrier Model Using Human Induced Pluripotent Stem Cells. Pharmaceutics 2022; 14:pharmaceutics14061308. [PMID: 35745880 PMCID: PMC9231001 DOI: 10.3390/pharmaceutics14061308] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 06/16/2022] [Accepted: 06/16/2022] [Indexed: 11/17/2022] Open
Abstract
The human blood–brain barrier (BBB) represents the interface of microvasculature and the central nervous system, regulating the transport of nutrients and protecting the brain from external threats. To gain a deeper understanding of (patho)physiological processes affecting the BBB, sophisticated models mimicking the in vivo situation are required. Currently, most in vitro models are cultivated on stiff, semipermeable, and non-biodegradable Transwell® membrane inserts, not adequately mimicking the complexity of the extracellular environment of the native human BBB. To overcome these disadvantages, we developed three-dimensional electrospun scaffolds resembling the natural structure of the human extracellular matrix. The polymer fibers of the scaffold imitate collagen fibrils of the human basement membrane, exhibiting excellent wettability and biomechanical properties, thus facilitating cell adhesion, proliferation, and migration. Cultivation of human induced pluripotent stem cells (hiPSCs) on these scaffolds enabled the development of a physiological BBB phenotype monitored via the formation of tight junctions and validated by the paracellular permeability of sodium fluorescein, further accentuating the non-linearity of TEER and barrier permeability. The novel in vitro model of the BBB forms a tight endothelial barrier, offering a platform to study barrier functions in a (patho)physiologically relevant context.
Collapse
|
32
|
Sánchez-Martínez JD, Valdés A, Gallego R, Suárez-Montenegro ZJ, Alarcón M, Ibañez E, Alvarez-Rivera G, Cifuentes A. Blood–Brain Barrier Permeability Study of Potential Neuroprotective Compounds Recovered From Plants and Agri-Food by-Products. Front Nutr 2022; 9:924596. [PMID: 35782945 PMCID: PMC9243654 DOI: 10.3389/fnut.2022.924596] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Accepted: 05/13/2022] [Indexed: 11/30/2022] Open
Abstract
Plants and agri-food by-products represent a wide and renewable source of bioactive compounds with neuroprotective properties. In this research, various green extraction techniques were employed to recover bioactive molecules from Kalanchoe daigremontiana (kalanchoe), epicarp of Cyphomandra betacea (tamarillo), and cooperage woods from Robinia pseudoacacia (acacia) and Nothofagus pumilio (lenga), as well as a reference extract (positive control) from Rosmarinus officinalis L. (rosemary). The neuroprotective capacity of these plant extracts was evaluated in a set of in vitro assays, including enzymatic [acetylcholinesterase (AChE), butyrylcholinesterase (BChE), and lipoxygenase (LOX)] and antioxidant [ABTS, and reactive oxygen and nitrogen species (ROS and RNS)] bioactivity tests. Extracts were also submitted to a parallel artificial membrane permeability assay mimicking the blood–brain barrier (PAMPA-BBB) and to two cell viability assays in HK-2 and SH-SY5Y cell lines. Comprehensive phytochemical profiling based on liquid chromatography coupled to quadrupole-time-of-flight mass spectrometry (LC-Q-TOF-MS) analysis showed enriched content of phenolic and terpenoid compounds in the target extracts. Moreover, in vitro bioactivity tests showed promising neuroprotective capacity, particularly for supercritical-fluid extraction (SFE) extract from acacia (ABTS IC50 = 0.11 μg ml−1; ROS IC50 = 1.56 μg ml−1; AChE IC50 = 4.23 μg ml−1; BChE IC50 = 1.20 μg ml−1; and LOX IC50 = 4.37 μg ml−1), whereas PAMPA-BBB assays revealed high perfusion capacity of some representative compounds, such as phenolic acids or flavonoids. Regarding cytotoxic assays, tamarillo and rosemary SFE extracts can be considered as non-toxic, acacia SFE extract and lenga pressurized liquid extraction (PLE) extract as mild-cytotoxic, and kalanchoe as highly toxic extracts. The obtained results demonstrate the great potential of the studied biomass extracts to be transformed into valuable food additives, food supplements, or nutraceuticals with promising neuroprotective properties.
Collapse
Affiliation(s)
- José David Sánchez-Martínez
- Laboratory of Foodomics, Institute of Food Science Research, CIAL, Spanish National Research Council (CSIC) - Universidad Autónoma de Madrid (UAM), Madrid, Spain
| | - Alberto Valdés
- Laboratory of Foodomics, Institute of Food Science Research, CIAL, Spanish National Research Council (CSIC) - Universidad Autónoma de Madrid (UAM), Madrid, Spain
| | - Rocio Gallego
- Laboratory of Foodomics, Institute of Food Science Research, CIAL, Spanish National Research Council (CSIC) - Universidad Autónoma de Madrid (UAM), Madrid, Spain
| | - Zully Jimena Suárez-Montenegro
- Laboratory of Foodomics, Institute of Food Science Research, CIAL, Spanish National Research Council (CSIC) - Universidad Autónoma de Madrid (UAM), Madrid, Spain
| | - Marina Alarcón
- Area of Food Technology, Faculty of Chemical Sciences and Technologies, University of Castilla-La Mancha, Ciudad Real, Spain
| | - Elena Ibañez
- Laboratory of Foodomics, Institute of Food Science Research, CIAL, Spanish National Research Council (CSIC) - Universidad Autónoma de Madrid (UAM), Madrid, Spain
| | - Gerardo Alvarez-Rivera
- Laboratory of Foodomics, Institute of Food Science Research, CIAL, Spanish National Research Council (CSIC) - Universidad Autónoma de Madrid (UAM), Madrid, Spain
- *Correspondence: Gerardo Alvarez-Rivera
| | - Alejandro Cifuentes
- Laboratory of Foodomics, Institute of Food Science Research, CIAL, Spanish National Research Council (CSIC) - Universidad Autónoma de Madrid (UAM), Madrid, Spain
- Alejandro Cifuentes
| |
Collapse
|
33
|
Ding Y, Jiang X, Kim Y. Relational graph convolutional networks for predicting blood-brain barrier penetration of drug molecules. Bioinformatics 2022; 38:2826-2831. [PMID: 35561199 PMCID: PMC9113341 DOI: 10.1093/bioinformatics/btac211] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Revised: 03/28/2022] [Accepted: 04/05/2022] [Indexed: 02/03/2023] Open
Abstract
MOTIVATION Evaluating the blood-brain barrier (BBB) permeability of drug molecules is a critical step in brain drug development. Traditional methods for the evaluation require complicated in vitro or in vivo testing. Alternatively, in silico predictions based on machine learning have proved to be a cost-efficient way to complement the in vitro and in vivo methods. However, the performance of the established models has been limited by their incapability of dealing with the interactions between drugs and proteins, which play an important role in the mechanism behind the BBB penetrating behaviors. To address this limitation, we employed the relational graph convolutional network (RGCN) to handle the drug-protein interactions as well as the properties of each individual drug. RESULTS The RGCN model achieved an overall accuracy of 0.872, an area under the receiver operating characteristic (AUROC) of 0.919 and an area under the precision-recall curve (AUPRC) of 0.838 for the testing dataset with the drug-protein interactions and the Mordred descriptors as the input. Introducing drug-drug similarity to connect structurally similar drugs in the data graph further improved the testing results, giving an overall accuracy of 0.876, an AUROC of 0.926 and an AUPRC of 0.865. In particular, the RGCN model was found to greatly outperform the LightGBM base model when evaluated with the drugs whose BBB penetration was dependent on drug-protein interactions. Our model is expected to provide high-confidence predictions of BBB permeability for drug prioritization in the experimental screening of BBB-penetrating drugs. AVAILABILITY AND IMPLEMENTATION The data and the codes are freely available at https://github.com/dingyan20/BBB-Penetration-Prediction. SUPPLEMENTARY INFORMATION Supplementary data are available at Bioinformatics online.
Collapse
Affiliation(s)
- Yan Ding
- Center for Secure Artificial Intelligence for Healthcare, School of Biomedical Informatics, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Xiaoqian Jiang
- Center for Secure Artificial Intelligence for Healthcare, School of Biomedical Informatics, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Yejin Kim
- To whom correspondence should be addressed.
| |
Collapse
|
34
|
Nasrin S, Islam MN, Tayab MA, Nasrin MS, Siddique MAB, Emran TB, Reza ASMA. Chemical profiles and pharmacological insights of Anisomeles indica Kuntze: An experimental chemico-biological interaction. Biomed Pharmacother 2022; 149:112842. [PMID: 35325851 DOI: 10.1016/j.biopha.2022.112842] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2021] [Revised: 03/14/2022] [Accepted: 03/16/2022] [Indexed: 12/22/2022] Open
Abstract
Anisomeles indica (L.) Kuntze is an ethnomedicinally important plant that has long been used in traditional medicine to treat a variety of ailments, including dyspepsia, abdominal pain, colic, allergies, inflammation, and rheumatic arthritis. However, the scientific framework underlying these medicinal properties is not well known. This study aimed to investigate the antidepressive, antidiarrheal, thrombolytic, and anti-inflammatory potential of a methanol extract of A. indica (MeOH-AI). The potential bioactive compounds in the MeOH-AI were identified using gas chromatography-mass spectrometry (GC-MS), and antidepressant activities were evaluated using the tail suspension test (TST) and forced swim test (FST). Antidiarrheal effects were also assayed in castor oil-induced diarrhea and gastrointestinal motility studies. The anti-inflammatory activities were explored by examining the effects on protein inhibition and denaturation in heat- and hypotonic solution-induced hemolysis assays. The thrombolytic activity was evaluated using the clot lysis test in human blood. BIOVIA and Schrödinger Maestro (v11.1) were applied for docking analysis to determine binding interactions, and the absorption, distribution, metabolisms, excretion/toxicity (ADME/T) properties of bioactive compounds were explored using a web-based method. The GC-MS analysis of MeOH-AI revealed the presence of several bioactive compounds. MeOH-AI administration resulted in significant (p < 0.01) reductions in the immobility times for both the FST and TST compared with those in the control group. MeOH-AI also induced significant (p < 0.01) reductions in castor oil-induced diarrhea severity and gastrointestinal motility in a mouse model. In addition, the in vitro anti-inflammatory and thrombolytic activity studies produced remarkable responses. The binding assay showed that 4-dehydroxy-N-(4,5-methylenedioxy-2-nitrobenzylidene) tyramine interacts favorably with monoamine oxidase and serotonin and M3 muscarinic acetylcholine receptors, displaying good pharmacokinetic properties, which may mediate the effects of MeOH-AI on depression and diarrhea. Overall, the research findings indicated that MeOH-AI has significant antidepressant, antidiarrheal, and anti-inflammatory effects and may represent an alternative source of novel therapeutic factors.
Collapse
Affiliation(s)
- Suaad Nasrin
- Department of Pharmacy, International Islamic University Chittagong, Chittagong 4318, Bangladesh
| | - Mohammad Nazmul Islam
- Department of Pharmacy, International Islamic University Chittagong, Chittagong 4318, Bangladesh
| | - Mohammed Abu Tayab
- Department of Pharmacy, International Islamic University Chittagong, Chittagong 4318, Bangladesh
| | - Mst Samima Nasrin
- Department of Pharmacy, International Islamic University Chittagong, Chittagong 4318, Bangladesh; Department of Biochemistry and Molecular Biology, University of Chittagong, Chittagong 4331, Bangladesh
| | - Md Abu Bakar Siddique
- Institute of National Analytical Research and Service (INARS), Bangladesh Council of Scientific and Industrial Research (BCSIR), Dhaka 1205, Bangladesh
| | - Talha Bin Emran
- Department of Pharmacy, BGC Trust University Bangladesh, Chittagong 4381, Bangladesh; Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, Dhaka 1207, Bangladesh.
| | - A S M Ali Reza
- Department of Pharmacy, International Islamic University Chittagong, Chittagong 4318, Bangladesh; Department of Biochemistry and Molecular Biology, University of Chittagong, Chittagong 4331, Bangladesh.
| |
Collapse
|
35
|
Cheng G, Liu Y, Ma R, Cheng G, Guan Y, Chen X, Wu Z, Chen T. Anti-Parkinsonian Therapy: Strategies for Crossing the Blood-Brain Barrier and Nano-Biological Effects of Nanomaterials. NANO-MICRO LETTERS 2022; 14:105. [PMID: 35426525 PMCID: PMC9012800 DOI: 10.1007/s40820-022-00847-z] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Accepted: 03/12/2022] [Indexed: 05/08/2023]
Abstract
Parkinson's disease (PD), a neurodegenerative disease that shows a high incidence in older individuals, is becoming increasingly prevalent. Unfortunately, there is no clinical cure for PD, and novel anti-PD drugs are therefore urgently required. However, the selective permeability of the blood-brain barrier (BBB) poses a huge challenge in the development of such drugs. Fortunately, through strategies based on the physiological characteristics of the BBB and other modifications, including enhancement of BBB permeability, nanotechnology can offer a solution to this problem and facilitate drug delivery across the BBB. Although nanomaterials are often used as carriers for PD treatment, their biological activity is ignored. Several studies in recent years have shown that nanomaterials can improve PD symptoms via their own nano-bio effects. In this review, we first summarize the physiological features of the BBB and then discuss the design of appropriate brain-targeted delivery nanoplatforms for PD treatment. Subsequently, we highlight the emerging strategies for crossing the BBB and the development of novel nanomaterials with anti-PD nano-biological effects. Finally, we discuss the current challenges in nanomaterial-based PD treatment and the future trends in this field. Our review emphasizes the clinical value of nanotechnology in PD treatment based on recent patents and could guide researchers working in this area in the future.
Collapse
Affiliation(s)
- Guowang Cheng
- Key Laboratory of Modern Preparation of Traditional Chinese Medicine, Ministry of Education, Jiangxi University of Chinese Medicine, Nanchang, 330004, People's Republic of China
| | - Yujing Liu
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, 510405, People's Republic of China
| | - Rui Ma
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, 510405, People's Republic of China
| | - Guopan Cheng
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, 510405, People's Republic of China
| | - Yucheng Guan
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, 510405, People's Republic of China
| | - Xiaojia Chen
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau, 999078, People's Republic of China
| | - Zhenfeng Wu
- Key Laboratory of Modern Preparation of Traditional Chinese Medicine, Ministry of Education, Jiangxi University of Chinese Medicine, Nanchang, 330004, People's Republic of China.
| | - Tongkai Chen
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, 510405, People's Republic of China.
| |
Collapse
|
36
|
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
|
37
|
Zarekiani P, Nogueira Pinto H, Hol EM, Bugiani M, de Vries HE. The neurovascular unit in leukodystrophies: towards solving the puzzle. Fluids Barriers CNS 2022; 19:18. [PMID: 35227276 PMCID: PMC8887016 DOI: 10.1186/s12987-022-00316-0] [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: 12/22/2021] [Accepted: 02/11/2022] [Indexed: 12/11/2022] Open
Abstract
The neurovascular unit (NVU) is a highly organized multicellular system localized in the brain, formed by neuronal, glial (astrocytes, oligodendrocytes, and microglia) and vascular (endothelial cells and pericytes) cells. The blood-brain barrier, a complex and dynamic endothelial cell barrier in the brain microvasculature that separates the blood from the brain parenchyma, is a component of the NVU. In a variety of neurological disorders, including Alzheimer's disease, multiple sclerosis, and stroke, dysfunctions of the NVU occurs. There is, however, a lack of knowledge regarding the NVU function in leukodystrophies, which are rare monogenic disorders that primarily affect the white matter. Since leukodystrophies are rare diseases, human brain tissue availability is scarce and representative animal models that significantly recapitulate the disease are difficult to develop. The introduction of human induced pluripotent stem cells (hiPSC) now makes it possible to surpass these limitations while maintaining the ability to work in a biologically relevant human context and safeguarding the genetic background of the patient. This review aims to provide further insights into the NVU functioning in leukodystrophies, with a special focus on iPSC-derived models that can be used to dissect neurovascular pathophysiology in these diseases.
Collapse
Affiliation(s)
- Parand Zarekiani
- Department of Pathology, Amsterdam Neuroscience, Amsterdam UMC, Vrije Universiteit Amsterdam, de Boelelaan 1117, Amsterdam, The Netherlands
- Amsterdam Leukodystrophy Center, Amsterdam UMC, Amsterdam, The Netherlands
- Department of Molecular Cell Biology and Immunology, Amsterdam Neuroscience, Amsterdam UMC, Vrije Universiteit Amsterdam, De Boelelaan 1117, Amsterdam, The Netherlands
| | - Henrique Nogueira Pinto
- Department of Molecular Cell Biology and Immunology, Amsterdam Neuroscience, Amsterdam UMC, Vrije Universiteit Amsterdam, De Boelelaan 1117, Amsterdam, The Netherlands
- Department of Translational Neuroscience, University Medical Center Utrecht Brain Center, Utrecht University, Utrecht, The Netherlands
| | - Elly M Hol
- Department of Translational Neuroscience, University Medical Center Utrecht Brain Center, Utrecht University, Utrecht, The Netherlands
| | - Marianna Bugiani
- Department of Pathology, Amsterdam Neuroscience, Amsterdam UMC, Vrije Universiteit Amsterdam, de Boelelaan 1117, Amsterdam, The Netherlands
- Amsterdam Leukodystrophy Center, Amsterdam UMC, Amsterdam, The Netherlands
| | - Helga E de Vries
- Department of Molecular Cell Biology and Immunology, Amsterdam Neuroscience, Amsterdam UMC, Vrije Universiteit Amsterdam, De Boelelaan 1117, Amsterdam, The Netherlands.
| |
Collapse
|
38
|
Pan C, Gao Q, Kim BS, Han Y, Gao G. The Biofabrication of Diseased Artery In Vitro Models. MICROMACHINES 2022; 13:mi13020326. [PMID: 35208450 PMCID: PMC8874977 DOI: 10.3390/mi13020326] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Revised: 02/10/2022] [Accepted: 02/17/2022] [Indexed: 11/16/2022]
Abstract
As the leading causes of global death, cardiovascular diseases are generally initiated by artery-related disorders such as atherosclerosis, thrombosis, and aneurysm. Although clinical treatments have been developed to rescue patients suffering from artery-related disorders, the underlying pathologies of these arterial abnormalities are not fully understood. Biofabrication techniques pave the way to constructing diseased artery in vitro models using human vascular cells, biomaterials, and biomolecules, which are capable of recapitulating arterial pathophysiology with superior performance compared with conventional planar cell culture and experimental animal models. This review discusses the critical elements in the arterial microenvironment which are important considerations for recreating biomimetic human arteries with the desired disorders in vitro. Afterward, conventionally biofabricated platforms for the investigation of arterial diseases are summarized, along with their merits and shortcomings, followed by a comprehensive review of advanced biofabrication techniques and the progress of their applications in establishing diseased artery models.
Collapse
Affiliation(s)
- Chen Pan
- Institute of Engineering Medicine, Beijing Institute of Technology, Beijing 100081, China; (C.P.); (Q.G.)
- School of Mechanical Engineering, Beijing Institute of Technology, Beijing 100081, China;
| | - Qiqi Gao
- Institute of Engineering Medicine, Beijing Institute of Technology, Beijing 100081, China; (C.P.); (Q.G.)
- School of Medical Technology, Beijing Institute of Technology, Beijing 100081, China
| | - Byoung-Soo Kim
- School of Biomedical Convergence Engineering, Pusan National University, Yangsan 626841, Korea
- Correspondence: (B.-S.K.); (G.G.)
| | - Yafeng Han
- School of Mechanical Engineering, Beijing Institute of Technology, Beijing 100081, China;
| | - Ge Gao
- Institute of Engineering Medicine, Beijing Institute of Technology, Beijing 100081, China; (C.P.); (Q.G.)
- School of Medical Technology, Beijing Institute of Technology, Beijing 100081, China
- Correspondence: (B.-S.K.); (G.G.)
| |
Collapse
|
39
|
Molins Gutiérrez G, Martorell J, Salazar-Martin AG, Balcells M. A Dynamic, In Vitro BBB Model to Study the Effects of Varying Levels of Shear Stress. Methods Mol Biol 2022; 2492:175-190. [PMID: 35733045 DOI: 10.1007/978-1-0716-2289-6_10] [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: 06/15/2023]
Abstract
The blood-brain barrier (BBB) consists of a tight network of blood capillaries in the brain that separate the circulatory system from the central nervous system. Its particular properties are based on the dynamic interaction between cerebral endothelial cells and other surrounding cells, especially astrocytes. We have designed and synthesized a three-dimensional scaffold that recapitulates the main hallmarks of the BBB extracellular matrix and serves as a platform to co-culture human brain microvascular endothelial cells and human cortical astrocytes. The scaffold can be exposed to flow, thereby allowing the study of flow-mediated pathways at the BBB.
Collapse
Affiliation(s)
- Gemma Molins Gutiérrez
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Jordi Martorell
- IQS School of Engineering, Universitat Ramon Llull, Barcelona, Spain.
| | - Antonio G Salazar-Martin
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Mercedes Balcells
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA, USA
- IQS School of Engineering, Universitat Ramon Llull, Barcelona, Spain
| |
Collapse
|
40
|
Gundersen ET, Førde JL, Tislevoll BS, Leitch C, Barratt G, Gjertsen BT, Herfindal L. Repurposing chlorpromazine for anti-leukaemic therapy by nanoparticle encapsulation. Int J Pharm 2021; 612:121296. [PMID: 34793932 DOI: 10.1016/j.ijpharm.2021.121296] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Revised: 10/26/2021] [Accepted: 11/11/2021] [Indexed: 12/12/2022]
Abstract
Treatment of acute myeloid leukaemia (AML) relies on decades-old drugs, and while recent years have seen some breakthroughs, AML is still characterised by poor prognosis and survival rate. Drug repurposing can expedite the preclinical development of new therapies, and by nanocarrier encapsulation, the number of potentially viable drug candidates can be further expanded. The anti-psychotic drug chlorpromazine (CPZ) has been identified as a candidate for repurposing for AML therapy. Nanoencapsulation may improve the suitability of CPZ for the treatment of AML by reducing its effect on the central nervous system. Using the emulsion-evaporation technique, we have developed PEGylated PLGA nanoparticles loaded with CPZ for AML therapy. The nanoparticles were characterised to be between 150 and 300 nm by DLS, of spherical morphology by TEM, with a drug loading of at least 6.0% (w/w). After an initial burst release of adsorbed drug, the remaining 80% of the drug was retained in the PLGA nanoparticles for at least 24 h. The CPZ-loaded nanoparticles had equal cytotoxic potential towards AML cells to free CPZ, but acted more slowly, in line with the protracted drug release. Crucially, nanoparticles injected intravenously into zebrafish larvae did not accumulate in the brain, and nanoencapsulation also prevented CPZ from crossing an artificial membrane model. This demonstrates that the purpose for nanoencapsulation of CPZ is fulfilled, namely avoiding effects on the central nervous system while retaining the anti-AML activity of the drug.
Collapse
Affiliation(s)
- Edvin Tang Gundersen
- Centre for Pharmacy, Department of Clinical Science, University of Bergen, Bergen, Norway; Hospital Pharmacies Enterprise, Western Norway, Bergen, Norway
| | - Jan-Lukas Førde
- Centre for Pharmacy, Department of Clinical Science, University of Bergen, Bergen, Norway; Department of Internal Medicine, Haukeland University Hospital, Bergen, Norway
| | - Benedicte Sjo Tislevoll
- Centre of Cancer Biomarkers CCBIO, Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Calum Leitch
- Centre of Cancer Biomarkers CCBIO, Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Gillian Barratt
- Université Paris-Saclay, CNRS, Institut Galien Paris-Saclay, Châtenay-Malabry, France
| | - Bjørn Tore Gjertsen
- Centre of Cancer Biomarkers CCBIO, Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Lars Herfindal
- Centre for Pharmacy, Department of Clinical Science, University of Bergen, Bergen, Norway.
| |
Collapse
|
41
|
Blanchard JW, Victor MB, Tsai LH. Dissecting the complexities of Alzheimer disease with in vitro models of the human brain. Nat Rev Neurol 2021; 18:25-39. [PMID: 34750588 DOI: 10.1038/s41582-021-00578-6] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/05/2021] [Indexed: 12/12/2022]
Abstract
Alzheimer disease (AD) is the most prevalent type of dementia. It is marked by severe memory loss and cognitive decline, and currently has limited effective treatment options. Although individuals with AD have common neuropathological hallmarks, emerging data suggest that the disease has a complex polygenic aetiology, and more than 25 genetic loci have been linked to an elevated risk of AD and dementia. Nevertheless, our ability to decipher the cellular and molecular mechanisms that underlie genetic susceptibility to AD, and its progression and severity, remains limited. Here, we discuss ongoing efforts to leverage genomic data from patients using cellular reprogramming technologies to recapitulate complex brain systems and build in vitro discovery platforms. Much attention has already been given to methodologies to derive major brain cell types from pluripotent stem cells. We therefore focus on technologies that combine multiple cell types to recreate anatomical and physiological properties of human brain tissue in vitro. We discuss the advances in the field for modelling four domains that have come into view as key contributors to the pathogenesis of AD: the blood-brain barrier, myelination, neuroinflammation and neuronal circuits. We also highlight opportunities for the field to further interrogate the complex genetic and environmental factors of AD using in vitro models.
Collapse
Affiliation(s)
- Joel W Blanchard
- Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, MA, USA.,Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA, USA.,Department of Neuroscience, Black Family Stem Cell Institute, Ronald M. Loeb Center for Alzheimer's Disease, Icahn School of Medicine at Mt. Sinai, New York, NY, USA
| | - Matheus B Victor
- Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, MA, USA.,Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Li-Huei Tsai
- Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, MA, USA. .,Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA, USA. .,Broad Institute of Harvard and MIT, Cambridge, MA, USA.
| |
Collapse
|
42
|
Radan M, Djikic T, Obradovic D, Nikolic K. Application of in vitro PAMPA technique and in silico computational methods for blood-brain barrier permeability prediction of novel CNS drug candidates. Eur J Pharm Sci 2021; 168:106056. [PMID: 34740787 DOI: 10.1016/j.ejps.2021.106056] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Revised: 10/09/2021] [Accepted: 10/31/2021] [Indexed: 11/17/2022]
Abstract
Permeability assessment of small molecules through the blood-brain barrier (BBB) plays a significant role in the development of effective central nervous system (CNS) drug candidates. Since in vivo methods for BBB permeability estimation require a lot of time and resources, in silico and in vitro approaches are becoming increasingly popular nowadays for faster and more economical predictions in early phases of drug discovery. In this work, through application of in vitro parallel artificial membrane permeability assay (PAMPA-BBB) and in silico computational methods we aimed to examine the passive permeability of eighteen compounds, which affect serotonin and dopamine levels in the CNS. The data set was consisted of novel six human dopamine transporter (hDAT) substrates that were previously identified as the most promising lead compounds for further optimisation to achieve neuroprotective effect, twelve approved CNS drugs, and their related compounds. Firstly, PAMPA methods was used to experimentally determine effective BBB permeability (Pe) for all studied compounds and obtained results were further submitted for quantitative structure permeability relationship (QSPR) analysis. QSPR models were built by using three different statistical methods: stepwise multiple linear regression (MLR), partial least square (PLS), and support-vector machine (SVM), while their predictive capability was tested through internal and external validation. Obtained statistical parameters (MLR- R2pred=-0.10; PLS- R2pred=0.64, r2m=0.69, r/2m=0.44; SVM- R2pred=0.57, r2m=0.72, r/2m=0.55) indicated that the SVM model is superior over others. The most important molecular descriptors (H0p and SolvEMt_3D) were identified and used to propose structural modifications of the examined compounds in order to improve their BBB permeability. Moreover, steered molecular dynamics (SMD) simulation was employed to comprehensively investigate the permeability pathway of compounds through a lipid bilayer. Taken together, the created QSPR model could be used as a reliable and fast pre-screening tool for BBB permeability prediction of structurally related CNS compounds, while performed MD simulations provide a good foundation for future in silico examination.
Collapse
Affiliation(s)
- Milica Radan
- University of Belgrade - Faculty of Pharmacy, Department of Pharmaceutical, Chemistry, Vojvode Stepe 450, 11000 Belgrade, Serbia
| | - Teodora Djikic
- University of Belgrade - Faculty of Pharmacy, Department of Pharmaceutical, Chemistry, Vojvode Stepe 450, 11000 Belgrade, Serbia.
| | - Darija Obradovic
- University of Belgrade - Faculty of Pharmacy, Department of Pharmaceutical, Chemistry, Vojvode Stepe 450, 11000 Belgrade, Serbia
| | - Katarina Nikolic
- University of Belgrade - Faculty of Pharmacy, Department of Pharmaceutical, Chemistry, Vojvode Stepe 450, 11000 Belgrade, Serbia.
| |
Collapse
|
43
|
Lynch MJ, Gobbo OL. Advances in Non-Animal Testing Approaches towards Accelerated Clinical Translation of Novel Nanotheranostic Therapeutics for Central Nervous System Disorders. NANOMATERIALS (BASEL, SWITZERLAND) 2021; 11:2632. [PMID: 34685073 PMCID: PMC8538557 DOI: 10.3390/nano11102632] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Revised: 09/21/2021] [Accepted: 10/01/2021] [Indexed: 12/11/2022]
Abstract
Nanotheranostics constitute a novel drug delivery system approach to improving systemic, brain-targeted delivery of diagnostic imaging agents and pharmacological moieties in one rational carrier platform. While there have been notable successes in this field, currently, the clinical translation of such delivery systems for the treatment of neurological disorders has been limited by the inadequacy of correlating in vitro and in vivo data on blood-brain barrier (BBB) permeation and biocompatibility of nanomaterials. This review aims to identify the most contemporary non-invasive approaches for BBB crossing using nanotheranostics as a novel drug delivery strategy and current non-animal-based models for assessing the safety and efficiency of such formulations. This review will also address current and future directions of select in vitro models for reducing the cumbersome and laborious mandate for testing exclusively in animals. It is hoped these non-animal-based modelling approaches will facilitate researchers in optimising promising multifunctional nanocarriers with a view to accelerating clinical testing and authorisation applications. By rational design and appropriate selection of characterised and validated models, ranging from monolayer cell cultures to organ-on-chip microfluidics, promising nanotheranostic particles with modular and rational design can be screened in high-throughput models with robust predictive power. Thus, this article serves to highlight abbreviated research and development possibilities with clinical translational relevance for developing novel nanomaterial-based neuropharmaceuticals for therapy in CNS disorders. By generating predictive data for prospective nanomedicines using validated in vitro models for supporting clinical applications in lieu of requiring extensive use of in vivo animal models that have notable limitations, it is hoped that there will be a burgeoning in the nanotherapy of CNS disorders by virtue of accelerated lead identification through screening, optimisation through rational design for brain-targeted delivery across the BBB and clinical testing and approval using fewer animals. Additionally, by using models with tissue of human origin, reproducible therapeutically relevant nanomedicine delivery and individualised therapy can be realised.
Collapse
Affiliation(s)
- Mark J. Lynch
- School of Pharmacy and Pharmaceutical Sciences, Panoz Building, Trinity College Dublin, D02 PN40 Dublin, Ireland
| | - Oliviero L. Gobbo
- School of Pharmacy and Pharmaceutical Sciences, Panoz Building, Trinity College Dublin, D02 PN40 Dublin, Ireland
| |
Collapse
|
44
|
Huang K, Castiaux A, Podicheti R, Rusch DB, Martin RS, Baker LA. A Hybrid Nanofiber/Paper Cell Culture Platform for Building a 3D Blood-brain Barrier Model. SMALL METHODS 2021; 5:2100592. [PMID: 34541301 PMCID: PMC8445000 DOI: 10.1002/smtd.202100592] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Indexed: 05/16/2023]
Abstract
The blood brain barrier (BBB) protects the central nervous system from toxins and pathogens in the blood by regulating permeation of molecules through the barrier interface. In vitro BBB models described to date reproduce some aspects of BBB functionality, but also suffer from incomplete phenotypic expression of brain endothelial traits, difficulty in reproducibility and fabrication, or overall cost. To address these limitations, we describe a three-dimensional (3D) BBB model based on a hybrid paper/nanofiber scaffold. The cell culture platform utilizes lens paper as a framework to accommodate 3D culture of astrocytes. An electrospun nanofiber layer is coated onto one face of the paper to mimic the basement membrane and support growth of an organized two-dimensional layer of endothelial cells (ECs). Human induced pluripotent stem cell-derived ECs and astrocytes are co-cultured to develop a human BBB model. Morphological and spatial organization of model are validated with confocal microscopy. Measurements of transendothelial resistance and permeability demonstrate the BBB model develops a high-quality barrier and responds to hyperosmolar treatments. RNA-sequencing shows introduction of astrocytes both regulates EC tight junction proteins and improves endothelial phenotypes related to vasculogenesis. This model shows promise as a model platform for future in vitro studies of the BBB.
Collapse
Affiliation(s)
- Kaixiang Huang
- Department of Chemistry, Indiana University Bloomington, 800 E. Kirkwood Avenue, Bloomington, Indiana 47405, USA
| | - Andre Castiaux
- Department of Chemistry and Center for Additive Manufacturing, Saint Louis University, 3501 Laclede Avenue, St. Louis, Missouri 63103, USA
| | - Ram Podicheti
- Center for Genomics and Bioinformatics, Indiana University Bloomington, 1001 East Third St., Bloomington, Indiana 47405, USA
| | - Douglas B Rusch
- Center for Genomics and Bioinformatics, Indiana University Bloomington, 1001 East Third St., Bloomington, Indiana 47405, USA
| | - R Scott Martin
- Department of Chemistry and Center for Additive Manufacturing, Saint Louis University, 3501 Laclede Avenue, St. Louis, Missouri 63103, USA
| | - Lane A Baker
- Department of Chemistry, Indiana University Bloomington, 800 E. Kirkwood Avenue, Bloomington, Indiana 47405, USA
| |
Collapse
|
45
|
Hanafy AS, Dietrich D, Fricker G, Lamprecht A. Blood-brain barrier models: Rationale for selection. Adv Drug Deliv Rev 2021; 176:113859. [PMID: 34246710 DOI: 10.1016/j.addr.2021.113859] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Revised: 06/21/2021] [Accepted: 07/01/2021] [Indexed: 01/21/2023]
Abstract
Brain delivery is a broad research area, the outcomes of which are far hindered by the limited permeability of the blood-brain barrier (BBB). Over the last century, research has been revealing the BBB complexity and the crosstalk between its cellular and molecular components. Pathologically, BBB alterations may precede as well as be concomitant or lead to brain diseases. To simulate the BBB and investigate options for drug delivery, several in vitro, in vivo, ex vivo, in situ and in silico models are used. Hundreds of drug delivery vehicles successfully pass preclinical trials but fail in clinical settings. Inadequate selection of BBB models is believed to remarkably impact the data reliability leading to unsatisfactory results in clinical trials. In this review, we suggest a rationale for BBB model selection with respect to the addressed research question and downstream applications. The essential considerations of an optimal BBB model are discussed.
Collapse
Affiliation(s)
- Amira Sayed Hanafy
- Department of Pharmaceutics, Institute of Pharmacy, University of Bonn, Bonn, Germany; Department of Pharmaceutics and Pharmaceutical Technology, Faculty of Pharmacy, Pharos University in Alexandria, Alexandria, Egypt
| | - Dirk Dietrich
- Department of Neurosurgery, University Hospital Bonn, Bonn, Germany
| | - Gert Fricker
- Institute of Pharmacy and Molecular Biotechnology, Ruprecht-Karls University, Heidelberg, Germany
| | - Alf Lamprecht
- Department of Pharmaceutics, Institute of Pharmacy, University of Bonn, Bonn, Germany.
| |
Collapse
|
46
|
Jenkins EPW, Finch A, Gerigk M, Triantis IF, Watts C, Malliaras GG. Electrotherapies for Glioblastoma. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2021; 8:e2100978. [PMID: 34292672 PMCID: PMC8456216 DOI: 10.1002/advs.202100978] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Revised: 05/20/2021] [Indexed: 05/08/2023]
Abstract
Non-thermal, intermediate frequency (100-500 kHz) electrotherapies present a unique therapeutic strategy to treat malignant neoplasms. Here, pulsed electric fields (PEFs) which induce reversible or irreversible electroporation (IRE) and tumour-treating fields (TTFs) are reviewed highlighting the foundations, advances, and considerations of each method when applied to glioblastoma (GBM). Several biological aspects of GBM that contribute to treatment complexity (heterogeneity, recurrence, resistance, and blood-brain barrier(BBB)) and electrophysiological traits which are suggested to promote glioma progression are described. Particularly, the biological responses at the cellular and molecular level to specific parameters of the electrical stimuli are discussed offering ways to compare these parameters despite the lack of a universally adopted physical description. Reviewing the literature, a disconnect is found between electrotherapy techniques and how they target the biological complexities of GBM that make treatment difficult in the first place. An attempt is made to bridge the interdisciplinary gap by mapping biological characteristics to different methods of electrotherapy, suggesting important future research topics and directions in both understanding and treating GBM. To the authors' knowledge, this is the first paper that attempts an in-tandem assessment of the biological effects of different aspects of intermediate frequency electrotherapy methods, thus offering possible strategies toward GBM treatment.
Collapse
Affiliation(s)
- Elise P. W. Jenkins
- Division of Electrical EngineeringDepartment of EngineeringUniversity of CambridgeCambridgeCB3 0FAUK
| | - Alina Finch
- Institute of Cancer and Genomic ScienceUniversity of BirminghamBirminghamB15 2TTUK
| | - Magda Gerigk
- Division of Electrical EngineeringDepartment of EngineeringUniversity of CambridgeCambridgeCB3 0FAUK
| | - Iasonas F. Triantis
- Department of Electrical and Electronic EngineeringCity, University of LondonLondonEC1V 0HBUK
| | - Colin Watts
- Institute of Cancer and Genomic ScienceUniversity of BirminghamBirminghamB15 2TTUK
| | - George G. Malliaras
- Division of Electrical EngineeringDepartment of EngineeringUniversity of CambridgeCambridgeCB3 0FAUK
| |
Collapse
|
47
|
Appelt-Menzel A, Oerter S, Mathew S, Haferkamp U, Hartmann C, Jung M, Neuhaus W, Pless O. Human iPSC-Derived Blood-Brain Barrier Models: Valuable Tools for Preclinical Drug Discovery and Development? ACTA ACUST UNITED AC 2021; 55:e122. [PMID: 32956578 DOI: 10.1002/cpsc.122] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Translating basic biological knowledge into applications remains a key issue for effectively tackling neurodegenerative, neuroinflammatory, or neuroendocrine disorders. Efficient delivery of therapeutics across the neuroprotective blood-brain barrier (BBB) still poses a demanding challenge for drug development targeting central nervous system diseases. Validated in vitro models of the BBB could facilitate effective testing of drug candidates targeting the brain early in the drug discovery process during lead generation. We here review the potential of mono- or (isogenic) co-culture BBB models based on brain capillary endothelial cells (BCECs) derived from human-induced pluripotent stem cells (hiPSCs), and compare them to several available BBB in vitro models from primary human or non-human cells and to rodent in vivo models, as well as to classical and widely used barrier models [Caco-2, parallel artificial membrane permeability assay (PAMPA)]. In particular, we are discussing the features and predictivity of these models and how hiPSC-derived BBB models could impact future discovery and development of novel CNS-targeting therapeutics. © 2020 The Authors.
Collapse
Affiliation(s)
- Antje Appelt-Menzel
- Fraunhofer Institute for Silicate Research ISC, Translational Center Regenerative Therapies (TLC-RT), Röntgenring 11, Würzburg, Germany.,University Hospital Würzburg, Chair Tissue Engineering and Regenerative Medicine (TERM), Röntgenring 11, Würzburg, Germany
| | - Sabrina Oerter
- Fraunhofer Institute for Silicate Research ISC, Translational Center Regenerative Therapies (TLC-RT), Röntgenring 11, Würzburg, Germany.,University Hospital Würzburg, Chair Tissue Engineering and Regenerative Medicine (TERM), Röntgenring 11, Würzburg, Germany
| | - Sanjana Mathew
- University Hospital Würzburg, Chair Tissue Engineering and Regenerative Medicine (TERM), Röntgenring 11, Würzburg, Germany
| | - Undine Haferkamp
- Fraunhofer IME ScreeningPort, Schnackenburgallee 114, Hamburg, Germany
| | - Carla Hartmann
- University Hospital Halle, University Clinic and Outpatient Clinic for Psychiatry, Psychotherapy, and Psychosomatic Medicine, Julius-Kuehn-Strasse 7, Halle (Saale), Germany
| | - Matthias Jung
- University Hospital Halle, University Clinic and Outpatient Clinic for Psychiatry, Psychotherapy, and Psychosomatic Medicine, Julius-Kuehn-Strasse 7, Halle (Saale), Germany
| | - Winfried Neuhaus
- AIT Austrian Institute of Technology GmbH, Center Health and Bioresources, Competence Unit Molecular Diagnostics, Giefinggasse 4, Vienna, Austria
| | - Ole Pless
- Fraunhofer IME ScreeningPort, Schnackenburgallee 114, Hamburg, Germany
| |
Collapse
|
48
|
Mesiti F, Gaspar A, Chavarria D, Maruca A, Rocca R, Gil Martins E, Barreiro S, Silva R, Fernandes C, Gul S, Keminer O, Alcaro S, Borges F. Mapping Chromone-3-Phenylcarboxamide Pharmacophore: Quid Est Veritas? J Med Chem 2021; 64:11169-11182. [PMID: 34269579 DOI: 10.1021/acs.jmedchem.1c00510] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Chromone-3-phenylcarboxamides (Crom-1 and Crom-2) were identified as potent, selective, and reversible inhibitors of human monoamine oxidase B (hMAO-B). Since they exhibit some absorption, distribution, metabolism, and excretion (ADME)-toxicity liabilities, new derivatives were synthesized to map the chemical structural features that compose the pharmacophore, a process vital for lead optimization. Structure-activity relationship data, supported by molecular docking studies, provided a rationale for the contribution of the heterocycle's rigidity, the carbonyl group, and the benzopyran heteroatom for hMAO-B inhibitory activity. From the study, N-(3-chlorophenyl)-4H-thiochromone-3-carboxamide (31) (hMAO-B IC50 = 1.52 ± 0.15 nM) emerged as a reversible tight binding inhibitor with an improved pharmacological profile. In in vitro ADME-toxicity studies, compound 31 showed a safe cytotoxicity profile in Caco-2, SH-SY5Y, HUVEC, HEK-293, and MCF-7 cells, did not present cardiotoxic effects, and did not affect P-gp transport activity. Compound 31 also protected SH-SY5Y cells from iron(III)-induced damage. Collectively, these studies highlighted compound 31 as the first-in-class and a suitable candidate for in vivo preclinical investigation.
Collapse
Affiliation(s)
- Francesco Mesiti
- Dipartimento di Scienze della Salute, Università "Magna Græcia" di Catanzaro, Campus "Salvatore Venuta", Catanzaro 88100, Italy.,Net4Science srl, Academic Spinoff, Università "Magna Græcia" di Catanzaro, Campus "Salvatore Venuta", Catanzaro 88100, Italy.,CIQUP/Department of Chemistry and Biochemistry, Faculty of Sciences, University of Porto, Porto 4169-007, Portugal
| | - Alexandra Gaspar
- CIQUP/Department of Chemistry and Biochemistry, Faculty of Sciences, University of Porto, Porto 4169-007, Portugal
| | - Daniel Chavarria
- CIQUP/Department of Chemistry and Biochemistry, Faculty of Sciences, University of Porto, Porto 4169-007, Portugal
| | - Annalisa Maruca
- Dipartimento di Scienze della Salute, Università "Magna Græcia" di Catanzaro, Campus "Salvatore Venuta", Catanzaro 88100, Italy.,Net4Science srl, Academic Spinoff, Università "Magna Græcia" di Catanzaro, Campus "Salvatore Venuta", Catanzaro 88100, Italy
| | - Roberta Rocca
- Net4Science srl, Academic Spinoff, Università "Magna Græcia" di Catanzaro, Campus "Salvatore Venuta", Catanzaro 88100, Italy.,Department of Experimental and Clinical Medicine, "Magna Græcia" University of Catanzaro, Campus "S. Venuta", Viale Europa, Germaneto, Catanzaro 88100, Italy
| | - Eva Gil Martins
- UCIBIO-REQUIMTE, Laboratory of Toxicology, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, Porto 4050-313, Portugal
| | - Sandra Barreiro
- UCIBIO-REQUIMTE, Laboratory of Toxicology, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, Porto 4050-313, Portugal
| | - Renata Silva
- UCIBIO-REQUIMTE, Laboratory of Toxicology, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, Porto 4050-313, Portugal
| | - Carlos Fernandes
- CIQUP/Department of Chemistry and Biochemistry, Faculty of Sciences, University of Porto, Porto 4169-007, Portugal
| | - Sheraz Gul
- Fraunhofer Institute for Translational Medicine and Pharmacology, Hamburg 22525, Germany.,Fraunhofer Cluster of Excellence for Immune-Mediated Diseases CIMD, Hamburg Site, Hamburg 22525, Germany
| | - Oliver Keminer
- Fraunhofer Institute for Translational Medicine and Pharmacology, Hamburg 22525, Germany.,Fraunhofer Cluster of Excellence for Immune-Mediated Diseases CIMD, Hamburg Site, Hamburg 22525, Germany
| | - Stefano Alcaro
- Dipartimento di Scienze della Salute, Università "Magna Græcia" di Catanzaro, Campus "Salvatore Venuta", Catanzaro 88100, Italy
| | - Fernanda Borges
- CIQUP/Department of Chemistry and Biochemistry, Faculty of Sciences, University of Porto, Porto 4169-007, Portugal
| |
Collapse
|
49
|
Staicu CE, Jipa F, Axente E, Radu M, Radu BM, Sima F. Lab-on-a-Chip Platforms as Tools for Drug Screening in Neuropathologies Associated with Blood-Brain Barrier Alterations. Biomolecules 2021; 11:916. [PMID: 34205550 PMCID: PMC8235582 DOI: 10.3390/biom11060916] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Revised: 06/15/2021] [Accepted: 06/16/2021] [Indexed: 12/19/2022] Open
Abstract
Lab-on-a-chip (LOC) and organ-on-a-chip (OOC) devices are highly versatile platforms that enable miniaturization and advanced controlled laboratory functions (i.e., microfluidics, advanced optical or electrical recordings, high-throughput screening). The manufacturing advancements of LOCs/OOCs for biomedical applications and their current limitations are briefly discussed. Multiple studies have exploited the advantages of mimicking organs or tissues on a chip. Among these, we focused our attention on the brain-on-a-chip, blood-brain barrier (BBB)-on-a-chip, and neurovascular unit (NVU)-on-a-chip applications. Mainly, we review the latest developments of brain-on-a-chip, BBB-on-a-chip, and NVU-on-a-chip devices and their use as testing platforms for high-throughput pharmacological screening. In particular, we analyze the most important contributions of these studies in the field of neurodegenerative diseases and their relevance in translational personalized medicine.
Collapse
Affiliation(s)
- Cristina Elena Staicu
- Department of Anatomy, Animal Physiology and Biophysics, Faculty of Biology, University of Bucharest, 050095 Bucharest, Romania;
- Center for Advanced Laser Technologies, National Institute for Laser, Plasma and Radiation Physics, 077125 Măgurele, Romania; (F.J.); (E.A.); (F.S.)
| | - Florin Jipa
- Center for Advanced Laser Technologies, National Institute for Laser, Plasma and Radiation Physics, 077125 Măgurele, Romania; (F.J.); (E.A.); (F.S.)
| | - Emanuel Axente
- Center for Advanced Laser Technologies, National Institute for Laser, Plasma and Radiation Physics, 077125 Măgurele, Romania; (F.J.); (E.A.); (F.S.)
| | - Mihai Radu
- Department of Life and Environmental Physics, ‘Horia Hulubei’ National Institute for Physics and Nuclear Engineering, 077125 Măgurele, Romania;
| | - Beatrice Mihaela Radu
- Department of Anatomy, Animal Physiology and Biophysics, Faculty of Biology, University of Bucharest, 050095 Bucharest, Romania;
| | - Felix Sima
- Center for Advanced Laser Technologies, National Institute for Laser, Plasma and Radiation Physics, 077125 Măgurele, Romania; (F.J.); (E.A.); (F.S.)
| |
Collapse
|
50
|
申 杰, 杨 迪, 陈 梦, 郭 新. [Effects of length and chemical modification on the activation of vascular endothelial cells induced by multi walled carbon nanotubes]. BEIJING DA XUE XUE BAO. YI XUE BAN = JOURNAL OF PEKING UNIVERSITY. HEALTH SCIENCES 2021; 53:439-446. [PMID: 34145842 PMCID: PMC8220036 DOI: 10.19723/j.issn.1671-167x.2021.03.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 01/07/2021] [Indexed: 06/12/2023]
Abstract
OBJECTIVE To investigate the effects of multi-walled carbon nanotubes (MWCNTs) with different length or chemical modification on endothelial cell activation and to explore the role of nucleotide-binding oligomerization domain-like receptor family pyrin domain containing 3 (NLRP3) inflammasome. METHODS MWCNTs were characterized by dynamic light scattering (DLS) after being suspended in culture medium. The immortalized mouse cerebral microvascular endothelial cell line b.End3 was treated with short MWCNTs (S-MWCNT, 0.5 to 2 μm), long MWCNTs (L-MWCNT, 10 to 30 μm) and the above long MWCNTs functionalized by carboxyl-(L-MWCNT-COOH), amino-(L-MWCNT-NH2) or hydroxyl-(L-MWCNT-OH) modification. Cytotoxicity of MWCNTs in b.End3 cells was determined by cell counting kit-8 (CCK-8) assay and lactate dehydrogenase (LDH) release assay, and non-toxic low dose was selected for subsequent experiments. Effects of all types of MWCNTs on the endothelial activation of b.End3 were determined by the measurement of vascular cell adhesion molecule-1 (VCAM-1) concentration in cell supernatant and adhesion assay of human monocytic cell line THP-1 to b.End3.To further elucidate the mechanism involved, the protein expressions of nucleotide-binding oligomerization domain-like receptor family pyrin domain containing 3(NLRP3) in cells treated with S-MWCNT, L-MWCNT and L-MWCNT-COOH were measured by Western blot. RESULTS At a higher concentration (125 μg/cm2) and treated for 24 h, all types of MWCNTs significantly inhibited viability of b.End3 cells. At a sub-toxic concentration (6.25 μg/cm2), all types of MWCNTs treated for 12 h significantly induced the activation of b.End3 cells, as evidenced by the elevated VCAM-1 release and THP-1 adhesion. Compared with S-MWCNT, L-MWCNT significantly promoted endothelial cell activation. L-MWCNT and L-MWCNT-COOH activated b.End3 cells to a similar extent. Furthermore, treatment with S-MWCNT, L-MWCNT and L-MWCNT-COOH increased NLRP3 expression in a time-dependent manner at 6.25 μg/cm2. Compared with S-MWCNT, cells treated with L-MWCNT for 4 h and 12 h exhibited significantly increased protein expressions of NLRP3. However, no significant differences were detected in the level of NLRP3 protein in cells treated with L-MWCNT and L-MWCNT-COOH. CONCLUSION Compared with the surface chemical modification, length changes of MWCNTs exerted more influence on endothelial cell activation, which may be related to the activation of NLRP3 inflammasome. Our study contributes further understanding of the impact of MWCNTs on endothelial cells, which may have implications for the improvement of safety evaluation of MWCNTs.
Collapse
Affiliation(s)
- 杰 申
- />北京大学公共卫生学院劳动卫生与环境卫生学系,北京 100191Department of Occupational and Environmental Health, Peking University School of Public Health, Beijing 100191, China
| | - 迪 杨
- />北京大学公共卫生学院劳动卫生与环境卫生学系,北京 100191Department of Occupational and Environmental Health, Peking University School of Public Health, Beijing 100191, China
| | - 梦圆 陈
- />北京大学公共卫生学院劳动卫生与环境卫生学系,北京 100191Department of Occupational and Environmental Health, Peking University School of Public Health, Beijing 100191, China
| | - 新彪 郭
- />北京大学公共卫生学院劳动卫生与环境卫生学系,北京 100191Department of Occupational and Environmental Health, Peking University School of Public Health, Beijing 100191, China
| |
Collapse
|