1
|
Han Y, Zhou S, Xiang H, Zhang Q, Zhang H, Zhang Y, Lin Q, Zhang Z, Zhang L, Huang S. A brain-targeting dl-3-n-butylphthalide prodrug to treat ischemic stroke. Eur J Med Chem 2025; 290:117474. [PMID: 40120494 DOI: 10.1016/j.ejmech.2025.117474] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2025] [Revised: 02/24/2025] [Accepted: 02/28/2025] [Indexed: 03/25/2025]
Abstract
Stroke is an acute cerebrovascular disease worldwide, featured by stubbornly high mortality and morbidity. 3-n-butylphthalide (NBP) is a first-line agent to treat ischemic stroke, but its unsatisfactorily intracephalic bioavailability is detrimental to the therapeutic efficacy. We have previously developed a series of prodrugs analogous to NBP, however, their conversion rate in the targeted sites cannot meet the clinical demand. Hence, it is imperative to ameliorate the bioavailability and cellular uptake efficiency of the prodrugs, which may be driven by potential energy consuming cationic transporters. Accordingly, we fabricated a novel prodrug named DB-10, which were used tertiary amine to possess active targeting capacity and connected through amide bonds. Moreover, it is noteworthy that DB-10 could rapidly convert into active original drug in the brain to treat acute injury caused by ischemia-reperfusion (I/R). The cytotoxicity of DB-10 was equivalent to that of NBP and would not cause obviously undesired damage in vivo. These encouraging results demonstrate that DB-10 could easily increase the accumulation in the brain site, and significantly improve the treatment effect for ischemic stroke, indicating this biosafety prodrug holds tremendous potential for clinical application prospects.
Collapse
Affiliation(s)
- Yikun Han
- Key Laboratory of Drug Targeting and Drug Delivery Systems of Ministry of Education, West China School of Pharmacy, Sichuan University, Chengdu, 610065, China
| | - Shuhao Zhou
- College of Polymer Science and Engineering, Sichuan University, Chengdu, 610000, China
| | - Honglin Xiang
- Key Laboratory of Drug Targeting and Drug Delivery Systems of Ministry of Education, West China School of Pharmacy, Sichuan University, Chengdu, 610065, China
| | - Qiang Zhang
- Key Laboratory of Drug Targeting and Drug Delivery Systems of Ministry of Education, West China School of Pharmacy, Sichuan University, Chengdu, 610065, China
| | - Hanming Zhang
- Key Laboratory of Drug Targeting and Drug Delivery Systems of Ministry of Education, West China School of Pharmacy, Sichuan University, Chengdu, 610065, China
| | - Yicong Zhang
- Key Laboratory of Drug Targeting and Drug Delivery Systems of Ministry of Education, West China School of Pharmacy, Sichuan University, Chengdu, 610065, China
| | - Qing Lin
- Key Laboratory of Drug Targeting and Drug Delivery Systems of Ministry of Education, West China School of Pharmacy, Sichuan University, Chengdu, 610065, China
| | - Zhirong Zhang
- Key Laboratory of Drug Targeting and Drug Delivery Systems of Ministry of Education, West China School of Pharmacy, Sichuan University, Chengdu, 610065, China
| | - Ling Zhang
- College of Polymer Science and Engineering, Sichuan University, Chengdu, 610000, China; Med-X Center for Materials, Sichuan University, Chengdu, 610000, China.
| | - Shiqi Huang
- College of Polymer Science and Engineering, Sichuan University, Chengdu, 610000, China.
| |
Collapse
|
2
|
Králová A, Montaser AB, Tampio J, Adla SK, Jalkanen A, Rysä J, Huttunen KM. A novel paracetamol derivative alleviates lipopolysaccharide-induced neuroinflammation. Eur J Pharmacol 2025; 995:177409. [PMID: 39986592 DOI: 10.1016/j.ejphar.2025.177409] [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: 10/01/2024] [Revised: 02/10/2025] [Accepted: 02/19/2025] [Indexed: 02/24/2025]
Abstract
Neuroinflammation has been implicated as a pathological contributor to several neurodegenerative disorders. Increasing evidence suggests that paracetamol (PCM, acetaminophen) has unappreciated anti-neuroinflammatory properties. However, PCM possesses hepatotoxicity in higher dosages, which are needed for achieving therapeutic concentrations in the brain. To lessen this effect and improve drug efficacy, PCM was in this study converted into an L-type amino acid transporter 1 (LAT1)-utilizing derivative and tested whether this LAT1-mediated delivery approach could enhance the relief of neuroinflammation, using both in vitro and in vivo lipopolysaccharide (LPS)-stimulated models. The gained results confirmed the derivative's improved transport into mouse primary astrocytes, immortalized microglia (BV2), and human immortalized microglia (SV40) via LAT1. In the LPS-stimulated BV2 model, the derivative effectively reduced the prostaglandin E2 (PGE2) level by 57% compared to the LPS treatment. Moreover, a more profound reduction of brain PGE2 production was confirmed in the LPS-stimulated mouse model. Finally, the global proteome of the whole mouse brain revealed that the derivative was able to reverse the altered expression of several inflammatory biomarkers, including ras-related C3 botulinum toxin substrate 1 (Rac1), cytochrome c oxidase subunit 2 (COX2), phospholipid phosphatase-related protein type 2 (Plppr2), ubiquitin-conjugating enzyme E2 variant 1 (Ube2v1) and A-kinase anchor protein 1, mitochondrial (Akap1).
Collapse
Affiliation(s)
- Adéla Králová
- School of Pharmacy, Faculty of Health Sciences, University of Eastern Finland, P.O. Box 1627, FI-70211, Kuopio, Finland.
| | - Ahmed B Montaser
- School of Pharmacy, Faculty of Health Sciences, University of Eastern Finland, P.O. Box 1627, FI-70211, Kuopio, Finland
| | - Janne Tampio
- School of Pharmacy, Faculty of Health Sciences, University of Eastern Finland, P.O. Box 1627, FI-70211, Kuopio, Finland
| | - Santosh Kumar Adla
- School of Pharmacy, Faculty of Health Sciences, University of Eastern Finland, P.O. Box 1627, FI-70211, Kuopio, Finland
| | - Aaro Jalkanen
- School of Pharmacy, Faculty of Health Sciences, University of Eastern Finland, P.O. Box 1627, FI-70211, Kuopio, Finland
| | - Jaana Rysä
- School of Pharmacy, Faculty of Health Sciences, University of Eastern Finland, P.O. Box 1627, FI-70211, Kuopio, Finland
| | - Kristiina M Huttunen
- School of Pharmacy, Faculty of Health Sciences, University of Eastern Finland, P.O. Box 1627, FI-70211, Kuopio, Finland
| |
Collapse
|
3
|
Yu D, Liu M, Ding Q, Wu Y, Wang T, Song L, Li X, Qian K, Cheng Z, Gu M, Li Z. Molecular imaging-guided diagnosis and treatment integration for brain diseases. Biomaterials 2025; 316:123021. [PMID: 39705925 DOI: 10.1016/j.biomaterials.2024.123021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2024] [Revised: 12/03/2024] [Accepted: 12/13/2024] [Indexed: 12/23/2024]
Abstract
In practical clinical scenarios, improved diagnostic methods have been developed for the precise visualization of molecular targets using molecular imaging in brain diseases. Recently, the introduction of innovative molecular imaging modalities across both macroscopic and mesoscopic dimensions, with remarkable specificity and spatial resolution, has expanded the scope of applications beyond diagnostic testing, with the potential to guide therapeutic interventions, offering real-time feedback in the context of brain therapy. The molecular imaging-guided integration of diagnosis and treatment holds the potential to revolutionize disease management by enabling the real-time monitoring of treatment responses and therapy adjustments. Given the vibrant and ever-evolving nature of this field, this review provides an integrated picture on molecular image-guided diagnosis and treatment integration for brain diseases involving the basic concepts, significant breakthroughs, and recent trends. In addition, based on the current achievements, some critical challenges are also discussed.
Collapse
Affiliation(s)
- Donghu Yu
- Brain Glioma Center & Department of Neurosurgery, International Science and Technology Cooperation Base for Research and Clinical Techniques for Brain Glioma Diagnosis and Treatment, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Menghao Liu
- Tsinghua-Peking Center for Life Sciences, Tsinghua University, Beijing, 100084, China
| | - Qihang Ding
- Department of Chemistry, Korea University, Seoul, 02841, South Korea.
| | - Youxian Wu
- Brain Glioma Center & Department of Neurosurgery, International Science and Technology Cooperation Base for Research and Clinical Techniques for Brain Glioma Diagnosis and Treatment, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Tianqing Wang
- Brain Glioma Center & Department of Neurosurgery, International Science and Technology Cooperation Base for Research and Clinical Techniques for Brain Glioma Diagnosis and Treatment, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Litong Song
- State Key Laboratory of Drug Research, Molecular Imaging Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Xiaoyu Li
- State Key Laboratory of Drug Research, Molecular Imaging Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Kun Qian
- State Key Laboratory of Drug Research, Molecular Imaging Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Zhen Cheng
- State Key Laboratory of Drug Research, Molecular Imaging Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China.
| | - Meijia Gu
- School of Pharmaceutical Sciences, Wuhan University, Wuhan, 430071, China.
| | - Zhiqiang Li
- Brain Glioma Center & Department of Neurosurgery, International Science and Technology Cooperation Base for Research and Clinical Techniques for Brain Glioma Diagnosis and Treatment, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China.
| |
Collapse
|
4
|
Kulkarni R, Kumari S, Dhapola R, Sharma P, Singh SK, Medhi B, HariKrishnaReddy D. Association Between the Gut Microbiota and Alzheimer's Disease: An Update on Signaling Pathways and Translational Therapeutics. Mol Neurobiol 2025; 62:4499-4519. [PMID: 39460901 DOI: 10.1007/s12035-024-04545-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Accepted: 10/10/2024] [Indexed: 10/28/2024]
Abstract
Alzheimer's disease (AD) is a cognitive disease with high morbidity and mortality. In AD patients, the diversity of the gut microbiota is altered, which influences pathology through the gut-brain axis. Probiotic therapy alleviates pathological and psychological consequences by restoring the diversity of the gut microbial flora. This study addresses the role of altered gut microbiota in the progression of neuroinflammation, which is a major hallmark of AD. This process begins with the activation of glial cells, leading to the release of proinflammatory cytokines and the modulation of cholinergic anti-inflammatory pathways. Short-chain fatty acids, which are bacterial metabolites, provide neuroprotective effects and maintain blood‒brain barrier integrity. Furthermore, the gut microbiota stimulates oxidative stress and mitochondrial dysfunction, which promote AD progression. The signaling pathways involved in gut dysbiosis-mediated neuroinflammation-mediated promotion of AD include cGAS-STING, C/EBPβ/AEP, RAGE, TLR4 Myd88, and the NLRP3 inflammasome. Preclinical studies have shown that natural extracts such as Ganmaidazao extract, isoorentin, camelia oil, Sparassis crispa-1, and xanthocerasides improve gut health and can delay the worsening of AD. Clinical studies using probiotics such as Bifidobacterium spp., yeast beta-glucan, and drugs such as sodium oligomannate and rifaximine have shown improvements in gut health, resulting in the amelioration of AD symptoms. This study incorporates the most current research on the pathophysiology of AD involving the gut microbiota and highlights the knowledge gaps that need to be filled to develop potent therapeutics against AD.
Collapse
Affiliation(s)
- Rutweek Kulkarni
- Advanced Pharmacology and Neuroscience Laboratory, Department of Pharmacology, School of Health Sciences, Central University of Punjab, Bathinda, 151401, Punjab, India
| | - Sneha Kumari
- Advanced Pharmacology and Neuroscience Laboratory, Department of Pharmacology, School of Health Sciences, Central University of Punjab, Bathinda, 151401, Punjab, India
| | - Rishika Dhapola
- Advanced Pharmacology and Neuroscience Laboratory, Department of Pharmacology, School of Health Sciences, Central University of Punjab, Bathinda, 151401, Punjab, India
| | - Prajjwal Sharma
- Advanced Pharmacology and Neuroscience Laboratory, Department of Pharmacology, School of Health Sciences, Central University of Punjab, Bathinda, 151401, Punjab, India
| | - Sunil K Singh
- Department of Biochemistry, School of Basic Sciences, Central University of Punjab, Ghudda, Bathinda, Punjab, India
| | - Bikash Medhi
- Department of Pharmacology, Post Graduate Institute of Medical Education and Research, Chandigarh, 160012, India
| | - Dibbanti HariKrishnaReddy
- Advanced Pharmacology and Neuroscience Laboratory, Department of Pharmacology, School of Health Sciences, Central University of Punjab, Bathinda, 151401, Punjab, India.
| |
Collapse
|
5
|
Tega Y, Kawauchi Y, Akanuma SI, Inagaki M, Tachikawa M, Hosoya KI. In vitro characterization of taurine transport using the human brain microvascular endothelial cell line as a human blood-brain barrier model. Drug Metab Pharmacokinet 2025; 61:101040. [PMID: 39923278 DOI: 10.1016/j.dmpk.2024.101040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Revised: 09/24/2024] [Accepted: 11/13/2024] [Indexed: 02/11/2025]
Abstract
Taurine, a sulfur-containing β-amino acid, has various roles in the brain including cellular osmoregulation and neuroprotection. For adequate supply to the brain, taurine has to pass through the blood-brain barrier (BBB); however, the associated mechanism behind crossing the human BBB is not fully understood. Therefore, we characterized taurine transport in vitro using the human brain microvascular endothelial (hCMEC/D3) cell line, a model of human BBB function. [3H]Taurine uptake by hCMEC/D3 cells exhibited time-, as well as extracellular Na+- and Cl--dependence. The uptake was saturable with a Km of 19 μM and was inhibited by GABA at an IC50 of 328 μM, which were similar to Km values of taurine transporter (TauT)-mediated transport of taurine and GABA, respectively, suggesting that TauT is a major contributor to taurine uptake. For distribution to the brain, taurine must undergo cellular efflux after uptake. Taurine efflux from hCMEC/D3 cells increased for at least 60 min, and monocarboxylate transporter 7 (MCT7)-targeted siRNA significantly reduced MCT7 mRNA levels and [3H]taurine efflux by 93 % and 12 %, respectively, suggesting that MCT7 partly contributes to taurine efflux from hCMEC/D3 cells. Taken together, these results suggest that TauT and MCT7 function cooperatively in the human BBB.
Collapse
Affiliation(s)
- Yuma Tega
- Department of Pharmaceutics, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, Toyama, Japan
| | - Yusuke Kawauchi
- Department of Pharmaceutics, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, Toyama, Japan
| | - Shin-Ichi Akanuma
- Department of Pharmaceutics, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, Toyama, Japan.
| | - Mai Inagaki
- Graduate School of Biomedical Sciences, Tokushima University, Tokushima, Japan
| | - Masanori Tachikawa
- Graduate School of Biomedical Sciences, Tokushima University, Tokushima, Japan
| | - Ken-Ichi Hosoya
- Department of Pharmaceutics, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, Toyama, Japan
| |
Collapse
|
6
|
Tang S, Han EL, Mitchell MJ. Peptide-functionalized nanoparticles for brain-targeted therapeutics. Drug Deliv Transl Res 2025:10.1007/s13346-025-01840-w. [PMID: 40164912 DOI: 10.1007/s13346-025-01840-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/17/2025] [Indexed: 04/02/2025]
Abstract
Despite the rapid development of nanoparticle (NP)-based drug delivery systems, intravenous delivery of drugs to the brain remains a major challenge due to various biological barriers. To achieve therapeutic effects, NP-encapsulated drugs must avoid accumulation in off-target organs and selectively deliver to the brain, successfully cross the blood-brain barrier (BBB), and reach the target cells in the brain. Conjugating receptor-specific ligands to the surface of NPs is a promising technique for engineering NPs to overcome these barriers. Specifically, peptides as brain-targeting ligands have been of increasing interest given their ease of synthesis, low cytotoxicity, and strong affinity to target proteins. The success of peptides as targeting ligands is largely due to the diverse strategies of designing and modifying peptides with favorable properties, including membrane permeability and multi-receptor targeting. Here, we review the design and implementation of peptide-functionalized NP systems for neurological disease applications. We also explore advances in rational peptide design strategies for brain targeting, including using generative deep-learning models to computationally design new peptides.
Collapse
Affiliation(s)
- Sophia Tang
- Department of Bioengineering, School of Engineering and Applied Science, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Emily L Han
- Department of Bioengineering, School of Engineering and Applied Science, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Michael J Mitchell
- Department of Bioengineering, School of Engineering and Applied Science, University of Pennsylvania, Philadelphia, PA, 19104, USA.
- Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA.
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA.
- Penn Institute for RNA Innovation, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA.
- Institute for Immunology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA.
- Cardiovascular Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA.
- Institute for Regenerative Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA.
| |
Collapse
|
7
|
Chung KJ, Abdelhafez YG, Spencer BA, Jones T, Tran Q, Nardo L, Chen MS, Sarkar S, Medici V, Lyo V, Badawi RD, Cherry SR, Wang G. Quantitative PET imaging and modeling of molecular blood-brain barrier permeability. Nat Commun 2025; 16:3076. [PMID: 40159510 PMCID: PMC11955546 DOI: 10.1038/s41467-025-58356-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Accepted: 03/19/2025] [Indexed: 04/02/2025] Open
Abstract
Neuroimaging of blood-brain barrier permeability has been instrumental in identifying its broad involvement in neurological and systemic diseases. However, current methods evaluate the blood-brain barrier mainly as a structural barrier. Here we developed a non-invasive positron emission tomography method in humans to measure the blood-brain barrier permeability of molecular radiotracers that cross the blood-brain barrier through its molecule-specific transport mechanism. Our method uses high-temporal resolution dynamic imaging and kinetic modeling for multiparametric imaging and quantification of the blood-brain barrier permeability-surface area product of molecular radiotracers. We show, in humans, our method can resolve blood-brain barrier permeability across three radiotracers and demonstrate its utility in studying brain aging and brain-body interactions in metabolic dysfunction-associated steatotic liver inflammation. Our method opens new directions to effectively study the molecular permeability of the human blood-brain barrier in vivo using the large catalogue of available molecular positron emission tomography tracers.
Collapse
Affiliation(s)
- Kevin J Chung
- Department of Radiology, University of California Davis Health, Sacramento, CA, USA
| | - Yasser G Abdelhafez
- Department of Radiology, University of California Davis Health, Sacramento, CA, USA
| | - Benjamin A Spencer
- Department of Radiology, University of California Davis Health, Sacramento, CA, USA
| | - Terry Jones
- Department of Radiology, University of California Davis Health, Sacramento, CA, USA
| | - Quyen Tran
- Department of Radiology, University of California Davis Health, Sacramento, CA, USA
| | - Lorenzo Nardo
- Department of Radiology, University of California Davis Health, Sacramento, CA, USA
| | - Moon S Chen
- Department of Internal Medicine, University of California Davis Health, Sacramento, CA, USA
| | - Souvik Sarkar
- Department of Internal Medicine, University of California Davis Health, Sacramento, CA, USA
- Division of Gastroenterology and Hepatology, University of California Davis Health, Sacramento, CA, USA
| | - Valentina Medici
- Department of Internal Medicine, University of California Davis Health, Sacramento, CA, USA
- Division of Gastroenterology and Hepatology, University of California Davis Health, Sacramento, CA, USA
| | - Victoria Lyo
- Department of Surgery, University of California Davis Health, Sacramento, CA, USA
- Center for Alimentary and Metabolic Sciences, University of California Davis Health, Sacramento, CA, USA
| | - Ramsey D Badawi
- Department of Radiology, University of California Davis Health, Sacramento, CA, USA
- Department of Biomedical Engineering, University of California at Davis, Davis, CA, USA
| | - Simon R Cherry
- Department of Radiology, University of California Davis Health, Sacramento, CA, USA
- Department of Biomedical Engineering, University of California at Davis, Davis, CA, USA
| | - Guobao Wang
- Department of Radiology, University of California Davis Health, Sacramento, CA, USA.
| |
Collapse
|
8
|
Pomeroy J, Borczyk M, Kawalec M, Hajto J, Carlson E, Svärd S, Verma S, Bareke E, Boratyńska-Jasińska A, Dymkowska D, Mellado-Ibáñez A, Laight D, Zabłocki K, Occhipinti A, Majewska L, Angione C, Majewski J, Yegutkin GG, Korostynski M, Zabłocka B, Górecki DC. Spatiotemporal diversity in molecular and functional abnormalities in the mdx dystrophic brain. Mol Med 2025; 31:108. [PMID: 40114059 PMCID: PMC11924731 DOI: 10.1186/s10020-025-01109-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Accepted: 01/28/2025] [Indexed: 03/22/2025] Open
Abstract
Duchenne muscular dystrophy (DMD) is characterized by progressive muscle degeneration and neuropsychiatric abnormalities. Loss of full-length dystrophins is both necessary and sufficient to initiate DMD. These isoforms are expressed in the hippocampus, cerebral cortex (Dp427c), and cerebellar Purkinje cells (Dp427p). However, our understanding of the consequences of their absence, which is crucial for developing targeted interventions, remains inadequate. We combined RNA sequencing with genome-scale metabolic modelling (GSMM), immunodetection, and mitochondrial assays to investigate dystrophic alterations in the brains of the mdx mouse model of DMD. The cerebra and cerebella were analysed separately to discern the roles of Dp427c and Dp427p, respectively. Investigating these regions at 10 days (10d) and 10 weeks (10w) followed the evolution of abnormalities from development to early adulthood. These time points also encompass periods before onset and during muscle inflammation, enabling assessment of the potential damage caused by inflammatory mediators crossing the dystrophic blood-brain barrier. For the first time, we demonstrated that transcriptomic and functional dystrophic alterations are unique to the cerebra and cerebella and vary substantially between 10d and 10w. The common anomalies involved altered numbers of retained introns and spliced exons across mdx transcripts, corresponding with alterations in the mRNA processing pathways. Abnormalities in the cerebra were significantly more pronounced in younger mice. The top enriched pathways included those related to metabolism, mRNA processing, and neuronal development. GSMM indicated dysregulation of glucose metabolism, which corresponded with GLUT1 protein downregulation. The cerebellar dystrophic transcriptome, while significantly altered, showed an opposite trajectory to that of the cerebra, with few changes identified at 10 days. These late defects are specific and indicate an impact on the functional maturation of the cerebella that occurs postnatally. Although no classical neuroinflammation markers or microglial activation were detected at 10 weeks, specific differences indicate that inflammation impacts DMD brains. Importantly, some dystrophic alterations occur late and may therefore be amenable to therapeutic intervention, offering potential avenues for mitigating DMD-related neuropsychiatric defects.
Collapse
Affiliation(s)
- Joanna Pomeroy
- School of Medicine, Pharmacy and Biomedical Sciences, University of Portsmouth, White Swan Road, Portsmouth, PO1 2DT, UK
| | - Malgorzata Borczyk
- Department of Molecular Neuropharmacology, Maj Institute of Pharmacology, Polish Academy of Sciences, 12 Smętna Str., 31-343, Krakow, Poland
| | - Maria Kawalec
- Molecular Biology Unit, Mossakowski Medical Research Institute, Polish Academy of Sciences, Warsaw, Poland
| | - Jacek Hajto
- Department of Molecular Neuropharmacology, Maj Institute of Pharmacology, Polish Academy of Sciences, 12 Smętna Str., 31-343, Krakow, Poland
| | - Emma Carlson
- Department of Human Genetics, McGill University, Montreal, QC, H3A 1B1, Canada
| | - Samuel Svärd
- MediCity Research Laboratory and InFLAMES Flagship, University of Turku, Turku, Finland
| | - Suraj Verma
- School of Computing, Engineering and Digital Technologies, Teesside University, Middlesbrough, UK
| | - Eric Bareke
- Department of Human Genetics, McGill University, Montreal, QC, H3A 1B1, Canada
| | - Anna Boratyńska-Jasińska
- Molecular Biology Unit, Mossakowski Medical Research Institute, Polish Academy of Sciences, Warsaw, Poland
| | - Dorota Dymkowska
- Laboratory of Cellular Metabolism, Nencki Institute of Experimental Biology, Warsaw, Poland
| | - Alvaro Mellado-Ibáñez
- School of Medicine, Pharmacy and Biomedical Sciences, University of Portsmouth, White Swan Road, Portsmouth, PO1 2DT, UK
| | - David Laight
- School of Medicine, Pharmacy and Biomedical Sciences, University of Portsmouth, White Swan Road, Portsmouth, PO1 2DT, UK
| | - Krzysztof Zabłocki
- Laboratory of Cellular Metabolism, Nencki Institute of Experimental Biology, Warsaw, Poland
| | - Annalisa Occhipinti
- School of Computing, Engineering and Digital Technologies, Teesside University, Middlesbrough, UK
| | - Loydie Majewska
- Department of Pediatrics, McGill University, McGill Health Centre Glen Site, 1001 Decarie Blvd, EM02210, Montreal, QC, H4A 3J1, Canada
| | - Claudio Angione
- School of Computing, Engineering and Digital Technologies, Teesside University, Middlesbrough, UK
| | - Jacek Majewski
- Department of Human Genetics, McGill University, Montreal, QC, H3A 1B1, Canada
| | - Gennady G Yegutkin
- MediCity Research Laboratory and InFLAMES Flagship, University of Turku, Turku, Finland
| | - Michal Korostynski
- Department of Molecular Neuropharmacology, Maj Institute of Pharmacology, Polish Academy of Sciences, 12 Smętna Str., 31-343, Krakow, Poland
| | - Barbara Zabłocka
- Molecular Biology Unit, Mossakowski Medical Research Institute, Polish Academy of Sciences, Warsaw, Poland
| | - Dariusz C Górecki
- School of Medicine, Pharmacy and Biomedical Sciences, University of Portsmouth, White Swan Road, Portsmouth, PO1 2DT, UK.
| |
Collapse
|
9
|
Taggi V, Schäfer AM, Kinzi J, Ritz D, Seibert I, Oswald S, Zu Schwabedissen HEM. Targeted and Untargeted Proteomics-based Comparison of Adenoviral Infected hCMEC/D3 and hBMEC as a Human Brain Endothelial Cells to Study the OATP2B1 Transporter. Mol Neurobiol 2025:10.1007/s12035-025-04807-7. [PMID: 40085356 DOI: 10.1007/s12035-025-04807-7] [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: 11/04/2024] [Accepted: 02/22/2025] [Indexed: 03/16/2025]
Abstract
The blood-brain barrier (BBB) is essential for central nervous system (CNS) homeostasis by regulating permeability between the bloodstream and brain. This study evaluates the immortalized human brain capillary endothelial cell lines hCMEC/D3 and hBMEC for their use as a brain endothelial cells to investigate the OATP2B1 transporter following adenoviral infection. We assessed the impact of adenoviral-mediated OATP2B1 expression on BBB marker proteins and transporters using targeted and untargeted mass spectrometry-based proteomics. Targeted proteomics identified measurable levels of endothelial markers PECAM1 and CDH5 in hCMEC/D3, whereas these markers were undetectable in hBMEC. Both cell lines exhibited similar Pgp levels, while BCRP was absent in hCMEC/D3. The expression of uptake transporters was also evaluated, revealing comparable levels of GLUT1, ENT1, MCT1 and OAT7 in hCMEC/D3 and hBMEC. Although OATP2B1 levels did not significantly increase post-infection in targeted proteomics, untargeted proteomics confirmed enhanced OATP2B1 expression. Other BBB markers and transporters remained unaffected in both cell lines. Notably, hCMEC/D3 demonstrated a stronger BBB phenotype, indicated by higher expression of BBB markers and transporters, while adenoviral infection was more effective in hBMEC. The differences between targeted and untargeted proteomics underscore the need for diverse methods to verify protein expression levels. This comparative analysis provides insights into the strengths and limitations of hCMEC/D3 and hBMEC for BBB research, particularly regarding drug transport mechanisms.
Collapse
Affiliation(s)
- Valerio Taggi
- Biopharmacy, Department of Pharmaceutical Sciences, University of Basel, Klingelbergstrasse 50, 4056, Basel, Switzerland
| | - Anima M Schäfer
- Biopharmacy, Department of Pharmaceutical Sciences, University of Basel, Klingelbergstrasse 50, 4056, Basel, Switzerland
| | - Jonny Kinzi
- Biopharmacy, Department of Pharmaceutical Sciences, University of Basel, Klingelbergstrasse 50, 4056, Basel, Switzerland
| | - Danilo Ritz
- Proteomics Core Facility, Biozentrum, University of Basel, Basel, Switzerland
| | - Isabell Seibert
- Biopharmacy, Department of Pharmaceutical Sciences, University of Basel, Klingelbergstrasse 50, 4056, Basel, Switzerland
| | - Stefan Oswald
- Institute of Pharmacology and Toxicology, Rostock University Medical Center, Rostock, Germany
| | | |
Collapse
|
10
|
Strazielle N, Blondel S, Confais J, El Khoury R, Contamin H, Ghersi-Egea JF. Molecular determinants of neuroprotection in blood-brain interfaces of the cynomolgus monkey. Front Pharmacol 2025; 16:1523819. [PMID: 40144668 PMCID: PMC11936797 DOI: 10.3389/fphar.2025.1523819] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2024] [Accepted: 02/14/2025] [Indexed: 03/28/2025] Open
Abstract
The blood-brain barrier (BBB) formed by the cerebral microvessel endothelium and the blood-CSF barrier (BCSFB) formed by the choroid plexus epithelium impact the cerebral bioavailability of drugs and endogenous molecules that contribute to neuroinflammatory and neurodegenerative diseases. Species specificities in tight junction proteins and efflux transporters governing the barrier functions of these interfaces hamper the direct translation of pharmacokinetic and pathophysiological data from rodents to human. We defined the molecular composition of tight junctions and identified the efflux transporters present at the BBB and BCSFB of cynomolgus monkey to assess whether this species is a relevant alternative to rodents. Choroid plexuses, cerebral microvessels, cortex and cerebellum were isolated from adult cynomolgus monkeys, and analysed by RT-qPCR and immunohistochemistry. Results were compared with data available in the literature for rat and human. In monkeys as in rat and human, claudin-5 in the BBB and claudin-1, -2, -3 in the BCSFB were landmark tight junction proteins. ABCB1 was strictly associated with the BBB, and ABCC1 was predominant at the BCSFB compared to the BBB. The monkey, like human, differed from rat by the localization of ABCG2 protein in choroidal vessels, a low expression of ABCC4 and SLC22A8 in the BBB, and the presence of SLC47A1 at the BCSFB. While the main characteristics of brain barriers are common to all three species, cynomolgus monkey and human share specificities in the expression and localization of selected claudins and efflux transporters that are not met in rat.
Collapse
Affiliation(s)
- Nathalie Strazielle
- BIP Facility, Fluid Team, Lyon Neurosciences Research Center, INSERM U1028, UMR CNRS, University Lyon 1, Bron, France
- Brain-i, Lyon, France
| | - Sandrine Blondel
- BIP Facility, Fluid Team, Lyon Neurosciences Research Center, INSERM U1028, UMR CNRS, University Lyon 1, Bron, France
| | | | - Rita El Khoury
- BIP Facility, Fluid Team, Lyon Neurosciences Research Center, INSERM U1028, UMR CNRS, University Lyon 1, Bron, France
| | | | - Jean-François Ghersi-Egea
- BIP Facility, Fluid Team, Lyon Neurosciences Research Center, INSERM U1028, UMR CNRS, University Lyon 1, Bron, France
| |
Collapse
|
11
|
Hammarlund-Udenaes M, Loryan I. Assessing central nervous system drug delivery. Expert Opin Drug Deliv 2025; 22:421-439. [PMID: 39895003 DOI: 10.1080/17425247.2025.2462767] [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: 10/15/2024] [Revised: 01/15/2025] [Accepted: 01/31/2025] [Indexed: 02/04/2025]
Abstract
INTRODUCTION Delivering drugs to the central nervous system (CNS) remains a major challenge due to the blood-brain barrier, restricting the entry of drugs into the brain. This limitation contributes to the ongoing lack of effective treatments for CNS diseases. To improve the process of drug discovery and development, it is crucial to streamline methods that measure clinically relevant parameters, allowing for good selection of drug candidates. AREA COVERED In this paper, we discuss the essential prerequisites for successful CNS drug delivery and review relevant methods. We emphasize the need for closer collaboration between in vitro and in vivo scientists to improve the relevance of these methods and increase the success rate of developing effective CNS therapies. While our focus is on small molecule drugs, we also touch on some aspects of larger molecules. EXPERT OPINION Significant progress has been made in recent years in method development and their application. However, there is still work to be done before the use of in silico models, in vitro cell systems, and AI can consistently offer meaningful correlations and relationships to clinical data. This gap is partly due to limited patient data, but a lot can be achieved through in vivo research in animal models.
Collapse
Affiliation(s)
| | - Irena Loryan
- Translational PKPD Group, Department of Pharmacy, Uppsala University, Uppsala, Sweden
| |
Collapse
|
12
|
Zou L, Chien HC, Pade D, Li Y, Nguyen M, Bhamidipati RK, Wang Z, Enogieru OJ, Wahlstrom J. Considerations in K p,uu,brain-based Strategy for Selecting CNS-targeted Drug Candidates with Sufficient Target Coverage and Substantial Pharmacodynamic Effect. AAPS J 2025; 27:52. [PMID: 40021548 DOI: 10.1208/s12248-025-01035-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2024] [Accepted: 02/04/2025] [Indexed: 03/03/2025] Open
Abstract
Kp,uu,brain is a critical parameter for evaluating the brain penetration of CNS-targeted compounds, reflecting the ratio of unbound drug concentration in the brain to that in the plasma. While Kp,uu,brain is widely used in the pharmaceutical industry to assess brain exposure, the fidelity of translating Kp,uu,brain to target coverage and pharmacodynamic (PD) effect remains uncertain. This study explores the effectiveness of Kp,uu,brain-based strategies in identifying drug candidates with sufficient target coverage and substantial PD effect. By analyzing reported Kp,uu,brain, unbound drug concentrations in the brain and IC50 values against pharmacological targets for 17 drugs including anticonvulsants, antidepressants, antipsychotics, and antimicrobials, our study demonstrated that while in vitro and in vivo models work well for rank ordering compounds with high Kp,uu,brain, this parameter does not necessarily translate into adequate target coverage (Cu/IC50). In addition, by leveraging PK and PD profiles of 18 drugs measured from human glioblastoma tumors, our study showed that target coverage (glioblastoma Cu/5xIC50) generally correlates well with PD effect. Additionally, Kp,uu,brain tumor is a better indicator for glioblastoma PD effect than Kp,uu,brain, suggesting that intact BBB model may not adequately reflect the barrier heterogeneity in brain tumors such as glioblastoma. In conclusion, while Kp,uu,brain provides an insight on the extent of brain penetration, our study highlighted the need for integrative approaches combining Kp,uu,brain data with comprehensive PK/PD analysis to prioritize CNS-targeted drug candidates with sufficient target coverage and substantial PD effect.
Collapse
Affiliation(s)
- Ling Zou
- Pharmacokinetics and Drug Metabolism, Amgen Inc., South San Francisco, California, USA
| | - Huan-Chieh Chien
- Pharmacokinetics and Drug Metabolism, Amgen Inc., South San Francisco, California, USA
| | - Devendra Pade
- Pharmacokinetics and Drug Metabolism, Amgen Inc., South San Francisco, California, USA
| | - Yanfei Li
- Pharmacokinetics and Drug Metabolism, Amgen Inc., South San Francisco, California, USA
| | - Minhkhoi Nguyen
- Pharmacokinetics and Drug Metabolism, Amgen Inc., South San Francisco, California, USA
| | | | - Zhe Wang
- Pharmacokinetics and Drug Metabolism, Amgen Inc., South San Francisco, California, USA
| | | | - Jan Wahlstrom
- Pharmacokinetics and Drug Metabolism, Amgen Inc., Thousand Oaks, California, 91320, USA.
| |
Collapse
|
13
|
Tournier N, Langer O. Imaging the Activity of Efflux Transporters at the Blood-Brain Barrier in Neurologic Diseases: Radiotracer Selection Criteria. J Nucl Med 2025:jnumed.124.269322. [PMID: 40015923 DOI: 10.2967/jnumed.124.269322] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2024] [Accepted: 02/10/2025] [Indexed: 03/01/2025] Open
Abstract
Efflux transporters of the adenosine triphosphate-binding cassette (ABC) superfamily, such as P-glycoprotein (P-gp/ABCB1) and breast cancer resistance protein (BCRP/ABCG2), are highly expressed at the blood-brain barrier (BBB), where they contribute to maintaining brain homeostasis. P-gp may serve as an imaging biomarker to assess the contribution of BBB functionality rather than integrity to the onset or progression of various neurologic diseases. Considerable efforts have been made to develop radiolabeled P-gp substrates to assess cerebral P-gp activity with PET. However, initially developed radiotracers have limited clinical utility as they lack sensitivity to detect moderate, physiologically relevant changes in cerebral P-gp activity. Learning from this molecular imaging area has called for specific criteria, different from those classically used for other central nervous system targets, for developing and selecting suitable PET tracers to study ABC transporter activity at the BBB in different neurologic diseases.
Collapse
Affiliation(s)
- Nicolas Tournier
- Laboratoire d'Imagerie Biomédicale Multimodale, Service Hospitalier Frédéric Joliot, Université Paris-Saclay, CEA, CNRS, INSERM, Orsay, France;
| | - Oliver Langer
- Department of Clinical Pharmacology, Medical University of Vienna, Vienna, Austria; and
- Department of Biomedical Imaging and Image-Guided Therapy, Division of Nuclear Medicine, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
14
|
Meng Q, Zhang X, Chen Y, Yang H, Liu J, Yang Z, Lei J, Lu F, Hao D, Feng L, Wang Y. Nanoparticles modified with glucose analogs to enhance the permeability of the blood-brain barrier and their accumulation in the epileptic brain. J Mater Chem B 2025; 13:2810-2819. [PMID: 39871635 DOI: 10.1039/d4tb02476a] [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: 01/29/2025]
Abstract
Drug delivery for epilepsy treatment faces enormous challenges, where the sole focus on enhancing the ability of drugs to penetrate the blood-brain barrier (BBB) through ligand modification is insufficient because of the absence of seizure-specific drug accumulation. In this study, an amphipathic drug carrier with a glucose transporter (GLUT)-targeting capability was synthesised by conjugating 2-deoxy-2-amino-D-glucose (2-DG) to the model carrier DSPE-PEG2k. A 2-DG-modified nano drug delivery system (NDDS) possessing robust stability and favourable biocompatibility was then fabricated using the nanoprecipitation method. The results showed that the 2-DG-modified NDDS exhibited enhanced cellular uptake by brain capillary endothelial cells and neuronal cells. Most importantly, the 2-DG-modified NDDS exhibited enhanced BBB penetration and brain accumulation, especially in the epileptic brain, thus achieving seizure-based on-demand drug delivery. Our study provides a simple and smart strategy for the delivery of anti-seizure medicines.
Collapse
Affiliation(s)
- Qian Meng
- Department of Neurology, the First Affiliated Hospital of Anhui Medical University, Hefei 230022, Anhui, P. R. China.
| | - Xiaoyu Zhang
- Department of Neurology, the First Affiliated Hospital of Anhui Medical University, Hefei 230022, Anhui, P. R. China.
- Anhui Public Health Clinical Center, Hefei 230041, Anhui, P. R. China
| | - Yuwen Chen
- Department of Neurology, the First Affiliated Hospital of Anhui Medical University, Hefei 230022, Anhui, P. R. China.
| | - Hao Yang
- School of Basic Medical Sciences, Anhui Medical University, Hefei 230032, Anhui, P. R. China.
| | - Jinshuai Liu
- Department of Neurology, the First Affiliated Hospital of Anhui Medical University, Hefei 230022, Anhui, P. R. China.
| | - Zifan Yang
- Department of Neurology, the First Affiliated Hospital of Anhui Medical University, Hefei 230022, Anhui, P. R. China.
| | - Jianxiang Lei
- Department of Neurology, the First Affiliated Hospital of Anhui Medical University, Hefei 230022, Anhui, P. R. China.
| | - Fengqing Lu
- Department of Neurology, the First Affiliated Hospital of Anhui Medical University, Hefei 230022, Anhui, P. R. China.
| | - Dengyuan Hao
- State Key Laboratory of Polymer Physics and Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, Jilin, P. R. China
| | - Lijie Feng
- School of Basic Medical Sciences, Anhui Medical University, Hefei 230032, Anhui, P. R. China.
- Biopharmaceutical Research Institute, Anhui Medical University, Hefei 230032, Anhui, P. R. China
| | - Yu Wang
- Department of Neurology, the First Affiliated Hospital of Anhui Medical University, Hefei 230022, Anhui, P. R. China.
- Anhui Public Health Clinical Center, Hefei 230041, Anhui, P. R. China
| |
Collapse
|
15
|
Jackwerth M, Mairinger S, Rausch I, Weber M, Jorda A, Nics L, Langsteger W, Zeitlinger M, Hacker M, Langer O. Effect of probenecid on the whole-body disposition of 6-bromo-7-[ 11C]methylpurine in humans assessed with long axial field-of-view PET/CT. Eur J Nucl Med Mol Imaging 2025:10.1007/s00259-025-07121-5. [PMID: 39920267 DOI: 10.1007/s00259-025-07121-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Accepted: 01/27/2025] [Indexed: 02/09/2025]
Abstract
PURPOSE Multidrug resistance-associated proteins (MRPs) have a widespread tissue distribution. They play an important role in drug disposition and drug-drug interactions (DDIs) and have been associated with various diseases. PET with 6-bromo-7-[11C]methylpurine ([11C]BMP) has been used to assess MRP1 function in the brain and lungs of mice. [11C]BMP crosses cellular membranes by passive diffusion followed by intracellular conjugation with glutathione and MRP1-mediated efflux of the radiolabelled glutathione-conjugate. In this study, we assessed the effect of the prototypical organic anion transporter inhibitor probenecid on the whole-body disposition of [11C]BMP to examine its suitability for measuring the function of MRP1 and possibly other MRP subtypes across multiple tissues. METHODS Seven healthy volunteers (3 women, 4 men) underwent two dynamic whole-body PET scans on a long axial field-of-view (LAFOV) PET/CT system after intravenous injection of [11C]BMP, without and with pre-treatment with a single oral dose of probenecid. Volumes of interest were outlined for several MRP-expressing tissues (cerebral cortex, cerebellum, choroid plexus, retina, lungs, myocardium, skeletal muscle, kidneys, and liver). Tissue time-activity curves were corrected for the contribution of vascular radioactivity and the elimination rate constant (kE, h- 1) was calculated as a parameter for tissue MRP function. RESULTS Radioactivity was primarily excreted into the urinary bladder and urinary clearance was significantly decreased after probenecid administration (- 50 ± 16%). Following probenecid administration, kE was significantly decreased in the kidneys (- 43 ± 20%), liver (- 18 ± 15%), myocardium (- 16 ± 12%), skeletal muscle (- 51 ± 34%), and retina (- 57 ± 29%, non-blood-corrected). CONCLUSION Our study highlights the great potential of LAFOV PET/CT to assess drug disposition and transporter-mediated DDIs in humans at a whole-body, multi-tissue level. Due to the slow elimination of [11C]BMP-derived radioactivity from the human brain, [11C]BMP appears unsuitable to measure cerebral MRP1 function in humans, but it may be used to assess the function of MRP1 and possibly other MRP subtypes in various peripheral tissues. TRIAL REGISTRATION EudraCT 2021-006348-29. Registered 15 December 2021.
Collapse
Affiliation(s)
- Matthias Jackwerth
- Department of Clinical Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Severin Mairinger
- Department of Clinical Pharmacology, Medical University of Vienna, Vienna, Austria
- Department of Biomedical Imaging and Image-guided Therapy, Medical University of Vienna, Vienna, Austria
| | - Ivo Rausch
- QIMP Team, Center for Medical Physics and Biomedical Engineering, Medical University of Vienna, Vienna, Austria
| | - Maria Weber
- Department of Clinical Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Anselm Jorda
- Department of Clinical Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Lukas Nics
- Department of Biomedical Imaging and Image-guided Therapy, Medical University of Vienna, Vienna, Austria
| | - Werner Langsteger
- Department of Biomedical Imaging and Image-guided Therapy, Medical University of Vienna, Vienna, Austria
| | - Markus Zeitlinger
- Department of Clinical Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Marcus Hacker
- Department of Biomedical Imaging and Image-guided Therapy, Medical University of Vienna, Vienna, Austria
| | - Oliver Langer
- Department of Clinical Pharmacology, Medical University of Vienna, Vienna, Austria.
- Department of Biomedical Imaging and Image-guided Therapy, Medical University of Vienna, Vienna, Austria.
| |
Collapse
|
16
|
Saadh MJ, Allela OQB, Kareem RA, Sanghvi G, Menon SV, Sharma P, Tomar BS, Sharma A, Sameer HN, Hamad AK, Athab ZH, Adil M. From Gut to Brain: The Impact of Short-Chain Fatty Acids on Brain Cancer. Neuromolecular Med 2025; 27:10. [PMID: 39821841 DOI: 10.1007/s12017-025-08830-8] [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: 12/17/2024] [Accepted: 01/06/2025] [Indexed: 01/19/2025]
Abstract
The primary source of short-chain fatty acids (SCFAs), now recognized as critical mediators of host health, particularly in the context of neurobiology and cancer development, is the gut microbiota's fermentation of dietary fibers. Recent research highlights the complex influence of SCFAs, such as acetate, propionate, and butyrate, on brain cancer progression. These SCFAs impact immune modulation and the tumor microenvironment, particularly in brain tumors like glioma. They play a critical role in regulating cellular processes, including apoptosis, cell differentiation, and inflammation. Moreover, studies have linked SCFAs to maintaining the integrity of the blood-brain barrier (BBB), suggesting a protective role in preventing tumor infiltration and enhancing anti-tumor immunity. As our understanding of the gut-brain axis deepens, it becomes increasingly important to investigate SCFAs' therapeutic potential in brain cancer management. Looking into how SCFAs affect brain tumor cells and the environment around them could lead to new ways to prevent and treat these diseases, which could lead to better outcomes for people who are dealing with these challenging cancers.
Collapse
Affiliation(s)
- Mohamed J Saadh
- Faculty of Pharmacy, Middle East University, Amman, 11831, Jordan.
| | | | | | - Gaurav Sanghvi
- Department of Microbiology, Faculty of Science, Marwadi University Research Center, Marwadi University, Rajkot, Gujarat, 360003, India
| | - Soumya V Menon
- Department of Chemistry and Biochemistry, School of Sciences, JAIN (Deemed to Be University), Bangalore, Karnataka, India
| | - Pawan Sharma
- Department of Sciences, Vivekananda Global University, Jaipur, Rajasthan, 303012, India
| | - Balvir S Tomar
- Institute of Pediatric Gastroenterology and Hepatology, National Institute of Medical Sciences, NIMS University Rajasthan, Jaipur, India
| | - Aanchal Sharma
- Department of Medical Lab Sciences, Chandigarh Group of Colleges-Jhanjeri, Mohali, Punjab, 140307, India
| | - Hayder Naji Sameer
- Collage of Pharmacy, National University of Science and Technology, Dhi Qar, 64001, Iraq
| | | | - Zainab H Athab
- Department of Pharmacy, Al-Zahrawi University College, Karbala, Iraq
| | - Mohaned Adil
- Pharmacy College, Al-Farahidi University, Baghdad, Iraq
| |
Collapse
|
17
|
Gülave B, Lesmana A, de Lange EC, van Hasselt JGC. Do P-glycoprotein-mediated drug-drug interactions at the blood-brain barrier impact morphine brain distribution? J Pharmacokinet Pharmacodyn 2025; 52:11. [PMID: 39776000 PMCID: PMC11706904 DOI: 10.1007/s10928-024-09957-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Accepted: 12/01/2024] [Indexed: 01/11/2025]
Abstract
P-glycoprotein (P-gp) is a key efflux transporter and may be involved in drug-drug interactions (DDIs) at the blood-brain barrier (BBB), which could lead to changes in central nervous system (CNS) drug exposure. Morphine is a P-gp substrate and therefore a potential victim drug for P-gp mediated DDIs. It is however unclear if P-gp inhibitors can induce clinically relevant changes in morphine CNS exposure. Here, we used a physiologically-based pharmacokinetic (PBPK) model-based approach to evaluate the potential impact of DDIs on BBB transport of morphine by clinically relevant P-gp inhibitor drugs.The LeiCNS-PK3.0 PBPK model was used to simulate morphine distribution at the brain extracellular fluid (brainECF) for different clinical intravenous dosing regimens of morphine, alone or in combination with a P-gp inhibitor. We included 34 commonly used P-gp inhibitor drugs, with inhibitory constants and expected clinical P-gp inhibitor concentrations derived from literature. The DDI impact was evaluated by the change in brainECF exposure for morphine alone or in combination with different inhibitors. Our analysis demonstrated that P-gp inhibitors had a negligible effect on morphine brainECF exposure in the majority of simulated population, caused by low P-gp inhibition. Sensitivity analyses showed neither major effects of increasing the inhibitory concentration nor changing the inhibitory constant on morphine brainECF exposure. In conclusion, P-gp mediated DDIs on morphine BBB transport for the evaluated P-gp inhibitors are unlikely to induce meaningful changes in clinically relevant morphine CNS exposure. The developed CNS PBPK modeling approach provides a general approach for evaluating BBB transporter DDIs in humans.
Collapse
Affiliation(s)
- Berfin Gülave
- Division of Systems Pharmacology and Pharmacy, Leiden Academic Center for Drug Research, Leiden University, Einsteinweg 55, Leiden, 2333 CC, The Netherlands
| | - Ariel Lesmana
- Division of Systems Pharmacology and Pharmacy, Leiden Academic Center for Drug Research, Leiden University, Einsteinweg 55, Leiden, 2333 CC, The Netherlands
| | - Elizabeth Cm de Lange
- Division of Systems Pharmacology and Pharmacy, Leiden Academic Center for Drug Research, Leiden University, Einsteinweg 55, Leiden, 2333 CC, The Netherlands
| | - J G Coen van Hasselt
- Division of Systems Pharmacology and Pharmacy, Leiden Academic Center for Drug Research, Leiden University, Einsteinweg 55, Leiden, 2333 CC, The Netherlands.
| |
Collapse
|
18
|
Ogata S, Ito S, Masuda T, Ohtsuki S. Isolation method of brain microvessels from small frozen human brain tissue for blood-brain barrier protein expression analysis. Fluids Barriers CNS 2024; 21:106. [PMID: 39702171 DOI: 10.1186/s12987-024-00609-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2024] [Accepted: 12/10/2024] [Indexed: 12/21/2024] Open
Abstract
BACKGROUND Protein expression analysis of isolated brain microvessels provides valuable insights into the function of the blood-brain barrier (BBB). However, isolation of brain microvessels from human brain tissue, particularly in small quantities, poses significant challenges. This study presents a method for isolating brain microvessels from a small amount of frozen human brain tissue, adapting techniques from an established mouse brain capillary isolation method. METHODS Brain microvessel fractions were obtained from approximately 0.3 g of frozen human brain tissue (frontal cortex) using a bead homogenizer for homogenization, followed by purification with a combination of cell strainers and glass beads. Protein expression in the isolated human microvessel fractions and whole-brain lysates was analyzed by western blot and proteomic analysis. RESULTS Microscopic imaging confirmed the successful isolation of brain microvessels from frozen human brain tissue. Protein quantification assays demonstrated that the microvessel fraction yielded sufficient protein for detailed expression analysis. Western blot analysis revealed an enrichment of BBB-selective proteins including multidrug resistance 1 (MDR1)/ATP-binding cassette sub-family B member 1 (ABCB1), glucose transporter protein type 1 (GLUT1)/solute carrier family 2 member 1 (SLC2A1), and claudin 5 (CLDN5), in the brain microvessel fraction compared to whole-brain lysates. Multiple reaction monitoring quantification of six BBB-selective proteins-MDR1, breast cancer resistance protein (BCRP)/ATP binding cassette subfamily G member 2 (ABCG2), GLUT1, monocarboxylate transporter 1 (MCT1)/solute carrier family 16 member 1 (SLC16A1), transferrin receptor, and CLDN5-revealed expression levels consistent with those observed in larger human brain samples. Sequential Window Acquisition of all Theoretical Mass Spectra (SWATH-MS)-based quantitative proteomics further demonstrated significant enrichment of human microvascular endothelial cells in the isolated fraction, corroborating the findings from mouse models. CONCLUSIONS We successfully developed a method for isolation of brain microvessels from a small amount of frozen human brain tissue, facilitating detailed study of BBB proteome in aging or pathological conditions. This technique provides valuable insights into BBB dysfunction in central nervous system disorders and holds potential for improving brain-targeted drug delivery strategies.
Collapse
Affiliation(s)
- Seiryo Ogata
- Department of Pharmaceutical Microbiology, Graduate School of Pharmaceutical Sciences, Kumamoto University, 5-1 Oe-honmachi, Chuo-ku, Kumamoto, 862-0973, Japan
- Department of Environmental Medicine and Molecular Toxicology, Tohoku University Graduate School of Medicine, 2-1 Seiryo-machi, Aoba-ku, Sendai, 980-8575, Japan
| | - Shingo Ito
- Department of Pharmaceutical Microbiology, Graduate School of Pharmaceutical Sciences, Kumamoto University, 5-1 Oe-honmachi, Chuo-ku, Kumamoto, 862-0973, Japan.
- Department of Pharmaceutical Microbiology, Faculty of Life Sciences, Kumamoto University, 5-1 Oe-honmachi, Chuo-ku, Kumamoto, 862-0973, Japan.
| | - Takeshi Masuda
- Department of Pharmaceutical Microbiology, Graduate School of Pharmaceutical Sciences, Kumamoto University, 5-1 Oe-honmachi, Chuo-ku, Kumamoto, 862-0973, Japan
- Department of Pharmaceutical Microbiology, Faculty of Life Sciences, Kumamoto University, 5-1 Oe-honmachi, Chuo-ku, Kumamoto, 862-0973, Japan
- Institute for Advanced Biosciences, Keio University, Nipponkoku 403-1, Daihouji, Tsuruoka, Yamagata, 997-0017, Japan
| | - Sumio Ohtsuki
- Department of Pharmaceutical Microbiology, Graduate School of Pharmaceutical Sciences, Kumamoto University, 5-1 Oe-honmachi, Chuo-ku, Kumamoto, 862-0973, Japan
- Department of Pharmaceutical Microbiology, Faculty of Life Sciences, Kumamoto University, 5-1 Oe-honmachi, Chuo-ku, Kumamoto, 862-0973, Japan
| |
Collapse
|
19
|
You H, Geng S, Li S, Imani M, Brambilla D, Sun T, Jiang C. Recent advances in biomimetic strategies for the immunotherapy of glioblastoma. Biomaterials 2024; 311:122694. [PMID: 38959533 DOI: 10.1016/j.biomaterials.2024.122694] [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: 04/08/2024] [Revised: 06/22/2024] [Accepted: 06/26/2024] [Indexed: 07/05/2024]
Abstract
Immunotherapy is regarded as one of the most promising approaches for treating tumors, with a multitude of immunotherapeutic thoughts currently under consideration for the lethal glioblastoma (GBM). However, issues with immunotherapeutic agents, such as limited in vivo stability, poor blood-brain barrier (BBB) penetration, insufficient GBM targeting, and represented monotherapy, have hindered the success of immunotherapeutic interventions. Moreover, even with the aid of conventional drug delivery systems, outcomes remain suboptimal. Biomimetic strategies seek to overcome these formidable drug delivery challenges by emulating nature's intelligent structures and functions. Leveraging the variety of biological structures and functions, biomimetic drug delivery systems afford a versatile platform with enhanced biocompatibility for the co-delivery of diverse immunotherapeutic agents. Moreover, their inherent capacity to traverse the BBB and home in on GBM holds promise for augmenting the efficacy of GBM immunotherapy. Thus, this review begins by revisiting the various thoughts and agents on immunotherapy for GBM. Then, the barriers to successful GBM immunotherapy are analyzed, and the corresponding biomimetic strategies are explored from the perspective of function and structure. Finally, the clinical translation's current state and prospects of biomimetic strategy are addressed. This review aspires to provide fresh perspectives on the advancement of immunotherapy for GBM.
Collapse
Affiliation(s)
- Haoyu You
- Key Laboratory of Smart Drug Delivery/Innovative Center for New Drug Development of Immune Inflammatory Diseases (Ministry of Education), Minhang Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Department of Pharmaceutics, School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Shuo Geng
- Key Laboratory of Smart Drug Delivery/Innovative Center for New Drug Development of Immune Inflammatory Diseases (Ministry of Education), Minhang Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Department of Pharmaceutics, School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Shangkuo Li
- Key Laboratory of Smart Drug Delivery/Innovative Center for New Drug Development of Immune Inflammatory Diseases (Ministry of Education), Minhang Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Department of Pharmaceutics, School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Mohammad Imani
- Department of Science, Iran Polymer and Petrochemical Institute, Tehran 14977-13115, Iran; Center for Nanoscience and Nanotechnology, Institute for Convergence Science & Technology, Tehran 14588-89694, Iran
| | - Davide Brambilla
- Faculty of Pharmacy, University of Montreal, Montreal Quebec H3T 1J4, Canada
| | - Tao Sun
- Key Laboratory of Smart Drug Delivery/Innovative Center for New Drug Development of Immune Inflammatory Diseases (Ministry of Education), Minhang Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Department of Pharmaceutics, School of Pharmacy, Fudan University, Shanghai 201203, China.
| | - Chen Jiang
- Key Laboratory of Smart Drug Delivery/Innovative Center for New Drug Development of Immune Inflammatory Diseases (Ministry of Education), Minhang Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Department of Pharmaceutics, School of Pharmacy, Fudan University, Shanghai 201203, China
| |
Collapse
|
20
|
Zhou M, Lin Y, Chen H, Zhao M, Zeng Y, Hu X, Tang P, Fu Y, Wei L, Han L. Brain-tumor-seeking and serpin-inhibiting outer membrane vesicles restore plasmin-mediated attacks against brain metastases. J Control Release 2024; 375:116-126. [PMID: 39236899 DOI: 10.1016/j.jconrel.2024.09.003] [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: 04/30/2024] [Revised: 08/19/2024] [Accepted: 09/02/2024] [Indexed: 09/07/2024]
Abstract
Many chemotherapeutic and molecular targeted drugs have been used to treat brain metastases, e.g., anti-angiogenic vandetanib. However, the blood-brain barrier and brain-specific resistance mechanisms make these systemic therapeutic approaches inefficacious. Brain metastatic cancer cells could mimic neurons to upregulate multiple serpins and secrete them into the extracellular environment to reduce local plasmin production to promote L1CAM-mediated vessel co-option and resist anti-angiogenesis therapy. Here, we developed brain-tumor-seeking and serpin-inhibiting outer membrane vesicles (DE@OMVs) to traverse across the blood-brain barrier, bypass neurons, and specially enter metastatic cancer cells via targeting GRP94 and vimentin. Through specific delivery of dexamethasone and embelin, reduced serpin secretion, restored plasmin production, significant L1CAM inactivation and tumor cell apoptosis were specially found in intracranial metastatic regions, leading to delayed tumor growth and prolonged survival in mice with brain metastases. By combining the brain-tumor-seeking properties with the regulation of the serpin/plasminogen activator/plasmin/L1CAM axis, this study provides a potent and highly-selective systemic therapeutic option for brain metastases.
Collapse
Affiliation(s)
- Mengyuan Zhou
- Jiangsu Key Laboratory of Neuropsychiatric Diseases Research, College of Pharmaceutical Sciences, Soochow University, Suzhou 215123, China; Jiangsu Key Laboratory of Infection and Immunity, Institutes of Biology and Medical Sciences, Soochow University, Suzhou 215123, China
| | - Yuanyuan Lin
- Jiangsu Key Laboratory of Neuropsychiatric Diseases Research, College of Pharmaceutical Sciences, Soochow University, Suzhou 215123, China; Jiangsu Key Laboratory of Infection and Immunity, Institutes of Biology and Medical Sciences, Soochow University, Suzhou 215123, China
| | - Haiyan Chen
- Jiangsu Key Laboratory of Neuropsychiatric Diseases Research, College of Pharmaceutical Sciences, Soochow University, Suzhou 215123, China
| | - Mei Zhao
- Jiangsu Key Laboratory of Neuropsychiatric Diseases Research, College of Pharmaceutical Sciences, Soochow University, Suzhou 215123, China
| | - Yuteng Zeng
- Jiangsu Key Laboratory of Neuropsychiatric Diseases Research, College of Pharmaceutical Sciences, Soochow University, Suzhou 215123, China
| | - Xiaoxiao Hu
- Jiangsu Key Laboratory of Neuropsychiatric Diseases Research, College of Pharmaceutical Sciences, Soochow University, Suzhou 215123, China
| | - Puxian Tang
- Jiangsu Key Laboratory of Neuropsychiatric Diseases Research, College of Pharmaceutical Sciences, Soochow University, Suzhou 215123, China
| | - Yuxuan Fu
- Jiangsu Key Laboratory of Infection and Immunity, Institutes of Biology and Medical Sciences, Soochow University, Suzhou 215123, China
| | - Lin Wei
- Department of Infectious Diseases, The First Affiliated Hospital of Anhui Medical University, Hefei 230032, Anhui, China; School of Life Sciences, Anhui Medical University, Hefei 230032, Anhui, China.
| | - Liang Han
- Jiangsu Key Laboratory of Neuropsychiatric Diseases Research, College of Pharmaceutical Sciences, Soochow University, Suzhou 215123, China; Jiangsu Province Engineering Research Center of Precision Diagnostics and Therapeutics Development, Soochow University, Suzhou 215123, China.
| |
Collapse
|
21
|
Mairinger S, Jackwerth M, Chalampalakis Z, Rausch I, Weber M, Wölfl-Duchek M, Pracher L, Nics L, Pahnke J, Langsteger W, Hacker M, Zeitlinger M, Langer O. First-in-human evaluation of 6-bromo-7-[ 11C]methylpurine, a PET tracer for assessing the function of multidrug resistance-associated proteins in different tissues. Eur J Nucl Med Mol Imaging 2024; 51:3900-3911. [PMID: 39060376 PMCID: PMC11527933 DOI: 10.1007/s00259-024-06851-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Accepted: 07/14/2024] [Indexed: 07/28/2024]
Abstract
PURPOSE Multidrug resistance-associated protein 1 (MRP1) is a transport protein with a widespread tissue distribution, which has been implicated in the pathophysiology of Alzheimer's and chronic respiratory disease. PET with 6-bromo-7-[11C]methylpurine ([11C]BMP) has been used to measure MRP1 function in rodents. In this study, [11C]BMP was for the first time characterised in humans to assess the function of MRP1 and other MRP subtypes in different tissues. METHODS Thirteen healthy volunteers (7 men, 6 women) underwent dynamic whole-body PET scans on a long axial field-of-view (LAFOV) PET/CT system after intravenous injection of [11C]BMP. Three subjects of each sex were scanned a second time to assess reproducibility. Volumes of interest were outlined for MRP-expressing tissues (cerebral cortex, cerebellum, choroid plexus, retina, lungs, myocardium, kidneys, and liver). From the time-activity curves, the elimination rate constant (kE, h- 1) was derived as a parameter for tissue MRP function and its test-retest variability (TRTV, %) was calculated. Radiation dosimetry was calculated using the Medical Internal Radiation Dose (MIRD) methodology. RESULTS Mean kE and corresponding TRTV values were: cerebral cortex: 0.055 ± 0.010 h- 1 (- 4 ± 24%), cerebellum: 0.033 ± 0.009 h- 1 (1 ± 39%), choroid plexus: 0.292 ± 0.059 h- 1 (0.1 ± 16%), retina: 0.234 ± 0.045 h- 1 (30 ± 38%), lungs: 0.875 ± 0.095 h- 1 (- 3 ± 11%), myocardium: 0.641 ± 0.105 h- 1 (11 ± 25%), kidneys: 1.378 ± 0.266 h- 1 (14 ± 16%), and liver: 0.685 ± 0.072 h- 1 (7 ± 9%). Significant sex differences were found for kE in the cerebellum, lungs and kidneys. Effective dose was 4.67 ± 0.18 µSv/MBq for men and 4.55 ± 0.18 µSv/MBq for women. CONCLUSION LAFOV PET/CT with [11C]BMP potentially allows for simultaneous assessment of MRP function in multiple human tissues. Mean TRTV of kE in different tissues was in an acceptable range, except for the retina. The radiation dosimetry of [11C]BMP was in the typical range of 11C-tracers. LAFOV PET/CT holds great potential to assess at a whole-body, multi-tissue level molecular targets relevant for drug disposition in humans. TRIAL REGISTRATION EudraCT 2021-006348-29. Registered 15 December 2021.
Collapse
Affiliation(s)
- Severin Mairinger
- Department of Clinical Pharmacology, Medical University of Vienna, Vienna, Austria
- Department of Biomedical Imaging and Image-guided Therapy, Medical University of Vienna, Vienna, Austria
| | - Matthias Jackwerth
- Department of Clinical Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Zacharias Chalampalakis
- QIMP Team, Center for Medical Physics and Biomedical Engineering, Medical University of Vienna, Vienna, Austria
| | - Ivo Rausch
- QIMP Team, Center for Medical Physics and Biomedical Engineering, Medical University of Vienna, Vienna, Austria
| | - Maria Weber
- Department of Clinical Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Michael Wölfl-Duchek
- Department of Clinical Pharmacology, Medical University of Vienna, Vienna, Austria
- Department of Biomedical Imaging and Image-guided Therapy, Medical University of Vienna, Vienna, Austria
| | - Lena Pracher
- Department of Clinical Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Lukas Nics
- Department of Biomedical Imaging and Image-guided Therapy, Medical University of Vienna, Vienna, Austria
| | - Jens Pahnke
- Translational Neurodegeneration Research and Neuropathology Lab, Department of Clinical Medicine (KlinMed), Medical Faculty, University of Oslo, Oslo, Norway
- Section of Neuropathology Research, Department of Pathology, Clinics for Laboratory Medicine (KLM), Oslo University Hospital, Oslo, Norway
- Institute of Nutritional Medicine (INUM) and Lübeck Institute of Dermatology (LIED), University of Lübeck and University Medical Center Schleswig-Holstein, Lübeck, Germany
- Department of Pharmacology, Faculty of Medicine and Life Sciences, University of Latvia, Rīga, Latvia
- School of Neurobiology, Biochemistry and Biophysics, The Georg S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Werner Langsteger
- Department of Biomedical Imaging and Image-guided Therapy, Medical University of Vienna, Vienna, Austria
| | - Marcus Hacker
- Department of Biomedical Imaging and Image-guided Therapy, Medical University of Vienna, Vienna, Austria
| | - Markus Zeitlinger
- Department of Clinical Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Oliver Langer
- Department of Clinical Pharmacology, Medical University of Vienna, Vienna, Austria.
- Department of Biomedical Imaging and Image-guided Therapy, Medical University of Vienna, Vienna, Austria.
| |
Collapse
|
22
|
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
|
23
|
Kanjanasirirat P, Saengsawang W, Ketsawatsomkron P, Asavapanumas N, Borwornpinyo S, Soodvilai S, Hongeng S, Charoensutthivarakul S. GDNF and cAMP significantly enhance in vitro blood-brain barrier integrity in a humanized tricellular transwell model. Heliyon 2024; 10:e39343. [PMID: 39492921 PMCID: PMC11530796 DOI: 10.1016/j.heliyon.2024.e39343] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Revised: 10/11/2024] [Accepted: 10/11/2024] [Indexed: 11/05/2024] Open
Abstract
Blood-brain barrier (BBB) is a crucial membrane safeguarding neural tissue by controlling the molecular exchange between blood and the brain. However, assessing BBB permeability presents challenges for central nervous system (CNS) drug development. In vitro studies of BBB-permeable agents before animal testing are essential to mitigate failures. Improved in vitro models are needed to mimic physiologically relevant BBB integrity. Here, we established an in vitro human-derived triculture BBB model, coculturing hCMEC/D3 with primary astrocytes and pericytes in a transwell format. This study found that the triculture BBB model exhibited significantly higher paracellular tightness (TEER 147.6 ± 6.5 Ω × cm2) than its monoculture counterpart (106.3 ± 1.0 Ω × cm2). Additionally, BBB permeability in the triculture model was significantly lower. While GDNF and cAMP have been shown to promote BBB integrity in monoculture models, their effect in our model was previously unreported. Our study demonstrates that both GDNF and cAMP increased TEER values (around 200 Ω × cm2 for each; 237.6 ± 17.7 Ω × cm2 for co-treatment) compared to untreated control, and decreased BBB permeability, mediated by increased claudin-5 expression. In summary, this humanized triculture BBB model, enhanced by GDNF and cAMP, offers an alternative for exploring in vitro drug penetration into the human brain.
Collapse
Affiliation(s)
- Phongthon Kanjanasirirat
- School of Bioinnovation and Bio-Based Product Intelligence, Faculty of Science, Mahidol University, Bangkok, 10400, Thailand
- Excellent Center for Drug Discovery (ECDD), Faculty of Science, Mahidol University, Bangkok, 10400, Thailand
- Department of Pathobiology, Faculty of Science, Mahidol University, Bangkok, 10400, Thailand
| | - Witchuda Saengsawang
- Department of Basic Biomedical Sciences, Dr. William M. Scholl College of Podiatric Medicine, Rosalind Franklin University of Medicine and Science, North Chicago, IL, 60064, USA
| | - Pimonrat Ketsawatsomkron
- Chakri Naruebodindra Medical Institute, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Samut Prakarn, 10540, Thailand
| | - Nithi Asavapanumas
- Chakri Naruebodindra Medical Institute, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Samut Prakarn, 10540, Thailand
| | - Suparerk Borwornpinyo
- Excellent Center for Drug Discovery (ECDD), Faculty of Science, Mahidol University, Bangkok, 10400, Thailand
- Department of Biotechnology, Faculty of Science, Mahidol University, Bangkok, 10400, Thailand
| | - Sunhapas Soodvilai
- Excellent Center for Drug Discovery (ECDD), Faculty of Science, Mahidol University, Bangkok, 10400, Thailand
- Department of Physiology, Faculty of Science, Mahidol University, Bangkok, 10400, Thailand
| | - Suradej Hongeng
- Excellent Center for Drug Discovery (ECDD), Faculty of Science, Mahidol University, Bangkok, 10400, Thailand
- Department of Pediatrics, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok, 10400, Thailand
| | - Sitthivut Charoensutthivarakul
- School of Bioinnovation and Bio-Based Product Intelligence, Faculty of Science, Mahidol University, Bangkok, 10400, Thailand
- Excellent Center for Drug Discovery (ECDD), Faculty of Science, Mahidol University, Bangkok, 10400, Thailand
- Center for Neuroscience, Faculty of Science, Mahidol University, Bangkok, 10400, Thailand
| |
Collapse
|
24
|
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
|
25
|
Lubitz LJ, Haffner MP, Rieger H, Leneweit G. Elevated Cellular Uptake of Succinimide- and Glucose-Modified Liposomes for Blood-Brain Barrier Transfer and Glioblastoma Therapy. Biomedicines 2024; 12:2135. [PMID: 39335648 PMCID: PMC11430759 DOI: 10.3390/biomedicines12092135] [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: 08/20/2024] [Revised: 09/03/2024] [Accepted: 09/06/2024] [Indexed: 09/30/2024] Open
Abstract
The uptake of four liposomal formulations was tested with the murine endothelial cell line bEnd.3 and the human glioblastoma cell line U-87 MG. All formulations were composed of DPPC, cholesterol, 5 mol% of mPEG (2000 Da, conjugated to DSPE), and the dye DiD. Three of the formulations had an additional PEG chain (nominally 5000 Da, conjugated to DSPE) with either succinimide (NHS), glucose (PEG-bound at C-6), or 4-aminophenyl β-D-glucopyranoside (bound at C-1) as ligands at the distal end. Measuring the uptake kinetics at 1 h and 3 h for liposomal incubation concentrations of 100 µM, 500 µM, and 1000 µM, we calculated the liposomal uptake saturation S and the saturation half-time t1/2. We show that only succinimide has an elevated uptake in bEnd.3 cells, which makes it a very promising and so far largely unexplored candidate for BBB transfer and brain cancer therapies. Half-times are uniform at low concentrations but diversify for high concentrations for bEnd.3 cells. Contrary, U-87 MG cells show almost identical saturations for all three ligands, making a uniform uptake mechanism likely. Only mPEG liposomes stay at 60% of the saturation for ligand-coated liposomes. Half-times are diverse at low concentrations but unify at high concentrations for U-87 MG cells.
Collapse
Affiliation(s)
- Larissa J Lubitz
- ABNOBA GmbH, 75223 Niefern-Öschelbronn, Germany
- Carl Gustav Carus-Institute,75223 Niefern-Öschelbronn, Germany
- Department of Chemical and Process Engineering, Institute of Mechanical Process Engineering and Mechanics, Karlsruhe Institute of Technology, 76131 Karlsruhe, Germany
| | | | - Harden Rieger
- ABNOBA GmbH, 75223 Niefern-Öschelbronn, Germany
- Carl Gustav Carus-Institute,75223 Niefern-Öschelbronn, Germany
| | - Gero Leneweit
- ABNOBA GmbH, 75223 Niefern-Öschelbronn, Germany
- Carl Gustav Carus-Institute,75223 Niefern-Öschelbronn, Germany
- Department of Chemical and Process Engineering, Institute of Mechanical Process Engineering and Mechanics, Karlsruhe Institute of Technology, 76131 Karlsruhe, Germany
| |
Collapse
|
26
|
Netzahualcoyotzi C, Santillán-Cigales JJ, Adalid-Peralta LV, Velasco I. Infiltration of immune cells to the brain and its relation to the pathogenesis of Alzheimer's and Parkinson's diseases. J Neurochem 2024; 168:2316-2334. [PMID: 38549444 DOI: 10.1111/jnc.16106] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2023] [Revised: 02/22/2024] [Accepted: 03/13/2024] [Indexed: 10/04/2024]
Abstract
The neurovascular unit, composed of vascular endothelium, vascular smooth muscle, extracellular matrix components, pericytes, astrocytes, microglia, and neurons, allows the highly regulated exchange of molecules and the limited trafficking of cells to the brain through coordinated signaling activity. The passage of peripheral immune cells to the brain parenchyma is observed when there is clear damage to the barriers of this neurovascular unit, as occurs in traumatic brain injury. The possibility of leukocyte infiltration to the brain in neurodegenerative conditions has been proposed. In this review, we focus on describing the evidence for peripheral immune cell infiltration to the brain in the two most frequent neurodegenerative diseases: Alzheimer's and Parkinson's diseases. In particular, we address the mechanisms that promote the passage of these cells into the brain under such pathological conditions. We also discuss the relevance of the resulting cellular interactions, which provide evidence that the presence of peripheral immune cells in the brain is a key point in these neurodegenerative diseases.
Collapse
Affiliation(s)
- Citlalli Netzahualcoyotzi
- Instituto de Fisiología Celular-Neurociencias, Universidad Nacional Autónoma de México, Ciudad de México, Mexico
- Laboratorio de Reprogramación Celular, Instituto Nacional de Neurología y Neurocirugía Manuel Velasco Suárez, Ciudad de México, Mexico
| | - Juan Jair Santillán-Cigales
- Instituto de Fisiología Celular-Neurociencias, Universidad Nacional Autónoma de México, Ciudad de México, Mexico
| | - Laura Virginia Adalid-Peralta
- Laboratorio de Reprogramación Celular, Instituto Nacional de Neurología y Neurocirugía Manuel Velasco Suárez, Ciudad de México, Mexico
| | - Iván Velasco
- Instituto de Fisiología Celular-Neurociencias, Universidad Nacional Autónoma de México, Ciudad de México, Mexico
- Laboratorio de Reprogramación Celular, Instituto Nacional de Neurología y Neurocirugía Manuel Velasco Suárez, Ciudad de México, Mexico
| |
Collapse
|
27
|
Hu J, Li Y, Dong C, Wei H, Liao K, Wei J, Zhao C, Chaudhary A, Chen J, Xu H, Zhong K, Liang SH, Wang L, Ye W. Discovery and evaluation of a novel 18F-labeled vasopressin 1a receptor PET ligand with peripheral binding specificity. Acta Pharm Sin B 2024; 14:4014-4027. [PMID: 39309503 PMCID: PMC11413668 DOI: 10.1016/j.apsb.2024.05.033] [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: 01/19/2024] [Revised: 05/06/2024] [Accepted: 05/13/2024] [Indexed: 09/25/2024] Open
Abstract
The arginine-vasopressin (AVP) hormone plays a pivotal role in regulating various physiological processes, such as hormone secretion, cardiovascular modulation, and social behavior. Recent studies have highlighted the V1a receptor as a promising therapeutic target. In-depth insights into V1a receptor-related pathologies, attained through in vivo imaging and quantification in both peripheral organs and the central nervous system (CNS), could significantly advance the development of effective V1a inhibitors. To address this need, we develop a novel V1a-targeted positron emission tomography (PET) ligand, [18F]V1A-2303 ([18F]8), which demonstrates favorable in vitro binding affinity and selectivity for the V1a receptor. Specific tracer binding in peripheral tissues was also confirmed through rigorous cell uptake studies, autoradiography, biodistribution assessments. Furthermore, [18F]8 was employed in PET imaging and arterial blood sampling studies in healthy rhesus monkeys to assess its brain permeability and specificity, whole-body distribution, and kinetic properties. Our research indicated [18F]8 as a valuable tool for noninvasively studying V1a receptors in peripheral organs, and as a foundational element for the development of next-generation, brain-penetrant ligands specifically designed for the CNS.
Collapse
Affiliation(s)
- Junqi Hu
- Center of Cyclotron and PET Radiopharmaceuticals, Department of Nuclear Medicine & Key Laboratory of Basic and Translational Research on Radiopharmaceuticals, the First Affiliated Hospital of Jinan University, Guangzhou 510630, China
| | - Yinlong Li
- Department of Radiology and Imaging Sciences, Emory University, Atlanta, GA 30322, USA
| | - Chenchen Dong
- Center of Cyclotron and PET Radiopharmaceuticals, Department of Nuclear Medicine & Key Laboratory of Basic and Translational Research on Radiopharmaceuticals, the First Affiliated Hospital of Jinan University, Guangzhou 510630, China
| | - Huiyi Wei
- Center of Cyclotron and PET Radiopharmaceuticals, Department of Nuclear Medicine & Key Laboratory of Basic and Translational Research on Radiopharmaceuticals, the First Affiliated Hospital of Jinan University, Guangzhou 510630, China
| | - Kai Liao
- Center of Cyclotron and PET Radiopharmaceuticals, Department of Nuclear Medicine & Key Laboratory of Basic and Translational Research on Radiopharmaceuticals, the First Affiliated Hospital of Jinan University, Guangzhou 510630, China
| | - Junjie Wei
- Center of Cyclotron and PET Radiopharmaceuticals, Department of Nuclear Medicine & Key Laboratory of Basic and Translational Research on Radiopharmaceuticals, the First Affiliated Hospital of Jinan University, Guangzhou 510630, China
| | - Chunyu Zhao
- Department of Radiology and Imaging Sciences, Emory University, Atlanta, GA 30322, USA
| | - Ahmad Chaudhary
- Department of Radiology and Imaging Sciences, Emory University, Atlanta, GA 30322, USA
| | - Jiahui Chen
- Department of Radiology and Imaging Sciences, Emory University, Atlanta, GA 30322, USA
| | - Hao Xu
- Center of Cyclotron and PET Radiopharmaceuticals, Department of Nuclear Medicine & Key Laboratory of Basic and Translational Research on Radiopharmaceuticals, the First Affiliated Hospital of Jinan University, Guangzhou 510630, China
| | - Ke Zhong
- Department of Pharmacy, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China
| | - Steven H. Liang
- Department of Radiology and Imaging Sciences, Emory University, Atlanta, GA 30322, USA
| | - Lu Wang
- Center of Cyclotron and PET Radiopharmaceuticals, Department of Nuclear Medicine & Key Laboratory of Basic and Translational Research on Radiopharmaceuticals, the First Affiliated Hospital of Jinan University, Guangzhou 510630, China
| | - Weijian Ye
- Center of Cyclotron and PET Radiopharmaceuticals, Department of Nuclear Medicine & Key Laboratory of Basic and Translational Research on Radiopharmaceuticals, the First Affiliated Hospital of Jinan University, Guangzhou 510630, China
| |
Collapse
|
28
|
Shan Y, Cen Y, Xu X, Li P, Chen J, Nie Z, Zhang J. The ATP-Binding Cassette Transporter-Mediated Efflux Transport of Ganciclovir at the Blood-Brain Barrier. Eur J Drug Metab Pharmacokinet 2024; 49:609-617. [PMID: 38963639 DOI: 10.1007/s13318-024-00908-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/16/2024] [Indexed: 07/05/2024]
Abstract
BACKGROUND AND OBJECTIVE Recent studies have highlighted the key role of the ATP-binding cassette (ABC) transporters, including the P-glycoprotein (P-gp), the breast cancer resistance protein (BCRP), and the multi-drug resistance protein 4 (MRP4) in limiting the brain distribution of several antiviral agents. In this study, we investigated whether the inhibition of these transporters increases the permeability of the blood-brain barrier (BBB) to ganciclovir. METHODS A microdialysis and high-performance liquid chromatographic method was developed to monitor the concentrations of unbound ganciclovir in the brain interstitial fluid and plasma, with and without the administration of ABC transporter inhibitors. Pharmacokinetic parameters, including the area under the plasma concentration-time curve from time 0 to time of the last measurable analyte concentration (AUC0-t,plasma), the area under the brain interstitial fluid concentration-time curve from time 0 to time of the last measurable analyte concentration (AUC0-t,brain), and the unbound brain-to-plasma concentration ratio (Kp,uu,brain) were calculated. RESULTS The mean AUC0-t,plasma, AUC0-t,brain, and Kp,uu,brain in rats who received ganciclovir (30 mg/kg, intraperitoneal) alone were 1090 min·µg/mL, 150 min·µg/mL, and 14%, respectively. After the administration of tariquidar (inhibitor of P-gp), Ko143 (inhibitor of BCRP), or MK-571 (inhibitor of MRP4), the Kp,uu,brain of ganciclovir increased to 31 ± 2.1%, 26 ± 1.3%, and 32 ± 2.0%, respectively. CONCLUSIONS The findings of this study suggest that ABC transporters P-gp, BCRP, and MRP4 mediate the efflux of ganciclovir at the BBB and that the inhibition of these transporters facilitates the penetration of the BBB by ganciclovir.
Collapse
MESH Headings
- Ganciclovir/pharmacokinetics
- Animals
- Blood-Brain Barrier/metabolism
- Rats
- Male
- Antiviral Agents/pharmacokinetics
- Antiviral Agents/pharmacology
- ATP-Binding Cassette Transporters/metabolism
- ATP-Binding Cassette Transporters/antagonists & inhibitors
- Rats, Sprague-Dawley
- Biological Transport
- Microdialysis/methods
- Chromatography, High Pressure Liquid/methods
- Brain/metabolism
- ATP Binding Cassette Transporter, Subfamily G, Member 2/metabolism
- ATP Binding Cassette Transporter, Subfamily G, Member 2/antagonists & inhibitors
- ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism
- ATP Binding Cassette Transporter, Subfamily B, Member 1/antagonists & inhibitors
- Diketopiperazines/pharmacokinetics
Collapse
Affiliation(s)
- Yuheng Shan
- Department of Neurology, Characteristic Medical Centre of People's Armed Police Force, Tianjin, 300162, People's Republic of China
| | - Yuying Cen
- Department of Neurology, Beijing Anzhen Hospital, Capital Medical University, Beijing, 100011, People's Republic of China
| | - Xiaojiao Xu
- Department of Neurology, The First Medical Centre, Chinese PLA General Hospital, No. 28, Fuxing Road, Beijing, 100853, People's Republic of China
| | - Ping Li
- Department of Neurology, Characteristic Medical Centre of People's Armed Police Force, Tianjin, 300162, People's Republic of China
| | - Jing Chen
- Department of Neurology, Characteristic Medical Centre of People's Armed Police Force, Tianjin, 300162, People's Republic of China
| | - Zhiyong Nie
- Beijing Institute of Pharmacology and Toxicology, Beijing, 100850, People's Republic of China.
| | - Jiatang Zhang
- Department of Neurology, The First Medical Centre, Chinese PLA General Hospital, No. 28, Fuxing Road, Beijing, 100853, People's Republic of China.
| |
Collapse
|
29
|
Loiola RA, Hachani J, Duban-Deweer S, Sevin E, Bugno P, Kowalska A, Rizzi E, Shimizu F, Kanda T, Mysiorek C, Mazurek M, Gosselet F. Secretome of brain microvascular endothelial cells promotes endothelial barrier tightness and protects against hypoxia-induced vascular leakage. Mol Med 2024; 30:132. [PMID: 39187765 PMCID: PMC11348522 DOI: 10.1186/s10020-024-00897-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Accepted: 08/12/2024] [Indexed: 08/28/2024] Open
Abstract
Cell-based therapeutic strategies have been proposed as an alternative for brain and blood vessels repair after stroke, but their clinical application is hampered by potential adverse effects. We therefore tested the hypothesis that secretome of these cells might be used instead to still focus on cell-based therapeutic strategies. We therefore characterized the composition and the effect of the secretome of brain microvascular endothelial cells (BMECs) on primary in vitro human models of angiogenesis and vascular barrier. Two different secretome batches produced in high scale (scHSP) were analysed by mass spectrometry. Human primary CD34+-derived endothelial cells (CD34+-ECs) were used as well as in vitro models of EC monolayer (CMECs) and blood-brain barrier (BBB). Cells were also exposed to oxygen-glucose deprivation (OGD) conditions and treated with scHSP during reoxygenation. Protein yield and composition of scHSP batches showed good reproducibility. scHSP increased CD34+-EC proliferation, tubulogenesis, and migration. Proteomic analysis of scHSP revealed the presence of growth factors and proteins modulating cell metabolism and inflammatory pathways. scHSP improved the integrity of CMECs, and upregulated the expression of junctional proteins. Such effects were mediated through the activation of the interferon pathway and downregulation of Wnt signalling. Furthermore, OGD altered the permeability of both CMECs and BBB, while scHSP prevented the OGD-induced vascular leakage in both models. These effects were mediated through upregulation of junctional proteins and regulation of MAPK/VEGFR2. Finally, our results highlight the possibility of using secretome from BMECs as a therapeutic alternative to promote brain angiogenesis and to protect from ischemia-induced vascular leakage.
Collapse
Affiliation(s)
- Rodrigo Azevedo Loiola
- UR 2465, Laboratory of the Blood-Brain Barrier (LBHE), Sciences Faculty Jean Perrin, Artois University, 62300, Lens, France
| | - Johan Hachani
- UR 2465, Laboratory of the Blood-Brain Barrier (LBHE), Sciences Faculty Jean Perrin, Artois University, 62300, Lens, France
| | - Sophie Duban-Deweer
- UR 2465, Laboratory of the Blood-Brain Barrier (LBHE), Sciences Faculty Jean Perrin, Artois University, 62300, Lens, France
| | - Emmanuel Sevin
- UR 2465, Laboratory of the Blood-Brain Barrier (LBHE), Sciences Faculty Jean Perrin, Artois University, 62300, Lens, France
| | - Paulina Bugno
- Pure Biologics S.A., Duńska 11, 54-427, Wroclaw, Poland
| | | | - Eleonora Rizzi
- UR 2465, Laboratory of the Blood-Brain Barrier (LBHE), Sciences Faculty Jean Perrin, Artois University, 62300, Lens, France
| | - Fumitaka Shimizu
- Department of Neurology and Clinical Neuroscience, Graduate School of Medicine, Yamaguchi University, Ube, Japan
| | - Takashi Kanda
- Department of Neurology and Clinical Neuroscience, Graduate School of Medicine, Yamaguchi University, Ube, Japan
| | - Caroline Mysiorek
- UR 2465, Laboratory of the Blood-Brain Barrier (LBHE), Sciences Faculty Jean Perrin, Artois University, 62300, Lens, France
| | | | - Fabien Gosselet
- UR 2465, Laboratory of the Blood-Brain Barrier (LBHE), Sciences Faculty Jean Perrin, Artois University, 62300, Lens, France.
| |
Collapse
|
30
|
Matsuo K, Nagamatsu J, Nagata K, Umeda R, Shiota T, Morimoto S, Suzuki N, Aoki M, Okano H, Nakamori M, Nishihara H. Establishment of a novel amyotrophic lateral sclerosis patient ( TARDBP N345K/+)-derived brain microvascular endothelial cell model reveals defective Wnt/β-catenin signaling: investigating diffusion barrier dysfunction and immune cell interaction. Front Cell Dev Biol 2024; 12:1357204. [PMID: 39211392 PMCID: PMC11357944 DOI: 10.3389/fcell.2024.1357204] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2023] [Accepted: 07/25/2024] [Indexed: 09/04/2024] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a major neurodegenerative disease for which there is currently no curative treatment. The blood-brain barrier (BBB), multiple physiological functions formed by mainly specialized brain microvascular endothelial cells (BMECs), serves as a gatekeeper to protect the central nervous system (CNS) from harmful molecules in the blood and aberrant immune cell infiltration. The accumulation of evidence indicating that alterations in the peripheral milieu can contribute to neurodegeneration within the CNS suggests that the BBB may be a previously overlooked factor in the pathogenesis of ALS. Animal models suggest BBB breakdown may precede neurodegeneration and link BBB alteration to the disease progression or even onset. However, the lack of a useful patient-derived model hampers understanding the pathomechanisms of BBB dysfunction and the development of BBB-targeted therapies. In this study, we differentiated BMEC-like cells from human induced pluripotent stem cells (hiPSCs) derived from ALS patients to investigate BMEC functions in ALS patients. TARDBP N345K/+ carrying patient-derived BMEC-like cells exhibited increased permeability to small molecules due to loss of tight junction in the absence of neurodegeneration or neuroinflammation, highlighting that BMEC abnormalities in ALS are not merely secondary consequences of disease progression. Furthermore, they exhibited increased expression of cell surface adhesion molecules like ICAM-1 and VCAM-1, leading to enhanced immune cell adhesion. BMEC-like cells derived from hiPSCs with other types of TARDBP gene mutations (TARDBP K263E/K263E and TARDBP G295S/G295S) introduced by genome editing technology did not show such BMEC dysfunction compared to the isogenic control. Interestingly, transactive response DNA-binding protein 43 (TDP-43) was mislocalized to cytoplasm in TARDBP N345K/+ carrying model. Wnt/β-catenin signaling was downregulated in the ALS patient (TARDBP N345K/+)-derived BMEC-like cells and its activation rescued the leaky barrier phenotype and settled down VCAM-1 expressions. These results indicate that TARDBP N345K/+ carrying model recapitulated BMEC abnormalities reported in brain samples of ALS patients. This novel patient-derived BMEC-like cell is useful for the further analysis of the involvement of vascular barrier dysfunctions in the pathogenesis of ALS and for promoting therapeutic drug discovery targeting BMEC.
Collapse
Affiliation(s)
- Kinya Matsuo
- Department of Neurology and Clinical Neuroscience, Yamaguchi University Graduate School of Medicine, Yamaguchi, Japan
| | - Jun Nagamatsu
- Yamaguchi University Graduate School of Medicine, Yamaguchi, Japan
| | - Kazuhiro Nagata
- Yamaguchi University Graduate School of Medicine, Yamaguchi, Japan
| | - Ryusei Umeda
- Yamaguchi University Graduate School of Medicine, Yamaguchi, Japan
| | - Takaya Shiota
- Yamaguchi University Graduate School of Medicine, Yamaguchi, Japan
| | - Satoru Morimoto
- Keio University, Regenerative Medicine Research Center, Kawasaki, Kanagawa, Japan
| | - Naoki Suzuki
- Department of Neurology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Masashi Aoki
- Department of Neurology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Hideyuki Okano
- Keio University, Regenerative Medicine Research Center, Kawasaki, Kanagawa, Japan
| | - Masayuki Nakamori
- Department of Neurology and Clinical Neuroscience, Yamaguchi University Graduate School of Medicine, Yamaguchi, Japan
| | - Hideaki Nishihara
- Department of Neurology and Clinical Neuroscience, Yamaguchi University Graduate School of Medicine, Yamaguchi, Japan
| |
Collapse
|
31
|
Han J, Wang Y, Wei P, Lu D, Shan Y. Unveiling the hidden connection: the blood-brain barrier's role in epilepsy. Front Neurol 2024; 15:1413023. [PMID: 39206290 PMCID: PMC11349696 DOI: 10.3389/fneur.2024.1413023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2024] [Accepted: 07/18/2024] [Indexed: 09/04/2024] Open
Abstract
Epilepsy is characterized by abnormal synchronous electrical activity of neurons in the brain. The blood-brain barrier, which is mainly composed of endothelial cells, pericytes, astrocytes and other cell types and is formed by connections between a variety of cells, is the key physiological structure connecting the blood and brain tissue and is critical for maintaining the microenvironment in the brain. Physiologically, the blood-brain barrier controls the microenvironment in the brain mainly by regulating the passage of various substances. Disruption of the blood-brain barrier and increased leakage of specific substances, which ultimately leading to weakened cell junctions and abnormal regulation of ion concentrations, have been observed during the development and progression of epilepsy in both clinical studies and animal models. In addition, disruption of the blood-brain barrier increases drug resistance through interference with drug trafficking mechanisms. The changes in the blood-brain barrier in epilepsy mainly affect molecular pathways associated with angiogenesis, inflammation, and oxidative stress. Further research on biomarkers is a promising direction for the development of new therapeutic strategies.
Collapse
Affiliation(s)
| | | | | | | | - Yongzhi Shan
- Department of Neurosurgery, Xuanwu Hospital Capital Medical University, Beijing, China
| |
Collapse
|
32
|
Lentzas A, de Gooijer MC, Zuidema S, Meurs A, Çitirikkaya CH, Venekamp N, Beijnen JH, van Tellingen O. ATP-binding cassette transporter inhibitor potency and substrate drug affinity are critical determinants of successful drug delivery enhancement to the brain. Fluids Barriers CNS 2024; 21:62. [PMID: 39103921 DOI: 10.1186/s12987-024-00562-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Accepted: 07/28/2024] [Indexed: 08/07/2024] Open
Abstract
BACKGROUND Pharmacotherapy for brain diseases is severely compromised by the blood-brain barrier (BBB). ABCB1 and ABCG2 are drug transporters that restrict drug entry into the brain and their inhibition can be used as a strategy to boost drug delivery and pharmacotherapy for brain diseases. METHODS We employed elacridar and tariquidar in mice to explore the conditions for effective inhibition at the BBB. Abcg2;Abcb1a/b knockout (KO), Abcb1a/b KO, Abcg2 KO and wild-type (WT) mice received a 3 h i.p. infusion of a cocktail of 8 typical substrate drugs in combination with elacridar or tariquidar at a range of doses. Abcg2;Abcb1a/b KO mice were used as the reference for complete inhibition, while single KO mice were used to assess the potency to inhibit the remaining transporter. Brain and plasma drug levels were measured by LC-MS/MS. RESULTS Complete inhibition of ABCB1 at the BBB is achieved when the elacridar plasma level reaches 1200 nM, whereas tariquidar requires at least 4000 nM. Inhibition of ABCG2 is more difficult. Elacridar inhibits ABCG2-mediated efflux of weak but not strong ABCG2 substrates. Strikingly, tariquidar does not enhance the brain uptake of any ABCG2-subtrate drug. Similarly, elacridar, but not tariquidar, was able to inhibit its own brain efflux in ABCG2-proficient mice. The plasma protein binding of elacridar and tariquidar was very high but similar in mouse and human plasma, facilitating the translation of mouse data to humans. CONCLUSIONS This work shows that elacridar is an effective pharmacokinetic-enhancer for the brain delivery of ABCB1 and weaker ABCG2 substrate drugs when a plasma concentration of 1200 nM is exceeded.
Collapse
Affiliation(s)
- Aristeidis Lentzas
- Division of Pharmacology, The Netherlands Cancer Institute, Room H3.010, Plesmanlaan 121, Amsterdam, 1066 CX, The Netherlands
| | - Mark C de Gooijer
- Division of Pharmacology, The Netherlands Cancer Institute, Room H3.010, Plesmanlaan 121, Amsterdam, 1066 CX, The Netherlands
- Faculty of Biology, Medicine and Health, University of Manchester, Manchester, M1 3WE, UK
- The Christie NHS Foundation Trust, Manchester, M20 4BX, UK
| | - Stefanie Zuidema
- Division of Pharmacology, The Netherlands Cancer Institute, Room H3.010, Plesmanlaan 121, Amsterdam, 1066 CX, The Netherlands
| | - Amber Meurs
- Division of Pharmacology, The Netherlands Cancer Institute, Room H3.010, Plesmanlaan 121, Amsterdam, 1066 CX, The Netherlands
| | - Ceren H Çitirikkaya
- Division of Pharmacology, The Netherlands Cancer Institute, Room H3.010, Plesmanlaan 121, Amsterdam, 1066 CX, The Netherlands
| | - Nikkie Venekamp
- Division of Pharmacology, The Netherlands Cancer Institute, Room H3.010, Plesmanlaan 121, Amsterdam, 1066 CX, The Netherlands
- Department of Pharmacy, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Jos H Beijnen
- Department of Pharmacy, The Netherlands Cancer Institute, Amsterdam, The Netherlands
- Division of Pharmacoepidemiology and Clinical Pharmacology, Department of Pharmaceutical Sciences, Faculty of Science, Utrecht University, Universiteitsweg 99, Utrecht, 3584 CG, The Netherlands
| | - Olaf van Tellingen
- Division of Pharmacology, The Netherlands Cancer Institute, Room H3.010, Plesmanlaan 121, Amsterdam, 1066 CX, The Netherlands.
- Mouse Cancer Clinic, The Netherlands Cancer Institute, Amsterdam, The Netherlands.
| |
Collapse
|
33
|
Zhou X, Azimi M, Handin N, Riselli A, Vora B, Chun E, Yee SW, Artursson P, Giacomini KM. Proteomic Profiling Reveals Age-Related Changes in Transporter Proteins in the Human Blood-Brain Barrier. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.26.604313. [PMID: 39091855 PMCID: PMC11291171 DOI: 10.1101/2024.07.26.604313] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 08/04/2024]
Abstract
The Blood-Brain Barrier (BBB) is a crucial, selective barrier that regulates the entry of molecules including nutrients, environmental toxins, and therapeutic medications into the brain. This function relies heavily on brain endothelial cell proteins, particularly transporters and tight junction proteins. The BBB continues to develop postnatally, adapting its selective barrier function across different developmental phases, and alters with aging and disease. Here we present a global proteomics analysis focused on the ontogeny and aging of proteins in human brain microvessels (BMVs), predominantly composed of brain endothelial cells. Our proteomic profiling quantified 6,223 proteins and revealed possible age-related alteration in BBB permeability due to basement membrane component changes through the early developmental stage and age-dependent changes in transporter expression. Notable changes in expression levels were observed with development and age in nutrient transporters and transporters that play critical roles in drug disposition. This research 1) provides important information on the mechanisms that drive changes in the metabolic content of the brain with age and 2) enables the creation of physiologically based pharmacokinetic models for CNS drug distribution across different life stages.
Collapse
Affiliation(s)
- Xujia Zhou
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, California, United States
| | - Mina Azimi
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, California, United States
| | - Niklas Handin
- Department of Pharmacy, Uppsala University, Uppsala, Sweden
| | - Andrew Riselli
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, California, United States
| | - Bianca Vora
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, California, United States
| | - Eden Chun
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, California, United States
| | - Sook Wah Yee
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, California, United States
| | - Per Artursson
- Department of Pharmacy, Uppsala University, Uppsala, Sweden
| | - Kathleen M Giacomini
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, California, United States
| |
Collapse
|
34
|
Chung KJ, Abdelhafez YG, Spencer BA, Jones T, Tran Q, Nardo L, Chen MS, Sarkar S, Medici V, Lyo V, Badawi RD, Cherry SR, Wang G. Quantitative PET imaging and modeling of molecular blood-brain barrier permeability. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.07.26.24311027. [PMID: 39108503 PMCID: PMC11302722 DOI: 10.1101/2024.07.26.24311027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 08/12/2024]
Abstract
Blood-brain barrier (BBB) disruption is involved in the pathogenesis and progression of many neurological and systemic diseases. Non-invasive assessment of BBB permeability in humans has mainly been performed with dynamic contrast-enhanced magnetic resonance imaging, evaluating the BBB as a structural barrier. Here, we developed a novel non-invasive positron emission tomography (PET) method in humans to measure the BBB permeability of molecular radiotracers that cross the BBB through different transport mechanisms. Our method uses high-temporal resolution dynamic imaging and kinetic modeling to jointly estimate cerebral blood flow and tracer-specific BBB transport rate from a single dynamic PET scan and measure the molecular permeability-surface area (PS) product of the radiotracer. We show our method can resolve BBB PS across three PET radiotracers with greatly differing permeabilities, measure reductions in BBB PS of 18F-fluorodeoxyglucose (FDG) in healthy aging, and demonstrate a possible brain-body association between decreased FDG BBB PS in patients with metabolic dysfunction-associated steatotic liver inflammation. Our method opens new directions to efficiently study the molecular permeability of the human BBB in vivo using the large catalogue of available molecular PET tracers.
Collapse
Affiliation(s)
- Kevin J Chung
- Department of Radiology, University of California Davis Health, Sacramento, CA
| | - Yasser G Abdelhafez
- Department of Radiology, University of California Davis Health, Sacramento, CA
| | - Benjamin A Spencer
- Department of Radiology, University of California Davis Health, Sacramento, CA
| | - Terry Jones
- Department of Radiology, University of California Davis Health, Sacramento, CA
| | - Quyen Tran
- Department of Radiology, University of California Davis Health, Sacramento, CA
| | - Lorenzo Nardo
- Department of Radiology, University of California Davis Health, Sacramento, CA
| | - Moon S Chen
- Department of Internal Medicine, University of California Davis Health, Sacramento, CA
| | - Souvik Sarkar
- Department of Internal Medicine, University of California Davis Health, Sacramento, CA
| | - Valentina Medici
- Department of Internal Medicine, University of California Davis Health, Sacramento, CA
- Division of Gastroenterology and Hepatology, University of California Davis Health, Sacramento, CA
| | - Victoria Lyo
- Department of Surgery, University of California Davis Health, Sacramento, CA
- Center for Alimentary and Metabolic Sciences, University of California Davis Health, Sacramento, CA
| | - Ramsey D Badawi
- Department of Radiology, University of California Davis Health, Sacramento, CA
- Department of Biomedical Engineering, University of California at Davis, Davis, CA
| | - Simon R Cherry
- Department of Biomedical Engineering, University of California at Davis, Davis, CA
- Department of Radiology, University of California Davis Health, Sacramento, CA
| | - Guobao Wang
- Department of Radiology, University of California Davis Health, Sacramento, CA
| |
Collapse
|
35
|
F Martins ML, Heydari P, Li W, Martínez-Chávez A, El Yattouti M, Lebre MC, Beijnen JH, Schinkel AH. The role of drug efflux and uptake transporters in the plasma pharmacokinetics and tissue disposition of morphine and its main metabolites. Toxicol Appl Pharmacol 2024; 490:117040. [PMID: 39032800 DOI: 10.1016/j.taap.2024.117040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Revised: 07/04/2024] [Accepted: 07/15/2024] [Indexed: 07/23/2024]
Abstract
Morphine is a widely used opioid for the treatment of pain. Differences in drug transporter expression and activity may contribute to variability in morphine pharmacokinetics and response. Using appropriate mouse models, we investigated the impact of the efflux transporters ABCB1 and ABCG2 and the OATP uptake transporters on the pharmacokinetics of morphine, morphine-3-glucuronide (M3G), and M6G. Upon subcutaneous administration of morphine, its plasma exposure in Abcb1a/1b-/-;Abcg2-/--, Abcb1a/1b-/-;Abcg2-/-;Oatp1a/1b-/-;Oatp2b1-/- (Bab12), and Oatp1a/1b-/-;Oatp2b1-/- mice was similar to that found in wild-type mice. Forty minutes after dosing, morphine brain accumulation increased by 2-fold when mouse (m)Abcb1 and mAbcg2 were ablated. Relative recovery of morphine in small intestinal content was significantly reduced in all the knockout strains. In the absence of mOatp1a/1b and mOatp2b1, plasma levels of M3G were markedly increased, suggesting a lower elimination rate. Moreover, Oatp-deficient mice displayed reduced hepatic and intestinal M3G accumulation. Mouse Oatps similarly affected plasma and tissue disposition of subcutaneously administered M6G. Human OATP1B1/1B3 transporters modestly contribute to the liver accumulation of M6G. In summary, mAbcb1, in combination with mAbcg2, limits morphine brain penetration and its net intestinal absorption. Variation in ABCB1 activity due to genetic polymorphisms/mutations and/or environmental factors might, therefore, partially affect morphine tissue exposure in patients. The ablation of mOatp1a/1b increases plasma exposure and decreases the liver and small intestinal disposition of M3G and M6G. Since the contribution of human OATP1B1/1B3 to M6G liver uptake was quite modest, the risks of undesirable drug interactions or interindividual variation related to OATP activity are likely negligible.
Collapse
Affiliation(s)
- Margarida L F Martins
- The Netherlands Cancer Institute, Division of Pharmacology, Amsterdam, the Netherlands
| | - Paniz Heydari
- The Netherlands Cancer Institute, Division of Pharmacology, Amsterdam, the Netherlands; Utrecht University, Faculty of Science, Department of Pharmaceutical Sciences, Division of Pharmacology, Utrecht, the Netherlands
| | - Wenlong Li
- The Netherlands Cancer Institute, Division of Pharmacology, Amsterdam, the Netherlands
| | - Alejandra Martínez-Chávez
- The Netherlands Cancer Institute, Division of Pharmacology, Amsterdam, the Netherlands; The Netherlands Cancer Institute, Department of Pharmacy & Pharmacology, Amsterdam, the Netherlands
| | - Malika El Yattouti
- The Netherlands Cancer Institute, Department of Pharmacy & Pharmacology, Amsterdam, the Netherlands
| | - Maria C Lebre
- The Netherlands Cancer Institute, Division of Pharmacology, Amsterdam, the Netherlands
| | - Jos H Beijnen
- The Netherlands Cancer Institute, Division of Pharmacology, Amsterdam, the Netherlands; The Netherlands Cancer Institute, Department of Pharmacy & Pharmacology, Amsterdam, the Netherlands; Utrecht University, Faculty of Science, Department of Pharmaceutical Sciences, Division of Pharmacoepidemiology & Clinical Pharmacology, Utrecht, the Netherlands
| | - Alfred H Schinkel
- The Netherlands Cancer Institute, Division of Pharmacology, Amsterdam, the Netherlands.
| |
Collapse
|
36
|
Helgudóttir SS, Johnsen KB, Routhe LG, Rasmussen CLM, Thomsen MS, Moos T. Upregulation of Transferrin Receptor 1 (TfR1) but Not Glucose Transporter 1 (GLUT1) or CD98hc at the Blood-Brain Barrier in Response to Valproic Acid. Cells 2024; 13:1181. [PMID: 39056763 PMCID: PMC11275047 DOI: 10.3390/cells13141181] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Revised: 07/08/2024] [Accepted: 07/09/2024] [Indexed: 07/28/2024] Open
Abstract
BACKGROUND Transferrin receptor 1 (TfR1), glucose transporter 1 (GLUT1), and CD98hc are candidates for targeted therapy at the blood-brain barrier (BBB). Our objective was to challenge the expression of TfR1, GLUT1, and CD98hc in brain capillaries using the histone deacetylase inhibitor (HDACi) valproic acid (VPA). METHODS Primary mouse brain capillary endothelial cells (BCECs) and brain capillaries isolated from mice injected intraperitoneally with VPA were examined using RT-qPCR and ELISA. Targeting to the BBB was performed by injecting monoclonal anti-TfR1 (Ri7217)-conjugated gold nanoparticles measured using ICP-MS. RESULTS In BCECs co-cultured with glial cells, Tfrc mRNA expression was significantly higher after 6 h VPA, returning to baseline after 24 h. In vivo Glut1 mRNA expression was significantly higher in males, but not females, receiving VPA, whereas Cd98hc mRNA expression was unaffected by VPA. TfR1 increased significantly in vivo after VPA, whereas GLUT1 and CD98hc were unchanged. The uptake of anti-TfR1-conjugated nanoparticles was unaltered by VPA despite upregulated TfR expression. CONCLUSIONS VPA upregulates TfR1 in brain endothelium in vivo and in vitro. VPA does not increase GLUT1 and CD98hc proteins. The increase in TfR1 does not result in higher anti-TfR1 antibody targetability, suggesting targeting sufficiently occurs with available transferrin receptors without further contribution from accessory VPA-induced TfR1.
Collapse
Affiliation(s)
- Steinunn Sara Helgudóttir
- Neurobiology Research and Drug Delivery (NRD), Department of Health Science and Technology, Aalborg University, 9260 Gistrup, Denmark; (S.S.H.); (L.G.R.); (C.L.M.R.)
| | - Kasper Bendix Johnsen
- Section for Biotherapeutic Engineering and Drug Targeting, Department of Health Technology, Technical University of Denmark, 2800 Lyngby, Denmark;
| | - Lisa Greve Routhe
- Neurobiology Research and Drug Delivery (NRD), Department of Health Science and Technology, Aalborg University, 9260 Gistrup, Denmark; (S.S.H.); (L.G.R.); (C.L.M.R.)
| | - Charlotte Laurfelt Munch Rasmussen
- Neurobiology Research and Drug Delivery (NRD), Department of Health Science and Technology, Aalborg University, 9260 Gistrup, Denmark; (S.S.H.); (L.G.R.); (C.L.M.R.)
| | - Maj Schneider Thomsen
- Neurobiology Research and Drug Delivery (NRD), Department of Health Science and Technology, Aalborg University, 9260 Gistrup, Denmark; (S.S.H.); (L.G.R.); (C.L.M.R.)
| | - Torben Moos
- Neurobiology Research and Drug Delivery (NRD), Department of Health Science and Technology, Aalborg University, 9260 Gistrup, Denmark; (S.S.H.); (L.G.R.); (C.L.M.R.)
| |
Collapse
|
37
|
Liang Y, Fan T, Bai M, Cui N, Li W, Wang J, Guan Y. Chikusetsu Saponin IVa liposomes modified with a retro-enantio peptide penetrating the blood-brain barrier to suppress pyroptosis in acute ischemic stroke rats. J Nanobiotechnology 2024; 22:393. [PMID: 38965602 PMCID: PMC11223377 DOI: 10.1186/s12951-024-02641-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2024] [Accepted: 06/15/2024] [Indexed: 07/06/2024] Open
Abstract
BACKGROUND The therapeutic strategies for acute ischemic stroke were faced with substantial constraints, emphasizing the necessity to safeguard neuronal cells during cerebral ischemia to reduce neurological impairments and enhance recovery outcomes. Despite its potential as a neuroprotective agent in stroke treatment, Chikusetsu saponin IVa encounters numerous challenges in clinical application. RESULT Brain-targeted liposomes modified with THRre peptides showed substantial uptake by bEnd. 3 and PC-12 cells and demonstrated the ability to cross an in vitro blood-brain barrier model, subsequently accumulating in PC-12 cells. In vivo, they could significantly accumulate in rat brain. Treatment with C-IVa-LPs-THRre notably reduced the expression of proteins in the P2RX7/NLRP3/Caspase-1 pathway and inflammatory factors. This was evidenced by decreased cerebral infarct size and improved neurological function in MCAO rats. CONCLUSION The findings indicate that C-IVa-LPs-THRre could serve as a promising strategy for targeting cerebral ischemia. This approach enhances drug concentration in the brain, mitigates pyroptosis, and improves the neuroinflammatory response associated with stroke.
Collapse
Affiliation(s)
- Yitong Liang
- Department of Pharmacy, Xijing Hospital, Air Force Medical University, Changle West Road 127, Xi'an, Shaanxi, China
| | - Tingting Fan
- Department of Pharmacy, Xijing Hospital, Air Force Medical University, Changle West Road 127, Xi'an, Shaanxi, China
| | - Min Bai
- Department of Geriatrics, Xijing Hospital, Air Force Medical University, Changle West Road 127, Xi'an, Shaanxi, China
| | - Na Cui
- Department of Pharmacy, Xijing Hospital, Air Force Medical University, Changle West Road 127, Xi'an, Shaanxi, China
| | - Wangting Li
- Department of Pharmacy, Xijing Hospital, Air Force Medical University, Changle West Road 127, Xi'an, Shaanxi, China
| | - Jingwen Wang
- Department of Pharmacy, Xijing Hospital, Air Force Medical University, Changle West Road 127, Xi'an, Shaanxi, China.
| | - Yue Guan
- Department of Pharmacy, Xijing Hospital, Air Force Medical University, Changle West Road 127, Xi'an, Shaanxi, China.
| |
Collapse
|
38
|
Ramsay E, Montaser AB, Niitsu K, Urtti A, Auriola S, Huttunen KM, Uchida Y, Kidron H, Terasaki T. Transporter Protein Expression of Corneal Epithelium in Rabbit and Porcine: Evaluation of Models for Ocular Drug Transport Study. Mol Pharm 2024; 21:3204-3217. [PMID: 38809137 PMCID: PMC11474527 DOI: 10.1021/acs.molpharmaceut.3c01210] [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] [Revised: 05/14/2024] [Accepted: 05/15/2024] [Indexed: 05/30/2024]
Abstract
The transcorneal route is the main entry route for drugs to the intraocular parts, after topical administration. The outer surface, the corneal epithelium (CE), forms the rate-limiting barrier for drug permeability. Information about the role and protein expression of drug and amino acid transporter proteins in the CE is sparse and lacking. The aim of our study was to characterize transporter protein expression in rabbit and porcine CE to better understand potential drug and nutrient absorption after topical administration. Proteins, mainly Abc and Slc transporters, were characterized with quantitative targeted absolute proteomics and global untargeted proteomics methods. In the rabbit CE, 24 of 48 proteins were detected in the targeted approach, and 21 of these were quantified. In the porcine CE, 26 of 58 proteins were detected in the targeted approach, and 20 of these were quantified. Among these, 15 proteins were quantified in both animals: 4f2hc (Slc3a2), Aqp0, Asct1 (Slc1a4), Asct2 (Slc1a5), Glut1 (Slc2a1), Hmit (Slc2a13), Insr, Lat1 (Slc7a5), Mct1 (Slc16a1), Mct2 (Slc16a7), Mct4 (Slc16a3), Mrp 4 (Abcc4), Na+/K+-ATPase, Oatp3a1 (Slco3a1), and Snat2 (Slc38a2). Overall, the global proteomics results supported the targeted proteomics results. Organic anion transporting polypeptide Oatp3a1 was detected and quantified for the first time in both rabbit (1.4 ± 0.4 fmol/cm2) and porcine (11.1 ± 5.3 fmol/cm2) CE. High expression levels were observed for L-type amino acid transporter, Lat1, which was quantified with newly selected extracellular domain peptides in rabbit (48.9 ± 11.8 fmol/cm2) and porcine (37.6 ± 11.5 fmol/cm2) CE. The knowledge of transporter protein expression in ocular barriers is a key factor in the successful design of new ocular drugs, pharmacokinetic modeling, understanding ocular diseases, and the translation to human.
Collapse
Affiliation(s)
- Eva Ramsay
- Drug
Research Programme, Division of Pharmaceutical Biosciences, Faculty
of Pharmacy, University of Helsinki, 00014 Helsinki, Finland
| | - Ahmed B. Montaser
- School
of Pharmacy, University of Eastern Finland, Yliopistonranta 1 C, 70211 Kuopio, Finland
| | - Kanako Niitsu
- School
of Pharmacy, University of Eastern Finland, Yliopistonranta 1 C, 70211 Kuopio, Finland
| | - Arto Urtti
- Drug
Research Programme, Division of Pharmaceutical Biosciences, Faculty
of Pharmacy, University of Helsinki, 00014 Helsinki, Finland
- School
of Pharmacy, University of Eastern Finland, Yliopistonranta 1 C, 70211 Kuopio, Finland
| | - Seppo Auriola
- School
of Pharmacy, University of Eastern Finland, Yliopistonranta 1 C, 70211 Kuopio, Finland
| | - Kristiina M. Huttunen
- School
of Pharmacy, University of Eastern Finland, Yliopistonranta 1 C, 70211 Kuopio, Finland
| | - Yasuo Uchida
- Department
of Molecular Systems Pharmaceutics, Graduate School of Biomedical
and Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima 734-0037, Japan
| | - Heidi Kidron
- Drug
Research Programme, Division of Pharmaceutical Biosciences, Faculty
of Pharmacy, University of Helsinki, 00014 Helsinki, Finland
| | - Tetsuya Terasaki
- School
of Pharmacy, University of Eastern Finland, Yliopistonranta 1 C, 70211 Kuopio, Finland
| |
Collapse
|
39
|
Carstens G, Verbeek MM, Rohlwink UK, Figaji AA, te Brake L, van Laarhoven A. Metabolite transport across central nervous system barriers. J Cereb Blood Flow Metab 2024; 44:1063-1077. [PMID: 38546534 PMCID: PMC11179608 DOI: 10.1177/0271678x241241908] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Revised: 02/02/2024] [Accepted: 02/27/2024] [Indexed: 06/13/2024]
Abstract
Metabolomic analysis of cerebrospinal fluid (CSF) is used to improve diagnostics and pathophysiological understanding of neurological diseases. Alterations in CSF metabolite levels can partly be attributed to changes in brain metabolism, but relevant transport processes influencing CSF metabolite concentrations should be considered. The entry of molecules including metabolites into the central nervous system (CNS), is tightly controlled by the blood-brain, blood-CSF, and blood-spinal cord barriers, where aquaporins and membrane-bound carrier proteins regulate influx and efflux via passive and active transport processes. This report therefore provides reference for future CSF metabolomic work, by providing a detailed summary of the current knowledge on the location and function of the involved transporters and routing of metabolites from blood to CSF and from CSF to blood.
Collapse
Affiliation(s)
- Gesa Carstens
- Department of Internal Medicine and Radboud Center of Infectious Diseases (RCI), Radboud University Medical Center, Nijmegen, Netherlands
| | - Marcel M Verbeek
- Departments of Neurology and Human Genetics, Radboud University Medical Center, Donders Institute for Brain, Cognition, and Behavior, Nijmegen, Netherlands
| | - Ursula K Rohlwink
- Division of Neurosurgery, Department of Surgery, Neuroscience Institute, University of Cape Town, Cape Town, South Africa
| | - Anthony A Figaji
- Division of Neurosurgery, Department of Surgery, Neuroscience Institute, University of Cape Town, Cape Town, South Africa
| | - Lindsey te Brake
- Department of Pharmacy, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Arjan van Laarhoven
- Department of Internal Medicine and Radboud Center of Infectious Diseases (RCI), Radboud University Medical Center, Nijmegen, Netherlands
| |
Collapse
|
40
|
Baltira C, Aronica E, Elmquist WF, Langer O, Löscher W, Sarkaria JN, Wesseling P, de Gooijer MC, van Tellingen O. The impact of ATP-binding cassette transporters in the diseased brain: Context matters. Cell Rep Med 2024; 5:101609. [PMID: 38897176 PMCID: PMC11228798 DOI: 10.1016/j.xcrm.2024.101609] [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: 08/22/2023] [Revised: 02/20/2024] [Accepted: 05/16/2024] [Indexed: 06/21/2024]
Abstract
ATP-binding cassette (ABC) transporters facilitate the movement of diverse molecules across cellular membranes, including those within the CNS. While most extensively studied in microvascular endothelial cells forming the blood-brain barrier (BBB), other CNS cell types also express these transporters. Importantly, disruptions in the CNS microenvironment during disease can alter transporter expression and function. Through this comprehensive review, we explore the modulation of ABC transporters in various brain pathologies and the context-dependent consequences of these changes. For instance, downregulation of ABCB1 may exacerbate amyloid beta plaque deposition in Alzheimer's disease and facilitate neurotoxic compound entry in Parkinson's disease. Upregulation may worsen neuroinflammation by aiding chemokine-mediated CD8 T cell influx into multiple sclerosis lesions. Overall, ABC transporters at the BBB hinder drug entry, presenting challenges for effective pharmacotherapy. Understanding the context-dependent changes in ABC transporter expression and function is crucial for elucidating the etiology and developing treatments for brain diseases.
Collapse
Affiliation(s)
- Chrysiida Baltira
- Division of Pharmacology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Eleonora Aronica
- Amsterdam University Medical Centers, University of Amsterdam, Amsterdam Neuroscience, Department of (Neuro)Pathology, Amsterdam, the Netherlands; Stichting Epilepsie Instellingen Nederland (SEIN), Heemstede, the Netherlands
| | - William F Elmquist
- Brain Barriers Research Center, Department of Pharmaceutics, College of Pharmacy, University of Minnesota, Minneapolis, MN, USA
| | - Oliver Langer
- Department of Clinical Pharmacology, Medical University of Vienna, Vienna, Austria; Department of Biomedical Imaging and Image-guided Therapy, Medical University of Vienna, Vienna, Austria
| | - Wolfgang Löscher
- Translational Neuropharmacology Lab, NIFE, Department of Experimental Otology of the ENT Clinics, Hannover Medical School, Hannover, Germany; Center for Systems Neuroscience, Hannover, Germany
| | - Jann N Sarkaria
- Department of Radiation Oncology, Mayo Clinic, Rochester, MN, USA
| | - Pieter Wesseling
- Department of Pathology, Amsterdam University Medical Centers, Amsterdam, the Netherlands; Laboratory for Childhood Cancer Pathology, Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands
| | - Mark C de Gooijer
- Division of Pharmacology, The Netherlands Cancer Institute, Amsterdam, the Netherlands; Faculty of Biology, Medicine and Health, University of Manchester; The Christie NHS Foundation Trust, Manchester, UK.
| | - Olaf van Tellingen
- Division of Pharmacology, The Netherlands Cancer Institute, Amsterdam, the Netherlands; Mouse Cancer Clinic, The Netherlands Cancer Institute, Amsterdam, the Netherlands.
| |
Collapse
|
41
|
Bhatt M, Lazzarin E, Alberto-Silva AS, Domingo G, Zerlotti R, Gradisch R, Bazzone A, Sitte HH, Stockner T, Bossi E. Unveiling the crucial role of betaine: modulation of GABA homeostasis via SLC6A1 transporter (GAT1). Cell Mol Life Sci 2024; 81:269. [PMID: 38884791 PMCID: PMC11335192 DOI: 10.1007/s00018-024-05309-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Revised: 06/04/2024] [Accepted: 06/06/2024] [Indexed: 06/18/2024]
Abstract
Betaine is an endogenous osmolyte that exhibits therapeutic potential by mitigating various neurological disorders. However, the underlying cellular and molecular mechanisms responsible for its neuroprotective effects remain puzzling.In this study, we describe a possible mechanism behind the positive impact of betaine in preserving neurons from excitotoxicity. Here we demonstrate that betaine at low concentration modulates the GABA uptake by GAT1 (slc6a1), the predominant GABA transporter in the central nervous system. This modulation occurs through the temporal inhibition of the transporter, wherein prolonged occupancy by betaine impedes the swift transition of the transporter to the inward conformation. Importantly, the modulatory effect of betaine on GAT1 is reversible, as the blocking of GAT1 disappears with increased extracellular GABA. Using electrophysiology, mass spectroscopy, radiolabelled cellular assay, and molecular dynamics simulation we demonstrate that betaine has a dual role in GAT1: at mM concentration acts as a slow substrate, and at µM as a temporal blocker of GABA, when it is below its K0.5. Given this unique modulatory characteristic and lack of any harmful side effects, betaine emerges as a promising neuromodulator of the inhibitory pathways improving GABA homeostasis via GAT1, thereby conferring neuroprotection against excitotoxicity.
Collapse
Affiliation(s)
- Manan Bhatt
- Department of Biotechnology and Life Science, Laboratory of Cellular and Molecular Physiology, University of Insubria, Via J. H. Dunant 3, 21100, Varese, Italy
| | - Erika Lazzarin
- Center for Physiology and Pharmacology, Institute of Pharmacology, Medical University of Vienna, 1090, Vienna, Austria
| | - Ana Sofia Alberto-Silva
- Center for Physiology and Pharmacology, Institute of Pharmacology, Medical University of Vienna, 1090, Vienna, Austria
| | - Guido Domingo
- Department of Biotechnology and Life Science, Laboratory of Cellular and Molecular Physiology, University of Insubria, Via J. H. Dunant 3, 21100, Varese, Italy
| | - Rocco Zerlotti
- Nanion Technologies GmbH, Ganghoferstr. 70a, 80339, Munich, Germany
| | - Ralph Gradisch
- Institute of Pharmacology and Toxicology, University of Zurich, Zurich, Switzerland
| | - Andre Bazzone
- Nanion Technologies GmbH, Ganghoferstr. 70a, 80339, Munich, Germany
| | - Harald H Sitte
- Center for Physiology and Pharmacology, Institute of Pharmacology, Medical University of Vienna, 1090, Vienna, Austria
- Hourani Center for Applied Scientific Research, Al-Ahliyya Amman University, Amman, 19328, Jordan
- Center for Addiction Research and Science, Medical University of Vienna, 1090, Vienna, Austria
| | - Thomas Stockner
- Center for Physiology and Pharmacology, Institute of Pharmacology, Medical University of Vienna, 1090, Vienna, Austria
| | - Elena Bossi
- Department of Biotechnology and Life Science, Laboratory of Cellular and Molecular Physiology, University of Insubria, Via J. H. Dunant 3, 21100, Varese, Italy.
- Centre for Neuroscience, University of Insubria, 21100, Varese, Italy.
| |
Collapse
|
42
|
Squalli Houssaini A, Lamrabet S, Nshizirungu JP, Senhaji N, Sekal M, Karkouri M, Bennis S. Glioblastoma Vaccines as Promising Immune-Therapeutics: Challenges and Current Status. Vaccines (Basel) 2024; 12:655. [PMID: 38932383 PMCID: PMC11209492 DOI: 10.3390/vaccines12060655] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Revised: 05/02/2024] [Accepted: 05/08/2024] [Indexed: 06/28/2024] Open
Abstract
Glioblastoma (GBM) is the most common and aggressive malignant brain tumor. Standard treatments including surgical resection, radiotherapy, and chemotherapy, have failed to significantly improve the prognosis of glioblastoma patients. Currently, immunotherapeutic approaches based on vaccines, chimeric antigen-receptor T-cells, checkpoint inhibitors, and oncolytic virotherapy are showing promising results in clinical trials. The combination of different immunotherapeutic approaches is proving satisfactory and promising. In view of the challenges of immunotherapy and the resistance of glioblastomas, the treatment of these tumors requires further efforts. In this review, we explore the obstacles that potentially influence the efficacy of the response to immunotherapy and that should be taken into account in clinical trials. This article provides a comprehensive review of vaccine therapy for glioblastoma. In addition, we identify the main biomarkers, including isocitrate dehydrogenase, epidermal growth factor receptor, and telomerase reverse transcriptase, known as potential immunotherapeutic targets in glioblastoma, as well as the current status of clinical trials. This paper also lists proposed solutions to overcome the obstacles facing immunotherapy in glioblastomas.
Collapse
Affiliation(s)
- Asmae Squalli Houssaini
- Laboratory of Biomedical and Translational Research, Faculty of Medicine, Pharmacy and Dental Medicine of Fez, Sidi Mohamed Ben Abdellah University, Fez 30070, Morocco;
| | - Salma Lamrabet
- Laboratory of Biomedical and Translational Research, Faculty of Medicine, Pharmacy and Dental Medicine of Fez, Sidi Mohamed Ben Abdellah University, Fez 30070, Morocco;
| | - Jean Paul Nshizirungu
- Biology Department, School of Science, College of Science and Technology, University of Rwanda, Kigali P.O. Box 3900, Rwanda;
| | - Nadia Senhaji
- Department of Biology, Faculty of Sciences, Moulay Ismail University, Meknes 50000, Morocco;
| | - Mohammed Sekal
- Laboratory of Epidemiology and Research in Health Sciences, Faculty of Medicine, Pharmacy and Dental Medicine of Fez, Sidi Mohamed Ben Abdellah University, Fez 30070, Morocco;
| | - Mehdi Karkouri
- Department of Pathological Anatomy, Ibn Rochd University Hospital of Casablanca, Casablanca 20250, Morocco;
- Laboratory of Cellular and molecular Pathology, Faculty of Medicine and Pharmacy of Casablanca, Hassan II University, Casablanca 20360, Morocco
| | - Sanae Bennis
- Laboratory of Biomedical and Translational Research, Faculty of Medicine, Pharmacy and Dental Medicine of Fez, Sidi Mohamed Ben Abdellah University, Fez 30070, Morocco;
| |
Collapse
|
43
|
Prieto Garcia L, Vildhede A, Nordell P, Ahlström C, Montaser AB, Terasaki T, Lennernäs H, Sjögren E. Physiologically based pharmacokinetics modeling and transporter proteomics to predict systemic and local liver and muscle disposition of statins. CPT Pharmacometrics Syst Pharmacol 2024; 13:1029-1043. [PMID: 38576225 PMCID: PMC11179708 DOI: 10.1002/psp4.13139] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Revised: 02/22/2024] [Accepted: 03/25/2024] [Indexed: 04/06/2024] Open
Abstract
Statins are used to reduce liver cholesterol levels but also carry a dose-related risk of skeletal muscle toxicity. Concentrations of statins in plasma are often used to assess efficacy and safety, but because statins are substrates of membrane transporters that are present in diverse tissues, local differences in intracellular tissue concentrations cannot be ruled out. Thus, plasma concentration may not be an adequate indicator of efficacy and toxicity. To bridge this gap, we used physiologically based pharmacokinetic (PBPK) modeling to predict intracellular concentrations of statins. Quantitative data on transporter clearance were scaled from in vitro to in vivo conditions by integrating targeted proteomics and transporter kinetics data. The developed PBPK models, informed by proteomics, suggested that organic anion-transporting polypeptide 2B1 (OATP2B1) and multidrug resistance-associated protein 1 (MRP1) play a pivotal role in the distribution of statins in muscle. Using these PBPK models, we were able to predict the impact of alterations in transporter function due to genotype or drug-drug interactions on statin systemic concentrations and exposure in liver and muscle. These results underscore the potential of proteomics-guided PBPK modeling to scale transporter clearance from in vitro data to real-world implications. It is important to evaluate the role of drug transporters when predicting tissue exposure associated with on- and off-target effects.
Collapse
Affiliation(s)
- Luna Prieto Garcia
- Department of Pharmaceutical Bioscience, Translational Drug Discovery and DevelopmentUppsala UniversityUppsalaSweden
- DMPK, Research and Early Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZenecaGothenburgSweden
| | - Anna Vildhede
- DMPK, Research and Early Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZenecaGothenburgSweden
| | - Pär Nordell
- DMPK, Research and Early Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZenecaGothenburgSweden
| | - Christine Ahlström
- DMPK, Research and Early Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZenecaGothenburgSweden
| | - Ahmed B. Montaser
- School of Pharmacy, Faculty of Health SciencesUniversity of Eastern FinlandKuopioFinland
| | - Tetsuya Terasaki
- School of Pharmacy, Faculty of Health SciencesUniversity of Eastern FinlandKuopioFinland
| | - Hans Lennernäs
- Department of Pharmaceutical Bioscience, Translational Drug Discovery and DevelopmentUppsala UniversityUppsalaSweden
| | - Erik Sjögren
- Department of Pharmaceutical Bioscience, Translational Drug Discovery and DevelopmentUppsala UniversityUppsalaSweden
| |
Collapse
|
44
|
Sóskuti E, Szilvásy N, Temesszentandrási-Ambrus C, Urbán Z, Csíkvári O, Szabó Z, Kecskeméti G, Pusztai É, Gáborik Z. Applicability of MDR1 Overexpressing Abcb1KO-MDCKII Cell Lines for Investigating In Vitro Species Differences and Brain Penetration Prediction. Pharmaceutics 2024; 16:736. [PMID: 38931858 PMCID: PMC11207571 DOI: 10.3390/pharmaceutics16060736] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Revised: 05/22/2024] [Accepted: 05/24/2024] [Indexed: 06/28/2024] Open
Abstract
Implementing the 3R initiative to reduce animal experiments in brain penetration prediction for CNS-targeting drugs requires more predictive in vitro and in silico models. However, animal studies are still indispensable to obtaining brain concentration and determining the prediction performance of in vitro models. To reveal species differences and provide reliable data for IVIVE, in vitro models are required. Systems overexpressing MDR1 and BCRP are widely used to predict BBB penetration, highlighting the impact of the in vitro system on predictive performance. In this study, endogenous Abcb1 knock-out MDCKII cells overexpressing MDR1 of human, mouse, rat or cynomolgus monkey origin were used. Good correlations between ERs of 83 drugs determined in each cell line suggest limited species specificities. All cell lines differentiated CNS-penetrating compounds based on ERs with high efficiency and sensitivity. The correlation between in vivo and predicted Kp,uu,brain was the highest using total ER of human MDR1 and BCRP and optimized scaling factors. MDR1 interactors were tested on all MDR1 orthologs using digoxin and quinidine as substrates. We found several examples of inhibition dependent on either substrate or transporter abundance. In summary, this assay system has the potential for early-stage brain penetration screening. IC50 comparison between orthologs is complex; correlation with transporter abundance data is not necessarily proportional and requires the understanding of modes of transporter inhibition.
Collapse
Affiliation(s)
- Emőke Sóskuti
- Charles River Laboratories Hungary, H-1117 Budapest, Hungary; (E.S.); (N.S.); (C.T.-A.); (Z.U.); (O.C.)
- Doctoral School of Semmelweis University, Molecular Medicine Division, H-1085 Budapest, Hungary
| | - Nóra Szilvásy
- Charles River Laboratories Hungary, H-1117 Budapest, Hungary; (E.S.); (N.S.); (C.T.-A.); (Z.U.); (O.C.)
| | | | - Zoltán Urbán
- Charles River Laboratories Hungary, H-1117 Budapest, Hungary; (E.S.); (N.S.); (C.T.-A.); (Z.U.); (O.C.)
| | - Olivér Csíkvári
- Charles River Laboratories Hungary, H-1117 Budapest, Hungary; (E.S.); (N.S.); (C.T.-A.); (Z.U.); (O.C.)
| | - Zoltán Szabó
- Department of Medical Chemistry, Albert Szent-Györgyi Medical School, University of Szeged, H-6720 Szeged, Hungary; (Z.S.); (G.K.)
| | - Gábor Kecskeméti
- Department of Medical Chemistry, Albert Szent-Györgyi Medical School, University of Szeged, H-6720 Szeged, Hungary; (Z.S.); (G.K.)
| | - Éva Pusztai
- Department of Chemical and Environmental Process Engineering, Faculty of Chemical Technology and Biotechnology, Budapest University of Technology and Economics, H-1111 Budapest, Hungary;
| | - Zsuzsanna Gáborik
- Charles River Laboratories Hungary, H-1117 Budapest, Hungary; (E.S.); (N.S.); (C.T.-A.); (Z.U.); (O.C.)
| |
Collapse
|
45
|
Porkoláb G, Mészáros M, Szecskó A, Vigh JP, Walter FR, Figueiredo R, Kálomista I, Hoyk Z, Vizsnyiczai G, Gróf I, Jan JS, Gosselet F, Pirity MK, Vastag M, Hudson N, Campbell M, Veszelka S, Deli MA. Synergistic induction of blood-brain barrier properties. Proc Natl Acad Sci U S A 2024; 121:e2316006121. [PMID: 38748577 PMCID: PMC11126970 DOI: 10.1073/pnas.2316006121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Accepted: 04/05/2024] [Indexed: 05/27/2024] Open
Abstract
Blood-brain barrier (BBB) models derived from human stem cells are powerful tools to improve our understanding of cerebrovascular diseases and to facilitate drug development for the human brain. Yet providing stem cell-derived endothelial cells with the right signaling cues to acquire BBB characteristics while also retaining their vascular identity remains challenging. Here, we show that the simultaneous activation of cyclic AMP and Wnt/β-catenin signaling and inhibition of the TGF-β pathway in endothelial cells robustly induce BBB properties in vitro. To target this interaction, we present a small-molecule cocktail named cARLA, which synergistically enhances barrier tightness in a range of BBB models across species. Mechanistically, we reveal that the three pathways converge on Wnt/β-catenin signaling to mediate the effect of cARLA via the tight junction protein claudin-5. We demonstrate that cARLA shifts the gene expressional profile of human stem cell-derived endothelial cells toward the in vivo brain endothelial signature, with a higher glycocalyx density and efflux pump activity, lower rates of endocytosis, and a characteristic endothelial response to proinflammatory cytokines. Finally, we illustrate how cARLA can improve the predictive value of human BBB models regarding the brain penetration of drugs and targeted nanoparticles. Due to its synergistic effect, high reproducibility, and ease of use, cARLA has the potential to advance drug development for the human brain by improving BBB models across laboratories.
Collapse
Affiliation(s)
- Gergő Porkoláb
- Institute of Biophysics, Biological Research Centre, Hungarian Research Network, SzegedH-6726, Hungary
- Doctoral School of Biology, University of Szeged, SzegedH-6720, Hungary
| | - Mária Mészáros
- Institute of Biophysics, Biological Research Centre, Hungarian Research Network, SzegedH-6726, Hungary
| | - Anikó Szecskó
- Institute of Biophysics, Biological Research Centre, Hungarian Research Network, SzegedH-6726, Hungary
- Doctoral School of Biology, University of Szeged, SzegedH-6720, Hungary
| | - Judit P. Vigh
- Institute of Biophysics, Biological Research Centre, Hungarian Research Network, SzegedH-6726, Hungary
- Doctoral School of Biology, University of Szeged, SzegedH-6720, Hungary
| | - Fruzsina R. Walter
- Institute of Biophysics, Biological Research Centre, Hungarian Research Network, SzegedH-6726, Hungary
| | | | - Ildikó Kálomista
- In Vitro Metabolism Laboratory, Gedeon Richter, BudapestH-1103, Hungary
| | - Zsófia Hoyk
- Institute of Biophysics, Biological Research Centre, Hungarian Research Network, SzegedH-6726, Hungary
| | - Gaszton Vizsnyiczai
- Institute of Biophysics, Biological Research Centre, Hungarian Research Network, SzegedH-6726, Hungary
| | - Ilona Gróf
- Institute of Biophysics, Biological Research Centre, Hungarian Research Network, SzegedH-6726, Hungary
| | - Jeng-Shiung Jan
- Department of Chemical Engineering, National Cheng Kung University, Tainan70101, Taiwan
| | - Fabien Gosselet
- Laboratoire de la Barriére Hémato-Encéphalique, Université d’Artois, Lens62307, France
| | - Melinda K. Pirity
- Institute of Genetics, Biological Research Centre, Hungarian Research Network, SzegedH-6726, Hungary
| | - Monika Vastag
- In Vitro Metabolism Laboratory, Gedeon Richter, BudapestH-1103, Hungary
| | - Natalie Hudson
- Smurfit Institute of Genetics, Trinity College Dublin, DublinD02 VF25, Ireland
| | - Matthew Campbell
- Smurfit Institute of Genetics, Trinity College Dublin, DublinD02 VF25, Ireland
| | - Szilvia Veszelka
- Institute of Biophysics, Biological Research Centre, Hungarian Research Network, SzegedH-6726, Hungary
| | - Mária A. Deli
- Institute of Biophysics, Biological Research Centre, Hungarian Research Network, SzegedH-6726, Hungary
| |
Collapse
|
46
|
Hasan I, Guo B, Zhang J, Chang C. Advances in Antioxidant Nanomedicines for Imaging and Therapy of Alzheimer's Disease. Antioxid Redox Signal 2024; 40:863-888. [PMID: 36070437 DOI: 10.1089/ars.2022.0107] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Significance: Reactive oxygen species (ROS) are crucial signaling molecules in the regulation of numerous physiological activities including the formation and function of the central nervous system (CNS). So far, many functional antioxidant nanomedicines with ROS scavenging capability to reduce oxidative stress in Alzheimer's disease (AD) have been developed for both imaging and therapy of AD. Recent Advances: This review focuses on the most recent advances in antioxidant nanomedicines such as ROS-scavenging nanoparticles (NPs), NPs with intrinsic antioxidant activity, and drug-loaded antioxidant NPs for AD theranostics. In addition to antioxidant nanomedicines, the emerging phototherapy treatment paradigms and the promising preclinic drug carriers, such as exosomes and liposomes, are also introduced. Critical Issues: In general, excessive generation of ROS can cause lipid peroxidation, oxidative DNA, as well as protein damage, aggravating pathogenic alterations, accumulation of amyloid-beta plaques and neurofibrillary tangles in the brain. These negative factors further cause cell death, which is the beginning of AD. Future Directions: We anticipate that this review will help researchers in the area of preclinical research and clinical translation of antioxidant nanomedicines for AD imaging and therapy.
Collapse
Affiliation(s)
- Ikram Hasan
- School of Biomedical Engineering, Health Science Center, Shenzhen University, Shenzhen, China
| | - Bing Guo
- School of Science and Shenzhen Key Laboratory of Flexible Printed Electronics Technology, Harbin Institute of Technology, Shenzhen, China
| | - Jian Zhang
- School of Pharmaceutical Sciences, Health Science Center, Shenzhen University, Shenzhen, China
| | - Chunqi Chang
- School of Biomedical Engineering, Health Science Center, Shenzhen University, Shenzhen, China
| |
Collapse
|
47
|
Huttunen KM. Improving drug delivery to the brain: the prodrug approach. Expert Opin Drug Deliv 2024; 21:683-693. [PMID: 38738934 DOI: 10.1080/17425247.2024.2355180] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Accepted: 05/10/2024] [Indexed: 05/14/2024]
Abstract
INTRODUCTION The prodrug approach has been thought to be a simple solution to improve brain drug delivery for decades. Nevertheless, it still comes as a surprise that there is relatively little success in the field. The best example anti-parkinsonian drug levodopa has been serendipitously discovered to be a transporter-utilizing brain-delivered prodrug rather than a rationally developed one. AREAS COVERED The lack of success can mainly be explained by the insufficient understanding of the role of membrane proteins that can facilitate drug delivery at dynamic barriers, such as the blood-brain barrier (BBB), but also by the sparse knowledge of prodrug bioconverting enzymes in the brain. This review summarizes the current status of the prodrug attempts that have been developed in the past to improve brain drug delivery. EXPERT OPINION With the expandingly improved analytical and computational technologies, it is anticipated that enhanced brain drug delivery will be eventually achieved for most of the central nervous system (CNS) acting drugs. However, this requires that carrier-mediated (pro)drug delivery methods are implemented in the very early phases of the drug development processes and not as a last step to survive a problematic investigational drug candidate.
Collapse
Affiliation(s)
- Kristiina M Huttunen
- School of Pharmacy, Faculty of Health Sciences, University of Eastern Finland, Kuopio, Finland
| |
Collapse
|
48
|
Chasseigneaux S, Cochois-Guégan V, Lecorgne L, Lochus M, Nicolic S, Blugeon C, Jourdren L, Gomez-Zepeda D, Tenzer S, Sanquer S, Nivet-Antoine V, Menet MC, Laplanche JL, Declèves X, Cisternino S, Saubaméa B. Fasting upregulates the monocarboxylate transporter MCT1 at the rat blood-brain barrier through PPAR δ activation. Fluids Barriers CNS 2024; 21:33. [PMID: 38589879 PMCID: PMC11003008 DOI: 10.1186/s12987-024-00526-8] [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: 10/28/2023] [Accepted: 02/29/2024] [Indexed: 04/10/2024] Open
Abstract
BACKGROUND The blood-brain barrier (BBB) is pivotal for the maintenance of brain homeostasis and it strictly regulates the cerebral transport of a wide range of endogenous compounds and drugs. While fasting is increasingly recognized as a potential therapeutic intervention in neurology and psychiatry, its impact upon the BBB has not been studied. This study was designed to assess the global impact of fasting upon the repertoire of BBB transporters. METHODS We used a combination of in vivo and in vitro experiments to assess the response of the brain endothelium in male rats that were fed ad libitum or fasted for one to three days. Brain endothelial cells were acutely purified and transcriptionaly profiled using RNA-Seq. Isolated brain microvessels were used to assess the protein expression of selected BBB transporters through western blot. The molecular mechanisms involved in the adaptation to fasting were investigated in primary cultured rat brain endothelial cells. MCT1 activity was probed by in situ brain perfusion. RESULTS Fasting did not change the expression of the main drug efflux ATP-binding cassette transporters or P-glycoprotein activity at the BBB but modulated a restrictive set of solute carrier transporters. These included the ketone bodies transporter MCT1, which is pivotal for the brain adaptation to fasting. Our findings in vivo suggested that PPAR δ, a major lipid sensor, was selectively activated in brain endothelial cells in response to fasting. This was confirmed in vitro where pharmacological agonists and free fatty acids selectively activated PPAR δ, resulting in the upregulation of MCT1 expression. Moreover, dosing rats with a specific PPAR δ antagonist blocked the upregulation of MCT1 expression and activity induced by fasting. CONCLUSIONS Altogether, our study shows that fasting affects a selected set of BBB transporters which does not include the main drug efflux transporters. Moreover, we describe a previously unknown selective adaptive response of the brain vasculature to fasting which involves PPAR δ and is responsible for the up-regulation of MCT1 expression and activity. Our study opens new perspectives for the metabolic manipulation of the BBB in the healthy or diseased brain.
Collapse
Affiliation(s)
- Stéphanie Chasseigneaux
- Optimisation Thérapeutique en Neuropsychopharmacologie, Université Paris Cité, Inserm, 4 avenue de l'Observatoire, 75006, Paris, France
| | - Véronique Cochois-Guégan
- Optimisation Thérapeutique en Neuropsychopharmacologie, Université Paris Cité, Inserm, 4 avenue de l'Observatoire, 75006, Paris, France
| | - Lucas Lecorgne
- Optimisation Thérapeutique en Neuropsychopharmacologie, Université Paris Cité, Inserm, 4 avenue de l'Observatoire, 75006, Paris, France
| | - Murielle Lochus
- Optimisation Thérapeutique en Neuropsychopharmacologie, Université Paris Cité, Inserm, 4 avenue de l'Observatoire, 75006, Paris, France
| | - Sophie Nicolic
- Optimisation Thérapeutique en Neuropsychopharmacologie, Université Paris Cité, Inserm, 4 avenue de l'Observatoire, 75006, Paris, France
| | - Corinne Blugeon
- Département de biologie, GenomiqueENS, Institut de Biologie de l'ENS (IBENS), École normale supérieure, CNRS, INSERM, Université PSL, 75005, Paris, France
| | - Laurent Jourdren
- Département de biologie, GenomiqueENS, Institut de Biologie de l'ENS (IBENS), École normale supérieure, CNRS, INSERM, Université PSL, 75005, Paris, France
| | - David Gomez-Zepeda
- Helmholtz-Institute for Translational Oncology Mainz (HI-TRON Mainz), A Hemlholtz Institute of the DKFZ, Mainz, Germany
- German Cancer Research Center (DKFZ) Heidelberg, Division 191, 69120, Heidelberg, Germany
- Institute of Immunology, University Medical Center of the Johannes-Gutenberg University, Mainz, Germany
| | - Stefan Tenzer
- Helmholtz-Institute for Translational Oncology Mainz (HI-TRON Mainz), A Hemlholtz Institute of the DKFZ, Mainz, Germany
- German Cancer Research Center (DKFZ) Heidelberg, Division 191, 69120, Heidelberg, Germany
- Institute of Immunology, University Medical Center of the Johannes-Gutenberg University, Mainz, Germany
- Research Center for Immunotherapy (FZI), University Medical Center of the Johannes-Gutenberg University, Mainz, Germany
| | | | - Valérie Nivet-Antoine
- AP-HP Biochimie générale, Hôpital Necker Enfants Malades, Université Paris Cité, Inserm, Innovations Thérapeutiques en Hémostase, Paris, France
| | - Marie-Claude Menet
- Institut de Chimie Physique, CNRS UMR8000, Université Paris-Saclay, 91400, Orsay, France
| | - Jean-Louis Laplanche
- Optimisation Thérapeutique en Neuropsychopharmacologie, Université Paris Cité, Inserm, 4 avenue de l'Observatoire, 75006, Paris, France
| | - Xavier Declèves
- Optimisation Thérapeutique en Neuropsychopharmacologie, Université Paris Cité, Inserm, 4 avenue de l'Observatoire, 75006, Paris, France
| | - Salvatore Cisternino
- Optimisation Thérapeutique en Neuropsychopharmacologie, Université Paris Cité, Inserm, 4 avenue de l'Observatoire, 75006, Paris, France
| | - Bruno Saubaméa
- Optimisation Thérapeutique en Neuropsychopharmacologie, Université Paris Cité, Inserm, 4 avenue de l'Observatoire, 75006, Paris, France.
| |
Collapse
|
49
|
Kim BS, Kim JU, Lee J, Ryu KM, Kim SH, Hwang NS. Decellularized brain extracellular matrix based NGF-releasing cryogel for brain tissue engineering in traumatic brain injury. J Control Release 2024; 368:140-156. [PMID: 38373473 DOI: 10.1016/j.jconrel.2024.02.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Revised: 02/05/2024] [Accepted: 02/12/2024] [Indexed: 02/21/2024]
Abstract
Traumatic brain injuries(TBI) pose significant challenges to human health, specifically neurological disorders and related motor activities. After TBI, the injured neuronal tissue is known for hardly regenerated and recovered to their normal neuron physiology and tissue compositions. For this reason, tissue engineering strategies that promote neuronal regeneration have gained increasing attention. This study explored the development of a novel neural tissue regeneration cryogel by combining brain-derived decellularized extracellular matrix (ECM) with heparin sulfate crosslinking that can perform nerve growth factor (NGF) release ability. Morphological and mechanical characterizations of the cryogels were performed to assess their suitability as a neural regeneration platform. After that, the heparin concnentration dependent effects of varying NGF concentrations on cryogel were investigated for their controlled release and impact on neuronal cell differentiation. The results revealed a direct correlation between the concentration of released NGF and the heparin sulfate ratio in cryogel, indicating that the cryogel can be tailored to carry higher loads of NGF with heparin concentration in cryogel that induced higher neuronal cell differentiation ratio. Furthermore, the study evaluated the NGF loaded cryogels on neuronal cell proliferation and brain tissue regeneration in vivo. The in vivo results suggested that the NGF loaded brain ECM derived cryogel significantly affects the regeneration of brain tissue. Overall, this research contributes to the development of advanced neural tissue engineering strategies and provides valuable insights into the design of regenerative cryogels that can be customized for specific therapeutic applications.
Collapse
Affiliation(s)
- Beom-Seok Kim
- Interdisciplinary Program in Bioengineering, Seoul National University, Seoul 08826, Republic of Korea
| | - Jeong-Uk Kim
- School of Chemical and Biological Engineering, Institute of Chemical Processes, Seoul National University, Seoul 08826, Republic of Korea
| | - Jaewoo Lee
- School of Chemical and Biological Engineering, Institute of Chemical Processes, Seoul National University, Seoul 08826, Republic of Korea
| | - Kyung Min Ryu
- School of Chemical and Biological Engineering, Institute of Chemical Processes, Seoul National University, Seoul 08826, Republic of Korea
| | - Su-Hwan Kim
- Department of Chemical Engineering (BK21 FOUR), Dong-A University, Busan 49315, Republic of Korea
| | - Nathaniel S Hwang
- Interdisciplinary Program in Bioengineering, Seoul National University, Seoul 08826, Republic of Korea; School of Chemical and Biological Engineering, Institute of Chemical Processes, Seoul National University, Seoul 08826, Republic of Korea; Bio-MAX Institute, Institute of Bio-Engineering, Seoul National University, Seoul 08826, Republic of Korea; Institute of Engineering Research, Seoul National University, Seoul 08826, Republic of Korea.
| |
Collapse
|
50
|
Puris E, Saveleva L, Auriola S, Gynther M, Kanninen KM, Fricker G. Sex-specific changes in protein expression of membrane transporters in the brain cortex of 5xFAD mouse model of Alzheimer's disease. Front Pharmacol 2024; 15:1365051. [PMID: 38572427 PMCID: PMC10989684 DOI: 10.3389/fphar.2024.1365051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Accepted: 02/27/2024] [Indexed: 04/05/2024] Open
Abstract
Membrane transporters playing an important role in the passage of drugs, metabolites and nutrients across the membranes of the brain cells have been shown to be involved in pathogenesis of Alzheimer's disease (AD). However, little is known about sex-specific changes in transporter protein expression at the brain in AD. Here, we investigated sex-specific alterations in protein expression of three ATP-binding cassette (ABC) and five solute carriers (SLC) transporters in the prefrontal cortex of a commonly used model of familial AD (FAD), 5xFAD mice. Sensitive liquid chromatography tandem mass spectrometry-based quantitative targeted absolute proteomic analysis was applied for absolute quantification of transporter protein expression. We compared the changes in transporter protein expressions in 7-month-old male and female 5xFAD mice versus sex-matched wild-type mice. The study revealed a significant sex-specific increase in protein expression of ABCC1 (p = 0.007) only in male 5xFAD mice as compared to sex-matched wild-type animals. In addition, the increased protein expression of glucose transporter 1 (p = 0.01), 4F2 cell-surface antigen heavy chain (p = 0.01) and long-chain fatty acid transport protein 1 (p = 0.02) were found only in female 5xFAD mice as compared to sex-matched wild-type animals. Finally, protein expression of alanine/serine/cysteine/threonine transporter 1 was upregulated in both male (p = 0.02) and female (p = 0.002) 5xFAD mice. The study provides important information about sex-specific changes in brain cortical transporter expression in 5xFAD mice, which will facilitate drug development of therapeutic strategies for AD targeting these transporters and drug delivery research.
Collapse
Affiliation(s)
- Elena Puris
- Institute of Pharmacy and Molecular Biotechnology, Ruprecht-Karls-University, Heidelberg, Germany
| | - Liudmila Saveleva
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Seppo Auriola
- School of Pharmacy, University of Eastern Finland, Kuopio, Finland
| | - Mikko Gynther
- School of Pharmacy, University of Eastern Finland, Kuopio, Finland
| | - Katja M. Kanninen
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Gert Fricker
- Institute of Pharmacy and Molecular Biotechnology, Ruprecht-Karls-University, Heidelberg, Germany
| |
Collapse
|