1
|
Soliman Y, Al-Khodor J, Yildirim Köken G, Mustafaoglu N. A guide for blood-brain barrier models. FEBS Lett 2024. [PMID: 39533665 DOI: 10.1002/1873-3468.15053] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Revised: 10/18/2024] [Accepted: 10/20/2024] [Indexed: 11/16/2024]
Abstract
Understanding the intricate mechanisms underlying brain-related diseases hinges on unraveling the pivotal role of the blood-brain barrier (BBB), an essential dynamic interface crucial for maintaining brain equilibrium. This review offers a comprehensive analysis of BBB physiology, delving into its cellular and molecular components while exploring a wide range of in vivo and in vitro BBB models. Notably, recent advancements in 3D cell culture techniques are explicitly discussed, as they have significantly improved the fidelity of BBB modeling by enabling the replication of physiologically relevant environments under flow conditions. Special attention is given to the cellular aspects of in vitro BBB models, alongside discussions on advances in stem cell technologies, providing valuable insights into generating robust cellular systems for BBB modeling. The diverse array of cell types used in BBB modeling, depending on their sources, is meticulously examined in this comprehensive review, scrutinizing their respective derivation protocols and implications. By synthesizing diverse approaches, this review sheds light on the improvements of BBB models to capture physiological conditions, aiding in understanding BBB interactions in health and disease conditions to foster clinical developments.
Collapse
Affiliation(s)
- Yomna Soliman
- Faculty of Engineering and Natural Sciences, Sabancı University, Istanbul, Turkey
- Faculty of Pharmacy, Mansoura University, Egypt
| | - Jana Al-Khodor
- Faculty of Engineering and Natural Sciences, Sabancı University, Istanbul, Turkey
| | | | - Nur Mustafaoglu
- Faculty of Engineering and Natural Sciences, Sabancı University, Istanbul, Turkey
- Sabancı University Nanotechnology Research and Application Center, Istanbul, Turkey
| |
Collapse
|
2
|
Kim J, Yoon T, Lee S, Kim PJ, Kim Y. Reconstitution of human tissue barrier function for precision and personalized medicine. LAB ON A CHIP 2024; 24:3347-3366. [PMID: 38895863 DOI: 10.1039/d4lc00104d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/21/2024]
Abstract
Tissue barriers in a body, well known as tissue-to-tissue interfaces represented by endothelium of the blood vessels or epithelium of organs, are essential for maintaining physiological homeostasis by regulating molecular and cellular transports. It is crucial for predicting drug response to understand physiology of tissue barriers through which drugs are absorbed, distributed, metabolized and excreted. Since the FDA Modernization Act 2.0, which prompts the inception of alternative technologies for animal models, tissue barrier chips, one of the applications of organ-on-a-chip or microphysiological system (MPS), have only recently been utilized in the context of drug development. Recent advancements in stem cell technology have brightened the prospects for the application of tissue barrier chips in personalized medicine. In past decade, designing and engineering these microfluidic devices, and demonstrating the ability to reconstitute tissue functions were main focus of this field. However, the field is now advancing to the next level of challenges: validating their utility in drug evaluation and creating personalized models using patient-derived cells. In this review, we briefly introduce key design parameters to develop functional tissue barrier chip, explore the remarkable recent progress in the field of tissue barrier chips and discuss future perspectives on realizing personalized medicine through the utilization of tissue barrier chips.
Collapse
Affiliation(s)
- Jaehoon Kim
- George W. Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, GA 30332, USA.
| | - Taehee Yoon
- George W. Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, GA 30332, USA.
- Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA 30332, USA
| | - Sungryeong Lee
- George W. Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, GA 30332, USA.
| | - Paul J Kim
- Department of Psychiatry & Behavioral Sciences, School of Medicine, Emory University, Atlanta, GA, 30322, USA
| | - YongTae Kim
- George W. Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, GA 30332, USA.
- Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA 30332, USA
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA 30332, USA
- Institute for Electronics and Nanotechnology, Georgia Institute of Technology, Atlanta, GA 30332, USA
| |
Collapse
|
3
|
Oezen G, Kraus L, Schentarra EM, Bolten JS, Huwyler J, Fricker G. Aluminum and ABC transporter activity. ENVIRONMENTAL TOXICOLOGY AND PHARMACOLOGY 2024; 108:104451. [PMID: 38648870 DOI: 10.1016/j.etap.2024.104451] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/12/2024] [Revised: 04/06/2024] [Accepted: 04/16/2024] [Indexed: 04/25/2024]
Abstract
Aluminum is the third most common element on Earth´s crust and despite its wide use in our workaday life it has been associated with several health risks after overexposure. In the present study the impact of aluminum salts upon ABC transporter activity was studied in the P-GP-expressing human blood-brain barrier cell line hCMEC/D3, in MDCKII cells overexpressing BCRP and MRP2, respectively, and in freshly isolated, functionally intact kidney tubules from Atlantic killifish (Fundulus heteroclitus), which express the analog ABC transporters, P-gp, Bcrp and Mrp2. In contrast to previous findings with heavy metals salts (cadmium(II) chloride or mercury(II) chloride), which have a strong inhibitory effect on ABC transporter activity, or zinc(II) chloride and sodium arsenite, which have a stimulatory effect upon ABC transport function, the results indicate no modulatory effect of aluminum salts on the efflux activity of the human ABC transporters P-GP, BCRP and MRP2 nor on the analog transporters P-gp, Bcrp and Mrp2.
Collapse
Affiliation(s)
- Goezde Oezen
- Institute of Pharmacy and Molecular Biotechnology, Ruprecht-Karls University, Heidelberg 69120, Germany; Mount Desert Island Biological Laboratory, Salisbury Cove, ME 04672, United States
| | - Lisa Kraus
- Institute of Pharmacy and Molecular Biotechnology, Ruprecht-Karls University, Heidelberg 69120, Germany; Mount Desert Island Biological Laboratory, Salisbury Cove, ME 04672, United States
| | - Eva-Maria Schentarra
- Institute of Pharmacy and Molecular Biotechnology, Ruprecht-Karls University, Heidelberg 69120, Germany; Mount Desert Island Biological Laboratory, Salisbury Cove, ME 04672, United States
| | - Jan Stephan Bolten
- Mount Desert Island Biological Laboratory, Salisbury Cove, ME 04672, United States; Department of Pharmaceutical Sciences, University of Basel, Basel 4056, Switzerland
| | - Joerg Huwyler
- Mount Desert Island Biological Laboratory, Salisbury Cove, ME 04672, United States; Department of Pharmaceutical Sciences, University of Basel, Basel 4056, Switzerland
| | - Gert Fricker
- Institute of Pharmacy and Molecular Biotechnology, Ruprecht-Karls University, Heidelberg 69120, Germany; Mount Desert Island Biological Laboratory, Salisbury Cove, ME 04672, United States.
| |
Collapse
|
4
|
Ho YS, Torres-Vergara P, Penny J. Regulation of the ATP-binding cassette transporters ABCB1, ABCG2 and ABCC5 by nuclear receptors in porcine blood-brain barrier endothelial cells. Br J Pharmacol 2023; 180:3092-3109. [PMID: 37476954 DOI: 10.1111/bph.16196] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Revised: 05/26/2023] [Accepted: 06/22/2023] [Indexed: 07/22/2023] Open
Abstract
BACKGROUND AND PURPOSE Blood-brain barrier (BBB) ABCB1, ABCG2 and ABCC5 transporters influence central therapeutic drug distribution. Transporter expression is regulated by the NR3C1, NR1I3 and NR1I2 nuclear receptors, but their precise roles in brain are poorly understood. We investigated the effects of selective ligand-based activation of NR3C1, NR1I3, NR1I2 and NR2B1 in porcine brain endothelial cells (PBECs). EXPERIMENTAL APPROACH Primary cultures of PBECs were exposed to NR3C1, NR1I3 and NR1I2 ligands and ABCB1, ABCG2 and ABCC5 transporter activities determined by measuring intracellular accumulation of fluorescent probes. Western blotting was used to determine the effects of receptor ligands on expression of ABCB1, ABCG2, ABCC5, NR1I2, NR1I3, NR3C1 and NR2B1. Fluorescent immunocytochemistry was employed to assess the effects of receptor ligands on the cellular localisation of NR1I2 and NR1I3. KEY RESULTS The NR1I2 agonist rifampicin significantly up-regulated ABCG2 activity, which is counteracted by co-treatment with NR1I2 antagonist l-sulforaphane. The NR1I3 agonist 6-(4-chlorophenyl)-imidazo[2,1-b]thiazole-5-carbaldehyde and inverse agonist meclizine significantly down-regulated ABCB1, ABCG2 and ABCC5 activity. NR3C1 agonist dexamethasone significantly increased ABCB1, ABCG2 and ABCC5 activity and ABCG2 and ABCC5 protein expression, which was counteracted by co-treatment with the NR3C1 antagonist mifepristone. This first study demonstrates that NR1I3 and NR3C1 regulate ABCC5 activity and protein expression in BBB endothelial cells. CONCLUSIONS AND IMPLICATIONS In PBECs, expression of key ATP-binding cassette (ABC) transporters and nuclear receptors is differentially regulated by NR1I3, NR1I2, NR3C1 and NR2B1. This will help to better understand the response of the BBB to physiological and pharmacological activation of nuclear receptors.
Collapse
Affiliation(s)
- Yu Siong Ho
- Division of Pharmacy and Optometry, School of Health Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, UK
| | - Pablo Torres-Vergara
- Departamento de Farmacia, Facultad de Farmacia, Universidad de Concepción, Concepción, Chile
| | - Jeffrey Penny
- Division of Pharmacy and Optometry, School of Health Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, UK
| |
Collapse
|
5
|
Wupperfeld D, Fricker G, Bois De Fer B, Frank L, Wehrle A, Popovic B. Essential phospholipids decrease apoptosis and increase membrane transport in human hepatocyte cell lines. Lipids Health Dis 2022; 21:91. [PMID: 36153592 PMCID: PMC9508738 DOI: 10.1186/s12944-022-01698-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Accepted: 09/03/2022] [Indexed: 11/10/2022] Open
Abstract
Abstract
Background
Essential phospholipids (EPL) have hepatoprotective effects across many liver diseases/conditions. The impact of EPL on hepatocyte function in vitro was investigated.
Methods
Effects of noncytotoxic concentrations of EPL (0.1 and 0.25 mg/ml), and its constituents, polyenylphosphatidylcholine (PPC) and phosphatidylinositol (PI) (both at 0.1 and 1 mg/ml), on membrane fluidity, apoptosis and extracellular transport versus controls were investigated in human hepatocyte cell lines (HepG2, HepaRG, steatotic HepaRG).
Results
Significantly increased membrane fluidity occurred with all 3 phospholipids (PLs) in HepG2 cultures, and with PI (1 mg/ml) in steatotic HepaRG cells. Significantly decreased tamoxifen-induced apoptosis was observed in HepG2 cells with EPL, PPC and PI. Breast cancer resistance protein (BCRP) activity was significantly increased by EPL and PI in HepG2 cells. Multidrug resistance-associated protein 2 (MRP-2) activity was unaffected by any PL in HepG2 cells, and significantly increased by EPL, PI and PPC (1 mg/ml) in HepaRG cells, and by PI (1 mg/ml) in steatotic HepaRG cells. Bile salt export protein (BSEP) activity in HepG2 cells and steatotic HepaRG cells was significantly increased by EPL (0.25 mg/ml), and PPC (both concentrations), but not by PI. The PLs had no effects on HepaRG cell BSEP activity. P-glycoprotein (P-GP) activity was significantly increased by all compounds in HepG2 cells. PI (1 mg/ml) significantly increased P-GP activity in HepaRG and steatotic HepaRG cells.
Conclusions
EPL, PPC, and PI increased hepatocyte membrane fluidity, decreased apoptosis and increased hepatocellular export, all of which may improve liver function. These in-vitro investigations provide valuable insights into the mechanism of action of EPL.
Collapse
|
6
|
PCSK9 acts as a key regulator of Aβ clearance across the blood-brain barrier. Cell Mol Life Sci 2022; 79:212. [PMID: 35344086 PMCID: PMC8960591 DOI: 10.1007/s00018-022-04237-x] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Revised: 02/25/2022] [Accepted: 03/08/2022] [Indexed: 12/18/2022]
Abstract
Despite the neurodegenerative disorder Alzheimer's disease (AD) is the most common form of dementia in late adult life, there is currently no therapy available to prevent the onset or slow down the progression of AD. The progressive cognitive decline in AD correlates with a successive accumulation of cerebral amyloid-β (Aβ) due to impaired clearance mechanisms. A significant percentage is removed by low-density lipoprotein receptor-related protein 1 (LRP1)-mediated transport across the blood-brain barrier (BBB) into the periphery. Circulating proprotein convertase subtilisin/kexin type 9 (PCSK9) binds to members of the low-density lipoprotein receptor protein family at the cell surface and targets them for lysosomal degradation, which reduces the number of functional receptors. However, the adverse impact of PCSK9 on LRP1-mediated brain Aβ clearance remains elusive. By using an established BBB model, we identified reduced LRP1-mediated brain-to-blood Aβ clearance due to PCSK9 across different endothelial monolayer in vitro. Consequently, the repetitive application of FDA-approved monoclonal anti-PCSK9 antibodies into 5xFAD mice decreased the cerebral Aβ burden across variants and aggregation state, which was not reproducible in brain endothelial-specific LRP1-/- 5xFAD mice. The peripheral PCSK9 inhibition reduced Aβ pathology in prefrontal cortex and hippocampus-brain areas critically involved in memory processing-and prevented disease-related impairment in hippocampus-dependent memory formation. Our data suggest that peripheral inhibition of PCSK9 by already available therapeutic antibodies may be a novel and easily applicable potential AD treatment.
Collapse
|
7
|
Szerémy P, Tauberné Jakab K, Baráth S, Apjok A, Filkor K, Holló Z, Márki‐Zay J, Kappelmayer J, Sipka S, Krajcsi P, Toldi G. Determination of Reference Values of MDR‐ABC Transporter Activities in CD3+ Lymphocytes of Healthy Volunteers Using a Flow Cytometry Based Method. CYTOMETRY PART B-CLINICAL CYTOMETRY 2018; 96:469-474. [DOI: 10.1002/cyto.b.21729] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/18/2018] [Revised: 07/30/2018] [Accepted: 08/01/2018] [Indexed: 12/11/2022]
Affiliation(s)
- Péter Szerémy
- MDQuest Ltd Szeged Hungary
- SOLVO Biotechnology Budaörs Hungary
| | | | - Sándor Baráth
- Department of Laboratory MedicineUniversity of Debrecen Debrecen Hungary
| | - András Apjok
- MDQuest Ltd Szeged Hungary
- SOLVO Biotechnology Budaörs Hungary
| | | | | | | | - János Kappelmayer
- Department of Laboratory MedicineUniversity of Debrecen Debrecen Hungary
| | - Sándor Sipka
- 3rd Department of Internal MedicineUniversity of Debrecen Debrecen Hungary
| | | | | |
Collapse
|
8
|
Storck SE, Hartz AM, Bernard J, Wolf A, Kachlmeier A, Mahringer A, Weggen S, Pahnke J, Pietrzik CU. The concerted amyloid-beta clearance of LRP1 and ABCB1/P-gp across the blood-brain barrier is linked by PICALM. Brain Behav Immun 2018; 73:21-33. [PMID: 30041013 PMCID: PMC7748946 DOI: 10.1016/j.bbi.2018.07.017] [Citation(s) in RCA: 81] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2018] [Revised: 07/10/2018] [Accepted: 07/20/2018] [Indexed: 01/18/2023] Open
Abstract
The accumulation of neurotoxic amyloid-beta (Aβ) in the brain is a characteristic hallmark of Alzheimer's disease (AD). The blood-brain barrier (BBB) provides a large surface area and has been shown to be an important mediator for removal of brain Aβ. Both, the ABC transporter P-glycoprotein (ABCB1/P-gp) and the receptor low-density lipoprotein receptor-related protein 1 (LRP1) have been implicated to play crucial roles in Aβ efflux from brain. Here, with immunoprecipitation experiments, co-immunostainings and dual inhibition of ABCB1/P-gp and LRP1, we show that both proteins are functionally linked, mediating a concerted transcytosis of Aβ through endothelial cells. Late-onset AD risk factor Phosphatidylinositol binding clathrin assembly protein (PICALM) is associated with both ABCB1/P-gp and LRP1 representing a functional link and guiding both proteins through the brain endothelium. Together, our results give more mechanistic insight on Aβ transport across the BBB and show that the functional interplay of different clearance proteins is needed for the rapid removal of Aβ from the brain.
Collapse
Affiliation(s)
- Steffen E. Storck
- Institute for Pathobiochemistry, University Medical Center of the Johannes Gutenberg-University, Mainz, Germany
| | - Anika M.S. Hartz
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY, United States,Department of Pharmacology and Nutritional Sciences, College of Medicine, University of Kentucky, Lexington, KY, United States
| | - Jessica Bernard
- Institute for Pathobiochemistry, University Medical Center of the Johannes Gutenberg-University, Mainz, Germany
| | - Andrea Wolf
- Department of Pharmacy Practice and Pharmaceutical Sciences, College of Pharmacy, University of Minnesota, Duluth, MN, United States
| | - André Kachlmeier
- Institute for Pathobiochemistry, University Medical Center of the Johannes Gutenberg-University, Mainz, Germany
| | - Anne Mahringer
- Institute of Pharmacy and Molecular Biotechnology, University of Heidelberg, Germany
| | - Sascha Weggen
- Department of Neuropathology, Heinrich Heine University Duesseldorf, Duesseldorf, Germany
| | - Jens Pahnke
- University of Oslo (UiO) & Oslo University Hospital (OUS), Department of Neuro-/Pathology, Oslo, Norway,University of Lübeck (UzL), LIED, Lübeck, Germany,Leibniz-Institute of Plant Biochemistry (IPB), Department for Bioorganic Chemistry, Halle, Germany,University of Latvia (UL), Department of Pharmacology, Riga, Latvia
| | - Claus U. Pietrzik
- Institute for Pathobiochemistry, University Medical Center of the Johannes Gutenberg-University, Mainz, Germany,Corresponding author at: University Medical Center of the Johannes Gutenberg-University of Mainz, Institute for Pathobiochemistry, Molecular Neurodegeneration, Duesbergweg 6, 55099 Mainz, Germany. (C.U. Pietrzik)
| |
Collapse
|
9
|
Yasuda K, Ganguly S, Schuetz EG. Pheophorbide A: Fluorescent Bcrp Substrate to Measure Oral Drug-Drug Interactions in Real-Time In Vivo. Drug Metab Dispos 2018; 46:1725-1733. [PMID: 30111622 DOI: 10.1124/dmd.118.083584] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2018] [Accepted: 08/13/2018] [Indexed: 01/19/2023] Open
Abstract
We investigated whether pheophorbide A (PhA) could serve as a selective breast cancer resistance protein (BCRP) substrate (victim) to screen in vivo using fluorescent live animal imaging for transporter-mediated interactions with orally administered inhibitors (perpetrators), and whether that could be coupled with serum sampling to measure the systemic concentration of PhA with a fast-throughput in vitro fluorescent assay. PhA is a breakdown product of chlorophyll and is highly fluorescent in the near-infrared (NIR) spectrum. Whole-body NIR fluorescence was greater in the Bcrp KO compared with wild-type (WT) mice fed a regular diet containing chlorophyll and PhA, with fluorescence in WT mice confined to the intestine. PhA intestinal enterocyte fluorescence, after removing lumen contents, was greater in Bcrp knockout (KO) mice versus WT mice due to PhA enterocyte absorption and lack of PhA efflux by Bcrp. This difference was eliminated by maintaining the mice on an alfalfa (chlorophyll/PhA)-free diet. The area under the fluorescence ratio-time curve up to 6 hours (AUCFL 0-6 h) of orally administrated PhA was 3.5 times greater in the Bcrp KO mice compared with WT mice, and the PhA serum concentration was 50-fold higher in KO mice. Pretreatment with known BCRP inhibitors lapatinib, curcumin, elacridar, pantoprazole, and sorafenib, at clinically relevant doses, significantly increased PhA AUCFL 0-6 h by 2.4-, 2.3-, 2.2-, 1.5-, and 1.4-fold, respectively, whereas the area under PhA serum concentration-time curve calculated up to 6 hours (AUCSerum 0-6 h) increased by 13.8-, 7.8-, 5.2-, 2.02-, and 1.45-fold, respectively, and corresponded to their hierarchy as in vitro BCRP inhibitors. Our results demonstrate that live animal imaging using PhA can be used to identify BCRP inhibitors and to assess the potential for BCRP-mediated clinical drug-drug interactions.
Collapse
Affiliation(s)
- Kazuto Yasuda
- Department of Pharmaceutical Sciences, St. Jude Children's Research Hospital, Memphis, Tennessee (K.Y., S.G., E.G.S.); and Cancer and Developmental Biology Track, University of Tennessee Health Science Center, Memphis, Tennessee (S.G.)
| | - Samit Ganguly
- Department of Pharmaceutical Sciences, St. Jude Children's Research Hospital, Memphis, Tennessee (K.Y., S.G., E.G.S.); and Cancer and Developmental Biology Track, University of Tennessee Health Science Center, Memphis, Tennessee (S.G.)
| | - Erin G Schuetz
- Department of Pharmaceutical Sciences, St. Jude Children's Research Hospital, Memphis, Tennessee (K.Y., S.G., E.G.S.); and Cancer and Developmental Biology Track, University of Tennessee Health Science Center, Memphis, Tennessee (S.G.)
| |
Collapse
|
10
|
Effect of amyloid beta on ATP-binding cassette transporter expression and activity in porcine brain microvascular endothelial cells. Biochim Biophys Acta Gen Subj 2018; 1862:2314-2322. [PMID: 30048740 DOI: 10.1016/j.bbagen.2018.07.021] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2018] [Revised: 07/19/2018] [Accepted: 07/20/2018] [Indexed: 11/24/2022]
Abstract
BACKGROUND Deposition of amyloid-β peptide (Aβ(1-42)) within the brain is characteristic of Alzheimer's disease. Little is known of the effects of Aβ(1-42) on blood-brain barrier (BBB) ATP-binding Cassette (ABC) efflux transporters which influence BBB permeability. The effects of Aβ(1-42) on ABCB1, ABCC5 and ABCG2 activity and expression and pregnane X receptor (PXR) and constitutive androstane receptor (CAR) transcription factors expression were determined in primary porcine brain endothelial cells (PBECs). METHODS The effect of Aβ(1-42) on transporter activity was determined by measurement of intracellular accumulation of the fluorescent probes calcein (ABCB1), GS-MF (ABCC5) and Hoechst 33342 (ABCG2). Expression of transporters and transcription factors was assessed by Western blotting. RESULTS Treatment of PBECs with Aβ(1-42) significantly decreased activity of ABCB1 (Aβ(1-42) at 10 μg/ml, 25 μg/ml and 50 μg/ml), ABCC5 (Aβ(1-42) at 25 μg/ml and 50 μg/ml) and ABCG2 (Aβ(1-42) at 10 μg/ml, 25 μg/ml and 50 μg/ml). Aβ(1-42) also significantly decreased expression of ABCB1 (p < 0.05 at 25 μg/ml and 50 μg/ml), ABCG2 (p < 0.05 at 25 μg/ml and p ≤ 0.001 at 50 μg/ml), ABCC5 (p < 0.05 at 25 μg/ml and 50 μg/ml), PXR (p < 0.05 at 10 μg/ml, 25 μg/ml and 50 μg/ml Aβ(1-42)) and CAR (p < 0.05 at 25 μg/ml and 50 μg/ml Aβ(1-42)). CONCLUSION Aβ(1-42) inhibits multiple ABC transporters and PXR and CAR in PBECs. GENERAL SIGNIFICANCE Aβ(1-42) reduces ABC transporter activity and expression in BBB endothelial cells and has the potential to influence BBB permeability characteristics.
Collapse
|
11
|
Neuhaus W, Piontek A, Protze J, Eichner M, Mahringer A, Subileau EA, Lee IFM, Schulzke JD, Krause G, Piontek J. Reversible opening of the blood-brain barrier by claudin-5-binding variants of Clostridium perfringens enterotoxin's claudin-binding domain. Biomaterials 2018; 161:129-143. [DOI: 10.1016/j.biomaterials.2018.01.028] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2017] [Revised: 01/15/2018] [Accepted: 01/18/2018] [Indexed: 02/06/2023]
|
12
|
Zinc chloride rapidly stimulates efflux transporters in renal proximal tubules of killifish ( Fundulus heteroclitus ). Toxicol Appl Pharmacol 2017; 334:88-99. [DOI: 10.1016/j.taap.2017.09.001] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2017] [Revised: 08/04/2017] [Accepted: 09/01/2017] [Indexed: 12/26/2022]
|
13
|
Nielsen SSE, Siupka P, Georgian A, Preston JE, Tóth AE, Yusof SR, Abbott NJ, Nielsen MS. Improved Method for the Establishment of an In Vitro Blood-Brain Barrier Model Based on Porcine Brain Endothelial Cells. J Vis Exp 2017. [PMID: 28994773 DOI: 10.3791/56277] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
The aim of this protocol presents an optimized procedure for the purification and cultivation of pBECs and to establish in vitro blood-brain barrier (BBB) models based on pBECs in mono-culture (MC), MC with astrocyte-conditioned medium (ACM), and non-contact co-culture (NCC) with astrocytes of porcine or rat origin. pBECs were isolated and cultured from fragments of capillaries from the brain cortices of domestic pigs 5-6 months old. These fragments were purified by careful removal of meninges, isolation and homogenization of grey matter, filtration, enzymatic digestion, and centrifugation. To further eliminate contaminating cells, the capillary fragments were cultured with puromycin-containing medium. When 60-95% confluent, pBECs growing from the capillary fragments were passaged to permeable membrane filter inserts and established in the models. To increase barrier tightness and BBB characteristic phenotype of pBECs, the cells were treated with the following differentiation factors: membrane permeant 8-CPT-cAMP (here abbreviated cAMP), hydrocortisone, and a phosphodiesterase inhibitor, RO-20-1724 (RO). The procedure was carried out over a period of 9-11 days, and when establishing the NCC model, the astrocytes were cultured 2-8 weeks in advance. Adherence to the described procedures in the protocol has allowed the establishment of endothelial layers with highly restricted paracellular permeability, with the NCC model showing an average transendothelial electrical resistance (TEER) of 1249 ± 80 Ω cm2, and paracellular permeability (Papp) for Lucifer Yellow of 0.90 10-6 ± 0.13 10-6 cm sec-1 (mean ± SEM, n=55). Further evaluation of this pBEC phenotype showed good expression of the tight junctional proteins claudin 5, ZO-1, occludin and adherens junction protein p120 catenin. The model presented can be used for a range of studies of the BBB in health and disease and, with the highly restrictive paracellular permeability, this model is suitable for studies of transport and intracellular trafficking.
Collapse
Affiliation(s)
- Simone S E Nielsen
- Lundbeck Foundation Research Initiative on Brain Barriers and Drug Delivery, Department of Biomedicine, Aarhus University
| | - Piotr Siupka
- Lundbeck Foundation Research Initiative on Brain Barriers and Drug Delivery, Department of Biomedicine, Aarhus University
| | - Ana Georgian
- Institute of Pharmaceutical Science, King's College London
| | - Jane E Preston
- Institute of Pharmaceutical Science, King's College London
| | - Andrea E Tóth
- Lundbeck Foundation Research Initiative on Brain Barriers and Drug Delivery, Department of Biomedicine, Aarhus University
| | - Siti R Yusof
- Institute of Pharmaceutical Science, King's College London; HICoE Centre for Drug Research, Universiti Sains Malaysia
| | - N Joan Abbott
- Institute of Pharmaceutical Science, King's College London;
| | - Morten S Nielsen
- Lundbeck Foundation Research Initiative on Brain Barriers and Drug Delivery, Department of Biomedicine, Aarhus University;
| |
Collapse
|
14
|
Muthiah D, Callaghan R. Dual effects of the PI3K inhibitor ZSTK474 on multidrug efflux pumps in resistant cancer cells. Eur J Pharmacol 2017; 815:127-137. [PMID: 28912036 DOI: 10.1016/j.ejphar.2017.09.001] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2017] [Revised: 08/30/2017] [Accepted: 09/01/2017] [Indexed: 12/13/2022]
Abstract
ZSTK474 is a potent phosphoinositide 3-kinase (PI3K) inhibitor that reduces cell proliferation via G1-arrest. However, there is little information on the susceptibility of this anticancer drug to resistance conferred by the multidrug pumps P-glycoprotein (ABCB1) and ABCG2. We have demonstrated that ZSTK474 generated cytotoxicity in cells over-expressing either pump with potency similar to that in drug sensitive cells. In addition, the co-administration of ZSTK474 with the cytotoxic anti-cancer drugs vinblastine and mitoxantrone caused a potentiated cytotoxic effect in both drug sensitive and efflux pump expressing cells. These observations suggest that ZSTK474 is unaffected by the presence of multidrug efflux pumps and may circumvent their activities. Indeed, ZSTK474 increased the cellular accumulation of calcein-AM and mitoxantrone in cells expressing ABCB1 and ABCG2, respectively. ZSTK474 treatment also resulted in reduced expression of both efflux pumps in multidrug resistant cancer cells. Measurement of ABCB1 or ABCG2 mRNA levels demonstrated that the reduction was not due to altered transcription. Similarly, inhibitor studies showed that the proteasomal degradation pathway for ABCB1 and the lysosomal route for ABCG2 degradation were unaffected by ZSTK474. Thus the mechanism underlying reduced ABCB1 and ABCG2 levels caused by ZSTK474 was due to a reduction in overall protein synthesis; a process influenced by the PI3K pathway. In summary, ZSTK474 is not susceptible to efflux by the resistance mediators ABCB1 and ABCG2. Moreover, it inhibits the drug transport function of the pumps and leads to a reduction in their cellular expression levels. Our observations demonstrate that ZSTK474 is a powerful anticancer drug.
Collapse
Affiliation(s)
- Divya Muthiah
- Division of Biomedical Science & Biochemistry, Research School of Biology and Medical School, The Australian National University, Building 134, Linnaeus Way, Canberra, ACT 2601, Australia
| | - Richard Callaghan
- Division of Biomedical Science & Biochemistry, Research School of Biology and Medical School, The Australian National University, Building 134, Linnaeus Way, Canberra, ACT 2601, Australia.
| |
Collapse
|
15
|
Bernard-Gauthier V, Bailey JJ, Mossine AV, Lindner S, Vomacka L, Aliaga A, Shao X, Quesada CA, Sherman P, Mahringer A, Kostikov A, Grand’Maison M, Rosa-Neto P, Soucy JP, Thiel A, Kaplan DR, Fricker G, Wängler B, Bartenstein P, Schirrmacher R, Scott PJH. A Kinome-Wide Selective Radiolabeled TrkB/C Inhibitor for in Vitro and in Vivo Neuroimaging: Synthesis, Preclinical Evaluation, and First-in-Human. J Med Chem 2017; 60:6897-6910. [DOI: 10.1021/acs.jmedchem.7b00396] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Affiliation(s)
- Vadim Bernard-Gauthier
- Department of Oncology,
Division of Oncological Imaging, University of Alberta, Edmonton, Alberta T6G 2R3, Canada
| | - Justin J. Bailey
- Department of Oncology,
Division of Oncological Imaging, University of Alberta, Edmonton, Alberta T6G 2R3, Canada
| | - Andrew V. Mossine
- Division of Nuclear Medicine, Department
of Radiology, The University of Michigan Medical School, Ann Arbor, Michigan 48109, United States
| | - Simon Lindner
- Department of Nuclear Medicine, Ludwig-Maximilians-University of Munich, Marchioninistrasse 15, Munich 81377, Germany
| | - Lena Vomacka
- Department of Nuclear Medicine, Ludwig-Maximilians-University of Munich, Marchioninistrasse 15, Munich 81377, Germany
| | - Arturo Aliaga
- Translational Neuroimaging Laboratory, McGill Centre
for Studies in Aging, Douglas Mental Health University Institute, 6875 Boulevard LaSalle, Montreal, Quebec H4H 1R3, Canada
| | - Xia Shao
- Division of Nuclear Medicine, Department
of Radiology, The University of Michigan Medical School, Ann Arbor, Michigan 48109, United States
| | - Carole A. Quesada
- Division of Nuclear Medicine, Department
of Radiology, The University of Michigan Medical School, Ann Arbor, Michigan 48109, United States
| | - Phillip Sherman
- Division of Nuclear Medicine, Department
of Radiology, The University of Michigan Medical School, Ann Arbor, Michigan 48109, United States
| | - Anne Mahringer
- Institute of Pharmacy and Molecular Biotechnology, University of Heidelberg, Heidelberg 69120, Germany
| | - Alexey Kostikov
- McConnell
Brain Imaging Centre, Montreal Neurological Institute, McGill University, 3801 University Street, Montreal, Quebec H3A 2B4, Canada
| | | | - Pedro Rosa-Neto
- Translational Neuroimaging Laboratory, McGill Centre
for Studies in Aging, Douglas Mental Health University Institute, 6875 Boulevard LaSalle, Montreal, Quebec H4H 1R3, Canada
| | - Jean-Paul Soucy
- Department of Neurology and Neurosurgery, Montreal Neurological Institute, McGill University, 3801 University Street, Montreal, Quebec H3A 2B4, Canada
| | - Alexander Thiel
- McConnell
Brain Imaging Centre, Montreal Neurological Institute, McGill University, 3801 University Street, Montreal, Quebec H3A 2B4, Canada
- Jewish General Hospital, Lady Davis Institute, Montreal, Quebec HT3 1E2, Canada
| | - David R. Kaplan
- Program in Neurosciences and Mental Health, Hospital for Sick Children, Toronto, Ontario M5G 0A4, Canada
- Department of Molecular
Genetics, University of Toronto, Toronto, Ontario M5S1A8, Canada
| | - Gert Fricker
- Institute of Pharmacy and Molecular Biotechnology, University of Heidelberg, Heidelberg 69120, Germany
| | - Björn Wängler
- Molecular
Imaging and Radiochemistry, Department of Clinical Radiology and Nuclear
Medicine, Medical Faculty Mannheim of Heidelberg University, Theodor-Kutzer-Ufer
1-3, Mannheim 68167, Germany
| | - Peter Bartenstein
- Department of Nuclear Medicine, Ludwig-Maximilians-University of Munich, Marchioninistrasse 15, Munich 81377, Germany
| | - Ralf Schirrmacher
- Department of Oncology,
Division of Oncological Imaging, University of Alberta, Edmonton, Alberta T6G 2R3, Canada
| | - Peter J. H. Scott
- Division of Nuclear Medicine, Department
of Radiology, The University of Michigan Medical School, Ann Arbor, Michigan 48109, United States
- The Interdepartmental Program in Medicinal
Chemistry, University of Michigan, Ann Arbor, Michigan 48109, United States
| |
Collapse
|
16
|
Approaches for the discovery of novel positron emission tomography radiotracers for brain imaging. Clin Transl Imaging 2017. [DOI: 10.1007/s40336-017-0221-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
|
17
|
Helms HC, Abbott NJ, Burek M, Cecchelli R, Couraud PO, Deli MA, Förster C, Galla HJ, Romero IA, Shusta EV, Stebbins MJ, Vandenhaute E, Weksler B, Brodin B. In vitro models of the blood-brain barrier: An overview of commonly used brain endothelial cell culture models and guidelines for their use. J Cereb Blood Flow Metab 2016; 36:862-90. [PMID: 26868179 PMCID: PMC4853841 DOI: 10.1177/0271678x16630991] [Citation(s) in RCA: 527] [Impact Index Per Article: 65.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2015] [Accepted: 01/05/2016] [Indexed: 12/12/2022]
Abstract
The endothelial cells lining the brain capillaries separate the blood from the brain parenchyma. The endothelial monolayer of the brain capillaries serves both as a crucial interface for exchange of nutrients, gases, and metabolites between blood and brain, and as a barrier for neurotoxic components of plasma and xenobiotics. This "blood-brain barrier" function is a major hindrance for drug uptake into the brain parenchyma. Cell culture models, based on either primary cells or immortalized brain endothelial cell lines, have been developed, in order to facilitate in vitro studies of drug transport to the brain and studies of endothelial cell biology and pathophysiology. In this review, we aim to give an overview of established in vitro blood-brain barrier models with a focus on their validation regarding a set of well-established blood-brain barrier characteristics. As an ideal cell culture model of the blood-brain barrier is yet to be developed, we also aim to give an overview of the advantages and drawbacks of the different models described.
Collapse
Affiliation(s)
- Hans C Helms
- Department of Pharmacy, University of Copenhagen, Denmark
| | - N Joan Abbott
- Institute of Pharmaceutical Science, King's College London, UK
| | - Malgorzata Burek
- Klinik und Poliklinik für Anästhesiologie, University of Wurzburg, Germany
| | | | - Pierre-Olivier Couraud
- Institut Cochin, INSERM U1016, CNRS UMR8104, Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Maria A Deli
- Institute of Biophysics, Biological Research Centre, HAS, Szeged, Hungary
| | - Carola Förster
- Klinik und Poliklinik für Anästhesiologie, University of Wurzburg, Germany
| | - Hans J Galla
- Institute of Biochemistry, University of Muenster, Germany
| | - Ignacio A Romero
- Department of Biological Sciences, The Open University, Walton Hall, Milton Keynes, UK
| | - Eric V Shusta
- Department of Chemical and Biological Engineering, University of Wisconsin-Madison, WI, USA
| | - Matthew J Stebbins
- Department of Chemical and Biological Engineering, University of Wisconsin-Madison, WI, USA
| | | | - Babette Weksler
- Division of Hematology and Medical Oncology, Weill Cornell Medical College, NY, USA
| | - Birger Brodin
- Department of Pharmacy, University of Copenhagen, Denmark
| |
Collapse
|
18
|
Caetano-Pinto P, Janssen MJ, Gijzen L, Verscheijden L, Wilmer MJ, Masereeuw R. Fluorescence-Based Transport Assays Revisited in a Human Renal Proximal Tubule Cell Line. Mol Pharm 2016; 13:933-44. [DOI: 10.1021/acs.molpharmaceut.5b00821] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Affiliation(s)
- Pedro Caetano-Pinto
- Department
of Pharmacology and Toxicology, Radboud university medical center, Radboud Institute for Molecular Life Sciences, 6500 HB Nijmegen, The Netherlands
- Division
of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, 3508 TB Utrecht, The Netherlands
| | - Manoe J. Janssen
- Department
of Pharmacology and Toxicology, Radboud university medical center, Radboud Institute for Molecular Life Sciences, 6500 HB Nijmegen, The Netherlands
- Division
of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, 3508 TB Utrecht, The Netherlands
| | - Linda Gijzen
- Department
of Pharmacology and Toxicology, Radboud university medical center, Radboud Institute for Molecular Life Sciences, 6500 HB Nijmegen, The Netherlands
| | - Laurens Verscheijden
- Department
of Pharmacology and Toxicology, Radboud university medical center, Radboud Institute for Molecular Life Sciences, 6500 HB Nijmegen, The Netherlands
| | - Martijn J.G. Wilmer
- Department
of Pharmacology and Toxicology, Radboud university medical center, Radboud Institute for Molecular Life Sciences, 6500 HB Nijmegen, The Netherlands
| | - Rosalinde Masereeuw
- Department
of Pharmacology and Toxicology, Radboud university medical center, Radboud Institute for Molecular Life Sciences, 6500 HB Nijmegen, The Netherlands
- Division
of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, 3508 TB Utrecht, The Netherlands
| |
Collapse
|
19
|
Brain delivery of camptothecin by means of solid lipid nanoparticles: formulation design, in vitro and in vivo studies. Int J Pharm 2012; 439:49-62. [PMID: 23046667 DOI: 10.1016/j.ijpharm.2012.09.054] [Citation(s) in RCA: 76] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2012] [Revised: 09/26/2012] [Accepted: 09/29/2012] [Indexed: 02/05/2023]
Abstract
For the purpose of brain delivery upon intravenous injection, formulations of camptothecin-loaded solid lipid nanoparticles (SLN), prepared by hot high pressure homogenisation, were designed. Incorporation of camptothecin in the hydrophobic and acidic environment of SLN matrix was chosen to stabilise the lactone ring, which is essential for its antitumour activity, and for avoiding premature loss of drug on the way to target camptothecin to the brain. A multivariate approach was used to assess the influence of the qualitative and quantitative composition on the physicochemical properties of camptothecin-loaded SLN in comparison to plain SLN. Mean particle sizes of ≤200 nm, homogenous size distributions and high encapsulation efficiencies (>90%) were achieved for the most suitable formulations. In vitro release studies in plasma, showed a prolonged release profile of camptothecin from SLN, confirming the physical stability of the particles under physiological pH. A higher affinity of the SLN to the porcine brain capillary endothelial cells (BCEC) was shown in comparison to macrophages. MTT studies in BCEC revealed a moderate decrease in the cell viability of camptothecin, when incorporated in SLN compared to free camptothecin in solution. In vivo studies in rats showed that fluorescently labelled SLN were detected in the brain after i.v. administration. This study indicates that the camptothecin-loaded SLN are a promising drug brain delivery system worth to explore further for brain tumour therapy.
Collapse
|
20
|
Cantrill CA, Skinner RA, Rothwell NJ, Penny JI. An immortalised astrocyte cell line maintains the in vivo phenotype of a primary porcine in vitro blood-brain barrier model. Brain Res 2012; 1479:17-30. [PMID: 22940232 DOI: 10.1016/j.brainres.2012.08.031] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2011] [Revised: 08/01/2012] [Accepted: 08/16/2012] [Indexed: 12/11/2022]
Abstract
Whilst it is well documented that all components of the neurovascular unit contribute to the restrictive nature of the blood-brain barrier (BBB), astrocytes have been identified as the cellular component most likely to play an essential role in maintaining the barrier properties. The aim of this study was to examine the impact of the rat astrocyte cell line, CTX-TNA2, on the structural and functional characteristics of an in vitro BBB and determine the capacity of this astrocyte cell line to maintain the BBB phenotype. Co-culture of the CTX-TNA2 cells with primary porcine brain endothelial cells produced an in vitro BBB model which retains key features of the in vivo BBB. High transendothelial electrical resistances, comparable to those reported in vivo, were obtained. Ultrastructural analysis revealed distinct intercellular tight junction protein complexes and immunocytochemistry confirmed expression of the tight junction proteins ZO-1 and occludin. Western blotting and fluorescent tracer assays confirmed expression and functional activity of P-glycoprotein (ABCB1) and breast cancer resistance protein (ABCG2) efflux transporters. Studies employing Alexa-fluor 555-conjugated human transferrin revealed temperature-sensitive internalisation indicating the BBB model retains functional receptor-mediated transferrin uptake. The findings of this study indicate that a robust BBB model has been produced and this is the first report of the inductive capacity of the CTX-TNA2 cell line. Since this in vitro BBB model possesses many key characteristics of the BBB in vivo it has the potential to be a valuable tool for the study of biochemical and physiological processes associated with the BBB.
Collapse
Affiliation(s)
- Carina A Cantrill
- School of Pharmacy and Pharmaceutical Sciences, Stopford Building, University of Manchester, Manchester M13 9PT, UK.
| | | | | | | |
Collapse
|
21
|
Immobilized Ru(bpy)32+ Electrochemiluminescence Coupled with Microdialysis Sampling for On-line Studying Drug-protein Interaction. CHINESE JOURNAL OF ANALYTICAL CHEMISTRY 2012. [DOI: 10.1016/s1872-2040(11)60548-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
|
22
|
Li C, Zhang L, Zhou L, Wo SK, Lin G, Zuo Z. Comparison of intestinal absorption and disposition of structurally similar bioactive flavones in Radix Scutellariae. AAPS JOURNAL 2011; 14:23-34. [PMID: 22167378 DOI: 10.1208/s12248-011-9310-9] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/12/2011] [Accepted: 11/11/2011] [Indexed: 02/02/2023]
Abstract
Radix Scutellariae is a commonly used herbal medicine. Baicalein, wogonin, and oroxylin A are three major bioactive flavones in Radix Scutellariae and share similar chemical structures. The intestinal absorption and disposition of baicalein have been systematically investigated by our group before. In this study, the intestinal absorption and disposition of wogonin and oroxylin A were further explored and compared with the profiles of baicalein to find potential structure-activity relationship. Absorptive models including Caco-2 cell monolayer model and rat in situ single-pass intestinal perfusion model as well as in vitro enzymatic kinetic study were employed in the current study. The absorption of baicalein, wogonin, and oroxylin A were favorable with wogonin showing the highest permeability based on two absorptive models. However, three flavones underwent a fast and extensive phase II metabolism. The intestinal metabolism of three flavones exhibited species difference between human and rat. Oroxylin A demonstrated the highest intrinsic clearance of glucuronidation among three flavones. The multidrug resistance proteins might be involved in the efflux of their intracellularly formed conjugated metabolites. The pathway of intestinal absorption and disposition of B, W, and OA was similar. However, the extent of permeability and metabolism was different among three flavones which might be due to the number and position of the hydroxyl group.
Collapse
Affiliation(s)
- Chenrui Li
- School of Pharmacy, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong SAR, China
| | | | | | | | | | | |
Collapse
|
23
|
Manda VK, Mittapalli RK, Bohn KA, Adkins CE, Lockman PR. Nicotine and cotinine increases the brain penetration of saquinavir in rat. J Neurochem 2010; 115:1495-507. [PMID: 20950334 DOI: 10.1111/j.1471-4159.2010.07054.x] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Endothelial tight junctions and efflux transporters of the blood-brain barrier (BBB) significantly limit brain accumulation of many drugs, including protease inhibitors such as saquinavir. The cholinergic agonist nicotine is one of the most commonly used drugs in the world and the incidence is even higher in the human immune deficiency virus population (∼ 70%). We examined the ability of nicotine and its primary metabolite cotinine to modify brain uptake of saquinavir in rats. Both nicotine and cotinine at pharmacological concentrations matching those in smokers, increased brain saquinavir uptake by two fold. Co-perfusion with nicotinic receptor antagonists and passive permeability markers showed that the effect was not caused by receptor activation or BBB permeability disruption. Transport inhibition studies demonstrated that brain saquinavir uptake is limited by multiple efflux transporters, P-glycoprotein (P-gp), breast cancer resistance protein and multidrug resistance-associated protein. In situ perfusion and in vitro experiments using a classical P-gp substrate rhodamine 123 linked the effect of nicotine to inhibition of BBB P-gp transport. The effect was confirmed in vivo in chronic 14 day nicotine administration animals. These data suggest nicotine increases antiretroviral drug exposure to brain and may represent a significant in vivo drug-drug interaction at the BBB. Although this may slightly benefit CNS antiretroviral efficacy, it may also expose the brain to potential serious neurotoxicity.
Collapse
Affiliation(s)
- Vamshi K Manda
- Texas Tech University Health Sciences Center, School of Pharmacy, Department of Pharmaceutical Sciences, Amarillo, Texas 79106-1712, USA
| | | | | | | | | |
Collapse
|
24
|
Sauer SW, Opp S, Mahringer A, Kamiński MM, Thiel C, Okun JG, Fricker G, Morath MA, Kölker S. Glutaric aciduria type I and methylmalonic aciduria: simulation of cerebral import and export of accumulating neurotoxic dicarboxylic acids in in vitro models of the blood-brain barrier and the choroid plexus. Biochim Biophys Acta Mol Basis Dis 2010; 1802:552-60. [PMID: 20302929 DOI: 10.1016/j.bbadis.2010.03.003] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2009] [Revised: 02/08/2010] [Accepted: 03/05/2010] [Indexed: 10/19/2022]
Abstract
Intracerebral accumulation of neurotoxic dicarboxylic acids (DCAs) plays an important pathophysiological role in glutaric aciduria type I and methylmalonic aciduria. Therefore, we investigated the transport characteristics of accumulating DCAs - glutaric (GA), 3-hydroxyglutaric (3-OH-GA) and methylmalonic acid (MMA) - across porcine brain capillary endothelial cells (pBCEC) and human choroid plexus epithelial cells (hCPEC) representing in vitro models of the blood-brain barrier (BBB) and the choroid plexus respectively. We identified expression of organic acid transporters 1 (OAT1) and 3 (OAT3) in pBCEC on mRNA and protein level. For DCAs tested, transport from the basolateral to the apical site (i.e. efflux) was higher than influx. Efflux transport of GA, 3-OH-GA, and MMA across pBCEC was Na(+)-dependent, ATP-independent, and was inhibited by the OAT substrates para-aminohippuric acid (PAH), estrone sulfate, and taurocholate, and the OAT inhibitor probenecid. Members of the ATP-binding cassette transporter family or the organic anion transporting polypeptide family, namely MRP2, P-gp, BCRP, and OATP1B3, did not mediate transport of GA, 3-OH-GA or MMA confirming the specificity of efflux transport via OATs. In hCPEC, cellular import of GA was dependent on Na(+)-gradient, inhibited by NaCN, and unaffected by probenecid suggesting a Na(+)-dependent DCA transporter. Specific transport of GA across hCPEC, however, was not found. In conclusion, our results indicate a low but specific efflux transport for GA, 3-OH-GA, and MMA across pBCEC, an in vitro model of the BBB, via OAT1 and OAT3 but not across hCPEC, an in vitro model of the choroid plexus.
Collapse
Affiliation(s)
- Sven W Sauer
- Department of General Pediatrics, Division of Inborn Metabolic Diseases, University Children's Hospital Heidelberg, D-69120 Heidelberg, Germany.
| | | | | | | | | | | | | | | | | |
Collapse
|