1
|
Cheng K, Wang Y, He Y, Tian Y, Li J, Chen C, Xu X, Wu Z, Yu H, Chen X, Wu Y, Song W, Dong Z, Xu H, Xie P. Upregulation of carbonic anhydrase 1 beneficial for depressive disorder. Acta Neuropathol Commun 2023; 11:59. [PMID: 37013604 PMCID: PMC10071615 DOI: 10.1186/s40478-023-01545-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Accepted: 03/06/2023] [Indexed: 04/05/2023] Open
Abstract
Carbonic Anhydrase 1 (CAR1) is a zinc-metalloenzyme that catalyzes the hydration of carbon dioxide, and the alteration of CAR1 has been implicated in neuropsychiatric disorders. However, the mechanism underlying the role of CAR1 in major depressive disorder (MDD) remains largely unknown. In this study, we report the decreased level of CAR1 in MDD patients and depression-like model rodents. We found that CAR1 is expressed in hippocampal astrocytes and CAR1 regulates extracellular bicarbonate concentration and pH value in the partial hilus. Ablation of the CAR1 gene increased the activity of granule cells via decreasing their miniature inhibitory postsynaptic currents (mIPSC), and caused depression-like behaviors in CAR1-knockout mice. Astrocytic CAR1 expression rescued the deficits in mIPSCs of granule cells and reduced depression-like behaviors in CAR1 deficient mice. Furthermore, pharmacological activation of CAR1 and overexpression of CAR1 in the ventral hippocampus of mice improved depressive behaviors. These findings uncover a critical role of CAR1 in the MDD pathogenesis and its therapeutic potential.
Collapse
Affiliation(s)
- Ke Cheng
- NHC Key Laboratory of Diagnosis and Treatment on Brain Functional Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
- Institute of Neuroscience, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Yue Wang
- NHC Key Laboratory of Diagnosis and Treatment on Brain Functional Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
- Institute of Neuroscience, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Yong He
- NHC Key Laboratory of Diagnosis and Treatment on Brain Functional Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Yu Tian
- NHC Key Laboratory of Diagnosis and Treatment on Brain Functional Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Junjie Li
- Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, Chongqing, 400014, China
- Chongqing Key Laboratory of Translational Medical Research in Cognitive Development and Learning and Memory Disorders, Children's Hospital of Chongqing Medical University, Chongqing, 400014, China
| | - Chong Chen
- NHC Key Laboratory of Diagnosis and Treatment on Brain Functional Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Xingzhe Xu
- NHC Key Laboratory of Diagnosis and Treatment on Brain Functional Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Zhonghao Wu
- NHC Key Laboratory of Diagnosis and Treatment on Brain Functional Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Heming Yu
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Xiangyu Chen
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Yili Wu
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Institute of Aging, Zhejiang Provincial Clinical Research Center for Mental Disorders, Key Laboratory of Alzheimer's Disease of Zhejiang Province, School of Mental Health and Kangning Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, 325000, China
| | - Weihong Song
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Institute of Aging, Zhejiang Provincial Clinical Research Center for Mental Disorders, Key Laboratory of Alzheimer's Disease of Zhejiang Province, School of Mental Health and Kangning Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, 325000, China.
| | - Zhifang Dong
- Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, Chongqing, 400014, China.
- Chongqing Key Laboratory of Translational Medical Research in Cognitive Development and Learning and Memory Disorders, Children's Hospital of Chongqing Medical University, Chongqing, 400014, China.
| | - Huatai Xu
- Institute of Neuroscience, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, 200031, China.
- Shanghai Center for Brain Science and Brain-Inspired Intelligence Technology, Shanghai, 201210, China.
| | - Peng Xie
- NHC Key Laboratory of Diagnosis and Treatment on Brain Functional Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China.
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China.
| |
Collapse
|
2
|
Königschulte W, Civai C, Hildebrand P, Gaber TJ, Fink GR, Zepf FD. Effects of serotonin depletion and dopamine depletion on bimodal divided attention. World J Biol Psychiatry 2020; 21:183-194. [PMID: 30295116 DOI: 10.1080/15622975.2018.1532110] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/28/2022]
Abstract
Objectives: This study aimed to explore the effects of acute phenylalanine tyrosine depletion (APTD) and acute tryptophan depletion (ATD) on bimodal divided attention. A balanced amino acid mixture (BAL) served as control condition.Methods: Fifty-three healthy adults (final analyzed sample was N = 49, age: M = 23.8 years) were randomly assigned to APTD, ATD or BAL in a double-blind, between-subject approach. Divided attention was assessed after 4 h. Blood samples were taken before and 6 h after challenge intake.Results: Amino acid concentrations following challenge intake significantly decreased (all P ≤ 0.01). There was a significant difference in the mean reaction time (RT) towards auditory stimuli, but not towards visual stimuli between the groups. Post-hoc comparison of mean RTs (auditory stimuli) showed a significant difference between ATD (RT = 604.0 ms, SD = 56.9 ms) and APTD (RT = 556.4 ms, SD = 54.2 ms; P = 0.037), but no RT difference between ATD and BAL or APTD and BAL (RT = 573.6 ms, SD = 45.7 ms).Conclusions: The results indicate a possible dissociation between the effects of a diminished brain 5-HT and DA synthesis on the performance in a bimodal divided attention task. The difference was exclusively observed within the RT towards auditory signals.
Collapse
Affiliation(s)
- W Königschulte
- Clinic for Child and Adolescent Psychiatry, Psychosomatics and Psychotherapy, RWTH Aachen University, Aachen, Germany.,JARA Translational Brain Medicine, Aachen & Jülich, Germany
| | - C Civai
- School of Psychology, University of Kent, Canterbury, UK
| | - P Hildebrand
- Clinic for Child and Adolescent Psychiatry, Psychosomatics and Psychotherapy, RWTH Aachen University, Aachen, Germany.,JARA Translational Brain Medicine, Aachen & Jülich, Germany
| | - T J Gaber
- Clinic for Child and Adolescent Psychiatry, Psychosomatics and Psychotherapy, RWTH Aachen University, Aachen, Germany.,JARA Translational Brain Medicine, Aachen & Jülich, Germany
| | - G R Fink
- Cognitive Neuroscience, Institute of Neuroscience and Medicine (INM-3), Research Centre Jülich, Jülich, Germany.,Department of Neurology, University of Cologne, Cologne, Germany
| | - F D Zepf
- Clinic for Child and Adolescent Psychiatry, Psychosomatics and Psychotherapy, RWTH Aachen University, Aachen, Germany.,JARA Translational Brain Medicine, Aachen & Jülich, Germany.,Department of Child and Adolescent Psychiatry, Psychosomatic Medicine and Psychotherapy, Jena University Hospital, Friedrich Schiller University Jena, Germany.,Centre and Discipline of Child and Adolescent Psychiatry, Psychosomatics and Psychotherapy, School of Medicine, Division of Psychiatry and Clinical Neurosciences & Division of Paediatrics and Child Health, Faculty of Medicine and Health Sciences, The University of Western Australia, Perth, Australia.,Telethon Kids Institute, Perth, Australia
| |
Collapse
|
3
|
Di Marco A, Gonzalez Paz O, Fini I, Vignone D, Cellucci A, Battista MR, Auciello G, Orsatti L, Zini M, Monteagudo E, Khetarpal V, Rose M, Dominguez C, Herbst T, Toledo-Sherman L, Summa V, Muñoz-Sanjuán I. Application of an in Vitro Blood–Brain Barrier Model in the Selection of Experimental Drug Candidates for the Treatment of Huntington’s Disease. Mol Pharm 2019; 16:2069-2082. [DOI: 10.1021/acs.molpharmaceut.9b00042] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | - Vinod Khetarpal
- CHDI Management, CHDI Foundation, Center Drive Los Angeles 6080, California, United States
| | - Mark Rose
- CHDI Management, CHDI Foundation, Center Drive Los Angeles 6080, California, United States
| | - Celia Dominguez
- CHDI Management, CHDI Foundation, Center Drive Los Angeles 6080, California, United States
| | - Todd Herbst
- CHDI Management, CHDI Foundation, Center Drive Los Angeles 6080, California, United States
| | - Leticia Toledo-Sherman
- CHDI Management, CHDI Foundation, Center Drive Los Angeles 6080, California, United States
| | | | - Ignacio Muñoz-Sanjuán
- CHDI Management, CHDI Foundation, Center Drive Los Angeles 6080, California, United States
| |
Collapse
|
4
|
Bongiovanni R, Mchaourab AS, McClellan F, Elsworth J, Double M, Jaskiw GE. Large neutral amino acids levels in primate cerebrospinal fluid do not confirm competitive transport under baseline conditions. Brain Res 2016; 1648:372-379. [DOI: 10.1016/j.brainres.2016.08.011] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2016] [Revised: 08/08/2016] [Accepted: 08/09/2016] [Indexed: 01/27/2023]
|
5
|
Blood-brain barrier transport of an essential amino acid after cerebral ischemia reperfusion injury. ACTA NEUROCHIRURGICA. SUPPLEMENT 2013; 118:297-302. [PMID: 23564153 DOI: 10.1007/978-3-7091-1434-6_58] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Under pathophysiological conditions such as -cerebral ischemia-reperfusion (IR), damage to cerebrovascular endothelial cells causes alterations in the blood-brain barrier (BBB) function that can exacerbate neuronal cell injury and death. Clarifying changes in BBB transport in the early period of IR is important for understanding BBB function during therapy after cerebral ischemia. The present study was aimed at clarifying changes during IR in the BBB transport of L-phenylalanine (Phe) as a substrate of L-type amino acid transporter 1. An IR model was produced in mice by blood recirculation following occlusion of the middle cerebral artery. Permeability of the BBB to [(3)H]Phe was measured after IR injury using the brain perfusion method. Confocal microscopy of the IR injury showed no brain penetration of fluorescent tracer, thus confirming BBB integrity during 45 min of ischemia. Tight junction opening was not observed at 30 min after reperfusion following ischemia for 45 min. At the time of IR, [(3)H]Phe uptake into the brain appeared saturated. The Michaelis constant and maximum transport velocity in the IR group was reduced by 22 % compared with those in controls. These results suggest that the intrinsic transport clearance of Phe is slightly decreased in the early phase of IR.
Collapse
|
6
|
Suenderhauf C, Hammann F, Huwyler J. Computational prediction of blood-brain barrier permeability using decision tree induction. Molecules 2012; 17:10429-45. [PMID: 22941223 PMCID: PMC6269008 DOI: 10.3390/molecules170910429] [Citation(s) in RCA: 55] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2012] [Revised: 08/17/2012] [Accepted: 08/27/2012] [Indexed: 12/15/2022] Open
Abstract
Predicting blood-brain barrier (BBB) permeability is essential to drug development, as a molecule cannot exhibit pharmacological activity within the brain parenchyma without first transiting this barrier. Understanding the process of permeation, however, is complicated by a combination of both limited passive diffusion and active transport. Our aim here was to establish predictive models for BBB drug permeation that include both active and passive transport. A database of 153 compounds was compiled using in vivo surface permeability product (logPS) values in rats as a quantitative parameter for BBB permeability. The open source Chemical Development Kit (CDK) was used to calculate physico-chemical properties and descriptors. Predictive computational models were implemented by machine learning paradigms (decision tree induction) on both descriptor sets. Models with a corrected classification rate (CCR) of 90% were established. Mechanistic insight into BBB transport was provided by an Ant Colony Optimization (ACO)-based binary classifier analysis to identify the most predictive chemical substructures. Decision trees revealed descriptors of lipophilicity (aLogP) and charge (polar surface area), which were also previously described in models of passive diffusion. However, measures of molecular geometry and connectivity were found to be related to an active drug transport component.
Collapse
Affiliation(s)
- Claudia Suenderhauf
- Division of Pharmaceutical Technology, Department of Pharmaceutical Sciences, University of Basel, Klingelbergstrasse 50, CH-4056 Basel, Switzerland
| | - Felix Hammann
- Division of Pharmaceutical Technology, Department of Pharmaceutical Sciences, University of Basel, Klingelbergstrasse 50, CH-4056 Basel, Switzerland
- Psychiatric Hospital of the University of Basel, Wilhelm-Klein-Str. 27, 4012 Basel, Switzerland
| | - Jörg Huwyler
- Division of Pharmaceutical Technology, Department of Pharmaceutical Sciences, University of Basel, Klingelbergstrasse 50, CH-4056 Basel, Switzerland
| |
Collapse
|
7
|
Avdeef A. How well can in vitro brain microcapillary endothelial cell models predict rodent in vivo blood-brain barrier permeability? Eur J Pharm Sci 2011; 43:109-24. [PMID: 21514381 DOI: 10.1016/j.ejps.2011.04.001] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2010] [Revised: 02/06/2011] [Accepted: 04/03/2011] [Indexed: 01/16/2023]
Abstract
The object of the study was to improve the blood-brain barrier (BBB) permeability in vitro-invivo correlations (IVIVC) between in vitro brain microcapillary endothelial cell (BMEC) models and the well-tested rodent in situ brain perfusion technique. Porcine, bovine, rat, mouse, and human in vitro BMEC apparent permeability values, P(e), (14 studies from several laboratories: 229 P(e), 60 compounds) were analyzed by a novel biophysical model encoded in a weighted nonlinear regression procedure to determine the aqueous boundary layer (ABL) thickness and the paracellular parameters: porosity-pathlength (dual-pore model), pore radius, and water channel electrostatic potential. The refined parameters were then used to transform the P(e) values into the transendothelial permeability (P(c)) values. Porcine BMEC mono-culture models showed tight junctions comparable to those reported in several Caco-2 studies. Bovine cultures were somewhat leakier. In the human primary cultured cell and the hCMEC/D3 cell line data, IVIVC based on P(e) values has r(2) = 0.14. With transformed permeability values, r(2) = 0.58. Comparable improvements were found in the other species data. By using the in vitro transendothelial P(c) values in place of the apparent P(e) values, IVIVC can be dramatically improved.
Collapse
|
8
|
Changes in kinetics of amino acid uptake at the ageing ovine blood-cerebrospinal fluid barrier. Neurobiol Aging 2010; 33:121-33. [PMID: 20138405 DOI: 10.1016/j.neurobiolaging.2010.01.015] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2009] [Revised: 01/15/2010] [Accepted: 01/19/2010] [Indexed: 01/05/2023]
Abstract
Amino acids (AA) in brain are precisely controlled by blood-brain barriers, which undergo a host of changes in both morphology and function during ageing. The effect of these age-related changes on AA homeostasis in brain is not well described. This study investigated the kinetics of four AA (Leu, Phe, Ala and Lys) uptakes at young and old ovine choroid plexus (CP), the blood-cerebrospinal fluid (CSF) barrier (BCB), and measured AA concentrations in CSF and plasma samples. In old sheep, the weight of lateral CP increased, so did the ratio of CP/brain. The expansion of the CP is consistent with clinical observation of thicker leptomeninges in old age. AA concentrations in old CSF, plasma and their ratio were different from the young. Both V(max) and K(m) of Phe and Lys were significant higher compared to the young, indicating higher trans-stimulation in old BCB. Cross-competition and kinetic inhibition studies found the sensitivity and specificity of these transporters were impaired in old BCB. These changes may be the first signs of a compromised barrier system in ageing brain leading increased AA influx into the brain causing neurotoxicity.
Collapse
|
9
|
Geldenhuys WJ, Manda VK, Mittapalli RK, Van der Schyf CJ, Crooks PA, Dwoskin LP, Allen DD, Lockman PR. Predictive screening model for potential vector-mediated transport of cationic substrates at the blood-brain barrier choline transporter. Bioorg Med Chem Lett 2010; 20:870-7. [PMID: 20053562 PMCID: PMC2818856 DOI: 10.1016/j.bmcl.2009.12.079] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2009] [Accepted: 12/18/2009] [Indexed: 11/30/2022]
Abstract
A set of semi-rigid cyclic and acyclic bis-quaternary ammonium analogs, which were part of a drug discovery program aimed at identifying antagonists at neuronal nicotinic acetylcholine receptors, were investigated to determine structural requirements for affinity at the blood-brain barrier choline transporter (BBB CHT). This transporter may have utility as a drug delivery vector for cationic molecules to access the central nervous system. In the current study, a virtual screening model was developed to aid in rational drug design/ADME of cationic nicotinic antagonists as BBB CHT ligands. Four 3D-QSAR comparative molecular field analysis (CoMFA) models were built which could predict the BBB CHT affinity for a test set with an r(2) <0.5 and cross-validated q(2) of 0.60, suggesting good predictive capability for these models. These models will allow the rapid in silico screening of binding affinity at the BBB CHT of both known nicotinic receptor antagonists and virtual compound libraries with the goal of informing the design of brain bioavailable quaternary ammonium analogs that are high affinity selective nicotinic receptor antagonists.
Collapse
Affiliation(s)
- Werner J Geldenhuys
- Department of Pharmaceutical Sciences, Texas Tech University Health Sciences Center, School of Pharmacy, Amarillo, TX 79106-1712, USA.
| | | | | | | | | | | | | | | |
Collapse
|
10
|
Witt KA, Ronaldson PT, Sandoval KE, Davis TP. CNS Delivery of Peptides Across the BBB Using the Dual-Artery In Situ Brain Perfusion Model. NEUROMETHODS 2010. [DOI: 10.1007/978-1-60761-529-3_11] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
|
11
|
Thomas FC, Taskar K, Rudraraju V, Goda S, Thorsheim HR, Gaasch JA, Mittapalli RK, Palmieri D, Steeg PS, Lockman PR, Smith QR. Uptake of ANG1005, a novel paclitaxel derivative, through the blood-brain barrier into brain and experimental brain metastases of breast cancer. Pharm Res 2009; 26:2486-94. [PMID: 19774344 DOI: 10.1007/s11095-009-9964-5] [Citation(s) in RCA: 163] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2009] [Accepted: 08/21/2009] [Indexed: 12/21/2022]
Abstract
PURPOSE We evaluated the uptake of angiopep-2 paclitaxel conjugate, ANG1005, into brain and brain metastases of breast cancer in rodents. Most anticancer drugs show poor delivery to brain tumors due to limited transport across the blood-brain barrier (BBB). To overcome this, a 19-amino acid peptide (angiopep-2) was developed that binds to low density lipoprotein receptor-related protein (LRP) receptors at the BBB and has the potential to deliver drugs to brain by receptor-mediated transport. METHODS The transfer coefficient (K(in)) for brain influx was measured by in situ rat brain perfusion. Drug distribution was determined at 30 min after i.v. injection in mice bearing intracerebral MDA-MB-231BR metastases of breast cancer. RESULTS The BBB K(in) for (125)I-ANG1005 uptake (7.3 +/- 0.2 x 10(-3) mL/s/g) exceeded that for (3)H-paclitaxel (8.5 +/- 0.5 x 10(-5)) by 86-fold. Over 70% of (125)I-ANG1005 tracer stayed in brain after capillary depletion or vascular washout. Brain (125)I-ANG1005 uptake was reduced by unlabeled angiopep-2 vector and by LRP ligands, consistent with receptor transport. In vivo uptake of (125)I-ANG1005 into vascularly corrected brain and brain metastases exceeded that of (14)C-paclitaxel by 4-54-fold. CONCLUSIONS The results demonstrate that ANG1005 shows significantly improved delivery to brain and brain metastases of breast cancer compared to free paclitaxel.
Collapse
Affiliation(s)
- Fancy C Thomas
- Department of Pharmaceutical Sciences, Texas Tech University Health Sciences Center, Amarillo, TX 79106, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
12
|
Dagenais C, Avdeef A, Tsinman O, Dudley A, Beliveau R. P-glycoprotein deficient mouse in situ blood-brain barrier permeability and its prediction using an in combo PAMPA model. Eur J Pharm Sci 2009; 38:121-37. [PMID: 19591928 DOI: 10.1016/j.ejps.2009.06.009] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2009] [Accepted: 06/25/2009] [Indexed: 01/06/2023]
Abstract
The purpose of the study was to assess the permeability of mouse blood-brain barrier (BBB) to a diverse set of compounds in the absence of P-glycoprotein (Pgp) mediated efflux, to predict it using an in combo PAMPA model, and to explore its role in brain penetration classification (BPC). The initial brain uptake (K(in)) of 19 compounds in both wild-type and Pgp mutant [mdr1a(-/-)] CF-1 mice was determined by the in situ brain perfusion technique. PAMPA measurements were performed, and the values were used to develop an in combo model, including Abraham descriptors. Published rodent K(in) values were used to enhance the dataset and validate the model. The model predicted 92% of the variance of the training set permeability. In all, 182 K(in) values were considered in this study, spanning four log orders of magnitude and where Pgp decreased brain uptake by as much as 14-fold. The calculated permeability-surface area (PS) values along with literature reported brain tissue binding were used to group molecules in terms of their brain penetration classification. The in situ BBB permeability can be predicted by the in combo PAMPA model to a satisfactory degree, and can be used as a lower-cost, high throughput first-pass screening method for BBB passive permeability.
Collapse
|
13
|
Deuther-Conrad W, Patt JT, Lockman PR, Allen DD, Patt M, Schildan A, Ganapathy V, Steinbach J, Sabri O, Brust P. Norchloro-fluoro-homoepibatidine (NCFHEB) - a promising radioligand for neuroimaging nicotinic acetylcholine receptors with PET. Eur Neuropsychopharmacol 2008; 18:222-9. [PMID: 17728108 DOI: 10.1016/j.euroneuro.2007.07.002] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/20/2007] [Revised: 06/27/2007] [Accepted: 07/05/2007] [Indexed: 11/15/2022]
Abstract
Cholinergic neurotransmission depends on the integrity of nicotinic acetylcholine receptors (nAChRs), and impairment of both is characteristic for various neurodegenerative diseases. Visualization of specific receptor subtypes by positron emission tomography (PET) has potential to assist with diagnosis of such neurodegenerative diseases and with design of suitable therapeutic approaches. The goal of our study was to evaluate in vivo the potential of (18)F-labelled (+)- and (-)-norchloro-fluoro-homoepibatidine ([(18)F]NCFHEB) in comparison to 2-[(18)F]F-A-85380 as PET tracers. In the brains of NMRI mice, highest levels of radioactivity were detected at 20 min post-injection of (+)-[(18)F]NCFHEB, (-)-[(18)F]NCFHEB, and 2-F-[(18)F]-A-85380 (7.45, 5.60, and 3.2% ID/g tissue, respectively). No marked pharmacological adverse effects were observed at 25 mug NCFHEB/kg. Uptake studies in RBE4 cells and in situ perfusion studies suggest an interaction of epibatidine and NCFHEB with the carrier-mediated choline transport at the blood-brain barrier. The data indicate that (+)- and (-)-[(18)F]NCFHEB have potential for further development as PET tracers.
Collapse
Affiliation(s)
- W Deuther-Conrad
- Institute of Interdisciplinary Isotope Research, Permoserstrasse 15, 04318 Leipzig, Germany.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
14
|
Singhal T, Narayanan TK, Jain V, Mukherjee J, Mantil J. 11C-l-Methionine Positron Emission Tomography in the Clinical Management of Cerebral Gliomas. Mol Imaging Biol 2007; 10:1-18. [DOI: 10.1007/s11307-007-0115-2] [Citation(s) in RCA: 134] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2007] [Revised: 06/30/2007] [Accepted: 07/31/2007] [Indexed: 10/22/2022]
|
15
|
Anderson PJ, Wood SJ, Francis DE, Coleman L, Anderson V, Boneh A. Are Neuropsychological Impairments in Children with Early-Treated Phenylketonuria (PKU) Related to White Matter Abnormalities or Elevated Phenylalanine Levels? Dev Neuropsychol 2007; 32:645-68. [PMID: 17931123 DOI: 10.1080/87565640701375963] [Citation(s) in RCA: 72] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
|
16
|
Park S, Sinko PJ. The blood-brain barrier sodium-dependent multivitamin transporter: a molecular functional in vitro-in situ correlation. Drug Metab Dispos 2005; 33:1547-54. [PMID: 16033951 DOI: 10.1124/dmd.105.005231] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
The molecular mechanism of biotin brain uptake was investigated using an in vitro bovine blood-brain barrier (BBB) cell model and an in situ mouse brain perfusion technique. A functional uptake/transport correlation of the in vitro and in situ characteristics of biotin uptake was investigated. Morphological and immunochemical characteristics (e.g., factor VIII expression) of the primary culture of brain microvessel endothelial cells (BMECs) were confirmed. Gene expression of the multidrug resistance (Mdr1) and sodium-dependent multivitamin (SMVT) transporters was also determined in BMECs. Biotin transport was saturable and Na(+)-dependent at the luminal side of the BBB. The estimated half-saturation concentrations (K(m)) of biotin uptake in vitro and in situ were 49.1 and 35.5 microM, respectively, supporting the presence of a carrier-mediated biotin transport system. Inhibition studies using various biotin derivatives and structural analogs demonstrated the structural requirements for biotin-SMVT interaction. Desthiobiotin and pantothenic acid significantly inhibited the uptake of biotin, whereas 2-iminobiotin and diaminobiotin were very weak inhibitors. Based on our results, there was a good correlation between the in vitro and in situ BBB models, suggesting that when a single membrane transporter is involved in substrate uptake, flexibility in choosing the experimental model can be afforded. The current results are also consistent with the suggestion that the properties of the BBB are likely to be organ-specific rather than species-specific. Further mechanistic and comparative studies are needed to validate these results. In conclusion, the in vitro transporter-based mechanism studies produced valuable molecular functional transport results that correlated well with in situ results.
Collapse
Affiliation(s)
- Seonghee Park
- Rutgers University, Ernest Mario School of Pharmacy, 160 Frelinghuysen Road, Piscataway, NJ 08854, USA
| | | |
Collapse
|
17
|
Liu X, Tu M, Kelly RS, Chen C, Smith BJ. Development of a computational approach to predict blood-brain barrier permeability. Drug Metab Dispos 2004; 32:132-9. [PMID: 14709630 DOI: 10.1124/dmd.32.1.132] [Citation(s) in RCA: 163] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
The objectives of this study were to generate a data set of blood-brain barrier (BBB) permeability values for drug-like compounds and to develop a computational model to predict BBB permeability from structure. The BBB permeability, expressed as permeability-surface area product (PS, quantified as logPS), was determined for 28 structurally diverse drug-like compounds using the in situ rat brain perfusion technique. A linear model containing three descriptors, logD, van der Waals surface area of basic atoms, and polar surface area, was developed based on 23 compounds in our data set, where the penetration across the BBB was assumed to occur primarily by passive diffusion. The correlation coefficient (R(2)) and standard deviation (S.D.) of the model-predicted logPS against the observed are 0.74 and 0.50, respectively. If an outlier was removed from the training data set, the R(2) and S.D. were 0.80 and 0.44, respectively. This new model was tested in two literature data sets, resulting in an R(2) of 0.77 to 0.94 and a S.D. of 0.38 to 0.51. For comparison, four literature models, logP, logD, log(D. MW(-0.5)), and linear free energy relationship, were tested using the set of 23 compounds primarily crossing the BBB by passive diffusion, resulting in an R(2) of 0.33 to 0.61 and a S.D. of 0.59 to 0.76. In summary, we have generated the largest PS data set and developed a robust three-descriptor model that can quantitatively predict BBB permeability. This model may be used in a drug discovery setting to predict the BBB permeability of new chemical entities.
Collapse
Affiliation(s)
- Xingrong Liu
- Department of Pharmacokinetics, Dynamics and Metabolism, Groton Laboratories, Pfizer Global Research and Development, MS 8220-4167, Eastern Point Road, Groton, CT 06340, USA.
| | | | | | | | | |
Collapse
|
18
|
Geldenhuys WJ, Lockman PR, McAfee JH, Fitzpatrick KT, Van der Schyf CJ, Allen DD. Molecular modeling studies on the active binding site of the blood–brain barrier choline transporter. Bioorg Med Chem Lett 2004; 14:3085-92. [PMID: 15149650 DOI: 10.1016/j.bmcl.2004.04.020] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2004] [Revised: 04/09/2004] [Accepted: 04/10/2004] [Indexed: 10/26/2022]
Abstract
The blood-brain barrier choline transporter may have utility as a drug delivery vector to the central nervous system. Surprisingly, this transporter has as yet not been cloned and expressed. We therefore initiated a 3D-QSAR study to develop predictive models for compound binding and identify structural features important for binding to this transporter. In vivo experimental data were obtained from in situ rat brain perfusion studies. Comparative molecular field analysis (CoMFA) and comparative molecular similarity index analysis (CoMSIA) methods were used to build the models. The best cross-validated CoMFA q(2) was found to be 0.47 and the non-cross-validated r(2) was 0.95. CoMSIA hydrophobic cross-validated q(2) was 0.37 and the non-cross-validated r(2) was 0.85. These models rendered a useful approximation for binding requirements in the BBB-choline transporter and, until such time as the cloned transporter becomes available, may have significant utility in developing a predictive model for the rational design of drugs targeted to the brain.
Collapse
Affiliation(s)
- Werner J Geldenhuys
- Department of Pharmaceutical Sciences, Texas Tech University Health Sciences Center, School of Pharmacy, Amarillo, TX 79106, USA
| | | | | | | | | | | |
Collapse
|
19
|
Lockman PR, Mumper RJ, Allen DD. Evaluation of blood-brain barrier thiamine efflux using the in situ rat brain perfusion method. J Neurochem 2003; 86:627-34. [PMID: 12859676 DOI: 10.1046/j.1471-4159.2003.01888.x] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Thiamine is an essential, positively charged (under physiologic conditions), water-soluble vitamin requiring transport into brain. Brain thiamine deficiency has been linked to neurodegenerative disease by subsequent impairment of thiamine-dependent enzymes used in brain glucose/energy metabolism. In this report, we evaluate brain uptake and efflux of [3H]thiamine using the in situ rat brain perfusion technique. To confirm brain distribution was not related to blood-brain barrier endothelial cell uptake, we compared parenchymal and cell distribution of [3H]thiamine using capillary depletion. Our work supports previous literature findings suggesting blood-brain barrier thiamine uptake is via a carrier-mediated transport mechanism, yet extends the literature by redefining the kinetics with more sensitive methodology. Significantly, [3H]thiamine brain accumulation was influenced by a considerable efflux rate. Evaluation of the efflux mechanism demonstrated increased stimulation by the presence of increased vascular thiamine. The influx transport mechanism and efflux rate were each comparable throughout brain regions despite documented differences in glucose and thiamine metabolism. The observation that [3H]thiamine blood-brain barrier influx and efflux is regionally homogenous may have significant relevance to neurodegenerative disease linked to thiamine deficiency.
Collapse
Affiliation(s)
- P R Lockman
- Department of Pharmaceutical Sciences, School of Pharmacy, Texas Tech University HSC, Amarillo, Texas 79106-1712, USA
| | | | | |
Collapse
|
20
|
Lockman PR, Koziara J, Roder KE, Paulson J, Abbruscato TJ, Mumper RJ, Allen DD. In vivo and in vitro assessment of baseline blood-brain barrier parameters in the presence of novel nanoparticles. Pharm Res 2003; 20:705-13. [PMID: 12751624 DOI: 10.1023/a:1023492015851] [Citation(s) in RCA: 70] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
PURPOSE Nanoparticles have advantage as CNS drug delivery vehicles given they disguise drug permeation limiting characteristics. Conflicting toxicological data, however, is published with regard to blood-brain barrier integrity and gross mortality. METHODS To address this issue two novel nanoparticle types: "emulsifying wax/Brij 78" and "Brij 72/Tween 80 nanoparticles were evaluated in vivo for effect on cerebral perfusion flow, barrier integrity, and permeability using the in situ brain perfusion technique. Additional evaluation was completed in vitro using bovine brain microvessel endothelial cells for effect on integrity, permeability, cationic transport interactions, and tight junction protein expression. RESULTS In the presence of either nanoparticle formulation, no overall significant differences were observed for cerebral perfusion flow in vivo. Furthermore, observed in vitro and in vivo data showed no statistical changes in barrier integrity, membrane permeability, or facilitated choline transport. Western blot analyses of occludin and claudin-1 confirmed no protein expression changes with incubation of either nanoparticle. CONCLUSIONS The nanoparticle formulations appear to have no effect on primary BBB parameters in established in vitro and in vivo blood-brain barrier models.
Collapse
Affiliation(s)
- Paul R Lockman
- Department of Pharmaceutical Sciences, School of Pharmacy, Texas Tech University HSC, 1300 So. Coulter Dr., Amarillo, Texas 79106-1712, USA
| | | | | | | | | | | | | |
Collapse
|
21
|
Abstract
The availability of amino acids in the brain is regulated by the blood-brain barrier (BBB) large neutral amino acid transporter type 1 (LAT1) isoform, which is characterized by a high affinity (low Km) for substrate large neutral amino acids. The hypothesis that brain amino acid transport activity can be altered with single nucleotide polymorphisms was tested in the present studies with site-directed mutagenesis of the BBB LAT1. The rabbit has a high Km LAT1 large neutral amino acid transporter, as compared to the low Km neutral amino acid transporter at the human or rat BBB. The rabbit LAT1 was cloned from a rabbit brain capillary cDNA library. Alignment of the amino acid sequences of rabbit, human, and rat LAT1 revealed two radical amino acid residues that differ in the rabbit relative to the rat or human LAT1. The G219D mutation had a modest effect on the Km and Vmax of tryptophan transport via cloned rabbit LAT1 in frog oocytes, but the W234L variant reduced the Km by 64% and the Vmax by 96%. Conversely, LAT1 transport of either tryptophan or phenylalanine was nearly normalized when the double mutation W234L/G219D variant was produced. These studies show that marked changes in the affinity and capacity of the LAT1 are caused by single nucleotide polymorphisms and that phenotype can be restored with a double mutation.
Collapse
Affiliation(s)
- Ruben J Boado
- Department of Medicine, UCLA School of Medicine, Los Angeles, California 90024, USA.
| | | | | |
Collapse
|
22
|
Mann GE, Yudilevich DL, Sobrevia L. Regulation of amino acid and glucose transporters in endothelial and smooth muscle cells. Physiol Rev 2003; 83:183-252. [PMID: 12506130 DOI: 10.1152/physrev.00022.2002] [Citation(s) in RCA: 319] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
While transport processes for amino acids and glucose have long been known to be expressed in the luminal and abluminal membranes of the endothelium comprising the blood-brain and blood-retinal barriers, it is only within the last decades that endothelial and smooth muscle cells derived from peripheral vascular beds have been recognized to rapidly transport and metabolize these nutrients. This review focuses principally on the mechanisms regulating amino acid and glucose transporters in vascular endothelial cells, although we also summarize recent advances in the understanding of the mechanisms controlling membrane transport activity and expression in vascular smooth muscle cells. We compare the specificity, ionic dependence, and kinetic properties of amino acid and glucose transport systems identified in endothelial cells derived from cerebral, retinal, and peripheral vascular beds and review the regulation of transport by vasoactive agonists, nitric oxide (NO), substrate deprivation, hypoxia, hyperglycemia, diabetes, insulin, steroid hormones, and development. In view of the importance of NO as a modulator of vascular tone under basal conditions and in disease and chronic inflammation, we critically review the evidence that transport of L-arginine and glucose in endothelial and smooth muscle cells is modulated by bacterial endotoxin, proinflammatory cytokines, and atherogenic lipids. The recent colocalization of the cationic amino acid transporter CAT-1 (system y(+)), nitric oxide synthase (eNOS), and caveolin-1 in endothelial plasmalemmal caveolae provides a novel mechanism for the regulation of NO production by L-arginine delivery and circulating hormones such insulin and 17beta-estradiol.
Collapse
Affiliation(s)
- Giovanni E Mann
- Centre for Cardiovascular Biology and Medicine, Guy's, King's, and St. Thomas' School of Biomedical Sciences, King's College London, London, United Kingdom.
| | | | | |
Collapse
|
23
|
Zielke HR, Zielke CL, Baab PJ, Collins RM. Large neutral amino acids auto exchange when infused by microdialysis into the rat brain: implication for maple syrup urine disease and phenylketonuria. Neurochem Int 2002; 40:347-54. [PMID: 11792465 DOI: 10.1016/s0197-0186(01)00077-8] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Maple syrup urine disease (MSUD) and phenylketonuria (PKU) are associated with accumulation of large neutral amino acids (LNAA) in blood and tissues and a decrease of other LNAA not directly related to the enzyme defects. One characteristic shared by both the elevated and decreased amino acids is that all are substrates for transport via the large neutral amino acid transporter. In this study, the blood brain barrier was effectively bypassed using microdialysis to determine the immediate effect of infused phenylalanine, tyrosine, 2-amino-2-norborane-carboxylic acid (BCH), and leucine and alpha-ketoisocaproate on extracellular levels of LNAA. The concentration of non-infused LNAA increased in the interstitial fluid, presumably due to trans-stimulated exchange of these LNAA from intracellular pools as the infused LNAA entered the cells. Such trans-stimulated exchange can potentially deplete cells of multiple essential LNAA. It is proposed that brain cells in disorders such as MSUD and PKU may be subject to two mechanisms that limit the availability of a full complement of these amino acids: competition for transport of LNAAs at the blood brain barrier and trans-stimulated exchange out of neuronal cells for subsequent metabolism or sequestration in the periphery.
Collapse
Affiliation(s)
- H Ronald Zielke
- Department of Pediatrics, University of Maryland, Pediatric Research, 10-035 BRB, 655 W Baltimore Street, Baltimore, MD 21201, USA.
| | | | | | | |
Collapse
|
24
|
Lockman PR, Roder KE, Allen DD. Inhibition of the rat blood-brain barrier choline transporter by manganese chloride. J Neurochem 2001; 79:588-94. [PMID: 11701762 DOI: 10.1046/j.1471-4159.2001.00589.x] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Choline transport has been characterized by multiple mechanisms including the blood-brain barrier (BBB), and high- and low-affinity systems. Each mechanism has unique locations and characteristics yet retain some similarities. Previous studies have demonstrated cationic competition by monovalent cations at the BBB and cation divalent manganese in the high-affinity system. To evaluate the effects of divalent manganese inhibition as well as other cationic metals at the BBB choline transporter, brain choline uptake was evaluated in the presence of certain metals of interest in Fischer-344 rats using the in situ brain perfusion technique. Brain choline uptake was inhibited in the presence of Cd(2+) (73 +/- 2%) and Mn(2+) (44 +/- 6%), whereas no inhibition was observed with Cu(2+) and Al(3+). Furthermore, it was found that manganese caused a reduction in brain choline uptake and significant regional choline uptake inhibition in the frontal and parietal cortex, the hippocampus and the caudate putamen (45 +/- 3%, 68 +/- 18%, 58 +/- 9% and 46 +/- 15%, respectively). These results suggest that choline uptake into the CNS can be inhibited by divalent cationic metals and monovalent cations. In addition, the choline transporter may be a means by which manganese enters the brain.
Collapse
Affiliation(s)
- P R Lockman
- Department of Pharmaceutical Sciences, Texas Tech University Health Science Center, Amarillo, Texas 79106-1712, USA
| | | | | |
Collapse
|
25
|
Mahar Doan KM, Boje KM. Theoretical pharmacokinetic and pharmacodynamic simulations of drug delivery mediated by blood--brain barrier transporters. Biopharm Drug Dispos 2000; 21:261-78. [PMID: 11512126 DOI: 10.1002/bdd.242] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
Pharmacokinetic/pharmacodynamic simulations were performed to assess the feasibility of central nervous system (CNS) drug delivery via endogenous transporters resident at the blood-brain barrier (BBB). Pharmacokinetic models were derived for intravenous bolus dosing of a hypothetical drug in the absence or presence of an endogenous, competing transport inhibitor. These models were linked to CNS pharmacodynamic models where the effect sites were either cell surface receptors or intracellularly localized enzymes. The response of the dependent parameter, the duration of effect (t(dur)), was examined in relationship to changes in the independent parameters, i.e. dose, elimination rate constant (k(e1)), BBB transport parameters (K(m1) and V(max1)) and EC(50) (effective concentration that elicits a 50% response). As expected, t(dur) increased with (a) increases in drug doses, (b) decreases in k(e1) or (c) decreases in EC(50), irrespective of the effect site. Surprisingly, endogenous transport inhibition produced decreases in drug terminal half-life and corresponding decreases in t(dur). Interestingly, t(dur) was independent of assigned transporter K(m) and V(max) when the dose/EC(50) ratio (dose/EC(50)) was >1 (irrespective of endogenous transport inhibition), but highly dependent on K(m1) and V(max1) when dose/EC(50) was (a) <1 (no endogenous transport inhibition) or (b) equal to 1 (with endogenous transport inhibition). Oral input of the endogenous transport inhibitor produced a decrease in t(dur) when the dose/EC(50) range was 0.1-10. These simulations highlight that (a) systemic pharmacokinetic and BBB transport parameters influence t(dur), (b) drug terminal half-life is inversely related to circulating levels of endogenous inhibitors, and (c) oral ingestion of endogenous transport inhibitor(s) reduces t(dur). Overall, these simulations provide insight for the feasibility of rational CNS drug design/delivery via endogenous transporters.
Collapse
Affiliation(s)
- K M Mahar Doan
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, University at Buffalo, Buffalo, NY 14260, USA
| | | |
Collapse
|
26
|
Abstract
In most regions of the brain, the uptake of glutamate and other anionic excitatory amino acids from the circulation is limited by the blood-brain barrier (BBB). In most animals, the BBB is formed by the brain vascular endothelium, which contains cells that are joined by multiple bands of tight junctions. These junctions effectively close off diffusion through intercellular pores; as a result, most solutes cross the BBB either by diffusing across the lipoid endothelial cell membranes or by being transported across by specific carriers. Glutamate transport at the BBB has been studied by both in vitro cell uptake assays and in vivo perfusion methods. The results demonstrate that at physiologic plasma concentrations, glutamate flux from plasma into brain is mediated by a high affinity transport system at the BBB. Efflux from brain back into plasma appears to be driven in large part by a sodium-dependent active transport system at the capillary abluminal membrane. Glutamate concentration in brain interstitial fluid is only a fraction of that of plasma and is maintained fairly independently of small fluctuations in plasma concentration. Restricted brain passage is also observed for several excitatory glutamate analogs, including domoic acid and kynurenic acid. In summary, the BBB is one component of a regulatory system that helps maintain brain interstitial fluid glutamate concentration independently of the circulation.
Collapse
Affiliation(s)
- Q R Smith
- Department of Pharmaceutical Sciences, Texas Tech University Health Sciences Center, Amarillo, TX 79106, USA
| |
Collapse
|
27
|
Metting TL, Burgio DE, Terry AV, Beach JW, McCurdy CR, Allen DD. Inhibition of brain choline uptake by isoarecolone and lobeline derivatives: implications for potential vector-mediated brain drug delivery. Neurosci Lett 1998; 258:25-8. [PMID: 9876043 DOI: 10.1016/s0304-3940(98)00871-4] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Delivery of certain compounds to brain is restricted by the nature of the blood-brain barrier (BBB). Many valuable pharmaceuticals are excluded from the CNS due to hydrophilicity or charge. These limitations have been overcome by numerous methods. One method we use is to take advantage of saturable nutrient transporters located at the barrier. These systems transport hydrophilic and charged nutrients into brain such as choline, a quaternized neurotransmitter precursor. Using knowledge of their substrate specificity, it is possible to deliver agents into brain using these nutrient carriers. In this report, derivatives of lobeline and isoarecolone were evaluated to determine if they may gain access to brain by the blood-brain barrier basic amine transporter using the in situ brain perfusion technique. These compounds do bind the blood-brain barrier basic amine transporter and may enter brain by this transport system.
Collapse
Affiliation(s)
- T L Metting
- Department of Pharmaceutical Sciences, School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo 79106-1712, USA
| | | | | | | | | | | |
Collapse
|
28
|
Rousselle CH, Lefauconnier JM, Allen DD. Evaluation of anesthetic effects on parameters for the in situ rat brain perfusion technique. Neurosci Lett 1998; 257:139-42. [PMID: 9870339 DOI: 10.1016/s0304-3940(98)00827-1] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
Studies of drug distribution to brain should be controlled for the experimental method used. Numerous methods have been employed to ascertain brain distribution and many of these approaches use anesthetic agents. The in situ rat brain perfusion method is one of the most sensitive and widely used methods for evaluating brain distribution profiles. There has been no evaluation of the effects of anesthetic agents on parameters associated with this method (i.e. cerebral perfusion fluid flow, brain vascular volume and blood-brain barrier permeability). We evaluated the effects of the anesthetic agents pentobarbital and ketamine combinations on these baseline parameters. The results suggest that the anesthetic agent has no effect on these parameters and anesthetic selection is open to the choice of the investigator when using the perfusion method.
Collapse
|
29
|
Möller HE, Weglage J, Wiedermann D, Ullrich K. Blood-brain barrier phenylalanine transport and individual vulnerability in phenylketonuria. J Cereb Blood Flow Metab 1998; 18:1184-91. [PMID: 9809507 DOI: 10.1097/00004647-199811000-00004] [Citation(s) in RCA: 53] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
In vivo nuclear magnetic resonance spectroscopy can be used to measure intracerebral phenylalanine (Phe) concentrations in patients with phenylketonuria (PKU). Stationary levels, obtained under free nutrition, as well as time courses after an oral Phe load (100 mg/kg) were investigated in 11 PKU patients and were correlated with the individual clinical outcome. At blood levels around 1.2 mmol/L, brain Phe was 0.41 to 0.73 mmol/L in clinically "typical" patients, but less than 0.15 mmol/L in three untreated, normally intelligent, adult women. Kinetic investigations revealed higher transport Michaelis constants and lower ratios of the brain influx and consumption rates in these women than in the "typical" control patients (Kt,app = 0.45 to 1.10 mmol/L versus 0.10 mmol/L; T(max)/v(met) = 2.55 to 3.19 versus 7.8 to 14.0). Such variations seem to be major causative factors for the individual vulnerability to PKU.
Collapse
Affiliation(s)
- H E Möller
- Center for In Vivo Microscopy, Duke University Medical Center, Durham, North Carolina 27710, USA
| | | | | | | |
Collapse
|
30
|
Möller HE, Weglage J, Wiedermann D, Vermathen P, Bick U, Ullrich K. Kinetics of phenylalanine transport at the human blood-brain barrier investigated in vivo. Brain Res 1997; 778:329-37. [PMID: 9459550 DOI: 10.1016/s0006-8993(97)01054-8] [Citation(s) in RCA: 41] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
In vivo proton magnetic resonance spectroscopy was used to investigate intracerebral phenylalanine (Phe) concentrations in nine patients with classical phenylketonuria (PKU). The study included serial examinations (n = 31; plasma Phe levels: 0.47-2.24 mmol/l) of patients either receiving a Phe-restricted diet (200 mg Phe per day; four patients) or a diet rich in Phe (1000 mg Phe per day; three patients). No spectrum showed metabolic abnormalities besides elevated Phe. Difference spectroscopy yielded intracerebral Phe concentrations between 0.20 and 0.76 mmol/l. Regional variations between parieto-occipital periventricular brain, frontal brain, and cerebellum were not statistically significant. Data could be fitted assuming saturable Phe transport into the brain, based on a symmetric Michaelis-Menten model (characterized by an apparent Michaelis transport constant, K(t,app), and a maximum transport velocity, Tmax) and constant Phe consumption in the brain cells (described by a velocity Vmax). Non-linear least-squares fitting of the combined data from all patients yielded K(t,app) = 0.16 +/- 0.11 mmol/l and (Tmax / Vmax) = 9.0 +/- 4.1. Carrier saturation and competitive inhibition of the influx of other large neutral amino acids can be expected at plasma Phe levels usually found in PKU patients.
Collapse
Affiliation(s)
- H E Möller
- Center for In Vivo Microscopy, Duke University Medical Center, Durham, NC 27710, USA.
| | | | | | | | | | | |
Collapse
|
31
|
Thomas SA, Segal MB. Saturation kinetics, specificity and NBMPR sensitivity of thymidine entry into the central nervous system. Brain Res 1997; 760:59-67. [PMID: 9237518 DOI: 10.1016/s0006-8993(97)00276-x] [Citation(s) in RCA: 22] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
It was not until the development of a technique that could measure the brain uptake of slowly moving substrates, that the saturable transport system at the blood-brain barrier (BBB) for the pyrimidine deoxyribonucleoside, thymidine, was demonstrated. The aim of this present study was to further characterize this saturable uptake system at the blood-brain and blood-CSF barriers in terms of specificity, 6-(4-nitrobenzyl)thio-9-beta-D-ribofuranosylpurine (NBMPR) sensitivity and saturation kinetics by means of the in situ brain perfusion technique in anaesthetized guinea pigs. The results indicated that the transport system identified for [3H]thymidine can also transport other pyrimidine deoxyribonucleosides (deoxycytidine) and pyrimidine ribonucleosides (uridine) and is partially NBMPR-sensitive. In addition, guanosine, monocarboxylic acids, hexoses or amino acids were not substrates for the transport system. Further studies revealed that the transport system for [3H]thymidine at the BBB has a low affinity (Km 0.20 +/- 0.06 mM), but a relatively high capacity (Vmax 1.06 +/- 0.08 nmol min(-1) g(-1)). Overall, this study is indicative of a NBMPR-sensitive (es) facilitative transport system for [3H]thymidine and the likely presence of a NBMPR-insensitive and/or sodium-dependent transport system of the N2 (cit) type at the blood-brain and blood-CSF barriers of the guinea pig.
Collapse
Affiliation(s)
- S A Thomas
- Sherrington School of Physiology, UMDS St. Thomas' Hospital Campus, University of London, UK.
| | | |
Collapse
|
32
|
Williams WM, Chang MC, Hayakawa T, Grange E, Rapoport SI. In vivo incorporation from plasma of radiolabeled palmitate and arachidonate into rat brain microvessels. Microvasc Res 1997; 53:163-6. [PMID: 9143548 DOI: 10.1006/mvre.1996.1984] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Affiliation(s)
- W M Williams
- Laboratory of Neurosciences, National Institute on Aging, National Institutes of Health, Bethesda, Maryland 20892, USA
| | | | | | | | | |
Collapse
|
33
|
Benrabh H, Lefauconnier JM. Glutamate is transported across the rat blood-brain barrier by a sodium-independent system. Neurosci Lett 1996; 210:9-12. [PMID: 8762179 DOI: 10.1016/0304-3940(96)12635-5] [Citation(s) in RCA: 38] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Transport of L-glutamate from blood to brain in equithesin-anesthetized rats was examined using in situ brain perfusion combined with multiple-time/graphical analysis. In situ perfusion allowed precise control of the composition of the perfusate, which was necessary for a detailed investigation of glutamate transport, while multiple time/graphical analysis permitted evaluation of the rapidly reversible volume and the period when the influx was unidirectional. Glutamate had no reversible volume and efflux from brain occurred after 30 s of perfusion. The in situ transfer coefficient (Kin) ranged from 0.74 +/- 0.07 mul/s per g in parietal cortex to 0.44 +/- 0.07 mul/s per g in hippocampus. L-Glutamate uptake was unaffected by removal of sodium from the perfusate, reduced by 5 mM L-glutamate, L-homocysteate, L-aspartate, plasma and 0.1 mM L-glutamate, while L-cystine did not reduce its uptake. These results suggest that the transport system for glutamate is saturated mainly by L-glutamate at physiological conditions and that it is not the sodium-independent x-C system since glutamate transport was not reduced by L-cystine except in hippocampus and that it was responsive to L-aspartate.
Collapse
Affiliation(s)
- H Benrabh
- INSERM U26, Hopital F. Widal, Paris, France
| | | |
Collapse
|
34
|
Smith QR. Brain perfusion systems for studies of drug uptake and metabolism in the central nervous system. PHARMACEUTICAL BIOTECHNOLOGY 1996; 8:285-307. [PMID: 8791815 DOI: 10.1007/978-1-4899-1863-5_15] [Citation(s) in RCA: 82] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Affiliation(s)
- Q R Smith
- Neurochemistry and Brain Transport Section, National Institute on Aging, National Institutes of Health, Bethesda, Maryland 20892, USA
| |
Collapse
|
35
|
Tamai I, Senmaru M, Terasaki T, Tsuji A. Na(+)- and Cl(-)-dependent transport of taurine at the blood-brain barrier. Biochem Pharmacol 1995; 50:1783-93. [PMID: 8615856 DOI: 10.1016/0006-2952(95)02046-2] [Citation(s) in RCA: 63] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
The characteristics of carrier-mediated transport of taurine at the blood-brain barrier (BBB) were studied by using primary cultured bovine brain capillary endothelial cells (BCECs), in situ brain perfusion and brain capillary depletion methods in rats. The uptake of [3H]taurine by cultured cells showed that the active transporter functions on both the luminal and antiluminal membranes of BCECs. The kinetic parameters for the saturable transport of taurine were estimated to be: for the luminal uptake, the Michaelis constant, Kt, was 12.1 +/- 0.5 microM, and the maximum uptake rate, Jmax, was 4.32 +/- 0.05 nmol/30 min/mg protein; for the antiluminal uptake, Kt was 13.6 +/- 2.4 microM and Jmax was 2.81 +/- 0.22 nmol/30 min/mg protein. The luminal and antiluminal uptakes of [3H]taurine were each dependent on both Na+ Cl-. Stoichiometric analyses suggest that two Na+ and one Cl- are associated with the luminal uptake of one taurine molecule. beta-Amino acids such as beta-alanine and hypotaurine strongly inhibited the uptake of [3H]taurine, whereas alpha- and gamma-amino acids had little or no effect. Furthermore, by in situ brain perfusion and in vivo brain capillary depletion methods, the carrier-mediated transport found by in vitro experiments was confirmed to function for the translocation of the taurine molecule from the vascular space into the brain. From these results, it was concluded that a Na+ and Cl- gradient-dependent transport (uptake) system for taurine exists in both the luminal and the antiluminal membranes of BCECs.
Collapse
Affiliation(s)
- I Tamai
- Department of Pharmaceutics, Faculty of Pharmaceutical Sciences, Kanazawa University, Japan
| | | | | | | |
Collapse
|
36
|
Benrabh H, Bourre JM, Lefauconnier JM. Taurine transport at the blood-brain barrier: an in vivo brain perfusion study. Brain Res 1995; 692:57-65. [PMID: 8548320 DOI: 10.1016/0006-8993(95)00648-a] [Citation(s) in RCA: 45] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Taurine transport into six brain regions of equithesin-anesthetized rats was studied by the in situ brain perfusion technique. This technique gives both accurate measurements of cerebrovascular amino acid transport and allows complete control of the perfusate amino acid composition. Final wash procedure showed that taurine efflux occurred rapidly from endothelial cells. The taurine influx into endothelial cells was sodium and chloride dependent suggesting that the sodium and chloride gradients are the principal source of energy for taurine transport into endothelial cells. Taurine transport could be fitted by a model with saturable components. The kinetic constants in the parietal cortex were 1.4 x 10(-4) mumol/s/g for the apparent Vmax and 0.078 mM for the apparent Km. Competition experiments in the presence of sodium ions showed that [14C]taurine uptake was strongly inhibited by the structural analogs of taurine, hypotaurine and beta-alanine.
Collapse
Affiliation(s)
- H Benrabh
- INSERM U 26, Hopital Fernand Widal, Paris, France
| | | | | |
Collapse
|
37
|
Tang JP, Melethil S. Effect of aging on the kinetics of blood-brain barrier uptake of tryptophan in rats. Pharm Res 1995; 12:1085-91. [PMID: 7494807 DOI: 10.1023/a:1016283003747] [Citation(s) in RCA: 22] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
PURPOSE The purpose of this investigation was to examine the effect of aging on the blood-brain barrier (BBB) transport of tryptophan. METHODS A well established in-situ brain perfusion technique was used to examine brain uptake of 14C-tryptophan in 2-, 12- and 24-month old Sprague-Dawley rats; perfusate tryptophan concentrations ranged from 0.00175 to 2 mM. Uptake data were modeled using non-linear regression analysis. RESULTS Permeability-surface area product (PA) for tryptophan was significantly lower in 12- and 24-month old rats, as compared to the 2-month old animals. A transport model consisting of both saturable (Michaelis-Menten type) and non-saturable components best described brain uptake of tryptophan in all 3 age groups. However, age-dependent differences in BBB transport parameters of tryptophan were observed. For the saturable component, both Vmax and Km were significantly lower in the 12- and 24-month old rats, as compared to the youngest group of rats. These results suggest that transporter mobility, number and affinity for tryptophan are altered in older rats. Values for Kd, the rate constant for non-saturable brain tryptophan transport, were also significantly lower in animals of the two older age groups. Interestingly, PA values for thiourea, a compound believed to be transporter across BBB by diffusion, were also lower in these two age groups. CONCLUSIONS Aging decreases the ability of the BBB to transport the neutral amino acid tryptophan.
Collapse
Affiliation(s)
- J P Tang
- School of Pharmacy, University of Missouri-Kansas City 64108, USA
| | | |
Collapse
|
38
|
|
39
|
Shirai A, Naito M, Tatsuta T, Dong J, Hanaoka K, Mikami K, Oh-hara T, Tsuruo T. Transport of cyclosporin A across the brain capillary endothelial cell monolayer by P-glycoprotein. BIOCHIMICA ET BIOPHYSICA ACTA 1994; 1222:400-4. [PMID: 7913624 DOI: 10.1016/0167-4889(94)90047-7] [Citation(s) in RCA: 62] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
P-glycoprotein, a multidrug transporter protein, exists in the brain capillary endothelium. To study the function of P-glycoprotein in brain capillary endothelium as a barrier against cyclosporin A, we examined the interaction of cyclosporin A with P-glycoprotein expressed in cultured brain capillary endothelial cells (MBEC4). P-glycoprotein of MBEC4 specifically bound [125I]iodoaryl azidoprazosin, and the binding was inhibited by cyclosporin A and vincristine. Intracellular accumulation of cyclosporin A in MBEC4 was about one-third the amount accumulated in mouse aortic endothelial cells (MAEC3), a cell line that did not express P-glycoprotein. The reduced accumulation of cyclosporin A in MBEC4 was increased by verapamil, a competitive inhibitor of transport function of P-glycoprotein. Cyclosporin A was preferentially transported from basal to apical side when the cell monolayer of MBEC4 was formed; however this transendothelial transport was not observed across cell monolayer of MAEC3. Verapamil inhibited the transendothelial transport of cyclosporin A across the MBEC4 monolayer. Thus P-glycoprotein in brain capillary endothelium could transport cyclosporin A across the endothelium from the basal to the apical side. These observations suggest that P-glycoprotein is involved in the complex function of the blood-brain barrier as a secretory detoxifying transporter of cyclosporin A.
Collapse
Affiliation(s)
- A Shirai
- Institute of Molecular and Cellular Biosciences, University of Tokyo, Japan
| | | | | | | | | | | | | | | |
Collapse
|
40
|
Wadhwani KC, Rapoport SI. Transport properties of vertebrate blood-nerve barrier: comparison with blood-brain barrier. Prog Neurobiol 1994; 43:235-79. [PMID: 7816928 DOI: 10.1016/0301-0082(94)90002-7] [Citation(s) in RCA: 22] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Affiliation(s)
- K C Wadhwani
- Laboratory of Neurosciences, NIA, NIH, Bethesda, MD 20892
| | | |
Collapse
|
41
|
Chikhale EG, Ng KY, Burton PS, Borchardt RT. Hydrogen bonding potential as a determinant of the in vitro and in situ blood-brain barrier permeability of peptides. Pharm Res 1994; 11:412-9. [PMID: 8008709 DOI: 10.1023/a:1018969222130] [Citation(s) in RCA: 162] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
With the exception of various central nervous system (CNS)-required nutrients for which specific, saturable transport systems exist, the passage of most water-soluble solutes through the blood-brain barrier (BBB) is believed to depend largely on the lipid solubility of the solutes. Most peptides, therefore, do not enter the CNS because of their hydrophilic character. Recently, utilizing homologous series of model peptides and Caco-2 cell monolayers as a model of the intestinal mucosa, it was concluded that the principal determinant of peptide transport across the intestinal cellular membrane is the energy required to desolvate the polar amide bonds in the peptide (P. S. Burton et al., adv. Drug Deliv. Rev. 7:365, 1991). To determine whether this correlation can be extended to the BBB, the permeabilities of the same peptides were determined using an in vitro as well as an in situ BBB model. The peptides, blocked on the N- and C-terminal ends, consisted of D-phenylalanine (F) residues: AcFNH2, AcF2NH2, AcF3NH2, AcF2(NMeF)NH2, AcF(NMeF)2NH2, Ac(NMeF)3NH2, and Ac(NMeF)3NHMe. A good correlation among the permeabilities of these model peptides across the bovine brain microvessel endothelial cell (BBMEC) monolayers, an in vitro model of the BBB, and their permeabilities across the BBB in situ was observed (r = 0.928, P < 0.05). The permeabilities of these peptides did not correlate with the octanol-buffer partition coefficients of the peptides (r = 0.389 in vitro and r = 0.155 in situ; P < 0.05).(ABSTRACT TRUNCATED AT 250 WORDS)
Collapse
Affiliation(s)
- E G Chikhale
- Department of Pharmaceutical Chemistry, University of Kansas, Lawrence 66045
| | | | | | | |
Collapse
|
42
|
Hammerstad JP, Pate BD, Hewitt KA, Chan GL, Ruth TJ, Calne DB. The transport of L-6-fluorodopa and its metabolites from blood to cerebrospinal fluid and brain. Ann Neurol 1993; 34:603-8. [PMID: 8215248 DOI: 10.1002/ana.410340415] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
The transport of L-6-fluorodopa and its major metabolites from the blood to the brain, cerebrospinal fluid (CSF), and muscle was studied in carbidopa-pretreated cynomolgus monkeys. A bolus intravenous injection of 18F-L-6-fluorodopa was followed by serial positron emission tomography scans and sampling of cisternal CSF and arterial blood. The relative concentrations of L-6-fluorodopa and its metabolites were determined in blood plasma and CSF by high-performance liquid chromatography. Raising the blood concentration of phenylalanine by intraperitoneal injection markedly reduced the accumulation of tracer in the brain. This indicates that L-6-fluorodopa and 3-O-methylfluorodopa, like native L-dopa and its O-methylated derivative, are transported at the brain capillary by the large neutral amino acid carrier-mediated system, which is subject to saturation and competition by other large neutral amino acids (such as phenylalanine) at physiological plasma concentrations. In contrast, administration of phenylalanine had no effect on the accumulation of tracer either in muscle, or as L-6-fluorodopa and 3-O-methylfluorodopa, in CSF. This suggests that the transport of L-dopa and its derivatives at the blood-CSF barrier differs from the transport at the blood-brain barrier and also that measurement of CSF L-dopa is not a good index of the transport and pharmacokinetics of L-dopa in the brain. However, the effect of phenylalanine administration in reducing the concentration of fluorohomovanillic acid in the CSF suggests that the concentration of homovanillic acid in the CSF is an accurate reflection of dopamine turnover in the brain.
Collapse
Affiliation(s)
- J P Hammerstad
- Neurodegenerative Disorders Centre, University of British Columbia, Vancouver, Canada
| | | | | | | | | | | |
Collapse
|
43
|
Smith QR. Drug delivery to brain and the role of carrier-mediated transport. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 1993; 331:83-93. [PMID: 8333351 DOI: 10.1007/978-1-4615-2920-0_14] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
In summary, the results suggest that carrier-mediated transport can be used to augment the brain delivery of a wide variety of hydrophilic therapeutic drugs. A large number of carriers are now known to be present at the brain capillary endothelium, and in many instances these carriers have been shown to mediate the brain uptake of exogenous drugs. The findings with D,L-NAM demonstrate that brain delivery can be improved through design of selective, high affinity agents. Although NAM was developed for the large neutral amino acid carrier, high affinity drugs could be produced for other systems, as shown by the work of Schein et al. with nitrogen mustard monosaccharides and by the work of Deves and Krupka on choline derivatives. Lastly, the method may allow some selectivity of delivery because of differential expression of transport carriers between tissues and in various disease states.
Collapse
Affiliation(s)
- Q R Smith
- Laboratory of Neurosciences, National Institute on Aging, National Institutes of Health, Bethesda, MD 20892
| |
Collapse
|
44
|
Tatsuta T, Naito M, Oh-hara T, Sugawara I, Tsuruo T. Functional involvement of P-glycoprotein in blood-brain barrier. J Biol Chem 1992. [DOI: 10.1016/s0021-9258(19)88713-6] [Citation(s) in RCA: 139] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022] Open
|
45
|
Vaalburg W, Coenen HH, Crouzel C, Elsinga PH, Långström B, Lemaire C, Meyer GJ. Amino acids for the measurement of protein synthesis in vivo by PET. INTERNATIONAL JOURNAL OF RADIATION APPLICATIONS AND INSTRUMENTATION. PART B, NUCLEAR MEDICINE AND BIOLOGY 1992; 19:227-37. [PMID: 1601675 DOI: 10.1016/0883-2897(92)90011-m] [Citation(s) in RCA: 39] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
In principle, PET in combination with amino acids labelled with positron-emitting radionuclides and kinetic metabolic models, can quantify local protein synthesis rates in tissue in vivo. These PET measurements have clinical potential in, for example, oncology, neurology and psychiatry. An optimal positron-emitting amino acid for the measurement of PSR has a high protein incorporation, can easily be prepared by automated equipment and has minimal non-protein radioactive metabolites. Presently L-[methyl-11C]methionine, L-[1-11C]leucine, L-[1-11C]tyrosine, L-[1-11C]phenylalanine, L-[1-11C]methionine and L-[2-18F]fluorotyrosine are under evaluation in normal volunteers and/or in patients. Several other amino acids are suggested. No comparison of the clinical usefulness of the different amino acids in man is yet available. Because of the longer half life of 18F compared to 11C, [18F]fluoro amino acids may have advantages over [11C]amino acids for the investigation of tissue with relative slow protein synthesis, such as brain, and for application in institutions with an off site, but nearby cyclotron. The half life of [13N]amino acids is considered to be too short for flexible clinical application. As yet no metabolic compartmental model has been investigated for [13N]amino acids. For routine application reliable preparation of the radiopharmaceutical is essential. Of all the amino acids under evaluation, a reliable, high yield, easy to automate production procedure is available for L-[methyl-11C]methionine only. It is however unlikely that this tracer can accurately measure PSR because of its non-protein metabolism. For the other amino acids the main problems in production are associated with complex multistep syntheses and/or low radiochemical yields, complex purification methods and the need to isolate the L-enantiomer. The kinetic metabolic models under investigation, consist of 4 or 5 compartments depending on the necessity to compensate for labelled metabolites. The metabolic profile of the amino acids is mainly extracted from animal experiments. Because of the number and amount of labelled metabolites in plasma, [11C]carboxylic labelled amino acids are preferred to amino acids with carbon-11 in another position. As yet no recommendation can be given on the optimal labelled amino acid(s) for PSR measurement in vivo nor on the methods to prepare the amino acids reported for this purpose.
Collapse
Affiliation(s)
- W Vaalburg
- Department of Nuclear Medicine, University Hospital, Groningen, The Netherlands
| | | | | | | | | | | | | |
Collapse
|
46
|
Møller SE. Effect of aspartame and protein, administered in phenylalanine-equivalent doses, on plasma neutral amino acids, aspartate, insulin and glucose in man. PHARMACOLOGY & TOXICOLOGY 1991; 68:408-12. [PMID: 1946186 DOI: 10.1111/j.1600-0773.1991.tb01262.x] [Citation(s) in RCA: 19] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Six human males each received 0.56 g phenylalanine (Phe) in the form of 1.0 g aspartame or 12.2 g bovine albumin in 200 ml water or water alone. Venous blood samples collected before consumption and during the following 4 hr were assayed for plasma levels of large, neutral amino acids (LNAA), aspartate, insulin and glucose. The area under the curve for plasma Phe was 40% greater, although not significant, after aspartame compared with albumin intake. The indicated increased clearance rate of plasma Phe after albumin may be caused by the significant increase of insulin, on which aspartame had no effect. There was a significant main effect of aspartame for plasma tyrosine but not for tryptophan, valine, isoleucine or leucine. Plasma aspartate was significantly increased at 0.25 hr after the aspartame intake. The percentage Phe/LNAA decreased slightly in response to albumin but increased 55% after aspartame and remained significantly increased for 2 hr. Tyrosine/LNAA increased and tryptophan/LNAA decreased modestly after aspartame intake. The study showed that the intake of aspartame in a not unrealistically high dose produced a marked and persistent increase of the availability of Phe to the brain, which was not observed after protein intake. The study indicated, furthermore, that Phe was cleared faster from the plasma after consumption of protein compared with aspartame.
Collapse
Affiliation(s)
- S E Møller
- Clinical Research Laboratory, St. Hans Hospital, Roskilde, Denmark
| |
Collapse
|
47
|
Takada Y, Greig NH, Vistica DT, Rapoport SI, Smith QR. Affinity of antineoplastic amino acid drugs for the large neutral amino acid transporter of the blood-brain barrier. Cancer Chemother Pharmacol 1991; 29:89-94. [PMID: 1760863 DOI: 10.1007/bf00687316] [Citation(s) in RCA: 43] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
The relative affinity of six anticancer amino acid drugs for the neutral amino acid carrier of the blood-brain barrier was examined in rats using an in situ brain perfusion technique. Affinity was evaluated from the concentration-dependent inhibition of L-[14C]-leucine uptake into rat brain during perfusion at tracer leucine concentrations and in the absence of competing amino acids. Of the six drugs tested, five, including melphalan, azaserine, acivicin, 6-diazo-5-oxo-L-norleucine, and buthionine sulfoximine, exhibited only low affinity for the carrier, displaying transport inhibition constants (Ki, concentrations producing 50% inhibition) ranging from 0.09 to 4.7 mM. However, one agent - D,L-2-amino-7-bis[(2-chloroethyl)amino]- 1,2,3,4-tetrahydro-2-naphthoic acid (D,L-NAM) - demonstrated remarkably high affinity for the carrier, showing a Ki value of approximately 0.2 microM. The relative affinity (1/Ki) of D,L-NAM was greater than 100-fold that of the other drugs and greater than 10-fold that of any compound previously tested. As the blood-brain barrier penetrability of most endogenous neutral amino acids is related to their carrier affinity, the results suggest that D,L-NAM may be a promising agent which may show enhanced uptake and distribution to brain tumors.
Collapse
Affiliation(s)
- Y Takada
- Laboratory of Neurosciences, National Institute on Aging, National Institutes of Health, Bethesda, MD 20892
| | | | | | | | | |
Collapse
|
48
|
Smith QR. The blood-brain barrier and the regulation of amino acid uptake and availability to brain. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 1991; 291:55-71. [PMID: 1927691 DOI: 10.1007/978-1-4684-5931-9_6] [Citation(s) in RCA: 28] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Affiliation(s)
- Q R Smith
- Laboratory of Neurosciences, National Institute on Aging, National Institutes of Health, Bethesda, Maryland
| |
Collapse
|
49
|
Brust P, Diemer NH. Blood-brain transfer of L-phenylalanine declines after peripheral but not central nervous administration of vasopressin. J Neurochem 1990; 55:2098-104. [PMID: 2230811 DOI: 10.1111/j.1471-4159.1990.tb05801.x] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
To determine whether a previously reported effect of vasopressin on blood-brain transfer of leucine extends to other large neutral amino acids, we measured the regional blood-brain transfer of L-phenylalanine with the integral technique. Intravenous co-injection of L-phenylalanine and arginine vasopressin (30 nmol to 10 pmol) resulted in a decrease of the permeability-surface area (PaS) product of phenylalanine of between 11 and 39%. In addition, the peptide elicited a decrease of the cerebral blood flow of between 11 and 56% combined with a drastic decrease of the cardiac output (32-64%) and an elevation of the blood pressure to approximately 150% of control values. However, we found no changes of the cardiac output, the blood pressure, or the PaS product of phenylalanine after microdialysis (30 min, 5 microliters min-1) of arginine vasopressin (15 mumol L-1) into the dorsal hippocampus, but cerebral blood flow was decreased. The results support the hypothesis that arginine vasopressin receptors at the blood-brain barrier are involved in the regulation of large neutral amino acid transfer from blood to brain and indicate that these receptors are located at the luminal membrane of the endothelial cells.
Collapse
Affiliation(s)
- P Brust
- Department of Cell Biology and Regulation, Karl Marx University, Leipzig, G.D.R
| | | |
Collapse
|
50
|
Greig NH, Soncrant TT, Shetty HU, Momma S, Smith QR, Rapoport SI. Brain uptake and anticancer activities of vincristine and vinblastine are restricted by their low cerebrovascular permeability and binding to plasma constituents in rat. Cancer Chemother Pharmacol 1990; 26:263-8. [PMID: 2369790 DOI: 10.1007/bf02897227] [Citation(s) in RCA: 63] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Unidirectional blood-brain barrier transfer of the lipophilic anticancer agents vincristine and vinblastine was studied in anesthetized rats, using an isolated, in situ brain perfusion technique. Drug binding to plasma constituents was also measured. Despite the high lipophilicity of these agents (the log octanol/physiological saline partition coefficient equalled 2.14 and 1.68, respectively), the cerebrovascular permeability-surface area product, PA, of vincristine in plasma was only 0.49 x 10(-4) ml s-1 g-1 for parietal cerebral cortex, whereas that of vinblastine was too low for determination. These values are similar to those of water-soluble, poorly diffusible nonelectrolytes. The PAs were significantly higher in the absence of plasma protein, being 1.24 x 10(-4) and 5.36 x 10(-4) ml s-1 g-1, respectively. Even these values, determined by brain perfusion of protein-free buffer, were lower than would be expected from the lipophilicity of the agents. The results suggest that additional factors, such as steric hindrance and molecular charge distribution, related to the chemical and geometric structure and the large size of vincristine and vinblastine (molecular weight, 825 and 814 daltons, respectively) restrict their passage across the blood-brain barrier. As a consequence of their paradoxically low permeability at the blood-brain barrier and restrictive binding to plasma and blood constituents, doses of both agents that cause significant inhibition of extracerebral Walker 256 carcinosarcoma tumor implants in rat have no effect on tumor located in the brain.
Collapse
Affiliation(s)
- N H Greig
- Laboratory of Neurosciences, National Institute on Aging, National Institutes of Health, Bethesda, MD 20892
| | | | | | | | | | | |
Collapse
|