1
|
Miah MK, Bickel U, Mehvar R. Effects of hepatic ischemia-reperfusion injury on the blood-brain barrier permeability to [ 14C] and [ 13C]sucrose. Metab Brain Dis 2017; 32:1903-1912. [PMID: 28779418 DOI: 10.1007/s11011-017-0069-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/21/2017] [Accepted: 07/11/2017] [Indexed: 01/28/2023]
Abstract
Hepatic encephalopathy that is associated with severe liver failure may compromise the blood-brain barrier (BBB) integrity. However, the effects of less severe liver diseases, in the absence of overt encephalopathy, on the BBB are not well understood. The goal of the current study was to investigate the effects of hepatic ischemia-reperfusion (IR) injury on the BBB tight junction permeability to small, hydrophilic molecules using the widely used [14C]sucrose and recently-proposed alternative [13C]sucrose as markers. Rats were subjected to 20 min of hepatic ischemia or sham surgery, followed by 8 h of reperfusion before administration of a single bolus dose of [14C] or [13C]sucrose and collection of serial (0-30 min) blood and plasma and terminal brain samples. The concentrations of [14C] and [13C]sucrose in the samples were determined by measurement of total radioactivity (nonspecific) and LC-MS/MS (specific), respectively. IR injury significantly increased the blood, plasma, and brain concentrations of both [14C] and [13C]sucrose. However, when the brain concentrations were corrected for their respective area under the blood concentration-time curve, only [14C]sucrose showed significantly higher (30%) BBB permeability values in the IR animals. Because [13C]sucrose is a more specific BBB permeability marker, these data indicate that our animal model of hepatic IR injury does not affect the BBB tight junction permeability to small, hydrophilic molecules. Methodological differences among studies of the effects of liver diseases on the BBB permeability may confound the conclusions of such studies.
Collapse
Affiliation(s)
- Mohammad K Miah
- Department of Pharmaceutical Sciences, School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, TX, USA
- School of Pharmacy, University of Pittsburgh, Pittsburgh, PA, USA
| | - Ulrich Bickel
- Department of Pharmaceutical Sciences, School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, TX, USA.
- Center for Blood-Brain Barrier Research, Texas Tech University Health Sciences Center, Amarillo, TX, USA.
| | - Reza Mehvar
- Department of Pharmaceutical Sciences, School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, TX, USA.
- Center for Blood-Brain Barrier Research, Texas Tech University Health Sciences Center, Amarillo, TX, USA.
- Department of Biomedical and Pharmaceutical Sciences, School of Pharmacy, Chapman University, 9401 Jeronimo Road, Irvine, CA, USA.
| |
Collapse
|
2
|
Evaluation of [ 14C] and [ 13C]Sucrose as Blood-Brain Barrier Permeability Markers. J Pharm Sci 2017; 106:1659-1669. [PMID: 28238901 DOI: 10.1016/j.xphs.2017.02.011] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2016] [Revised: 02/10/2017] [Accepted: 02/13/2017] [Indexed: 12/30/2022]
Abstract
Nonspecific quantitation of [14C]sucrose in blood and brain has been routinely used as a quantitative measure of the in vivo blood-brain barrier (BBB) integrity. However, the reported apparent brain uptake clearance (Kin) of the marker varies widely (∼100-fold). We investigated the accuracy of the use of the marker in comparison with a stable isotope of sucrose ([13C]sucrose) measured by a specific liquid chromatography-tandem mass spectrometry method. Rats received single doses of each marker, and the Kin values were determined. Surprisingly, the Kin value of [13C]sucrose was 6- to 7-fold lower than that of [14C]sucrose. Chromatographic fractionation after in vivo administration of [14C]sucrose indicated that the majority of the brain content of radioactivity belonged to compounds other than the intact [14C]sucrose. However, mechanistic studies failed to reveal any substantial metabolism of the marker. The octanol:water partition coefficient of [14C]sucrose was >2-fold higher than that of [13C]sucrose, indicating the presence of lipid-soluble impurities in the [14C]sucrose solution. Our data indicate that [14C]sucrose overestimates the true BBB permeability to sucrose. We suggest that specific quantitation of the stable isotope (13C) of sucrose is a more accurate alternative to the current widespread use of the radioactive sucrose as a BBB marker.
Collapse
|
3
|
Abstract
Sleep and its disorders are known to affect the functions of essential organs and systems in the body. However, very little is known about how the blood-brain barrier (BBB) is regulated. A few years ago, we launched a project to determine the impact of sleep fragmentation and chronic sleep restriction on BBB functions, including permeability to fluorescent tracers, tight junction protein expression and distribution, glucose and other solute transporter activities, and mediation of cellular mechanisms. Recent publications and relevant literature allow us to summarize here the sleep-BBB interactions in five sections: (1) the structural basis enabling the BBB to serve as a huge regulatory interface; (2) BBB transport and permeation of substances participating in sleep-wake regulation; (3) the circadian rhythm of BBB function; (4) the effect of experimental sleep disruption maneuvers on BBB activities, including regional heterogeneity, possible threshold effect, and reversibility; and (5) implications of sleep disruption-induced BBB dysfunction in neurodegeneration and CNS autoimmune diseases. After reading the review, the general audience should be convinced that the BBB is an important mediating interface for sleep-wake regulation and a crucial relay station of mind-body crosstalk. The pharmaceutical industry should take into consideration that sleep disruption alters the pharmacokinetics of BBB permeation and CNS drug delivery, being attentive to the chrono timing and activation of co-transporters in subjects with sleep disorders.
Collapse
Affiliation(s)
- Weihong Pan
- 1 Biopotentials Sleep Center, Baton Rouge, LA 70809
| | - Abba J Kastin
- 2 Blood-Brain Barrier Group, Pennington Biomedical Research Center, Baton Rouge, LA, USA
| |
Collapse
|
4
|
Chodobski A, Ghersi-Egea JF, Nicholson C, Nagaraja TN, Szmydynger-Chodobska J. The quest for a better insight into physiology of fluids and barriers of the brain: the exemplary career of Joseph D. Fenstermacher. Fluids Barriers CNS 2015; 12:1. [PMID: 25745556 PMCID: PMC4350980 DOI: 10.1186/2045-8118-12-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2014] [Accepted: 12/18/2014] [Indexed: 12/27/2022] Open
Abstract
In June 2014 Dr. Joseph D. Fenstermacher celebrated his 80th birthday, which was honored by the symposium held in New London, NH, USA. This review discusses Fenstermacher's contribution to the field of fluids and barriers of the CNS. Specifically, his fundamental work on diffusion of molecules within the brain extracellular space and the research on properties of the blood-brain barrier in health and disease are described. Fenstermacher's early research on cerebrospinal fluid dynamics and the regulation of cerebral blood flow is also reviewed, followed by the discussion of his more recent work involving the use of magnetic resonance imaging.
Collapse
Affiliation(s)
- Adam Chodobski
- Department of Emergency Medicine, Neurotrauma and Brain Barriers Research Laboratory, The Warren Alpert Medical School of Brown University, Coro Center West, Room 112, 1 Hoppin Street, Providence, RI 02903 USA
| | - Jean-François Ghersi-Egea
- Blood-Brain Interface Group, Oncoflam Team and BIP Platform INSERM U 1028, CNRS UMR5292 Lyon Neuroscience Research Center, Faculté de Médecine RTH Laennec, Rue Guillaume Paradin, Cedex 08, 69372 Lyon, France
| | - Charles Nicholson
- Department of Neuroscience and Physiology, NYU School of Medicine, MSB 460, 550 First Avenue, New York, NY 10016 USA
| | - Tavarekere N Nagaraja
- Department of Anesthesiology, Henry Ford Hospital, 2799 West Grand Blvd., Detroit, MI 48202-2689 USA
| | - Joanna Szmydynger-Chodobska
- Department of Emergency Medicine, Neurotrauma and Brain Barriers Research Laboratory, The Warren Alpert Medical School of Brown University, Coro Center West, Room 112, 1 Hoppin Street, Providence, RI 02903 USA
| |
Collapse
|
5
|
Dashti SR, Spalding A, Kadner RJ, Yao T, Kumar A, Sun DA, LaRocca R. Targeted intraarterial anti-VEGF therapy for medically refractory radiation necrosis in the brain. J Neurosurg Pediatr 2015; 15:20-5. [PMID: 25360851 DOI: 10.3171/2014.9.peds14198] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Radiation necrosis (RN) is a serious complication that can occur in up to 10% of brain radiotherapy cases, with the incidence dependent on both dose and brain location. Available medical treatment for RN includes steroids, vitamin E, pentoxifylline, and hyperbaric oxygen. In a significant number of patients, however, RN is medically refractory and the patients experience progressive neurological decline, disabling headaches, and decreased quality of life. Vascular endothelial growth factor (VEGF) is a known mediator of cerebral edema in RN. Recent reports have shown successful treatment of RN with intravenous bevacizumab, a monoclonal antibody for VEGF. Bevacizumab, however, is associated with significant systemic complications including sinus thrombosis, pulmonary embolus, gastrointestinal tract perforation, wound dehiscence, and severe hypertension. Using lower drug doses may decrease systemic exposure and reduce complication rates. By using an intraarterial route for drug administration following blood-brain barrier disruption (BBBD), the authors aim to lower the bevacizumab dose while increasing target delivery. In the present report, the authors present the cases of 2 pediatric patients with cerebral arteriovenous malformations, who presented with medically intractable RN following stereotactic radiosurgery. They received a single intraarterial infusion of 2.5 mg/kg bevacizumab after hyperosmotic BBBD. At mean follow-up duration of 8.5 months, the patients had significant and durable clinical and radiographic response. Both patients experienced resolution of their previously intractable headaches and reversal of cushingoid features as they were successfully weaned off steroids. One of the patients regained significant motor strength. There was an associated greater than 70% reduction in cerebral edema. Intraarterial administration of a single low dose of bevacizumab after BBBD was safe and resulted in durable clinical and radiographic improvements at concentrations well below those required for the typical systemic intravenous route. Advantages over the intravenous route may include higher concentration of drug delivery to the affected brain, decreased systemic toxicity, and a significantly lower cost.
Collapse
|
6
|
Abstract
This issue of the journal honors Professor Henry McIlwain for his contributions to our knowledge of neurochemistry, as a pioneer (an important contributor already in the 1950s), as a scientist, and as a teacher of great influence and help to the next generation of neurochemists. It is fitting that in his semi-retirement he turns his interest to the history and background of our discipline and demonstrates to us that there is a great deal to learn from the past. In today's explosion of knowledge and new approaches, and the consequent rush to do the work, we tend to forget not only the important past accomplishments but also the past mistakes not to be repeated. It is worthwhile from time to time to take stock, to look back at the path that led to the present. This paper is an attempt to explore this retrospection by a discussion of some of the background of research on cerebral amino acid transport. Emphasis for the purpose is on illustration, with arbitrarily selected examples rather than an exhaustive review of the subject.
Collapse
Affiliation(s)
- A Lajtha
- Center for Neurochemistry, The Nathan S. Kline Institute for Psychiatric Research, Ward's Island, New York, NY 10035, U.S.A
| |
Collapse
|
7
|
Cattelotte J, André P, Ouellet M, Bourasset F, Scherrmann JM, Cisternino S. In situ mouse carotid perfusion model: glucose and cholesterol transport in the eye and brain. J Cereb Blood Flow Metab 2008; 28:1449-59. [PMID: 18446168 DOI: 10.1038/jcbfm.2008.34] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
The in situ mouse brain perfusion method for measuring blood-brain barrier permeability was adapted to assess transport of solutes at the blood-brain and blood-eye barriers. The procedure was checked with radiolabeled markers in oxygenated bicarbonate-buffered fluid infused for 30 to 120 sec via a carotid artery. Vascular flow estimated with diazepam was 2.2-fold lower in the eye than in the brain. The vascular volume and the integrity markers sucrose and inulin indicated that a perfusion flow rate of 2.5 mL/min preserved the physical integrity of these organs. However, the brain vasculature integrity was more sensitive to acute perfusion pressure than the eye vasculature. The functional capacities of blood barriers were assessed with D-glucose; its transport followed Michaelis-Menten kinetics with an apparent K(m) of 7.6 mmol/L and a V(max) of 23 micromol/sec per g in the brain, and a K(m) of 22.9 mmol/L and a V(max) of 40 micromol/sec per g in the eye. The transport of cholesterol to the brain and eye was significantly enhanced by adding the Abca1 inhibitor probucol, suggesting an Abca1-mediated efflux at the mouse brain and eye blood barriers. Thus in situ carotid perfusion is suitable for elucidating transport processes at the blood-brain and blood-eye barriers.
Collapse
Affiliation(s)
- Julie Cattelotte
- Department of Pharmacokinetics, Faculty of Pharmacy, INSERM U705, CNRS UMR 7157, Université Paris Descartes, Paris, France
| | | | | | | | | | | |
Collapse
|
8
|
Nagaraja TN, Keenan KA, Brown SL, Fenstermacher JD, Knight RA. Relative distribution of plasma flow markers and red blood cells across BBB openings in acute cerebral ischemia. Neurol Res 2007; 29:78-80. [PMID: 17427280 DOI: 10.1179/174313206x153815] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022]
Abstract
Acute blood-brain barrier (BBB) opening in cerebral ischemia is an often observed but seldom studied phenomenon. Increased permeability has been implicated with several consequences including exacerbating ischemic injury, leading to hemorrhagic transformation (HT) and also predictive of chronic damage and a way of delivering therapeutics to the diseased parts of brain. Very few studies have investigated the 'size' of such acute openings. Herein the blood-brain distribution of fluorescent isothiocyanate (FITC)- labeled red blood cells (RBCs; approximately 5 tm in diameter) and two different sized plasma flow markers in cerebral microvessels was studied by laser scanning confocal microscopy (LSCM) 6 and 24 hours after the onset of a 3 hour period of focal ischemia. At hour 6, Evans blue-tagged albumin [EB-Alb; molecular weight (MW)= 68 kDa, Stokes-Einstein radius=37 A], a marker of both plasma flow and BBB opening, was seen both inside and around microvessels whereas the RBCs were only intravascular. FITC-labeled dextran (FITC-dextran; MW=2000 kDa, Stokes-Einstein radius = approximately 150 A), another plasma flow tracer, had not leaked across the BBB into the tissue at this time. At hour 24, both RBCs and FITC-dextran were found extravascularly along with EB-Alb. We postulate that smaller sized openings in BBB at hour 6 limited the leaking of the two large tracers (RBCs and FITC-dextran) and that such size-dependency was lost by 24 hours with the progression of the ischemic injury.
Collapse
|
9
|
Abstract
The transport system for the cytokine tumor necrosis factor-alpha (TNFalpha) at the blood-brain barrier (BBB) enables an enhanced yet saturable entry of TNFalpha from blood to the CNS. This review focuses on the selective upregulation of the transport system for TNFalpha at the BBB that is specific for type of pathology, region, and time. The upregulation is reflected by increased CNS tissue uptake of radiolabeled TNFalpha after iv injection in mice and by inhibition of this increase with excess non-radiolabeled TNFalpha. (1) Spinal cord injury (SCI): upregulation of TNFalpha uptake after thoracic transection is seen in the delayed phase of BBB disruption at the lumbar spinal cord. Thoracic SCI by compression, however, has a longer lasting impact on TNFalpha transport that involves thoracic and lumbar spinal cord, in contrast to the upregulation confined to the lumbar region in lumbar SCI by compression. Regardless, the uptake of TNFalpha by spinal cord does not parallel BBB disruption as measured by the leakage of radiolabeled albumin. (2) Experimental autoimmune encephalomyelitis (EAE): the increase in the differential permeability to TNFalpha is seen in all CNS regions (brain and cervical, thoracic, and lumbar spinal cord) and has a distinct time course and reversibility. Exogenous TNFalpha has biphasic effects in modulating functional scores. The BBB, a dynamically regulated barrier, is actively involved in disease processes.
Collapse
Affiliation(s)
- W Pan
- VA Medical Center and Tulane University School of Medicine, New Orleans, LA 70112-1262, USA.
| | | |
Collapse
|
10
|
Guérin CJ, Nolan CC, Mavroudis G, Lister T, Davidson GM, Holton JL, Ray DE. The dynamics of blood-brain barrier breakdown in an experimental model of glial cell degeneration. Neuroscience 2001; 103:873-83. [PMID: 11301198 DOI: 10.1016/s0306-4522(01)00015-x] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
Abstract
This study was undertaken to investigate the dynamics of blood-brain barrier breakdown in an in vivo rat model of selective CNS vulnerability. 1,3-Dinitrobenzene was used to induce rapid glial degeneration in highly defined areas of the brainstem. Leakage of fluorescent dextran was used to demonstrate the breakdown of the blood-brain barrier, and antibodies to glial and neuronal specific proteins to assess the accompanying cell changes. Beginning 18 h after a toxic dose of dinitrobenzene and before loss of glial ensheathment, a sub-population of blood vessels became permeable to fluorescent dextrans below 500,000 mol. wt in size. By 24h most macroglial cells had been lost from within susceptible areas and vascular leakage had reached peak levels. Macrophage invasion was detected three days following dinitrobenzene. Vessels continued to leak up to four days after the lesion was formed, but by six days blood-brain barrier integrity was largely re-established. Multiple tracer injections over time demonstrated that a single sub-population of vessels was leaking during the experimental period. From these findings we conclude that blood-brain barrier breakdown in this model system is highly selective, graded in extent and molecular weight specificity and not a direct consequence of astrocyte degeneration or microglial activation. This system could be useful in modeling human CNS pathological processes with a vascular component and for understanding in vivo glial blood-brain barrier interactions.
Collapse
Affiliation(s)
- C J Guérin
- Medical Research Council Toxicology Unit, Neurotoxicology Group, Hodgkin Building, Lancaster Road, LE1 9HN, Leicester, UK.
| | | | | | | | | | | | | |
Collapse
|
11
|
Abstract
The blood-brain barrier minimizes the entry of molecules into brain tissue. This restriction arises by the presence of tight junctions (zonulae occludens) between adjacent endothelial cells and a relative paucity of pinocytotic vesicles within endothelium of cerebral arterioles, capillaries, and venules. Many types of stimuli can alter the permeability characteristics of the blood-brain barrier. Acute increases in arterial blood pressure beyond the autoregulatory capacity of cerebral blood vessels, application of hyperosmolar solutions, application of various inflammatory mediators known to be elevated during brain injury, and/or activation of blood-borne elements such as leukocytes can produce changes in permeability of the blood-brain barrier. The second messenger systems that account for increases in permeability of the blood-brain barrier during pathophysiologic conditions, however, remain poorly defined. This review will summarize studies that have examined factors that influence disruption of the blood-brain barrier, and will discuss the contribution of various cellular second messenger pathways in disruption of the blood-brain barrier during pathophysiologic conditions.
Collapse
Affiliation(s)
- W G Mayhan
- Department of Physiology and Biophysics, University of Nebraska Medical Center, Omaha 68198-4575, USA.
| |
Collapse
|
12
|
Preston E, Webster J, Small D. Characteristics of sustained blood-brain barrier opening and tissue injury in a model for focal trauma in the rat. J Neurotrauma 2001; 18:83-92. [PMID: 11200252 DOI: 10.1089/089771501750055794] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Minor stab wounding of rodent brain by needle or razor blade is a standard model for immunohistochemical investigations of secondary neuronal degeneration and scarring. Opening of the blood-brain barrier (BBB) to plasma molecules and inflammatory cells is integral to the secondary injury process. To facilitate quantitative study of these BBB phenomena, we tested the utility of a stereotaxic wire knife as a minimally invasive way for modeling of focal trauma and bleeding in brain parenchyma and substantial, reproducible BBB damage. Adult rats were anesthetized, and through a skull burr hole, the 0.3-mm dia guide cannula housing a laterally extendable tungsten wire (0.13 mm dia) was inserted into the right striatum. A layering of horizontal disk-like cuts (3 mm dia) was made, producing a cylindrical lesion of approximately 18 mm3 volume, approximately 2.7% of the cerebral hemisphere. Transfer constants (Ki) for blood to brain permeation of [3H]sucrose measured from 30 min to 2 weeks postlesion showed sustained BBB leakiness; for example, mean Ki +/- SEM (nL.g(-1) x s(-1)) for a standard, matrix-dissected forebrain sample enclosing the lesion were 7.2 +/- 1.2 (day 1 postlesion), 8.1 +/-1.4 (day 3), 5.4 +/- 0.8 (day 14) compared with values for contralateral nonlesioned forebrain ranging from 1.3 +/- 0.05 to 1.6 +/- 0.3 (n = 3-4 samples per time point). Analysis of the simultaneous transport of [14C]sucrose (MW = 342 Da) and [3H]inulin (MW approximately 5,000) showed significantly larger upward increments in Ki for sucrose than inulin, indicating a pore-like opening mechanism. Significant edema was measured 3 days postlesion. A reactive glial response was indicated by an increase in S100beta by 6 h and a glial scar forming around the lesion by 7 days. Secondary brain injury was indicated by a 10% loss of hemisphere mass, measured at 2 months. The wire knife enabled tailoring of interstitial trauma with a minimum of extraneous injury and supported reproducible measurements of sustained BBB injury using relatively few animals.
Collapse
Affiliation(s)
- E Preston
- Institute for Biological Sciences, National Research Council of Canada, Ottawa, Ontario.
| | | | | |
Collapse
|
13
|
Rapoport SI. Osmotic opening of the blood-brain barrier: principles, mechanism, and therapeutic applications. Cell Mol Neurobiol 2000; 20:217-30. [PMID: 10696511 DOI: 10.1023/a:1007049806660] [Citation(s) in RCA: 305] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
1. Osmotic opening of the blood-brain barrier by intracarotid infusion of a hypertonic arabinose or mannitol solution is mediated by vasodilatation and shrinkage of cerebrovascular endothelial cells, with widening of the interendothelial tight junctions to an estimated radius of 200 A. The effect may be facilitated by calcium-mediated contraction of the endothelial cytoskeleton. 2. The marked increase in apparent blood-brain barrier permeability to intravascular substances (10-fold for small molecules) following the osmotic procedure is due to both increased diffusion and bulk fluid flow across the tight junctions. The permeability effect is largely reversed within 10 min. 3. In experimental animals, the osmotic method has been used to grant wide access to the brain of water-soluble drugs, peptides, antibodies, boron compounds for neutron capture therapy, and viral vectors for gene therapy. The method also has been used together with anticancer drugs to treat patients with metastatic or primary brain tumors, with some success and minimal morbidity.
Collapse
Affiliation(s)
- S I Rapoport
- Section on Brain Physiology and Metabolism, National Institute on Aging, National Institutes of Health, Bethesda, Maryland 20892, USA.
| |
Collapse
|
14
|
Mackic JB, Stins M, Jovanovic S, Kim KS, Bartus RT, Zlokovic BV. Cereport (RMP-7) increases the permeability of human brain microvascular endothelial cell monolayers. Pharm Res 1999; 16:1360-5. [PMID: 10496650 DOI: 10.1023/a:1018938722768] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
PURPOSE To study Cereport (RMP-7, bradykinin B2 agonist) effects on human brain microvascular endothelial cell (HBMEC) monolayer permeability. METHODS HBMEC grown on transwell membranes were exposed to Cereport. The monolayer permeability was determined with [14C]-inulin (MW. 5,200) and [3H]-dextran (MW. 70,000). RESULTS Cereport increased the HBMEC permeability to [14C]-inulin, but not to [3H]-dextran. The effect was transient, maximal at 15 min (i.e., 79.3% increase), and polarized to the basolateral membrane. An inverted U, dose-response curve was observed with active concentrations of Cereport from 0.01 to 0.5 nmol/L, the plateau maximal effect between 0.5 and 10 nmol/L, and loss of activity at the highest concentration, i.e., 20 nmol/L. Cyclic AMP-specific phosphodiesterase 3 (PDE3) inhibitor rolipram (10 micromol/L) abolished Cereport effects, while cGMP-specific PDE5 inhibitor, zaniprast (50 micromol/L) enhanced by 31% (p < 0.05) the effect of 0.1 nmolL Cereport. Unlabeled Cereport displaced [125I]-bradykinin and/or [125I]-Cereport from the basolateral side. There was no specific Cereport binding to the apical side. CONCLUSIONS Cereport exerts specific time, dose and size dependent actions on HMBEC monolayer that are restricted to the basolateral membrane. Its effects can be further modulated through changes in cAMP and cGMP second messenger systems.
Collapse
Affiliation(s)
- J B Mackic
- Department of Neurological Surgery, USC School of Medicine, Los Angeles, California 90033, USA
| | | | | | | | | | | |
Collapse
|
15
|
Upregulation of tumor necrosis factor alpha transport across the blood-brain barrier after acute compressive spinal cord injury. J Neurosci 1999. [PMID: 10212323 DOI: 10.1523/jneurosci.19-09-03649.1999] [Citation(s) in RCA: 68] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Tumor necrosis factor alpha (TNF) is a cytokine that is involved in the inflammatory process after CNS injury and is implicated in neuroregeneration. A saturable transport system for TNF located at the blood-brain barrier (BBB) is responsible for the limited entry of TNF from blood to the CNS in normal mice. After partial disruption of the BBB by compression of the lumbar spinal cord, permeability to TNF was increased not only in the lumbar spinal cord but also in brain and distal spinal cord segments, where the BBB remained intact. The increase in the entry of TNF to the CNS followed a biphasic temporal pattern, with a first peak immediately after injury and a second peak starting on day 3; these changes lasted longer than the mere disruption of the BBB. The increased entry of TNF was abolished by addition of excess unlabeled TNF, showing that the transport system for TNF remained saturable after spinal cord injury (SCI) and providing evidence that the enhanced entry of TNF could not be explained by diffusion or leakage. This study adds strong support for our concept that the saturable transport system for TNF across the BBB can be upregulated in the diseased state, and it suggests that the BBB is actively involved in the modulation of the processes of degeneration and regeneration after SCI.
Collapse
|
16
|
Abstract
Pharmacological therapy, present and future, will undoubtedly continue to play a large role within the overall management of patients with severe head injury. Nevertheless, limited clinical data are available to evaluate the effect of severe head injury on pharmacokinetics. The disruption of the blood-brain barrier secondary to trauma and/or subsequent hyperosmolar therapy can be expected to result in higher than expected brain drug concentrations. Aggressive dietary protein supplementation may result in increased oxidative drug metabolism. These effects may counterbalance inhibitory influences on drug metabolism secondary to cytokine release during the acute phase response. Alterations in protein binding can also be anticipated with the hypoalbuminaemia and increases in alpha 1-acid glycoprotein typically observed in these patients. Based on studies in other patient populations, moderate hypothermia, a treatment strategy in patients with head injury, can decrease drug metabolism. The pharmacokinetics of the following drugs in patients with severe head injury have been studied: phenytoin, pentobarbital (pentobarbitone), thiopental (thiopentone), tirilazad, and the agents used as marker substrates, antipyrine, lorazepam and indocynanine green (ICG). Several studies have documented increase in metabolism over time with phenytoin, pentobarbital, thiopental, antipyrine and lorazepam. Increases in tirilazad clearance were also observed but attributed to concurrent phenytoin therapy. No changes in the pharmacokinetics of ICG were apparent following head injury. With the frequent use of potent inhibitors of drug metabolism (e.g., cimetidine, ciprofloxacin) the potential for drug interaction is high in patients with severe head injury. Additional pharmacokinetic investigations are recommended to optimise pharmacological outcomes in patients with severe head injury.
Collapse
Affiliation(s)
- B A Boucher
- Department of Clinical Pharmacy, University of Tennessee, Memphis, USA.
| | | |
Collapse
|
17
|
Abstract
The microvessel wall is a barrier for the delivery of various therapeutic agents to tumor cells. Tumor microvessels are, in general, more permeable to macromolecules than normal vessels. The hyperpermeability is presumably due to the existence of large pore structures in the vessel wall, induced by various cytokines. The cutoff pore size is tumor dependent, as determined by transport studies of nanoparticles. The vascular permeability is heterogeneous in tumors and dependent on physicochemical properties of molecules as well as the ultrastructure of the vessel wall. The ultrastructure is dynamic and can be modulated by the tumor microenvironment. The microenvironment itself can be altered by the transvascular transport because the transport may facilitate angiogenesis, reduce blood flow, and induce interstitial hypertension in tumors. Future studies of transport need to address mechanisms of the barrier formation and emphasize development of novel strategies for circumventing or exploiting the vascular barrier.
Collapse
Affiliation(s)
- F Yuan
- Department of Biomedical Engineering, Duke University, Durham, NC 27708, USA
| |
Collapse
|
18
|
Pan W, Banks WA, Kastin AJ. Blood-brain barrier permeability to ebiratide and TNF in acute spinal cord injury. Exp Neurol 1997; 146:367-73. [PMID: 9270046 DOI: 10.1006/exnr.1997.6533] [Citation(s) in RCA: 81] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Spinal cord injury (SCI) in mammals has a poor outcome because of a lack of regeneration. Alteration of the local environment after injury may induce regeneration. However, the passage of blood-borne or exogenous neurotrophic substances through the blood-brain barrier (BBB) is not well characterized in either normal or injured states. We investigated the permeability of the BBB in normal and injured states to two markers of permeability (albumin and sucrose), to a peptide (ebiratide), and to a cytokine [tumor necrosis factor-alpha(TNF)]. We found that in normal mice the cervical and lumbar areas of the spinal cord were more permeable than the thoracic area and the brain to all four substances. The penetration of the alpha-MSH/ACTH analogue ebiratide and of TNF, substances that have saturable transport systems across the BBB and may be involved in regenerative processes in the CNS, followed a regional pattern of differential permeability comparable to that of albumin and sucrose. Complete transection at the lumbar level induced a temporal change in the permeability of the BBB. The increased permeability, as measured by the radioactively labeled tracers albumin and sucrose, was most apparent in the lumbar region proximal to the transection. After SCI, the permeability to ebiratide remained unchanged, suggesting that disruption of the BBB did not affect the transport system for ebiratide. By contrast, the increase of permeability to TNF exceeded that detected by the markers albumin and sucrose. This enhanced permeability was inhibited by excess unlabeled TNF in the blood, showing saturability. This suggests that the transport system for TNF may be activated in SCI.
Collapse
Affiliation(s)
- W Pan
- VA Medical Center and Tulane University School of Medicine, New Orleans, Louisiana 70146, USA
| | | | | |
Collapse
|
19
|
Preston E, Foster DO. Evidence for pore-like opening of the blood-brain barrier following forebrain ischemia in rats. Brain Res 1997; 761:4-10. [PMID: 9247060 DOI: 10.1016/s0006-8993(97)00323-5] [Citation(s) in RCA: 37] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
The nature of the delayed blood-brain barrier (BBB) opening that occurs in rats subjected to forebrain ischemia by the technique of two-vessel (carotid) occlusion plus hypovolemic hypotension (2VO ischemia) was probed by examining the simultaneous, trans-barrier movement of two hydrophilic, normally poorly permeative solutes of markedly different molecular size: sucrose (MW = 342) and inulin (MW approximately 5000). Pentobarbital-anesthetized, male, Sprague-Dawley rats (342-374 g) were subjected to 10 min of 2VO ischemia (tympanic temperature, 37.5-38.0 degrees C); 6 h later they were reanesthetized and, along with non-ischemic controls, injected i.v. with [14C]sucrose and [3H]inulin. Transfer constants (Kis) for blood-to-brain movement of the tracers and Vis (apparent initial volumes of tracer distribution) were determined for six brain regions by the multiple-time, graphical method (tracer circulation times from 3 to 30 min). Vis differed little or insignificantly between the two tracers, or between control and post-ischemic rats; the values did not suggest appreciable endothelial binding of either tracer that might lead to its uptake by adsorptive-phase endocytosis. In the controls, regional Kis +/- S.E.M. (nl g(-1) s(-1)) for inulin ranged from 0.18 +/- 0.04 to 0.31 +/- 0.09 and were significantly lower (P < 0.01) than Kis for sucrose (1.53 +/- 0.16-1.91 +/- 0.29). The Ki ratio (sucrose/inulin) across brain regions (mean, 6.6; S.E.M., 0.6) was much lower than would be expected according to the concept that movement of most organic non-electrolytes across the intact BBB occurs by dissolution in and diffusion through endothelial cell plasma membranes, at a rate proportional to the lipid solubility and diffusivity of the solute. This finding is interpreted as indicating that a portion of the transfer of sucrose and inulin occurred by a mechanism other than dissolution-diffusion (e.g., via pores or vesicles). In the post-ischemic rats, Kis for both tracers were elevated significantly (P < 0.01) in parietal cortex, striatum, hippocampus, and midbrain. The post-ischemic increases (delta Kis) in these regions were greater for sucrose (1.90-3.31 nl g(-1) s(-1)) than for inulin (0.80-1.33). Across brain regions the ratio between sucrose delta Ki and inulin delta Ki averaged 2.9 (S.E.M., 0.2), a value significantly greater than the ratio of 1 that would be expected were the BBB opening due to an enhancement of micropinocytosis and vesicular transport. The correspondence of the mean delta Ki ratio with the ratio of the free diffusion coefficients of the tracers (D(f, suc)/D(f, inu) = 2.9; water, 38 degrees C) suggests that the delayed opening of the BBB following 2VO ischemia involves the formation of trans- or paracellular, aqueous pores or channels.
Collapse
Affiliation(s)
- E Preston
- Institute for Biological Sciences, National Research Council of Canada, Ottawa, Ont.
| | | |
Collapse
|
20
|
Pan W, Banks WA, Kennedy MK, Gutierrez EG, Kastin AJ. Differential permeability of the BBB in acute EAE: enhanced transport of TNT-alpha. THE AMERICAN JOURNAL OF PHYSIOLOGY 1996; 271:E636-42. [PMID: 8897850 DOI: 10.1152/ajpendo.1996.271.4.e636] [Citation(s) in RCA: 50] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Impairment of the blood-brain barrier (BBB) in experimental autoimmune encephalomyelitis (EAE) has been frequently attributed to disruption, without much consideration of saturable transport processes. In mice with EAE, we studied the permeability of the BBB to radioactively labeled albumin and sucrose, markers of BBB disruption, and tumor necrosis factor-alpha (TNF-alpha), a cytokine transported across the BBB by a saturable system and thought to play a role in the pathogenesis of EAE. Permeation of the BBB was increased to all three substances during the acutely ill stage, was greatest in the lumbar spine, and returned to normal with recovery. The change in BBB permeability to sucrose was greater than to the larger albumin and is consistent with a partial disruption of the BBB. The enhanced permeability to TNF-alpha was comparable to that for sucrose, even though TNF-alpha is similar in size to albumin. This paradoxically high uptake of TNF-alpha could be explained by an enhancement of its endogenous saturable transport system. Thus the changes in BBB function during EAE extend beyond disruption to include changes in the saturable transport systems for substances involved in the disease process.
Collapse
Affiliation(s)
- W Pan
- Department of Neuroscience, Tulane University School of Medicine, New Orleans, Louisiana, USA
| | | | | | | | | |
Collapse
|
21
|
Başkaya MK, Rao AM, Puckett L, Prasad MR, Dempsey RJ. Effect of difluoromethylornithine treatment on regional ornithine decarboxylase activity and edema formation after experimental brain injury. J Neurotrauma 1996; 13:85-92. [PMID: 9094379 DOI: 10.1089/neu.1996.13.85] [Citation(s) in RCA: 25] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
This study examined the effect of difluoromethylornithine (DFMO) on regional activities of ornithine decarboxylase (ODC) and edema formation in bilateral cerebral cortex and hippocampus after a unilateral controlled cortical-impact (CCI) injury in rats. To measure the activity of ODC, the brains of injured and control rats were frozen in situ at 30 min, 3, 6, and 24 h after CCI brain injury of moderate severity. The specific gravity, an indicator of edema formation, was examined in decapitated animals at corresponding time points. Brain injury induced significant increases of ODC in the ipsilateral hippocampus, adjacent and injury-site cortices, and in the contralateral cortex and hippocampus at 3 and 6 h after injury. No significant edema formation was found in any brain region at 30 min after injury. A significant edema formation was first found only in the injury-site cortex at 3 h after injury. At 6 and 24 h after injury, significant edema was found in all regions ipsilateral to the injury-site. At 24 h after injury, significant but less severe edema was also found in the contralateral cortex and hippocampus. DFMO, an irreversible inhibitor of ODC, abolished the increase in ODC in all regions. It also attenuated edema formation in the adjacent cortex and in the contralateral cortex and hippocampus. These findings indicate that polyamines may play a role in posttraumatic brain edema formation, particularly in important brain regions remote from the injury-site.
Collapse
Affiliation(s)
- M K Başkaya
- Department of Surgery, University of Kentucky Medical Center, Lexington, 40536-0084, USA
| | | | | | | | | |
Collapse
|
22
|
|
23
|
Cloughesy TF, Black KL. Pharmacological blood-brain barrier modification for selective drug delivery. J Neurooncol 1995; 26:125-32. [PMID: 8787854 DOI: 10.1007/bf01060218] [Citation(s) in RCA: 48] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Vasoactive agents have been identified through studies of peritumoral edema and effects on systemic capillaries. Abnormal blood-brain barrier or blood-tumor barrier can develop transient increases in permeability with the intraarterial delivery of vasoactive agents. Normal blood-brain barrier resists the effects of these compounds because of a biochemical barrier that may inactivate or become inert to vasoactive agents. Vasoactive compounds, including leukotrienes, bradykinin, and histamine appear to selectively increase permeability in abnormal brain capillaries. Intracarotid infusion of leukotrienes, bradykinin, and other vasoactive agents can increase drug delivery to diseased tissue.
Collapse
|
24
|
|
25
|
Inamura T, Nomura T, Bartus RT, Black KL. Intracarotid infusion of RMP-7, a bradykinin analog: a method for selective drug delivery to brain tumors. J Neurosurg 1994; 81:752-8. [PMID: 7931623 DOI: 10.3171/jns.1994.81.5.0752] [Citation(s) in RCA: 123] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
The bradykinin analog, RMP-7, was investigated for its ability to selectively increase uptake of molecular tracers in RG2 glial tumors. When infused in low doses (0.1 microgram/kg/min) through the intracarotid artery ipsilateral to RG2 gliomas in rats, RMP-7 significantly increased the permeability of tumor capillaries to methotrexate and to four other tracers of varying molecular weights, compared to intracarotid infusion of vehicle alone. Tracers used to examine permeability included radiolabeled alpha-aminoisobutyric acid (M(r) 103 D), sucrose (M(r) 342 D), methotrexate (M(r) 454.5 D), inulin (M(r) 5000 D), and dextran (M(r) 70,000 D). Permeability was expressed as the unidirectional transfer constant, Ki (microliters/gm/min). The permeability (Ki) of tumors in the RMP-7 group compared to the vehicle control group was as follows: alpha-aminoisobutyric acid, 35.3 +/- 9.11 versus 12.7 +/- 4.56 (p < 0.001); sucrose, 16.5 +/- 3.83 versus 9.28 +/- 3.12 (p < 0.05); methotrexate, 26.3 +/- 10.3 versus 8.98 +/- 6.78 (p < 0.005); inulin, 13.5 +/- 3.23 versus 6.55 +/- 4.32 (p < 0.005); dextran, 15.2 +/- 3.42 versus 1.47 +/- 1.24 (p < 0.001). The permeability of RG2 gliomas to high-molecular-weight dextran (70,000 D) was 10.3-fold higher in the RMP-7 group than in the vehicle control group. Intracarotid infusion of RMP-7 did not significantly increase the blood volume in tumor or brain tissue. The permeability of normal brain capillaries was unaffected by intracarotid infusion of 0.1 microgram/kg/min RMP-7 relative to that achieved in tumor. These data support the idea that intracarotid infusion of RMP-7 will be a useful technique for selective delivery of antitumor compounds to brain tumors.
Collapse
Affiliation(s)
- T Inamura
- Brain Research Institute, Jonsson Comprehensive Cancer Center, University of California, Los Angeles, Medical Center
| | | | | | | |
Collapse
|
26
|
Abstract
Bradykinin, infused in low doses (10 micrograms/kg/min) through the carotid artery ipsilateral to RG2 glioma in rats, significantly increased the permeability in tumor capillaries to six different tracers of varying molecular weights compared with intracarotid infusion of saline alone. Permeability in normal brain capillaries was not significantly increased by intracarotid bradykinin infusion. Tracers used to examined permeability included radiolabeled alpha-aminoisobutyric acid (AIB; MW 103), sucrose (MW 342.3), inulin (MW 5000), and dextran (MW 70,000), horseradish peroxidase (HRP) and Evans blue (EB). Permeability was expressed as the unidirectional transfer constant K(i) (microliter/g/min). The permeabilities (K(i)) of tumors in the bradykinin group versus the control saline group for AIB, sucrose, inulin, and dextran were 25.91 +/- 6.78 vs. 13.95 +/- 4.29 (p < 0.01), 17.90 +/- 2.65 vs. 10.75 +/- 4.55 (p < 0.01), 23.92 +/- 6.99 vs. 6.20 +/- 4.37 (p < 0.01), and 17.84 +/- 1.00 vs. 1.47 +/- 1.24 (p < 0.001), respectively (mean +/- SD). Permeability of RG2 gliomas to high molecular weight dextran (70,000) was 12-fold higher in the bradykinin group than in the saline infusion group. Intracarotid infusion of bradykinin did not significantly increase the blood volume in tumor or brain tissue despite its known vasodilative effect. The permeability of normal brain capillaries was unaffected by intracarotid bradykinin infusion. The increased permeability was reversed 20 min after stopping the intracarotid infusion. Electron microscopic and gross qualitative analysis was performed using HRP and EB. Intracarotid bradykinin infusion increased HRP and EB within tumor tissue but not normal tissue.(ABSTRACT TRUNCATED AT 250 WORDS)
Collapse
Affiliation(s)
- T Inamura
- Brain Research Institute, Jonsson Comprehensive Cancer Center, UCLA Medical Center 90024
| | | |
Collapse
|
27
|
Osmotic Opening of the Blood-Brain Barrier and Brain Tumor Chemotherapy. ACTA ACUST UNITED AC 1994. [DOI: 10.1016/b978-0-12-185291-7.50009-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register]
|
28
|
Schmitz MP, Combs DJ, Dempsey RJ. Difluoromethylornithine decreases postischemic brain edema and blood-brain barrier breakdown. Neurosurgery 1993; 33:882-7; discussion 887-8. [PMID: 8264888 DOI: 10.1227/00006123-199311000-00016] [Citation(s) in RCA: 26] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
Abstract
Brain polyamines have been associated with posttraumatic vasogenic edema and blood-brain barrier (BBB) breakdown seen in some models of brain injury. We hypothesized that the inhibition of the enzyme responsible for polyamine production with the decarboxylase difluoromethylornithine (DFMO) may decrease BBB breakdown after a focal brain ischemic stroke. Thirty-two cats underwent 8 hours of middle cerebral artery occlusion and one of four treatments: sham operation (Sham), ischemia (Isc), ischemia/DFMO (Isc/DF), and ischemia/DFMO/putrescine (Isc/DF/PU). The regional brain specific gravity and the volume of Evans blue (EB) extravasation were measured at the time of death. The groups were monitored for temperature, heart rate, blood pressure, and arterial blood gases, and the values did not differ outside normal physiological ranges. EB results were expressed as the percentage of the hemisphere stained and showed the following: Sham, 2.23%; Isc, 32.8%; Isc/DF, 5.6%; Isc/DF/PU, 36.3%. As a measure of BBB, ischemia increased EB staining; DFMO pretreatment decreased the amount of EB staining to control levels; and the polyamine putrescine abolished the protective effect of DFMO (all significant at P = 0.05). DFMO pretreatment also resulted in a significant (P = 0.05) return to control values for specific gravity in the EB-stained regions (1.0328) of ischemic animals. This effect was present primarily in the white matter. Treatment with DFMO, an ornithine decarboxylase inhibitor, significantly decreased postischemic BBB breakdown and vasogenic edema in this model.
Collapse
Affiliation(s)
- M P Schmitz
- Department of Surgery, University of Kentucky, Chandler Medical Center, Lexington
| | | | | |
Collapse
|
29
|
Difluoromethylornithine Decreases Postischemic Brain Edema and Blood-Brain Barrier Breakdown. Neurosurgery 1993. [DOI: 10.1097/00006123-199311000-00016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
|
30
|
Ziylan ZY, Bernard GC, Lefauconnier JM, Durand GA, Bourre JM. Effect of dietary n-3 fatty acid deficiency on blood-to-brain transfer of sucrose, alpha-aminoisobutyric acid and phenylalanine in the rat. Neurosci Lett 1992; 137:9-13. [PMID: 1625821 DOI: 10.1016/0304-3940(92)90286-g] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Possible alterations in blood-to-brain unidirectional transport of sucrose (mol. wt., 342), alpha-aminoisobutyric acid (mol. wt., 104), and L-phenylalanine (mol. wt., 165) induced by a diet deficient in n-3 polyunsaturated fatty acids were studied with respect to blood-brain barrier function. Two groups of rats were for to two generations with a semisynthetic diet. One group of rats was fed a peanut oil+rapeseed oil diet which contained both essential fatty acids: linoleic acid (18:2 n-6) and alpha-linolenic acid, (18:3 n-3). Another group was fed a diet of peanut oil, this diet (containing 18:2 n-6) was deficient in alpha-linolenic acid. The experiments were performed at 6 months of age. Unidirectional transfer rate constants (Ki) of sucrose, alpha-aminoisobutyric acid and L-phenylalanine were measured. The diet based on peanut oil (deficient in n-3) caused a greater blood-to-brain transport of sucrose but not of alpha-aminoisobutyric acid or L-phenylalanine. These observations indicate that regardless of the mechanisms involved, alterations in essential fatty acids induced by diet can modulate to some extent the blood-brain transport of hydrophilic molecules without a carrier.
Collapse
Affiliation(s)
- Z Y Ziylan
- Department of Physiology, Istanbul Faculty of Medicine, Turkey
| | | | | | | | | |
Collapse
|
31
|
Ziylan YZ, Lefauconnier JM, Ates N, Bernard G, Bourre JM. Age-dependent alteration in regional cerebrovascular permeability during drug-induced epilepsy. Mech Ageing Dev 1992; 62:319-27. [PMID: 1583915 DOI: 10.1016/0047-6374(92)90116-u] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Age-related changes in blood-brain barrier permeability were investigated during pentylenetetrazol-induced seizures in rats aged from 15 days to 120 days. Tracers such as [14C]sucrose and [3H]inulin which diffuse very slowly across the intact endothelium were simultaneously injected i.v. in rats treated with pentylenetetrazol (PTZ) or in control animals. Permeability-surface area products (PA) were determined in 9 brain regions. Pentylenetetrazol-induced seizures caused a significant increase in PA for both sucrose and inulin in all brain regions studied. Blood-brain barrier dysfunction was present only in animals in which the mean arterial blood pressure rose at seizure onset. Although increased blood-brain barrier permeability was found partly in similar areas in both young and adult rat brains, in adults the increase was the highest in the preoptic area, septum, colliculus inferior, hypothalamus and in the cerebellum while the increase was comparatively much smaller in the same areas of young brains. The increase in blood-brain barrier permeability was extremely high in the hippocampus, hypothalamus and cerebellum of 15-day-old rat brain and, was least affected in the corpus striatum and cerebral cortex in contrast to older rats. From the results obtained it may be concluded that the increased cerebrovascular permeability induced by pentylenetetrazol differs markedly in localization in young and adult rats. The age-dependent increased blood-brain barrier integrity is not over all dependent on variations in the blood pressure, but rather on progressive maturation of capillaries and changes in their internal structure, and local phenomena in neuronal activity during the seizures.
Collapse
Affiliation(s)
- Y Z Ziylan
- Department of Physiology, Istanbul Faculty of Medicine, Capa, Turkey
| | | | | | | | | |
Collapse
|
32
|
Abstract
1. Experimental determinations of blood-brain barrier permeability from measurements of uptake of tracers by brain are limited in two ways. First, there are experimental limitations that are specific to the particular method being used. These limitations include the range of permeability values that a particular method can reliably determine (sensitivity); whether repeated experiments are possible in the same animal; whether regional values can be determined; and to what degree the chemical composition of the perfusate passing through the brain can be controlled. 2. A second set of limitations on permeability determinations is of a more general, physiological nature. These constraints apply to a greater or lesser degree to all experimental determinations, and may limit the accuracy and reliability of the permeability values obtained; although certain reliable upper and lower bounds can be determined. These general factors include: capillary heterogeneity (i.e. variations in capillary lengths, surface areas, blood flows, etc.); the possibility of binding to circulating plasma proteins, especially albumin; and the possibility of capillary recruitment and de-recruitment with changes in cerebral blood flow.
Collapse
Affiliation(s)
- P J Robinson
- Laboratory of Neurosciences, National Institute on Aging, National Institutes of Health, Bethesda, Maryland
| |
Collapse
|
33
|
Ziylan YZ, Lefauconnier JM, Bernard G, Bourre JM. Blood-brain barrier permeability: regional alterations after acute and chronic administration of ethinyl estradiol. Neurosci Lett 1990; 118:181-4. [PMID: 2274267 DOI: 10.1016/0304-3940(90)90621-f] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
In this study we measured the effect of acute and chronic estrogen treatment on cerebrovascular permeability to sucrose and inulin. Animals were subcutaneously injected once with 0.1 micrograms/rat of ethinyl estradiol or injected daily with the same drug dose for 3 weeks. Control rats received the same amount of arachis oil vehicle. Three weeks treatment but not the single injection of ethinyl estradiol produced significant increases in the cerebrovascular permeability-surface area product for sucrose and inulin in almost all brain regions.
Collapse
Affiliation(s)
- Y Z Ziylan
- Department of Physiology, Istanbul Faculty of Medicine, Turkey
| | | | | | | |
Collapse
|
34
|
Robinson PJ, Rapoport SI. Model for drug uptake by brain tumors: effects of osmotic treatment and of diffusion in brain. J Cereb Blood Flow Metab 1990; 10:153-61. [PMID: 2303532 DOI: 10.1038/jcbfm.1990.30] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
A mathematical model describing drug uptake into brain tumors, directly from blood and indirectly from neighboring tissue, is presented. The model quantitatively describes uptake into tumor, brain surrounding tumor (BST), and normal brain and uptake following reversible osmotic blood-brain barrier (BBB) and blood-tumor barrier disruption. It employs published data on the time course for reclosure of the BBB following osmotic treatment and on the brain and tumor uptake of [14C]alpha-aminoisobutyric acid by Walker 256 carcinomas and C6 gliomas implanted into the rat brain. Constant infusion and bolus injection infusion schedules are considered. In untreated brain, the BST acts as a sink, reducing the integrated exposure of the adjacent tumor to the drug, whereas following osmotic treatment, tumor exposure to drug is enhanced, not only by increased delivery from blood but also by diffusion (and bulk flow) from neighboring brain. The model provides a quantitative framework for examining the efficacy of osmotic treatment to enhance chemotherapy of brain tumors.
Collapse
Affiliation(s)
- P J Robinson
- Laboratory of Neurosciences, National Institute on Aging, National Institutes of Health, Bethesda, Maryland
| | | |
Collapse
|
35
|
Armstrong BK, Smith Q, Rapoport SI, Strohalm J, Kopeček J, Duncan R. Osmotic opening of the blood-brain barrier permeability to N-(2-hydroxypropyl)methacrylamide copolymers. Effect of polymer -Mw charge and hydrophobicity. J Control Release 1989. [DOI: 10.1016/0168-3659(89)90015-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
|
36
|
Ziylan YZ, Lefauconnier JM, Bernard G, Bourre JM. Regional alterations in blood-to-brain transfer of alpha-aminoisobutyric acid and sucrose, after chronic administration and withdrawal of dexamethasone. J Neurochem 1989; 52:684-9. [PMID: 2918309 DOI: 10.1111/j.1471-4159.1989.tb02509.x] [Citation(s) in RCA: 24] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
The effect of dexamethasone administration and withdrawal was studied with respect to blood-brain barrier function. The tracers alpha-[3H]aminoisobutyric acid (AIB) (MW 104) and [14C]sucrose (MW 342), which have a low permeability across the intact endothelium, were simultaneously injected intravenously in rats treated with dexamethasone and placebo-treated control animals or in rats in which dexamethasone treatment was discontinued 3 days before the experiment. Unidirectional transfer constants (Ki) were determined in discrete brain regions. Steroid administration reduced the rate of influx of AIB and sucrose, whereas discontinuation of drug resulted in an increased permeability. These findings suggest that when exposure to glucocorticoids is prolonged, the efficiency of medical treatment of CNS diseases may decrease due to reduction of drug delivery to CNS. Thus, these experimental findings may have particular importance in the clinical setting of drug administration when considering the combination of steroids with other drugs, and may aid in understanding better the pathogenesis of some types of brain edema seen in patients from whom corticosteroid therapy has been withdrawn.
Collapse
Affiliation(s)
- Y Z Ziylan
- Unité de Neurotoxicologie, INSERM U. 26, Hôpital Fernand Widal, Paris, France
| | | | | | | |
Collapse
|
37
|
Ziylan YZ, LeFauconnier JM, Bernard G, Bourre JM. Effect of dexamethasone on transport of alpha-aminoisobutyric acid and sucrose across the blood-brain barrier. J Neurochem 1988; 51:1338-42. [PMID: 3171581 DOI: 10.1111/j.1471-4159.1988.tb01094.x] [Citation(s) in RCA: 48] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
The effect of glucocorticoids on the blood-brain barrier (BBB) was studied in rats following a single injection or 3 days of dexamethasone administration. Tracers with a low permeability across the intact endothelium, [14C]sucrose and alpha-[3H]aminoisobutyric acid ([3H]AIB), were simultaneously injected intravenously in untreated rats or in rats treated with dexamethasone. Unidirectional blood-to-brain transfer constants (Ki) in 14 regions of the rat brain were determined. In regions of control brain, average Ki values for AIB and sucrose were approximately 0.0020 and 0.00060 ml g-1 min-1, respectively. The lowest transfer constants were found in caudate nucleus, hippocampus, white matter, and cerebellum. In dexamethasone-treated animals, Ki values for both sucrose and AIB markedly decreased by 30-50% in almost all brain regions. These results indicate that a single injection or 3 days of treatment with dexamethasone causes an apparent reduction in the normal BBB permeability, and dexamethasone may greatly interfere with drug delivery into brain. These observations may have an importance for the administration of drugs in brain disease in the presence of steroids.
Collapse
Affiliation(s)
- Y Z Ziylan
- Unité de Neurotoxicologie, INSERM U. 26, Hôpital Fernand Widal, Paris, France
| | | | | | | |
Collapse
|
38
|
Robinson PJ. Facilitation of drug entry into brain by osmotic opening of the blood-brain barrier. Clin Exp Pharmacol Physiol 1987; 14:887-901. [PMID: 3329074 DOI: 10.1111/j.1440-1681.1987.tb02425.x] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
1. After osmotic opening, the blood-brain barrier (BBB) has been shown to reclose more rapidly to large than to small neutral, water-soluble molecules. Quantitative analysis of these data supports the creation of interendothelial pores with radii of about 200 A through which such molecules pass by both restricted diffusion and bulk flow (with solute drag) from blood to brain. 2. The major reduction in BBB permeability from 6 to 35 min following osmotic opening seems to be due to a reduction in bulk flow by a factor of about 10, rather than marked decreases in pore densities or effective pore size. On this basis, quantitative predictions of brain uptake of neutral, water soluble substances are made for various times after osmotic opening of the BBB, as a function of molecular size. 3. Implications of these results are discussed for enhancement of uptake of drugs, including enzymes and certain anti-cancer agents, by the brain. 4. The idea of a 'therapeutic window' as the period of time, following reversible osmotic opening, during which the permeability of the BBB is enhanced significantly for a particular compound, is introduced. Since the BBB is normally highly impermeable to plasma proteins, the effect of BBB opening on the uptake of highly protein-bound drugs is discussed briefly. 5. The effect of molecular charge on the passage of molecules through interendothelial pores into the brain is also discussed.
Collapse
Affiliation(s)
- P J Robinson
- Department of Mathematics, University of Queensland, St Lucia, Australia
| |
Collapse
|
39
|
Warnke PC, Blasberg RG, Groothuis DR. The effect of hyperosmotic blood-brain barrier disruption on blood-to-tissue transport in ENU-induced gliomas. Ann Neurol 1987; 22:300-5. [PMID: 3118762 DOI: 10.1002/ana.410220304] [Citation(s) in RCA: 47] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Hyperosmotic blood-brain barrier disruption transiently increases the rate of blood-to-tissue transport of water-soluble compounds to normal brain and has been used in brain tumor patients to increase the delivery of chemotherapeutic drugs. This method remains controversial; debate concerns the extent to which it increases drug delivery to brain tumors. Ethylnitrosourea (ENU)-induced gliomas in rats have the lowest rate of blood-to-tissue transfer of the water-soluble compound alpha-aminoisobutyric acid of all experimental brain tumors studied to date. To gain further understanding about the effects of hyperosmotic blood-brain barrier disruption in brain tumors, we measured the unidirectional blood-to-tissue transfer constant of alpha-aminoisobutyric acid in ENU-induced brain tumors in rats after hyperosmotic disruption. Hyperosmotic blood-brain barrier disruption with 1.6 osmolar mannitol resulted in an averaged whole-tumor transfer constant of 0.013 +/- 0.003 (standard error) mL/(g/min), compared to a transfer constant of 0.007 +/- 0.002 mL/(g/min) for ENU-induced gliomas in the contralateral undisrupted hemisphere, a difference that was not significant. In contrast, hyperosmotic blood-brain barrier disruption produced a large and significant increase in the transfer constant of alpha-aminoisobutyric acid in tumor-free cortex (from 0.002 +/- 0.001 to 0.05 +/- 0.011 mL/[g/min]) and in tumor-free corpus callosum (from 0.001 +/- 0.003 to 0.017 +/- 0.005 mL/[g/min]). Hyperosmotic blood-brain barrier disruption does not appear to be an efficient method with which to increase the rate of delivery of water-soluble drugs to brain tumors but does result in a significant increase in the delivery rate of these drugs to normal brain tissue.
Collapse
Affiliation(s)
- P C Warnke
- Department of Neurology, Northwestern University Medical School, Chicago, IL
| | | | | |
Collapse
|
40
|
Armstrong BK, Robinson PJ, Rapoport SI. Size-dependent blood-brain barrier opening demonstrated with [14C]sucrose and a 200,000-Da [3H]dextran. Exp Neurol 1987; 97:686-96. [PMID: 2442027 DOI: 10.1016/0014-4886(87)90125-7] [Citation(s) in RCA: 20] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
The reclosure of the blood-brain barrier following osmotic opening was investigated by determining the product of permeability times surface area for two neutral, water-soluble compounds differing widely in molecular size. [14C]Sucrose (Mr 340 Da, radius 5A) and [3H]dextran (Mr 200,000 Da, radius 100 A) were simultaneously injected i.v., and their regional permeability times surface areas were calculated at 6, 10, 35, and 55 min after the blood-brain barrier was opened by a 30-s infusion of 1.8 m L(+)-arabinose into the right external carotid artery. The control permeability times surface area product was about 10(-5) cm3 s-1 g-1 brain for sucrose and negligible for dextran. It increased to 4 X 10(-4) cm3 s-1 g-1 brain and 10(-4) cm3 s-1 g-1 brain for sucrose and dextran, respectively, at 6 min after opening of the blood-brain barrier. Thereafter, permeability-surface area products for both substances declined. Dextran had significantly lower (P less than 0.05) values than sucrose at all times. The ratios of permeability times surface areas of [14C]sucrose to those to [3H]dextran were consistent with restricted diffusion through pores or slits at 35 and 55 min after blood-brain barrier opening, but at 6 and 10 min these ratios were less than the ratio of their free diffusion coefficients, indicative of bulk fluid flow with solute drag from blood to brain. A previously measured increase in brain water content following opening of the blood-brain barrier together with the present results, suggest the creation of slits approximately 400 A in width after osmotic treatment. Reduction in bulk fluid flow from blood to brain appears to be the major cause for the reduction of permeability times surface areas for both sucrose and dextran as the blood-brain barrier recloses.
Collapse
|
41
|
Rapoport SI, Robinson PJ. Tight-junctional modification as the basis of osmotic opening of the blood-brain barrier. Ann N Y Acad Sci 1986; 481:250-67. [PMID: 3468860 DOI: 10.1111/j.1749-6632.1986.tb27155.x] [Citation(s) in RCA: 97] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Osmotic opening of the blood-brain barrier (BBB) most likely is mediated by modification of interendothelial tight junctions, subsequent to shrinkage of cerebrovascular endothelial cells, and not by stimulation of transendothelial vesicular transport or by channel formation. This paper summarizes evidence for this conclusion: osmotic BBB opening is mediated by endothelial cell shrinkage, electron microscopy, with single or serial thin sections, demonstrates penetration of intravascular tracer into brain via tight junctional complexes, the BBB remains open after the brain is fixed, osmotic BBB opening is rapidly reversible, and is insensitive to phenothiazines, and BBB closure following osmotic treatment is size-dependent, indicative of a sieve (pore) mechanism with bulk flow. The entire mechanism of vesicular transport in normal tissue is, furthermore, in doubt, because vesicles that are found in tissue layers connected by tight junctions (e.g., frog nerve perineurium and capillary endoneurium) do not support macromolecular transport.
Collapse
|
42
|
Kim YS, Lee MH, Wisniewski HM. Aluminum induced reversible change in permeability of the blood-brain barrier to [14C]sucrose. Brain Res 1986; 377:286-91. [PMID: 3730864 DOI: 10.1016/0006-8993(86)90870-x] [Citation(s) in RCA: 28] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
To determine whether aluminum alters the permeability of the blood-brain barrier (BBB), 4 groups of rats were given an intraperitoneal injection of aluminum chloride, aluminum lactate, aluminum hydroxide or physiological saline. Two hours later, [14C]sucrose was injected, and brain radioactivity was measured from 5 different brain regions. The permeability capillary surface area (PA) was calculated by the dual compartment model (plasma-brain) proposed by Rapoport et al. The PAs for [14C]sucrose were significantly elevated in all brain regions of the animals injected with the aluminum chloride or aluminum lactate. However, the aluminum hydroxide group showed no BBB permeability change. In the second experiment, the reversibility of the aluminum induced BBB change was examined. PA was determined at 2, 4 or 24 h after exposure to aluminum lactate. Significant permeability changes were observed at 2 and 4 h after aluminum. However, the difference disappeared by 24 h. These findings indicate that exposure to a high level of aluminum alters the function of BBB in the rat and the aluminum induced BBB change is reversible. An increase in the blood aluminum level and time after exposure appear to be important factors associated with the BBB permeability change. Implications of the results are discussed in terms of aluminum's potential action on the endothelial cells.
Collapse
|
43
|
Todd NV, Picozzi P, Crockard HA. Quantitative measurement of cerebral blood flow and cerebral blood volume after cerebral ischaemia. J Cereb Blood Flow Metab 1986; 6:338-41. [PMID: 3711160 DOI: 10.1038/jcbfm.1986.57] [Citation(s) in RCA: 29] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
CBF obtained by the hydrogen clearance technique and cerebral blood volume (CBV) calculated from the [14C]dextran space were measured in three groups of rats subjected to temporary four-vessel occlusion to produce 15 min of ischaemia, followed by 60 min of reperfusion. In the control animals, mean CBF was 93 +/- 6 ml 100 g-1 min-1, which fell to 5.5 +/- 0.5 ml 100 g-1 min-1 during ischaemia. There was a marked early postischaemic hyperaemia (262 +/- 18 ml 100 g-1 min-1), but 1 h after the onset of ischaemia, there was a significant hypoperfusion (51 +/- 3 ml 100 g-1 min-1). Mean cortical dextran space was 1.58 +/- 0.09 ml 100 g-1 prior to ischaemia. Early in reperfusion there was a significant increase in CBV (1.85 +/- 0.24 ml 100 g-1) with a decrease during the period of hypoperfusion (1.33 +/- 0.03 ml 100 g-1). Therefore, following a period of temporary ischaemia, there are commensurate changes in CBF and CBV, and alterations in the permeability-surface area product at this time may be due to variations in surface area and not necessarily permeability.
Collapse
|
44
|
Preston E, Haas N. Defining the lower limits of blood-brain barrier permeability: factors affecting the magnitude and interpretation of permeability-area products. J Neurosci Res 1986; 16:709-19. [PMID: 3098986 DOI: 10.1002/jnr.490160411] [Citation(s) in RCA: 25] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Experimental alteration in the restricted permeability of the blood-brain barrier to polar, blood-borne molecules is often quantitated in the rat with use of 14C-sucrose or 3H-mannitol delivered as a test substance into the circulation. The underlying principle is to relate the quantity of saccharide that has permeated into brain parenchyma, after an arbitrary time period, to some index of the circulating tracer level. This study indicates that to correct the radioactivity level in the brain tissue for intravascular tracer, it is an erroneous practice to estimate the latter as the product of tissue blood volume and the tracer concentration measured in a systematic blood sample. Dissected brain tissue was found to have a lower hematocrit and thereby larger plasma/tracer compartment per unit blood volume than femoral arterial blood. It is further shown that, although commercially supplied stocks of 14C-sucrose or 3H-mannitol may contain only small quantities of radioactive impurities, their inclusion in injectates and preferential uptake into brain may cause significant overestimation of permeability to the parent tracer. It is also confirmed that magnitude of permeability-area (PA) products for permeation of purified sucrose or mannitol into brain varies inversely with the length of time allotted for tracer circulation in the bloodstream. This finding is at variance with the assumptions of a two-compartment (plasma/brain) diffusion model underlying such measurements and supports a recently published model for blood-to-brain transfer based on multiple uptake compartments in brain parenchyma. The factors compromising PA measurement identified in this study may partly underlie variations in PA values published from several laboratories that had been attributed to genetic differences in laboratory rats.
Collapse
|
45
|
Storm AJ, van der Kogel AJ, Nooter K. Effect of X-irradiation on the pharmacokinetics of methotrexate in rats: alteration of the blood-brain barrier. EUROPEAN JOURNAL OF CANCER & CLINICAL ONCOLOGY 1985; 21:759-64. [PMID: 4018118 DOI: 10.1016/0277-5379(85)90275-5] [Citation(s) in RCA: 25] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
This study was designed to evaluate the effects of brain irradiation on the permeability of the blood-brain barrier for methotrexate (MTX). Female WAG/Rij rats were cranially irradiated with a single dose of 20 Gy of 300 kV X-rays. At different times (1-15 days) after the exposure the rats were injected intravenously with MTX (25 mg/kg body wt). Irradiation had hardly any effect on the MTX concentrations in the plasma, heart and kidneys as determined by high-performance liquid chromatography. However, irradiation resulted in a significant increase of MTX (determined by 125I-radioimmunoassay) in brain tissue per gram wet weight (187.6 +/- 17.9 pmol/g vs 46.4 +/- 29.3 pmol/g in unirradiated brain). This change in permeability of the blood-brain barrier lasted for about 9 days. The MTX elimination from the irradiated brain was the same as that from the non-irradiated brain. This indicates that only the MTX uptake and not the elimination by the brain was affected by the irradiation treatment.
Collapse
|
46
|
Picozzi P, Todd NV, Crockard AH. The role of cerebral blood volume changes in brain specific-gravity measurements. J Neurosurg 1985; 62:704-10. [PMID: 2580960 DOI: 10.3171/jns.1985.62.5.0704] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Cerebral blood volume (CBV) was calculated in gerbils from specific-gravity (SG) changes between normal and saline-perfused brains. Furthermore, changes in CBV were investigated during ischemia using carbon-14-labeled dextran (MW 70,000) as an intravascular marker. Both data were used to evaluate the possible error due to a change in CBV on the measurement of ischemic brain edema by the SG method. The methodological error found was 0.0004 for a 100% CBV change. This error is insignificant, being less than the standard deviation in the SG measured for the gerbil cortex (SG 1.0494 +/- 0.0006). Thus, CBV changes are not responsible for the SG variations observed during the first phase of ischemia. These variations are better explained as an increase of brain water content during ischemia.
Collapse
|
47
|
Kessler RM, Goble JC, Bird JH, Girton ME, Doppman JL, Rapoport SI, Barranger JA. Measurement of blood-brain barrier permeability with positron emission tomography and [68Ga]EDTA. J Cereb Blood Flow Metab 1984; 4:323-8. [PMID: 6432806 DOI: 10.1038/jcbfm.1984.48] [Citation(s) in RCA: 37] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
Positron emission tomography (PET) was employed to examine time-dependent changes in blood-brain barrier (BBB) permeability to [68Ga]ethylenediaminetetraacetate (EDTA) in the rhesus monkey, following reversible barrier opening by intracarotid infusion of a hypertonic mannitol solution. The PET technique, when combined with measurements of plasma radioactivity, provided a quantitative measure of the cerebrovascular permeability-area product (PA) at different times following mannitol infusion. Hypertonic mannitol treatment reversibly increased PA to [68Ga]EDTA more than 10-fold; much of the barrier effect was over by 10 min after mannitol treatment. The results show that PET can be used to measure transient changes in BBB integrity in specific brain regions, under in vivo, noninvasive conditions.
Collapse
|
48
|
Koenig H, Goldstone AD, Lu CY. Blood brain barrier breakdown in brain edema following cold injury is mediated by microvascular polyamines. Biochem Biophys Res Commun 1983; 116:1039-48. [PMID: 6418170 DOI: 10.1016/s0006-291x(83)80247-2] [Citation(s) in RCA: 77] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
A focal freeze injury to rat cerebral cortex induces an early (less than 5 min) increase in brain ornithine decarboxylase activity and an accumulation of polyamines involving cerebral microvessels. This polyamine synthesis correlates with the abnormal increase in microvascular permeability, monitored by uptake of Evans Blue and sod. fluorescein. The ornithine decarboxylase inhibitor alpha-difluoromethylornithine suppressed the injury-induced increment in spermidine and spermine and microvascular permeability. Putrescine nullified alpha-difluoromethylornithine inhibition and restored microvessel spermidine and spermine and the pathological increase in microvascular permeability. These results indicate that polyamine synthesis is obligatory for blood-brain barrier breakdown. alpha-Difluoromethylornithine may be useful in the treatment of vasogenic brain edema.
Collapse
|