Generic placeholder image

Current Pharmaceutical Design

Editor-in-Chief

ISSN (Print): 1381-6128
ISSN (Online): 1873-4286

Review Article

The Blood-Brain Barrier Interface in Diabetes Mellitus: Dysfunctions, Mechanisms and Approaches to Treatment

Author(s): William A. Banks*

Volume 26, Issue 13, 2020

Page: [1438 - 1447] Pages: 10

DOI: 10.2174/1381612826666200325110014

Price: $65

Abstract

Diabetes mellitus (DM) is one of the most common diseases in the world. Among its effects are an increase in the risk of cognitive impairment, including Alzheimer’s disease, and blood-brain barrier (BBB) dysfunction. DM is characterized by high blood glucose levels that are caused by either lack of insulin (Type I) or resistance to the actions of insulin (Type II). The phenotypes of these two types are dramatically different, with Type I animals being thin, with low levels of leptin as well as insulin, whereas Type II animals are often obese with high levels of both leptin and insulin. The best characterized change in BBB dysfunction is that of disruption. The brain regions that are disrupted, however, vary between Type I vs Type II DM, suggesting that factors other than hyperglycemia, perhaps hormonal factors such as leptin and insulin, play a regionally diverse role in BBB vulnerability or protection. Some BBB transporters are also altered in DM, including P-glycoprotein, lowdensity lipoprotein receptor-related protein 1, and the insulin transporter as other functions of the BBB, such as brain endothelial cell (BEC) expression of matrix metalloproteinases (MMPs) and immune cell trafficking. Pericyte loss secondary to the increased oxidative stress of processing excess glucose through the Krebs cycle is one mechanism that has shown to result in BBB disruption. Vascular endothelial growth factor (VEGF) induced by advanced glycation endproducts can increase the production of matrix metalloproteinases, which in turn affects tight junction proteins, providing another mechanism for BBB disruption as well as effects on P-glycoprotein. Through the enhanced expression of the redox-related mitochondrial transporter ABCB10, redox-sensitive transcription factor NF-E2 related factor-2 (Nrf2) inhibits BEC-monocyte adhesion. Several potential therapies, in addition to those of restoring euglycemia, can prevent some aspects of BBB dysfunction. Carbonic anhydrase inhibition decreases glucose metabolism and so reduces oxidative stress, preserving pericytes and blocking or reversing BBB disruption. Statins or N-acetylcysteine can reverse the BBB opening in some models of DM, fibroblast growth factor-21 improves BBB permeability through an Nrf2-dependent pathway, and nifedipine or VEGF improves memory in DM models. In summary, DM alters various aspects of BBB function through a number of mechanisms. A variety of treatments based on those mechanisms, as well as restoration of euglycemia, may be able to restore BBB functions., including reversal of BBB disruption.

Keywords: Blood-brain barrier, diabetes mellitus, disruption, p-glycoprotein, therapeutic, BEC-monocyte adhesion.

[1]
Zimmet P, Alberti KG, Magliano DJ, Bennett PH. Diabetes mellitus statistics on prevalence and mortality: facts and fallacies. Nat Rev Endocrinol 2016; 12(10): 616-22.
[http://dx.doi.org/10.1038/nrendo.2016.105] [PMID: 27388988]
[2]
Krolewski AS, Warram JH. Natural history of diabetes mellitus. Principles and practice of endocrinology and metabolism. Philadelphia: lippincott williams & wilkins.. 2001; 1320-7.
[3]
Banting FG, Best CH. The internal secretion of the pancreas. 1922. Indian J Med Res 2007; 125(3): 251-66.
[PMID: 17582843]
[4]
Licinio J, Caglayan S, Ozata M, et al. Phenotypic effects of leptin replacement on morbid obesity, diabetes mellitus, hypogonadism, and behavior in leptin-deficient adults. Proc Natl Acad Sci USA 2004; 101(13): 4531-6.
[http://dx.doi.org/10.1073/pnas.0308767101] [PMID: 15070752]
[5]
Chen H, Charlat O, Tartaglia LA, et al. Evidence that the diabetes gene encodes the leptin receptor: identification of a mutation in the leptin receptor gene in db/db mice. Cell 1996; 84(3): 491-5.
[http://dx.doi.org/10.1016/S0092-8674(00)81294-5] [PMID: 8608603]
[6]
Butler PC, Chou J, Carter WB, et al. Effects of meal ingestion on plasma amylin concentration in NIDDM and nondiabetic humans. Diabetes 1990; 39(6): 752-6.
[http://dx.doi.org/10.2337/diab.39.6.752] [PMID: 2189768]
[7]
Neuwelt E, Abbott NJ, Abrey L, et al. Strategies to advance translational research into brain barriers. Lancet Neurol 2008; 7(1): 84-96.
[http://dx.doi.org/10.1016/S1474-4422(07)70326-5] [PMID: 18093565]
[8]
Ginter E, Simko V. Type 2 diabetes mellitus, pandemic in 21st century. Adv Exp Med Biol 2012; 771: 42-50.
[http://dx.doi.org/10.1007/978-1-4614-5441-0_6] [PMID: 23393670]
[9]
Banks WA, Willoughby LM, Thomas DR, Morley JE. Insulin resistance syndrome in the elderly: assessment of functional, biochemical, metabolic, and inflammatory status. Diabetes Care 2007; 30(9): 2369-73.
[http://dx.doi.org/10.2337/dc07-0649] [PMID: 17536070]
[10]
Reske-Nielsen E, Lundbæk K, Rafaelsen OJ. Pathological changes in the central and peripheral nervous system of young long-term diabetics : I. Diabetic encephalopathy. Diabetologia 1966; 1(3-4): 233-41.
[http://dx.doi.org/10.1007/BF01257917] [PMID: 24173307]
[11]
Ott A, Stolk RP, van Harskamp F, Pols HA, Hofman A, Breteler MM. Diabetes mellitus and the risk of dementia: The Rotterdam Study. Neurology 1999; 53(9): 1937-42.
[http://dx.doi.org/10.1212/WNL.53.9.1937] [PMID: 10599761]
[12]
U’Ren RC, Riddle MC, Lezak MD, Bennington-Davis M. The mental efficiency of the elderly person with type II diabetes mellitus. J Am Geriatr Soc 1990; 38(5): 505-10.
[http://dx.doi.org/10.1111/j.1532-5415.1990.tb02398.x] [PMID: 2332570]
[13]
Huber JD. Diabetes, cognitive function, and the blood-brain barrier. Curr Pharm Des 2008; 14(16): 1594-600.
[http://dx.doi.org/10.2174/138161208784705441] [PMID: 18673200]
[14]
Rhea EM, Raber J, Banks WA. ApoE and cerebral insulin: Trafficking, receptors, and resistance. Neurobiol Dis 2020; 137: 104755-5.
[http://dx.doi.org/10.1016/j.nbd.2020.104755] [PMID: 31978603]
[15]
Sweeney MD, Zhao Z, Montagne A, Nelson AR, Zlokovic BV. Blood-brain barrier: from physiology to disease and back. Physiol Rev 2019; 99(1): 21-78.
[http://dx.doi.org/10.1152/physrev.00050.2017] [PMID: 30280653]
[16]
Campos-Bedolla P, Walter FR, Veszelka S, Deli MA. Role of the blood-brain barrier in the nutrition of the central nervous system. Arch Med Res 2014; 45(8): 610-38.
[http://dx.doi.org/10.1016/j.arcmed.2014.11.018] [PMID: 25481827]
[17]
Deane R, Sagare A, Zlokovic BV. The role of the cell surface LRP and soluble LRP in blood-brain barrier Abeta clearance in Alzheimer’s disease. Curr Pharm Des 2008; 14(16): 1601-5.
[http://dx.doi.org/10.2174/138161208784705487] [PMID: 18673201]
[18]
Banks WA. The blood-brain barrier as an endocrine tissue. Nat Rev Endocrinol 2019; 15(8): 444-55.
[http://dx.doi.org/10.1038/s41574-019-0213-7]
[19]
Erickson MA, Banks WA. Neuroimmune axes of the blood-brain barriers and blood-brain interfaces: Bases for physiological regulation, disease states, and pharmacological interventions. Pharmacol Rev 2018; 70(2): 278-314.
[http://dx.doi.org/10.1124/pr.117.014647] [PMID: 29496890]
[20]
Banks WA. From blood-brain barrier to blood-brain interface: new opportunities for CNS drug delivery. Nat Rev Drug Discov 2016; 15(4): 275-92.
[http://dx.doi.org/10.1038/nrd.2015.21] [PMID: 26794270]
[21]
Bradbury M. The concept of a blood-brain barrier. New York: John Wiley and Sons Ltd. 1979.
[22]
Brightman MW, Reese TS. Junctions between intimately apposed cell membranes in the vertebrate brain. J Cell Biol 1969; 40(3): 648-77.
[http://dx.doi.org/10.1083/jcb.40.3.648] [PMID: 5765759]
[23]
Reese TS, Karnovsky MJ. Fine structural localization of a blood-brain barrier to exogenous peroxidase. J Cell Biol 1967; 34(1): 207-17.
[http://dx.doi.org/10.1083/jcb.34.1.207] [PMID: 6033532]
[24]
Cong X, Kong W. Endothelial tight junctions and their regulatory signaling pathways in vascular homeostasis and disease. Cell Signal 2020; 66: 109485-5.
[http://dx.doi.org/10.1016/j.cellsig.2019.109485] [PMID: 31770579]
[25]
Tjakra M, Wang Y, Vania V, et al. Overview of crosstalk between multiple factor of transcytosis in blood brain barrier. Front Neurosci 2020; 13: 1436-6.
[http://dx.doi.org/10.3389/fnins.2019.01436] [PMID: 32038141]
[26]
Toth AE, Holst MR, Nielsen MS. Vesicular transport machinery in brain endothelial cells: what we know and what we don’t. Curr Pharm Des 2020; 26(13): 1405-16.
[http://dx.doi.org/10.2174/1381612826666200212113421] [PMID: 32048959]
[27]
Preston JE, Joan Abbott N, Begley DJ. Transcytosis of macromolecules at the blood-brain barrier. Adv Pharmacol 2014; 71: 147-63.
[http://dx.doi.org/10.1016/bs.apha.2014.06.001] [PMID: 25307216]
[28]
Hardebo JE, Owman C. Enzymatic barrier mechanisms for neurotransmitter monoamines and their precursors at the blood-brain barrier Pathophysiology of the blood-brain barrier. Elsevier Amsterdam 1990; pp. 41-55.
[29]
Keller BT, Borchardt RT. Cultured bovine brain capillary endothelial cells (BBCEC) - a blood-brain barrier model for studying the binding and internalization of insulin and insulin-like growth factor 1. Fed Proc 1987; 46: 416.
[30]
Ding Y, Wang R, Zhang J, et al. Potential regulation mechanisms of p-gp in the blood-brain barrier in hypoxia. Curr Pharm Des 2019; 25(10): 1041-51.
[http://dx.doi.org/10.2174/1381612825666190610140153] [PMID: 31187705]
[31]
Banks WA. Critical roles of efflux systems in health and disease. Efflux transporters and the blood-brain barrier 2005; 21-53.
[32]
Fromm MF. Importance of P-glycoprotein at blood-tissue barriers. Trends Pharmacol Sci 2004; 25(8): 423-9.
[http://dx.doi.org/10.1016/j.tips.2004.06.002] [PMID: 15276711]
[33]
Donahue JE, Flaherty SL, Johanson CE, et al. RAGE, LRP-1, and amyloid-beta protein in Alzheimer’s disease. Acta Neuropathol 2006; 112(4): 405-15.
[http://dx.doi.org/10.1007/s00401-006-0115-3] [PMID: 16865397]
[34]
van Assema DM, Lubberink M, Bauer M, et al. Blood-brain barrier P-glycoprotein function in Alzheimer’s disease. Brain 2012; 135(Pt 1): 181-9.
[http://dx.doi.org/10.1093/brain/awr298] [PMID: 22120145]
[35]
Nicolazzo JA, Banks WA. Decreased blood-brain barrier expression of P-glycoprotein in Alzheimer’s disease: impact on pathogenesis and brain access of therapeutic agents. Ther Deliv 2011; 2(7): 841-4.
[http://dx.doi.org/10.4155/tde.11.65] [PMID: 22833898]
[36]
Hartz AMS, Miller DS, Bauer B. Restoring blood-brain barrier P-glycoprotein reduces brain amyloid-beta in a mouse model of Alzheimer’s disease. Mol Pharmacol 2010; 77(5): 715-23.
[http://dx.doi.org/10.1124/mol.109.061754] [PMID: 20101004]
[37]
Schinkel AH, Wagenaar E, Mol CAAM, van Deemter L. P-glycoprotein in the blood-brain barrier of mice influences the brain penetration and pharmacological activity of many drugs. J Clin Invest 1996; 97(11): 2517-24.
[http://dx.doi.org/10.1172/JCI118699] [PMID: 8647944]
[38]
Tsuji A, Terasaki T, Takabatake Y, et al. P-glycoprotein as the drug efflux pump in primary cultured bovine brain capillary endothelial cells. Life Sci 1992; 51(18): 1427-37.
[http://dx.doi.org/10.1016/0024-3205(92)90537-Y] [PMID: 1357522]
[39]
Cordon-Cardo C, O’Brien JP, Casals D, et al. Multidrug-resistance gene (P-glycoprotein) is expressed by endothelial cells at blood-brain barrier sites. Proc Natl Acad Sci USA 1989; 86(2): 695-8.
[http://dx.doi.org/10.1073/pnas.86.2.695] [PMID: 2563168]
[40]
Davson H, Segal MB. Special aspects of the blood-brain barrier Physiology of the CSF and blood-brain barriers. Boca Raton: CRC Press 1996; pp. 303-485.
[41]
Oldendorf WH. Carrier-mediated blood-brain barrier transport of short-chain monocarboxylic organic acids. Am J Physiol 1973; 224(6): 1450-3.
[http://dx.doi.org/10.1152/ajplegacy.1973.224.6.1450] [PMID: 4712154]
[42]
Fishman JB, Rubin JB, Handrahan JV, Connor JR, Fine RE. Receptor-mediated transcytosis of transferrin across the blood-brain barrier. J Neurosci Res 1987; 18(2): 299-304.
[http://dx.doi.org/10.1002/jnr.490180206] [PMID: 3694713]
[43]
Cserr HF, Berman BJ. Iodide and thiocyanate efflux from brain following injection into rat caudate nucleus. Am J Physiol 1978; 235(4): F331-7.
[PMID: 696872]
[44]
Lipinski CA, Lombardo F, Dominy BW, Feeney PJ. Experimental and computational approaches to estimate solubility and permeability in drug discovery and developmental settings. Adv Drug Deliv Rev 1997; 23: 3-25.
[http://dx.doi.org/10.1016/S0169-409X(96)00423-1]
[45]
Stein WD. Transport and diffusion across cell membranes. London: Academic Press, Inc. 1986.
[46]
Broadwell RD, Baker-Cairns BJ, Friden PM, Oliver C, Villegas JC. Transcytosis of protein through the mammalian cerebral epithelium and endothelium. III. Receptor-mediated transcytosis through the blood-brain barrier of blood-borne transferrin and antibody against the transferrin receptor. Exp Neurol 1996; 142(1): 47-65.
[http://dx.doi.org/10.1006/exnr.1996.0178] [PMID: 8912898]
[47]
Broadwell RD, Balin BJ, Salcman M. Transcytotic pathway for blood-borne protein through the blood-brain barrier. Proc Natl Acad Sci USA 1988; 85(2): 632-6.
[http://dx.doi.org/10.1073/pnas.85.2.632] [PMID: 2448779]
[48]
Wang Y, Zhang JH, Sheng J, Shao A. Immunoreactive cells after cerebral ischemia. Front Immunol 2019; 10: 2781-1.
[http://dx.doi.org/10.3389/fimmu.2019.02781] [PMID: 31849964]
[49]
Greenwood J, Heasman SJ, Alvarez JI, Pratt A. Lyck r, Engelhardt B. Review: Leukocyte-endothelial cell crosstalk at the blood-brain barrier: a prerequisite for successful immune cell entry to the brain. Neuropathol Appl Neurobiol 2011; 37: 24-39.
[http://dx.doi.org/10.1111/j.1365-2990.2010.01140.x] [PMID: 20946472]
[50]
Persidsky Y, Ramirez SH, Haorah J, Kanmogne GD. Blood-brain barrier: structural components and function under physiologic and pathologic conditions. J Neuroimmune Pharmacol 2006; 1(3): 223-36.
[http://dx.doi.org/10.1007/s11481-006-9025-3] [PMID: 18040800]
[51]
Banks WA. Role of the blood-brain barrier in communication between the central nervous system and the peripheral tissues Blood-spinal cord and brain barriers in health and disease. Amsterdam: Elsevier, Inc. 2004; pp. 73-81.
[http://dx.doi.org/10.1016/B978-012639011-7/50012-7]
[52]
Davis TP, Sanchez-Covarubias L, Tome ME. P-glycoprotein trafficking as a therapeutic target to optimize CNS drug delivery. Adv Pharmacol 2014; 71: 25-44.
[http://dx.doi.org/10.1016/bs.apha.2014.06.009] [PMID: 25307213]
[53]
Miller DS. ABC transporter regulation by signaling at the blood-brain barrier: relevance to pharmacology. Adv Pharmacol 2014; 71: 1-24.
[http://dx.doi.org/10.1016/bs.apha.2014.06.008] [PMID: 25307212]
[54]
Banks WA. Brain meets body: the blood-brain barrier as an endocrine interface. Endocrinology 2012; 153(9): 4111-9.
[http://dx.doi.org/10.1210/en.2012-1435] [PMID: 22778219]
[55]
Mándi Y, Ocsovszki I, Szabo D, Nagy Z, Nelson J, Molnar J. Nitric oxide production and MDR expression by human brain endothelial cells. Anticancer Res 1998; 18(4C): 3049-52.
[PMID: 9713508]
[56]
Engström L, Ruud J, Eskilsson A, et al. Lipopolysaccharide-induced fever depends on prostaglandin E2 production specifically in brain endothelial cells. Endocrinology 2012; 153(10): 4849-61.
[http://dx.doi.org/10.1210/en.2012-1375] [PMID: 22872578]
[57]
McGuire TR, Trickler WJ, Hock L, Vrana A, Hoie EB, Miller DW. Release of prostaglandin E-2 in bovine brain endothelial cells after exposure to three unique forms of the antifungal drug amphotericin-B: role of COX-2 in amphotericin-B induced fever. Life Sci 2003; 72(23): 2581-90.
[http://dx.doi.org/10.1016/S0024-3205(03)00172-3] [PMID: 12672504]
[58]
Verma S, Nakaoke R, Dohgu S, Banks WA. Release of cytokines by brain endothelial cells: A polarized response to lipopolysaccharide. Brain Behav Immun 2006; 20(5): 449-55.
[http://dx.doi.org/10.1016/j.bbi.2005.10.005] [PMID: 16309883]
[59]
Hawkins BT, Davis TP. The blood-brain barrier/neurovascular unit in health and disease. Pharmacol Rev 2005; 57(2): 173-85.
[http://dx.doi.org/10.1124/pr.57.2.4] [PMID: 15914466]
[60]
Dore-Duffy P. Pericytes: pluripotent cells of the blood brain barrier. Curr Pharm Des 2008; 14(16): 1581-93.
[http://dx.doi.org/10.2174/138161208784705469] [PMID: 18673199]
[61]
Dore-Duffy P, Katychev A, Wang X, Van Buren E. CNS microvascular pericytes exhibit multipotential stem cell activity. J Cereb Blood Flow Metab 2006; 26(5): 613-24.
[http://dx.doi.org/10.1038/sj.jcbfm.9600272] [PMID: 16421511]
[62]
Dore-Duffy P, Owen C, Balabanov R, Murphy S, Beaumont T, Rafols JA. Pericyte migration from the vascular wall in response to traumatic brain injury. Microvasc Res 2000; 60(1): 55-69.
[http://dx.doi.org/10.1006/mvre.2000.2244] [PMID: 10873515]
[63]
Hudson LC, Bragg DC, Tompkins MB, Meeker RB. Astrocytes and microglia differentially regulate trafficking of lymphocyte subsets across brain endothelial cells. Brain Res 2005; 1058(1-2): 148-60.
[http://dx.doi.org/10.1016/j.brainres.2005.07.071] [PMID: 16137663]
[64]
Starr JM, Wardlaw J, Ferguson K, MacLullich A, Deary IJ, Marshall I. Increased blood-brain barrier permeability in type II diabetes demonstrated by gadolinium magnetic resonance imaging. J Neurol Neurosurg Psychiatry 2003; 74(1): 70-6.
[http://dx.doi.org/10.1136/jnnp.74.1.70] [PMID: 12486269]
[65]
Abuhaiba SI, Cordeiro M, Amorim A, et al. Occipital blood-brain barrier permeability is an independent predictor of visual outcome in type 2 diabetes, irrespective of the retinal barrier: A longitudinal study. J Neuroendocrinol 2018; 30(1): 30.
[http://dx.doi.org/10.1111/jne.12566] [PMID: 29247551]
[66]
Janelidze S, Hertze J, Nägga K, et al. Increased blood-brain barrier permeability is associated with dementia and diabetes but not amyloid pathology or APOE genotype. Neurobiol Aging 2017; 51: 104-12.
[http://dx.doi.org/10.1016/j.neurobiolaging.2016.11.017] [PMID: 28061383]
[67]
Dai J, Vrensen GF, Schlingemann RO. Blood-brain barrier integrity is unaltered in human brain cortex with diabetes mellitus. Brain Res 2002; 954(2): 311-6.
[http://dx.doi.org/10.1016/S0006-8993(02)03294-8] [PMID: 12414115]
[68]
Vavilala MS, Richards TL, Roberts JS, et al. Change in blood-brain barrier permeability during pediatric diabetic ketoacidosis treatment. Pediatr Crit Care Med 2010; 11(3): 332-8.
[PMID: 19838141]
[69]
Xu Z, Zeng W, Sun J, et al. The quantification of blood-brain barrier disruption using dynamic contrast-enhanced magnetic resonance imaging in aging rhesus monkeys with spontaneous type 2 diabetes mellitus. Neuroimage 2017; 158: 480-7.
[http://dx.doi.org/10.1016/j.neuroimage.2016.07.017] [PMID: 27402601]
[70]
Fujihara R, Chiba Y, Nakagawa T, et al. Albumin microvascular leakage in brains with diabetes mellitus. Microsc Res Tech 2016; 79(9): 833-7.
[http://dx.doi.org/10.1002/jemt.22708] [PMID: 27333535]
[71]
Chehade JM, Haas MJ, Mooradian AD. Diabetes-related changes in rat cerebral occludin and zonula occludens-1 (ZO-1) expression. Neurochem Res 2002; 27(3): 249-52.
[http://dx.doi.org/10.1023/A:1014892706696] [PMID: 11958524]
[72]
Hawkins BT, Lundeen TF, Norwood KM, Brooks HL, Egleton RD. Increased blood-brain barrier permeability and altered tight junctions in experimental diabetes in the rat: contribution of hyperglycaemia and matrix metalloproteinases. Diabetologia 2007; 50(1): 202-11.
[http://dx.doi.org/10.1007/s00125-006-0485-z] [PMID: 17143608]
[73]
Huber JD, VanGilder RL, Houser KA. Streptozotocin-induced diabetes progressively increases blood-brain barrier permeability in specific brain regions in rats. Am J Physiol Heart Circ Physiol 2006; 291(6): H2660-8.
[http://dx.doi.org/10.1152/ajpheart.00489.2006] [PMID: 16951046]
[74]
Salameh TS, Mortell WG, Logsdon AF, Butterfield DA, Banks WA. Disruption of the hippocampal and hypothalamic blood-brain barrier in a diet-induced obese model of type II diabetes: prevention and treatment by the mitochondrial carbonic anhydrase inhibitor, topiramate. Fluids Barriers CNS 2019; 16(1): 1.
[http://dx.doi.org/10.1186/s12987-018-0121-6] [PMID: 30616618]
[75]
Salameh TS, Shah GN, Price TO, Hayden MR, Banks WA. Blood-brain barrier disruption and neurovascular unit dysfunction in diabetic mice: protection with the mitochondrial carbonic anhydrase inhibitor topiramate. J Pharmacol Exp Ther 2016; 359(3): 452-9.
[http://dx.doi.org/10.1124/jpet.116.237057] [PMID: 27729477]
[76]
Rhea EM, Banks WA. Role of the blood-brain barrier in central nervous system insulin resistance. Front Neurosci 2019; 13: 521.
[http://dx.doi.org/10.3389/fnins.2019.00521] [PMID: 31213970]
[77]
Liu H, Xu X, Yang Z, Deng Y, Liu X, Xie L. Impaired function and expression of P-glycoprotein in blood-brain barrier of streptozotocin-induced diabetic rats. Brain Res 2006; 1123(1): 245-52.
[http://dx.doi.org/10.1016/j.brainres.2006.09.061] [PMID: 17074306]
[78]
Wu KC, Pan HJ, Yin HS, Chen MR, Lu SC, Lin CJ. Change in P-glycoprotein and caveolin protein expression in brain striatum capillaries in New Zealand obese mice with type 2 diabetes. Life Sci 2009; 85(23-26): 775-81.
[http://dx.doi.org/10.1016/j.lfs.2009.10.014] [PMID: 19891976]
[79]
Reichel V, Burghard S, John I, Huber O. P-glycoprotein and breast cancer resistance protein expression and function at the blood-brain barrier and blood-cerebrospinal fluid barrier (choroid plexus) in streptozotocin-induced diabetes in rats. Brain Res 2011; 1370: 238-45.
[http://dx.doi.org/10.1016/j.brainres.2010.11.012] [PMID: 21075088]
[80]
Maeng HJ, Kim MH, Jin HE, et al. Functional induction of P-glycoprotein in the blood-brain barrier of streptozotocin-induced diabetic rats: evidence for the involvement of nuclear factor-kappaB, a nitrosative stress-sensitive transcription factor, in the regulation. Drug Metab Dispos 2007; 35(11): 1996-2005.
[http://dx.doi.org/10.1124/dmd.107.015800] [PMID: 17664251]
[81]
Hong H, Liu LP, Liao JM, Wang TS, Ye FY. Wu.J. Downregulation of LRP1 at the blood-brain barrier in streptozotocin-induced diabetic mice. Neuropharmacology 2009; 56: 1054-9.
[http://dx.doi.org/10.1016/j.neuropharm.2009.03.001] [PMID: 19285094]
[82]
Liu LP, Hong H, Liao JM, et al. Upregulation of RAGE at the blood-brain barrier in streptozotocin-induced diabetic mice. Synapse 2009; 63(8): 636-42.
[http://dx.doi.org/10.1002/syn.20644] [PMID: 19347957]
[83]
Peila R, Rodriguez BL, Launer LJ. Type 2 diabetes, APOE gene, and the risk for dementia and related pathologies: The Honolulu-Asia Aging Study. Diabetes 2002; 51(4): 1256-62.
[http://dx.doi.org/10.2337/diabetes.51.4.1256] [PMID: 11916953]
[84]
Liu Y, Liu H, Yang J, et al. Increased amyloid beta-peptide (1-40) level in brain of streptozotocin-induced diabetic rats. Neuroscience 2008; 153(3): 796-802.
[http://dx.doi.org/10.1016/j.neuroscience.2008.03.019] [PMID: 18424002]
[85]
Qu ZS, Tian Q, Zhou XW, et al. [Alteration of beta-amyloid and glutamate transporter in the brain of diabetes rats and the underlying mechanism]. Zhongguo Yi Xue Ke Xue Yuan Xue Bao 2005; 27(6): 708-11.
[PMID: 16447643]
[86]
Shuli S, Yongmei Z, Zhiwei Z, Zhijuan J. beta-Amyloid and its binding protein in the hippocampus of diabetic mice: effect of APP17 peptide. Neuroreport 2001; 12(15): 3317-9.
[http://dx.doi.org/10.1097/00001756-200110290-00034] [PMID: 11711878]
[87]
Higashida T, Kreipke CW, Rafols JA, et al. The role of hypoxia-inducible factor-1α, aquaporin-4, and matrix metalloproteinase-9 in blood-brain barrier disruption and brain edema after traumatic brain injury. J Neurosurg 2011; 114(1): 92-101.
[http://dx.doi.org/10.3171/2010.6.JNS10207] [PMID: 20617879]
[88]
Rom S, Zuluaga-Ramirez V, Gajghate S, et al. Hyperglycemia-driven neuroinflammation compromises BBB leading to memory loss in both diabetes mellitus (DM) type 1 and type 2 mouse models. Mol Neurobiol 2019; 56(3): 1883-96.
[http://dx.doi.org/10.1007/s12035-018-1195-5] [PMID: 29974394]
[89]
Shimizu F, Sano Y, Tominaga O, Maeda T, Abe MA, Kanda T. Advanced glycation end-products disrupt the blood-brain barrier by stimulating the release of transforming growth factor-β by pericytes and vascular endothelial growth factor and matrix metalloproteinase-2 by endothelial cells in vitro. Neurobiol Aging 2013; 34(7): 1902-12.
[http://dx.doi.org/10.1016/j.neurobiolaging.2013.01.012] [PMID: 23428182]
[90]
Mooradian AD. Blood-brain barrier choline transport is reduced in diabetic rats. Diabetes 1987; 36(10): 1094-7.
[http://dx.doi.org/10.2337/diab.36.10.1094] [PMID: 3653526]
[91]
Kastin AJ, Akerstrom V. Glucose and insulin increase the transport of leptin through the blood-brain barrier in normal mice but not in streptozotocin-diabetic mice. Neuroendocrinology 2001; 73(4): 237-42.
[http://dx.doi.org/10.1159/000054640] [PMID: 11340337]
[92]
Mayhan WG, Patel KP. Acute effects of glucose on reactivity of cerebral microcirculation: role of activation of protein kinase C. Am J Physiol 1995; 269(4 Pt. 2): H1297-302.
[PMID: 7485561]
[93]
Banks WA, Jaspan JB, Kastin AJ. Effect of diabetes mellitus on the permeability of the blood-brain barrier to insulin. Peptides 1997; 18(10): 1577-84.
[http://dx.doi.org/10.1016/S0196-9781(97)00238-6] [PMID: 9437719]
[94]
Kaiyala KJ, Prigeon RL, Kahn SE, Woods SC, Schwartz MW. Obesity induced by a high-fat diet is associated with reduced brain insulin transport in dogs. Diabetes 2000; 49(9): 1525-33.
[http://dx.doi.org/10.2337/diabetes.49.9.1525] [PMID: 10969837]
[95]
Heni M, Schöpfer P, Peter A, et al. Evidence for altered transport of insulin across the blood-brain barrier in insulin-resistant humans. Acta Diabetol 2014; 51(4): 679-81.
[http://dx.doi.org/10.1007/s00592-013-0546-y] [PMID: 24370925]
[96]
Banks WA, Niehoff ML, Ponzio NM, Erickson MA, Zalcman SS. Pharmacokinetics and modeling of immune cell trafficking: quantifying differential influences of target tissues versus lymphocytes in SJL and lipopolysaccharide-treated mice. J Neuroinflammation 2012; 9: 231.
[http://dx.doi.org/10.1186/1742-2094-9-231] [PMID: 23034075]
[97]
Hu P, Thinschmidt JS, Yan Y, et al. CNS inflammation and bone marrow neuropathy in type 1 diabetes. Am J Pathol 2013; 183(5): 1608-20.
[http://dx.doi.org/10.1016/j.ajpath.2013.07.009] [PMID: 24160325]
[98]
Soltésová D, Veselá A, Mravec B, Herichová I. Daily profile of glut1 and glut4 expression in tissues inside and outside the blood-brain barrier in control and streptozotocin-treated rats. Physiol Res 2013; 62(Suppl. 1): S115-24.
[PMID: 24329691]
[99]
van de Ven KC, van der Graaf M, Tack CJ, Heerschap A, de Galan BE. Steady-state brain glucose concentrations during hypoglycemia in healthy humans and patients with type 1 diabetes. Diabetes 2012; 61(8): 1974-7.
[http://dx.doi.org/10.2337/db11-1778] [PMID: 22688331]
[100]
Minamizono A, Tomi M, Hosoya K. Inhibition of dehydroascorbic acid transport across the rat blood-retinal and -brain barriers in experimental diabetes. Biol Pharm Bull 2006; 29(10): 2148-50.
[http://dx.doi.org/10.1248/bpb.29.2148] [PMID: 17015969]
[101]
Mooradian AD, Smith TL. The effect of experimentally induced diabetes mellitus on the lipid order and composition of rat cerebral microvessels. Neurosci Lett 1992; 145(2): 145-8.
[http://dx.doi.org/10.1016/0304-3940(92)90007-T] [PMID: 1465210]
[102]
Price TO, Eranki V, Banks WA, Ercal N, Shah GN. Topiramate treatment protects blood-brain barrier pericytes from hyperglycemia-induced oxidative damage in diabetic mice. Endocrinology 2012; 153(1): 362-72.
[http://dx.doi.org/10.1210/en.2011-1638] [PMID: 22109883]
[103]
Shah GN, Morofuji Y, Banks WA, Price TO. High glucose-induced mitochondrial respiration and reactive oxygen species in mouse cerebral pericytes is reversed by pharmacological inhibition of mitochondrial carbonic anhydrases: Implications for cerebral microvascular disease in diabetes. Biochem Biophys Res Commun 2013; 440(2): 354-8.
[http://dx.doi.org/10.1016/j.bbrc.2013.09.086] [PMID: 24076121]
[104]
Shah GN, Price TO, Banks WA, et al. Pharmacological inhibition of mitochondrial carbonic anhydrases protects mouse cerebral pericytes from high glucose-induced oxidative stress and apoptosis. J Pharmacol Exp Ther 2013; 344(3): 637-45.
[http://dx.doi.org/10.1124/jpet.112.201400] [PMID: 23249625]
[105]
Li W, Maloney RE, Circu ML, Alexander JS, Aw TY. Acute carbonyl stress induces occludin glycation and brain microvascular endothelial barrier dysfunction: role for glutathione-dependent metabolism of methylglyoxal. Free Radic Biol Med 2013; 54: 51-61.
[http://dx.doi.org/10.1016/j.freeradbiomed.2012.10.552] [PMID: 23108103]
[106]
Corem N, Anzi S, Gelb S, Ben-Zvi A. Leptin receptor deficiency induces early, transient and hyperglycaemia-independent blood-brain barrier dysfunction. Sci Rep 2019; 9(1): 2884.
[http://dx.doi.org/10.1038/s41598-019-39230-1] [PMID: 30814586]
[107]
Mäe MA, Li T, Bertuzzi G, et al. Prolonged systemic hyperglycemia does not cause pericyte loss and permeability at the mouse blood-brain barrier. Sci Rep 2018; 8(1): 17462.
[http://dx.doi.org/10.1038/s41598-018-35576-0] [PMID: 30498224]
[108]
Taylor SL, Trudeau D, Arnold B, et al. VEGF can protect against blood brain barrier dysfunction, dendritic spine loss and spatial memory impairment in an experimental model of diabetes. Neurobiol Dis 2015; 78: 1-11.
[http://dx.doi.org/10.1016/j.nbd.2015.03.022] [PMID: 25829228]
[109]
Sajja RK, Prasad S, Tang S, Kaisar MA, Cucullo L. Blood-brain barrier disruption in diabetic mice is linked to Nrf2 signaling deficits: Role of ABCB10? Neurosci Lett 2017; 653: 152-8.
[http://dx.doi.org/10.1016/j.neulet.2017.05.059] [PMID: 28572033]
[110]
Sandoval DA, Obici S, Seeley RJ. Targeting the CNS to treat type 2 diabetes. Nat Rev Drug Discov 2009; 8(5): 386-98.
[http://dx.doi.org/10.1038/nrd2874] [PMID: 19404312]
[111]
Woods SC, Lotter EC, McKay LD, Porte D Jr. Chronic intracerebroventricular infusion of insulin reduces food intake and body weight of baboons. Nature 1979; 282(5738): 503-5.
[http://dx.doi.org/10.1038/282503a0] [PMID: 116135]
[112]
Schwartz MW, Figlewicz DP, Baskin DG, Woods SC, Porte D Jr. Insulin in the brain: a hormonal regulator of energy balance. Endocr Rev 1992; 13(3): 387-414.
[PMID: 1425482]
[113]
Machida T, Takata F, Matsumoto J, et al. Contribution of thrombin-reactive brain pericytes to blood-brain barrier dysfunction in an in vivo mouse model of obesity-associated diabetes and an in vitro rat model. PLoS One 2017; 12(5): e0177447
[http://dx.doi.org/10.1371/journal.pone.0177447] [PMID: 28489922]
[114]
Tsuji A, Tamai I. Blood-brain barrier function of P-glycoprotein. Adv Drug Deliv Rev 1997; 25: 287-98.
[http://dx.doi.org/10.1016/S0169-409X(97)00504-8]
[115]
Taylor EM. The impact of efflux transporters in the brain on the development of drugs for CNS disorders. Clin Pharmacokinet 2002; 41(2): 81-92.
[http://dx.doi.org/10.2165/00003088-200241020-00001] [PMID: 11888329]
[116]
Begley DJ. ABC transporters and the blood-brain barrier. Curr Pharm Des 2004; 10(12): 1295-312.
[http://dx.doi.org/10.2174/1381612043384844] [PMID: 15134482]
[117]
Juhler M, Barry DI, Offner H, Konat G, Klinken L, Paulson OB. Blood-brain and blood-spinal cord barrier permeability during the course of experimental allergic encephalomyelitis in the rat. Brain Res 1984; 302(2): 347-55.
[http://dx.doi.org/10.1016/0006-8993(84)90249-X] [PMID: 6610460]
[118]
Logsdon AF, Meabon JS, Cline MM, et al. Blast exposure elicits blood-brain barrier disruption and repair mediated by tight junction integrity and nitric oxide dependent processes. Sci Rep 2018; 8(1): 11344.
[http://dx.doi.org/10.1038/s41598-018-29341-6] [PMID: 30054495]
[119]
He J, Hsuchou H, He Y, Kastin AJ, Wang Y, Pan W. Sleep restriction impairs blood-brain barrier function. J Neurosci 2014; 34(44): 14697-706.
[http://dx.doi.org/10.1523/JNEUROSCI.2111-14.2014] [PMID: 25355222]
[120]
Nakagawa S, Castro V, Toborek M. Infection of human pericytes by HIV-1 disrupts the integrity of the blood-brain barrier. J Cell Mol Med 2012; 16(12): 2950-7.
[http://dx.doi.org/10.1111/j.1582-4934.2012.01622.x] [PMID: 22947176]
[121]
Ryu JK, McLarnon JG. A leaky blood-brain barrier, fibrinogen infiltration and microglial reactivity in inflamed Alzheimer’s disease brain. J Cell Mol Med 2009; 13(9A): 2911-25.
[http://dx.doi.org/10.1111/j.1582-4934.2008.00434.x] [PMID: 18657226]
[122]
Liu H, Zhang D, Xu X, et al. Attenuated function and expression of P-glycoprotein at blood-brain barrier and increased brain distribution of phenobarbital in streptozotocin-induced diabetic mice. Eur J Pharmacol 2007; 561(1-3): 226-32.
[http://dx.doi.org/10.1016/j.ejphar.2007.01.062] [PMID: 17349620]
[123]
Sun YN, Liu LB, Xue YX, Wang P. Effects of insulin combined with idebenone on blood-brain barrier permeability in diabetic rats. J Neurosci Res 2015; 93(4): 666-77.
[http://dx.doi.org/10.1002/jnr.23511] [PMID: 25421718]
[124]
Chen F, Dong RR, Zhong KL, et al. Antidiabetic drugs restore abnormal transport of amyloid-β across the blood-brain barrier and memory impairment in db/db mice. Neuropharmacology 2016; 101: 123-36.
[http://dx.doi.org/10.1016/j.neuropharm.2015.07.023] [PMID: 26211973]
[125]
Zanotto C, Simão F, Gasparin MS, et al. Exendin-4 reverses biochemical and functional alterations in the blood-brain and blood-CSF barriers in diabetic rats. Mol Neurobiol 2017; 54(3): 2154-66.
[http://dx.doi.org/10.1007/s12035-016-9798-1] [PMID: 26927659]
[126]
Fukuda S, Nakagawa S, Tatsumi R, et al. Glucagon-like peptide-1 strengthens the barrier integrity in primary cultures of rat brain endothelial cells under basal and hyperglycemic conditions. J Mol Neurosci 2016; 59(2): 211-9.
[http://dx.doi.org/10.1007/s12031-015-0696-1] [PMID: 26659380]
[127]
Aggarwal A, Khera A, Singh I, Sandhir R. S-nitrosoglutathione prevents blood-brain barrier disruption associated with increased matrix metalloproteinase-9 activity in experimental diabetes. J Neurochem 2015; 132(5): 595-608.
[http://dx.doi.org/10.1111/jnc.12939] [PMID: 25187090]
[128]
Kayano R, Morofuji Y, Nakagawa S, et al. In vitro analysis of drugs that improve hyperglycemia-induced blood-brain barrier dysfunction. Biochem Biophys Res Commun 2018; 503(3): 1885-90.
[http://dx.doi.org/10.1016/j.bbrc.2018.07.131] [PMID: 30060956]
[129]
Mooradian AD, Haas MJ, Batejko O, Hovsepyan M, Feman SS. Statins ameliorate endothelial barrier permeability changes in the cerebral tissue of streptozotocin-induced diabetic rats. Diabetes 2005; 54(10): 2977-82.
[http://dx.doi.org/10.2337/diabetes.54.10.2977] [PMID: 16186401]
[130]
Jain S, Sharma BM, Sharma B. Calcium channel blockade and peroxisome proliferator activated receptor γ agonism diminish cognitive loss and preserve endothelial function during diabetes mellitus. Curr Neurovasc Res 2016; 13(1): 33-44.
[http://dx.doi.org/10.2174/1567202613666151203233500] [PMID: 26648342]
[131]
Yu Z, Lin L, Jiang Y, et al. Recombinant FGF21 protects against blood-brain barrier leakage through Nrf2 upregulation in type 2 diabetes mice. Mol Neurobiol 2019; 56(4): 2314-27.
[http://dx.doi.org/10.1007/s12035-018-1234-2] [PMID: 30022432]
[132]
Ugochukwu NH, Mukes JD, Figgers CL. Ameliorative effects of dietary caloric restriction on oxidative stress and inflammation in the brain of streptozotocin-induced diabetic rats. Clin Chim Acta 2006; 370(1-2): 165-73.
[http://dx.doi.org/10.1016/j.cca.2006.02.003] [PMID: 16546151]
[133]
de Senna PN, Xavier LL, Bagatini PB, et al. Physical training improves non-spatial memory, locomotor skills and the blood brain barrier in diabetic rats. Brain Res 2015; 1618: 75-82.
[http://dx.doi.org/10.1016/j.brainres.2015.05.026] [PMID: 26032744]

Rights & Permissions Print Cite
© 2024 Bentham Science Publishers | Privacy Policy