In vitro models and systems for evaluating the dynamics of drug delivery to the healthy and diseased brain
Tài liệu tham khảo
Aird, 2007, Phenotypic heterogeneity of the endothelium: II. Representative vascular beds, Circ. Res., 100, 174, 10.1161/01.RES.0000255690.03436.ae
Gloor, 2001, Molecular and cellular permeability control at the blood–brain barrier, Brain Res. Rev., 36, 258, 10.1016/S0165-0173(01)00102-3
Zlokovic, 2008, The blood-brain barrier in health and chronic neurodegenerative disorders, Neuron, 57, 178, 10.1016/j.neuron.2008.01.003
Daneman, 2012, The blood–brain barrier in health and disease, Ann. Neurol., 72, 648, 10.1002/ana.23648
Lakin, 2007, Modeling intracranial pressures in microgravity: the influence of the blood-brain barrier, Aviat. Space Environ. Med., 78, 932, 10.3357/ASEM.2060.2007
Janmaleki, 2016, Impact of simulated microgravity on cytoskeleton and viscoelastic properties of endothelial cell, Sci. Rep., 6, 10.1038/srep32418
Kou, 2018, Transporter-guided delivery of nanoparticles to improve drug permeation across cellular barriers and drug exposure to selective cell types, Front. Pharmacol., 9, 27, 10.3389/fphar.2018.00027
Luissint, 2012, Tight junctions at the blood brain barrier: physiological architecture and disease-associated dysregulation, Fluids Barriers CNS, 9, 23, 10.1186/2045-8118-9-23
Luissint, 2012, Guanine nucleotide-binding protein Gαi2: a new partner of Claudin-5 that regulates tight junction integrity in human brain endothelial cells, J. Cereb. Blood Flow Metab., 32, 860, 10.1038/jcbfm.2011.202
Anderson, 2009, Physiology and function of the tight junction, Cold Spring Harb. Perspect. Biol., 1, 10.1101/cshperspect.a002584
Daugherty, 2007, Regulation of heterotypic claudin compatibility, J. Biol. Chem., 282, 30005, 10.1074/jbc.M703547200
Nitta, 2003, Size-selective loosening of the blood-brain barrier in claudin-5-deficient mice, J. Cell Biol., 161, 653, 10.1083/jcb.200302070
Daneman, 2010, The Mouse blood-brain barrier transcriptome: a new resource for understanding the development and function of brain endothelial cells, PLoS One, 5, 10.1371/journal.pone.0013741
Furuse, 2010, Molecular basis of the core structure of tight junctions, Cold Spring Harb. Perspect. Biol., 2, 10.1101/cshperspect.a002907
Masuda, 2011, LSR defines cell corners for tricellular tight junction formation in epithelial cells, J. Cell Sci., 124, 548, 10.1242/jcs.072058
Wolburg-Buchholz, 2009, Loss of astrocyte polarity marks blood–brain barrier impairment during experimental autoimmune encephalomyelitis, Acta Neuropathol., 118, 219, 10.1007/s00401-009-0558-4
Ueno, 2010, Transporters in the Brain Endothelial Barrier, Curr. Med. Chem., 17, 1125, 10.2174/092986710790827816
Kido, 2000, Functional clarification of MCT1-mediated transport of monocarboxylic acids at the blood-brain barrier using in vitro cultured cells and in vivo BUI studies, Pharm. Res., 17, 55, 10.1023/A:1007518525161
Stewart, 1981, Developing nervous tissue induces formation of blood-brain barrier characteristics in invading endothelial cells: a study using quail-chick transplantation chimeras, Dev. Biol., 84, 183, 10.1016/0012-1606(81)90382-1
Lee, 2003, SSeCKS regulates angiogenesis and tight junction formation in blood-brain barrier, Nat. Med., 9, 900, 10.1038/nm889
Wosik, 2007, controls occludin function and is required for blood–brain barrier maintenance: relevance to multiple sclerosis, J. Neurosci., 27, 9032, 10.1523/JNEUROSCI.2088-07.2007
Daneman, 2010, Pericytes are required for blood-brain barrier integrity during embryogenesis, Nature, 468, 562, 10.1038/nature09513
Dohgu, 2005, Brain pericytes contribute to the induction and up-regulation of blood–brain barrier functions through transforming growth factor-β production, Brain Res., 1038, 208, 10.1016/j.brainres.2005.01.027
Daneman, 2009, Wnt/β-catenin signaling is required for CNS, but not non-CNS, angiogenesis, Proc. Natl. Acad. Sci. U. S. A., 106, 641, 10.1073/pnas.0805165106
Macdonald, 2010, Endothelial cell heterogeneity of blood-brain barrier gene expression along the cerebral microvasculature, J. Neurosci. Res., 88, 1457
Gross, 1992, Chapter 31: Circumventricular organ capillaries, 219, 10.1016/S0079-6123(08)62338-9
Cullen, 2011, GPR124, an orphan G protein-coupled receptor, is required for CNS-specific vascularization and establishment of the blood–brain barrier, Proc. Natl. Acad. Sci. U. S. A., 108, 5759, 10.1073/pnas.1017192108
Pardridge, 2005, The blood-brain barrier: bottleneck in brain drug development, NeuroRx: the journal of the American Society for Experimental, Neurotherapeutics, 2, 3, 10.1602/neurorx.2.1.3
Fischer, 1998, Blood-brain barrier permeation: molecular parameters governing passive diffusion, J. Membr. Biol., 165, 201, 10.1007/s002329900434
Courade, 2001, Acetaminophen distribution in the rat central nervous system, Life Sci., 69, 1455, 10.1016/S0024-3205(01)01228-0
Abbott, 2010, Structure and function of the blood–brain barrier, Neurobiol. Dis., 37, 13, 10.1016/j.nbd.2009.07.030
Serlin, 2015, Anatomy and physiology of the blood–brain barrier, 2
Mueckler, 2013, The SLC2 (GLUT) family of membrane transporters, Mol. Asp. Med., 34, 121, 10.1016/j.mam.2012.07.001
Mosconi, 2008, Hippocampal hypometabolism predicts cognitive decline from normal aging, Neurobiol. Aging, 29, 676, 10.1016/j.neurobiolaging.2006.12.008
Landau, 2011, Associations between cognitive, functional, and FDG-PET measures of decline in AD and MCI, Neurobiol. Aging, 32, 1207, 10.1016/j.neurobiolaging.2009.07.002
Simpson, 2007, Supply and demand in cerebral energy metabolism: the role of nutrient transporters, J. Cereb. Blood Flow Metab., 27, 1766, 10.1038/sj.jcbfm.9600521
Patching, 2016, Glucose transporters at the blood-brain barrier: function, regulation and gateways for drug delivery, Mol. Neurobiol., 1
Wirth, 2014, Transport of thyroid hormone in brain, Front. Endocrinol., 5, 98, 10.3389/fendo.2014.00098
Smith, 1998, Blood-brain barrier amino acid transport, Introduction Blood–Brain Barrier, 1, 188, 10.1017/CBO9780511570216.020
Oldendorf, 1976, Amino acid assignment to one of three blood-brain barrier amino acid carriers, Am. J. Physiol., 230, 94, 10.1152/ajplegacy.1976.230.1.94
Kiernan, 2011, Amyotrophic lateral sclerosis, Lancet, 377, 942, 10.1016/S0140-6736(10)61156-7
Beart, 2007, Transporters for -glutamate: an update on their molecular pharmacology and pathological involvement, Br. J. Pharmacol., 150, 5, 10.1038/sj.bjp.0706949
Li, 2009, Soluble oligomers of amyloid β protein facilitate hippocampal long-term depression by disrupting neuronal glutamate uptake, Neuron, 62, 788, 10.1016/j.neuron.2009.05.012
Garrido-Sanabria, 2008, Impaired expression and function of group II metabotropic glutamate receptors in pilocarpine-treated chronically epileptic rats, Brain Res., 1240, 165, 10.1016/j.brainres.2008.08.084
Pardridge, 2012, Drug transport across the blood–brain barrier, J. Cereb. Blood Flow Metab., 32, 1959, 10.1038/jcbfm.2012.126
Bynoe, 2015, Adenosine receptor signaling: a key to opening the blood–brain door, Fluids Barriers CNS, 12, 1, 10.1186/s12987-015-0017-7
Carman, 2011, Adenosine receptor signaling modulates permeability of the blood–brain barrier, J. Neurosci., 31, 13272, 10.1523/JNEUROSCI.3337-11.2011
Eyal, 2009, Drug interactions at the blood-brain barrier: fact or fantasy?, Pharmacol. Ther., 123, 80, 10.1016/j.pharmthera.2009.03.017
Hagenbuch, 2004, Organic anion transporting polypeptides of the OATP/SLC21 family: phylogenetic classification as OATP/SLCO superfamily, new nomenclature and molecular/functional properties, Pflugers Arch., 447, 653, 10.1007/s00424-003-1168-y
Rizwan, 2007, Organic anion transporters of the SLC22 family: biopharmaceutical, physiological, and pathological roles, Pharm. Res., 24, 450, 10.1007/s11095-006-9181-4
Jonker, 2004, Pharmacological and physiological functions of the polyspecific organic cation transporters: OCT1, 2, and 3 (SLC22A1-3), J. Pharmacol. Exp. Ther., 308, 2, 10.1124/jpet.103.053298
Löscher, 2005, Role of drug efflux transporters in the brain for drug disposition and treatment of brain diseases, Prog. Neurobiol., 76, 22, 10.1016/j.pneurobio.2005.04.006
Potschka, 2004, Inhibition of multidrug transporters by verapamil or probenecid does not alter blood-brain barrier penetration of levetiracetam in rats, Epilepsy Res., 58, 85, 10.1016/j.eplepsyres.2003.12.007
Kamiie, 2008, Quantitative atlas of membrane transporter proteins: development and application of a highly sensitive simultaneous LC/MS/MS method combined with novel in-silico peptide selection criteria, Pharm. Res., 25, 1469, 10.1007/s11095-008-9532-4
Kuhnke, 2007, MDR1-P-Glycoprotein (ABCB1) mediates transport of alzheimer’s amyloid-β peptides—implications for the mechanisms of Aβ clearance at the blood–brain barrier, Brain Pathol., 17, 347, 10.1111/j.1750-3639.2007.00075.x
Hartz, 2010, Restoring blood-brain barrier P-glycoprotein reduces brain amyloid-beta in a mouse model of Alzheimer's disease, Mol. Pharmacol., 77, 715, 10.1124/mol.109.061754
van Assema, 2012, Blood-brain barrier P-glycoprotein function in Alzheimer's disease, Brain, 135, 181, 10.1093/brain/awr298
Daneman, 2015, The blood–brain barrier, Cold Spring Harb. Perspect. Biol., 7, 10.1101/cshperspect.a020412
Miller, 2014, Signaling pathways that regulate basal ABC transporter activity at the blood-brain barrier, Curr. Pharm. Des., 20, 1463, 10.2174/13816128113199990457
Bankstahl, 2008, Tariquidar-induced P-glycoprotein inhibition at the rat blood–brain barrier studied with (R)-11C-verapamil and PET, J. Nucl. Med., 49, 1328, 10.2967/jnumed.108.051235
Bart, 2003, Quantitative assessment of P-glycoprotein function in the rat blood–brain barrier by distribution volume of [11 C] verapamil measured with PET, NeuroImage, 20, 1775, 10.1016/S1053-8119(03)00405-1
Hartz, 2010, Estrogen receptor β signaling through phosphatase and tensin homolog/phosphoinositide 3-kinase/Akt/glycogen synthase kinase 3 down-regulates blood-brain barrier breast cancer resistance protein, J. Pharmacol. Exp. Ther., 334, 467, 10.1124/jpet.110.168930
Erickson, 2013, Blood–brain barrier dysfunction as a cause and consequence of Alzheimer's disease, J. Cereb. Blood Flow Metab., 33, 1500, 10.1038/jcbfm.2013.135
De Boer, 2007, Drug targeting to the brain, Annu. Rev. Pharmacol. Toxicol., 47, 323, 10.1146/annurev.pharmtox.47.120505.105237
Singh, 2013, Low levels of copper disrupt brain amyloid-β homeostasis by altering its production and clearance, Proc. Natl. Acad. Sci. U. S. A., 110, 14771, 10.1073/pnas.1302212110
Deane, 2009, Clearance of amyloid-beta peptide across the blood-brain barrier: implication for therapies in Alzheimer's disease, CNS Neurol. Disord. Drug Targets, 8, 16, 10.2174/187152709787601867
Donahue, 2006, RAGE, LRP-1, and amyloid-beta protein in Alzheimer's disease, Acta Neuropathol., 112, 405, 10.1007/s00401-006-0115-3
Somjen, 2004
Oldendorf, 1977, The large apparent work capability of the blood-brain barrier: a study of the mitochondrial content of capillary endothelial cells in brain and other tissues of the rat, Ann. Neurol., 1, 409, 10.1002/ana.410010502
Taylor, 2006, Transporters involved in regulation of intracellular pH in primary cultured rat brain endothelial cells, J. Physiol., 576, 769, 10.1113/jphysiol.2006.117374
O’Donnell, 2014, Chapter four-blood–brain barrier Na transporters in ischemic stroke, Adv. Pharmacol., 71, 113, 10.1016/bs.apha.2014.06.011
Lam, 2009, Cerebral microvascular endothelial cell Na/H exchange: evidence for the presence of NHE1 and NHE2 isoforms and regulation by arginine vasopressin, Am. J. Phys. Cell Phys., 297, C278, 10.1152/ajpcell.00093.2009
Nicola, 2008, Transport activities involved in intracellular pH recovery following acid and alkali challenges in rat brain microvascular endothelial cells, Pflügers Arch. Eur. J. Physiol., 456, 801, 10.1007/s00424-007-0441-x
Abbott, 2013, Blood–brain barrier structure and function and the challenges for CNS drug delivery, J. Inherit. Metab. Dis., 36, 437, 10.1007/s10545-013-9608-0
Sloan, 2012, Analytical and biological methods for probing the blood-brain barrier, Annu. Rev. Anal. Chem. (Palo Alto, Calif.), 5, 505, 10.1146/annurev-anchem-062011-143002
Kingwell, 2016, Drug delivery: new targets for drug delivery across the BBB, Nat. Rev. Drug Discov., 15, 84, 10.1038/nrd.2016.14
Pardridge, 2015, Targeted delivery of protein and gene medicines through the blood–brain barrier, Clin. Pharmacol. Ther., 97, 347, 10.1002/cpt.18
Ransohoff, 2003, Three or more routes for leukocyte migration into the central nervous system, Nat. Rev. Immunol., 3, 569, 10.1038/nri1130
Engelhardt, 2005, The ins and outs of T-lymphocyte trafficking to the CNS: anatomical sites and molecular mechanisms, Trends Immunol., 26, 485, 10.1016/j.it.2005.07.004
Puentes, 2016, Non-neuronal cells in ALS: role of glial, immune cells and blood-CNS barriers, Brain Pathol., 26, 248, 10.1111/bpa.12352
Bien, 2002, Destruction of neurons by cytotoxic T cells: a new pathogenic mechanism in Rasmussen's encephalitis, Ann. Neurol., 51, 311, 10.1002/ana.10100
O’Brien, 2011, Amyloid precursor protein processing and Alzheimer’s disease, Annu. Rev. Neurosci., 34, 185, 10.1146/annurev-neuro-061010-113613
van de Haar, 2016, Blood-brain barrier leakage in patients with early Alzheimer disease, Radiology, 281, 527, 10.1148/radiol.2016152244
Brown, 2011, Cerebral microvascular pathology in ageing and neurodegeneration, Neuropathol. Appl. Neurobiol., 37, 56, 10.1111/j.1365-2990.2010.01139.x
Sharma, 2004
Sharma, 2012, The blood-brain barrier in Alzheimer's disease: novel therapeutic targets and nanodrug delivery, Int. Rev. Neurobiol., 102, 47, 10.1016/B978-0-12-386986-9.00003-X
Förster, 2008, Differential effects of hydrocortisone and TNFα on tight junction proteins in an in vitro model of the human blood–brain barrier, J. Physiol., 586, 1937, 10.1113/jphysiol.2007.146852
de Vries, 1996, The influence of cytokines on the integrity of the blood-brain barrier in vitro, J. Neuroimmunol., 64, 37, 10.1016/0165-5728(95)00148-4
Wong, 1999, Cadherin-5 redistribution at sites of TNF-α and IFN-γ-induced permeability in mesenteric venules, Am. J. Phys. Heart Circ. Phys., 276, H736
Zehendner, 2011, Caspase-3 contributes to ZO-1 and Cl-5 tight-junction disruption in rapid anoxic neurovascular unit damage, PLoS One, 6, 10.1371/journal.pone.0016760
Lopez-Ramirez, 2012, Role of caspases in cytokine-induced barrier breakdown in human brain endothelial cells, J. Immunol., 189, 3130, 10.4049/jimmunol.1103460
Zlokovic, 2011, Neurovascular pathways to neurodegeneration in Alzheimer's disease and other disorders, Nat. Rev. Neurosci., 12, 723, 10.1038/nrn3114
Soto-Rojas, 2015
Tolleson, 2013, Advances in the mechanisms of Parkinson’s disease, Discov. Med., 15, 61
Berman, 1999, Dopamine oxidation alters mitochondrial respiration and induces permeability transition in brain mitochondria, J. Neurochem., 73, 1127, 10.1046/j.1471-4159.1999.0731127.x
Kortekaas, 2005, Blood–brain barrier dysfunction in parkinsonian midbrain in vivo, Ann. Neurol., 57, 176, 10.1002/ana.20369
Hirano, 2008, Dissociation of metabolic and neurovascular responses to levodopa in the treatment of Parkinson's disease, J. Neurosci., 28, 4201, 10.1523/JNEUROSCI.0582-08.2008
Ohlin, 2011, Vascular endothelial growth factor is upregulated by L-dopa in the parkinsonian brain: implications for the development of dyskinesia, Brain, 134, 2339, 10.1093/brain/awr165
Lee, 2013, Disruption of the blood-brain barrier in Parkinson's disease: curse or route to a cure?, Front. Biosci., 19, 272, 10.2741/4206
Chen, 2008, Caffeine protects against MPTP-induced blood-brain barrier dysfunction in mouse striatum, J. Neurochem., 107, 1147
Carvey, 2005, 6-Hydroxydopamine-induced alterations in blood–brain barrier permeability, Eur. J. Neurosci., 22, 1158, 10.1111/j.1460-9568.2005.04281.x
Bradaric, 2012, Evidence for angiogenesis in Parkinson’s disease, incidental Lewy body disease, and progressive supranuclear palsy, J. Neural Transm., 119, 59, 10.1007/s00702-011-0684-8
Wong, 2004, Cytokines, nitric oxide, and cGMP modulate the permeability of an in vitro model of the human blood–brain barrier, Exp. Neurol., 190, 446, 10.1016/j.expneurol.2004.08.008
Dunckley, 2007, Whole-genome analysis of sporadic amyotrophic lateral sclerosis, N. Engl. J. Med., 357, 775, 10.1056/NEJMoa070174
Brettschneider, 2006, Axonal damage markers in cerebrospinal fluid are increased in ALS, Neurology, 66, 852, 10.1212/01.wnl.0000203120.85850.54
Henkel, 2009, Decreased mRNA expression of tight junction proteins in lumbar spinal cords of patients with ALS, Neurology, 72, 1614, 10.1212/WNL.0b013e3181a41228
Miyazaki, 2011, Disruption of neurovascular unit prior to motor neuron degeneration in amyotrophic lateral sclerosis, J. Neurosci. Res., 89, 718, 10.1002/jnr.22594
Garbuzova-Davis, 2012, Impaired blood–brain/spinal cord barrier in ALS patients, Brain Res., 1469, 114, 10.1016/j.brainres.2012.05.056
Zhong, 2008, ALS-causing SOD1 mutants generate vascular changes prior to motor neuron degeneration, Nat. Neurosci., 11, 420, 10.1038/nn2073
Nicaise, 2009, Impaired blood–brain and blood–spinal cord barriers in mutant SOD1-linked ALS rat, Brain Res., 1301, 152, 10.1016/j.brainres.2009.09.018
Winkler, 2012, Blood–spinal cord barrier pericyte reductions contribute to increased capillary permeability, J. Cereb. Blood Flow Metab., 32, 1841, 10.1038/jcbfm.2012.113
Liebner, 2000, Claudin-1 and claudin-5 expression and tight junction morphology are altered in blood vessels of human glioblastoma multiforme, Acta Neuropathol., 100, 323, 10.1007/s004010000180
Van Vliet, 2007, Blood–brain barrier leakage may lead to progression of temporal lobe epilepsy, Brain, 130, 521, 10.1093/brain/awl318
Korn, 2005, Focal cortical dysfunction and blood–brain barrier disruption in patients with Postconcussion syndrome, J. Clin. Neurophysiol., 22, 1, 10.1097/01.WNP.0000150973.24324.A7
Van Vliet, 2015, Blood–brain barrier dysfunction, seizures and epilepsy, 26
Cornford, 1995, High expression of the gluti glucose transporter in human brain hemangioblastoma endothelium, J. Neuropathol. Exp. Neurol., 54, 842, 10.1097/00005072-199511000-00011
Clos, 2010, Therapeutic removal of amyloid deposits in cutaneous amyloidosis by localised intra-lesional injections of anti-amyloid antibodies, Exp. Dermatol., 19, 904, 10.1111/j.1600-0625.2010.01121.x
Mamikonyan, 2007, Anti-Aβ1-11 antibody binds to different β-amyloid species, inhibits fibril formation, and disaggregates preformed fibrils but not the most toxic oligomers, J. Biol. Chem., 282, 22376, 10.1074/jbc.M700088200
Solomon, 1997, Disaggregation of Alzheimer β-amyloid by site-directed mAb, Proc. Natl. Acad. Sci. U. S. A., 94, 4109, 10.1073/pnas.94.8.4109
Sigurdsson, 2006, Immunotherapy for conformational diseases, Curr. Pharm. Des., 12, 2569, 10.2174/138161206777698837
Hrncic, 2000, Antibody-mediated resolution of light chain-associated amyloid deposits, Am. J. Pathol., 157, 1239, 10.1016/S0002-9440(10)64639-1
Kitazawa, 2009, Immunization with amyloid-β attenuates inclusion body myositis-like myopathology and motor impairment in a transgenic mouse model, J. Neurosci., 29, 6132, 10.1523/JNEUROSCI.1150-09.2009
Chauhan, 2007, Intracerebroventricular passive immunization with anti-oligoAβ antibody in TgCRND8, J. Neurosci. Res., 85, 451, 10.1002/jnr.21110
Masliah, 2005, Aβ vaccinaton effects on plaque pathology in the absence of encephalitis in Alzheimer disease, Neurology, 64, 129, 10.1212/01.WNL.0000148590.39911.DF
Schenk, 1999, Immunization with amyloid-β attenuates Alzheimer-disease-like pathology in the PDAPP mouse, Nature, 400, 173, 10.1038/22124
Orgogozo, 2003, Subacute meningoencephalitis in a subset of patients with AD after Aβ42 immunization, Neurology, 61, 46, 10.1212/01.WNL.0000073623.84147.A8
Nicoll, 2003, Neuropathology of human Alzheimer disease after immunization with amyloid-β peptide: a case report, Nat. Med., 9, 448, 10.1038/nm840
Ferrer, 2004, Neuropathology and pathogenesis of encephalitis following amyloid β immunization in Alzheimer's disease, Brain Pathol., 14, 11, 10.1111/j.1750-3639.2004.tb00493.x
Pisani, 2012, Increased blood-cerebrospinal fluid transfer of albumin in advanced Parkinson’s disease, J. Neuroinflammation, 9, 1186, 10.1186/1742-2094-9-188
Mullin, 2015, Pathogenic mechanisms of neurodegeneration in Parkinson disease, Neurol. Clin., 33, 1, 10.1016/j.ncl.2014.09.010
Ivens, 2007, TGF-β receptor-mediated albumin uptake into astrocytes is involved in neocortical epileptogenesis, Brain, 130, 535, 10.1093/brain/awl317
Fabene, 2008, A role for leukocyte-endothelial adhesion mechanisms in epilepsy, Nat. Med., 14, 1377, 10.1038/nm.1878
Librizzi, 2012, Seizure-induced brain-borne inflammation sustains seizure recurrence and blood–brain barrier damage, Ann. Neurol., 72, 82, 10.1002/ana.23567
Zeng, 2009, The mammalian target of rapamycin signaling pathway mediates epileptogenesis in a model of temporal lobe epilepsy, J. Neurosci., 29, 6964, 10.1523/JNEUROSCI.0066-09.2009
Gottesman, 2002, Multidrug resistance in cancer: role of ATP-dependent transporters, Nat. Rev. Cancer, 2, 48, 10.1038/nrc706
Meguro, 1999, Neocortical and hippocampal glucose hypometabolism following neurotoxic lesions of the entorhinal and perirhinal cortices in the non-human primate as shown by PET, Implications Alzheimers Dis., 122, 1519
Higgins, 2003, Transgenic mouse models of Alzheimer's disease: phenotype and application, Behav. Pharmacol., 14, 419
Ambudkar, 1999, Biochemical, cellular, and pharmacological aspects of the multidrug transporter, Annu. Rev. Pharmacol. Toxicol., 39, 361, 10.1146/annurev.pharmtox.39.1.361
Takano, 2006, Evaluation of in vivo P-Glycoprotein function at the blood–brain barrier among MDR1 gene polymorphisms by using 11C-Verapamil, J. Nucl. Med., 47, 1427
Cirrito, 2005, P-glycoprotein deficiency at the blood-brain barrier increases amyloid-β deposition in an Alzheimer disease mouse model, J. Clin. Investig., 115, 3285, 10.1172/JCI25247
Hsiao, 2006, Verapamil P-glycoprotein transport across the rat blood-brain barrier: cyclosporine, a concentration inhibition analysis, and comparison with human data, J. Pharmacol. Exp. Ther., 317, 704, 10.1124/jpet.105.097931
Elsinga, 1996, Carbon-11-labeled daunorubicin and verapamil for probing p-glycoprotein in tumors with PET, J. Nucl. Med., 37, 1571
Ikoma, 2006, Quantitative analysis of 11C-Verapamil transfer at the human blood–brain barrier for evaluation of p-glycoprotein function, J. Nucl. Med., 47, 1531
Dauer, 2003, Parkinson's disease, Neuron, 39, 889, 10.1016/S0896-6273(03)00568-3
Forno, 1986, Locus ceruleus lesions and eosinophilic inclusions in MPTP-treated monkeys, Ann. Neurol., 20, 449, 10.1002/ana.410200403
Waerzeggers, 2010, Mouse models in neurological disorders: applications of non-invasive imaging, Biochim. Biophys. Acta, 1802, 819, 10.1016/j.bbadis.2010.04.009
Tansey, 2007, Neuroinflammatory mechanisms in Parkinson's disease: potential environmental triggers, pathways, and targets for early therapeutic intervention, Exp. Neurol., 208, 1, 10.1016/j.expneurol.2007.07.004
Geldenhuys, 2012, Novel models for assessing blood–brain barrier drug permeation, Expert Opin. Drug Metab. Toxicol., 8, 647, 10.1517/17425255.2012.677433
Andersson, 2011, Models for predicting blood-brain barrier permeation, Drug Discov. Today, 16, 472
Mayer, 2009, Evolutionary conservation of vertebrate blood-brain barrier chemoprotective mechanisms in Drosophila, J. Neurosci., 29, 3538, 10.1523/JNEUROSCI.5564-08.2009
Jeong, 2008, Functional and developmental analysis of the blood–brain barrier in zebrafish, Brain Res. Bull., 75, 619, 10.1016/j.brainresbull.2007.10.043
El Andaloussi, 2013, Exosomes for targeted siRNA delivery across biological barriers, Adv. Drug Deliv. Rev., 65, 391, 10.1016/j.addr.2012.08.008
Alvarez-Erviti, 2011, Delivery of siRNA to the mouse brain by systemic injection of targeted exosomes, Nat. Biotechnol., 29, 341, 10.1038/nbt.1807
Burgess, 2011, Targeted delivery of neural stem cells to the brain using MRI-guided focused ultrasound to disrupt the blood-brain barrier, PLoS One, 6, 10.1371/journal.pone.0027877
Hammarlund-Udenaes, 2014
Banks, 2016, From blood-brain barrier to blood-brain interface: new opportunities for CNS drug delivery, Nat. Rev. Drug Discov., 15, 275, 10.1038/nrd.2015.21
Khawli, 2013, Drug delivery across the blood–brain barrier, Mol. Pharm., 10, 1471, 10.1021/mp400170b
Misra, 2003, Drug delivery to the central nervous system: a review, J. Pharm. Pharm. Sci., 6, 252
Lee, 2009, High-throughput analysis of alzheimer’s β-amyloid aggregation using a microfluidic self-assembly of monomersf, Anal. Chem., 81, 2751, 10.1021/ac802701z
Ying, 2010, Dual-targeting daunorubicin liposomes improve the therapeutic efficacy of brain glioma in animals, J. Control. Release, 141, 183, 10.1016/j.jconrel.2009.09.020
Aday, 2013, Stem cell-based human blood & brain barrier models for drug discovery and delivery, Trends Biotechnol., 34, 382, 10.1016/j.tibtech.2016.01.001
Meairs, 2015, Facilitation of drug transport across the blood–brain barrier with ultrasound and microbubbles, Pharmaceutics, 7, 275, 10.3390/pharmaceutics7030275
Chen, 2012, Modern methods for delivery of drugs across the blood–brain barrier, Adv. Drug Deliv. Rev., 64, 640, 10.1016/j.addr.2011.11.010
Schorey, 2008, Exosome function: from tumor immunology to pathogen biology, Traffic, 9, 871, 10.1111/j.1600-0854.2008.00734.x
Yu, 2014, Therapeutic bispecific antibodies cross the blood-brain barrier in nonhuman primates, Sci. Transl. Med., 6, 261ra154, 10.1126/scitranslmed.3009835
Hassanpour-Tamrin, 2017, Nanoscale optoregulation of neural stem cell differentiation by intracellular alteration of redox balance, Adv. Funct. Mater., 27, 10.1002/adfm.201701420
Ruck, 2015, Blood-brain barrier modeling: challenges and perspectives, Neural Regen. Res., 10, 889, 10.4103/1673-5374.158342
Naik, 2012, In vitro blood–brain barrier models: current and perspective technologies, J. Pharm. Sci., 101, 1337, 10.1002/jps.23022
Tietz, 2015, Brain barriers: crosstalk between complex tight junctions and adherens junctions, J. Cell Biol., 209, 493, 10.1083/jcb.201412147
Omolara, 2011, In vitro modeling of the blood-brain barrier: simplicity versus complexity, Curr. Pharm. Des., 17, 2755, 10.2174/138161211797440159
Booth, 2012, Characterization of a microfluidic in vitro model of the blood-brain barrier (μBBB), Lab Chip, 12, 1784, 10.1039/c2lc40094d
Griep, 2013, BBB on chip: microfluidic platform to mechanically and biochemically modulate blood-brain barrier function, Biomed. Microdevices, 15, 145, 10.1007/s10544-012-9699-7
Achyuta, 2013, A modular approach to create a neurovascular unit-on-a-chip, Lab Chip, 13, 542, 10.1039/C2LC41033H
Ogunshola, 2011, In vitro modeling of the blood-brain barrier: simplicity versus complexity, Curr. Pharm. Des., 17, 2755, 10.2174/138161211797440159
Cecchelli, 1999, In vitro model for evaluating drug transport across the blood–brain barrier, Adv. Drug Deliv. Rev., 36, 165, 10.1016/S0169-409X(98)00083-0
Tóth, 2011, Patented in vitro blood-brain barrier models in CNS drug discovery, Recent Patents CNS Drug Discov., 6, 107, 10.2174/157488911795933910
Nakagawa, 2009, A new blood–brain barrier model using primary rat brain endothelial cells, pericytes and astrocytes, Neurochem. Int., 54, 253, 10.1016/j.neuint.2008.12.002
Stanimirovic, 2015, Blood–brain barrier models: in vitro to in vivo translation in preclinical development of CNS-targeting biotherapeutics, Expert Opin. Drug Discovery, 10, 141, 10.1517/17460441.2015.974545
Culot, 2008, An in vitro blood-brain barrier model for high throughput (HTS) toxicological screening, Toxicol. in Vitro, 22, 799, 10.1016/j.tiv.2007.12.016
Cucullo, 2005, Drug delivery and in vitro models of the blood-brain barrier, Curr. Opin. Drug Discov. Dev., 8, 89
Yousif, 2013, Laminin isoforms in endothelial and perivascular basement membranes, Cell Adhes. Migr., 7, 101, 10.4161/cam.22680
Hartmann, 2007, The impact of glia-derived extracellular matrices on the barrier function of cerebral endothelial cells: an in vitro study, Exp. Cell Res., 313, 1318, 10.1016/j.yexcr.2007.01.024
Nakagawa, 2007, Pericytes from brain microvessels strengthen te barrier integrity in primary cultures of rat brain endothelial cells, Cell. Mol. Neurobiol., 27, 687, 10.1007/s10571-007-9195-4
Watson, 2013, Modelling the endothelial blood-CNS barriers: a method for the production of robust in vitro models of the rat blood-brain barrier and blood-spinal cord barrier, BMC Neurosci., 14, 1, 10.1186/1471-2202-14-59
Coisne, 2005, Mouse syngenic in vitro blood–brain barrier model: a new tool to examine inflammatory events in cerebral endothelium, Lab. Investig., 85, 734, 10.1038/labinvest.3700281
Patabendige, 2013, Establishment of a simplified in vitro porcine blood–brain barrier model with high transendothelial electrical resistance, Brain Res., 1521, 1, 10.1016/j.brainres.2012.06.057
Freeman, 2014, t-PA, but not desmoteplase, induces plasmin-dependent opening of a blood-brain barrier model under normoxic and ischaemic conditions, Brain Res., 1565, 63, 10.1016/j.brainres.2014.03.027
Muruganandam, 2002, Selection of phage-displayed llama single-domain antibodies that transmigrate across human blood–brain barrier endothelium, FASEB J., 16, 240, 10.1096/fj.01-0343fje
Cucullo, 2013, A new dynamic in vitro modular capillaries-venules modular system: cerebrovascular physiology in a box, BMC Neurosci., 14, 10.1186/1471-2202-14-18
Cucullo, 2008, Immortalized human brain endothelial cells and flow-based vascular modeling: a marriage of convenience for rational neurovascular studies, J. Cereb. Blood Flow Metab., 28, 312, 10.1038/sj.jcbfm.9600525
Hoshi, 2013, Quantitative atlas of blood–brain barrier transporters, receptors, and tight junction proteins in rats and common marmoset, J. Pharm. Sci., 102, 3343, 10.1002/jps.23575
Uchida, 2011, Quantitative targeted absolute proteomics of human blood–brain barrier transporters and receptors, J. Neurochem., 117, 333, 10.1111/j.1471-4159.2011.07208.x
Ito, 2011, Quantitative membrane protein expression at the blood–brain barrier of adult and younger cynomolgus monkeys, J. Pharm. Sci., 100, 3939, 10.1002/jps.22487
Ohtsuki, 2014, Quantitative targeted proteomics for understanding the blood–brain barrier: towards pharmacoproteomics, Expert Rev. Proteomics, 11, 303, 10.1586/14789450.2014.893830
Chun, 2011, The proteome of mouse brain microvessel membranes and basal lamina, J. Cereb. Blood Flow Metab., 31, 2267, 10.1038/jcbfm.2011.104
Warren, 2009, Comparative gene expression profiles of ABC transporters in brain microvessel endothelial cells and brain in five species including human, Pharmacol. Res., 59, 404, 10.1016/j.phrs.2009.02.007
Davidson, 2008, Structure, function, and evolution of bacterial ATP-binding cassette systems, Microbiol. Mol. Biol. Rev., 72, 317, 10.1128/MMBR.00031-07
Katoh, 2006, Kinetic analyses for species differences in P-glycoprotein-mediated drug transport, J. Pharm. Sci., 95, 2673, 10.1002/jps.20686
Gómez, 2013, GRL-04810 and GRL-05010, difluoride-containing nonpeptidic HIV-1 protease inhibitors (PIs) that inhibit the replication of multi-PI-resistant HIV-1 in vitro and possess favorable lipophilicity that may allow blood-brain barrier penetration, Antimicrob. Agents Chemother., 57, 6110, 10.1128/AAC.01420-13
Garberg, 2005, In vitro models for the blood–brain barrier, Toxicol. in Vitro, 19, 299, 10.1016/j.tiv.2004.06.011
Franke, 2000, Primary cultures of brain microvessel endothelial cells: a valid and flexible model to study drug transport through the blood–brain barrier in vitro, Brain Res. Protocol., 5, 248, 10.1016/S1385-299X(00)00020-9
Boveri, 2005, Induction of blood-brain barrier properties in cultured brain capillary endothelial cells: comparison between primary glial cells and C6 cell line, Glia, 51, 187, 10.1002/glia.20189
Bernas, 2010, Establishment of primary cultures of human brain microvascular endothelial cells to provide an in vitro cellular model of the blood-brain barrier, Nat. Protoc., 5, 1265, 10.1038/nprot.2010.76
Cioni, 2012, Expression of tight junction and drug efflux transporter proteins in an in vitro model of human blood–brain barrier, Front. Psychiatry, 3, 47, 10.3389/fpsyt.2012.00047
Bowman, 1983, Brain microvessel endothelial cells in tissue culture: a model for study of blood-brain barrier permeability, Ann. Neurol., 14, 396, 10.1002/ana.410140403
Miller, 1992, Application of cultured endothelial cells of the brain microvasculature in the study of the blood-brain barrier, J. Tissue Cult. Methods, 14, 217, 10.1007/BF01409014
Jeliazkova-Mecheva, 2003, A porcine astrocyte/endothelial cell co-culture model of the blood–brain barrier, Brain Res. Protocol., 12, 91, 10.1016/j.brainresprot.2003.08.004
Abbott, 2012, An improved in vitro blood–brain barrier model: rat brain endothelial cells co-cultured with astrocytes, Astrocytes, 415, 10.1007/978-1-61779-452-0_28
Navone, 2013, Isolation and expansion of human and mouse brain microvascular endothelial cells, Nat. Protoc., 8, 1680, 10.1038/nprot.2013.107
Wilhelm, 1949, I.n.A. Krizbai, In vitro models of the blood–brain barrier for the study of drug delivery to the brain, Mol. Pharm., 11
Aschner, 2006, Blood-brain barrier and cell-cell interactions: methods for establishing in vitro models of the blood-brain barrier and transport measurements, Cell-Cell Interactions, 1
Wolff, 2015, In vitro blood–brain barrier models—An overview of established models and new microfluidic approaches, J. Pharm. Sci., 104, 2727, 10.1002/jps.24329
Muruganandam, 1997, Development of immortalized human cerebromicrovascular endothelial cell line as an in vitro model of the human blood-brain barrier, FASEB J., 11, 1187, 10.1096/fasebj.11.13.9367354
Weksler, 2005, Blood-brain barrier-specific properties of a human adult brain endothelial cell line, FASEB J., 19, 1872, 10.1096/fj.04-3458fje
Weksler, 2013, The hCMEC/D3 cell line as a model of the human blood brain barrier, Fluids Barriers CNS, 10, 16, 10.1186/2045-8118-10-16
Prudhomme, 1996, Studies of Plasmodium falciparum cytoadherence using immortalized human brain capillary endothelial cells, Int. J. Parasitol., 26, 647, 10.1016/0020-7519(96)00027-6
Stins, 2001, Bacterial invasion and transcytosis in transfected human brain microvascular endothelial cells, Microb. Pathog., 30, 19, 10.1006/mpat.2000.0406
Kusch-Poddar, 2005, Evaluation of the immortalized human brain capillary endothelial cell line BB19 as a human cell culture model for the blood–brain barrier, Brain Res., 1064, 21, 10.1016/j.brainres.2005.10.014
Daniels, 2013, Immortalized human cerebral microvascular endothelial cells maintain the properties of primary cells in an in vitro model of immune migration across the blood brain barrier, J. Neurosci. Methods, 212, 173, 10.1016/j.jneumeth.2012.10.001
Urich, 2012, Transcriptional profiling of human brain endothelial cells reveals key properties crucial for predictive in vitro blood-brain barrier models, PLoS One, 7, 10.1371/journal.pone.0038149
Carl, 2010, ABC and SLC transporter expression and pot substrate characterization across the human CMEC/D3 blood−brain barrier cell line, Mol. Pharm., 7, 1057, 10.1021/mp900178j
Poller, 2008, The human brain endothelial cell line hCMEC/D3 as a human blood-brain barrier model for drug transport studies, J. Neurochem., 107, 1358, 10.1111/j.1471-4159.2008.05730.x
Eigenmann, 2013, Comparative study of four immortalized human brain capillary endothelial cell lines, hCMEC/D3, hBMEC, TY10, and BB19, and optimization of culture conditions, for an in vitro blood–brain barrier model for drug permeability studies, Fluids Barriers CNS, 10, 1, 10.1186/2045-8118-10-33
Shimizu, 2011, Peripheral nerve pericytes modify the blood–nerve barrier function and tight junctional molecules through the secretion of various soluble factors, J. Cell. Physiol., 226, 255, 10.1002/jcp.22337
Major, 1985, Establishment of a line of human fetal glial cells that supports JC virus multiplication, Proc. Natl. Acad. Sci. U. S. A., 82, 1257, 10.1073/pnas.82.4.1257
Shayan, 2011, Murine in vitro model of the blood–brain barrier for evaluating drug transport, Eur. J. Pharm. Sci., 42, 148, 10.1016/j.ejps.2010.11.005
Dehouck, 1990, An easier, reproducible, and mass-production method to study the blood–brain barrier in vitro, J. Neurochem., 54, 1798, 10.1111/j.1471-4159.1990.tb01236.x
Zysk, 2001, Pneumolysin is the main inducer of cytotoxicity to brain microvascular endothelial cells caused by Streptococcus pneumoniae, Infect. Immun., 69, 845, 10.1128/IAI.69.2.845-852.2001
Kis, 2001, Adrenomedullin regulates blood–brain barrier functions in vitro, Neuroreport, 12, 4139, 10.1097/00001756-200112210-00055
Deli, 2003, N, N-diethyl-2-[4-(phenylmethyl) phenoxy] ethanamine increases the permeability of primary mouse cerebral endothelial cell monolayers, Inflamm. Res., 52, s39, 10.1007/s000110300045
Smith, 2007, Primary porcine brain microvascular endothelial cells: biochemical and functional characterisation as a model for drug transport and targeting, J. Drug Target., 15, 253, 10.1080/10611860701288539
Hutamekalin, 2008, Effect of nicotine and polyaromtic hydrocarbons on cerebral endothelial cells, Cell Biol. Int., 32, 198, 10.1016/j.cellbi.2007.08.026
Ohtsuki, 2007, Exogenous expression of claudin-5 induces barrier properties in cultured rat brain capillary endothelial cells, J. Cell. Physiol., 210, 81, 10.1002/jcp.20823
Watanabe, 2013, Paracellular barrier and tight junction protein expression in the immortalized brain endothelial cell lines bEND. 3, bEND. 5 and mouse brain endothelial cell 4, Biol. Pharm. Bull., 36, 492, 10.1248/bpb.b12-00915
Cecchelli, 2014, A stable and reproducible human blood-brain barrier model derived from hematopoietic stem cells, PLoS One, 9, 10.1371/journal.pone.0099733
Lippmann, 2012, Derivation of blood-brain barrier endothelial cells from human pluripotent stem cells, Nat. Biotechnol., 30, 783, 10.1038/nbt.2247
Lu, 2012, A novel hypothesis of blood-brain barrier (BBB) development and in vitro BBB model: neural stem cell is the driver of BBB formation and maintenance, J. Exp. Integr. Med., 2, 39, 10.5455/jeim.041211.hp.002
Aday, 2016, Stem cell-based human blood–brain barrier models for drug discovery and delivery, Trends Biotechnol., 34, 382, 10.1016/j.tibtech.2016.01.001
Boyer-Di Ponio, 2014, Instruction of circulating endothelial progenitors in vitro towards specialized blood-brain barrier and arterial phenotypes, PLoS One, 9
Lippmann, 2014, A retinoic acid-enhanced, multicellular human blood-brain barrier model derived from stem cell sources, Sci. Rep., 4
Abbott, 2005, Dynamics of CNS barriers: evolution, differentiation, and modulation, Cell. Mol. Neurobiol., 25, 5, 10.1007/s10571-004-1374-y
Berezowski, 2004, Transport screening of drug cocktails through an in vitro blood-brain barrier: is it a good strategy for increasing the throughput of the discovery pipeline?, Pharm. Res., 21, 756, 10.1023/B:PHAM.0000026424.78528.11
Pottiez, 2011, A differential proteomic approach identifies structural and functional components that contribute to the differentiation of brain capillary endothelial cells, J. Proteome, 75, 628, 10.1016/j.jprot.2011.09.002
Deracinois, 2013, Glial-cell-mediated re-induction of the blood-brain barrier phenotype in brain capillary endothelial cells: a differential gel electrophoresis study, Proteomics, 13, 1185, 10.1002/pmic.201200166
Sá-Pereira, 2012, Neurovascular unit: a focus on pericytes, Mol. Neurobiol., 45, 327, 10.1007/s12035-012-8244-2
Hori, 2004, A pericyte-derived angiopoietin-1 multimeric complex induces occludin gene expression in brain capillary endothelial cells through Tie-2 activation in vitro, J. Neurochem., 89, 503, 10.1111/j.1471-4159.2004.02343.x
Janzer, 1987, Astrocytes induce blood–brain barrier properties in endothelial cells, Nature, 325, 253, 10.1038/325253a0
Hatherell, 2011, Development of a three-dimensional, all-human in vitro model of the blood–brain barrier using mono-, co-, and tri-cultivation Transwell models, J. Neurosci. Methods, 199, 223, 10.1016/j.jneumeth.2011.05.012
Hayashi, 2004, Effects of hypoxia on endothelial/pericytic co-culture model of the blood–brain barrier, Regul. Pept., 123, 77, 10.1016/j.regpep.2004.05.023
Cucullo, 2002, A new dynamic in vitro model for the multidimensional study of astrocyte–endothelial cell interactions at the blood–brain barrier, Brain Res., 951, 243, 10.1016/S0006-8993(02)03167-0
Krizanac-Bengez, 2003, Effects of transient loss of shear stress on blood–brain barrier endothelium: role of nitric oxide and IL-6, Brain Res., 977, 239, 10.1016/S0006-8993(03)02689-1
McAllister, 2001, Mechanisms of glucose transport at the blood–brain barrier: an in vitro study, Brain Res., 904, 20, 10.1016/S0006-8993(01)02418-0
Stanness, 1997, Morphological and functional characterization of an in vitro blood–brain barrier model, Brain Res., 771, 329, 10.1016/S0006-8993(97)00829-9
Stanness, 1999, A new model of the blood–brain barrier: co-culture of neuronal, endothelial and glial cells under dynamic conditions, Neuroreport, 10, 3725, 10.1097/00001756-199912160-00001
Cucullo, 2011, The role of shear stress in blood-brain barrier endothelial physiology, BMC Neurosci., 12, 40, 10.1186/1471-2202-12-40
Berezowski, 2004, Contribution of glial cells and pericytes to the mRNA profiles of P-glycoprotein and multidrug resistance-associated proteins in an in vitro model of the blood–brain barrier, Brain Res., 1018, 1, 10.1016/j.brainres.2004.05.092
Grabb, 1995, Neoplastic and pharmacological influence on the permeability of an in vitro blood-brain barrier, J. Neurosurg., 82, 1053, 10.3171/jns.1995.82.6.1053
Hoheisel, 1998, Hydrocortisone reinforces the blood–brain barrier properties in a serum free cell culture system, Biochem. Biophys. Res. Commun., 244, 312, 10.1006/bbrc.1997.8051
Helms, 2016, In vitro models of the blood–brain barrier: an overview of commonly used brain endothelial cell culture models and guidelines for their use, J. Cereb. Blood Flow Metab., 36, 862, 10.1177/0271678X16630991
Cucullo, 2013, A new dynamic in vitro modular capillaries-venules modular system: cerebrovascular physiology in a box, BMC Neurosci., 14, 18, 10.1186/1471-2202-14-18
Xue, 2013, A novel brain neurovascular unit model with neurons, astrocytes and microvascular endothelial cells of rat, Int. J. Biol. Sci., 9, 174, 10.7150/ijbs.5115
Abbott, 2006, Astrocyte–endothelial interactions at the blood–brain barrier, Nat. Rev. Neurosci., 7, 41, 10.1038/nrn1824
Alvarez, 2013, Glial influence on the blood brain barrier, Glia, 61, 1939, 10.1002/glia.22575
Wolburg, 1994, Modulation of tight junction structure in blood-brain barrier endothelial cells. Effects of tissue culture, second messengers and cocultured astrocytes, J. Cell Sci., 107, 1347, 10.1242/jcs.107.5.1347
Kuntz, 2014, Stroke-induced brain parenchymal injury drives blood–brain barrier early leakage kinetics: a combined in vivo/in vitro study, J. Cereb. Blood Flow Metab., 34, 95, 10.1038/jcbfm.2013.169
Demeuse, 2002, Compartmentalized coculture of rat brain endothelial cells and astrocytes: a syngenic model to study the blood–brain barrier, J. Neurosci. Methods, 121, 21, 10.1016/S0165-0270(02)00225-X
Vandenhaute, 2012, Case study: adapting in vitro blood–brain barrier models for use in early-stage drug discovery, Drug Discov. Today, 17, 285, 10.1016/j.drudis.2011.10.006
DeBault, 1980, Gamma-glutamyl transpeptidase in isolated brain endothelial cells: induction by glial cells in vitro, Science, 207, 653, 10.1126/science.6101511
Vastag, 2009, Current in vitro and in silico models of blood-brain barrier penetration: a practical view, Curr. Opin. Drug Discov. Dev., 12, 115
Gaillard, 2001, Establishment and functional characterization of an in vitro model of the blood–brain barrier, comprising a co-culture of brain capillary endothelial cells and astrocytes, Eur. J. Pharm. Sci., 12, 215, 10.1016/S0928-0987(00)00123-8
Hori, 2004, Functional expression of rat ABCG2 on the luminal side of brain capillaries and its enhancement by astrocyte-derived soluble factor (s), J. Neurochem., 90, 526, 10.1111/j.1471-4159.2004.02537.x
Gaillard, 2000, Astrocytes increase the functional expression of P-glycoprotein in an in vitro model of the blood-brain barrier, Pharm. Res., 17, 1198, 10.1023/A:1026406528530
Al Ahmad, 2011, Astrocytes and pericytes differentially modulate blood–brain barrier characteristics during development and hypoxic insult, J. Cereb. Blood Flow Metab., 31, 693, 10.1038/jcbfm.2010.148
Terasaki, 2003, New approaches to in vitro models of blood–brain barrier drug transport, Drug Discov. Today, 8, 944, 10.1016/S1359-6446(03)02858-7
Zenker, 2003, Human blood-derived macrophages enhance barrier function of cultured primary bovine and human brain capillary endothelial cells, J. Physiol., 551, 1023, 10.1113/jphysiol.2003.045880
Rudolph, 2016, Postarrest stalling rather than crawling favors CD8+ over CD4+ T-cell migration across the blood–brain barrier under flow in vitro, Eur. J. Immunol., 46, 2187, 10.1002/eji.201546251
Choi, 2013, Size-controllable networked neurospheres as a 3D neuronal tissue model for Alzheimer's disease studies, Biomaterials, 34, 2938, 10.1016/j.biomaterials.2013.01.038
Hudecz, 2014, Reproducibility in biological models of the blood-brain barrier, Eur. J. Nanomed., 6, 185, 10.1515/ejnm-2014-0021
Siddharthan, 2007, Human astrocytes/astrocyte-conditioned medium and shear stress enhance the barrier properties of human brain microvascular endothelial cells, Brain Res., 1147, 39, 10.1016/j.brainres.2007.02.029
Prabhakarpandian, 2013, SyM-BBB: a microfluidic blood brain barrier model, Lab Chip, 13, 1093, 10.1039/c2lc41208j
Alcendor, 2013, Neurovascular unit on a chip: implications for translational applications, Stem Cell Res. Ther., 4, 1, 10.1186/scrt379
Booth, 2014, Permeability analysis of neuroactive drugs through a dynamic microfluidic in vitro blood–brain barrier model, Ann. Biomed. Eng., 42, 2379, 10.1007/s10439-014-1086-5
Park, 2015, Three-dimensional brain-on-a-chip with an interstitial level of flow and its application as an in vitro model of Alzheimer's disease, Lab Chip, 15, 141, 10.1039/C4LC00962B
Kato-Negishi, 2010, A neurospheroid network-stamping method for neural transplantation to the brain, Biomaterials, 31, 8939, 10.1016/j.biomaterials.2010.08.008
Vedadghavami, 2017, Manufacturing of hydrogel biomaterials with controlled mechanical properties for tissue engineering applications, Acta Biomater., 62, 42, 10.1016/j.actbio.2017.07.028
Huh, 2012, Microengineered physiological biomimicry: organs-on-chips, Lab Chip, 12, 2156, 10.1039/c2lc40089h
Herland, 2016, Distinct contributions of astrocytes and pericytes to neuroinflammation identified in a 3d human blood-brain barrier on a chip, PLoS One, 11, 10.1371/journal.pone.0150360
Kim, 2016, Contributions of microbiome and mechanical deformation to intestinal bacterial overgrowth and inflammation in a human gut-on-a-chip, Proc. Natl. Acad. Sci. U. S. A., 113, E7, 10.1073/pnas.1522193112
Hasan, 2015, A multilayered microfluidic blood vessel-like structure, Biomed. Microdevices, 17, 1, 10.1007/s10544-015-9993-2
Wolf, 2016, Bioengineered vascular constructs as living models for in vitro cardiovascular research, Drug Discov. Today, 21, 1446, 10.1016/j.drudis.2016.04.017
Liu, 2016, Open source 3D-printing approach for economic and fast engineering of perfusable vessel-like channels within cell-laden hydrogels, 3D Print. Addit. Manuf., 3, 22, 10.1089/3dp.2015.0025
Zarifi, 2018, Noncontact and nonintrusive microwave-microfluidic flow sensor for energy and biomedical engineering, Sci. Rep., 8, 139, 10.1038/s41598-017-18621-2
Mousavi Shaegh, 2016, A microfluidic optical platform for real-time monitoring of pH and oxygen in microfluidic bioreactors and organ-on-chip devices, Biomicrofluidics, 10, 044111, 10.1063/1.4955155
Riahi, 2016, Automated microfluidic platform of bead-based electrochemical immunosensor integrated with bioreactor for continual monitoring of cell secreted biomarkers, Sci. Rep., 6, 10.1038/srep24598
Wang, 2014, Engineering interconnected 3D vascular networks in hydrogels using molded sodium alginate lattice as the sacrificial template, Lab Chip, 14, 2709, 10.1039/C4LC00069B
Borenstein, 2010, Tissue equivalents based on cell-seeded biodegradable microfluidic constructs, Dent. Mater., 3, 1833
Hasan, 2016, Micro and nanotechnologies in heart valve tissue engineering, Biomaterials, 103, 278, 10.1016/j.biomaterials.2016.07.001
Gonzalvo, 2007, Bench-to-bedside review: brain-lung interaction in the critically ill–a pending issue revisited, Crit. Care, 11, 1, 10.1186/cc5930
Basch, 1970, Increased permeability of the blood-brain barrier following experimental thermal injury of the skin, J. Vasc. Res., 7, 357, 10.1159/000157851
Ott, 2004, Blood–brain barrier permeability to ammonia in liver failure: a critical reappraisal, Neurochem. Int., 44, 185, 10.1016/S0197-0186(03)00153-0
Zaki, 1983, Potential toxins of acute liver failure and their effects on blood-brain barrier permeability, Experientia, 39, 988, 10.1007/BF01989765
Hawkins, 2005, The blood-brain barrier/neurovascular unit in health and disease, Pharmacol. Rev., 57, 173, 10.1124/pr.57.2.4
Lv, 2010, Tumour necrosis factor-α affects blood–brain barrier permeability and tight junction-associated occludin in acute liver failure, Liver Int., 30, 1198, 10.1111/j.1478-3231.2010.02211.x
Liu, 2009
Wagner, 2013, A dynamic multi-organ-chip for long-term cultivation and substance testing proven by 3D human liver and skin tissue co-culture, Lab Chip, 13, 3538, 10.1039/c3lc50234a
Maschmeyer, 2015, A four-organ-chip for interconnected long-term co-culture of human intestine, liver, skin and kidney equivalents, Lab Chip, 15, 2688, 10.1039/C5LC00392J
Materne, 2015, A multi-organ chip co-culture of neurospheres and liver equivalents for long-term substance testing, J. Biotechnol., 205, 36, 10.1016/j.jbiotec.2015.02.002
Zhang, 2017, Multisensor-integrated organs-on-chips platform for automated and continual in situ monitoring of organoid behaviors, Proc. Natl. Acad. Sci. U. S. A., 114, E2293, 10.1073/pnas.1612906114
Rezaei Kolahchi, 2016, Microfluidic-based multi-organ platforms for drug discovery, Micromachines, 7, 162, 10.3390/mi7090162
Xu, 2015, An in vitro biomimetic multi-organ microfluidic chip system to test lung cancer metastasis, Am. J. Respir. Crit. Care Med., 191, A5014
Lee, 2017, Leukotriene B4-Mediated Neutrophil recruitment causes pulmonary capillaritis during lethal fungal sepsis, Cell Host Microbe, 23, 121, 10.1016/j.chom.2017.11.009
Seddon, 2009, Drug interactions with lipid membranes, Chem. Soc. Rev., 38, 2509, 10.1039/b813853m
Wolak, 2013, Diffusion of macromolecules in the brain: implications for drug delivery, Mol. Pharm., 10, 1492, 10.1021/mp300495e
Kaznessis, 2005, A review of methods for computational prediction of blood-brain partitioning, Curr. Med. Chem., 5, 185
Pardridge, 1999, Blood-brain barrier biology and methodology, J. Neurovirol., 5, 556, 10.3109/13550289909021285
Young, 1988, Development of a new physicochemical model for brain penetration and its application to the design of centrally acting H2 receptor histamine antagonists, J. Med. Chem., 31, 656, 10.1021/jm00398a028
Eddy, 1997, In vitro models to predict blood-brain barrier permeability, Adv. Drug Deliv. Rev., 23, 185, 10.1016/S0169-409X(96)00435-8
Reichel, 1998, Potential of immobilized artificial membranes for predicting drug penetration across the blood−brain barrier, Pharm. Res., 15, 1270, 10.1023/A:1011904311149
Feng, 2002, Assessment of blood-brain barrier penetration: in silico, in vitro and in vivo, Curr. Drug Metab., 3, 647, 10.2174/1389200023337063
Lohmann, 2002, Predicting blood-brain barrier permeability of drugs: evaluation of different in vitro assays, J. Drug Target., 10, 263, 10.1080/10611860290031903
Fan, 2010, Insights for predicting blood-brain barrier penetration of CNS targeted molecules using QSPR approaches, J. Chem. Inf. Model., 50, 1123, 10.1021/ci900384c
Carpenter, 2014, A method to predict blood-brain barrier permeability of drug-like compounds using molecular dynamics simulations, Biophys. J., 107, 630, 10.1016/j.bpj.2014.06.024
Doniger, 2002, Predicting CNS permeability of drug molecules: comparison of neural network and support vector machine algorithms, J. Comput. Biol., 9, 849, 10.1089/10665270260518317
Wu, 2012, Comparison of prediction models for blood brain barrier permeability and analysis of the molecular descriptors, Die Pharmazie, 67, 628
Adenot, 2004, Blood-brain barrier permeation models: discriminating between potential CNS and non-CNS drugs including P-glycoprotein substrates, J. Chem. Inf. Comput. Sci., 44, 239, 10.1021/ci034205d
van de Waterbeemd, 2002, High-throughput and in silico techniques in drug metabolism and pharmacokinetics, Curr. Opin. Drug Discov. Dev., 5, 33
Crivori, 2000, Predicting blood-brain barrier permeation from three-dimensional molecular structure, J. Med. Chem., 43, 2204, 10.1021/jm990968+
Rose, 2002, Modeling blood-brain barrier partitioning using the electrotopological state, J. Chem. Inf. Comput. Sci., 42, 651, 10.1021/ci010127n
Lobell, 2003, Recent advances in the prediction of blood–brain partitioning from molecular structure, J. Pharm. Sci., 92, 360, 10.1002/jps.10282
Abraham, 2004, The factors that influence permeation across the blood–brain barrier, Eur. J. Med. Chem., 39, 235, 10.1016/j.ejmech.2003.12.004
Winkler, 2004, Modelling blood–brain barrier partitioning using Bayesian neural nets, J. Mol. Graph. Model., 22, 499, 10.1016/j.jmgm.2004.03.010
Zhang, 2004, A new nonlinear equation for the tissue/blood partition coefficients of neutral compounds, J. Pharm. Sci., 93, 1595, 10.1002/jps.20084
Dureja, 2006, Topochemical models for the prediction of permeability through blood–brain barrier, Int. J. Pharm., 323, 27, 10.1016/j.ijpharm.2006.05.042
Hemmateenejad, 2006, Accurate prediction of the blood–brain partitioning of a large set of solutes using ab initio calculations and genetic neural network modeling, J. Comput. Chem., 27, 1125, 10.1002/jcc.20437
Wan, 2007, High-throughput screening of drug-brain tissue binding and in silico prediction for assessment of central nervous system drug delivery, J. Med. Chem., 50, 4606, 10.1021/jm070375w
Bendels, 2008, In silico prediction of brain and CSF permeation of small molecules using PLS regression models, Eur. J. Med. Chem., 43, 1581, 10.1016/j.ejmech.2007.11.011
Abraham, 1994, Hydrogen bonding. 33. Factors that influence the distribution of solutes between blood and brain, J. Pharm. Sci., 83, 1257, 10.1002/jps.2600830915
Abraham, 1995, Hydrogen-bonding. Part 36. Determination of blood brain distribution using octanol-water partition coefficients, Drug Des. Discov., 13, 123
Abraham, 2002, Application of hydrogen bonding calculations in property based drug design, Drug Discov. Today, 7, 1056, 10.1016/S1359-6446(02)02478-9
Levin, 1980, Relationship of octanol/water partition coefficient and molecular weight to rat brain capillary permeability, J. Med. Chem., 23, 682, 10.1021/jm00180a022
Kelder, 1999, Polar molecular surface as a dominating determinant for oral absorption and brain penetration of drugs, Pharm. Res., 16, 1514, 10.1023/A:1015040217741
Clark, 1999, Rapid calculation of polar molecular surface area and its application to the prediction of transport phenomena. 2. Prediction of blood–brain barrier penetration, J. Pharm. Sci., 88, 815, 10.1021/js980402t
Gupta, 2015, Qualitative and quantitative structure–activity relationship modelling for predicting blood-brain barrier permeability of structurally diverse chemicals, SAR QSAR Environ. Res., 26, 95, 10.1080/1062936X.2014.994562
Hou, 2003, ADME evaluation in drug discovery. 3. Modeling blood-brain barrier partitioning using simple molecular descriptors, J. Chem. Inf. Comput. Sci., 43, 2137, 10.1021/ci034134i
Clark, 2003, In silico prediction of blood–brain barrier permeation, Drug Discov. Today, 8, 927, 10.1016/S1359-6446(03)02827-7
Weaver, 2008, The importance of the domain of applicability in QSAR modeling, J. Mol. Graph. Model., 26, 1315, 10.1016/j.jmgm.2008.01.002
Borhani, 2012, The future of molecular dynamics simulations in drug discovery, J. Comput. Aided Mol. Des., 26, 15, 10.1007/s10822-011-9517-y
Deng, 2009, Computations of standard binding free energies with molecular dynamics simulations, J. Phys. Chem. B, 113, 2234, 10.1021/jp807701h
Kumar, 2013, Can we predict blood brain barrier permeability of ligands using computational approaches?, Interdisc. Sci., 5, 95, 10.1007/s12539-013-0158-9
Suenderhauf, 2012, Computational prediction of blood-brain barrier permeability using decision tree induction, Molecules, 17, 10429, 10.3390/molecules170910429
Dolghih, 2012, Predicting efflux ratios and blood-brain barrier penetration from chemical structure: combining passive permeability with active efflux by P-glycoprotein, ACS Chem. Neurosci., 4, 361, 10.1021/cn3001922
Jiang, 2015, A method to predict different mechanisms for blood–brain barrier permeability of CNS activity compounds in Chinese herbs using support vector machine, J. Bioinforma. Comput. Biol., 14, 1650005, 10.1142/S0219720016500050
Geldenhuys, 2015, Molecular determinants of blood-brain barrier permeation, Ther. Deliv., 6, 961, 10.4155/tde.15.32
Allen, 2001, Characterization of the blood–brain barrier choline transporter using the in situ rat brain perfusion technique, J. Neurochem., 76, 1032, 10.1046/j.1471-4159.2001.00093.x
Gratton, 1997, Molecular factors influencing drug transfer across the blood-brain barrier, J. Pharm. Pharmacol., 49, 1211, 10.1111/j.2042-7158.1997.tb06072.x
Liu, 2004, Development of a computational approach to predict blood-brain barrier permeability, Drug Metab. Dispos., 32, 132, 10.1124/dmd.32.1.132
Re, 2011, Functionalization of liposomes with ApoE-derived peptides at different density affects cellular uptake and drug transport across a blood-brain barrier model, Nanomedicine, 7, 551, 10.1016/j.nano.2011.05.004
Salvati, 2013, Liposomes functionalized to overcome the blood–brain barrier and to target amyloid-β peptide: the chemical design affects the permeability across an in vitro model, Int. J. Nanomedicine, 8, 1749
Leonor Pinzon-Daza, 2013, Nanoparticle-and liposome-carried drugs: new strategies for active targeting and drug delivery across blood-brain barrier, Curr. Drug Metab., 14, 625, 10.2174/1389200211314060001
Bana, 2014, Liposomes bi-functionalized with phosphatidic acid and an ApoE-derived peptide affect Aβ aggregation features and cross the blood–brain-barrier: implications for therapy of Alzheimer disease, Nanomedicine, 10, 1583, 10.1016/j.nano.2013.12.001
Salvati, 2010, vol. 156, 71
Martin-Banderas, 2011, Nanostructures for drug delivery to the brain, Curr. Med. Chem., 18, 5303, 10.2174/092986711798184262
Tosi, 2008, Polymeric nanoparticles for the drug delivery to the central nervous system, Expert Opin. Drug Deliv., 5, 155, 10.1517/17425247.5.2.155
Tosi, 2013, Potential use of polymeric nanoparticles for drug delivery across the blood-brain barrier, Curr. Med. Chem., 20, 2212, 10.2174/0929867311320170006
Clemens-Hemmelmann, 2016, Amphiphilic copolymers shuttle drugs across the blood–brain barrier, Macromol. Biosci., 16, 655, 10.1002/mabi.201500388
Beg, 2011, Dendrimers as novel systems for delivery of neuropharmaceuticals to the brain, CNS Neurol. Disord., 10, 576, 10.2174/187152711796235023
Gaillard, 2014, Blood-to-brain drug delivery using nanocarriers, 433
Bullen, 2011, Evaluation of biotinylated PAMAM dendrimer toxicity in models of the blood brain barrier: a biophysical and cellular approach, J. Biomater. Nanobiotechnol., 2, 485, 10.4236/jbnb.2011.225059
Roney, 2005, Targeted nanoparticles for drug delivery through the blood–brain barrier for Alzheimer's disease, J. Control. Release, 108, 193, 10.1016/j.jconrel.2005.07.024
Garbayo, 2013, Drug development in Parkinson's disease: from emerging molecules to innovative drug delivery systems, Maturitas, 76, 272, 10.1016/j.maturitas.2013.05.019
Wilson, 2008, Targeted delivery of tacrine into the brain with polysorbate 80-coated poly(n-butylcyanoacrylate) nanoparticles, Eur. J. Pharm. Biopharm., 70, 75, 10.1016/j.ejpb.2008.03.009
Wilson, 2008, Poly (n-butylcyanoacrylate) nanoparticles coated with polysorbate 80 for the targeted delivery of rivastigmine into the brain to treat Alzheimer's disease, Brain Res., 1200, 159, 10.1016/j.brainres.2008.01.039
Huang, 2013, Angiopep-conjugated nanoparticles for targeted long-term gene therapy of Parkinson's disease, Pharm. Res., 30, 2549, 10.1007/s11095-013-1005-8
Santra, 2001, Conjugation of biomolecules with luminophore-doped silica nanoparticles for photostable biomarkers, Anal. Chem., 73, 4988, 10.1021/ac010406+
Wang, 2013, Functionalized silica nanoparticles: a platform for fluorescence imaging at the cell and small animal levels, Acc. Chem. Res., 46, 1367, 10.1021/ar3001525
He, 2008, In vivo study of biodistribution and urinary excretion of surface-modified silica nanoparticles, Anal. Chem., 80, 9597, 10.1021/ac801882g
Gao, 2013, Glioma targeting and blood–brain barrier penetration by dual-targeting doxorubincin liposomes, Biomaterials, 34, 5628, 10.1016/j.biomaterials.2013.03.097
Hanada, 2014, Cell-based in vitro blood–brain barrier model can rapidly evaluate nanoparticles’ brain permeability in association with particle size and surface modification, Int. J. Mol. Sci., 15, 1812, 10.3390/ijms15021812
Liu, 2014, In vitro and in vivo studies on the transport of PEGylated silica nanoparticles across the blood–brain barrier, ACS Appl. Mater. Interfaces, 6, 2131, 10.1021/am405219u
Jain, 2012, Nanobiotechnology-based strategies for crossing the blood-brain barrier, Nanomedicine, 7, 1225, 10.2217/nnm.12.86
Aryal, 2014, Ultrasound-mediated blood–brain barrier disruption for targeted drug delivery in the central nervous system, Adv. Drug Deliv. Rev., 72, 94, 10.1016/j.addr.2014.01.008
Magnin, 2015, Magnetic resonance-guided motorized transcranial ultrasound system for blood-brain barrier permeabilization along arbitrary trajectories in rodents, J. Ther. Ultrasound, 3, 22, 10.1186/s40349-015-0044-5
Babak, 2012, Activation of signaling pathways following localized delivery of systemically administered neurotrophic factors across the blood–brain barrier using focused ultrasound and microbubbles, Phys. Med. Biol., 57, N65, 10.1088/0031-9155/57/7/N65
Nance, 2014, Non-invasive delivery of stealth, brain-penetrating nanoparticles across the blood − brain barrier using MRI-guided focused ultrasound, J. Control. Release, 189, 123, 10.1016/j.jconrel.2014.06.031
Samiotaki, 2012, A quantitative pressure and microbubble-size dependence study of focused ultrasound-induced blood-brain barrier opening reversibility in vivo using MRI, Magn. Reson. Med., 67, 769, 10.1002/mrm.23063
Seyfried, 2008, Mannitol Enhances Delivery of Marrow Stromal Cells to the Brain after Experimental Intracerebral Hemorrhage, Brain Res., 1224, 12, 10.1016/j.brainres.2008.05.080
Tung, 2011, The mechanism of interaction between focused ultrasound and microbubbles in blood-brain barrier opening in mice, J. Acoust. Soc. Am., 130, 3059, 10.1121/1.3646905