Diabetes Metab J.  2022 Mar;46(2):222-238. 10.4093/dmj.2021.0146.

Glial and Vascular Cell Regulation of the Blood-Brain Barrier in Diabetes

Affiliations
  • 1National Drug Clinical Trial Institution, Second Affiliated Hospital, Army Medical University, Chongqing, China

Abstract

As a structural barrier, the blood-brain barrier (BBB) is located at the interface between the brain parenchyma and blood, and modulates communication between the brain and blood microenvironment to maintain homeostasis. The BBB is composed of endothelial cells, basement membrane, pericytes, and astrocytic end feet. BBB impairment is a distinguishing and pathogenic factor in diabetic encephalopathy. Diabetes causes leakage of the BBB through downregulation of tight junction proteins, resulting in impaired functioning of endothelial cells, pericytes, astrocytes, microglia, nerve/glial antigen 2-glia, and oligodendrocytes. However, the temporal regulation, mechanisms of molecular and signaling pathways, and consequences of BBB impairment in diabetes are not well understood. Consequently, the efficacy of therapies diabetes targeting BBB leakage still lags behind the requirements. This review summarizes the recent research on the effects of diabetes on BBB composition and the potential roles of glial and vascular cells as therapeutic targets for BBB disruption in diabetic encephalopathy.

Keyword

Astrocytes; Blood-brain barrier; Diabetes mellitus; Endothelial cells; Microglia; Oligodendrocyte precursor cells; Oligodendroglia; Pericytes

Figure

  • Fig. 1. The location and connection of glia (astrocytes, microglia, nerve/glial antigen 2 [NG2-glia], and oligodendrocytes) and vascular cells (endothelial cells and pericytes) in diabetic blood-brain barrier (BBB). As the major constituents of the BBB, endothelial cells connect via gap junctional proteins and works together with various other cell types, such as astrocytes, pericytes, microglia, NG2-glia, and oligodendrocytes. Their synergistic communication contributes to the functional properties of the BBB.

  • Fig. 2. The detrimental effects of diabetes on glia and vascular cells to increase blood-brain barrier (BBB) permeability. As a basic characteristic of diabetes, hyperglycemia function as the initiating injury factor of BBB leakage, and other injury factors induced by hyperglycemia further aggravate the damage of BBB, such as advanced glycation end-products, inflammation, oxidative stress, metalloproteinase, astrocyte apoptosis, microglia polarization to swallow astrocytic end-feet, nerve/glial antigen 2 (NG2-glia) proliferation, pericyte death, and lower endothelial tight junction coverage.

  • Fig. 3. Glial and vascular cell functions and mechanisms involved in diabetic blood-brain barrier (BBB) permeability. Astrocytes modulate diabetic BBB integrity via glial-derived neurotrophic factor, nitric oxide, tumor necrosis factor-α, reactive oxygen-nitrogen, advanced glycation end-products, transforming growth factor-β, glial cell line-derived neurotrophic factor, basic fibroblast growth factor, interleukin-6, steroids, the αvβ5-integrin/glycogen synthase kinase-3β/β-catenin signaling pathway, and noncanonical hedgehog signaling. Microglia regulate diabetic BBB integrity via claudin-5, cytokines, and chemokines. Nerve/glial antigen 2 (NG2-glia) cause diabetic BBB breakdown via metalloproteinase 9 secretion and protection via the transforming growth factor β-receptor-mitogen-activated protein/extracellular signal-regulated kinase kinase/extracellular signal-regulated kinase signaling pathway. Oligodendrocytes improve diabetic BBB permeability through various soluble factors. Endothelial cells modulate BBB integrity via junction proteins, vascular cell adhesion molecule-1, intercellular adhesion molecule-1, chemokine receptor, Calcium/calmodulin-dependent protein kinase kinase α and β, adenosine receptor 2a, the C3a/C3a receptor signaling pathway, and the fibroblast growth factor receptor 1/Kelch-like ECH-associating protein 1 (Keap1)/NF-E2 related factor-2 (Nrf2) activation pathway. Pericytes regulate BBB permeability via transforming growth factor-β, vascular endothelial growth factor, fibronectin, claudin-5, integrin α1, platelet-derived growth factors receptor-β1, connexin-43, monocyte chemoattractant protein-1, CCAAT/enhancer binding protein homologous protein, and transcription factor 4.


Reference

1. 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; 359:452–9.
Article
2. Bogush M, Heldt NA, Persidsky Y. Blood brain barrier injury in diabetes: unrecognized effects on brain and cognition. J Neuroimmune Pharmacol. 2017; 12:593–601.
Article
3. Mogi M, Horiuchi M. Neurovascular coupling in cognitive impairment associated with diabetes mellitus. Circ J. 2011; 75:1042–8.
Article
4. Sharma B, Singh N. Pitavastatin and 4’-hydroxy-3’-methoxyacetophenone (HMAP) reduce cognitive dysfunction in vascular dementia during experimental diabetes. Curr Neurovasc Res. 2010; 7:180–91.
Article
5. Stranahan AM, Hao S, Dey A, Yu X, Baban B. Blood-brain barrier breakdown promotes macrophage infiltration and cognitive impairment in leptin receptor-deficient mice. J Cereb Blood Flow Metab. 2016; 36:2108–21.
Article
6. Mijnhout GS, Scheltens P, Diamant M, Biessels GJ, Wessels AM, Simsek S, et al. Diabetic encephalopathy: a concept in need of a definition. Diabetologia. 2006; 49:1447–8.
Article
7. 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:H2660–8.
Article
8. Ninomiya T. Diabetes mellitus and dementia. Curr Diab Rep. 2014; 14:487.
Article
9. Hill J, Rom S, Ramirez SH, Persidsky Y. Emerging roles of pericytes in the regulation of the neurovascular unit in health and disease. J Neuroimmune Pharmacol. 2014; 9:591–605.
Article
10. Persidsky Y, Ramirez SH, Haorah J, Kanmogne GD. Bloodbrain barrier: structural components and function under physiologic and pathologic conditions. J Neuroimmune Pharmacol. 2006; 1:223–36.
Article
11. Chen CH, Mayo JN, Gourdie RG, Johnstone SR, Isakson BE, Bearden SE. The connexin 43/ZO-1 complex regulates cerebral endothelial F-actin architecture and migration. Am J Physiol Cell Physiol. 2015; 309:C600–7.
Article
12. Daneman R. The blood-brain barrier in health and disease. Ann Neurol. 2012; 72:648–72.
Article
13. Munji RN, Soung AL, Weiner GA, Sohet F, Semple BD, Trivedi A, et al. Profiling the mouse brain endothelial transcriptome in health and disease models reveals a core blood-brain barrier dysfunction module. Nat Neurosci. 2019; 22:1892–902.
Article
14. 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; 85:775–81.
Article
15. Liu H, Zhang D, Xu X, Liu X, Wang G, Xie L, 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; 561:226–32.
Article
16. 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; 1123:245–52.
Article
17. Banks WA. The blood-brain barrier interface in diabetes mellitus: dysfunctions, mechanisms and approaches to treatment. Curr Pharm Des. 2020; 26:1438–47.
Article
18. Qiao J, Lawson CM, Rentrup KF, Kulkarni P, Ferris CF. Evaluating blood-brain barrier permeability in a rat model of type 2 diabetes. J Transl Med. 2020; 18:256.
Article
19. Tsuneoka Y, Nishimura T, Oka JI. Fluorescein permeability of the blood-brain barrier is enhanced in juvenile- but not young adult-onset type 1 diabetes in rats. Biol Pharm Bull. 2021; 44:1088–92.
Article
20. 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.
Article
21. Acharya NK, Levin EC, Clifford PM, Han M, Tourtellotte R, Chamberlain D, et al. Diabetes and hypercholesterolemia increase blood-brain barrier permeability and brain amyloid deposition: beneficial effects of the LpPLA2 inhibitor darapladib. J Alzheimers Dis. 2013; 35:179–98.
Article
22. 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; 74:70–6.
Article
23. Xu Z, Zeng W, Sun J, Chen W, Zhang R, Yang Z, 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.
Article
24. 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.
Article
25. Bouchard P, Ghitescu LD, Bendayan M. Morpho-functional studies of the blood-brain barrier in streptozotocin-induced diabetic rats. Diabetologia. 2002; 45:1017–25.
Article
26. Dai J, Vrensen GF, Schlingemann RO. Blood-brain barrier integrity is unaltered in human brain cortex with diabetes mellitus. Brain Res. 2002; 954:311–6.
Article
27. Mae MA, Li T, Bertuzzi G, Raschperger E, Vanlandewijck M, He L, et al. Prolonged systemic hyperglycemia does not cause pericyte loss and permeability at the mouse blood-brain barrier. Sci Rep. 2018; 8:17462.
Article
28. Baird TA, Parsons MW, Barber PA, Butcher KS, Desmond PM, Tress BM, et al. The influence of diabetes mellitus and hyperglycaemia on stroke incidence and outcome. J Clin Neurosci. 2002; 9:618–26.
Article
29. Lee IK, Kim HS, Bae JH. Endothelial dysfunction: its relationship with acute hyperglycaemia and hyperlipidemia. Int J Clin Pract Suppl. 2002; 129:59–64.
30. Pricci F, Leto G, Amadio L, Iacobini C, Cordone S, Catalano S, et al. Oxidative stress in diabetes-induced endothelial dysfunction involvement of nitric oxide and protein kinase C. Free Radic Biol Med. 2003; 35:683–94.
Article
31. Salmi M, Stolen C, Jousilahti P, Yegutkin GG, Tapanainen P, Janatuinen T, et al. Insulin-regulated increase of soluble vascular adhesion protein-1 in diabetes. Am J Pathol. 2002; 161:2255–62.
Article
32. Konishi M, Sakaguchi M, Lockhart SM, Cai W, Li ME, Homan EP, et al. Endothelial insulin receptors differentially control insulin signaling kinetics in peripheral tissues and brain of mice. Proc Natl Acad Sci U S A. 2017; 114:E8478–87.
Article
33. Wang B, Chandrasekera PC, Pippin JJ. Leptin- and leptin receptor-deficient rodent models: relevance for human type 2 diabetes. Curr Diabetes Rev. 2014; 10:131–45.
Article
34. D’souza AM, Neumann UH, Glavas MM, Kieffer TJ. The glucoregulatory actions of leptin. Mol Metab. 2017; 6:1052–65.
Article
35. 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; 50:202–11.
Article
36. Hoffman WH, Stamatovic SM, Andjelkovic AV. Inflammatory mediators and blood brain barrier disruption in fatal brain edema of diabetic ketoacidosis. Brain Res. 2009; 1254:138–48.
Article
37. 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; 440:354–8.
Article
38. Zhong Y, Wang JJ, Zhang SX. Intermittent but not constant high glucose induces ER stress and inflammation in human retinal pericytes. Adv Exp Med Biol. 2012; 723:285–92.
Article
39. 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; 34:1902–12.
40. Chehade JM, Haas MJ, Mooradian AD. Diabetes-related changes in rat cerebral occludin and zonula occludens-1 (ZO1) expression. Neurochem Res. 2002; 27:249–52.
41. Dias IH, Griffiths HR. Oxidative stress in diabetes: circulating advanced glycation end products, lipid oxidation and vascular disease. Ann Clin Biochem. 2014; 51(Pt 2):125–7.
Article
42. Lu QY, Chen W, Lu L, Zheng Z, Xu X. Involvement of RhoA/ROCK1 signaling pathway in hyperglycemia-induced microvascular endothelial dysfunction in diabetic retinopathy. Int J Clin Exp Pathol. 2014; 7:7268–77.
43. Mika J, Zychowska M, Popiolek-Barczyk K, Rojewska E, Przewlocka B. Importance of glial activation in neuropathic pain. Eur J Pharmacol. 2013; 716:106–19.
Article
44. Mohammad G, Siddiquei MM. Role of matrix metalloproteinase-2 and -9 in the development of diabetic retinopathy. J Ocul Biol Dis Infor. 2012; 5:1–8.
Article
45. Muranyi M, Ding C, He Q, Lin Y, Li PA. Streptozotocin-induced diabetes causes astrocyte death after ischemia and reperfusion injury. Diabetes. 2006; 55:349–55.
Article
46. Jing L, Mai L, Zhang JZ, Wang JG, Chang Y, Dong JD, et al. Diabetes inhibits cerebral ischemia-induced astrocyte activation: an observation in the cingulate cortex. Int J Biol Sci. 2013; 9:980–8.
47. Min LJ, Mogi M, Shudou M, Jing F, Tsukuda K, Ohshima K, et al. Peroxisome proliferator-activated receptor-γ activation with angiotensin II type 1 receptor blockade is pivotal for the prevention of blood-brain barrier impairment and cognitive decline in type 2 diabetic mice. Hypertension. 2012; 59:1079–88.
Article
48. Kapural M, Krizanac-Bengez Lj, Barnett G, Perl J, Masaryk T, Apollo D, et al. Serum S-100beta as a possible marker of blood-brain barrier disruption. Brain Res. 2002; 940:102–4.
49. Hovsepyan MR, Haas MJ, Boyajyan AS, Guevorkyan AA, Mamikonyan AA, Myers SE, et al. Astrocytic and neuronal biochemical markers in the sera of subjects with diabetes mellitus. Neurosci Lett. 2004; 369:224–7.
Article
50. Alvarez JI, Katayama T, Prat A. Glial influence on the blood brain barrier. Glia. 2013; 61:1939–58.
Article
51. McConnell HL, Li Z, Woltjer RL, Mishra A. Astrocyte dysfunction and neurovascular impairment in neurological disorders: correlation or causation? Neurochem Int. 2019; 128:70–84.
Article
52. Gandhi GK, Ball KK, Cruz NF, Dienel GA. Hyperglycaemia and diabetes impair gap junctional communication among astrocytes. ASN Neuro. 2010; 2:e00030.
Article
53. Wang J, Li G, Wang Z, Zhang X, Yao L, Wang F, et al. High glucose-induced expression of inflammatory cytokines and reactive oxygen species in cultured astrocytes. Neuroscience. 2012; 202:58–68.
Article
54. Heithoff BP, George KK, Phares AN, Zuidhoek IA, MunozBallester C, Robel S. Astrocytes are necessary for blood-brain barrier maintenance in the adult mouse brain. Glia. 2021; 69:436–72.
Article
55. Davalos D, Ryu JK, Merlini M, Baeten KM, Le Moan N, Petersen MA, et al. Fibrinogen-induced perivascular microglial clustering is required for the development of axonal damage in neuroinflammation. Nat Commun. 2012; 3:1227.
Article
56. Shigemoto-Mogami Y, Hoshikawa K, Sato K. Activated microglia disrupt the blood-brain barrier and induce chemokines and cytokines in a rat in vitro model. Front Cell Neurosci. 2018; 12:494.
57. Huang Z, Tan S. P2X7 receptor as a potential target for major depressive disorder. Curr Drug Targets. 2021; 22:1108–20.
Article
58. Haruwaka K, Ikegami A, Tachibana Y, Ohno N, Konishi H, Hashimoto A, et al. Dual microglia effects on blood brain barrier permeability induced by systemic inflammation. Nat Commun. 2019; 10:5816.
Article
59. Wodarski R, Clark AK, Grist J, Marchand F, Malcangio M. Gabapentin reverses microglial activation in the spinal cord of streptozotocin-induced diabetic rats. Eur J Pain. 2009; 13:807–11.
Article
60. Richa R, Yadawa AK, Chaturvedi CM. Hyperglycemia and high nitric oxide level induced oxidative stress in the brain and molecular alteration in the neurons and glial cells of laboratory mouse, Mus musculus. Neurochem Int. 2017; 104:64–79.
Article
61. Ben Assayag E, Eldor R, Korczyn AD, Kliper E, Shenhar-Tsarfaty S, Tene O, et al. Type 2 diabetes mellitus and impaired renal function are associated with brain alterations and poststroke cognitive decline. Stroke. 2017; 48:2368–74.
62. Suenaga J, Hu X, Pu H, Shi Y, Hassan SH, Xu M, et al. White matter injury and microglia/macrophage polarization are strongly linked with age-related long-term deficits in neurological function after stroke. Exp Neurol. 2015; 272:109–19.
63. Wang G, Zhang J, Hu X, Zhang L, Mao L, Jiang X, et al. Microglia/macrophage polarization dynamics in white matter after traumatic brain injury. J Cereb Blood Flow Metab. 2013; 33:1864–74.
64. Girolamo F, Errede M, Longo G, Annese T, Alias C, Ferrara G, et al. Defining the role of NG2-expressing cells in experimental models of multiple sclerosis: a biofunctional analysis of the neurovascular unit in wild type and NG2 null mice. PLoS One. 2019; 14:e0213508.
Article
65. da Rosa PM, Meira LA, Souza DO, Bobermin LD, Quincozes-Santos A, Leite MC. High-glucose medium induces cellular differentiation and changes in metabolic functionality of oligodendroglia. Mol Biol Rep. 2019; 46:4817–26.
Article
66. Yatomi Y, Tanaka R, Shimada Y, Yamashiro K, Liu M, MitomeMishima Y, et al. Type 2 diabetes reduces the proliferation and survival of oligodendrocyte progenitor cells in ishchemic white matter lesions. Neuroscience. 2015; 289:214–23.
Article
67. Niu J, Tsai HH, Hoi KK, Huang N, Yu G, Kim K, et al. Aberrant oligodendroglial-vascular interactions disrupt the blood-brain barrier, triggering CNS inflammation. Nat Neurosci. 2019; 22:709–18.
Article
68. Ma S, Wang J, Wang Y, Dai X, Xu F, Gao X, et al. Diabetes mellitus impairs white matter repair and long-term functional deficits after cerebral ischemia. Stroke. 2018; 49:2453–63.
Article
69. Fu J, Tay SS, Ling EA, Dheen ST. High glucose alters the expression of genes involved in proliferation and cell-fate specification of embryonic neural stem cells. Diabetologia. 2006; 49:1027–38.
Article
70. Royland JE, Konat GW, Wiggins RC. Myelin gene activation: a glucose sensitive critical period in development. J Neurosci Res. 1993; 36:399–404.
Article
71. Rinholm JE, Hamilton NB, Kessaris N, Richardson WD, Bergersen LH, Attwell D. Regulation of oligodendrocyte development and myelination by glucose and lactate. J Neurosci. 2011; 31:538–48.
Article
72. Yoo DY, Yim HS, Jung HY, Nam SM, Kim JW, Choi JH, et al. Chronic type 2 diabetes reduces the integrity of the bloodbrain barrier by reducing tight junction proteins in the hippocampus. J Vet Med Sci. 2016; 78:957–62.
Article
73. Argaw AT, Gurfein BT, Zhang Y, Zameer A, John GR. VEGF-mediated disruption of endothelial CLN-5 promotes bloodbrain barrier breakdown. Proc Natl Acad Sci U S A. 2009; 106:1977–82.
Article
74. Vorbrodt AW, Dobrogowska DH, Tarnawski M, Meeker HC, Carp RI. Immunogold study of altered expression of some interendothelial junctional molecules in the brain blood microvessels of diabetic scrapie-infected mice. J Mol Histol. 2006; 37:27–35.
Article
75. Yan J, Zhang Z, Shi H. HIF-1 is involved in high glucose-induced paracellular permeability of brain endothelial cells. Cell Mol Life Sci. 2012; 69:115–28.
Article
76. Sajja RK, Prasad S, Cucullo L. Impact of altered glycaemia on blood-brain barrier endothelium: an in vitro study using the hCMEC/D3 cell line. Fluids Barriers CNS. 2014; 11:8.
77. Dobi A, Rosanaly S, Devin A, Baret P, Meilhac O, Harry GJ, et al. Advanced glycation end-products disrupt brain microvascular endothelial cell barrier: the role of mitochondria and oxidative stress. Microvasc Res. 2021; 133:104098.
Article
78. Rom S, Zuluaga-Ramirez V, Gajghate S, Seliga A, Winfield M, Heldt NA, 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; 56:1883–96.
Article
79. Rom S, Heldt NA, Gajghate S, Seliga A, Reichenbach NL, Persidsky Y. Hyperglycemia and advanced glycation end products disrupt BBB and promote occludin and claudin-5 protein secretion on extracellular microvesicles. Sci Rep. 2020; 10:7274.
Article
80. Wang S, Lv W, Zhang H, Liu Y, Li L, Jefferson JR, et al. Aging exacerbates impairments of cerebral blood flow autoregulation and cognition in diabetic rats. Geroscience. 2020; 42:1387–410.
Article
81. Liu Y, Zhang H, Wang S, Guo Y, Fang X, Zheng B, et al. Reduced pericyte and tight junction coverage in old diabetic rats are associated with hyperglycemia-induced cerebrovascular pericyte dysfunction. Am J Physiol Heart Circ Physiol. 2021; 320:H549–62.
Article
82. Wang S, Cao C, Chen Z, Bankaitis V, Tzima E, Sheibani N, et al. Pericytes regulate vascular basement membrane remodeling and govern neutrophil extravasation during inflammation. PLoS One. 2012; 7:e45499.
Article
83. Warmke N, Griffin KJ, Cubbon RM. Pericytes in diabetes-associated vascular disease. J Diabetes Complications. 2016; 30:1643–50.
Article
84. Kim LA, Wong LL, Amarnani DS, Bigger-Allen AA, Hu Y, Marko CK, et al. Characterization of cells from patient-derived fibrovascular membranes in proliferative diabetic retinopathy. Mol Vis. 2015; 21:673–87.
85. Patrick P, Price TO, Diogo AL, Sheibani N, Banks WA, Shah GN. Topiramate protects pericytes from glucotoxicity: role for mitochondrial CA VA in cerebromicrovascular disease in diabetes. J Endocrinol Diabetes. 2015; 2:1–7.
86. Seo JH, Maki T, Maeda M, Miyamoto N, Liang AC, Hayakawa K, et al. Oligodendrocyte precursor cells support blood-brain barrier integrity via TGF-β signaling. PLoS One. 2014; 9:e103174.
Article
87. Winkler EA, Bell RD, Zlokovic BV. Central nervous system pericytes in health and disease. Nat Neurosci. 2011; 14:1398–405.
Article
88. Hanisch UK, Kettenmann H. Microglia: active sensor and versatile effector cells in the normal and pathologic brain. Nat Neurosci. 2007; 10:1387–94.
Article
89. Igarashi Y, Utsumi H, Chiba H, Yamada-Sasamori Y, Tobioka H, Kamimura Y, et al. Glial cell line-derived neurotrophic factor induces barrier function of endothelial cells forming the blood-brain barrier. Biochem Biophys Res Commun. 1999; 261:108–12.
Article
90. Fu J, Li L, Huo D, Zhi S, Yang R, Yang B, et al. Astrocyte-derived TGFβ1 facilitates blood-brain barrier function via noncanonical hedgehog signaling in brain microvascular endothelial cells. Brain Sci. 2021; 11:77.
Article
91. Allen CL, Bayraktutan U. Antioxidants attenuate hyperglycaemia-mediated brain endothelial cell dysfunction and bloodbrain barrier hyperpermeability. Diabetes Obes Metab. 2009; 11:480–90.
Article
92. Yun JH, Park SW, Kim JH, Park YJ, Cho CH, Kim JH. Angiopoietin 2 induces astrocyte apoptosis via αvβ5-integrin signaling in diabetic retinopathy. Cell Death Dis. 2016; 7:e2101.
Article
93. da Fonseca AC, Matias D, Garcia C, Amaral R, Geraldo LH, Freitas C, et al. The impact of microglial activation on bloodbrain barrier in brain diseases. Front Cell Neurosci. 2014; 8:362.
Article
94. Jiao H, Wang Z, Liu Y, Wang P, Xue Y. Specific role of tight junction proteins claudin-5, occludin, and ZO-1 of the bloodbrain barrier in a focal cerebral ischemic insult. J Mol Neurosci. 2011; 44:130–9.
Article
95. Seyedsadr MS, Weinmann O, Amorim A, Ineichen BV, Egger M, Mirnajafi-Zadeh J, et al. Inactivation of sphingosine-1-phosphate receptor 2 (S1PR2) decreases demyelination and enhances remyelination in animal models of multiple sclerosis. Neurobiol Dis. 2019; 124:189–201.
Article
96. Yang Y, Salayandia VM, Thompson JF, Yang LY, Estrada EY, Yang Y. Attenuation of acute stroke injury in rat brain by minocycline promotes blood-brain barrier remodeling and alternative microglia/macrophage activation during recovery. J Neuroinflammation. 2015; 12:26.
Article
97. Arai K, Lo EH. An oligovascular niche: cerebral endothelial cells promote the survival and proliferation of oligodendrocyte precursor cells. J Neurosci. 2009; 29:4351–5.
Article
98. Kimura I, Dohgu S, Takata F, Matsumoto J, Watanabe T, Iwao T, et al. Oligodendrocytes upregulate blood-brain barrier function through mechanisms other than the PDGF-BB/ PDGFRα pathway in the barrier-tightening effect of oligodendrocyte progenitor cells. Neurosci Lett. 2020; 715:134594.
99. Maki T, Maeda M, Uemura M, Lo EK, Terasaki Y, Liang AC, et al. Potential interactions between pericytes and oligodendrocyte precursor cells in perivascular regions of cerebral white matter. Neurosci Lett. 2015; 597:164–9.
Article
100. Butt AM, Ibrahim M, Ruge FM, Berry M. Biochemical subtypes of oligodendrocyte in the anterior medullary velum of the rat as revealed by the monoclonal antibody Rip. Glia. 1995; 14:185–97.
Article
101. Orthmann-Murphy JL, Abrams CK, Scherer SS. Gap junctions couple astrocytes and oligodendrocytes. J Mol Neurosci. 2008; 35:101–16.
Article
102. Arai K, Lo EH. Astrocytes protect oligodendrocyte precursor cells via MEK/ERK and PI3K/Akt signaling. J Neurosci Res. 2010; 88:758–63.
Article
103. Kato S, Aoyama M, Kakita H, Hida H, Kato I, Ito T, et al. Endogenous erythropoietin from astrocyte protects the oligodendrocyte precursor cell against hypoxic and reoxygenation injury. J Neurosci Res. 2011; 89:1566–74.
Article
104. Wang Y, Su Y, Yu G, Wang X, Chen X, Yu B, et al. Reduced oligodendrocyte precursor cell impairs astrocytic development in early life stress. Adv Sci (Weinh). 2021; 8:e2101181.
Article
105. Yang Y, Kimura-Ohba S, Thompson JF, Salayandia VM, Cosse M, Raz L, et al. Vascular tight junction disruption and angiogenesis in spontaneously hypertensive rat with neuroinflammatory white matter injury. Neurobiol Dis. 2018; 114:95–110.
Article
106. Zhao Q, Zhang F, Yu Z, Guo S, Liu N, Jiang Y, et al. HDAC3 inhibition prevents blood-brain barrier permeability through Nrf2 activation in type 2 diabetes male mice. J Neuroinflammation. 2019; 16:103.
Article
107. Yu Z, Lin L, Jiang Y, Chin I, Wang X, Li X, et al. Recombinant FGF21 protects against blood-brain barrier leakage through Nrf2 upregulation in type 2 diabetes mice. Mol Neurobiol. 2019; 56:2314–27.
Article
108. 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; 93:666–77.
Article
109. Yamamoto M, Guo DH, Hernandez CM, Stranahan AM. Endothelial Adora2a activation promotes blood-brain barrier breakdown and cognitive impairment in mice with diet-induced insulin resistance. J Neurosci. 2019; 39:4179–92.
Article
110. Haseloff RF, Blasig IE, Bauer HC, Bauer H. In search of the astrocytic factor(s) modulating blood-brain barrier functions in brain capillary endothelial cells in vitro. Cell Mol Neurobiol. 2005; 25:25–39.
Article
111. Armulik A, Genove G, Mae M, Nisancioglu MH, Wallgard E, Niaudet C, et al. Pericytes regulate the blood-brain barrier. Nature. 2010; 468:557–61.
Article
112. Hori S, Ohtsuki S, Hosoya K, Nakashima E, Terasaki T. A pericyte-derived angiopoietin-1 multimeric complex induces occludin gene expression in brain capillary endothelial cells through Tie-2 activation in vitro. J Neurochem. 2004; 89:503–13.
Article
113. Daneman R, Zhou L, Kebede AA, Barres BA. Pericytes are required for blood-brain barrier integrity during embryogenesis. Nature. 2010; 468:562–6.
Article
114. Tien T, Muto T, Barrette K, Challyandra L, Roy S. Downregulation of connexin 43 promotes vascular cell loss and excess permeability associated with the development of vascular lesions in the diabetic retina. Mol Vis. 2014; 20:732–41.
115. Yang Y, Yang LY, Salayandia VM, Thompson JF, Torbey M, Yang Y. Treatment with atorvastatin during vascular remodeling promotes pericyte-mediated blood-brain barrier maturation following ischemic stroke. Transl Stroke Res. 2021; 12:905–22.
Article
Full Text Links
  • DMJ
Actions
Cited
CITED
export Copy
Close
Share
  • Twitter
  • Facebook
Similar articles
Copyright © 2024 by Korean Association of Medical Journal Editors. All rights reserved.     E-mail: koreamed@kamje.or.kr