全部 标题 作者
关键词 摘要

OALib Journal期刊
ISSN: 2333-9721
费用:99美元

查看量下载量

相关文章

更多...

Slice Cultures as a Model to Study Neurovascular Coupling and Blood Brain Barrier In Vitro

DOI: 10.1155/2011/646958

Full-Text   Cite this paper   Add to My Lib

Abstract:

Proper neuronal functioning depends on a strictly regulated interstitial environment and tight coupling of neuronal and metabolic activity involving adequate vascular responses. These functions take place at the blood brain barrier (BBB) composed of endothelial cells, basal lamina covered with pericytes, and the endfeet of perivascular astrocytes. In conventional in vitro models of the BBB, some of these components are missing. Here we describe a new model system for studying BBB and neurovascular coupling by using confocal microscopy and fluorescence staining protocols in organotypic hippocampal slice cultures. An elaborated network of vessels is retained in culture in spite of the absence of blood flow. Application of calcein-AM either from the interstitial or from the luminal side resulted in different staining patterns indicating the maintenance of a barrier. By contrast, the ethidium derivative MitoSox penetrated perivascular basal lamina and revealed free radical formation in contractile cells embracing the vessels, likely pericytes. 1. Introduction Proper function of the central nervous system requires a meticulously controlled interstitial environment. Since its composition largely differs from that of blood plasma, its maintenance relies on selective filtering and active transport processes at the blood brain barrier (BBB). In order to keep pace with the energetic demand of neuronal activity, cerebral blood flow is tightly regulated by multiple and only partially understood mechanisms termed as neurovascular coupling. The structural substrate for BBB and neurovascular coupling is the neurovascular unit composed of tight junction coupled endothelial cells, capillary basal lamina covered with smooth muscle cells (SMCs)/pericytes, and the endfeet of perivascular astrocytes [1]. Studies on BBB and neurovascular coupling are frequently done in vivo although the exact control of systemic effects is difficult. Accordingly, the conclusions which can be drawn need careful interpretation. Studies on acute brain slices gave us new insights on the regulation of capillary microcirculation [2–5] as well as on consequences of BBB disruption [6, 7]. However, brain slices represent acutely injured tissue with severed BBB and ongoing cell damage that might negatively interfere with the mechanisms of neurovascular coupling [8, 9]. Widely used in vitro models of BBB are based on different cocultures of endothelial cells and astrocytes [10]. However, such models dismiss the intimate influence of the surrounding nervous tissue, pericytes, and perivascular microglia

References

[1]  S. Banerjee and M. A. Bhat, “Neuron-glial interactions in blood-brain barrier formation,” Annual Review of Neuroscience, vol. 30, pp. 235–258, 2007.
[2]  M. Zonta, M. C. Angulo, S. Gobbo et al., “Neuron-to-astrocyte signaling is central to the dynamic control of brain microcirculation,” Nature Neuroscience, vol. 6, no. 1, pp. 43–50, 2003.
[3]  C. Iadecola, “Neurovascular regulation in the normal brain and in Alzheimer's disease,” Nature Reviews Neuroscience, vol. 5, no. 5, pp. 347–360, 2004.
[4]  S. J. Mulligan and B. A. MacVicar, “Calcium transients in astrocyte endfeet cause cerebrovascular constrictions,” Nature, vol. 431, no. 7005, pp. 195–199, 2004.
[5]  J. A. Filosa, A. D. Bonev, S. V. Straub et al., “Local potassium signaling couples neuronal activity to vasodilation in the brain,” Nature Neuroscience, vol. 9, no. 11, pp. 1397–1403, 2006.
[6]  E. Seiffert, J. P. Dreier, S. Ivens et al., “Lasting blood-brain barrier disruption induces epileptic focus in the rat somatosensory cortex,” Journal of Neuroscience, vol. 24, no. 36, pp. 7829–7836, 2004.
[7]  Y. David, L. P. Cacheaux, S. Ivens et al., “Astrocytic dysfunction in epileptogenesis: consequence of altered potassium and glutamate homeostasis?” Journal of Neuroscience, vol. 29, no. 34, pp. 10588–10599, 2009.
[8]  H. Girouard and C. Iadecola, “Neurovascular coupling in the normal brain and in hypertension, stroke, and Alzheimer disease,” Journal of Applied Physiology, vol. 100, no. 1, pp. 328–335, 2006.
[9]  J. A. Filosa, “Vascular tone and neurovascular coupling: considerations toward an improved in vitro model,” Front Neuroenergetics, vol. 2, article 16, 2010.
[10]  M. Gumbleton and K. L. Audus, “Progress and limitations in the use of in vitro cell cultures to serve as a permeability screen for the blood-brain barrier,” Journal of Pharmaceutical Sciences, vol. 90, no. 11, pp. 1681–1698, 2001.
[11]  B. H. Gahwiler and F. Hefti, “Guidance of acetylcholinesterase-containing fibres by target tissue in co-cultured brain slices,” Neuroscience, vol. 13, no. 3, pp. 681–689, 1984.
[12]  M. Frothscher and B. H. Gahwiler, “Synaptic organization of intracellularly stained CA3 pyramidal neurons in slice cultures of rat hippocampus,” Neuroscience, vol. 24, no. 2, pp. 541–551, 1988.
[13]  L. Stoppini, P. A. Buchs, and D. Muller, “A simple method for organotypic cultures of nervous tissue,” Journal of Neuroscience Methods, vol. 37, no. 2, pp. 173–182, 1991.
[14]  S. Duport, F. Robert, D. Muller, G. Grau, L. Parisi, and L. Stoppini, “An in vitro blood-brain barrier model: cocultures between endothelial cells and organotypic brain slice cultures,” Proceedings of the National Academy of Sciences of the United States of America, vol. 95, no. 4, pp. 1840–1845, 1998.
[15]  C. M. Zehendner, H. J. Luhmann, and C. R. W. Kuhlmann, “Studying the neurovascular unit: an improved blood-brain barrier model,” Journal of Cerebral Blood Flow and Metabolism, vol. 29, no. 12, pp. 1879–1884, 2009.
[16]  K. V. Moser, R. Schmidt-Kastner, H. Hinterhuber, and C. Humpel, “Brain capillaries and cholinergic neurons persist in organotypic brain slices in the absence of blood flow,” European Journal of Neuroscience, vol. 18, no. 1, pp. 85–94, 2003.
[17]  K. Bendfeldt, V. Radojevic, J. Kapfhammer, and C. Nitsch, “Basic fibroblast growth factor modulates density of blood vessels and preserves tight junctions in organotypic cortical cultures of mice: a new in vitro model of the blood-brain barrier,” Journal of Neuroscience, vol. 27, no. 12, pp. 3260–3267, 2007.
[18]  R. S. Camenzind, S. Chip, H. Gutmann, J. P. Kapfhammer, C. Nitsch, and K. Bendfeldt, “Preservation of transendothelial glucose transporter 1 and P-glycoprotein transporters in a cortical slice culture model of the blood-brain barrier,” Neuroscience, vol. 170, no. 1, pp. 361–371, 2010.
[19]  R. Kovács, R. Gutiérrez, A. Kivi, S. Schuchmann, S. Gabriel, and U. Heinemann, “Acute cell damage after low Mg-induced epileptiform activity in organotypic hippocampal slice cultures,” NeuroReport, vol. 10, no. 2, pp. 207–213, 1999.
[20]  R. Kovács, A. Rabanus, J. Otáhal et al., “Endogenous nitric oxide is a key promoting factor for initiation of seizure-like events in hippocampal and entorhinal cortex slices,” Journal of Neuroscience, vol. 29, no. 26, pp. 8565–8577, 2009.
[21]  S. Bolte and F. P. Cordelières, “A guided tour into subcellular colocalization analysis in light microscopy,” Journal of Microscopy, vol. 224, no. 3, pp. 213–232, 2006.
[22]  A. Andreasen and G. Danscher, “Optical slicing and 3-D characterization of hippocampal capillaries in the rat visualized by autometallographic silver enhancement of colloidal gold particles,” Histochemical Journal, vol. 29, no. 10, pp. 775–781, 1997.
[23]  R. Kovács, J. Kardos, U. Heinemann, and O. Kann, “Mitochondrial calcium ion and membrane potential transients follow the pattern of epileptiform discharges in hippocampal slice cultures,” Journal of Neuroscience, vol. 25, no. 17, pp. 4260–4269, 2005.
[24]  K. M. Robinson, M. S. Janes, and J. S. Beckman, “The selective detection of mitochondrial superoxide by live cell imaging,” Nature Protocols, vol. 3, no. 6, pp. 941–947, 2008.
[25]  K. Jandová, D. P?sler, L. L. Antonio et al., “Carbamazepine-resistance in the epileptic dentate gyrus of human hippocampal slices,” Brain, vol. 129, no. 12, pp. 3290–3306, 2006.
[26]  I. Manzini and D. Schild, “Multidrug resistance transporters in the olfactory receptor neurons of Xenopus laevis tadpoles,” Journal of Physiology, vol. 546, no. 2, pp. 375–385, 2003.
[27]  G. R. J. Gordon, H. B. Choi, R. L. Rungta, G. C. R. Ellis-Davies, and B. A. MacVicar, “Brain metabolism dictates the polarity of astrocyte control over arterioles,” Nature, vol. 456, no. 7223, pp. 745–750, 2008.
[28]  C. Capone, G. Faraco, J. Anrather, P. Zhou, and C. Iadecola, “Cyclooxygenase 1-derived prostaglandin E2 and EP1 receptors are required for the cerebrovascular dysfunction induced by angiotensin II,” Hypertension, vol. 55, no. 4, pp. 911–917, 2010.
[29]  M. Yemisci, Y. Gursoy-Ozdemir, A. Vural, A. Can, K. Topalkara, and T. Dalkara, “Pericyte contraction induced by oxidative-nitrative stress impairs capillary reflow despite successful opening of an occluded cerebral artery,” Nature Medicine, vol. 15, no. 9, pp. 1031–1037, 2009.
[30]  J. Noraberg, F. R. Poulsen, M. Blaabjerg et al., “Organotypic hippocampal slice cultures for studies of brain damage, neuroprotection and neurorepair,” Current Drug Targets: CNS and Neurological Disorders, vol. 4, no. 4, pp. 435–452, 2005.
[31]  B. Lassègue and R. E. Clempus, “Vascular NAD(P)H oxidases: specific features, expression, and regulation,” American Journal of Physiology, vol. 285, no. 2, pp. R277–R297, 2003.
[32]  M. Dai, A. Nuttall, Y. Yang, and X. Shi, “Visualization and contractile activity of cochlear pericytes in the capillaries of the spiral ligament,” Hearing Research, vol. 254, no. 1-2, pp. 100–107, 2009.
[33]  P. Ballabh, A. Braun, and M. Nedergaard, “The blood-brain barrier: an overview: structure, regulation, and clinical implications,” Neurobiology of Disease, vol. 16, no. 1, pp. 1–13, 2004.
[34]  H. Shalev, Y. Serlin, and A. Friedman, “Breaching the blood-brain barrier as a gate to psychiatric disorder,” Cardiovascular Psychiatry and Neurology, vol. 2009, Article ID 278531, 7 pages, 2009.
[35]  X. E. Ndode-Ekane, N. Hayward, O. Gr?hn, and A. Pitk?nen, “Vascular changes in epilepsy: functional consequences and association with network plasticity in pilocarpine-induced experimental epilepsy,” Neuroscience, vol. 166, no. 1, pp. 312–332, 2010.
[36]  C. Iadecola, L. Park, and C. Capone, “Threats to the mind: aging, amyloid, and hypertension,” Stroke, vol. 40, no. 3, supplement, pp. S40–S44, 2009.
[37]  R. Kovács, S. Schuchmann, S. Gabriel, O. Kann, J. Kardos, and U. Heinemann, “Free radical-mediated cell damage after experimental status epilepticus in hippocampal slice cultures,” Journal of Neurophysiology, vol. 88, no. 6, pp. 2909–2918, 2002.

Full-Text

comments powered by Disqus

Contact Us

service@oalib.com

QQ:3279437679

WhatsApp +8615387084133