Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Article
  • Published:

Astrocytes mediate neurovascular signaling to capillary pericytes but not to arterioles

A Publisher Correction to this article was published on 13 July 2020

A Corrigendum to this article was published on 26 July 2017

This article has been updated

Abstract

Active neurons increase their energy supply by dilating nearby arterioles and capillaries. This neurovascular coupling underlies blood oxygen level–dependent functional imaging signals, but its mechanism is controversial. Canonically, neurons release glutamate to activate metabotropic glutamate receptor 5 (mGluR5) on astrocytes, evoking Ca2+ release from internal stores, activating phospholipase A2 and generating vasodilatory arachidonic acid derivatives. However, adult astrocytes lack mGluR5, and knockout of the inositol 1,4,5-trisphosphate receptors that release Ca2+ from stores does not affect neurovascular coupling. We now show that buffering astrocyte Ca2+ inhibits neuronally evoked capillary dilation, that astrocyte [Ca2+]i is raised not by release from stores but by entry through ATP-gated channels, and that Ca2+ generates arachidonic acid via phospholipase D2 and diacylglycerol lipase rather than phospholipase A2. In contrast, dilation of arterioles depends on NMDA receptor activation and Ca2+-dependent NO generation by interneurons. These results reveal that different signaling cascades regulate cerebral blood flow at the capillary and arteriole levels.

This is a preview of subscription content, access via your institution

Access options

Buy this article

Prices may be subject to local taxes which are calculated during checkout

Figure 1: Neuronal activity evokes capillary dilation.
Figure 2: P2X1-evoked astrocyte Ca2+ signaling mediates capillary-level neurovascular coupling.
Figure 3: Neuronally evoked astrocyte [Ca2+]i rise depends on P2X1 receptors.
Figure 4: AA metabolites mediating stimulation-evoked capillary dilation.
Figure 5: PLD2, not PLA2, initiates neurovascular coupling at the capillary level.
Figure 6: Neurovascular signaling to arterioles is mediated by NMDAR and NOS activity, and not by astrocyte Ca2+.
Figure 7: Neurovascular signaling to capillaries in vivo is mediated by P2X1 receptors.

Similar content being viewed by others

Change history

  • 05 December 2016

    In the version of this article initially published, the abstract referred to diacylglycerol kinase; this should have been diacylglycerol lipase. The error has been corrected in the HTML and PDF versions of the article.

  • 13 July 2020

    A Correction to this paper has been published: https://doi.org/10.1038/s41593-020-0680-0

References

  1. Zonta, M. et al. Neuron-to-astrocyte signaling is central to the dynamic control of brain microcirculation. Nat. Neurosci. 6, 43–50 (2003).

    Article  CAS  PubMed  Google Scholar 

  2. Mulligan, S.J. & MacVicar, B.A. Calcium transients in astrocyte endfeet cause cerebrovascular constrictions. Nature 431, 195–199 (2004).

    Article  CAS  PubMed  Google Scholar 

  3. Takano, T. et al. Astrocyte-mediated control of cerebral blood flow. Nat. Neurosci. 9, 260–267 (2006).

    Article  CAS  PubMed  Google Scholar 

  4. Zimmermann, K.W. Der feinere Bau der Blutcapillaren. Z. Anat. Entwicklungsgesch. 68, 29–109 (1923).

    Article  Google Scholar 

  5. Peppiatt, C.M., Howarth, C., Mobbs, P. & Attwell, D. Bidirectional control of CNS capillary diameter by pericytes. Nature 443, 700–704 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Hall, C.N. et al. Capillary pericytes regulate cerebral blood flow in health and disease. Nature 508, 55–60 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Hamel, E. Perivascular nerves and the regulation of cerebrovascular tone. J. Appl. Physiol. 100, 1059–1064 (2006).

    Article  PubMed  Google Scholar 

  8. Attwell, D. et al. Glial and neuronal control of brain blood flow. Nature 468, 232–243 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Mishra, A., Hamid, A. & Newman, E.A. Oxygen modulation of neurovascular coupling in the retina. Proc. Natl. Acad. Sci. USA 108, 17827–17831 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Nizar, K. et al. In vivo stimulus-induced vasodilation occurs without IP3 receptor activation and may precede astrocytic calcium increase. J. Neurosci. 33, 8411–8422 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Winship, I.R., Plaa, N. & Murphy, T.H. Rapid astrocyte calcium signals correlate with neuronal activity and onset of the hemodynamic response in vivo. J. Neurosci. 27, 6268–6272 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Reeves, A.M., Shigetomi, E. & Khakh, B.S. Bulk loading of calcium indicator dyes to study astrocyte physiology: key limitations and improvements using morphological maps. J. Neurosci. 31, 9353–9358 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Lind, B.L., Brazhe, A.R., Jessen, S.B., Tan, F.C. & Lauritzen, M.J. Rapid stimulus-evoked astrocyte Ca2+ elevations and hemodynamic responses in mouse somatosensory cortex in vivo. Proc. Natl. Acad. Sci. USA 110, E4678–E4687 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Otsu, Y. et al. Calcium dynamics in astrocyte processes during neurovascular coupling. Nat. Neurosci. 18, 210–218 (2015).

    Article  CAS  PubMed  Google Scholar 

  15. Sun, W. et al. Glutamate-dependent neuroglial calcium signaling differs between young and adult brain. Science 339, 197–200 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Bonder, D.E. & McCarthy, K.D. Astrocytic Gq-GPCR-linked IP3R-dependent Ca2+ signaling does not mediate neurovascular coupling in mouse visual cortex in vivo. J. Neurosci. 34, 13139–13150 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Seifert, G. & Steinhäuser, C. Glial cells in the mouse hippocampus express AMPA receptors with an intermediate Ca2+ permeability. Eur. J. Neurosci. 7, 1872–1881 (1995).

    Article  CAS  PubMed  Google Scholar 

  18. Lalo, U., Pankratov, Y., Kirchhoff, F., North, R.A. & Verkhratsky, A. NMDA receptors mediate neuron-to-glia signaling in mouse cortical astrocytes. J. Neurosci. 26, 2673–2683 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Lalo, U. et al. P2X1 and P2X5 subunits form the functional P2X receptor in mouse cortical astrocytes. J. Neurosci. 28, 5473–5480 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Shigetomi, E., Jackson-Weaver, O., Huckstepp, R.T., O'Dell, T.J. & Khakh, B.S. TRPA1 channels are regulators of astrocyte basal calcium levels and long-term potentiation via constitutive D-serine release. J. Neurosci. 33, 10143–10153 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Gordon, G.R., Choi, H.B., Rungta, R.L., Ellis-Davies, G.C. & MacVicar, B.A. Brain metabolism dictates the polarity of astrocyte control over arterioles. Nature 456, 745–749 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Mishra, A. et al. Imaging pericytes and capillary diameter in brain slices and isolated retinae. Nat. Protoc. 9, 323–336 (2014).

    Article  CAS  PubMed  Google Scholar 

  23. Panatier, A. & Robitaille, R. Astrocytic mGluR5 and the tripartite synapse. Neuroscience 323, 29–34 (2016).

    Article  CAS  PubMed  Google Scholar 

  24. Butt, A.M. ATP: a ubiquitous gliotransmitter integrating neuron-glial networks. Semin. Cell Dev. Biol. 22, 205–213 (2011).

    Article  CAS  PubMed  Google Scholar 

  25. Haustein, M.D. et al. Conditions and constraints for astrocyte calcium signaling in the hippocampal mossy fiber pathway. Neuron 82, 413–429 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. North, R.A. Molecular physiology of P2X receptors. Physiol. Rev. 82, 1013–1067 (2002).

    Article  CAS  PubMed  Google Scholar 

  27. Shigetomi, E. et al. Imaging calcium microdomains within entire astrocyte territories and endfeet with GCaMPs expressed using adeno-associated viruses. J. Gen. Physiol. 141, 633–647 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Panatier, A. et al. Astrocytes are endogenous regulators of basal transmission at central synapses. Cell 146, 785–798 (2011).

    Article  CAS  PubMed  Google Scholar 

  29. Di Castro, M.A. et al. Local Ca2+ detection and modulation of synaptic release by astrocytes. Nat. Neurosci. 14, 1276–1284 (2011).

    Article  CAS  PubMed  Google Scholar 

  30. Meng, W., Tobin, J.R. & Busija, D.W. Glutamate-induced cerebral vasodilation is mediated by nitric oxide through N-methyl-D-aspartate receptors. Stroke 26, 857–862; discussion 863 (1995).

    Article  CAS  PubMed  Google Scholar 

  31. Stobart, J.L., Lu, L., Anderson, H.D., Mori, H. & Anderson, C.M. Astrocyte-induced cortical vasodilation is mediated by D-serine and endothelial nitric oxide synthase. Proc. Natl. Acad. Sci. USA 110, 3149–3154 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Alkayed, N.J. et al. Molecular characterization of an arachidonic acid epoxygenase in rat brain astrocytes. Stroke 27, 971–979 (1996).

    Article  CAS  PubMed  Google Scholar 

  33. Stephenson, D.T. et al. Calcium-sensitive cytosolic phospholipase A2 (cPLA2) is expressed in human brain astrocytes. Brain Res. 637, 97–105 (1994).

    Article  CAS  PubMed  Google Scholar 

  34. Cockcroft, S. Signalling roles of mammalian phospholipase D1 and D2. Cell. Mol. Life Sci. 58, 1674–1687 (2001).

    Article  CAS  PubMed  Google Scholar 

  35. Sarri, E., Pardo, R., Fensome-Green, A. & Cockcroft, S. Endogenous phospholipase D2 localizes to the plasma membrane of RBL-2H3 mast cells and can be distinguished from ADP ribosylation factor-stimulated phospholipase D1 activity by its specific sensitivity to oleic acid. Biochem. J. 369, 319–329 (2003).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Yemisci, M. et al. Pericyte contraction induced by oxidative-nitrative stress impairs capillary reflow despite successful opening of an occluded cerebral artery. Nat. Med. 15, 1031–1037 (2009).

    Article  CAS  PubMed  Google Scholar 

  37. Duffy, S. & MacVicar, B.A. Adrenergic calcium signaling in astrocyte networks within the hippocampal slice. J. Neurosci. 15, 5535–5550 (1995).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Calcinaghi, N. et al. Metabotropic glutamate receptor mGluR5 is not involved in the early hemodynamic response. J. Cereb. Blood Flow Metab. 31, e1–e10 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Srinivasan, R. et al. Ca2+ signaling in astrocytes from Ip3r2−/− mice in brain slices and during startle responses in vivo. Nat. Neurosci. 18, 708–717 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Wells, J.A. et al. A critical role for purinergic signalling in the mechanisms underlying generation of BOLD fMRI responses. J. Neurosci. 35, 5284–5292 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Wieraszko, A., Goldsmith, G. & Seyfried, T.N. Stimulation-dependent release of adenosine triphosphate from hippocampal slices. Brain Res. 485, 244–250 (1989).

    Article  CAS  PubMed  Google Scholar 

  42. Zhang, Y. et al. An RNA-sequencing transcriptome and splicing database of glia, neurons, and vascular cells of the cerebral cortex. J. Neurosci. 34, 11929–11947 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Tricoire, L. & Vitalis, T. Neuronal nitric oxide synthase expressing neurons: a journey from birth to neuronal circuits. Front. Neural Circuits 6, 82 (2012).

    Article  PubMed  PubMed Central  Google Scholar 

  44. Schmidt, H.H., Pollock, J.S., Nakane, M., Förstermann, U. & Murad, F. Ca2+/calmodulin-regulated nitric oxide synthases. Cell Calcium 13, 427–434 (1992).

    Article  CAS  PubMed  Google Scholar 

  45. Lindauer, U., Megow, D., Matsuda, H. & Dirnagl, U. Nitric oxide: a modulator, but not a mediator, of neurovascular coupling in rat somatosensory cortex. Am. J. Physiol. 277, H799–H811 (1999).

    CAS  PubMed  Google Scholar 

  46. Metea, M.R. & Newman, E.A. Glial cells dilate and constrict blood vessels: a mechanism of neurovascular coupling. J. Neurosci. 26, 2862–2870 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Lecrux, C., Kocharyan, A., Sandoe, C.H., Tong, X.K. & Hamel, E. Pyramidal cells and cytochrome P450 epoxygenase products in the neurovascular coupling response to basal forebrain cholinergic input. J. Cereb. Blood Flow Metab. 32, 896–906 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Farooqui, A.A., Yang, H.C., Rosenberger, T.A. & Horrocks, L.A. Phospholipase A2 and its role in brain tissue. J. Neurochem. 69, 889–901 (1997).

    Article  CAS  PubMed  Google Scholar 

  49. Zhang, Y. et al. Increased expression of two phospholipase D isoforms during experimentally induced hippocampal mossy fiber outgrowth. Glia 46, 74–83 (2004).

    Article  PubMed  Google Scholar 

  50. Blinder, P. et al. The cortical angiome: an interconnected vascular network with noncolumnar patterns of blood flow. Nat. Neurosci. 16, 889–897 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Biesecker et al. Glial cell calcium signaling mediates capillary regulation of blood flow in the retina. J. Neurosci. 36, 9435–9445 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Kur, J. & Newman, E.A. Purinergic control of vascular tone in the retina. J. Physiol. (Lond.) 592, 491–504 (2014).

    Article  CAS  Google Scholar 

  53. Peters, B.P. & Goldstein, I.J. The use of fluorescein-conjugated Bandeiraea simplicifolia B4-isolectin as a histochemical reagent for the detection of alpha-D-galactopyranosyl groups. Their occurrence in basement membranes. Exp. Cell Res. 120, 321–334 (1979).

    Article  CAS  PubMed  Google Scholar 

  54. Shen, Z., Lu, Z., Chhatbar, P.Y., O'Herron, P. & Kara, P. An artery-specific fluorescent dye for studying neurovascular coupling. Nat. Methods 9, 273–276 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. Nakatsuka, A., Mizuno, R., Ono, N., Nakayama, J. & Ohhashi, T. Arachidonic acid-induced COX-1 and COX-2-mediated vasodilation in rat gingival arterioles in vivo. Jpn. J. Physiol. 55, 293–302 (2005).

    Article  CAS  PubMed  Google Scholar 

  56. Ito, S. et al. Roles of stretch-activated cation channel and Rho-kinase in the spontaneous contraction of airway smooth muscle. Eur. J. Pharmacol. 552, 135–142 (2006).

    Article  CAS  PubMed  Google Scholar 

  57. Wang, M.H. et al. Cytochrome P450-derived arachidonic acid metabolism in the rat kidney: characterization of selective inhibitors. J. Pharmacol. Exp. Ther. 284, 966–973 (1998).

    CAS  PubMed  Google Scholar 

  58. Bley, K.R. et al. RO1138452 and RO3244794: characterization of structurally distinct, potent and selective IP (prostacyclin) receptor antagonists. Br. J. Pharmacol. 147, 335–345 (2006).

    Article  CAS  PubMed  Google Scholar 

  59. Myren, M., Olesen, J. & Gupta, S. Pharmacological and expression profile of the prostaglandin I2 receptor in the rat craniovascular system. Vascul. Pharmacol. 55, 50–58 (2011).

    Article  CAS  PubMed  Google Scholar 

  60. Shin, K.J. et al. Phospholipase A2-mediated Ca2+ influx by 2,2′,4,6-tetrachlorobiphenyl in PC12 cells. Toxicol. Appl. Pharmacol. 178, 37–43 (2002).

    Article  CAS  PubMed  Google Scholar 

  61. Smith, R.J. et al. Receptor-coupled signal transduction in human polymorphonuclear neutrophils: effects of a novel inhibitor of phospholipase C-dependent processes on cell responsiveness. J. Pharmacol. Exp. Ther. 253, 688–697 (1990).

    CAS  PubMed  Google Scholar 

  62. Su, W. et al. 5-Fluoro-2-indolyl des-chlorohalopemide (FIPI), a phospholipase D pharmacological inhibitor that alters cell spreading and inhibits chemotaxis. Mol. Pharmacol. 75, 437–446 (2009).

    Article  CAS  PubMed  Google Scholar 

  63. Scott, S.A. et al. Design of isoform-selective phospholipase D inhibitors that modulate cancer cell invasiveness. Nat. Chem. Biol. 5, 108–117 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  64. Weigl, L., Zidar, A., Gscheidlinger, R., Karel, A. & Hohenegger, M. Store operated Ca2+ influx by selective depletion of ryanodine sensitive Ca2+ pools in primary human skeletal muscle cells. Naunyn Schmiedebergs Arch. Pharmacol. 367, 353–363 (2003).

    Article  CAS  PubMed  Google Scholar 

  65. Rettinger, J. et al. Profiling at recombinant homomeric and heteromeric rat P2X receptors identifies the suramin analogue NF449 as a highly potent P2X1 receptor antagonist. Neuropharmacology 48, 461–468 (2005).

    Article  CAS  PubMed  Google Scholar 

  66. Soto, F., Lambrecht, G., Nickel, P., Stühmer, W. & Busch, A.E. Antagonistic properties of the suramin analogue NF023 at heterologously expressed P2X receptors. Neuropharmacology 38, 141–149 (1999).

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

We thank K. Zheng for help with two-photon microscopy, and M. Carandini, I. Christie, S. Cockcroft, M. Ford, R. Jolivet, S. Mastitskaya and S. Sullivan for comments on the manuscript. Supported by the European Research Council (BRAINPOWER to D.A. and NETSIGNAL to D.A.R.), a Fondation Leducq Transatlantic Network grant (to D.A.), a Wellcome Trust Programme Grant and Senior Investigator Award (075232 and 099222 to D.A.), a Wellcome Trust Senior Research Fellowship (095064 and 200893 to A.V.G.), a Wellcome Trust Principal Research Fellowship (101896 to D.A.R.), an EU FP7 grant (ITN EXTRABRAIN 606950 to D.A.R.) and an RSF grant (15-14-30000 to D.A.R.).

Author information

Authors and Affiliations

Authors

Contributions

A.M. and D.A. conceived the study. A.M. carried out all brain slice experiments, some immunocytochemistry, and analysis of brain slice and in vivo data; J.P.R. performed in vivo experiments and analyzed the resulting data; Y.C. carried out immunocytochemistry; A.V.G. and D.A.R. provided in vivo imaging expertise; A.M. and D.A. wrote the paper; all authors revised the paper.

Corresponding author

Correspondence to David Attwell.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Integrated supplementary information

Supplementary Figure 1 Schematic diagram of the mechanisms underlying neurovascular signalling to capillaries and arterioles.

Synaptic activity (top) evokes ATP release from post-synaptic neurons, which activates ionotropic ATP receptors containing P2X1 subunits on astrocytes, leading to a rise in [Ca2+]i via influx from the extracellular space. This rise in [Ca2+]i activates PLD2, resulting in the formation of phosphatidic acid (PA) which is converted into diacylglycerol (DAG), which is then further metabolized by DAG lipase into arachidonic acid (AA). AA is then metabolized, by the consecutive activity of COX1 and PGES, to produce PGE2, which dilates capillaries by acting on EP4 receptors, presumably on pericytes. Synaptic activation of Ca2+ entry through NMDA receptors increases NOS activity in interneurons, resulting in NO release onto arterioles to dilate them. The neurovascular coupling pathways involved in signalling to capillaries and arterioles are highlighted in blue, while the enzymes and receptors ruled out by our experiments are shown in red with black crosses.

Supplementary Figure 2 Stimulation-evoked field potentials and identification of vessels.

(a-b) A schematic (a) and a low-magnification image (b) of a cortical slice demonstrating the placement of the stimulation and recording electrodes. Cortical layers are indicated. Inset shows a high magnification image of the recording electrode near a capillary. (c) Stimulation-evoked fibre volley and field excitatory post-synaptic currents (fEPSCs) are not affected by U46619. NBQX blocks the fEPSCs but not the fibre volley and TTX blocks both the fibre volley and the fEPSCs. (d) Mean data (peak fEPSC amplitude measured 5 to 9 ms after the stimulation) showing the effect of U46619 and NBQX on fEPSCs. (e) Arterioles can be distinguished by the thick layer of smooth muscle cells (SMC, white brackets) that surround them, shown here alongside FITC-isolectin B4 (IB4) labeling of the basement membrane. (f, g) Capillaries are identified as smaller vessels that have only occasional pericyte cell bodies (arrowheads) outlined by the IB4-labeling. IB4 also labels microglia (asterisk). Vessel lumen is marked by red brackets. Data shown as mean±s.e.m.

Supplementary Figure 3 Constriction of cortical capillaries evoked by U46619 before each experiment.

(a) Example trace of a U46619 (200 nM) induced capillary constriction lasting at least 30 minutes. (b-x) Mean constriction of capillaries to U46619 in interleaved control and drug experiments for the: (b) voltage-gated sodium channel blocker TTX, (c) AMPA/KA receptor blocker NBQX, (d) NMDA receptor blocker D-AP5, (e) fast Ca2+ chelator BAPTA, (f) inhibitor of group I and II mGluRs S-MCPG, (g) P2Y1 blocker MRS2179, (h) TRPA1 blocker A967079, (i) P2X1 blocker NF449, (j) P2X1 blocker NF023, (k) COX1 blocker SC-560, (l) COX2 blocker NS-398, (m) epoxygenase blocker PPOH, (n) EP4 receptor blocker L-161,982, (o) IP receptor blocker CAY10441, (p) NO synthase blocker L-NNA, (q) PLA2 blocker MAFP, (r) PLC blocker U73122, (s) PLD blocker FIPI, (t) PLD1 blocker VU0155069, (u) PLD2 blocker CAY10594, (v) DAGL blocker RHC80267, (w) P2X1 agonist α,β-meATP, and (x) PGE2 in the presence or absence of the PLD blocker FIPI. P-values comparing U46619-evoked constrictions for control and drug experiments were >0.05 for all panels even without correcting for multiple comparisons. For experiments involving BAPTA (e), PPOH (m) and L-161,982 (n), U-46619 was applied in the presence of the relevant drug (see Methods). For all other experiments, vessels were pre-constricted with U46619 before drug application. Data shown as mean±s.e.m.

Supplementary Figure 4 Signaling to capillary pericytes is similar for different stimulation durations in P21 rats, and in P45 rats.

Mean dilation (left panels) and time traces of the diameter change (right panels) observed in cortical capillaries following electrical stimulation of neuronal activity (at 100 Hz for 0.2 sec, repeated once/sec) in the absence and presence of P2X1 blockers. (a-c) Capillary dilation evoked by short (200 ms; a), medium (5 sec; b) or long duration (1 min; c, reproduced from Fig. 2l) stimulation in slices from P21 rats is inhibited by the P2X1 blocker NF449 (100 nM). (d) Stimulation-evoked capillary dilation in slices from P21 rats is inhibited by a second P2X1 blocker NF023 (5 μM). (e) Stimulation-evoked capillary dilation in slices from P45 rats is also inhibited by the P2X1 blocker NF449 (100 nM). Data shown as mean±s.e.m.

Supplementary Figure 5 COX1 and PGES are expressed in astrocyte endfeet along vessels.

(a) AQP4-expressing astrocyte endfeet along arterioles (arrows) and capillaries (arrowheads) are immunoreactive for COX1. Vascular basement membrane is labeled with Alexa dye conjugated isolectin B4 (IB4). (b) COX2 is absent from GFAP-expressing astrocyte somata (white asterisk) and endfeet (arrowhead) but is expressed in neuronal cell bodies (black asterisk). (c) Epoxygenase (CYP2C11) is expressed in AQP4-expressing astrocyte endfeet along arterioles (arrows) but not in those along capillaries (arrowhead). Arterioles can be identified as larger vessels, often with space between layers of IB4-labeled basement membrane (white circle) where the vascular smooth muscle cells are located. (d) PGES is expressed in GFAP-expressing astrocyte cell bodies (white asterisk), processes and endfeet along vessels (arrowheads). (e) Control experiments where slices were incubated with the secondary antibody used to stain the enzyme shown in the corresponding panels in a-d, but with the primary antibody omitted, demonstrating the lack of non-specific binding. Control for the secondary antibody used to detect COX1 is shown in the top row, COX2 in the second row, epoxygenase in third row and PGES in the fourth row.

Supplementary Figure 6 PLA2 is expressed in GFAP-labeled astrocyte endfeet along the vasculature.

Arrowhead indicates a pericyte cell body.

Supplementary Figure 7 PLD is upstream of PGE2 in the signaling pathway.

(a) After preconstriction with 200 nM U46619, applying 1 μM PGE2 evokes a dilation. (b) A similar PGE2-evoked dilation was seen in the presence of 1 μM FIPI to block PLD1 and PLD2. (c) Quantification of data from experiments like those in panels a-b, showing that PLD inhibition does not affect the capillary dilation evoked by PGE2. Data shown as mean±s.e.m.

Supplementary Figure 8 PLD1 and PLD2 expression in the cortex.

(a, b) PLD1 was diffusely expressed in the cortical neuropil and appeared concentrated in endothelial cells along arterioles (arrow, a) but not capillaries (b). No PLD1 labeling was observed in AQP4-labeled endfeet (arrowheads). (c, d) PLD2 expression was detected in GFAP-expressing astrocyte endfeet along capillaries (arrowheads, c) as well as in astrocyte somata (asterisks, d). (e) Control experiments where slices were labeled with the secondary antibody used to detect the enzymes shown in the corresponding panels in a-d, but with the primary antibody omitted, demonstrating the lack of non-specific binding. Control for secondary antibodies used to detect PLD1 is shown in the top two panels and for PLD2 is shown in the last two panels (for these pictures illumination was applied appropriate for evoking fluorescence from the secondary antibody used for PLD1/PLD2 labeling and from the dye-conjugated IB4 or DAPI).

Supplementary Figure 9 Constriction of cortical arterioles evoked by U46619 before each experiment.

Mean constriction of arterioles to U46619 in interleaved control and drug experiments for the: (a) P2X1 blocker NF449, (b) PLA2 blocker MAFP and PLD2 blocker CAY10594 (experiments were on the same animals with interleaved controls in common), (c) fast Ca2+ chelator BAPTA, (d) NMDA receptor blocker D-AP5, (e) NO synthase blocker L-NNA. P-values comparing U46619-evoked constrictions for control and drug experiments were >0.05 for all panels even without correction for multiple comparisons. Data shown as mean±s.e.m.

Supplementary Figure 10 Experimental setup for puffing α,β-methylene-ATP.

(a) DIC image of slice showing a capillary. (b) DIC image of the slice surface showing position of the puff pipette. (c) Fluorescence image showing the pipette containing Alexa Fluor 594 positioned above the slice. (d-i) The spread of the Alexa Fluor 594 dye imaged at the start (d) and 5 consecutive seconds (e-i) following a 5 s puff at 5 psi. Note how the dye flows downwards and does not reach the location of the vessel at the top (demarcated by white dotted lines).

Supplementary information

Supplementary Text and Figures

Supplementary Figures 1–10 and Supplementary Table 1 (PDF 2471 kb)

Supplementary Methods Checklist (PDF 468 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Mishra, A., Reynolds, J., Chen, Y. et al. Astrocytes mediate neurovascular signaling to capillary pericytes but not to arterioles. Nat Neurosci 19, 1619–1627 (2016). https://doi.org/10.1038/nn.4428

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nn.4428

This article is cited by

Search

Quick links

Nature Briefing

Sign up for the Nature Briefing newsletter — what matters in science, free to your inbox daily.

Get the most important science stories of the day, free in your inbox. Sign up for Nature Briefing