Skip to main content

Advertisement

Log in

Distal activity patterns shape the spatial specificity of neurovascular coupling

  • Article
  • Published:

From Nature Neuroscience

View current issue Submit your manuscript

Abstract

Neurovascular coupling links brain activity to local changes in blood flow, forming the basis for non-invasive brain mapping. Using multiscale imaging, we investigated how vascular activity spatially relates to neuronal activity elicited by single whiskers across different columns and layers of mouse cortex. Here we show that mesoscopic hemodynamic signals quantitatively reflect neuronal activity across space but are composed of a highly heterogeneous pattern of responses across individual vessel segments that is poorly predicted by local neuronal activity. Rather, this heterogeneity is dependent on vessel directionality, specifically in thalamocortical input layer 4, where capillaries respond preferentially to neuronal activity patterns along their downstream perfusion domain. Thus, capillaries fine-tune blood flow based on distant activity and encode laminar-specific activity patterns. These findings imply that vascular anatomy sets a resolution limit on functional imaging signals, where individual blood vessels inaccurately report neuronal activity in their immediate vicinity but, instead, integrate activity patterns along the vascular arbor.

This is a preview of subscription content, log in via an institution to check access.

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Fig. 1: Changes in HbT are spatially well correlated to neuronal activity across cortical columns in mouse barrel cortex.
Fig. 2: The selectivity of vessel dilations is highly heterogenous and poorly reflects local excitatory activity in L2/3 and L4.
Fig. 3: Vessel response selectivity does not increase with capillary branch order.
Fig. 4: Vascular perfusion path directionality, but not vessel position per se, shapes vessel dilation selectivity in L4.
Fig. 5: Unlike neurons, vessels show a preference toward activity in the nearest of two diametrically opposite barrels in L4.
Fig. 6: Neuronal activity is more spatially confined than HbT in awake mice.
Fig. 7: Despite a general increase in selectivity, vessel response selectivity in L2/3 remains spatially heterogenous in awake mice.

Similar content being viewed by others

Data availability

The dataset necessary to interpret, verify and extend the results presented in this paper is available at https://doi.org/10.6084/m9.figshare.26121076 (ref. 79). The standardized ROI masks delineating the subregions of S1 and the individual barrels are included in this dataset.

References

  1. Mergenthaler, P., Lindauer, U., Dienel, G. A. & Meisel, A. Sugar for the brain: the role of glucose in physiological and pathological brain function. Trends Neurosci. 36, 587–597 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  2. Howarth, C., Gleeson, P. & Attwell, D. Updated energy budgets for neural computation in the neocortex and cerebellum. J. Cereb. Blood Flow Metab. 32, 1222–1232 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Bourquin, C., Poree, J., Lesage, F. & Provost, J. In vivo pulsatility measurement of cerebral microcirculation in rodents using dynamic ultrasound localization microscopy. IEEE Trans. Med. Imaging 41, 782–792 (2022).

    Article  PubMed  Google Scholar 

  4. Chen, X. et al. Assessment of single-vessel cerebral blood velocity by phase contrast fMRI. PLoS Biol. 19, e3000923 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Huber, L. et al. High-resolution CBV-fMRI allows mapping of laminar activity and connectivity of cortical input and output in human M1. Neuron 96, 1253–1263 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Renaudin, N. et al. Functional ultrasound localization microscopy reveals brain-wide neurovascular activity on a microscopic scale. Nat. Methods 19, 1004–1012 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Akbari, A., Gati, J. S., Zeman, P., Liem, B. & Menon, R. S. Layer dependence of monocular and binocular responses in human ocular dominance columns at 7T using VASO and BOLD. Preprint at bioRxiv https://doi.org/10.1101/2023.04.06.535924 (2023).

  8. Sirotin, Y. B. & Das, A. Anticipatory haemodynamic signals in sensory cortex not predicted by local neuronal activity. Nature 457, 475–479 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. O’Herron, P. et al. Neural correlates of single-vessel haemodynamic responses in vivo. Nature 534, 378–382 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  10. Boido, D. et al. Mesoscopic and microscopic imaging of sensory responses in the same animal. Nat. Commun. 10, 1110 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  11. Winder, A. T., Echagarruga, C., Zhang, Q. & Drew, P. J. Weak correlations between hemodynamic signals and ongoing neural activity during the resting state. Nat. Neurosci. 20, 1761–1769 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Harris, K. D. & Shepherd, G. M. The neocortical circuit: themes and variations. Nat. Neurosci. 18, 170–181 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. 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 

  14. Duvernoy, H. M., Delon, S. & Vannson, J. L. Cortical blood vessels of the human brain. Brain Res. Bull. 7, 519–579 (1981).

    Article  CAS  PubMed  Google Scholar 

  15. Kirst, C. et al. Mapping the fine-scale organization and plasticity of the brain vasculature. Cell 180, 780–795 (2020).

    Article  CAS  PubMed  Google Scholar 

  16. Jukovskaya, N., Tiret, P., Lecoq, J. & Charpak, S. What does local functional hyperemia tell about local neuronal activation? J. Neurosci. 31, 1579–1582 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Rungta, R. L., Chaigneau, E., Osmanski, B. F. & Charpak, S. Vascular compartmentalization of functional hyperemia from the synapse to the pia. Neuron 99, 362–375 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Drew, P. J. Neurovascular coupling: motive unknown. Trends Neurosci. 45, 809–819 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Chen, B. R., Kozberg, M. G., Bouchard, M. B., Shaik, M. A. & Hillman, E. M. A critical role for the vascular endothelium in functional neurovascular coupling in the brain. J. Am. Heart Assoc. 3, e000787 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  20. Emerson, G. G. & Segal, S. S. Endothelial cell pathway for conduction of hyperpolarization and vasodilation along hamster feed artery. Circ. Res. 86, 94–100 (2000).

    Article  CAS  PubMed  Google Scholar 

  21. Iadecola, C., Yang, G., Ebner, T. J. & Chen, G. Local and propagated vascular responses evoked by focal synaptic activity in cerebellar cortex. J. Neurophysiol. 78, 651–659 (1997).

    Article  CAS  PubMed  Google Scholar 

  22. Longden, T. A. et al. Capillary K+-sensing initiates retrograde hyperpolarization to increase local cerebral blood flow. Nat. Neurosci. 20, 717–726 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Schaeffer, S. & Iadecola, C. Revisiting the neurovascular unit. Nat. Neurosci. 24, 1198–1209 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. 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 

  26. Chow, B. W. et al. Caveolae in CNS arterioles mediate neurovascular coupling. Nature 579, 106–110 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Dana, H. et al. Thy1 transgenic mice expressing the red fluorescent calcium indicator jRGECO1a for neuronal population imaging in vivo. PLoS ONE 13, e0205444 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  28. Li, B. et al. Two-photon microscopic imaging of capillary red blood cell flux in mouse brain reveals vulnerability of cerebral white matter to hypoperfusion. J. Cereb. Blood Flow Metab. 40, 501–512 (2020).

    Article  PubMed  Google Scholar 

  29. Staiger, J. F. & Petersen, C. C. H. Neuronal circuits in barrel cortex for whisker sensory perception. Physiol. Rev. 101, 353–415 (2021).

    Article  PubMed  Google Scholar 

  30. Madisen, L. et al. A robust and high-throughput Cre reporting and characterization system for the whole mouse brain. Nat. Neurosci. 13, 133–140 (2010).

    Article  CAS  PubMed  Google Scholar 

  31. Tian, P. et al. Cortical depth-specific microvascular dilation underlies laminar differences in blood oxygenation level-dependent functional MRI signal. Proc. Natl Acad. Sci. USA 107, 15246–15251 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Yu, X., Qian, C., Chen, D. Y., Dodd, S. J. & Koretsky, A. P. Deciphering laminar-specific neural inputs with line-scanning fMRI. Nat. Methods 11, 55–58 (2014).

    Article  PubMed  Google Scholar 

  33. Rungta, R. L. et al. Diversity of neurovascular coupling dynamics along vascular arbors in layer II/III somatosensory cortex. Commun. Biol. 4, 855 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Mishra, A. et al. Astrocytes mediate neurovascular signaling to capillary pericytes but not to arterioles. Nat. Neurosci. 19, 1619–1627 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Kornfield, T. E. & Newman, E. A. Regulation of blood flow in the retinal trilaminar vascular network. J. Neurosci. 34, 11504–11513 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  36. Cai, C. et al. Stimulation-induced increases in cerebral blood flow and local capillary vasoconstriction depend on conducted vascular responses. Proc. Natl Acad. Sci. USA 115, E5796–E5804 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Grubb, S. et al. Precapillary sphincters maintain perfusion in the cerebral cortex. Nat. Commun. 11, 395 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Grant, R. I. et al. Organizational hierarchy and structural diversity of microvascular pericytes in adult mouse cortex. J. Cereb. Blood Flow Metab. 39, 411–425 (2019).

    Article  PubMed  Google Scholar 

  39. Gonzales, A. L. et al. Contractile pericytes determine the direction of blood flow at capillary junctions. Proc. Natl Acad. Sci. USA 117, 27022–27033 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Masamoto, K. & Kanno, I. Anesthesia and the quantitative evaluation of neurovascular coupling. J. Cereb. Blood Flow Metab. 32, 1233–1247 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Zuend, M. et al. Arousal-induced cortical activity triggers lactate release from astrocytes. Nat. Metab. 2, 179–191 (2020).

    Article  CAS  PubMed  Google Scholar 

  42. Echagarruga, C. T., Gheres, K. W., Norwood, J. N. & Drew, P. J. nNOS-expressing interneurons control basal and behaviorally evoked arterial dilation in somatosensory cortex of mice. eLife 9, e60533 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Lee, L. et al. Key aspects of neurovascular control mediated by specific populations of inhibitory cortical interneurons. Cereb. Cortex 30, 2452–2464 (2020).

    Article  CAS  PubMed  Google Scholar 

  44. Vo, T. T. et al. Parvalbumin interneuron activity drives fast inhibition-induced vasoconstriction followed by slow substance P-mediated vasodilation. Proc. Natl Acad. Sci. USA 120, e2220777120 (2023).

    Article  PubMed  PubMed Central  Google Scholar 

  45. Kocharyan, A., Fernandes, P., Tong, X. K., Vaucher, E. & Hamel, E. Specific subtypes of cortical GABA interneurons contribute to the neurovascular coupling response to basal forebrain stimulation. J. Cereb. Blood Flow Metab. 28, 221–231 (2008).

    Article  CAS  PubMed  Google Scholar 

  46. Krawchuk, M. B., Ruff, C. F., Yang, X., Ross, S. E. & Vazquez, A. L. Optogenetic assessment of VIP, PV, SOM and NOS inhibitory neuron activity and cerebral blood flow regulation in mouse somato-sensory cortex. J. Cereb. Blood Flow Metab. 40, 1427–1440 (2020).

    Article  CAS  PubMed  Google Scholar 

  47. Uhlirova, H. et al. Cell type specificity of neurovascular coupling in cerebral cortex. eLife 5, e14315 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  48. Del Franco, A. P., Chiang, P. P. & Newman, E. A. Dilation of cortical capillaries is not related to astrocyte calcium signaling. Glia 70, 508–521 (2022).

    Article  PubMed  Google Scholar 

  49. 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 

  50. Institoris, A. et al. Astrocytes amplify neurovascular coupling to sustained activation of neocortex in awake mice. Nat. Commun. 13, 7872 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Krogsgaard, A. et al. PV interneurons evoke astrocytic Ca2+ responses in awake mice, which contributes to neurovascular coupling. Glia 71, 1830–1846 (2023).

  52. Lia, A., Di Spiezio, A., Speggiorin, M. & Zonta, M. Two decades of astrocytes in neurovascular coupling. Front. Netw. Physiol. 3, 1162757 (2023).

    Article  PubMed  PubMed Central  Google Scholar 

  53. Berwick, J. et al. Fine detail of neurovascular coupling revealed by spatiotemporal analysis of the hemodynamic response to single whisker stimulation in rat barrel cortex. J. Neurophysiol. 99, 787–798 (2008).

    Article  CAS  PubMed  Google Scholar 

  54. Logothetis, N. K., Pauls, J., Augath, M., Trinath, T. & Oeltermann, A. Neurophysiological investigation of the basis of the fMRI signal. Nature 412, 150–157 (2001).

    Article  CAS  PubMed  Google Scholar 

  55. Mathiesen, C., Caesar, K., Akgoren, N. & Lauritzen, M. Modification of activity-dependent increases of cerebral blood flow by excitatory synaptic activity and spikes in rat cerebellar cortex. J. Physiol. 512, 555–566 (1998).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  56. Shih, Y. Y. et al. Ultra high-resolution fMRI and electrophysiology of the rat primary somatosensory cortex. Neuroimage 73, 113–120 (2013).

    Article  PubMed  Google Scholar 

  57. Cho, S. et al. Cortical layer-specific differences in stimulus selectivity revealed with high-field fMRI and single-vessel resolution optical imaging of the primary visual cortex. Neuroimage 251, 118978 (2022).

    Article  CAS  PubMed  Google Scholar 

  58. Jung, W. B., Im, G. H., Jiang, H. & Kim, S. G. Early fMRI responses to somatosensory and optogenetic stimulation reflect neural information flow. Proc. Natl Acad. Sci. USA 118, e2023265118 (2021).

  59. Nunes, D., Gil, R. & Shemesh, N. A rapid-onset diffusion functional MRI signal reflects neuromorphological coupling dynamics. Neuroimage 231, 117862 (2021).

    Article  PubMed  Google Scholar 

  60. Schmid, F., Barrett, M. J. P., Jenny, P. & Weber, B. Vascular density and distribution in neocortex. Neuroimage 197, 792–805 (2019).

    Article  PubMed  Google Scholar 

  61. Shih, A. Y. et al. The smallest stroke: occlusion of one penetrating vessel leads to infarction and a cognitive deficit. Nat. Neurosci. 16, 55–63 (2013).

    Article  CAS  PubMed  Google Scholar 

  62. Nishimura, N., Rosidi, N. L., Iadecola, C. & Schaffer, C. B. Limitations of collateral flow after occlusion of a single cortical penetrating arteriole. J. Cereb. Blood Flow Metab. 30, 1914–1927 (2010).

    Article  PubMed  PubMed Central  Google Scholar 

  63. Koo, B. B. et al. Age-related effects on cortical thickness patterns of the rhesus monkey brain. Neurobiol. Aging 33, 200.e23–200.e31 (2012).

  64. Defelipe, J. The evolution of the brain, the human nature of cortical circuits, and intellectual creativity. Front. Neuroanat. 5, 29 (2011).

    Article  PubMed  PubMed Central  Google Scholar 

  65. Roe, A. W., Winberry, J. E. & Friedman, R. M. Study of single and multidigit activation in monkey somatosensory cortex using voltage-sensitive dye imaging. Neurophotonics 4, 031219 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  66. Shaw, K. et al. Neurovascular coupling and oxygenation are decreased in hippocampus compared to neocortex because of microvascular differences. Nat. Commun. 12, 3190 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  67. Schmid, F., Reichold, J., Weber, B. & Jenny, P. The impact of capillary dilation on the distribution of red blood cells in artificial networks. Am. J. Physiol. Heart Circ. Physiol. 308, H733–H742 (2015).

    Article  CAS  PubMed  Google Scholar 

  68. Epp, R., Schmid, F., Weber, B. & Jenny, P. Predicting vessel diameter changes to up-regulate biphasic blood flow during activation in realistic microvascular networks. Front. Physiol. 11, 566303 (2020).

    Article  PubMed  PubMed Central  Google Scholar 

  69. Hartmann, D. A. et al. Brain capillary pericytes exert a substantial but slow influence on blood flow. Nat. Neurosci. 24, 633–645 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  70. Nelson, A. R. et al. Channelrhodopsin excitation contracts brain pericytes and reduces blood flow in the aging mouse brain in vivo. Front. Aging Neurosci. 12, 108 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  71. McDowell, K. P., Berthiaume, A. A., Tieu, T., Hartmann, D. A. & Shih, A. Y. VasoMetrics: unbiased spatiotemporal analysis of microvascular diameter in multi-photon imaging applications. Quant. Imaging Med. Surg. 11, 969–982 (2021).

    Article  PubMed  PubMed Central  Google Scholar 

  72. Cuttler, A. S. et al. Characterization of Pdgfrb-Cre transgenic mice reveals reduction of ROSA26 reporter activity in remodeling arteries. Genesis 49, 673–680 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  73. Rungta, R. L., Osmanski, B. F., Boido, D., Tanter, M. & Charpak, S. Light controls cerebral blood flow in naive animals. Nat. Commun. 8, 14191 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  74. Valley, M. T. et al. Separation of hemodynamic signals from GCaMP fluorescence measured with wide-field imaging. J. Neurophysiol. 123, 356–366 (2020).

    Article  CAS  PubMed  Google Scholar 

  75. Belanger, S., de Souza, B. O., Casanova, C. & Lesage, F. Correlation of hemodynamic and fluorescence signals under resting state conditions in mice’s barrel field cortex. Neurosci. Lett. 616, 177–181 (2016).

    Article  CAS  PubMed  Google Scholar 

  76. Guevara, E., Sadekova, N., Girouard, H. & Lesage, F. Optical imaging of resting-state functional connectivity in a novel arterial stiffness model. Biomed. Opt. Express 4, 2332–2346 (2013).

    Article  PubMed  PubMed Central  Google Scholar 

  77. Vanni, M. P., Chan, A. W., Balbi, M., Silasi, G. & Murphy, T. H. Mesoscale mapping of mouse cortex reveals frequency-dependent cycling between distinct macroscale functional modules. J. Neurosci. 37, 7513–7533 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  78. Pachitariu, M. et al. Suite2p: beyond 10,000 neurons with standard two-photon microscopy. Preprint at bioRxiv https://doi.org/10.1101/061507 (2017).

  79. Martineau, E. et al. Widefield and two-photon recordings of neuronal and vascular changes during single whisker stimulation in the mouse barrel cortex. figshare https://doi.org/10.6084/m9.figshare.26121076 (2024).

Download references

Acknowledgements

This work was supported by a Natural Sciences and Engineering Research Council of Canada discovery grant (RGPIN-2020-05276), a Canadian Institutes of Health Research project grant (no. 455513) and an Azrieli Future Leader in Canadian Brain Research grant from the Brain Canada Foundation through the Canada Brain Research Fund, with the financial support of Health Canada and the Azrieli Foundation to R.L.R. and an ERA-NET NEURON (JTC2022) grant with financial support from Fonds de Recherche du Québec. R.L.R. holds a Canada Research Chair in Neurovascular Interactions. A.M. was supported by a Mitacs Accelerate Studentship (IT28768) and a Bourse de Mérite from the Faculty of Medicine. We thank V. Linder (Maine Medical Center) for PDGFRβ-Cre mice; I. Laplante, P. Kwemo and L. Zana for colony and laboratory management; M. Abran and S. Bélanger for technical assistance; and P. Rompré for statistical advice. Schematics in Figs. 1a, 2a, 5a and 6a were created with BioRender.

Author information

Authors and Affiliations

Authors

Contributions

É.M. and R.L.R. designed the study. É.M., N.E. and R.L.R. performed experiments. É.M. and A.M. developed analysis procedures, analyzed data and interpreted results, with R.L.R. É.M. and R.L.R. wrote the paper, with feedback from A.M. R.L.R. supervised the research. All authors agreed on the final version of the paper.

Corresponding author

Correspondence to Ravi L. Rungta.

Ethics declarations

Competing interests

The authors declare no competing interests.

Peer review

Peer review information

Nature Neuroscience thanks Tzu-Hao Chao, Anna Devor, Anusha Mishra and the other, anonymous, reviewer(s) for their contribution to the peer review of this work.

Additional information

Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Extended data

Extended Data Fig. 1 Examples of vessel dilations poorly reflecting the selectivity of neighboring excitatory neurons.

(a) Example image of average jRGECO1a fluorescence in response to the stimulation of the associated whisker (green) superimposed onto an image of the vasculature (magenta), measured in layer 2/3 from a thy1-jRGECO1a-expressing mouse. Vessels are labelled by retro-orbitally injecting Alexa-680 Dextran (2000 kDa). (b) Diameter changes of each vessel segment and (c) average changes in jRGECO1a fluorescence (ΔF/F0) from neurons surrounding these vessels following the stimulation of the associated whisker (blue) or its neighbor (orange). (d) Example images of average GCaMP6f fluorescence (green) in response to the stimulation of the associated whisker (left) or its neighbor (right) superimposed onto an image of the vasculature (red), measured in layer 4 from a Scnn1a-Tg3-Cre::GCaMP6f mouse. Vessels are labelled by retro-orbitally injecting TexasRed Dextran (70 kDa). (e) Diameter changes of each vessel segment and (f) average change in GCaMP6f fluorescence from neurons surrounding these vessels (neuron #1–5) following the stimulation of the associated whisker (blue) or its neighbor (orange). Scale bars = 20 µm (a) and 10 µm (d). Signals were extracted by manually drawing ROIs over the neurons and without neuropil subtraction.

Extended Data Fig. 2 A greater proportion of neurons in the L4 barrel are selective for their associated whisker than in L2/3.

(a-b) False-color images of GCaMP6f fluorescence during the stimulation of the C2 (a) or D2 (b) whisker measured in the C2 barrel in layer 4 of a Scnn1a-Tg3-Cre::GCaMP6f mouse (average of 9 and 11 trials respectively). Dashed lines represent the boundary of the C2 L4 barrel. Fluorescence changes on the right side of the barrel are obscured by surface vessels. (c) Image illustrating the selectivity of neuronal cell bodies within the imaged barrel, segmented with the assistance of suite2P. (d) Average change in GCaMP6f fluorescence (ΔF/F0), after neuropil subtraction, from example neuronal somas following the stimulation of the associated whisker (C2, blue) or its neighbor (D2, orange). A 200 ms moving average was applied to the traces for representation. SI are calculated on unfiltered signals. (e) Global distribution of neuronal selectivity indexes in the barrel ROI in layer 2/3 (left) and layer 4 (right) showing that roughly twice as many neuronal somas were selective for their associated whisker in L4 compared with L2/3 in sedated mice (L2/3: N = 6 mice (4M, 2F), n = 6 recordings, 113 excited neurons out of 251 segmented; L4: N = 10 mice (5M, 5F), n = 12 recordings, 299 excited neurons out of 477 segmented). (f) Percentage of selective neuronal somas (SI > 0.1) in L2/3 and L4, calculated per recording (p = 0.0415; L2/3: n = 6 vs L4: n = 12; Two-tailed Mann-Whitney test). (g) Selectivity of the neuropil signal, extracted by removing the neuronal ROIs from the barrel ROI, in layer 2/3 and layer 4 (p = 0.1675; L2/3: n = 6 vs L4: n = 12; Two-tailed Student’s T-test). Scale bars = 50 µm (a-b). (f-g) *: p < 0.05, ns: non-significant. Error bars (f-g) represent the SEM.

Extended Data Fig. 3 Arteriole dilation onset tends to be faster in L4 than in L2/3.

(a) Normalized arteriole diameter changes in L2/3 and L4 following a 5 Hz 4 second stimulation of the whisker associated with its barrel (L2/3: N = 10 mice (5M, 5F); n = 11 responsive arterioles; L4: N = 9 mice (4M, 5F), n = 14 responsive arterioles). Two responsive arterioles in L2/3 and 1 responsive arteriole in L4 were excluded from this analysis as the fit was too uncertain to accurately calculate an onset. (b-c) Onsets were calculated by fitting a sigmoid to the rise of each response and calculating the time to 10% (b, p = 0.0745, Two-tailed Student’s t-test) and 25% (c, p = 0.106, Two-tailed Student’s t-test) of the peak, as previously described33. Shaded areas (a) and error bars (b-c) represent the SEM. All mice were under dexmedetomidine sedation.

Extended Data Fig. 4 Vessel dilation selectivity across capillary branches in L2/3.

(a) Raster plots of diameter changes of responding vessels following the stimulation of the associated (left) or neighboring (right) whisker in L2/3, split by branch order and sorted by ascending selectivity index. Dashed lines represent stimulation onset and offset. (b) Average vessel diameter changes of different branch orders following the stimulation of the associated (blue) or neighboring (orange) whisker. (c) Vessel dilation selectivity is independent of branch order in L2/3. (d) Correlation between vessel dilation selectivity index and cortical depth. (a-d; N = 13 mice (8M, 5F), arterioles: n = 13/15, 1st order: n = 12/20, 2nd / 3rd order: n = 48/61, 4th / 5th order: n = 29/49, 6th - 8th order: n = 7/11). Shaded areas (b) and error bars (c) represent the SEM. All mice were under dexmedetomidine sedation.

Extended Data Fig. 5 Vessel dilation selectivity across capillary branches in L4.

(a) Raster plots of diameter changes of responding vessels following the stimulation of the associated (left) or neighboring (right) whisker from Scnn1a-Tg3-Cre::GCaMP6f (L4) mice, split by branch order and sorted by ascending selectivity index. Dashed lines represent stimulation onset and offset. (b) Average vessel diameter changes of different branch orders following the stimulation of the associated (blue) or neighboring (orange) whisker. (c) Vessel dilation selectivity is independent of branch order in L4. (d) Correlation between vessel dilation selectivity index and cortical depth. (a-d; N = 10 mice (5M, 5F), arterioles: n = 15/15, 1st order: n = 11/14, 2nd / 3rd order: n = 37/53, 4th / 5th order: n = 39/70, 6th - 8th order: n = 16/36). Shaded areas (b) and error bars (c) represent the SEM. All mice were under dexmedetomidine sedation.

Extended Data Fig. 6 RBC velocity and flow selectivity across branch order and cortical depth.

(a) Example image of average jRGECO1a fluorescence in response to the stimulation of the associated whisker (green) superimposed onto an image of the vasculature (magenta), measured in L2/3. (b) Example image of a linescan (distance/time) within a capillary (left) and average increase in RBC velocity and flow in the capillary (right) following the stimulation of the associated (blue) or neighboring (orange) whisker. (c-d) Average change in RBC velocity (c) and flow (d), across different branch orders (N = 16 mice, (10M, 6F)), following the stimulation of the associated (blue) or neighboring (orange) whisker. (e-f) Correlation between cortical depth and selectivity indexes for increases in RBC velocity (e) or RBC flow (f). Of note, cortical depth had little impact on the selectivity of RBC dynamics, with only increases in RBC velocity being very slightly more selective with depth in higher order capillaries (RBC Velocity: R2adj = 0.08875, Overall regression: p = 0.0204, Depth: p = 0.2007, BranchOrder: p = 0.0154, Depth*BranchOrder: p = 0.0389; RBC Flow: Adjusted R2adj = -0.01495, Overall regression: p = 0.4905, Depth: p = 0.1407, BranchOrder: p = 0.1621, Depth*BranchOrder: p = 0.1463;MLR). Shaded areas (c-d) represent the SEM. Results are pooled from thy1-jRGECO1a-expressing (L2/3) and Scnn1a-Tg3-Cre::GCaMP6f (L4) mice. The numbers of vessels and mice in each group is identical to those in Fig. 3g, h. All mice were under dexmedetomidine sedation.

Extended Data Fig. 7 2D-vessel position relative to the associated and neighboring whisker representation centroid.

(a) Schematic representation of the formula used to calculate the normalized axial position on the C2-D2 axis and the relative proximity of each vessel to the barrel column’s centroid. (b-c) Normalized axial position (b) and relative proximity index (c) values for five hypothetical vessel positions on the cortical surface.

Extended Data Fig. 8 Vessels that were poorly tuned to their associated whisker have a preference towards activity in one of two diametrically opposite barrels in L4.

(a) Example image highlighting two neighboring capillary networks emerging from the same penetrating arteriole, but perfusing spatial domains in different directions. These networks were recorded over two imaging sessions while stimulating a different whisker pair. (b-c) Dilation measurements from each network showing that (b) vessels along network 1 become gradually less selective towards the associated whisker (C2), and more selective to D2, while (c) vessels along network 2 become more selective to C2, and less responsive to either neighbor of C2. (d) Changes in vessel selectivity depending on the stimulated whisker pair (N = 6 mice (3M, 3F), arterioles: n = 7, 1st order: n = 7, 2nd/3rd order: n = 25, 4th/5th order: n = 32, 6th-8th order: n = 12). (e-f) Results in (d) split between vessels that were non-selective (e, N = 6 mice, arterioles: n = 5, 1st order: n = 5, 2nd/3rd order: n = 14, 4th/5th order: n = 22, 6th-8th order: n = 6) or selective (f, N = 6 mice, arterioles: n = 2, 1st order: n = 2, 2nd/3rd order: n = 11, 4th/5th order: n = 10, 6th-8th order: n = 6) toward the associated whisker, showing that vessels with poor selectivity towards the associated whisker were more selective towards one of the two neighbors and vice versa (e-f, StimulatedWhiskerPair: p = 0.0002 and p = 0.0087 respectively, RM-Two-way ANOVA). Non-responsive vessels when stimulating either neighboring whisker were assigned a value of 0. The shaded area in (d) represents the cut-off value (0.2268) for splitting selective and non-selective vessels, corresponding to the mean selectivity index when stimulating the associated-neighbor1 pair (in absolute value). Absolute values were used for this analysis in order to quantify if selectivity improved or deteriorated in each subgroup when stimulating both neighbors. Shaded areas (b-c) represent the stimulation. (e-f) * p < 0.05, Sidak’s multiple comparisons. Refer to Supplementary Tables 38 and 39 for exact p-values. Scale bars = 50 µm.

Extended Data Fig. 9 In awake mice, both neuronal activity and [HbT] elevations are more spatially specific in the late-phase of the response.

(a) jRGECO1a responses during the early and late phase (red shaded areas, AUCt = 4:5 and AUCt = 7:8, respectively) were extracted and compared to (b) changes in [HbT] during the matching phase in awake mice. An offset of +0.479 seconds, was applied to each phase for the [HbT] signal to account for the delay in NVC. This offset was determined by calculating a transfer function between the [HbT] and jRGECO1a signals33,79. (c) Spatial tuning of the neuronal and [HbT] response in awake mice for each phase, showing both neuronal and [HbT] responses are more spatially specific in the late phase of the response (N = 6 mice (3M, 3F); Associated barrel: n = 19 barrels, 1-away: n = 26 barrels, 2-away: n = 14 barrels). (d) The AUC of vessel dilations from two-photon experiments in L2/3 of awake mice during the early and late phase (AUCt = 4.479: 5.479 and AUCt = 7.479: 8.479, respectively) were extracted and compared. (e-g) Dilation selectivity during the early (e) and late phase (f) of the response in awake mice, grouped by branch order (g, N = 5 mice (2M, 3F), arterioles: n = 11/12, 1st order: n = 12/14, 2nd / 3rd order: n = 43/57, 4th / 5th order: n = 25/50, 6th order: n = 1/3 for each phase, RM-Two-way ANOVA with Tukey’s multiple comparisons). (h-i) Changes in dilation selectivity in the early (h) and late (i) phase, as a function of vascular path directionality. Traces in (a, b and d) are identical to those presented in Fig. 6c, d, and Fig. 7c, and serve only to represent the phases used for this analysis. Shaded traces (a,b and d) and error bars (c, e and f) represent the SEM. The single dilating 6th order vessel (e-g) was not included in the analysis. (g) * p < 0.05, ** p < 0.01, **** p < 0.0001 Tukey’s multiple comparisons. Refer to Supplementary Table 41 for exact p-values.

Extended Data Fig. 10 Spatial diversity of L2/3 neuronal responses is maintained in awake mice.

(a, b) Images illustrating the selectivity of neuronal cell bodies in L2/3 within the imaged barrel, segmented with the assistance of suite2P, in sedated (a) and awake (b) thy1-jRGECO1a-expressing mice. (c) Global distribution of neuronal selectivity indexes in the barrel ROI in layer 2/3 in awake mice (L2/3: N = 5 mice (2M, 3F), n = 9 recordings, 293 excited neurons out of 588 segmented). Of note, only recordings from the barrel in the center of the image were included, as the majority of this barrel was visible for segmentation (9 out 12 total recordings). (d) Percentage of selective neuronal somas (SI > 0.1) in awake and sedated mice, calculated per recording, showing that a similar proportion of cells in the L2/3 are selective for their associated whisker (p = 0.8639; Sedated: n = 6 vs Awake: n = 9; Two-tailed Mann-Whitney test). (e) Selectivity of the neuropil signal, extracted by subtracting the neuronal ROIs from the barrel ROI, in awake and sedated mice (p = 0.0169; Sedated: n = 6 vs Awake: n = 9; Two-tailed Student’s T-test). Error bars (d-e) represent the SEM. Scale bars = 50 µm (a-b).

Supplementary information

Supplementary Information

Legends for Supplementary Videos 1–3 and Tables 1–44.

Reporting Summary

Supplementary Video 1

Neuronal activity in L2/3 during C2 or D2 whisker stimulation in sedated mice. Imaging of jRGECO1a expressed in L2/3 excitatory neurons during the stimulation of the C2 (top left, bottom: blue) or D2 (top right, bottom: yellow) whiskers. Whiskers are stimulated from 0 s to 4 s. Images represent the average of seven C2 stimulations and 13 D2 stimulations. Between-frame movement was corrected by applying a rigid alignment algorithm (Methods) on simultaneously acquired images of the vasculature (Alexa 680–dextran) and then applying the same translation vectors to the images from the neuronal channel. Same recording as the example in Fig. 2b. The ROI used to extract signal from the C2 barrel is traced in white.

Supplementary Video 2

Neuronal activity in L2/3 during C2 or D2 whisker stimulation in sedated mice. Imaging of GCaMP6f expressed in L4 neurons during the stimulation of the C2 (top left, bottom: green) or D2 (top right, bottom: magenta) whiskers. Whiskers are stimulated from 0 s to 4 s. Images represent the average of 11 C2 stimulations and nine D2 stimulations. Between-frame movement was corrected by applying a rigid alignment algorithm (Methods) on simultaneously acquired images of the vasculature (Texas Red–dextran) and then applying the same translation vectors to the images from the neuronal channel. Same recording as the example in Fig. 2g. The ROIs used to extract signal from the C2 and D2 barrels are traced in white.

Supplementary Video 3

Neuronal activity in L2/3 during C2 or D2 whisker stimulation in awake mice. Imaging of jRGECO1a expressed in L2/3 excitatory neurons during the stimulation of the C2 (top left, bottom: blue) or D2 (top right, bottom: yellow) whiskers. Whiskers are stimulated from 0 s to 4 s. Images represent the average of 10 C2 stimulations and 10 D2 stimulations. Between-frame movement was corrected by applying a rigid alignment algorithm (Methods) on simultaneously acquired images of the vasculature (Alexa 680–dextran) and then applying the same translation vectors to the images from the neuronal channel. Same recording as the example in Fig. 7a. The ROIs used to extract signal from the C2 and D2 barrels are traced in white.

Rights and permissions

Springer Nature or its licensor (e.g. a society or other partner) holds exclusive rights to this article under a publishing agreement with the author(s) or other rightsholder(s); author self-archiving of the accepted manuscript version of this article is solely governed by the terms of such publishing agreement and applicable law.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Martineau, É., Malescot, A., Elmkinssi, N. et al. Distal activity patterns shape the spatial specificity of neurovascular coupling. Nat Neurosci (2024). https://doi.org/10.1038/s41593-024-01756-7

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1038/s41593-024-01756-7

  • Springer Nature America, Inc.

Navigation