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:

Pericyte degeneration leads to neurovascular uncoupling and limits oxygen supply to brain

Abstract

Pericytes are perivascular mural cells of brain capillaries. They are positioned centrally in the neurovascular unit between endothelial cells, astrocytes and neurons. This position allows them to regulate key neurovascular functions of the brain. The role of pericytes in the regulation of cerebral blood flow (CBF) and neurovascular coupling remains, however, under debate. Using loss-of-function pericyte-deficient mice, here we show that pericyte degeneration diminishes global and individual capillary CBF responses to neuronal stimuli, resulting in neurovascular uncoupling, reduced oxygen supply to the brain and metabolic stress. Neurovascular deficits lead over time to impaired neuronal excitability and neurodegenerative changes. Thus, pericyte degeneration as seen in neurological disorders such as Alzheimer's disease may contribute to neurovascular dysfunction and neurodegeneration associated with human disease.

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

Access options

Rent or buy this article

Prices vary by article type

from$1.95

to$39.95

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

Figure 1: Delayed capillary dilation and reduced capillary red blood cell flow in response to a hindlimb stimulus in 1- to 2-month-old pericyte-deficient Pdgfrb+/− mice.
Figure 2: Impaired hemodynamic response in 1- to 2-month-old pericyte-deficient Pdgfrb+/− mice determined by IOS imaging (530 nm) at a time point when neuronal activity is unaffected as determined by LFP recordings.
Figure 3: Arteriolar vasoactivity and endothelium-dependent vasodilation in 1- to 2-month-old pericyte-deficient Pdgfrb+/− mice and age-matched littermate controls.
Figure 4: Diminished brain tissue oxygen levels and oxygen delivery in young pericyte-deficient Pdgfrb+/− mice.
Figure 5: Metabolic changes in 1- to 2-month-old pericyte-deficient Pdgfrb+/− mice.
Figure 6: Cortical neuronal activity, neuronal number, neuritic density and behavior in pericyte-deficient Pdgfrb+/− mice 1–2 and 6–8 months old, along with age-matched littermate controls.

Similar content being viewed by others

References

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  2. Zlokovic, B.V. Neurovascular pathways to neurodegeneration in Alzheimer's disease and other disorders. Nat. Rev. Neurosci. 12, 723–738 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Iadecola, C. The pathobiology of vascular dementia. Neuron 80, 844–866 (2013).

    Article  CAS  PubMed  Google Scholar 

  4. Iadecola, C. & Nedergaard, M. Glial regulation of the cerebral microvasculature. Nat. Neurosci. 10, 1369–1376 (2007).

    Article  CAS  PubMed  Google Scholar 

  5. MacVicar, B.A. & Newman, E.A. Astrocyte regulation of blood flow in the brain. Cold Spring Harb. Perspect. Biol. 7, a020388 (2015).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  6. Rouget, C. Memoire sur le developpement, la structures et les proprietes des capillaries sanguins et lymphatiques. Arch. Physiol. Norm. Pathol. 5, 603–633 (1873).

    Google Scholar 

  7. Armulik, A., Genové, G. & Betsholtz, C. Pericytes: developmental, physiological, and pathological perspectives, problems, and promises. Dev. Cell 21, 193–215 (2011).

    Article  CAS  PubMed  Google Scholar 

  8. Winkler, E.A., Bell, R.D. & Zlokovic, B.V. Central nervous system pericytes in health and disease. Nat. Neurosci. 14, 1398–1405 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Sweeney, M.D., Ayyadurai, S. & Zlokovic, B.V. Pericytes of the neurovascular unit: key functions and signaling pathways. Nat. Neurosci. 19, 771–783 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Armulik, A. et al. Pericytes regulate the blood-brain barrier. Nature 468, 557–561 (2010).

    Article  CAS  PubMed  Google Scholar 

  11. Daneman, R., Zhou, L., Kebede, A.A. & Barres, B.A. Pericytes are required for blood-brain barrier integrity during embryogenesis. Nature 468, 562–566 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Bell, R.D. et al. Pericytes control key neurovascular functions and neuronal phenotype in the adult brain and during brain aging. Neuron 68, 409–427 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. 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 

  14. 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 

  15. 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 

  16. Fernández-Klett, F., Offenhauser, N., Dirnagl, U., Priller, J. & Lindauer, U. Pericytes in capillaries are contractile in vivo, but arterioles mediate functional hyperemia in the mouse brain. Proc. Natl. Acad. Sci. USA 107, 22290–22295 (2010).

    Article  PubMed  PubMed Central  Google Scholar 

  17. Hill, R.A. et al. Regional blood flow in the normal and ischemic brain is controlled by arteriolar smooth muscle cell contractility and not by capillary pericytes. Neuron 87, 95–110 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Iadecola, C. Neurovascular regulation in the normal brain and in Alzheimer's disease. Nat. Rev. Neurosci. 5, 347–360 (2004).

    Article  CAS  PubMed  Google Scholar 

  19. Montagne, A. et al. Blood-brain barrier breakdown in the aging human hippocampus. Neuron 85, 296–302 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Farkas, E. & Luiten, P.G. Cerebral microvascular pathology in aging and Alzheimer's disease. Prog. Neurobiol. 64, 575–611 (2001).

    Article  CAS  PubMed  Google Scholar 

  21. Baloyannis, S.J. & Baloyannis, I.S. The vascular factor in Alzheimer's disease: a study in Golgi technique and electron microscopy. J. Neurol. Sci. 322, 117–121 (2012).

    Article  CAS  PubMed  Google Scholar 

  22. Sengillo, J.D. et al. Deficiency in mural vascular cells coincides with blood-brain barrier disruption in Alzheimer's disease. Brain Pathol. 23, 303–310 (2013).

    Article  PubMed  Google Scholar 

  23. Halliday, M.R. et al. Accelerated pericyte degeneration and blood-brain barrier breakdown in apolipoprotein E4 carriers with Alzheimer's disease. J. Cereb. Blood Flow Metab. 36, 216–227 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Winkler, E.A. et al. Blood-spinal cord barrier breakdown and pericyte reductions in amyotrophic lateral sclerosis. Acta Neuropathol. 125, 111–120 (2013).

    Article  CAS  PubMed  Google Scholar 

  25. 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 

  26. Montagne, A. et al. Brain imaging of neurovascular dysfunction in Alzheimer's disease. Acta Neuropathol. 131, 687–707 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. 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 

  28. Chow, N. et al. Serum response factor and myocardin mediate arterial hypercontractility and cerebral blood flow dysregulation in Alzheimer's phenotype. Proc. Natl. Acad. Sci. USA 104, 823–828 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Kim, T.N. et al. Line-scanning particle image velocimetry: an optical approach for quantifying a wide range of blood flow speeds in live animals. PLoS One 7, e38590 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Hillman, E.M.C. Optical brain imaging in vivo: techniques and applications from animal to man. J. Biomed. Opt. 12, 051402 (2007).

    Article  PubMed  Google Scholar 

  31. Sirotin, Y.B., Hillman, E.M.C., Bordier, C. & Das, A. Spatiotemporal precision and hemodynamic mechanism of optical point spreads in alert primates. Proc. Natl. Acad. Sci. USA 106, 18390–18395 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Iadecola, C. et al. SOD1 rescues cerebral endothelial dysfunction in mice overexpressing amyloid precursor protein. Nat. Neurosci. 2, 157–161 (1999).

    Article  CAS  PubMed  Google Scholar 

  33. Wu, Z. et al. Role of the MEOX2 homeobox gene in neurovascular dysfunction in Alzheimer disease. Nat. Med. 11, 959–965 (2005).

    Article  CAS  PubMed  Google Scholar 

  34. Sakadzić, S. et al. Two-photon high-resolution measurement of partial pressure of oxygen in cerebral vasculature and tissue. Nat. Methods 7, 755–759 (2010).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  35. Kasischke, K.A. et al. Two-photon NADH imaging exposes boundaries of oxygen diffusion in cortical vascular supply regions. J. Cereb. Blood Flow Metab. 31, 68–81 (2011).

    Article  CAS  PubMed  Google Scholar 

  36. Baraghis, E. et al. Two-photon microscopy of cortical NADH fluorescence intensity changes: correcting contamination from the hemodynamic response. J. Biomed. Opt. 16, 106003 (2011).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  37. Dromparis, P. & Michelakis, E.D. Mitochondria in vascular health and disease. Annu. Rev. Physiol. 75, 95–126 (2013).

    Article  CAS  PubMed  Google Scholar 

  38. Barros, L.F. Metabolic signaling by lactate in the brain. Trends Neurosci. 36, 396–404 (2013).

    Article  CAS  PubMed  Google Scholar 

  39. Ishii, Y. et al. Characterization of neuroprogenitor cells expressing the PDGF beta-receptor within the subventricular zone of postnatal mice. Mol. Cell. Neurosci. 37, 507–518 (2008).

    Article  CAS  PubMed  Google Scholar 

  40. Lindahl, P., Johansson, B.R., Levéen, P. & Betsholtz, C. Pericyte loss and microaneurysm formation in PDGF-B-deficient mice. Science 277, 242–245 (1997).

    Article  CAS  PubMed  Google Scholar 

  41. Winkler, E.A., Bell, R.D. & Zlokovic, B.V. Pericyte-specific expression of PDGF beta receptor in mouse models with normal and deficient PDGF beta receptor signaling. Mol. Neurodegener. 5, 32 (2010).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  42. Hartmann, D.A. et al. Pericyte structure and distribution in the cerebral cortex revealed by high-resolution imaging of transgenic mice. Neurophotonics 2, 041402 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  43. Attwell, D., Mishra, A., Hall, C.N., O'Farrell, F.M. & Dalkara, T. What is a pericyte? J. Cereb. Blood Flow Metab. 36, 451–455 (2016).

    Article  CAS  PubMed  Google Scholar 

  44. Winkler, E.A., Sagare, A.P. & Zlokovic, B.V. The pericyte: a forgotten cell type with important implications for Alzheimer's disease? Brain Pathol. 24, 371–386 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Kawamura, H. et al. Effects of angiotensin II on the pericyte-containing microvasculature of the rat retina. J. Physiol. (Lond.) 561, 671–683 (2004).

    Article  CAS  Google Scholar 

  46. Yamanishi, S., Katsumura, K., Kobayashi, T. & Puro, D.G. Extracellular lactate as a dynamic vasoactive signal in the rat retinal microvasculature. Am. J. Physiol. Heart Circ. Physiol. 290, H925–H934 (2006).

    Article  CAS  PubMed  Google Scholar 

  47. Oishi, K., Kamiyashiki, T. & Ito, Y. Isometric contraction of microvascular pericytes from mouse brain parenchyma. Microvasc. Res. 73, 20–28 (2007).

    Article  CAS  PubMed  Google Scholar 

  48. Dai, M., Nuttall, A., Yang, Y. & Shi, X. Visualization and contractile activity of cochlear pericytes in the capillaries of the spiral ligament. Hear. Res. 254, 100–107 (2009).

    Article  PubMed  PubMed Central  Google Scholar 

  49. Park, L. et al. Age-dependent neurovascular dysfunction and damage in a mouse model of cerebral amyloid angiopathy. Stroke 45, 1815–1821 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  51. Ragan, T. et al. Serial two-photon tomography for automated ex vivo mouse brain imaging. Nat. Methods 9, 255–258 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Tsai, P.S. et al. Correlations of neuronal and microvascular densities in murine cortex revealed by direct counting and colocalization of nuclei and vessels. J. Neurosci. 29, 14553–14570 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Brown, C.E., Aminoltejari, K., Erb, H., Winship, I.R. & Murphy, T.H. In vivo voltage-sensitive dye imaging in adult mice reveals that somatosensory maps lost to stroke are replaced over weeks by new structural and functional circuits with prolonged modes of activation within both the peri-infarct zone and distant sites. J. Neurosci. 29, 1719–1734 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Bell, R.D. et al. Apolipoprotein E controls cerebrovascular integrity via cyclophilin A. Nature 485, 512–516 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. Harrison, T.C., Sigler, A. & Murphy, T.H. Simple and cost-effective hardware and software for functional brain mapping using intrinsic optical signal imaging. J. Neurosci. Methods 182, 211–218 (2009).

    Article  PubMed  Google Scholar 

  56. Frostig, R.D., Lieke, E.E., Ts'o, D.Y. & Grinvald, A. Cortical functional architecture and local coupling between neuronal activity and the microcirculation revealed by in vivo high-resolution optical imaging of intrinsic signals. Proc. Natl. Acad. Sci. USA 87, 6082–6086 (1990).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. 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 

  58. Sagare, A.P. et al. Pericyte loss influences Alzheimer-like neurodegeneration in mice. Nat. Commun. 4, 2932 (2013).

    Article  PubMed  CAS  Google Scholar 

  59. Hesterberg, T. Bootstrap. Wiley Interdiscip. Rev. Comput. Stat. 3, 497–526 (2011).

    Article  Google Scholar 

  60. Efron, B. & Tibshirani, R. An Introduction to the Bootstrap (Chapman & Hall, 1993).

Download references

Acknowledgements

We would like to thank R. Angus for statistical discussions. This research was supported by National Institutes of Health grants R01 AG023084, R01 AG039452, R01 NS034467 and R01 NS100459 to B.V.Z.; R01 NS091230 to S.S.; and R01 EB000790, R24 NS092986 and P01 NS055104 to D.A.B.; National Natural Science Foundation of China grant 31371116 to Y.Z.; and American Heart Association grant SDG7600037 to S.S.

Author information

Authors and Affiliations

Authors

Contributions

K.K., A.R.N. and A.R. contributed to manuscript preparation and to experimental design and analysis, and conducted experiments. S.V.R. and S.S. contributed to experimental design and to data analysis and interpretation, and conducted experiments. A.A., D.L. and Y.W. conducted and analyzed experiments. P.S.T. contributed to three-dimensional angiography analysis and software. Z.Z. contributed to experimental design and analysis. Y.Z. contributed to data analysis. D.A.B. contributed to project design. B.V.Z. supervised and designed all experiments and analysis, and wrote the manuscript.

Corresponding author

Correspondence to Berislav V Zlokovic.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Integrated supplementary information

Supplementary Figure 1 The average sigmoid parametric curves of capillary dilation time courses and average time to 50% peak capillary diameter dilation computed from individual capillary responses averaged per animal in control Pdgfrb+/+ mice (top, red traces, n = 12 mice, 37 individual capillaries) and pericyte-deficient Pdgfrb+/− mice (bottom, blue traces, n = 9 mice, 33 individual capillaries).

The time to 50% peak dilation corresponds to the time at which the sigmoid curve reaches 50% of its maximal value, as illustrated by the intercepts of the horizontal dashed line at 0.5 on the ordinate with the curves (diameter increase) and the vertical dashed line intercepts with the abscissa (time to 50% peak dilation). Arrows below the abscissa illustrate time intercepts showing 50% peak capillary diameter dilation per mouse in each group. As in Fig. 1c, diameter changes are expressed relative to the respective basal capillary diameter prior to stimulation (value set as 0) and maximal diameter after stimulation (value set as 1). The average sigmoid curves were derived from individual sigmoid curves fitted to individual capillary responses per animal. The sigmoid fits for individual capillary responses with their intercepts with the horizontal dashed line at 0.5 on the ordinate (diameter increase) and the vertical dashed line intercepts (time to 50% peak dilation) with the abscissa are not shown because of the high degree of overlap, which makes difficult to distinguish responses between individual capillaries. However, data (circles) in Fig. 1d were derived from averaging the individual capillary responses per mouse (time to 50% peak dilation) from sigmoid fits of individual vessel dilation time courses per animal, and then the individual times for 50% peak dilation values were averaged per animal.

Supplementary Figure 2 Pericyte capillary coverage in 1- to 2-month-old Pdgfrb+/− mice and age-matched littermate controls.

(a) Representative confocal microscopy images of pericyte coverage (CD13, magenta) of lectin-positive brain endothelial capillary profiles (< 6 μm in diameter; lectin, blue) in the stimulated cortical S1 area in Pdgfrb+/+ and Pdgfrb+/− mice. (b) Quantification of CD13-positive pericyte coverage of cortical capillaries in Pdgfrb+/+ and Pdgfrb+/− mice from a. Mean ± 95% CI; n = 5 Pdgfrb+/+ and 6 Pdgfrb+/− mice per group (t-test, single tail, equal variance: t = 11.36, p = 6.1e-7). In each animal 5 randomly selected fields from the cortex were analyzed in 6 non-adjacent sections (~100 μm apart) and averaged per mouse to obtain individual values (circles) as illustrated. (c) Vascular smooth muscle cell actin (SMa; yellow) and pericyte marker CD13 in Pdgfrb+/+ control mice illustrating that pericytes lining lectin-positive endothelial capillary profiles are largely negative for SMa. An SMα-positive arteriole (arrow) is shown for comparison.

Source data

Supplementary Figure 3 Baseline vessel diameters, RBC flow velocity and thickness of the arteriole smooth muscle cell layer in 1- to 2-month-old pericyte-deficient Pdgfrb+/− mice and age-matched littermate controls.

(a) Average baseline in vivo vessel diameters (mean ± 95% CI) prior to stimulation determined for 21 total arterioles from 9 Pdgfrb+/+ mice, 27 total arterioles from 11 Pdgfrb+/− mice, 57 total capillaries from 10 Pdgfrb+/+ mice and 40 total capillaries from 10 Pdgfrb+/− mice (arterioles: t-test, equal variance: t = 0.27, p = 0.79; capillaries: t-test, equal variance: t = 1.04, p = 0.31). (b) Average (mean ± 95% CI) baseline in vivo RBC velocity acquired prior to stimulation in 14 total arterioles from 6 Pdgfrb+/+ mice and 23 total arterioles from 10 Pdgfrb+/− mice, and 32 total capillaries from 9 Pdgfrb+/+ mice and 36 total capillaries from 12 Pdgfrb+/− mice (t-test, equal variance: arterioles: t = 0.23 p = 0.82, capillaries: t = 0.33, p = 0.74). (c) Representative images of the smooth muscle cell layer (SMα, red) thickness and lack of pericyte coverage (CD13, magenta) on arterioles. (d) Thickness of the arteriolar smooth muscle cell layer determined by confocal microscopy analysis. Mean ± 95% CI from 42 arterioles from 7 Pdgfrb+/+ mice and 45 arterioles from 6 Pdgfrb+/− mice (t-test, equal variance: t = 1.14, p = 0.28).Two-tail t-tests used.

Source data

Supplementary Figure 4 Astrocyte coverage of capillary wall, astrocyte and microglia numbers and capillary length in 1- to 2-month-old pericyte-deficient Pdgfrb+/− mice and age-matched littermate controls.

(a) Confocal microscopy of immunolabeled aquaporin-4-positive astrocytic endfoot coverage of microvessels (lectin), GFAP-positive astrocytes, and Iba1-positive microglia in the S1 cortical region. (b-d) Quantification (mean ± 95% CI) of aquaporin-4-positive (AQP4+) astrocytic endfoot coverage (t-test, equal variance: t = 0.24, p = 0.82) (b), the number of GFAP-positive astrocytes (t-test, equal variance: t = 0.93, p = 0.38) (c), and Iba1-positive microglia (t-test, equal variance: t = 0.26, p = 0.80) (d) from 5 Pdgfrb+/+ and 5 Pdgfrb+/− mice at 1-2 months of age; in each animal 6 randomly selected fields from the cortex were analyzed in 5 nonadjacent sections (~100 μm apart) and data were averaged per mouse to obtain individual values (circles) as illustrated. (e) 3D reconstructions of 880 x 330 x 1000 μm sections of Pdgfrb+/+ and Pdgfrb+/− vasculature in the S1 somatosensory cortex region. Scale bar = 200 μm. Insets: Single 50 μm thick slices horizontally through cortex cortex layer IV. (f) Comparison of vascular density with depth in cortex of Pdgfrb+/− and Pdgfrb+/+ mice for all vessels <35 μm diameter (top; t-test, single tail, equal variance: t = 1.68, p = 0.07), and vascular density of capillaries (< 6 μm diameter; t-test, single tail, equal variance: t = 1.77, p = 0.06) and larger vessels between 6-35 μm (bottom; t-test, two tail, equal variance: t = 0.10, p = 0.92) measured in Pdgfrb+/− and Pdgfrb+/+ mice. Lines represent average values ± 95% CI from n = 4 mice per group. T-tests based on average vessel density through all cortical layers per mouse.

Source data

Supplementary Figure 5 Capillary length, pericyte coverage and vessel dilation in young Meox2+/− mice.

(a) Representative image of pericyte coverage (CD13, magenta; left) of lectin-positive brain endothelial capillary profiles (< 6 μm in diameter; lectin, blue; middle) and overlay (right) in Meox2+/− and Meox2+/+ control mice. (b,c) Quantification (mean ± 95% CI) of total capillary length calculated from the length of lectin-positive endothelial profiles < 6 μm in diameter (b, Mann-Whitney U test, single tail, p = 0.05), and pericyte coverage of lectin-positive brain endothelial capillary profiles (c, Mann-Whitney U test, p = 1.00) in 3 control Meox2+/+ and 3 Meox2+/− mice (b), and 5 control Meox2+/+ and 3 Meox2+/− mice (c). In each animal 6 randomly selected fields from the cortex were analyzed in 5 nonadjacent sections (~100 μm apart) and data were averaged per mouse to obtain individual values (circles) as illustrated. (d, e) Average time (mean ± CI) to 50% peak capillary diameter (d, Mann-Whitney U test, p = 0.79) (d) or 50% peak arteriole diameter (e, Mann-Whitney U test, p = 0.55) was determined after an electrical hind limb stimulation (10 s, 10 Hz, 2 ms pulse duration) in capillaries from 7 control Meox2+/+ (16 total capillaries) mice and 3 Meox2+/− mice (15 total capillaries), and arterioles from 6 control Meox2+/+ mice (13 total arterioles) and 3 Meox2+/− mice (12 total arterioles). Sigmoid parametric fit analysis was performed as for figure 1 c–e. Bootstrapped in panels b–e.

Source data

Supplementary Figure 6 Lactate and glucose plasma levels in 1- to 2-month-old pericyte-deficient Pdgfrb+/− mice and age-matched littermate controls.

(a) Serum lactate levels (mean + 95% CI) in 5 Pdgfrb+/+ and 5 Pdgfrb+/− mice (t-test, equal variance: t = 0.51, p = 0.63). (b) Serum glucose levels (mean ± 95% CI.) in 6 Pdgfrb+/+ and 4 Pdgfrb+/− mice (t-test, equal variance: t = 0.08, p = 0.94).

Source data

Supplementary Figure 7 Cortical neuronal activity in 1- to 2-month-old pericyte-deficient Pdgfrb+/− mice and age-matched littermate controls.

(a) Representative pseudo-colored voltage-sensitive dye (VSD) image sequences of cortical neuronal activity in the S1 region in response to a 300 ms mechanical hind limb stimulus in Pdgfrb+/+ and Pdgfrb+/− mice. Hind limb region (HL) is indicated by dashed line. Scale bar = 0.5 mm. (b-d) Representative VSD intensity traces (b), time to peak (Mann-Whitney U test, p = 0.84; bootstrapped) (c) and peak fluorescence change (t-test, two tailed, equal variance: t = 0.11, p = 0.92) (d) in Pdgfrb+/+ and Pdgfrb+/− mice. Arrow in (b) indicates peak VSD signal for Pdgfrb+/− (blue) and littermate control Pdgfrb+/+ (red) mice. Mean ± 95% CI, from n = 5 mice per group.

Source data

Supplementary Figure 8 Cerebral blood flow response and pericyte coverage in 6- to 8-month-old pericyte-deficient Pdgfrb+/− mice and age-matched littermate controls.

(a) Cerebral blood flow (CBF) response to an electrical hind limb stimulus (60 s, 7 Hz, 2 ms pulse duration) determined by laser doppler flowmetry (LDF) as a percentage of baseline change in 4 Pdgfrb+/− mice and 4 Pdgfrb+/+ littermate controls at 6-8 months of age. Circles denote individual values derived from 3 independent LDF measurements per mouse; mean + 95% CI (Mann-Whitney U test, p = 0.03; bootstrapped). (b) Representative confocal microscopy images of pericyte coverage (CD13, magenta) of lectin-positive brain endothelial capillary profiles (< 6 μm in diameter; lectin, blue) in the S1 cortical area in Pdgfrb+/+ and Pdgfrb+/− mice at 6-8 mo of age. (c) Quantification (mean ± 95% CI) of CD13-positive pericyte coverage of cortical capillaries in 4 Pdgfrb+/+ and 4 Pdgfrb+/− mice as in b (t-test, single tail, equal variance: t = 4.89, p = 0.001). In each animal 6 randomly selected fields from the cortex were analyzed in 5 nonadjacent sections (~100 μm apart) and data were averaged per mouse to obtain individual values (circles) as illustrated.

Source data

Supplementary information

Supplementary Text and Figures

Supplementary Figures 1–8 and Supplementary Table 1 (PDF 1134 kb)

Supplementary Methods Checklist (PDF 539 kb)

Illustration of diameter change upon stimulus in a pericyte-covered capillary in vivo in a pericyte-deficient Pdgfrb+/−;dsRed mouse using TPLSM.

Beginning of the movie shows a still image indicating pericyte location on a vessel before zooming to the magnification used for time-lapse imaging. The second part of the movie shows time-lapse imaging of vessel dilation in response to stimulus. Dashed lines indicate vessel's pre-stimulus diameter. Arrowheads indicate maximal extent of dilation during stimulus. Appearance of white dot in upper left corner indicates application of stimulus. Images were acquired at 1.57 s/frame and played at 2 fps. (AVI 1048 kb)

Illustration of a minimal diameter change upon stimulus in a pericyte-absent capillary in vivo in a pericyte-deficient Pdgfrb+/−;dsRed mouse using TPLSM.

Beginning of the movie shows a still image of a pericyte-free vessel before zooming to the magnification used for time-lapse imaging. The second part of the movie shows time-lapse imaging of vessel dilation in response to stimulus. Dashed lines indicate vessel's pre-stimulus diameter. Arrowheads indicate maximal extent of dilation during stimulus. Appearance of white dot in upper left corner indicates application of stimulus. Images were acquired at 1.57 s/frame and played at 2 fps. (AVI 969 kb)

Source data

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Kisler, K., Nelson, A., Rege, S. et al. Pericyte degeneration leads to neurovascular uncoupling and limits oxygen supply to brain. Nat Neurosci 20, 406–416 (2017). https://doi.org/10.1038/nn.4489

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nn.4489

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