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.

  • Letter
  • Published:

Blood–brain barrier breakdown is an early biomarker of human cognitive dysfunction

Abstract

Vascular contributions to cognitive impairment are increasingly recognized1,2,3,4,5 as shown by neuropathological6,7, neuroimaging4,8,9,10,11, and cerebrospinal fluid biomarker4,12 studies. Moreover, small vessel disease of the brain has been estimated to contribute to approximately 50% of all dementias worldwide, including those caused by Alzheimer’s disease (AD)3,4,13. Vascular changes in AD have been typically attributed to the vasoactive and/or vasculotoxic effects of amyloid-β (Aβ)3,11,14, and more recently tau15. Animal studies suggest that Aβ and tau lead to blood vessel abnormalities and blood–brain barrier (BBB) breakdown14,15,16. Although neurovascular dysfunction3,11 and BBB breakdown develop early in AD1,4,5,8,9,10,12,13, how they relate to changes in the AD classical biomarkers Aβ and tau, which also develop before dementia17, remains unknown. To address this question, we studied brain capillary damage using a novel cerebrospinal fluid biomarker of BBB-associated capillary mural cell pericyte, soluble platelet-derived growth factor receptor-β8,18, and regional BBB permeability using dynamic contrast-enhanced magnetic resonance imaging8,9,10. Our data show that individuals with early cognitive dysfunction develop brain capillary damage and BBB breakdown in the hippocampus irrespective of Alzheimer’s Aβ and/or tau biomarker changes, suggesting that BBB breakdown is an early biomarker of human cognitive dysfunction independent of Aβ and tau.

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

Fig. 1: Early brain capillary damage and BBB breakdown in the human HC and PHG in individuals with an increased CDR score is independent of Aβ and tau status.
Fig. 2: Early brain capillary damage and BBB breakdown in human HC and PHG in individuals with an increased CDR score is independent of HC and PHG volume.
Fig. 3: Early brain capillary damage and BBB breakdown in human HC and PHG in individuals with an increased cognitive domain impairment is independent of Aβ and tau status.
Fig. 4: Early brain capillary damage and BBB breakdown in human HC and PHG in individuals with an increased cognitive domain impairment is independent of HC and PHG volume.

Similar content being viewed by others

Data availability

All data included in this study are available in Supplementary Tables containing detailed statistical analyses and in the accompanying Source Data. The data that support the findings of this study are available from the corresponding author upon reasonable request.

References

  1. Snyder, H. M. et al. Vascular contributions to cognitive impairment and dementia including Alzheimer’s disease. Alzheimers Dement. 11, 710–717 (2015).

    Article  Google Scholar 

  2. Gottesman, R. F. et al. Association between midlife vascular risk factors and estimated brain amyloid deposition. JAMA 317, 1443–1450 (2017).

    Article  CAS  Google Scholar 

  3. Iadecola, C. The neurovascular unit coming of age: a journey through neurovascular coupling in health and disease. Neuron 96, 17–42 (2017).

    Article  CAS  Google Scholar 

  4. Sweeney, M. D., Sagare, A. P. & Zlokovic, B. V. Blood–brain barrier breakdown in Alzheimer disease and other neurodegenerative disorders. Nat. Rev. Neurol. 14, 133–150 (2018).

    Article  CAS  Google Scholar 

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

    Article  CAS  Google Scholar 

  6. Arvanitakis, Z., Capuano, A. W., Leurgans, S. E., Bennett, D. A. & Schneider, J. A. Relation of cerebral vessel disease to Alzheimer’s disease dementia and cognitive function in elderly people: a cross-sectional study. Lancet Neurol. 15, 934–943 (2016).

    Article  CAS  Google Scholar 

  7. Toledo, J. B. et al. Contribution of cerebrovascular disease in autopsy confirmed neurodegenerative disease cases in the National Alzheimer’s Coordinating Centre. Brain 136, 2697–2706 (2013).

    Article  Google Scholar 

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

    Article  CAS  Google Scholar 

  9. van de Haar, H. J. et al. Neurovascular unit impairment in early Alzheimer’s disease measured with magnetic resonance imaging. Neurobiol. Aging 45, 190–196 (2016).

    Article  Google Scholar 

  10. van de Haar, H. J. et al. Blood–brain barrier leakage in patients with early Alzheimer disease. Radiology 281, 527–535 (2016).

    Article  Google Scholar 

  11. Kisler, K., Nelson, A. R., Montagne, A. & Zlokovic, B. V. Cerebral blood flow regulation and neurovascular dysfunction in Alzheimer disease. Nat. Rev. Neurosci. 18, 419–434 (2017).

    Article  CAS  Google Scholar 

  12. Iturria-Medina, Y. et al. Early role of vascular dysregulation on late-onset Alzheimer’s disease based on multifactorial data-driven analysis. Nat. Commun. 7, 11934 (2016).

    Article  CAS  Google Scholar 

  13. Wardlaw, J. M. et al. Neuroimaging standards for research into small vessel disease and its contribution to ageing and neurodegeneration. Lancet Neurol. 12, 822–838 (2013).

    Article  Google Scholar 

  14. Montagne, A., Zhao, Z. & Zlokovic, B. V. Alzheimer’s disease: a matter of blood–brain barrier dysfunction? J. Exp. Med. 214, 3151–3169 (2017).

    Article  CAS  Google Scholar 

  15. Bennett, R. E. et al. Tau induces blood vessel abnormalities and angiogenesis-related gene expression in P301L transgenic mice and human Alzheimer’s disease. Proc. Natl Acad. Sci. USA 115, E1289–E1298 (2018).

    Article  CAS  Google Scholar 

  16. Blair, L. J. et al. Tau depletion prevents progressive blood–brain barrier damage in a mouse model of tauopathy. Acta Neuropathol. Commun. 3, 8 (2015).

    Article  Google Scholar 

  17. Jack, C. R. Jr. et al. NIA-AA Research Framework: toward a biological definition of Alzheimer’s disease. Alzheimers Dement. 14, 535–562 (2018).

    Article  Google Scholar 

  18. Sagare, A. P., Sweeney, M. D., Makshanoff, J. & Zlokovic, B. V. Shedding of soluble platelet-derived growth factor receptor-β from human brain pericytes. Neurosci. Lett. 607, 97–101 (2015).

    Article  CAS  Google Scholar 

  19. Pan, C. et al. Diagnostic values of cerebrospinal fluid t-tau and Aβ42 using meso scale discovery assays for Alzheimer’s disease. J. Alzheimers Dis. 45, 709–719 (2015).

    Article  CAS  Google Scholar 

  20. Roe, C. M. et al. Amyloid imaging and CSF biomarkers in predicting cognitive impairment up to 7.5 years later. Neurology 80, 1784–1791 (2013).

    Article  CAS  Google Scholar 

  21. Bondi, M. W. et al. Neuropsychological criteria for mild cognitive impairment improves diagnostic precision, biomarker associations, and progression rates. J. Alzheimers Dis. 42, 275–289 (2014).

    Article  Google Scholar 

  22. 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  Google Scholar 

  23. 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  Google Scholar 

  24. 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  Google Scholar 

  25. Vanlandewijck, M. et al. A molecular atlas of cell types and zonation in the brain vasculature. Nature 554, 475–480 (2018).

    Article  CAS  Google Scholar 

  26. Trost, A. et al. Brain and retinal pericytes: origin, function and role. Front. Cell. Neurosci. 10, 20 (2016).

    Article  Google Scholar 

  27. Mendelson, K., Swendeman, S., Saftig, P. & Blobel, C. P. Stimulation of platelet-derived growth factor receptor β (PDGFRβ) activates ADAM17 and promotes metalloproteinase-dependent cross-talk between the PDGFRβ and epidermal growth factor receptor (EGFR) signaling pathways. J. Biol. Chem. 285, 25024–25032 (2010).

    Article  CAS  Google Scholar 

  28. Sweeney, M. D., Sagare, A. P. & Zlokovic, B. V. Cerebrospinal fluid biomarkers of neurovascular dysfunction in mild dementia and Alzheimer’s disease. J. Cereb. Blood Flow Metab. 35, 1055–1068 (2015).

    Article  CAS  Google Scholar 

  29. Wallin, A., Blennow, K. & Rosengren, L. Cerebrospinal fluid markers of pathogenetic processes in vascular dementia, with special reference to the subcortical subtype. Alzheimer Dis. Assoc. Disord. 13 Suppl 3, S102–S105 (1999).

    CAS  PubMed  Google Scholar 

  30. 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  Google Scholar 

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

    Article  CAS  Google Scholar 

  32. Spellman, T. et al. Hippocampal-prefrontal input supports spatial encoding in working memory. Nature 522, 309–314 (2015).

    Article  CAS  Google Scholar 

  33. Tan, Z. et al. Dynamic ErbB4 activity in hippocampal-prefrontal synchrony and top-down attention in rodents. Neuron 98, 380–393.e4 (2018).

    Article  CAS  Google Scholar 

  34. Croxson, P. L. et al. Quantitative investigation of connections of the prefrontal cortex in the human and macaque using probabilistic diffusion tractography. J. Neurosci. 25, 8854–8866 (2005).

    Article  CAS  Google Scholar 

  35. O’Shea, A., Cohen, R. A., Porges, E. C., Nissim, N. R. & Woods, A. J. Cognitive aging and the hippocampus in older adults. Front. Aging Neurosci. 8, 298 (2016).

    Article  Google Scholar 

  36. Glikmann-Johnston, Y., Oren, N., Hendler, T. & Shapira-Lichter, I. Distinct functional connectivity of the hippocampus during semantic and phonemic fluency. Neuropsychologia 69, 39–49 (2015).

    Article  Google Scholar 

  37. Rodríguez-Aranda, C. et al. Neuroanatomical correlates of verbal fluency in early Alzheimer’s disease and normal aging. Brain Lang. 155–156, 24–35 (2016).

    Article  Google Scholar 

  38. Grahn, J. A., Parkinson, J. A. & Owen, A. M. The cognitive functions of the caudate nucleus. Prog. Neurobiol. 86, 141–155 (2008).

    Article  Google Scholar 

  39. Provost, J.-S., Hanganu, A. & Monchi, O. Neuroimaging studies of the striatum in cognition Part I: healthy individuals. Front. Syst. Neurosci. 9, 140 (2015).

    Article  Google Scholar 

  40. Drachman, D. A. The amyloid hypothesis, time to move on: amyloid is the downstream result, not cause, of Alzheimer’s disease. Alzheimers Dement. 10, 372–380 (2014).

    Article  Google Scholar 

  41. Jak, A. J. et al. Quantification of five neuropsychological approaches to defining mild cognitive impairment. Am. J. Geriatr. Psychiatry 17, 368–375 (2009).

    Article  Google Scholar 

  42. Jak, A. J. et al. Neuropsychological criteria for mild cognitive impairment and dementia risk in the Framingham Heart Study. J. Int. Neuropsychol. Soc. 22, 937–943 (2016).

    Article  Google Scholar 

  43. Delis D., Kramer J., Kaplan E. & Ober B. California Verbal Learning Test. (Psychological Corporation: San Antonio, 2000).

  44. Nation, D. A. et al. Antemortem pulse pressure elevation predicts cerebrovascular disease in autopsy-confirmed Alzheimer’s disease. J. Alzheimers Dis. 30, 595–603 (2012).

    Article  Google Scholar 

  45. Bangen, K. J. et al. Aggregate effects of vascular risk factors on cerebrovascular changes in autopsy-confirmed Alzheimer’s disease. Alzheimers Dement. 11, 394–403.e1 (2015).

    Article  Google Scholar 

  46. Hölttä, M. et al. Evaluating amyloid-β oligomers in cerebrospinal fluid as a biomarker for Alzheimer’s disease. PLoS ONE 8, e66381 (2013).

    Article  Google Scholar 

  47. Sengupta, U. et al. Tau oligomers in cerebrospinal fluid in Alzheimer’s disease. Ann. Clin. Transl. Neurol. 4, 226–235 (2017).

    Article  CAS  Google Scholar 

  48. Verbeek, M. M., Otte-Höller, I., Wesseling, P., Ruiter, D. J. & de Waal, R. M. Induction of alpha-smooth muscle actin expression in cultured human brain pericytes by transforming growth factor-beta 1. Am. J. Pathol. 144, 372–382 (1994).

    CAS  PubMed  PubMed Central  Google Scholar 

  49. Barnes, S. R. et al. ROCKETSHIP: a flexible and modular software tool for the planning, processing and analysis of dynamic MRI studies. BMC Med. Imaging 15, 19 (2015).

    Article  Google Scholar 

  50. Patlak, C. S. & Blasberg, R. G. Graphical evaluation of blood-to-brain transfer constants from multiple-time uptake data. Generalizations. J. Cereb. Blood Flow Metab. 5, 584–590 (1985).

    Article  CAS  Google Scholar 

  51. Fischl, B. FreeSurfer. Neuroimage 62, 774–781 (2012).

    Article  Google Scholar 

  52. Fischl, B. et al. Whole brain segmentation: automated labeling of neuroanatomical structures in the human brain. Neuron 33, 341–355 (2002).

    Article  CAS  Google Scholar 

  53. Desikan, R. S. et al. An automated labeling system for subdividing the human cerebral cortex on MRI scans into gyral based regions of interest. Neuroimage 31, 968–980 (2006).

    Article  Google Scholar 

  54. Fischl, B. & Dale, A. M. Measuring the thickness of the human cerebral cortex from magnetic resonance images. Proc. Natl Acad. Sci. USA 97, 11050–11055 (2000).

    Article  CAS  Google Scholar 

  55. Fischl, B., Sereno, M. I., Tootell, R. B. & Dale, A. M. High-resolution intersubject averaging and a coordinate system for the cortical surface. Hum. Brain Mapp. 8, 272–284 (1999).

    Article  CAS  Google Scholar 

  56. Dinov, I. et al. Neuroimaging study designs, computational analyses and dataprovenance using the LONI pipeline. PLoS ONE 5, e13070 (2010).

    Article  Google Scholar 

  57. Sepehrband, F. et al. Neuroanatomical morphometric characterization of sex differences in youth using statistical learning. Neuroimage 172, 217–227 (2018).

    Article  Google Scholar 

  58. Su, Y. et al. Quantitative analysis of PiB-PET with FreeSurfer ROIs. PLoS ONE 8, e73377 (2013).

    Article  CAS  Google Scholar 

  59. Gordon, B. A. et al. Longitudinal β-amyloid deposition and hippocampal volume in preclinical Alzheimer disease and suspected non-Alzheimer disease pathophysiology. JAMA Neurol. 73, 1192–1200 (2016).

    Article  Google Scholar 

  60. Glickman, M. E., Rao, S. R. & Schultz, M. R. False discovery rate control is a recommended alternative to Bonferroni-type adjustments in health studies. J. Clin. Epidemiol. 67, 850–857 (2014).

    Article  Google Scholar 

Download references

Acknowledgements

The work of B.V.Z. is supported by the National Institutes of Health (NIH) grant nos. R01AG023084, R01NS090904, R01NS034467, R01AG039452, 1R01NS100459, 5P01AG052350, and 5P50AG005142 in addition to the Alzheimer’s Association strategic 509279 grant, Cure Alzheimer’s Fund, and the Foundation Leducq Transatlantic Network of Excellence for the Study of Perivascular Spaces in Small Vessel Disease reference no. 16 CVD 05. D.B and M.G.H. are supported by the L.K. Whittier Foundation, grant nos. P01AG052350, RO1AG054434, and R01AG055770. D.N. is supported by the NIH grant no. R21AG055034 and Alzheimer’s Association grant no. AARG-17–532905. Enrollment of participants into the Washington University Knight ADRC is supported by NIH grant nos. P50AG05681 (J.C.M.), P01AG03991 (J.C.M.), and P01AG026276 (J.C.M.). Enrollment of participants into the USC ADRC is supported by NIH grant no. 5P50AG005142 (H.C.C.).

Author information

Authors and Affiliations

Authors

Contributions

D.A.N., M.D.S., A.M., A.P.S., and B.V.Z. designed the research study and analyzed and interpreted the data. M.D.S., A.M., A.P.S., L.M.D., M.P., F.S., and D.P.B. performed the experiments and analyzed the data. L.M.D. and A.R.N. prepared and submitted the study to the IRB. M.G.H., T.L.S.B., A.M.F., J.M.R., L.S.S., J.C.M., H.C.C., M.L., and A.W.T. recruited the participants and performed and provided the imaging scans. A.P.S., M.G.H., T.L.S.B., A.M.F., J.M.R., L.S.S., J.C.M., H.C.C., M.L., and A.W.T. provided critical reading of the manuscript. D.A.N., M.D.S., and A.M. contributed to manuscript writing and B.V.Z. wrote the manuscript.

Corresponding author

Correspondence to Berislav V. Zlokovic.

Ethics declarations

Competing interests

The authors declare no competing interests.

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 ADAM10 mediates sPDGFRβ shedding in human brain pericytes in vitro.

a, Primary human brain vascular SMCs and pericytes were subjected to treatment with ionomycin (2.5 µM), a calcium ionophore that activates ADAM10, or control treatment (media only); media was immunoprecipitated to measure sPDGFRβ by quantitative western blot. Compared to pericytes, SMCs shed extremely low levels of sPDGFRβ, which was not significantly increased by ionomycin. Pericytes shed high basal levels of sPDGFRβ that was significantly increased fivefold by treatment with ionomycin, which activated ADAM10. To further determine the involvement of ADAM10, ionomycin treatment was conducted in the presence of the pharmacological inhibition of ADAM10 with marimastat (4 µM), which inhibits ADAM10 by binding to active-site zinc, and genetic small interfering RNA (siRNA) knockdown of ADAM10. Both pharmacological (marimastat) and genetic (siRNA) inhibition of ADAM10 significantly reduced sPDGFRβ shedding activated by ionomycin by >90 and 75%, respectively. b, The siRNA ADAM10 knockdown efficiency in this study was 85%, as shown by the western blot analysis. Data was generated from n = 3–6 independent culture experiments and plotted as means ± s.e.m. a, SMC data by two-tailed Student’s t-test; pericyte data by ANOVA with Tukey post-hoc test. b, Two-tailed Student’s t-test. Significance set at α = 0.05 for all analyses.

Extended Data Fig. 2 CSF sPDGFRβ increases with CDR impairment, independent of Aβ and tau, and reflects BBB breakdown.

ab, Site-specific analysis of CSF sPDGFRβ and the standard AD biomarkers, Aβ42 and pTau, indicates an early increase in sPDGFRβ with increasing CDR at both independent clinical sites, USC (a) and Washington University (b). There were no changes in Aβ42 and pTau at the USC site (a), whereas Aβ42, but not pTau, was altered at the Washington University site; supports Fig. 1a–c. cd, Site-specific analysis of CSF sPDGFRβ increases with CDR, independent of CSF Aβ42 and pTau status at two independent sites, USC (c) and Washington University (d); supports Fig. 1d–f. ef, CSF sPDGFRβ is associated with BBB breakdown. CSF sPDGFRβ positively correlates with the conventional biochemical biomarkers of BBB breakdown including the CSF/plasma albumin ratio (Qalb) (e) and CSF fibrinogen (f); supports Figs. 1 and 3. g, CSF sPDGFRβ is increased with CDR, independent of amyloid positivity by 11C-PiB-PET; supports Fig. 1d–f. hi, No differences were observed in CSF Aβ (h) and tau (i) oligomer levels in individuals with CDR 0 versus CDR 0.5; supports Fig. 1d–f. jk, Increases in CSF sPDGFRβ (j) and regional BBB Ktrans in the HC and PHG (k) of individuals with CDR 0.5 versus CDR 0 remain significant after statistically controlling for the impact of CSF tau oligomers; supports Fig. 1d–f. ad, gi, The box and whisker plot lines indicate the median values, the boxes indicate the IQR, and the whiskers indicate the minimum and maximum values. ad, g, Significance tests from ANCOVAs. ef, Statistical significance determined by Pearson correlation coefficient (r). hi, Significance by two-tailed Student’s t-test at α = 0.05. jk, ANCOVA models representing estimated marginal means ± s.e.m. The brackets denote the sample size (n) in each analysis.

Extended Data Fig. 3 sPDGFRβ increases with CDR independent of VRFs, and no change in other neurovascular unit biomarkers.

ac, CSF sPDGFRβ is increased with CDR, independent of VRF burden in the combined site analysis (a) and at two independent clinical sites from USC (b) and Washington University (c). VRFs 0–1: no or 1 VRF. VRFs 2+: 2 or more VRFs. See Supplementary Table 1 for the list of VRFs; supports Fig. 1a–f. The box and whisker plot lines indicate the median values, the boxes indicate the IQR, and the whiskers indicate the minimum and maximum values. Significance tests from ANCOVAs. The brackets denote the sample size (n) in each analysis.

Extended Data Fig. 4 Other CSF biomarkers of the neurovascular unit are not altered with CDR cognitive impairment.

ac, CSF markers of glial, inflammatory, or neuronal injury exhibited no significant differences between unimpaired and impaired individuals on CDR, including S100B, IL-6, TNFα, or NSE in the combined site analysis (a) and similarly in the site-specific analysis of individuals from USC (b) and Washington University (c); supports Fig. 1a–c. The box and whisker plot lines indicate the median values, the boxes indicate the IQR, and the whiskers indicate the minimum and maximum values. Significance tests from ANCOVAs. The brackets denote the sample size (n) in each analysis.

Extended Data Fig. 5 Regional BBB breakdown Ktrans increases with CDR independent of CSF Aβ, tau, and VRFs, and relates to sPDGFRβ only in hippocampal gray matter regions.

ab, An increase in Ktrans values in the HC, PHG, and the CA1, CA3, and dentate gyrus HC subfields, with increasing CDR (a), but not in other brain regions including the superior frontal cortical gyrus and inferior temporal cortical gyrus, white matter regions including the subcortical white matter fibers, corpus callosum, and internal capsule, and deep gray matter regions including the thalamus, caudate nucleus, and striatum (b). cd, Additional brain regions showed no significant differences in Ktrans BBB permeability values in individuals with CDR 0 and CDR 0.5, regardless of CSF Aβ42 (c) or pTau (d) status. ef, VRF burden does not influence an increase in the Ktrans BBB permeability values with increasing CDR in the HC, PHG, and HC subfields (that is, CA1, CA3, and dentate gyrus) (e), and no change in the Ktrans BBB permeability values in other brain regions (f). See Supplementary Table 1 for the list of VRFs. af, Supports Fig. 1g–k. gj, CSF sPDGFRβ is associated with BBB breakdown measured by neuroimaging in hippocampal gray matter regions (gh), but not in white matter regions (ij); supports Figs. 1 and 3. af, The box and whisker plot lines indicate the median values, the boxes indicate the IQR, and the whiskers indicate the minimum and maximum values. Significance tests after FDR correction from ANCOVAs. gj, Statistical significance determined by Pearson correlation coefficient (r) . The brackets denote the sample size (n) in each analysis; applies to all regions within each panel.

Extended Data Fig. 6 CSF sPDGFRβ increases with CDR impairment, independent of Aβ, tau, and VRFs.

ab, Site-specific analysis of CSF sPDGFRβ and the standard AD biomarkers, Aβ42 and pTau, indicates an early increase in sPDGFRβ with increasing impaired domains at both independent clinical sites, USC (a) and Washington University (b); supports Fig. 3a–c. cd, Site-specific analysis of CSF sPDGFRβ indicates increases with the number of impaired cognitive domains, independent of CSF Aβ42 and pTau status at two independent sites, USC (c) and Washington University (d); supports Fig. 3d–f. eg, CSF sPDGFRβ is increased with the increasing number of impaired cognitive domains, independent of VRF burden in the combined site analysis (e) and at two independent clinical sites, USC (f) and Washington University (g). VRFs 0–1: no or 1 VRF. VRFs 2+: 2 or more VRFs. See Supplementary Table 2 for the list of VRFs. Supports Fig. 3a–f. ag, The box and whisker plot lines indicate the median values, the boxes indicate the IQR, and the whiskers indicate the minimum and maximum values. Significance tests from ANCOVAs. The brackets denote the sample size (n) in each analysis.

Extended Data Fig. 7 BBB breakdown is independent of amyloid and tau oligomers.

a, CSF sPDGFRβ is increased with impaired cognitive domains, independent of amyloid positivity by PiB-PET; supports Fig. 3d–f. bc, No differences were observed in CSF Aβ (b) and tau (c) oligomer levels in individuals with 0 or 1+ impaired cognitive domains. de, Increases in CSF sPDGFRβ (d) and regional BBB Ktrans in the HC and PHG (e) of individuals with 1+ versus 0 cognitive domain impairment remain significant after statistically controlling for the impact of CSF tau oligomers; supports Fig. 3d–f. ac, The box and whisker plot lines indicate the median values, the boxes indicate the IQR, and the whiskers indicate the minimum and maximum values. a, Significance tests from ANCOVAs. bc, Significance by two-tailed Student’s t-test at α = 0.05. de, ANCOVA models representing the estimated marginal means ± s.e.m. The brackets denote the sample size (n) in each analysis.

Extended Data Fig. 8 Other CSF biomarkers of the neurovascular unit are not altered with cognitive domain impairment.

ac, CSF markers of glial, inflammatory, or neuronal injury exhibited no significant differences between unimpaired and impaired individuals on neuropsychological exams, including S100B, IL-6*, TNFα, or NSEin the combined site analysis (a) or in the site-specific analysis of individuals from USC (b) or from Washington University (c). ac, The box and whisker plot lines indicate the median values, the boxes indicate the IQR, and the whiskers indicate the minimum and maximum values. Significance tests after FDR correction from ANCOVAs with post-hoc Bonferroni comparisons. The brackets denote the sample size (n) in each analysis. aAnalysis did not survive significance after FDR correction. bIndividual group comparison P values reported because the omnibus test was P < 0.05 but the post-hoc group comparisons were null. Supports Fig. 3a–c.

Extended Data Fig. 9 Regional BBB breakdown Ktrans increases with cognitive domain impairment, independent of CSF Aβ, tau, and VRFs.

ab, An increase in Ktrans values in the HC, PHG, and the CA1, CA3, and dentate gyrus HC subfields with increasing cognitive impairment measured by the number of impaired cognitive domains (a), but not in other brain regions, including the superior frontal cortical gyrus and the inferior temporal cortical gyrus, white matter regions including the subcortical white matter fibers, corpus callosum, and internal capsule, and deep gray matter regions including thalamus, caudate nucleus, and striatum (b). cd, Additional brain regions showed no significant difference in Ktrans BBB permeability in individuals with 0 and 1+ impaired cognitive domains, regardless of CSF Aβ42 (c) and pTau (d) status. ef, Ktrans BBB permeability is increased with increasing cognitive domain impairment in the HC, PHG, and HC subfields (that is, CA1, CA3, and dentate gyrus), independent of VRF burden (e), but not in other brain regions (f). VRFs 0–1: no or 1 VRF; VRFs 2+: 2 or more VRFs. See Supplementary Table 2 for the list of VRFs. af, The box and whisker plot lines indicate the median values, the boxes indicate the IQR, and the whiskers indicate the minimum and maximum values. Significance tests after FDR correction from ANCOVAs. The brackets denote the sample size (n) in each analysis; applies to all regions within each panel. Supports Fig. 3g–k.

Extended Data Fig. 10 CSF sPDGFRβ and medial temporal BBB permeability Ktrans values are not correlated with age, indicating that changes in CSF sPDGFRβ and Ktrans capture processes relating to cognitive impairment are independent of normal aging.

In CDR 0 individuals, age does not correlate with CSF sPDGFRβ (a) or regional Ktrans in the HC (c) and PHG (e). Similarly, in CDR 0.5 individuals, age does not correlate with CSF sPDGFRβ (a) or regional Ktrans in the HC (c) and PHG (e). Statistical significance determined by Pearson correlation coefficient (r); the brackets denote the sample size (n) in each analysis. Supports Figs. 1 and 3.

Supplementary information

Supplementary Table

Supplementary Tables 1–10

Reporting Summary

Source data

Source Data

Full annotated Western blot gels

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Nation, D.A., Sweeney, M.D., Montagne, A. et al. Blood–brain barrier breakdown is an early biomarker of human cognitive dysfunction. Nat Med 25, 270–276 (2019). https://doi.org/10.1038/s41591-018-0297-y

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41591-018-0297-y

This article is cited by

Search

Quick links

Nature Briefing: Translational Research

Sign up for the Nature Briefing: Translational Research newsletter — top stories in biotechnology, drug discovery and pharma.

Get what matters in translational research, free to your inbox weekly. Sign up for Nature Briefing: Translational Research