Becaplermin; Receptor, Platelet-Derived Growth Factor beta; Becaplermin/metabolism; Becaplermin/pharmacology; Brain/metabolism; Humans; Receptor, Platelet-Derived Growth Factor beta/metabolism; Receptor, Platelet-Derived Growth Factor beta/pharmacology; Alzheimer Disease/metabolism; Pericytes; Alzheimer Disease; Brain; Medicine (miscellaneous); Biochemistry, Genetics and Molecular Biology (all); Agricultural and Biological Sciences (all); General Agricultural and Biological Sciences; General Biochemistry, Genetics and Molecular Biology
Abstract :
[en] Platelet-derived growth factor-BB (PDGF-BB):PDGF receptor-β (PDGFRβ) signalling in brain pericytes is critical to the development, maintenance and function of a healthy blood-brain barrier (BBB). Furthermore, BBB impairment and pericyte loss in Alzheimer's disease (AD) is well documented. We found that PDGF-BB:PDGFRβ signalling components were altered in human AD brains, with a marked reduction in vascular PDGFB. We hypothesised that reduced PDGF-BB:PDGFRβ signalling in pericytes may impact on the BBB. We therefore tested the effects of PDGF-BB on primary human brain pericytes in vitro to define pathways related to BBB function. Using pharmacological inhibitors, we dissected distinct aspects of the PDGF-BB response that are controlled by extracellular signal-regulated kinase (ERK) and Akt pathways. PDGF-BB promotes the proliferation of pericytes and protection from apoptosis through ERK signalling. In contrast, PDGF-BB:PDGFRβ signalling through Akt augments pericyte-derived inflammatory secretions. It may therefore be possible to supplement PDGF-BB signalling to stabilise the cerebrovasculature in AD.
Disciplines :
Neurology
Author, co-author :
Smyth, Leon C D ; Department of Pharmacology and Clinical Pharmacology, Faculty of Medical and Health Sciences, University of Auckland, 85 Park Road, Grafton, Auckland, 1023, New Zealand ; Centre for Brain Research, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand ; Centre for Free Radical Research, Department of Pathology and Biomedical Science, University of Otago, Christchurch, New Zealand ; Center for Brain Immunology and Glia, Department of Pathology and Immunology, Washington University in St Louis, Missouri, USA
Highet, Blake; Centre for Brain Research, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand ; Department of Anatomy with Medical Imaging, Faculty of Medical and Health Sciences, University of Auckland, 85 Park Road, Auckland, 1023, New Zealand
Jansson, Deidre ; Department of Pharmacology and Clinical Pharmacology, Faculty of Medical and Health Sciences, University of Auckland, 85 Park Road, Grafton, Auckland, 1023, New Zealand ; Centre for Brain Research, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand ; Department of Psychiatry and Behavioural Sciences, University of Washington, Seattle, Washington, USA
Wu, Jane; Centre for Brain Research, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand ; Department of Anatomy with Medical Imaging, Faculty of Medical and Health Sciences, University of Auckland, 85 Park Road, Auckland, 1023, New Zealand
Rustenhoven, Justin; Department of Pharmacology and Clinical Pharmacology, Faculty of Medical and Health Sciences, University of Auckland, 85 Park Road, Grafton, Auckland, 1023, New Zealand ; Centre for Brain Research, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand ; Center for Brain Immunology and Glia, Department of Pathology and Immunology, Washington University in St Louis, Missouri, USA
Aalderink, Miranda; Department of Pharmacology and Clinical Pharmacology, Faculty of Medical and Health Sciences, University of Auckland, 85 Park Road, Grafton, Auckland, 1023, New Zealand ; Centre for Brain Research, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
Tan, Adelie; Centre for Brain Research, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand ; Department of Anatomy with Medical Imaging, Faculty of Medical and Health Sciences, University of Auckland, 85 Park Road, Auckland, 1023, New Zealand
Li, Susan ; Department of Pharmacology and Clinical Pharmacology, Faculty of Medical and Health Sciences, University of Auckland, 85 Park Road, Grafton, Auckland, 1023, New Zealand ; Centre for Brain Research, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
Johnson, Rebecca; Department of Pharmacology and Clinical Pharmacology, Faculty of Medical and Health Sciences, University of Auckland, 85 Park Road, Grafton, Auckland, 1023, New Zealand ; Centre for Brain Research, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
Coppieters't Wallant, Natacha ; Université de Liège - ULiège > GIGA > GIGA Neurosciences - Nervous system disorders and therapy
Handley, Renee; Centre for Brain Research, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand ; School of Biological Sciences, University of Auckland, Auckland, New Zealand
Narayan, Pritika ; Centre for Brain Research, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand ; School of Biological Sciences, University of Auckland, Auckland, New Zealand
Singh-Bains, Malvindar K; Centre for Brain Research, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand ; Department of Anatomy with Medical Imaging, Faculty of Medical and Health Sciences, University of Auckland, 85 Park Road, Auckland, 1023, New Zealand
Schweder, Patrick; Centre for Brain Research, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand ; Auckland City Hospital, 2 Park Road, Auckland, 1023, New Zealand
Turner, Clinton; Centre for Brain Research, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand ; Auckland City Hospital, 2 Park Road, Auckland, 1023, New Zealand
Mee, Edward W; Centre for Brain Research, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand ; Auckland City Hospital, 2 Park Road, Auckland, 1023, New Zealand
Heppner, Peter; Centre for Brain Research, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand ; Auckland City Hospital, 2 Park Road, Auckland, 1023, New Zealand
Correia, Jason; Centre for Brain Research, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand ; Auckland City Hospital, 2 Park Road, Auckland, 1023, New Zealand
Park, Thomas I-H ; Department of Pharmacology and Clinical Pharmacology, Faculty of Medical and Health Sciences, University of Auckland, 85 Park Road, Grafton, Auckland, 1023, New Zealand ; Centre for Brain Research, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
Curtis, Maurice A; Centre for Brain Research, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand ; Department of Anatomy with Medical Imaging, Faculty of Medical and Health Sciences, University of Auckland, 85 Park Road, Auckland, 1023, New Zealand
Faull, Richard L M; Centre for Brain Research, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand ; Department of Anatomy with Medical Imaging, Faculty of Medical and Health Sciences, University of Auckland, 85 Park Road, Auckland, 1023, New Zealand
Dragunow, Mike ; Department of Pharmacology and Clinical Pharmacology, Faculty of Medical and Health Sciences, University of Auckland, 85 Park Road, Grafton, Auckland, 1023, New Zealand. m.dragunow@auckland.ac.nz ; Centre for Brain Research, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand. m.dragunow@auckland.ac.nz
We would like to thank the donors for their generous gift of brain tissue for research. We also thank the staff at Auckland Hospital, the Hugh Green Biobank, the Neurological Foundation Human Brain Bank and Sheryl Feng and Marika Eszes. We acknowledge the following funding bodies for their support of this research programme: the Hugh Green Foundation, the Health Research Council of New Zealand, the Sir Thomas and Lady Duncan Trust and the Coker Trust. Dr. Leon Smyth was funded by a Neurological Foundation Philip Wrightson Postdoctoral Fellowship. The Accuri C6 was partially funded by Lottery Health New Zealand.
Zlokovic, B. V. Neurovascular mechanisms of Alzheimer’s neurodegeneration. Trends Neurosci. 28, 202–208 (2005).
Yang, A. C. et al. A human brain vascular atlas reveals diverse cell mediators of Alzheimer’s disease risk. bioRxiv https://doi.org/10.1101/2021.04.26.441262 (2021).
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).
Zhang, X. et al. High-resolution mapping of brain vasculature and its impairment in the hippocampus of Alzheimer’s disease mice. Natl. Sci. Rev. 6, 1223–1238 (2019).
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).
Nortley, R. et al. Amyloid-beta oligomers constrict human capillaries in Alzheimer’s disease via signaling to pericytes. Science 365, eaav9518 (2019).
Nation, D. A. et al. Blood–brain barrier breakdown is an early biomarker of human cognitive dysfunction. Nat. Med. 10.1038/s41591-018-0297-y (2019).
Montagne, A. et al. APOE4 leads to blood–brain barrier dysfunction predicting cognitive decline. Nature 10.1038/s41586-020-2247-3 (2020).
Sagare, A. P. et al. Pericyte loss influences Alzheimer-like neurodegeneration in mice. Nat. Commun. 4, 2932 (2013).
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).
Bourassa, P., Tremblay, C., Schneider, J. A., Bennett, D. A. & Calon, F. Brain mural cell loss in the parietal cortex in Alzheimer’s disease correlates with cognitive decline and TDP‐43 pathology. Neuropathol. Appl. Neurobiol. https://doi.org/10.1111/nan.12599 (2020).
Montagne, A. et al. Blood-brain barrier breakdown in the aging human hippocampus. Neuron 85, 296–302 (2015).
Lindahl, P., Johansson, B. R., Levéen, P. & Betsholtz, C. Pericyte loss and microaneurysm formation in PDGF-B-deficient mice. Science 277, 242–245 (1997).
Smyth, L. C. D. et al. Unique and shared inflammatory profiles of human brain endothelia and pericytes. J. Neuroinflamm. 15, 138 (2018).
Vanlandewijck, M. et al. A molecular atlas of cell types and zonation in the brain vasculature. Nature 554, 475–480 (2018).
Armulik, A. et al. Pericytes regulate the blood–brain barrier. Nature 468, 557–561 (2010).
Paul, G. et al. The adult human brain harbors multipotent perivascular mesenchymal stem cells. PLoS ONE 7, e35577 (2012).
Lindblom, P. et al. Endothelial PDGF-B retention is required for proper investment of pericytes in the microvessel wall. Genes Dev. 17, 1835–1840 (2003).
Nakamura, K. et al. Perlecan regulates pericyte dynamics in the maintenance and repair of the blood-brain barrier. J. Cell Biol. 218, 3506–3525 (2019).
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).
Nikolakopoulou, A. M., Zhao, Z., Montagne, A. & Zlokovic, B. V. Regional early and progressive loss of brain pericytes but not vascular smooth muscle cells in adult mice with disrupted platelet-derived growth factor receptor-β signaling. PLoS ONE 12, e0176225 (2017).
Vazquez-Liébanas, E. V. et al. Endothelium-derived PDGF-B is essential for mural cell maintenance and endothelial cell quiescence in the adult brain. Cold Spring Harb. Brain Barriers 185 (2021).
Arango-Lievano, M. et al. Topographic reorganization of cerebrovascular mural cells under seizure conditions. Cell Rep. 23, 1045–1059 (2018).
Geraldes, P. et al. Activation of PKC-δ and SHP-1 by hyperglycemia causes vascular cell apoptosis and diabetic retinopathy. Nat. Med. 15, 1298–1306 (2009).
Chintalgattu, V. et al. Coronary microvascular pericytes are the cellular target of sunitinib malate-induced cardiotoxicity. Sci. Transl. Med. 5, 187ra69 (2013).
Ma, Q. et al. Blood-brain barrier-associated pericytes internalize and clear aggregated amyloid-β42 by LRP1-dependent apolipoprotein E isoform-specific mechanism. Mol. Neurodegener. 13, 57 (2018).
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).
Jansson, D. et al. Interferon-γ blocks signalling through PDGFRβ in human brain pericytes. J. Neuroinflamm. 13, 249 (2016).
Gate, D. et al. Clonally expanded CD8 T cells patrol the cerebrospinal fluid in Alzheimer’s disease. Nature 10.1038/s41586-019-1895-7 (2020).
Stubbing, L. A. et al. Synthesis of peptide homo‐ and heterodimers as potential mimics of platelet‐derived growth factor BB. Peptide Sci. https://doi.org/10.1002/pep2.24150 (2020).
Smyth, L. C. D. et al. Markers for human brain pericytes and smooth muscle cells. J. Chem. Neuroanat. (2018) https://doi.org/10.1016/j.jchemneu.2018.06.001 (2018).
Highet, B. et al. fISHing with immunohistochemistry for housekeeping gene changes in Alzheimer’s disease using an automated quantitative analysis workflow. J. Neurochem. 157, 1270–1283 (2021).
Allen, M. et al. Human whole genome genotype and transcriptome data for Alzheimer’s and other neurodegenerative diseases. Sci. Data 3, 160089 (2016).
Allen, M. et al. Gene expression, methylation and neuropathology correlations at progressive supranuclear palsy risk loci. Acta Neuropathol. 132, 197–211 (2016).
Porsch, H., Mehić, M., Olofsson, B., Heldin, P. & Heldin, C.-H. Platelet-derived growth factor β-receptor, transforming growth factor β type I receptor, and CD44 protein modulate each other’s signaling and stability. J. Biol. Chem. 289, 19747–19757 (2014).
He, L. et al. Analysis of the brain mural cell transcriptome. Sci. Rep. 6, 35108 (2016).
Yamamoto, H., Crow, M., Cheng, L., Lakatta, E. & Kinsella, J. PDGF receptor-to-nucleus signaling of p91 (STAT1α) transcription factor in rat smooth muscle cells. Exp. Cell Res. 222, 125–130 (1996).
Jastrzębski, K. et al. Multiple routes of endocytic internalization of PDGFRβ contribute to PDGF-induced STAT3 signaling. J. Cell Sci. 130, 577–589 (2017).
Yu, Y. et al. PDGF stimulates pulmonary vascular smooth muscle cell proliferation by upregulating TRPC6 expression. Am. J. Physiol. Cell Physiol. 284, C316–C330 (2003).
Lennartsson, J. et al. The Fer tyrosine kinase is important for platelet-derived growth factor-BB-induced signal transducer and activator of transcription 3 (STAT3) protein phosphorylation, colony formation in soft agar, and tumor growth in vivo. J. Biol. Chem. 288, 15736–15744 (2013).
Shen, J. et al. PDGFR-β as a positive regulator of tissue repair in a mouse model of focal cerebral ischemia. J. Cereb. Blood Flow. Metab. 32, 353–367 (2012).
Hellström, M., Kalén, M., Lindahl, P., Abramsson, A. & Betsholtz, C. Role of PDGF-B and PDGFR- β in recruitment of vascular smooth muscle cells and pericytes during embryonic blood vessel formation in the mouse. Development 126, 3047–3055 (1999).
Vazquez-Liebanas, E. et al. Adult-induced genetic ablation distinguishes PDGFB roles in blood-brain barrier maintenance and development. J. Cereb. Blood Flow Metab. https://doi.org/10.1177/0271678X211056395 (2021).
Sweeney, M. D., Ayyadurai, S. & Zlokovic, B. V. Pericytes of the neurovascular unit: key functions and signaling pathways. Nat. Neurosci. 19, 771–783 (2016).
Zlokovic, B. V. Neurovascular pathways to neurodegeneration in Alzheimer’s disease and other disorders. Nat. Rev. Neurosci. 12, 723–738 (2011).
Jew, B. et al. Accurate estimation of cell composition in bulk expression through robust integration of single-cell information. Nat. Commun. 11, 1–11 (2020).
Yang, A. C. et al. Physiological blood–brain transport is impaired with age by a shift in transcytosis. Nature https://doi.org/10.1038/s41586-020-2453-z (2020).
Pfau, S. J. et al. Vascular and perivascular cell profiling reveals the molecular and cellular bases of blood-brain barrier heterogeneity. bioRxiv https://doi.org/10.1101/2021.04.26.441465 (2021).
Roth, M. et al. Regulator of G-protein signaling 5 regulates the shift from perivascular to parenchymal pericytes in the chronic phase after stroke. FASEB J. https://doi.org/10.1096/fj.201900153R (2019).
Shen, J. et al. PDGFR-β restores blood-brain barrier functions in a mouse model of focal cerebral ischemia. J. Cereb. Blood Flow Metabo. https://doi.org/10.1177/0271678X18769515 (2018).
Schmahl, J., Raymond, C. S. & Soriano, P. PDGF signaling specificity is mediated through multiple immediate early genes. Nat. Genet. 39, 52–60 (2007).
Wu, J., Bohanan, C. S., Neumann, J. C. & Lingrel, J. B. KLF2 transcription factor modulates blood vessel maturation through smooth muscle cell migration. J. Biol. Chem. 283, 3942–3950 (2008).
Deaton, R. A., Gan, Q. & Owens, G. K. Sp1-dependent activation of KLF4 is required for PDGF-BB-induced phenotypic modulation of smooth muscle. Am. J. Physiol. 296, H1027–H1037 (2009).
Olson, L. E. & Soriano, P. PDGFRβ signaling regulates mural cell plasticity and inhibits fat development. Dev. Cell 20, 815–826 (2011).
Romashkova, J. A. & Makarov, S. S. NF-κB is a target of AKT in anti-apoptotic PDGF signalling. Nature 401, 86–90 (1999).
Rustenhoven, J. et al. TGF-beta1 regulates human brain pericyte inflammatory processes involved in neurovasculature function. J. Neuroinflamm. 13, 37 (2016).
Rustenhoven, J., Jansson, D., Smyth, L. C. & Dragunow, M. Brain pericytes as mediators of neuroinflammation. Trends Pharmacol. Sci. 38, 291–304 (2017).
Kang, T.-Y. et al. Pericytes enable effective angiogenesis in the presence of proinflammatory signals. Proc. Natl. Acad. Sci. https://doi.org/10.1073/pnas.1913373116 (2019).
Dohgu, S. et al. Brain pericytes contribute to the induction and up-regulation of blood–brain barrier functions through transforming growth factor-β production. Brain Res. 1038, 208–215 (2005).
Chen, J. et al. CD146 coordinates brain endothelial cell-pericyte communication for blood-brain barrier development. Proc. Natl. Acad. Sci. USA 114, E7622–E7631 (2017).
Duan, L. et al. PDGFRβ cells rapidly relay inflammatory signal from the circulatory system to neurons via chemokine CCL2. Neuron 100, 183–200.e8 (2018).
Klement, W. et al. A pericyte‐glia scarring develops at the leaky capillaries in the hippocampus during seizure activity. Epilepsia 60, 1399–1411 (2019).
Jansson, D. et al. A role for human brain pericytes in neuroinflammation. J. Neuroinflamm. 11, 104 (2014).
Waldvogel, H. J., Curtis, M. A., Baer, K., Rees, M. I. & Faull, R. L. M. Immunohistochemical staining of post-mortem adult human brain sections. Nat. Protoc. 1, 2719–2732 (2007).
Singh-Bains, M. K., Mehrabi, N. F., Tan, A. Y. S., Faull, R. L. M. & Dragunow, M. Preparation, construction and high-throughput automated analysis of human brain tissue microarrays for neurodegenerative disease drug development. Nat. Protoc. 16, 2308–2343 (2021).
Narayan, P. J. et al. Assessing fibrinogen extravasation into Alzheimer’s disease brain using high-content screening of brain tissue microarrays. J. Neurosci. Methods 247, 41–49 (2015).
Gibbons, H. M. et al. Cellular composition of human glial cultures from adult biopsy brain tissue. J. Neurosci. Methods 166, 89–98 (2007).
Rustenhoven, J. et al. Modelling physiological and pathological conditions to study pericyte biology in brain function and dysfunction. BMC Neurosci. 19, 6 (2018).
Park, T. I.-H. et al. Cultured pericytes from human brain show phenotypic and functional differences associated with differential CD90 expression. Sci. Rep. 6, 26587 (2016).
Rustenhoven, J. et al. An anti-inflammatory role for C/EBPδ in human brain pericytes. Sci. Rep. 5, 12132 (2015).
Jansson, D. et al. Cardiac glycosides target barrier inflammation of the vasculature, meninges and choroid plexus. Commun. Biol. 4, 1–17 (2021).
Park, T. I.-H. et al. Adult human brain neural progenitor cells (NPCs) and fibroblast-like cells have similar properties in vitro but Only NPCs differentiate into neurons. PLoS ONE 7, e37742 (2012).
Grimsey, N. L., Narayan, P. J., Dragunow, M. & Glass, M. A novel high-throughput assay for the quantitative assessment of receptor trafficking. Clin. Exp. Pharmacol. Physiol. 35, 1377–1382 (2008).