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:

The human brain NGF metabolic pathway is impaired in the pre-clinical and clinical continuum of Alzheimers disease

Abstract

The NGF metabolic pathway entails the proteins that mature pro-nerve growth factor (proNGF) to NGF and those that degrade NGF. Basal forebrain cholinergic neurons require NGF for maintenance of cholinergic phenotype, are critical for cognition, and degenerate early in Alzheimer’s disease (AD). In AD, NGF metabolism is altered, but it is not known whether this is an early phenomenon, nor how it relates to AD pathology and symptomology. We acquired dorsolateral/medial prefrontal cortex samples from individuals with Alzheimer’s dementia, Mild Cognitive Impairment (MCI), or no cognitive impairment with high (HA-NCI) and low (LA-NCI) brain Aβ from the Religious Orders Study. Cortical proNGF protein, but not mRNA, was higher in AD, MCI, and HA-NCI, while mature NGF was lower. Plasminogen protein was higher in MCI and AD brain tissue, with plasminogen mRNA not likewise elevated, suggesting diminished activation of the proNGF convertase, plasmin. The plasminogen activator tPA was lower in HA-NCI while neuroserpin, the CNS tPA inhibitor, was higher in AD and MCI cortical samples. Matrix metalloproteinase 9 (MMP9), which degrades NGF, was overactive in MCI and AD. Transcription of the MMP9 inhibitor TIMP1 was lower in HA-NCI. ProNGF levels correlated with plasminogen, neuroserpin, and VAChT while NGF correlated with MMP9 activity. In NCI, proNGF correlated with cerebral Aβ and tau deposition and to cognitive performance. In summary, proNGF maturation is impaired in preclinical and clinical AD while mature NGF degradation is enhanced. These differences correlate with cognition, pathology, and cholinergic tone, and may suggest novel biomarkers and therapeutic targets.

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

Access options

Buy this article

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

Fig. 1: Schematic representation of the NGF metabolic pathway and its compromise in the continuum of Alzheimer’s disease.
Fig. 2: Normal NGF synthesis in the continuum of AD pathology is accompanied by an increase of proNGF and decrease of mNGF, beginning at preclinical stages, as well as abnormal expression of proteins participating in proNGF maturation.
Fig. 3: Increased levels and protease activity of MMP9, a mature-NGF degrading protease, and diminished expression of its endogenous inhibitor, TIMP1, at AD clinical stages in dorsolateral/medial prefrontal cortex homogenates.
Fig. 4: Association between elevations of dorsolateral/medial prefrontal cortex proNGF protein levels and hallmarks of Alzheimer’s neuropathology and to cognitive scores in individuals with no cognitive impairment (n= 59).
Fig. 5: VAChT expression across the AD continuum and its association with proNGF and neuroserpin expression.

Similar content being viewed by others

References

  1. Jack CR, Knopman DS, Jagust WJ, Petersen RC, Weiner MW, Aisen PS, et al. Tracking pathophysiological processes in Alzheimer’s disease: an updated hypothetical model of dynamic biomarkers. Lancet Neurol. 2013;12:207–16.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  2. Sperling RA, Aisen PS, Beckett LA, Bennett DA, Craft S, Fagan AM, et al. Toward defining the preclinical stages of Alzheimer’s disease: Recommendations from the National Institute on Aging-Alzheimer’s Association workgroups on diagnostic guidelines for Alzheimer’s disease. Alzheimer’s Dement. 2011;7:280–92.

    Article  Google Scholar 

  3. Gauthier S, Albert M, Fox N, Goedert M, Kivipelto M, Mestre-Ferrandiz J, et al. Why has therapy development for dementia failed in the last two decades? Alzheimer’s Dement. 2016;12:60–4.

    Article  Google Scholar 

  4. McDade E, Bateman RJ. Stop Alzheimer’s before it starts. Nature. 2017;547:153.

    Article  CAS  PubMed  Google Scholar 

  5. Epelbaum S, Genthon R, Cavedo E, Habert MO, Lamari F, Gagliardi G, et al. Preclinical Alzheimer’s disease: A systematic review of the cohorts underlying the concept. Alzheimer’s Dementia. 2017;13:454–67.

    Article  PubMed  Google Scholar 

  6. Lanctôt KL, Herrmann N, Yau KK, Khan LR, Liu BA, LouLou MM, et al. Efficacy and safety of cholinesterase inhibitors in Alzheimer’s disease: a meta-analysis. Can Med Assoc J. 2003;169:557–64.

    Google Scholar 

  7. Perry E, Tomlinson B, Blessed G, Perry R, Cross A, Crow T. Noradrenergic and cholinergic systems in senile dementia of Alzheimer type. Lancet. 1981;318:149.

    Article  Google Scholar 

  8. Davies P, Maloney A. Selective loss of central cholinergic neurons in Alzheimer’s disease. Lancet. 1976;308:1403.

    Article  Google Scholar 

  9. Bowen DM, Smith CB, White P, Davison AN. Neurotransmitter-related enzymes and indices of hypoxia in senile dementia and other abiotrophies. Brain: a J Neurol. 1976;99:459–96.

    Article  CAS  Google Scholar 

  10. Coyle JT, Price DL, Delong MR. Alzheimer’s disease: a disorder of cortical cholinergic innervation. Science 1983;219:1184–90.

    Article  CAS  PubMed  Google Scholar 

  11. Pearson R, Sofroniew M, Cuello A, Powell T, Eckenstein F, Esiri M, et al. Persistence of cholinergic neurons in the basal nucleus in a brain with senile dementia of the Alzheimer’s type demonstrated by immunohistochemical staining for choline acetyltransferase. Brain Res. 1983;289:375–9.

    Article  CAS  PubMed  Google Scholar 

  12. Whitehouse PJ, Price DL, Struble RG, Clark AW, Coyle JT, Delon MR. Alzheimer’s disease and senile dementia: loss of neurons in the basal forebrain. Science. 1982;215:1237–9.

    Article  CAS  PubMed  Google Scholar 

  13. Jacobs RW, Butcher LL. Pathology of the basal forebrain in Alzheimer’s disease and other dementias. The biological substrates of Alzheimer’s disease. New York: Academic Press; 1986. p. 87–100.

    Google Scholar 

  14. DeKosky DST, Harbaugh RE, Schmitt FA, Bakay RA, Chui HC, Knopman DS, et al. Cortical biopsy in Alzheimer’s disease: diagnostic accuracy and neurochemical, neuropathological, and cognitive correlations. Ann Neurol. 1992;32:625–32.

    Article  CAS  PubMed  Google Scholar 

  15. Giacobini E, Spiegel R, Enz A, Veroff A, Cutler N. Inhibition of acetyl-and butyryl-cholinesterase in the cerebrospinal fluid of patients with Alzheimer’s disease by rivastigmine: correlation with cognitive benefit. J Neural Transm. 2002;109:1053–65.

    Article  CAS  PubMed  Google Scholar 

  16. Hefti F. Nerve growth factor promotes survival of septal cholinergic neurons after fimbrial transections. J Neurosci: Off J Soc Neurosci. 1986;6:2155–62.

    Article  CAS  Google Scholar 

  17. Cuello AC. Trophic responses of forebrain cholinergic neurons. Prog Brain Res. 1993;98:265-.

    Article  CAS  PubMed  Google Scholar 

  18. Gage FH, Armstrong DM, Williams LR, Varon S. Morphological response of axotomized septal neurons to nerve growth factor. J Comp Neurol. 1988;269:147–55.

    Article  CAS  PubMed  Google Scholar 

  19. Debeir T, Saragovi HU, Cuello AC. A nerve growth factor mimetic TrkA antagonist causes withdrawal of cortical cholinergic boutons in the adult rat. Proc Natl Acad Sci. 1999;96:4067–72.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Goedert M, Fine A, Hunt S, Ullrich A. Nerve growth factor mRNA in peripheral and central rat tissues and in the human central nervous system: lesion effects in the rat brain and levels in Alzheimer’s disease. Mol Brain Res. 1986;1:85–92.

    Article  Google Scholar 

  21. Fahnestock M, Yu G, Michalski B, Mathew S, Colquhoun A, Ross GM, et al. The nerve growth factor precursor proNGF exhibits neurotrophic activity but is less active than mature nerve growth factor. J Neurochem. 2004;89:581–92.

    Article  CAS  PubMed  Google Scholar 

  22. Ioannou MS, Fahnestock M. ProNGF, but Not NGF, switches from neurotrophic to apoptotic activity in response to reductions in TrkA receptor levels. Int J Mol Sci. 2017;18:599.

    Article  PubMed Central  CAS  Google Scholar 

  23. Pedraza CE, Podlesniy P, Vidal N, Arévalo JC, Lee R, Hempstead B, et al. Pro-NGF isolated from the human brain affected by Alzheimer’s disease induces neuronal apoptosis mediated by p75NTR. Am J Pathol. 2005;166:533–43.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Nykjaer A, Lee R, Teng KK, Jansen P, Madsen P, Nielsen MS, et al. Sortilin is essential for proNGF-induced neuronal cell death. Nature. 2004;427:843–8.

    Article  CAS  PubMed  Google Scholar 

  25. Fahnestock M, Michalski B, Xu B, Coughlin MD. The precursor pro-nerve growth factor is the predominant form of nerve growth factor in brain and is increased in Alzheimer’s disease. Mol Cell Neurosci. 2001;18:210–20.

    Article  CAS  PubMed  Google Scholar 

  26. Bruno MA, Cuello AC. Activity-dependent release of precursor nerve growth factor, conversion to mature nerve growth factor, and its degradation by a protease cascade. Proc Natl Acad Sci. 2006;103:6735–40.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Osterwalder T, Contartese J, Stoeckli E, Kuhn T, Sonderegger P. Neuroserpin, an axonally secreted serine protease inhibitor. EMBO J. 1996;15:2944–53.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Krueger SR, Ghisu G-P, Cinelli P, Gschwend TP, Osterwalder T, Wolfer DP, et al. Expression of neuroserpin, an inhibitor of tissue plasminogen activator, in the developing and adult nervous system of the mouse. J Neurosci. 1997;17:8984–96.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Cawston TE, Galloway WA, Mercer E, Murphy G, Reynolds JJ. Purification of rabbit bone inhibitor of collagenase. Biochemical J. 1981;195:159–65.

    Article  CAS  Google Scholar 

  30. Docherty AJ, Lyons A, Smith BJ, Wright EM, Stephens PE, Harris TJ, et al. Sequence of human tissue inhibitor of metalloproteinases and its identity to erythroid-potentiating activity. Nature. 1985;318:66.

    Article  CAS  PubMed  Google Scholar 

  31. Allard S, Leon WC, Pakavathkumar P, Bruno MA, Ribeiro-da-Silva A, Cuello AC. Impact of the NGF maturation and degradation pathway on the cortical cholinergic system phenotype. J Neurosci. 2012;32:2002–12.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Allard S, Jacobs ML, Do Carmo S, Cuello AC. Compromise of cortical proNGF maturation causes selective retrograde atrophy in cholinergic nucleus basalis neurons. Neurobiol Aging. 2018;67:10–20.

    Article  CAS  PubMed  Google Scholar 

  33. Bruno MA, Mufson EJ, Wuu J, Cuello AC. Increased matrix metalloproteinase-9 activity in mild cognitive impairment. J Neuropathol Exp Neurol. 2009;68:1309.

    Article  CAS  PubMed  Google Scholar 

  34. Bruno MA, Leon WC, Fragoso G, Mushynski WE, Almazan G, Cuello AC. Amyloid β-induced nerve growth factor dysmetabolism in Alzheimer disease. J Neuropathol Exp Neurol. 2009;68:857–69.

    Article  CAS  PubMed  Google Scholar 

  35. Fabbro S, Seeds NW. Plasminogen activator activity is inhibited while neuroserpin is up‐regulated in the Alzheimer disease brain. J Neurochem. 2009;109:303–15.

    Article  CAS  PubMed  Google Scholar 

  36. Iulita MF, Bistue Millon MB, Pentz R, Aguilar LF, Do Carmo S, Allard S, et al. Differential deregulation of NGF and BDNF neurotrophins in a transgenic rat model of Alzheimer’s disease. Neurobiol Dis. 2017;108:307–23.

    Article  CAS  PubMed  Google Scholar 

  37. A Bennett D, A Schneider J, Arvanitakis Z, S Wilson R. Overview and findings from the religious orders study. Curr Alzheimer Res. 2012;9:628–45.

    Article  PubMed  Google Scholar 

  38. Bennett D, Schneider J, Arvanitakis Z, Kelly J, Aggarwal N, Shah R, et al. Neuropathology of older persons without cognitive impairment from two community-based studies. Neurology. 2006;66:1837–44.

    Article  CAS  PubMed  Google Scholar 

  39. McKhann G, Drachman D, Folstein M, Katzman R, Price D, Stadlan EM. Clinical diagnosis of Alzheimer’s disease: report of the NINCDS‐ADRDA Work Group* under the auspices of Department of Health and Human Services Task Force on Alzheimer’s Disease. Neurology. 1984;34:939-.

    Article  CAS  PubMed  Google Scholar 

  40. Iulita MF, Do Carmo S, Ower AK, Fortress AM, Aguilar LF, Hanna M, et al. Nerve growth factor metabolic dysfunction in Down’s syndrome brains. Brain. 2014;137:860–72.

    Article  PubMed  PubMed Central  Google Scholar 

  41. Locke S, Yousefpour N, Mannarino M, Xing S, Yashmin F, Bourassa V, et al. Peripheral and central nervous system alterations in a rat model of inflammatory arthritis. Pain. 2020. https://doi.org/10.1097/j.pain.0000000000001837.

  42. Mirra SS, Heyman A, McKeel D, Sumi S, Crain BJ, Brownlee L, et al. The Consortium to establish a registry for Alzheimer’s Disease (CERAD): Part II. Standardization of the neuropathologic assessment of Alzheimer’s disease. Neurology. 1991;41:479.

    Article  CAS  PubMed  Google Scholar 

  43. Head E, Lott IT. Down syndrome and beta-amyloid deposition. Curr Opin Neurol. 2004;17:95–100.

    Article  CAS  PubMed  Google Scholar 

  44. Lott IT, Head E. Dementia in Down syndrome: unique insights for Alzheimer disease research. Nat Rev Neurol. 2019;15:135–47.

    Article  PubMed  PubMed Central  Google Scholar 

  45. Parks WC, Wilson CL, López-Boado YS. Matrix metalloproteinases as modulators of inflammation and innate immunity. Nat Rev Immunol. 2004;4:617.

    Article  CAS  PubMed  Google Scholar 

  46. Khokha R, Murthy A, Weiss A. Metalloproteinases and their natural inhibitors in inflammation and immunity. Nat Rev Immunol. 2013;13:649.

    Article  CAS  PubMed  Google Scholar 

  47. Nissinen L, Kähäri V-M. Matrix metalloproteinases in inflammation. Biochimica et Biophysica Acta (BBA)-Gen Subj. 2014;1840:2571–80.

    Article  CAS  Google Scholar 

  48. Heneka MT, Carson MJ, El Khoury J, Landreth GE, Brosseron F, Feinstein DL, et al. Neuroinflammation in Alzheimer’s disease. Lancet Neurol. 2015;14:388–405.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Rogers J. Principles for central nervous system inflammation research: a call for a consortium approach. Alzheimer’s Dement. 2018;14:1553–9.

    Article  Google Scholar 

  50. Cuello AC. Early and late CNS inflammation in Alzheimer’s disease: two extremes of a continuum? Trends Pharmacol Sciences. 2017;38:956–66.

    Article  CAS  Google Scholar 

  51. Iulita MF, Caraci F, Cuello AC. A link between nerve growth factor metabolic deregulation and amyloid-beta-driven inflammation in down syndrome. CNS Neurol Disorders Drug Targets. 2016;15:434–47.

  52. Iulita MF, Cuello AC. Nerve growth factor metabolic dysfunction in Alzheimer’s disease and down syndrome. Trends Pharmacol Sci. 2014;35:338–48.

    Article  CAS  PubMed  Google Scholar 

  53. Rosas‐Ballina M, Tracey K. Cholinergic control of inflammation. J Intern Med. 2009;265:663–79.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  54. Counts SE, Mufson EJ. The role of nerve growth factor receptors in cholinergic basal forebrain degeneration in prodromal Alzheimer disease. J Neuropathol Exp Neurol. 2005;64:263–72.

    Article  CAS  PubMed  Google Scholar 

  55. Mesulam M, Shaw P, Mash D, Weintraub S. Cholinergic nucleus basalis tauopathy emerges early in the aging‐MCI‐AD continuum. Ann Neurol: Off J Am Neurological Assoc Child Neurol Soc. 2004;55:815–28.

    Article  CAS  Google Scholar 

  56. Cooper JD, Salehi A, Delcroix J-D, Howe CL, Belichenko PV, Chua-Couzens J, et al. Failed retrograde transport of NGF in a mouse model of Down’s syndrome: reversal of cholinergic neurodegenerative phenotypes following NGF infusion. Proc Natl Acad Sci. 2001;98:10439–44.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. Salehi A, Delcroix J-D, Belichenko PV, Zhan K, Wu C, Valletta JS, et al. Increased App expression in a mouse model of Down’s syndrome disrupts NGF transport and causes cholinergic neuron degeneration. Neuron. 2006;51:29–42.

    Article  CAS  PubMed  Google Scholar 

  58. Venero J, Knüsel B, Beck K, Hefti F. Expression of neurotrophin and trk receptor genes in adult rats with fimbria transections: effect of intraventricular nerve growth factor and brain-derived neurotrophic factor administration. Neuroscience. 1994;59:797–815.

    Article  CAS  PubMed  Google Scholar 

  59. Figueiredo B, Skup M, Bedard A, Tetzlaff W, Cuello A. Differential expression of p140trk, p75NGFR and growth-associated phosphoprotein-43 genes in nucleus basalis magnocellularis, thalamus and adjacent cortex following neocortical infarction and nerve growth factor treatment. Neuroscience. 1995;68:29–45.

    Article  CAS  PubMed  Google Scholar 

  60. Gnahn H, Hefti F, Heumann R, Schwab M, Thoenen H. NGF-mediated increase of choline acetyltransferase (ChAT) in the neonatal rat forebrain: evidence for a physiological role of NGF in the brain? Devel Brain Res. 1983;9:45–52.

    Article  CAS  Google Scholar 

  61. Stephens P, Cuello A, Sofroniew M, Pearson R, Tagari P. Effect of unilateral decortication on choline acetyltransferase activity in the nucleus basalis and other areas of the rat brain. J Neurochem. 1985;45:1021–6.

    Article  CAS  PubMed  Google Scholar 

  62. Hartikka J, Hefti F. Development of septal cholinergic neurons in culture: plating density and glial cells modulate effects of NGF on survival, fiber growth, and expression of transmitter-specific enzymes. J Neurosci. 1988;8:2967–85.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  63. Pongrac JL, Rylett RJ. NGF-induction of the expression of ChAT mRNA in PC12 cells and primary cultures of embryonic rat basal forebrain. Mol Brain Res. 1998;62:25–34.

    Article  CAS  PubMed  Google Scholar 

  64. Berse B, Lopez-Coviella I, Blusztajn JK. Activation of TrkA by nerve growth factor upregulates expression of the cholinergic gene locus but attenuates the response to ciliary neurotrophic growth factor. Biochemical J. 1999;342:301–8.

    Article  CAS  Google Scholar 

  65. Madziar B, Lopez‐Coviella I, Zemelko V, Berse B. Regulation of cholinergic gene expression by nerve growth factor depends on the phosphatidylinositol‐3′‐kinase pathway. J Neurochem. 2005;92:767–79.

    Article  CAS  PubMed  Google Scholar 

  66. Villarin JM, McCurdy EP, Martínez JC, Hengst U. Local synthesis of dynein cofactors matches retrograde transport to acutely changing demands. Nat Commun. 2016;7:13865.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  67. Yoon BC, Jung H, Dwivedy A, O’Hare CM, Zivraj KH, Holt CE. Local translation of extranuclear lamin B promotes axon maintenance. Cell. 2012;148:752–64.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  68. Iulita MF, Ower A, Barone C, Pentz R, Gubert P, Romano C, et al. An inflammatory and trophic disconnect biomarker profile revealed in Down syndrome plasma: Relation to cognitive decline and longitudinal evaluation. Alzheimer’s Dementia. 2016;12:1132–48.

    Article  PubMed  Google Scholar 

  69. Iulita MF, Ganesh A, Pentz R, Flores Aguilar L, Gubert P, Ducatenzeiler A, et al. Identification and preliminary validation of a plasma profile associated with cognitive decline in dementia and at-risk individuals: a retrospective cohort analysis. J Alzheimer’s Dis. 2019;67:327–41.

    Article  CAS  Google Scholar 

  70. Hanzel CE, Iulita MF, Eyjolfsdottir H, Hjorth E, Schultzberg M, Eriksdotter M, et al. Analysis of matrix metallo-proteases and the plasminogen system in mild cognitive impairment and Alzheimer’s disease cerebrospinal fluid. J Alzheimer’s Dis. 2014;40:667–78.

    Article  CAS  Google Scholar 

  71. E Counts S, He B, G Prout J, Michalski B, Farotti L, Fahnestock M, et al. Cerebrospinal fluid proNGF: a putative biomarker for early Alzheimer’s disease. Curr Alzheimer Res. 2016;13:800–8.

    Article  CAS  Google Scholar 

  72. Blennow K, Zetterberg H. Biomarkers for Alzheimer’s disease: current status and prospects for the future. J Intern Med. 2018;284:643–63.

    Article  CAS  PubMed  Google Scholar 

  73. Everitt BJ, Robbins TW. Central cholinergic systems and cognition. Annu Rev Psychol. 1997;48:649–84.

    Article  CAS  PubMed  Google Scholar 

  74. Mesulam M. The cholinergic lesion of Alzheimer’s disease: pivotal factor or side show?. Learn Memory. 2004;11:43–9.

    Article  Google Scholar 

  75. Gilmor ML, Erickson JD, Varoqui H, Hersh LB, Bennett DA, Cochran EJ, et al. Preservation of nucleus basalis neurons containing choline acetyltransferase and the vesicular acetylcholine transporter in the elderly with mild cognitive impairment and early Alzheimer’s disease. J Comp Neurol. 1999;411:693–704.

    Article  CAS  PubMed  Google Scholar 

  76. DeKosky ST, Ikonomovic MD, Styren SD, Beckett L, Wisniewski S, Bennett DA, et al. Upregulation of choline acetyltransferase activity in hippocampus and frontal cortex of elderly subjects with mild cognitive impairment. Annals Neurol: Off J Am Neurological Assoc Child Neurol Soc. 2002;51:145–55.

    Article  CAS  Google Scholar 

  77. Cuello A, Bruno A, Bell K. NGF-cholinergic dependency in brain aging, MCI and Alzheimer’s disease. Curr Alzheimer Res. 2007;4:351–8.

    Article  CAS  PubMed  Google Scholar 

  78. Zissimopoulos J, Crimmins E, Clair PS, editors. The value of delaying Alzheimer’s disease onset. Forum for Health Economics and Policy; 2015: De Gruyter.

  79. Hampel H, Mesulam M-M, Cuello AC, Farlow MR, Giacobini E, Grossberg GT, et al. The cholinergic system in the pathophysiology and treatment of Alzheimer’s disease. Brain 2018;141:1917–33.

    Article  PubMed  PubMed Central  Google Scholar 

  80. Cavedo E, Dubois B, Colliot O, Lista S, Croisile B, Tisserand GL, et al. Reduced regional cortical thickness rate of change in donepezil-treated subjects with suspected prodromal Alzheimer’s disease. J Clin Psych. 2016;77:1631–8.

    Article  Google Scholar 

  81. Dubois B, Chupin M, Hampel H, Lista S, Cavedo E, Croisile B, et al. Donepezil decreases annual rate of hippocampal atrophy in suspected prodromal Alzheimer’s disease. Alzheimers Dement 2015;11:1041–9.

    Article  PubMed  Google Scholar 

  82. Fox C, Richardson K, Maidment ID, Savva GM, Matthews FE, Smithard D, et al. Anticholinergic medication use and cognitive impairment in the older population: the medical research council cognitive function and ageing study. J Am Geriatrics Soc. 2011;59:1477–83.

    Article  Google Scholar 

  83. Risacher SL, McDonald BC, Tallman EF, West JD, Farlow MR, Unverzagt FW, et al. Association between anticholinergic medication use and cognition, brain metabolism, and brain atrophy in cognitively normal older adults. JAMA Neurol. 2016;73:721–32.

    Article  PubMed  PubMed Central  Google Scholar 

  84. Campbell NL, Lane KA, Gao S, Boustani MA, Unverzagt F. Anticholinergics influence transition from normal cognition to mild cognitive impairment in older adults in primary care. Pharmacotherapy: J Hum Pharmacol Drug Ther. 2018;38:511–9.

    Article  CAS  Google Scholar 

  85. Richardson K, Fox C, Maidment I, Steel N, Loke YK, Arthur A, et al. Anticholinergic drugs and risk of dementia: case-control study. bmj 2018;361:k1315.

    Article  PubMed  PubMed Central  Google Scholar 

  86. Schmitz TW, Mur M, Aghourian M, Bedard MA, Spreng RN, Alzheimer’s Disease, Neuroimaging I. Longitudinal Alzheimer’s Degeneration Reflects the Spatial Topography of Cholinergic Basal Forebrain Projections. Cell Rep. 2018;24:38–46.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  87. Schmitz TW, Nathan Spreng R, Alzheimer’s Disease Neuroimaging I. Basal forebrain degeneration precedes and predicts the cortical spread of Alzheimer’s pathology. Nat Commun. 2016;7:13249.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  88. Nitsch RM, Slack BE, Wurtman RJ, Growdon JH. Release of Alzheimer amyloid precursor derivatives stimulated by activation of muscarinic acetylcholine receptors. Science. 1992;258:304–7.

    Article  CAS  PubMed  Google Scholar 

  89. Fisher A. Cholinergic modulation of amyloid precursor protein processing with emphasis on M1 muscarinic receptor: perspectives and challenges in treatment of Alzheimer’s disease. J Neurochemistry. 2012;120:22–33.

    Article  CAS  Google Scholar 

  90. Hall H, Iulita MF, Gubert P, Flores Aguilar L, Ducatenzeiler A, Fisher A, et al. AF710B, an M1/sigma-1 receptor agonist with long-lasting disease-modifying properties in a transgenic rat model of Alzheimer’s disease. Alzheimers Dement. 2018;14:811–23.

    Article  PubMed  Google Scholar 

Download references

Acknowledgements

The authors express gratitude to the participants in the ROS study as well as to the team at the RUSH Medical Center for performing the cognitive testing and neuropathological analyses. The authors thank Dr. Daniel Lawrence from the Michigan Center for Integrative Research in Critical Care, USA for graciously providing the anti-neuroserpin antibody used in this study. The authors are also grateful for the revisions and suggestions provided by Drs Ezio Giacobini, Harald Hampel, and Giancarlo Pepeu on this manuscript. ACC acknowledges financial support from the Canadian Institutes of Health Research (CIHR) and the Alzheimer Society of Canada. He holds the McGill University Charles E. Frosst/Merck Chair in Pharmacology and is a member of the Canadian Consortium of Neurodegeneration in Aging. ACC wishes to thank Merck Canada for their unrestricted support. DAB was supported by grants P30AG10161 and R01AG15819 from the NIA. RP was the recipient of a Student Fellowship from the McGill Integrated Program in Neuroscience and CIHR Doctoral Award. MFI acknowledges support from a Bourse Postdoctorale from the Fonds de Recherche du Quebec Santé (FRQS). The funding bodies had no role in the design of the study or in the collection, analysis, and interpretation of data or in writing the manuscript.

Author information

Authors and Affiliations

Authors

Contributions

RP, MFI, and ACC conceived and designed the study. RP measured the proteins and transcripts in brain with guidance from MFI, AD, and ACC. DAB provided brain tissue as well as neuropathological and cognitive data. RP generated and analyzed the data. RP, MFI, and ACC wrote the manuscript. All authors have read and revised the final version of the manuscript.

Corresponding author

Correspondence to A. Claudio Cuello.

Ethics declarations

Conflict of interest

The authors declare that they have no conflict of interest.

Additional information

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

Supplementary information

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Pentz, R., Iulita, M.F., Ducatenzeiler, A. et al. The human brain NGF metabolic pathway is impaired in the pre-clinical and clinical continuum of Alzheimers disease. Mol Psychiatry 26, 6023–6037 (2021). https://doi.org/10.1038/s41380-020-0797-2

Download citation

  • Received:

  • Revised:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41380-020-0797-2

This article is cited by

Search

Quick links