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.

  • Review Article
  • Published:

Selective vulnerability in neurodegenerative diseases

Abstract

Neurodegenerative diseases have two general characteristics that are so fundamental we usually take them for granted. The first is that the pathology associated with the disease only affects particular neurons (‘selective neuronal vulnerability’); the second is that the pathology worsens with time and impacts more regions in a stereotypical and predictable fashion. The mechanisms underpinning selective neuronal and regional vulnerability have been difficult to dissect, but the recent application of whole-genome technologies, the development of mouse models that reproduce spatial and temporal features of the pathology, and the identification of intrinsic morphological, electrophysiological, and biochemical properties of vulnerable neurons are beginning to shed some light on these fundamental features of neurodegenerative diseases. Here we detail our emerging understanding of the underlying biology of selective neuronal vulnerability and outline some of the areas in which our understanding is incomplete.

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: Regions and neurons that are vulnerable in neurodegenerative diseases.

Similar content being viewed by others

References

  1. Roberts, G. W., Nash, M., Ince, P. G., Royston, M. C. & Gentleman, S. M. On the origin of Alzheimer’s disease: a hypothesis. Neuroreport 4, 7–9 (1993).

    Article  CAS  PubMed  Google Scholar 

  2. Hyman, B. T., Van Hoesen, G. W., Damasio, A. R. & Barnes, C. L. Alzheimer’s disease: cell-specific pathology isolates the hippocampal formation. Science 225, 1168–1170 (1984).

    Article  CAS  PubMed  Google Scholar 

  3. Morrison, B. M., Hof, P. R. & Morrison, J. H. Determinants of neuronal vulnerability in neurodegenerative diseases. Ann. Neurol. 44 (Suppl 1), S32–S44 (1998).

    Article  CAS  PubMed  Google Scholar 

  4. Morrison, J. H. & Hof, P. R. Selective vulnerability of corticocortical and hippocampal circuits in aging and Alzheimer’s disease. Prog. Brain Res. 136, 467–486 (2002).

    Article  CAS  PubMed  Google Scholar 

  5. Stranahan, A. M. & Mattson, M. P. Selective vulnerability of neurons in layer II of the entorhinal cortex during aging and Alzheimer’s disease. Neural Plast. 2010, 108190, https://doi.org/10.1155/2010/108190 (2010).

    Article  CAS  Google Scholar 

  6. Whitehouse, P. J. et al. Alzheimer’s disease and senile dementia: loss of neurons in the basal forebrain. Science 215, 1237–1239 (1982).

    Article  CAS  PubMed  Google Scholar 

  7. Davies, P. & Maloney, A. J. Selective loss of central cholinergic neurons in Alzheimer’s disease. Lancet 2, 1403 (1976).

    Article  CAS  PubMed  Google Scholar 

  8. Bondareff, W., Mountjoy, C. Q. & Roth, M. Loss of neurons of origin of the adrenergic projection to cerebral cortex (nucleus locus coeruleus) in senile dementia. Neurology 32, 164–168 (1982).

    Article  CAS  PubMed  Google Scholar 

  9. Greenamyre, J. T. & Young, A. B. Excitatory amino acids and Alzheimer’s disease. Neurobiol. Aging 10, 593–602 (1989).

    Article  CAS  PubMed  Google Scholar 

  10. Chin, J. et al. Reelin depletion in the entorhinal cortex of human amyloid precursor protein transgenic mice and humans with Alzheimer’s disease. J. Neurosci. 27, 2727–2733 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Morrison, J. H. et al. A monoclonal antibody to non-phosphorylated neurofilament protein marks the vulnerable cortical neurons in Alzheimer’s disease. Brain Res. 416, 331–336 (1987).

    Article  CAS  PubMed  Google Scholar 

  12. Bussière, T. et al. Progressive degeneration of nonphosphorylated neurofilament protein-enriched pyramidal neurons predicts cognitive impairment in Alzheimer’s disease: stereologic analysis of prefrontal cortex area 9. J. Comp. Neurol. 463, 281–302 (2003).

    Article  PubMed  CAS  Google Scholar 

  13. Hof, P. R., Nimchinsky, E. A., Celio, M. R., Bouras, C. & Morrison, J. H. Calretinin-immunoreactive neocortical interneurons are unaffected in Alzheimer’s disease. Neurosci. Lett. 152, 145–148 (1993).

    Article  CAS  PubMed  Google Scholar 

  14. Iwamoto, N. & Emson, P. C. Demonstration of neurofibrillary tangles in parvalbumin-immunoreactive interneurones in the cerebral cortex of Alzheimer-type dementia brain. Neurosci. Lett. 128, 81–84 (1991).

    Article  CAS  PubMed  Google Scholar 

  15. Fu, H. et al. Tau pathology induces excitatory neuron loss, grid cell dysfunction, and spatial memory deficits reminiscent of early Alzheimer’s disease. Neuron 93, 533–541.e5 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Sarter, M. & Bruno, J. P. The neglected constituent of the basal forebrain corticopetal projection system: GABAergic projections. Eur. J. Neurosci. 15, 1867–1873 (2002).

    Article  PubMed  Google Scholar 

  17. Mattson, M. P. & Magnus, T. Ageing and neuronal vulnerability. Nat. Rev. Neurosci. 7, 278–294 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Muratore, C. R. et al. Cell-type dependent Alzheimer’s disease phenotypes: probing the biology of selective neuronal vulnerability. Stem Cell Rep. 9, 1868–1884 (2017).

    Article  CAS  Google Scholar 

  19. Brichta, L. & Greengard, P. Molecular determinants of selective dopaminergic vulnerability in Parkinson’s disease: an update. Front. Neuroanat. 8, 152 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  20. Surmeier, D. J., Obeso, J. A. & Halliday, G. M. Parkinson’s disease is not simply a prion disorder. J. Neurosci. 37, 9799–9807 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Double, K. L. Neuronal vulnerability in Parkinson’s disease. Parkinsonism Relat. Disord. 18 (Suppl 1), S52–S54 (2012).

    Article  PubMed  Google Scholar 

  22. Jellinger, K. A. Post mortem studies in Parkinson’s disease–is it possible to detect brain areas for specific symptoms? J. Neural Transm. Suppl. 56, 1–29 (1999).

    Article  CAS  PubMed  Google Scholar 

  23. Halliday, G. M. et al. Neuropathology of immunohistochemically identified brainstem neurons in Parkinson’s disease. Ann. Neurol. 27, 373–385 (1990).

    Article  CAS  PubMed  Google Scholar 

  24. Henderson, J. M., Carpenter, K., Cartwright, H. & Halliday, G. M. Degeneration of the centré median-parafascicular complex in Parkinson’s disease. Ann. Neurol. 47, 345–352 (2000).

    Article  CAS  PubMed  Google Scholar 

  25. Harding, A. J., Stimson, E., Henderson, J. M. & Halliday, G. M. Clinical correlates of selective pathology in the amygdala of patients with Parkinson’s disease. Brain 125, 2431–2445 (2002).

    Article  PubMed  Google Scholar 

  26. Kingsbury, A. E. et al. Brain stem pathology in Parkinson’s disease: an evaluation of the Braak staging model. Mov. Disord. 25, 2508–2515 (2010).

    Article  PubMed  Google Scholar 

  27. Braak, H. et al. Staging of brain pathology related to sporadic Parkinson’s disease. Neurobiol. Aging 24, 197–211 (2003).

    Article  PubMed  Google Scholar 

  28. Walker, D. G. et al. Changes in properties of serine 129 phosphorylated α-synuclein with progression of Lewy-type histopathology in human brains. Exp. Neurol. 240, 190–204 (2013).

    Article  CAS  PubMed  Google Scholar 

  29. Saxena, S. & Caroni, P. Selective neuronal vulnerability in neurodegenerative diseases: from stressor thresholds to degeneration. Neuron 71, 35–48 (2011).

    Article  CAS  PubMed  Google Scholar 

  30. Boillée, S., Vande Velde, C. & Cleveland, D. W. ALS: a disease of motor neurons and their nonneuronal neighbors. Neuron 52, 39–59 (2006).

    Article  PubMed  CAS  Google Scholar 

  31. Roselli, F. & Caroni, P. From intrinsic firing properties to selective neuronal vulnerability in neurodegenerative diseases. Neuron 85, 901–910 (2015).

    Article  CAS  PubMed  Google Scholar 

  32. Alexianu, M. E. et al. The role of calcium-binding proteins in selective motoneuron vulnerability in amyotrophic lateral sclerosis. Ann. Neurol. 36, 846–858 (1994).

    Article  CAS  PubMed  Google Scholar 

  33. Kihira, T., Yoshida, S., Yoshimasu, F., Wakayama, I. & Yase, Y. Involvement of Onuf’s nucleus in amyotrophic lateral sclerosis. J. Neurol. Sci. 147, 81–88 (1997).

    Article  CAS  PubMed  Google Scholar 

  34. von Lewinski, F. & Keller, B. U. Ca2+, mitochondria and selective motoneuron vulnerability: implications for ALS. Trends Neurosci. 28, 494–500 (2005).

    Article  CAS  Google Scholar 

  35. Mills, K. R. The natural history of central motor abnormalities in amyotrophic lateral sclerosis. Brain 126, 2558–2566 (2003).

    Article  CAS  PubMed  Google Scholar 

  36. Varrone, A. et al. Voxel-based comparison of rCBF SPET images in frontotemporal dementia and Alzheimer’s disease highlights the involvement of different cortical networks. Eur. J. Nucl. Med. Mol. Imaging 29, 1447–1454 (2002).

    Article  PubMed  Google Scholar 

  37. Rabinovici, G. D. et al. Distinct MRI atrophy patterns in autopsy-proven Alzheimer’s disease and frontotemporal lobar degeneration. Am. J. Alzheimers Dis. Other Demen. 22, 474–488 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Seeley, W. W. et al. Early frontotemporal dementia targets neurons unique to apes and humans. Ann. Neurol. 60, 660–667 (2006).

    Article  PubMed  Google Scholar 

  39. Seeley, W. W. Selective functional, regional, and neuronal vulnerability in frontotemporal dementia. Curr. Opin. Neurol. 21, 701–707 (2008).

    Article  PubMed  PubMed Central  Google Scholar 

  40. Dickson, D. W., Kouri, N., Murray, M. E. & Josephs, K. A. Neuropathology of frontotemporal lobar degeneration-tau (FTLD-tau). J. Mol. Neurosci. 45, 384–389 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Galvan, L., André, V. M., Wang, E. A., Cepeda, C. & Levine, M. S. Functional differences between direct and indirect striatal output pathways in Huntington’s disease. J. Huntingtons Dis. 1, 17–25 (2012).

    Article  PubMed  PubMed Central  Google Scholar 

  42. Morigaki, R. & Goto, S. Striatal vulnerability in Huntington’s disease: neuroprotection versus neurotoxicity. Brain Sci. 7, E63 (2017).

    Article  PubMed  CAS  Google Scholar 

  43. Reiner, A. et al. Differential loss of striatal projection neurons in Huntington disease. Proc. Natl. Acad. Sci. USA 85, 5733–5737 (1988).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Richfield, E. K., Maguire-Zeiss, K. A., Vonkeman, H. E. & Voorn, P. Preferential loss of preproenkephalin versus preprotachykinin neurons from the striatum of Huntington’s disease patients. Ann. Neurol. 38, 852–861 (1995).

    Article  CAS  PubMed  Google Scholar 

  45. Braak, H. & Braak, E. Neuropathological stageing of Alzheimer-related changes. Acta Neuropathol. 82, 239–259 (1991).

    Article  CAS  PubMed  Google Scholar 

  46. Brettschneider, J. et al. Stages of pTDP-43 pathology in amyotrophic lateral sclerosis. Ann. Neurol. 74, 20–38 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Josephs, K. A. et al. Staging TDP-43 pathology in Alzheimer’s disease. Acta Neuropathol. 127, 441–450 (2014).

    Article  CAS  PubMed  Google Scholar 

  48. Braak, H. & Del Tredici, K. Neuropathological staging of brain pathology in sporadic Parkinson’s disease: separating the wheat from the chaff. J. Parkinsons Dis. 7 s1, S71–S85 (2017).

    Article  Google Scholar 

  49. Braak, H. et al. Amyotrophic lateral sclerosis–a model of corticofugal axonal spread. Nat. Rev. Neurol. 9, 708–714 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Walker, L. C. & Jucker, M. Neurodegenerative diseases: expanding the prion concept. Annu. Rev. Neurosci. 38, 87–103 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Brettschneider, J., Del Tredici, K., Lee, V. M. & Trojanowski, J. Q. Spreading of pathology in neurodegenerative diseases: a focus on human studies. Nat. Rev. Neurosci. 16, 109–120 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Goedert, M., Eisenberg, D. S. & Crowther, R. A. Propagation of tau aggregates and neurodegeneration. Annu. Rev. Neurosci. 40, 189–210 (2017).

    Article  CAS  PubMed  Google Scholar 

  53. Mudher, A. et al. What is the evidence that tau pathology spreads through prion-like propagation? Acta Neuropathol. Commun 5, 99 (2017).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  54. Jucker, M. & Walker, L. C. Self-propagation of pathogenic protein aggregates in neurodegenerative diseases. Nature 501, 45–51 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. Spina, S. et al. The tauopathy associated with mutation +3 in intron 10 of Tau: characterization of the MSTD family. Brain 131, 72–89 (2008).

    Article  PubMed  Google Scholar 

  56. Ghetti, B. et al. Invited review: Frontotemporal dementia caused by microtubule-associated protein tau gene (MAPT) mutations: a chameleon for neuropathology and neuroimaging. Neuropathol. Appl. Neurobiol. 41, 24–46 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. Walker, L. C. Proteopathic strains and the heterogeneity of neurodegenerative diseases. Annu. Rev. Genet. 50, 329–346 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  58. Schmidt, M. et al. Peptide dimer structure in an Aβ(1-42) fibril visualized with cryo-EM. Proc. Natl. Acad. Sci. USA 112, 11858–11863 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  59. Gremer, L. et al. Fibril structure of amyloid-β(1-42) by cryo-electron microscopy. Science 358, 116–119 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  60. Fitzpatrick, A. W. P. et al. Cryo-EM structures of tau filaments from Alzheimer’s disease. Nature 547, 185–190 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  61. Langfelder, P. & Horvath, S. WGCNA: an R package for weighted correlation network analysis. BMC Bioinformatics 9, 559 (2008).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  62. Gamazon, E. R. et al. A gene-based association method for mapping traits using reference transcriptome data. Nat. Genet. 47, 1091–1098 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  63. Matarin, M. et al. A genome-wide gene-expression analysis and database in transgenic mice during development of amyloid or tau pathology. Cell Rep. 10, 633–644 (2015).

    Article  CAS  PubMed  Google Scholar 

  64. Ryan, B. J., Hoek, S., Fon, E. A. & Wade-Martins, R. Mitochondrial dysfunction and mitophagy in Parkinson’s: from familial to sporadic disease. Trends Biochem. Sci. 40, 200–210 (2015).

    Article  CAS  PubMed  Google Scholar 

  65. Hardy, J. Catastrophic cliffs: a partial suggestion for selective vulnerability in neurodegenerative diseases. Biochem. Soc. Trans. 44, 659–661 (2016).

    Article  CAS  PubMed  Google Scholar 

  66. Ciryam, P., Tartaglia, G. G., Morimoto, R. I., Dobson, C. M. & Vendruscolo, M. Widespread aggregation and neurodegenerative diseases are associated with supersaturated proteins. Cell Rep. 5, 781–790 (2013).

    Article  CAS  PubMed  Google Scholar 

  67. Ciryam, P., Kundra, R., Morimoto, R. I., Dobson, C. M. & Vendruscolo, M. Supersaturation is a major driving force for protein aggregation in neurodegenerative diseases. Trends Pharmacol. Sci. 36, 72–77 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  68. Ciryam, P. et al. A transcriptional signature of Alzheimer’s disease is associated with a metastable subproteome at risk for aggregation. Proc. Natl. Acad. Sci. USA 113, 4753–4758 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  69. Ciryam, P. et al. Spinal motor neuron protein supersaturation patterns are associated with inclusion body formation in ALS. Proc. Natl. Acad. Sci. USA 114, E3935–E3943 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  70. Freer, R. et al. A protein homeostasis signature in healthy brains recapitulates tissue vulnerability to Alzheimer’s disease. Sci. Adv. 2, e1600947 (2016).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  71. Hardy, J. Expression of normal sequence pathogenic proteins for neurodegenerative disease contributes to disease risk: ‘permissive templating’ as a general mechanism underlying neurodegeneration. Biochem. Soc. Trans. 33, 578–581 (2005).

    Article  CAS  PubMed  Google Scholar 

  72. Tiernan, C. T. et al. Protein homeostasis gene dysregulation in pretangle-bearing nucleus basalis neurons during the progression of Alzheimer’s disease. Neurobiol. Aging 42, 80–90 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  73. Johnson, N. R. et al. Evidence for sortilin modulating regional accumulation of human tau prions in transgenic mice. Proc. Natl. Acad. Sci. USA 114, E11029–E11036 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  74. Kundra, R., Ciryam, P., Morimoto, R. I., Dobson, C. M. & Vendruscolo, M. Protein homeostasis of a metastable subproteome associated with Alzheimer’s disease. Proc. Natl. Acad. Sci. USA 114, E5703–E5711 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  75. Tsvetkov, A. S. et al. Proteostasis of polyglutamine varies among neurons and predicts neurodegeneration. Nat. Chem. Biol. 9, 586–592 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  76. Guedes-Dias, P. et al. Mitochondrial dynamics and quality control in Huntington’s disease. Neurobiol. Dis. 90, 51–57 (2016).

    Article  CAS  PubMed  Google Scholar 

  77. Le Grand, J. N. et al. Specific distribution of the autophagic protein GABARAPL1/GEC1 in the developing and adult mouse brain and identification of neuronal populations expressing GABARAPL1/GEC1. PLoS One 8, e63133 (2013).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  78. Fimia, G. M. et al. Ambra1 regulates autophagy and development of the nervous system. Nature 447, 1121–1125 (2007).

    Article  CAS  PubMed  Google Scholar 

  79. Chung, Y. H. et al. Decreased expression of calretinin in the cerebral cortex and hippocampus of SOD1G93A transgenic mice. Brain Res. 1035, 105–109 (2005).

    Article  CAS  PubMed  Google Scholar 

  80. Greene, J. G., Dingledine, R. & Greenamyre, J. T. Gene expression profiling of rat midbrain dopamine neurons: implications for selective vulnerability in Parkinsonism. Neurobiol. Dis. 18, 19–31 (2005).

    Article  CAS  PubMed  Google Scholar 

  81. Mendez, I. et al. Cell type analysis of functional fetal dopamine cell suspension transplants in the striatum and substantia nigra of patients with Parkinson’s disease. Brain 128, 1498–1510 (2005).

    Article  PubMed  Google Scholar 

  82. Shaw, P. J. & Eggett, C. J. Molecular factors underlying selective vulnerability of motor neurons to neurodegeneration in amyotrophic lateral sclerosis. J. Neurol. 247 (Suppl 1), I17–I27 (2000).

    Article  PubMed  Google Scholar 

  83. Simonian, N. A. & Hyman, B. T. Functional alterations in neural circuits in Alzheimer’s disease. Neurobiol. Aging 16, 305–309 (1995).

    Article  CAS  PubMed  Google Scholar 

  84. Wang, Y. & Mattson, M. P. L-type Ca2+ currents at CA1 synapses, but not CA3 or dentate granule neuron synapses, are increased in 3xTgAD micein an age-dependent manner. Neurobiol. Aging 35, 88–95 (2014).

    Article  PubMed  CAS  Google Scholar 

  85. Sulzer, D. Multiple hit hypotheses for dopamine neuron loss in Parkinson’s disease. Trends Neurosci. 30, 244–250 (2007).

    Article  CAS  PubMed  Google Scholar 

  86. Kanning, K. C., Kaplan, A. & Henderson, C. E. Motor neuron diversity in development and disease. Annu. Rev. Neurosci. 33, 409–440 (2010).

    Article  CAS  PubMed  Google Scholar 

  87. Lin, M. T. & Beal, M. F. Mitochondrial dysfunction and oxidative stress in neurodegenerative diseases. Nature 443, 787–795 (2006).

    Article  CAS  PubMed  Google Scholar 

  88. Chan, C. S., Gertler, T. S. & Surmeier, D. J. Calcium homeostasis, selective vulnerability and Parkinson’s disease. Trends Neurosci. 32, 249–256 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  89. Hirsch, E. C., Jenner, P. & Przedborski, S. Pathogenesis of Parkinson’s disease. Mov. Disord. 28, 24–30 (2013).

    Article  CAS  PubMed  Google Scholar 

  90. Mattsson, N., Schott, J. M., Hardy, J., Turner, M. R. & Zetterberg, H. Selective vulnerability in neurodegeneration: insights from clinical variants of Alzheimer’s disease. J. Neurol. Neurosurg. Psychiatry 87, 1000–1004 (2016).

    Article  PubMed  Google Scholar 

  91. Surmeier, D. J., Guzman, J. N. & Sanchez-Padilla, J. Calcium, cellular aging, and selective neuronal vulnerability in Parkinson’s disease. Cell Calcium 47, 175–182 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  92. Bolam, J. P. & Pissadaki, E. K. Living on the edge with too many mouths to feed: why dopamine neurons die. Mov. Disord. 27, 1478–1483 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  93. Bender, A. et al. High levels of mitochondrial DNA deletions in substantia nigra neurons in aging and Parkinson disease. Nat. Genet. 38, 515–517 (2006).

    Article  CAS  PubMed  Google Scholar 

  94. Bender, A. et al. Dopaminergic midbrain neurons are the prime target for mitochondrial DNA deletions. J. Neurol. 255, 1231–1235 (2008).

    Article  PubMed  Google Scholar 

  95. Kraytsberg, Y. et al. Mitochondrial DNA deletions are abundant and cause functional impairment in aged human substantia nigra neurons. Nat. Genet. 38, 518–520 (2006).

    Article  CAS  PubMed  Google Scholar 

  96. Schapira, A. H. et al. The Royal Kings and Queens Parkinson’s Disease Research Group. Mitochondrial function in Parkinson’s disease. Ann. Neurol. 32 (Suppl), S116–S124 (1992).

    Article  CAS  PubMed  Google Scholar 

  97. Braak, H. & Braak, E. Development of Alzheimer-related neurofibrillary changes in the neocortex inversely recapitulates cortical myelogenesis. Acta Neuropathol. 92, 197–201 (1996).

    Article  CAS  PubMed  Google Scholar 

  98. Richter, C., Park, J. W. & Ames, B. N. Normal oxidative damage to mitochondrial and nuclear DNA is extensive. Proc. Natl. Acad. Sci. USA 85, 6465–6467 (1988).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  99. Choi, D. W. Ionic dependence of glutamate neurotoxicity. J. Neurosci. 7, 369–379 (1987).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  100. Morrison, J. H. Differential vulnerability, connectivity, and cell typology. Neurobiol. Aging 14, 51–54 (1993). discussion 55–56.

    Article  CAS  PubMed  Google Scholar 

  101. Magnusson, K. R., Brim, B. L. & Das, S. R. Selective vulnerabilities of N-methyl-D-aspartate (NMDA) receptors during brain aging. Front. Aging Neurosci. 2, 11 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  102. Mishizen-Eberz, A. J. et al. Biochemical and molecular studies of NMDA receptor subunits NR1/2A/2B in hippocampal subregions throughout progression of Alzheimer’s disease pathology. Neurobiol. Dis. 15, 80–92 (2004).

    Article  CAS  PubMed  Google Scholar 

  103. Lee, H. G. et al. Aberrant expression of metabotropic glutamate receptor 2 in the vulnerable neurons of Alzheimer’s disease. Acta Neuropathol. 107, 365–371 (2004).

    Article  CAS  PubMed  Google Scholar 

  104. Laslo, P., Lipski, J. & Funk, G. D. Differential expression of Group I metabotropic glutamate receptors in motoneurons at low and high risk for degeneration in ALS. Neuroreport 12, 1903–1908 (2001).

    Article  CAS  PubMed  Google Scholar 

  105. Tomiyama, M. et al. Expression of metabotropic glutamate receptor mRNAs in the human spinal cord: implications for selective vulnerability of spinal motor neurons in amyotrophic lateral sclerosis. J. Neurol. Sci. 189, 65–69 (2001).

    Article  CAS  PubMed  Google Scholar 

  106. Götz, J., Schonrock, N., Vissel, B. & Ittner, L. M. Alzheimer’s disease selective vulnerability and modeling in transgenic mice. J. Alzheimers Dis. 18, 243–251 (2009).

    Article  PubMed  Google Scholar 

  107. Lorenzo, L. E., Barbe, A., Portalier, P., Fritschy, J. M. & Bras, H. Differential expression of GABAA and glycine receptors in ALS-resistant vs. ALS-vulnerable motoneurons: possible implications for selective vulnerability of motoneurons. Eur. J. Neurosci 23, 3161–3170 (2006).

    Article  PubMed  Google Scholar 

  108. Nitrini, R. Selective vulnerability of von Economo neurons in frontotemporal dementia. Dement. Neuropsychol. 2, 164 (2008).

    PubMed  PubMed Central  Google Scholar 

  109. Seeley, W. W. et al. Divergent social functioning in behavioral variant frontotemporal dementia and Alzheimer disease: reciprocal networks and neuronal evolution. Alzheimer Dis. Assoc. Disord. 21, S50–S57 (2007).

    Article  PubMed  Google Scholar 

  110. Zeron, M. M. et al. Increased sensitivity to N-methyl-D-aspartate receptor-mediated excitotoxicity in a mouse model of Huntington’s disease. Neuron 33, 849–860 (2002).

    Article  CAS  PubMed  Google Scholar 

  111. Landwehrmeyer, G. B., Standaert, D. G., Testa, C. M., Penney, J. B. Jr. & Young, A. B. NMDA receptor subunit mRNA expression by projection neurons and interneurons in rat striatum. J. Neurosci. 15, 5297–5307 (1995).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  112. Küppenbender, K. D. et al. Expression of NMDA receptor subunit mRNAs in neurochemically identified projection and interneurons in the human striatum. J. Comp. Neurol. 419, 407–421 (2000).

    Article  PubMed  Google Scholar 

  113. Han, I., You, Y., Kordower, J. H., Brady, S. T. & Morfini, G. A. Differential vulnerability of neurons in Huntington’s disease: the role of cell type-specific features. J. Neurochem. 113, 1073–1091 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  114. Okamoto, S. et al. Balance between synaptic versus extrasynaptic NMDA receptor activity influences inclusions and neurotoxicity of mutant huntingtin. Nat. Med. 15, 1407–1413 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  115. Milnerwood, A. J. et al. Early increase in extrasynaptic NMDA receptor signaling and expression contributes to phenotype onset in Huntington’s disease mice. Neuron 65, 178–190 (2010).

    Article  CAS  PubMed  Google Scholar 

  116. Ginsberg, S. D., Che, S., Counts, S. E. & Mufson, E. J. Single cell gene expression profiling in Alzheimer’s disease. NeuroRx 3, 302–318 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  117. Liang, W. S. et al. Altered neuronal gene expression in brain regions differentially affected by Alzheimer’s disease: a reference data set. Physiol. Genomics 33, 240–256 (2008).

    Article  CAS  PubMed  Google Scholar 

  118. Taylor, R. C. & Dillin, A. Aging as an event of proteostasis collapse. Cold Spring Harb. Perspect. Biol. 3, a004440 (2011).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  119. Labbadia, J. & Morimoto, R. I. The biology of proteostasis in aging and disease. Annu. Rev. Biochem. 84, 435–464 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  120. Kaushik, S. & Cuervo, A. M. Proteostasis and aging. Nat. Med 21, 1406–1415 (2015).

    Article  CAS  PubMed  Google Scholar 

  121. Wojda, U., Salinska, E. & Kuznicki, J. Calcium ions in neuronal degeneration. IUBMB Life 60, 575–590 (2008).

    Article  CAS  PubMed  Google Scholar 

  122. Squier, T. C. Oxidative stress and protein aggregation during biological aging. Exp. Gerontol. 36, 1539–1550 (2001).

    Article  CAS  PubMed  Google Scholar 

  123. Soreq, L. et al. Major shifts in glial regional identity are a transcriptional hallmark of human brain aging. Cell Rep. 18, 557–570 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  124. Sun, S. et al. Translational profiling identifies a cascade of damage initiated in motor neurons and spreading to glia in mutant SOD1-mediated ALS. Proc. Natl. Acad. Sci. USA 112, E6993–E7002 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  125. Ginsberg, S.D. et al. Selective decline of neurotrophin and neurotrophin receptor genes within CA1 pyramidal neurons and hippocampus proper: correlation with cognitive performance and neuropathology in mild cognitive impairment and Alzheimer’s disease. Hippocampus (2017).

  126. Fombonne, J., Rabizadeh, S., Banwait, S., Mehlen, P. & Bredesen, D. E. Selective vulnerability in Alzheimer’s disease: amyloid precursor protein and p75(NTR) interaction. Ann. Neurol. 65, 294–303 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  127. Perez, S. E. et al. Rac1b increases with progressive tau pathology within cholinergic nucleus basalis neurons in Alzheimer’s disease. Am. J. Pathol. 180, 526–540 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  128. Gerschütz, A. et al. Neuron-specific alterations in signal transduction pathways associated with Alzheimer’s disease. J. Alzheimers Dis. 40, 135–142 (2014).

    Article  PubMed  CAS  Google Scholar 

  129. Shu, S. et al. Selective degeneration of entorhinal-CA1 synapses in Alzheimer’s disease via activation of DAPK1. J. Neurosci. 36, 10843–10852 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  130. Di Giovannantonio, L. G. et al. Otx2 selectively controls the neurogenesis of specific neuronal subtypes of the ventral tegmental area and compensates En1-dependent neuronal loss and MPTP vulnerability. Dev. Biol. 373, 176–183 (2013).

    Article  PubMed  CAS  Google Scholar 

  131. Oliveira, M. A. P., Balling, R., Smidt, M. P. & Fleming, R. M. T. Embryonic development of selectively vulnerable neurons in Parkinson’s disease. NPJ Parkinsons Dis. 3, 21 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  132. Osborne, P. B., Halliday, G. M., Cooper, H. M. & Keast, J. R. Localization of immunoreactivity for deleted in colorectal cancer (DCC), the receptor for the guidance factor netrin-1, in ventral tier dopamine projection pathways in adult rodents. Neuroscience 131, 671–681 (2005).

    Article  CAS  PubMed  Google Scholar 

  133. Nichterwitz, S. et al. Laser capture microscopy coupled with Smart-seq2 for precise spatial transcriptomic profiling. Nat. Commun. 7, 12139 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  134. McKeever, P. M. et al. Cholinergic neuron gene expression differences captured by translational profiling in a mouse model of Alzheimer’s disease. Neurobiol. Aging 57, 104–119 (2017).

    Article  CAS  PubMed  Google Scholar 

  135. Keren-Shaul, H. et al. A unique microglia type associated with restricting development of Alzheimer’s disease. Cell 169, 1276–1290.e17 (2017).

    Article  CAS  PubMed  Google Scholar 

  136. Gosselin, D. et al. An environment-dependent transcriptional network specifies human microglia identity. Science 356, eaal3222 (2017).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  137. Lake, B. B. et al. Neuronal subtypes and diversity revealed by single-nucleus RNA sequencing of the human brain. Science 352, 1586–1590 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  138. Habib, N. et al. Massively parallel single-nucleus RNA-seq with DroNc-seq. Nat. Methods 14, 955–958 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  139. Lee, J. H. et al. Highly multiplexed subcellular RNA sequencing in situ. Science 343, 1360–1363 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  140. Silva, M. C. et al. Human iPSC-derived neuronal model of tau-A152T frontotemporal dementia reveals tau-mediated mechanisms of neuronal vulnerability. Stem Cell Rep. 7, 325–340 (2016).

    Article  CAS  Google Scholar 

  141. Wang, C. et al. Scalable production of iPSC-derived human neurons to identify tau-lowering compounds by high-content screening. Stem Cell Rep. 9, 1221–1233 (2017).

    Article  CAS  Google Scholar 

  142. Imamura, K. et al. Calcium dysregulation contributes to neurodegeneration in FTLD patient iPSC-derived neurons. Sci. Rep. 6, 34904 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  143. Swistowski, A. et al. Efficient generation of functional dopaminergic neurons from human induced pluripotent stem cells under defined conditions. Stem Cells 28, 1893–1904 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  144. Roessler, R., Boddeke, E. & Copray, S. Induced pluripotent stem cell technology and direct conversion: new possibilities to study and treat Parkinson’s disease. Stem Cell Rev. 9, 505–513 (2013).

    Article  CAS  Google Scholar 

  145. Sundberg, M. et al. Improved cell therapy protocols for Parkinson’s disease based on differentiation efficiency and safety of hESC-, hiPSC-, and non-human primate iPSC-derived dopaminergic neurons. Stem Cells 31, 1548–1562 (2013).

    Article  CAS  PubMed  Google Scholar 

  146. Zhang, Y. et al. Rapid single-step induction of functional neurons from human pluripotent stem cells. Neuron 78, 785–798 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  147. Sun, A. X. et al. Direct induction and functional maturation of forebrain GABAergic neurons from human pluripotent stem cells. Cell Rep. 16, 1942–1953 (2016).

    Article  CAS  PubMed  Google Scholar 

  148. Yang, N. et al. Generation of pure GABAergic neurons by transcription factor programming. Nat. Methods 14, 621–628 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  149. Miller, J. D. et al. Human iPSC-based modeling of late-onset disease via progerin-induced aging. Cell Stem Cell 13, 691–705 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  150. Victor, M.B. et al. Striatal neurons directly converted from Huntington’s disease patient fibroblasts recapitulate age-associated disease phenotypes. Nat Neurosci. 21, 341–352 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

The authors thank D. Dickson for critical reading of the manuscript. K.E.D. acknowledges the support of NIH (NS074874 and AG056151), Cure Alzheimer’s Fund, the Tau Consortium, and the Brightfocus Foundation. This work was also supported by a grant from the NIH (AG056673) and the Alzheimer’s Association (AARF-17-505009) to H.J.F.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Karen E. Duff.

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.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Fu, H., Hardy, J. & Duff, K.E. Selective vulnerability in neurodegenerative diseases. Nat Neurosci 21, 1350–1358 (2018). https://doi.org/10.1038/s41593-018-0221-2

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41593-018-0221-2

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