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.

  • Perspective
  • Published:

Connectome-based modelling of neurodegenerative diseases: towards precision medicine and mechanistic insight

Abstract

Neurodegenerative diseases are the most common cause of dementia. Although their underlying molecular pathologies have been identified, there is substantial heterogeneity in the patterns of progressive brain alterations across and within these diseases. Recent advances in neuroimaging methods have revealed that pathological proteins accumulate along specific macroscale brain networks, implicating the network architecture of the brain in the system-level pathophysiology of neurodegenerative diseases. However, the extent to which ‘network-based neurodegeneration’ applies across the wide range of neurodegenerative disorders remains unclear. Here, we discuss the state-of-the-art of neuroimaging-based connectomics for the mapping and prediction of neurodegenerative processes. We review findings supporting brain networks as passive conduits through which pathological proteins spread. As an alternative view, we also discuss complementary work suggesting that network alterations actively modulate the spreading of pathological proteins between connected brain regions. We conclude this Perspective by proposing an integrative framework in which connectome-based models can be advanced along three dimensions of innovation: incorporating parameters that modulate propagation behaviour on the basis of measurable biological features; building patient-tailored models that use individual-level information and allowing model parameters to interact dynamically over time. We discuss promises and pitfalls of these strategies for improving disease insights and moving towards precision medicine.

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: Clinico-pathological spectrum of neurodegenerative dementia syndromes.
Fig. 2: Common methods for assessing the influence of network architecture on disease progression.
Fig. 3: Brain networks as conduits and drivers of disease progression.
Fig. 4: The next generation of connectome-based models can innovate along three dimensions.

Similar content being viewed by others

References

  1. Elahi, F. M. & Miller, B. L. A clinicopathological approach to the diagnosis of dementia. Nat. Rev. Neurol. 13, 457–476 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  2. Graff-Radford, J. et al. New insights into atypical Alzheimer’s disease in the era of biomarkers. Lancet Neurol. 20, 222–234 (2021).

    Article  PubMed  PubMed Central  Google Scholar 

  3. Ossenkoppele, R. et al. The behavioural/dysexecutive variant of Alzheimer’s disease: clinical, neuroimaging and pathological features. Brain 138, 2732–2749 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  4. Warren, J. D., Fletcher, P. D. & Golden, H. L. The paradox of syndromic diversity in Alzheimer disease. Nat. Rev. Neurol. 8, 451–464 (2012).

    Article  CAS  PubMed  Google Scholar 

  5. Beach, T. G., Monsell, S. E., Phillips, L. E. & Kukull, W. Accuracy of the clinical diagnosis of Alzheimer disease at National Institute on Aging Alzheimer Disease Centers, 2005–2010. J. Neuropathol. Exp. Neurol. 71, 266–273 (2012).

    Article  PubMed  Google Scholar 

  6. Seeley, W. W. Mapping neurodegenerative disease onset and progression. Cold Spring Harb. Perspect. Biol. 9, a023622 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  7. Pearson, R. C., Esiri, M. M., Hiorns, R. W., Wilcock, G. K. & Powell, T. P. Anatomical correlates of the distribution of the pathological changes in the neocortex in Alzheimer disease. Proc. Natl Acad. Sci. USA 82, 4531–4534 (1985).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Saper, C. B., Wainer, B. H. & German, D. C. Axonal and transneuronal transport in the transmission of neurological disease: potential role in system degenerations, including Alzheimer’s disease. Neuroscience 23, 389–398 (1987).

    Article  CAS  PubMed  Google Scholar 

  9. Braak, H. & Braak, E. Neuropathological staging of Alzheimer-related changes. Acta Neuropathologica 82, 239–259 (1991).

    Article  CAS  PubMed  Google Scholar 

  10. Jucker, M. & Walker, L. C. Propagation and spread of pathogenic protein assemblies in neurodegenerative diseases. Nat. Neurosci. 21, 1341–1349 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. 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 

  12. Prusiner, S. B. Prions. Proc. Natl Acad. Sci. USA 95, 13363–13383 (1998).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Seeley, W. W., Crawford, R. K., Zhou, J., Miller, B. L. & Greicius, M. D. Neurodegenerative diseases target large-scale human brain networks. Neuron 62, 42–52 (2009). This landmark study maps dementia syndromes onto the degeneration of syndrome-specific functional networks.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Zhou, J., Gennatas, E. D., Kramer, J. H., Miller, B. L. & Seeley, W. W. Predicting regional neurodegeneration from the healthy brain functional connectome. Neuron 73, 1216–1227 (2012). This important paper was among the first to empirically model brain networks as constraints for brain atrophy across various neurodegenerative diseases.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Calafate, S. et al. Synaptic contacts enhance cell-to-cell tau pathology propagation. Cell Rep. 11, 1176–1183 (2015).

    Article  CAS  PubMed  Google Scholar 

  16. de Calignon, A. et al. Propagation of tau pathology in a model of early Alzheimer’s disease. Neuron 73, 685–697 (2012).

    Article  PubMed  PubMed Central  Google Scholar 

  17. Bero, A. W. et al. Neuronal activity regulates the regional vulnerability to amyloid-β deposition. Nat. Neurosci. 14, 750–756 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Busche, M. A. & Hyman, B. T. Synergy between amyloid-β and tau in Alzheimer’s disease. Nat. Neurosci. 23, 1183–1193 (2020).

    Article  CAS  PubMed  Google Scholar 

  19. Busche, M. A. et al. Tau impairs neural circuits, dominating amyloid-β effects, in Alzheimer models in vivo. Nat. Neurosci. 22, 57–64 (2019). This paper investigates the mutual interplay between pathological proteins and alterations in neuronal activity, and vice versa, reinvigorating the idea that networks may have an active role in disease spread.

    Article  CAS  PubMed  Google Scholar 

  20. Busche, M. A. et al. Critical role of soluble amyloid-β for early hippocampal hyperactivity in a mouse model of Alzheimer’s disease. Proc. Natl Acad. Sci. USA 109, 8740–8745 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Wu, J. W. et al. Neuronal activity enhances tau propagation and tau pathology in vivo. Nat. Neurosci. 19, 1085–1092 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Grothe, M. J. & Teipel, S. J. & Alzheimer’s Disease Neuroimaging Initiative. Spatial patterns of atrophy, hypometabolism, and amyloid deposition in Alzheimer’s disease correspond to dissociable functional brain networks. Hum. Brain Mapp. 37, 35–53 (2016).

    Article  PubMed  Google Scholar 

  23. Pereira, J. B. et al. Amyloid and tau accumulate across distinct spatial networks and are differentially associated with brain connectivity. eLife 8, e50830 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Buckner, R. L. et al. Cortical hubs revealed by intrinsic functional connectivity: mapping, assessment of stability, and relation to Alzheimer’s disease. J. Neurosci. 29, 1860–1873 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Buckner, R. L. et al. Molecular, structural, and functional characterization of Alzheimer’s disease: evidence for a relationship between default activity, amyloid, and memory. J. Neurosci. 25, 7709–7717 (2005). This seminal paper explores properties linking brain functional organization and patterns of β-amyloid deposition in Alzheimer disease.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Palmqvist, S. et al. Earliest accumulation of β-amyloid occurs within the default-mode network and concurrently affects brain connectivity. Nat. Commun. 8, 1214 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  27. Villeneuve, S. et al. Existing Pittsburgh compound-B positron emission tomography thresholds are too high: statistical and pathological evaluation. Brain 138, 2020–2033 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  28. Hoenig, M. C. et al. Networks of tau distribution in Alzheimer’s disease. Brain 141, 568–581 (2018).

    Article  PubMed  Google Scholar 

  29. Jones, D. T. et al. Tau, amyloid, and cascading network failure across the Alzheimer’s disease spectrum. Cortex 97, 143–159 (2017). This paper directly investigated thecascading network failurehypothesis by examining the interplay between accumulation of pathological proteins and brain network activity in vivo.

    Article  PubMed  PubMed Central  Google Scholar 

  30. Hansson, O. et al. Tau pathology distribution in Alzheimer’s disease corresponds differentially to cognition-relevant functional brain networks. Front. Neurosci. 11, 167 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  31. Bejanin, A. et al. Tau pathology and neurodegeneration contribute to cognitive impairment in Alzheimer’s disease. Brain 140, 3286–3300 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  32. Ossenkoppele, R. et al. Tau PET patterns mirror clinical and neuroanatomical variability in Alzheimer’s disease. Brain 139, 1551–1567 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  33. Schöll, M. et al. Distinct 18F-AV-1451 tau PET retention patterns in early- and late-onset Alzheimer’s disease. Brain 140, 2286–2294 (2017).

    Article  PubMed  Google Scholar 

  34. Zeighami, Y. et al. Network structure of brain atrophy in de novo Parkinson’s disease. eLife 4, e08440 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  35. Fürtjes, A. E., Cole, J. H., Couvy-Duchesne, B. & Ritchie, S. J. A quantified comparison of cortical atlases on the basis of trait morphometricity. Cortex 158, 110–126 (2023).

    Article  PubMed  Google Scholar 

  36. Laumann, T. O. et al. Functional system and areal organization of a highly sampled individual human Brain. Neuron 87, 657–670 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. 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  Google Scholar 

  38. Bassett, D. S., Zurn, P. & Gold, J. I. On the nature and use of models in network neuroscience. Nat. Rev. Neurosci. 19, 566–578 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Bassett, D. S. & Sporns, O. Network neuroscience. Nat. Neurosci. 20, 353–364 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Franzmeier, N. et al. Functional connectivity associated with tau levels in ageing, Alzheimer’s, and small vessel disease. Brain 142, 1093–1107 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  41. Adams, J. N., Maass, A., Harrison, T. M., Baker, S. L. & Jagust, W. J. Cortical tau deposition follows patterns of entorhinal functional connectivity in aging. eLife 8, e49132 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Vogel, J. W. et al. Spread of pathological tau proteins through communicating neurons in human Alzheimer’s disease. Nat. Commun. 11, 2612 (2020). This study used epidemic-spread simulations to provide empirical evidence for the spread of tau pathology through brain networks in humans.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Lee, W. J. et al. Regional Aβ-tau interactions promote onset and acceleration of Alzheimer’s disease tau spreading. Neuron 110, 1932–1943.e5 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. de Flores, R. et al. Medial temporal lobe networks in Alzheimer’s disease: structural and molecular vulnerabilities. J. Neurosci. 42, 2131–2141 (2022).

    Article  PubMed  PubMed Central  Google Scholar 

  45. Sintini, I. et al. Functional connectivity to the premotor cortex maps onto longitudinal brain neurodegeneration in progressive apraxia of speech. Neurobiol. Aging 120, 105–116 (2022).

    Article  PubMed  PubMed Central  Google Scholar 

  46. Franzmeier, N. et al. Tau deposition patterns are associated with functional connectivity in primary tauopathies. Nat. Commun. 13, 1362 (2022). This article uniquely marries post mortem neuropathology with in vivo assessed functional connectivity to suggest a role of network spread in 4R primary tauopathies.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Cope, T. E. et al. Tau burden and the functional connectome in Alzheimer’s disease and progressive supranuclear palsy. Brain 141, 550–567 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  48. Sintini, I. et al. Tau and amyloid relationships with resting-state functional connectivity in atypical Alzheimer’s disease. Cereb. Cortex 31, 1693–1706 (2021).

    Article  PubMed  Google Scholar 

  49. Therriault, J. et al. Intrinsic connectivity of the human brain provides scaffold for tau aggregation in clinical variants of Alzheimer’s disease. Sci. Transl. Med 14, eabc8693 (2022).

    Article  CAS  PubMed  Google Scholar 

  50. Franzmeier, N. et al. Functional brain architecture is associated with the rate of tau accumulation in Alzheimer’s disease. Nat. Commun. 11, 347 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Franzmeier, N. et al. Patient-centered connectivity-based prediction of tau pathology spread in Alzheimer’s disease. Sci. Adv. 6, eabd1327 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Brown, J. A. et al. Patient-tailored, connectivity-based forecasts of spreading brain atrophy. Neuron 104, 856–868.e5 (2019). An important study highlighting the potential of individualized network information for making person-specific predictions about future neurodegeneration.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Yau, Y. et al. Network connectivity determines cortical thinning in early Parkinson’s disease progression. Nat. Commun. 9, 12 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Raj, A., Kuceyeski, A. & Weiner, M. A network diffusion model of disease progression in dementia. Neuron 73, 1204–1215 (2012). This important paper was the first to apply network diffusion simulations to explain the role of networks in constraining brain atrophy in neurodegenerative diseases.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. Iturria-Medina, Y., Sotero, R. C., Toussaint, P. J. & Evans, A. C., Alzheimer’s Disease Neuroimaging Initiative. Epidemic spreading model to characterize misfolded proteins propagation in aging and associated neurodegenerative disorders. PLoS Comput. Biol. 10, e1003956 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  56. Garbarino, S. & Lorenzi, M., Alzheimer’s Disease Neuroimaging Initiative. Investigating hypotheses of neurodegeneration by learning dynamical systems of protein propagation in the brain. NeuroImage 235, 117980 (2021).

    Article  CAS  PubMed  Google Scholar 

  57. Raj, A. & Powell, F. Models of network spread and network degeneration in brain disorders. Biol. Psychiatry Cogn. Neurosci. Neuroimaging 3, 788–797 (2018).

    PubMed  Google Scholar 

  58. Raj, A. & Powell, F. Network model of pathology spread recapitulates neurodegeneration and selective vulnerability in Huntington’s disease. NeuroImage 235, 118008 (2021).

    Article  CAS  PubMed  Google Scholar 

  59. Raj, A. et al. Network diffusion model of progression predicts longitudinal patterns of atrophy and metabolism in Alzheimer’s disease. Cell Rep. 10, 359–369 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  60. Abdelgawad, A. et al. Predicting longitudinal brain atrophy in Parkinson’s disease using a susceptible-infected-removed agent-based model. Netw. Neurosci. https://doi.org/10.1162/netn_a_00296 (2023).

    Article  Google Scholar 

  61. Schäfer, A., Mormino, E. C. & Kuhl, E. Network diffusion modeling explains longitudinal tau PET data. Front. Neurosci. 14, 566876 (2020).

    Article  PubMed  PubMed Central  Google Scholar 

  62. Damasceno, P. F. et al. Connectome-mediated prediction of future tau-PET burden in Alzheimer’s disease. J. Biotechnol. Biomed. 244-260 (2022).

  63. Yang, F. et al. Longitudinal predictive modeling of tau progression along the structural connectome. NeuroImage 237, 118126 (2021).

    Article  CAS  PubMed  Google Scholar 

  64. Oh, S. W. et al. A mesoscale connectome of the mouse brain. Nature 508, 207–214 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  65. Cornblath, E. J. et al. Computational modeling of tau pathology spread reveals patterns of regional vulnerability and the impact of a genetic risk factor. Sci. Adv. 7, eabg6677 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  66. Henderson, M. X. et al. Spread of α-synuclein pathology through the brain connectome is modulated by selective vulnerability and predicted by network analysis. Nat. Neurosci. 22, 1248–1257 (2019). This important study applied network diffusion models used in human studies to animal models of α-synuclein spread and further explored concepts of regional vulnerability.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  67. Thompson, T. B., Chaggar, P., Kuhl, E. & Goriely, A., Alzheimer’s Disease Neuroimaging Initiative. Protein–protein interactions in neurodegenerative diseases: a conspiracy theory. PLoS Comput. Biol. 16, e1008267 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  68. Fornari, S., Schäfer, A., Jucker, M., Goriely, A. & Kuhl, E. Prion-like spreading of Alzheimer’s disease within the brain’s connectome. J. R. Soc. Interface 16, 20190356 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  69. Zheng, Y.-Q. et al. Local vulnerability and global connectivity jointly shape neurodegenerative disease propagation. PLoS Biol. 17, e3000495 (2019). One of the first studies to incorporate gene expression and connectivity together into a single model predicting brain alterations (in Parkinson disease).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  70. Fornari, S., Schäfer, A., Kuhl, E. & Goriely, A. Spatially-extended nucleation-aggregation-fragmentation models for the dynamics of prion-like neurodegenerative protein-spreading in the brain and its connectome. J. Theor. Biol. 486, 110102 (2020).

    Article  PubMed  Google Scholar 

  71. Weickenmeier, J., Jucker, M., Goriely, A. & Kuhl, E. A physics-based model explains the prion-like features of neurodegeneration in Alzheimer’s disease, Parkinson’s disease, and amyotrophic lateral sclerosis. J. Mech. Phys. Solids 124, 264–281 (2019).

    Article  Google Scholar 

  72. Váša, F. & Mišić, B. Null models in network neuroscience. Nat. Rev. Neurosci. 23, 493–504 (2022).

    Article  PubMed  Google Scholar 

  73. Suárez, L. E., Richards, B. A., Lajoie, G. & Misic, B. Learning function from structure in neuromorphic networks. Nat. Mach. Intell. 3, 771–786 (2021).

    Article  Google Scholar 

  74. Shine, J. M. et al. Computational models link cellular mechanisms of neuromodulation to large-scale neural dynamics. Nat. Neurosci. 24, 765–776 (2021).

    Article  CAS  PubMed  Google Scholar 

  75. Filippi, M. et al. Changes in functional and structural brain connectome along the Alzheimer’s disease continuum. Mol. Psychiatry 25, 230–239 (2020).

    Article  PubMed  Google Scholar 

  76. Jonkman, L. E. et al. Relationship between β-amyloid and structural network topology in decedents without dementia. Neurology 95, e532–e544 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  77. Corriveau-Lecavalier, N. et al. A quadratic function of activation in individuals at risk of Alzheimer’s disease. Alzheimers Dement. 12, e12139 (2020).

    Google Scholar 

  78. Corriveau-Lecavalier, N., Natasha Rajah, M., Mellah, S. & Belleville, S. Latent patterns of task-related functional connectivity in relation to regions of hyperactivation in individuals at risk of Alzheimer’s disease. NeuroImage Clin. 30, 102643 (2021).

    Article  PubMed  PubMed Central  Google Scholar 

  79. Billette, O. V. et al. Novelty-related fMRI responses of precuneus and medial temporal regions in individuals at risk for Alzheimer disease. Neurology https://doi.org/10.1212/WNL.0000000000200667 (2022).

    Article  PubMed  PubMed Central  Google Scholar 

  80. Zott, B. et al. A vicious cycle of β amyloid-dependent neuronal hyperactivation. Science 365, 559–565 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  81. Busche, M. A. & Konnerth, A. Neuronal hyperactivity — a key defect in Alzheimer’s disease? Bioessays 37, 624–632 (2015).

    Article  PubMed  Google Scholar 

  82. Palop, J. J. & Mucke, L. Amyloid-β-induced neuronal dysfunction in Alzheimer’s disease: from synapses toward neural networks. Nat. Neurosci. 13, 812–818 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  83. Ovsepian, S. V., O’Leary, V. B., Zaborszky, L., Ntziachristos, V. & Dolly, J. O. Amyloid plaques of Alzheimer’s disease as hotspots of glutamatergic activity. Neuroscientist 25, 288–297 (2019).

    Article  CAS  PubMed  Google Scholar 

  84. Busche, M. A. et al. Clusters of hyperactive neurons near amyloid plaques in a mouse model of Alzheimer’s disease. Science 321, 1686–1689 (2008).

    Article  CAS  PubMed  Google Scholar 

  85. Zhou, B., Lu, J. G., Siddu, A., Wernig, M. & Südhof, T. C. Synaptogenic effect of APP-Swedish mutation in familial Alzheimer’s disease. Sci. Transl. Med. 14, eabn9380 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  86. Cirrito, J. R. et al. Synaptic activity regulates interstitial fluid amyloid-beta levels in vivo. Neuron 48, 913–922 (2005).

    Article  CAS  PubMed  Google Scholar 

  87. Hallinan, G. I., Vargas-Caballero, M., West, J. & Deinhardt, K. Tau misfolding efficiently propagates between individual intact hippocampal neurons. J. Neurosci. 39, 9623–9632 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  88. Yamada, K. et al. Neuronal activity regulates extracellular tau in vivo. J. Exp. Med. 211, 387–393 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  89. Rodriguez, G. A., Barrett, G. M., Duff, K. E. & Hussaini, S. A. Chemogenetic attenuation of neuronal activity in the entorhinal cortex reduces Aβ and tau pathology in the hippocampus. PLoS Biol. 18, e3000851 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  90. Palop, J. J. & Mucke, L. Epilepsy and cognitive impairments in Alzheimer disease. Arch. Neurol. 66, 435–440 (2009).

    Article  PubMed  PubMed Central  Google Scholar 

  91. Minkeviciene, R. et al. Amyloid β-induced neuronal hyperexcitability triggers progressive epilepsy. J. Neurosci. 29, 3453–3462 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  92. Lam, A. D. et al. Association of epileptiform abnormalities and seizures in Alzheimer disease. Neurology 95, e2259–e2270 (2020).

    Article  PubMed  PubMed Central  Google Scholar 

  93. Berron, D. et al. Higher CSF tau levels are related to hippocampal hyperactivity and object mnemonic discrimination in older adults. J. Neurosci. 39, 8788–8797 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  94. Huijbers, W. et al. Tau accumulation in clinically normal older adults is associated with hippocampal hyperactivity. J. Neurosci. 39, 548–556 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  95. Huijbers, W. et al. Amyloid-β deposition in mild cognitive impairment is associated with increased hippocampal activity, atrophy and clinical progression. Brain 138, 1023–1035 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  96. Quiroz, Y. T. et al. Hippocampal hyperactivation in presymptomatic familial Alzheimer’s disease. Ann. Neurol. 68, 865–875 (2010).

    Article  PubMed  PubMed Central  Google Scholar 

  97. O’Brien, J. L. et al. Longitudinal fMRI in elderly reveals loss of hippocampal activation with clinical decline. Neurology 74, 1969–1976 (2010).

    Article  PubMed  PubMed Central  Google Scholar 

  98. Bakker, A. et al. Reduction of hippocampal hyperactivity improves cognition in amnestic mild cognitive impairment. Neuron 74, 467–474 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  99. Bakker, A., Albert, M. S., Krauss, G., Speck, C. L. & Gallagher, M. Response of the medial temporal lobe network in amnestic mild cognitive impairment to therapeutic intervention assessed by fMRI and memory task performance. NeuroImage Clin. 7, 688–698 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  100. Sperling, R. A. et al. Amyloid deposition is associated with impaired default network function in older persons without dementia. Neuron 63, 178–188 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  101. Schultz, A. P. et al. Longitudinal degradation of the default/salience network axis in symptomatic individuals with elevated amyloid burden. NeuroImage Clin. 26, 102052 (2020).

    Article  PubMed  Google Scholar 

  102. Schultz, A. P. et al. Phases of hyperconnectivity and hypoconnectivity in the default mode and salience networks track with amyloid and tau in clinically normal individuals. J. Neurosci. 37, 4323–4331 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  103. Pereira, J. B. et al. Untangling the association of amyloid-β and tau with synaptic and axonal loss in Alzheimer’s disease. Brain 144, 310–324 (2021).

    Article  PubMed  Google Scholar 

  104. Hillary, F. G. & Grafman, J. H. Injured brains and adaptive networks: the benefits and costs of hyperconnectivity. Trends Cogn. Sci. 21, 385–401 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  105. Elman, J. A. et al. Neural compensation in older people with brain amyloid-β deposition. Nat. Neurosci. 17, 1316–1318 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  106. Leal, S. L., Landau, S. M., Bell, R. K. & Jagust, W. J. Hippocampal activation is associated with longitudinal amyloid accumulation and cognitive decline. eLife 6, 622978 (2017).

    Article  Google Scholar 

  107. Marks, S. M., Lockhart, S. N., Baker, S. L. & Jagust, W. J. Tau and β-amyloid are associated with medial temporal lobe structure, function, and memory encoding in normal aging. J. Neurosci. 37, 3192–3201 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  108. Adams, J. N. et al. Reduced repetition suppression in aging is driven by tau-related hyperactivity in medial temporal lobe. J. Neurosci. 41, 3917–3931 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  109. Mutlu, J. et al. Distinct influence of specific versus global connectivity on the different Alzheimer’s disease biomarkers. Brain 140, 3317–3328 (2017).

    Article  PubMed  Google Scholar 

  110. Putcha, D. et al. Hippocampal hyperactivation associated with cortical thinning in Alzheimer’s disease signature regions in non-demented elderly adults. J. Neurosci. 31, 17680–17688 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  111. Harris, S. S., Wolf, F., De Strooper, B. & Busche, M. A. Tipping the scales: peptide-dependent dysregulation of neural circuit dynamics in Alzheimer’s disease. Neuron 107, 417–435 (2020).

    Article  CAS  PubMed  Google Scholar 

  112. Mondragón-Rodríguez, S., Gu, N., Manseau, F. & Williams, S. Alzheimer’s transgenic model is characterized by very early brain network alterations and β-CTF fragment accumulation: reversal by β-secretase inhibition. Front. Cell. Neurosci. 12, 121 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  113. Gordon, B. A. et al. Task-evoked fMRI changes in attention networks are associated with preclinical Alzheimer’s disease biomarkers. Neurobiol. Aging 36, 1771–1779 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  114. Maass, A. et al. Alzheimer’s pathology targets distinct memory networks in the ageing brain. Brain 142, 2492–2509 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  115. Sepulcre, J. et al. Tau and amyloid β proteins distinctively associate to functional network changes in the aging brain. Alzheimers Dement. 13, 1261–1269 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  116. Corriveau-Lecavalier, N., Mellah, S., Clément, F. & Belleville, S. Evidence of parietal hyperactivation in individuals with mild cognitive impairment who progressed to dementia: a longitudinal fMRI study. NeuroImage Clin. 24, 101958 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  117. Foster, C. M., Kennedy, K. M., Horn, M. M., Hoagey, D. A. & Rodrigue, K. M. Both hyper- and hypo-activation to cognitive challenge are associated with increased beta-amyloid deposition in healthy aging: a nonlinear effect. NeuroImage 166, 285–292 (2018).

    Article  CAS  PubMed  Google Scholar 

  118. Kelberman, M. A. et al. Age-dependent dysregulation of locus coeruleus firing in a transgenic rat model of Alzheimer’s disease. Neurobiol. Aging https://doi.org/10.1016/j.neurobiolaging.2023.01.016 (2023).

    Article  PubMed  Google Scholar 

  119. Ittner, L. M. et al. Dendritic function of tau mediates amyloid-beta toxicity in Alzheimer’s disease mouse models. Cell 142, 387–397 (2010).

    Article  CAS  PubMed  Google Scholar 

  120. Roberson, E. D. et al. Amyloid-β/Fyn-induced synaptic, network, and cognitive impairments depend on tau levels in multiple mouse models of Alzheimer’s disease. J. Neurosci. 31, 700–711 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  121. Roberson, E. D. et al. Reducing endogenous tau ameliorates amyloid beta-induced deficits in an Alzheimer’s disease mouse model. Science 316, 750–754 (2007).

    Article  CAS  PubMed  Google Scholar 

  122. DeVos, S. L. et al. Antisense reduction of tau in adult mice protects against seizures. J. Neurosci. 33, 12887–12897 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  123. Holth, J. K. et al. Tau loss attenuates neuronal network hyperexcitability in mouse and Drosophila genetic models of epilepsy. J. Neurosci. 33, 1651–1659 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  124. Shao, E. et al. TAU ablation in excitatory neurons and postnatal TAU knockdown reduce epilepsy, SUDEP, and autism behaviors in a Dravet syndrome model. Sci. Transl. Med 14, eabm5527 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  125. Brown, J. et al. Tau in cerebrospinal fluid induces neuronal hyperexcitability and alters hippocampal theta oscillations. Acta Neuropathol. Commun. 11, 67 (2023).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  126. Ranasinghe, K. G. et al. Altered excitatory and inhibitory neuronal subpopulation parameters are distinctly associated with tau and amyloid in Alzheimer’s disease. eLife 11, e77850 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  127. Bassil, F. et al. Amyloid-beta (Aβ) plaques promote seeding and spreading of alpha-synuclein and tau in a mouse model of Lewy body disorders with Aβ pathology. Neuron 105, 260–275.e6 (2020).

    Article  CAS  PubMed  Google Scholar 

  128. Wu, Q. et al. Neuronal activity modulates alpha-synuclein aggregation and spreading in organotypic brain slice cultures and in vivo. Acta Neuropathol. 140, 831–849 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  129. Fortin, D. L. et al. Neural activity controls the synaptic accumulation of alpha-synuclein. J. Neurosci. 25, 10913–10921 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  130. Yamada, K. & Iwatsubo, T. Extracellular α-synuclein levels are regulated by neuronal activity. Mol. Neurodegener. 13, 9 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  131. Ramalingam, N. et al. Dynamic physiological α-synuclein S129 phosphorylation is driven by neuronal activity. npjParkinsons Dis. 9, 4 (2023).

    CAS  Google Scholar 

  132. Kulkarni, A. S. et al. Perturbation of in vivo neural activity following α-synuclein seeding in the olfactory bulb. J. Parkinsons Dis. 10, 1411–1427 (2020).

    Article  PubMed  PubMed Central  Google Scholar 

  133. Dagra, A. et al. Synuclein-induced dysregulation of neuronal activity contributes to murine dopamine neuron vulnerability. npj Parkinsons Dis. 7, 76 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  134. Abós, A. et al. Discriminating cognitive status in Parkinson’s disease through functional connectomics and machine learning. Sci. Rep. 7, 45347 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  135. Hassan, M. et al. Functional connectivity disruptions correlate with cognitive phenotypes in Parkinson’s disease. NeuroImage Clin. 14, 591–601 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  136. Hacker, C. D., Perlmutter, J. S., Criswell, S. R., Ances, B. M. & Snyder, A. Z. Resting state functional connectivity of the striatum in Parkinson’s disease. Brain 135, 3699–3711 (2012).

    Article  PubMed  PubMed Central  Google Scholar 

  137. Gratton, C. et al. Emergent functional network effects in Parkinson disease. Cereb. Cortex 29, 2509–2523 (2019).

    Article  PubMed  Google Scholar 

  138. De Micco, R. et al. Functional connectomics and disease progression in drug-naive Parkinson’s disease patients. Mov. Disord. 36, 1603–1616 (2021).

    Article  PubMed  Google Scholar 

  139. Luo, C. et al. Decreased resting-state interhemispheric functional connectivity in Parkinson’s disease. Biomed. Res. Int. 2015, 692684 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  140. Belete, D. et al. Association between antiepileptic drugs and incident Parkinson disease. JAMA Neurol. 80, 183–187 (2023).

    Article  PubMed  Google Scholar 

  141. Herzog, J. J. et al. TDP-43 dysfunction restricts dendritic complexity by inhibiting CREB activation and altering gene expression. Proc. Natl Acad. Sci. USA 117, 11760–11769 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  142. Wang, I.-F., Wu, L.-S., Chang, H.-Y. & Shen, C.-K. J. TDP-43, the signature protein of FTLD-U, is a neuronal activity-responsive factor. J. Neurochem. 105, 797–806 (2008).

    Article  CAS  PubMed  Google Scholar 

  143. Hosaka, Y. et al. Reduced TDP-43 expression improves neuronal activities in a Drosophila model of Perry syndrome. eBioMedicine 21, 218–227 (2017).

    Google Scholar 

  144. Dopper, E. G. P. et al. Structural and functional brain connectivity in presymptomatic familial frontotemporal dementia. Neurology 83, e19–e26 (2014).

    Article  PubMed  Google Scholar 

  145. Filippi, M. et al. Functional network connectivity in the behavioral variant of frontotemporal dementia. Cortex 49, 2389–2401 (2013).

    Article  PubMed  Google Scholar 

  146. Whitwell, J. L. et al. Imaging signatures of molecular pathology in behavioral variant frontotemporal dementia. J. Mol. Neurosci. 45, 372–378 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  147. Ranasinghe, K. G. et al. Distinct subtypes of behavioral variant frontotemporal dementia based on patterns of network degeneration. JAMA Neurol. 73, 1078–1088 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  148. Farb, N. A. S. et al. Abnormal network connectivity in frontotemporal dementia: evidence for prefrontal isolation. Cortex 49, 1856–1873 (2013).

    Article  PubMed  Google Scholar 

  149. Zhou, J. et al. Divergent network connectivity changes in behavioural variant frontotemporal dementia and Alzheimer’s disease. Brain 133, 1352–1367 (2010).

    Article  PubMed  PubMed Central  Google Scholar 

  150. Battistella, G. et al. Differential intrinsic functional connectivity changes in semantic variant primary progressive aphasia. NeuroImage Clin. 22, 101797 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  151. Guo, C. C. et al. Anterior temporal lobe degeneration produces widespread network-driven dysfunction. Brain 136, 2979–2991 (2013).

    Article  PubMed  PubMed Central  Google Scholar 

  152. Popal, H., Quimby, M., Hochberg, D., Dickerson, B. C. & Collins, J. A. Altered functional connectivity of cortical networks in semantic variant primary progressive aphasia. NeuroImage Clin. 28, 102494 (2020).

    Article  PubMed  PubMed Central  Google Scholar 

  153. Lee, S. E. et al. Thalamo-cortical network hyperconnectivity in preclinical progranulin mutation carriers. NeuroImage Clin. 22, 101751 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  154. Lui, H. et al. Progranulin deficiency promotes circuit-specific synaptic pruning by microglia via complement activation. Cell 165, 921–935 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  155. Gregory, S. et al. Operationalizing compensation over time in neurodegenerative disease. Brain 140, 1158–1165 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  156. McColgan, P. et al. Neurofilament light associated connectivity in young-adult Huntington’s disease is related to neuronal genes. Brain 145, 3953–3967 (2022).

    Article  PubMed  PubMed Central  Google Scholar 

  157. Frere, S. & Slutsky, I. Alzheimer’s disease: from firing instability to homeostasis network collapse. Neuron 97, 32–58 (2018).

    Article  CAS  PubMed  Google Scholar 

  158. Styr, B. & Slutsky, I. Imbalance between firing homeostasis and synaptic plasticity drives early-phase Alzheimer’s disease. Nat. Neurosci. 21, 463–473 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  159. Delbeuck, X., Van der Linden, M. & Collette, F. Alzheimer’s disease as a disconnection syndrome? Neuropsychol. Rev. 13, 79–92 (2003).

    Article  CAS  PubMed  Google Scholar 

  160. Catani, M. & Ffytche, D. H. The rises and falls of disconnection syndromes. Brain 128, 2224–2239 (2005).

    Article  PubMed  Google Scholar 

  161. Pasquini, L. et al. Medial temporal lobe disconnection and hyperexcitability across Alzheimer’s disease stages. J. Alzheimers Dis. Rep. 3, 103–112 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  162. Tahmasian, M. et al. The lower hippocampus global connectivity, the higher its local metabolism in Alzheimer disease. Neurology 84, 1956–1963 (2015).

    Article  CAS  PubMed  Google Scholar 

  163. Jones, D. T. et al. Cascading network failure across the Alzheimer’s disease spectrum. Brain 139, 547–562 (2016).

    Article  PubMed  Google Scholar 

  164. Hampton, O. L. et al. Resting-state functional connectivity and amyloid burden influence longitudinal cortical thinning in the default mode network in preclinical Alzheimer’s disease. NeuroImage Clin. 28, 102407 (2020).

    Article  PubMed  PubMed Central  Google Scholar 

  165. Harrison, T. M. et al. Tau deposition is associated with functional isolation of the hippocampus in aging. Nat. Commun. 10, 4900 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  166. Pasquini, L. et al. Increased intrinsic activity of medial-temporal lobe subregions is associated with decreased cortical thickness of medial-parietal areas in patients with Alzheimer’s disease dementia. J. Alzheimers Dis. 51, 313–326 (2016).

    Article  CAS  PubMed  Google Scholar 

  167. Stoub, T. R. et al. Hippocampal disconnection contributes to memory dysfunction in individuals at risk for Alzheimer’s disease. Proc. Natl Acad. Sci. USA 103, 10041–10045 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  168. Wisse, L. E. M. et al. Hippocampal disconnection in early Alzheimer’s disease: a 7 tesla MRI study. J. Alzheimers Dis. 45, 1247–1256 (2015).

    Article  PubMed  Google Scholar 

  169. Mandino, F. et al. The lateral entorhinal cortex is a hub for local and global dysfunction in early Alzheimer’s disease states. J. Cereb. Blood Flow. Metab. 42, 1616–1631 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  170. Walsh, D. M. & Selkoe, D. J. A critical appraisal of the pathogenic protein spread hypothesis of neurodegeneration. Nat. Rev. Neurosci. 17, 251–260 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  171. Peng, C., Trojanowski, J. Q. & Lee, V. M.-Y. Protein transmission in neurodegenerative disease. Nat. Rev. Neurol. 16, 199–212 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  172. Olmi, S., Petkoski, S., Guye, M., Bartolomei, F. & Jirsa, V. Controlling seizure propagation in large-scale brain networks. PLoS Comput. Biol. 15, e1006805 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  173. Shah, P. et al. Local structural connectivity directs seizure spread in focal epilepsy. Preprint at bioRxiv https://doi.org/10.1101/406793 (2018).

    Article  Google Scholar 

  174. Millán, A. P. et al. Epidemic models characterize seizure propagation and the effects of epilepsy surgery in individualized brain networks based on MEG and invasive EEG recordings. Sci. Rep. 12, 4086 (2022).

    Article  PubMed  PubMed Central  Google Scholar 

  175. Tai, X. Y. et al. Hyperphosphorylated tau in patients with refractory epilepsy correlates with cognitive decline: a study of temporal lobe resections. Brain 139, 2441–2455 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  176. Seguin, C. et al. Communication dynamics in the human connectome shape the cortex-wide propagation of direct electrical stimulation. Neuron 111, 1391–1401.e5 (2023).

    Article  CAS  PubMed  Google Scholar 

  177. Buldyrev, S. V., Parshani, R., Paul, G., Stanley, H. E. & Havlin, S. Catastrophic cascade of failures in interdependent networks. Nature 464, 1025–1028 (2010).

    Article  CAS  PubMed  Google Scholar 

  178. Yang, Y., Nishikawa, T. & Motter, A. E. Small vulnerable sets determine large network cascades in power grids. Science 358, eaan3184 (2017).

    Article  PubMed  Google Scholar 

  179. Brockmann, D. & Helbing, D. The hidden geometry of complex, network-driven contagion phenomena. Science 342, 1337–1342 (2013).

    Article  CAS  PubMed  Google Scholar 

  180. Spreng, R. N. & Turner, G. R. Structural covariance of the default network in healthy and pathological aging. J. Neurosci. 33, 15226–15234 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  181. Alexander-Bloch, A., Raznahan, A., Bullmore, E. & Giedd, J. The convergence of maturational change and structural covariance in human cortical networks. J. Neurosci. 33, 2889–2899 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  182. Raznahan, A. et al. Patterns of coordinated anatomical change in human cortical development: a longitudinal neuroimaging study of maturational coupling. Neuron 72, 873–884 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  183. Arnatkeviciute, A., Fulcher, B. D., Bellgrove, M. A. & Fornito, A. Where the genome meets the connectome: understanding how genes shape human brain connectivity. NeuroImage 244, 118570 (2021).

    Article  CAS  PubMed  Google Scholar 

  184. Richiardi, J. et al. Brain networks. Correlated gene expression supports synchronous activity in brain networks. Science 348, 1241–1244 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  185. Mrdjen, D. et al. The basis of cellular and regional vulnerability in Alzheimer’s disease. Acta Neuropathol. 138, 729–749 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  186. Haak, K. V. & Beckmann, C. F. Understanding brain organisation in the face of functional heterogeneity and functional multiplicity. NeuroImage 220, 117061 (2020).

    Article  PubMed  Google Scholar 

  187. Margulies, D. S. et al. Situating the default-mode network along a principal gradient of macroscale cortical organization. Proc. Natl Acad. Sci. USA 113, 12574–12579 (2016). A seminal paper revealing the hierarchical organization of macroscale spatial gradients of the human connectome.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  188. Burt, J. B. et al. Hierarchy of transcriptomic specialization across human cortex captured by structural neuroimaging topography. Nat. Neurosci. 21, 1251–1259 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  189. Mesulam, M. M. From sensation to cognition. Brain 121, 1013–1052 (1998).

    Article  PubMed  Google Scholar 

  190. Sydnor, V. J. et al. Neurodevelopment of the association cortices: patterns, mechanisms, and implications for psychopathology. Neuron 109, 2820–2846 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  191. Bolt, T. et al. A parsimonious description of global functional brain organization in three spatiotemporal patterns. Nat. Neurosci. 25, 1093–1103 (2022).

    Article  CAS  PubMed  Google Scholar 

  192. Vogel, J. W. et al. Conserved whole-brain spatiomolecular gradients shape adult brain functional organization. Preprint at bioRxiv https://doi.org/10.1101/2022.09.18.508425 (2022).

    Article  PubMed  PubMed Central  Google Scholar 

  193. Huntenburg, J. M. et al. A systematic relationship between functional connectivity and intracortical myelin in the human cerebral cortex. Cereb. Cortex 27, 981–997 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  194. Goulas, A. et al. The natural axis of transmitter receptor distribution in the human cerebral cortex. Proc. Natl Acad. Sci. USA 118, e2020574118 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  195. Hansen, J. et al. Mapping neurotransmitter systems to the structural and functional organization of the human neocortex. Nat. Neurosci. 25, 1569–1581 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  196. Brown, J. A., Lee, A. J., Pasquini, L. & Seeley, W. W. A dynamic gradient architecture generates brain activity states. Neuroimage 261, 119526 (2022).

    Article  PubMed  Google Scholar 

  197. Raut, R. V., Snyder, A. Z. & Raichle, M. E. Hierarchical dynamics as a macroscopic organizing principle of the human brain. Proc. Natl Acad. Sci. USA 117, 20890–20897 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  198. Lowe, A. J. et al. Targeting age-related differences in brain and cognition with multimodal imaging and connectome topography profiling. Hum. Brain Mapp. 40, 5213–5230 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  199. Iaccarino, L. et al. Spatial relationships between molecular pathology and neurodegeneration in the Alzheimer’s disease continuum. Cereb. Cortex 31, 1–14 (2021).

    Article  PubMed  Google Scholar 

  200. Vlassenko, A. G. et al. Spatial correlation between brain aerobic glycolysis and amyloid-β (Aβ) deposition. Proc. Natl Acad. Sci. USA 107, 17763–17767 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  201. Vértes, P. E. et al. Gene transcription profiles associated with inter-modular hubs and connection distance in human functional magnetic resonance imaging networks. Philos. Trans. R. Soc. Lond. B Biol. Sci. 371, 0362 (2016).

    Article  Google Scholar 

  202. Paquola, C. et al. Microstructural and functional gradients are increasingly dissociated in transmodal cortices. PLoS Biol. 17, e3000284 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  203. Huntenburg, J. M., Bazin, P.-L. & Margulies, D. S. Large-scale gradients in human cortical organization. Trends Cogn. Sci. 22, 21–31 (2018).

    Article  PubMed  Google Scholar 

  204. Shafiei, G. et al. Topographic gradients of intrinsic dynamics across neocortex. eLife 9, e62116 (2020).

    CAS  Google Scholar 

  205. Mahjoory, K., Schoffelen, J.-M., Keitel, A. & Gross, J. The frequency gradient of human resting-state brain oscillations follows cortical hierarchies. eLife 9, e53715 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  206. Grothe, M. J. et al. Molecular properties underlying regional vulnerability to Alzheimer’s disease pathology. Brain 141, 2755–2771 (2018).

    PubMed  PubMed Central  Google Scholar 

  207. Sepulcre, J. et al. Neurogenetic contributions to amyloid beta and tau spreading in the human cortex. Nat. Med. 24, 1910–1918 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  208. Montal, V. et al. Network tau spreading is vulnerable to the expression gradients of APOE and glutamatergic-related genes. Sci. Transl. Med 14, eabn7273 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  209. Altmann, A. et al. Analysis of brain atrophy and local gene expression in genetic frontotemporal dementia. Brain Commun. 2, fcaa122 (2020).

    Article  PubMed  PubMed Central  Google Scholar 

  210. Fu, H. et al. A tau homeostasis signature is linked with the cellular and regional vulnerability of excitatory neurons to tau pathology. Nat. Neurosci. 22, 47–56 (2019).

    Article  CAS  PubMed  Google Scholar 

  211. Leng, K. et al. Molecular characterization of selectively vulnerable neurons in Alzheimer’s disease. Nat. Neurosci. 24, 276–287 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  212. Kamath, T. et al. Single-cell genomic profiling of human dopamine neurons identifies a population that selectively degenerates in Parkinson’s disease. Nat. Neurosci. 25, 588–595 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  213. Nana, A. L. et al. Neurons selectively targeted in frontotemporal dementia reveal early stage TDP-43 pathobiology. Acta Neuropathol. 137, 27–46 (2019).

    Article  PubMed  Google Scholar 

  214. 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 

  215. Sebastian Pineda, S. et al. Single-cell profiling of the human primary motor cortex in ALS and FTLD. Preprint at bioRxiv https://doi.org/10.1101/2021.07.07.451374 (2021).

    Article  Google Scholar 

  216. Freeze, B., Pandya, S., Zeighami, Y. & Raj, A. Regional transcriptional architecture of Parkinson’s disease pathogenesis and network spread. Brain 142, 3072–3085 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  217. Anand, C., Maia, P. D., Torok, J., Mezias, C. & Raj, A. The effects of microglia on tauopathy progression can be quantified using Nexopathy in silico (Nexis) models. Sci. Rep. 12, 21170 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  218. Rahayel, S. et al. Differentially targeted seeding reveals unique pathological alpha-synuclein propagation patterns. Brain 145, 1743–1756 (2022).

    Article  PubMed  Google Scholar 

  219. Shafiei, G. et al. Network structure and transcriptomic vulnerability shape atrophy in frontotemporal dementia. Brain 146, 321–336 (2023).

    Article  PubMed  Google Scholar 

  220. Adewale, Q., Khan, A. F., Carbonell, F. & Iturria-Medina, Y., Alzheimer’s Disease Neuroimaging Initiative. Integrated transcriptomic and neuroimaging brain model decodes biological mechanisms in aging and Alzheimer’s disease. eLife 10, e62589 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  221. Mezias, C., LoCastro, E., Xia, C. & Raj, A. Connectivity, not region-intrinsic properties, predicts regional vulnerability to progressive tau pathology in mouse models of disease. Acta Neuropathol. Commun. 5, 61 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  222. Jellinger, K. A. Interaction between pathogenic proteins in neurodegenerative disorders. J. Cell. Mol. Med. 16, 1166–1183 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  223. Robinson, J. L. et al. The development and convergence of co-pathologies in Alzheimer’s disease. Brain 144, 953–962 (2021).

    Article  PubMed  PubMed Central  Google Scholar 

  224. Irwin, D. J. et al. CSF tau and β-amyloid predict cerebral synucleinopathy in autopsied Lewy body disorders. Neurology 90, e1038–e1046 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  225. Josephs, K. A. et al. TDP-43 is a key player in the clinical features associated with Alzheimer’s disease. Acta Neuropathol. 127, 811–824 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  226. Bartzokis, G. Age-related myelin breakdown: a developmental model of cognitive decline and Alzheimer’s disease. Neurobiol. Aging 25, 5–18 (2004). Author reply 49–62.

    Article  CAS  PubMed  Google Scholar 

  227. Braak, H. & Tredici, K. D.Poor and protracted myelination as a contributory factor to neurodegenerative disorders. Neurobiol. Aging 25, 19–23 (2004).

    Article  CAS  PubMed  Google Scholar 

  228. Rubinski, A. et al. Higher levels of myelin are associated with higher resistance against tau pathology in Alzheimer’s disease. Alz. Res. Ther. 14, 139 (2022).

    Article  CAS  Google Scholar 

  229. Sakae, N. et al. Clinicopathologic subtype of Alzheimer’s disease presenting as corticobasal syndrome. Alzheimers Dement. 15, 1218–1228 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  230. Zachlod, D., Bludau, S., Cichon, S., Palomero-Gallagher, N. & Amunts, K. Combined analysis of cytoarchitectonic, molecular and transcriptomic patterns reveal differences in brain organization across human functional brain systems. NeuroImage 257, 119286 (2022).

    Article  PubMed  Google Scholar 

  231. Khan, A. F. et al. Personalized brain models identify neurotransmitter receptor changes in Alzheimer’s disease. Brain 145, 1785–1804 (2022).

    Article  PubMed  Google Scholar 

  232. Hansen, J. Y. et al. Local molecular and global connectomic contributions to cross-disorder cortical abnormalities. Nat. Commun. 13, 4682 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  233. Vogel, J. W. et al. Four distinct trajectories of tau deposition identified in Alzheimer’s disease. Nat. Med. 27, 871–881 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  234. Levitis, E. et al. Differentiating amyloid beta spread in autosomal dominant and sporadic Alzheimer’s disease. Brain Commun. 4, fcac085 (2022).

    Article  PubMed  PubMed Central  Google Scholar 

  235. Lee, W. J. et al. Dynamic network model reveals distinct tau spreading patterns in early- and late-onset Alzheimer disease. Alzheimers Res. Ther. 14, 121 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  236. Finn, E. S. et al. Functional connectome fingerprinting: identifying individuals using patterns of brain connectivity. Nat. Neurosci. 18, 1664–1671 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  237. Vogel, J. W. & Hansson, O. Subtypes of Alzheimer’s disease: questions, controversy, and meaning. Trends Neurosci. 45, 342–345 (2022).

    Article  CAS  PubMed  Google Scholar 

  238. Franzmeier, N. et al. Left frontal cortex connectivity underlies cognitive reserve in prodromal Alzheimer disease. Neurology 88, 1054–1061 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  239. Ewers, M. et al. Segregation of functional networks is associated with cognitive resilience in Alzheimer’s disease. Brain 144, 2176–2185 (2021).

    Article  PubMed  PubMed Central  Google Scholar 

  240. Rittman, T. et al. Functional network resilience to pathology in presymptomatic genetic frontotemporal dementia. Neurobiol. Aging 77, 169–177 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  241. Tsvetanov, K. A. et al. Brain functional network integrity sustains cognitive function despite atrophy in presymptomatic genetic frontotemporal dementia. Alzheimers Dement. 17, 500–514 (2021).

    Article  CAS  PubMed  Google Scholar 

  242. Birn, R. M. et al. The effect of scan length on the reliability of resting-state fMRI connectivity estimates. NeuroImage 83, 550–558 (2013).

    Article  PubMed  Google Scholar 

  243. Gordon, E. M. et al. Precision functional mapping of individual human brains. Neuron 95, 791–807.e7 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  244. Kong, R. et al. Spatial topography of individual-specific cortical networks predicts human cognition, personality, and emotion. Cereb. Cortex 29, 2533–2551 (2019).

    Article  PubMed  Google Scholar 

  245. Kong, R. et al. Individual-specific areal-level parcellations improve functional connectivity prediction of behavior. Cereb. Cortex 31, 4477–4500 (2021).

    Article  PubMed  PubMed Central  Google Scholar 

  246. Zhou, J. et al. Deep learning sequence-based ab initio prediction of variant effects on expression and disease risk. Nat. Genet. 50, 1171–1179 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  247. Kelley, D. R. et al. Sequential regulatory activity prediction across chromosomes with convolutional neural networks. Genome Res. 28, 739–750 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  248. Agarwal, V. & Shendure, J. Predicting mRNA abundance directly from genomic sequence using deep convolutional neural networks. Cell Rep. 31, 107663 (2020).

    Article  CAS  PubMed  Google Scholar 

  249. Avsec, Ž. et al. Effective gene expression prediction from sequence by integrating long-range interactions. Nat. Methods 18, 1196–1203 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  250. Basu, M., Wang, K., Ruppin, E. & Hannenhalli, S. Predicting tissue-specific gene expression from whole blood transcriptome. Sci. Adv. 7, eabd6991 (2021).

    Article  CAS  PubMed  Google Scholar 

  251. Jiang, Y. et al. Large-scale plasma proteomic profiling identifies a high-performance biomarker panel for Alzheimer’s disease screening and staging. Alzheimers Dement. 18, 88–102 (2022).

    Article  CAS  PubMed  Google Scholar 

  252. Phillips, B. et al. Proteome wide association studies of LRRK2 variants identify novel causal and druggable for Parkinson’s disease. npj Parkinsons Dis. 9, 107 (2023).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  253. Jacobs, H. I. L. et al. Structural tract alterations predict downstream tau accumulation in amyloid-positive older individuals. Nat. Neurosci. 21, 424–431 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  254. Pichet Binette, A. et al. Bundle-specific associations between white matter microstructure and Aβ and tau pathology in preclinical Alzheimer’s disease. eLife 10, e62929 (2021).

    Google Scholar 

  255. Wen, Q. et al. Tau-related white-matter alterations along spatially selective pathways. NeuroImage 226, 117560 (2021).

    Article  PubMed  Google Scholar 

  256. Alexandersen, C. G., de Haan, W., Bick, C. & Goriely, A. A multi-scale model explains oscillatory slowing and neuronal hyperactivity in Alzheimer’s disease. J. R. Soc. Interface 20, 20220607 (2023).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  257. Villemagne, V. L. et al. Longitudinal assessment of Aβ and cognition in aging and Alzheimer disease. Ann. Neurol. 69, 181–192 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  258. Villain, N. et al. Regional dynamics of amyloid-β deposition in healthy elderly, mild cognitive impairment and Alzheimer’s disease: a voxelwise PiB–PET longitudinal study. Brain 135, 2126–2139 (2012).

    Article  PubMed  Google Scholar 

  259. Jack, C. R. Jr et al. Brain β-amyloid load approaches a plateau. Neurology 80, 890–896 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  260. Phillips, J. S. et al. Rates of longitudinal change in 18F‐flortaucipir PET vary by brain region, cognitive impairment, and age in atypical Alzheimer’s disease. Alzheimers Dement. 18, 1235–1247 (2022).

    Article  CAS  PubMed  Google Scholar 

  261. Nishihira, Y. et al. Sporadic amyotrophic lateral sclerosis of long duration is associated with relatively mild TDP-43 pathology. Acta Neuropathol. 117, 45–53 (2009).

    Article  CAS  PubMed  Google Scholar 

  262. Mišić, B. et al. Cooperative and competitive spreading dynamics on the human connectome. Neuron 86, 1518–1529 (2015).

    Article  PubMed  Google Scholar 

  263. Robinson, J. L. et al. Neurodegenerative disease concomitant proteinopathies are prevalent, age-related and APOE4-associated. Brain 141, 2181–2193 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  264. Rahimi, J. & Kovacs, G. G. Prevalence of mixed pathologies in the aging brain. Alzheimers Res. Ther. 6, 82 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  265. Jbabdi, S., Sotiropoulos, S. N., Savio, A. M., Graña, M. & Behrens, T. E. J. Model-based analysis of multishell diffusion MR data for tractography: how to get over fitting problems. Magn. Reson. Med. 68, 1846–1855 (2012).

    Article  PubMed  PubMed Central  Google Scholar 

  266. Schilling, K. G. et al. Limits to anatomical accuracy of diffusion tractography using modern approaches. NeuroImage 185, 1–11 (2019).

    Article  PubMed  Google Scholar 

  267. Schiavi, S. et al. A new method for accurate in vivo mapping of human brain connections using microstructural and anatomical information. Sci. Adv. 6, eaba8245 (2020).

    Article  PubMed  PubMed Central  Google Scholar 

  268. Cieslak, M. et al. QSIPrep: an integrative platform for preprocessing and reconstructing diffusion MRI data. Nat. Methods 18, 775–778 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  269. Kucyi, A. et al. Intracranial electrophysiology reveals reproducible intrinsic functional connectivity within human brain networks. J. Neurosci. 38, 4230–4242 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  270. Gratton, C. et al. Defining individual-specific functional neuroanatomy for precision psychiatry. Biol. Psychiatry 88, 28–39 (2020).

    Article  PubMed  Google Scholar 

  271. Corriveau-Lecavalier, N. et al. Default mode network failure and neurodegeneration across aging and amnestic and dysexecutive Alzheimer’s disease. Brain Commun. 5, fcad058 (2023).

    Article  PubMed  PubMed Central  Google Scholar 

  272. Satterthwaite, T. D. et al. Motion artifact in studies of functional connectivity: characteristics and mitigation strategies. Hum. Brain Mapp. 40, 2033–2051 (2019).

    Article  PubMed  Google Scholar 

  273. Raimondo, L. et al. Advances in resting state fMRI acquisitions for functional connectomics. NeuroImage 243, 118503 (2021).

    Article  CAS  PubMed  Google Scholar 

  274. Eyler, L. T. et al. Resting state abnormalities of the default mode network in mild cognitive impairment: a systematic review and meta-analysis. J. Alzheimers Dis. 70, 107–120 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  275. Badhwar, A. et al. Resting-state network dysfunction in Alzheimer’s disease: a systematic review and meta-analysis. Alzheimers Dement. 8, 73–85 (2017).

    Google Scholar 

  276. Lawrence, S. J. D., Formisano, E., Muckli, L. & de Lange, F. P. Laminar fMRI: applications for cognitive neuroscience. NeuroImage 197, 785–791 (2019).

    Article  PubMed  Google Scholar 

  277. Braak, H. & Del Tredici, K. Spreading of tau pathology in sporadic Alzheimer’s disease along cortico-cortical top-down connections. Cereb. Cortex 28, 3372–3384 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  278. Lemoine, L., Saint-Aubert, L., Nennesmo, I., Gillberg, P.-G. & Nordberg, A. Cortical laminar tau deposits and activated astrocytes in Alzheimer’s disease visualised by 3H-THK5117 and 3H-deprenyl autoradiography. Sci. Rep. 7, 45496 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  279. Maynard, K. R. et al. Transcriptome-scale spatial gene expression in the human dorsolateral prefrontal cortex. Nat. Neurosci. 24, 425–436 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  280. Priovoulos, N. et al. High-resolution in vivo imaging of human locus coeruleus by magnetization transfer MRI at 3T and 7T. NeuroImage 168, 427–436 (2018).

    Article  PubMed  Google Scholar 

  281. Brendel, M. et al. Assessment of 18F-PI-2620 as a biomarker in progressive supranuclear palsy. JAMA Neurol. 77, 1408–1419 (2020).

    Article  PubMed  Google Scholar 

  282. Abeyasinghe, P. M. et al. Tracking Huntington’s disease progression using motor, functional, cognitive, and imaging markers. Mov. Disord. 36, 2282–2292 (2021).

    Article  PubMed  PubMed Central  Google Scholar 

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

    Article  PubMed  Google Scholar 

  284. Johnson, E. L. & Knight, R. T. Intracranial recordings and human memory. Curr. Opin. Neurobiol. 31, 18–25 (2015).

    Article  CAS  PubMed  Google Scholar 

  285. Schurr, R. & Mezer, A. A. The glial framework reveals white matter fiber architecture in human and primate brains. Science 374, 762–767 (2021).

    Article  CAS  PubMed  Google Scholar 

  286. Yushkevich, P. A. et al. Three-dimensional mapping of neurofibrillary tangle burden in the human medial temporal lobe. Brain 144, 2784–2797 (2021).

    Article  PubMed  PubMed Central  Google Scholar 

  287. Tisdall, M. D. et al. Ex vivo MRI and histopathology detect novel iron-rich cortical inflammation in frontotemporal lobar degeneration with tau versus TDP-43 pathology. NeuroImage Clin. 33, 102913 (2022).

    Article  PubMed  Google Scholar 

  288. Kadakia, K. T., Beckman, A. L., Ross, J. S. & Krumholz, H. M. Leveraging open science to accelerate research. N. Engl. J. Med. 384, e61 (2021).

    Article  PubMed  Google Scholar 

  289. Poldrack, R. A. et al. Scanning the horizon: towards transparent and reproducible neuroimaging research. Nat. Rev. Neurosci. 18, 115–126 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  290. Besançon, L. et al. Open science saves lives: lessons from the COVID-19 pandemic. BMC Med. Res. Methodol. 21, 117 (2021).

    Article  PubMed  PubMed Central  Google Scholar 

  291. Boes, A. D. et al. Network localization of neurological symptoms from focal brain lesions. Brain 138, 3061–3075 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  292. Crossley, N. A. et al. The hubs of the human connectome are generally implicated in the anatomy of brain disorders. Brain 137, 2382–2395 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  293. Ferguson, M. A. et al. A human memory circuit derived from brain lesions causing amnesia. Nat. Commun. 10, 3497 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  294. Tetreault, A. M. et al. Network localization of clinical, cognitive, and neuropsychiatric symptoms in Alzheimer’s disease. Brain 143, 1249–1260 (2020).

    Article  PubMed  PubMed Central  Google Scholar 

  295. Jones, D. et al. A computational model of neurodegeneration in Alzheimer’s disease. Nat. Commun. 13, 1643 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  296. Biel, D. et al. Combining tau-PET and fMRI meta-analyses for patient-centered prediction of cognitive decline in Alzheimer’s disease. Alzheimers Res. Ther. 14, 166 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  297. Larivière, S. et al. Network-based atrophy modeling in the common epilepsies: a worldwide ENIGMA study. Sci. Adv. 6, eabc6457 (2020).

    Article  PubMed  PubMed Central  Google Scholar 

  298. Leliveld, S. R. et al. Insolubility of disrupted-in-schizophrenia 1 disrupts oligomer-dependent interactions with nuclear distribution element 1 and is associated with sporadic mental disease. J. Neurosci. 28, 3839–3845 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  299. Zhu, S., Abounit, S., Korth, C. & Zurzolo, C. Transfer of disrupted-in-schizophrenia 1 aggregates between neuronal-like cells occurs in tunnelling nanotubes and is promoted by dopamine. Open Biol. 7, 160328 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  300. Shafiei, G. et al. Spatial patterning of tissue volume loss in schizophrenia reflects brain network architecture. Biol. Psychiatry 87, 727–735 (2020).

    Article  CAS  PubMed  Google Scholar 

  301. Chopra, S. et al. Network-based spreading of grey matter changes across different stages of psychosis. Preprint at bioRxiv https://doi.org/10.1101/2022.01.11.22268989 (2022).

    Article  Google Scholar 

  302. Devi, L. & Ohno, M. Mechanisms that lessen benefits of β-secretase reduction in a mouse model of Alzheimer’s disease. Transl. Psychiatry 3, e284 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  303. Sanchez, P. E. et al. Levetiracetam suppresses neuronal network dysfunction and reverses synaptic and cognitive deficits in an Alzheimer’s disease model. Proc. Natl Acad. Sci. USA 109, E2895–E2903 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  304. Klink, K. et al. Targeting hippocampal hyperactivity with real-time fMRI neurofeedback: protocol of a single-blind randomized controlled trial in mild cognitive impairment. BMC Psychiatry https://doi.org/10.1186/s12888-021-03091-8 (2021).

    Article  PubMed  PubMed Central  Google Scholar 

  305. Ríos, A. S. et al. Optimal deep brain stimulation sites and networks for stimulation of the fornix in Alzheimer’s disease. Nat. Commun. 13, 7707 (2022).

    Article  PubMed  PubMed Central  Google Scholar 

  306. Koch, G. et al. Precuneus magnetic stimulation for Alzheimer’s disease: a randomized, sham-controlled trial. Brain 145, 3776–3786 (2022).

    Article  PubMed  PubMed Central  Google Scholar 

  307. Chard, D. T. et al. Mind the gap: from neurons to networks to outcomes in multiple sclerosis. Nat. Rev. Neurol. 17, 173–184 (2021).

    Article  PubMed  Google Scholar 

  308. Markello, R. D. & Misic, B. Comparing spatial null models for brain maps. NeuroImage 236, 118052 (2021).

    Article  PubMed  Google Scholar 

  309. Sporns, O. Graph theory methods: applications in brain networks. Dialogues Clin. Neurosci. 20, 111–121 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  310. LaPoint, M. R., Baker, S. L., Landau, S. M., Harrison, T. M. & Jagust, W. J. Rates of β-amyloid deposition indicate widespread simultaneous accumulation throughout the brain. Neurobiol. Aging 115, 1–11 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  311. Whittington, A., Sharp, D. J. & Gunn, R. N., Alzheimer’s Disease Neuroimaging Initiative. Spatiotemporal distribution of β-amyloid in Alzheimer disease is the result of heterogeneous regional carrying capacities. J. Nucl. Med. 59, 822–827 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  312. Meisl, G. et al. In vivo rate-determining steps of tau seed accumulation in Alzheimer’s disease. Sci. Adv. 7, eabh1448 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  313. Dewenter, A. et al. Disentangling the effects of Alzheimer’s and small vessel disease on white matter fiber tracts. Brain 164, 678–689 (2023).

    Article  Google Scholar 

  314. Fu, H., Hardy, J. & Duff, K. E. Selective vulnerability in neurodegenerative diseases. Nat. Neurosci. 21, 1350–1358 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  315. Arnatkeviciute, A. et al. Genetic influences on hub connectivity of the human connectome. Nat. Commun. 12, 4237 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  316. Arnatkeviciute, A., Fulcher, B. D., Bellgrove, M. A. & Fornito, A. Imaging transcriptomics of brain disorders. Biol. Psychiatry Glob. Open Sci. https://doi.org/10.1016/j.bpsgos.2021.10.002 (2021).

    Article  PubMed  PubMed Central  Google Scholar 

  317. Appel, S. H. A unifying hypothesis for the cause of amyotrophic lateral sclerosis, parkinsonism, and Alzheimer disease. Ann. Neurol. 10, 499–505 (1981).

    Article  CAS  PubMed  Google Scholar 

  318. Salehi, A. et al. Increased App expression in a mouse model of Down’s syndrome disrupts NGF transport and causes cholinergic neuron degeneration. Neuron 51, 29–42 (2006).

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

The authors thank P. Chaggar for comments and suggestions on the manuscript. J.W.V. acknowledges support from the SciLifeLab & Wallenberg Data Driven Life Science Program (grant no. KAW 2020.0239). D.S.B. acknowledges support from the Michael J. Fox Foundation ASAP Initiative.

Author information

Authors and Affiliations

Authors

Contributions

J.W.V., M.E., N.C.-L., N.F., J.A.B. and A.M. researched data for the article and along with J.B.P. made a substantial contribution to discussion of content, wrote, reviewed and edited the manuscript before submission. A substantial contribution to the discussion of content, reviewing and editing of the manuscript before submission was also made by H.B., W.W.S., D.S.B. and D.T.J.

Corresponding authors

Correspondence to Jacob W. Vogel or Michael Ewers.

Ethics declarations

Competing interests

M.E. and N.F. receive research funding from Eli Lilly. M.E. is a consultant for Prevail Therapeutics.

Peer review

Peer review information

Nature Reviews Neuroscience thanks A. Fornito and B. Misic for their contribution to the peer review of this work.

Additional information

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

Glossary

Amnestic

Presenting with primary impairment in memory and memory-related processes.

APOE

The strongest genetic risk factor for sporadic Alzheimer disease with three common alleles (ε2, ε3 and ε4): ε4 is a risk factor and ε2 is protective against Alzheimer disease relative to the ε3 allele.

Diffusion-weighted imaging

(DWI). A type of structural MRI sequence that measures the motion of water molecules in the brain. The water in the brain diffuses more easily along the direction of the white matter fibres (myelinated axons) than perpendicular to them and diffusion anisotropy can be used to reconstruct the anatomical connections.

Electrophysiology

A branch of physiology that studies the electrical properties of neurons and brain tissues by using electrodes placed directly on the exposed surface of the brain to record electrical activity from the cerebral cortex.

Functional MRI

(fMRI). A type of MRI sequence that measures changes in blood flow that occur with brain activity and can be used to identify the functional brain connections, which are obtained from the temporal correlations in the activity between pairs of grey matter regions.

Graph theory

A mathematical system used to study pairwise relationships between objects. In graphs, objects are represented as ‘nodes’, whereas their relationships are represented as ‘edges’.

Neurodegenerative diseases

A disorder characterized by the accumulation of pathological protein aggregates, which result in progressive damage to neurons and brain connections that are essential for cognition and/or sensorimotor functions.

Overfitting

An undesirable situation when a mathematical model gives accurate predictions of data that were used to train it, but not of new, unseen data.

Positron emission tomography

(PET). A type of molecular imaging technique that uses radiotracers on a fluorine radioisotope, which cross the blood–brain barrier and can bind to neurons that show abnormal pathological processes such as hypometabolism, tau accumulation or amyloid deposition.

Structural MRI

(sMRI). The most common non-invasive neuroimaging modality used to quantify grey and white matter atrophy in neurodegenerative disorders through the use of strong magnetic fields that produce contrast between different brain tissue classes.

Synucleinopathies

A class of neurodegenerative disorders characterized by the abnormal accumulation of α-synuclein aggregates in the brain, which can lead to the development of inclusions called Lewy bodies inside the neurons. Parkinson disease and dementia with Lewy bodies are the most common forms of synucleinopathies.

Tauopathy

A class of neurodegenerative diseases that involve the abnormal aggregation of tau into neurofibrillary tangles in the brain. Depending on the major tau isoforms appearing in the aggregates, tauopathies can be classified into groups on the basis of the number of repeats (R): 3R tauopathies (for example, Pick disease), 4R tauopathies (for example, progressive supranuclear palsy) and mixed 3R/4R tauopathies (for example, Alzheimer disease).

Rights and permissions

Springer Nature or its licensor (e.g. a society or other partner) holds exclusive rights to this article under a publishing agreement with the author(s) or other rightsholder(s); author self-archiving of the accepted manuscript version of this article is solely governed by the terms of such publishing agreement and applicable law.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Vogel, J.W., Corriveau-Lecavalier, N., Franzmeier, N. et al. Connectome-based modelling of neurodegenerative diseases: towards precision medicine and mechanistic insight. Nat. Rev. Neurosci. 24, 620–639 (2023). https://doi.org/10.1038/s41583-023-00731-8

Download citation

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41583-023-00731-8

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