Skip to main content

Advertisement

Log in

Current advances in neuronal intranuclear inclusion disease

  • Review Article
  • Published:
Neurological Sciences Aims and scope Submit manuscript

Abstract

Neuronal intranuclear inclusion disease (NIID) is a rare but probably underdiagnosed neurodegenerative disorder due to pathogenic GGC expansions in the NOTCH2NLC gene. In this review, we summarize recent developments in the inheritance features, pathogenesis, and histopathologic and radiologic features of NIID that subvert the previous perceptions of NIID. GGC repeat sizes determine the age of onset and clinical phenotypes of NIID patients. Anticipation may be absent in NIID but paternal bias is observed in NIID pedigrees. Eosinophilic intranuclear inclusions in skin tissues once considered pathological hallmarks of NIID can also present in other GGC repeat diseases. Diffusion-weighted imaging (DWI) hyperintensity along the corticomedullary junction once considered the imaging hallmark of NIID can frequently be absent in muscle weakness and parkinsonism phenotype of NIID. Besides, DWI abnormalities can appear years after the onset of predominant symptoms and may even disappear completely with disease progression. Moreover, continuous reports of NOTCH2NLC GGC expansions in patients with other neurodegenerative diseases lead to the proposal of a new concept of NOTCH2NLC-related GGC repeat expansion disorders (NRED). However, by reviewing the previous literature, we point out the limitations of these studies and provide evidence that these patients are actually suffering from neurodegenerative phenotypes of NIID.

This is a preview of subscription content, log in via an institution to check access.

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Fig. 1

Similar content being viewed by others

Data Availability

The authors confirm that the data supporting the findings of this study are available within the article and its supplementary materials.

References

  1. Lindenberg R, Rubinstein LJ, Herman MM, Haydon GB (1968) A light and electron microscopy study of an unusual widespread nuclear inclusion body disease. A possible residuum of an old herpesvirus infection. Acta Neuropathol 10(1):54–73. https://doi.org/10.1007/BF00690510

    Article  CAS  PubMed  Google Scholar 

  2. Sone J, Tanaka F, Koike H, Inukai A, Katsuno M, Yoshida M et al (2011) Skin biopsy is useful for the antemortem diagnosis of neuronal intranuclear inclusion disease. Neurology 76(16):1372–6. https://doi.org/10.1212/WNL.0b013e3182166e13

    Article  CAS  Google Scholar 

  3. Sone J, Kitagawa N, Sugawara E, Iguchi M, Nakamura R, Koike H et al (2014) Neuronal intranuclear inclusion disease cases with leukoencephalopathy diagnosed via skin biopsy. J Neurol Neurosurg Psychiatr 85(3):354–6. https://doi.org/10.1136/jnnp-2013-306084

    Article  Google Scholar 

  4. Ishiura H, Shibata S, Yoshimura J, Suzuki Y, Qu W, Doi K et al (2019) Noncoding CGG repeat expansions in neuronal intranuclear inclusion disease, oculopharyngodistal myopathy and an overlapping disease. Nat Genet 51(8):1222–32. https://doi.org/10.1038/s41588-019-0458-z

    Article  CAS  PubMed  Google Scholar 

  5. Huang XR, Tang BS, Jin P, Guo JF (2022) The phenotypes and mechanisms of NOTCH2NLC-related GGC repeat expansion disorders: a comprehensive review. Mol Neurobiol 59(1):523–34. https://doi.org/10.1007/s12035-021-02616-2

    Article  CAS  PubMed  Google Scholar 

  6. Pearson CE, Nichol Edamura K, Cleary JD (2005) Repeat instability: mechanisms of dynamic mutations. Nat Rev Genet 6(10):729–742. https://doi.org/10.1038/nrg1689

    Article  CAS  PubMed  Google Scholar 

  7. Malik I, Kelley CP, Wang ET, Todd PK (2021) Molecular mechanisms underlying nucleotide repeat expansion disorders. Nat Rev Mol Cell Biol 22(9):589–607. https://doi.org/10.1038/s41580-021-00382-6

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Paulson H (2018) Repeat expansion diseases. Handb Clin Neurol 147:105–123. https://doi.org/10.1016/B978-0-444-63233-3.00009-9

    Article  PubMed  PubMed Central  Google Scholar 

  9. Tian Y, Wang JL, Huang W, Zeng S, Jiao B, Liu Z et al (2019) Expansion of human-specific GGC repeat in neuronal intranuclear inclusion disease-related disorders. Am J Hum Genet 105(1):166–76. https://doi.org/10.1016/j.ajhg.2019.05.013

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Deng J, Zhou B, Yu J, Han X, Fu J, Li X et al (2022) Genetic origin of sporadic cases and RNA toxicity in neuronal intranuclear inclusion disease. J Med Genet 59(5):462–9. https://doi.org/10.1136/jmedgenet-2020-107649

    Article  CAS  PubMed  Google Scholar 

  11. Tian Y, Zhou L, Gao J, Jiao B, Zhang SZ, Xiao Q et al (2022) Clinical features of NOTCH2NLC-related neuronal intranuclear inclusion disease. J Neurol Neurosur Ps. https://doi.org/10.1136/jnnp-2022-329772

    Article  Google Scholar 

  12. Cabal-Herrera AM, Tassanakijpanich N, Salcedo-Arellano MJ, Hagerman RJ (2020) Fragile X-associated tremor/ataxia syndrome (FXTAS): pathophysiology and clinical implications. Int J Mol Sci 21(12):4391. https://doi.org/10.3390/ijms21124391

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Jiao B, Zhou L, Zhou Y, Weng L, Liao X, Tian Y et al (2020) Identification of expanded repeats in NOTCH2NLC in neurodegenerative dementias. Neurobiol Aging 89:142e1-e7. https://doi.org/10.1016/j.neurobiolaging.2020.01.010

    Article  CAS  Google Scholar 

  14. Ma D, Tan YJ, Ng ASL, Ong HL, Sim W, Lim WK et al (2020) Association of NOTCH2NLC repeat expansions with Parkinson disease. JAMA Neurol 77(12):1559–1563. https://doi.org/10.1001/jamaneurol.2020.3023

    Article  PubMed  Google Scholar 

  15. Yuan Y, Liu Z, Hou X, Li W, Ni J, Huang L et al (2020) Identification of GGC repeat expansion in the NOTCH2NLC gene in amyotrophic lateral sclerosis. Neurology 95(24):e3394–e405. https://doi.org/10.1212/WNL.0000000000010945

    Article  CAS  Google Scholar 

  16. Shi CH, Fan Y, Yang J, Yuan YP, Shen S, Liu F et al (2021) NOTCH2NLC intermediate-length repeat expansions are associated with Parkinson disease. Ann Neurol 89(1):182–7. https://doi.org/10.1002/ana.25925

    Article  CAS  PubMed  Google Scholar 

  17. Chen H, Lu L, Wang B, Cui G, Wang X, Wang Y et al (2020) Re-defining the clinicopathological spectrum of neuronal intranuclear inclusion disease. Ann Clin Transl Neurol 7(10):1930–41. https://doi.org/10.1002/acn3.51189

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Fukuda H, Yamaguchi D, Nyquist K, Yabuki Y, Miyatake S, Uchiyama Y et al (2021) Father-to-offspring transmission of extremely long NOTCH2NLC repeat expansions with contractions: genetic and epigenetic profiling with long-read sequencing. Clin Epigenetics 13(1):204. https://doi.org/10.1186/s13148-021-01192-5

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Boivin M, Charlet-Berguerand N (2022) Trinucleotide CGG repeat diseases: an expanding field of polyglycine proteins? Front Genet 13:843014. https://doi.org/10.3389/fgene.2022.843014

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Ridley RM, Frith CD, Crow TJ, Conneally PM (1988) Anticipation in Huntington’s disease is inherited through the male line but may originate in the female. J Med Genet 25(9):589–595. https://doi.org/10.1136/jmg.25.9.589

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Orr HT, Chung MY, Banfi S, Kwiatkowski TJ Jr, Servadio A, Beaudet AL et al (1993) Expansion of an unstable trinucleotide CAG repeat in spinocerebellar ataxia type 1. Nat Genet 4(3):221–226. https://doi.org/10.1038/ng0793-221

    Article  CAS  PubMed  Google Scholar 

  22. Reyniers E, Vits L, De Boulle K, Van Roy B, Van Velzen D, de Graaff E et al (1993) The full mutation in the FMR-1 gene of male fragile X patients is absent in their sperm. Nat Genet 4(2):143–146. https://doi.org/10.1038/ng0693-143

    Article  CAS  PubMed  Google Scholar 

  23. Yu J, Deng J, Guo X, Shan J, Luan X, Cao L et al (2021) The GGC repeat expansion in NOTCH2NLC is associated with oculopharyngodistal myopathy type 3. Brain 144(6):1819–1832. https://doi.org/10.1093/brain/awab077

    Article  PubMed  PubMed Central  Google Scholar 

  24. Fiddes IT, Lodewijk GA, Mooring M, Bosworth CM, Ewing AD, Mantalas GL et al (2018) Human-specific NOTCH2NL genes affect Notch signaling and cortical neurogenesis. Cell. 173(6):1356–1369. https://doi.org/10.1016/j.cell.2018.03.051

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Sone J, Mitsuhashi S, Fujita A, Mizuguchi T, Hamanaka K, Mori K et al (2019) Long-read sequencing identifies GGC repeat expansions in NOTCH2NLC associated with neuronal intranuclear inclusion disease. Nat Genet 51(8):1215–1221. https://doi.org/10.1038/s41588-019-0459-y

    Article  CAS  PubMed  Google Scholar 

  26. Krzyzosiak WJ, Sobczak K, Wojciechowska M, Fiszer A, Mykowska A, Kozlowski P (2012) Triplet repeat RNA structure and its role as pathogenic agent and therapeutic target. Nucleic Acids Res 40(1):11–26. https://doi.org/10.1093/nar/gkr729

    Article  CAS  PubMed  Google Scholar 

  27. Zhong S, Lian Y, Luo W, Luo R, Wu X, Ji J et al (2021) Upstream open reading frame with NOTCH2NLC GGC expansion generates polyglycine aggregates and disrupts nucleocytoplasmic transport: implications for polyglycine diseases. Acta Neuropathol 142(6):1003–1023. https://doi.org/10.1007/s00401-021-02375-3

    Article  CAS  PubMed  Google Scholar 

  28. Yu J, Liufu T, Zheng Y, Xu J, Meng L, Zhang W et al (2022) CGG repeat expansion in NOTCH2NLC causes mitochondrial dysfunction and progressive neurodegeneration in Drosophila model. Proc Natl Acad Sci USA. 119(41):e2208649119. https://doi.org/10.1073/pnas.2208649119

  29. Liu Q, Zhang K, Kang Y, Li Y, Deng P, Li Y et al (2022) Expression of expanded GGC repeats within NOTCH2NLC causes behavioral deficits and neurodegeneration in a mouse model of neuronal intranuclear inclusion disease. Sci Adv. 8(47):eadd6391. https://doi.org/10.1126/sciadv.add6391

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Asamitsu S, Yabuki Y, Ikenoshita S, Kawakubo K, Kawasaki M, Usuki S et al (2021) CGG repeat RNA G-quadruplexes interact with FMRpolyG to cause neuronal dysfunction in fragile X-related tremor/ataxia syndrome. Sci Adv 7(3):eabd9440. https://doi.org/10.1126/sciadv.abd9440

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Sone J, Mori K, Inagaki T, Katsumata R, Takagi S, Yokoi S et al (2016) Clinicopathological features of adult-onset neuronal intranuclear inclusion disease. Brain 139(Pt 12):3170–86. https://doi.org/10.1093/brain/aww249

    Article  PubMed  PubMed Central  Google Scholar 

  32. Hunsaker MR, Greco CM, Spath MA, Smits AP, Navarro CS, Tassone F et al (2011) Widespread non-central nervous system organ pathology in fragile X premutation carriers with fragile X-associated tremor/ataxia syndrome and CGG knock-in mice. Acta Neuropathol 122(4):467–79. https://doi.org/10.1007/s00401-011-0860-9

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Gokden M, Al-Hinti JT, Harik SI (2009) Peripheral nervous system pathology in fragile X tremor/ataxia syndrome (FXTAS). Neuropathology 29(3):280–4. https://doi.org/10.1111/j.1440-1789.2008.00948.x

    Article  Google Scholar 

  34. Toko M, Ohshita T, Kurashige T, Morino H, Kume K, Yamashita H et al (2021) FXTAS is difficult to differentiate from neuronal intranuclear inclusion disease through skin biopsy: a case report. BMC Neurol 21(1):396. https://doi.org/10.1186/s12883-021-02425-z

    Article  PubMed  PubMed Central  Google Scholar 

  35. Ogasawara M, Eura N, Nagaoka U, Sato T, Arahata H, Hayashi T et al (2022) Intranuclear inclusions in skin biopsies are not limited to neuronal intranuclear inclusion disease but can also be seen in oculopharyngodistal myopathy. Neuropathol Appl Neurobiol 48(3):e12787. https://doi.org/10.1111/nan.12787

    Article  CAS  PubMed  Google Scholar 

  36. Ogasawara M, Eura N, Iida A, Kumutpongpanich T, Minami N, Nonaka I et al (2022) Intranuclear inclusions in muscle biopsy can differentiate oculopharyngodistal myopathy and oculopharyngeal muscular dystrophy. Acta Neuropathol Commun 10(1):176. https://doi.org/10.1186/s40478-022-01482-w

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Boivin M, Deng J, Pfister V, Grandgirard E, Oulad-Abdelghani M, Morlet B et al (2021) Translation of GGC repeat expansions into a toxic polyglycine protein in NIID defines a novel class of human genetic disorders: the polyG diseases. Neuron 109(11):1825-1835e5. https://doi.org/10.1016/j.neuron.2021.03.038

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Abe K, Fujita M (2017) Over 10 years MRI observation of a patient with neuronal intranuclear inclusion disease. BMJ Case Rep 2017. https://doi.org/10.1136/bcr-2016-218790

  39. Wang Y, Wang B, Wang L, Yao S, Zhao J, Zhong S et al (2020) Diagnostic indicators for adult-onset neuronal intranuclear inclusion disease. Clin Neuropathol 39(1):7–18. https://doi.org/10.5414/NP301203

    Article  CAS  PubMed  Google Scholar 

  40. Liu YH, Chou YT, Chang FP, Lee WJ, Guo YC, Chou CT et al (2022) Neuronal intranuclear inclusion disease in patients with adult-onset non-vascular leukoencephalopathy. Brain 145(9):3010–3021. https://doi.org/10.1093/brain/awac135

    Article  PubMed  Google Scholar 

  41. Yokoi S, Yasui K, Hasegawa Y, Niwa K, Noguchi Y, Tsuzuki T et al (2016) Pathological background of subcortical hyperintensities on diffusion-weighted images in a case of neuronal intranuclear inclusion disease. Clin Neuropathol 35(6):375–380. https://doi.org/10.5414/NP300961

    Article  PubMed  Google Scholar 

  42. Chen L, Wu L, Li S, Huang Q, Xiong J, Hong D et al (2018) A long time radiological follow-up of neuronal intranuclear inclusion disease: two case reports. Medicine (Baltimore) 97(49):e13544. https://doi.org/10.1097/MD.0000000000013544

    Article  PubMed  Google Scholar 

  43. Kawarabayashi T, Nakamura T, Seino Y, Hirohata M, Mori F, Wakabayashi K et al (2018) Disappearance of MRI imaging signals in a patient with neuronal intranuclear inclusion disease. J Neurol Sci 388:1–3. https://doi.org/10.1016/j.jns.2018.02.038

    Article  PubMed  Google Scholar 

  44. Eisenmenger L, Porter MC, Carswell CJ, Thompson A, Mead S, Rudge P et al (2016) Evolution of diffusion-weighted magnetic resonance imaging signal abnormality in sporadic Creutzfeldt-Jakob disease, with histopathological correlation. JAMA Neurol 73(1):76–84. https://doi.org/10.1001/jamaneurol.2015.3159

    Article  PubMed  PubMed Central  Google Scholar 

  45. Sugiyama A, Sato N, Kimura Y, Maekawa T, Enokizono M, Saito Y et al (2017) MR imaging features of the cerebellum in adult-onset neuronal intranuclear inclusion disease: 8 cases. AJNR Am J Neuroradiol 38(11):2100–4. https://doi.org/10.3174/ajnr.A5336

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Sun QY, Xu Q, Tian Y, Hu ZM, Qin LX, Yang JX et al (2020) Expansion of GGC repeat in the human-specific NOTCH2NLC gene is associated with essential tremor. Brain 143(1):222–233. https://doi.org/10.1093/brain/awz372

    Article  PubMed  Google Scholar 

  47. Liao YC, Chang FP, Huang HW, Chen TB, Chou YT, Hsu SL et al (2022) GGC repeat expansion of NOTCH2NLC in Taiwanese patients with inherited neuropathies. Neurology 98(2):e199–e206. https://doi.org/10.1212/WNL.0000000000013008

    Article  CAS  Google Scholar 

  48. Ogasawara M, Iida A, Kumutpongpanich T, Ozaki A, Oya Y, Konishi H et al (2020) CGG expansion in NOTCH2NLC is associated with oculopharyngodistal myopathy with neurological manifestations. Acta Neuropathol Commun 8(1):204. https://doi.org/10.1186/s40478-020-01084-4

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Westenberger A, Klein C (2020) Essential phenotypes of NOTCH2NLC-related repeat expansion disorder. Brain 143(1):5–8. https://doi.org/10.1093/brain/awz404

    Article  PubMed  Google Scholar 

  50. Ng ASL, Lim WK, Xu Z, Ong HL, Tan YJ, Sim WY et al (2020) NOTCH2NLC GGC repeat expansions are associated with sporadic essential tremor: variable disease expressivity on long-term follow-up. Ann Neurol 88(3):614–8. https://doi.org/10.1002/ana.25803

    Article  CAS  PubMed  Google Scholar 

  51. Chen H, Lu L, Wang B, Hua X, Wan B, Sun M et al (2020) Essential tremor as the early symptom of NOTCH2NLC gene-related repeat expansion disorder. Brain 143(7):e56. https://doi.org/10.1093/brain/awaa142

    Article  PubMed  Google Scholar 

  52. Yang D, Cen Z, Wang L, Chen X, Liu P, Wang H et al (2022) Neuronal intranuclear inclusion disease tremor-dominant subtype: a mimicker of essential tremor. Eur J Neurol 29(2):450–8. https://doi.org/10.1111/ene.15169

    Article  PubMed  Google Scholar 

  53. Vermilion J, Johnson M, Srinivasan J, Mink JW (2019) Neuronal intranuclear inclusion disease: longitudinal case report of motor and nonmotor symptoms. J Child Neurol 34(13):801–5. https://doi.org/10.1177/0883073819860566

    Article  PubMed  PubMed Central  Google Scholar 

  54. Wang ZY, Guo JJ, Wang M, Wang ZX, Hong DJ, Yu XF (2020) Adult-onset neuronal intranuclear inclusion disease mimicking Parkinson’s disease in a Chinese patient: a case report and literature reviews. Neuro Endocrinol Lett 41(4):155–161

    PubMed  Google Scholar 

  55. Paviour DC, Revesz T, Holton JL, Evans A, Olsson JE, Lees AJ (2005) Neuronal intranuclear inclusion disease: report on a case originally diagnosed as dopa-responsive dystonia with Lewy bodies. Mov Disord 20(10):1345–1349. https://doi.org/10.1002/mds.20559

    Article  PubMed  Google Scholar 

  56. Liu P, Yang D, Zhang F, Chen S, Xie F, Luo Y et al (2022) The role of NOTCH2NLC in Parkinson’s disease: a clinical, neuroimaging, and pathological study. Eur J Neurol 29(6):1610–8. https://doi.org/10.1111/ene.15283

    Article  PubMed  Google Scholar 

  57. Sugiyama A, Takeda T, Koide M, Yokota H, Mukai H, Kitayama Y et al (2021) Coexistence of neuronal intranuclear inclusion disease and amyotrophic lateral sclerosis: an autopsy case. BMC Neurol 21(1):273. https://doi.org/10.1186/s12883-021-02306-5

    Article  PubMed  PubMed Central  Google Scholar 

  58. Mann DM, Rollinson S, Robinson A, Bennion Callister J, Thompson JC, Snowden JS et al (2013) Dipeptide repeat proteins are present in the p62 positive inclusions in patients with frontotemporal lobar degeneration and motor neurone disease associated with expansions in C9ORF72. Acta Neuropathol Commun 1:68. https://doi.org/10.1186/2051-5960-1-68

    Article  PubMed  PubMed Central  Google Scholar 

  59. Lee SM, Asress S, Hales CM, Gearing M, Vizcarra JC, Fournier CN et al (2019) TDP-43 cytoplasmic inclusion formation is disrupted in C9orf72-associated amyotrophic lateral sclerosis/frontotemporal lobar degeneration. Brain Commun 1(1):fcz014. https://doi.org/10.1093/braincomms/fcz014

    Article  PubMed  PubMed Central  Google Scholar 

  60. Davidson JM, Chung RS, Lee A (2022) The converging roles of sequestosome-1/p62 in the molecular pathways of amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD). Neurobiol Dis 166:105653. https://doi.org/10.1016/j.nbd.2022.105653

    Article  CAS  PubMed  Google Scholar 

  61. Al-Sarraj S, King A, Troakes C, Smith B, Maekawa S, Bodi I et al (2011) p62 positive, TDP-43 negative, neuronal cytoplasmic and intranuclear inclusions in the cerebellum and hippocampus define the pathology of C9orf72-linked FTLD and MND/ALS. Acta Neuropathol 122(6):691–702. https://doi.org/10.1007/s00401-011-0911-2

    Article  CAS  PubMed  Google Scholar 

  62. Fan Y, Xu Y, Shi C (2022) NOTCH2NLC-related disorders: the widening spectrum and genotype-phenotype correlation. J Med Genet 59(1):1–9. https://doi.org/10.1136/jmedgenet-2021-107883

    Article  CAS  PubMed  Google Scholar 

  63. Sone J, Hishikawa N, Koike H, Hattori N, Hirayama M, Nagamatsu M et al (2005) Neuronal intranuclear hyaline inclusion disease showing motor-sensory and autonomic neuropathy. Neurology 65(10):1538–1543. https://doi.org/10.1212/01.wnl.0000184490.22527.90

    Article  CAS  Google Scholar 

Download references

Author information

Authors and Affiliations

Authors

Corresponding authors

Correspondence to Hao Chen or Guiyun Cui.

Ethics declarations

Ethical approval

None.

Conflict of interest

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

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

Bao, L., Zuo, D., Li, Q. et al. Current advances in neuronal intranuclear inclusion disease. Neurol Sci 44, 1881–1889 (2023). https://doi.org/10.1007/s10072-023-06677-0

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s10072-023-06677-0

Keywords

Navigation