Regional pattern of beta-amyloid accumulation in the preclinical and clinical states of Alzheimer’s disease

Cover Page


Cite item

Full Text

Abstract

Amyloid- (A ) plaque accumulation in the brain is a hallmark of Alzheimer’s disease (AD). The concept of preclinical AD implies that A deposits may accumulate in the brain years prior to the clinical manifestations of AD. In this study, we measured binding potentials (BP) of different brain regions using positron emission tomography (PET) study with A radiotracer N-methyl-[11C]2-(4ґ-methylaminophenyl)-6-hydroxybenzothiazole ([11C]PIB) in 16 patients with mild to moderate dementia of Alzheimer’s type (DAT) and 223 cognitively normal individuals aged 50 to 86 years old. Mean cortical BP was calculated from binding potentials of brain regions prone to A accumulation and was used as a measure to define threshold value for abnormal elevation of [11C]PIB uptake in cognitively normal individuals. In both groups, with low (n=181) or high (n = 42) A accumulation, the highest [11C]PIB BP was demonstrated in the precuneus. In DAT patients, A accumulation was substantially increased in all regions, with the precuneus and prefrontal cortex having the highest [11C] PIB BP. We suggest that the precuneus may be brain region with the earliest involvement in the A plaque accumulation.

 

About the authors

A. G. Vlassenko

Departments of Radiology and Neurology and the Knight Alzheimer’s Disease Research Center, Washington University School of Medicine

Email: andrei@npg.wustl.edu
United States, St. Louis, MO, USA

J. C. Morris

Departments of Radiology and Neurology and the Knight Alzheimer’s Disease Research Center, Washington University School of Medicine

Email: andrei@npg.wustl.edu
United States, St. Louis

M. A. Minton

Departments of Radiology and Neurology and the Knight Alzheimer’s Disease Research Center, Washington University School of Medicine

Author for correspondence.
Email: andrei@npg.wustl.edu
United States, St. Louis

References

  1. Agdeppa E.D., Kepe V., Liu J. et al. Binding characteristics of radiofluorinated 6-dialkylamino-2-naphthylethylidene derivatives as positron emission tomography imaging probes for beta-amyloid plaques in Alzheimer’s disease. J. Neurosci. 2001; 21: RC189.
  2. Berg L. Clinical Dementia Rating (CDR). Psychopharmacol. Bull. 1988; 24: 637–639.
  3. Braak H., Braak Е. Neuropathological stageing of Alzheimer-related changes. Acta Neuropathol. (Berl.) 1991; 82: 239–259.
  4. Bradley K.M., O’Sullivan V.T., Soper N.D. et al. Cerebral perfusion SPET correlated with Braak pathological stage in Alzheimer’s disease. Brain 2002; 125: 1772–1781.
  5. Cavanna A.E., Trimble M.R. The precuneus: a review of its functional anatomy and behavioral correlates. Brain 2006; 129: 564–583.
  6. Corder E.H., Saunders A.M., Strittmatter W.J. et al. Gene dose of apolipoprotein E type 4 allele and the risk of Alzheimer’s disease in late onset families. Science 1993; 261: 921–923.
  7. Dickerson B.C. New frontiers in computational analysis of human hippocampal anatomy. Hippocampus 2009; 19: 507–509.
  8. Edison P., Archer H.A., Hinz R. et al. Amyloid, hypometabolism, and cognition in Alzheimer disease. An [11C]PIB and [18F]FDG PET study. Neurology 2007; 68: 501–508.
  9. Fagan A.M., Mintun M.A., Mach R.H. et al. Inverse relation between in vivo amyloid imaging load and cerebrospinal fluid Abeta42 in humans. Ann. Neurol. 2006; 59: 512–519.
  10. Hardy J., Allsop D. Amyloid deposition as the central event in the aetiology of Alzheimer’s disease. Trends Pharmacol. Sci. 1991; 12: 383–388.
  11. Hardy J., Duff K., Hardy K.G. et al. Genetic dissection of Alzheimer’s disease and related dementias: amyloid and its relationship to tau. Nat. Neurosci. 1998; 1: 355–358.
  12. Hardy J., Selkoe D.J. The amyloid hypothesis of Alzheimer’s disease: progress and problems on the road to therapeutics. Science 2002; 297: 353–356.
  13. Hardy J.A., Higgins G.A. Alzheimer’s disease: the amyloid cascade hypothesis. Science 1992; 256: 184–185.
  14. Hedden T., Van Dijk K.R., Becker J.A. et al. Disruption of functional connectivity in clinically normal older adults harboring amyloid burden. J. Neurosci. 2009; 29: 12686–12694.
  15. Huang C., Wahlund L.O., Svensson L. et al. Cingulate cortex hypoperfusion predicts Alzheimer’s disease in mild cognitive impairment. BMC Neurol. 2002; 2: 9.
  16. Hulette C.M., Welsh-Bohmer K.A., Murray M.G. et al. Neuropathological and neuropsychological changes in «normal» aging: evidence for preclinical Alzheimer disease in cognitively normal individuals. J. Neuropathol. Exp. Neurol. 1998; 57: 1168–1174.
  17. Ikonomovic M.D., Klunk W.E., Abrahamson E.E. et al. Post-mortem correlates of in vivo PiB-PET amyloid imaging in a typical case of Alzheimer’s disease. Brain 2008; 131: 1630–1645.
  18. Kemp P.M., Hoffmann S.A., Holmes C. et al. The contribution of statistical parametric mapping in the assessment of precuneal and medial temporal lobe perfusion by 99mTc-HMPAO SPECT in mild Alzheimer’s and Lewy body dementia. Nucl. Med. Commun. 2005; 26: 1099–1106.
  19. Klunk W.E., Engler H., Nordberg A. et al. Imaing brain amyloid in Alzheimer’s disease with Pittsburgh Compound-B. Ann. Neurol. 2004; 55: 306–319.
  20. Kogure D., Matsuda H., Ohnishi T. et al. Longitudinal evaluation of early Alzheimer’s disease using brain perfusion SPECT. J. Nucl. Med. 2000; 41: 1155–1162.
  21. Leinonen V., Alafuzoff I., Aalto S. et al. Assessment of beta-amyloid in a frontal cortical brain biopsy specimen and by positron emission tomography with carbon 11-labeled Pittsburgh Compound B. Arch. Neurol. 2008; 65: 1304–1309.
  22. Logan J., Fowler J.S., Volkow N.D. et al. Graphical analysis of reversible radioligand binding from time-activity measurements applied to [N-11C-methyl]-(-)-cocaine PET studies in human subjects. J. Cereb. Blood Flow Metab. 1990; 10: 740–747.
  23. Logan J., Fowler J.S., Volkow N.D. et al. Distribution volume ratios without blood sampling from graphical analysis of PET data. J. Cereb. Blood Flow Metab. 1996; 16: 834–840.
  24. Lustig C., Snyder A.Z., Bhakta M. et al. Functional deactivations: change with age and dementia of the Alzheimer type. Proc. Natl. Acad. Sci. U.S.A 2003; 100: 14504–14509.
  25. Minoshima S., Giordani B., Berent S. et al. Metabolic reduction in the posterior cingulate cortex in very early Alzheimer’s disease. Ann. Neurol. 1997; 42: 85–94.
  26. Mintun M.A., Larossa G.N., Sheline Y.I. et al. [11C]PIB in a nondemented population: potential antecedent marker of Alzheimer disease. Neurology 2006; 67: 446–452.
  27. Morris J.C. The Clinical Dementia Rating (CDR): current version and scoring rules. Neurology 1993; 43: 2412–2414.
  28. Morris J.C., Price A.L. Pathologic correlates of nondemented aging, mild cognitive impairment, and early-stage Alzheimer’s disease. J. Mol. Neurosci. 2001; 17: 101–118.
  29. Morris J.C., Roe C.M., Grant E.A. et al. Pittsburgh compound B imaging and prediction of progression from cognitive normality to symptomatic Alzheimer disease. Arch Neurol. 2009; 66: 1469–1475.
  30. Morris J.C., Roe C.M., Xiong C. et al. APOE predicts amyloid-beta but not tau Alzheimer pathology in cognitively normal aging. Ann. Neurol. 2010; 67: 122–131.
  31. Price D.L., Sisodia S.S. Mutant genes in familial Alzheimer’s disease and transgenic models. Annu. Rev. Neurosci. 1998; 21: 479–505.
  32. Price J.C., Klunk W.E., Lopresti B.J. et al. Kinetic modeling of amyloid binding in humans using PET imaging and Pittsburgh Compound- B. J. Cereb. Blood Flow Metab. 2005; 25: 1528–1547.
  33. Raichle M.E., MacLeod A.M., Snyder A.Z. et al. A default mode of brain function. Proc. Natl. Acad. Sci USA 2001; 98: 676–682.
  34. Reiman E.M., Caselli R.J., Yun L.S. et al. Preclinical evidence of Alzheimer’s disease in persons homozygous for the epsilon 4 allele for apolipoprotein E. N. Engl. J. Med. 1996; 334: 752–758.
  35. Sakamoto S., Matsuda H., Asada T. et al. Apolipoprotein E genotype and early Alzheimer’s disease: a longitudinal SPECT study. J. Neuroimaging 2003; 13: 113–123.
  36. Salmon E., Collette F., Degueldre C. et al. Voxel-based analysis of confounding effects of age and dementia severity on cerebral metabolism in Alzheimer’s disease. Hum. Brain Mapp. 2000; 10: 39–48.
  37. Selemon L.D., Goldman-Rakic P.S. Common cortical and subcortical targets of the dorsolateral prefrontal and posterior parietal cortices in the rhesus monkey: evidence for a distributed neural network subserving spatially guided behavior. J. Neurosci. 1988; 8: 4049–4068.
  38. Selkoe D.J. Soluble oligomers of the amyloid beta-protein impair synaptic plasticity and behavior. Behav. Brain Res. 2008; 192: 106–113.
  39. Sheline Y.I., Raichle M.E., Snyder A.Z. et al. Amyloid plaques disrupt resting state default mode network connectivity in cognitively normal elderly. Biol. Psychiatry 2010; 67: 584–587.
  40. Shulman G.L., Fiez J.A., Corbetta M. et al. Common blood flow changes across visual tasks: II. Decreases in cerebral cortex. J. Cognit. Neurosci. 1997; 9: 648–663.
  41. Small G.W., Ercoli L.M., Silverman D.H. et al. Cerebral metabolic and cognitive decline in persons at genetic risk for Alzheimer’s disease. Proc. Natl. Acad. Sci. USA 2000; 97: 6037–6042.
  42. Small S.A., Duff K. Linking Abeta and tau in late-onset Alzheimer’s disease: a dual pathway hypothesis. Neuron 2008; 60: 534–542.
  43. Storandt M., Mintun M.A, Head D., Morris J.C. Cognitive decline and brain volume loss as signatures of cerebral amyloid-beta peptide deposition identified with Pittsburgh compound B: cognitive decline associated with Abeta deposition. Arch. Neurol. 2009; 66: 1476–1481.
  44. Tanzi R.E., Kovacs D.M., Kim T.W. et al. The gene defects responsible for familial Alzheimer’s disease. Neurobiol. Dis. 1996; 3: 159–168.
  45. Tomlinson B.E., Blessed G., Roth M. Observations on the brains of non-demented old people. J. Neurol. Sci. 1968; 7: 331–356.
  46. Verhoef N., Wilson A.A., Takeshita S. et al. In vivo imaging of Alzheimer disease β-amyloid with [ 1 1 C]SB-13 PET. Am. J. Geriatr. Psychiatry 2004; 12: 584–595.
  47. Vaishnavi S.N., Vlassenko A.G., Rundle M.M. et al. Regional aerobic glycolysis in the human brain. [published online ahead of print September 13 2010] Proc. Natl. Acad. Sci. USA. www.pnas.org/cgi/doi/10.1073/pnas.1010459107.
  48. Vlassenko A.G., Vaishnavi N.S., Couture L. et al. Spatial correlation between brain aerobic glycolysis and A deposition. [published online ahead of print September 13 2010] Proc. Natl. Acad. Sci. USA www.pnas.org/cgi/doi/10.1073/pnas.1010461107.
  49. Wong D.F., Rosenberg P.B., Zhou Y. et al. In vivo imaging of amyloid deposition in Alzheimer disease using the radioligand 18F-AV-45 (florbetapir [corrected] F 18). J. Nucl. Med. 2010; 51: 913–920.

Supplementary files

Supplementary Files
Action
1. JATS XML

Copyright (c) 2010 Vlassenko A.G., Morris J.C., Minton M.A.

Creative Commons License
This work is licensed under a Creative Commons Attribution 4.0 International License.

СМИ зарегистрировано Федеральной службой по надзору в сфере связи, информационных технологий и массовых коммуникаций (Роскомнадзор).
Регистрационный номер и дата принятия решения о регистрации СМИ: серия ПИ № ФС 77-83204 от 12.05.2022.


This website uses cookies

You consent to our cookies if you continue to use our website.

About Cookies