Elsevier

NeuroImage

Volume 54, Supplement 1, January 2011, Pages S204-S217
NeuroImage

Identification of amyloid plaques in retinas from Alzheimer's patients and noninvasive in vivo optical imaging of retinal plaques in a mouse model

https://doi.org/10.1016/j.neuroimage.2010.06.020Get rights and content

Abstract

Noninvasive monitoring of β-amyloid (Aβ) plaques, the neuropathological hallmarks of Alzheimer's disease (AD), is critical for AD diagnosis and prognosis. Current visualization of Aβ plaques in brains of live patients and animal models is limited in specificity and resolution. The retina as an extension of the brain presents an appealing target for a live, noninvasive optical imaging of AD if disease pathology is manifested there. We identified retinal Aβ plaques in postmortem eyes from AD patients (n = 8) and in suspected early stage cases (n = 5), consistent with brain pathology and clinical reports; plaques were undetectable in age-matched non-AD individuals (n = 5). In APPSWE/PS1∆E9 transgenic mice (AD-Tg; n = 18) but not in non-Tg wt mice (n = 10), retinal Aβ plaques were detected following systemic administration of curcumin, a safe plaque-labeling fluorochrome. Moreover, retinal plaques were detectable earlier than in the brain and accumulated with disease progression. An immune-based therapy effective in reducing brain plaques, significantly reduced retinal Aβ plaque burden in immunized versus non-immunized AD mice (n = 4 mice per group). In live AD-Tg mice (n = 24), systemic administration of curcumin allowed noninvasive optical imaging of retinal Aβ plaques in vivo with high resolution and specificity; plaques were undetectable in non-Tg wt mice (n = 11). Our discovery of Aβ specific plaques in retinas from AD patients, and the ability to noninvasively detect individual retinal plaques in live AD mice establish the basis for developing high-resolution optical imaging for early AD diagnosis, prognosis assessment and response to therapies.

Introduction

Alzheimer's disease (AD) is a common and devastating age-dependent neurodegenerative condition. At present, a definite diagnosis of AD is determined after brain autopsy by detecting accumulation of the hallmark proteolytic products of amyloid precursor protein (APP), β-amyloid peptides (Aβ), which form extracellular aggregates termed Aβ plaques, and by the presence of intracellular neurofibrillary tangles (Hardy & Selkoe, 2002, Sisodia & Price, 1995). Aβ plaques are believed to contribute to disrupted cellular activities and communication in the brain, leading to neurotoxic inflammation and neuronal death (McGeer & McGeer, 2002, Wyss-Coray, 2006). Major efforts have been invested in developing tools to enable noninvasive detection of amyloid plaques (e.g., through the skull) in living AD patients and animal models (Hintersteiner et al., 2005, Klunk et al., 2004, Nakada et al., 2008, Ng et al., 2007). However, such noninvasive monitoring of Aβ plaques is still clinically challenging, and is of limited resolution and availability (Klunk et al., 2005, Lockhart et al., 2007, Toyama et al., 2005). Optical detection constitutes a powerful, high-resolution and specific tool for in vivo imaging (Fujimoto and Farkas, 2009), as demonstrated using multiphoton microscopy to detect Aβ plaques in the mouse brain via an invasive cranial window (Meyer-Luehmann et al., 2008).

An alternative noninvasive approach to visualize amyloid plaques in AD patients at high resolution could be a direct optical imaging of the retina, provided that Aβ plaques are formed in patients’ retinas and share properties with those in the brain. Choosing the retina as a target for visualization of brain disease also relies on it being a direct extension of the brain, having the potential to faithfully reflect AD brain's pathology. Retinal abnormalities in AD patients, some of which were detected at early stages, have been described in the past (Berisha et al., 2007, Blanks et al., 1996, Hinton et al., 1986, Katz & Rimmer, 1989, Sadun et al., 1987, Trick et al., 1989). These changes, however, mostly related to the nerve fiber layer (NFL) thickness reduction, seem to appear in additional pathologies of the eye and the brain including ocular hypertension, glaucoma, demyelinating optic neuritis, multiple sclerosis, and Parkinson's disease (Jindahra et al., 2010, Parisi, 2003). Detection of ganglion cell death in retinas of live AD mouse models has been recently documented (Cordeiro et al., 2010); however, this phenomenon is common to multiple neurodegenerative disorders including glaucoma and age-related macular degeneration. In addition, postmortem lenses from AD patients were reported to exhibit specific cytosolic nanometer-size Aβ aggregates and supranuclear cataracts in the peripheral lens (Goldstein et al., 2003). Encouraging results in APPSWE and Presenilin (PS) 1∆E9 transgenic mice carrying the human mutated genes causing an early-onset familial AD were recently reported: these mice were found to develop human Aβ deposits in their retinal tissues at advanced stages of the disease (Ning et al., 2008, Perez et al., 2009).

The existing reports emphasized the need for unequivocal identification of Aβ plaque hallmark pathology of Alzheimer's disease in AD patients, especially at an early stage, for noninvasive in vivo detection of retinal plaques, and demonstration of their response to therapeutic intervention. Here, we report the presence of Aβ plaques in retinas of postmortem eyes from AD patients, and moreover, in retinas from those suspected as early stage cases. Using the APPSWE/PS1∆E9 transgenic (AD-Tg) mice, we provide evidence for the formation of Aβ plaques in the retina prior to their manifestation in the brain. Further, in these mice we found that an immune-based therapy, using a weak agonist of a myelin-derived peptide loaded onto dendritic cells (Koronyo-Hamaoui et al., 2009), was effective in reducing Aβ plaques in the retinas to an extent similar to that observed in the brain. Importantly, we were able to demonstrate that systemic injection of curcumin (diferuloylmethane), a natural and safe fluorochrome that binds and labels Aβ plaques (Garcia-Alloza et al., 2006, Yang et al., 2005), into live AD mice allows high-resolution and specific noninvasive in vivo visualization of retinal Aβ plaques.

Section snippets

Mice

Double transgenic mice (females and males in equal numbers) harboring the chimeric mouse/human APP (APPSWE) and mutant human presenilin 1 (PS1∆E9) genes, and their aged-matched wt littermates, were purchased from the Jackson Laboratories (Bar Harbor, ME, strain #4462). Both APPSWE and PS1∆E9 mutations are associated with early-onset Alzheimer's disease, and their expression is directed to CNS neurons. The “humanized” APPSWE transgene allows the mice to produce and secrete a human Aβ peptide.

Curcumin binds to retinal Aβ plaques in a mouse model of Alzheimer's disease

We first assessed the potential of using curcumin for the development of an in vivo noninvasive method to detect retinal Aβ plaques. To this end, we first verified that curcumin could specifically ex vivo label Aβ plaques in the retinas of AD-Tg mice. Retinal whole-mounts and cross-sections from AD-Tg and non-Tg (wt) mice were co-stained with curcumin and four different anti-Aβ mAbs (Fig. 1). Retinal Aβ plaques, co-labeled with curcumin and 11A5-B10 or 12F4 mAbs [recognizing the C-terminal

Discussion

The present study demonstrates that a noninvasive in vivo monitoring of AD hallmark pathology via optical imaging with high specificity and resolution is feasible through the retina. Two key issues towards translation of this approach for the detection of Alzheimer's in humans have been accomplished here: in vivo imaging of Aβ plaques in the retina of live AD animals, and identification of amyloid plaques in retinas from human Alzheimer's disease patients, even before clinical symptoms allow

Conclusions

This study provides the first demonstration of Aβ plaques in postmortem retinas from suspected and definite AD patients that reflected AD brain pathology. In addition, systemic administration of curcumin to AD mice resulted in specific in vivo labeling of retinal Aβ plaques. This novel approach enabled noninvasive and high-resolution monitoring of individual retinal Aβ plaques in live AD mice. Finally, curcumin-visualized retinal plaques were shown to decrease in number and size following

Competing interests statement

The authors declare that no conflict of interest exists.

Acknowledgments

We thank Drs. J.Y. Hwang, Y. Kohanzadeh, A.G. Nowatzyk, K.V. Ramanujan and K. Wawrowsky for useful discussions and imaging collaboration. M.S. holds the Maurice and Ilse Katz Professorial Chair in Neuroimmunology at The Weizmann Institute of Science, Israel. This work was supported in part by the Marciano Family Foundation, the U.S. Navy Bureau of Medicine and Surgery, R01 EY13431 and M01 RR00425, the Winnick Family Foundation, and the University of Southern California Alzheimer's Disease

References (50)

  • D.J. Selkoe

    Soluble oligomers of the amyloid β-protein impair synaptic plasticity and behavior

    Behav. Brain Res.

    (2008)
  • E.S. Wachman et al.

    AOTF microscope for imaging with increased speed and spectral versatility

    Biophys. J.

    (1997)
  • F. Yang et al.

    Curcumin inhibits formation of amyloid β oligomers and fibrils, binds plaques, and reduces amyloid in vivo

    J. Biol. Chem.

    (2005)
  • P. Anand et al.

    Bioavailability of curcumin: problems and promises

    Mol. Pharm.

    (2007)
  • R.A. Armstrong

    β-Amyloid plaques: stages in life history or independent origin?

    Dement. Geriatr. Cogn. Disord.

    (1998)
  • W. Baschong et al.

    Control of autofluorescence of archival formaldehyde-fixed, paraffin-embedded tissue in confocal laser scanning microscopy (CLSM)

    J. Histochem. Cytochem.

    (2001)
  • F. Berisha et al.

    Retinal abnormalities in early Alzheimer's disease

    Invest. Ophthalmol. Vis. Sci.

    (2007)
  • Burton, K., Jeong, J., Wachsmann-Hogiu, S., Farkas, D.L., 2009. Spectral optical imaging in biology and medicine. In:...
  • M.F. Cordeiro et al.

    Imaging multiple phases of neurodegeneration: a novel approach to assessing cell death in vivo

    Cell Death Dis.

    (2010)
  • N. Dhillon et al.

    Phase II trial of curcumin in patients with advanced pancreatic cancer

    Clin. Cancer Res.

    (2008)
  • M.L. Ford et al.

    An MHC anchor-substituted analog of myelin oligodendrocyte glycoprotein 35–55 induces IFN-gamma and autoantibodies in the absence of experimental autoimmune encephalomyelitis and optic neuritis

    Eur. J. Immunol.

    (2004)
  • M. Garcia-Alloza et al.

    Curcumin labels amyloid pathology in vivo, disrupts existing plaques, and partially restores distorted neurites in an Alzheimer mouse model

    J. Neurochem.

    (2007)
  • L. Guo et al.

    Targeting amyloid-β in glaucoma treatment

    Proc. Natl. Acad. Sci. U. S. A.

    (2007)
  • J. Hardy et al.

    The amyloid hypothesis of Alzheimer's disease: progress and problems on the road to therapeutics

    Science

    (2002)
  • Cited by (498)

    View all citing articles on Scopus
    1

    These authors contributed equally to this study.

    2

    These authors contributed equally to this study.

    View full text