Skip to main content
Erschienen in: Translational Neurodegeneration 1/2021

Open Access 01.12.2021 | Review

Advances in retina imaging as potential biomarkers for early diagnosis of Alzheimer’s disease

verfasst von: Ying Zhang, Yanjiang Wang, Ce Shi, Meixiao Shen, Fan Lu

Erschienen in: Translational Neurodegeneration | Ausgabe 1/2021

Abstract

As the most common form of dementia, Alzheimer’s disease (AD) is characterized by progressive cognitive impairments and constitutes a major social burden. Currently, the invasiveness and high costs of tests have limited the early detection and intervention of the disease. As a unique window of the brain, retinal changes can reflect the pathology of the brain. In this review, we summarize current understanding of retinal structures in AD, mild cognitive impairment (MCI) and preclinical AD, focusing on neurodegeneration and microvascular changes measured using optical coherence tomography (OCT) and optical coherence tomography angiography (OCTA) technologies. The literature suggests that the impairment of retinal microvascular network and neural microstructure exists in AD, MCI and even preclinical AD. These findings provide valuable insights into a better understanding of disease pathogenesis and demonstrate that retinal changes are potential biomarkers for early diagnosis of AD and monitoring of disease progression.
Abkürzungen
AD
Alzheimer’s disease
MCI
Mild cognitive impairment
OCT
Optical coherence tomography
OCTA
Optical coherence tomography angiography
MRI
Magnetic resonance imaging
CT
Computed tomography
RNFL
Retinal nerve fiber layer
GCL
Ganglion cell layer
IPL
Inner plexiform layer
INL
Inner nuclear layer
OPL
Outer plexiform layer
ONL
Outer nuclear layer
GC-IPL
Ganglion cell-inner plexiform layer
GCC
Ganglion cell complex
pRNFL
Peripapillary retinal nerve fiber layer
mRNFL
Macular retinal nerve fiber layer
VD
Vessel density
FD
Fractal dimension
FAZ
Foveal avascular zone
VLD
Vessel length density
RPC
Radial peripapillary capillary
SRCP
Superficial retinal capillary plexus
DRCP
Deep retinal capillary plexus
Amyloid-beta

Background

Alzheimer’s disease (AD) is a leading cause of dementia and the most common chronic neurodegenerative disease leading to cognitive impairment in the elderly. AD is pathologically characterized by abnormal extracellular senile plaques consisting of amyloid-beta (Aβ) peptide and intracellular neurofibrillary tangles composed of hyperphosphorylated tau protein. Other disease-specific signs include cerebral amyloid angiopathy resulting from amyloid deposition on microvascular walls, neuron and synaptic loss, inflammation, and gliosis [14]. It has been estimated that by 2050 up to 131 million people would suffer from AD [5]. Despite the advances in understanding the pathogenesis and clinical practice over the past decades, current clinical treatments are limited as most patients have advanced AD with irreversible neuronal damage. It is generally believed that early intervention can slow the progression of AD. Epidemiologic research has reported that one-year delay in the onset of AD would reduce the projected global burden by 9 million [6]. Therefore, preclinical detection of neurodegeneration will be crucial for preventing dementia caused by AD and developing new treatments.
Neuroimaging is the most widely used method for early diagnosis of neurodegenerative pathology of AD. With the advent of different neuroimaging technologies, quantitative assessment of cerebral structures and metabolic states in specific regions has become possible. Both magnetic resonance imaging (MRI) and computed tomography (CT) can identify cortical and hippocampal atrophy. In addition, Aβ deposits can be quantified by amyloid positron emission tomography [7, 8]. Besides, the cerebrospinal fluid test has been thought to be able to reflect the pathology of AD through decreased amyloid and increased tau levels [9, 10]. The above methods, however, are invasive, expensive or time-consuming, which may not be suitable for early large-scale screening of AD. Thus, there is an urgent need for an early, noninvasive and cost-efficient tool to identify AD biomarkers, which could help detect AD pathology in mild cognitive impairment (MCI) stage, even in asymptomatic preclinical AD stage. Such strategy could help predict those at a high risk of developing dementia in a large-scale population.
As a window to the brain, the retina provides a unique opportunity to study the pathophysiology of many ophthalmic and neurodegenerative diseases. A growing body of evidence has indicated that both the brain and the retina are affected in AD and these pathologic changes are significantly correlated [11]. Some evidence has also shown that the retinal nerve fiber layer (RNFL) thinning, which reflects the loss of retinal ganglion cells (RGCs), is significantly associated with brain atrophy [12, 13]. The optical coherence tomography (OCT) has enabled the imaging and quantification of RGCs and their axons.
In addition to the neurological defects, evidence has shown that the vascular factors also play a crucial role in the occurrence and development of AD [14, 15]. With the advancement of technology, the retinal microvasculature can be visualized and quantified non-invasively using optical coherence tomography angiography (OCTA). Compared to the traditional color fundus photography, OCTA has advantages of visualizing retinal microvasculature at the micrometer level. OCT and OCTA imaging of the retina are potential screening tools for early diagnosis of AD and identification of the risk of disease progression. In this review, we summarize recent development on the applications of OCT and OCTA as retinal imaging tools to study the pathophysiology of AD at different stages. We also discuss the clinical implications of these findings and what we can do in future research.

Why is the retina a window to the brain?

The retina originates from the neural tube as a part of the central nervous system during embryonic development [16]. The RNFL is mainly composed of axons, while the ganglion cell- inner plexiform layer (GC-IPL) mainly contains cell bodies and dendrites. In terms of the brain parenchyma, the white matter of the brain is mainly composed of axons, while the gray matter is mainly composed of cell bodies and dendrites of neurons. Research has demonstrated that thinning of the GC-IPL is significantly associated with the gray matter volume obtained from MRI scans, suggesting that the retinal ganglion cells are potential markers of cerebral neurodegeneration [17].
In terms of the retinal microvasculature around macula, the inner layer of retina is supplied by the ophthalmic artery through the central retinal artery. The ophthalmic artery originates from the internal carotid artery, similar to the intracranial artery. The external retina is supplied by choroid vessels. The blood supply of the optic disc is derived from the posterior ciliary artery circulation, with the retinal circulation supplying the surface nerve fiber layer [1821]. The retinal arterioles and venules, with diameters from 100 μm to 300 μm, have similar anatomical features and physiological properties with cerebral small vessels, which provides a unique and accessible “window” to study the subclinical microvascular pathology of the brain (Fig. 1).
As one of the main pathological features of AD, Aβ is not only associated with blood vessels but also occurs in the retinal neural structure. Researchers have discovered that the retinal plaques are detectable earlier than those in the brain and accumulate with disease progression in animal models [22]. Koronyo Y and colleagues [11] detected curcumin-labeled Aβ plaques in patients in vivo and showed that the retinal Aβ burden was quantitatively correlated with the amyloid burden in the brain. With regard to other pathological states, patients with raised intracranial pressure often develop optic disc edema, and fundus findings including papilledema and hemorrhage are common in patients with spontaneous subarachnoid hemorrhage [23]. A large standard deviation in retinal arterial width is associated with intracranial arterial stenosis, which indicates that the retinal vessels may provide insights into the cerebrovascular network [24].

Advances in imaging of retinal neural structure and microvasculature based on OCT and OCTA

OCT is an invasive imaging tool with advantages of high resolution, excellent repeatability, fast scanning speed, and high convenience, which can provide quantitative analyses of individual retinal layers, including the inner boundary membrane, RNFL, ganglion cell layer (GCL), inner plexiform layer (IPL), inner nuclear layer (INL), outer plexiform layer (OPL), outer nuclear layer (ONL), outer membrane, photoreceptor cell layer, and retinal pigment epithelium [25, 26]. The peripapillary RNFL (pRNFL), which converges to form the optic nerve, is composed of axons of ganglion cells and is thickest around the optic disc. The ganglion cells are mainly concentrated in the macular area. High-resolution spectral-domain OCT also allows measurement of the thickness of the pRNFL, the GCL, the GC-IPL, and the ganglion cell complex (GCC), which is composed of pRNFL and ganglion cell bodies and dendrites. The INL contains cell bodies of horizontal, bipolar and amacrine cells, and the ONL contains cell bodies of rods and cones. The two neuropils consisting of synaptic contacts divide the above nerve cell layers. The different degrees of light absorption and scattering in individual retinal layers serve as the basis for imaging. Compared with the conventional time-domain OCT devices, the spectral-domain OCT technology offers faster scanning speed and higher axial resolutions. Recently, swept-source OCT has been shown to be more accurate in quantitative segmented retinal layer analyses with increased speed and resolution. The current equipment and built-in software have enabled us to assess the thickness of each retinal layer in different quadrants of macula and optic disc within a minute.
Previous studies based on fundus photography have demonstrated that the quantitative retinal vascular parameters including fractal dimension (FD), central retinal artery equivalent, and central retinal vein equivalent, as well as the qualitative retinopathy, are associated with dementia [27, 28]. Nevertheless, findings from other studies are inconsistent, especially in the early stage of AD [2932]. This discrepancy may be because that the relatively large blood vessels detected by the fundus camera were not sufficient to reflect the subtle pathology in the early stage of the disease.
As a milestone in the development of retinal imaging technology, the OCTA can detect the movement of red blood cells in the vascular lumen via changes in OCT signal measured by multiple scans in the same cross section. Combined with continuous en-face blood cell movement information, complete three-dimensional imaging of retinal and choroid vessels can be achieved. This is a novel imaging technique that can noninvasively and rapidly depict the microvasculature in different retinal layers with high resolution [3336]. In addition to the retinal vessel density (VD), FD [37], which represents the retinal capillary complexity, can be obtained from OCTA to detect subtle microvascular changes in the early stage of retinopathies [38] and neurodegenerative diseases [39]. In the following, we summarize relevant studies of OCT/OCTA in AD, MCI and preclinical AD patients (Tables 1 and 2, respectively).
Table 1
Studies on OCT parameters in patients with AD, MCI and preclinical AD
Authors
Year of publication
Layer
Subjects
Alteration
Parisi et al. [40]
2001
pRNFL
AD
Thinning
Iseri et al. [41]
2006
pRNFL, mRNFL
AD
Thinning
Berisha et al. [42]
2007
pRNFL
AD
Thinner in the superior quadrant
Lu et al. [43]
2010
pRNFL
AD
Thinner in the superior and inferior quadrants
Moschos et al. [44]
2012
pRNFL
AD
Thinning
Kirbas et al. [45]
2013
pRNFL
AD
Thinner in the superior quadrant
Marziani E et al. [46]
2013
mRNFL
mRNFL + GCL
AD
Thinning
Garcia-Martin et al. [47]
2016
pRNFL, mRNFL GCL, IPL, INL, OPL, ONL, RPE
AD
Thinner pRNFL, mRNFL, GCL, and IPL
Gharbiya et al. [48]
2014
pRNFL
AD
No difference
Larrosa et al. [49]
2014
pRNFL
AD
Thinning except the nasal quadrant.
Kromer et al. [50]
2014
pRNFL
AD
Thinner in the nasal superior sector
Polo V et al. [51]
2017
pRNFL
AD
Thinner in the superior and inferior quadrants.
Salobrar-Garcia et al. [52]
2015
pRNFL, mRNFL
AD
Thinner mRNFL
Eraslan et al. [53]
2015
pRNFL, GCC
AD
Thinning
La Morgia et al. [54]
2016
pRNFL
AD
Thinning
Cunha et al. [55]
2017
pRNFL
AD
Thinner in the temporal superior quadrants and globally
Liu et al. [56]
2015
pRNFL
AD
Mild and moderate AD had thinner pRNFL in the superior quadrant. Severe AD had thinner pRNFL in the superior and inferior quadrants.
Ferrari L et al. [57]
2017
pRNFL, pGC-IPL
AD
MCI
No difference in mild AD. Moderate AD had thinner pRNFL and pGC-IPL. MCI had thinner pRNFL.
Paquet et al. [58]
2007
pRNFL
AD
MCI
Thinning
Kesler et al. [59]
2011
pRNFL
AD
MCI
AD had thinner pRNFL in the superior and inferior quadrants. MCI had thinner pRNFL only in the inferior quadrant.
Ascaso et al. [60]
2014
pRNFL
AD
MCI
AD had thinner pRNFL in all sectors. MCI had thinner pRNFL in all sectors except the temporal quadrant.
Gao et al. [61]
2015
pRNFL
AD
MCI
Thinning
Oktem et al. [62]
2015
pRNFL
AD
MCI
Thinning
Cheung et al. [63]
2015
pRNFL, GC-IPL
AD
MCI
AD had thinner pRNFL in the superior quadrant and thinner mGC-IPL in all sectors.
MCI had thinner mGC-IPL in the majority of sectors.
Shao et al. [64]
2018
pRNFL, GC-IPL
AD
MCI
Thinning
Almeida et al. [65]
2019
pRNFL, mRNFL
MCI
Thinner mRNFL, mGC-IPL and mGCC
Santos et al. [66]
2018
pRNFL, mRNFL, GCL, IPL, OPL, INL, ONL
Preclinical AD
Thinner mRNFL, ONL and IPL
López-Cuenca et al. [67]
2020
pRNFL, mRNFL, GCL, IPL, INL, OPL, ONL, RPE
Preclinical AD
Thinner mRNFL, IPL, INL and OPL
Snyder et al. [68]
2016
pRNFL, mRNFL, GCL, IPL, INL, OPL, ONL,
Preclinical AD
Thicker IPL
van de Kreeke et al. [69]
2019
pRNFL, mRNFL, GCL, IPL,
Preclinical AD
No difference
van de Kreeke et al. [70]
2020
pRNFL, mRNFL, GCL, IPL,
Preclinical AD
No difference in the rate of change of any retinal layers
OCT optical coherence tomography; AD Alzheimer’s disease; MCI mild cognitive impairment; pRNFL peripapillary retinal nerve fiber layer; mRNFL macular retinal nerve fiber layer; GCL ganglion cell layer; IPL inner plexiform layer; GC-IPL ganglion cell-inner plexiform layer; pGC-IPL peripapillary ganglion cell-inner plexiform layer; mGCC macular ganglion cell complex; INL inner nuclear layer; OPL outer plexiform layer; ONL outer nuclear layer; RPE retinal pigment epithelium
Table 2
Studies on OCTA parameters in patients with AD, MCI and preclinical AD
Authors
Year of publication
Parameters
Subjects
Alteration
Bulut M et al. [71]
2018
SRCP-VD, DRCP-VD, FAZ
AD
Lower VD, enlarged FAZ.
Lahme L et al. [72]
2018
SRCP-flow density, DRCP-flow density, Peripapillary-VD
AD
Lower SRCP-flow density
Jiang et al. [73]
2017
SRCP-VD, DRCP-VD
AD
MCI
AD had lower VD of SRCP and DRCP. MCI had lower VD of DRCP in the superior nasal quadrant
Stephen P et al. [74]
2019
SRCP-VD, FAZ
AD
MCI
AD had lower SRCP-VD. No difference in MCI
Zhang et al. [75]
2019
SRCP-VD, DRCP-VD, RPC
MCI or early AD
Lower SRCP-VD
Wu et al. [76]
2020
SRCP-VD, DRCP-VD, FAZ
AD
MCI
Lower DRCP-VD
O’Bryhim et al. [77]
2018
SRCP-VD, FAZ
Preclinical AD
Lower VD, enlarged FAZ
van de Kreeke et al. [78]
2020
Macular-VD, Peripapillary-VD, FAZ
Preclinical AD
Higher VD, no difference in FAZ.
OCT optical coherence tomography; AD Alzheimer’s disease; MCI mild cognitive impairment; SRCP-VD superficial retinal capillary plexus-vessel density; DRCP-VD deep retinal capillary plexus-vessel density; RPC radial peripapillary capillary; FAZ foveal avascular zone

Changes in retinal neural structure in AD, MCI, and preclinical AD

Changes in RNFL and GCC in AD, MCI, and preclinical AD

The compositions of retinal RNFL and GC-IPL correspond to white and grey matter components of the brain, therefore, they are widely regarded as neural parameters reflecting the progression of AD. At advanced stage of AD, a recent meta-analysis also found a significant reduction in the overall mean pRNFL thickness as well as in the mean pRNFL thickness of all four quadrants [79]. In addition, some studies have reported significant pRNFL thinning in individual quadrants. In most studies, the superior [4143, 45, 49, 51, 55, 61] and inferior [41, 43, 49, 51] quadrants showed significantly greater thinning in patients with AD compared with controls, whereas the temporal [49, 55] and nasal [41, 61] quadrants were found to be significantly thinner only in a small number of studies. This pattern of quadrantal change in the retina may indicate that the magnocellular ganglion cells located in the extramacular retina are vulnerable to AD pathology [54]. In addition, another explanation is that the concentration of axon bundles is highest in the superior and inferior quadrants [80]. The characteristic quadrantal loss in the RNFL may have better diagnostic efficacy in distinguishing different types of dementia. In a recent study, researchers divided AD patients into different subgroups (mild, moderate and severe AD) and found that the pRNFL degeneration paralleled dementia progression [56]. The pRNFL decreased significantly in the superior quadrant in mild and moderate AD patients, compared to the controls. In severe AD, the loss of pRNFL occurred not only in the superior quadrant but also in the inferior quadrant. Others have pointed out that the size of the RGC is 10 to 20 times the diameter of axons, which may lead to more significant changes in the thickness of GC-IPL and GCC compared to the pRNFL [63]. Several studies have also found changes in the GC-IPL and GCC in AD. Significant thinning of the average GCC has been associated with AD [53]. A recent meta-analysis also showed that the average thickness of GC-IPL was significantly decreased in AD and the thinning occurred in most sectors around the fovea, except the supratemporal sector [81]. In addition, a series of studies has shown that the cognitive scores are significantly correlated with macular parameters rather than the pRNFL thickness [41, 57, 65]. A large population-based study in Japan discovered that the presence of dementia was inversely associated with full macular thickness and GCC thickness but not with the pRNFL thickness [82]. Another population-based study in Germany also showed that the GCC volume was more strongly related to the global function than was the pRNFL thickness [83]. In line with the above results, some also inferred that the GCC thickness performed better than pRNFL thickness in detecting the disease status based on the area under the curve value [84]. The divisions of the retina seem to have better diagnostic performance due to the less influence from individual variation [85]. These results indicate that the macular parameters are more useful than peripapillary parameters.
Similarly, pRNFL, GC-IPL or GCC thinning has also been found in MCI patients [5664, 79, 86]. Others even discovered thickening and thinning of regions adjacent to each other in MCI patients, indicating that the two layers undergo dynamic changes during progression from MCI to AD [87]. Interestingly, the mean thickness of GC-IPL and GCC in MCI patients is significantly reduced compared with that in controls, while changes in the pRNFL thickness are not significant [65]. The above findings suggest that the ganglion cells around macula, reflected by the thickness of GC-IPL or GCC, are sensitive in detecting neural structural changes in the initial stage of MCI.
In addition, a 27-month longitudinal study on preclinical AD showed significant changes in macular RNFL (mRNFL) rather than pRNFL over time [66]. The mRNFL thinning was significantly correlated with increased neocortical accumulation of Aβ and impaired performance in a task of audiovisual integration efficiency. Another study also suggested that the axonal loss in the mRNFL occurred earlier than pRNFL degeneration and this retinal parameter was expected to reflect the subtle disruption of white matter integrity in the preclinical AD stage [67]. However, several other studies did not report similar significant results [68, 69]. Current cross-sectional studies have reported no statistically significant differences in the pRNFL or GCL between preclinical AD and controls. A longitudinal follow-up study also showed no difference in the rate of change of the above layers [70]. Therefore, the diagnostic efficacy of retinal neural structural parameters and their relationship with the progression of AD should be further studied especially in the preclinical AD stage.

Other retinal layer changes in AD, MCI, and preclinical AD

In additional to the neurodegenerative changes, neuroinflammation in retina and brain is also involved in AD progression [88]. Retinal glial cells including Muller cells, astrocytes and microglia cells are involved in immune and inflammatory responses and may lead to changes in the outer layer of the retina. The outer layer is relatively less studied and the results are inconsistent. Some studies in AD patients have found remarkable thickening of the ONL [64, 89], while others revealed no significant decrease of the outer retinal thickness including the ONL [90]. Furthermore, there is no significant difference in IPL and INL, albeit with OPL thinning [89]. However, inconsistent with the above results based on OCT devices, postmortem examination of retinas of AD patients showed thinning of pRNFL, GCL, IPL, INL, OPL and ONL [91]. The researchers further pointed out that the retinal structural damage may be attributed to the retrograde trans-synaptic degeneration, deposition of Aβ plaques, as well as the consequent neurotoxicity.
In MCI subjects, some studies have found OPL thinning and ONL thickening [89], while others revealed no significant difference in the ONL [90]. Only a few studies have focused on the outer retinal layers in preclinical AD and the results are controversial. A recent study found significant decreases in the IPL, INL and OPL in ApoE ɛ4 carriers with a family history of AD compared to the non-carriers without a family history [67]. Another study also showed a decrease in IPL and ONL volumes over a 27-month follow-up in older adults with multiple risk factors for AD, compared to the healthy control subjects [66]. Inconsistently, a cross-sectional study reported no significant differences in INL, ONL and OPL [68]. Besides, remarkable IPL thickening has been reported as well. As for IPL, a team showed no difference in cross-sectional observation or in the rate of change [69, 70]. These findings suggest that the individual retinal layers play different roles at different stages of AD, and the thickening and thinning of the outer layer, which contains a variety of cells, is not specific compared to the RNFL and GCC. Of course, the differences in diagnostic criteria, inclusion and exclusion criteria, and devices may lead to the heterogeneity of the results. Further large longitudinal studies are needed to confirm the effectiveness of individual retinal layers, especially in the preclinical AD stage.

Retinal microvascular changes in AD, MCI and preclinical AD

Retinal microvascular OCTA findings are relatively less reported. In general, retinal microvascular damage can be observed at all stages of AD. A report on monozygotic twin pairs showed significantly decreased VD in the superficial retinal capillary plexus (SRCP) in twins with AD compared with cognitively normal twins, indicating that the changes of SRCP may serve as a possible biomarker for AD based on OCTA [92]. Several recent studies have also shown decreased SRCP or deep retinal capillary plexus (DRCP), as well as enlarged foveal avascular zone (FAZ) in AD subjects [7174, 76]. The FAZ enlargement may be due to the dropout of vasculature within the fovea. Besides, a significantly reduced flow density in radial peripapillary capillaries (RPC) with larger vascular channels in the peripapillary region has also been found in AD patients [72]. Most studies found that the vascular parameters were significantly correlated with cognitive scores [71, 7477]. However, some studies also showed no association between retinal vascular parameters and cognitive function [72, 73]. The inconsistency may result from ambiguous clinical staging of disease progression with a small sample size.
In addition, some studies have shown a tendency of microvascular density loss along the development of AD, which indicates that the retinal vascular impairment reflected as reduced microvascular density by OCTA can serve as a marker for monitoring disease progression [73]. Meanwhile, MCI individuals had significantly declined parafoveal SRCP VD and adjusted flow index, while no difference was found in the RPC [75]. Contrary to the above study, other studies have demonstrated no statistically significant difference in SRCP VD [73, 74, 76]. Interestingly, two studies found lower microvascular density in DRCP rather than in SRCP in MCI patients [73, 76]. This may be because that the smaller microvasculature of the DRCP is more susceptible to disease development than larger vessels of the SRCP, suggesting that the deep microvascular density around macula is more sensitive in detecting and monitoring progression of MCI at the early stage.
So far, there are only two relevant studies focusing on preclinical AD. Notably, one study revealed enlargement of the FAZ in individuals with preclinical AD with positive positron emission tomography scan biomarkers [77]. However, the other showed no statistical difference in FAZ size between preclinical AD and healthy individuals. Even more, they found increased vessel density in preclinical AD subjects, which may be caused by the increased blood flow secondary to hypoxia [78].
In sum, research on the role of microvascular pathology as reflected by OCTA in early diagnosis of AD is still in the preliminary stage. The relationship between vascular change and neurodegenerative changes needs to be further investigated in more studies with longer follow-up durations.

Conclusion

In conclusion, accumulating evidence has shown that the retina could provide valuable insights into the early diagnosis of AD. Retinal neuronal structural and microvascular imaging by OCT/OCTA is potentially useful for large-scale population screening or monitoring responses to therapies in AD patients. Before full application in clinical practice, further research is needed to validate whether the findings on retinal neuronal loss and microvascular damage in MCI or even in preclinical AD patients are related with the progression of cognitive impairment and brain neuronal/vascular loss in individual AD patients, based on longitudinal data of multi-center studies. Histopathological studies are also needed to understand the pathophysiological mechanisms underlying the retinal neuronal and microvascular alterations, which will facilitate the clinical applications of retinal OCT/OCTA imaging in large-scale population screening and monitoring and the development of new therapies.

Acknowledgements

Not applicable.
Not applicable.
Not applicable.

Competing interests

All authors claim that there are no conflicts of interest.
Open AccessThis article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://​creativecommons.​org/​licenses/​by/​4.​0/​. The Creative Commons Public Domain Dedication waiver (http://​creativecommons.​org/​publicdomain/​zero/​1.​0/​) applies to the data made available in this article, unless otherwise stated in a credit line to the data.
Literatur
2.
Zurück zum Zitat Savva GM, Wharton SB, Ince PG, Forster G, Matthews FE, Brayne C. Age, neuropathology, and dementia. N Engl J Med. 2009;360:2302–9.PubMedCrossRef Savva GM, Wharton SB, Ince PG, Forster G, Matthews FE, Brayne C. Age, neuropathology, and dementia. N Engl J Med. 2009;360:2302–9.PubMedCrossRef
3.
Zurück zum Zitat Ramirez AI, de Hoz R, Salobrar-Garcia E, Salazar JJ, Rojas B, Ajoy D, et al. The role of microglia in retinal neurodegeneration: Alzheimer's disease, Parkinson, and glaucoma. Front Aging Neurosci. 2017;9:214.PubMedPubMedCentralCrossRef Ramirez AI, de Hoz R, Salobrar-Garcia E, Salazar JJ, Rojas B, Ajoy D, et al. The role of microglia in retinal neurodegeneration: Alzheimer's disease, Parkinson, and glaucoma. Front Aging Neurosci. 2017;9:214.PubMedPubMedCentralCrossRef
4.
Zurück zum Zitat Fernández-Albarral JA, Salobrar-García E, Martínez-Páramo R, Ramírez AI, de Hoz R, Ramírez JM, et al. Retinal glial changes in Alzheimer's disease - a review. J Optom. 2019;12:198–207.PubMedCrossRef Fernández-Albarral JA, Salobrar-García E, Martínez-Páramo R, Ramírez AI, de Hoz R, Ramírez JM, et al. Retinal glial changes in Alzheimer's disease - a review. J Optom. 2019;12:198–207.PubMedCrossRef
5.
Zurück zum Zitat Prince M, Wimo A, Guerchet M, Ali GC, Wu YT, Prina M, et al. World Alzheimer Report 2015. The Global Impact of Dementia: An Analysis of Prevalence, Incidence, Cost and Trends. London: Alzheimer’s Disease International (ADI); 2015. Prince M, Wimo A, Guerchet M, Ali GC, Wu YT, Prina M, et al. World Alzheimer Report 2015. The Global Impact of Dementia: An Analysis of Prevalence, Incidence, Cost and Trends. London: Alzheimer’s Disease International (ADI); 2015.
6.
Zurück zum Zitat Brookmeyer R, Johnson E, Ziegler-Graham K, Arrighi HM. Forecasting the global burden of Alzheimer's disease. Alzheimers Dement. 2007;3:186–91.PubMedCrossRef Brookmeyer R, Johnson E, Ziegler-Graham K, Arrighi HM. Forecasting the global burden of Alzheimer's disease. Alzheimers Dement. 2007;3:186–91.PubMedCrossRef
7.
Zurück zum Zitat Martínez G, Vernooij RW, Fuentes Padilla P, Zamora J, Flicker L, Bonfill CX. 18F PET with flutemetamol for the early diagnosis of Alzheimer's disease dementia and other dementias in people with mild cognitive impairment (MCI). Cochrane Database Syst Rev. 2017;11:Cd012884.PubMed Martínez G, Vernooij RW, Fuentes Padilla P, Zamora J, Flicker L, Bonfill CX. 18F PET with flutemetamol for the early diagnosis of Alzheimer's disease dementia and other dementias in people with mild cognitive impairment (MCI). Cochrane Database Syst Rev. 2017;11:Cd012884.PubMed
8.
Zurück zum Zitat Rabinovici GD, Gatsonis C, Apgar C, Chaudhary K, Gareen I, Hanna L, et al. Association of amyloid positron emission tomography with subsequent change in clinical management among medicare beneficiaries with mild cognitive impairment or dementia. JAMA. 2019;321:1286–94.PubMedPubMedCentralCrossRef Rabinovici GD, Gatsonis C, Apgar C, Chaudhary K, Gareen I, Hanna L, et al. Association of amyloid positron emission tomography with subsequent change in clinical management among medicare beneficiaries with mild cognitive impairment or dementia. JAMA. 2019;321:1286–94.PubMedPubMedCentralCrossRef
9.
Zurück zum Zitat Kaerst L, Kuhlmann A, Wedekind D, Stoeck K, Lange P, Zerr I. Using cerebrospinal fluid marker profiles in clinical diagnosis of dementia with Lewy bodies, Parkinson's disease, and Alzheimer's disease. J Alzheimers Dis. 2014;38:63–73.PubMedCrossRef Kaerst L, Kuhlmann A, Wedekind D, Stoeck K, Lange P, Zerr I. Using cerebrospinal fluid marker profiles in clinical diagnosis of dementia with Lewy bodies, Parkinson's disease, and Alzheimer's disease. J Alzheimers Dis. 2014;38:63–73.PubMedCrossRef
10.
Zurück zum Zitat Struyfs H, Van Broeck B, Timmers M, Fransen E, Sleegers K, Van Broeckhoven C, et al. Diagnostic accuracy of cerebrospinal fluid amyloid-β isoforms for early and differential dementia diagnosis. J Alzheimers Dis. 2015;45:813–22.PubMedCrossRef Struyfs H, Van Broeck B, Timmers M, Fransen E, Sleegers K, Van Broeckhoven C, et al. Diagnostic accuracy of cerebrospinal fluid amyloid-β isoforms for early and differential dementia diagnosis. J Alzheimers Dis. 2015;45:813–22.PubMedCrossRef
11.
Zurück zum Zitat Koronyo Y, Biggs D, Barron E, Boyer DS, Pearlman JA, Au WJ, et al. Retinal amyloid pathology and proof-of-concept imaging trial in Alzheimer's disease. JCI Insight. 2017;2. Koronyo Y, Biggs D, Barron E, Boyer DS, Pearlman JA, Au WJ, et al. Retinal amyloid pathology and proof-of-concept imaging trial in Alzheimer's disease. JCI Insight. 2017;2.
12.
Zurück zum Zitat Shi Z, Cao X, Hu J, Jiang L, Mei X, Zheng H, et al. Retinal nerve fiber layer thickness is associated with hippocampus and lingual gyrus volumes in nondemented older adults. Prog Neuro-Psychopharmacol Biol Psychiatry. 2020;99:109824.CrossRef Shi Z, Cao X, Hu J, Jiang L, Mei X, Zheng H, et al. Retinal nerve fiber layer thickness is associated with hippocampus and lingual gyrus volumes in nondemented older adults. Prog Neuro-Psychopharmacol Biol Psychiatry. 2020;99:109824.CrossRef
13.
Zurück zum Zitat Shi Z, Zheng H, Hu J, Jiang L, Cao X, Chen Y, et al. Retinal nerve fiber layer thinning is associated with brain atrophy: a longitudinal study in nondemented older adults. Front Aging Neurosci. 2019;11:69.PubMedPubMedCentralCrossRef Shi Z, Zheng H, Hu J, Jiang L, Cao X, Chen Y, et al. Retinal nerve fiber layer thinning is associated with brain atrophy: a longitudinal study in nondemented older adults. Front Aging Neurosci. 2019;11:69.PubMedPubMedCentralCrossRef
14.
Zurück zum Zitat Gommer ED, Martens EG, Aalten P, Shijaku E, Verhey FR, Mess WH, et al. Dynamic cerebral autoregulation in subjects with Alzheimer's disease, mild cognitive impairment, and controls: evidence for increased peripheral vascular resistance with possible predictive value. J Alzheimers Dis. 2012;30:805–13.PubMedCrossRef Gommer ED, Martens EG, Aalten P, Shijaku E, Verhey FR, Mess WH, et al. Dynamic cerebral autoregulation in subjects with Alzheimer's disease, mild cognitive impairment, and controls: evidence for increased peripheral vascular resistance with possible predictive value. J Alzheimers Dis. 2012;30:805–13.PubMedCrossRef
15.
Zurück zum Zitat Lin YF, Smith AV, Aspelund T, Betensky RA, Smoller JW, Gudnason V, et al. Genetic overlap between vascular pathologies and Alzheimer's dementia and potential causal mechanisms. Alzheimers Dement. 2019;15:65–75.PubMedCrossRef Lin YF, Smith AV, Aspelund T, Betensky RA, Smoller JW, Gudnason V, et al. Genetic overlap between vascular pathologies and Alzheimer's dementia and potential causal mechanisms. Alzheimers Dement. 2019;15:65–75.PubMedCrossRef
16.
Zurück zum Zitat Patton N, Aslam T, Macgillivray T, Pattie A, Deary IJ, Dhillon B. Retinal vascular image analysis as a potential screening tool for cerebrovascular disease: a rationale based on homology between cerebral and retinal microvasculatures. J Anat. 2005;206:319–48.PubMedPubMedCentralCrossRef Patton N, Aslam T, Macgillivray T, Pattie A, Deary IJ, Dhillon B. Retinal vascular image analysis as a potential screening tool for cerebrovascular disease: a rationale based on homology between cerebral and retinal microvasculatures. J Anat. 2005;206:319–48.PubMedPubMedCentralCrossRef
17.
Zurück zum Zitat Ong YT, Hilal S, Cheung CY, Venketasubramanian N, Niessen WJ, Vrooman H, et al. Retinal neurodegeneration on optical coherence tomography and cerebral atrophy. Neurosci Lett. 2015;584:12–6.PubMedCrossRef Ong YT, Hilal S, Cheung CY, Venketasubramanian N, Niessen WJ, Vrooman H, et al. Retinal neurodegeneration on optical coherence tomography and cerebral atrophy. Neurosci Lett. 2015;584:12–6.PubMedCrossRef
18.
Zurück zum Zitat Hayreh SS. The 1994 Von Sallman lecture. The optic nerve head circulation in health and disease. Exp Eye Res. 1995;61:259–72.PubMedCrossRef Hayreh SS. The 1994 Von Sallman lecture. The optic nerve head circulation in health and disease. Exp Eye Res. 1995;61:259–72.PubMedCrossRef
19.
Zurück zum Zitat Hayreh SS. The blood supply of the optic nerve head and the evaluation of it - myth and reality. Prog Retin Eye Res. 2001;20:563–93.PubMedCrossRef Hayreh SS. The blood supply of the optic nerve head and the evaluation of it - myth and reality. Prog Retin Eye Res. 2001;20:563–93.PubMedCrossRef
20.
Zurück zum Zitat Ryan SJ, Sadda SR. Ryan's retinal imaging and diagnostics (1st Ed.). Saunders; 2013. Ryan SJ, Sadda SR. Ryan's retinal imaging and diagnostics (1st Ed.). Saunders; 2013.
21.
Zurück zum Zitat JMea R. Glia and blood retinal barrier: effects of ocular hypertension. In: Cardiovascular Disease II. iConcept Press Ltd; 2014. JMea R. Glia and blood retinal barrier: effects of ocular hypertension. In: Cardiovascular Disease II. iConcept Press Ltd; 2014.
22.
Zurück zum Zitat Koronyo-Hamaoui M, Koronyo Y, Ljubimov AV, Miller CA, Ko MK, Black KL, et al. Identification of amyloid plaques in retinas from Alzheimer's patients and noninvasive in vivo optical imaging of retinal plaques in a mouse model. Neuroimage. 2011;54(Suppl 1):S204–17.PubMedCrossRef Koronyo-Hamaoui M, Koronyo Y, Ljubimov AV, Miller CA, Ko MK, Black KL, et al. Identification of amyloid plaques in retinas from Alzheimer's patients and noninvasive in vivo optical imaging of retinal plaques in a mouse model. Neuroimage. 2011;54(Suppl 1):S204–17.PubMedCrossRef
23.
Zurück zum Zitat Entezari M, Azhari S, Ramezani A. Fundus findings in spontaneous subarachnoid hemorrhage and their correlation with neurologic characteristics. Eur J Ophthalmol. 2009;19:460–5.PubMedCrossRef Entezari M, Azhari S, Ramezani A. Fundus findings in spontaneous subarachnoid hemorrhage and their correlation with neurologic characteristics. Eur J Ophthalmol. 2009;19:460–5.PubMedCrossRef
24.
Zurück zum Zitat Rhee EJ, Chung PW, Wong TY, Song SJ. Relationship of retinal vascular caliber variation with intracranial arterial stenosis. Microvasc Res. 2016;108:64–8.PubMedCrossRef Rhee EJ, Chung PW, Wong TY, Song SJ. Relationship of retinal vascular caliber variation with intracranial arterial stenosis. Microvasc Res. 2016;108:64–8.PubMedCrossRef
26.
Zurück zum Zitat Pazos M, Dyrda AA, Biarnés M, Gómez A, Martín C, Mora C, et al. Diagnostic accuracy of Spectralis SD OCT automated macular layers segmentation to discriminate normal from early glaucomatous eyes. Ophthalmology. 2017;124:1218–28.PubMedCrossRef Pazos M, Dyrda AA, Biarnés M, Gómez A, Martín C, Mora C, et al. Diagnostic accuracy of Spectralis SD OCT automated macular layers segmentation to discriminate normal from early glaucomatous eyes. Ophthalmology. 2017;124:1218–28.PubMedCrossRef
27.
Zurück zum Zitat Frost S, Kanagasingam Y, Sohrabi H, Vignarajan J, Bourgeat P, Salvado O, et al. Retinal vascular biomarkers for early detection and monitoring of Alzheimer's disease. Transl Psychiatry. 2013;3:e233.PubMedPubMedCentralCrossRef Frost S, Kanagasingam Y, Sohrabi H, Vignarajan J, Bourgeat P, Salvado O, et al. Retinal vascular biomarkers for early detection and monitoring of Alzheimer's disease. Transl Psychiatry. 2013;3:e233.PubMedPubMedCentralCrossRef
28.
Zurück zum Zitat Deal JA, Sharrett AR, Rawlings AM, Gottesman RF, Bandeen-Roche K, Albert M, et al. Retinal signs and 20-year cognitive decline in the atherosclerosis risk in communities study. Neurology. 2018;90:e1158–66.PubMedPubMedCentralCrossRef Deal JA, Sharrett AR, Rawlings AM, Gottesman RF, Bandeen-Roche K, Albert M, et al. Retinal signs and 20-year cognitive decline in the atherosclerosis risk in communities study. Neurology. 2018;90:e1158–66.PubMedPubMedCentralCrossRef
29.
Zurück zum Zitat McGrory S, Ballerini L, Okely JA, Ritchie SJ, Doubal FN, Doney AS, et al. Retinal microvascular features and cognitive change in the Lothian-birth cohort 1936. Alzheimers Dement (Amst). 2019;11:500–9.PubMedPubMedCentralCrossRef McGrory S, Ballerini L, Okely JA, Ritchie SJ, Doubal FN, Doney AS, et al. Retinal microvascular features and cognitive change in the Lothian-birth cohort 1936. Alzheimers Dement (Amst). 2019;11:500–9.PubMedPubMedCentralCrossRef
30.
Zurück zum Zitat McGrory S, Cameron JR, Pellegrini E, Warren C, Doubal FN, Deary IJ, et al. The application of retinal fundus camera imaging in dementia: a systematic review. Alzheimers Dement (Amst). 2017;6:91–107.CrossRef McGrory S, Cameron JR, Pellegrini E, Warren C, Doubal FN, Deary IJ, et al. The application of retinal fundus camera imaging in dementia: a systematic review. Alzheimers Dement (Amst). 2017;6:91–107.CrossRef
31.
Zurück zum Zitat Lee CS, Tyring AJ, Wu Y, Xiao S, Rokem AS, DeRuyter NP, et al. Generating retinal flow maps from structural optical coherence tomography with artificial intelligence. Sci Rep. 2019;9:5694.PubMedPubMedCentralCrossRef Lee CS, Tyring AJ, Wu Y, Xiao S, Rokem AS, DeRuyter NP, et al. Generating retinal flow maps from structural optical coherence tomography with artificial intelligence. Sci Rep. 2019;9:5694.PubMedPubMedCentralCrossRef
32.
Zurück zum Zitat Jung NY, Han JC, Ong YT, Cheung CY, Chen CP, Wong TY, et al. Retinal microvasculature changes in amyloid-negative subcortical vascular cognitive impairment compared to amyloid-positive Alzheimer's disease. J Neurol Sci. 2019;396:94–101.PubMedCrossRef Jung NY, Han JC, Ong YT, Cheung CY, Chen CP, Wong TY, et al. Retinal microvasculature changes in amyloid-negative subcortical vascular cognitive impairment compared to amyloid-positive Alzheimer's disease. J Neurol Sci. 2019;396:94–101.PubMedCrossRef
33.
Zurück zum Zitat Jia Y, Tan O, Tokayer J, Potsaid B, Wang Y, Liu JJ, et al. Split-spectrum amplitude-decorrelation angiography with optical coherence tomography. Opt Express. 2012;20:4710–25.PubMedPubMedCentralCrossRef Jia Y, Tan O, Tokayer J, Potsaid B, Wang Y, Liu JJ, et al. Split-spectrum amplitude-decorrelation angiography with optical coherence tomography. Opt Express. 2012;20:4710–25.PubMedPubMedCentralCrossRef
34.
Zurück zum Zitat Spaide RF, Klancnik Jr JM Jr, Cooney MJ. Retinal vascular layers imaged by fluorescein angiography and optical coherence tomography angiography. JAMA Ophthalmol. 2015;133:45–50.CrossRefPubMed Spaide RF, Klancnik Jr JM Jr, Cooney MJ. Retinal vascular layers imaged by fluorescein angiography and optical coherence tomography angiography. JAMA Ophthalmol. 2015;133:45–50.CrossRefPubMed
35.
Zurück zum Zitat Kashani AH, Chen CL, Gahm JK, Zheng F, Richter GM, Rosenfeld PJ, et al. Optical coherence tomography angiography: a comprehensive review of current methods and clinical applications. Prog Retin Eye Res. 2017;60:66–100.PubMedPubMedCentralCrossRef Kashani AH, Chen CL, Gahm JK, Zheng F, Richter GM, Rosenfeld PJ, et al. Optical coherence tomography angiography: a comprehensive review of current methods and clinical applications. Prog Retin Eye Res. 2017;60:66–100.PubMedPubMedCentralCrossRef
36.
Zurück zum Zitat Savastano MC, Lumbroso B, Rispoli M. In vivo characterization of retinal vascularization morphology using optical coherence tomography angiography. Retina. 2015;35:2196–203.PubMedCrossRef Savastano MC, Lumbroso B, Rispoli M. In vivo characterization of retinal vascularization morphology using optical coherence tomography angiography. Retina. 2015;35:2196–203.PubMedCrossRef
37.
Zurück zum Zitat Jiang H, Debuc DC, Rundek T, Lam BL, Wright CB, Shen M, et al. Automated segmentation and fractal analysis of high-resolution non-invasive capillary perfusion maps of the human retina. Microvasc Res. 2013;89:172–5.PubMedPubMedCentralCrossRef Jiang H, Debuc DC, Rundek T, Lam BL, Wright CB, Shen M, et al. Automated segmentation and fractal analysis of high-resolution non-invasive capillary perfusion maps of the human retina. Microvasc Res. 2013;89:172–5.PubMedPubMedCentralCrossRef
38.
Zurück zum Zitat Chen Q, Ma Q, Wu C, Tan F, Chen F, Wu Q, et al. Macular vascular fractal dimension in the deep capillary layer as an early indicator of microvascular loss for retinopathy in type 2 diabetic patients. Invest Ophthalmol Vis Sci. 2017;58:3785–94.PubMedCrossRef Chen Q, Ma Q, Wu C, Tan F, Chen F, Wu Q, et al. Macular vascular fractal dimension in the deep capillary layer as an early indicator of microvascular loss for retinopathy in type 2 diabetic patients. Invest Ophthalmol Vis Sci. 2017;58:3785–94.PubMedCrossRef
39.
Zurück zum Zitat Shi C, Chen Y, Kwapong WR, Tong Q, Wu S, Zhou Y, et al. Characterization by fractal dimension analysis of the retinal capillary network in Parkinson disease. Retina. 2020;40:1483–91.PubMedCrossRef Shi C, Chen Y, Kwapong WR, Tong Q, Wu S, Zhou Y, et al. Characterization by fractal dimension analysis of the retinal capillary network in Parkinson disease. Retina. 2020;40:1483–91.PubMedCrossRef
40.
Zurück zum Zitat Parisi V, Restuccia R, Fattapposta F, Mina C, Bucci MG, Pierelli F. Morphological and functional retinal impairment in Alzheimer's disease patients. Clin Neurophysiol. 2001;112:1860–7.PubMedCrossRef Parisi V, Restuccia R, Fattapposta F, Mina C, Bucci MG, Pierelli F. Morphological and functional retinal impairment in Alzheimer's disease patients. Clin Neurophysiol. 2001;112:1860–7.PubMedCrossRef
41.
Zurück zum Zitat Iseri PK, Altinaş O, Tokay T, Yüksel N. Relationship between cognitive impairment and retinal morphological and visual functional abnormalities in Alzheimer disease. J Neuroophthalmol. 2006;26:18–24.PubMedCrossRef Iseri PK, Altinaş O, Tokay T, Yüksel N. Relationship between cognitive impairment and retinal morphological and visual functional abnormalities in Alzheimer disease. J Neuroophthalmol. 2006;26:18–24.PubMedCrossRef
42.
Zurück zum Zitat Berisha F, Feke GT, Trempe CL, McMeel JW, Schepens CL. Retinal abnormalities in early Alzheimer's disease. Invest Ophthalmol Vis Sci. 2007;48:2285–9.PubMedCrossRef Berisha F, Feke GT, Trempe CL, McMeel JW, Schepens CL. Retinal abnormalities in early Alzheimer's disease. Invest Ophthalmol Vis Sci. 2007;48:2285–9.PubMedCrossRef
43.
Zurück zum Zitat Lu Y, Li Z, Zhang X, Ming B, Jia J, Wang R, et al. Retinal nerve fiber layer structure abnormalities in early Alzheimer's disease: evidence in optical coherence tomography. Neurosci Lett. 2010;480:69–72.PubMedCrossRef Lu Y, Li Z, Zhang X, Ming B, Jia J, Wang R, et al. Retinal nerve fiber layer structure abnormalities in early Alzheimer's disease: evidence in optical coherence tomography. Neurosci Lett. 2010;480:69–72.PubMedCrossRef
44.
Zurück zum Zitat Moschos MM, Markopoulos I, Chatziralli I, Rouvas A, Papageorgiou SG, Ladas I, et al. Structural and functional impairment of the retina and optic nerve in Alzheimer's disease. Curr Alzheimer Res. 2012;9:782–8.PubMedCrossRef Moschos MM, Markopoulos I, Chatziralli I, Rouvas A, Papageorgiou SG, Ladas I, et al. Structural and functional impairment of the retina and optic nerve in Alzheimer's disease. Curr Alzheimer Res. 2012;9:782–8.PubMedCrossRef
45.
Zurück zum Zitat Kirbas S, Turkyilmaz K, Anlar O, Tufekci A, Durmus M. Retinal nerve fiber layer thickness in patients with Alzheimer disease. J Neuroophthalmol. 2013;33:58–61.PubMedCrossRef Kirbas S, Turkyilmaz K, Anlar O, Tufekci A, Durmus M. Retinal nerve fiber layer thickness in patients with Alzheimer disease. J Neuroophthalmol. 2013;33:58–61.PubMedCrossRef
46.
Zurück zum Zitat Marziani E, Pomati S, Ramolfo P, Cigada M, Giani A, Mariani C, et al. Evaluation of retinal nerve fiber layer and ganglion cell layer thickness in Alzheimer's disease using spectral-domain optical coherence tomography. Invest Ophthalmol Vis Sci. 2013;54:5953–8.PubMedCrossRef Marziani E, Pomati S, Ramolfo P, Cigada M, Giani A, Mariani C, et al. Evaluation of retinal nerve fiber layer and ganglion cell layer thickness in Alzheimer's disease using spectral-domain optical coherence tomography. Invest Ophthalmol Vis Sci. 2013;54:5953–8.PubMedCrossRef
47.
Zurück zum Zitat Garcia-Martin E, Bambo MP, Marques ML, Satue M, Otin S, Larrosa JM, et al. Ganglion cell layer measurements correlate with disease severity in patients with Alzheimer's disease. Acta Ophthalmol. 2016;94:e454–9.PubMedCrossRef Garcia-Martin E, Bambo MP, Marques ML, Satue M, Otin S, Larrosa JM, et al. Ganglion cell layer measurements correlate with disease severity in patients with Alzheimer's disease. Acta Ophthalmol. 2016;94:e454–9.PubMedCrossRef
48.
Zurück zum Zitat Gharbiya M, Trebbastoni A, Parisi F, Manganiello S, Cruciani F, D'Antonio F, et al. Choroidal thinning as a new finding in Alzheimer's disease: evidence from enhanced depth imaging spectral domain optical coherence tomography. J Alzheimers Dis. 2014;40:907–17.PubMedCrossRef Gharbiya M, Trebbastoni A, Parisi F, Manganiello S, Cruciani F, D'Antonio F, et al. Choroidal thinning as a new finding in Alzheimer's disease: evidence from enhanced depth imaging spectral domain optical coherence tomography. J Alzheimers Dis. 2014;40:907–17.PubMedCrossRef
49.
Zurück zum Zitat Larrosa JM, Garcia-Martin E, Bambo MP, Pinilla J, Polo V, Otin S, et al. Potential new diagnostic tool for Alzheimer's disease using a linear discriminant function for Fourier domain optical coherence tomography. Invest Ophthalmol Vis Sci. 2014;55:3043–51.PubMedCrossRef Larrosa JM, Garcia-Martin E, Bambo MP, Pinilla J, Polo V, Otin S, et al. Potential new diagnostic tool for Alzheimer's disease using a linear discriminant function for Fourier domain optical coherence tomography. Invest Ophthalmol Vis Sci. 2014;55:3043–51.PubMedCrossRef
50.
Zurück zum Zitat Kromer R, Serbecic N, Hausner L, Froelich L, Aboul-Enein F, Beutelspacher SC. Detection of retinal nerve fiber layer defects in Alzheimer's disease using SD-OCT. Front Psychiatry. 2014;5:22.PubMedPubMedCentralCrossRef Kromer R, Serbecic N, Hausner L, Froelich L, Aboul-Enein F, Beutelspacher SC. Detection of retinal nerve fiber layer defects in Alzheimer's disease using SD-OCT. Front Psychiatry. 2014;5:22.PubMedPubMedCentralCrossRef
51.
Zurück zum Zitat Polo V, Rodrigo MJ, Garcia-Martin E, Otin S, Larrosa JM, Fuertes MI, et al. Visual dysfunction and its correlation with retinal changes in patients with Alzheimer's disease. Eye (Lond). 2017;31:1034–41.CrossRef Polo V, Rodrigo MJ, Garcia-Martin E, Otin S, Larrosa JM, Fuertes MI, et al. Visual dysfunction and its correlation with retinal changes in patients with Alzheimer's disease. Eye (Lond). 2017;31:1034–41.CrossRef
52.
Zurück zum Zitat Salobrar-Garcia E, Hoyas I, Leal M, de Hoz R, Rojas B, Ramirez AI, et al. Analysis of retinal peripapillary segmentation in early Alzheimer's disease patients. Biomed Res Int. 2015;2015:636548.PubMedPubMedCentralCrossRef Salobrar-Garcia E, Hoyas I, Leal M, de Hoz R, Rojas B, Ramirez AI, et al. Analysis of retinal peripapillary segmentation in early Alzheimer's disease patients. Biomed Res Int. 2015;2015:636548.PubMedPubMedCentralCrossRef
53.
Zurück zum Zitat Eraslan M, Çerman E, Çekiç O, Balci S, Dericioğlu V, Sahin Ö, et al. Neurodegeneration in ocular and central nervous systems: optical coherence tomography study in normal-tension glaucoma and Alzheimer disease. Turk Med Sci. 2015;45:1106–14.CrossRef Eraslan M, Çerman E, Çekiç O, Balci S, Dericioğlu V, Sahin Ö, et al. Neurodegeneration in ocular and central nervous systems: optical coherence tomography study in normal-tension glaucoma and Alzheimer disease. Turk Med Sci. 2015;45:1106–14.CrossRef
54.
Zurück zum Zitat La Morgia C, Ross-Cisneros FN, Koronyo Y, Hannibal J, Gallassi R, Cantalupo G, et al. Melanopsin retinal ganglion cell loss in Alzheimer disease. Ann Neurol. 2016;79:90–109.PubMedCrossRef La Morgia C, Ross-Cisneros FN, Koronyo Y, Hannibal J, Gallassi R, Cantalupo G, et al. Melanopsin retinal ganglion cell loss in Alzheimer disease. Ann Neurol. 2016;79:90–109.PubMedCrossRef
55.
Zurück zum Zitat Cunha JP, Proenca R, Dias-Santos A, Almeida R, Águas H, Alves M, et al. OCT in Alzheimer's disease: thinning of the RNFL and superior hemiretina. Graefes Arch Clin Exp Ophthalmol. 2017;255:1827–35.PubMedCrossRef Cunha JP, Proenca R, Dias-Santos A, Almeida R, Águas H, Alves M, et al. OCT in Alzheimer's disease: thinning of the RNFL and superior hemiretina. Graefes Arch Clin Exp Ophthalmol. 2017;255:1827–35.PubMedCrossRef
56.
Zurück zum Zitat Liu D, Zhang L, Li Z, Zhang X, Wu Y, Yang H, et al. Thinner changes of the retinal nerve fiber layer in patients with mild cognitive impairment and Alzheimer's disease. BMC Neurol. 2015;15:14.PubMedPubMedCentralCrossRef Liu D, Zhang L, Li Z, Zhang X, Wu Y, Yang H, et al. Thinner changes of the retinal nerve fiber layer in patients with mild cognitive impairment and Alzheimer's disease. BMC Neurol. 2015;15:14.PubMedPubMedCentralCrossRef
57.
Zurück zum Zitat Ferrari L, Huang SC, Magnani G, Ambrosi A, Comi G, Leocani L. Optical coherence tomography reveals retinal neuroaxonal thinning in frontotemporal dementia as in Alzheimer's disease. J Alzheimers Dis. 2017;56:1101–7.PubMedCrossRef Ferrari L, Huang SC, Magnani G, Ambrosi A, Comi G, Leocani L. Optical coherence tomography reveals retinal neuroaxonal thinning in frontotemporal dementia as in Alzheimer's disease. J Alzheimers Dis. 2017;56:1101–7.PubMedCrossRef
58.
Zurück zum Zitat Paquet C, Boissonnot M, Roger F, Dighiero P, Gil R, Hugon J. Abnormal retinal thickness in patients with mild cognitive impairment and Alzheimer's disease. Neurosci Lett. 2007;420:97–9.PubMedCrossRef Paquet C, Boissonnot M, Roger F, Dighiero P, Gil R, Hugon J. Abnormal retinal thickness in patients with mild cognitive impairment and Alzheimer's disease. Neurosci Lett. 2007;420:97–9.PubMedCrossRef
59.
Zurück zum Zitat Kesler A, Vakhapova V, Korczyn AD, Naftaliev E, Neudorfer M. Retinal thickness in patients with mild cognitive impairment and Alzheimer's disease. Clin Neurol Neurosurg. 2011;113:523–6.PubMedCrossRef Kesler A, Vakhapova V, Korczyn AD, Naftaliev E, Neudorfer M. Retinal thickness in patients with mild cognitive impairment and Alzheimer's disease. Clin Neurol Neurosurg. 2011;113:523–6.PubMedCrossRef
60.
Zurück zum Zitat Ascaso FJ, Cruz N, Modrego PJ, Santabárbara J, Pascual LF, Lobo A, et al. Retinal alterations in mild cognitive impairment and Alzheimer's disease: an optical coherence tomography study. J Neurol. 2014;261:1522–30.PubMedCrossRef Ascaso FJ, Cruz N, Modrego PJ, Santabárbara J, Pascual LF, Lobo A, et al. Retinal alterations in mild cognitive impairment and Alzheimer's disease: an optical coherence tomography study. J Neurol. 2014;261:1522–30.PubMedCrossRef
61.
Zurück zum Zitat Gao L, Liu Y, Li X, Bai Q, Liu P. Abnormal retinal nerve fiber layer thickness and macula lutea in patients with mild cognitive impairment and Alzheimer's disease. Arch Gerontol Geriatr. 2015;60:162–7.PubMedCrossRef Gao L, Liu Y, Li X, Bai Q, Liu P. Abnormal retinal nerve fiber layer thickness and macula lutea in patients with mild cognitive impairment and Alzheimer's disease. Arch Gerontol Geriatr. 2015;60:162–7.PubMedCrossRef
62.
Zurück zum Zitat Oktem EO, Derle E, Kibaroglu S, Oktem C, Akkoyun I, Can U. The relationship between the degree of cognitive impairment and retinal nerve fiber layer thickness. Neurol Sci. 2015;36:1141–6.PubMedCrossRef Oktem EO, Derle E, Kibaroglu S, Oktem C, Akkoyun I, Can U. The relationship between the degree of cognitive impairment and retinal nerve fiber layer thickness. Neurol Sci. 2015;36:1141–6.PubMedCrossRef
63.
Zurück zum Zitat Cheung CY, Ong YT, Hilal S, Ikram MK, Low S, Ong YL, et al. Retinal ganglion cell analysis using high-definition optical coherence tomography in patients with mild cognitive impairment and Alzheimer's disease. J Alzheimers Dis. 2015;45:45–56.PubMedCrossRef Cheung CY, Ong YT, Hilal S, Ikram MK, Low S, Ong YL, et al. Retinal ganglion cell analysis using high-definition optical coherence tomography in patients with mild cognitive impairment and Alzheimer's disease. J Alzheimers Dis. 2015;45:45–56.PubMedCrossRef
64.
Zurück zum Zitat Shao Y, Jiang H, Wei Y, Shi Y, Shi C, Wright CB, et al. Visualization of focal thinning of the ganglion cell-inner plexiform layer in patients with mild cognitive impairment and Alzheimer's disease. J Alzheimers Dis. 2018;64:1261–73.PubMedCrossRef Shao Y, Jiang H, Wei Y, Shi Y, Shi C, Wright CB, et al. Visualization of focal thinning of the ganglion cell-inner plexiform layer in patients with mild cognitive impairment and Alzheimer's disease. J Alzheimers Dis. 2018;64:1261–73.PubMedCrossRef
65.
Zurück zum Zitat Almeida ALM, Pires LA, Figueiredo EA, Costa-Cunha LVF, Zacharias LC, Preti RC, et al. Correlation between cognitive impairment and retinal neural loss assessed by swept-source optical coherence tomography in patients with mild cognitive impairment. Alzheimers Dement (Amst). 2019;11:659–69.CrossRef Almeida ALM, Pires LA, Figueiredo EA, Costa-Cunha LVF, Zacharias LC, Preti RC, et al. Correlation between cognitive impairment and retinal neural loss assessed by swept-source optical coherence tomography in patients with mild cognitive impairment. Alzheimers Dement (Amst). 2019;11:659–69.CrossRef
66.
Zurück zum Zitat Santos CY, Johnson LN, Sinoff SE, Festa EK, Heindel WC, Snyder PJ. Change in retinal structural anatomy during the preclinical stage of Alzheimer's disease. Alzheimers Dement (Amst). 2018;10:196–209.PubMedPubMedCentralCrossRef Santos CY, Johnson LN, Sinoff SE, Festa EK, Heindel WC, Snyder PJ. Change in retinal structural anatomy during the preclinical stage of Alzheimer's disease. Alzheimers Dement (Amst). 2018;10:196–209.PubMedPubMedCentralCrossRef
67.
Zurück zum Zitat López-Cuenca I, de Hoz R, Salobrar-García E, Elvira-Hurtado L, Rojas P, Fernández-Albarral JA, et al. Macular thickness decrease in asymptomatic subjects at high genetic risk of developing Alzheimer's disease: an OCT study. J Clin Med. 2020;9:1728.PubMedCentralCrossRef López-Cuenca I, de Hoz R, Salobrar-García E, Elvira-Hurtado L, Rojas P, Fernández-Albarral JA, et al. Macular thickness decrease in asymptomatic subjects at high genetic risk of developing Alzheimer's disease: an OCT study. J Clin Med. 2020;9:1728.PubMedCentralCrossRef
68.
Zurück zum Zitat Snyder PJ, Johnson LN, Lim YY, Santos CY, Alber J, Maruff P, Fernández B, et al. Nonvascular retinal imaging markers of preclinical Alzheimer's disease. Alzheimers Dement (Amst). 2016;4:169–78.PubMedPubMedCentralCrossRef Snyder PJ, Johnson LN, Lim YY, Santos CY, Alber J, Maruff P, Fernández B, et al. Nonvascular retinal imaging markers of preclinical Alzheimer's disease. Alzheimers Dement (Amst). 2016;4:169–78.PubMedPubMedCentralCrossRef
69.
Zurück zum Zitat van de Kreeke JA, Nguyen HT, den Haan J, Konijnenberg E, Tomassen J, den Braber A, et al. Retinal layer thickness in preclinical Alzheimer's disease. Acta Ophthalmol. 2019;97:798–804.PubMedPubMedCentralCrossRef van de Kreeke JA, Nguyen HT, den Haan J, Konijnenberg E, Tomassen J, den Braber A, et al. Retinal layer thickness in preclinical Alzheimer's disease. Acta Ophthalmol. 2019;97:798–804.PubMedPubMedCentralCrossRef
70.
Zurück zum Zitat van de Kreeke JA, Nguyen HT, Konijnenberg E, Tomassen J, den Braber A, Ten Kate M, et al. Longitudinal retinal layer changes in preclinical Alzheimer's disease. Acta Ophthalmol. 2020;18. van de Kreeke JA, Nguyen HT, Konijnenberg E, Tomassen J, den Braber A, Ten Kate M, et al. Longitudinal retinal layer changes in preclinical Alzheimer's disease. Acta Ophthalmol. 2020;18.
71.
Zurück zum Zitat Bulut M, Kurtulus F, Gozkaya O, Erol MK, Cengiz A, Akıdan M, et al. Evaluation of optical coherence tomography angiographic findings in Alzheimer's type dementia. Br J Ophthalmol. 2018;102:233–7.PubMedCrossRef Bulut M, Kurtulus F, Gozkaya O, Erol MK, Cengiz A, Akıdan M, et al. Evaluation of optical coherence tomography angiographic findings in Alzheimer's type dementia. Br J Ophthalmol. 2018;102:233–7.PubMedCrossRef
72.
Zurück zum Zitat Lahme L, Esser EL, Mihailovic N, Schubert F, Lauermann J, Johnen A, et al. Evaluation of ocular perfusion in Alzheimer's disease using optical coherence tomography angiography. J Alzheimers Dis. 2018;66:1745–52.PubMedCrossRef Lahme L, Esser EL, Mihailovic N, Schubert F, Lauermann J, Johnen A, et al. Evaluation of ocular perfusion in Alzheimer's disease using optical coherence tomography angiography. J Alzheimers Dis. 2018;66:1745–52.PubMedCrossRef
73.
Zurück zum Zitat Jiang H, Wei Y, Shi Y, Wright CB, Sun X, Gregori G, et al. Altered macular microvasculature in mild cognitive impairment and Alzheimer disease. J Neuroophthalmol. 2018;38:292–8.PubMedPubMedCentralCrossRef Jiang H, Wei Y, Shi Y, Wright CB, Sun X, Gregori G, et al. Altered macular microvasculature in mild cognitive impairment and Alzheimer disease. J Neuroophthalmol. 2018;38:292–8.PubMedPubMedCentralCrossRef
74.
Zurück zum Zitat Yoon SP, Grewal DS, Thompson AC, Polascik BW, Dunn C, Burke JR, et al. Retinal microvascular and neurodegenerative changes in Alzheimer's disease and mild cognitive impairment compared with control participants. Ophthalmol Retina. 2019;3:489–99.PubMedPubMedCentralCrossRef Yoon SP, Grewal DS, Thompson AC, Polascik BW, Dunn C, Burke JR, et al. Retinal microvascular and neurodegenerative changes in Alzheimer's disease and mild cognitive impairment compared with control participants. Ophthalmol Retina. 2019;3:489–99.PubMedPubMedCentralCrossRef
75.
Zurück zum Zitat Zhang YS, Zhou N, Knoll BM, Samra S, Ward MR, Weintraub S, et al. Parafoveal vessel loss and correlation between peripapillary vessel density and cognitive performance in amnestic mild cognitive impairment and early Alzheimer's disease on optical coherence tomography angiography. PLoS One. 2019;14:e0214685.PubMedPubMedCentralCrossRef Zhang YS, Zhou N, Knoll BM, Samra S, Ward MR, Weintraub S, et al. Parafoveal vessel loss and correlation between peripapillary vessel density and cognitive performance in amnestic mild cognitive impairment and early Alzheimer's disease on optical coherence tomography angiography. PLoS One. 2019;14:e0214685.PubMedPubMedCentralCrossRef
76.
Zurück zum Zitat Wu J, Zhang X, Azhati G, Li T, Xu G, Liu F. Retinal microvascular attenuation in mental cognitive impairment and Alzheimer's disease by optical coherence tomography angiography. Acta Ophthalmol. 2020;98:e781–7.PubMed Wu J, Zhang X, Azhati G, Li T, Xu G, Liu F. Retinal microvascular attenuation in mental cognitive impairment and Alzheimer's disease by optical coherence tomography angiography. Acta Ophthalmol. 2020;98:e781–7.PubMed
77.
Zurück zum Zitat O'Bryhim BE, Apte RS, Kung N, Coble D, Van Stavern GP. Association of preclinical Alzheimer disease with optical coherence tomographic angiography findings. JAMA Ophthalmol. 2018;136:1242–8.PubMedPubMedCentralCrossRef O'Bryhim BE, Apte RS, Kung N, Coble D, Van Stavern GP. Association of preclinical Alzheimer disease with optical coherence tomographic angiography findings. JAMA Ophthalmol. 2018;136:1242–8.PubMedPubMedCentralCrossRef
78.
Zurück zum Zitat van de Kreeke JA, Nguyen HT, Konijnenberg E, Tomassen J, den Braber A, Ten Kate M, et al. Optical coherence tomography angiography in preclinical Alzheimer's disease. Br J Ophthalmol. 2020;104:157–61.PubMedCrossRef van de Kreeke JA, Nguyen HT, Konijnenberg E, Tomassen J, den Braber A, Ten Kate M, et al. Optical coherence tomography angiography in preclinical Alzheimer's disease. Br J Ophthalmol. 2020;104:157–61.PubMedCrossRef
79.
Zurück zum Zitat Thomson KL, Yeo JM, Waddell B, Cameron JR, Pal S. A systematic review and meta-analysis of retinal nerve fiber layer change in dementia, using optical coherence tomography. Alzheimers Dement (Amst). 2015;1:136–43.PubMedPubMedCentralCrossRef Thomson KL, Yeo JM, Waddell B, Cameron JR, Pal S. A systematic review and meta-analysis of retinal nerve fiber layer change in dementia, using optical coherence tomography. Alzheimers Dement (Amst). 2015;1:136–43.PubMedPubMedCentralCrossRef
80.
Zurück zum Zitat Alber J, Goldfarb D, Thompson LI, Arthur E, Hernandez K, Cheng D, et al. Developing retinal biomarkers for the earliest stages of Alzheimer's disease: what we know, what we don't, and how to move forward. Alzheimers Dement. 2020;16:229–43.PubMedCrossRef Alber J, Goldfarb D, Thompson LI, Arthur E, Hernandez K, Cheng D, et al. Developing retinal biomarkers for the earliest stages of Alzheimer's disease: what we know, what we don't, and how to move forward. Alzheimers Dement. 2020;16:229–43.PubMedCrossRef
81.
Zurück zum Zitat Chan VTT, Sun Z, Tang S, Chen LJ, Wong A, Tham CC, et al. Spectral-domain OCT measurements in Alzheimer's disease: a systematic review and meta-analysis. Ophthalmology. 2019;126:497–510.PubMedCrossRef Chan VTT, Sun Z, Tang S, Chen LJ, Wong A, Tham CC, et al. Spectral-domain OCT measurements in Alzheimer's disease: a systematic review and meta-analysis. Ophthalmology. 2019;126:497–510.PubMedCrossRef
82.
Zurück zum Zitat Ito Y, Sasaki M, Takahashi H, Nozaki S, Matsuguma S, Motomura K, et al. Quantitative assessment of the retina using OCT and associations with cognitive function. Ophthalmology. 2020;127:107–18.PubMedCrossRef Ito Y, Sasaki M, Takahashi H, Nozaki S, Matsuguma S, Motomura K, et al. Quantitative assessment of the retina using OCT and associations with cognitive function. Ophthalmology. 2020;127:107–18.PubMedCrossRef
83.
Zurück zum Zitat Ward DD, Mauschitz MM, Bönniger MM, Merten N, Finger RP, Breteler MMB. Association of retinal layer measurements and adult cognitive function: a population-based study. Neurology. 2020;95:e1144–52.PubMedCrossRef Ward DD, Mauschitz MM, Bönniger MM, Merten N, Finger RP, Breteler MMB. Association of retinal layer measurements and adult cognitive function: a population-based study. Neurology. 2020;95:e1144–52.PubMedCrossRef
84.
Zurück zum Zitat Sen S, Saxena R, Vibha D, Tripathi M, Sharma P, Phuljhele S, et al. Detection of structural and electrical disturbances in macula and optic nerve in Alzheimer's patients and their correlation with disease severity. Semin Ophthalmol. 2020;35:116–25.PubMedCrossRef Sen S, Saxena R, Vibha D, Tripathi M, Sharma P, Phuljhele S, et al. Detection of structural and electrical disturbances in macula and optic nerve in Alzheimer's patients and their correlation with disease severity. Semin Ophthalmol. 2020;35:116–25.PubMedCrossRef
85.
Zurück zum Zitat Mwanza JC, Oakley JD, Budenz DL, Chang RT, Knight OJ, Feuer WJ. Macular ganglion cell-inner plexiform layer: automated detection and thickness reproducibility with spectral domain-optical coherence tomography in glaucoma. Invest Ophthalmol Vis Sci. 2011;52:8323–9.PubMedPubMedCentralCrossRef Mwanza JC, Oakley JD, Budenz DL, Chang RT, Knight OJ, Feuer WJ. Macular ganglion cell-inner plexiform layer: automated detection and thickness reproducibility with spectral domain-optical coherence tomography in glaucoma. Invest Ophthalmol Vis Sci. 2011;52:8323–9.PubMedPubMedCentralCrossRef
86.
Zurück zum Zitat Wu Y, Wang XN, Wang N, Han Y, Ma D, Lu Y. Regularity changes of the retinal nerve fiber layer and macular ganglion cell complex in patients with the amnestic mild cognitive impairment. Int J Neurosci. 2018;128:849–53.PubMedCrossRef Wu Y, Wang XN, Wang N, Han Y, Ma D, Lu Y. Regularity changes of the retinal nerve fiber layer and macular ganglion cell complex in patients with the amnestic mild cognitive impairment. Int J Neurosci. 2018;128:849–53.PubMedCrossRef
87.
Zurück zum Zitat Lad EM, Mukherjee D, Stinnett SS, Cousins SW, Potte GG, Burke JR, et al. Evaluation of inner retinal layers as biomarkers in mild cognitive impairment to moderate Alzheimer's disease. PLoS One. 2018;13:e0192646.PubMedPubMedCentralCrossRef Lad EM, Mukherjee D, Stinnett SS, Cousins SW, Potte GG, Burke JR, et al. Evaluation of inner retinal layers as biomarkers in mild cognitive impairment to moderate Alzheimer's disease. PLoS One. 2018;13:e0192646.PubMedPubMedCentralCrossRef
88.
Zurück zum Zitat Nilson AN, English KC, Gerson JE, Barton Whittle T, Nicolas Crain C, et al. Tau oligomers associate with inflammation in the brain and retina of tauopathy mice and in neurodegenerative diseases. J Alzheimers Dis. 2017;55:1083–99.PubMedCrossRef Nilson AN, English KC, Gerson JE, Barton Whittle T, Nicolas Crain C, et al. Tau oligomers associate with inflammation in the brain and retina of tauopathy mice and in neurodegenerative diseases. J Alzheimers Dis. 2017;55:1083–99.PubMedCrossRef
89.
Zurück zum Zitat Salobrar-García E, de Hoz R, Ramírez AI, López-Cuenca I, Rojas P, Vazirani R, et al. Changes in visual function and retinal structure in the progression of Alzheimer's disease. PLoS One. 2019;14:e0220535.PubMedPubMedCentralCrossRef Salobrar-García E, de Hoz R, Ramírez AI, López-Cuenca I, Rojas P, Vazirani R, et al. Changes in visual function and retinal structure in the progression of Alzheimer's disease. PLoS One. 2019;14:e0220535.PubMedPubMedCentralCrossRef
90.
Zurück zum Zitat Uchida A, Pillai JA, Bermel R, Bonner-Jackson A, Rae-Grant A, Fernandez H, et al. Outer retinal assessment using spectral-domain optical coherence tomography in patients with Alzheimer's and Parkinson's disease. Invest Ophthalmol Vis Sci. 2018;59:2768–77.PubMedPubMedCentralCrossRef Uchida A, Pillai JA, Bermel R, Bonner-Jackson A, Rae-Grant A, Fernandez H, et al. Outer retinal assessment using spectral-domain optical coherence tomography in patients with Alzheimer's and Parkinson's disease. Invest Ophthalmol Vis Sci. 2018;59:2768–77.PubMedPubMedCentralCrossRef
91.
Zurück zum Zitat Asanad S, Ross-Cisneros FN, Nassisi M, Barron E, Karanjia R, Sadun AA. The retina in Alzheimer's disease: histomorphometric analysis of an ophthalmologic biomarker. Invest Ophthalmol Vis Sci. 2019;60:1491–500.PubMedPubMedCentralCrossRef Asanad S, Ross-Cisneros FN, Nassisi M, Barron E, Karanjia R, Sadun AA. The retina in Alzheimer's disease: histomorphometric analysis of an ophthalmologic biomarker. Invest Ophthalmol Vis Sci. 2019;60:1491–500.PubMedPubMedCentralCrossRef
92.
Zurück zum Zitat Grewal DS, Polascik BW, Hoffmeyer GC, Fekrat S. Assessment of differences in retinal microvasculature using OCT angiography in Alzheimer's disease: a twin discordance report. Ophthalmic Surg Lasers Imaging Retina. 2018;49:440–4.PubMedCrossRef Grewal DS, Polascik BW, Hoffmeyer GC, Fekrat S. Assessment of differences in retinal microvasculature using OCT angiography in Alzheimer's disease: a twin discordance report. Ophthalmic Surg Lasers Imaging Retina. 2018;49:440–4.PubMedCrossRef
Metadaten
Titel
Advances in retina imaging as potential biomarkers for early diagnosis of Alzheimer’s disease
verfasst von
Ying Zhang
Yanjiang Wang
Ce Shi
Meixiao Shen
Fan Lu
Publikationsdatum
01.12.2021
Verlag
BioMed Central
Erschienen in
Translational Neurodegeneration / Ausgabe 1/2021
Elektronische ISSN: 2047-9158
DOI
https://doi.org/10.1186/s40035-021-00230-9

Weitere Artikel der Ausgabe 1/2021

Translational Neurodegeneration 1/2021 Zur Ausgabe

Neu in den Fachgebieten Neurologie und Psychiatrie

Typ-2-Diabetes und Depression folgen oft aufeinander

14.05.2024 Typ-2-Diabetes Nachrichten

Menschen mit Typ-2-Diabetes sind überdurchschnittlich gefährdet, in den nächsten Jahren auch noch eine Depression zu entwickeln – und umgekehrt. Besonders ausgeprägt ist die Wechselbeziehung laut GKV-Daten bei jüngeren Erwachsenen.

Schützt Olivenöl vor dem Tod durch Demenz?

10.05.2024 Morbus Alzheimer Nachrichten

Konsumieren Menschen täglich 7 Gramm Olivenöl, ist ihr Risiko, an einer Demenz zu sterben, um mehr als ein Viertel reduziert – und dies weitgehend unabhängig von ihrer sonstigen Ernährung. Dafür sprechen Auswertungen zweier großer US-Studien.

Darf man die Behandlung eines Neonazis ablehnen?

08.05.2024 Gesellschaft Nachrichten

In einer Leseranfrage in der Zeitschrift Journal of the American Academy of Dermatology möchte ein anonymer Dermatologe bzw. eine anonyme Dermatologin wissen, ob er oder sie einen Patienten behandeln muss, der eine rassistische Tätowierung trägt.

Chirurginnen und Chirurgen sind stark suizidgefährdet

07.05.2024 Suizid Nachrichten

Der belastende Arbeitsalltag wirkt sich negativ auf die psychische Gesundheit der Angehörigen ärztlicher Berufsgruppen aus. Chirurginnen und Chirurgen bilden da keine Ausnahme, im Gegenteil.