Научная статья на тему 'Retinal Optical Coherence Tomography in Neurodegenerative Diseases (Review)'

Retinal Optical Coherence Tomography in Neurodegenerative Diseases (Review) Текст научной статьи по специальности «Медицинские технологии»

CC BY
474
270
i Надоели баннеры? Вы всегда можете отключить рекламу.
Ключевые слова
Parkinson’s disease / Alzheimer’s disease / retina / biomarker / optical coherence tomography / neurodegeneration

Аннотация научной статьи по медицинским технологиям, автор научной работы — S.N. Svetozarskiy, S.V. Kopishinskaya

Increased aging of the population makes problems of the diagnosis and treatment of neurodegenerative diseases socially more significant. The ability to use the retina as a “window” to the central nervous system has attracted great attention in recent years. Optical coherence tomography (OCT) is a non-invasive method for in vivo studies of various conditions to generate high-resolution images of the tissue cross sections under study. Retinal OCT parameters are considered to be potential surrogate biomarkers of early-stage neurodegenerative disorders, and have already been included in the guidelines for diagnosing neuromyelitis optica. This review summarizes and analyzes the current information on retinal changes according to OCT data in neurodegeneration in vitro and in vivo in Alzheimer’s and Parkinson’s diseases. The application of ultra-high resolution OCT for the diagnosis of the early stages of neurodegeneration is also considered. Morphological and functional links and possible mechanisms for the retinal lesions in Alzheimer’s and Parkinson’s diseases, and their similarities in glaucoma are discussed. The efficacy of using this method in the diagnosis of neurodegenerative processes at an early stage is likely to be increased by the development of instrumentation and improvements in the design study for carrying out investigations in different groups of patients, including those having hereditary diseases of the nervous system.

i Надоели баннеры? Вы всегда можете отключить рекламу.
iНе можете найти то, что вам нужно? Попробуйте сервис подбора литературы.
i Надоели баннеры? Вы всегда можете отключить рекламу.

Текст научной работы на тему «Retinal Optical Coherence Tomography in Neurodegenerative Diseases (Review)»

Retinal Optical Coherence Tomography

in Neurodegenerative Diseases (Review)

DOI 10.17691/stm2015.7.1.14

Received October 16, 2014

S.N. Svetozarskiy, Clinical Resident, Department of Eye Diseases;

S.V. Kopishinskaya, PhD, Associate Professor, Department of Neurology, Psychiatry and Addiction Medicine,

Postgraduate Faculty

Nizhny Novgorod State Medical Academy, 10/1 Minin and Pozharsky Square, Nizhny Novgorod, 603005,

Russian Federation

Increased aging of the population makes problems of the diagnosis and treatment of neurodegenerative diseases socially more significant.

The ability to use the retina as a “window” to the central nervous system has attracted great attention in recent years. Optical coherence

tomography (OCT) is a non-invasive method for in vivo studies of various conditions to generate high-resolution images of the tissue

cross sections under study. Retinal OCT parameters are considered to be potential surrogate biomarkers of early-stage neurodegenerative

disorders, and have already been included in the guidelines for diagnosing neuromyelitis optica. This review summarizes and analyzes the

current information on retinal changes according to OCT data in neurodegeneration in vitro and in vivo in Alzheimer’s and Parkinson’s diseases.

The application of ultra-high resolution OCT for the diagnosis of the early stages of neurodegeneration is also considered. Morphological and

functional links and possible mechanisms for the retinal lesions in Alzheimer’s and Parkinson’s diseases, and their similarities in glaucoma are

discussed. The efficacy of using this method in the diagnosis of neurodegenerative processes at an early stage is likely to be increased by the

development of instrumentation and improvements in the design study for carrying out investigations in different groups of patients, including

those having hereditary diseases of the nervous system.

Key words: retina; biomarker; optical coherence tomography; Parkinson’s disease; Alzheimer’s disease; neurodegeneration.

Introduction. An increasingly ageing population in developed countries has made the problem of diagnosis and treatment of neurodegenerative diseases especially topical. Currently, numerous neuroprotective drugs are being developed, while reliable biomarkers and methods for monitoring the neurodegenerative process are scanty [1]. There are several markers which indirectly reflect the activity the process of apoptosis. In particular, there are methods for determining the concentration of pro-apoptotic proteins in the blood, but these are not widely used in medical practice. Magnetic resonance imaging is not a reliable method of neuronal degeneration evaluation, because it does not show a high level of correlation with the activity of the neurodegenerative process in short-term studies [2]. In neurodegenerative diseases the progressive loss of neurons increases with age, but no objective method for the measurement of this has been developed.

The retina and optic nerve form part of the central nervous system (CNS). The first three types of neurons of the visual analyzer — the photoreceptors, bipolar and ganglion cells, forming a vertical pathway of signal transmission, are located in the retina [3]. Between them there are amacrine and horizontal cells, which provide signal transmission and modulation in the horizontal direction [4]. The retinal nerve fiber layer consists of unmyelinated regions of the axons of the ganglion cells, which are covered with myelin where they leave the eyeball via the mesh-like plate of the sclera, and form the optic nerve. The retina as a “window” to the CNS

has attracted special attention in recent years [5-9]. There are several immutable anatomical prerequisites for this: the physiological transparency of the eyeball media, the unique structure of the retina with its low concentration of glial cells, and the absence of myelin. Furthermore, advances in science and technology have led to the development of new bioimaging techniques such as optical coherence tomography (OCT).

Optical coherence tomography. OCT is a non-invasive method for in vivo studies that yields high-resolution cross sectional images of tissues. The cross section image is made by combining multiple axial depth reflectivity profiles (A-scans). The image quality depends on the speed A-scans acquisition and their resolution [10-12]. The first commercially available OCT devices had a speed of about 400 A-scans per second, axial resolutions of 8-10 |jm and a depth sufficient to explore all the layers of the retina. Modern high and ultra-high resolution OCT devices provide speeds of 20,000-52,000 scans per second and resolutions of better than 1-4 |jm. Their high speed makes it possible to reduce the number of motion artifacts, while the high resolution enables research to be carried out, not only at a tissue level, but at cellular and subcellular levels as well. The depth of scanning has also increased, so choroid structure investigations have become feasible [13, 14].

In recent years retinal OCT has increasingly been used in the study of neurological diseases. Firstly for demyelinating diseases — multiple sclerosis and neuromyelitis optica,

Corresponding author: Svetozarskiy Sergey, e-mail: svetozarskij@rambler.ru

уттжтжтжтжттттжтжтжтжтттт^ттттжтттт^тттт^тититттттиттг/'

116 STM J 2015 — vol. 7, No.1 S.N. Svetozarskiy, S.V. Kopishinskaya

REVIEWS OF UTILIZATION

and now the use of retinal OCT in these diseases has been recognised and is being actively developed [15-20]. Assessment of the retinal nerve fiber layer thickness is included in the guidelines for diagnosing neuromyelitis optica [21]. Research is being carried on in groups of patients with Alzheimer's and Parkinson's diseases, mild cognitive syndrome, various forms of optic nerve atrophy, stroke, spinocerebellar ataxia, hereditary diseases of the CNS, obstructive sleep apnea and prion diseases (Creutzfeldt-Jakob disease) [22-29].

This method is promising in relation to monitoring the activity of neurodegenerative processes in the course of studying the efficacy of pharmaceutical preparations [7, 30]. The high reproducibility of the results of examinations of the same patient by different specialists, and a high index of agreement by experts on the data obtained, makes OCT a well-suited technique for multi-center studies [3135]. The prevalence of diffuse retinal changes over local abnormalities in neurodegenerative disorders determines the final conclusion made according to the findings of the OCT technique: focusing not on a qualitative description of optical phenomena, but mainly on the retinal thickness and its volume parameters. In this regard, most commonly used standard protocols include a quantitative assessment of the retinal ganglion cell layer and retinal nerve fiber layer thickness presented by the axons of the ganglion cells, and of the total macular volume (total volume of retinal tissue in the yellow spot area (macula) within a disc of a given diameter). This review summarizes the experience of applying OCT for the diagnosis of retinal neurodegenerative processes in experimental work and in the most common neurodegenerative diseases — Alzheimer's and Parkinson's diseases.

Experimental OCT diagnosis of retinal neurodegeneration. Damage to major subcellular organelles — the nucleus, mitochondria, Golgi apparatus — provides information regarding the onset of programmed cell death, apoptosis, much sooner than the appearance of the first changes at a cellular level. Under physiological conditions, the mitochondria in a cell are working as a structural and functional network. When a programme of apoptosis is trigged, a collapse of the mitochondrial network occurs, and the mitochondria become independent structures. Soon structural changes occur, including an increase in the permeability of the mitochondrial membrane, leading to the release of apoptotic molecules, such as cytochrome C, into the cytosol, and caspase cascade activation takes place [36, 37].

When studying apoptosis in cultured retinal ganglion cells, Tudor et al. [38] found a number of early morphological and optical changes. The mitochondrial network degradation when the mitochondria become independent structures occurs within 20 min of exposure to staurosporine, and after 60 min, the release of cytochrome C, which is an indicator of mitochondrial membrane disintegration, can be detected. Thus, data has been obtained in the time “window” between the beginning of the functional changes and the structural ones in the mitochondria. Using these morphological findings, the authors have developed a programme for analysis using an ultra-high resolution

OCT device, which allows them, automatically, to be able to distinguish between a control sample, a sample in the early stages of apoptosis 20 min after the staurosporine exposure, and a sample 30-60 min after such exposure, with an accuracy of 85%. The first stage involves the initial processing to remove noise, detect the cover-glass position and to correct imaging artifacts, caused by any tilting of the object under test. Then, the part of the image, containing the greatest number of cells (the region of interest, ROI), is selected automatically on the basis of the surrounding pixel intensities and adjustments for the cover-glass position. Each ROI is assessed, using 65 parameters such as entropy, range, standard deviation contrast, correlation coefficient, energy and homogeneity for each co-occurrence matrices along one of three X, Y, and Z coordinates, in order to reveal significant features in the OCT images of the retinal ganglion cells under physiological conditions and at different stages of apoptosis. Using a Gaussian mixture model a multidimensional distribution of the indicators, so-called feature space, is generated, in which clusters of healthy and apoptotic cell images are marked out, to provide the basis for the analysis programme. The distance from the point in the feature space, representing the cell clusters in the tested sample, to the healthy cell clusters point allows differentiation of the control sample from a sample at an early stage of apoptosis. The authors can state that the change in the optical parameters of the ganglion cell culture occurs, primarily, due to the reconfiguration of the mitochondrial network. Similar results were obtained in a study of retinal explants (ex vivo). Thus, this demonstrates the possibility of non-invasive quantitative assessment of the condition of retinal neurons using the OCT technique, and its application for the diagnosis of neurodegeneration at an early stage.

Retinal OCT has occupied an important place in experiments, for assessing the in vivo effects of neurotrophic drugs and the effects of cell therapy. In particular, OCT has been used for the study of retinal ganglion cell axon regeneration after optic nerve damage and subsequent stem cell therapy [39]. Experimental studies aimed at improving the quality of the images have played a significant role in the development of the OCT method and its application in the diagnosis of neurodegenerative processes. This may be achieved, not only by improving the technical performance of the device, but of the software as well; for example, by averaging a greater number of images [40]. Such techniques have helped to achieve a higher level of matching of the histological and optical picture from the internal limiting membrane of the retina to the outer layers of the choroid [41].

Retinal OCT in Alzheimer’s disease. Alzheimer's disease (AD) is a steadily progressing neurodegenerative disease in which neuritic plaques (accumulation of beta-amyloid) and neurofibrillary tangles (containing tau protein) are formed in the CNS [42, 43]. Visual symptoms are often early complaints in AD and are accompanied by such pathological changes in the structures of the optic tract, as the deposition of beta-amyloid, the deterioration of blood flow in the retinal vessels, vacuolar degeneration of the ganglion cells and their axons within the optic nerve,

Retinal Optical Coherence Tomography in Neurodegenerative Diseases STM J 2015 — vol. 7, No.1 117

REVIEWS OF UTILIZATION

reduction of retinal nerve fiber layer thickness and neuron atrophy in the occipital lobe [44-52].

There are different opinions concerning the decrease in retinal nerve fiber layer thickness in AD [53]. One of the hypotheses is based on the fact that the pathogenic process in AD occurs not only in the cortex, but also in other parts of the CNS, particularly in the retina. A reduction in the number of retinal ganglion cells by 36.4% [54] is accompanied by the growth of glial cells, and by the deposition of beta-amyloid and tau protein in various layers of the retina and the optic nerve, which is similar to the processes going on in the cortex [46]. However, such a deposition of AD-associated proteins and their precursors in the retina is observed not only in AD but also in retinitis pigmentosa, age-related macular degeneration, other diseases, and during normal ageing [55, 56]. A second hypothesis concerns secondary lesions of the retinal neurons, focusing on the mechanisms of retrograde transsynaptic degeneration in the direction from the geniculo-cortical to the retino-geniculate pathway and anterograde degeneration in the cortico-geniculate pathway [57-60]. A third hypothesis was inspired by epidemiological data: glaucoma develops more commonly, and has the most severe course, in patients with AD [6163]. Indeed, pathological processes in AD and normal pressure glaucoma are quite similar at the level of the lamina cribrosa — the exit site of the optic nerve [64]. The pressure gradient at the level of the lamina cribrosa is determined by the difference between the intraocular pressure and the retrobulbar pressure of the cerebrospinal fluid (CSF). An increase in the gradient due to raised intraocular pressure, or reduced CSF pressure, leads to lamina cribrosa deformation in the direction of the retrobulbar space and to damage of the retinal nerve fiber layer. In severe AD, there is a decrease in CSF pressure, which may be caused by a reduction in its rate of production in the choroid plexus of the brain ventricles [65]. In AD, in addition to age-related physiological atrophy, there is a marked stromal fibrosis and deposition of amyloid-beta, immunoglobulins and complement system fragments on the basal membrane of the choroid plexus [66]. As a result, the rates of CSF production and regeneration in AD are reduced, while the CSF volume increases due to the atrophy of the brain [67]. Each of these mechanisms — primary, retrograde degeneration and glaucoma degeneration of the retinal ganglion cell and their axons (retinal nerve fiber layer) is likely to play a role at particular stages of the development of AD. New studies, considering the subtype, severity and duration of the disease, are needed.

According to the OCT data, the reduction of the retinal nerve fiber layer thickness is already determined by the early stages of AD, and in mild cognitive syndrome, in the absence of changes in the visual field and visual acuity, and is progressing with the development of the disease [68-72]. In the course of a meta-analysis, which included seven studies and involved the eyes of 324 patients with AD, it was confirmed that, in AD, a statistically significant decrease in the thickness of the retinal nerve fiber layer occurs in all quadrants [73]. In the study of the macular volume [74] and the retinal ganglion cell layer thickness [69, 75] a statistically significant reduction was also found,

both parameters correlating with dementia (mini-mental state examination score, MMSE). A correlation has also been found between the nerve fiber layer thickness and a number of pattern-electroretinogram characteristics, with the P50-N95 amplitude, in particular [76]. Defects in the lower part of the visual field correspond to a greater reduction in nerve fiber layer thickness in the upper half of the retina [47]. No correlation was found between the retinal nerve fiber layer thickness and the investigation data on visual evoked potentials [77]. The role of choroid thickness as an AD biomarker is currently being studied. Statistically significant reductions in the choroid thickness in AD patients have been identified according both to OCT data and to histological studies [78, 79].

Retinal OCT in Parkinson’s disease. Parkinson's disease (PD) is a progressive multisystem neurodegenerative disease, which manifests itself through a number of motor and non-motor symptoms, developing due to the loss of dopaminergic neurons in the substantia nigra and other areas of the CNS [80]. In the retina, A18 amacrine cells are the main dopaminergic neurons [81]. They are involved in horizontal impulse transmission from the bipolar cells to other subtypes of amacrine cells, and further, to the retinal ganglion cells. The lack of dopamine results in disturbances to central vision, and to decreased colour vision and contrast sensitivity [82, 83]. Dopamine is also involved in retinal trophism, circadian rhythm formation, eye growth, and cell death [84-86]. Such symptoms as dry eyes, double vision, impairment of the visual searching functions, reading difficulties, fixation problems and complex visual hallucinations have been described in PD [80, 87-89].

The first study devoted to evaluating the nerve fiber layer thickness in PD, using the OCT technique, was performed in 2004 [90]. Most subsequent studies have shown a decrease in the retinal nerve fiber layer thickness and macular volume. In the meta-analysis, which included 13 studies, involving the eyes of 644 patients with PD, a statistically significant decrease in the retinal nerve fiber layer thickness in PD was found in all quadrants [91]. In analyzing the research results, the role of the device parameters must be taken into account. This is confirmed by the recent data obtained by Satue et al. [92] in a group of 153 patients with PD on two different OCT instruments — Cirrus and Spectralis. In both cases, the average retinal nerve fiber layer thickness was reduced in patients with PD compared to the control group. According to the information received through the Spectralis device, the retinal thickness in patients with PD was significantly reduced in all areas of the macula zone except for the fovea. However, with the Cirrus device, a difference in the thickness of the retina in the fovea and in two other areas was also revealed, but in the remaining six regions no statistically significant differences were found.

It is interesting to note that the majority of patients with PD have visual field defects similar to those in glaucoma, often in the absence of the nerve fiber layer thickness reduction at normal intraocular pressure [93]. In an OCT study, where all the retinal layers were measured [93], a statistically significant reduction was determined not only in nerve fiber layer thickness, but also in the ganglion cell layer, and the

118 STM J 2015 — vol. 7, No.1 S.N. Svetozarskiy, S.V. Kopishinskaya

REVIEWS OF UTILIZATION

inner and outer plexiform layers. Increased thickness was found in the inner nuclear layer (bipolar, amacrine, horizontal and MUller cells). Patients with secondary Parkinsonism syndrome were identified to have specific changes of retinal neuronal architecture [94]. Progressive supranuclear palsy is characterised by a thickening of the outer nuclear layer, and, in multisystem atrophy, in the outer plexiform layer; retinal thickness is decreased in both diseases.

Half of patients with PD were noted to have interocular asymmetry, with different retinal thickness in the fovea, according to the OCT data [95]. No differences were found in the papillomacular bundle thickness compared with the control group [92]. The authors found a statistically significant correlation between retinal thickness and the results of motor independence measurements according to the Schwab-England ADL scale, and the unified Parkinson's disease rating scale, UPDRS. In earlier works [96] a correlation between the foveal thickness and UPDRS measurements was also found. There was no correlation between the thickness of the inner retinal layers and contrast sensitivity [97].

Obviously, the axons are the first target in the neurodegenerative process, but our understanding of the relationships between the dopamine level, amacrine cells and the loss of ganglion cell axons in PD is still not complete.

OCT perspectives and limitations in the diagnosis of neurodegenerative processes. Diagnostic accuracy of OCT in retinal neurodegenerative diseases may be achieved only in the case of observing a particular judiciously selecting research algorithm [7]. The first step includes identification of a concomitant ophthalmic pathology: optically significant factors — cataracts, vitreous opacities, and diseases that damage the ganglion cell layer and nerve fiber layer — glaucoma, degenerative myopia, optic disk edema and a history of optic neuritis [98]. This requires thorough recording of the medical history and the availability of diagnostic tools in the clinic — a slit lamp, tonometer, refractometer, or their installation in the OCT device. Secondly, the physician must constantly control the quality of the images obtained [99-102]. Low signal intensity, incorrect selection of the study area, and patient tremor may significantly distort the results.

The first experience of the application of OCT in the diagnosis of neurodegenerative processes has revealed a number of unsolved problems, presenting a wide field for investigations. Databases of the normal values for each OCT device linked to the distribution by age, gender and ethnicity, are incomplete and need to be confirmed by more extensive research. Furthermore, mean values of the physiological reduction of retinal layer thickness with age should be collected in the course of a long-term follow-up.

Prospective application of this method in the diagnosis of neurodegenerative processes at an early stage is connected, on the one hand, with technological advances,

i.e. the introduction of ultra-high resolution OCT devices, allowing specialists to study the retinal structure at cellular and subcellular levels, and with achieving high quality images of the deep structures to study the role of the choroid in neuronal death. On the other hand, it is important

to improve the design of the research. In this respect continuing investigations in those groups of patients with different degrees of severity of disease and clinical phenotypes, and in gene carriers of hereditary diseases of the CNS, will help in the study of the early preclinical stages of neurodegenerative diseases.

Research Funding and Conflict of Interest. The study was not funded by any source, and there are no conflicts of interest related to this study.

Acknowledgments. We thank Prof. Igor Smetankin and Prof. Alexander Gustov for their help in scientific revision a manuscript.

References

1. Kramer P.A., Ravi S., Chacko B., Johnson M.S., Darley-Usmar V.M. A review of the mitochondrial and glycolytic metabolism in human platelets and leukocytes: implications for their use as bioenergetic biomarkers. Redox Biol 2014 Jan; 2: 206-210, http://dx.doi.org/10.10167j.redox.2013.12.026.

2. Daumer M., Neuhaus A., Morrissey S., Hintzen R., Ebers G.C. MRI as an outcome in multiple sclerosis clinical trials. Neurology 2009 Feb; 72(8): 705-711, http://dx.doi. org/10.1212/01.wnl.0000336916.38629.43.

3. Masland R.H. The neuronal organization of the retina. Neuron 2012 Oct; 76(2): 266-280, http://dx.doi.org/10.1016/ j.neuron.2012.10.002.

4. Thoreson W.B., Mangel S.C. Lateral interactions in the outer retina. Prog Retin Eye Res 2012 Sep; 31(5): 407-441, http://dx.doi.org/10.1016/j.preteyeres.2012.04.003.

5. Bambo M.P., Garcia-Martin E., Pinilla J., Herrero R., Satue M., Otin S., Fuertes I., Marques M.L., Pablo L.E. Detection of retinal nerve fiber layer degeneration in patients with Alzheimer's disease using optical coherence tomography: searching new biomarkers. Acta Ophthalmol 2014 Nov; 92(7): e581-e582, http://dx.doi.org/doi:10.1111/aos.12374.

6. Fairless R., Williams S.K., Hoffmann D.B., Stojic A., Hochmeister S., Schmitz F., Storch M.K., Diem R. Preclinical retinal neurodegeneration in a model of multiple sclerosis. J Neurosci 2012 Apr; 32(16): 5585-5597, http://dx.doi. org/10.1523/JNEUROSCI.5705-11.2012.

7. Greenberg B.M., Frohman E. Optical coherence tomography as a potential readout in clinical trials. Ther Adv Neurol Disord 2010 May; 3(3): 153-60, http://dx.doi.org/10.117 7/1756285610368890.

8. Guo L., Duggan J., Cordeiro M.F. Alzheimer's disease and retinal neurodegeneration. Curr Alzheimer Res 2010 Feb; 7(1): 3-14, http://dx.doi.org/10.2174/156720510790274491.

9. Fjeldstad A.S., Carlson N.G., Rose J.W. Optical coherence tomography as a biomarker in multiple sclerosis. Expert Opin MedDiagn 2012 Nov; 6(6): 593-604, http://dx.doi. org/10.1517/17530059.2012.719496.

10. Fujimoto J.G., Brezinski M.E., Tearney G.J., Boppart S.A., Bouma B., Hee M.R., Southern J.F., Swanson E.A. Optical biopsy and imaging using optical coherence tomography. Nat Med 1995 Sep; 1(9): 970-972, http://dx.doi. org/10.1038/nm0995-970.

11. Abtahian F., Jang I.K. Optical coherence tomography: basics, current application and future potential. Curr Opin Pharmacol 2012 Oct; 12(5): 583-591, http://dx.doi.org/10.1016/ j.coph.2012.07.015.

12. Gladkova N.D. Opticheskaya kogerentnaya tomografiya

'шшттшттштштшттшттштштшттшттштштшттшттштштшттштш/ш/ы

Retinal Optical Coherence Tomography in Neurodegenerative Diseases STM J 2015 — vol. 7, No.1 119

REVIEWS OF UTILIZATION

v ryadu metodov meditsinskoy vizualizatsii [Optical coherence tomography in a series of medical imaging]. Nizhny Novgorod: Institute of Applied Physics RAS; 2005; 324 p.

13. Adhi M., Duker J.S. Optical coherence tomography —

current and future applications. Curr Opin Ophthalmol 2013 May; 24(3): 213-221, http://dx.doi.org/10.1097/

ICU.0b013e32835f8bf8.

14. Regatieri C.V., Branchini L., Fujimoto J.G., Duker J.S. Choroidal imaging using spectral-domain optical coherence tomography. Retina 2012 May; 32(5): 865-876, http://dx.doi. org/doi:10.1097/IAE.0b013e318251a3a8.

15. Parisi V., Manni G., Spadaro M., Colacino G., Restuccia R., Marchi S., Bucci M.G., Pierelli F. Correlation between morphological and functional retinal impairment in multiple sclerosis patients. Invest Ophthalmol Vis Sci 1999 Oct; 40(11): 2520-2527.

16. Wang X., Jia Y., Spain R., Potsaid B., Liu J.J.,

Baumann B., Hornegger J., Fujimoto J.G., Wu Q., Huang D. Optical coherence tomography angiography of optic nerve head and parafovea in multiple sclerosis. Br J Ophthalmol 2014; 98(10): 1368-1373, http://dx.doi.org/doM0.1136/

bjophthalmol-2013-304547.

17. Garcia-Martin E., Calvo B., Malve M., Herrero R., Fuertes I., Ferreras A., Larrosa J.M., Polo V., Pablo L.E. Three-dimensional geometries representing the retinal nerve fiber layer in multiple sclerosis, optic neuritis, and healthy eyes. Ophthalmic Res 2013; 50(1): 72-81, http://dx.doi. org/10.1159/000350413.

18. Balk L., Tewarie P., Killestein J., Polman C., Uitdehaag B., Petzold A. Disease course heterogeneity and OCT in multiple sclerosis. Mult Scler 2014 Jan; 20(9): 11981206, http://dx.doi.org/10.1177/1352458513518626.

19. Balk L.J., Twisk J.W., Steenwijk M.D., Daams M., Tewarie P., Killestein J., Uitdehaag B.M., Polman C.H., Petzold A. A dam for retrograde axonal degeneration in multiple sclerosis? J Neurol Neurosurg Psychiatry 2014 Jul; 85(7): 782-789, http://dx.doi.org/10.1136/jnnp-2013-306902.

20. Schinzel J., Zimmermann H., Paul F., Ruprecht K., Hahn K., Brandt A.U., Dorr J. Relations of low contrast visual acuity, quality of life and multiple sclerosis functional composite: a cross-sectional analysis. BMC Neurol 2014 Feb; 14: 31, http://dx.doi.org/10.1186/1471-2377-14-31.

21. Trebst C., Jarius S., Berthele A., Paul F., Schippling S., Wildemann B., Borisow N., Kleiter I., Aktas O., KQmpfel T.; Neuromyelitis Optica Study Group (NEMOS). Update on the diagnosis and treatment of neuromyelitis optica: recommendations of the Neuromyelitis Optica Study Group (NEMOS). J Neurol 2014 Jan; 261(1): 1-16, http://dx.doi. org/10.1007/s00415-013-7169-7.

22. Pula J.H., Towle V.L., Staszak V.M., Cao D., Bernard J.T., Gomez C.M. Retinal nerve fibre layer and macular thinning in spinocerebellar ataxia and cerebellar multisystem atrophy. Neuroophthalmology 2011 Jun; 35(3): 108-114, http://dx.doi.org/10.3109/01658107.2011.580898.

23. Wiethoff S., Zhour A., Schols L., Fischer M.D. Retinal nerve fibre layer loss in hereditary spastic paraplegias is restricted to complex phenotypes. BMC Neurol 2012 Nov; 12: 143, http://dx.doi.org/10.1186/1471-2377-12-143.

24. Wang D., Li Y., Wang C., Xu L., You Q.S., Wang Y.X., Zhao L., Wei W.B., Zhao X., Jonas J.B. Localized retinal nerve fiber layer defects and stroke. Stroke 2014 Jun; 45(6): 16511566, http://dx.doi.org/10.1161/STROKEAHA.113.004629.

25. McNeill A., Roberti G., Lascaratos G., Hughes D.,

Mehta A., Garway-Heath D.F., Schapira A.H. Retinal thinning in Gaucher disease patients and carriers: results of a pilot study. Mol Genet Metab 2013 Jun; 109(2): 221-223, http:// dx.doi.org/10.1016/j.ymgme.2013.04.001.

26. Garcia-Martin E., Satue M., Otin S., Fuertes I., Alarcia R., Larrosa J.M., Polo V., Pablo L.E. Retina measurements for diagnosis of Parkinson disease. Retina 2014 May; 34(5): 971980, http://dx.doi.org/10.1097/IAE.0000000000000028.

27. Garcia-Martin E.S., Rojas B., Ramirez A.I., de Hoz R., Salazar J.J., Yubero R., Gil P., Trivino A., Ramirez J.M. Macular thickness as a potential biomarker of mild Alzheimer's disease. Ophthalmology 2014 May; 121(5): 1149-1151, http://dx.doi. org/10.1016/j.ophtha.2013.12.023.

28. Garcia-Martin E., Pablo L.E., Gazulla J., Vela A., Larrosa J.M., Polo V., Marques M.L., Alfaro J. Retinal segmentation as noninvasive technique to demonstrate hyperplasia in ataxia of Charlevoix-Saguenay. Invest Ophthalmol Vis Sci 2013 Oct; 54(10): 7137-7142. http://dx.doi. org/10.1167/iovs.13-12726.

29. Kopishinskaya S., Svetozarskiy S., Antonova V., Gustov A. The first data on retinal optical coherence tomography parameters in Huntington's disease. Eur J Neurol 2014 May; 21(Suppl 1): 36.

30. Yavas G.F., Yilmaz O., KQsbeci T., OztQrk F. The effect of levodopa and dopamine agonists on optic nerve head in Parkinson disease. Eur J Ophthalmol 2007 Sep-Oct; 17(5): 812-816.

31. Zhang N., Hoffmeyer G.C., Young E.S., Burns R.E., Winter K.P., Stinnett S.S., Toth C.A., Jaffe G.J. Optical coherence tomography reader agreement in neovascular age-related macular degeneration. Am J Ophthalmol 2007 Jul; 144(1): 37-44, http://dx.doi.org/10.1016/j.ajo.2007.03.056.

32. Syc S.B., Warner C.V., Hiremath G.S., Farrell S.K., Ratchford J.N., Conger A., Frohman T., Cutter G., Balcer L.J., Frohman E.M., Calabresi P.A. Reproducibility of high-resolution optical coherence tomography in multiple sclerosis. Mult Scler 2010 Jul; 16(7): 829-839, http://dx.doi.org/10.1177/13524585 10371640.

33. Tewarie P., Balk L., Costello F., Green A., Martin R., Schippling S., Petzold A. The OSCAR-IB consensus criteria for retinal OCT quality assessment. PLoS One 2012; 7(4): e34823, http://dx.doi.org/10.1371/journal.pone.0034823.

iНе можете найти то, что вам нужно? Попробуйте сервис подбора литературы.

34. Schippling S., Balk L., Costello F., Albrecht P., Balcer L., Calabresi P., Frederiksen J., Frohman E., Green A., Klistorner A., Outteryck O., Paul F., Plant G., Traber G., Vermersch P., Villoslada P., Wolf S., Petzold A. Quality control for retinal OCT in multiple sclerosis: validation of the OSCAR-IB criteria. Mult Scler 2014, [Epub ahead of print], http://dx.doi. org/10.1177/1352458514538110.

35. Alizadeh Y., Panjtanpanah M.R., Mohammadi M.J., Behboudi H., Kazemnezhad L.E. Reproducibility of optical coherence tomography retinal nerve fiber layer thickness measurements before and after pupil dilation. J Ophthalmic Vis Res 2014 Jan; 9(1): 38-43.

36. Hroudova J., Singh N., Fisar Z. Mitochondrial dysfunctions in neurodegenerative diseases: relevance to Alzheimer's disease. Biomed Res Int 2014; 2014: 175062, http://dx.doi.org/10.1155/2014/175062.

37. Kurokawa M., Kornbluth S. Caspases and kinases in a death grip. Cell 2009 Sep; 138(5): 838-854, http://dx.doi. org/10.1016/j.cell.2009.08.021.

38. Tudor D., Kajic V., Rey S., Erchova I., Povazay B., Hofer B., Powell K.A., Marshall D., Rosin P.L., Drexler W.,

уттжтжтжтжттттжтжтжтжтттт^ттттжтттт^тттт^тититттттиттг/'

120 STM J 2015 — vol. 7, No.1 S.N. Svetozarskiy, S.V. Kopishinskaya

REVIEWS OF UTILIZATION

Morgan J.E. Non-invasive detection of early retinal neuronal degeneration by ultrahigh resolution optical coherence tomography. PLoS One 2014 Apr; 9(4): e93916, http://dx.doi. org/10.1371/journal.pone.0093916.

39. Mead B., Logan A., Berry M., Leadbeater W., Scheven B.A. Intravitreally transplanted dental pulp stem cells promote neuroprotection and axon regeneration of retinal ganglion cells after optic nerve injury. Invest Ophthalmol Vis Sci 2013 Nov; 54(12): 7544-7556, http://dx.doi.org/10.1167/ iovs.13-13045.

40. Berger A., Cavallero S., Dominguez E., Barbe P.,

Simonutti M., Sahel J.A., Sennlaub F., Raoul W., Paques M., Bemelmans A.P. Spectral-domain optical coherence

tomography of the rodent eye: highlighting layers of the outer retina using signal averaging and comparison with histology. PLoS One 2014 May; 9(5): e96494, http://dx.doi.org/10.1371/ journal.pone.0096494.

41. Branchini L., Regatieri C.V., Flores-Moreno I., Baumann B., Fujimoto J.G., Duker J.S. Reproducibility of choroidal thickness measurements across three spectral domain optical coherence tomography systems. Ophthalmology 2012 Jan; 119(1): 119-123, http://dx.doi.org/10.1016/ j.ophtha.2011.07.002.

42. Serrano-Pozo A., Frosch M.P., Masliah E., Hyman B.T. Neuropathological alterations in Alzheimer disease. Cold Spring Harb Perspect Med 2011 Sep; 1(1): a006189, http:// dx.doi.org/10.1101/cshperspect.a006189.

43. Nelson P.T., Alafuzoff I., Bigio E.H., Bouras C., Braak H., Cairns N.J., et al. Correlation of Alzheimer disease neuropathologic changes with cognitive status: a review of the literature. J Neuropathol Exp Neurol 2012 May; 71(5): 362381, http://dx.doi.org/10.1097/NEN.0b013e31825018f7.

44. Hinton D.R., Sadun A.A., Blanks J.C., Miller C.A. Optic-nerve degeneration in Alzheimer's disease. N Engl J Med 1986 Aug; 315(8): 485-487, http://dx.doi.org/10.1056/ NEJM198608213150804.

45. Ning A., Cui J., To E., Ashe K.H., Matsubara J. Amyloid-P deposits lead to retinal degeneration in a mouse model of Alzheimer disease. Invest Ophthalmol Vis Sci 2008 Nov; 49(11): 5136-5143, http://dx.doi.org/10.1167/iovs.08-1849.

46. Perez S.E., Lumayag S., Kovacs B., Mufson E.J., Xu S. P-amyloid deposition and functional impairment in the retina of the APPswe/PS1AE9 transgenic mouse model of Alzheimer's disease. Invest Ophthalmol Vis Sci 2009 Feb; 50(2): 793-800, http://dx.doi.org/10.1167/iovs.08-2384.

47. Berisha F., Feke G.T., Trempe C.L., McMeel J.W., Schepens C.L. Retinal abnormalities in early Alzheimer's disease. Invest Ophthalmol Vis Sci 2007 May; 48(5): 22852289, http://dx.doi.org/10.1167/iovs.06-1029.

48. Chiu K., Chan T.F., Wu A., Leung I.Y., So K.F., Chang R.C. Neurodegeneration of the retina in mouse models of Alzheimer's disease: what can we learn from the retina? Age (Dordr) 2012 Jun; 34(3): 633-649, http://dx.doi.org/10.1007/ s11357-011-9260-2.

49. Frost S., Kanagasingam Y., Sohrabi H., Vignarajan J., Bourgeat P., Salvado O., Villemagne V., Rowe C.C., Macaulay S.L., Szoeke C., Ellis K.A., Ames D., Masters C.L., Rainey-Smith S., Martins R.N.; AIBL Research Group. Retinal vascular biomarkers for early detection and monitoring of Alzheimer's disease. Transl Psychiatry 2013 Feb; 3(2): e233, http://dx.doi.org/10.1038/tp.2012.150.

50. Koronyo-Hamaoui M., Koronyo Y., Ljubimov A.V., Miller C.A., Ko M.K., Black K.L., Schwartz M., Farkas D.L.

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 Jan; 54(Suppl 1): S204-S217, http://dx.doi.org/10.1016/j.neuroimage.2010.06.0 20.

51. Tsai Y., Lu B., Ljubimov A.V., Girman S., Ross-Cisneros F.N., Sadun A.A., Svendsen C.N., Cohen R.M., Wang S. Ocular changes in TgF344-AD rat model of Alzheimer's disease. Invest Ophthalmol Vis Sci 2014 Jan; 55(1): 523-534, http://dx.doi.org/10.1167/iovs.13-12888.

52. Simao L.M. The contribution of optical coherence

tomography in neurodegenerative diseases. Curr Opin Ophthalmol 2013 Nov; 24(6): 521-527, http://dx.doi.

org/10.1097/ICU.0000000000000000.

53. Jindahra P., Plant G.T. Retinal nerve fibre layer thinning in Alzheimer Disease. Chapter 14. In: The clinical spectrum of Alzheimers disease — the charge toward comprehensive diagnostic and therapeutic strategies. Edited by De La Monte S. InTech; 2011; p. 279-294, http://dx.doi.org/10.5772/16891.

54. Blanks J.C., Schmidt S.Y., Torigoe Y., Porrello K.V, Hinton D.R., Blanks R.H. Retinal pathology in Alzheimer's disease. II. Regional neuron loss and glial changes in GCL. Neurobiol Aging 1996 May-Jun; 17(3): 385-395, http://dx.doi. org/10.1016/0197-4580(96)00009-7.

55. Loffler K.U., Edward D.P., Tso M.O. Immunoreactivity against tau, amyloid precursor protein, and beta-amyloid in the human retina. Invest Ophthalmol Vis Sci 1995 Jan; 36(1): 24-31.

56. Ho W.L., Leung Y., Tsang A.W., So K.F., Chiu K., Chang R.C. Review: tauopathy in the retina and optic nerve: does it shadow pathological changes in the brain? Mol Vis 2012; 18: 2700-2710.

57. Jindahra P., Petrie A., Plant G.T. Retrograde transsynaptic retinal ganglion cell loss identified by optical coherence tomography. Brain 2009 Mar; 132(3): 628-634, http://dx.doi. org/10.1093/brain/awp001.

58. Bridge H., Jindahra P., Barbur J., Plant G.T. Imaging reveals optic tract degeneration in hemianopia. Invest Ophthalmol Vis Sci 2011 Jan 21; 52(1): 382-388, http://dx.doi. org/10.1167/iovs.10-5708.

59. Jindahra P., Petrie A., Plant G.T. The time course of retrograde trans-synaptic degeneration following occipital lobe damage in humans. Brain 2012 Feb; 135(2): 534-541, http:// dx.doi.org/10.1093/brain/awr324.

60. Millington R.S., Yasuda C.L., Jindahra P., Jenkinson M., Barbur J.L., Kennard C., Cendes F., Plant G.T., Bridge H. Quantifying the pattern of optic tract degeneration in human hemianopia. J Neurol Neurosurg Psychiatry 2014 Apr; 85(4): 379-386, http://dx.doi.org/10.1136/jnnp-2013-306577.

61. Tamura H., Kawakami H., Kanamoto T., Kato T., Yokoyama T., Sasaki K., Izumi Y., Matsumoto M., Mishima H.K. High frequency of open-angle glaucoma in Japanese patients with Alzheimer's disease. J Neurol Sci 2006 Jul; 246(1-2): 7983, http://dx.doi.org/10.1016/j.jns.2006.02.009.

62. Bayer A.U., Ferrari F., Erb C. High occurrence rate of glaucoma among patients with Alzheimer's disease. Eur Neurol 2002; 47(3): 165-168, http://dx.doi.org/10.1159/000047976.

63. Bayer A.U., Ferrari F. Severe progression of glaucomatous optic neuropathy in patients with Alzheimer's disease. Eye (Lond) 2002 Mar; 16(2): 209-212.

64. Wostyn P., De Groot V., Van Dam D., Audenaert K., De Deyn P.P. The role of low intracranial pressure in the development of glaucoma in patients with Alzheimer's disease.

Retinal Optical Coherence Tomography in Neurodegenerative Diseases STM J 2015 — vol. 7, No.1 121

REVIEWS OF UTILIZATION

Prog Retin Eye Res 2014 Mar; 39: 107-108, http://dx.doi. org/10.1016/j.preteyeres.2013.12.002.

65. Ott B.R., Cohen R.A., Gongvatana A., Okonkwo O.C., Johanson C.E., Stopa E.G., Donahue J.E., Silverberg G.D.; Alzheimer's Disease Neuroimaging Initiative. Brain ventricular volume and cerebrospinal fluid biomarkers of Alzheimer's disease. J Alzheimers Dis 2010; 20(2): 647-657, http://dx.doi. org/10.3233/JAD-2010-1406.

66. Serot J.M., Bene M.C., Faure G.C. Choroid plexus, aging of the brain, and Alzheimer's disease. Front Biosci 2003 May; 8: s515-s521.

67. Johanson C.E., Duncan J.A. 3rd, Klinge P.M., Brinker T., Stopa E.G., Silverberg G.D. Multiplicity of cerebrospinal fluid functions: new challenges in health and disease. Cerebrospinal Fluid Res 2008 May 14; 5: 10, http://dx.doi.org/10.1186/1743-8454-5-10.

68. 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 Jun; 420(2): 97-99, http://dx.doi.org/10.1016/ j.neulet.2007.02.090.

69. Marziani E., Pomati S., Ramolfo P., Cigada M., Giani A., Mariani C., Staurenghi G. 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 Sep; 54(9): 5953-5958, http://dx.doi. org/doi:10.1167/iovs.13-12046.

70. Kirbas S., Turkyilmaz K., Anlar O., Tufekci A, Durmus M. Retinal nerve fiber layer thickness in patients with Alzheimer disease. J Neuroophthalmol 2013 Mar; 33(1): 58-61, http:// dx.doi.org/10.1097/WNO.0b013e318267fd5f.

71. Shi Z., Wu Y., Wang M., Cao J., Feng W., Cheng Y., Li C., Shen Y. Greater attenuation of retinal nerve fiber layer thickness in Alzheimer's disease patients. J Alzheimers Dis 2014; 40(2): 277-283, http://dx.doi.org/10.3233/JAD-131898.

72. Kromer R., Serbecic N., Hausner L., Froelich L., Aboul-Enein F., Beutelspacher S.C. Detection of retinal nerve fiber layer defects in Alzheimer's disease using SD-OCT. Front Psychiatry 2014; 5: 22, http://dx.doi.org/10.3389/ fpsyt.2014.00022.

73. He X.-F., Liu Y.-T., Peng C., Zhang F., Zhuang S., Zhang J.S. Optical coherence tomography assessed retinal nerve fiber layer thickness in patients with Alzheimer's disease: a meta-analysis. Int J Ophthalmol 2012; 5(3): 401-405, http:// dx.doi.org/10.3980/j.issn.2222-3959.2012.03.30.

74. Iseri P.K., Altina§ O., Tokay T., YQksel N. Relationship between cognitive impairment and retinal morphological and visual functional abnormalities in Alzheimer disease. J Neuroophthalmol 2006 Mar; 26(1): 18-24, http://dx.doi. org/10.1097/01.wno.0000204645.56873.26.

75. Bayhan H.A., Bayhan S.A., Celikbilek A., Tanik N., GQrdal C. Evaluation of the chorioretinal thickness changes in Alzheimer's disease using spectral-domain optical coherence tomography. Clin Experiment Ophthalmol 2014 Jul, [Epub ahead of print], http://dx.doi.org/10.1111/ceo.12386.

76. Parisi V., Restuccia R., Fattapposta F., Mina C., Bucci M.G., Pierelli F. Morphological and functional retinal impairment in Alzheimer's disease patients. Clin Neurophysiol 2001 Oct; 112(10): 1860-1867, http://dx.doi.org/10.1016/ S1388-2457(01)00620-4.

77. Kromer R., Serbecic N., Hausner L., Froelich L., Beutelspacher S.C. Comparison of visual evoked potentials and retinal nerve fiber layer thickness in Alzheimer's disease.

Front Neurol 2013; 4: 203, http://dx.doi.org/10.3389/

fneur.2013.00203.

78. Gharbiya M., Trebbastoni A., Parisi F., Manganiello S., Cruciani F., D'Antonio F., De Vico U., Imbriano L., Campanelli A., De Lena C. 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(4): 907-917, http://dx.doi.org/10.3233/JAD-132039.

79. Khoo T.K., Yarnall A.J., Duncan G.W., Coleman S., O'Brien J.T., Brooks D.J., Barker R.A., Burn D.J. The spectrum of nonmotor symptoms in early Parkinson disease. Neurology

2013 Jan 15; 80(3): 276-281, http://dx.doi.org/10.1212/ WNL.0b013e31827deb74.

80. Archibald N.K., Clarke M.P., Mosimann U.P., Burn D.J. The retina in Parkinson's disease. Brain 2009; 132: 11281145, http://dx.doi.org/10.1093/brain/awp068.

81. Hajee M.E., March W.F., Lazzaro D.R., Wolintz A.H.,

Shrier E.M., Glazman S., Bodis-Wollner I.G. Inner retinal layer thinning in Parkinson disease. Arch Ophthalmol 2009 Jun; 127(6): 737-741, http://dx.doi.org/10.1001/

archophthalmol.2009.106.

82. Bodis-Wollner I. Foveal vision is impaired in Parkinson's disease. Parkinsonism Relat Disord 2013 Jan; 19(1): 1-14, http://dx.doi.org/10.1016Zj.parkreldis.2012.07.012.

83. He Q., Xu H.P., Wang P., Tian N. Dopamine D1 receptors regulate the light dependent development of retinal synaptic responses. PLoS One 2013 Nov 19; 8(11): e79625, http://dx.doi.org/10.1371/journal.pone.0079625.

84. Hwang C.K., Chaurasia S.S., Jackson C.R., Chan G.C., Storm D.R., luvone P.M. Circadian rhythm of contrast sensitivity is regulated by a dopamine-neuronal PAS-domain protein 2-adenylyl cyclase 1 signaling pathway in retinal ganglion cells. J Neurosci 2013 Sep; 33(38): 14989-14997, http://dx.doi. org/10.1523/JNEUROSCI.2039-13.2013.

85. Aung M.H., Park H.N., Han M.K., Obertone T.S., Abey J., Aseem F., Thule P.M., luvone P.M., Pardue M.T. Dopamine deficiency contributes to early visual dysfunction in a rodent model of type 1 diabetes. J Neurosci 2014 Jan; 34(3): 726-736, http://dx.doi.org/10.1523/JNEUROSCI.3483-13.2014.

86. Archibald N.K., Clarke M.P., Mosimann U.P., Burn D.J. Visual symptoms in Parkinson's disease and Parkinson's disease dementia. Mov Disord 2011 Nov; 26(13): 2387-2395, http://dx.doi.org/10.1002/mds.23891.

87. Archibald N.K., Hutton S.B., Clarke M.P., Mosimann U.P., Burn D.J. Visual exploration in Parkinson's disease and Parkinson's disease dementia. Brain 2013 Mar; 136(Pt 3): 739-750, http://dx.doi.org/10.1093/brain/awt005.

88. Urwyler P., Nef T., Killen A., Collerton D., Thomas A., Burn D., McKeith I., Mosimann U.P. Visual complaints and visual hallucinations in Parkinson's disease. Parkinsonism Relat Disord 2014 Mar; 20(3): 318-222, http://dx.doi.org/10.1016/j.p arkreldis.2013.12.009.

89. Inzelberg R., Ramirez J.A., Nisipeanu P., Ophir A. Retinal nerve fiber layer thinning in Parkinson disease. Vision Res 2004 Nov; 44(24): 2793-2797, http://dx.doi.org/10.1016/ j.visres.2004.06.009.

90. Yu J.G., Feng Y.F., Xiang Y., Huang J.H., Savini G., Parisi V., Yang W.J., Fu X.A. Retinal nerve fiber layer thickness changes in Parkinson disease: a meta-analysis. PLoS One

2014 Jan; 9(1): e85718, http://dx.doi.org/10.1371/journal. pone.0085718.

91. Tsironi E.E., Dastiridou A., Katsanos A.,

уттжтжтжтжттттжтжтжтжтттт^ттттжтттт^тттт^тититттттиттг/'

122 STM J 2015 — vol. 7, No.1 S.N. Svetozarskiy, S.V. Kopishinskaya

REVIEWS OF UTILIZATION

Dardiotis E., Veliki S., Patramani G., Zacharaki F., Ralli S., Hadjigeorgiou G.M. Perimetric and retinal nerve fiber layer findings in patients with Parkinson's disease. BMC Ophthalmol 2012 Oct; 12: 54, http://dx.doi.org/10.1186/1471-2415-12-54.

92. Satue M., Seral M., Otin S., Alarcia R., Herrero R., Bambo M.P., Fuertes M.I., Pablo L.E., Garcia-Martin E. Retinal thinning and correlation with functional disability in patients with Parkinson's disease. Br J Ophthalmol 2014 Mar; 98(3): 350355, http://dx.doi.org/10.1136/bjophthalmol-2013-304152.

93. Garcia-Martin E., Larrosa J.M., Polo V., Satue M., Marques M.L., Alarcia R., Seral M., Fuertes I., Otin S., Pablo L.E. Distribution of retinal layer atrophy in patients with Parkinson disease and association with disease severity and duration. Am J Ophthalmol 2014 Feb; 157(2): 470-478.e2, http://dx.doi.org/10.1016/j.ajo.2013.09.028.

94. Schneider M., Moller H.P., Lauda F., Tumani H., Ludolph A.C., Kassubek J., Pinkhardt E.H. Retinal single-layer analysis in Parkinsonian syndromes: an optical coherence tomography study. J Neural Transm 2014 Jan; 121(1): 41-47, http://dx.doi.org/10.1007/s00702-013-1072-3.

95. Shrier E.M., Adam C.R., Spund B., Glazman S., Bodis-Wollner I. Interocular asymmetry of foveal thickness in Parkinson disease. J Ophthalmol 2012; 2012: 728457, http://

97. Adam C.R., Shrier E., Ding Y., Glazman S., Bodis-Wollner I. Correlation of inner retinal thickness evaluated by spectral-domain optical coherence tomography and contrast sensitivity in Parkinson disease. J Neuroophthalmol

2013 Jun; 33(2): 137-142, http://dx.doi.org/10.1097/

WNO.0b013e31828c4e1a.

98. Bambo M.P., Garcia-Martin E., Otin S., Sancho E.,

Fuertes I., Herrero R., Satue M., Pablo L. Influence of cataract surgery on repeatability and measurements of spectral domain optical coherence tomography. Br J Ophthalmol 2014 Jan; 98(1): 52-58, http://dx.doi.org/10.1136/bjophthalmol-2013-

303752.

99. Smetankin I.G., Agarkova D.I. Confocal microscopy and optical coherent tomography for evaluation of the anatomical and functional state of corneal wound (in vivo) after cataract phacoemulcification. Sovremennye tehnologii v medicine 2012; 3: 89-92.

100. Hood D.C., Raza A.S. On improving the use of OCT imaging for detecting glaucomatous damage. Br J Ophthalmol

2014 Jul; 98(Suppl 2): ii1-ii9, http://dx.doi.org/10.1136/ bjophthalmol-2014-305156.

101. Rao H.L., Addepalli U.K., Yadav R.K., Senthil S., Choudhari N.S., Garudadri C.S. Effect of scan quality on

i Надоели баннеры? Вы всегда можете отключить рекламу.