Skip to main content
Advertisement
Browse Subject Areas
?

Click through the PLOS taxonomy to find articles in your field.

For more information about PLOS Subject Areas, click here.

  • Loading metrics

Increased risk of Alzheimer’s disease among patients with age-related macular degeneration: A nationwide population-based study

  • Li-Yen Wen ,

    Contributed equally to this work with: Li-Yen Wen, Lei Wan

    Roles Data curation, Formal analysis, Investigation, Methodology, Writing – original draft

    Affiliation School of Chinese Medicine, China Medical University, Taichung, Taiwan

  • Lei Wan ,

    Contributed equally to this work with: Li-Yen Wen, Lei Wan

    Roles Conceptualization, Validation, Visualization, Writing – review & editing

    Affiliations School of Chinese Medicine, China Medical University, Taichung, Taiwan, Department of Medical Laboratory Science and Biotechnology, Asia University, Taichung, Taiwan, Department of Obstetrics and Gynecology, China Medical University Hospital, Taichung, Taiwan

  • Jung-Nien Lai,

    Roles Funding acquisition, Supervision

    Affiliation School of Chinese Medicine, China Medical University, Taichung, Taiwan

  • Chih Sheng Chen,

    Roles Funding acquisition, Project administration, Resources

    Affiliation Division of Chinese Medicine, Asia University Hospital, Taichung, Taiwan

  • Jamie Jiin-Yi Chen,

    Roles Data curation, Formal analysis, Software

    Affiliations School of Chinese Medicine, China Medical University, Taichung, Taiwan, Department of Ophthalmology and Department of Molecular Genetics, China Medical University Hospital, Taichung, Taiwan

  • Ming-Yen Wu,

    Roles Conceptualization, Software, Validation, Visualization

    Affiliations School of Chinese Medicine, China Medical University, Taichung, Taiwan, Department of Ophthalmology and Department of Molecular Genetics, China Medical University Hospital, Taichung, Taiwan

  • Kai-Chieh Hu,

    Roles Writing – review & editing

    Affiliation Management office for Health Data, China Medical University Hospital, Taichung, Taiwan

  • Lu-Ting Chiu,

    Roles Data curation, Formal analysis, Methodology, Software

    Affiliation Management office for Health Data, China Medical University Hospital, Taichung, Taiwan

  • Peng-Tai Tien ,

    Roles Project administration, Resources, Supervision

    miketien913@gmail.com (PTT); irisluu2396@gmail.com (HJL)

    Affiliations Department of Ophthalmology and Department of Molecular Genetics, China Medical University Hospital, Taichung, Taiwan, Graduate Institute of Clinical Medical Science, College of Medicine, China Medical University, Taichung, Taiwan

  • Hui-Ju Lin

    Roles Conceptualization, Funding acquisition, Project administration, Resources, Supervision

    miketien913@gmail.com (PTT); irisluu2396@gmail.com (HJL)

    Affiliations School of Chinese Medicine, China Medical University, Taichung, Taiwan, Department of Ophthalmology and Department of Molecular Genetics, China Medical University Hospital, Taichung, Taiwan

Abstract

Objective

This study aimed to investigate the risk of Alzheimer’s disease among patients with age-related macular degeneration and its association with confounding comorbidities.

Method

This was a population-based, retrospective cohort study. By accessing data from the National Health Insurance Research Database of Taiwan, we identified 10,578 patients aged 50–100 years who were newly diagnosed with age-related macular degeneration between 2000 and 2012 and 10,578 non- age-related macular degeneration individuals. The comorbidities assessed were osteoporosis, diabetes, cirrhosis, cerebrovascular disease, chronic kidney disease, hypertension, hyperlipidemia, coronary artery disease, and chronic obstructive pulmonary disease.

Results

Patients with age-related macular degeneration had a 1.23-fold increased risk of their condition advancing to Alzheimer’s disease (aHR = 1.23, 95% CI = 1.04–1.46). The younger patients were diagnosed with age-related macular degeneration, the more likely patients got Alzheimer’s disease (50–64 age group: aHR = 1.97, 95% CI = 1.04–3.73; 65–79 age group: aHR = 1.27, 95% CI = 1.02–1.58; 80–100 age group: aHR = 1.06, 95% CI = 0.78–1.45). In addition, there were significantly higher risks of Alzheimer’s disease for patients with cirrhosis (aHR = 1.50, 95% CI = 1.09–2.06) in the age-related macular degeneration cohort than in the non-age-related macular degeneration cohort.

Conclusion

Patients with age-related macular degeneration may exhibit a higher risk of Alzheimer’s disease than people without age-related macular degeneration.

Introduction

Alzheimer’s disease (AD) is a neurodegenerative disorder characterized by progressive and irreversible cognitive impairment. AD is the most common cause of dementia in the elderly, with an estimated 44 million people having AD worldwide, and the prevalence is predicted to triple by 2050 [1]. The characteristic neuropathological change in AD is the presence of amyloid-beta (Aβ) plaques and neurofibrillary tangles in brain tissues [2, 3].

The exact etiology and pathogenesis of AD remain unclear. Over the past two decades, the amyloid cascade hypothesis has been one of the most acceptable theories [4, 5]. Aβ stems from a transmembrane protein of neuronal cells termed amyloid precursor protein (APP). Aβ is generated when APP is sequentially cleaved by β-secretase and γ-secretase [6, 7]. The production and clearance imbalance of Aβ results in abnormal Aβ deposition in the neuropil, which is considered the main cause of synaptic loss and neuronal cell death in AD [8, 9].

Age-related macular degeneration (AMD) is another neurodegenerative disease that mainly affects the aging population. The disease results in progressive central vision loss and is the fourth leading cause of blindness worldwide [10, 11]. The hallmark pathology of AMD is ocular drusen and extracellular deposits located between the retinal pigment epithelium (RPE) and Bruch’s membrane [12]. Similar to senile plaques in AD, Aβ is also an important constituent in drusen [1315]. Previous study has demonstrated that the Aβ in drusen correlates with the location of photoreceptors and RPE cell degeneration [16]. Moreover, oxidative stress, neuroinflammation, and complement activation have been implicated in both AD and AMD [1719]. AD and AMD share risk factors such as cigarette smoking and diabetes [2023]. In addition, genetic studies have also noted that the apolipoprotein E (APOE) epsilon‑4 and cysteine proteinase inhibitor cystatin C gene (CST3) increase the risk for both disorders [24, 25].

Because of the similarity in pathologic features between AMD and AD, there has been an increasing interest in investigating the potential link between the two diseases in recent years. However, most studies on the association between AMD and AD have shown paradoxical results. Several publications have documented that AMD was associated with cognitive impairment and dementia [2629]. On the contrary, an epidemiologic study reported that there was no significant increase of the risk of dementia and AD following AMD [30]. Most past studies contained small sample size or only adjusted for a few factors. Therefore, we used a national population-based dataset from Taiwan National Health Insurance to investigate the association between AMD and AD.

Methods

Data resource

The requirement for informed consent was waived by both the National Health Insurance (NHI) Administration and China Medical University Hospital Research Ethics Committee.

Taiwan adopted a single-payer NHI program in March 1995. It is mandatory for Taiwanese citizens to enroll in the program; the coverage rate of NHI is over 99 percent. We used the Longitudinal Health Insurance Database 2010 (LHID 2010), which contains the claims data of 1,000,000 beneficiaries randomly sampled from the National Health Insurance Research Database (NHIRD). No significant differences in age and sex were found between the LHID 2010 and the enrollees in the mother NHIRD. Patient demographic characteristics included encrypted identification numbers, sex, dates of birth and death, diagnostic data, and procedures. The diagnostic data were coded according to the International Classification of Diseases, Ninth Revision, Clinical Modification (ICD-9-CM). The accuracy of the diagnosis codes of common conditions in NHIRD has been validated [3133]. This study was approved by China Medical University Hospital Research Ethics Committee.

Sample participant, outcome, and comorbidity

We included 10,578 individuals aged at 50–100 years who were newly diagnosed with AMD (ICD-9-CM code 362.50, 362.51, and 362.52) by certified ophthalmologists between 2000 and 2010 in the AMD group. The date of the first outpatient visits or inpatient visit for AMD was defined as index date. For comparison, each AMD patient was frequency matched with an individual without AMD based on gender, age (in 5-year bands), and index-year. The exclusion criteria was patients with a pre-existing diagnosis of AD (ICD-9-CM 331) in both AMD cohort and control cohort. All study subjects were followed up until the appearance of AD, death, withdrawal from the insurance program or the end of study period (December 31, 2013).

All included patients were followed from the index date until the occurrence of the endpoint, withdrawal from the NHI program, or the end of 2013. The potential comorbidities included osteoporosis (ICD-9-CM code 733), diabetes (ICD-9-CM code 250 and A181), cirrhosis (ICD-9-CM code 571 and A347), cerebrovascular disease (ICD-9-CM code 342.34, and 430–438), chronic kidney disease (ICD-9-CM code 403.11, 403.91, 404.12, 404.92, 585, 586, v42.0, v45.1, v56.0, and v56.8), hypertension (ICD-9-CM code 401–405, A260 and A269), hyperlipidemia (ICD-9-CM code 272), coronary artery disease (ICD-9-CM code 410–414), and chronic obstructive pulmonary disease (ICD-9-CM code 491, 492, 493, and 496).

Statistical analysis

Descriptive statistics of demographics and comorbidities were summarized by counts, percentages, averages, and standard deviations (SDs) for the AMD and non-case cohort. Chi-square and Wilcoxon rank-sum tests were used to examine the differences in the characteristic distributions between the two cohorts. The cumulative incidence for AD was estimated using Kaplan-Meier methods and significance of the difference between the AMD cohort and the non-case cohort were tested using log-rank test. The incidence rate was calculated as the number of events divided by the person-years during the follow-up. The univariate Cox proportional hazards model was used to compute the hazard ratio (HR) and the 95% confidence interval (CI) for AD in the AMD cohort when compared to the non-case cohort. And the multivariate Cox proportional hazards model with covariates of gender, age, and comorbidities was used to compute the adjusted hazard ratio (aHR) and the 95% CI for AD in the AMD cohort when compared to the non-case cohort. Data analysis in this study was performed using SAS statistical software (Version 9.4 for Windows; SAS Institute Inc., Cary, NC, USA), with statistical significance set at a p value < 0.05.

Results

Table 1 reveals the demographic characteristics and the prevalence of comorbidities in both cohorts. The mean ages in the AMD and non-AMD group were 70.5 (±9.54, standard deviation [SD]) and 70.4 (±9.60, SD) years old, respectively, with the mean follow-up time of 5.68 (±3.81, SD) and 5.48 (±3.84, SD) years. The presence of osteoporosis, diabetes, cirrhosis, cerebrovascular disease, chronic kidney disease, hypertension, hyperlipidemia, coronary artery disease and chronic obstructive pulmonary disease was significantly higher in patients with AMD than controls.

thumbnail
Table 1. Demographic characteristics and presence of comorbidities in patients with and without age-related macular degeneration (AMD).

https://doi.org/10.1371/journal.pone.0250440.t001

Table 2 demonstrates that there was a higher risk of developing AD in the AMD cohort than the control cohort (aHR = 1.23, 95% CI = 1.04–1.46), with incidences of 5.36 and 3.92 per 1,000 person-years in the AMD and non-AMD groups, respectively. Compared to the 50-64-year subgroup, the subgroups with individuals 65–79 years old and more than 80 years old have a significant 3.67 (95% CI = 2.69–5.01) and 7.53 (95% CI = 5.38–10.5) risk for AD. Among all of comorbidities, patients with cerebrovascular disease and chronic obstructive pulmonary disease tended to have a higher risk for the development of AD (aHR = 1.44, 95% CI = 1.19–1.73 and aHR = 1.20, 95% CI = 1.01–1.44). Compared to the patients without any comorbidities, patients with at least one comorbidity also have a significant 1.23 (95% CI = 1.04–1.47) risk for AD.

thumbnail
Table 2. Cox proportional HRs for risk of Alzheimer’s disease in patients with and without AMD, in different gender, age groups, and in patients with preexisting comorbidities.

https://doi.org/10.1371/journal.pone.0250440.t002

Table 3 presents the risk for AD between patients with and without AMD stratified by gender, age, and comorbidities. Compared with the women without AMD, the AMD women had a significant higher risk of AD (aHR = 1.31, 95% CI = 1.04–1.66). And the younger patients were diagnosed with AMD, the more likely patients got AD (50–64 age group: aHR = 1.97, 95% CI = 1.04–3.73; 65–79 age group: aHR = 1.27, 95% CI = 1.02–1.58; 80–100 age group: aHR = 1.06, 95% CI = 0.78–1.45). Moreover, AMD patients with cirrhosis exhibited 1.50 (95% CI = 1.09–2.06) higher risk of AD compared with the non-AMD group. Comparing to without AMD cohort, the risk of AD for AMD in patients no matter with or without any comorbidities (aHR = 1.19, 95% CI = 1.00–1.42 and aHR = 2.19, 95% CI = 1.13–4.09) were also significantly higher.

thumbnail
Table 3. Incidence rates, hazard ratios, and confidence intervals of Alzheimer’s disease in patients with AMD stratified by gender, age and comorbidity.

https://doi.org/10.1371/journal.pone.0250440.t003

Table 4 presents the incidence rate of AD among different types of AMD. There were no statistically significant differences in the risk of AD between with and without macular degeneration (ICD-9-CM code: 362.50), nonexudative senile macular degeneration (ICD-9-CM code: 362.51), and exudative senile macular degeneration (ICD-9-CM code: 362.52) patients.

thumbnail
Table 4. Incidence rates, hazard ratios, and confidence intervals of Alzheimer’s disease among different type of AMD.

https://doi.org/10.1371/journal.pone.0250440.t004

The Kaplan–Meier plot of the cumulative incidence of Alzheimer’s disease in AMD and comparison cohort is depicted in Fig 1. After 11 years of follow-up, the AMD group had a significantly higher cumulative proportion of AD patients than did the non-AMD group (log-rank test (p = 0.003)). Fig 2 presents that there was a considerably different cumulative incidence of among each age group (log-rank p <0.0001).

thumbnail
Fig 1. Kaplan-Meier plot of the cumulative incidence of Alzheimer’s disease in patients with and without AMD.

https://doi.org/10.1371/journal.pone.0250440.g001

thumbnail
Fig 2. Kaplan-Meier plot of the cumulative incidence of Alzheimer’s disease in each age group.

https://doi.org/10.1371/journal.pone.0250440.g002

Discussion

This large-scale, nationwide, retrospective cohort study showed that patients with AMD exhibited a 1.23-fold higher risk of developing AD (aHR = 1.23, 95% CI = 1.04–1.46) than patients without AMD. Old age, cerebrovascular disease, and chronic obstructive pulmonary disease were also risk factors for AD. AMD patients who were female, younger age and with cirrhosis had a significant higher risk of developing AD.

Several publications have documented that AMD is associated with increased risk of AD or cognitive impairment. In 2006, the Blue Mountains Eye Study showed that people with late AMD were associated with lower Mini-Mental State Examination scores than people without AMD [34]. A cross-sectional study reported that AD frequency was significantly increased in the AMD group. The nonexudative AMD subtype was associated with worse cognitive function than the exudative AMD subtype [35]. Similarly, the population-based cohort study in Taiwan by Tsai suggested that patients with AMD had a greater risk of developing AD, although the nonexudative AMD subtype alone showed a conventional level of significance in stratified analysis [36]. However, neither the nonexudative nor the exudative AMD subtype was a significant risk factor for AD in our stratified analysis. The follow-up duration in our study is longer than that of Tsai (2000–2012 vs. 2001–2009), which may contribute to this finding. In a recent paper by Choi, AMD was associated with a higher risk of AD, even after considering lifestyle behaviors, including smoking, alcohol intake, and physical activity [37]. In contrast, the results obtained by Keenan et al. suggested that there is no positive association between AMD and AD. However, in England, most patients with AMD were admitted for receiving intravitreal anti-vascular endothelial growth factor therapy, which is the standard treatment for exudative AMD [30].

Aging is the primary risk factor for both AD and AMD [38]. Oxidative stress is considered to be an imbalance between free radicals and antioxidants, which is the fundamental mechanism contributing to the aging process [39]. Previous studies indicated that oxidative stress may be the common underlying pathological mechanism between AD and AMD [17]. Researchers have found that the levels of lipid peroxidation end products, oxidized proteins, and advanced glycation end products are increased in AD [40, 41]. Likewise, the elevation of oxidative stress was observed in the retina of patients with AMD [42, 43]. In 2011, a study showed that oxidative stress upregulated β-secretase and γ-secretase activities, resulting in increased Aβ production [44]. Common pathogenic pathways could explain the association between AMD and AD.

The strength of this study is the use of NHIRD, a nationwide population-based database, which covers over 99% of medical claims data of citizens in Taiwan, reducing the likelihood of selection and participation bias. However, some limitations should be noted. First, some confounding variables were unavailable in NHIRD. Smoking habit, alcohol consumption, body mass index, physical activity, and family history of AD could not be adjusted in the data analysis. Second, NHIRD lacks imaging findings, neuropsychological assessment results, and laboratory data. Third, AD, AMD, and additional comorbidities cases were identified using ICD-9-CM codes. Same as all claims databases, the accuracy of diagnosis codes is still a concern. However, several studies have validated the positive predictive value of the diagnostic ICD9-CM codes for common diseases, such as ischemic stroke, hypertension, diabetes and all cancers [3133]. Finally, AD and AMD are not usually diagnosed in the early stages, which could lead to misclassification bias in our study.

In conclusion, our study suggests that patients with AMD have a higher risk of AD than people without AMD. The exact pathological factors are still poorly understood, and further studies need to be conducted to explore the underlying mechanism between AD and AMD.

Acknowledgments

We would like to thank all the participants for their enthusiastic participation in this study.

References

  1. 1. Mendiola-Precoma J, Berumen LC, Padilla K, Garcia-Alcocer G. Therapies for Prevention and Treatment of Alzheimer’s Disease. Biomed Res Int. 2016;2016:2589276. Epub 2016/08/23. pmid:27547756; PubMed Central PMCID: PMC4980501.
  2. 2. De-Paula VJ, Radanovic M, Diniz BS, Forlenza OV. Alzheimer’s disease. Sub-cellular biochemistry. 2012;65:329–52. Epub 2012/12/12. pmid:23225010.
  3. 3. Octave J. Alzheimer disease: cellular and molecular aspects. Bulletin et memoires de l’Academie royale de medecine de Belgique. 2005;160(10–12):445–9; discussion 50. pmid:16768248
  4. 4. Barage SH, Sonawane KD. Amyloid cascade hypothesis: Pathogenesis and therapeutic strategies in Alzheimer’s disease. Neuropeptides. 2015;52:1–18. Epub 2015/07/08. pmid:26149638.
  5. 5. Korczyn AD. The amyloid cascade hypothesis. Alzheimer’s & dementia. 2008;4(3):176–8. pmid:18631966
  6. 6. Chow VW, Mattson MP, Wong PC, Gleichmann M. An overview of APP processing enzymes and products. Neuromolecular medicine. 2010;12(1):1–12. pmid:20232515
  7. 7. O’Brien RJ, Wong PC. Amyloid precursor protein processing and Alzheimer’s disease. Annual review of neuroscience. 2011;34:185–204. pmid:21456963
  8. 8. Shankar GM, Walsh DM. Alzheimer’s disease: synaptic dysfunction and Aβ. Molecular neurodegeneration. 2009;4(1):48. pmid:19930651
  9. 9. Mawuenyega KG, Sigurdson W, Ovod V, Munsell L, Kasten T, Morris JC, et al. Decreased clearance of CNS β-amyloid in Alzheimer’s disease. Science. 2010;330(6012):1774–. pmid:21148344
  10. 10. Jonas JB, Cheung CMG, Panda-Jonas S. Updates on the Epidemiology of Age-Related Macular Degeneration. Asia Pac J Ophthalmol (Phila). 2017;6(6):493–7. Epub 2017/09/15. pmid:28906084.
  11. 11. Lim LS, Mitchell P, Seddon JM, Holz FG, Wong TY. Age-related macular degeneration. The Lancet. 2012;379(9827):1728–38.
  12. 12. Al Gwairi O, Thach L, Zheng W, Osman N, Little PJ. Cellular and molecular pathology of age-related macular degeneration: potential role for proteoglycans. Journal of ophthalmology. 2016;2016. pmid:27563459
  13. 13. Yoshida T, Ohno-Matsui K, Ichinose S, Sato T, Iwata N, Saido TC, et al. The potential role of amyloid β in the pathogenesis of age-related macular degeneration. The Journal of clinical investigation. 2005;115(10):2793–800. pmid:16167083
  14. 14. Ashok A, Singh N, Chaudhary S, Bellamkonda V, Kritikos AE, Wise AS, et al. Retinal Degeneration and Alzheimer’s Disease: An Evolving Link. International Journal of Molecular Sciences. 2020;21(19):7290. pmid:33023198
  15. 15. Dentchev T, Milam AH, Lee VM-Y, Trojanowski JQ, Dunaief JL. Amyloid-β is found in drusen from some age-related macular degeneration retinas, but not in drusen from normal retinas. American journal of ophthalmology. 2003;136(4):787–.
  16. 16. Dentchev T, Milam AH, Lee VM, Trojanowski JQ, Dunaief JL. Amyloid-beta is found in drusen from some age-related macular degeneration retinas, but not in drusen from normal retinas. Molecular vision. 2003;9:184–90. Epub 2003/05/24. pmid:12764254.
  17. 17. Cerman E, Eraslan M, Cekic O. Age-related macular degeneration and Alzheimer disease. Turk J Med Sci. 2015;45(5):1004–9. Epub 2016/01/08. pmid:26738339.
  18. 18. Agostinho P, A Cunha R, Oliveira C. Neuroinflammation, oxidative stress and the pathogenesis of Alzheimer’s disease. Current pharmaceutical design. 2010;16(25):2766–78. pmid:20698820
  19. 19. Hollyfield JG, Bonilha VL, Rayborn ME, Yang X, Shadrach KG, Lu L, et al. Oxidative damage–induced inflammation initiates age-related macular degeneration. Nature medicine. 2008;14(2):194–8. pmid:18223656
  20. 20. Zhang J, Chen C, Hua S, Liao H, Wang M, Xiong Y, et al. An updated meta-analysis of cohort studies: diabetes and risk of Alzheimer’s disease. Diabetes research and clinical practice. 2017;124:41–7. pmid:28088029
  21. 21. Chen X, Rong SS, Xu Q, Tang FY, Liu Y, Gu H, et al. Diabetes mellitus and risk of age-related macular degeneration: a systematic review and meta-analysis. PloS one. 2014;9(9):e108196. pmid:25238063
  22. 22. Durazzo TC, Mattsson N, Weiner MW, Initiative AsDN. Smoking and increased Alzheimer’s disease risk: a review of potential mechanisms. Alzheimer’s & Dementia. 2014;10:S122–S45.
  23. 23. Neuner B, Komm A, Wellmann J, Dietzel M, Pauleikhoff D, Walter J, et al. Smoking history and the incidence of age-related macular degeneration—results from the Muenster Aging and Retina Study (MARS) cohort and systematic review and meta-analysis of observational longitudinal studies. Addictive behaviors. 2009;34(11):938–47. pmid:19539431
  24. 24. Williams MA, McKay GJ, Carson R, Craig D, Silvestri G, Passmore P. Age-Related Macular Degeneration–Associated Genes in Alzheimer Disease. The American Journal of Geriatric Psychiatry. 2015;23(12):1290–6. pmid:26419733
  25. 25. Butler JM, Sharif U, Ali M, McKibbin M, Thompson JP, Gale R, et al. A missense variant in CST3 exerts a recessive effect on susceptibility to age-related macular degeneration resembling its association with Alzheimer’s disease. Human genetics. 2015;134(7):705–15. pmid:25893795
  26. 26. Wong TY, Klein R, Nieto FJ, Moraes SA, Mosley TH, Couper DJ, et al. Is early age-related maculopathy related to cognitive function? The Atherosclerosis Risk in Communities Study. American journal of ophthalmology. 2002;134(6):828–35. pmid:12470750
  27. 27. Pham TQ, Kifley A, Mitchell P, Wang JJ. Relation of age-related macular degeneration and cognitive impairment in an older population. Gerontology. 2006;52(6):353–8. pmid:16902306
  28. 28. Klaver CC, Ott A, Hofman A, Assink JJ, Breteler MM, de Jong PT. Is age-related maculopathy associated with Alzheimer’s disease?: the Rotterdam Study. American journal of epidemiology. 1999;150(9):963–8. pmid:10547142
  29. 29. Rong SS, Lee BY, Kuk AK, Yu XT, Li SS, Li J, et al. Comorbidity of dementia and age-related macular degeneration calls for clinical awareness: a meta-analysis. British Journal of Ophthalmology. 2019;103(12):1777–83. pmid:31000510
  30. 30. Keenan TD, Goldacre R, Goldacre MJ. Associations between age-related macular degeneration, Alzheimer disease, and dementia: record linkage study of hospital admissions. JAMA Ophthalmol. 2014;132(1):63–8. Epub 2013/11/16. pmid:24232933.
  31. 31. Hsieh C-Y, Chen C-H, Li C-Y, Lai M-L. Validating the diagnosis of acute ischemic stroke in a National Health Insurance claims database. Journal of the Formosan Medical Association. 2015;114(3):254–9. pmid:24140108
  32. 32. Sung S-F, Hsieh C-Y, Lin H-J, Chen Y-W, Yang Y-HK, Li C-Y. Validation of algorithms to identify stroke risk factors in patients with acute ischemic stroke, transient ischemic attack, or intracerebral hemorrhage in an administrative claims database. International journal of cardiology. 2016;215:277–82. pmid:27128546
  33. 33. Kao WH, Hong JH, See LC, Yu HP, Hsu JT, Chou IJ, et al. Validity of cancer diagnosis in the National Health Insurance database compared with the linked National Cancer Registry in Taiwan. Pharmacoepidemiology and drug safety. 2018;27(10):1060–6. pmid:28815803
  34. 34. Hong T, Mitchell P, Burlutsky G, Liew G, Wang JJ. Visual Impairment, Hearing Loss and Cognitive Function in an Older Population: Longitudinal Findings from the Blue Mountains Eye Study. PloS one. 2016;11(1):e0147646. Epub 2016/01/26. pmid:26808979; PubMed Central PMCID: PMC4726694.
  35. 35. Seden D, Alime G, Kadir D, Serpil D, Levent T, Ozlem T. Is Alzheimer disease related to age-related macular degeneration? Turk J Med Sci. 2015;45(5):1115–21. Epub 2016/01/08. pmid:26738356.
  36. 36. Tsai DC, Chen SJ, Huang CC, Yuan MK, Leu HB. Age-Related Macular Degeneration and Risk of Degenerative Dementia among the Elderly in Taiwan: A Population-Based Cohort Study. Ophthalmology. 2015;122(11):2327–35 e2. Epub 2015/09/05. pmid:26337003.
  37. 37. Choi S, Jahng WJ, Park SM, Jee D. Association of Age-Related Macular Degeneration on Alzheimer or Parkinson Disease: A Retrospective Cohort Study. Am J Ophthalmol. 2020;210:41–7. Epub 2019/11/13. pmid:31712068.
  38. 38. Biscetti L, Luchetti E, Vergaro A, Menduno P, Cagini C, Parnetti L. Associations of Alzheimer’s disease with macular degeneration. Frontiers in bioscience (Elite edition). 2017;9:174–91. pmid:27814598
  39. 39. Pizzino G, Irrera N, Cucinotta M, Pallio G, Mannino F, Arcoraci V, et al. Oxidative stress: harms and benefits for human health. Oxidative medicine and cellular longevity. 2017;2017. pmid:28819546
  40. 40. Chen Z, Zhong C. Oxidative stress in Alzheimer’s disease. Neuroscience bulletin. 2014;30(2):271–81. pmid:24664866
  41. 41. Huang WJ, Zhang X, Chen WW. Role of oxidative stress in Alzheimer’s disease. Biomedical reports. 2016;4(5):519–22. pmid:27123241
  42. 42. Bellezza I. Oxidative stress in age-related macular degeneration: Nrf2 as therapeutic target. Frontiers in pharmacology. 2018;9:1280. pmid:30455645
  43. 43. Beatty S, Koh H-H, Phil M, Henson D, Boulton M. The role of oxidative stress in the pathogenesis of age-related macular degeneration. Survey of ophthalmology. 2000;45(2):115–34. pmid:11033038
  44. 44. Mouton-Liger F, Paquet C, Dumurgier J, Bouras C, Pradier L, Gray F, et al. Oxidative stress increases BACE1 protein levels through activation of the PKR-eIF2α pathway. Biochimica et Biophysica Acta (BBA)-Molecular Basis of Disease. 2012;1822(6):885–96. pmid:22306812