1
|
Dhapola R, Sharma P, Kumari S, Vellingiri B, Medhi B, HariKrishnaReddy D. Exploring Retinal Neurodegeneration in Alzheimer's Disease: A Molecular and Cellular Perspective. Neurotox Res 2025; 43:22. [PMID: 40216597 DOI: 10.1007/s12640-025-00744-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2025] [Revised: 02/21/2025] [Accepted: 04/01/2025] [Indexed: 05/03/2025]
Abstract
Increasing evidence of ocular impairments in Alzheimer's disease (AD) has drawn the attention of researchers worldwide towards retinal neurodegeneration in AD. The AD-associated changes observed in the retina include visual discrepancies, pupil size modulations, retinal nerve layer changes, retinal blood flow alterations and histopathological changes. The brain cells that act as pathological triggers for the progression of retinal neurodegeneration associated with AD are microglia, astrocytes and neurons. Various molecular pathways lead to structural and functional abnormalities in the retina, significantly affecting the brain including Aβ accumulation, apoptosis, inflammation and oxidative stress. Therapeutic agents under development that ameliorate disease conditions by targeting retinal anomalies include mesenchymal stem cell-conditioned media, BDNF, glatiramer acetate, salvianolic acid B, Lycium barbarum extract and exosomes. Investigating real-time alterations in the retina in AD may not only affect diagnostic approaches but also help to clarify neuropathological pathways and offer helpful measurements for assessing novel therapeutic approaches for AD.
Collapse
Affiliation(s)
- Rishika Dhapola
- Advanced Pharmacology and Neuroscience Laboratory, Department of Pharmacology, School of Health Sciences, Central University of Punjab, Ghudda, Bathinda, 151401, Punjab, India
| | - Prajjwal Sharma
- Advanced Pharmacology and Neuroscience Laboratory, Department of Pharmacology, School of Health Sciences, Central University of Punjab, Ghudda, Bathinda, 151401, Punjab, India
| | - Sneha Kumari
- Advanced Pharmacology and Neuroscience Laboratory, Department of Pharmacology, School of Health Sciences, Central University of Punjab, Ghudda, Bathinda, 151401, Punjab, India
| | - Balachandar Vellingiri
- Human Cytogenetics and Stem Cell Laboratory, Department of Zoology, School of Basic Sciences, Central University of Punjab, Bathinda, 151401, Punjab, India
| | - Bikash Medhi
- Department of Pharmacology, Post Graduate Institute of Medical Education and Research, Chandigarh, 160012, India
| | - Dibbanti HariKrishnaReddy
- Advanced Pharmacology and Neuroscience Laboratory, Department of Pharmacology, School of Health Sciences, Central University of Punjab, Ghudda, Bathinda, 151401, Punjab, India.
| |
Collapse
|
2
|
Olivares Ordoñez MA, Smith RC, Yiu G, Liu YA. Retinal Microstructural and Microvascular Changes in Alzheimer Disease: A Review. Int Ophthalmol Clin 2025; 65:59-67. [PMID: 39710907 PMCID: PMC11817161 DOI: 10.1097/iio.0000000000000549] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2024]
Abstract
"The eyes are a window to the brain," prompting the investigation of whether retinal biomarkers can indicate Alzheimer disease (AD) and cognitive impairment. AD is a neurodegenerative condition with a lengthy preclinical phase where pathologic changes in the central nervous system (CNS) occur before clinical symptoms. Mild cognitive impairment (MCI) often precedes AD. As part of the CNS, the retina exhibits similar pathologic changes related to AD as those seen in the brains of patients with MCI. Noninvasive imaging technologies such as optical coherence tomography (OCT) and optical coherence tomography angiography (OCTA) allow high-resolution visualization of the retina, providing an opportunity to screen and monitor AD noninvasively. In this review, we summarize the relationship between AD and retinal pathology detected by OCT and OCTA. The most common findings in patients with AD include peripapillary retinal nerve fiber layer thinning, decreased macular thickness, an enlarged foveal avascular zone, and decreased vascular densities in the superficial and deep capillary plexuses. These retinal changes correlate with magnetic resonance imaging (MRI) findings of cerebral atrophy, positron emission tomography (PET) findings of increased amyloid load, and neuropsychological testing results suggesting cognitive dysfunction. We conclude that retinal microstructural and microvascular abnormalities may serve as biomarkers for the early detection and clinical monitoring of AD and as tools for evaluating potential treatment effects. Future studies should focus on standardizing protocols for in vivo ophthalmic imaging to measure retinal pathology in AD and MCI.
Collapse
Affiliation(s)
| | | | - Glenn Yiu
- Department of Ophthalmology & Vision Science, University of California, Davis, Sacramento, CA
| | - Yin Allison Liu
- Department of Ophthalmology & Vision Science, University of California, Davis, Sacramento, CA
- Department of Neurology, University of California, Davis, Sacramento, CA
- Department of Neurological Surgery, University of California, Davis, Sacramento, CA
| |
Collapse
|
3
|
Ganguly A, Babu SS, Ghosh S, Velyutham R, Kapusetti G. Advances and future trends in the detection of beta-amyloid: A comprehensive review. Med Eng Phys 2025; 135:104269. [PMID: 39922648 DOI: 10.1016/j.medengphy.2024.104269] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2024] [Revised: 11/26/2024] [Accepted: 12/01/2024] [Indexed: 02/10/2025]
Abstract
The neurodegenerative condition known as Alzheimer's disease is typified by the build-up of beta-amyloid plaques within the brain. The timely and precise identification of beta-amyloid is essential for understanding disease progression and developing effective therapeutic interventions. This comprehensive review explores the diverse landscape of beta-amyloid detection methods, ranging from traditional immunoassays to cutting-edge technologies. The review critically examines the strengths and limitations of established techniques such as ELISA, PET, and MRI, providing insights into their roles in research and clinical settings. Emerging technologies, including electrochemical methods, nanotechnology, fluorescence techniques, point-of-care devices, and machine learning integration, are thoroughly discussed, emphasizing recent breakthroughs and their potential for revolutionizing beta-amyloid detection. Furthermore, the review delves into the challenges associated with current detection methods, such as sensitivity, specificity, and accessibility. By amalgamating knowledge from multidisciplinary approaches, this review aims to guide researchers, clinicians, and policymakers in navigating the complex landscape of beta-amyloid detection, ultimately contributing to advancements in Alzheimer's disease diagnostics and therapeutics.
Collapse
Affiliation(s)
- Atri Ganguly
- Department of Medical Devices, National Institute of Pharmaceutical Education and Research - Kolkata, -700054, India
| | - Srivalliputtur Sarath Babu
- Department of Medical Devices, National Institute of Pharmaceutical Education and Research - Kolkata, -700054, India
| | - Sumanta Ghosh
- Divison of Applied Oral Science, The University of Hong Kong, SAR, Hong Kong
| | - Ravichandiran Velyutham
- Department of Medical Devices, National Institute of Pharmaceutical Education and Research - Kolkata, -700054, India.
| | - Govinda Kapusetti
- Department of Medical Devices, National Institute of Pharmaceutical Education and Research - Kolkata, -700054, India.
| |
Collapse
|
4
|
Hong S, Baek SH, Lai MKP, Arumugam TV, Jo DG. Aging-associated sensory decline and Alzheimer's disease. Mol Neurodegener 2024; 19:93. [PMID: 39633396 PMCID: PMC11616278 DOI: 10.1186/s13024-024-00776-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Accepted: 11/08/2024] [Indexed: 12/07/2024] Open
Abstract
Multisensory decline is common as people age, and aging is the primary risk of Alzheimer's Disease (AD). Recent studies have begun to shed light on the possibility that age-related sensory decline could accelerate AD pathogenesis, or be a prodromal indicator of AD. Sensory impairments, specifically in taste and smell, often emerge before cognitive symptoms in AD, indicating their potential as early biomarkers. Olfactory dysfunction has been frequently associated with AD and may offer valuable insights into early detection. Hearing impairment is significantly associated with AD, but its causal impact on AD progression remains unclear. The review also discusses visual and tactile deficits in AD, including retinal thinning and changes in tactile perception, highlighting their links to disease progression. Focusing on molecular mechanisms, the review explores the roles of amyloid-β (Aβ) accumulation and tau protein pathology in sensory decline and their bidirectional relationship with AD. In summary, the evidence presented conclusively supports advocating for an integrated approach to understanding AD and sensory decline, to enhance early detection, implementing preventive strategies, and developing therapeutic interventions for AD. This approach underscores the significance of sensory health in addressing neurodegenerative diseases, particularly AD.
Collapse
Affiliation(s)
- Suji Hong
- The School of Pharmacy, Sungkyunkwan University, Suwon, 16419, Republic of Korea
| | - Seung-Hyun Baek
- The School of Pharmacy, Sungkyunkwan University, Suwon, 16419, Republic of Korea
| | - Mitchell K P Lai
- Department of Pharmacology, Yong Loo Lin School of Medicine, Singapore, 117600, Singapore
| | - Thiruma V Arumugam
- The School of Pharmacy, Sungkyunkwan University, Suwon, 16419, Republic of Korea.
- Centre for Cardiovascular Biology and Disease Research, La Trobe Institute for Molecular Science, Department of Microbiology, Anatomy, Physiology and Pharmacology, School of Agriculture, Biomedicine and Environment, La Trobe University, Melbourne, 3086, Australia.
| | - Dong-Gyu Jo
- The School of Pharmacy, Sungkyunkwan University, Suwon, 16419, Republic of Korea.
- Biomedical Institute for Convergence at SKKU (BICS), Suwon, 16419, Republic of Korea.
- Department of Health Sciences and Technology, SAIHST, Sungkyunkwan University, Seoul, 06355, Republic of Korea.
- Institute of Quantum Biophysics, Sungkyunkwan University, Suwon, 16419, Republic of Korea.
| |
Collapse
|
5
|
Jin Z, Chen X, Jiang C, Feng X, Shang K, Li J, Ren Q, Zhou C. Investigating ocular biomarkers and differential diagnosis of Alzheimer's disease and vascular cognitive impairment based on multimodal imaging. JOURNAL OF BIOMEDICAL OPTICS 2024; 29:126003. [PMID: 39735398 PMCID: PMC11672116 DOI: 10.1117/1.jbo.29.12.126003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/10/2024] [Revised: 12/03/2024] [Accepted: 12/10/2024] [Indexed: 12/31/2024]
Abstract
SIGNIFICANCE The eye can be used as a potential monitoring window for screening, diagnosis, and monitoring of neurological diseases. Alzheimer's disease (AD) and vascular cognitive impairment (VCI) are common causes of cognitive impairment and may share many similarities in ocular signs. Multimodal ophthalmic imaging is a technology to quantify pupillary light reaction, retinal reflectance spectrum, and hemodynamics. This provides multidimensional ocular metrics from a non-invasive approach to ocular biomarkers and differential diagnosis of AD and VCI. AIM We aim to investigate the changing pattern of ocular metrics in patients with AD and VCI using multimodal ophthalmic imaging. APPROACH Patients with subjective cognitive complaints in the memory clinic were subdivided into AD, VCI, and cognitively healthy individuals using neuropsychological and neuroimaging examinations, including positron emission tomography. All subjects underwent a medical history review, blood pressure measurement, medical optometry, intraocular pressure measurement, and custom-built multimodal ophthalmic imaging. Multidimensional parameters were analyzed by one-way analysis of variance and post hoc comparisons. RESULTS This study included 19 patients with AD, 24 patients with VCI, and 37 cognitively healthy age- and sex-matched subjects. Both AD and VCI patients showed abnormal pupillary light reactions, including decreased resting pupil diameter, pupil constriction amplitude, and maximum constriction velocity. Compared with the control group, the AD group presented increased retinal reflectance at 548 nm, whereas the VCI group presented an increased resistivity index and decreased blowout score in retinal hemodynamics. CONCLUSIONS We demonstrate that pupillary light reaction-related neurodegeneration is the common pathological change in both AD and VCI. In addition, AD is characterized by alterations in retinal spectral signatures, whereas VCI is characterized by alterations in retinal hemodynamics. These findings suggest that multimodal ophthalmic imaging may have the potential to be used as a screening tool for detecting AD and VCI.
Collapse
Affiliation(s)
- Zi Jin
- Eye Hospital, Wenzhou Medical University, National Engineering Research Center of Ophthalmology and Optometry, Wenzhou, China
- Eye Hospital, Wenzhou Medical University, National Clinical Research Center for Ocular Diseases, Wenzhou, China
- Peking University Shenzhen Graduate School, Institute of Biomedical Engineering, Shenzhen, China
- Institute of Biomedical Engineering, Shenzhen Bay Laboratory, Shenzhen, China
| | - Xuhui Chen
- Peking University Shenzhen Hospital, Department of Neurology, Shenzhen, China
| | - Chunxia Jiang
- Peking University Shenzhen Hospital, Department of Ophthalmology, Shenzhen, China
| | - Ximeng Feng
- Peking University Shenzhen Graduate School, Institute of Biomedical Engineering, Shenzhen, China
- Institute of Biomedical Engineering, Shenzhen Bay Laboratory, Shenzhen, China
- Peking University, College of Future Technology, Department of Biomedical Engineering, Beijing, China
| | - Kun Shang
- Shanghai University of Medicine and Health Sciences, College of Medical Instruments, Shanghai, China
| | - Jinying Li
- Peking University Shenzhen Hospital, Department of Ophthalmology, Shenzhen, China
| | - Qiushi Ren
- Peking University Shenzhen Graduate School, Institute of Biomedical Engineering, Shenzhen, China
- Institute of Biomedical Engineering, Shenzhen Bay Laboratory, Shenzhen, China
- Peking University, College of Future Technology, Department of Biomedical Engineering, Beijing, China
| | - Chuanqing Zhou
- Shanghai University of Medicine and Health Sciences, College of Medical Instruments, Shanghai, China
| |
Collapse
|
6
|
Jin Z, Chen X, Jiang C, Feng X, Zou D, Lu Y, Li J, Ren Q, Zhou C. Predicting the cognitive impairment with multimodal ophthalmic imaging and artificial neural network for community screening. Br J Ophthalmol 2024; 108:1737-1742. [PMID: 38697799 DOI: 10.1136/bjo-2023-323283] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2023] [Accepted: 04/18/2024] [Indexed: 05/05/2024]
Abstract
BACKGROUND/AIMS To investigate the comprehensive prediction ability for cognitive impairment in a general elder population using the combination of the multimodal ophthalmic imaging and artificial neural networks. METHODS Patients with cognitive impairment and cognitively healthy individuals were recruited. All subjects underwent medical history, blood pressure measurement, the Montreal Cognitive Assessment, medical optometry, intraocular pressure and custom-built multimodal ophthalmic imaging, which integrated pupillary light reaction, multispectral imaging, laser speckle contrast imaging and retinal oximetry. Multidimensional parameters were analysed by Student's t-test. Logistic regression analysis and back-propagation neural network (BPNN) were used to identify the predictive capability for cognitive impairment. RESULTS This study included 104 cognitive impairment patients (61.5% female; mean (SD) age, 68.3 (9.4) years), and 94 cognitively healthy age-matched and sex-matched subjects (56.4% female; mean (SD) age, 65.9 (7.6) years). The variation of most parameters including decreased pupil constriction amplitude (CA), relative CA, average constriction velocity, venous diameter, venous blood flow and increased centred retinal reflectance in 548 nm (RC548) in cognitive impairment was consistent with previous studies while the reduced flow acceleration index and oxygen metabolism were reported for the first time. Compared with the logistic regression model, BPNN had better predictive performance (accuracy: 0.91 vs 0.69; sensitivity: 93.3% vs 61.70%; specificity: 90.0% vs 68.66%). CONCLUSIONS This study demonstrates retinal spectral signature alteration, neurodegeneration and angiopathy occur concurrently in cognitive impairment. The combination of multimodal ophthalmic imaging and BPNN can be a useful tool for predicting cognitive impairment with high performance for community screening.
Collapse
Affiliation(s)
- Zi Jin
- Department of Biomedical Engineering, Peking University Shenzhen Graduate School, Shenzhen, China
- Department of Biomedical Engineering, Shenzhen Bay Laboratory, Shenzhen, China
| | - Xuhui Chen
- Department of Neurology, Peking University Shenzhen Hospital, Shenzhen, China
| | - Chunxia Jiang
- Department of Ophthalmology, Peking University Shenzhen Hospital, Shenzhen, China
| | - Ximeng Feng
- Department of Biomedical Engineering, Peking University Shenzhen Graduate School, Shenzhen, China
- Department of Biomedical Engineering, Shenzhen Bay Laboratory, Shenzhen, China
- Department of Biomedical Engineering, Peking University, Beijing, China
- Institute of Medical Technology, Peking University Health Science Centre, Beijing, China
| | - Da Zou
- Department of Biomedical Engineering, Peking University Shenzhen Graduate School, Shenzhen, China
- Department of Biomedical Engineering, Shenzhen Bay Laboratory, Shenzhen, China
- Department of Biomedical Engineering, Peking University, Beijing, China
- Institute of Medical Technology, Peking University Health Science Centre, Beijing, China
| | - Yanye Lu
- Department of Biomedical Engineering, Peking University, Beijing, China
- Institute of Medical Technology, Peking University Health Science Centre, Beijing, China
| | - Jinying Li
- Department of Ophthalmology, Peking University Shenzhen Hospital, Shenzhen, China
| | - Qiushi Ren
- Department of Biomedical Engineering, Peking University Shenzhen Graduate School, Shenzhen, China
- Department of Biomedical Engineering, Shenzhen Bay Laboratory, Shenzhen, China
- Department of Biomedical Engineering, Peking University, Beijing, China
- Institute of Medical Technology, Peking University Health Science Centre, Beijing, China
| | - Chuanqing Zhou
- College of Medical Instruments, Shanghai University of Medicine and Health Sciences, Shanghai, China
| |
Collapse
|
7
|
Cao Q, Yang S, Wang X, Sun H, Chen W, Wang Y, Gao J, Wu Y, Yang Q, Chen X, Yuan S, Xiao M, Nedergaard M, Huo Y, Liu Q. Transport of β-amyloid from brain to eye causes retinal degeneration in Alzheimer's disease. J Exp Med 2024; 221:e20240386. [PMID: 39316084 PMCID: PMC11448872 DOI: 10.1084/jem.20240386] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2024] [Revised: 07/03/2024] [Accepted: 08/12/2024] [Indexed: 09/25/2024] Open
Abstract
The eye is closely connected to the brain, providing a unique window to detect pathological changes in the brain. In this study, we discovered β-amyloid (Aβ) deposits along the ocular glymphatic system in patients with Alzheimer's disease (AD) and 5×FAD transgenic mouse model. Interestingly, Aβ from the brain can flow into the eyes along the optic nerve through cerebrospinal fluid (CSF), causing retinal degeneration. Aβ is mainly observed in the optic nerve sheath, the neural axon, and the perivascular space, which might represent the critical steps of the Aβ transportation from the brain to the eyes. Aquaporin-4 facilitates the influx of Aβ in brain-eye transport and out-excretion of the retina, and its absence or loss of polarity exacerbates brain-derived Aβ induced damage and visual impairment. These results revealed brain-to-eye Aβ transport as a major contributor to AD retinopathy, highlighting a new therapeutic avenue in ocular and neurodegenerative disease.
Collapse
Affiliation(s)
- Qiuchen Cao
- Department of Ophthalmology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
- Jiangsu Province Key Laboratory of Neurodegeneration, Nanjing Medical University, Nanjing, China
- Department of Ophthalmology, Baylor College of Medicine, Houston, TX, USA
- Department of Cellular Biology and Anatomy, Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, GA, USA
| | - Shige Yang
- Department of Ophthalmology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Xiaowei Wang
- Faculty of Medical and Health Sciences, Center for Translational Neuromedicine, University of Copenhagen, Copenhagen, Denmark
- Department of Neurosurgery, Center for Translational Neuromedicine, University of Rochester Medical Center, Rochester, NY, USA
| | - Huaiqing Sun
- Jiangsu Province Key Laboratory of Neurodegeneration, Nanjing Medical University, Nanjing, China
- Department of Neurology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Weijie Chen
- Department of Ophthalmology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Yuliang Wang
- Department of Immunology, Key Laboratory of Immune Microenvironment and Diseases, Nanjing Medical University, Nanjing, China
| | - Junying Gao
- Jiangsu Province Key Laboratory of Neurodegeneration, Nanjing Medical University, Nanjing, China
| | - Yanchi Wu
- Department of Ophthalmology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Qiuhua Yang
- Department of Cellular Biology and Anatomy, Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, GA, USA
| | - Xue Chen
- Department of Ophthalmology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Songtao Yuan
- Department of Ophthalmology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Ming Xiao
- Jiangsu Province Key Laboratory of Neurodegeneration, Nanjing Medical University, Nanjing, China
- Nanjing Brain Hospital, Brain Institute, Nanjing Medical University , Nanjing, China
| | - Maiken Nedergaard
- Faculty of Medical and Health Sciences, Center for Translational Neuromedicine, University of Copenhagen, Copenhagen, Denmark
- Department of Neurosurgery, Center for Translational Neuromedicine, University of Rochester Medical Center, Rochester, NY, USA
| | - Yuqing Huo
- Department of Ophthalmology, Baylor College of Medicine, Houston, TX, USA
- Department of Cellular Biology and Anatomy, Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, GA, USA
| | - Qinghuai Liu
- Department of Ophthalmology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| |
Collapse
|
8
|
Sheehan N, Bannai D, Silverstein SM, Lizano P. Neuroretinal Alterations in Schizophrenia and Bipolar Disorder: An Updated Meta-analysis. Schizophr Bull 2024; 50:1067-1082. [PMID: 38954839 PMCID: PMC11349028 DOI: 10.1093/schbul/sbae102] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 07/04/2024]
Abstract
Schizophrenia (SZ) and bipolar disorder (BD) are characterized by major symptomatic, cognitive, and neuroanatomical changes. Recent studies have used optical coherence tomography (OCT) to investigate retinal changes in SZ and BD, but their unique and shared changes require further evaluation. Articles were identified using PubMed and Google Scholar. 39 studies met the inclusion criteria. Diagnostic groups were proband (SZ/BD combined), SZ, BD, and healthy control (HC) eyes. Meta-analyses utilized fixed and random effects models when appropriate, and publication bias was corrected using trim-and-fill analysis ("meta" package in R). Results are reported as standardized mean differences with 95% CIs. Data from 3145 patient eyes (1956 SZ, 1189 BD) and 3135 HC eyes were included. Studies identified thinning of the peripapillary retinal nerve fiber layer (pRNFL, overall and in 2 subregions), m-Retina (overall and all subregions), mGCL-IPL, mIPL, and mRPE in SZ patients. BD showed thinning of the pRNFL (overall and in each subregion), pGCC, and macular Retina (in 5 subregions), but no changes in thickness or volume for the total retina. Neither SZ nor BD patients demonstrated significant changes in the fovea, mRNFL, mGCL, mGCC, mINL, mOPL, mONL, or choroid thicknesses. Moderating effects of age, illness duration, and smoking on retinal structures were identified. This meta-analysis builds upon previous literature in this field by incorporating recent OCT studies and examining both peripapillary and macular retinal regions with respect to psychotic disorders. Overall, this meta-analysis demonstrated both peripapillary and macular structural retinal abnormalities in people with SZ or BD compared with HCs.
Collapse
Affiliation(s)
- Nora Sheehan
- Department of Psychiatry, Beth Israel Deaconess Medical Center, Boston, MA, USA
- Division of Translational Neuroscience, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Deepthi Bannai
- Department of Psychiatry, Beth Israel Deaconess Medical Center, Boston, MA, USA
- Division of Translational Neuroscience, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Steven M Silverstein
- Department of Psychiatry, University of Rochester Medical Center, Rochester, NY, USA
- Department of Neuroscience, University of Rochester Medical Center, Rochester, NY, USA
- Department of Ophthalmology, University of Rochester Medical Center, Rochester, NY, USA
| | - Paulo Lizano
- Department of Psychiatry, Beth Israel Deaconess Medical Center, Boston, MA, USA
- Division of Translational Neuroscience, Beth Israel Deaconess Medical Center, Boston, MA, USA
- Department of Psychiatry, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
9
|
Vagiakis I, Bakirtzis C, Andravizou A, Pirounides D. Unlocking the Potential of Vessel Density and the Foveal Avascular Zone in Optical Coherence Tomography Angiography as Biomarkers in Alzheimer's Disease. Healthcare (Basel) 2024; 12:1589. [PMID: 39201148 PMCID: PMC11353459 DOI: 10.3390/healthcare12161589] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Revised: 08/06/2024] [Accepted: 08/07/2024] [Indexed: 09/02/2024] Open
Abstract
Alzheimer's disease is the most prevalent form of dementia. Apart from its traditional clinical diagnostic methods, novel ocular imaging biomarkers have the potential to significantly enhance the diagnosis of Alzheimer's disease. Ophthalmologists might be able to play a crucial role in this multidisciplinary approach, aiding in the early detection and diagnosis of Alzheimer's disease through the use of advanced retinal imaging techniques. This systematic literature review the utilization of optical coherence tomography angiography biomarkers, specifically vessel density and the foveal avascular zone, for the diagnosis of Alzheimer's disease. A comprehensive search was performed across multiple academic journal databases, including 11 relevant studies. The selected studies underwent thorough analysis to assess the potential of these optical coherence tomography angiography biomarkers as diagnostic tools for Alzheimer's disease. The assessment of vessel density and the foveal avascular zone have emerged as a promising avenue for identifying and diagnosing Alzheimer's disease. However, it is imperative to acknowledge that further targeted investigations are warranted to address the inherent limitations of the existing body of literature. These limitations encompass various factors such as modest sample sizes, heterogeneity among study populations, disparities in optical coherence tomography angiography imaging protocols, and inconsistencies in the reported findings. In order to establish the clinical utility and robustness of these biomarkers in Alzheimer's disease diagnosis, future research endeavors should strive to overcome these limitations by implementing larger-scale studies characterized by standardized protocols and comprehensive assessments.
Collapse
Affiliation(s)
- Iordanis Vagiakis
- Department of Ophthalmology, AHEPA University Hospital, 54626 Thessaloniki, Greece;
| | - Christos Bakirtzis
- Second Department of Neurology, School of Medicine, Aristotle University of Thessaloniki, 54621 Thessaloniki, Greece;
| | - Athina Andravizou
- Second Department of Neurology, School of Medicine, Aristotle University of Thessaloniki, 54621 Thessaloniki, Greece;
| | - Demetrios Pirounides
- Department of Ophthalmology, AHEPA University Hospital, 54626 Thessaloniki, Greece;
| |
Collapse
|
10
|
Gaire BP, Koronyo Y, Fuchs DT, Shi H, Rentsendorj A, Danziger R, Vit JP, Mirzaei N, Doustar J, Sheyn J, Hampel H, Vergallo A, Davis MR, Jallow O, Baldacci F, Verdooner SR, Barron E, Mirzaei M, Gupta VK, Graham SL, Tayebi M, Carare RO, Sadun AA, Miller CA, Dumitrascu OM, Lahiri S, Gao L, Black KL, Koronyo-Hamaoui M. Alzheimer's disease pathophysiology in the Retina. Prog Retin Eye Res 2024; 101:101273. [PMID: 38759947 PMCID: PMC11285518 DOI: 10.1016/j.preteyeres.2024.101273] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2023] [Revised: 04/23/2024] [Accepted: 05/10/2024] [Indexed: 05/19/2024]
Abstract
The retina is an emerging CNS target for potential noninvasive diagnosis and tracking of Alzheimer's disease (AD). Studies have identified the pathological hallmarks of AD, including amyloid β-protein (Aβ) deposits and abnormal tau protein isoforms, in the retinas of AD patients and animal models. Moreover, structural and functional vascular abnormalities such as reduced blood flow, vascular Aβ deposition, and blood-retinal barrier damage, along with inflammation and neurodegeneration, have been described in retinas of patients with mild cognitive impairment and AD dementia. Histological, biochemical, and clinical studies have demonstrated that the nature and severity of AD pathologies in the retina and brain correspond. Proteomics analysis revealed a similar pattern of dysregulated proteins and biological pathways in the retina and brain of AD patients, with enhanced inflammatory and neurodegenerative processes, impaired oxidative-phosphorylation, and mitochondrial dysfunction. Notably, investigational imaging technologies can now detect AD-specific amyloid deposits, as well as vasculopathy and neurodegeneration in the retina of living AD patients, suggesting alterations at different disease stages and links to brain pathology. Current and exploratory ophthalmic imaging modalities, such as optical coherence tomography (OCT), OCT-angiography, confocal scanning laser ophthalmoscopy, and hyperspectral imaging, may offer promise in the clinical assessment of AD. However, further research is needed to deepen our understanding of AD's impact on the retina and its progression. To advance this field, future studies require replication in larger and diverse cohorts with confirmed AD biomarkers and standardized retinal imaging techniques. This will validate potential retinal biomarkers for AD, aiding in early screening and monitoring.
Collapse
Affiliation(s)
- Bhakta Prasad Gaire
- Department of Neurosurgery, Maxine Dunitz Neurosurgical Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Yosef Koronyo
- Department of Neurosurgery, Maxine Dunitz Neurosurgical Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Dieu-Trang Fuchs
- Department of Neurosurgery, Maxine Dunitz Neurosurgical Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Haoshen Shi
- Department of Neurosurgery, Maxine Dunitz Neurosurgical Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Altan Rentsendorj
- Department of Neurosurgery, Maxine Dunitz Neurosurgical Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Ron Danziger
- Department of Neurology, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Jean-Philippe Vit
- Department of Neurosurgery, Maxine Dunitz Neurosurgical Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Nazanin Mirzaei
- Department of Neurosurgery, Maxine Dunitz Neurosurgical Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Jonah Doustar
- Department of Neurosurgery, Maxine Dunitz Neurosurgical Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Julia Sheyn
- Department of Neurosurgery, Maxine Dunitz Neurosurgical Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Harald Hampel
- Sorbonne University, Alzheimer Precision Medicine (APM), AP-HP, Pitié-Salpêtrière Hospital, Paris, France
| | - Andrea Vergallo
- Sorbonne University, Alzheimer Precision Medicine (APM), AP-HP, Pitié-Salpêtrière Hospital, Paris, France
| | - Miyah R Davis
- Department of Neurosurgery, Maxine Dunitz Neurosurgical Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Ousman Jallow
- Department of Neurosurgery, Maxine Dunitz Neurosurgical Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Filippo Baldacci
- Sorbonne University, Alzheimer Precision Medicine (APM), AP-HP, Pitié-Salpêtrière Hospital, Paris, France; Department of Clinical and Experimental Medicine, Neurology Unit, University of Pisa, Pisa, Italy
| | | | - Ernesto Barron
- Department of Ophthalmology, David Geffen School of Medicine at University of California Los Angeles, Los Angeles, CA, USA; Doheny Eye Institute, Los Angeles, CA, USA
| | - Mehdi Mirzaei
- Department of Clinical Medicine, Health and Human Sciences, Macquarie Medical School, Macquarie University, Sydney, NSW, Australia
| | - Vivek K Gupta
- Department of Clinical Medicine, Health and Human Sciences, Macquarie Medical School, Macquarie University, Sydney, NSW, Australia
| | - Stuart L Graham
- Department of Clinical Medicine, Health and Human Sciences, Macquarie Medical School, Macquarie University, Sydney, NSW, Australia; Department of Clinical Medicine, Macquarie University, Sydney, NSW, Australia
| | - Mourad Tayebi
- School of Medicine, Western Sydney University, Campbelltown, NSW, Australia
| | - Roxana O Carare
- Department of Clinical Neuroanatomy, University of Southampton, Southampton, UK
| | - Alfredo A Sadun
- Department of Ophthalmology, David Geffen School of Medicine at University of California Los Angeles, Los Angeles, CA, USA; Doheny Eye Institute, Los Angeles, CA, USA
| | - Carol A Miller
- Department of Pathology Program in Neuroscience, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | | | - Shouri Lahiri
- Department of Neurology, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Liang Gao
- Department of Bioengineering, University of California Los Angeles, Los Angeles, CA, USA
| | - Keith L Black
- Department of Neurosurgery, Maxine Dunitz Neurosurgical Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Maya Koronyo-Hamaoui
- Department of Neurosurgery, Maxine Dunitz Neurosurgical Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA; Department of Neurology, Cedars-Sinai Medical Center, Los Angeles, CA, USA; Department of Biomedical Sciences, Division of Applied Cell Biology and Physiology, Cedars-Sinai Medical Center, Los Angeles, CA, USA.
| |
Collapse
|
11
|
Sun Z, Zhang B, Smith S, Atan D, Khawaja AP, Stuart KV, Luben RN, Biradar MI, McGillivray T, Patel PJ, Khaw PT, Petzold A, Foster PJ, the UK Biobank Eye and Vision Consortium. Structural correlations between brain magnetic resonance image-derived phenotypes and retinal neuroanatomy. Eur J Neurol 2024; 31:e16288. [PMID: 38716763 PMCID: PMC11235673 DOI: 10.1111/ene.16288] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2023] [Revised: 03/08/2024] [Accepted: 03/12/2024] [Indexed: 07/28/2024]
Abstract
BACKGROUND AND PURPOSE The eye is a well-established model of brain structure and function, yet region-specific structural correlations between the retina and the brain remain underexplored. Therefore, we aim to explore and describe the relationships between the retinal layer thicknesses and brain magnetic resonance image (MRI)-derived phenotypes in UK Biobank. METHODS Participants with both quality-controlled optical coherence tomography (OCT) and brain MRI were included in this study. Retinal sublayer thicknesses and total macular thickness were derived from OCT scans. Brain image-derived phenotypes (IDPs) of 153 cortical and subcortical regions were processed from MRI scans. We utilized multivariable linear regression models to examine the association between retinal thickness and brain regional volumes. All analyses were corrected for multiple testing and adjusted for confounders. RESULTS Data from 6446 participants were included in this study. We identified significant associations between volumetric brain MRI measures of subregions in the occipital lobe (intracalcarine cortex), parietal lobe (postcentral gyrus), cerebellum (lobules VI, VIIb, VIIIa, VIIIb, and IX), and deep brain structures (thalamus, hippocampus, caudate, putamen, pallidum, and accumbens) and the thickness of the innermost retinal sublayers and total macular thickness (all p < 3.3 × 10-5). We did not observe statistically significant associations between brain IDPs and the thickness of the outer retinal sublayers. CONCLUSIONS Thinner inner and total retinal thicknesses are associated with smaller volumes of specific brain regions. Notably, these relationships extend beyond anatomically established retina-brain connections.
Collapse
Affiliation(s)
- Zihan Sun
- National Institute for Health Research Biomedical Research Centre at Moorfields Eye Hospital National Health Service Foundation Trust and University College London Institute of OphthalmologyLondonUK
| | - Bing Zhang
- National Clinical Research Centre for Ocular Diseases, Eye HospitalWenzhou Medical UniversityWenzhouChina
| | - Stephen Smith
- Wellcome Centre for Integrative Neuroimaging (WIN Functional Magnetic Resonance Imaging Building)University of OxfordOxfordUK
| | - Denize Atan
- Bristol Eye HospitalUniversity Hospitals Bristol and Weston NHS Foundation TrustBristolUK
- Translational Health Sciences, Bristol Medical SchoolUniversity of BristolBristolUK
| | - Anthony P. Khawaja
- National Institute for Health Research Biomedical Research Centre at Moorfields Eye Hospital National Health Service Foundation Trust and University College London Institute of OphthalmologyLondonUK
| | - Kelsey V. Stuart
- National Institute for Health Research Biomedical Research Centre at Moorfields Eye Hospital National Health Service Foundation Trust and University College London Institute of OphthalmologyLondonUK
| | - Robert N. Luben
- National Institute for Health Research Biomedical Research Centre at Moorfields Eye Hospital National Health Service Foundation Trust and University College London Institute of OphthalmologyLondonUK
| | - Mahantesh I. Biradar
- National Institute for Health Research Biomedical Research Centre at Moorfields Eye Hospital National Health Service Foundation Trust and University College London Institute of OphthalmologyLondonUK
| | | | - Praveen J. Patel
- National Institute for Health Research Biomedical Research Centre at Moorfields Eye Hospital National Health Service Foundation Trust and University College London Institute of OphthalmologyLondonUK
| | - Peng T. Khaw
- National Institute for Health Research Biomedical Research Centre at Moorfields Eye Hospital National Health Service Foundation Trust and University College London Institute of OphthalmologyLondonUK
| | - Axel Petzold
- Queen Square Institute of Neurology, University College London, Department of Molecular NeurosciencesMoorfields Eye Hospital and National Hospital for Neurology and NeurosurgeryLondonUK
- Departments of Neurology and Ophthalmology and Expertise Center for Neuro‐ophthalmologyAmsterdam University Medical CentreAmsterdamthe Netherlands
| | - Paul J. Foster
- National Institute for Health Research Biomedical Research Centre at Moorfields Eye Hospital National Health Service Foundation Trust and University College London Institute of OphthalmologyLondonUK
| | | |
Collapse
|
12
|
Dumitrascu OM, Doustar J, Fuchs DT, Koronyo Y, Sherman DS, Miller MS, Johnson KO, Carare RO, Verdooner SR, Lyden PD, Schneider JA, Black KL, Koronyo-Hamaoui M. Retinal peri-arteriolar versus peri-venular amyloidosis, hippocampal atrophy, and cognitive impairment: exploratory trial. Acta Neuropathol Commun 2024; 12:109. [PMID: 38943220 PMCID: PMC11212356 DOI: 10.1186/s40478-024-01810-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Accepted: 06/02/2024] [Indexed: 07/01/2024] Open
Abstract
The relationship between amyloidosis and vasculature in cognitive impairment and Alzheimer's disease (AD) pathogenesis is increasingly acknowledged. We conducted a quantitative and topographic assessment of retinal perivascular amyloid plaque (AP) distribution in individuals with both normal and impaired cognition. Using a retrospective dataset of scanning laser ophthalmoscopy fluorescence images from twenty-eight subjects with varying cognitive states, we developed a novel image processing method to examine retinal peri-arteriolar and peri-venular curcumin-positive AP burden. We further correlated retinal perivascular amyloidosis with neuroimaging measures and neurocognitive scores. Our study unveiled that peri-arteriolar AP counts surpassed peri-venular counts throughout the entire cohort (P < 0.0001), irrespective of the primary, secondary, or tertiary vascular branch location, with a notable increase among cognitively impaired individuals. Moreover, secondary branch peri-venular AP count was elevated in the cognitively impaired (P < 0.01). Significantly, peri-venular AP count, particularly in secondary and tertiary venules, exhibited a strong correlation with clinical dementia rating, Montreal cognitive assessment score, hippocampal volume, and white matter hyperintensity count. In conclusion, our exploratory analysis detected greater peri-arteriolar versus peri-venular amyloidosis and a marked elevation of amyloid deposition in secondary branch peri-venular regions among cognitively impaired subjects. These findings underscore the potential feasibility of retinal perivascular amyloid imaging in predicting cognitive decline and AD progression. Larger longitudinal studies encompassing diverse populations and AD-biomarker confirmation are warranted to delineate the temporal-spatial dynamics of retinal perivascular amyloid deposition in cognitive impairment and the AD continuum.
Collapse
Affiliation(s)
- Oana M Dumitrascu
- Departments of Neurology, Mayo Clinic, AZ, 13400 E. Shea Blvd, Scottsdale, AZ, 85259, USA.
| | - Jonah Doustar
- Department of Neurosurgery, Cedars-Sinai Medical Center, Maxine Dunitz Neurosurgical Institute, 127 S. San Vicente Blvd., Los Angeles, CA, 90048, USA
| | - Dieu-Trang Fuchs
- Department of Neurosurgery, Cedars-Sinai Medical Center, Maxine Dunitz Neurosurgical Institute, 127 S. San Vicente Blvd., Los Angeles, CA, 90048, USA
| | - Yosef Koronyo
- Department of Neurosurgery, Cedars-Sinai Medical Center, Maxine Dunitz Neurosurgical Institute, 127 S. San Vicente Blvd., Los Angeles, CA, 90048, USA
| | - Dale S Sherman
- Department of Physical Medicine and Rehabilitation, Cedars-Sinai Medical Center, 127 S. San Vicente Blvd., Los Angeles, CA, 90048, USA
| | - Michelle Shizu Miller
- Department of Neurosurgery, Cedars-Sinai Medical Center, Maxine Dunitz Neurosurgical Institute, 127 S. San Vicente Blvd., Los Angeles, CA, 90048, USA
- Department of Neurosurgery, Tulane University School of Medicine, 1415 Tulane Ave, New Orleans, LA, 70112, USA
| | - Kenneth O Johnson
- NeuroVision Imaging LLC, 1395 Garden Hwy, Sacramento, CA, 95833, USA
| | - Roxana O Carare
- Department of Clinical Neuroanatomy, University of Southampton, University Road Southampton, Southampton, SO17 1BJ, UK
| | | | - Patrick D Lyden
- Department of Physiology and Neuroscience, Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, 1501 San Pablo St, Los Angeles, CA, 90033, USA
| | - Julie A Schneider
- Department of Pathology, Department of Neurological Sciences, Alzheimer's Disease Research Center, Rush Medical College, Rush University, 600 S. Paulina St., Chicago, IL, 60612, USA
| | - Keith L Black
- Department of Neurosurgery, Cedars-Sinai Medical Center, Maxine Dunitz Neurosurgical Institute, 127 S. San Vicente Blvd., Los Angeles, CA, 90048, USA
| | - Maya Koronyo-Hamaoui
- Department of Neurosurgery, Cedars-Sinai Medical Center, Maxine Dunitz Neurosurgical Institute, 127 S. San Vicente Blvd., Los Angeles, CA, 90048, USA.
- Department of Neurology, Cedars-Sinai Medical Center, 127 S. San Vicente Blvd., Los Angeles, CA, 90048, USA.
- Division of Applied Cell Biology and Physiology, Department of Biomedical Sciences, Cedars-Sinai Medical Center, 127 S. San Vicente Blvd., Los Angeles, CA, 90048, USA.
| |
Collapse
|
13
|
Duman Sastim D, Elboga G, Elboga U, Gungor K. Evaluation of the relationship between FDG-PET hypometabolism and retinal layer thickness in patients with Alzheimer's disease. Acta Neurol Belg 2024; 124:987-993. [PMID: 38546932 DOI: 10.1007/s13760-024-02511-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2023] [Accepted: 02/23/2024] [Indexed: 06/01/2024]
Abstract
We aimed to investigate the diagnostic value of Optical coherence tomography (OCT) in Alzheimer's disease (AD) and to assess the correlation between OCT and fluorodeoxyglucose (FDG)-positron emission tomography (PET) which shows high diagnostic agreement with findings from postmortem histopathology-the gold standard method. Patients who were diagnosed with AD-related dementia were selected for the study. Patients with a mini mental test (MMT) score between 18 and 23 were included in the study (n = 31). Volunteers with MMT ≥ 28 and no cognitive impairment were included in the study as the control group (n = 31). OCT imaging was performed in the patient and control groups after detailed ophthalmological examinations including visual acuity and intraocular pressure measurements. Brain glucose metabolism measurement was performed using 18 F-FDG PET/computed tomography. When adjusted for age and sex, mean retinal nerve fiber layer thickness (RNFL) thickness showed a significant difference between groups and the RNFL thickness in the superior temporal and superior nasal quadrants in AD-related mild dementia group showed a significant difference (p < 0.05). Furthermore, only the RNFL thickness in the inferior nasal quadrant of the right eye showed a significant difference between the groups (p = 0.016). It is thought that OCT is a promising imaging method in the elderly population due to its low-cost, non-invasive and easily applicability, and therefore, it may contribute in the future as a tool in the periodic follow-up of patients diagnosed with AD.
Collapse
Affiliation(s)
- Demet Duman Sastim
- Department of Psychiatry, Tunceli State Hospital, Merkez, 62000, Merkez/Tunceli, Turkey.
| | - Gulcin Elboga
- Faculty of Medicine, Department of Psychiatry, Gaziantep University, Gaziantep, Turkey
| | - Umut Elboga
- Faculty of Medicine, Department of Nuclear Medicine, Gaziantep University, Gaziantep, Turkey
| | - Kivanc Gungor
- Faculty of Medicine, Department of Ophthalmology, Gaziantep University, Gaziantep, Turkey
| |
Collapse
|
14
|
Abyadeh M, Gupta V, Paulo JA, Mahmoudabad AG, Shadfar S, Mirshahvaladi S, Gupta V, Nguyen CT, Finkelstein DI, You Y, Haynes PA, Salekdeh GH, Graham SL, Mirzaei M. Amyloid-beta and tau protein beyond Alzheimer's disease. Neural Regen Res 2024; 19:1262-1276. [PMID: 37905874 PMCID: PMC11467936 DOI: 10.4103/1673-5374.386406] [Citation(s) in RCA: 34] [Impact Index Per Article: 34.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2023] [Revised: 08/08/2023] [Accepted: 09/07/2023] [Indexed: 11/02/2023] Open
Abstract
ABSTRACT The aggregation of amyloid-beta peptide and tau protein dysregulation are implicated to play key roles in Alzheimer's disease pathogenesis and are considered the main pathological hallmarks of this devastating disease. Physiologically, these two proteins are produced and expressed within the normal human body. However, under pathological conditions, abnormal expression, post-translational modifications, conformational changes, and truncation can make these proteins prone to aggregation, triggering specific disease-related cascades. Recent studies have indicated associations between aberrant behavior of amyloid-beta and tau proteins and various neurological diseases, such as Alzheimer's disease, Parkinson's disease, and amyotrophic lateral sclerosis, as well as retinal neurodegenerative diseases like Glaucoma and age-related macular degeneration. Additionally, these proteins have been linked to cardiovascular disease, cancer, traumatic brain injury, and diabetes, which are all leading causes of morbidity and mortality. In this comprehensive review, we provide an overview of the connections between amyloid-beta and tau proteins and a spectrum of disorders.
Collapse
Affiliation(s)
| | - Vivek Gupta
- Department of Clinical Medicine, Faculty of Medicine, Health and Human Sciences, Macquarie Medical School, Macquarie University, Macquarie Park, North Ryde, Sydney, NSW, Australia
| | - Joao A. Paulo
- Department of Cell Biology, Harvard Medical School, Boston, MA, USA
| | | | - Sina Shadfar
- Department of Clinical Medicine, Faculty of Medicine, Health and Human Sciences, Macquarie Medical School, Macquarie University, Macquarie Park, North Ryde, Sydney, NSW, Australia
| | - Shahab Mirshahvaladi
- Department of Clinical Medicine, Faculty of Medicine, Health and Human Sciences, Macquarie Medical School, Macquarie University, Macquarie Park, North Ryde, Sydney, NSW, Australia
| | - Veer Gupta
- School of Medicine, Deakin University, Geelong, VIC, Australia
| | - Christine T.O. Nguyen
- Department of Optometry and Vision Sciences, School of Health Sciences, Faculty of Medicine Dentistry and Health Sciences, University of Melbourne, Parkville, VIC, Australia
| | - David I. Finkelstein
- The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, VIC, Australia
| | - Yuyi You
- Department of Clinical Medicine, Faculty of Medicine, Health and Human Sciences, Macquarie Medical School, Macquarie University, Macquarie Park, North Ryde, Sydney, NSW, Australia
| | - Paul A. Haynes
- School of Natural Sciences, Macquarie University, Macquarie Park, NSW, Australia
| | - Ghasem H. Salekdeh
- School of Natural Sciences, Macquarie University, Macquarie Park, NSW, Australia
| | - Stuart L. Graham
- Department of Clinical Medicine, Faculty of Medicine, Health and Human Sciences, Macquarie Medical School, Macquarie University, Macquarie Park, North Ryde, Sydney, NSW, Australia
| | - Mehdi Mirzaei
- Department of Clinical Medicine, Faculty of Medicine, Health and Human Sciences, Macquarie Medical School, Macquarie University, Macquarie Park, North Ryde, Sydney, NSW, Australia
| |
Collapse
|
15
|
Tan YY, Kang HG, Lee CJ, Kim SS, Park S, Thakur S, Da Soh Z, Cho Y, Peng Q, Lee K, Tham YC, Rim TH, Cheng CY. Prognostic potentials of AI in ophthalmology: systemic disease forecasting via retinal imaging. EYE AND VISION (LONDON, ENGLAND) 2024; 11:17. [PMID: 38711111 PMCID: PMC11071258 DOI: 10.1186/s40662-024-00384-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Accepted: 04/17/2024] [Indexed: 05/08/2024]
Abstract
BACKGROUND Artificial intelligence (AI) that utilizes deep learning (DL) has potential for systemic disease prediction using retinal imaging. The retina's unique features enable non-invasive visualization of the central nervous system and microvascular circulation, aiding early detection and personalized treatment plans for personalized care. This review explores the value of retinal assessment, AI-based retinal biomarkers, and the importance of longitudinal prediction models in personalized care. MAIN TEXT This narrative review extensively surveys the literature for relevant studies in PubMed and Google Scholar, investigating the application of AI-based retina biomarkers in predicting systemic diseases using retinal fundus photography. The study settings, sample sizes, utilized AI models and corresponding results were extracted and analysed. This review highlights the substantial potential of AI-based retinal biomarkers in predicting neurodegenerative, cardiovascular, and chronic kidney diseases. Notably, DL algorithms have demonstrated effectiveness in identifying retinal image features associated with cognitive decline, dementia, Parkinson's disease, and cardiovascular risk factors. Furthermore, longitudinal prediction models leveraging retinal images have shown potential in continuous disease risk assessment and early detection. AI-based retinal biomarkers are non-invasive, accurate, and efficient for disease forecasting and personalized care. CONCLUSION AI-based retinal imaging hold promise in transforming primary care and systemic disease management. Together, the retina's unique features and the power of AI enable early detection, risk stratification, and help revolutionizing disease management plans. However, to fully realize the potential of AI in this domain, further research and validation in real-world settings are essential.
Collapse
Affiliation(s)
| | - Hyun Goo Kang
- Division of Retina, Severance Eye Hospital, Yonsei University College of Medicine, Seoul, South Korea
| | - Chan Joo Lee
- Division of Cardiology, Severance Cardiovascular Hospital, Yonsei University College of Medicine, Seoul, South Korea
| | - Sung Soo Kim
- Division of Retina, Severance Eye Hospital, Yonsei University College of Medicine, Seoul, South Korea
| | - Sungha Park
- Division of Cardiology, Severance Cardiovascular Hospital, Yonsei University College of Medicine, Seoul, South Korea
| | - Sahil Thakur
- Singapore Eye Research Institute, Singapore National Eye Centre, Singapore, Singapore
| | - Zhi Da Soh
- Singapore Eye Research Institute, Singapore National Eye Centre, Singapore, Singapore
- Department of Ophthalmology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Yunnie Cho
- Mediwhale Inc, Seoul, Republic of Korea
- Department of Education and Human Resource Development, Seoul National University Hospital, Seoul, South Korea
| | - Qingsheng Peng
- Singapore Eye Research Institute, Singapore National Eye Centre, Singapore, Singapore
| | - Kwanghyun Lee
- Department of Ophthalmology, National Health Insurance Service Ilsan Hospital, Goyang, Republic of Korea
| | - Yih-Chung Tham
- Singapore Eye Research Institute, Singapore National Eye Centre, Singapore, Singapore
- Department of Ophthalmology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Centre for Innovation and Precision Eye Health, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Ophthalmology and Visual Sciences Academic Clinical Program, Singapore, Singapore
| | - Tyler Hyungtaek Rim
- Singapore Eye Research Institute, Singapore National Eye Centre, Singapore, Singapore.
- Mediwhale Inc, Seoul, Republic of Korea.
| | - Ching-Yu Cheng
- Singapore Eye Research Institute, Singapore National Eye Centre, Singapore, Singapore
- Department of Ophthalmology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Centre for Innovation and Precision Eye Health, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Ophthalmology and Visual Sciences Academic Clinical Program, Singapore, Singapore
| |
Collapse
|
16
|
Wang CY, Sadrieh FK, Shen YT, Chen SE, Kim S, Chen V, Raghavendra A, Wang D, Saeedi O, Tao Y. MEMO: dataset and methods for robust multimodal retinal image registration with large or small vessel density differences. BIOMEDICAL OPTICS EXPRESS 2024; 15:3457-3479. [PMID: 38855695 PMCID: PMC11161385 DOI: 10.1364/boe.516481] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Revised: 03/20/2024] [Accepted: 04/18/2024] [Indexed: 06/11/2024]
Abstract
The measurement of retinal blood flow (RBF) in capillaries can provide a powerful biomarker for the early diagnosis and treatment of ocular diseases. However, no single modality can determine capillary flowrates with high precision. Combining erythrocyte-mediated angiography (EMA) with optical coherence tomography angiography (OCTA) has the potential to achieve this goal, as EMA can measure the absolute RBF of retinal microvasculature and OCTA can provide the structural images of capillaries. However, multimodal retinal image registration between these two modalities remains largely unexplored. To fill this gap, we establish MEMO, the first public multimodal EMA and OCTA retinal image dataset. A unique challenge in multimodal retinal image registration between these modalities is the relatively large difference in vessel density (VD). To address this challenge, we propose a segmentation-based deep-learning framework (VDD-Reg), which provides robust results despite differences in vessel density. VDD-Reg consists of a vessel segmentation module and a registration module. To train the vessel segmentation module, we further designed a two-stage semi-supervised learning framework (LVD-Seg) combining supervised and unsupervised losses. We demonstrate that VDD-Reg outperforms existing methods quantitatively and qualitatively for cases of both small VD differences (using the CF-FA dataset) and large VD differences (using our MEMO dataset). Moreover, VDD-Reg requires as few as three annotated vessel segmentation masks to maintain its accuracy, demonstrating its feasibility.
Collapse
Affiliation(s)
- Chiao-Yi Wang
- Department of Bioengineering, University of Maryland, College Park, MD 20742, USA
| | | | - Yi-Ting Shen
- Department of Electrical and Computer Engineering, University of Maryland, College Park, MD 20742, USA
| | - Shih-En Chen
- Department of Ophthalmology and Visual Sciences, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Sarah Kim
- Department of Ophthalmology and Visual Sciences, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Victoria Chen
- Department of Ophthalmology and Visual Sciences, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Achyut Raghavendra
- Department of Ophthalmology and Visual Sciences, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Dongyi Wang
- Department of Biological and Agricultural Engineering, University of Arkansas, Fayetteville, AR 72701, USA
| | - Osamah Saeedi
- Department of Ophthalmology and Visual Sciences, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Yang Tao
- Department of Bioengineering, University of Maryland, College Park, MD 20742, USA
| |
Collapse
|
17
|
Ho K, Bodi NE, Sharma TP. Normal-Tension Glaucoma and Potential Clinical Links to Alzheimer's Disease. J Clin Med 2024; 13:1948. [PMID: 38610712 PMCID: PMC11012506 DOI: 10.3390/jcm13071948] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2024] [Revised: 03/21/2024] [Accepted: 03/25/2024] [Indexed: 04/14/2024] Open
Abstract
Glaucoma is a group of optic neuropathies and the world's leading cause of irreversible blindness. Normal-tension glaucoma (NTG) is a subtype of glaucoma that is characterized by a typical pattern of peripheral retinal loss, in which the patient's intraocular pressure (IOP) is considered within the normal range (<21 mmHg). Currently, the only targetable risk factor for glaucoma is lowering IOP, and patients with NTG continue to experience visual field loss after IOP-lowering treatments. This demonstrates the need for a better understanding of the pathogenesis of NTG and underlying mechanisms leading to neurodegeneration. Recent studies have found significant connections between NTG and cerebral manifestations, suggesting NTG as a neurodegenerative disease beyond the eye. Gaining a better understanding of NTG can potentially provide new Alzheimer's Disease diagnostics capabilities. This review identifies the epidemiology, current biomarkers, altered fluid dynamics, and cerebral and ocular manifestations to examine connections and discrepancies between the mechanisms of NTG and Alzheimer's Disease.
Collapse
Affiliation(s)
- Kathleen Ho
- Eugene and Marilyn Glick Eye Institute, Department of Ophthalmology, Indiana University School of Medicine, Indianapolis, IN 46202, USA;
| | - Nicole E. Bodi
- Pharmacology and Toxicology, Indiana University School of Medicine, Indianapolis, IN 46202, USA;
| | - Tasneem P. Sharma
- Eugene and Marilyn Glick Eye Institute, Department of Ophthalmology, Indiana University School of Medicine, Indianapolis, IN 46202, USA;
- Pharmacology and Toxicology, Indiana University School of Medicine, Indianapolis, IN 46202, USA;
- Stark Neurosciences Research Institute, Indianapolis, IN 46202, USA
| |
Collapse
|
18
|
Dumitrascu OM, Doustar J, Fuchs DT, Koronyo Y, Sherman DS, Miller MS, Johnson KO, Carare RO, Verdooner SR, Lyden PD, Schneider JA, Black KL, Koronyo-Hamaoui M. Distinctive retinal peri-arteriolar versus peri-venular amyloid plaque distribution correlates with the cognitive performance. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.02.27.580733. [PMID: 38464292 PMCID: PMC10925252 DOI: 10.1101/2024.02.27.580733] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/12/2024]
Abstract
Introduction The vascular contribution to Alzheimer's disease (AD) is tightly connected to cognitive performance across the AD continuum. We topographically describe retinal perivascular amyloid plaque (AP) burden in subjects with normal or impaired cognition. Methods Using scanning laser ophthalmoscopy, we quantified retinal peri-arteriolar and peri-venular curcumin-positive APs in the first, secondary and tertiary branches in twenty-eight subjects. Perivascular AP burden among cognitive states was correlated with neuroimaging and cognitive measures. Results Peri-arteriolar exceeded peri-venular AP count (p<0.0001). Secondary branch AP count was significantly higher in cognitively impaired (p<0.01). Secondary small and tertiary peri-venular AP count strongly correlated with clinical dementia rating, hippocampal volumes, and white matter hyperintensity count. Discussion Our topographic analysis indicates greater retinal amyloid accumulation in the retinal peri-arteriolar regions overall, and distal peri-venular regions in cognitively impaired individuals. Larger longitudinal studies are warranted to understand the temporal-spatial relationship between vascular dysfunction and perivascular amyloid deposition in AD. Highlights Retinal peri-arteriolar region exhibits more amyloid compared with peri-venular regions.Secondary retinal vascular branches have significantly higher perivascular amyloid burden in subjects with impaired cognition, consistent across sexes.Cognitively impaired individuals have significantly greater retinal peri-venular amyloid deposits in the distal small branches, that correlate with CDR and hippocampal volumes.
Collapse
|
19
|
Kelly L, Brown C, Michalik D, Hawkes CA, Aldea R, Agarwal N, Salib R, Alzetani A, Ethell DW, Counts SE, de Leon M, Fossati S, Koronyo‐Hamaoui M, Piazza F, Rich SA, Wolters FJ, Snyder H, Ismail O, Elahi F, Proulx ST, Verma A, Wunderlich H, Haack M, Dodart JC, Mazer N, Carare RO. Clearance of interstitial fluid (ISF) and CSF (CLIC) group-part of Vascular Professional Interest Area (PIA), updates in 2022-2023. Cerebrovascular disease and the failure of elimination of Amyloid-β from the brain and retina with age and Alzheimer's disease: Opportunities for therapy. Alzheimers Dement 2024; 20:1421-1435. [PMID: 37897797 PMCID: PMC10917045 DOI: 10.1002/alz.13512] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Revised: 08/30/2023] [Accepted: 08/30/2023] [Indexed: 10/30/2023]
Abstract
This editorial summarizes advances from the Clearance of Interstitial Fluid and Cerebrospinal Fluid (CLIC) group, within the Vascular Professional Interest Area (PIA) of the Alzheimer's Association International Society to Advance Alzheimer's Research and Treatment (ISTAART). The overarching objectives of the CLIC group are to: (1) understand the age-related physiology changes that underlie impaired clearance of interstitial fluid (ISF) and cerebrospinal fluid (CSF) (CLIC); (2) understand the cellular and molecular mechanisms underlying intramural periarterial drainage (IPAD) in the brain; (3) establish novel diagnostic tests for Alzheimer's disease (AD), cerebral amyloid angiopathy (CAA), retinal amyloid vasculopathy, amyloid-related imaging abnormalities (ARIA) of spontaneous and iatrogenic CAA-related inflammation (CAA-ri), and vasomotion; and (4) establish novel therapies that facilitate IPAD to eliminate amyloid β (Aβ) from the aging brain and retina, to prevent or reduce AD and CAA pathology and ARIA side events associated with AD immunotherapy.
Collapse
Affiliation(s)
- Louise Kelly
- Faculty of MedicineUniversity of SouthamptonSouthamptonHampshireUK
| | | | - Daniel Michalik
- Faculty of MedicineUniversity of SouthamptonSouthamptonHampshireUK
| | | | - Roxana Aldea
- Roche Pharma Research & Early DevelopmentRoche Innovation Center BaselBaselSwitzerland
| | - Nivedita Agarwal
- Neuroradiology sectionScientific Institute IRCCS Eugenio MedeaBosisio Parini, LCItaly
| | - Rami Salib
- Faculty of MedicineUniversity of SouthamptonSouthamptonHampshireUK
| | - Aiman Alzetani
- Faculty of MedicineUniversity of SouthamptonSouthamptonHampshireUK
| | | | - Scott E. Counts
- Dept. Translational NeuroscienceDept. Family MedicineMichigan State UniversityGrand RapidsMichiganUSA
| | - Mony de Leon
- Brain Health Imaging InstituteDepartment of RadiologyWeill Cornell MedicineNew YorkNew YorkUSA
| | | | - Maya Koronyo‐Hamaoui
- Departments of NeurosurgeryNeurology, and Biomedical SciencesMaxine Dunitz Neurosurgical Research InstituteCedars‐Sinai Medical CenterLos AngelesCaliforniaUSA
| | | | | | | | - Heather Snyder
- Alzheimer's AssociationMedical & Scientific RelationsChicagoIllinoisUSA
| | - Ozama Ismail
- Icahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
| | - Fanny Elahi
- Icahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
| | | | - Ajay Verma
- Formation Venture Engineering FoundryTopsfieldMassachusettsUSA
| | | | | | | | | | - Roxana O. Carare
- Faculty of MedicineUniversity of SouthamptonSouthamptonHampshireUK
| |
Collapse
|
20
|
Robbins CB, Rathinavelu J, Ma JP, Soundararajan S, Stinnett SS, Liu AJ, Johnson KG, Grewal DS, Fekrat S. Peripapillary Optical Coherence Tomography Angiography in Alzheimer's Disease, Mild Cognitive Impairment, and Normal Cognition. Ophthalmic Surg Lasers Imaging Retina 2024; 55:78-84. [PMID: 38346150 DOI: 10.3928/23258160-20240107-01] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/15/2024]
Abstract
OBJECTIVE This study aimed to identify peripapillary microvascular changes in Alzheimer's disease (AD) and mild cognitive impairment (MCI). PATIENTS AND METHODS In this prospective study, 66 eyes of 36 subjects with AD, 119 eyes of 63 with MCI, and 513 eyes of 265 controls with normal cognition were enrolled. Peripapillary capillary perfusion density (CPD), capillary flux index (CFI), and retinal nerve fiber layer (RNFL) thickness were determined. RESULTS Average CPD differed significantly between all three groups (P = 0.001), being significantly greater in AD vs controls (0.446 ± 0.015 vs 0.439 ± 0.017, P = 0.001) and MCI vs controls (0.443 ± 0.020 vs 0.439 ± 0.017, P = 0.007) but not AD vs MCI (P = 0.69). CFI and average RNFL thickness did not significantly differ among groups (all P > 0.05). CONCLUSION Peripapillary CPD is increased in eyes with AD or MCI compared to controls despite similar RNFL thickness. [Ophthalmic Surg Lasers Imaging Retina 2024;55:78-84.].
Collapse
|
21
|
Poudel P, Frost SM, Eslick S, Sohrabi HR, Taddei K, Martins RN, Hone E. Hyperspectral Retinal Imaging as a Non-Invasive Marker to Determine Brain Amyloid Status. J Alzheimers Dis 2024; 100:S131-S152. [PMID: 39121128 DOI: 10.3233/jad-240631] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/11/2024]
Abstract
Background As an extension of the central nervous system (CNS), the retina shares many similarities with the brain and can manifest signs of various neurological diseases, including Alzheimer's disease (AD). Objective To investigate the retinal spectral features and develop a classification model to differentiate individuals with different brain amyloid levels. Methods Sixty-six participants with varying brain amyloid-β protein levels were non-invasively imaged using a hyperspectral retinal camera in the wavelength range of 450-900 nm in 5 nm steps. Multiple retina features from the central and superior views were selected and analyzed to identify their variability among individuals with different brain amyloid loads. Results The retinal reflectance spectra in the 450-585 nm wavelengths exhibited a significant difference in individuals with increasing brain amyloid. The retinal features in the superior view showed higher inter-subject variability. A classification model was trained to differentiate individuals with varying amyloid levels using the spectra of extracted retinal features. The performance of the spectral classification model was dependent upon retinal features and showed 0.758-0.879 accuracy, 0.718-0.909 sensitivity, 0.764-0.912 specificity, and 0.745-0.891 area under curve for the right eye. Conclusions This study highlights the spectral variation of retinal features associated with brain amyloid loads. It also demonstrates the feasibility of the retinal hyperspectral imaging technique as a potential method to identify individuals in the preclinical phase of AD as an inexpensive alternative to brain imaging.
Collapse
Affiliation(s)
- Purna Poudel
- Alzheimer's Research Australia, Ralph and Patricia Sarich Neuroscience Research Institute, Nedlands, WA, Australia
- Centre of Excellence for Alzheimer's Disease Research and Care, School of Medical and Health Sciences, Edith Cowan University, Joondalup, WA, Australia
| | - Shaun M Frost
- Commonwealth Scientific and Industrial Research Organisation (CSIRO), Kensington, WA, Australia
- Australian e-Health Research Centre, Floreat, WA, Australia
| | - Shaun Eslick
- Lifespan Health and Wellbeing Research Centre, Macquarie Medical School, Macquarie University, Macquarie Park, NSW, Australia
| | - Hamid R Sohrabi
- Alzheimer's Research Australia, Ralph and Patricia Sarich Neuroscience Research Institute, Nedlands, WA, Australia
- Centre for Healthy Ageing, Health Futures Institute, Murdoch University, Perth, WA, Australia
| | - Kevin Taddei
- Alzheimer's Research Australia, Ralph and Patricia Sarich Neuroscience Research Institute, Nedlands, WA, Australia
- Centre of Excellence for Alzheimer's Disease Research and Care, School of Medical and Health Sciences, Edith Cowan University, Joondalup, WA, Australia
- Lions Alzheimer's Foundation, Perth, WA, Australia
| | - Ralph N Martins
- Alzheimer's Research Australia, Ralph and Patricia Sarich Neuroscience Research Institute, Nedlands, WA, Australia
- Centre of Excellence for Alzheimer's Disease Research and Care, School of Medical and Health Sciences, Edith Cowan University, Joondalup, WA, Australia
- Lifespan Health and Wellbeing Research Centre, Macquarie Medical School, Macquarie University, Macquarie Park, NSW, Australia
- Lions Alzheimer's Foundation, Perth, WA, Australia
| | - Eugene Hone
- Alzheimer's Research Australia, Ralph and Patricia Sarich Neuroscience Research Institute, Nedlands, WA, Australia
- Centre of Excellence for Alzheimer's Disease Research and Care, School of Medical and Health Sciences, Edith Cowan University, Joondalup, WA, Australia
- Lions Alzheimer's Foundation, Perth, WA, Australia
| |
Collapse
|
22
|
Lee CS, Ferguson AN, Gibbons LE, Walker R, Su YR, Krakauer C, Brush M, Kam J, Larson EB, Arterburn DE, Crane PK. Eye Adult Changes in Thought (Eye ACT) Study: Design and Report on the Inaugural Cohort. J Alzheimers Dis 2024; 100:309-320. [PMID: 38875039 PMCID: PMC11556780 DOI: 10.3233/jad-240203] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/16/2024]
Abstract
Background Conflicting research on retinal biomarkers of Alzheimer's disease and related dementias (AD/ADRD) is likely related to limited sample sizes, study design, and protocol differences. Objective The prospective Eye Adult Changes in Thought (Eye ACT) seeks to address these gaps. Methods Eye ACT participants are recruited from ACT, an ongoing cohort of dementia-free, older adults followed biennially until AD/ADRD, and undergo visual function and retinal imaging assessment either in clinic or at home. Results 330 participants were recruited as of 03/2023. Compared to ACT participants not in Eye ACT (N = 1868), Eye ACT participants (N = 330) are younger (mean age: 70.3 versus 71.2, p = 0.014), newer to ACT (median ACT visits since baseline: 3 versus 4, p < 0.001), have more years of education (17.7 versus 16.2, p < 0.001) and had lower rates of visual impairment (12% versus 22%, p < 0.001). Compared to those seen in clinic (N = 300), Eye ACT participants seen at home (N = 30) are older (77.2 versus 74.9, p = 0.015), more frequently female (60% versus 49%, p = 0.026), and have significantly worse visual acuity (71.1 versus 78.9 Early Treatment Diabetic Retinopathy Study letters, p < 0.001) and contrast sensitivity (-1.9 versus -2.1 mean log units at 3 cycles per degree, p = 0.002). Cognitive scores and retinal imaging measurements are similar between the two groups. Conclusions Participants assessed at home had significantly worse visual function than those seen in clinic. By including these participants, Eye ACT provides a unique longitudinal cohort for evaluating potential retinal biomarkers of dementia.
Collapse
Affiliation(s)
- Cecilia S Lee
- Department of Ophthalmology, University of Washington, Seattle, Washington, United States
- The Roger and Angie Karalis Johnson Retina Center, Seattle, Washington
| | - Alina N Ferguson
- Department of Ophthalmology, University of Washington, Seattle, Washington, United States
- The Roger and Angie Karalis Johnson Retina Center, Seattle, Washington
- University of Washington School of Medicine, Seattle, Washington
| | - Laura E Gibbons
- Department of Medicine, University of Washington, Seattle, Washington
| | - Rod Walker
- Kaiser Permanente Washington Health Research Institute, Seattle, Washington
| | - Yu-Ru Su
- Kaiser Permanente Washington Health Research Institute, Seattle, Washington
| | - Chloe Krakauer
- Kaiser Permanente Washington Health Research Institute, Seattle, Washington
| | | | - Jason Kam
- Kaiser Permanente Washington, Seattle, Washington
| | - Eric B Larson
- Department of Medicine, University of Washington, Seattle, Washington
| | - David E Arterburn
- Kaiser Permanente Washington Health Research Institute, Seattle, Washington
| | - Paul K Crane
- Department of Medicine, University of Washington, Seattle, Washington
| |
Collapse
|
23
|
Donato L, Mordà D, Scimone C, Alibrandi S, D’Angelo R, Sidoti A. Bridging Retinal and Cerebral Neurodegeneration: A Focus on Crosslinks between Alzheimer-Perusini's Disease and Retinal Dystrophies. Biomedicines 2023; 11:3258. [PMID: 38137479 PMCID: PMC10741418 DOI: 10.3390/biomedicines11123258] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Revised: 12/02/2023] [Accepted: 12/07/2023] [Indexed: 12/24/2023] Open
Abstract
In the early stages of Alzheimer-Perusini's disease (AD), individuals often experience vision-related issues such as color vision impairment, reduced contrast sensitivity, and visual acuity problems. As the disease progresses, there is a connection with glaucoma and age-related macular degeneration (AMD) leading to retinal cell death. The retina's involvement suggests a link with the hippocampus, where most AD forms start. A thinning of the retinal nerve fiber layer (RNFL) due to the loss of retinal ganglion cells (RGCs) is seen as a potential AD diagnostic marker using electroretinography (ERG) and optical coherence tomography (OCT). Amyloid beta fragments (Aβ), found in the eye's vitreous and aqueous humor, are also present in the cerebrospinal fluid (CSF) and accumulate in the retina. Aβ is known to cause tau hyperphosphorylation, leading to its buildup in various retinal layers. However, diseases like AD are now seen as mixed proteinopathies, with deposits of the prion protein (PrP) and α-synuclein found in affected brains and retinas. Glial cells, especially microglial cells, play a crucial role in these diseases, maintaining immunoproteostasis. Studies have shown similarities between retinal and brain microglia in terms of transcription factor expression and morphotypes. All these findings constitute a good start to achieving better comprehension of neurodegeneration in both the eye and the brain. New insights will be able to bring the scientific community closer to specific disease-modifying therapies.
Collapse
Affiliation(s)
- Luigi Donato
- Department of Biomedical and Dental Sciences and Morphofunctional Imaging, Division of Medical Biotechnologies and Preventive Medicine, University of Messina, 98122 Messina, Italy; (L.D.); (C.S.); (R.D.); (A.S.)
- Department of Biomolecular Strategies, Genetics, Cutting-Edge Therapies, Euro-Mediterranean Institute of Science and Technology (I.E.ME.S.T.), 90139 Palermo, Italy;
| | - Domenico Mordà
- Department of Biomolecular Strategies, Genetics, Cutting-Edge Therapies, Euro-Mediterranean Institute of Science and Technology (I.E.ME.S.T.), 90139 Palermo, Italy;
- Department of Veterinary Sciences, University of Messina, 98122 Messina, Italy
| | - Concetta Scimone
- Department of Biomedical and Dental Sciences and Morphofunctional Imaging, Division of Medical Biotechnologies and Preventive Medicine, University of Messina, 98122 Messina, Italy; (L.D.); (C.S.); (R.D.); (A.S.)
- Department of Biomolecular Strategies, Genetics, Cutting-Edge Therapies, Euro-Mediterranean Institute of Science and Technology (I.E.ME.S.T.), 90139 Palermo, Italy;
| | - Simona Alibrandi
- Department of Biomedical and Dental Sciences and Morphofunctional Imaging, Division of Medical Biotechnologies and Preventive Medicine, University of Messina, 98122 Messina, Italy; (L.D.); (C.S.); (R.D.); (A.S.)
- Department of Biomolecular Strategies, Genetics, Cutting-Edge Therapies, Euro-Mediterranean Institute of Science and Technology (I.E.ME.S.T.), 90139 Palermo, Italy;
| | - Rosalia D’Angelo
- Department of Biomedical and Dental Sciences and Morphofunctional Imaging, Division of Medical Biotechnologies and Preventive Medicine, University of Messina, 98122 Messina, Italy; (L.D.); (C.S.); (R.D.); (A.S.)
| | - Antonina Sidoti
- Department of Biomedical and Dental Sciences and Morphofunctional Imaging, Division of Medical Biotechnologies and Preventive Medicine, University of Messina, 98122 Messina, Italy; (L.D.); (C.S.); (R.D.); (A.S.)
| |
Collapse
|
24
|
Suh A, Ong J, Kamran SA, Waisberg E, Paladugu P, Zaman N, Sarker P, Tavakkoli A, Lee AG. Retina Oculomics in Neurodegenerative Disease. Ann Biomed Eng 2023; 51:2708-2721. [PMID: 37855949 DOI: 10.1007/s10439-023-03365-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Accepted: 09/05/2023] [Indexed: 10/20/2023]
Abstract
Ophthalmic biomarkers have long played a critical role in diagnosing and managing ocular diseases. Oculomics has emerged as a field that utilizes ocular imaging biomarkers to provide insights into systemic diseases. Advances in diagnostic and imaging technologies including electroretinography, optical coherence tomography (OCT), confocal scanning laser ophthalmoscopy, fluorescence lifetime imaging ophthalmoscopy, and OCT angiography have revolutionized the ability to understand systemic diseases and even detect them earlier than clinical manifestations for earlier intervention. With the advent of increasingly large ophthalmic imaging datasets, machine learning models can be integrated into these ocular imaging biomarkers to provide further insights and prognostic predictions of neurodegenerative disease. In this manuscript, we review the use of ophthalmic imaging to provide insights into neurodegenerative diseases including Alzheimer Disease, Parkinson Disease, Amyotrophic Lateral Sclerosis, and Huntington Disease. We discuss recent advances in ophthalmic technology including eye-tracking technology and integration of artificial intelligence techniques to further provide insights into these neurodegenerative diseases. Ultimately, oculomics opens the opportunity to detect and monitor systemic diseases at a higher acuity. Thus, earlier detection of systemic diseases may allow for timely intervention for improving the quality of life in patients with neurodegenerative disease.
Collapse
Affiliation(s)
- Alex Suh
- Tulane University School of Medicine, New Orleans, LA, USA.
| | - Joshua Ong
- Michigan Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Sharif Amit Kamran
- Human-Machine Perception Laboratory, Department of Computer Science and Engineering, University of Nevada, Reno, Reno, NV, USA
| | - Ethan Waisberg
- University College Dublin School of Medicine, Belfield, Dublin, Ireland
| | - Phani Paladugu
- Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA, USA
| | - Nasif Zaman
- Human-Machine Perception Laboratory, Department of Computer Science and Engineering, University of Nevada, Reno, Reno, NV, USA
| | - Prithul Sarker
- Human-Machine Perception Laboratory, Department of Computer Science and Engineering, University of Nevada, Reno, Reno, NV, USA
| | - Alireza Tavakkoli
- Human-Machine Perception Laboratory, Department of Computer Science and Engineering, University of Nevada, Reno, Reno, NV, USA
| | - Andrew G Lee
- Center for Space Medicine, Baylor College of Medicine, Houston, TX, USA
- Department of Ophthalmology, Blanton Eye Institute, Houston Methodist Hospital, 6560 Fannin St #450, Houston, TX, 77030, USA
- The Houston Methodist Research Institute, Houston Methodist Hospital, Houston, TX, USA
- Departments of Ophthalmology, Neurology and Neurosurgery, Weill Cornell Medicine, New York, NY, USA
- Department of Ophthalmology, University of Texas Medical Branch, Galveston, TX, USA
- University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Texas A&M College of Medicine, Bryan, TX, USA
- Department of Ophthalmology, The University of Iowa Hospitals and Clinics, Iowa City, IA, USA
| |
Collapse
|
25
|
Bernardin F, Gauld C, Martin VP, Laprévote V, Dondé C. The 68 symptoms of the clinical high risk for psychosis: Low similarity among fourteen screening questionnaires. Psychiatry Res 2023; 330:115592. [PMID: 37948888 DOI: 10.1016/j.psychres.2023.115592] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Revised: 10/28/2023] [Accepted: 11/04/2023] [Indexed: 11/12/2023]
Abstract
The Clinical High Risk for psychosis (CHR) is a heterogeneous condition with multiple symptoms. CHR screening is challenging in routine care, as a wide variety of questionnaires exists. We propose to explore the extent to which these questionnaires differ or overlap in item content. We performed a systematic and quantitative analysis of item content in a set of widely-used CHR screening questionnaires. Items were extracted from questionnaires and reworded according to the Structured Interview for Psychosis-Risk Syndromes (SIPS). Then, symptoms were generated from individual items. The Jaccard Index was calculated to assess content overlap. The 14 analysed questionnaires were composed of 347 items, from which 198 symptoms were generated and, in turn, collapsed into 68 distinct symptoms. Positive symptoms were the most commonly represented. The overall overlap across questionnaires showed weak similarity (Jaccard = 0.19±0.50). CHR screening questionnaires might evaluate the same broad clinical construct, but have different scopes within that construct, and may be more or less comprehensive than one another. Clinicians and researchers should be mindful of the specific features of each instrument for optimal CHR screening.
Collapse
Affiliation(s)
- Florent Bernardin
- Centre Psychothérapique de Nancy, Pôle Transversal Médico-Technique et Clinique, Centre de, Liaison et d'Intervention Précoce, Laxou F-54520, France; Institut National de la Santé et de la Recherche Médical, INSERM U1114, Pôle de Psychiatrie, Fédération de Médecine Translationnelle de Strasbourg, Centre Hospitalier Régional Universitaire, de Strasbourg, Université de Strasbourg, Strasbourg, France.
| | - Christophe Gauld
- Department of Child Psychiatry, Université de Lyon, 59 Bd Pinel, Lyon F-69000, France; Université Claude Bernard Lyon 1, CNRS, INSERM, Centre de Recherche en Neurosciences de, Lyon CRNL U1028 UMR5292, PSYR2, Bron F-69500, France
| | - Vincent P Martin
- Univ. Bordeaux, CNRS, Bordeaux INP, LaBRI, UMR 5800, Talence F-33400, France; Univ. Bordeaux, CNRS, SANPSY, UMR 6033, Bordeaux F-33000, France
| | - Vincent Laprévote
- Centre Psychothérapique de Nancy, Pôle Transversal Médico-Technique et Clinique, Centre de, Liaison et d'Intervention Précoce, Laxou F-54520, France; Institut National de la Santé et de la Recherche Médical, INSERM U1114, Pôle de Psychiatrie, Fédération de Médecine Translationnelle de Strasbourg, Centre Hospitalier Régional Universitaire, de Strasbourg, Université de Strasbourg, Strasbourg, France; Université de Lorraine, Faculté de Médecine, Vandœuvre-lès-Nancy F-54500 France
| | - Clément Dondé
- Univ. Grenoble Alpes, Grenoble F-38000, France; INSERM, U1216, Grenoble F-38000, France; Psychiatry Department, CHU Grenoble Alpes, Grenoble F-38000, France.
| |
Collapse
|
26
|
Li C, Zhu X, Yang K, Ju Y, Shi K, Xiao Y, Su B, Lu F, Cui L, Li M. Relationship of retinal capillary plexus and ganglion cell complex with mild cognitive impairment and dementia. Eye (Lond) 2023; 37:3743-3750. [PMID: 37270614 PMCID: PMC10698172 DOI: 10.1038/s41433-023-02592-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Revised: 05/08/2023] [Accepted: 05/17/2023] [Indexed: 06/05/2023] Open
Abstract
OBJECTIVE To investigate relationship of the retinal capillary plexus (RCP) and ganglion cell complex (GCC) with mild cognitive impairment (MCI) and dementia in a community-based study1. METHODS This cross-sectional study incorporated the participants of the Jidong Eye Cohort Study. Optical coherence tomography angiography was performed to obtain RCP vessel density and GCC thickness with detailed segments. The Mini-mental State Examination and Montreal Cognitive Assessment were used to assess cognitive status by professional neuropsychologists. Participants were thus divided into three groups: normal, mild cognitive impairment, and dementia. Multivariable analysis was used to measure relationship of ocular parameters with cognitive impairment. RESULTS Of the 2678 participants, the mean age was 44.1 ± 11.7 years. MCI and dementia occurred in 197 (7.4%) and 80 (3%) participants, respectively. Compared to the normal group, the adjusted odds ratio (OR) with the 95% confidence interval was 0.76 (0.65-0.90) for the correlation of lower deep RCP with MCI. We found the following items significantly associated with dementia compared with the normal group: a superficial (OR, 0.68 [0.54-0.86]) and deep (OR, 0.75 [0.57-0.99]) RCP, as well as the GCC (OR, 0.68 [0.54-0.85]). Compared to the MCI group, those with dementia had decreased GCC (OR, 0.75 [0.58-0.97]). CONCLUSIONS Decreased deep RCP density was associated with MCI. Decreased superficial and deep RCP and the thin GCC were correlated with dementia. These implied that the retinal microvasculature may develop into a promising non-invasive imaging marker to predict severity of cognitive impairment.
Collapse
Affiliation(s)
- Chunmei Li
- National Clinical Research Center for Ocular Diseases, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China
- National Engineering Research Center of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China
| | - Xiaoxuan Zhu
- National Clinical Research Center for Ocular Diseases, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China
- National Engineering Research Center of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China
| | - Kai Yang
- National Clinical Research Center for Ocular Diseases, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China
- National Engineering Research Center of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China
| | - Ying Ju
- National Clinical Research Center for Ocular Diseases, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China
- National Engineering Research Center of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China
| | - Keai Shi
- National Clinical Research Center for Ocular Diseases, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China
- National Engineering Research Center of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China
| | - Yunfan Xiao
- National Clinical Research Center for Ocular Diseases, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China
- National Engineering Research Center of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China
| | - Binbin Su
- National Clinical Research Center for Ocular Diseases, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China
- National Engineering Research Center of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China
| | - Fan Lu
- National Clinical Research Center for Ocular Diseases, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China.
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China.
- National Engineering Research Center of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China.
| | - Lele Cui
- National Clinical Research Center for Ocular Diseases, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China.
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China.
- National Engineering Research Center of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China.
| | - Ming Li
- National Clinical Research Center for Ocular Diseases, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China.
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China.
- National Engineering Research Center of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China.
| |
Collapse
|
27
|
Santos-Ortega Á, Alba-Linero C, Urbinati F, Rocha-de-Lossada C, Orti R, Reyes-Bueno JA, Garzón-Maldonado FJ, Serrano V, de Rojas-Leal C, de la Cruz-Cosme C, España-Contreras M, Rodríguez-Calvo-de-Mora M, García-Casares N. Structural and Functional Retinal Changes in Patients with Mild Cognitive Impairment with and without Diabetes. J Clin Med 2023; 12:7035. [PMID: 38002648 PMCID: PMC10672424 DOI: 10.3390/jcm12227035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2023] [Revised: 11/01/2023] [Accepted: 11/07/2023] [Indexed: 11/26/2023] Open
Abstract
Our objective is to analyze retinal changes using optical coherence tomography angiography (OCT-A) in patients with mild cognitive impairment (MCI) to characterize structural and vascular alterations. This cross-sectional study involved 117 eyes: 39 eyes from patients with MCI plus diabetes (DM-MCI), 39 eyes from patients with MCI but no diabetes (MCI); and 39 healthy control eyes (C). All patients underwent a visual acuity measurement, a structural OCT, an OCT-A, and a neuropsychological examination. Our study showed a thinning of retinal nerve fiber layer thickness (RNFL) and a decrease in macular thickness when comparing the MCI-DM group to the C group (p = 0.008 and p = 0.016, respectively). In addition, an increase in arteriolar thickness (p = 0.016), a reduction in superficial capillary plexus density (p = 0.002), and a decrease in ganglion cell thickness (p = 0.027) were found when comparing the MCI-DM group with the MCI group. Diabetes may exacerbate retinal vascular changes when combined with mild cognitive impairment.
Collapse
Affiliation(s)
| | - Carmen Alba-Linero
- Department of Ophthalmology, Hospital Universitario Virgen de la Victoria, 29010 Malaga, Spain
- Department of Ophthalmology, Faculty of Medicine, University of Malaga, 29016 Malaga, Spain;
| | - Facundo Urbinati
- Department of Ophthalmology, Hospital Regional Universitario, 29011 Malaga, Spain; (F.U.); (C.R.-d.-L.); (M.E.-C.); (M.R.-C.-d.-M.)
| | - Carlos Rocha-de-Lossada
- Department of Ophthalmology, Hospital Regional Universitario, 29011 Malaga, Spain; (F.U.); (C.R.-d.-L.); (M.E.-C.); (M.R.-C.-d.-M.)
- Qvision, Opththalmology Department, VITHAS Almería Hospital, 04120 Almería, Spain
- Ophthalmology Department, VITHAS Málaga, 29016 Malaga, Spain
- Department of Surgery, Faculty of Medicine, Ophthalmology Area Doctor Fedriani, University of Sevilla, 41004 Sevilla, Spain
| | - Rafael Orti
- Department of Ophthalmology, Faculty of Medicine, University of Malaga, 29016 Malaga, Spain;
| | | | - Francisco Javier Garzón-Maldonado
- Department of Neurology, Hospital Virgen de la Victoria, 29010 Malaga, Spain; (F.J.G.-M.); (V.S.); (C.d.R.-L.); (C.d.l.C.-C.)
- Instituto de Investigación Biomédica de Málaga (IBIMA), 29010 Malaga, Spain;
| | - Vicente Serrano
- Department of Neurology, Hospital Virgen de la Victoria, 29010 Malaga, Spain; (F.J.G.-M.); (V.S.); (C.d.R.-L.); (C.d.l.C.-C.)
| | - Carmen de Rojas-Leal
- Department of Neurology, Hospital Virgen de la Victoria, 29010 Malaga, Spain; (F.J.G.-M.); (V.S.); (C.d.R.-L.); (C.d.l.C.-C.)
- Instituto de Investigación Biomédica de Málaga (IBIMA), 29010 Malaga, Spain;
| | - Carlos de la Cruz-Cosme
- Department of Neurology, Hospital Virgen de la Victoria, 29010 Malaga, Spain; (F.J.G.-M.); (V.S.); (C.d.R.-L.); (C.d.l.C.-C.)
- Instituto de Investigación Biomédica de Málaga (IBIMA), 29010 Malaga, Spain;
| | - Manuela España-Contreras
- Department of Ophthalmology, Hospital Regional Universitario, 29011 Malaga, Spain; (F.U.); (C.R.-d.-L.); (M.E.-C.); (M.R.-C.-d.-M.)
| | - Marina Rodríguez-Calvo-de-Mora
- Department of Ophthalmology, Hospital Regional Universitario, 29011 Malaga, Spain; (F.U.); (C.R.-d.-L.); (M.E.-C.); (M.R.-C.-d.-M.)
- Qvision, Opththalmology Department, VITHAS Almería Hospital, 04120 Almería, Spain
- Ophthalmology Department, VITHAS Málaga, 29016 Malaga, Spain
| | - Natalia García-Casares
- Instituto de Investigación Biomédica de Málaga (IBIMA), 29010 Malaga, Spain;
- Department of Medicine, Faculty of Medicine, University of Malaga, 29016 Malaga, Spain
- Centro de Investigaciones Médico-Sanitarias (CIMES), University of Malaga, 29016 Malaga, Spain
| |
Collapse
|
28
|
Sugasini D, Park JC, McAnany JJ, Kim TH, Ma G, Yao X, Antharavally B, Oroskar A, Oroskar AA, Layden BT, Subbaiah PV. Improvement of retinal function in Alzheimer disease-associated retinopathy by dietary lysophosphatidylcholine-EPA/DHA. Sci Rep 2023; 13:9179. [PMID: 37280266 PMCID: PMC10244360 DOI: 10.1038/s41598-023-36268-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2023] [Accepted: 05/31/2023] [Indexed: 06/08/2023] Open
Abstract
Alzheimer disease (AD) is the most prevalent cause of dementia in the elderly. Although impaired cognition and memory are the most prominent features of AD, abnormalities in visual functions often precede them, and are increasingly being used as diagnostic and prognostic markers for the disease. Retina contains the highest concentration of the essential fatty acid docosahexaenoic acid (DHA) in the body, and its deficiency is associated with several retinal diseases including diabetic retinopathy and age related macular degeneration. In this study, we tested the hypothesis that enriching retinal DHA through a novel dietary approach could ameliorate symptoms of retinopathy in 5XFAD mice, a widely employed model of AD. The results show that 5XFAD mice have significantly lower retinal DHA compared to their wild type littermates, and feeding the lysophosphatidylcholine (LPC) form of DHA and eicosapentaenoic acid (EPA) rapidly normalizes the DHA levels, and increases retinal EPA by several-fold. On the other hand, feeding similar amounts of DHA and EPA in the form of triacylglycerol had only modest effects on retinal DHA and EPA. Electroretinography measurements after 2 months of feeding the experimental diets showed a significant improvement in a-wave and b-wave functions by the LPC-diet, whereas the TAG-diet had only a modest benefit. Retinal amyloid β levels were decreased by about 50% by the LPC-DHA/EPA diet, and by about 17% with the TAG-DHA/EPA diet. These results show that enriching retinal DHA and EPA through dietary LPC could potentially improve visual abnormalities associated with AD.
Collapse
Affiliation(s)
- Dhavamani Sugasini
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, University of Illinois, Chicago, IL, 60612, USA.
| | - Jason C Park
- Department of Ophthalmology and Visual Sciences, University of Illinois, Chicago, IL, 60612, USA
| | - J Jason McAnany
- Department of Ophthalmology and Visual Sciences, University of Illinois, Chicago, IL, 60612, USA
| | - Tae-Hoon Kim
- Department of Biomedical Engineering, University of Illinois, Chicago, IL, 60607, USA
| | - Guangying Ma
- Department of Biomedical Engineering, University of Illinois, Chicago, IL, 60607, USA
| | - Xincheng Yao
- Department of Ophthalmology and Visual Sciences, University of Illinois, Chicago, IL, 60612, USA
- Department of Biomedical Engineering, University of Illinois, Chicago, IL, 60607, USA
| | | | - Anil Oroskar
- Orochem Technologies, Inc, Naperville, IL, 60563, USA
| | | | - Brian T Layden
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, University of Illinois, Chicago, IL, 60612, USA
- Jesse Brown VA Medical Center, Chicago, IL, 60612, USA
| | - Papasani V Subbaiah
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, University of Illinois, Chicago, IL, 60612, USA.
- Jesse Brown VA Medical Center, Chicago, IL, 60612, USA.
| |
Collapse
|
29
|
Ying W. Phenomic Studies on Diseases: Potential and Challenges. PHENOMICS (CHAM, SWITZERLAND) 2023; 3:285-299. [PMID: 36714223 PMCID: PMC9867904 DOI: 10.1007/s43657-022-00089-4] [Citation(s) in RCA: 39] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/28/2021] [Revised: 11/21/2022] [Accepted: 11/24/2022] [Indexed: 01/23/2023]
Abstract
The rapid development of such research field as multi-omics and artificial intelligence (AI) has made it possible to acquire and analyze the multi-dimensional big data of human phenomes. Increasing evidence has indicated that phenomics can provide a revolutionary strategy and approach for discovering new risk factors, diagnostic biomarkers and precision therapies of diseases, which holds profound advantages over conventional approaches for realizing precision medicine: first, the big data of patients' phenomes can provide remarkably richer information than that of the genomes; second, phenomic studies on diseases may expose the correlations among cross-scale and multi-dimensional phenomic parameters as well as the mechanisms underlying the correlations; and third, phenomics-based studies are big data-driven studies, which can significantly enhance the possibility and efficiency for generating novel discoveries. However, phenomic studies on human diseases are still in early developmental stage, which are facing multiple major challenges and tasks: first, there is significant deficiency in analytical and modeling approaches for analyzing the multi-dimensional data of human phenomes; second, it is crucial to establish universal standards for acquirement and management of phenomic data of patients; third, new methods and devices for acquirement of phenomic data of patients under clinical settings should be developed; fourth, it is of significance to establish the regulatory and ethical guidelines for phenomic studies on diseases; and fifth, it is important to develop effective international cooperation. It is expected that phenomic studies on diseases would profoundly and comprehensively enhance our capacity in prevention, diagnosis and treatment of diseases.
Collapse
Affiliation(s)
- Weihai Ying
- Med-X Research Institute and School of Biomedical Engineering, Shanghai Jiao Tong University, 1954 Huashan Road, Shanghai, 200030 China
- Collaborative Innovation Center for Genetics and Development, Shanghai, 200043 China
| |
Collapse
|
30
|
Ashraf G, McGuinness M, Khan MA, Obtinalla C, Hadoux X, van Wijngaarden P. Retinal imaging biomarkers of Alzheimer's disease: A systematic review and meta-analysis of studies using brain amyloid beta status for case definition. ALZHEIMER'S & DEMENTIA (AMSTERDAM, NETHERLANDS) 2023; 15:e12421. [PMID: 37250908 PMCID: PMC10210353 DOI: 10.1002/dad2.12421] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Revised: 03/03/2023] [Accepted: 03/07/2023] [Indexed: 05/31/2023]
Abstract
Introduction We performed a systematic review and meta-analysis of the association between retinal imaging parameters and Alzheimer's disease (AD). Methods PubMed, EMBASE, and Scopus were systematically searched for prospective and observational studies. Included studies had AD case definition based on brain amyloid beta (Aβ) status. Study quality assessment was performed. Random-effects meta-analyses of standardized mean difference, correlation, and diagnostic accuracy were conducted. Results Thirty-eight studies were included. There was weak evidence of peripapillary retinal nerve fiber layer thinning on optical coherence tomography (OCT) (p = 0.14, 11 studies, n = 828), increased foveal avascular zone area on OCT-angiography (p = 0.18, four studies, n = 207), and reduced arteriole and venule vessel fractal dimension on fundus photography (p < 0.001 and p = 0.08, respectively, three studies, n = 297) among AD cases. Discussion Retinal imaging parameters appear to be associated with AD. Small study sizes and heterogeneity in imaging methods and reporting make it difficult to determine utility of these changes as AD biomarkers. Highlights We performed a systematic review on retinal imaging and Alzheimer's disease (AD).We only included studies in which cases were based on brain amyloid beta status.Several retinal biomarkers were associated with AD but clinical utility is uncertain.Studies should focus on biomarker-defined AD and use standardized imaging methods.
Collapse
Affiliation(s)
- Gizem Ashraf
- Centre for Eye Research AustraliaRoyal Victorian Eye and Ear HospitalMelbourneVictoriaAustralia
- OphthalmologyDepartment of SurgeryUniversity of MelbourneMelbourneVictoriaAustralia
| | - Myra McGuinness
- Centre for Eye Research AustraliaRoyal Victorian Eye and Ear HospitalMelbourneVictoriaAustralia
- Centre for Epidemiology and BiostatisticsMelbourne School of Population and Global HealthUniversity of MelbourneMelbourneVictoriaAustralia
| | - Muhammad Azaan Khan
- Faculty of Medicine and HealthUniversity of New South WalesSydneyNew South WalesAustralia
| | - Czarina Obtinalla
- Discipline of OrthopticsSchool of Allied HealthHuman Services & SportCollege of ScienceHealth & EngineeringLa Trobe UniversityMelbourneVictoriaAustralia
| | - Xavier Hadoux
- Centre for Eye Research AustraliaRoyal Victorian Eye and Ear HospitalMelbourneVictoriaAustralia
| | - Peter van Wijngaarden
- Centre for Eye Research AustraliaRoyal Victorian Eye and Ear HospitalMelbourneVictoriaAustralia
- OphthalmologyDepartment of SurgeryUniversity of MelbourneMelbourneVictoriaAustralia
| |
Collapse
|
31
|
Carrero L, Antequera D, Alcalde I, Megías D, Figueiro-Silva J, Merayo-Lloves J, Municio C, Carro E. Disturbed circadian rhythm and retinal degeneration in a mouse model of Alzheimer's disease. Acta Neuropathol Commun 2023; 11:55. [PMID: 37004084 PMCID: PMC10067208 DOI: 10.1186/s40478-023-01529-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Accepted: 02/11/2023] [Indexed: 04/03/2023] Open
Abstract
The circadian clock is synchronized to the 24 h day by environmental light which is transmitted from the retina to the suprachiasmatic nucleus (SCN) primarily via the retinohypothalamic tract (RHT). Circadian rhythm abnormalities have been reported in neurodegenerative disorders such as Alzheimer's disease (AD). Whether these AD-related changes are a result of the altered clock gene expression, retina degeneration, including the dysfunction in RHT transmission, loss of retinal ganglion cells and its electrophysiological capabilities, or a combination of all of these pathological mechanisms, is not known. Here, we evaluated transgenic APP/PS1 mouse model of AD and wild-type mice at 6- and 12-month-old, as early and late pathological stage, respectively. We noticed the alteration of circadian clock gene expression not only in the hypothalamus but also in two extra-hypothalamic brain regions, cerebral cortex and hippocampus, in APP/PS1 mice. These alterations were observed in 6-month-old transgenic mice and were exacerbated at 12 months of age. This could be explained by the reduced RHT projections in the SCN of APP/PS1 mice, correlating with downregulation of hypothalamic GABAergic response in APP/PS1 mice in advanced stage of pathology. Importantly, we also report retinal degeneration in APP/PS1 mice, including Aβ deposits and reduced choline acetyltransferase levels, loss of melanopsin retinal ganglion cells and functional integrity mainly of inner retina layers. Our findings support the theory that retinal degeneration constitutes an early pathological event that directly affects the control of circadian rhythm in AD.
Collapse
Affiliation(s)
- Laura Carrero
- Group of Neurodegenerative Diseases, Hospital Universitario 12 de Octubre Research Institute (imas12), 28041, Madrid, Spain
- Network Center for Biomedical Research in Neurodegenerative Diseases (CIBERNED), ISCIII, Madrid, Spain
- Autonoma de Madrid University, Madrid, Spain
| | - Desireé Antequera
- Network Center for Biomedical Research in Neurodegenerative Diseases (CIBERNED), ISCIII, Madrid, Spain
- Neurobiology of Alzheimer's Disease Unit, Functional Unit for Research into Chronic Diseases, Instituto de Salud Carlos III, Madrid, Spain
| | - Ignacio Alcalde
- Instituto Universitario Fernández-Vega, Universidad de Oviedo and Fundación de Investigación Oftalmológica, Oviedo, Spain
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Oviedo, Spain
| | - Diego Megías
- Advanced Optical Microscopy Unit, Unidades Centrales Científico-Técnicas, Instituto de Salud Carlos III, Madrid, Spain
| | - Joana Figueiro-Silva
- Institute of Medical Genetics, University of Zurich, Zurich, Switzerland
- Department of Molecular Life Science, University of Zurich, Zurich, Switzerland
| | - Jesús Merayo-Lloves
- Instituto Universitario Fernández-Vega, Universidad de Oviedo and Fundación de Investigación Oftalmológica, Oviedo, Spain
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Oviedo, Spain
| | - Cristina Municio
- Group of Neurodegenerative Diseases, Hospital Universitario 12 de Octubre Research Institute (imas12), 28041, Madrid, Spain.
- Network Center for Biomedical Research in Neurodegenerative Diseases (CIBERNED), ISCIII, Madrid, Spain.
| | - Eva Carro
- Network Center for Biomedical Research in Neurodegenerative Diseases (CIBERNED), ISCIII, Madrid, Spain.
- Neurobiology of Alzheimer's Disease Unit, Functional Unit for Research into Chronic Diseases, Instituto de Salud Carlos III, Madrid, Spain.
| |
Collapse
|
32
|
Gaber HA, Aly EM, Mohamed ES, Elfouly M, Talaat MS, El-Sayed ESM. Prognosis of Biomarker of Alzheimer’s Disease in the Function of the Retina and Secondary Molecular Structure Variation of the Retina and Brain. Int J Alzheimers Dis 2023; 2023:9775921. [PMID: 37035098 PMCID: PMC10076121 DOI: 10.1155/2023/9775921] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Revised: 02/06/2023] [Accepted: 03/15/2023] [Indexed: 03/31/2023] Open
Abstract
Alzheimer’s disease (AD) is one of the most serious neurodegenerative diseases in the globe. As a result, there is an acute need to discover indications that allow for early disease detection. There is growing scientific data showing the similarities between the eye and other central nervous system components, suggesting that information obtained in ophthalmic research might be valuable in the study and diagnosis of AD. Fifty male albino Wistar rats were separated into five groups: the first group served as control, and the other four groups of animals were administrated aluminium chloride (AlCl3) in a dose of 100 mg/kg body weight (b.w.) for 2, 4, 6, and 8 weeks, respectively. Insights into the function of the retina by electroretinogram (ERG) and the changes thought to have occurred in the molecular structure of the retina and brain using Fourier transform infrared spectroscopy (FTIR) as a result of AD progression induced by AlCl3 in rats were done. Moreover, the measurement of acetylcholinesterase (AchE) was done. After 6 and 8 weeks of AlCl3 injection, there was a substantial reduction (
) in a- and b-wave amplitudes and a significant rise (
) in implicit time compared to controls. A significant elevation (
) of AchE content was observed after 4, 6, and 8 weeks. FTIR revealed a significant increase (
) of β-turn and β-sheet content associated with significant decrease (
) of α-helix content for all groups administrated with AlCl3. Our findings suggest that retinal biomarkers such as ERG of the retina may be used as a screening tool for detection of AD. Secondary structural changes in the proteins of the retina and the brain were similar in AD rats’ model and precede retinal dysfunction.
Collapse
|
33
|
Moon S, Jeon S, Seo SK, Kim DE, Jung NY, Kim SJ, Lee MJ, Lee J, Kim EJ. Comparison of Retinal Structural and Neurovascular Changes between Patients with and without Amyloid Pathology. J Clin Med 2023; 12:jcm12041310. [PMID: 36835845 PMCID: PMC9964845 DOI: 10.3390/jcm12041310] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2023] [Revised: 01/29/2023] [Accepted: 02/01/2023] [Indexed: 02/10/2023] Open
Abstract
To evaluate whether an impaired anterior visual pathway (retinal structures with microvasculature) is associated with underlying beta-amyloid (Aβ) pathologies in patients with Alzheimer's disease dementia (ADD) and mild cognitive impairment (MCI), we compared retinal structural and vascular factors in each subgroup with positive or negative amyloid biomarkers. Twenty-seven patients with dementia, thirty-five with MCI, and nine with cognitively unimpaired (CU) controls were consecutively recruited. All participants were divided into positive Aβ (A+) or negative Aβ (A-) pathology based on amyloid positron emission tomography or cerebrospinal fluid Aβ. The retinal circumpapillary retinal nerve fiber layer thickness (cpRNFLT), macular ganglion cell/inner plexiform layer thickness (mGC/IPLT), and microcirculation of the macular superficial capillary plexus were measured using optical coherence tomography (OCT) and OCT angiography. One eye of each participant was included in the analysis. Retinal structural and vascular factors significantly decreased in the following order: dementia < MCI < CU controls. The A+ group had significantly lower microcirculation in the para- and peri-foveal temporal regions than did the A-. However, the structural and vascular parameters did not differ between the A+ and A- with dementia. The cpRNFLT was unexpectedly greater in the A+ than in the A- with MCI. mGC/IPLT was lower in the A+ CU than in the A- CU. Our findings suggest that retinal structural changes may occur in the preclinical and early stages of dementia but are not highly specific to AD pathophysiology. In contrast, decreased temporal macula microcirculation may be used as a biomarker for the underlying Aβ pathology.
Collapse
Affiliation(s)
- Sangwoo Moon
- Department of Ophthalmology, Pusan National University Hospital, Pusan National University School of Medicine and Biomedical Research Institute, Busan 49241, Republic of Korea
| | - Sumin Jeon
- Department of Neurology, Pusan National University Hospital, Pusan National University School of Medicine and Biomedical Research Institute, Busan 49241, Republic of Korea
| | - Sook Kyeong Seo
- Department of Neurology, Pusan National University Hospital, Pusan National University School of Medicine and Biomedical Research Institute, Busan 49241, Republic of Korea
| | - Da Eun Kim
- Department of Neurology, Pusan National University Hospital, Pusan National University School of Medicine and Biomedical Research Institute, Busan 49241, Republic of Korea
| | - Na-Yeon Jung
- Department of Neurology, Pusan National University Yangsan Hospital, Pusan National University School of Medicine, Yangsan 50612, Republic of Korea
| | - Seung Joo Kim
- Department of Neurology, Gyeongsang National University School of Medicine and Gyeongsang National University Changwon Hospital, Changwon 51472, Republic of Korea
| | - Myung Jun Lee
- Department of Neurology, Pusan National University Hospital, Pusan National University School of Medicine and Biomedical Research Institute, Busan 49241, Republic of Korea
| | - Jiwoong Lee
- Department of Ophthalmology, Pusan National University Hospital, Pusan National University School of Medicine and Biomedical Research Institute, Busan 49241, Republic of Korea
- Correspondence: (J.L.); (E.-J.K.)
| | - Eun-Joo Kim
- Department of Neurology, Pusan National University Hospital, Pusan National University School of Medicine and Biomedical Research Institute, Busan 49241, Republic of Korea
- Correspondence: (J.L.); (E.-J.K.)
| |
Collapse
|
34
|
Latina V, De Introna M, Caligiuri C, Loviglio A, Florio R, La Regina F, Pignataro A, Ammassari-Teule M, Calissano P, Amadoro G. Immunotherapy with Cleavage-Specific 12A12mAb Reduces the Tau Cleavage in Visual Cortex and Improves Visuo-Spatial Recognition Memory in Tg2576 AD Mouse Model. Pharmaceutics 2023; 15:pharmaceutics15020509. [PMID: 36839831 PMCID: PMC9965010 DOI: 10.3390/pharmaceutics15020509] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Revised: 01/25/2023] [Accepted: 01/31/2023] [Indexed: 02/05/2023] Open
Abstract
Tau-targeted immunotherapy is a promising approach for treatment of Alzheimer's disease (AD). Beyond cognitive decline, AD features visual deficits consistent with the manifestation of Amyloid β-protein (Aβ) plaques and neurofibrillary tangles (NFT) in the eyes and higher visual centers, both in animal models and affected subjects. We reported that 12A12-a monoclonal cleavage-specific antibody (mAb) which in vivo neutralizes the neurotoxic, N-terminal 20-22 kDa tau fragment(s)-significantly reduces the retinal accumulation in Tg(HuAPP695Swe)2576 mice of both tau and APP/Aβ pathologies correlated with local inflammation and synaptic deterioration. Here, we report the occurrence of N-terminal tau cleavage in the primary visual cortex (V1 area) and the beneficial effect of 12A12mAb treatment on phenotype-associated visuo-spatial deficits in this AD animal model. We found out that non-invasive administration of 12 A12mAb markedly reduced the pathological accumulation of both truncated tau and Aβ in the V1 area, correlated to significant improvement in visual recognition memory performance along with local increase in two direct readouts of cortical synaptic plasticity, including the dendritic spine density and the expression level of activity-regulated cytoskeleton protein Arc/Arg3.1. Translation of these findings to clinical therapeutic interventions could offer an innovative tau-directed opportunity to delay or halt the visual impairments occurring during AD progression.
Collapse
Affiliation(s)
- Valentina Latina
- European Brain Research Institute (EBRI), Viale Regina Elena 295, 00161 Rome, Italy
| | - Margherita De Introna
- Institute of Translational Pharmacology (IFT), National Research Council (CNR), Via Fosso del Cavaliere 100, 00133 Rome, Italy
- IRCCS Santa Lucia Foundation (FSL), Centro di Ricerca Europeo sul Cervello (CERC), Via Fosso del Fiorano 64-65, 00143 Rome, Italy
| | - Chiara Caligiuri
- IRCCS Santa Lucia Foundation (FSL), Centro di Ricerca Europeo sul Cervello (CERC), Via Fosso del Fiorano 64-65, 00143 Rome, Italy
| | - Alessia Loviglio
- European Brain Research Institute (EBRI), Viale Regina Elena 295, 00161 Rome, Italy
| | - Rita Florio
- European Brain Research Institute (EBRI), Viale Regina Elena 295, 00161 Rome, Italy
| | - Federico La Regina
- European Brain Research Institute (EBRI), Viale Regina Elena 295, 00161 Rome, Italy
| | - Annabella Pignataro
- Institute of Translational Pharmacology (IFT), National Research Council (CNR), Via Fosso del Cavaliere 100, 00133 Rome, Italy
- IRCCS Santa Lucia Foundation (FSL), Centro di Ricerca Europeo sul Cervello (CERC), Via Fosso del Fiorano 64-65, 00143 Rome, Italy
| | - Martine Ammassari-Teule
- IRCCS Santa Lucia Foundation (FSL), Centro di Ricerca Europeo sul Cervello (CERC), Via Fosso del Fiorano 64-65, 00143 Rome, Italy
- Institute of Biochemistry and Cell Biology (IBBC), National Research Council (CNR), Via Ercole Ramarini 32, 00015 Rome, Italy
| | - Pietro Calissano
- European Brain Research Institute (EBRI), Viale Regina Elena 295, 00161 Rome, Italy
| | - Giuseppina Amadoro
- European Brain Research Institute (EBRI), Viale Regina Elena 295, 00161 Rome, Italy
- Institute of Translational Pharmacology (IFT), National Research Council (CNR), Via Fosso del Cavaliere 100, 00133 Rome, Italy
- Correspondence: ; Tel.: +39-06-49255252
| |
Collapse
|
35
|
Dysfunction of the glutamatergic photoreceptor synapse in the P301S mouse model of tauopathy. Acta Neuropathol Commun 2023; 11:5. [PMID: 36631898 PMCID: PMC9832799 DOI: 10.1186/s40478-022-01489-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Accepted: 12/04/2022] [Indexed: 01/13/2023] Open
Abstract
Tauopathies, including Alzheimer's disease, are characterized by retinal ganglion cell loss associated with amyloid and phosphorylated tau deposits. We investigated the functional impact of these histopathological alterations in the murine P301S model of tauopathy. Visual impairments were demonstrated by a decrease in visual acuity already detectable at 6 months, the onset of disease. Visual signals to the cortex and retina were delayed at 6 and 9 months, respectively. Surprisingly, the retinal output signal was delayed at the light onset and advanced at the light offset. This antagonistic effect, due to a dysfunction of the cone photoreceptor synapse, was associated with changes in the expression of the vesicular glutamate transporter and a microglial reaction. This dysfunction of retinal glutamatergic synapses suggests a novel interpretation for visual deficits in tauopathies and it highlights the potential value of the retina for the diagnostic assessment and the evaluation of therapies in Alzheimer's disease and other tauopathies.
Collapse
|
36
|
Mozdbar S, Petersen M, Zhang F, Johnson L, Tolman A, Nyalakonda R, Gutierrez A, O’Bryant S. Application of Structural Retinal Biomarkers to Detect Cognitive Impairment in a Primary Care Setting. J Alzheimers Dis Rep 2022; 6:749-755. [PMID: 36721487 PMCID: PMC9837730 DOI: 10.3233/adr-220070] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2022] [Accepted: 11/11/2022] [Indexed: 12/14/2022] Open
Abstract
Background Despite the diagnostic accuracy of advanced neurodiagnostic procedures, the detection of Alzheimer's disease (AD) remains poor in primary care. There is an urgent need for screening tools to aid in the detection of early AD. Objective This study examines the predictive ability of structural retinal biomarkers in detecting cognitive impairment in a primary care setting. Methods Participants were recruited from Alzheimer's Disease in Primary Care (ADPC) study. As part of the ADPC Retinal Biomarker Study (ADPC RBS), visual acuity, an ocular history questionnaire, eye pressure, optical coherence tomography (OCT) imaging, and fundus imaging was performed. Results Data were examined on n = 91 participants. The top biomarkers for predicting cognitive impairment included the inferior quadrant of the outer retinal layers, all four quadrants of the peripapillary retinal nerve fiber layer, and the inferior quadrant of the macular retinal nerve fiber layer. Conclusion The current data provides strong support for continued investigation into structural retinal biomarkers, particularly the retinal nerve fiber layer, as screening tools for AD.
Collapse
Affiliation(s)
- Sima Mozdbar
- University of North Texas Health Science Center, Department of Pharmacology & Neuroscience, Fort Worth, TX, USA,University of North Texas Health Science Center, North Texas Eye Research Institute, Fort Worth, TX, USA,Anne Burnett Marion School of Medicine at Texas Christian University, Fort Worth, TX, USA,Correspondence to: Sima Mozdbar, OD, MPH, FAAO, 3500 Camp Bowie Blvd, Fort Worth, TX 76107, USA. Tel.: +1 817 735 2197; Fax: +1 817 735 7983; E-mail:
| | - Melissa Petersen
- University of North Texas Health Science Center, Texas College of Osteopathic Medicine, Fort Worth, TX, USA,University of North Texas Health Science Center, Institute for Translational Research, Fort Worth, TX, USA
| | - Fan Zhang
- University of North Texas Health Science Center, Texas College of Osteopathic Medicine, Fort Worth, TX, USA,University of North Texas Health Science Center, Institute for Translational Research, Fort Worth, TX, USA
| | - Leigh Johnson
- University of North Texas Health Science Center, Department of Pharmacology & Neuroscience, Fort Worth, TX, USA,University of North Texas Health Science Center, Institute for Translational Research, Fort Worth, TX, USA
| | - Alex Tolman
- Anne Burnett Marion School of Medicine at Texas Christian University, Fort Worth, TX, USA
| | - Ramyashree Nyalakonda
- University of North Texas Health Science Center, Texas College of Osteopathic Medicine, Fort Worth, TX, USA
| | - Alejandra Gutierrez
- Anne Burnett Marion School of Medicine at Texas Christian University, Fort Worth, TX, USA
| | - Sid O’Bryant
- University of North Texas Health Science Center, Texas College of Osteopathic Medicine, Fort Worth, TX, USA,University of North Texas Health Science Center, Institute for Translational Research, Fort Worth, TX, USA
| |
Collapse
|
37
|
Xia X, Qin Q, Peng Y, Wang M, Yin Y, Tang Y. Retinal Examinations Provides Early Warning of Alzheimer's Disease. J Alzheimers Dis 2022; 90:1341-1357. [PMID: 36245377 DOI: 10.3233/jad-220596] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Patients with Alzheimer's disease have difficulty maintaining independent living abilities as the disease progresses, causing an increased burden of care on family caregivers and the healthcare system and related financial strain. This patient group is expected to continue to expand as life expectancy climbs. Current diagnostics for Alzheimer's disease are complex, unaffordable, and invasive without regard to diagnosis quality at early stages, which urgently calls for more technical improvements for diagnosis specificity. Optical coherence tomography or tomographic angiography has been shown to identify retinal thickness loss and lower vascular density present earlier than symptom onset in these patients. The retina is an extension of the central nervous system and shares anatomic and functional similarities with the brain. Ophthalmological examinations can be an efficient tool to offer a window into cerebral pathology with the merit of easy operation. In this review, we summarized the latest observations on retinal pathology in Alzheimer's disease and discussed the feasibility of retinal imaging in diagnostic prediction, as well as limitations in current retinal examinations for Alzheimer's disease diagnosis.
Collapse
Affiliation(s)
- Xinyi Xia
- Innovation Center for Neurological Disorders, Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, China.,National Center for Neurological Disorders, National Clinical Research Center for Geriatric Diseases, Beijing, China
| | - Qi Qin
- Innovation Center for Neurological Disorders, Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, China.,National Center for Neurological Disorders, National Clinical Research Center for Geriatric Diseases, Beijing, China
| | - Yankun Peng
- Department of Ophthalmology, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Meng Wang
- Innovation Center for Neurological Disorders, Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Yunsi Yin
- Innovation Center for Neurological Disorders, Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Yi Tang
- Innovation Center for Neurological Disorders, Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
38
|
Chua J, Li C, Ho LKH, Wong D, Tan B, Yao X, Gan A, Schwarzhans F, Garhöfer G, Sng CCA, Hilal S, Venketasubramanian N, Cheung CY, Fischer G, Vass C, Wong TY, Chen CLH, Schmetterer L. A multi-regression framework to improve diagnostic ability of optical coherence tomography retinal biomarkers to discriminate mild cognitive impairment and Alzheimer’s disease. Alzheimers Res Ther 2022; 14:41. [PMID: 35272711 PMCID: PMC8908577 DOI: 10.1186/s13195-022-00982-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Accepted: 02/23/2022] [Indexed: 11/24/2022]
Abstract
Background Diagnostic performance of optical coherence tomography (OCT) to detect Alzheimer’s disease (AD) and mild cognitive impairment (MCI) remains limited. We assessed whether compensating the circumpapillary retinal nerve fiber layer (cpRNFL) thickness for multiple demographic and anatomical factors as well as the combination of macular layers improves the detection of MCI and AD. Methods This cross-sectional study of 62 AD (n = 92 eyes), 108 MCI (n = 158 eyes), and 55 cognitively normal control (n = 86 eyes) participants. Macular ganglion cell complex (mGCC) thickness was extracted. Circumpapillary retinal nerve fiber layer (cpRNFL) measurement was compensated for several ocular factors. Thickness measurements and their corresponding areas under the receiver operating characteristic curves (AUCs) were compared between the groups. The main outcome measure was OCT thickness measurements. Results Participants with MCI/AD showed significantly thinner measured and compensated cpRNFL, mGCC, and altered retinal vessel density (p < 0.05). Compensated RNFL outperformed measured RNFL for discrimination of MCI/AD (AUC = 0.74 vs 0.69; p = 0.026). Combining macular and compensated cpRNFL parameters provided the best detection of MCI/AD (AUC = 0.80 vs 0.69; p < 0.001). Conclusions and relevance Accounting for interindividual variations of ocular anatomical features in cpRNFL measurements and incorporating macular information may improve the identification of high-risk individuals with early cognitive impairment. Supplementary Information The online version contains supplementary material available at 10.1186/s13195-022-00982-0.
Collapse
|
39
|
Papageorgiou E, Tsirelis D, Lazari K, Siokas V, Dardiotis E, Tsironi EE. Visual disorders and driving ability in persons with dementia: A mini review. Front Hum Neurosci 2022; 16:932820. [DOI: 10.3389/fnhum.2022.932820] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2022] [Accepted: 11/11/2022] [Indexed: 11/30/2022] Open
Abstract
BackgroundImpaired driving ability in patients with Alzheimer’s disease (AD) is associated with a decline in cognitive processes and a deterioration of their basic sensory visual functions. Although a variety of ocular abnormalities have been described in patients with AD, little is known about the impact of those visual disorders on their driving performance.AimAim of this mini-review is to provide an update on the driving ability of patients with dementia and summarize the primary visual disorders affecting their driving behavior.MethodsDatabases were screened for studies investigating dementia, associated visual abnormalities and driving ability.ResultsThere is consistent evidence that dementia affects driving ability. Patients with dementia present with a variety of visual disorders, such as visual acuity reduction, visual field defects, impaired contrast sensitivity, decline in color vision and age-related pathological changes, that may have a negative impact on their driving ability. However, there is a paucity in studies describing the impact of oculovisual decline on the driving ability of AD subjects. A bidirectional association between cognitive and visual impairment (VI) has been described.ConclusionGiven the bidirectional association between VI and dementia, vision screening and cognitive assessment of the older driver should aim to identify at-risk individuals and employ timely strategies for treatment of both cognitive and ocular problems. Future studies should characterize the basic visual sensory status of AD patients participating in driving studies, and investigate the impact of vision abnormalities on their driving performance.
Collapse
|
40
|
Multi-modal retinal scanning to measure retinal thickness and peripheral blood vessels in multiple sclerosis. Sci Rep 2022; 12:20472. [PMID: 36443364 PMCID: PMC9705292 DOI: 10.1038/s41598-022-24312-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Accepted: 11/14/2022] [Indexed: 11/29/2022] Open
Abstract
Our purpose was to investigate changes to the retina in multiple sclerosis (MS) using established and novel modes of retinal image acquisition and analysis. 72 participants with MS and 80 healthy volunteers underwent retinal scanning with optical coherence tomography (OCT) and ultra-widefield (UWF) scanning laser ophthalmoscopy (SLO), over a two-year period. Changes in retinal nerve fibre layer (RNFL) thickness, macular volume and retinal blood vessel diameter were measured and parameters were then tested for associations with MS. Measurements from OCT showed that individuals with MS had a thinner RNFL and reduced macular volume when compared to healthy volunteers. On UWF images, participants with MS had reduced arterial widths in the inferior nasal quadrant of both eyes and reduced venous widths in the inferior nasal quadrant of right eyes. Longitudinal analysis showed that participants with MS had an accelerated annual rate of RNFL thinning in several regions of the retina. In conclusion, the assessment of OCT showed thinning of the RNFL and macula in concordance with previous reports on MS, while analysis of blood vessels in the retinal periphery from UWF-SLO images revealed novel changes.
Collapse
|
41
|
Mroczkowska S, Shokr H, Benavente-Pérez A, Negi A, Bentham P, Gherghel D. Retinal Microvascular Dysfunction Occurs Early and Similarly in Mild Alzheimer's Disease and Primary-Open Angle Glaucoma Patients. J Clin Med 2022; 11:6702. [PMID: 36431179 PMCID: PMC9717733 DOI: 10.3390/jcm11226702] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Revised: 11/06/2022] [Accepted: 11/11/2022] [Indexed: 11/16/2022] Open
Abstract
Purpose: To assess the similarities and differences in retinal microvascular function between mild Alzheimer’s disease (AD) patients, early-stage primary open angle glaucoma (POAG) patients and healthy controls. Methods: Retinal vessel reactivity to flickering light was assessed in 10 AD, 19 POAG and 20 healthy age matched control patients by means of dynamic retinal vessel analysis (DVA, IMEDOS, GmbH, Jena, Germany) according to an established protocol. All patients additionally underwent BP measurements and blood analysis for glucose and lipid metabolism markers. Results: AD and POAG patients demonstrated comparable alterations in retinal artery reactivity, in the form of an increased arterial reaction time (RT) to flicker light on the final flicker cycle (p = 0.009), which was not replicated by healthy controls (p > 0.05). Furthermore, the sequential changes in RT on progressing from flicker one to flicker three were found to differ between healthy controls and the two disease groups (p = 0.001). Conclusion: AD and POAG patients demonstrate comparable signs of vascular dysfunction in their retinal arteries at the early stages of their disease process. This provides support for the concept of a common underlying vascular aetiology in these two neurodegenerative diseases.
Collapse
Affiliation(s)
- Stephanie Mroczkowska
- Vascular Research Laboratory, Ophthalmic Research Group, College Health and Life Sciences, Aston University, Birmingham B4 7ET, UK
- Eye and Vision Research Group, School of Health Professions, Plymouth University, Plymouth PL4 8AA, UK
| | - Hala Shokr
- Vascular Research Laboratory, Ophthalmic Research Group, College Health and Life Sciences, Aston University, Birmingham B4 7ET, UK
- Pharmacy Division, Faculty of Biology, Medicine and Health, University of Manchester, Manchester M13 9PL, UK
| | - Alexandra Benavente-Pérez
- Vascular Research Laboratory, Ophthalmic Research Group, College Health and Life Sciences, Aston University, Birmingham B4 7ET, UK
| | - Anil Negi
- Medical Innovation Development and Research Unit, University Hospitals Birmingham NHS Foundation Trust, Birmingham B15 1NT, UK
| | - Peter Bentham
- Medical Innovation Development and Research Unit, University Hospitals Birmingham NHS Foundation Trust, Birmingham B15 1NT, UK
| | - Doina Gherghel
- Vascular Research Laboratory, Ophthalmic Research Group, College Health and Life Sciences, Aston University, Birmingham B4 7ET, UK
- Division of Cardiovascular Sciences, University of Manchester, Manchester M13 9PL, UK
| |
Collapse
|
42
|
Buscho S, Palacios E, Xia F, Shi S, Li S, Luisi J, Kayed R, Motamedi M, Zhang W, Liu H. Longitudinal characterization of retinal vasculature alterations with optical coherence tomography angiography in a mouse model of tauopathy. Exp Eye Res 2022; 224:109240. [PMID: 36096190 PMCID: PMC10162407 DOI: 10.1016/j.exer.2022.109240] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Revised: 08/05/2022] [Accepted: 08/29/2022] [Indexed: 01/16/2023]
Abstract
Tauopathies are a family of neurodegenerative diseases which predominately afflict the rapidly growing aging population suffering from various brain disorders including Alzheimer's disease, frontotemporal dementia with parkinsonism-17 and Pick disease. As the only visually accessible region of the central nervous system, in recent years, the retina has attracted extensive attention for its potential as a target for visualizing and quantifying emerging biomarkers of neurodegenerative diseases. Our previous study has found that retinal vascular inflammation and leakage occur at the very early stage of tauopathic mouse model. Here, we aimed to non-invasively visualize age-dependent alterations of retinal vasculature assessing the potential for using changes in retinal vasculature as the biomarker for the early diagnosis of tauopathy. Optical coherence tomography angiography (OCTA), a non-invasive depth-resolved high-resolution imaging technique was used to visualize and quantify tauopathy-induced alterations of retinal vasculature in P301S transgenic mice overexpressing the P301S mutant form of human tau and age-matched wild type littermate mice at 3, 6 and 10 months of age. We observed significant alterations of vascular features in the intermediate capillary plexus (ICP) and deep capillary plexus (DCP) but not in the superficial vascular complex (SVC) of P301S mice at early stages of tauopathy. With aging, alterations of vascular features in P301S mice became more prominent in all three vascular plexuses. Staining of retinal vasculature in flatmounts and trypsin digests of P301S mice at 10 months of age revealed decreased vessel density and increased acellular capillary formation, indicating that vascular degeneration also occurs during tauopathy. Overall, our results demonstrate that the changes in retinal vascular features accelerate during the progression of tauopathy. Vessels in the ICP and DCP may be more susceptible to tauopathy than vessels in the SVC. Since changes in retinal vasculature often precede tau pathology in the brain, non-invasive identification of retinal vascular alterations with OCTA may be a useful biomarker for the early diagnosis of tauopathy and monitoring its progression.
Collapse
Affiliation(s)
- Seth Buscho
- Department of Ophthalmology & Visual Sciences, University of Texas Medical Branch, Galveston, TX, USA
| | - Erick Palacios
- Department of Ophthalmology & Visual Sciences, University of Texas Medical Branch, Galveston, TX, USA
| | - Fan Xia
- Department of Ophthalmology & Visual Sciences, University of Texas Medical Branch, Galveston, TX, USA
| | - Shuizhen Shi
- Department of Ophthalmology & Visual Sciences, University of Texas Medical Branch, Galveston, TX, USA
| | - Shengguo Li
- Department of Ophthalmology & Visual Sciences, University of Texas Medical Branch, Galveston, TX, USA
| | - Jonathan Luisi
- Department of Ophthalmology & Visual Sciences, University of Texas Medical Branch, Galveston, TX, USA; Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, TX, USA
| | - Rakez Kayed
- Department of Neurology, University of Texas Medical Branch, Galveston, TX, USA
| | - Massoud Motamedi
- Department of Ophthalmology & Visual Sciences, University of Texas Medical Branch, Galveston, TX, USA
| | - Wenbo Zhang
- Department of Ophthalmology & Visual Sciences, University of Texas Medical Branch, Galveston, TX, USA; Departments of Neuroscience, Cell Biology & Anatomy, University of Texas Medical Branch, Galveston, TX, USA; Institute for Human Infections and Immunity, University of Texas Medical Branch, Galveston, TX, USA.
| | - Hua Liu
- Department of Ophthalmology & Visual Sciences, University of Texas Medical Branch, Galveston, TX, USA.
| |
Collapse
|
43
|
Vij R, Arora S. A systematic survey of advances in retinal imaging modalities for Alzheimer's disease diagnosis. Metab Brain Dis 2022; 37:2213-2243. [PMID: 35290546 DOI: 10.1007/s11011-022-00927-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Accepted: 02/04/2022] [Indexed: 01/06/2023]
Abstract
Recent advances in retinal imaging pathophysiology have shown a new function for biomarkers in Alzheimer's disease diagnosis and prognosis. The significant improvements in Optical coherence tomography (OCT) retinal imaging have led to significant clinical translation, particularly in Alzheimer's disease detection. This systematic review will provide a comprehensive overview of retinal imaging in clinical applications, with a special focus on biomarker analysis for use in Alzheimer's disease detection. Articles on OCT retinal imaging in Alzheimer's disease diagnosis were identified in PubMed, Google Scholar, IEEE Xplore, and Research Gate databases until March 2021. Those studies using simultaneous retinal imaging acquisition were chosen, while those using sequential techniques were rejected. "Alzheimer's disease" and "Dementia" were searched alone and in combination with "OCT" and "retinal imaging". Approximately 1000 publications were searched, and after deleting duplicate articles, 145 relevant studies focused on the diagnosis of Alzheimer's disease utilizing retinal imaging were chosen for study. OCT has recently been demonstrated to be a valuable technique in clinical practice as according to this survey, 57% of the researchers employed optical coherence tomography, 19% used ocular fundus imaging, 13% used scanning laser ophthalmoscopy, and 11% have used multimodal imaging to diagnose Alzheimer disease. Retinal imaging has become an important diagnostic technique for Alzheimer's disease. Given the scarcity of available literature, it is clear that future prospective trials involving larger and more homogeneous groups are necessary, and the work can be expanded by evaluating its significance utilizing a machine-learning platform rather than simply using statistical methodologies.
Collapse
Affiliation(s)
- Richa Vij
- School of Computer Science & Engineering, Shri Mata Vaishno Devi University, Katra, Jammu and Kashmir, 182320, India
| | - Sakshi Arora
- School of Computer Science & Engineering, Shri Mata Vaishno Devi University, Katra, Jammu and Kashmir, 182320, India.
| |
Collapse
|
44
|
Giuliani G, Sborgia G, Niro A, Castellana F, Lampignano L, Puzo P, Pascale A, Pastore V, Buonamassa R, Galati R, Bordinone M, Cassano F, Clemente A, Landini L, Scotti G, Gaudiomonte M, Guglielmi A, Semeraro R, Santoro M, Alessio G, Sardone R, Boscia F. Correlation between retinal vessel rarefaction and psychometric measures in an older Southern Italian population. Front Aging Neurosci 2022; 14:999796. [PMID: 36212041 PMCID: PMC9541429 DOI: 10.3389/fnagi.2022.999796] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Accepted: 09/06/2022] [Indexed: 12/22/2022] Open
Abstract
Objective To explore the linear association between inner retinal layers thickness and macular capillary density compared to variations of global cognition evaluated by psychometric measures in a cohort of Mediterranean subjects aged 65+ years. Materials and methods We performed a cross-sectional analysis of 574 participants aged 65 years+ drawn from a population-based Southern Italian study. All subjects underwent neurological evaluations, including global cognitive screening, the Mini-Mental State Examination (MMSE) and frontal assessment battery (FAB), together with an ophthalmic examination including optical coherence tomography (OCT) and OCT-Angiography. We assessed the average thickness of the ganglion cell complex (GCC) and the retinal nerve fiber layer (RNFL), the foveal avascular zone area, and vascular density (VD) of superficial (SVD) and deep (DVD) capillary plexi at the foveal and parafoveal area. Linear regression was applied to assess associations of ocular measurements with MMSE and FAB scores. Results In the linear regression model, foveal DVD (beta = 0.01, 95% CI:0.004–0.052), whole DVD (beta = 0.04, 95% CI:0.02–0.08), and whole SVD (beta = 0.04, 95% CI:0.02–0.07) showed a positive association with MMSE. In addition, foveal SVD (beta = 0.01, 95% CI:0.003–0.05) and whole SVD (beta = 0.03, 95% CI:0.004–0.08) were positively associated with the FAB score. We found no further significant association between the MMSE score or the FAB score and the average thickness of the GCC and RNFL, and FAZ area. Conclusion A direct linear association between the VD of the macular capillary plexi with global and frontal cognitive functions was observed in elderly subjects.
Collapse
Affiliation(s)
- Gianluigi Giuliani
- Department of Medical Science, Neuroscience and Sense Organs, Eye Clinic, University of Bari, Bari, Italy
| | - Giancarlo Sborgia
- Department of Medical Science, Neuroscience and Sense Organs, Eye Clinic, University of Bari, Bari, Italy
| | - Alfredo Niro
- Eye Clinic, Hospital “SS Annunziata,” ASL Taranto, Taranto, Italy
- *Correspondence: Alfredo Niro,
| | - Fabio Castellana
- Unit of Research Methodology and Data Sciences for Population Health, “Salus in Apulia Study,” National Institute of Gastroenterology “Saverio de Bellis,” Research Hospital, Bari, Italy
| | - Luisa Lampignano
- Unit of Research Methodology and Data Sciences for Population Health, “Salus in Apulia Study,” National Institute of Gastroenterology “Saverio de Bellis,” Research Hospital, Bari, Italy
| | - Pasquale Puzo
- Department of Medical Science, Neuroscience and Sense Organs, Eye Clinic, University of Bari, Bari, Italy
| | - Angelo Pascale
- Department of Medical Science, Neuroscience and Sense Organs, Eye Clinic, University of Bari, Bari, Italy
| | - Valentina Pastore
- Department of Medical Science, Neuroscience and Sense Organs, Eye Clinic, University of Bari, Bari, Italy
| | - Rosa Buonamassa
- Department of Medical Science, Neuroscience and Sense Organs, Eye Clinic, University of Bari, Bari, Italy
| | - Roberta Galati
- Department of Medical Science, Neuroscience and Sense Organs, Eye Clinic, University of Bari, Bari, Italy
| | - Marco Bordinone
- Department of Medical Science, Neuroscience and Sense Organs, Eye Clinic, University of Bari, Bari, Italy
| | - Flavio Cassano
- Department of Medical Science, Neuroscience and Sense Organs, Eye Clinic, University of Bari, Bari, Italy
| | - Arcangelo Clemente
- Department of Medical Science, Neuroscience and Sense Organs, Eye Clinic, University of Bari, Bari, Italy
| | - Luca Landini
- Department of Medical Science, Neuroscience and Sense Organs, Eye Clinic, University of Bari, Bari, Italy
| | - Giacomo Scotti
- Department of Medical Science, Neuroscience and Sense Organs, Eye Clinic, University of Bari, Bari, Italy
| | - Marida Gaudiomonte
- Department of Medical Science, Neuroscience and Sense Organs, Eye Clinic, University of Bari, Bari, Italy
| | - Antonella Guglielmi
- Department of Medical Science, Neuroscience and Sense Organs, Eye Clinic, University of Bari, Bari, Italy
| | - Roberto Semeraro
- Department of Medical Science, Neuroscience and Sense Organs, Eye Clinic, University of Bari, Bari, Italy
| | - Michele Santoro
- Department of Medical Science, Neuroscience and Sense Organs, Eye Clinic, University of Bari, Bari, Italy
| | - Giovanni Alessio
- Department of Medical Science, Neuroscience and Sense Organs, Eye Clinic, University of Bari, Bari, Italy
| | - Rodolfo Sardone
- Unit of Research Methodology and Data Sciences for Population Health, “Salus in Apulia Study,” National Institute of Gastroenterology “Saverio de Bellis,” Research Hospital, Bari, Italy
| | - Francesco Boscia
- Department of Medical Science, Neuroscience and Sense Organs, Eye Clinic, University of Bari, Bari, Italy
| |
Collapse
|
45
|
Wang X, Wang Y, Liu H, Zhu X, Hao X, Zhu Y, Xu B, Zhang S, Jia X, Weng L, Liao X, Zhou Y, Tang B, Zhao R, Jiao B, Shen L. Macular Microvascular Density as a Diagnostic Biomarker for Alzheimer’s Disease. J Alzheimers Dis 2022; 90:139-149. [DOI: 10.3233/jad-220482] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Background: Some previous studies showed abnormal pathological and vascular changes in the retina of patients with Alzheimer’s disease (AD). However, whether retinal microvascular density is a diagnostic indicator for AD remains unclear. Objective: This study evaluated the macular vessel density (m-VD) in the superficial capillary plexus and fovea avascular zone (FAZ) area in AD, explored their correlations with clinical parameters, and finally confirmed an optimal machine learning model for AD diagnosis. Methods: 77 patients with AD and 145 healthy controls (HCs) were enrolled. The m-VD and the FAZ area were measured using optical coherence tomography angiography (OCTA) in all participants. Additionally, AD underwent neuropsychological assessment, brain magnetic resonance imaging scan, cerebrospinal fluid (CSF) biomarker detection, and APOE ɛ4 genotyping. Finally, the performance of machine learning algorithms based on the OCTA measurements was evaluated by Python programming language. Results: The m-VD was noticeably decreased in AD compared with HCs. Moreover, m-VD in the fovea, superior inner, inferior inner, nasal inner subfields, and the whole inner ring declined significantly in mild AD, while it was more serious in moderate/severe AD. However, no significant difference in the FAZ was noted between AD and HCs. Furthermore, we found that m-VD exhibited a significant correlation with cognitive function, medial temporal atrophy and Fazekas scores, and APOE ɛ4 genotypes. No significant correlations were observed between m-VD and CSF biomarkers. Furthermore, results revealed the Adaptive boosting algorithm exhibited the best diagnostic performance for AD. Conclusion: Macular vascular density could serve as a diagnostic biomarker for AD.
Collapse
Affiliation(s)
- Xin Wang
- Department of Neurology, Xiangya Hospital, CentralSouth University, Changsha, China
| | - Yaqin Wang
- Health Management Center, the Third Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Hui Liu
- Department of Neurology, Xiangya Hospital, CentralSouth University, Changsha, China
| | - Xiangyu Zhu
- Department of Neurology, Xiangya Hospital, CentralSouth University, Changsha, China
| | - Xiaoli Hao
- Department of Neurology, Xiangya Hospital, CentralSouth University, Changsha, China
| | - Yuan Zhu
- Department of Neurology, Xiangya Hospital, CentralSouth University, Changsha, China
| | - Bei Xu
- Eye Center of Xiangya Hospital, Central South University, Changsha, China
| | - Sizhe Zhang
- Department of Neurology, Xiangya Hospital, CentralSouth University, Changsha, China
| | - Xiaoliang Jia
- School of Computer Science and Engineering, Central South University, Changsha, China
| | - Ling Weng
- Department of Neurology, Xiangya Hospital, CentralSouth University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Central South University, Changsha, China
- Engineering Research Center of Hunan Province in Cognitive Impairment Disorders, Central South University, Changsha, China
- Hunan International Scientific and Technological Cooperation Base of Neurodegenerative and Neurogenetic Diseases, Changsha, China
- Key Laboratory of Hunan Province inNeurodegenerative Disorders, Central South University, Changsha, China
| | - Xinxin Liao
- National Clinical Research Center for Geriatric Disorders, Central South University, Changsha, China
- Engineering Research Center of Hunan Province in Cognitive Impairment Disorders, Central South University, Changsha, China
- Hunan International Scientific and Technological Cooperation Base of Neurodegenerative and Neurogenetic Diseases, Changsha, China
- Key Laboratory of Hunan Province inNeurodegenerative Disorders, Central South University, Changsha, China
- Department of Geriatrics, Xiangya Hospital, Central South University, Changsha, China
| | - Yafang Zhou
- National Clinical Research Center for Geriatric Disorders, Central South University, Changsha, China
- Engineering Research Center of Hunan Province in Cognitive Impairment Disorders, Central South University, Changsha, China
- Hunan International Scientific and Technological Cooperation Base of Neurodegenerative and Neurogenetic Diseases, Changsha, China
- Key Laboratory of Hunan Province inNeurodegenerative Disorders, Central South University, Changsha, China
- Department of Geriatrics, Xiangya Hospital, Central South University, Changsha, China
| | - Beisha Tang
- Department of Neurology, Xiangya Hospital, CentralSouth University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Central South University, Changsha, China
- Engineering Research Center of Hunan Province in Cognitive Impairment Disorders, Central South University, Changsha, China
- Hunan International Scientific and Technological Cooperation Base of Neurodegenerative and Neurogenetic Diseases, Changsha, China
- Key Laboratory of Hunan Province inNeurodegenerative Disorders, Central South University, Changsha, China
| | - Rongchang Zhao
- School of Computer Science and Engineering, Central South University, Changsha, China
| | - Bin Jiao
- Department of Neurology, Xiangya Hospital, CentralSouth University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Central South University, Changsha, China
- Engineering Research Center of Hunan Province in Cognitive Impairment Disorders, Central South University, Changsha, China
- Hunan International Scientific and Technological Cooperation Base of Neurodegenerative and Neurogenetic Diseases, Changsha, China
- Key Laboratory of Hunan Province inNeurodegenerative Disorders, Central South University, Changsha, China
| | - Lu Shen
- Department of Neurology, Xiangya Hospital, CentralSouth University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Central South University, Changsha, China
- Engineering Research Center of Hunan Province in Cognitive Impairment Disorders, Central South University, Changsha, China
- Hunan International Scientific and Technological Cooperation Base of Neurodegenerative and Neurogenetic Diseases, Changsha, China
- Key Laboratory of Hunan Province inNeurodegenerative Disorders, Central South University, Changsha, China
| |
Collapse
|
46
|
Cheung CY, Wong WLE, Hilal S, Kan CN, Gyanwali B, Tham YC, Schmetterer L, Xu D, Lee ML, Hsu W, Venketasubramanian N, Tan BY, Wong TY, Chen CPLH. Deep-learning retinal vessel calibre measurements and risk of cognitive decline and dementia. Brain Commun 2022; 4:fcac212. [PMID: 36043139 PMCID: PMC9416061 DOI: 10.1093/braincomms/fcac212] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Revised: 06/07/2022] [Accepted: 08/16/2022] [Indexed: 10/14/2023] Open
Abstract
Previous studies have explored the associations of retinal vessel calibre, measured from retinal photographs or fundus images using semi-automated computer programs, with cognitive impairment and dementia, supporting the concept that retinal blood vessels reflect microvascular changes in the brain. Recently, artificial intelligence deep-learning algorithms have been developed for the fully automated assessment of retinal vessel calibres. Therefore, we aimed to determine whether deep-learning-based retinal vessel calibre measurements are predictive of risk of cognitive decline and dementia. We conducted a prospective study recruiting participants from memory clinics at the National University Hospital and St. Luke's Hospital in Singapore; all participants had comprehensive clinical and neuropsychological examinations at baseline and annually for up to 5 years. Fully automated measurements of retinal arteriolar and venular calibres from retinal fundus images were estimated using a deep-learning system. Cox regression models were then used to assess the relationship between baseline retinal vessel calibre and the risk of cognitive decline and developing dementia, adjusting for age, gender, ethnicity, education, cerebrovascular disease status, hypertension, hyperlipidemia, diabetes, and smoking. A total of 491 participants were included in this study, of whom 254 developed cognitive decline over 5 years. In multivariable models, narrower retinal arteriolar calibre (hazard ratio per standard deviation decrease = 1.258, P = 0.008) and wider retinal venular calibre (hazard ratio per standard deviation increase = 1.204, P = 0.037) were associated with increased risk of cognitive decline. Among participants with cognitive impairment but no dementia at baseline (n = 212), 44 progressed to have incident dementia; narrower retinal arteriolar calibre was also associated with incident dementia (hazard ratio per standard deviation decrease = 1.624, P = 0.021). In summary, deep-learning-based measurement of retinal vessel calibre was associated with risk of cognitive decline and dementia.
Collapse
Affiliation(s)
- Carol Y Cheung
- Department of Ophthalmology and Visual Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Win Lee Edwin Wong
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117600, Singapore
- Memory Ageing and Cognition Centre, National University Health System, Singapore 119074, Singapore
| | - Saima Hilal
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117600, Singapore
- Memory Ageing and Cognition Centre, National University Health System, Singapore 119074, Singapore
- Saw Swee Hock School of Public Health, National University of Singapore and National University Health System, Singapore 117549, Singapore
| | - Cheuk Ni Kan
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117600, Singapore
- Memory Ageing and Cognition Centre, National University Health System, Singapore 119074, Singapore
| | - Bibek Gyanwali
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117600, Singapore
- Memory Ageing and Cognition Centre, National University Health System, Singapore 119074, Singapore
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117600, Singapore
| | - Yih Chung Tham
- Singapore Eye Research Institute, Singapore National Eye Centre, Singapore 169856, Singapore
- Ophthalmology and Visual Sciences Academic Clinical Programme, Duke-NUS Medical School, Singapore 169857, Singapore
| | - Leopold Schmetterer
- Singapore Eye Research Institute, Singapore National Eye Centre, Singapore 169856, Singapore
- School of Chemical and Biomedical Engineering, Nanyang Technological University, Singapore 639798, Singapore
- Department of Clinical Pharmacology, Medical University Vienna, Vienna 1090, Austria
- Austria Center for Medical Physics and Biomedical Engineering, Medical University Vienna, Vienna 1090, Austria
- Institute of Molecular and Clinical Ophthalmology, Basel, Switzerland
| | - Dejiang Xu
- School of Computing, National University of Singapore, Singapore 117417, Singapore
| | - Mong Li Lee
- School of Computing, National University of Singapore, Singapore 117417, Singapore
| | - Wynne Hsu
- School of Computing, National University of Singapore, Singapore 117417, Singapore
| | | | - Boon Yeow Tan
- St. Luke's Hospital, Singapore, Singapore 659674, Singapore
| | - Tien Yin Wong
- Singapore Eye Research Institute, Singapore National Eye Centre, Singapore 169856, Singapore
- Ophthalmology and Visual Sciences Academic Clinical Programme, Duke-NUS Medical School, Singapore 169857, Singapore
| | - Christopher P L H Chen
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117600, Singapore
- Memory Ageing and Cognition Centre, National University Health System, Singapore 119074, Singapore
- Department of Psychological Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117600, Singapore
| |
Collapse
|
47
|
Matei N, Leahy S, Blair NP, Burford J, Rahimi M, Shahidi M. Retinal Vascular Physiology Biomarkers in a 5XFAD Mouse Model of Alzheimer's Disease. Cells 2022; 11:2413. [PMID: 35954257 PMCID: PMC9368483 DOI: 10.3390/cells11152413] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Revised: 07/30/2022] [Accepted: 07/30/2022] [Indexed: 11/17/2022] Open
Abstract
BACKGROUND Alzheimer's disease (AD) is a neurodegenerative disorder that affects the brain and retina and lacks reliable biomarkers for early diagnosis. As amyloid beta (Aβ) manifestations emerge prior to clinical symptoms and plaques of amyloid may cause vascular damage, identification of retinal vascular biomarkers may improve knowledge of AD pathophysiology and potentially serve as therapeutic targets. The purpose of the current study was to test the hypothesis that retinal hemodynamic and oxygen metrics are altered in 5XFAD mice. METHODS Thirty-two male mice were evaluated at 3 months of age: sixteen 5XFAD transgenic and sixteen wild-type mice. Spectral-domain optical coherence tomography, vascular oxygen tension, and blood flow imaging were performed in one eye of each mouse. After imaging, the imaged and fellow retinal tissues were submitted for histological sectioning and amyloid protein analysis, respectively. Protein analysis was also performed on the brain tissues. RESULTS Retinal physiological changes in venous diameter and blood velocity, arterial and venous oxygen contents, coupled with anatomical alterations in the thickness of retinal cell layers were detected in 5XFAD mice. Moreover, an increase in Aβ42 levels in both the retina and brain tissues was observed in 5XFAD mice. Significant changes in retinal oxygen delivery, metabolism, or extraction fraction were not detected. Based on compiled data from both groups, arterial oxygen content was inversely related to venous blood velocity and nerve fiber/ganglion cell layer thickness. CONCLUSIONS Concurrent alterations in retinal hemodynamic and oxygen metrics, thickness, and tissue Aβ42 protein levels in 5XFAD mice at 3 months of age corresponded to previously reported findings in human AD. Overall, these results suggest that this mouse model can be utilized for studying pathophysiology of AD and evaluating potential therapies.
Collapse
Affiliation(s)
- Nathanael Matei
- Department of Ophthalmology, University of Southern California, Los Angeles, CA 90033, USA
| | - Sophie Leahy
- Department of Ophthalmology, University of Southern California, Los Angeles, CA 90033, USA
| | - Norman P. Blair
- Department of Ophthalmology and Visual Sciences, University of Illinois at Chicago, Chicago, IL 60612, USA
| | - James Burford
- Department of Ophthalmology, University of Southern California, Los Angeles, CA 90033, USA
| | - Mansour Rahimi
- Department of Ophthalmology, University of Southern California, Los Angeles, CA 90033, USA
| | - Mahnaz Shahidi
- Department of Ophthalmology, University of Southern California, Los Angeles, CA 90033, USA
| |
Collapse
|
48
|
Rai H, Gupta S, Kumar S, Yang J, Singh SK, Ran C, Modi G. Near-Infrared Fluorescent Probes as Imaging and Theranostic Modalities for Amyloid-Beta and Tau Aggregates in Alzheimer's Disease. J Med Chem 2022; 65:8550-8595. [PMID: 35759679 DOI: 10.1021/acs.jmedchem.1c01619] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
A person suspected of having Alzheimer's disease (AD) is clinically diagnosed for the presence of principal biomarkers, especially misfolded amyloid-beta (Aβ) and tau proteins in the brain regions. Existing radiotracer diagnostic tools, such as PET imaging, are expensive and have limited availability for primary patient screening and pre-clinical animal studies. To change the status quo, small-molecular near-infrared (NIR) probes have been rapidly developed, which may serve as an inexpensive, handy imaging tool to comprehend the dynamics of pathogenic progression in AD and assess therapeutic efficacy in vivo. This Perspective summarizes the biochemistry of Aβ and tau proteins and then focuses on structurally diverse NIR probes with coverages of their spectroscopic properties, binding affinity toward Aβ and tau species, and theranostic effectiveness. With the summarized information and perspective discussions, we hope that this paper may serve as a guiding tool for designing novel in vivo imaging fluoroprobes with theranostic capabilities in the future.
Collapse
Affiliation(s)
- Himanshu Rai
- Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (BHU), Varanasi, U.P.-221005, India
| | - Sarika Gupta
- Molecular Science Laboratory, National Institute of Immunology, New Delhi-110067, India
| | - Saroj Kumar
- Department of Biophysics, All India Institute of Medical Sciences, New Delhi-110029, India
| | - Jian Yang
- School of Medicine, Shanghai University, Shanghai 200444, China
| | - Sushil K Singh
- Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (BHU), Varanasi, U.P.-221005, India
| | - Chongzhao Ran
- Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts 02129, United States
| | - Gyan Modi
- Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (BHU), Varanasi, U.P.-221005, India
| |
Collapse
|
49
|
Moran C, Xu ZY, Mehta H, Gillies M, Karayiannis C, Beare R, Chen C, Srikanth V. Neuroimaging and cognitive correlates of retinal Optical Coherence Tomography (OCT) measures at late middle age in a twin sample. Sci Rep 2022; 12:9562. [PMID: 35688899 PMCID: PMC9187769 DOI: 10.1038/s41598-022-13662-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Accepted: 05/26/2022] [Indexed: 11/09/2022] Open
Abstract
Sharing in embryology and function between the eye and brain has led to interest in whether assessments of the eye reflect brain changes seen in neurodegeneration. We aimed to examine the associations between measures of retinal layer thickness using optical coherence tomography (OCT) and multimodal measures of brain structure and function. Using a convenient sample of twins discordant for type 2 diabetes, we performed cognitive testing, structural brain MRI (tissue volumetry), diffusion tensor imaging (white matter microstructure), and arterial spin labelling (cerebral blood flow). OCT images were recorded and retinal thickness maps generated. We used mixed level modelling to examine the relationship between retinal layer thicknesses and brain measures. We enrolled 35 people (18 pairs, mean age 63.8 years, 63% female). Ganglion cell layer thickness was positively associated with memory, speed, gray matter volume, and altered mean diffusivity. Ganglion cell layer thickness was strongly positively associated with regional cerebral blood flow. We found only a limited number of associations between other retinal layer thickness and measures of brain structure or function. Ganglion cell layer thickness showed consistent associations with a range of brain measures suggesting it may have utility as a marker for future dementia risk.
Collapse
Affiliation(s)
- Chris Moran
- National Centre for Healthy Ageing, Melbourne, Australia.,Department of Geriatric Medicine, Peninsula Health and Central Clinical School, Monash University, Melbourne, Australia.,Department of Aged Care, Alfred Health, Melbourne, Australia
| | - Zheng Yang Xu
- Royal Free London NHS Foundation Trust, London, UK.,UCL Medical School, London, UK
| | - Hemal Mehta
- Royal Free London NHS Foundation Trust, London, UK.,Macular Research Group, University of Sydney, Sydney, Australia
| | - Mark Gillies
- Macular Research Group, University of Sydney, Sydney, Australia
| | - Chris Karayiannis
- National Centre for Healthy Ageing, Melbourne, Australia.,Department of Geriatric Medicine, Peninsula Health and Central Clinical School, Monash University, Melbourne, Australia
| | - Richard Beare
- National Centre for Healthy Ageing, Melbourne, Australia.,Department of Geriatric Medicine, Peninsula Health and Central Clinical School, Monash University, Melbourne, Australia
| | - Christine Chen
- Department of Ophthalmology, Monash Health, Melbourne, Australia
| | - Velandai Srikanth
- National Centre for Healthy Ageing, Melbourne, Australia. .,Department of Geriatric Medicine, Peninsula Health and Central Clinical School, Monash University, Melbourne, Australia.
| |
Collapse
|
50
|
Kim HM, Han JW, Park YJ, Bae JB, Woo SJ, Kim KW. Association Between Retinal Layer Thickness and Cognitive Decline in Older Adults. JAMA Ophthalmol 2022; 140:683-690. [PMID: 35616950 PMCID: PMC9136677 DOI: 10.1001/jamaophthalmol.2022.1563] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Question Is retinal layer thickness associated with cognitive decline in an older population? Findings In this cohort study including 430 community-dwelling participants in Korea, baseline macular retinal nerve fiber layer (RNFL) thickness was associated with baseline cognitive function scores and follow-up cognitive decline. Meaning These findings suggest that macular RNFL thickness could be considered a predictive biomarker for evaluating cognitive function in older individuals. Importance Retinal layer thickness is hypothesized to be related to cognitive function in patients with mild cognitive impairment (MCI) and Alzheimer disease (AD). However, longitudinal cohort studies of the healthy older population are scarce. Objective To investigate the association between retinal layer thickness and cognitive impairment and future cognitive decline in a community-based population cohort. Design, Setting, and Participants A total of 430 randomly sampled community-dwelling Korean individuals 60 years or older participated in the baseline assessment (mean [SD], 76.3 [6.6] years) 215 of whom completed a mean (SD) of 5.4 (0.6) years (range, 4.1-6.2 years) of follow-up. Using spectral-domain optical coherence tomography, the study team assessed the thickness of 6 retinal layers in the macular region, the peripapillary retinal nerve fiber layers (RNFLs), and the subfoveal choroid at baseline. Exposures Age, sex, education, diabetes, hypertension, and apolipoprotein E4 gene status. Main Outcomes and Measures Retinal layer thickness and cognitive function test scores were analyzed. Results This study included 430 participants (female, 208 [48.6%]). Baseline macular RNFL thickness was associated with baseline Consortium to Establish a Registry for Alzheimer’s Disease (CERAD) score (coefficient [β] = 0.077; 95% CI, 0.054-0.100; P = .04 for total macular area) and Mini-Mental State Examination (MMSE) score (coefficient [β] = 0.082; 95% CI, 0.063-0.101; P = .03 for total macular area). A thinner baseline total macular RNFL thickness (lowest quartile, <231 μm) was associated with a larger decline in the CERAD and MMSE scores during the follow-up period (P = .003 and P = .01, respectively). Furthermore, participants with baseline total macular RNFL thickness below the lowest quartile cutoff value presented a greater decline in cognitive scores and a higher prevalence of cognitive impairment and Alzheimer disease than those with RNFL thickness above the lowest quartile cutoff value. Conclusions and Relevance In this study, macular RNFL thickness could be used as a prognostic biomarker of long-term cognitive decline in adults 60 years or older. However, to confirm these results, further large-scale population-based studies should be performed.
Collapse
Affiliation(s)
- Hyeong Min Kim
- Department of Ophthalmology, Seoul National University College of Medicine, Seoul National University Bundang Hospital, Seongnam, Republic of Korea
| | - Ji Won Han
- Department of Neuropsychiatry, Seoul National University Bundang Hospital, Seongnam, Republic of Korea
| | - Young Joo Park
- Department of Ophthalmology, Kangwon National University School of Medicine, Kangwon National University Hospital, Chuncheon, Republic of Korea
| | - Jong Bin Bae
- Department of Neuropsychiatry, Seoul National University Bundang Hospital, Seongnam, Republic of Korea
| | - Se Joon Woo
- Department of Ophthalmology, Seoul National University College of Medicine, Seoul National University Bundang Hospital, Seongnam, Republic of Korea
| | - Ki Woong Kim
- Department of Neuropsychiatry, Seoul National University Bundang Hospital, Seongnam, Republic of Korea.,Department of Psychiatry and Behavioral Science, Seoul National University College of Medicine, Seoul, Republic of Korea.,Department of Brain and Cognitive Science, Seoul National University College of Natural Sciences, Seoul, Republic of Korea
| |
Collapse
|