1
|
Soundara Pandi SP, Winter H, Smith MR, Harkin K, Bojdo J. Preclinical Retinal Disease Models: Applications in Drug Development and Translational Research. Pharmaceuticals (Basel) 2025; 18:293. [PMID: 40143072 PMCID: PMC11944893 DOI: 10.3390/ph18030293] [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: 01/26/2025] [Revised: 02/10/2025] [Accepted: 02/18/2025] [Indexed: 03/28/2025] Open
Abstract
Retinal models play a pivotal role in translational drug development, bridging preclinical research and therapeutic applications for both ocular and systemic diseases. This review highlights the retina as an ideal organ for studying advanced therapies, thanks to its immune privilege, vascular and neuronal networks, accessibility, and advanced imaging capabilities. Preclinical retinal disease models offer unparalleled insights into inflammation, angiogenesis, fibrosis, and hypoxia, utilizing clinically translatable bioimaging tools like fundoscopy, optical coherence tomography, confocal scanning laser ophthalmoscopy, fluorescein angiography, optokinetic tracking, and electroretinography. These models have driven innovations in anti-inflammatory, anti-angiogenic, and neuroprotective strategies, with broader implications for systemic diseases such as rheumatoid arthritis, Alzheimer's, and fibrosis-related conditions. By emphasizing the integration of the 3Rs principles and novel imaging modalities, this review highlights how retinal research not only enhances therapeutic precision but also minimizes ethical concerns, paving the way for more predictive and human-relevant approaches in drug development.
Collapse
Affiliation(s)
| | - Hanagh Winter
- Medinect Bioservices Ltd., Belfast BT7 1NF, UK; (S.P.S.P.); (H.W.); (M.R.S.); (K.H.)
| | - Madeleine R. Smith
- Medinect Bioservices Ltd., Belfast BT7 1NF, UK; (S.P.S.P.); (H.W.); (M.R.S.); (K.H.)
| | - Kevin Harkin
- Medinect Bioservices Ltd., Belfast BT7 1NF, UK; (S.P.S.P.); (H.W.); (M.R.S.); (K.H.)
- Wellcome-Wolfson Institute for Experimental Medicine, Queen’s University Belfast, Belfast BT9 7BL, UK
| | - James Bojdo
- Medinect Bioservices Ltd., Belfast BT7 1NF, UK; (S.P.S.P.); (H.W.); (M.R.S.); (K.H.)
| |
Collapse
|
2
|
Hosseinpour Mashkani SM, Bishop DP, Westerhausen MT, Adlard PA, Golzan SM. Alterations in zinc, copper, and iron levels in the retina and brain of Alzheimer's disease patients and the APP/PS1 mouse model. Metallomics 2024; 16:mfae053. [PMID: 39520546 PMCID: PMC11630249 DOI: 10.1093/mtomcs/mfae053] [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: 04/18/2024] [Accepted: 11/06/2024] [Indexed: 11/16/2024]
Abstract
Transition metals like copper (Cu), iron (Fe), and zinc (Zn) are vital for normal central nervous system function and are also linked to neurodegeneration, particularly in the onset and progression of Alzheimer's disease (AD). Their alterations in AD, identified prior to amyloid plaque aggregation, offer a unique target for staging pre-amyloid AD. However, analysing their levels in the brain is extremely challenging, necessitating the development of alternative approaches. Here, we utilized laser ablation-inductively coupled plasma-mass spectrometry and solution nebulization-inductively coupled plasma-mass spectrometry to quantitatively measure Cu, Fe, and Zn concentrations in the retina and hippocampus samples obtained from human donors (i.e. AD and healthy controls), and in the amyloid precursor protein/presenilin 1 (APP/PS1) mouse model of AD and wild-type (WT) controls, aged 9 and 18 months. Our findings revealed significantly elevated Cu, Fe, and Zn levels in the retina (*P < .05, P < .01, and P < .001) and hippocampus (*P < .05, *P < .05, and *P < .05) of human AD samples compared to healthy controls. Conversely, APP/PS1 mouse models exhibited notably lower metal levels in the same regions compared to WT mice-Cu, Fe, and Zn levels in the retina (**P < .01, *P < .05, and *P < .05) and hippocampus (**P < .01, **P < .01, and *P < .05). The contrasting metal profiles in human and mouse samples, yet similar patterns within each species' retina and brain, suggest the retina mirrors cerebral metal dyshomoeostasis in AD. Our findings lay the groundwork for staging pre-AD pathophysiology through assessment of transition metal levels in the retina.
Collapse
Affiliation(s)
- Seyed Mostafa Hosseinpour Mashkani
- Institute for Biomedical Materials and Devices, School of Mathematical and Physical Sciences, Faculty of Science, University of Technology Sydney, Sydney, NSW 2007, Australia
| | - David P Bishop
- Hyphenated Mass Spectrometry Laboratory, School of Mathematical and Physical Sciences, University of Technology Sydney, Broadway, Sydney, NSW 2007, Australia
| | - Mika T Westerhausen
- Hyphenated Mass Spectrometry Laboratory, School of Mathematical and Physical Sciences, University of Technology Sydney, Broadway, Sydney, NSW 2007, Australia
| | - Paul A Adlard
- Synaptic Neurobiology Laboratory, The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Melbourne 3010, Australia
| | - S Mojtaba Golzan
- Vision Science Group (Orthoptics Discipline), Graduate School of Health, University of Technology Sydney, Sydney, NSW 2007, Australia
| |
Collapse
|
3
|
Vander Wall R, Basavarajappa D, Palanivel V, Sharma S, Gupta V, Klistoner A, Graham S, You Y. VEP Latency Delay Reflects Demyelination Beyond the Optic Nerve in the Cuprizone Model. Invest Ophthalmol Vis Sci 2024; 65:50. [PMID: 39576623 PMCID: PMC11587907 DOI: 10.1167/iovs.65.13.50] [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: 06/20/2024] [Accepted: 09/24/2024] [Indexed: 11/24/2024] Open
Abstract
Purpose Remyelination therapies are advancing for multiple sclerosis, focusing on visual pathways and using visual evoked potentials (VEPs) for de/remyelination processes. While the cuprizone (CZ) model and VEPs are core tools in preclinical trials, many overlook the posterior visual pathway. This study aimed to assess functional and structural changes across the murine visual pathway during de/remyelination. Methods One group of C57BL/6 mice underwent a CZ diet for 6 weeks to simulate demyelination, with a subset returning to a regular diet to induce remyelination. An additional group was fed a protracted CZ diet for 12 weeks to maintain chronic demyelination. Visual function was evaluated using electrophysiological recordings, including scotopic threshold responses (STRs) and electroretinograms (ERGs), with VEPs serving as a key biomarker for overall pathway health. Tissues from eyes, brains, and optic nerves (ONs) were collected at different time points for structural analysis. Results Our results demonstrated significant effects on VEPs, including increased N1 latencies and reduced amplitudes in the CZ mouse model. However, retinal function remained unaffected, as evidenced by unchanged STRs, ERGs, and retinal ganglion cell counts. Analysis of ONs revealed morphological changes, characterized by a significantly decreased axon diameter in the core region compared to the subpial region. Additionally, there was a significant increase in the g-ratio of the core region at 12 weeks CZ compared to controls. Immunofluorescence further demonstrated a decrease in myelin basic protein levels at 6 and 12 weeks in CZ animals. Interestingly, the dorsal lateral geniculate nucleus and primary visual cortex (V1) exhibited similar myelin changes, correlating with VEP latency alterations. Conclusions These data reveal that interpreting VEP latency solely as a marker for ON demyelination is incomplete. Previous preclinical studies have overlooked the posterior visual pathways, necessitating a broader interpretation of VEP latency to cover the entire visual pathway.
Collapse
Affiliation(s)
- Roshana Vander Wall
- Faculty of Medicine Health and Human Sciences, Macquarie University, North Ryde, NSW, Australia
| | - Devaraj Basavarajappa
- Faculty of Medicine Health and Human Sciences, Macquarie University, North Ryde, NSW, Australia
| | - Viswanthram Palanivel
- Faculty of Medicine Health and Human Sciences, Macquarie University, North Ryde, NSW, Australia
| | - Samridhi Sharma
- Garvan Institute of Medical Research, Sydney, NSW, Australia
| | - Vivek Gupta
- Faculty of Medicine Health and Human Sciences, Macquarie University, North Ryde, NSW, Australia
| | - Alexander Klistoner
- Faculty of Medicine Health and Human Sciences, Macquarie University, North Ryde, NSW, Australia
| | - Stuart Graham
- Faculty of Medicine Health and Human Sciences, Macquarie University, North Ryde, NSW, Australia
- Save Sight Institute, Sydney University, Sydney, NSW, Australia
| | - Yuyi You
- Faculty of Medicine Health and Human Sciences, Macquarie University, North Ryde, NSW, Australia
- Save Sight Institute, Sydney University, Sydney, NSW, Australia
| |
Collapse
|
4
|
Abbasi M, Gupta V, Chitranshi N, Moustardas P, Ranjbaran R, Graham SL. Molecular Mechanisms of Glaucoma Pathogenesis with Implications to Caveolin Adaptor Protein and Caveolin-Shp2 Axis. Aging Dis 2024; 15:2051-2068. [PMID: 37962455 PMCID: PMC11346403 DOI: 10.14336/ad.2023.1012] [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: 09/01/2023] [Accepted: 10/12/2023] [Indexed: 11/15/2023] Open
Abstract
Glaucoma is a common retinal disorder characterized by progressive optic nerve damage, resulting in visual impairment and potential blindness. Elevated intraocular pressure (IOP) is a major risk factor, but some patients still experience disease progression despite IOP-lowering treatments. Genome-wide association studies have linked variations in the Caveolin1/2 (CAV-1/2) gene loci to glaucoma risk. Cav-1, a key protein in caveolae membrane invaginations, is involved in signaling pathways and its absence impairs retinal function. Recent research suggests that Cav-1 is implicated in modulating the BDNF/TrkB signaling pathway in retinal ganglion cells, which plays a critical role in retinal ganglion cell (RGC) health and protection against apoptosis. Understanding the interplay between these proteins could shed light on glaucoma pathogenesis and provide potential therapeutic targets.
Collapse
Affiliation(s)
- Mojdeh Abbasi
- Macquarie Medical School, Faculty of Medicine, Health and Human Sciences, Macquarie University, North Ryde, Sydney, NSW 2109, Australia.
- Division of Ophthalmology, Department of Biomedical and Clinical Sciences, Linköping University, Linköping Sweden.
| | - Vivek Gupta
- Macquarie Medical School, Faculty of Medicine, Health and Human Sciences, Macquarie University, North Ryde, Sydney, NSW 2109, Australia.
| | - Nitin Chitranshi
- Macquarie Medical School, Faculty of Medicine, Health and Human Sciences, Macquarie University, North Ryde, Sydney, NSW 2109, Australia.
| | - Petros Moustardas
- Division of Ophthalmology, Department of Biomedical and Clinical Sciences, Linköping University, Linköping Sweden.
| | - Reza Ranjbaran
- Diagnostic Laboratory Sciences and Technology Research Center, School of Paramedical Sciences, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Stuart L. Graham
- Macquarie Medical School, Faculty of Medicine, Health and Human Sciences, Macquarie University, North Ryde, Sydney, NSW 2109, Australia.
| |
Collapse
|
5
|
Cai C, Gu C, Meng C, He S, Thashi L, Deji D, Zheng Z, Qiu Q. Therapeutic Effects of Metformin on Central Nervous System Diseases: A Focus on Protection of Neurovascular Unit. Pharm Res 2024; 41:1907-1920. [PMID: 39375240 DOI: 10.1007/s11095-024-03777-0] [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: 02/20/2024] [Accepted: 09/26/2024] [Indexed: 10/09/2024]
Abstract
Metformin is one of the most commonly used oral hypoglycemic drugs in clinical practice, with unique roles in neurodegeneration and vascular lesions. Neurodegeneration and vasculopathy coexist in many diseases and typically affect the neurovascular unit (NVU), a minimal structural and functional unit in the central nervous system. Its components interact with one another and are indispensable for maintaining tissue homeostasis. This review focuses on retinal (diabetic retinopathy, retinitis pigmentosa) and cerebral (ischemic stroke, Alzheimer's disease) diseases to explore the effects of metformin on the NVU. Metformin has a preliminarily confirmed therapeutic effect on the retinal NUV, affecting many of its components, such as photoreceptors (cones and rods), microglia, ganglion, Müller, and vascular endothelial cells. Since it rapidly penetrates the blood-brain barrier (BBB) and accumulates in the brain, metformin also has an extensively studied neuronal protective effect in neuronal diseases. Its mechanism affects various NVU components, including pericytes, astrocytes, microglia, and vascular endothelial cells, mainly serving to protect the BBB. Regulating the inflammatory response in NVU (especially neurons and microglia) may be the main mechanism of metformin in improving central nervous system related diseases. Metformin may be a potential drug for treating diseases associated with NVU deterioration, however, more trials are needed to validate its timing, duration, dose, clinical effects, and side effects.
Collapse
Affiliation(s)
- Chunyang Cai
- Department of Ophthalmology, Tong Ren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, No. 1111 Xianxia Road, Changning District, Shanghai, 200050, PR China
- Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, PR China
- National Clinical Research Center for Eye Diseases; Shanghai Key Laboratory of Ocular Fundus Diseases; Shanghai Engineering Center for Visual Science and Photomedicine; Shanghai engineering center for precise diagnosis and treatment of eye diseases, Shanghai, PR China
| | - Chufeng Gu
- Department of Ophthalmology, Fuzhou University Affiliated Provincial Hospital, Fujian Provincial Hospital, Shengli Clinical College of Fujian Medical University, Fuzhou, Fujian, PR China
| | - Chunren Meng
- Department of Ophthalmology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, PR China
| | - Shuai He
- Department of Ophthalmology, Tong Ren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, No. 1111 Xianxia Road, Changning District, Shanghai, 200050, PR China
| | - Lhamo Thashi
- Department of Ophthalmology, Shigatse People's Hospital, Shigatse, Tibet, PR China
| | - Draga Deji
- Department of Ophthalmology, Shigatse People's Hospital, Shigatse, Tibet, PR China
| | - Zhi Zheng
- Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, PR China.
- National Clinical Research Center for Eye Diseases; Shanghai Key Laboratory of Ocular Fundus Diseases; Shanghai Engineering Center for Visual Science and Photomedicine; Shanghai engineering center for precise diagnosis and treatment of eye diseases, Shanghai, PR China.
| | - Qinghua Qiu
- Department of Ophthalmology, Tong Ren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, No. 1111 Xianxia Road, Changning District, Shanghai, 200050, PR China.
- Department of Ophthalmology, Shigatse People's Hospital, Shigatse, Tibet, PR China.
- High Altitude Ocular Disease Research Center of People's Hospital of Shigatse City and Tongren Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, PR China.
| |
Collapse
|
6
|
Carrero L, Antequera D, Municio C, Carro E. Circadian rhythm disruption and retinal dysfunction: a bidirectional link in Alzheimer's disease? Neural Regen Res 2024; 19:1967-1972. [PMID: 38227523 DOI: 10.4103/1673-5374.390962] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Accepted: 11/07/2023] [Indexed: 01/17/2024] Open
Abstract
Dysfunction in circadian rhythms is a common occurrence in patients with Alzheimer's disease. A predominant function of the retina is circadian synchronization, carrying information to the brain through the retinohypothalamic tract, which projects to the suprachiasmatic nucleus. Notably, Alzheimer's disease hallmarks, including amyloid-β, are present in the retinas of Alzheimer's disease patients, followed/associated by structural and functional disturbances. However, the mechanistic link between circadian dysfunction and the pathological changes affecting the retina in Alzheimer's disease is not fully understood, although some studies point to the possibility that retinal dysfunction could be considered an early pathological process that directly modulates the circadian rhythm.
Collapse
Affiliation(s)
- Laura Carrero
- Group of Neurodegenerative Diseases, Hospital Universitario 12 de Octubre Research Institute (imas12), Madrid, Spain; Network Center for Biomedical Research in Neurodegenerative Diseases (CIBERNED), ISCIII, Madrid, Spain
- PhD Program in Neuroscience, Autonoma de Madrid University, Madrid, Spain
| | - Desireé Antequera
- Neurobiology of Alzheimer's Disease Unit, Functional Unit for Research into Chronic Diseases, Instituto de Salud Carlos III, Madrid, Spain; Network Centre for Biomedical Research in Neurodegenerative Diseases (CIBERNED), ISCIII, Madrid, Spain
| | - Cristina Municio
- Group of Neurodegenerative Diseases, Hospital Universitario 12 de Octubre Research Institute (imas12), Madrid, Spain; Network Center for Biomedical Research in Neurodegenerative Diseases (CIBERNED), ISCIII, Madrid, Spain
| | - Eva Carro
- Neurobiology of Alzheimer's Disease Unit, Functional Unit for Research into Chronic Diseases, Instituto de Salud Carlos III, Madrid, Spain; Network Centre for Biomedical Research in Neurodegenerative Diseases (CIBERNED), ISCIII, Madrid, Spain
| |
Collapse
|
7
|
Sánchez-Puebla L, de Hoz R, Salobrar-García E, Arias-Vázquez A, González-Jiménez M, Ramírez AI, Fernández-Albarral JA, Matamoros JA, Elvira-Hurtado L, Saido TC, Saito T, Nieto Vaquero C, Cuartero MI, Moro MA, Salazar JJ, López-Cuenca I, Ramírez JM. Age-Related Retinal Layer Thickness Changes Measured by OCT in APPNL-F/NL-F Mice: Implications for Alzheimer's Disease. Int J Mol Sci 2024; 25:8221. [PMID: 39125789 PMCID: PMC11312090 DOI: 10.3390/ijms25158221] [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: 05/29/2024] [Revised: 07/11/2024] [Accepted: 07/25/2024] [Indexed: 08/12/2024] Open
Abstract
In Alzheimer's disease (AD), transgenic mouse models have established links between abnormalities in the retina and those in the brain. APPNL-F/NL-F is a murine, humanized AD model that replicates several pathological features observed in patients with AD. Research has focused on obtaining quantitative parameters from optical coherence tomography (OCT) in AD. The aim of this study was to analyze, in a transversal case-control study using manual retinal segmentation via SD-OCT, the changes occurring in the retinal layers of the APPNL/F-NF/L AD model in comparison to C57BL/6J mice (WT) at 6, 9, 12, 15, 17, and 20 months of age. The analysis focused on retinal thickness in RNFL-GCL, IPL, INL, OPL, and ONL based on the Early Treatment Diabetic Retinopathy Study (ETDRS) sectors. Both APPNL-F/NL-F-model and WT animals exhibited thickness changes at the time points studied. While WT showed significant changes in INL, OPL, and ONL, the AD model showed changes in all retinal layers analyzed. The APPNL-F/NL-F displayed significant thickness variations in the analyzed layers except for the IPL compared to related WT. These thickness changes closely resembled those found in humans during preclinical stages, as well as during mild and moderate AD stages, making this AD model behave more similarly to the disease in humans.
Collapse
Affiliation(s)
- Lidia Sánchez-Puebla
- Ramon Castroviejo Institute for Ophthalmic Research, Complutense University of Madrid, 28040 Madrid, Spain; (L.S.-P.); (R.d.H.); (E.S.-G.); (A.A.-V.); (M.G.-J.); (A.I.R.); (J.A.F.-A.); (J.A.M.); (L.E.-H.); (J.J.S.)
- Health Research Institute of the Hospital Clínico San Carlos (IdISSC), 28040 Madrid, Spain
- Department of Immunology, Ophthalmology and ENT, School of Medicine, Complutense University of Madrid, 28040 Madrid, Spain
| | - Rosa de Hoz
- Ramon Castroviejo Institute for Ophthalmic Research, Complutense University of Madrid, 28040 Madrid, Spain; (L.S.-P.); (R.d.H.); (E.S.-G.); (A.A.-V.); (M.G.-J.); (A.I.R.); (J.A.F.-A.); (J.A.M.); (L.E.-H.); (J.J.S.)
- Health Research Institute of the Hospital Clínico San Carlos (IdISSC), 28040 Madrid, Spain
- Department of Immunology, Ophthalmology and ENT, Faculty of Optics and Optometry, Complutense University of Madrid, 28040 Madrid, Spain
| | - Elena Salobrar-García
- Ramon Castroviejo Institute for Ophthalmic Research, Complutense University of Madrid, 28040 Madrid, Spain; (L.S.-P.); (R.d.H.); (E.S.-G.); (A.A.-V.); (M.G.-J.); (A.I.R.); (J.A.F.-A.); (J.A.M.); (L.E.-H.); (J.J.S.)
- Health Research Institute of the Hospital Clínico San Carlos (IdISSC), 28040 Madrid, Spain
- Department of Immunology, Ophthalmology and ENT, Faculty of Optics and Optometry, Complutense University of Madrid, 28040 Madrid, Spain
| | - Alberto Arias-Vázquez
- Ramon Castroviejo Institute for Ophthalmic Research, Complutense University of Madrid, 28040 Madrid, Spain; (L.S.-P.); (R.d.H.); (E.S.-G.); (A.A.-V.); (M.G.-J.); (A.I.R.); (J.A.F.-A.); (J.A.M.); (L.E.-H.); (J.J.S.)
| | - María González-Jiménez
- Ramon Castroviejo Institute for Ophthalmic Research, Complutense University of Madrid, 28040 Madrid, Spain; (L.S.-P.); (R.d.H.); (E.S.-G.); (A.A.-V.); (M.G.-J.); (A.I.R.); (J.A.F.-A.); (J.A.M.); (L.E.-H.); (J.J.S.)
| | - Ana I. Ramírez
- Ramon Castroviejo Institute for Ophthalmic Research, Complutense University of Madrid, 28040 Madrid, Spain; (L.S.-P.); (R.d.H.); (E.S.-G.); (A.A.-V.); (M.G.-J.); (A.I.R.); (J.A.F.-A.); (J.A.M.); (L.E.-H.); (J.J.S.)
- Health Research Institute of the Hospital Clínico San Carlos (IdISSC), 28040 Madrid, Spain
- Department of Immunology, Ophthalmology and ENT, Faculty of Optics and Optometry, Complutense University of Madrid, 28040 Madrid, Spain
| | - José A. Fernández-Albarral
- Ramon Castroviejo Institute for Ophthalmic Research, Complutense University of Madrid, 28040 Madrid, Spain; (L.S.-P.); (R.d.H.); (E.S.-G.); (A.A.-V.); (M.G.-J.); (A.I.R.); (J.A.F.-A.); (J.A.M.); (L.E.-H.); (J.J.S.)
- Health Research Institute of the Hospital Clínico San Carlos (IdISSC), 28040 Madrid, Spain
- Department of Immunology, Ophthalmology and ENT, Faculty of Optics and Optometry, Complutense University of Madrid, 28040 Madrid, Spain
| | - José A. Matamoros
- Ramon Castroviejo Institute for Ophthalmic Research, Complutense University of Madrid, 28040 Madrid, Spain; (L.S.-P.); (R.d.H.); (E.S.-G.); (A.A.-V.); (M.G.-J.); (A.I.R.); (J.A.F.-A.); (J.A.M.); (L.E.-H.); (J.J.S.)
- Department of Immunology, Ophthalmology and ENT, Faculty of Optics and Optometry, Complutense University of Madrid, 28040 Madrid, Spain
| | - Lorena Elvira-Hurtado
- Ramon Castroviejo Institute for Ophthalmic Research, Complutense University of Madrid, 28040 Madrid, Spain; (L.S.-P.); (R.d.H.); (E.S.-G.); (A.A.-V.); (M.G.-J.); (A.I.R.); (J.A.F.-A.); (J.A.M.); (L.E.-H.); (J.J.S.)
| | - Takaomi C. Saido
- Brain Science Institute, RIKEN, Laboratory for Proteolytic Neuroscience, Wako 351-0198, Japan;
| | - Takashi Saito
- Institute of Brain Science, Faculty of Medical Sciences, Nagoya City University, Nagoya 467-8601, Japan;
| | - Carmen Nieto Vaquero
- Neurovascular Pathophysiology, Cardiovascular Risk Factor and Brain Function Programme, Centro Nacional de Investigaciones Cardiovasculares (CNIC), 28029 Madrid, Spain; (C.N.V.); (M.A.M.)
- Hospital 12 de Octubre Research Institute (i + 12), 28041 Madrid, Spain;
- University Institute for Research in Neurochemistry, Complutense University of Madrid (UCM), 28040 Madrid, Spain
| | - María I. Cuartero
- Hospital 12 de Octubre Research Institute (i + 12), 28041 Madrid, Spain;
- University Institute for Research in Neurochemistry, Complutense University of Madrid (UCM), 28040 Madrid, Spain
- Department of Pharmacology and Toxicology, Faculty of Medicine, Complutense University of Madrid (UCM), 28040 Madrid, Spain
| | - María A. Moro
- Neurovascular Pathophysiology, Cardiovascular Risk Factor and Brain Function Programme, Centro Nacional de Investigaciones Cardiovasculares (CNIC), 28029 Madrid, Spain; (C.N.V.); (M.A.M.)
| | - Juan J. Salazar
- Ramon Castroviejo Institute for Ophthalmic Research, Complutense University of Madrid, 28040 Madrid, Spain; (L.S.-P.); (R.d.H.); (E.S.-G.); (A.A.-V.); (M.G.-J.); (A.I.R.); (J.A.F.-A.); (J.A.M.); (L.E.-H.); (J.J.S.)
- Health Research Institute of the Hospital Clínico San Carlos (IdISSC), 28040 Madrid, Spain
- Department of Immunology, Ophthalmology and ENT, Faculty of Optics and Optometry, Complutense University of Madrid, 28040 Madrid, Spain
| | - Inés López-Cuenca
- Ramon Castroviejo Institute for Ophthalmic Research, Complutense University of Madrid, 28040 Madrid, Spain; (L.S.-P.); (R.d.H.); (E.S.-G.); (A.A.-V.); (M.G.-J.); (A.I.R.); (J.A.F.-A.); (J.A.M.); (L.E.-H.); (J.J.S.)
- Health Research Institute of the Hospital Clínico San Carlos (IdISSC), 28040 Madrid, Spain
- Department of Immunology, Ophthalmology and ENT, Faculty of Optics and Optometry, Complutense University of Madrid, 28040 Madrid, Spain
| | - José M. Ramírez
- Ramon Castroviejo Institute for Ophthalmic Research, Complutense University of Madrid, 28040 Madrid, Spain; (L.S.-P.); (R.d.H.); (E.S.-G.); (A.A.-V.); (M.G.-J.); (A.I.R.); (J.A.F.-A.); (J.A.M.); (L.E.-H.); (J.J.S.)
- Health Research Institute of the Hospital Clínico San Carlos (IdISSC), 28040 Madrid, Spain
- Department of Immunology, Ophthalmology and ENT, School of Medicine, Complutense University of Madrid, 28040 Madrid, Spain
| |
Collapse
|
8
|
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
|
9
|
Rinaldi M, Pezone A, Quadrini GI, Abbadessa G, Laezza MP, Passaro ML, Porcellini A, Costagliola C. Targeting shared pathways in tauopathies and age-related macular degeneration: implications for novel therapies. Front Aging Neurosci 2024; 16:1371745. [PMID: 38633983 PMCID: PMC11021713 DOI: 10.3389/fnagi.2024.1371745] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Accepted: 03/18/2024] [Indexed: 04/19/2024] Open
Abstract
The intricate parallels in structure and function between the human retina and the central nervous system designate the retina as a prospective avenue for understanding brain-related processes. This review extensively explores the shared physiopathological mechanisms connecting age-related macular degeneration (AMD) and proteinopathies, with a specific focus on tauopathies. The pivotal involvement of oxidative stress and cellular senescence emerges as key drivers of pathogenesis in both conditions. Uncovering these shared elements not only has the potential to enhance our understanding of intricate neurodegenerative diseases but also sets the stage for pioneering therapeutic approaches in AMD.
Collapse
Affiliation(s)
- Michele Rinaldi
- Department of Neurosciences, Reproductive Sciences and Dentistry, University of Naples Federico II, Naples, Italy
| | - Antonio Pezone
- Department of Biology, University of Naples Federico II, Naples, Italy
| | - Gaia Italia Quadrini
- Department of Neurosciences, Reproductive Sciences and Dentistry, University of Naples Federico II, Naples, Italy
| | - Gianmarco Abbadessa
- Division of Neurology, Department of Advanced Medical and Surgical Sciences, University of Campania Luigi Vanvitelli, Naples, Italy
| | - Maria Paola Laezza
- Department of Neurosciences, Reproductive Sciences and Dentistry, University of Naples Federico II, Naples, Italy
- Department of Medicine and Health Sciences “V. Tiberio”, University of Molise, Campobasso, Italy
| | - Maria Laura Passaro
- Department of Neurosciences, Reproductive Sciences and Dentistry, University of Naples Federico II, Naples, Italy
- Department of Medicine and Health Sciences “V. Tiberio”, University of Molise, Campobasso, Italy
| | | | - Ciro Costagliola
- Department of Neurosciences, Reproductive Sciences and Dentistry, University of Naples Federico II, Naples, Italy
| |
Collapse
|
10
|
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
|
11
|
Casciano F, Zauli E, Celeghini C, Caruso L, Gonelli A, Zauli G, Pignatelli A. Retinal Alterations Predict Early Prodromal Signs of Neurodegenerative Disease. Int J Mol Sci 2024; 25:1689. [PMID: 38338966 PMCID: PMC10855697 DOI: 10.3390/ijms25031689] [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: 12/21/2023] [Revised: 01/25/2024] [Accepted: 01/27/2024] [Indexed: 02/12/2024] Open
Abstract
Neurodegenerative diseases are an increasingly common group of diseases that occur late in life with a significant impact on personal, family, and economic life. Among these, Alzheimer's disease (AD) and Parkinson's disease (PD) are the major disorders that lead to mild to severe cognitive and physical impairment and dementia. Interestingly, those diseases may show onset of prodromal symptoms early after middle age. Commonly, the evaluation of these neurodegenerative diseases is based on the detection of biomarkers, where functional and structural magnetic resonance imaging (MRI) have shown a central role in revealing early or prodromal phases, although it can be expensive, time-consuming, and not always available. The aforementioned diseases have a common impact on the visual system due to the pathophysiological mechanisms shared between the eye and the brain. In Parkinson's disease, α-synuclein deposition in the retinal cells, as well as in dopaminergic neurons of the substantia nigra, alters the visual cortex and retinal function, resulting in modifications to the visual field. Similarly, the visual cortex is modified by the neurofibrillary tangles and neuritic amyloid β plaques typically seen in the Alzheimer's disease brain, and this may reflect the accumulation of these biomarkers in the retina during the early stages of the disease, as seen in postmortem retinas of AD patients. In this light, the ophthalmic evaluation of retinal neurodegeneration could become a cost-effective method for the early diagnosis of those diseases, overcoming the limitations of functional and structural imaging of the deep brain. This analysis is commonly used in ophthalmic practice, and interest in it has risen in recent years. This review will discuss the relationship between Alzheimer's disease and Parkinson's disease with retinal degeneration, highlighting how retinal analysis may represent a noninvasive and straightforward method for the early diagnosis of these neurodegenerative diseases.
Collapse
Affiliation(s)
- Fabio Casciano
- Department of Translational Medicine and LTTA Centre, University of Ferrara, 44121 Ferrara, Italy
| | - Enrico Zauli
- Department of Translational Medicine, University of Ferrara, 44121 Ferrara, Italy
| | - Claudio Celeghini
- Department of Translational Medicine, University of Ferrara, 44121 Ferrara, Italy
| | - Lorenzo Caruso
- Department of Environment and Prevention Sciences, University of Ferrara, 44121 Ferrara, Italy
| | - Arianna Gonelli
- Department of Translational Medicine, University of Ferrara, 44121 Ferrara, Italy
| | - Giorgio Zauli
- Research Department, King Khaled Eye Specialistic Hospital, Riyadh 12329, Saudi Arabia
| | - Angela Pignatelli
- Department of Neuroscience and Rehabilitation, University of Ferrara, 44124 Ferrara, Italy
| |
Collapse
|
12
|
Huh MG, Kim YK, Lee J, Shin YI, Lee YJ, Choe S, Kim DW, Jeong Y, Jeoung JW, Park KH. Relative Risks for Dementia among Individuals with Glaucoma: A Meta-Analysis of Observational Cohort Studies. KOREAN JOURNAL OF OPHTHALMOLOGY 2023; 37:490-500. [PMID: 37899286 PMCID: PMC10721395 DOI: 10.3341/kjo.2023.0059] [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: 05/25/2023] [Revised: 09/27/2023] [Accepted: 10/05/2023] [Indexed: 10/31/2023] Open
Abstract
PURPOSE To investigate the relative risks (RRs) for dementia among individuals with glaucoma. METHODS We conducted a search of PubMed, Web of Science, Scopus, and Cochrane databases for observational cohort studies examining the association between glaucoma and dementia until March 2023. Two authors independently screened all titles and abstracts according to predefined inclusion and exclusion criteria. Pooled RR and 95% confidence intervals (CIs) were generated using random-effect models. RESULTS The meta-analysis included 18 cohort studies conducted in eight countries and involving 4,975,325 individuals. The pooled RR for the association between glaucoma and all-cause dementia was 1.314 (95% CI, 1.099-1.572; I2 = 95%). The pooled RRs for the associations of open-angle glaucoma with Alzheimer dementia and Parkinson disease were 1.287 (95% CI, 1.007-1.646; I2 = 96%) and 1.233 (95% CI, 0.677-2.243; I2 = 73%), respectively. The pooled RRs for the associations of angle-closure glaucoma with all-cause dementia and Alzheimer dementia were 0.978 (95% CI, 0.750-1.277; I2 = 17%) and 0.838 (95% CI, 0.421-1.669; I2 = 16%), respectively. No evidence of publication bias was detected in the Begg-Mazumdar adjusted rank correlation test (p = 0.47). CONCLUSIONS Based on current observational cohort studies, there is evidence supporting that glaucoma is a risk factor for dementia in the adult population.
Collapse
Affiliation(s)
- Min Gu Huh
- Department of Ophthalmology, Seoul National University Hospital, Seoul,
Korea
- Department of Ophthalmology, Seoul National University College of Medicine, Seoul,
Korea
| | - Young Kook Kim
- Department of Ophthalmology, Seoul National University Hospital, Seoul,
Korea
- Department of Ophthalmology, Seoul National University College of Medicine, Seoul,
Korea
- EyeLight Data Science Laboratory, Seoul National University College of Medicine, Seoul,
Korea
| | - Jaekyoung Lee
- Department of Ophthalmology, Seoul National University Hospital, Seoul,
Korea
- Department of Ophthalmology, Seoul National University College of Medicine, Seoul,
Korea
| | - Young In Shin
- Department of Ophthalmology, Seoul National University Hospital, Seoul,
Korea
- Department of Ophthalmology, Seoul National University College of Medicine, Seoul,
Korea
| | - Yun Jeong Lee
- Department of Ophthalmology, Seoul National University College of Medicine, Seoul,
Korea
- Department of Ophthalmology, Chungnam National University Hospital, Daejeon,
Korea
| | - Sooyeon Choe
- Department of Ophthalmology, Seoul National University College of Medicine, Seoul,
Korea
- Department of Ophthalmology, Chungnam National University Hospital, Daejeon,
Korea
| | - Dai Woo Kim
- Department of Ophthalmology, Kyungpook National University Hospital, Kyungpook National University School of Medicine, Daegu,
Korea
| | - Yoon Jeong
- Department of Ophthalmology, Seoul National University Hospital, Seoul,
Korea
- Department of Ophthalmology, Seoul National University College of Medicine, Seoul,
Korea
| | - Jin Wook Jeoung
- Department of Ophthalmology, Seoul National University Hospital, Seoul,
Korea
- Department of Ophthalmology, Seoul National University College of Medicine, Seoul,
Korea
| | - Ki Ho Park
- Department of Ophthalmology, Seoul National University Hospital, Seoul,
Korea
- Department of Ophthalmology, Seoul National University College of Medicine, Seoul,
Korea
| |
Collapse
|
13
|
Basavarajappa D, Galindo-Romero C, Gupta V, Agudo-Barriuso M, Gupta VB, Graham SL, Chitranshi N. Signalling pathways and cell death mechanisms in glaucoma: Insights into the molecular pathophysiology. Mol Aspects Med 2023; 94:101216. [PMID: 37856930 DOI: 10.1016/j.mam.2023.101216] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Revised: 09/25/2023] [Accepted: 10/02/2023] [Indexed: 10/21/2023]
Abstract
Glaucoma is a complex multifactorial eye disease manifesting in retinal ganglion cell (RGC) death and optic nerve degeneration, ultimately causing irreversible vision loss. Research in recent years has significantly enhanced our understanding of RGC degenerative mechanisms in glaucoma. It is evident that high intraocular pressure (IOP) is not the only contributing factor to glaucoma pathogenesis. The equilibrium of pro-survival and pro-death signalling pathways in the retina strongly influences the function and survival of RGCs and optic nerve axons in glaucoma. Molecular evidence from human retinal tissue analysis and a range of experimental models of glaucoma have significantly contributed to unravelling these mechanisms. Accumulating evidence reveals a wide range of molecular signalling pathways that can operate -either alone or via intricate networks - to induce neurodegeneration. The roles of several molecules, including neurotrophins, interplay of intracellular kinases and phosphates, caveolae and adapter proteins, serine proteases and their inhibitors, nuclear receptors, amyloid beta and tau, and how their dysfunction affects retinal neurons are discussed in this review. We further underscore how anatomical alterations in various animal models exhibiting RGC degeneration and susceptibility to glaucoma-related neuronal damage have helped to characterise molecular mechanisms in glaucoma. In addition, we also present different regulated cell death pathways that play a critical role in RGC degeneration in glaucoma.
Collapse
Affiliation(s)
- Devaraj Basavarajappa
- Macquarie Medical School, Faculty of Medicine and Health Sciences, Macquarie University, Sydney, Australia.
| | - Caridad Galindo-Romero
- Experimental Ophthalmology Group, Instituto Murciano de Investigación Biosanitaria Virgen de la Arrixaca (IMIB-Arrixaca) & Ophthalmology Department, Universidad de Murcia, Murcia, Spain
| | - Vivek Gupta
- Macquarie Medical School, Faculty of Medicine and Health Sciences, Macquarie University, Sydney, Australia
| | - Marta Agudo-Barriuso
- Experimental Ophthalmology Group, Instituto Murciano de Investigación Biosanitaria Virgen de la Arrixaca (IMIB-Arrixaca) & Ophthalmology Department, Universidad de Murcia, Murcia, Spain
| | - Veer B Gupta
- School of Medicine, Deakin University, Melbourne, VIC, Australia
| | - Stuart L Graham
- Macquarie Medical School, Faculty of Medicine and Health Sciences, Macquarie University, Sydney, Australia
| | - Nitin Chitranshi
- Macquarie Medical School, Faculty of Medicine and Health Sciences, Macquarie University, Sydney, Australia.
| |
Collapse
|
14
|
Wang MD, Zhang S, Liu XY, Wang PP, Zhu YF, Zhu JR, Lv CS, Li SY, Liu SF, Wen L. Salvianolic acid B ameliorates retinal deficits in an early-stage Alzheimer's disease mouse model through downregulating BACE1 and Aβ generation. Acta Pharmacol Sin 2023; 44:2151-2168. [PMID: 37420104 PMCID: PMC10618533 DOI: 10.1038/s41401-023-01125-3] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Accepted: 06/08/2023] [Indexed: 07/09/2023]
Abstract
Alzheimer's disease (AD) is a neurodegenerative disease with subtle onset, early diagnosis remains challenging. Accumulating evidence suggests that the emergence of retinal damage in AD precedes cognitive impairment, and may serve as a critical indicator for early diagnosis and disease progression. Salvianolic acid B (Sal B), a bioactive compound isolated from the traditional Chinese medicinal herb Salvia miltiorrhiza, has been shown promise in treating neurodegenerative diseases, such as AD and Parkinson's disease. In this study we investigated the therapeutic effects of Sal B on retinopathy in early-stage AD. One-month-old transgenic mice carrying five familial AD mutations (5×FAD) were treated with Sal B (20 mg·kg-1·d-1, i.g.) for 3 months. At the end of treatment, retinal function and structure were assessed, cognitive function was evaluated in Morris water maze test. We showed that 4-month-old 5×FAD mice displayed distinct structural and functional deficits in the retinas, which were significantly ameliorated by Sal B treatment. In contrast, untreated, 4-month-old 5×FAD mice did not exhibit cognitive impairment compared to wild-type mice. In SH-SY5Y-APP751 cells, we demonstrated that Sal B (10 μM) significantly decreased BACE1 expression and sorting into the Golgi apparatus, thereby reducing Aβ generation by inhibiting the β-cleavage of APP. Moreover, we found that Sal B effectively attenuated microglial activation and the associated inflammatory cytokine release induced by Aβ plaque deposition in the retinas of 5×FAD mice. Taken together, our results demonstrate that functional impairments in the retina occur before cognitive decline, suggesting that the retina is a valuable reference for early diagnosis of AD. Sal B ameliorates retinal deficits by regulating APP processing and Aβ generation in early AD, which is a potential therapeutic intervention for early AD treatment.
Collapse
Affiliation(s)
- Meng-Dan Wang
- State Key Laboratory of Cellular Stress Biology, Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, Xiang'an Hospital, School of Medicine, Xiamen University, Xiamen, 361102, China
- Xiamen Key Laboratory for TCM Dampness Disease, Neurology & Immunology Research, Department of Traditional Chinese Medicine, Xiang'an Hospital, School of Medicine, Xiamen University, Xiamen, 361102, China
| | - Shuo Zhang
- State Key Laboratory of Cellular Stress Biology, Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, Xiang'an Hospital, School of Medicine, Xiamen University, Xiamen, 361102, China
- Xiamen Key Laboratory for TCM Dampness Disease, Neurology & Immunology Research, Department of Traditional Chinese Medicine, Xiang'an Hospital, School of Medicine, Xiamen University, Xiamen, 361102, China
| | - Xing-Yang Liu
- State Key Laboratory of Cellular Stress Biology, Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, Xiang'an Hospital, School of Medicine, Xiamen University, Xiamen, 361102, China
- Xiamen Key Laboratory for TCM Dampness Disease, Neurology & Immunology Research, Department of Traditional Chinese Medicine, Xiang'an Hospital, School of Medicine, Xiamen University, Xiamen, 361102, China
| | - Pan-Pan Wang
- State Key Laboratory of Cellular Stress Biology, Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, Xiang'an Hospital, School of Medicine, Xiamen University, Xiamen, 361102, China
- Xiamen Key Laboratory for TCM Dampness Disease, Neurology & Immunology Research, Department of Traditional Chinese Medicine, Xiang'an Hospital, School of Medicine, Xiamen University, Xiamen, 361102, China
| | - Yi-Fan Zhu
- State Key Laboratory of Cellular Stress Biology, Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, Xiang'an Hospital, School of Medicine, Xiamen University, Xiamen, 361102, China
- Xiamen Key Laboratory for TCM Dampness Disease, Neurology & Immunology Research, Department of Traditional Chinese Medicine, Xiang'an Hospital, School of Medicine, Xiamen University, Xiamen, 361102, China
| | - Jun-Rong Zhu
- State Key Laboratory of Cellular Stress Biology, Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, Xiang'an Hospital, School of Medicine, Xiamen University, Xiamen, 361102, China
- Xiamen Key Laboratory for TCM Dampness Disease, Neurology & Immunology Research, Department of Traditional Chinese Medicine, Xiang'an Hospital, School of Medicine, Xiamen University, Xiamen, 361102, China
| | - Chong-Shan Lv
- State Key Laboratory of Cellular Stress Biology, Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, Xiang'an Hospital, School of Medicine, Xiamen University, Xiamen, 361102, China
- Xiamen Key Laboratory for TCM Dampness Disease, Neurology & Immunology Research, Department of Traditional Chinese Medicine, Xiang'an Hospital, School of Medicine, Xiamen University, Xiamen, 361102, China
| | - Shi-Ying Li
- Eye Institute of Xiamen University, Department of Ophthalmology, Xiang'an Hospital, School of Medicine, Xiamen University, Xiamen, 361102, China.
| | - Sui-Feng Liu
- Zhongshan Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, 361000, China.
| | - Lei Wen
- State Key Laboratory of Cellular Stress Biology, Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, Xiang'an Hospital, School of Medicine, Xiamen University, Xiamen, 361102, China.
- Xiamen Key Laboratory for TCM Dampness Disease, Neurology & Immunology Research, Department of Traditional Chinese Medicine, Xiang'an Hospital, School of Medicine, Xiamen University, Xiamen, 361102, China.
| |
Collapse
|
15
|
Martucci A, Di Giuliano F, Minosse S, Pocobelli G, Nucci C, Garaci F. MRI and Clinical Biomarkers Overlap between Glaucoma and Alzheimer's Disease. Int J Mol Sci 2023; 24:14932. [PMID: 37834380 PMCID: PMC10573932 DOI: 10.3390/ijms241914932] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2023] [Accepted: 09/22/2023] [Indexed: 10/15/2023] Open
Abstract
Glaucoma is the leading cause of blindness worldwide. It is classically associated with structural and functional changes in the optic nerve head and retinal nerve fiber layer, but the damage is not limited to the eye. The involvement of the central visual pathways and disruption of brain network organization have been reported using advanced neuroimaging techniques. The brain structural changes at the level of the areas implied in processing visual information could justify the discrepancy between signs and symptoms and underlie the analogy of this disease with neurodegenerative dementias, such as Alzheimer's disease, and with the complex group of pathologies commonly referred to as "disconnection syndromes." This review aims to summarize the current state of the art on the use of advanced neuroimaging techniques in glaucoma and Alzheimer's disease, highlighting the emerging biomarkers shared by both diseases.
Collapse
Affiliation(s)
- Alessio Martucci
- Ophthalmology Unit, Department of Experimental Medicine, University of Rome “Tor Vergata”, 00133 Rome, Italy; (A.M.); (G.P.)
| | - Francesca Di Giuliano
- Neuroradiology Unit, Department of Biomedicine and Prevention, University of Rome “Tor Vergata”, 00133 Rome, Italy;
| | - Silvia Minosse
- Diagnostic Imaging Unit, Department of Biomedicine and Prevention, University of Rome “Tor Vergata”, 00133 Rome, Italy; (S.M.); (F.G.)
| | - Giulio Pocobelli
- Ophthalmology Unit, Department of Experimental Medicine, University of Rome “Tor Vergata”, 00133 Rome, Italy; (A.M.); (G.P.)
| | - Carlo Nucci
- Ophthalmology Unit, Department of Experimental Medicine, University of Rome “Tor Vergata”, 00133 Rome, Italy; (A.M.); (G.P.)
| | - Francesco Garaci
- Diagnostic Imaging Unit, Department of Biomedicine and Prevention, University of Rome “Tor Vergata”, 00133 Rome, Italy; (S.M.); (F.G.)
- San Raffaele Cassino, 03043 Frosinone, Italy
| |
Collapse
|
16
|
Chitranshi N, Rajput R, Godinez A, Pushpitha K, Mirzaei M, Basavarajappa D, Gupta V, Sharma S, You Y, Galliciotti G, Salekdeh GH, Baker MS, Graham SL, Gupta VK. Neuroserpin gene therapy inhibits retinal ganglion cell apoptosis and promotes functional preservation in glaucoma. Mol Ther 2023; 31:2056-2076. [PMID: 36905120 PMCID: PMC10362384 DOI: 10.1016/j.ymthe.2023.03.008] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Revised: 01/27/2023] [Accepted: 03/07/2023] [Indexed: 03/12/2023] Open
Abstract
Our research has proven that the inhibitory activity of the serine protease inhibitor neuroserpin (NS) is impaired because of its oxidation deactivation in glaucoma. Using genetic NS knockout (NS-/-) and NS overexpression (NS+/+ Tg) animal models and antibody-based neutralization approaches, we demonstrate that NS loss is detrimental to retinal structure and function. NS ablation was associated with perturbations in autophagy and microglial and synaptic markers, leading to significantly enhanced IBA1, PSD95, beclin-1, and LC3-II/LC3-I ratio and reduced phosphorylated neurofilament heavy chain (pNFH) levels. On the other hand, NS upregulation promoted retinal ganglion cell (RGC) survival in wild-type and NS-/- glaucomatous mice and increased pNFH expression. NS+/+Tg mice demonstrated decreased PSD95, beclin-1, LC3-II/LC3-I ratio, and IBA1 following glaucoma induction, highlighting its protective role. We generated a novel reactive site NS variant (M363R-NS) resistant to oxidative deactivation. Intravitreal administration of M363R-NS was observed to rescue the RGC degenerative phenotype in NS-/- mice. These findings demonstrate that NS dysfunction plays a key role in the glaucoma inner retinal degenerative phenotype and that modulating NS imparts significant protection to the retina. NS upregulation protected RGC function and restored biochemical networks associated with autophagy and microglial and synaptic function in glaucoma.
Collapse
Affiliation(s)
- Nitin Chitranshi
- Faculty of Medicine and Health Sciences, Macquarie University, Sydney, NSW, Australia.
| | - Rashi Rajput
- Faculty of Medicine and Health Sciences, Macquarie University, Sydney, NSW, Australia
| | - Angela Godinez
- Faculty of Medicine and Health Sciences, Macquarie University, Sydney, NSW, Australia
| | - Kanishka Pushpitha
- Faculty of Medicine and Health Sciences, Macquarie University, Sydney, NSW, Australia
| | - Mehdi Mirzaei
- Faculty of Medicine and Health Sciences, Macquarie University, Sydney, NSW, Australia
| | - Devaraj Basavarajappa
- Faculty of Medicine and Health Sciences, Macquarie University, Sydney, NSW, Australia
| | - Veer Gupta
- School of Medicine, Deakin University, Melbourne, VIC, Australia
| | - Samridhi Sharma
- Faculty of Medicine and Health Sciences, Macquarie University, Sydney, NSW, Australia
| | - Yuyi You
- Faculty of Medicine and Health Sciences, Macquarie University, Sydney, NSW, Australia
| | - Giovanna Galliciotti
- Institute of Neuropathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Ghasem H Salekdeh
- School of Natural Sciences, Macquarie University, Macquarie Park, NSW 2109, Australia
| | - Mark S Baker
- Faculty of Medicine and Health Sciences, Macquarie University, Sydney, NSW, Australia
| | - Stuart L Graham
- Faculty of Medicine and Health Sciences, Macquarie University, Sydney, NSW, Australia
| | - Vivek K Gupta
- Faculty of Medicine and Health Sciences, Macquarie University, Sydney, NSW, Australia.
| |
Collapse
|
17
|
Maran JJ, Adesina MM, Green CR, Kwakowsky A, Mugisho OO. The central role of the NLRP3 inflammasome pathway in the pathogenesis of age-related diseases in the eye and the brain. Ageing Res Rev 2023; 88:101954. [PMID: 37187367 DOI: 10.1016/j.arr.2023.101954] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Revised: 05/08/2023] [Accepted: 05/12/2023] [Indexed: 05/17/2023]
Abstract
With increasing age, structural changes occur in the eye and brain. Neuronal death, inflammation, vascular disruption, and microglial activation are among many of the pathological changes that can occur during ageing. Furthermore, ageing individuals are at increased risk of developing neurodegenerative diseases in these organs, including Alzheimer's disease (AD), Parkinson's disease (PD), glaucoma and age-related macular degeneration (AMD). Although these diseases pose a significant global public health burden, current treatment options focus on slowing disease progression and symptomatic control rather than targeting underlying causes. Interestingly, recent investigations have proposed an analogous aetiology between age-related diseases in the eye and brain, where a process of chronic low-grade inflammation is implicated. Studies have suggested that patients with AD or PD are also associated with an increased risk of AMD, glaucoma, and cataracts. Moreover, pathognomonic amyloid-β and α-synuclein aggregates, which accumulate in AD and PD, respectively, can be found in ocular parenchyma. In terms of a common molecular pathway that underpins these diseases, the nucleotide-binding domain, leucine-rich-containing family, and pyrin domain-containing-3 (NLRP3) inflammasome is thought to play a vital role in the manifestation of all these diseases. This review summarises the current evidence regarding cellular and molecular changes in the brain and eye with age, similarities between ocular and cerebral age-related diseases, and the role of the NLRP3 inflammasome as a critical mediator of disease propagation in the eye and the brain during ageing.
Collapse
Affiliation(s)
- Jack J Maran
- Buchanan Ocular Therapeutics Unit, Department of Ophthalmology and the New Zealand National Eye Centre, University of Auckland, New Zealand
| | - Moradeke M Adesina
- Buchanan Ocular Therapeutics Unit, Department of Ophthalmology and the New Zealand National Eye Centre, University of Auckland, New Zealand
| | - Colin R Green
- Department of Ophthalmology and the New Zealand National Eye Centre, Faculty of Medical and Health Sciences, University of Auckland, New Zealand
| | - Andrea Kwakowsky
- Pharmacology and Therapeutics, School of Medicine, Galway Neuroscience Centre, University of Galway, Galway, Ireland
| | - Odunayo O Mugisho
- Buchanan Ocular Therapeutics Unit, Department of Ophthalmology and the New Zealand National Eye Centre, University of Auckland, New Zealand.
| |
Collapse
|
18
|
Maran JJ, Adesina MM, Green CR, Kwakowsky A, Mugisho OO. Retinal inner nuclear layer thickness in the diagnosis of cognitive impairment explored using a C57BL/6J mouse model. Sci Rep 2023; 13:8150. [PMID: 37208533 DOI: 10.1038/s41598-023-35229-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Accepted: 05/15/2023] [Indexed: 05/21/2023] Open
Abstract
Major neurocognitive disorder (NCD) affects over 55 million people worldwide and is characterized by cognitive impairment (CI). This study aimed to develop a non-invasive diagnostic test for CI based upon retinal thickness measurements explored in a mouse model. Discrimination indices and retinal layer thickness of healthy C57BL/6J mice were quantified through a novel object recognition test (NORT) and ocular coherence tomography (OCT), respectively. Based on criteria from the Diagnostic and statistical manual of mental disorders 5th ed. (DSM-V), a diagnostic test was generated by transforming data into rolling monthly averages and categorizing mice into those with and without CI and those with a high or low decline in retinal layer thickness. Only inner nuclear layer thickness had a statistically significant relationship with discrimination indices. Furthermore, our diagnostic test was 85.71% sensitive and 100% specific for diagnosing CI, with a positive predictive value of 100%. These findings have potential clinical implications for the early diagnosis of CI in NCD. However, further investigation in comorbid mice and humans is warranted.
Collapse
Affiliation(s)
- Jack J Maran
- Buchanan Ocular Therapeutics Unit, Department of Ophthalmology and The New Zealand National Eye Centre, University of Auckland, Auckland, New Zealand
| | - Moradeke M Adesina
- Department of Ophthalmology and The New Zealand National Eye Centre, Faculty of Medical and Health Sciences, University of Auckland, Private Bag 92019, Auckland, New Zealand
| | - Colin R Green
- Department of Ophthalmology and The New Zealand National Eye Centre, Faculty of Medical and Health Sciences, University of Auckland, Private Bag 92019, Auckland, New Zealand
| | - Andrea Kwakowsky
- Centre for Brain Research, University of Auckland, Auckland, New Zealand
- Department of Anatomy and Medical Imaging, University of Auckland, Auckland, New Zealand
- Pharmacology and Therapeutics, School of Medicine, Galway Neuroscience Centre, University of Galway, Galway, Ireland
| | - Odunayo O Mugisho
- Buchanan Ocular Therapeutics Unit, Department of Ophthalmology and The New Zealand National Eye Centre, University of Auckland, Auckland, New Zealand.
- Department of Ophthalmology and The New Zealand National Eye Centre, Faculty of Medical and Health Sciences, University of Auckland, Private Bag 92019, Auckland, New Zealand.
| |
Collapse
|
19
|
Hosseinpour Mashkani SM, Bishop DP, Raoufi-Rad N, Adlard PA, Shimoni O, Golzan SM. Distribution of Copper, Iron, and Zinc in the Retina, Hippocampus, and Cortex of the Transgenic APP/PS1 Mouse Model of Alzheimer's Disease. Cells 2023; 12:cells12081144. [PMID: 37190053 DOI: 10.3390/cells12081144] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Revised: 03/30/2023] [Accepted: 03/30/2023] [Indexed: 05/17/2023] Open
Abstract
A mis-metabolism of transition metals (i.e., copper, iron, and zinc) in the brain has been recognised as a precursor event for aggregation of Amyloid-β plaques, a pathological hallmark of Alzheimer's disease (AD). However, imaging cerebral transition metals in vivo can be extremely challenging. As the retina is a known accessible extension of the central nervous system, we examined whether changes in the hippocampus and cortex metal load are also mirrored in the retina. Laser ablation inductively coupled plasma-mass spectrometry (LA-ICP-MS) was used to visualise and quantify the anatomical distribution and load of Cu, Fe, and Zn in the hippocampus, cortex, and retina of 9-month-old Amyloid Precursor Protein/Presenilin 1 (APP/PS1, n = 10) and Wild Type (WT, n = 10) mice. Our results show a similar metal load trend between the retina and the brain, with the WT mice displaying significantly higher concentrations of Cu, Fe, and Zn in the hippocampus (p < 0.05, p < 0.0001, p < 0.01), cortex (p < 0.05, p = 0.18, p < 0.0001) and the retina (p < 0.001, p = 0.01, p < 0.01) compared with the APP/PS1 mice. Our findings demonstrate that dysfunction of the cerebral transition metals in AD is also extended to the retina. This could lay the groundwork for future studies on the assessment of transition metal load in the retina in the context of early AD.
Collapse
Affiliation(s)
- Seyed Mostafa Hosseinpour Mashkani
- Institute for Biomedical Materials and Devices, School of Mathematical and Physical Sciences, Faculty of Science, University of Technology Sydney, 15 Broadway, Sydney, NSW 2007, Australia
| | - David P Bishop
- Hyphenated Mass Spectrometry Laboratory (HyMaS), School of Mathematical and Physical Sciences, Faculty of Science, University of Technology Sydney, 15 Broadway, Sydney, NSW 2007, Australia
| | - Newsha Raoufi-Rad
- Institute for Biomedical Materials and Devices, School of Mathematical and Physical Sciences, Faculty of Science, University of Technology Sydney, 15 Broadway, Sydney, NSW 2007, Australia
| | - Paul A Adlard
- Synaptic Neurobiology Laboratory, The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Melbourne, VIC 3000, Australia
| | - Olga Shimoni
- Institute for Biomedical Materials and Devices, School of Mathematical and Physical Sciences, Faculty of Science, University of Technology Sydney, 15 Broadway, Sydney, NSW 2007, Australia
| | - S Mojtaba Golzan
- Vision Science Group, Graduate School of Health (GSH), University of Technology Sydney, 15 Broadway, Sydney, NSW 2007, Australia
| |
Collapse
|
20
|
Fernández-Albarral JA, Salobrar-García E, Matamoros JA, Fernández-Mendívil C, del Sastre E, Chen L, de Hoz R, López-Cuenca I, Sánchez-Puebla L, Ramírez JM, Salazar JJ, Lopez MG, Ramírez AI. Microglial Hemoxygenase-1 Deletion Reduces Inflammation in the Retina of Old Mice with Tauopathy. Antioxidants (Basel) 2022; 11:2151. [PMID: 36358522 PMCID: PMC9686584 DOI: 10.3390/antiox11112151] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Revised: 10/27/2022] [Accepted: 10/27/2022] [Indexed: 09/26/2023] Open
Abstract
Tauopathies such as Alzheimer's disease are characterized by the accumulation of neurotoxic aggregates of tau protein. With aging and, especially, in Alzheimer's patients, the inducible enzyme heme oxygenase 1 (HO-1) progressively increases in microglia, causing iron accumulation, neuroinflammation, and neurodegeneration. The retina is an organ that can be readily accessed and can reflect changes that occur in the brain. In this context, we evaluated how the lack of microglial HO-1, using mice that do not express HO-1 in microglia (HMO-KO), impacts retinal macro and microgliosis of aged subjects (18 months old mice) subjected to tauopathy by intrahippocampal delivery of AAV-hTauP301L (TAU). Our results show that although tauopathy, measured as anti-TAUY9 and anti-AT8 positive immunostaining, was not observed in the retina of WT-TAU or HMO-KO+TAU mice, a morphometric study of retinal microglia and macroglia showed significant retinal changes in the TAU group compared to the WT group, such as: (i) increased number of activated microglia, (ii) retraction of microglial processes, (iii) increased number of CD68+ microglia, and (iv) increased retinal area occupied by GFAP (AROA) and C3 (AROC3). This retinal inflammatory profile was reduced in HMO-KO+TAU mice. Conclusion: Reduction of microglial HO-1 could be beneficial to prevent tauopathy-induced neuroinflammation.
Collapse
Affiliation(s)
- José A. Fernández-Albarral
- Instituto de Investigaciones Oftalmológicas Ramón Castroviejo, Grupo UCM 920105, IdISSC, Universidad Complutense de Madrid, 28040 Madrid, Spain
| | - Elena Salobrar-García
- Instituto de Investigaciones Oftalmológicas Ramón Castroviejo, Grupo UCM 920105, IdISSC, Universidad Complutense de Madrid, 28040 Madrid, Spain
- Facultad de Óptica y Optometría, Departamento de Inmunología, Oftalmología y ORL, Universidad Complutense de Madrid, 28037 Madrid, Spain
| | - José A. Matamoros
- Instituto de Investigaciones Oftalmológicas Ramón Castroviejo, Grupo UCM 920105, IdISSC, Universidad Complutense de Madrid, 28040 Madrid, Spain
- Facultad de Óptica y Optometría, Departamento de Inmunología, Oftalmología y ORL, Universidad Complutense de Madrid, 28037 Madrid, Spain
| | - Cristina Fernández-Mendívil
- Instituto Teófilo Hernando for Drug Discovery, Department of Pharmacology, School of Medicine, Universidad Autónoma Madrid, 28029 Madrid, Spain
- Instituto de Investigación Sanitario (IIS-IP), Hospital Universitario de La Princesa, 28006 Madrid, Spain
| | - Eric del Sastre
- Instituto Teófilo Hernando for Drug Discovery, Department of Pharmacology, School of Medicine, Universidad Autónoma Madrid, 28029 Madrid, Spain
- Instituto de Investigación Sanitario (IIS-IP), Hospital Universitario de La Princesa, 28006 Madrid, Spain
| | - Lejing Chen
- Instituto de Investigaciones Oftalmológicas Ramón Castroviejo, Grupo UCM 920105, IdISSC, Universidad Complutense de Madrid, 28040 Madrid, Spain
| | - Rosa de Hoz
- Instituto de Investigaciones Oftalmológicas Ramón Castroviejo, Grupo UCM 920105, IdISSC, Universidad Complutense de Madrid, 28040 Madrid, Spain
- Facultad de Óptica y Optometría, Departamento de Inmunología, Oftalmología y ORL, Universidad Complutense de Madrid, 28037 Madrid, Spain
| | - Inés López-Cuenca
- Instituto de Investigaciones Oftalmológicas Ramón Castroviejo, Grupo UCM 920105, IdISSC, Universidad Complutense de Madrid, 28040 Madrid, Spain
| | - Lidia Sánchez-Puebla
- Instituto de Investigaciones Oftalmológicas Ramón Castroviejo, Grupo UCM 920105, IdISSC, Universidad Complutense de Madrid, 28040 Madrid, Spain
| | - José M. Ramírez
- Instituto de Investigaciones Oftalmológicas Ramón Castroviejo, Grupo UCM 920105, IdISSC, Universidad Complutense de Madrid, 28040 Madrid, Spain
- Facultad de Medicina, Departamento de Inmunología, Oftalmología y ORL, Universidad Complutense de Madrid, 28040 Madrid, Spain
| | - Juan J. Salazar
- Instituto de Investigaciones Oftalmológicas Ramón Castroviejo, Grupo UCM 920105, IdISSC, Universidad Complutense de Madrid, 28040 Madrid, Spain
- Facultad de Óptica y Optometría, Departamento de Inmunología, Oftalmología y ORL, Universidad Complutense de Madrid, 28037 Madrid, Spain
| | - Manuela G. Lopez
- Instituto Teófilo Hernando for Drug Discovery, Department of Pharmacology, School of Medicine, Universidad Autónoma Madrid, 28029 Madrid, Spain
- Instituto de Investigación Sanitario (IIS-IP), Hospital Universitario de La Princesa, 28006 Madrid, Spain
| | - Ana I. Ramírez
- Instituto de Investigaciones Oftalmológicas Ramón Castroviejo, Grupo UCM 920105, IdISSC, Universidad Complutense de Madrid, 28040 Madrid, Spain
- Facultad de Óptica y Optometría, Departamento de Inmunología, Oftalmología y ORL, Universidad Complutense de Madrid, 28037 Madrid, Spain
| |
Collapse
|
21
|
Shi H, Yin Z, Koronyo Y, Fuchs DT, Sheyn J, Davis MR, Wilson JW, Margeta MA, Pitts KM, Herron S, Ikezu S, Ikezu T, Graham SL, Gupta VK, Black KL, Mirzaei M, Butovsky O, Koronyo-Hamaoui M. Regulating microglial miR-155 transcriptional phenotype alleviates Alzheimer's-induced retinal vasculopathy by limiting Clec7a/Galectin-3 + neurodegenerative microglia. Acta Neuropathol Commun 2022; 10:136. [PMID: 36076283 PMCID: PMC9461176 DOI: 10.1186/s40478-022-01439-z] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Accepted: 08/29/2022] [Indexed: 11/16/2022] Open
Abstract
Single cell RNA sequencing studies identified novel neurodegeneration-associated microglial (MGnD/DAM) subtypes activated around cerebral amyloid plaques. Micro-RNA (miR)-155 of the TREM2-APOE pathway was shown to be a key transcriptional regulator of MGnD microglial phenotype. Despite growing interest in studying manifestations of Alzheimer's disease (AD) in the retina, a CNS organ accessible to noninvasive high-resolution imaging, to date MGnD microglia have not been studied in the AD retina. Here, we discovered the presence and increased populations of Clec7a+ and Galectin-3+ MGnD microglia in retinas of transgenic APPSWE/PS1L166P AD-model mice. Conditionally targeting MGnD microglia by miR-155 ablation via the tamoxifen-inducible CreERT2 system in APPSWE/PS1L166P mice diminished retinal Clec7a+ and Galectin-3+ microglial populations while increasing homeostatic P2ry12+ microglia. Retinal MGnD microglia were often adhering to microvessels; their depletion protected the inner blood-retina barrier and reduced vascular amyloidosis. Microglial miR-155 depletion further limits retinal inflammation. Mass spectrometry analysis revealed enhanced retinal PI3K-Akt signaling and predicted IL-8 and Spp1 decreases in mice with microglia-specific miR-155 knockout. Overall, this study identified MGnD microglia in APPSWE/PS1L166P mouse retina. Transcriptional regulation of these dysfunctional microglia mitigated retinal inflammation and vasculopathy. The protective effects of microglial miR-155 ablation should shed light on potential treatments for retinal inflammation and vascular damage during AD and other ocular diseases.
Collapse
Affiliation(s)
- Haoshen Shi
- Department of Neurosurgery, Maxine Dunitz Neurosurgical Research Institute, Cedars-Sinai Medical Center, 127 S. San Vicente Blvd., Los Angeles, CA, A6212, USA
| | - Zhuoran Yin
- Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Yosef Koronyo
- Department of Neurosurgery, Maxine Dunitz Neurosurgical Research Institute, Cedars-Sinai Medical Center, 127 S. San Vicente Blvd., Los Angeles, CA, A6212, USA
| | - Dieu-Trang Fuchs
- Department of Neurosurgery, Maxine Dunitz Neurosurgical Research Institute, Cedars-Sinai Medical Center, 127 S. San Vicente Blvd., Los Angeles, CA, A6212, USA
| | - Julia Sheyn
- Department of Neurosurgery, Maxine Dunitz Neurosurgical Research Institute, Cedars-Sinai Medical Center, 127 S. San Vicente Blvd., Los Angeles, CA, A6212, USA
| | - Miyah R Davis
- Department of Neurosurgery, Maxine Dunitz Neurosurgical Research Institute, Cedars-Sinai Medical Center, 127 S. San Vicente Blvd., Los Angeles, CA, A6212, USA
| | - Jered W Wilson
- Department of Neurosurgery, Maxine Dunitz Neurosurgical Research Institute, Cedars-Sinai Medical Center, 127 S. San Vicente Blvd., Los Angeles, CA, A6212, USA
| | - Milica A Margeta
- Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Department of Ophthalmology, Massachusetts Eye and Ear Infirmary, Harvard Medical School, Boston, MA, USA
| | - Kristen M Pitts
- Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Department of Ophthalmology, Massachusetts Eye and Ear Infirmary, Harvard Medical School, Boston, MA, USA
| | - Shawn Herron
- Department of Pharmacology and Experimental Therapeutics, Boston University School of Medicine, Boston, MA, USA
| | - Seiko Ikezu
- Department of Neuroscience, Mayo Clinic Florida, Jacksonville, FL, USA
| | - Tsuneya Ikezu
- Department of Neuroscience, Mayo Clinic Florida, Jacksonville, FL, USA
| | - Stuart L Graham
- Department of Health and Human Sciences, Macquarie University, Sydney, NSW, Australia
| | - Vivek K Gupta
- Department of Health and Human Sciences, Macquarie University, Sydney, NSW, Australia
| | - Keith L Black
- Department of Neurosurgery, Maxine Dunitz Neurosurgical Research Institute, Cedars-Sinai Medical Center, 127 S. San Vicente Blvd., Los Angeles, CA, A6212, USA
| | - Mehdi Mirzaei
- Department of Molecular Sciences, Macquarie University, Sydney, NSW, Australia
- Department of Clinical Medicine, Department of Molecular Sciences and Australian Proteome Analysis Facility, Macquarie University, Sydney, NSW, Australia
| | - Oleg Butovsky
- Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Evergrande Center for Immunologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Maya Koronyo-Hamaoui
- Department of Neurosurgery, Maxine Dunitz Neurosurgical Research Institute, Cedars-Sinai Medical Center, 127 S. San Vicente Blvd., Los Angeles, CA, A6212, USA.
- Department of Biomedical Sciences, Division of Applied Cell Biology and Physiology, Cedars-Sinai Medical Center, Los Angeles, CA, USA.
| |
Collapse
|
22
|
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
|
23
|
Concomitant Retinal Alterations in Neuronal Activity and TNFα Pathway Are Detectable during the Pre-Symptomatic Stage in a Mouse Model of Alzheimer's Disease. Cells 2022; 11:cells11101650. [PMID: 35626688 PMCID: PMC9140134 DOI: 10.3390/cells11101650] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Revised: 04/21/2022] [Accepted: 05/13/2022] [Indexed: 12/10/2022] Open
Abstract
The pre-symptomatic stage of Alzheimer’s disease (AD) is associated with increased amyloid-β (Aβ) precursor protein (APP) processing and Aβ accumulation in the retina and hippocampus. Because neuronal dysfunctions are among the earliest AD-related alterations, we asked whether they are already detectable in the retina during the pre-symptomatic stage in a APPswePS1dE9 (APP/PS1) mouse model. The age chosen for the study (3–4 months) corresponds to the pre-symptomatic stage because no retinal Aβ was detected, in spite of the presence of βCTF (the first cleavage product of APP). We observed an increase in ERG amplitudes in APP/PS1 mice in comparison to the controls, which indicated an increased retinal neuron activity. These functional changes coincided with an increased expression of retinal TNFα and its receptors type-1 (TNFR1). Consistently, the IkB expression increased in APP/PS1 mice with a greater proportion of the phosphorylated protein (P-IkB) over total IkB, pointing to the putative involvement of the NFkB pathway. Because TNFα plays a crucial role in the control of neuronal excitability, it is likely that, as in the hippocampus, TNFα signaling via the TNFR1/NFkB pathway may be also involved in early, AD-associated, retinal neuron hyperexcitability. These results further demonstrate the interest of the retina for early disease detection with a potential to assess future therapeutic strategies.
Collapse
|
24
|
Zhang J, Shi L, Shen Y. The retina: A window in which to view the pathogenesis of Alzheimer's disease. Ageing Res Rev 2022; 77:101590. [PMID: 35192959 DOI: 10.1016/j.arr.2022.101590] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Revised: 01/14/2022] [Accepted: 02/12/2022] [Indexed: 01/01/2023]
Abstract
Alzheimer's disease (AD) is the most familiar type of dementia affecting elderly populations worldwide. Studies of AD patients and AD transgenic mice have revealed alterations in the retina similar to alterations which occur in the AD brain. Moreover, AD retinal pathology occurs even earlier than AD brain pathology. Importantly, non-invasive imaging techniques can be utilized for retinal observation due to the unique optical transparency of the eye, which acts as a convenient window in which preclinical pathology in the AD brain can be monitored. In this review, we overview the existing literature covering different forms of AD retinal pathology and propose a basis for the clinical application of using the retina as a window to view AD during preclinical and clinical stages.
Collapse
Affiliation(s)
- Jie Zhang
- Institute on Aging and Brain Disorders, The First Affiliated Hospital of USTC, Neurodegenerative Disorder Research Center, School of Life Science, Division of Life Science and Medicine, University of Science and Technology of China, Hefei 230026, China
| | - Lei Shi
- Institute on Aging and Brain Disorders, The First Affiliated Hospital of USTC, Neurodegenerative Disorder Research Center, School of Life Science, Division of Life Science and Medicine, University of Science and Technology of China, Hefei 230026, China
| | - Yong Shen
- Institute on Aging and Brain Disorders, The First Affiliated Hospital of USTC, Neurodegenerative Disorder Research Center, School of Life Science, Division of Life Science and Medicine, University of Science and Technology of China, Hefei 230026, China; Center for Excellence in Brain Sciences and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China.
| |
Collapse
|
25
|
Yeh TC, Kuo CT, Chou YB. Retinal Microvascular Changes in Mild Cognitive Impairment and Alzheimer's Disease: A Systematic Review, Meta-Analysis, and Meta-Regression. Front Aging Neurosci 2022; 14:860759. [PMID: 35572135 PMCID: PMC9096234 DOI: 10.3389/fnagi.2022.860759] [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: 01/23/2022] [Accepted: 03/14/2022] [Indexed: 01/03/2023] Open
Abstract
Background The remarkable increase in prevalence and significant morbidity of neurodegenerative diseases pose a tremendous burden for the health care system. Changes in retinal microvasculature metrics associated with Alzheimer's disease (AD) and mild cognitive impairment (MCI) may provide opportunities for early diagnosis and intervention. However, the role of retinal vascular biomarkers remains controversial. We aim to perform a systematic review, meta-analysis and meta-regression to evaluate the comprehensive retinal microvasculature changes in patients with AD and MCI. Methods We conducted a literature search on PubMed, MEDLINE, and EMBASE to identify studies published before May 2021 which assessed the measurements of optical coherence tomography angiography (OCTA) between AD, MCI with healthy control eyes, including foveal avascular zone (FAZ), vessel density (VD) of peripapillary, superficial and deep capillary plexus, and choroidal thickness using a random-effect model. We also performed meta-regression and subgroup analysis and assessed heterogeneity and publication bias to evaluate potential sources of bias. Results Compared with control eyes, VD of superficial capillary plexus was significantly lower in AD [standardized mean difference (SMD): −0.48; 95% CI (−0.70 to −0.27); p = 0.04] and MCI eyes [SMD: −0.42; 95% CI (−0.81 to −0.03); p = 0.03], as well as reduced VD of deep capillary plexus [SMD: −1.19; 95% CI (−2.00 to −0.38]; p < 0.001], [SMD: −0.53; 95% CI (−0.85 to −0.22); p < 0.001]. FAZ was significantly enlarged in AD eyes [SMD: 0.54; 95% CI (0.09 to 0.99); p = 0.02]. The meta-regression analysis showed that the OCTA machine type and macular scan size significantly influenced the variation of VD and FAZ between AD and control eyes (p < 0.05). Conclusion Our results highlight the potential of OCTA as a biomarker to detect early microvasculature deficits in AD and MCI. Notably, the macular scan size and different OCTA machine type could explain the heterogeneity observed in literatures. This information might be useful for future longitudinal study design to evaluate the role of OCTA in monitoring disease progression and treatment efficacy.
Collapse
Affiliation(s)
- Tsai-Chu Yeh
- Department of Ophthalmology, Taipei Veterans General Hospital, Taipei, Taiwan
- School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Chun-Tung Kuo
- Institute of Health Behaviors and Community Sciences, College of Public Health, National Taiwan University, Taipei, Taiwan
- School of Public Health, National Defense Medical Center, Taipei, Taiwan
| | - Yu-Bai Chou
- Department of Ophthalmology, Taipei Veterans General Hospital, Taipei, Taiwan
- School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
- *Correspondence: Yu-Bai Chou
| |
Collapse
|
26
|
Wu H, Lei Z, Ou Y, Shi X, Xu Q, Shi K, Ding J, Zhao Q, Wang X, Cai X, Liu X, Lou J, Liu X. Computed Tomography Density and β-Amyloid Deposition of Intraorbital Optic Nerve May Assist in Diagnosing Mild Cognitive Impairment and Alzheimer's Disease: A 18F-Flutemetamol Positron Emission Tomography/Computed Tomography Study. Front Aging Neurosci 2022; 14:836568. [PMID: 35370601 PMCID: PMC8970307 DOI: 10.3389/fnagi.2022.836568] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Accepted: 01/26/2022] [Indexed: 11/24/2022] Open
Abstract
Objective The aim was to study whether the computed tomography (CT) density and β-amyloid (Aβ) level of intraorbital optic nerve could assist in diagnosing mild cognitive impairment (MCI) and Alzheimer's disease (AD). Methods A total of sixty subjects were recruited in our study, including nine normal control (NC) subjects (i.e., 4 men and 5 women), twenty four MCI subjects (i.e., 11 men and 13 women), and twenty seven AD subjects (i.e., 14 men and 13 women). All subjects conducted 18F-flutemetamol amyloid positron emission tomography (PET)/CT imaging. Blinded to the clinical information of the subjects, two physicians independently measured and calculated the standardized uptake value ratio (SUVR) of the bilateral occipital cortex, SUVR of the bilateral intraorbital optic nerve, and CT density of the bilateral intraorbital optic nerve by using GE AW 4.5 Workstation. Results Between AD and NC groups, the differences of the bilateral intraorbital optic nerve SUVR were statistically significant; between AD and MCI groups, the differences of the left intraorbital optic nerve SUVR were statistically significant. Between any two of the three groups, the differences in the bilateral intraorbital optic nerve density were statistically significant. The bilateral occipital SUVR was positively correlated with the bilateral intraorbital optic nerve SUVR and negatively correlated with the bilateral intraorbital optic nerve density. Bilateral intraorbital optic nerve SUVR was negatively correlated with the bilateral intraorbital optic nerve density. The area under the receiver operating characteristic (ROC) curve of multiple logistic regression was 0.9167 (for MCI vs. NC) and 0.8951 (for AD vs. MCI). The Montreal Cognitive Assessment (MoCA) and Mini-Mental State Examination (MMSE) scores were positively associated with the intraorbital optic nerve density and were negatively associated with the intraorbital optic nerve SUVR. The regression equation of MoCA was y = 16.37-0.9734 × x 1 + 0.5642 × x 2-3.127 × x 3 + 0.0275 × x 4; the R 2 was 0.848. The regression equation of MMSE was y = 19.57-1.633 × x 1 + 0.4397 × x 2-1.713 × x 3 + 0.0424 × x 4; the R 2 was 0.827. Conclusion The CT density and Aβ deposition of the intraorbital optic nerve were associated with Aβ deposition of the occipital cortex and the severity of cognitive impairment. The intraorbital optic nerve CT density and intraorbital optic nerve Aβ deposition could assist in diagnosing MCI and AD.
Collapse
Affiliation(s)
- Han Wu
- Department of Nuclear Medicine, Huashan Hospital, Fudan University, Shanghai, China
- Department of Nuclear Medicine, Pudong Hospital, Fudan University, Shanghai, China
| | - Zhe Lei
- Department of Nuclear Medicine, Huashan Hospital, Fudan University, Shanghai, China
- Department of Nuclear Medicine, Pudong Hospital, Fudan University, Shanghai, China
| | - Yinghui Ou
- Department of Nuclear Medicine, Huashan Hospital, Fudan University, Shanghai, China
- Department of Nuclear Medicine, Pudong Hospital, Fudan University, Shanghai, China
| | - Xin Shi
- Department of Nuclear Medicine, Huashan Hospital, Fudan University, Shanghai, China
| | - Qian Xu
- Department of Nuclear Medicine, Huashan Hospital, Fudan University, Shanghai, China
| | - Keqing Shi
- Department of Nuclear Medicine, Huashan Hospital, Fudan University, Shanghai, China
| | - Jing Ding
- Department of Neurology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Qianhua Zhao
- Department of Neurology, Huashan Hospital, Fudan University, Shanghai, China
| | - Xiuzhe Wang
- Department of Neurology, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University, Shanghai, China
| | - Xiaolong Cai
- Department of Neurology, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University, Shanghai, China
| | - Xueyuan Liu
- Department of Neurology, Tenth People’s Hospital affiliated to Tongji University, Shanghai, China
| | - Jingjing Lou
- Department of Nuclear Medicine, Pudong Hospital, Fudan University, Shanghai, China
| | - Xingdang Liu
- Department of Nuclear Medicine, Huashan Hospital, Fudan University, Shanghai, China
- Department of Nuclear Medicine, Pudong Hospital, Fudan University, Shanghai, China
| |
Collapse
|
27
|
Bogolepova A, Makhnovich E, Kovalenko E, Osinovskaya N. Potential biomarkers of early diagnosis of Alzheimer’s disease. Zh Nevrol Psikhiatr Im S S Korsakova 2022; 122:7-14. [DOI: 10.17116/jnevro20221220917] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
|
28
|
Little K, Llorián-Salvador M, Scullion S, Hernández C, Simó-Servat O, Del Marco A, Bosma E, Vargas-Soria M, Carranza-Naval MJ, Van Bergen T, Galbiati S, Viganò I, Musi CA, Schlingemann R, Feyen J, Borsello T, Zerbini G, Klaassen I, Garcia-Alloza M, Simó R, Stitt AW. Common pathways in dementia and diabetic retinopathy: understanding the mechanisms of diabetes-related cognitive decline. Trends Endocrinol Metab 2022; 33:50-71. [PMID: 34794851 DOI: 10.1016/j.tem.2021.10.008] [Citation(s) in RCA: 55] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Revised: 09/06/2021] [Accepted: 10/29/2021] [Indexed: 12/14/2022]
Abstract
Type 2 diabetes (T2D) is associated with multiple comorbidities, including diabetic retinopathy (DR) and cognitive decline, and T2D patients have a significantly higher risk of developing Alzheimer's disease (AD). Both DR and AD are characterized by a number of pathological mechanisms that coalesce around the neurovascular unit, including neuroinflammation and degeneration, vascular degeneration, and glial activation. Chronic hyperglycemia and insulin resistance also play a significant role, leading to activation of pathological mechanisms such as increased oxidative stress and the accumulation of advanced glycation end-products (AGEs). Understanding these common pathways and the degree to which they occur simultaneously in the brain and retina during diabetes will provide avenues to identify T2D patients at risk of cognitive decline.
Collapse
Affiliation(s)
- Karis Little
- Wellcome-Wolfson Institute for Experimental Medicine, Queen's University Belfast, Belfast, UK
| | - María Llorián-Salvador
- Wellcome-Wolfson Institute for Experimental Medicine, Queen's University Belfast, Belfast, UK
| | - Sarah Scullion
- Wellcome-Wolfson Institute for Experimental Medicine, Queen's University Belfast, Belfast, UK
| | - Cristina Hernández
- Vall d'Hebron Research Institute and CIBERDEM (ISCIII), Barcelona, Spain
| | - Olga Simó-Servat
- Vall d'Hebron Research Institute and CIBERDEM (ISCIII), Barcelona, Spain
| | - Angel Del Marco
- Division of Physiology, School of Medicine, Instituto de Investigacion Biomedica de Cadiz (INIBICA), Universidad de Cadiz, Cadiz, Spain
| | - Esmeralda Bosma
- Ocular Angiogenesis Group, University of Amsterdam, Amsterdam, The Netherlands
| | - Maria Vargas-Soria
- Division of Physiology, School of Medicine, Instituto de Investigacion Biomedica de Cadiz (INIBICA), Universidad de Cadiz, Cadiz, Spain
| | - Maria Jose Carranza-Naval
- Division of Physiology, School of Medicine, Instituto de Investigacion Biomedica de Cadiz (INIBICA), Universidad de Cadiz, Cadiz, Spain
| | | | - Silvia Galbiati
- Complications of Diabetes Unit, Diabetes Research Institute, IRCCS Ospedale San Raffaele, Milano, Italy
| | - Ilaria Viganò
- Complications of Diabetes Unit, Diabetes Research Institute, IRCCS Ospedale San Raffaele, Milano, Italy
| | - Clara Alice Musi
- Università Degli Studi di Milano and Istituto di Ricerche Farmacologiche Mario Negri- IRCCS, Milano, Italy
| | - Reiner Schlingemann
- Ocular Angiogenesis Group, University of Amsterdam, Amsterdam, The Netherlands; Department of Ophthalmology, University of Lausanne, Jules Gonin Eye Hospital, Lausanne, Switzerland
| | | | - Tiziana Borsello
- Università Degli Studi di Milano and Istituto di Ricerche Farmacologiche Mario Negri- IRCCS, Milano, Italy
| | - Gianpaolo Zerbini
- Complications of Diabetes Unit, Diabetes Research Institute, IRCCS Ospedale San Raffaele, Milano, Italy
| | - Ingeborg Klaassen
- Ocular Angiogenesis Group, University of Amsterdam, Amsterdam, The Netherlands
| | - Monica Garcia-Alloza
- Division of Physiology, School of Medicine, Instituto de Investigacion Biomedica de Cadiz (INIBICA), Universidad de Cadiz, Cadiz, Spain
| | - Rafael Simó
- Vall d'Hebron Research Institute and CIBERDEM (ISCIII), Barcelona, Spain.
| | - Alan W Stitt
- Wellcome-Wolfson Institute for Experimental Medicine, Queen's University Belfast, Belfast, UK.
| | | |
Collapse
|
29
|
Progressive impairments in executive function in the APP/PS1 model of Alzheimer's disease as measured by translatable touchscreen testing. Neurobiol Aging 2021; 108:58-71. [PMID: 34509856 DOI: 10.1016/j.neurobiolaging.2021.08.004] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2019] [Revised: 07/19/2021] [Accepted: 08/04/2021] [Indexed: 02/08/2023]
Abstract
Executive function deficits in Alzheimer's disease (AD) occur early in disease progression and may be predictive of cognitive decline. However, no preclinical studies have identified deficits in rewarded executive function in the commonly used APPSwe/PS1∆E9 (APP/PS1) mouse model. To address this, we assessed 12-26 month old APP/PS1 mice on rewarded reversal and/or extinction tasks. 16-month-old, but not 13- or 26-month-old, APP/PS1 mice showed an attenuated rate of extinction. Reversal deficits were seen in 22-month-old, but not 13-month-old APP/PS1 animals. We then confirmed that impairments in reversal were unrelated to previously reported visual impairments in both AD mouse models and humans. Age, but not genotype, had a significant effect on markers of retinal health, indicating the deficits seen in APP/PS1 mice were directly related to cognition. This is the first characterisation of rewarded executive function in APP/PS1 mice, and has great potential to facilitate translation from preclinical models to the clinic.
Collapse
|
30
|
Zhao W, Lv X, Wu G, Zhou X, Tian H, Qu X, Sun H, He Y, Zhang Y, Wang C, Tian J. Glaucoma Is Not Associated With Alzheimer's Disease or Dementia: A Meta-Analysis of Cohort Studies. Front Med (Lausanne) 2021; 8:688551. [PMID: 34504851 PMCID: PMC8423132 DOI: 10.3389/fmed.2021.688551] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Accepted: 06/28/2021] [Indexed: 11/13/2022] Open
Abstract
Background: Previous studies evaluating the relationships of glaucoma with Alzheimer's disease (AD) and dementia showed inconsistent results. We performed a meta-analysis of cohort studies to evaluate the association between glaucoma with incidence of AD, all-cause dementia, and non-AD dementia. Methods: Cohort studies which evaluated the association between glaucoma with incidence of AD, all-cause dementia, and non-AD dementia in adult population with multivariate analyses were identified by systematic search of PubMed, Embase, and Cochrane's Library databases. A random-effects model incorporating the potential intra-study heterogeneity was used for the meta-analysis. Results: Eleven cohort studies including 4,645,925 participants were included. Results showed that compared to those without glaucoma at baseline, adult patients with glaucoma was not independently associated with increased incidence of AD [adjusted risk ratio (RR): 1.03, 95% confidence interval (CI): 0.93-1.05, P = 0.55; I 2 = 83%], all-cause dementia (adjusted RR: 1.08, 95% CI: 0.97-1.19, P = 0.15; I 2 = 79%), or non-AD dementia (adjusted RR: 1.05 95% CI: 0.91-1.21, P = 0.49; I 2 = 82%). Sensitivity analyses by excluding one study at a time did not significantly affect the results of the meta-analyses. Moreover, subgroup analyses showed consistent results in meta-analysis of prospective or retrospective cohort studies, and in meta-analysis of patients with primary open-angle glaucoma or primary angle-closure glaucoma (P-values for subgroup difference all > 0.05). Conclusions: Current evidence from cohort studies did not support that glaucoma is an independent risk factor of AD, all-cause dementia, or non-AD dementia in adult population.
Collapse
Affiliation(s)
- Wenmei Zhao
- Zunyi Medical University, Zunyi, China.,Department of Emergency, Guizhou Provincial People's Hospital, Guiyang, China
| | - Xia Lv
- Department of Emergency, Guizhou Provincial People's Hospital, Guiyang, China
| | - Guangjie Wu
- Department of Emergency, Guizhou Provincial People's Hospital, Guiyang, China
| | - Xia Zhou
- Department of Emergency, Guizhou Provincial People's Hospital, Guiyang, China
| | - Helan Tian
- Department of Emergency, Guizhou Provincial People's Hospital, Guiyang, China
| | - Xiang Qu
- Department of Emergency, Guizhou Provincial People's Hospital, Guiyang, China
| | - Hongpeng Sun
- Department of Emergency, Guizhou Provincial People's Hospital, Guiyang, China
| | - Yingying He
- Department of Emergency, Guizhou Provincial People's Hospital, Guiyang, China
| | - Yingyue Zhang
- Department of Emergency, Guizhou Provincial People's Hospital, Guiyang, China
| | - Chuan Wang
- Department of Emergency, Guizhou Provincial People's Hospital, Guiyang, China
| | - Jinyong Tian
- Department of Emergency, Guizhou Provincial People's Hospital, Guiyang, China
| |
Collapse
|
31
|
Tadokoro K, Yamashita T, Kimura S, Nomura E, Ohta Y, Omote Y, Takemoto M, Hishikawa N, Morihara R, Morizane Y, Abe K. Retinal Amyloid Imaging for Screening Alzheimer's Disease. J Alzheimers Dis 2021; 83:927-934. [PMID: 34366344 DOI: 10.3233/jad-210327] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
BACKGROUND Cost-effective and noninvasive methods for in vivo imaging of amyloid deposition are needed to screen Alzheimer's disease (AD). Although retinal amyloid is a possible diagnostic marker of AD, there are very few studies on in vivo retinal amyloid imaging. OBJECTIVE To examine the usefulness of in vivo imaging of retinal amyloid in AD patients. METHODS To examine amyloid deposition, 30 Japanese subjects (10 normal control (NC), 7 with mild cognitive impairment (MCI), and 13 with AD) underwent a complete ophthalmic examination, including fundus imaging by scanning laser ophthalmoscopy before and after oral curcumin intake. RESULTS Retinal amyloid deposition was greater in AD than in NC subjects (*p < 0.05) while MCI showed a slight but insignificant increase of retinal amyloid deposition relative to NC subjects. Retinal amyloid deposition was correlated with whole gray matter atrophy (r = 0.51, *p < 0.05) but not with the cognitive score of the Mini-Mental State Examination, nor with medial temporal lobe atrophy. CONCLUSION The present noninvasive in vivo detection of retinal amyloid deposition is useful for screening AD patients.
Collapse
Affiliation(s)
- Koh Tadokoro
- Department of Neurology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Kita-ku, Okayama, Japan
| | - Toru Yamashita
- Department of Neurology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Kita-ku, Okayama, Japan
| | - Shuhei Kimura
- Department of Ophthalmology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Kita-ku, Okayama, Japan
| | - Emi Nomura
- Department of Neurology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Kita-ku, Okayama, Japan
| | - Yasuyuki Ohta
- Department of Neurology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Kita-ku, Okayama, Japan
| | - Yoshio Omote
- Department of Neurology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Kita-ku, Okayama, Japan
| | - Mami Takemoto
- Department of Neurology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Kita-ku, Okayama, Japan
| | - Nozomi Hishikawa
- Department of Neurology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Kita-ku, Okayama, Japan
| | - Ryuta Morihara
- Department of Neurology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Kita-ku, Okayama, Japan
| | - Yuki Morizane
- Department of Ophthalmology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Kita-ku, Okayama, Japan
| | - Koji Abe
- Department of Neurology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Kita-ku, Okayama, Japan
| |
Collapse
|
32
|
Chitranshi N, Kumar A, Sheriff S, Gupta V, Godinez A, Saks D, Sarkar S, Shen T, Mirzaei M, Basavarajappa D, Abyadeh M, Singh SK, Dua K, Zhang KYJ, Graham SL, Gupta V. Identification of Novel Cathepsin B Inhibitors with Implications in Alzheimer's Disease: Computational Refining and Biochemical Evaluation. Cells 2021; 10:cells10081946. [PMID: 34440715 PMCID: PMC8391575 DOI: 10.3390/cells10081946] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2021] [Revised: 07/23/2021] [Accepted: 07/27/2021] [Indexed: 02/07/2023] Open
Abstract
Amyloid precursor protein (APP), upon proteolytic degradation, forms aggregates of amyloid β (Aβ) and plaques in the brain, which are pathological hallmarks of Alzheimer’s disease (AD). Cathepsin B is a cysteine protease enzyme that catalyzes the proteolytic degradation of APP in the brain. Thus, cathepsin B inhibition is a crucial therapeutic aspect for the discovery of new anti-Alzheimer’s drugs. In this study, we have employed mixed-feature ligand-based virtual screening (LBVS) by integrating pharmacophore mapping, docking, and molecular dynamics to detect small, potent molecules that act as cathepsin B inhibitors. The LBVS model was generated by using hydrophobic (HY), hydrogen bond acceptor (HBA), and hydrogen bond donor (HBD) features, using a dataset of 24 known cathepsin B inhibitors of both natural and synthetic origins. A validated eight-feature pharmacophore hypothesis (Hypo III) was utilized to screen the Maybridge chemical database. The docking score, MM-PBSA, and MM-GBSA methodology was applied to prioritize the lead compounds as virtual screening hits. These compounds share a common amide scaffold, and showed important interactions with Gln23, Cys29, His110, His111, Glu122, His199, and Trp221. The identified inhibitors were further evaluated for cathepsin-B-inhibitory activity. Our study suggests that pyridine, acetamide, and benzohydrazide compounds could be used as a starting point for the development of novel therapeutics.
Collapse
Affiliation(s)
- Nitin Chitranshi
- Faculty of Medicine, Health and Human Sciences, Macquarie University, F10A, 2 Technology Place, North Ryde, NSW 2109, Australia; (S.S.); (A.G.); (D.S.); (S.S.); (T.S.); (M.M.); (D.B.); (S.L.G.)
- Correspondence: (N.C.); (V.G.); Tel.: +61-(02)-9850-2804 (N.C.)
| | - Ashutosh Kumar
- Center for Biosystems Dynamics Research, Laboratory for Structural Bioinformatics, RIKEN, 1-7-22 Suehiro, Tsurumi, Yokohama 230-0045, Kanagawa, Japan; (A.K.); (K.Y.J.Z.)
| | - Samran Sheriff
- Faculty of Medicine, Health and Human Sciences, Macquarie University, F10A, 2 Technology Place, North Ryde, NSW 2109, Australia; (S.S.); (A.G.); (D.S.); (S.S.); (T.S.); (M.M.); (D.B.); (S.L.G.)
| | - Veer Gupta
- School of Medicine, Faculty of Health, Deakin University, Geelong, VIC 3220, Australia;
| | - Angela Godinez
- Faculty of Medicine, Health and Human Sciences, Macquarie University, F10A, 2 Technology Place, North Ryde, NSW 2109, Australia; (S.S.); (A.G.); (D.S.); (S.S.); (T.S.); (M.M.); (D.B.); (S.L.G.)
| | - Danit Saks
- Faculty of Medicine, Health and Human Sciences, Macquarie University, F10A, 2 Technology Place, North Ryde, NSW 2109, Australia; (S.S.); (A.G.); (D.S.); (S.S.); (T.S.); (M.M.); (D.B.); (S.L.G.)
| | - Soumalya Sarkar
- Faculty of Medicine, Health and Human Sciences, Macquarie University, F10A, 2 Technology Place, North Ryde, NSW 2109, Australia; (S.S.); (A.G.); (D.S.); (S.S.); (T.S.); (M.M.); (D.B.); (S.L.G.)
| | - Ting Shen
- Faculty of Medicine, Health and Human Sciences, Macquarie University, F10A, 2 Technology Place, North Ryde, NSW 2109, Australia; (S.S.); (A.G.); (D.S.); (S.S.); (T.S.); (M.M.); (D.B.); (S.L.G.)
| | - Mehdi Mirzaei
- Faculty of Medicine, Health and Human Sciences, Macquarie University, F10A, 2 Technology Place, North Ryde, NSW 2109, Australia; (S.S.); (A.G.); (D.S.); (S.S.); (T.S.); (M.M.); (D.B.); (S.L.G.)
| | - Devaraj Basavarajappa
- Faculty of Medicine, Health and Human Sciences, Macquarie University, F10A, 2 Technology Place, North Ryde, NSW 2109, Australia; (S.S.); (A.G.); (D.S.); (S.S.); (T.S.); (M.M.); (D.B.); (S.L.G.)
| | - Morteza Abyadeh
- Cell Science Research Center, Department of Molecular Systems Biology, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran 1665659911, Iran;
| | - Sachin K. Singh
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara 144411, Punjab, India;
| | - Kamal Dua
- Discipline of Pharmacy, Graduate School of Health, University of Technology Sydney, Ultimo, NSW 2007, Australia;
- Australian Research Centre in Complementary and Integrative Medicine, Faculty of Health, University of Technology Sydney, Ultimo, NSW 2007, Australia
| | - Kam Y. J. Zhang
- Center for Biosystems Dynamics Research, Laboratory for Structural Bioinformatics, RIKEN, 1-7-22 Suehiro, Tsurumi, Yokohama 230-0045, Kanagawa, Japan; (A.K.); (K.Y.J.Z.)
| | - Stuart L. Graham
- Faculty of Medicine, Health and Human Sciences, Macquarie University, F10A, 2 Technology Place, North Ryde, NSW 2109, Australia; (S.S.); (A.G.); (D.S.); (S.S.); (T.S.); (M.M.); (D.B.); (S.L.G.)
| | - Vivek Gupta
- Faculty of Medicine, Health and Human Sciences, Macquarie University, F10A, 2 Technology Place, North Ryde, NSW 2109, Australia; (S.S.); (A.G.); (D.S.); (S.S.); (T.S.); (M.M.); (D.B.); (S.L.G.)
- Correspondence: (N.C.); (V.G.); Tel.: +61-(02)-9850-2804 (N.C.)
| |
Collapse
|
33
|
Guo L, Ravindran N, Shamsher E, Hill D, Cordeiro MF. Retinal Changes in Transgenic Mouse Models of Alzheimer's Disease. Curr Alzheimer Res 2021; 18:89-102. [PMID: 33855942 DOI: 10.2174/1567205018666210414113634] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2020] [Revised: 02/09/2021] [Accepted: 04/05/2021] [Indexed: 11/22/2022]
Abstract
Alzheimer's disease (AD) is a neurodegenerative disorder, the most common form of dementia. AD is characterised by amyloid-β (Aβ) plaques and neurofibrillary tangles (NFT) in the brain, in association with neuronal loss and synaptic failure, causing cognitive deficits. Accurate and early diagnosis is currently unavailable in lifespan, hampering early intervention of potential new treatments. Visual deficits have been well documented in AD patients, and the pathological changes identified in the brain are also believed to be found in the retina, an integral part of the central nervous system. Retinal changes can be detected by real-time non-invasive imaging, due to the transparent nature of the ocular media, potentially allowing an earlier diagnosis as well as monitoring disease progression and treatment outcome. Animal models are essential for AD research, and this review has a focus on retinal changes in various transgenic AD mouse models with retinal imaging and immunohistochemical analysis as well as therapeutic effects in those models. We also discuss the limitations of transgenic AD models in clinical translations.
Collapse
Affiliation(s)
- Li Guo
- Glaucoma & Retinal Neurodegeneration Research Group, Institute of Ophthalmology, University College London, London, United Kingdom
| | - Nivedita Ravindran
- Glaucoma & Retinal Neurodegeneration Research Group, Institute of Ophthalmology, University College London, London, United Kingdom
| | - Ehtesham Shamsher
- Glaucoma & Retinal Neurodegeneration Research Group, Institute of Ophthalmology, University College London, London, United Kingdom
| | - Daniel Hill
- Glaucoma & Retinal Neurodegeneration Research Group, Institute of Ophthalmology, University College London, London, United Kingdom
| | - M Francesca Cordeiro
- Glaucoma & Retinal Neurodegeneration Research Group, Institute of Ophthalmology, University College London, London, United Kingdom
| |
Collapse
|
34
|
Araya-Arriagada J, Bello F, Shivashankar G, Neira D, Durán-Aniotz C, Acosta ML, Escobar MJ, Hetz C, Chacón M, Palacios AG. Retinal Ganglion Cells Functional Changes in a Mouse Model of Alzheimer's Disease Are Linked with Neurotransmitter Alterations. J Alzheimers Dis 2021; 82:S5-S18. [PMID: 33749647 DOI: 10.3233/jad-201195] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
BACKGROUND Alzheimer's disease (AD) is the most prevalent form of dementia worldwide. This neurodegenerative syndrome affects cognition, memory, behavior, and the visual system, particularly the retina. OBJECTIVE This work aims to determine whether the 5xFAD mouse, a transgenic model of AD, displays changes in the function of retinal ganglion cells (RGCs) and if those alterations are correlated with changes in the expression of glutamate and gamma-aminobutyric acid (GABA) neurotransmitters. METHODS In young (2-3-month-old) and adult (6-7-month-old) 5xFAD and WT mice, we have studied the physiological response, firing rate, and burst of RGCs to various types of visual stimuli using a multielectrode array system. RESULTS The firing rate and burst response in 5xFAD RGCs showed hyperactivity at the early stage of AD in young mice, whereas hypoactivity was seen at the later stage of AD in adults. The physiological alterations observed in 5xFAD correlate well with an increase in the expression of glutamate in the ganglion cell layer in young and adults. GABA staining increased in the inner nuclear and plexiform layer, which was more pronounced in the adult than the young 5xFAD retina, altering the excitation/inhibition balance, which could explain the observed early hyperactivity and later hypoactivity in RGC physiology. CONCLUSION These findings indicate functional changes may be caused by neurochemical alterations of the retina starting at an early stage of the AD disease.
Collapse
Affiliation(s)
- Joaquín Araya-Arriagada
- Centro Interdisciplinario de Neurociencia de Valparaíso, Universidad de Valparaíso, Valparaíso, Chile.,Escuela de Tecnología Médica, Facultad de Salud, Universidad Santo Tomás, Chile
| | - Felipe Bello
- Department of Engineering Informatics, Universidad de Santiago, Santiago, Chile
| | - Gaganashree Shivashankar
- School of Optometry and Vision Science; Centre for Brain Research; Brain Research New Zealand; The University of Auckland, Auckland, New Zealand
| | - David Neira
- Centro Interdisciplinario de Neurociencia de Valparaíso, Universidad de Valparaíso, Valparaíso, Chile
| | - Claudia Durán-Aniotz
- Center for Social and Cognitive Neuroscience, School of Psychology, Universidad Adolfo Ibáñez, Santiago de Chile, Chile
| | - Mónica L Acosta
- School of Optometry and Vision Science; Centre for Brain Research; Brain Research New Zealand; The University of Auckland, Auckland, New Zealand
| | - María José Escobar
- Departamento de Electrónica, Universidad Técnica Federico Santa María, Valparaíso, Chile
| | - Claudio Hetz
- Biomedical Neuroscience Institute, Universidad de Chile, Santiago, Chile
| | - Max Chacón
- Department of Engineering Informatics, Universidad de Santiago, Santiago, Chile
| | - Adrián G Palacios
- Centro Interdisciplinario de Neurociencia de Valparaíso, Universidad de Valparaíso, Valparaíso, Chile
| |
Collapse
|
35
|
Retinal hyperspectral imaging in the 5xFAD mouse model of Alzheimer's disease. Sci Rep 2021; 11:6387. [PMID: 33737550 PMCID: PMC7973540 DOI: 10.1038/s41598-021-85554-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Accepted: 02/20/2021] [Indexed: 11/08/2022] Open
Abstract
Hyperspectral imaging of the retina has recently been posited as a potentially useful form of spectroscopy of amyloid-beta (Aβ) protein in the eyes of those with Alzheimer's disease (AD). The concept of using the retina as a biomarker for AD is an attractive one, as current screening tools for AD are either expensive or inaccessible. Recent studies have investigated hyperspectral imaging in Aβ models however these studies have been in younger mice. Here we characterised hyperspectral reflectance profile in 6 to 17 months old 5xFAD mice and compare this to Aβ in isolated preparations. Hyperspectral imaging was conducted across two preparations of Aβ using a custom built bench ophthalmoscope. In the in vitro condition, 1 mg of purified human Aβ42 was solubilised and left to aggregate for 72 h. This soluble/insoluble Aβ mixture was then imaged by suspending the solution at a pipette tip and compared against phosphate buffered saline (PBS) control (n = 10 ROIs / group). In the in vivo condition, a 5xFAD transgenic mouse model was used and retinae were imaged at the age of 6 (n = 9), 12 (n = 9) and 17 months (n = 8) with age matched wildtype littermates as control (n = 12, n = 13, n = 15 respectively). In the vitro condition, hyperspectral imaging of the solution showed greater reflectance compared with vehicle (p < 0.01), with the greatest differences occurring in the short visible spectrum (< 500 nm). In the in vivo preparation, 5xFAD showed greater hyperspectral reflectance at all ages (6, 12, 17 months, p < 0.01). These differences were noted most in the short wavelengths at younger ages, with an additional peak appearing at longer wavelengths (~ 550 nm) with advancing age. This study shows that the presence of Aβ (soluble/insoluble mixture) can increase the hyperspectral reflectance profile in vitro as well as in vivo. Differences were evident in the short wavelength spectrum (< 500 nm) in vitro and were preserved when imaged through the ocular media in the in vivo conditions. With advancing age a second hump around ~ 550 nm became more apparent. Hyperspectral imaging of the retina does not require the use of contrast agents and is a potentially useful and non-invasive biomarker for AD.
Collapse
|
36
|
Deng L, Gupta VK, Wu Y, Pushpitha K, Chitranshi N, Gupta VB, Fitzhenry MJ, Moghaddam MZ, Karl T, Salekdeh GH, Graham SL, Haynes PA, Mirzaei M. Mouse model of Alzheimer's disease demonstrates differential effects of early disease pathology on various brain regions. Proteomics 2021; 21:e2000213. [PMID: 33559908 DOI: 10.1002/pmic.202000213] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2020] [Revised: 01/31/2021] [Accepted: 02/01/2021] [Indexed: 12/17/2022]
Abstract
Different parts of the brain are affected distinctively in various stages of the Alzheimer's disease (AD) pathogenesis. Identifying the biochemical changes in specific brain regions is key to comprehend the neuropathological mechanisms in early pre-symptomatic phases of AD. Quantitative proteomics profiling of four distinct areas of the brain of young APP/PS1 mouse model of AD was performed followed by biochemical pathway enrichment analysis. Findings revealed fundamental compositional and functional shifts even in the early stages of the disease. This novel study highlights unique proteome and biochemical pathway alterations in specific regions of the brain that underlie the early stages of AD pathology and will provide a framework for future longitudinal studies. The proteomics data were deposited into the ProteomeXchange Consortium via PRIDE with the identifier PXD019192.
Collapse
Affiliation(s)
- Liting Deng
- Department of Molecular Sciences, Faculty of Science and Engineering, Macquarie University, Sydney, New South Wales, Australia
| | - Vivek K Gupta
- Faculty of Medicine and Health and Human Sciences, Macquarie University, Sydney, New South Wales, Australia
| | - Yunqi Wu
- Department of Molecular Sciences, Faculty of Science and Engineering, Macquarie University, Sydney, New South Wales, Australia
| | - Kanishka Pushpitha
- Faculty of Medicine and Health and Human Sciences, Macquarie University, Sydney, New South Wales, Australia
| | - Nitin Chitranshi
- Faculty of Medicine and Health and Human Sciences, Macquarie University, Sydney, New South Wales, Australia
| | - Veer B Gupta
- School of Medicine, Deakin University, Geelong, Victoria, Australia
| | - Matthew J Fitzhenry
- Australian Proteome Analysis Facility (APAF), Macquarie University, Sydney, New South Wales, Australia
| | | | - Tim Karl
- School of Medicine, Western Sydney University, Penrith, New South Wales, Australia
| | - Ghasem Hosseini Salekdeh
- Department of Molecular Sciences, Faculty of Science and Engineering, Macquarie University, Sydney, New South Wales, Australia
| | - Stuart L Graham
- Faculty of Medicine and Health and Human Sciences, Macquarie University, Sydney, New South Wales, Australia
| | - Paul A Haynes
- Department of Molecular Sciences, Faculty of Science and Engineering, Macquarie University, Sydney, New South Wales, Australia
| | - Mehdi Mirzaei
- Faculty of Medicine and Health and Human Sciences, Macquarie University, Sydney, New South Wales, Australia
| |
Collapse
|
37
|
Latina V, Giacovazzo G, Cordella F, Balzamino BO, Micera A, Varano M, Marchetti C, Malerba F, Florio R, Ercole BB, La Regina F, Atlante A, Coccurello R, Di Angelantonio S, Calissano P, Amadoro G. Systemic delivery of a specific antibody targeting the pathological N-terminal truncated tau peptide reduces retinal degeneration in a mouse model of Alzheimer's Disease. Acta Neuropathol Commun 2021; 9:38. [PMID: 33750467 PMCID: PMC7942014 DOI: 10.1186/s40478-021-01138-1] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Accepted: 02/26/2021] [Indexed: 12/28/2022] Open
Abstract
Retina and optic nerve are sites of extra-cerebral manifestations of Alzheimer's Disease (AD). Amyloid-β (Aβ) plaques and neurofibrillary tangles of hyperphosphorylated tau protein are detected in eyes from AD patients and transgenic animals in correlation with inflammation, reduction of synapses, visual deficits, loss of retinal cells and nerve fiber. However, neither the pathological relevance of other post-translational tau modifications-such as truncation with generation of toxic fragments-nor the potential neuroprotective action induced by their in vivo clearance have been investigated in the context of AD retinal degeneration. We have recently developed a monoclonal tau antibody (12A12mAb) which selectively targets the neurotoxic 20-22 kDa NH2-derived peptide generated from pathological truncation at the N-terminal domain of tau without cross-reacting with its full-length normal protein. Previous studies have shown that 12A12mAb, when intravenously (i.v.)-injected into 6-month-old Tg2576 animals, markedly improves their AD-like, behavioural and neuropathological syndrome. By taking advantage of this well-established tau-directed immunization regimen, we found that 12A12mAb administration also exerts a beneficial action on biochemical, morphological and metabolic parameters (i.e. APP/Aβ processing, tau hyperphosphorylation, neuroinflammation, synaptic proteins, microtubule stability, mitochondria-based energy production, neuronal death) associated with ocular injury in the AD phenotype. These findings prospect translational implications in the AD field by: (1) showing for the first time that cleavage of tau takes part in several pathological changes occurring in vivo in affected retinas and vitreous bodies and that its deleterious effects are successfully antagonized by administration of the specific 12A12mAb; (2) shedding further insights on the tight connections between neurosensory retina and brain, in particular following tau-based immunotherapy. In our view, the parallel response we detected in this preclinical animal model, both in the eye and in the hippocampus, following i.v. 12A12mAb injection opens novel diagnostic and therapeutic avenues for the clinical management of cerebral and extracerebral AD signs in human beings.
Collapse
Affiliation(s)
- Valentina Latina
- European Brain Research Institute (EBRI), Viale Regina Elena 295, 00161 Rome, Italy
| | - Giacomo Giacovazzo
- IRCSS Santa Lucia Foundation, Via Fosso del Fiorano 64-65, 00143 Rome, Italy
| | - Federica Cordella
- Department of Physiology and Pharmacology, University of Rome La Sapienza, Piazzale Aldo Moro 5, 00185 Rome, Italy
- Center for Life Nanoscience, Istituto Italiano Di Tecnologia, Viale Regina Elena 291, 00161 Rome, Italy
| | - Bijorn Omar Balzamino
- Research Laboratories in Ophthalmology, IRCCS - Fondazione Bietti, Via Santo Stefano Rotondo, 6, 00184 Rome, Italy
| | - Alessandra Micera
- Research Laboratories in Ophthalmology, IRCCS - Fondazione Bietti, Via Santo Stefano Rotondo, 6, 00184 Rome, Italy
| | - Monica Varano
- Research Laboratories in Ophthalmology, IRCCS - Fondazione Bietti, Via Santo Stefano Rotondo, 6, 00184 Rome, Italy
| | - Cristina Marchetti
- European Brain Research Institute (EBRI), Viale Regina Elena 295, 00161 Rome, Italy
| | - Francesca Malerba
- 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
| | - Bruno Bruni Ercole
- 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
| | - Anna Atlante
- Institute of Biomembranes, Bioenergetics and Molecular Biotechnologies (IBIOM)-CNR, Via Amendola 122/O, 70126 Bari, Italy
| | - Roberto Coccurello
- IRCSS Santa Lucia Foundation, Via Fosso del Fiorano 64-65, 00143 Rome, Italy
- Institute for Complex System (ISC)-CNR, Via dei Taurini 19, 00185 Rome, Italy
| | - Silvia Di Angelantonio
- Department of Physiology and Pharmacology, University of Rome La Sapienza, Piazzale Aldo Moro 5, 00185 Rome, Italy
- Center for Life Nanoscience, Istituto Italiano Di Tecnologia, Viale Regina Elena 291, 00161 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
| |
Collapse
|
38
|
Wang L, Mao X. Role of Retinal Amyloid-β in Neurodegenerative Diseases: Overlapping Mechanisms and Emerging Clinical Applications. Int J Mol Sci 2021; 22:2360. [PMID: 33653000 PMCID: PMC7956232 DOI: 10.3390/ijms22052360] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Revised: 02/23/2021] [Accepted: 02/23/2021] [Indexed: 02/03/2023] Open
Abstract
Amyloid-β (Aβ) accumulations have been identified in the retina for neurodegeneration-associated disorders like Alzheimer's disease (AD), glaucoma, and age-related macular degeneration (AMD). Elevated retinal Aβ levels were associated with progressive retinal neurodegeneration, elevated cerebral Aβ accumulation, and increased disease severity with a decline in cognition and vision. Retinal Aβ accumulation and its pathological effects were demonstrated to occur prior to irreversible neurodegeneration, which highlights its potential in early disease detection and intervention. Using the retina as a model of the brain, recent studies have focused on characterizing retinal Aβ to determine its applicability for population-based screening of AD, which warrants a further understanding of how Aβ manifests between these disorders. While current treatments directly targeting Aβ accumulations have had limited results, continued exploration of Aβ-associated pathological pathways may yield new therapeutic targets for preserving cognition and vision. Here, we provide a review on the role of retinal Aβ manifestations in these distinct neurodegeneration-associated disorders. We also discuss the recent applications of retinal Aβ for AD screening and current clinical trial outcomes for Aβ-associated treatment approaches. Lastly, we explore potential future therapeutic targets based on overlapping mechanisms of pathophysiology in AD, glaucoma, and AMD.
Collapse
Affiliation(s)
- Liang Wang
- Miller School of Medicine, University of Miami, Miami, FL 33136, USA;
| | - Xiaobo Mao
- Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| |
Collapse
|
39
|
Salobrar-García E, López-Cuenca I, Sánchez-Puebla L, de Hoz R, Fernández-Albarral JA, Ramírez AI, Bravo-Ferrer I, Medina V, Moro MA, Saido TC, Saito T, Salazar JJ, Ramírez JM. Retinal Thickness Changes Over Time in a Murine AD Model APP NL-F/NL-F. Front Aging Neurosci 2021; 12:625642. [PMID: 33542683 PMCID: PMC7852550 DOI: 10.3389/fnagi.2020.625642] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2020] [Accepted: 12/15/2020] [Indexed: 11/13/2022] Open
Abstract
Background: Alzheimer's disease (AD) may present retinal changes before brain pathology, suggesting the retina as an accessible biomarker of AD. The present work is a diachronic study using spectral domain optical coherence tomography (SD-OCT) to determine the total retinal thickness and retinal nerve fiber layer (RNFL) thickness in an APPNL−F/NL−F mouse model of AD at 6, 9, 12, 15, 17, and 20 months old compared to wild type (WT) animals. Methods: Total retinal thickness and RNFL thickness were determined. The mean total retinal thickness was analyzed following the Early Treatment Diabetic Retinopathy Study sectors. RNFL was measured in six sectors of axonal ring scans around the optic nerve. Results: In the APPNL−F/NL−F group compared to WT animals, the total retinal thickness changes observed were the following: (i) At 6-months-old, a significant thinning in the outer temporal sector was observed; (ii) at 15-months-old a significant thinning in the inner temporal and in the inner and outer inferior retinal sectors was noticed; (iii) at 17-months-old, a significant thickening in the inferior and nasal sectors was found in both inner and outer rings; and (iv) at 20-months-old, a significant thinning in the inner ring of nasal, temporal, and inferior retina and in the outer ring of superior and temporal retina was seen. In RNFL thickness, there was significant thinning in the global analysis and in nasal and inner-temporal sectors at 6 months old. Thinning was also found in the supero-temporal and nasal sectors and global value at 20 months old. Conclusions: In the APPNL−F/NL−F AD model, the retinal thickness showed thinning, possibly produced by neurodegeneration alternating with thickening caused by deposits and neuroinflammation in some areas of the retina. These changes over time are similar to those observed in the human retina and could be a biomarker for AD. The APPNL−F/NL−F AD model may help us better understand the different retinal changes during the progression of AD.
Collapse
Affiliation(s)
- Elena Salobrar-García
- Ramon Castroviejo Ophthalmological Research Institute, Complutense University of Madrid, Madrid, Spain.,Department of Immunology, Ophthalmology and Ear, Nose, and Throat, Faculty of Optics and Optometry, Complutense University of Madrid, Madrid, Spain
| | - Inés López-Cuenca
- Ramon Castroviejo Ophthalmological Research Institute, Complutense University of Madrid, Madrid, Spain
| | - Lídia Sánchez-Puebla
- Ramon Castroviejo Ophthalmological Research Institute, Complutense University of Madrid, Madrid, Spain
| | - Rosa de Hoz
- Ramon Castroviejo Ophthalmological Research Institute, Complutense University of Madrid, Madrid, Spain.,Department of Immunology, Ophthalmology and Ear, Nose, and Throat, Faculty of Optics and Optometry, Complutense University of Madrid, Madrid, Spain
| | - José A Fernández-Albarral
- Ramon Castroviejo Ophthalmological Research Institute, Complutense University of Madrid, Madrid, Spain
| | - Ana I Ramírez
- Ramon Castroviejo Ophthalmological Research Institute, Complutense University of Madrid, Madrid, Spain.,Department of Immunology, Ophthalmology and Ear, Nose, and Throat, Faculty of Optics and Optometry, Complutense University of Madrid, Madrid, Spain
| | - Isabel Bravo-Ferrer
- Department of Pharmacology and Toxicology, Faculty of Medicine, Complutense University of Madrid, Madrid, Spain.,Edinburgh Medical School, UK Dementia Research Institute, University of Edinburgh, Edinburgh, United Kingdom
| | - Violeta Medina
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - María A Moro
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Takaomi C Saido
- Laboratory for Proteolytic Neuroscience, Brain Science Institute, RIKEN, Wako, Japan
| | - Takashi Saito
- Department of Neurocognitive Science, Institute of Brain Science, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Juan J Salazar
- Ramon Castroviejo Ophthalmological Research Institute, Complutense University of Madrid, Madrid, Spain.,Department of Immunology, Ophthalmology and Ear, Nose, and Throat, Faculty of Optics and Optometry, Complutense University of Madrid, Madrid, Spain
| | - José M Ramírez
- Ramon Castroviejo Ophthalmological Research Institute, Complutense University of Madrid, Madrid, Spain.,Department of Immunology, Ophthalmology and ENT, School of Medicine, Complutense University of Madrid, Madrid, Spain
| |
Collapse
|
40
|
Vit JP, Fuchs DT, Angel A, Levy A, Lamensdorf I, Black KL, Koronyo Y, Koronyo-Hamaoui M. Color and contrast vision in mouse models of aging and Alzheimer's disease using a novel visual-stimuli four-arm maze. Sci Rep 2021; 11:1255. [PMID: 33441984 PMCID: PMC7806734 DOI: 10.1038/s41598-021-80988-0] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2020] [Accepted: 12/31/2020] [Indexed: 12/14/2022] Open
Abstract
We introduce a novel visual-stimuli four-arm maze (ViS4M) equipped with spectrally- and intensity-controlled LED emitters and dynamic grayscale objects that relies on innate exploratory behavior to assess color and contrast vision in mice. Its application to detect visual impairments during normal aging and over the course of Alzheimer’s disease (AD) is evaluated in wild-type (WT) and transgenic APPSWE/PS1∆E9 murine models of AD (AD+) across an array of irradiance, chromaticity, and contrast conditions. Substantial color and contrast-mode alternation deficits appear in AD+ mice at an age when hippocampal-based memory and learning is still intact. Profiling of timespan, entries and transition patterns between the different arms uncovers variable AD-associated impairments in contrast sensitivity and color discrimination, reminiscent of tritanomalous defects documented in AD patients. Transition deficits are found in aged WT mice in the absence of alternation decline. Overall, ViS4M is a versatile, controlled device to measure color and contrast-related vision in aged and diseased mice.
Collapse
Affiliation(s)
- Jean-Philippe Vit
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, 127 S. San Vicente Blvd., Los Angeles, CA, 90048, USA.,Biobehavioral Research Core, 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
| | - Ariel Angel
- Pharmaseed Ltd., 9 Hamazmera St., 74047, Ness Ziona, Israel
| | - Aharon Levy
- Pharmaseed Ltd., 9 Hamazmera St., 74047, Ness Ziona, Israel
| | | | - Keith L Black
- 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
| | - Maya Koronyo-Hamaoui
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, 127 S. San Vicente Blvd., Los Angeles, CA, 90048, USA. .,Department of Neurosurgery, Maxine Dunitz Neurosurgical Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA.
| |
Collapse
|
41
|
Vandenabeele M, Veys L, Lemmens S, Hadoux X, Gelders G, Masin L, Serneels L, Theunis J, Saito T, Saido TC, Jayapala M, De Boever P, De Strooper B, Stalmans I, van Wijngaarden P, Moons L, De Groef L. The App NL-G-F mouse retina is a site for preclinical Alzheimer's disease diagnosis and research. Acta Neuropathol Commun 2021; 9:6. [PMID: 33407903 PMCID: PMC7788955 DOI: 10.1186/s40478-020-01102-5] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Accepted: 12/13/2020] [Indexed: 12/13/2022] Open
Abstract
In this study, we report the results of a comprehensive phenotyping of the retina of the AppNL-G-F mouse. We demonstrate that soluble Aβ accumulation is present in the retina of these mice early in life and progresses to Aβ plaque formation by midlife. This rising Aβ burden coincides with local microglia reactivity, astrogliosis, and abnormalities in retinal vein morphology. Electrophysiological recordings revealed signs of neuronal dysfunction yet no overt neurodegeneration was observed and visual performance outcomes were unaffected in the AppNL-G-F mouse. Furthermore, we show that hyperspectral imaging can be used to quantify retinal Aβ, underscoring its potential as a biomarker for AD diagnosis and monitoring. These findings suggest that the AppNL-G-F retina mimics the early, preclinical stages of AD, and, together with retinal imaging techniques, offers unique opportunities for drug discovery and fundamental research into preclinical AD.
Collapse
Affiliation(s)
- Marjan Vandenabeele
- Neural Circuit Development and Regeneration Research Group, Department of Biology, University of Leuven (KU Leuven), Naamsestraat 61, Box 2464, 3000, Leuven, Belgium
- Leuven Brain Institute, Leuven, Belgium
| | - Lien Veys
- Neural Circuit Development and Regeneration Research Group, Department of Biology, University of Leuven (KU Leuven), Naamsestraat 61, Box 2464, 3000, Leuven, Belgium
- Leuven Brain Institute, Leuven, Belgium
| | - Sophie Lemmens
- Department of Ophthalmology, University Hospitals Leuven, Leuven, Belgium
- Research Group Ophthalmology, Department of Neurosciences, KU Leuven, Leuven, Belgium
| | - Xavier Hadoux
- Centre for Eye Research Australia, Royal Victorian Eye and Ear Hospital, East Melbourne, Australia
- Ophthalmology, Department of Surgery, University of Melbourne, Parkville, Australia
| | - Géraldine Gelders
- Neural Circuit Development and Regeneration Research Group, Department of Biology, University of Leuven (KU Leuven), Naamsestraat 61, Box 2464, 3000, Leuven, Belgium
- Leuven Brain Institute, Leuven, Belgium
| | - Luca Masin
- Neural Circuit Development and Regeneration Research Group, Department of Biology, University of Leuven (KU Leuven), Naamsestraat 61, Box 2464, 3000, Leuven, Belgium
- Leuven Brain Institute, Leuven, Belgium
| | - Lutgarde Serneels
- Leuven Brain Institute, Leuven, Belgium
- Center for Brain and Disease Research, Flemish Institute for Biotechnology (VIB), Leuven, Belgium
- Laboratory for the Research of Neurodegenerative Diseases, Department of Neurosciences, KU Leuven, Leuven, Belgium
| | - Jan Theunis
- Health Unit, Flemish Institute for Technological Research (VITO), Mol, Belgium
| | - Takashi Saito
- Laboratory for Proteolytic Neuroscience, RIKEN Center for Brain Science, Saitama, Japan
- Department of Neurocognitive Science, Institute of Brain Science, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
- Interuniversity Microelectronics Centre (Imec), Leuven, Belgium
| | - Takaomi C Saido
- Laboratory for Proteolytic Neuroscience, RIKEN Center for Brain Science, Saitama, Japan
| | - Murali Jayapala
- Interuniversity Microelectronics Centre (Imec), Leuven, Belgium
| | - Patrick De Boever
- Center for Brain and Disease Research, Flemish Institute for Biotechnology (VIB), Leuven, Belgium
- Laboratory for the Research of Neurodegenerative Diseases, Department of Neurosciences, KU Leuven, Leuven, Belgium
- Center of Environmental Sciences, Hasselt University, Diepenbeek, Belgium
- Department of Biology, University of Antwerp, Wilrijk, Belgium
| | - Bart De Strooper
- Leuven Brain Institute, Leuven, Belgium
- Center for Brain and Disease Research, Flemish Institute for Biotechnology (VIB), Leuven, Belgium
- Laboratory for the Research of Neurodegenerative Diseases, Department of Neurosciences, KU Leuven, Leuven, Belgium
| | - Ingeborg Stalmans
- Department of Ophthalmology, University Hospitals Leuven, Leuven, Belgium
- Research Group Ophthalmology, Department of Neurosciences, KU Leuven, Leuven, Belgium
| | - Peter van Wijngaarden
- Centre for Eye Research Australia, Royal Victorian Eye and Ear Hospital, East Melbourne, Australia
- Ophthalmology, Department of Surgery, University of Melbourne, Parkville, Australia
| | - Lieve Moons
- Neural Circuit Development and Regeneration Research Group, Department of Biology, University of Leuven (KU Leuven), Naamsestraat 61, Box 2464, 3000, Leuven, Belgium
- Leuven Brain Institute, Leuven, Belgium
| | - Lies De Groef
- Neural Circuit Development and Regeneration Research Group, Department of Biology, University of Leuven (KU Leuven), Naamsestraat 61, Box 2464, 3000, Leuven, Belgium.
- Leuven Brain Institute, Leuven, Belgium.
| |
Collapse
|
42
|
Simons ES, Smith MA, Dengler-Crish CM, Crish SD. Retinal ganglion cell loss and gliosis in the retinofugal projection following intravitreal exposure to amyloid-beta. Neurobiol Dis 2021; 147:105146. [PMID: 33122075 DOI: 10.1016/j.nbd.2020.105146] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2020] [Revised: 10/09/2020] [Accepted: 10/23/2020] [Indexed: 01/07/2023] Open
Abstract
Pathological accumulations of amyloid-beta (Aβ) peptide are found in retina early in Alzheimer's disease, yet its effects on retinal neuronal structure remain unknown. To investigate this, we injected fibrillized Aβ1-42 protein into the eye of adult C57BL/6 J mice and analyzed the retina, optic nerve (ON), and the superior colliculus (SC), the primary retinal target in mice. We found that retinal Aβ exposure stimulated microglial activation and retinal ganglion cell (RGC) loss as early as 1-week post-injection. Pathology was not limited to the retina, but propagated into other areas of the central nervous system. Microgliosis spread throughout the retinal projection (retina, ON, and SC), with multiplex protein quantitation demonstrating an increase in endogenously produced Aβ in the ON and SC corresponding to the injected retinas. Surprisingly, this pathology spread to the opposite side, with unilateral Aβ eye injections driving increased Aβ levels, neuroinflammation, and RGC death in the opposite, un-injected retinal projection. As Aβ-mediated microglial activation has been shown to propagate Aβ pathology, we also investigated the role of the Aβ-binding microglial scavenger receptor CD36 in this pathology. Transgenic mice lacking the CD36 receptor were resistant to Aβ-induced inflammation and RGC death up to 2 weeks following exposure. These results indicate that Aβ pathology drives regional neuropathology in the retina and does not remain isolated to the affected eye, but spreads throughout the nervous system. Further, CD36 may serve as a promising target to prevent Aβ-mediated inflammatory damage.
Collapse
Affiliation(s)
- E S Simons
- Northeast Ohio Medical University, Rootstown, OH 44272, United States; Kent State Biomedical Sciences Graduate Program, Kent, OH 44240, United States
| | - M A Smith
- Northeast Ohio Medical University, Rootstown, OH 44272, United States; Kent State Biomedical Sciences Graduate Program, Kent, OH 44240, United States; Akron Children's Hospital, Rebecca D. Considine Research Institute, Akron, OH 44302, United States
| | - C M Dengler-Crish
- Northeast Ohio Medical University, Rootstown, OH 44272, United States; Kent State Biomedical Sciences Graduate Program, Kent, OH 44240, United States
| | - S D Crish
- Northeast Ohio Medical University, Rootstown, OH 44272, United States; Kent State Biomedical Sciences Graduate Program, Kent, OH 44240, United States.
| |
Collapse
|
43
|
Retinal capillary degeneration and blood-retinal barrier disruption in murine models of Alzheimer's disease. Acta Neuropathol Commun 2020; 8:202. [PMID: 33228786 PMCID: PMC7686701 DOI: 10.1186/s40478-020-01076-4] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2020] [Accepted: 11/11/2020] [Indexed: 01/17/2023] Open
Abstract
Extensive effort has been made studying retinal pathology in Alzheimer’s disease (AD) to improve early noninvasive diagnosis and treatment. Particularly relevant are vascular changes, which appear prominent in early brain pathogenesis and could predict cognitive decline. Recently, we identified platelet-derived growth factor receptor beta (PDGFRβ) deficiency and pericyte loss associated with vascular Aβ deposition in the neurosensory retina of mild cognitively impaired (MCI) and AD patients. However, the pathological mechanisms of retinal vascular changes and their possible relationships with vascular amyloidosis, pericyte loss, and blood-retinal barrier (BRB) integrity remain unknown. Here, we evaluated the retinas of transgenic APPSWE/PS1ΔE9 mouse models of AD (ADtg mice) and wild-type mice at different ages for capillary degeneration, PDGFRβ expression, vascular amyloidosis, permeability and inner BRB tight-junction molecules. Using a retinal vascular isolation technique followed by periodic acid-Schiff or immunofluorescent staining, we discovered significant retinal capillary degeneration in ADtg mice compared to age- and sex-matched wild-type mice (P < 0.0001). This small vessel degeneration reached significance in 8-month-old mice (P = 0.0035), with males more susceptible than females. Degeneration of retinal capillaries also progressively increased with age in healthy mice (P = 0.0145); however, the phenomenon was significantly worse during AD-like progression (P = 0.0001). A substantial vascular PDGFRβ deficiency (~ 50% reduction, P = 0.0017) along with prominent vascular Aβ deposition was further detected in the retina of ADtg mice, which inversely correlated with the extent of degenerated capillaries (Pearson’s r = − 0.8, P = 0.0016). Importantly, tight-junction alterations such as claudin-1 downregulation and increased BRB permeability, demonstrated in vivo by retinal fluorescein imaging and ex vivo following injection of FITC-dextran (2000 kD) and Texas Red-dextran (3 kD), were found in ADtg mice. Overall, the identification of age- and Alzheimer’s-dependent retinal capillary degeneration and compromised BRB integrity starting at early disease stages in ADtg mice could contribute to the development of novel targets for AD diagnosis and therapy.
Collapse
|
44
|
Retinal Degeneration and Alzheimer's Disease: An Evolving Link. Int J Mol Sci 2020; 21:ijms21197290. [PMID: 33023198 PMCID: PMC7582766 DOI: 10.3390/ijms21197290] [Citation(s) in RCA: 81] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2020] [Revised: 09/15/2020] [Accepted: 09/25/2020] [Indexed: 12/13/2022] Open
Abstract
Age-related macular degeneration (AMD) and glaucoma are degenerative conditions of the retina and a significant cause of irreversible blindness in developed countries. Alzheimer’s disease (AD), the most common dementia of the elderly, is often associated with AMD and glaucoma. The cardinal features of AD include extracellular accumulation of amyloid β (Aβ) and intracellular deposits of hyper-phosphorylated tau (p-tau). Neuroinflammation and brain iron dyshomeostasis accompany Aβ and p-tau deposits and, together, lead to progressive neuronal death and dementia. The accumulation of Aβ and iron in drusen, the hallmark of AMD, and Aβ and p-tau in retinal ganglion cells (RGC), the main retinal cell type implicated in glaucoma, and accompanying inflammation suggest overlapping pathology. Visual abnormalities are prominent in AD and are believed to develop before cognitive decline. Some are caused by degeneration of the visual cortex, while others are due to RGC loss or AMD-associated retinal degeneration. Here, we review recent information on Aβ, p-tau, chronic inflammation, and iron dyshomeostasis as common pathogenic mechanisms linking the three degenerative conditions, and iron chelation as a common therapeutic option for these disorders. Additionally discussed is the role of prion protein, infamous for prion disorders, in Aβ-mediated toxicity and, paradoxically, in neuroprotection.
Collapse
|
45
|
Das R, Chinnathambi S. Actin-mediated Microglial Chemotaxis via G-Protein Coupled Purinergic Receptor in Alzheimer's Disease. Neuroscience 2020; 448:325-336. [PMID: 32941933 DOI: 10.1016/j.neuroscience.2020.09.024] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Revised: 09/05/2020] [Accepted: 09/07/2020] [Indexed: 02/08/2023]
Abstract
Alzheimer's disease (AD) is a neurodegenerative disease mainly associated with aging, oxidative stress and genetic mutations. There are two pathological proteins involved in AD; Amyloid-β peptide and microtubule-associated protein Tau (MAPT). The β- and γ-secretase enzyme cleaves the Amyloid precursor protein, which results in the formation of extracellular plaques in brain. While, Tau undergoes hyperphosphorylation and other post-translational modifications (PTMs), which eventually generates Tau oligomers, and intracellular neurofibrillary tangles (NFTs) in neurons. Moreover, the brain-resident glia and infiltrated macrophages elevate the level of CNS inflammation, which trigger the oxidative damage of neuronal circuits by reactive oxygen species (ROS) and Nitric oxide (NO). Microglia is the primary immune cell in the CNS, which is continuously surveilling the neuronal synapses and pathogen invasion. Microglia in the resting state is called 'Ramified', which possess long surveilling extensions with a small cell body. But, upon activation, microglia retracts the cellular extensions and transform into round migratory cells, called as 'Amoeboid' state. Activated microglia undergoes actin remodeling by forming lamellipodia and filopodia, which directs the migratory axis while podosomes formed are involved in extracellular matrix degradation for invasion. Protein-aggregates in malfunctioning synapses and in CNS milieu can be detected by microglia, which results in its activation and migration. Subsequently, the phagocytosis of synapses leads to the inflammatory burst and memory loss. The extracellular nucleotides released from damaged neurons and the cytokine-chemokine gradients allow the neighboring microglia and macrophages to migrate-infiltrate at the site of neuronal-damage. The ionotropic (P2XR) and metabotropic (P2YR) purinergic receptor recognize extracellular ATP/ADP, which propagates through the intracellular calcium signaling, chemotaxis, phagocytosis and inflammation. The P2Y receptors give 'find me' or 'eat me' signals to microglia to either migrate or phagocytose cellular debris. Further, the actin cytoskeleton helps microglia to mediate directed chemotaxis and neuronal repair during neurodegeneration. Hence, we aim to emphasize the connection between purinergic signaling and actin-driven mechanical movements of microglia for migration and inflammation in AD.
Collapse
Affiliation(s)
- Rashmi Das
- Neurobiology Group, Division of Biochemical Sciences, CSIR-National Chemical Laboratory, Dr. Homi Bhabha Road, 411008 Pune, India; Academy of Scientific and Innovative Research (AcSIR), 411008 Pune, India
| | - Subashchandrabose Chinnathambi
- Neurobiology Group, Division of Biochemical Sciences, CSIR-National Chemical Laboratory, Dr. Homi Bhabha Road, 411008 Pune, India; Academy of Scientific and Innovative Research (AcSIR), 411008 Pune, India.
| |
Collapse
|
46
|
Mirzaei N, Shi H, Oviatt M, Doustar J, Rentsendorj A, Fuchs DT, Sheyn J, Black KL, Koronyo Y, Koronyo-Hamaoui M. Alzheimer's Retinopathy: Seeing Disease in the Eyes. Front Neurosci 2020; 14:921. [PMID: 33041751 PMCID: PMC7523471 DOI: 10.3389/fnins.2020.00921] [Citation(s) in RCA: 78] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Accepted: 08/10/2020] [Indexed: 01/18/2023] Open
Abstract
The neurosensory retina emerges as a prominent site of Alzheimer's disease (AD) pathology. As a CNS extension of the brain, the neuro retina is easily accessible for noninvasive, high-resolution imaging. Studies have shown that along with cognitive decline, patients with mild cognitive impairment (MCI) and AD often suffer from visual impairments, abnormal electroretinogram patterns, and circadian rhythm disturbances that can, at least in part, be attributed to retinal damage. Over a decade ago, our group identified the main pathological hallmark of AD, amyloid β-protein (Aβ) plaques, in the retina of patients including early-stage clinical cases. Subsequent histological, biochemical and in vivo retinal imaging studies in animal models and in humans corroborated these findings and further revealed other signs of AD neuropathology in the retina. Among these signs, hyperphosphorylated tau, neuronal degeneration, retinal thinning, vascular abnormalities and gliosis were documented. Further, linear correlations between the severity of retinal and brain Aβ concentrations and plaque pathology were described. More recently, extensive retinal pericyte loss along with vascular platelet-derived growth factor receptor-β deficiency were discovered in postmortem retinas of MCI and AD patients. This progressive loss was closely associated with increased retinal vascular amyloidosis and predicted cerebral amyloid angiopathy scores. These studies brought excitement to the field of retinal exploration in AD. Indeed, many questions still remain open, such as queries related to the temporal progression of AD-related pathology in the retina compared to the brain, the relations between retinal and cerebral changes and whether retinal signs can predict cognitive decline. The extent to which AD affects the retina, including the susceptibility of certain topographical regions and cell types, is currently under intense investigation. Advances in retinal amyloid imaging, hyperspectral imaging, optical coherence tomography, and OCT-angiography encourage the use of such modalities to achieve more accurate, patient- and user-friendly, noninvasive detection and monitoring of AD. In this review, we summarize the current status in the field while addressing the many unknowns regarding Alzheimer's retinopathy.
Collapse
Affiliation(s)
- Nazanin Mirzaei
- Department of Neurosurgery, Maxine Dunitz Neurosurgical Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA, United States
| | - Haoshen Shi
- Department of Neurosurgery, Maxine Dunitz Neurosurgical Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA, United States
| | - Mia Oviatt
- Department of Neurosurgery, Maxine Dunitz Neurosurgical Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA, United States
| | - Jonah Doustar
- Department of Neurosurgery, Maxine Dunitz Neurosurgical Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA, United States
| | - Altan Rentsendorj
- Department of Neurosurgery, Maxine Dunitz Neurosurgical Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA, United States
| | - Dieu-Trang Fuchs
- Department of Neurosurgery, Maxine Dunitz Neurosurgical Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA, United States
| | - Julia Sheyn
- Department of Neurosurgery, Maxine Dunitz Neurosurgical Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA, United States
| | - Keith L. Black
- Department of Neurosurgery, Maxine Dunitz Neurosurgical Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA, United States
| | - Yosef Koronyo
- Department of Neurosurgery, Maxine Dunitz Neurosurgical Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA, United States
| | - Maya Koronyo-Hamaoui
- Department of Neurosurgery, Maxine Dunitz Neurosurgical Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA, United States
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, United States
| |
Collapse
|
47
|
Gupta VB, Chitranshi N, den Haan J, Mirzaei M, You Y, Lim JK, Basavarajappa D, Godinez A, Di Angelantonio S, Sachdev P, Salekdeh GH, Bouwman F, Graham S, Gupta V. Retinal changes in Alzheimer's disease- integrated prospects of imaging, functional and molecular advances. Prog Retin Eye Res 2020; 82:100899. [PMID: 32890742 DOI: 10.1016/j.preteyeres.2020.100899] [Citation(s) in RCA: 83] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2020] [Revised: 08/24/2020] [Accepted: 08/27/2020] [Indexed: 12/31/2022]
Abstract
Alzheimer's Disease (AD) is a devastating neurodegenerative disorder of the brain, clinically characterised by cognitive deficits that gradually worsen over time. There is, at present, no established cure, or disease-modifying treatments for AD. As life expectancy increases globally, the number of individuals suffering from the disease is projected to increase substantially. Cumulative evidence indicates that AD neuropathological process is initiated several years, if not decades, before clinical signs are evident in patients, and diagnosis made. While several imaging, cognitive, CSF and blood-based biomarkers have been proposed for the early detection of AD; their sensitivity and specificity in the symptomatic stages is highly variable and it is difficult to justify their use in even earlier, pre-clinical stages of the disease. Research has identified potentially measurable functional, structural, metabolic and vascular changes in the retina during early stages of AD. Retina offers a distinctively accessible insight into brain pathology and current and developing ophthalmic technologies have provided us with the possibility of detecting and characterising subtle, disease-related changes. Recent human and animal model studies have further provided mechanistic insights into the biochemical pathways that are altered in the retina in disease, including amyloid and tau deposition. This information coupled with advances in molecular imaging has allowed attempts to monitor biochemical changes and protein aggregation pathology in the retina in AD. This review summarises the existing knowledge that informs our understanding of the impact of AD on the retina and highlights some of the gaps that need to be addressed. Future research will integrate molecular imaging innovation with functional and structural changes to enhance our knowledge of the AD pathophysiological mechanisms and establish the utility of monitoring retinal changes as a potential biomarker for AD.
Collapse
Affiliation(s)
- Veer B Gupta
- School of Medicine, Deakin University, VIC, Australia
| | - Nitin Chitranshi
- Faculty of Medicine Health and Human Sciences, Macquarie University, North Ryde, NSW, 2109, Australia
| | - Jurre den Haan
- Department of Neurology, Alzheimer Center Amsterdam, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, the Netherlands
| | - Mehdi Mirzaei
- Faculty of Medicine Health and Human Sciences, Macquarie University, North Ryde, NSW, 2109, Australia
| | - Yuyi You
- Faculty of Medicine Health and Human Sciences, Macquarie University, North Ryde, NSW, 2109, Australia
| | - Jeremiah Kh Lim
- Optometry and Vision Science, College of Nursing and Health Sciences, Bedford Park, South Australia, 5042, Australia
| | - Devaraj Basavarajappa
- Faculty of Medicine Health and Human Sciences, Macquarie University, North Ryde, NSW, 2109, Australia
| | - Angela Godinez
- Faculty of Medicine Health and Human Sciences, Macquarie University, North Ryde, NSW, 2109, Australia
| | - Silvia Di Angelantonio
- Center for Life Nanoscience, Istituto Italiano di Tecnologia, Rome, Italy; Department of Physiology and Pharmacology, Sapienza University of Rome, Rome, Italy
| | - Perminder Sachdev
- Centre for Healthy Brain and Ageing (CHeBA), School of Psychiatry, Faculty of Medicine, University of New South Wales, Sydney, New South Wales, Australia; Neuropsychiatric Institute, Prince of Wales Hospital, Sydney, New South Wales, Australia
| | - Ghasem H Salekdeh
- Department of Molecular Systems Biology, Cell Science Research Center, Royan, Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Femke Bouwman
- Department of Neurology, Alzheimer Center Amsterdam, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, the Netherlands
| | - Stuart Graham
- Faculty of Medicine Health and Human Sciences, Macquarie University, North Ryde, NSW, 2109, Australia; Save Sight Institute, Sydney University, Sydney, NSW, 2000, Australia.
| | - Vivek Gupta
- Faculty of Medicine Health and Human Sciences, Macquarie University, North Ryde, NSW, 2109, Australia.
| |
Collapse
|
48
|
Chibhabha F, Yaqi Y, Li F. Retinal involvement in Alzheimer's disease (AD): evidence and current progress on the non-invasive diagnosis and monitoring of AD-related pathology using the eye. Rev Neurosci 2020; 31:/j/revneuro.ahead-of-print/revneuro-2019-0119/revneuro-2019-0119.xml. [PMID: 32804680 DOI: 10.1515/revneuro-2019-0119] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2019] [Accepted: 06/04/2020] [Indexed: 12/22/2022]
Abstract
Alzheimer's disease (AD) is a common form of age-related dementia that mostly affects the aging population. Clinically, it is a disease characterized by impaired memory and progressive cognitive decline. Although the pathological hallmarks of AD have been traditionally described with a general confinement in the brain, recent studies have shown similar pathological changes in the retina, which is a developmental outgrowth of the forebrain. These AD-related neurodegenerative changes in the retina have been implicated to cause early visual problems in AD even before cognitive impairment becomes apparent. With recent advances in research, the commonly held view that AD-related cerebral pathology causes visual dysfunction through disruption of central visual pathways has been re-examined. Currently, several studies have already explored how AD manifests in the retina and the possibility of using the same retina as a window to non-invasively examine AD-related pathology in the brain. Non-invasive screening of AD through the retina has the potential to improve on early detection and management of the disease since the majority of AD cases are usually diagnosed very late. The purpose of this review is to provide evidence on the involvement of the retina in AD and to suggest a possible direction for future research into the non-invasive screening, diagnosis, and monitoring of AD using the retina.
Collapse
Affiliation(s)
- Fidelis Chibhabha
- Department of Anatomy and Neurobiology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou510080,China
- Department of Anatomy, Faculty of Medicine, Midlands State University, P. Bag 9055, Senga, Gweru, Zimbabwe
- and Guangdong Provincial Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080,China
| | - Yang Yaqi
- Department of Anatomy and Neurobiology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou510080,China
- and Guangdong Provincial Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080,China
| | - Feng Li
- Department of Anatomy and Neurobiology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou510080,China
- and Guangdong Provincial Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080,China
| |
Collapse
|
49
|
Lim JKH, Li QX, He Z, Vingrys AJ, Chinnery HR, Mullen J, Bui BV, Nguyen CTO. Retinal Functional and Structural Changes in the 5xFAD Mouse Model of Alzheimer's Disease. Front Neurosci 2020; 14:862. [PMID: 32903645 PMCID: PMC7438734 DOI: 10.3389/fnins.2020.00862] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Accepted: 07/24/2020] [Indexed: 12/18/2022] Open
Abstract
Alzheimer’s disease is characterized by the aberrant deposition of protein in the brain and is the leading cause of dementia worldwide. Increasingly, there have been reports of the presence of these protein hallmarks in the retina. In this study, we assayed the retina of 5xFAD mice, a transgenic model of amyloid deposition known to exhibit dementia-like symptoms with age. Using OCT, we found that the retinal nerve fiber layer was thinner in 5xFAD at 6, 12, and 17 months of age compared with wild-type littermates, but the inner plexiform layer was thicker at 6 months old. Retinal function showed reduced ganglion cell responses to light in 5xFAD at 6, 12, and 17 months of age. This functional loss was observed in the outer retina at 17 months of age but not in younger mice. We showed using immunohistochemistry and ELISA that soluble and insoluble amyloid was present in the retina and brain at all ages. In conclusion, we report that amyloid is present in brain and retina of 5xFAD mice and that the pattern of neuronal dysfunction occurs in the inner retina at the early ages and progresses to encompass the outer retina with age. This implies that the inner retina is more sensitive to amyloid changes in early disease and that the outer retina is also affected with disease progression.
Collapse
Affiliation(s)
- Jeremiah K H Lim
- Department of Optometry and Vision Sciences, University of Melbourne, Parkville, VIC, Australia.,Optometry and Vision Science, College of Nursing and Health Sciences, Flinders University, Bedford Park, SA, Australia
| | - Qiao-Xin Li
- Florey Institute of Neuroscience and Mental Health, Parkville, VIC, Australia
| | - Zheng He
- Department of Optometry and Vision Sciences, University of Melbourne, Parkville, VIC, Australia
| | - Algis J Vingrys
- Department of Optometry and Vision Sciences, University of Melbourne, Parkville, VIC, Australia
| | - Holly R Chinnery
- Department of Optometry and Vision Sciences, University of Melbourne, Parkville, VIC, Australia
| | - Jamie Mullen
- AstraZeneca Neuroscience, Cambridge, MA, United States
| | - Bang V Bui
- Department of Optometry and Vision Sciences, University of Melbourne, Parkville, VIC, Australia
| | - Christine T O Nguyen
- Department of Optometry and Vision Sciences, University of Melbourne, Parkville, VIC, Australia
| |
Collapse
|
50
|
Lee S, Jiang K, McIlmoyle B, To E, Xu QA, Hirsch-Reinshagen V, Mackenzie IR, Hsiung GYR, Eadie BD, Sarunic MV, Beg MF, Cui JZ, Matsubara JA. Amyloid Beta Immunoreactivity in the Retinal Ganglion Cell Layer of the Alzheimer's Eye. Front Neurosci 2020; 14:758. [PMID: 32848548 PMCID: PMC7412634 DOI: 10.3389/fnins.2020.00758] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Accepted: 06/29/2020] [Indexed: 01/04/2023] Open
Abstract
Alzheimer’s disease (AD) is the most prevalent form of dementia, accounting for 60–70% of all dementias. AD is often under-diagnosed and recognized only at a later, more advanced stage, and this delay in diagnosis has been suggested as a contributing factor in the numerous unsuccessful AD treatment trials. Although there is no known cure for AD, early diagnosis is important for disease management and care. A hallmark of AD is the deposition of amyloid-β (Aβ)-containing senile neuritic plaques and neurofibrillary tangles composed of hyperphosporylated tau in the brain. However, current in vivo methods to quantify Aβ in the brain are invasive, requiring radioactive tracers and positron emission tomography. Toward development of alternative methods to assess AD progression, we focus on the retinal manifestation of AD pathology. The retina is an extension of the central nervous system uniquely accessible to light-based, non-invasive ophthalmic imaging. However, earlier studies in human retina indicate that the literature is divided on the presence of Aβ in the AD retina. To help resolve this disparity, this study assessed retinal tissues from neuropathologically confirmed AD cases to determine the regional distribution of Aβ in retinal wholemounts and to inform on future retinal image studies targeting Aβ. Concurrent post-mortem brain tissues were also collected. Neuropathological cortical assessments including neuritic plaque (NP) scores and cerebral amyloid angiopathy (CAA) were correlated with retinal Aβ using immunohistochemistry, confocal microscopy, and quantitative image analysis. Aβ load was compared between AD and control (non-AD) eyes. Our results indicate that levels of intracellular and extracellular Aβ retinal deposits were significantly higher in AD than controls. Mid-peripheral Aβ levels were greater than central retina in both AD and control eyes. In AD retina, higher intracellular Aβ was associated with lower NP score, while higher extracellular Aβ was associated with higher CAA score. Our data support the feasibility of using the retinal tissue to assess ocular Aβ as a surrogate measure of Aβ in the brain of individuals with AD. Specifically, mid-peripheral retina possesses more Aβ deposition than central retina, and thus may be the optimal location for future in vivo ocular imaging.
Collapse
Affiliation(s)
- Sieun Lee
- Department of Ophthalmology and Visual Sciences, Faculty of Medicine, The University of British Columbia, Vancouver, BC, Canada.,School of Engineering Science, Simon Fraser University, Burnaby, BC, Canada
| | - Kailun Jiang
- Department of Surgery, Division of Ophthalmology, University of Calgary, Calgary, AB, Canada
| | - Brandon McIlmoyle
- Department of Family Medicine, Queen's University, Kingston, ON, Canada
| | - Eleanor To
- Department of Ophthalmology and Visual Sciences, Faculty of Medicine, The University of British Columbia, Vancouver, BC, Canada
| | - Qinyuan Alis Xu
- Department of Ophthalmology and Visual Sciences, Faculty of Medicine, The University of British Columbia, Vancouver, BC, Canada
| | - Veronica Hirsch-Reinshagen
- Department of Pathology, Vancouver General Hospital, The University of British Columbia, Vancouver, BC, Canada
| | - Ian R Mackenzie
- Department of Pathology, Vancouver General Hospital, The University of British Columbia, Vancouver, BC, Canada
| | - Ging-Yuek R Hsiung
- Division of Neurology, Department of Medicine, Faculty of Medicine, The University of British Columbia, Vancouver, BC, Canada
| | - Brennan D Eadie
- Department of Ophthalmology and Visual Sciences, Dalhousie University, Halifax, NS, Canada
| | - Marinko V Sarunic
- School of Engineering Science, Simon Fraser University, Burnaby, BC, Canada
| | - Mirza Faisal Beg
- School of Engineering Science, Simon Fraser University, Burnaby, BC, Canada
| | - Jing Z Cui
- Department of Ophthalmology and Visual Sciences, Faculty of Medicine, The University of British Columbia, Vancouver, BC, Canada
| | - Joanne A Matsubara
- Department of Ophthalmology and Visual Sciences, Faculty of Medicine, The University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|