Bridging Retinal and Cerebral Neurodegeneration: A Focus on Crosslinks between Alzheimer–Perusini’s Disease and Retinal Dystrophies
Abstract
:1. Retinal Degeneration in Alzheimer’s Disease (AD)
2. Histopathological Alterations in Different Eye Cytotypes Due to β-Amyloid Production
3. Retinal Histopathological Abnormalities in AD Mouse Models
4. The Role of the Prion Protein in Retinal Allostasis
5. Different Microglial Phenotypes and Aβ Clearance
6. Brain–Retina Microglia Axes and Alzheimer–Perusini’s Continuum
7. The Retina as a Gateway to Early AD Diagnosis
8. Retinal Organoids (RO) and Other Culture Systems
9. Rhodopsin Quantification
10. Conclusions and Future Scenarios
- Enhancing the sensitivity and specificity of certain techniques (as ones mentioned in the previous paragraphs);
- Establishing definitive cut-offs and thresholds (which indicate when a certain accumulation of non-functioning rhodopsin or when a specific modification of the molecule is unequivocally indicative of an emerging neurodegenerative progression);
- Developing a comprehensive diagnostic framework that encompasses various stages and provides refined guidelines for individual evaluations;
- Ensuring global accessibility to advanced equipment and technologies.
Author Contributions
Funding
Institutional Review Board Statement
Informed Consent Statement
Data Availability Statement
Conflicts of Interest
References
- Moons, L.; De Groef, L. Multimodal retinal imaging to detect and understand Alzheimer’s and Parkinson’s disease. Curr. Opin. Neurobiol. 2022, 72, 1–7. [Google Scholar] [CrossRef] [PubMed]
- Shi, H.; Koronyo, Y.; Rentsendorj, A.; Fuchs, D.T.; Sheyn, J.; Black, K.L.; Mirzaei, N.; Koronyo-Hamaoui, M. Retinal Vasculopathy in Alzheimer’s Disease. Front. Neurosci. 2021, 15, 731614. [Google Scholar] [CrossRef] [PubMed]
- Yoon, S.P.; Grewal, D.S.; Thompson, A.C.; Polascik, B.W.; Dunn, C.; Burke, J.R.; Fekrat, S. Retinal Microvascular and Neurodegenerative Changes in Alzheimer’s Disease and Mild Cognitive Impairment Compared with Control Participants. Ophthalmol. Retin. 2019, 3, 489–499. [Google Scholar] [CrossRef]
- Ashok, A.; Singh, N.; Chaudhary, S.; Bellamkonda, V.; Kritikos, A.E.; Wise, A.S.; Rana, N.; McDonald, D.; Ayyagari, R. Retinal Degeneration and Alzheimer’s Disease: An Evolving Link. Int. J. Mol. Sci. 2020, 21, 7290. [Google Scholar] [CrossRef] [PubMed]
- Asanad, S.; Ross-Cisneros, F.N.; Nassisi, M.; Barron, E.; Karanjia, R.; Sadun, A.A. The Retina in Alzheimer’s Disease: Histomorphometric Analysis of an Ophthalmologic Biomarker. Investig. Ophthalmol. Vis. Sci. 2019, 60, 1491–1500. [Google Scholar] [CrossRef]
- Gupta, V.; Gupta, V.B.; Chitranshi, N.; Gangoda, S.; Vander Wall, R.; Abbasi, M.; Golzan, M.; Dheer, Y.; Shah, T.; Avolio, A.; et al. One protein, multiple pathologies: Multifaceted involvement of amyloid beta in neurodegenerative disorders of the brain and retina. Cell Mol. Life Sci. 2016, 73, 4279–4297. [Google Scholar] [CrossRef]
- Ukalovic, K.; Cao, S.; Lee, S.; Tang, Q.; Beg, M.F.; Sarunic, M.V.; Hsiung, G.R.; Mackenzie, I.R.; Hirsch-Reinshagen, V.; Cui, J.Z.; et al. Drusen in the Peripheral Retina of the Alzheimer’s Eye. Curr. Alzheimer Res. 2018, 15, 743–750. [Google Scholar] [CrossRef]
- Blanks, J.C.; Schmidt, S.Y.; Torigoe, Y.; Porrello, K.V.; Hinton, D.R.; Blanks, R.H. Retinal pathology in Alzheimer’s disease. II. Regional neuron loss and glial changes in GCL. Neurobiol. Aging 1996, 17, 385–395. [Google Scholar] [CrossRef]
- Guo, L.Y.; Alekseev, O.; Li, Y.; Song, Y.; Dunaief, J.L. Iron increases APP translation and amyloid-beta production in the retina. Exp. Eye Res. 2014, 129, 31–37. [Google Scholar] [CrossRef]
- Ohno-Matsui, K. Parallel findings in age-related macular degeneration and Alzheimer’s disease. Prog. Retin. Eye Res. 2011, 30, 217–238. [Google Scholar] [CrossRef] [PubMed]
- Ong, S.S.; Proia, A.D.; Whitson, H.E.; Farsiu, S.; Doraiswamy, P.M.; Lad, E.M. Ocular amyloid imaging at the crossroad of Alzheimer’s disease and age-related macular degeneration: Implications for diagnosis and therapy. J. Neurol. 2019, 266, 1566–1577. [Google Scholar] [CrossRef] [PubMed]
- Donato, L.; Morda, D.; Scimone, C.; Alibrandi, S.; D’Angelo, R.; Sidoti, A. How Many Alzheimer-Perusini’s Atypical Forms Do We Still Have to Discover? Biomedicines 2023, 11, 2035. [Google Scholar] [CrossRef] [PubMed]
- den Haan, J.; Morrema, T.H.J.; Verbraak, F.D.; de Boer, J.F.; Scheltens, P.; Rozemuller, A.J.; Bergen, A.A.B.; Bouwman, F.H.; Hoozemans, J.J. Amyloid-beta and phosphorylated tau in post-mortem Alzheimer’s disease retinas. Acta Neuropathol. Commun. 2018, 6, 147. [Google Scholar] [CrossRef]
- Koronyo, Y.; Biggs, D.; Barron, E.; Boyer, D.S.; Pearlman, J.A.; Au, W.J.; Kile, S.J.; Blanco, A.; Fuchs, D.T.; Ashfaq, A.; et al. Retinal amyloid pathology and proof-of-concept imaging trial in Alzheimer’s disease. JCI Insight 2017, 2, e93621. [Google Scholar] [CrossRef] [PubMed]
- Shah, T.M.; Gupta, S.M.; Chatterjee, P.; Campbell, M.; Martins, R.N. Beta-amyloid sequelae in the eye: A critical review on its diagnostic significance and clinical relevance in Alzheimer’s disease. Mol. Psychiatry 2017, 22, 353–363. [Google Scholar] [CrossRef]
- Cerquera-Jaramillo, M.A.; Nava-Mesa, M.O.; Gonzalez-Reyes, R.E.; Tellez-Conti, C.; de-la-Torre, A. Visual Features in Alzheimer’s Disease: From Basic Mechanisms to Clinical Overview. Neural Plast. 2018, 2018, 2941783. [Google Scholar] [CrossRef]
- Berisha, F.; Feke, G.T.; Trempe, C.L.; McMeel, J.W.; Schepens, C.L. Retinal abnormalities in early Alzheimer’s disease. Investig. Ophthalmol. Vis. Sci. 2007, 48, 2285–2289. [Google Scholar] [CrossRef] [PubMed]
- Liu, D.; Zhang, L.; Li, Z.; Zhang, X.; Wu, Y.; Yang, H.; Min, B.; Zhang, X.; Ma, D.; Lu, Y. Thinner changes of the retinal nerve fiber layer in patients with mild cognitive impairment and Alzheimer’s disease. BMC Neurol. 2015, 15, 14. [Google Scholar] [CrossRef] [PubMed]
- Marziani, E.; Pomati, S.; Ramolfo, P.; Cigada, M.; Giani, A.; Mariani, C.; Staurenghi, G. Evaluation of retinal nerve fiber layer and ganglion cell layer thickness in Alzheimer’s disease using spectral-domain optical coherence tomography. Investig. Ophthalmol. Vis. Sci. 2013, 54, 5953–5958. [Google Scholar] [CrossRef]
- Polo, V.; Rodrigo, M.J.; Garcia-Martin, E.; Otin, S.; Larrosa, J.M.; Fuertes, M.I.; Bambo, M.P.; Pablo, L.E.; Satue, M. Visual dysfunction and its correlation with retinal changes in patients with Alzheimer’s disease. Eye 2017, 31, 1034–1041. [Google Scholar] [CrossRef]
- Jones-Odeh, E.; Hammond, C.J. How strong is the relationship between glaucoma, the retinal nerve fibre layer, and neurodegenerative diseases such as Alzheimer’s disease and multiple sclerosis? Eye 2015, 29, 1270–1284. [Google Scholar] [CrossRef]
- Sivak, J.M. The aging eye: Common degenerative mechanisms between the Alzheimer’s brain and retinal disease. Investig. Ophthalmol. Vis. Sci. 2013, 54, 871–880. [Google Scholar] [CrossRef] [PubMed]
- Liao, H.; Zhu, Z.; Peng, Y. Potential Utility of Retinal Imaging for Alzheimer’s Disease: A Review. Front. Aging Neurosci. 2018, 10, 188. [Google Scholar] [CrossRef] [PubMed]
- Ngolab, J.; Honma, P.; Rissman, R.A. Reflections on the Utility of the Retina as a Biomarker for Alzheimer’s Disease: A Literature Review. Neurol. Ther. 2019, 8, 57–72. [Google Scholar] [CrossRef] [PubMed]
- Tsokolas, G.; Tsaousis, K.T.; Diakonis, V.F.; Matsou, A.; Tyradellis, S. Optical Coherence Tomography Angiography in Neurodegenerative Diseases: A Review. Eye Brain 2020, 12, 73–87. [Google Scholar] [CrossRef]
- Ratnayaka, J.A.; Serpell, L.C.; Lotery, A.J. Dementia of the eye: The role of amyloid beta in retinal degeneration. Eye 2015, 29, 1013–1026. [Google Scholar] [CrossRef]
- Liu, B.; Rasool, S.; Yang, Z.; Glabe, C.G.; Schreiber, S.S.; Ge, J.; Tan, Z. Amyloid-peptide vaccinations reduce beta-amyloid plaques but exacerbate vascular deposition and inflammation in the retina of Alzheimer’s transgenic mice. Am. J. Pathol. 2009, 175, 2099–2110. [Google Scholar] [CrossRef] [PubMed]
- Liu, C.; Zhang, C.W.; Zhou, Y.; Wong, W.Q.; Lee, L.C.; Ong, W.Y.; Yoon, S.O.; Hong, W.; Fu, X.Y.; Soong, T.W.; et al. APP upregulation contributes to retinal ganglion cell degeneration via JNK3. Cell Death Differ. 2018, 25, 663–678. [Google Scholar] [CrossRef]
- Zhao, Y.; Bhattacharjee, S.; Jones, B.M.; Hill, J.M.; Clement, C.; Sambamurti, K.; Dua, P.; Lukiw, W.J. Beta-Amyloid Precursor Protein (betaAPP) Processing in Alzheimer’s Disease (AD) and Age-Related Macular Degeneration (AMD). Mol. Neurobiol. 2015, 52, 533–544. [Google Scholar] [CrossRef]
- Prasad, T.; Zhu, P.; Verma, A.; Chakrabarty, P.; Rosario, A.M.; Golde, T.E.; Li, Q. Amyloid beta peptides overexpression in retinal pigment epithelial cells via AAV-mediated gene transfer mimics AMD-like pathology in mice. Sci. Rep. 2017, 7, 3222. [Google Scholar] [CrossRef]
- Rong, S.S.; Lee, B.Y.; Kuk, A.K.; Yu, X.T.; Li, S.S.; Li, J.; Guo, Y.; Yin, Y.; Osterbur, D.L.; Yam, J.C.S.; et al. Comorbidity of dementia and age-related macular degeneration calls for clinical awareness: A meta-analysis. Br. J. Ophthalmol. 2019, 103, 1777–1783. [Google Scholar] [CrossRef] [PubMed]
- Striebel, J.F.; Race, B.; Leung, J.M.; Schwartz, C.; Chesebro, B. Prion-induced photoreceptor degeneration begins with misfolded prion protein accumulation in cones at two distinct sites: Cilia and ribbon synapses. Acta Neuropathol. Commun. 2021, 9, 17. [Google Scholar] [CrossRef] [PubMed]
- Ornek, N.; Dag, E.; Ornek, K. Corneal sensitivity and tear function in neurodegenerative diseases. Curr. Eye Res. 2015, 40, 423–428. [Google Scholar] [CrossRef] [PubMed]
- Dong, Z.; Luo, A.; Gan, Y.; Li, J. Amyloid Beta Deposition Could Cause Corneal Epithelial Cell Degeneration Associated with Increasing Apoptosis in APPswePS1 Transgenic Mice. Curr. Eye Res. 2018, 43, 1326–1333. [Google Scholar] [CrossRef] [PubMed]
- Lee, E.J.; Kim, T.W.; Lee, D.S.; Kim, H.; Park, Y.H.; Kim, J.; Lee, J.W.; Kim, S. Increased CSF tau level is correlated with decreased lamina cribrosa thickness. Alzheimers Res. Ther. 2016, 8, 6. [Google Scholar] [CrossRef]
- Dragan, M.C.; Leonard, T.K.; Lozano, A.M.; McAndrews, M.P.; Ng, K.; Ryan, J.D.; Tang-Wai, D.F.; Wynn, J.S.; Hoffman, K.L. Pupillary responses and memory-guided visual search reveal age-related and Alzheimer’s-related memory decline. Behav. Brain Res. 2017, 322, 351–361. [Google Scholar] [CrossRef]
- Iijima, A.; Haida, M.; Ishikawa, N.; Ueno, A.; Minamitani, H.; Shinohara, Y. Re-evaluation of tropicamide in the pupillary response test for Alzheimer’s disease. Neurobiol. Aging 2003, 24, 789–796. [Google Scholar] [CrossRef] [PubMed]
- Scinto, L.F. ApoE allelic variability influences pupil response to cholinergic challenge and cognitive impairment. Genes. Brain Behav. 2007, 6, 209–215. [Google Scholar] [CrossRef]
- Kawasaki, A.; Ouanes, S.; Crippa, S.V.; Popp, J. Early-Stage Alzheimer’s Disease Does Not Alter Pupil Responses to Colored Light Stimuli. J. Alzheimers Dis. 2020, 75, 1273–1282. [Google Scholar] [CrossRef]
- Kremen, W.S.; Panizzon, M.S.; Elman, J.A.; Granholm, E.L.; Andreassen, O.A.; Dale, A.M.; Gillespie, N.A.; Gustavson, D.E.; Logue, M.W.; Lyons, M.J.; et al. Pupillary dilation responses as a midlife indicator of risk for Alzheimer’s disease: Association with Alzheimer’s disease polygenic risk. Neurobiol. Aging 2019, 83, 114–121. [Google Scholar] [CrossRef]
- Kerbage, C.; Sadowsky, C.H.; Jennings, D.; Cagle, G.D.; Hartung, P.D. Alzheimer’s disease diagnosis by detecting exogenous fluorescent signal of ligand bound to Beta amyloid in the lens of human eye: An exploratory study. Front. Neurol. 2013, 4, 62. [Google Scholar] [CrossRef] [PubMed]
- Melov, S.; Wolf, N.; Strozyk, D.; Doctrow, S.R.; Bush, A.I. Mice transgenic for Alzheimer disease beta-amyloid develop lens cataracts that are rescued by antioxidant treatment. Free Radic. Biol. Med. 2005, 38, 258–261. [Google Scholar] [CrossRef]
- Sun, S.W.; Nishioka, C.; Labib, W.; Liang, H.F. Axonal Terminals Exposed to Amyloid-beta May Not Lead to Pre-Synaptic Axonal Damage. J. Alzheimers Dis. 2015, 45, 1139–1148. [Google Scholar] [CrossRef] [PubMed]
- Nishioka, C.; Liang, H.F.; Barsamian, B.; Sun, S.W. Amyloid-beta induced retrograde axonal degeneration in a mouse tauopathy model. Neuroimage 2019, 189, 180–191. [Google Scholar] [CrossRef]
- Sadun, A.A.; Bassi, C.J. Optic nerve damage in Alzheimer’s disease. Ophthalmology 1990, 97, 9–17. [Google Scholar] [CrossRef] [PubMed]
- Bayer, A.U.; Keller, O.N.; Ferrari, F.; Maag, K.P. Association of glaucoma with neurodegenerative diseases with apoptotic cell death: Alzheimer’s disease and Parkinson’s disease. Am. J. Ophthalmol. 2002, 133, 135–137. [Google Scholar] [CrossRef] [PubMed]
- Wostyn, P.; Audenaert, K.; De Deyn, P.P. Alzheimer’s disease-related changes in diseases characterized by elevation of intracranial or intraocular pressure. Clin. Neurol. Neurosurg. 2008, 110, 101–109. [Google Scholar] [CrossRef] [PubMed]
- Lenoir, H.; Sieroff, E. Visual perceptual disorders in Alzheimer’s disease. Geriatr. Psychol. Neuropsychiatr. Vieil. 2019, 17, 307–316. [Google Scholar] [CrossRef]
- Wright, L.M.; Stein, T.D.; Jun, G.; Chung, J.; McConnell, K.; Fiorello, M.; Siegel, N.; Ness, S.; Xia, W.; Turner, K.L.; et al. Association of Cognitive Function with Amyloid-beta and Tau Proteins in the Vitreous Humor. J. Alzheimers Dis. 2019, 68, 1429–1438. [Google Scholar] [CrossRef]
- Hart, N.J.; Koronyo, Y.; Black, K.L.; Koronyo-Hamaoui, M. Ocular indicators of Alzheimer’s: Exploring disease in the retina. Acta Neuropathol. 2016, 132, 767–787. [Google Scholar] [CrossRef]
- Ho, W.L.; Leung, Y.; Tsang, A.W.; So, K.F.; Chiu, K.; Chang, R.C. Review: Tauopathy in the retina and optic nerve: Does it shadow pathological changes in the brain? Mol. Vis. 2012, 18, 2700–2710. [Google Scholar] [PubMed]
- Chiasseu, M.; Cueva Vargas, J.L.; Destroismaisons, L.; Vande Velde, C.; Leclerc, N.; Di Polo, A. Tau Accumulation, Altered Phosphorylation, and Missorting Promote Neurodegeneration in Glaucoma. J. Neurosci. 2016, 36, 5785–5798. [Google Scholar] [CrossRef] [PubMed]
- Rhein, V.; Song, X.; Wiesner, A.; Ittner, L.M.; Baysang, G.; Meier, F.; Ozmen, L.; Bluethmann, H.; Drose, S.; Brandt, U.; et al. Amyloid-beta and tau synergistically impair the oxidative phosphorylation system in triple transgenic Alzheimer’s disease mice. Proc. Natl. Acad. Sci. USA 2009, 106, 20057–20062. [Google Scholar] [CrossRef] [PubMed]
- Michael, R.; Otto, C.; Lenferink, A.; Gelpi, E.; Montenegro, G.A.; Rosandic, J.; Tresserra, F.; Barraquer, R.I.; Vrensen, G.F. Absence of amyloid-beta in lenses of Alzheimer patients: A confocal Raman microspectroscopic study. Exp. Eye Res. 2014, 119, 44–53. [Google Scholar] [CrossRef] [PubMed]
- Choi, S.I.; Lee, B.; Woo, J.H.; Jeong, J.B.; Jun, I.; Kim, E.K. APP processing and metabolism in corneal fibroblasts and epithelium as a potential biomarker for Alzheimer’s disease. Exp. Eye Res. 2019, 182, 167–174. [Google Scholar] [CrossRef] [PubMed]
- Einarsdottir, A.B.; Hardarson, S.H.; Kristjansdottir, J.V.; Bragason, D.T.; Snaedal, J.; Stefansson, E. Retinal oximetry imaging in Alzheimer’s disease. J. Alzheimers Dis. 2016, 49, 79–83. [Google Scholar] [CrossRef]
- Olafsdottir, O.B.; Saevarsdottir, H.S.; Hardarson, S.H.; Hannesdottir, K.H.; Traustadottir, V.D.; Karlsson, R.A.; Einarsdottir, A.B.; Jonsdottir, K.D.; Stefansson, E.; Snaedal, J. Retinal oxygen metabolism in patients with mild cognitive impairment. Alzheimers Dement 2018, 10, 340–345. [Google Scholar] [CrossRef]
- Koronyo, Y.; Salumbides, B.C.; Black, K.L.; Koronyo-Hamaoui, M. Alzheimer’s disease in the retina: Imaging retinal abeta plaques for early diagnosis and therapy assessment. Neurodegener. Dis. 2012, 10, 285–293. [Google Scholar] [CrossRef] [PubMed]
- Cheung, C.Y.; Ong, Y.T.; Ikram, M.K.; Ong, S.Y.; Li, X.; Hilal, S.; Catindig, J.A.; Venketasubramanian, N.; Yap, P.; Seow, D.; et al. Microvascular network alterations in the retina of patients with Alzheimer’s disease. Alzheimers Dement. 2014, 10, 135–142. [Google Scholar] [CrossRef]
- Frost, S.; Kanagasingam, Y.; Sohrabi, H.; Vignarajan, J.; Bourgeat, P.; Salvado, O.; Villemagne, V.; Rowe, C.C.; Macaulay, S.L.; Szoeke, C.; et al. Retinal vascular biomarkers for early detection and monitoring of Alzheimer’s disease. Transl. Psychiatry 2013, 3, e233. [Google Scholar] [CrossRef]
- Shi, H.; Koronyo, Y.; Rentsendorj, A.; Regis, G.C.; Sheyn, J.; Fuchs, D.T.; Kramerov, A.A.; Ljubimov, A.V.; Dumitrascu, O.M.; Rodriguez, A.R.; et al. Identification of early pericyte loss and vascular amyloidosis in Alzheimer’s disease retina. Acta Neuropathol. 2020, 139, 813–836. [Google Scholar] [CrossRef] [PubMed]
- Liu, S.S.; Zhu, S.Q. [Correlation between Alzheimer disease and cataract]. Zhonghua Yan Ke Za Zhi 2017, 53, 314–316. [Google Scholar] [CrossRef] [PubMed]
- Frederikse, P.H.; Zigler, J.S., Jr. Presenilin expression in the ocular lens. Curr. Eye Res. 1998, 17, 947–952. [Google Scholar] [CrossRef]
- Ho, C.Y.; Troncoso, J.C.; Knox, D.; Stark, W.; Eberhart, C.G. Beta-amyloid, phospho-tau and alpha-synuclein deposits similar to those in the brain are not identified in the eyes of Alzheimer’s and Parkinson’s disease patients. Brain Pathol. 2014, 24, 25–32. [Google Scholar] [CrossRef] [PubMed]
- Kwak, D.E.; Ko, T.; Koh, H.S.; Ji, Y.W.; Shin, J.; Kim, K.; Kim, H.Y.; Lee, H.K.; Kim, Y. Alterations of aqueous humor Abeta levels in Abeta-infused and transgenic mouse models of Alzheimer disease. PLoS ONE 2020, 15, e0227618. [Google Scholar] [CrossRef] [PubMed]
- Prakasam, A.; Muthuswamy, A.; Ablonczy, Z.; Greig, N.H.; Fauq, A.; Rao, K.J.; Pappolla, M.A.; Sambamurti, K. Differential accumulation of secreted AbetaPP metabolites in ocular fluids. J. Alzheimers Dis. 2010, 20, 1243–1253. [Google Scholar] [CrossRef] [PubMed]
- Cunha, J.P.; Proenca, R.; Dias-Santos, A.; Melancia, D.; Almeida, R.; Aguas, H.; Santos, B.O.; Alves, M.; Ferreira, J.; Papoila, A.L.; et al. Choroidal thinning: Alzheimer’s disease and aging. Alzheimers Dement 2017, 8, 11–17. [Google Scholar] [CrossRef]
- Trebbastoni, A.; Marcelli, M.; Mallone, F.; D’Antonio, F.; Imbriano, L.; Campanelli, A.; de Lena, C.; Gharbiya, M. Attenuation of Choroidal Thickness in Patients with Alzheimer Disease: Evidence From an Italian Prospective Study. Alzheimer Dis. Assoc. Disord. 2017, 31, 128–134. [Google Scholar] [CrossRef]
- Rizzo, M.; Anderson, S.W.; Dawson, J.; Nawrot, M. Vision and cognition in Alzheimer’s disease. Neuropsychologia 2000, 38, 1157–1169. [Google Scholar] [CrossRef] [PubMed]
- Chiu, K.; Chan, T.F.; Wu, A.; Leung, I.Y.; So, K.F.; Chang, R.C. Neurodegeneration of the retina in mouse models of Alzheimer’s disease: What can we learn from the retina? Age 2012, 34, 633–649. [Google Scholar] [CrossRef] [PubMed]
- Guo, L.; Salt, T.E.; Luong, V.; Wood, N.; Cheung, W.; Maass, A.; Ferrari, G.; Russo-Marie, F.; Sillito, A.M.; Cheetham, M.E.; et al. Targeting amyloid-beta in glaucoma treatment. Proc. Natl. Acad. Sci. USA 2007, 104, 13444–13449. [Google Scholar] [CrossRef]
- Edwards, M.M.; Rodriguez, J.J.; Gutierrez-Lanza, R.; Yates, J.; Verkhratsky, A.; Lutty, G.A. Retinal macroglia changes in a triple transgenic mouse model of Alzheimer’s disease. Exp. Eye Res. 2014, 127, 252–260. [Google Scholar] [CrossRef]
- Joly, S.; Lamoureux, S.; Pernet, V. Nonamyloidogenic processing of amyloid beta precursor protein is associated with retinal function improvement in aging male APP(swe)/PS1DeltaE9 mice. Neurobiol. Aging 2017, 53, 181–191. [Google Scholar] [CrossRef] [PubMed]
- Walsh, D.T.; Bresciani, L.; Saunders, D.; Manca, M.F.; Jen, A.; Gentleman, S.M.; Jen, L.S. Amyloid beta peptide causes chronic glial cell activation and neuro-degeneration after intravitreal injection. Neuropathol. Appl. Neurobiol. 2005, 31, 491–502. [Google Scholar] [CrossRef] [PubMed]
- Chiasseu, M.; Alarcon-Martinez, L.; Belforte, N.; Quintero, H.; Dotigny, F.; Destroismaisons, L.; Vande Velde, C.; Panayi, F.; Louis, C.; Di Polo, A. Tau accumulation in the retina promotes early neuronal dysfunction and precedes brain pathology in a mouse model of Alzheimer’s disease. Mol. Neurodegener. 2017, 12, 58. [Google Scholar] [CrossRef] [PubMed]
- Jarosz-Griffiths, H.H.; Noble, E.; Rushworth, J.V.; Hooper, N.M. Amyloid-beta Receptors: The Good, the Bad, and the Prion Protein. J. Biol. Chem. 2016, 291, 3174–3183. [Google Scholar] [CrossRef] [PubMed]
- Lauren, J.; Gimbel, D.A.; Nygaard, H.B.; Gilbert, J.W.; Strittmatter, S.M. Cellular prion protein mediates impairment of synaptic plasticity by amyloid-beta oligomers. Nature 2009, 457, 1128–1132. [Google Scholar] [CrossRef]
- Nygaard, H.B.; van Dyck, C.H.; Strittmatter, S.M. Fyn kinase inhibition as a novel therapy for Alzheimer’s disease. Alzheimers Res. Ther. 2014, 6, 8. [Google Scholar] [CrossRef] [PubMed]
- Altmeppen, H.C.; Puig, B.; Dohler, F.; Thurm, D.K.; Falker, C.; Krasemann, S.; Glatzel, M. Proteolytic processing of the prion protein in health and disease. Am. J. Neurodegener. Dis. 2012, 1, 15–31. [Google Scholar]
- Asai, M.; Hattori, C.; Szabo, B.; Sasagawa, N.; Maruyama, K.; Tanuma, S.; Ishiura, S. Putative function of ADAM9, ADAM10, and ADAM17 as APP alpha-secretase. Biochem. Biophys. Res. Commun. 2003, 301, 231–235. [Google Scholar] [CrossRef]
- Asthana, A.; Baksi, S.; Ashok, A.; Karmakar, S.; Mammadova, N.; Kokemuller, R.; Greenlee, M.H.; Kong, Q.; Singh, N. Prion protein facilitates retinal iron uptake and is cleaved at the beta-site: Implications for retinal iron homeostasis in prion disorders. Sci. Rep. 2017, 7, 9600. [Google Scholar] [CrossRef]
- Castle, A.R.; Gill, A.C. Physiological Functions of the Cellular Prion Protein. Front. Mol. Biosci. 2017, 4, 19. [Google Scholar] [CrossRef]
- Ezpeleta, J.; Boudet-Devaud, F.; Pietri, M.; Baudry, A.; Baudouin, V.; Alleaume-Butaux, A.; Dagoneau, N.; Kellermann, O.; Launay, J.M.; Schneider, B. Protective role of cellular prion protein against TNFalpha-mediated inflammation through TACE alpha-secretase. Sci. Rep. 2017, 7, 7671. [Google Scholar] [CrossRef]
- Singh, N. The role of iron in prion disease and other neurodegenerative diseases. PLoS Pathog. 2014, 10, e1004335. [Google Scholar] [CrossRef]
- Singh, N.; Singh, A.; Das, D.; Mohan, M.L. Redox control of prion and disease pathogenesis. Antioxid. Redox Signal 2010, 12, 1271–1294. [Google Scholar] [CrossRef] [PubMed]
- Ramana, K.V. Tumor necrosis factor-alpha converting enzyme: Implications for ocular inflammatory diseases. Int. J. Biochem. Cell Biol. 2010, 42, 1076–1079. [Google Scholar] [CrossRef] [PubMed]
- Frigg, R.; Wenzel, A.; Samardzija, M.; Oesch, B.; Wariwoda, H.; Navarini, A.A.; Seeliger, M.W.; Tanimoto, N.; Reme, C.; Grimm, C. The prion protein is neuroprotective against retinal degeneration in vivo. Exp. Eye Res. 2006, 83, 1350–1358. [Google Scholar] [CrossRef] [PubMed]
- Loubet, D.; Dakowski, C.; Pietri, M.; Pradines, E.; Bernard, S.; Callebert, J.; Ardila-Osorio, H.; Mouillet-Richard, S.; Launay, J.M.; Kellermann, O.; et al. Neuritogenesis: The prion protein controls beta1 integrin signaling activity. FASEB J. 2012, 26, 678–690. [Google Scholar] [CrossRef] [PubMed]
- Alleaume-Butaux, A.; Nicot, S.; Pietri, M.; Baudry, A.; Dakowski, C.; Tixador, P.; Ardila-Osorio, H.; Haeberle, A.M.; Bailly, Y.; Peyrin, J.M.; et al. Double-Edge Sword of Sustained ROCK Activation in Prion Diseases through Neuritogenesis Defects and Prion Accumulation. PLoS Pathog. 2015, 11, e1005073. [Google Scholar] [CrossRef] [PubMed]
- Kim, H.J.; Choi, H.S.; Park, J.H.; Kim, M.J.; Lee, H.G.; Petersen, R.B.; Kim, Y.S.; Park, J.B.; Choi, E.K. Regulation of RhoA activity by the cellular prion protein. Cell Death Dis. 2017, 8, e2668. [Google Scholar] [CrossRef] [PubMed]
- Bertuchi, F.R.; Bourgeon, D.M.; Landemberger, M.C.; Martins, V.R.; Cerchiaro, G. PrPC displays an essential protective role from oxidative stress in an astrocyte cell line derived from PrPC knockout mice. Biochem. Biophys. Res. Commun. 2012, 418, 27–32. [Google Scholar] [CrossRef] [PubMed]
- Dupiereux, I.; Falisse-Poirrier, N.; Zorzi, W.; Watt, N.T.; Thellin, O.; Zorzi, D.; Pierard, O.; Hooper, N.M.; Heinen, E.; Elmoualij, B. Protective effect of prion protein via the N-terminal region in mediating a protective effect on paraquat-induced oxidative injury in neuronal cells. J. Neurosci. Res. 2008, 86, 653–659. [Google Scholar] [CrossRef] [PubMed]
- Haigh, C.L.; Collins, S.J. Endoproteolytic cleavage as a molecular switch regulating and diversifying prion protein function. Neural Regen. Res. 2016, 11, 238–239. [Google Scholar] [CrossRef] [PubMed]
- Zeng, F.; Watt, N.T.; Walmsley, A.R.; Hooper, N.M. Tethering the N-terminus of the prion protein compromises the cellular response to oxidative stress. J. Neurochem. 2003, 84, 480–490. [Google Scholar] [CrossRef] [PubMed]
- Zeng, L.; Zou, W.; Wang, G. Cellular prion protein (PrP(C)) and its role in stress responses. Int. J. Clin. Exp. Med. 2015, 8, 8042–8050. [Google Scholar] [PubMed]
- Brown, D.R.; Clive, C.; Haswell, S.J. Antioxidant activity related to copper binding of native prion protein. J. Neurochem. 2001, 76, 69–76. [Google Scholar] [CrossRef] [PubMed]
- Singh, A.; Haldar, S.; Horback, K.; Tom, C.; Zhou, L.; Meyerson, H.; Singh, N. Prion protein regulates iron transport by functioning as a ferrireductase. J. Alzheimers Dis. 2013, 35, 541–552. [Google Scholar] [CrossRef]
- Wille, H.; Requena, J.R. The Structure of PrP(Sc) Prions. Pathogens 2018, 7, 20. [Google Scholar] [CrossRef] [PubMed]
- Ramirez, A.I.; de Hoz, R.; Salobrar-Garcia, E.; Salazar, J.J.; Rojas, B.; Ajoy, D.; Lopez-Cuenca, I.; Rojas, P.; Trivino, A.; Ramirez, J.M. The Role of Microglia in Retinal Neurodegeneration: Alzheimer’s Disease, Parkinson, and Glaucoma. Front. Aging Neurosci. 2017, 9, 214. [Google Scholar] [CrossRef]
- Surguchov, A.; McMahan, B.; Masliah, E.; Surgucheva, I. Synucleins in ocular tissues. J. Neurosci. Res. 2001, 65, 68–77. [Google Scholar] [CrossRef]
- Moore, K.L.; Persaud, T.V.; Torchia, M.G. The Developing Human: Clinically Oriented Embryology, 1st ed.; Elsevier: Amsterdam, The Netherlands, 2012. [Google Scholar]
- Alliot, F.; Godin, I.; Pessac, B. Microglia derive from progenitors, originating from the yolk sac, and which proliferate in the brain. Brain Res. Dev. Brain Res. 1999, 117, 145–152. [Google Scholar] [CrossRef]
- Ashwell, K. Microglia and cell death in the developing mouse cerebellum. Brain Res. Dev. Brain Res. 1990, 55, 219–230. [Google Scholar] [CrossRef]
- Li, F.; Jiang, D.; Samuel, M.A. Microglia in the developing retina. Neural Dev. 2019, 14, 12. [Google Scholar] [CrossRef] [PubMed]
- Ginhoux, F.; Greter, M.; Leboeuf, M.; Nandi, S.; See, P.; Gokhan, S.; Mehler, M.F.; Conway, S.J.; Ng, L.G.; Stanley, E.R.; et al. Fate mapping analysis reveals that adult microglia derive from primitive macrophages. Science 2010, 330, 841–845. [Google Scholar] [CrossRef] [PubMed]
- Diaz-Araya, C.M.; Provis, J.M.; Penfold, P.L.; Billson, F.A. Development of microglial topography in human retina. J. Comp. Neurol. 1995, 363, 53–68. [Google Scholar] [CrossRef] [PubMed]
- Santos, A.M.; Calvente, R.; Tassi, M.; Carrasco, M.C.; Martin-Oliva, D.; Marin-Teva, J.L.; Navascues, J.; Cuadros, M.A. Embryonic and postnatal development of microglial cells in the mouse retina. J. Comp. Neurol. 2008, 506, 224–239. [Google Scholar] [CrossRef] [PubMed]
- Ashwell, K.W.; Hollander, H.; Streit, W.; Stone, J. The appearance and distribution of microglia in the developing retina of the rat. Vis. Neurosci. 1989, 2, 437–448. [Google Scholar] [CrossRef]
- Martin-Estebane, M.; Navascues, J.; Sierra-Martin, A.; Martin-Guerrero, S.M.; Cuadros, M.A.; Carrasco, M.C.; Marin-Teva, J.L. Onset of microglial entry into developing quail retina coincides with increased expression of active caspase-3 and is mediated by extracellular ATP and UDP. PLoS ONE 2017, 12, e0182450. [Google Scholar] [CrossRef]
- Marin-Teva, J.L.; Calvente, R.; Cuadros, M.A.; Almendros, A.; Navascues, J. Circumferential migration of ameboid microglia in the margin of the developing quail retina. Glia 1999, 27, 226–238. [Google Scholar] [CrossRef]
- Provis, J.M.; Diaz, C.M.; Penfold, P.L. Microglia in human retina: A heterogeneous population with distinct ontogenies. Perspect. Dev. Neurobiol. 1996, 3, 213–222. [Google Scholar]
- Chen, L.; Yang, P.; Kijlstra, A. Distribution, markers, and functions of retinal microglia. Ocul. Immunol. Inflamm. 2002, 10, 27–39. [Google Scholar] [CrossRef] [PubMed]
- Pei, Y.F.; Rhodin, J.A. The prenatal development of the mouse eye. Anat. Rec. 1970, 168, 105–125. [Google Scholar] [CrossRef] [PubMed]
- Sernagor, E.; Eglen, S.J.; Wong, R.O. Development of retinal ganglion cell structure and function. Prog. Retin. Eye Res. 2001, 20, 139–174. [Google Scholar] [CrossRef] [PubMed]
- Silverman, S.M.; Wong, W.T. Microglia in the Retina: Roles in Development, Maturity, and Disease. Annu. Rev. Vis. Sci. 2018, 4, 45–77. [Google Scholar] [CrossRef] [PubMed]
- Walker, D.G.; Lue, L.F. Immune phenotypes of microglia in human neurodegenerative disease: Challenges to detecting microglial polarization in human brains. Alzheimers Res. Ther. 2015, 7, 56. [Google Scholar] [CrossRef]
- Luo, C.; Jian, C.; Liao, Y.; Huang, Q.; Wu, Y.; Liu, X.; Zou, D.; Wu, Y. The role of microglia in multiple sclerosis. Neuropsychiatr. Dis. Treat. 2017, 13, 1661–1667. [Google Scholar] [CrossRef] [PubMed]
- Akinrinmade, O.A.; Chetty, S.; Daramola, A.K.; Islam, M.U.; Thepen, T.; Barth, S. CD64: An Attractive Immunotherapeutic Target for M1-type Macrophage Mediated Chronic Inflammatory Diseases. Biomedicines 2017, 5, 56. [Google Scholar] [CrossRef] [PubMed]
- Holloway, O.G.; Canty, A.J.; King, A.E.; Ziebell, J.M. Rod microglia and their role in neurological diseases. Semin. Cell Dev. Biol. 2019, 94, 96–103. [Google Scholar] [CrossRef]
- Walker, F.R.; Beynon, S.B.; Jones, K.A.; Zhao, Z.; Kongsui, R.; Cairns, M.; Nilsson, M. Dynamic structural remodelling of microglia in health and disease: A review of the models, the signals and the mechanisms. Brain Behav. Immun. 2014, 37, 1–14. [Google Scholar] [CrossRef] [PubMed]
- Torres-Platas, S.G.; Comeau, S.; Rachalski, A.; Bo, G.D.; Cruceanu, C.; Turecki, G.; Giros, B.; Mechawar, N. Morphometric characterization of microglial phenotypes in human cerebral cortex. J. Neuroinflammation 2014, 11, 12. [Google Scholar] [CrossRef]
- Nimmerjahn, A.; Kirchhoff, F.; Helmchen, F. Resting microglial cells are highly dynamic surveillants of brain parenchyma in vivo. Science 2005, 308, 1314–1318. [Google Scholar] [CrossRef] [PubMed]
- Karperien, A.; Ahammer, H.; Jelinek, H.F. Quantitating the subtleties of microglial morphology with fractal analysis. Front. Cell Neurosci. 2013, 7, 3. [Google Scholar] [CrossRef] [PubMed]
- Davis, B.M.; Salinas-Navarro, M.; Cordeiro, M.F.; Moons, L.; De Groef, L. Characterizing microglia activation: A spatial statistics approach to maximize information extraction. Sci. Rep. 2017, 7, 1576. [Google Scholar] [CrossRef] [PubMed]
- Giordano, K.R.; Denman, C.R.; Dubisch, P.S.; Akhter, M.; Lifshitz, J. An update on the rod microglia variant in experimental and clinical brain injury and disease. Brain Commun. 2021, 3, fcaa227. [Google Scholar] [CrossRef] [PubMed]
- Taylor, S.E.; Morganti-Kossmann, C.; Lifshitz, J.; Ziebell, J.M. Rod microglia: A morphological definition. PLoS ONE 2014, 9, e97096. [Google Scholar] [CrossRef] [PubMed]
- de Hoz, R.; Gallego, B.I.; Ramirez, A.I.; Rojas, B.; Salazar, J.J.; Valiente-Soriano, F.J.; Aviles-Trigueros, M.; Villegas-Perez, M.P.; Vidal-Sanz, M.; Trivino, A.; et al. Rod-like microglia are restricted to eyes with laser-induced ocular hypertension but absent from the microglial changes in the contralateral untreated eye. PLoS ONE 2013, 8, e83733. [Google Scholar] [CrossRef]
- Yuan, T.F.; Liang, Y.X.; Peng, B.; Lin, B.; So, K.F. Local proliferation is the main source of rod microglia after optic nerve transection. Sci. Rep. 2015, 5, 10788. [Google Scholar] [CrossRef] [PubMed]
- Dubbelaar, M.L.; Kracht, L.; Eggen, B.J.L.; Boddeke, E. The Kaleidoscope of Microglial Phenotypes. Front. Immunol. 2018, 9, 1753. [Google Scholar] [CrossRef]
- Fernandez-Arjona, M.D.M.; Grondona, J.M.; Granados-Duran, P.; Fernandez-Llebrez, P.; Lopez-Avalos, M.D. Microglia Morphological Categorization in a Rat Model of Neuroinflammation by Hierarchical Cluster and Principal Components Analysis. Front. Cell Neurosci. 2017, 11, 235. [Google Scholar] [CrossRef]
- Okajima, T.; Tsuruta, F. Microglial dynamics during brain development. Neural Regen. Res. 2018, 13, 222–223. [Google Scholar] [CrossRef]
- Perez-Pouchoulen, M.; VanRyzin, J.W.; McCarthy, M.M. Morphological and Phagocytic Profile of Microglia in the Developing Rat Cerebellum. eNeuro 2015, 2, ENEURO.0036-15.2015. [Google Scholar] [CrossRef]
- Davis, E.J.; Foster, T.D.; Thomas, W.E. Cellular forms and functions of brain microglia. Brain Res. Bull. 1994, 34, 73–78. [Google Scholar] [CrossRef]
- Kreutzberg, G.W. Microglia: A sensor for pathological events in the CNS. Trends Neurosci. 1996, 19, 312–318. [Google Scholar] [CrossRef]
- Koso, H.; Tsuhako, A.; Lai, C.Y.; Baba, Y.; Otsu, M.; Ueno, K.; Nagasaki, M.; Suzuki, Y.; Watanabe, S. Conditional rod photoreceptor ablation reveals Sall1 as a microglial marker and regulator of microglial morphology in the retina. Glia 2016, 64, 2005–2024. [Google Scholar] [CrossRef] [PubMed]
- Bachstetter, A.D.; Ighodaro, E.T.; Hassoun, Y.; Aldeiri, D.; Neltner, J.H.; Patel, E.; Abner, E.L.; Nelson, P.T. Rod-shaped microglia morphology is associated with aging in 2 human autopsy series. Neurobiol. Aging 2017, 52, 98–105. [Google Scholar] [CrossRef] [PubMed]
- Zeng, H.Y.; Green, W.R.; Tso, M.O. Microglial activation in human diabetic retinopathy. Arch. Ophthalmol. 2008, 126, 227–232. [Google Scholar] [CrossRef] [PubMed]
- Zhao, L.; Zabel, M.K.; Wang, X.; Ma, W.; Shah, P.; Fariss, R.N.; Qian, H.; Parkhurst, C.N.; Gan, W.B.; Wong, W.T. Microglial phagocytosis of living photoreceptors contributes to inherited retinal degeneration. EMBO Mol. Med. 2015, 7, 1179–1197. [Google Scholar] [CrossRef]
- O’Koren, E.G.; Yu, C.; Klingeborn, M.; Wong, A.Y.W.; Prigge, C.L.; Mathew, R.; Kalnitsky, J.; Msallam, R.A.; Silvin, A.; Kay, J.N.; et al. Microglial Function Is Distinct in Different Anatomical Locations during Retinal Homeostasis and Degeneration. Immunity 2019, 50, 723–737.e727. [Google Scholar] [CrossRef]
- Tang, P.H.; Pierson, M.J.; Heuss, N.D.; Gregerson, D.S. A subpopulation of activated retinal macrophages selectively migrated to regions of cone photoreceptor stress, but had limited effect on cone death in a mouse model for type 2 Leber congenital amaurosis. Mol. Cell Neurosci. 2017, 85, 70–81. [Google Scholar] [CrossRef] [PubMed]
- Tan, Y.L.; Yuan, Y.; Tian, L. Microglial regional heterogeneity and its role in the brain. Mol. Psychiatry 2020, 25, 351–367. [Google Scholar] [CrossRef]
- Hendrickx, D.A.; Schuurman, K.G.; van Draanen, M.; Hamann, J.; Huitinga, I. Enhanced uptake of multiple sclerosis-derived myelin by THP-1 macrophages and primary human microglia. J. Neuroinflamm. 2014, 11, 64. [Google Scholar] [CrossRef] [PubMed]
- Choi, S.; Guo, L.; Cordeiro, M.F. Retinal and Brain Microglia in Multiple Sclerosis and Neurodegeneration. Cells 2021, 10, 1507. [Google Scholar] [CrossRef]
- Baik, S.H.; Kang, S.; Son, S.M.; Mook-Jung, I. Microglia contributes to plaque growth by cell death due to uptake of amyloid beta in the brain of Alzheimer’s disease mouse model. Glia 2016, 64, 2274–2290. [Google Scholar] [CrossRef]
- Cao, S.; Fisher, D.W.; Rodriguez, G.; Yu, T.; Dong, H. Comparisons of neuroinflammation, microglial activation, and degeneration of the locus coeruleus-norepinephrine system in APP/PS1 and aging mice. J. Neuroinflamm. 2021, 18, 10. [Google Scholar] [CrossRef] [PubMed]
- Grimaldi, A.; Brighi, C.; Peruzzi, G.; Ragozzino, D.; Bonanni, V.; Limatola, C.; Ruocco, G.; Di Angelantonio, S. Inflammation, neurodegeneration and protein aggregation in the retina as ocular biomarkers for Alzheimer’s disease in the 3xTg-AD mouse model. Cell Death Dis. 2018, 9, 685. [Google Scholar] [CrossRef]
- Vandenabeele, M.; Veys, L.; Lemmens, S.; Hadoux, X.; Gelders, G.; Masin, L.; Serneels, L.; Theunis, J.; Saito, T.; Saido, T.C.; et al. The App(NL-G-F) mouse retina is a site for preclinical Alzheimer’s disease diagnosis and research. Acta Neuropathol. Commun. 2021, 9, 6. [Google Scholar] [CrossRef] [PubMed]
- Yao, K.; Zu, H.B. Microglial polarization: Novel therapeutic mechanism against Alzheimer’s disease. Inflammopharmacology 2020, 28, 95–110. [Google Scholar] [CrossRef]
- Ma, W.; Cojocaru, R.; Gotoh, N.; Gieser, L.; Villasmil, R.; Cogliati, T.; Swaroop, A.; Wong, W.T. Gene expression changes in aging retinal microglia: Relationship to microglial support functions and regulation of activation. Neurobiol. Aging 2013, 34, 2310–2321. [Google Scholar] [CrossRef]
- Grimaldi, A.; Pediconi, N.; Oieni, F.; Pizzarelli, R.; Rosito, M.; Giubettini, M.; Santini, T.; Limatola, C.; Ruocco, G.; Ragozzino, D.; et al. Neuroinflammatory Processes, A1 Astrocyte Activation and Protein Aggregation in the Retina of Alzheimer’s Disease Patients, Possible Biomarkers for Early Diagnosis. Front. Neurosci. 2019, 13, 925. [Google Scholar] [CrossRef]
- Okunuki, Y.; Mukai, R.; Nakao, T.; Tabor, S.J.; Butovsky, O.; Dana, R.; Ksander, B.R.; Connor, K.M. Retinal microglia initiate neuroinflammation in ocular autoimmunity. Proc. Natl. Acad. Sci. USA 2019, 116, 9989–9998. [Google Scholar] [CrossRef]
- Koizumi, T.; Kerkhofs, D.; Mizuno, T.; Steinbusch, H.W.M.; Foulquier, S. Vessel-Associated Immune Cells in Cerebrovascular Diseases: From Perivascular Macrophages to Vessel-Associated Microglia. Front. Neurosci. 2019, 13, 1291. [Google Scholar] [CrossRef]
- Lopez-de-Eguileta, A.; Cervero, A.; Ruiz de Sabando, A.; Sanchez-Juan, P.; Casado, A. Ganglion Cell Layer Thinning in Alzheimer’s Disease. Medicina 2020, 56, 553. [Google Scholar] [CrossRef] [PubMed]
- Zhao, H.; Chang, R.; Che, H.; Wang, J.; Yang, L.; Fang, W.; Xia, Y.; Li, N.; Ma, Q.; Wang, X. Hyperphosphorylation of tau protein by calpain regulation in retina of Alzheimer’s disease transgenic mouse. Neurosci. Lett. 2013, 551, 12–16. [Google Scholar] [CrossRef]
- Chiquita, S.; Rodrigues-Neves, A.C.; Baptista, F.I.; Carecho, R.; Moreira, P.I.; Castelo-Branco, M.; Ambrosio, A.F. The Retina as a Window or Mirror of the Brain Changes Detected in Alzheimer’s Disease: Critical Aspects to Unravel. Mol. Neurobiol. 2019, 56, 5416–5435. [Google Scholar] [CrossRef] [PubMed]
- Markram, H. The blue brain project. Nat. Rev. Neurosci. 2006, 7, 153–160. [Google Scholar] [CrossRef]
- Sharpee, T.O.; Destexhe, A.; Kawato, M.; Sekulic, V.; Skinner, F.K.; Wojcik, D.K.; Chintaluri, C.; Cserpan, D.; Somogyvari, Z.; Kim, J.K.; et al. 25th Annual Computational Neuroscience Meeting: CNS-2016. BMC Neurosci. 2016, 17 (Suppl. S1), 54. [Google Scholar] [CrossRef] [PubMed]
- Ge, Y.J.; Xu, W.; Ou, Y.N.; Qu, Y.; Ma, Y.H.; Huang, Y.Y.; Shen, X.N.; Chen, S.D.; Tan, L.; Zhao, Q.H.; et al. Retinal biomarkers in Alzheimer’s disease and mild cognitive impairment: A systematic review and meta-analysis. Ageing Res. Rev. 2021, 69, 101361. [Google Scholar] [CrossRef] [PubMed]
- Lenahan, C.; Sanghavi, R.; Huang, L.; Zhang, J.H. Rhodopsin: A Potential Biomarker for Neurodegenerative Diseases. Front. Neurosci. 2020, 14, 326. [Google Scholar] [CrossRef] [PubMed]
- Cheignon, C.; Tomas, M.; Bonnefont-Rousselot, D.; Faller, P.; Hureau, C.; Collin, F. Oxidative stress and the amyloid beta peptide in Alzheimer’s disease. Redox Biol. 2018, 14, 450–464. [Google Scholar] [CrossRef] [PubMed]
- Fernandez-Albarral, J.A.; Salobrar-Garcia, E.; Martinez-Paramo, R.; Ramirez, A.I.; de Hoz, R.; Ramirez, J.M.; Salazar, J.J. Retinal glial changes in Alzheimer’s disease—A review. J. Optom. 2019, 12, 198–207. [Google Scholar] [CrossRef]
- Nilson, A.N.; English, K.C.; Gerson, J.E.; Barton Whittle, T.; Nicolas Crain, C.; Xue, J.; Sengupta, U.; Castillo-Carranza, D.L.; Zhang, W.; Gupta, P.; et al. Tau Oligomers Associate with Inflammation in the Brain and Retina of Tauopathy Mice and in Neurodegenerative Diseases. J. Alzheimers Dis. 2017, 55, 1083–1099. [Google Scholar] [CrossRef]
- Movio, M.I.; de Lima-Vasconcellos, T.H.; Dos Santos, G.B.; Echeverry, M.B.; Colombo, E.; Mattos, L.S.; Resende, R.R.; Kihara, A.H. Retinal organoids from human-induced pluripotent stem cells: From studying retinal dystrophies to early diagnosis of Alzheimer’s and Parkinson’s disease. Semin. Cell Dev. Biol. 2023, 144, 77–86. [Google Scholar] [CrossRef]
- Golzan, S.M.; Goozee, K.; Georgevsky, D.; Avolio, A.; Chatterjee, P.; Shen, K.; Gupta, V.; Chung, R.; Savage, G.; Orr, C.F.; et al. Retinal vascular and structural changes are associated with amyloid burden in the elderly: Ophthalmic biomarkers of preclinical Alzheimer’s disease. Alzheimers Res. Ther. 2017, 9, 13. [Google Scholar] [CrossRef] [PubMed]
- Gupta, V.B.; Chitranshi, N.; den Haan, J.; Mirzaei, M.; You, Y.; Lim, J.K.; Basavarajappa, D.; Godinez, A.; Di Angelantonio, S.; Sachdev, P.; et al. Retinal changes in Alzheimer’s disease- integrated prospects of imaging, functional and molecular advances. Prog. Retin. Eye Res. 2021, 82, 100899. [Google Scholar] [CrossRef]
- Johnson, E.C.B.; Dammer, E.B.; Duong, D.M.; Ping, L.; Zhou, M.; Yin, L.; Higginbotham, L.A.; Guajardo, A.; White, B.; Troncoso, J.C.; et al. Large-scale proteomic analysis of Alzheimer’s disease brain and cerebrospinal fluid reveals early changes in energy metabolism associated with microglia and astrocyte activation. Nat. Med. 2020, 26, 769–780. [Google Scholar] [CrossRef] [PubMed]
- Mirzaei, M.; Pushpitha, K.; Deng, L.; Chitranshi, N.; Gupta, V.; Rajput, R.; Mangani, A.B.; Dheer, Y.; Godinez, A.; McKay, M.J.; et al. Upregulation of Proteolytic Pathways and Altered Protein Biosynthesis Underlie Retinal Pathology in a Mouse Model of Alzheimer’s Disease. Mol. Neurobiol. 2019, 56, 6017–6034. [Google Scholar] [CrossRef] [PubMed]
- Penney, J.; Ralvenius, W.T.; Tsai, L.H. Modeling Alzheimer’s disease with iPSC-derived brain cells. Mol. Psychiatry 2020, 25, 148–167. [Google Scholar] [CrossRef] [PubMed]
- Artero Castro, A.; Rodriguez Jimenez, F.J.; Jendelova, P.; Erceg, S. Deciphering retinal diseases through the generation of three dimensional stem cell-derived organoids: Concise Review. Stem Cells 2019, 37, 1496–1504. [Google Scholar] [CrossRef] [PubMed]
- Brighi, C.; Cordella, F.; Chiriatti, L.; Soloperto, A.; Di Angelantonio, S. Retinal and Brain Organoids: Bridging the Gap Between in vivo Physiology and in vitro Micro-Physiology for the Study of Alzheimer’s Diseases. Front. Neurosci. 2020, 14, 655. [Google Scholar] [CrossRef]
- Jiang, H.; Wang, J.; Levin, B.E.; Baumel, B.S.; Camargo, C.J.; Signorile, J.F.; Rundek, T. Retinal Microvascular Alterations as the Biomarkers for Alzheimer Disease: Are We There Yet? J. Neuroophthalmol. 2021, 41, 251–260. [Google Scholar] [CrossRef] [PubMed]
- Xu, M.; Zhang, L.; Liu, G.; Jiang, N.; Zhou, W.; Zhang, Y. Pathological Changes in Alzheimer’s Disease Analyzed Using Induced Pluripotent Stem Cell-Derived Human Microglia-Like Cells. J. Alzheimers Dis. 2019, 67, 357–368. [Google Scholar] [CrossRef]
- Abud, E.M.; Ramirez, R.N.; Martinez, E.S.; Healy, L.M.; Nguyen, C.H.H.; Newman, S.A.; Yeromin, A.V.; Scarfone, V.M.; Marsh, S.E.; Fimbres, C.; et al. iPSC-Derived Human Microglia-like Cells to Study Neurological Diseases. Neuron 2017, 94, 278–293.e9. [Google Scholar] [CrossRef] [PubMed]
- Henstridge, C.M.; Spires-Jones, T.L. Modeling Alzheimer’s disease brains in vitro. Nat. Neurosci. 2018, 21, 899–900. [Google Scholar] [CrossRef] [PubMed]
- Campos, P.B.; Paulsen, B.S.; Rehen, S.K. Accelerating neuronal aging in in vitro model brain disorders: A focus on reactive oxygen species. Front. Aging Neurosci. 2014, 6, 292. [Google Scholar] [CrossRef] [PubMed]
- Grenier, K.; Kao, J.; Diamandis, P. Three-dimensional modeling of human neurodegeneration: Brain organoids coming of age. Mol. Psychiatry 2020, 25, 254–274. [Google Scholar] [CrossRef] [PubMed]
- Cideciyan, A.V.; Jacobson, S.G.; Aleman, T.S.; Gu, D.; Pearce-Kelling, S.E.; Sumaroka, A.; Acland, G.M.; Aguirre, G.D. In vivo dynamics of retinal injury and repair in the rhodopsin mutant dog model of human retinitis pigmentosa. Proc. Natl. Acad. Sci. USA 2005, 102, 5233–5238. [Google Scholar] [CrossRef] [PubMed]
- Xiong, B.; Bellen, H.J. Rhodopsin homeostasis and retinal degeneration: Lessons from the fly. Trends Neurosci. 2013, 36, 652–660. [Google Scholar] [CrossRef]
- Di Pierdomenico, J.; Garcia-Ayuso, D.; Pinilla, I.; Cuenca, N.; Vidal-Sanz, M.; Agudo-Barriuso, M.; Villegas-Perez, M.P. Early Events in Retinal Degeneration Caused by Rhodopsin Mutation or Pigment Epithelium Malfunction: Differences and Similarities. Front. Neuroanat. 2017, 11, 14. [Google Scholar] [CrossRef]
- Ni, J.D.; Baik, L.S.; Holmes, T.C.; Montell, C. A rhodopsin in the brain functions in circadian photoentrainment in Drosophila. Nature 2017, 545, 340–344. [Google Scholar] [CrossRef] [PubMed]
- Reeves, B.C.; Karimy, J.K.; Kundishora, A.J.; Mestre, H.; Cerci, H.M.; Matouk, C.; Alper, S.L.; Lundgaard, I.; Nedergaard, M.; Kahle, K.T. Glymphatic System Impairment in Alzheimer’s Disease and Idiopathic Normal Pressure Hydrocephalus. Trends Mol. Med. 2020, 26, 285–295. [Google Scholar] [CrossRef]
- Stojanovic, A.; Stitham, J.; Hwa, J. Critical role of transmembrane segment zinc binding in the structure and function of rhodopsin. J. Biol. Chem. 2004, 279, 35932–35941. [Google Scholar] [CrossRef]
- Ugarte, M.; Osborne, N.N. Zinc in the retina. Prog. Neurobiol. 2001, 64, 219–249. [Google Scholar] [CrossRef] [PubMed]
- del Valle, L.J.; Ramon, E.; Canavate, X.; Dias, P.; Garriga, P. Zinc-induced decrease of the thermal stability and regeneration of rhodopsin. J. Biol. Chem. 2003, 278, 4719–4724. [Google Scholar] [CrossRef] [PubMed]
- Das, N.; Raymick, J.; Sarkar, S. Role of metals in Alzheimer’s disease. Metab. Brain Dis. 2021, 36, 1627–1639. [Google Scholar] [CrossRef] [PubMed]
- Doustar, J.; Torbati, T.; Black, K.L.; Koronyo, Y.; Koronyo-Hamaoui, M. Optical Coherence Tomography in Alzheimer’s Disease and Other Neurodegenerative Diseases. Front. Neurol. 2017, 8, 701. [Google Scholar] [CrossRef] [PubMed]
- Ehler, M.; Dobrosotskaya, J.; Cunningham, D.; Wong, W.T.; Chew, E.Y.; Czaja, W.; Bonner, R.F. Modeling Photo-Bleaching Kinetics to Create High Resolution Maps of Rod Rhodopsin in the Human Retina. PLoS ONE 2015, 10, e0131881. [Google Scholar] [CrossRef]
- Jackson, G.R.; Owsley, C.; McGwin, G., Jr. Aging and dark adaptation. Vision. Res. 1999, 39, 3975–3982. [Google Scholar] [CrossRef]
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content. |
© 2023 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (https://creativecommons.org/licenses/by/4.0/).
Share and Cite
Donato, L.; Mordà, D.; Scimone, C.; Alibrandi, S.; D’Angelo, R.; Sidoti, A. Bridging Retinal and Cerebral Neurodegeneration: A Focus on Crosslinks between Alzheimer–Perusini’s Disease and Retinal Dystrophies. Biomedicines 2023, 11, 3258. https://doi.org/10.3390/biomedicines11123258
Donato L, Mordà D, Scimone C, Alibrandi S, D’Angelo R, Sidoti A. Bridging Retinal and Cerebral Neurodegeneration: A Focus on Crosslinks between Alzheimer–Perusini’s Disease and Retinal Dystrophies. Biomedicines. 2023; 11(12):3258. https://doi.org/10.3390/biomedicines11123258
Chicago/Turabian StyleDonato, Luigi, Domenico Mordà, Concetta Scimone, Simona Alibrandi, Rosalia D’Angelo, and Antonina Sidoti. 2023. "Bridging Retinal and Cerebral Neurodegeneration: A Focus on Crosslinks between Alzheimer–Perusini’s Disease and Retinal Dystrophies" Biomedicines 11, no. 12: 3258. https://doi.org/10.3390/biomedicines11123258
APA StyleDonato, L., Mordà, D., Scimone, C., Alibrandi, S., D’Angelo, R., & Sidoti, A. (2023). Bridging Retinal and Cerebral Neurodegeneration: A Focus on Crosslinks between Alzheimer–Perusini’s Disease and Retinal Dystrophies. Biomedicines, 11(12), 3258. https://doi.org/10.3390/biomedicines11123258