1
|
Ilina A, Khavinson V, Linkova N, Petukhov M. Neuroepigenetic Mechanisms of Action of Ultrashort Peptides in Alzheimer's Disease. Int J Mol Sci 2022; 23:ijms23084259. [PMID: 35457077 PMCID: PMC9032300 DOI: 10.3390/ijms23084259] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Revised: 04/07/2022] [Accepted: 04/09/2022] [Indexed: 12/23/2022] Open
Abstract
Epigenetic regulation of gene expression is necessary for maintaining higher-order cognitive functions (learning and memory). The current understanding of the role of epigenetics in the mechanism of Alzheimer’s disease (AD) is focused on DNA methylation, chromatin remodeling, histone modifications, and regulation of non-coding RNAs. The pathogenetic links of this disease are the misfolding and aggregation of tau protein and amyloid peptides, mitochondrial dysfunction, oxidative stress, impaired energy metabolism, destruction of the blood–brain barrier, and neuroinflammation, all of which lead to impaired synaptic plasticity and memory loss. Ultrashort peptides are promising neuroprotective compounds with a broad spectrum of activity and without reported side effects. The main aim of this review is to analyze the possible epigenetic mechanisms of the neuroprotective action of ultrashort peptides in AD. The review highlights the role of short peptides in the AD pathophysiology. We formulate the hypothesis that peptide regulation of gene expression can be mediated by the interaction of short peptides with histone proteins, cis- and transregulatory DNA elements and effector molecules (DNA/RNA-binding proteins and non-coding RNA). The development of therapeutic agents based on ultrashort peptides may offer a promising addition to the multifunctional treatment of AD.
Collapse
Affiliation(s)
- Anastasiia Ilina
- Department of Biogerontology, Saint Petersburg Institute of Bioregulation and Gerontology, 19711 Saint Petersburg, Russia; (V.K.); (N.L.)
- Department of General Pathology and Pathological Physiology, Institute of Experimental Medicine, 197376 Saint Petersburg, Russia
- Correspondence: ; Tel.: +7-(953)145-89-58
| | - Vladimir Khavinson
- Department of Biogerontology, Saint Petersburg Institute of Bioregulation and Gerontology, 19711 Saint Petersburg, Russia; (V.K.); (N.L.)
- Group of Peptide Regulation of Aging, Pavlov Institute of Physiology, Russian Academy of Sciences, 199034 Saint Petersburg, Russia
| | - Natalia Linkova
- Department of Biogerontology, Saint Petersburg Institute of Bioregulation and Gerontology, 19711 Saint Petersburg, Russia; (V.K.); (N.L.)
| | - Mikhael Petukhov
- Department of Molecular Radiation Biophysics, Petersburg Nuclear Physics Institute Named after B.P. Konstantinov, NRC “Kurchatov Institute”, 188300 Gatchina, Russia;
- Group of Biophysics, Higher Engineering and Technical School, Peter the Great St. Petersburg Polytechnic University, 195251 Saint Petersburg, Russia
| |
Collapse
|
2
|
Nigalye AK, Hess K, Pundlik SJ, Jeffrey BG, Cukras CA, Husain D. Dark Adaptation and Its Role in Age-Related Macular Degeneration. J Clin Med 2022; 11:jcm11051358. [PMID: 35268448 PMCID: PMC8911214 DOI: 10.3390/jcm11051358] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Revised: 02/18/2022] [Accepted: 02/26/2022] [Indexed: 01/04/2023] Open
Abstract
Dark adaptation (DA) refers to the slow recovery of visual sensitivity in darkness following exposure to intense or prolonged illumination, which bleaches a significant amount of the rhodopsin. This natural process also offers an opportunity to understand cellular function in the outer retina and evaluate for presence of disease. How our eyes adapt to darkness can be a key indicator of retinal health, which can be altered in the presence of certain diseases, such as age-related macular degeneration (AMD). A specific focus on clinical aspects of DA measurement and its significance to furthering our understanding of AMD has revealed essential findings underlying the pathobiology of the disease. The process of dark adaptation involves phototransduction taking place mainly between the photoreceptor outer segments and the retinal pigment epithelial (RPE) layer. DA occurs over a large range of luminance and is modulated by both cone and rod photoreceptors. In the photopic ranges, rods are saturated and cone cells adapt to the high luminance levels. However, under scotopic ranges, cones are unable to respond to the dim luminance and rods modulate the responses to lower levels of light as they can respond to even a single photon. Since the cone visual cycle is also based on the Muller cells, measuring the impairment in rod-based dark adaptation is thought to be particularly relevant to diseases such as AMD, which involves both photoreceptors and RPE. Dark adaptation parameters are metrics derived from curve-fitting dark adaptation sensitivities over time and can represent specific cellular function. Parameters such as the cone-rod break (CRB) and rod intercept time (RIT) are particularly sensitive to changes in the outer retina. There is some structural and functional continuum between normal aging and the AMD pathology. Many studies have shown an increase of the rod intercept time (RIT), i.e., delays in rod-mediated DA in AMD patients with increasing disease severity determined by increased drusen grade, pigment changes and the presence of subretinal drusenoid deposits (SDD) and association with certain morphological features in the peripheral retina. Specifications of spatial testing location, repeatability of the testing, ease and availability of the testing device in clinical settings, and test duration in elderly population are also important. We provide a detailed overview in light of all these factors.
Collapse
Affiliation(s)
- Archana K. Nigalye
- Retina Service, Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, 243 Charles St., Boston, MA 02114, USA;
| | - Kristina Hess
- National Eye Institute, National Institutes of Health, Bethesda, MD 20892, USA; (K.H.); (B.G.J.)
| | - Shrinivas J. Pundlik
- Schepens Eye Research Institute of Mass Eye and Ear, Harvard Medical School Department of Ophthalmology, Boston, MA 02114, USA;
| | - Brett G. Jeffrey
- National Eye Institute, National Institutes of Health, Bethesda, MD 20892, USA; (K.H.); (B.G.J.)
| | - Catherine A. Cukras
- National Eye Institute, National Institutes of Health, Bethesda, MD 20892, USA; (K.H.); (B.G.J.)
- Correspondence: (C.A.C.); (D.H.); Tel.: +1-(301)435-5061 (C.A.C.); +1-617-573-4371 (D.H.); Fax: +1-617-573-3698 (D.H.)
| | - Deeba Husain
- Retina Service, Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, 243 Charles St., Boston, MA 02114, USA;
- Correspondence: (C.A.C.); (D.H.); Tel.: +1-(301)435-5061 (C.A.C.); +1-617-573-4371 (D.H.); Fax: +1-617-573-3698 (D.H.)
| |
Collapse
|
3
|
Lee MJ, Bhattarai D, Jang H, Baek A, Yeo IJ, Lee S, Miller Z, Lee S, Hong JT, Kim DE, Lee W, Kim KB. Macrocyclic Immunoproteasome Inhibitors as a Potential Therapy for Alzheimer's Disease. J Med Chem 2021; 64:10934-10950. [PMID: 34309393 PMCID: PMC10913540 DOI: 10.1021/acs.jmedchem.1c00291] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Previously, we reported that immunoproteasome (iP)-targeting linear peptide epoxyketones improve cognitive function in mouse models of Alzheimer's disease (AD) in a manner independent of amyloid β. However, these compounds' clinical prospect for AD is limited due to potential issues, such as poor brain penetration and metabolic instability. Here, we report the development of iP-selective macrocyclic peptide epoxyketones prepared by a ring-closing metathesis reaction between two terminal alkenes attached at the P2 and P3/P4 positions of linear counterparts. We show that a lead macrocyclic compound DB-60 (20) effectively inhibits the catalytic activity of iP in ABCB1-overexpressing cells (IC50: 105 nM) and has metabolic stability superior to its linear counterpart. DB-60 (20) also lowered the serum levels of IL-1α and ameliorated cognitive deficits in Tg2576 mice. The results collectively suggest that macrocyclic peptide epoxyketones have improved CNS drug properties than their linear counterparts and offer promising potential as an AD drug candidate.
Collapse
Affiliation(s)
- Min Jae Lee
- Department of Pharmaceutical Sciences, University of Kentucky, 789 South Limestone, Lexington, KY 40536-0596, USA
| | - Deepak Bhattarai
- Department of Pharmaceutical Sciences, University of Kentucky, 789 South Limestone, Lexington, KY 40536-0596, USA
| | - Hyeryung Jang
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul 08826, Republic of Korea
| | - Ahreum Baek
- Department of Bioscience and Biotechnology, Konkuk University, Seoul 05029, Republic of Korea
| | - In Jun Yeo
- College of Pharmacy, Chungbuk National University, Cheongju, Chungbuk 28160, Republic of Korea
| | - Seongsoo Lee
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul 08826, Republic of Korea
| | - Zachary Miller
- Department of Pharmaceutical Sciences, University of Kentucky, 789 South Limestone, Lexington, KY 40536-0596, USA
| | - Sukyeong Lee
- Verna and Marrs McLean Department of Biochemistry and Molecular Biology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Jin Tae Hong
- College of Pharmacy, Chungbuk National University, Cheongju, Chungbuk 28160, Republic of Korea
| | - Dong-Eun Kim
- Department of Bioscience and Biotechnology, Konkuk University, Seoul 05029, Republic of Korea
| | - Wooin Lee
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul 08826, Republic of Korea
| | - Kyung Bo Kim
- Department of Pharmaceutical Sciences, University of Kentucky, 789 South Limestone, Lexington, KY 40536-0596, USA
| |
Collapse
|
4
|
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
|
5
|
Wu J, Gao G, Shi F, Xie H, Yang Q, Liu D, Qu S, Qin H, Zhang C, Xu GT, Liu F, Zhang J. Activated microglia-induced neuroinflammatory cytokines lead to photoreceptor apoptosis in Aβ-injected mice. J Mol Med (Berl) 2021; 99:713-728. [PMID: 33575853 DOI: 10.1007/s00109-021-02046-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Revised: 01/24/2021] [Accepted: 01/26/2021] [Indexed: 12/16/2022]
Abstract
Age-related macular degeneration (AMD) is mainly characterized by the progressive accumulation of drusen deposits and loss of photoreceptors and retinal pigment epithelial (RPE) cells. Because amyloid β (Aβ) is the main component of drusen, Aβ-induced activated microglia most likely lead to neuroinflammation and play a critical role in the pathogenesis of AMD. However, the relationship between activated microglia-mediated neuroinflammatory cytokines and photoreceptor death has not been clarified. By subretinal injection of Aβ42 in mice, we mimicked an inflammatory milieu of AMD to better understand how activated microglia-induced neuroinflammatory cytokines lead to photoreceptor apoptosis in the AMD progression. We demonstrated that subretinal injection of Aβ42 induces microglial activation and increases inflammatory cytokine release, which gives rise to photoreceptor apoptosis in mice. Our results were verified in vitro by co-culture of Aβ42 activated primary microglia and the photoreceptor cell line 661W. We also demonstrated that the p38 mitogen-activated protein kinase (MAPK) signaling pathway was involved in Aβ42-induced microglial activation and inflammatory cytokine release. Overall, our findings indicate that activated microglia-derived neuroinflammatory cytokines could contribute to photoreceptor apoptosis under the stimulation of Aβ42. Moreover, this study may provide a potential therapeutic approach for AMD. KEY MESSAGES: Further explore the association between activated microglia-derived neuroinflammatory cytokine secretion and photoreceptor apoptosis under the stimulation of Aβ42. Subretinal injection of Aβ42 induces the activation of microglia and increases proinflammatory cytokines IL-1β and COX-2 expression in the retina, which could give rise to the deterioration of visual function and aggravate photoreceptor apoptosis in mice. Primary microglial are activated and the levels of proinflammatory cytokines are increased by Aβ42 stimulation, which could increase the apoptosis of photoreceptor cell line 661W in vitro. The p38 MAPK signaling pathway is involved in microglial activation and photoreceptor apoptosis under Aβ42 treatment.
Collapse
Affiliation(s)
- Jing Wu
- Department of Ophthalmology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Ge Gao
- Center for Translational Neurodegeneration and Regenerative Therapy, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Fanjun Shi
- Department of Ophthalmology, the Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Hai Xie
- Department of Regenerative Medicine and Department of Pharmacology, Tongji University School of Medicine, Shanghai, China
| | - Qian Yang
- Department of Regenerative Medicine and Department of Pharmacology, Tongji University School of Medicine, Shanghai, China
| | - Dandan Liu
- Department of Regenerative Medicine and Department of Pharmacology, Tongji University School of Medicine, Shanghai, China
| | - Sichang Qu
- Department of Ophthalmology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Haifeng Qin
- Department of Ophthalmology, Shanghai Changhai Hospital, Shanghai, China
| | - Chaoyang Zhang
- Department of Ophthalmology, Shanghai General Hospital (Shanghai First People's Hospital), Shanghai Jiao Tong University, Shanghai, 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, China
| | - Guo-Tong Xu
- Department of Regenerative Medicine and Department of Pharmacology, Tongji University School of Medicine, Shanghai, China
| | - Fang Liu
- Department of Ophthalmology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China.
| | - Jingfa Zhang
- Department of Ophthalmology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China. .,Department of Regenerative Medicine and Department of Pharmacology, Tongji University School of Medicine, Shanghai, China. .,Department of Ophthalmology, Shanghai General Hospital (Shanghai First People's Hospital), Shanghai Jiao Tong University, Shanghai, 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, China.
| |
Collapse
|
6
|
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
|
7
|
Innate Immunity in Age-Related Macular Degeneration. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1256:121-141. [PMID: 33848000 DOI: 10.1007/978-3-030-66014-7_5] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Multiple lines of investigation have demonstrated that inflammation plays significant roles in etiology of age-related macular degeneration (AMD). Although interventional trials in AMD therapy targeting inflammatory pathways have been conducted, they have not yet been successful and a detailed understanding as to why some have failed is still elusive. One limitation is the relative dearth of information on how immune cells interact with retinal cells to generate AMD phenotypes at each disease stage. Here, we summarize current research evidence and hypotheses regarding potential pathogenic roles of innate immune cells in the eye, which include resident retinal microglia, macrophages derived from infiltrating systemic monocytes, and macrophages resident in the choroid. We relate recent findings regarding the physiology, function, and cellular interactions involving innate immune cells in the retina and choroid to AMD-related processes, including: (1) drusen formation and regression, (2) the onset and spread of degeneration in late atrophic AMD, and (3) the initiation, growth, and exudation of neovascular vessels in late "wet" AMD. Understanding how innate immune cells contribute to specific AMD phenotypes can assist in generating a comprehensive view on the inflammatory etiology of AMD and aid in identifying anti-inflammatory therapeutic strategies and selecting appropriate clinical outcomes for the planned interventions.
Collapse
|
8
|
Bhattarai D, Lee MJ, Baek A, Yeo IJ, Miller Z, Baek YM, Lee S, Kim DE, Hong JT, Kim KB. LMP2 Inhibitors as a Potential Treatment for Alzheimer’s Disease. J Med Chem 2020; 63:3763-3783. [DOI: 10.1021/acs.jmedchem.0c00416] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Affiliation(s)
- Deepak Bhattarai
- Department of Pharmaceutical Sciences, University of Kentucky, 789 South Limestone, Lexington, Kentucky 40536-0596, United States
| | - Min Jae Lee
- Department of Pharmaceutical Sciences, University of Kentucky, 789 South Limestone, Lexington, Kentucky 40536-0596, United States
| | - Ahruem Baek
- Department of Bioscience and Biotechnology, Konkuk University, Seoul 05029, Republic of Korea
| | - In Jun Yeo
- College of Pharmacy, Chungbuk National University, Cheongju, Chungbuk 28160, Republic of Korea
| | - Zachary Miller
- Department of Pharmaceutical Sciences, University of Kentucky, 789 South Limestone, Lexington, Kentucky 40536-0596, United States
| | - Yu Mi Baek
- Department of Bioscience and Biotechnology, Konkuk University, Seoul 05029, Republic of Korea
| | - Sukyeong Lee
- Verna and Marrs McLean Department of Biochemistry and Molecular Biology, Baylor College of Medicine, Houston, Texas 77030, United States
| | - Dong-Eun Kim
- Department of Bioscience and Biotechnology, Konkuk University, Seoul 05029, Republic of Korea
| | - Jin Tae Hong
- College of Pharmacy, Chungbuk National University, Cheongju, Chungbuk 28160, Republic of Korea
| | - Kyung Bo Kim
- Department of Pharmaceutical Sciences, University of Kentucky, 789 South Limestone, Lexington, Kentucky 40536-0596, United States
| |
Collapse
|
9
|
A dual inhibitor of the proteasome catalytic subunits LMP2 and Y attenuates disease progression in mouse models of Alzheimer's disease. Sci Rep 2019; 9:18393. [PMID: 31804556 PMCID: PMC6895163 DOI: 10.1038/s41598-019-54846-z] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2019] [Accepted: 11/15/2019] [Indexed: 12/18/2022] Open
Abstract
The immunoproteasome (iP) is a variant of the constitutive proteasome (cP) that is abundantly expressed in immune cells which can also be induced in somatic cells by cytokines such as TNF-α or IFN-γ. Accumulating evidence support that the iP is closely linked to multiple facets of inflammatory response, eventually leading to the development of several iP inhibitors as potential therapeutic agents for autoimmune diseases. Recent studies also found that the iP is upregulated in reactive glial cells surrounding amyloid β (Aβ) deposits in brains of Alzheimer’s disease (AD) patients, but the role it plays in the pathogenesis of AD remains unclear. In this study, we investigated the effects of several proteasome inhibitors on cognitive function in AD mouse models and found that YU102, a dual inhibitor of the iP catalytic subunit LMP2 and the cP catalytic subunit Y, ameliorates cognitive impairments in AD mouse models without affecting Aβ deposition. The data obtained from our investigation revealed that YU102 suppresses the secretion of inflammatory cytokines from microglial cells. Overall, this study indicates that there may exist a potential link between LMP2/Y and microglia-mediated neuroinflammation and that inhibition of these subunits may offer a new therapeutic strategy for AD.
Collapse
|
10
|
Nettleton JA, Salem N. International Society for the Study of Fatty Acids and Lipids 2018 Symposium: Arachidonic and Docosahexaenoic Acids in Infant Development. ANNALS OF NUTRITION AND METABOLISM 2019; 74:83-91. [PMID: 30616237 DOI: 10.1159/000495906] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/09/2018] [Accepted: 11/30/2018] [Indexed: 12/16/2022]
Affiliation(s)
| | - Norman Salem
- DSM Nutritional Products, Inc, Columbia, Maryland, USA
| |
Collapse
|