1
|
Li M, Qi X, Tao L. Potential role of βB1 crystallin in cataract formation:a systematic review. Arch Biochem Biophys 2025; 770:110463. [PMID: 40355021 DOI: 10.1016/j.abb.2025.110463] [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: 12/15/2024] [Revised: 03/22/2025] [Accepted: 05/08/2025] [Indexed: 05/14/2025]
Abstract
βB1 crystallin is a soluble structural protein of the lens, which plays an important role in maintaining lens transparency and cell homeostasis. βB1 crystallin has conservative dual structural domains, each of which contains two Greek key motifs. Gene mutation or post-translational modification can affect the structure and function of βB1 crystallin, leading to abnormal protein aggregation and the occurrence of cataracts. This article will review the protein structure, post-translational modification, and related gene mutations of βB1 crystallin. Understanding these molecular mechanisms of βB1crystallin mutations not only aids in clarifying the pathogenesis of cataracts but also provides potential targets for pharmacological interventions.
Collapse
Affiliation(s)
- Muzi Li
- The Second School of Clinical Medicine, Anhui Medical University, 15 Feicui Road, Hefei, Anhui, China
| | - Xiaoxuan Qi
- Department of Ophthalmology, The Second Affiliated Hospital of Anhui Medical University, 678 Furong Road, Hefei, Anhui, China
| | - Liming Tao
- Department of Ophthalmology, The Second Affiliated Hospital of Anhui Medical University, 678 Furong Road, Hefei, Anhui, China.
| |
Collapse
|
2
|
Schaffer N, Mitias S, Guo Y, Bernstein SL, Lindberg I. The neuronal chaperone proSAAS is highly expressed in the retina. PLoS One 2025; 20:e0321867. [PMID: 40378115 DOI: 10.1371/journal.pone.0321867] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2024] [Accepted: 03/12/2025] [Indexed: 05/18/2025] Open
Abstract
The many layers of the neuroretina contain a complex, interconnected network of specialized neurons that both process visual stimuli and conduct processed information to higher brain areas. Neural networks rely on proteostatic control mechanisms to maintain proper protein homeostasis both in cell bodies as well as within synapses; protein chaperones play an important role in regulating and supporting this process. ProSAAS is a small neuronal chaperone that functions as an anti-aggregant in in vitro assays and is released upon depolarization in neuronal primary cultures. We here report a potential role for proSAAS in the retina. A review of human and mouse retinal RNAseq studies reveals that proSAAS expression is abundant within the retina. Single cell sequencing data from mouse and human studies show that proSAAS levels are highest in retinal ganglion cells (RGCs) and horizontal cells. Using proSAAS antibodies in combination with antisera to known retinal cell markers in mouse retinal sections, we confirm RNAseq data showing that proSAAS expression is highest in RGCs and horizontal cells. The proSAAS signal is concentrated within the ganglion cell layer and the inner plexiform layer, a dense synaptic layer connecting retinal neurons. Western blotting of mouse retinal extracts indicates the presence of two processed proSAAS forms, a 21 kDa C-terminally processed form, and a small 13 kDa species which, based on antibody specificity, likely represents an internal fragment. This fragment is also found in extracts prepared from human retinas. Taken together, our data provide support for the hypothesis that retinal synapses utilize the proSAAS chaperone to support visual signaling.
Collapse
Affiliation(s)
- Nicholas Schaffer
- Department of Neurobiology, University of Maryland School of Medicine, Baltimore, Maryland, United States of America
| | - Samira Mitias
- Department of Neurobiology, University of Maryland School of Medicine, Baltimore, Maryland, United States of America
| | - Yan Guo
- Department of Ophthalmology, University of Maryland School of Medicine, Baltimore, Maryland, United States of America
| | - Steven L Bernstein
- Department of Ophthalmology, University of Maryland School of Medicine, Baltimore, Maryland, United States of America
| | - Iris Lindberg
- Department of Neurobiology, University of Maryland School of Medicine, Baltimore, Maryland, United States of America
| |
Collapse
|
3
|
Skowronska-Krawczyk D, Finnemann SC, Grant MB, Held K, Hu Z, Lu YR, Malek G, Sennlaub F, Sparrow J, D'Amore PA. Features that distinguish age-related macular degeneration from aging. Exp Eye Res 2025; 254:110303. [PMID: 39986366 PMCID: PMC11975485 DOI: 10.1016/j.exer.2025.110303] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2025] [Revised: 02/14/2025] [Accepted: 02/18/2025] [Indexed: 02/24/2025]
Abstract
Age-related macular degeneration (AMD) is a complex, multifactorial retinal degenerative disease that is influenced by both genetic and environmental factors. However, the strongest risk factor for AMD is advanced age. Several physiological processes are observed in aging tissues including a low level of chronic inflammation (inflammaging), changed lipid and energy metabolism, and senescence. Nevertheless, whereas everyone ages, only a subset of the population develops AMD. The purpose of this review is to delineate the differences on a cellular and molecular level between natural aging changes and those observed in AMD. We provide a unique perspective on how genetic and environmental components modulate aging in the eye, as well as the specific role of the aging RPE and retina in the pathogenesis of AMD. Topics discussed include the mechanism of aging and its relation to the mechanism of AMD, current animal models that can be used to recapitulate some aspects of the pathology, and potential interventions that shift the balance towards healthy aging and therefore attenuate, prevent or delay the initiation of the disease.
Collapse
Affiliation(s)
| | | | - Maria B Grant
- Department of Ophthalmology and Visual Sciences, Marnix E. Heersink School of Medicine University of Alabama at Birmingham, Alabama, USA
| | - Katherine Held
- Ophthalmology Discovery Research, AbbVie Inc., Irvine, CA, USA
| | - Zhengping Hu
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Boston, MA, USA; Departments of Ophthalmology and Pathology, Harvard Medical School, Boston, MA, USA
| | | | - Goldis Malek
- Duke University, Departments of Ophthalmology, Pathology, and Cell Biology, Albert Eye Research Institute, Durham, NC, USA
| | - Florian Sennlaub
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, 17 rue Moreau, F-75012, Paris, France
| | - Janet Sparrow
- Departments of Ophthalmology and Pathology and Cell Biology, Columbia University Medical Center, New York, NY, USA
| | - Patricia A D'Amore
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Boston, MA, USA; Departments of Ophthalmology and Pathology, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
4
|
Campello L, Brooks MJ, Fadl BR, Choi HS, Pal S, Swaroop A. Transcriptional Heterogeneity and Differential Response of Rod Photoreceptor Pathway Uncovered by Single-Cell RNA Sequencing of the Aging Mouse Retina. Aging Cell 2025; 24:e70001. [PMID: 39954235 PMCID: PMC12073905 DOI: 10.1111/acel.70001] [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: 08/18/2024] [Revised: 11/27/2024] [Accepted: 01/21/2025] [Indexed: 02/17/2025] Open
Abstract
Visual function deteriorates throughout the natural course of aging. Age-related structural and functional adaptations are observed in the retina, the light-sensitive neuronal tissue of the eye where visual perception begins. Molecular mechanisms underlying retinal aging are still poorly understood, highlighting the need to identify biomarkers for better prognosis and alleviation of aging-associated vision impairment. Here, we investigate dynamics of transcriptional dysregulation in the retina and identify affected pathways within distinct retinal cell types. Using an optimized protocol for single-cell RNA sequencing of mouse retinas at 3, 12, 18, and 24 months, we detect a progressive increase in the number of differentially expressed genes across all retinal cell types. The extent and direction of expression changes varies, with photoreceptor, bipolar, and Müller cells showing the maximum number of differentially expressed genes at all age groups. Furthermore, our analyses uncover transcriptionally distinct, heterogeneous subpopulations of rod photoreceptors and bipolar cells, distributed across distinct areas of the retina. Our findings provide a plausible molecular explanation for enhanced susceptibility of rod cells to aging and correlate with the observed loss of scotopic sensitivity in elderly individuals.
Collapse
Affiliation(s)
- Laura Campello
- Neurobiology, Neurodegeneration and Repair Laboratory, National Eye InstituteNational Institutes of HealthBethesdaMarylandUSA
| | - Matthew J. Brooks
- Neurobiology, Neurodegeneration and Repair Laboratory, National Eye InstituteNational Institutes of HealthBethesdaMarylandUSA
| | - Benjamin R. Fadl
- Neurobiology, Neurodegeneration and Repair Laboratory, National Eye InstituteNational Institutes of HealthBethesdaMarylandUSA
| | - Hyo Sub Choi
- Neurobiology, Neurodegeneration and Repair Laboratory, National Eye InstituteNational Institutes of HealthBethesdaMarylandUSA
| | - Soumitra Pal
- Neurobiology, Neurodegeneration and Repair Laboratory, National Eye InstituteNational Institutes of HealthBethesdaMarylandUSA
| | - Anand Swaroop
- Neurobiology, Neurodegeneration and Repair Laboratory, National Eye InstituteNational Institutes of HealthBethesdaMarylandUSA
| |
Collapse
|
5
|
Meng Y, Zhang J, Liu Y, Zhu Y, Lv H, Xia F, Guo Q, Shi Q, Qiu C, Wang J. The biomedical application of inorganic metal nanoparticles in aging and aging-associated diseases. J Adv Res 2025; 71:551-570. [PMID: 38821357 DOI: 10.1016/j.jare.2024.05.023] [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: 10/30/2023] [Revised: 05/10/2024] [Accepted: 05/22/2024] [Indexed: 06/02/2024] Open
Abstract
Aging and aging-associated diseases (AAD), including neurodegenerative disease, cancer, cardiovascular diseases, and diabetes, are inevitable process. With the gradual improvement of life style, life expectancy is gradually extended. However, the extended lifespan has not reduced the incidence of disease, and most elderly people are in ill-health state in their later years. Hence, understanding aging and AAD are significant for reducing the burden of the elderly. Inorganic metal nanoparticles (IMNPs) predominantly include gold, silver, iron, zinc, titanium, thallium, platinum, cerium, copper NPs, which has been widely used to prevent and treat aging and AAD due to their superior properties (essential metal ions for human body, easily synthesis and modification, magnetism). Therefore, a systematic review of common morphological alternations of senescent cells, altered genes and signal pathways in aging and AAD, and biomedical applications of IMNPs in aging and AAD is crucial for the further research and development of IMNPs in aging and AAD. This review focus on the existing research on cellular senescence, aging and AAD, as well as the applications of IMNPs in aging and AAD in the past decade. This review aims to provide cutting-edge knowledge involved with aging and AAD, the application of IMNPs in aging and AAD to promote the biomedical application of IMNPs in aging and AAD.
Collapse
Affiliation(s)
- Yuqing Meng
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao-di Herbs, Artemisinin Research Center, and Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Junzhe Zhang
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao-di Herbs, Artemisinin Research Center, and Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Yanqing Liu
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao-di Herbs, Artemisinin Research Center, and Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Yongping Zhu
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao-di Herbs, Artemisinin Research Center, and Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Haining Lv
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao-di Herbs, Artemisinin Research Center, and Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Fei Xia
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao-di Herbs, Artemisinin Research Center, and Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Qiuyan Guo
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao-di Herbs, Artemisinin Research Center, and Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Qianli Shi
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao-di Herbs, Artemisinin Research Center, and Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Chong Qiu
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao-di Herbs, Artemisinin Research Center, and Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China.
| | - Jigang Wang
- Department of Urology, Shenzhen Clinical Research Centre for Geriatrics, Shenzhen People's Hospital; The First Affiliated Hospital, Southern University of Science and Technology, Shenzhen 518020, Guangdong, China; State Key Laboratory of Antiviral Drugs, School of Pharmacy, Henan University, Kaifeng 475004, China.
| |
Collapse
|
6
|
Song HB, Campello L, Mondal A, Chen HY, English MA, Glen M, Vanlandingham P, Farjo R, Swaroop A. Sex-specific attenuation of photoreceptor degeneration by reserpine in a rhodopsin P23H rat model of autosomal dominant retinitis pigmentosa. eLife 2025; 14:RP103888. [PMID: 40231721 PMCID: PMC11999695 DOI: 10.7554/elife.103888] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/16/2025] Open
Abstract
Inherited retinal degenerations (IRDs) constitute a group of clinically and genetically diverse vision-impairing disorders. Retinitis pigmentosa (RP), the most common form of IRD, is characterized by gradual dysfunction and degeneration of rod photoreceptors, followed by the loss of cone photoreceptors. Recently, we identified reserpine as a lead molecule for maintaining rod survival in mouse and human retinal organoids as well as in the rd16 mouse, which phenocopy Leber congenital amaurosis caused by mutations in the cilia-centrosomal gene CEP290 (Chen et al., 2023). Here, we show the therapeutic potential of reserpine in a rhodopsin P23H rat model of autosomal dominant RP. At postnatal day (P) 68, when males and females are analyzed together, the reserpine-treated rats exhibit higher rod-derived scotopic b-wave amplitudes compared to the controls with little or no change in scotopic a-wave or cone-derived photopic b-wave. Interestingly, the reserpine-treated female rats display enhanced scotopic a- and b-waves and photopic b-wave responses at P68, along with a better contrast threshold and increased outer nuclear layer thickness. The female rats demonstrate better preservation of both rod and cone photoreceptors following reserpine treatment. Retinal transcriptome analysis reveals sex-specific responses to reserpine, with significant upregulation of phototransduction genes and proteostasis-related pathways, and notably, genes associated with stress response. This study builds upon our previously reported results reaffirming the potential of reserpine for gene-agnostic treatment of IRDs and emphasizes the importance of biological sex in retinal disease research and therapy development.
Collapse
Affiliation(s)
- Hyun Beom Song
- Neurobiology, Neurodegeneration and Repair Laboratory, National Eye Institute, National Institutes of HealthBethesdaUnited States
- Department of Biomedical Sciences, Seoul National University College of MedicineSeoulRepublic of Korea
| | - Laura Campello
- Neurobiology, Neurodegeneration and Repair Laboratory, National Eye Institute, National Institutes of HealthBethesdaUnited States
| | - Anupam Mondal
- Neurobiology, Neurodegeneration and Repair Laboratory, National Eye Institute, National Institutes of HealthBethesdaUnited States
| | - Holly Y Chen
- Neurobiology, Neurodegeneration and Repair Laboratory, National Eye Institute, National Institutes of HealthBethesdaUnited States
| | - Milton A English
- Neurobiology, Neurodegeneration and Repair Laboratory, National Eye Institute, National Institutes of HealthBethesdaUnited States
| | - Michael Glen
- Neurobiology, Neurodegeneration and Repair Laboratory, National Eye Institute, National Institutes of HealthBethesdaUnited States
| | | | | | - Anand Swaroop
- Neurobiology, Neurodegeneration and Repair Laboratory, National Eye Institute, National Institutes of HealthBethesdaUnited States
| |
Collapse
|
7
|
Sun J, Liu Y, Chen Z. Melatonin and retinal cell damage: molecular and biological functions. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025; 398:3199-3212. [PMID: 39520554 DOI: 10.1007/s00210-024-03575-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Accepted: 10/27/2024] [Indexed: 11/16/2024]
Abstract
The indoleamine hormone, melatonin, is produced in the pineal gland and has an essential role in many physiological functions. The pineal gland is considered to be the most important organ for producing melatonin. Nevertheless, it is important to point out that the eye is also capable of producing melatonin, and has its own circadian rhythm in producing this hormone. Melatonin is mainly produced by a subpopulation of photoreceptors in a diurnal rhythm. Numerous in vitro and in vivo studies have shown the beneficial effects of melatonin in eye-related disorders. These diseases primarily affect retinal cells, highlighting the therapeutic potential of melatonin, especially in the retina. Melatonin's ability to regulate oxidative stress response pathways and modulate the expression of antioxidant genes makes it a promising candidate for mitigating retinal cell damage. Moreover, melatonin can modulate inflammatory pathways such as NF-кB and further reduce retinal damage, as well as affecting programmed cell death such as apoptosis and autophagy in retinal cells. Therefore, the goal of this review is to explore the ways in which melatonin protects retinal cells from damage and ischemia. We discuss the mechanisms involved in order to gain valuable understanding of the possible therapeutic applications of melatonin in protection of retinal cells and treatment of retinal disorders.
Collapse
Affiliation(s)
- Jingwen Sun
- Harbin 242 Hospital, Harbin, Heilongjiang, 150000, China
| | - Yan Liu
- Harbin 242 Hospital, Harbin, Heilongjiang, 150000, China
| | - Zhangming Chen
- Harbin 242 Hospital, Harbin, Heilongjiang, 150000, China.
| |
Collapse
|
8
|
Durán-Cristiano SC, de Diego-García L, Martín-Gil A, Carracedo G. The Role of the Ubiquitin System in Eye Diseases. Life (Basel) 2025; 15:504. [PMID: 40141848 PMCID: PMC11943997 DOI: 10.3390/life15030504] [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: 01/30/2025] [Revised: 03/11/2025] [Accepted: 03/19/2025] [Indexed: 03/28/2025] Open
Abstract
The ubiquitin-proteasome system (UPS) is a fundamental process that regulates various biological functions, including immune response, cell cycle, oxidative stress, migration, and cellular proliferation. This system is responsible for the degradation of proteins, while proteasomes play a significant role in mechanisms involved in health and human diseases. The participation of the UPS in immune response is particularly relevant, leading to the involvement of immunoproteasomes. This specialized proteasome is involved in the processing and presentation of antigenic peptides, making it crucial for proper immune function. Moreover, the impact of the UPS is considered essential in understanding several diseases, such as neurodegenerative disorders, infections, and vascular diseases. The dysregulation of the UPS may contribute to the pathogenesis of these conditions, highlighting its importance as a potential therapeutic target. Interestingly, the UPS is also related to ocular structures, playing a role in visual perception and ocular homeostasis. This involvement in the regulation of various ocular processes suggests its potential impact on both anterior and posterior eye pathologies. This review aims to discuss the general considerations of the UPS and provide information about its participation in anterior and posterior eye pathologies. By understanding its role in ocular health and disease, researchers and clinicians may explore novel therapeutic strategies targeting the UPS for the treatment of various eye conditions. In conclusion, the UPS is a crucial player in biological processes, with far-reaching implications in health and disease, including the anterior and posterior segments of the eye. Further research in this field may lead to the development of innovative therapies and a better understanding of the complex mechanisms underlying various eye disorders.
Collapse
Affiliation(s)
| | - Laura de Diego-García
- Department of Biochemistry and Molecular Biology, Faculty of Optics and Optometry, Universidad Complutense de Madrid, 28037 Madrid, Spain
| | - Alba Martín-Gil
- Department of Optometry and Vision, Faculty of Optics and Optometry, Universidad Complutense de Madrid, 28037 Madrid, Spain; (A.M.-G.); (G.C.)
| | - Gonzalo Carracedo
- Department of Optometry and Vision, Faculty of Optics and Optometry, Universidad Complutense de Madrid, 28037 Madrid, Spain; (A.M.-G.); (G.C.)
| |
Collapse
|
9
|
Koller A, Preishuber-Pflügl J, Mayr D, Brunner SM, Ladek AM, Runge C, Aigner L, Reitsamer HA, Trost A. Cysteinyl leukotriene receptor 1 modulates retinal immune cells, vascularity and proteolytic activity in aged mice. Aging (Albany NY) 2025; 17:308-328. [PMID: 39891615 PMCID: PMC11892928 DOI: 10.18632/aging.206193] [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: 03/20/2024] [Accepted: 01/21/2025] [Indexed: 02/03/2025]
Abstract
Cysteinyl leukotrienes (CysLTs) modulate the immune response, the microvasculature, cell stress and the endosomal-lysosomal system, and are involved in cellular aging. Interestingly, CysLT receptor 1 (Cysltr1) is highly expressed in the retina, a tissue that is strongly affected by the aging process. Thus, we performed an introductory examination to determine a potential importance of Cysltr1 for cells in the neurovascular unit using qPCR and immunofluorescence analysis, and on proteolytic activity in the retinas of aged mice. Aged mice (~84 weeks) were treated orally with vehicle or 10 mg/kg montelukast (MTK), a specific Cysltr1 inhibitor, for 8 weeks, 5x/week. The retinas of young mice (~11 weeks) served as controls. Compared with young control mice, aged mice exhibited increased numbers of microglia and a reduced retinal capillary diameter, but these age-dependent changes were abrogated by MTK treatment. Retinal protein levels of the ubiquitin binding protein sequestosome-1 were amplified by aging, but were reduced by MTK treatment. Interestingly, retinal proteasome activity was decreased in aged mice, whereas Cysltr1 inhibition increased this activity. The reduction in immune cells caused by Cysltr1 suppression may dampen neuroinflammation, a known promoter of tissue aging. Additionally, an increase in capillary diameter after Cysltr1 inhibition could have a beneficial effect on blood flow in aged individuals. Furthermore, the increase in proteolytic activity upon Cysltr1 inhibition could prevent the accumulation of toxic deposits, which is a hallmark of aged tissue. Overall, Cysltr1 is a promising target for modulating the impact of aging on retinal tissue.
Collapse
Affiliation(s)
- Andreas Koller
- Department of Ophthalmology and Optometry, Research Program for Experimental Ophthalmology and Glaucoma Research, University Hospital of the Paracelsus Medical University, Salzburg 5020, Austria
| | - Julia Preishuber-Pflügl
- Department of Ophthalmology and Optometry, Research Program for Experimental Ophthalmology and Glaucoma Research, University Hospital of the Paracelsus Medical University, Salzburg 5020, Austria
| | - Daniela Mayr
- Department of Ophthalmology and Optometry, Research Program for Experimental Ophthalmology and Glaucoma Research, University Hospital of the Paracelsus Medical University, Salzburg 5020, Austria
| | - Susanne Maria Brunner
- Department of Ophthalmology and Optometry, Research Program for Experimental Ophthalmology and Glaucoma Research, University Hospital of the Paracelsus Medical University, Salzburg 5020, Austria
| | - Anja-Maria Ladek
- Department of Ophthalmology and Optometry, Research Program for Experimental Ophthalmology and Glaucoma Research, University Hospital of the Paracelsus Medical University, Salzburg 5020, Austria
| | - Christian Runge
- Department of Ophthalmology and Optometry, Research Program for Experimental Ophthalmology and Glaucoma Research, University Hospital of the Paracelsus Medical University, Salzburg 5020, Austria
| | - Ludwig Aigner
- Institute of Molecular Regenerative Medicine, Paracelsus Medical University, Salzburg 5020, Austria
| | - Herbert Anton Reitsamer
- Department of Ophthalmology and Optometry, Research Program for Experimental Ophthalmology and Glaucoma Research, University Hospital of the Paracelsus Medical University, Salzburg 5020, Austria
| | - Andrea Trost
- Department of Ophthalmology and Optometry, Research Program for Experimental Ophthalmology and Glaucoma Research, University Hospital of the Paracelsus Medical University, Salzburg 5020, Austria
| |
Collapse
|
10
|
Dey PN, Singh N, Zelinger L, Batz Z, Nellissery J, White Carreiro ND, Qian H, Li T, Fariss RN, Dong L, Swaroop A. Loss of paired immunoglobin-like type 2 receptor B gene associated with age-related macular degeneration impairs photoreceptor function in mouse retina. Hum Mol Genet 2025; 34:64-76. [PMID: 39532089 PMCID: PMC12034095 DOI: 10.1093/hmg/ddae161] [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: 08/10/2024] [Revised: 10/10/2024] [Accepted: 11/01/2024] [Indexed: 11/16/2024] Open
Abstract
Genome-wide association studies have uncovered mostly non-coding variants at over 60 genetic loci linked to susceptibility for age-related macular degeneration (AMD). To ascertain the causal gene at the PILRB/PILRA locus, we used a CRISPR strategy to produce germline deletions in the mouse paired immunoglobin-like type 2 receptor (Pilr) genes that encode highly related activating (PILRB) and inhibitory (PILRA) receptors. We show that a combined loss of Pilrb1 and Pilrb2, but not Pilra, leads to an early but relatively stationary defect as the electroretinography (ERG) amplitudes of Pilrb1/2-/- mice exhibit a marked reduction as early as postnatal day 15 and do not show additional significant decrease at 3 and 12-months. No alterations are evident in Müller glia, microglia, bipolar, amacrine and horizontal cells based on immunohistochemistry using cell-type specific markers. PILRB immunostaining is specifically detected at the proximal part of photoreceptor outer segment. Reduced expression of select calcium-regulated phototransduction and synapse-associated proteins, including GCAP1 and 2, PDE6b, AIPL1, PSD95, and CTBP1 indicates dysregulation of calcium homeostasis as a possible mechanism of retinal phenotype in Pilrb1/2-/- mice. Our studies suggest a novel function of PILRB in retinal photoreceptors and an association of PILRB, but not PILRA, with AMD pathogenesis.
Collapse
Affiliation(s)
- Partha Narayan Dey
- Neurobiology, Neurodegeneration & Repair Laboratory, National Eye Institute, National Institutes of Health, 6 Center Drive, MSC0610, Bethesda, MD 20892, United States
| | - Nivedita Singh
- Neurobiology, Neurodegeneration & Repair Laboratory, National Eye Institute, National Institutes of Health, 6 Center Drive, MSC0610, Bethesda, MD 20892, United States
| | - Lina Zelinger
- Neurobiology, Neurodegeneration & Repair Laboratory, National Eye Institute, National Institutes of Health, 6 Center Drive, MSC0610, Bethesda, MD 20892, United States
| | - Zachary Batz
- Neurobiology, Neurodegeneration & Repair Laboratory, National Eye Institute, National Institutes of Health, 6 Center Drive, MSC0610, Bethesda, MD 20892, United States
| | - Jacob Nellissery
- Neurobiology, Neurodegeneration & Repair Laboratory, National Eye Institute, National Institutes of Health, 6 Center Drive, MSC0610, Bethesda, MD 20892, United States
| | - Noor D White Carreiro
- Biological Imaging Core, National Eye Institute, National Institutes of Health, 6 Center Drive, MSC0610, Bethesda, MD 20892, United States
| | - Haohua Qian
- Visual Function Core Facility, National Eye Institute, National Institutes of Health, 6 Center Drive, MSC0610, Bethesda, MD 20892, United States
| | - Tiansen Li
- Neurobiology, Neurodegeneration & Repair Laboratory, National Eye Institute, National Institutes of Health, 6 Center Drive, MSC0610, Bethesda, MD 20892, United States
| | - Robert N Fariss
- Biological Imaging Core, National Eye Institute, National Institutes of Health, 6 Center Drive, MSC0610, Bethesda, MD 20892, United States
| | - Lijin Dong
- Genetic Engineering Core, National Eye Institute, National Institutes of Health, 6 Center Drive, MSC0610, Bethesda, MD 20892, United States
| | - Anand Swaroop
- Neurobiology, Neurodegeneration & Repair Laboratory, National Eye Institute, National Institutes of Health, 6 Center Drive, MSC0610, Bethesda, MD 20892, United States
| |
Collapse
|
11
|
Qian H, Chen W, Yuan G, Luo M, Zhang L, Wu B, Huang H, Xu J, Wang Q, Li M. RTA408 alleviates retinal ganglion cells damage in mouse glaucoma by inhibiting excessive autophagy. PLoS One 2024; 19:e0313446. [PMID: 39527591 PMCID: PMC11554135 DOI: 10.1371/journal.pone.0313446] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2024] [Accepted: 10/23/2024] [Indexed: 11/16/2024] Open
Abstract
BACKGROUND Glaucoma, characterized by a high incidence and significant ocular harm, has been elucidated through various mechanisms. Excessive autophagy leading to the loss of retinal ganglion cells (RGCs) is suggested as one potential cause for visual impairment in glaucoma. METHODS A glaucoma model was established through anterior chamber injection of silicone oil in mice. RTA408 and the positive control tafluprost were administered for intervention. The efficacy was preliminarily assessed by intraocular pressure measurement. HE staining and fluorescent staining were used to assess RGC loss, while fluorescent staining and western blot were employed to evaluate the expression of Nrf2. The role of autophagy in the pathogenesis of glaucoma was investigated by artificially modulating autophagy levels. RESULTS In glaucomatous mice, RTA408 significantly reduces the apoptosis levels of RGCs and decreases RGC loss. Further investigations reveal a notable upregulation of autophagy levels in glaucomatous mice, with RGC loss being associated with autophagy. RTA408 promotes the expression of Nrf2 and downstream antioxidant molecules, enhancing the antioxidant system while downregulating mitochondrial autophagy levels. This reduces RGC apoptosis and loss, demonstrating a protective effect against glaucoma. CONCLUSION Autophagy mediates the occurrence of glaucoma in mice, promoting RGC apoptosis. RTA408 alleviates RGCs damage by inhibiting excessive autophagy in the context of glaucoma.
Collapse
Affiliation(s)
- Hongmei Qian
- Department of Ophthalmology, Shaoxing People’s Hospital, Shaoxing, Zhejiang, China
| | - Wei Chen
- Department of Ophthalmology, Shaoxing People’s Hospital, Shaoxing, Zhejiang, China
| | - Guomei Yuan
- Department of Ophthalmology, Shaoxing People’s Hospital, Shaoxing, Zhejiang, China
| | - Man Luo
- Department of Ophthalmology, Shaoxing People’s Hospital, Shaoxing, Zhejiang, China
| | - Li Zhang
- Department of Ophthalmology, Shaoxing People’s Hospital, Shaoxing, Zhejiang, China
| | - Biao Wu
- Department of Ophthalmology, Shaoxing People’s Hospital, Shaoxing, Zhejiang, China
| | - Hanshi Huang
- Department of Ophthalmology, Shaoxing People’s Hospital, Shaoxing, Zhejiang, China
| | - Jiahao Xu
- Department of Ophthalmology, Shaoxing People’s Hospital, Shaoxing, Zhejiang, China
| | - Qiong Wang
- Department of Ophthalmology, Shaoxing People’s Hospital, Shaoxing, Zhejiang, China
| | - Mengyun Li
- Medical Research Center, Shaoxing People’s Hospital, Shaoxing, Zhejiang, China
| |
Collapse
|
12
|
Tundo GR, Cavaterra D, Pandino I, Zingale GA, Giammaria S, Boccaccini A, Michelessi M, Roberti G, Tanga L, Carnevale C, Figus M, Grasso G, Coletta M, Bocedi A, Oddone F, Sbardella D. The Delayed Turnover of Proteasome Processing of Myocilin upon Dexamethasone Stimulation Introduces the Profiling of Trabecular Meshwork Cells' Ubiquitylome. Int J Mol Sci 2024; 25:10017. [PMID: 39337505 PMCID: PMC11432723 DOI: 10.3390/ijms251810017] [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: 08/01/2024] [Revised: 09/11/2024] [Accepted: 09/13/2024] [Indexed: 09/30/2024] Open
Abstract
Glaucoma is chronic optic neuropathy whose pathogenesis has been associated with the altered metabolism of Trabecular Meshwork Cells, which is a cell type involved in the synthesis and remodeling of the trabecular meshwork, the main drainage pathway of the aqueous humor. Starting from previous findings supporting altered ubiquitin signaling, in this study, we investigated the ubiquitin-mediated turnover of myocilin (MYOC/TIGR gene), which is a glycoprotein with a recognized role in glaucoma pathogenesis, in a human Trabecular Meshwork strain cultivated in vitro in the presence of dexamethasone. This is a validated experimental model of steroid-induced glaucoma, and myocilin upregulation by glucocorticoids is a phenotypic marker of Trabecular Meshwork strains. Western blotting and native-gel electrophoresis first uncovered that, in the presence of dexamethasone, myocilin turnover by proteasome particles was slower than in the absence of the drug. Thereafter, co-immunoprecipitation, RT-PCR and gene-silencing studies identified STUB1/CHIP as a candidate E3-ligase of myocilin. In this regard, dexamethasone treatment was found to downregulate STUB1/CHIP levels by likely promoting its proteasome-mediated turnover. Hence, to strengthen the working hypothesis about global alterations of ubiquitin-signaling, the first profiling of TMCs ubiquitylome, in the presence and absence of dexamethasone, was here undertaken by diGLY proteomics. Application of this workflow effectively highlighted a robust dysregulation of key pathways (e.g., phospholipid signaling, β-catenin, cell cycle regulation) in dexamethasone-treated Trabecular Meshwork Cells, providing an ubiquitin-centered perspective around the effect of glucocorticoids on metabolism and glaucoma pathogenesis.
Collapse
Affiliation(s)
- Grazia Raffaella Tundo
- Department of Clinical Sciences and Translational Medicine, University of Tor Vergata, 00133 Rome, Italy
| | - Dario Cavaterra
- Department of Chemical Sciences and Technologies, University of Tor Vergata, 00133 Rome, Italy (A.B.)
| | - Irene Pandino
- IRCCS-Fondazione Bietti, 00168 Rome, Italy (G.R.); (M.C.)
| | | | - Sara Giammaria
- IRCCS-Fondazione Bietti, 00168 Rome, Italy (G.R.); (M.C.)
| | | | | | - Gloria Roberti
- IRCCS-Fondazione Bietti, 00168 Rome, Italy (G.R.); (M.C.)
| | - Lucia Tanga
- IRCCS-Fondazione Bietti, 00168 Rome, Italy (G.R.); (M.C.)
| | | | - Michele Figus
- Department of Surgical, Medical, Molecular Pathology and Critical Care Medicine, University of Pisa, 56124 Pisa, Italy;
| | - Giuseppe Grasso
- Department of Chemical Sciences, University of Catania, 95125 Catania, Italy;
| | | | - Alessio Bocedi
- Department of Chemical Sciences and Technologies, University of Tor Vergata, 00133 Rome, Italy (A.B.)
| | | | | |
Collapse
|
13
|
Ma Z, Hao J, Yang Z, Zhang M, Xin J, Bi H, Guo D. Research Progress on the Role of Ubiquitination in Eye Diseases. Cell Biochem Biophys 2024; 82:1825-1836. [PMID: 38913283 DOI: 10.1007/s12013-024-01381-y] [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] [Accepted: 06/19/2024] [Indexed: 06/25/2024]
Abstract
The occurrence and development of ophthalmic diseases are related to the dysfunction of eye tissues. Ubiquitin is an important form of protein post-translational modification, which plays an essential role in the occurrence and development of diseases through specific modification of target proteins. Ubiquitination governs a variety of intracellular signal transduction processes, including proteasome degradation, DNA damage repair, and cell cycle progression. Studies have found that ubiquitin can play a role in eye diseases such as cataracts, glaucoma, keratopathy, retinopathy, and eye tumors. In this paper, the role of protein ubiquitination in eye diseases was reviewed.
Collapse
Affiliation(s)
- Zhongyu Ma
- Shandong University of Traditional Chinese Medicine, Jinan, 250002, China
| | - Jiawen Hao
- Shandong University of Traditional Chinese Medicine, Jinan, 250002, China
| | - Zhaohui Yang
- Shandong University of Traditional Chinese Medicine, Jinan, 250002, China
| | - Miao Zhang
- Shandong University of Traditional Chinese Medicine, Jinan, 250002, China
| | - Jizhao Xin
- Shandong University of Traditional Chinese Medicine, Jinan, 250002, China
| | - Hongsheng Bi
- Affiliated Eye Hospital of Shandong University of Traditional Chinese Medicine, Jinan, 250002, China.
- Shandong Provincial Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Therapy of Ocular Diseases, Shandong Academy of Eye Disease Prevention and Therapy, Shandong Provincial Clinical Research Center of Ophthalmology and Children Visual Impairment Prevention and Control, Shandong Engineering Technology Research Center of Visual Intelligence, Shandong Academy of Health and Myopia Prevention and Control of Children and Adolescents, Jinan, 250002, China.
- Medical College of Optometry and Ophthalmology, Shandong University of Traditional Chinese Medicine, Jinan, 250002, China.
| | - Dadong Guo
- Affiliated Eye Hospital of Shandong University of Traditional Chinese Medicine, Jinan, 250002, China.
- Shandong Provincial Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Therapy of Ocular Diseases, Shandong Academy of Eye Disease Prevention and Therapy, Shandong Provincial Clinical Research Center of Ophthalmology and Children Visual Impairment Prevention and Control, Shandong Engineering Technology Research Center of Visual Intelligence, Shandong Academy of Health and Myopia Prevention and Control of Children and Adolescents, Jinan, 250002, China.
- Medical College of Optometry and Ophthalmology, Shandong University of Traditional Chinese Medicine, Jinan, 250002, China.
| |
Collapse
|
14
|
Cvekl A, Vijg J. Aging of the eye: Lessons from cataracts and age-related macular degeneration. Ageing Res Rev 2024; 99:102407. [PMID: 38977082 DOI: 10.1016/j.arr.2024.102407] [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: 03/11/2024] [Revised: 06/18/2024] [Accepted: 07/01/2024] [Indexed: 07/10/2024]
Abstract
Aging is the greatest risk factor for chronic human diseases, including many eye diseases. Geroscience aims to understand the effects of the aging process on these diseases, including the genetic, molecular, and cellular mechanisms that underlie the increased risk of disease over the lifetime. Understanding of the aging eye increases general knowledge of the cellular physiology impacted by aging processes at various biological extremes. Two major diseases, age-related cataract and age-related macular degeneration (AMD) are caused by dysfunction of the lens and retina, respectively. Lens transparency and light refraction are mediated by lens fiber cells lacking nuclei and other organelles, which provides a unique opportunity to study a single aging hallmark, i.e., loss of proteostasis, within an environment of limited metabolism. In AMD, local dysfunction of the photoreceptors/retinal pigmented epithelium/Bruch's membrane/choriocapillaris complex in the macula leads to the loss of photoreceptors and eventually loss of central vision, and is driven by nearly all the hallmarks of aging and shares features with Alzheimer's disease, Parkinson's disease, cardiovascular disease, and diabetes. The aging eye can function as a model for studying basic mechanisms of aging and, vice versa, well-defined hallmarks of aging can be used as tools to understand age-related eye disease.
Collapse
Affiliation(s)
- Ales Cvekl
- Departments of Genetics and Ophthalmology and Visual Sciences, Albert Einstein College of Medicine, Bronx, NY 10461, USA.
| | - Jan Vijg
- Departments of Genetics and Ophthalmology and Visual Sciences, Albert Einstein College of Medicine, Bronx, NY 10461, USA.
| |
Collapse
|
15
|
Bejarano E, Domenech-Bendaña A, Avila-Portillo N, Rowan S, Edirisinghe S, Taylor A. Glycative stress as a cause of macular degeneration. Prog Retin Eye Res 2024; 101:101260. [PMID: 38521386 PMCID: PMC11699537 DOI: 10.1016/j.preteyeres.2024.101260] [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: 01/05/2024] [Revised: 03/08/2024] [Accepted: 03/11/2024] [Indexed: 03/25/2024]
Abstract
People are living longer and rates of age-related diseases such as age-related macular degeneration (AMD) are accelerating, placing enormous burdens on patients and health care systems. The quality of carbohydrate foods consumed by an individual impacts health. The glycemic index (GI) is a kinetic measure of the rate at which glucose arrives in the blood stream after consuming various carbohydrates. Consuming diets that favor slowly digested carbohydrates releases sugar into the bloodstream gradually after consuming a meal (low glycemic index). This is associated with reduced risk for major age-related diseases including AMD, cardiovascular disease, and diabetes. In comparison, consuming the same amounts of different carbohydrates in higher GI diets, releases glucose into the blood rapidly, causing glycative stress as well as accumulation of advanced glycation end products (AGEs). Such AGEs are cytotoxic by virtue of their forming abnormal proteins and protein aggregates, as well as inhibiting proteolytic and other protective pathways that might otherwise selectively recognize and remove toxic species. Using in vitro and animal models of glycative stress, we observed that consuming higher GI diets perturbs metabolism and the microbiome, resulting in a shift to more lipid-rich metabolomic profiles. Interactions between aging, diet, eye phenotypes and physiology were observed. A large body of laboratory animal and human clinical epidemiologic data indicates that consuming lower GI diets, or lower glycemia diets, is protective against features of early AMD (AMDf) in mice and AMD prevalence or AMD progression in humans. Drugs may be optimized to diminish the ravages of higher glycemic diets. Human trials are indicated to determine if AMD progression can be retarded using lower GI diets. Here we summarized the current knowledge regarding the pathological role of glycative stress in retinal dysfunction and how dietary strategies might diminish retinal disease.
Collapse
Affiliation(s)
- Eloy Bejarano
- Department of Biomedical Sciences, School of Health Sciences and Veterinary School, Universidad Cardenal Herrera-CEU, CEU Universities, Valencia, Spain
| | - Alicia Domenech-Bendaña
- Department of Biomedical Sciences, School of Health Sciences and Veterinary School, Universidad Cardenal Herrera-CEU, CEU Universities, Valencia, Spain
| | | | - Sheldon Rowan
- JM USDA Human Nutrition Research Center on Aging at Tufts University, United States
| | - Sachini Edirisinghe
- Tufts University Friedman School of Nutrition Science and Policy, United States
| | - Allen Taylor
- Tufts University Friedman School of Nutrition Science and Policy, United States.
| |
Collapse
|
16
|
Bisen AC, Dubey A, Agrawal S, Biswas A, Rawat KS, Srivastava S, Bhatta RS. Recent updates on ocular disease management with ophthalmic ointments. Ther Deliv 2024; 15:463-480. [PMID: 38888757 DOI: 10.1080/20415990.2024.2346047] [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: 10/31/2023] [Accepted: 04/18/2024] [Indexed: 06/20/2024] Open
Abstract
Ophthalmic diseases can result in permanent vision loss and blindness. Convenient topical and systemic treatments are preferred to address these sight-threatening conditions. However, the unique anatomy of the eye presents challenges for drug delivery. Various ophthalmic ointment formulations have been developed to enhance bioavailability in the eye to prolong residence time and improve corneal permeability. This article explores a wide range of ocular diseases affecting individuals globally and how ointments are used to manage them. From eye to ocular barriers, this review focuses on published scientific research and formulation strategies for severe ocular complications using conventional topical ointments. Additionally, it delves through patented technologies and marketed formulations supporting the use of ointments in ocular drug delivery.
Collapse
Affiliation(s)
- Amol Chhatrapati Bisen
- Pharmaceutics & Pharmacokinetics Division, CSIR-Central Drug Research Institute, Lucknow, 226031, Uttar Pradesh, India
- Academy of Scientific & Innovative Research (AcSIR), Ghaziabad, 201002, Uttar Pradesh, India
- Sophisticated Analytical Instrument Facility and Research, CSIR-Central Drug Research Institute, Lucknow, 226031, Uttar Pradesh, India
| | - Ayush Dubey
- Pharmaceutics & Pharmacokinetics Division, CSIR-Central Drug Research Institute, Lucknow, 226031, Uttar Pradesh, India
- School of Pharmaceutical Sciences, CSJM University, Kanpur, 208024, Uttar Pradesh, India
| | - Sristi Agrawal
- Pharmaceutics & Pharmacokinetics Division, CSIR-Central Drug Research Institute, Lucknow, 226031, Uttar Pradesh, India
- Academy of Scientific & Innovative Research (AcSIR), Ghaziabad, 201002, Uttar Pradesh, India
| | - Arpon Biswas
- Pharmaceutics & Pharmacokinetics Division, CSIR-Central Drug Research Institute, Lucknow, 226031, Uttar Pradesh, India
| | - Kundan Singh Rawat
- Prof. Rajendra Singh Nanoscience & Chemistry D.S.B. Campus, Kumaun University, Nainital, 263001, Uttarakhand, India
| | - Saurabh Srivastava
- Pharmaceutics & Pharmacokinetics Division, CSIR-Central Drug Research Institute, Lucknow, 226031, Uttar Pradesh, India
- School of Pharmaceutical Sciences, CSJM University, Kanpur, 208024, Uttar Pradesh, India
| | - Rabi Sankar Bhatta
- Pharmaceutics & Pharmacokinetics Division, CSIR-Central Drug Research Institute, Lucknow, 226031, Uttar Pradesh, India
- Academy of Scientific & Innovative Research (AcSIR), Ghaziabad, 201002, Uttar Pradesh, India
| |
Collapse
|
17
|
Chen PY, Shen M, Cai SQ, Tang ZW. Association Between Atopic Dermatitis and Aging: Clinical Observations and Underlying Mechanisms. J Inflamm Res 2024; 17:3433-3448. [PMID: 38828054 PMCID: PMC11144009 DOI: 10.2147/jir.s467099] [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: 03/29/2024] [Accepted: 05/17/2024] [Indexed: 06/05/2024] Open
Abstract
As one of the most prevalent chronic inflammatory skin diseases, atopic dermatitis (AD) increasingly affects the aging population. Amid the ongoing global aging trend, it's essential to recognize the intricate relationship between AD and aging. This paper reviews existing knowledge, summarizing clinical observations of associations between AD and aging-related diseases in various systems, including endocrine, cardiovascular, and neurological. Additionally, it discusses major theories explaining the correlation, encompassing skin-mucosal barriers, systemic inflammation and stress, genes, signal transduction, and environmental and behavioral factors. The association between AD and aging holds significant importance, both in population and basic perspectives. While further research is warranted, this paper aims to inspire deeper exploration of inflammation/allergy-aging dynamics and the timely management of elderly patients with AD.
Collapse
Affiliation(s)
- Peng-Yu Chen
- Department of Dermatology, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, People’s Republic of China
| | - Minxue Shen
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, 410008, People’s Republic of China
- Hunan Engineering Research Center of Skin Health and Disease; Hunan Key Laboratory of Skin Cancer and Psoriasis (Xiangya Hospital), Changsha, 410008, People’s Republic of China
- Department of Social Medicine and Health Management, Xiangya School of Public Health, Central South University, Changsha, 410078, People’s Republic of China
| | - Sui-Qing Cai
- Department of Dermatology, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, People’s Republic of China
| | - Zhen-Wei Tang
- Department of Dermatology, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, People’s Republic of China
| |
Collapse
|
18
|
Nithianandam V, Sarkar S, Feany MB. Pathways controlling neurotoxicity and proteostasis in mitochondrial complex I deficiency. Hum Mol Genet 2024; 33:860-871. [PMID: 38324746 PMCID: PMC11070137 DOI: 10.1093/hmg/ddae018] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Revised: 01/02/2024] [Accepted: 01/24/2024] [Indexed: 02/09/2024] Open
Abstract
Neuromuscular disorders caused by dysfunction of the mitochondrial respiratory chain are common, severe and untreatable. We recovered a number of mitochondrial genes, including electron transport chain components, in a large forward genetic screen for mutations causing age-related neurodegeneration in the context of proteostasis dysfunction. We created a model of complex I deficiency in the Drosophila retina to probe the role of protein degradation abnormalities in mitochondrial encephalomyopathies. Using our genetic model, we found that complex I deficiency regulates both the ubiquitin/proteasome and autophagy/lysosome arms of the proteostasis machinery. We further performed an in vivo kinome screen to uncover new and potentially druggable mechanisms contributing to complex I related neurodegeneration and proteostasis failure. Reduction of RIOK kinases and the innate immune signaling kinase pelle prevented neurodegeneration in complex I deficiency animals. Genetically targeting oxidative stress, but not RIOK1 or pelle knockdown, normalized proteostasis markers. Our findings outline distinct pathways controlling neurodegeneration and protein degradation in complex I deficiency and introduce an experimentally facile model in which to study these debilitating and currently treatment-refractory disorders.
Collapse
Affiliation(s)
- Vanitha Nithianandam
- Department of Pathology, Brigham and Women’s Hospital, Harvard Medical School, 77 Avenue Louis Pasteur, Boston, MA 02115, United States
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, 5425 Wisconsin Avenue, Chevy Chase, MD 20815, United States
| | - Souvarish Sarkar
- Department of Pathology, Brigham and Women’s Hospital, Harvard Medical School, 77 Avenue Louis Pasteur, Boston, MA 02115, United States
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, 5425 Wisconsin Avenue, Chevy Chase, MD 20815, United States
| | - Mel B Feany
- Department of Pathology, Brigham and Women’s Hospital, Harvard Medical School, 77 Avenue Louis Pasteur, Boston, MA 02115, United States
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, 5425 Wisconsin Avenue, Chevy Chase, MD 20815, United States
| |
Collapse
|
19
|
Brener A, Lorber D, Reuveny A, Toledano H, Porat-Kuperstein L, Lebenthal Y, Weizman E, Olender T, Volk T. Sedentary Behavior Impacts on the Epigenome and Transcriptome: Lessons from Muscle Inactivation in Drosophila Larvae. Cells 2023; 12:2333. [PMID: 37830547 PMCID: PMC10571804 DOI: 10.3390/cells12192333] [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: 08/31/2023] [Revised: 09/18/2023] [Accepted: 09/19/2023] [Indexed: 10/14/2023] Open
Abstract
The biological mechanisms linking sedentary lifestyles and metabolic derangements are incompletely understood. In this study, temporal muscle inactivation in Drosophila larvae carrying a temperature-sensitive mutation in the shibire (shi1) gene was induced to mimic sedentary behavior during early life and study its transcriptional outcome. Our findings indicated a significant change in the epigenetic profile, as well as the genomic profile, of RNA Pol II binding in the inactive muscles relative to control, within a relatively short time period. Whole-genome analysis of RNA-Pol II binding to DNA by muscle-specific targeted DamID (TaDa) protocol revealed that muscle inactivity altered Pol II binding in 121 out of 2010 genes (6%), with a three-fold enrichment of genes coding for lncRNAs. The suppressed protein-coding genes included genes associated with longevity, DNA repair, muscle function, and ubiquitin-dependent proteostasis. Moreover, inducing muscle inactivation exerted a multi-level impact upon chromatin modifications, triggering an altered epigenetic balance of active versus inactive marks. The downregulated genes in the inactive muscles included genes essential for muscle structure and function, carbohydrate metabolism, longevity, and others. Given the multiple analogous genes in Drosophila for many human genes, extrapolating our findings to humans may hold promise for establishing a molecular link between sedentary behavior and metabolic diseases.
Collapse
Affiliation(s)
- Avivit Brener
- Pediatric Endocrinology and Diabetes Institute, Dana-Dwek Children’s Hospital, Tel Aviv Sourasky Medical Center, Affiliated with the Faculty of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel; (A.B.); (Y.L.)
| | - Dana Lorber
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot 7610001, Israel; (D.L.); (A.R.); (T.O.)
| | - Adriana Reuveny
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot 7610001, Israel; (D.L.); (A.R.); (T.O.)
| | - Hila Toledano
- Department of Human Biology, Faculty of Natural Sciences, University of Haifa, Haifa 3498838, Israel; (H.T.); (L.P.-K.)
| | - Lilach Porat-Kuperstein
- Department of Human Biology, Faculty of Natural Sciences, University of Haifa, Haifa 3498838, Israel; (H.T.); (L.P.-K.)
| | - Yael Lebenthal
- Pediatric Endocrinology and Diabetes Institute, Dana-Dwek Children’s Hospital, Tel Aviv Sourasky Medical Center, Affiliated with the Faculty of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel; (A.B.); (Y.L.)
| | - Eviatar Weizman
- G-INCPM, Weizmann Institute of Science, Rehovot 7610001, Israel;
| | - Tsviya Olender
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot 7610001, Israel; (D.L.); (A.R.); (T.O.)
| | - Talila Volk
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot 7610001, Israel; (D.L.); (A.R.); (T.O.)
| |
Collapse
|
20
|
Ma X, Nan Y, Huang C, Li X, Yang Y, Jiang W, Ye M, Liu Q, Niu Y, Yuan L. Expression of αA-crystallin (CRYAA) in vivo and in vitro models of age-related cataract and the effect of its silencing on HLEB3 cells. Aging (Albany NY) 2023; 15:204754. [PMID: 37253645 DOI: 10.18632/aging.204754] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2023] [Accepted: 05/16/2023] [Indexed: 06/01/2023]
Abstract
AIM To investigate the expression of αA-crystallin (CRYAA) in age-related cataract (ARC) models and its role in lens epithelial cells (LECs). METHODS We used Flow cytometry to detect the apoptosis and cell cycle in HLEB3 cells and Real-time fluorescence quantitative polymerase chain reaction to detect the expression of CRYAA mRNA in HLEB3 and in rabbit lens. The expression of CRYAA in HLEB3 cells and rabbit lenses as well as the proteins related to apoptosis and autophagy in transfected cells were detected by western blotting. The lens structure in rabbits was investigated using hematoxylin-eosin staining. Protein thermostability assay was performed to detect the thermal stability of rabbit lens proteins. CCK- 8 assay was used to detect the viability of transfected cells, and the transfection was recorded by fluorescence photography. RESULTS Hydrogen peroxide can promote apoptosis and arrest the cell cycle in HLEB3 cells, and naphthalene can cause cataract formation and damage the structure of the lens in rabbits. Both ARC models can reduce the expression of CRYAA. The expression of CRYAA silencing increased apoptosis and autophagy in HLEB3 cells.
Collapse
Affiliation(s)
- Xiaoling Ma
- Ningxia Medical University Key Laboratory of Ningxia Minority Medicine Modernization Ministry of Education, Ningxia Medical University, Yinchuan 750004, Ningxia, China
| | - Yi Nan
- Ningxia Medical University Key Laboratory of Ningxia Minority Medicine Modernization Ministry of Education, Ningxia Medical University, Yinchuan 750004, Ningxia, China
| | - Can Huang
- Ningxia Medical University Key Laboratory of Ningxia Minority Medicine Modernization Ministry of Education, Ningxia Medical University, Yinchuan 750004, Ningxia, China
| | - Xiangyang Li
- School of Traditional Chinese Medicine, Ningxia Medical University, Yinchuan 750004, Ningxia, China
| | - Yifan Yang
- Ningxia Medical University Key Laboratory of Ningxia Minority Medicine Modernization Ministry of Education, Ningxia Medical University, Yinchuan 750004, Ningxia, China
| | - Wenjie Jiang
- Ningxia Medical University Key Laboratory of Ningxia Minority Medicine Modernization Ministry of Education, Ningxia Medical University, Yinchuan 750004, Ningxia, China
| | - Mengyi Ye
- School of Traditional Chinese Medicine, Ningxia Medical University, Yinchuan 750004, Ningxia, China
| | - Qian Liu
- School of Clinical Medicine, Ningxia Medical University, Yinchuan 750004, Ningxia, China
| | - Yang Niu
- Ningxia Medical University Key Laboratory of Ningxia Minority Medicine Modernization Ministry of Education, Ningxia Medical University, Yinchuan 750004, Ningxia, China
- School of Traditional Chinese Medicine, Ningxia Medical University, Yinchuan 750004, Ningxia, China
| | - Ling Yuan
- Ningxia Medical University Key Laboratory of Ningxia Minority Medicine Modernization Ministry of Education, Ningxia Medical University, Yinchuan 750004, Ningxia, China
- School of Pharmacy, Ningxia Medical University, Yinchuan 750004, Ningxia Hui Autonomous Region, China
| |
Collapse
|
21
|
Disatham J, Brennan L, Cvekl A, Kantorow M. Multiomics Analysis Reveals Novel Genetic Determinants for Lens Differentiation, Structure, and Transparency. Biomolecules 2023; 13:693. [PMID: 37189439 PMCID: PMC10136076 DOI: 10.3390/biom13040693] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Revised: 04/13/2023] [Accepted: 04/16/2023] [Indexed: 05/17/2023] Open
Abstract
Recent advances in next-generation sequencing and data analysis have provided new gateways for identification of novel genome-wide genetic determinants governing tissue development and disease. These advances have revolutionized our understanding of cellular differentiation, homeostasis, and specialized function in multiple tissues. Bioinformatic and functional analysis of these genetic determinants and the pathways they regulate have provided a novel basis for the design of functional experiments to answer a wide range of long-sought biological questions. A well-characterized model for the application of these emerging technologies is the development and differentiation of the ocular lens and how individual pathways regulate lens morphogenesis, gene expression, transparency, and refraction. Recent applications of next-generation sequencing analysis on well-characterized chicken and mouse lens differentiation models using a variety of omics techniques including RNA-seq, ATAC-seq, whole-genome bisulfite sequencing (WGBS), chip-seq, and CUT&RUN have revealed a wide range of essential biological pathways and chromatin features governing lens structure and function. Multiomics integration of these data has established new gene functions and cellular processes essential for lens formation, homeostasis, and transparency including the identification of novel transcription control pathways, autophagy remodeling pathways, and signal transduction pathways, among others. This review summarizes recent omics technologies applied to the lens, methods for integrating multiomics data, and how these recent technologies have advanced our understanding ocular biology and function. The approach and analysis are relevant to identifying the features and functional requirements of more complex tissues and disease states.
Collapse
Affiliation(s)
- Joshua Disatham
- Charles E. Schmidt College of Medicine, Florida Atlantic University, Boca Raton, FL 33431, USA; (J.D.); (L.B.)
| | - Lisa Brennan
- Charles E. Schmidt College of Medicine, Florida Atlantic University, Boca Raton, FL 33431, USA; (J.D.); (L.B.)
| | - Ales Cvekl
- Departments of Ophthalmology and Visual Sciences and Genetics, Albert Einstein College of Medicine, Bronx, NY 10461, USA;
| | - Marc Kantorow
- Charles E. Schmidt College of Medicine, Florida Atlantic University, Boca Raton, FL 33431, USA; (J.D.); (L.B.)
| |
Collapse
|