201
|
Beykin G, Stell L, Halim MS, Nuñez M, Popova L, Nguyen BT, Groth SL, Dennis A, Li Z, Atkins M, Khavari T, Wang SY, Chang R, Fisher AC, Sepah YJ, Goldberg JL. Phase 1b Randomized Controlled Study of Short Course Topical Recombinant Human Nerve Growth Factor (rhNGF) for Neuroenhancement in Glaucoma: Safety, Tolerability, and Efficacy Measure Outcomes. Am J Ophthalmol 2022; 234:223-234. [PMID: 34780798 PMCID: PMC8821405 DOI: 10.1016/j.ajo.2021.11.002] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Revised: 11/01/2021] [Accepted: 11/02/2021] [Indexed: 02/03/2023]
Abstract
PURPOSE No approved therapies directly target retinal ganglion cells (RGCs) for neuroprotection or neuroenhancement in glaucoma. Recombinant human nerve growth factor (rhNGF) has been shown to promote RGC survival and function in animal models of optic neuropathy. Here we evaluate the safety, tolerability, and efficacy of short-term, high-dose rhNGF eye drops versus placebo in a cohort of glaucoma patients. DESIGN This was a prospective, phase 1b, single-center, randomized, double-masked, vehicle-controlled, parallel-group study. METHODS This study was designed to assess safety and tolerability as well as short-term neuroenhancement of structure and function (clinicaltrials.gov NCT02855450). A total of 60 open-angle glaucoma patients were randomized 40:20 to receive either 180 μg/mL rhNGF or vehicle control eye drops in both eyes, 3 times daily for 8 weeks, with a 24-week post-treatment follow-up. One eye was officially selected as the study eye, although both eyes were studied and dosed. Primary endpoints were safety, as assessed by adverse events, and tolerability, as assessed by patient-reported outcomes. Secondary outcome measures included best corrected visual acuity (BCVA), Humphrey visual field, electroretinograpy (ERG), and optical coherence tomography (OCT) of retinal nerve fiber layer (RNFL) thickness at baseline, after 8 weeks of treatment, and at 4 and 24 weeks after treatment (12 and 32 weeks total). RESULTS Of the 60 randomized patients, 23 were female (38%) and the average age was 66.1 years. Through week 32, there were no treatment-related serious adverse events, including no unexpectedly severe progression of optic neuropathy, no adverse events affecting ocular function or pressure, and no drug-related systemic toxicity. Topical high-dose rhNGF was tolerated well, with a low level of symptom burden mainly eliciting periocular ache (in 52% of treated group and 5% of placebo group) and only 3 patients (7.5%) discontinuing treatment because of discomfort, of whom 1 patient (2.5%) prematurely withdrew from the study. There were no statistically significant differences in global indices of Humphrey visual field and no meaningful differences in total, quadrant, or clock-hour mean RNFL thickness between the groups, although both of these function and structure measures showed nonsignificant trends toward significance in favor of rhNGF. Real-world participant data was used to generate an estimate of cohort size needed to power subsequent studies. CONCLUSIONS Use of rhNGF is safe and tolerable in a topical 180-μg/mL formulation. Although no statistically significant short-term neuroenhancement was detected in this trial, given the strong effects of NGF in preclinical models and the trends detected in this study, analysis for efficacy in a neuroprotection trial is warranted. NOTE: Publication of this article is sponsored by the American Ophthalmological Society.
Collapse
|
202
|
Treatment of Glaucoma with Natural Products and Their Mechanism of Action: An Update. Nutrients 2022; 14:nu14030534. [PMID: 35276895 PMCID: PMC8840399 DOI: 10.3390/nu14030534] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Revised: 01/15/2022] [Accepted: 01/18/2022] [Indexed: 02/07/2023] Open
Abstract
Glaucoma is one of the leading causes of irreversible blindness. It is generally caused by increased intraocular pressure, which results in damage of the optic nerve and retinal ganglion cells, ultimately leading to visual field dysfunction. However, even with the use of intraocular pressure-lowering eye drops, the disease still progresses in some patients. In addition to mechanical and vascular dysfunctions of the eye, oxidative stress, neuroinflammation and excitotoxicity have also been implicated in the pathogenesis of glaucoma. Hence, the use of natural products with antioxidant and anti-inflammatory properties may represent an alternative approach for glaucoma treatment. The present review highlights recent preclinical and clinical studies on various natural products shown to possess neuroprotective properties for retinal ganglion cells, which thereby may be effective in the treatment of glaucoma. Intraocular pressure can be reduced by baicalein, forskolin, marijuana, ginsenoside, resveratrol and hesperidin. Alternatively, Ginkgo biloba, Lycium barbarum, Diospyros kaki, Tripterygium wilfordii, saffron, curcumin, caffeine, anthocyanin, coenzyme Q10 and vitamins B3 and D have shown neuroprotective effects on retinal ganglion cells via various mechanisms, especially antioxidant, anti-inflammatory and anti-apoptosis mechanisms. Extensive studies are still required in the future to ensure natural products' efficacy and safety to serve as an alternative therapy for glaucoma.
Collapse
|
203
|
Leung CKS, Ren ST, Chan PPM, Wan KHN, Kam AKW, Lai GWK, Chiu VSM, Ko MWL, Yiu CKF, Yu MCY. Nicotinamide riboside as a neuroprotective therapy for glaucoma: study protocol for a randomized, double-blind, placebo-control trial. Trials 2022; 23:45. [PMID: 35039056 PMCID: PMC8762963 DOI: 10.1186/s13063-021-05968-1] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Accepted: 12/23/2021] [Indexed: 12/18/2022] Open
Abstract
Background Whereas lowering the intraocular pressure (IOP) can slow optic nerve degeneration in glaucoma, many patients with glaucoma continue to develop progressive loss in vision despite a significant reduction in IOP. No treatment has been shown to be effective for neuroprotection in glaucoma. We set out to conduct a randomized controlled trial to investigate whether nicotinamide riboside (NR), a nicotinamide adenine dinucleotide precursor, is effective to slow optic nerve degeneration in patients with primary open-angle glaucoma (POAG). We hypothesize that patients treated with NR have a slower rate of progressive retinal nerve fiber layer (RNFL) thinning compared with those treated with placebo. Methods This is a randomized, double-blind, placebo-controlled, parallel-group, multi-center study including 125 patients with POAG. Patients will be randomized to receive 300 mg NR or placebo for 24 months. Clinical examination, optical coherence tomography imaging of the RNFL, and visual field (VF) test will be performed at the baseline, 1 month, 4 months, and then at 2-month intervals until 24 months. The primary outcome measure is the rate of RNFL thinning measured over 24 months. The secondary outcome measures include (1) time to VF progression, (2) time to progressive RNFL/ganglion cell inner plexiform layer (GCIPL) thinning, and (3) the rate of change of VF sensitivity over 24 months (to investigate neuroprotection) and 1 month (to investigate neuroenhancement). The rates of RNFL thinning and VF sensitivity decline between treatment groups will be compared with linear mixed modeling. Survival analysis will be performed to compare the differences in time from baseline to VF progression and time from baseline to progressive RNFL/GCIPL thinning between treatment groups using Cox proportional hazards models. Discussion Outcome measures in glaucoma neuroprotection trials have been centered on the detection of VF progression, which may take years to develop and confirm. In addition to addressing whether NR has a neuroprotective/neuroenhancement effect in glaucoma patients, this study will demonstrate the feasibility of studying neuroprotection in a relatively short trial period (24 months) by comparing the rates of progressive RNFL thinning, a more reproducible and objective outcome measure compared with VF endpoints, between treatment groups. Trial registration Chinese Clinical Trial Registry 1900021998
Collapse
Affiliation(s)
- Christopher Kai Shun Leung
- Department of Ophthalmology, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, People's Republic of China. .,Hong Kong Eye Hospital, Hong Kong, People's Republic of China. .,Department of Ophthalmology and Visual Sciences, The Chinese University of Hong Kong, Hong Kong, People's Republic of China.
| | - Seraph Tianmin Ren
- Department of Ophthalmology and Visual Sciences, The Chinese University of Hong Kong, Hong Kong, People's Republic of China
| | - Poemen Pui Man Chan
- Hong Kong Eye Hospital, Hong Kong, People's Republic of China.,Department of Ophthalmology and Visual Sciences, The Chinese University of Hong Kong, Hong Kong, People's Republic of China
| | - Kelvin Ho Nam Wan
- Hong Kong Eye Hospital, Hong Kong, People's Republic of China.,Department of Ophthalmology and Visual Sciences, The Chinese University of Hong Kong, Hong Kong, People's Republic of China
| | - Aziz Ka Wai Kam
- Department of Ophthalmology and Visual Sciences, The Chinese University of Hong Kong, Hong Kong, People's Republic of China
| | - Gilda Wing Ki Lai
- Department of Ophthalmology, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, People's Republic of China
| | - Vivian Sheung Man Chiu
- Department of Ophthalmology, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, People's Republic of China
| | - Match Wai Lun Ko
- Department of Mechanical Engineering, University of Hong Kong, Hong Kong, People's Republic of China
| | - Cedric Ka Fai Yiu
- Department of Applied Mathematics, Hong Kong Polytechnic University, Hong Kong, People's Republic of China
| | | |
Collapse
|
204
|
Liu P, Wang F, Song Y, Wang M, Zhang X. Current situation and progress of drugs for reducing intraocular pressure. Ther Adv Chronic Dis 2022; 13:20406223221140392. [PMID: 36479139 PMCID: PMC9720821 DOI: 10.1177/20406223221140392] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Accepted: 11/03/2022] [Indexed: 12/05/2022] Open
Abstract
Glaucoma, the most common cause of irreversible blindness worldwide, usually causes characteristic optic nerve damage. Pathological intraocular pressure (IOP) elevation is a major risk factor. Drug reduction of IOP is the preferred treatment for clinicians because it can delay the progression of disease. However, the traditional IOP-lowering drugs currently used by patients may be poorly tolerated. Therefore, in recent years, some new drugs have been put into clinical application or in clinical phase I–III studies. They have a better IOP-lowering effect and fewer adverse reactions. Because glaucoma is a chronic disease, drugs need to be administered continuously for a long time. For patients, good compliance and high drug bioavailability have a positive effect on the prognosis of the disease. Therefore, clinicians and scientists have developed drug delivery systems to solve this complex problem. In addition, natural compounds and dietary supplements have a good effect of reducing IOP, and they can also protect the optic nerve through antioxidant action. We summarize the current traditional drugs, new drugs, sustained-release drug delivery systems, and complementary drugs and outline the mechanism of action and clinical effects of these drugs on glaucoma and their recent advances.
Collapse
Affiliation(s)
- Peiyu Liu
- Jiangxi Provincial Key Laboratory for Ophthalmology, Nanchang University School of Ophthalmology & Optometry, Jiangxi Research Institute of Ophthalmology & Visual Science, Affiliated Eye Hospital of Nanchang University, Nanchang, China
| | - Feifei Wang
- Jiangxi Provincial Key Laboratory for Ophthalmology, Nanchang University School of Ophthalmology & Optometry, Jiangxi Research Institute of Ophthalmology & Visual Science, Affiliated Eye Hospital of Nanchang University, Nanchang, China
| | - Yuning Song
- Jiangxi Provincial Key Laboratory for Ophthalmology, Nanchang University School of Ophthalmology & Optometry, Jiangxi Research Institute of Ophthalmology & Visual Science, Affiliated Eye Hospital of Nanchang University, Nanchang, China
| | - Menghui Wang
- Jiangxi Provincial Key Laboratory for Ophthalmology, Nanchang University School of Ophthalmology & Optometry, Jiangxi Research Institute of Ophthalmology & Visual Science, Affiliated Eye Hospital of Nanchang University, Nanchang, China
| | - Xu Zhang
- Jiangxi Provincial Key Laboratory for Ophthalmology, Nanchang University School of Ophthalmology & Optometry, Jiangxi Research Institute of Ophthalmology & Visual Science, Affiliated Eye Hospital of Nanchang University, 463 Bayi Road, Nanchang 330006, China
| |
Collapse
|
205
|
Zhou DB, Castanos MV, Geyman L, Rich CA, Tantraworasin A, Ritch R, Rosen RB. Mitochondrial Dysfunction in Primary Open-angle Glaucoma Characterized by Flavoprotein Fluorescence at the Optic Nerve Head. Ophthalmol Glaucoma 2021; 5:413-420. [PMID: 34968754 DOI: 10.1016/j.ogla.2021.12.006] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2021] [Revised: 12/18/2021] [Accepted: 12/22/2021] [Indexed: 11/30/2022]
Abstract
OBJECTIVE To investigate the presence of flavoprotein fluorescence (FPF) at the optic nerve head (ONH) rim as a marker of mitochondrial dysfunction in primary open-angle glaucoma (POAG) and control eyes. DESIGN Retrospective cross-sectional study, with patients recruited from the New York Eye and Ear Infirmary of Mount Sinai. SUBJECTS, PARTICIPANTS, AND/OR CONTROLS A total of 86 eyes (50 eyes of 30 POAG patents and 36 eyes of 20 controls) were enrolled. The presence of POAG was defined by circumpapillary retinal nerve fiber layer thickness below the bottom fifth percentile of the normative database, glaucomatous ONH changes, and visual field defects on 24-2 tests. METHODS, INTERVENTION, OR TESTING POAG and control eyes were imaged using the OcuMet Beacon. A 23°x23° infrared scan was obtained, and an FPF scan was performed within a capture field spanning 13 degrees in diameter. The ONH margins on the infrared image were identified by software algorithms. FPF then was measured within an elliptical annulus around the ONH rim, with the inner and outer boundaries corresponding to 0.5 to 1.1 times the ONH rim size. MAIN OUTCOMES MEASURES FPF at the OHN rim in POAG and control eyes. RESULTS Differences in FPF between POAG and control eyes were characterized through mixed-effects logistic regression, adjusted for age and interocular pressure. FPF was significantly higher in POAG versus control eyes, with a mean±SD of 46.4±27.9 versus 28.0±11.7 (P<0.001), respectively. Evaluation of anatomical quadrants revealed greater FPF in POAG versus control eyes at the temporal (P=0.001), superior (P<0.001), nasal (P=0.002), and inferior (P=0.001) quadrants. Among POAG eyes, FPF showed correlation to visual field mean deviation (P<0.001), visual field pattern standard deviation (P=0.003), and circumpapillary retinal nerve fiber thickness (P=0.001) on linear mixed-effects models. CONCLUSIONS Higher FPF in POAG versus control eyes suggests the presence of mitochondrial dysfunction at the ONH rim in eyes with glaucomatous damage. The degree of FPF corresponds to disease severity, as measured by visual field and nerve fiber layer thickness metrics. FPF may thus represent a metabolic indicator of disease status that reveals the extent of injury in glaucoma.
Collapse
Affiliation(s)
- Davis B Zhou
- Department of Ophthalmology, New York Eye and Ear Infirmary of Mount Sinai; Department of Ophthalmology, Icahn School of Medicine at Mount Sinai
| | - Maria V Castanos
- Department of Ophthalmology, New York Eye and Ear Infirmary of Mount Sinai; Department of Ophthalmology, Icahn School of Medicine at Mount Sinai
| | - Lawrence Geyman
- Einhorn Clinical Research Center, New York Eye and Ear Infirmary of Mount Sinai, New York; Department of Ophthalmology, Illinois Eye and Ear Infirmary, Chicago, Illinois
| | | | - Apichat Tantraworasin
- Clinical Epidemiology and Clinical Statistic Center, and Department of Surgery, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Robert Ritch
- Einhorn Clinical Research Center, New York Eye and Ear Infirmary of Mount Sinai, New York
| | - Richard B Rosen
- Department of Ophthalmology, New York Eye and Ear Infirmary of Mount Sinai; Department of Ophthalmology, Icahn School of Medicine at Mount Sinai.
| |
Collapse
|
206
|
Lo Faro V, Nolte IM, Ten Brink JB, Snieder H, Jansonius NM, Bergen AA. Mitochondrial Genome Study Identifies Association Between Primary Open-Angle Glaucoma and Variants in MT-CYB, MT-ND4 Genes and Haplogroups. Front Genet 2021; 12:781189. [PMID: 34976016 PMCID: PMC8719162 DOI: 10.3389/fgene.2021.781189] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Accepted: 11/29/2021] [Indexed: 11/21/2022] Open
Abstract
Background and purpose: Primary open-angle glaucoma (POAG) is an optic neuropathy characterized by death of retinal ganglion cells and atrophy of the optic nerve head. The susceptibility of the optic nerve to damage has been shown to be mediated by mitochondrial dysfunction. In this study, we aimed to determine a possible association between mitochondrial SNPs or haplogroups and POAG. Methods: Mitochondrial DNA single nucleotide polymorphisms (mtSNPs) were genotyped using the Illumina Infinium Global Screening Array-24 (GSA) 700K array set. Genetic analyses were performed in a POAG case-control study involving the cohorts, Groningen Longitudinal Glaucoma Study-Lifelines Cohort Study and Amsterdam Glaucoma Study, including 721 patients and 1951 controls in total. We excluded samples not passing quality control for nuclear genotypes and samples with low call rate for mitochondrial variation. The mitochondrial variants were analyzed both as SNPs and haplogroups. These were determined with the bioinformatics software HaploGrep, and logistic regression analysis was used for the association, as well as for SNPs. Results: Meta-analysis of the results from both cohorts revealed a significant association between POAG and the allele A of rs2853496 [odds ratio (OR) = 0.64; p = 0.006] within the MT-ND4 gene, and for the T allele of rs35788393 (OR = 0.75; p = 0.041) located in the MT-CYB gene. In the mitochondrial haplogroup analysis, the most significant p-value was reached by haplogroup K (p = 1.2 × 10−05), which increases the risk of POAG with an OR of 5.8 (95% CI 2.7–13.1). Conclusion: We identified an association between POAG and polymorphisms in the mitochondrial genes MT-ND4 (rs2853496) and MT-CYB (rs35788393), and with haplogroup K. The present study provides further evidence that mitochondrial genome variations are implicated in POAG. Further genetic and functional studies are required to substantiate the association between mitochondrial gene polymorphisms and POAG and to define the pathophysiological mechanisms of mitochondrial dysfunction in glaucoma.
Collapse
Affiliation(s)
- Valeria Lo Faro
- Department of Ophthalmology, University of Groningen, University Medical Center Groningen, Groningen, Netherlands
- Department of Clinical Genetics, Amsterdam University Medical Center (AMC), Amsterdam, Netherlands
| | - Ilja M. Nolte
- Department of Epidemiology, University of Groningen, University Medical Center Groningen, Groningen, Netherlands
| | - Jacoline B. Ten Brink
- Department of Clinical Genetics, Amsterdam University Medical Center (AMC), Amsterdam, Netherlands
| | - Harold Snieder
- Department of Epidemiology, University of Groningen, University Medical Center Groningen, Groningen, Netherlands
| | - Nomdo M. Jansonius
- Department of Ophthalmology, University of Groningen, University Medical Center Groningen, Groningen, Netherlands
| | - Arthur A. Bergen
- Department of Clinical Genetics, Amsterdam University Medical Center (AMC), Amsterdam, Netherlands
- Department of Ophthalmology, Amsterdam UMC, Amsterdam, Netherlands
- *Correspondence: Arthur A. Bergen,
| |
Collapse
|
207
|
Manrique-Caballero CL, Kellum JA, Gómez H, De Franco F, Giacchè N, Pellicciari R. Innovations and Emerging Therapies to Combat Renal Cell Damage: NAD + As a Drug Target. Antioxid Redox Signal 2021; 35:1449-1466. [PMID: 33499758 PMCID: PMC8905249 DOI: 10.1089/ars.2020.8066] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Revised: 01/21/2021] [Accepted: 01/21/2021] [Indexed: 12/29/2022]
Abstract
Significance: Acute kidney injury (AKI) is a common and life-threatening complication in hospitalized and critically ill patients. It is defined by an abrupt deterioration in renal function, clinically manifested by increased serum creatinine levels, decreased urine output, or both. To execute all its functions, namely excretion of waste products, fluid/electrolyte balance, and hormone synthesis, the kidney requires incredible amounts of energy in the form of adenosine triphosphate. Recent Advances: Adequate mitochondrial functioning and nicotinamide adenine dinucleotide (NAD+) homeostasis are essential to meet these high energetic demands. NAD+ is a ubiquitous essential coenzyme to many cellular functions. NAD+ as an electron acceptor mediates metabolic pathways such as oxidative phosphorylation (OXPHOS) and glycolysis, serves as a cosubstrate of aging molecules (i.e., sirtuins), participates in DNA repair mechanisms, and mediates mitochondrial biogenesis. Critical Issues: In many forms of AKI and chronic kidney disease, renal function deterioration has been associated with mitochondrial dysfunction and NAD+ depletion. Based on this, therapies aiming to restore mitochondrial function and increase NAD+ availability have gained special attention in the last two decades. Future Directions: Experimental and clinical studies have shown that by restoring mitochondrial homeostasis and increasing renal tubulo-epithelial cells, NAD+ availability, AKI incidence, and chronic long-term complications are significantly decreased. This review covers some general epidemiological and pathophysiological concepts; describes the role of mitochondrial homeostasis and NAD+ metabolism; and analyzes the underlying rationale and role of NAD+ aiming therapies as promising preventive and therapeutic strategies for AKI. Antioxid. Redox Signal. 35, 1449-1466.
Collapse
Affiliation(s)
- Carlos L. Manrique-Caballero
- Center for Critical Care Nephrology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Department of Critical Care Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - John A. Kellum
- Center for Critical Care Nephrology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Department of Critical Care Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Hernando Gómez
- Center for Critical Care Nephrology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Department of Critical Care Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | | | | | | |
Collapse
|
208
|
Sokolov D, Sechrest ER, Wang Y, Nevin C, Du J, Kolandaivelu S. Nuclear NAD +-biosynthetic enzyme NMNAT1 facilitates development and early survival of retinal neurons. eLife 2021; 10:e71185. [PMID: 34878972 PMCID: PMC8754432 DOI: 10.7554/elife.71185] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2021] [Accepted: 12/07/2021] [Indexed: 11/13/2022] Open
Abstract
Despite mounting evidence that the mammalian retina is exceptionally reliant on proper NAD+ homeostasis for health and function, the specific roles of subcellular NAD+ pools in retinal development, maintenance, and disease remain obscure. Here, we show that deletion of the nuclear-localized NAD+ synthase nicotinamide mononucleotide adenylyltransferase-1 (NMNAT1) in the developing murine retina causes early and severe degeneration of photoreceptors and select inner retinal neurons via multiple distinct cell death pathways. This severe phenotype is associated with disruptions to retinal central carbon metabolism, purine nucleotide synthesis, and amino acid pathways. Furthermore, transcriptomic and immunostaining approaches reveal dysregulation of a collection of photoreceptor and synapse-specific genes in NMNAT1 knockout retinas prior to detectable morphological or metabolic alterations. Collectively, our study reveals previously unrecognized complexity in NMNAT1-associated retinal degeneration and suggests a yet-undescribed role for NMNAT1 in gene regulation during photoreceptor terminal differentiation.
Collapse
Affiliation(s)
- David Sokolov
- Department of Ophthalmology and Visual Sciences, Eye Institute, One Medical Center Drive, West Virginia UniversityMorgantownUnited States
| | - Emily R Sechrest
- Department of Ophthalmology and Visual Sciences, Eye Institute, One Medical Center Drive, West Virginia UniversityMorgantownUnited States
| | - Yekai Wang
- Department of Ophthalmology and Visual Sciences, Eye Institute, One Medical Center Drive, West Virginia UniversityMorgantownUnited States
- Department of Biochemistry, One Medical Center Drive, West Virginia UniversityMorgantownUnited States
| | - Connor Nevin
- Department of Ophthalmology and Visual Sciences, Eye Institute, One Medical Center Drive, West Virginia UniversityMorgantownUnited States
| | - Jianhai Du
- Department of Ophthalmology and Visual Sciences, Eye Institute, One Medical Center Drive, West Virginia UniversityMorgantownUnited States
- Department of Biochemistry, One Medical Center Drive, West Virginia UniversityMorgantownUnited States
| | - Saravanan Kolandaivelu
- Department of Ophthalmology and Visual Sciences, Eye Institute, One Medical Center Drive, West Virginia UniversityMorgantownUnited States
- Department of Biochemistry, One Medical Center Drive, West Virginia UniversityMorgantownUnited States
| |
Collapse
|
209
|
Jassim AH, Fan Y, Pappenhagen N, Nsiah NY, Inman DM. Oxidative Stress and Hypoxia Modify Mitochondrial Homeostasis During Glaucoma. Antioxid Redox Signal 2021; 35:1341-1357. [PMID: 33736457 PMCID: PMC8817702 DOI: 10.1089/ars.2020.8180] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Aims: Cellular response to hypoxia can include transition from respiration to glycolysis via upregulation of glycolytic enzymes and transporters, as well as mitophagy induction to eliminate surplus mitochondria. Our purpose was to evaluate the impact of hypoxia-inducible factor-1α (HIF-1α) stabilization on mitochondrial homeostasis and oxidative stress in a chronic model of glaucoma. Results: Retina and optic nerve (ON) were evaluated from young and aged DBA/2J (D2) glaucoma model mice and the control strain, the DBA/2-Gpnmb+. Hypoxic retinal ganglion cells (RGCs) were observed in young and aged D2 retina, with a significant increase in HIF-1α protein in the aged D2 retina. Reactive oxygen species observed in young D2 retina and ON were followed by significant decreases in antioxidant capacity in aged D2 retina and ON. HIF-1α targets such as neuron-specific glucose transporter-3 and lactate dehydrogenase were decreased or unchanged, respectively, in aged D2 retina despite an increased hypoxia response in RGCs. Mitochondrial mass was decreased in aged D2 retina concomitant with decreased mitochondrially encoded electron transport chain transcripts despite a stable nuclear-encoded TFAM (mitochondrial transcription factor), suggesting a breakdown in the nuclear-mitochondrial communication. Decreased mitophagy-associated proteins p62 and Rheb were observed in aged D2 retina, although p62 was significantly increased in the aged D2 ON. Innovation and Conclusion: The increased reactive oxygen species concomitant with HIF-1α upregulation despite reduced glucose transporters, mis-match of nuclear- and mitochondrial-encoded transcripts, and signs of reduced mitophagy suggest that retinas from D2 mice with chronic intraocular pressure elevation transition to pseudohypoxia without consistent metabolic reprogramming before significant RGC loss. Antioxid. Redox Signal. 35, 1341-1357.
Collapse
Affiliation(s)
- Assraa Hassan Jassim
- Department of Pharmaceutical Sciences, Northeast Ohio Medical University, Rootstown, Ohio, USA
| | - Yan Fan
- Department of Pharmaceutical Sciences, North Texas Eye Research Institute, University of North Texas Health Science Center, Fort Worth, Texas, USA
| | - Nathaniel Pappenhagen
- Department of Pharmaceutical Sciences, North Texas Eye Research Institute, University of North Texas Health Science Center, Fort Worth, Texas, USA
- Kent State University School of Biomedical Sciences, Kent, Ohio, USA
| | - Nana Yaa Nsiah
- Department of Pharmaceutical Sciences, North Texas Eye Research Institute, University of North Texas Health Science Center, Fort Worth, Texas, USA
| | - Denise M. Inman
- Department of Pharmaceutical Sciences, North Texas Eye Research Institute, University of North Texas Health Science Center, Fort Worth, Texas, USA
- Address correspondence to: Dr. Denise M. Inman, Department of Pharmaceutical Sciences, North Texas Eye Research Institute, University of North Texas Health Science Center, 3500 Camp Bowie Boulevard, Fort Worth, TX 76107, USA
| |
Collapse
|
210
|
Association between Daily Niacin Intake and Glaucoma: National Health and Nutrition Examination Survey. Nutrients 2021; 13:nu13124263. [PMID: 34959814 PMCID: PMC8709149 DOI: 10.3390/nu13124263] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Revised: 11/15/2021] [Accepted: 11/22/2021] [Indexed: 12/15/2022] Open
Abstract
Background and Aims: To determine the relationship between dietary intake of niacin and glaucoma using the data from the 2005 to 2008 National Health and Nutrition Examination Survey (NHANES). Methods: Subjects aged 40 years and older who participated in the dietary intake interview and vision health questionnaire of NHANES were included in the study. Glaucoma diagnosis by self-report was utilized. Additionally, glaucoma diagnosis by fundus imaging and International Society Geographical and Epidemiological Ophthalmology (ISGEO) criteria was used in subjects with available qualified retinal imaging. Survey logistic regression analyses were used to examine the association between daily niacin consumption and glaucoma. Results: A total of 5768 participants were included in the study. There was a significant decrease in the crude odds of self-reported glaucoma in the third (OR 0.57, 95% Cl 0.43–0.76; p < 0.001) and fourth (OR 0.57, 95% Cl 0.37–0.90; p = 0.018) quartiles of daily niacin consumption, which equated to 21.01 to 28.22 mg/day and greater than 28.22 mg/day, respectively. A similar trend was found using fundus imaging of subjects with niacin intake in the third (OR 0.42, 95% Cl 0.25–0.72; p = 0.002) and fourth (OR 0.36, 95% Cl 0.20–0.67; p = 0.002) quartiles. After adjusting for covariates, the odds of glaucoma based on fundus imaging remained significantly lower for niacin intake in the third (OR 0.49, 95% Cl 0.28–0.87; p = 0.016) and fourth (OR 0.48, 95% Cl 0.26–0.89; p = 0.022) quartile levels. Using ISGEO criteria, there was no significant association between glaucoma and daily niacin consumption. Conclusions: Greater niacin intake may be associated with a lower chance of developing glaucoma.
Collapse
|
211
|
De Moraes CG, John SWM, Williams PA, Blumberg DM, Cioffi GA, Liebmann JM. Nicotinamide and Pyruvate for Neuroenhancement in Open-Angle Glaucoma: A Phase 2 Randomized Clinical Trial. JAMA Ophthalmol 2021; 140:11-18. [PMID: 34792559 DOI: 10.1001/jamaophthalmol.2021.4576] [Citation(s) in RCA: 75] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022]
Abstract
Importance Open-angle glaucoma may continue to progress despite significant lowering of intraocular pressure (IOP). Preclinical research has suggested that enhancing mitochondrial function and energy production may enhance retinal ganglion cell survival in animal models of glaucoma, but there is scant information on its effectiveness in a clinical setting. Objective To test the hypothesis that a combination of nicotinamide and pyruvate can improve retinal ganglion cell function in human glaucoma as measured with standard automated perimetry. Design, Setting, and Participants In this phase 2, randomized, double-blind, placebo-controlled clinical trial at a single academic institution, 197 patients were assessed for eligibility. Of these, 42 patients with treated open-angle glaucoma and moderate visual field loss in at least 1 eye were selected for inclusion and randomized. A total of 32 completed the study and were included in the final analysis. The mean (SD) age was 64.6 (9.8) years. Twenty-one participants (66%) were female. Participant race and ethnicity data were collected via self-report to ensure the distribution reflected that observed in clinical practice in the US but are not reported here to protect patient privacy. Recruitment took place in April 2019 and patients were monitored through December 2020. Data were analyzed from January to May 2021. Interventions Ascending oral doses of nicotinamide (1000 to 3000 mg) and pyruvate (1500 to 3000 mg) vs placebo (2:1 randomization). Main Outcomes and Measures Number of visual field test locations improving beyond normal variability in the study eye. Secondary end points were the rates of change of visual field global indices (mean deviation [MD], pattern standard deviation [PSD], and visual field index [VFI]). Results Twenty-two of 29 participants (76%) randomized to the intervention group and 12 of 13 participants (92%) randomized to placebo received their allocation, and 32 participants (32 eyes; ratio 21:11) completed the study (21 from the intervention group and 11 from the placebo group). Median (IQR) follow-up time was 2.2 (2.0-2.4) months. No serious adverse events were reported during the study. The number of improving test locations was significantly higher in the treatment group than in the placebo group (median [IQR], 15 [6-25] vs 7 [6-11]; P = .005). Rates of change of PSD suggested improvement with treatment compared with placebo (median, -0.06 vs 0.02 dB per week; 95% CI, 0.02 to 0.24; P = .02) but not MD (0.04 vs -0.002 dB per week; 95% CI, -0.27 to 0.09; P = .35) or VFI (0.09 vs -0.02% per week; 95% CI, -0.53 to 0.36; P = .71). Conclusions and Relevance A combination of nicotinamide and pyruvate yielded significant short-term improvement in visual function, supporting prior experimental research suggesting a role for these agents in neuroprotection for individuals with glaucoma and confirming the need for long-term studies to establish their usefulness in slowing progression. Trial Registration ClinicalTrials.gov Identifier: NCT03797469.
Collapse
Affiliation(s)
- Carlos Gustavo De Moraes
- Bernard and Shirlee Brown Glaucoma Research Laboratory, Department of Ophthalmology, Columbia University Irving Medical Center, New York, New York
| | - Simon W M John
- Bernard and Shirlee Brown Glaucoma Research Laboratory, Department of Ophthalmology, Columbia University Irving Medical Center, New York, New York.,Howard Hughes Medical Institute, Chevy Chase, Maryland.,Zuckerman Mind Brain Behavior Institute, Columbia University, New York, New York
| | - Pete A Williams
- Division of Eye and Vision, Department of Clinical Neuroscience, St Erik Eye Hospital, Karolinska Institutet, Stockholm, Sweden
| | - Dana M Blumberg
- Bernard and Shirlee Brown Glaucoma Research Laboratory, Department of Ophthalmology, Columbia University Irving Medical Center, New York, New York
| | - George A Cioffi
- Bernard and Shirlee Brown Glaucoma Research Laboratory, Department of Ophthalmology, Columbia University Irving Medical Center, New York, New York
| | - Jeffrey M Liebmann
- Bernard and Shirlee Brown Glaucoma Research Laboratory, Department of Ophthalmology, Columbia University Irving Medical Center, New York, New York
| |
Collapse
|
212
|
Liu P, Huang H, Fang F, Liu L, Li L, Feng X, Chen W, Dalal R, Sun Y, Hu Y. Neuronal NMNAT2 Overexpression Does Not Achieve Significant Neuroprotection in Experimental Autoimmune Encephalomyelitis/Optic Neuritis. Front Cell Neurosci 2021; 15:754651. [PMID: 34707482 PMCID: PMC8542903 DOI: 10.3389/fncel.2021.754651] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Accepted: 09/10/2021] [Indexed: 11/20/2022] Open
Abstract
Optic neuritis, inflammation, and demyelination of the optic nerve (ON), is one of the most common clinical manifestations of multiple sclerosis; affected patients suffer persistent visual symptoms due to ON degeneration and secondary retinal ganglion cell (RGC) death. The mouse experimental autoimmune encephalomyelitis (EAE) model replicates optic neuritis and significant RGC soma and axon loss. Nicotinamide mononucleotide adenylyltransferases (NMNATs) are NAD+-synthetic enzymes that have been shown to be essential for axon integrity, activation of which significantly delays axonal Wallerian degeneration. NMNAT2, which is enriched in axons, has been proposed as a promising therapeutic target for axon injury-induced neurodegeneration. We therefore investigated whether activation of NMNAT2 can be used as a gene therapy strategy for neuroprotection in EAE/optic neuritis. To avoid the confounding effects in inflammatory cells, which play important roles in EAE initiation and progression, we used an RGC-specific promoter to drive the expression of the long half-life NMNAT2 mutant in mouse RGCs in vivo. However, optical coherence tomography in vivo retina imaging did not reveal significant protection of the ganglion cell complex, and visual function assays, pattern electroretinography, and optokinetic response also showed no improvement in mice with NMNAT2 overexpression. Postmortem histological analysis of retina wholemounts and semithin sections of ON confirmed the in vivo results: NMNAT2 activation in RGCs does not provide significant neuroprotection of RGCs in EAE/optic neuritis. Our studies suggest that a different degenerative mechanism than Wallerian degeneration is involved in autoimmune inflammatory axonopathy and that NMNAT2 may not be a major contributor to this mechanism.
Collapse
Affiliation(s)
- Pingting Liu
- Department of Ophthalmology, Stanford University School of Medicine, Palo Alto, CA, United States
| | - Haoliang Huang
- Department of Ophthalmology, Stanford University School of Medicine, Palo Alto, CA, United States
| | - Fang Fang
- Department of Ophthalmology, Stanford University School of Medicine, Palo Alto, CA, United States.,Department of Ophthalmology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Liang Liu
- Department of Ophthalmology, Stanford University School of Medicine, Palo Alto, CA, United States
| | - Liang Li
- Department of Ophthalmology, Stanford University School of Medicine, Palo Alto, CA, United States
| | - Xue Feng
- Department of Ophthalmology, Stanford University School of Medicine, Palo Alto, CA, United States
| | - Wei Chen
- Department of Ophthalmology, Stanford University School of Medicine, Palo Alto, CA, United States
| | - Roopa Dalal
- Department of Ophthalmology, Stanford University School of Medicine, Palo Alto, CA, United States
| | - Yang Sun
- Department of Ophthalmology, Stanford University School of Medicine, Palo Alto, CA, United States
| | - Yang Hu
- Department of Ophthalmology, Stanford University School of Medicine, Palo Alto, CA, United States
| |
Collapse
|
213
|
Yang Y, Li X, Wang J, Tan J, Fitzmaurice B, Nishina PM, Sun K, Tian W, Liu W, Liu X, Chang B, Zhu X. A missense mutation in Pitx2 leads to early-onset glaucoma via NRF2-YAP1 axis. Cell Death Dis 2021; 12:1017. [PMID: 34716303 PMCID: PMC8556256 DOI: 10.1038/s41419-021-04331-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 10/12/2021] [Accepted: 10/18/2021] [Indexed: 12/21/2022]
Abstract
Glaucoma is a leading cause of blindness, affecting 70 million people worldwide. Owing to the similarity in anatomy and physiology between human and mouse eyes and the ability to genetically manipulate mice, mouse models are an invaluable resource for studying mechanisms underlying disease phenotypes and for developing therapeutic strategies. Here, we report the discovery of a new mouse model of early-onset glaucoma that bears a transversion substitution c. G344T, which results in a missense mutation, p. R115L in PITX2. The mutation causes an elevation in intraocular pressure (IOP) and progressive death of retinal ganglion cells (RGC). These ocular phenotypes recapitulate features of pathologies observed in human glaucoma. Increased oxidative stress was evident in the inner retina. We demonstrate that the mutant PITX2 protein was not capable of binding to Nuclear factor-like 2 (NRF2), which regulates Pitx2 expression and nuclear localization, and to YAP1, which is necessary for co-initiation of transcription of downstream targets. PITX2-mediated transcription of several antioxidant genes were also impaired. Treatment with N-Acetyl-L-cysteine exerted a profound neuroprotective effect on glaucoma-associated neuropathies, presumably through inhibition of oxidative stress. Our study demonstrates that a disruption of PITX2 leads to glaucoma optic pathogenesis and provides a novel early-onset glaucoma model that will enable elucidation of mechanisms underlying the disease as well as to serve as a resource to test new therapeutic strategies.
Collapse
Affiliation(s)
- Yeming Yang
- Sichuan Provincial Key Laboratory for Human Disease Gene Study, Center for Medical Genetics, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, 610072, Chengdu, Sichuan, China
- Henan Eye Institute, Henan Eye Hospital, People's Hospital of Zhengzhou University, Henan Provincial People's Hospital, 450003, Zhengzhou, Henan, China
- Research Unit for Blindness Prevention of Chinese Academy of Medical Sciences (2019RU026), Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, 610072, Chengdu, Sichuan, China
| | - Xiao Li
- Sichuan Provincial Key Laboratory for Human Disease Gene Study, Center for Medical Genetics, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, 610072, Chengdu, Sichuan, China
| | - Jieping Wang
- The Jackson Laboratory, Bar Harbor, ME, 04609, USA
| | - Junkai Tan
- Xiamen Eye Center, Xiamen University, 361006, Xiamen, Fujian, China
| | | | | | - Kuanxiang Sun
- Sichuan Provincial Key Laboratory for Human Disease Gene Study, Center for Medical Genetics, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, 610072, Chengdu, Sichuan, China
| | - Wanli Tian
- Sichuan Provincial Key Laboratory for Human Disease Gene Study, Center for Medical Genetics, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, 610072, Chengdu, Sichuan, China
| | - Wenjing Liu
- Sichuan Provincial Key Laboratory for Human Disease Gene Study, Center for Medical Genetics, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, 610072, Chengdu, Sichuan, China
| | - Xuyang Liu
- Xiamen Eye Center, Xiamen University, 361006, Xiamen, Fujian, China.
- Department of Ophthalmology, Shenzhen People's Hospital, the 2nd Clinical Medical College, Jinan University, 518020, Shenzhen, China.
| | - Bo Chang
- The Jackson Laboratory, Bar Harbor, ME, 04609, USA.
| | - Xianjun Zhu
- Sichuan Provincial Key Laboratory for Human Disease Gene Study, Center for Medical Genetics, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, 610072, Chengdu, Sichuan, China.
- Henan Eye Institute, Henan Eye Hospital, People's Hospital of Zhengzhou University, Henan Provincial People's Hospital, 450003, Zhengzhou, Henan, China.
- Research Unit for Blindness Prevention of Chinese Academy of Medical Sciences (2019RU026), Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, 610072, Chengdu, Sichuan, China.
| |
Collapse
|
214
|
Sharif NA. Therapeutic Drugs and Devices for Tackling Ocular Hypertension and Glaucoma, and Need for Neuroprotection and Cytoprotective Therapies. Front Pharmacol 2021; 12:729249. [PMID: 34603044 PMCID: PMC8484316 DOI: 10.3389/fphar.2021.729249] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Accepted: 08/18/2021] [Indexed: 12/11/2022] Open
Abstract
Damage to the optic nerve and the death of associated retinal ganglion cells (RGCs) by elevated intraocular pressure (IOP), also known as glaucoma, is responsible for visual impairment and blindness in millions of people worldwide. The ocular hypertension (OHT) and the deleterious mechanical forces it exerts at the back of the eye, at the level of the optic nerve head/optic disc and lamina cribosa, is the only modifiable risk factor associated with glaucoma that can be treated. The elevated IOP occurs due to the inability of accumulated aqueous humor (AQH) to egress from the anterior chamber of the eye due to occlusion of the major outflow pathway, the trabecular meshwork (TM) and Schlemm’s canal (SC). Several different classes of pharmaceutical agents, surgical techniques and implantable devices have been developed to lower and control IOP. First-line drugs to promote AQH outflow via the uveoscleral outflow pathway include FP-receptor prostaglandin (PG) agonists (e.g., latanoprost, travoprost and tafluprost) and a novel non-PG EP2-receptor agonist (omidenepag isopropyl, Eybelis®). TM/SC outflow enhancing drugs are also effective ocular hypotensive agents (e.g., rho kinase inhibitors like ripasudil and netarsudil; and latanoprostene bunod, a conjugate of a nitric oxide donor and latanoprost). One of the most effective anterior chamber AQH microshunt devices is the Preserflo® microshunt which can lower IOP down to 10–13 mmHg. Other IOP-lowering drugs and devices on the horizon will be also discussed. Additionally, since elevated IOP is only one of many risk factors for development of glaucomatous optic neuropathy, a treatise of the role of inflammatory neurodegeneration of the optic nerve and retinal ganglion cells and appropriate neuroprotective strategies to mitigate this disease will also be reviewed and discussed.
Collapse
Affiliation(s)
- Najam A Sharif
- Global Alliances and External Research, Ophthalmology Innovation Center, Santen Inc., Emeryville, CA, United States
| |
Collapse
|
215
|
Immune responses to injury and their links to eye disease. Transl Res 2021; 236:52-71. [PMID: 34051364 PMCID: PMC8380715 DOI: 10.1016/j.trsl.2021.05.005] [Citation(s) in RCA: 90] [Impact Index Per Article: 22.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 05/12/2021] [Accepted: 05/24/2021] [Indexed: 12/31/2022]
Abstract
The eye is regarded as an immune privileged site. Since the presence of a vasculature would impair vision, the vasculature of the eye is located outside of the central light path. As a result, many regions of the eye evolved mechanisms to deliver immune cells to sites of dysgenesis, injury, or in response to the many age-related pathologies. While the purpose of these immune responses is reparative or protective, cytokines released by immune cells compromise visual acuity by inducing inflammation and fibrosis. The response to traumatic or pathological injury is distinct in different regions of the eye. Age-related diseases impact both the anterior and posterior segment and lead to reduced quality of life and blindness. Here we focus attention on the role that inflammation and fibrosis play in the progression of age-related pathologies of the cornea and the lens as well as in glaucoma, the formation of epiretinal membranes, and in proliferative vitreoretinopathy.
Collapse
Key Words
- 2ryERM
- A T-helper cell that expresses high levels of IL-17 which can suppress T-regulatory cell function
- A cytokine expressed early during inflammation that attracts neutrophils
- A cytokine expressed early during inflammation that attracts neutrophils, sometimes referred to as monocyte chemoattractant protein-1 (MCP-1))
- A mouse model that lacks functional T and B cells and used to study the immune response
- A pigmented mouse strain used for research and known to mount a primarily Th1 response to infection
- A protein encoded by the ADGRE1 gene that, in mice, is expressed primarily on macrophages
- A strain of pigmented mice used in glaucoma research
- ACAID
- APCs
- ASC
- An albino mouse strain used for research and known to mount a primarily Th2 response to infection
- Antigen Presenting Cells, this class includes dendritic cells and monocytes
- BALB/c
- BM
- C57BL6
- CCL2
- CD45
- CNS
- CXCL1
- Central Nervous System
- Cluster of differentiation 45 antigen
- DAMPs
- DBA/2J
- EBM
- ECM
- EMT
- ERM
- Epithelial Basement Membrane
- F4/80
- FGF2
- HA =hyaluronic acid
- HSK
- HSP
- HSPGs
- HSV
- ICN
- IL-20
- IL6
- ILM
- IOP
- Inner (or internal) limiting membrane
- Interleukin 6
- Interleukin-20
- MAGP1
- MHC-II
- Major histocompatibility complex type II, a class of MHC proteins typically found only on APCs
- Microfibril-associated glycoprotein 1
- N-cad
- N-cadherin
- NEI
- NK
- National Eye Institute
- Natural killer T cells
- PCO
- PDGF
- PDR
- PVD
- PVR
- Platelet derived growth factor
- Posterior capsular opacification
- RGC
- RPE
- RRD
- Rag1-/-
- Retinal ganglion cells
- Retinal pigment epithelial cells
- SMAD
- Sons of Mothers Against Decapentaplegic, SMADs are a class of molecules that mediate TGF and bone morphogenetic protein signaling
- T-helper cell 1 response, proinflammatory adaptive response involving interferon gamma and associated with autoimmunity
- T-helper cell 2 response involving IgE and interleukins 4,5, and 13, also induces the anti-inflammatory interleukin 10 family cytokines
- T-regulatory cell
- TG
- TGF1
- TM
- TNF
- Th1
- Th17
- Th2
- Transforming growth factor 1
- Treg
- Tumor necrosis factor a cytokine produced during inflammation
- VEGF
- Vascular endothelial growth factor
- WHO
- World Health Organization
- anterior chamber immune deviation
- anterior subcapsular cataracts
- basement membrane
- damage-associated molecular patterns
- epiretinal membrane
- epiretinal membrane secondary to disease pathology
- epithelial-mesenchymal transition
- extracellular matrix
- fibroblast growth factor 2, also referred to as basic FGF
- heat shock protein
- heparan sulfate proteoglycans
- herpes simplex virus
- herpes stromal keratitis
- iERM
- idiopathic epiretinal membrane
- intraepithelial corneal nerves
- intraocular pressure
- mTOR
- mechanistic target of rapamycin, a protein kinase encoded by the MTOR genes that regulates a variety of signal transduction events including cell growth, autophagy and actin cytoskeleton
- posterior vitreous detachment
- proliferative diabetic retinopathy
- proliferative vitreoretinopathy
- rhegmatogenous (rupture, tear) retinal detachment
- trabecular meshwork
- trigeminal ganglion
- αSMA
- α−Smooth muscle actin, a class of actin expressed in mesenchymal cells
Collapse
|
216
|
Abstract
Optic nerve health is essential for proper function of the visual system. However, the pathophysiology of certain neurodegenerative disease processes affecting the optic nerve, such as glaucoma, is not fully understood. Recently, it was hypothesized that a lack of proper clearance of neurotoxins contributes to neurodegenerative diseases. The ability to clear metabolic waste is essential for tissue homeostasis in mammals, including humans. While the brain lacks the traditional lymphatic drainage system identified in other anatomical regions, there is growing evidence of a glymphatic system in the central nervous system, which structurally includes the optic nerve. Named to acknowledge the supportive role of astroglial cells, this perivascular fluid drainage system is essential to remove toxic metabolites from the central nervous system. Herein, we review existing literature describing the physiology and dysfunction of the glymphatic system specifically as it relates to the optic nerve. We summarize key imaging studies demonstrating the existence of a glymphatic system in the optic nerves of wild-type rodents, aquaporin 4-null rodents, and humans; glymphatic imaging studies in diseases where the optic nerve is impaired; and current evidence regarding pharmacological and lifestyle interventions that may help promote glymphatic function to improve optic nerve health. We conclude by highlighting future research directions that could be applied to improve imaging detection and guide therapeutic interventions for diseases affecting the optic nerve.
Collapse
Affiliation(s)
- Anisha Kasi
- Department of Ophthalmology, NYU Grossman School of Medicine, NYU Langone Health, New York University, New York, NY, USA
| | - Crystal Liu
- Department of Ophthalmology, NYU Grossman School of Medicine, NYU Langone Health, New York University, New York, NY, USA
| | - Muneeb A Faiq
- Department of Ophthalmology, NYU Grossman School of Medicine, NYU Langone Health, New York University, New York, NY, USA
| | - Kevin C Chan
- Department of Ophthalmology; Department of Radiology; Neuroscience Institute, NYU Grossman School of Medicine, NYU Langone Health; Center for Neural Science, College of Arts and Science, New York University, New York, NY, USA
| |
Collapse
|
217
|
Non-drug interventions in glaucoma: Putative roles for lifestyle, diet and nutritional supplements. Surv Ophthalmol 2021; 67:675-696. [PMID: 34563531 DOI: 10.1016/j.survophthal.2021.09.002] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Revised: 09/15/2021] [Accepted: 09/20/2021] [Indexed: 02/06/2023]
Abstract
Glaucoma is a major ocular neurodegenerative disease characterized by progressive retinal ganglion cells degeneration and sight loss. Current treatment options have been limited to reducing intraocular pressure (IOP), known as the leading risk factor for this disease; however, glaucoma can develop even with low or normal IOP and progress despite controlling IOP values. Lifestyle, dietary habits, and supplementation may influence some of the risk factors and pathophysiological mechanisms underlying glaucoma development and progression; thus, the role of this complementary and alternative medicine in glaucoma has received great interest from both patients and ophthalmologists. We provide a summary of the current evidence concerning the relationship between lifestyle, dietary habits, and effects of supplements on the incidence and progression of glaucoma and their targets and associated mechanisms. The data suggest the existence of a therapeutic potential that needs to be further explored with both preclinical and rigorous clinical studies.
Collapse
|
218
|
Enz TJ, Tribble JR, Williams PA. Comparison of Glaucoma-Relevant Transcriptomic Datasets Identifies Novel Drug Targets for Retinal Ganglion Cell Neuroprotection. J Clin Med 2021; 10:3938. [PMID: 34501387 PMCID: PMC8432026 DOI: 10.3390/jcm10173938] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2021] [Revised: 08/10/2021] [Accepted: 08/27/2021] [Indexed: 12/20/2022] Open
Abstract
Glaucoma is a leading cause of blindness and is characterized by the progressive dysfunction and irreversible death of retinal ganglion cells. We aimed to identify shared differentially expressed genes (DE genes) between different glaucoma relevant models of retinal ganglion cell injury using existing RNA-sequencing data, thereby discovering targets for neuroprotective therapies. A comparison of DE genes from publicly available transcriptomic datasets identified 12 shared DE genes. The Comparative Toxicogenomics Database (CTD) was screened for compounds targeting a significant proportion of the identified DE genes. Forty compounds were identified in the CTD that interact with >50% of these shared DE genes. We next validated this approach by testing select compounds for an effect on retinal ganglion cell survival using a mouse retinal explant model. Folic acid, genistein, SB-431542, valproic acid, and WY-14643 (pirinixic acid) were tested. Folic acid, valproic acid, and WY-14643 demonstrated significant protection against retinal ganglion cell death in this model. The increasing prevalence of open access-omics data presents a resource to discover targets for future therapeutic investigation.
Collapse
Affiliation(s)
| | | | - Pete A. Williams
- Department of Clinical Neuroscience, Division of Eye and Vision, St. Erik Eye Hospital, Karolinska Institutet, 171 64 Stockholm, Sweden; (T.J.E.); (J.R.T.)
| |
Collapse
|
219
|
Dammak A, Huete-Toral F, Carpena-Torres C, Martin-Gil A, Pastrana C, Carracedo G. From Oxidative Stress to Inflammation in the Posterior Ocular Diseases: Diagnosis and Treatment. Pharmaceutics 2021; 13:1376. [PMID: 34575451 PMCID: PMC8467715 DOI: 10.3390/pharmaceutics13091376] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 08/16/2021] [Accepted: 08/20/2021] [Indexed: 12/17/2022] Open
Abstract
Most irreversible blindness observed with glaucoma and retina-related ocular diseases, including age-related macular degeneration and diabetic retinopathy, have their origin in the posterior segment of the eye, making their physiopathology both complex and interconnected. In addition to the age factor, these diseases share the same mechanism disorder based essentially on oxidative stress. In this context, the imbalance between the production of reactive oxygen species (ROS) mainly by mitochondria and their elimination by protective mechanisms leads to chronic inflammation. Oxidative stress and inflammation share a close pathophysiological process, appearing simultaneously and suggesting a relationship between both mechanisms. The biochemical end point of these two biological alarming systems is the release of different biomarkers that can be used in the diagnosis. Furthermore, oxidative stress, initiating in the vulnerable tissue of the posterior segment, is closely related to mitochondrial dysfunction, apoptosis, autophagy dysfunction, and inflammation, which are involved in each disease progression. In this review, we have analyzed (1) the oxidative stress and inflammatory processes in the back of the eye, (2) the importance of biomarkers, detected in systemic or ocular fluids, for the diagnosis of eye diseases based on recent studies, and (3) the treatment of posterior ocular diseases, based on long-term clinical studies.
Collapse
Affiliation(s)
- Azza Dammak
- Ocupharm Group Research, Faculty of Optic and Optometry, University Complutense of Madrid, C/Arcos del Jalon 118, 28037 Madrid, Spain; (A.D.); (F.H.-T.); (C.C.-T.); (A.M.-G.); (C.P.)
| | - Fernando Huete-Toral
- Ocupharm Group Research, Faculty of Optic and Optometry, University Complutense of Madrid, C/Arcos del Jalon 118, 28037 Madrid, Spain; (A.D.); (F.H.-T.); (C.C.-T.); (A.M.-G.); (C.P.)
| | - Carlos Carpena-Torres
- Ocupharm Group Research, Faculty of Optic and Optometry, University Complutense of Madrid, C/Arcos del Jalon 118, 28037 Madrid, Spain; (A.D.); (F.H.-T.); (C.C.-T.); (A.M.-G.); (C.P.)
| | - Alba Martin-Gil
- Ocupharm Group Research, Faculty of Optic and Optometry, University Complutense of Madrid, C/Arcos del Jalon 118, 28037 Madrid, Spain; (A.D.); (F.H.-T.); (C.C.-T.); (A.M.-G.); (C.P.)
| | - Cristina Pastrana
- Ocupharm Group Research, Faculty of Optic and Optometry, University Complutense of Madrid, C/Arcos del Jalon 118, 28037 Madrid, Spain; (A.D.); (F.H.-T.); (C.C.-T.); (A.M.-G.); (C.P.)
| | - Gonzalo Carracedo
- Ocupharm Group Research, Faculty of Optic and Optometry, University Complutense of Madrid, C/Arcos del Jalon 118, 28037 Madrid, Spain; (A.D.); (F.H.-T.); (C.C.-T.); (A.M.-G.); (C.P.)
- Department of Optometry and Vsiion, Faculty of Optic and Optometry, University Complutense of Madrid, C/Arcos del Jalon 118, 28037 Madrid, Spain
| |
Collapse
|
220
|
Tang J, Zhuo Y, Li Y. Effects of Iron and Zinc on Mitochondria: Potential Mechanisms of Glaucomatous Injury. Front Cell Dev Biol 2021; 9:720288. [PMID: 34447755 PMCID: PMC8383321 DOI: 10.3389/fcell.2021.720288] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Accepted: 07/22/2021] [Indexed: 12/26/2022] Open
Abstract
Glaucoma is the most substantial cause of irreversible blinding, which is accompanied by progressive retinal ganglion cell damage. Retinal ganglion cells are energy-intensive neurons that connect the brain and retina, and depend on mitochondrial homeostasis to transduce visual information through the brain. As cofactors that regulate many metabolic signals, iron and zinc have attracted increasing attention in studies on neurons and neurodegenerative diseases. Here, we summarize the research connecting iron, zinc, neuronal mitochondria, and glaucomatous injury, with the aim of updating and expanding the current view of how retinal ganglion cells degenerate in glaucoma, which can reveal novel potential targets for neuroprotection.
Collapse
Affiliation(s)
- Jiahui Tang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Yehong Zhuo
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Yiqing Li
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
221
|
Hurley DJ, Irnaten M, O’Brien C. Metformin and Glaucoma-Review of Anti-Fibrotic Processes and Bioenergetics. Cells 2021; 10:cells10082131. [PMID: 34440899 PMCID: PMC8394782 DOI: 10.3390/cells10082131] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 08/13/2021] [Accepted: 08/16/2021] [Indexed: 02/06/2023] Open
Abstract
Glaucoma is the leading cause of irreversible blindness globally. With an aging population, disease incidence will rise with an enormous societal and economic burden. The treatment strategy revolves around targeting intraocular pressure, the principle modifiable risk factor, to slow progression of disease. However, there is a clear unmet clinical need to find a novel therapeutic approach that targets and halts the retinal ganglion cell (RGC) degeneration that occurs with fibrosis. RGCs are highly sensitive to metabolic fluctuations as a result of multiple stressors and thus their viability depends on healthy mitochondrial functioning. Metformin, known for its use in type 2 diabetes, has come to the forefront of medical research in multiple organ systems. Its use was recently associated with a 25% reduced risk of glaucoma in a large population study. Here, we discuss its application to glaucoma therapy, highlighting its effect on fibrotic signalling pathways, mitochondrial bioenergetics and NAD oxidation.
Collapse
Affiliation(s)
- Daire J. Hurley
- Department of Ophthalmology, Mater Misericordiae University Hospital, Eccles Street, D07 R2WY Dublin, Ireland; (M.I.); (C.O.)
- School of Medicine, University College Dublin, Belfield, D04 V1W8 Dublin, Ireland
- Correspondence:
| | - Mustapha Irnaten
- Department of Ophthalmology, Mater Misericordiae University Hospital, Eccles Street, D07 R2WY Dublin, Ireland; (M.I.); (C.O.)
| | - Colm O’Brien
- Department of Ophthalmology, Mater Misericordiae University Hospital, Eccles Street, D07 R2WY Dublin, Ireland; (M.I.); (C.O.)
- School of Medicine, University College Dublin, Belfield, D04 V1W8 Dublin, Ireland
| |
Collapse
|
222
|
Wu L, Liu J, Chang X, Zheng Y. The therapeutic effect of Healaflow in glaucoma surgery. Am J Transl Res 2021; 13:9729-9735. [PMID: 34540102 PMCID: PMC8430195] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2021] [Accepted: 02/25/2021] [Indexed: 06/13/2023]
Abstract
OBJECTIVE To explore the clinical effectiveness of Healaflow in primary angle-closure glaucoma surgery. METHODS From August 2018 to July 2019, 100 primary angle-closure glaucoma patients admitted to our ophthalmology department were divided into a control group (trabeculectomy alone) and an observation group (trabeculectomy + Halver), with 50 patients in each group, and 53 eyes in the control group and 56 eyes in the observation group. All the patients were followed up for half a year. The vision, intraocular pressure, bleb morphology, efficacy, and adverse reactions were evaluated. RESULTS There was no significant difference in the visual acuity between the two groups of patients during the follow-up (P > 0.05). The intraocular pressure levels in the two groups of patients during the follow-up were significantly lower than they were before the treatment (P < 0.05), and the difference in the intraocular pressure levels between the 6th month postoperative groups was significant (P < 0.05). During the last follow-up, 45 eyes in the control group developed functional filtration blebs, and 8 eyes had non-functioning filtration blebs. There were 55 functional filtration blebs and 1 non-functional filtration bleb in the observation group (χ2 = 4.731, P = 0.030). The total effective rate in the observation group was higher than it was in the control group (92.86% VS 77.36%, χ2 = 4.058, P = 0.044). During the follow-up period, the control group had 2 eyes with anterior chambers, 1 eye with low intraocular pressure, and 1 eye with an iris adhesion. The observation group had 1 eye with an anterior chamber, and no significant difference in the complications between the groups was evident (P > 0.05). CONCLUSION Healaflow is of great value in maintaining functional filtration blebs and in controlling and stabilizing intraocular pressure. It is safe and reliable in clinical application, and it helps to reduce the unstable intraocular pressure after glaucoma surgery caused by scar adhesions in the filtration channel.
Collapse
Affiliation(s)
- Liping Wu
- Department of Glaucoma and Cataract, Aier Eye Hospital Group Hankou Hospital of Wuhan Aier Eye HospitalWuhan, Hubei, China
| | - Junjie Liu
- Department of Fundas, Aier Eye Hospital Group Hankou Hospital of Wuhan Aier Eye HospitalWuhan, Hubei, China
| | - Xiaoke Chang
- Department of Glaucoma and Cataract, Aier Eye Hospital Group Hankou Hospital of Wuhan Aier Eye HospitalWuhan, Hubei, China
| | - Yijun Zheng
- Department of Glaucoma and Cataract, Aier Eye Hospital Group Hankou Hospital of Wuhan Aier Eye HospitalWuhan, Hubei, China
| |
Collapse
|
223
|
Donahue RJ, Fehrman RL, Gustafson JR, Nickells RW. BCLX L gene therapy moderates neuropathology in the DBA/2J mouse model of inherited glaucoma. Cell Death Dis 2021; 12:781. [PMID: 34376637 PMCID: PMC8355227 DOI: 10.1038/s41419-021-04068-x] [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: 03/08/2021] [Revised: 07/26/2021] [Accepted: 07/28/2021] [Indexed: 12/19/2022]
Abstract
Axonal degeneration of retinal ganglion cells (RGCs) causes blindness in glaucoma. Currently, there are no therapies that target axons to prevent them from degenerating. Activation of the BAX protein has been shown to be the determining step in the intrinsic apoptotic pathway that causes RGCs to die in glaucoma. A putative role for BAX in axonal degeneration is less well elucidated. BCLXL (BCL2L1) is the primary antagonist of BAX in RGCs. We developed a mCherry-BCLXL fusion protein, which prevented BAX recruitment and activation to the mitochondria in tissue culture cells exposed to staurosporine. This fusion protein was then packaged into adeno-associated virus serotype 2, which was used to transduce RGCs after intravitreal injection and force its overexpression. Transduced RGCs express mCherry-BCLXL throughout their somas and axons along the entire optic tract. In a model of acute optic nerve crush, the transgene prevented the recruitment of a GFP-BAX fusion protein to mitochondria and provided long-term somal protection up to 12 weeks post injury. To test the efficacy in glaucoma, DBA/2J mice were transduced at 5 months of age, just prior to the time they begin to exhibit ocular hypertension. Gene therapy with mCherry-BCLXL did not affect the longitudinal history of intraocular pressure elevation compared to naive mice but did robustly attenuate both RGC soma pathology and axonal degeneration in the optic nerve at both 10.5 and 12 months of age. BCLXL gene therapy is a promising candidate for glaucoma therapy.
Collapse
Affiliation(s)
- Ryan J Donahue
- Department of Ophthalmology and Visual Sciences, University of Wisconsin-Madison, Madison, WI, USA
- Cellular and Molecular Pathology Graduate Program, University of Wisconsin-Madison, Madison, WI, USA
| | - Rachel L Fehrman
- Department of Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Jenna R Gustafson
- Department of Neurobiology and Anatomy, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Robert W Nickells
- Department of Ophthalmology and Visual Sciences, University of Wisconsin-Madison, Madison, WI, USA.
- McPherson Eye Research Institute, University of Wisconsin-Madison, Madison, WI, USA.
| |
Collapse
|
224
|
Guo X, Zhou J, Starr C, Mohns EJ, Li Y, Chen EP, Yoon Y, Kellner CP, Tanaka K, Wang H, Liu W, Pasquale LR, Demb JB, Crair MC, Chen B. Preservation of vision after CaMKII-mediated protection of retinal ganglion cells. Cell 2021; 184:4299-4314.e12. [PMID: 34297923 PMCID: PMC8530265 DOI: 10.1016/j.cell.2021.06.031] [Citation(s) in RCA: 90] [Impact Index Per Article: 22.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Revised: 04/22/2021] [Accepted: 06/24/2021] [Indexed: 12/18/2022]
Abstract
Retinal ganglion cells (RGCs) are the sole output neurons that transmit visual information from the retina to the brain. Diverse insults and pathological states cause degeneration of RGC somas and axons leading to irreversible vision loss. A fundamental question is whether manipulation of a key regulator of RGC survival can protect RGCs from diverse insults and pathological states, and ultimately preserve vision. Here, we report that CaMKII-CREB signaling is compromised after excitotoxic injury to RGC somas or optic nerve injury to RGC axons, and reactivation of this pathway robustly protects RGCs from both injuries. CaMKII activity also promotes RGC survival in the normal retina. Further, reactivation of CaMKII protects RGCs in two glaucoma models where RGCs degenerate from elevated intraocular pressure or genetic deficiency. Last, CaMKII reactivation protects long-distance RGC axon projections in vivo and preserves visual function, from the retina to the visual cortex, and visually guided behavior.
Collapse
Affiliation(s)
- Xinzheng Guo
- Department of Ophthalmology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Jing Zhou
- Department of Ophthalmology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Christopher Starr
- Department of Ophthalmology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Ethan J Mohns
- Department of Neuroscience, Yale University School of Medicine, New Haven, CT 06511, USA
| | - Yidong Li
- Department of Neuroscience, Yale University School of Medicine, New Haven, CT 06511, USA
| | | | - Yonejung Yoon
- Department of Neurosurgery, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Christopher P Kellner
- Department of Neurosurgery, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Kohichi Tanaka
- Laboratory of Molecular Neuroscience, School of Biomedical Sciences and Medical Research Institute, Tokyo Medical and Dental University, Tokyo, Japan
| | - Hongbing Wang
- Department of Physiology, Michigan State University, East Lansing, MI 48824, USA
| | - Wei Liu
- Department of Ophthalmology and Visual Sciences, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Louis R Pasquale
- Department of Ophthalmology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Jonathan B Demb
- Department of Neuroscience, Yale University School of Medicine, New Haven, CT 06511, USA; Department of Ophthalmology and Visual Science, Yale University School of Medicine, New Haven, CT 06511, USA; Department of Cellular and Molecular Physiology, Yale University School of Medicine, New Haven, CT 06511, USA
| | - Michael C Crair
- Department of Neuroscience, Yale University School of Medicine, New Haven, CT 06511, USA; Department of Ophthalmology and Visual Science, Yale University School of Medicine, New Haven, CT 06511, USA
| | - Bo Chen
- Department of Ophthalmology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Department of Cell, Developmental and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA.
| |
Collapse
|
225
|
Shestopalov VI, Spurlock M, Gramlich OW, Kuehn MH. Immune Responses in the Glaucomatous Retina: Regulation and Dynamics. Cells 2021; 10:1973. [PMID: 34440742 PMCID: PMC8391899 DOI: 10.3390/cells10081973] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Revised: 07/27/2021] [Accepted: 07/28/2021] [Indexed: 12/27/2022] Open
Abstract
Glaucoma is a multifactorial disease resulting in progressive vision loss due to retinal ganglion cell (RGC) dysfunction and death. Early events in the pathobiology of the disease include oxidative, metabolic, or mechanical stress that acts upon RGC, causing these to rapidly release danger signals, including extracellular ATP, resulting in micro- and macroglial activation and neuroinflammation. Danger signaling also leads to the formation of inflammasomes in the retina that enable maturation of proinflammatory cytokines such IL-1β and IL-18. Chronic neuroinflammation can have directly damaging effects on RGC, but it also creates a proinflammatory environment and compromises the immune privilege of the retina. In particular, continuous synthesis of proinflammatory mediators such as TNFα, IL-1β, and anaphylatoxins weakens the blood-retina barrier and recruits or activates T-cells. Recent data have demonstrated that adaptive immune responses strongly exacerbate RGC loss in animal models of the disease as T-cells appear to target heat shock proteins displayed on the surface of stressed RGC to cause their apoptotic death. It is possible that dysregulation of these immune responses contributes to the continued loss of RGC in some patients.
Collapse
Affiliation(s)
- Valery I. Shestopalov
- Department of Ophthalmology, Miller School of Medicine, University of Miami, Miami, FL 33101, USA;
- Department of Cell and Developmental Biology, Miller School of Medicine, University of Miami, Miami, FL 33101, USA;
- Graduate Program in Neuroscience, Miller School of Medicine, University of Miami, Miami, FL 33101, USA
- Kharkevich Institute for Information Transmission Problems, RAS, 127051 Moscow, Russia
| | - Markus Spurlock
- Department of Cell and Developmental Biology, Miller School of Medicine, University of Miami, Miami, FL 33101, USA;
- Graduate Program in Neuroscience, Miller School of Medicine, University of Miami, Miami, FL 33101, USA
| | - Oliver W. Gramlich
- Department of Veterans Affairs, Center for the Prevention and Treatment of Visual Loss, Iowa City, IA 52246, USA;
- Department of Ophthalmology and Visual Sciences, University of Iowa, Iowa City, IA 52242, USA
- Department of Neuroscience and Pharmacology, University of Iowa, Iowa City, IA 52242, USA
| | - Markus H. Kuehn
- Department of Veterans Affairs, Center for the Prevention and Treatment of Visual Loss, Iowa City, IA 52246, USA;
- Department of Ophthalmology and Visual Sciences, University of Iowa, Iowa City, IA 52242, USA
- Interdisciplinary Graduate Program in Genetics, University of Iowa, Iowa City, IA 52242, USA
| |
Collapse
|
226
|
Colbert MK, Ho LC, van der Merwe Y, Yang X, McLellan GJ, Hurley SA, Field AS, Yun H, Du Y, Conner IP, Parra C, Faiq MA, Fingert JH, Wollstein G, Schuman JS, Chan KC. Diffusion Tensor Imaging of Visual Pathway Abnormalities in Five Glaucoma Animal Models. Invest Ophthalmol Vis Sci 2021; 62:21. [PMID: 34410298 PMCID: PMC8383913 DOI: 10.1167/iovs.62.10.21] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Purpose To characterize the visual pathway integrity of five glaucoma animal models using diffusion tensor imaging (DTI). Methods Two experimentally induced and three genetically determined models of glaucoma were evaluated. For inducible models, chronic IOP elevation was achieved via intracameral injection of microbeads or laser photocoagulation of the trabecular meshwork in adult rodent eyes. For genetic models, the DBA/2J mouse model of pigmentary glaucoma, the LTBP2 mutant feline model of congenital glaucoma, and the transgenic TBK1 mouse model of normotensive glaucoma were compared with their respective genetically matched healthy controls. DTI parameters, including fractional anisotropy, axial diffusivity, and radial diffusivity, were evaluated along the optic nerve and optic tract. Results Significantly elevated IOP relative to controls was observed in each animal model except for the transgenic TBK1 mice. Significantly lower fractional anisotropy and higher radial diffusivity were observed along the visual pathways of the microbead- and laser-induced rodent models, the DBA/2J mice, and the LTBP2-mutant cats compared with their respective healthy controls. The DBA/2J mice also exhibited lower axial diffusivity, which was not observed in the other models examined. No apparent DTI change was observed in the transgenic TBK1 mice compared with controls. Conclusions Chronic IOP elevation was accompanied by decreased fractional anisotropy and increased radial diffusivity along the optic nerve or optic tract, suggestive of disrupted microstructural integrity in both inducible and genetic glaucoma animal models. The effects on axial diffusivity differed between models, indicating that this DTI metric may represent different aspects of pathological changes over time and with severity.
Collapse
Affiliation(s)
- Max K Colbert
- Department of Ophthalmology, NYU Grossman School of Medicine, NYU Langone Health, New York University, New York, New York, United States
| | - Leon C Ho
- Department of Ophthalmology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States
| | - Yolandi van der Merwe
- Department of Ophthalmology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States
| | - Xiaoling Yang
- Department of Ophthalmology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States
| | - Gillian J McLellan
- Department of Ophthalmology and Visual Sciences, University of Wisconsin - Madison, Madison, Wisconsin, United States.,McPherson Eye Research Institute, University of Wisconsin - Madison, Madison, Wisconsin, United States
| | - Samuel A Hurley
- Department of Radiology, University of Wisconsin - Madison, Madison, Wisconsin, United States
| | - Aaron S Field
- Department of Radiology, University of Wisconsin - Madison, Madison, Wisconsin, United States
| | - Hongmin Yun
- Department of Ophthalmology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States
| | - Yiqin Du
- Department of Ophthalmology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States
| | - Ian P Conner
- Department of Ophthalmology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States
| | - Carlos Parra
- Department of Ophthalmology, NYU Grossman School of Medicine, NYU Langone Health, New York University, New York, New York, United States
| | - Muneeb A Faiq
- Department of Ophthalmology, NYU Grossman School of Medicine, NYU Langone Health, New York University, New York, New York, United States
| | - John H Fingert
- Department of Ophthalmology and Visual Sciences, University of Iowa College of Medicine, Iowa City, Iowa, United States
| | - Gadi Wollstein
- Department of Ophthalmology, NYU Grossman School of Medicine, NYU Langone Health, New York University, New York, New York, United States.,Center for Neural Science, College of Arts and Science, New York University, New York, New York, United States.,Department of Biomedical Engineering, New York University Tandon School of Engineering, Brooklyn, New York, United States
| | - Joel S Schuman
- Department of Ophthalmology, NYU Grossman School of Medicine, NYU Langone Health, New York University, New York, New York, United States.,Center for Neural Science, College of Arts and Science, New York University, New York, New York, United States.,Department of Biomedical Engineering, New York University Tandon School of Engineering, Brooklyn, New York, United States.,Neuroscience Institute, NYU Grossman School of Medicine, NYU Langone Health, New York University, New York, New York, United States
| | - Kevin C Chan
- Department of Ophthalmology, NYU Grossman School of Medicine, NYU Langone Health, New York University, New York, New York, United States.,Center for Neural Science, College of Arts and Science, New York University, New York, New York, United States.,Department of Biomedical Engineering, New York University Tandon School of Engineering, Brooklyn, New York, United States.,Neuroscience Institute, NYU Grossman School of Medicine, NYU Langone Health, New York University, New York, New York, United States.,Department of Radiology, NYU Grossman School of Medicine, NYU Langone Health, New York University, New York, New York, United States
| |
Collapse
|
227
|
Amato R, Rossino MG, Cammalleri M, Timperio AM, Fanelli G, Dal Monte M, Pucci L, Casini G. The Potential of Lisosan G as a Possible Treatment for Glaucoma. Front Pharmacol 2021; 12:719951. [PMID: 34393798 PMCID: PMC8355587 DOI: 10.3389/fphar.2021.719951] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2021] [Accepted: 07/19/2021] [Indexed: 12/12/2022] Open
Abstract
Lisosan G (LG), a fermented powder obtained from whole grains, is a nutritional supplement containing a variety of metabolites with documented antioxidant properties. We have recently demonstrated that orally administered LG protects diabetic rodent retinas from oxidative stress, inflammation, apoptosis, blood-retinal barrier disruption, and functional damage. Here, we investigated whether LG may exert protective effects in a model of glaucoma and measured the amounts of selected LG components that reach the retina after oral LG administration. Six-month-old DBA/2J mice were given an aqueous LG solution in place of drinking water for 2 mo. During the 2 mo of treatment with LG, the intraocular pressure (IOP) was monitored and the retinal ganglion cell (RGC) functional activity was recorded with pattern-electroretinography (PERG). At the end of the 2-mo period, the expression of oxidative stress and inflammatory markers was measured with qPCR, and RGC survival or macroglial activation were assessed with immunofluorescence. Alternatively, LG was administered by gavage and the concentrations of four of the main LG components (nicotinamide, gallic acid, 4-hydroxybenzoic acid, and quercetin) were measured in the retinas in the following 24 h using mass spectrometry. LG treatment in DBA/2J mice did not influence IOP, but it affected RGC function since PERG amplitude was increased and PERG latency was decreased with respect to untreated DBA/2J mice. This improvement of RGC function was concomitant with a significant decrease of both oxidative stress and inflammation marker expression, of RGC loss, and of macroglial activation. All four LG metabolites were found in the retina, although with different proportions with respect to the amount in the dose of administered LG, and with different temporal profiles in the 24 h following administration. These findings are consistent with neuroenhancing and neuroprotective effects of LG in glaucoma that are likely to derive from its powerful antioxidant properties. The co-occurrence of different metabolites in LG may provide an added value to their beneficial effects and indicate LG as a basis for the potential treatment of a variety of retinal pathologies.
Collapse
Affiliation(s)
- Rosario Amato
- Department of Biology, University of Pisa, Pisa, Italy
| | | | - Maurizio Cammalleri
- Department of Biology, University of Pisa, Pisa, Italy.,Interdepartmental Research Center Nutrafood "Nutraceuticals and Food for Health", University of Pisa, Pisa, Italy
| | - Anna Maria Timperio
- Department of Ecological and Biological Sciences, University of Tuscia, Viterbo, Italy
| | - Giuseppina Fanelli
- Department of Agriculture and Forest Sciences, University of Tuscia, Viterbo, Italy
| | - Massimo Dal Monte
- Department of Biology, University of Pisa, Pisa, Italy.,Interdepartmental Research Center Nutrafood "Nutraceuticals and Food for Health", University of Pisa, Pisa, Italy
| | - Laura Pucci
- National Research Council, Institute of Agricultural Biology and Biotechnology (IBBA), Pisa, Italy
| | - Giovanni Casini
- Department of Biology, University of Pisa, Pisa, Italy.,Interdepartmental Research Center Nutrafood "Nutraceuticals and Food for Health", University of Pisa, Pisa, Italy
| |
Collapse
|
228
|
Hamada K, Shinozaki Y, Namekata K, Matsumoto M, Ohno N, Segawa T, Kashiwagi K, Harada T, Koizumi S. Loss of P2Y 1 receptors triggers glaucoma-like pathology in mice. Br J Pharmacol 2021; 178:4552-4571. [PMID: 34309010 DOI: 10.1111/bph.15637] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 06/22/2021] [Accepted: 07/16/2021] [Indexed: 11/30/2022] Open
Abstract
BACKGROUND AND PURPOSE Glaucoma, the leading cause of blindness, damages the retinal ganglion cells. Elevated intraocular pressure (IOP) is a high-risk factor for glaucoma, so topical hypotensive drugs are usually used for treatment. Because not all patients do not respond adequately to current treatments, there is a need to identify a new molecular target to reduce IOP. Here, we have assessed the role of P2Y1 receptors in mediating elevated IOP. EXPERIMENTAL APPROACH P2Y1 receptor agonist was instilled into the eyes of mice, and the IOP changes were measured by a rebound-type tonometer. Expression of P2Y1 receptors was estimated by immunohistochemistry. Ocular function was measured by a multifocal electroretinogram. KEY RESULTS A single dose of the P2Y1 receptor agonist transiently reduced IOP and such effects were absent in P2Y1 receptor-deficient (P2Y1 KO) mice. P2Y1 receptors were functionally expressed in the ciliary body, trabecular meshwork and Schlemm's canal. Activation of P2Y1 receptors negatively regulated aquaporin 4 (AQP4) function but up-regulated endothelial NOS (eNOS). P2Y1 KO mice showed chronic ocular hypertension regardless of age. P2Y1 KO mice at 3 months old showed no damage to retinal ganglion cells, whereas 12-month-old mice showed a significant loss of these cells and impairment of ocular functions. Damage to retinal ganglion cells was attenuated by chronic administration of an IOP-reducing agent. CONCLUSION AND IMPLICATIONS Activation of P2Y1 receptors reduced IOP via dual pathways including AQP4 and eNOS. Loss of P2Y1 receptors resulted in glaucomatous optic neuropathy, suggesting that P2Y1 receptors might provide an effective target in the treatment of glaucoma.
Collapse
Affiliation(s)
- Kentaro Hamada
- Department of Neuropharmacology, Interdisciplinary Graduate School of Medicine, University of Yamanashi, Yamanashi, Japan
| | - Youichi Shinozaki
- Department of Neuropharmacology, Interdisciplinary Graduate School of Medicine, University of Yamanashi, Yamanashi, Japan.,GLIA Center, Interdisciplinary Graduate School of Medicine, University of Yamanashi, Yamanashi, Japan
| | - Kazuhiko Namekata
- Visual Research Project, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| | - Mami Matsumoto
- Division of Ultrastructural Research, National Institute of Physiological Sciences, Aichi, Japan
| | - Nobuhiko Ohno
- Division of Ultrastructural Research, National Institute of Physiological Sciences, Aichi, Japan.,Department of Anatomy, Jichi Medical University, Tochigi, Japan
| | - Takahiro Segawa
- Center for Life Science Research, Interdisciplinary Graduate School of Medicine, University of Yamanashi, Yamanashi, Japan
| | - Kenji Kashiwagi
- Department of Ophthalmology, Interdisciplinary Graduate School of Medicine, University of Yamanashi, Yamanashi, Japan
| | - Takayuki Harada
- Visual Research Project, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| | - Schuichi Koizumi
- Department of Neuropharmacology, Interdisciplinary Graduate School of Medicine, University of Yamanashi, Yamanashi, Japan.,GLIA Center, Interdisciplinary Graduate School of Medicine, University of Yamanashi, Yamanashi, Japan
| |
Collapse
|
229
|
Ye Z, Li X, Zheng D, Pei S, Cheng P, Zhang L, Zhu L. Intravitreally Injected Methylene Blue Protects Retina against Acute Ocular Hypertension in Rats. Curr Eye Res 2021; 47:91-101. [PMID: 34165383 DOI: 10.1080/02713683.2021.1948062] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
Purpose: To assess the neuroprotective effects of methylene blue (MB) in a rat model of acute ocular hypertension (AOH) and explore its possible mechanisms.Methods: Our AOH rat model was obtained with anterior chamber perfusion for 60 min. After that, 100 μM MB was injected into the vitreous cavity immediately after injury. Electroretinogram, fundus photography, optical coherence tomography (OCT) and retina morphology examination were utilized to quantify retinal damage before surgery, as well as 7, 14 and 28 days after. The average number of surviving retinal ganglion cells (RGCs) was counted after fluorescent retrograde labelling with 4% DiI. And TUNEL assay was used to investigate retinal cell apoptosis at 24 hours after AOH. Nrf2 and BACE1 in the retina were determined by RT-qPCR analysis.Results: AOH did produce a severe degeneration effect on the whole retinal layer. Intravitreally injected MB maintained certain retinal thickness after AOH, reduced the destruction of electroretinograms, and enhanced RGCs survival. The average number of TUNEL-labelled cells statistically reduced in the MB-treated retina tissue compared with retina treated with normal saline. The relative mRNA level of Nrf2 was also much higher in the MB-treated retinas after AOH, and the expression of BACE1 had a decline in the AOH + MB group.Conclusions: MB can protect the retina from AOH injury and the possible mechanism might involve the inhibition of BACE1 expression and the activation of Nrf2 antioxidant pathway.
Collapse
Affiliation(s)
- Zhiqiang Ye
- Institute of Advanced Materials for Nano-Bio Applications, School of Ophthalmology and Optometry, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Xiaoli Li
- Institute of Advanced Materials for Nano-Bio Applications, School of Ophthalmology and Optometry, Wenzhou Medical University, Wenzhou, Zhejiang, China.,Henan Provincial People's Hospital and People's Hospital of Zhengzhou University, Henan Eye Institute, Henan Eye Hospital, Zhengzhou, Henan, China
| | - Dongliang Zheng
- Institute of Advanced Materials for Nano-Bio Applications, School of Ophthalmology and Optometry, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Shuaili Pei
- Institute of Advanced Materials for Nano-Bio Applications, School of Ophthalmology and Optometry, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Pei Cheng
- Institute of Advanced Materials for Nano-Bio Applications, School of Ophthalmology and Optometry, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Lishu Zhang
- Institute of Advanced Materials for Nano-Bio Applications, School of Ophthalmology and Optometry, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Lin Zhu
- Institute of Advanced Materials for Nano-Bio Applications, School of Ophthalmology and Optometry, Wenzhou Medical University, Wenzhou, Zhejiang, China
| |
Collapse
|
230
|
Jassim AH, Inman DM, Mitchell CH. Crosstalk Between Dysfunctional Mitochondria and Inflammation in Glaucomatous Neurodegeneration. Front Pharmacol 2021; 12:699623. [PMID: 34366851 PMCID: PMC8334009 DOI: 10.3389/fphar.2021.699623] [Citation(s) in RCA: 67] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Accepted: 06/22/2021] [Indexed: 12/11/2022] Open
Abstract
Mitochondrial dysfunction and excessive inflammatory responses are both sufficient to induce pathology in age-dependent neurodegenerations. However, emerging evidence indicates crosstalk between damaged mitochondrial and inflammatory signaling can exacerbate issues in chronic neurodegenerations. This review discusses evidence for the interaction between mitochondrial damage and inflammation, with a focus on glaucomatous neurodegeneration, and proposes that positive feedback resulting from this crosstalk drives pathology. Mitochondrial dysfunction exacerbates inflammatory signaling in multiple ways. Damaged mitochondrial DNA is a damage-associated molecular pattern, which activates the NLRP3 inflammasome; priming and activation of the NLRP3 inflammasome, and the resulting liberation of IL-1β and IL-18 via the gasdermin D pore, is a major pathway to enhance inflammatory responses. The rise in reactive oxygen species induced by mitochondrial damage also activates inflammatory pathways, while blockage of Complex enzymes is sufficient to increase inflammatory signaling. Impaired mitophagy contributes to inflammation as the inability to turnover mitochondria in a timely manner increases levels of ROS and damaged mtDNA, with the latter likely to stimulate the cGAS-STING pathway to increase interferon signaling. Mitochondrial associated ER membrane contacts and the mitochondria-associated adaptor molecule MAVS can activate NLRP3 inflammasome signaling. In addition to dysfunctional mitochondria increasing inflammation, the corollary also occurs, with inflammation reducing mitochondrial function and ATP production; the resulting downward spiral accelerates degeneration. Evidence from several preclinical models including the DBA/2J mouse, microbead injection and transient elevation of IOP, in addition to patient data, implicates both mitochondrial damage and inflammation in glaucomatous neurodegeneration. The pressure-dependent hypoxia and the resulting metabolic vulnerability is associated with mitochondrial damage and IL-1β release. Links between mitochondrial dysfunction and inflammation can occur in retinal ganglion cells, microglia cells and astrocytes. In summary, crosstalk between damaged mitochondria and increased inflammatory signaling enhances pathology in glaucomatous neurodegeneration, with implications for other complex age-dependent neurodegenerations like Alzheimer's and Parkinson's disease.
Collapse
Affiliation(s)
- Assraa Hassan Jassim
- Department of Basic and Translational Science, University of Pennsylvania, Philadelphia, PA, United States
| | - Denise M. Inman
- Department of Pharmaceutical Sciences, North Texas Eye Research Institute, University of North Texas Health Science Center, Fort Worth, TX, United States
| | - Claire H. Mitchell
- Department of Basic and Translational Science, University of Pennsylvania, Philadelphia, PA, United States
- Department of Ophthalmology, University of Pennsylvania, Philadelphia, PA, United States
- Department of Physiology, University of Pennsylvania, Philadelphia, PA, United States
| |
Collapse
|
231
|
Hopkins EL, Gu W, Kobe B, Coleman MP. A Novel NAD Signaling Mechanism in Axon Degeneration and its Relationship to Innate Immunity. Front Mol Biosci 2021; 8:703532. [PMID: 34307460 PMCID: PMC8295901 DOI: 10.3389/fmolb.2021.703532] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Accepted: 06/28/2021] [Indexed: 12/21/2022] Open
Abstract
Axon degeneration represents a pathological feature of many neurodegenerative diseases, including Alzheimer's disease and Parkinson's disease where axons die before the neuronal soma, and axonopathies, such as Charcot-Marie-Tooth disease and hereditary spastic paraplegia. Over the last two decades, it has slowly emerged that a central signaling pathway forms the basis of this process in many circumstances. This is an axonal NAD-related signaling mechanism mainly regulated by the two key proteins with opposing roles: the NAD-synthesizing enzyme NMNAT2, and SARM1, a protein with NADase and related activities. The crosstalk between the axon survival factor NMNAT2 and pro-degenerative factor SARM1 has been extensively characterized and plays an essential role in maintaining the axon integrity. This pathway can be activated in necroptosis and in genetic, toxic or metabolic disorders, physical injury and neuroinflammation, all leading to axon pathology. SARM1 is also known to be involved in regulating innate immunity, potentially linking axon degeneration to the response to pathogens and intercellular signaling. Understanding this NAD-related signaling mechanism enhances our understanding of the process of axon degeneration and enables a path to the development of drugs for a wide range of neurodegenerative diseases.
Collapse
Affiliation(s)
- Eleanor L. Hopkins
- John van Geest Centre for Brain Repair, Department of Clinical Neurosciences, University of Cambridge, Cambridge, United Kingdom
| | - Weixi Gu
- School of Chemistry and Molecular Biosciences, Institute for Molecular Bioscience and Australian Infectious Diseases Research Centre, The University of Queensland, Brisbane, QLD, Australia
| | - Bostjan Kobe
- School of Chemistry and Molecular Biosciences, Institute for Molecular Bioscience and Australian Infectious Diseases Research Centre, The University of Queensland, Brisbane, QLD, Australia
| | - Michael P. Coleman
- John van Geest Centre for Brain Repair, Department of Clinical Neurosciences, University of Cambridge, Cambridge, United Kingdom
| |
Collapse
|
232
|
Lenaers G, Neutzner A, Le Dantec Y, Jüschke C, Xiao T, Decembrini S, Swirski S, Kieninger S, Agca C, Kim US, Reynier P, Yu-Wai-Man P, Neidhardt J, Wissinger B. Dominant optic atrophy: Culprit mitochondria in the optic nerve. Prog Retin Eye Res 2021; 83:100935. [PMID: 33340656 DOI: 10.1016/j.preteyeres.2020.100935] [Citation(s) in RCA: 60] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Revised: 12/05/2020] [Accepted: 12/09/2020] [Indexed: 12/14/2022]
Abstract
Dominant optic atrophy (DOA) is an inherited mitochondrial disease leading to specific degeneration of retinal ganglion cells (RGCs), thus compromising transmission of visual information from the retina to the brain. Usually, DOA starts during childhood and evolves to poor vision or legal blindness, affecting the central vision, whilst sparing the peripheral visual field. In 20% of cases, DOA presents as syndromic disorder, with secondary symptoms affecting neuronal and muscular functions. Twenty years ago, we demonstrated that heterozygous mutations in OPA1 are the most frequent molecular cause of DOA. Since then, variants in additional genes, whose functions in many instances converge with those of OPA1, have been identified by next generation sequencing. OPA1 encodes a dynamin-related GTPase imported into mitochondria and located to the inner membrane and intermembrane space. The many OPA1 isoforms, resulting from alternative splicing of three exons, form complex homopolymers that structure mitochondrial cristae, and contribute to fusion of the outer membrane, thus shaping the whole mitochondrial network. Moreover, OPA1 is required for oxidative phosphorylation, maintenance of mitochondrial genome, calcium homeostasis and regulation of apoptosis, thus making OPA1 the Swiss army-knife of mitochondria. Understanding DOA pathophysiology requires the understanding of RGC peculiarities with respect to OPA1 functions. Besides the tremendous energy requirements of RGCs to relay visual information from the eye to the brain, these neurons present unique features related to their differential environments in the retina, and to the anatomical transition occurring at the lamina cribrosa, which parallel major adaptations of mitochondrial physiology and shape, in the pre- and post-laminar segments of the optic nerve. Three DOA mouse models, with different Opa1 mutations, have been generated to study intrinsic mechanisms responsible for RGC degeneration, and these have further revealed secondary symptoms related to mitochondrial dysfunctions, mirroring the more severe syndromic phenotypes seen in a subgroup of patients. Metabolomics analyses of cells, mouse organs and patient plasma mutated for OPA1 revealed new unexpected pathophysiological mechanisms related to mitochondrial dysfunction, and biomarkers correlated quantitatively to the severity of the disease. Here, we review and synthesize these data, and propose different approaches for embracing possible therapies to fulfil the unmet clinical needs of this disease, and provide hope to affected DOA patients.
Collapse
Affiliation(s)
- Guy Lenaers
- MitoLab Team, UMR CNRS 6015 - INSERM U1083, Institut MitoVasc, Angers University and Hospital, Angers, France.
| | - Albert Neutzner
- Department of Biomedicine, University Hospital Basel, University of Basel, Basel, Switzerland; Department of Ophthalmology University Hospital Basel, University of Basel, Basel, Switzerland.
| | - Yannick Le Dantec
- MitoLab Team, UMR CNRS 6015 - INSERM U1083, Institut MitoVasc, Angers University and Hospital, Angers, France
| | - Christoph Jüschke
- Human Genetics, Faculty VI - School of Medicine and Health Sciences, University of Oldenburg, Oldenburg, Germany
| | - Ting Xiao
- Molecular Genetics Laboratory, Institute for Ophthalmic Research, Center for Ophthalmology, University of Tübingen, Tübingen, Germany
| | - Sarah Decembrini
- Department of Biomedicine, University Hospital Basel, University of Basel, Basel, Switzerland; Department of Ophthalmology University Hospital Basel, University of Basel, Basel, Switzerland
| | - Sebastian Swirski
- Human Genetics, Faculty VI - School of Medicine and Health Sciences, University of Oldenburg, Oldenburg, Germany
| | - Sinja Kieninger
- Molecular Genetics Laboratory, Institute for Ophthalmic Research, Center for Ophthalmology, University of Tübingen, Tübingen, Germany
| | - Cavit Agca
- Molecular Biology, Genetics and Bioengineering Program, Sabanci University, Istanbul, Turkey; Nanotechnology Research and Application Center (SUNUM), Sabanci University, Istanbul, Turkey
| | - Ungsoo S Kim
- Kim's Eye Hospital, Seoul, South Korea; Cambridge Centre for Brain Repair and MRC Mitochondrial Biology Unit, Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK; Cambridge Eye Unit, Addenbrooke's Hospital, Cambridge University Hospitals, Cambridge, UK; Moorfields Eye Hospital, London, UK
| | - Pascal Reynier
- MitoLab Team, UMR CNRS 6015 - INSERM U1083, Institut MitoVasc, Angers University and Hospital, Angers, France; Department of Biochemistry, University Hospital of Angers, Angers, France
| | - Patrick Yu-Wai-Man
- Cambridge Centre for Brain Repair and MRC Mitochondrial Biology Unit, Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK; Cambridge Eye Unit, Addenbrooke's Hospital, Cambridge University Hospitals, Cambridge, UK; Moorfields Eye Hospital, London, UK; UCL Institute of Ophthalmology, University College London, London, UK
| | - John Neidhardt
- Human Genetics, Faculty VI - School of Medicine and Health Sciences, University of Oldenburg, Oldenburg, Germany; Research Center Neurosensory Science, University Oldenburg, Oldenburg, Germany.
| | - Bernd Wissinger
- Molecular Genetics Laboratory, Institute for Ophthalmic Research, Center for Ophthalmology, University of Tübingen, Tübingen, Germany.
| |
Collapse
|
233
|
Belforte N, Agostinone J, Alarcon-Martinez L, Villafranca-Baughman D, Dotigny F, Cueva Vargas JL, Di Polo A. AMPK hyperactivation promotes dendrite retraction, synaptic loss, and neuronal dysfunction in glaucoma. Mol Neurodegener 2021; 16:43. [PMID: 34187514 PMCID: PMC8243567 DOI: 10.1186/s13024-021-00466-z] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Accepted: 06/09/2021] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND The maintenance of complex dendritic arbors and synaptic transmission are processes that require a substantial amount of energy. Bioenergetic decline is a prominent feature of chronic neurodegenerative diseases, yet the signaling mechanisms that link energy stress with neuronal dysfunction are poorly understood. Recent work has implicated energy deficits in glaucoma, and retinal ganglion cell (RGC) dendritic pathology and synapse disassembly are key features of ocular hypertension damage. RESULTS We show that adenosine monophosphate-activated protein kinase (AMPK), a conserved energy biosensor, is strongly activated in RGC from mice with ocular hypertension and patients with primary open angle glaucoma. Our data demonstrate that AMPK triggers RGC dendrite retraction and synapse elimination. We show that the harmful effect of AMPK is exerted through inhibition of the mammalian target of rapamycin complex 1 (mTORC1). Attenuation of AMPK activity restores mTORC1 function and rescues dendrites and synaptic contacts. Strikingly, AMPK depletion promotes recovery of light-evoked retinal responses, improves axonal transport, and extends RGC survival. CONCLUSIONS This study identifies AMPK as a critical nexus between bioenergetic decline and RGC dysfunction during pressure-induced stress, and highlights the importance of targeting energy homeostasis in glaucoma and other neurodegenerative diseases.
Collapse
Affiliation(s)
- Nicolas Belforte
- Department of Neuroscience, Université de Montréal, Succursale centre-ville 6128, Montréal, Québec, H3C 3J7, Canada.,Centre de recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), 900 Saint Denis Street, Montréal, Québec, H2X 0A9, Canada
| | - Jessica Agostinone
- Department of Neuroscience, Université de Montréal, Succursale centre-ville 6128, Montréal, Québec, H3C 3J7, Canada.,Centre de recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), 900 Saint Denis Street, Montréal, Québec, H2X 0A9, Canada
| | - Luis Alarcon-Martinez
- Department of Neuroscience, Université de Montréal, Succursale centre-ville 6128, Montréal, Québec, H3C 3J7, Canada.,Centre de recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), 900 Saint Denis Street, Montréal, Québec, H2X 0A9, Canada
| | - Deborah Villafranca-Baughman
- Department of Neuroscience, Université de Montréal, Succursale centre-ville 6128, Montréal, Québec, H3C 3J7, Canada.,Centre de recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), 900 Saint Denis Street, Montréal, Québec, H2X 0A9, Canada
| | - Florence Dotigny
- Centre de recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), 900 Saint Denis Street, Montréal, Québec, H2X 0A9, Canada
| | - Jorge L Cueva Vargas
- Department of Neuroscience, Université de Montréal, Succursale centre-ville 6128, Montréal, Québec, H3C 3J7, Canada.,Centre de recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), 900 Saint Denis Street, Montréal, Québec, H2X 0A9, Canada
| | - Adriana Di Polo
- Department of Neuroscience, Université de Montréal, Succursale centre-ville 6128, Montréal, Québec, H3C 3J7, Canada. .,Centre de recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), 900 Saint Denis Street, Montréal, Québec, H2X 0A9, Canada.
| |
Collapse
|
234
|
Muench NA, Patel S, Maes ME, Donahue RJ, Ikeda A, Nickells RW. The Influence of Mitochondrial Dynamics and Function on Retinal Ganglion Cell Susceptibility in Optic Nerve Disease. Cells 2021; 10:cells10071593. [PMID: 34201955 PMCID: PMC8306483 DOI: 10.3390/cells10071593] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2021] [Revised: 06/15/2021] [Accepted: 06/17/2021] [Indexed: 12/30/2022] Open
Abstract
The important roles of mitochondrial function and dysfunction in the process of neurodegeneration are widely acknowledged. Retinal ganglion cells (RGCs) appear to be a highly vulnerable neuronal cell type in the central nervous system with respect to mitochondrial dysfunction but the actual reasons for this are still incompletely understood. These cells have a unique circumstance where unmyelinated axons must bend nearly 90° to exit the eye and then cross a translaminar pressure gradient before becoming myelinated in the optic nerve. This region, the optic nerve head, contains some of the highest density of mitochondria present in these cells. Glaucoma represents a perfect storm of events occurring at this location, with a combination of changes in the translaminar pressure gradient and reassignment of the metabolic support functions of supporting glia, which appears to apply increased metabolic stress to the RGC axons leading to a failure of axonal transport mechanisms. However, RGCs themselves are also extremely sensitive to genetic mutations, particularly in genes affecting mitochondrial dynamics and mitochondrial clearance. These mutations, which systemically affect the mitochondria in every cell, often lead to an optic neuropathy as the sole pathologic defect in affected patients. This review summarizes knowledge of mitochondrial structure and function, the known energy demands of neurons in general, and places these in the context of normal and pathological characteristics of mitochondria attributed to RGCs.
Collapse
Affiliation(s)
- Nicole A. Muench
- Department of Ophthalmology and Visual Sciences, University of Wisconsin-Madison, Madison, WI 53706, USA; (N.A.M.); (S.P.); (R.J.D.)
| | - Sonia Patel
- Department of Ophthalmology and Visual Sciences, University of Wisconsin-Madison, Madison, WI 53706, USA; (N.A.M.); (S.P.); (R.J.D.)
| | - Margaret E. Maes
- Institute of Science and Technology Austria, 3400 Klosterneuburg, Austria;
| | - Ryan J. Donahue
- Department of Ophthalmology and Visual Sciences, University of Wisconsin-Madison, Madison, WI 53706, USA; (N.A.M.); (S.P.); (R.J.D.)
- Boston Children’s Hospital, Harvard Medical School, Harvard University, Boston, MA 02115, USA
| | - Akihiro Ikeda
- Department of Medical Genetics, University of Wisconsin-Madison, Madison, WI 53706, USA;
- McPherson Eye Research Institute, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Robert W. Nickells
- Department of Ophthalmology and Visual Sciences, University of Wisconsin-Madison, Madison, WI 53706, USA; (N.A.M.); (S.P.); (R.J.D.)
- McPherson Eye Research Institute, University of Wisconsin-Madison, Madison, WI 53705, USA
- Correspondence:
| |
Collapse
|
235
|
Wang X, Lou N, Eberhardt A, Yang Y, Kusk P, Xu Q, Förstera B, Peng S, Shi M, Ladrón-de-Guevara A, Delle C, Sigurdsson B, Xavier ALR, Ertürk A, Libby RT, Chen L, Thrane AS, Nedergaard M. An ocular glymphatic clearance system removes β-amyloid from the rodent eye. Sci Transl Med 2021; 12:12/536/eaaw3210. [PMID: 32213628 DOI: 10.1126/scitranslmed.aaw3210] [Citation(s) in RCA: 152] [Impact Index Per Article: 38.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2018] [Revised: 08/24/2019] [Accepted: 12/30/2019] [Indexed: 12/31/2022]
Abstract
Despite high metabolic activity, the retina and optic nerve head lack traditional lymphatic drainage. We here identified an ocular glymphatic clearance route for fluid and wastes via the proximal optic nerve in rodents. β-amyloid (Aβ) was cleared from the retina and vitreous via a pathway dependent on glial water channel aquaporin-4 (AQP4) and driven by the ocular-cranial pressure difference. After traversing the lamina barrier, intra-axonal Aβ was cleared via the perivenous space and subsequently drained to lymphatic vessels. Light-induced pupil constriction enhanced efflux, whereas atropine or raising intracranial pressure blocked efflux. In two distinct murine models of glaucoma, Aβ leaked from the eye via defects in the lamina barrier instead of directional axonal efflux. The results suggest that, in rodents, the removal of fluid and metabolites from the intraocular space occurs through a glymphatic pathway that might be impaired in glaucoma.
Collapse
Affiliation(s)
- Xiaowei Wang
- Center for Translational Neuromedicine, Faculty of Medical and Health Sciences, University of Copenhagen, Blegdamsvej 3B, 2200 Copenhagen N, Denmark.,Center for Translational Neuromedicine, University of Rochester Medical School, Elmwood Avenue 601, Rochester, NY 14642, USA
| | - Nanhong Lou
- Center for Translational Neuromedicine, University of Rochester Medical School, Elmwood Avenue 601, Rochester, NY 14642, USA
| | - Allison Eberhardt
- Center for Translational Neuromedicine, University of Rochester Medical School, Elmwood Avenue 601, Rochester, NY 14642, USA
| | - Yujia Yang
- Center for Eye Disease and Development, Vision Science Graduate Program, and School of Optometry, University of California Berkeley, Berkeley, CA 94720, USA
| | - Peter Kusk
- Center for Translational Neuromedicine, Faculty of Medical and Health Sciences, University of Copenhagen, Blegdamsvej 3B, 2200 Copenhagen N, Denmark
| | - Qiwu Xu
- Center for Translational Neuromedicine, University of Rochester Medical School, Elmwood Avenue 601, Rochester, NY 14642, USA
| | - Benjamin Förstera
- Institute for Stroke and Dementia Research, Klinikum der Universität München, Ludwig Maximilians University of Munich (LMU), 81377 Munich, Germany.,Institute for Tissue Engineering and Regenerative Medicine (iTERM), Helmholtz Center München, 85764 Munich, Germany
| | - Sisi Peng
- Center for Translational Neuromedicine, University of Rochester Medical School, Elmwood Avenue 601, Rochester, NY 14642, USA
| | - Meng Shi
- Center for Eye Disease and Development, Vision Science Graduate Program, and School of Optometry, University of California Berkeley, Berkeley, CA 94720, USA
| | - Antonio Ladrón-de-Guevara
- Center for Translational Neuromedicine, University of Rochester Medical School, Elmwood Avenue 601, Rochester, NY 14642, USA
| | - Christine Delle
- Center for Translational Neuromedicine, Faculty of Medical and Health Sciences, University of Copenhagen, Blegdamsvej 3B, 2200 Copenhagen N, Denmark
| | - Björn Sigurdsson
- Center for Translational Neuromedicine, Faculty of Medical and Health Sciences, University of Copenhagen, Blegdamsvej 3B, 2200 Copenhagen N, Denmark
| | - Anna L R Xavier
- Center for Translational Neuromedicine, Faculty of Medical and Health Sciences, University of Copenhagen, Blegdamsvej 3B, 2200 Copenhagen N, Denmark
| | - Ali Ertürk
- Institute for Stroke and Dementia Research, Klinikum der Universität München, Ludwig Maximilians University of Munich (LMU), 81377 Munich, Germany.,Institute for Tissue Engineering and Regenerative Medicine (iTERM), Helmholtz Center München, 85764 Munich, Germany
| | - Richard T Libby
- Department of Ophthalmology, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Lu Chen
- Center for Eye Disease and Development, Vision Science Graduate Program, and School of Optometry, University of California Berkeley, Berkeley, CA 94720, USA.
| | - Alexander S Thrane
- Center for Translational Neuromedicine, Faculty of Medical and Health Sciences, University of Copenhagen, Blegdamsvej 3B, 2200 Copenhagen N, Denmark.,Department of Ophthalmology, Haukeland University Hospital, Jonas Lies Vei 65, 5021 Bergen, Norway
| | - Maiken Nedergaard
- Center for Translational Neuromedicine, Faculty of Medical and Health Sciences, University of Copenhagen, Blegdamsvej 3B, 2200 Copenhagen N, Denmark. .,Center for Translational Neuromedicine, University of Rochester Medical School, Elmwood Avenue 601, Rochester, NY 14642, USA
| |
Collapse
|
236
|
Carrella S, Massa F, Indrieri A. The Role of MicroRNAs in Mitochondria-Mediated Eye Diseases. Front Cell Dev Biol 2021; 9:653522. [PMID: 34222230 PMCID: PMC8249810 DOI: 10.3389/fcell.2021.653522] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Accepted: 05/20/2021] [Indexed: 12/15/2022] Open
Abstract
The retina is among the most metabolically active tissues with high-energy demands. The peculiar distribution of mitochondria in cells of retinal layers is necessary to assure the appropriate energy supply for the transmission of the light signal. Photoreceptor cells (PRs), retinal pigment epithelium (RPE), and retinal ganglion cells (RGCs) present a great concentration of mitochondria, which makes them particularly sensitive to mitochondrial dysfunction. To date, visual loss has been extensively correlated to defective mitochondrial functions. Many mitochondrial diseases (MDs) show indeed neuro-ophthalmic manifestations, including retinal and optic nerve phenotypes. Moreover, abnormal mitochondrial functions are frequently found in the most common retinal pathologies, i.e., glaucoma, age-related macular degeneration (AMD), and diabetic retinopathy (DR), that share clinical similarities with the hereditary primary MDs. MicroRNAs (miRNAs) are established as key regulators of several developmental, physiological, and pathological processes. Dysregulated miRNA expression profiles in retinal degeneration models and in patients underline the potentiality of miRNA modulation as a possible gene/mutation-independent strategy in retinal diseases and highlight their promising role as disease predictive or prognostic biomarkers. In this review, we will summarize the current knowledge about the participation of miRNAs in both rare and common mitochondria-mediated eye diseases. Definitely, given the involvement of miRNAs in retina pathologies and therapy as well as their use as molecular biomarkers, they represent a determining target for clinical applications.
Collapse
Affiliation(s)
| | - Filomena Massa
- Telethon Institute of Genetics and Medicine, Naples, Italy
| | - Alessia Indrieri
- Telethon Institute of Genetics and Medicine, Naples, Italy.,Institute for Genetic and Biomedical Research, National Research Council (CNR), Milan, Italy
| |
Collapse
|
237
|
Systemic Treatment with Nicotinamide Riboside Is Protective in Two Mouse Models of Retinal Ganglion Cell Damage. Pharmaceutics 2021; 13:pharmaceutics13060893. [PMID: 34208613 PMCID: PMC8235058 DOI: 10.3390/pharmaceutics13060893] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Revised: 05/20/2021] [Accepted: 06/10/2021] [Indexed: 11/26/2022] Open
Abstract
Glaucoma etiology often includes retinal ganglion cell (RGC) death associated with elevated intraocular pressure (IOP). However, even when IOP is managed well, disease can progress. It is thus important to develop therapeutic approaches that directly protect RGCs in an IOP-independent manner. Compromised nicotinamide adenine dinucleotide (NAD+) metabolism occurs in neurodegenerative diseases, including models of glaucoma. Here we report testing the protective effects of prophylactically systemically administered nicotinamide riboside (NR), a NAD+ precursor, in a mouse model of acute RGC damage (optic nerve crush (ONC)), and in a chronic model of RGC degeneration (ocular hypertension induced by intracameral injection of microbeads). For both models, treatment enhanced RGC survival, assessed by counting cells in retinal flatmounts immunostained for Brn3a+. In the ONC model, treatment preserved RGC function, as assessed by pattern electroretinogram, and suppressed retinal inflammation, as assessed by immunofluorescence staining of retinal fixed sections for glial fibrillary acidic protein (GFAP). This is the first study to demonstrate that systemic treatment with NR is protective in acute and chronic models of RGC damage. The protection is significant and, considering that NR is highly bioavailable in and well-tolerated by humans, may support the proposition of prospective human subject studies.
Collapse
|
238
|
Abstract
The outer retina is nourished from the choroid, a capillary bed just inside the sclera. O2, glucose, and other nutrients diffuse out of the choroid and then filter through a monolayer of retinal pigment epithelium (RPE) cells to fuel the retina. Recent studies of energy metabolism have revealed striking differences between retinas and RPE cells in the ways that they extract energy from fuels. The purpose of this review is to suggest and evaluate the hypothesis that the retina and RPE have complementary metabolic roles that make them depend on each other for survival and for their abilities to perform essential and specialized functions. Expected final online publication date for the Annual Review of Vision Science, Volume 7 is September 2021. Please see http://www.annualreviews.org/page/journal/pubdates for revised estimates.
Collapse
Affiliation(s)
- James B Hurley
- Departments of Biochemistry and Ophthalmology, University of Washington, Seattle, Washington 98115, USA;
| |
Collapse
|
239
|
Neuroprotection in Glaucoma: NAD +/NADH Redox State as a Potential Biomarker and Therapeutic Target. Cells 2021; 10:cells10061402. [PMID: 34198948 PMCID: PMC8226607 DOI: 10.3390/cells10061402] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 05/24/2021] [Accepted: 05/27/2021] [Indexed: 02/07/2023] Open
Abstract
Glaucoma is the leading cause of irreversible blindness worldwide. Its prevalence and incidence increase exponentially with age and the level of intraocular pressure (IOP). IOP reduction is currently the only therapeutic modality shown to slow glaucoma progression. However, patients still lose vision despite best treatment, suggesting that other factors confer susceptibility. Several studies indicate that mitochondrial function may underlie both susceptibility and resistance to developing glaucoma. Mitochondria meet high energy demand, in the form of ATP, that is required for the maintenance of optimum retinal ganglion cell (RGC) function. Reduced nicotinamide adenine dinucleotide (NAD+) levels have been closely correlated to mitochondrial dysfunction and have been implicated in several neurodegenerative diseases including glaucoma. NAD+ is at the centre of various metabolic reactions culminating in ATP production—essential for RGC function. In this review we present various pathways that influence the NAD+(H) redox state, affecting mitochondrial function and making RGCs susceptible to degeneration. Such disruptions of the NAD+(H) redox state are generalised and not solely induced in RGCs because of high IOP. This places the NAD+(H) redox state as a potential systemic biomarker for glaucoma susceptibility and progression; a hypothesis which may be tested in clinical trials and then translated to clinical practice.
Collapse
|
240
|
Modeling Retinal Ganglion Cell Dysfunction in Optic Neuropathies. Cells 2021; 10:cells10061398. [PMID: 34198840 PMCID: PMC8227951 DOI: 10.3390/cells10061398] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Revised: 05/24/2021] [Accepted: 06/03/2021] [Indexed: 12/13/2022] Open
Abstract
As in glaucoma and other optic neuropathies cellular dysfunction often precedes cell death, the assessment of retinal ganglion cell (RGC) function represents a key outcome measure for neuroprotective strategies aimed at targeting distressed but still viable cells. RGC dysfunction can be assessed with the pattern electroretinogram (PERG), a sensitive measure of electrical activity of RGCs that is recorded non-invasively in human subjects and mouse models. Here, we offer a conceptual framework based on an intuitive state-transition model used for disease management in patients to identify progressive, potentially reversible stages of RGC dysfunction leading to cell death in mouse models of glaucoma and other optic neuropathies. We provide mathematical equations to describe state-transitions with a set of modifiable parameters that alter the time course and severity of state-transitions, which can be used for hypothesis testing and fitting experimental PERG data. PERG dynamics as a function of physiological stimuli are also used to differentiate phenotypic and altered RGC response dynamics, to assess susceptibility to stressors and to assess reversible dysfunction upon pharmacological treatment.
Collapse
|
241
|
Multifactorial Pathogenic Processes of Retinal Ganglion Cell Degeneration in Glaucoma towards Multi-Target Strategies for Broader Treatment Effects. Cells 2021; 10:cells10061372. [PMID: 34199494 PMCID: PMC8228726 DOI: 10.3390/cells10061372] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Revised: 05/14/2021] [Accepted: 05/29/2021] [Indexed: 02/06/2023] Open
Abstract
Glaucoma is a chronic neurodegenerative disease characterized by apoptosis of retinal ganglion cell (RGC) somas, degeneration of axons, and loss of synapses at dendrites and axon terminals. Glaucomatous neurodegeneration encompasses multiple triggers, multiple cell types, and multiple molecular pathways through the etiological paths with biomechanical, vascular, metabolic, oxidative, and inflammatory components. As much as intrinsic responses of RGCs themselves, divergent responses and intricate interactions of the surrounding glia also play decisive roles for the cell fate. Seen from a broad perspective, multitarget treatment strategies have a compelling pathophysiological basis to more efficiently manipulate multiple pathogenic processes at multiple injury sites in such a multifactorial neurodegenerative disease. Despite distinct molecular programs for somatic and axonal degeneration, mitochondrial dysfunction and glia-driven neuroinflammation present interdependent processes with widespread impacts in the glaucomatous retina and optic nerve. Since dysfunctional mitochondria stimulate inflammatory responses and proinflammatory mediators impair mitochondria, mitochondrial restoration may be immunomodulatory, while anti-inflammatory treatments protect mitochondria. Manipulation of these converging routes may thus allow a unified treatment strategy to protect RGC axons, somas, and synapses. This review presents an overview of recent research advancements with emphasis on potential treatment targets to achieve the best treatment efficacy to preserve visual function in glaucoma.
Collapse
|
242
|
Wang J, Struebing FL, Geisert EE. Commonalities of optic nerve injury and glaucoma-induced neurodegeneration: Insights from transcriptome-wide studies. Exp Eye Res 2021; 207:108571. [PMID: 33844961 PMCID: PMC9890784 DOI: 10.1016/j.exer.2021.108571] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2020] [Revised: 03/28/2021] [Accepted: 04/02/2021] [Indexed: 02/03/2023]
Abstract
Glaucoma is a collection of diseases that lead to an irreversible vision loss due to damage of retinal ganglion cells (RGCs). Although the underlying events leading to RGC death are not fully understood, recent research efforts are beginning to define the genetic changes that play a critical role in the initiation and progression of glaucomatous injury and RGC death. Several genetic and experimental animal models have been developed to mimic glaucomatous neurodegeneration. These models differ in many respects but all result in the loss of RGCs. Assessing transcriptional changes across different models could provide a more complete perspective on the molecular drivers of RGC degeneration. For the past several decades, changes in the retinal transcriptome during neurodegeneration process were defined using microarray methods, RNA sequencing and now single cell RNA sequencing. It is understood that these methods have strengths and weaknesses due to technical differences and variations in the analytical tools used. In this review, we focus on the use of transcriptome-wide expression profiling of the changes occurring as RGCs are lost across different glaucoma models. Commonalities of optic nerve crush and glaucoma-induced neurodegeneration are identified and discussed.
Collapse
Affiliation(s)
- Jiaxing Wang
- Emory Eye Center, Department of Ophthalmology, Emory University, 1365B Clifton Road NE, Atlanta, GA, 30322, USA
| | - Felix L. Struebing
- Center for Neuropathology and Prion Research, Ludwig Maximilian University of Munich, Germany,Department for Translational Brain Research, German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
| | - Eldon E. Geisert
- Emory Eye Center, Department of Ophthalmology, Emory University, 1365B Clifton Road NE, Atlanta, GA, 30322, USA,Corresponding author: (E.E. Geisert)
| |
Collapse
|
243
|
Hubens WHG, Kievit MT, Berendschot TTJM, de Coo IFM, Smeets HJM, Webers CAB, Gorgels TGMF. Plasma GDF-15 concentration is not elevated in open-angle glaucoma. PLoS One 2021; 16:e0252630. [PMID: 34048486 PMCID: PMC8162581 DOI: 10.1371/journal.pone.0252630] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Accepted: 05/10/2021] [Indexed: 12/26/2022] Open
Abstract
Aim Recently, the level of growth differentiation factor 15 (GDF-15) in blood, was proposed as biomarker to detect mitochondrial dysfunction. In the current study, we evaluate this biomarker in open-angle glaucoma (OAG), as there is increasing evidence that mitochondrial dysfunction plays a role in the pathophysiology of this disease. Methods Plasma GDF-15 concentrations were measured with ELISA in 200 OAG patients and 61 age-matched controls (cataract without glaucoma). The OAG patient group consisted of high tension glaucoma (HTG; n = 162) and normal tension glaucoma (NTG; n = 38). Groups were compared using the Kruskal-Wallis nonparametric test with Dunn’s multiple comparison post-hoc correction. GDF-15 concentration was corrected for confounders identified with forward linear regression models. Results Before correcting for confounders, median plasma GDF-15 levels was significantly lower in the combined OAG group (p = 0.04), but not when analysing HTG and NTG patients separately. Forward linear regression analysis showed that age, gender, smoking and systemic hypertension were significant confounders affecting GDF-15 levels. After correction for these confounders, GDF-15 levels in OAG patients were no longer significantly different from controls. Subgroup analysis of the glaucoma patients did not show a correlation between disease severity and plasma GDF-15, but did reveal that for NTG patients, intake of dietary supplements, which potentially improve mitochondrial function, correlated with lower plasma GDF-15. Conclusion The present study suggests that plasma GDF-15 is not suited as biomarker of mitochondrial dysfunction in OAG patients.
Collapse
Affiliation(s)
- Wouter H G Hubens
- University Eye Clinic Maastricht, Maastricht University Medical Center, Maastricht, The Netherlands.,School for Mental Health and Neuroscience, Maastricht University, Maastricht, The Netherlands
| | - Mariëlle T Kievit
- University Eye Clinic Maastricht, Maastricht University Medical Center, Maastricht, The Netherlands.,School for Mental Health and Neuroscience, Maastricht University, Maastricht, The Netherlands
| | - Tos T J M Berendschot
- University Eye Clinic Maastricht, Maastricht University Medical Center, Maastricht, The Netherlands.,Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, The Netherlands
| | - Irenaeus F M de Coo
- Department of Toxicogenomics, Maastricht University, Maastricht, The Netherlands
| | - Hubert J M Smeets
- Department of Toxicogenomics, Maastricht University, Maastricht, The Netherlands
| | - Carroll A B Webers
- University Eye Clinic Maastricht, Maastricht University Medical Center, Maastricht, The Netherlands
| | - Theo G M F Gorgels
- University Eye Clinic Maastricht, Maastricht University Medical Center, Maastricht, The Netherlands
| |
Collapse
|
244
|
Wilkison SJ, Bright CL, Vancini R, Song DJ, Bomze HM, Cartoni R. Local Accumulation of Axonal Mitochondria in the Optic Nerve Glial Lamina Precedes Myelination. Front Neuroanat 2021; 15:678501. [PMID: 34093141 PMCID: PMC8173055 DOI: 10.3389/fnana.2021.678501] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Accepted: 04/26/2021] [Indexed: 11/18/2022] Open
Abstract
Mitochondria are essential for neurons and must be optimally distributed along their axon to fulfill local functions. A high density of mitochondria has been observed in retinal ganglion cell (RGC) axons of an unmyelinated region of the optic nerve, called the glial lamina (GL) in mouse (lamina cribrosa in human). In glaucoma, the world's leading cause of irreversible blindness, the GL is the epicenter of RGC degeneration and is connected to mitochondrial dysfunction. It is generally accepted that the local accumulation of mitochondria in the GL is established due to the higher energy requirement of unmyelinated axons. Here we revisit the connection between mitochondrial positioning and myelin in RGC axons. We show that the high density of mitochondria in the GL is restricted to larger axons and is established before myelination. Thus, contrary to a longstanding belief in the field, the myelination pattern is not responsible for the establishment of the local accumulation of mitochondria in GL axons. Our findings open new research avenues likely critical to understanding the pathophysiology of glaucoma.
Collapse
Affiliation(s)
- Samantha J Wilkison
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Durham, NC, United States.,Department of Ophthalmology, Duke University School of Medicine, Durham, NC, United States
| | - Cora L Bright
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Durham, NC, United States.,Department of Ophthalmology, Duke University School of Medicine, Durham, NC, United States
| | - Ricardo Vancini
- Department of Pathology, Duke University School of Medicine, Durham, NC, United States
| | - Daniel J Song
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Durham, NC, United States.,Department of Ophthalmology, Duke University School of Medicine, Durham, NC, United States
| | - Howard M Bomze
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Durham, NC, United States.,Department of Ophthalmology, Duke University School of Medicine, Durham, NC, United States
| | - Romain Cartoni
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Durham, NC, United States.,Department of Ophthalmology, Duke University School of Medicine, Durham, NC, United States
| |
Collapse
|
245
|
Groth B, Venkatakrishnan P, Lin SJ. NAD + Metabolism, Metabolic Stress, and Infection. Front Mol Biosci 2021; 8:686412. [PMID: 34095234 PMCID: PMC8171187 DOI: 10.3389/fmolb.2021.686412] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Accepted: 05/05/2021] [Indexed: 12/26/2022] Open
Abstract
Nicotinamide adenine dinucleotide (NAD+) is an essential metabolite with wide-ranging and significant roles in the cell. Defects in NAD+ metabolism have been associated with many human disorders; it is therefore an emerging therapeutic target. Moreover, NAD+ metabolism is perturbed during colonization by a variety of pathogens, either due to the molecular mechanisms employed by these infectious agents or by the host immune response they trigger. Three main biosynthetic pathways, including the de novo and salvage pathways, contribute to the production of NAD+ with a high degree of conservation from bacteria to humans. De novo biosynthesis, which begins with l-tryptophan in eukaryotes, is also known as the kynurenine pathway. Intermediates of this pathway have various beneficial and deleterious effects on cellular health in different contexts. For example, dysregulation of this pathway is linked to neurotoxicity and oxidative stress. Activation of the de novo pathway is also implicated in various infections and inflammatory signaling. Given the dynamic flexibility and multiple roles of NAD+ intermediates, it is important to understand the interconnections and cross-regulations of NAD+ precursors and associated signaling pathways to understand how cells regulate NAD+ homeostasis in response to various growth conditions. Although regulation of NAD+ homeostasis remains incompletely understood, studies in the genetically tractable budding yeast Saccharomyces cerevisiae may help provide some molecular basis for how NAD+ homeostasis factors contribute to the maintenance and regulation of cellular function and how they are regulated by various nutritional and stress signals. Here we present a brief overview of recent insights and discoveries made with respect to the relationship between NAD+ metabolism and selected human disorders and infections, with a particular focus on the de novo pathway. We also discuss how studies in budding yeast may help elucidate the regulation of NAD+ homeostasis.
Collapse
Affiliation(s)
- Benjamin Groth
- Department of Microbiology and Molecular Genetics, College of Biological Sciences, University of California, Davis, Davis, CA, United States
| | - Padmaja Venkatakrishnan
- Department of Microbiology and Molecular Genetics, College of Biological Sciences, University of California, Davis, Davis, CA, United States
| | - Su-Ju Lin
- Department of Microbiology and Molecular Genetics, College of Biological Sciences, University of California, Davis, Davis, CA, United States
| |
Collapse
|
246
|
Sun X, Cao B, Naval-Sanchez M, Pham T, Sun YBY, Williams B, Heazlewood SY, Deshpande N, Li J, Kraus F, Rae J, Nguyen Q, Yari H, Schröder J, Heazlewood CK, Fulton M, Hatwell-Humble J, Das Gupta K, Kapetanovic R, Chen X, Sweet MJ, Parton RG, Ryan MT, Polo JM, Nefzger CM, Nilsson SK. Nicotinamide riboside attenuates age-associated metabolic and functional changes in hematopoietic stem cells. Nat Commun 2021; 12:2665. [PMID: 33976125 PMCID: PMC8113506 DOI: 10.1038/s41467-021-22863-0] [Citation(s) in RCA: 65] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2019] [Accepted: 03/29/2021] [Indexed: 12/13/2022] Open
Abstract
With age, hematopoietic stem cells (HSC) undergo changes in function, including reduced regenerative potential and loss of quiescence, which is accompanied by a significant expansion of the stem cell pool that can lead to haematological disorders. Elevated metabolic activity has been implicated in driving the HSC ageing phenotype. Here we show that nicotinamide riboside (NR), a form of vitamin B3, restores youthful metabolic capacity by modifying mitochondrial function in multiple ways including reduced expression of nuclear encoded metabolic pathway genes, damping of mitochondrial stress and a decrease in mitochondrial mass and network-size. Metabolic restoration is dependent on continuous NR supplementation and accompanied by a shift of the aged transcriptome towards the young HSC state, more youthful bone marrow cellular composition and an improved regenerative capacity in a transplant setting. Consequently, NR administration could support healthy ageing by re-establishing a more youthful hematopoietic system.
Collapse
Affiliation(s)
- Xuan Sun
- Biomedical Manufacturing Commonwealth Scientific and Industrial Research Organisation (CSIRO), Melbourne, VIC, Australia
- Australian Regenerative Medicine Institute, Monash University, Melbourne, VIC, Australia
| | - Benjamin Cao
- Biomedical Manufacturing Commonwealth Scientific and Industrial Research Organisation (CSIRO), Melbourne, VIC, Australia
- Australian Regenerative Medicine Institute, Monash University, Melbourne, VIC, Australia
| | - Marina Naval-Sanchez
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD, Australia
| | - Tony Pham
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD, Australia
| | - Yu Bo Yang Sun
- Australian Regenerative Medicine Institute, Monash University, Melbourne, VIC, Australia
- Monash Biomedicine Discovery Institute, Melbourne, VIC, Australia
- Department of Anatomy and Developmental Biology, Monash University, Melbourne, VIC, Australia
| | - Brenda Williams
- Biomedical Manufacturing Commonwealth Scientific and Industrial Research Organisation (CSIRO), Melbourne, VIC, Australia
- Australian Regenerative Medicine Institute, Monash University, Melbourne, VIC, Australia
| | - Shen Y Heazlewood
- Biomedical Manufacturing Commonwealth Scientific and Industrial Research Organisation (CSIRO), Melbourne, VIC, Australia
- Australian Regenerative Medicine Institute, Monash University, Melbourne, VIC, Australia
| | - Nikita Deshpande
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD, Australia
| | - Jinhua Li
- Monash Biomedicine Discovery Institute, Melbourne, VIC, Australia
- Department of Anatomy and Developmental Biology, Monash University, Melbourne, VIC, Australia
| | - Felix Kraus
- Monash Biomedicine Discovery Institute, Melbourne, VIC, Australia
- Department of Biochemistry and Molecular Biology, Monash University, Melbourne, VIC, Australia
| | - James Rae
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD, Australia
| | - Quan Nguyen
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD, Australia
| | - Hamed Yari
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD, Australia
| | - Jan Schröder
- Australian Regenerative Medicine Institute, Monash University, Melbourne, VIC, Australia
- Monash Biomedicine Discovery Institute, Melbourne, VIC, Australia
- Department of Anatomy and Developmental Biology, Monash University, Melbourne, VIC, Australia
| | - Chad K Heazlewood
- Biomedical Manufacturing Commonwealth Scientific and Industrial Research Organisation (CSIRO), Melbourne, VIC, Australia
- Australian Regenerative Medicine Institute, Monash University, Melbourne, VIC, Australia
| | - Madeline Fulton
- Biomedical Manufacturing Commonwealth Scientific and Industrial Research Organisation (CSIRO), Melbourne, VIC, Australia
- Australian Regenerative Medicine Institute, Monash University, Melbourne, VIC, Australia
| | - Jessica Hatwell-Humble
- Biomedical Manufacturing Commonwealth Scientific and Industrial Research Organisation (CSIRO), Melbourne, VIC, Australia
- Australian Regenerative Medicine Institute, Monash University, Melbourne, VIC, Australia
| | - Kaustav Das Gupta
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD, Australia
- IMB Centre for Inflammation and Disease Research, The University of Queensland, St. Lucia, QLD, Australia
- Australian Infectious Diseases Research Centre, The University of Queensland, St. Lucia, QLD, Australia
| | - Ronan Kapetanovic
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD, Australia
- IMB Centre for Inflammation and Disease Research, The University of Queensland, St. Lucia, QLD, Australia
- Australian Infectious Diseases Research Centre, The University of Queensland, St. Lucia, QLD, Australia
| | - Xiaoli Chen
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD, Australia
| | - Matthew J Sweet
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD, Australia
- IMB Centre for Inflammation and Disease Research, The University of Queensland, St. Lucia, QLD, Australia
- Australian Infectious Diseases Research Centre, The University of Queensland, St. Lucia, QLD, Australia
| | - Robert G Parton
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD, Australia
- Centre for Microscopy and Microanalysis, The University of Queensland, St. Lucia, QLD, Australia
| | - Michael T Ryan
- Monash Biomedicine Discovery Institute, Melbourne, VIC, Australia
- Department of Biochemistry and Molecular Biology, Monash University, Melbourne, VIC, Australia
| | - Jose M Polo
- Australian Regenerative Medicine Institute, Monash University, Melbourne, VIC, Australia
- Monash Biomedicine Discovery Institute, Melbourne, VIC, Australia
- Department of Anatomy and Developmental Biology, Monash University, Melbourne, VIC, Australia
| | - Christian M Nefzger
- Australian Regenerative Medicine Institute, Monash University, Melbourne, VIC, Australia.
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD, Australia.
- Monash Biomedicine Discovery Institute, Melbourne, VIC, Australia.
- Department of Anatomy and Developmental Biology, Monash University, Melbourne, VIC, Australia.
| | - Susan K Nilsson
- Biomedical Manufacturing Commonwealth Scientific and Industrial Research Organisation (CSIRO), Melbourne, VIC, Australia.
- Australian Regenerative Medicine Institute, Monash University, Melbourne, VIC, Australia.
| |
Collapse
|
247
|
Shi Y, Ye D, Huang R, Xu Y, Lu P, Chen H, Huang J. Down Syndrome Critical Region 1 Reduces Oxidative Stress-Induced Retinal Ganglion Cells Apoptosis via CREB-Bcl-2 Pathway. Invest Ophthalmol Vis Sci 2021; 61:23. [PMID: 33104163 PMCID: PMC7594594 DOI: 10.1167/iovs.61.12.23] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Purpose Irreversible retina ganglion cell (RGC) loss is a key process during glaucoma progression. Down syndrome critical region 1 (DSCR1) has been shown to have protective effects against neuronal death. In this study, we aimed to investigate the neuroprotective mechanisms of DSCR1 on RGCs. Methods DBA/2J mice and optic nerve crush (ONC) rat model were used for vivo assays. Oxidative stress model of primary RGCs was carried out with in vitro transduction. DSCR1 protein localization was assessed by immunofluorescence. Differential protein expression was validated by Western blot, and gene expression was detected by real-time PCR. TUNEL was used to identify cell apoptosis, and 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide was used to analyze cell viability. Results Significant upregulation of DSCR1 was observed in DBA/2J mice, ONC rat model, and RGCs treated with H2O2, reaching peaks at the age of 6 months in DBA/2J mice, 5 days after ONC in rats, and 24 hours after H2O2 treatment in RGCs, respectively. DSCR1 was shown to be expressed in the ganglion cell layer. In vitro, overexpressed DSCR1 significantly promoted phosphorylation of cyclic AMP response element binding protein (CREB), B-cell lymphoma 2 (Bcl-2) expression, and RGC survival rate while reducing cleaved caspase 3 expression in H2O2-treated RGCs. On the other hand, the opposite effects were shown after knockdown of DSCR1. In addition, silencing of CREB inhibited expression of DSCR1. Conclusions Our results suggested that DSCR1 might protect the RGCs against oxidative stress via the CREB–Bcl-2 pathway, which may provide a theoretical basis for future treatments of glaucoma.
Collapse
Affiliation(s)
- Yuxun Shi
- State Key Laboratory of Ophthalmology, Department of Glaucoma, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Dan Ye
- State Key Laboratory of Ophthalmology, Department of Glaucoma, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Rong Huang
- State Key Laboratory of Ophthalmology, Department of Glaucoma, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Yue Xu
- State Key Laboratory of Ophthalmology, Department of Glaucoma, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Peng Lu
- State Key Laboratory of Ophthalmology, Department of Glaucoma, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Hailiu Chen
- State Key Laboratory of Ophthalmology, Department of Glaucoma, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Jingjing Huang
- State Key Laboratory of Ophthalmology, Department of Glaucoma, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
248
|
Kompella UB, Hartman RR, Patil MA. Extraocular, periocular, and intraocular routes for sustained drug delivery for glaucoma. Prog Retin Eye Res 2021; 82:100901. [PMID: 32891866 PMCID: PMC8317199 DOI: 10.1016/j.preteyeres.2020.100901] [Citation(s) in RCA: 64] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2019] [Revised: 08/22/2020] [Accepted: 08/30/2020] [Indexed: 02/07/2023]
Abstract
Although once daily anti-glaucoma drug therapy is a current clinical reality, most therapies require multiple dosing and there is an unmet need to develop convenient, safe, and effective sustained release drug delivery systems for long-term treatment to improve patient adherence and outcomes. One of the first sustained release drug delivery systems was approved for the reduction of intraocular pressure in glaucoma patients. It is a polymeric reservoir-type insert delivery system, Ocusert™, placed under the eyelid and on the ocular surface for zero-order drug release over one week. The insert, marketed in two strengths, released pilocarpine on the eye surface. While many clinicians appreciated this drug product, it was eventually discontinued. No similar sustained release non-invasive drug delivery system has made it to the market to date for treating glaucoma. Drug delivery systems under development include punctal plugs, ring-type systems, contact lenses, implants, microspheres, nanospheres, gels, and other depot systems placed in the extraocular, periocular, or intraocular regions including intracameral, supraciliary, and intravitreal spaces. This article discusses the advantages and disadvantages of the various routes of administration and delivery systems for sustained glaucoma therapy. It also provides the reader with some examples and discussion of drug delivery systems that could potentially be applied for glaucoma treatment. Interestingly, one intracamerally injected implant, Durysta™, was approved recently for sustained intraocular pressure reduction. However, long-term acceptance of such devices has yet to be established. The ultimate success of the delivery system will depend on efficacy relative to eye drop dosing, safety, reimbursement options, and patient acceptance. Cautious development efforts are warranted considering prior failed approaches for sustained glaucoma drug delivery. Neuroprotective approaches for glaucoma therapy including cell, gene, protein, and drug-combination therapies, mostly administered intravitreally, are also rapidly progressing towards assessment in humans.
Collapse
Affiliation(s)
- Uday B Kompella
- Department of Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO, USA; Department of Ophthalmology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA; Department of Bioengineering, University of Colorado Anschutz Medical Campus, Aurora, CO, USA; Colorado Center for Nanomedicine and Nanosafety, University of Colorado Anschutz Medical Campus, Aurora, CO, USA.
| | - Rachel R Hartman
- Department of Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Madhoosudan A Patil
- Department of Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| |
Collapse
|
249
|
Liu H, Xiu Y, Zhang Q, Xu Y, Wan Q, Tao L. Silencing microRNA‑29b‑3p expression protects human trabecular meshwork cells against oxidative injury via upregulation of RNF138 to activate the ERK pathway. Int J Mol Med 2021; 47:101. [PMID: 33907817 PMCID: PMC8054636 DOI: 10.3892/ijmm.2021.4934] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Accepted: 02/26/2021] [Indexed: 12/25/2022] Open
Abstract
In recent years, the potential involvement of numerous microRNAs (miRNAs) in glaucoma has been widely reported. However, the role of microRNA-29b-3p (miR-29b-3p) in the pathogenesis of glaucoma remains unknown. This study aimed to explore the biological role and regulatory mechanism of miR-29b-3p in the oxidative injury of human trabecular meshwork (HTM) cells induced by H2O2 stimulation. By establishing a glaucoma rat model, the effects of miR-29-3p in glaucoma were detected in vivo. Our findings demonstrated that miR-29b-3p was upregulated in a glaucoma model and antagomiR-29b-3p alleviated the symptoms of glaucoma. In vitro assays revealed that miR-29b-3p expression was significantly upregulated in HTM cells with H2O2 stimulation. Knockdown of miR-29b-3p alleviated H2O2-induced oxidative injury in HTM cells by promoting cell viability, and inhibiting cell apoptosis, reactive oxygen species generation and extracellular matrix production. Subsequently, it was found that E3 ubiquitin-protein ligase RNF138 (RNF138) was a downstream target of miR-29b-3p. RNF138 expression was downregulated in HTM cells with H2O2 stimulation. RNF138 knockdown significantly rescued the protective effect of miR-29b-3p inhibitor on HTM cells under oxidative injury. Additionally, miR-29b-3p silencing activated the ERK pathway via upregulating RNF138. Collectively, silencing of miR-29b-3p protected HTM cells against oxidative injury by upregulation of RNF138 to activate the ERK pathway.
Collapse
Affiliation(s)
- Heting Liu
- Department of Ophthalmology, The Second Hospital of Anhui Medical University, Hefei, Anhui 230601, P.R. China
| | - Yanghui Xiu
- Eye Institute and Xiamen Eye Center, Affiliated Xiamen University, Xiamen, Fujian 361000, P.R. China
| | - Qing Zhang
- Department of Ophthalmology, The Second Hospital of Anhui Medical University, Hefei, Anhui 230601, P.R. China
| | - Yuxin Xu
- Department of Ophthalmology, The Second Hospital of Anhui Medical University, Hefei, Anhui 230601, P.R. China
| | - Qianqian Wan
- Department of Ophthalmology, The Second Hospital of Anhui Medical University, Hefei, Anhui 230601, P.R. China
| | - Liming Tao
- Department of Ophthalmology, The Second Hospital of Anhui Medical University, Hefei, Anhui 230601, P.R. China
| |
Collapse
|
250
|
Tribble JR, Otmani A, Sun S, Ellis SA, Cimaglia G, Vohra R, Jöe M, Lardner E, Venkataraman AP, Domínguez-Vicent A, Kokkali E, Rho S, Jóhannesson G, Burgess RW, Fuerst PG, Brautaset R, Kolko M, Morgan JE, Crowston JG, Votruba M, Williams PA. Nicotinamide provides neuroprotection in glaucoma by protecting against mitochondrial and metabolic dysfunction. Redox Biol 2021; 43:101988. [PMID: 33932867 PMCID: PMC8103000 DOI: 10.1016/j.redox.2021.101988] [Citation(s) in RCA: 119] [Impact Index Per Article: 29.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Revised: 04/06/2021] [Accepted: 04/20/2021] [Indexed: 12/16/2022] Open
Abstract
Nicotinamide adenine dinucleotide (NAD) is a REDOX cofactor and metabolite essential for neuronal survival. Glaucoma is a common neurodegenerative disease in which neuronal levels of NAD decline. We assess the effects of nicotinamide (a precursor to NAD) on retinal ganglion cells (the affected neuron in glaucoma) in normal physiological conditions and across a range of glaucoma relevant insults including mitochondrial stress and axon degenerative insults. We demonstrate retinal ganglion cell somal, axonal, and dendritic neuroprotection by nicotinamide in rodent models which represent isolated ocular hypertensive, axon degenerative, and mitochondrial degenerative insults. We performed metabolomics enriched for small molecular weight metabolites for the retina, optic nerve, and superior colliculus which demonstrates that ocular hypertension induces widespread metabolic disruption, including consistent changes to α-ketoglutaric acid, creatine/creatinine, homocysteine, and glycerophosphocholine. This metabolic disruption is prevented by nicotinamide. Nicotinamide provides further neuroprotective effects by increasing oxidative phosphorylation, buffering and preventing metabolic stress, and increasing mitochondrial size and motility whilst simultaneously dampening action potential firing frequency. These data support continued determination of the utility of long-term nicotinamide treatment as a neuroprotective therapy for human glaucoma. Nicotinamide is neuroprotective in cell and animal models that recapitulate isolated features of glaucoma. Systemic nicotinamide administration has limited molecular side-effects on visual system tissue under basal conditions. Nicotinamide provides a robust reversal in the disease metabolic profile of glaucomatous animals. Nicotinamide increases oxidative phosphorylation, buffers and prevents metabolic stress, and increases mitochondrial size.
Collapse
Affiliation(s)
- James R Tribble
- Department of Clinical Neuroscience, Division of Eye and Vision, St. Erik Eye Hospital, Karolinska Institutet, Stockholm, Sweden.
| | - Amin Otmani
- Department of Clinical Neuroscience, Division of Eye and Vision, St. Erik Eye Hospital, Karolinska Institutet, Stockholm, Sweden.
| | - Shanshan Sun
- School of Optometry and Vision Sciences, Cardiff University, Cardiff, UK.
| | - Sevannah A Ellis
- The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Melbourne, Australia.
| | - Gloria Cimaglia
- Department of Clinical Neuroscience, Division of Eye and Vision, St. Erik Eye Hospital, Karolinska Institutet, Stockholm, Sweden; School of Optometry and Vision Sciences, Cardiff University, Cardiff, UK.
| | - Rupali Vohra
- Department of Veterinary and Animal Sciences, Pathobiological Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Denmark; Department of Drug Design and Pharmacology, Eye Translational Research Unit, Faculty of Health and Medical Sciences, University of Copenhagen, Denmark.
| | - Melissa Jöe
- Department of Clinical Neuroscience, Division of Eye and Vision, St. Erik Eye Hospital, Karolinska Institutet, Stockholm, Sweden.
| | - Emma Lardner
- Department of Clinical Neuroscience, Division of Eye and Vision, St. Erik Eye Hospital, Karolinska Institutet, Stockholm, Sweden.
| | - Abinaya P Venkataraman
- Department of Clinical Neuroscience, Division of Eye and Vision, St. Erik Eye Hospital, Karolinska Institutet, Stockholm, Sweden.
| | - Alberto Domínguez-Vicent
- Department of Clinical Neuroscience, Division of Eye and Vision, St. Erik Eye Hospital, Karolinska Institutet, Stockholm, Sweden.
| | - Eirini Kokkali
- School of Optometry and Vision Sciences, Cardiff University, Cardiff, UK.
| | - Seungsoo Rho
- School of Optometry and Vision Sciences, Cardiff University, Cardiff, UK; Department of Ophthalmology, CHA Bundang Medical Center, CHA University, Seongnam, Republic of Korea.
| | - Gauti Jóhannesson
- Department of Clinical Sciences, Ophthalmology, Umeå University, Umeå, Sweden; Wallenberg Centre of Molecular Medicine, Umeå University, Umeå, Sweden.
| | | | - Peter G Fuerst
- WWAMI Medical Education Program, University of Idaho, Moscow, ID, USA.
| | - Rune Brautaset
- Department of Clinical Neuroscience, Division of Eye and Vision, St. Erik Eye Hospital, Karolinska Institutet, Stockholm, Sweden.
| | - Miriam Kolko
- Department of Drug Design and Pharmacology, Eye Translational Research Unit, Faculty of Health and Medical Sciences, University of Copenhagen, Denmark; Department of Ophthalmology, Copenhagen University Hospital, Rigshospitalet-Glostrup, Glostrup, Denmark.
| | - James E Morgan
- School of Optometry and Vision Sciences, Cardiff University, Cardiff, UK; Cardiff Eye Unit, University Hospital Wales, Cardiff, UK; School of Medicine, Cardiff University, Cardiff, UK.
| | - Jonathan G Crowston
- Centre for Eye Research Australia, Royal Victorian Eye and Ear Hospital, Melbourne, Australia; Singapore Eye Research Institute, Singapore National Eye Centre, Singapore, Singapore; Centre for Vision Research, Neuroscience and Behavioural Disorders, Duke-NUS, Singapore, Singapore.
| | - Marcela Votruba
- School of Optometry and Vision Sciences, Cardiff University, Cardiff, UK; Cardiff Eye Unit, University Hospital Wales, Cardiff, UK.
| | - Pete A Williams
- Department of Clinical Neuroscience, Division of Eye and Vision, St. Erik Eye Hospital, Karolinska Institutet, Stockholm, Sweden.
| |
Collapse
|