1
|
Lee R, Ong J, Waisberg E, Mader T, Berdahl J, Suh A, Panzo N, Memon H, Sampige R, Katsev B, Kadipasaoglu CM, Mason CE, Beheshti A, Zwart SR, Smith SM, Lee AG. Potential Risks of Ocular Molecular and Cellular Changes in Spaceflight. Semin Ophthalmol 2025:1-11. [PMID: 40094398 DOI: 10.1080/08820538.2025.2471443] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/16/2025] [Revised: 02/12/2025] [Accepted: 02/17/2025] [Indexed: 03/19/2025]
Abstract
PURPOSE Many fundamental cellular and molecular changes are known to occur in biological systems during spaceflight, including oxidative stress, DNA damage, mitochondrial damage, epigenetic factors, telomere lengthening, and microbial shifts. We can apply the consequences of these molecular changes in ocular cells, such as the retinal ganglion cells and corneal epithelium, to identify ophthalmologic risks during spaceflight. This review aims to discuss the potential molecular changes in greater detail and apply the principles to ocular cells and ophthalmic disease risk in astronauts. METHODS A targeted, relevant search of the literature on the topic and related topics of ocular surface and spaceflight was conducted with scholarly databases PubMed, Web of Science, and Embase from inception to July2024 with search terms "oxidative stress"; "DNA damage"; "Mitochondrial Dysfunction"; "Epigenetics"; "Telomeres"; "Microbiome"; "ocular cells"; "spaceflight"; "microgravity"; "radiation." RESULTS A total of 115 articles were included following screening and eligibility assessment. Key findings include molecular changes and their contributions to ophthalmic diseases like cataracts, spaceflight-associated neuro-ocular syndrome, and dry eye syndrome. CONCLUSION This review provides a comprehensive overview of risks to vision associated with long-duration spaceflight missions beyond low Earth orbit (LEO). Further investigation into targeted countermeasures is imperative to mitigate vision-threatening sequelae in astronauts undertaking deep-space exploration.
Collapse
Affiliation(s)
- Ryung Lee
- Touro College of Osteopathic Medicine, New York, NY, USA
| | - Joshua Ong
- Department of Ophthalmology and Visual Sciences, University of Michigan Kellogg Eye Center, Ann Arbor, Michigan, USA
| | - Ethan Waisberg
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
| | | | - John Berdahl
- Vance Thompson Vision, Sioux Falls, South Dakota, USA
| | - Alex Suh
- Tulane School of Medicine, New Orleans, Louisiana, USA
| | | | - Hamza Memon
- Texas A&M School of Medicine, Bryan, TX, USA
| | - Ritu Sampige
- Center for Space Medicine, Baylor College of Medicine, Houston, TX, USA
| | - Blake Katsev
- Department of Ophthalmology and Visual Sciences, Washington University, Saint Louis, MO, USA
| | | | - Christopher E Mason
- Department of Physiology, Biophysics and Systems Biology, Weill Cornell Medicine, New York, NY, USA
| | - Afshin Beheshti
- Center for Space Biomedicine, McGowan Institute for Regenerative Medicine, Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Sara R Zwart
- Human Health and Performance Directorate, NASA Johnson Space Center, Houston, TX, USA
| | - Scott M Smith
- Human Health and Performance Directorate, NASA Johnson Space Center, Houston, TX, USA
| | - Andrew G Lee
- Department of Ophthalmology, Blanton Eye Institute, Houston Methodist Hospital, Houston, TX, USA
- Departments of Ophthalmology, Neurology, and Neurosurgery, Weill Cornell Medicine, New York, New York, USA
- Department of Ophthalmology, University of Texas Medical Branch, Galveston, TX, USA
- University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Department of Ophthalmology, The University of Iowa Hospitals and Clinics, Iowa City, Iowa, USA
| |
Collapse
|
2
|
Cao H, Jiang J, Chen L, Gao L. Mimicomes: Mimicking Multienzyme System by Artificial Design. Adv Healthc Mater 2025; 14:e2402372. [PMID: 39380346 DOI: 10.1002/adhm.202402372] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Revised: 09/05/2024] [Indexed: 10/10/2024]
Abstract
Enzymes are widely distributed in organelles of cells, which are capable of carrying out specific catalytic reactions. In general, several enzymes collaborate to facilitate complex reactions and engage in vital biochemical processes within cells, which are also called cascade systems. The cascade systems are highly efficient, and their dysfunction is associated with a multitude of endogenous diseases. The advent of nanotechnology makes it possible to mimic these cascade systems in nature and realize partial functions of natural biological processes both in vitro and in vivo. To emphasize the significance of artificial cascade systems, mimicomes is first proposed, a new concept that refers to the artificial cascade catalytic systems. Typically, mimicomes are able to mimic specific natural biochemical catalytic processes or facilitate the overall catalytic efficiency of cascade systems. Subsequently, the evolution and development of different types of mimicomes in recent decades are elucidated exhaustedly, from the natural enzyme-based mimicomes (immobilized enzyme and vesicle mimicomes) to the nanozyme-based mimicomes and enzyme-nanozyme hybrid mimicomes. In conclusion, the remaining challenges in the design of multifunctional mimicomes and their potential applications are summarized, offering insights into their future prospects.
Collapse
Affiliation(s)
- Haolin Cao
- CAS Engineering Laboratory for Nanozyme, Key Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China
| | - Jing Jiang
- CAS Engineering Laboratory for Nanozyme, Key Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China
| | - Lei Chen
- CAS Engineering Laboratory for Nanozyme, Key Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China
- Nanozyme Laboratory in Zhongyuan, Henan Academy of Innovations in Medical Science, Zhengzhou, Henan, 450052, China
| | - Lizeng Gao
- CAS Engineering Laboratory for Nanozyme, Key Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China
- Nanozyme Laboratory in Zhongyuan, Henan Academy of Innovations in Medical Science, Zhengzhou, Henan, 450052, China
- Nanozyme Medical Center, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan, 450001, China
| |
Collapse
|
3
|
Singh H, Singh R, Singh A, Singh H, Singh G, Kaur S, Singh B. Role of oxidative stress in diabetes-induced complications and their management with antioxidants. Arch Physiol Biochem 2024; 130:616-641. [PMID: 37571852 DOI: 10.1080/13813455.2023.2243651] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Revised: 05/30/2023] [Accepted: 06/02/2023] [Indexed: 08/13/2023]
Abstract
Diabetes mellitus (DM) is a huge global health issue and one of the most studied diseases, with a large global prevalence. Oxidative stress is a cytotoxic consequence of the excessive development of ROS and suppression of the antioxidant defense system for ROS elimination, which accelerates the progression of diabetes complications such as diabetic neuropathy, retinopathy, and nephropathy. Hyperglycaemia induced oxidative stress causes the activation of seven major pathways implicated in the pathogenesis of diabetic complications. These pathways increase the production of ROS and RNS, which contributes to dysregulated autophagy, gene expression changes, and the development of numerous pro-inflammatory mediators which may eventually lead to diabetic complications. This review will illustrate that oxidative stress plays a vital role in the pathogenesis of diabetic complications, and the use of antioxidants will help to reduce oxidative stress and thus may alleviate diabetic complications.
Collapse
Affiliation(s)
- Hasandeep Singh
- Department of Pharmaceutical Sciences, Guru Nanak Dev University, Amritsar, 143005, India
| | - Rajanpreet Singh
- Department of Pharmaceutical Sciences, Guru Nanak Dev University, Amritsar, 143005, India
| | - Arshdeep Singh
- Department of Pharmaceutical Sciences, Guru Nanak Dev University, Amritsar, 143005, India
| | - Harshbir Singh
- Department of Pharmaceutical Sciences, Guru Nanak Dev University, Amritsar, 143005, India
| | - Gurpreet Singh
- Department of Pharmaceutical Sciences, Guru Nanak Dev University, Amritsar, 143005, India
| | - Sarabjit Kaur
- Department of Pharmaceutical Sciences, Guru Nanak Dev University, Amritsar, 143005, India
| | - Balbir Singh
- Department of Pharmaceutical Sciences, Guru Nanak Dev University, Amritsar, 143005, India
| |
Collapse
|
4
|
Choi JY, Han E, Yoo TK. Application of ChatGPT-4 to oculomics: a cost-effective osteoporosis risk assessment to enhance management as a proof-of-principles model in 3PM. EPMA J 2024; 15:659-676. [PMID: 39635018 PMCID: PMC11612069 DOI: 10.1007/s13167-024-00378-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Accepted: 08/20/2024] [Indexed: 12/07/2024]
Abstract
Background Oculomics is an emerging medical field that focuses on the study of the eye to detect and understand systemic diseases. ChatGPT-4 is a highly advanced AI model with multimodal capabilities, allowing it to process text and statistical data. Osteoporosis is a chronic condition presenting asymptomatically but leading to fractures if untreated. Current diagnostic methods like dual X-ray absorptiometry (DXA) are costly and involve radiation exposure. This study aims to develop a cost-effective osteoporosis risk prediction tool using ophthalmological data and ChatGPT-4 based on oculomics, aligning with predictive, preventive, and personalized medicine (3PM) principles. Working hypothesis and methods We hypothesize that leveraging ophthalmological data (oculomics) combined with AI-driven regression models developed by ChatGPT-4 can significantly improve the predictive accuracy for osteoporosis risk. This integration will facilitate earlier detection, enable more effective preventive strategies, and support personalized treatment plans tailored to individual patients. We utilized DXA and ophthalmological data from the Korea National Health and Nutrition Examination Survey to develop and validate osteopenia and osteoporosis prediction models. Ophthalmological and demographic data were integrated into logistic regression analyses, facilitated by ChatGPT-4, to create prediction formulas. These models were then converted into calculator software through automated coding by ChatGPT-4. Results ChatGPT-4 automatically developed prediction models based on key predictors of osteoporosis and osteopenia included age, gender, weight, and specific ophthalmological conditions such as cataracts and early age-related macular degeneration, and successfully implemented a risk calculator tool. The oculomics-based models outperformed traditional methods, with area under the curve of the receiver operating characteristic values of 0.785 for osteopenia and 0.866 for osteoporosis in the validation set. The calculator demonstrated high sensitivity and specificity, providing a reliable tool for early osteoporosis screening. Conclusions and expert recommendations in the framework of 3PM This study illustrates the value of integrating ophthalmological data into multi-level diagnostics for osteoporosis, significantly improving the accuracy of health risk assessment and the identification of at-risk individuals. Aligned with the principles of 3PM, this approach fosters earlier detection and enables the development of individualized patient profiles, facilitating personalized and targeted treatment strategies. This study also highlights the potential of AI, specifically ChatGPT-4, in developing accessible, cost-effective, and radiation-free screening tools for advancing 3PM in clinical practice. Our findings emphasize the importance of a holistic approach, incorporating comprehensive health indices and interdisciplinary collaboration, to deliver personalized management plans. Preventive strategies should focus on lifestyle modifications and targeted interventions to enhance bone health, thereby preventing the progression of osteoporosis and contributing to overall patient well-being. Supplementary Information The online version contains supplementary material available at 10.1007/s13167-024-00378-0.
Collapse
Affiliation(s)
- Joon Yul Choi
- Department of Biomedical Engineering, Yonsei University, Wonju, South Korea
| | - Eoksoo Han
- Electronics and Telecommunications Research Institute (ETRI), Daejeon, South Korea
| | - Tae Keun Yoo
- Department of Ophthalmology, Hangil Eye Hospital, 35 Bupyeong-Daero, Bupyeong-Gu, Incheon, 21388 South Korea
| |
Collapse
|
5
|
Chen S, Sun D, Zhang S, Xu L, Wang N, Li H, Xu X, Wei F. TIN2 modulates FOXO1 mitochondrial shuttling to enhance oxidative stress-induced apoptosis in retinal pigment epithelium under hyperglycemia. Cell Death Differ 2024; 31:1487-1505. [PMID: 39080375 PMCID: PMC11519896 DOI: 10.1038/s41418-024-01349-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Revised: 07/12/2024] [Accepted: 07/18/2024] [Indexed: 10/30/2024] Open
Abstract
Progressive dysfunction of the retinal pigment epithelium (RPE) and the adjacent photoreceptor cells in the outer retina plays a pivotal role in the pathogenesis of diabetic retinopathy (DR). Here, we observed a marked increase in oxidative stress-induced apoptosis in parallel with higher expression of telomeric protein TIN2 in RPE cells under hyperglycemia in vivo and in vitro. Delving deeper, we confirm that high glucose-induced elevation of mitochondria-localized TIN2 compromises mitochondrial activity and weakens the intrinsic antioxidant defense, thereby leading to the activation of mitochondria-dependent apoptotic pathways. Mechanistically, mitochondrial TIN2 promotes the phosphorylation of FOXO1 and its relocation to the mitochondria. Such translocation of transcription factor FOXO1 not only promotes its binding to the D-loop region of mitochondrial DNA-resulting in the inhibition of mitochondrial respiration-but also hampers its availability to nuclear target DNA, thereby undermining the intrinsic antioxidant defense. Moreover, TIN2 knockdown effectively mitigates oxidative-induced apoptosis in diabetic mouse RPE by preserving mitochondrial homeostasis, which concurrently prevents secondary photoreceptor damage. Our study proposes the potential of TIN2 as a promising molecular target for therapeutic interventions for diabetic retinopathy, which emphasizes the potential significance of telomeric proteins in the regulation of metabolism and mitochondrial function. Created with BioRender ( https://www.biorender.com/ ).
Collapse
Affiliation(s)
- Shimei Chen
- Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China
- National Clinical Research Center for Eye Diseases; Shanghai Key Laboratory of Ocular Fundus Disease; Shanghai Engineering Center for Visual Science and Photo Medicine; Shanghai Engineering Center for Precise Diagnosis and Treatment of Eye Diseases, Shanghai, 200080, China
| | - Dandan Sun
- Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China
- National Clinical Research Center for Eye Diseases; Shanghai Key Laboratory of Ocular Fundus Disease; Shanghai Engineering Center for Visual Science and Photo Medicine; Shanghai Engineering Center for Precise Diagnosis and Treatment of Eye Diseases, Shanghai, 200080, China
| | - Shuchang Zhang
- Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China
- National Clinical Research Center for Eye Diseases; Shanghai Key Laboratory of Ocular Fundus Disease; Shanghai Engineering Center for Visual Science and Photo Medicine; Shanghai Engineering Center for Precise Diagnosis and Treatment of Eye Diseases, Shanghai, 200080, China
| | - Li Xu
- Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China
- National Clinical Research Center for Eye Diseases; Shanghai Key Laboratory of Ocular Fundus Disease; Shanghai Engineering Center for Visual Science and Photo Medicine; Shanghai Engineering Center for Precise Diagnosis and Treatment of Eye Diseases, Shanghai, 200080, China
| | - Ning Wang
- Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China
- National Clinical Research Center for Eye Diseases; Shanghai Key Laboratory of Ocular Fundus Disease; Shanghai Engineering Center for Visual Science and Photo Medicine; Shanghai Engineering Center for Precise Diagnosis and Treatment of Eye Diseases, Shanghai, 200080, China
| | - Huiming Li
- Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Xun Xu
- Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China.
- National Clinical Research Center for Eye Diseases; Shanghai Key Laboratory of Ocular Fundus Disease; Shanghai Engineering Center for Visual Science and Photo Medicine; Shanghai Engineering Center for Precise Diagnosis and Treatment of Eye Diseases, Shanghai, 200080, China.
| | - Fang Wei
- Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China.
- National Clinical Research Center for Eye Diseases; Shanghai Key Laboratory of Ocular Fundus Disease; Shanghai Engineering Center for Visual Science and Photo Medicine; Shanghai Engineering Center for Precise Diagnosis and Treatment of Eye Diseases, Shanghai, 200080, China.
| |
Collapse
|
6
|
Wood JPM, Chidlow G, Wall GM, Casson RJ. N-acetylcysteine amide and di- N-acetylcysteine amide protect retinal cells in culture via an antioxidant action. Exp Eye Res 2024; 248:110074. [PMID: 39251120 DOI: 10.1016/j.exer.2024.110074] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Revised: 09/03/2024] [Accepted: 09/04/2024] [Indexed: 09/11/2024]
Abstract
Reactive oxygen species (ROS) play a significant role in toxicity to the retina in a variety of diseases. N-acetylcysteine (NAC), N-acetylcysteine amide (NACA) and the dimeric di-N-acetylcysteine amide (diNACA) were evaluated in terms of protecting retinal cells, in vitro, in a variety of stress models. Three types of rat retinal cell cultures were utilized in the study: macroglial-only cell cultures, neuron-only retinal ganglion cell (RGC) cultures, and mixed cultures containing retinal glia and neurons. Ability of test agents to attenuate oxidative stress in all cultures was ascertained. In addition, capability of agents to protect against a variety of alternate clinically-relevant stressors, including excitotoxins and mitochondrial electron transport chain inhibitors, was also evaluated. Capacity of test agents to elevate cellular levels of reduced glutathione under normal and compromised conditions was also determined. NAC, NACA and diNACA demonstrated concentration-dependent cytoprotection against oxidative stress in all cultures. These three compounds, however, had differing effects against a variety of alternate insults to retinal cells. The most protective agent was NACA, which was most potent against the most stressors (including oxidative stress, mitochondrial impairment by antimycin A and azide, and glutamate-induced excitotoxicity). Similar to NAC, NACA increased glutathione levels in non-injured cells, although diNACA did not, suggesting a different, unknown mechanism of antioxidant activity for the latter. In support of this, diNACA was the only agent to attenuate rotenone-induced toxicity in mitochondria. NAC, NACA and diNACA exhibited varying degrees of antioxidant activity, i.e., protected cultured rat retinal cells from a variety of stressors which were designed to mimic aspects of the pathology of different retinal diseases. A general rank order of activity was observed: NACA ≥ diNACA > NAC. These results warrant further exploration of NACA and diNACA as antioxidant therapeutics for the treatment of retinal diseases, particularly those involving oxidative stress. Furthermore, we have defined the battery of tests carried out as the "Wood, Chidlow, Wall and Casson (WCWC) Retinal Antioxidant Indices"; we believe that these are of great value for screening molecules for potential to reduce retinal oxidative stress in a range of retinal diseases.
Collapse
Affiliation(s)
- John P M Wood
- Discipline of Ophthalmology & Visual Sciences, Level 7 Adelaide Health and Medical Sciences Building, University of Adelaide, North Terrace, Adelaide, SA, 5000, Australia; South Australian Institute of Ophthalmology, Royal Adelaide Hospital, Port Road, SA 5000, Australia.
| | - Glyn Chidlow
- Discipline of Ophthalmology & Visual Sciences, Level 7 Adelaide Health and Medical Sciences Building, University of Adelaide, North Terrace, Adelaide, SA, 5000, Australia; South Australian Institute of Ophthalmology, Royal Adelaide Hospital, Port Road, SA 5000, Australia
| | | | - Robert J Casson
- Discipline of Ophthalmology & Visual Sciences, Level 7 Adelaide Health and Medical Sciences Building, University of Adelaide, North Terrace, Adelaide, SA, 5000, Australia; South Australian Institute of Ophthalmology, Royal Adelaide Hospital, Port Road, SA 5000, Australia
| |
Collapse
|
7
|
Zhang X, Yao S, Zhang L, Zhang B, Yang M, Guo Q, Xu J, Wang Z, Lei B, Jin X. Mitochondrial functional impairment in ARL3-mutation related rod-cone dystrophy. FASEB Bioadv 2024; 6:555-564. [PMID: 39512837 PMCID: PMC11539027 DOI: 10.1096/fba.2023-00138] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2023] [Revised: 08/20/2024] [Accepted: 08/26/2024] [Indexed: 11/15/2024] Open
Abstract
Mitochondria are vital for retinal cell function and survival, and there is growing evidence linking mitochondrial dysfunction to retinal degenerations. Although ARL3 mutations have been linked to multiple forms of retinal degeneration, the relationship between ARL3 and mitochondria remains unexplored. Herein, we investigated the effects of ARL3 T31A , ARL3 C118F , and ARL3 T31A/C118F mutations on mitochondrial function in fibroblasts obtained from patients with ARL3-related rod-cone dystrophy. Our findings revealed that these mutations led to a decrease in mitochondrial respiration, an increase in the accumulation mitochondrial reactive oxygen species (ROS), and induction of apoptosis in fibroblasts. Additionally, we conducted a comparative analysis of the effects of ARL3T31A, ARL3C118F, and ARL3T31A/C118F proteins on mitochondria in ARPE-19 cells. Results showed that ARL3T31A and ARL3T31A/C118F not only affected mitochondrial function but also induced apoptosis in ARPE-19 cells. Conversely, ARL3C118F primarily influenced cell apoptosis with minimal effects on mitochondrial function in ARPE-19 cells. Transcriptome analysis further suggested the involvement of respiratory electron transport, response to ROS, and apoptotic signaling pathways in ARL3T31A/C118F cells. Our study demonstrated that ARL3-related mutations play a significant role in the diversity of mitochondrial function, providing novel insights into the functional analysis of ARL3-related mutations.
Collapse
Affiliation(s)
- Xiaoli Zhang
- Institute of Neuroscience and The Third Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
| | - Shun Yao
- Henan Eye Institute, People's Hospital of Zhengzhou University, Henan Provincial People's HospitalZhengzhouChina
- Eye instituteHenan Academy of Innovations in Medical ScienceZhengzhouChina
| | - Lujia Zhang
- Institute of Neuroscience and The Third Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
| | - Beisi Zhang
- Henan Eye Institute, People's Hospital of Zhengzhou University, Henan Provincial People's HospitalZhengzhouChina
| | - Mingzhu Yang
- Henan Eye Institute, People's Hospital of Zhengzhou University, Henan Provincial People's HospitalZhengzhouChina
| | - Qingge Guo
- Henan Eye Institute, People's Hospital of Zhengzhou University, Henan Provincial People's HospitalZhengzhouChina
| | - Jin Xu
- Henan Eye Institute, People's Hospital of Zhengzhou University, Henan Provincial People's HospitalZhengzhouChina
- Eye instituteHenan Academy of Innovations in Medical ScienceZhengzhouChina
| | - Zhongfeng Wang
- Institute of Neuroscience and The Third Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain ScienceInstitutes of Brain Science, Fudan UniversityShanghaiChina
| | - Bo Lei
- Henan Eye Institute, People's Hospital of Zhengzhou University, Henan Provincial People's HospitalZhengzhouChina
- Eye instituteHenan Academy of Innovations in Medical ScienceZhengzhouChina
| | - Xiuxiu Jin
- Institute of Neuroscience and The Third Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
- Eye instituteHenan Academy of Innovations in Medical ScienceZhengzhouChina
| |
Collapse
|
8
|
Warwick AM, Bomze HM, Wang L, Hao Y, Stinnett SS, Gospe SM. Hypoxia-mediated rescue of retinal ganglion cells deficient in mitochondrial complex I is independent of the hypoxia-inducible factor pathway. Sci Rep 2024; 14:24114. [PMID: 39406814 PMCID: PMC11480089 DOI: 10.1038/s41598-024-75916-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2024] [Accepted: 10/09/2024] [Indexed: 10/19/2024] Open
Abstract
Continuous exposure to environmental hypoxia (11% O2) has been shown to markedly slow the progressive degeneration of retinal ganglion cells (RGCs) in a mouse model of mitochondrial optic neuropathy with RGC-specific deletion of the key mitochondrial complex I accessory subunit ndufs4. As a first step toward identifying the therapeutic mechanism of hypoxia in this model, we conducted a series of experiments to investigate the role of the hypoxia-inducible factor (HIF) regulatory pathway in RGC neuroprotection. Vglut2-Cre; ndufs4loxP/loxP mice were crossed with strains bearing floxed alleles of the negative HIF regulatory vhl or of the two major HIF α-subunit isoforms, Hif1α and Hif2α. Deletion of vhl within ndufs4-deficient RGCs failed to prevent RGC degeneration under normoxia, indicating that HIF activation is not sufficient to achieve RGC rescue. Furthermore, the rescue of ndufs4-deficient RGCs by hypoxia remained robust despite genetic inactivation of Hif1α and Hif2α. Our findings demonstrate that the HIF pathway is entirely dispensable to the rescue of RGCs by hypoxia. Future efforts to uncover key HIF-independent molecular pathways induced by hypoxia in this mouse model may be of therapeutic relevance to mitochondrial optic neuropathies such as Leber hereditary optic neuropathy.
Collapse
Affiliation(s)
- Alexander M Warwick
- Department of Ophthalmology, Duke University School of Medicine, Durham, NC, 27710, USA
| | - Howard M Bomze
- Department of Ophthalmology, Duke University School of Medicine, Durham, NC, 27710, USA
| | - Luyu Wang
- Department of Ophthalmology, Duke University School of Medicine, Durham, NC, 27710, USA
| | - Ying Hao
- Department of Ophthalmology, Duke University School of Medicine, Durham, NC, 27710, USA
| | - Sandra S Stinnett
- Department of Ophthalmology, Duke University School of Medicine, Durham, NC, 27710, USA
| | - Sidney M Gospe
- Department of Ophthalmology, Duke University School of Medicine, Durham, NC, 27710, USA.
- Department of Ophthalmology, Box 3712 Med Center, Duke University, 2351 Erwin Road, Durham, NC, 27710, USA.
| |
Collapse
|
9
|
Hernandez M, Recalde S, Bezunartea J, Moreno-Orduña M, Belza I, Chas-Prat A, Perugini E, Garcia-Layana A, Fernández-Robredo P. The Scavenging Activity of Coenzyme Q 10 Plus a Nutritional Complex on Human Retinal Pigment Epithelial Cells. Int J Mol Sci 2024; 25:8070. [PMID: 39125641 PMCID: PMC11311961 DOI: 10.3390/ijms25158070] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Revised: 07/19/2024] [Accepted: 07/21/2024] [Indexed: 08/12/2024] Open
Abstract
Age-related macular degeneration (AMD) and diabetic retinopathy (DR) are common retinal diseases responsible for most blindness in working-age and elderly populations. Oxidative stress and mitochondrial dysfunction play roles in these pathogenesis, and new therapies counteracting these contributors could be of great interest. Some molecules, like coenzyme Q10 (CoQ10), are considered beneficial to maintain mitochondrial homeostasis and contribute to the prevention of cellular apoptosis. We investigated the impact of adding CoQ10 (Q) to a nutritional antioxidant complex (Nutrof Total®; N) on the mitochondrial status and apoptosis in an in vitro hydrogen peroxide (H2O2)-induced oxidative stress model in human retinal pigment epithelium (RPE) cells. H2O2 significantly increased 8-OHdG levels (p < 0.05), caspase-3 (p < 0.0001) and TUNEL intensity (p < 0.01), and RANTES (p < 0.05), caspase-1 (p < 0.05), superoxide (p < 0.05), and DRP-1 (p < 0.05) levels, and also decreased IL1β, SOD2, and CAT gene expression (p < 0.05) vs. control. Remarkably, Q showed a significant recovery in IL1β gene expression, TUNEL, TNFα, caspase-1, and JC-1 (p < 0.05) vs. H2O2, and NQ showed a synergist effect in caspase-3 (p < 0.01), TUNEL (p < 0.0001), mtDNA, and DRP-1 (p < 0.05). Our results showed that CoQ10 supplementation is effective in restoring/preventing apoptosis and mitochondrial stress-related damage, suggesting that it could be a valid strategy in degenerative processes such as AMD or DR.
Collapse
Affiliation(s)
- Maria Hernandez
- Retinal Pathologies and New Therapies Group, Experimental Ophthalmology Laboratory, Department of Ophthalmology, Clinica Universidad de Navarra, Navarra Institute for Health Research, IdiSNA, (RICORS-TERAV), 31008 Pamplona, Spain; (M.H.); (S.R.); (J.B.); (A.C.-P.); (A.G.-L.)
| | - Sergio Recalde
- Retinal Pathologies and New Therapies Group, Experimental Ophthalmology Laboratory, Department of Ophthalmology, Clinica Universidad de Navarra, Navarra Institute for Health Research, IdiSNA, (RICORS-TERAV), 31008 Pamplona, Spain; (M.H.); (S.R.); (J.B.); (A.C.-P.); (A.G.-L.)
| | - Jaione Bezunartea
- Retinal Pathologies and New Therapies Group, Experimental Ophthalmology Laboratory, Department of Ophthalmology, Clinica Universidad de Navarra, Navarra Institute for Health Research, IdiSNA, (RICORS-TERAV), 31008 Pamplona, Spain; (M.H.); (S.R.); (J.B.); (A.C.-P.); (A.G.-L.)
| | - Maite Moreno-Orduña
- Retinal Pathologies and New Therapies Group, Experimental Ophthalmology Laboratory, Department of Ophthalmology, Clinica Universidad de Navarra, 31008 Pamplona, Spain; (M.M.-O.); (I.B.); (E.P.)
| | - Idoia Belza
- Retinal Pathologies and New Therapies Group, Experimental Ophthalmology Laboratory, Department of Ophthalmology, Clinica Universidad de Navarra, 31008 Pamplona, Spain; (M.M.-O.); (I.B.); (E.P.)
| | - Ainara Chas-Prat
- Retinal Pathologies and New Therapies Group, Experimental Ophthalmology Laboratory, Department of Ophthalmology, Clinica Universidad de Navarra, Navarra Institute for Health Research, IdiSNA, (RICORS-TERAV), 31008 Pamplona, Spain; (M.H.); (S.R.); (J.B.); (A.C.-P.); (A.G.-L.)
| | - Elena Perugini
- Retinal Pathologies and New Therapies Group, Experimental Ophthalmology Laboratory, Department of Ophthalmology, Clinica Universidad de Navarra, 31008 Pamplona, Spain; (M.M.-O.); (I.B.); (E.P.)
| | - Alfredo Garcia-Layana
- Retinal Pathologies and New Therapies Group, Experimental Ophthalmology Laboratory, Department of Ophthalmology, Clinica Universidad de Navarra, Navarra Institute for Health Research, IdiSNA, (RICORS-TERAV), 31008 Pamplona, Spain; (M.H.); (S.R.); (J.B.); (A.C.-P.); (A.G.-L.)
| | - Patricia Fernández-Robredo
- Retinal Pathologies and New Therapies Group, Experimental Ophthalmology Laboratory, Department of Ophthalmology, Clinica Universidad de Navarra, Navarra Institute for Health Research, IdiSNA, (RICORS-TERAV), 31008 Pamplona, Spain; (M.H.); (S.R.); (J.B.); (A.C.-P.); (A.G.-L.)
| |
Collapse
|
10
|
Ramm L, Spoerl E, Terai N, Herber R, Pillunat LE. Association Between Corneal Changes and Retinal Oximetry in Diabetes Mellitus. Clin Ophthalmol 2024; 18:1235-1243. [PMID: 38737594 PMCID: PMC11088381 DOI: 10.2147/opth.s456020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Accepted: 04/21/2024] [Indexed: 05/14/2024] Open
Abstract
Purpose Diabetes mellitus (DM) causes different corneal changes that are associated with the severity of diabetic retinopathy. To identify the pathophysiological reasons for this, corneal tomography and optical densitometry (COD) were combined with retinal oximetry. Methods Patients with DM and healthy subjects were included in this pilot study. Spatially resolved corneal thickness and COD were assessed using the Pentacam HR (Oculus). The pachymetry difference (PACDiff) was calculated as an indicator of an increase in the peripheral corneal thickness. Oxygen saturation (SO2) of the retinal vessels was measured using the Retinal Vessel Analyzer (Imedos Systems UG). Subsequently, the associations between corneal and retinal parameters were analyzed. Results Data from 30 patients with DM were compared with those from 30 age-matched healthy subjects. In DM, arterial (P = 0.048) and venous (P < 0.001) SO2 levels were increased, and arteriovenous SO2 difference was decreased (P < 0.001). In patients, PACDiff was higher than that in healthy subjects (P < 0.05), indicating a stronger increase in peripheral corneal thickness. The COD was reduced in DM (P = 0.004). The PACDiff of concentric rings with a diameter of 4 mm (r = -0.404; P = 0.033) to 8 mm (r = -0.522; P = 0.004) was inversely correlated with the arteriovenous SO2 difference. Furthermore, PACDiff 4 mm was negatively associated with arterial SO2 (r = -0.389; P = 0.041), and the COD of the peripheral corneal areas correlated positive with arterial SO2 (COD total 10-12 mm: r = 0.408; P = 0.025). Conclusion These associations might indicate a common pathogenesis of corneal and retinal changes in DM, which could be caused by reduced oxygen supply, mitochondrial dysfunction, oxidative stress, and cytokine effects.
Collapse
Affiliation(s)
- Lisa Ramm
- Department of Ophthalmology, University Hospital Carl Gustav Carus, TU Dresden, Dresden, 01307, Germany
| | - Eberhard Spoerl
- Department of Ophthalmology, University Hospital Carl Gustav Carus, TU Dresden, Dresden, 01307, Germany
| | - Naim Terai
- Department of Ophthalmology, University Hospital Carl Gustav Carus, TU Dresden, Dresden, 01307, Germany
| | - Robert Herber
- Department of Ophthalmology, University Hospital Carl Gustav Carus, TU Dresden, Dresden, 01307, Germany
| | - Lutz E Pillunat
- Department of Ophthalmology, University Hospital Carl Gustav Carus, TU Dresden, Dresden, 01307, Germany
| |
Collapse
|
11
|
Buthelezi LM, Munsamy AJ, Mashige KP. Inflammatory mechanisms contributing to retinal alterations in HIV infection and long-term ART. South Afr J HIV Med 2024; 25:1548. [PMID: 38628910 PMCID: PMC11019112 DOI: 10.4102/sajhivmed.v25i1.1548] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Accepted: 01/24/2024] [Indexed: 04/19/2024] Open
Abstract
People living with HIV (PLWH) may face an increased risk of eye complications associated with ageing, chronic inflammation, and the toxicity arising from long-term antiretroviral therapy (ART). This review aims to understand how inflammatory pathways contribute to retinal alterations observed in PLWH on long-term ART. This review was conducted using four electronic database searches, namely Scopus, Hinari, Google Scholar, and PubMed; from 1996 (when ART became available) until January 2022, without language restriction. Sources from clinical trials, meta-analyses, randomised controlled trials, and systematic reviews were used. Dysregulated para-inflammation (chronic inflammation) damages the blood-retina barrier, resulting in the altered retinal immune privilege and leading to the development of retinal and blood vessel changes. There is an interplay between the effects of the disease versus ART. ART causes mitochondrial toxicity, which affects the retinal ganglion cells and retinal pigment epithelium (RPE) due to oxidative stress. Infection by HIV also affects retinal microglia, which contributes to RPE damage. Both of these mechanisms affect the blood vessels. Assessing the integrity of the inner and outer blood-retina barrier is a pivotal point in pinpointing the pathogenesis of inner retinal alterations. Optical coherence tomography is a valuable tool to assess these changes. There is a paucity of research to understand how these structural changes may affect visual function, such as contrast sensitivity and colour vision.
Collapse
Affiliation(s)
- Lungile M Buthelezi
- Department of Optometry, School of Health Sciences, University of KwaZulu-Natal, Durban, South Africa
| | - Alvin J Munsamy
- Department of Optometry, School of Health Sciences, University of KwaZulu-Natal, Durban, South Africa
| | - Khathutshelo P Mashige
- Department of Optometry, School of Health Sciences, University of KwaZulu-Natal, Durban, South Africa
| |
Collapse
|
12
|
Landowski M, Gogoi P, Ikeda S, Ikeda A. Roles of transmembrane protein 135 in mitochondrial and peroxisomal functions - implications for age-related retinal disease. FRONTIERS IN OPHTHALMOLOGY 2024; 4:1355379. [PMID: 38576540 PMCID: PMC10993500 DOI: 10.3389/fopht.2024.1355379] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/06/2024]
Abstract
Aging is the most significant risk factor for age-related diseases in general, which is true for age-related diseases in the eye including age-related macular degeneration (AMD). Therefore, in order to identify potential therapeutic targets for these diseases, it is crucial to understand the normal aging process and how its mis-regulation could cause age-related diseases at the molecular level. Recently, abnormal lipid metabolism has emerged as one major aspect of age-related symptoms in the retina. Animal models provide excellent means to identify and study factors that regulate lipid metabolism in relation to age-related symptoms. Central to this review is the role of transmembrane protein 135 (TMEM135) in the retina. TMEM135 was identified through the characterization of a mutant mouse strain exhibiting accelerated retinal aging and positional cloning of the responsible mutation within the gene, indicating the crucial role of TMEM135 in regulating the normal aging process in the retina. Over the past decade, the molecular functions of TMEM135 have been explored in various models and tissues, providing insights into the regulation of metabolism, particularly lipid metabolism, through its action in multiple organelles. Studies indicated that TMEM135 is a significant regulator of peroxisomes, mitochondria, and their interaction. Here, we provide an overview of the molecular functions of TMEM135 which is crucial for regulating mitochondria, peroxisomes, and lipids. The review also discusses the age-dependent phenotypes in mice with TMEM135 perturbations, emphasizing the importance of a balanced TMEM135 function for the health of the retina and other tissues including the heart, liver, and adipose tissue. Finally, we explore the potential roles of TMEM135 in human age-related retinal diseases, connecting its functions to the pathobiology of AMD.
Collapse
Affiliation(s)
- Michael Landowski
- Department of Medical Genetics, University of Wisconsin-Madison, Madison, WI, United States
- McPherson Eye Research Institute, University of Wisconsin-Madison, Madison, WI, United States
| | - Purnima Gogoi
- Department of Medical Genetics, University of Wisconsin-Madison, Madison, WI, United States
| | - Sakae Ikeda
- Department of Medical Genetics, University of Wisconsin-Madison, Madison, WI, United States
- McPherson Eye Research Institute, University of Wisconsin-Madison, Madison, WI, United States
| | - Akihiro Ikeda
- Department of Medical Genetics, University of Wisconsin-Madison, Madison, WI, United States
- McPherson Eye Research Institute, University of Wisconsin-Madison, Madison, WI, United States
| |
Collapse
|
13
|
Upreti S, Nag TC, Ghosh MP. Trolox aids coenzyme Q 10 in neuroprotection against NMDA induced damage via upregulation of VEGF in rat model of glutamate excitotoxicity. Exp Eye Res 2024; 238:109740. [PMID: 38056553 DOI: 10.1016/j.exer.2023.109740] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Revised: 10/19/2023] [Accepted: 11/24/2023] [Indexed: 12/08/2023]
Abstract
Glutamate induced damage to retinal ganglion cells (RGCs) requires tight physiological regulation of the N-methyl-D-aspartate (NMDA) receptors. Previously, studies have demonstrated the neuroprotective abilities of antioxidants like coenzyme Q10 (CoQ10) and vitamin E analogs like α-tocopherol against neuropathies resulting from NMDA insult, but have failed to shed light on the effect of CoQ10 and trolox, a hydrophilic analog of vitamin E, on glaucomatous neurodegeneration. In the current study, we wanted to investigate whether the combined effect of trolox with CoQ10 could alleviate NMDA-induced death of retinal cells while also trying to elucidate the underlying mechanism in relation to the yet unexplained role of vascular endothelial growth factor (VEGF) in NMDA-mediated excitotoxicity. After successful NMDA-induced degeneration, we followed it up with the treatment of combination of Trolox and CoQ10. The structural damage by NMDA was repaired significantly and retina retained structural integrity comparable to levels of control in the treatment group of Trolox and CoQ10. Detection of ROS generation after NMDA insult showed that together, Trolox and CoQ10 could significantly bring down the high levels of free radicals while also rescuing mitochondrial membrane potential (MMP). A significant increase in NMDA receptor Grin2A by CoQ10 alone as well as by CoQ10 and trolox was accompanied by a lowered Grin2B receptor expression, suggesting neuroprotective action of Trolox and CoQ10. Subsequently, lowered VEGFR1 and VEGFR2 receptor expression by NMDA treatment also recovered when subjected to combined treatment of Trolox and CoQ10. Western blot analyses also indicated the same whereby Trolox and CoQ10 could increase the diminished levels of phosphorylated VEGFR2. Immunofluorescence studies also indicated a positive correlation between recovered VEGFR2 and NMDAR2A levels and diminished levels of NMDAR2D, confirming the results obtained by RT-PCR analysis. This is the first report in our knowledge that demonstrates the efficacy of trolox in combination with CoQ10 highlighting the importance of maintaining VEGF levels that are lowered in ocular diseases due to NMDA-related toxicities.
Collapse
Affiliation(s)
- Shikha Upreti
- Ocular Pharmacology and Therapeutics Lab, Centre for Medical Biotechnology, Amity Institute of Biotechnology, Amity University Uttar Pradesh, Noida, 201313, India.
| | - Tapas Chandra Nag
- Department of Anatomy, All India Institute of Medical Sciences, Ansari Nagar, New Delhi, 110029, India.
| | - Madhumita P Ghosh
- Ocular Pharmacology and Therapeutics Lab, Centre for Medical Biotechnology, Amity Institute of Biotechnology, Amity University Uttar Pradesh, Noida, 201313, India.
| |
Collapse
|
14
|
Kim S, Chae JB, Kim D, Park CW, Sim Y, Lee H, Park G, Lee J, Hong S, Jana B, Kim C, Chung H, Ryu JH. Supramolecular Senolytics via Intracellular Oligomerization of Peptides in Response to Elevated Reactive Oxygen Species Levels in Aging Cells. J Am Chem Soc 2023; 145:21991-22008. [PMID: 37664981 DOI: 10.1021/jacs.3c06898] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/05/2023]
Abstract
Senolytics, which eliminate senescent cells from tissues, represent an emerging therapeutic strategy for various age-related diseases. Most senolytics target antiapoptotic proteins, which are overexpressed in senescent cells, limiting specificity and inducing severe side effects. To overcome these limitations, we constructed self-assembling senolytics targeting senescent cells with an intracellular oligomerization system. Intracellular aryl-dithiol-containing peptide oligomerization occurred only inside the mitochondria of senescent cells due to selective localization of the peptides by RGD-mediated cellular uptake into integrin αvβ3-overexpressed senescent cells and elevated levels of reactive oxygen species, which can be used as a chemical fuel for disulfide formation. This oligomerization results in an artificial protein-like nanoassembly with a stable α-helix secondary structure, which can disrupt the mitochondrial membrane via multivalent interactions because the mitochondrial membrane of senescent cells has weaker integrity than that of normal cells. These three specificities (integrin αvβ3, high ROS, and weak mitochondrial membrane integrity) of senescent cells work in combination; therefore, this intramitochondrial oligomerization system can selectively induce apoptosis of senescent cells without side effects on normal cells. Significant reductions in key senescence markers and amelioration of retinal degeneration were observed after elimination of the senescent retinal pigment epithelium by this peptide senolytic in an age-related macular degeneration mouse model and in aged mice, and this effect was accompanied by improved visual function. This system provides a strategy for the treatment of age-related diseases using supramolecular senolytics.
Collapse
Affiliation(s)
- Sangpil Kim
- Department of Chemistry, Ulsan National Institute of Science and Technology (UNIST), Ulsan 44919, Republic of Korea
| | - Jae-Byoung Chae
- Department of Ophthalmology, Konkuk University School of Medicine, Seoul 05029, Republic of Korea
| | - Dohyun Kim
- Department of Chemistry, Ulsan National Institute of Science and Technology (UNIST), Ulsan 44919, Republic of Korea
| | - Chul-Woo Park
- Department of Ophthalmology, Konkuk University School of Medicine, Seoul 05029, Republic of Korea
| | - Youjung Sim
- Department of Chemistry, Ulsan National Institute of Science and Technology (UNIST), Ulsan 44919, Republic of Korea
| | - Hyungwoo Lee
- Department of Ophthalmology, Konkuk University School of Medicine, Seoul 05029, Republic of Korea
| | - Gaeun Park
- Department of Chemistry, Ulsan National Institute of Science and Technology (UNIST), Ulsan 44919, Republic of Korea
| | - Jaeeun Lee
- Department of Chemistry, Ulsan National Institute of Science and Technology (UNIST), Ulsan 44919, Republic of Korea
| | - Seongho Hong
- Department of Chemistry, Ulsan National Institute of Science and Technology (UNIST), Ulsan 44919, Republic of Korea
| | - Batakrishna Jana
- Department of Chemistry, Ulsan National Institute of Science and Technology (UNIST), Ulsan 44919, Republic of Korea
| | - Chaekyu Kim
- Fusion Biotechnology, Ulsan 44919, Republic of Korea
| | - Hyewon Chung
- Department of Ophthalmology, Konkuk University School of Medicine, Seoul 05029, Republic of Korea
| | - Ja-Hyoung Ryu
- Department of Chemistry, Ulsan National Institute of Science and Technology (UNIST), Ulsan 44919, Republic of Korea
| |
Collapse
|
15
|
Becherucci V, Bacci GM, Marziali E, Sodi A, Bambi F, Caputo R. The New Era of Therapeutic Strategies for the Treatment of Retinitis Pigmentosa: A Narrative Review of Pathomolecular Mechanisms for the Development of Cell-Based Therapies. Biomedicines 2023; 11:2656. [PMID: 37893030 PMCID: PMC10604477 DOI: 10.3390/biomedicines11102656] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Revised: 09/20/2023] [Accepted: 09/25/2023] [Indexed: 10/29/2023] Open
Abstract
Retinitis pigmentosa, defined more properly as cone-rod dystrophy, is a paradigm of inherited diffuse retinal dystrophies, one of the rare diseases with the highest prevalence in the worldwide population and one of the main causes of low vision in the pediatric and elderly age groups. Advancements in and the understanding of molecular biology and gene-editing technologies have raised interest in laying the foundation for new therapeutic strategies for rare diseases. As a consequence, new possibilities for clinicians and patients are arising due to the feasibility of treating such a devastating disorder, reducing its complications. The scope of this review focuses on the pathomolecular mechanisms underlying RP better to understand the prospects of its treatment using innovative approaches.
Collapse
Affiliation(s)
- Valentina Becherucci
- Cell Factory Meyer, Children’s Hospital A. Meyer Istituti di Ricovero e Cura a Carattere Scientifico (IRCCS), University of Florence, 50139 Florence, Italy; (V.B.); (F.B.)
| | - Giacomo Maria Bacci
- Pediatric Ophthalmology Unit, Children’s Hospital A. Meyer Istituti di Ricovero e Cura a Carattere Scientifico (IRCCS), University of Florence, 50139 Florence, Italy; (E.M.); (R.C.)
| | - Elisa Marziali
- Pediatric Ophthalmology Unit, Children’s Hospital A. Meyer Istituti di Ricovero e Cura a Carattere Scientifico (IRCCS), University of Florence, 50139 Florence, Italy; (E.M.); (R.C.)
| | - Andrea Sodi
- Department of Neuroscience, Psychology, Drug Research and Child Health, University of Florence, 50139 Florence, Italy;
| | - Franco Bambi
- Cell Factory Meyer, Children’s Hospital A. Meyer Istituti di Ricovero e Cura a Carattere Scientifico (IRCCS), University of Florence, 50139 Florence, Italy; (V.B.); (F.B.)
| | - Roberto Caputo
- Pediatric Ophthalmology Unit, Children’s Hospital A. Meyer Istituti di Ricovero e Cura a Carattere Scientifico (IRCCS), University of Florence, 50139 Florence, Italy; (E.M.); (R.C.)
| |
Collapse
|
16
|
Liu J, Si H, Huang D, Lu D, Zou S, Qi D, Pei X, Huang S, Li Z. Mechanisms of Extraorbital Lacrimal Gland Aging in Mice: An Integrative Analysis of the Temporal Transcriptome. Invest Ophthalmol Vis Sci 2023; 64:18. [PMID: 37695604 PMCID: PMC10501490 DOI: 10.1167/iovs.64.12.18] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Accepted: 08/16/2023] [Indexed: 09/12/2023] Open
Abstract
Purpose This study used high-throughput RNA sequencing (RNA-Seq) and bioinformatics analysis to investigate the altered transcriptome profile of aging lacrimal glands in mice that occurs over the course of a 24-hour cycle. Methods Male C57BL/6J mice aged 12 weeks (young) and 20 months (aging) were housed in a pathogen-free setting with a 12-hour light/12-hour dark cycle. Throughout a 24-hour cycle, mouse extraorbital lacrimal glands (ELGs) were collected at eight time points at three-hour intervals. To prepare for the high-throughput RNA-Seq, whole mRNA was extracted. Differentially expressed genes (DEGs) in the young and aging groups were subjected to bioinformatic analysis based on diurnal patterns. Furthermore, the cell populations in which significant DEGs express and signaling pathways occur were validated at the single-cell RNA sequencing (scRNA-seq) level. Results The total transcriptome composition was significantly altered in aging ELGs compared with that in young mouse ELGs at eight time points during the 24-hour cycle, with 864 upregulated and 228 downregulated DEGs, which were primarily enriched in inflammatory pathways. Further comparative analysis of the point-to-point transcriptome revealed that aging ELGs underwent alterations in the temporal transcriptome profile in several pathways, including the inflammation-related, metabolism-related, mitochondrial bioenergetic function-associated, synaptome neural activity-associated, cell processes-associated, DNA processing-associated and fibrosis-associated pathways. Most of these pathways occurred separately in distinct cell populations. Conclusions Transcriptome profiles of aging lacrimal glands undergo considerable diurnal time-dependent changes; this finding offers a comprehensive source of information to better understand the pathophysiology of lacrimal gland aging and its underlying mechanisms.
Collapse
Affiliation(s)
- Jiangman Liu
- Department of Ophthalmology, Zhengzhou University People's Hospital, Henan Provincial People's Hospital, Zhengzhou, Henan, China
| | - Hongli Si
- Department of Ophthalmology, Zhengzhou University People's Hospital, Henan Provincial People's Hospital, Zhengzhou, Henan, China
| | - Duliurui Huang
- Department of Ophthalmology, Zhengzhou University People's Hospital, Henan Provincial People's Hospital, Zhengzhou, Henan, China
| | - Dingli Lu
- Henan Eye Institute, Henan Eye Hospital, and Henan Key Laboratory of Ophthalmology and Visual Science, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, People's Hospital of Henan University, Zhengzhou, China
| | - Sen Zou
- Henan Eye Institute, Henan Eye Hospital, and Henan Key Laboratory of Ophthalmology and Visual Science, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, People's Hospital of Henan University, Zhengzhou, China
| | - Di Qi
- Henan Eye Institute, Henan Eye Hospital, and Henan Key Laboratory of Ophthalmology and Visual Science, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, People's Hospital of Henan University, Zhengzhou, China
| | - Xiaoting Pei
- Henan Eye Institute, Henan Eye Hospital, and Henan Key Laboratory of Ophthalmology and Visual Science, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, People's Hospital of Henan University, Zhengzhou, China
| | - Shenzhen Huang
- Henan Eye Institute, Henan Eye Hospital, and Henan Key Laboratory of Ophthalmology and Visual Science, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, People's Hospital of Henan University, Zhengzhou, China
| | - Zhijie Li
- Henan Eye Institute, Henan Eye Hospital, and Henan Key Laboratory of Ophthalmology and Visual Science, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, People's Hospital of Henan University, Zhengzhou, China
| |
Collapse
|
17
|
Zaman K, Nguyen V, Prokai-Tatrai K, Prokai L. Proteomics-Based Identification of Retinal Protein Networks Impacted by Elevated Intraocular Pressure in the Hypertonic Saline Injection Model of Experimental Glaucoma. Int J Mol Sci 2023; 24:12592. [PMID: 37628770 PMCID: PMC10454042 DOI: 10.3390/ijms241612592] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Revised: 08/03/2023] [Accepted: 08/05/2023] [Indexed: 08/27/2023] Open
Abstract
Elevated intraocular pressure is considered a major cause of glaucomatous retinal neurodegeneration. To facilitate a better understanding of the underlying molecular processes and mechanisms, we report a study focusing on alterations of the retina proteome by induced ocular hypertension in a rat model of the disease. Glaucomatous processes were modeled through sclerosing the aqueous outflow routes of the eyes by hypertonic saline injections into an episcleral vein. Mass spectrometry-based quantitative retina proteomics using a label-free shotgun methodology identified over 200 proteins significantly affected by ocular hypertension. Various facets of glaucomatous pathophysiology were revealed through the organization of the findings into protein interaction networks and by pathway analyses. Concentrating on retinal neurodegeneration as a characteristic process of the disease, elevated intraocular pressure-induced alterations in the expression of selected proteins were verified by targeted proteomics based on nanoflow liquid chromatography coupled with nano-electrospray ionization tandem mass spectrometry using the parallel reaction monitoring method of data acquisition. Acquired raw data are shared through deposition to the ProteomeXchange Consortium (PXD042729), making a retina proteomics dataset on the selected animal model of glaucoma available for the first time.
Collapse
Affiliation(s)
| | | | - Katalin Prokai-Tatrai
- Department of Pharmacology and Neuroscience, University of North Texas Health Science Center, Fort Worth, TX 76107, USA; (K.Z.); (V.N.)
| | - Laszlo Prokai
- Department of Pharmacology and Neuroscience, University of North Texas Health Science Center, Fort Worth, TX 76107, USA; (K.Z.); (V.N.)
| |
Collapse
|
18
|
Cakir B, Tomita Y, Yagi H, Romfh P, Allen W, Ko M, Chen P, Fu Z, Vakhshoori D, Smith LEH. In vivo noninvasive mitochondrial redox assessment of the optic nerve head to predict disease. PNAS NEXUS 2023; 2:pgad148. [PMID: 37265545 PMCID: PMC10230116 DOI: 10.1093/pnasnexus/pgad148] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Revised: 03/19/2023] [Accepted: 04/11/2023] [Indexed: 06/03/2023]
Abstract
Eye diseases are diagnosed by visualizing often irreversible structural changes occurring late in disease progression, such as retinal ganglion cell loss in glaucoma. The retina and optic nerve head have high mitochondrial energy need. Early mitochondrial/energetics dysfunction may predict vulnerability to permanent structural changes. In the in vivo murine eye, we used light-based resonance Raman spectroscopy (RRS) to assess noninvasively the redox states of mitochondria and hemoglobin which reflect availability of electron donors (fuel) and acceptors (oxygen). As proof of principle, we demonstrated that the mitochondrial redox state at the optic nerve head correlates with later retinal ganglion loss after acute intraocular pressure (IOP) elevation. This technology can potentially map the metabolic health of eye tissue in vivo complementary to optical coherence tomography, defining structural changes. Early detection (and normalization) of mitochondrial dysfunction before irreversible damage could lead to prevention of permanent neural loss.
Collapse
Affiliation(s)
- Bertan Cakir
- Department of Ophthalmology, Boston Children’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Yohei Tomita
- Department of Ophthalmology, Boston Children’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Hitomi Yagi
- Department of Ophthalmology, Boston Children’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | | | - William Allen
- Department of Ophthalmology, Boston Children’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Minji Ko
- Department of Ophthalmology, Boston Children’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Peili Chen
- Pendar Technologies, Cambridge, MA 02138, USA
| | - Zhongjie Fu
- Department of Ophthalmology, Boston Children’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | | | - Lois E H Smith
- Department of Ophthalmology, Boston Children’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
19
|
Calbiague García V, Cadiz B, Herrera P, Díaz A, Schmachtenberg O. Evaluation of Photobiomodulation and Boldine as Alternative Treatment Options in Two Diabetic Retinopathy Models. Int J Mol Sci 2023; 24:ijms24097918. [PMID: 37175628 PMCID: PMC10178531 DOI: 10.3390/ijms24097918] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Revised: 04/18/2023] [Accepted: 04/19/2023] [Indexed: 05/15/2023] Open
Abstract
Diabetic retinopathy causes progressive and irreversible damage to the retina through activation of inflammatory processes, overproduction of oxidative species, and glial reactivity, leading to changes in neuronal function and finally ischemia, edema, and hemorrhages. Current treatments are invasive and mostly applied at advanced stages, stressing the need for alternatives. To this end, we tested two unconventional and potentially complementary non-invasive treatment options: Photobiomodulation, the stimulation with near-infrared light, has shown promising results in ameliorating retinal pathologies and insults in several studies but remains controversial. Boldine, on the other hand, is a potent natural antioxidant and potentially useful to prevent free radical-induced oxidative stress. To establish a baseline, we first evaluated the effects of diabetic conditions on the retina with immunofluorescence, histological, and ultrastructural analysis in two diabetes model systems, obese LepRdb/db mice and organotypic retinal explants, and then tested the potential benefits of photobiomodulation and boldine treatment in vitro on retinal explants subjected to high glucose concentrations, mimicking diabetic conditions. Our results suggest that the principal subcellular structures affected by these conditions were mitochondria in the inner segment of photoreceptors, which displayed morphological changes in both model systems. In retinal explants, lactate metabolism, assayed as an indicator of mitochondrial function, was altered, and decreased photoreceptor viability was observed, presumably as a consequence of increased oxidative-nitrosative stress. The latter was reduced by boldine treatment in vitro, while photobiomodulation improved mitochondrial metabolism but was insufficient to prevent retinal structural damage caused by high glucose. These results warrant further research into alternative and complementary treatment options for diabetic retinopathy.
Collapse
Affiliation(s)
- Víctor Calbiague García
- Ph. D. Program in Neuroscience, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso 2360102, Chile
- Centro Interdisciplinario de Neurociencias de Valparaíso (CINV), Facultad de Ciencias, Universidad de Valparaíso, Valparaíso 2360102, Chile
| | - Bárbara Cadiz
- Centro Interdisciplinario de Neurociencias de Valparaíso (CINV), Facultad de Ciencias, Universidad de Valparaíso, Valparaíso 2360102, Chile
| | - Pablo Herrera
- Instituto de Biología, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso 2360102, Chile
| | - Alejandra Díaz
- Centro Interdisciplinario de Neurociencias de Valparaíso (CINV), Facultad de Ciencias, Universidad de Valparaíso, Valparaíso 2360102, Chile
| | - Oliver Schmachtenberg
- Centro Interdisciplinario de Neurociencias de Valparaíso (CINV), Facultad de Ciencias, Universidad de Valparaíso, Valparaíso 2360102, Chile
- Instituto de Biología, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso 2360102, Chile
| |
Collapse
|
20
|
Yaribeygi H, Maleki M, Butler AE, Jamialahmadi T, Sahebkar A. Sodium-glucose cotransporter 2 inhibitors and mitochondrial functions: state of the art. EXCLI JOURNAL 2023; 22:53-66. [PMID: 36814854 PMCID: PMC9939776 DOI: 10.17179/excli2022-5482] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Accepted: 12/21/2022] [Indexed: 02/24/2023]
Abstract
Sodium-glucose cotransporter 2 inhibitors (SGLT2is) are a class of newly introduced antidiabetic drugs with potent hypoglycemic effects. Recent evidence suggests that these drugs have extraglycemic impacts and are therefore able to provide additional benefits beyond glucose lowering. Mitochondrial dysfunction is a central facet of many disorders that negatively impacts many tissues and organs, especially in the setting of diabetes. Therefore, it would be hugely beneficial if an antidiabetic drug could also provide mitochondrial benefits to improve cellular function and reduce the risk of diabetic complications. In this review, we have surveyed the literature for possible mitochondrial benefits of SGLT2is and we discuss the possible mechanisms involved.
Collapse
Affiliation(s)
- Habib Yaribeygi
- Research Center of Physiology, Semnan University of Medical Sciences, Semnan, Iran
| | - Mina Maleki
- Urology and Nephrology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Alexandra E. Butler
- Research Department, Royal College of Surgeons in Ireland Bahrain, Adliya, 15503, Bahrain
| | - Tannaz Jamialahmadi
- Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- Surgical Oncology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Amirhossein Sahebkar
- Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
- School of Medicine, The University of Western Australia, Perth, Australia
- Department of Biotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
21
|
Russell MW, Muste JC, Seth K, Kumar M, Rich CA, Singh RP, Traboulsi EI. Functional imaging of mitochondria in genetically confirmed retinal dystrophies using flavoprotein fluorescence. Ophthalmic Genet 2022; 43:834-840. [PMID: 36384402 DOI: 10.1080/13816810.2022.2144903] [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: 11/18/2022]
Abstract
BACKGROUND Whether by indirect oxidative stress or direct genetic defect, various genetic retinal dystrophies involve mitochondrial stress. Mitochondrial flavoprotein fluorescence (FPF), reported as either average signal intensity or variability (heterogeneity), may serve as a direct, quantifiable marker of oxidative stress. MATERIALS AND METHODS This observational study enrolled patients with genetically confirmed retinal dystrophies between January and December 2021. Patients with concomitant maculopathy and ocular hypertension were excluded. Patients were FPF imaged with OcuMet Beacon® third generation device during routine outpatient visit. RESULTS The final analysis cohort included 242 images from 157 patients. Mean FPF intensity was significantly increased between age matched controls and patients with confirmed rod-cone dystrophy, Stargardt disease, Bardet-Biedl syndrome (BBS), and Mitochondrial ATP synthase mutation (P ≤ 0.007). Mean FPF heterogeneity was significantly increased between age matched controls and patients with confirmed rod-cone dystrophy, Stargardt disease, and BBS (P ≤ 0.011). FPF lesions were noted to correlate with Fundus Autofluorescence (FAF) lesions in diseases examined. CONCLUSIONS FPF intensity and heterogeneity significantly increased in patients with retinal dystrophies. The correlation of FPF lesions with FAF lesions implies FPF may be a clinically useful biomarker in patients with IRDs.
Collapse
Affiliation(s)
- Matthew W Russell
- Center for Ophthalmic Bioinformatics, Cole Eye Institute, Cleveland Clinic, Cleveland, Ohio, USA.,Department of Education, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, Ohio, USA
| | - Justin C Muste
- Center for Ophthalmic Bioinformatics, Cole Eye Institute, Cleveland Clinic, Cleveland, Ohio, USA.,Cleveland Clinic, Cole Eye Institute, Cleveland, Ohio, USA
| | - Kanika Seth
- Center for Ophthalmic Bioinformatics, Cole Eye Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Madhukar Kumar
- Center for Ophthalmic Bioinformatics, Cole Eye Institute, Cleveland Clinic, Cleveland, Ohio, USA.,Department of Education, Case Western Reserve University School of Medicine, Cleveland, Ohio, USA
| | | | - Rishi P Singh
- Center for Ophthalmic Bioinformatics, Cole Eye Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | | |
Collapse
|
22
|
Anand S, Trounce IA, Gangoda L. Role of extracellular vesicles in mitochondrial eye diseases. IUBMB Life 2022; 74:1264-1272. [PMID: 36308309 PMCID: PMC10947567 DOI: 10.1002/iub.2687] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Accepted: 10/14/2022] [Indexed: 03/20/2024]
Abstract
Extracellular vesicles (EVs) are small packages that are released by almost all types of cells. While the role of EVs in pathogenesis of certain diseases such as cancer is well established, EVs role in ocular health and disease is still at early stages of investigation. Given the significant role of EVs in pathological development and progression of diseases such as cancer, EVs present a similar opportunity for investigation in ocular pathophysiology. Studies have shown the presence of EVs in fluids from the ocular environment have close links with ocular health and disease. Hence, the cargo carried in EVs from ocular fluids can be used for monitoring disease phenotypes or therapeutic outcomes in eye-related disorders. Furthermore, in recent times EVs have increasingly gained attention as therapeutics and drug-delivery vehicles for treatment of eye diseases. There is a close relationship between EVs and mitochondria functioning with mitochondria dysfunction leading to a significant number of ophthalmic disorders. This review discusses the current knowledge of EVs in visual systems with a special focus on eye diseases resulting from dysfunctional mitochondria.
Collapse
Affiliation(s)
- Sushma Anand
- Centre for Eye Research AustraliaRoyal Victorian Eye and Ear HospitalEast MelbourneVictoriaAustralia
- Opthalmology, Department of SurgeryUniversity of MelbourneMelbourneVictoriaAustralia
| | - Ian A. Trounce
- Centre for Eye Research AustraliaRoyal Victorian Eye and Ear HospitalEast MelbourneVictoriaAustralia
- Opthalmology, Department of SurgeryUniversity of MelbourneMelbourneVictoriaAustralia
| | - Lahiru Gangoda
- Centre for Eye Research AustraliaRoyal Victorian Eye and Ear HospitalEast MelbourneVictoriaAustralia
- Opthalmology, Department of SurgeryUniversity of MelbourneMelbourneVictoriaAustralia
- The Walter and Eliza Hall Institute of Medical Research (WEHI)MelbourneVictoriaAustralia
- Department of Medical BiologyUniversity of MelbourneMelbourneVictoriaAustralia
| |
Collapse
|
23
|
Noh SE, Lee SJ, Lee TG, Park KS, Kim JH. Inhibition of Cellular Senescence Hallmarks by Mitochondrial Transplantation in Senescence-induced ARPE-19 cells. Neurobiol Aging 2022; 121:157-165. [DOI: 10.1016/j.neurobiolaging.2022.11.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Revised: 10/03/2022] [Accepted: 11/01/2022] [Indexed: 11/09/2022]
|
24
|
Galindez SM, Keightley A, Koulen P. Differential distribution of steroid hormone signaling networks in the human choroid-retinal pigment epithelial complex. BMC Ophthalmol 2022; 22:406. [PMID: 36266625 PMCID: PMC9583547 DOI: 10.1186/s12886-022-02585-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Accepted: 08/22/2022] [Indexed: 11/10/2022] Open
Abstract
Background The retinal pigment epithelium (RPE), a layer of pigmented cells that lies between the neurosensory retina and the underlying choroid, plays a critical role in maintaining the functional integrity of photoreceptor cells and in mediating communication between the neurosensory retina and choroid. Prior studies have demonstrated neurotrophic effects of select steroids that mitigate the development and progression of retinal degenerative diseases via an array of distinct mechanisms of action. Methods Here, we identified major steroid hormone signaling pathways and their key functional protein constituents controlling steroid hormone signaling, which are potentially involved in the mitigation or propagation of retinal degenerative processes, from human proteome datasets with respect to their relative abundances in the retinal periphery, macula, and fovea. Results Androgen, glucocorticoid, and progesterone signaling networks were identified and displayed differential distribution patterns within these three anatomically distinct regions of the choroid-retinal pigment epithelial complex. Classical and non-classical estrogen and mineralocorticoid receptors were not identified. Conclusion Identified differential distribution patterns suggest both selective susceptibility to chronic neurodegenerative disease processes, as well as potential substrates for drug target discovery and novel drug development focused on steroid signaling pathways in the choroid-RPE.
Collapse
Affiliation(s)
- Sydney M Galindez
- School of Medicine, Vision Research Center, Department of Ophthalmology, University of Missouri - Kansas City School of Medicine, 2411 Holmes St, Kansas City, MO, 64108, USA
| | - Andrew Keightley
- School of Medicine, Vision Research Center, Department of Ophthalmology, University of Missouri - Kansas City School of Medicine, 2411 Holmes St, Kansas City, MO, 64108, USA
| | - Peter Koulen
- School of Medicine, Vision Research Center, Department of Ophthalmology, University of Missouri - Kansas City School of Medicine, 2411 Holmes St, Kansas City, MO, 64108, USA. .,Department of Biomedical Sciences, University of Missouri - Kansas City School of Medicine, Kansas City, MO, USA.
| |
Collapse
|
25
|
Wang J, Li M, Geng Z, Khattak S, Ji X, Wu D, Dang Y. Role of Oxidative Stress in Retinal Disease and the Early Intervention Strategies: A Review. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:7836828. [PMID: 36275903 PMCID: PMC9586758 DOI: 10.1155/2022/7836828] [Citation(s) in RCA: 43] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Revised: 08/05/2022] [Accepted: 08/11/2022] [Indexed: 02/05/2023]
Abstract
The retina, owing to its cellular anatomy and physical location, is susceptible to generating reactive oxygen species (ROS), which are associated with several major retinal diseases. When ROS exceeds the body's natural antioxidants, the retina is in a state of oxidative stress, which is recognized as the pathogenesis of retinal diseases. The early stage of the pathogenic process is an adaptive change in which oxidative stress and endogenous defense mechanisms occur. If no treatment is applied, the retinal diseases will progress to the pathological stage with neuronal and vascular dysfunction or damage and even blindness. This review summarizes the role of oxidative stress in several common retinal diseases, including retinitis pigmentosa, age-related macular degeneration, diabetic retinopathy, glaucoma, and retinopathy of prematurity. In addition, we discuss the early intervention strategies for these diseases. An outline is provided to identify potential intervention targets for further research. Early intervention for retinal diseases is necessary and urgent and may offer hope to improve patients' quality of life through functional vision.
Collapse
Affiliation(s)
- Jun Wang
- School of Basic Medical Sciences, Henan University, Kaifeng, China
- Henan International Joint Laboratory for Nuclear Protein Regulation, Henan University, Kaifeng, China
| | - Mengling Li
- College of Acu-Moxibustion and Massage, Shaanxi University of Chinese Medicine, Xianyang, China
| | - Ziyue Geng
- School of Clinical Medicine, Henan University, Kaifeng, Henan, China
| | - Saadullah Khattak
- Henan International Joint Laboratory for Nuclear Protein Regulation, Henan University, Kaifeng, China
| | - Xinying Ji
- Henan International Joint Laboratory for Nuclear Protein Regulation, Henan University, Kaifeng, China
| | - Dongdong Wu
- Henan International Joint Laboratory for Nuclear Protein Regulation, Henan University, Kaifeng, China
| | - Yalong Dang
- Henan International Joint Laboratory for Nuclear Protein Regulation, Henan University, Kaifeng, China
- Sanmenxia Central Hospital, Sanmenxia, Henan, China
| |
Collapse
|
26
|
Mauri S, Favaro M, Bernardo G, Mazzotta GM, Ziviani E. Mitochondrial autophagy in the sleeping brain. Front Cell Dev Biol 2022; 10:956394. [PMID: 36092697 PMCID: PMC9449320 DOI: 10.3389/fcell.2022.956394] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Accepted: 07/13/2022] [Indexed: 11/13/2022] Open
Abstract
A significant percentage of the mitochondrial mass is replaced on a daily basis via mechanisms of mitochondrial quality control. Through mitophagy (a selective type of autophagy that promotes mitochondrial proteostasis) cells keep a healthy pool of mitochondria, and prevent oxidative stress and inflammation. Furthermore, mitophagy helps adapting to the metabolic demand of the cells, which changes on a daily basis.Core components of the mitophagy process are PINK1 and Parkin, which mutations are linked to Parkinson’s Disease. The crucial role of PINK1/Parkin pathway during stress-induced mitophagy has been extensively studied in vitro in different cell types. However, recent advances in the field allowed discovering that mitophagy seems to be only slightly affected in PINK1 KO mice and flies, putting into question the physiological relevance of this pathway in vivo in the whole organism. Indeed, several cell-specific PINK1/Parkin-independent mitophagy pathways have been recently discovered, which appear to be activated under physiological conditions such as those that promote mitochondrial proteome remodeling during differentiation or in response to specific physiological stimuli.In this Mini Review we want to summarize the recent advances in the field, and add another level of complexity by focusing attention on a potentially important aspect of mitophagy regulation: the implication of the circadian clock. Recent works showed that the circadian clock controls many aspects of mitochondrial physiology, including mitochondrial morphology and dynamic, respiratory activity, and ATP synthesis. Furthermore, one of the essential functions of sleep, which is controlled by the clock, is the clearance of toxic metabolic compounds from the brain, including ROS, via mechanisms of proteostasis. Very little is known about a potential role of the clock in the quality control mechanisms that maintain the mitochondrial repertoire healthy during sleep/wake cycles. More importantly, it remains completely unexplored whether (dys)function of mitochondrial proteostasis feedbacks to the circadian clockwork.
Collapse
Affiliation(s)
| | | | | | | | - Elena Ziviani
- *Correspondence: Gabriella M. Mazzotta, Elena Ziviani,
| |
Collapse
|
27
|
Shao Z, Chwa M, Atilano SR, Park J, Karageozian H, Karageozian V, Kenney MC. The Transcriptome Profile of Retinal Pigment Epithelium and Müller Cell Lines Protected by Risuteganib Against Hydrogen Peroxide Stress. J Ocul Pharmacol Ther 2022; 38:513-526. [PMID: 35731128 PMCID: PMC9508878 DOI: 10.1089/jop.2022.0015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022] Open
Abstract
Purpose: Oxidative stress contributes to the pathogenesis of vision-impairing diseases. In the retina, retinal pigment epithelium (RPE) and Müller cells support neuronal homeostasis, but also contribute to pathological development under stressed conditions. Recent studies found that the investigational drug risuteganib (RSG) has a good safety profile, provided protection in experimental models, and improved visual acuity in patients. The present in vitro study evaluated the effects of RSG in RPE and Müller cell lines stressed with the oxidant hydrogen peroxide (H2O2). Methods: Human RPE (ARPE-19) and Müller (MIO-M1) cell lines were treated with various combinations of RSG and H2O2. Trypan blue assay was used to investigate the effect of compounds on cell viability. Gene expression was measured using RNA sequencing to identify regulated genes and the biological processes and pathways involved. Results: Trypan blue assay found RSG pre-treatment significantly protected against H2O2-induced cell death in ARPE-19 and MIO-M1 cells. Transcriptome analysis found H2O2 regulated genes in several disease-relevant biological processes, including cell adhesion, migration, death, and proliferation; ECM organization; angiogenesis; metabolism; and immune system processes. RSG pre-treatment modulated these gene expression profiles in the opposite direction of H2O2. Pathway analysis found genes in integrin, AP-1, and syndecan signaling pathways were regulated. Expression of selected RSG-regulated genes was validated using qRT-PCR. Conclusions: RSG protected cultured human RPE and Müller cell lines against H2O2-induced cell death and mitigated the associated transcriptome changes in biological processes and pathways relevant to the pathogenesis of retinal diseases. These results demonstrate RSG reduced oxidative stress-induced toxicity in two retinal cell lines with potential relevance to the treatment of human diseases.
Collapse
Affiliation(s)
- Zixuan Shao
- Allegro Ophthalmics, LLC, San Juan Capistrano, California, USA
| | - Marilyn Chwa
- Gavin Herbert Eye Institute, University of California Irvine, Irvine, California, USA
| | - Shari R Atilano
- Gavin Herbert Eye Institute, University of California Irvine, Irvine, California, USA
| | - John Park
- Allegro Ophthalmics, LLC, San Juan Capistrano, California, USA
| | | | | | - M Cristina Kenney
- Gavin Herbert Eye Institute, University of California Irvine, Irvine, California, USA.,Department of Pathology and Laboratory Medicine, University of California Irvine, Irvine, California, USA
| |
Collapse
|
28
|
Tools and Biomarkers for the Study of Retinal Ganglion Cell Degeneration. Int J Mol Sci 2022; 23:ijms23084287. [PMID: 35457104 PMCID: PMC9025234 DOI: 10.3390/ijms23084287] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Revised: 04/03/2022] [Accepted: 04/08/2022] [Indexed: 11/17/2022] Open
Abstract
The retina is part of the central nervous system, its analysis may provide an idea of the health and functionality, not only of the retina, but also of the entire central nervous system, as has been shown in Alzheimer’s or Parkinson’s diseases. Within the retina, the ganglion cells (RGC) are the neurons in charge of processing and sending light information to higher brain centers. Diverse insults and pathological states cause degeneration of RGC, leading to irreversible blindness or impaired vision. RGCs are the measurable endpoints in current research into experimental therapies and diagnosis in multiple ocular pathologies, like glaucoma. RGC subtype classifications are based on morphological, functional, genetical, and immunohistochemical aspects. Although great efforts are being made, there is still no classification accepted by consensus. Moreover, it has been observed that each RGC subtype has a different susceptibility to injury. Characterizing these subtypes together with cell death pathway identification will help to understand the degenerative process in the different injury and pathological models, and therefore prevent it. Here we review the known RGC subtypes, as well as the diagnostic techniques, probes, and biomarkers for programmed and unprogrammed cell death in RGC.
Collapse
|
29
|
Heckel E, Cagnone G, Agnihotri T, Cakir B, Das A, Kim JS, Kim N, Lavoie G, Situ A, Pundir S, Sun Y, Wünnemann F, Pierce KA, Dennis C, Mitchell GA, Chemtob S, Rezende FA, Andelfinger G, Clish CB, Roux PP, Sapieha P, Smith LE, Joyal JS. Triglyceride-derived fatty acids reduce autophagy in a model of retinal angiomatous proliferation. JCI Insight 2022; 7:e154174. [PMID: 35167498 PMCID: PMC8986067 DOI: 10.1172/jci.insight.154174] [Citation(s) in RCA: 6] [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: 08/16/2021] [Accepted: 02/09/2022] [Indexed: 11/17/2022] Open
Abstract
Dyslipidemia and autophagy have been implicated in the pathogenesis of blinding neovascular age-related macular degeneration (NV-AMD). VLDL receptor (VLDLR), expressed in photoreceptors with a high metabolic rate, facilitates the uptake of triglyceride-derived fatty acids. Since fatty acid uptake is reduced in Vldlr-/- tissues, more remain in circulation, and the retina is fuel deficient, driving the formation in mice of neovascular lesions reminiscent of retinal angiomatous proliferation (RAP), a subtype of NV-AMD. Nutrient scarcity and energy failure are classically mitigated by increasing autophagy. We found that excess circulating lipids restrained retinal autophagy, which contributed to pathological angiogenesis in the Vldlr-/- RAP model. Triglyceride-derived fatty acid sensed by free fatty acid receptor 1 (FFAR1) restricted autophagy and oxidative metabolism in photoreceptors. FFAR1 suppressed transcription factor EB (TFEB), a master regulator of autophagy and lipid metabolism. Reduced TFEB, in turn, decreased sirtuin-3 expression and mitochondrial respiration. Metabolomic signatures of mouse RAP-like retinas were consistent with a role in promoting angiogenesis. This signature was also found in human NV-AMD vitreous. Restoring photoreceptor autophagy in Vldlr-/- retinas, either pharmacologically or by deleting Ffar1, enhanced metabolic efficiency and suppressed pathological angiogenesis. Dysregulated autophagy by circulating lipids might therefore contribute to the energy failure of photoreceptors driving neovascular eye diseases, and FFAR1 may be a target for intervention.
Collapse
Affiliation(s)
- Emilie Heckel
- Department of Pharmacology, University of Montreal, Montreal, Quebec, Canada
| | - Gael Cagnone
- Department of Pharmacology, University of Montreal, Montreal, Quebec, Canada
| | - Tapan Agnihotri
- Department of Pharmacology and Therapeutics, McGill University, Montreal, Quebec, Canada
| | - Bertan Cakir
- Department of Ophthalmology, Boston Children’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Ashim Das
- Department of Pharmacology, University of Montreal, Montreal, Quebec, Canada
| | - Jin Sung Kim
- Department of Pharmacology and Therapeutics, McGill University, Montreal, Quebec, Canada
| | - Nicholas Kim
- Department of Pharmacology and Therapeutics, McGill University, Montreal, Quebec, Canada
| | - Geneviève Lavoie
- Department of Pathology and Cell Biology, Institute for Research in Immunology and Cancer (IRIC), and
| | - Anu Situ
- Department of Pediatrics, University of Montreal, Montreal, Quebec, Canada
| | - Sheetal Pundir
- Department of Pharmacology and Therapeutics, McGill University, Montreal, Quebec, Canada
| | - Ye Sun
- Department of Ophthalmology, Boston Children’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Florian Wünnemann
- Department of Pediatrics, University of Montreal, Montreal, Quebec, Canada
| | - Kerry A. Pierce
- Metabolomics Platform, Broad Institute of MIT and Harvard University, Cambridge, Massachusetts, USA
| | - Courtney Dennis
- Metabolomics Platform, Broad Institute of MIT and Harvard University, Cambridge, Massachusetts, USA
| | - Grant A. Mitchell
- Department of Pediatrics, University of Montreal, Montreal, Quebec, Canada
| | - Sylvain Chemtob
- Department of Pharmacology, University of Montreal, Montreal, Quebec, Canada
- Department of Pharmacology and Therapeutics, McGill University, Montreal, Quebec, Canada
- Department of Pediatrics, University of Montreal, Montreal, Quebec, Canada
- Department of Ophthalmology, University of Montreal, Montreal, Quebec, Canada
| | - Flavio A. Rezende
- Department of Ophthalmology, University of Montreal, Montreal, Quebec, Canada
| | - Gregor Andelfinger
- Department of Pediatrics, University of Montreal, Montreal, Quebec, Canada
| | - Clary B. Clish
- Metabolomics Platform, Broad Institute of MIT and Harvard University, Cambridge, Massachusetts, USA
| | - Philippe P. Roux
- Department of Pathology and Cell Biology, Institute for Research in Immunology and Cancer (IRIC), and
| | - Przemyslaw Sapieha
- Department of Ophthalmology, University of Montreal, Montreal, Quebec, Canada
| | - Lois E.H. Smith
- Department of Ophthalmology, Boston Children’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Jean-Sébastien Joyal
- Department of Pharmacology, University of Montreal, Montreal, Quebec, Canada
- Department of Pharmacology and Therapeutics, McGill University, Montreal, Quebec, Canada
- Department of Pediatrics, University of Montreal, Montreal, Quebec, Canada
- Department of Ophthalmology, University of Montreal, Montreal, Quebec, Canada
| |
Collapse
|
30
|
Chiang MC, Chern E. Current Development, Obstacle and Futural Direction of Induced Pluripotent Stem Cell and Mesenchymal Stem Cell Treatment in Degenerative Retinal Disease. Int J Mol Sci 2022; 23:ijms23052529. [PMID: 35269671 PMCID: PMC8910526 DOI: 10.3390/ijms23052529] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Revised: 02/20/2022] [Accepted: 02/23/2022] [Indexed: 11/26/2022] Open
Abstract
Degenerative retinal disease is one of the major causes of vision loss around the world. The past several decades have witnessed emerging development of stem cell treatment for retinal disease. Nevertheless, sourcing stem cells remains controversial due to ethical concerns and their rarity. Furthermore, induced pluripotent stem cells (iPSCs) and mesenchymal stem cells (MSCs) are both isolated from patients’ mature tissues; thus, issues such as avoiding moral controversy and adverse events related to immunosuppression and obtaining a large number of cells have opened a new era in regenerative medicine. This review focuses on the current application and development, clinical trials, and latest research of stem cell therapy, as well as its limitations and future directions.
Collapse
|
31
|
Clementi ME, Pizzoferrato M, Bianchetti G, Brancato A, Sampaolese B, Maulucci G, Tringali G. Cytoprotective Effect of Idebenone through Modulation of the Intrinsic Mitochondrial Pathway of Apoptosis in Human Retinal Pigment Epithelial Cells Exposed to Oxidative Stress Induced by Hydrogen Peroxide. Biomedicines 2022; 10:biomedicines10020503. [PMID: 35203712 PMCID: PMC8962354 DOI: 10.3390/biomedicines10020503] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Revised: 02/17/2022] [Accepted: 02/18/2022] [Indexed: 02/04/2023] Open
Abstract
Idebenone is a ubiquinone short-chain synthetic analog with antioxidant properties, which is believed to restore mitochondrial ATP synthesis. As such, idebenone is investigated in numerous clinical trials for diseases of mitochondrial aetiology and it is authorized as a drug for the treatment of Leber’s hereditary optic neuropathy. Mitochondria of retinal pigment epithelium (RPE) are particularly vulnerable to oxidative damage associated with cellular senescence. Therefore, the aim of this study was to explore idebenone’s cytoprotective effect and its underlying mechanism. We used a human-RPE cell line (ARPE-19) exposed to idebenone pre-treatment for 24 h followed by conditions inducing H2O2 oxidative damage for a further 24 h. We found that idebenone: (a) ameliorated H2O2-lowered cell viability in the RPE culture; (b) activated Nrf2 signaling pathway by promoting Nrf2 nuclear translocation; (c) increased Bcl-2 protein levels, leaving unmodified those of Bax, thereby reducing the Bax/Bcl-2 ratio; (d) maintained the mitochondrial membrane potential (ΔΨm) at physiological levels, preserving the functionality of mitochondrial respiratory complexes and counteracting the excessive production of ROS; and (e) reduced mitochondrial cytochrome C-mediated caspase-3 activity. Taken together, our findings show that idebenone protects RPE from oxidative damage by modulating the intrinsic mitochondrial pathway of apoptosis, suggesting its possible role in retinal epitheliopathies associated with mitochondrial dysfunction.
Collapse
Affiliation(s)
- Maria Elisabetta Clementi
- Institute of Chemical Sciences and Technologies “Giulio Natta” (SCITEC)-CNR, Largo F. Vito 1, 00168 Rome, Italy; (M.E.C.); (B.S.)
| | - Michela Pizzoferrato
- Pharmacology Section, Department of Health Care Surveillance and Bioethics, Università Cattolica del Sacro Cuore, Largo F. Vito 1, 00168 Rome, Italy;
- Fondazione Policlinico Universitario Agostino Gemelli IRCSS, 00168 Rome, Italy; (G.B.); (G.M.)
| | - Giada Bianchetti
- Fondazione Policlinico Universitario Agostino Gemelli IRCSS, 00168 Rome, Italy; (G.B.); (G.M.)
- Biophysics Section, Neuroscience Department, Università Cattolica Del Sacro Cuore, Largo F. Vito 1, 00168 Rome, Italy
| | - Anna Brancato
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties of Excellence “G. D’Alessandro”, University of Palermo, 90127 Palermo, Italy;
| | - Beatrice Sampaolese
- Institute of Chemical Sciences and Technologies “Giulio Natta” (SCITEC)-CNR, Largo F. Vito 1, 00168 Rome, Italy; (M.E.C.); (B.S.)
| | - Giuseppe Maulucci
- Fondazione Policlinico Universitario Agostino Gemelli IRCSS, 00168 Rome, Italy; (G.B.); (G.M.)
- Biophysics Section, Neuroscience Department, Università Cattolica Del Sacro Cuore, Largo F. Vito 1, 00168 Rome, Italy
| | - Giuseppe Tringali
- Pharmacology Section, Department of Health Care Surveillance and Bioethics, Università Cattolica del Sacro Cuore, Largo F. Vito 1, 00168 Rome, Italy;
- Biophysics Section, Neuroscience Department, Università Cattolica Del Sacro Cuore, Largo F. Vito 1, 00168 Rome, Italy
- Correspondence: ; Tel.: +39-063-015-4367
| |
Collapse
|
32
|
Shahandeh A, Bui BV, Finkelstein DI, Nguyen CTO. Effects of Excess Iron on the Retina: Insights From Clinical Cases and Animal Models of Iron Disorders. Front Neurosci 2022; 15:794809. [PMID: 35185447 PMCID: PMC8851357 DOI: 10.3389/fnins.2021.794809] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Accepted: 12/17/2021] [Indexed: 01/19/2023] Open
Abstract
Iron plays an important role in a wide range of metabolic pathways that are important for neuronal health. Excessive levels of iron, however, can promote toxicity and cell death. An example of an iron overload disorder is hemochromatosis (HH) which is a genetic disorder of iron metabolism in which the body’s ability to regulate iron absorption is altered, resulting in iron build-up and injury in several organs. The retina was traditionally assumed to be protected from high levels of systemic iron overload by the blood-retina barrier. However, recent data shows that expression of genes that are associated with HH can disrupt retinal iron metabolism. Thus, the effects of iron overload on the retina have become an area of research interest, as excessively high levels of iron are implicated in several retinal disorders, most notably age–related macular degeneration. This review is an effort to highlight risk factors for excessive levels of systemic iron build-up in the retina and its potential impact on the eye health. Information is integrated across clinical and preclinical animal studies to provide insights into the effects of systemic iron loading on the retina.
Collapse
Affiliation(s)
- Ali Shahandeh
- Department of Optometry and Vision Sciences, Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, Parkville, VIC, Australia
| | - Bang V. Bui
- Department of Optometry and Vision Sciences, Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, Parkville, VIC, Australia
| | - David I. Finkelstein
- Florey Department of Neuroscience and Mental Health, University of Melbourne, Parkville, VIC, Australia
| | - Christine T. O. Nguyen
- Department of Optometry and Vision Sciences, Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, Parkville, VIC, Australia
- *Correspondence: Christine T. O. Nguyen,
| |
Collapse
|
33
|
Role of Creatine Supplementation in Conditions Involving Mitochondrial Dysfunction: A Narrative Review. Nutrients 2022; 14:nu14030529. [PMID: 35276888 PMCID: PMC8838971 DOI: 10.3390/nu14030529] [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: 12/14/2021] [Revised: 01/24/2022] [Accepted: 01/24/2022] [Indexed: 12/14/2022] Open
Abstract
Creatine monohydrate (CrM) is one of the most widely used nutritional supplements among active individuals and athletes to improve high-intensity exercise performance and training adaptations. However, research suggests that CrM supplementation may also serve as a therapeutic tool in the management of some chronic and traumatic diseases. Creatine supplementation has been reported to improve high-energy phosphate availability as well as have antioxidative, neuroprotective, anti-lactatic, and calcium-homoeostatic effects. These characteristics may have a direct impact on mitochondrion's survival and health particularly during stressful conditions such as ischemia and injury. This narrative review discusses current scientific evidence for use or supplemental CrM as a therapeutic agent during conditions associated with mitochondrial dysfunction. Based on this analysis, it appears that CrM supplementation may have a role in improving cellular bioenergetics in several mitochondrial dysfunction-related diseases, ischemic conditions, and injury pathology and thereby could provide therapeutic benefit in the management of these conditions. However, larger clinical trials are needed to explore these potential therapeutic applications before definitive conclusions can be drawn.
Collapse
|
34
|
Olchawa MM, Herrnreiter AM, Skumatz CMB, Krzysztynska-Kuleta OI, Mokrzynski KT, Burke JM, Sarna TJ. The Inhibitory Effect of Blue Light on Phagocytic Activity by ARPE-19 Cells. Photochem Photobiol 2022; 98:1110-1121. [PMID: 35067943 DOI: 10.1111/php.13596] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Revised: 01/12/2022] [Accepted: 01/18/2022] [Indexed: 11/30/2022]
Abstract
Chronic exposure of the retina to short wavelength visible light is a risk factor in pathogenesis of age-related macular degeneration. The proper functioning and survival of photoreceptors depends on efficient phagocytosis of photoreceptor outer segments (POS) by retinal pigment epithelium. The purpose of this study was to analyze the phagocytic activity of blue light-treated ARPE-19 cells, and to examine whether the observed effects could be related to altered levels of POS phagocytosis receptor proteins and/or to oxidation of cellular proteins and lipids. POS phagocytosis was measured by flow cytometry. Phagocytosis receptor proteins αv and β5 integrin subunits and Mer tyrosine kinase (MerTK) were quantified by western blotting. The intact functional heterodimer αvβ5 was quantified by immunoprecipitation followed by immunoblotting. Cellular protein and lipid hydroperoxides were analyzed by coumarin boronic acid probe and iodometric assay, respectively. Cell irradiation induced reversible inhibition of specific phagocytosis and transient reductions in phagocytosis receptor proteins. Full recovery of functional heterodimer was apparent. Significant photooxidation of cellular proteins and lipids was observed. The results indicate that transient inhibition of specific phagocytosis by blue light could be related to the reduction in phagocytosis receptor proteins. Such changes may arise from oxidative modifications of cell phagocytic machinery components.
Collapse
Affiliation(s)
- Magdalena M Olchawa
- Department of Biophysics, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, 30-387, Krakow, Poland.,Department of Ophthalmology, Eye Institute, Medical College of Wisconsin, Milwaukee, WI, 53226, USA
| | - Anja M Herrnreiter
- Department of Ophthalmology, Eye Institute, Medical College of Wisconsin, Milwaukee, WI, 53226, USA.,Department of Pharmacology and Toxicology, Medical College of Wisconsin, Milwaukee, WI, 53226, USA
| | - Christine M B Skumatz
- Department of Ophthalmology, Eye Institute, Medical College of Wisconsin, Milwaukee, WI, 53226, USA.,Department of Ophthalmology and Visual Sciences, Eye Institute, Medical College of Wisconsin, Milwaukee, WI, 53226, USA
| | - Olga I Krzysztynska-Kuleta
- Department of Biophysics, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, 30-387, Krakow, Poland
| | - Krystian T Mokrzynski
- Department of Biophysics, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, 30-387, Krakow, Poland
| | - Janice M Burke
- Department of Ophthalmology, Eye Institute, Medical College of Wisconsin, Milwaukee, WI, 53226, USA.,Emeritus Professor of Ophthalmology
| | - Tadeusz J Sarna
- Department of Biophysics, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, 30-387, Krakow, Poland
| |
Collapse
|
35
|
Shukal DK, Malaviya PB, Sharma T. Role of the AMPK signalling pathway in the aetiopathogenesis of ocular diseases. Hum Exp Toxicol 2022; 41:9603271211063165. [PMID: 35196887 DOI: 10.1177/09603271211063165] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
BACKGROUND AMP-activated protein kinase (AMPK) plays a precise role as a master regulator of cellular energy homeostasis. AMPK is activated in response to the signalling cues that exhaust cellular ATP levels such as hypoxia, ischaemia, glucose depletion and heat shock. As a central regulator of both lipid and glucose metabolism, AMPK is considered to be a potential therapeutic target for the treatment of various diseases, including eye disorders. OBJECTIVE To review all the shreds of evidence concerning the role of the AMPK signalling pathway in the pathogenesis of ocular diseases. METHOD Scientific data search and review of available information evaluating the influence of AMPK signalling on ocular diseases. RESULTS Review highlights the significance of AMPK signalling in the aetiopathogenesis of ocular diseases, including cataract, glaucoma, diabetic retinopathy, retinoblastoma, age-related macular degeneration, corneal diseases, etc. The review also provides the information on the AMPK-associated pathways with reference to ocular disease, which includes mitochondrial biogenesis, autophagy and regulation of inflammatory response. CONCLUSION The study concludes the role of AMPK in ocular diseases. There is growing interest in the therapeutic utilization of the AMPK pathway for ocular disease treatment. Furthermore, inhibition of AMPK signalling might represent more pertinent strategy than AMPK activation for ocular disease treatment. Such information will guide the development of more effective AMPK modulators for ocular diseases.[Formula: see text].
Collapse
Affiliation(s)
- Dhaval K Shukal
- 534329Iladevi Cataract and IOL Research Centre, Memnagar, Ahmedabad, Gujarat, India.,76793Manipal Academy of Higher Education, Mangalore, Karnataka, India
| | - Pooja B Malaviya
- 534329Iladevi Cataract and IOL Research Centre, Memnagar, Ahmedabad, Gujarat, India.,76793Manipal Academy of Higher Education, Mangalore, Karnataka, India
| | - Tusha Sharma
- 534329Iladevi Cataract and IOL Research Centre, Memnagar, Ahmedabad, Gujarat, India
| |
Collapse
|
36
|
Mitochondria dynamics in the aged mice eye and the role in the RPE phagocytosis. Exp Eye Res 2021; 213:108800. [PMID: 34688622 DOI: 10.1016/j.exer.2021.108800] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Revised: 09/30/2021] [Accepted: 10/19/2021] [Indexed: 12/15/2022]
Abstract
Aging is a predominant risk factor for various eye diseases. Age-related macular degeneration (AMD) is the leading cause of blindness in the elderly, and its etiology remains unclear. Fragmented and dysfunctional mitochondria are associated with age-related diseases. The retinal pigment epithelium (RPE), a polarized cell layer that functions in visual pigment recycling and degeneration, is suspected as the primary region site of AMD. In the present study, we investigated the relationship between mitochondrial dysfunction and RPE aging. Compared to young mice, aged pigmented mice (C57BL/6J, 12-month-old) exhibit decreased visual function without retinal thinning. Consistently, the rhodopsin expression level decreased in the outer segment of aged mice. Moreover, the cell volume of the RPE increased in aged animals. Interestingly, the expression of mitochondria dynamics-related proteins, including Drp1, was altered in the RPE-choroid complex but not in the neural retina after aging. Electron microscopy revealed that mitochondrial size decreased and cristae width increased in aged RPE. The photoreceptor outer segment (POS) treatment of ARPE-19 cells causes Drp1 activation. Furthermore, pharmacological suppression of mitochondrial fission improved the phagocytosis of the POS. These findings indicate that mitochondrial dysfunction and fission in RPE impede phagocytosis and cause retardation of the visual cycle, which can be one of the age-related defects in the retina that may contribute to the onset of AMD.
Collapse
|
37
|
Prokai-Tatrai K, Zaman K, Nguyen V, De La Cruz DL, Prokai L. Proteomics-Based Retinal Target Engagement Analysis and Retina-Targeted Delivery of 17β-Estradiol by the DHED Prodrug for Ocular Neurotherapy in Males. Pharmaceutics 2021; 13:1392. [PMID: 34575465 PMCID: PMC8466286 DOI: 10.3390/pharmaceutics13091392] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Revised: 08/26/2021] [Accepted: 08/27/2021] [Indexed: 12/21/2022] Open
Abstract
We examined the impact of 17β-estradiol (E2) eye drops on the modulation of the proteome profile in the male rat retina. With discovery-driven proteomics, we have identified proteins that were regulated by our treatment. These proteins were assembled to several bioinformatics-based networks implicating E2's beneficial effects on the male rat retina in a broad context of ocular neuroprotection including the maintenance of retinal homeostasis, facilitation of efficient disposal of damaged proteins, and mitochondrial respiratory chain biogenesis. We have also shown for the first time that the hormone's beneficial effects on the male retina can be constrained to this target site by treatment with the bioprecursor prodrug, DHED. A large concentration of E2 was produced after DHED eye drops not only in male rat retinae but also in those of rabbits. However, DHED treatment did not increase circulating E2 levels, thereby ensuring therapeutic safety in males. Targeted proteomics focusing on selected biomarkers of E2's target engagement further confirmed the prodrug's metabolism to E2 in the male retina and indicated that the retinal impact of DHED treatment was identical to that of the direct E2 treatment. Altogether, our study shows the potential of topical DHED therapy for an efficacious and safe protection of the male retina without the unwanted hormonal side-effects associated with current estrogen therapies.
Collapse
Affiliation(s)
- Katalin Prokai-Tatrai
- Department of Pharmacology and Neuroscience, University of North Texas Health Science Center, Fort Worth, TX 76107, USA; (K.Z.); (V.N.); (D.L.D.L.C.); (L.P.)
| | | | | | | | | |
Collapse
|
38
|
Min J, Zeng T, Roux M, Lazar D, Chen L, Tudzarova S. The Role of HIF1α-PFKFB3 Pathway in Diabetic Retinopathy. J Clin Endocrinol Metab 2021; 106:2505-2519. [PMID: 34019671 PMCID: PMC8372643 DOI: 10.1210/clinem/dgab362] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/07/2021] [Indexed: 12/13/2022]
Abstract
Diabetic retinopathy (DR) is the leading cause of blindness for adults in developed countries. Both microvasculopathy and neurodegeneration are implicated in mechanisms of DR development, with neuronal impairment preceding microvascular abnormalities, which is often underappreciated in the clinic. Most current therapeutic strategies, including anti-vascular endothelial growth factor (anti-VEGF)-antibodies, aim at treating the advanced stages (diabetic macular edema and proliferative diabetic retinopathy) and fail to target the neuronal deterioration. Hence, new therapeutic approach(es) intended to address both vascular and neuronal impairment are urgently needed. The hypoxia-inducible factor 1α (HIF1α)-6-phosphofructo-2-kinase/fructose-2,6-bisphosphatase 3 (PFKFB3) pathway is critically implicated in the islet pathology of diabetes. Recent evidence highlighted the pathway relevance for pathologic angiogenesis and neurodegeneration, two key aspects in DR. PFKFB3 is key to the sprouting angiogenesis, along with VEGF, by determining the endothelial tip-cell competition. Also, PFKFB3-driven glycolysis compromises the antioxidative capacity of neurons leading to neuronal loss and reactive gliosis. Therefore, the HIF1α-PFKFB3 signaling pathway is unique as being a pervasive pathological component across multiple cell types in the retina in the early as well as late stages of DR. A metabolic point-of-intervention based on HIF1α-PFKFB3 targeting thus deserves further consideration in DR.
Collapse
Affiliation(s)
- Jie Min
- Department of Endocrinology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Larry L. Hillblom Islet Research Center, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | - Tianshu Zeng
- Department of Endocrinology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Margaretha Roux
- Groote Schuur and Red Cross Children’s Hospital, University of Cape Town, South Africa
| | - David Lazar
- Lazar Retina Ophthalmology, Los Angeles, CA, USA
| | - Lulu Chen
- Department of Endocrinology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Lulu Chen, PhD, Department of Endocrinology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Ave, Wuhan, Hubei, 430022, China.
| | - Slavica Tudzarova
- Larry L. Hillblom Islet Research Center, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
- Correspondence: Slavica Tudzarova, PhD, Larry Hillblom Islet Research Center, University of California Los Angeles, 10833 Le Conte Ave, CHS 33-165, Los Angeles, CA 90095, USA.
| |
Collapse
|
39
|
Kador PF, Salvi R. Multifunctional Redox Modulators Protect Auditory, Visual, and Cognitive Function. Antioxid Redox Signal 2021; 36:1136-1157. [PMID: 34162214 PMCID: PMC9221172 DOI: 10.1089/ars.2021.0129] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Accepted: 06/18/2021] [Indexed: 12/26/2022]
Abstract
Significance: Oxidative stress contributes to vision, hearing and neurodegenerative disorders. Currently, no treatments prevent these disorders; therefore, there is an urgent need for redox modulators that can prevent these disorders. Recent Advances: Oxidative stress is associated with the generation of reactive oxygen species (ROS) and reactive nitrogen species, metal dyshomeostasis, and mitochondrial dysfunction. Here, we discuss the role that oxidative stress and metal dyshomeostasis play in hearing loss, visual impairments, and neurodegeneration and discuss the benefits of a new class of multifunctional redox modulators (MFRMs) that suppress sensory and neural degeneration. MFRMs not only reduce free radicals but also independently bind transition metals associated with the generation of hydroxyl radicals. The MFRMs redistribute zinc from neurotoxic amyloid beta zinc (Aβ:Zn) complexes to the cytoplasm, facilitating the degradation of Aβ plaques by matrix metalloprotease-2 (MMP-2). Although MFRMs bind copper (Cu1+, Cu2+), iron (Fe2+, Fe3+), zinc (Zn2+), and manganese (Mn2+), they do not deplete free cytoplasmic Zn+2 and they protect mitochondria from Mn+2-induced dysfunction. Oral administration of MFRMs reduce ROS-induced cataracts, protect the retina from light-induced degeneration, reduce neurotoxic Aβ:Zn plaque formation, and protect auditory hair cells from noise-induced hearing loss. Critical Issues: Regulation of redox balance is essential for clinical efficacy in maintaining sensory functions. Future Directions: Future use of these MFRMs requires additional pharmacokinetic, pharmacodynamics, and toxicological data to bring them into widespread clinical use. Additional animal studies are also needed to determine whether MFRMs can prevent neurodegeneration, dementia, and other forms of vision and hearing loss.
Collapse
Affiliation(s)
- Peter F. Kador
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Richard Salvi
- Center for Hearing and Deafness, University at Buffalo, Buffalo, New York, USA
| |
Collapse
|
40
|
Lem DW, Davey PG, Gierhart DL, Rosen RB. A Systematic Review of Carotenoids in the Management of Age-Related Macular Degeneration. Antioxidants (Basel) 2021; 10:1255. [PMID: 34439503 PMCID: PMC8389280 DOI: 10.3390/antiox10081255] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 07/30/2021] [Accepted: 08/02/2021] [Indexed: 01/14/2023] Open
Abstract
Age-related macular degeneration (AMD) remains a leading cause of modifiable vision loss in older adults. Chronic oxidative injury and compromised antioxidant defenses represent essential drivers in the development of retinal neurodegeneration. Overwhelming free radical species formation results in mitochondrial dysfunction, as well as cellular and metabolic imbalance, which becomes exacerbated with increasing age. Thus, the depletion of systemic antioxidant capacity further proliferates oxidative stress in AMD-affected eyes, resulting in loss of photoreceptors, neuroinflammation, and ultimately atrophy within the retinal tissue. The aim of this systematic review is to examine the neuroprotective potential of the xanthophyll carotenoids lutein, zeaxanthin, and meso-zeaxanthin on retinal neurodegeneration for the purpose of adjunctive nutraceutical strategy in the management of AMD. A comprehensive literature review was performed to retrieve 55 eligible publications, using four database searches from PubMed, Embase, Cochrane Library, and the Web of Science. Epidemiology studies indicated an enhanced risk reduction against late AMD with greater dietary consumption of carotenoids, meanwhile greater concentrations in macular pigment demonstrated significant improvements in visual function among AMD patients. Collectively, evidence strongly suggests that carotenoid vitamin therapies offer remarkable synergic protection in the neurosensory retina, with the potential to serve as adjunctive nutraceutical therapy in the management of established AMD, albeit these benefits may vary among different stages of disease.
Collapse
Affiliation(s)
- Drake W. Lem
- College of Optometry, Western University of Health Sciences, Pomona, CA 91766, USA;
| | | | | | - Richard B. Rosen
- Department of Ophthalmology, New York Eye and Ear Infirmary of Mount Sinai, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA;
| |
Collapse
|
41
|
Jiang K, Nellissery J, Swaroop A. Determination of Mitochondrial Respiration and Glycolysis in Ex Vivo Retinal Tissue Samples. J Vis Exp 2021. [PMID: 34424254 PMCID: PMC11375468 DOI: 10.3791/62914] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022] Open
Abstract
Mitochondrial respiration is a critical energy-generating pathway in all cells, especially retinal photoreceptors that possess a highly active metabolism. In addition, photoreceptors also exhibit high aerobic glycolysis like cancer cells. Precise measurements of these metabolic activities can provide valuable insights into cellular homeostasis under physiological conditions and in disease states. High throughput microplate-based assays have been developed to measure mitochondrial respiration and various metabolic activities in live cells. However, a vast majority of these are developed for cultured cells and have not been optimized for intact tissue samples and for application ex vivo. Described here is a detailed step-by-step protocol, using microplate-based fluorescence technology, to directly measure oxygen consumption rate (OCR) as an indicator of mitochondrial respiration, as well as extracellular acidification rate (ECAR) as an indicator of glycolysis, in intact ex vivo retinal tissue. This method has been used to successfully assess metabolic activities in adult mouse retina and demonstrate its application in investigating cellular mechanisms of aging and disease.
Collapse
Affiliation(s)
- Ke Jiang
- Neurobiology, Neurodegeneration & Repair Laboratory, National Eye Institute, National Institutes of Health;
| | - Jacob Nellissery
- Neurobiology, Neurodegeneration & Repair Laboratory, National Eye Institute, National Institutes of Health
| | - Anand Swaroop
- Neurobiology, Neurodegeneration & Repair Laboratory, National Eye Institute, National Institutes of Health;
| |
Collapse
|
42
|
Guerra MH, Yumnamcha T, Singh LP, Ibrahim AS. Relative Contribution of Different Mitochondrial Oxidative Phosphorylation Components to the Retinal Pigment Epithelium Barrier Function: Implications for RPE-Related Retinal Diseases. Int J Mol Sci 2021; 22:8130. [PMID: 34360894 PMCID: PMC8348500 DOI: 10.3390/ijms22158130] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Revised: 07/19/2021] [Accepted: 07/23/2021] [Indexed: 12/19/2022] Open
Abstract
Disruption of retinal pigment epithelial (RPE) barrier integrity is involved in the pathology of several blinding retinal diseases including age-related macular degeneration (AMD) and diabetic retinopathy (DR), but the underlying causes and pathophysiology are not completely well-defined. Mitochondria dysfunction has often been considered as a potential candidate implicated in such a process. In this study, we aimed to dissect the role of different mitochondrial components; specifically, those of oxidative phosphorylation (OxPhos), in maintaining the barrier functionality of RPE. Electric cell-substrate impedance sensing (ECIS) technology was used to collect multi-frequency electrical impedance data to assess in real-time the barrier formation of the RPE cells. For this purpose, the human retinal pigment epithelial cell line-ARPE-19-was used and treated with varying concentrations of specific mitochondrial inhibitors that target different steps in OxPhos: Rotenone for complex I (the largest protein complex in the electron transport chain (ETC)); oligomycin for ATP synthase; and carbonyl cyanide-p-trifluoromethoxyphenyl hydrazone (FCCP) for uncoupling ATP synthesis from the accompanying ETC. Furthermore, data were modeled using the ECIS-Zθ software to investigate in depth the effects of these inhibitors on three separate barrier parameters: cell-cell interactions (Rb), cell-matrix interactions (α), and the cell membrane capacitance (Cm). The viability of ARPE-19 cells was determined by lactate dehydrogenase (LDH) Cytotoxicity Assay. The ECIS program's modeling demonstrated that FCCP and thus OxPhos uncoupling disrupt the barrier function in the ARPE-19 cells across all three components of the total resistance (Rb, α, and Cm) in a dose-dependent manner. On the other hand, oligomycin and thus ATP synthase inhibition mostly affects the ARPE-19 cells' attachment to their substrate evident by a significant decrease in α resistance in a dose-dependent manner, both at the end and throughout the duration of the experiment. On the contrary, rotenone and complex I inhibition mostly affect the ARPE-19 paracellular resistance Rb in a dose-dependent manner compared to basolateral resistance α or Cm. Our results clearly demonstrate differential roles for different mitochondrial components in maintaining RPE cell functionality in which uncoupling of OxPhos is a major contributing factor to the disruption barrier function. Such differences can be used in investigating gene expression as well as for screening of selective agents that improve the OxPhos coupling efficiency to be used in the therapeutic approach for treating RPE-related retinal diseases.
Collapse
Affiliation(s)
- Michael H. Guerra
- Department of Ophthalmology, Visual and Anatomical Sciences, School of Medicine, Wayne State University, 540 East Canfield, Detroit, MI 48201, USA; (M.H.G.); (T.Y.); (L.P.S.)
| | - Thangal Yumnamcha
- Department of Ophthalmology, Visual and Anatomical Sciences, School of Medicine, Wayne State University, 540 East Canfield, Detroit, MI 48201, USA; (M.H.G.); (T.Y.); (L.P.S.)
| | - Lalit P. Singh
- Department of Ophthalmology, Visual and Anatomical Sciences, School of Medicine, Wayne State University, 540 East Canfield, Detroit, MI 48201, USA; (M.H.G.); (T.Y.); (L.P.S.)
| | - Ahmed S. Ibrahim
- Department of Ophthalmology, Visual and Anatomical Sciences, School of Medicine, Wayne State University, 540 East Canfield, Detroit, MI 48201, USA; (M.H.G.); (T.Y.); (L.P.S.)
- Department of Pharmacology, School of Medicine, Wayne State University, 540 East Canfield, Detroit, MI 48201, USA
- Department of Biochemistry, Faculty of Pharmacy, Mansoura University, Mansoura 35516, Egypt
| |
Collapse
|
43
|
All-Trans Retinoic Acid Increases DRP1 Levels and Promotes Mitochondrial Fission. Cells 2021; 10:cells10051202. [PMID: 34068960 PMCID: PMC8156392 DOI: 10.3390/cells10051202] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Revised: 05/07/2021] [Accepted: 05/10/2021] [Indexed: 02/07/2023] Open
Abstract
In the heart, mitochondrial homeostasis is critical for sustaining normal function and optimal responses to metabolic and environmental stressors. Mitochondrial fusion and fission are thought to be necessary for maintaining a robust population of mitochondria, and disruptions in mitochondrial fission and/or fusion can lead to cellular dysfunction. The dynamin-related protein (DRP1) is an important mediator of mitochondrial fission. In this study, we investigated the direct effects of the micronutrient retinoid all-trans retinoic acid (ATRA) on the mitochondrial structure in vivo and in vitro using Western blot, confocal, and transmission electron microscopy, as well as mitochondrial network quantification using stochastic modeling. Our results showed that ATRA increases DRP1 protein levels, increases the localization of DRP1 to mitochondria in isolated mitochondrial preparations. Our results also suggested that ATRA remodels the mitochondrial ultrastructure where the mitochondrial area and perimeter were decreased and the circularity was increased. Microscopically, mitochondrial network remodeling is driven by an increased rate of fission over fusion events in ATRA, as suggested by our numerical modeling. In conclusion, ATRA results in a pharmacologically mediated increase in the DRP1 protein. It also results in the modulation of cardiac mitochondria by promoting fission events, altering the mitochondrial network, and modifying the ultrastructure of mitochondria in the heart.
Collapse
|
44
|
Kim JY, Park S, Park HJ, Kim SH, Lew H, Kim GJ. PEDF-Mediated Mitophagy Triggers the Visual Cycle by Enhancing Mitochondrial Functions in a H 2O 2-Injured Rat Model. Cells 2021; 10:cells10051117. [PMID: 34066394 PMCID: PMC8148157 DOI: 10.3390/cells10051117] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 05/03/2021] [Accepted: 05/04/2021] [Indexed: 12/24/2022] Open
Abstract
Retinal degenerative diseases result from oxidative stress and mitochondrial dysfunction, leading to the loss of visual acuity. Damaged retinal pigment epithelial (RPE) and photoreceptor cells undergo mitophagy. Pigment epithelium-derived factor (PEDF) protects from oxidative stress in RPE and improves mitochondrial functions. Overexpression of PEDF in placenta-derived mesenchymal stem cells (PD-MSCs; PD-MSCsPEDF) provides therapeutic effects in retinal degenerative diseases. Here, we investigated whether PD-MSCsPEDF restored the visual cycle through a mitophagic mechanism in RPE cells in hydrogen peroxide (H2O2)-injured rat retinas. Compared with naïve PD-MSCs, PD-MSCsPEDF augmented mitochondrial biogenesis and translation markers as well as mitochondrial respiratory states. In the H2O2-injured rat model, intravitreal administration of PD-MSCsPEDF restored total retinal layer thickness compared to that of naïve PD-MSCs. In particular, PTEN-induced kinase 1 (PINK1), which is the major mitophagy marker, exhibited increased expression in retinal layers and RPE cells after PD-MSCPEDF transplantation. Similarly, expression of the visual cycle enzyme retinol dehydrogenase 11 (RDH11) showed the same patterns as PINK1 levels, resulting in improved visual activity. Taken together, these findings suggest that PD-MSCsPEDF facilitate mitophagy and restore the loss of visual cycles in H2O2-injured rat retinas and RPE cells. These data indicate a new strategy for next-generation MSC-based treatment of retinal degenerative diseases.
Collapse
Affiliation(s)
- Jae Yeon Kim
- Department of Biomedical Science, CHA University, Seongnam 13488, Korea; (J.Y.K.); (S.P.); (H.J.P.); (S.H.K.)
- Research Institute of Placental Science, CHA University, Seongnam 13488, Korea
| | - Sohae Park
- Department of Biomedical Science, CHA University, Seongnam 13488, Korea; (J.Y.K.); (S.P.); (H.J.P.); (S.H.K.)
| | - Hee Jung Park
- Department of Biomedical Science, CHA University, Seongnam 13488, Korea; (J.Y.K.); (S.P.); (H.J.P.); (S.H.K.)
| | - Se Ho Kim
- Department of Biomedical Science, CHA University, Seongnam 13488, Korea; (J.Y.K.); (S.P.); (H.J.P.); (S.H.K.)
| | - Helen Lew
- CHA Bundang Medical Center, Department of Ophthalmology, CHA University, Seongnam 13496, Korea;
| | - Gi Jin Kim
- Department of Biomedical Science, CHA University, Seongnam 13488, Korea; (J.Y.K.); (S.P.); (H.J.P.); (S.H.K.)
- Research Institute of Placental Science, CHA University, Seongnam 13488, Korea
- Correspondence: ; Tel.: +82-31-881-7145
| |
Collapse
|
45
|
Beeson C, Peterson YK, Perron N, Bandyopadhyay M, Nasarre C, Beeson G, Comer RF, Lindsey CC, Schnellmann RG, Rohrer B. Newly Identified Chemicals Preserve Mitochondrial Capacity and Decelerate Loss of Photoreceptor Cells in Murine Retinal Degeneration Models. J Ocul Pharmacol Ther 2021; 37:367-378. [PMID: 33945330 DOI: 10.1089/jop.2020.0140] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
Purpose: Metabolic stress and associated mitochondrial dysfunction are implicated in retinal degeneration irrespective of the underlying cause. We identified seven unique chemicals from a Chembridge DiverSET screen and tested their protection against photoreceptor cell death in cell- and animal-based approaches. Methods: Calcium overload (A23187) was triggered in 661W murine photoreceptor-derived cells, and changes in redox potential and real-time changes in cellular metabolism were assessed using the MTT and Seahorse Biosciences XF assay, respectively. Cheminformatics to compare structures, and biodistribution in the living pig eye aided in selection of the lead compound. In-situ, retinal organ cultures of rd1 mouse and S334ter-line-3 rat were tested, in-vivo the light-induced retinal degeneration in albino Balb/c mice was used, assessing photoreceptor cell numbers histologically. Results: Of the seven chemicals, six were protective against A23187- and IBMX-induced loss of mitochondrial capacity, as measured by viability and respirometry in 661W cells. Cheminformatic analyses identified a unique pharmacophore with 6 physico-chemical features based on two compounds (CB11 and CB12). The protective efficacy of CB11 was further shown by reducing photoreceptor cell loss in retinal explants from two retinitis pigmentosa rodent models. Using eye drops, CB11 targeting to the pig retina was confirmed. The same eye drops decreased photoreceptor cell loss in light-stressed Balb/c mice. Conclusions: New chemicals were identified that protect from mitochondrial damage and lead to improved mitochondrial function. Using ex-vivo and in-vivo models, CB11 decreased the loss of photoreceptor cells in murine models of retinal degeneration and may be effective as treatment for different retinal dystrophies.
Collapse
Affiliation(s)
- Craig Beeson
- Department of Drug Discovery and Biomedical Sciences, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Yuri K Peterson
- Department of Drug Discovery and Biomedical Sciences, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Nathan Perron
- Department of Ophthalmology, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Mausumi Bandyopadhyay
- Department of Ophthalmology, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Cecile Nasarre
- Department of Ophthalmology, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Gyda Beeson
- Department of Drug Discovery and Biomedical Sciences, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Richard F Comer
- Department of Drug Discovery and Biomedical Sciences, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Christopher C Lindsey
- Department of Drug Discovery and Biomedical Sciences, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Rick G Schnellmann
- Department of Pharmacology and Toxicology, University of Arizona, Tucson, Arizona, USA.,Research Service, Southern Arizona VA Healthcare System, Tucson, Arizona, USA
| | - Bärbel Rohrer
- Department of Ophthalmology, Medical University of South Carolina, Charleston, South Carolina, USA.,Research Service, Ralph H Johnson VA Medical Center, Charleston, South Carolina, USA
| |
Collapse
|
46
|
Toma C, De Cillà S, Palumbo A, Garhwal DP, Grossini E. Oxidative and Nitrosative Stress in Age-Related Macular Degeneration: A Review of Their Role in Different Stages of Disease. Antioxidants (Basel) 2021; 10:antiox10050653. [PMID: 33922463 PMCID: PMC8145578 DOI: 10.3390/antiox10050653] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Revised: 04/20/2021] [Accepted: 04/21/2021] [Indexed: 12/14/2022] Open
Abstract
Although the exact pathogenetic mechanisms leading to age-related macular degeneration (AMD) have not been clearly identified, oxidative damage in the retina and choroid due to an imbalance between local oxidants/anti-oxidant systems leading to chronic inflammation could represent the trigger event. Different in vitro and in vivo models have demonstrated the involvement of reactive oxygen species generated in a highly oxidative environment in the development of drusen and retinal pigment epithelium (RPE) changes in the initial pathologic processes of AMD; moreover, recent evidence has highlighted the possible association of oxidative stress and neovascular AMD. Nitric oxide (NO), which is known to play a key role in retinal physiological processes and in the regulation of choroidal blood flow, under pathologic conditions could lead to RPE/photoreceptor degeneration due to the generation of peroxynitrite, a potentially cytotoxic tyrosine-nitrating molecule. Furthermore, the altered expression of the different isoforms of NO synthases could be involved in choroidal microvascular changes leading to neovascularization. The purpose of this review was to investigate the different pathways activated by oxidative/nitrosative stress in the pathogenesis of AMD, focusing on the mechanisms leading to neovascularization and on the possible protective role of anti-vascular endothelial growth factor agents in this context.
Collapse
Affiliation(s)
- Caterina Toma
- Eye Clinic, University Hospital Maggiore Della Carità, 28100 Novara, Italy; (C.T.); (S.D.C.); (A.P.)
| | - Stefano De Cillà
- Eye Clinic, University Hospital Maggiore Della Carità, 28100 Novara, Italy; (C.T.); (S.D.C.); (A.P.)
- Department of Health Sciences, University East Piedmont “A. Avogadro”, 28100 Novara, Italy
| | - Aurelio Palumbo
- Eye Clinic, University Hospital Maggiore Della Carità, 28100 Novara, Italy; (C.T.); (S.D.C.); (A.P.)
| | - Divya Praveen Garhwal
- Laboratory of Physiology and Experimental Surgery, Department of Translational Medicine, University East Piedmont “A. Avogadro”, 28100 Novara, Italy;
| | - Elena Grossini
- Laboratory of Physiology and Experimental Surgery, Department of Translational Medicine, University East Piedmont “A. Avogadro”, 28100 Novara, Italy;
- Correspondence: ; Tel.:+39-0321-660526
| |
Collapse
|
47
|
Zhou B, Fang L, Dong Y, Yang J, Chen X, Zhang N, Zhu Y, Huang T. Mitochondrial quality control protects photoreceptors against oxidative stress in the H 2O 2-induced models of retinal degeneration diseases. Cell Death Dis 2021; 12:413. [PMID: 33879768 PMCID: PMC8058096 DOI: 10.1038/s41419-021-03660-5] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Revised: 03/22/2021] [Accepted: 03/22/2021] [Indexed: 12/26/2022]
Abstract
Retinal degeneration diseases (RDDs) are common and devastating eye diseases characterized by the degeneration of photoreceptors, which are highly associated with oxidative stress. Previous studies reported that mitochondrial dysfunction is associated with various neurodegenerative diseases. However, the role of mitochondrial proteostasis mainly regulated by mitophagy and mitochondrial unfolded protein response (mtUPR) in RDDs is unclear. We hypothesized that the mitochondrial proteostasis is neuroprotective against oxidative injury in RDDs. In this study, the data from our hydrogen peroxide (H2O2)-treated mouse retinal cone cell line (661w) model of RDDs showed that nicotinamide riboside (NR)-activated mitophagy increased the expression of LC3B II and PINK1, and promoted the co-localization of LC3 and mitochondria, as well as PINK1 and Parkin in the H2O2-treated 661w cells. However, the NR-induced mitophagy was remarkably reversed by chloroquine (CQ) and cyclosporine A (CsA), mitophagic inhibitors. In addition, doxycycline (DOX), an inducer of mtUPR, up-regulated the expression of HSP60 and CHOP, the key proteins of mtUPR. Activation of both mitophagy and mtUPR increased the cell viability and reduced the level of apoptosis and oxidative damage in the H2O2-treated 661w cells. Furthermore, both mitophagy and mtUPR played a protective effect on mitochondria by increasing mitochondrial membrane potential and maintaining mitochondrial mass. By contrast, the inhibition of mitophagy by CQ or CsA reversed the beneficial effect of mitophagy in the H2O2-treated 661w cells. Together, our study suggests that the mitophagy and mtUPR pathways may serve as new therapeutic targets to delay the progression of RDDs through enhancing mitochondrial proteostasis.
Collapse
Affiliation(s)
- Biting Zhou
- Department of Ophthalmology, The First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian, China
| | - Lijun Fang
- Department of Ophthalmology, Fujian Medical University Union Hospital, Fuzhou, Fujian, China
| | - Yanli Dong
- Qiqihaer Food and Drug Control Center, Qiqihaer, Heilongjiang, China
| | - Juhua Yang
- Department of Bioengineering and Biopharmaceutics, School of Pharmacy, Fujian Medical University, Fuzhou, Fujian, China
| | - Xiaole Chen
- Department of Bioengineering and Biopharmaceutics, School of Pharmacy, Fujian Medical University, Fuzhou, Fujian, China
| | - Nanwen Zhang
- Department of Pharmacology, School of Pharmacy, Fujian Medical University, Fuzhou, Fujian, China
| | - Yihua Zhu
- Department of Ophthalmology, The First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian, China.
| | - Tianwen Huang
- Department of Neurology, Fujian Medical University Union Hospital, Fuzhou, Fujian, China.
- Fujian Key Laboratory of Vascular Aging (Fujian Medical University), Fuzhou, Fujian, China.
| |
Collapse
|
48
|
Castelli V, Paladini A, d'Angelo M, Allegretti M, Mantelli F, Brandolini L, Cocchiaro P, Cimini A, Varrassi G. Taurine and oxidative stress in retinal health and disease. CNS Neurosci Ther 2021; 27:403-412. [PMID: 33621439 PMCID: PMC7941169 DOI: 10.1111/cns.13610] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Revised: 12/19/2020] [Accepted: 12/21/2020] [Indexed: 12/16/2022] Open
Abstract
Retinal disorders are leading causes of blindness and are due to an imbalance between reactive oxygen species and antioxidant scavenger (in favor of pro‐oxidant species) or a disruption of redox signaling and control. Indeed, it is well known that oxidative stress is one of the leading causes of retinal degenerative diseases. Different approaches using nutraceuticals resulted in protective effects in these disorders. This review will discuss the impact of oxidative stress in retinal neurodegenerative diseases and the potential strategies for avoiding or counteracting oxidative damage in retinal tissues, with a specific focus on taurine. Increasing data indicate that taurine may be effective in slowing down the progression of degenerative retinal diseases, thus suggesting that taurine can be a promising candidate for the prevention or as adjuvant treatment of these diseases. The mechanism by which taurine supplementation acts is mainly related to the reduction of oxidative stress. In particular, it has been demonstrated to improve retinal reduced glutathione, malondialdehyde, superoxide dismutase, and catalase activities. Antiapoptotic effects are also involved; however, the protective mechanisms exerted by taurine against retinal damage remain to be further investigated.
Collapse
Affiliation(s)
- Vanessa Castelli
- Department of Life, Health and Environmental Sciences, University of L'Aquila, L'Aquila, Italy
| | - Antonella Paladini
- Department of Life, Health and Environmental Sciences, University of L'Aquila, L'Aquila, Italy
| | - Michele d'Angelo
- Department of Life, Health and Environmental Sciences, University of L'Aquila, L'Aquila, Italy
| | | | | | | | | | - Annamaria Cimini
- Department of Life, Health and Environmental Sciences, University of L'Aquila, L'Aquila, Italy.,Sbarro Institute for Cancer Research and Molecular Medicine and Center for Biotechnology, Temple University, Philadelphia, PA, USA
| | | |
Collapse
|
49
|
George AK, Homme RP, Stanisic D, Tyagi SC, Singh M. Protecting the aging eye with hydrogen sulfide. Can J Physiol Pharmacol 2021; 99:161-170. [PMID: 32721225 DOI: 10.1139/cjpp-2020-0216] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Research demonstrates that senescence is associated with tissue and organ dysfunction, and the eye is no exception. Sequelae arising from aging have been well defined as distinct clinical entities and vision impairment has significant psychosocial consequences. Retina and adjacent tissues like retinal pigmented epithelium and choroid are the key structures that are required for visual perception. Any structural and functional changes in retinal layers and blood retinal barrier could lead to age-related macular degeneration, diabetic retinopathy, and glaucoma. Further, there are significant oxygen gradients in the eye that can lead to excessive reactive oxygen species, resulting in endoplasmic reticulum and mitochondrial stress response. These radicals are source of functional and morphological impairment in retinal pigmented epithelium and retinal ganglion cells. Therefore, ocular diseases could be summarized as disturbance in the redox homeostasis. Hyperhomocysteinemia is a risk factor and causes vascular occlusive disease of the retina. Interestingly, hydrogen sulfide (H2S) has been proven to be an effective antioxidant agent, and it can help treat diseases by alleviating stress and inflammation. Concurrent glutamate excitotoxicity, endoplasmic reticulum stress, and microglia activation are also linked to stress; thus, H2S may offer additional interventional strategy. A refined understanding of the aging eye along with H2S biology and pharmacology may help guide newer therapies for the eye.
Collapse
Affiliation(s)
- Akash K George
- Eye and Vision Science Laboratory, Department of Physiology, University of Louisville School of Medicine, Louisville, KY, USA
| | - Rubens P Homme
- Eye and Vision Science Laboratory, Department of Physiology, University of Louisville School of Medicine, Louisville, KY, USA
| | - Dragana Stanisic
- Department of Dentistry, Faculty of Medical Sciences, University of Kragujevac, Kragujevac, Serbia
| | - Suresh C Tyagi
- Department of Physiology, University of Louisville School of Medicine, Louisville, KY, USA
| | - Mahavir Singh
- Eye and Vision Science Laboratory, Department of Physiology, University of Louisville School of Medicine, Louisville, KY, USA
| |
Collapse
|
50
|
Tozawa T, Nishimura A, Ueno T, Shikata A, Taura Y, Yoshida T, Nakagawa N, Wada T, Kosugi S, Uehara T, Takenouchi T, Kosaki K, Chiyonobu T. Complex hereditary spastic paraplegia associated with episodic visual loss caused by ACO2 variants. Hum Genome Var 2021; 8:4. [PMID: 33500398 PMCID: PMC7838304 DOI: 10.1038/s41439-021-00136-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Revised: 12/15/2020] [Accepted: 12/15/2020] [Indexed: 11/09/2022] Open
Abstract
Most patients with homozygous or compound heterozygous pathogenic ACO2 variants present with muscular hypotonia features, namely, infantile cerebellar-retinal degeneration. Recently, two studies reported rare familial cases of ACO2 variants presenting as complex hereditary spastic paraplegia (HSP) with broad clinical spectra. Here, we report the case of a 20-year-old Japanese woman with complex HSP caused by compound heterozygous ACO2 variants, revealing a new phenotype of episodic visual loss during febrile illness.
Collapse
Affiliation(s)
- Takenori Tozawa
- Department of Pediatrics, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan. .,Department of Pediatrics, Ayabe City Hospital, Ayabe, Japan.
| | - Akira Nishimura
- Department of Neonatology, Japanese Red Cross Society Kyoto Daiichi Hospital, Kyoto, Japan
| | - Tamaki Ueno
- Department of Pediatrics, Ayabe City Hospital, Ayabe, Japan.,Department of Pediatrics, Tokai Central Hospital, Kakamigahara, Japan
| | - Akane Shikata
- Kyoto Prefectural Maizuru Rehabilitation Center for Children, Maizuru, Japan
| | - Yoshihiro Taura
- Department of Pediatrics, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Takeshi Yoshida
- Department of Pediatrics, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Naoko Nakagawa
- Department of Medical Ethics/Medical Genetics, Kyoto University School of Public Health, Kyoto, Japan
| | - Takahito Wada
- Department of Medical Ethics/Medical Genetics, Kyoto University School of Public Health, Kyoto, Japan
| | - Shinji Kosugi
- Department of Medical Ethics/Medical Genetics, Kyoto University School of Public Health, Kyoto, Japan
| | - Tomoko Uehara
- Center for Medical Genetics, Keio University School of Medicine, Tokyo, Japan
| | - Toshiki Takenouchi
- Department of Pediatrics, Keio University School of Medicine, Tokyo, Japan
| | - Kenjiro Kosaki
- Center for Medical Genetics, Keio University School of Medicine, Tokyo, Japan
| | - Tomohiro Chiyonobu
- Department of Pediatrics, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| |
Collapse
|