1
|
Ben S, Zheng Q, Zhao Y, Xia J, Mu W, Yao M, Yan B, Jiang Q. Tear Fluid-Based Metabolomics Profiling in Chronic Dacryocystitis Patients. J Proteome Res 2025; 24:224-233. [PMID: 39670809 DOI: 10.1021/acs.jproteome.4c00592] [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] [Indexed: 12/14/2024]
Abstract
Chronic dacryocystitis (CD) can result in severe complications and vision impairment due to ongoing microbial infections and persistent tearing. Tear fluid, which contains essential components vital for maintaining ocular surface health, has been investigated for its potential in the noninvasive identification of ocular biomarkers through metabolomics analysis. In this study, we employed UHPLC-MS/MS to analyze the tear metabolome of CD patients. UHPLC-MS/MS analysis of tear samples from CD patients revealed significant metabolic alterations. Compared with the control group, 298 metabolites were elevated, while 142 were decreased. KEGG pathway analysis suggested that these changes primarily affected arginine and proline metabolism, biosynthesis of amino acids, and phenylalanine biosynthesis in CD. Notably, 3-dehydroquinic acid, anthranilic acid, citric acid, and l-isoleucine emerged as potential biomarker candidates of CD with high diagnostic accuracy (AUC = 0.94). These findings suggest that tear fluid metabolism, particularly amino acid biosynthesis, plays a significant role in the pathogenesis of CD. Uncovering these metabolic products and pathways provides valuable insights into the mechanisms underlying CD and paves the way for the development of diagnostic tools and targeted therapies.
Collapse
Affiliation(s)
- Shuai Ben
- Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China
- The Affiliated Eye Hospital, Nanjing Medical University, Nanjing 210029, China
| | - Qun Zheng
- The Affiliated Eye Hospital, Nanjing Medical University, Nanjing 210029, China
- Nanjing Aier Eye Hospital, Nanjing 210000, China
| | - Ya Zhao
- Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China
| | - Jiao Xia
- Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China
| | - Wan Mu
- Eye Institute and Department of Ophthalmology, Eye and ENT Hospital, State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai 200030, China
| | - Mudi Yao
- Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China
| | - Biao Yan
- Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China
| | - Qin Jiang
- The Affiliated Eye Hospital, Nanjing Medical University, Nanjing 210029, China
| |
Collapse
|
2
|
Amiri-Dashatan N, Etemadi SM, Besharati S, Farahani M, Moghaddam AK. Dysregulation of amino acids balance as potential serum-metabolite biomarkers for diagnosis and prognosis of diabetic retinopathy: a metabolomics study. J Diabetes Metab Disord 2024; 23:2031-2042. [PMID: 39610496 PMCID: PMC11599686 DOI: 10.1007/s40200-024-01462-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/26/2024] [Accepted: 06/23/2024] [Indexed: 11/30/2024]
Abstract
Objectives Diabetic retinopathy (DR), an earnest complication of diabetes, is one of the most common causes of blindness worldwide. This study aimed to investigate the altered metabolites in the serum of non-DR (NDR) and DR including non-proliferative diabetic retinopathy (NPDR), and proliferative diabetic retinopathy (PDR) subjects. Methods In this study, the 1HNMR platform was applied to reveal the discriminating serum metabolites in three diabetic groups based on the status of their complications: T2D or NDR (n = 15), NPDR, (n = 15), and PDR (n = 15) groups. Multivariate analyses include principal component analysis (PCA) and Partial Least Structures-Discriminant Analysis (PLS-DA) analysis that were performed using R software. The main metabolic pathways were also revealed by KEGG pathway enrichment analysis. Results The results revealed the significantly different metabolites include 10 metabolites of the NPDR versus PDR group, 24 metabolites of the PDR versus NDR group, and 25 metabolites of the NPDR versus NDR group. The results showed that the significantly altered metabolites in DR compared with NDR serum samples mainly belonged to amino acids. The most important pathways between NPDR/PDR, and NDR/DR groups include ascorbate and aldarate metabolism, galactose metabolism, glutathione metabolism, and tryptophan metabolism, respectively. In addition, some metabolites were detected for the first time. Conclusions We created a metabolomics profile for NDR, PDR and NPDR groups. The impairment in the ascorbate/aldarate, galactose, and especially amino acids metabolism was identified as metabolic dysregulation associated with DR, which may provide new insights into potential pathogenesis pathways for DR. Graphical Abstract
Collapse
Affiliation(s)
- Nasrin Amiri-Dashatan
- Zanjan Metabolic Diseases Research Center, Zanjan University of Medical Sciences, Zanjan, Iran
| | | | - Shahin Besharati
- Zanjan Metabolic Diseases Research Center, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Masoumeh Farahani
- Proteomics Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Arezoo Karimi Moghaddam
- Department of Ophthalmology, School of Medicine, Vali-E-Asr Hospital, Zanjan University of Medical sciences, Zanjan, Iran
| |
Collapse
|
3
|
Wang H, Tao Y, Han J, Shen J, Mu H, Wang Z, Wang J, Jin X, Zhang Q, Yang Y, Lin J, Sun M, Ma X, Ren L, LeBlanc AK, Xu J, Hua Y, Sun W. Disrupting YAP1-mediated glutamine metabolism induces synthetic lethality alongside ODC1 inhibition in osteosarcoma. Cell Oncol (Dordr) 2024; 47:1845-1861. [PMID: 39115605 DOI: 10.1007/s13402-024-00967-1] [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] [Accepted: 06/06/2024] [Indexed: 10/11/2024] Open
Abstract
PURPOSE Osteosarcoma, a highly malignant primary bone tumor primarily affecting adolescents, frequently develops resistance to initial chemotherapy, leading to metastasis and limited treatment options. Our study aims to uncover novel therapeutic targets for metastatic and recurrent osteosarcoma. METHODS In this study, we proved the potential of modulating the YAP1-regulated glutamine metabolic pathway to augment the response of OS to DFMO. We initially employed single-cell transcriptomic data to gauge the activation level of polyamine metabolism in MTAP-deleted OS patients. This was further substantiated by transcriptome sequencing data from recurrent and non-recurrent patient tissues, confirming the activation of polyamine metabolism in progressive OS. Through high-throughput drug screening, we pinpointed CIL56, a YAP1 inhibitor, as a promising candidate for a combined therapeutic strategy with DFMO. In vivo, we utilized PDX and CDX models to validate the therapeutic efficacy of this drug combination. In vitro, we conducted western blot analysis, qPCR analysis, immunofluorescence staining, and PuMA experiments to monitor alterations in molecular expression, distribution, and tumor metastasis capability. We employed CCK-8 and colony formation assays to assess the proliferative capacity of cells in the experimental group. We used flow cytometry and reactive oxygen probes to observe changes in ROS and glutamine metabolism within the cells. Finally, we applied RNA-seq in tandem with metabolomics to identify metabolic alterations in OS cells treated with a DFMO and CIL56 combination. This enabled us to intervene and validate the role of the YAP1-mediated glutamine metabolic pathway in DFMO resistance. RESULTS Through single-cell RNA-seq data analysis, we pinpointed a subset of late-stage OS cells with significantly upregulated polyamine metabolism. This upregulation was further substantiated by transcriptomic profiling of recurrent and non-recurrent OS tissues. High-throughput drug screening revealed a promising combination strategy involving DFMO and CIL56. DFMO treatment curbs the phosphorylation of YAP1 protein in OS cells, promoting nuclear entry and initiating the YAP1-mediated glutamine metabolic pathway. This reduces intracellular ROS levels, countering DFMO's anticancer effect. The therapeutic efficacy of DFMO can be amplified both in vivo and in vitro by combining it with the YAP1 inhibitor CIL56 or the glutaminase inhibitor CB-839. This underscores the significant potential of targeting the YAP1-mediated glutamine metabolic pathway to enhance efficacy of DFMO. CONCLUSION Our findings elucidate YAP1-mediated glutamine metabolism as a crucial bypass mechanism against DFMO, following the inhibition of polyamine metabolism. Our study provides valuable insights into the potential role of DFMO in an "One-two Punch" therapy of metastatic and recurrent osteosarcoma.
Collapse
Affiliation(s)
- Hongsheng Wang
- Department of Orthopedics, Shanghai General Hospital, School of Medicine, Shanghai Jiao Tong University, 100 Haining Road, Shanghai, 200080, China
- Shanghai Bone Tumor Institution, Shanghai, China
| | - Yining Tao
- Department of Orthopedics, Shanghai General Hospital, School of Medicine, Shanghai Jiao Tong University, 100 Haining Road, Shanghai, 200080, China
- Shanghai Bone Tumor Institution, Shanghai, China
| | - Jing Han
- Department of Orthopedics, Shanghai General Hospital, School of Medicine, Shanghai Jiao Tong University, 100 Haining Road, Shanghai, 200080, China
- Shanghai Bone Tumor Institution, Shanghai, China
| | - Jiakang Shen
- Department of Orthopedics, Shanghai General Hospital, School of Medicine, Shanghai Jiao Tong University, 100 Haining Road, Shanghai, 200080, China
- Shanghai Bone Tumor Institution, Shanghai, China
| | - Haoran Mu
- Department of Orthopedics, Shanghai General Hospital, School of Medicine, Shanghai Jiao Tong University, 100 Haining Road, Shanghai, 200080, China
- Shanghai Bone Tumor Institution, Shanghai, China
| | - Zhuoying Wang
- Department of Orthopedics, Shanghai General Hospital, School of Medicine, Shanghai Jiao Tong University, 100 Haining Road, Shanghai, 200080, China
- Shanghai Bone Tumor Institution, Shanghai, China
| | - Jinzeng Wang
- National Research Center for Translational Medicine (Shanghai), State Key Laboratory of Medical Genomics, Shanghai Institute of Hematology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xinmeng Jin
- Department of Orthopedics, Shanghai General Hospital, School of Medicine, Shanghai Jiao Tong University, 100 Haining Road, Shanghai, 200080, China
- Shanghai Bone Tumor Institution, Shanghai, China
| | - Qi Zhang
- Department of Biochemistry and Molecular Cell Biology, Shanghai Key Laboratory for Tumor Microenvironment and Inflammation, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yuqin Yang
- Department of Laboratory Animal Center, Shanghai General Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Jun Lin
- Department of Pathology, Shanghai General Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Mengxiong Sun
- Department of Orthopedics, Shanghai General Hospital, School of Medicine, Shanghai Jiao Tong University, 100 Haining Road, Shanghai, 200080, China
- Shanghai Bone Tumor Institution, Shanghai, China
| | - Xiaojun Ma
- Department of Orthopedics, Shanghai General Hospital, School of Medicine, Shanghai Jiao Tong University, 100 Haining Road, Shanghai, 200080, China
- Shanghai Bone Tumor Institution, Shanghai, China
| | - Ling Ren
- Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Amy K LeBlanc
- Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Jing Xu
- Department of Orthopedics, Shanghai General Hospital, School of Medicine, Shanghai Jiao Tong University, 100 Haining Road, Shanghai, 200080, China.
- Shanghai Bone Tumor Institution, Shanghai, China.
| | - Yingqi Hua
- Department of Orthopedics, Shanghai General Hospital, School of Medicine, Shanghai Jiao Tong University, 100 Haining Road, Shanghai, 200080, China.
- Shanghai Bone Tumor Institution, Shanghai, China.
| | - Wei Sun
- Department of Orthopedics, Shanghai General Hospital, School of Medicine, Shanghai Jiao Tong University, 100 Haining Road, Shanghai, 200080, China.
- Shanghai Bone Tumor Institution, Shanghai, China.
| |
Collapse
|
4
|
Bouhlel J, Caffin F, Gros-Désormeaux F, Douki T, Benoist JF, Castelli FA, Chu-Van E, Piérard C, Junot C, Fenaille F. Metabolomics Analysis of Rabbit Plasma after Ocular Exposure to Vapors of Sulfur Mustard. Metabolites 2024; 14:349. [PMID: 39057672 PMCID: PMC11279318 DOI: 10.3390/metabo14070349] [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: 06/11/2024] [Revised: 06/19/2024] [Accepted: 06/20/2024] [Indexed: 07/28/2024] Open
Abstract
Sulfur mustard (SM) is a highly potent alkylating vesicant agent and remains a relevant threat to both civilians and military personnel. The eyes are the most sensitive organ after airborne SM exposure, causing ocular injuries with no antidote or specific therapeutics available. In order to identify relevant biomarkers and to obtain a deeper understanding of the underlying biochemical events, we performed an untargeted metabolomics analysis using liquid chromatography coupled to high-resolution mass spectrometry of plasma samples from New Zealand white rabbits ocularly exposed to vapors of SM. Metabolic profiles (332 unique metabolites) from SM-exposed (n = 16) and unexposed rabbits (n = 8) were compared at different time intervals from 1 to 28 days. The observed time-dependent changes in metabolic profiles highlighted the profound dysregulation of the sulfur amino acids, the phenylalanine, the tyrosine and tryptophan pathway, and the polyamine and purine biosynthesis, which could reflect antioxidant and anti-inflammatory activities. Taurine and 3,4-dihydroxy-phenylalanine (Dopa) seem to be specifically related to SM exposure and correspond well with the different phases of ocular damage, while the dysregulation of adenosine, polyamines, and acylcarnitines might be related to ocular neovascularization. Additionally, neither cysteine, N-acetylcysteine, or guanine SM adducts were detected in the plasma of exposed rabbits at any time point. Overall, our study provides an unprecedented view of the plasma metabolic changes post-SM ocular exposure, which may open up the development of potential new treatment strategies.
Collapse
Affiliation(s)
- Jihéne Bouhlel
- CEA, INRAE, Département Médicaments et Technologies pour la Santé (DMTS), SPI, MetaboHUB-IDF, Université Paris-Saclay, 91191 Gif-sur-Yvette, France; (J.B.); (J.-F.B.); (F.A.C.); (E.C.-V.); (C.J.)
| | - Fanny Caffin
- Institut de Recherche Biomédicale des Armées (IRBA), 91223 Brétigny-sur-Orge, France; (F.C.); (F.G.-D.)
| | - Fanny Gros-Désormeaux
- Institut de Recherche Biomédicale des Armées (IRBA), 91223 Brétigny-sur-Orge, France; (F.C.); (F.G.-D.)
| | - Thierry Douki
- CEA, CNRS, IRIG, SyMMES, Université Grenoble Alpes, 38000 Grenoble, France;
| | - Jean-François Benoist
- CEA, INRAE, Département Médicaments et Technologies pour la Santé (DMTS), SPI, MetaboHUB-IDF, Université Paris-Saclay, 91191 Gif-sur-Yvette, France; (J.B.); (J.-F.B.); (F.A.C.); (E.C.-V.); (C.J.)
- Biochemistry Laboratory, APHP, Hôpital Universitaire Necker Enfants Malades, 75015 Paris, France
| | - Florence A. Castelli
- CEA, INRAE, Département Médicaments et Technologies pour la Santé (DMTS), SPI, MetaboHUB-IDF, Université Paris-Saclay, 91191 Gif-sur-Yvette, France; (J.B.); (J.-F.B.); (F.A.C.); (E.C.-V.); (C.J.)
| | - Emeline Chu-Van
- CEA, INRAE, Département Médicaments et Technologies pour la Santé (DMTS), SPI, MetaboHUB-IDF, Université Paris-Saclay, 91191 Gif-sur-Yvette, France; (J.B.); (J.-F.B.); (F.A.C.); (E.C.-V.); (C.J.)
| | - Christophe Piérard
- Institut de Recherche Biomédicale des Armées (IRBA), 91223 Brétigny-sur-Orge, France; (F.C.); (F.G.-D.)
| | - Christophe Junot
- CEA, INRAE, Département Médicaments et Technologies pour la Santé (DMTS), SPI, MetaboHUB-IDF, Université Paris-Saclay, 91191 Gif-sur-Yvette, France; (J.B.); (J.-F.B.); (F.A.C.); (E.C.-V.); (C.J.)
| | - François Fenaille
- CEA, INRAE, Département Médicaments et Technologies pour la Santé (DMTS), SPI, MetaboHUB-IDF, Université Paris-Saclay, 91191 Gif-sur-Yvette, France; (J.B.); (J.-F.B.); (F.A.C.); (E.C.-V.); (C.J.)
| |
Collapse
|
5
|
Fucito M, Spedicato M, Felletti S, Yu AC, Busin M, Pasti L, Franchina FA, Cavazzini A, De Luca C, Catani M. A Look into Ocular Diseases: The Pivotal Role of Omics Sciences in Ophthalmology Research. ACS MEASUREMENT SCIENCE AU 2024; 4:247-259. [PMID: 38910860 PMCID: PMC11191728 DOI: 10.1021/acsmeasuresciau.3c00067] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Revised: 02/06/2024] [Accepted: 02/07/2024] [Indexed: 06/25/2024]
Abstract
Precision medicine is a new medical approach which considers both population characteristics and individual variability to provide customized healthcare. The transition from traditional reactive medicine to personalized medicine is based on a biomarker-driven process and a deep knowledge of biological mechanisms according to which the development of diseases occurs. In this context, the advancements in high-throughput omics technologies represent a unique opportunity to discover novel biomarkers and to provide an unbiased picture of the biological system. One of the medical fields in which omics science has started to be recently applied is that of ophthalmology. Ocular diseases are very common, and some of them could be highly disabling, thus leading to vision loss and blindness. The pathogenic mechanism of most ocular diseases may be dependent on various genetic and environmental factors, whose effect has not been yet completely understood. In this context, large-scale omics approaches are fundamental to have a comprehensive evaluation of the whole system and represent an essential tool for the development of novel therapies. This Review summarizes the recent advancements in omics science applied to ophthalmology in the last ten years, in particular by focusing on proteomics, metabolomics and lipidomics applications from an analytical perspective. The role of high-efficiency separation techniques coupled to (high-resolution) mass spectrometry ((HR)MS) is also discussed, as well as the impact of sampling, sample preparation and data analysis as integrating parts of the analytical workflow.
Collapse
Affiliation(s)
- Maurine Fucito
- Department
of Chemical, Pharmaceutical and Agricultural Sciences, University of Ferrara, via L. Borsari 46, 44121 Ferrara, Italy
| | - Matteo Spedicato
- Department
of Chemical, Pharmaceutical and Agricultural Sciences, University of Ferrara, via L. Borsari 46, 44121 Ferrara, Italy
| | - Simona Felletti
- Department
of Environmental and Prevention Sciences, University of Ferrara, via L. Borsari 46, Ferrara 44121, Italy
| | - Angeli Christy Yu
- Department
of Translational Medicine and for Romagna, University of Ferrara, via Aldo Moro 8, 44124 Ferrara, Italy
| | - Massimo Busin
- Department
of Translational Medicine and for Romagna, University of Ferrara, via Aldo Moro 8, 44124 Ferrara, Italy
| | - Luisa Pasti
- Department
of Environmental and Prevention Sciences, University of Ferrara, via L. Borsari 46, Ferrara 44121, Italy
| | - Flavio A. Franchina
- Department
of Chemical, Pharmaceutical and Agricultural Sciences, University of Ferrara, via L. Borsari 46, 44121 Ferrara, Italy
| | - Alberto Cavazzini
- Department
of Chemical, Pharmaceutical and Agricultural Sciences, University of Ferrara, via L. Borsari 46, 44121 Ferrara, Italy
- Council
for Agricultural Research and Economics, via della Navicella 2/4, Rome 00184, Italy
| | - Chiara De Luca
- Department
of Chemical, Pharmaceutical and Agricultural Sciences, University of Ferrara, via L. Borsari 46, 44121 Ferrara, Italy
| | - Martina Catani
- Department
of Chemical, Pharmaceutical and Agricultural Sciences, University of Ferrara, via L. Borsari 46, 44121 Ferrara, Italy
| |
Collapse
|
6
|
Zhang L, Buonfiglio F, Fieß A, Pfeiffer N, Gericke A. Retinopathy of Prematurity-Targeting Hypoxic and Redox Signaling Pathways. Antioxidants (Basel) 2024; 13:148. [PMID: 38397746 PMCID: PMC10885953 DOI: 10.3390/antiox13020148] [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: 12/30/2023] [Revised: 01/19/2024] [Accepted: 01/21/2024] [Indexed: 02/25/2024] Open
Abstract
Retinopathy of prematurity (ROP) is a proliferative vascular ailment affecting the retina. It is the main risk factor for visual impairment and blindness in infants and young children worldwide. If left undiagnosed and untreated, it can progress to retinal detachment and severe visual impairment. Geographical variations in ROP epidemiology have emerged over recent decades, attributable to differing levels of care provided to preterm infants across countries and regions. Our understanding of the causes of ROP, screening, diagnosis, treatment, and associated risk factors continues to advance. This review article aims to present the pathophysiological mechanisms of ROP, including its treatment. Specifically, it delves into the latest cutting-edge treatment approaches targeting hypoxia and redox signaling pathways for this condition.
Collapse
Affiliation(s)
| | | | | | | | - Adrian Gericke
- Department of Ophthalmology, University Medical Center, Johannes Gutenberg University Mainz, Langenbeckstrasse 1, 55131 Mainz, Germany; (L.Z.); (F.B.); (A.F.); (N.P.)
| |
Collapse
|
7
|
Zhang JY, Greenwald MJ, Rodriguez SH. Gut Microbiome and Retinopathy of Prematurity. THE AMERICAN JOURNAL OF PATHOLOGY 2023; 193:1683-1690. [PMID: 36780985 DOI: 10.1016/j.ajpath.2023.01.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Revised: 01/17/2023] [Accepted: 01/26/2023] [Indexed: 02/13/2023]
Abstract
Retinopathy of prematurity (ROP), a leading cause of childhood blindness worldwide, is strongly associated with gestational age and weight at birth. Yet, many extremely preterm infants never develop ROP or develop only mild ROP with spontaneous regression. In addition, a myriad of other factors play a role in the retinal pathology, one of which may include the early gut microbiome. The complications associated with early gestational age include dysbiosis of the dynamic neonatal gut microbiome, as evidenced by the development of often concomitant conditions, such as necrotizing enterocolitis. Given this, alongside growing evidence for a gut-retina axis, there is an increasing interest in how the early intestinal environment may play a role in the pathophysiology of ROP. Potential mechanisms include dysregulation of vascular endothelial growth factor and insulin-like growth factor 1. Furthermore, the gut microbiome may be impacted by other known risk factors for ROP, such as intermittent hypoxia and sepsis treated with antibiotics. This mini-review summarizes the literature supporting these proposed avenues, establishing a foundation to guide future studies.
Collapse
Affiliation(s)
- Jason Y Zhang
- Pritzker School of Medicine, University of Chicago, Chicago, Illinois; Department of Ophthalmology and Visual Science, University of Chicago, Chicago, Illinois
| | - Mark J Greenwald
- Department of Ophthalmology and Visual Science, University of Chicago, Chicago, Illinois
| | - Sarah H Rodriguez
- Department of Ophthalmology and Visual Science, University of Chicago, Chicago, Illinois.
| |
Collapse
|
8
|
Błaszkiewicz M, Walulik A, Florek K, Górecki I, Sławatyniec O, Gomułka K. Advances and Perspectives in Relation to the Molecular Basis of Diabetic Retinopathy-A Review. Biomedicines 2023; 11:2951. [PMID: 38001952 PMCID: PMC10669459 DOI: 10.3390/biomedicines11112951] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Revised: 10/26/2023] [Accepted: 10/27/2023] [Indexed: 11/26/2023] Open
Abstract
Diabetes mellitus (DM) is a growing problem nowadays, and diabetic retinopathy (DR) is its predominant complication. Currently, DR diagnosis primarily relies on fundoscopic examination; however, novel biomarkers may facilitate that process and make it widely available. In this current review, we delve into the intricate roles of various factors and mechanisms in DR development, progression, prediction, and their association with therapeutic approaches linked to the underlying pathogenic pathways. Specifically, we focus on advanced glycation end products, vascular endothelial growth factor (VEGF), asymmetric dimethylarginine, endothelin-1, and the epigenetic regulation mediated by microRNAs (miRNAs) in the context of DR.
Collapse
Affiliation(s)
- Michał Błaszkiewicz
- Student Scientific Group of Adult Allergology, Wroclaw Medical University, 50-369 Wroclaw, Poland
| | - Agata Walulik
- Student Scientific Group of Adult Allergology, Wroclaw Medical University, 50-369 Wroclaw, Poland
| | - Kamila Florek
- Student Scientific Group of Adult Allergology, Wroclaw Medical University, 50-369 Wroclaw, Poland
| | - Ignacy Górecki
- Student Scientific Group of Adult Allergology, Wroclaw Medical University, 50-369 Wroclaw, Poland
| | - Olga Sławatyniec
- Student Scientific Group of Adult Allergology, Wroclaw Medical University, 50-369 Wroclaw, Poland
| | - Krzysztof Gomułka
- Clinical Department of Internal Medicine, Pneumology and Allergology, Wroclaw Medical University, 50-369 Wroclaw, Poland
| |
Collapse
|
9
|
Yang W, Xia F, Mei F, Shi S, Robichaux WG, Lin W, Zhang W, Liu H, Cheng X. Upregulation of Epac1 Promotes Pericyte Loss by Inducing Mitochondrial Fission, Reactive Oxygen Species Production, and Apoptosis. Invest Ophthalmol Vis Sci 2023; 64:34. [PMID: 37651112 PMCID: PMC10476449 DOI: 10.1167/iovs.64.11.34] [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: 03/30/2023] [Accepted: 08/09/2023] [Indexed: 09/01/2023] Open
Abstract
Purpose The pathogenic mechanisms behind the development of ischemic retinopathy are complex and poorly understood. This study investigates the involvement of exchange protein directly activated by cAMP (Epac)1 signaling in pericyte injury during ischemic retinopathy, including diabetic retinopathy, a disease that threatens vision. Methods Mouse models of retinal ischemia-reperfusion injury and type 1 diabetes induced by streptozotocin were used to investigate the pathogenesis of these diseases. The roles of Epac1 signaling in the pathogenesis of ischemic retinopathy were determined by an Epac1 knockout mouse model. The cellular and molecular mechanisms of Epac1-mediated pericyte dysfunction in response to high glucose were investigated by specific modulation of Epac1 activity in primary human retinal pericytes using Epac1-specific RNA interference and a pharmacological inhibitor. Results Ischemic injury or diabetes-induced retinal capillary degeneration were associated with an increased expression of Epac1 in the mouse retinal vasculature, including both endothelial cells and pericytes. Genetic deletion of Epac1 protected ischemic injury-induced pericyte loss and capillary degeneration in the mouse retina. Furthermore, high glucose-induced Epac1 expression in retinal pericytes was accompanied by increased Drp1 phosphorylation, mitochondrial fission, reactive oxygen species production, and caspase 3 activation. Inhibition of Epac1 via RNA interference or pharmacological approaches blocked high glucose-mediated mitochondrial dysfunction and caspase 3 activation. Conclusions Our study reveals an important role of Epac1 signaling in mitochondrial dynamics, reactive oxygen species production, and apoptosis in retinal pericytes and identifies Epac1 as a therapeutic target for treating ischemic retinopathy.
Collapse
Affiliation(s)
- Wenli Yang
- Department of Integrative Biology and Pharmacology, University of Texas Health Science Center, Houston, Texas, United States
- Texas Therapeutics Institute, University of Texas Health Science Center, Houston, Texas, United States
| | - Fan Xia
- Department of Ophthalmology & Visual Sciences, University of Texas Medical Branch, Galveston, Texas, United States
| | - Fang Mei
- Department of Integrative Biology and Pharmacology, University of Texas Health Science Center, Houston, Texas, United States
- Texas Therapeutics Institute, University of Texas Health Science Center, Houston, Texas, United States
| | - Shuizhen Shi
- Department of Ophthalmology & Visual Sciences, University of Texas Medical Branch, Galveston, Texas, United States
| | - William G. Robichaux
- Department of Integrative Biology and Pharmacology, University of Texas Health Science Center, Houston, Texas, United States
- Texas Therapeutics Institute, University of Texas Health Science Center, Houston, Texas, United States
| | - Wei Lin
- Department of Integrative Biology and Pharmacology, University of Texas Health Science Center, Houston, Texas, United States
- Texas Therapeutics Institute, University of Texas Health Science Center, Houston, Texas, United States
| | - Wenbo Zhang
- Department of Ophthalmology & Visual Sciences, University of Texas Medical Branch, Galveston, Texas, United States
- Department of Neurobiology, University of Texas Medical Branch, Galveston, Texas, United States
| | - Hua Liu
- Department of Ophthalmology & Visual Sciences, University of Texas Medical Branch, Galveston, Texas, United States
| | - Xiaodong Cheng
- Department of Integrative Biology and Pharmacology, University of Texas Health Science Center, Houston, Texas, United States
- Texas Therapeutics Institute, University of Texas Health Science Center, Houston, Texas, United States
| |
Collapse
|
10
|
Chen C, Ding P, Yan W, Wang Z, Lan Y, Yan X, Li T, Han J. Pharmacological roles of lncRNAs in diabetic retinopathy with a focus on oxidative stress and inflammation. Biochem Pharmacol 2023; 214:115643. [PMID: 37315816 DOI: 10.1016/j.bcp.2023.115643] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Revised: 06/03/2023] [Accepted: 06/07/2023] [Indexed: 06/16/2023]
Abstract
Diabetic retinopathy (DR) is a complication caused by abnormal glucose metabolism, which affects the vision and quality of life of patients and severely impacts the society at large.DR has a complex pathogenic process. Evidence from multiple studies have shown that oxidative stress and inflammation play pivotal roles in DR.Additionally, with the rapid development of various genetic detection methods, the abnormal expression of long non-coding RNAs (lncRNAs) have been confirmed to promote the development of DR.Research has demonstrated the potential of lncRNAs as ideal biomarkers and theranostic targets in DR. In this narrative review, we will focus on the research results on mechanisms underlying DR, list lncRNAs confirmed to be closely related to these mechanisms, and discuss their potential clinical application value and limitations.
Collapse
Affiliation(s)
- Chengming Chen
- Department of Ophthalmology, Tangdu Hospital, The Air Force Military Medical University, Xi'an 710038, China; Department of Ophthalmology, The 900th Hospital of Joint Logistic Support Force, PLA (Clinical Medical College of Fujian Medical University, Dongfang Hospital Affiliated to Xiamen University), Fuzhou 350025, China
| | - Peng Ding
- Department of Thoracic Surgery, Tangdu Hospital, The Air Force Military Medical University, Xi'an 710038, China
| | - Weiming Yan
- Department of Ophthalmology, The 900th Hospital of Joint Logistic Support Force, PLA (Clinical Medical College of Fujian Medical University, Dongfang Hospital Affiliated to Xiamen University), Fuzhou 350025, China
| | - Zhaoyang Wang
- Department of Thoracic Surgery, Tangdu Hospital, The Air Force Military Medical University, Xi'an 710038, China
| | - Yanyan Lan
- College of Rehabilitation Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou 350122, China
| | - Xiaolong Yan
- Department of Thoracic Surgery, Tangdu Hospital, The Air Force Military Medical University, Xi'an 710038, China.
| | - Tian Li
- School of Basic Medicine, Fourth Military Medical University, Xi'an 710032, China.
| | - Jing Han
- Department of Ophthalmology, Tangdu Hospital, The Air Force Military Medical University, Xi'an 710038, China.
| |
Collapse
|
11
|
Fevereiro-Martins M, Marques-Neves C, Guimarães H, Bicho M. Retinopathy of prematurity: A review of pathophysiology and signaling pathways. Surv Ophthalmol 2023; 68:175-210. [PMID: 36427559 DOI: 10.1016/j.survophthal.2022.11.007] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Revised: 11/15/2022] [Accepted: 11/18/2022] [Indexed: 11/25/2022]
Abstract
Retinopathy of prematurity (ROP) is a vasoproliferative disorder of the retina and a leading cause of visual impairment and childhood blindness worldwide. The disease is characterized by an early stage of retinal microvascular degeneration, followed by neovascularization that can lead to subsequent retinal detachment and permanent visual loss. Several factors play a key role during the different pathological stages of the disease. Oxidative and nitrosative stress and inflammatory processes are important contributors to the early stage of ROP. Nitric oxide synthase and arginase play important roles in ischemia/reperfusion-induced neurovascular degeneration. Destructive neovascularization is driven by mediators of the hypoxia-inducible factor pathway, such as vascular endothelial growth factor and metabolic factors (succinate). The extracellular matrix is involved in hypoxia-induced retinal neovascularization. Vasorepulsive molecules (semaphorin 3A) intervene preventing the revascularization of the avascular zone. This review focuses on current concepts about signaling pathways and their mediators, involved in the pathogenesis of ROP, highlighting new potentially preventive and therapeutic modalities. A better understanding of the intricate molecular mechanisms underlying the pathogenesis of ROP should allow the development of more effective and targeted therapeutic agents to reduce aberrant vasoproliferation and facilitate physiological retinal vascular development.
Collapse
Affiliation(s)
- Mariza Fevereiro-Martins
- Laboratório de Genética and Grupo Ecogenética e Saúde Humana, Instituto de Saúde Ambiental, Faculdade de Medicina, Universidade de Lisboa, Portugal; Instituto de Investigação Científica Bento da Rocha Cabral, Lisboa, Portugal; Departamento de Oftalmologia, Hospital Cuf Descobertas, Lisboa, Portugal.
| | - Carlos Marques-Neves
- Centro de Estudos das Ci.¼ncias da Visão, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal; Grupo Ecogenética e Saúde Humana, Instituto de Saúde Ambiental, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal.
| | - Hercília Guimarães
- Departamento de Ginecologia-Obstetrícia e Pediatria, Faculdade de Medicina, Universidade do Porto, Porto, Portugal.
| | - Manuel Bicho
- Laboratório de Genética and Grupo Ecogenética e Saúde Humana, Instituto de Saúde Ambiental, Faculdade de Medicina, Universidade de Lisboa, Portugal; Instituto de Investigação Científica Bento da Rocha Cabral, Lisboa, Portugal.
| |
Collapse
|
12
|
New insight of metabolomics in ocular diseases in the context of 3P medicine. EPMA J 2023; 14:53-71. [PMID: 36866159 PMCID: PMC9971428 DOI: 10.1007/s13167-023-00313-9] [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: 11/01/2022] [Accepted: 01/09/2023] [Indexed: 02/19/2023]
Abstract
Metabolomics refers to the high-through untargeted or targeted screening of metabolites in biofluids, cells, and tissues. Metabolome reflects the functional states of cells and organs of an individual, influenced by genes, RNA, proteins, and environment. Metabolomic analyses help to understand the interaction between metabolism and phenotype and reveal biomarkers for diseases. Advanced ocular diseases can lead to vision loss and blindness, reducing patients' quality of life and aggravating socio-economic burden. Contextually, the transition from reactive medicine to the predictive, preventive, and personalized (PPPM / 3P) medicine is needed. Clinicians and researchers dedicate a lot of efforts to explore effective ways for disease prevention, biomarkers for disease prediction, and personalized treatments, by taking advantages of metabolomics. In this way, metabolomics has great clinical utility in the primary and secondary care. In this review, we summarized much progress achieved by applying metabolomics to ocular diseases and pointed out potential biomarkers and metabolic pathways involved to promote 3P medicine approach in healthcare.
Collapse
|
13
|
Wei Q, Deng Y, Yang Q, Zhan A, Wang L. The markers to delineate different phenotypes of macrophages related to metabolic disorders. Front Immunol 2023; 14:1084636. [PMID: 36814909 PMCID: PMC9940311 DOI: 10.3389/fimmu.2023.1084636] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Accepted: 01/17/2023] [Indexed: 02/05/2023] Open
Abstract
Macrophages have a wide variety of roles in physiological and pathological conditions, making them promising diagnostic and therapeutic targets in diseases, especially metabolic disorders, which have attracted considerable attention in recent years. Owing to their heterogeneity and polarization, the phenotypes and functions of macrophages related to metabolic disorders are diverse and complicated. In the past three decades, the rapid progress of macrophage research has benefited from the emergence of specific molecular markers to delineate different phenotypes of macrophages and elucidate their role in metabolic disorders. In this review, we analyze the functions and applications of commonly used and novel markers of macrophages related to metabolic disorders, facilitating the better use of these macrophage markers in metabolic disorder research.
Collapse
Affiliation(s)
- Quxing Wei
- Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine, Guangdong Pharmaceutical University, Guangzhou, China.,Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education of China, Guangzhou, China.,Guangdong Traditional Chinese Medicine Key Laboratory for Metabolic Diseases, Guangdong Pharmaceutical University, Guangzhou, China.,Institute of Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou, China
| | - Yanyue Deng
- Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine, Guangdong Pharmaceutical University, Guangzhou, China.,Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education of China, Guangzhou, China.,Guangdong Traditional Chinese Medicine Key Laboratory for Metabolic Diseases, Guangdong Pharmaceutical University, Guangzhou, China.,Institute of Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou, China
| | - Qianqian Yang
- Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine, Guangdong Pharmaceutical University, Guangzhou, China.,Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education of China, Guangzhou, China.,Guangdong Traditional Chinese Medicine Key Laboratory for Metabolic Diseases, Guangdong Pharmaceutical University, Guangzhou, China.,Institute of Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou, China
| | - Angyu Zhan
- Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine, Guangdong Pharmaceutical University, Guangzhou, China.,Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education of China, Guangzhou, China.,Guangdong Traditional Chinese Medicine Key Laboratory for Metabolic Diseases, Guangdong Pharmaceutical University, Guangzhou, China.,Institute of Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou, China
| | - Lexun Wang
- Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine, Guangdong Pharmaceutical University, Guangzhou, China.,Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education of China, Guangzhou, China.,Guangdong Traditional Chinese Medicine Key Laboratory for Metabolic Diseases, Guangdong Pharmaceutical University, Guangzhou, China.,Institute of Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou, China
| |
Collapse
|
14
|
Guo X, Xing Y, Jin W. Role of ADMA in the pathogenesis of microvascular complications in type 2 diabetes mellitus. Front Endocrinol (Lausanne) 2023; 14:1183586. [PMID: 37152974 PMCID: PMC10160678 DOI: 10.3389/fendo.2023.1183586] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Accepted: 04/11/2023] [Indexed: 05/09/2023] Open
Abstract
Diabetic microangiopathy is a typical and severe problem in diabetics, including diabetic retinopathy, diabetic nephropathy, diabetic neuropathy, and diabetic cardiomyopathy. Patients with type 2 diabetes and diabetic microvascular complications have significantly elevated levels of Asymmetric dimethylarginine (ADMA), which is an endogenous inhibitor of nitric oxide synthase (NOS). ADMA facilitates the occurrence and progression of microvascular complications in type 2 diabetes through its effects on endothelial cell function, oxidative stress damage, inflammation, and fibrosis. This paper reviews the association between ADMA and microvascular complications of diabetes and elucidates the underlying mechanisms by which ADMA contributes to these complications. It provides a new idea and method for the prevention and treatment of microvascular complications in type 2 diabetes.
Collapse
Affiliation(s)
| | | | - Wei Jin
- *Correspondence: Yiqiao Xing, ; Wei Jin,
| |
Collapse
|
15
|
Doman AJ, Tommasi S, Perkins MV, McKinnon RA, Mangoni AA, Nair PC. Chemical similarities and differences among inhibitors of nitric oxide synthase, arginase and dimethylarginine dimethylaminohydrolase-1: implications for the design of novel enzyme inhibitors modulating the nitric oxide pathway. Bioorg Med Chem 2022; 72:116970. [DOI: 10.1016/j.bmc.2022.116970] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Revised: 08/01/2022] [Accepted: 08/18/2022] [Indexed: 11/02/2022]
|
16
|
Aldosari DI, Malik A, Alhomida AS, Ola MS. Implications of Diabetes-Induced Altered Metabolites on Retinal Neurodegeneration. Front Neurosci 2022; 16:938029. [PMID: 35911994 PMCID: PMC9328693 DOI: 10.3389/fnins.2022.938029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Accepted: 06/09/2022] [Indexed: 11/13/2022] Open
Abstract
Diabetic retinopathy (DR) is one of the major complications of diabetic eye diseases, causing vision loss and blindness worldwide. The concept of diabetic retinopathy has evolved from microvascular disease into more complex neurovascular disorders. Early in the disease progression of diabetes, the neuronal and glial cells are compromised before any microvascular abnormalities clinically detected by the ophthalmoscopic examination. This implies understanding the pathophysiological mechanisms at the early stage of disease progression especially due to diabetes-induced metabolic alterations to damage the neural retina so that early intervention and treatments options can be identified to prevent and inhibit the progression of DR. Hyperglycemia has been widely considered the major contributor to the progression of the retinal damage, even though tight control of glucose does not seem to have a bigger effect on the incidence or progression of retinal damage that leads to DR. Emerging evidence suggests that besides diabetes-induced hyperglycemia, dyslipidemia and amino acid defects might be a major contributor to the progression of early neurovascular retinal damage. In this review, we have discussed recent advances in the alterations of key metabolites of carbohydrate, lipid, and amino acids and their implications for neurovascular damage in DR.
Collapse
|
17
|
Sun Y, Kong L, Zhang AH, Han Y, Sun H, Yan GL, Wang XJ. A Hypothesis From Metabolomics Analysis of Diabetic Retinopathy: Arginine-Creatine Metabolic Pathway May Be a New Treatment Strategy for Diabetic Retinopathy. Front Endocrinol (Lausanne) 2022; 13:858012. [PMID: 35399942 PMCID: PMC8987289 DOI: 10.3389/fendo.2022.858012] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Accepted: 03/01/2022] [Indexed: 12/31/2022] Open
Abstract
Diabetic retinopathy is one of the serious complications of diabetes, which the leading causes of blindness worldwide, and its irreversibility renders the existing treatment methods unsatisfactory. Early detection and timely intervention can effectively reduce the damage caused by diabetic retinopathy. Metabolomics is a branch of systems biology and a powerful tool for studying pathophysiological processes, which can help identify the characteristic metabolic changes marking the progression of diabetic retinopathy, discover potential biomarkers to inform clinical diagnosis and treatment. This review provides an update on the known metabolomics biomarkers of diabetic retinopathy. Through comprehensive analysis of biomarkers, we found that the arginine biosynthesis is closely related to diabetic retinopathy. Meanwhile, creatine, a metabolite with arginine as a precursor, has attracted our attention due to its important correlation with diabetic retinopathy. We discuss the possibility of the arginine-creatine metabolic pathway as a therapeutic strategy for diabetic retinopathy.
Collapse
Affiliation(s)
- Ye Sun
- National Chinmedomics Research Center and National Traditional Chinese Medicine (TCM) Key Laboratory of Serum Pharmacochemistry, Department of Pharmaceutical Analysis, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Ling Kong
- National Chinmedomics Research Center and National Traditional Chinese Medicine (TCM) Key Laboratory of Serum Pharmacochemistry, Department of Pharmaceutical Analysis, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Ai-Hua Zhang
- National Chinmedomics Research Center and National Traditional Chinese Medicine (TCM) Key Laboratory of Serum Pharmacochemistry, Department of Pharmaceutical Analysis, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Ying Han
- National Chinmedomics Research Center and National Traditional Chinese Medicine (TCM) Key Laboratory of Serum Pharmacochemistry, Department of Pharmaceutical Analysis, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Hui Sun
- National Chinmedomics Research Center and National Traditional Chinese Medicine (TCM) Key Laboratory of Serum Pharmacochemistry, Department of Pharmaceutical Analysis, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Guang-Li Yan
- National Chinmedomics Research Center and National Traditional Chinese Medicine (TCM) Key Laboratory of Serum Pharmacochemistry, Department of Pharmaceutical Analysis, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Xi-Jun Wang
- National Chinmedomics Research Center and National Traditional Chinese Medicine (TCM) Key Laboratory of Serum Pharmacochemistry, Department of Pharmaceutical Analysis, Heilongjiang University of Chinese Medicine, Harbin, China
- State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau, Macau SAR, China
- National Engineering Laboratory for the Development of Southwestern Endangered Medicinal Materials, Guangxi Botanical Garden of Medicinal Plant, Nanning, China
| |
Collapse
|
18
|
Effect of the ethyl acetate extract of Sophora flavescens Aiton on diabetic retinopathy based on untargeted retinal metabolomics. J Chromatogr B Analyt Technol Biomed Life Sci 2022; 1198:123233. [DOI: 10.1016/j.jchromb.2022.123233] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2021] [Revised: 02/15/2022] [Accepted: 03/26/2022] [Indexed: 12/22/2022]
|
19
|
Peters KS, Rivera E, Warden C, Harlow PA, Mitchell SL, Calcutt MW, Samuels DC, Brantley MA. Plasma Arginine and Citrulline are Elevated in Diabetic Retinopathy. Am J Ophthalmol 2022; 235:154-162. [PMID: 34587493 DOI: 10.1016/j.ajo.2021.09.021] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2020] [Revised: 09/16/2021] [Accepted: 09/17/2021] [Indexed: 12/20/2022]
Abstract
PURPOSE To determine if plasma levels of six arginine-related and citrulline-related metabolites (arginine, citrulline, asymmetric dimethylarginine [ADMA], ornithine, proline, and argininosuccinate) differ between patients with type 2 diabetes and diabetic retinopathy (DR) and type 2 diabetic controls or between patients with proliferative DR (PDR) and non-proliferative DR (NPDR). DESIGN Cross-sectional study. METHODS Adults with type 2 diabetes were recruited from the Vanderbilt Eye Institute. Exclusion criteria included non-diabetic retinal disease. Plasma metabolite levels were quantified in 159 diabetic controls and 156 DR patients (92 NPDR, 64 PDR) using isotope dilution liquid chromatography-tandem mass spectrometry (LC-MS/MS). Metabolite levels were compared using Wilcoxon Rank Sum test and logistic regressions adjusting for age, sex, hemoglobin A1c, diabetes duration, statin use, and angiotensin-converting enzyme inhibitor or angiotensin II receptor blocker use. A secondary analysis that included creatinine in the regression model was performed for the subset of patients with available creatinine values (135 diabetic controls, 100 DR patients [58 NPDR, 42 PDR]). RESULTS Multivariable logistic regression analyses determined that arginine (OR = 1.20, [1.06-1.38], P = .0067) and citrulline (OR = 1.53, [1.20-1.98], P = .0025) were significantly elevated in DR patients compared to diabetic controls. While ADMA differed between NPDR and PDR patients in the primary analysis (OR = 1.56, [1.15-2.16], P = .0051), it was not significantly different when adjusting for creatinine (OR = 1.30, [0.90-1.91], P = .15). CONCLUSIONS Plasma arginine and citrulline were significantly elevated in type 2 diabetic patients with DR compared to diabetic controls. None of the tested metabolites significantly differed between NPDR and PDR patients in the adjusted analysis.
Collapse
|
20
|
Bunch KL, Abdelrahman AA, Caldwell RB, Caldwell RW. Novel Therapeutics for Diabetic Retinopathy and Diabetic Macular Edema: A Pathophysiologic Perspective. Front Physiol 2022; 13:831616. [PMID: 35250632 PMCID: PMC8894892 DOI: 10.3389/fphys.2022.831616] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Accepted: 01/17/2022] [Indexed: 12/12/2022] Open
Abstract
Diabetic retinopathy (DR) and diabetic macular edema (DME) are retinal complications of diabetes that can lead to loss of vision and impaired quality of life. The current gold standard therapies for treatment of DR and DME focus on advanced disease, are invasive, expensive, and can trigger adverse side-effects, necessitating the development of more effective, affordable, and accessible therapies that can target early stage disease. The pathogenesis and pathophysiology of DR is complex and multifactorial, involving the interplay between the effects of hyperglycemia, hyperlipidemia, hypoxia, and production of reactive oxygen species (ROS) in the promotion of neurovascular dysfunction and immune cell polarization to a proinflammatory state. The pathophysiology of DR provides several therapeutic targets that have the potential to attenuate disease progression. Current novel DR and DME therapies under investigation include erythropoietin-derived peptides, inducers of antioxidant gene expression, activators of nitric oxide/cyclic GMP signaling pathways, and manipulation of arginase activity. This review aims to aid understanding of DR and DME pathophysiology and explore novel therapies that capitalize on our knowledge of these diabetic retinal complications.
Collapse
Affiliation(s)
- Katharine L. Bunch
- Department of Pharmacology and Toxicology, Medical College of Georgia, Augusta University, Augusta, GA, United States
- James and Jean Culver Vision Discovery Institute, Medical College of Georgia, Augusta University, Augusta, GA, United States
| | - Ammar A. Abdelrahman
- Department of Pharmacology and Toxicology, Medical College of Georgia, Augusta University, Augusta, GA, United States
- James and Jean Culver Vision Discovery Institute, Medical College of Georgia, Augusta University, Augusta, GA, United States
| | - Ruth B. Caldwell
- James and Jean Culver Vision Discovery Institute, Medical College of Georgia, Augusta University, Augusta, GA, United States
- Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, Augusta, GA, United States
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, GA, United States
| | - R. William Caldwell
- Department of Pharmacology and Toxicology, Medical College of Georgia, Augusta University, Augusta, GA, United States
- James and Jean Culver Vision Discovery Institute, Medical College of Georgia, Augusta University, Augusta, GA, United States
- *Correspondence: R. William Caldwell,
| |
Collapse
|
21
|
Du X, Yang L, Kong L, Sun Y, Shen K, Cai Y, Sun H, Zhang B, Guo S, Zhang A, Wang X. Metabolomics of various samples advancing biomarker discovery and pathogenesis elucidation for diabetic retinopathy. Front Endocrinol (Lausanne) 2022; 13:1037164. [PMID: 36387907 PMCID: PMC9646596 DOI: 10.3389/fendo.2022.1037164] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/05/2022] [Accepted: 10/10/2022] [Indexed: 11/28/2022] Open
Abstract
Diabetic retinopathy (DR) is a universal microvascular complication of diabetes mellitus (DM), which is the main reason for global sight damage/loss in middle-aged and/or older people. Current clinical analyses, like hemoglobin A1c, possess some importance as prognostic indicators for DR severity, but no effective circulating biomarkers are used for DR in the clinic currently, and studies on the latent pathophysiology remain lacking. Recent developments in omics, especially metabolomics, continue to disclose novel potential biomarkers in several fields, including but not limited to DR. Therefore, based on the overview of metabolomics, we reviewed progress in analytical technology of metabolomics, the prominent roles and the current status of biomarkers in DR, and the update of potential biomarkers in various DR-related samples via metabolomics, including tear as well as vitreous humor, aqueous humor, retina, plasma, serum, cerebrospinal fluid, urine, and feces. In this review, we underscored the in-depth analysis and elucidation of the common biomarkers in different biological samples based on integrated results, namely, alanine, lactate, and glutamine. Alanine may participate in and regulate glucose metabolism through stimulating N-methyl-D-aspartate receptors and subsequently suppressing insulin secretion, which is the potential pathogenesis of DR. Abnormal lactate could cause extensive oxidative stress and neuroinflammation, eventually leading to retinal hypoxia and metabolic dysfunction; on the other hand, high-level lactate may damage the structure and function of the retinal endothelial cell barrier via the G protein-coupled receptor 81. Abnormal glutamine indicates a disturbance of glutamate recycling, which may affect the activation of Müller cells and proliferation via the PPP1CA-YAP-GS-Gln-mTORC1 pathway.
Collapse
Affiliation(s)
- Xiaohui Du
- National Chinmedomics Research Center, National TCM Key Laboratory of Serum Pharmacochemistry, Metabolomics Laboratory, Department of Pharmaceutical Analysis, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Le Yang
- State Key Laboratory of Dampness Syndrome, the Second Affiliated Hospital Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Ling Kong
- National Chinmedomics Research Center, National TCM Key Laboratory of Serum Pharmacochemistry, Metabolomics Laboratory, Department of Pharmaceutical Analysis, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Ye Sun
- National Chinmedomics Research Center, National TCM Key Laboratory of Serum Pharmacochemistry, Metabolomics Laboratory, Department of Pharmaceutical Analysis, Heilongjiang University of Chinese Medicine, Harbin, China
- State Key Laboratory of Dampness Syndrome, the Second Affiliated Hospital Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Kunshuang Shen
- National Chinmedomics Research Center, National TCM Key Laboratory of Serum Pharmacochemistry, Metabolomics Laboratory, Department of Pharmaceutical Analysis, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Ying Cai
- National Chinmedomics Research Center, National TCM Key Laboratory of Serum Pharmacochemistry, Metabolomics Laboratory, Department of Pharmaceutical Analysis, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Hui Sun
- National Chinmedomics Research Center, National TCM Key Laboratory of Serum Pharmacochemistry, Metabolomics Laboratory, Department of Pharmaceutical Analysis, Heilongjiang University of Chinese Medicine, Harbin, China
- *Correspondence: Hui Sun, ; Xijun Wang,
| | - Bo Zhang
- National Chinmedomics Research Center, National TCM Key Laboratory of Serum Pharmacochemistry, Metabolomics Laboratory, Department of Pharmaceutical Analysis, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Sifan Guo
- National Chinmedomics Research Center, National TCM Key Laboratory of Serum Pharmacochemistry, Metabolomics Laboratory, Department of Pharmaceutical Analysis, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Aihua Zhang
- National Chinmedomics Research Center, National TCM Key Laboratory of Serum Pharmacochemistry, Metabolomics Laboratory, Department of Pharmaceutical Analysis, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Xijun Wang
- National Chinmedomics Research Center, National TCM Key Laboratory of Serum Pharmacochemistry, Metabolomics Laboratory, Department of Pharmaceutical Analysis, Heilongjiang University of Chinese Medicine, Harbin, China
- State Key Laboratory of Dampness Syndrome, the Second Affiliated Hospital Guangzhou University of Chinese Medicine, Guangzhou, China
- State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau, Macau SAR, China
- *Correspondence: Hui Sun, ; Xijun Wang,
| |
Collapse
|
22
|
Buraczynska M, Zakrocka I. Arginase Gene Polymorphism Increases Risk of Diabetic Retinopathy in Type 2 Diabetes Mellitus Patients. J Clin Med 2021; 10:jcm10225407. [PMID: 34830689 PMCID: PMC8620112 DOI: 10.3390/jcm10225407] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Revised: 11/16/2021] [Accepted: 11/16/2021] [Indexed: 12/22/2022] Open
Abstract
Studies have demonstrated that polymorphic variants of arginase 1 gene (ARG1) are involved in human diseases, such as coronary heart disease, hypertension, and diabetes. Our study aimed to investigate the association between ARG1 rs2781666 single nucleotide polymorphism (SNP) and diabetic retinopathy (DR) in type 2 diabetes (T2DM) patients. Polymorphism was genotyped in 740 T2DM patients and 400 healthy individuals. A significant difference in the genotype distribution was observed between the patients and the controls. The T allele and TT genotype were associated with an increased risk of T2DM (OR 1.4, 95% CI 1.14–1.72, p = 0.001 and OR 2.16, 95% CI 1.23–3.80, p = 0.007, respectively). When the T2DM subjects were stratified into DR+ and DR− subgroups, the T allele and TT genotype frequencies were significantly higher in the DR+ group compared to the DR− group, demonstrating OR 1.68 (1.33–2.12), p < 0.0001 and OR 2.39 (1.36–4.18), p = 0.002, respectively. Logistic regression analysis was applied to determine the interaction between the ARG1 genotypes and other risk factors. Only ARG1 rs2781666 SNP was a significant risk predictor of DR (p = 0.003). In conclusion, this is the first report discussing the effect of ARG1 polymorphism on the microvascular complications that are associated with diabetes. Our findings demonstrate that ARG1 rs2781666 SNP is significantly associated with an increased susceptibility to DR in T2DM patients.
Collapse
|
23
|
Abstract
Purpose Diabetic retinopathy (DR), a common microvascular complication of diabetes, is the leading cause of acquired blindness in the working-age population. Individuals with diabetes still develop DR despite appropriate glycemic and blood pressure control, highlighting the pressing need to identify useful biomarkers for risk stratification. The purpose of this review is to systematically summarize potential metabolic biomarkers and pathways of DR, which could facilitate developing an understanding of the disease mechanisms, as well as new therapeutic measures. Methods We searched PubMed and Web of Science for relevant metabolomics studies on humans published before September 30, 2020. Information regarding authors, title, publication date, study subjects, analytical platforms, methods of statistical analysis, biological samples, directions of change of potential metabolic biomarkers, and predictive values of metabolic biomarker panels was extracted, and the quality of the studies was assessed. Pathway analysis, including enrichment analysis and topology analysis, was derived from integrating differential metabolites using MetaboAnalyst 3.0, based on the Kyoto Encyclopedia of Genes and Genomes and Human Metabolome Database. Results We found nine studies focused on the identification of potential biomarkers. Repeatedly identified metabolites including l-glutamine, l-lactic acid, pyruvic acid, acetic acid, l-glutamic acid, d-glucose, l-alanine, l-threonine, citrulline, l-lysine, and succinic acid were found to be potential biomarkers of DR. It was observed that l-glutamine and citrulline changed in all biological samples. Dysregulation of metabolic pathways involved amino acid and energy metabolism. Conclusions This review summarizes potential biomarkers and metabolic pathways, providing insights into new pathogenic pathways for this microvascular complication of diabetes.
Collapse
Affiliation(s)
- Xiao-Wen Hou
- School of Public Health, Medical College of Soochow University, Suzhou, China
| | - Ying Wang
- School of Public Health, Medical College of Soochow University, Suzhou, China
| | - Chen-Wei Pan
- School of Public Health, Medical College of Soochow University, Suzhou, China
| |
Collapse
|
24
|
Development of a new nano arginase HPLC capillary column for the fast screening of arginase inhibitors and evaluation of their binding affinity. J Chromatogr B Analyt Technol Biomed Life Sci 2021; 1175:122751. [PMID: 33991957 DOI: 10.1016/j.jchromb.2021.122751] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Revised: 04/26/2021] [Accepted: 04/27/2021] [Indexed: 11/21/2022]
Abstract
A simple and rapid Nano LC method has been developed for the screening of arginase inhibitors. The method is based on the immobilization of biotinylated arginase on a neutravidin functionalized nano HPLC capillary column. The arginase immobilization step performed by frontal analysis is very fast and only takes a few minutes. The miniaturized capillary column of 170 nL (length 5 cm, internal diameter 75 μm) significantly decreased the required amount of used enzyme (25 pmol). This was of significance importance when working with less available or expensive purified enzyme. Non-selective adsorption of the organic monolith matrix was reduced (<6%) and the arginase efficient yield was high (92%). The resultant affinity capillary columns showed excellent repeatability and long lifetime. The arginase reaction product was achieved within 60 s and the immobilized arginase retained 97% of the initial activity beyond 90 days. This novel approach can thus be used for the fast evaluation of recognition assay induced bya series of inhibitor molecules (caffeic acid phenylamide, chlorogenic acid, piceatannol, nor-NOHA acetate) and plant extracts.
Collapse
|
25
|
Hydrogen Sulfide: Novel Endogenous and Exogenous Modulator of Oxidative Stress in Retinal Degeneration Diseases. Molecules 2021; 26:molecules26092411. [PMID: 33919146 PMCID: PMC8122398 DOI: 10.3390/molecules26092411] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Revised: 04/14/2021] [Accepted: 04/19/2021] [Indexed: 11/29/2022] Open
Abstract
Oxidative stress (OS) damage can cause significant injury to cells, which is related to the occurrence and development of many diseases. This pathological process is considered to be the first step to trigger the death of outer retinal neurons, which is related to the pathology of retinal degenerative diseases. Hydrogen sulfide (H2S) has recently received widespread attention as a physiological signal molecule and gas neuromodulator and plays an important role in regulating OS in eyes. In this article, we reviewed the OS responses and regulatory mechanisms of H2S and its donors as endogenous and exogenous regulators in retinal degenerative diseases. Understanding the relevant mechanisms will help to identify the therapeutic potential of H2S in retinal degenerative diseases.
Collapse
|
26
|
Catalani E, Silvestri F, Bongiorni S, Taddei AR, Fanelli G, Rinalducci S, De Palma C, Perrotta C, Prantera G, Cervia D. Retinal damage in a new model of hyperglycemia induced by high-sucrose diets. Pharmacol Res 2021; 166:105488. [PMID: 33582248 DOI: 10.1016/j.phrs.2021.105488] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Revised: 02/08/2021] [Accepted: 02/08/2021] [Indexed: 12/13/2022]
Abstract
Loss of retinal neurons may precede clinical signs of diabetic retinopathy (DR). We studied for the first time the effects of hyperglycemia on the visual system of the fruit fly Drosophila melanogaster to characterize a model for glucose-induced retinal neurodegeneration, thus complementing more traditional vertebrate systems. Adult flies were fed with increased high-sucrose regimens which did not modify the locomotion ability, muscle phenotype and mobility after 10 days. The increased availability of dietary sucrose induced hyperglycemia and phosphorylation of Akt in fat tissue, without significant effects on adult growth and viability, consistent with the early phase of insulin signaling and a low impact on the overall metabolic profile of flies at short term. Noteworthy, high-sucrose diets significantly decreased Drosophila responsiveness to the light as a consequence of vision defects. Hyperglycemia did not alter the gross anatomical architecture of the external eye phenotype although a progressive damage of photosensitive units was observed. Appreciable levels of cleaved caspase 3 and nitrotyrosine were detected in the internal retina network as well as punctate staining of Light-Chain 3 and p62, and accumulated autophagosomes, indicating apoptotic features, peroxynitrite formation and autophagy turnover defects. In summary, our results in Drosophila support the view that hyperglycemia induced by high-sucrose diets lead to eye defects, apoptosis/autophagy dysregulation, oxidative stress, and visual dysfunctions which are evolutionarily conserved, thus offering a meaningful opportunity of using a simple in vivo model to study the pathophysiology of neuroretinal alterations that develop in patients at the early stages of DR.
Collapse
Affiliation(s)
- Elisabetta Catalani
- Department for Innovation in Biological, Agro-food and Forest Systems (DIBAF), Università degli Studi della Tuscia, largo dell'Università snc, 01100 Viterbo, Italy
| | - Federica Silvestri
- Department for Innovation in Biological, Agro-food and Forest Systems (DIBAF), Università degli Studi della Tuscia, largo dell'Università snc, 01100 Viterbo, Italy
| | - Silvia Bongiorni
- Department of Ecological and Biological Sciences (DEB), Università degli Studi della Tuscia, largo dell'Università snc, 01100 Viterbo, Italy
| | - Anna Rita Taddei
- Section of Electron Microscopy, Great Equipment Center, Università degli Studi della Tuscia, largo dell'Università snc, 01100 Viterbo, Italy
| | - Giuseppina Fanelli
- Department of Ecological and Biological Sciences (DEB), Università degli Studi della Tuscia, largo dell'Università snc, 01100 Viterbo, Italy
| | - Sara Rinalducci
- Department of Ecological and Biological Sciences (DEB), Università degli Studi della Tuscia, largo dell'Università snc, 01100 Viterbo, Italy
| | - Clara De Palma
- Department of Medical Biotechnology and Translational Medicine (BioMeTra), Università degli Studi di Milano, via L. Vanvitelli 32, 20129 Milano, Italy
| | - Cristiana Perrotta
- Department of Biomedical and Clinical Sciences "Luigi Sacco" (DIBIC), Università degli Studi di Milano, via G.B. Grassi 74, 20157 Milano, Italy
| | - Giorgio Prantera
- Department of Ecological and Biological Sciences (DEB), Università degli Studi della Tuscia, largo dell'Università snc, 01100 Viterbo, Italy
| | - Davide Cervia
- Department for Innovation in Biological, Agro-food and Forest Systems (DIBAF), Università degli Studi della Tuscia, largo dell'Università snc, 01100 Viterbo, Italy.
| |
Collapse
|
27
|
Xuan Q, Ouyang Y, Wang Y, Wu L, Li H, Luo Y, Zhao X, Feng D, Qin W, Hu C, Zhou L, Liu X, Zou H, Cai C, Wu J, Jia W, Xu G. Multiplatform Metabolomics Reveals Novel Serum Metabolite Biomarkers in Diabetic Retinopathy Subjects. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2020; 7:2001714. [PMID: 33240754 PMCID: PMC7675050 DOI: 10.1002/advs.202001714] [Citation(s) in RCA: 67] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Revised: 08/10/2020] [Indexed: 05/12/2023]
Abstract
Diabetic retinopathy (DR) is the main cause of vision loss or blindness in working age adults worldwide. The lack of effective diagnostic biomarkers for DR leads to unsatisfactory curative treatments. To define potential metabolite biomarkers for DR diagnosis, a multiplatform-based metabolomics study is performed. In this study, a total of 905 subjects with diabetes without DR (NDR) and with DR at different clinical stages are recruited. Multiplatform metabolomics methods are used to characterize the serum metabolic profiles and to screen and validate the DR biomarkers. Based on the criteria p < 0.05 and false-discovery rate < 0.05, 348 and 290 metabolites are significantly associated with the pathogenesis of DR and early-stage DR, respectively. The biomarker panel consisting of 12-hydroxyeicosatetraenoic acid (12-HETE) and 2-piperidone exhibited better diagnostic performance than hemoglobin A1c (HbA1c) in differentiating DR from diabetes, with AUCs of 0.946 versus 0.691 and 0.928 versus 0.648 in the discovery and validation sets, respectively. In addition, this panel showed higher sensitivity in early-stage DR detection than HbA1c. In conclusion, this multiplatform-based metabolomics study comprehensively revealed the metabolic dysregulation associated with DR onset and progression. The defined biomarker panel can be used for detection of DR and early-stage DR.
Collapse
Affiliation(s)
- Qiuhui Xuan
- CAS Key Laboratory of Separation Science for Analytical ChemistryDalian Institute of Chemical PhysicsChinese Academy of Sciences457 Zhongshan RoadDalian116023China
- University of Chinese Academy of SciencesBeijing100049China
| | - Yang Ouyang
- CAS Key Laboratory of Separation Science for Analytical ChemistryDalian Institute of Chemical PhysicsChinese Academy of Sciences457 Zhongshan RoadDalian116023China
- University of Chinese Academy of SciencesBeijing100049China
| | - Yanfeng Wang
- CAS Key Laboratory of Separation Science for Analytical ChemistryDalian Institute of Chemical PhysicsChinese Academy of Sciences457 Zhongshan RoadDalian116023China
- University of Chinese Academy of SciencesBeijing100049China
| | - Liang Wu
- Shanghai Diabetes Institute, Shanghai Key Laboratory of Diabetes Mellitus, Shanghai Clinical Center for Endocrine and Metabolic Diseases, Metabolic diseases biobankShanghai JiaoTong University Affiliated Sixth People's HospitalShanghai200233China
| | - Huating Li
- Shanghai Diabetes Institute, Shanghai Key Laboratory of Diabetes Mellitus, Shanghai Clinical Center for Endocrine and Metabolic Diseases, Metabolic diseases biobankShanghai JiaoTong University Affiliated Sixth People's HospitalShanghai200233China
| | - Yuanyuan Luo
- CAS Key Laboratory of Separation Science for Analytical ChemistryDalian Institute of Chemical PhysicsChinese Academy of Sciences457 Zhongshan RoadDalian116023China
- University of Chinese Academy of SciencesBeijing100049China
| | - Xinjie Zhao
- CAS Key Laboratory of Separation Science for Analytical ChemistryDalian Institute of Chemical PhysicsChinese Academy of Sciences457 Zhongshan RoadDalian116023China
| | - Disheng Feng
- CAS Key Laboratory of Separation Science for Analytical ChemistryDalian Institute of Chemical PhysicsChinese Academy of Sciences457 Zhongshan RoadDalian116023China
- University of Chinese Academy of SciencesBeijing100049China
| | - Wangshu Qin
- CAS Key Laboratory of Separation Science for Analytical ChemistryDalian Institute of Chemical PhysicsChinese Academy of Sciences457 Zhongshan RoadDalian116023China
| | - Chunxiu Hu
- CAS Key Laboratory of Separation Science for Analytical ChemistryDalian Institute of Chemical PhysicsChinese Academy of Sciences457 Zhongshan RoadDalian116023China
| | - Lina Zhou
- CAS Key Laboratory of Separation Science for Analytical ChemistryDalian Institute of Chemical PhysicsChinese Academy of Sciences457 Zhongshan RoadDalian116023China
| | - Xinyu Liu
- CAS Key Laboratory of Separation Science for Analytical ChemistryDalian Institute of Chemical PhysicsChinese Academy of Sciences457 Zhongshan RoadDalian116023China
| | - Haidong Zou
- Department of OphthalmologyFirst People's Hospital of ShanghaiShanghai Jiao Tong UniversityShanghaiChina
| | - Chun Cai
- Shanghai Diabetes Institute, Shanghai Key Laboratory of Diabetes Mellitus, Shanghai Clinical Center for Endocrine and Metabolic Diseases, Metabolic diseases biobankShanghai JiaoTong University Affiliated Sixth People's HospitalShanghai200233China
| | - Jiarui Wu
- Key Laboratory of Systems Biology, CAS Center for Excellence in Molecular Cell Science, Institute of Biochemistry and Cell Biology, University of Chinese Academy of SciencesChinese Academy of Sciences320 Yue‐Yang RoadShanghai200031China
- Key Laboratory of Systems BiologyCAS Center for Excellence in Molecular Cell ScienceInstitute of Biochemistry and Cell BiologyUniversity of Chinese Academy of SciencesChinese Academy of Sciences320 Yue‐Yang RoadShanghai200031China
| | - Weiping Jia
- Shanghai Diabetes Institute, Shanghai Key Laboratory of Diabetes Mellitus, Shanghai Clinical Center for Endocrine and Metabolic Diseases, Metabolic diseases biobankShanghai JiaoTong University Affiliated Sixth People's HospitalShanghai200233China
- Shanghai Diabetes InstituteShanghai Key Laboratory of Diabetes MellitusShanghai Clinical Center for Endocrine and Metabolic DiseasesShanghai Jiaotong University Affiliated Sixth People's HospitalShanghai200233China
| | - Guowang Xu
- CAS Key Laboratory of Separation Science for Analytical ChemistryDalian Institute of Chemical PhysicsChinese Academy of Sciences457 Zhongshan RoadDalian116023China
- University of Chinese Academy of SciencesBeijing100049China
- CAS Key Laboratory of Separation Science for Analytical ChemistryDalian Institute of Chemical PhysicsChinese Academy of SciencesDalian116023China
| |
Collapse
|
28
|
Luo Y, Cui HP, Liu Y, Chen L. Metabolomics and biomarkers in ocular matrix: beyond ocular diseases. Int J Ophthalmol 2020; 13:991-1003. [PMID: 32566514 DOI: 10.18240/ijo.2020.06.21] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2020] [Accepted: 03/23/2020] [Indexed: 12/15/2022] Open
Abstract
According to the recent report, there are 870 million people suffer from ocular diseases worldwide. The present approaches for diagnosis are morphological examination, imaging examination and immunological examination, regrettably, they lack of sensitivity and difficult to make a definite diagnosis in the early stage. Systemic biology as an effective method has been used in clinical diagnosis and treatment for diseases, especially metabolomics which is more attractive with high sensitivity and accuracy. Although previous researches had been confirmed that endogenous metabolites in the ocular matrix play a crucial role in the progress of diseases related diseases, the standard protocols and systematic summary about the biomarker researches based on ocular matrix has not been established. This review article highlights the pretreatment for ocular matrix and the new biomarkers expressed by the eye diseases, expected to promote the application of biomarkers in the diagnosis and treatment of eye diseases.
Collapse
Affiliation(s)
- Yun Luo
- School of Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou 510006, Guangdong Province, China
| | - Hong-Pei Cui
- Department of Ophthalmology, Henan Provincial People's Hospital, Zhengzhou 450003, Henan Province, China
| | - Yi Liu
- School of Chinese Medicine, Southern Medical University, Guangzhou 510515, Guangdong Province, China
| | - Lei Chen
- School of Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou 510006, Guangdong Province, China
| |
Collapse
|
29
|
Akintunde JK, Akintola TE, Hammed MO, Amoo CO, Adegoke AM, Ajisafe LO. Naringin protects against Bisphenol-A induced oculopathy as implication of cataract in hypertensive rat model. Biomed Pharmacother 2020; 126:110043. [PMID: 32172062 DOI: 10.1016/j.biopha.2020.110043] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2020] [Revised: 02/14/2020] [Accepted: 02/23/2020] [Indexed: 12/12/2022] Open
Abstract
People who have experienced high blood pressure are at greater risk of susceptibility to other health problems including oculopathy. The patients with these experiences do not have adequate treatment and those who do; spend much funds on the drug purchase. The study examines the protective effect of naringin (NRG) against ocular impairment in L-NAME induced hypertensive rat on exposure to a cellular disruptor. Fifty-six adult male albino rats were randomly distributed into eight (n = 7) groups. Group I: control animals, Group II was treated with Nω-nitro-L-arginine methyl ester hydrochloride (L-NAME), Group III was treated with 50 mg/kg Bisphenol-A, Group IV was treated with L-NAME +50 mg/kg Bisphenol-A. Group V was administered with L-NAME +80 mg/kg NRG. Group VI was administered with 50 Mg/kg BPA + 80 mg/kg NRG. Group VII was administered with L-NAME+50 mg/kg Bisphenol-A +80 mg/kg NRG. Lastly, group VIII was treated with 80 mg/kg NRG alone for 14 days. Naringin prevented hypertension and ocular dysfunction by depleting the activities of angiotensin-converting enzymes, arginase, aldose-reductase and phosphodiesterase-51 (PDE-51) with corresponding down-regulation of inflammatory markers including TNF-α and IL-B. Moreover, ocular impairment was remarkably reduced by NRG as manifested by the decreased activities of AChE, BuChE, MAO-A and enzymes of ATP hydrolysis (ATPase, ADPase, AMPase) and adenosine deaminase with resultant increased NO level. Also, ocular expression of CD43 transcript, caspaace-9 and tumor suppressor P53 proteins were suppressed on treatment with NRG. This study corroborates the view that NRG may be a useful therapy in alleviating inflammatory markers, apoptosis and metabolic nucleotides disorders via the NOS/cGMP/PKG signaling pathways in hypertensive rat model on exposure to a cellular disruptor.
Collapse
Affiliation(s)
- J K Akintunde
- Applied Biochemistry and Molecular Toxicology Research Group, Department of Biochemistry, College of Biosciences, Federal University of Agriculture, Abeokuta, Nigeria.
| | - T E Akintola
- Applied Biochemistry and Molecular Toxicology Research Group, Department of Biochemistry, College of Biosciences, Federal University of Agriculture, Abeokuta, Nigeria
| | - M O Hammed
- Applied Biochemistry and Molecular Toxicology Research Group, Department of Biochemistry, College of Biosciences, Federal University of Agriculture, Abeokuta, Nigeria
| | - C O Amoo
- Applied Biochemistry and Molecular Toxicology Research Group, Department of Biochemistry, College of Biosciences, Federal University of Agriculture, Abeokuta, Nigeria
| | - A M Adegoke
- Cancer Research and Molecular Biology Laboratories, Department of Biochemistry, Faculty of Basic Medical Sciences, College of Medicine, University of Ibadan, Nigeria
| | - L O Ajisafe
- Cancer Research and Molecular Biology Laboratories, Department of Biochemistry, Faculty of Basic Medical Sciences, College of Medicine, University of Ibadan, Nigeria
| |
Collapse
|
30
|
Liu X, Zhang Y, Liang H, Zhang Y, Xu Y. microRNA-499-3p inhibits proliferation and promotes apoptosis of retinal cells in diabetic retinopathy through activation of the TLR4 signaling pathway by targeting IFNA2. Gene 2020; 741:144539. [PMID: 32160960 DOI: 10.1016/j.gene.2020.144539] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2019] [Accepted: 03/06/2020] [Indexed: 12/21/2022]
Abstract
microRNAs (miRNAs) are involved in the physiological and pathophysiological processes of diabetes and its microvascular and macrovascular complications. Hence, the aim of the study was to investigate whether miR-499-3p played an important role in diabetic retinopathy. Diabetic retinopathy was developed in rats by intraperitoneal injection of streptozocin (STZ), followed by collection of retinal tissues and preparation of retinal cells. Immunohistochemical staining was used to detect expression of interferon alpha 2 (IFNA2). RT-qPCR was used to determine the expression of miR-499-3p. Bioinformatics website and dual luciferase reporter gene assay were used to validate the targeting relationship between miR-499-3p and IFNA2. Gain- and loss-of-function assays were performed to explore the functional roles of aberrantly expressed miR-499-3p and IFNA2 in retinal cell proliferation by MTT, and apoptosis by flow cytometry. In retinal tissues and cells of diabetic rats, IFNA2 expression was reduced, and miR-499-3p expression increased to activate the toll-like receptor 4 (TLR4) signaling pathway. IFNA2 was a target gene of miR-499-3p and negatively regulated by miR-499-3p. Further, downregulated miR-499-3p promoted retinal cell proliferation while suppressing apoptosis to alleviate diabetic retinopathy. All in all, miR-499-3p promoted retinopathy by enhancing activation of the TLR4 signaling pathway, which provides a new therapeutic target for diabetic retinopathy.
Collapse
Affiliation(s)
- Xiaomeng Liu
- Department of Endocrinology (2(nd) Ward), Linyi People's Hospital, Linyi 276000, PR China
| | - Yuanyuan Zhang
- Department of Endocrinology (2(nd) Ward), Linyi People's Hospital, Linyi 276000, PR China
| | - Hongwei Liang
- Department of Healthcare, Linyi People's Hospital, Linyi 276000, PR China
| | - Yusong Zhang
- Department of Image, Linyi People's Hospital, Linyi 276000, PR China
| | - Yanchao Xu
- Department of Endocrinology (2(nd) Ward), Linyi People's Hospital, Linyi 276000, PR China.
| |
Collapse
|
31
|
Is the Arginase Pathway a Novel Therapeutic Avenue for Diabetic Retinopathy? J Clin Med 2020; 9:jcm9020425. [PMID: 32033258 PMCID: PMC7073619 DOI: 10.3390/jcm9020425] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Accepted: 01/31/2020] [Indexed: 12/12/2022] Open
Abstract
Diabetic retinopathy (DR) is the leading cause of blindness in working age Americans. Clinicians diagnose DR based on its characteristic vascular pathology, which is evident upon clinical exam. However, extensive research has shown that diabetes causes significant neurovascular dysfunction prior to the development of clinically apparent vascular damage. While laser photocoagulation and/or anti-vascular endothelial growth factor (VEGF) therapies are often effective for limiting the late-stage vascular pathology, we still do not have an effective treatment to limit the neurovascular dysfunction or promote repair during the early stages of DR. This review addresses the role of arginase as a mediator of retinal neurovascular injury and therapeutic target for early stage DR. Arginase is the ureohydrolase enzyme that catalyzes the production of L-ornithine and urea from L-arginine. Arginase upregulation has been associated with inflammation, oxidative stress, and peripheral vascular dysfunction in models of both types of diabetes. The arginase enzyme has been identified as a therapeutic target in cardiovascular disease and central nervous system disease including stroke and ischemic retinopathies. Here, we discuss and review the literature on arginase-induced retinal neurovascular dysfunction in models of DR. We also speculate on the therapeutic potential of arginase in DR and its related underlying mechanisms.
Collapse
|
32
|
Tekin K, Tekin MI. Oxidative stress and diabetic retinopathy. Pathology 2020. [DOI: 10.1016/b978-0-12-815972-9.00003-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
33
|
Azul L, Leandro A, Boroumand P, Klip A, Seiça R, Sena CM. Increased inflammation, oxidative stress and a reduction in antioxidant defense enzymes in perivascular adipose tissue contribute to vascular dysfunction in type 2 diabetes. Free Radic Biol Med 2020; 146:264-274. [PMID: 31698080 DOI: 10.1016/j.freeradbiomed.2019.11.002] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/03/2019] [Revised: 10/19/2019] [Accepted: 11/03/2019] [Indexed: 01/27/2023]
Abstract
BACKGROUND Perivascular adipose tissue (PVAT) surrounds most large blood vessels and plays an important role in vascular homeostasis. The present study was conducted to investigate the contribution of PVAT to vascular dysfunction in a rat model of type 2 diabetes. MATERIAL AND METHODS Several in vivo parameters such as lipid profile (total cholesterol and triglyceride systemic levels), fasting glucose levels, glucose tolerance and insulin sensitivity (through glucose and insulin tolerance tests, respectively) were determined in Goto-Kakizaki (GK) diabetic rats and compared with control Wistar rats. At the vascular level, endothelial dependent and independent relaxation and contraction studies were performed in aortic rings in the absence (PVAT-) or in the presence (PVAT+) of thoracic PVAT. We also evaluated vascular oxidative stress and performed western blots, PCR and immunohistochemistry analysis of cytokines and various enzymes in PVAT. RESULTS Endothelium-dependent relaxation to acetylcholine, assessed by wire myography, was impaired in GK rats and improved by the antioxidant TEMPOL and by the TLR4 inhibitor, CLI-095 suggesting an increase in oxidative stress and inflammation. In addition, vascular superoxide and peroxynitrite production was increased in the vascular wall of diabetic rats, accompanied by reduced nitric oxide bioavailability. The presence of PVAT had an anticontractile effect in response to phenylephrine in Wistar rats that was lost in GK rats. Western blot and immunohistochemistry analysis revealed that PVAT phenotype shifts, under diabetic conditions, towards a proinflammatory (with increment in CRP, CCL2, CD36), pro-oxidant (increased levels of aldose reductase, and reduced levels of antioxidant deference enzymes) and vasoconstriction state. CONCLUSION Our data suggest that this rat model of type 2 diabetes is associated with perivascular adipose dysfunction that contributes to oxidative stress, inflammation and endothelial dysfunction.
Collapse
Affiliation(s)
- Lara Azul
- Institute of Physiology, iCBR, Faculty of Medicine, University of Coimbra, Portugal
| | - Adriana Leandro
- Institute of Physiology, iCBR, Faculty of Medicine, University of Coimbra, Portugal
| | - Parastoo Boroumand
- Cell Biology Program, The Hospital for Sick Children, Toronto, Ontario, M5G 0A4, Canada; Department of Biochemistry, University of Toronto, Toronto, Ontario, M5S 1A8, Canada
| | - Amira Klip
- Cell Biology Program, The Hospital for Sick Children, Toronto, Ontario, M5G 0A4, Canada; Department of Biochemistry, University of Toronto, Toronto, Ontario, M5S 1A8, Canada
| | - Raquel Seiça
- Institute of Physiology, iCBR, Faculty of Medicine, University of Coimbra, Portugal
| | - Cristina M Sena
- Institute of Physiology, iCBR, Faculty of Medicine, University of Coimbra, Portugal.
| |
Collapse
|
34
|
Wang H, Fang J, Chen F, Sun Q, Xu X, Lin SH, Liu K. Metabolomic profile of diabetic retinopathy: a GC-TOFMS-based approach using vitreous and aqueous humor. Acta Diabetol 2020; 57:41-51. [PMID: 31089930 DOI: 10.1007/s00592-019-01363-0] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/25/2019] [Accepted: 05/03/2019] [Indexed: 12/12/2022]
Abstract
AIM To identify the potential metabolite markers in diabetic retinopathy (DR) by using gas chromatography coupled with time-of-flight mass spectrometry (GC-TOFMS). METHODS GC-TOFMS spectra were acquired from vitreous and aqueous humor (AH) samples of patients with DR and non-diabetic participants. Comparative analysis was used to elucidate the distinct metabolites of DR. Metabolic pathway was employed to explicate the metabolic reprogramming pathways involved in DR. Logistic regression and receiver-operating characteristic analyses were carried out to select and validate the biomarker metabolites and establish a therapeutic model. RESULTS Comparative analysis showed a clear separation between disease and control groups. Eight differentiating metabolites from AH and 15 differentiating metabolites from vitreous were highlighted. Out of these 23 metabolites, 11 novel metabolites have not been detected previously. Pathway analysis identified nine pathways (three in AH and six in vitreous) as the major disturbed pathways associated with DR. The abnormal of gluconeogenesis, ascorbate-aldarate metabolism, valine-leucine-isoleucine biosynthesis, and arginine-proline metabolism might weigh the most in the development of DR. The AUC of the logistic regression model established by D-2,3-Dihydroxypropanoic acid, isocitric acid, fructose 6-phosphate, and L-Lactic acid in AH was 0.965. The AUC established by pyroglutamic acid and pyruvic acid in vitreous was 0.951. CONCLUSIONS These findings have expanded our understanding of identified metabolites and revealed for the first time some novel metabolites in DR. These results may provide useful information to explore the mechanism and may eventually allow the development of metabolic biomarkers for prognosis and novel therapeutic strategies for the management of DR.
Collapse
Affiliation(s)
- Haiyan Wang
- Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Key Laboratory of Ocular Fundus Diseases, Shanghai Engineering Center for Visual Science and Photomedicine, Shanghai, China
| | - Junwei Fang
- College of Basic Medical Sciences, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Fenge Chen
- Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Key Laboratory of Ocular Fundus Diseases, Shanghai Engineering Center for Visual Science and Photomedicine, Shanghai, China
| | - Qian Sun
- Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Key Laboratory of Ocular Fundus Diseases, Shanghai Engineering Center for Visual Science and Photomedicine, Shanghai, China
| | - Xiaoyin Xu
- Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Key Laboratory of Ocular Fundus Diseases, Shanghai Engineering Center for Visual Science and Photomedicine, Shanghai, China
| | - Shu-Hai Lin
- College of Basic Medical Sciences, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, Xiamen, China.
| | - Kun Liu
- Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Key Laboratory of Ocular Fundus Diseases, Shanghai Engineering Center for Visual Science and Photomedicine, Shanghai, China.
| |
Collapse
|
35
|
Sumarriva K, Uppal K, Ma C, Herren DJ, Wang Y, Chocron IM, Warden C, Mitchell SL, Burgess LG, Goodale MP, Osborn MP, Ferreira AJ, Law JC, Cherney EF, Jones DP, Brantley MA. Arginine and Carnitine Metabolites Are Altered in Diabetic Retinopathy. Invest Ophthalmol Vis Sci 2019; 60:3119-3126. [PMID: 31323682 PMCID: PMC6645705 DOI: 10.1167/iovs.19-27321] [Citation(s) in RCA: 61] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Purpose To determine plasma metabolite and metabolic pathway differences between patients with type 2 diabetes with diabetic retinopathy (DR) and without retinopathy (diabetic controls), and between patients with proliferative DR (PDR) and nonproliferative DR (NPDR). Methods Using high-resolution mass spectrometry with liquid chromatography, untargeted metabolomics was performed on plasma samples from 83 DR patients and 90 diabetic controls. Discriminatory metabolic features were identified through partial least squares discriminant analysis, and linear regression was used to adjust for age, sex, diabetes duration, and hemoglobin A1c. Pathway analysis was performed using Mummichog 2.0. Results In the adjusted analysis, 126 metabolic features differed significantly between DR patients and diabetic controls. Pathway analysis revealed alterations in the metabolism of amino acids, leukotrienes, niacin, pyrimidine, and purine. Arginine, citrulline, glutamic γ-semialdehyde, and dehydroxycarnitine were key contributors to these pathway differences. A total of 151 features distinguished PDR patients from NPDR patients, and pathway analysis revealed alterations in the β-oxidation of saturated fatty acids, fatty acid metabolism, and vitamin D3 metabolism. Carnitine was a major contributor to the pathway differences. Conclusions This study demonstrates that arginine and citrulline-related pathways are dysregulated in DR, and fatty acid metabolism is altered in PDR patients compared with NPDR patients.
Collapse
Affiliation(s)
- Katherine Sumarriva
- Vanderbilt Eye Institute, Vanderbilt University Medical Center, Nashville, Tennessee, United States
| | - Karan Uppal
- Department of Medicine, Emory University, Atlanta, Georgia, United States
| | - Chunyu Ma
- Department of Medicine, Emory University, Atlanta, Georgia, United States
| | - David J Herren
- Vanderbilt Eye Institute, Vanderbilt University Medical Center, Nashville, Tennessee, United States
| | - Yating Wang
- Department of Medicine, Emory University, Atlanta, Georgia, United States
| | - Isaac M Chocron
- Vanderbilt Eye Institute, Vanderbilt University Medical Center, Nashville, Tennessee, United States
| | - Cassandra Warden
- Vanderbilt Eye Institute, Vanderbilt University Medical Center, Nashville, Tennessee, United States
| | - Sabrina L Mitchell
- Vanderbilt Eye Institute, Vanderbilt University Medical Center, Nashville, Tennessee, United States
| | - L Goodwin Burgess
- Vanderbilt Eye Institute, Vanderbilt University Medical Center, Nashville, Tennessee, United States
| | - Megan P Goodale
- Vanderbilt Eye Institute, Vanderbilt University Medical Center, Nashville, Tennessee, United States
| | - Melissa P Osborn
- Vanderbilt Eye Institute, Vanderbilt University Medical Center, Nashville, Tennessee, United States
| | - Allison J Ferreira
- Vanderbilt Eye Institute, Vanderbilt University Medical Center, Nashville, Tennessee, United States
| | - Janice C Law
- Vanderbilt Eye Institute, Vanderbilt University Medical Center, Nashville, Tennessee, United States
| | - Edward F Cherney
- Vanderbilt Eye Institute, Vanderbilt University Medical Center, Nashville, Tennessee, United States
| | - Dean P Jones
- Department of Medicine, Emory University, Atlanta, Georgia, United States
| | - Milam A Brantley
- Vanderbilt Eye Institute, Vanderbilt University Medical Center, Nashville, Tennessee, United States
| |
Collapse
|
36
|
Cantó A, Olivar T, Romero FJ, Miranda M. Nitrosative Stress in Retinal Pathologies: Review. Antioxidants (Basel) 2019; 8:antiox8110543. [PMID: 31717957 PMCID: PMC6912788 DOI: 10.3390/antiox8110543] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2019] [Revised: 10/30/2019] [Accepted: 11/06/2019] [Indexed: 12/20/2022] Open
Abstract
Nitric oxide (NO) is a gas molecule with diverse physiological and cellular functions. In the eye, NO is used to maintain normal visual function as it is involved in photoreceptor light transduction. In addition, NO acts as a rapid vascular endothelial relaxant, is involved in the control of retinal blood flow under basal conditions and mediates the vasodilator responses of different substances such as acetylcholine, bradykinin, histamine, substance P or insulin. However, the retina is rich in polyunsaturated lipid membranes and is sensitive to the action of reactive oxygen and nitrogen species. Products generated from NO (i.e., dinitrogen trioxide (N2O3) and peroxynitrite) have great oxidative damaging effects. Oxygen and nitrogen species can react with biomolecules (lipids, proteins and DNA), potentially leading to cell death, and this is particularly important in the retina. This review focuses on the role of NO in several ocular diseases, including diabetic retinopathy, retinitis pigmentosa, glaucoma or age-related macular degeneration (AMD).
Collapse
Affiliation(s)
- Antolin Cantó
- Departamento Ciencias Biomédicas, Facultad de Ciencias de la Salud, Universidad Cardenal Herrera-CEU, CEU Universities, 64315 Valencia, Spain; (A.C.); (T.O.)
| | - Teresa Olivar
- Departamento Ciencias Biomédicas, Facultad de Ciencias de la Salud, Universidad Cardenal Herrera-CEU, CEU Universities, 64315 Valencia, Spain; (A.C.); (T.O.)
| | - Francisco Javier Romero
- Departamento de Ciencias Biomédicas, Universidad Europea de Valencia, 46010 Valencia, Spain;
| | - María Miranda
- Departamento Ciencias Biomédicas, Facultad de Ciencias de la Salud, Universidad Cardenal Herrera-CEU, CEU Universities, 64315 Valencia, Spain; (A.C.); (T.O.)
- Correspondence: ; Tel.: +34-961369000
| |
Collapse
|
37
|
Narayanan SP, Shosha E, D Palani C. Spermine oxidase: A promising therapeutic target for neurodegeneration in diabetic retinopathy. Pharmacol Res 2019; 147:104299. [PMID: 31207342 PMCID: PMC7011157 DOI: 10.1016/j.phrs.2019.104299] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/31/2018] [Revised: 04/23/2019] [Accepted: 06/05/2019] [Indexed: 12/20/2022]
Abstract
Diabetic Retinopathy (DR), is a significant public health issue and the leading cause of blindness in working-aged adults worldwide. The vision loss associated with DR affects patients' quality of life and has negative social and psychological effects. In the past, diabetic retinopathy was considered as a vascular disease; however, it is now recognized to be a neuro-vascular disease of the retina. Current therapies for DR, such as laser photocoagulation and anti-VEGF therapy, treat advanced stages of the disease, particularly the vasculopathy and have adverse side effects. Unavailability of effective treatments to prevent the incidence or progression of DR is a major clinical problem. There is a great need for therapeutic interventions capable of preventing retinal damage in DR patients. A growing body of evidence shows that neurodegeneration is an early event in DR pathogenesis. Therefore, studies of the underlying mechanisms that lead to neurodegeneration are essential for identifying new therapeutic targets in the early stages of DR. Deregulation of the polyamine metabolism is implicated in various neurodegenerative diseases, cancer, renal failure, and diabetes. Spermine Oxidase (SMOX) is a highly inducible enzyme, and its dysregulation can alter polyamine homeostasis. The oxidative products of polyamine metabolism are capable of inducing cell damage and death. The current review provides insight into the SMOX-regulated molecular mechanisms of cellular damage and dysfunction, and its potential as a therapeutic target for diabetic retinopathy. Structural and functional changes in the diabetic retina and the mechanisms leading to neuronal damage (excitotoxicity, loss of neurotrophic factors, oxidative stress, mitochondrial dysfunction etc.) are also summarized in this review. Furthermore, existing therapies and new approaches to neuroprotection are discussed.
Collapse
Affiliation(s)
- S Priya Narayanan
- Clinical and Experimental Therapeutics, College of Pharmacy, University of Georgia, Augusta, GA, United States; Augusta University Culver Vision Discovery Institute, Augusta, GA, United States; Vascular Biology Center, Augusta University, Augusta, GA, United States; VA Medical Center, Augusta, GA, United States.
| | - Esraa Shosha
- Augusta University Culver Vision Discovery Institute, Augusta, GA, United States; Vascular Biology Center, Augusta University, Augusta, GA, United States; Clinical Pharmacy Department, Faculty of Pharmacy, Cairo University, Cairo, Egypt
| | - Chithra D Palani
- Clinical and Experimental Therapeutics, College of Pharmacy, University of Georgia, Augusta, GA, United States; Augusta University Culver Vision Discovery Institute, Augusta, GA, United States; Vascular Biology Center, Augusta University, Augusta, GA, United States
| |
Collapse
|
38
|
Age-Dependent Oxidative Stress Elevates Arginase 1 and Uncoupled Nitric Oxide Synthesis in Skeletal Muscle of Aged Mice. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2019; 2019:1704650. [PMID: 31205583 PMCID: PMC6530149 DOI: 10.1155/2019/1704650] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/07/2018] [Accepted: 03/04/2019] [Indexed: 12/21/2022]
Abstract
Aging is associated with reduced muscle mass (sarcopenia) and poor bone quality (osteoporosis), which together increase the incidence of falls and bone fractures. It is widely appreciated that aging triggers systemic oxidative stress, which can impair myoblast cell survival and differentiation. We previously reported that arginase plays an important role in oxidative stress-dependent bone loss. We hypothesized that arginase activity is dysregulated with aging in muscles and may be involved in muscle pathophysiology. To investigate this, we analyzed arginase activity and its expression in skeletal muscles of young and aged mice. We found that arginase activity and arginase 1 expression were significantly elevated in aged muscles. We also demonstrated that SOD2, GPx1, and NOX2 increased with age in skeletal muscle. Most importantly, we also demonstrated elevated levels of peroxynitrite formation and uncoupling of eNOS in aged muscles. Our in vitro studies using C2C12 myoblasts showed that the oxidative stress treatment increased arginase activity, decreased cell survival, and increased apoptotic markers. These effects were reversed by treatment with an arginase inhibitor, 2(S)-amino-6-boronohexanoic acid (ABH). Our study provides strong evidence that L-arginine metabolism is altered in aged muscle and that arginase inhibition could be used as a novel therapeutic target for age-related muscle complications.
Collapse
|
39
|
Jiang MN, Zhou YY, Hua DH, Yang JY, Hu ML, Xing YQ. Vagal Nerve Stimulation Attenuates Ischemia-Reperfusion Induced Retina Dysfunction in Acute Ocular Hypertension. Front Neurosci 2019; 13:87. [PMID: 30804746 PMCID: PMC6378858 DOI: 10.3389/fnins.2019.00087] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2018] [Accepted: 01/25/2019] [Indexed: 12/19/2022] Open
Abstract
Purpose: The present study aimed to investigate whether cervical vagal nerve stimulation (VNS) could prevent retinal ganglion cell (RGC) loss and retinal dysfunction after ischemia/reperfusion (I/R) injury. Methods: First, rats were randomly divided into sham group (n = 4) and VNS group (n = 12). Activation of the nodose ganglia (NOG), nucleus of the solitary tract (NTS), superior salivatory nucleus (SSN), and pterygopalatine ganglion (PPG) neural circuit were evaluated by c-fos expression at 0 h after sham VNS and at 0 h (n = 4), 6 h (n = 4), 72 h (n = 4) after VNS. Secondly, rats were randomly assigned to I/R group (pressure-induced retinal ischemia for 1 h and reperfusion for 1 h in the right eye, n = 16) and I/R+VNS group (right cervical VNS for 2 h during the I/R period, n = 16). The left eye of each rat served as a control. Electroretinogram (ERG), RGC numbers, tumor necrosis factor-α (TNF-α) and vasoactive intestinal polypeptide (VIP) levels in retina were determined. Additionally, the level of VIP in PPG was evaluated. Results: In the first part of the study, compared with the sham group, the VNS group exhibited significantly increased expression of c-fos in NOG, NTS, SSN, and PPG tissues at 0, 6, and 72 h. In the second part of the study, compared with left eyes, retinal function in right eyes (as assessed by the a-wave, b-wave and the oscillatory potential amplitudes of ERG and RGC data) was significantly decreased by I/R. The decreased retinal function was attenuated by VNS. In addition, I/R induced an increase in inflammation, which was reflected by elevated TNF-α expression in the retina. VNS significantly attenuated the increase in I/R-induced inflammation. Moreover, VIP expression in the retina and PPG, which may contribute to the inhibition of the inflammatory response, was significantly increased after VNS. Conclusion: VNS could protect against retinal I/R injury by downregulating TNF-α. Upregulation of VIP expression due to activation of the NOG-NTS-SSN-PPG neural circuit may underlie to the protective effects of VNS.
Collapse
Affiliation(s)
- Meng-Nan Jiang
- Eye Center, Renmin Hospital of Wuhan University, Wuhan, China
| | - Yu-Yang Zhou
- Eye Center, Renmin Hospital of Wuhan University, Wuhan, China
| | - Di-Hao Hua
- Eye Center, Renmin Hospital of Wuhan University, Wuhan, China
| | - Jia-Yi Yang
- Eye Center, Renmin Hospital of Wuhan University, Wuhan, China
| | - Man-Li Hu
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Yi-Qiao Xing
- Eye Center, Renmin Hospital of Wuhan University, Wuhan, China
| |
Collapse
|
40
|
Laíns I, Gantner M, Murinello S, Lasky-Su JA, Miller JW, Friedlander M, Husain D. Metabolomics in the study of retinal health and disease. Prog Retin Eye Res 2018; 69:57-79. [PMID: 30423446 DOI: 10.1016/j.preteyeres.2018.11.002] [Citation(s) in RCA: 100] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2018] [Revised: 10/06/2018] [Accepted: 11/07/2018] [Indexed: 02/06/2023]
Abstract
Metabolomics is the qualitative and quantitative assessment of the metabolites (small molecules < 1.5 kDa) in body fluids. The metabolites are the downstream of the genetic transcription and translation processes and also downstream of the interactions with environmental exposures; thus, they are thought to closely relate to the phenotype, especially for multifactorial diseases. In the last decade, metabolomics has been increasingly used to identify biomarkers in disease, and it is currently recognized as a very powerful tool with great potential for clinical translation. The metabolome and the associated pathways also help improve our understanding of the pathophysiology and mechanisms of disease. While there has been increasing interest and research in metabolomics of the eye, the application of metabolomics to retinal diseases has been limited, even though these are leading causes of blindness. In this manuscript, we perform a comprehensive summary of the tools and knowledge required to perform a metabolomics study, and we highlight essential statistical methods for rigorous study design and data analysis. We review available protocols, summarize the best approaches, and address the current unmet need for information on collection and processing of tissues and biofluids that can be used for metabolomics of retinal diseases. Additionally, we critically analyze recent work in this field, both in animal models and in human clinical disease, including diabetic retinopathy and age-related macular degeneration. Finally, we identify opportunities for future research applying metabolomics to improve our current assessment and understanding of mechanisms of vitreoretinal diseases, and to hence improve patient assessment and care.
Collapse
Affiliation(s)
- Inês Laíns
- Retina Service, Massachusetts Eye and Ear, Harvard Medical School, 243 Charles Street, Boston, MA, 02114, United States; Faculty of Medicine, University of Coimbra, 3000 Coimbra, Portugal.
| | - Mari Gantner
- Lowy Medical Research Institute, La Jolla, CA, 92037, United States; Scripps Research Institute, La Jolla, CA, 92037, United States.
| | - Salome Murinello
- Lowy Medical Research Institute, La Jolla, CA, 92037, United States; Scripps Research Institute, La Jolla, CA, 92037, United States.
| | - Jessica A Lasky-Su
- Systems Genetics and Genomics Unit, Channing Division of Network Medicine Brigham and Women's Hospital and Harvard Medical School, 75 Francis Street, Boston, MA, 02115, United States.
| | - Joan W Miller
- Retina Service, Massachusetts Eye and Ear, Harvard Medical School, 243 Charles Street, Boston, MA, 02114, United States.
| | - Martin Friedlander
- Lowy Medical Research Institute, La Jolla, CA, 92037, United States; Scripps Research Institute, La Jolla, CA, 92037, United States.
| | - Deeba Husain
- Retina Service, Massachusetts Eye and Ear, Harvard Medical School, 243 Charles Street, Boston, MA, 02114, United States.
| |
Collapse
|
41
|
Sahajpal NS, Goel RK, Chaubey A, Aurora R, Jain SK. Pathological Perturbations in Diabetic Retinopathy: Hyperglycemia, AGEs, Oxidative Stress and Inflammatory Pathways. Curr Protein Pept Sci 2018; 20:92-110. [DOI: 10.2174/1389203719666180928123449] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2018] [Revised: 08/10/2018] [Accepted: 08/29/2017] [Indexed: 01/02/2023]
Abstract
Diabetic retinopathy (DR) remains the leading cause of blindness in working-aged adults
around the world. The proliferative diabetic retinopathy (PDR) and diabetic macular edema (DME) are
the severe vision threatening stages of the disorder. Although, a huge body of research exists in elaborating
the pathological mechanisms that lead to the development of DR, the certainty and the correlation
amongst these pathways remain ambiguous. The complexity of DR lies in the multifactorial pathological
perturbations that are instrumental in both the disease development and its progression. Therefore, a holistic
perspective with an understanding of these pathways and their correlation may explain the pathogenesis
of DR as a unifying mechanism. Hyperglycemia, oxidative stress and inflammatory pathways
are the crucial components that are implicated in the pathogenesis of DR. Of these, hyperglycemia appears
to be the initiating central component around which other pathological processes operate. Thus,
this review discusses the role of hyperglycemia, oxidative stress and inflammation in the pathogenesis of
DR, and highlights the cross-talk amongst these pathways in an attempt to understand the complex interplay
of these mechanisms. Further, an effort has been made to identify the knowledge gap and the key
players in each pathway that may serve as potential therapeutic drug targets.
Collapse
Affiliation(s)
- Nikhil Shri Sahajpal
- Department of Pharmaceutical Sciences, Guru Nanak Dev University, Amritsar, Punjab, India
| | - Rajesh Kumar Goel
- Department of Pharmaceutical Sciences and Drug Research, Punjabi University, Patiala, India
| | - Alka Chaubey
- Cytogenetics Laboratory, Greenwood Genetic Center, Greenwood, South Carolina, SC, United States
| | - Rohan Aurora
- The International School Bangalore, Karnataka, India
| | - Subheet Kumar Jain
- Department of Pharmaceutical Sciences, Guru Nanak Dev University, Amritsar, Punjab, India
| |
Collapse
|
42
|
Hobson-Gutierrez SA, Carmona-Fontaine C. The metabolic axis of macrophage and immune cell polarization. Dis Model Mech 2018; 11:11/8/dmm034462. [PMID: 29991530 PMCID: PMC6124558 DOI: 10.1242/dmm.034462] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
The extracellular space of solid tumors ranges from being well-nurtured to being completely ischemic and can serve as a source of intratumoral heterogeneity, determining the behavior and molecular profiles of malignant and stromal cells. Here, we discuss how the metabolic tumor microenvironment modulates the phenotypes of the immune cells that infiltrate tumors, with an emphasis on tumor-associated macrophages. These cells constitute a diverse population that has pro-tumoral and anti-inflammatory properties, and are likened to anti-inflammatory ‘M2’ macrophages. Recent findings show how different metabolic microenvironments specify an array of phenotypic changes in macrophages. In tumors, extracellular metabolite levels vary predictably according to proximity to the vasculature, and phenotypic changes in tumor-associated macrophages and in other immune cells are also predictable. We speculate that this ‘metabolic axis’ of macrophage polarization modulates – and is modulated by – the response to inflammatory cues, creating a wide variety of possible phenotypic states. Understanding how extracellular metabolites influence cell phenotypes allows us to predict how tumor-associated macrophages and other tumor cells might change, with the aim of harnessing this predictability for therapy. Overall, we describe an emerging picture in which chemokines, growth factors and the metabolic tumor microenvironment act together to determine the phenotypes of tumor-infiltrating immune cells. Summary: Extracellular metabolites can mediate cell communication and change transcriptional and functional aspects of tumor and normal cells. This Review focuses on how these metabolic cues affect the immune system with a special focus on tumor-associated macrophages.
Collapse
Affiliation(s)
- Spencer A Hobson-Gutierrez
- Center for Genomics and Systems Biology, Department of Biology, New York University, New York City, NY 10003, USA
| | - Carlos Carmona-Fontaine
- Center for Genomics and Systems Biology, Department of Biology, New York University, New York City, NY 10003, USA
| |
Collapse
|
43
|
Shosha E, Xu Z, Narayanan SP, Lemtalsi T, Fouda AY, Rojas M, Xing J, Fulton D, Caldwell RW, Caldwell RB. Mechanisms of Diabetes-Induced Endothelial Cell Senescence: Role of Arginase 1. Int J Mol Sci 2018; 19:ijms19041215. [PMID: 29673160 PMCID: PMC5979610 DOI: 10.3390/ijms19041215] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2018] [Revised: 04/13/2018] [Accepted: 04/14/2018] [Indexed: 12/17/2022] Open
Abstract
We have recently found that diabetes-induced premature senescence of retinal endothelial cells is accompanied by NOX2-NADPH oxidase-induced increases in the ureohydrolase enzyme arginase 1 (A1). Here, we used genetic strategies to determine the specific involvement of A1 in diabetes-induced endothelial cell senescence. We used A1 knockout mice and wild type mice that were rendered diabetic with streptozotocin and retinal endothelial cells (ECs) exposed to high glucose or transduced with adenovirus to overexpress A1 for these experiments. ABH [2(S)-Amino-6-boronohexanoic acid] was used to inhibit arginase activity. We used Western blotting, immunolabeling, quantitative PCR, and senescence associated β-galactosidase (SA β-Gal) activity to evaluate senescence. Analyses of retinal tissue extracts from diabetic mice showed significant increases in mRNA expression of the senescence-related proteins p16INK4a, p21, and p53 when compared with non-diabetic mice. SA β-Gal activity and p16INK4a immunoreactivity were also increased in retinal vessels from diabetic mice. A1 gene deletion or pharmacological inhibition protected against the induction of premature senescence. A1 overexpression or high glucose treatment increased SA β-Gal activity in cultured ECs. These results demonstrate that A1 is critically involved in diabetes-induced senescence of retinal ECs. Inhibition of arginase activity may therefore be an effective therapeutic strategy to alleviate diabetic retinopathy by preventing premature senescence.
Collapse
Affiliation(s)
- Esraa Shosha
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA.
- Charlie Norwood VA Medical Center, Augusta, GA 30904, USA.
| | - Zhimin Xu
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA.
- Charlie Norwood VA Medical Center, Augusta, GA 30904, USA.
| | - S Priya Narayanan
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA.
- Charlie Norwood VA Medical Center, Augusta, GA 30904, USA.
- Department of Occupational Therapy, College of Allied Health Sciences, Augusta University, Augusta, GA 30912, USA.
| | - Tahira Lemtalsi
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA.
- Charlie Norwood VA Medical Center, Augusta, GA 30904, USA.
| | - Abdelrahman Y Fouda
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA.
- Charlie Norwood VA Medical Center, Augusta, GA 30904, USA.
| | - Modesto Rojas
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA.
- Charlie Norwood VA Medical Center, Augusta, GA 30904, USA.
| | - Ji Xing
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA.
| | - David Fulton
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA.
| | - R William Caldwell
- Department of Pharmacology and Toxicology, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA.
| | - Ruth B Caldwell
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA.
- Charlie Norwood VA Medical Center, Augusta, GA 30904, USA.
| |
Collapse
|
44
|
Chen M, Zhao J, Ali IHA, Marry S, Augustine J, Bhuckory M, Lynch A, Kissenpfennig A, Xu H. Cytokine Signaling Protein 3 Deficiency in Myeloid Cells Promotes Retinal Degeneration and Angiogenesis through Arginase-1 Up-Regulation in Experimental Autoimmune Uveoretinitis. THE AMERICAN JOURNAL OF PATHOLOGY 2018; 188:1007-1020. [PMID: 29452101 DOI: 10.1016/j.ajpath.2017.12.021] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/02/2017] [Revised: 11/14/2017] [Accepted: 12/07/2017] [Indexed: 01/01/2023]
Abstract
The suppressor of cytokine signaling protein 3 (SOCS3) critically controls immune cell activation, although its role in macrophage polarization and function remains controversial. Using experimental autoimmune uveoretinitis (EAU) as a model, we show that inflammation-mediated retinal degeneration is exaggerated and retinal angiogenesis is accelerated in mice with SOCS3 deficiency in myeloid cells (LysMCre/+SOCS3fl/fl). At the acute stage of EAU, the population of infiltrating neutrophils was increased and the population of macrophages decreased in LysMCre/+SOCS3fl/fl mice compared with that in wild-type (WT) mice. Real-time RT-PCR showed that the expression of tumor necrosis factor-α, IL-1β, interferon-γ, granulocyte-macrophage colony-stimulating factor, and arginase-1 was significantly higher in the LysMCre/+SOCS3fl/fl EAU retina in contrast to the WT EAU retina. The percentage of arginase-1+ infiltrating cells was significantly higher in the LysMCre/+SOCS3fl/fl EAU retina than that in the WT EAU retina. In addition, bone marrow-derived macrophages and neutrophils from the LysMCre/+SOCS3fl/fl mice express significantly higher levels of chemokine (C-C motif) ligand 2 and arginase-1 compared with those from WT mice. Inhibition of arginase using an l-arginine analog amino-2-borono-6-hexanoic suppressed inflammation-induced retinal angiogenesis without affecting the severity of inflammation. Our results suggest that SOCS3 critically controls the phenotype and function of macrophages and neutrophils under inflammatory conditions and loss of SOCS3 promotes the angiogenic phenotype of the cells through up-regulation of arginase-1.
Collapse
Affiliation(s)
- Mei Chen
- Centre for Experimental Medicine, School of Medicine, Dentistry & Biomedical Science, Queen's University Belfast, Belfast, United Kingdom
| | - Jiawu Zhao
- Centre for Experimental Medicine, School of Medicine, Dentistry & Biomedical Science, Queen's University Belfast, Belfast, United Kingdom
| | - Imran H A Ali
- Centre for Experimental Medicine, School of Medicine, Dentistry & Biomedical Science, Queen's University Belfast, Belfast, United Kingdom
| | - Stephen Marry
- Centre for Experimental Medicine, School of Medicine, Dentistry & Biomedical Science, Queen's University Belfast, Belfast, United Kingdom
| | - Josy Augustine
- Centre for Experimental Medicine, School of Medicine, Dentistry & Biomedical Science, Queen's University Belfast, Belfast, United Kingdom
| | - Mohajeet Bhuckory
- Centre for Experimental Medicine, School of Medicine, Dentistry & Biomedical Science, Queen's University Belfast, Belfast, United Kingdom
| | - Aisling Lynch
- Centre for Experimental Medicine, School of Medicine, Dentistry & Biomedical Science, Queen's University Belfast, Belfast, United Kingdom
| | - Adrien Kissenpfennig
- Centre for Experimental Medicine, School of Medicine, Dentistry & Biomedical Science, Queen's University Belfast, Belfast, United Kingdom
| | - Heping Xu
- Centre for Experimental Medicine, School of Medicine, Dentistry & Biomedical Science, Queen's University Belfast, Belfast, United Kingdom.
| |
Collapse
|
45
|
Polat N, Ozer MA, Parlakpinar H, Vardi N, Aksungur Z, Ozhan O, Yildiz A, Turkoz Y. Effects of molsidomine on retinal ischemia/reperfusion injury in rabbits. Biotech Histochem 2018; 93:188-197. [PMID: 29323543 DOI: 10.1080/10520295.2017.1406616] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022] Open
Abstract
We investigated the effect of molsidomine (MOL) on ischemia/reperfusion (I/R) injury. Rabbits were assigned to four groups: group 1, sham; group 2, I/R; group 3, MOL treatment for 4 days after I/R; group 4, MOL treatment for 1 day before I/R and 3 days after I/R. Retinal I/R was produced by elevating the intraocular pressure to 150 mm Hg for 60 min. Seven days after I/R, the eyes were enucleated. Retinal changes were examined using histochemistry. The levels of malondialdehyde (MDA), glutathione (GSH), superoxide dismutase (SOD), glutathione peroxidase (GSH-Px) and catalase (CAT) also were measured. We found a significant increase in the thickness of the outer nuclear layer of group 3 compared to the other groups. In groups 3 and 4, caspase-3 stained cells in the ganglion cell layer were decreased compared to group 2. We found a significant increase in caspase-3 stained cells in the inner nuclear layer (INL) of group 2 compared to the other groups. We found a significant increase in caspase-3 stained cells in group 3 compared to group 4 in the INL. The MDA level in group 2 was significantly higher than group 1 and MOL significantly decreased MDA levels in groups 3 and 4. We found that MOL protected the retina from I/R injury by enhancing antioxidative effects and inhibiting apoptosis of retinal cells.
Collapse
Affiliation(s)
- N Polat
- a Department of Ophthalmology , Inonu University School of Medicine , Malatya
| | - M A Ozer
- b Department of Ophthalmology , Giresun University School of Medicine , Giresun
| | | | | | - Z Aksungur
- e Biochemistry , Inonu University School of Medicine , Malatya , Turkey
| | - O Ozhan
- c Departments of Pharmacology
| | | | - Y Turkoz
- e Biochemistry , Inonu University School of Medicine , Malatya , Turkey
| |
Collapse
|
46
|
Dehdashtian E, Mehrzadi S, Yousefi B, Hosseinzadeh A, Reiter RJ, Safa M, Ghaznavi H, Naseripour M. Diabetic retinopathy pathogenesis and the ameliorating effects of melatonin; involvement of autophagy, inflammation and oxidative stress. Life Sci 2018; 193:20-33. [DOI: 10.1016/j.lfs.2017.12.001] [Citation(s) in RCA: 163] [Impact Index Per Article: 23.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2017] [Revised: 11/19/2017] [Accepted: 12/01/2017] [Indexed: 12/12/2022]
|
47
|
Ischemic Retinopathies: Oxidative Stress and Inflammation. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2017; 2017:3940241. [PMID: 29410732 PMCID: PMC5749295 DOI: 10.1155/2017/3940241] [Citation(s) in RCA: 99] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/17/2017] [Accepted: 11/20/2017] [Indexed: 12/15/2022]
Abstract
Ischemic retinopathies (IRs), such as retinopathy of prematurity (ROP), diabetic retinopathy (DR), and (in many cases) age-related macular degeneration (AMD), are ocular disorders characterized by an initial phase of microvascular changes that results in ischemia, followed by a second phase of abnormal neovascularization that may culminate into retinal detachment and blindness. IRs are complex retinal conditions in which several factors play a key role during the development of the different pathological stages of the disease. Increasing evidence reveals that oxidative stress and inflammatory processes are important contributors to the pathogenesis of IRs. Despite the beneficial effects of the photocoagulation and anti-VEGF therapy during neovascularization phase, the need to identify novel targets to prevent initial phases of these ocular pathologies is still needed. In this review, we provide an update on the involvement of oxidative stress and inflammation in the progression of IRs and address some therapeutic interventions by using antioxidants and anti-inflammatory agents.
Collapse
|
48
|
Hua Y, Yao W, Ji P, Wei Y. Integrated metabonomic-proteomic studies on blood enrichment effects of Angelica sinensis on a blood deficiency mice model. PHARMACEUTICAL BIOLOGY 2017; 55:853-863. [PMID: 28140733 PMCID: PMC6130503 DOI: 10.1080/13880209.2017.1281969] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/14/2016] [Revised: 10/19/2016] [Accepted: 01/10/2017] [Indexed: 05/18/2023]
Abstract
CONTEXT Angelica sinensis (Oliv.) Diels (Umbelliferae) (AS) is a well-known Traditional Chinese Medicine (TCM) that enriches and regulates the blood. OBJECTIVE An integrated metabonomic and proteomic method was developed and applied to study the blood enrichment effects and mechanisms of AS on blood deficiency (BD) mouse model. MATERIALS AND METHODS Forty mice were randomly divided into the control, BD, High-dose of AS (ASH), Middle-dose of AS (ASM), and Low-dose of AS (ASL) groups. BD model mice were established by injecting N-acetylphenylhydrazine (APH) and cyclophosphamide (CTX) (ip). The aqueous extract of AS was administered at three dose of 20, 10, or 5 g/kg b. wt. orally for 7 consecutive days before/after APH and CTX administration. Gas chromatography-mass spectrometry (GC-MS) combined with pattern recognition method and 2D gel electrophoresis (2-DE) proteomics were performed in this study to discover the underlying hematopoietic regulation mechanisms of AS on BD mouse model. RESULTS Unlike in the control group, the HSP90 and arginase levels increased significantly (p < 0.05) in the BD group, but the levels of carbonic anhydrase, GAPDH, catalase, fibrinogen, GSTP, carboxylesterase and hem binding protein in the BD group decreased significantly (p < 0.05). Unlike the levels in the BD group, the levels of these biomarkers were regulated to a normal state near the control group in the ASM group. Unlike in the control group, l-alanine, arachidonic acid, l-valine, octadecanoic acid, glycine, hexadecanoic acid, l-threonine, butanoic acid, malic acid, l-proline and propanoic acid levels increased significantly (p < 0.05) in the BD group, the levels of d-fructose in the BD group decreased significantly (p < 0.05). The relative concentrations of 12 endogenous metabolites were also significantly affected by the ASL, ASM, and ASH treatments. Notably, most of the altered BD-related metabolites were restored to normal state after ASM administration. CONCLUSION AS can promote hematopoietic activities, inhibit production of reactive oxygen species, regulate energy metabolism, increase antiapoptosis, and potentially contribute to the blood enrichment effects of AS against APH- and CTX-induced BD mice.
Collapse
Affiliation(s)
- Yongli Hua
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, Gansu Province, People’s Republic of China
| | - Wangling Yao
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, Gansu Province, People’s Republic of China
| | - Peng Ji
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, Gansu Province, People’s Republic of China
| | - Yanming Wei
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, Gansu Province, People’s Republic of China
- CONTACT Yanming WeiCollege of Veterinary Medicine, Gansu Agricultural University, Lanzhou, Gansu Province730070, People’s Republic of China
| |
Collapse
|
49
|
Liew G, Lei Z, Tan G, Joachim N, Ho IV, Wong TY, Mitchell P, Gopinath B, Crossett B. Metabolomics of Diabetic Retinopathy. Curr Diab Rep 2017; 17:102. [PMID: 28940103 DOI: 10.1007/s11892-017-0939-3] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
PURPOSE OF REVIEW Metabolomics is the study of dysregulated metabolites in biological materials. We reviewed the use of the technique to elucidate the genetic and environmental factors that contribute to the development of diabetic retinopathy. RECENT FINDINGS With regard to metabolomic studies of diabetic retinopathy, the field remains in its infancy with few studies published to date and little replication of results. Vitreous and serum samples are the main tissues examined, and dysregulation in pathways such as the pentose phosphate pathway, arginine to proline pathway, polyol pathway, and ascorbic acidic pathways have been reported. Few studies have examined the metabolomic underpinnings of diabetic retinopathy. Further research is required to replicate findings to date and determine longitudinal associations with disease.
Collapse
Affiliation(s)
- Gerald Liew
- Centre for Vision Research, Westmead Millennium Institute of Medical Research, University of Sydney, Westmead, Sydney, 2145, Australia.
- South West Retina, Retina Associates, Liverpool, Sydney, Australia.
| | - Zhou Lei
- Duke-NUS School of Medicine, National University of Singapore, Singapore, Singapore
| | - Gavin Tan
- Duke-NUS School of Medicine, National University of Singapore, Singapore, Singapore
| | - Nichole Joachim
- Centre for Vision Research, Westmead Millennium Institute of Medical Research, University of Sydney, Westmead, Sydney, 2145, Australia
| | - I-Van Ho
- South West Retina, Retina Associates, Liverpool, Sydney, Australia
- Save Sight Institute, University of Sydney, Sydney, Australia
- Australian School of Advanced Medicine, Macquarie University, Sydney, Australia
| | - Tien Y Wong
- Duke-NUS School of Medicine, National University of Singapore, Singapore, Singapore
| | - Paul Mitchell
- Centre for Vision Research, Westmead Millennium Institute of Medical Research, University of Sydney, Westmead, Sydney, 2145, Australia
| | - Bamini Gopinath
- Centre for Vision Research, Westmead Millennium Institute of Medical Research, University of Sydney, Westmead, Sydney, 2145, Australia
| | - Ben Crossett
- Mass Spectrometry Core Facility, Building D17, University of Sydney, Sydney, Australia
| |
Collapse
|
50
|
Crooke A, Huete-Toral F, Colligris B, Pintor J. The role and therapeutic potential of melatonin in age-related ocular diseases. J Pineal Res 2017; 63. [PMID: 28658514 DOI: 10.1111/jpi.12430] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/14/2017] [Accepted: 06/23/2017] [Indexed: 12/20/2022]
Abstract
The eye is continuously exposed to solar UV radiation and pollutants, making it prone to oxidative attacks. In fact, oxidative damage is a major cause of age-related ocular diseases including cataract, glaucoma, age-related macular degeneration, and diabetic retinopathy. As the nature of lens cells, trabecular meshwork cells, retinal ganglion cells, retinal pigment epithelial cells, and photoreceptors is postmitotic, autophagy plays a critical role in their cellular homeostasis. In age-related ocular diseases, this process is impaired, and thus, oxidative damage becomes irreversible. Other conditions such as low-grade chronic inflammation and angiogenesis also contribute to the development of retinal diseases (glaucoma, age-related macular degeneration and diabetic retinopathy). As melatonin is known to have remarkable qualities such as antioxidant/antinitridergic, mitochondrial protector, autophagy modulator, anti-inflammatory, and anti-angiogenic, it can represent a powerful tool to counteract all these diseases. The present review analyzes the role and therapeutic potential of melatonin in age-related ocular diseases, focusing on nitro-oxidative stress, autophagy, inflammation, and angiogenesis mechanisms.
Collapse
Affiliation(s)
- Almudena Crooke
- Department of Biochemistry and Molecular Biology IV, Group Ocupharm, Faculty of Optics and Optometry, Universidad Complutense de Madrid, Madrid, Spain
| | - Fernando Huete-Toral
- Department of Biochemistry and Molecular Biology IV, Group Ocupharm, Faculty of Optics and Optometry, Universidad Complutense de Madrid, Madrid, Spain
| | - Basilio Colligris
- Department of Biochemistry and Molecular Biology IV, Group Ocupharm, Faculty of Optics and Optometry, Universidad Complutense de Madrid, Madrid, Spain
| | - Jesús Pintor
- Department of Biochemistry and Molecular Biology IV, Group Ocupharm, Faculty of Optics and Optometry, Universidad Complutense de Madrid, Madrid, Spain
| |
Collapse
|