51
|
Clarkson-Townsend DA, Bales KL, Marsit CJ, Pardue MT. Light Environment Influences Developmental Programming of the Metabolic and Visual Systems in Mice. Invest Ophthalmol Vis Sci 2021; 62:22. [PMID: 33861321 PMCID: PMC8083116 DOI: 10.1167/iovs.62.4.22] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Accepted: 03/27/2021] [Indexed: 02/06/2023] Open
Abstract
Purpose Light is a salient cue that can influence neurodevelopment and the immune system. Light exposure out of sync with the endogenous clock causes circadian disruption and chronic disease. Environmental light exposure may contribute to developmental programming of metabolic and neurological systems but has been largely overlooked in Developmental Origins of Health and Disease (DOHaD) research. Here, we investigated whether developmental light exposure altered programming of visual and metabolic systems. Methods Pregnant mice and pups were exposed to control light (12:12 light:dark) or weekly light cycle inversions (circadian disruption [CD]) until weaning, after which male and female offspring were housed in control light and longitudinally measured to evaluate differences in growth (weight), glucose tolerance, visual function (optomotor response), and retinal function (electroretinogram), with and without high fat diet (HFD) challenge. Retinal microglia and macrophages were quantified by positive Iba1 and CD11b immunofluorescence. Results CD exposure caused impaired visual function and increased retinal immune cell expression in adult offspring. When challenged with HFD, CD offspring also exhibited altered retinal function and sex-specific impairments in glucose tolerance. Conclusions Overall, these findings suggest that the light environment contributes to developmental programming of the metabolic and visual systems, potentially promoting a pro-inflammatory milieu in the retina and increasing the risk of visual disease later in life.
Collapse
Affiliation(s)
- Danielle A. Clarkson-Townsend
- Gangarosa Department of Environmental Health, Rollins School of Public Health, Emory University, Atlanta, Georgia, United States
- Center for Visual and Neurocognitive Rehabilitation, Atlanta VA Healthcare System, Decatur, Georgia, United States
| | - Katie L. Bales
- Center for Visual and Neurocognitive Rehabilitation, Atlanta VA Healthcare System, Decatur, Georgia, United States
- Department of Ophthalmology, Emory University, Atlanta, Georgia, United States
| | - Carmen J. Marsit
- Gangarosa Department of Environmental Health, Rollins School of Public Health, Emory University, Atlanta, Georgia, United States
| | - Machelle T. Pardue
- Center for Visual and Neurocognitive Rehabilitation, Atlanta VA Healthcare System, Decatur, Georgia, United States
- Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, Georgia, United States
| |
Collapse
|
52
|
Chen YW, Huang YP, Wu PC, Chiang WY, Wang PH, Chen BY. The Functional Vision Protection Effect of Danshensu via Dopamine D1 Receptors: In Vivo Study. Nutrients 2021; 13:nu13030978. [PMID: 33803057 PMCID: PMC8002943 DOI: 10.3390/nu13030978] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Revised: 03/09/2021] [Accepted: 03/15/2021] [Indexed: 12/22/2022] Open
Abstract
Danshensu, a traditional herb-based active component (Salvia miltiorrhiza Bunge), has garnered attention, due to its safety, nutritional value, and antioxidant effects, along with cardiovascular-protective and neuroprotective abilities; however, its effect on the retinal tissues and functional vision has not been fully studied. The objective of this study was to analyze the protective effect of danshensu on retinal tissues and functional vision in vivo in a mouse model of light-induced retinal degeneration. High energy light-evoked visual damage was confirmed by the loss in structural tissue integrity in the retina accompanied by a decline in visual acuity and visual contrast sensitivity function (VCSF), whereas the retina tissue exhibited severe Müller cell gliosis. Although danshensu treatment did not particularly reduce light-evoked damage to the photoreceptors, it significantly prevented Müller cell gliosis. Danshensu exerted protective effects against light-evoked deterioration on low spatial frequency-based VCSF as determined by the behavioral optomotor reflex method. Additionally, the protective effect of danshensu on VCSF can be reversed and blocked by the injection of a dopamine D1 receptor antagonist (SCH 23390). This study demonstrated that the major functional vision promotional effect of danshensu in vivo was through the dopamine D1 receptors enhancement pathway, rather than the structural protection of the retinas.
Collapse
Affiliation(s)
- Yun-Wen Chen
- Department of Ophthalmology, Kaohsiung Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Kaohsiung 88301, Taiwan; (Y.-W.C.); (P.-C.W.); (W.-Y.C.)
| | - Yun-Ping Huang
- Department of Optometry, Chung Shan Medical University, Taichung 40201, Taiwan; (Y.-P.H.); (P.-H.W.)
| | - Pei-Chang Wu
- Department of Ophthalmology, Kaohsiung Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Kaohsiung 88301, Taiwan; (Y.-W.C.); (P.-C.W.); (W.-Y.C.)
| | - Wei-Yu Chiang
- Department of Ophthalmology, Kaohsiung Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Kaohsiung 88301, Taiwan; (Y.-W.C.); (P.-C.W.); (W.-Y.C.)
| | - Ping-Hsun Wang
- Department of Optometry, Chung Shan Medical University, Taichung 40201, Taiwan; (Y.-P.H.); (P.-H.W.)
| | - Bo-Yie Chen
- Department of Optometry, Chung Shan Medical University, Taichung 40201, Taiwan; (Y.-P.H.); (P.-H.W.)
- Department of Ophthalmology, Chung Shan Medical University Hospital, Taichung 40201, Taiwan
- Correspondence: ; Tel.: +886-4-2473-0022 (ext. 12319)
| |
Collapse
|
53
|
Madrakhimov SB, Yang JY, Kim JH, Han JW, Park TK. mTOR-dependent dysregulation of autophagy contributes to the retinal ganglion cell loss in streptozotocin-induced diabetic retinopathy. Cell Commun Signal 2021; 19:29. [PMID: 33637094 PMCID: PMC7913405 DOI: 10.1186/s12964-020-00698-4] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Accepted: 12/15/2020] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Neurodegeneration, an early event in the pathogenesis of diabetic retinopathy (DR), precedes clinically detectable microvascular damage. Autophagy dysregulation is considered a potential cause of neuronal cell loss, however underlying mechanisms remain unclear. The mechanistic target of rapamycin (mTOR) integrates diverse environmental signals to coordinate biological processes, including autophagy. Here, we investigated the role of mTOR signaling in neuronal cell death in DR. METHODS Diabetes was induced by a single intraperitoneal injection of streptozotocin and tissue samples were harvested at 1, 2, 3, 4, and 6 months of diabetes. Early-stage of DR was investigated in 1-month-diabetic mice treated with phlorizin (two daily subcutaneous injections at a dose of 200 mg/kg of body weight during the last 7 full days of the experiment and the morning of the 8th day, 3 h before sacrifice) or rapamycin (daily intraperitoneal injections, at a dose of 3 mg/kg for the same period as for phlorizin treatment). The effect of autophagy modulation on retinal ganglion cells was investigated in 3-months-diabetic mice treated with phlorizin (two daily subcutaneous injections during the last 10 full days of the experiment and the morning of the 11th day, 3 h before sacrifice) or MHY1485 (daily i.p. injections, at a dose of 10 mg/kg for the same period as for phlorizin treatment). Tissue samples obtained from treated/untreated diabetic mice and age-matched controls were used for Western blot and histologic analysis. RESULTS mTOR-related proteins and glucose transporter 1 (GLUT1) was upregulated at 1 month and downregulated in the following period up to 6 months. Diabetes-induced neurodegeneration was characterized by an increase of apoptotic marker-cleaved caspase 3, a decrease of the total number of cells, and NeuN immunoreactivity in the ganglion cell layer, as well as an increase of autophagic protein. Insulin-independent glycemic control restored the mTOR pathway activity and GLUT1 expression, along with a decrease of autophagic and apoptotic proteins in 3-months-diabetic mice neuroretina. However, blockade of autophagy using MHY1485 resulted in a more protective effect on ganglion cells compared with phlorizin treatment. CONCLUSION Collectively, our study describes the mechanisms of neurodegeneration through the hyperglycemia/ mTOR/ autophagy/ apoptosis pathway. Video Abstract.
Collapse
Affiliation(s)
- Sanjar Batirovich Madrakhimov
- Department of Interdisciplinary Program in Biomedical Science, Soonchunhyang Graduate School, Bucheon Hospital, Bucheon, South Korea
- Laboratory for Translational Research On Retinal and Macular Degeneration, Soonchunhyang University Hospital Bucheon, Bucheon, South Korea
| | - Jin Young Yang
- Department of Interdisciplinary Program in Biomedical Science, Soonchunhyang Graduate School, Bucheon Hospital, Bucheon, South Korea
- Laboratory for Translational Research On Retinal and Macular Degeneration, Soonchunhyang University Hospital Bucheon, Bucheon, South Korea
| | - Jin Ha Kim
- Department of Ophthalmology, Soonchunhyang University Hospital Bucheon, Bucheon, South Korea
| | - Jung Woo Han
- Department of Ophthalmology, Soonchunhyang University Hospital Bucheon, Bucheon, South Korea
| | - Tae Kwann Park
- Department of Interdisciplinary Program in Biomedical Science, Soonchunhyang Graduate School, Bucheon Hospital, Bucheon, South Korea
- Laboratory for Translational Research On Retinal and Macular Degeneration, Soonchunhyang University Hospital Bucheon, Bucheon, South Korea
- Department of Ophthalmology, Soonchunhyang University Hospital Bucheon, Bucheon, South Korea
- Department of Ophthalmology, College of Medicine, Soonchunhyang University, Choongchungnam-do, Cheonan, South Korea
- Department of Ophthalmology, College of Medicine, Soonchunhyang University Bucheon Hospital, Bucheon, South Korea
- Ex Lumina Therapeutics and Technologies. Co., Ltd., Bucheon, South Korea
| |
Collapse
|
54
|
Fei X, He X, Tai Z, Wang H, Qu S, Chen L, Hu Q, Fang J, Jiang Y. Electroacupuncture alleviates diabetic neuropathic pain in rats by suppressing P2X3 receptor expression in dorsal root ganglia. Purinergic Signal 2020; 16:491-502. [PMID: 33011961 PMCID: PMC7855163 DOI: 10.1007/s11302-020-09728-9] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Accepted: 09/02/2020] [Indexed: 02/07/2023] Open
Abstract
Diabetic neuropathic pain (DNP) is a troublesome diabetes complication all over the world. P2X3 receptor (P2X3R), a purinergic receptor from dorsal root ganglion (DRG), has important roles in neuropathic pain pathology and nociceptive sensations. Here, we investigated the involvement of DRG P2X3R and the effect of 2 Hz electroacupuncture (EA) on DNP. We monitored the rats' body weight, fasting blood glucose level, paw withdrawal thresholds, and paw withdrawal latency, and evaluated P2X3R expression in DRG. We found that P2X3R expression is upregulated on DNP, while 2 Hz EA is analgesic against DNP and suppresses P2X3R expression in DRG. To evaluate P2X3R involvement in pain modulation, we then treated the animals with A317491, a P2X3R specific antagonist, or α β-me ATP, a P2X3R agonist. We found that A317491 alleviates hyperalgesia, while α β-me ATP blocks EA's analgesic effects. Our findings indicated that 2 Hz EA alleviates DNP, possibly by suppressing P2X3R upregulation in DRG.
Collapse
Affiliation(s)
- Xueyu Fei
- Key Laboratory of Acupuncture and Neurology of Zhejiang Province, Department of Neurobiology and Acupuncture Research, The Third Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Xiaofen He
- Key Laboratory of Acupuncture and Neurology of Zhejiang Province, Department of Neurobiology and Acupuncture Research, The Third Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Zhaoxia Tai
- Key Laboratory of Acupuncture and Neurology of Zhejiang Province, Department of Neurobiology and Acupuncture Research, The Third Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Hanzhi Wang
- Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Siying Qu
- Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Luhang Chen
- Key Laboratory of Acupuncture and Neurology of Zhejiang Province, Department of Neurobiology and Acupuncture Research, The Third Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Qunqi Hu
- Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Jianqiao Fang
- Key Laboratory of Acupuncture and Neurology of Zhejiang Province, Department of Neurobiology and Acupuncture Research, The Third Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, 310053, China.
| | - Yongliang Jiang
- Key Laboratory of Acupuncture and Neurology of Zhejiang Province, Department of Neurobiology and Acupuncture Research, The Third Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, 310053, China.
| |
Collapse
|
55
|
Abstract
Diabetic retinopathy is now well understood as a neurovascular disease. Significant deficits early in diabetes are found in the inner retina that consists of bipolar cells that receive inputs from rod and cone photoreceptors, ganglion cells that receive inputs from bipolar cells, and amacrine cells that modulate these connections. These functional deficits can be measured in vivo in diabetic humans and animal models using the electroretinogram (ERG) and behavioral visual testing. Early effects of diabetes on both the human and animal model ERGs are changes to the oscillatory potentials that suggest dysfunctional communication between amacrine cells and bipolar cells as well as ERG measures that suggest ganglion cell dysfunction. These are coupled with changes in contrast sensitivity that suggest inner retinal changes. Mechanistic in vitro neuronal studies have suggested that these inner retinal changes are due to decreased inhibition in the retina, potentially due to decreased gamma aminobutyric acid (GABA) release, increased glutamate release, and increased excitation of retinal ganglion cells. Inner retinal deficits in dopamine levels have also been observed that can be reversed to limit inner retinal damage. Inner retinal targets present a promising new avenue for therapies for early-stage diabetic eye disease.
Collapse
|
56
|
Flood MD, Wellington AJ, Cruz LA, Eggers ED. Early diabetes impairs ON sustained ganglion cell light responses and adaptation without cell death or dopamine insensitivity. Exp Eye Res 2020; 200:108223. [PMID: 32910942 DOI: 10.1016/j.exer.2020.108223] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Revised: 08/17/2020] [Accepted: 09/03/2020] [Indexed: 10/23/2022]
Abstract
Retinal signaling under dark-adapted conditions is perturbed during early diabetes. Additionally, dopamine, the main neuromodulator of retinal light adaptation, is diminished in diabetic retinas. However, it is not known if this dopamine deficiency changes how the retina responds to increased light or dopamine. Here we determine whether light adaptation is impaired in the diabetic retina, and investigate potential mechanism(s) of impairment. Diabetes was induced in C57BL/6J male mice via 3 intraperitoneal injections of streptozotocin (75 mg/kg) and confirmed by blood glucose levels more than 200 mg/dL. After 6 weeks, whole-cell recordings of light-evoked and spontaneous inhibitory postsynaptic currents (IPSCs) or excitatory postsynaptic currents (EPSCs) were made from rod bipolar cells and ON sustained ganglion cells, respectively. Light responses were recorded before and after D1 receptor (D1R) activation (SKF-38393, 20 μM) or light adaptation (background of 950 photons·μm-2 ·s-1). Retinal whole mounts were stained for either tyrosine hydroxylase and activated caspase-3 or GAD65/67, GlyT1 and RBPMS and imaged. D1R activation and light adaptation both decreased inhibition, but the disinhibition was not different between control and diabetic rod bipolar cells. However, diabetic ganglion cell light-evoked EPSCs were increased in the dark and showed reduced light adaptation. No differences were found in light adaptation of spontaneous EPSC parameters, suggesting upstream changes. No changes in cell density were found for dopaminergic, glycinergic or GABAergic amacrine cells, or ganglion cells. Thus, in early diabetes, ON sustained ganglion cells receive excessive excitation under dark- and light-adapted conditions. Our results show that this is not attributable to loss in number or dopamine sensitivity of inhibitory amacrine cells or loss of dopaminergic amacrine cells.
Collapse
Affiliation(s)
- Michael D Flood
- Departments of Physiology and Biomedical Engineering, P.O. Box 245051, University of Arizona, Tucson, AZ, 85724, USA.
| | - Andrea J Wellington
- Departments of Physiology and Biomedical Engineering, P.O. Box 245051, University of Arizona, Tucson, AZ, 85724, USA.
| | - Luis A Cruz
- Departments of Physiology and Biomedical Engineering, P.O. Box 245051, University of Arizona, Tucson, AZ, 85724, USA.
| | - Erika D Eggers
- Departments of Physiology and Biomedical Engineering, P.O. Box 245051, University of Arizona, Tucson, AZ, 85724, USA.
| |
Collapse
|
57
|
Zhang Z, Zhou Y, Zhao H, Xu J, Yang X. Association Between Pathophysiological Mechanisms of Diabetic Retinopathy and Parkinson's Disease. Cell Mol Neurobiol 2020; 42:665-675. [PMID: 32880791 DOI: 10.1007/s10571-020-00953-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Accepted: 08/22/2020] [Indexed: 11/27/2022]
Abstract
Diabetic retinopathy, the most common complication of diabetes, is a neurodegenerative disease in the eye. And Parkinson's disease, affecting the health of 1-2% of people over 60 years old throughout the world, is the second largest neurodegenerative disease in the brain. As the understanding of diabetic retinopathy and Parkinson's disease deepens, the two diseases are found to show correlation in incidence, similarity in clinical presentation, and close association in pathophysiological mechanisms. To reveal the association between pathophysiological mechanisms of the two disease, in this review, the shared pathophysiological factors of diabetic retinopathy and Parkinson's disease are summarized and classified into dopaminergic system, circadian rhythm, neurotrophic factors, α-synuclein, and Wnt signaling pathways. Furthermore, similar and different mechanisms so far as the shared pathophysiological factors of the two disorders are discussed systematically. Finally, a brief summary and new perspectives are presented to provide new directions for further efforts on the association, exploration, and clinical prevention and treatment of diabetic retinopathy and Parkinson's disease.
Collapse
Affiliation(s)
- Zhuoqing Zhang
- Department of Ophthalmology, The Affiliated Hospital of Kunming University of Science and Technology, Kunming, Yunnan, China
- Department of Ophthalmology, The First People's Hospital of Yunnan Province, Kunming, Yunnan, China
| | - Yikun Zhou
- Department of Endocrinology, The First People's Hospital of Yunnan Province, Kunming, Yunnan, China
| | - Haiyan Zhao
- Department of Ophthalmology, The Affiliated Hospital of Kunming University of Science and Technology, Kunming, Yunnan, China
- Department of Ophthalmology, The First People's Hospital of Yunnan Province, Kunming, Yunnan, China
| | - Jinghui Xu
- Department of Ophthalmology, The Affiliated Hospital of Kunming University of Science and Technology, Kunming, Yunnan, China
- Department of Ophthalmology, The First People's Hospital of Yunnan Province, Kunming, Yunnan, China
| | - Xiaochun Yang
- Department of Ophthalmology, The Affiliated Hospital of Kunming University of Science and Technology, Kunming, Yunnan, China.
- Department of Ophthalmology, The First People's Hospital of Yunnan Province, Kunming, Yunnan, China.
| |
Collapse
|
58
|
Luo HH, Li J, Feng XF, Sun XY, Li J, Yang X, Fang ZZ. Plasma phenylalanine and tyrosine and their interactions with diabetic nephropathy for risk of diabetic retinopathy in type 2 diabetes. BMJ Open Diabetes Res Care 2020; 8:8/1/e000877. [PMID: 32883686 PMCID: PMC7473660 DOI: 10.1136/bmjdrc-2019-000877] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/03/2019] [Revised: 02/29/2020] [Accepted: 06/19/2020] [Indexed: 02/06/2023] Open
Abstract
OBJECTIVE Tight control of hyperglycemia reduces risk of diabetic retinopathy (DR), but the residual risk remains high. This study aimed to explore relationships between plasma phenylalanine and tyrosine with DR in type 2 diabetes (T2D) and interactions between the two amino acids, and their secondary interaction with renal dysfunction. RESEARCH DESIGN AND METHODS We extracted data of 1032 patients with T2D from tertiary hospital consecutively from May 2015 to August 2016. Binary logistic regression models with restricted cubic spline were used to check potential non-linear associations and to obtain ORs and 95% CIs of variables under study. Addictive interaction was estimated using relative excess risk due to interaction, attributable proportion due to interaction and synergy index. Area under the receiver operating characteristic curve was used to check increased predictive values. RESULTS Of 1032 patients, 162 suffered from DR. Copresence of low phenylalanine and low tyrosine increased DR risk (OR 6.01, 95% CI 1.35 to 26.8), while either of them alone did not have a significant effect with significant additive interaction. Presence of diabetic nephropathy further increased the OR of copresence of low phenylalanine and low tyrosine for DR to 25.9 (95% CI 8.71 to 76.9) with a significant additive interaction. Inclusion of phenylalanine and tyrosine in a traditional risk factor model significantly increased area under the curve from 0.81 to 0.83 (95% CI 0.80 to 0.86). CONCLUSION Plasma low phenylalanine and low tyrosine worked independently and synergistically to increase the risk of DR in T2D. Presence of renal dysfunction further amplified the effect of copresence of low phenylalanine and low tyrosine on DR risk.
Collapse
Affiliation(s)
- Hui-Huan Luo
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Tianjin Medical University, Tianjin, China
- Chinese Academy of Sciences, Dalian Institute of Chemical Physics, Dalian, China
| | - Juan Li
- Department of Radiotherapy Oncology, The Second Affiliated Hospital of Dalian Medical University, Dalian Medical University, Dalian, China
| | - Xiao-Fei Feng
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Tianjin Medical University, Tianjin, China
| | - Xiao-Yu Sun
- Chinese Academy of Sciences, Dalian Institute of Chemical Physics, Dalian, China
| | - Jing Li
- Department of Epidemiology and Biostatistics, School of Public Health, Tianjin Medical University, Tianjin, China
| | - Xilin Yang
- Department of Epidemiology and Biostatistics, School of Public Health, Tianjin Medical University, Tianjin, China
- Tianjin Key Laboratory of Environment, Nutrition and Public Health, Tianjin, China
| | - Zhong-Ze Fang
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Tianjin Medical University, Tianjin, China
- Chinese Academy of Sciences, Dalian Institute of Chemical Physics, Dalian, China
- Tianjin Key Laboratory of Environment, Nutrition and Public Health, Tianjin, China
| |
Collapse
|
59
|
Abstract
Diabetic retinopathy (DR) is a frequent complication of diabetes mellitus and an increasingly common cause of visual impairment. Blood vessel damage occurs as the disease progresses, leading to ischemia, neovascularization, blood-retina barrier (BRB) failure and eventual blindness. Although detection and treatment strategies have improved considerably over the past years, there is room for a better understanding of the pathophysiology of the diabetic retina. Indeed, it has been increasingly realized that DR is in fact a disease of the retina's neurovascular unit (NVU), the multi-cellular framework underlying functional hyperemia, coupling neuronal computations to blood flow. The accumulating evidence reveals that both neurochemical (synapses) and electrical (gap junctions) means of communications between retinal cells are affected at the onset of hyperglycemia, warranting a global assessment of cellular interactions and their role in DR. This is further supported by the recent data showing down-regulation of connexin 43 gap junctions along the vascular relay from capillary to feeding arteriole as one of the earliest indicators of experimental DR, with rippling consequences to the anatomical and physiological integrity of the retina. Here, recent advancements in our knowledge of mechanisms controlling the retinal neurovascular unit will be assessed, along with their implications for future treatment and diagnosis of DR.
Collapse
|
60
|
Motz CT, Chesler KC, Allen RS, Bales KL, Mees LM, Feola AJ, Maa AY, Olson DE, Thule PM, Iuvone PM, Hendrick AM, Pardue MT. Novel Detection and Restorative Levodopa Treatment for Preclinical Diabetic Retinopathy. Diabetes 2020; 69:1518-1527. [PMID: 32051147 PMCID: PMC7306127 DOI: 10.2337/db19-0869] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2019] [Accepted: 02/09/2020] [Indexed: 12/21/2022]
Abstract
Diabetic retinopathy (DR) is diagnosed clinically by directly viewing retinal vascular changes during ophthalmoscopy or through fundus photographs. However, electroretinography (ERG) studies in humans and rodents have revealed that retinal dysfunction is demonstrable prior to the development of visible vascular defects. Specifically, delays in dark-adapted ERG oscillatory potential (OP) implicit times in response to dim-flash stimuli (<-1.8 log cd · s/m2) occur prior to clinically recognized DR. Animal studies suggest that retinal dopamine deficiency underlies these early functional deficits. In this study, we randomized individuals with diabetes, without clinically detectable retinopathy, to treatment with either low- or high-dose Sinemet (levodopa plus carbidopa) for 2 weeks and compared their ERG findings with those of control subjects (no diabetes). We assessed dim-flash-stimulated OP delays using a novel handheld ERG system (RETeval) at baseline and 2 and 4 weeks. RETeval recordings identified significant OP implicit time delays in individuals with diabetes without retinopathy compared with age-matched control subjects (P < 0.001). After 2 weeks of Sinemet treatment, OP implicit times were restored to control values, and these improvements persisted even after a 2-week washout. We conclude that detection of dim-flash OP delays could provide early detection of DR and that Sinemet treatment may reverse retinal dysfunction.
Collapse
Affiliation(s)
- Cara T Motz
- Center for Visual and Neurocognitive Rehabilitation, Atlanta Veterans Affairs Medical Center, Decatur, GA
| | - Kyle C Chesler
- Center for Visual and Neurocognitive Rehabilitation, Atlanta Veterans Affairs Medical Center, Decatur, GA
- Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA
| | - Rachael S Allen
- Center for Visual and Neurocognitive Rehabilitation, Atlanta Veterans Affairs Medical Center, Decatur, GA
- Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA
| | - Katie L Bales
- Center for Visual and Neurocognitive Rehabilitation, Atlanta Veterans Affairs Medical Center, Decatur, GA
- Department of Ophthalmology, Emory University, Atlanta, GA
| | - Lukas M Mees
- Center for Visual and Neurocognitive Rehabilitation, Atlanta Veterans Affairs Medical Center, Decatur, GA
| | - Andrew J Feola
- Center for Visual and Neurocognitive Rehabilitation, Atlanta Veterans Affairs Medical Center, Decatur, GA
- Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA
| | - April Y Maa
- Center for Visual and Neurocognitive Rehabilitation, Atlanta Veterans Affairs Medical Center, Decatur, GA
- Department of Ophthalmology, Emory University, Atlanta, GA
| | - Darin E Olson
- Division of Endocrinology, Metabolism and Lipids, Emory University, Atlanta, GA
- Medical Service, Atlanta Veterans Affairs Medical Center, Decatur, GA
| | - Peter M Thule
- Division of Endocrinology, Metabolism and Lipids, Emory University, Atlanta, GA
- Medical Service, Atlanta Veterans Affairs Medical Center, Decatur, GA
| | - P Michael Iuvone
- Department of Ophthalmology, Emory University, Atlanta, GA
- Department of Pharmacology and Chemical Biology, Emory University, Atlanta, GA
| | | | - Machelle T Pardue
- Center for Visual and Neurocognitive Rehabilitation, Atlanta Veterans Affairs Medical Center, Decatur, GA
- Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA
- Department of Ophthalmology, Emory University, Atlanta, GA
| |
Collapse
|
61
|
Affiliation(s)
- Thomas J Wubben
- Kellogg Eye Center, Department of Ophthalmology and Visual Sciences, University of Michigan, Ann Arbor, MI
| |
Collapse
|
62
|
Abstract
Patients with diabetes continue to suffer from impaired visual performance before the appearance of overt damage to the retinal microvasculature and later sight-threatening complications. This diabetic retinopathy (DR) has long been thought to start with endothelial cell oxidative stress. Yet newer data surprisingly finds that the avascular outer retina is the primary site of oxidative stress before microvascular histopathology in experimental DR. Importantly, correcting this early oxidative stress is sufficient to restore vision and mitigate the histopathology in diabetic models. However, translating these promising results into the clinic has been stymied by an absence of methods that can measure and optimize anti-oxidant treatment efficacy in vivo. Here, we review imaging approaches that address this problem. In particular, diabetes-induced oxidative stress impairs dark-light regulation of subretinal space hydration, which regulates the distribution of interphotoreceptor binding protein (IRBP). IRBP is a vision-critical, anti-oxidant, lipid transporter, and pro-survival factor. We show how optical coherence tomography can measure subretinal space oxidative stress thus setting the stage for personalizing anti-oxidant treatment and prevention of impactful declines and loss of vision in patients with diabetes.
Collapse
|
63
|
Hendrick A, Smith J, Stelton C, Barb S, Yan J, Cribbs B, Jain N, Yeh S, Hubbard GB, He L, Dhakal S, Iuvone PM. Dopamine metabolite levels in the vitreous of diabetic and non-diabetic humans. Exp Eye Res 2020; 195:108040. [PMID: 32360553 DOI: 10.1016/j.exer.2020.108040] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2020] [Revised: 04/03/2020] [Accepted: 04/21/2020] [Indexed: 01/12/2023]
Abstract
Animal studies suggest that the retinal dysfunction in diabetic subjects that precedes overt clinical vasculopathy may be due to a retinal dopamine deficit. We analyzed levels of dopamine (DA) and its primary metabolite, 3,4-dihydroxyphenylacetic acid (DOPAC), in the vitreous of diabetic and non-diabetic human subjects. Adult patients undergoing pars plana vitrectomy for non-hemorrhagic indications were prospectively recruited from the Emory Eye Center in Atlanta, GA. Vitreous samples were analyzed using high performance liquid chromatography (HPLC) to measure levels of DOPAC and DA in the vitreous specimens. Vitreous samples from 9 diabetic patients and 20 from non-diabetic patients were analyzed. No eyes had apparent diabetic retinopathy. Mean normalized DA concentration in vitreous of diabetic subjects was 0.76 ± 0.12 pg/μL vs. 0.73 ± 0.08 pg/μL in non-diabetic vitreous (p = 0.849). DOPAC concentration was 8.84 ± 0.74 pg/μL in vitreous of diabetic subjects vs. 9.22 ± 0.56 pg/μL in vitreous of non-diabetic subjects (p = 0.691). No difference was observed in the concentrations of DA and DOPAC in the vitreous of people without diabetes compared to those with diabetes without retinopathy.
Collapse
Affiliation(s)
- Andrew Hendrick
- Department of Ophthalmology, Emory Eye Center, Emory University School of Medicine, Atlanta, GA, USA.
| | - Jesse Smith
- Department of Ophthalmology, Emory Eye Center, Emory University School of Medicine, Atlanta, GA, USA
| | - Chris Stelton
- Department of Ophthalmology, Emory Eye Center, Emory University School of Medicine, Atlanta, GA, USA
| | - Scott Barb
- Department of Ophthalmology, Emory Eye Center, Emory University School of Medicine, Atlanta, GA, USA
| | - Jiong Yan
- Department of Ophthalmology, Emory Eye Center, Emory University School of Medicine, Atlanta, GA, USA
| | - Blaine Cribbs
- Department of Ophthalmology, Emory Eye Center, Emory University School of Medicine, Atlanta, GA, USA
| | - Nieraj Jain
- Department of Ophthalmology, Emory Eye Center, Emory University School of Medicine, Atlanta, GA, USA
| | - Steve Yeh
- Department of Ophthalmology, Emory Eye Center, Emory University School of Medicine, Atlanta, GA, USA
| | - G Baker Hubbard
- Department of Ophthalmology, Emory Eye Center, Emory University School of Medicine, Atlanta, GA, USA
| | - Li He
- Department of Ophthalmology, Emory Eye Center, Emory University School of Medicine, Atlanta, GA, USA
| | - Susov Dhakal
- Department of Ophthalmology, Emory Eye Center, Emory University School of Medicine, Atlanta, GA, USA
| | - P Michael Iuvone
- Department of Ophthalmology, Emory Eye Center, Emory University School of Medicine, Atlanta, GA, USA; Department Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, GA, USA
| |
Collapse
|
64
|
Pavlenko TA, Chesnokova NB, Nodel MR, Kim AR, Ugrumov MV. Molecular Mechanisms and Clinical Manifestations of Catecholamine Dysfunction in the Eye in Parkinson's Disease As a Basis for Developing Early Diagnosis. Acta Naturae 2020; 12:52-62. [PMID: 32742727 PMCID: PMC7385097 DOI: 10.32607/actanaturae.10906] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Accepted: 04/16/2020] [Indexed: 01/08/2023] Open
Abstract
This review provides information on the non-motor peripheral manifestations of Parkinson's disease (PD) associated with a pathology of the visual analyzer and the auxiliary apparatus of the eye. The relationship between neurodegenerative processes that take place in the brain and in the eye opens new prospects to use preventive ophthalmologic examination to diagnose PD long before the characteristic motor symptoms appear. This will encourage the use of neuroprotective therapy, which stops, or at least slows down, neuronal death, instead of the current replacement therapy with dopamine agonists. An important result of an eye examination of patients with PD may be a non-invasive identification of new peripheral biomarkers manifesting themselves as changes in the composition of the lacrimal fluid.
Collapse
Affiliation(s)
- T. A. Pavlenko
- Helmholtz Moscow Research Institute of Eye Diseases of Ministry of Health of the Russian Federation, Moscow, 105062 Russia
| | - N. B. Chesnokova
- Helmholtz Moscow Research Institute of Eye Diseases of Ministry of Health of the Russian Federation, Moscow, 105062 Russia
| | - M. R. Nodel
- Sechenov First Moscow State Medical University, Moscow, 119991 Russia
- Pirogov Russian National Research Medical University, Russian Clinical and Research Center of Gerontology, Moscow, 129226 Russia
| | - A. R. Kim
- Koltzov Institute of Developmental Biology of Russian Academy of Sciences, Moscow, 119334 Russia
| | - M. V. Ugrumov
- Koltzov Institute of Developmental Biology of Russian Academy of Sciences, Moscow, 119334 Russia
| |
Collapse
|
65
|
Ou K, Copland DA, Theodoropoulou S, Mertsch S, Li Y, Liu J, Schrader S, Liu L, Dick AD. Treatment of diabetic retinopathy through neuropeptide Y-mediated enhancement of neurovascular microenvironment. J Cell Mol Med 2020; 24:3958-3970. [PMID: 32141716 PMCID: PMC7171318 DOI: 10.1111/jcmm.15016] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2019] [Revised: 12/02/2019] [Accepted: 12/27/2019] [Indexed: 12/12/2022] Open
Abstract
Diabetic retinopathy (DR) is one of the most severe clinical manifestations of diabetes mellitus and a major cause of blindness. DR is principally a microvascular disease, although the pathogenesis also involves metabolic reactive intermediates which induce neuronal and glial activation resulting in disruption of the neurovascular unit and regulation of the microvasculature. However, the impact of neural/glial activation in DR remains controversial, notwithstanding our understanding as to when neural/glial activation occurs in the course of disease. The objective of this study was to determine a potential protective role of neuropeptide Y (NPY) using an established model of DR permissive to N-methyl-D-aspartate (NMDA)-induced excitotoxic apoptosis of retinal ganglion cells (RGC) and vascular endothelial growth factor (VEGF)-induced vascular leakage. In vitro evaluation using primary retinal endothelial cells demonstrates that NPY promotes vascular integrity, demonstrated by maintained tight junction protein expression and reduced permeability in response to VEGF treatment. Furthermore, ex vivo assessment of retinal tissue explants shows that NPY can protect RGC from excitotoxic-induced apoptosis. In vivo clinical imaging and ex vivo tissue analysis in the diabetic model permitted assessment of NPY treatment in relation to neural and endothelial changes. The neuroprotective effects of NPY were confirmed by attenuating NMDA-induced retinal neural apoptosis and able to maintain inner retinal vascular integrity. These findings could have important clinical implications and offer novel therapeutic approaches for the treatment in the early stages of DR.
Collapse
Affiliation(s)
- Kepeng Ou
- College of Pharmacy, National and Local Joint Engineering Research Center of Targeted and Innovative Therapeutics, Chongqing Key Laboratory of Kinase Modulators as Innovative Medicine, Chongqing University of Arts and Sciences, Chongqing, China.,Laboratory for Experimental Ophthalmology, University of Düsseldorf, Düsseldorf, Germany.,Academic Unit of Ophthalmology, Bristol Medical School, University of Bristol, Bristol, UK
| | - David A Copland
- Academic Unit of Ophthalmology, Bristol Medical School, University of Bristol, Bristol, UK
| | - Sofia Theodoropoulou
- Academic Unit of Ophthalmology, Bristol Medical School, University of Bristol, Bristol, UK
| | - Sonja Mertsch
- Laboratory for Experimental Ophthalmology, University of Düsseldorf, Düsseldorf, Germany.,Department of Ophthalmology, Faculty of Medicine and Health Sciences, University of Oldenburg, Oldenburg, Germany
| | - Youjian Li
- College of Pharmacy, National and Local Joint Engineering Research Center of Targeted and Innovative Therapeutics, Chongqing Key Laboratory of Kinase Modulators as Innovative Medicine, Chongqing University of Arts and Sciences, Chongqing, China.,Academic Unit of Ophthalmology, Bristol Medical School, University of Bristol, Bristol, UK
| | - Jian Liu
- Academic Unit of Ophthalmology, Bristol Medical School, University of Bristol, Bristol, UK
| | - Stefan Schrader
- Laboratory for Experimental Ophthalmology, University of Düsseldorf, Düsseldorf, Germany.,Department of Ophthalmology, Faculty of Medicine and Health Sciences, University of Oldenburg, Oldenburg, Germany
| | - Lei Liu
- Academic Unit of Ophthalmology, Bristol Medical School, University of Bristol, Bristol, UK
| | - Andrew D Dick
- Academic Unit of Ophthalmology, Bristol Medical School, University of Bristol, Bristol, UK.,National Institute for Health Research (NIHR) Biomedical Research Centre at Moorfields Eye Hospital, University College London Institute of Ophthalmology, London, UK
| |
Collapse
|
66
|
Moore-Dotson JM, Eggers ED. Reductions in Calcium Signaling Limit Inhibition to Diabetic Retinal Rod Bipolar Cells. Invest Ophthalmol Vis Sci 2020; 60:4063-4073. [PMID: 31560762 PMCID: PMC6779064 DOI: 10.1167/iovs.19-27137] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/02/2022] Open
Abstract
Purpose The balance of neuronal excitation and inhibition is important for proper retinal signaling. A previous report showed that diabetes selectively reduces light-evoked inhibition to the retinal dim light rod pathway, changing this balance. Here, changes in mechanisms of retinal inhibitory synaptic transmission after 6 weeks of diabetes are investigated. Methods Diabetes was induced in C57BL/6J mice by three intraperitoneal injections of streptozotocin (STZ, 75 mg/kg), and confirmed by blood glucose levels more than 200 mg/dL. After 6 weeks, whole-cell voltage-clamp recordings of electrically evoked inhibitory postsynaptic currents from rod bipolar cells and light-evoked excitatory postsynaptic currents from A17-amacrine cells were made in dark-adapted retinal slices. Results Diabetes shortened the timecourse of directly activated lateral GABAergic inhibitory amacrine cell inputs to rod bipolar cells. The timing of GABA release onto rod bipolar cells depends on a prolonged amacrine cell calcium signal that is reduced by slow calcium buffering. Therefore, the effects of calcium buffering with EGTA-acetoxymethyl ester (AM) on diabetic GABAergic signaling were tested. EGTA-AM reduced GABAergic signaling in diabetic retinas more strongly, suggesting that diabetic amacrine cells have reduced calcium signals. Additionally, the timing of release from reciprocal inhibitory inputs to diabetic rod bipolar cells was reduced, but the activation of the A17 amacrine cells responsible for this inhibition was not changed. Conclusions These results suggest that reduced light-evoked inhibitory input to rod bipolar cells is due to reduced and shortened calcium signals in presynaptic GABAergic amacrine cells. A reduction in calcium signaling may be a common mechanism limiting inhibition in the retina.
Collapse
Affiliation(s)
- Johnnie M Moore-Dotson
- Departments of Physiology and Biomedical Engineering, University of Arizona, Tucson, Arizona, United States
| | - Erika D Eggers
- Departments of Physiology and Biomedical Engineering, University of Arizona, Tucson, Arizona, United States
| |
Collapse
|
67
|
Kurczab R, Kucwaj-Brysz K, Śliwa P. The Significance of Halogen Bonding in Ligand-Receptor Interactions: The Lesson Learned from Molecular Dynamic Simulations of the D 4 Receptor. Molecules 2019; 25:E91. [PMID: 31881785 PMCID: PMC6983170 DOI: 10.3390/molecules25010091] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2019] [Revised: 12/22/2019] [Accepted: 12/23/2019] [Indexed: 11/21/2022] Open
Abstract
Recently, a computational approach combining a structure-activity relationship library containing pairs of halogenated ligands and their corresponding unsubstituted ligands (called XSAR) with QM-based molecular docking and binding free energy calculations was developed and used to search for amino acids frequently targeted by halogen bonding, also known as XB hot spots. However, the analysis of ligand-receptor complexes with halogen bonds obtained by molecular docking provides a limited ability to study the role and significance of halogen bonding in biological systems. Thus, a set of molecular dynamics simulations for the dopamine D4 receptor, recently crystallized with the antipsychotic drug nemonapride (5WIU), and the five XSAR sets were performed to verify the identified hot spots for halogen bonding, in other words, primary (V5x40), and secondary (S5x43, S5x461 and H6x55). The simulations confirmed the key role of halogen bonding with V5x40 and H6x55 and supported S5x43 and S5x461. The results showed that steric restrictions and the topology of the molecular core have a crucial impact on the stabilization of the ligand-receptor complex by halogen bonding.
Collapse
Affiliation(s)
- Rafał Kurczab
- Department of Medicinal Chemistry, Maj Institute of Pharmacology, Polish Academy of Sciences, Smętna 12, 31-343 Cracow, Poland;
| | - Katarzyna Kucwaj-Brysz
- Department of Medicinal Chemistry, Maj Institute of Pharmacology, Polish Academy of Sciences, Smętna 12, 31-343 Cracow, Poland;
- Department of Technology and Biotechnology of Drugs, Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna 9, 30-688 Cracow, Poland
| | - Paweł Śliwa
- Faculty of Chemical Engineering and Technology, Cracow University of Technology, Warszawska 24, 31-155 Cracow, Poland;
| |
Collapse
|
68
|
Rojo Arias JE, Economopoulou M, Juárez López DA, Kurzbach A, Au Yeung KH, Englmaier V, Merdausl M, Schaarschmidt M, Ader M, Morawietz H, Funk RHW, Jászai J. VEGF-Trap is a potent modulator of vasoregenerative responses and protects dopaminergic amacrine network integrity in degenerative ischemic neovascular retinopathy. J Neurochem 2019; 153:390-412. [PMID: 31550048 DOI: 10.1111/jnc.14875] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2019] [Revised: 09/06/2019] [Accepted: 09/18/2019] [Indexed: 12/17/2022]
Abstract
Retinal hypoxia triggers abnormal vessel growth and microvascular hyper-permeability in ischemic retinopathies. Whereas vascular endothelial growth factor A (VEGF-A) inhibitors significantly hinder disease progression, their benefits to retinal neurons remain poorly understood. Similar to humans, oxygen-induced retinopathy (OIR) mice exhibit severe retinal microvascular malformations and profound neuronal dysfunction. OIR mice are thus a phenocopy of human retinopathy of prematurity, and a proxy for investigating advanced stages of proliferative diabetic retinopathy. Hence, the OIR model offers an excellent platform for assessing morpho-functional responses of the ischemic retina to anti-angiogenic therapies. Using this model, we investigated the retinal responses to VEGF-Trap (Aflibercept), an anti-angiogenic agent recognizing ligands of VEGF receptors 1 and 2 that possesses regulatory approval for the treatment of neovascular age-related macular degeneration, macular edema secondary to retinal vein occlusion and diabetic macular edema. Our results indicate that Aflibercept not only reduces the severity of retinal microvascular aberrations but also significantly improves neuroretinal function. Aflibercept administration significantly enhanced light-responsiveness, as revealed by electroretinographic examinations, and led to increased numbers of dopaminergic amacrine cells. Additionally, retinal transcriptional profiling revealed the concerted regulation of both angiogenic and neuronal targets, including transcripts encoding subunits of transmitter receptors relevant to amacrine cell function. Thus, Aflibercept represents a promising therapeutic alternative for the treatment of further progressive ischemic retinal neurovasculopathies beyond the set of disease conditions for which it has regulatory approval. Cover Image for this issue: doi: 10.1111/jnc.14743.
Collapse
Affiliation(s)
- Jesús E Rojo Arias
- Department of Anatomy, Medical Faculty Carl Gustav Carus, Technische Universität Dresden, Saxony, Germany
| | - Matina Economopoulou
- Department of Ophthalmology, University Hospital Carl Gustav Carus, Technische Universität Dresden, Saxony, Germany
| | - David A Juárez López
- Department of Anatomy, Medical Faculty Carl Gustav Carus, Technische Universität Dresden, Saxony, Germany
| | - Anica Kurzbach
- Medizinische Klinik III, Universitätsklinikum Carl Gustav Carus, Technische Universität Dresden, Saxony, Germany.,German Center for Diabetes Research (DZD e.V.), München-Neuherberg, Germany
| | - Kwan H Au Yeung
- Department of Anatomy, Medical Faculty Carl Gustav Carus, Technische Universität Dresden, Saxony, Germany
| | - Vanessa Englmaier
- Department of Anatomy, Medical Faculty Carl Gustav Carus, Technische Universität Dresden, Saxony, Germany
| | - Marie Merdausl
- Department of Anatomy, Medical Faculty Carl Gustav Carus, Technische Universität Dresden, Saxony, Germany
| | - Martin Schaarschmidt
- Department of Anatomy, Medical Faculty Carl Gustav Carus, Technische Universität Dresden, Saxony, Germany
| | - Marius Ader
- DFG-Center for Regenerative Therapies Dresden, Cluster of Excellence, Saxony, Germany
| | - Henning Morawietz
- Department of Medicine III, University Hospital Carl Gustav Carus, Division of Vascular Endothelium and Microcirculation, Technische Universität Dresden, Saxony, Germany
| | - Richard H W Funk
- Department of Anatomy, Medical Faculty Carl Gustav Carus, Technische Universität Dresden, Saxony, Germany
| | - József Jászai
- Department of Anatomy, Medical Faculty Carl Gustav Carus, Technische Universität Dresden, Saxony, Germany
| |
Collapse
|
69
|
Yin N, Yang YL, Cheng S, Wang HN, Hu X, Miao Y, Li F, Wang Z. Dopamine D2 Receptor-Mediated Modulation of Rat Retinal Ganglion Cell Excitability. Neurosci Bull 2019; 36:230-242. [PMID: 31606861 DOI: 10.1007/s12264-019-00431-3] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2019] [Accepted: 06/09/2019] [Indexed: 12/19/2022] Open
Abstract
Ganglion cells (RGCs) are the sole output neurons of the retinal circuity. Here, we investigated whether and how dopamine D2 receptors modulate the excitability of dissociated rat RGCs. Application of the selective D2 receptor agonist quinpirole inhibited outward K+ currents, which were mainly mediated by glybenclamide- and 4-aminopyridine-sensitive channels, but not the tetraethylammonium-sensitive channel. In addition, quinpirole selectively enhanced Nav1.6 voltage-gated Na+ currents. The intracellular cAMP/protein kinase A, Ca2+/calmodulin-dependent protein kinase II, and mitogen-activated protein kinase/extracellular signal-regulated kinase signaling pathways were responsible for the effects of quinpirole on K+ and Na+ currents, while phospholipase C/protein kinase C signaling was not involved. Under current-clamp conditions, the number of action potentials evoked by positive current injection was increased by quinpirole. Our results suggest that D2 receptor activation increases RGC excitability by suppressing outward K+ currents and enhancing Nav1.6 currents, which may affect retinal visual information processing.
Collapse
Affiliation(s)
- Ning Yin
- Department of Neurology, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Yu-Long Yang
- Department of Neurology, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Shuo Cheng
- Department of Neurology, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Hong-Ning Wang
- Department of Neurology, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Xin Hu
- Department of Neurology, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Yanying Miao
- Department of Neurology, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Fang Li
- Department of Neurology, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Zhongfeng Wang
- Department of Neurology, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Zhongshan Hospital, Fudan University, Shanghai, 200032, China.
| |
Collapse
|
70
|
Mees LM, Coulter MM, Chrenek MA, Motz CT, Landis EG, Boatright JH, Pardue MT. Low-Intensity Exercise in Mice Is Sufficient to Protect Retinal Function During Light-Induced Retinal Degeneration. Invest Ophthalmol Vis Sci 2019; 60:1328-1335. [PMID: 30933260 PMCID: PMC6445616 DOI: 10.1167/iovs.18-25883] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Purpose We previously reported that a specific treadmill running exercise regimen protects against light-induced retinal degeneration (LIRD) in mice. We hypothesized that this protective effect varies with running intensity. To test this, mice undergoing LIRD were run at different treadmill speeds and retinal function was assessed. Methods BALB/c mice were assigned to LIRD groups at varying treadmill speeds-0, 5, 10, or 20 m/min labeled inactive, low, medium, and high, respectively-and compared with naïve mice exposed to standard lighting (50 lux; naïve). Following 2 weeks of exercise, a subset of mice were exposed to toxic light (10,000 lux; LIRD) for 4 hours. After 5 additional days of exercise, retinal function was assessed by ERG. Corticosterone levels in serum and cathepsin B (CTSB) protein levels in muscle, brain, serum, and retina were measured. The retinal gene expression of complement factor 1qa (C1qa) and CTSB were measured. Results The low+LIRD and medium+LIRD exercise groups had greater a- and b-wave ERG amplitudes when compared with the inactive+LIRD group (P < 0.02). The high+LIRD mice only differed from the inactive+LIRD mice in their dark-adapted b-waves. Serum corticosterone increased in the high+LIRD mice (P < 0.006). Retinal CTSB protein levels were higher in the low+LIRD versus high+LIRD mice (P < 0.004) but were otherwise unchanged. Exercise of any intensity decreased C1qa gene expression. Conclusions Faster running did not additionally protect against LIRD, but it did increase serum corticosterone, suggesting stress-induced limits to exercise benefits. Unexpectedly, exercise did not increase CTSB proteins levels in muscle or serum, suggesting that it may not mediate exercise effects. Our results have implications for the use of low-intensity exercise as a vision loss treatment.
Collapse
Affiliation(s)
- Lukas M Mees
- Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, Georgia, United States.,Atlanta Veterans Administration Center for Visual and Neurocognitive Rehabilitation, Decatur, Georgia, United States
| | - Monica M Coulter
- Atlanta Veterans Administration Center for Visual and Neurocognitive Rehabilitation, Decatur, Georgia, United States
| | - Micah A Chrenek
- Department of Ophthalmology, Emory University, Atlanta, Georgia, United States
| | - Cara T Motz
- Atlanta Veterans Administration Center for Visual and Neurocognitive Rehabilitation, Decatur, Georgia, United States
| | - Erica G Landis
- Atlanta Veterans Administration Center for Visual and Neurocognitive Rehabilitation, Decatur, Georgia, United States.,Neuroscience Program, Laney Graduate School, Emory University, Atlanta, Georgia, United States
| | - Jeffrey H Boatright
- Atlanta Veterans Administration Center for Visual and Neurocognitive Rehabilitation, Decatur, Georgia, United States.,Department of Ophthalmology, Emory University, Atlanta, Georgia, United States
| | - Machelle T Pardue
- Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, Georgia, United States.,Atlanta Veterans Administration Center for Visual and Neurocognitive Rehabilitation, Decatur, Georgia, United States
| |
Collapse
|
71
|
Cui RZ, Wang L, Qiao SN, Wang YC, Wang X, Yuan F, Weng SJ, Yang XL, Zhong YM. ON-Type Retinal Ganglion Cells are Preferentially Affected in STZ-Induced Diabetic Mice. Invest Ophthalmol Vis Sci 2019; 60:1644-1656. [PMID: 30995300 DOI: 10.1167/iovs.18-26359] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Purpose We investigate morphologic and physiologic alterations of ganglion cells (GCs) in a streptozocin (STZ)-induced diabetic mouse model. Methods Experiments were conducted in flat-mount retinas of mice 3 months after the induction of diabetes. Changes in morphology of four subtypes of GCs (ON-type RGA2 [ON-RGA2], OFF-type RGA2 [OFF-RGA2], ON-type RGC1 [ON-RGC1], and ON-OFF type RGD2 [ON-OFF RGD2]) were characterized in Thy1-YFP transgenic mice. Using whole-cell patch-clamp recording, passive membrane properties and action potential (AP) firing properties were further investigated in transient ON- and OFF-RGA2 cells. Results Morphologic parameters were significantly altered in the dendrites branching in the ON sublamina of the inner plexiform layer (IPL) for ON-RGA2 cells and ON-OFF RGD2 cells. Much less significant changes, if any, were seen in those arborizing in the OFF sublamina of the IPL for OFF-RGA2 and ON-OFF RGD2 cells. No detectable changes in morphology were seen in RGC1 cells. Electrophysiologically, increased resting membrane potentials and decreased membrane capacitance were found in transient ON-RGA2 cells, but not in transient OFF-RGA2 cells. Similar alterations in AP firing properties, such as an increase in AP width and reduction in maximum spiking rate, were shared by these two subtypes. Furthermore, in response to depolarizing current injections, both cells generated more APs suggesting an enhanced excitability of these cells in diabetic conditions. Conclusions These differential changes in morphology and electrophysiology in subtypes of GCs may be responsible for reduced contrast sensitivity known to occur during the early stage of diabetic retinopathy.
Collapse
Affiliation(s)
- Run-Ze Cui
- Department of Neurology, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Zhongshan Hospital, Fudan University, Shanghai, People's Republic of China.,Department of Ophthalmology, Zhongshan Hospital, Fudan University, Shanghai, People's Republic of China
| | - Lu Wang
- Department of Neurology, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Zhongshan Hospital, Fudan University, Shanghai, People's Republic of China.,Department of Ophthalmology, Zhongshan Hospital, Fudan University, Shanghai, People's Republic of China
| | - Sheng-Nan Qiao
- Department of Neurology, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Zhongshan Hospital, Fudan University, Shanghai, People's Republic of China.,Department of Ophthalmology, Zhongshan Hospital, Fudan University, Shanghai, People's Republic of China
| | - Yong-Chen Wang
- Department of Neurology, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Zhongshan Hospital, Fudan University, Shanghai, People's Republic of China.,Department of Ophthalmology, Zhongshan Hospital, Fudan University, Shanghai, People's Republic of China
| | - Xin Wang
- Department of Neurology, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Zhongshan Hospital, Fudan University, Shanghai, People's Republic of China.,Department of Ophthalmology, Zhongshan Hospital, Fudan University, Shanghai, People's Republic of China
| | - Fei Yuan
- Department of Ophthalmology, Zhongshan Hospital, Fudan University, Shanghai, People's Republic of China
| | - Shi-Jun Weng
- Department of Neurology, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Zhongshan Hospital, Fudan University, Shanghai, People's Republic of China.,Department of Ophthalmology, Zhongshan Hospital, Fudan University, Shanghai, People's Republic of China
| | - Xiong-Li Yang
- Department of Neurology, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Zhongshan Hospital, Fudan University, Shanghai, People's Republic of China.,Department of Ophthalmology, Zhongshan Hospital, Fudan University, Shanghai, People's Republic of China
| | - Yong-Mei Zhong
- Department of Neurology, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Zhongshan Hospital, Fudan University, Shanghai, People's Republic of China.,Department of Ophthalmology, Zhongshan Hospital, Fudan University, Shanghai, People's Republic of China
| |
Collapse
|
72
|
Abstract
PURPOSE OF REVIEW To introduce recent advances in the understanding of diabetic retinopathy and to summarize current and emerging strategies to treat this common and complex cause of vision loss. RECENT FINDINGS Advances in retinal imaging and functional analysis indicate that retinal vascular and neural pathologies exist long before the development of clinically visible retinopathy. Such diagnostics could facilitate risk stratification and selective early intervention in high-risk patients. Antagonists of the vascular endothelial growth factor pathway effectively reduce vision loss in diabetes and promote regression of disease severity. Promising new strategies to treat diabetic retinopathy involve novel systemic diabetes therapy and ocular therapies that antagonize angiogenic growth factor signaling, improve blood-retina barrier function and neurovascular coupling, modulate neuroretinal metabolism, or provide neuroprotection. Long considered a pure microvasculopathy, diabetic retinopathy in fact affects the neural and vascular retina as well as neurovascular communication. Emerging therapies include those that target neuroretinal dysfunction in addition to those modulating vascular biology.
Collapse
Affiliation(s)
- Avinash Honasoge
- Department of Ophthalmology and Visual Sciences, Washington University School of Medicine, 660 S. Euclid Ave. 8096, St. Louis, MO, 63108, USA
| | - Eric Nudleman
- Shiley Eye Institute, University of California, San Diego, La Jolla, CA, USA
| | - Morton Smith
- Department of Ophthalmology and Visual Sciences, Washington University School of Medicine, 660 S. Euclid Ave. 8096, St. Louis, MO, 63108, USA
| | - Rithwick Rajagopal
- Department of Ophthalmology and Visual Sciences, Washington University School of Medicine, 660 S. Euclid Ave. 8096, St. Louis, MO, 63108, USA.
| |
Collapse
|
73
|
Sergeys J, Etienne I, Van Hove I, Lefevere E, Stalmans I, Feyen JHM, Moons L, Van Bergen T. Longitudinal In Vivo Characterization of the Streptozotocin-Induced Diabetic Mouse Model: Focus on Early Inner Retinal Responses. Invest Ophthalmol Vis Sci 2019; 60:807-822. [PMID: 30811545 DOI: 10.1167/iovs.18-25372] [Citation(s) in RCA: 56] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Purpose The goal of this study was to perform an extensive temporal characterization of the early pathologic processes in the streptozotocin (STZ)-induced diabetic retinopathy (DR) mouse model, beyond the vascular phenotype, and to investigate the potential of clinically relevant compounds in attenuating these processes. Methods Visual acuity and contrast sensitivity (CS) were studied in the mouse STZ model until 24 weeks postdiabetes onset. ERG, spectral domain optical coherence tomography (SD-OCT), leukostasis, and immunohistochemistry were applied to investigate neurodegeneration, inflammation, and gliosis during early-, mid- and late-phase diabetes. Aflibercept or triamcinolone acetonide (TAAC) was administered to investigate their efficacy on the aforementioned processes. Results Visual acuity and CS loss started at 4 and 18 weeks postdiabetes onset, respectively, and progressively declined over time. ERG amplitudes were diminished and OP latencies increased after 6 weeks, whereas SD-OCT revealed retinal thinning from 4 weeks postdiabetes. Immunohistochemical analyses linked these findings to retinal ganglion and cholinergic amacrine cell loss at 4 and 8 weeks postdiabetes onset, respectively, which was further decreased after aflibercept administration. The number of adherent leukocytes was augmented after 2 weeks, whereas increased micro- and macroglia reactivity was present from 4 weeks postdiabetes. Aflibercept or TAAC showed improved efficacy on inflammation and gliosis. Conclusions STZ-induced diabetic mice developed early pathologic DR hallmarks, from which inflammation seemed the initial trigger, leading to further development of functional and morphologic retinal changes. These findings indicate that the mouse STZ model is suitable to study novel integrative non-vascular therapies to treat early DR.
Collapse
Affiliation(s)
- Jurgen Sergeys
- Neural Circuit Development and Regeneration Research Group, Department of Biology, Zoological Institute, KU Leuven, Leuven, Belgium
| | | | - Inge Van Hove
- Neural Circuit Development and Regeneration Research Group, Department of Biology, Zoological Institute, KU Leuven, Leuven, Belgium.,Oxurion NV, Leuven, Belgium
| | - Evy Lefevere
- Neural Circuit Development and Regeneration Research Group, Department of Biology, Zoological Institute, KU Leuven, Leuven, Belgium
| | - Ingeborg Stalmans
- Laboratory of Experimental Ophthalmology, Department of Neurosciences, O&N II, KU Leuven, Leuven, Belgium
| | | | - Lieve Moons
- Neural Circuit Development and Regeneration Research Group, Department of Biology, Zoological Institute, KU Leuven, Leuven, Belgium
| | | |
Collapse
|
74
|
Allen RS, Feola A, Motz CT, Ottensmeyer AL, Chesler KC, Dunn R, Thulé PM, Pardue MT. Retinal Deficits Precede Cognitive and Motor Deficits in a Rat Model of Type II Diabetes. Invest Ophthalmol Vis Sci 2019; 60:123-133. [PMID: 30640976 DOI: 10.1167/iovs.18-25110] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Purpose To investigate the temporal appearance of retinal, cognitive, and motor deficits in Goto-Kakizaki (GK) rats, a spontaneously occurring, polygenic model of type II diabetes. GK rats develop impaired insulin secretion at 2 weeks and fasting hyperglycemia at 4 weeks. Methods In male and female GK rats and Wistar controls, glucose tolerance test (hyperglycemia) and electroretinogram (ERG, retinal function) were performed at 4 and 8 weeks of age. Spectral domain-optical coherence tomography (retinal structure) was assessed at 6 weeks. Spatial alternation (cognitive function) and number of entries (exploratory behavior) were assessed via Y-maze at 4, 5, 6, 7, and 8 weeks. Rotarod (motor function) was performed at 4, 6, and 8 weeks. Results By 4 weeks, the GK rats exhibited significant glucose intolerance (P < 0.001) and retinal deficits, including delays in ERG implicit times (flicker, P < 0.01; oscillatory potentials, P < 0.001). In addition, the GK rats showed greater ERG amplitudes (P < 0.001) and thinner retinas (P < 0.001). At 7 weeks, the GK rats showed deficits in cognitive function (P < 0.001) and exploratory behavior (P < 0.01). However, no motor function deficits were observed by 8 weeks. Interestingly, the male GK rats showed greater hyperglycemia (P < 0.05), but the female rats showed greater ERG delays (P < 0.001). Conclusions In GK rats, retinal function deficits developed prior to cognitive or motor deficits. Future studies will investigate common mechanistic links, long-term functional and vascular changes, and whether early retinal deficits can predict cognitive dysfunction or late-stage retinal disease.
Collapse
Affiliation(s)
- Rachael S Allen
- Atlanta VA Center for Visual and Neurocognitive Rehabilitation, Atlanta VA Health Care System, Decatur, Georgia, United States.,Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, Georgia, United States
| | - Andrew Feola
- Atlanta VA Center for Visual and Neurocognitive Rehabilitation, Atlanta VA Health Care System, Decatur, Georgia, United States.,Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, Georgia, United States
| | - Cara T Motz
- Atlanta VA Center for Visual and Neurocognitive Rehabilitation, Atlanta VA Health Care System, Decatur, Georgia, United States
| | - Amy L Ottensmeyer
- Atlanta VA Center for Visual and Neurocognitive Rehabilitation, Atlanta VA Health Care System, Decatur, Georgia, United States.,Department of Ophthalmology, Emory University, Atlanta, Georgia, United States
| | - Kyle C Chesler
- Atlanta VA Center for Visual and Neurocognitive Rehabilitation, Atlanta VA Health Care System, Decatur, Georgia, United States.,Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, Georgia, United States
| | - Ryan Dunn
- Atlanta VA Center for Visual and Neurocognitive Rehabilitation, Atlanta VA Health Care System, Decatur, Georgia, United States.,Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, Georgia, United States
| | - Peter M Thulé
- Section Endocrinology & Metabolism, Atlanta VA Health Care System & Emory University School of Medicine, Decatur, Georgia, United States
| | - Machelle T Pardue
- Atlanta VA Center for Visual and Neurocognitive Rehabilitation, Atlanta VA Health Care System, Decatur, Georgia, United States.,Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, Georgia, United States
| |
Collapse
|
75
|
Lakstygal AM, de Abreu MS, Lifanov DA, Wappler-Guzzetta EA, Serikuly N, Alpsyshov ET, Wang D, Wang M, Tang Z, Yan D, Demin KA, Volgin AD, Amstislavskaya TG, Wang J, Song C, Alekseeva P, Kalueff AV. Zebrafish models of diabetes-related CNS pathogenesis. Prog Neuropsychopharmacol Biol Psychiatry 2019; 92:48-58. [PMID: 30476525 DOI: 10.1016/j.pnpbp.2018.11.016] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/25/2018] [Revised: 11/18/2018] [Accepted: 11/22/2018] [Indexed: 12/12/2022]
Abstract
Diabetes mellitus (DM) is a common metabolic disorder that affects multiple organ systems. DM also affects brain processes, contributing to various CNS disorders, including depression, anxiety and Alzheimer's disease. Despite active research in humans, rodent models and in-vitro systems, the pathogenetic link between DM and brain disorders remains poorly understood. Novel translational models and new model organisms are therefore essential to more fully study the impact of DM on CNS. The zebrafish (Danio rerio) is a powerful novel model species to study metabolic and CNS disorders. Here, we discuss how DM alters brain functions and behavior in zebrafish, and summarize their translational relevance to studying DM-related CNS pathogenesis in humans. We recognize the growing utility of zebrafish models in translational DM research, as they continue to improve our understanding of different brain pathologies associated with DM, and may foster the discovery of drugs that prevent or treat these diseases.
Collapse
Affiliation(s)
- Anton M Lakstygal
- Institute of Translational Biomedicine, St. Petersburg State University, St. Petersburg, Russia; Laboratory of Preclinical Bioscreening, Granov Russian Research Center of Radiology and Surgical Technologies, Ministry of Healthcare of Russian Federation, Pesochny, Russia
| | - Murilo S de Abreu
- Bioscience Institute, University of Passo Fundo (UPF), Passo Fundo, RS, Brazil; The International Zebrafish Neuroscience Research Consortium (ZNRC), Slidell, LA, USA
| | - Dmitry A Lifanov
- Institute of Translational Biomedicine, St. Petersburg State University, St. Petersburg, Russia; Laboratory of Preclinical Bioscreening, Granov Russian Research Center of Radiology and Surgical Technologies, Ministry of Healthcare of Russian Federation, Pesochny, Russia; School of Pharmacy, Southwest University, Chongqing, China
| | | | - Nazar Serikuly
- School of Pharmacy, Southwest University, Chongqing, China
| | | | - DongMei Wang
- School of Pharmacy, Southwest University, Chongqing, China
| | - MengYao Wang
- School of Pharmacy, Southwest University, Chongqing, China
| | - ZhiChong Tang
- School of Pharmacy, Southwest University, Chongqing, China
| | - DongNi Yan
- School of Pharmacy, Southwest University, Chongqing, China
| | - Konstantin A Demin
- Institute of Experimental Medicine, Almazov National Medical Research Centre, Ministry of Healthcare of Russian Federation, St. Petersburg, Russia; Laboratory of Biological Psychiatry, Institute of Translational Biomedicine, St. Petersburg State University, St. Petersburg, Russia
| | - Andrey D Volgin
- Scientific Research Institute of Physiology and Basic Medicine, Novosibirsk, Russia
| | | | - JiaJia Wang
- Institute for Marine Drugs and Nutrition, Guangdong Ocean University, Zhanjiang, China; Marine Medicine Development Center, Shenzhen Institute, Guangdong Ocean University, Shenzhen, China
| | - Cai Song
- Institute for Marine Drugs and Nutrition, Guangdong Ocean University, Zhanjiang, China; Marine Medicine Development Center, Shenzhen Institute, Guangdong Ocean University, Shenzhen, China
| | - Polina Alekseeva
- Institute of Experimental Medicine, Almazov National Medical Research Centre, Ministry of Healthcare of Russian Federation, St. Petersburg, Russia
| | - Allan V Kalueff
- School of Pharmacy, Southwest University, Chongqing, China; Institute of Experimental Medicine, Almazov National Medical Research Centre, Ministry of Healthcare of Russian Federation, St. Petersburg, Russia; Laboratory of Biological Psychiatry, Institute of Translational Biomedicine, St. Petersburg State University, St. Petersburg, Russia; Scientific Research Institute of Physiology and Basic Medicine, Novosibirsk, Russia; Ural Federal University, Ekaterinburg, Russia; Russian Scientific Center of Radiology and Surgical Technologies, Ministry of Healthcare of Russian Federation, Pesochny, Russia; ZENEREI Research Center, Slidell, LA, USA.
| |
Collapse
|
76
|
Felder-Schmittbuhl MP, Buhr ED, Dkhissi-Benyahya O, Hicks D, Peirson SN, Ribelayga CP, Sandu C, Spessert R, Tosini G. Ocular Clocks: Adapting Mechanisms for Eye Functions and Health. Invest Ophthalmol Vis Sci 2019; 59:4856-4870. [PMID: 30347082 PMCID: PMC6181243 DOI: 10.1167/iovs.18-24957] [Citation(s) in RCA: 55] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Vision is a highly rhythmic function adapted to the extensive changes in light intensity occurring over the 24-hour day. This adaptation relies on rhythms in cellular and molecular processes, which are orchestrated by a network of circadian clocks located within the retina and in the eye, synchronized to the day/night cycle and which, together, fine-tune detection and processing of light information over the 24-hour period and ensure retinal homeostasis. Systematic or high throughput studies revealed a series of genes rhythmically expressed in the retina, pointing at specific functions or pathways under circadian control. Conversely, knockout studies demonstrated that the circadian clock regulates retinal processing of light information. In addition, recent data revealed that it also plays a role in development as well as in aging of the retina. Regarding synchronization by the light/dark cycle, the retina displays the unique property of bringing together light sensitivity, clock machinery, and a wide range of rhythmic outputs. Melatonin and dopamine play a particular role in this system, being both outputs and inputs for clocks. The retinal cellular complexity suggests that mechanisms of regulation by light are diverse and intricate. In the context of the whole eye, the retina looks like a major determinant of phase resetting for other tissues such as the retinal pigmented epithelium or cornea. Understanding the pathways linking the cell-specific molecular machineries to their cognate outputs will be one of the major challenges for the future.
Collapse
Affiliation(s)
- Marie-Paule Felder-Schmittbuhl
- Centre National de la Recherche Scientifique, Université de Strasbourg, Institut des Neurosciences Cellulaires et Intégratives (UPR 3212), Strasbourg, France
| | - Ethan D Buhr
- Department of Ophthalmology, University of Washington Medical School, Seattle, Washington, United States
| | - Ouria Dkhissi-Benyahya
- Univ Lyon, Université Claude Bernard Lyon 1, Inserm, Stem Cell and Brain Research Institute U1208, Bron, France
| | - David Hicks
- Centre National de la Recherche Scientifique, Université de Strasbourg, Institut des Neurosciences Cellulaires et Intégratives (UPR 3212), Strasbourg, France
| | - Stuart N Peirson
- Sleep and Circadian Neuroscience Institute (SCNi), Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, United Kingdom
| | - Christophe P Ribelayga
- Ruiz Department of Ophthalmology and Visual Science, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, Texas, United States
| | - Cristina Sandu
- Centre National de la Recherche Scientifique, Université de Strasbourg, Institut des Neurosciences Cellulaires et Intégratives (UPR 3212), Strasbourg, France
| | - Rainer Spessert
- Institute of Functional and Clinical Anatomy, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Gianluca Tosini
- Neuroscience Institute and Department of Pharmacology & Toxicology, Morehouse School of Medicine, Atlanta, Georgia, United States
| |
Collapse
|
77
|
Mazade RE, Flood MD, Eggers ED. Dopamine D1 receptor activation reduces local inner retinal inhibition to light-adapted levels. J Neurophysiol 2019; 121:1232-1243. [PMID: 30726156 PMCID: PMC6485729 DOI: 10.1152/jn.00448.2018] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2018] [Revised: 02/04/2019] [Accepted: 02/05/2019] [Indexed: 01/26/2023] Open
Abstract
During adaptation from dim to bright environments, changes in retinal signaling are mediated, in part, by dopamine. Dopamine is released with light and can modulate retinal receptive fields, neuronal coupling, inhibitory receptors, and rod pathway inhibition. However, it is unclear how dopamine affects inner retinal inhibition to cone bipolar cells, which relay visual information from photoreceptors to ganglion cells and are important signal processing sites. We tested the hypothesis that dopamine (D)1 receptor activation is sufficient to elicit light-adapted inhibitory changes. Local light-evoked inhibition and spontaneous activity were measured from OFF cone bipolar cells in dark-adapted mouse retinas while stimulating D1 receptors, which are located on bipolar, horizontal, and inhibitory amacrine cells. The D1 agonist SKF38393 reduced local inhibitory light-evoked response magnitude and increased response transience, which mimicked changes measured with light adaptation. D1-mediated reductions in local inhibition were more pronounced for glycinergic than GABAergic inputs, comparable with light adaptation. The effects of D1 receptors on light-evoked input were similar to the effects on spontaneous input. D1 receptor activation primarily decreased glycinergic spontaneous current frequency, similar to light adaptation, suggesting mainly a presynaptic amacrine cell site of action. These results expand the role of dopamine to include signal modulation of cone bipolar cell local inhibition. In this role, D1 receptor activation, acting primarily through glycinergic amacrine cells, may be an important mechanism for the light-adapted reduction in OFF bipolar cell inhibition since the actions are similar and dopamine is released during light adaptation. NEW & NOTEWORTHY Retinal adaptation to different luminance conditions requires the adjustment of local circuits for accurate signaling of visual scenes. Understanding mechanisms behind luminance adaptation at different retinal levels is important for understanding how the retina functions in a dynamic environment. In the mouse, we show that dopamine pathways reduce inner retinal inhibition similar to increased background luminance, suggesting the two are linked and highlighting a possible mechanism for light adaptation at an early retinal processing center.
Collapse
Affiliation(s)
- Reece E Mazade
- Departments of Physiology and Biomedical Engineering, University of Arizona , Tucson, Arizona
| | - Michael D Flood
- Departments of Physiology and Biomedical Engineering, University of Arizona , Tucson, Arizona
| | - Erika D Eggers
- Departments of Physiology and Biomedical Engineering, University of Arizona , Tucson, Arizona
| |
Collapse
|
78
|
Zafar S, Sachdeva M, Frankfort BJ, Channa R. Retinal Neurodegeneration as an Early Manifestation of Diabetic Eye Disease and Potential Neuroprotective Therapies. Curr Diab Rep 2019; 19:17. [PMID: 30806815 PMCID: PMC7192364 DOI: 10.1007/s11892-019-1134-5] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
PURPOSE OF REVIEW Diabetic retinopathy (DR) is a major cause of visual impairment and blindness throughout the world. Microvascular changes have long been regarded central to disease pathogenesis. In recent years, however, retinal neurodegeneration is increasingly being hypothesized to occur prior to the vascular changes classically associated with DR and contribute to disease pathogenesis. RECENT FINDINGS There is growing structural and functional evidence from human and animal studies that suggests retinal neurodegeneration to be an early component of DR. Identification of new therapeutic targets is an ongoing area of research with several different molecules undergoing testing in animal models for their neuroprotective properties and for possible use in humans. Retinal neurodegeneration may play a central role in DR pathogenesis. As new therapies are developed, it will be important to develop criteria for clinically defining retinal neurodegeneration. A standardization of the methods for monitoring neurodegeneration along with more sensitive means of detecting preclinical damage is also needed.
Collapse
Affiliation(s)
- Sidra Zafar
- Wilmer Eye Institute, Johns Hopkins Hospital, Baltimore, MD 21287, USA
| | - Mira Sachdeva
- Wilmer Eye Institute, Johns Hopkins Hospital, Baltimore, MD 21287, USA
| | | | - Roomasa Channa
- Wilmer Eye Institute, Johns Hopkins Hospital, Baltimore, MD 21287, USA
- Department of Ophthalmology, Baylor College of Medicine, Houston, TX, USA
| |
Collapse
|
79
|
Sundstrom JM, Hernández C, Weber SR, Zhao Y, Dunklebarger M, Tiberti N, Laremore T, Simó-Servat O, Garcia-Ramirez M, Barber AJ, Gardner TW, Simó R. Proteomic Analysis of Early Diabetic Retinopathy Reveals Mediators of Neurodegenerative Brain Diseases. Invest Ophthalmol Vis Sci 2019; 59:2264-2274. [PMID: 29847632 PMCID: PMC5935294 DOI: 10.1167/iovs.17-23678] [Citation(s) in RCA: 72] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Purpose Current evidence suggests that retinal neurodegeneration is an early event in the pathogenesis of diabetic retinopathy. Our main goal was to examine whether, in the diabetic human retina, common proteins and pathways are shared with brain neurodegenerative diseases. Methods A proteomic analysis was performed on three groups of postmortem retinas matched by age: nondiabetic control retinas (n = 5), diabetic retinas without glial activation (n = 5), and diabetic retinas with glial activation (n = 5). Retinal lysates from each group were pooled and run on an SDS-PAGE gel. Bands were analyzed sequentially by liquid chromatography-mass spectrometry (LC/MS) using an Orbitrap Mass Spectrometer. Results A total of 2190 proteins were identified across all groups. To evaluate the association of the identified proteins with neurological signaling, significant signaling pathways belonging to the category “Neurotransmitters and Other Nervous System Signaling” were selected for analysis. Pathway analysis revealed that “Neuroprotective Role of THOP1 in Alzheimer's Disease” and “Unfolded Protein Response” pathways were uniquely enriched in control retinas. By contrast, “Dopamine Degradation” and “Parkinson's Signaling” were enriched only in diabetic retinas with glial activation. The “Neuregulin Signaling,” “Synaptic Long Term Potentiation,” and “Amyloid Processing” pathways were enriched in diabetic retinas with no glial activation. Conclusions Diabetes-induced retinal neurodegeneration and brain neurodegenerative diseases, such as Alzheimer's and Parkinson's diseases, share common pathogenic pathways. These findings suggest that the study of neurodegeneration in the diabetic retina could be useful to further understand the neurodegenerative processes that occur in the brain of persons with diabetes.
Collapse
Affiliation(s)
- Jeffrey M Sundstrom
- Penn State Hershey Eye Center, Penn State College of Medicine, Hershey, Pennsylvania, United States
| | - Cristina Hernández
- Institut de Recerca Hospital Universitari Vall d'Hebron (VHIR), Barcelona, Spain.,Instituto de Salud Carlos III (CIBERDEM), Barcelona, Spain
| | - Sarah R Weber
- Penn State Hershey Eye Center, Penn State College of Medicine, Hershey, Pennsylvania, United States
| | - Yuanjun Zhao
- Penn State Hershey Eye Center, Penn State College of Medicine, Hershey, Pennsylvania, United States
| | - Mitchell Dunklebarger
- Penn State Hershey Eye Center, Penn State College of Medicine, Hershey, Pennsylvania, United States
| | | | - Tatiana Laremore
- Proteomics and Mass Spectrometry Core Facility, Penn State University, Pennsylvania, United States
| | - Olga Simó-Servat
- Institut de Recerca Hospital Universitari Vall d'Hebron (VHIR), Barcelona, Spain.,Instituto de Salud Carlos III (CIBERDEM), Barcelona, Spain
| | - Marta Garcia-Ramirez
- Institut de Recerca Hospital Universitari Vall d'Hebron (VHIR), Barcelona, Spain.,Instituto de Salud Carlos III (CIBERDEM), Barcelona, Spain
| | - Alistair J Barber
- Penn State Hershey Eye Center, Penn State College of Medicine, Hershey, Pennsylvania, United States
| | - Thomas W Gardner
- Kellogg Eye Center, University of Michigan Medical School, Ann Arbor, Michigan, United States
| | - Rafael Simó
- Institut de Recerca Hospital Universitari Vall d'Hebron (VHIR), Barcelona, Spain.,Instituto de Salud Carlos III (CIBERDEM), Barcelona, Spain
| |
Collapse
|
80
|
Matzinger M, Fischhuber K, Heiss EH. Activation of Nrf2 signaling by natural products-can it alleviate diabetes? Biotechnol Adv 2018; 36:1738-1767. [PMID: 29289692 PMCID: PMC5967606 DOI: 10.1016/j.biotechadv.2017.12.015] [Citation(s) in RCA: 140] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2017] [Revised: 12/19/2017] [Accepted: 12/26/2017] [Indexed: 02/06/2023]
Abstract
Type 2 diabetes mellitus (DM) has reached pandemic proportions and effective prevention strategies are wanted. Its onset is accompanied by cellular distress, the nuclear factor erythroid 2-related factor 2 (Nrf2) is a transcription factor boosting cytoprotective responses, and many phytochemicals activate Nrf2 signaling. Thus, Nrf2 activation by natural products could presumably alleviate DM. We summarize function, regulation and exogenous activation of Nrf2, as well as diabetes-linked and Nrf2-susceptible forms of cellular stress. The reported amelioration of insulin resistance, β-cell dysfunction and diabetic complications by activated Nrf2 as well as the status quo of Nrf2 in precision medicine for DM are reviewed.
Collapse
Affiliation(s)
- Manuel Matzinger
- University of Vienna, Department of Pharmacognosy, Althanstrasse 14, 1090 Vienna, Austria
| | - Katrin Fischhuber
- University of Vienna, Department of Pharmacognosy, Althanstrasse 14, 1090 Vienna, Austria
| | - Elke H Heiss
- University of Vienna, Department of Pharmacognosy, Althanstrasse 14, 1090 Vienna, Austria.
| |
Collapse
|
81
|
Lee SE, Han K, Baek JY, Ko KS, Lee KU, Koh EH. Association Between Diabetic Retinopathy and Parkinson Disease: The Korean National Health Insurance Service Database. J Clin Endocrinol Metab 2018; 103:3231-3238. [PMID: 29982669 DOI: 10.1210/jc.2017-02774] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2017] [Accepted: 06/27/2018] [Indexed: 12/13/2022]
Abstract
CONTEXT Studies have shown an association between diabetes and Parkinson disease (PD). The retina is a part of the central nervous system; it was proposed that diabetic retinopathy (DR) and PD share common pathophysiology of dopamine deficiency. However, no epidemiologic studies have investigated the relationship between these two diseases. OBJECTIVE We assessed the association between DR and incident PD using a population-based database. DESIGN/SETTING/PARTICIPANTS Using the Korean National Health Insurance Service database, 14,912,368 participants who underwent regular health checkup from 2005 to 2008 were included. Subjects were classified into non-diabetes, diabetes without DR, and diabetes with DR groups at baseline and followed up until the date of PD incidence, death, or 31 December 2013. Cox proportional hazards regression analysis was used to evaluate the association between DR and incident PD. RESULTS During the period, 34,834 subjects were newly diagnosed with PD. The incidence of PD was 2.74, 8.39, and 15.51 per 10,000 person-years for the non-diabetes, diabetes without DR, and diabetes with DR groups, respectively. In multivariate Cox proportional hazard models, DR groups were associated with significantly higher risk of PD than non-diabetes or diabetes without DR groups even after adjusting for age, sex, fasting plasma glucose level, insulin usage, and other possible risk factors. CONCLUSION Concurrent DR was associated with an increased risk of incident PD. Future studies are necessary to investigate the mechanism of increased risk of PD in DR including dopamine deficiency in the central nervous system and long-lasting poor glycemic control.
Collapse
Affiliation(s)
- Seung Eun Lee
- Department of Internal Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Kyungdo Han
- Department of Biostatistics, The Catholic University of Korea, Seoul, Korea
| | - Ji Yeon Baek
- Department of Internal Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Kyung Soo Ko
- Department of Internal Medicine, Cardiovascular and Metabolic Disease Center, Inje University Sanggye Paik Hospital, Inje University College of Medicine, Seoul, Korea
| | - Ki-Up Lee
- Department of Internal Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Eun Hee Koh
- Department of Internal Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | | |
Collapse
|
82
|
Kim MK, Aung MH, Mees L, Olson DE, Pozdeyev N, Iuvone PM, Thule PM, Pardue MT. Dopamine Deficiency Mediates Early Rod-Driven Inner Retinal Dysfunction in Diabetic Mice. Invest Ophthalmol Vis Sci 2018; 59:572-581. [PMID: 29372256 PMCID: PMC5788047 DOI: 10.1167/iovs.17-22692] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Purpose Electroretinograms (ERGs) are abnormal in diabetic retinas before the appearance of vascular lesions, providing a possible biomarker for diabetic vision loss. Previously, we reported that decreased retinal dopamine (DA) levels in diabetic rodents contributed to early visual and retinal dysfunction. In the current study, we examined whether oscillatory potentials (OPs) could serve as a potential marker for detecting early inner retinal dysfunction due to retinal DA deficiency. Methods Retinal function was tested with dark-adapted ERGs, taken at 3, 4, and 5 weeks after diabetes induction with streptozotocin. Electrical responses were analyzed and correlations were made with previously reported retinal DA levels. The effect of restoring systemic DA levels or removing DA from the retina in diabetic mice on OPs was assessed using L-3,4-dihydroxyphenylalanine (L-DOPA) treatments and retina-specific tyrosine hydroxylase (Th) knockout mice (rTHKO), respectively. Results Diabetic animals had significantly delayed OPs compared to control animals in response to dim, but not bright, flash stimuli. L-DOPA treatment preserved OP implicit time in diabetic mice. Diabetic rTHKO mice had further delayed OPs compared to diabetic mice with normal retinal Th, with L-DOPA treatment also providing benefit. Decreasing retinal DA levels significantly correlated with increasing OP delays mediated by rod pathways. Conclusions Our data suggest that inner retinal dysfunction in early-stage diabetes is mediated by rod-pathway deficits and DA deficiencies. OP delays may be used to determine the earliest functional deficits in diabetic retinopathy and to establish an early treatment window for DA therapies that may prevent progressive vision loss.
Collapse
Affiliation(s)
- Moon K Kim
- Department of Ophthalmology, Emory University, Atlanta, Georgia, United States
| | - Moe H Aung
- Neuroscience, Emory University, Atlanta, Georgia, United States
| | - Lukas Mees
- Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, Georgia, United States.,Center for Visual and Neurocognitive Rehabilitation, Atlanta Veterans Affairs Medical Center, Decatur, Georgia, United States
| | - Darin E Olson
- Division of Endocrinology, Metabolism and Lipids, Emory University, Atlanta, Georgia, United States.,Medical Service, Atlanta Veterans Affairs Medical Center, Decatur, Georgia, United States
| | - Nikita Pozdeyev
- Department of Ophthalmology, Emory University, Atlanta, Georgia, United States.,Department of Pharmacology, Emory University, Atlanta, Georgia, United States
| | - P Michael Iuvone
- Department of Ophthalmology, Emory University, Atlanta, Georgia, United States.,Neuroscience, Emory University, Atlanta, Georgia, United States.,Department of Pharmacology, Emory University, Atlanta, Georgia, United States
| | - Peter M Thule
- Division of Endocrinology, Metabolism and Lipids, Emory University, Atlanta, Georgia, United States.,Medical Service, Atlanta Veterans Affairs Medical Center, Decatur, Georgia, United States
| | - Machelle T Pardue
- Department of Ophthalmology, Emory University, Atlanta, Georgia, United States.,Neuroscience, Emory University, Atlanta, Georgia, United States.,Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, Georgia, United States.,Center for Visual and Neurocognitive Rehabilitation, Atlanta Veterans Affairs Medical Center, Decatur, Georgia, United States
| |
Collapse
|
83
|
Pardue MT, Allen RS. Neuroprotective strategies for retinal disease. Prog Retin Eye Res 2018; 65:50-76. [PMID: 29481975 PMCID: PMC6081194 DOI: 10.1016/j.preteyeres.2018.02.002] [Citation(s) in RCA: 158] [Impact Index Per Article: 22.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2017] [Revised: 02/14/2018] [Accepted: 02/20/2018] [Indexed: 12/20/2022]
Abstract
Diseases that affect the eye, including photoreceptor degeneration, diabetic retinopathy, and glaucoma, affect 11.8 million people in the US, resulting in vision loss and blindness. Loss of sight affects patient quality of life and puts an economic burden both on individuals and the greater healthcare system. Despite the urgent need for treatments, few effective options currently exist in the clinic. Here, we review research on promising neuroprotective strategies that promote neuronal survival with the potential to protect against vision loss and retinal cell death. Due to the large number of neuroprotective strategies, we restricted our review to approaches that we had direct experience with in the laboratory. We focus on drugs that target survival pathways, including bile acids like UDCA and TUDCA, steroid hormones like progesterone, therapies that target retinal dopamine, and neurotrophic factors. In addition, we review rehabilitative methods that increase endogenous repair mechanisms, including exercise and electrical stimulation therapies. For each approach, we provide background on the neuroprotective strategy, including history of use in other diseases; describe potential mechanisms of action; review the body of research performed in the retina thus far, both in animals and in humans; and discuss considerations when translating each treatment to the clinic and to the retina, including which therapies show the most promise for each retinal disease. Despite the high incidence of retinal diseases and the complexity of mechanisms involved, several promising neuroprotective treatments provide hope to prevent blindness. We discuss attractive candidates here with the goal of furthering retinal research in critical areas to rapidly translate neuroprotective strategies into the clinic.
Collapse
Affiliation(s)
- Machelle T Pardue
- Center for Visual and Neurocognitive Rehabilitation, Atlanta VA Medical Center, 1670 Clairmont Road, Decatur, GA, 30033, USA; Department of Biomedical Engineering, Georgia Institute of Technology, 313 Ferst Drive, Atlanta, GA, 30332, USA.
| | - Rachael S Allen
- Center for Visual and Neurocognitive Rehabilitation, Atlanta VA Medical Center, 1670 Clairmont Road, Decatur, GA, 30033, USA
| |
Collapse
|
84
|
Joyce DS, Feigl B, Kerr G, Roeder L, Zele AJ. Melanopsin-mediated pupil function is impaired in Parkinson's disease. Sci Rep 2018; 8:7796. [PMID: 29773814 PMCID: PMC5958070 DOI: 10.1038/s41598-018-26078-0] [Citation(s) in RCA: 55] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2017] [Accepted: 04/30/2018] [Indexed: 01/16/2023] Open
Abstract
Parkinson's disease (PD) is characterised by non-motor symptoms including sleep and circadian disruption. Melanopsin-expressing intrinsically photosensitive Retinal Ganglion Cells (ipRGC) transmit light signals to brain areas controlling circadian rhythms and the pupil light reflex. To determine if non-motor symptoms observed in PD are linked to ipRGC dysfunction, we evaluated melanopsin and rod/cone contributions to the pupil response in medicated participants with PD (n = 17) and controls (n = 12). Autonomic tone was evaluated by measuring pupillary unrest in darkness. In the PD group, there is evidence for an attenuated post-illumination pupil response (PIPR) amplitude and reduced pupil constriction amplitude, and PIPR amplitudes did not correlate with measures of sleep quality, retinal nerve fibre layer thickness, disease severity, or medication dosage. Both groups exhibited similar pupillary unrest. We show that melanopsin- and the rod/cone-photoreceptor contributions to the pupil control pathway are impaired in people with early-stage PD who have no clinically observable ophthalmic abnormalities. Given that ipRGCs project to brain targets involved in arousal, sleep and circadian rhythms, ipRGC dysfunction may underpin some of the non-motor symptoms observed in PD.
Collapse
Affiliation(s)
- Daniel S Joyce
- Institute of Health and Biomedical Innovation, Queensland University of Technology (QUT), Brisbane, Australia
- Visual Science Laboratory, School of Optometry and Vision Science, Queensland University of Technology (QUT), Brisbane, Australia
- Department of Psychiatry and Behavioral Sciences, School of Medicine, Stanford University, Stanford, USA
| | - Beatrix Feigl
- Institute of Health and Biomedical Innovation, Queensland University of Technology (QUT), Brisbane, Australia
- Medical Retina Laboratory, School of Biomedical Sciences, Queensland University of Technology (QUT), Brisbane, Australia
- Queensland Eye Institute, Brisbane, Australia
| | - Graham Kerr
- Institute of Health and Biomedical Innovation, Queensland University of Technology (QUT), Brisbane, Australia
- Movement Neuroscience Program, Queensland University of Technology (QUT), Brisbane, Australia
| | - Luisa Roeder
- Institute of Health and Biomedical Innovation, Queensland University of Technology (QUT), Brisbane, Australia
- Movement Neuroscience Program, Queensland University of Technology (QUT), Brisbane, Australia
| | - Andrew J Zele
- Institute of Health and Biomedical Innovation, Queensland University of Technology (QUT), Brisbane, Australia.
- Visual Science Laboratory, School of Optometry and Vision Science, Queensland University of Technology (QUT), Brisbane, Australia.
| |
Collapse
|
85
|
Allen RS, Hanif AM, Gogniat MA, Prall BC, Haider R, Aung MH, Prunty MC, Mees LM, Coulter MM, Motz CT, Boatright JH, Pardue MT. TrkB signalling pathway mediates the protective effects of exercise in the diabetic rat retina. Eur J Neurosci 2018. [PMID: 29537701 DOI: 10.1111/ejn.13909] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Diabetic retinopathy is a leading cause of vision loss. Treatment options for early retinopathy are sparse. Exercise protects dying photoreceptors in models of retinal degeneration, thereby preserving vision. We tested the protective effects of exercise on retinal and cognitive deficits in a type 1 diabetes model and determined whether the TrkB pathway mediates this effect. Hyperglycaemia was induced in Long Evans rats via streptozotocin injection (STZ; 100 mg/kg). Following confirmed hyperglycaemia, both control and diabetic rats underwent treadmill exercise for 30 min, 5 days/week at 0 m/min (inactive groups) or 15 m/min (active groups) for 8 weeks. A TrkB receptor antagonist (ANA-12), or vehicle, was injected 2.5 h before exercise training. We measured spatial frequency and contrast sensitivity using optokinetic tracking biweekly post-STZ; retinal function using electroretinography at 4 and 8 weeks; and cognitive function and exploratory behaviour using Y-maze at 8 weeks. Retinal neurotrophin-4 was measured using ELISA. Compared with non-diabetic controls, diabetic rats showed significantly reduced spatial frequency and contrast sensitivity, delayed electroretinogram oscillatory potential and flicker implicit times and reduced cognitive function and exploratory behaviour. Exercise interventions significantly delayed the appearance of all deficits, except for exploratory behaviour. Treatment with ANA-12 significantly reduced this protection, suggesting a TrkB-mediated mechanism. Despite this, no changes in retinal neurotrohin-4 were observed with diabetes or exercise. Exercise protected against early visual and cognitive dysfunction in diabetic rats, suggesting that exercise interventions started after hyperglycaemia diagnosis may be a beneficial treatment. The translational potential is high, given that exercise treatment is non-invasive, patient controlled and inexpensive.
Collapse
Affiliation(s)
- Rachael S Allen
- Center for Visual and Neurocognitive Rehabilitation, Atlanta VA Medical Center, 1670 Clairmont Road, Decatur, GA, 30033, USA.,Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA, USA
| | - Adam M Hanif
- Center for Visual and Neurocognitive Rehabilitation, Atlanta VA Medical Center, 1670 Clairmont Road, Decatur, GA, 30033, USA
| | - Marissa A Gogniat
- Center for Visual and Neurocognitive Rehabilitation, Atlanta VA Medical Center, 1670 Clairmont Road, Decatur, GA, 30033, USA.,Department of Ophthalmology, Emory University, Atlanta, GA, USA
| | - Brian C Prall
- Center for Visual and Neurocognitive Rehabilitation, Atlanta VA Medical Center, 1670 Clairmont Road, Decatur, GA, 30033, USA.,Department of Ophthalmology, Emory University, Atlanta, GA, USA.,Neuroscience Program, Emory University, Atlanta, GA, USA
| | - Raza Haider
- Center for Visual and Neurocognitive Rehabilitation, Atlanta VA Medical Center, 1670 Clairmont Road, Decatur, GA, 30033, USA
| | - Moe H Aung
- Center for Visual and Neurocognitive Rehabilitation, Atlanta VA Medical Center, 1670 Clairmont Road, Decatur, GA, 30033, USA.,Department of Ophthalmology, Emory University, Atlanta, GA, USA.,Neuroscience Program, Emory University, Atlanta, GA, USA
| | - Megan C Prunty
- Center for Visual and Neurocognitive Rehabilitation, Atlanta VA Medical Center, 1670 Clairmont Road, Decatur, GA, 30033, USA
| | - Lukas M Mees
- Center for Visual and Neurocognitive Rehabilitation, Atlanta VA Medical Center, 1670 Clairmont Road, Decatur, GA, 30033, USA.,Department of Ophthalmology, Emory University, Atlanta, GA, USA
| | - Monica M Coulter
- Center for Visual and Neurocognitive Rehabilitation, Atlanta VA Medical Center, 1670 Clairmont Road, Decatur, GA, 30033, USA
| | - Cara T Motz
- Center for Visual and Neurocognitive Rehabilitation, Atlanta VA Medical Center, 1670 Clairmont Road, Decatur, GA, 30033, USA
| | - Jeffrey H Boatright
- Center for Visual and Neurocognitive Rehabilitation, Atlanta VA Medical Center, 1670 Clairmont Road, Decatur, GA, 30033, USA.,Department of Ophthalmology, Emory University, Atlanta, GA, USA.,Neuroscience Program, Emory University, Atlanta, GA, USA
| | - Machelle T Pardue
- Center for Visual and Neurocognitive Rehabilitation, Atlanta VA Medical Center, 1670 Clairmont Road, Decatur, GA, 30033, USA.,Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA, USA.,Neuroscience Program, Emory University, Atlanta, GA, USA
| |
Collapse
|
86
|
Wu HJ, Li XY, Qian WJ, Li Q, Wang SY, Ji M, Ma YY, Gao F, Sun XH, Wang X, Miao Y, Yang XL, Wang Z. Dopamine D1 receptor-mediated upregulation of BKCa
currents modifies Müller cell gliosis in a rat chronic ocular hypertension model. Glia 2018; 66:1507-1519. [PMID: 29508439 DOI: 10.1002/glia.23321] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2017] [Revised: 02/15/2018] [Accepted: 02/19/2018] [Indexed: 11/10/2022]
Affiliation(s)
- Hang-Jing Wu
- Department of Neurology; Institutes of Brain Science, State Key Laboratory of Medical Neurobiology, Zhongshan Hospital, Fudan University; Shanghai 200032 China
| | - Xue-Yan Li
- Department of Neurology; Institutes of Brain Science, State Key Laboratory of Medical Neurobiology, Zhongshan Hospital, Fudan University; Shanghai 200032 China
| | - Wen-Jing Qian
- Department of Neurology; Institutes of Brain Science, State Key Laboratory of Medical Neurobiology, Zhongshan Hospital, Fudan University; Shanghai 200032 China
| | - Qian Li
- Department of Neurology; Institutes of Brain Science, State Key Laboratory of Medical Neurobiology, Zhongshan Hospital, Fudan University; Shanghai 200032 China
| | - Shu-Yue Wang
- Department of Neurology; Institutes of Brain Science, State Key Laboratory of Medical Neurobiology, Zhongshan Hospital, Fudan University; Shanghai 200032 China
| | - Min Ji
- Department of Ophthalmology at Eye & ENT Hospital; Fudan University; Shanghai 200031 China
| | - Yuan-Yuan Ma
- Department of Neurology; Institutes of Brain Science, State Key Laboratory of Medical Neurobiology, Zhongshan Hospital, Fudan University; Shanghai 200032 China
| | - Feng Gao
- Department of Ophthalmology at Eye & ENT Hospital; Fudan University; Shanghai 200031 China
| | - Xing-Huai Sun
- Department of Ophthalmology at Eye & ENT Hospital; Fudan University; Shanghai 200031 China
| | - Xin Wang
- Department of Neurology; Institutes of Brain Science, State Key Laboratory of Medical Neurobiology, Zhongshan Hospital, Fudan University; Shanghai 200032 China
| | - Yanying Miao
- Department of Neurology; Institutes of Brain Science, State Key Laboratory of Medical Neurobiology, Zhongshan Hospital, Fudan University; Shanghai 200032 China
| | - Xiong-Li Yang
- Department of Neurology; Institutes of Brain Science, State Key Laboratory of Medical Neurobiology, Zhongshan Hospital, Fudan University; Shanghai 200032 China
| | - Zhongfeng Wang
- Department of Neurology; Institutes of Brain Science, State Key Laboratory of Medical Neurobiology, Zhongshan Hospital, Fudan University; Shanghai 200032 China
| |
Collapse
|
87
|
Rha J, Jones SK, Fidler J, Banerjee A, Leung SW, Morris KJ, Wong JC, Inglis GAS, Shapiro L, Deng Q, Cutler AA, Hanif AM, Pardue MT, Schaffer A, Seyfried NT, Moberg KH, Bassell GJ, Escayg A, García PS, Corbett AH. The RNA-binding protein, ZC3H14, is required for proper poly(A) tail length control, expression of synaptic proteins, and brain function in mice. Hum Mol Genet 2018; 26:3663-3681. [PMID: 28666327 DOI: 10.1093/hmg/ddx248] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2017] [Accepted: 06/20/2017] [Indexed: 12/30/2022] Open
Abstract
A number of mutations in genes that encode ubiquitously expressed RNA-binding proteins cause tissue specific disease. Many of these diseases are neurological in nature revealing critical roles for this class of proteins in the brain. We recently identified mutations in a gene that encodes a ubiquitously expressed polyadenosine RNA-binding protein, ZC3H14 (Zinc finger CysCysCysHis domain-containing protein 14), that cause a nonsyndromic, autosomal recessive form of intellectual disability. This finding reveals the molecular basis for disease and provides evidence that ZC3H14 is essential for proper brain function. To investigate the role of ZC3H14 in the mammalian brain, we generated a mouse in which the first common exon of the ZC3H14 gene, exon 13 is removed (Zc3h14Δex13/Δex13) leading to a truncated ZC3H14 protein. We report here that, as in the patients, Zc3h14 is not essential in mice. Utilizing these Zc3h14Δex13/Δex13mice, we provide the first in vivo functional characterization of ZC3H14 as a regulator of RNA poly(A) tail length. The Zc3h14Δex13/Δex13 mice show enlarged lateral ventricles in the brain as well as impaired working memory. Proteomic analysis comparing the hippocampi of Zc3h14+/+ and Zc3h14Δex13/Δex13 mice reveals dysregulation of several pathways that are important for proper brain function and thus sheds light onto which pathways are most affected by the loss of ZC3H14. Among the proteins increased in the hippocampi of Zc3h14Δex13/Δex13 mice compared to control are key synaptic proteins including CaMK2a. This newly generated mouse serves as a tool to study the function of ZC3H14 in vivo.
Collapse
Affiliation(s)
- Jennifer Rha
- Department of Biochemistry, Emory University School of Medicine, Atlanta, GA 30322, USA.,Graduate Program in Biochemistry, Cell, and Developmental Biology, Emory University, Atlanta, GA 30322, USA
| | - Stephanie K Jones
- Department of Biology.,Graduate Program in Genetics and Molecular Biology, Emory University, Atlanta, GA 30322, USA
| | - Jonathan Fidler
- Department of Anesthesiology, Emory University School of Medicine & Research Division, Atlanta VA Medical Center, Atlanta, GA 30322, USA
| | | | | | - Kevin J Morris
- Graduate Program in Biochemistry, Cell, and Developmental Biology, Emory University, Atlanta, GA 30322, USA.,Department of Biology
| | - Jennifer C Wong
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - George Andrew S Inglis
- Graduate Program in Genetics and Molecular Biology, Emory University, Atlanta, GA 30322, USA.,Department of Human Genetics, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Lindsey Shapiro
- Graduate Program in Genetics and Molecular Biology, Emory University, Atlanta, GA 30322, USA.,Graduate Program in Neuroscience, Emory University, Atlanta, GA 30322, USA
| | - Qiudong Deng
- Department of Biochemistry, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Alicia A Cutler
- Graduate Program in Biochemistry, Cell, and Developmental Biology, Emory University, Atlanta, GA 30322, USA.,Department of Pharmacology, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Adam M Hanif
- Department of Opthamology, Emory University School of Medicine & Research Division, & Atlanta VA Medical Center, Atlanta, GA 30322, USA
| | - Machelle T Pardue
- Department of Opthamology, Emory University School of Medicine & Research Division, & Atlanta VA Medical Center, Atlanta, GA 30322, USA
| | - Ashleigh Schaffer
- Department of Genetics and Genome Sciences, School of Medicine, Case Western Reserve University, Cleveland, OH 44106-4955, USA
| | - Nicholas T Seyfried
- Department of Biochemistry, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Kenneth H Moberg
- Department of Cell Biology, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Gary J Bassell
- Department of Cell Biology, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Andrew Escayg
- Graduate Program in Genetics and Molecular Biology, Emory University, Atlanta, GA 30322, USA
| | - Paul S García
- Department of Anesthesiology, Emory University School of Medicine & Research Division, Atlanta VA Medical Center, Atlanta, GA 30322, USA
| | | |
Collapse
|
88
|
Gnaz couples the circadian and dopaminergic system to G protein-mediated signaling in mouse photoreceptors. PLoS One 2017; 12:e0187411. [PMID: 29088301 PMCID: PMC5663513 DOI: 10.1371/journal.pone.0187411] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2017] [Accepted: 10/19/2017] [Indexed: 01/08/2023] Open
Abstract
The mammalian retina harbors a circadian clockwork that regulates vision and promotes healthiness of retinal neurons, mainly through directing the rhythmic release of the neurohormones dopamine—acting on dopamine D4 receptors—and melatonin—acting on MT1 and MT2 receptors. The gene Gnaz—a unique Gi/o subfamily member—was seen in the present study to be expressed in photoreceptors where its protein product Gαz shows a daily rhythm in its subcellular localization. Apart from subcellular localization, Gnaz displays a daily rhythm in expression—with peak values at night—in preparations of the whole retina, microdissected photoreceptors and photoreceptor-related pinealocytes. In retina, Gnaz rhythmicity was observed to persist under constant darkness and to be abolished in retina deficient for Clock or dopamine D4 receptors. Furthermore, circadian regulation of Gnaz was disturbed in the db/db mouse, a model of diabetic retinopathy. The data of the present study suggest that Gnaz links the circadian clockwork—via dopamine acting on D4 receptors—to G protein-mediated signaling in intact but not diabetic retina.
Collapse
|
89
|
Chesnokova NB, Pavlenko TA, Ugrumov MV. [Ophthalmic disorders as a manifestation of Parkinson's disease]. Zh Nevrol Psikhiatr Im S S Korsakova 2017; 117:124-131. [PMID: 29053133 DOI: 10.17116/jnevro201711791124-131] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Parkinson's disease is a severe neurodegenerative disease accompanied with the degeneration of dopaminergic neurons in the central and peripheral nervous system. The diagnosis of Parkinson's disease can still be made only on the stage of irreversible and nearly total degeneration of the nigrostriatum dopaminergic system and exhaustion of brain compensatory mechanisms that explains the low efficacy of therapy. Ophthalmic pathology is one of the nonmotor symptoms of Parkinson's disease. This can be explained firstly by the fact that eye is a 'peripheral part of brain' and secondly by the involvement of dopaminergic neurons (dopamine-producing cells) that are subject to the selective degeneration during Parkinson's disease in the regulation of visual function in the eye and brain. Dopaminergic neurons and dopamine receptors are present in all structures of the eye. Parkinson's disease cause abnormalities not only in the retina but in the whole optic tract and can be considered as peripheral manifestations of the disease that precede the well-known motor dysfunctions. This review describes ophthalmological symptoms of Parkinson's disease, possible pathophysiological mechanisms of their development, optical disorders in experimental models of Parkinson's disease and also the perspectives of experimental and clinical studies of visual disorders for the development of preclinical diagnosis of Parkinson's disease.
Collapse
Affiliation(s)
- N B Chesnokova
- Helmholtz Moscow Research Institute of Eye Diseases, Moscow, Russia
| | - T A Pavlenko
- Helmholtz Moscow Research Institute of Eye Diseases, Moscow, Russia
| | - M V Ugrumov
- Koltzov Institute of Developmental Biology, Russian of Sciences, Moscow, Russia
| |
Collapse
|
90
|
Neurodegeneration in diabetic retinopathy: Potential for novel therapies. Vision Res 2017; 139:82-92. [PMID: 28988945 DOI: 10.1016/j.visres.2017.06.014] [Citation(s) in RCA: 64] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2017] [Revised: 06/12/2017] [Accepted: 06/14/2017] [Indexed: 11/20/2022]
Abstract
The complex pathology of diabetic retinopathy (DR) affects both vascular and neural tissue. The characteristics of neurodegeneration are well-described in animal models but have more recently been confirmed in the clinical setting, mostly by using non-invasive imaging approaches such as spectral domain optical coherence tomography (SD-OCT). The most frequent observations report loss of tissue in the nerve fiber layer and inner plexiform layer, confirming earlier findings from animal models. In several cases the reduction in inner retinal layers is reported in patients with little evidence of vascular lesions or macular edema, suggesting that degenerative loss of neural tissue in the inner retina can occur after relatively short durations of diabetes. Animal studies also suggest that neurodegeneration leading to retinal thinning is not limited to cell death and tissue loss but also includes changes in neuronal morphology, reduced synaptic protein expression and alterations in neurotransmission, including changes in expression of neurotransmitter receptors as well as neurotransmitter release, reuptake and metabolism. The concept of neurodegeneration as an early component of DR introduces the possibility to explore alternative therapies to prevent the onset of vision loss, including neuroprotective therapies and drugs targeting individual neurotransmitter systems, as well as more general neuroprotective approaches to preserve the integrity of the neural retina. In this review we consider some of the evidence for progressive retinal neurodegeneration in diabetes, and explore potential neuroprotective therapies.
Collapse
|
91
|
Liu LL, Spix NJ, Zhang DQ. NMDA Receptors Contribute to Retrograde Synaptic Transmission from Ganglion Cell Photoreceptors to Dopaminergic Amacrine Cells. Front Cell Neurosci 2017; 11:279. [PMID: 28959188 PMCID: PMC5603656 DOI: 10.3389/fncel.2017.00279] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2017] [Accepted: 08/29/2017] [Indexed: 11/13/2022] Open
Abstract
Recently, a line of evidence has demonstrated that the vertebrate retina possesses a novel retrograde signaling pathway. In this pathway, phototransduction is initiated by the photopigment melanopsin, which is expressed in a small population of retinal ganglion cells. These ganglion cell photoreceptors then signal to dopaminergic amacrine cells (DACs) through glutamatergic synapses, influencing visual light adaptation. We have previously demonstrated that in Mg2+-containing solution, α-amino-3-hydroxyl-5-methyl-4-isoxazole-propionate (AMPA) receptors mediate this glutamatergic transmission. Here, we demonstrate that removing extracellular Mg2+ enhances melanopsin-based DAC light responses at membrane potentials more negative than −40 mV. Melanopsin-based responses in Mg2+-free solution were profoundly suppressed by the selective N-methyl-D-aspartate (NMDA) receptor antagonist D-AP5. In addition, application of NMDA to the retina produced excitatory inward currents in DACs. These data strongly suggest that DACs express functional NMDA receptors. We further found that in the presence of Mg2+, D-AP5 reduced the peak amplitude of melanopsin-based DAC responses by ~70% when the cells were held at their resting membrane potential (−50 mV), indicating that NMDA receptors are likely to contribute to retrograde signal transmission to DACs under physiological conditions. Moreover, our data show that melanopsin-based NMDA-receptor-mediated responses in DACs are suppressed by antagonists specific to either the NR2A or NR2B receptor subtype. Immunohistochemical results show that NR2A and NR2B subunits are expressed on DAC somata and processes. These results suggest that DACs express functional NMDA receptors containing both NR2A and NR2B subunits. Collectively, our data reveal that, along with AMPA receptors, NR2A- and NR2B-containing NMDA receptors mediate retrograde signal transmission from ganglion cell photoreceptors to DACs.
Collapse
Affiliation(s)
- Lei-Lei Liu
- Eye Research Institute, Oakland UniversityRochester, MI, United States
| | - Nathan J Spix
- Eye Research Institute, Oakland UniversityRochester, MI, United States
| | - Dao-Qi Zhang
- Eye Research Institute, Oakland UniversityRochester, MI, United States
| |
Collapse
|
92
|
Vanlandingham PA, Nuno DJ, Quiambao AB, Phelps E, Wassel RA, Ma JX, Farjo KM, Farjo RA. Inhibition of Stat3 by a Small Molecule Inhibitor Slows Vision Loss in a Rat Model of Diabetic Retinopathy. Invest Ophthalmol Vis Sci 2017; 58:2095-2105. [PMID: 28395025 PMCID: PMC5386345 DOI: 10.1167/iovs.16-20641] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Purpose Diabetic retinopathy is a leading cause of vision loss. Previous studies have shown signaling pathways mediated by Stat3 (signal transducer and activator of transcription 3) play a primary role in diabetic retinopathy progression. This study tested CLT-005, a small molecule inhibitor of Stat3, for its dose-dependent therapeutic effects on vision loss in a rat model of diabetic retinopathy. Methods Brown Norway rats were administered streptozotocin (STZ) to induce diabetes. CLT-005 was administered daily by oral gavage for 16 weeks at concentrations of 125, 250, or 500 mg/kg, respectively, beginning 4 days post streptozotocin administration. Systemic and ocular drug concentration was quantified with mass spectrometry. Visual function was monitored at 2-week intervals from 6 to 16 weeks using optokinetic tracking to measure visual acuity and contrast sensitivity. The presence and severity of cataracts was visually monitored and correlated to visual acuity. The transcription and translation of multiple angiogenic factors and inflammatory cytokines were measured by real-time polymerase chain reaction and Multiplex immunoassay. Results Streptozotocin-diabetic rats sustain progressive vision loss over 16 weeks, and this loss in visual function is rescued in a dose-dependent manner by CLT-005. This positive therapeutic effect correlates to the positive effects of CLT-005 on vascular leakage and the presence of inflammatory cytokines in the retina. Conclusions The present study indicates that Stat3 inhibition has strong therapeutic potential for the treatment of vision loss in diabetic retinopathy.
Collapse
Affiliation(s)
| | - Didier J Nuno
- Charlesson LLC, Oklahoma City, Oklahoma, United States
| | | | - Eric Phelps
- Charlesson LLC, Oklahoma City, Oklahoma, United States
| | - Ronald A Wassel
- Charlesson LLC, Oklahoma City, Oklahoma, United States 2EyeCRO, LLC, Oklahoma City, Oklahoma, United States
| | - Jian-Xing Ma
- Department of Physiology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, United States
| | - Krysten M Farjo
- Department of Physiology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, United States
| | - Rafal A Farjo
- Charlesson LLC, Oklahoma City, Oklahoma, United States 2EyeCRO, LLC, Oklahoma City, Oklahoma, United States
| |
Collapse
|
93
|
Lindsley CW, Hopkins CR. Return of D4 Dopamine Receptor Antagonists in Drug Discovery. J Med Chem 2017; 60:7233-7243. [DOI: 10.1021/acs.jmedchem.7b00151] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Affiliation(s)
| | - Corey R. Hopkins
- Department
of Pharmaceutical Sciences, College of Pharmacy, University of Nebraska Medical Center, Omaha, Nebraska 68198-6125, United States
| |
Collapse
|
94
|
Oxidative stress and diabetic retinopathy: development and treatment. Eye (Lond) 2017; 31:1122-1130. [PMID: 28452994 DOI: 10.1038/eye.2017.64] [Citation(s) in RCA: 141] [Impact Index Per Article: 17.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2015] [Accepted: 03/14/2017] [Indexed: 02/07/2023] Open
Abstract
Diabetic retinopathy (DR) is the most common microvascular complication in diabetic patients and one of the main causes of acquired blindness in the world. From the 90s until date, the incidence of this complication has increased. Reactive oxygen species (ROS) is a free radical with impaired electron that usually participates in the redox mechanisms of some body molecules such as enzymes, proteins, and so on. In normal biological conditions, ROS is maintained in equilibrium, however its overproduction can lead to biological process called oxidative stress and this is considered the main pathogenesis of DR. The retina is susceptible to ROS because of high-energy demands and exposure to light. When the balance is broken, ROS produces retinal cell injury by interacting with the cellular components. This article describes the possible role of oxidative stress in the development of DR and proposes some treatment options based on its stages. The review of the topic shows that blindness caused by DR can be avoided by early detection and timely treatment.
Collapse
|
95
|
Gardner TW, Davila JR. The neurovascular unit and the pathophysiologic basis of diabetic retinopathy. Graefes Arch Clin Exp Ophthalmol 2016; 255:1-6. [PMID: 27832340 DOI: 10.1007/s00417-016-3548-y] [Citation(s) in RCA: 119] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2016] [Revised: 10/25/2016] [Accepted: 10/31/2016] [Indexed: 01/17/2023] Open
Abstract
PURPOSE To relate the concept of the retinal neurovascular unit and its alterations in diabetes to the pathophysiology of diabetic retinopathy. METHODS Case illustrations and conceptual frameworks are presented that illustrate adaptive and maladaptive "dis-integration" of the retinal neurovascular unit with the progression of diabetes. RESULTS Retinopathy treatment should address pathophysiologic processes rather than pathologic lesions as is current practice. CONCLUSIONS Future improvements in the treatment of diabetic retinopathy requires deeper understanding of the cellular and molecular changes induced by diabetes, coupled with the use of quantitative phenotyping methods that assess the pathophysiologic processes.
Collapse
Affiliation(s)
- Thomas W Gardner
- Department of Ophthalmology and Visual Sciences, W.K. Kellogg Eye Center, University of Michigan Medical School, 1000 Wall St, Ann Arbor, MI, 48105, USA.
| | - Jose R Davila
- Department of Ophthalmology and Visual Sciences, W.K. Kellogg Eye Center, University of Michigan Medical School, 1000 Wall St, Ann Arbor, MI, 48105, USA
| |
Collapse
|
96
|
Lahouaoui H, Coutanson C, Cooper HM, Bennis M, Dkhissi-Benyahya O. Diabetic retinopathy alters light-induced clock gene expression and dopamine levels in the mouse retina. Mol Vis 2016; 22:959-69. [PMID: 27559292 PMCID: PMC4974849] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2016] [Accepted: 08/03/2016] [Indexed: 10/28/2022] Open
Abstract
PURPOSE Diabetic retinopathy is one of the most common consequences of diabetes that affects millions of working-age adults worldwide and leads to progressive degeneration of the retina, visual loss, and blindness. Diabetes is associated with circadian disruption of the central and peripheral circadian clocks, but the mechanisms responsible for such alterations are unknown. Using a streptozotocin (STZ)-induced model of diabetes, we investigated whether diabetes alters 1) the circadian regulation of clock genes in the retina and in the central clocks, 2) the light response of clock genes in the retina, and/or 3) light-driven retinal dopamine (DA), a major output marker of the retinal clock. METHODS To quantify circadian expression of clock and clock-controlled genes, retinas and suprachiasmatic nucleus (SCN) from the same animals were collected every 4 h in circadian conditions, 12 weeks post-diabetes. Induction of Per1, Per2, and c-fos mRNAs was quantified in the retina after the administration of a pulse of monochromatic light (480 nm, 1.17×10(14) photons/cm(2)/s, 15 min) at circadian time 16. Gene expression was assessed with real-time reverse transcription PCR (RT-PCR). Pooled retinas from the control and STZ-diabetic mice were collected 2 h after light ON and light OFF (Zeitgeber time (ZT)2 and ZT14), and DA and its metabolite were analyzed with high-performance liquid chromatography (HPLC). RESULTS We found variable effects of diabetes on the expression of clock genes in the retina and only slight differences in phase and/or amplitude in the SCN. c-fos and Per1 induction by a 480 nm light pulse was abolished in diabetic animals at 12 weeks post-induction of diabetes in comparison with the control mice, suggesting a deficit in light-induced neuronal activation of the retinal clock. Finally, we quantified a 56% reduction in the total number of tyrosine hydroxylase (TH) immunopositive cells, associated with a decrease in DA levels during the subjective day (ZT2). CONCLUSIONS These findings demonstrate that diabetes affects the molecular machinery and the light response of the retinal clock and alters the light-driven retinal DA level.
Collapse
Affiliation(s)
- Hasna Lahouaoui
- Univ Lyon, Université Claude Bernard Lyon 1, Inserm, Stem Cell and Brain Research Institute, Bron, France,Laboratory of Pharmacology, Neurobiology and Behavior, University Cadi Ayyad, Marrakech, Morocco
| | - Christine Coutanson
- Univ Lyon, Université Claude Bernard Lyon 1, Inserm, Stem Cell and Brain Research Institute, Bron, France
| | - Howard M. Cooper
- Univ Lyon, Université Claude Bernard Lyon 1, Inserm, Stem Cell and Brain Research Institute, Bron, France
| | - Mohamed Bennis
- Laboratory of Pharmacology, Neurobiology and Behavior, University Cadi Ayyad, Marrakech, Morocco
| | - Ouria Dkhissi-Benyahya
- Univ Lyon, Université Claude Bernard Lyon 1, Inserm, Stem Cell and Brain Research Institute, Bron, France
| |
Collapse
|
97
|
Moore-Dotson JM, Beckman JJ, Mazade RE, Hoon M, Bernstein AS, Romero-Aleshire MJ, Brooks HL, Eggers ED. Early Retinal Neuronal Dysfunction in Diabetic Mice: Reduced Light-Evoked Inhibition Increases Rod Pathway Signaling. Invest Ophthalmol Vis Sci 2016; 57:1418-30. [PMID: 27028063 PMCID: PMC4819579 DOI: 10.1167/iovs.15-17999] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Purpose Recent studies suggest that the neural retinal response to light is compromised in diabetes. Electroretinogram studies suggest that the dim light retinal rod pathway is especially susceptible to diabetic damage. The purpose of this study was to determine whether diabetes alters rod pathway signaling. Methods Diabetes was induced in C57BL/6J mice by three intraperitoneal injections of streptozotocin (STZ; 75 mg/kg), and confirmed by blood glucose levels > 200 mg/dL. Six weeks after the first injection, whole-cell voltage clamp recordings of spontaneous and light-evoked inhibitory postsynaptic currents from rod bipolar cells were made in dark-adapted retinal slices. Light-evoked excitatory currents from rod bipolar and AII amacrine cells, and spontaneous excitatory currents from AII amacrine cells were also measured. Receptor inputs were pharmacologically isolated. Immunohistochemistry was performed on whole mounted retinas. Results Rod bipolar cells had reduced light-evoked inhibitory input from amacrine cells but no change in excitatory input from rod photoreceptors. Reduced light-evoked inhibition, mediated by both GABAA and GABAC receptors, increased rod bipolar cell output onto AII amacrine cells. Spontaneous release of GABA onto rod bipolar cells was increased, which may limit GABA availability for light-evoked release. These physiological changes occurred in the absence of retinal cell loss or changes in GABAA receptor expression levels. Conclusions Our results indicate that early diabetes causes deficits in the rod pathway leading to decreased light-evoked rod bipolar cell inhibition and increased rod pathway output that provide a basis for the development of early diabetic visual deficits.
Collapse
Affiliation(s)
| | - Jamie J Beckman
- Graduate Interdisciplinary Program in Physiological Sciences, University of Arizona, Tucson, Arizona, United States
| | - Reece E Mazade
- Graduate Interdisciplinary Program in Physiological Sciences, University of Arizona, Tucson, Arizona, United States
| | - Mrinalini Hoon
- Department of Biological Structure, University of Washington, Seattle, Washington, United States
| | - Adam S Bernstein
- Department of Physiology, University of Arizona, Tucson, Arizona, United States
| | | | - Heddwen L Brooks
- Department of Physiology, University of Arizona, Tucson, Arizona, United States
| | - Erika D Eggers
- Department of Physiology, University of Arizona, Tucson, Arizona, United States 4Department of Biomedical Engineering, University of Arizona, Tucson, Arizona, United States
| |
Collapse
|
98
|
Guzman DC, Olguín HJ, García EH, Peraza AV, de la Cruz DZ, Soto MP. Mechanisms involved in the development of diabetic retinopathy induced by oxidative stress. Redox Rep 2016; 22:10-16. [PMID: 27420399 DOI: 10.1080/13510002.2016.1205303] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
BACKGROUND Diabetic retinopathy (DR) is one of the main complications in patients with diabetes and has been the leading cause of visual loss since 1990. Oxidative stress is a biological process resulting from excessive production of reactive oxygen species (ROS). This process contributes to the development of many diseases and disease complications. ROS interact with various cellular components to induce cell injury. Fortunately, there is an antioxidan t system that protects organisms against ROS. Indeed, when ROS exceed antioxidant capacity, the resulting cell injury can cause diverse physiological and pathological changes that could lead to a disease like DR. OBJECTIVE This paper reviews the possible mechanisms of common and novel biomarkers involved in the development of DR and explores how these biomarkers could be used to monitor the damage induced by oxidative stress in DR, which is a significant complication in people with diabetes. CONCLUSION The poor control of glucemy in pacients with DB has been shown contribute to the development of complications in eyes as DR.
Collapse
Affiliation(s)
| | - Hugo Juárez Olguín
- b Laboratory of Pharmacology , National Institute of Pediatrics , Mexico.,c Faculty of Medicine, Department of Pharmacology , National Autonomous University of Mexico , Mexico
| | | | | | - Diego Zamora de la Cruz
- c Faculty of Medicine, Department of Pharmacology , National Autonomous University of Mexico , Mexico
| | - Monica Punzo Soto
- b Laboratory of Pharmacology , National Institute of Pediatrics , Mexico
| |
Collapse
|
99
|
Ilaiwy A, Liu M, Parry TL, Bain JR, Newgard CB, Schisler JC, Muehlbauer MJ, Despa F, Willis MS. Human amylin proteotoxicity impairs protein biosynthesis, and alters major cellular signaling pathways in the heart, brain and liver of humanized diabetic rat model in vivo. Metabolomics 2016; 12:95. [PMID: 28775675 PMCID: PMC5538143 DOI: 10.1007/s11306-016-1022-9] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
INTRODUCTION Chronic hypersecretion of the 37 amino acid amylin is common in type 2 diabetics (T2D). Recent studies implicate human amylin aggregates cause proteotoxicity (cell death induced by misfolded proteins) in both the brain and the heart. OBJECTIVES Identify systemic mechanisms/markers by which human amylin associated with cardiac and brain defects might be identified. METHODS We investigated the metabolic consequences of amyloidogenic and cytotoxic amylin oligomers in heart, brain, liver, and plasma using non-targeted metabolomics analysis in a rat model expressing pancreatic human amylin (HIP model). RESULTS Four metabolites were significantly different in 3 or more of the the four compartments (heart, brain, liver, and plasma) in HIP rats. When compared to a T2D rat model, HIP hearts uniquely had significant DECREASES in five amino acids (lysine, alanine, tyrosine, phenylalanine, serine), with phenylalanine decreased across all four tissues investigated, including plasma. In contrast, significantly INCREASED circulating phenylalanine is reported in diabetics in multiple recent studies. CONCLUSION DECREASED phenylalanine may serve as a unique marker of cardiac and brain dysfunction due to hyperamylinemia that can be differentiated from alterations in T2D in the plasma. While the deficiency in phenylalanine was seen across tissues including plasma and could be monitored, reduced tyrosine was seen only in the brain. The 50% reduction in phenylalanine and tyrosine in HIP brains is significant given their role in supporting brain chemistry as a precursor for catecholamines (dopamine, norepinephrine, epinephrine), which may contribute to the increased morbidity and mortality in diabetics at a multi-system level beyond the effects on glucose metabolism.
Collapse
Affiliation(s)
| | - Miao Liu
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, KY, USA
| | - Traci L Parry
- McAllister Heart Institute, University of North Carolina, Chapel Hill, NC, USA
| | - James R Bain
- Sarah W. Stedman Nutrition and Metabolism Center, Duke Molecular Physiology Institute, Duke University Medical Center, Durham, NC, USA
| | - Christopher B Newgard
- Sarah W. Stedman Nutrition and Metabolism Center, Duke Molecular Physiology Institute, Duke University Medical Center, Durham, NC, USA
| | - Jonathan C Schisler
- Department of Pharmacology, University of North Carolina, Chapel Hill, NC, USA
| | - Michael J Muehlbauer
- Sarah W. Stedman Nutrition and Metabolism Center, Duke Molecular Physiology Institute, Duke University Medical Center, Durham, NC, USA
| | - Florin Despa
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, KY, USA
| | - Monte S Willis
- Department of Pharmacology, University of North Carolina, Chapel Hill, NC, USA
| |
Collapse
|
100
|
Abstract
Ocular clocks, first identified in the retina, are also found in the retinal pigment epithelium (RPE), cornea, and ciliary body. The retina is a complex tissue of many cell types and considerable effort has gone into determining which cell types exhibit clock properties. Current data suggest that photoreceptors as well as inner retinal neurons exhibit clock properties with photoreceptors dominating in nonmammalian vertebrates and inner retinal neurons dominating in mice. However, these differences may in part reflect the choice of circadian output, and it is likely that clock properties are widely dispersed among many retinal cell types. The phase of the retinal clock can be set directly by light. In nonmammalian vertebrates, direct light sensitivity is commonplace among body clocks, but in mice only the retina and cornea retain direct light-dependent phase regulation. This distinguishes the retina and possibly other ocular clocks from peripheral oscillators whose phase depends on the pace-making properties of the hypothalamic central brain clock, the suprachiasmatic nuclei (SCN). However, in mice, retinal circadian oscillations dampen quickly in isolation due to weak coupling of its individual cell-autonomous oscillators, and there is no evidence that retinal clocks are directly controlled through input from other oscillators. Retinal circadian regulation in both mammals and nonmammalian vertebrates uses melatonin and dopamine as dark- and light-adaptive neuromodulators, respectively, and light can regulate circadian phase indirectly through dopamine signaling. The melatonin/dopamine system appears to have evolved among nonmammalian vertebrates and retained with modification in mammals. Circadian clocks in the eye are critical for optimum visual function where they play a role fine tuning visual sensitivity, and their disruption can affect diseases such as glaucoma or retinal degeneration syndromes.
Collapse
Affiliation(s)
- Joseph C Besharse
- Department of Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, Milwaukee, WI
| | - Douglas G McMahon
- Department of Biological Sciences, Vanderbilt University, Nashville, TN
| |
Collapse
|