1
|
Liu HH, Cullen PF, Sivak JM, Gronert K, Flanagan JG. Protective Effects of Lipoxin A 4 and B 4 Signaling on the Inner Retina in a Mouse Model of Experimental Glaucoma. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.17.575414. [PMID: 38293224 PMCID: PMC10827219 DOI: 10.1101/2024.01.17.575414] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/01/2024]
Abstract
Glaucoma is a common neurodegenerative disease characterized by progressive degeneration of retinal ganglion cells (RGCs) and the retinal nerve fiber layer (RNFL), resulting in a gradual decline of vision. A recent study by our groups indicated that the levels of lipoxins A4 (LXA4) and B4 (LXB4) in the retina and optic nerve decrease following acute injury, and that restoring their function is neuroprotective. Lipoxins are members of the specialized pro-resolving mediator (SPM) family and play key roles to mitigate and resolve chronic inflammation and tissue damage. Yet, knowledge about lipoxin neuroprotective activity remains limited. Here we investigate the in vivo efficacy of exogenous LXA4 and LXB4 administration on the inner retina in a mouse model of chronic experimental glaucoma. To investigate the contribution of LXA4 signaling we used transgenic knockout (KO) mice lacking the two mouse LXA4 receptors (Fpr2/Fpr3-/-). Functional and structural changes of inner retinal neurons were assessed longitudinally using electroretinogram (ERG) and optical coherence tomography (OCT). At the end of the experiment, retinal samples were harvested for immunohistological assessment. While both lipoxins generated protective trends, only LXB4 treatment was significant, and consistently more efficacious than LXA4 in all endpoints. Both lipoxins also appeared to dramatically reduce Müller glial reactivity following injury. In comparison, Fpr2/Fpr3 deletion significantly worsened inner retinal injury and function, consistent with an essential protective role for endogenous LXA4. Together, these results support further exploration of lipoxin signaling as a treatment for glaucomatous neurodegeneration.
Collapse
Affiliation(s)
- Hsin-Hua Liu
- Herbert Wertheim School of Optometry and Vision Science, University of California at Berkeley, Berkeley, California, United States
| | - Paul F. Cullen
- Herbert Wertheim School of Optometry and Vision Science, University of California at Berkeley, Berkeley, California, United States
| | - Jeremy M. Sivak
- Department of Ophthalmology and Vision Science, University of Toronto, Toronto, Ontario, Canada
- Krembil Research Institute, University Health Network, Toronto, Ontario, Canada
| | - Karsten Gronert
- Herbert Wertheim School of Optometry and Vision Science, University of California at Berkeley, Berkeley, California, United States
| | - John G. Flanagan
- Herbert Wertheim School of Optometry and Vision Science, University of California at Berkeley, Berkeley, California, United States
| |
Collapse
|
2
|
Tsioti I, Steiner BL, Escher P, Zinkernagel MS, Benz PM, Kokona D. Systemic Lipopolysaccharide Exposure Exacerbates Choroidal Neovascularization in Mice. Ocul Immunol Inflamm 2024; 32:19-30. [PMID: 36441988 DOI: 10.1080/09273948.2022.2147547] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Accepted: 11/08/2022] [Indexed: 11/29/2022]
Abstract
This study aims to investigate the effect of a systemic lipopolysaccharide (LPS) stimulus in the course of laser-induced choroidal neovascularization (CNV) in C57BL/6 J mice. A group of CNV-subjected mice received 1 mg/kg LPS via the tail vein immediately after CNV induction. Mouse eyes were monitored in vivo with fluorescein angiography for 2 weeks. In situ hybridization and flow cytometry were performed in the retina at different time points. LPS led to increased fluorescein leakage 3 days after CNV, correlated with a large influx of monocyte-derived macrophages and increase of pro-inflammatory microglia/macrophages in the retina. Additionally, LPS enhanced Vegfα mRNA expression by Glul-expressing cells but not Aif1 positive microglia/macrophages in the laser lesion. These findings suggest that systemic LPS exposure has transient detrimental effects in the course of CNV through activation of microglia/macrophages to a pro-inflammatory phenotype and supports the important role of these cells in the CNV course.
Collapse
Affiliation(s)
- Ioanna Tsioti
- Department of Ophthalmology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
- Department of BioMedical Research, University of Bern, Bern, Switzerland
| | - Beatrice L Steiner
- Department of Ophthalmology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
- Department of BioMedical Research, University of Bern, Bern, Switzerland
| | - Pascal Escher
- Department of Ophthalmology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
- Department of BioMedical Research, University of Bern, Bern, Switzerland
| | - Martin S Zinkernagel
- Department of Ophthalmology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
- Department of BioMedical Research, University of Bern, Bern, Switzerland
| | - Peter M Benz
- Department of CardioMetabolic Diseases Research, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach, Germany
| | - Despina Kokona
- Department of Ophthalmology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
- Department of BioMedical Research, University of Bern, Bern, Switzerland
| |
Collapse
|
3
|
Mei T, Wu J, Wu K, Zhao M, Luo J, Liu X, Shang B, Xu W, Yang Z, Lai Y, Liu C, Gong H, Gao X, Zhuo Y, Lin M, Zhao L. Lipocalin 2 induces visual impairment by promoting ferroptosis in retinal ischemia-reperfusion injury. ANNALS OF TRANSLATIONAL MEDICINE 2023; 11:3. [PMID: 36760251 PMCID: PMC9906199 DOI: 10.21037/atm-22-3298] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Accepted: 11/07/2022] [Indexed: 01/07/2023]
Abstract
Background Retinal ischemia-reperfusion (RIR) is a common pathological condition that can lead to retinal ganglion cell (RGC) death and visual impairment. However, the pathogenesis of RGC loss and visual impairment caused by retinal ischemia remains unclear. Methods A mouse model of elevated intraocular pressure (IOP)-induced RIR injury was used. Flash visual evoked potentials (FVEPs) and electroretinography (ERG) recordings were performed to assess visual function. The structural integrity of the retina and the number of RGC were assessed using hematoxylin and eosin (HE) staining and retinal flat mounts. Ferroptosis was evaluated by testing the levels of glutathione (GSH), malondialdehyde (MDA), glutathione peroxidase (GPX4), and ferritin light chains (FTL) in the retina of wild-type (WT) and lipocalin-2 transgenic (LCN2-TG) mice after RIR injury. Results We found that LCN2 was mainly expressed in the RGC layer in the retina of wild-type mice and remarkably upregulated after RIR injury. Compared with wild-type mice, aggravated RGC death and visual impairment were exhibited in LCN2-TG mice with RIR injury. Moreover, LCN2 overexpression activated glial cells and upregulated proinflammatory factors. More importantly, we found that LCN2 strongly promoted ferroptosis signaling in RGC death and visual impairment. Liproxstatin-1, an inhibitor of ferroptosis, could significantly ameliorate RGC death and visual impairment. Furthermore, we found significantly alleviated RGC death and retinal damage in LCN2 heterozygous knockout mice. Conclusions Our study provides important insights linking upregulated LCN2-mediated promotion of ferroptosis to RGC death and visual function impairment in the pathogenesis of ischemic retinopathy.
Collapse
Affiliation(s)
- Tingfang Mei
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, China
| | - Jinwen Wu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, China
| | - Keling Wu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, China
| | - Minglei Zhao
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, China
| | - Jingyi Luo
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, China
| | - Xinqi Liu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, China;,Guangdong Province Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Bizhi Shang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, China
| | - Wenchang Xu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, China
| | - Zeqiu Yang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, China
| | - Yuhua Lai
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, China
| | - Chujun Liu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, China
| | - Haijun Gong
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, China;,Department of Ophthalmology, Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Xinbo Gao
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, China
| | - Yehong Zhuo
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, China
| | - Mingkai Lin
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, China
| | - Ling Zhao
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, China
| |
Collapse
|
4
|
Peng H, Han W, Ma B, Dai S, Long J, Zhou S, Li H, Chen B. Autophagy and senescence of rat retinal precursor cells under high glucose. Front Endocrinol (Lausanne) 2023; 13:1047642. [PMID: 36686430 PMCID: PMC9846177 DOI: 10.3389/fendo.2022.1047642] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/18/2022] [Accepted: 12/05/2022] [Indexed: 01/05/2023] Open
Abstract
Backgrounds Diabetic retinopathy (DR) is a common diabetic ocular disease characterized by retinal ganglion cell (RGC) changes. An abnormal environment, hyperglycemia, may progressively alter the structure and function of RGCs, which is a primary pathological feature of retinal neurodegeneration in DR. Accumulated studies confirmed autophagy and senescence play a vital role in DR; however, the underlying mechanisms need to be clarified. Methods This study included the microarray expression profiling dataset GSE60436 from Gene Expression Omnibus (GEO) to conduct the bioinformatics analysis. The R software was used to identify autophagy-related genes (ARGs) that were differentially expressed in fibrovascular membranes (FVMs) and normal retinas. Co-expression and tissue-specific expression were elicited for the filtered genes. The genes were then analyzed by ontology (GO) enrichment analysis, Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analysis and Gene Set Enrichment Analysis (GSEA). R28 cells were cultured with high glucose, detected by reverse transcription-quantitative (RT-qPCR) and stained by apoptosis kit. Results In the retina, 31 differentially expressed ARGs (24 up-regulated genes) were discovered and enriched. The enrichment results revealed that differentially expressed ARGs were significantly enriched in autophagy, apoptosis, aging, and neural function. Four hub genes (i.e., TP53, CASP1, CCL2, and CASP1) were significantly up-regulated. Upregulation of cellular autophagy and apoptosis level was detected in the hyperglycemia model in vitro. Conclusions Our results provide evidence for the autophagy and cellular senescence mechanisms involved in retinal hyperglycemia injury, and the protective function of autophagy is limited. Further study may favour understanding the disease progression and neuroprotection of DR.
Collapse
Affiliation(s)
- Hanhan Peng
- Department of Ophthalmology, Second Xiangya Hospital, Central South University, Changsha, China
- Hunan Clinical Research Centre of Ophthalmic Disease, Changsha, China
| | - Wentao Han
- Department of Ophthalmology, Second Xiangya Hospital, Central South University, Changsha, China
- Hunan Clinical Research Centre of Ophthalmic Disease, Changsha, China
| | - Benteng Ma
- Department of Ophthalmology, Second Xiangya Hospital, Central South University, Changsha, China
- Hunan Clinical Research Centre of Ophthalmic Disease, Changsha, China
| | - Shirui Dai
- Department of Ophthalmology, Second Xiangya Hospital, Central South University, Changsha, China
- Hunan Clinical Research Centre of Ophthalmic Disease, Changsha, China
| | - Jianfeng Long
- Department of Ophthalmology, Second Xiangya Hospital, Central South University, Changsha, China
- Hunan Clinical Research Centre of Ophthalmic Disease, Changsha, China
| | - Shu Zhou
- Department of Ophthalmology, Second Xiangya Hospital, Central South University, Changsha, China
- Hunan Clinical Research Centre of Ophthalmic Disease, Changsha, China
| | - Haoyu Li
- Department of Ophthalmology, Second Xiangya Hospital, Central South University, Changsha, China
- Hunan Clinical Research Centre of Ophthalmic Disease, Changsha, China
| | - Baihua Chen
- Department of Ophthalmology, Second Xiangya Hospital, Central South University, Changsha, China
- Hunan Clinical Research Centre of Ophthalmic Disease, Changsha, China
| |
Collapse
|
5
|
Xu QA, Boerkoel P, Hirsch-Reinshagen V, Mackenzie IR, Hsiung GYR, Charm G, To EF, Liu AQ, Schwab K, Jiang K, Sarunic M, Beg MF, Pham W, Cui J, To E, Lee S, Matsubara JA. Müller cell degeneration and microglial dysfunction in the Alzheimer's retina. Acta Neuropathol Commun 2022; 10:145. [PMID: 36199154 PMCID: PMC9533552 DOI: 10.1186/s40478-022-01448-y] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Accepted: 09/18/2022] [Indexed: 01/26/2023] Open
Abstract
Amyloid beta (Aβ) deposits in the retina of the Alzheimer's disease (AD) eye may provide a useful diagnostic biomarker for AD. This study focused on the relationship of Aβ with macroglia and microglia, as these glial cells are hypothesized to play important roles in homeostasis and clearance of Aβ in the AD retina. Significantly higher Aβ load was found in AD compared to controls, and specifically in the mid-peripheral region. AD retina showed significantly less immunoreactivity against glial fibrillary acidic protein (GFAP) and glutamine synthetase (GS) compared to control eyes. Immunoreactivity against ionized calcium binding adapter molecule-1 (IBA-1), a microglial marker, demonstrated a higher level of microgliosis in AD compared to control retina. Within AD retina, more IBA-1 immunoreactivity was present in the mid-peripheral retina, which contained more Aβ than the central AD retina. GFAP co-localized rarely with Aβ, while IBA-1 co-localized with Aβ in more layers of control than AD donor retina. These results suggest that dysfunction of the Müller and microglial cells may be key features of the AD retina.
Collapse
Affiliation(s)
- Qinyuan Alis Xu
- grid.17091.3e0000 0001 2288 9830MD Undergraduate Program, University of British Columbia, Vancouver, BC Canada ,grid.17063.330000 0001 2157 2938Postgraduate Medical Education, Department of Family and Community Medicine, University of Toronto, Barrie, ON Canada
| | - Pierre Boerkoel
- grid.17091.3e0000 0001 2288 9830MD Undergraduate Program, University of British Columbia, Vancouver, BC Canada
| | - Veronica Hirsch-Reinshagen
- grid.17091.3e0000 0001 2288 9830Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC Canada
| | - Ian R. Mackenzie
- grid.17091.3e0000 0001 2288 9830Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC Canada
| | - Ging-Yuek Robin Hsiung
- grid.17091.3e0000 0001 2288 9830Division of Neurology, Department of Medicine, University of British Columbia, Vancouver, BC Canada
| | - Geoffrey Charm
- grid.17091.3e0000 0001 2288 9830Department of Ophthalmology and Visual Sciences, Eye Care Centre, University of British Columbia, 2550 Willow Street, Vancouver, BC V5Z3N9 Canada
| | - Elliott F. To
- grid.17091.3e0000 0001 2288 9830Department of Ophthalmology and Visual Sciences, Eye Care Centre, University of British Columbia, 2550 Willow Street, Vancouver, BC V5Z3N9 Canada
| | - Alice Q. Liu
- grid.17091.3e0000 0001 2288 9830MD Undergraduate Program, University of British Columbia, Vancouver, BC Canada
| | - Katerina Schwab
- grid.17091.3e0000 0001 2288 9830MD Undergraduate Program, University of British Columbia, Vancouver, BC Canada
| | - Kailun Jiang
- grid.17091.3e0000 0001 2288 9830Department of Ophthalmology and Visual Sciences, Eye Care Centre, University of British Columbia, 2550 Willow Street, Vancouver, BC V5Z3N9 Canada
| | - Marinko Sarunic
- grid.61971.380000 0004 1936 7494School of Engineering Sciences, Simon Fraser University, Burnaby, BC Canada
| | - Mirza Faisal Beg
- grid.61971.380000 0004 1936 7494School of Engineering Sciences, Simon Fraser University, Burnaby, BC Canada
| | - Wellington Pham
- grid.152326.10000 0001 2264 7217Vanderbilt University Institute of Imaging Science, Vanderbilt University School of Medicine, Nashville, USA
| | - Jing Cui
- grid.17091.3e0000 0001 2288 9830Department of Ophthalmology and Visual Sciences, Eye Care Centre, University of British Columbia, 2550 Willow Street, Vancouver, BC V5Z3N9 Canada
| | - Eleanor To
- grid.17091.3e0000 0001 2288 9830Department of Ophthalmology and Visual Sciences, Eye Care Centre, University of British Columbia, 2550 Willow Street, Vancouver, BC V5Z3N9 Canada
| | - Sieun Lee
- grid.61971.380000 0004 1936 7494School of Engineering Sciences, Simon Fraser University, Burnaby, BC Canada ,grid.4563.40000 0004 1936 8868Mental Health & Clinical Neurosciences, School of Medicine, University of Nottingham, University of Nottingham, Nottingham, England
| | - Joanne A. Matsubara
- grid.17091.3e0000 0001 2288 9830Department of Ophthalmology and Visual Sciences, Eye Care Centre, University of British Columbia, 2550 Willow Street, Vancouver, BC V5Z3N9 Canada
| |
Collapse
|
6
|
Balaratnasingam C, An D, Hein M, Yu P, Yu DY. Studies of the retinal microcirculation using human donor eyes and high-resolution clinical imaging: Insights gained to guide future research in diabetic retinopathy. Prog Retin Eye Res 2022; 94:101134. [PMID: 37154065 DOI: 10.1016/j.preteyeres.2022.101134] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Revised: 09/18/2022] [Accepted: 10/03/2022] [Indexed: 11/06/2022]
Abstract
The microcirculation plays a key role in delivering oxygen to and removing metabolic wastes from energy-intensive retinal neurons. Microvascular changes are a hallmark feature of diabetic retinopathy (DR), a major cause of irreversible vision loss globally. Early investigators have performed landmark studies characterising the pathologic manifestations of DR. Previous works have collectively informed us of the clinical stages of DR and the retinal manifestations associated with devastating vision loss. Since these reports, major advancements in histologic techniques coupled with three-dimensional image processing has facilitated a deeper understanding of the structural characteristics in the healthy and diseased retinal circulation. Furthermore, breakthroughs in high-resolution retinal imaging have facilitated clinical translation of histologic knowledge to detect and monitor progression of microcirculatory disturbances with greater precision. Isolated perfusion techniques have been applied to human donor eyes to further our understanding of the cytoarchitectural characteristics of the normal human retinal circulation as well as provide novel insights into the pathophysiology of DR. Histology has been used to validate emerging in vivo retinal imaging techniques such as optical coherence tomography angiography. This report provides an overview of our research on the human retinal microcirculation in the context of the current ophthalmic literature. We commence by proposing a standardised histologic lexicon for characterising the human retinal microcirculation and subsequently discuss the pathophysiologic mechanisms underlying key manifestations of DR, with a focus on microaneurysms and retinal ischaemia. The advantages and limitations of current retinal imaging modalities as determined using histologic validation are also presented. We conclude with an overview of the implications of our research and provide a perspective on future directions in DR research.
Collapse
Affiliation(s)
- Chandrakumar Balaratnasingam
- Lions Eye Institute, Nedlands, Western Australia, Australia; Centre for Ophthalmology and Visual Science, University of Western Australia, Perth, Australia; Department of Ophthalmology, Sir Charles Gairdner Hospital, Western Australia, Australia.
| | - Dong An
- Lions Eye Institute, Nedlands, Western Australia, Australia; Centre for Ophthalmology and Visual Science, University of Western Australia, Perth, Australia
| | - Martin Hein
- Lions Eye Institute, Nedlands, Western Australia, Australia; Centre for Ophthalmology and Visual Science, University of Western Australia, Perth, Australia
| | - Paula Yu
- Lions Eye Institute, Nedlands, Western Australia, Australia; Centre for Ophthalmology and Visual Science, University of Western Australia, Perth, Australia
| | - Dao-Yi Yu
- Lions Eye Institute, Nedlands, Western Australia, Australia; Centre for Ophthalmology and Visual Science, University of Western Australia, Perth, Australia
| |
Collapse
|
7
|
Liberski S, Wichrowska M, Kocięcki J. Aflibercept versus Faricimab in the Treatment of Neovascular Age-Related Macular Degeneration and Diabetic Macular Edema: A Review. Int J Mol Sci 2022; 23:9424. [PMID: 36012690 PMCID: PMC9409486 DOI: 10.3390/ijms23169424] [Citation(s) in RCA: 37] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2022] [Revised: 08/16/2022] [Accepted: 08/17/2022] [Indexed: 12/02/2022] Open
Abstract
Diabetic macular edema (DME) and neovascular age-related macular degeneration (nAMD) are common retinal vascular diseases responsible for most blindness in the working-age and older population in developed countries. Currently, anti-VEGF agents that block VEGF family ligands, including ranibizumab, bevacizumab (off-label use), brolucizumab, and aflibercept, are the first-line treatment for nAMD and DME. However, due to the complex pathophysiological background of nAMD and DME, non-response, resistance during anti-VEGF therapy, and relapses of the disease are still observed. Moreover, frequent injections are a psychological and economic burden for patients, leading to inadequate adhesion to therapy and a higher risk of complications. Therefore, therapeutic methods are strongly needed to develop and improve, allowing for more satisfactory disease management and lower treatment burden. Currently, the Ang/Tie-2 pathway is a promising therapeutic target for retinal vascular diseases. Faricimab is the first bispecific monoclonal antibody for intravitreal use that can neutralize VEGF and Ang-2. Due to the prolonged activity, faricimab allows extending the interval between successive injections up to three or four months in nAMD and DME patients, which can be a significant benefit for patients and an alternative to implanted drug delivery systems.
Collapse
Affiliation(s)
- Sławomir Liberski
- Department of Ophthalmology, Poznan University of Medical Sciences, ul. Augustyna Szamarzewskiego 84, 61-848 Poznan, Poland
| | - Małgorzata Wichrowska
- Department of Ophthalmology, Poznan University of Medical Sciences, ul. Augustyna Szamarzewskiego 84, 61-848 Poznan, Poland
- Doctoral School, Poznan University of Medical Sciences, ul. Bukowska 70, 60-812 Poznan, Poland
| | - Jarosław Kocięcki
- Department of Ophthalmology, Poznan University of Medical Sciences, ul. Augustyna Szamarzewskiego 84, 61-848 Poznan, Poland
| |
Collapse
|
8
|
Peña JS, Vazquez M. Harnessing the Neuroprotective Behaviors of Müller Glia for Retinal Repair. FRONT BIOSCI-LANDMRK 2022; 27:169. [PMID: 35748245 PMCID: PMC9639582 DOI: 10.31083/j.fbl2706169] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Revised: 04/18/2022] [Accepted: 05/05/2022] [Indexed: 11/29/2022]
Abstract
Progressive and irreversible vision loss in mature and aging adults creates a health and economic burden, worldwide. Despite the advancements of many contemporary therapies to restore vision, few approaches have considered the innate benefits of gliosis, the endogenous processes of retinal repair that precede vision loss. Retinal gliosis is fundamentally driven by Müller glia (MG) and is characterized by three primary cellular mechanisms: hypertrophy, proliferation, and migration. In early stages of gliosis, these processes have neuroprotective potential to halt the progression of disease and encourage synaptic activity among neurons. Later stages, however, can lead to glial scarring, which is a hallmark of disease progression and blindness. As a result, the neuroprotective abilities of MG have remained incompletely explored and poorly integrated into current treatment regimens. Bioengineering studies of the intrinsic behaviors of MG hold promise to exploit glial reparative ability, while repressing neuro-disruptive MG responses. In particular, recent in vitro systems have become primary models to analyze individual gliotic processes and provide a stepping stone for in vivo strategies. This review highlights recent studies of MG gliosis seeking to harness MG neuroprotective ability for regeneration using contemporary biotechnologies. We emphasize the importance of studying gliosis as a reparative mechanism, rather than disregarding it as an unfortunate clinical prognosis in diseased retina.
Collapse
Affiliation(s)
- Juan S. Peña
- Department of Biomedical Engineering, Rutgers, The State
University of New Jersey, Piscataway (08854), New Jersey, USA
| | - Maribel Vazquez
- Department of Biomedical Engineering, Rutgers, The State
University of New Jersey, Piscataway (08854), New Jersey, USA
| |
Collapse
|
9
|
Tajima K, Akanuma SI, Ohishi Y, Yoshida Y, Bauer B, Kubo Y, Inouye M, Hosoya KI. Freshly isolated retinal capillaries to determine efflux transporter function at the inner BRB. J Control Release 2022; 343:434-442. [DOI: 10.1016/j.jconrel.2022.01.037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Revised: 01/11/2022] [Accepted: 01/22/2022] [Indexed: 11/26/2022]
|
10
|
Kobayashi Y, Watanabe S, Ong ALC, Shirai M, Yamashiro C, Ogata T, Higashijima F, Yoshimoto T, Hayano T, Asai Y, Sasai N, Kimura K. Early manifestations and differential gene expression associated with photoreceptor degeneration in Prom1-deficient retina. Dis Model Mech 2021; 14:272527. [PMID: 34664634 PMCID: PMC8628633 DOI: 10.1242/dmm.048962] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Accepted: 10/08/2021] [Indexed: 12/13/2022] Open
Abstract
Retinitis pigmentosa (RP) and macular dystrophy (MD) are characterized by gradual photoreceptor death in the retina and are often associated with genetic mutations, including those in the prominin-1 (Prom1) gene. Prom1-knockout (KO) mice recapitulate key features of these diseases including light-dependent retinal degeneration and constriction of retinal blood vessels. The mechanisms underlying such degeneration have remained unclear, however. We here analysed early events associated with retinal degeneration in Prom1-KO mice. We found that photoreceptor cell death and glial cell activation occur between 2 and 3 weeks after birth. Whereas gene expression was not affected at 2 weeks, the expression of several genes was altered at 3 weeks in the Prom1-KO retina, with the expression of that for endothelin-2 (Edn2) being markedly upregulated. Expression of Edn2 was also induced by light stimulation in Prom1-KO mice reared in the dark. Treatment with endothelin receptor antagonists attenuated photoreceptor cell death, gliosis and retinal vessel stenosis in Prom1-KO mice. Our findings thus reveal early manifestations of retinal degeneration in a model of RP/MD and suggest potential therapeutic agents for these diseases. This article has an associated First Person interview with the first author of the paper.
Collapse
Affiliation(s)
- Yuka Kobayashi
- Department of Ophthalmology, Yamaguchi University Graduate School of Medicine, 1-1-1 Minami-kogushi, Ube 755-0046, Japan
| | - Shizuka Watanabe
- Developmental Biomedical Science, Division of Biological Sciences, Nara Institute of Science and Technology, 8916-5 Takayama-cho, Ikoma 630-0192, Japan
| | - Agnes Lee Chen Ong
- Developmental Biomedical Science, Division of Biological Sciences, Nara Institute of Science and Technology, 8916-5 Takayama-cho, Ikoma 630-0192, Japan
| | - Manabu Shirai
- Omics Research Center (ORC), National Cerebral and Cardiovascular Center, 6-1 Kishibe Shinmachi, Suita, Osaka 564-8565, Japan
| | - Chiemi Yamashiro
- Department of Ophthalmology, Yamaguchi University Graduate School of Medicine, 1-1-1 Minami-kogushi, Ube 755-0046, Japan
| | - Tadahiko Ogata
- Department of Ophthalmology, Yamaguchi University Graduate School of Medicine, 1-1-1 Minami-kogushi, Ube 755-0046, Japan
| | - Fumiaki Higashijima
- Department of Ophthalmology, Yamaguchi University Graduate School of Medicine, 1-1-1 Minami-kogushi, Ube 755-0046, Japan
| | - Takuya Yoshimoto
- Department of Ophthalmology, Yamaguchi University Graduate School of Medicine, 1-1-1 Minami-kogushi, Ube 755-0046, Japan
| | - Takahide Hayano
- Department of Systems Bioinformatics, Yamaguchi University Graduate School of Medicine, 1-1-1 Minami-kogushi, Ube 755-0046, Japan
| | - Yoshiyuki Asai
- Department of Systems Bioinformatics, Yamaguchi University Graduate School of Medicine, 1-1-1 Minami-kogushi, Ube 755-0046, Japan
| | - Noriaki Sasai
- Developmental Biomedical Science, Division of Biological Sciences, Nara Institute of Science and Technology, 8916-5 Takayama-cho, Ikoma 630-0192, Japan
| | - Kazuhiro Kimura
- Department of Ophthalmology, Yamaguchi University Graduate School of Medicine, 1-1-1 Minami-kogushi, Ube 755-0046, Japan
| |
Collapse
|
11
|
Hamdani EH, Popek M, Frontczak-Baniewicz M, Utheim TP, Albrecht J, Zielińska M, Chaudhry FA. Perturbation of astroglial Slc38 glutamine transporters by NH 4 + contributes to neurophysiologic manifestations in acute liver failure. FASEB J 2021; 35:e21588. [PMID: 34169573 DOI: 10.1096/fj.202001712rr] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Revised: 03/17/2021] [Accepted: 03/25/2021] [Indexed: 02/07/2023]
Abstract
Ammonia is considered the main pathogenic toxin in hepatic encephalopathy (HE). However, the molecular mechanisms involved have been disputed. As altered glutamatergic and GABAergic neurotransmission has been reported in HE, we investigated whether four members of the solute carrier 38 (Slc38) family of amino acid transporters-involved in the replenishment of glutamate and GABA-contribute to ammonia neurotoxicity in HE. We show that ammonium ion exerts multiple actions on the Slc38 transporters: It competes with glutamine for the binding to the system N transporters Slc38a3 and Slc38a5, consequently inhibiting bidirectional astroglial glutamine transport. It also competes with H+ , Na+ , and K+ for uncoupled permeation through the same transporters, which may perturb astroglial intracellular pH, membrane potential, and K+ -buffering. Knockdown of Slc38a3 in mice results in cerebral cortical edema and disrupted neurotransmitter synthesis mimicking events contributing to HE development. Finally, in a mouse model of acute liver failure (ALF), we demonstrate the downregulation of Slc38a3 protein, impeded astroglial glutamine release, and cytotoxic edema. Altogether, we demonstrate contribution of Slc38 transporters to the ammonia-induced impairment of glutamine recycling between astrocytes and neurons, a phenomenon underlying acute ammonia neurotoxicity in the setting of ALF.
Collapse
Affiliation(s)
- El Hassan Hamdani
- Department of Molecular Medicine, University of Oslo (UiO), Oslo, Norway.,Institute of Behavioural Science, Oslo Metropolitan University, Oslo, Norway
| | - Mariusz Popek
- Neurotoxicology Department, Mossakowski Medical Research Institute PAS, Warsaw, Poland
| | | | - Tor Paaske Utheim
- Department of Medical Biochemistry, Oslo University Hospital, Oslo, Norway.,Department of Plastic and Reconstructive Surgery, Oslo University Hospital, Oslo, Norway
| | - Jan Albrecht
- Neurotoxicology Department, Mossakowski Medical Research Institute PAS, Warsaw, Poland
| | - Magdalena Zielińska
- Neurotoxicology Department, Mossakowski Medical Research Institute PAS, Warsaw, Poland
| | - Farrukh Abbas Chaudhry
- Department of Molecular Medicine, University of Oslo (UiO), Oslo, Norway.,Department of Plastic and Reconstructive Surgery, Oslo University Hospital, Oslo, Norway
| |
Collapse
|
12
|
Harnessing Astrocytes and Müller Glial Cells in the Retina for Survival and Regeneration of Retinal Ganglion Cells. Cells 2021; 10:cells10061339. [PMID: 34071545 PMCID: PMC8229010 DOI: 10.3390/cells10061339] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Revised: 05/24/2021] [Accepted: 05/26/2021] [Indexed: 01/17/2023] Open
Abstract
Astrocytes have been associated with the failure of axon regeneration in the central nervous system (CNS), as it undergoes reactive gliosis in response to damages to the CNS and functions as a chemical and physical barrier to axon regeneration. However, beneficial roles of astrocytes have been extensively studied in the spinal cord over the years, and a growing body of evidence now suggests that inducing astrocytes to become more growth-supportive can promote axon regeneration after spinal cord injury (SCI). In retina, astrocytes and Müller cells are known to undergo reactive gliosis after damage to retina and/or optic nerve and are hypothesized to be either detrimental or beneficial to survival and axon regeneration of retinal ganglion cells (RGCs). Whether they can be induced to become more growth-supportive after retinal and optic nerve injury has yet to be determined. In this review, we pinpoint the potential molecular pathways involved in the induction of growth-supportive astrocytes in the spinal cord and suggest that stimulating the activation of these pathways in the retina could represent a new therapeutic approach to promoting survival and axon regeneration of RGCs in retinal degenerative diseases.
Collapse
|
13
|
Diabetic retinal neurodegeneration as a form of diabetic retinopathy. Int Ophthalmol 2021; 41:3223-3248. [PMID: 33954860 DOI: 10.1007/s10792-021-01864-4] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Accepted: 04/08/2021] [Indexed: 12/11/2022]
Abstract
PURPOSE To review the evidence supporting diabetic retinal neurodegeneration (DRN) as a form of diabetic retinopathy. METHOD Review of literature. RESULTS DRN is recognized to be a part of retinopathy in patients with diabetes mellitus (DM), in addition to the well-established diabetic retinal vasculopathy (DRV). DRN has been noted in the early stages of DM, before the onset of clinically evident diabetic retinopathy. The occurrence of DRN has been confirmed in animal models of DM, histopathological examination of donor's eyes from diabetic individuals and assessment of neural structure and function in humans. DRN involves alterations in retinal ganglion cells, photoreceptors, amacrine cells and bipolar cells, and is thought to be driven by glutamate, oxidative stress and dysregulation of neuroprotective factors in the retina. Potential therapeutic options for DRN are under evaluation. CONCLUSIONS Literature is divided on the temporal relation between DRN and DRV, with evidence of both precedence and simultaneous occurrence. The relationship between DRN and multi-system neuropathy in DM is yet to be evaluated critically.
Collapse
|
14
|
Park YS, Kim HL, Lee SH, Zhang Y, Kim IB. Expression of the Endoplasmic Reticulum Stress Marker GRP78 in the Normal Retina and Retinal Degeneration Induced by Blue LED Stimuli in Mice. Cells 2021; 10:cells10050995. [PMID: 33922686 PMCID: PMC8145904 DOI: 10.3390/cells10050995] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Revised: 04/19/2021] [Accepted: 04/22/2021] [Indexed: 01/12/2023] Open
Abstract
Retinal degeneration is a leading cause of blindness. The unfolded protein response (UPR) is an endoplasmic reticulum (ER) stress response that affects cell survival and death and GRP78 forms a representative protective response. We aimed to determine the exact localization of GRP78 in an animal model of light-induced retinal degeneration. Dark-adapted mice were exposed to blue light-emitting diodes and retinas were obtained at 24 h and 72 h after exposure. In the normal retina, we found that GRP78 was rarely detected in the photoreceptor cells while it was expressed in the perinuclear space of the cell bodies in the inner nuclear and ganglion cell layers. After injury, the expression of GRP78 in the outer nuclear and inner plexiform layers increased in a time-dependent manner. However, an increased GRP78 expression was not observed in damaged photoreceptor cells in the outer nuclear layer. GRP78 was located in the perinuclear space and ER lumen of glial cells and the ER developed in glial cells during retinal degeneration. These findings suggest that GRP78 and the ER response are important for glial cell activation in the retina during photoreceptor degeneration.
Collapse
Affiliation(s)
- Yong Soo Park
- Department of Anatomy, College of Medicine, The Catholic University of Korea, 222 Banpo-daero, Seocho-gu, Seoul 06591, Korea; (Y.S.P.); (S.H.L.); (Y.Z.)
- Catholic Neuroscience Institute, College of Medicine, The Catholic University of Korea, 222 Banpo-daero, Seocho-gu, Seoul 06591, Korea
| | - Hong-Lim Kim
- Integrative Research Support Center, College of Medicine, The Catholic University of Korea, 222 Banpo-daero, Seocho-gu, Seoul 06591, Korea;
| | - Seung Hee Lee
- Department of Anatomy, College of Medicine, The Catholic University of Korea, 222 Banpo-daero, Seocho-gu, Seoul 06591, Korea; (Y.S.P.); (S.H.L.); (Y.Z.)
- Catholic Neuroscience Institute, College of Medicine, The Catholic University of Korea, 222 Banpo-daero, Seocho-gu, Seoul 06591, Korea
| | - Yan Zhang
- Department of Anatomy, College of Medicine, The Catholic University of Korea, 222 Banpo-daero, Seocho-gu, Seoul 06591, Korea; (Y.S.P.); (S.H.L.); (Y.Z.)
- Department of Biomedicine and Health Sciences, Graduate School, The Catholic University of Korea, 222 Banpo-daero, Seocho-gu, Seoul 06591, Korea
| | - In-Beom Kim
- Department of Anatomy, College of Medicine, The Catholic University of Korea, 222 Banpo-daero, Seocho-gu, Seoul 06591, Korea; (Y.S.P.); (S.H.L.); (Y.Z.)
- Catholic Neuroscience Institute, College of Medicine, The Catholic University of Korea, 222 Banpo-daero, Seocho-gu, Seoul 06591, Korea
- Integrative Research Support Center, College of Medicine, The Catholic University of Korea, 222 Banpo-daero, Seocho-gu, Seoul 06591, Korea;
- Department of Biomedicine and Health Sciences, Graduate School, The Catholic University of Korea, 222 Banpo-daero, Seocho-gu, Seoul 06591, Korea
- Catholic Institute for Applied Anatomy, College of Medicine, The Catholic University of Korea, 222 Banpo-daero, Seocho-gu, Seoul 06591, Korea
- Correspondence: ; Tel.: +82-2-2258-7263
| |
Collapse
|
15
|
Ortega JT, Parmar T, Golczak M, Jastrzebska B. Protective Effects of Flavonoids in Acute Models of Light-Induced Retinal Degeneration. Mol Pharmacol 2020; 99:60-77. [PMID: 33154094 DOI: 10.1124/molpharm.120.000072] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Accepted: 10/26/2020] [Indexed: 12/17/2022] Open
Abstract
Degeneration of photoreceptors caused by excessive illumination, inherited mutations, or aging is the principal pathology of blinding diseases. Pharmacological compounds that stabilize the visual receptor rhodopsin and modulate the cellular pathways triggering death of photoreceptors could avert this pathology. Interestingly, flavonoids can modulate the cellular processes, such as oxidative stress, inflammatory responses, and apoptosis, that are activated during retinal degeneration. As we found previously, flavonoids also bind directly to unliganded rod opsin, enhancing its folding, stability, and regeneration. In addition, flavonoids stimulate rhodopsin gene expression. Thus, we evaluated the effect of two main dietary flavonoids, quercetin and myricetin, in ATP-binding cassette subfamily A member 4 -/- /retinol dehydrogenase 8 -/- and wild-type BALB/c mice susceptible to light-induced photoreceptor degeneration. Using in vivo imaging, such as optical coherence tomography, scanning laser ophthalmoscopy, and histologic assessment of retinal morphology, we found that treatment with these flavonoids prior to light insult remarkably protected retina from deterioration and preserved its function. Using high-performance liquid chromatography-mass spectrometry analysis, we detected these flavonoids in the eye upon their intraperitoneal administration. The molecular events associated with the protective effect of quercetin and myricetin were related to the elevated expression of photoreceptor-specific proteins, rhodopsin and cone opsins, decreased expression of the specific inflammatory markers, and the shift of the equilibrium between cell death regulators BCL2-associated X protein (BAX) and B-cell lymphoma 2 toward an antiapoptotic profile. These results were confirmed in photoreceptor-derived 661W cells treated with either H2O2 or all-trans-retinal stressors implicated in the mechanism of retinal degeneration. Altogether, flavonoids could have significant prophylactic value for retinal degenerative diseases. SIGNIFICANCE STATEMENT: Flavonoids commonly present in food exhibit advantageous effects in blinding diseases. They bind to and stabilize unliganded rod opsin, which in excess accelerates degenerative processes in the retina. Additionally, flavonoids enhance the expression of the visual receptors, rod and cone opsins; inhibit the inflammatory reactions; and induce the expression of antiapoptotic markers in the retina, preventing the degeneration in vivo. Thus, flavonoids could have a prophylactic value for retinal degenerative diseases.
Collapse
Affiliation(s)
- Joseph T Ortega
- Department of Pharmacology, Cleveland Center for Membrane and Structural Biology, School of Medicine, Case Western Reserve University, Cleveland, Ohio
| | - Tanu Parmar
- Department of Pharmacology, Cleveland Center for Membrane and Structural Biology, School of Medicine, Case Western Reserve University, Cleveland, Ohio
| | - Marcin Golczak
- Department of Pharmacology, Cleveland Center for Membrane and Structural Biology, School of Medicine, Case Western Reserve University, Cleveland, Ohio
| | - Beata Jastrzebska
- Department of Pharmacology, Cleveland Center for Membrane and Structural Biology, School of Medicine, Case Western Reserve University, Cleveland, Ohio
| |
Collapse
|
16
|
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: 1.8] [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
|
17
|
Menon M, Mohammadi S, Davila-Velderrain J, Goods BA, Cadwell TD, Xing Y, Stemmer-Rachamimov A, Shalek AK, Love JC, Kellis M, Hafler BP. Single-cell transcriptomic atlas of the human retina identifies cell types associated with age-related macular degeneration. Nat Commun 2019; 10:4902. [PMID: 31653841 PMCID: PMC6814749 DOI: 10.1038/s41467-019-12780-8] [Citation(s) in RCA: 187] [Impact Index Per Article: 37.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2018] [Accepted: 09/27/2019] [Indexed: 12/19/2022] Open
Abstract
Genome-wide association studies (GWAS) have identified genetic variants associated with age-related macular degeneration (AMD), one of the leading causes of blindness in the elderly. However, it has been challenging to identify the cell types associated with AMD given the genetic complexity of the disease. Here we perform massively parallel single-cell RNA sequencing (scRNA-seq) of human retinas using two independent platforms, and report the first single-cell transcriptomic atlas of the human retina. Using a multi-resolution network-based analysis, we identify all major retinal cell types, and their corresponding gene expression signatures. Heterogeneity is observed within macroglia, suggesting that human retinal glia are more diverse than previously thought. Finally, GWAS-based enrichment analysis identifies glia, vascular cells, and cone photoreceptors to be associated with the risk of AMD. These data provide a detailed analysis of the human retina, and show how scRNA-seq can provide insight into cell types involved in complex, inflammatory genetic diseases.
Collapse
Affiliation(s)
- Madhvi Menon
- Broad Institute of MIT and Harvard, Cambridge, MA, 02142, USA
- Departments of Ophthalmology and Neurology, Harvard Medical School, Boston, MA, 02115, USA
- Evergrande Center for Immunologic Diseases, Harvard Medical School, Boston, MA, 02115, USA
| | - Shahin Mohammadi
- Broad Institute of MIT and Harvard, Cambridge, MA, 02142, USA
- MIT Computer Science and Artificial Intelligence Laboratory, Cambridge, MA, 02139, USA
| | - Jose Davila-Velderrain
- Broad Institute of MIT and Harvard, Cambridge, MA, 02142, USA
- MIT Computer Science and Artificial Intelligence Laboratory, Cambridge, MA, 02139, USA
| | - Brittany A Goods
- Broad Institute of MIT and Harvard, Cambridge, MA, 02142, USA
- Institute for Medical Engineering and Science and Department of Chemistry, MIT, Cambridge, MA, 02139, USA
- Koch Institute for Integrative Cancer Research, MIT, Cambridge, MA, 02142, USA
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, 02139, USA
| | - Tanina D Cadwell
- Evergrande Center for Immunologic Diseases, Harvard Medical School, Boston, MA, 02115, USA
| | - Yu Xing
- Evergrande Center for Immunologic Diseases, Harvard Medical School, Boston, MA, 02115, USA
| | | | - Alex K Shalek
- Broad Institute of MIT and Harvard, Cambridge, MA, 02142, USA
- Institute for Medical Engineering and Science and Department of Chemistry, MIT, Cambridge, MA, 02139, USA
- Koch Institute for Integrative Cancer Research, MIT, Cambridge, MA, 02142, USA
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, 02139, USA
| | - John Christopher Love
- Broad Institute of MIT and Harvard, Cambridge, MA, 02142, USA
- Koch Institute for Integrative Cancer Research, MIT, Cambridge, MA, 02142, USA
| | - Manolis Kellis
- Broad Institute of MIT and Harvard, Cambridge, MA, 02142, USA
- Evergrande Center for Immunologic Diseases, Harvard Medical School, Boston, MA, 02115, USA
- MIT Computer Science and Artificial Intelligence Laboratory, Cambridge, MA, 02139, USA
| | - Brian P Hafler
- Broad Institute of MIT and Harvard, Cambridge, MA, 02142, USA.
- Departments of Ophthalmology and Neurology, Harvard Medical School, Boston, MA, 02115, USA.
- Evergrande Center for Immunologic Diseases, Harvard Medical School, Boston, MA, 02115, USA.
- Department of Ophthalmology and Visual Science, Yale School of Medicine, New Haven, CT, 06510, USA.
| |
Collapse
|
18
|
Cellular Mechanisms of Angiogenesis in Neonatal Rat Models of Retinal Neurodegeneration. Int J Mol Sci 2019; 20:ijms20194759. [PMID: 31557901 PMCID: PMC6801463 DOI: 10.3390/ijms20194759] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2019] [Revised: 09/23/2019] [Accepted: 09/23/2019] [Indexed: 11/17/2022] Open
Abstract
Νeuronal and glial cells play an important role in the development of vasculature in the retina. In this study, we investigated whether re-vascularization occurs in retinal neurodegenerative injury models. To induce retinal injury, N-methyl-D-aspartic acid (NMDA, 200 nmol) or kainic acid (KA, 20 nmol) was injected into the vitreous chamber of the eye on postnatal day (P)7. Morphological changes in retinal neurons and vasculature were assessed on P14, P21, and P35. Prevention of vascular growth and regression of some capillaries were observed on P14 in retinas of NMDA- and KA-treated eyes. However, vascular growth and re-vascularization started on P21, and the retinal vascular network was established by P35 in retinas with neurodegenerative injuries. The re-vascularization was suppressed by a two-day treatment with KRN633, an inhibitor of VEGF receptor tyrosine kinase, on P21 and P22. Astrocytes and Müller cells expressed vascular endothelial growth factor (VEGF), and the distribution pattern of VEGF was almost the same between the control and the NMDA-induced retinal neurodegenerative injury model, except for the difference in the thickness of the inner retinal layer. During re-vascularization, angiogenic sprouts from pre-existing blood vessels were present along the network of fibronectins formed by astrocytes. These results suggest that glial cells contribute to angiogenesis in neonatal rat models of retinal neurodegeneration.
Collapse
|
19
|
Kezic JM, McMenamin PG. Systemic exposure to CpG-ODN elicits low-grade inflammation in the retina. Exp Eye Res 2019; 186:107708. [PMID: 31242444 DOI: 10.1016/j.exer.2019.107708] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2019] [Revised: 06/18/2019] [Accepted: 06/20/2019] [Indexed: 01/04/2023]
Abstract
Previous studies have reported that topical exposure to the toll-like receptor (TLR) 9 ligand CpG-ODN causes widespread ocular inflammation, including retinal microglial activation and posterior segment inflammation. Here we sought to determine the effects of systemic exposure to CpG-ODN in the retina and whether this inflammatory response was altered with Cx3cr1 deficiency or hyperglycemia. Male non-diabetic Cx3cr1+/gfp and Cx3cr1gfp/gfp littermates (normoglycemic controls) and Cx3cr1+/gfpIns2Akitaand Cx3cr1gfp/gfpIns2Akita diabetic mice were injected intraperitoneally with 40 μg CpG-ODN. Immunofluorescence staining was performed 1 week later to assess the expression of MHC Class II and glial fibrillary acidic protein (GFAP), as well as to identify morphological changes to microglia and changes in retinal macrophage cell density. Systemic exposure to CpG-ODN induced the upregulated expression of both GFAP on retinal Müller cells and MHC Class II on the retinal vasculature. Additionally, there was an increased accumulation of macrophages in the subretinal space 1 week after exposure to systemic CpG-ODN as well as characteristic morphological changes to microglia indicative of an activated phenotype. These preliminary studies demonstrate that low-grade inflammatory changes were not enhanced in Cx3cr1-deficient or diabetic mice, indicating that the inflammatory response to systemic CpG-ODN in the retina is unaltered in the context of Cx3cr1 deficiency or prolonged hyperglycemia.
Collapse
Affiliation(s)
- Jelena M Kezic
- Department of Anatomy and Developmental Biology, Monash University, Clayton, Victoria, Australia.
| | - Paul G McMenamin
- Department of Anatomy and Developmental Biology, Monash University, Clayton, Victoria, Australia
| |
Collapse
|
20
|
Mohammad HMF, Sami MM, Makary S, Toraih EA, Mohamed AO, El-Ghaiesh SH. Neuroprotective effect of levetiracetam in mouse diabetic retinopathy: Effect on glucose transporter-1 and GAP43 expression. Life Sci 2019; 232:116588. [PMID: 31226418 DOI: 10.1016/j.lfs.2019.116588] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2019] [Revised: 06/14/2019] [Accepted: 06/17/2019] [Indexed: 01/12/2023]
Abstract
AIMS Retinopathy is a neurodegenerative complication associating diabetes mellitus. Diabetic retinopathy (DR) is the primary reason of visual loss during early adulthood. DR has a complicated multifactorial pathophysiology initiated by hyperglycaemia-induced ischaemic neurodegenerative retinal changes, followed by vision-threatening consequences. The main therapeutic modalities for DR involve invasive delivery of intravitreal antiangiogenic agents as well as surgical interventions. The current work aimed to explore the potential anti-inflammatory and retinal neuroprotective effects of levetiracetam. MAIN METHODS This study was performed on alloxan-induced diabetes in mice (n: 21). After 10 weeks, a group of diabetic animals (n: 7) was treated with levetiracetam (25 mg/kg) for six weeks. Retinal tissues were dissected and paraffin-fixed for examination using (1) morphometric analysis with haematoxylin and eosin (HE), (2) immunohistochemistry (GLUT1, GFAP and GAP43), and (3) RT-PCR-detected expression of retinal inflammatory and apoptotic mediators (TNF-α, IL6, iNOS, NF-κB and Tp53). KEY FINDINGS Diabetic mice developed disorganized and debilitated retinal layers with upregulation of the gliosis marker GFAP and downregulation of the neuronal plasticity marker GAP43. Additionally, diabetic retinae showed increased transcription of NF-κB, TNF-α, IL6, iNOS and Tp53. Levetiracetam-treated mice showed downregulation of retinal GLUT1 with relief and regression of retinal inflammation and improved retinal structural organization. SIGNIFICANCE Levetiracetam may represent a potential neuroprotective agent in DR. The data presented herein supported an anti-inflammatory role of levetiracetam. However, further clinical studies may be warranted to confirm the effectiveness and safety of levetiracetam in DR patients.
Collapse
Affiliation(s)
- Hala M F Mohammad
- Department of Clinical Pharmacology, Faculty of Medicine, Suez Canal University, Ismailia, Egypt; Central Lab., Center of Excellence in Molecular and Cellular Medicine (CEMCM), Faculty of Medicine, Suez Canal University, Ismailia, Egypt
| | - Manal M Sami
- Department of Pathology, Faculty of Medicine, Suez Canal University, Ismailia, Egypt
| | - Samy Makary
- Department of Medical Physiology, Faculty of Medicine, Suez Canal University, Ismailia, Egypt
| | - Eman A Toraih
- Genetics Unit, Department of Histology and Cell Biology, Faculty of Medicine, Suez Canal University, Ismailia 41522, Egypt; Molecular Lab, Center of Excellence of Molecular and Cellular Medicine, Suez Canal University, Ismailia, Egypt
| | - Amany O Mohamed
- Department of Medical Biochemistry, Faculty of Medicine, Assiut University, Assiut, Egypt
| | - Sabah H El-Ghaiesh
- Department of Pharmacology, Faculty of Medicine, Tanta University, Tanta, Egypt; Department of Pharmacology, Faculty of Medicine, University of Tabuk, Tabuk, Saudi Arabia.
| |
Collapse
|
21
|
Yang H, Liu C, Fan H, Chen B, Huang D, Zhang L, Zhang Q, An J, Zhao J, Wang Y, Hao D. Sonic Hedgehog Effectively Improves Oct4-Mediated Reprogramming of Astrocytes into Neural Stem Cells. Mol Ther 2019; 27:1467-1482. [PMID: 31153826 DOI: 10.1016/j.ymthe.2019.05.006] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2018] [Revised: 05/01/2019] [Accepted: 05/04/2019] [Indexed: 01/19/2023] Open
Abstract
Irreversible neuron loss following spinal cord injury (SCI) usually results in persistent neurological dysfunction. The generation of autologous neural stem cells (NSCs) holds great potential for neural replenishment therapies and drug screening in SCI. Our recent studies demonstrated that mature astrocytes from the spinal cord can directly revert back to a pluripotent state under appropriate signals. However, in previous attempts, the reprogramming of astrocytes into induced NSCs (iNSCs) was unstable, inefficient, and frequently accompanied by generation of intermediate precursors. It remained unknown how to further increase the efficiency of astrocyte reprogramming into iNSCs. Here, we show that mature astrocytes could be directly converted into iNSCs by a single transcription factor, Oct4, and that the iNSCs displayed typical neurosphere morphology, authentic NSC gene expression, self-renewal capacity, and multipotency. Strikingly, Oct4-driven reprogramming of astrocytes into iNSCs was potentiated with continuous sonic hedgehog (Shh) stimulation, as demonstrated by a sped-up reprogramming and increased conversion efficiency. Moreover, the iNSC-derived neurons possessed functionality as neurons. Importantly, crosstalk between Sox2/Shh-targeted downstream signals and phosphatidylinositol 3-kinase/cyclin-dependent kinase 2/Smad ubiquitin regulatory factor 2 (PI3K/Cdk2/Smurf2) signaling is likely involved in the mechanisms underlying this cellular event. The highly efficient reprogramming of astrocytes to generate iNSCs will provide an alternative therapeutic approach for SCI using autologous cells.
Collapse
Affiliation(s)
- Hao Yang
- Translational Medicine Center, Hong Hui Hospital, Xi'an Jiaotong University, Shaanxi 710054, China.
| | - Cuicui Liu
- Translational Medicine Center, Hong Hui Hospital, Xi'an Jiaotong University, Shaanxi 710054, China
| | - Hong Fan
- Translational Medicine Center, Hong Hui Hospital, Xi'an Jiaotong University, Shaanxi 710054, China
| | - Bo Chen
- Translational Medicine Center, Hong Hui Hospital, Xi'an Jiaotong University, Shaanxi 710054, China
| | - Dageng Huang
- Department of Spine Surgery, Hong Hui Hospital, Xi'an Jiaotong University, Shaanxi 710054, China
| | - Lingling Zhang
- Translational Medicine Center, Hong Hui Hospital, Xi'an Jiaotong University, Shaanxi 710054, China
| | - Qian Zhang
- Translational Medicine Center, Hong Hui Hospital, Xi'an Jiaotong University, Shaanxi 710054, China
| | - Jing An
- Translational Medicine Center, Hong Hui Hospital, Xi'an Jiaotong University, Shaanxi 710054, China
| | - Jingjing Zhao
- Translational Medicine Center, Hong Hui Hospital, Xi'an Jiaotong University, Shaanxi 710054, China
| | - Yi Wang
- Translational Medicine Center, Hong Hui Hospital, Xi'an Jiaotong University, Shaanxi 710054, China
| | - Dingjun Hao
- Translational Medicine Center, Hong Hui Hospital, Xi'an Jiaotong University, Shaanxi 710054, China; Department of Spine Surgery, Hong Hui Hospital, Xi'an Jiaotong University, Shaanxi 710054, China.
| |
Collapse
|
22
|
Chiquita S, Rodrigues-Neves AC, Baptista FI, Carecho R, Moreira PI, Castelo-Branco M, Ambrósio AF. The Retina as a Window or Mirror of the Brain Changes Detected in Alzheimer's Disease: Critical Aspects to Unravel. Mol Neurobiol 2019; 56:5416-5435. [PMID: 30612332 DOI: 10.1007/s12035-018-1461-6] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2018] [Accepted: 12/17/2018] [Indexed: 11/24/2022]
Abstract
Alzheimer's disease is the most frequent cause of dementia worldwide, representing a global health challenge, with a massive impact on the quality of life of Alzheimer's disease patients and their relatives. The diagnosis of Alzheimer's disease constitutes a real challenge, because the symptoms manifest years after the first degenerative changes occurring in the brain and the diagnosis is based on invasive and/or expensive techniques. Therefore, there is an urgent need to identify new reliable biomarkers to detect Alzheimer's disease at an early stage. Taking into account the evidence for visual deficits in Alzheimer's disease patients, sometimes even before the appearance of the first disease symptoms, and that the retina is an extension of the brain, the concept of the retina as a window to look into the brain or a mirror of the brain has received increasing interest in recent years. However, only a few studies have assessed the changes occurring in the retina and the brain at the same time points. Unlike previous reviews on this subject, which are mainly focused on brain changes, we organized this review by comprehensively summarizing findings related with structural, functional, cellular, and molecular parameters in the retina reported in both Alzheimer's disease patients and animal models. Moreover, we separated the studies that assessed only the retina, and those that assessed both the retina and brain, which are few but allow establishing correlations between the retina and brain. This review also highlights some inconsistent results in the literature as well as relevant missing gaps in this field.
Collapse
Affiliation(s)
- Samuel Chiquita
- iCBR, Coimbra Institute for Clinical and Biomedical Research, Faculty of Medicine, University of Coimbra, Coimbra, Portugal
- CNC.IBILI Consortium, University of Coimbra, Coimbra, Portugal
| | - Ana C Rodrigues-Neves
- iCBR, Coimbra Institute for Clinical and Biomedical Research, Faculty of Medicine, University of Coimbra, Coimbra, Portugal
- CNC.IBILI Consortium, University of Coimbra, Coimbra, Portugal
| | - Filipa I Baptista
- iCBR, Coimbra Institute for Clinical and Biomedical Research, Faculty of Medicine, University of Coimbra, Coimbra, Portugal
- CNC.IBILI Consortium, University of Coimbra, Coimbra, Portugal
| | - Rafael Carecho
- iCBR, Coimbra Institute for Clinical and Biomedical Research, Faculty of Medicine, University of Coimbra, Coimbra, Portugal
- CNC.IBILI Consortium, University of Coimbra, Coimbra, Portugal
| | - Paula I Moreira
- CNC.IBILI Consortium, University of Coimbra, Coimbra, Portugal
- CNC, Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
- Institute of Physiology, Faculty of Medicine, University of Coimbra, Coimbra, Portugal
| | - Miguel Castelo-Branco
- CNC.IBILI Consortium, University of Coimbra, Coimbra, Portugal
- CIBIT, Coimbra Institute for Biomedical Imaging and Translational Research, ICNAS, Institute of Nuclear Sciences Applied to Health, University of Coimbra, Coimbra, Portugal
| | - António F Ambrósio
- iCBR, Coimbra Institute for Clinical and Biomedical Research, Faculty of Medicine, University of Coimbra, Coimbra, Portugal.
- CNC.IBILI Consortium, University of Coimbra, Coimbra, Portugal.
| |
Collapse
|
23
|
Mat Nor N, Guo CX, Rupenthal ID, Chen YS, Green CR, Acosta ML. Sustained Connexin43 Mimetic Peptide Release From Loaded Nanoparticles Reduces Retinal and Choroidal Photodamage. Invest Ophthalmol Vis Sci 2018; 59:3682-3693. [PMID: 30029255 DOI: 10.1167/iovs.17-22829] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Purpose To evaluate the long-term effect on inflammation and inflammasome activation of intravitreally delivered connexin43 mimetic peptide (Cx43MP) in saline or incorporated within nanoparticles (NPs) for the treatment of the light-damaged rat eye. Methods Light-induced damage to the retina was created by exposure of adult albino Sprague-Dawley rats to intense light for 24 hours. A single dose of Cx43MP, Cx43MP-NPs, or saline was injected intravitreally at 2 hours after onset of light damage. Fluorescein isothiocyanate (FITC)-labelled Cx43MP-NPs were intravitreally injected to confirm delivery into the retina. Electroretinogram (ERG) recordings were performed at 24 hours, 1 week, and 2 weeks post cessation of light damage. The retinal and choroidal layers were analyzed in vivo using optical coherence tomography (OCT) and immunohistochemistry was performed on harvested tissues using glial fibrillary acidic protein (GFAP), leukocyte common antigen (CD45), and Cx43 antibodies. Results FITC was visualized 30 minutes after injection in the ganglion cell layer and in the choroid. Cx43MP and Cx43MP-NP treatments improved a-wave and b-wave function of the ERG compared with saline-injected eyes at 1 week and 2 weeks post treatment, and prevented photoreceptor loss by 2 weeks post treatment. Inflammation was also reduced and this was in parallel with downregulation of Cx43 expression. Conclusions The slow release of Cx43MP incorporated into NPs is more effective at treating retinal injury than a single dose of native Cx43MP in solution by reducing inflammation and maintaining both retinal structure and function. This NP preparation has clinical relevance as it reduces possible ocular complications associated with repeated intravitreal injections.
Collapse
Affiliation(s)
- Nasir Mat Nor
- School of Optometry and Vision Science, University of Auckland, Auckland, New Zealand.,Faculty of Medicine, University of Sultan Zainal Abidin, Kuala Terengganu, Malaysia.,New Zealand National Eye Centre, University of Auckland, Auckland, New Zealand
| | - Cindy X Guo
- School of Optometry and Vision Science, University of Auckland, Auckland, New Zealand.,New Zealand National Eye Centre, University of Auckland, Auckland, New Zealand
| | - Ilva D Rupenthal
- New Zealand National Eye Centre, University of Auckland, Auckland, New Zealand.,Department of Ophthalmology, University of Auckland, Auckland, New Zealand.,Buchanan Ocular Therapeutics Unit, University of Auckland, Auckland, New Zealand
| | - Ying-Shan Chen
- New Zealand National Eye Centre, University of Auckland, Auckland, New Zealand.,Department of Ophthalmology, University of Auckland, Auckland, New Zealand.,Buchanan Ocular Therapeutics Unit, University of Auckland, Auckland, New Zealand
| | - Colin R Green
- New Zealand National Eye Centre, University of Auckland, Auckland, New Zealand.,Department of Ophthalmology, University of Auckland, Auckland, New Zealand
| | - Monica L Acosta
- School of Optometry and Vision Science, University of Auckland, Auckland, New Zealand.,New Zealand National Eye Centre, University of Auckland, Auckland, New Zealand
| |
Collapse
|
24
|
Yoo S, Blackshaw S. Regulation and function of neurogenesis in the adult mammalian hypothalamus. Prog Neurobiol 2018; 170:53-66. [PMID: 29631023 DOI: 10.1016/j.pneurobio.2018.04.001] [Citation(s) in RCA: 89] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2017] [Revised: 02/20/2018] [Accepted: 04/05/2018] [Indexed: 12/11/2022]
Abstract
Over the past two decades, evidence has accumulated that neurogenesis can occur in both the juvenile and adult mammalian hypothalamus. Levels of hypothalamic neurogenesis can be regulated by dietary, environmental and hormonal signals. Since the hypothalamus has a central role in controlling a broad range of homeostatic physiological processes, these findings may have far ranging behavioral and medical implications. However, many questions in the field remain unresolved, including the cells of origin of newborn hypothalamic neurons and the extent to which these cells actually regulate hypothalamic-controlled behaviors. In this manuscript, we conduct a critical review of the literature on postnatal hypothalamic neurogenesis in mammals, lay out the main outstanding controversies in the field, and discuss how best to advance our knowledge of this fascinating but still poorly understood process.
Collapse
Affiliation(s)
- Sooyeon Yoo
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Seth Blackshaw
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA; Department of Ophthalmology, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA; Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA; Center for Human Systems Biology, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA; Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA.
| |
Collapse
|
25
|
Rübsam A, Parikh S, Fort PE. Role of Inflammation in Diabetic Retinopathy. Int J Mol Sci 2018; 19:ijms19040942. [PMID: 29565290 PMCID: PMC5979417 DOI: 10.3390/ijms19040942] [Citation(s) in RCA: 470] [Impact Index Per Article: 78.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2018] [Revised: 03/09/2018] [Accepted: 03/17/2018] [Indexed: 02/07/2023] Open
Abstract
Diabetic retinopathy is a common complication of diabetes and remains the leading cause of blindness among the working-age population. For decades, diabetic retinopathy was considered only a microvascular complication, but the retinal microvasculature is intimately associated with and governed by neurons and glia, which are affected even prior to clinically detectable vascular lesions. While progress has been made to improve the vascular alterations, there is still no treatment to counteract the early neuro-glial perturbations in diabetic retinopathy. Diabetes is a complex metabolic disorder, characterized by chronic hyperglycemia along with dyslipidemia, hypoinsulinemia and hypertension. Increasing evidence points to inflammation as one key player in diabetes-associated retinal perturbations, however, the exact underlying molecular mechanisms are not yet fully understood. Interlinked molecular pathways, such as oxidative stress, formation of advanced glycation end-products and increased expression of vascular endothelial growth factor have received a lot of attention as they all contribute to the inflammatory response. In the current review, we focus on the involvement of inflammation in the pathophysiology of diabetic retinopathy with special emphasis on the functional relationships between glial cells and neurons. Finally, we summarize recent advances using novel targets to inhibit inflammation in diabetic retinopathy.
Collapse
Affiliation(s)
- Anne Rübsam
- Department of Ophthalmology and Visual Sciences, University of Michigan, Ann Arbor, MI 48105, USA.
| | - Sonia Parikh
- Department of Ophthalmology and Visual Sciences, University of Michigan, Ann Arbor, MI 48105, USA.
| | - Patrice E Fort
- Department of Ophthalmology and Visual Sciences, University of Michigan, Ann Arbor, MI 48105, USA.
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI 48105, USA.
| |
Collapse
|
26
|
Oda S, Tsuneoka Y, Yoshida S, Adachi-Akahane S, Ito M, Kuroda M, Funato H. Immunolocalization of muscarinic M1 receptor in the rat medial prefrontal cortex. J Comp Neurol 2018; 526:1329-1350. [PMID: 29424434 PMCID: PMC5900831 DOI: 10.1002/cne.24409] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2017] [Revised: 01/23/2018] [Accepted: 01/27/2018] [Indexed: 12/20/2022]
Abstract
The medial prefrontal cortex (mPFC) has been considered to participate in many higher cognitive functions, such as memory formation and spatial navigation. These cognitive functions are modulated by cholinergic afferents via muscarinic acetylcholine receptors. Previous pharmacological studies have strongly suggested that the M1 receptor (M1R) is the most important subtype among muscarinic receptors to perform these cognitive functions. Actually, M1R is abundant in mPFC. However, the proportion of somata containing M1R among cortical cellular types, and the precise intracellular localization of M1R remain unclear. In this study, to clarify the precise immunolocalization of M1R in rat mPFC, we examined three major cellular types, pyramidal neurons, inhibitory neurons, and astrocytes. M1R immunopositivity signals were found in the majority of the somata of both pyramidal neurons and inhibitory neurons. In pyramidal neurons, strong M1R immunopositivity signals were usually found throughout their somata and dendrites including spines. On the other hand, the signal strength of M1R immunopositivity in the somata of inhibitory neurons significantly varied. Some neurons showed strong signals. Whereas about 40% of GAD67‐immunopositive neurons and 30% of parvalbumin‐immunopositive neurons (PV neurons) showed only weak signals. In PV neurons, M1R immunopositivity signals were preferentially distributed in somata. Furthermore, we found that many astrocytes showed substantial M1R immunopositivity signals. These signals were also mainly distributed in their somata. Thus, the distribution pattern of M1R markedly differs between cellular types. This difference might underlie the cholinergic modulation of higher cognitive functions subserved by mPFC.
Collapse
Affiliation(s)
- Satoko Oda
- Department of Anatomy, Faculty of Medicine, Toho University, Tokyo, 143-8540, Japan
| | - Yousuke Tsuneoka
- Department of Anatomy, Faculty of Medicine, Toho University, Tokyo, 143-8540, Japan
| | - Sachine Yoshida
- Department of Anatomy, Faculty of Medicine, Toho University, Tokyo, 143-8540, Japan.,Precursory Research for Embryonic Science and Technology (PRESTO), Japan Science and Technology Agency, Saitama, 332-0012, Japan
| | - Satomi Adachi-Akahane
- Department of Physiology, Faculty of Medicine, Toho University, Tokyo, 143-8540, Japan
| | - Masanori Ito
- Department of Physiology, Faculty of Medicine, Toho University, Tokyo, 143-8540, Japan
| | - Masaru Kuroda
- Department of Anatomy, Faculty of Medicine, Toho University, Tokyo, 143-8540, Japan
| | - Hiromasa Funato
- Department of Anatomy, Faculty of Medicine, Toho University, Tokyo, 143-8540, Japan.,International institute for integrative sleep medicine (IIIS), Tsukuba University, Ibaraki, 305-8575, Japan
| |
Collapse
|
27
|
Hachana S, Bhat M, Sénécal J, Huppé-Gourgues F, Couture R, Vaucher E. Expression, distribution and function of kinin B 1 receptor in the rat diabetic retina. Br J Pharmacol 2018; 175:968-983. [PMID: 29285756 DOI: 10.1111/bph.14138] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2017] [Revised: 11/22/2017] [Accepted: 12/15/2017] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND AND PURPOSE The kinin B1 receptor contributes to vascular inflammation and blood-retinal barrier breakdown in diabetic retinopathy (DR). We investigated the changes in expression, cellular localization and vascular inflammatory effect of B1 receptors in retina of streptozotocin diabetic rats. EXPERIMENTAL APPROACH The distribution of B1 receptors on retinal cell types was investigated by immunocytochemistry. Effects of B1 receptor agonist, R-838, and antagonist, R-954, on retinal leukocyte adhesion, gene expression of kinin and VEGF systems, B1 receptor immunoreactivity, microgliosis and capillary leakage were measured. Effect of B1 receptor siRNA on gene expression was also assessed. KEY RESULTS mRNA levels of the kinin and VEGF systems were significantly enhanced at 2 weeks in streptozotocin (STZ)-retina compared to control-retina and were further increased at 6 weeks. B1 receptor mRNA levels remained increased at 6 months. B1 receptor immunolabelling was detected in vascular layers of the retina, on glial and ganglion cells. Intravitreal R-838 amplified B1 and B2 receptor gene expression, B1 receptor levels (immunodetection), leukostasis and vascular permeability at 2 weeks in STZ-retina. Topical application (eye drops) of R-954 reversed these increases in B1 receptors, leukostasis and vascular permeability. Intravitreal B1 receptor siRNA inhibited gene expression of kinin and VEGF systems in STZ-retina. Microgliosis was unaffected by R-838 or R-954 in STZ-retina. CONCLUSION AND IMPLICATIONS Our results support the detrimental role of B1 receptors on endothelial and glial cells in acute and advanced phases of DR. Topical application of the B1 receptor antagonist R-954 seems a feasible therapeutic approach for the treatment of DR.
Collapse
Affiliation(s)
- Soumaya Hachana
- École d'optométrie, Université de Montréal, Montréal, QC, Canada.,Département de Pharmacologie et Physiologie, Université de Montréal, Montréal, QC, Canada
| | - Menakshi Bhat
- École d'optométrie, Université de Montréal, Montréal, QC, Canada.,Département de Pharmacologie et Physiologie, Université de Montréal, Montréal, QC, Canada
| | - Jacques Sénécal
- Département de Pharmacologie et Physiologie, Université de Montréal, Montréal, QC, Canada
| | | | - Réjean Couture
- Département de Pharmacologie et Physiologie, Université de Montréal, Montréal, QC, Canada
| | - Elvire Vaucher
- École d'optométrie, Université de Montréal, Montréal, QC, Canada
| |
Collapse
|
28
|
Stifter J, Ulbrich F, Goebel U, Böhringer D, Lagrèze WA, Biermann J. Neuroprotection and neuroregeneration of retinal ganglion cells after intravitreal carbon monoxide release. PLoS One 2017; 12:e0188444. [PMID: 29176876 PMCID: PMC5703485 DOI: 10.1371/journal.pone.0188444] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2017] [Accepted: 11/07/2017] [Indexed: 01/10/2023] Open
Abstract
PURPOSE Retinal ischemia induces apoptosis leading to neurodegeneration and vision impairment. Carbon monoxide (CO) in gaseous form showed cell-protective and anti-inflammatory effects after retinal ischemia-reperfusion-injury (IRI). These effects were also demonstrated for the intravenously administered CO-releasing molecule (CORM) ALF-186. This article summarizes the results of intravitreally released CO to assess its suitability as a neuroprotective and neuroregenerative agent. METHODS Water-soluble CORM ALF-186 (25 μg), PBS, or inactivated ALF (iALF) (all 5 μl) were intravitreally applied into the left eyes of rats directly after retinal IRI for 1 h. Their right eyes remained unaffected and were used for comparison. Retinal tissue was harvested 24 h after intervention to analyze mRNA or protein expression of Caspase-3, pERK1/2, p38, HSP70/90, NF-kappaB, AIF-1 (allograft inflammatory factor), TNF-α, and GAP-43. Densities of fluorogold-prelabeled retinal ganglion cells (RGC) were examined in flat-mounted retinae seven days after IRI and were expressed as mean/mm2. The ability of RGC to regenerate their axon was evaluated two and seven days after IRI using retinal explants in laminin-1-coated cultures. Immunohistochemistry was used to analyze the different cell types growing out of the retinal explants. RESULTS Compared to the RGC-density in the contralateral right eyes (2804±214 RGC/mm2; data are mean±SD), IRI+PBS injection resulted in a remarkable loss of RGC (1554±159 RGC/mm2), p<0.001. Intravitreally injected ALF-186 immediately after IRI provided RGC protection and reduced the extent of RGC-damage (IRI+PBS 1554±159 vs. IRI+ALF 2179±286, p<0.001). ALF-186 increased the IRI-mediated phosphorylation of MAP-kinase p38. Anti-apoptotic and anti-inflammatory effects were detectable as Caspase-3, NF-kappaB, TNF-α, and AIF-1 expression were significantly reduced after IRI+ALF in comparison to IRI+PBS or IRI+iALF. Gap-43 expression was significantly increased after IRI+ALF. iALF showed effects similar to PBS. The intrinsic regenerative potential of RGC-axons was induced to nearly identical levels after IRI and ALF or iALF-treatment under growth-permissive conditions, although RGC viability differed significantly in both groups. Intravitreal CO further increased the IRI-induced migration of GFAP-positive cells out of retinal explants and their transdifferentiation, which was detected by re-expression of beta-III tubulin and nestin. CONCLUSION Intravitreal CORM ALF-186 protected RGC after IRI and stimulated their axons to regenerate in vitro. ALF conveyed anti-apoptotic, anti-inflammatory, and growth-associated signaling after IRI. CO's role in neuroregeneration and its effect on retinal glial cells needs further investigation.
Collapse
Affiliation(s)
- Julia Stifter
- Eye Center, Medical Center—University of Freiburg, Killianstrasse 5, Freiburg, Germany
- Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Felix Ulbrich
- Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Department of Anesthesiology and Intensive Care, Medical Center—University of Freiburg, Hugstetter Strasse 55, Freiburg, Germany
| | - Ulrich Goebel
- Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Department of Anesthesiology and Intensive Care, Medical Center—University of Freiburg, Hugstetter Strasse 55, Freiburg, Germany
| | - Daniel Böhringer
- Eye Center, Medical Center—University of Freiburg, Killianstrasse 5, Freiburg, Germany
- Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Wolf Alexander Lagrèze
- Eye Center, Medical Center—University of Freiburg, Killianstrasse 5, Freiburg, Germany
- Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Julia Biermann
- Eye Center, Medical Center—University of Freiburg, Killianstrasse 5, Freiburg, Germany
- Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Department of Ophthalmology, University of Muenster Medical Center, Domagkstrasse 15, Muenster, Germany
| |
Collapse
|
29
|
Someya E, Mori A, Sakamoto K, Ishii K, Nakahara T. Stimulation of μ-opioid receptors dilates retinal arterioles by neuronal nitric oxide synthase-derived nitric oxide in rats. Eur J Pharmacol 2017; 803:124-129. [DOI: 10.1016/j.ejphar.2017.03.043] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2016] [Revised: 03/09/2017] [Accepted: 03/20/2017] [Indexed: 01/31/2023]
|
30
|
Acharya NK, Qi X, Goldwaser EL, Godsey GA, Wu H, Kosciuk MC, Freeman TA, Macphee CH, Wilensky RL, Venkataraman V, Nagele RG. Retinal pathology is associated with increased blood-retina barrier permeability in a diabetic and hypercholesterolaemic pig model: Beneficial effects of the LpPLA 2 inhibitor Darapladib. Diab Vasc Dis Res 2017; 14:200-213. [PMID: 28301218 DOI: 10.1177/1479164116683149] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Using a porcine model of diabetes mellitus and hypercholesterolaemia, we previously showed that diabetes mellitus and hypercholesterolaemia is associated with a chronic increase in blood-brain barrier permeability in the cerebral cortex, leading to selective binding of immunoglobulin G and deposition of amyloid-beta1-42 peptide in pyramidal neurons. Treatment with Darapladib (GlaxoSmithKline, SB480848), an inhibitor of lipoprotein-associated phospholipase-A2, alleviated these effects. Here, investigation of the effects of chronic diabetes mellitus and hypercholesterolaemia on the pig retina revealed a corresponding increased permeability of the blood-retina barrier coupled with a leak of plasma components into the retina, alterations in retinal architecture, selective IgG binding to neurons in the ganglion cell layer, thinning of retinal layers due to cell loss and increased glial fibrillary acidic protein expression in Müller cells, all of which were curtailed by treatment with Darapladib. These findings suggest that chronic diabetes mellitus and hypercholesterolaemia induces increased blood-retina barrier permeability that may be linked to altered expression of blood-retina barrier-associated tight junction proteins, claudin and occludin, leading to structural changes in the retina consistent with diabetic retinopathy. Additionally, results suggest that drugs with vascular anti-inflammatory properties, such as Darapladib, may have beneficial effects on eye diseases strongly linked to vascular abnormalities such as diabetic retinopathy and age-related macular degeneration.
Collapse
Affiliation(s)
- Nimish K Acharya
- 1 Biomarker Discovery Center, New Jersey Institute for Successful Aging, School of Osteopathic Medicine, Rowan University, Stratford, NJ, USA
- 2 Department of Geriatrics and Gerontology, School of Osteopathic Medicine, Rowan University, Stratford, NJ, USA
- 3 Department of Neurosurgery, University of Pennsylvania, Philadelphia, PA, USA
| | - Xin Qi
- 4 Thomas Jefferson University, Philadelphia, PA, USA
- 5 Department of Ophthalmology, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Eric L Goldwaser
- 1 Biomarker Discovery Center, New Jersey Institute for Successful Aging, School of Osteopathic Medicine, Rowan University, Stratford, NJ, USA
- 2 Department of Geriatrics and Gerontology, School of Osteopathic Medicine, Rowan University, Stratford, NJ, USA
- 6 Graduate School of Biomedical Sciences, Rowan University, Stratford, NJ, USA
| | - George A Godsey
- 2 Department of Geriatrics and Gerontology, School of Osteopathic Medicine, Rowan University, Stratford, NJ, USA
- 6 Graduate School of Biomedical Sciences, Rowan University, Stratford, NJ, USA
| | - Hao Wu
- 6 Graduate School of Biomedical Sciences, Rowan University, Stratford, NJ, USA
- 7 Department of Cell Biology, School of Osteopathic Medicine, Rowan University, Stratford, NJ, USA
| | - Mary C Kosciuk
- 1 Biomarker Discovery Center, New Jersey Institute for Successful Aging, School of Osteopathic Medicine, Rowan University, Stratford, NJ, USA
- 2 Department of Geriatrics and Gerontology, School of Osteopathic Medicine, Rowan University, Stratford, NJ, USA
| | - Theresa A Freeman
- 8 Department of Orthopaedic Surgery, Department of Dermatology and Cutaneous Biology, Sidney Kimmel Medical College, Thomas Jefferson University
| | | | - Robert L Wilensky
- 10 Hospital of the University of Pennsylvania, Philadelphia, PA, USA
| | - Venkat Venkataraman
- 7 Department of Cell Biology, School of Osteopathic Medicine, Rowan University, Stratford, NJ, USA
| | - Robert G Nagele
- 1 Biomarker Discovery Center, New Jersey Institute for Successful Aging, School of Osteopathic Medicine, Rowan University, Stratford, NJ, USA
- 2 Department of Geriatrics and Gerontology, School of Osteopathic Medicine, Rowan University, Stratford, NJ, USA
| |
Collapse
|
31
|
Dharmarajan S, Fisk DL, Sorenson CM, Sheibani N, Belecky-Adams TL. Microglia activation is essential for BMP7-mediated retinal reactive gliosis. J Neuroinflammation 2017; 14:76. [PMID: 28381236 PMCID: PMC5382432 DOI: 10.1186/s12974-017-0855-0] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2016] [Accepted: 03/27/2017] [Indexed: 02/08/2023] Open
Abstract
Background Our previous studies have shown that BMP7 is able to trigger activation of retinal macroglia. However, these studies showed the responsiveness of Müller glial cells and retinal astrocytes in vitro was attenuated in comparison to those in vivo, indicating other retinal cell types may be mediating the response of the macroglial cells to BMP7. In this study, we test the hypothesis that BMP7-mediated gliosis is the result of inflammatory signaling from retinal microglia. Methods Adult mice were injected intravitreally with BMP7 and eyes harvested 1, 3, or 7 days postinjection. Some mice were treated with PLX5622 (PLX) to ablate microglia and were subsequently injected with control or BMP7. Processed tissue was analyzed via immunofluorescence, RT-qPCR, or ELISA. In addition, cultures of retinal microglia were treated with vehicle, lipopolysaccharide, or BMP7 to determine the effects of BMP7-isolated cells. Results Mice injected with BMP7 showed regulation of various inflammatory markers at the RNA level, as well as changes in microglial morphology. Isolated retinal microglia also showed an upregulation of BMP-signaling components following treatment. In vitro treatment of retinal astrocytes with conditioned media from activated microglia upregulated RNA levels of gliosis markers. In the absence of microglia, the mouse retina showed a subdued gliosis and inflammatory response when exposed to BMP7. Conclusions Gliosis resulting from BMP7 is mediated through an inflammatory response from retinal microglia. Electronic supplementary material The online version of this article (doi:10.1186/s12974-017-0855-0) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Subramanian Dharmarajan
- Department of Biology, Indiana University-Purdue University Indianapolis, 723 W Michigan St, SL306, Indianapolis, IN, 46202, USA.,Center for Developmental and Regenerative Biology, Indiana University-Purdue University Indianapolis, 723 W Michigan St, Indianapolis, IN, 46202, USA
| | - Debra L Fisk
- Department of Ophthalmology and Visual Sciences, University of Wisconsin School of Medicine and Public Health, 1111 Highland Avenue, 9453 WIMR, Madison, WI, 53705, USA
| | - Christine M Sorenson
- Department of Pediatrics, University of Wisconsin School of Medicine and Public Health, 1111 Highland Avenue, 9453 WIMR, Madison, WI, 53705, USA
| | - Nader Sheibani
- Department of Ophthalmology and Visual Sciences, University of Wisconsin School of Medicine and Public Health, 1111 Highland Avenue, 9453 WIMR, Madison, WI, 53705, USA
| | - Teri L Belecky-Adams
- Department of Biology, Indiana University-Purdue University Indianapolis, 723 W Michigan St, SL306, Indianapolis, IN, 46202, USA. .,Center for Developmental and Regenerative Biology, Indiana University-Purdue University Indianapolis, 723 W Michigan St, Indianapolis, IN, 46202, USA.
| |
Collapse
|
32
|
Zalis MC, Johansson S, Englund-Johansson U. Immunocytochemical Profiling of Cultured Mouse Primary Retinal Cells. J Histochem Cytochem 2017; 65:223-239. [PMID: 28151698 PMCID: PMC5407564 DOI: 10.1369/0022155416689675] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
Primary retinal cell cultures and immunocytochemistry are important experimental platforms in ophthalmic research. Translation of retinal cells from their native environment to the in vitro milieu leads to cellular stress, jeopardizing their in vivo phenotype features. Moreover, the specificity and stability of many retinal immunochemical markers are poorly evaluated in retinal cell cultures. Hence, we here evaluated the expression profile of 17 retinal markers, that is, recoverin, rhodopsin, arrestin, Chx10, PKC, DCX, CRALBP, GS, vimentin, TPRV4, RBPMS, Brn3a, β-tubulin III, NeuN, MAP2, GFAP, and synaptophysin. At 7 and 18 days of culture, the marker expression profiles of mouse postnatal retinal cells were compared with their age-matched in vivo retinas. We demonstrate stable in vitro expression of all markers, except for arrestin and CRALBP. Differences in cellular expression and location of some markers were observed, both over time in culture and compared with the age-matched retina. We hypothesize that these differences are likely culture condition dependent. Taken together, we suggest a thorough evaluation of the antibodies in specific culture settings, before extrapolating the in vitro results to an in vivo setting. Moreover, the identification of specific cell types may require a combination of different genes expressed or markers with structural information.
Collapse
Affiliation(s)
- Marina C Zalis
- Division of Ophthalmology, Department of Clinical Sciences in Lund, Lund University, Lund, Sweden (MCZ, SJ, UEJ)
| | - Sebastian Johansson
- Division of Ophthalmology, Department of Clinical Sciences in Lund, Lund University, Lund, Sweden (MCZ, SJ, UEJ)
| | - Ulrica Englund-Johansson
- Division of Ophthalmology, Department of Clinical Sciences in Lund, Lund University, Lund, Sweden (MCZ, SJ, UEJ)
| |
Collapse
|
33
|
Jindal N, Banik A, Prabhakar S, Vaiphie K, Anand A. Alteration of Neurotrophic Factors After Transplantation of Bone Marrow Derived Lin-ve Stem Cell in NMDA-Induced Mouse Model of Retinal Degeneration. J Cell Biochem 2017; 118:1699-1711. [PMID: 27935095 DOI: 10.1002/jcb.25827] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2016] [Accepted: 12/06/2016] [Indexed: 02/02/2023]
Abstract
Retinal ganglion cell layer (RGCs) is one of the important layers of retina, depleted in Glaucoma. Loss of RGC neurons is a major cellular mechanism involved in its pathogenesis resulting in severe vision loss. Stem cell therapy has emerged as a potential strategy to arrest the apoptotic loss of RGCs and also replace the degenerative cells in damaged retina. Here, we have investigated the incorporation and survival of mouse bone marrow derived Lin-ve stem cells in N-methyl-d-aspartate (NMDA)-induced mouse model of retinal degeneration. Two days after intravitreal injection of NMDA (100 mM) showed significant decrease in ganglion cell number and increase in TUNEL positive apoptotic cells in retinal layers. The injury was further characterized by immunohistochemical expression of Brn3b, GFAP, Bcl2, pCREB, CNTF, GDNF, and BDNF in retinal layers. Lin-ve cells (100,000 dose) were intravitreally transplanted after 2 days of injury and evaluated after 7, 14, and 21 days of transplantation. Transplanted cells were found to have migrated from intravitreal space and incorporated into injured retina at 7, 14, and 21 days post-transplantation. At 21 days Brn3b, CNTF, and BDNF expression was found to be upregulated whereas GDNF was downregulated when compared to respective injury time points. Molecular data showed decrease in the expression of Brn3b, BDNF, CNTF, and GDNF post transplantation when compared with injury groups. This study reveals that Lin-ve stem cells may exert neuroprotective effect in damaged retina mediated by participation of neurotrophic factors induced by stem cell transplantation at the site of injury. J. Cell. Biochem. 118: 1699-1711, 2017. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Neeru Jindal
- Neuroscience Research Lab, Department of Neurology, PGIMER, Chandigarh 160012, India
| | - Avijit Banik
- Neuroscience Research Lab, Department of Neurology, PGIMER, Chandigarh 160012, India
| | - Sudesh Prabhakar
- Neuroscience Research Lab, Department of Neurology, PGIMER, Chandigarh 160012, India
| | - Kim Vaiphie
- Department of Histopathology, PGIMER, Chandigarh 160012, India
| | - Akshay Anand
- Neuroscience Research Lab, Department of Neurology, PGIMER, Chandigarh 160012, India
| |
Collapse
|
34
|
Mosley M, Shah C, Morse KA, Miloro SA, Holmes MM, Ahern TH, Forger NG. Patterns of cell death in the perinatal mouse forebrain. J Comp Neurol 2017; 525:47-64. [PMID: 27199256 PMCID: PMC5116296 DOI: 10.1002/cne.24041] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2016] [Revised: 05/16/2016] [Accepted: 05/17/2016] [Indexed: 12/15/2022]
Abstract
The importance of cell death in brain development has long been appreciated, but many basic questions remain, such as what initiates or terminates the cell death period. One obstacle has been the lack of quantitative data defining exactly when cell death occurs. We recently created a "cell death atlas," using the detection of activated caspase-3 (AC3) to quantify apoptosis in the postnatal mouse ventral forebrain and hypothalamus, and found that the highest rates of cell death were seen at the earliest postnatal ages in most regions. Here we have extended these analyses to prenatal ages and additional brain regions. We quantified cell death in 16 forebrain regions across nine perinatal ages from embryonic day (E) 17 to postnatal day (P) 11 and found that cell death peaks just after birth in most regions. We found greater cell death in several regions in offspring delivered vaginally on the day of parturition compared with those of the same postconception age but still in utero at the time of collection. We also found massive cell death in the oriens layer of the hippocampus on P1 and in regions surrounding the anterior crossing of the corpus callosum on E18 as well as the persistence of large numbers of cells in those regions in adult mice lacking the pro-death Bax gene. Together these findings suggest that birth may be an important trigger of neuronal cell death and identify transient cell groups that may undergo wholesale elimination perinatally. J. Comp. Neurol. 525:47-64, 2017. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Morgan Mosley
- Neuroscience Institute, Georgia State University, Atlanta, Georgia, 30302
| | - Charisma Shah
- Neuroscience Institute, Georgia State University, Atlanta, Georgia, 30302
| | - Kiriana A Morse
- Department of Psychology, Center for Behavioral Neuroscience, Quinnipiac University, Hamden, Connecticut, 06518
| | - Stephen A Miloro
- Department of Psychology, Center for Behavioral Neuroscience, Quinnipiac University, Hamden, Connecticut, 06518
| | - Melissa M Holmes
- Department of Psychology, University of Toronto Mississauga, Mississauga, Ontario, L5L 1C6, Canada
| | - Todd H Ahern
- Department of Psychology, Center for Behavioral Neuroscience, Quinnipiac University, Hamden, Connecticut, 06518
| | - Nancy G Forger
- Neuroscience Institute, Georgia State University, Atlanta, Georgia, 30302
| |
Collapse
|
35
|
Yoo JY, Hwang CH, Hong HN. A Model of Glial Scarring Analogous to the Environment of a Traumatically Injured Spinal Cord Using Kainate. Ann Rehabil Med 2016; 40:757-768. [PMID: 27847705 PMCID: PMC5108702 DOI: 10.5535/arm.2016.40.5.757] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2015] [Accepted: 03/14/2016] [Indexed: 11/05/2022] Open
Abstract
OBJECTIVE To develop an in vitro model analogous to the environment of traumatic spinal cord injury (SCI), the authors evaluated change of astrogliosis following treatments with kainate and/or scratch, and degree of neurite outgrowth after treatment with a kainate inhibitor. METHODS Astrocytes were obtained from the rat spinal cord. Then, 99% of the cells were confirmed to be GFAP-positive astrocytes. For chemical injury, the cells were treated with kainate at different concentrations (10, 50 or 100 µM). For mechanical injury, two kinds of uniform scratches were made using a plastic pipette tip by removing strips of cells. For combined injury (S/K), scratch and kainate were provided. Cord neurons from rat embryos were plated onto culture plates immediately after the three kinds of injuries and some cultures were treated with a kainate inhibitor. RESULTS Astro-gliosis (glial fibrillary acidic protein [GFAP], vimentin, chondroitin sulfate proteoglycan [CSPG], rho-associated protein kinase [ROCK], and ephrin type-A receptor 4 [EphA4]) was most prominent after treatment with 50 µM kainate and extensive scratch injury in terms of single arm (p<0.001) and in the S/K-induced injury model in view of single or combination (p<0.001). Neurite outgrowth in the seeded spinal cord (β-III tubulin) was the least in the S/K-induced injury model (p<0.001) and this inhibition was reversed by the kainate inhibitor (p<0.001). CONCLUSION The current in vitro model combining scratch and kainate induced glial scarring and inhibitory molecules and restricted neurite outgrowth very strongly than either the mechanically or chemically-induced injury model; hence, it may be a useful tool for research on SCI.
Collapse
Affiliation(s)
- Jong Yoon Yoo
- Department of Rehabilitation Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Chang Ho Hwang
- Department of Physical Medicine and Rehabilitation, Ulsan University Hospital, University of Ulsan College of Medicine, Ulsan, Korea
| | - Hea Nam Hong
- Department of Anatomy, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| |
Collapse
|
36
|
Kim CR, Kim JH, Park HYL, Park CK. Ischemia Reperfusion Injury Triggers TNFα Induced-Necroptosis in Rat Retina. Curr Eye Res 2016; 42:771-779. [PMID: 27732109 DOI: 10.1080/02713683.2016.1227449] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
PURPOSE A recent study revealed a novel form of cell death, termed necroptosis, or programmed necrosis. Previous research indicated that after ischemia-reperfusion (IR) injury to the retina, Tumor Necrosis Factor α (TNFα) is increased, which may activate necroptosis. This study observed macroglial cell activation, and in particular, astrocyte activation, after the release of TNFα and other necroptosis factors in the rat retina due to IR. MATERIALS AND METHODS IR was induced in the retinas of adult male Sprague-Dawley rats by increasing the intraocular pressure to 160 mmHg and then allowing reperfusion. In addition, to inhibit necroptosis, Nec-1 (necrostatin-1) was injected intravitreally after IR. Rats were sacrificed after reperfusion at 12 hours, 1, 3, and 5 days, and 1 and 2 weeks. Retinas from each time point were analyzed by immunohistochemistry (IHC) and Western blotting (WB) to identify the initiator of necroptosis, TNFα, the expression of necroptosis factors, such as receptor interacting protein (RIP) 1, 3, and inactive caspase 8, and Brn3a. RESULTS Cell death in the IR-injured retinas was identified by cell counting. We found decreased retinal cell numbers in the inner and outer nuclear layers (INL and ONL), as well as in the ganglion cell layer (GCL). Increased glial cell activation was detected by using glial fibrillary acidic protein (GFAP) IHC. TNFα, RIP1, RIP3, and inactive caspase 8 were mainly expressed in the GCL after IR, as determined by IHC and WB. Nec-1 inhibited RIP1, a necroptosis factor, indicating protection against retinal cell loss after IR injury. CONCLUSIONS We showed that IR injury triggered increases in both activation of astrocytes and the expression of TNFα. In addition, TNFα, which was activated by IR, triggered the release of necroptosis factors, particularly, in GCL. Inhibition of necroptosis using Nec-1 decreased the level of RIP1 and retinal cell loss in IR-injured retinas.
Collapse
Affiliation(s)
- Cho Rong Kim
- a Department of Ophthalmology and Visual Science , Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea , Seoul , Korea
| | - Jie Hyun Kim
- a Department of Ophthalmology and Visual Science , Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea , Seoul , Korea
| | - Hae-Young Lopilly Park
- a Department of Ophthalmology and Visual Science , Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea , Seoul , Korea
| | - Chan Kee Park
- a Department of Ophthalmology and Visual Science , Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea , Seoul , Korea
| |
Collapse
|
37
|
Livne-Bar I, Lam S, Chan D, Guo X, Askar I, Nahirnyj A, Flanagan JG, Sivak JM. Pharmacologic inhibition of reactive gliosis blocks TNF-α-mediated neuronal apoptosis. Cell Death Dis 2016; 7:e2386. [PMID: 27685630 PMCID: PMC5059876 DOI: 10.1038/cddis.2016.277] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2015] [Revised: 07/25/2016] [Accepted: 07/29/2016] [Indexed: 01/03/2023]
Abstract
Reactive gliosis is an early pathological feature common to most neurodegenerative diseases, yet its regulation and impact remain poorly understood. Normally astrocytes maintain a critical homeostatic balance. After stress or injury they undergo rapid parainflammatory activation, characterized by hypertrophy, and increased polymerization of type III intermediate filaments (IFs), particularly glial fibrillary acidic protein and vimentin. However, the consequences of IF dynamics in the adult CNS remains unclear, and no pharmacologic tools have been available to target this mechanism in vivo. The mammalian retina is an accessible model to study the regulation of astrocyte stress responses, and their influence on retinal neuronal homeostasis. In particular, our work and others have implicated p38 mitogen-activated protein kinase (MAPK) signaling as a key regulator of glutamate recycling, antioxidant activity and cytokine secretion by astrocytes and related Müller glia, with potent influences on neighboring neurons. Here we report experiments with the small molecule inhibitor, withaferin A (WFA), to specifically block type III IF dynamics in vivo. WFA was administered in a model of metabolic retinal injury induced by kainic acid, and in combination with a recent model of debridement-induced astrocyte reactivity. We show that WFA specifically targets IFs and reduces astrocyte and Müller glial reactivity in vivo. Inhibition of glial IF polymerization blocked p38 MAPK-dependent secretion of TNF-α, resulting in markedly reduced neuronal apoptosis. To our knowledge this is the first study to demonstrate that pharmacologic inhibition of IF dynamics in reactive glia protects neurons in vivo.
Collapse
Affiliation(s)
- Izhar Livne-Bar
- Department of Vision Sciences, Krembil Research Institute, University Health Network, Toronto, Ontario, Canada.,School of Optometry, University of California at Berkeley, Berkeley, CA, USA
| | - Susy Lam
- Department of Vision Sciences, Krembil Research Institute, University Health Network, Toronto, Ontario, Canada
| | - Darren Chan
- Department of Vision Sciences, Krembil Research Institute, University Health Network, Toronto, Ontario, Canada
| | - Xiaoxin Guo
- Department of Vision Sciences, Krembil Research Institute, University Health Network, Toronto, Ontario, Canada
| | - Idil Askar
- Department of Vision Sciences, Krembil Research Institute, University Health Network, Toronto, Ontario, Canada
| | - Adrian Nahirnyj
- Department of Vision Sciences, Krembil Research Institute, University Health Network, Toronto, Ontario, Canada
| | - John G Flanagan
- School of Optometry, University of California at Berkeley, Berkeley, CA, USA
| | - Jeremy M Sivak
- Department of Vision Sciences, Krembil Research Institute, University Health Network, Toronto, Ontario, Canada.,Department of Ophthalmology and Vision Science, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
38
|
Sorrentino FS, Allkabes M, Salsini G, Bonifazzi C, Perri P. The importance of glial cells in the homeostasis of the retinal microenvironment and their pivotal role in the course of diabetic retinopathy. Life Sci 2016; 162:54-9. [PMID: 27497914 DOI: 10.1016/j.lfs.2016.08.001] [Citation(s) in RCA: 84] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2016] [Revised: 07/24/2016] [Accepted: 08/02/2016] [Indexed: 01/18/2023]
Abstract
Diabetic retinopathy (DR) is a remarkable microvascular complication of diabetes and it has been considered the leading cause of legal blindness in working-age adults in the world. Several overlapping and interrelated molecular pathways are involved in the development of this disease. DR is staged into different levels of severity, from the nonproliferative to the advanced proliferative form. Over the years the progression of DR evolves through a series of changes involving distinct types of specialized cells: neural, vascular and glial. Prior to the clinically observable vascular complications, hyperglycemia and inflammation affect retinal glial cells which undergo a wide range of structural and functional alterations. In this review, we provide an overview of the status of macroglia and microglia in the course of DR, trying to briefly take into account the complex biochemical mechanisms that affect the intimate relationship among neuroretina, vessels and glial cells.
Collapse
Affiliation(s)
| | - Michael Allkabes
- Department of Biomedical and Surgical Sciences, Division of Ophthalmology, University of Ferrara, Ferrara, Italy
| | - Giulia Salsini
- Department of Biomedical and Surgical Sciences, Division of Ophthalmology, University of Ferrara, Ferrara, Italy
| | - Claudio Bonifazzi
- Department of Biomedical and Surgical Sciences, Section of Human Physiology, University of Ferrara, Ferrara, Italy
| | - Paolo Perri
- Department of Biomedical and Surgical Sciences, Division of Ophthalmology, University of Ferrara, Ferrara, Italy
| |
Collapse
|
39
|
Chang SW, Kim HI, Kim GH, Park SJ, Kim IB. Increased Expression of Osteopontin in Retinal Degeneration Induced by Blue Light-Emitting Diode Exposure in Mice. Front Mol Neurosci 2016; 9:58. [PMID: 27504084 PMCID: PMC4958628 DOI: 10.3389/fnmol.2016.00058] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2016] [Accepted: 07/07/2016] [Indexed: 02/01/2023] Open
Abstract
Osteopontin (OPN) is a multifunctional adhesive glycoprotein that is implicated in a variety of pro-inflammatory as well as neuroprotective and repair-promoting effects in the brain. As a first step towards understanding the role of OPN in retinal degeneration (RD), we examined changes in OPN expression in a mouse model of RD induced by exposure to a blue light-emitting diode (LED). RD was induced in BALB/c mice by exposure to a blue LED (460 nm) for 2 h. Apoptotic cell death was evaluated by terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL) assay. In order to investigate changes in OPN in RD, western blotting and immunohistochemistry were performed. Anti-OPN labeling was compared to that of anti-glial fibrillary acidic protein (GFAP), which is a commonly used marker for retinal injury or stress including inflammation. OPN expression in RD retinas markedly increased at 24 h after exposure, was sustained through 72 h, and subsided at 120 h. Increased OPN expression was observed co-localized with microglial cells in the outer nuclear layer (ONL), outer plexiform layer (OPL), and subretinal space. Expression was restricted to the central retina in which photoreceptor cell death occurred. Interestingly, OPN expression in the ONL/OPL was closely associated with microglia, whereas most of the OPN plaques observed in the subretinal space were not. Immunogold electron microscopy demonstrated that OPN was distributed throughout the cytoplasm of microglia and in nearby fragments of degenerating photoreceptors. In addition, we found that OPN was induced more acutely and with greater region specificity than GFAP. These results indicate that OPN may be a more useful marker for retinal injury or stress, and furthermore act as a microglial pro-inflammatory mediator and a phagocytosis-inducing opsonin in the subretinal space. Taken together, our data suggest that OPN plays an important role in the pathogenesis of RD.
Collapse
Affiliation(s)
- Seung Wook Chang
- Department of Anatomy, College of Medicine, The Catholic University of Korea Seoul, Korea
| | - Hyung Il Kim
- Department of Anatomy, College of Medicine, The Catholic University of KoreaSeoul, Korea; Gyeongju St. Mary's Eye ClinicGyeongju, Korea
| | - Gyu Hyun Kim
- Department of Anatomy, College of Medicine, The Catholic University of KoreaSeoul, Korea; Catholic Neuroscience Institute, College of Medicine, The Catholic University of KoreaSeoul, Korea
| | - Su Jin Park
- Department of Anatomy, College of Medicine, The Catholic University of KoreaSeoul, Korea; Catholic Neuroscience Institute, College of Medicine, The Catholic University of KoreaSeoul, Korea
| | - In-Beom Kim
- Department of Anatomy, College of Medicine, The Catholic University of KoreaSeoul, Korea; Catholic Neuroscience Institute, College of Medicine, The Catholic University of KoreaSeoul, Korea; Catholic Institute for Applied Anatomy, College of Medicine, The Catholic University of KoreaSeoul, Korea
| |
Collapse
|
40
|
Gene expression changes in the retina following subretinal injection of human neural progenitor cells into a rodent model for retinal degeneration. Mol Vis 2016; 22:472-90. [PMID: 27217715 PMCID: PMC4872275] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2016] [Accepted: 05/13/2016] [Indexed: 12/03/2022] Open
Abstract
PURPOSE Retinal degenerative diseases (RDDs) affect millions of people and are the leading cause of vision loss. Although treatment options for RDDs are limited, stem and progenitor cell-based therapies have great potential to halt or slow the progression of vision loss. Our previous studies have shown that a single subretinal injection of human forebrain derived neural progenitor cells (hNPCs) into the Royal College of Surgeons (RCS) retinal degenerate rat offers long-term preservation of photoreceptors and visual function. Furthermore, neural progenitor cells are currently in clinical trials for treating age-related macular degeneration; however, the molecular mechanisms of stem cell-based therapies are largely unknown. This is the first study to analyze gene expression changes in the retina of RCS rats following subretinal injection of hNPCs using high-throughput sequencing. METHODS RNA-seq data of retinas from RCS rats injected with hNPCs (RCS(hNPCs)) were compared to sham surgery in RCS (RCS(sham)) and wild-type Long Evans (LE(sham)) rats. Differential gene expression patterns were determined with in silico analysis and confirmed with qRT-PCR. Function, biologic, cellular component, and pathway analyses were performed on differentially expressed genes and investigated with immunofluorescent staining experiments. RESULTS Analysis of the gene expression data sets identified 1,215 genes that were differentially expressed between RCS(sham) and LE(sham) samples. Additionally, 283 genes were differentially expressed between the RCS(hNPCs) and RCS(sham) samples. Comparison of these two gene sets identified 68 genes with inverse expression (termed rescue genes), including Pdc, Rp1, and Cdc42ep5. Functional, biologic, and cellular component analyses indicate that the immune response is enhanced in RCS(sham). Pathway analysis of the differential expression gene sets identified three affected pathways in RCS(hNPCs), which all play roles in phagocytosis signaling. Immunofluorescent staining detected the increased presence of macrophages and microglia in RCS(sham) retinas, which decreased in RCS(hNPCs) retinas similar to the patterns detected in LE(sham). CONCLUSIONS The results from this study provide evidence of the gene expression changes that occur following treatment with hNPCs in the degenerating retina. This information can be used in future studies to potentially enhance or predict responses to hNPC and other stem cell therapies for retinal degenerative diseases.
Collapse
|
41
|
Calado SM, Diaz-Corrales F, Silva GA. pEPito-driven PEDF Expression Ameliorates Diabetic Retinopathy Hallmarks. Hum Gene Ther Methods 2016; 27:79-86. [PMID: 26942449 DOI: 10.1089/hgtb.2015.169] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Diabetic retinopathy (DR) is one of the major complications of diabetes mellitus. It is characterized by retinal microvascular changes caused by chronic exposure to hyperglycemia, leading to low tissue oxygenation and ultimately to neovascularization. Laser photocoagulation and vitrectomy are the most efficient treatments for DR, but display severe side effects such as the destruction of the healthy retina. Another clinical approach uses antiangiogenic agents to prevent and delay progression of neovascularization, but these require recurrent local administrations that increase the possibility of retinal detachment, vitreous hemorrhage, and cataract formation. Studies in human diabetic retinas have revealed an imbalance between proangiogenic factors such as the vascular endothelial growth factor (VEGF) and antiangiogenic factors, such as pigment epithelial-derived factor (PEDF). This imbalance favors pathological angiogenesis contributing to DR, and can constitute a therapeutic target. Gene therapy was recently shown to be an adequate intervention for long-term treatment of several retinal pathologies. We have previously shown the newly engineered episomal vector pEPito to be able of sustained gene expression in the mouse retina. We here show that pEPito was able to overexpress PEDF for up to three months, both in in vitro cultures of human retinal pigment epithelial cells and in the retina of diabetic mice after a single subretinal injection. In vivo, in parallel with the increase in PEDF we observed a decrease in VEGF levels in injected compared with noninjected eyes and a significant effect on two hallmarks of DR: reduction of glucose transport (by glucose transporter GLUT1), and reduction of inflammation by decreased reactivity of microglia. Jointly, these results point to a significant therapeutic potential of gene therapy with pEPito-PEDF for the treatment of DR.
Collapse
Affiliation(s)
- Sofia M Calado
- 1 Doctoral Program in Biomedical Sciences, Department of Biomedical Sciences and Medicine, University of Algarve , Faro, Portugal .,2 CEDOC, NOVA Medical School, Faculdade de Ciências Médicas, Universidade Nova de Lisboa , Lisboa, Portugal
| | - Francisco Diaz-Corrales
- 3 Department of Cell Therapy and Regenerative Medicine, Andalusian Molecular Biology and Regenerative Medicine Centre (CABIMER) , Seville, Spain
| | - Gabriela A Silva
- 2 CEDOC, NOVA Medical School, Faculdade de Ciências Médicas, Universidade Nova de Lisboa , Lisboa, Portugal
| |
Collapse
|
42
|
Cre recombinase expression or topical tamoxifen treatment do not affect retinal structure and function, neuronal vulnerability or glial reactivity in the mouse eye. Neuroscience 2016; 325:188-201. [PMID: 27026593 DOI: 10.1016/j.neuroscience.2016.03.050] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2016] [Revised: 03/20/2016] [Accepted: 03/22/2016] [Indexed: 01/06/2023]
Abstract
Mice with a constitutive or tamoxifen-induced Cre recombinase (Cre) expression are frequently used research tools to allow the conditional deletion of target genes via the Cre-loxP system. Here we analyzed for the first time in a comprehensive and comparative way, whether retinal Cre expression or topical tamoxifen treatment itself would cause structural or functional changes, including changes in the expression profiles of molecular markers, glial reactivity and photoreceptor vulnerability. To this end, we characterized the transgenic α-Cre, Lmop-Cre and the tamoxifen-inducible CAGG-CreER™ mouse lines, all having robust Cre expression in the neuronal retina. In addition, we characterized the effects of topical tamoxifen treatment itself in wildtype mice. We performed morphometric analyses, immunohistochemical staining, in vivo ERG and angiography analyses and realtime RT-PCR analyses. Furthermore, the influence of Cre recombinase or topical tamoxifen exposure on neuronal vulnerability was studied by using light damage as a model for photoreceptor degeneration. Taken together, neither the expression of Cre, nor topical tamoxifen treatment caused detectable changes in retinal structure and function, the expression profiles of investigated molecular markers, glial reactivity and photoreceptor vulnerability. We conclude that the Cre-loxP system and its induction through tamoxifen is a safe and reliable method to delete desired target genes in the neural retina.
Collapse
|
43
|
Modeling long-term diabetes and related complications in rats. J Pharmacol Toxicol Methods 2016; 78:1-12. [DOI: 10.1016/j.vascn.2015.11.003] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2015] [Revised: 11/10/2015] [Accepted: 11/12/2015] [Indexed: 12/22/2022]
|
44
|
Liu YJ, Lian ZY, Liu G, Zhou HY, Yang HJ. RNA sequencing reveals retinal transcriptome changes in STZ-induced diabetic rats. Mol Med Rep 2016; 13:2101-9. [PMID: 26781437 PMCID: PMC4768987 DOI: 10.3892/mmr.2016.4793] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2015] [Accepted: 12/11/2015] [Indexed: 02/05/2023] Open
Abstract
The present study aimed to investigate changes in retinal gene expression in streptozotocin (STZ)‑induced diabetic rats using next‑generation sequencing, utilize transcriptome signatures to investigate the molecular mechanisms of diabetic retinopathy (DR), and identify novel strategies for the treatment of DR. Diabetes was chemically induced in 10‑week‑old male Sprague‑Dawley rats using STZ. Flash‑electroretinography (F‑ERG) was performed to evaluate the visual function of the rats. The retinas of the rats were removed to perform high throughput RNA sequence (RNA‑seq) analysis. The a‑wave, b‑wave, oscillatory potential 1 (OP1), OP2 and ∑OP amplitudes were significantly reduced in the diabetic group, compared with those of the control group (P<0.05). Furthermore, the implicit b‑wave duration 16 weeks post‑STZ induction were significantly longer in the diabetic rats, compared with the control rats (P<0.001). A total of 868 genes were identified, of which 565 were upregulated and 303 were downregulated. Among the differentially expressed genes (DEGs), 94 apoptotic genes and apoptosis regulatory genes, and 19 inflammatory genes were detected. The results of the KEGG pathway significant enrichment analysis revealed enrichment in cell adhesion molecules, complement and coagulation cascades, and antigen processing and presentation. Diabetes alters several transcripts in the retina, and RNA‑seq provides novel insights into the molecular mechanisms underlying DR.
Collapse
Affiliation(s)
- Yuan-Jie Liu
- Department of Human Anatomy, West China School of Preclinical and Forensic Medicine, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Zhi-Yun Lian
- Department of Neurology, West China Hospital of Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Geng Liu
- Department of Human Anatomy, West China School of Preclinical and Forensic Medicine, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Hong-Ying Zhou
- Department of Human Anatomy, West China School of Preclinical and Forensic Medicine, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Hui-Jun Yang
- Department of Human Anatomy, West China School of Preclinical and Forensic Medicine, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| |
Collapse
|
45
|
Sun Y, Ju M, Lin Z, Fredrick TW, Evans LP, Tian KT, Saba NJ, Morss PC, Pu WT, Chen J, Stahl A, Joyal JS, Smith LEH. SOCS3 in retinal neurons and glial cells suppresses VEGF signaling to prevent pathological neovascular growth. Sci Signal 2015; 8:ra94. [PMID: 26396267 DOI: 10.1126/scisignal.aaa8695] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Neurons and glial cells in the retina contribute to neovascularization, or the formation of abnormal new blood vessels, in proliferative retinopathy, a condition that can lead to vision loss or blindness. We identified a mechanism by which suppressor of cytokine signaling 3 (SOCS3) in neurons and glial cells prevents neovascularization. We found that Socs3 expression was increased in the retinal ganglion cell and inner nuclear layers after oxygen-induced retinopathy. Mice with Socs3 deficiency in neuronal and glial cells had substantially reduced vaso-obliterated retinal areas and increased pathological retinal neovascularization in response to oxygen-induced retinopathy, suggesting that loss of neuronal/glial SOCS3 increased both retinal vascular regrowth and pathological neovascularization. Furthermore, retinal expression of Vegfa (which encodes vascular endothelial growth factor A) was higher in these mice than in Socs3 flox/flox controls, indicating that neuronal and glial SOCS3 suppressed Vegfa expression during pathological conditions. Lack of neuronal and glial SOCS3 resulted in greater phosphorylation and activation of STAT3, which led to increased expression of its gene target Vegfa, and increased endothelial cell proliferation. In summary, SOCS3 in neurons and glial cells inhibited the STAT3-mediated secretion of VEGF from these cells, which suppresses endothelial cell activation, resulting in decreased endothelial cell proliferation and angiogenesis. These results suggest that neuronal and glial cell SOCS3 limits pathological retinal angiogenesis by suppressing VEGF signaling.
Collapse
Affiliation(s)
- Ye Sun
- Department of Ophthalmology, Harvard Medical School, Boston Children's Hospital, 300 Longwood Avenue, Boston, MA 02115, USA
| | - Meihua Ju
- Department of Ophthalmology, Harvard Medical School, Boston Children's Hospital, 300 Longwood Avenue, Boston, MA 02115, USA
| | - Zhiqiang Lin
- Department of Cardiology, Harvard Medical School, Boston Children's Hospital, Boston, MA 02115, USA
| | - Thomas W Fredrick
- Department of Ophthalmology, Harvard Medical School, Boston Children's Hospital, 300 Longwood Avenue, Boston, MA 02115, USA
| | - Lucy P Evans
- Department of Ophthalmology, Harvard Medical School, Boston Children's Hospital, 300 Longwood Avenue, Boston, MA 02115, USA
| | - Katherine T Tian
- Department of Ophthalmology, Harvard Medical School, Boston Children's Hospital, 300 Longwood Avenue, Boston, MA 02115, USA
| | - Nicholas J Saba
- Department of Ophthalmology, Harvard Medical School, Boston Children's Hospital, 300 Longwood Avenue, Boston, MA 02115, USA
| | - Peyton C Morss
- Department of Ophthalmology, Harvard Medical School, Boston Children's Hospital, 300 Longwood Avenue, Boston, MA 02115, USA
| | - William T Pu
- Department of Cardiology, Harvard Medical School, Boston Children's Hospital, Boston, MA 02115, USA. Harvard Stem Cell Institute, Harvard University, Cambridge, MA 02138, USA
| | - Jing Chen
- Department of Ophthalmology, Harvard Medical School, Boston Children's Hospital, 300 Longwood Avenue, Boston, MA 02115, USA
| | - Andreas Stahl
- University Eye Hospital Freiburg, Killianstr. 5, Freiburg 79106, Germany
| | - Jean-Sébastien Joyal
- Department of Pediatrics, Centre Hospitalier Universitaire (CHU) Sainte-Justine Research Center, Université de Montréal, Montréal, Québec H3T1C4, Canada
| | - Lois E H Smith
- Department of Ophthalmology, Harvard Medical School, Boston Children's Hospital, 300 Longwood Avenue, Boston, MA 02115, USA.
| |
Collapse
|
46
|
Interplay of proliferation and differentiation factors is revealed in the early human eye development. Graefes Arch Clin Exp Ophthalmol 2015; 253:2187-201. [PMID: 26255818 DOI: 10.1007/s00417-015-3128-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2015] [Revised: 07/20/2015] [Accepted: 07/28/2015] [Indexed: 10/23/2022] Open
Abstract
BACKGROUND Eye development is a consequence of numerous epithelial-to-mesenchymal interactions between the prospective lens ectoderm, outpocketings of the forebrain forming optic vesicles, and surrounding mesenchyme. How different cell types forming eye structures differentiate from their precursors, and which factors coordinate complex human eye development remains largely unknown. Proper differentiation of photoreceptors is of special interest because of their involvement in the appearance of degenerative retinal diseases. METHODS Here we analyze the spatiotemporal expression of neuronal markers nestin, protein gene product 9.5 (PGP9.5), and calcium binding protein (S100), proliferation marker (Ki-67), markers for cilia (alpha-tubulin), and cell stemness marker octamer-binding transcription factor 4 (Oct-4) in histological sections of 5-12 -week human eyes using immunohistochemical and immunofluorescence methods. RESULTS While during the investigated developmental period nestin shows strong expression in all mesenchymal derivatives, lens, optic stalk and inner neuroblastic layer, PGP9.5 and S100 expression characterizes only neural derivatives (optic nerve and neural retina). PGP9.5 is co-localized with nestin and S100 in the differentiating cells of the inner neuroblastic layer. Initially strong proliferation in all parts of the developing eye gradually ceases, especially in the outer neuroblastic layer. Proliferating Ki-67 positive cells co-localize with nestin in the retina, lens, and choroid. Strong Oct-4 and alpha-tubulin immunoreactivity in the retina and optic nerve gradually decreases, while they co-localize in outer neuroblastic and nerve fiber layers. CONCLUSIONS The described expression of investigated markers indicates their importance in eye growth and morphogenesis, while their spatially and temporally restricted pattern coincides with differentiation of initially immature cells into specific retinal cell lineages. Alterations in their spatiotemporal interplay might lead to disturbances of visual function.
Collapse
|
47
|
Therapeutical Strategies for Spinal Cord Injury and a Promising Autologous Astrocyte-Based Therapy Using Efficient Reprogramming Techniques. Mol Neurobiol 2015; 53:2826-2842. [DOI: 10.1007/s12035-015-9157-7] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2015] [Accepted: 03/19/2015] [Indexed: 01/01/2023]
|
48
|
|
49
|
Kawano J. Chemoarchitecture of glial fibrillary acidic protein (GFAP) and glutamine synthetase in the rat optic nerve: an immunohistochemical study. Okajimas Folia Anat Jpn 2015; 92:11-30. [PMID: 26448374 DOI: 10.2535/ofaj.92.11] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/05/2023]
Abstract
An immunohistochemical analysis of the chemoarchitecture of glial fibrillary acidic protein (GFAP) and glutamine synthetase (GS) was conducted in the rat optic nerve. The optic nerve has been divided into 3 regions: the intraretinal, unmyelinated, and myelinated regions. However, it currently remains unclear whether the chemoarchitecture of GFAP and GS is homogeneously organized, especially in the myelinated region. The intraretinal region was divided into intraretinal regions 1 (i1) and 2 (i2). GFAP immunoreactivity was very strong in the i2 and unmyelinated regions, and strong in the i1 region. GS immunoreactivity was moderate in the i1 and i2 regions, and weak in the unmyelinated region. The myelinated region was separated into myelinated regions 1 (m1) and 2 (m2). In the m1 region, GFAP immunoreactivity was strong and GS immunoreactivity was moderate; however, GFAP immunoreactivity was moderate and GS immunoreactivity was weak in the m2 region. Thus, the chemoarchitecture was heterogeneously organized in the myelinated region, with the i1, i2 and m1 regions being the main GS distribution sites. Moreover, most GS-immunoreactive glial cells were oligodendrocytes in the myelinated region. Since GS is a key enzyme in glutamate metabolism, these results may facilitate future investigations for a clearer understanding of glutamate metabolism.
Collapse
Affiliation(s)
- June Kawano
- Department of Morphological Sciences, Kagoshima University Graduate School of Medical and Dental Sciences
| |
Collapse
|
50
|
Cell transcriptional state alters genomic patterns of DNA double-strand break repair in human astrocytes. Nat Commun 2014; 5:5799. [PMID: 25517576 DOI: 10.1038/ncomms6799] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2014] [Accepted: 11/07/2014] [Indexed: 01/11/2023] Open
Abstract
The misrepair of DNA double-strand breaks in close spatial proximity within the nucleus can result in chromosomal rearrangements that are important in the pathogenesis of haematopoietic and solid malignancies. It is unknown why certain epigenetic states, such as those found in stem or progenitor cells, appear to facilitate neoplastic transformation. Here we show that altering the transcriptional state of human astrocytes alters patterns of DNA damage repair from ionizing radiation at a gene locus-specific and genome-wide level. Astrocytes induced into a reactive state exhibit increased DNA repair, compared with non-reactive cells, in actively transcribed chromatin after irradiation. In mapping these repair sites, we identify misrepair events and repair hotspots that are unique to each state. The precise characterization of genomic regions susceptible to mutation in specific transcriptional states provides new opportunities for addressing clonal evolution in solid cancers, in particular those where double-strand break induction is a cornerstone of clinical intervention.
Collapse
|