1
|
Soucy JR, Oswald J, Kriukov E, Phay M, Masland J, Pernstich C, Baranov P. Sustained neurotrophic factor cotreatment enhances donor and host retinal ganglion cell survival in mice. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.03.07.583961. [PMID: 38558999 PMCID: PMC10979869 DOI: 10.1101/2024.03.07.583961] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/04/2024]
Abstract
Retinal ganglion cells (RGCs) lack regenerative capacity in mammals, and their degeneration in glaucoma leads to irreversible blindness. Autologous and allogeneic RGC replacement with stem cell-derived neurons has been established as a promising strategy for vision restoration, but it has been limited by relatively low (<1%) survival rates of donor cells in the hostile microenvironment of a diseased retina and optic nerve. Brain-derived neurotrophic factor (BDNF) and glial-derived neurotrophic factor (GDNF) are known to support the survival of neurons, including RGCs in. vitro and in vivo. Here, we aim to improve donor RGC survival by supplementing our in vitro cultures and in transplants with a slow-release formulation of these neuroprotective agents. We show that slow-release BDNF/GDNF significantly enhances RGC differentiation, survival, and function in vitro. Furthermore, we demonstrated that BDNF/GDNF co-treatment improved mouse and human stem cell-derived donor RGC transplantation outcomes by 2.7- and 15-fold in mice, respectively. Lastly, we show that slow-release BDNF/GDNF provides neuroprotective effects on host RGCs, preserving retinal function in a model of optic neuropathy. Altogether, this approach of engineering the retinal microenvironment with slow-release neurotrophic factors significantly enhances both donor and host neuron survival, representing a promising approach for treating glaucoma and other optic neuropathies.
Collapse
|
2
|
Sekyi MT, Feri M, Desfor S, Atkinson KC, Golestany B, Beltran F, Tiwari-Woodruff SK. Demyelination and neurodegeneration early in experimental autoimmune encephalomyelitis contribute to functional deficits in the anterior visual pathway. Sci Rep 2024; 14:24048. [PMID: 39402114 PMCID: PMC11473523 DOI: 10.1038/s41598-024-73792-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Accepted: 09/20/2024] [Indexed: 10/17/2024] Open
Abstract
Impaired visual function is a prevalent feature of optic neuritis (ON) in multiple sclerosis (MS). Abnormal visual evoked potential (VEP) findings of increased latencies, reduced amplitudes and abnormal waveforms as well as decreased retinal nerve fiber layer (RNFL) assessed by optical coherence tomography (OCT) are hallmarks of ON-induced visual dysfunction. Here we utilized the experimental autoimmune encephalomyelitis (EAE) mouse model of MS to investigate the functional and pathological progression during early (before any clinical symptoms), peak (initial maximal clinical symptoms), and late (chronic disease for > 3 weeks) disease stages. Demyelination and initial stages of axon damage were observed in early EAE. Significant demyelination, inflammation, increased axon damage and impaired P1/N2 amplitudes and latencies by VEP were seen in middle and late EAE groups. A decrease in RNFL thickness by OCT was observed only during late EAE. NanoString analysis of optic nerves from late EAE indicated elevated inflammation-related genes, reduced myelin-related genes, and changes in axon degeneration-related genes. Early inflammatory demyelination and functional deficits of the visual pathway, if untreated, may lead to severe irrecoverable axon damage in EAE. These studies potentially help explain the progression of visual dysfunction during MS.
Collapse
Affiliation(s)
- Maria T Sekyi
- Division of Biomedical Sciences, School of Medicine, University of California Riverside, Rm 3140, Multidisciplinary Research Building, 900 University Ave, Riverside, CA, 92521, USA
| | - Micah Feri
- Division of Biomedical Sciences, School of Medicine, University of California Riverside, Rm 3140, Multidisciplinary Research Building, 900 University Ave, Riverside, CA, 92521, USA
| | - Shane Desfor
- Division of Biomedical Sciences, School of Medicine, University of California Riverside, Rm 3140, Multidisciplinary Research Building, 900 University Ave, Riverside, CA, 92521, USA
| | - Kelley C Atkinson
- Division of Biomedical Sciences, School of Medicine, University of California Riverside, Rm 3140, Multidisciplinary Research Building, 900 University Ave, Riverside, CA, 92521, USA
| | - Batis Golestany
- Division of Biomedical Sciences, School of Medicine, University of California Riverside, Rm 3140, Multidisciplinary Research Building, 900 University Ave, Riverside, CA, 92521, USA
| | - Fernando Beltran
- Division of Biomedical Sciences, School of Medicine, University of California Riverside, Rm 3140, Multidisciplinary Research Building, 900 University Ave, Riverside, CA, 92521, USA
| | - Seema K Tiwari-Woodruff
- Division of Biomedical Sciences, School of Medicine, University of California Riverside, Rm 3140, Multidisciplinary Research Building, 900 University Ave, Riverside, CA, 92521, USA.
| |
Collapse
|
3
|
Castoldi V, Zerbini G, Maestroni S, Viganò I, Rama P, Leocani L. Topical Nerve Growth Factor (NGF) restores electrophysiological alterations in the Ins2 Akita mouse model of diabetic retinopathy. Exp Eye Res 2023; 237:109693. [PMID: 37890756 DOI: 10.1016/j.exer.2023.109693] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Revised: 10/02/2023] [Accepted: 10/24/2023] [Indexed: 10/29/2023]
Abstract
People suffering from diabetes mellitus commonly have to face diabetic retinopathy (DR), an eye disease characterized by early retinal neurodegeneration and microvascular damage, progressively leading to sight loss. The Ins2Akita (Akita) diabetic mouse presents the characteristics of DR and experimental drugs can be tested on this model to check their efficacy before going to the clinic. Topical administration of Nerve Growth Factor (NGF) has been recently demonstrated to prevent DR in the Akita mouse, reverting the thinning of retinal layers and protecting the retinal ganglion cells (RGCs) from death. In this study, we characterize the effects of topical NGF on neuroretina function, quantified with the electroretinogram (ERG). In particular, we show that NGF can ameliorate RGC conduction in the retina of Akita mice, which correlates with a recovery of retinal nerve fiber plus ganglion cell layer (RNFL-GCL) structure. Overall, our preclinical results highlight that topical administration of NGF could be a promising therapeutic approach for DR, being capable of exerting a beneficial impact on retinal functionality.
Collapse
Affiliation(s)
- Valerio Castoldi
- Experimental Neurophysiology Unit, Institute of Experimental Neurology-INSPE, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Gianpaolo Zerbini
- Complications of Diabetes Unit, Diabetes Research Institute-DRI, IRCCS Ospedale San Raffaele, Milan, Italy
| | - Silvia Maestroni
- Complications of Diabetes Unit, Diabetes Research Institute-DRI, IRCCS Ospedale San Raffaele, Milan, Italy
| | - Ilaria Viganò
- Complications of Diabetes Unit, Diabetes Research Institute-DRI, IRCCS Ospedale San Raffaele, Milan, Italy
| | - Paolo Rama
- Cornea and Ocular Surface Unit, IRCCS Ospedale San Raffaele, Milan, Italy; Vita-Salute San Raffaele University, Milan, Italy
| | - Letizia Leocani
- Experimental Neurophysiology Unit, Institute of Experimental Neurology-INSPE, IRCCS San Raffaele Scientific Institute, Milan, Italy; Vita-Salute San Raffaele University, Milan, Italy.
| |
Collapse
|
4
|
Soucy JR, Aguzzi EA, Cho J, Gilhooley MJ, Keuthan C, Luo Z, Monavarfeshani A, Saleem MA, Wang XW, Wohlschlegel J, Baranov P, Di Polo A, Fortune B, Gokoffski KK, Goldberg JL, Guido W, Kolodkin AL, Mason CA, Ou Y, Reh TA, Ross AG, Samuels BC, Welsbie D, Zack DJ, Johnson TV. Retinal ganglion cell repopulation for vision restoration in optic neuropathy: a roadmap from the RReSTORe Consortium. Mol Neurodegener 2023; 18:64. [PMID: 37735444 PMCID: PMC10514988 DOI: 10.1186/s13024-023-00655-y] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Accepted: 09/07/2023] [Indexed: 09/23/2023] Open
Abstract
Retinal ganglion cell (RGC) death in glaucoma and other optic neuropathies results in irreversible vision loss due to the mammalian central nervous system's limited regenerative capacity. RGC repopulation is a promising therapeutic approach to reverse vision loss from optic neuropathies if the newly introduced neurons can reestablish functional retinal and thalamic circuits. In theory, RGCs might be repopulated through the transplantation of stem cell-derived neurons or via the induction of endogenous transdifferentiation. The RGC Repopulation, Stem Cell Transplantation, and Optic Nerve Regeneration (RReSTORe) Consortium was established to address the challenges associated with the therapeutic repair of the visual pathway in optic neuropathy. In 2022, the RReSTORe Consortium initiated ongoing international collaborative discussions to advance the RGC repopulation field and has identified five critical areas of focus: (1) RGC development and differentiation, (2) Transplantation methods and models, (3) RGC survival, maturation, and host interactions, (4) Inner retinal wiring, and (5) Eye-to-brain connectivity. Here, we discuss the most pertinent questions and challenges that exist on the path to clinical translation and suggest experimental directions to propel this work going forward. Using these five subtopic discussion groups (SDGs) as a framework, we suggest multidisciplinary approaches to restore the diseased visual pathway by leveraging groundbreaking insights from developmental neuroscience, stem cell biology, molecular biology, optical imaging, animal models of optic neuropathy, immunology & immunotolerance, neuropathology & neuroprotection, materials science & biomedical engineering, and regenerative neuroscience. While significant hurdles remain, the RReSTORe Consortium's efforts provide a comprehensive roadmap for advancing the RGC repopulation field and hold potential for transformative progress in restoring vision in patients suffering from optic neuropathies.
Collapse
Affiliation(s)
- Jonathan R Soucy
- Department of Ophthalmology, Schepens Eye Research Institute of Mass. Eye and Ear, Harvard Medical School, Boston, MA, USA
| | - Erika A Aguzzi
- The Institute of Ophthalmology, University College London, London, England, UK
| | - Julie Cho
- Spencer Center for Vision Research, Byers Eye Institute, Stanford University School of Medicine, Palo Alto, CA, USA
| | - Michael James Gilhooley
- The Institute of Ophthalmology, University College London, London, England, UK
- Moorfields Eye Hospital, London, England, UK
| | - Casey Keuthan
- Department of Ophthalmology, Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Ziming Luo
- Spencer Center for Vision Research, Byers Eye Institute, Stanford University School of Medicine, Palo Alto, CA, USA
| | - Aboozar Monavarfeshani
- Center for Brain Science and Department of Molecular and Cellular Biology, Harvard University, Cambridge, MA, USA
- Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA, USA
| | - Meher A Saleem
- Bascom Palmer Eye Institute, University of Miami Health System, Miami, FL, USA
| | - Xue-Wei Wang
- Department of Orthopaedic Surgery, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | | | - Petr Baranov
- Department of Ophthalmology, Schepens Eye Research Institute of Mass. Eye and Ear, Harvard Medical School, Boston, MA, USA
| | - Adriana Di Polo
- Department of Neuroscience, University of Montreal, Montreal, QC, Canada
- University of Montreal Hospital Research Centre, Montreal, QC, Canada
| | - Brad Fortune
- Discoveries in Sight Research Laboratories, Devers Eye Institute and Legacy Research Institute, Legacy Health, Portland, OR, USA
| | - Kimberly K Gokoffski
- Department of Ophthalmology, Roski Eye Institute, University of Southern California, Los Angeles, CA, USA
| | - Jeffrey L Goldberg
- Spencer Center for Vision Research, Byers Eye Institute, Stanford University School of Medicine, Palo Alto, CA, USA
| | - William Guido
- Department of Anatomical Sciences and Neurobiology, School of Medicine, University of Louisville, Louisville, KY, USA
| | - Alex L Kolodkin
- The Solomon H Snyder, Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Carol A Mason
- Departments of Pathology and Cell Biology, Neuroscience, and Ophthalmology, College of Physicians and Surgeons, Zuckerman Mind Brain Behavior Institute, Columbia University, New York, NY, USA
| | - Yvonne Ou
- Department of Ophthalmology, University of California, San Francisco, CA, USA
| | - Thomas A Reh
- Department of Biological Structure, University of Washington, Seattle, WA, USA
| | - Ahmara G Ross
- Departments of Ophthalmology and Neurology, University of Pennsylvania, Philadelphia, PA, USA
| | - Brian C Samuels
- Department of Ophthalmology and Visual Sciences, Callahan Eye Hospital, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Derek Welsbie
- Shiley Eye Institute and Viterbi Family Department of Ophthalmology, University of California, San Diego, CA, USA
| | - Donald J Zack
- Glaucoma Center of Excellence, Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, 21287 MD, USA
- Departments of Neuroscience, Molecular Biology & Genetics, and Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Thomas V Johnson
- Departments of Neuroscience, Molecular Biology & Genetics, and Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
- Cellular & Molecular Medicine Program, Johns Hopkins University School of Medicine, Baltimore, 21287 MD, USA.
| |
Collapse
|
5
|
Lagali PS, Shanmugalingam U, Baker AN, Mezey N, Smith PD, Coupland SG, Tsilfidis C. Assessment of the uniform field electroretinogram for mouse retinal ganglion cell functional analysis. Doc Ophthalmol 2023:10.1007/s10633-023-09933-y. [PMID: 37106219 DOI: 10.1007/s10633-023-09933-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Accepted: 03/26/2023] [Indexed: 04/29/2023]
Abstract
PURPOSE The uniform field electroretinogram (UF-ERG) has been suggested as an alternative to the pattern electroretinogram (PERG) for non-invasive assessment of retinal ganglion cell (RGC) function in primates. We evaluated the validity of the UF-ERG to assess mouse RGC activity in vivo. METHODS Unilateral optic nerve crush (ONC) was performed on adult C57BL/6J mice. Contralateral eyes with uncrushed optic nerves and eyes from surgically naive mice served as experimental controls. Electrophysiological visual assessment was performed at 12 weeks post-ONC. Flash-mediated visual-evoked cortical potentials (VEPs) were measured to confirm the robustness of the ONC procedure. Full-field flash ERGs were used to interrogate photoreceptor and retinal bipolar cell function. RGC function was assessed with pattern ERGs. Summed onset and offset UF-ERG responses to alternating dark and light uniform field flash stimuli of different intensities and wavelengths were recorded from ONC and control eyes, and relative differences were compared to the PERG results. Following electrophysiological analysis, RGC loss was monitored by immunohistochemical staining of the RGC marker protein, RBPMS, in post-mortem retinal tissues. RESULTS ONC dramatically impacts RGC integrity and optic nerve function, demonstrated by reduced RGC counts and near complete elimination of VEPs. ONC did not affect scotopic ERG a-wave and b-wave amplitudes, while PERG amplitudes of eyes subjected to ONC were reduced by approximately 50% compared to controls. Summation of ON and OFF UF-ERG responses did not reveal statistically significant differences between ONC and control eyes, regardless of visual stimulus. CONCLUSIONS PERG responses are markedly impaired upon ONC, while UF-ERG responses are not significantly affected by surgical trauma to RGC axons in mice. The more closely related pattern and uniform field ERGs recorded in primates suggests species-specific differences in RGC features or subpopulations corresponding to PERG and UF-ERG response generators, limiting the utility of the UF-ERG for mouse RGC functional analysis.
Collapse
Affiliation(s)
- Pamela S Lagali
- Neuroscience Program, Ottawa Hospital Research Institute, Ottawa, ON, K1H 8L6, Canada
- University of Ottawa Eye Institute, The Ottawa Hospital, Ottawa, ON, K1H 8L6, Canada
| | | | - Adam N Baker
- Neuroscience Program, Ottawa Hospital Research Institute, Ottawa, ON, K1H 8L6, Canada
- University of Ottawa Eye Institute, The Ottawa Hospital, Ottawa, ON, K1H 8L6, Canada
| | - Natalie Mezey
- Department of Neuroscience, Carleton University, Ottawa, ON, Canada
| | - Patrice D Smith
- Department of Neuroscience, Carleton University, Ottawa, ON, Canada
| | - Stuart G Coupland
- University of Ottawa Eye Institute, The Ottawa Hospital, Ottawa, ON, K1H 8L6, Canada
- Department of Ophthalmology, University of Ottawa, Ottawa, ON, K1H 8M5, Canada
- Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, ON, K1H 8M5, Canada
| | - Catherine Tsilfidis
- Neuroscience Program, Ottawa Hospital Research Institute, Ottawa, ON, K1H 8L6, Canada.
- University of Ottawa Eye Institute, The Ottawa Hospital, Ottawa, ON, K1H 8L6, Canada.
- Department of Ophthalmology, University of Ottawa, Ottawa, ON, K1H 8M5, Canada.
- Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, ON, K1H 8M5, Canada.
| |
Collapse
|
6
|
Xin J, Zhou L, Zhang L, Guo K, Yang D. Neuroprotective Effects of Human Adipose-Derived Mesenchymal Stem Cells in Oxygen-Induced Retinopathy. Cell Transplant 2023; 32:9636897231213309. [PMID: 38018498 PMCID: PMC10687918 DOI: 10.1177/09636897231213309] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Revised: 10/17/2023] [Accepted: 10/25/2023] [Indexed: 11/30/2023] Open
Abstract
This study was designed to provide evidence of the neuroprotective of human adipose-derived mesenchymal stem cells (hADSCs) in oxygen-induced retinopathy (OIR). In vivo, hADSCs were intravitreally injected into OIR mice. Various assessments, including HE (histological evaluation), TUNEL (terminal deoxynucleotidyl transferase dUTP nick end labeling) staining, electroretinogram (ERG) analysis, and retinal flat-mount examination, were performed separately at postnatal days 15 (P15) and 17 (P17) to evaluate neurological damage and functional changes. Western blot analysis of ciliary neurotrophic factor (CNTF), glial cell line-derived neurotrophic factor (GDNF), and brain-derived neurotrophic factor (BDNF) was conducted at P17 to elucidate the neuroprotective mechanism. The P17 OIR group exhibited a significant increase in vascular endothelial cell nuclei and neovascularization that breached the ILM (inner limiting membrane) to the P17 control group. In addition, the retinal nonperfusion areas in the P17 OIR group and the number of apoptotic retinal cells in the P15 OIR group were significantly higher than in the corresponding hADSCs treatment group and control group. There was no significant thickness change in the inner nuclear layer (INL) but the outer nuclear layer (ONL) in the P17 OIR treatment group compared with the P17 OIR group. The cell density in the INL and ONL at P17 in the hADSCs treatment group was not significantly different from the OIR group. The amplitude of a-wave and b-wave in scotopic ERG analysis for the P17 OIR group was significantly lower than in the P17 hADSCs treatment group and the P17 control group. Furthermore, the latency of the a-wave and b-wave in the P17 OIR group was significantly longer than in the P17 hADSCs treatment group and the P17 control group. In addition, the expression levels of CNTF and BDNF in the P17 OIR group were statistically higher than those in the P17 control group, whereas the expression of GDNF was statistically lower in the P17 OIR group, compared with the P17 control group. The expression of CNTF and GDNF in the P17 hADSCs treatment group was statistically higher than in the P17 OIR group. However, the expression of BDNF in the P17 hADSCs treatment group was statistically lower than in the P17 OIR group. This study provides evidence for the neuroprotective effects of hADSCs in OIR.
Collapse
Affiliation(s)
- Jifu Xin
- Department of Ophthalmology, The Affiliated Hospital of Inner Mongolia Medical University, Hohhot, China
| | - Lvlv Zhou
- Department of Ophthalmology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Ling Zhang
- Department of Ophthalmology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Kai Guo
- Department of Ophthalmology, The Affiliated Hospital of Inner Mongolia Medical University, Hohhot, China
| | - Dayong Yang
- Department of Ophthalmology, The Affiliated Hospital of Inner Mongolia Medical University, Hohhot, China
| |
Collapse
|
7
|
Van Hook MJ. Influences of Glaucoma on the Structure and Function of Synapses in the Visual System. Antioxid Redox Signal 2022; 37:842-861. [PMID: 35044228 PMCID: PMC9587776 DOI: 10.1089/ars.2021.0253] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Accepted: 12/31/2021] [Indexed: 11/12/2022]
Abstract
Significance: Glaucoma is an age-related neurodegenerative disorder of the visual system associated with sensitivity to intraocular pressure (IOP). It is the leading irreversible cause of vision loss worldwide, and vision loss results from damage and dysfunction of the retinal output neurons known as retinal ganglion cells (RGCs). Recent Advances: Elevated IOP and optic nerve injury triggers pruning of RGC dendrites, altered morphology of excitatory inputs from presynaptic bipolar cells, and disrupted RGC synaptic function. Less is known about RGC outputs, although evidence to date indicates that glaucoma is associated with altered mitochondrial and synaptic structure and function in RGC-projection targets in the brain. These early functional changes likely contribute to vision loss and might be a window into early diagnosis and treatment. Critical Issues: Glaucoma affects different RGC populations to varying extents and along distinct time courses. The influence of glaucoma on RGC synaptic function as well as the mechanisms underlying these effects remain to be determined. Since RGCs are an especially energetically demanding population of neurons, altered intracellular axon transport of mitochondria and mitochondrial function might contribute to RGC synaptic dysfunction in the retina and brain as well as RGC vulnerability in glaucoma. Future Directions: The mechanisms underlying differential RGC vulnerability remain to be determined. Moreover, the timing and mechanisms of RGCs synaptic dysfunction and degeneration will provide valuable insight into the disease process in glaucoma. Future work will be able to capitalize on these findings to better design diagnostic and therapeutic approaches to detect disease and prevent vision loss. Antioxid. Redox Signal. 37, 842-861.
Collapse
Affiliation(s)
- Matthew J. Van Hook
- Department of Ophthalmology & Visual Science and Truhlsen Eye Institute, University of Nebraska Medical Center, Omaha, Nebraska, USA
- Department of Cellular & Integrative Physiology, Truhlsen Eye Institute, University of Nebraska Medical Center, Omaha, Nebraska, USA
| |
Collapse
|
8
|
Peng J, Jin J, Su W, Shao W, Li W, Li Z, Yu H, Zheng Y, Zhong L. High-Mobility Group Box 1 Inhibitor BoxA Alleviates Neuroinflammation-Induced Retinal Ganglion Cell Damage in Traumatic Optic Neuropathy. Int J Mol Sci 2022; 23:ijms23126715. [PMID: 35743157 PMCID: PMC9223527 DOI: 10.3390/ijms23126715] [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: 05/23/2022] [Revised: 06/07/2022] [Accepted: 06/10/2022] [Indexed: 02/04/2023] Open
Abstract
Traumatic optic neuropathy (TON) is a significant cause of vision loss and irreversible blindness worldwide. It is defined as retinal ganglion cell death and axon degeneration caused by injury. Optic nerve crush (ONC), a well-validated model of TON, activates retinal microglia and initiates neuroinflammation. High-mobility group box 1 (HMGB1), a non-histone chromosomal binding protein in the nucleus of eukaryotic cells, is an important inducer of microglial activation and pro-inflammatory cytokine release. The purpose of this study was to examine the protective effects and mechanism of the HMGB1 inhibitor BoxA to neuroinflammation-induced retinal ganglion cells (RGCs) damage in traumatic optic neuropathy. For that purpose, an optic nerve crush model was established in C57BL/6J mice at 10–12 weeks. Model mice received an intravitreal injection of PBS and the HMGB1 inhibitor BoxA. Our data demonstrated that HMGB1 expression increased after optic nerve crush. Retinal ganglion cell function and morphology were damaged, and retinal ganglion cell numbers were reduced after optic nerve crush. Intravitreal injection of BoxA after ONC can alleviate damage. Furthermore, BoxA reduced microglial activation and expression levels of nuclear factor κB (NF-kB), nucleotide-binding domain, leucine-rich repeat containing protein 3 (NLRP3), and apoptosis-associated speck-like protein containing a CARD (ASC) in experimental ONC mice. In summary, HMGB1 mediates NLRP3 inflammasome via NF-kB to participate in retinal inflammatory injury after ONC. Thus, intravitreal injection of BoxA has potential therapeutic benefits for the effective treatment of RGC death to prevent TON.
Collapse
|
9
|
Losenkova K, Takeda A, Ragauskas S, Cerrada-Gimenez M, Vähätupa M, Kaja S, Paul ML, Schmies CC, Rolshoven G, Müller CE, Sandholm J, Jalkanen S, Kalesnykas G, Yegutkin GG. CD73 controls ocular adenosine levels and protects retina from light-induced phototoxicity. Cell Mol Life Sci 2022; 79:152. [PMID: 35212809 PMCID: PMC8881442 DOI: 10.1007/s00018-022-04187-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2021] [Revised: 01/28/2022] [Accepted: 02/04/2022] [Indexed: 01/03/2023]
Abstract
ATP and adenosine have emerged as important signaling molecules involved in vascular remodeling, retinal functioning and neurovascular coupling in the mammalian eye. However, little is known about the regulatory mechanisms of purinergic signaling in the eye. Here, we used three-dimensional multiplexed imaging, in situ enzyme histochemistry, flow cytometric analysis, and single cell transcriptomics to characterize the whole pattern of purine metabolism in mouse and human eyes. This study identified ecto-nucleoside triphosphate diphosphohydrolase-1 (NTPDase1/CD39), NTPDase2, and ecto-5′-nucleotidase/CD73 as major ocular ecto-nucleotidases, which are selectively expressed in the photoreceptor layer (CD73), optic nerve head, retinal vasculature and microglia (CD39), as well as in neuronal processes and cornea (CD39, NTPDase2). Specifically, microglial cells can create a spatially arranged network in the retinal parenchyma by extending and retracting their branched CD39high/CD73low processes and forming local “purinergic junctions” with CD39low/CD73− neuronal cell bodies and CD39high/CD73− retinal blood vessels. The relevance of the CD73–adenosine pathway was confirmed by flash electroretinography showing that pharmacological inhibition of adenosine production by injection of highly selective CD73 inhibitor PSB-12489 in the vitreous cavity of dark-adapted mouse eyes rendered the animals hypersensitive to prolonged bright light, manifested as decreased a-wave and b-wave amplitudes. The impaired electrical responses of retinal cells in PSB-12489-treated mice were not accompanied by decrease in total thickness of the retina or death of photoreceptors and retinal ganglion cells. Our study thus defines ocular adenosine metabolism as a complex and spatially integrated network and further characterizes the critical role of CD73 in maintaining the functional activity of retinal cells.
Collapse
Affiliation(s)
- Karolina Losenkova
- MediCity Research Laboratory and InFLAMES Flagship, University of Turku, Tykistökatu 6A, 20520, Turku, Finland
| | - Akira Takeda
- MediCity Research Laboratory and InFLAMES Flagship, University of Turku, Tykistökatu 6A, 20520, Turku, Finland
| | | | | | | | - Simon Kaja
- Experimentica Ltd., Kuopio, Finland.,Department of Ophthalmology, Loyola University Chicago, Stritch School of Medicine, Maywood, IL, USA
| | - Marius L Paul
- MediCity Research Laboratory and InFLAMES Flagship, University of Turku, Tykistökatu 6A, 20520, Turku, Finland.,Pharma Center Bonn, Pharmaceutical Institute, Pharmaceutical and Medicinal Chemistry, University of Bonn, Bonn, Germany
| | - Constanze C Schmies
- Pharma Center Bonn, Pharmaceutical Institute, Pharmaceutical and Medicinal Chemistry, University of Bonn, Bonn, Germany
| | - Georg Rolshoven
- Pharma Center Bonn, Pharmaceutical Institute, Pharmaceutical and Medicinal Chemistry, University of Bonn, Bonn, Germany
| | - Christa E Müller
- Pharma Center Bonn, Pharmaceutical Institute, Pharmaceutical and Medicinal Chemistry, University of Bonn, Bonn, Germany
| | - Jouko Sandholm
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, Turku, Finland
| | - Sirpa Jalkanen
- MediCity Research Laboratory and InFLAMES Flagship, University of Turku, Tykistökatu 6A, 20520, Turku, Finland
| | | | - Gennady G Yegutkin
- MediCity Research Laboratory and InFLAMES Flagship, University of Turku, Tykistökatu 6A, 20520, Turku, Finland.
| |
Collapse
|
10
|
Taiyab A, Akula M, Dham J, Deschamps P, Sheardown H, Williams T, Borrás T, West-Mays JA. Deletion of transcription factor AP-2β from the developing murine trabecular meshwork region leads to progressive glaucomatous changes. J Neurosci Res 2021; 100:638-652. [PMID: 34822722 DOI: 10.1002/jnr.24982] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Revised: 09/28/2021] [Accepted: 10/03/2021] [Indexed: 11/08/2022]
Abstract
Glaucoma is one of the leading causes of irreversible blindness and can result from abnormalities in anterior segment structures required for aqueous humor outflow, including the trabecular meshwork (TM) and Schlemm's canal (SC). Transcription factors such as AP-2β play critical roles in anterior segment development. Here, we show that the Mgp-Cre knock-in (Mgp-Cre.KI) mouse can be used to target the embryonic periocular mesenchyme giving rise to the TM and SC. Fate mapping of male and female mice indicates that AP-2β loss causes a decrease in iridocorneal angle cells derived from Mgp-Cre.KI-expressing populations compared to controls. Moreover, histological analyses revealed peripheral iridocorneal adhesions in AP-2β mutants that were accompanied by a decrease in expression of TM and SC markers, as observed using immunohistochemistry. In addition, rebound tonometry showed significantly higher intraocular pressure (IOP) that was correlated with a progressive significant loss of retinal ganglion cells, reduced retinal thickness, and reduced retinal function, as measured using an electroretinogram, in AP-2β mutants compared with controls, reflecting pathology described in late-stage glaucoma patients. Importantly, elevated IOP in AP-2β mutants was significantly reduced by treatment with latanoprost, a prostaglandin analog that increases unconventional outflow. These findings demonstrate that AP-2β is critical for TM and SC development, and that these mutant mice can serve as a model for understanding and treating progressive human primary angle-closure glaucoma.
Collapse
Affiliation(s)
- Aftab Taiyab
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, ON, Canada
| | - Monica Akula
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, ON, Canada
| | - Japnit Dham
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, ON, Canada
| | - Paula Deschamps
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, ON, Canada
| | - Heather Sheardown
- Department of Chemical Engineering, McMaster University, Hamilton, ON, Canada
| | - Trevor Williams
- Department of Craniofacial Biology, University of Colorado, Aurora, CO, USA
| | - Teresa Borrás
- Department of Ophthalmology, University of North Carolina School of Medicine, Chapel Hill, NC, USA
| | - Judith A West-Mays
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, ON, Canada
| |
Collapse
|
11
|
Di Pierdomenico J, Henderson DCM, Giammaria S, Smith VL, Jamet AJ, Smith CA, Hooper ML, Chauhan BC. Age and intraocular pressure in murine experimental glaucoma. Prog Retin Eye Res 2021; 88:101021. [PMID: 34801667 DOI: 10.1016/j.preteyeres.2021.101021] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2021] [Revised: 10/25/2021] [Accepted: 11/08/2021] [Indexed: 12/23/2022]
Abstract
Age and intraocular pressure (IOP) are the two most important risk factors for the development and progression of open-angle glaucoma. While IOP is commonly considered in models of experimental glaucoma (EG), most studies use juvenile or adult animals and seldom older animals which are representative of the human disease. This paper provides a concise review of how retinal ganglion cell (RGC) loss, the hallmark of glaucoma, can be evaluated in EG with a special emphasis on serial in vivo imaging, a parallel approach used in clinical practice. It appraises the suitability of EG models for the purpose of in vivo imaging and argues for the use of models that provide a sustained elevation of IOP, without compromise of the ocular media. In a study with parallel cohorts of adult (3-month-old, equivalent to 20 human years) and old (2-year-old, equivalent to 70 human years) mice, we compare the effects of elevated IOP on serial ganglion cell complex thickness and individual RGC dendritic morphology changes obtained in vivo. We also evaluate how age modulates the impact of elevated IOP on RGC somal and axonal density in histological analysis as well the density of melanopsin RGCs. We discuss the challenges of using old animals and emphasize the potential of single RGC imaging for understanding the pathobiology of RGC loss and evaluating new therapeutic avenues.
Collapse
Affiliation(s)
- Johnny Di Pierdomenico
- Retina and Optic Nerve Research Laboratory, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Delaney C M Henderson
- Retina and Optic Nerve Research Laboratory, Dalhousie University, Halifax, Nova Scotia, Canada; Department of Medical Neuroscience, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Sara Giammaria
- Department of Ophthalmology and Visual Sciences, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Victoria L Smith
- Retina and Optic Nerve Research Laboratory, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Aliénor J Jamet
- Retina and Optic Nerve Research Laboratory, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Corey A Smith
- Retina and Optic Nerve Research Laboratory, Dalhousie University, Halifax, Nova Scotia, Canada; Department of Ophthalmology and Visual Sciences, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Michele L Hooper
- Retina and Optic Nerve Research Laboratory, Dalhousie University, Halifax, Nova Scotia, Canada; Department of Physiology and Biophysics, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Balwantray C Chauhan
- Retina and Optic Nerve Research Laboratory, Dalhousie University, Halifax, Nova Scotia, Canada; Department of Medical Neuroscience, Dalhousie University, Halifax, Nova Scotia, Canada; Department of Ophthalmology and Visual Sciences, Dalhousie University, Halifax, Nova Scotia, Canada; Department of Physiology and Biophysics, Dalhousie University, Halifax, Nova Scotia, Canada.
| |
Collapse
|
12
|
Smith BJ, Côté PD, Tremblay F. Voltage-gated sodium channel-dependent retroaxonal modulation of photoreceptor function during post-natal development in mice. Dev Neurobiol 2021; 81:353-365. [PMID: 33248000 DOI: 10.1002/dneu.22793] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Revised: 11/16/2020] [Accepted: 11/17/2020] [Indexed: 11/11/2022]
Abstract
Juvenile (postnatal day 16) mice lacking Nav 1.6 channels (null-mutant Scn8admu ) have reduced photoreceptor function, which is unexpected given that Nav channels have not been detected in mouse photoreceptors and do not contribute appreciably to photoreceptor function in adults. We demonstrate that acute block of Nav channels with intravitreal TTX in juvenile (P16) wild-type mice has no effect on photoreceptor function. However, reduced light activity by prolonged dark adaptation from P8 caused significant reduction in photoreceptor function at P16. Injecting TTX into the retrobulbar space at P16 to specifically block Nav channels in the optic nerve also caused a reduction in photoreceptor function comparable to that seen at P16 in null-mutant Scn8a mice. In both P16 null-mutant Scn8admu and retrobulbar TTX-injected wild-type mice, photoreceptor function was restored following intravitreal injection of the TrkB receptor agonist 7,8-dihydroxyflavone, linking Nav -dependent retrograde transport to TrkB-dependent neurotrophic factor production pathways as a modulatory influence of photoreceptor function at P16. We also found that in Scn8admu mice, photoreceptor function recovers by P22-25 despite more precarious general health of the animal. Retrobulbar injection of TTX in the wild type still reduced the photoreceptor response at this age but to a lesser extent, suggesting that Nav -dependent modulation of photoreceptor function is largely transient, peaking soon after eye opening. Together, these results suggest that the general photosensitivity of the retina is modulated following eye opening by retrograde transport through activity-dependent retinal ganglion cell axonal signaling targeting TrkB receptors.
Collapse
Affiliation(s)
- Benjamin J Smith
- Department of Biology, Dalhousie University, Halifax, NS, Canada
| | - Patrice D Côté
- Department of Biology, Dalhousie University, Halifax, NS, Canada.,Department of Ophthalmology and Visual Sciences, Dalhousie University, Halifax, NS, Canada
| | - François Tremblay
- Department of Ophthalmology and Visual Sciences, Dalhousie University, Halifax, NS, Canada.,Department of Physiology and Biophysics, Dalhousie University, Halifax, NS, Canada.,Izaak Walton Killam Health Centre, Halifax, NS, Canada
| |
Collapse
|
13
|
Waldner DM, Ito K, Chen LL, Nguyen L, Chow RL, Lee A, Rancourt DE, Tremblay F, Stell WK, Bech-Hansen NT. Transgenic Expression of Cacna1f Rescues Vision and Retinal Morphology in a Mouse Model of Congenital Stationary Night Blindness 2A (CSNB2A). Transl Vis Sci Technol 2020; 9:19. [PMID: 33117610 PMCID: PMC7571326 DOI: 10.1167/tvst.9.11.19] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2020] [Accepted: 09/17/2020] [Indexed: 12/20/2022] Open
Abstract
Purpose Congenital stationary night blindness 2A (CSNB2A) is a genetic retinal disorder characterized by poor visual acuity, nystagmus, strabismus, and other signs of retinal dysfunction resulting from mutations in Cacna1f -the gene coding for the pore-forming subunit of the calcium channel CaV1.4. Mouse models of CSNB2A have shown that mutations causing the disease deleteriously affect photoreceptors and their synapses with second-order neurons. This study was undertaken to evaluate whether transgenic expression of Cacna1f could rescue morphology and visual function in a Cacna1f-KO model of CSNB2A. Methods Strategic creation, breeding and use of transgenic mouse lines allowed for Cre-driven retina-specific expression of Cacna1f in a CSNB2A model. Transgene expression and retinal morphology were investigated with immunohistochemistry in retinal wholemounts or cross-sections. Visual function was assessed by optokinetic response (OKR) analysis and electroretinography (ERG). Results Mosaic, prenatal expression of Cacna1f in the otherwise Cacna1f-KO retina was sufficient to rescue some visual function. Immunohistochemical analyses demonstrated wild-type-like photoreceptor and synaptic morphology in sections with transgenic expression of Cacna1f. Conclusions This report describes a novel system for Cre-inducible expression of Cacna1f in a Cacna1f-KO mouse model of CSNB2A and provides preclinical evidence for the potential use of gene therapy in the treatment of CSNB2A. Translational Relevance These data have relevance in the treatment of CSNB2A and in understanding how photoreceptor integration might be achieved in retinas in which photoreceptors have been lost, such as retinitis pigmentosa, age-related macular degeneration, and other degenerative conditions.
Collapse
Affiliation(s)
- Derek M Waldner
- Graduate Department of Neuroscience, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Kenichi Ito
- Department of Biochemistry and Molecular Biology, University of Calgary, Calgary, AB, Canada
| | - Li-Li Chen
- Department of Biology, University of Victoria, Victoria, BC, Canada
| | - Lisa Nguyen
- Department of Medical Genetics, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Robert L Chow
- Department of Biology, University of Victoria, Victoria, BC, Canada
| | - Amy Lee
- Department of Molecular Physiology and Biophysics, Department of Otolaryngology Head-Neck Surgery and Department of Neurology, University of Iowa, Iowa City, IA, USA
| | - Derrick E Rancourt
- Department of Biochemistry and Molecular Biology, University of Calgary, Calgary, AB, Canada
| | - Francois Tremblay
- Department of Ophthalmology and Visual Sciences, Faculty of Medicine, and Clinical Vision Sciences Program, Faculty of Health Dalhousie University, NS, Canada
| | - William K Stell
- Department of Cell Biology and Anatomy and Department of Surgery, Hotchkiss Brain Institute, and Alberta Children's Hospital Research Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - N Torben Bech-Hansen
- Department of Medical Genetics, and Department of Surgery, Alberta Children's Hospital Research Institute, and Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| |
Collapse
|
14
|
Fairless R, Williams SK, Katiyar R, Maxeiner S, Schmitz F, Diem R. ERG Responses in Mice with Deletion of the Synaptic Ribbon Component RIBEYE. Invest Ophthalmol Vis Sci 2020; 61:37. [PMID: 32437548 PMCID: PMC7405791 DOI: 10.1167/iovs.61.5.37] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Purpose To determine the influence of RIBEYE deletion and the resulting absence of synaptic ribbons on retinal light signaling by electroretinography. Methods Full-field flash electroretinograms (ERGs) were recorded in RIBEYE knock-out (KO) and wild-type (WT) littermate mice under photopic and scotopic conditions, with oscillatory potentials (OPs) extracted by digital filtering. Flicker ERGs and ERGs following intravitreal injection of pharmacological agents were also obtained under scotopic conditions. Results The a-wave amplitudes were unchanged between RIBEYE KO and WT mice; however, the b-wave amplitudes were reduced in KOs under scotopic, but not photopic, conditions. Increasing stimulation frequency led to a greater reduction in RIBEYE KO b-wave amplitudes compared with WTs. Furthermore, we observed prominent, supernormal OPs in RIBEYE KO mice in comparison with WT mice. Following intravitreal injections with l-2 amino-4-phosphonobutyric acid and cis-2,3 piperidine dicarboxylic acid to block ON and OFF responses at photoreceptor synapses, OPs were completely abolished in both mice types, indicating a synaptic origin of the prominent OPs in the KOs. Conversely, tetrodotoxin treatment to block voltage-gated Na+ channels/spiking neurons did not differentially affect OPs in WT and KO mice. Conclusions The decreased scotopic b-wave and decreased responses to increased stimulation frequencies are consistent with signaling malfunctions at photoreceptor and inner retinal ribbon synapses. Because phototransduction in the photoreceptor outer segments is unaffected in the KOs, their supernormal OPs presumably result from a dysfunction in retinal synapses. The relatively mild ERG phenotype in KO mice, particularly in the photopic range, is probably caused by compensatory mechanisms in retinal signaling pathways.
Collapse
|
15
|
Quantification of Changes in Visual Function During Disease Development in a Mouse Model of Pigmentary Glaucoma. J Glaucoma 2019; 27:828-841. [PMID: 30001268 DOI: 10.1097/ijg.0000000000001024] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
PURPOSE We investigated the relationship between visual parameters that are commonly affected during glaucomatous disease progression with functional measures of retina physiology using electroretinography and behavioral measures of visual function in a mouse model of glaucoma. Electroretinogram components measuring retinal ganglion cell (RGC) responses were determined using the non-invasive Ganzfeld flash electroretinography (fERG) to assess RGC loss in a mouse model of glaucoma. METHODS Intraocular pressure (IOP), behaviorally assessed measures of visual function, namely visual acuity and contrast sensitivity as well as fERG responses were recorded in 4- and 11-month-old male DBA/2 mice. Scotopic threshold response (STR) and photopic negative response components as well as oscillatory potentials (OPs) were isolated from fERG responses and correlated with IOP, optomotor reflex measurements, and RGC counts. RESULTS The 11-month-old DBA/2 mice had significantly elevated IOP, reduced visual performance, as assessed behaviorally, significant RGC loss, deficits in standardized fERG responses, reduced STRs, and differences in OP amplitudes and latencies, when compared with 4-month-old mice of the same strain. STRs and OPs correlated with some visual and physiological parameters. In addition, elevated IOP and RGC loss correlated positively with measures of visual function, specifically with surrogate measures of RGC function derived from fERG. CONCLUSIONS Our data suggest that RGC function as well as interactions of RGCs with other retinal cell types is impaired during glaucoma. In addition, a later OP wavelet denoted as OP4 in this study was identified as a very reproducible indicator of loss of visual function in the glaucoma mouse model.
Collapse
|
16
|
Agostinone J, Alarcon-Martinez L, Gamlin C, Yu WQ, Wong ROL, Di Polo A. Insulin signalling promotes dendrite and synapse regeneration and restores circuit function after axonal injury. Brain 2019; 141:1963-1980. [PMID: 29931057 PMCID: PMC6022605 DOI: 10.1093/brain/awy142] [Citation(s) in RCA: 90] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2017] [Accepted: 04/06/2018] [Indexed: 01/07/2023] Open
Abstract
Dendrite pathology and synapse disassembly are critical features of chronic neurodegenerative diseases. In spite of this, the capacity of injured neurons to regenerate dendrites has been largely ignored. Here, we show that, upon axonal injury, retinal ganglion cells undergo rapid dendritic retraction and massive synapse loss that preceded neuronal death. Human recombinant insulin, administered as eye drops or systemically after dendritic arbour shrinkage and prior to cell loss, promoted robust regeneration of dendrites and successful reconnection with presynaptic targets. Insulin-mediated regeneration of excitatory postsynaptic sites on retinal ganglion cell dendritic processes increased neuronal survival and rescued light-triggered retinal responses. Further, we show that axotomy-induced dendrite retraction triggered substantial loss of the mammalian target of rapamycin (mTOR) activity exclusively in retinal ganglion cells, and that insulin fully reversed this response. Targeted loss-of-function experiments revealed that insulin-dependent activation of mTOR complex 1 (mTORC1) is required for new dendritic branching to restore arbour complexity, while complex 2 (mTORC2) drives dendritic process extension thus re-establishing field area. Our findings demonstrate that neurons in the mammalian central nervous system have the intrinsic capacity to regenerate dendrites and synapses after injury, and provide a strong rationale for the use of insulin and/or its analogues as pro-regenerative therapeutics for intractable neurodegenerative diseases including glaucoma.
Collapse
Affiliation(s)
- Jessica Agostinone
- Department of Neuroscience, University of Montreal, Montreal, Quebec, Canada.,University of Montreal Hospital Research Center (CR-CHUM), University of Montreal, Montreal, Quebec, Canada
| | - Luis Alarcon-Martinez
- Department of Neuroscience, University of Montreal, Montreal, Quebec, Canada.,University of Montreal Hospital Research Center (CR-CHUM), University of Montreal, Montreal, Quebec, Canada
| | - Clare Gamlin
- Department of Biological Structure, University of Washington, 1959 NE Pacific Street, Seattle, Washington, USA
| | - Wan-Qing Yu
- Department of Biological Structure, University of Washington, 1959 NE Pacific Street, Seattle, Washington, USA
| | - Rachel O L Wong
- Department of Biological Structure, University of Washington, 1959 NE Pacific Street, Seattle, Washington, USA
| | - Adriana Di Polo
- Department of Neuroscience, University of Montreal, Montreal, Quebec, Canada.,University of Montreal Hospital Research Center (CR-CHUM), University of Montreal, Montreal, Quebec, Canada
| |
Collapse
|
17
|
Blandford SN, Hooper ML, Yabana T, Chauhan BC, Baldridge WH, Farrell SRM. Retinal Characterization of the Thy1-GCaMP3 Transgenic Mouse Line After Optic Nerve Transection. Invest Ophthalmol Vis Sci 2019; 60:183-191. [PMID: 30640971 DOI: 10.1167/iovs.18-25861] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Purpose GCaMP3 is a genetically encoded calcium indicator for monitoring intracellular calcium dynamics. We characterized the expression pattern and functional properties of GCaMP3 in the Thy1-GCaMP3 transgenic mouse retina. Methods To determine the specificity of GCaMP3 expression, Thy1-GCaMP3 (B6; CBA-Tg(Thy1-GCaMP3)6Gfng/J) retinas were processed for immunohistochemistry with anti-green fluorescent protein (anti-GFP, to enhance GCaMP3 fluorescence), anti-RBPMS (retinal ganglion cell [RGC]-specific marker), and antibodies against amacrine cell markers (ChAT, GABA, GAD67, syntaxin). Calcium imaging was used to characterize functional responses of GCaMP3-expressing (GCaMP+) cells by recording calcium transients evoked by superfusion of kainic acid (KA; 10, 50, or 100 μM). In a subset of animals, optic nerve transection (ONT) was performed 3, 5, or 7 days prior to calcium imaging. Results GFP immunoreactivity colocalized with RBPMS but not amacrine cell markers in both ONT and non-ONT (control) groups. Calcium transients evoked by KA were reduced after ONT (50 μM KA; ΔF/F0 [SD]; control: 1.00 [0.67], day 3: 0.50 [0.35], day 5: 0.31 [0.28], day 7: 0.35 [0.36]; P < 0.05 versus control). There was also a decrease in the number of GCaMP3+ cells after ONT (cells/mm2 [SD]; control: 2198 [453], day 3: 2224 [643], day 5: 1383 [375], day 7: 913 [178]; P < 0.05). Furthermore, the proportion of GCaMP3+ cells that responded to KA decreased after ONT (50 μM KA, 97%, 54%, 47%, and 58%; control, 3, 5, and 7 days, respectively). Conclusions Following ONT, functional RGC responses are lost prior to the loss of RGC somata, suggesting that anatomical markers of RGCs may underestimate the extent of RGC dysfunction.
Collapse
Affiliation(s)
- Stephanie N Blandford
- Department of Medical Neuroscience, Dalhousie University, Halifax, Nova Scotia, Canada.,Retina and Optic Nerve Research Laboratory, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Michele L Hooper
- Retina and Optic Nerve Research Laboratory, Dalhousie University, Halifax, Nova Scotia, Canada.,Department of Ophthalmology and Visual Sciences, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Takeshi Yabana
- Retina and Optic Nerve Research Laboratory, Dalhousie University, Halifax, Nova Scotia, Canada.,Tohoku University Graduate School of Medicine, Department of Ophthalmology, Sendai, Japan
| | - Balwantray C Chauhan
- Department of Medical Neuroscience, Dalhousie University, Halifax, Nova Scotia, Canada.,Retina and Optic Nerve Research Laboratory, Dalhousie University, Halifax, Nova Scotia, Canada.,Department of Ophthalmology and Visual Sciences, Dalhousie University, Halifax, Nova Scotia, Canada.,Nova Scotia Health Authority, Halifax, Nova Scotia, Canada
| | - William H Baldridge
- Department of Medical Neuroscience, Dalhousie University, Halifax, Nova Scotia, Canada.,Retina and Optic Nerve Research Laboratory, Dalhousie University, Halifax, Nova Scotia, Canada.,Department of Ophthalmology and Visual Sciences, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Spring R M Farrell
- Department of Medical Neuroscience, Dalhousie University, Halifax, Nova Scotia, Canada.,Retina and Optic Nerve Research Laboratory, Dalhousie University, Halifax, Nova Scotia, Canada.,Nova Scotia Health Authority, Halifax, Nova Scotia, Canada
| |
Collapse
|
18
|
Pérez de Lara MJ, Avilés-Trigueros M, Guzmán-Aránguez A, Valiente-Soriano FJ, de la Villa P, Vidal-Sanz M, Pintor J. Potential role of P2X7 receptor in neurodegenerative processes in a murine model of glaucoma. Brain Res Bull 2019; 150:61-74. [PMID: 31102752 DOI: 10.1016/j.brainresbull.2019.05.006] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2018] [Revised: 04/23/2019] [Accepted: 05/10/2019] [Indexed: 12/20/2022]
Abstract
Glaucoma is a common cause of visual impairment and blindness, characterized by retinal ganglion cell (RGC) death. The mechanisms that trigger the development of glaucoma remain unknown and have gained significant relevance in the study of this neurodegenerative disease. P2X7 purinergic receptors (P2X7R) could be involved in the regulation of the synaptic transmission and neuronal death in the retina through different pathways. The aim of this study was to characterize the molecular signals underlying glaucomatous retinal injury. The time-course of functional, morphological, and molecular changes in the glaucomatous retina of the DBA/2J mice were investigated. The expression and localization of P2X7R was analysed in relation with retinal markers. Caspase-3, JNK, and p38 were evaluated in control and glaucomatous mice by immunohistochemical and western-blot analysis. Furthermore, electroretinogram recordings (ERG) were performed to assess inner retina dysfunction. Glaucomatous mice exhibited changes in P2X7R expression as long as the pathology progressed. There was P2X7R overexpression in RGCs, the primary injured neurons, which correlated with the loss of function through ERG measurements. All analyzed MAPK and caspase-3 proteins were upregulated in the DBA/2J retinas suggesting a pro-apoptotic cell death. The increase in P2X7Rs presence may contribute, together with other factors, to the changes in retinal functionality and the concomitant death of RGCs. These findings provide evidence of possible intracellular pathways responsible for apoptosis regulation during glaucomatous degeneration.
Collapse
Affiliation(s)
- María J Pérez de Lara
- Department of Biochemistry and Molecular Biology IV, Faculty of Optics and Optometry, Complutense University of Madrid, c/Arcos de Jalón 118, E-28037, Madrid, Spain
| | - Marcelino Avilés-Trigueros
- Laboratory of Experimental Ophthalmology, Dept. of Ophthalmology, Faculty of Medicine, University of Murcia and Murcia Institute of Bio-Health Research (IMIB), E-30120, El Palmar, Murcia, Spain
| | - Ana Guzmán-Aránguez
- Department of Biochemistry and Molecular Biology IV, Faculty of Optics and Optometry, Complutense University of Madrid, c/Arcos de Jalón 118, E-28037, Madrid, Spain
| | - F Javier Valiente-Soriano
- Laboratory of Experimental Ophthalmology, Dept. of Ophthalmology, Faculty of Medicine, University of Murcia and Murcia Institute of Bio-Health Research (IMIB), E-30120, El Palmar, Murcia, Spain
| | - Pedro de la Villa
- Systems Biology Department, Faculty of Medicine, University of Alcalá, Alcalá de Henares, Madrid, Spain
| | - Manuel Vidal-Sanz
- Laboratory of Experimental Ophthalmology, Dept. of Ophthalmology, Faculty of Medicine, University of Murcia and Murcia Institute of Bio-Health Research (IMIB), E-30120, El Palmar, Murcia, Spain.
| | - Jesús Pintor
- Department of Biochemistry and Molecular Biology IV, Faculty of Optics and Optometry, Complutense University of Madrid, c/Arcos de Jalón 118, E-28037, Madrid, Spain
| |
Collapse
|
19
|
Eriksen AZ, Eliasen R, Oswald J, Kempen PJ, Melander F, Andresen TL, Young M, Baranov P, Urquhart AJ. Multifarious Biologic Loaded Liposomes that Stimulate the Mammalian Target of Rapamycin Signaling Pathway Show Retina Neuroprotection after Retina Damage. ACS NANO 2018; 12:7497-7508. [PMID: 30004669 PMCID: PMC6117751 DOI: 10.1021/acsnano.8b00596] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/23/2018] [Accepted: 07/13/2018] [Indexed: 05/08/2023]
Abstract
A common event in optic neuropathies is the loss of axons and death of retinal ganglion cells (RGCs) resulting in irreversible blindness. Mammalian target of rapamycin (mTOR) signaling pathway agonists have been shown to foster axon regeneration and RGC survival in animal models of optic nerve damage. However, many challenges remain in developing therapies that exploit cell growth and tissue remodeling including (i) activating/inhibiting cell pathways synergistically, (ii) avoiding tumorigenesis, and (iii) ensuring appropriate physiological tissue function. These challenges are further exacerbated by the need to overcome ocular physiological barriers and clearance mechanisms. Here we present liposomes loaded with multiple mTOR pathway stimulating biologics designed to enhance neuroprotection after retina damage. Liposomes were loaded with ciliary neurotrophic factor, insulin-like growth factor 1, a lipopeptide N-fragment osteopontin mimic, and lipopeptide phosphatase tension homologue inhibitors for either the ATP domain or the c-terminal tail. In a mouse model of N-methyl-d-aspartic acid induced RGC death, a single intravitreal administration of liposomes reduced both RGC death and loss of retina electrophysiological function. Furthermore, combining liposomes with transplantation of induced pluripotent stem cell derived RGCs led to an improved electrophysiological outcome in mice. The results presented here show that liposomes carrying multiple signaling pathway modulators can facilitate neuroprotection and transplant electrophysiological outcome.
Collapse
Affiliation(s)
- Anne Z. Eriksen
- DTU
Nanotech, Department of Micro- and Nanotechnology, Technical University of Denmark, 2800 Kongens Lyngby, Denmark
| | - Rasmus Eliasen
- DTU
Nanotech, Department of Micro- and Nanotechnology, Technical University of Denmark, 2800 Kongens Lyngby, Denmark
| | - Julia Oswald
- Schepens
Eye Research Institute, Massachusetts Eye and Ear, 20 Staniford Street, Boston, Massachusetts 02114, United States
| | - Paul J. Kempen
- DTU
Nanotech, Department of Micro- and Nanotechnology, Technical University of Denmark, 2800 Kongens Lyngby, Denmark
| | - Fredrik Melander
- DTU
Nanotech, Department of Micro- and Nanotechnology, Technical University of Denmark, 2800 Kongens Lyngby, Denmark
| | - Thomas L. Andresen
- DTU
Nanotech, Department of Micro- and Nanotechnology, Technical University of Denmark, 2800 Kongens Lyngby, Denmark
| | - Michael Young
- Schepens
Eye Research Institute, Massachusetts Eye and Ear, 20 Staniford Street, Boston, Massachusetts 02114, United States
| | - Petr Baranov
- Schepens
Eye Research Institute, Massachusetts Eye and Ear, 20 Staniford Street, Boston, Massachusetts 02114, United States
| | - Andrew J. Urquhart
- DTU
Nanotech, Department of Micro- and Nanotechnology, Technical University of Denmark, 2800 Kongens Lyngby, Denmark
| |
Collapse
|
20
|
Park SJ, Paik SS, Lee JY, Oh SJ, Kim IB. Blue-on-Green Flash Induces Maximal Photopic Negative Response and Oscillatory Potential and Serves as a Diagnostic Marker for Glaucoma in Rat Retina. Exp Neurobiol 2018; 27:210-216. [PMID: 30022872 PMCID: PMC6050416 DOI: 10.5607/en.2018.27.3.210] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2018] [Revised: 05/23/2018] [Accepted: 05/28/2018] [Indexed: 11/19/2022] Open
Abstract
The purpose of this study was to investigate the application of various electroretinography (ERG) to the diagnosis of inner retinal dysfunction induced by mild intraocular pressure (IOP) elevation in a rat glaucoma model. For inner retinal function measurements, available photopic ERG protocols were applied under various light conditions including monochromatic combinations, which complement conventional scotopic ERG. Three episcleral veins in the right eyes of Sprague-Dawley rats were cauterized to induce an experimental model of glaucoma, leading to mild IOP elevation. ERG responses were measured before surgery and at 1, 2, 4, and 8 weeks after cauterization. We first confirmed that the amplitude reduction in the standard photopic b-wave was almost comparable to the amplitudes of scotopic a- and b-waves in glaucomatous eyes over time. We have implemented additional photopic ERG protocols under different stimulus conditions, which consisted of a longer duration and different monochromatic combinations. Such a change in the stimulations resulted in more pronounced differences in response between the two groups. Especially in normal animals, blue stimulation on a green background produced the largest b-wave and photopic negative response (PhNR) amplitudes and caused more pronounced oscillatory potential (OP) wavelets (individual components). In glaucomatous eyes, blue stimulation on a green background significantly reduced PhNR amplitudes and abolished the robust OP components. These results, by providing the usefulness of blue on green combination, suggest the applicable photopic ERG protocol that complements the conventional ERG methods of accessing the progression of glaucomatous damage in the rat retina.
Collapse
Affiliation(s)
- Su Jin Park
- Department of Anatomy, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea.,Catholic Neuroscience Institute, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea
| | - Sun Sook Paik
- Department of Anatomy, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea.,Catholic Neuroscience Institute, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea
| | - Ji-Yeon Lee
- Department of Anatomy, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea
| | - Su-Ja Oh
- Department of Anatomy, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea
| | - In-Beom Kim
- Department of Anatomy, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea.,Catholic Neuroscience Institute, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea.,Catholic Institute for Applied Anatomy, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea
| |
Collapse
|
21
|
Ji S, Lin S, Chen J, Huang X, Wei CC, Li Z, Tang S. Neuroprotection of Transplanting Human Umbilical Cord Mesenchymal Stem Cells in a Microbead Induced Ocular Hypertension Rat Model. Curr Eye Res 2018; 43:810-820. [PMID: 29505314 DOI: 10.1080/02713683.2018.1440604] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
PURPOSE The purpose of this study is to investigate the potential therapeutic benefits of intravitreally transplanted human umbilical cord mesenchymal stem cells (UC-MSCs) in an animal model of microbead-injection-induced ocular hypertension (OHT). METHODS UC-MSCs were isolated from human umbilical cords and then cultured. The OHT model was induced via intracameral injection of polystyrene microbeads in Sprague-Dawley adult rat eyes. Fifty-four healthy adult rats were randomly divided into three groups: normal control, OHT model treated with intravitreal transplantation of UC-MSCs, or phosphate-buffered saline (PBS). Two days after OHT was induced, either 5 µl 105 UC-MSCs suspension or PBS was injected into the vitreous cavity of rats. UC-MSCs localization and integration were examined via immunohistochemistry. Neuroprotection was quantified by counting retinal ganglion cells (RGCs) and axons 2 weeks following transplantation. The expression levels of glial-derived neurotrophic factor (GDNF), brain-derived neurotrophic factor (BDNF), and glial fibrillary acidic protein (GFAP) were assessed via immunohistochemistry and Western blot. Functional recovery was assessed 2 weeks after transplantation via scotopic threshold response (STR) electroretinography. RESULTS Elevated IOP levels were sustained at least 3 weeks after intracameral microbead injection and the number of β-III-tubulin+ RGCs significantly declined compared to PBS-injected eyes. UC-MSCs survived for at least 2 weeks after intravitreal transplantation and predominantly located in the vitreous cavity. A fraction of cells migrated into the ganglion cell layer of host retina, but without differentiation. Intravitreal UC-MSC transplantation resulted in increased number of RGCs, axons, and increased expression of GDNF and BDNF but decreased expression of GFAP. Intravitreal delivery of UC-MSCs significantly improved the recovery of the positive STR. CONCLUSIONS Intravitreal transplantation of UC-MSCs revealed the neuroprotection in the microbead-injection induced OHT. The effects could be related to the secretion of tropic factors (BDNF and GDNF) and the modulation of glial cell activation.
Collapse
Affiliation(s)
- Shangli Ji
- a Aier School of Ophthalmology , Central South University , Changsha , Hunan , China
| | - Saiyue Lin
- b Department of Anatomy and Neurobiology, Xiangya School of Medicine , Central South University , Changsha , Hunan , China
| | - Jiansu Chen
- a Aier School of Ophthalmology , Central South University , Changsha , Hunan , China
| | - Xinping Huang
- c Department of Biology , ShenzhenHornetcorn Biotechnology Co., Ltd , Shenzhen , Guangdong , China
| | - Chih-Chang Wei
- c Department of Biology , ShenzhenHornetcorn Biotechnology Co., Ltd , Shenzhen , Guangdong , China
| | - Zhiyuan Li
- b Department of Anatomy and Neurobiology, Xiangya School of Medicine , Central South University , Changsha , Hunan , China
| | - Shibo Tang
- a Aier School of Ophthalmology , Central South University , Changsha , Hunan , China
| |
Collapse
|
22
|
Smith CA, Chauhan BC. In vivo imaging of adeno-associated viral vector labelled retinal ganglion cells. Sci Rep 2018; 8:1490. [PMID: 29367685 PMCID: PMC5784170 DOI: 10.1038/s41598-018-19969-9] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2017] [Accepted: 01/11/2018] [Indexed: 01/10/2023] Open
Abstract
A defining characteristic of optic neuropathies, such as glaucoma, is progressive loss of retinal ganglion cells (RGCs). Current clinical tests only provide weak surrogates of RGC loss, but the possibility of optically visualizing RGCs and quantifying their rate of loss could represent a radical advance in the management of optic neuropathies. In this study we injected two different adeno-associated viral (AAV) vector serotypes in the vitreous to enable green fluorescent protein (GFP) labelling of RGCs in wild-type mice for in vivo and non-invasive imaging. GFP-labelled cells were detected by confocal scanning laser ophthalmoscopy 1-week post-injection and plateaued in density at 4 weeks. Immunohistochemical analysis 5-weeks post-injection revealed labelling specificity to RGCs to be significantly higher with the AAV2-DCX-GFP vector compared to the AAV2-CAG-GFP vector. There were no adverse functional or structural effects of the labelling method as determined with electroretinography and optical coherence tomography, respectively. The RGC-specific positive and negative scotopic threshold responses had similar amplitudes between control and experimental eyes, while inner retinal thickness was also unchanged after injection. As a positive control experiment, optic nerve transection resulted in a progressive loss of labelled RGCs. AAV vectors provide strong and long-lasting GFP labelling of RGCs without detectable adverse effects.
Collapse
Affiliation(s)
- Corey A Smith
- Department of Physiology and Biophysics, Dalhousie University, 5850 College Street, PO Box 15000, Halifax, Nova Scotia, B3H 4R2, Canada.,Retina and Optic Nerve Research Laboratory, Dalhousie University, 5850 College Street, PO Box 15000, Halifax, Nova Scotia, B3H 4R2, Canada
| | - Balwantray C Chauhan
- Department of Physiology and Biophysics, Dalhousie University, 5850 College Street, PO Box 15000, Halifax, Nova Scotia, B3H 4R2, Canada. .,Retina and Optic Nerve Research Laboratory, Dalhousie University, 5850 College Street, PO Box 15000, Halifax, Nova Scotia, B3H 4R2, Canada. .,Department of Ophthalmology and Visual Sciences, Dalhousie University, 1276 South Park Street, 2W Victoria, Halifax, Nova Scotia, B3H 2Y9, Canada.
| |
Collapse
|
23
|
Choh V, Gurdita A, Tan B, Feng Y, Bizheva K, McCulloch DL, Joos KM. Isoflurane and ketamine:xylazine differentially affect intraocular pressure-associated scotopic threshold responses in Sprague-Dawley rats. Doc Ophthalmol 2017. [PMID: 28638951 DOI: 10.1007/s10633-017-9597-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
PURPOSE Amplitudes of electroretinograms (ERG) are enhanced during acute, moderate elevation of intraocular pressure (IOP) in rats anaesthetised with isoflurane. As anaesthetics alone are known to affect ERG amplitudes, the present study compares the effects of inhalant isoflurane and injected ketamine:xylazine on the scotopic threshold response (STR) in rats with moderate IOP elevation. METHODS Isoflurane-anaesthetised (n = 9) and ketamine:xylazine-anaesthetised (n = 6) rats underwent acute unilateral IOP elevation using a vascular loop anterior to the equator of the right eye. STRs to a luminance series (subthreshold to -3.04 log scotopic cd s/m2) were recorded from each eye of Sprague-Dawley rats before, during, and after IOP elevation. RESULTS Positive STR (pSTR) amplitudes for all conditions were significantly smaller (p = 0.0001) for isoflurane- than for ketamine:xylazine-anaesthetised rats. In addition, ketamine:xylazine was associated with a progressive increase in pSTR amplitudes over time (p = 0.0028). IOP elevation was associated with an increase in pSTR amplitude (both anaesthetics p < 0.0001). The absolute interocular differences in IOP-associated enhancement of pSTR amplitudes for ketamine:xylazine and isoflurane were similar (66.3 ± 35.5 vs. 54.2 ± 24.1 µV, respectively). However, the fold increase in amplitude during IOP elevation was significantly higher in the isoflurane- than in the ketamine:xylazine-anaesthetised rats (16.8 ± 29.7x vs. 2.1 ± 2.7x, respectively, p = 0.0004). CONCLUSIONS The anaesthetics differentially affect the STRs in the rat model with markedly reduced amplitudes with isoflurane compared to ketamine:xylazine. However, the IOP-associated enhancement is of similar absolute magnitude for the two anaesthetics, suggesting that IOP stress and anaesthetic effects operate on separate retinal mechanisms.
Collapse
Affiliation(s)
- Vivian Choh
- School of Optometry and Vision Science, University of Waterloo, Waterloo, ON, N2L 3G1, Canada.
| | - Akshay Gurdita
- School of Optometry and Vision Science, University of Waterloo, Waterloo, ON, N2L 3G1, Canada
| | - Bingyao Tan
- Department of Physics and Astronomy, University of Waterloo, Waterloo, ON, N2L 3G1, Canada
| | - Yunwei Feng
- School of Optometry and Vision Science, University of Waterloo, Waterloo, ON, N2L 3G1, Canada
| | - Kostadinka Bizheva
- School of Optometry and Vision Science, University of Waterloo, Waterloo, ON, N2L 3G1, Canada.,Department of Physics and Astronomy, University of Waterloo, Waterloo, ON, N2L 3G1, Canada
| | - Daphne L McCulloch
- School of Optometry and Vision Science, University of Waterloo, Waterloo, ON, N2L 3G1, Canada
| | - Karen M Joos
- Vanderbilt Eye Institute, Vanderbilt University Medical Center, Nashville, TN, USA
| |
Collapse
|
24
|
Test–retest reliability of scotopic full-field electroretinograms in rabbits. Doc Ophthalmol 2017; 134:157-165. [DOI: 10.1007/s10633-017-9582-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2016] [Accepted: 03/01/2017] [Indexed: 10/20/2022]
|
25
|
Leinonen H, Lipponen A, Gurevicius K, Tanila H. Normal Amplitude of Electroretinography and Visual Evoked Potential Responses in AβPP/PS1 Mice. J Alzheimers Dis 2016; 51:21-6. [PMID: 26836173 DOI: 10.3233/jad-150798] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Alzheimer's disease has been shown to affect vision in human patients and animal models. This may pose the risk of bias in behavior studies and therefore requires comprehensive investigation. We recorded electroretinography (ERG) under isoflurane anesthesia and visual evoked potentials (VEP) in awake amyloid expressing AβPPswe/PS1dE9 (AβPP/PS1) and wild-type littermate mice at a symptomatic age. The VEPs in response to patterned stimuli were normal in AβPP/PS1 mice. They also showed normal ERG amplitude but slightly shortened ERG latency in dark-adapted conditions. Our results indicate subtle changes in visual processing in aged male AβPP/PS1 mice specifically at a retinal level.
Collapse
|
26
|
Smith BJ, Côté PD, Tremblay F. Contribution of Na v1.8 sodium channels to retinal function. Neuroscience 2016; 340:279-290. [PMID: 27984182 DOI: 10.1016/j.neuroscience.2016.10.054] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2016] [Revised: 10/07/2016] [Accepted: 10/21/2016] [Indexed: 12/28/2022]
Abstract
We examined the contribution of the sodium channel isoform Nav1.8 to retinal function using the specific blocker A803467. We found that A803467 has little influence on the electroretinogram (ERG) a- and b-waves, but significantly reduces the oscillatory potentials (OPs) to 40-60% of their original amplitude, with significant changes in implicit time in the rod-driven range. To date, only two cell types were found in mouse to express Nav1.8; the starburst amacrine cells (SBACs), and a subtype of retinal ganglion cells (RGCs). When we recorded light responses from ganglion cells using a multielectrode array we found significant and opposing changes in two physiological groups of RGCs. ON-sustained cells showed significant decreases while transient ON-OFF cells showed significant increases. The effects on ON-OFF transient cells but not ON-sustained cells disappeared in the presence of an inhibitory cocktail. We have previously shown that RGCs have only a minor contribution to the OPs (Smith et al., 2014), therefore suggesting that SBACs might be a significant contributor to this ERG component. Targeting SBACs with the cholinergic neurotoxin ethylcholine mustard aziridinium (AF64A) caused a reduction in the amplitude of the OPs similar to A803467. Our results, both using the ERG and MEA recordings from RGCs, suggest that Nav1.8 plays a role in modulating specific aspects of the retinal physiology and that SBACs are a fundamental cellular contributor to the OPs in mice, a clear demonstration of the dichotomy between ERG b-wave and OPs.
Collapse
Affiliation(s)
- Benjamin J Smith
- Department of Biology, Dalhousie University, 1355 Oxford St., PO Box 15000, Halifax, NS B3H 4R2, Canada.
| | - Patrice D Côté
- Department of Biology, Dalhousie University, 1355 Oxford St., PO Box 15000, Halifax, NS B3H 4R2, Canada; Department of Ophthalmology and Visual Sciences, Dalhousie University, 1276 South Park St., PO Box 15000, Halifax, NS B3H 4R2, Canada.
| | - François Tremblay
- Department of Ophthalmology and Visual Sciences, Dalhousie University, 1276 South Park St., PO Box 15000, Halifax, NS B3H 4R2, Canada; Department of Physiology and Biophysics, Dalhousie University, 5850 College Street, PO Box 15000, Halifax, NS B3H 4R2, Canada.
| |
Collapse
|
27
|
Mead B, Tomarev S. Evaluating retinal ganglion cell loss and dysfunction. Exp Eye Res 2016; 151:96-106. [PMID: 27523467 DOI: 10.1016/j.exer.2016.08.006] [Citation(s) in RCA: 78] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2016] [Revised: 08/09/2016] [Accepted: 08/10/2016] [Indexed: 12/16/2022]
Abstract
Retinal ganglion cells (RGC) bear the sole responsibility of propagating visual stimuli to the brain. Their axons, which make up the optic nerve, project from the retina to the brain through the lamina cribrosa and in rodents, decussate almost entirely at the optic chiasm before synapsing at the superior colliculus. For many traumatic and degenerative ocular conditions, the dysfunction and/or loss of RGC is the primary determinant of visual loss and are the measurable endpoints in current research into experimental therapies. To actually measure these endpoints in rodent models, techniques must ascertain both the quantity of surviving RGC and their functional capacity. Quantification techniques include phenotypic markers of RGC, retrogradely transported fluorophores and morphological measurements of retinal thickness whereas functional assessments include electroretinography (flash and pattern) and visual evoked potential. The importance of the accuracy and reliability of these techniques cannot be understated, nor can the relationship between RGC death and dysfunction. The existence of up to 30 types of RGC complicates the measuring process, particularly as these may respond differently to disease and treatment. Since the above techniques may selectively identify and ignore particular subpopulations, their appropriateness as measures of RGC survival and function may be further limited. This review discusses the above techniques in the context of their subtype specificity.
Collapse
Affiliation(s)
- Ben Mead
- Section of Retinal Ganglion Cell Biology, Laboratory of Retinal Cell and Molecular Biology, National Eye Institute, National Institutes of Health, Bethesda, MD, 20892, USA.
| | - Stanislav Tomarev
- Section of Retinal Ganglion Cell Biology, Laboratory of Retinal Cell and Molecular Biology, National Eye Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| |
Collapse
|
28
|
Yukita M, Omodaka K, Machida S, Yasuda M, Sato K, Maruyama K, Nishiguchi KM, Nakazawa T. Brimonidine Enhances the Electrophysiological Response of Retinal Ganglion Cells through the Trk-MAPK/ERK and PI3K Pathways in Axotomized Eyes. Curr Eye Res 2016; 42:125-133. [PMID: 27314704 DOI: 10.3109/02713683.2016.1153112] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
PURPOSE To investigate changes in retinal ganglion cell (RGC) activity by measuring the positive scotopic threshold response (pSTR) of the electroretinogram (ERG) in axotomized eyes after brimonidine injection. METHODS In 50 adult Sprague-Dawley rats, the left eye was axotomized and injected with phosphate buffered saline (PBS) or brimonidine and the contralateral right eye was left untreated. Scotopic ERGs were recorded simultaneously from both eyes on days 1, 2, 3, 7, and 10 after the intravitreal injection, and the amplitude of the a- and b-waves and the pSTR were measured. Surviving RGCs in the flat-mounted retinas were counted 10 days after axotomy. In addition to brimonidine, K252a (an inhibitor of tyrosine kinase phosphorylation of the Trk receptors), U0126 (a MAPK/ERK kinase inhibitor), and LY294002 (phosphoinositide 3-kinases [PI3Ks]) were also injected intravitreally into the left eye, and ERGs were recorded using the same protocol. RESULTS The pSTR amplitude increased significantly in the axotomized eyes with brimonidine, to 122.9 ± 5.0%, 161.8 ± 8.3%, and 133.6 ± 8.1% on days 1, 2, and 3 (P < 0.01), respectively, compared to the axotomized eyes treated with PBS (control). The increased pSTR amplitude returned to normal (103.6 ± 6.7%) on day 7, although there were a greater number of surviving RGCs in the treatment groups than in the controls. The intravitreal injection of K252a, U0126, or LY294002 significantly attenuated the increase in pSTR induced by intravitreal brimonidine (P < 0.01). CONCLUSION Intravitreal brimonidine enhanced the survival and electrophysiological activity of the RGCs in rats. The mechanism of this electrophysiological change may involve activation of the Trk-MAPK/ERK and Trk-PI3K signals.
Collapse
Affiliation(s)
- Masayoshi Yukita
- a Department of Ophthalmology , Tohoku University Graduate School of Medicine , Sendai , Japan
| | - Kazuko Omodaka
- a Department of Ophthalmology , Tohoku University Graduate School of Medicine , Sendai , Japan
| | - Shigeki Machida
- b Department of Ophthalmology , Koshigaya Hospital, Dokkyo Medical University , Koshigaya , Japan.,c Department of Ophthalmology , Iwate Medical University School of Medicine , Morioka , Japan
| | - Masayuki Yasuda
- a Department of Ophthalmology , Tohoku University Graduate School of Medicine , Sendai , Japan
| | - Kota Sato
- a Department of Ophthalmology , Tohoku University Graduate School of Medicine , Sendai , Japan
| | - Kazuichi Maruyama
- a Department of Ophthalmology , Tohoku University Graduate School of Medicine , Sendai , Japan
| | - Koji M Nishiguchi
- d Department of Advanced Ophthalmic Medicine , Tohoku University Graduate School of Medicine , Sendai , Japan
| | - Toru Nakazawa
- a Department of Ophthalmology , Tohoku University Graduate School of Medicine , Sendai , Japan.,d Department of Advanced Ophthalmic Medicine , Tohoku University Graduate School of Medicine , Sendai , Japan.,e Department of Retinal Disease Control , Tohoku University Graduate School of Medicine , Sendai , Japan
| |
Collapse
|
29
|
Li X, Gaillard F, Monckton EA, Glubrecht DD, Persad ARL, Moser M, Sauvé Y, Godbout R. Loss of AP-2delta reduces retinal ganglion cell numbers and axonal projections to the superior colliculus. Mol Brain 2016; 9:62. [PMID: 27259519 PMCID: PMC4893287 DOI: 10.1186/s13041-016-0244-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2016] [Accepted: 05/29/2016] [Indexed: 11/10/2022] Open
Abstract
Background AP-2δ is the most divergent member of the Activating Protein-2 (TFAP2) family of transcription factors. AP-2δ is restricted to specific regions of the CNS, including a subset of ganglion cells in the retina. Retinal ganglion cells (RGCs), the only output neurons of the retina, are responsible for transmitting the visual signal to the brain. Results AP-2δ knockout results in loss of Brn3c (Pou4f3) expression in AP-2δ -positive RGCs. While AP-2δ-/- mice have morphologically normal retinas at birth, there is a significant reduction in retinal ganglion cell numbers by P21, after eye opening. Chromatin immunoprecipitation indicates that Brn3c is a target of AP-2δ in the retina. Using fluorochrome-conjugated cholera toxin subunit B to trace ganglion cell axons from the eye to the major visual pathways in the brain, we found 87 % and 32 % decreases in ipsilateral and contralateral projections, respectively, to the superior colliculus in AP-2δ-/- mice. In agreement with anatomical data, visually evoked responses recorded from the brain confirmed that retinal outputs to the brain are compromised. Conclusions AP-2δ is important for the maintenance of ganglion cell numbers in the retina. Loss of AP-2δ alters retinal axonal projections to visual centers of the brain, with ipsilaterial projections to the superior colliculus being the most dramatically affected. Our results have important implications for integration of the visual signal at the superior colliculus. Electronic supplementary material The online version of this article (doi:10.1186/s13041-016-0244-0) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Xiaodong Li
- Department of Oncology, Cross Cancer Institute, University of Alberta, 11560 University Avenue, Edmonton, AB, T6G 1Z2, Canada
| | - Frédéric Gaillard
- Department of Physiology, University of Alberta, 11560 University Avenue, Edmonton, AB, Canada.,Department of Ophthalmology, University of Alberta, 11560 University Avenue, Edmonton, AB, Canada
| | - Elizabeth A Monckton
- Department of Oncology, Cross Cancer Institute, University of Alberta, 11560 University Avenue, Edmonton, AB, T6G 1Z2, Canada
| | - Darryl D Glubrecht
- Department of Oncology, Cross Cancer Institute, University of Alberta, 11560 University Avenue, Edmonton, AB, T6G 1Z2, Canada
| | - Amit R L Persad
- Department of Oncology, Cross Cancer Institute, University of Alberta, 11560 University Avenue, Edmonton, AB, T6G 1Z2, Canada
| | - Markus Moser
- Max-Planck-Institute of Biochemistry, Martinsried, Germany
| | - Yves Sauvé
- Department of Physiology, University of Alberta, 11560 University Avenue, Edmonton, AB, Canada.,Department of Ophthalmology, University of Alberta, 11560 University Avenue, Edmonton, AB, Canada
| | - Roseline Godbout
- Department of Oncology, Cross Cancer Institute, University of Alberta, 11560 University Avenue, Edmonton, AB, T6G 1Z2, Canada.
| |
Collapse
|
30
|
Mao CA, Agca C, Mocko-Strand JA, Wang J, Ullrich-Lüter E, Pan P, Wang SW, Arnone MI, Frishman LJ, Klein WH. Substituting mouse transcription factor Pou4f2 with a sea urchin orthologue restores retinal ganglion cell development. Proc Biol Sci 2016; 283:20152978. [PMID: 26962139 PMCID: PMC4810862 DOI: 10.1098/rspb.2015.2978] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2015] [Accepted: 02/10/2016] [Indexed: 12/11/2022] Open
Abstract
Pou domain transcription factor Pou4f2 is essential for the development of retinal ganglion cells (RGCs) in the vertebrate retina. A distant orthologue of Pou4f2 exists in the genome of the sea urchin (class Echinoidea) Strongylocentrotus purpuratus (SpPou4f1/2), yet the photosensory structure of sea urchins is strikingly different from that of the mammalian retina. Sea urchins have no obvious eyes, but have photoreceptors clustered around their tube feet disc. The mechanisms that are associated with the development and function of photoreception in sea urchins are largely unexplored. As an initial approach to better understand the sea urchin photosensory structure and relate it to the mammalian retina, we asked whether SpPou4f1/2 could support RGC development in the absence of Pou4f2. To answer this question, we replaced genomic Pou4f2 with an SpPou4f1/2 cDNA. In Pou4f2-null mice, retinas expressing SpPou4f1/2 were outwardly identical to those of wild-type mice. SpPou4f1/2 retinas exhibited dark-adapted electroretinogram scotopic threshold responses, indicating functionally active RGCs. During retinal development, SpPou4f1/2 activated RGC-specific genes and in S. purpuratus, SpPou4f2 was expressed in photoreceptor cells of tube feet in a pattern distinct from Opsin4 and Pax6. Our results suggest that SpPou4f1/2 and Pou4f2 share conserved components of a gene network for photosensory development and they maintain their conserved intrinsic functions despite vast morphological differences in mouse and sea urchin photosensory structures.
Collapse
Affiliation(s)
- Chai-An Mao
- Department of Systems Biology, Unit 0950, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Cavit Agca
- Department of Systems Biology, Unit 0950, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | | | - Jing Wang
- College of Optometry, University of Houston, Houston, TX 77204, USA
| | | | - Ping Pan
- Department of Systems Biology, Unit 0950, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Steven W Wang
- Department of Systems Biology, Unit 0950, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Maria Ina Arnone
- Biology and Evolution of Marine Organisms, Stazione Zoologica Anton Dohrn, Naples 80121, Italy
| | - Laura J Frishman
- College of Optometry, University of Houston, Houston, TX 77204, USA
| | - William H Klein
- Department of Systems Biology, Unit 0950, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| |
Collapse
|
31
|
Porciatti V. Electrophysiological assessment of retinal ganglion cell function. Exp Eye Res 2015; 141:164-70. [PMID: 25998495 DOI: 10.1016/j.exer.2015.05.008] [Citation(s) in RCA: 158] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2014] [Revised: 04/17/2015] [Accepted: 05/17/2015] [Indexed: 01/22/2023]
Abstract
The function of retinal ganglion cells (RGCs) can be non-invasively assessed in experimental and genetic models of glaucoma by means of variants of the ERG technique that emphasize the activity of inner retina neurons. The best understood technique is the Pattern Electroretinogram (PERG) in response to contrast-reversing gratings or checkerboards, which selectively depends on the presence of functional RGCs. In glaucoma models, the PERG can be altered before histological loss of RGCs; PERG alterations may be either reversed with moderate IOP lowering or exacerbated with moderate IOP elevation. Under particular luminance-stimulus conditions, the Flash-ERG displays components that may reflect electrical activity originating in the proximal retina and be altered in some experimental glaucoma models (positive Scotopic Threshold response, pSTR; negative Scotopic Threshold Response, nSTR; Photopic Negative Response, PhNR; Oscillatory Potentials, OPs; multifocal ERG, mfERG). It is not yet known which of these components is most sensitive to glaucomatous damage. Electrophysiological assessment of RGC function appears to be a necessary outcome measure in experimental glaucoma models, which complements structural assessment and may even predict it. Neuroprotective strategies could be tested based on enhancement of baseline electrophysiological function that results in improved RGC survival. The use of electrophysiology in glaucoma models may be facilitated by specifically designed instruments that allow high throughput, robust assessment of electrophysiological function.
Collapse
Affiliation(s)
- Vittorio Porciatti
- Bascom Palmer Eye Institute, University of Miami Miller School of Medicine, McKnight Vision Research Center, 1638 NW 10th Ave., Miami, FL 33136, United States.
| |
Collapse
|
32
|
Deletion of myosin VI causes slow retinal optic neuropathy and age-related macular degeneration (AMD)-relevant retinal phenotype. Cell Mol Life Sci 2015; 72:3953-69. [PMID: 25939269 PMCID: PMC4575690 DOI: 10.1007/s00018-015-1913-3] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2014] [Revised: 04/20/2015] [Accepted: 04/23/2015] [Indexed: 11/16/2022]
Abstract
The unconventional myosin VI, a member of the actin-based motor protein family of myosins, is expressed in the retina. Its deletion was previously shown to reduce amplitudes of the a- and b-waves of the electroretinogram. Analyzing wild-type and myosin VI-deficient Snell’s Waltzer mice in more detail, the expression pattern of myosin VI in retinal pigment epithelium, outer limiting membrane, and outer plexiform layer could be linked with differential progressing ocular deficits. These encompassed reduced a-waves and b-waves and disturbed oscillatory potentials in the electroretinogram, photoreceptor cell death, retinal microglia infiltration, and formation of basal laminar deposits. A phenotype comprising features of glaucoma (neurodegeneration) and age-related macular degeneration could thus be uncovered that suggests dysfunction of myosin VI and its variable cargo adaptor proteins for membrane sorting and autophagy, as possible candidate mediators for both disease forms.
Collapse
|
33
|
Yukita M, Machida S, Nishiguchi KM, Tsuda S, Yokoyama Y, Yasuda M, Maruyama K, Nakazawa T. Molecular, anatomical and functional changes in the retinal ganglion cells after optic nerve crush in mice. Doc Ophthalmol 2015; 130:149-56. [PMID: 25560383 DOI: 10.1007/s10633-014-9478-2] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2014] [Accepted: 12/30/2014] [Indexed: 01/11/2023]
Abstract
PURPOSE Optic nerve crush (ONC) and subsequent axonal damage can be used in rodents to study the mechanism of retinal ganglion cell (RGC) degeneration. Here, we examined electroretinograms (ERGs) in post-ONC mice to investigate changes in the positive scotopic threshold response (pSTR). We then compared these changes with molecular and morphological changes to identify early objective biomarkers of RGC dysfunction. METHODS Fifty 12-week-old C57BL/6 mice were included. ONC was used to induce axonal injury in the right eye of each animal, with the left eye used as a control. The expression of the RGC markers Brn3a and Brn3b was measured on days 1, 2, 3, 5 and 7 after ONC with quantitative real-time PCR. ERGs were recorded under dark adaptation with the stimulus intensity increasing from -6.2 to 0.43 log cd-s/m(2) on days 1, 2, 3, 5, 7 and 10 after ONC. The pSTR, a- and b-wave amplitudes were measured. Inner retinal thickness around the optic nerve head was measured with spectral-domain optical coherence tomography on days 0, 2, 5, 7 and 10 after ONC. RESULTS The expression of Brn3a and Brn3b began to significantly decrease on day 1 and day 2, respectively (P < 0.01). The amplitude of the pSTR underwent rapid, significant deterioration on day 3, after which it fell gradually (P < 0.01), while the a- and b-wave amplitudes remained unchanged throughout the experiment. Inner retinal thickness gradually decreased, with the most significant reduction on day 10 (P < 0.01). CONCLUSIONS Decrease in pSTR likely reflected the early loss of RGC function after ONC and that declining expression of RGC-specific genes preceded anatomical and functional changes in the RGCs.
Collapse
Affiliation(s)
- Masayoshi Yukita
- Department of Ophthalmology, Tohoku University Graduate School of Medicine, 1-1 Seiryo, Aoba, Sendai, Miyagi, 980-8574, Japan
| | | | | | | | | | | | | | | |
Collapse
|
34
|
Ragauskas S, Leinonen H, Puranen J, Rönkkö S, Nymark S, Gurevicius K, Lipponen A, Kontkanen O, Puoliväli J, Tanila H, Kalesnykas G. Early retinal function deficit without prominent morphological changes in the R6/2 mouse model of Huntington's disease. PLoS One 2014; 9:e113317. [PMID: 25469887 PMCID: PMC4254453 DOI: 10.1371/journal.pone.0113317] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2014] [Accepted: 10/24/2014] [Indexed: 11/30/2022] Open
Abstract
Huntington’s disease (HD) is an inherited neurodegenerative disorder that primarily affects the medium-size GABAergic neurons of striatum. The R6/2 mouse line is one of the most widely used animal models of HD. Previously the hallmarks of HD-related pathology have been detected in photoreceptors and interneurons of R6/2 mouse retina. Here we aimed to explore the survival of retinal ganglion cells (RGCs) and functional integrity of distinct retinal cell populations in R6/2 mice. The pattern electroretinography (PERG) signal was lost at the age of 8 weeks in R6/2 mice in contrast to the situation in wild-type (WT) littermates. This defect may be attributable to a major reduction in photopic ERG responses in R6/2 mice which was more evident in b- than a-wave amplitudes. At the age of 4 weeks R6/2 mice had predominantly the soluble form of mutant huntingtin protein (mHtt) in the RGC layer cells, whereas the aggregated form of mHtt was found in the majority of those cells from the 12-week-old R6/2 mice and onwards. Retinal astrocytes did not contain mHtt deposits. The total numbers of RGC layer cells, retinal astrocytes as well as optic nerve axons did not differ between 18-week-old R6/2 mice and their WT controls. Our data indicate that mHtt deposition does not cause RGC degeneration or retinal astrocyte loss in R6/2 mice even at a late stage of HD-related pathology. However, due to functional deficits in the rod- and cone-pathways, the R6/2 mice suffer progressive deficits in visual capabilities starting as early as 4 weeks; at 8 weeks there is severe impairment. This should be taken into account in any behavioral testing conducted in R6/2 mice.
Collapse
Affiliation(s)
- Symantas Ragauskas
- Department of Ophthalmology, Institute of Clinical Medicine, School of Medicine, University of Eastern Finland, Kuopio, Finland
- Vilnius Gediminas Technical University, Department of Biochemistry, Vilnius, Lithuania
- State Research Institute for Innovative Medicine, Vilnius, Lithuania
- Experimentica Ltd., Kuopio, Finland
| | - Henri Leinonen
- Department of Neurobiology, A.I. Virtanen Institute, University of Eastern Finland, Kuopio, Finland
| | - Jooseppi Puranen
- Department of Ophthalmology, Institute of Clinical Medicine, School of Medicine, University of Eastern Finland, Kuopio, Finland
- Experimentica Ltd., Kuopio, Finland
| | - Seppo Rönkkö
- Department of Ophthalmology, Institute of Clinical Medicine, School of Medicine, University of Eastern Finland, Kuopio, Finland
| | - Soile Nymark
- Department of Electronics and Communications Engineering, BioMediTech, Tampere University of Technology, Tampere, Finland
| | - Kestutis Gurevicius
- Department of Neurobiology, A.I. Virtanen Institute, University of Eastern Finland, Kuopio, Finland
| | - Arto Lipponen
- Department of Neurobiology, A.I. Virtanen Institute, University of Eastern Finland, Kuopio, Finland
| | | | | | - Heikki Tanila
- Department of Neurobiology, A.I. Virtanen Institute, University of Eastern Finland, Kuopio, Finland
| | - Giedrius Kalesnykas
- Department of Ophthalmology, Institute of Clinical Medicine, School of Medicine, University of Eastern Finland, Kuopio, Finland
- Experimentica Ltd., Kuopio, Finland
- * E-mail:
| |
Collapse
|
35
|
Nashine S, Liu Y, Kim BJ, Clark AF, Pang IH. Role of C/EBP homologous protein in retinal ganglion cell death after ischemia/reperfusion injury. Invest Ophthalmol Vis Sci 2014; 56:221-31. [PMID: 25414185 DOI: 10.1167/iovs.14-15447] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
PURPOSE To investigate the role of C/EBP homologous protein (CHOP), a proapoptotic protein, and the unfolded protein response (UPR) marker that is involved in endoplasmic reticulum (ER) stress-mediated apoptosis in mouse retinal ganglion cell (RGC) death following ischemia/reperfusion (I/R) injury. METHODS Retinal I/R injury was induced in adult C57BL/6J wild-type (WT) and CHOP knockout (Chop(-/-)) mice by raising IOP to 120 mm Hg for 60 minutes. Expression of CHOP and other UPR markers was studied by Western blot and immunohistochemistry. Retinal ganglion cell counts were performed in retinal flat mounts stained with an RGC marker. Retinal ganglion cell function was evaluated by scotopic threshold response (STR) electroretinography. RESULTS In WT mice, retinal CHOP was upregulated by 30% in I/R-injured eyes compared to uninjured eyes 3 days after injury (P < 0.05). Immunohistochemistry confirmed CHOP upregulation specifically in RGCs. CHOP knockout did not affect baseline RGC density or STR amplitude. Ischemia/reperfusion injury decreased RGC densities and STR amplitudes in both WT and Chop(-/-) mice. However, survival of RGCs in I/R-injured Chop(-/-) mouse was 48% higher (P < 0.05) than that in I/R-injured WT mouse 3 days after I/R injury. Similarly, RGC density was significantly higher in Chop(-/-) eyes at 7, 14, and 28 days after I/R injury. Scotopic threshold response amplitudes of Chop(-/-) mice were significantly higher at 3 and 7 days after I/R than those of WT mice. CONCLUSIONS Absence of CHOP partially protects against RGC loss and reduction in retinal function after I/R injury, indicating that CHOP and, thus, ER stress play an important role in RGC apoptosis in retinal I/R injury.
Collapse
Affiliation(s)
- Sonali Nashine
- North Texas Eye Research Institute, University of North Texas Health Science Center, Fort Worth, Texas, United States Department of Cell Biology and Immunology, University of North Texas Health Science Center, Fort Worth, Texas, United States
| | - Yang Liu
- North Texas Eye Research Institute, University of North Texas Health Science Center, Fort Worth, Texas, United States
| | - Byung-Jin Kim
- North Texas Eye Research Institute, University of North Texas Health Science Center, Fort Worth, Texas, United States
| | - Abbot F Clark
- North Texas Eye Research Institute, University of North Texas Health Science Center, Fort Worth, Texas, United States Department of Cell Biology and Immunology, University of North Texas Health Science Center, Fort Worth, Texas, United States
| | - Iok-Hou Pang
- North Texas Eye Research Institute, University of North Texas Health Science Center, Fort Worth, Texas, United States Department of Pharmaceutical Sciences, University of North Texas Health Science Center, Fort Worth, Texas, United States
| |
Collapse
|