1
|
Wong NK, Yip SP, Huang CL. Establishing Functional Retina in a Dish: Progress and Promises of Induced Pluripotent Stem Cell-Based Retinal Neuron Differentiation. Int J Mol Sci 2023; 24:13652. [PMID: 37686457 PMCID: PMC10487913 DOI: 10.3390/ijms241713652] [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: 08/03/2023] [Revised: 08/28/2023] [Accepted: 08/30/2023] [Indexed: 09/10/2023] Open
Abstract
The human eye plays a critical role in vision perception, but various retinal degenerative diseases such as retinitis pigmentosa (RP), glaucoma, and age-related macular degeneration (AMD) can lead to vision loss or blindness. Although progress has been made in understanding retinal development and in clinical research, current treatments remain inadequate for curing or reversing these degenerative conditions. Animal models have limited relevance to humans, and obtaining human eye tissue samples is challenging due to ethical and legal considerations. Consequently, researchers have turned to stem cell-based approaches, specifically induced pluripotent stem cells (iPSCs), to generate distinct retinal cell populations and develop cell replacement therapies. iPSCs offer a novel platform for studying the key stages of human retinogenesis and disease-specific mechanisms. Stem cell technology has facilitated the production of diverse retinal cell types, including retinal ganglion cells (RGCs) and photoreceptors, and the development of retinal organoids has emerged as a valuable in vitro tool for investigating retinal neuron differentiation and modeling retinal diseases. This review focuses on the protocols, culture conditions, and techniques employed in differentiating retinal neurons from iPSCs. Furthermore, it emphasizes the significance of molecular and functional validation of the differentiated cells.
Collapse
Affiliation(s)
- Nonthaphat Kent Wong
- Department of Health Technology and Informatics, The Hong Kong Polytechnic University, Hong Kong, China;
- Centre for Eye and Vision Research (CEVR), Hong Kong Science Park, Hong Kong, China
| | - Shea Ping Yip
- Department of Health Technology and Informatics, The Hong Kong Polytechnic University, Hong Kong, China;
- Centre for Eye and Vision Research (CEVR), Hong Kong Science Park, Hong Kong, China
| | - Chien-Ling Huang
- Department of Health Technology and Informatics, The Hong Kong Polytechnic University, Hong Kong, China;
- Centre for Eye and Vision Research (CEVR), Hong Kong Science Park, Hong Kong, China
| |
Collapse
|
2
|
Subramani M, Van Hook MJ, Ahmad I. Reproducible generation of human retinal ganglion cells from banked retinal progenitor cells: analysis of target recognition and IGF-1-mediated axon regeneration. Front Cell Dev Biol 2023; 11:1214104. [PMID: 37519299 PMCID: PMC10373790 DOI: 10.3389/fcell.2023.1214104] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Accepted: 06/26/2023] [Indexed: 08/01/2023] Open
Abstract
The selective degeneration of retinal ganglion cells (RGCs) is a common feature in glaucoma, a complex group of diseases, leading to irreversible vision loss. Stem cell-based glaucoma disease modeling, cell replacement, and axon regeneration are viable approaches to understand mechanisms underlying glaucomatous degeneration for neuroprotection, ex vivo stem cell therapy, and therapeutic regeneration. These approaches require direct and facile generation of human RGCs (hRGCs) from pluripotent stem cells. Here, we demonstrate a method for rapid generation of hRGCs from banked human pluripotent stem cell-derived retinal progenitor cells (hRPCs) by recapitulating the developmental mechanism. The resulting hRGCs are stable, functional, and transplantable and have the potential for target recognition, demonstrating their suitability for both ex vivo stem cell approaches to glaucomatous degeneration and disease modeling. Additionally, we demonstrate that hRGCs derived from banked hRPCs are capable of regenerating their axons through an evolutionarily conserved mechanism involving insulin-like growth factor 1 and the mTOR axis, demonstrating their potential to identify and characterize the underlying mechanism(s) that can be targeted for therapeutic regeneration.
Collapse
Affiliation(s)
| | | | - Iqbal Ahmad
- Department of Ophthalmology and Visual Science, University of Nebraska Medical Center, Omaha, NE, United States
| |
Collapse
|
3
|
Lambuk L, Mohd Lazaldin MA, Ahmad S, Iezhitsa I, Agarwal R, Uskoković V, Mohamud R. Brain-Derived Neurotrophic Factor-Mediated Neuroprotection in Glaucoma: A Review of Current State of the Art. Front Pharmacol 2022; 13:875662. [PMID: 35668928 PMCID: PMC9163364 DOI: 10.3389/fphar.2022.875662] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Accepted: 04/28/2022] [Indexed: 12/14/2022] Open
Abstract
Retinal ganglion cells (RGCs) are neurons of the visual system that are responsible for transmitting signals from the retina to the brain via the optic nerve. Glaucoma is an optic neuropathy characterized by apoptotic loss of RGCs and degeneration of optic nerve fibers. Risk factors such as elevated intraocular pressure and vascular dysregulation trigger the injury that culminates in RGC apoptosis. In the event of injury, the survival of RGCs is facilitated by neurotrophic factors (NTFs), the most widely studied of which is brain-derived neurotrophic factor (BDNF). Its production is regulated locally in the retina, but transport of BDNF retrogradely from the brain to retina is also crucial. Not only that the interruption of this retrograde transport has been detected in the early stages of glaucoma, but significantly low levels of BDNF have also been detected in the sera and ocular fluids of glaucoma patients, supporting the notion that neurotrophic deprivation is a likely mechanism of glaucomatous optic neuropathy. Moreover, exogenous NTF including BDNF administration was shown reduce neuronal loss in animal models of various neurodegenerative diseases, indicating the possibility that exogenous BDNF may be a treatment option in glaucoma. Current literature provides an extensive insight not only into the sources, transport, and target sites of BDNF but also the intracellular signaling pathways, other pathways that influence BDNF signaling and a wide range of its functions. In this review, the authors discuss the neuroprotective role of BDNF in promoting the survival of RGCs and its possible application as a therapeutic tool to meet the challenges in glaucoma management. We also highlight the possibility of using BDNF as a biomarker in neurodegenerative disease such as glaucoma. Further we discuss the challenges and future strategies to explore the utility of BDNF in the management of glaucoma.
Collapse
Affiliation(s)
- Lidawani Lambuk
- Department of Immunology, School of Medical Sciences, Universiti Sains Malaysia, Kota Bharu, Malaysia
| | | | - Suhana Ahmad
- Department of Immunology, School of Medical Sciences, Universiti Sains Malaysia, Kota Bharu, Malaysia
| | - Igor Iezhitsa
- Department of Pharmacology and Therapeutics, School of Medicine, International Medical University, Kuala Lumpur, Malaysia
- Department of Pharmacology and Bioinformatics, Volgograd State Medical University, Volgograd, Russia
| | - Renu Agarwal
- Department of Pharmacology and Therapeutics, School of Medicine, International Medical University, Kuala Lumpur, Malaysia
| | - Vuk Uskoković
- TardigradeNano LLC, Irvine, CA, United States
- Department of Mechanical Engineering, San Diego State University, San Diego, CA, United States
| | - Rohimah Mohamud
- Department of Immunology, School of Medical Sciences, Universiti Sains Malaysia, Kota Bharu, Malaysia
| |
Collapse
|
4
|
Beros J, Rodger J, Harvey AR. Age Related Response of Neonatal Rat Retinal Ganglion Cells to Reduced TrkB Signaling in vitro and in vivo. Front Cell Dev Biol 2021; 9:671087. [PMID: 34150766 PMCID: PMC8213349 DOI: 10.3389/fcell.2021.671087] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Accepted: 05/12/2021] [Indexed: 01/19/2023] Open
Abstract
During development of retinofugal pathways there is naturally occurring cell death of at least 50% of retinal ganglion cells (RGCs). In rats, RGC death occurs over a protracted pre- and early postnatal period, the timing linked to the onset of axonal ingrowth into central visual targets. Gene expression studies suggest that developing RGCs switch from local to target-derived neurotrophic support during this innervation phase. Here we investigated, in vitro and in vivo, how RGC birthdate affects the timing of the transition from intra-retinal to target-derived neurotrophin dependence. RGCs were pre-labeled with 5-Bromo-2'-Deoxyuridine (BrdU) at embryonic (E) day 15 or 18. For in vitro studies, RGCs were purified from postnatal day 1 (P1) rat pups and cultured with or without: (i) brain derived neurotrophic factor (BDNF), (ii) blocking antibodies to BDNF and neurotrophin 4/5 (NT-4/5), or (iii) a tropomyosin receptor kinase B fusion protein (TrkB-Fc). RGC viability was quantified 24 and 48 h after plating. By 48 h, the survival of purified βIII-tubulin immunopositive E15 but not E18 RGCs was dependent on addition of BDNF to the culture medium. For E18 RGCs, in the absence of exogenous BDNF, addition of blocking antibodies or TrkB-Fc reduced RGC viability at both 24 and 48 h by 25-40%. While this decrease was not significant due to high variance, importantly, each blocking method also consistently reduced complex process expression in surviving RGCs. In vivo, survival of BrdU and Brn3a co-labeled E15 or E18 RGCs was quantified in rats 24 h after P1 or P5 injection into the eye or contralateral superior colliculus (SC) of BDNF and NT-4/5 antibodies, or serum vehicle. The density of E15 RGCs 24 h after P1 or P5 injection of blocking antibodies was reduced after SC but not intraretinal injection. Antibody injections into either site had little obvious impact on viability of the substantially smaller population of E18 RGCs. In summary, most early postnatal RGC death in the rat involves the elimination of early-born RGCs with their survival primarily dependent upon the availability of target derived BDNF during this time. In contrast, late-born RGC survival may be influenced by additional factors, suggesting an association between RGC birthdate and developmental death mechanisms.
Collapse
Affiliation(s)
- Jamie Beros
- School of Biological Sciences, The University of Western Australia, Crawley, WA, Australia
- Perron Institute for Neurological and Translational Science, Nedlands, WA, Australia
| | - Jennifer Rodger
- School of Biological Sciences, The University of Western Australia, Crawley, WA, Australia
- Perron Institute for Neurological and Translational Science, Nedlands, WA, Australia
| | - Alan R Harvey
- Perron Institute for Neurological and Translational Science, Nedlands, WA, Australia
- School of Human Sciences, The University of Western Australia, Crawley, WA, Australia
| |
Collapse
|
5
|
Jassim AH, Cavanaugh M, Shah JS, Willits R, Inman DM. Transcorneal Electrical Stimulation Reduces Neurodegenerative Process in a Mouse Model of Glaucoma. Ann Biomed Eng 2021; 49:858-870. [PMID: 32974756 PMCID: PMC7854493 DOI: 10.1007/s10439-020-02608-8] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Accepted: 09/02/2020] [Indexed: 12/19/2022]
Abstract
Glaucoma is a neurodegenerative disease in which the retinal ganglion cell axons of the optic nerve degenerate concomitant with synaptic changes in the retina, leading finally to death of the retinal ganglion cells (RGCs). Electrical stimulation has been used to improve neural regeneration in a variety of systems, including in diseases of the retina. Therefore, the focus of this study was to investigate whether transcorneal electrical stimulation (TES) in the DBA2/J mouse model of glaucoma could improve retinal or optic nerve pathology and serve as a minimally invasive treatment option. Mice (10 months-old) received 21 sessions of TES over 8 weeks, after which we evaluated RGC number, axon number, and anterograde axonal transport using histology and immunohistochemistry. To gain insight into the mechanism of proposed protection, we also evaluated inflammation by quantifying CD3+ T-cells and Iba1+ microglia; perturbations in metabolism were shown via the ratio pAMPK to AMPK, and changes in trophic support were tested using protein capillary electrophoresis. We found that TES resulted in RGC axon protection, a reduction in inflammatory cells and their activation, improved energy homeostasis, and a reduction of the cell death-associated p75NTR. Collectively, the data indicated that TES maintained axons, decreased inflammation, and increased trophic factor support, in the form of receptor presence and energy homeostasis, suggesting that electrical stimulation impacts several facets of the neurodegenerative process in glaucoma.
Collapse
Affiliation(s)
- Assraa Hassan Jassim
- Department of Pharmaceutical Sciences, Northeast Ohio Medical University, Rootstown, OH, USA
- Department of Basic and Translational Sciences, University of Pennsylvania, Philadelphia, PA, USA
| | - McKay Cavanaugh
- Department of Biomedical Engineering, University of Akron, Akron, OH, USA
| | | | - Rebecca Willits
- Department of Biomedical Engineering, University of Akron, Akron, OH, USA.
- Department of Chemical Engineering, Northeastern University, Boston, MA, USA.
| | - Denise M Inman
- Department of Pharmaceutical Sciences, Northeast Ohio Medical University, Rootstown, OH, USA.
- North Texas Eye Research Institute, UNT-HSC, 3500 Camp Bowie Blvd, Fort Worth, TX, 76107, USA.
| |
Collapse
|
6
|
Ahmad I, Teotia P, Erickson H, Xia X. Recapitulating developmental mechanisms for retinal regeneration. Prog Retin Eye Res 2019; 76:100824. [PMID: 31843569 DOI: 10.1016/j.preteyeres.2019.100824] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2019] [Revised: 12/06/2019] [Accepted: 12/11/2019] [Indexed: 12/18/2022]
Abstract
Degeneration of specific retinal neurons in diseases like glaucoma, age-related macular degeneration, and retinitis pigmentosa is the leading cause of irreversible blindness. Currently, there is no therapy to modify the disease-associated degenerative changes. With the advancement in our knowledge about the mechanisms that regulate the development of the vertebrate retina, the approach to treat blinding diseases through regenerative medicine appears a near possibility. Recapitulation of developmental mechanisms is critical for reproducibly generating cells in either 2D or 3D culture of pluripotent stem cells for retinal repair and disease modeling. It is the key for unlocking the neurogenic potential of Müller glia in the adult retina for therapeutic regeneration. Here, we examine the current status and potential of the regenerative medicine approach for the retina in the backdrop of developmental mechanisms.
Collapse
Affiliation(s)
- Iqbal Ahmad
- Department of Ophthalmology and Visual Science, University of Nebraska Medical Center, Omaha, NE, 68198, USA.
| | - Pooja Teotia
- Department of Ophthalmology and Visual Science, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Helen Erickson
- Department of Ophthalmology and Visual Science, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Xiaohuan Xia
- Center for Translational Neurodegeneration and Regenerative Therapy, Shanghai Tenth People's Hospital Affiliated to Tongji University School of Medicine, Shanghai, 200072, China
| |
Collapse
|
7
|
Alqawlaq S, Flanagan JG, Sivak JM. All roads lead to glaucoma: Induced retinal injury cascades contribute to a common neurodegenerative outcome. Exp Eye Res 2018; 183:88-97. [PMID: 30447198 DOI: 10.1016/j.exer.2018.11.005] [Citation(s) in RCA: 66] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2018] [Revised: 11/02/2018] [Accepted: 11/06/2018] [Indexed: 11/25/2022]
Abstract
Glaucoma describes a distinct optic neuropathy with complex etiology and a variety of associated risk factors, but with similar pathological endpoints. Risk factors such as age, increased intraocular pressure (IOP), low mean arterial pressure, and autoimmune disease, can all be associated with death of retinal ganglion cells (RGCs) and optic nerve head remodeling. Today, IOP management remains the standard of care, even though IOP elevation is not pathognomonic of glaucoma, and patients can continue to lose vision despite effective IOP control. A contemporary view of glaucoma as a complex, neurodegenerative disease has developed, along with the recognition of a need for new disease modifying retinal treatment strategies and improved outcomes. However, the distinction between risk factors triggering the disease process and retinal injury responses is not always clear. In this review, we attempt to distinguish between the various triggers, and their association with subsequent key RGC injury mechanisms. We propose that distinct glaucomatous risk factors result in similar retinal and optic nerve injury cascades, including oxidative and metabolic stress, glial reactivity, and altered inflammatory responses, which induce common molecular signals to induce RGC apoptosis. This organization forms a coherent disease framework and presents conserved targets for therapeutic intervention that are not limited to specific risk factors.
Collapse
Affiliation(s)
- Samih Alqawlaq
- Department of Vision Science, Krembil Research Institute, University Health Network, Toronto, ON, Canada; Vision Science Research Program, Ophthalmology and Vision Science, University of Toronto, Toronto, ON, Canada; Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
| | - John G Flanagan
- School of Optometry and Vision Science Program, University of California at Berkeley, Berkeley, CA, USA
| | - Jeremy M Sivak
- Department of Vision Science, Krembil Research Institute, University Health Network, Toronto, ON, Canada; Vision Science Research Program, Ophthalmology and Vision Science, University of Toronto, Toronto, ON, Canada; Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
8
|
Progress in Gene Therapy to Prevent Retinal Ganglion Cell Loss in Glaucoma and Leber's Hereditary Optic Neuropathy. Neural Plast 2018; 2018:7108948. [PMID: 29853847 PMCID: PMC5954906 DOI: 10.1155/2018/7108948] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2018] [Accepted: 04/04/2018] [Indexed: 12/24/2022] Open
Abstract
The eye is at the forefront of the application of gene therapy techniques to medicine. In the United States, a gene therapy treatment for Leber's congenital amaurosis, a rare inherited retinal disease, recently became the first gene therapy to be approved by the FDA for the treatment of disease caused by mutations in a specific gene. Phase III clinical trials of gene therapy for other single-gene defect diseases of the retina and optic nerve are also currently underway. However, for optic nerve diseases not caused by single-gene defects, gene therapy strategies are likely to focus on slowing or preventing neuronal death through the expression of neuroprotective agents. In addition to these strategies, there has also been recent interest in the potential use of precise genome editing techniques to treat ocular disease. This review focuses on recent developments in gene therapy techniques for the treatment of glaucoma and Leber's hereditary optic neuropathy (LHON). We discuss recent successes in clinical trials for the treatment of LHON using gene supplementation therapy, promising neuroprotective strategies that have been employed in animal models of glaucoma and the potential use of genome editing techniques in treating optic nerve disease.
Collapse
|
9
|
Beros J, Rodger J, Harvey AR. Developmental retinal ganglion cell death and retinotopicity of the murine retinocollicular projection. Dev Neurobiol 2017; 78:51-60. [PMID: 29134765 DOI: 10.1002/dneu.22559] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2017] [Revised: 11/07/2017] [Accepted: 11/10/2017] [Indexed: 12/13/2022]
Abstract
During mammalian visual system development, retinal ganglion cells (RGCs) undergo extensive apoptotic death. In mouse retina, approximately 50% of RGCs present at birth (postnatal day 0; P0) die by P5, at a time when axons innervate central targets such as the superior colliculus (SC). We examined whether RGCs that make short-range axonal targeting errors within the contralateral SC are more likely to be eliminated during the peak period of RGC death (P1-P5), compared with RGCs initially making more accurate retinotopic connections. A small volume (2.3 nL) of the retrograde nucleophilic dye Hoechst 33342 was injected into the superficial left SC of anesthetized neonatal C57Bl/6J mice at P1 (n = 5) or P4 (n = 8), and the contralateral retina wholemounted 12 hr later. Retrogradely labelled healthy and dying (pyknotic) RGCs were identified by morphological criteria and counted. The percentage of pyknotic RGCs was analyzed in relation to distance from the area of highest density RGC labelling, presumed to represent the most topographically accurate population. As expected, pyknotic RGC density at P1 was significantly greater than P4 (p < 0.05). At P4, the density of healthy RGCs 500-750 µm away from the central region was significantly less, although this was not reflected in altered pyknotic rates. However, at P1 there was a trend (p = 0.08) for an increased proportion of pyknotic RGCs, specifically in temporal parts of the retina outside the densely labelled center. Overall, the lack of consistent association between short-range targeting errors and cell death suggests that most postnatal RGC loss is not directly related to topographic accuracy. © 2017 Wiley Periodicals, Inc. Develop Neurobiol 78: 51-60, 2018.
Collapse
Affiliation(s)
- Jamie Beros
- School of Human Sciences, The University of Western Australia, Crawley, Western Australia, 6009, Australia.,School of Biological Sciences, The University of Western Australia, Crawley, Western Australia, 6009, Australia
| | - Jennifer Rodger
- School of Biological Sciences, The University of Western Australia, Crawley, Western Australia, 6009, Australia.,Perron Institute for Neurological and Translational Science, RR Block, QE II Medical Centre, Nedlands, Western Australia, 6009, Australia
| | - Alan R Harvey
- School of Human Sciences, The University of Western Australia, Crawley, Western Australia, 6009, Australia.,Perron Institute for Neurological and Translational Science, RR Block, QE II Medical Centre, Nedlands, Western Australia, 6009, Australia
| |
Collapse
|
10
|
Mysona BA, Zhao J, Bollinger KE. Role of BDNF/TrkB pathway in the visual system: Therapeutic implications for glaucoma. EXPERT REVIEW OF OPHTHALMOLOGY 2016; 12:69-81. [PMID: 28751923 DOI: 10.1080/17469899.2017.1259566] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
INTRODUCTION Neuroprotective therapeutics are needed to treat glaucoma, an optic neuropathy that results in death of retinal ganglion cells (RGCs). AREAS COVERED The BDNF/TrkB pathway is important for RGC survival. Temporal and spatial alterations in the BDNF/TrkB pathway occur in development and in response to acute optic nerve injury and to glaucoma. In animal models, BDNF supplementation is successful at slowing RGC death after acute optic nerve injury and in glaucoma, however, the BDNF/TrkB signaling is not the only pathway supporting long term RGC survival. EXPERT COMMENTARY Much remains to be discovered about the interaction between retrograde, anterograde, and retinal BDNF/TrkB signaling pathways in both neurons and glia. An ideal therapeutic agent for glaucoma likely has several modes of action that target multiple mechanisms of neurodegeneration including the BDNF/TrkB pathway.
Collapse
Affiliation(s)
- B A Mysona
- Augusta University Department of Cellular Biology and Anatomy, James and Jean Culver Vision Discovery Institute. Address: Augusta University Department of Cellular Biology and Anatomy, Health Sciences Campus, 1120 15th Street, Augusta, GA 30912, USA,
| | - J Zhao
- Medical College of Georgia, Department of Ophthalmology at Augusta University, James and Jean Culver Vision Discovery Institute. Address: Medical College of Georgia, Department of Ophthalmology at Augusta University, 1120 15th Street, Augusta, GA 30912, USA,
| | - K E Bollinger
- Medical College of Georgia, Department of Ophthalmology at Augusta University, Augusta University Department of Cellular Biology and Anatomy, James and Jean Culver Vision Discovery Institute. Address: Medical College of Georgia, Department of Ophthalmology at Augusta University, 1120 15th Street, Augusta, GA 30912, USA,
| |
Collapse
|
11
|
Teotia P, Chopra DA, Dravid SM, Van Hook MJ, Qiu F, Morrison J, Rizzino A, Ahmad I. Generation of Functional Human Retinal Ganglion Cells with Target Specificity from Pluripotent Stem Cells by Chemically Defined Recapitulation of Developmental Mechanism. Stem Cells 2016; 35:572-585. [PMID: 27709736 DOI: 10.1002/stem.2513] [Citation(s) in RCA: 57] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2016] [Revised: 09/06/2016] [Accepted: 09/12/2016] [Indexed: 01/07/2023]
Abstract
Glaucoma is a complex group of diseases wherein a selective degeneration of retinal ganglion cells (RGCs) lead to irreversible loss of vision. A comprehensive approach to glaucomatous RGC degeneration may include stem cells to functionally replace dead neurons through transplantation and understand RGCs vulnerability using a disease in a dish stem cell model. Both approaches require the directed generation of stable, functional, and target-specific RGCs from renewable sources of cells, that is, the embryonic stem cells and induced pluripotent stem cells. Here, we demonstrate a rapid and safe, stage-specific, chemically defined protocol that selectively generates RGCs across species, including human, by recapitulating the developmental mechanism. The de novo generated RGCs from pluripotent cells are similar to native RGCs at the molecular, biochemical, functional levels. They also express axon guidance molecules, and discriminate between specific and nonspecific targets, and are nontumorigenic. Stem Cells 2017;35:572-585.
Collapse
Affiliation(s)
- Pooja Teotia
- Department of Ophthalmology and Visual Sciences, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Divyan A Chopra
- Department of Pharmacology, Creighton University, Omaha, Nebraska, USA
| | | | - Matthew J Van Hook
- Department of Ophthalmology and Visual Sciences, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Fang Qiu
- Department of Biostatistics, College of Public Health, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - John Morrison
- Department of Ophthalmology, Casey Eye Institute, Oregon Health & Science University, Portland, Oregon, USA
| | - Angie Rizzino
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Iqbal Ahmad
- Department of Ophthalmology and Visual Sciences, University of Nebraska Medical Center, Omaha, Nebraska, USA
| |
Collapse
|
12
|
Reese BE, Keeley PW. Genomic control of neuronal demographics in the retina. Prog Retin Eye Res 2016; 55:246-259. [PMID: 27492954 DOI: 10.1016/j.preteyeres.2016.07.003] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2016] [Revised: 07/21/2016] [Accepted: 07/28/2016] [Indexed: 12/22/2022]
Abstract
The mature retinal architecture is composed of various types of neuron, each population differing in size and constrained to particular layers, wherein the cells achieve a characteristic patterning in their local organization. These demographic features of retinal nerve cell populations are each complex traits controlled by multiple genes affecting different processes during development, and their genetic determinants can be dissected by correlating variation in these traits with their genomic architecture across recombinant-inbred mouse strains. Using such a resource, we consider how the variation in the numbers of twelve different types of retinal neuron are independent of one another, including those sharing transcriptional regulation as well as those that are synaptically-connected, each mapping to distinct genomic loci. Using the populations of two retinal interneurons, the horizontal cells and the cholinergic amacrine cells, we present in further detail examples where the variation in neuronal number, as well as the variation in mosaic patterning or in laminar positioning, each maps to discrete genomic loci where allelic variants modulating these features must be present. At those loci, we identify candidate genes which, when rendered non-functional, alter those very demographic properties, and in turn, we identify candidate coding or regulatory variants that alter protein structure or gene expression, respectively, being prospective contributors to the variation in phenotype. This forward-genetic approach provides an alternative means for dissecting the molecular genetic control of neuronal population dynamics, with each genomic locus serving as a causal anchor from which we may ultimately understand the developmental principles responsible for the control of those traits.
Collapse
Affiliation(s)
- Benjamin E Reese
- Neuroscience Research Institute, University of California, Santa Barbara, CA 93106-5060, USA; Departments of Psychological & Brain Sciences, University of California, Santa Barbara, CA 93106-9660, USA.
| | - Patrick W Keeley
- Neuroscience Research Institute, University of California, Santa Barbara, CA 93106-5060, USA; Departments of Molecular, Cellular & Developmental Biology, University of California, Santa Barbara, CA 93106-9625, USA
| |
Collapse
|
13
|
Barnstable CJ, Reddy R, Li H, Horvath TL. Mitochondrial Uncoupling Protein 2 (UCP2) Regulates Retinal Ganglion Cell Number and Survival. J Mol Neurosci 2016; 58:461-9. [PMID: 26846222 PMCID: PMC4833669 DOI: 10.1007/s12031-016-0728-5] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2015] [Accepted: 01/27/2016] [Indexed: 12/29/2022]
Abstract
In the brain, mitochondrial uncoupling protein 2 (UCP2) has emerged as a stress signal associated with neuronal survival. In the retina, UCP2 is expressed primarily by retinal ganglion cells. Here, we investigated the functional relevance of UCP2 in the mouse retina. Increased expression of UCP2 significantly reduced apoptosis during the critical developmental period resulting in elevated numbers of retinal ganglion cells in the adult. Elevated UCP2 levels also protected against excitotoxic cell death induced by intraocular injection of either NMDA or kainic acid. In monolayer cultures of retinal cells, elevated UCP2 levels increased cell survival and rendered the cells independent of the survival-promoting effects of the neurotrophic factors BDNF and CNTF. Taken together, these data implicate UCP2 as an important regulator of retinal neuron survival both during development and in adult animals.
Collapse
Affiliation(s)
- Colin J Barnstable
- Department of Ophthalmology and Visual Science, Yale University School of Medicine, New Haven, CT, 06510, USA.
- Department of Neurobiology, Yale University School of Medicine, New Haven, CT, 06510, USA.
- Department of Neural and Behavioral Science, Penn State University College of Medicine, H109, 500 University Drive, Hershey, PA, 17033, USA.
| | - Rajini Reddy
- Department of Ophthalmology and Visual Science, Yale University School of Medicine, New Haven, CT, 06510, USA
| | - Hong Li
- Department of Ophthalmology and Visual Science, Yale University School of Medicine, New Haven, CT, 06510, USA
- Department of Neural and Behavioral Science, Penn State University College of Medicine, H109, 500 University Drive, Hershey, PA, 17033, USA
| | - Tamas L Horvath
- Department of Neurobiology, Yale University School of Medicine, New Haven, CT, 06510, USA
- Obstetrics, Gynecology and Reproductive Sciences, Yale University School of Medicine, New Haven, CT, 06510, USA
- Section of Comparative Medicine, Yale University School of Medicine, New Haven, CT, 06510, USA
| |
Collapse
|
14
|
Doozandeh A, Yazdani S. Neuroprotection in Glaucoma. J Ophthalmic Vis Res 2016; 11:209-20. [PMID: 27413504 PMCID: PMC4926571 DOI: 10.4103/2008-322x.183923] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2015] [Accepted: 08/03/2015] [Indexed: 11/04/2022] Open
Abstract
Glaucoma is a degenerative optic neuropathy characterized by retinal ganglion cell (RGC) loss and visual field defects. It is known that in some glaucoma patients, death of RGCs continues despite intraocular pressure (IOP) reduction. Neuroprotection in the field of glaucoma is defined as any treatment, independent of IOP reduction, which prevents RGC death. Glutamate antagonists, ginkgo biloba extract, neurotrophic factors, antioxidants, calcium channel blockers, brimonidine, glaucoma medications with blood regulatory effect and nitric oxide synthase inhibitors are among compounds with possible neuroprotective activity in preclinical studies. A few agents (such as brimonidine or memantine) with neuroprotective effects in experimental studies have advanced to clinical trials; however the results of clinical trials for these agents have not been conclusive. Nevertheless, lack of compelling clinical evidence has not prevented the off-label use of some of these compounds in glaucoma practice. Stem cell transplantation has been reported to halt experimental neurodegenerative disease processes in the absence of cell replacement. It has been hypothesized that transplantation of some types of stem cells activates multiple neuroprotective pathways via secretion of various factors. The advantage of this approach is a prolonged and targeted effect. Important concerns in this field include the secretion of unwanted harmful mediators, graft survival issues and tumorigenesis. Neuroprotection in glaucoma, pharmacologically or by stem cell transplantation, is an interesting subject waiting for broad and multidisciplinary collaborative studies to better clarify its role in clinical practice.
Collapse
Affiliation(s)
- Azadeh Doozandeh
- Ophthalmic Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Shahin Yazdani
- Ocular Tissue Engineering Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
15
|
Abstract
Glaucoma is a chronic optic neuropathy characterized by progressive damage to the optic nerve, death of retinal ganglion cells and ultimately visual field loss. It is one of the leading causes of irreversible loss of vision worldwide. The most important trigger of glaucomatous damage is elevated eye pressure, and the current standard approach in glaucoma therapy is reduction of intraocular pressure (IOP). However, despite the use of effective medications or surgical treatment leading to lowering of IOP, progression of glaucomatous changes and loss of vision among patients with glaucoma is common. Therefore, it is critical to prevent vision loss through additional treatment. To implement such treatment(s), it is imperative to identify pathophysiological changes in glaucoma and develop therapeutic methods taking into account neuroprotection. Currently, there is no method of neuroprotection with long-term proven effectiveness in the treatment of glaucoma. Among the most promising molecules shown to protect the retina and optic nerve are neurotrophic factors. Thus, the current focus is on the development of safe and non-invasive methods for the long-term elevation of the intraocular level of neurotrophins through advanced gene therapy and topical eye treatment and on the search for selective agonists of neurotrophin receptors affording more efficient neuroprotection.
Collapse
Affiliation(s)
- Anna Wójcik-Gryciuk
- Department of Ophthalmology, MSW Hospital, Warsaw, Poland
- Department of Neurophysiology, Nencki Institute of Experimental Biology, Warsaw, Poland
| | - Małgorzata Skup
- Department of Neurophysiology, Nencki Institute of Experimental Biology, Warsaw, Poland
| | | |
Collapse
|
16
|
The Acquisition of Target Dependence by Developing Rat Retinal Ganglion Cells. eNeuro 2015; 2:eN-NWR-0044-14. [PMID: 26464991 PMCID: PMC4586937 DOI: 10.1523/eneuro.0044-14.2015] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2014] [Revised: 06/22/2015] [Accepted: 06/23/2015] [Indexed: 11/23/2022] Open
Abstract
Similar to neurons in the peripheral nervous system, immature CNS-derived RGCs become dependent on target-derived neurotrophic support as their axons reach termination sites in the brain. To study the factors that influence this developmental transition we took advantage of the fact that rat RGCs are born, and target innervation occurs, over a protracted period of time. Early-born RGCs have axons in the SC by birth (P0), whereas axons from late-born RGCs do not innervate the SC until P4-P5. Birth dating RGCs using EdU allowed us to identify RGCs (1) with axons still growing toward targets, (2) transitioning to target dependence, and (3) entirely dependent on target-derived support. Using laser-capture microdissection we isolated ∼34,000 EdU+ RGCs and analyzed transcript expression by custom qPCR array. Statistical analyses revealed a difference in gene expression profiles in actively growing RGCs compared with target-dependent RGCs, as well as in transitional versus target-dependent RGCs. Prior to innervation RGCs expressed high levels of BDNF and CNTFR α but lower levels of neurexin 1 mRNA. Analysis also revealed greater expression of transcripts for signaling molecules such as MAPK, Akt, CREB, and STAT. In a supporting in vitro study, purified birth-dated P1 RGCs were cultured for 24-48 h with or without BDNF; lack of BDNF resulted in significant loss of early-born but not late-born RGCs. In summary, we identified several important changes in RGC signaling that may form the basis for the switch from target independence to dependence.
Collapse
|
17
|
Soares CA, Mason CA. Transient ipsilateral retinal ganglion cell projections to the brain: Extent, targeting, and disappearance. Dev Neurobiol 2015; 75:1385-401. [PMID: 25788284 DOI: 10.1002/dneu.22291] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2015] [Revised: 03/13/2015] [Accepted: 03/15/2015] [Indexed: 12/27/2022]
Abstract
During development of the mammalian eye, the first retinal ganglion cells (RGCs) that extend to the brain are located in the dorsocentral (DC) retina. These RGCs extend to either ipsilateral or contralateral targets, but the ipsilateral projections do not survive into postnatal periods. The function and means of disappearance of the transient ipsilateral projection are not known. We have followed the course of this transient early ipsilateral cohort of RGCs, paying attention to how far they extend, whether they enter targets and if so, which ones, and the time course of their disappearance. The DC ipsilateral RGC axons were traced using DiI labeling at E13.5 and E15.5 to compare the proportion of ipsi- versus contralateral projections during the first period of growth. In utero electroporation of E12.5 retina with GFP constructs was used to label axons that could be visualized at succeeding time points into postnatal ages. Our results show that the earliest ipsilateral axons grow along the cellular border of the brain, and are segregated from the laterally positioned contralateral axons from the same retinal origin. In agreement with previous reports, although many early RGCs extend ipsilaterally, after E16 their number rapidly declines. Nonetheless, some ipsilateral axons from the DC retina enter the superior colliculus and arborize minimally, but very few enter the dorsal lateral geniculate nucleus and those that do extend only short branches. While the mechanism of selective axonal disappearance remains elusive, these data give further insight into establishment of the visual pathways.
Collapse
Affiliation(s)
- Célia A Soares
- Department of Pathology and Cell Biology, College of Physicians and Surgeons, Columbia University, New York.,Life and Health Science Research Institute, School of Health Sciences, University of Minho, Braga, Portugal.,ICVS/3B's, PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Carol A Mason
- Department of Pathology and Cell Biology, College of Physicians and Surgeons, Columbia University, New York.,Department of Neuroscience, College of Physicians and Surgeons, Columbia University, New York.,Department of Ophthalmology, College of Physicians and Surgeons, Columbia University, New York
| |
Collapse
|
18
|
Liu Y, Xu X, Tang R, Chen G, Lei X, Gao L, Li W, Chen Y. Viability of primary cultured retinal neurons in a hyperglycemic condition. Neural Regen Res 2014; 8:410-9. [PMID: 25206682 PMCID: PMC4146128 DOI: 10.3969/j.issn.1673-5374.2013.05.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2012] [Accepted: 01/10/2013] [Indexed: 12/21/2022] Open
Abstract
The retina of Wistar rats within 1-3 days of birth were dissociated into a retinal cell suspension using 0.05% trypsin digestion. The cell suspension was incubated in Dulbecco's modified Eagle's medium for 24 hours, followed by neurobasal medium for 5-7 days. Nissl staining showed that 79.86% of primary cultured retinal cells were positive and immunocytochemical staining showed that the purity of anti-neurofilament heavy chain antibody-positive cells was 71.53%, indicating that the primary culture system of rat retinal neurons was a reliable and stable cell system with neurons as the predominant cell type. The primary cultured retinal neurons were further treated with 0, 5.5, 15, 25, and 35 mM glucose for 24, 48, and 72 hours. The thiazolyl blue tetrazolium bromide test and flow cytometry showed that with increasing glucose concentration and treatment duration, the viability of retinal neurons was reduced, and apoptosis increased. In particular, 35 mM glucose exhibited the most significant effect at 72 hours. Thus, rat retinal neurons treated with 35 mM glucose for 72 hours can be used to simulate a neuronal model of diabetic retinopathy.
Collapse
Affiliation(s)
- Yu Liu
- Department of Ophthalmology, Third Xiangya Hospital of Central South University, Changsha 410013, Hunan Province, China
| | - Xueliang Xu
- Department of Ophthalmology, Xiangya Hospital of Central South University, Changsha 410008, Hunan Province, China
| | - Renhong Tang
- Department of Ophthalmology, Third Xiangya Hospital of Central South University, Changsha 410013, Hunan Province, China
| | - Guoping Chen
- Department of Ophthalmology, Third Xiangya Hospital of Central South University, Changsha 410013, Hunan Province, China
| | - Xiang Lei
- Department of Ophthalmology, Third Xiangya Hospital of Central South University, Changsha 410013, Hunan Province, China
| | - Limo Gao
- Department of Ophthalmology, Third Xiangya Hospital of Central South University, Changsha 410013, Hunan Province, China
| | - Wenjie Li
- Department of Ophthalmology, Third Xiangya Hospital of Central South University, Changsha 410013, Hunan Province, China
| | - Yu Chen
- Department of Ophthalmology, Third Xiangya Hospital of Central South University, Changsha 410013, Hunan Province, China
| |
Collapse
|
19
|
Weber AJ, Harman CD. BDNF treatment and extended recovery from optic nerve trauma in the cat. Invest Ophthalmol Vis Sci 2013; 54:6594-604. [PMID: 23989190 DOI: 10.1167/iovs.13-12683] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
PURPOSE We examined the treatment period necessary to restore retinal and visual stability following trauma to the optic nerve. METHODS Cats received unilateral optic nerve crush and no treatment (NT), treatment of the injured eye with brain-derived neurotrophic factor (BDNF), or treatment of the injured eye combined with treatment of visual cortex for 2 or 4 weeks. After 1-, 2-, 4-, or 6-week survival periods, pattern electroretinograms (PERGs) were obtained and retinal ganglion cell (RGC) survival determined. RESULTS In the peripheral retina, RGC survival for NT, eye only, and eye + cortex animals was 55%, 78%, and 92%, respectively, at 1 week, and 31%, 60%, and 93%, respectively, at 2 weeks. PERGs showed a similar pattern of improvement. After 4 weeks, RGC survival was 7%, 29%, and 53% in each group, with PERGs in the dual-treated animals similar to the 1- to 2-week animals. For area centralis (AC), the NT, eye only, and eye + cortex animals showed 47%, 78%, and 82% survival, respectively, at 2 weeks, and 13%, 54%, and 81% survival, respectively, at 4 weeks. Removing the pumps at 2 weeks resulted in ganglion cell survival levels of 76% and 74% in the AC at 4 and 6 weeks postcrush, respectively. The PERGs from 2-week treated, but 4- and 6-week survival animals were comparable to those of the 2-week animals. CONCLUSIONS Treating the entire central visual pathway is important following optic nerve trauma. Long-term preservation of central vision may be achieved with as little as 2 weeks of treatment using this approach.
Collapse
Affiliation(s)
- Arthur J Weber
- Department of Physiology, Neuroscience Training Program, Michigan State University, East Lansing, Michigan
| | | |
Collapse
|
20
|
Lin B, Peng EB. Retinal ganglion cells are resistant to photoreceptor loss in retinal degeneration. PLoS One 2013; 8:e68084. [PMID: 23840814 PMCID: PMC3695938 DOI: 10.1371/journal.pone.0068084] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2013] [Accepted: 05/24/2013] [Indexed: 01/19/2023] Open
Abstract
The rapid and massive degeneration of photoreceptors in retinal degeneration might have a dramatic negative effect on retinal circuits downstream of photoreceptors. However, the impact of photoreceptor loss on the morphology and function of retinal ganglion cells (RGCs) is not fully understood, precluding the rational design of therapeutic interventions that can reverse the progressive loss of retinal function. The present study investigated the morphological changes in several identified RGCs in the retinal degeneration rd1 mouse model of retinitis pigmentosa (RP), using a combination of viral transfection, microinjection of neurobiotin and confocal microscopy. Individual RGCs were visualized with a high degree of detail using an adeno-associated virus (AAV) vector carrying the gene for enhanced green fluorescent protein (EGFP), allowed for large-scale surveys of the morphology of RGCs over a wide age range. Interestingly, we found that the RGCs of nine different types we encountered were especially resistant to photoreceptor degeneration, and retained their fine dendritic geometry well beyond the complete death of photoreceptors. In addition, the RGC-specific markers revealed a remarkable degree of stability in both morphology and numbers of two identified types of RGCs for up to 18 months of age. Collectively, our data suggest that ganglion cells, the only output cells of the retina, are well preserved morphologically, indicating the ganglion cell population might be an attractive target for treating vision loss.
Collapse
Affiliation(s)
- Bin Lin
- Department of Anatomy, The University of Hong Kong, Li Ka Shing Faculty of Medicine, Pokfulam, Hong Kong.
| | | |
Collapse
|
21
|
Fan Q, Huang WB, Zhang XL. TRPC6: an underlying target for human glaucoma. Int J Ophthalmol 2012; 5:523-6. [PMID: 22937518 DOI: 10.3980/j.issn.2222-3959.2012.04.23] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2012] [Accepted: 07/20/2012] [Indexed: 11/02/2022] Open
Abstract
Glaucoma is one of the leading causes of visual impairment and blindness worldwide. Of known risk factors for glaucoma, an increased in intraocular pressure is most highly correlated with glaucomatous damage. Irrespective of the cause, apoptosis of the retinal ganglion cells is the eventual outcome. It is widely accepted that glaucoma is a neurodegenerative disease that is strongly correlated with central nervous system disorders, such as Alzheimer's disease. These two disorders also share some similarities in pathogenic mechanisms. Recent studies suggest that the transient receptor potential canonical 6 channel could work together with brain-derived neurotrophic factor to promote neuron survival in brain and retina. In this study, we propose that transient receptor potential canonical 6 may contribute to the pathogenesis of human glaucoma and become a potential therapeutic target.
Collapse
Affiliation(s)
- Qian Fan
- Zhongshan Ophthalmic Center, State Key Laboratory of Ophthalmology, Sun Yat-sen University, Guangzhou 510060, Guangdong Province, China
| | | | | |
Collapse
|
22
|
Voyatzis S, Muzerelle A, Gaspar P, Nicol X. Modeling activity and target-dependent developmental cell death of mouse retinal ganglion cells ex vivo. PLoS One 2012; 7:e31105. [PMID: 22363559 PMCID: PMC3281910 DOI: 10.1371/journal.pone.0031105] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2011] [Accepted: 01/02/2012] [Indexed: 12/20/2022] Open
Abstract
Programmed cell death is widespread during the development of the central nervous system and serves multiple purposes including the establishment of neural connections. In the mouse retina a substantial reduction of retinal ganglion cells (RGCs) occurs during the first postnatal week, coinciding with the formation of retinotopic maps in the superior colliculus (SC). We previously established a retino-collicular culture preparation which recapitulates the progressive topographic ordering of RGC projections during early post-natal life. Here, we questioned whether this model could also be suitable to examine the mechanisms underlying developmental cell death of RGCs. Brn3a was used as a marker of the RGCs. A developmental decline in the number of Brn3a-immunolabelled neurons was found in the retinal explant with a timing that paralleled that observed in vivo. In contrast, the density of photoreceptors or of starburst amacrine cells increased, mimicking the evolution of these cell populations in vivo. Blockade of neural activity with tetrodotoxin increased the number of surviving Brn3a-labelled neurons in the retinal explant, as did the increase in target availability when one retinal explant was confronted with 2 or 4 collicular slices. Thus, this ex vivo model reproduces the developmental reduction of RGCs and recapitulates its regulation by neural activity and target availability. It therefore offers a simple way to analyze developmental cell death in this classic system. Using this model, we show that ephrin-A signaling does not participate to the regulation of the Brn3a population size in the retina, indicating that eprhin-A-mediated elimination of exuberant projections does not involve developmental cell death.
Collapse
Affiliation(s)
- Sylvie Voyatzis
- Institut National de la Santé et de la Recherche Médicale, Unité mixte de Recherche en Santé 839, Paris, France
- Université Pierre et Marie Curie, Paris, France
- Institut du Fer à Moulin, Paris, France
| | - Aude Muzerelle
- Institut National de la Santé et de la Recherche Médicale, Unité mixte de Recherche en Santé 839, Paris, France
- Université Pierre et Marie Curie, Paris, France
- Institut du Fer à Moulin, Paris, France
| | - Patricia Gaspar
- Institut National de la Santé et de la Recherche Médicale, Unité mixte de Recherche en Santé 839, Paris, France
- Université Pierre et Marie Curie, Paris, France
- Institut du Fer à Moulin, Paris, France
| | - Xavier Nicol
- Institut National de la Santé et de la Recherche Médicale, Unité mixte de Recherche en Santé 839, Paris, France
- Université Pierre et Marie Curie, Paris, France
- Institut du Fer à Moulin, Paris, France
| |
Collapse
|
23
|
Neurotrophic factors and the regeneration of adult retinal ganglion cell axons. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2012; 106:1-33. [PMID: 23211458 DOI: 10.1016/b978-0-12-407178-0.00002-8] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The adult central nervous system (CNS) has only a limited capacity to regenerate axons after injury. This is due to a number of factors including the presence of extrinsic inhibitory factors that limit plasticity, lack of effective trophic support, and intrinsic changes in neuronal responsiveness. In this review, we describe the expression and role of neurotrophins in retinal ganglion cells (RGCs) during development and adulthood, and the receptors and miscellaneous signaling systems that influence axonal regeneration after injury. The impact of exogenous neurotrophic factors on adult RGCs injured at different sites in the visual pathway is described for several modes of delivery, including recombinant factors, viral vectors, cell transplantation, as well as combinatorial treatments involving other pharmacotherapeutic agents. Indirect, off-target effects of neurotrophic factors on RGC axonal regeneration are also considered. There remain unresolved issues relating to optimal delivery of neurotrophic factors, and we emphasize the need to develop safe, reliable methods for the regulation of exogenous supply of these factors to the injured CNS.
Collapse
|
24
|
Dallimore EJ, Park KK, Pollett MA, Taylor JS, Harvey AR. The life, death and regenerative ability of immature and mature rat retinal ganglion cells are influenced by their birthdate. Exp Neurol 2010; 225:353-65. [DOI: 10.1016/j.expneurol.2010.07.007] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2010] [Revised: 06/30/2010] [Accepted: 07/12/2010] [Indexed: 11/17/2022]
|
25
|
Trophic responsiveness of purified postnatal and adult rat retinal ganglion cells. Cell Tissue Res 2010; 339:297-310. [PMID: 19936794 DOI: 10.1007/s00441-009-0897-4] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2009] [Accepted: 10/09/2009] [Indexed: 12/17/2022]
Abstract
Central neurons lose the ability for axonal re-growth during development and typically do not regenerate their axons following axotomy once they become mature unless given a growth-permissive environment i.e. peripheral nerve graft. In the present study, the growth responsiveness of purified retinal ganglion cells (RGCs) at different ages to neurotrophic factors and Schwann cell (SC)-secreted factors were examined directly. The purity of adult RGCs was 97% as assessed by retrograde labelling with 4,6-diamidino-2-phenylindole. The stability of cultures were demonstrated by long-term survival (30 days) in medium contained brain-derived neurotrophic factor (BDNF), ciliary neurotrophic factor (CNTF) and forskolin (F) (BCF). RGCs from postnatal (P) (P0, P4, P8, P21) and adult (P90) rats showed decreasing levels of survival and neuritogenesis when grown in BCF. In contrast, the opposite was observed in SC-conditioned medium (CM)-treated P0-P8 RGCs which were increasingly responsive. SCCM induced maximal neurite outgrowth in P8 RGCs via the activation of extracellular regulated kinase 1/2 (Erk1/2). Inhibition of mitogen-activated protein kinase-Erk1/2 signaling using an Erk1/2-specific inhibitor (UO126) abolished SCCM-induced Erk1/2 phosphorylation and neuritogenesis completely. Although both SCCM and BCF failed to sustain the same levels of growth in P21 or P90 cultures as observed in P8 cultures, SCCM promoted higher survival and neuritogenesis than BCF-treated adult RGCs. This study is the first report of adult rat RGC purification and demonstrates that mature RGCs need multiple factors for survival and neurite outgrowth.
Collapse
|
26
|
Barnstable CJ. Mitochondria and the regulation of free radical damage in the eye. J Ocul Biol Dis Infor 2009; 2:145-148. [PMID: 20046847 PMCID: PMC2798985 DOI: 10.1007/s12177-009-9036-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2009] [Accepted: 09/03/2009] [Indexed: 01/15/2023] Open
Abstract
Neuronal cell death can be determined by the overall level of reactive oxygen species (ROS) resulting from the combination of extrinsic sources and intrinsic production as a byproduct of oxidative phosphorylation. Key controllers of the intrinsic production of ROS are the mitochondrial uncoupling proteins (UCPs). By allowing a controlled leak of protons across the inner mitochondrial membrane activation of these proteins can decrease ROS and promote cell survival. In both primate models of Parkinson's disease and mouse models of seizures, increased activity of UCP2 significantly increased neuronal cells survival. In the retina UCP2 is expressed in many neurons and glial cells, but was not detected in rod photoreceptors. Retinal ganglion cell survival following excitotoxic damage was much greater in animals overexpressing UCP2. Traditional Chinese medicines, such as an extract of Cistanche tubulosa, may provide benefit by altering mitochondrial metabolism.
Collapse
Affiliation(s)
- Colin J. Barnstable
- Department of Neural and Behavioral Sciences, Penn State University College of Medicine, Hershey, PA 17033 USA
| |
Collapse
|
27
|
Guo Y, Johnson E, Cepurna W, Jia L, Dyck J, Morrison JC. Does elevated intraocular pressure reduce retinal TRKB-mediated survival signaling in experimental glaucoma? Exp Eye Res 2009; 89:921-33. [PMID: 19682984 DOI: 10.1016/j.exer.2009.08.003] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2009] [Revised: 06/16/2009] [Accepted: 08/02/2009] [Indexed: 12/11/2022]
Abstract
Reduced retrograde transport of neurotrophins (NT) and their receptors has been hypothesized to contribute directly to retinal ganglion cell (RGC) loss in glaucoma. However, strategies of supplementing NT and NT receptors have failed to avert ultimate RGC death in experimental glaucoma. This study examines the response of major components of the NT system and their interacting proteins in a rat glaucoma model. Unilateral chronic intraocular pressure (IOP) elevation was produced by episcleral vein injection of hypertonic saline (N = 99). Retinas were collected and grouped by extent of optic nerve injury. Quantitative reverse transcription PCR, western blot analysis and immunohistochemistry were used to determine mRNA and protein levels and protein localization. Out of three RGC-specific Brn3 proteins (Brn3a, b, and c), only Brn3a was significantly downregulated at the message level to 35 +/- 4% of fellow values with the severest nerve injury. With IOP elevation, no significant alterations were found in retinal mRNA levels for BDNF, NGF, NT-4/5 or NT-3. The abundance of mature retinal BDNF protein was not significantly affected by elevated IOP, while proBDNF protein decreased linearly with increasing injury grade (r(2) = 0.50). In retinas with the severest nerve injury, TrkB and TrkC receptor mRNA levels significantly declined to 67 +/- 9% and 44 +/- 5% of fellow values, respectively. However, the levels of TRKB protein and its phosphorylated form were unchanged. Message level for p75(NTR) was linearly upregulated up to 219 +/- 26% with increasing injury (r(2) = 0.46), but no alteration was detected at protein level. The mRNA expression of p75(NTR) apoptosis adaptor proteins NADE, NRIF, and Lingo1 were significantly downregulated in retinas with the greatest nerve injury. A positive correlation was found between injury extent and message levels for Jun (r(2) = 0.23) as well as Junb (r(2) = 0.27), and RGC labeling of activated JUN protein increased. Atf3 mRNA levels demonstrated a positive linear correlation to the extent of injury (r(2) = 0.53), resulting in a nearly five-fold increase (482 +/- 76%) in eyes with the greatest nerve damage. Among downstream pro-survival signaling components, Erk5 mRNA expression was linearly upregulated (r(2) = 0.32) up to 157 +/- 15% of fellow values in retinas with the severest nerve injury (p < 0.01). A slight positive correlation was found between NF-kappaB message levels and injury extent (r(2) = 0.12). Bcl-xl mRNA levels in the most severely injured retinas were significantly reduced to 83 +/- 7% by elevated IOP exposure. Message levels for Erk1/2, Akt1-3 or Bcl2 appeared unaffected. Elevated IOP did not alter mRNA levels of pro-apoptotic Bim, Bax, or p53. This study demonstrates that elevated IOP exposure does not result in a dramatic decrease in retinal levels of either BDNF or its receptor, TrkB. It shows that the responses of NT pathways to elevated IOP are complex, particularly with regard to the role of p75(NTR) and Atf3. A better understanding of the roles of these proteins in IOP-induced injury is likely to suggest informed strategies for neuroprotection in glaucoma.
Collapse
Affiliation(s)
- Ying Guo
- The Kenneth C. Swan Ocular Neurobiology Laboratory, Casey Eye Institute, Oregon Health and Science University, 3375 SW Terwilliger Blvd, Portland, OR 97239, USA.
| | | | | | | | | | | |
Collapse
|
28
|
Johnson EC, Guo Y, Cepurna WO, Morrison JC. Neurotrophin roles in retinal ganglion cell survival: lessons from rat glaucoma models. Exp Eye Res 2009; 88:808-15. [PMID: 19217904 PMCID: PMC2704056 DOI: 10.1016/j.exer.2009.02.004] [Citation(s) in RCA: 86] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2008] [Revised: 01/13/2009] [Accepted: 02/03/2009] [Indexed: 12/19/2022]
Abstract
The neurotrophin (NT) hypothesis proposes that the obstruction of retrograde transport at the optic nerve head results in the deprivation of neurotrophic support to retinal ganglion cells (RGC) leading to apoptotic cell death in glaucoma. An important corollary to this concept is the implication that appropriate enhancement of neurotrophic support will prolong the survival of injured RGC indefinitely. This hypothesis is, perhaps, the most widely recognized theory to explain RGC loss resulting from exposure of the eye to elevated intraocular pressure (IOP). Recent studies of NT signaling using rat glaucoma models, have examined the endogenous responses of the retina to pressure exposure as well as studies designed to augment NT signaling in order to rescue RGC from apoptosis following pressure-induced injury. The examination of these studies in this review reveals a number of consistent observations and provides direction for further investigations of this hypothesis.
Collapse
Affiliation(s)
- Elaine C Johnson
- The Kenneth C. Swan Ocular Neurobiology Laboratory, Casey Eye Institute, CERES, Oregon Health and Science University, Portland, OR 97239, USA.
| | | | | | | |
Collapse
|
29
|
Pérez-Rico C, de la Villa P, Reinoso-Suárez F, Gómez-Ramos P. Kainic acid intraocular injections during the postnatal critical period induce plastic changes in the visual system. Neurosci Res 2009; 63:244-50. [PMID: 19167438 DOI: 10.1016/j.neures.2008.12.005] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2008] [Revised: 12/24/2008] [Accepted: 12/26/2008] [Indexed: 11/26/2022]
Abstract
Changes in the retino-collicular projection and in the number of optic nerve (ON) axons in adult rats were analyzed after partial loss of retinal ganglion cells (RGCs), induced by intravitreal injections of kainic acid (KA) on postnatal days 2-3 (P2-P3) or 10-12 (P10-P12). KA injected at P2-P3 decreased the volume of the adult contralateral superior colliculus (SC) and the density of the retino-collicular contralateral projection, but maintained the neonatal pattern in the ipsilateral projection from the un-injected eye. ON axon number was significantly increased in the un-injected eye but decreased in the KA-injected eye. Thus, restriction of the ipsilateral retino-collicular projection and RGC death in the un-injected eye are modified by KA at P2-P3, during the postnatal critical period, but not at P10-P12, after it is over. We suggest that, in the SC contralateral to the KA-injected eye, the disappearance of axon terminals belonging to RGC killed by KA would decrease competition between ipsilateral and contralateral terminals, thus contributing to maintaining the neonatal pattern in the ipsilateral retino-collicular projection. The reduction in RGC death in the un-injected eye could also be related to the disappearance of RGC terminals in the contralateral SC, which would have increased neurotrophic factor availability.
Collapse
|
30
|
Abstract
Elevated intraocular pressure in glaucoma can injure retinal ganglion cells and trigger the spread of disease to connected target vision structures of the brain. Glaucomatous degeneration has been observed in retrobulbar and intracranial optic nerve, lateral geniculate nucleus, and visual cortex of the brain. Oxidative damage and glutamate toxicity are implicated in transsynaptic central nervous system injury in glaucoma, similar to other neurodegenerative diseases. The perception of glaucoma as a disorder of "visual neurons" within the eye and brain may contribute to understanding progressive disease, and encourage comprehensive treatment strategies to prevent vision loss in glaucoma.
Collapse
|
31
|
Spalding KL, Cui Q, Dharmarajan AM, Harvey AR. Injury-induced retinal ganglion cell loss in the neonatal rat retina. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2007; 572:447-51. [PMID: 17249608 DOI: 10.1007/0-387-32442-9_62] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/13/2023]
Affiliation(s)
- Kirsty L Spalding
- School of Anatomy & Human Biology, The University of Western Australia, Crawley, WA 6009, Australia
| | | | | | | |
Collapse
|
32
|
Lönngren U, Näpänkangas U, Lafuente M, Mayor S, Lindqvist N, Vidal-Sanz M, Hallböök F. The growth factor response in ischemic rat retina and superior colliculus after brimonidine pre-treatment. Brain Res Bull 2006; 71:208-18. [PMID: 17113948 DOI: 10.1016/j.brainresbull.2006.09.005] [Citation(s) in RCA: 37] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2006] [Revised: 08/08/2006] [Accepted: 09/06/2006] [Indexed: 10/24/2022]
Abstract
The alpha-2-adrenergic receptor agonist brimonidine has been shown to increase survival of retinal ganglion cells following ischemic injury to the rat retina. Increased expression of growth factors has been suggested to be involved in this action. We investigated expressional changes of growth factors and their receptors following transient retinal ischemia induced by selective ligature of ophthalmic vessels in rats pre-treated with vehicle or 0.5% brimonidine. In addition, analysis of expression in retinal samples following unilateral administration of brimonidine to normal tissue was performed. Tissue samples of retina and superior colliculus were collected at time points between 6h and 14 days of retinal reperfusion. Analysis of mRNA levels of the ligands BDNF, NT3, CNTF, FGF1, FGF2, FGF9 and HGF; as well as the receptors TrkB, TrkC, p75(NTR), CNTFRalpha, FGFR1, FGFR3, FGFR4 and HGFR were performed using qRT-PCR. The cell specific markers Thy1 and GFAP were analysed. We report transiently increased retinal levels of BDNF, NT3, p75(NTR), FGFR1 and HGFR and decreased levels of FGF9, HGF, TrkB, TrkC, FGFR4 and Thy1 following ischemia. The decreases were counteracted by brimonidine. Brimonidine treatment gave an increase in BDNF, NT3 and CNTF levels compared to the vehicle treated group. In superior colliculus increased levels of growth factor mRNA were found. In conclusion, transient ischemia has a profound effect on gene expression in rat retina. Alterations can also be seen in the superior colliculus but are smaller. Brimonidine pre-treatment attenuates an acute injury-induced response by decreasing the expression of several genes, among them p75(NTR). Brimonidine also causes a prolonged increase of several growth factors as well as receptors in retina and superior colliculus compared to the ischemic situation. The increased expression of several growth factors represents a coordinated growth factor system response that differs from the ischemia-induced changes and is likely part of the neuroprotective activity that is elicited by BMD pre-treatment.
Collapse
Affiliation(s)
- Ulrika Lönngren
- Department of Neuroscience, Uppsala University, Uppsala, Sweden
| | | | | | | | | | | | | |
Collapse
|
33
|
Harvey AR, Hu Y, Leaver SG, Mellough CB, Park K, Verhaagen J, Plant GW, Cui Q. Gene therapy and transplantation in CNS repair: The visual system. Prog Retin Eye Res 2006; 25:449-89. [PMID: 16963308 DOI: 10.1016/j.preteyeres.2006.07.002] [Citation(s) in RCA: 82] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Normal visual function in humans is compromised by a range of inherited and acquired degenerative conditions, many of which affect photoreceptors and/or retinal pigment epithelium. As a consequence the majority of experimental gene- and cell-based therapies are aimed at rescuing or replacing these cells. We provide a brief overview of these studies, but the major focus of this review is on the inner retina, in particular how gene therapy and transplantation can improve the viability and regenerative capacity of retinal ganglion cells (RGCs). Such studies are relevant to the development of new treatments for ocular conditions that cause RGC loss or dysfunction, for example glaucoma, diabetes, ischaemia, and various inflammatory and neurodegenerative diseases. However, RGCs and associated central visual pathways also serve as an excellent experimental model of the adult central nervous system (CNS) in which it is possible to study the molecular and cellular mechanisms associated with neuroprotection and axonal regeneration after neurotrauma. In this review we present the current state of knowledge pertaining to RGC responses to injury, neurotrophic and gene therapy strategies aimed at promoting RGC survival, and how best to promote the regeneration of RGC axons after optic nerve or optic tract injury. We also describe transplantation methods being used in attempts to replace lost RGCs or encourage the regrowth of RGC axons back into visual centres in the brain via peripheral nerve bridges. Cooperative approaches including novel combinations of transplantation, gene therapy and pharmacotherapy are discussed. Finally, we consider a number of caveats and future directions, such as problems associated with compensatory sprouting and the reformation of visuotopic maps, the need to develop efficient, regulatable viral vectors, and the need to develop different but sequential strategies that target the cell body and/or the growth cone at appropriate times during the repair process.
Collapse
Affiliation(s)
- Alan R Harvey
- School of Anatomy and Human Biology, The University of Western Australia, Crawley, WA 6009, Australia
| | | | | | | | | | | | | | | |
Collapse
|
34
|
Turner BA, Sparrow J, Cai B, Monroe J, Mikawa T, Hempstead BL. TrkB/BDNF signaling regulates photoreceptor progenitor cell fate decisions. Dev Biol 2006; 299:455-65. [PMID: 17005175 PMCID: PMC2623246 DOI: 10.1016/j.ydbio.2006.08.025] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2005] [Revised: 08/07/2006] [Accepted: 08/09/2006] [Indexed: 10/24/2022]
Abstract
Neurotrophins, via activation of Trk receptor tyrosine kinases, serve as mitogens, survival factors and regulators of arborization during retinal development. Brain-derived neurotrophic factor (BDNF) and TrkB regulate neuronal arborization and survival in late retinal development. However, TrkB is expressed during early retinal development where its functions are unclear. To assess TrkB/BDNF actions in the early chick retina, replication-incompetent retroviruses were utilized to over-express a dominant negative truncated form of TrkB (trunc TrkB), or BDNF and effects were assessed at E15. Clones expressing trunc TrkB were smaller than controls, and proliferation and apoptosis assays suggest that decreased clone size correlated with increased cell death when BDNF/TrkB signaling was impaired. Analysis of clonal composition revealed that trunc TrkB over-expression decreased photoreceptor numbers (41%) and increased cell numbers in the middle third of the inner nuclear layer (INL) (23%). Conversely, BDNF over-expression increased photoreceptor numbers (25%) and decreased INL numbers (17%). Photoreceptors over-expressing trunc TrkB demonstrated no increase in apoptosis nor abnormalities in lamination suggesting that TrkB activation is not required for photoreceptor cell survival or migration. These studies suggest that TrkB signaling regulates commitment to and/or differentiation of photoreceptor cells from retinal progenitor cells, identifying a novel role for TrkB/BDNF in regulating cell fate decisions.
Collapse
Affiliation(s)
- Brian A. Turner
- Department of Medicine, Weill Medical College of Cornell University, New York, New York, 10021
| | - Janet Sparrow
- Department of Ophthalmology, Columbia University, New York, New York, 10032
| | - Bolin Cai
- Department of Ophthalmology, Columbia University, New York, New York, 10032
| | - Julie Monroe
- Department of Medicine, Weill Medical College of Cornell University, New York, New York, 10021
| | - Takashi Mikawa
- Department of Cell Biology, Weill Medical College of Cornell University, New York, New York, 10021
| | - Barbara L. Hempstead
- Department of Medicine, Weill Medical College of Cornell University, New York, New York, 10021
- *Author to whom to address correspondence: Barbara L. Hempstead, Department of Medicine, Weill Medical College of Cornell University, 1300 York Avenue, Room C606, New York, New York, 10021, phone: 212-746-6215, fax: 212-746-8647,
| |
Collapse
|
35
|
Glorioso C, Sabatini M, Unger T, Hashimoto T, Monteggia LM, Lewis DA, Mirnics K. Specificity and timing of neocortical transcriptome changes in response to BDNF gene ablation during embryogenesis or adulthood. Mol Psychiatry 2006; 11:633-48. [PMID: 16702976 DOI: 10.1038/sj.mp.4001835] [Citation(s) in RCA: 78] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Brain-derived neurotrophic factor (BDNF) has been reported to be critical for the development of cortical inhibitory neurons. However, the effect of BDNF on the expression of transcripts whose protein products are involved in gamma amino butric acid (GABA) neurotransmission has not been assessed. In this study, gene expression profiling using oligonucleotide microarrays was performed in prefrontal cortical tissue from mice with inducible deletions of BDNF. Both embryonic and adulthood ablation of BDNF gave rise to many shared transcriptome changes. BDNF appeared to be required to maintain gene expression in the SST-NPY-TAC1 subclass of GABA neurons, although the absence of BDNF did not alter their general phenotype as inhibitory neurons. Furthermore, we observed expression alterations in genes encoding early-immediate genes (ARC, EGR1, EGR2, FOS, DUSP1, DUSP6) and critical cellular signaling systems (CDKN1c, CCND2, CAMK1g, RGS4). These BDNF-dependent gene expression changes may illuminate the biological basis for transcriptome changes observed in certain human brain disorders.
Collapse
Affiliation(s)
- C Glorioso
- Department of Psychiatry, University of Pittsburgh School of Medicine, PA 15261, USA
| | | | | | | | | | | | | |
Collapse
|
36
|
Murphy JA, Clarke DB. Target-derived neurotrophins may influence the survival of adult retinal ganglion cells when local neurotrophic support is disrupted: Implications for glaucoma. Med Hypotheses 2006; 67:1208-12. [PMID: 16806723 DOI: 10.1016/j.mehy.2006.04.049] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2006] [Accepted: 04/28/2006] [Indexed: 10/24/2022]
Abstract
Following target innervation, developing and neonatal retinal ganglion cells (RGCs) depend on neurotrophic factors from their target tissue for survival. This dependence is reduced for adult RGCs which rely primarily on trophic support from their local environment; however, some findings indicate that target tissue may play a role in the long-term survival of RGCs. We propose that a deficiency in neurotrophic factors from the target tissue may influence the survival of RGCs when local neurotrophic support is disrupted. Furthermore, we propose that this hypothesis may explain, at least in part, the progressive loss of RGCs in optic neuropathies such as glaucoma. Neurotrophic factors are present in the adult superior colliculus and they are trafficked to the retina; however, removal or lesioning of the adult target tissue results in little or no RGC loss for up to several months. In vitro, adult RGCs will survive when maintained by co-culturing these neurons with their target tissue. As well, the timing and pattern of adult RGC loss is consistent with that seen in glaucoma and in reports of delayed RGC loss following target-removal. Our hypothesis can be tested by selectively disrupting local neurotrophic support and evaluating RGC survival when target-derived neurotrophic support is maintained and when it is disrupted. Specifically, intravitreal injection of blocking antibodies could be used to disrupt local neurotrophic signaling, while aspiration of the superior colliculus will eliminate retrograde transport from the primary target tissue in rodents. The results of these experiments would provide valuable information concerning the influence of target-derived neurotrophic support when local neurotrophin signaling is disrupted. Specifically, this research could verify whether deficiencies in target-derived neurotrophic support play a role in RGC loss during glaucoma. A further understanding of this mechanism may lead to the development of effective neuroprotective strategies for treating glaucoma.
Collapse
Affiliation(s)
- J A Murphy
- Neuron Survival and Regeneration Laboratory, Department of Anatomy and Neurobiology, Dalhousie University, 5850 College Street, Halifax, NS, Canada
| | | |
Collapse
|
37
|
Dunlop SA, Rodger J, Beazley LD. Compensatory and transneuronal plasticity after early collicular ablation. J Comp Neurol 2006; 500:1117-26. [PMID: 17183539 DOI: 10.1002/cne.21221] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
Plasticity within the visual system was assessed in the quokka wallaby following unilateral superior collicular (SC) ablation at postnatal days (P) 8-10, prior to the arrival of retinal ganglion cell (RGC) axons. At maturity (P100), projections were traced from the eye opposite the ablation, and total RGC numbers were estimated for both eyes. Ablations were partial (28-89% of SC remaining) or complete (0-5% of SC remaining). Projections to the visual centers showed significant bilateral (P < 0.05) increases in absolute volume. Minor anomalous projections also formed within the deep, surviving non-retino-recipient layers of the ablated SC and via a small bundle of RGC axons recrossing the midline to innervate discrete patches in the SC contralateral to the lesion. Total absolute volume of projections did not differ between partial and complete ablations; moreover, values did not differ from normal (P > 0.05). Compared with normal, total RGC numbers were significantly (P < 0.05) reduced in the eye opposite the ablation but increased (P < 0.05) in the other eye. Consequently, the sum of the two RGC populations did not differ from normal (P > 0.05). As in rodents, the visual system in quokka compensates following injury by maintaining a set volume of arborization but does so by forming only minor anomalous projections. Furthermore, increased RGC numbers in the eye ipsilateral to the lesion indicate that compensation occurs transneuronally, thus maintaining total numbers of projecting neurons. The implication is that the visual system acts in concert following unilateral injury to maintain set values for RGC terminal arbors as well as their cell bodies.
Collapse
Affiliation(s)
- Sarah A Dunlop
- School of Animal Biology, Western Australian Institute of Medical Research, The University of Western Australia, Crawley, Australia 6907.
| | | | | |
Collapse
|
38
|
Spalding KL, Dharmarajan AM, Harvey AR. Caspase-independent retinal ganglion cell death after target ablation in the neonatal rat. Eur J Neurosci 2005; 21:33-45. [PMID: 15654841 DOI: 10.1111/j.1460-9568.2004.03826.x] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
In neonatal rats, superior colliculus (SC) ablation results in a massive and rapid increase in retinal ganglion cell (RGC) death that peaks about 24 h post-lesion (PL). Naturally occurring cell death during normal development, and RGC death after axonal injury in neonatal and adult rats, has primarily been ascribed to apoptosis. Given that normal developmental cell death is reported to involve caspase 3 activation, and blocking caspase activity in adults reduces axotomy-induced death, we examined whether blocking caspases in vivo reduces RGC death after neonatal SC lesions. Neither general nor specific caspase inhibitors increased neonatal RGC survival 6 and 24 h PL. These inhibitors were, however, effective in blocking caspases in another well-defined in vitro apoptosis model, the corpus luteum. Caspase 3 protein and mRNA levels in retinas from normal and SC-lesioned neonatal rats were assessed 3, 6 and 24 h after SC removal using immunohistochemistry, western and northern blots and quantitative real-time polymerase chain reaction. Terminal deoxynucleotidyl transferase-mediated dUTP nick-end labelling (TUNEL) was used to independently monitor retinal cell death. The polymerase chain reaction data showed a small but insignificant increase in caspase 3 mRNA in retinas 24 h PL. Western blot analysis did not reveal a significant shift to cleaved (activated) caspase 3 protein. There was a small increase in the number of cleaved caspase 3 immunolabelled cells in the ganglion cell layer 24 h PL but this represented only a fraction of the death revealed by TUNEL. Together, these data indicate that, unlike the situation in adults, most lesion-induced RGC death in neonatal rats occurs independently of caspase activation.
Collapse
Affiliation(s)
- Kirsty L Spalding
- School of Anatomy and Human Biology, The University of Western Australia, Crawley, WA, Australia.
| | | | | |
Collapse
|