1
|
Haine L, Taylor I, Vaughan M. The Psychological Impact of Amblyopia Treatment: A Systematic Literature Review. Br Ir Orthopt J 2025; 21:1-14. [PMID: 39831149 PMCID: PMC11740718 DOI: 10.22599/bioj.426] [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: 09/06/2024] [Accepted: 12/24/2024] [Indexed: 01/22/2025] Open
Abstract
Aim The aim of this literature review was to determine if a consensus could be reached on whether amblyopia treatment causes distress to patients and/or their guardians, and if so, establish the impact of this reported psychological distress upon paediatric patients and/or their parents/guardians. Methods A systematic review of the literature was conducted of all publications written in English. Search terms included both MeSH terms and alternatives related to amblyopia and psychological distress. Evidence quality was assessed using an adapted Newcastle-Ottawa Score (NOS) and evaluation of the literature was used to form a narrative synthesis of the findings. Results Initial searches yielded 7,838 titles in total, with 25 peer reviewed papers published between 1999 and 2021 meeting the study inclusion criteria. Factors such as the presence of strabismus, moderate and severe amblyopic density, occlusive patch treatment and patching during school age increase the likelihood of experiencing distress as a result of amblyopia treatment. Conclusions Both parents/guardians and patients can experience psychological distress as a result of undertaking amblyopia treatment. School-aged children and those receiving occlusion therapy in the form of patching report higher distress than infants and young-children, and those receiving atropine occlusion therapy or refractive correction only. Further study measuring the physiological markers of distress such as Cortisol and BDNF, is recommended.
Collapse
|
2
|
Ebrahimi M, Thompson P, Lauer AK, Sivaprasad S, Perry G. The retina-brain axis and diabetic retinopathy. Eur J Ophthalmol 2023; 33:2079-2095. [PMID: 37259525 DOI: 10.1177/11206721231172229] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/02/2023]
Abstract
Diabetic retinopathy (DR) is a major contributor to permanent vision loss and blindness. Changes in retinal neurons, glia, and microvasculature have been the focus of intensive study in the quest to better understand DR. However, the impact of diabetes on the rest of the visual system has received less attention. There are reports of associations of changes in the visual system with preclinical and clinical manifestations of diabetes. Simultaneous investigation of the retina and the brain may shed light on the mechanisms underlying neurodegeneration in diabetics. Additionally, investigating the links between DR and other neurodegenerative disorders of the brain including Alzheimer's and Parkinson's disease may reveal shared mechanisms for neurodegeneration and potential therapy options.
Collapse
Affiliation(s)
- Moein Ebrahimi
- School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Network of Immunity in Infection, Malignancy, and Autoimmunity, Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Paul Thompson
- Imaging Genetics Center, Mark and Mary Stevens Institute for Neuroimaging and Informatics, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Andreas K Lauer
- Department of Ophthalmology, Casey Eye Institute, Oregon Health & Science University, Portland, Oregon, USA
| | - Sobha Sivaprasad
- National Institute of Health and Care Research Moorfields Biomedical Research Centre, Moorfields Eye Hospital, London, UK
| | - George Perry
- Department of Neuroscience, Developmental and Regenerative Biology, University of Texas and San Antonio, San Antonio, TX, USA
| |
Collapse
|
3
|
Duffy KR, Crowder NA, Heynen AJ, Bear MF. Comparative analysis of structural modifications induced by monocular retinal inactivation and monocular deprivation in the developing cat lateral geniculate nucleus. J Comp Neurol 2023; 531:1244-1260. [PMID: 37139534 PMCID: PMC10330129 DOI: 10.1002/cne.25493] [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: 08/07/2022] [Revised: 04/10/2023] [Accepted: 04/13/2023] [Indexed: 05/05/2023]
Abstract
During a critical period of postnatal life, monocular deprivation (MD) by eyelid closure reduces the size of neurons in layers of the dorsal lateral geniculate nucleus (dLGN) connected to the deprived eye and shifts cortical ocular dominance in favor of the non-deprived eye. Temporary inactivation of the non-deprived eye can promote superior recovery from the effects of long-term MD compared to conventional occlusion therapy. In the current study, we assessed the modification of neuron size in the dLGN as a means of measuring the impact of a brief period of monocular inactivation (MI) imposed at different postnatal ages. The biggest impact of MI was observed when it occurred at the peak of the critical period. Unlike the effect of MD, structural plasticity following MI was observed in both the binocular and monocular segments of the dLGN. With increasing age, the capacity for inactivation to alter postsynaptic cell size diminished but was still significant beyond the critical period. In comparison to MD, inactivation produced effects that were about double in magnitude and exhibited efficacy at older ages. Notwithstanding the large neural alterations precipitated by MI, its effects were remediated with a short period of binocular experience, and vision through the previously inactivated eye fully recovered. These results demonstrate that MI is a potent means of modifying the visual pathway and does so at ages when occlusion is ineffective. The efficacy and longevity of inactivation to elicit plasticity highlight its potential to ameliorate disorders of the visual system such as amblyopia.
Collapse
Affiliation(s)
- Kevin R Duffy
- Department of Psychology & Neuroscience, Dalhousie University, Halifax, Canada
| | - Nathan A Crowder
- Department of Psychology & Neuroscience, Dalhousie University, Halifax, Canada
| | - Arnold J Heynen
- Picower Institute for Learning and Memory, Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | - Mark F Bear
- Picower Institute for Learning and Memory, Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| |
Collapse
|
4
|
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
|
5
|
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
|
6
|
Li B, Zou Y, Yin X, Tang X, Fan H. Expression of brain-derived neurotrophic factor in the lateral geniculate body of monocular form deprivation amblyopic kittens. Eur J Ophthalmol 2020; 31:2724-2730. [PMID: 32873060 DOI: 10.1177/1120672120953341] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE The present study compared the expression of brain-derived neurotrophic factor (BDNF) in the lateral geniculate body between form deprivation amblyopia kittens and normal kittens to examine the significance of BDNF in the lateral geniculate body in the pathogenesis of amblyopia. METHODS Twenty kittens were divided into control group (n = 10) and deprivation group (n = 10). A black opaque eye mask was placed to cover the right eye of the deprivation group. Pattern visual-evoked potentials (PVEPs) were detected weekly in all kittens .After the kittens in the deprivation group developed monocular amblyopia, the lateral geniculate bodies of all kittens were removed. The expression of BDNF in the lateral geniculate body of the two groups was compared by immunohistochemistry and Western blotting. RESULTS The latency of the P100 wave in the right eye of the deprivation group was longer than that of the left eye and that of the right eye of the control group (p < 0.05), and the amplitude decreased (p < 0.05). The number and average optical density of BDNF-positive cells in the deprivation group were lower than those in the control group (p < 0.05), and the expression of BDNF in the deprivation group was lower than that in the control group (p < 0.05). CONCLUSIONS The expression of BDNF in the lateral geniculate body of the amblyopic kittens decreased, and the decrease in BDNF promoted the development of amblyopia. These results demonstrate that BDNF in the lateral geniculate body plays an important role in visual development.
Collapse
Affiliation(s)
- Bo Li
- Department of Ophthalmology, Suining Central Hospital, Suining, China.,Department of Ophthalmology, affiliated Hospital of North Sichuan Medical College, Nanchong, China.,Department of Optometry, North Sichuan Medical College, Nanchong, China
| | - Yunchun Zou
- Department of Ophthalmology, affiliated Hospital of North Sichuan Medical College, Nanchong, China.,Department of Optometry, North Sichuan Medical College, Nanchong, China
| | - Ximin Yin
- Department of Ophthalmology, affiliated Hospital of North Sichuan Medical College, Nanchong, China.,Department of Optometry, North Sichuan Medical College, Nanchong, China
| | - Xiuping Tang
- Department of Ophthalmology, affiliated Hospital of North Sichuan Medical College, Nanchong, China.,Department of Optometry, North Sichuan Medical College, Nanchong, China
| | - Haobo Fan
- Department of Ophthalmology, affiliated Hospital of North Sichuan Medical College, Nanchong, China.,Department of Optometry, North Sichuan Medical College, Nanchong, China
| |
Collapse
|
7
|
Eastlake K, Luis J, Limb GA. Potential of Müller Glia for Retina Neuroprotection. Curr Eye Res 2020; 45:339-348. [PMID: 31355675 DOI: 10.1080/02713683.2019.1648831] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2019] [Accepted: 07/19/2019] [Indexed: 12/26/2022]
Abstract
Müller glia constitute the main glial cells of the retina. They are spatially distributed along this tissue, facilitating their close membrane interactions with all retinal neurons. Müller glia are characterized by their active metabolic functions, which are neuroprotective in nature. Although they can become reactive under pathological conditions, leading to their production of inflammatory and neurotoxic factors, their main metabolic functions confer neuroprotection to the retina, resulting in the promotion of neural cell repair and survival. In addition to their protective metabolic features, Müller glia release several neurotrophic factors and antioxidants into the retinal microenvironment, which are taken up by retinal neurons for their survival. This review summarizes the Müller glial neuroprotective mechanisms and describes advances made on the clinical application of these factors for the treatment of retinal degenerative diseases. It also discusses prospects for the use of these cells as a vehicle to deliver neuroprotective factors into the retina.
Collapse
Affiliation(s)
- Karen Eastlake
- UCL Institute of Ophthalmology and NIHR Biomedical Research Centre at Moorfields Eye Hospital, London, UK
| | - Joshua Luis
- UCL Institute of Ophthalmology and NIHR Biomedical Research Centre at Moorfields Eye Hospital, London, UK
| | - G Astrid Limb
- UCL Institute of Ophthalmology and NIHR Biomedical Research Centre at Moorfields Eye Hospital, London, UK
| |
Collapse
|
8
|
Choi JA, Jung IY, Jee D. The Relationship Between the Sighting Eye and Functional and Structural Asymmetries in Glaucoma. ACTA ACUST UNITED AC 2018; 59:5447-5454. [DOI: 10.1167/iovs.18-24083] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Affiliation(s)
- Jin A Choi
- Department of Ophthalmology and Visual Science, St. Vincent's Hospital, College of Medicine, the Catholic University of Korea, Seoul, Korea
| | - Il-Young Jung
- Department of Ophthalmology and Visual Science, St. Vincent's Hospital, College of Medicine, the Catholic University of Korea, Seoul, Korea
| | - Donghyun Jee
- Department of Ophthalmology and Visual Science, St. Vincent's Hospital, College of Medicine, the Catholic University of Korea, Seoul, Korea
| |
Collapse
|
9
|
Structural changes of the macula and optic nerve head in the remaining eyes after enucleation for retinoblastoma: an optical coherence tomography study. BMC Ophthalmol 2017; 17:251. [PMID: 29246122 PMCID: PMC5732405 DOI: 10.1186/s12886-017-0650-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2017] [Accepted: 12/05/2017] [Indexed: 01/06/2023] Open
Abstract
BACKGROUND To describe objectively the possible structural changes of the macula and optic nerve head in the free eyes of unilateral cured retinoblastoma patients and, also after enucleation using spectral domain optical coherence tomography. METHODS A cross sectional study involving 60 patients subdivided into three groups; 15 unilateral RB patients in whom enucleation was indicated as a sole treatment performed earlier in life [(study group (I)], 15 unilateral RB patients who had completely regressed disease with a preserved eye [(study group (II)] and 30 age and sex matched healthy controls. The remaining and free eyes in study groups and right eyes of control group had full ophthalmological examination, static automated perimetry and optical coherence tomography of the macula and optic nerve head. RESULTS In study group (II); a significant thinning of total macula, central fovea, ganglion cell layer (GCL), ganglion cell complex (GCC), and some sectors of outer nuclear layer (P- values ≤0.05) was found with no significant difference in peripapillary nerve fiber layer (pRNFL) thickness and optic nerve head parameters compared to the control group and the study group (I). A significantly thickened total macula, GCL, GCC, and pRNFL in study group (I) compared to study group (II). Thickened pRNFL was significantly correlated to standard automated perimetry pattern deviations. No significant difference was found between study group (I) and control group. CONCLUSION Retinoblastoma eyes characterized by thinning of central fovea, GCL, GCC compared to the control group. After unilateral enucleation, increased GCC and pRNFL thicknesses were detected compared to retinoblastoma group.
Collapse
|
10
|
Walton MMG, Pallus A, Fleuriet J, Mustari MJ, Tarczy-Hornoch K. Neural mechanisms of oculomotor abnormalities in the infantile strabismus syndrome. J Neurophysiol 2017; 118:280-299. [PMID: 28404829 DOI: 10.1152/jn.00934.2016] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2016] [Revised: 04/03/2017] [Accepted: 04/07/2017] [Indexed: 02/08/2023] Open
Abstract
Infantile strabismus is characterized by numerous visual and oculomotor abnormalities. Recently nonhuman primate models of infantile strabismus have been established, with characteristics that closely match those observed in human patients. This has made it possible to study the neural basis for visual and oculomotor symptoms in infantile strabismus. In this review, we consider the available evidence for neural abnormalities in structures related to oculomotor pathways ranging from visual cortex to oculomotor nuclei. These studies provide compelling evidence that a disturbance of binocular vision during a sensitive period early in life, whatever the cause, results in a cascade of abnormalities through numerous brain areas involved in visual functions and eye movements.
Collapse
Affiliation(s)
- Mark M G Walton
- Washington National Primate Research Center, University of Washington, Seattle, Washington;
| | - Adam Pallus
- Washington National Primate Research Center, University of Washington, Seattle, Washington.,Department of Ophthalmology, University of Washington, Seattle, Washington
| | - Jérome Fleuriet
- Washington National Primate Research Center, University of Washington, Seattle, Washington.,Department of Ophthalmology, University of Washington, Seattle, Washington
| | - Michael J Mustari
- Washington National Primate Research Center, University of Washington, Seattle, Washington.,Department of Ophthalmology, University of Washington, Seattle, Washington.,Department of Biological Structure, University of Washington, Seattle, Washington; and
| | - Kristina Tarczy-Hornoch
- Department of Ophthalmology, University of Washington, Seattle, Washington.,Seattle Children's Hospital, Seattle, Washington
| |
Collapse
|
11
|
Chumak T, Rüttiger L, Lee SC, Campanelli D, Zuccotti A, Singer W, Popelář J, Gutsche K, Geisler HS, Schraven SP, Jaumann M, Panford-Walsh R, Hu J, Schimmang T, Zimmermann U, Syka J, Knipper M. BDNF in Lower Brain Parts Modifies Auditory Fiber Activity to Gain Fidelity but Increases the Risk for Generation of Central Noise After Injury. Mol Neurobiol 2015; 53:5607-27. [PMID: 26476841 PMCID: PMC5012152 DOI: 10.1007/s12035-015-9474-x] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2015] [Accepted: 10/05/2015] [Indexed: 11/24/2022]
Abstract
For all sensory organs, the establishment of spatial and temporal cortical resolution is assumed to be initiated by the first sensory experience and a BDNF-dependent increase in intracortical inhibition. To address the potential of cortical BDNF for sound processing, we used mice with a conditional deletion of BDNF in which Cre expression was under the control of the Pax2 or TrkC promoter. BDNF deletion profiles between these mice differ in the organ of Corti (BDNFPax2-KO) versus the auditory cortex and hippocampus (BDNFTrkC-KO). We demonstrate that BDNFPax2-KO but not BDNFTrkC-KO mice exhibit reduced sound-evoked suprathreshold ABR waves at the level of the auditory nerve (wave I) and inferior colliculus (IC) (wave IV), indicating that BDNF in lower brain regions but not in the auditory cortex improves sound sensitivity during hearing onset. Extracellular recording of IC neurons of BDNFPax2 mutant mice revealed that the reduced sensitivity of auditory fibers in these mice went hand in hand with elevated thresholds, reduced dynamic range, prolonged latency, and increased inhibitory strength in IC neurons. Reduced parvalbumin-positive contacts were found in the ascending auditory circuit, including the auditory cortex and hippocampus of BDNFPax2-KO, but not of BDNFTrkC-KO mice. Also, BDNFPax2-WT but not BDNFPax2-KO mice did lose basal inhibitory strength in IC neurons after acoustic trauma. These findings suggest that BDNF in the lower parts of the auditory system drives auditory fidelity along the entire ascending pathway up to the cortex by increasing inhibitory strength in behaviorally relevant frequency regions. Fidelity and inhibitory strength can be lost following auditory nerve injury leading to diminished sensory outcome and increased central noise.
Collapse
Affiliation(s)
- Tetyana Chumak
- Department of Auditory Neuroscience, Institute of Experimental Medicine, Academy of Sciences of the Czech Republic, Vídeňská 1083, 142 20, Prague, Czech Republic
| | - Lukas Rüttiger
- Department of Otolaryngology, Hearing Research Centre Tübingen, Molecular Physiology of Hearing, University of Tübingen, Elfriede-Aulhorn-Str. 5, 72076, Tübingen, Germany
| | - Sze Chim Lee
- Department of Otolaryngology, Hearing Research Centre Tübingen, Molecular Physiology of Hearing, University of Tübingen, Elfriede-Aulhorn-Str. 5, 72076, Tübingen, Germany
| | - Dario Campanelli
- Department of Otolaryngology, Hearing Research Centre Tübingen, Molecular Physiology of Hearing, University of Tübingen, Elfriede-Aulhorn-Str. 5, 72076, Tübingen, Germany
| | - Annalisa Zuccotti
- Department of Otolaryngology, Hearing Research Centre Tübingen, Molecular Physiology of Hearing, University of Tübingen, Elfriede-Aulhorn-Str. 5, 72076, Tübingen, Germany.,Department of Clinical Neurobiology, University Hospital and DKFZ Heidelberg, In Neuenheimer Feld 280, 69120, Heidelberg, Germany
| | - Wibke Singer
- Department of Otolaryngology, Hearing Research Centre Tübingen, Molecular Physiology of Hearing, University of Tübingen, Elfriede-Aulhorn-Str. 5, 72076, Tübingen, Germany
| | - Jiří Popelář
- Department of Auditory Neuroscience, Institute of Experimental Medicine, Academy of Sciences of the Czech Republic, Vídeňská 1083, 142 20, Prague, Czech Republic
| | - Katja Gutsche
- Instituto de Biologíay Genética Molecular, Universidad de Valladolid y Consejo Superior de Investigaciones Científicas, E-47003, Valladolid, Spain
| | - Hyun-Soon Geisler
- Department of Otolaryngology, Hearing Research Centre Tübingen, Molecular Physiology of Hearing, University of Tübingen, Elfriede-Aulhorn-Str. 5, 72076, Tübingen, Germany
| | - Sebastian Philipp Schraven
- Department of Otolaryngology, Plastic, Aesthetic and Reconstructive Head and Neck Surgery, Comprehensive Hearing Center, University of Würzburg, Josef-Schneider-Straße 11, 97080, Würzburg, Germany
| | - Mirko Jaumann
- Department of Otolaryngology, Hearing Research Centre Tübingen, Molecular Physiology of Hearing, University of Tübingen, Elfriede-Aulhorn-Str. 5, 72076, Tübingen, Germany
| | | | - Jing Hu
- Centre for Integrative Neuroscience, University of Tübingen, Otfried-Müller-Straße 25, 72076, Tübingen, Germany
| | - Thomas Schimmang
- Instituto de Biologíay Genética Molecular, Universidad de Valladolid y Consejo Superior de Investigaciones Científicas, E-47003, Valladolid, Spain
| | - Ulrike Zimmermann
- Department of Otolaryngology, Hearing Research Centre Tübingen, Molecular Physiology of Hearing, University of Tübingen, Elfriede-Aulhorn-Str. 5, 72076, Tübingen, Germany
| | - Josef Syka
- Department of Auditory Neuroscience, Institute of Experimental Medicine, Academy of Sciences of the Czech Republic, Vídeňská 1083, 142 20, Prague, Czech Republic
| | - Marlies Knipper
- Department of Otolaryngology, Hearing Research Centre Tübingen, Molecular Physiology of Hearing, University of Tübingen, Elfriede-Aulhorn-Str. 5, 72076, Tübingen, Germany.
| |
Collapse
|
12
|
Mwachaka PM, Saidi H, Odula PO, Mandela PI. Effect of Monocular Deprivation on Rabbit Neural Retinal Cell Densities. J Ophthalmic Vis Res 2015; 10:144-50. [PMID: 26425316 PMCID: PMC4568611 DOI: 10.4103/2008-322x.163770] [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] [Indexed: 11/04/2022] Open
Abstract
PURPOSE To describe the effect of monocular deprivation on densities of neural retinal cells in rabbits. METHODS Thirty rabbits, comprised of 18 subject and 12 control animals, were included and monocular deprivation was achieved through unilateral lid suturing in all subject animals. The rabbits were observed for three weeks. At the end of each week, 6 experimental and 3 control animals were euthanized, their retinas was harvested and processed for light microscopy. Photomicrographs of the retina were taken and imported into FIJI software for analysis. RESULTS Neural retinal cell densities of deprived eyes were reduced along with increasing period of deprivation. The percentage of reductions were 60.9% (P < 0.001), 41.6% (P = 0.003), and 18.9% (P = 0.326) for ganglion, inner nuclear, and outer nuclear cells, respectively. In non-deprived eyes, cell densities in contrast were increased by 116% (P < 0.001), 52% (P < 0.001) and 59.6% (P < 0.001) in ganglion, inner nuclear, and outer nuclear cells, respectively. CONCLUSION In this rabbit model, monocular deprivation resulted in activity-dependent changes in cell densities of the neural retina in favour of the non-deprived eye along with reduced cell densities in the deprived eye.
Collapse
Affiliation(s)
| | - Hassan Saidi
- Department of Human Anatomy, University of Nairobi, Nairobi, Kenya
| | | | | |
Collapse
|
13
|
Willoughby CL, Fleuriet J, Walton MM, Mustari MJ, McLoon LK. Adaptation of slow myofibers: the effect of sustained BDNF treatment of extraocular muscles in infant nonhuman primates. Invest Ophthalmol Vis Sci 2015; 56:3467-83. [PMID: 26030102 DOI: 10.1167/iovs.15-16852] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
PURPOSE We evaluated promising new treatment options for strabismus. Neurotrophic factors have emerged as a potential treatment for oculomotor disorders because of diverse roles in signaling to muscles and motor neurons. Unilateral treatment with sustained release brain-derived neurotrophic factor (BDNF) to a single lateral rectus muscle in infant monkeys was performed to test the hypothesis that strabismus would develop in correlation with extraocular muscle (EOM) changes during the critical period for development of binocularity. METHODS The lateral rectus muscles of one eye in two infant macaques were treated with sustained delivery of BDNF for 3 months. Eye alignment was assessed using standard photographic methods. Muscle specimens were analyzed to examine the effects of BDNF on the density, morphology, and size of neuromuscular junctions, as well as myofiber size. Counts were compared to age-matched controls. RESULTS No change in eye alignment occurred with BDNF treatment. Compared to control muscle, neuromuscular junctions on myofibers expressing slow myosins had a larger area. Myofibers expressing slow myosin had larger diameters, and the percentage of myofibers expressing slow myosins increased in the proximal end of the muscle. Expression of BDNF was examined in control EOM, and observed to have strongest immunoreactivity outside the endplate zone. CONCLUSIONS We hypothesize that the oculomotor system adapted to sustained BDNF treatment to preserve normal alignment. Our results suggest that BDNF treatment preferentially altered myofibers expressing slow myosins. This implicates BDNF signaling as influencing the slow twitch properties of EOM.
Collapse
Affiliation(s)
- Christy L Willoughby
- Graduate Program in Neuroscience, University of Minnesota, Minneapolis, Minnesota, United States 2Department of Ophthalmology and Visual Neurosciences, University of Minnesota, Minneapolis, Minnesota, United States
| | - Jérome Fleuriet
- Washington National Primate Research Center, Seattle, Washington, United States 4Department of Ophthalmology, University of Washington, Seattle, Washington, United States
| | - Mark M Walton
- Washington National Primate Research Center, Seattle, Washington, United States
| | - Michael J Mustari
- Washington National Primate Research Center, Seattle, Washington, United States 4Department of Ophthalmology, University of Washington, Seattle, Washington, United States
| | - Linda K McLoon
- Graduate Program in Neuroscience, University of Minnesota, Minneapolis, Minnesota, United States 2Department of Ophthalmology and Visual Neurosciences, University of Minnesota, Minneapolis, Minnesota, United States
| |
Collapse
|
14
|
Low-intensity repetitive transcranial magnetic stimulation improves abnormal visual cortical circuit topography and upregulates BDNF in mice. J Neurosci 2014; 34:10780-92. [PMID: 25100609 DOI: 10.1523/jneurosci.0723-14.2014] [Citation(s) in RCA: 92] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Repetitive transcranial magnetic stimulation (rTMS) is increasingly used as a treatment for neurological and psychiatric disorders. Although the induced field is focused on a target region during rTMS, adjacent areas also receive stimulation at a lower intensity and the contribution of this perifocal stimulation to network-wide effects is poorly defined. Here, we examined low-intensity rTMS (LI-rTMS)-induced changes on a model neural network using the visual systems of normal (C57Bl/6J wild-type, n = 22) and ephrin-A2A5(-/-) (n = 22) mice, the latter possessing visuotopic anomalies. Mice were treated with LI-rTMS or sham (handling control) daily for 14 d, then fluorojade and fluororuby were injected into visual cortex. The distribution of dorsal LGN (dLGN) neurons and corticotectal terminal zones (TZs) was mapped and disorder defined by comparing their actual location with that predicted by injection sites. In the afferent geniculocortical projection, LI-rTMS decreased the abnormally high dispersion of retrogradely labeled neurons in the dLGN of ephrin-A2A5(-/-) mice, indicating geniculocortical map refinement. In the corticotectal efferents, LI-rTMS improved topography of the most abnormal TZs in ephrin-A2A5(-/-) mice without altering topographically normal TZs. To investigate a possible molecular mechanism for LI-rTMS-induced structural plasticity, we measured brain derived neurotrophic factor (BDNF) in the visual cortex and superior colliculus after single and multiple stimulations. BDNF was upregulated after a single stimulation for all groups, but only sustained in the superior colliculus of ephrin-A2A5(-/-) mice. Our results show that LI-rTMS upregulates BDNF, promoting a plastic environment conducive to beneficial reorganization of abnormal cortical circuits, information that has important implications for clinical rTMS.
Collapse
|
15
|
Stuart K, Summers MJ, Valenzuela MJ, Vickers JC. BDNF and COMT polymorphisms have a limited association with episodic memory performance or engagement in complex cognitive activity in healthy older adults. Neurobiol Learn Mem 2014; 110:1-7. [PMID: 24468545 DOI: 10.1016/j.nlm.2014.01.013] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2013] [Revised: 01/15/2014] [Accepted: 01/19/2014] [Indexed: 12/14/2022]
Abstract
Cognitive decline is a major factor in lowering the quality of life in older populations, and contributes substantially to social, economic, and health costs. As humans age, cognitive function decreases differentially, and individual differences in cognitive ageing are likely attributed to a range of causes, including environmental and genetic influences. The current study included 360 participants (240 females and 120 males) aged between 50 and 79years from the Tasmanian Healthy Brain Project. The brain-derived neurotrophic factor (BDNF) Val66Met and Catechol-O-Methyltransferase (COMT) Val158Met polymorphisms were examined for their association with visual and auditory episodic memory performance. The polymorphisms were also investigated for their association with reported life-long engagement in complex cognitive activity using a retrospective questionnaire. Relative to the demographic variables, the gene variations were found to have no association with episodic memory performance, with the exception of the COMT polymorphism on a single measure of auditory memory (RAVLT). Several other studies also demonstrated that these polymorphisms have no, small, or inconsistent effects on memory function. The BDNF Val66Met and COMT Val158Met polymorphisms were also found to be of little significance to active engagement in complex cognitive activity throughout most of the lifespan. An association was detected between BDNF Val66Met and engagement in cognitive activity in early life (p=.04, d=.23), however this did not reach significance when adjusted for multiple comparisons. The biological mechanisms that underlie engagement in cognitive activity are elusive, thus the potential relationship between BDNF Val66Met genotype and early life cognitive engagement warrants further investigation.
Collapse
Affiliation(s)
- Kimberley Stuart
- Wicking Dementia Research and Education Centre, University of Tasmania, Australia; School of Medicine, University of Tasmania, Tasmania, Australia
| | - Mathew James Summers
- Wicking Dementia Research and Education Centre, University of Tasmania, Australia; School of Psychology, University of Tasmania, Tasmania, Australia.
| | - Michael J Valenzuela
- Regenerative Neuroscience Group, Brain and Mind Research Institute, University of Sydney, Australia
| | - James C Vickers
- Wicking Dementia Research and Education Centre, University of Tasmania, Australia; School of Medicine, University of Tasmania, Tasmania, Australia
| |
Collapse
|
16
|
Viscomi MT, Molinari M. Remote neurodegeneration: multiple actors for one play. Mol Neurobiol 2014; 50:368-89. [PMID: 24442481 DOI: 10.1007/s12035-013-8629-x] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2013] [Accepted: 12/24/2013] [Indexed: 12/19/2022]
Abstract
Remote neurodegeneration significantly influences the clinical outcome in many central nervous system (CNS) pathologies, such as stroke, multiple sclerosis, and traumatic brain and spinal cord injuries. Because these processes develop days or months after injury, they are accompanied by a therapeutic window of opportunity. The complexity and clinical significance of remote damage is prompting many groups to examine the factors of remote degeneration. This research is providing insights into key unanswered questions, opening new avenues for innovative neuroprotective therapies. In this review, we evaluate data from various remote degeneration models to describe the complexity of the systems that are involved and the importance of their interactions in reducing damage and promoting recovery after brain lesions. Specifically, we recapitulate the current data on remote neuronal degeneration, focusing on molecular and cellular events, as studied in stroke and brain and spinal cord injury models. Remote damage is a multifactorial phenomenon in which many components become active in specific time frames. Days, weeks, or months after injury onset, the interplay between key effectors differentially affects neuronal survival and functional outcomes. In particular, we discuss apoptosis, inflammation, oxidative damage, and autophagy-all of which mediate remote degeneration at specific times. We also review current findings on the pharmacological manipulation of remote degeneration mechanisms in reducing damage and sustaining outcomes. These novel treatments differ from those that have been proposed to limit primary lesion site damage, representing new perspectives on neuroprotection.
Collapse
Affiliation(s)
- Maria Teresa Viscomi
- Experimental Neurorehabilitation Laboratory, Santa Lucia Foundation I.R.C.C.S., Via del Fosso di Fiorano 65, 00143, Rome, Italy,
| | | |
Collapse
|
17
|
Environmental enrichment extends ocular dominance plasticity into adulthood and protects from stroke-induced impairments of plasticity. Proc Natl Acad Sci U S A 2014; 111:1150-5. [PMID: 24395770 DOI: 10.1073/pnas.1313385111] [Citation(s) in RCA: 108] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Ocular dominance (OD) plasticity in mouse primary visual cortex (V1) declines during postnatal development and is absent beyond postnatal day 110 if mice are raised in standard cages (SCs). An enriched environment (EE) promotes OD plasticity in adult rats. Here, we explored cellular mechanisms of EE in mouse V1 and the therapeutic potential of EE to prevent impairments of plasticity after a cortical stroke. Using in vivo optical imaging, we observed that monocular deprivation in adult EE mice (i) caused a very strong OD plasticity previously only observed in 4-wk-old animals, (ii) restored already lost OD plasticity in adult SC-raised mice, and (iii) preserved OD plasticity after a stroke in the primary somatosensory cortex. Using patch-clamp electrophysiology in vitro, we also show that (iv) local inhibition was significantly reduced in V1 slices of adult EE mice and (v) the GABA/AMPA ratio was like that in 4-wk-old SC-raised animals. These observations were corroborated by in vivo analyses showing that diazepam treatment significantly reduced the OD shift of EE mice after monocular deprivation. Taken together, EE extended the sensitive phase for OD plasticity into late adulthood, rejuvenated V1 after 4 mo of SC-rearing, and protected adult mice from stroke-induced impairments of cortical plasticity. The EE effect was mediated most likely by preserving low juvenile levels of inhibition into adulthood, which potentially promoted adaptive changes in cortical circuits.
Collapse
|
18
|
Ferrari E, Gu C, Niranjan D, Restani L, Rasetti-Escargueil C, Obara I, Geranton SM, Arsenault J, Goetze TA, Harper CB, Nguyen TH, Maywood E, O'Brien J, Schiavo G, Wheeler DW, Meunier FA, Hastings M, Edwardson JM, Sesardic D, Caleo M, Hunt SP, Davletov B. Synthetic self-assembling clostridial chimera for modulation of sensory functions. Bioconjug Chem 2013; 24:1750-9. [PMID: 24011174 PMCID: PMC3901392 DOI: 10.1021/bc4003103] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Clostridial neurotoxins reversibly block neuronal communication for weeks and months. While these proteolytic neurotoxins hold great promise for clinical applications and the investigation of brain function, their paralytic activity at neuromuscular junctions is a stumbling block. To redirect the clostridial activity to neuronal populations other than motor neurons, we used a new self-assembling method to combine the botulinum type A protease with the tetanus binding domain, which natively targets central neurons. The two parts were produced separately and then assembled in a site-specific way using a newly introduced 'protein stapling' technology. Atomic force microscopy imaging revealed dumbbell shaped particles which measure ∼23 nm. The stapled chimera inhibited mechanical hypersensitivity in a rat model of inflammatory pain without causing either flaccid or spastic paralysis. Moreover, the synthetic clostridial molecule was able to block neuronal activity in a defined area of visual cortex. Overall, we provide the first evidence that the protein stapling technology allows assembly of distinct proteins yielding new biomedical properties.
Collapse
Affiliation(s)
- Enrico Ferrari
- MRC Laboratory of Molecular Biology , Cambridge, United Kingdom
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
19
|
Jeanneteau F, Chao MV. Are BDNF and glucocorticoid activities calibrated? Neuroscience 2013; 239:173-95. [PMID: 23022538 PMCID: PMC3581703 DOI: 10.1016/j.neuroscience.2012.09.017] [Citation(s) in RCA: 104] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2012] [Revised: 09/04/2012] [Accepted: 09/06/2012] [Indexed: 12/22/2022]
Abstract
One hypothesis to account for the onset and severity of neurological disorders is the loss of trophic support. Indeed, changes in the levels and activities of brain-derived neurotrophic factor (BDNF) occur in numerous neurodegenerative and neuropsychiatric diseases. A deficit promotes vulnerability whereas a gain of function facilitates recovery by enhancing survival, synapse formation and synaptic plasticity. Implementation of 'BDNF therapies', however, faces numerous methodological and pharmacokinetic issues. Identifying BDNF mimetics that activate the BDNF receptor or downstream targets of BDNF signaling represent an alternative approach. One mechanism that shows great promise is to study the interplay of BDNF and glucocorticoid hormones, a major class of natural steroid secreted during stress reactions and in synchrony with circadian rhythms. While small amounts of glucocorticoids support normal brain function, excess stimulation by these steroid hormones precipitates stress-related affective disorders. To date, however, because of the paucity of knowledge of underlying cellular mechanisms, deleterious effects of glucocorticoids are not prevented following extreme stress. In the present review, we will discuss the complementary roles shared by BDNF and glucocorticoids in synaptic plasticity, and delineate possible signaling mechanisms mediating these effects.
Collapse
Affiliation(s)
- F Jeanneteau
- Skirball Institute of Biomolecular Medicine, Department of Cell Biology, NYU School of Medicine, New York, NY 10016, USA.
| | | |
Collapse
|
20
|
Abstract
Brain-derived neurotrophic factor (BDNF)--a member of a small family of secreted proteins that includes nerve growth factor, neurotrophin 3 and neurotrophin 4--has emerged as a key regulator of neural circuit development and function. The expression, secretion and actions of BDNF are directly controlled by neural activity, and secreted BDNF is capable of mediating many activity-dependent processes in the mammalian brain, including neuronal differentiation and growth, synapse formation and plasticity, and higher cognitive functions. This Review summarizes some of the recent progress in understanding the cellular and molecular mechanisms underlying neurotrophin regulation of neural circuits. The focus of the article is on BDNF, as this is the most widely expressed and studied neurotrophin in the mammalian brain.
Collapse
|
21
|
Evidence for anterograde transport and transcytosis of botulinum neurotoxin A (BoNT/A). J Neurosci 2011; 31:15650-9. [PMID: 22049408 DOI: 10.1523/jneurosci.2618-11.2011] [Citation(s) in RCA: 125] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
Botulinum neurotoxin type A (BoNT/A) is a metalloprotease that blocks synaptic transmission via the cleavage of SNAP-25 (synaptosomal-associated protein of 25 kDa). BoNT/A is successfully used in clinical neurology for the treatment of several neuromuscular pathologies and pain syndromes. Despite its widespread use, relatively little is known on BoNT/A intracellular trafficking in neurons. Using the visual pathway as a model system, here we show that catalytically active BoNT/A is capable of undergoing anterograde axonal transport and transcytosis. Following BoNT/A injection into the rat eye, significant levels of BoNT/A-cleaved SNAP-25 appeared in the retinorecipient layers of the superior colliculus (SC). Anterograde propagation of BoNT/A effects required axonal transport, ruling out a systemic spread of the toxin. Cleaved SNAP-25 was present in presynaptic structures of the tectum, but retinal terminals were devoid of the immunoreactivity, indicative of transcytosis. Experiments based on sequential administration of BoNT/A and BoNT/E showed a persistent catalytic activity of BoNT/A in tectal cells following its injection into the retina. Our findings demonstrate that catalytically active BoNT/A is anterogradely transported from the eye to the SC and transcytosed to tectal synapses. These data are important for a more complete understanding of the mechanisms of action of BoNT/A.
Collapse
|
22
|
Abstract
One of the fundamental features of retinal ganglion cells (RGCs) is that dendrites of individual RGCs are confined to one or a few narrow strata within the inner plexiform layer (IPL), and each RGC synapses only with a small group of presynaptic bipolar and amacrine cells with axons/dendrites ramified in the same strata to process distinct visual features. The underlying mechanisms which control the development of this laminar-restricted distribution pattern of RGC dendrites have been extensively studied, and it is still an open question whether the dendritic pattern of RGCs is determined by molecular cues or by activity-dependent refinement. Accumulating evidence suggests that both molecular cues and activity-dependent refinement might regulate RGC dendrites in a cell subtype-specific manner. However, identification of morphological subtypes of RGCs before they have achieved their mature dendritic pattern is a major challenge in the study of RGC dendritic development. This problem is now being circumvented through the use of molecular markers in genetically engineered mouse lines to identify RGC subsets early during development. Another unanswered fundamental question in the study of activity-dependent refinement of RGC dendrites is how changes in synaptic activity lead to the changes in dendritic morphology. Recent studies have started to shed light on the molecular basis of activity-dependent dendritic refinement of RGCs by showing that some molecular cascades control the cytoskeleton reorganization of RGCs.
Collapse
Affiliation(s)
- Ning Tian
- Department of Ophthalmology and Visual Science, University of Utah School of Medicine, Salt Lake City, USA.
| |
Collapse
|
23
|
Cerri C, Restani L, Caleo M. Callosal contribution to ocular dominance in rat primary visual cortex. Eur J Neurosci 2010; 32:1163-9. [PMID: 20726891 DOI: 10.1111/j.1460-9568.2010.07363.x] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Ocular dominance (OD) plasticity triggered by monocular eyelid suture is a classic paradigm for studying experience-dependent changes in neural connectivity. Recently, rodents have become the most popular model for studies of OD plasticity. It is therefore important to determine how OD is determined in the rodent primary visual cortex. In particular, cortical cells receive considerable inputs from the contralateral hemisphere via callosal axons, but the role of these connections in controlling eye preference remains controversial. Here we have examined the role of callosal connections in binocularity of the visual cortex in naïve young rats. We recorded cortical responses evoked by stimulation of each eye before and after acute silencing, via stereotaxic tetrodotoxin (TTX) injection, of the lateral geniculate nucleus ipsilateral to the recording site. This protocol allowed us to isolate visual responses transmitted via the corpus callosum. Cortical binocularity was assessed by visual evoked potential (VEP) and single-unit recordings. We found that acute silencing of afferent geniculocortical input produced a very significant reduction in the contralateral-to-ipsilateral (C/I) VEP ratio, and a marked shift towards the ipsilateral eye in the OD distribution of cortical cells. Analysis of absolute strength of each eye indicated a dramatic decrease in contralateral eye responses following TTX, while those of the ipsilateral eye were reduced but maintained a more evident input. We conclude that callosal connections contribute to normal OD mainly by carrying visual input from the ipsilateral eye. These data have important implications for the interpretation of OD plasticity following alterations of visual experience.
Collapse
|
24
|
Restani L, Cerri C, Pietrasanta M, Gianfranceschi L, Maffei L, Caleo M. Functional masking of deprived eye responses by callosal input during ocular dominance plasticity. Neuron 2010; 64:707-18. [PMID: 20005826 DOI: 10.1016/j.neuron.2009.10.019] [Citation(s) in RCA: 61] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/14/2009] [Indexed: 10/20/2022]
Abstract
Monocular deprivation (MD) is a well-known paradigm of experience-dependent plasticity in which cortical neurons exhibit a shift of ocular dominance (OD) toward the open eye. The mechanisms underlying this form of plasticity are incompletely understood. Here we demonstrate the involvement of callosal connections in the synaptic modifications occurring during MD. Rats at the peak of the critical period were deprived for 7 days, resulting in the expected OD shift toward the open eye. Acute microinjection of the activity blocker muscimol into the visual cortex contralateral to the recording site restored binocularity of cortical cells. Continuous silencing of callosal input throughout the period of MD also resulted in substantial attenuation of the OD shift. Blockade of interhemispheric communication selectively enhanced deprived eye responses with no effect on open eye-driven activity. We conclude that callosal inputs play a key role in functional weakening of less active connections during OD plasticity.
Collapse
Affiliation(s)
- Laura Restani
- Istituto di Neuroscienze, Consiglio Nazionale delle Ricerche, via G. Moruzzi 1, 56100 Pisa, Italy
| | | | | | | | | | | |
Collapse
|
25
|
Setting the pace for retinal development: environmental enrichment acts through insulin-like growth factor 1 and brain-derived neurotrophic factor. J Neurosci 2009; 29:10809-19. [PMID: 19726638 DOI: 10.1523/jneurosci.1857-09.2009] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Environmental enrichment strongly affects visual system maturation both at retinal and cortical levels. Which molecular pathways are activated by an enriched environment (EE) to regulate visual system development has not been clarified. Here, we show that early [postnatal day 1 (P1) to P7] insulin-like growth factor 1 (IGF-1) injections in the eyes of non-EE rat pups mimic EE effects both in increasing BDNF levels in the retinal ganglion cell layer at P10 and in determining a more adult-like retinal acuity, assessed with pattern electroretinogram at P25. Blocking IGF-1 action in EE animals during the same early postnatal time window by injecting the IGF-1 receptor antagonist JB1 prevents EE effects both on BDNF expression and on retinal acuity maturation. Reducing BDNF expression in the retina of IGF-1-treated rats prevents IGF-1 effects on retinal acuity development. Finally, we show that tyrosine hydroxylase (TH) expression is increased in the retina of P10 EE and IGF-1-treated rats and that blocking TH expression in EE animals prevents EE from affecting retinal acuity development. Thus, early levels of IGF-1 play a key role in mediating EE effects on retinal development through an action that requires BDNF and involves dopaminergic amacrine cell network.
Collapse
|
26
|
Wong J, Webster MJ, Cassano H, Weickert CS. Changes in alternative brain-derived neurotrophic factor transcript expression in the developing human prefrontal cortex. Eur J Neurosci 2009; 29:1311-22. [PMID: 19519623 DOI: 10.1111/j.1460-9568.2009.06669.x] [Citation(s) in RCA: 57] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
In this study, we determined when and through which promoter brain-derived neurotrophic factor (BDNF) transcription is regulated during the protracted period of human frontal cortex development. Using quantitative real-time polymerase chain reaction, we examined the expression of the four most abundant alternative 5' exons of the BDNF gene (exons I, II, IV, and VI) in RNA extracted from the prefrontal cortex. We found that expression of transcripts I-IX and VI-IX was highest during infancy, whereas that of transcript II-IX was lowest just after birth, slowly increasing to reach a peak in toddlers. Transcript IV-IX was significantly upregulated within the first year of life, and was maintained at this level until school age. Quantification of BDNF protein revealed that levels followed a similar developmental pattern as transcript IV-IX. In situ hybridization of mRNA in cortical sections showed the highest expression in layers V and VI for all four BDNF transcripts, whereas moderate expression was observed in layers II and III. Interestingly, although low expression of BDNF was observed in cortical layer IV, this BDNF mRNA low-zone decreased in prominence with age and showed an increase in neuronal mRNA localization. In summary, our findings show that dynamic regulation of BDNF expression occurs through differential use of alternative promoters during the development of the human prefrontal cortex, particularly in the younger age groups, when the prefrontal cortex is more plastic.
Collapse
Affiliation(s)
- Jenny Wong
- Schizophrenia Research Institute, Sydney, Australia
| | | | | | | |
Collapse
|
27
|
Fujita R, Ueda M, Fujiwara K, Ueda H. Prothymosin-alpha plays a defensive role in retinal ischemia through necrosis and apoptosis inhibition. Cell Death Differ 2008; 16:349-58. [PMID: 18989338 DOI: 10.1038/cdd.2008.159] [Citation(s) in RCA: 57] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
Prothymosin-alpha (ProTalpha) causes a switch in cell death mode from necrosis to neurotrophin-reversible apoptosis in primary cultured cortical neurons. In the present study, post-ischemic administration (3 or 24 h, intravenously) of recombinant mouse ProTalpha without neurotrophins completely prevented ischemia-induced retinal damage accompanying necrosis and apoptosis, as well as dysfunction assessed by electroretinogram. Treatments with anti-erythropoietin (EPO) or brain-derived neurotrophic factor (BDNF) immunoglobulin G (IgG) reversed ProTalpha-induced inhibition of apoptosis. ProTalpha upregulated retinal EPO and BDNF levels in the presence of ischemia. Moreover, intravitreous administration of anti-ProTalpha IgG or an antisense oligodeoxynucleotide for ProTalpha accelerated ischemia-induced retinal damage. We also observed that ischemia treatment caused a depletion of ProTalpha from retinal cells. Altogether, these results suggest that the systemic administration of ProTalpha switches ischemia-induced necrosis to apoptosis, which in turn is inhibited by neurotrophic factors upregulated by ProTalpha and ischemia. ProTalpha released upon ischemic stress was found to have a defensive role in retinal ischemia.
Collapse
Affiliation(s)
- R Fujita
- Division of Molecular Pharmacology and Neuroscience, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | | | | | | |
Collapse
|
28
|
Experience-Dependent Transfer of Otx2 Homeoprotein into the Visual Cortex Activates Postnatal Plasticity. Cell 2008; 134:508-20. [DOI: 10.1016/j.cell.2008.05.054] [Citation(s) in RCA: 380] [Impact Index Per Article: 22.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2007] [Revised: 04/22/2008] [Accepted: 05/05/2008] [Indexed: 11/21/2022]
|
29
|
Xu HP, Tian N. Glycine receptor-mediated synaptic transmission regulates the maturation of ganglion cell synaptic connectivity. J Comp Neurol 2008; 509:53-71. [PMID: 18425804 PMCID: PMC2952449 DOI: 10.1002/cne.21727] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
It is well documented that neuronal activity is required for the developmental segregation of retinal ganglion cell (RGC) synaptic connectivity with ON and OFF bipolar cells in mammalian retina. Our recent study showed that light deprivation preferentially blocked the developmental RGC dendritic redistribution from the center to sublamina a of the inner plexiform layer (IPL). To determine whether OFF signals in visual stimulation are required for OFF RGC dendritic development, the light-evoked responses and dendritic stratification patterns of RGCs in Spastic mutant mice, in which the OFF signal transmission in the rod pathway is largely blocked due to a reduction of glycine receptor (GlyR) expression, were quantitatively studied at different ages and rearing conditions. The dendritic distribution in the IPL of these mice was indistinguishable from wildtype controls at the age of postnatal day (P)12. However, the adult Spastic mutants had altered RGC light-evoked synaptic inputs from ON and OFF pathways, which could not be mimicked by pharmacologically blocking of glycinergic synaptic transmission on age-matched wildtype animals. Spastic mutation also blocked the developmental redistribution of RGC dendrites from the center to sublamina a of the IPL, which mimicked the effects induced by light deprivation on wildtype animals. Moreover, light deprivation of the Spastic mutants had no additional impact on the RGC dendritic distribution and light response patterns. We interpret these results as that visual stimulation regulates the maturation of RGC synaptic activity and connectivity primarily through GlyR-mediated synaptic transmission.
Collapse
Affiliation(s)
- Hong-ping Xu
- Department of Ophthalmology and Visual Science, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Ning Tian
- Department of Ophthalmology and Visual Science, Yale University School of Medicine, New Haven, CT 06520, USA
- Department of Neurobiology, Yale University School of Medicine, New Haven, CT 06520, USA
| |
Collapse
|
30
|
Grishanin RN, Yang H, Liu X, Donohue-Rolfe K, Nune GC, Zang K, Xu B, Duncan JL, LaVail MM, Copenhagen DR, Reichardt LF. Retinal TrkB receptors regulate neural development in the inner, but not outer, retina. Mol Cell Neurosci 2008; 38:431-43. [PMID: 18511296 PMCID: PMC2849660 DOI: 10.1016/j.mcn.2008.04.004] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2007] [Revised: 04/05/2008] [Accepted: 04/09/2008] [Indexed: 01/19/2023] Open
Abstract
BDNF signaling through its TrkB receptor plays a pivotal role in activity-dependent refinement of synaptic connectivity of retinal ganglion cells. Additionally, studies using TrkB knockout mice have suggested that BDNF/TrkB signaling is essential for the development of photoreceptors and for synaptic communication between photoreceptors and second order retinal neurons. Thus the action of BDNF on refinement of synaptic connectivity of retinal ganglion cells could be a direct effect in the inner retina, or it could be secondary to its proposed role in rod maturation and in the formation of rod to bipolar cell synaptic transmission. To address this matter we have conditionally eliminated TrkB within the retina. We find that rod function and synaptic transmission to bipolar cells is not compromised in these conditional knockout mice. Consistent with previous work, we find that inner retina neural development is regulated by retinal BDNF/TrkB signaling. Specifically we show here also that the complexity of neuronal processes of dopaminergic cells is reduced in conditional TrkB knockout mice. We conclude that retinal BDNF/TrkB signaling has its primary role in the development of inner retinal neuronal circuits, and that this action is not a secondary effect due to the loss of visual signaling in the outer retina.
Collapse
Affiliation(s)
- Ruslan N. Grishanin
- Neuroscience Program, School of Medicine, University of California San Francisco, San Francisco, California 94143
- Department of Physiology, School of Medicine, University of California San Francisco, San Francisco, California 94143
| | - Haidong Yang
- Beckman Vision Center, Department of Ophthalmology, School of Medicine, University of California San Francisco, San Francisco, California 94143
| | - Xiaorong Liu
- Department of Physiology, School of Medicine, University of California San Francisco, San Francisco, California 94143
- Beckman Vision Center, Department of Ophthalmology, School of Medicine, University of California San Francisco, San Francisco, California 94143
| | - Kate Donohue-Rolfe
- Beckman Vision Center, Department of Ophthalmology, School of Medicine, University of California San Francisco, San Francisco, California 94143
| | - George C. Nune
- Beckman Vision Center, Department of Ophthalmology, School of Medicine, University of California San Francisco, San Francisco, California 94143
| | - Keling Zang
- Department of Physiology, School of Medicine, University of California San Francisco, San Francisco, California 94143
| | - Baoji Xu
- Pharmacology Department, Georgetown University, Washington, DC 20057
| | - Jacque L. Duncan
- Beckman Vision Center, Department of Ophthalmology, School of Medicine, University of California San Francisco, San Francisco, California 94143
| | - Matthew M. LaVail
- Neuroscience Program, School of Medicine, University of California San Francisco, San Francisco, California 94143
- Beckman Vision Center, Department of Ophthalmology, School of Medicine, University of California San Francisco, San Francisco, California 94143
| | - David R. Copenhagen
- Neuroscience Program, School of Medicine, University of California San Francisco, San Francisco, California 94143
- Department of Physiology, School of Medicine, University of California San Francisco, San Francisco, California 94143
- Beckman Vision Center, Department of Ophthalmology, School of Medicine, University of California San Francisco, San Francisco, California 94143
| | - Louis F. Reichardt
- Neuroscience Program, School of Medicine, University of California San Francisco, San Francisco, California 94143
- Department of Physiology, School of Medicine, University of California San Francisco, San Francisco, California 94143
| |
Collapse
|
31
|
Butowt R, von Bartheld CS. Conventional kinesin-I motors participate in the anterograde axonal transport of neurotrophins in the visual system. J Neurosci Res 2008; 85:2546-56. [PMID: 17243173 DOI: 10.1002/jnr.21165] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Retinal ganglion cells (RGCs) anterogradely transport neurotrophins to the midbrain tectum/superior colliculus with significant downstream effects. The molecular mechanism of this type of axonal transport of neurotrophins is not well characterized. We identified kinesin-I proteins as a motor participating in the anterograde axonal movement of vesicular structures containing radiolabeled neurotrophins along the optic nerve. RT-PCR analysis of purified murine RGCs showed that adult RGCs express all known members of the kinesin-I family. After intraocular injection of (125)I-brain-derived neurotrophic factor (BDNF) into the adult mouse or (125)I-neurotrophin-3 (NT-3) into the embryonic chicken eye, radioactivity was efficiently immunoprecipitated from the optic nerve lysates by anti-kinesin heavy chain and anti-kinesin light chain monoclonal antibodies (H2 and L1). Immunoreactivity for the BDNF receptor trkB is also present in the immunoprecipitates obtained by the anti-kinesin-I antibodies. The delivery of the H2 antibody in vivo into the mouse RGCs substantially reduced anterograde axonal transport of (125)I-BDNF. Anterograde transport of BDNF was not diminished in kinesin light chain 1 (KLC1) knockout mice. However, this may be due to redundancy in functions between two different isoforms of KLC present in the RGCs, as it was described previously for kinesin heavy chains (Kanai et al. [ 2000] J Neurosci 20:6374-6384). These data indicate that kinesin-I is a protein motor that participates in the anterograde axonal transport of neurotrophins in the chicken and mouse visual pathways.
Collapse
Affiliation(s)
- Rafal Butowt
- Department of Physiology and Cell Biology, University of Nevada School of Medicine, Reno, Nevada, USA.
| | | |
Collapse
|
32
|
Fujieda H, Sasaki H. Expression of brain-derived neurotrophic factor in cholinergic and dopaminergic amacrine cells in the rat retina and the effects of constant light rearing. Exp Eye Res 2007; 86:335-43. [PMID: 18093585 DOI: 10.1016/j.exer.2007.11.005] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2007] [Revised: 10/26/2007] [Accepted: 11/08/2007] [Indexed: 10/22/2022]
Abstract
Brain-derived neurotrophic factor (BDNF) regulates many aspects of neuronal development, including survival, axonal and dendritic growth and synapse formation. Despite recent advances in our understanding of the functional significance of BDNF in retinal development, the retinal cell types expressing BDNF remains poorly defined. The goal of the present study was to determine the localization of BDNF in the mammalian retina, with special focus on the subtypes of amacrine cells, and to characterize, at the cellular level, the effects of constant light exposure during early postnatal period on retinal expression of BDNF. Retinas from 3-week-old rats reared in a normal light cycle or constant light were subjected to double immunofluorescence staining using antibodies to BDNF and retinal cell markers. BDNF immunoreactivity was localized to ganglion cells, cholinergic amacrine cells and dopaminergic amacrine cells, but not to AII amacrine cells regardless of rearing conditions. Approximately 75% of BDNF-positive cells in the inner nuclear layer were cholinergic amacrine cells in animals reared in a normal lighting condition. While BDNF immunoreactivity in ganglion cells and cholinergic amacrine cells was significantly increased by constant light rearing, which in dopaminergic amacrine cells was apparently unaltered. The overall structure of the retina and the density of ganglion cells, cholinergic amacrine cells and AII amacrine cells were unaffected by rearing conditions, whereas the density of dopaminergic amacrine cells was significantly increased by constant light rearing. The present results indicate that cholinergic amacrine cells are the primary source of BDNF in the inner nuclear layer of the rat retina and provide the first evidence that cholinergic amacrine cells may be involved in the visual activity-dependent regulation of retinal development through the production of BDNF. The present data also suggest that the production or survival of dopaminergic amacrine cells is regulated by early visual experience.
Collapse
Affiliation(s)
- Hiroki Fujieda
- Department of Anatomy, School of Medicine, Tokyo Women's Medical University, Tokyo, Japan.
| | | |
Collapse
|
33
|
Liu X, Grishanin RN, Tolwani RJ, Rentería RC, Xu B, Reichardt LF, Copenhagen DR. Brain-derived neurotrophic factor and TrkB modulate visual experience-dependent refinement of neuronal pathways in retina. J Neurosci 2007; 27:7256-67. [PMID: 17611278 PMCID: PMC2579893 DOI: 10.1523/jneurosci.0779-07.2007] [Citation(s) in RCA: 67] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2007] [Revised: 05/25/2007] [Accepted: 05/26/2007] [Indexed: 01/19/2023] Open
Abstract
Sensory experience refines neuronal structure and functionality. The visual system has proved to be a productive model system to study this plasticity. In the neonatal retina, the dendritic arbors of a large proportion of ganglion cells are diffuse in the inner plexiform layer. With maturation, many of these arbors become monolaminated. Visual deprivation suppresses this remodeling. Little is known of the molecular mechanisms controlling maturational and experience-dependent refinement. Here, we tested the hypothesis that brain-derived neurotrophic factor (BDNF), which is known to regulate dendritic branching and synaptic function in the brain, modulates the developmental and visual experience-dependent refinement of retinal ganglion cells. We used a transgenic mouse line, in which a small number of ganglion cells were labeled with yellow fluorescence protein, to delineate their dendritic structure in vivo. We found that transgenic overexpression of BDNF accelerated the laminar refinement of ganglion cell dendrites, whereas decreased TrkB expression or retina-specific deletion of TrkB, the cognate receptor for BDNF, retarded it. BDNF-TrkB signaling regulated the maturational formation of new branches in ON but not the bilaminated ON-OFF ganglion cells. Furthermore, BDNF overexpression overrides the requirement for visual inputs to stimulate laminar refinement and dendritic branching of ganglion cells. These experiments reveal a previously unrecognized action of BDNF and TrkB in controlling cell-specific, experience-dependent remodeling of neuronal structures in the visual system.
Collapse
Affiliation(s)
- Xiaorong Liu
- Department of Ophthalmology
- Department of Physiology
- Program in Neuroscience, and
| | | | - Ravi J. Tolwani
- Department of Comparative Medicine, Stanford University School of Medicine, Stanford, California 94305, and
| | - René C. Rentería
- Department of Ophthalmology
- Department of Physiology
- Program in Neuroscience, and
| | - Baoji Xu
- Department of Pharmacology, Georgetown University Medical School, Washington, DC 20057
| | - Louis F. Reichardt
- Department of Physiology
- Program in Neuroscience, and
- Howard Hughes Medical Institute, University of California, San Francisco, San Francisco, California 94143
| | - David R. Copenhagen
- Department of Ophthalmology
- Department of Physiology
- Program in Neuroscience, and
| |
Collapse
|
34
|
Marshak S, Nikolakopoulou AM, Dirks R, Martens GJ, Cohen-Cory S. Cell-autonomous TrkB signaling in presynaptic retinal ganglion cells mediates axon arbor growth and synapse maturation during the establishment of retinotectal synaptic connectivity. J Neurosci 2007; 27:2444-56. [PMID: 17344382 PMCID: PMC6672515 DOI: 10.1523/jneurosci.4434-06.2007] [Citation(s) in RCA: 71] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
BDNF contributes to the activity-dependent establishment and refinement of visual connectivity. In Xenopus, BDNF applications in the optic tectum influence retinal ganglion cell (RGC) axon branching and promote synapse formation and stabilization. The expression patterns of BDNF and TrkB suggest that BDNF specifically regulates the maturation of RGC axons at the target. It is possible, however, that BDNF modulates retinotectal synaptic connectivity by differentially influencing presynaptic RGC axons and postsynaptic tectal cells. Here, we combined single-cell expression of a dominant-negative TrkB-enhanced green fluorescent protein (GFP) fusion protein with confocal microscopy imaging in live Xenopus tadpoles to differentiate between presynaptic and postsynaptic actions of BDNF. Disruption of TrkB signaling in individual RGCs influenced the branching and synaptic maturation of presynaptic axon arbors. Specifically, GFP-TrkB.T1 overexpression increased the proportion of axons with immature, growth cone-like morphology, decreased axon branch stability, and increased axon arbor degeneration. In addition, GFP-TrkB.T1 overexpression reduced the number of red fluorescent protein-synaptobrevin-labeled presynaptic specializations per axon terminal. In contrast, overexpression of GFP-TrkB.T1 in tectal neurons did not alter synaptic number or the morphology or dynamic behavior of their dendritic arbors. Electron microscopy analysis revealed a significant decrease in the number of mature synaptic profiles and in the number of docked synaptic vesicles at retinotectal synapses made by RGC axons expressing GFP-TrkB.T1. Together, our results demonstrate that presynaptic TrkB signaling in RGCs is a key determinant in the establishment of visual connectivity and indicate that changes in tectal neuron synaptic connectivity are secondary to the BDNF-elicited enhanced stability and growth of presynaptic RGCs.
Collapse
Affiliation(s)
- Sonya Marshak
- Department of Neurobiology and Behavior, University of California, Irvine, Irvine, California 92697, and
| | | | - Ron Dirks
- Department of Molecular Animal Physiology, Nijmegen Center for Molecular Life Sciences, Radboud University Nijmegen, 6525 GA Nijmegen, The Netherlands
| | - Gerard J. Martens
- Department of Molecular Animal Physiology, Nijmegen Center for Molecular Life Sciences, Radboud University Nijmegen, 6525 GA Nijmegen, The Netherlands
| | - Susana Cohen-Cory
- Department of Neurobiology and Behavior, University of California, Irvine, Irvine, California 92697, and
| |
Collapse
|
35
|
Landi S, Cenni MC, Maffei L, Berardi N. Environmental enrichment effects on development of retinal ganglion cell dendritic stratification require retinal BDNF. PLoS One 2007; 2:e346. [PMID: 17406670 PMCID: PMC1829175 DOI: 10.1371/journal.pone.0000346] [Citation(s) in RCA: 56] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2006] [Accepted: 03/12/2007] [Indexed: 11/18/2022] Open
Abstract
A well-known developmental event of retinal maturation is the progressive segregation of retinal ganglion cell (RGC) dendrites into a and b sublaminae of the inner plexiform layer (IPL), a morphological rearrangement crucial for the emergence of the ON and OFF pathways. The factors regulating this process are not known, although electrical activity has been demonstrated to play a role. Here we report that Environmental Enrichment (EE) accelerates the developmental segregation of RGC dendrites and prevents the effects exerted on it by dark rearing (DR). Development of RGC stratification was analyzed in a line of transgenic mice expressing plasma-membrane marker green fluorescent protein (GFP) under the control of Thy-1 promoter; we visualized the a and b sublaminae of the IPL by using an antibody selectively directed against a specific marker of cholinergic neurons. EE precociously increases Brain Derived Neurotrophic Factor (BDNF) in the retina, in parallel with the precocious segregation of RGC dendrites; in addition, EE counteracts retinal BDNF reduction in DR retinas and promotes a normal segregation of RGC dendrites. Blocking retinal BDNF by means of antisense oligos blocks EE effects on the maturation of RGC dendritic stratification. Thus, EE affects the development of RGC dendritic segregation and retinal BDNF is required for this effect to take place, suggesting that BDNF could play an important role in the emergence of the ON and OFF pathways.
Collapse
|
36
|
Landi S, Sale A, Berardi N, Viegi A, Maffei L, Cenni MC. Retinal functional development is sensitive to environmental enrichment: a role for BDNF. FASEB J 2006; 21:130-9. [PMID: 17135370 DOI: 10.1096/fj.06-6083com] [Citation(s) in RCA: 70] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Retina has long been considered less plastic than cortex or hippocampus, the very sites of experience-dependent plasticity. Now, we show that retinal development is responsive to the experience provided by an enriched environment (EE): the maturation of retinal acuity, which is a sensitive index of retinal circuitry development, is strongly accelerated in EE rats. This effect is present also in rats exposed to EE up to P10, that is before eye opening, suggesting that factors sufficient to trigger retinal acuity development are affected by EE during the first days of life. Brain derived neurotrophic factor (BDNF) is precociously expressed in the ganglion cell layer of EE with respect to non-EE rats and reduction of BDNF expression in EE animals counteracts EE effects on retinal acuity. Thus, EE controls the development of retinal circuitry, and this action depends on retinal BDNF expression.
Collapse
Affiliation(s)
- S Landi
- Laboratorio di Neurobiologia, Scuola Normale Superiore c/o Istituto di Neuroscienze del CNR, Via G. Moruzzi, 1, 56100 Pisa, Italy.
| | | | | | | | | | | |
Collapse
|
37
|
Hofer SB, Mrsic-Flogel TD, Bonhoeffer T, Hübener M. Lifelong learning: ocular dominance plasticity in mouse visual cortex. Curr Opin Neurobiol 2006; 16:451-9. [PMID: 16837188 DOI: 10.1016/j.conb.2006.06.007] [Citation(s) in RCA: 100] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2006] [Accepted: 06/29/2006] [Indexed: 11/28/2022]
Abstract
Ocular dominance plasticity has long served as a successful model for examining how cortical circuits are shaped by experience. In this paradigm, altered retinal activity caused by unilateral eye-lid closure leads to dramatic shifts in the binocular response properties of neurons in the visual cortex. Much of the recent progress in identifying the cellular and molecular mechanisms underlying ocular dominance plasticity has been achieved by using the mouse as a model system. In this species, monocular deprivation initiated in adulthood also causes robust ocular dominance shifts. Research on ocular dominance plasticity in the mouse is starting to provide insight into which factors mediate and influence cortical plasticity in juvenile and adult animals.
Collapse
Affiliation(s)
- Sonja B Hofer
- Max-Planck-Institut für Neurobiologie, Am Klopferspitz 18, 82152 Martinsried, Germany
| | | | | | | |
Collapse
|
38
|
Avwenagha O, Bird MM, Lieberman AR, Yan Q, Campbell G. Patterns of expression of brain-derived neurotrophic factor and tyrosine kinase B mRNAs and distribution and ultrastructural localization of their proteins in the visual pathway of the adult rat. Neuroscience 2006; 140:913-28. [PMID: 16626872 DOI: 10.1016/j.neuroscience.2006.02.056] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2005] [Revised: 02/23/2006] [Accepted: 02/23/2006] [Indexed: 01/19/2023]
Abstract
We have examined the cellular and subcellular distribution and the patterns of expression of brain-derived neurotrophic factor (BDNF), and of its high affinity receptor, tyrosine kinase B (TrkB), in retinorecipient regions of the brain, including the superior colliculus, the lateral geniculate nucleus and the olivary pretectal nucleus. In the retinorecipient layers of the superior colliculus, BDNF protein and mRNA were present in the cell bodies of a subpopulation of neurons, and BDNF protein was present in the neuropil as punctate or fiber-like structures. In the lateral geniculate nucleus, however, BDNF mRNA was not detected, and BDNF protein was restricted to punctate and fiber-like structures in the neuropil, especially in the most superficial part of the dorsal lateral geniculate nucleus, just below the optic tract. At the ultrastructural level, BDNF protein was localized predominantly to axon terminals containing round synaptic vesicles and pale mitochondria with irregular cristae, which made asymmetric (Gray type I) synaptic specializations (R-boutons). Enucleation of one eye was followed by loss of BDNF immunoreactivity and disappearance of BDNF-positive R-boutons in the contralateral visual centers, confirming the retinal origin of at least most of these terminals. TrkB was present in postsynaptic densities apposed to immunoreactive R-boutons in the superior colliculus and lateral geniculate nucleus, and was also associated with axonal and dendritic microtubules. These findings suggest that BDNF is synthesized by a subpopulation of retinal ganglion cells and axonally transported to visual centers where this neurotrophin is assumed to play important roles in visual system maintenance and/or in modulating the excitatory retinal input to neurons in these centers.
Collapse
Affiliation(s)
- O Avwenagha
- Department of Anatomy and Developmental Biology, University College London, Gower Street, London WC1E 6BT, UK
| | | | | | | | | |
Collapse
|