1
|
Nittoli V, Sepe RM, Coppola U, D'Agostino Y, De Felice E, Palladino A, Vassalli QA, Locascio A, Ristoratore F, Spagnuolo A, D'Aniello S, Sordino P. A comprehensive analysis of neurotrophins and neurotrophin tyrosine kinase receptors expression during development of zebrafish. J Comp Neurol 2018; 526:1057-1072. [DOI: 10.1002/cne.24391] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2017] [Revised: 11/30/2017] [Accepted: 12/18/2017] [Indexed: 01/01/2023]
Affiliation(s)
- Valeria Nittoli
- Department of Biology and Evolution of Marine Organisms; Stazione Zoologica Anton Dohrn, Villa Comunale; Naples 80121 Italy
| | - Rosa M. Sepe
- Department of Biology and Evolution of Marine Organisms; Stazione Zoologica Anton Dohrn, Villa Comunale; Naples 80121 Italy
| | - Ugo Coppola
- Department of Biology and Evolution of Marine Organisms; Stazione Zoologica Anton Dohrn, Villa Comunale; Naples 80121 Italy
| | - Ylenia D'Agostino
- Department of Biology and Evolution of Marine Organisms; Stazione Zoologica Anton Dohrn, Villa Comunale; Naples 80121 Italy
| | - Elena De Felice
- Department of Biology and Evolution of Marine Organisms; Stazione Zoologica Anton Dohrn, Villa Comunale; Naples 80121 Italy
| | - Antonio Palladino
- Department of Biology and Evolution of Marine Organisms; Stazione Zoologica Anton Dohrn, Villa Comunale; Naples 80121 Italy
| | - Quirino A. Vassalli
- Department of Biology and Evolution of Marine Organisms; Stazione Zoologica Anton Dohrn, Villa Comunale; Naples 80121 Italy
| | - Annamaria Locascio
- Department of Biology and Evolution of Marine Organisms; Stazione Zoologica Anton Dohrn, Villa Comunale; Naples 80121 Italy
| | - Filomena Ristoratore
- Department of Biology and Evolution of Marine Organisms; Stazione Zoologica Anton Dohrn, Villa Comunale; Naples 80121 Italy
| | - Antonietta Spagnuolo
- Department of Biology and Evolution of Marine Organisms; Stazione Zoologica Anton Dohrn, Villa Comunale; Naples 80121 Italy
| | - Salvatore D'Aniello
- Department of Biology and Evolution of Marine Organisms; Stazione Zoologica Anton Dohrn, Villa Comunale; Naples 80121 Italy
| | - Paolo Sordino
- Department of Biology and Evolution of Marine Organisms; Stazione Zoologica Anton Dohrn, Villa Comunale; Naples 80121 Italy
| |
Collapse
|
2
|
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: 3.6] [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
|
3
|
Matsumoto H, Kataoka K, Tsoka P, Connor KM, Miller JW, Vavvas DG. Strain difference in photoreceptor cell death after retinal detachment in mice. Invest Ophthalmol Vis Sci 2014; 55:4165-74. [PMID: 24854853 DOI: 10.1167/iovs.14-14238] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
PURPOSE To evaluate the potential for mouse genetic background to effect photoreceptor cell death in response to experimental retinal detachment (RD). METHODS Retinal detachment was induced in three inbred mouse strains (C57BL/6, BALB/c, and B6129SF2) by subretinal injection of sodium hyaluronate. A time course of photoreceptor cell death was assessed by TUNEL assay. Total photoreceptor cell death was analyzed through comparing the outer nuclear layer (ONL)/inner nuclear layer (INL) ratio 7 days post RD. Western blot analysis or quantitative real-time PCR (qPCR) were performed to assess cell death signaling, expression of endogenous neurotrophin, and levels of apoptosis inhibitors 24 hours after RD. Inflammatory cytokine secretion and inflammatory cell infiltration were quantified by ELISA and immunostaining, respectively. RESULTS The peak of photoreceptor cell death after RD was at 24 hours in all strains. Photoreceptor cell death as well as monocyte chemoattractant protein 1 and interleukin 6 secretion at 24 hours after RD was the highest in BALB/c, followed in order of magnitude by C57BL/6 and B6129SF2. Conversely, nerve growth factor expression and ONL/INL ratio were the lowest in BALB/c. Apoptosis signaling was higher in C57BL/6, whereas necroptosis signaling was higher in C57BL/6 and BALB/c. Autophagic signaling was higher in BALB/c. X-linked inhibitor of apoptosis (XIAP) and survivin protein levels were lower in C57BL/6 and BALB/c, respectively. Macrophage/microglia infiltration was higher in C57BL/6 and BALB/c at 24 hours after RD. CONCLUSIONS Photoreceptor cell death after RD was significantly different among the three strains, suggesting the presence of genetic factors that affect photoreceptor cell death after RD.
Collapse
Affiliation(s)
- Hidetaka Matsumoto
- Retina Service, Angiogenesis Laboratory, Massachusetts Eye and Ear Infirmary, Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts, United States
| | - Keiko Kataoka
- Retina Service, Angiogenesis Laboratory, Massachusetts Eye and Ear Infirmary, Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts, United States
| | - Pavlina Tsoka
- Retina Service, Angiogenesis Laboratory, Massachusetts Eye and Ear Infirmary, Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts, United States
| | - Kip M Connor
- Retina Service, Angiogenesis Laboratory, Massachusetts Eye and Ear Infirmary, Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts, United States
| | - Joan W Miller
- Retina Service, Angiogenesis Laboratory, Massachusetts Eye and Ear Infirmary, Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts, United States
| | - Demetrios G Vavvas
- Retina Service, Angiogenesis Laboratory, Massachusetts Eye and Ear Infirmary, Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts, United States
| |
Collapse
|
4
|
De Felice E, Porreca I, Alleva E, De Girolamo P, Ambrosino C, Ciriaco E, Germanà A, Sordino P. Localization of BDNF expression in the developing brain of zebrafish. J Anat 2014; 224:564-74. [PMID: 24588510 DOI: 10.1111/joa.12168] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/27/2014] [Indexed: 12/14/2022] Open
Abstract
The brain-derived neurotrophic factor (BDNF) gene is expressed in differentiating and post-mitotic neurons of the zebrafish embryo, where it has been implicated in Huntington's disease. Little is known, however, about the full complement of neuronal cell types that express BDNF in this important vertebrate model. Here, we further explored the transcriptional profiles during the first week of development using real-time quantitative polymerase chain reaction (RT-qPCR) and whole-mount in situ hybridization (WISH). RT-qPCR results revealed a high level of maternal contribution followed by a steady increase of zygotic transcription, consistent with the notion of a prominent role of BDNF in neuronal maturation and maintenance. Based on WISH, we demonstrate for the first time that BDNF expression in the developing brain of zebrafish is structure specific. Anatomical criteria and co-staining with genetic markers (shh, pax2a, emx1, krox20, lhx2b and lhx9) visualized major topological domains of BDNF-positive cells in the pallium, hypothalamus, posterior tuberculum and optic tectum. Moreover, the relative timing of BDNF transcription in the eye and tectum may illustrate a mechanism for coordinated development of the retinotectal system. Taken together, our results are compatible with a local delivery and early role of BDNF in the developing brain of zebrafish, adding basic knowledge to the study of neurotrophin functions in neural development and disease.
Collapse
Affiliation(s)
- E De Felice
- Laboratory of Cellular and Developmental Biology, Stazione Zoologica Anton Dohrn, Naples, Italy; Department of Morphology, Biochemistry, Physiology and Animal Productions, Section of Morphology, University of Messina, Messina, Italy
| | | | | | | | | | | | | | | |
Collapse
|
5
|
Gatta C, Castaldo L, Cellerino A, de Girolamo P, Lucini C, D'Angelo L. Brain derived neurotrophic factor in the retina of the teleost N. furzeri. Ann Anat 2014; 196:192-6. [PMID: 24629406 DOI: 10.1016/j.aanat.2014.01.002] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2013] [Revised: 01/02/2014] [Accepted: 01/08/2014] [Indexed: 12/27/2022]
Abstract
BDNF plays an important role in the development and maintenance of visual circuitries in the retina and brain visual centers. In adulthood, BDNF signaling is involved in neural protection and regeneration of retina. In this survey, we investigated the expression of BDNF in the retina of adult Nothobranchius furzeri, a teleost fish employed for age research. After describing the retina of N. furzeri and confirming that the structure is organized in layers as in all vertebrates, we have studied the localization of BDNF mRNA and protein throughout the retinal layers. BDNF mRNA is detectable in all layers, whereas the protein is lacking in the photoreceptors. The occurrence of BDNF provides new insights on its role in the retina, particularly in view of age-related disease of retina.
Collapse
Affiliation(s)
- Claudia Gatta
- Department of Veterinary Medicine and Animal Productions, University of Naples Federico II, via Veterinaria, 1, I-80137 Napoli, Italy
| | - Luciana Castaldo
- Department of Veterinary Medicine and Animal Productions, University of Naples Federico II, via Veterinaria, 1, I-80137 Napoli, Italy
| | - Alessandro Cellerino
- Scuola Normale Superiore of Pisa, Laboratory of Biology, c/o Institute of Biophysics of CNR, via Moruzzi, 1, I-56100 Pisa, Italy
| | - Paolo de Girolamo
- Department of Veterinary Medicine and Animal Productions, University of Naples Federico II, via Veterinaria, 1, I-80137 Napoli, Italy
| | - Carla Lucini
- Department of Veterinary Medicine and Animal Productions, University of Naples Federico II, via Veterinaria, 1, I-80137 Napoli, Italy
| | - Livia D'Angelo
- Department of Veterinary Medicine and Animal Productions, University of Naples Federico II, via Veterinaria, 1, I-80137 Napoli, Italy.
| |
Collapse
|
6
|
Manni L, Rocco ML, Bianchi P, Soligo M, Guaragna M, Barbaro SP, Aloe L. Nerve growth factor: basic studies and possible therapeutic applications. Growth Factors 2013; 31:115-22. [PMID: 23777359 DOI: 10.3109/08977194.2013.804073] [Citation(s) in RCA: 68] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
The nerve growth factor (NGF) belongs to a family of neurotrophic factors called neurotrophins. It was discovered as a molecule that stimulates the survival and maturation of developing neurons in the peripheral nervous system and has later been shown to protect adult neurons in the degenerating mammalian brain. Basic and clinical studies have been undertaken to use NGF as a therapeutic agent aimed at restoring and maintaining neuronal function in the central nervous system and to determine the mechanisms to safely deliver the molecule into the brain. Recent studies have also recognized that the role of NGF extends far beyond the horizon of nerve cells and even beyond the peripheral and central nervous system. Studies published from our laboratory have shown that topical application of NGF possesses a protective action on human pressure ulcer, corneal ulcer and glaucoma. Here, we will review these studies, supporting the therapeutic potential of NGF.
Collapse
Affiliation(s)
- Luigi Manni
- Institute of Translational Pharmacology, National Research Council, Rome, Italy
| | | | | | | | | | | | | |
Collapse
|
7
|
Ola MS, Nawaz MI, El-Asrar AA, Abouammoh M, Alhomida AS. Reduced levels of brain derived neurotrophic factor (BDNF) in the serum of diabetic retinopathy patients and in the retina of diabetic rats. Cell Mol Neurobiol 2013; 33:359-67. [PMID: 23271640 DOI: 10.1007/s10571-012-9901-8] [Citation(s) in RCA: 78] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2012] [Accepted: 12/13/2012] [Indexed: 01/18/2023]
Abstract
Diabetic retinopathy (DR) is widely recognized as a neurovascular disease. Retina, being a neuronal tissue of the eye, produces neurotrophic factors for its maintenance. However, diabetes dysregulates their levels and thereby may damage the retina. Among neurotrophins, brain derived neurotrophic factor (BDNF) is the most abundant in the retina. In this study, we investigated the level of BDNF in the serum of patients with DR and also in the serum and retina of streptozotocin-induced diabetic rats. The level of BDNF was significantly decreased in the serum of proliferative diabetic retinopathy patients as compared to that of non-diabetic healthy controls (25.5 ± 8.5-10.0 ± 8.1 ng/ml, p < 0.001) as well as compared to that of diabetic patients with no retinopathy (21.8 ± 4.7-10.0 ± 8.1 ng/ml, p < 0.001), as measured by ELISA techniques. The levels of BDNF in the serum and retina of diabetic rats were also significantly reduced compared to that of non-diabetic controls (p < 0.05). In addition, the expression level of tropomyosin-related kinase B (TrkB) was significantly decreased in diabetic rat retina compared to that of non-diabetic controls as determined by Western blotting technique. Caspase-3 activity was increased in diabetic rat retina after 3 weeks of diabetes and remained elevated until 10 weeks, which negatively correlated with the level of BDNF (r = -0.544, p = 0.013). Our results indicate that reduced levels of BDNF in diabetes may cause apoptosis and neurodegeneration early in diabetic retina, which may lead to neuro-vascular damage later in DR.
Collapse
Affiliation(s)
- M Shamsul Ola
- Department of Biochemistry, College of Science, King Saud University, Riyadh, 11415, Saudi Arabia.
| | | | | | | | | |
Collapse
|
8
|
The molecular basis of retinal ganglion cell death in glaucoma. Prog Retin Eye Res 2012; 31:152-81. [DOI: 10.1016/j.preteyeres.2011.11.002] [Citation(s) in RCA: 565] [Impact Index Per Article: 47.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2011] [Revised: 10/28/2011] [Accepted: 11/01/2011] [Indexed: 12/14/2022]
|
9
|
Jee D, Lee WK. Inhibitory Effect of Intravitreal Injection of Bevacizumab on Nerve Growth Factor. Curr Eye Res 2011; 37:408-15. [DOI: 10.3109/02713683.2011.632108] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
|
10
|
Overexpression of neurotrophin-3 stimulates a second wave of dopaminergic amacrine cell genesis after birth in the mouse retina. J Neurosci 2011; 31:12663-73. [PMID: 21880927 DOI: 10.1523/jneurosci.1100-11.2011] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Dopaminergic amacrine (DA) cells play multiple and important roles in retinal function. Neurotrophins are known to modulate the number and morphology of DA cells, but the underlying regulatory mechanisms are unclear. Here, we investigate how neurotrophin-3 (NT-3) regulates DA cell density in the mouse retina. We demonstrate that overexpression of NT-3 upregulates DA cell number and leads to a consequent increase in the density of DA cell dendrites. To examine the mechanisms of DA cell density increase, we further investigate the effect of NT-3 overexpression on retinal apoptosis and mitosis during development. We find that NT-3 does not affect the well known wave of retinal cell apoptosis that normally occurs during the first 2 weeks after birth. Instead, overexpression of NT-3 promotes additional mitosis of DA cells at postnatal day 4, but does not affect cell mitosis before birth, the peak period of amacrine cell genesis in wild-type retinas. We next show that retinal explants cultured from birth to day 7 without extra NT-3 produced by lens exhibit similar number of DA cells as in wild type, further supporting the notion that postnatal overexpression of lens-derived NT-3 affects DA cell number. Moreover, the additional mitosis after birth in NT-3-overexpressing mice does not occur in calretinin-positive amacrine cells or PKC-positive rod ON bipolar cells. Thus, the NT-3-triggered wave of cell mitosis after birth is specific for the retinal DA cells.
Collapse
|
11
|
Santos E, Romero-Alemán M, Monzón-Mayor M, Lang D, Rodger J, Yanes C. Expression of BDNF and NT-3 during the ontogeny and regeneration of the lacertidian (Gallotia galloti) visual system. Dev Neurobiol 2011; 71:836-53. [DOI: 10.1002/dneu.20939] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
12
|
Germanà A, Sánchez-Ramos C, Guerrera MC, Calavia MG, Navarro M, Zichichi R, García-Suárez O, Pérez-Piñera P, Vega JA. Expression and cell localization of brain-derived neurotrophic factor and TrkB during zebrafish retinal development. J Anat 2010; 217:214-22. [PMID: 20649707 DOI: 10.1111/j.1469-7580.2010.01268.x] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022] Open
Abstract
Brain-derived neurotrophic factor (BDNF) signaling through TrkB regulates different 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 and TrkB in the retina, the cell types in the retina that express BDNF and TrkB, and the variations in their levels of expression during development, remain poorly defined. The goal of the present study is to determine the age-dependent changes in the levels of expression and localization of BDNF and TrkB in the zebrafish retina. Zebrafish retinas from 10 days post-fertilization (dpf) to 180 dpf were used to perform PCR, Western blot and immunohistochemistry. Both BDNF and TrkB mRNAs, and BDNF and full-length TrkB proteins were detected at all ages sampled. The localization of these proteins in the retina was very similar at all time points studied. BDNF immunoreactivity was found in the outer nuclear layer, the outer plexiform layer and the inner plexiform layer, whereas TrkB immunoreactivity was observed in the inner plexiform layer and, to a lesser extent, in the ganglion cell layer. These results demonstrate that the pattern of expression of BDNF and TrkB in the retina of zebrafish remains unchanged during postembryonic development and adult life. Because TrkB expression in retina did not change with age, cells expressing TrkB may potentially be able to respond during the entire lifespan of zebrafish to BDNF either exogenously administered or endogenously produced, acting through paracrine mechanisms.
Collapse
Affiliation(s)
- A Germanà
- Dipartmento di Morfologia, Biochimica, Fisiologia e Produzione Animale, Sezione di Morfologia, Università di Messina, Italy
| | | | | | | | | | | | | | | | | |
Collapse
|
13
|
Butowt R, von Bartheld CS. Fates of neurotrophins after retrograde axonal transport: phosphorylation of p75NTR is a sorting signal for delayed degradation. J Neurosci 2009; 29:10715-29. [PMID: 19710323 PMCID: PMC2761711 DOI: 10.1523/jneurosci.2512-09.2009] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2009] [Revised: 07/15/2009] [Accepted: 07/20/2009] [Indexed: 12/16/2022] Open
Abstract
Neurotrophins can mediate survival or death of neurons. Opposing functions of neurotrophins are based on binding of these ligands to two distinct types of receptors: trk receptors and p75NTR. Previous work showed that target-derived NGF induces cell death, whereas BDNF and NT-3 enhance survival of neurons in the isthmo-optic nucleus of avian embryos. To determine the fate of retrogradely transported neurotrophins and test whether their sorting differs between neurotrophins mediating survival- or death-signaling pathways, we traced receptor-binding, sorting, and degradation kinetics of target-applied radiolabeled neurotrophins that bind in this system to trk receptors (BDNF, NT-3) or only to p75NTR (NGF). At the ultrastructural level, the p75NTR-bound NGF accumulates with a significant delay in multivesicular bodies and organelles of the degradation pathway on arrival in the cell body when compared with trk-bound BDNF or NT-3. This delayed lysosomal accumulation was restricted to target-derived NGF, but was not seen when NGF was supplied to the soma in vitro. The kinase inhibitors K252a and Gö6976 alter the kinetics of organelle accumulation: phosphorylation of p75NTR is a sorting signal for delayed sequestering of p75NTR-bound NGF in multivesicular bodies and delayed degradation in lysosomes when compared with trk-bound neurotrophins. Mutagenesis and mass spectrometry studies indicate that p75NTR is phosphorylated by conventional protein kinase C on serine 266. We conclude that, in addition to the known phosphorylation of trks, the phosphorylation of p75NTR can also significantly affect neuronal survival in vivo by changing the intracellular sorting and degradation kinetics of its ligands and thus signaling duration.
Collapse
Affiliation(s)
- Rafal Butowt
- Department of Physiology and Cell Biology, University of Nevada School of Medicine, Reno, Nevada 89557
| | | |
Collapse
|
14
|
Liu X, Robinson ML, Schreiber AM, Wu V, Lavail MM, Cang J, Copenhagen DR. Regulation of neonatal development of retinal ganglion cell dendrites by neurotrophin-3 overexpression. J Comp Neurol 2009; 514:449-58. [PMID: 19350645 DOI: 10.1002/cne.22016] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
The morphology of dendrites constrains and reflects the nature of synaptic inputs to neurons. The visual system has served as a useful model to show how visual function is determined by the arborization patterns of neuronal processes. In retina, light ON and light OFF responding ganglion cells selectively elaborate their dendritic arbors in distinct sublamina, where they receive, respectively, inputs from ON and OFF bipolar cells. During neonatal maturation, the bilaminarly distributed dendritic arbors of ON-OFF retinal ganglion cells (RGCs) are refined to more narrowly localized monolaminar structures characteristic of ON or OFF RGCs. Recently, brain-derived neurotrophic factor (BDNF) has been shown to regulate this laminar refinement, and to enhance the development of dendritic branches selectively of ON RGCs. Although other related neurotrophins are known to regulate neuronal process formation in the central nervous system, little is known about their action in maturing retina. Here, we report that overexpression of neurotrophin-3 (NT-3) in the eye accelerates RGC laminar refinement before eye opening. Furthermore, NT-3 overexpression increases dendritic branch number but reduces dendritic elongation preferentially in ON-OFF RGCs, a process that also occurs before eye opening. NT-3 overexpression does affect dendritic maturation in ON RGCs, but to a much less degree. Taken together, our results suggest that NT-3 and BDNF exhibit overlapping effects in laminar refinement but distinct RGC-cell-type specific effects in shaping dendritic arborization during postnatal development.
Collapse
Affiliation(s)
- Xiaorong Liu
- Department of Neurobiology and Physiology, Northwestern University, Evanston, Illinois 60208, USA.
| | | | | | | | | | | | | |
Collapse
|
15
|
Sposato V, Bucci MG, Coassin M, Russo MA, Lambiase A, Aloe L. Reduced NGF level and TrkA protein and TrkA gene expression in the optic nerve of rats with experimentally induced glaucoma. Neurosci Lett 2008; 446:20-4. [PMID: 18817846 DOI: 10.1016/j.neulet.2008.09.024] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2008] [Revised: 09/04/2008] [Accepted: 09/07/2008] [Indexed: 11/16/2022]
Abstract
Glaucoma (GL) is an optic neuropathy characterized by progressive loss of visual field due to retinal cell death and optic nerve (ON) degeneration, usually in response to abnormal elevated intraocular pressure (EIOP). It has previously demonstrated that cells of the ON express nerve growth factor (NGF) and NGF-receptors. Relatively little is known, however, about their role on the ON of the glaucomatous eye. The aim of the study was to elucidate this aspect. Using a rat model of GL we investigated the response of NGF and NGF-receptors in the ON of subjects with experimentally induced EIOP. Our results show that EIOP significantly impairs the presence of NGF and NGF-receptor proteins and TrkA gene expression in the ON of glaucomatous eye. These findings suggest that NGF and NGF-receptor might be important signals for the ON response in the EIOP.
Collapse
Affiliation(s)
- Valentina Sposato
- Institute of Neurobiology and Molecular Medicine, Section of Neurobiology, National Research Council (CNR), Via del Fosso di Fiorano 64/65, I-00143 Rome, Italy
| | | | | | | | | | | |
Collapse
|
16
|
Vissio PG, Cánepa MM, Maggese MC. Brain-derived neurotrophic factor (BDNF)-like immunoreactivity localization in the retina and brain of Cichlasoma dimerus (Teleostei, Perciformes). Tissue Cell 2008; 40:261-70. [PMID: 18343472 DOI: 10.1016/j.tice.2008.01.001] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2007] [Revised: 01/11/2008] [Accepted: 01/14/2008] [Indexed: 12/21/2022]
Abstract
Brain-derived neurotrophic factor (BDNF) is a neurotrophin involved in the development and maintenance of vertebrate nervous systems. Although there were several studies in classical animal models, scarce information for fish was available. The main purpose of this study was to analyze the distribution of BDNF in the brain and retina of the cichlid fish Cichlasoma dimerus. By immunohistochemistry we detected BDNF-like immunoreactive cells in the cytoplasm and the nuclei of the ganglion cell layer and the inner nuclear layer of the retina. In the optic tectum, BDNF-like immunoreactivity was detected in the nucleus of neurons of the stratum periventriculare and the stratum marginale and in neurons of the intermediate layers. In the hypothalamus we found BDNF-like immunoreactivity mainly in the cytoplasm of the nucleus lateralis tuberis and the nucleus of the lateral recess. To confirm the nuclear and cytoplasm localization of BDNF we performed subcellular fractionation, followed by Western blot, detecting a 39 kDa immunoreactive-band corresponding to a possible precursor form of BDNF in both fractions. BDNF-like immunoreactivity was distributed in areas related with photoreception (retina), the integration center of retinal projections (optic tectum) and the control center of background and stress adaptation (hypothalamus). These results provide baseline anatomical information for future research about the role of neurotrophins in the adult fish central nervous system.
Collapse
Affiliation(s)
- P G Vissio
- CONICET, Laboratorio de Embriología Animal, DBBE, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Ciudad Universitaria, C1428EHA Buenos Aires, Argentina.
| | | | | |
Collapse
|
17
|
Sun X, Xu X, Wang F, Zhang X, Ho PC, Liu H, Qian J, Yu Z, Lu H, Xu W. Nerve Growth Factor Helps Protect Retina in Experimental Retinal Detachment. Ophthalmologica 2007; 222:58-61. [DOI: 10.1159/000109281] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
|
18
|
Sun X, Xu X, Wang F, Zhang X, Yu Z, Lu H, Ho PC. Effects of nerve growth factor for retinal cell survival in experimental retinal detachment. Curr Eye Res 2007; 32:765-72. [PMID: 17882709 DOI: 10.1080/02713680701531082] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Abstract
PURPOSE To investigate the neuron protective effect of recombined nerve growth factor (rNGF) on retinal cell damage induced by experimental retinal detachment. METHODS Experimental retinal detachment models were created in Sprague-Dawley rats by subretinal injection of sodium hyaluronate. Intravitreal injection of rNGF (5 microg/eye) or phosphate-buffered saline (PBS) was separately applied every 4 days after retinal detachment. The rat eyes were then observed and sacrificed at various time points. Morphologic changes were observed by light microscopy, electron microscopy, and cell counts. Apoptosis of retinal cells was detected by TUNEL assay. RESULTS After retinal detachment, most eyes from NGF-treated groups showed better organized structure of retinal cells than those from the PBS-treated control groups. Cell counts indicated that the nuclei numbers in the outer nuclear layer (ONL), inner nuclear layer (INL), and ganglion cell layer (GCL) of NGF-treated groups were significantly more than those from PBS-treated control group (p < 0.05) after retinal reattachment. TUNEL-positive cells were identified in ONL, INL, and GCL. They peaked at the fourth day after retinal detachment in both the NGF-treated groups and the control groups. But the cell counts of apoptosis revealed that the NGF-treated retina had less TUNEL-positive cells than the control groups (p < 0.05). CONCLUSIONS The results showed that intravitreal injection of exogenous NGF can protect retinal cells from degeneration and apoptosis in experimental retinal detachment. It may exert its neuroprotection effect by preventing the apoptosis of retinal cells after retinal detachment.
Collapse
Affiliation(s)
- Xiaodong Sun
- Eye Research Institute of Shanghai JiaoTong University, 85 Wu Jin Road, Shanghai 200080, China
| | | | | | | | | | | | | |
Collapse
|
19
|
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: 70] [Impact Index Per Article: 4.1] [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
|
20
|
Calamusa M, Pattabiraman PP, Pozdeyev N, Iuvone PM, Cellerino A, Domenici L. Specific alterations of tyrosine hydroxylase immunopositive cells in the retina of NT-4 knock out mice. Vision Res 2007; 47:1523-36. [PMID: 17350071 DOI: 10.1016/j.visres.2007.01.017] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2006] [Revised: 01/26/2007] [Accepted: 01/26/2007] [Indexed: 11/23/2022]
Abstract
To assess the effect of NT-4 deprivation on maturation of retinal circuitry, we investigated a mouse with targeted deletion of the gene encoding nt-4 (nt-4(-/-)). In particular, we studied neurons immunostained by an antibody recognizing tyrosine hydroxylase (TH), the rate limiting enzyme for dopamine (DA) synthesis. We found that TH immunopositive processes were altered in the retina of nt-4(-/-). Alteration of TH immunopositive processes in nt-4(-/-) mice resulted in changes of DA turnover, as assessed by high-pressure liquid chromatography measurements. These findings suggest that retinal NT-4 plays a role in the morphological maturation of dopaminergic retinal cells.
Collapse
Affiliation(s)
- Martina Calamusa
- Istituto di Neuroscienze, Consiglio Nazionale delle Ricerche (CNR), Via G. Moruzzi 1, 56100 Pisa, Italy
| | | | | | | | | | | |
Collapse
|
21
|
Fischer AJ, Stanke JJ, Aloisio G, Hoy H, Stell WK. Heterogeneity of horizontal cells in the chicken retina. J Comp Neurol 2007; 500:1154-71. [PMID: 17183536 DOI: 10.1002/cne.21236] [Citation(s) in RCA: 72] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Despite numerous reports that different markers are expressed by horizontal cells in the avian retina, it remains unknown whether different types of horizontal cells can be defined by differences in their immunocytochemical profiles. The purpose of this study was to rectify this deficiency. We identified horizontal cells by indirect immunofluorescence with antibodies to calretinin, trkA, GABA, Prox1, AP2alpha, Pax6, islet1, and Lim1 + 2. We found two major groups of horizontal cells, those that express trkA and those that express calretinin. The trkA-immunoreactive (-IR) horizontal cells had small, round somata and robust, bulbous dendritic endings, whereas calretinin-IR horizontal cells had large, polygonal cell bodies and fine, diffuse dendritic endings, both contacting the calbindin-IR pedicles of double cones. Weak gamma-aminobutyric acid (GABA) immunoreactivity was observed only in a few of the trkA-IR horizontal cells, whereas the overlap of calretinin and GABA immunoreactivities was 100%. The majority of trkA-IR horizontal cells expressed islet1, and the majority of calretinin-IR horizontal cells expressed Lim1 + 2, AP2alpha, and Pax6. Islet1 immunoreactivity was observed in a small fraction of calretinin-IR/non-trkA-IR cells. In agreement with previous reports, we detected Prox1 immunoreactivity in all types of horizontal cells. These immunolabeling profiles suggest that there are four immunochemically distinct subtypes of horizontal cells in the postnatal chick retina, which may match the four types that have been observed in Golgi-impregnated pigeon and turtle retinas.
Collapse
Affiliation(s)
- Andy J Fischer
- Department of Neuroscience, The Ohio State University, Columbus, Ohio 43210, USA.
| | | | | | | | | |
Collapse
|
22
|
Santos E, Monzón-Mayor M, Romero-Alemán M, Yanes C. Distribution of neurotrophin-3 during the ontogeny and regeneration of the lizard (Gallotia galloti) visual system. Dev Neurobiol 2007; 68:31-44. [DOI: 10.1002/dneu.20566] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
|
23
|
von Bartheld CS, Fritzsch B. Comparative analysis of neurotrophin receptors and ligands in vertebrate neurons: tools for evolutionary stability or changes in neural circuits? BRAIN, BEHAVIOR AND EVOLUTION 2006; 68:157-72. [PMID: 16912469 DOI: 10.1159/000094085] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
To better understand the role of multiple neurotrophin ligands and their receptors in vertebrate brain evolution, we examined the distribution of trk neurotrophin receptors in representatives of several vertebrate classes. Trk receptors are largely expressed in homologous neuronal populations among different species/classes of vertebrates. In many neurons, trkB and trkC receptors are co-expressed. TrkB and trkC receptors are primarily found in neurons with more restricted, specialized dendritic and axonal fields that are thought to be involved in discriminative or 'analytical' functions. The neurotrophin receptor trkA is expressed predominantly in neurons with larger, overlapping dendritic fields with more heterogeneous connections ('integrative' or 'modulatory' systems) such as nociceptive and sympathetic autonomic nervous system, locus coeruleus and cholinergic basal forebrain. Surveys of trk receptor expression and function in the peripheral nervous system of different vertebrate classes reveal trends ranging from dependency on a single neurotrophin to a more complex dependency on increasing numbers of neurotrophins and their receptors, for example, in taste and inner ear innervation. Gene deletion studies in mice provide evidence for a complex regulation of neuronal survival of sensory ganglion cells by different neurotrophins. Although expression of neurotrophins and their receptors is predominantly conserved in most circuits, increasing diversity of neurotrophin ligands and their receptors and a more complex dependency of neurons on neurotrophins might have facilitated the formation of at least some new neuronal entities.
Collapse
|
24
|
Fischer AJ, Skorupa D, Schonberg DL, Walton NA. Characterization of glucagon-expressing neurons in the chicken retina. J Comp Neurol 2006; 496:479-94. [PMID: 16572462 PMCID: PMC2565864 DOI: 10.1002/cne.20937] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
We recently identified large glucagon-expressing neurons that densely ramify neurites in the peripheral edge of the retina and regulate the proliferation of progenitors in the circumferential marginal zone (CMZ) of the postnatal chicken eye (Fischer et al. [2005] J Neurosci 25:10157-10166). However, nothing is known about the transmitters and proteins that are expressed by the glucagon-expressing neurons in the avian retina. We used antibodies to cell-distinguishing markers to better characterize the different types of glucagon-expressing neurons. We found that the large glucagon-expressing neurons were immunoreactive for substance P, neurofilament, Pax6, AP2alpha, HuD, calretinin, trkB, and trkC. Colocalization of glucagon and substance P in the large glucagon-expressing neurons indicates that these cells are the "bullwhip cells" that have been briefly described by Ehrlich et al. ([1987] J Comp Neurol 266:220-233). Similar to the bullwhip cells, the conventional glucagon-expressing amacrine cells were immunoreactive for calretinin, HuD, Pax6, and AP2alpha. Unlike bullwhip cells, the conventional glucagon-expressing amacrine cells were immunoreactive for GABA. While glucagon-immunoreactive amacrine cells were negative for substance P in central regions of the retina, a subset of this type of amacrine cell was immunoreactive for substance P in far peripheral regions of the retina. An additional type of glucagon/substance P-expressing neuron, resembling the bullwhip cells, was found in far peripheral and dorsal regions of the retina. Based on morphology, distribution within the retina, and histological markers, we conclude that there may be four different types of glucagon-expressing neurons in the avian retina.
Collapse
Affiliation(s)
- Andy J Fischer
- Department of Neuroscience, Ohio State University, Columbus, 43210, USA.
| | | | | | | |
Collapse
|
25
|
Abstract
Progress in the study of the molecular mechanisms that regulate neuronal differentiation has been quite impressive in recent years, and promises to continue to an equally fast pace. This should not lead us into a sense of complacency, however, because there are still significant barriers that cannot be overcome by simply conducting the same type of experiments that we have been performing thus far. This article will describe some of these challenges, while highlighting the conceptual and methodological breakthroughs that will be necessary to overcome them.
Collapse
Affiliation(s)
- Ruben Adler
- Department of Ophthalmology and Neuroscience The Johns Hopkins University School of Medicine Baltimore, MD 21287-9257, USA.
| |
Collapse
|
26
|
Steffensky M, Steinbach K, Schwarz U, Schlosshauer B. Differential impact of semaphorin 3E and 3A on CNS axons. Int J Dev Neurosci 2005; 24:65-72. [PMID: 16338115 DOI: 10.1016/j.ijdevneu.2005.10.007] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2005] [Revised: 10/24/2005] [Accepted: 10/26/2005] [Indexed: 11/20/2022] Open
Abstract
During the development of the central nervous system (CNS), the correct wiring of outgrowing neurites is mediated by antagonistic mechanisms. Aberrant growth is prevented by repulsive factors such as semaphorins. Expression of the ligands Sema3A and -3E and the receptors neuropilin Npn-1, -2a and -2b in the chick visual system were analyzed by RT-PCR. Whereas Sema3A and its major receptor Npn-1 were abundant, Sema3E and Npn-2 isoform expression was highly restricted and developmentally regulated. Peak expression occurred during retinal axon innervation of the tectum. Functional in vitro assays with recombinant proteins revealed a topography-specific growth cone collapsing activity of Sema3A for tectal axons. Interestingly, whereas tectal axons collapsed in a topographic-specific manner only in the presence of Sema3A, retinal axons responded only to Sema3E. The collapsing activity was intracellularly mediated by cGMP. For a detailed analysis of neuronal responses to sempahorins, time lapse video recording was performed. When tectal and retinal axons were pre-exposed to brain-derived neurotrophic factor (BDNF), a protective effect was evident only in the case of retinal axons. Our results suggest a molecular mechanism whereby ingrowth of retinal axons into the tectum can be regulated by Sema3E/BDNF modulation without disturbing tectal axon growth out of the tectum mediated by Sema3A.
Collapse
Affiliation(s)
- Marion Steffensky
- NMI Naturwissenschaftliches und Medizinisches Institut, an der Universität Tübingen, Markwiesenstr. 55, D-72770 Reutlingen, Germany
| | | | | | | |
Collapse
|
27
|
Butowt R, von Bartheld CS. Anterograde axonal transport of BDNF and NT-3 by retinal ganglion cells: roles of neurotrophin receptors. Mol Cell Neurosci 2005; 29:11-25. [PMID: 15866043 DOI: 10.1016/j.mcn.2005.02.004] [Citation(s) in RCA: 36] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2004] [Revised: 12/22/2004] [Accepted: 02/04/2005] [Indexed: 11/15/2022] Open
Abstract
Retinal ganglion cells (RGCs) transport exogenous neurotrophins anterogradely to the midbrain tectum/superior colliculus with significant downstream effects. We determined contributions of neurotrophin receptors for anterograde transport of intraocularly injected radiolabeled neurotrophins. In adult rodents, anterograde transport of brain-derived neurotrophic factor (BDNF) was receptor-mediated, and transport of exogenous BDNF and neurotrophin-3 (NT-3) was more efficient, per RGC, in rodents than chicks. RT-PCR and Western blot analysis of purified murine RGCs showed that adult RGCs express the p75 receptor. Anterograde transport of BDNF or NT-3 was not diminished in p75 knock-out mice (with unaltered final numbers of RGCs), but BDNF transport was substantially reduced by co-injected trkB antibodies. In chick embryos, however, p75 antisense or co-injected p75 antibodies significantly attenuated anterograde transport of NT-3 by RGCs. Thus, neither BDNF nor NT-3 utilizes p75 for anterograde transport in adult rodent RGCs, while anterograde NT-3 transport requires the p75 receptor in embryonic chicken RGCs.
Collapse
Affiliation(s)
- Rafal Butowt
- Department of Physiology and Cell Biology, Mailstop 352, University of Nevada School of Medicine, Reno, NV 89557, USA
| | | |
Collapse
|
28
|
Gehler S, Gallo G, Veien E, Letourneau PC. p75 neurotrophin receptor signaling regulates growth cone filopodial dynamics through modulating RhoA activity. J Neurosci 2004; 24:4363-72. [PMID: 15128850 PMCID: PMC6729430 DOI: 10.1523/jneurosci.0404-04.2004] [Citation(s) in RCA: 101] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
The mechanisms by which neurotrophins regulate growth cone motility are unclear. We investigated the role of the p75 neurotrophin receptor (p75NTR) in mediating neurotrophin-induced increases in filopodial length. Our data demonstrate that neurotrophin binding to p75NTR is necessary and sufficient to regulate filopodial dynamics. Furthermore, retinal and dorsal root ganglion growth cones from p75 mutant mice are insensitive to neurotrophins but display enhanced filopodial lengths comparable with neurotrophin-treated wild-type growth cones. This suggests unoccupied p75NTR negatively regulates filopodia length. Furthermore, p75NTR regulates RhoA activity to mediate filopodial dynamics. Constitutively active RhoA blocks neurotrophin-induced increases in filopodial length, whereas inhibition of RhoA enhances filopodial lengths, similar to neurotrophin treatment. BDNF treatment of retinal neurons results in reduced RhoA activity. Furthermore, p75 mutant neurons display reduced levels of activated RhoA compared with wild-type counterparts, consistent with the enhanced filopodial lengths observed on mutant growth cones. These observations suggest that neurotrophins regulate filopodial dynamics by depressing the activation of RhoA that occurs through p75NTR signaling.
Collapse
Affiliation(s)
- Scott Gehler
- Department of Neuroscience, University of Minnesota, Minneapolis, Minnesota 55455, USA
| | | | | | | |
Collapse
|
29
|
Lindqvist N, Näpänkangas U, Lindblom J, Hallböök F. Proopiomelanocortin and melanocortin receptors in the adult rat retino-tectal system and their regulation after optic nerve transection. Eur J Pharmacol 2004; 482:85-94. [PMID: 14660008 DOI: 10.1016/j.ejphar.2003.10.011] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
The aim of this study was to characterise the expression of the melanocortin system in the normal and injured rat visual system. Using real-time polymerase chain reaction and immunohistochemistry, we detected melanocortin MC(3), MC(4) and MC(5) receptors and proopiomelanocortin in adult retina and superior colliculus. Melanocortin MC(4) receptor mRNA was the most abundant receptor. Melanocortin MC(3), MC(4) and MC(5) receptors were localised to the ganglion cell and inner nuclear layers and the melanocortin MC(3) and MC(4) receptors were localised to retinal ganglion cells. Transection of the optic nerve leads to ganglion cell death and both melanocortin receptor and proopiomelanocortin expression decreased in superior colliculus after transection whereas the expression was unchanged or even increased in the retina. alpha-Melanocyte-stimulating hormone elicited neurite outgrowth from embryonic retinal explants. Together, these data implicate a role for the melanocortin system in the adult rat retina and that melanocortins can stimulate neurite growth from retinal neurons.
Collapse
MESH Headings
- Animals
- Chick Embryo
- Female
- Optic Nerve Injuries/genetics
- Optic Nerve Injuries/metabolism
- Pro-Opiomelanocortin/biosynthesis
- Pro-Opiomelanocortin/genetics
- RNA, Messenger/biosynthesis
- RNA, Messenger/genetics
- Rats
- Receptor, Melanocortin, Type 3/biosynthesis
- Receptor, Melanocortin, Type 3/genetics
- Receptor, Melanocortin, Type 4/biosynthesis
- Receptor, Melanocortin, Type 4/genetics
- Receptors, Corticotropin/genetics
- Receptors, Corticotropin/metabolism
- Receptors, Melanocortin/biosynthesis
- Receptors, Melanocortin/genetics
- Retina/metabolism
- Superior Colliculi/metabolism
Collapse
Affiliation(s)
- Niclas Lindqvist
- Unit for Developmental Neuroscience, Department of Neuroscience, Biomedical Center, Uppsala University, Box 587, S-75123 Uppsala, Sweden
| | | | | | | |
Collapse
|
30
|
Srinivasan B, Roque CH, Hempstead BL, Al-Ubaidi MR, Roque RS. Microglia-derived pronerve growth factor promotes photoreceptor cell death via p75 neurotrophin receptor. J Biol Chem 2004; 279:41839-45. [PMID: 15277529 DOI: 10.1074/jbc.m402872200] [Citation(s) in RCA: 71] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Reports implicating microglia-derived nerve growth factor (NGF) during programmed cell death in the developing chick retina led us to investigate its possible role in degenerative retinal disease. Freshly isolated activated retinal microglia expressed high molecular weight forms of neurotrophins including that of nerve growth factor (NGF), brain-derived neurotrophic factor, neurotrophin-3, and neurotrophin-4. Conditioned media from cultured retinal microglia (MGCM) consistently yielded a approximately 32-kDa NGF-reactive band when supplemented with bovine serum albumin (BSA) or protease inhibitors (PI); and promoted cell death that was suppressed by NGF immunodepletion in a mouse photoreceptor cell line (661w). The approximately 32 kDa protein was partially purified (MGCM/p32) and was highly immunoreactive with a polyclonal anti-pro-NGF antibody. Both MGCM/p32 and recombinant pro-NGF protein promoted cell death in 661w cultures. Increased levels of pro-NGF mRNA and protein were observed in the RCS rat model of retinal dystrophy. MGCM-mediated cell death was reversed by p75NTR antiserum in p75NTR(+)/trkA(-) 661w cells. Our study shows that a approximately 32 kDa pro-NGF protein released by activated retinal microglia promoted degeneration of cultured photoreceptor cells. Moreover, our study suggests that defective post-translational processing of NGF might be involved in photoreceptor cell loss in retinal dystrophy.
Collapse
Affiliation(s)
- Bhooma Srinivasan
- Department of Cell Biology and Genetics, University of North Texas Health Science Center, Fort Worth, Texas 76107, USA
| | | | | | | | | |
Collapse
|
31
|
Chytrova G, Johnson JE. Spontaneous retinal activity modulates BDNF trafficking in the developing chick visual system. Mol Cell Neurosci 2004; 25:549-57. [PMID: 15080885 DOI: 10.1016/j.mcn.2003.11.009] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2003] [Revised: 11/06/2003] [Accepted: 11/19/2003] [Indexed: 11/18/2022] Open
Abstract
Both neuronal activity and neurotrophin signaling play critical roles in normal CNS development. This study examined whether spontaneous retinal activity (SRA) also governs the axonal transport of endogenous brain-derived neurotrophic factor (BDNF) protein within the developing chick visual system. In previous work, we have found that during the normal period of SRA, retinal BDNF protein levels decrease by about 50% while BDNF mRNA levels remain elevated. Here, we show that the blockade of SRA with tetrodotoxin (TTX), or the blockade of axonal transport with colchicine, both reversed the normal mismatch between retinal BDNF mRNA and protein. The axonal transport of retinal-derived BDNF in segments of the optic nerve as well as tectal-derived BDNF protein transported in segments of the optic tract were both significantly reduced after very brief periods of activity blockade. These results suggest that normal SRA plays a role in regulating the axonal transport of endogenous BDNF protein.
Collapse
Affiliation(s)
- Gabriela Chytrova
- Neuroscience Program, Wake Forest University School of Medicine, Winston-Salem, NC 27157, USA
| | | |
Collapse
|
32
|
Bonnet D, Garcia M, Vecino E, Lorentz JG, Sahel J, Hicks D. Brain-derived neurotrophic factor signalling in adult pig retinal ganglion cell neurite regeneration in vitro. Brain Res 2004; 1007:142-51. [PMID: 15064145 DOI: 10.1016/j.brainres.2004.02.023] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/11/2004] [Indexed: 11/26/2022]
Abstract
Brain-derived neurotrophic factor (BDNF) has been implicated in stimulating retinal ganglion cell (RGC) survival and axonal regeneration in rodent animal models in vivo and in vitro, but very little data are available on neurotrophin effects in higher mammals. We hence analysed BDNF signalling in primary cultures of adult pig RGC. As detected by immunohistochemistry, HPLC analysis and RT-PCR, BDNF protein and mRNA were present within pig retina in vivo and in vitro, where it may be involved in baseline RGC neuritogenesis. Initial dose-response studies established optimal effects were induced by 20 ng/ml BDNF, leading to an approximately threefold increase in neurite length. We analysed the respective contributions of phosphatidyl inositol 3 kinase (PI3K) and mitogen activated protein kinase (MAPK) cascades to BDNF-induced neurite regeneration. Addition of either the PI3K inhibitor wortmannin or the MAPK inhibitor U0126 blocked 50-100% BDNF-induced neurite elongation; U0126 also significantly reduced neurite regeneration below untreated control levels. The trk receptor inhibitor K252a had no observable effect on neurite regeneration or morphology. These data hence demonstrate that BDNF is a potent stimulator of neurite growth in RGC prepared from an adult large mammal retina, and that at least two signalling pathways are causally involved. BDNF-based therapy may be of potential use in treating RGC degeneration in humans.
Collapse
Affiliation(s)
- Delphine Bonnet
- Laboratoire de Physiopathologie Cellulaire et Moléculaire de la Rétine, INSERM U. 592, Clinique Médicale A, Centre Hospitalier Universitaire, BP 426, 67091 Strasbourg, France
| | | | | | | | | | | |
Collapse
|
33
|
TrkB receptor signaling regulates developmental death dynamics, but not final number, of retinal ganglion cells. J Neurosci 2003. [PMID: 14602830 DOI: 10.1523/jneurosci.23-31-10137.2003] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
We investigated the effects of endogenous neurotrophin signaling on the death-survival of immature retinal ganglion cells (RGCs) in vivo. Null mutation of brain-derived neurotrophic factor [(BDNF) alone or in combination with neurotrophin 4 (NT4)] increases the peak rate of developmental RGC death as compared with normal. Null mutation of NT4 alone is ineffective. Null mutation of the full-length trkB (trkBFL) receptor catalytic domain produces a dose-dependent increase in the peak RGC death rate that is negatively correlated with retinal levels of trkBFL protein and phosphorylated (activated) trkBFL. Depletion of target-derived trkB ligands by injection of trkB-Fc fusion protein into the superior colliculus increases the peak rate of RGC death compared with trkA-Fc-treated and normal animals. Adult trkBFL+/- mice have a normal number of RGCs, despite an elevated peak death rate of immature RGCs. Thus, target-derived BDNF modulates the dynamics of developmental RGC death through trkBFL activation, but BDNF/trkB-independent mechanisms determine the final number of RGCs.
Collapse
|
34
|
Duprey-Díaz MV, Blagburn JM, Blanco RE. Neurotrophin-3 and TrkC in the frog visual system: changes after axotomy. Brain Res 2003; 982:54-63. [PMID: 12915240 DOI: 10.1016/s0006-8993(03)02948-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Neurotrophins are potent regulators of the survival of different neuronal populations in the CNS. Little is known of the immunodistribution of neurotrophin-3 (NT-3) and tyrosine kinase C (TrkC) receptor in the frog visual system, which can successfully regenerate and recover vision after injury. In this study we show that both NT-3 and TrkC are present in the frog retina and tectum, and that their distribution changes after optic nerve transection. Both NT-3 and TrkC are present in the ganglion cell layer, inner nuclear layer, nerve fiber layer and outer plexiform layer, and in Müller cells of control retinas. Quantification of identified RGCs shows that there are only small changes in the proportion, or intensity, of NT-3 immunostained cells surviving after axotomy and regeneration. Müller cell staining, however, is increased. TrkC staining in the retina does not change after axotomy. In the tectum, NT-3 immunoreactivity is present in the retinorecipient layer 9, and in radial processes of neurons and ependymoglia. TrkC is present in ependymoglia and in tectal neurons. After axotomy or colchicine treatment fewer NT-3-immunoreactive processes are present in layer 9 and there is decreased staining of tectal neurons. These data are consistent with the hypothesis that NT-3 is synthesized in the retina and anterogradely transported to the tectum. TrkC immunostaining, on the other hand, increases in tectal cells after optic nerve transection, suggesting that it may be regulated by the supply of NT-3 from the retina.
Collapse
Affiliation(s)
- Mildred V Duprey-Díaz
- Institute of Neurobiology, University of Puerto Rico Medical Sciences Campus, 201 Boulevard del Valle, San Juan 00901, Puerto Rico
| | | | | |
Collapse
|
35
|
Ghazi-Nouri SMS, Assi A, Limb GA, Scott RAH, von Bussmann K, Humphrey I, Luthert PJ, Charteris DG. Laser photocoagulation alters the pattern of staining for neurotrophin-4, GFAP, and CD68 in human retina. Br J Ophthalmol 2003; 87:488-92. [PMID: 12642316 PMCID: PMC1771631 DOI: 10.1136/bjo.87.4.488] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
AIMS To investigate the staining pattern of neurotrophin-3 (NT3), neurotrophin-4 (NT4), and brain derived neurotrophic factor (BDNF) as well as glial fibrillary acid protein (GFAP) and CD68 in lasered human retina. METHODS Retinal laser photocoagulation was performed on four patients (two males, two females) with choroidal malignant melanoma 1-6 days before enucleation. Three other enucleated eyes with malignant melanoma and three normal cadaveric donor eyes were used as controls. Immunohistochemistry was performed to investigate the pattern of staining of NT3, NT4, BDNF, GFAP, and CD68 in 7 mm sections of fixed specimens. RESULTS Expression of NT4 was detected in the inner and outer nuclear layers of all the retinal sections examined but no NT3 and BDNF staining was seen. NT4 staining was found to be less intense in lasered and melanoma controls compared to normal cadaveric donor retinas. There was an upregulation of GFAP expression in both lasered and control eyes with melanoma in comparison with normal controls. CD68 staining was only observed in retinal pigment epithelium and choroid of lasered eyes. CONCLUSION NT4 is expressed in inner and outer nuclear layers of normal human retina and its expression is downregulated following laser photocoagulation. This occurs in parallel with an increased expression of GFAP suggesting that reactive changes in Muller cells may be responsible for reduced NT4 staining. Expression of CD68 at the site of laser injury is consistent with a wound healing process as a response to local damage.
Collapse
|
36
|
Wang X, Butowt R, von Bartheld CS. Presynaptic neurotrophin-3 increases the number of tectal synapses, vesicle density, and number of docked vesicles in chick embryos. J Comp Neurol 2003; 458:62-77. [PMID: 12577323 DOI: 10.1002/cne.10558] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
To determine whether presynaptically derived neurotrophins may contribute to synaptic plasticity, we examined whether neurotrophin-3 (NT-3) changed the number, size, vesicle content, or vesicle distribution of synapses within the retinorecipient layers of the chick optic tectum. In this system, endogenous NT-3 derives presynaptically from retinal ganglion cell axons. Retinotectal synapses comprise the majority of synapses in superficial tectal layers, as demonstrated by destruction of retinotectal input by intraocular application of the drug monensin. To examine the effect of increased or decreased levels of NT-3, either exogenous NT-3 or monoclonal NT-3 blocking antibodies were injected into the optic tectum of 19-day-old chick embryos, spiked with radiolabeled protein to verify the success of injections and estimate effective concentrations. After 48 hours, the ultrastructure of superficial tectal layers was analyzed and compared with samples from control tecta injected with cytochrome C. NT-3 increased the number of synapses, synaptic vesicles/profile, synaptic vesicle densities, the number of docked vesicles, and the length of the synaptic profile. Deprivation of anterogradely transported endogenous NT-3 with NT-3 antibodies resulted in the opposite effect: decreased numbers of synapses, decreased vesicle densities, and decreased numbers of docked vesicles. Brain-derived neurotrophic factor (BDNF) had a largely different effect than NT-3. BDNF increased the density of vesicles and deprivation of endogenous TrkB ligands with TrkB fusion protein reduced the density of vesicles in the synapses, without effects on synapse number or docked vesicles. We conclude that anterogradely transported NT-3 affects synapse strength in a way that differs from that of presumably postsynaptic-derived BDNF.
Collapse
Affiliation(s)
- Xiaoxia Wang
- Department of Physiology and Cell Biology, University of Nevada School of Medicine, Reno, Nevada 89557, USA
| | | | | |
Collapse
|
37
|
Duprey-Díaz MV, Soto I, Blagburn JM, Blanco RE. Changes in brain-derived neurotrophic factor and trkB receptor in the adult Rana pipiens retina and optic tectum after optic nerve injury. J Comp Neurol 2002; 454:456-69. [PMID: 12455009 DOI: 10.1002/cne.10451] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
In this study we used immunocytochemistry to investigate the distribution of brain-derived neurotrophic factor (BDNF) and its receptor tyrosine kinase (trkB) in retina and optic tectum of the frog Rana pipiens during regeneration after axotomy. We also measured changes in BDNF mRNA in retina and tectum. Retrograde labeling was used to identify retinal ganglion cells (RGCs) prior to quantification of the BDNF immunoreactivity. In control animals, BDNF was found in the majority of RGCs and displaced amacrine cells and in some cells in the inner nuclear layer (INL). After axotomy, BDNF immunoreactivity was reduced in RGCs but increased in the INL. BDNF mRNA levels in the retina remained high before and after axotomy. Three months after axotomy, after reconnection to the target, the staining intensity of many of the surviving RGCs had partially recovered. In the control tectum, BDNF staining was present in ependymoglial cells and in neurons throughout layers 4, 6, 8, and 9. After axotomy, BDNF staining in tectal neurons became more intense, even though mRNA synthesis was transiently down-regulated. In control retinas, trkB receptor immunostaining was present in most RGCs; no significant changes were observed after axotomy. In control tectum, trkB was detected only in ependymoglial cells. After axotomy, many neuronal cell bodies were transiently labeled. Our data are consistent with the hypothesis that a considerable fraction of the BDNF normally present in RGCs is acquired from their targets in the tectum. However, there are also intraretinal sources of BDNF that could contribute to the survival of RGCs.
Collapse
Affiliation(s)
- Mildred V Duprey-Díaz
- Institute of Neurobiology and Department of Anatomy, University of Puerto Rico Medical Sciences Campus, San Juan, Puerto Rico 00901
| | | | | | | |
Collapse
|
38
|
Local and target-derived brain-derived neurotrophic factor exert opposing effects on the dendritic arborization of retinal ganglion cells in vivo. J Neurosci 2002. [PMID: 12196587 DOI: 10.1523/jneurosci.22-17-07639.2002] [Citation(s) in RCA: 77] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
The dendritic and axonal arbors of developing retinal ganglion cells (RGCs) are exposed to two sources of BDNF: RGC dendrites are exposed to BDNF locally within the retina, and RGC axons are exposed to BDNF at the target, the optic tectum. Our previous studies demonstrated that increasing tectal BDNF levels promotes RGC axon terminal arborization, whereas increasing retinal BDNF levels inhibits RGC dendritic arborization. These results suggested that differential neurotrophic action at the axon versus dendrite might be responsible for the opposing effects of BDNF on RGC axonal versus dendritic arborization. To explore this possibility, we examined the effects of altering BDNF levels at the optic tectum on the elaboration of RGC dendritic arbors in the retina. Increasing tectal BDNF levels resulted in a significant increase in dendritic branching, whereas neutralizing endogenous tectal BDNF with function-blocking antibodies significantly decreased dendritic arbor complexity. Thus, RGC dendritic arbors react in opposing manners to retinal- versus tectal-derived BDNF. Alterations in retinal BDNF levels, however, did not affect axon terminal arborization. Thus, RGC dendritic arborization is controlled in a complementary manner by both local and target-derived sources of BDNF, whereas axon arborization is modulated solely by neurotrophic interactions at the target. Together, our results indicate that developing RGCs modulate dendritic arborization by integrating signals from discrete sources of BDNF in the eye and brain. Differential integration of spatially discrete neurotrophin signals within a single neuron may therefore finely tune afferent and efferent neuronal connectivity.
Collapse
|
39
|
Sheedlo HJ, Srinivasan B, Brun-Zinkernagel AM, Roque CH, Lambert W, Wordinger RJ, Roque RS. Expression of p75(NTR) in photoreceptor cells of dystrophic rat retinas. BRAIN RESEARCH. MOLECULAR BRAIN RESEARCH 2002; 103:71-9. [PMID: 12106693 DOI: 10.1016/s0169-328x(02)00185-7] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
Although a gene mutation in the Royal College of Surgeons (RCS) dystrophic rat results in defective phagocytosis and in accumulation of debris in the subretinal space, the molecular mechanisms leading to photoreceptor cell death remain unclear. In this study, the expression of p75(NTR), the low-affinity neurotrophin receptor incriminated in the apoptosis of developing neurons, was investigated at various stages of retinal degeneration in dystrophic rats using immunohistochemistry, in situ reverse transcription polymerase chain reaction (RT-PCR), Western blot, and relative RT-PCR. In normal adult retinas, p75(NTR) immunolabeling was observed mainly in the outer limiting membrane, with punctate labeling in the inner nuclear and ganglion cell layers. In 18- to 30-day-old dystrophic retinas, the immunostaining appeared to increase especially in the photoreceptor outer and inner segments. Dense staining was also observed in the retinal pigment epithelium (RPE) and choroid. In 60-day-old dystrophic rat retinas, the density of immunolabeling for p75(NTR) increased dramatically in the remaining inner retina, especially in the inner nuclear, inner plexiform, and ganglion cell layers. Post-embedding immunogold labeling of normal retinas verified the distribution of p75(NTR) in photoreceptor cells within the inner segments, cell bodies, and outer segments. The apparent increased intensity in p75(NTR) immunostaining in dystrophic retinas was verified by Western blots and densitometric analyses. In situ RT-PCR and relative RT-PCR further established increased synthesis of p75(NTR) in dystrophic retinas. The increased levels of p75(NTR) in the RPE and photoreceptor cells, the initial sites of injury, during retinal degeneration in dystrophic rats strongly suggest that altered expression of p75(NTR) may be directly involved in photoreceptor death.
Collapse
Affiliation(s)
- Harold J Sheedlo
- Department of Pathology and Anatomy, University of North Texas Health Science Center, 3500 Camp Bowie Blvd., Fort Worth, TX 76107, USA
| | | | | | | | | | | | | |
Collapse
|
40
|
Karlsson M, Lindqvist N, Mayordomo R, Hallböök F. Overlapping and specific patterns of GDNF, c-ret and GFR alpha mRNA expression in the developing chicken retina. Mech Dev 2002; 114:161-5. [PMID: 12175504 DOI: 10.1016/s0925-4773(02)00045-x] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
GDNF and the GDNF receptors, c-Ret, GFR alpha 1 and 2 mRNA is expressed in the developing chicken retina. GDNF labelling was mainly found in embryonic day 4-5 retina but weak labelling could also be found over scattered retinal cells at later stages. c-ret labelling was found over ganglion cells, amacrine and horizontal cells; the preferred GDNF receptor (GFR alpha 1) over amacrine and horizontal cells; and the less preferred GDNF receptor (GFR alpha 2) over ganglion cells, amacrine cells and photoreceptors.
Collapse
Affiliation(s)
- Miriam Karlsson
- Department of Neuroscience, Uppsala University, Box 587, 751 23, Uppsala, Sweden
| | | | | | | |
Collapse
|
41
|
Reis RAM, Cabral da Silva MC, Loureiro dos Santos NE, Bampton E, Taylor JSH, de Mello FG, Linden R. Sympathetic neuronal survival induced by retinal trophic factors. JOURNAL OF NEUROBIOLOGY 2002; 50:13-23. [PMID: 11748629 DOI: 10.1002/neu.10008] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Neuronal survival in the vertebrate peripheral nervous system depends on neurotrophic factors available from target tissues. In an attempt to identify novel survival factors, we have studied the effect of secreted factors from retinal cells on the survival of chick sympathetic ganglion neurons. Embryonic day 10 sympathetic neurons undergo programmed cell death after 48 h without appropriate levels of nerve growth factor (NGF). Retina Conditioned Media (RCM) from explants of embryonic day 11 retinas maintained for 4 days in vitro supported 90% of E10 chick sympathetic neurons after 48 h. Conditioned medium from purified chick retinal Muller glial cells supported nearly 100% of E10 chick sympathetic neurons. Anti-NGF (1 microg/mL) blocked the survival effect of NGF, but did not block the trophic effect of RCM. Neither BDNF nor NT4 (0.1-50 ng/mL) supported E10 sympathetic neuron survival. Incubation of chimeric immunoglobulin-receptors TrkA, TrkB, or TrkC had no effect on RCM-induced sympathetic neuron survival. The survival effects were not blocked by anti-GDNF, anti-TGFbeta, and anti-CNTF and were not mimicked by FGFb (0.1-10 nM). LY294002 at 50 microM, but not PD098059 blocked sympathetic survival induced by RCM. Further, the combination of RCM and NGF did not result in an increase in neuronal survival compared with NGF alone (82% survival after 48 h). The secreted factor in RCM is retained in subfractions with a molecular weight above 100 kDa, binds to heparin, and is unaffected by dialysis, but is heat sensitive. Our results indicate the presence of a high-molecular weight retinal secreted factor that supports sympathetic neurons in culture.
Collapse
Affiliation(s)
- Ricardo A M Reis
- Laboratório de Neuroquímica, IBCCF(o), UFRJ, Rio de Janeiro 21949-900, Brazil.
| | | | | | | | | | | | | |
Collapse
|
42
|
Sorting of internalized neurotrophins into an endocytic transcytosis pathway via the Golgi system: Ultrastructural analysis in retinal ganglion cells. J Neurosci 2001. [PMID: 11698603 DOI: 10.1523/jneurosci.21-22-08915.2001] [Citation(s) in RCA: 56] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Subcellular pathways and accumulation of internalized radiolabeled neurotrophins NGF, BDNF, and NT-3 were examined in retinal ganglion cells (RGCs) of chick embryos by using quantitative electron microscopic autoradiography. All three neurotrophins accumulated in endosomes and multivesicular bodies. BDNF and NGF also concentrated at the plasma membrane, whereas NT-3 accumulated transiently in the Golgi system. The enhanced targeting of NT-3 to the Golgi system correlated with the anterograde axonal transport of this neurotrophin. Anterograde transport of NT-3, but not its internalization, was significantly attenuated by the tyrosine kinase (trk) inhibitor K252a. Abolishment of trk activity with K252a shifted NT-3 (and BDNF) away from the Golgi system and into a lysosomal pathway, indicating that trk activity regulated sorting of the ligand-receptor complex. Cross-linking of neurotrophins and immunoprecipitation with antibodies to the neurotrophin receptors p75, trkA, trkB, and trkC showed that the large majority of exogenous, receptor-bound NT-3 was bound to trkC in RGC somata, but during anterograde transport in the optic nerve most receptor-bound NT-3 was associated with p75, and after arrival and release in the optic tectum transferred to presumably postsynaptic trkC. These results reveal remarkable and unexpected differences in the intracellular pathways and fates of different neurotrophins within the same cell type. They provide first evidence for an endocytic pathway of internalized neurotrophic factors via the Golgi system before anterograde transport and transcytosis. The results challenge the belief that after internalization all neurotrophins are rapidly degraded in lysosomes.
Collapse
|
43
|
Tu S, Debski EA. Neurotrophins, but not depolarization, regulate substance P expression in the developing optic tectum. ACTA ACUST UNITED AC 2001. [PMID: 11438942 DOI: 10.1002/neu.1047] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Neurotransmitter expression can be regulated by both activity and neurotrophins in a number of in vitro systems. We examined whether either of these factors was likely to play a role in the in vivo optic nerve-dependent regulation of a substance P-like immunoreactive (SP-ir) population of cells in the developing optic tectum of the frog. In contrast to our previous results with the adult system, blocking tectal cell responses to glutamate release by retinal ganglion cells with 6-cyano-7-nitroquinoxaline-2,3 dione (CNQX) did not affect the percent of SP-ir cells in the developing tectum. Treatment with d-(-)-2-amino-5-phosphonovaleric acid (d-AP-5) was also ineffective in this regard, although both it and CNQX treatment disrupted visual map topography. Chronic treatment with brain-derived neurotrophic factor (BDNF) and neurotrophin-4/5 (NT-4/5) produced increases in SP-ir cells in the treated lobes of normal animals, which were significant in the case of NT-4/5. Both substances also prevented the decrease of SP cells that would otherwise occur in the deafferented lobe of unilaterally optic nerve-transected tadpoles. These changes in the percent of SP-ir cells occurred without any detectable changes in the overall number of tectal cells. NGF had no effect on SP expression. Nor did it affect topographic map formation, which was disrupted by treatment with either BDNF or NT-4/5. Our results demonstrate that different mechanisms regulate SP expression in the developing and adult tectum. They indicate that neurotrophin levels in the developing optic tectum may selectively regulate a specific neuropeptide-expressing population of cells.
Collapse
Affiliation(s)
- S Tu
- School of Biological Sciences, University of Kentucky, Lexington, Kentucky 40506, USA
| | | |
Collapse
|
44
|
Lund RD, Kwan AS, Keegan DJ, Sauvé Y, Coffey PJ, Lawrence JM. Cell transplantation as a treatment for retinal disease. Prog Retin Eye Res 2001; 20:415-49. [PMID: 11390255 DOI: 10.1016/s1350-9462(01)00003-9] [Citation(s) in RCA: 104] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
It has been shown that photoreceptor degeneration can be limited in experimental animals by transplantation of fresh RPE to the subretinal space. There is also evidence that retinal cell transplants can be used to reconstruct retinal circuitry in dystrophic animals. Here we describe and review recent developments that highlight the necessary steps that should be taken prior to embarking on clinical trials in humans.
Collapse
Affiliation(s)
- R D Lund
- Institute of Ophthalmology, Bath Street, EC1V 9EL, London, UK
| | | | | | | | | | | |
Collapse
|
45
|
Dünker N, Schuster N, Krieglstein K. TGF-β modulates programmed cell death in the retina of the developing chick embryo. Development 2001; 128:1933-42. [PMID: 11493517 DOI: 10.1242/dev.128.11.1933] [Citation(s) in RCA: 64] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Programmed cell death (PCD) is a key phenomenon in the regulation of cell number in multicellular organisms. We have shown that reduction of endogenous transforming growth factor β (TGF-β) prevents apoptotic PCD of neurons in the developing peripheral and central nervous system, suggesting that TGF-β is an important mediator of ontogenetic neuron death. Previous studies suggested that there are other pro-apoptotic molecules, nerve growth factor (NGF) and brain-derived neurotrophic factor, that induce cell death in the nervous system. In the developing chick retina, NGF induces PCD by activation of the p75 receptor. We have studied the role of TGF-β and its putative interdependence with NGF-mediated PCD in the chick retina. We found that TGF-β is present in the developing chick retina during the period of PCD and is essentially required to regulate PCD of retinal cells. TGF-β2, TGF-β3 and the ligand-binding TGF-β receptor can be detected immunocytochemically in the central retina, a region where apoptosis is most prominent during the early period of PCD. Application of a TGF-β-neutralizing antibody to chick embryos in ovo resulted in a decrease in the number of TUNEL-positive cells and a reduction of free nucleosome levels. In terms of magnitude, reduction of PCD caused by the neutralization of endogenous TGF-β was equivalent to that seen after anti-NGF application. Neutralization of both factors did not result in a further decrease in apoptosis, indicating that NGF and TGF-β may act on the same cell population. Furthermore, neutralization of TGF-β did not affect the expression of NGF or the p75-receptor. Our results suggest that TGF-β and NGF are both required to regulate cell death in the chick retina in vivo.
Collapse
Affiliation(s)
- N Dünker
- University of Saarland, Department of Anatomy, D-66421 Homburg/Saar, Germany
| | | | | |
Collapse
|
46
|
Von Bartheld CS, Johnson JE. Target-derived BDNF (brain-derived neurotrophic factor) is essential for the survival of developing neurons in the isthmo-optic nucleus. J Comp Neurol 2001; 433:550-64. [PMID: 11304717 DOI: 10.1002/cne.1159] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
Neurons in the peripheral nervous system depend on single neurotrophic factors, whereas those in the brain are thought to utilize many different trophic factors. This study examined whether some neurons in the brain critically depend on a single trophic factor during development. Neurons in the isthmo-optic nucleus (ION) of chick embryos respond to exogenous brain-derived neurotrophic factor (BDNF). Relatively high concentrations of endogenous BDNF were present in the ION of 14-18-day-old chick embryos. ION target cells in the retina were immunolabeled for BDNF but showed surprisingly low levels of BDNF mRNA. These data suggest that ION target cells derive some BDNF from other retinal sources. No BDNF mRNA was detected in the ION itself. ION neurons had a very efficient retrograde transport system for BDNF and exogenous BDNF arrived in the ION intact. When the ION was deprived of endogenous trkB ligands by injection of trkB fusion proteins in the eye, cell death of ION neurons was enhanced, and this effect was mimicked by BDNF-specific blocking antibodies in the eye. TrkB fusion proteins in the retina induced cell death of ION neurons prior to visible effects on ION target cells in the retina. Immunolabel for endogenous BDNF was sparse in pyknotic ION neurons, suggesting that ION neurons with low BDNF content were eliminated by apoptosis. These data show that BDNF is an essential target-derived trophic factor for developing ION neurons and thereby validate the neurotrophic hypothesis for at least one neuronal population in the brain.
Collapse
Affiliation(s)
- C S Von Bartheld
- Department of Physiology and Cell Biology, University of Nevada School of Medicine, Reno, Nevada 89557, USA.
| | | |
Collapse
|
47
|
Haamedi SN, Karten HJ, Djamgoz MB. Nerve growth factor induces light adaptive cellular and synaptic plasticity in the outer retina of fish. J Comp Neurol 2001; 431:397-404. [PMID: 11223810 DOI: 10.1002/1096-9861(20010319)431:4<397::aid-cne1078>3.0.co;2-p] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Recent evidence suggests that neurotrophins can be involved in short-term synaptic plasticity in parts of the central nervous system. In the present study, the possible role of nerve growth factor (NGF) in inducing morphologic (cellular and subcellular) changes in the outer retina of carp was assessed. The effects of NGF on cone photomechanical movements (PMMs) and horizontal cell (HC) spinule formation were measured. NGF-induced cone contraction and formation of HC spinules in the dark-adapted retina were consistent with its role in light adaptation. These effects were dose dependent in the range of 5--250 nM. Because cone contraction and HC spinule formation have previously been shown to be controlled by dopamine (DA), nitric oxide (NO), or both, the possibility that the effects of NGF could be occurring by means of release of DA and/or NO was tested. Haloperidol (HAL), a nonspecific DA receptor blocker, or 2-(4-carboxyphenyl)-4,4,5,5-tetramethyl-imidazoline-1-oxyl-3-oxide potassium (cPTIO), a NO scavenger, was applied in combination with NGF to dark-adapted eyecups. The results showed that both HAL and cPTIO significantly blocked the effects of NGF on cone PMMs and HC spinule formation. In conclusion, (1) NGF represents a novel light-adaptive signalling mechanism in the outer retina of fish; and (2) NGF-induced cone contraction and HC spinule formation in the retina together with our previous observation would suggest that the effects of NGF may be mediated through NO by means of DA.
Collapse
Affiliation(s)
- S N Haamedi
- Neurobiology Group, Department of Biology, Imperial College of Science, Technology and Medicine, London SW7 2AZ, United Kingdom.
| | | | | |
Collapse
|
48
|
Fuhrmann S, Chow L, Reh TA. Molecular control of cell diversification in the vertebrate retina. Results Probl Cell Differ 2001; 31:69-91. [PMID: 10929402 DOI: 10.1007/978-3-540-46826-4_5] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/17/2023]
Affiliation(s)
- S Fuhrmann
- Department of Biological Structure, University of Washington, Seattle 98195, USA
| | | | | |
Collapse
|
49
|
Karlsson M, Mayordomo R, Reichardt LF, Catsicas S, Karten HJ, Hallböök F. Nerve growth factor is expressed by postmitotic avian retinal horizontal cells and supports their survival during development in an autocrine mode of action. Development 2001; 128:471-9. [PMID: 11171331 PMCID: PMC2710126 DOI: 10.1242/dev.128.4.471] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Cell death in the developing retina is regulated, but so far little is known about what factors regulate the cell death. Several neurotrophic factors and receptors, including the neurotrophins and Trk receptors, are expressed during the critical time. We have studied the developing avian retina with respect to the role of nerve growth factor (NGF) in these processes. Our starting point for the work was that NGF and its receptor TrkA are expressed in a partially overlapping pattern in the inner nuclear layer of the developing retina. Our results show that TrkA and NGF-expressing cells are postmitotic. The first NGF-expressing cells were found on the vitreal side of the central region of E5.5-E6 retina. This pattern changed and NGF-expressing cells identified as horizontal cells were later confined to the external inner nuclear layer. We show that these horizontal cells co-express TrkA and NGF, unlike a subpopulation of amacrine cells that only expresses TrkA. In contrast to the horizontal cells, which survive, the majority of the TrkA-expressing amacrine cells die during a period of cell death in the inner nuclear layer. Intraocular injections of NGF protein rescued the dying amacrine cells and injection of antisense oligonucleotides for NGF that block its synthesis, caused death among the TrkA-expressing horizontal cells, which normally would survive. Our results suggest that NGF supports the survival of TrkA expressing avian horizontal cells in an autocrine mode of action in the retina of E10-E12 chicks. The cells co-express TrkA and NGF and the role for NGF is to maintain the TrkA-expressing horizontal cells. The TrkA-expressing amacrine cells are not supported by NGF and subsequently die. In addition to the effect on survival, our results suggest that NGF plays a role in horizontal cell plasticity.
Collapse
Affiliation(s)
- Miriam Karlsson
- Department of Neuroscience, Unit of Developmental Neuroscience, Biomedical Center, Uppsala University, S-751 23 Uppsala, Sweden
| | - Raquel Mayordomo
- Department of Neuroscience, Unit of Developmental Neuroscience, Biomedical Center, Uppsala University, S-751 23 Uppsala, Sweden
| | | | - Stefan Catsicas
- IBCM, Medical Faculty, University of Lausanne, Lausanne, Switzerland
| | - Harvey J. Karten
- Department of Neuroscience, University of California, San Diego, La Jolla, CA 92093, USA
| | - Finn Hallböök
- Department of Neuroscience, Unit of Developmental Neuroscience, Biomedical Center, Uppsala University, S-751 23 Uppsala, Sweden
| |
Collapse
|
50
|
Viant MR, Millam JR, Delany ME, Fry DM. Regulation of brain-derived neurotrophic factor messenger RNA levels in avian hypothalamic slice cultures. Neuroscience 2000; 99:373-80. [PMID: 10938443 DOI: 10.1016/s0306-4522(00)00167-6] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
Mechanisms regulating the expression of brain-derived neurotrophic factor, a member of the neurotrophin family, have been extensively studied in the rat cerebral cortex, hippocampus and cerebellum. In contrast, little is known regarding the regulation of this growth factor in the hypothalamus. Here we present an analysis of the regulation of brain-derived neurotrophic factor messenger RNA levels in chick embryo hypothalamic slice cultures following exposure to potassium chloride, glutamate agonists and sex steroids. Following a week in chemically-defined media the tissue was depolarized by exposure to 50 mM potassium chloride for 6h, resulting in a significant 4.2-fold increase in the level of brain-derived neurotrophic factor messenger RNA. This result is consistent with studies of other brain regions. Similar 6-h acute exposures of the hypothalamic cultures to 25 microM N-methyl-D-aspartic acid, 25 microM kainic acid and 25 microM alpha-amino-3-hydroxy-5-methyl-isoxazole-4-propionic acid also significantly increased messenger RNA levels 2.5-, 2.1- and 1.4-fold, respectively. It was previously reported that brain-derived neurotrophic factor levels within the rat cerebral cortex, olfactory bulb and hippocampus are altered by exposure to 17beta-estradiol. Here we show that in hypothalamic slice cultures neither acute nor chronic treatments with 10 and 100 nM 17beta-estradiol and 10nM testosterone significantly altered the steady-state level of this growth factor. These findings show that neuronal activity, induced by glutamate agonists and potassium chloride, can regulate brain-derived neurotrophic factor messenger RNA levels within embryonic hypothalamic slice cultures. This regulation could play a critical role in the modulation of programmed cell death and synaptic maturation during development of the hypothalamus.
Collapse
Affiliation(s)
- M R Viant
- Department of Animal Science, University of California, Davis, CA 95616, USA
| | | | | | | |
Collapse
|