Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters










Publication year range
1.
Proc Natl Acad Sci U S A ; 102(33): 11900-5, 2005 Aug 16.
Article in English | MEDLINE | ID: mdl-16079201

ABSTRACT

Age-related macular degeneration (AMD) is a late-onset, multifactorial, neurodegenerative disease of the retina and the leading cause of irreversible vision loss in the elderly in the Western world. We describe here a murine model that combines three known AMD risk factors: advanced age, high fat cholesterol-rich (HF-C) diet, and apolipoprotein E (apoE) genotype. Eyes of aged, targeted replacement mice expressing human apoE2, apoE3, or apoE4 and maintained on a HF-C diet show apoE isoform-dependent pathologies of differential severity. ApoE4 mice are the most severely affected. They develop a constellation of changes that mimic the pathology associated with human AMD. These alterations include diffuse sub-retinal pigment epithelial deposits, drusenoid deposits, thickened Bruch's membrane, and atrophy, hypopigmentation, and hyperpigmentation of the retinal pigment epithelium. In extreme cases, apoE4 mice also develop marked choroidal neovascularization, a hallmark of exudative AMD. Neither age nor HF-C diet alone is sufficient to elicit these changes. We document choroidal neovascularization and other AMD-like ocular pathologies in an animal model that exploits known AMD risk factors. The model is additionally attractive because it is not complicated by invasive experimental intervention. Our findings in this model implicate the human apoE E4 allele as a susceptibility gene for AMD and support the hypothesis that common pathogenic mechanisms may underlie AMD and Alzheimer's disease.


Subject(s)
Aging/physiology , Alleles , Apolipoproteins E/genetics , Apolipoproteins E/metabolism , Retinal Degeneration/genetics , Retinal Degeneration/pathology , Animal Feed , Animals , Cholesterol/pharmacology , Female , Humans , Male , Mice , Mice, Transgenic , Microscopy, Electron , Models, Biological , Retinal Degeneration/metabolism , Vascular Endothelial Growth Factor A/metabolism
2.
Eye (Lond) ; 19(4): 455-63, 2005 Apr.
Article in English | MEDLINE | ID: mdl-15184939

ABSTRACT

PURPOSE: Vitreous and retinal amino-acid concentrations were evaluated in a primate model of central retinal artery occlusion (CRAO) to study the role of glutamate excitotoxicity in acute retinal ischaemia. METHODS: Unilateral, acute CRAO was produced by temporary clamping of the central retinal artery for 190 min in four elderly rhesus monkeys. Fundus photography, fluorescein angiography, and electroretinogram were performed before and during CRAO, and after unclamping the artery. Vitreous samples were obtained before and after CRAO in both eyes, and analysed for 13 amino-acid concentrations using high-pressure liquid chromatography. The animals were killed 350 min after retinal reperfusion, and the retinal tissue was submitted for amino-acid analysis. RESULTS: In all four eyes, the macula showed the 'cherry red spot'. The CRAO was confirmed by fluorescein angiography and decreased b-wave on electroretinogram. Retinal histology confirmed ischaemic changes in the inner retina. Changes in all 13 vitreous amino-acid concentrations after CRAO (including glutamate) were not significantly different between study and control eyes (P = 0.09 to 0.82). All retinal amino-acid concentrations (including glutamate) were not significantly different between two eyes (P = 0.07-0.93). CONCLUSIONS: In the primate model of acute inner retinal ischaemia induced by transient CRAO, we were unable to detect significantly elevated concentrations of vitreous and retinal glutamate. Our primate model has the advantage of closely modelling the CRAO in humans. Further basic and clinical studies are needed to elucidate the role of glutamate excitotoxicity in retinal ischaemia.


Subject(s)
Amino Acids/metabolism , Retina/metabolism , Retinal Artery Occlusion/metabolism , Vitreous Body/metabolism , Acute Disease , Animals , Chromatography, High Pressure Liquid/methods , Disease Models, Animal , Electroretinography , Fluorescein Angiography/methods , Fundus Oculi , Glutamic Acid/metabolism , Macaca mulatta , Retinal Artery Occlusion/etiology , Specimen Handling/methods
3.
Microsc Res Tech ; 50(2): 103-11, 2000 Jul 15.
Article in English | MEDLINE | ID: mdl-10891874

ABSTRACT

This review discusses the expression and cellular localization of the neuropeptide somatostatin (SRIF) and one of the SRIF subtype (sst) receptors, sst(2A) in the mammalian retina. SRIF immunoreactivity is predominantly localized to a sparse population of amacrine and displaced amacrine cells in the ganglion cell layer in several mammalian retinas including the rat, rabbit, cat, and primate. These cells, characterized by multiple processes, form a sparse network in the inner plexiform layer (IPL) in all retinal regions. Very few processes are also in the outer plexiform layer. In contrast to the predominant distribution of SRIF processes to the IPL, there is a widespread distribution of sst(2A) immunoreactivity to both the inner and outer retina in all mammalian retinas studied to date. In rabbit retina, sst(2A) immunoreactivity is predominant in rod bipolar cells and in sparse wide-field amacrine cells. In the rat retina, sst(2A) immunoreactivity is localized to several neuronal cell types-cone photoreceptors, horizontal cells, rod and cone bipolar cells, and amacrine cells. Reverse-transcriptase-polymerase chain reaction analysis found that sst(2A) mRNA is expressed in the rat retina, while sst(2B) mRNA is not detected. Finally, in the primate retina sst(2) immunoreactivity is predominant in cone photoreceptors, with additional immunostained cell bodies and processes in the inner retina. These findings indicate that SRIF may modulate several neuronal cell types in the retina, and that it has a broad influence on both scotopic and photopic visual pathways.


Subject(s)
Receptors, Somatostatin/metabolism , Retina/physiology , Somatostatin/metabolism , Animals , Humans , Immunohistochemistry , Mammals , Paracrine Communication , RNA, Messenger/analysis , Receptors, Somatostatin/analysis , Receptors, Somatostatin/genetics , Retina/cytology , Retina/metabolism , Retinal Cone Photoreceptor Cells/metabolism , Retinal Rod Photoreceptor Cells/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Somatostatin/analysis
4.
Microsc Res Tech ; 50(2): 124-9, 2000 Jul 15.
Article in English | MEDLINE | ID: mdl-10891876

ABSTRACT

The rodent retina is a particularly attractive model for the study of neuronal developmental processes since considerable neurogenesis, cellular migration, phenotypic differentiation of retinal cell types and synaptogenesis occurs postnatally. In addition, the retina is readily accessible to surgical intervention, pharmacological manipulation, and local suppression of gene expression-tools that can be utilized to study mechanisms underlying the development of retinal neurons and their interconnections that form distinct functional circuits. Here, I review our studies describing the ontogeny of a specific retinal interneuron, the AII amacrine cell, an integral element in the rod (scotopic) pathway. Specifically, we used a number of approaches to examine the potential role of neurotrophic factors on the morphological and neurochemical differentiation of the AII.


Subject(s)
Nerve Growth Factors/metabolism , Retina/metabolism , Animals , Brain-Derived Neurotrophic Factor/genetics , Brain-Derived Neurotrophic Factor/metabolism , Culture Techniques , Immunohistochemistry , In Situ Hybridization , Interneurons/metabolism , Nerve Growth Factor/genetics , Nerve Growth Factor/metabolism , Oligonucleotides, Antisense/pharmacology , Parvalbumins/analysis , Parvalbumins/metabolism , RNA, Messenger/analysis , Receptor, trkB/metabolism , Retina/drug effects , Retina/growth & development , Retinal Rod Photoreceptor Cells/metabolism , Thionucleotides/pharmacology
5.
Brain Res Mol Brain Res ; 77(1): 95-103, 2000 Apr 14.
Article in English | MEDLINE | ID: mdl-10814835

ABSTRACT

Foveal cone photoreceptors are morphologically distinct and, presumably, express unique transcripts. We have identified a cDNA clone encoding the protein tyrosine phosphatase (PTP), phosphatase of regenerating liver 1 (PRL-1) in a screen for genes that are enriched in monkey fovea. PRL-1 was originally isolated as an immediate early gene in regenerating liver [R.H. Diamond, D.E. Cressman, T.M. Laz, C.S. Abrams, R. Taub, PRL-1, a unique nuclear protein tyrosine phosphatase, affects cell growth, Mol. Cell Biol. 14 (1994) 3752-3762]. On cDNA Southern blots of human and monkey retina, radiolabeled PRL-1 cDNA hybridized to a single mRNA species of about 2.5 kb that was most intense in fovea-enriched samples. The monkey PRL-1 deduced amino acid sequence is identical to human, rat and mouse PRL-1. Affinity-purified antibodies directed against PRL-1 preferentially labeled cone photoreceptor cells and a subpopulation of bipolar cells in monkey retina. Immunoreactivity in cones was confined to red and green, but not to blue, cones and was restricted to the outer segments. Immunolocalization also revealed that PRL-1 protein expression was non-nuclear, suggesting that its function in the retina may be unrelated to its role in other tissues where it is expressed primarily in nuclei. Although both foveal and extrafoveal cones were PRL-1 reactive, the high abundance of PRL-1 mRNAs detected in monkey fovea correlates with the high concentration of cones in the fovea. The PRL-1 gene is located on chromosome 6q within an interval that also contains the genes that cause two hereditary retinal dystrophies. These studies demonstrate novel expression of the PRL-1 gene in the neural retina and suggest the phosphatase activity of PRL-1 may modulate normal cone photoreceptor cell function.


Subject(s)
Immediate-Early Proteins/genetics , Protein Tyrosine Phosphatases/genetics , Retinal Cone Photoreceptor Cells/enzymology , Animals , Cell Cycle Proteins , Cloning, Molecular , Humans , Immediate-Early Proteins/analysis , Immunohistochemistry , Macaca fascicularis , Membrane Proteins , Mice , Neoplasm Proteins , Protein Tyrosine Phosphatases/analysis , Rats , Retinal Cone Photoreceptor Cells/cytology , Reverse Transcriptase Polymerase Chain Reaction , Sequence Analysis, DNA
6.
J Comp Neurol ; 421(2): 275-87, 2000 May 29.
Article in English | MEDLINE | ID: mdl-10813787

ABSTRACT

Tachykinin (TK) peptides act on retinal neurons through neurokinin (NK) receptors. We examined the expression of neurokinin-1 (NK1; the substance P receptor), NK3 [the neurokinin B (NKB) receptor], and TK peptides in developing rat retinas. NK1 immunolabeling was found in newborn retinas in rare amacrine cells and in putative ganglion cells. At postnatal day 2 (PND 2), NK1 immunostaining was reduced greatly among ganglion cells, and it appeared in many amacrine cells and in fibers in the inner plexiform layer (IPL), with the highest density in laminae 1, 3, and 5. A similar pattern was found at PND 7. At PND 12, interplexiform NK1-immunoreactive (-IR) cells were detected, and NK1-IR fibers in the IPL were concentrated in lamina 2, similar to what was seen in adults. NK3 was expressed mainly by OFF-cone bipolar cells, and the developmental pattern of NK3 was compared with that of cone bipolar cells that were labeled with antibodies to recoverin. Immature recoverin-IR cone bipolar cells were seen at PND 2. NK3 immunolabeling was detected first in the outer plexiform layer and in sparse bipolar cell somata at PND 10, when recoverin-IR cone bipolar cells are nearly mature. By PND 15, both the NK3 immunostaining pattern and the recoverin immunostaining pattern were similar to the patterns seen in adults. TK immunoreactivity was present at PND 0 in amacrine cells and displaced amacrine cells. By PND 10, the morphologic maturation of TK-IR cells was complete. These findings indicate that, in early postnatal retinas, substance P may act on NK1 receptors, whereas NKB/NK3 interactions are unlikely, suggesting that there are different levels of importance for different TK peptides in the developing retina.


Subject(s)
Receptors, Neurokinin-1/metabolism , Receptors, Neurokinin-3/metabolism , Retina/metabolism , Animals , Animals, Newborn , Female , Pregnancy , Rats , Rats, Sprague-Dawley , Retina/growth & development , Retinal Cone Photoreceptor Cells/metabolism , Retinal Ganglion Cells/metabolism , Tachykinins/metabolism
7.
J Neurobiol ; 41(3): 376-84, 1999 Nov 15.
Article in English | MEDLINE | ID: mdl-10526316

ABSTRACT

The rodent retina undergoes considerable postnatal neurogenesis and phenotypic differentiation, and it is likely that diffusible neurotrophic factors contribute to this development and to the subsequent formation of functional retinal circuitry. Accordingly, perturbation of specific neurotrophin ligand-receptor interactions has provided valuable information as to the fundamental processes underlying this development. In the present studies we have built upon our previous observation that suppression of expression of trk(B), the high-affinity receptor for brain-derived neurotrophic factor (BDNF), in the postnatal rat retina results in the alteration of a specific interneuron in the rod pathway-the parvalbumin (PV)-immunoreactive AII amacrine cell. Here, we isolated retinas from newborn rats and maintained them in organotypic culture for up to 14 days (approximating the time of eye opening, in vivo) in the presence of individual neurotrophins [BDNF or nerve growth factor (NGF)]. We then examined histological sections of cultures for PV immunoreactivity. In control cultures, only sparse PV-immunostained cells were observed. In cultures supplemented with NGF, numerous lightly immunostained somata were present in the inner nuclear layer (INL) at the border of the inner plexiform layer (IPL). Many of these cells had rudimentary dendritic arborizations in the IPL. Cultures supplemented with BDNF displayed numerous well-immunostained somata at the INL/IPL border that gave rise to elaborate dendritic arborizations that approximated the morphology of mature AII amacrine cells in vivo. These observations indicate that neurotrophins have specific effects upon the neurochemical and, perhaps, morphological differentiation of an important interneuron in a specific functional retinal circuit.


Subject(s)
Brain-Derived Neurotrophic Factor/pharmacology , Parvalbumins/analysis , Retina/chemistry , Retina/growth & development , Retinal Ganglion Cells/chemistry , Animals , Brain-Derived Neurotrophic Factor/physiology , Immunohistochemistry , Organ Culture Techniques , Parvalbumins/physiology , Rats , Rats, Sprague-Dawley , Retina/cytology , Retinal Ganglion Cells/drug effects , Retinal Ganglion Cells/physiology
8.
Brain Res Dev Brain Res ; 115(1): 41-7, 1999 Jun 08.
Article in English | MEDLINE | ID: mdl-10366701

ABSTRACT

Neuronal programmed cell death, or apoptosis, occurs during development, following injury or in certain disease processes, and is regulated by members of the B-cell leukemia-2 (Bcl-2) protein family. These molecules include both positive and negative regulators of cell death and act by selective dimerization that results in permissive or inhibitory effects on a cascade of cellular events, including mitochondrial release of cytochrome c, stimulation of cysteine protease activity and subsequent cellular deterioration. Here, we have characterized the expression of the cell death agonist, Bad, in the postnatal rat retina and forebrain. Isolation, subsequent amplification by RT-PCR and DNA sequence analysis revealed that retinal Bad was identical to Bad expressed in the developing and adult rat brain. Using a polyclonal antibody to Bad, we determined that, in the retina, on the day of birth (postnatal day-0, PND-0) Bad immunoreactivity was expressed primarily by retinal ganglion cells, some cells in the inner neuroblastic layer (NBL) and an indistinct plexus of processes in the inner plexiform layer (IPL). On PND-7, Bad immunoreactivity was observed in most cells in the ganglion cell layer (GCL), numerous cells scattered throughout the inner nuclear layer (INL), a lightly stained IPL and in a distinct band of immunostained fibers in the forming outer plexiform layer (OPL). By PND-15, Bad immunoreactivity was present in cells in the GCL, in some cells in the proximal INL and in horizontal cell processes in the OPL. The IPL was only faintly labeled. In the adult retina, specific Bad immunostaining was confined to large cells in the ganglion cell layer (presumed ganglion cells), occasional lightly stained horizontal cells and their processes in the OPL and to occasional small, lightly stained cells in the proximal INL (presumed amacrine cells) and GCL (presumed displaced amacrine cells). Again, the interposed IPL was faintly labeled. In the brain, Bad immunoreactive cells were scattered throughout the forebrain parenchyma but were particularly concentrated in neurons of the cerebral cortex, hippocampus and amygdala. Bad immunoreactivity was heaviest in these cells at PND-7, distinctly weaker at PND-10 and absent by PND-24. At all time points examined, Bad immunoreactivity was present in epithelial cells of the choroid plexus, as previously reported in the adult rat brain. These data suggest that Bad is transiently expressed by various cell types in the perinatal retina, particularly ganglion cells, and in discrete forebrain regions. In the context of corroborative observations, Bad expression may be regulated in response to acute ischemia and may act as a control point for retinal neuronal apoptosis.


Subject(s)
Carrier Proteins/genetics , Promoter Regions, Genetic , Prosencephalon/growth & development , Retina/growth & development , Animals , Cell Death , Immunohistochemistry , Rats , Rats, Sprague-Dawley , Reverse Transcriptase Polymerase Chain Reaction , bcl-Associated Death Protein
9.
Brain Res Dev Brain Res ; 111(1): 107-17, 1998 Nov 01.
Article in English | MEDLINE | ID: mdl-9804913

ABSTRACT

In the adult rabbit, rat and cat retina, parvalbumin (PV) immunoreactivity is primarily localized to a population of narrow-field, bistratified amacrine cells, the AII amacrine cells-major interneurons of the rod pathway. This investigation examines the postnatal development of PV immunoreactivity in order to better understand the ontogeny of the AII amacrine cell population and the formation of the rod pathway. Rabbit retinas at various postnatal ages were processed for immunohistochemistry using a monoclonal antibody directed to PV and analyzed morphometrically. On the day of birth, PV immunoreactive cell bodies are numerous in the proximal inner nuclear layer (INL) in all retinal regions. These cells have a primary process directed towards the inner plexiform layer (IPL). At postnatal day (PND) 2, a few faint immunoreactive processes are observed in the IPL. At PND 4, well-stained processes are observed to ramify mainly in the proximal IPL. At PND 6, strongly immunoreactive processes are present in both the distal and proximal IPL, and at PND 10 they form a continuous, dense plexus in both levels of the IPL. By PND 10, the morphology of PV immunoreactive cells is similar to PV immunoreactive cells in adult retinas. The density of PV immunoreactive cells in the proximal INL increases from PND 2 to PND 5, then it gradually decreases to adult values, while the total number of PV immunoreactive cell bodies increases until PND 10. PV immunoreactive amacrine cells at PND 2, as in the adult, are nonrandomly distributed across the retinal surface. These studies show that PV immunoreactive amacrine cells have a developmental profile that is similar to several other amacrine cell types. This includes the elaboration of processes in the IPL during the first postnatal week and a mature appearance towards the end of the second week of life, about the time of eye opening. These observations indicate that the AII amacrine cell may participate in the processing of visual information at eye opening.


Subject(s)
Aging/metabolism , Animals, Newborn/metabolism , Parvalbumins/metabolism , Retina/metabolism , Animals , Animals, Newborn/growth & development , Cell Count , Immunohistochemistry , Rabbits , Retina/cytology , Retina/growth & development
10.
J Comp Neurol ; 389(3): 496-507, 1997 Dec 22.
Article in English | MEDLINE | ID: mdl-9414009

ABSTRACT

Tachykinin (TK) peptides influence neuronal activity in the inner retina of mammals. The aim of this investigation was to determine the cellular localization of the neurokinin 1 receptor (NK1), whose preferred ligand is the TK peptide substance P (SP), in the rat retina. These studies used a polyclonal antiserum directed to the C-terminus of rat NK1. The majority of NK1-immunoreactive (IR) cells were located in the proximal inner nuclear layer (INL), and very rarely they were found in the distal INL. Some small and large NK1-IR somata were present in the ganglion cell layer. NK1-IR processes were densely distributed across the inner plexiform layer (IPL) with a maximum density over lamina 2 of the IPL. Immunoreactive processes also crossed the INL and ramified in the outer plexiform layer where they formed a sparse meshwork. NK1-IR processes were rarely observed in the optic nerve fiber layer. Double-label immunofluorescence studies with different histochemical markers for bipolar cells indicated that NK1 immunoreactivity was not present in bipolar cells. Together, these observations indicate that NK1 immunoreactivity is predominantly expressed by amacrine, displaced amacrine, interplexiform, and some ganglion cells. Double-label immunofluorescence experiments were also performed to characterize NK1-containing amacrine cells. Sixty-one percent of the gamma-aminobutyric acid (GABA)-IR cells, 71% of the large tyrosine hydroxylase (TH)-IR cells, and 100% of the small TH-IR cells contained NK1 immunoreactivity. In addition, most (91%) of the NK1-IR cells had GABA immunoreactivity. In contrast, vasoactive intestinal polypeptide-, TK-, choline acetyltransferase-, and parvalbumin-IR amacrine tells did not express NK1 immunoreactivity. Overall, the present findings suggest that SP acts directly upon several cell populations, including GABA-containing amacrine cells and ganglion cells, to influence visual information processing in the inner retina.


Subject(s)
Neurons/chemistry , Receptors, Neurokinin-1/analysis , Retina/chemistry , Animals , Immunohistochemistry , Rats , Rats, Sprague-Dawley , Retina/cytology
11.
J Comp Neurol ; 375(2): 212-24, 1996 Nov 11.
Article in English | MEDLINE | ID: mdl-8915826

ABSTRACT

gamma-Aminobutyric acid (GABA) plasma membrane transporters (GATs) influence synaptic neurotransmission by high-affinity uptake and release of GABA. The distribution and cellular localization of GAT-1, GAT-2, and GAT-3 in the rat retina have been evaluated by using affinity-purified polyclonal antibodies directed to the C terminus of each of these GAT subtypes. Small GAT-1-immunoreactive cell bodies were located in the proximal inner nuclear layer (INL) and ganglion cell layer (GCL), and processes were distributed to all laminae of the interplexiform layer (IPL). Varicose processes were in the optic fiber layer (OFL) and the outer plexiform layer (OPL). Weak GAT-1 immunostaining surrounded cells in the INL and GCL, and it was found in the OFL and OPL and in numerous processes in the outer nuclear layer (ONL) that ended at the outer limiting membrane. GAT-1 is therefore strongly expressed by amacrine, displaced amacrine, and interplexiform cells and weakly expressed by Müller cells. GAT-2 immunostaining was observed in the retina pigment epithelium and the nonpigmented ciliary epithelium. GAT-3 immunoreactivity was distributed to the OFL, to all laminae of the IPL, GCL and INL, and to processes in the ONL that ended at the outer limiting membrane. Small GAT-3-immunoreactive cell bodies were in the proximal INL and GCL. GAT-3 is therefore strongly expressed by Müller cells, and by some amacrine and displaced amacrine cells. Together, these observations demonstrate a heterologous distribution of GATs in the retina. These transporters are likely to take up GABA from, and perhaps release GABA into, the synaptic cleft and extracellular space. This suggests that GATs regulate GABA levels in these areas and thus influence synaptic neurotransmission.


Subject(s)
Eye Proteins/analysis , Membrane Proteins/analysis , Membrane Transport Proteins , Nerve Tissue Proteins/analysis , Organic Anion Transporters , Retina/chemistry , gamma-Aminobutyric Acid/metabolism , Amino Acid Sequence , Animals , Carrier Proteins/analysis , Epithelium/chemistry , Female , GABA Plasma Membrane Transport Proteins , Immunohistochemistry , Male , Molecular Sequence Data , Pigment Epithelium of Eye/chemistry , Rats , Rats, Sprague-Dawley , Sequence Homology, Amino Acid
12.
Proc Natl Acad Sci U S A ; 93(22): 12564-9, 1996 Oct 29.
Article in English | MEDLINE | ID: mdl-8901622

ABSTRACT

trkB is the high-affinity receptor for brain-derived neurotrophic factor (BDNF), a trophic molecule with demonstrated effects on the survival and differentiation of a wide variety of neuronal populations. In the mammalian retina, trkB is localized to both ganglion cells and numerous cells in the inner nuclear layer. Much information on the role of BDNF in neuronal development has been derived from the study of trkB- and BDNF-deficient mutant mice. This includes an attenuation of the numbers of cortical neurons immunopositive for the calcium-binding proteins, parvalbumin, and calbindin. Unfortunately, these mutant animals typically fail to survive for > 24-48 hr after birth. Since most retinal neuronal differentiation occurs postnatally, we have devised an alternative scheme to suppress the expression of trkB in the retina to examine the role of BDNF on the postnatal development of neurons of the inner retina. Neonatal rats were treated with intraocular injection of an antisense oligonucleotide (1-2 microliters of 10-100 microM solution) targeted to the trkB mRNA. Immunohistochemistry with a polyclonal antibody to trkB showed that the expression of trkB in retinal neurons was suppressed 48-72 hr following a single injection. Northern blot analysis demonstrated that antisense treatment had no effect on the level of trkB mRNA, even after multiple injections. This suggests an effect of trkB antisense treatment on protein translation, but not on RNA transcription. No alterations were observed in the thickness of retinal cellular or plexiform layers, suggesting that BDNF is not the sole survival factor for these neurons. There were, however, alterations in the patterns of immunostaining for parvalbumin, a marker for the narrow-field, bistratified AII amacrine cell-a central element of the rod (scotopic) pathway. This was evidenced by a decrease in both the number of immunostained somata (> 50%) and in the intensity of immunolabeling. However, the immunostaining pattern of calbindin was not affected. These studies suggest that the ligands for trkB have specific effects on the neurochemical phenotypic expression of inner retinal neurons and in the development of a well-defined retinal circuit.


Subject(s)
Brain-Derived Neurotrophic Factor/physiology , Neurons/drug effects , Oligonucleotides, Antisense/pharmacology , Receptor Protein-Tyrosine Kinases/biosynthesis , Receptors, Nerve Growth Factor/biosynthesis , Retina/growth & development , Animals , Blotting, Northern , Calbindins , Electrophoresis, Agar Gel , Mice , Microscopy, Fluorescence , Nerve Tissue Proteins/analysis , Neurons/metabolism , Parvalbumins/analysis , Phenotype , Rats , Receptor, trkB , S100 Calcium Binding Protein G/analysis
13.
J Comp Neurol ; 368(4): 597-607, 1996 May 13.
Article in English | MEDLINE | ID: mdl-8744446

ABSTRACT

The members of the trk family of tyrosine receptor kinases, trkA, trkB, and trkC, are the functional receptors for neurotrophins, a family of related neurotrophic factors. In this study, we investigated 1) the distribution of neurotrophin receptors in the developing and adult rat digestive tract with a pan-trk antibody that recognizes all known trks and 2) the cellular localization of trk-encoding mRNAs in the adult gut with single-stranded RNA probes specific for trkA, trkB, and trkC. In the developing myenteric plexus, trk immunoreactivity was present at embryonic day (ED) 14. Cells and fibers immunoreactive for trk could be visualized in the myenteric plexus at ED 16. At this age, dense staining was found in thick bundles of fibers in proximity to the myenteric plexus in the longitudinal muscle and in association with blood vessels in the mesentery. At ED 18, trk immunoreactivity was also seen in thin processes running from the myenteric plexus into the circular muscle, and in fibers and cells in intrapancreatic ganglia. By ED 20, immunoreactive staining was quite dense in both the myenteric and submucosal plexuses. At birth, virtually all enteric ganglia displayed strong trk immunoreactivity; the intensity of the staining at this age made it difficult to discern individual cells. During postnatal development, there was a decrease in cell body staining and an increase in the density of trk-containing fibers that became widely distributed to the gut wall and pancreas. The adult pattern of trk immunoreactivity was established between postnatal days 5 and 10. In adults, trk immunoreactivity was found in numerous enteric and intrapancreatic ganglion cells and in dense networks of fibers innervating all the layers of the gut, the pancreas, and vasculature. The trkC mRNA was expressed in adult enteric ganglion cells of both the myenteric and submucous plexus. By contrast, the trkA and trkB mRNAs could not be detected in enteric ganglia. All three trk mRNAs were expressed in dorsal root ganglia, which were used as positive controls. The density and wide distribution of trk immunoreactivity together with its persistence in adulthood support the concept that neurotrophins play a broad role in the digestive system from development through adult life, perhaps being involved in differentiation, phenotypic expression, and tissue maintenance. The presence of trkC mRNA in enteric neurons along with recent evidence that neurotrophin-3 plays a role in the development of the enteric nervous system suggest that trkC and neurotrophin-3 are a major neurotrophin system in the gastrointestinal tract.


Subject(s)
Enteric Nervous System/chemistry , Proto-Oncogene Proteins/genetics , RNA, Messenger/analysis , Receptor Protein-Tyrosine Kinases/analysis , Receptor Protein-Tyrosine Kinases/genetics , Receptors, Nerve Growth Factor/analysis , Receptors, Nerve Growth Factor/genetics , Animals , Embryonic and Fetal Development/physiology , Enteric Nervous System/anatomy & histology , Enteric Nervous System/growth & development , Histocytochemistry , Immunohistochemistry , In Situ Hybridization , Rats , Rats, Sprague-Dawley , Receptor, trkA
14.
J Comp Neurol ; 365(3): 491-503, 1996 Feb 12.
Article in English | MEDLINE | ID: mdl-8822184

ABSTRACT

Somatostatin (SRIF) is a neuroactive peptide that is distributed throughout the nervous system, including the retina. This peptide has been localized to populations of amacrine cells in a variety of vertebrate species. In the rabbit retina, SRIF immunoreactivity is present in a sparse population of medium to large neurons (13.72 microns in diameter, or 147.84 mu 2) in the ganglion cell layer and in a small number of neurons in the inner nuclear layer. These cells display a preferential distribution to the inferior retina, with the highest density near the ventral and ventrolateral retinal margins (11.33 cells/mm2). SRIF-immunoreactive cells have two to five primary processes that arborize in the proximal inner plexiform layer (IPL). These give rise to a plexus of finer processes in the distal IPL. Occasional immunoreactive processes are also present in the outer plexiform layer. In the IPL, these laminar networks are present in all retinal regions. In addition, SRIF-immunoreactive cells often have a fine-caliber axonlike process that eminates from the soma or perisomal region. These processes travel for great distances across the retina in either the nerve fiber layer or in the distal IPL but are never seen to enter the optic nerve head. In addition, the number of SRIF-immunoreactive somata remains unchanged following transection of the optic nerve. Taken together, these data indicate that SRIF-immunoreactive neurons of the rabbit retina are displaced amacrine cells. Furthermore, the sparse distribution of SRIF-immunoreactive somata, the wide-ranging, asymmetric arborization of their cellular processes, and previous pharmacological studies suggest that these neurons mediate a broad modulatory role in retinal function.


Subject(s)
Neurons/chemistry , Rabbits/metabolism , Retina/chemistry , Somatostatin/analysis , Animals , Axons/chemistry , Cell Count , Cell Size , Female , Immunohistochemistry , Male , Microscopy, Electron , Optic Nerve/physiology , Rabbits/anatomy & histology , Retina/cytology
15.
J Comp Neurol ; 356(1): 132-42, 1995 May 22.
Article in English | MEDLINE | ID: mdl-7629307

ABSTRACT

Parvalbumin (PV) is a calcium-binding protein localized to selected neurons in the nervous system, including the retina. This investigation evaluated the distribution of PV immunoreactivity in the rabbit retina using immunohistochemistry with a monoclonal antibody directed to carp PV. In the inner nuclear layer (INL), PV immunoreactivity was present in horizontal and amacrine cells. In the ganglion cell layer, PV immunostaining was confined to somata that are likely to be both displaced amacrine cells and ganglion cells. PV-immunoreactive (IR) amacrine cells were positioned in the proximal INL adjacent to the inner plexiform layer (IPL). These cells usually gave rise to a single primary process, which arborized into two distinct bands in the IPL. In sublamina a, the processes were thin and had large, irregular endings. In sublamina b, multiple processes branched from the primary process and were characterized by varicosities and spines. PV-IR amacrine cell bodies measured from 8 to 10 microns in diameter. Their density was highest in the visual streak and lowest in the periphery of the superior retina. The average number of PV-IR amacrine cells was 464,045 cells per retina (N = 3), and the average regularity index of the PV-IR cell mosaic was 3.23. PV-IR amacrine cells were further characterized by double-label immunofluorescence experiments using antibodies to PV and tyrosine hydroxylase (TH). Varicose TH-IR processes were in close apposition to many PV-IR amacrine cells and often formed "ring structures" around them. Together, these morphological, quantitative, and histochemical observations indicate that PV immunoreactivity in the INL is localized predominantly to AII amacrine cells, and therefore it is a valuable marker for the identification of this cell type.


Subject(s)
Parvalbumins/metabolism , Retinal Ganglion Cells/cytology , Retinal Rod Photoreceptor Cells/cytology , Animals , Antibodies, Monoclonal , Ganglia/cytology , Ganglia/immunology , Ganglia/metabolism , Immunohistochemistry , Parvalbumins/immunology , Rabbits , Retinal Ganglion Cells/immunology , Retinal Ganglion Cells/metabolism , Retinal Rod Photoreceptor Cells/immunology , Retinal Rod Photoreceptor Cells/metabolism , Visual Pathways/cytology , Visual Pathways/metabolism
16.
Vis Neurosci ; 12(2): 215-22, 1995.
Article in English | MEDLINE | ID: mdl-7786843

ABSTRACT

The neurotrophins, including nerve growth factor (NGF), brain-derived neurotrophic factor (BDNF), neurotrophin-3 (NT-3) and NT-4/5 are important in a variety of developmental processes in the peripheral and central nervous systems. These molecules bind to a low-affinity receptor and to distinct high-affinity receptors. The high-affinity receptor for NGF is the proto-oncogene product, p140trkA(trkA). Isoforms of p140trkA, p145trkB(trkB), and p140trkC(trkC), are the primary high-affinity receptors for BDNF and NT-3, respectively. We evaluated the developmental regulation of the high-affinity neurotrophin receptors in the rat retina using polyclonal antibodies directed to a highly conserved region of the C-terminus of the p140trkA isoforms (pantrk) and antibodies directed to unique amino-acid sequences of p140trkA, p145trkB, and p140trkC. Immunoreactivities for trkA and trkB, as well as pantrk, were detected in the developing retina and showed similar distributions. At similar antibody concentrations, trkC immunoreactivity was not detected. In the embryo, immunoreactivties were present in cells located throughout the neuroblastic retina, especially in the inner retinal layers, and in fibers in the nerve fiber layer and optic nerve. In the newborn retina, immunoreactivities for these two receptor isoforms were localized to numerous somata in the inner nuclear layer (INL), as well as to cells in the ganglion cell layer (GCL) and axons in the nerve fiber layer and optic nerve. A similar pattern of immunostaining persisted throughout the first postnatal week. By postnatal day-10, immunostaining was confined to large-diameter cells in the GCL, both heavily stained and lightly stained cells in the INL and a plexus of processes in the inner plexiform layer (IPL).(ABSTRACT TRUNCATED AT 250 WORDS)


Subject(s)
Proto-Oncogene Proteins/biosynthesis , Receptor Protein-Tyrosine Kinases/biosynthesis , Receptors, Nerve Growth Factor/biosynthesis , Retina/metabolism , Animals , Animals, Newborn , Axons/chemistry , Female , Fetus , Fluorescent Antibody Technique , Immunoenzyme Techniques , Nerve Fibers/chemistry , Neurons/chemistry , Optic Nerve/chemistry , Optic Nerve/embryology , Optic Nerve/growth & development , Optic Nerve/metabolism , Pregnancy , Proto-Oncogene Proteins/analysis , Proto-Oncogene Proteins/genetics , Proto-Oncogenes , Rats , Rats, Sprague-Dawley , Receptor Protein-Tyrosine Kinases/analysis , Receptor Protein-Tyrosine Kinases/genetics , Receptor, trkA , Receptors, Nerve Growth Factor/analysis , Receptors, Nerve Growth Factor/genetics , Retina/chemistry , Retina/embryology , Retina/growth & development
17.
Brain Res ; 345(1): 186-91, 1985 Oct 14.
Article in English | MEDLINE | ID: mdl-3933762

ABSTRACT

The functional role of dopamine in frog retina was examined in a combined neurochemical, immunohistochemical and electrophysiological study. Dopamine and serotonin are the primary monoamines present in the retina and they are localized to amacrine cells which have distinct morphologies. Intravitreal injection of 6-hydroxydopamine was found to produce a selective depletion of retinal dopamine content and elimination of tyrosine hydroxylase-like immunoreactivity. Electroretinograms from 6-hydroxydopamine-treated retinas demonstrated enhanced oscillatory potentials and a lengthening of the b-wave implicit time compared to vehicle control retinas; both of these changes in the electroretinogram were reversed by the dopamine agonist apomorphine. These observations support earlier suggestions that dopamine-containing amacrine cells are part of a retinal feedback system that generates oscillatory potentials and plays a role in light adaptation.


Subject(s)
Dopamine/physiology , Retina/physiology , Animals , Apomorphine/pharmacology , Electroretinography , Feedback , Hydroxydopamines/pharmacology , Oxidopamine , Rana pipiens
SELECTION OF CITATIONS
SEARCH DETAIL
...