Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
Genes Cells ; 25(6): 402-412, 2020 Jun.
Article in English | MEDLINE | ID: mdl-32215989

ABSTRACT

Di- and trimethylation of lysine 27 on histone 3 (H3K27me2/3) is a critical gene repression mechanism. We previously showed that down-regulation of the H3K27 demethylase, Jumonji domain-containing protein 3 (JMJD3), resulted in a reduced number of protein kinase C (PKC)α-positive rod ON-bipolar cells. In this work, we focused on the role of another H3K27 demethylase, ubiquitously transcribed tetratricopeptide repeat X chromosome (UTX), in retinal development. UTX was expressed in the retinal progenitor cells of the embryonic mouse retina and was observed in the inner nuclear layer during late retinal development and in the mature retina. The short hairpin RNA-mediated knockdown of Utx in a mouse retinal explant led to a reduced number of PKCα-positive rod ON-bipolar cells. However, other retinal subtypes were unaffected by this knockdown. Using a retina-specific knockout of Utx in mice, the in vivo effects of UTX down-regulation were examined. Again, the number of PKCα-positive rod ON-bipolar cells was reduced, and no other apparent phenotypes, including retinal progenitor proliferation, apoptosis or differentiation, were observed. Finally, we examined retina-specific Utx and Jmjd3 double-knockout mice and found that although the number of rod ON-bipolar cells was reduced, no additional effects from the loss of Utx and Jmjd3 were observed. Taken together, our data show that UTX contributes to retinal differentiation in a lineage-specific manner.


Subject(s)
Cell Differentiation/genetics , Histone Demethylases/metabolism , Retina/metabolism , Retinal Bipolar Cells/metabolism , Animals , Apoptosis/genetics , Cell Lineage , Cell Proliferation/genetics , Gene Expression Regulation, Developmental/genetics , Gene Knockdown Techniques , Histone Demethylases/genetics , Jumonji Domain-Containing Histone Demethylases/genetics , Jumonji Domain-Containing Histone Demethylases/metabolism , Methylation , Mice , Mice, Knockout , Protein Kinase C-alpha/metabolism , RNA Interference , Retina/embryology , Retina/enzymology , Retinal Bipolar Cells/cytology , Retinal Bipolar Cells/enzymology , Retinal Rod Photoreceptor Cells/cytology , Retinal Rod Photoreceptor Cells/metabolism
2.
Histol Histopathol ; 33(10): 1059-1073, 2018 Oct.
Article in English | MEDLINE | ID: mdl-29741202

ABSTRACT

A growing number of studies have revealed the functional neuroarchitecture of the microbat retina and suggested that microbats can see using their eyes. To better understand the organization of the microbat retina, quantitative analysis of protein kinase C alpha (PKCα)- and tyrosine hydroxylase (TH)-immunoreactive (IR) cells was conducted on the greater horseshoe bat (Rhinolophus ferrumequinum) retina. As a result, PKCα immunoreactivity was observed in rod bipolar cells, consistent with previous studies on other mammalian retinas. PKCα-IR cell distribution in the inner nuclear layer showed regional differences in density, with the highest density found in the nasal retina. The average density of PKCα-IR cells was 10,487±441 cells/mm² (mean ± S.D.; n=4), with a total of 43,077±1,843 cells/retina. TH-IR cells in the Rhinolophus ferrumequinum retina could be classified into four types based on soma location and ramification in the inner plexiform layer: conventional amacrine, displaced amacrine, interplexiform, and intercalated cells. The majority of TH-IR cells were conventional amacrine cells. TH-IR cells were nonrandomly distributed at low density over the retina. The average density was 29.7±3.1 cells/mm² (mean ± S.D.; n=3), with a total of 124.0±11.3 cells/retina. TH-IR processes showed varicosities and formed ring-like structures encircling AII amacrine cells. Our study provides the foundation for understanding the neurochemical architecture of the microbat retina and supports the notion that the eyes do play a role in the visual system of microbats.


Subject(s)
Chiroptera/metabolism , Fluorescent Antibody Technique , Protein Kinase C-alpha/metabolism , Retinal Neurons/enzymology , Tyrosine 3-Monooxygenase/metabolism , Amacrine Cells/enzymology , Animals , Biomarkers/metabolism , Chiroptera/classification , Female , Male , Retinal Bipolar Cells/enzymology
3.
Invest Ophthalmol Vis Sci ; 56(8): 4961-74, 2015 Jul.
Article in English | MEDLINE | ID: mdl-26230760

ABSTRACT

PURPOSE: Protein kinase C α (PKCα) is abundantly expressed in rod bipolar cells (RBCs) in the retina, yet the physiological function of PKCα in these cells is not well understood. To elucidate the role of PKCα in visual processing in the eye, we examined the effect of genetic deletion of PKCα on the ERG and on RBC light responses in the mouse. METHODS: Immunofluorescent labeling was performed on wild-type (WT), TRPM1 knockout, and PKCα knockout (PKC-KO) retina. Scotopic and photopic ERGs were recorded from WT and PKC-KO mice. Light responses of RBCs were measured using whole-cell recordings in retinal slices from WT and PKC-KO mice. RESULTS: Protein kinase C alpha expression in RBCs is correlated with the activity state of the cell. Rod bipolar cells dendrites are a major site of PKCα phosphorylation. Electroretinogram recordings indicated that loss of PKCα affects the scotopic b-wave, including a larger peak amplitude, longer implicit time, and broader width of the b-wave. There were no differences in the ERG a- or c-wave between PKCα KO and WT mice, indicating no measurable effect of PKCα in photoreceptors or the RPE. The photopic ERG was unaffected consistent with the lack of detectable PKCα in cone bipolar cells. Whole-cell recordings from RBCs in PKC-KO retinal slices revealed that, compared with WT, RBC light responses in the PKC-KO retina are delayed and of longer duration. CONCLUSIONS: Protein kinase C alpha plays an important modulatory role in RBCs, regulating both the peak amplitude and temporal properties of the RBC light response in the rod visual pathway.


Subject(s)
DNA/genetics , Gene Expression Regulation , Protein Kinase C-alpha/genetics , Retinal Bipolar Cells/enzymology , Retinal Diseases/genetics , Retinal Rod Photoreceptor Cells/enzymology , Visual Pathways/enzymology , Animals , Blotting, Western , Disease Models, Animal , Electroretinography , Genetic Therapy/methods , Immunohistochemistry , Mice , Mice, Knockout , Patch-Clamp Techniques , Protein Kinase C-alpha/biosynthesis , Retinal Bipolar Cells/pathology , Retinal Diseases/enzymology , Retinal Diseases/physiopathology , Retinal Rod Photoreceptor Cells/pathology , Visual Pathways/physiopathology
4.
Exp Eye Res ; 115: 230-8, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23856406

ABSTRACT

The N-methyl-d-aspartate (NMDA) receptor-induced apoptosis is implicated in the pathological mechanisms of neural tissues, increasing the release of reactive oxygen species (ROS), resulting in a type of apoptotic cell death called excitotoxicity. Although intrinsic mechanisms to remove ROS, such as antioxidant enzymes, are provided by the tissue, the association between NMDA-induced excitotoxicity and antioxidative enzymes is not well understood. In this study, we focused on superoxide dismutase 1 (SOD1), an antioxidant enzyme, and investigated the role of SOD1 in the NMDA-induced neuronal cell death in the retina. NMDA was intravitreally injected into wild-type (WT) and SOD1 total knock-out (SOD1-deficient) mice. The number of TUNEL-positive cells in the retinal ganglion cell layer (GCL) and inner nuclear layer (INL) counted in the retinal sections and flatmount retinas were significantly higher in the SOD1-deficient mice than the WT mice after NMDA injection. Visual function assessed by dark-adapted electroretinogram (ERG) showed that the amplitudes of a-wave, b-wave, and oscillatory potential 2 were significantly reduced in the NMDA-injected SOD1-deficient mice. The level of ROS in the GCL and INL, measured using dihydroethidium, and the number of positive cells for γ-H2AX, a marker for DNA double strand breaks, and 8-OHdG, a marker for DNA oxidation, in the GCL were significantly increased in the SOD1-deficient mice after NMDA injection. We also measured mRNA and protein levels of SOD1 and SOD2 in the retina of WT mice, to find that mRNA and protein levels of SOD1, but not SOD2, were significantly reduced after NMDA injection. SOD1 deficiency exacerbated NMDA-induced damage to the inner retinal neurons, and NMDA reduced SOD1 levels in the retina of WT mice. Therefore, SOD1 protected retinal neurons against NMDA-induced retinal neurotoxicity, and NMDA-induced SOD1 reduction may be involved in neuronal vulnerability to excitotoxicity.


Subject(s)
Amacrine Cells/enzymology , Excitatory Amino Acid Agonists/toxicity , N-Methylaspartate/toxicity , Retinal Bipolar Cells/enzymology , Retinal Ganglion Cells/enzymology , Retinal Horizontal Cells/enzymology , Superoxide Dismutase/physiology , Amacrine Cells/drug effects , Amacrine Cells/pathology , Animals , Apoptosis/drug effects , Aspartic Acid/metabolism , Dark Adaptation , Electroretinography , Fluorescent Antibody Technique, Indirect , Immunoblotting , In Situ Nick-End Labeling , Intravitreal Injections , Mice , Mice, Inbred C57BL , Mice, Knockout , RNA, Messenger/metabolism , Reactive Oxygen Species/metabolism , Real-Time Polymerase Chain Reaction , Retinal Bipolar Cells/drug effects , Retinal Bipolar Cells/pathology , Retinal Ganglion Cells/drug effects , Retinal Ganglion Cells/pathology , Retinal Horizontal Cells/drug effects , Retinal Horizontal Cells/pathology , Superoxide Dismutase-1
5.
Cell Physiol Biochem ; 29(3-4): 417-30, 2012.
Article in English | MEDLINE | ID: mdl-22508049

ABSTRACT

Rod outer segment membrane guanylate cyclase (ROS-GC1) is a bimodal Ca(2+) signal transduction switch. Lowering [Ca(2+)](i) from 200 to 20 nM progressively turns it "ON" as does raising [Ca(2+)](i) from 500 to 5000 nM. The mode operating at lower [Ca(2+)](i) plays a vital role in phototransduction in both rods and cones. The physiological function of the mode operating at elevated [Ca(2+)](i) is not known. Through comprehensive studies on mice involving gene deletions, biochemistry, immunohistochemistry, electroretinograms and single cell recordings, the present study demonstrates that the Ca(2+)-sensor S100B coexists with and is physiologically linked to ROS-GC1 in cones but not in rods. It up-regulates ROS-GC1 activity with a K(1/2) for Ca(2+) greater than 500 nM and modulates the transmission of neural signals to cone ON-bipolar cells. Furthermore, a possibility is raised that under pathological conditions where [Ca(2+)](i) levels rise to and perhaps even enter the micromolar range, the S100B signaling switch will be turned "ON" causing an explosive production of CNG channel opening and further rise in [Ca(2+)](i) in cone outer segments. The findings define a new cone-specific Ca(2+)-dependent feature of photoreceptors and expand our understanding of the operational principles of phototransduction machinery.


Subject(s)
Calcium/metabolism , Guanylate Cyclase/metabolism , Nerve Growth Factors/metabolism , Receptors, Cell Surface/metabolism , Retinal Cone Photoreceptor Cells/enzymology , Rod Cell Outer Segment/enzymology , S100 Proteins/metabolism , Animals , Cyclic GMP/genetics , Cyclic GMP/metabolism , Enzyme Activation , Guanylate Cyclase/genetics , Immunohistochemistry , Light Signal Transduction , Mice , Mice, Knockout , Nerve Growth Factors/genetics , Receptors, Cell Surface/genetics , Retinal Bipolar Cells/enzymology , Retinal Bipolar Cells/metabolism , Retinal Bipolar Cells/physiology , Retinal Cone Photoreceptor Cells/metabolism , Retinal Cone Photoreceptor Cells/physiology , Rod Cell Outer Segment/metabolism , Rod Cell Outer Segment/physiology , S100 Calcium Binding Protein beta Subunit , S100 Proteins/genetics , Synaptic Membranes/enzymology , Synaptic Membranes/metabolism , Synaptic Membranes/physiology
6.
J Mol Histol ; 42(5): 483-9, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21822613

ABSTRACT

HPC-1/syntaxin 1A (STX1A) is abundantly expressed in neurons. STX1A is believed to regulate exocytosis in synaptic vesicles. In our recent studies, STX1A knockout (KO) mice showed normal development, and basal synaptic transmission in cultured hippocampal neurons appeared to be normal. However, behavioral abnormalities were observed in STX1A KO mice. In the normal rodent retina, the STX1A protein is expressed in two synaptic layers (plexiform layers). Here, to evaluate the effects of the loss of STX1A on retinal structure, we examined the retinal layer structure in STX1A KO mice using hematoxylin staining and immunostaining. We found that the general layer structures in the retina were preserved in all genotypes. However, the outer plexiform layer (OPL) was significantly thicker in KO and heterozygous mutant (HT) mice compared with that in wild-type (WT) mice. No significant differences were observed in the thicknesses of the other layers. Immunostaining for protein kinase C α showed that the alignment of rod bipolar cell bodies in the inner nuclear layer (INL) was slightly disrupted in HT and KO retinas. Furthermore, the dendrites of these cells in the OPL of KO mice were sparse, compared to those in WT mice. Our results show that STX1A KO mice have increased thickness of the OPL and changes in the morphology of the INL that may contribute to the change in OPL thickness. We suggest that STX1A may play a role in the structural formation of the INL and OPL in the retina.


Subject(s)
Retina/cytology , Retina/metabolism , Syntaxin 1/deficiency , Animals , Genotype , Immunohistochemistry , Mice , Mice, Knockout , Protein Kinase C/metabolism , Retinal Bipolar Cells/cytology , Retinal Bipolar Cells/enzymology , Retinal Neurons/cytology , Retinal Neurons/metabolism , Staining and Labeling , Syntaxin 1/metabolism
7.
Invest Ophthalmol Vis Sci ; 51(11): 6051-8, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20554612

ABSTRACT

PURPOSE: Protein kinase (PKC)-α is abundant in retinal bipolar cells. This study was performed to explore its role in visual processing. METHODS: PKCα-knockout (Prkca(-/-)) mice and control animals were examined by using electroretinography (ERG), light microscopy, and immunocytochemistry. RESULTS: The Prkca(-/-) mice showed no signs of retinal degeneration up to 12 months of age, but ERG measurements indicated a decelerated increase in the ascending limb of the scotopic (rod-sensitive) b-wave as well as a delayed return to baseline. These results suggest that PKCα is an important modulator that affects bipolar cell signal transduction and termination. Confocal microscopy of retinal sections showed that PKCα co-localized with calbindin, which indicates a PKCα localization in close proximity to the horizontal cell terminals. In addition, the implicit time of the ERG c-wave originating from the retinal pigment epithelium (RPE) and the recovery of photoreceptors from bleaching conditions were substantially faster in the knockout mice than in the wild-type control animals. CONCLUSIONS: These results suggest that PKCα is a modulator of rod-bipolar cell function by accelerating glutamate-driven signal transduction and termination. This modulation is of importance in the switch between scotopic and photopic vision. Furthermore, PKCα seems to play a role in RPE function.


Subject(s)
Protein Kinase C-alpha/physiology , Retinal Bipolar Cells/enzymology , Retinal Rod Photoreceptor Cells/enzymology , Vision, Ocular/physiology , Animals , Blotting, Southern , Dark Adaptation , Electroretinography , Female , Genotype , Immunohistochemistry , Light , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Microscopy , Polymerase Chain Reaction , Retinal Bipolar Cells/radiation effects , Retinal Rod Photoreceptor Cells/radiation effects
8.
Vis Neurosci ; 25(2): 167-77, 2008.
Article in English | MEDLINE | ID: mdl-18442439

ABSTRACT

This study investigated the anatomical consequences of a photoreceptor toxin, iodoacetic acid (IAA), in the rabbit retina. Retinae were examined 2 weeks, 1, 3, and 6 months after systemic IAA injection. The retinae were processed using standard histological methods to assess the gross morphology and topographical distribution of damage, and by immunohistochemistry to examine specific cell populations in the retina. Degeneration was restricted to the photoreceptors and was most common in the ventral retina and visual streak. In damaged regions, the outer nuclear layer was reduced in thickness or eliminated entirely, with a concomitant loss of immunoreactivity for rhodopsin. However, the magnitude of the effect varied between animals with the same IAA dose and survival time, suggesting individual differences in the bioavailability of the toxin. In all eyes, the inner retina remained intact, as judged by the thickness of the inner nuclear layer, and by the pattern of immunoreactivity for protein kinase C-alpha (rod bipolar cells) and calbindin D-28 (horizontal cells). Müller cell stalks became immunoreactive for glial fibrillary acidic protein (GFAP) even in IAA-treated retinae that had no signs of cell loss, indicating a response of the retina to the toxin. However, no marked hypertrophy or proliferation of Müller cells was observed with either GFAP or vimentin immunohistochemistry. Thus the selective, long lasting damage to the photoreceptors produced by this toxin did not lead to a reorganization of the surviving cells, at least with survival as long as 6 months, in contrast to the remodeling of the inner retina that is observed in inherited retinal degenerations such as retinitis pigmentosa and retinal injuries such as retinal detachment.


Subject(s)
Iodoacetic Acid/poisoning , Photoreceptor Cells, Vertebrate/drug effects , Retina/drug effects , Animals , Calbindins , Cell Nucleus/pathology , Cell Survival/drug effects , Glial Fibrillary Acidic Protein/metabolism , Immunohistochemistry , Injections, Intravenous , Iodoacetic Acid/administration & dosage , Nerve Degeneration/chemically induced , Nerve Degeneration/pathology , Nerve Degeneration/physiopathology , Protein Kinase C-alpha/metabolism , Rabbits , Retina/metabolism , Retina/pathology , Retina/physiopathology , Retinal Bipolar Cells/enzymology , Retinal Horizontal Cells/metabolism , S100 Calcium Binding Protein G/metabolism , Time Factors
9.
Cell Physiol Biochem ; 21(1-3): 161-72, 2008.
Article in English | MEDLINE | ID: mdl-18209483

ABSTRACT

The activity of cells critically depends on the control of their cytosolic free calcium ion (Ca(2+)) concentration. The objective of the present study was to identify mechanisms of action underlying the control of the gain of intracellular Ca(2+) release by circulating gonadal steroid hormones. Acute stimulation of isolated neurons with progesterone led to IP(3)R-mediated Ca(2+) transients that depend on the activation of the PI3 kinase/Akt/PKB signaling pathway. These results were confirmed at the molecular level and phosphorylation of IP(3)R type 1 by Akt/PKB was identified as the mechanism of action. Hence, it is likely that circulating gonadal steroid hormones control neuronal activity including phosporylation status through receptor- and kinase-mediated signaling. With a direct control of the gain of the Ca(2+) second messenger system as a signaling gatekeeper for neuronal activity the present study identifies a novel pathway for interaction of the endocrine and central nervous system.


Subject(s)
Calcium Signaling/drug effects , Inositol 1,4,5-Trisphosphate/metabolism , Progesterone/pharmacology , Proto-Oncogene Proteins c-akt/metabolism , Animals , Inositol 1,4,5-Trisphosphate Receptors/chemistry , Ion Channel Gating/drug effects , Mice , Phosphorylation/drug effects , Receptors, Progesterone/metabolism , Retinal Bipolar Cells/cytology , Retinal Bipolar Cells/drug effects , Retinal Bipolar Cells/enzymology
10.
Proc Natl Acad Sci U S A ; 104(20): 8514-9, 2007 May 15.
Article in English | MEDLINE | ID: mdl-17485676

ABSTRACT

Members of the carbonic anhydrase (CA) family play an important role in the regulation of pH, CO(2), ion, and water transport. CA IV and CA XIV are membrane-bound isozymes expressed in the eye. CA IV immunostaining is limited to the choriocapillaris overlying the retina, whereas CA XIV is expressed within the retina in Müller glial cells and retinal pigment epithelium. Here, we have characterized the physiological and morphological phenotype of the CA IV-null, CA XIV-null, and CA IV/CA XIV-double-null mouse retinas. Flash electroretinograms performed at 2, 7, and 10 months of age showed that the rod/cone a-wave, b-wave, and cone b-wave were significantly reduced (26-45%) in the CA XIV-null mice compared with wild-type littermates. Reductions in the dark-adapted response were not progressive between 2 and 10 months, and no differences in retinal morphology were observed between wild-type and CA XIV-null mice. Müller cells and rod bipolar cells had a normal appearance. Retinas of CA IV-null mice showed no functional or morphological differences compared with normal littermates. However, CA IV/CA XIV double mutants showed a greater deficit in light response than the CA XIV-null retina. Our results indicate that CA XIV, which regulates extracellular pH and pCO(2), plays an important part in producing a normal retinal light response. A larger functional deficit in the CA IV/CA XIV double mutants suggests that CA IV can also contribute to pH regulation, at least in the absence of CA XIV.


Subject(s)
Carbonic Anhydrases/deficiency , Light , Retina/physiopathology , Retina/radiation effects , Animals , Carbonic Anhydrase IV/deficiency , Electroretinography , Genotype , Mice , Mice, Knockout , Photic Stimulation , Retina/cytology , Retina/enzymology , Retinal Bipolar Cells/cytology , Retinal Bipolar Cells/enzymology , Retinal Bipolar Cells/radiation effects
11.
Mol Vis ; 12: 389-98, 2006 Apr 18.
Article in English | MEDLINE | ID: mdl-16636658

ABSTRACT

PURPOSE: Gene inactivation with homologous recombination in mice is a widely used tool to study gene function. However, many proteins play essential roles in a number of tissues and germline gene inactivation often results in embryonic lethality. To overcome this limitation and to dissect the functions of essential genes beyond embryonic development, we generated mouse rod opsin promoter-controlled cre transgenic mice with a goal of obtaining transgenic lines with a range of Cre activity in rod photoreceptors. METHODS: Transgenic mice expressing Cre recombinase directed by a long or short mouse opsin promoter were generated. Candidate Cre-expressing lines were identified with RT-PCR and Western blot analysis. Potentially useful Cre-expressing lines were characterized further with immunohistochemistry, PCR, and functional analysis using a Cre-activatable lacZ reporter mouse strain (R26R) to determine temporal and spatial patterns of Cre expression. Retinal function and morphology in these mouse lines were analyzed with electroretinography (ERG) and light microscopy of hematoxylin and eosin stained retinal sections. RESULTS: Transgenic mice expressing Cre in rod photoreceptors were generated. Characterization of candidate photoreceptor-specific Cre mice using immunohistochemistry and functional assays demonstrated that an efficient Cre-mediated recombination occurred in rod photoreceptor cells in one mouse line and a mosaic Cre-mediated recombination occurred in rod photoreceptors and rod bipolar cells in another mouse line. Further analysis of these mice with ERG and morphological examination suggested that the retinas of eight-month-old adults were normal. CONCLUSIONS: We have generated transgenic mice expressing Cre recombinase in rod photoreceptors. One transgenic mouse line was capable of carrying out efficient Cre-mediated recombination in rod photoreceptors. Another transgenic mouse line was capable of carrying out mosaic Cre-mediated recombination in rod photoreceptors and bipolar cells across the whole retina. These mice will be useful tools for Cre/lox-based gene activation and inactivation, as well as genetic mosaics, in rod photoreceptors and rod bipolar cells.


Subject(s)
Integrases/metabolism , Mice, Transgenic/genetics , Promoter Regions, Genetic , Retinal Rod Photoreceptor Cells/enzymology , Rod Opsins/genetics , Animals , Gene Expression , Integrases/genetics , Mice , Mosaicism , Recombination, Genetic , Retina/cytology , Retina/enzymology , Retinal Bipolar Cells/enzymology , Tissue Distribution
12.
Neurobiol Dis ; 21(1): 43-9, 2006 Jan.
Article in English | MEDLINE | ID: mdl-16023354

ABSTRACT

Nitric oxide (NO) can play either a neuroprotective or a neurotoxic role in diverse neurodegenerative conditions. This study investigated the differential expression of neuronal nitric oxide synthase (nNOS) in the streptozotocin-induced diabetic rat retina to clarify the involvement of NO produced from neurons in the early pathogenesis of diabetic retinopathy. A decrease in thickness of the outer retina was evident at 12 and 24 weeks after onset of diabetes. nNOS was immunolocalized in two subtypes of amacrine cells, displaced amacrine cells and in some bipolar cells in the normal retinas. The densities of each type of nNOS-expressing neuron showed no significant differences in the diabetic retinas with the exception of the bipolar cells. The numbers of nNOS bipolar cells at 12 weeks of diabetes increased threefold, showing dendritic polarity of nNOS expression. Protein levels of nNOS increased throughout the diabetic retinas reaching a peak value at 24 weeks of diabetes. Thus, diabetes up-regulates the expression of nNOS in bipolar cells, and NO from these cells may aggravate the degeneration of the outer retina in the diabetic retinas.


Subject(s)
Amacrine Cells/enzymology , Diabetic Retinopathy/metabolism , Diabetic Retinopathy/pathology , Nitric Oxide Synthase Type I/metabolism , Retinal Bipolar Cells/enzymology , Amacrine Cells/pathology , Animals , Blotting, Western , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Experimental/pathology , Male , Nitric Oxide/metabolism , Rats , Rats, Sprague-Dawley , Retinal Bipolar Cells/pathology , Up-Regulation/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...