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










Publication year range
1.
PLoS One ; 16(12): e0259505, 2021.
Article in English | MEDLINE | ID: mdl-34882677

ABSTRACT

PURPOSE: The purpose of this study was to investigate neuronal and vascular functional deficits in the retina and their association in a diabetic mouse model. We measured electroretinography (ERG) responses and choroidal and retinal blood flow (ChBF, RBF) with magnetic resonance imaging (MRI) in healthy and diabetic mice under basal conditions and under hypercapnic challenge. METHODS: Ins2Akita diabetic (Diab, n = 8) and age-matched, wild-type C57BL/6J mice (Ctrl, n = 8) were studied under room air and moderate hypercapnia (5% CO2). Dark-adapted ERG a-wave, b-wave, and oscillatory potentials (OPs) were measured for a series of flashes. Regional ChBF and RBF under air and hypercapnia were measured using MRI in the same mice. RESULTS: Under room air, Diab mice had compromised ERG b-wave and OPs (e.g., b-wave amplitude was 422.2±10.7 µV in Diab vs. 600.1±13.9 µV in Ctrl, p < 0.001). Under hypercapnia, OPs and b-wave amplitudes were significantly reduced in Diab (OPs by 30.3±3.0% in Diab vs. -3.0±3.6% in Ctrl, b-wave by 17.9±1.4% in Diab vs. 1.3±0.5% in Ctrl). Both ChBF and RBF had significant differences in regional blood flow, with Diab mice having substantially lower blood flow in the nasal region (ChBF was 5.4±1.0 ml/g/min in Diab vs. 8.6±1.0 ml/g/min in Ctrl, RBF was 0.91±0.10 ml/g/min in Diab vs. 1.52±0.24 ml/g/min in Ctrl). Under hypercapnia, ChBF increased in both Ctrl and Diab without significant group difference (31±7% in Diab vs. 17±7% in Ctrl, p > 0.05), but an increase in RBF was not detected for either group. CONCLUSIONS: Inner retinal neuronal function and both retinal and choroidal blood flow were impaired in Diab mice. Hypercapnia further compromised inner retinal neuronal function in diabetes, while the blood flow response was not affected, suggesting that the diabetic retina has difficulty adapting to metabolic challenges due to factors other than impaired blood flow regulation.


Subject(s)
Choroid/blood supply , Diabetes Mellitus, Experimental/complications , Hypercapnia/diagnostic imaging , Retina/physiopathology , Animals , Choroid/diagnostic imaging , Diabetes Mellitus, Experimental/diagnostic imaging , Diabetes Mellitus, Experimental/physiopathology , Electroretinography , Hypercapnia/etiology , Insulin/genetics , Magnetic Resonance Imaging , Male , Mice , Mice, Inbred C57BL , Retina/diagnostic imaging
2.
Invest Ophthalmol Vis Sci ; 62(10): 20, 2021 08 02.
Article in English | MEDLINE | ID: mdl-34410299

ABSTRACT

Purpose: Diabetic retinopathy results in vision loss with changes to both retinal blood vessels and neural retina. Recent studies have revealed that animal models of diabetes demonstrate early loss of visual function. We explored the time course of retinal change in three different mouse models of diabetes in a longitudinal study using in vivo measures of retinal structure (optical coherence tomography [OCT]) and visual function (optomotor and pupillary responses). Methods: OCT analysis of retinal microstructure, optokinetic response as a measure of visual acuity, and pupillary response to light stimulation were compared among the db/db, Ins2Akita, and streptozotocin (STZ)-induced mouse models of diabetes at 1.5, 3, 6, and 9 months of diabetes. Results: The db/db, Ins2Akita, and STZ-induced models of diabetes all exhibited vision loss and retinal thinning as disease progressed. Both structural changes and functional measures were significantly correlated with the blood glucose levels. Despite this, vision loss and retinal thinning were not consistently correlated, except for the inner retinal layer thickness at 6 months of diabetes. Conclusions: This longitudinal study compiled structural measures and functional outcome data for type 1 and 2 diabetes mouse models commonly used for diabetes studies and demonstrated an overall decline in retinal-related health in conjunction with weight change and blood glucose alterations. The relationship between the structural change and functional outcome could be correlative but is not necessarily causative, as retinal thinning was not sufficient to explain visual acuity decline.


Subject(s)
Diabetes Mellitus, Experimental/diagnosis , Diabetic Retinopathy/pathology , Retina/pathology , Retinal Vessels/pathology , Tomography, Optical Coherence/methods , Visual Acuity/physiology , Animals , Diabetes Mellitus, Experimental/physiopathology , Diabetic Retinopathy/physiopathology , Follow-Up Studies , Male , Mice , Mice, Inbred C57BL , Retina/physiopathology , Retinal Vessels/physiopathology
3.
J Gerontol A Biol Sci Med Sci ; 71(4): 427-30, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26297941

ABSTRACT

Over the past decade, a large number of discoveries have shown that interventions (genetic, pharmacological, and nutritional) increase the lifespan of invertebrates and laboratory rodents. Therefore, the possibility of developing antiaging interventions for humans has gone from a dream to a reality. However, it has also become apparent that we need more information than just lifespan to evaluate the translational potential of any proposed antiaging intervention to humans. Information is needed on how an intervention alters the "healthspan" of an animal, that is, how the physiological functions that change with age are altered. In this report, we describe the utility and the limitations of assays in mice currently available for measuring a wide range of physiological functions that potentially impact quality of life. We encourage investigators and reviewers alike to expect at minimum an overall assessment of health in several domains across several ages before an intervention is labeled as "increasing healthspan." In addition, it is important that investigators indicate any tests in which the treated group did worse or did not differ statistically from controls because overall health is a complex phenotype, and no intervention discovered to date improves every aspect of health. Finally, we strongly recommend that functional measurements be performed in both males and females so that sex differences in the rate of functional decline in different domains are taken into consideration.


Subject(s)
Aging/physiology , Animals , Female , Humans , Longevity/physiology , Male , Mice , Models, Animal , Quality of Life , Sex Factors , Translational Research, Biomedical
4.
Dev Neurobiol ; 74(7): 692-706, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24408883

ABSTRACT

The mouse visual system is immature when the eyes open two weeks after birth. As in other mammals, some of the maturation that occurs in the subsequent weeks is known to depend on visual experience. Development of the retina, which as the first stage of vision provides the visual information to the brain, also depends on light-driven activity for proper development but has been less well studied than visual cortical development. The critical properties for retinal encoding of images include detection of contrast and responsiveness to the broad range of temporal stimulus frequencies present in natural stimuli. Here we show that contrast detection threshold and temporal frequency response characteristics of ON and OFF retinal ganglion cells (RGCs), which are poor at eye opening, subsequently undergo maturation, improving RGC performance. Further, we find that depriving mice of visual experience from before birth by rearing them in the dark causes ON and OFF RGCs to have smaller receptive field centers but does not affect their contrast detection threshold development. The modest developmental increase in temporal frequency responsiveness of RGCs in mice reared on a normal light cycle was inhibited by dark rearing only in ON but not OFF RGCs. Thus, these RGC response characteristics are in many ways unaffected by the experience-dependent changes to synaptic and spontaneous activity known to occur in the mouse retina in the two weeks after eye opening, but specific differences are apparent in the ON vs. OFF RGC populations.


Subject(s)
Contrast Sensitivity/physiology , Retina/growth & development , Retina/physiology , Retinal Ganglion Cells/physiology , Sensory Deprivation/physiology , Vision, Ocular/physiology , Action Potentials , Age Factors , Animals , Darkness , Differential Threshold/physiology , Evoked Potentials, Visual , Female , Housing, Animal , Male , Mice, Inbred C57BL , Microelectrodes , Photic Stimulation , Tissue Culture Techniques
5.
Invest Ophthalmol Vis Sci ; 53(10): 6488-94, 2012 Sep 21.
Article in English | MEDLINE | ID: mdl-22915034

ABSTRACT

PURPOSE: To investigate ocular blood flow and visual function in the Ins2(Akita) diabetic retinopathy mouse model at early and late time points after onset of hyperglycemia. METHODS: Mice heterozygous for the Ins2(Akita) mutation, which become hyperglycemic at approximately 4 weeks old, were studied at 2.5 and 7.5 months of age, with age-matched wild-type littermates used as controls. Retinal and choroidal blood flows were noninvasively imaged at 42 × 42 × 400 µm using magnetic resonance imaging. Visual function was measured using optokinetic tracking to determine spatial frequency and contrast thresholds from the same mice. RESULTS: At 2.5 months, choroidal blood flow was significantly reduced (P < 0.01) by 20% in Ins2(Akita) mice (n = 13) compared with age-matched controls (n = 16), whereas retinal blood flow and visual function were not significantly affected (P > 0.05). At 7.5 months, both choroidal and retinal blood flow were significantly reduced (P < 0.05) by 27% and 28%, respectively, in Ins2(Akita) mice (n = 11) compared with age-matched controls (n = 15). Visual functions were also significantly worse (P < 0.05) in Ins2(Akita) mice at 7.5 months, as indicated by a 19% decreased spatial frequency threshold and 135% increased contrast threshold compared with age-matched controls. The magnitudes of the blood flow and vision deficits, however, were not correlated. CONCLUSIONS: Although both choroidal and retinal blood flow and vision were altered after prolonged diabetes in the Ins2(Akita) mouse, choroidal blood flow was reduced even in young diabetic animals, suggesting ocular blood flow deficit could be an early pathological change in diabetic retinopathy.


Subject(s)
Diabetic Retinopathy/pathology , Diabetic Retinopathy/physiopathology , Regional Blood Flow/physiology , Retinal Vessels/physiopathology , Animals , Choroid/blood supply , Choroid/physiology , Contrast Sensitivity/physiology , Diabetic Retinopathy/genetics , Disease Models, Animal , Early Diagnosis , Female , Hyperglycemia/genetics , Hyperglycemia/physiopathology , Insulin/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Photic Stimulation/methods , Retina/pathology , Retina/physiopathology , Retinal Vessels/pathology , Sensory Thresholds/physiology
6.
Behav Brain Res ; 226(2): 601-5, 2012 Jan 15.
Article in English | MEDLINE | ID: mdl-21963766

ABSTRACT

Diabetic retinopathy can lead to progressive loss of vision and is a leading cause of blindness. The Ins2(Akita) mouse model of diabetes develops significant retinal and systemic pathology, but how these affect visual behavior is unknown. Here, we show that Ins2(Akita) mice have progressive, quantifiable vision deficits in an optomotor behavior. This mouse line is a promising model in which to understand the contribution of retinal neuronal injury during the chronic hyperglycemia and hypoinsulinemia of diabetes to deficits in vision.


Subject(s)
Contrast Sensitivity , Diabetes Mellitus, Experimental/psychology , Diabetic Retinopathy/psychology , Insulin/genetics , Visual Perception/genetics , Animals , Blood Glucose/metabolism , Diabetes Mellitus, Experimental/blood , Diabetes Mellitus, Experimental/genetics , Diabetic Retinopathy/blood , Diabetic Retinopathy/genetics , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Photic Stimulation/methods , Psychomotor Performance , Vision, Ocular/genetics
7.
Invest Ophthalmol Vis Sci ; 52(9): 6766-73, 2011 Aug 29.
Article in English | MEDLINE | ID: mdl-21757588

ABSTRACT

PURPOSE: The optomotor reflex of DBA/2J (D2), DBA/2J-Gpnmb+ (D2-Gpnmb+), and C57BL/6J (B6) mouse strains was assayed, and the retinal ganglion cell (RGC) firing patterns, direction selectivity, vestibulomotor function and central vision was compared between the D2 and B6 mouse lines. METHODS: Intraocular pressure (IOP) measurements, real-time PCR, and immunohistochemical analysis were used to assess the time course of glaucomatous changes in D2 retinas. Behavioral analyses of optomotor head-turning reflex, visible platform Morris water maze and Rotarod measurements were conducted to test vision and vestibulomotor function. Electroretinogram (ERG) measurements were used to assay outer retinal function. The multielectrode array (MEA) technique was used to characterize RGC spiking and direction selectivity in D2 and B6 retinas. RESULTS: Progressive increase in IOP and loss of Brn3a signals in D2 animals were consistent with glaucoma progression starting after 6 months of age. D2 mice showed no response to visual stimulation that evoked robust optomotor responses in B6 mice at any age after eye opening. Spatial frequency threshold was also not measurable in the D2-Gpnmb+ strain control. ERG a- and b-waves, central vision, vestibulomotor function, the spiking properties of ON, OFF, ON-OFF, and direction-selective RGCs were normal in young D2 mice. CONCLUSIONS: The D2 strain is characterized by a lack of optomotor reflex before IOP elevation and RGC degeneration are observed. This behavioral deficit is D2 strain-specific, but is independent of retinal function and glaucoma. Caution is advised when using the optomotor reflex to follow glaucoma progression in D2 mice.


Subject(s)
Glaucoma/physiopathology , Head Movements/physiology , Reflex/physiology , Retinal Ganglion Cells/physiology , Animals , Behavior, Animal , Disease Models, Animal , Electroretinography , Gene Expression , Glaucoma/genetics , Glaucoma/metabolism , Immunohistochemistry , Intraocular Pressure , Mice , Mice, Inbred C57BL , Mice, Inbred DBA , Photic Stimulation , Polymerase Chain Reaction , RNA/genetics , Transcription Factor Brn-3A/biosynthesis , Transcription Factor Brn-3A/genetics
8.
J Neurophysiol ; 106(2): 895-904, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21613583

ABSTRACT

Development of the mammalian visual system is not complete at birth but continues postnatally well after eye opening. Although numerous studies have revealed changes in the development of the thalamus and visual cortex during this time, less is known about the development of response properties of retinal ganglion cells (RGCs). Here, we mapped functional receptive fields of mouse RGCs using a Gaussian white noise checkerboard stimulus and a multielectrode array to record from retinas at eye opening, 3 days later, and 4 wk after birth, when visual responses are essentially mature. Over this time, the receptive field center size of ON and OFF RGC populations decreased. The average receptive field center size of ON RGCs was larger than that of OFF RGCs at eye opening, but they decreased to the same size in the adult. Firing properties were also immature at eye opening. RGCs had longer latencies, lower frequencies of firing, and lower sensitivity than in the adult. Hence, the dramatic maturation of the visual system during the first weeks of visual experience includes the retina.


Subject(s)
Action Potentials/physiology , Eye/growth & development , Photic Stimulation/methods , Retinal Ganglion Cells/cytology , Retinal Ganglion Cells/physiology , Visual Fields/physiology , Animals , Animals, Newborn , Eye/cytology , Mice , Mice, Inbred C57BL
9.
J Neurosci ; 31(19): 7089-101, 2011 May 11.
Article in English | MEDLINE | ID: mdl-21562271

ABSTRACT

Sustained increase in intraocular pressure represents a major risk factor for eye disease, yet the cellular mechanisms of pressure transduction in the posterior eye are essentially unknown. Here we show that the mouse retina expresses mRNA and protein for the polymodal transient receptor potential vanilloid 4 (TRPV4) cation channel known to mediate osmotransduction and mechanotransduction. TRPV4 antibodies labeled perikarya, axons, and dendrites of retinal ganglion cells (RGCs) and intensely immunostained the optic nerve head. Müller glial cells, but not retinal astrocytes or microglia, also expressed TRPV4 immunoreactivity. The selective TRPV4 agonists 4α-PDD and GSK1016790A elevated [Ca2+]i in dissociated RGCs in a dose-dependent manner, whereas the TRPV1 agonist capsaicin had no effect on [Ca2+](RGC). Exposure to hypotonic stimulation evoked robust increases in [Ca2+](RGC). RGC responses to TRPV4-selective agonists and hypotonic stimulation were absent in Ca2+ -free saline and were antagonized by the nonselective TRP channel antagonists Ruthenium Red and gadolinium, but were unaffected by the TRPV1 antagonist capsazepine. TRPV4-selective agonists increased the spiking frequency recorded from intact retinas recorded with multielectrode arrays. Sustained exposure to TRPV4 agonists evoked dose-dependent apoptosis of RGCs. Our results demonstrate functional TRPV4 expression in RGCs and suggest that its activation mediates response to membrane stretch leading to elevated [Ca2+]i and augmented excitability. Excessive Ca2+ influx through TRPV4 predisposes RGCs to activation of Ca2+ -dependent proapoptotic signaling pathways, indicating that TRPV4 is a component of the response mechanism to pathological elevations of intraocular pressure.


Subject(s)
Apoptosis/physiology , Calcium/metabolism , Retinal Ganglion Cells/physiology , TRPV Cation Channels/metabolism , Animals , Apoptosis/drug effects , Axons/metabolism , Capsaicin/pharmacology , Dendrites/metabolism , Dose-Response Relationship, Drug , Electrophysiology , Immunohistochemistry , Leucine/analogs & derivatives , Leucine/pharmacology , Mechanotransduction, Cellular/physiology , Mice , RNA, Messenger/genetics , RNA, Messenger/metabolism , Retinal Ganglion Cells/drug effects , Sulfonamides/pharmacology , TRPV Cation Channels/genetics
10.
Proc Natl Acad Sci U S A ; 107(40): 17374-8, 2010 Oct 05.
Article in English | MEDLINE | ID: mdl-20855606

ABSTRACT

Melanopsin-expressing, intrinsically photosensitive retinal ganglion cells (ipRGCs) form a light-sensitive system separate from rods and cones. Direct light stimulation of ipRGCs can regulate many nonimage-forming visual functions such as photoentrainment of circadian rhythms and pupil responses, and can intensify migraine headache in adults. In mice, ipRGCs are light responsive as early as the day of birth. In contrast, their eyelids do not open until 12-13 d after birth (P12-13), and light signaling from rods and cones does not begin until approximately P10. No physiological or behavioral function is established for ipRGCs in neonates before the onset of rod and cone signaling. Here we report that mouse pups as young as P6 will completely turn away from a light. Light-induced responses of ipRGCs could be readily recorded in retinas of pups younger than P9, and we found no evidence for rod- and cone-mediated visual signaling to the RGCs of these younger mice. These results confirm that negative phototaxis is evident before the onset of rod- and cone-mediated visual signaling, and well before the onset of image-forming vision. Negative phototaxis was absent in mice lacking melanopsin. We conclude that light activation of melanopsin ipRGCs is necessary and sufficient for negative phototaxis. These results strongly suggest that light activation of ipRGCs may regulate physiological functions such as sleep/wake cycles in preterm and neonatal infants.


Subject(s)
Animals, Newborn , Avoidance Learning/physiology , Light Signal Transduction/physiology , Light , Rod Opsins/metabolism , Animals , Behavior, Animal/physiology , Humans , Infant, Newborn , Mice , Mice, Knockout , Photic Stimulation , Retinal Ganglion Cells/cytology , Retinal Ganglion Cells/physiology , Rod Opsins/genetics
11.
J Neurosci ; 27(27): 7245-55, 2007 Jul 04.
Article in English | MEDLINE | ID: mdl-17611277

ABSTRACT

Glutamatergic neurotransmission requires vesicular glutamate transporters (VGLUTs) to sequester glutamate into synaptic vesicles. Generally, VGLUT1 and VGLUT2 isoforms show complementary expression in the CNS and retina. However, little is known about whether isoform-specific expression serves distinct pathways and physiological functions. Here, by examining visual functions in VGLUT1-null mice, we demonstrate that visual signaling from photoreceptors to retinal output neurons requires VGLUT1. However, photoentrainment and pupillary light responses are preserved. We provide evidence that melanopsin-containing, intrinsically photosensitive retinal ganglion cells (RGCs), signaling via VGLUT2 pathways, support these non-image-forming functions. We conclude that VGLUT1 is essential for transmitting visual signals from photoreceptors to second- and third-order neurons, but VGLUT1 is not necessary for intrinsic visual functions. Furthermore, melanopsin and VGLUT2 expression in a subset of RGCs immediately after birth strongly supports the idea that intrinsic vision can function well before rod- and cone-mediated signaling has matured.


Subject(s)
Photoreceptor Cells/physiology , Signal Transduction/physiology , Synapses/physiology , Vesicular Glutamate Transport Protein 1/physiology , Vision, Ocular/physiology , Animals , Evoked Potentials, Visual/physiology , Mice , Mice, Knockout , Photic Stimulation/methods , Protein Isoforms/physiology , Rats , Rats, Long-Evans , Retinal Ganglion Cells/physiology
12.
J Neurosci ; 27(27): 7256-67, 2007 Jul 04.
Article in English | MEDLINE | ID: mdl-17611278

ABSTRACT

Sensory experience refines neuronal structure and functionality. The visual system has proved to be a productive model system to study this plasticity. In the neonatal retina, the dendritic arbors of a large proportion of ganglion cells are diffuse in the inner plexiform layer. With maturation, many of these arbors become monolaminated. Visual deprivation suppresses this remodeling. Little is known of the molecular mechanisms controlling maturational and experience-dependent refinement. Here, we tested the hypothesis that brain-derived neurotrophic factor (BDNF), which is known to regulate dendritic branching and synaptic function in the brain, modulates the developmental and visual experience-dependent refinement of retinal ganglion cells. We used a transgenic mouse line, in which a small number of ganglion cells were labeled with yellow fluorescence protein, to delineate their dendritic structure in vivo. We found that transgenic overexpression of BDNF accelerated the laminar refinement of ganglion cell dendrites, whereas decreased TrkB expression or retina-specific deletion of TrkB, the cognate receptor for BDNF, retarded it. BDNF-TrkB signaling regulated the maturational formation of new branches in ON but not the bilaminated ON-OFF ganglion cells. Furthermore, BDNF overexpression overrides the requirement for visual inputs to stimulate laminar refinement and dendritic branching of ganglion cells. These experiments reveal a previously unrecognized action of BDNF and TrkB in controlling cell-specific, experience-dependent remodeling of neuronal structures in the visual system.


Subject(s)
Brain-Derived Neurotrophic Factor/physiology , Receptor, trkB/physiology , Retina/growth & development , Retina/metabolism , Visual Pathways/growth & development , Visual Pathways/metabolism , Age Factors , Animals , Brain-Derived Neurotrophic Factor/biosynthesis , Brain-Derived Neurotrophic Factor/genetics , Humans , Mice , Mice, Transgenic , Rats , Receptor, trkB/biosynthesis , Receptor, trkB/genetics , Retina/physiology , Sensory Deprivation/physiology , Vision, Ocular/physiology , Visual Pathways/physiology
13.
J Neurosci ; 26(46): 11857-69, 2006 Nov 15.
Article in English | MEDLINE | ID: mdl-17108159

ABSTRACT

Parallel ON and OFF pathways conduct visual signals from bipolar cells in the retina to higher centers in the brain. ON responses are thought to originate by exclusive use of metabotropic glutamate receptor 6 (mGluR6) expressed in retinal ON bipolar cells. Paradoxically, we find ON responses in retinal ganglion cells of mGluR6-null mice, but they occur at long latency. The long-latency ON responses are not blocked by metabotropic glutamate or cholinergic receptor antagonists and are not produced by activation of receptive field surrounds. We show that these longer-latency ON responses are initiated in the OFF pathways. Our results expose a previously unrecognized intrinsic property of OFF retinal pathways that generates responses to light onset. In mGluR6-null mice, long-latency ON responses are observed in the visual cortex, indicating that they can be conducted reliably to higher visual areas. In wild-type (WT) mice, APB (DL-2-amino-4-phosphonobutyric acid), an mGluR6 agonist, blocks normal, short-latency ON responses but unmasks longer-latency ones. We find that these potentially confusing ON responses in the OFF pathway are actively suppressed in WT mice via two pharmacologically separable retinal circuits that are activated by the ON system in the retina. Consequently, we propose that a major function of the signaling of the ON pathway to the OFF pathway is suppression of these mistimed, and therefore inappropriate, light-evoked responses.


Subject(s)
Neural Inhibition/physiology , Neurons/physiology , Receptors, Metabotropic Glutamate/metabolism , Retina/physiology , Visual Cortex/physiology , Visual Pathways/physiology , Action Potentials/drug effects , Action Potentials/physiology , Animals , Excitatory Amino Acid Agonists/pharmacology , Excitatory Amino Acid Antagonists/pharmacology , Female , Glutamic Acid/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Neural Inhibition/drug effects , Neurons/drug effects , Reaction Time/drug effects , Reaction Time/physiology , Receptors, Metabotropic Glutamate/drug effects , Receptors, Metabotropic Glutamate/genetics , Retina/drug effects , Retinal Bipolar Cells/drug effects , Retinal Bipolar Cells/physiology , Retinal Ganglion Cells/drug effects , Retinal Ganglion Cells/physiology , Synaptic Transmission/drug effects , Synaptic Transmission/physiology , Visual Cortex/drug effects , Visual Pathways/drug effects
14.
J Neurosci ; 26(23): 6269-81, 2006 Jun 07.
Article in English | MEDLINE | ID: mdl-16763034

ABSTRACT

Neuronal pentraxins (NPs) define a family of proteins that are homologous to C-reactive and acute-phase proteins in the immune system and have been hypothesized to be involved in activity-dependent synaptic plasticity. To investigate the role of NPs in vivo, we generated mice that lack one, two, or all three NPs. NP1/2 knock-out mice exhibited defects in the segregation of eye-specific retinal ganglion cell (RGC) projections to the dorsal lateral geniculate nucleus, a process that involves activity-dependent synapse formation and elimination. Retinas from mice lacking NP1 and NP2 had cholinergically driven waves of activity that occurred at a frequency similar to that of wild-type mice, but several other parameters of retinal activity were altered. RGCs cultured from these mice exhibited a significant delay in functional maturation of glutamatergic synapses. Other developmental processes, such as pathfinding of RGCs at the optic chiasm and hippocampal long-term potentiation and long-term depression, appeared normal in NP-deficient mice. These data indicate that NPs are necessary for early synaptic refinements in the mammalian retina and dorsal lateral geniculate nucleus. We speculate that NPs exert their effects through mechanisms that parallel the known role of short pentraxins outside the CNS.


Subject(s)
C-Reactive Protein/physiology , Geniculate Bodies/physiology , Nerve Tissue Proteins/physiology , Neurons/metabolism , Retina/physiology , Synapses/physiology , Visual Pathways/physiology , Animals , Cells, Cultured , Geniculate Bodies/growth & development , Glutamic Acid/metabolism , Hippocampus/physiology , Mice , Mice, Knockout , Neuronal Plasticity , Retina/cytology , Retina/growth & development , Retinal Ganglion Cells/physiology , Synapses/metabolism , Synaptic Transmission/physiology , Visual Pathways/growth & development
15.
Neuron ; 48(5): 797-809, 2005 Dec 08.
Article in English | MEDLINE | ID: mdl-16337917

ABSTRACT

The visual cortex is organized into retinotopic maps that preserve an orderly representation of the visual world, achieved by topographically precise inputs from the lateral geniculate nucleus. We show here that geniculocortical mapping is imprecise when the waves of spontaneous activity in the retina during the first postnatal week are disrupted genetically. This anatomical mapping defect is present by postnatal day 8 and has functional consequences, as revealed by optical imaging and microelectrode recording in adults. Pharmacological disruption of these retinal waves during the first week phenocopies the mapping defect, confirming both the site and the timing of the disruption in neural activity responsible for the defect. Analysis shows that the geniculocortical miswiring is not a trivial or necessary consequence of the retinogeniculate defect. Our findings demonstrate that disrupting early spontaneous activity in the eye alters thalamic connections to the cortex.


Subject(s)
Animals, Newborn/physiology , Brain Mapping , Retina/physiology , Visual Cortex/physiology , Animals , Bridged Bicyclo Compounds, Heterocyclic/pharmacology , Geniculate Bodies/physiology , Mice , Mice, Knockout , Neurons, Afferent/physiology , Nicotinic Agonists/pharmacology , Pyridines/pharmacology , Receptors, Nicotinic/deficiency , Receptors, Nicotinic/physiology , Retina/drug effects , Synaptic Transmission/physiology , Time Factors , Visual Cortex/growth & development , Visual Fields/physiology
16.
Vis Neurosci ; 22(3): 263-74, 2005.
Article in English | MEDLINE | ID: mdl-16079002

ABSTRACT

Calcium ion (Ca(2+)) signaling has been widely implicated in developmental events in the retina, but little is known about the specific mechanisms utilized by developing neurons to decrease intracellular Ca(2+). Using immunocytochemistry, we determined the expression profiles of all known isoforms of a key Ca(2+) transporter, the plasma membrane Ca(2+) ATPase (PMCA), in the rat retina. During the first postnatal week, the four PMCA isoforms were expressed in patterns that differed from their expression in the adult retina. At birth, PMCA1 was found in the ventricular zone and nascent cell processes in the distal retina as well as in ganglion and amacrine cells. After the first postnatal week, PMCA1 became restricted to photoreceptors and cone bipolar cells. By P10 (by postnatal day 10), most inner retinal PMCA consisted of PMCA2 and PMCA3. Prominent PMCA4 expression appeared after the first postnatal week and was confined primarily to the ON sublamina of the inner plexiform layer (IPL). The four PMCA isoforms could play distinct functional roles in the development of the mammalian retina even before synaptic circuits are established. Their expression patterns are consistent with the hypothesis that inner and outer retinal neurons have different Ca(2+) handling needs.


Subject(s)
Calcium-Transporting ATPases/metabolism , Cation Transport Proteins/metabolism , Neurons/enzymology , Retina/cytology , Retina/growth & development , Age Factors , Amino Acid Transport Systems/metabolism , Animals , Animals, Newborn , Calcium-Transporting ATPases/classification , Cation Transport Proteins/classification , Choline O-Acetyltransferase/metabolism , Diagnostic Imaging/methods , Glutamate Decarboxylase/metabolism , Immunohistochemistry/methods , Isoenzymes/metabolism , Nerve Tissue Proteins/metabolism , Plasma Membrane Calcium-Transporting ATPases , Rats , Rats, Long-Evans , Tyrosine 3-Monooxygenase/metabolism , Vesicular Inhibitory Amino Acid Transport Proteins , Vesicular Transport Proteins/metabolism
17.
Nat Neurosci ; 8(8): 1022-7, 2005 Aug.
Article in English | MEDLINE | ID: mdl-16025107

ABSTRACT

In mammals, retinal ganglion cell (RGC) projections initially intermingle and then segregate into a stereotyped pattern of eye-specific layers in the dorsal lateral geniculate nucleus (dLGN). Here we found that in mice deficient for ephrin-A2, ephrin-A3 and ephrin-A5, eye-specific inputs segregated but the shape and location of eye-specific layers were profoundly disrupted. In contrast, mice that lacked correlated retinal activity did not segregate eye-specific inputs. Inhibition of correlated neural activity in ephrin mutants led to overlapping retinal projections that were located in inappropriate regions of the dLGN. Thus, ephrin-As and neural activity act together to control patterning of eye-specific retinogeniculate layers.


Subject(s)
Body Patterning/physiology , Ephrin-A2/physiology , Ephrin-A3/physiology , Ephrin-A5/physiology , Geniculate Bodies/physiology , Retinal Ganglion Cells/physiology , Synaptic Transmission/physiology , Animals , Brain Mapping , Ephrin-A2/deficiency , Ephrin-A3/deficiency , Ephrin-A5/deficiency , Mice , Mice, Knockout , Receptor, EphA2/deficiency , Receptor, EphA3/deficiency , Receptor, EphA5/deficiency , Visual Pathways/physiology
18.
Neuron ; 41(6): 839-41, 2004 Mar 25.
Article in English | MEDLINE | ID: mdl-15046714

ABSTRACT

Taurine, a multifunctional amino acid prevalent in developing nervous tissues, regulates the number of rod photoreceptors in developing postnatal rodent retina. In this issue of Neuron, Young and Cepko show that taurine acts via GlyRalpha2 subunit-containing glycine receptors expressed by retinal progenitor cells at birth.


Subject(s)
Ion Channel Gating/physiology , Receptors, Glycine/metabolism , Retinal Rod Photoreceptor Cells/growth & development , Retinal Rod Photoreceptor Cells/metabolism , Stem Cells/metabolism , Taurine/metabolism , Animals , Cell Differentiation/drug effects , Cell Differentiation/genetics , Cell Division/drug effects , Cell Division/genetics , Cell Lineage/drug effects , Cell Lineage/genetics , Humans , Ion Channel Gating/drug effects , Receptors, Glycine/drug effects , Receptors, Glycine/genetics , Retinal Rod Photoreceptor Cells/drug effects , Stem Cells/drug effects , Taurine/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...