Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 33
Filter
Add more filters










Publication year range
1.
Cephalalgia ; 39(13): 1623-1634, 2019 11.
Article in English | MEDLINE | ID: mdl-29940781

ABSTRACT

OBJECTIVE: To review clinical and pre-clinical evidence supporting the role of visual pathways, from the eye to the cortex, in the development of photophobia in headache disorders. BACKGROUND: Photophobia is a poorly understood light-induced phenomenon that emerges in a variety of neurological and ophthalmological conditions. Over the years, multiple mechanisms have been proposed to explain its causes; however, scarce research and lack of systematic assessment of photophobia in patients has made the search for answers quite challenging. In the field of headaches, significant progress has been made recently on how specific visual networks contribute to photophobia features such as light-induced intensification of headache, increased perception of brightness and visual discomfort, which are frequently experienced by migraineurs. Such progress improved our understanding of the phenomenon and points to abnormal processing of light by both cone/rod-mediated image-forming and melanopsin-mediated non-image-forming visual pathways, and the consequential transfer of photic signals to multiple brain regions involved in sensory, autonomic and emotional regulation. CONCLUSION: Photophobia phenotype is diverse, and the relative contribution of visual, trigeminal and autonomic systems may depend on the disease it emerges from. In migraine, photophobia could result from photic activation of retina-driven pathways involved in the regulation of homeostasis, making its association with headache more complex than previously thought.


Subject(s)
Headache/physiopathology , Photophobia/physiopathology , Visual Pathways/physiopathology , Animals , Blindness/physiopathology , Brain Stem/physiopathology , Color , Headache/complications , Humans , Light/adverse effects , Mesencephalon/physiopathology , Mice , Migraine Disorders/complications , Migraine Disorders/physiopathology , Photic Stimulation/adverse effects , Photophobia/etiology , Retinal Ganglion Cells/physiology , Retinal Rod Photoreceptor Cells/physiology , Retinal Rod Photoreceptor Cells/radiation effects , Rod Opsins/physiology , Somatosensory Cortex/physiopathology , Thalamus/physiopathology
2.
Vis Neurosci ; 35: E004, 2018 01.
Article in English | MEDLINE | ID: mdl-29905117

ABSTRACT

A unique class of intrinsically photosensitive retinal ganglion cells in mammalian retinae has been recently discovered and characterized. These neurons can generate visual signals in the absence of inputs from rods and cones, the conventional photoreceptors in the visual system. These light sensitive ganglion cells (mRGCs) express the non-rod, non-cone photopigment melanopsin and play well documented roles in modulating pupil responses to light, photoentrainment of circadian rhythms, mood, sleep and other adaptive light functions. While most research efforts in mammals have focused on mRGCs in retina, recent studies reveal that melanopsin is expressed in non-retinal tissues. For example, light-evoked melanopsin activation in extra retinal tissue regulates pupil constriction in the iris and vasodilation in the vasculature of the heart and tail. As another example of nonretinal melanopsin expression we report here the previously unrecognized localization of this photopigment in nerve fibers within the cornea. Surprisingly, we were unable to detect light responses in the melanopsin-expressing corneal fibers in spite of our histological evidence based on genetically driven markers and antibody staining. We tested further for melanopsin localization in cell bodies of the trigeminal ganglia (TG), the principal nuclei of the peripheral nervous system that project sensory fibers to the cornea, and found expression of melanopsin mRNA in a subset of TG neurons. However, neither electrophysiological recordings nor calcium imaging revealed any light responsiveness in the melanopsin positive TG neurons. Given that we found no light-evoked activation of melanopsin-expressing fibers in cornea or in cell bodies in the TG, we propose that melanopsin protein might serve other sensory functions in the cornea. One justification for this idea is that melanopsin expressed in Drosophila photoreceptors can serve as a temperature sensor.


Subject(s)
Cornea/metabolism , Gene Expression Regulation/physiology , Rod Opsins/genetics , Trigeminal Ganglion/metabolism , Animals , Cell Body/metabolism , Cells, Cultured , Dependovirus/genetics , Electrophysiology , Female , Guinea Pigs , Immunohistochemistry , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Nerve Fibers/metabolism , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Rod Opsins/metabolism , Transfection
3.
PLoS One ; 11(2): e0149501, 2016.
Article in English | MEDLINE | ID: mdl-26895233

ABSTRACT

To understand visual functions mediated by intrinsically photosensitive melanopsin-expressing retinal ganglion cells (mRGCs), it is important to elucidate axonal projections from these cells into the brain. Initial studies reported that melanopsin is expressed only in retinal ganglion cells within the eye. However, recent studies in Opn4-Cre mice revealed Cre-mediated marker expression in multiple brain areas. These discoveries complicate the use of melanopsin-driven genetic labeling techniques to identify retinofugal projections specifically from mRGCs. To restrict labeling to mRGCs, we developed a recombinant adeno-associated virus (AAV) carrying a Cre-dependent reporter (human placental alkaline phosphatase) that was injected into the vitreous of Opn4-Cre mouse eyes. The labeling observed in the brain of these mice was necessarily restricted specifically to retinofugal projections from mRGCs in the injected eye. We found that mRGCs innervate multiple nuclei in the basal forebrain, hypothalamus, amygdala, thalamus and midbrain. Midline structures tended to be bilaterally innervated, whereas the lateral structures received mostly contralateral innervation. As validation of our approach, we found projection patterns largely corresponded with previously published results; however, we have also identified a few novel targets. Our discovery of projections to the central amygdala suggests a possible direct neural pathway for aversive responses to light in neonates. In addition, projections to the accessory optic system suggest that mRGCs play a direct role in visual tracking, responses that were previously attributed to other classes of retinal ganglion cells. Moreover, projections to the zona incerta raise the possibility that mRGCs could regulate visceral and sensory functions. However, additional studies are needed to investigate the actual photosensitivity of mRGCs that project to the different brain areas. Also, there is a concern of "overlabeling" with very sensitive reporters that uncover low levels of expression. Light-evoked signaling from these cells must be shown to be of sufficient sensitivity to elicit physiologically relevant responses.


Subject(s)
Retina/metabolism , Retinal Ganglion Cells/metabolism , Rod Opsins/biosynthesis , Alkaline Phosphatase/genetics , Alkaline Phosphatase/metabolism , Animals , Brain/cytology , Brain/metabolism , Dependovirus/genetics , GPI-Linked Proteins/genetics , GPI-Linked Proteins/metabolism , Genes, Reporter , Humans , Injections, Intraocular , Integrases/genetics , Isoenzymes/genetics , Isoenzymes/metabolism , Mice , Mice, Inbred C57BL , Retina/cytology
4.
Ophthalmology ; 120(12): 2706-2713, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24139125

ABSTRACT

PURPOSE: Fetal mice require light exposure in utero during early gestation for normal vascular development in the eye. Because angiogenic abnormalities in retinopathy of prematurity (ROP) are manifested in preterm infants, we investigated whether day length during early gestation was associated with severe ROP (SROP). DESIGN: Single-center, retrospective cohort study. PARTICIPANTS: We included a total of 343 premature infants (401-1250 g birth weight [BW], from 1998-2002): 684 eyes (1 eye each of 2 patients excluded) with 76 eyes developing SROP, defined as (1) classic threshold ROP in zone I or II, (2) type 1 ROP in zone I, or (3) in a few eyes, type 1 ROP in posterior zone II that was treated. METHODS: For each infant, average day length (ADL) was calculated during different cumulative time periods and time windows after the estimated date of conception (EDC). Multiple logistic regression analysis (with generalized estimating equations to account for inter-eye correlation) was performed. MAIN OUTCOME MEASURES: Association of ADL during early gestation with SROP. RESULTS: In a model evaluating all 684 eyes with 76 eyes developing SROP, BW, gestational age, multiple births, race, per capita income in the mother's residence ZIP code, and ADL during the first 90 days after the EDC were factors associated with the development of SROP. Each additional hour of ADL (90 days) decreased the likelihood of SROP by 28% (P = 0.015; odds ratio [OR], 0.72; 95% confidence interval [CI], 0.55-0.94). In a model evaluating the subset of 146 prethreshold ROP eyes with 76 eyes developing SROP, each additional hour of ADL during the first 105 days after the EDC decreased the likelihood of SROP by 46% (P = 0.001; OR, 0.54; 95% CI, 0.37-0.78). Time windows when ADL was most closely associated with SROP were 31 to 60 days and 61 to 90 days after the EDC for the all eyes and the prethreshold ROP eyes models, respectively. CONCLUSIONS: Higher ADL during early gestation was associated with a lower risk for SROP and may imply a role for prophylactic light treatment during early gestation to decrease the risk of SROP.


Subject(s)
Infant, Premature , Photoperiod , Pregnancy , Retinopathy of Prematurity/etiology , Cohort Studies , Female , Gestational Age , Humans , Infant, Newborn , Infant, Very Low Birth Weight , Male , Parity , Retrospective Studies , Risk Factors , Seasons , Time Factors
5.
Nature ; 494(7436): 243-6, 2013 Feb 14.
Article in English | MEDLINE | ID: mdl-23334418

ABSTRACT

Vascular patterning is critical for organ function. In the eye, there is simultaneous regression of embryonic hyaloid vasculature (important to clear the optical path) and formation of the retinal vasculature (important for the high metabolic demands of retinal neurons). These events occur postnatally in the mouse. Here we have identified a light-response pathway that regulates both processes. We show that when mice are mutated in the gene (Opn4) for the atypical opsin melanopsin, or are dark-reared from late gestation, the hyaloid vessels are persistent at 8 days post-partum and the retinal vasculature overgrows. We provide evidence that these vascular anomalies are explained by a light-response pathway that suppresses retinal neuron number, limits hypoxia and, as a consequence, holds local expression of vascular endothelial growth factor (VEGFA) in check. We also show that the light response for this pathway occurs in late gestation at about embryonic day 16 and requires the photopigment in the fetus and not the mother. Measurements show that visceral cavity photon flux is probably sufficient to activate melanopsin-expressing retinal ganglion cells in the mouse fetus. These data thus show that light--the stimulus for function of the mature eye--is also critical in preparing the eye for vision by regulating retinal neuron number and initiating a series of events that ultimately pattern the ocular blood vessels.


Subject(s)
Eye/blood supply , Eye/growth & development , Fetus/radiation effects , Light Signal Transduction/radiation effects , Light , Retinal Neurons/radiation effects , Rod Opsins/metabolism , Animals , Cell Count , Cell Hypoxia/radiation effects , Eye/metabolism , Eye/radiation effects , Female , Fetus/cytology , Fetus/embryology , Fetus/metabolism , Mice , Mice, Inbred C57BL , Neovascularization, Pathologic , Neovascularization, Physiologic/radiation effects , Photons , Retinal Ganglion Cells/cytology , Retinal Ganglion Cells/metabolism , Retinal Ganglion Cells/radiation effects , Retinal Neurons/cytology , Retinal Neurons/metabolism , Rod Opsins/deficiency , Rod Opsins/genetics , Vascular Endothelial Growth Factor A/metabolism
6.
PLoS One ; 8(12): e83974, 2013.
Article in English | MEDLINE | ID: mdl-24391855

ABSTRACT

Melanopsin-expressing retinal ganglion cells (mRGCs) in the eye play an important role in many light-activated non-image-forming functions including neonatal photoaversion and the adult pupillary light reflex (PLR). MRGCs rely on glutamate and possibly PACAP (pituitary adenylate cyclase-activating polypeptide) to relay visual signals to the brain. However, the role of these neurotransmitters for individual non-image-forming responses remains poorly understood. To clarify the role of glutamatergic signaling from mRGCs in neonatal aversion to light and in adult PLR, we conditionally deleted vesicular glutamate transporter (VGLUT2) selectively from mRGCs in mice. We found that deletion of VGLUT2 in mRGCs abolished negative phototaxis and light-induced distress vocalizations in neonatal mice, underscoring a necessary role for glutamatergic signaling. In adult mice, loss of VGLUT2 in mRGCs resulted in a slow and an incomplete PLR. We conclude that glutamatergic neurotransmission from mRGCs is required for neonatal photoaversion but is complemented by another non-glutamatergic signaling mechanism for the pupillary light reflex in adult mice. We speculate that this complementary signaling might be due to PACAP neurotransmission from mRGCs.


Subject(s)
Light , Pituitary Adenylate Cyclase-Activating Polypeptide/metabolism , Reflex, Pupillary/physiology , Retinal Ganglion Cells/metabolism , Rod Opsins/physiology , Synaptic Transmission/physiology , Vesicular Glutamate Transport Protein 2/physiology , Animals , Animals, Newborn , Behavior, Animal , Female , Immunoenzyme Techniques , Integrases/metabolism , Light Signal Transduction , Male , Mice , Mice, Knockout , Neurotransmitter Agents/metabolism , Photic Stimulation , Reflex, Pupillary/radiation effects , Retinal Ganglion Cells/radiation effects , Vision Disorders , Vision, Ocular/physiology , Vision, Ocular/radiation effects
7.
PLoS One ; 7(9): e43787, 2012.
Article in English | MEDLINE | ID: mdl-23028470

ABSTRACT

Melanopsin-expressing intrinsically photosensitive retinal ganglion cells (ipRGCs) are the only functional photoreceptive cells in the eye of newborn mice. Through postnatal day 9, in the absence of functional rods and cones, these ipRGCs mediate a robust avoidance behavior to a light source, termed negative phototaxis. To determine whether this behavior is associated with an aversive experience in neonatal mice, we characterized light-induced vocalizations and patterns of neuronal activation in regions of the brain involved in the processing of aversive and painful stimuli. Light evoked distinct melanopsin-dependent ultrasonic vocalizations identical to those emitted under stressful conditions, such as isolation from the litter. In contrast, light did not evoke the broad-spectrum calls elicited by acute mechanical pain. Using markers of neuronal activation, we found that light induced the immediate-early gene product Fos in the posterior thalamus, a brain region associated with the enhancement of responses to mechanical stimulation of the dura by light, and thought to be the basis for migrainous photophobia. Additionally, light induced the phosphorylation of extracellular-related kinase (pERK) in neurons of the central amygdala, an intracellular signal associated with the processing of the aversive aspects of pain. However, light did not activate Fos expression in the spinal trigeminal nucleus caudalis, the primary receptive field for painful stimulation to the head. We conclude that these light-evoked vocalizations and the distinct pattern of brain activation in neonatal mice are consistent with a melanopsin-dependent neural pathway involved in processing light as an aversive but not acutely painful stimulus.


Subject(s)
Light , Retinal Ganglion Cells/metabolism , Rod Opsins/metabolism , Vocalization, Animal/physiology , Amygdala/metabolism , Animals , Extracellular Signal-Regulated MAP Kinases/metabolism , Mice , Photic Stimulation , Proto-Oncogene Proteins c-fos/metabolism , Thalamus/metabolism , Trigeminal Ganglion/metabolism , Vision, Ocular/physiology
8.
J Neurosci ; 31(35): 12663-73, 2011 Aug 31.
Article in English | MEDLINE | ID: mdl-21880927

ABSTRACT

Dopaminergic amacrine (DA) cells play multiple and important roles in retinal function. Neurotrophins are known to modulate the number and morphology of DA cells, but the underlying regulatory mechanisms are unclear. Here, we investigate how neurotrophin-3 (NT-3) regulates DA cell density in the mouse retina. We demonstrate that overexpression of NT-3 upregulates DA cell number and leads to a consequent increase in the density of DA cell dendrites. To examine the mechanisms of DA cell density increase, we further investigate the effect of NT-3 overexpression on retinal apoptosis and mitosis during development. We find that NT-3 does not affect the well known wave of retinal cell apoptosis that normally occurs during the first 2 weeks after birth. Instead, overexpression of NT-3 promotes additional mitosis of DA cells at postnatal day 4, but does not affect cell mitosis before birth, the peak period of amacrine cell genesis in wild-type retinas. We next show that retinal explants cultured from birth to day 7 without extra NT-3 produced by lens exhibit similar number of DA cells as in wild type, further supporting the notion that postnatal overexpression of lens-derived NT-3 affects DA cell number. Moreover, the additional mitosis after birth in NT-3-overexpressing mice does not occur in calretinin-positive amacrine cells or PKC-positive rod ON bipolar cells. Thus, the NT-3-triggered wave of cell mitosis after birth is specific for the retinal DA cells.


Subject(s)
Amacrine Cells/physiology , Dopamine/metabolism , Gene Expression Regulation, Developmental/physiology , Neurogenesis/physiology , Neurotrophin 3/metabolism , Retina/cytology , Animals , Animals, Newborn , Bromodeoxyuridine/metabolism , Calbindin 2 , Cell Cycle/genetics , Cell Death , Gene Expression Regulation, Developmental/genetics , In Situ Nick-End Labeling/methods , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neurogenesis/genetics , Neurotrophin 3/genetics , Protein Kinase C/metabolism , S100 Calcium Binding Protein G/metabolism , Tyrosine 3-Monooxygenase/metabolism
9.
J Neurosci ; 31(8): 2769-80, 2011 Feb 23.
Article in English | MEDLINE | ID: mdl-21414899

ABSTRACT

Inhibitory interneurons play a critical role in coordinating the activity of neural circuits. To explore the mechanisms that direct the organization of inhibitory circuits, we analyzed the involvement of tropomyosin-related kinase B (TrkB) in the assembly and maintenance of GABAergic inhibitory synapses between Golgi and granule cells in the mouse cerebellar cortex. We show that TrkB acts directly within each cell-type to regulate synaptic differentiation. TrkB is required not only for assembly, but also maintenance of these synapses and acts, primarily, by regulating the localization of synaptic constituents. Postsynaptically, TrkB controls the localization of a scaffolding protein, gephyrin, but acts at a step subsequent to the localization of a cell adhesion molecule, Neuroligin-2. Importantly, TrkB is required for the localization of an Ig superfamily cell adhesion molecule, Contactin-1, in Golgi and granule cells and the absence of Contactin-1 also results in deficits in inhibitory synaptic development. Thus, our findings demonstrate that TrkB controls the assembly and maintenance of GABAergic synapses and suggest that TrkB functions, in part, through promoting synaptic adhesion.


Subject(s)
Cell Differentiation/physiology , Cerebellar Cortex/enzymology , Cerebellar Cortex/growth & development , Receptor, trkB/physiology , Synapses/physiology , gamma-Aminobutyric Acid/physiology , Animals , Cell Adhesion/genetics , Cell Adhesion/physiology , Cell Differentiation/genetics , Interneurons/cytology , Interneurons/enzymology , Mice , Mice, Knockout , Mice, Transgenic , Synapses/enzymology , Synapses/genetics , Synaptic Transmission/genetics , Tropomyosin/physiology
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.
Neuron ; 62(2): 230-41, 2009 Apr 30.
Article in English | MEDLINE | ID: mdl-19409268

ABSTRACT

In the few days prior to eye-opening in mice, the excitatory drive underlying waves switches from cholinergic to glutamatergic. Here, we describe the unique synaptic and spatiotemporal properties of waves generated by the retina's glutamatergic circuits. First, knockout mice lacking vesicular glutamate transporter type 1 do not have glutamatergic waves, but continue to exhibit cholinergic waves, demonstrating that the two wave-generating circuits are linked. Second, simultaneous outside-out patch and whole-cell recordings reveal that retinal waves are accompanied by transient increases in extrasynaptic glutamate, directly demonstrating the existence of glutamate spillover during waves. Third, the initiation rate and propagation speed of retinal waves, as assayed by calcium imaging, are sensitive to pharmacological manipulations of spillover and inhibition, demonstrating a role for both signaling pathways in shaping the spatiotemporal properties of glutamatergic retinal waves.


Subject(s)
Glutamic Acid/metabolism , Retinal Ganglion Cells/physiology , Synapses/physiology , Synaptic Transmission/physiology , Amino Acid Transport Systems, Acidic/deficiency , Animals , Animals, Newborn , Aspartic Acid/pharmacology , Calcium/metabolism , Dihydro-beta-Erythroidine/pharmacology , Drug Interactions , Excitatory Amino Acid Agonists/pharmacology , Excitatory Amino Acid Antagonists/pharmacology , GABA Antagonists/pharmacology , In Vitro Techniques , Mice , Mice, Inbred C57BL , Mice, Knockout , Models, Biological , N-Methylaspartate/pharmacology , Neural Inhibition/drug effects , Neural Inhibition/physiology , Nicotinic Antagonists/pharmacology , Patch-Clamp Techniques/methods , Pyridazines/pharmacology , Quinoxalines/pharmacology , Retinal Ganglion Cells/drug effects , Synapses/genetics , Synaptic Transmission/drug effects , Time Factors , Valine/analogs & derivatives , Valine/pharmacology , Vesicular Glutamate Transport Protein 1/deficiency , Vesicular Glutamate Transport Protein 1/genetics
12.
J Comp Neurol ; 514(5): 449-58, 2009 Jun 10.
Article in English | MEDLINE | ID: mdl-19350645

ABSTRACT

The morphology of dendrites constrains and reflects the nature of synaptic inputs to neurons. The visual system has served as a useful model to show how visual function is determined by the arborization patterns of neuronal processes. In retina, light ON and light OFF responding ganglion cells selectively elaborate their dendritic arbors in distinct sublamina, where they receive, respectively, inputs from ON and OFF bipolar cells. During neonatal maturation, the bilaminarly distributed dendritic arbors of ON-OFF retinal ganglion cells (RGCs) are refined to more narrowly localized monolaminar structures characteristic of ON or OFF RGCs. Recently, brain-derived neurotrophic factor (BDNF) has been shown to regulate this laminar refinement, and to enhance the development of dendritic branches selectively of ON RGCs. Although other related neurotrophins are known to regulate neuronal process formation in the central nervous system, little is known about their action in maturing retina. Here, we report that overexpression of neurotrophin-3 (NT-3) in the eye accelerates RGC laminar refinement before eye opening. Furthermore, NT-3 overexpression increases dendritic branch number but reduces dendritic elongation preferentially in ON-OFF RGCs, a process that also occurs before eye opening. NT-3 overexpression does affect dendritic maturation in ON RGCs, but to a much less degree. Taken together, our results suggest that NT-3 and BDNF exhibit overlapping effects in laminar refinement but distinct RGC-cell-type specific effects in shaping dendritic arborization during postnatal development.


Subject(s)
Dendrites/physiology , Eye/growth & development , Neurotrophin 3/metabolism , Retinal Ganglion Cells/physiology , Analysis of Variance , Animals , Blotting, Western , Brain-Derived Neurotrophic Factor/metabolism , Dendrites/ultrastructure , Eye/cytology , Eye/metabolism , Immunohistochemistry , Mice , Mice, Transgenic , Microscopy, Confocal , Neurotrophin 3/genetics , Rats , Retinal Ganglion Cells/cytology
13.
Mol Cell Neurosci ; 38(3): 431-43, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18511296

ABSTRACT

BDNF signaling through its TrkB receptor plays a pivotal role in activity-dependent refinement of synaptic connectivity of retinal ganglion cells. Additionally, studies using TrkB knockout mice have suggested that BDNF/TrkB signaling is essential for the development of photoreceptors and for synaptic communication between photoreceptors and second order retinal neurons. Thus the action of BDNF on refinement of synaptic connectivity of retinal ganglion cells could be a direct effect in the inner retina, or it could be secondary to its proposed role in rod maturation and in the formation of rod to bipolar cell synaptic transmission. To address this matter we have conditionally eliminated TrkB within the retina. We find that rod function and synaptic transmission to bipolar cells is not compromised in these conditional knockout mice. Consistent with previous work, we find that inner retina neural development is regulated by retinal BDNF/TrkB signaling. Specifically we show here also that the complexity of neuronal processes of dopaminergic cells is reduced in conditional TrkB knockout mice. We conclude that retinal BDNF/TrkB signaling has its primary role in the development of inner retinal neuronal circuits, and that this action is not a secondary effect due to the loss of visual signaling in the outer retina.


Subject(s)
Receptor, trkB/physiology , Retina/growth & development , Retina/metabolism , Animals , Brain-Derived Neurotrophic Factor/deficiency , Brain-Derived Neurotrophic Factor/genetics , Brain-Derived Neurotrophic Factor/physiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Nerve Net/growth & development , Nerve Net/metabolism , Receptor, trkB/deficiency , Receptor, trkB/genetics , Rod Cell Outer Segment/metabolism , Rod Cell Outer Segment/physiology , Signal Transduction/genetics , Signal Transduction/physiology
14.
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
15.
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
16.
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
17.
J Neurosci ; 26(27): 7201-11, 2006 Jul 05.
Article in English | MEDLINE | ID: mdl-16822977

ABSTRACT

Transmission of visual signals at the first retinal synapse is associated with changes in calcium concentration in photoreceptors and bipolar cells. We investigated how loss of plasma membrane Ca2+ ATPase isoform 2 (PMCA2), the calcium transporter isoform with the highest affinity for Ca2+/calmodulin, affects transmission of rod- and cone-mediated responses. PMCA2 expression in the neuroblast layer was observed soon after birth; in the adult, PMCA2 was expressed in inner segments and synaptic terminals of rod photoreceptors, in rod bipolar cells, and in most inner retinal neurons but was absent from cones. To determine the role of PMCA2 in retinal signaling, we compared morphology and light responses of retinas from control mice and deafwaddler dfw2J mice, which lack functional PMCA2 protein. The cytoarchitecture of retinas from control and dfw2J mice was indistinguishable at the light microscope level. Suction electrode recordings revealed no difference in the sensitivity or amplitude of outer segment light responses of control and dfw2J rods. However, rod-mediated ERG b-wave responses in dfw2J mice were approximately 45% smaller and significantly slower than those of control mice. Furthermore, recordings from individual rod bipolar cells showed that the sensitivity of transmission at the rod output synapse was reduced by approximately 50%. No changes in the amplitude or timing of cone-mediated ERG responses were observed. These results suggest that PMCA2-mediated Ca2+ extrusion modulates the amplitude and timing of the high-sensitivity rod pathway to a much greater extent than that of the cone pathway.


Subject(s)
Calcium-Transporting ATPases/metabolism , Calcium/metabolism , Cation Transport Proteins/metabolism , Dark Adaptation/physiology , Retina/physiology , Vision, Ocular/physiology , Animals , Calcium-Transporting ATPases/genetics , Cation Transport Proteins/genetics , Cell Membrane/metabolism , Evoked Potentials, Visual/physiology , Female , Gene Expression Regulation, Developmental/physiology , Male , Mice , Mice, Inbred BALB C , Mice, Inbred CBA , Mice, Neurologic Mutants , Photic Stimulation , Plasma Membrane Calcium-Transporting ATPases , Retina/cytology , Retina/growth & development , Retinal Bipolar Cells/physiology , Retinal Cone Photoreceptor Cells/physiology , Retinal Rod Photoreceptor Cells/physiology , Synapses/physiology
18.
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
19.
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
20.
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
SELECTION OF CITATIONS
SEARCH DETAIL
...