Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 57
Filtrar
1.
J Neurosci ; 39(16): 3041-3056, 2019 04 17.
Artigo em Inglês | MEDLINE | ID: mdl-30737308

RESUMO

The mammalian visual system operates over an extended range of ambient light levels by switching between rod and cone photoreceptors. Rod-driven vision is sluggish, highly sensitive, and operates in dim or scotopic lights, whereas cone-driven vision is brisk, less sensitive, and operates in bright or photopic lights. At intermediate or mesopic lights, vision transitions seamlessly from rod-driven to cone-driven, despite the profound differences in rod and cone response dynamics. The neural mechanisms underlying such a smooth handoff are not understood. Using an operant behavior assay, electrophysiological recordings, and mathematical modeling we examined the neural underpinnings of the mesopic visual transition in mice of either sex. We found that rods, but not cones, drive visual sensitivity to temporal light variations over much of the mesopic range. Surprisingly, speeding up rod photoresponse recovery kinetics in transgenic mice improved visual sensitivity to slow temporal variations, in the range where perceptual sensitivity is governed by Weber's law of sensation. In contrast, physiological processes acting downstream from phototransduction limit sensitivity to high frequencies and temporal resolution. We traced the paradoxical control of visual temporal sensitivity to rod photoresponses themselves. A scenario emerges where perceptual sensitivity is limited by: (1) the kinetics of neural processes acting downstream from phototransduction in scotopic lights, (2) rod response kinetics in mesopic lights, and (3) cone response kinetics as light levels rise into the photopic range.SIGNIFICANCE STATEMENT Our ability to detect flickering lights is constrained by the dynamics of the slowest step in the visual pathway. Cone photoresponse kinetics limit visual temporal sensitivity in bright (photopic) lights, whereas mechanisms in the inner retina limit sensitivity in dim (scotopic) lights. The neural mechanisms underlying the transition between scotopic and photopic vision in mesopic lights, when both rods are cones are active, are unknown. This study provides a missing link in this mechanism by establishing that rod photoresponse kinetics limit temporal sensitivity during the mesopic transition. Surprisingly, this range is where Weber's Law of Sensation governs temporal contrast sensitivity in mouse. Our results will help guide future studies of complex and dynamic interactions between rod-cone signals in the mesopic retina.


Assuntos
Comportamento de Escolha/fisiologia , Sensibilidades de Contraste/fisiologia , Visão Mesópica/fisiologia , Células Fotorreceptoras Retinianas Bastonetes/fisiologia , Animais , Condicionamento Operante/fisiologia , Feminino , Masculino , Camundongos , Modelos Teóricos
2.
Proc Natl Acad Sci U S A ; 110(13): 5181-6, 2013 Mar 26.
Artigo em Inglês | MEDLINE | ID: mdl-23479632

RESUMO

Stargardt type 3 (STGD3) disease is a juvenile macular dystrophy caused by mutations in the ELOVL4 (Elongation of very long chain fatty acids 4) gene. Its protein product, ELOVL4, is an elongase required for the biosynthesis of very long-chain polyunsaturated fatty acids (VLC-PUFAs). It is unclear whether photoreceptor degeneration in STGD3 is caused by loss of VLC-PUFAs or by mutated ELOVL4 protein trafficking/aggregation. We therefore generated conditional knockout (cKO) mice with Elovl4 ablated in rods or cones and compared their phenotypes to transgenic (TG) animals that express the human STGD3-causing ELOVL4(STGD3) allele. Gas chromatography-mass spectrometry was used to assess C30-C34 VLC-PUFA and N-retinylidene-N-retinylethanolamine content; electroretinography was used to measure phototransduction and outer retinal function; electron microscopy was used for retinal ultrastructure; and the optomotor tracking response was used to test scotopic and photopic visual performance. Elovl4 transcription and biosynthesis of C30-C34 VLC-PUFAs in rod cKO and TG retinas were reduced up to 98%, whereas the content of docosahexaenoic acid was diminished in TG, but not rod cKO, retinas. Despite the near-total loss of the retinal VLC-PUFA content, rod and cone cKO animals exhibited no electrophysiological or behavioral deficits, whereas the typical rod-cone dystrophic pattern was observed in TG animals. Our data suggest that photoreceptor-specific VLC-PUFA depletion is not sufficient to induce the STGD3 phenotype, because depletion alone had little effect on photoreceptor survival, phototransduction, synaptic transmission, and visual behavior.


Assuntos
Proteínas do Olho/metabolismo , Ácidos Graxos Insaturados/metabolismo , Degeneração Macular/congênito , Proteínas de Membrana/metabolismo , Células Fotorreceptoras de Vertebrados/metabolismo , Retina/metabolismo , Transmissão Sináptica , Visão Ocular , Animais , Sobrevivência Celular/genética , Modelos Animais de Doenças , Proteínas do Olho/genética , Ácidos Graxos Insaturados/genética , Humanos , Degeneração Macular/genética , Degeneração Macular/metabolismo , Degeneração Macular/patologia , Proteínas de Membrana/genética , Camundongos , Camundongos Knockout , Mutação , Células Fotorreceptoras de Vertebrados/patologia , Retina/patologia
3.
Mol Pharmacol ; 87(4): 747-65, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25657338

RESUMO

Cannabinoid CB1 receptors (CB1Rs) mediate the presynaptic effects of endocannabinoids in the central nervous system (CNS) and most behavioral effects of exogenous cannabinoids. Cannabinoid receptor-interacting protein 1a (CRIP1a) binds to the CB1R C-terminus and can attenuate constitutive CB1R-mediated inhibition of Ca(2+) channel activity. We now demonstrate cellular colocalization of CRIP1a at neuronal elements in the CNS and show that CRIP1a inhibits both constitutive and agonist-stimulated CB1R-mediated guanine nucleotide-binding regulatory protein (G-protein) activity. Stable overexpression of CRIP1a in human embryonic kidney (HEK)-293 cells stably expressing CB1Rs (CB1-HEK), or in N18TG2 cells endogenously expressing CB1Rs, decreased CB1R-mediated G-protein activation (measured by agonist-stimulated [(35)S]GTPγS (guanylyl-5'-[O-thio]-triphosphate) binding) in both cell lines and attenuated inverse agonism by rimonabant in CB1-HEK cells. Conversely, small-interfering RNA-mediated knockdown of CRIP1a in N18TG2 cells enhanced CB1R-mediated G-protein activation. These effects were not attributable to differences in CB1R expression or endocannabinoid tone because CB1R levels did not differ between cell lines varying in CRIP1a expression, and endocannabinoid levels were undetectable (CB1-HEK) or unchanged (N18TG2) by CRIP1a overexpression. In CB1-HEK cells, 4-hour pretreatment with cannabinoid agonists downregulated CB1Rs and desensitized agonist-stimulated [(35)S]GTPγS binding. CRIP1a overexpression attenuated CB1R downregulation without altering CB1R desensitization. Finally, in cultured autaptic hippocampal neurons, CRIP1a overexpression attenuated both depolarization-induced suppression of excitation and inhibition of excitatory synaptic activity induced by exogenous application of cannabinoid but not by adenosine A1 agonists. These results confirm that CRIP1a inhibits constitutive CB1R activity and demonstrate that CRIP1a can also inhibit agonist-stimulated CB1R signaling and downregulation of CB1Rs. Thus, CRIP1a appears to act as a broad negative regulator of CB1R function.


Assuntos
Proteínas de Transporte/metabolismo , Receptor CB1 de Canabinoide/metabolismo , Animais , Proteínas de Transporte/genética , Linhagem Celular , Cerebelo/metabolismo , Endocanabinoides/metabolismo , Proteínas de Ligação ao GTP/metabolismo , Hipocampo/citologia , Hipocampo/metabolismo , Humanos , Masculino , Camundongos , Neurônios/metabolismo , Ensaio Radioligante , Ratos Sprague-Dawley , Receptor CB1 de Canabinoide/agonistas , Transdução de Sinais
4.
J Neurosci ; 33(18): 7941-51, 2013 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-23637185

RESUMO

G-protein ß subunits perform essential neuronal functions as part of G-protein ßγ and Gß5-regulators of G-protein signaling (RGS) complexes. Both Gßγ and Gß5-RGS are obligate dimers that are thought to require the assistance of the cytosolic chaperonin CCT and a cochaperone, phosducin-like protein 1 (PhLP1) for dimer formation. To test this hypothesis in vivo, we deleted the Phlp1 gene in mouse (Mus musculus) retinal rod photoreceptor cells and measured the effects on G-protein biogenesis and visual signal transduction. In the PhLP1-depleted rods, Gßγ dimer formation was decreased 50-fold, resulting in a >10-fold decrease in light sensitivity. Moreover, a 20-fold reduction in Gß5 and RGS9-1 expression was also observed, causing a 15-fold delay in the shutoff of light responses. These findings conclusively demonstrate in vivo that PhLP1 is required for the folding and assembly of both Gßγ and Gß5-RGS9.


Assuntos
Proteínas do Olho/metabolismo , Proteínas de Ligação ao GTP/metabolismo , Retina/citologia , Células Fotorreceptoras Retinianas Bastonetes/metabolismo , Transdução de Sinais/fisiologia , Animais , Fenômenos Biofísicos/genética , Sensibilidades de Contraste/genética , Relação Dose-Resposta à Radiação , Estimulação Elétrica , Eletrorretinografia , Proteínas do Olho/genética , Subunidades beta da Proteína de Ligação ao GTP/metabolismo , Subunidades gama da Proteína de Ligação ao GTP/metabolismo , Regulação da Expressão Gênica/genética , Técnicas In Vitro , Luz , Potenciais da Membrana/genética , Proteínas de Membrana/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Técnicas de Patch-Clamp , Estimulação Luminosa , RNA Mensageiro/metabolismo , Degeneração Retiniana/genética , Degeneração Retiniana/patologia , Degeneração Retiniana/fisiopatologia , Acuidade Visual/genética
5.
J Neurosci ; 32(41): 14364-73, 2012 Oct 10.
Artigo em Inglês | MEDLINE | ID: mdl-23055507

RESUMO

The rate at which photoreceptors recover from excitation is thought to be critical for setting the temporal resolution of vision. Indeed, mutations in RGS9 (regulator of G-protein signaling 9) and R9AP (RGS9 anchor protein) proteins mediating rapid photoresponse recovery impair patients' ability to see moving objects. In this study, we analyzed temporal properties of retinal sensitivity and spatiotemporal aspects of visual behavior in R9AP knock-out mice. Surprisingly, we have found that this knock-out does not affect dim-light vision mediated by rods acting as single-photon counters. Under these conditions, vision was also unaffected in mice overexpressing R9AP in rods, which causes accelerated photoresponse recovery. However, in brighter light, slow photoresponse recovery in rods and cones impaired visual responses to high temporal frequency stimuli, as reported for the daylight vision of human patients. Therefore, the speed of photoresponse recovery can affect temporal resolution and motion detection when photoreceptors integrate signals from multiple photons but not when they act as single-photon counters.


Assuntos
Adaptação Ocular/fisiologia , Proteínas de Membrana/biossíntese , Percepção de Movimento/fisiologia , Estimulação Luminosa/métodos , Células Fotorreceptoras de Vertebrados/fisiologia , Visão Ocular/fisiologia , Adaptação Ocular/genética , Animais , Feminino , Masculino , Proteínas de Membrana/deficiência , Proteínas de Membrana/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fatores de Tempo
6.
J Neurosci ; 32(45): 15998-6006, 2012 Nov 07.
Artigo em Inglês | MEDLINE | ID: mdl-23136436

RESUMO

Light isomerizes 11-cis-retinal in a retinal rod and produces an active form of rhodopsin (Rh*) that binds to the G-protein transducin and activates the phototransduction cascade. Rh* is turned off by phosphorylation by rhodopsin kinase [G-protein-coupled receptor kinase 1 (GRK1)] and subsequent binding of arrestin. To evaluate the role of GRK1 in rod light response decay, we have generated the transgenic mouse RKS561L in which GRK1, which is normally present at only 2-3% of rhodopsin, is overexpressed by ∼12-fold. Overexpression of GRK1 increases the rate of Rh* phosphorylation and reduces the exponential decay constant of the response (τ(REC)) and the limiting time constant (τ(D)) both by ∼30%; these decreases are highly significant. Similar decreases are produced in Rv(-/-) rods, in which the GRK1-binding protein recoverin has been genetically deleted. These changes in response decay are produced by acceleration of light-activated phosphodiesterase (PDE*) decay rather than Rh* decay, because light-activated PDE* decay remains rate limiting for response decay in both RKS561L and Rv(-/-) rods. A model incorporating an effect of GRK1 on light-activated PDE* decay rate can satisfactorily account for the changes in response amplitude and waveform. Modulation of response decay in background light is nearly eliminated by deletion of recoverin. Our experiments indicate that rhodopsin kinase and recoverin, in addition to their well-known role in regulating the turning off of Rh*, can also modulate the decay of light-activated PDE*, and the effects of these proteins on light-activated PDE* decay may be responsible for the quickening of response recovery in background light.


Assuntos
Receptor Quinase 1 Acoplada a Proteína G/genética , Diester Fosfórico Hidrolases/metabolismo , Recoverina/metabolismo , Células Fotorreceptoras Retinianas Bastonetes/metabolismo , Rodopsina/metabolismo , Potenciais de Ação/fisiologia , Animais , Receptor Quinase 1 Acoplada a Proteína G/metabolismo , Camundongos , Camundongos Transgênicos , Fosforilação , Estimulação Luminosa , Recoverina/genética , Transducina/metabolismo
7.
J Biol Chem ; 287(18): 14873-9, 2012 Apr 27.
Artigo em Inglês | MEDLINE | ID: mdl-22371490

RESUMO

Two members of the R7 subfamily of regulators of G protein signaling, RGS7 and RGS11, are present at dendritic tips of retinal depolarizing bipolar cells (DBCs). Their involvement in the mGluR6/Gα(o)/TRPM1 pathway that mediates DBC light responses has been implicated. However, previous genetic studies employed an RGS7 mutant mouse that is hypomorphic, and hence the exact role of RGS7 in DBCs remains unclear. We have made a true RGS7-null mouse line with exons 6-8 deleted. The RGS7(-/-) mouse is viable and fertile but smaller in body size. Electroretinogram (ERG) b-wave implicit time in young RGS7(-/-) mice is prolonged at eye opening, but the phenotype disappears at 2 months of age. Expression levels of RGS6 and RGS11 are unchanged in RGS7(-/-) retina, but the Gß5S level is significantly reduced. By characterizing a complete RGS7 and RGS11 double knock-out (711dKO) mouse line, we found that Gß5S expression in the retinal outer plexiform layer is eliminated, as is the ERG b-wave. Ultrastructural defects akin to those of Gß5(-/-) mice are evident in 711dKO mice. In retinas of mice lacking RGS6, RGS7, and RGS11, Gß5S is undetectable, whereas levels of the photoreceptor-specific Gß5L remain unchanged. Whereas RGS6 alone sustains a significant amount of Gß5S expression in retina, the DBC-related defects in Gß5(-/-) mice are caused solely by a combined loss of RGS7 and RGS11. Our data support the notion that the role of Gß5 in the retina, and likely in the entire nervous system, is mediated exclusively by R7 RGS proteins.


Assuntos
Subunidades beta da Proteína de Ligação ao GTP/biossíntese , Proteínas RGS/metabolismo , Retina/metabolismo , Animais , Subunidades beta da Proteína de Ligação ao GTP/genética , Regulação da Expressão Gênica/genética , Camundongos , Camundongos Knockout , Proteínas RGS/genética , Retina/patologia
8.
Nat Genet ; 32(2): 254-60, 2002 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-12219089

RESUMO

Excessive phototransduction signaling is thought to be involved in light-induced and inherited retinal degeneration. Using knockout mice with defects in rhodopsin shut-off and transducin signaling, we show that two different pathways of photoreceptor-cell apoptosis are induced by light. Bright light induces apoptosis that is independent of transducin and accompanied by induction of the transcription factor AP-1. By contrast, low light induces an apoptotic pathway that requires transducin. We also provide evidence that additional genetic factors regulate sensitivity to light-induced damage. Our use of defined mouse mutants resolves some of the complexity underlying the mechanisms that regulate susceptibility to retinal degeneration.


Assuntos
Apoptose , Proteínas do Olho , Luz/efeitos adversos , Retina/efeitos da radiação , Animais , Arrestina/genética , Arrestina/metabolismo , Proteínas de Transporte , Dexametasona/metabolismo , Receptor Quinase 1 Acoplada a Proteína G , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Mutação , Células Fotorreceptoras de Vertebrados/fisiologia , Proteínas Quinases/genética , Proteínas Quinases/metabolismo , Proteínas/metabolismo , Retina/metabolismo , Retina/fisiopatologia , Rodopsina/metabolismo , Transdução de Sinais , Fator de Transcrição AP-1/antagonistas & inibidores , Fator de Transcrição AP-1/metabolismo , Transducina/metabolismo , cis-trans-Isomerases
9.
JCI Insight ; 8(13)2023 07 10.
Artigo em Inglês | MEDLINE | ID: mdl-37219953

RESUMO

Mechanical, thermal, and chemical pain sensation is conveyed by primary nociceptors, a subset of sensory afferent neurons. The intracellular regulation of the primary nociceptive signal is an area of active study. We report here the discovery of a Gß5-dependent regulatory pathway within mechanical nociceptors that restrains antinociceptive input from metabotropic GABA-B receptors. In mice with conditional knockout (cKO) of the gene that encodes Gß5 (Gnb5) targeted to peripheral sensory neurons, we demonstrate the impairment of mechanical, thermal, and chemical nociception. We further report the specific loss of mechanical nociception in Rgs7-Cre+/- Gnb5fl/fl mice but not in Rgs9-Cre+/- Gnb5fl/fl mice, suggesting that Gß5 might specifically regulate mechanical pain in regulator of G protein signaling 7-positive (Rgs7+) cells. Additionally, Gß5-dependent and Rgs7-associated mechanical nociception is dependent upon GABA-B receptor signaling since both were abolished by treatment with a GABA-B receptor antagonist and since cKO of Gß5 from sensory cells or from Rgs7+ cells potentiated the analgesic effects of GABA-B agonists. Following activation by the G protein-coupled receptor Mrgprd agonist ß-alanine, enhanced sensitivity to inhibition by baclofen was observed in primary cultures of Rgs7+ sensory neurons harvested from Rgs7-Cre+/- Gnb5fl/fl mice. Taken together, these results suggest that the targeted inhibition of Gß5 function in Rgs7+ sensory neurons might provide specific relief for mechanical allodynia, including that contributing to chronic neuropathic pain, without reliance on exogenous opioids.


Assuntos
Subunidades beta da Proteína de Ligação ao GTP , Proteínas RGS , Animais , Camundongos , Receptores de GABA-B/genética , Receptores de GABA-B/metabolismo , Subunidades beta da Proteína de Ligação ao GTP/genética , Subunidades beta da Proteína de Ligação ao GTP/metabolismo , Nociceptividade , Transdução de Sinais/fisiologia , Dor , Proteínas RGS/genética , Proteínas RGS/metabolismo
10.
Circ Res ; 107(11): 1345-9, 2010 Nov 26.
Artigo em Inglês | MEDLINE | ID: mdl-20864673

RESUMO

RATIONALE: Parasympathetic regulation of heart rate is mediated by acetylcholine binding to G protein-coupled muscarinic M2 receptors, which activate heterotrimeric G(i/o) proteins to promote G protein-coupled inwardly rectifying K(+) (GIRK) channel activation. Regulator of G protein signaling (RGS) proteins, which function to inactivate G proteins, are indispensable for normal parasympathetic control of the heart. However, it is unclear which of the more than 20 known RGS proteins function to negatively regulate and thereby ensure normal parasympathetic control of the heart. OBJECTIVE: To examine the specific contribution of RGS6 as an essential regulator of parasympathetic signaling in heart. METHODS AND RESULTS: We developed RGS6 knockout mice to determine the functional impact of loss of RGS6 on parasympathetic regulation of cardiac automaticity. RGS6 exhibited a uniquely robust expression in the heart, particularly in sinoatrial and atrioventricular nodal regions. Loss of RGS6 provoked dramatically exaggerated bradycardia in response to carbachol in mice and isolated perfused hearts and significantly enhanced the effect of carbachol on inhibition of spontaneous action potential firing in sinoatrial node cells. Consistent with a role of RGS6 in G protein inactivation, RGS6-deficient atrial myocytes exhibited a significant reduction in the time course of acetylcholine-activated potassium current (I(K)(ACh)) activation and deactivation, as well as the extent of I(K)(ACh) desensitization. CONCLUSIONS: RGS6 is a previously unrecognized, but essential, regulator of parasympathetic activation in heart, functioning to prevent parasympathetic override and severe bradycardia. These effects likely result from actions of RGS6 as a negative regulator of G protein activation of GIRK channels.


Assuntos
Potenciais de Ação/fisiologia , Frequência Cardíaca/fisiologia , Coração/fisiologia , Fibras Parassimpáticas Pós-Ganglionares/fisiologia , Proteínas RGS/fisiologia , Transdução de Sinais/fisiologia , Potenciais de Ação/genética , Animais , Bradicardia/genética , Bradicardia/metabolismo , Bradicardia/fisiopatologia , Células Cultivadas , Frequência Cardíaca/genética , Camundongos , Camundongos da Linhagem 129 , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteínas RGS/deficiência , Proteínas RGS/genética , Receptor Muscarínico M2/fisiologia , Transdução de Sinais/genética , Nó Sinoatrial/fisiologia
11.
Neural Plast ; 2012: 426437, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22292123

RESUMO

The cAMP-response element-binding protein (CREB) plays an important role in visual cortical plasticity that follows the disruption of sensory activity, as induced by dark rearing (DR). Recent findings indicate that the dorsal lateral geniculate nucleus (dLGN) of thalamus is also sensitive to altered sensory activity. DR disrupts retinogeniculate synaptic strength and pruning in mice, but only when DR starts one week after eye opening (delayed DR, DDR) and not after chronic DR (CDR) from birth. While DR upregulates CREB in visual cortex, whether it also modulates this pathway in dLGN remains unknown. Here we investigate the role of CREB in the dLGN of mice that were CDR or DDR using western blot and immunofluorescence. Similar to findings in visual cortex, CREB is upregulated in dLGN after CDR and DDR. These findings are consistent with the proposal that DR up-regulates the CREB pathway in response to decreased visual drive.


Assuntos
Proteína de Ligação a CREB/fisiologia , Escuridão , Corpos Geniculados/fisiologia , Córtex Visual/fisiologia , Animais , Animais Recém-Nascidos , Adaptação à Escuridão/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Privação Sensorial/fisiologia
12.
Cells ; 11(14)2022 07 14.
Artigo em Inglês | MEDLINE | ID: mdl-35883647

RESUMO

The retina, the accessible part of the central nervous system, has served as a model system to study the relationship between energy utilization and metabolite supply. When the metabolite supply cannot match the energy demand, retinal neurons are at risk of death. As the powerhouse of eukaryotic cells, mitochondria play a pivotal role in generating ATP, produce precursors for macromolecules, maintain the redox homeostasis, and function as waste management centers for various types of metabolic intermediates. Mitochondrial dysfunction has been implicated in the pathologies of a number of degenerative retinal diseases. It is well known that photoreceptors are particularly vulnerable to mutations affecting mitochondrial function due to their high energy demand and susceptibility to oxidative stress. However, it is unclear how defective mitochondria affect other retinal neurons. Nuclear respiratory factor 1 (Nrf1) is the major transcriptional regulator of mitochondrial biogenesis, and loss of Nrf1 leads to defective mitochondria biogenesis and eventually cell death. Here, we investigated how different retinal neurons respond to the loss of Nrf1. We provide in vivo evidence that the disruption of Nrf1-mediated mitochondrial biogenesis results in a slow, progressive degeneration of all retinal cell types examined, although they present different sensitivity to the deletion of Nrf1, which implicates differential energy demand and utilization, as well as tolerance to mitochondria defects in different neuronal cells. Furthermore, transcriptome analysis on rod-specific Nrf1 deletion uncovered a previously unknown role of Nrf1 in maintaining genome stability.


Assuntos
Fator 1 Nuclear Respiratório , Neurônios Retinianos , Mitocôndrias/genética , Mitocôndrias/metabolismo , Fator 1 Nuclear Respiratório/genética , Fator 1 Nuclear Respiratório/metabolismo , Biogênese de Organelas , Retina/metabolismo , Neurônios Retinianos/metabolismo
13.
J Neurosci ; 30(4): 1213-20, 2010 Jan 27.
Artigo em Inglês | MEDLINE | ID: mdl-20107049

RESUMO

The Ca(2+)-binding protein recoverin is thought to regulate rhodopsin kinase and to modulate the lifetime of the photoexcited state of rhodopsin (Rh*), the visual pigment of vertebrate rods. Recoverin has been postulated to inhibit the kinase in darkness, when Ca(2+) is high, and to be released from the disk membrane in light when Ca(2+) is low, accelerating rhodopsin phosphorylation and shortening the lifetime of Rh*. This proposal has remained controversial, in part because the normally rapid turnoff of Rh* has made Rh* modulation difficult to study in an intact rod. To circumvent this problem, we have made mice that underexpress rhodopsin kinase so that Rh* turnoff is rate limiting for the decay of the rod light response. We show that background light speeds the decay of Rh* turnoff, and that this no longer occurs in mice that have had recoverin knocked out. This is the first demonstration in an intact rod that light accelerates Rh* inactivation and that the Ca(2+)-binding protein recoverin may be required for the light-dependent modulation of Rh* lifetime.


Assuntos
Luz , Recoverina/efeitos da radiação , Células Fotorreceptoras Retinianas Bastonetes/metabolismo , Células Fotorreceptoras Retinianas Bastonetes/efeitos da radiação , Rodopsina/efeitos da radiação , Visão Ocular/efeitos da radiação , Animais , Cálcio/metabolismo , Sinalização do Cálcio/fisiologia , Sinalização do Cálcio/efeitos da radiação , Receptor Quinase 1 Acoplada a Proteína G/metabolismo , Receptor Quinase 1 Acoplada a Proteína G/efeitos da radiação , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Estimulação Luminosa , Tempo de Reação/fisiologia , Tempo de Reação/efeitos da radiação , Recoverina/metabolismo , Rodopsina/metabolismo , Fatores de Tempo , Visão Ocular/fisiologia
14.
J Neurosci ; 30(7): 2496-503, 2010 Feb 17.
Artigo em Inglês | MEDLINE | ID: mdl-20164334

RESUMO

Rpe65(-/-) mice are unable to produce 11-cis-retinal, the chromophore of visual pigments. Consequently, the pigment is present as the apoprotein opsin with a minute level of pigment containing 9-cis-retinal as chromophore. Notably, a 10-20% fraction of this opsin is mono-phosphorylated independently of light conditions. To determine the role of rhodopsin kinase (GRK1) in phosphorylating this opsin and to test whether eliminating this phosphorylation would accelerate photoreceptor degeneration, we generated the Rpe65(-/-)Grk1(-/-) mouse. The retinae of Rpe65(-/-)Grk1(-/-) mice had negligible opsin phosphorylation, extensive degeneration with decreased opsin levels, and diminished light-evoked rod responses relative to Rpe65(-/-) mice. These data show that opsin phosphorylation in the Rpe65(-/-) mouse is due to the action of GRK1 and is neuroprotective. However, despite the higher activity of unphosphorylated opsin, the severe loss of opsin in the rapidly degenerating Rpe65(-/-)Grk1(-/-) mice resulted in lower overall opsin activity and in higher rod sensitivity compared with Rpe65(-/-) mice. In Rpe65(-/-)Grk1(-/-)Gnat1(-/-) mice where transduction activation was blocked, degeneration was only partially prevented. Therefore, increased opsin activity in the absence of phosphorylation was not the only mechanism for the accelerated retinal degeneration. Finally, the deletion of GRK1 triggered retinal degeneration in Grk1(-/-) mice after 1 month, even in the absence of apo-opsin. This degeneration was independent of light conditions and occurred even in the absence of transducin in Grk1(-/-)Gnat1(-/-) mice. Taken together, our results demonstrate a light-independent mechanism for retinal degeneration in the absence of GRK1, suggesting a second, not previously recognized role for that kinase.


Assuntos
Receptor Quinase 1 Acoplada a Proteína G/deficiência , Degeneração Retiniana/genética , Transducina/metabolismo , Visão Ocular/genética , Adaptação Ocular/genética , Animais , Biofísica/métodos , Proteínas de Transporte/genética , Proteínas do Olho/genética , Subunidades alfa de Proteínas de Ligação ao GTP/deficiência , Camundongos , Camundongos Knockout , Opsinas/metabolismo , Fosforilação/genética , Estimulação Luminosa/métodos , Degeneração Retiniana/fisiopatologia , Células Fotorreceptoras Retinianas Bastonetes/fisiologia , cis-trans-Isomerases
15.
J Neurochem ; 119(3): 544-54, 2011 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-21883221

RESUMO

Gß5 is a divergent member of the signal-transducing G protein ß subunit family encoded by GNB5 and expressed principally in brain and neuronal tissue. Among heterotrimeric Gß isoforms, Gß5 is unique in its ability to heterodimerize with members of the R7 subfamily of the regulator of G protein signaling proteins that contain G protein-γ like domains. Previous studies employing Gnb5 knockout (KO) mice have shown that Gß5 is an essential stabilizer of such regulator of G protein signaling proteins and regulates the deactivation of retinal phototransduction and the proper functioning of retinal bipolar cells. However, little is known of the function of Gß5 in the brain outside the visual system. We show here that mice lacking Gß5 have a markedly abnormal neurologic phenotype that includes impaired development, tiptoe-walking, motor learning and coordination deficiencies, and hyperactivity. We further show that Gß5-deficient mice have abnormalities of neuronal development in cerebellum and hippocampus. We find that the expression of both mRNA and protein from multiple neuronal genes is dysregulated in Gnb5 KO mice. Taken together with previous observations from Gnb5 KO mice, our findings suggest a model in which Gß5 regulates dendritic arborization and/or synapse formation during development, in part by effects on gene expression.


Assuntos
Anormalidades Múltiplas/genética , Anormalidades Múltiplas/metabolismo , Encéfalo/anormalidades , Encéfalo/crescimento & desenvolvimento , Cerebelo/anormalidades , Subunidades beta da Proteína de Ligação ao GTP/deficiência , Regulação da Expressão Gênica no Desenvolvimento/genética , Hipocampo/anormalidades , Anormalidades Múltiplas/fisiopatologia , Animais , Encéfalo/metabolismo , Cerebelo/crescimento & desenvolvimento , Cerebelo/metabolismo , Subunidades beta da Proteína de Ligação ao GTP/genética , Subunidades beta da Proteína de Ligação ao GTP/fisiologia , Hipocampo/crescimento & desenvolvimento , Hipocampo/metabolismo , Camundongos , Camundongos da Linhagem 129 , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fenótipo
16.
Proc Natl Acad Sci U S A ; 105(17): 6243-8, 2008 Apr 29.
Artigo em Inglês | MEDLINE | ID: mdl-18434540

RESUMO

Heterotrimeric G proteins relay extracellular cues from heptahelical transmembrane receptors to downstream effector molecules. Composed of an alpha subunit with intrinsic GTPase activity and a betagamma heterodimer, the trimeric complex dissociates upon receptor-mediated nucleotide exchange on the alpha subunit, enabling each component to engage downstream effector targets for either activation or inhibition as dictated in a particular pathway. To mitigate excessive effector engagement and concomitant signal transmission, the Galpha subunit's intrinsic activation timer (the rate of GTP hydrolysis) is regulated spatially and temporally by a class of GTPase accelerating proteins (GAPs) known as the regulator of G protein signaling (RGS) family. The array of G protein-coupled receptors, Galpha subunits, RGS proteins and downstream effectors in mammalian systems is vast. Understanding the molecular determinants of specificity is critical for a comprehensive mapping of the G protein system. Here, we present the 2.9 A crystal structure of the enigmatic, neuronal G protein Galpha(o) in the GTP hydrolytic transition state, complexed with RGS16. Comparison with the 1.89 A structure of apo-RGS16, also presented here, reveals plasticity upon Galpha(o) binding, the determinants for GAP activity, and the structurally unique features of Galpha(o) that likely distinguish it physiologically from other members of the larger Galpha(i) family, affording insight to receptor, GAP and effector specificity.


Assuntos
Subunidades alfa Gi-Go de Proteínas de Ligação ao GTP/química , Proteínas RGS/química , Animais , Camundongos , Estrutura Secundária de Proteína , Estrutura Terciária de Proteína
17.
J Comp Neurol ; 529(15): 3513-3532, 2021 10.
Artigo em Inglês | MEDLINE | ID: mdl-34245014

RESUMO

The mammalian retina contains more than 40 retinal ganglion cell (RGC) subtypes based on their unique morphologies, functions, and molecular profiles. Among them, intrinsically photosensitive RGCs (ipRGCs) are the first specified RGC type emerging from a common retinal progenitor pool during development. Previous work has shown that T-box transcription factor T-brain 2 (Tbr2) is essential for the formation and maintenance of ipRGCs, and that Tbr2-expressing RGCs activate Opn4 expression upon native ipRGC ablation, suggesting that Tbr2+ RGCs contain a reservoir for ipRGCs. However, the identity of Tbr2+ RGCs has not been fully vetted. Here, using genetic sparse labeling and single cell recording, we showed that Tbr2-expressing retinal neurons include RGCs and a subset of GABAergic displaced amacrine cells (dACs). Most Tbr2+ RGCs are intrinsically photosensitive and morphologically resemble native ipRGCs with identical retinofugal projections. Tbr2+ RGCs also include a unique and rare Pou4f1-expressing OFF RGC subtype. Using a loss-of-function strategy, we have further demonstrated that Tbr2 is essential for the survival of these RGCs and dACs, as well as maintaining the expression of Opn4. These data set a strong foundation to study how Tbr2 regulates ipRGC development and survival, as well as the expression of molecular machinery regulating intrinsic photosensitivity.


Assuntos
Células Ganglionares da Retina/metabolismo , Proteínas com Domínio T/biossíntese , Proteínas com Domínio T/genética , Animais , Dendritos/química , Dendritos/metabolismo , Feminino , Expressão Gênica , Masculino , Camundongos , Camundongos da Linhagem 129 , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Células Ganglionares da Retina/química , Proteínas com Domínio T/análise
18.
Neuron ; 51(4): 409-16, 2006 Aug 17.
Artigo em Inglês | MEDLINE | ID: mdl-16908407

RESUMO

Signaling through G protein-coupled receptors (GPCRs) underlies many cellular processes, yet it is not known which molecules determine the duration of signaling in intact cells. Two candidates are G protein-coupled receptor kinases (GRKs) and Regulators of G protein signaling (RGSs), deactivation enzymes for GPCRs and G proteins, respectively. Here we investigate whether GRK or RGS governs the overall rate of recovery of the light response in mammalian rod photoreceptors, a model system for studying GPCR signaling. We show that overexpression of rhodopsin kinase (GRK1) increases phosphorylation of the GPCR rhodopsin but has no effect on photoresponse recovery. In contrast, overexpression of the photoreceptor RGS complex (RGS9-1.Gbeta5L.R9AP) dramatically accelerates response recovery. Our results show that G protein deactivation is normally at least 2.5 times slower than rhodopsin deactivation, resolving a long-standing controversy concerning the mechanism underlying the recovery of rod visual transduction.


Assuntos
Expressão Gênica/fisiologia , Proteínas RGS/metabolismo , Recuperação de Função Fisiológica/fisiologia , Células Fotorreceptoras Retinianas Bastonetes/fisiologia , Animais , Western Blotting/métodos , Receptor Quinase 1 Acoplada a Proteína G/genética , Receptor Quinase 1 Acoplada a Proteína G/metabolismo , Subunidades beta da Proteína de Ligação ao GTP/genética , Subunidades beta da Proteína de Ligação ao GTP/metabolismo , Expressão Gênica/efeitos da radiação , Técnicas In Vitro , Luz , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Fosforilação/efeitos da radiação , Estimulação Luminosa/métodos , Proteínas RGS/genética , Recuperação de Função Fisiológica/genética , Retina/citologia , Fatores de Tempo , Visão Ocular/fisiologia
19.
J Neurosci ; 29(45): 14287-98, 2009 Nov 11.
Artigo em Inglês | MEDLINE | ID: mdl-19906976

RESUMO

Heterotrimeric kinesin-II is a molecular motor localized to the inner segment, connecting cilium and axoneme of mammalian photoreceptors. Our purpose was to identify the role of kinesin-II in anterograde intraflagellar transport by photoreceptor-specific deletions of kinesin family member 3A (KIF3A), its obligatory motor subunit. In cones lacking KIF3A, membrane proteins involved in phototransduction did not traffic to the outer segments resulting in complete absence of a photopic electroretinogram and progressive cone degeneration. Rod photoreceptors lacking KIF3A degenerated rapidly between 2 and 4 weeks postnatally, but the phototransduction components including rhodopsin trafficked to the outer segments during the course of degeneration. Furthermore, KIF3A deletion did not affect synaptic anterograde trafficking. The results indicate that trafficking of membrane proteins to the outer segment is dependent on kinesin-II in cone, but not rod photoreceptors, even though rods and cones share similar structures, and closely related phototransduction polypeptides.


Assuntos
Cinesinas/metabolismo , Proteínas de Membrana/metabolismo , Proteínas Associadas aos Microtúbulos/metabolismo , Células Fotorreceptoras Retinianas Cones/fisiologia , Células Fotorreceptoras Retinianas Bastonetes/fisiologia , Animais , Cinesinas/genética , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Transporte Proteico/fisiologia , Retina/crescimento & desenvolvimento , Retina/fisiopatologia , Retina/ultraestrutura , Células Fotorreceptoras Retinianas Cones/ultraestrutura , Degeneração Retiniana/fisiopatologia , Pigmentos da Retina/metabolismo , Rodopsina/metabolismo , Sinapses/fisiologia , Fatores de Tempo
20.
Invest Ophthalmol Vis Sci ; 60(10): 3584-3594, 2019 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-31425583

RESUMO

Purpose: Heterozygous mutations in the essential X-linked gene CASK associate with optic nerve hypoplasia (ONH) and other retinal disorders in girls. CASK+/- heterozygous knockout mice with mosaic CASK expression exhibit ONH with a loss of retinal ganglion cells (RGCs) but no changes in retinal morphology. It remains unclear if CASK deficiency selectively affects RGCs or also affects other retinal cells. Furthermore, it is not known if CASK expression in RGCs is critical for optic nerve (ON) development and maintenance. Methods: The visual behavior of CASK+/- mice was assessed and electroretinography (ERG) was performed. Using a mouse line with a floxed CASK gene that expresses approximately 40% CASK globally in all cells (hypomorph) under hemizygous and homozygous conditions, we investigated effects of CASK reduction on the retina and ON. CASK then was completely deleted from RGCs to examine its cell-autonomous role. Finally, for the first time to our knowledge, we describe a hemizygous CASK missense mutation in a boy with ONH. Results: CASK+/- heterozygous mutant mice display reduced visual contrast sensitivity, but ERG is indistinguishable from wildtype. CASK hypomorph mice exhibit ONH, but deletion of CASK from RGCs in this background does not exacerbate the condition. The boy with ONH harbors a missense mutation (p.Pro673Leu) that destabilizes CASK and weakens the crucial CASK-neurexin interaction. Conclusions: Our results demonstrate that mosaic or global reduction in CASK expression and/or function disproportionately affects RGCs. CASK expression in RGCs does not appear critical for cell survival, indicating a noncell autonomous role for CASK in the development of ON.


Assuntos
Regulação Enzimológica da Expressão Gênica/fisiologia , Guanilato Quinases/genética , Hipoplasia do Nervo Óptico/genética , Animais , Sobrevivência Celular , Pré-Escolar , Sensibilidades de Contraste/fisiologia , Eletrorretinografia , Feminino , Humanos , Immunoblotting , Imuno-Histoquímica , Hibridização In Situ , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Mutação de Sentido Incorreto , Hipoplasia do Nervo Óptico/fisiopatologia , Retina/fisiopatologia , Células Ganglionares da Retina/enzimologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA