Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 20
Filtrar
1.
J Biol Chem ; 299(12): 105412, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-37918805

RESUMEN

A major unsolved question in vertebrate photoreceptor biology is the mechanism of rhodopsin transport to the outer segment. In rhodopsin-like class A G protein-coupled receptors, hydrophobic interactions between C-terminal α-helix 8 (H8), and transmembrane α-helix-1 (TM1) have been shown to be important for transport to the plasma membrane, however whether this interaction is important for rhodopsin transport to ciliary rod outer segments is not known. We examined the crystal structures of vertebrate rhodopsins and class A G protein-coupled receptors and found a conserved network of predicted hydrophobic interactions. In Xenopus rhodopsin (xRho), this interaction corresponds to F313, L317, and L321 in H8 and M57, V61, and L68 in TM1. To evaluate the role of H8-TM1 hydrophobic interactions in rhodopsin transport, we expressed xRho-EGFP where hydrophobic residues were mutated in Xenopus rods and evaluated the efficiency of outer segment enrichment. We found that substituting L317 and M57 with hydrophilic residues had the strongest impact on xRho mislocalization. Substituting hydrophilic amino acids at positions L68, F313, and L321 also had a significant impact. Replacing L317 with M resulted in significant mislocalization, indicating that the hydrophobic interaction between residues 317 and 57 is exquisitely sensitive. The corresponding experiment in bovine rhodopsin expressed in HEK293 cells had a similar effect, showing that the H8-TM1 hydrophobic network is essential for rhodopsin transport in mammalian species. Thus, for the first time, we show that a hydrophobic interaction between H8 and TM1 is critical for efficient rhodopsin transport to the vertebrate photoreceptor ciliary outer segment.


Asunto(s)
Células Fotorreceptoras Retinianas Bastones , Rodopsina , Animales , Bovinos , Humanos , Células HEK293 , Interacciones Hidrofóbicas e Hidrofílicas , Receptores Acoplados a Proteínas G/metabolismo , Células Fotorreceptoras Retinianas Bastones/metabolismo , Rodopsina/genética , Rodopsina/química , Segmento Externo de la Célula en Bastón/metabolismo , Vertebrados
2.
Pflugers Arch ; 473(9): 1493-1516, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-33880652

RESUMEN

Retinal photoreceptors are neurons that convert dynamically changing patterns of light into electrical signals that are processed by retinal interneurons and ultimately transmitted to vision centers in the brain. They represent the essential first step in seeing without which the remainder of the visual system is rendered moot. To support this role, the major functions of photoreceptors are segregated into three main specialized compartments-the outer segment, the inner segment, and the pre-synaptic terminal. This compartmentalization is crucial for photoreceptor function-disruption leads to devastating blinding diseases for which therapies remain elusive. In this review, we examine the current understanding of the molecular and physical mechanisms underlying photoreceptor functional compartmentalization and highlight areas where significant knowledge gaps remain.


Asunto(s)
Membrana Celular/metabolismo , Terminales Presinápticos/metabolismo , Neuronas Retinianas/metabolismo , Segmento Interno de las Células Fotorreceptoras Retinianas/metabolismo , Segmento Externo de las Células Fotorreceptoras Retinianas/metabolismo , Animales , Humanos , Células Fotorreceptoras de Vertebrados/metabolismo , Transporte de Proteínas/fisiología
3.
Proc Natl Acad Sci U S A ; 109(1): 203-8, 2012 Jan 03.
Artículo en Inglés | MEDLINE | ID: mdl-22184246

RESUMEN

Proteins segregate into discrete subcellular compartments via a variety of mechanisms, including motor protein transport, local binding, and diffusion barriers. This physical separation of cell functions serves, in part, as a mechanism for controlling compartment activity by allowing regulation of local protein concentrations. In this study we explored how soluble protein size impacts access to the confined space within the retinal photoreceptor outer segment signaling compartment and discovered a strikingly steep relationship. We find that GFP monomers, dimers, and trimers expressed transgenically in frog rods are present in the outer segment at 1.8-, 2.9-, and 6.8-fold lower abundances, relative to the cell body, than the small soluble fluorescent marker, calcein. Theoretical analysis, based on statistical-mechanical models of molecular access to polymer meshes, shows that these observations can be explained by the steric hindrance of molecules occupying the highly constrained spaces between outer segment disc membranes. This mechanism may answer a long-standing question of how the soluble regulatory protein, arrestin, is effectively excluded from the outer segments of dark-adapted rods and cones. Generally, our results suggest an alternate mode for the control of protein access to cell domains based on dynamic, size-dependent compartmental partitioning that does not require diffusion barriers, active transport, or large numbers of immobile binding sites.


Asunto(s)
Cilios/metabolismo , Proteínas Fluorescentes Verdes/metabolismo , Células Fotorreceptoras Retinianas Bastones/ultraestructura , Animales , Compartimento Celular , Citoplasma/metabolismo , Modelos Biológicos , Células Fotorreceptoras Retinianas Bastones/citología , Células Fotorreceptoras Retinianas Bastones/metabolismo , Solubilidad , Xenopus laevis
4.
Front Cell Dev Biol ; 9: 636737, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33614665

RESUMEN

Functional compartmentalization of cells is a universal strategy for segregating processes that require specific components, undergo regulation by modulating concentrations of those components, or that would be detrimental to other processes. Primary cilia are hair-like organelles that project from the apical plasma membranes of epithelial cells where they serve as exclusive compartments for sensing physical and chemical signals in the environment. As such, molecules involved in signal transduction are enriched within cilia and regulating their ciliary concentrations allows adaptation to the environmental stimuli. The highly efficient organization of primary cilia has been co-opted by major sensory neurons, olfactory cells and the photoreceptor neurons that underlie vision. The mechanisms underlying compartmentalization of cilia are an area of intense current research. Recent findings have revealed similarities and differences in molecular mechanisms of ciliary protein enrichment and its regulation among primary cilia and sensory cilia. Here we discuss the physiological demands on photoreceptors that have driven their evolution into neurons that rely on a highly specialized cilium for signaling changes in light intensity. We explore what is known and what is not known about how that specialization appears to have driven unique mechanisms for photoreceptor protein and membrane compartmentalization.

5.
J Cell Biol ; 218(11): 3753-3772, 2019 11 04.
Artículo en Inglés | MEDLINE | ID: mdl-31594805

RESUMEN

In neurons, peripheral membrane proteins are enriched in subcellular compartments, where they play key roles, including transducing and transmitting information. However, little is known about the mechanisms underlying their compartmentalization. To explore the roles of hydrophobic and electrostatic interactions, we engineered probes consisting of lipidation motifs attached to fluorescent proteins by variously charged linkers and expressed them in Xenopus rod photoreceptors. Quantitative live cell imaging showed dramatic differences in distributions and dynamics of the probes, including presynapse and ciliary OS enrichment, depending on lipid moiety and protein surface charge. Opposing extant models of ciliary enrichment, most probes were weakly membrane bound and diffused through the connecting cilium without lipid binding chaperone protein interactions. A diffusion-binding-transport model showed that ciliary enrichment of a rhodopsin kinase probe occurs via recycling as it perpetually leaks out of the ciliary OS. The model accounts for weak membrane binding of peripheral membrane proteins and a leaky connecting cilium diffusion barrier.


Asunto(s)
Proteínas de la Membrana/metabolismo , Células Fotorreceptoras de Invertebrados/metabolismo , Animales , Xenopus laevis
6.
J Cell Biol ; 217(8): 2831-2849, 2018 08 06.
Artículo en Inglés | MEDLINE | ID: mdl-29945903

RESUMEN

Physical properties of primary cilia membranes in living cells were examined using two independent, high-spatiotemporal-resolution approaches: fast tracking of single quantum dot-labeled G protein-coupled receptors and a novel two-photon super-resolution fluorescence recovery after photobleaching of protein ensemble. Both approaches demonstrated the cilium membrane to be partitioned into corralled domains spanning 274 ± 20 nm, within which the receptors are transiently confined for 0.71 ± 0.09 s. The mean membrane diffusion coefficient within the corrals, Dm1 = 2.9 ± 0.41 µm2/s, showed that the ciliary membranes were among the most fluid encountered. At longer times, the apparent membrane diffusion coefficient, Dm2 = 0.23 ± 0.05 µm2/s, showed that corral boundaries impeded receptor diffusion 13-fold. Mathematical simulations predict the probability of G protein-coupled receptors crossing corral boundaries to be 1 in 472. Remarkably, latrunculin A, cytochalasin D, and jasplakinolide treatments altered the corral permeability. Ciliary membranes are thus partitioned into highly fluid membrane nanodomains that are delimited by filamentous actin.


Asunto(s)
Citoesqueleto de Actina/fisiología , Cilios/metabolismo , Citoesqueleto de Actina/metabolismo , Animales , Línea Celular , Membrana Celular/metabolismo , Membrana Celular/ultraestructura , Cilios/ultraestructura , Ratones , Puntos Cuánticos/análisis
7.
J Gen Physiol ; 128(4): 473-85, 2006 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-17001087

RESUMEN

In vertebrate rods, photoisomerization of the 11-cis retinal chromophore of rhodopsin to the all-trans conformation initiates a biochemical cascade that closes cGMP-gated channels and hyperpolarizes the cell. All-trans retinal is reduced to retinol and then removed to the pigment epithelium. The pigment epithelium supplies fresh 11-cis retinal to regenerate rhodopsin. The recent discovery that tens of nanomolar retinal inhibits cloned cGMP-gated channels at low [cGMP] raised the question of whether retinoid traffic across the plasma membrane of the rod might participate in the signaling of light. Native channels in excised patches from rods were very sensitive to retinoid inhibition. Perfusion of intact rods with exogenous 9- or 11-cis retinal closed cGMP-gated channels but required higher than expected concentrations. Channels reopened after perfusing the rod with cellular retinoid binding protein II. PDE activity, flash response kinetics, and relative sensitivity were unchanged, ruling out pharmacological activation of the phototransduction cascade. Bleaching of rhodopsin to create all-trans retinal and retinol inside the rod did not produce any measurable channel inhibition. Exposure of a bleached rod to 9- or 11-cis retinal did not elicit channel inhibition during the period of rhodopsin regeneration. Microspectrophotometric measurements showed that exogenous 9- or 11-cis retinal rapidly cross the plasma membrane of bleached rods and regenerate their rhodopsin. Although dark-adapted rods could also take up large quantities of 9-cis retinal, which they converted to retinol, the time course was slow. Apparently cGMP-gated channels in intact rods are protected from the inhibitory effects of retinoids that cross the plasma membrane by a large-capacity buffer. Opsin, with its chromophore binding pocket occupied (rhodopsin) or vacant, may be an important component. Exceptionally high retinoid levels, e.g., associated with some retinal degenerations, could overcome the buffer, however, and impair sensitivity or delay the recovery after exposure to bright light.


Asunto(s)
Canales Iónicos/fisiología , Células Fotorreceptoras Retinianas Bastones/fisiología , Retinoides/farmacología , 1-Metil-3-Isobutilxantina/farmacología , 3',5'-GMP Cíclico Fosfodiesterasas/metabolismo , Ambystoma , Animales , GMP Cíclico/biosíntesis , Canales Catiónicos Regulados por Nucleótidos Cíclicos , Diterpenos , Guanilato Ciclasa/metabolismo , Canales Iónicos/antagonistas & inhibidores , Luz , Microespectrofotometría , Técnicas de Placa-Clamp , Células Fotorreceptoras Retinianas Bastones/efectos de los fármacos , Células Fotorreceptoras Retinianas Bastones/efectos de la radiación , Retinaldehído/metabolismo , Retinaldehído/farmacología , Retinoides/metabolismo , Proteínas de Unión al Retinol/farmacología , Proteínas Plasmáticas de Unión al Retinol , Rodopsina/metabolismo , Segmento Externo de la Célula en Bastón/metabolismo , Vitamina A/farmacología
8.
Mol Biol Cell ; 28(4): 554-566, 2017 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-27974638

RESUMEN

Resolution limitations of optical systems are major obstacles for determining whether proteins are enriched within cell compartments. Here we use an approach to determine the degree of membrane protein ciliary enrichment that quantitatively accounts for the differences in sampling of the ciliary and apical membranes inherent to confocal microscopes. Theory shows that cilia will appear more than threefold brighter than the surrounding apical membrane when the densities of fluorescently labeled proteins are the same, thus providing a benchmark for ciliary enrichment. Using this benchmark, we examined the ciliary enrichment signals of two G protein-coupled receptors (GPCRs)-the somatostatin receptor 3 and rhodopsin. Remarkably, we found that the C-terminal VxPx motif, required for efficient enrichment of rhodopsin within rod photoreceptor sensory cilia, inhibited enrichment of the somatostatin receptor in primary cilia. Similarly, VxPx inhibited primary cilium enrichment of a chimera of rhodopsin and somatostatin receptor 3, where the dual Ax(S/A)xQ ciliary targeting motifs within the third intracellular loop of the somatostatin receptor replaced the third intracellular loop of rhodopsin. Rhodopsin was depleted from primary cilia but gained access, without being enriched, with the dual Ax(S/A)xQ motifs. Ciliary enrichment of these GPCRs thus operates via distinct mechanisms in different cells.


Asunto(s)
Receptores de Somatostatina/metabolismo , Células Fotorreceptoras Retinianas Bastones/metabolismo , Rodopsina/metabolismo , Secuencia de Aminoácidos , Animales , Bovinos , Técnicas de Cultivo de Célula/métodos , Cilios/metabolismo , Fluorescencia , Humanos , Microscopía Fluorescente/métodos , Unión Proteica , Receptores Acoplados a Proteínas G/metabolismo , Células Fotorreceptoras Retinianas Bastones/fisiología
9.
Invest Ophthalmol Vis Sci ; 47(5): 2185-94, 2006 May.
Artículo en Inglés | MEDLINE | ID: mdl-16639031

RESUMEN

PURPOSE: To investigate the impact of aryl hydrocarbon receptor-interacting protein-like (AIPL)-1 on photoreception in rods. METHODS: Photoresponses of mouse rods expressing lowered amounts of AIPL1 were studied by single-cell and electroretinogram (ERG) recordings. Phototransduction protein levels and enzymatic activities were determined in biochemical assays. Ca2+ dynamics were probed with a fluorescent dye. Comparisons were made to rods expressing mutant Y99C guanylate cyclase activating protein (GCAP)-1, to understand which effects arose from elevated dark levels of cGMP and Ca2+. RESULTS: Except for PDE, transduction protein levels were normal in low-AIPL1 retinas, as were guanylate cyclase (GC), rhodopsin kinase (RK), and normalized phosphodiesterase (PDE) activities. Y99C and low-AIPL1 rods were more sensitive to flashes than normal, but flash responses of low-AIPL1 rods showed an abnormal delay, reduced rate of increase, and longer recovery not present in Y99C rod responses. In addition, low-AIPL1 rods but not Y99C rods failed to reach the normal light-induced minimum in Ca2+ concentration. CONCLUSIONS: Reduced AIPL1 delayed the photoresponse, decreased its amplification constant, slowed a rate-limiting step in its recovery, and limited the light-induced decrease in Ca2+. Not all changes were attributable to decreased PDE or to elevated cGMP and Ca2+ in darkness. Therefore, AIPL1 directly or indirectly affects more than one component of phototransduction.


Asunto(s)
Proteínas Portadoras/fisiología , Células Fotorreceptoras Retinianas Bastones/fisiología , Visión Ocular/fisiología , Proteínas Adaptadoras Transductoras de Señales , Animales , Calcio/metabolismo , Cromatografía de Gases , GMP Cíclico/metabolismo , Electrorretinografía , Ácidos Grasos/metabolismo , Quinasa 1 del Receptor Acoplado a Proteína-G/metabolismo , Guanilato Ciclasa/metabolismo , Proteínas Activadoras de la Guanilato-Ciclasa/metabolismo , Ratones , Ratones Transgénicos , Hidrolasas Diéster Fosfóricas/metabolismo , Estimulación Luminosa , Células Fotorreceptoras Retinianas Bastones/efectos de la radiación , Transducina/metabolismo
10.
J Neurosci ; 24(27): 6078-85, 2004 Jul 07.
Artículo en Inglés | MEDLINE | ID: mdl-15240799

RESUMEN

Guanylyl cyclase-activating proteins (GCAPs) are Ca2+-binding proteins that activate guanylyl cyclase when free Ca2+ concentrations in retinal rods and cones fall after illumination and inhibit the cyclase when free Ca2+ reaches its resting level in the dark. Several forms of retinal dystrophy are caused by mutations in GUCA1A, the gene coding for GCAP1. To investigate the cellular mechanisms affected by the diseased state, we created transgenic mice that express GCAP1 with a Tyr99Cys substitution (Y99C GCAP1) found in human patients with a late-onset retinal dystrophy (Payne et al., 1998). Y99C GCAP1 shifted the Ca2+ sensitivity of the guanylyl cyclase in photoreceptors, keeping it partially active at 250 nM free Ca2+, the normal resting Ca2+ concentration in darkness. The enhanced activity of the cyclase in the dark increased cyclic nucleotide-gated channel activity and elevated the rod outer segment Ca2+ concentration in darkness, measured by using fluo-5F and laser spot microscopy. In different lines of transgenic mice the magnitude of this effect rose with the Y99C GCAP1 expression. Surprisingly, there was little change in the rod photoresponse, indicating that dynamic Ca2+-dependent regulation of cGMP synthesis was preserved. However, the photoreceptors in these mice degenerated, and the rate of the cell loss increased with the level of the transgene expression, unlike in transgenic mice that overexpressed normal GCAP1. These results provide the first direct evidence that a mutation linked to congenital blindness increases Ca2+ in the outer segment, which may trigger the apoptotic process.


Asunto(s)
Proteínas de Unión al Calcio/genética , Calcio/metabolismo , Líquido Intracelular/metabolismo , Células Fotorreceptoras de Vertebrados/metabolismo , Degeneración Retiniana/genética , Sustitución de Aminoácidos , Animales , Apoptosis/genética , Proteínas de Unión al Calcio/metabolismo , Bovinos , Separación Celular , Adaptación a la Oscuridad/genética , Adaptación a la Oscuridad/fisiología , Electrofisiología , Electrorretinografía , Guanilato Ciclasa/metabolismo , Proteínas Activadoras de la Guanilato-Ciclasa , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Mutación Missense , Estimulación Luminosa , Células Fotorreceptoras de Vertebrados/patología , Receptores de Superficie Celular/metabolismo , Retina/química , Retina/metabolismo , Retina/patología , Degeneración Retiniana/metabolismo , Degeneración Retiniana/patología
11.
J Gen Physiol ; 119(2): 129-45, 2002 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-11815664

RESUMEN

Vertebrate rod photoreceptors adjust their sensitivity as they adapt during exposure to steady light. Light adaptation prevents the rod from saturating and significantly extends its dynamic range. We examined the time course of the onset of light adaptation in bullfrog rods and compared it with the projected onset of feedback reactions thought to underlie light adaptation on the molecular level. We found that adaptation developed in two distinct temporal phases: (1) a fast phase that operated within seconds after the onset of illumination, which is consistent with most previous reports of a 1-2-s time constant for the onset of adaptation; and (2) a slow phase that engaged over tens of seconds of continuous illumination. The fast phase desensitized the rods as much as 80-fold, and was observed at every light intensity tested. The slow phase was observed only at light intensities that suppressed more than half of the dark current. It provided an additional sensitivity loss of up to 40-fold before the rod saturated. Thus, rods achieved a total degree of adaptation of approximately 3,000-fold. Although the fast adaptation is likely to originate from the well characterized Ca(2+)-dependent feedback mechanisms regulating the activities of several phototransduction cascade components, the molecular mechanism underlying slow adaptation is unclear. We tested the hypothesis that the slow adaptation phase is mediated by cGMP dissociation from noncatalytic binding sites on the cGMP phosphodiesterase, which has been shown to reduce the lifetime of activated phosphodiesterase in vitro. Although cGMP dissociated from the noncatalytic binding sites in intact rods with kinetics approximating that for the slow adaptation phase, this hypothesis was ruled out because the intensity of light required for cGMP dissociation far exceeded that required to evoke the slow phase. Other possible mechanisms are discussed.


Asunto(s)
Adaptación Ocular/fisiología , Células Fotorreceptoras Retinianas Bastones/fisiología , 3',5'-GMP Cíclico Fosfodiesterasas/metabolismo , Animales , Técnicas In Vitro , Cinética , Luz , Modelos Biológicos , Rana catesbeiana , Células Fotorreceptoras Retinianas Bastones/enzimología , Factores de Tiempo
12.
Mol Vis ; 11: 1236-45, 2005 Dec 30.
Artículo en Inglés | MEDLINE | ID: mdl-16402024

RESUMEN

PURPOSE: The concentration of enhanced green fluorescent protein (EGFP) in individual photoreceptor cells of live mouse retina was quantified and correlated with physiological measurements of cell function. METHODS: EGFP protein levels in the retinas of mice injected subretinally by either one of two serotypes of adeno-associated virus (AAV; AAV2/5.CMV.EGFP; AAV2/2.CMV.EGFP) were quantified with a photon-counting confocal laser scanning microscope and compared with those of transgenic mice whose retinas expressed EGFP under the beta-actin (pbetaAct) or human L/M-cone opsin (pLMCOps) promoter. Single-cell suction pipette recordings of single rods and whole-field electroretinograms (ERGs) were performed to assess retinal cell function. RESULTS: The highest levels of EGFP (680 microM) were in the retinal pigment epithelium (RPE) cells of the AAV-transduced eyes. Living photoreceptors of pbetaAct.EGFP mice contained 270 microM EGFP, while their bipolars had 440 microM. The cones of pLMCOps.EGFP mice expressed 60 microM protein. The amplitudes of the major components of ERGs were within the normal range for all transgenic animals examined, and single cell recordings from living pbetaAct.EGFP rods were indistinguishable from those of controls. CONCLUSIONS: EGFP levels in individual cells of live mouse retinas can be quantified, so that the efficacy of gene transfer methods can be quantified. Concentrations of several hundred microM are not deleterious to normal function of photoreceptors and bipolar cells. This approach can also be used to quantify levels of biologically active EGFP fusion proteins.


Asunto(s)
Técnicas de Transferencia de Gen , Proteínas Fluorescentes Verdes/farmacocinética , Proteínas Fluorescentes Verdes/envenenamiento , Retina/efectos de los fármacos , Retina/metabolismo , Animales , Dependovirus/genética , Electrofisiología , Electrorretinografía , Embrión de Mamíferos/metabolismo , Vectores Genéticos , Proteínas Fluorescentes Verdes/administración & dosificación , Inyecciones , Ratones , Ratones Endogámicos C57BL , Microscopía Confocal , Concentración Osmolar , Estimulación Luminosa , Células Fotorreceptoras de Vertebrados/metabolismo , Retina/citología , Retina/fisiología , Células Bipolares de la Retina/metabolismo , Distribución Tisular
13.
Methods Mol Biol ; 1271: 309-23, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25697532

RESUMEN

High-resolution multiphoton imaging of live cells has become an invaluable method to study protein dynamics in highly compartmentalized subcellular environments. Here we describe procedures that we recently developed to quantify rhodopsin mobility within and between retinal rod photoreceptor light signaling microcompartments, the disc membrane lobules, using multiphoton fluorescence relaxation after photoconversion.


Asunto(s)
Células Fotorreceptoras/metabolismo , Rodopsina/química , Rodopsina/metabolismo , Receptores Acoplados a Proteínas G/química , Receptores Acoplados a Proteínas G/metabolismo , Transducción de Señal/fisiología
14.
Adv Exp Med Biol ; 514: 37-60, 2002.
Artículo en Inglés | MEDLINE | ID: mdl-12596914

RESUMEN

The photoresponse of a rod wanes over time in steady illumination, as light loses its efficacy in generating the response. Such desensitization is adaptive because it extends the range of ambient light levels over which the rod signals changes in light intensity by several orders of magnitude. Adaptation begins to unfold rapidly after the onset of light with a time constant of approximately 1 s, causing the rod's sensitivity to steady light to decrease by nearly two log units. Thereafter, a much slower phase of adaptation evolves with a time constant of 9 s. During this phase the rod's sensitivity decreases by an additional log unit. Both phases are dependent upon the light-induced fall in intracellular Ca2+. The fast phase of light adaptation can be attributed to Ca2+ feedback processes regulating the lifetime ofphotoactivated rhodopsin, cGMP synthesis and sensitivity of the cGMP-gated channel to cGMP. Although the mechanism(s) of the slow phase is not yet known, it appears to include further regulation of the lifetime of photoactivated rhodopsin.


Asunto(s)
Luz , Células Fotorreceptoras Retinianas Bastones/fisiología , Adaptación Biológica , Animales , Calcio/metabolismo , Modelos Biológicos , Fosforilación , Células Fotorreceptoras Retinianas Bastones/metabolismo , Rodopsina/metabolismo , Factores de Tiempo
15.
PLoS One ; 8(11): e80059, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24260336

RESUMEN

The rod outer segment (OS), comprised of tightly stacked disk membranes packed with rhodopsin, is in a dynamic equilibrium governed by a diurnal rhythm with newly synthesized membrane inserted at the OS base balancing membrane loss from the distal tip via disk shedding. Using transgenic Xenopus and live cell confocal imaging, we found OS axial variation of fluorescence intensity in cells expressing a fluorescently tagged rhodopsin transgene. There was a light synchronized fluctuation in intensity, with higher intensity in disks formed at night and lower intensity for those formed during the day. This fluctuation was absent in constant light or dark conditions. There was also a slow modulation of the overall expression level that was not synchronized with the lighting cycle or between cells in the same retina. The axial variations of other membrane-associated fluorescent proteins, eGFP-containing two geranylgeranyl acceptor sites and eGFP fused to the transmembrane domain of syntaxin, were greatly reduced or not detectable, respectively. In acutely light-adapted rods, an arrestin-eGFP fusion protein also exhibited axial variation. Both the light-sensitive Rho-eGFP and arrestin-eGFP banding were in phase with the previously characterized birefringence banding (Kaplan, Invest. Ophthalmol. Vis. Sci. 21, 395-402 1981). In contrast, endogenous rhodopsin did not exhibit such axial variation. Thus, there is an axial inhomogeneity in membrane composition or structure, detectable by the rhodopsin transgene density distribution and regulated by the light cycle, implying a light-regulated step for disk assembly in the OS. The impact of these results on the use of chimeric proteins with rhodopsin fused to fluorescent proteins at the carboxyl terminus is discussed.


Asunto(s)
Proteínas Fluorescentes Verdes/metabolismo , Rodopsina/metabolismo , Segmento Externo de la Célula en Bastón/metabolismo , Xenopus/metabolismo , Animales , Arrestina/metabolismo , Birrefringencia , Relojes Circadianos/fisiología , Luz , Proteínas Qa-SNARE/metabolismo , Retina/metabolismo
16.
Vision Res ; 75: 11-8, 2012 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-22922002

RESUMEN

Most cells in the human body elaborate cilia which serve a wide variety of functions, including cell and tissue differentiation during development, sensing physical and chemical properties of the extracellular milieu and mechanical force generation. Common among cilia is the transduction of external stimuli into signals that regulate the activities of the cilia and the cells that possess them. These functions require the transport and localization of specialized proteins to the cilium, a process that many recent studies have shown to be vital for normal cell function and, ultimately, the health of the organism. Here we discuss several mechanisms proposed for the transport and localization of soluble and peripheral membrane proteins to, or their exclusion from the ciliary compartment with a focus on how the structure of the cytoplasm and the size and shape of proteins influence these processes. Additionally, we examine the impact of cell and protein structure on our ability to accurately measure the relative concentrations of fluorescently tagged proteins amongst various cellular domains, which is integral to our understanding of the molecular mechanisms underlying protein localization and transport.


Asunto(s)
Proteínas de la Membrana/metabolismo , Células Fotorreceptoras de Vertebrados/metabolismo , Transducción de Señal/fisiología , Cilios/metabolismo , Humanos , Transporte de Proteínas/fisiología
17.
J Gen Physiol ; 140(3): 249-66, 2012 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-22891277

RESUMEN

G protein-coupled receptor (GPCR) cascades rely on membrane protein diffusion for signaling and are generally found in spatially constrained subcellular microcompartments. How the geometry of these microcompartments impacts cascade activities, however, is not understood, primarily because of the inability of current live cell-imaging technologies to resolve these small structures. Here, we examine the dynamics of the GPCR rhodopsin within discrete signaling microcompartments of live photoreceptors using a novel high resolution approach. Rhodopsin fused to green fluorescent protein variants, either enhanced green fluorescent protein (EGFP) or the photoactivatable PAGFP (Rho-E/PAGFP), was expressed transgenically in Xenopus laevis rod photoreceptors, and the geometries of light signaling microcompartments formed by lamellar disc membranes and their incisure clefts were resolved by confocal imaging. Multiphoton fluorescence relaxation after photoconversion experiments were then performed with a Ti-sapphire laser focused to the diffraction limit, which produced small sub-cubic micrometer volumes of photoconverted molecules within the discrete microcompartments. A model of molecular diffusion was developed that allows the geometry of the particular compartment being examined to be specified. This was used to interpret the experimental results. Using this unique approach, we showed that rhodopsin mobility across the disc surface was highly heterogeneous. The overall relaxation of Rho-PAGFP fluorescence photoactivated within a microcompartment was biphasic, with a fast phase lasting several seconds and a slow phase of variable duration that required up to several minutes to reach equilibrium. Local Rho-EGFP diffusion within defined compartments was monotonic, however, with an effective lateral diffusion coefficient D(lat) = 0.130 ± 0.012 µm(2)s(-1). Comparison of rhodopsin-PAGFP relaxation time courses with model predictions revealed that microcompartment geometry alone may explain both fast local rhodopsin diffusion and its slow equilibration across the greater disc membrane. Our approach has for the first time allowed direct examination of GPCR dynamics within a live cell signaling microcompartment and a quantitative assessment of the impact of compartment geometry on GPCR activity.


Asunto(s)
Microdominios de Membrana/metabolismo , Células Fotorreceptoras Retinianas Bastones/metabolismo , Rodopsina/metabolismo , Secuencia de Aminoácidos , Animales , Recuperación de Fluorescencia tras Fotoblanqueo , Proteínas Fluorescentes Verdes/genética , Fototransducción , Microdominios de Membrana/química , Microscopía de Fluorescencia por Excitación Multifotónica , Modelos Biológicos , Datos de Secuencia Molecular , Proteínas Recombinantes/genética , Rodopsina/análisis , Rodopsina/genética , Xenopus
18.
J Gen Physiol ; 135(3): 173-96, 2010 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-20176852

RESUMEN

Transport of proteins to and from cilia is crucial for normal cell function and survival, and interruption of transport has been implicated in degenerative and neoplastic diseases. It has been hypothesized that the ciliary axoneme and structures adjacent to and including the basal bodies of cilia impose selective barriers to the movement of proteins into and out of the cilium. To examine this hypothesis, using confocal and multiphoton microscopy we determined the mobility of the highly soluble photoactivatable green fluorescent protein (PAGFP) in the connecting cilium (CC) of live Xenopus retinal rod photoreceptors, and in the contiguous subcellular compartments bridged by the CC, the inner segment (IS) and the outer segment (OS). The estimated axial diffusion coefficients are D(CC) = 2.8 +/- 0.3, D(IS) = 5.2 +/- 0.6, and D(OS) = 0.079 +/- 0.009 microm(2) s(-1). The results establish that the CC does not pose a major barrier to protein diffusion within the rod cell. However, the results also reveal that axial diffusion in each of the rod's compartments is substantially retarded relative to aqueous solution: the axial diffusion of PAGFP was retarded approximately 18-, 32- and 1,000-fold in the IS, CC, and OS, respectively, with approximately 20-fold of the reduction in the OS attributable to tortuosity imposed by the lamellar disc membranes. Previous investigation of PAGFP diffusion in passed, spherical Chinese hamster ovary cells yielded D(CHO) = 20 microm(2) s(-1), and estimating cytoplasmic viscosity as D(aq)/D(CHO) = 4.5, the residual 3- to 10-fold reduction in PAGFP diffusion is ascribed to sub-optical resolution structures in the IS, CC, and OS compartments.


Asunto(s)
Proteínas Fluorescentes Verdes/metabolismo , Cilio Conector de los Fotorreceptores/metabolismo , Segmento Interno de las Células Fotorreceptoras Retinianas/metabolismo , Células Fotorreceptoras Retinianas Bastones/metabolismo , Algoritmos , Animales , Animales Modificados Genéticamente , Transporte Biológico , Difusión , Colorantes Fluorescentes/metabolismo , Líquido Intracelular , Modelos Biológicos , Xenopus laevis
19.
Trends Cell Biol ; 16(11): 560-8, 2006 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-16996267

RESUMEN

The dynamic localization of proteins within cells is often determined by environmental stimuli. In retinal photoreceptors, light exposure results in the massive translocation of three key signal transduction proteins, transducin, arrestin and recoverin, into and out of the outer segment compartment where phototransduction takes place. This phenomenon has rapidly taken the center stage of photoreceptor cell biology, thanks to the introduction of new quantitative and transgenic approaches. Here, we discuss evidence that intracellular protein translocation contributes to adaptation of photoreceptors to diurnal changes in ambient light intensity and summarize the current debate on whether it is driven by diffusion or molecular motors.


Asunto(s)
Péptidos y Proteínas de Señalización Intracelular/metabolismo , Luz , Células Fotorreceptoras de Vertebrados/química , Animales , Difusión , Humanos , Proteínas Motoras Moleculares , Transporte de Proteínas
20.
J Cell Sci ; 117(Pt 14): 3049-59, 2004 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-15197244

RESUMEN

The hypothesis is tested that enhanced green fluorescent protein (EGFP) can be used to quantify the aqueous spaces of living cells, using as a model transgenic Xenopus rods. Consistent with the hypothesis, regions of rods having structures that exclude EGFP, such as the mitochondrial-rich ellipsoid and the outer segments, have highly reduced EGFP fluorescence. Over a 300-fold range of expression the average EGFP concentration in the outer segment was approximately half that in the most intensely fluorescent regions of the inner segment, in quantitative agreement with prior X-ray diffraction estimates of outer segment cytoplasmic volume. In contrast, the fluorescence of soluble arrestin-EGFP fusion protein in the dark adapted rod outer segment was approximately threefold lower than predicted by the EGFP distribution, establishing that the fusion protein is not equilibrated with the cytoplasm. Arrestin-EGFP mass was conserved during a large-scale, light-driven redistribution in which approximately 40% of the protein in the inner segment moved to the outer segment in less than 30 minutes.


Asunto(s)
Arrestina/metabolismo , Citoplasma/metabolismo , Proteínas Fluorescentes Verdes/metabolismo , Proteínas Recombinantes de Fusión/metabolismo , Células Fotorreceptoras Retinianas Bastones/citología , Animales , Animales Modificados Genéticamente , Arrestina/genética , Células CHO , Compartimento Celular , Cricetinae , Cricetulus , Adaptación a la Oscuridad , Proteínas Fluorescentes Verdes/genética , Luz , Microscopía Confocal , Proteínas Recombinantes de Fusión/genética , Células Fotorreceptoras Retinianas Bastones/metabolismo , Xenopus
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA