Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
1.
Hum Mol Genet ; 31(7): 1035-1050, 2022 03 31.
Artículo en Inglés | MEDLINE | ID: mdl-34652420

RESUMEN

Heteromeric Kv2.1/Kv8.2 channels are voltage-gated potassium channels localized to the photoreceptor inner segment. They carry IKx, which is largely responsible for setting the photoreceptor resting membrane potential. Mutations in Kv8.2 result in childhood-onset cone dystrophy with supernormal rod response (CDSRR). We generated a Kv8.2 knockout (KO) mouse and examined retinal signaling and photoreceptor degeneration to gain deeper insight into the complex phenotypes of this disease. Using electroretinograms, we show that there were delayed or reduced signaling from rods depending on the intensity of the light stimulus, consistent with reduced capacity for light-evoked changes in membrane potential. The delayed response was not seen ex vivo where extracellular potassium levels were controlled by the perfusion buffer, so we propose the in vivo alteration is influenced by genotype-associated ionic imbalance. We observed mild retinal degeneration. Signaling from cones was reduced but there was no loss of cone density. Loss of Kv8.2 altered responses to flickering light with responses attenuated at high frequencies and altered in shape at low frequencies. The Kv8.2 KO line on an all-cone retina background had reduced cone-driven ERG b wave amplitudes and underwent degeneration. Altogether, we provide insight into how a deficit in the dark current affects the health and function of photoreceptors.


Asunto(s)
Canales de Potasio con Entrada de Voltaje , Degeneración Retiniana , Enfermedades de la Retina , Animales , Electrorretinografía , Ratones , Canales de Potasio con Entrada de Voltaje/genética , Retina/fisiología , Células Fotorreceptoras Retinianas Conos/fisiología , Degeneración Retiniana/genética
2.
Adv Exp Med Biol ; 1415: 269-276, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37440044

RESUMEN

Photoreceptors (PRs) in the neural retina convert photon capture into an electrical signal that is communicated across a chemical synapse to second-order neurons in the retina and on through the rest of the visual pathway. This information is decoded in the visual cortex to create images. The activity of PRs depends on the concerted action of several voltage-gated ion channels that will be discussed in this chapter.


Asunto(s)
Células Fotorreceptoras , Retina , Células Fotorreceptoras/metabolismo , Retina/metabolismo , Transducción de Señal , Sinapsis/metabolismo , Canales Iónicos/metabolismo , Canales Catiónicos Regulados por Nucleótidos Cíclicos/metabolismo , Células Fotorreceptoras Retinianas Conos/fisiología
3.
Exp Eye Res ; 170: 108-116, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29486162

RESUMEN

The 14-3-3 family of proteins has undergone considerable expansion in higher eukaryotes with humans and mice expressing seven isoforms (ß, ε, η, γ, θ, ζ, and σ) from seven distinct genes (YWHAB, YWAHE, YWHAH, YWHAG, YWHAQ, YWHAZ, and SFN). Growing evidence indicates that while highly conserved, these isoforms are not entirely functionally redundant as they exhibit unique tissue expression profiles, subcellular localization, and biochemical functions. A key limitation in our understanding of 14-3-3 biology lies in our limited knowledge of cell-type specific 14-3-3 expression. Here we provide a characterization of 14-3-3 expression in whole retina and isolated rod photoreceptors using reverse-transcriptase digital droplet PCR. We find that all 14-3-3 genes with the exception of SFN are expressed in mouse retina with YWHAQ and YWHAE being the most highly expressed. Rod photoreceptors are enriched in YWHAE (14-3-3 ε). Immunohistochemistry revealed that 14-3-3 ε and 14-3-3 ζ exhibit unique distributions in photoreceptors with 14-3-3 ε restricted to the inner segment and 14-3-3 ζ localized to the outer segment. Our data demonstrates that, in the retina, 14-3-3 isoforms likely serve specific functions as they exhibit unique expression levels and cell-type specificity. As such, future investigations into 14-3-3 function in rod photoreceptors should be centered on 14-3-3 ε and 14-3-3 ζ, depending on the subcellular region of question.


Asunto(s)
Proteínas 14-3-3/genética , Regulación de la Expresión Génica/fisiología , Retina/metabolismo , Células Fotorreceptoras Retinianas Bastones/metabolismo , Animales , Western Blotting , Femenino , Inmunohistoquímica , Masculino , Ratones , Ratones Endogámicos C3H , Ratones Endogámicos C57BL , Plásmidos , Isoformas de Proteínas/genética , ARN Mensajero/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
4.
Am J Physiol Heart Circ Physiol ; 304(5): H687-96, 2013 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-23262137

RESUMEN

Vascular endothelial growth factor receptor-2 (VEGFR2) is a receptor tyrosine kinase that is expressed in endothelial cells and regulates angiogenic signal transduction under both physiological and pathological conditions. VEGFR2 turnover at the plasma membrane (PM) is regulated by its transport through endocytic and secretory transport pathways. Short-range cargo trafficking along actin filaments is commonly regulated by motor proteins of myosin superfamily. In the current study, performed in primary human endothelial cells, we demonstrate that unconventional myosin 1c (Myo1c; class I family member) regulates the localization of VEGFR2 at the PM. We further demonstrate that the recruitment of VEGFR2 to the PM and its colocalization with Myo1c and caveolin-1 occur in response to VEGF-A (VEGF) stimulation. In addition, VEGF-induced delivery of VEGFR2 to the cell surface requires Myo1c; surface VEGFR2 levels are reduced in the absence of Myo1c and, more importantly, are restored by the overexpression of wild-type but not mutant Myo1c. Subcellular density gradient fractionation revealed that partitioning of VEGFR2 into caveolin-1- and Myo1c-enriched membrane fractions is dependent on VEGF stimulation. Myo1c depletion resulted in increased VEGF-induced VEGFR2 transport to the lysosomes for degradation and was rescued by applying either brefeldin A, which blocks trafficking between the endoplasmic reticulum and the Golgi complex, or dynasore, an inhibitor of dynamin-mediated endocytosis. Myo1c depletion also reduced VEGF-induced VEGFR2 phosphorylation at Y1175 and phosphorylation-dependent activation of ERK1/2 and c-Src kinase, leading to reduced cell proliferation and cell migration. This is the first report demonstrating that Myo1c is an important mediator of VEGF-induced VEGFR2 delivery to the cell surface and plays a role in angiogenic signaling.


Asunto(s)
Células Endoteliales/metabolismo , Proteínas Motoras Moleculares/metabolismo , Miosina Tipo I/metabolismo , Neovascularización Fisiológica/fisiología , Transducción de Señal/fisiología , Receptor 2 de Factores de Crecimiento Endotelial Vascular/metabolismo , Antimaláricos/farmacología , Brefeldino A/farmacología , Caveolina 1/metabolismo , Membrana Celular/metabolismo , Movimiento Celular/fisiología , Proliferación Celular , Cloroquina/farmacología , Endocitosis/efectos de los fármacos , Endocitosis/fisiología , Células Endoteliales/citología , Células Endoteliales de la Vena Umbilical Humana , Humanos , Hidrazonas/farmacología , Microdominios de Membrana/metabolismo , Proteínas Motoras Moleculares/genética , Miosina Tipo I/genética , Inhibidores de la Síntesis de la Proteína/farmacología , Transporte de Proteínas/efectos de los fármacos , Transporte de Proteínas/fisiología , ARN Mensajero/metabolismo , Vías Secretoras/efectos de los fármacos , Vías Secretoras/fisiología
5.
J Biol Chem ; 286(42): 36749-61, 2011 Oct 21.
Artículo en Inglés | MEDLINE | ID: mdl-21880737

RESUMEN

The α5ß1 integrin heterodimer regulates many processes that contribute to embryonic development and angiogenesis, in both physiological and pathological contexts. As one of the major adhesion complexes on endothelial cells, it plays a vital role in adhesion and migration along the extracellular matrix. We recently showed that angiogenesis is modulated by syntaxin 6, a Golgi- and endosome-localized t-SNARE, and that it does so by regulating the post-Golgi trafficking of VEGFR2. Here we show that syntaxin 6 is also required for α5ß1 integrin-mediated adhesion of endothelial cells to, and migration along, fibronectin. We demonstrate that syntaxin 6 and α5ß1 integrin colocalize in EEA1-containing early endosomes, and that functional inhibition of syntaxin 6 leads to misrouting of ß1 integrin to the degradation pathway (late endosomes and lysosomes) rather transport along recycling pathway from early endosomes; an increase in the pool of ubiquitinylated α5 integrin and its lysosome-dependent degradation; reduced cell spreading on fibronectin; decreased Rac1 activation; and altered Rac1 localization. Collectively, our data show that functional syntaxin 6 is required for the regulation of α5ß1-mediated endothelial cell movement on fibronectin. These syntaxin 6-regulated membrane trafficking events control outside-in signaling via haptotactic and chemotactic mechanisms.


Asunto(s)
Movimiento Celular/fisiología , Células Endoteliales/metabolismo , Fibronectinas , Integrina alfa5/metabolismo , Integrina beta1/metabolismo , Proteínas Qa-SNARE/metabolismo , Adhesión Celular/fisiología , Membrana Celular/metabolismo , Células Cultivadas , Endosomas/metabolismo , Células Endoteliales/citología , Activación Enzimática/fisiología , Humanos , Lisosomas/metabolismo , Transporte de Proteínas/fisiología , Proteolisis , Transducción de Señal/fisiología , Ubiquitinación/fisiología , Proteínas de Transporte Vesicular/metabolismo , Proteína de Unión al GTP rac1/metabolismo
6.
Front Neurosci ; 16: 821059, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35401099

RESUMEN

Alzheimer's disease (AD) belongs to a class of diseases characterized by progressive accumulation and aggregation of pathogenic proteins, particularly Aß proteins. Genetic analysis has identified UBQLN1 as an AD candidate gene. Ubiquilin-1 levels reduce with AD progression, suggesting a potential loss-of-function mechanism. The ubiquilin-1 protein is involved in protein quality control (PQC), which plays essential roles in cellular growth and normal cell function. Ubiquilin-1 regulates γ-secretase by increasing endoproteolysis of PS1, a key γ-secretase component. Presently, the effects of ubiquilin-1 on cellular physiology as well as Aß-related events require further investigation. Here, we investigated the effects of ubiquilin-1 on cellular growth and viability in association with APP (amyloid-ß protein precursor), APP processing-related ß-secretase (BACE1, BACE) and γ-secretase using cell and animal-based models. We showed that loss-of-function in Drosophila ubqn suppresses human APP and human BACE phenotypes in wing veins and altered cell number and tissue compartment size in the wing. Additionally, we performed cell-based studies and showed that silencing UBQLN1 reduced cell viability and increased caspase-3 activity. Overexpression of UBQLN1 significantly reduced Aß levels. Furthermore, pharmacological inhibition of γ-secretase increased ubiquilin-1 protein levels, suggesting a mechanism that regulates ubiquilin-1 levels which may associate with reduced Aß reduction by inhibiting γ-secretase. Collectively, our results support not only a loss-of-function mechanism of ubiquilin-1 in association with AD, but also support the significance of targeting ubiquilin-1-mediated PQC as a potential therapeutic strategy for AD.

7.
Front Cell Neurosci ; 14: 595523, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33250719

RESUMEN

Vision, hearing, smell, taste, and touch are the tools used to perceive and navigate the world. They enable us to obtain essential resources such as food and highly desired resources such as mates. Thanks to the investments in biomedical research the molecular unpinning's of human sensation are rivaled only by our knowledge of sensation in the laboratory mouse. Humans rely heavily on vision whereas mice use smell as their dominant sense. Both modalities have many features in common, starting with signal detection by highly specialized primary sensory neurons-rod and cone photoreceptors (PR) for vision, and olfactory sensory neurons (OSN) for the smell. In this chapter, we provide an overview of how these two types of primary sensory neurons operate while highlighting the similarities and distinctions.

8.
Front Cell Neurosci ; 14: 589494, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33173469

RESUMEN

Transducin mediates signal transduction in a classical G protein-coupled receptor (GPCR) phototransduction cascade. Interactions of transducin with the receptor and the effector molecules had been extensively investigated and are currently defined at the atomic level. However, partners and functions of rod transducin α (Gαt 1) and ßγ (Gß1γ1) outside the visual pathway are not well-understood. In particular, light-induced redistribution of rod transducin from the outer segment to the inner segment and synaptic terminal (IS/ST) allows Gαt1 and/or Gß1γ1 to modulate synaptic transmission from rods to rod bipolar cells (RBCs). Protein-protein interactions underlying this modulation are largely unknown. We discuss known interactors of transducin in the rod IS/ST compartment and potential pathways leading to the synaptic effects of light-dispersed Gαt1 and Gß1γ1. Furthermore, we show that a prominent non-GPCR guanine nucleotide exchange factor (GEF) and a chaperone of Gα subunits, resistance to inhibitors of cholinesterase 8A (Ric-8A) protein, is expressed throughout the retina including photoreceptor cells. Recent structures of Ric-8A alone and in complexes with Gα subunits have illuminated the structural underpinnings of the Ric-8A activities. We generated a mouse model with conditional knockout of Ric-8A in rods in order to begin defining the functional roles of the protein in rod photoreceptors and the retina. Our analysis suggests that Ric-8A is not an obligate chaperone of Gαt1. Further research is needed to investigate probable roles of Ric-8A as a GEF, trafficking chaperone, or a mediator of the synaptic effects of Gαt1.

9.
Front Cell Dev Biol ; 4: 51, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27376061

RESUMEN

Spatial regulation of exocytosis relies on the exocyst, a hetero-octameric protein complex that tethers vesicles to fusion sites at the plasma membrane. Nevertheless, our understanding of mechanisms regulating exocyst assembly/disassembly, localization, and function are incomplete. Here, we have exploited a panel of anti-Sec6 monoclonal antibodies (mAbs) to probe possible configurational changes accompanying transitions in exocyst function in epithelial MDCK cells. Sec6 is quantitatively associated with Sec8 in high molecular weight complexes, as shown by gel filtration and co-immunoprecipitation studies. We mapped epitopes recognized by more than 20 distinct mAbs to one of six Sec6 segments. Surprisingly, mAbs that bound epitopes in each segment labeled distinct subcellular structures. In general, antibodies to epitopes in N-terminal domains labeled Sec6 in either cytosolic or nuclear pools, whereas those that bound epitopes in C-terminal domains labeled membrane-associated Sec6. In this latter group, we identified antibodies that labeled distinct Sec6 populations at the apical junctional complex, desmosomes, endoplasmic reticulum and vimentin-type intermediate filaments. That each antibody was specific was verified by both Sec6 RNAi and competition with fusion proteins containing each domain. Comparison of non-polarized and polarized cells revealed that many Sec6 epitopes either redistribute or become concealed during epithelial polarization. Transitions in exocyst configurations may be regulated in part by the actions of Ral GTPases, because the exposure of Sec6 C-terminal domain epitopes at the plasma membrane is significantly reduced upon RalA RNAi. To determine whether spatio-temporal changes in epitope accessibility was correlated with differential stability of interactions between Sec6 and other exocyst subunits, we quantified relative amounts of each subunit that co-immunoprecipitated with Sec6 when antibodies to N-terminal or C-terminal epitopes were used. Antibodies to Sec6NT co-precipitated substantially more Sec5, -10, -15, Exo70 and -84 than did those to Sec6CT. In contrast, antibodies to Sec6CT co-precipitated more Sec3 and Sec8 than did those to Sec6NT. These results are consistent with a model in which exocyst activation during periods of rapid membrane expansion is accompanied by molecular rearrangements within the holocomplex or association with accessory proteins, which expose the Sec6 C-terminal domain when the complex is membrane-bound and conceal it when the complex is cytoplasmic.

10.
PLoS One ; 8(4): e61857, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23626741

RESUMEN

The formation and maintenance of cell-cell junctions, both under physiological and pathological conditions, requires the targeting and trafficking of junctional proteins. Proteins of the syntaxin (Stx)-family localize to a variety of subcellular membranes and contribute to intracellular transport of cargo by regulating vesicle fusion events at these sites. Unlike plasma membrane localized Stxs, the roles of endosome- and Golgi-localized stx proteins in epithelial morphogenesis are less understood. Here we show that Stx16- an endosome- and Golgi-localized target-membrane soluble N-ethylmaleimide attachment protein receptor (t-SNARE) that plays a role in membrane trafficking between these compartments - is essential for lumen development. In cultured Madin Darby Canine Kidney (MDCK) cells, Stx16 was selectively upregulated as sparsely plated cells attained confluency. Stx16-depleted confluent monolayers consistently showed lower transepithelial resistance than control monolayers, and failed to maintain endogenous and ectopically expressed E-cadherin at the adherens junctions due to decreased recycling. We further found that whereas cysts formed by MDCK cells cultured in Matrigel have a single hollow lumen, those formed by stx16-depleted counterparts had multiple lumens, due to abnormal orientiation of the mitotic spindle. Finally, a similar role for stx16 function in vivo is indicated by our analysis of pronephric-duct development in zebrafish expressing the claudinB:lynGFP transgene; lack of stx16 function in this structure (in stx16-morphant embryos) led to the development of enlarged, torturous pronephric ducts with more than one lumen. Taken together, our in vitro and in vivo studies establish a role for Stx16 in maintaining the integrity of cell-cell junctions, and thereby in morphogenesis of the kidney epithelial lumen.


Asunto(s)
Regulación del Desarrollo de la Expresión Génica , Uniones Intercelulares/metabolismo , Riñón/metabolismo , Sintaxina 16/metabolismo , Animales , Cadherinas/genética , Cadherinas/metabolismo , Recuento de Células , Membrana Celular/metabolismo , Membrana Celular/ultraestructura , Colágeno , Perros , Combinación de Medicamentos , Embrión no Mamífero , Endosomas/metabolismo , Endosomas/ultraestructura , Aparato de Golgi/metabolismo , Aparato de Golgi/ultraestructura , Uniones Intercelulares/genética , Uniones Intercelulares/ultraestructura , Riñón/crecimiento & desarrollo , Riñón/ultraestructura , Laminina , Células de Riñón Canino Madin Darby , Transporte de Proteínas , Proteoglicanos , Proteínas SNARE/genética , Proteínas SNARE/metabolismo , Transducción de Señal , Huso Acromático/metabolismo , Huso Acromático/ultraestructura , Sintaxina 16/genética , Transgenes , Pez Cebra
11.
Biosci Rep ; 32(4): 383-91, 2012 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-22489884

RESUMEN

Intracellular membrane trafficking along endocytic and secretory transport pathways plays a critical role in diverse cellular functions including both developmental and pathological processes. Briefly, proteins and lipids destined for transport to distinct locations are collectively assembled into vesicles and delivered to their target site by vesicular fusion. SNARE (soluble N-ethylmaleimide-sensitive factor-attachment protein receptor) proteins are required for these events, during which v-SNAREs (vesicle SNAREs) interact with t-SNAREs (target SNAREs) to allow transfer of cargo from donor vesicle to target membrane. Recently, the t-SNARE family member, syntaxin-6, has been shown to play an important role in the transport of proteins that are key to diverse cellular dynamic processes. In this paper, we briefly discuss the specific role of SNAREs in various mammalian cell types and comprehensively review the various roles of the Golgi- and endosome-localized t-SNARE, syntaxin-6, in membrane trafficking during physiological as well as pathological conditions.


Asunto(s)
Membranas Intracelulares/metabolismo , Proteínas Qa-SNARE/fisiología , Animales , Endocitosis , Células Endoteliales/metabolismo , Células Endoteliales/fisiología , Humanos , Transporte de Proteínas , Proteínas Qa-SNARE/metabolismo , Proteínas SNARE/metabolismo , Proteínas SNARE/fisiología , Vesículas Secretoras/metabolismo
12.
PLoS One ; 7(9): e44572, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22962618

RESUMEN

The soluble form of vascular endothelial growth factor receptor 1 (sVEGFR-1/sFlt1) is generated by alternative splicing of the FLT1 gene. Secretion of sFlt1 from endothelial cells plays an important role in blood vessel sprouting and morphogenesis. However, excess sFlt1 secretion is associated with diseases such as preeclampsia and chronic kidney disease. To date, the secretory transport process involved in the secretion of sFlt1 is poorly understood. In the present study, we investigated the itinerary of sFlt1 trafficking along the secretory pathway. To understand the timecourse of sFlt1 secretion, endothelial cells stably expressing sFlt1 were metabolically radiolabeled with [(35)S]-methionine and cysteine. Our results indicate that after initial synthesis the levels of secreted [(35)S]-sFlt1 in the extracellular medium peaks at 8 hours. Treatment with brefeldin A (BFA), a drug which blocks trafficking between the endoplasmic reticulum (ER) and the Golgi complex, inhibited extracellular release of sFlt1 suggesting that ER to Golgi and intra-Golgi trafficking of sFlt1 are essential for its secretion. Furthermore, we show that ectopic expression of dominant-negative mutant forms of Arf1, Arf6, and Rab11 as well as siRNA-mediated knockdown of these GTPases block secretion of sFlt1 during normoxic and hypoxic conditions suggesting role for these small GTPases. This work is the first to report role of regulatory proteins involved in sFlt1 trafficking along the secretory pathway and may provide insights and new molecular targets for the modulation of sFlt-1 release during physiological and pathological conditions.


Asunto(s)
Factor 1 de Ribosilacion-ADP/genética , Factores de Ribosilacion-ADP/genética , Retículo Endoplásmico/metabolismo , Aparato de Golgi/metabolismo , Receptor 1 de Factores de Crecimiento Endotelial Vascular/genética , Proteínas de Unión al GTP rab/genética , Factor 1 de Ribosilacion-ADP/antagonistas & inhibidores , Factor 1 de Ribosilacion-ADP/metabolismo , Factor 6 de Ribosilación del ADP , Factores de Ribosilacion-ADP/antagonistas & inhibidores , Factores de Ribosilacion-ADP/metabolismo , Aminoácidos/metabolismo , Brefeldino A/farmacología , Línea Celular Tumoral , Retículo Endoplásmico/efectos de los fármacos , Retículo Endoplásmico/genética , Expresión Génica/efectos de los fármacos , Aparato de Golgi/efectos de los fármacos , Aparato de Golgi/genética , Células Endoteliales de la Vena Umbilical Humana , Humanos , Plásmidos , Estructura Terciaria de Proteína , Inhibidores de la Síntesis de la Proteína/farmacología , Transporte de Proteínas/efectos de los fármacos , ARN Interferente Pequeño/genética , Transducción de Señal/efectos de los fármacos , Solubilidad , Radioisótopos de Azufre , Transfección , Receptor 1 de Factores de Crecimiento Endotelial Vascular/metabolismo , Proteínas de Unión al GTP rab/antagonistas & inhibidores , Proteínas de Unión al GTP rab/metabolismo
13.
Fly (Austin) ; 4(3): 204-12, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-20495373

RESUMEN

Proper activation of the Ras/MAPK pathway is broadly required during development, and in many cases, signal transduction downstream of the receptor is linear. Thus, different mechanisms exist to properly regulate the large number of specific developmental outputs that are required by the activation of this pathway. Previously, we have reported a regulated cytoplasmic sequestration of phosphorylated MAPK (pMAPK) in developing Drosophila compound eyes and wings "called MAPK Cytoplasmic Hold". In the developing wing, we have shown that cytoplasmic hold promotes the differentiation of wing vein tissue, while pMAPK nuclear translocation regulates growth and division. We had also suggested that the Ras pathway signals for inducing cell growth and cell division split upstream of the nuclear translocation of MAPK itself. Here, we further refine the role of MAPK in Drosophila. We report evidence that suggests, for the first time, that the phosphorylation of MAPK is itself another step in the regulation of cell growth and division in both Drosophila wing and eye cells. We show that inhibition of MAPK phosphorylation, or pMAPK nuclear translocation, is sufficient to block cell growth, but not cell division. These data suggest that non-phosphorylated MAPK is sufficient to induce cell division, but not cell growth, once inside the nucleus of the cell.


Asunto(s)
Ojo Compuesto de los Artrópodos/crecimiento & desarrollo , Drosophila/crecimiento & desarrollo , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Alas de Animales/crecimiento & desarrollo , Animales , División Celular , Aumento de la Célula , Ojo Compuesto de los Artrópodos/enzimología , Drosophila/enzimología , Proteínas de Drosophila/metabolismo , Proteínas del Ojo/metabolismo , Femenino , Masculino , Proteínas del Tejido Nervioso/metabolismo , Fosforilación , Proteínas Quinasas/metabolismo , Alas de Animales/enzimología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA