Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
Más filtros

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
J Am Soc Nephrol ; 31(3): 544-559, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-32047005

RESUMEN

BACKGROUND: Understanding podocyte-specific responses to injury at a systems level is difficult because injury leads to podocyte loss or an increase of extracellular matrix, altering glomerular cellular composition. Finding a window into early podocyte injury might help identify molecular pathways involved in the podocyte stress response. METHODS: We developed an approach to apply proteome analysis to very small samples of purified podocyte fractions. To examine podocytes in early disease states in FSGS mouse models, we used podocyte fractions isolated from individual mice after chemical induction of glomerular disease (with Doxorubicin or LPS). We also applied single-glomerular proteome analysis to tissue from patients with FSGS. RESULTS: Transcriptome and proteome analysis of glomeruli from patients with FSGS revealed an underrepresentation of podocyte-specific genes and proteins in late-stage disease. Proteome analysis of purified podocyte fractions from FSGS mouse models showed an early stress response that includes perturbations of metabolic, mechanical, and proteostasis proteins. Additional analysis revealed a high correlation between the amount of proteinuria and expression levels of the mechanosensor protein Filamin-B. Increased expression of Filamin-B in podocytes in biopsy samples from patients with FSGS, in single glomeruli from proteinuric rats, and in podocytes undergoing mechanical stress suggests that this protein has a role in detrimental stress responses. In Drosophila, nephrocytes with reduced filamin homolog Cher displayed altered filtration capacity, but exhibited no change in slit diaphragm structure. CONCLUSIONS: We identified conserved mechanisms of the podocyte stress response through ultrasensitive proteome analysis of human glomerular FSGS tissue and purified native mouse podocytes during early disease stages. This approach enables systematic comparisons of large-scale proteomics data and phenotype-to-protein correlation.


Asunto(s)
Filaminas/genética , Regulación de la Expresión Génica , Glomeruloesclerosis Focal y Segmentaria/patología , Proteómica/métodos , Estrés Fisiológico/genética , Animales , Células Cultivadas , Modelos Animales de Enfermedad , Glomeruloesclerosis Focal y Segmentaria/genética , Humanos , Ratones , Podocitos/metabolismo , Proteinuria/genética , Proteinuria/fisiopatología , Distribución Aleatoria , Ratas
2.
Kidney Int ; 95(4): 846-858, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30770218

RESUMEN

Recent human genetic studies have suggested an intriguing link between ciliary signaling defects and altered DNA damage responses in nephronophthisis (NPH) and related ciliopathies. However, the molecular mechanism and the role of altered DNA damage response in kidney degeneration and fibrosis have remained elusive. We recently identified the kinase-regulated DNA damage response target Apoptosis Antagonizing Transcription Factor (AATF) as a master regulator of the p53 response. Here, we characterized the phenotype of mice with genetic deletion of Aatf in tubular epithelial cells. Mice were born without an overt phenotype, but gradually developed progressive kidney disease. Histology was notable for severe tubular atrophy and interstitial fibrosis as well as cysts at the corticomedullary junction, hallmarks of human nephronophthisis. Aatf deficiency caused ciliary defects as well as an accumulation of DNA double strand breaks. In addition to its role as a p53 effector, we found that AATF suppressed RNA:DNA hybrid (R loop) formation, a known cause of DNA double strand breaks, and enabled DNA double strand break repair in vitro. Genome-wide transcriptomic analysis of Aatf deficient tubular epithelial cells revealed several deregulated pathways that could contribute to the nephronophthisis phenotype, including alterations in the inflammatory response and anion transport. These results suggest that AATF is a regulator of primary cilia and a modulator of the DNA damage response, connecting two pathogenetic mechanisms in nephronophthisis and related ciliopathies.


Asunto(s)
Proteínas Reguladoras de la Apoptosis/metabolismo , Cilios/patología , Roturas del ADN de Doble Cadena , Enfermedades Renales Quísticas/genética , Túbulos Renales/patología , Proteínas Nucleares/metabolismo , Animales , Apoptosis/genética , Proteínas Reguladoras de la Apoptosis/genética , Biopsia , Línea Celular Tumoral , Cilios/genética , Modelos Animales de Enfermedad , Células Epiteliales/citología , Células Epiteliales/patología , Fibrosis , Humanos , Enfermedades Renales Quísticas/patología , Túbulos Renales/citología , Ratones , Ratones Noqueados , Proteínas Nucleares/genética , Cultivo Primario de Células , Estructuras R-Loop/genética , Proteínas Represoras/metabolismo , Transducción de Señal/genética
3.
J Am Soc Nephrol ; 29(8): 2060-2068, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-29794128

RESUMEN

Background Three different cell types constitute the glomerular filter: mesangial cells, endothelial cells, and podocytes. However, to what extent cellular heterogeneity exists within healthy glomerular cell populations remains unknown.Methods We used nanodroplet-based highly parallel transcriptional profiling to characterize the cellular content of purified wild-type mouse glomeruli.Results Unsupervised clustering of nearly 13,000 single-cell transcriptomes identified the three known glomerular cell types. We provide a comprehensive online atlas of gene expression in glomerular cells that can be queried and visualized using an interactive and freely available database. Novel marker genes for all glomerular cell types were identified and supported by immunohistochemistry images obtained from the Human Protein Atlas. Subclustering of endothelial cells revealed a subset of endothelium that expressed marker genes related to endothelial proliferation. By comparison, the podocyte population appeared more homogeneous but contained three smaller, previously unknown subpopulations.Conclusions Our study comprehensively characterized gene expression in individual glomerular cells and sets the stage for the dissection of glomerular function at the single-cell level in health and disease.


Asunto(s)
Células Endoteliales/metabolismo , Perfilación de la Expresión Génica , Glomérulos Renales/fisiología , Células Mesangiales/metabolismo , Podocitos/metabolismo , Análisis de Secuencia de ARN , Animales , Células Cultivadas , Regulación de la Expresión Génica , Glomérulos Renales/citología , Masculino , Ratones , Ratones Endogámicos , Valores de Referencia
4.
Hum Mol Genet ; 21(26): 5528-38, 2012 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-23026745

RESUMEN

Nephronophthisis (NPH) is a genetically heterogenous kidney disease and represents the most common genetic cause for end-stage renal disease in children. It is caused by the mutation of genes encoding for the nephrocystin proteins (NPHPs) which localize to primary cilia or centrosomes, classifying this disease as a 'ciliopathy'. Recently, it has been shown that NPHP4 acts as a potent negative regulator of mammalian Hippo signalling by interacting with the Lats protein kinase and controlling the phosphorylation of the oncogenic transcriptional activator TAZ. Here, we demonstrate that NPHP9, another NPH family member, also controls TAZ activity by a distinct mechanism. NPHP9, which is also called NEK8, directly interacted with TAZ and induced nuclear translocation of the TAZ/NPHP9 protein complex. Binding of NPHP9 to TAZ was enhanced in a TAZ mutant that lost its ability to bind 14-3-3, suggesting that 14-3-3 and NPHP9 may compete for TAZ binding, with 14-3-3 favouring cytoplasmic retention and NPHP9 mediating nuclear delivery. Consistently, co-expression of NPHP4, which inhibits TAZ phosphorylation at the 14-3-3 binding site through the inhibition of Lats kinase activity, induced efficient nuclear delivery of the TAZ/NPHP9 protein pair. Consistent with a role for TAZ in controlling proliferation and tumorigenesis, the downregulation of NPHP9 inhibited the TAZ-dependent proliferation of hippo-responsive normal epithelial and also breast cancer cells. As NPHP9 has been shown to be upregulated in breast cancer, these data do not only support a critical role for TAZ/hippo signalling in the pathogenesis of NPH but may also imply a possible role for NPHP9 in TAZ-mediated tumorigenesis.


Asunto(s)
Cilios/patología , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Enfermedades Renales Quísticas/genética , Proteínas Quinasas/metabolismo , Proteínas 14-3-3/genética , Proteínas 14-3-3/metabolismo , Núcleo Celular/metabolismo , Proliferación Celular , Centrosoma/metabolismo , Cilios/metabolismo , Regulación hacia Abajo , Técnica del Anticuerpo Fluorescente , Regulación de la Expresión Génica , Células HEK293 , Humanos , Inmunoprecipitación , Péptidos y Proteínas de Señalización Intracelular/genética , Enfermedades Renales Quísticas/patología , Luciferasas/metabolismo , Mutación , Quinasas Relacionadas con NIMA , Fosforilación , Plásmidos , Proteínas Quinasas/genética , Proteínas/genética , Proteínas/metabolismo , Transducción de Señal , Transactivadores , Factores de Transcripción , Proteínas Coactivadoras Transcripcionales con Motivo de Unión a PDZ
5.
Am J Physiol Renal Physiol ; 303(10): F1473-85, 2012 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-22975019

RESUMEN

Inflammation conveys the development of glomerular injury and is a major cause of progressive kidney disease. NF-κB signaling is among the most important regulators of proinflammatory signaling. Its role in podocytes, the epithelial cells at the kidney filtration barrier, is poorly understood. Here, we inhibited NF-κB signaling in podocytes by specific ablation of the NF-κB essential modulator (NEMO, IKKγ). Podocyte-specific NEMO-deficient mice (NEMO(pko)) were viable and did not show proteinuria or overt changes in kidney morphology. After induction of glomerulonephritis, both NEMO(pko) and control mice developed significant proteinuria. However, NEMO(pko) mice recovered much faster, showing rapid remission of proteinuria and restoration of podocyte morphology. Interestingly, quantification of infiltrating macrophages, T-lymphocytes, and granulocytes at day 7 revealed no significant difference between wild-type and NEMO(pko). To further investigate the underlying mechanisms, we created a stable NEMO knockdown mouse podocyte cell line. Again, no overt changes in morphology were observed. Translocation of NF-κB to the nucleus after stimulation with TNFα or IL-1 was sufficiently inhibited. Moreover, secretion of proinflammatory chemokines from podocytes after stimulation with TNFα or IL-1 was significantly reduced in NEMO-deficient podocytes and in glomerular samples obtained at day 7 after induction of nephrotoxic nephritis. Collectively, these results show that proinflammatory activity of NF-κB in podocytes aggravates proteinuria in experimental glomerulonephritis in mice. Based on these data, it may be speculated that immunosuppressive drugs may not only target professional immune cells but also podocytes directly to convey their beneficial effects in various types of glomerulonephritis.


Asunto(s)
Glomerulonefritis/metabolismo , FN-kappa B/metabolismo , Podocitos/metabolismo , Proteinuria/metabolismo , Transducción de Señal/fisiología , Animales , Modelos Animales de Enfermedad , Glomerulonefritis/patología , Células HEK293 , Humanos , Quinasa I-kappa B/genética , Quinasa I-kappa B/metabolismo , Inflamación/metabolismo , Inflamación/patología , Interleucina-1/farmacología , Macrófagos/metabolismo , Macrófagos/patología , Ratones , Ratones Noqueados , Podocitos/efectos de los fármacos , Podocitos/patología , Proteinuria/patología , Interferencia de ARN , Transducción de Señal/efectos de los fármacos , Linfocitos T/metabolismo , Linfocitos T/patología , Factor de Necrosis Tumoral alfa/farmacología
6.
Nat Commun ; 13(1): 4099, 2022 07 14.
Artículo en Inglés | MEDLINE | ID: mdl-35835746

RESUMEN

Hypertension and kidney disease have been repeatedly associated with genomic variants and alterations of lysine metabolism. Here, we combined stable isotope labeling with untargeted metabolomics to investigate lysine's metabolic fate in vivo. Dietary 13C6 labeled lysine was tracked to lysine metabolites across various organs. Globally, lysine reacts rapidly with molecules of the central carbon metabolism, but incorporates slowly into proteins and acylcarnitines. Lysine metabolism is accelerated in a rat model of hypertension and kidney damage, chiefly through N-alpha-mediated degradation. Lysine administration diminished development of hypertension and kidney injury. Protective mechanisms include diuresis, further acceleration of lysine conjugate formation, and inhibition of tubular albumin uptake. Lysine also conjugates with malonyl-CoA to form a novel metabolite Nε-malonyl-lysine to deplete malonyl-CoA from fatty acid synthesis. Through conjugate formation and excretion as fructoselysine, saccharopine, and Nε-acetyllysine, lysine lead to depletion of central carbon metabolites from the organism and kidney. Consistently, lysine administration to patients at risk for hypertension and kidney disease inhibited tubular albumin uptake, increased lysine conjugate formation, and reduced tricarboxylic acid (TCA) cycle metabolites, compared to kidney-healthy volunteers. In conclusion, lysine isotope tracing mapped an accelerated metabolism in hypertension, and lysine administration could protect kidneys in hypertensive kidney disease.


Asunto(s)
Hipertensión , Riñón , Lisina , Albúminas/metabolismo , Animales , Carbono/metabolismo , Modelos Animales de Enfermedad , Hipertensión/metabolismo , Riñón/metabolismo , Lisina/metabolismo , Malonil Coenzima A/metabolismo , Ratas
7.
J Am Soc Nephrol ; 19(10): 1871-8, 2008 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-18614772

RESUMEN

Classically, infants with mutations in NPHS1, which encodes nephrin, present with nephrotic syndrome within the first 3 mo of life (congenital nephrotic syndrome of the Finnish-type), and children with mutations in NPHS2, which encodes podocin, present later with steroid-resistant nephrotic syndrome. Recently, however, NPHS2 mutations have been identified in children with congenital nephrotic syndrome. Whether NPHS1 mutations similarly account for some cases of childhood steroid-resistant nephrotic syndrome is unknown. In this study, 160 patients who belonged to 142 unrelated families and presented with nephrotic syndrome at least 3 mo after birth were screened for NPHS1 variants once mutations in NPHS2 had been excluded. Compound heterozygous NPHS1 mutations were identified in one familial case and nine sporadic cases. Mutations included protein-truncating nonsense and frameshift mutations, as well as splice-site and missense variants. Mutations were classified as "severe" or "mild" using prediction algorithms and functional assays. Most missense variants trafficked normally to the plasma membrane and maintained the ability to form nephrin homodimers and to heterodimerize with NEPH1, suggesting retained function. The presence of at least one "mild" mutation in these patients likely explains the later onset and milder course of disease. These results broaden the spectrum of renal disease related to nephrin mutations.


Asunto(s)
Edad de Inicio , Proteínas de la Membrana/genética , Mutación/genética , Síndrome Nefrótico/epidemiología , Síndrome Nefrótico/genética , Algoritmos , Niño , Preescolar , Mapeo Cromosómico , Estudios de Cohortes , Femenino , Humanos , Lactante , Masculino , Síndrome Nefrótico/patología , Índice de Severidad de la Enfermedad
8.
Sci Rep ; 8(1): 11042, 2018 07 23.
Artículo en Inglés | MEDLINE | ID: mdl-30038331

RESUMEN

Fabry disease is a lysosomal storage disorder resulting from impaired alpha-galactosidase A (α-Gal A) enzyme activity due to mutations in the GLA gene. Currently, powerful diagnostic tools and in vivo research models to study Fabry disease are missing, which is a major obstacle for further improvements in diagnosis and therapy. Here, we explore the utility of urine-derived primary cells of Fabry disease patients. Viable cells were isolated and cultured from fresh urine void. The obtained cell culture, modeling the renal epithelium, is characterized by patient-specific information. We demonstrate that this non-invasive source of patient cells provides an adequate cellular in vivo model as cells exhibit decreased α-Gal A enzyme activity and concomitant globotriaosylceramide accumulation. Subsequent quantitative proteomic analyses revealed dysregulation of endosomal and lysosomal proteins indicating an involvement of the Coordinated Lysosomal Expression and Regulation (CLEAR) network in the disease pathology. This proteomic pattern resembled data from our previously described human podocyte model of Fabry disease. Taken together, the employment of urine-derived primary cells of Fabry disease patients might have diagnostic and prognostic implications in the future. Our findings pave the way towards a more detailed understanding of pathophysiological mechanisms and may allow the development of future tailored therapeutic strategies.


Asunto(s)
Enfermedad de Fabry/diagnóstico , Enfermedad de Fabry/orina , Orina/citología , Adulto , Anciano , Enfermedad de Fabry/metabolismo , Femenino , Humanos , Masculino , Persona de Mediana Edad , Proteómica/métodos , Trihexosilceramidas/metabolismo , alfa-Galactosidasa/metabolismo
9.
Genes Brain Behav ; 17(8): e12516, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-30133126

RESUMEN

Adhesion molecules of the immunoglobulin superfamily (IgSF) are essential for neuronal synapse development across evolution and control various aspects of synapse formation and maturation. Neph2, also known as Kirrel3, is an IgSF adhesion molecule implicated in synapse formation, synaptic transmission and ultrastructure. In humans, defects in the NEPH2 gene have been associated with neurodevelopmental disorders such as Jacobsen syndrome, intellectual disability, and autism-spectrum disorders. However, the precise role in development and function of the nervous system is still unclear. Here, we present the histomorphological and phenotypical analysis of a constitutive Neph2-knockout mouse line. Knockout mice display defects in auditory sensory processing, motor skills, and hyperactivity in the home-cage analysis. Olfactory, memory and metabolic testing did not differ from controls. Despite the wide-spread expression of Neph2 in various brain areas, no gross anatomic defects could be observed. Neph2 protein could be located at the cerebellar pinceaux. It interacted with the pinceau core component neurofascin and other synaptic proteins thus suggesting a possible role in cerebellar synapse formation and circuit assembly. Our results suggest that Neph2/Kirrel3 acts on the synaptic ultrastructural level and neuronal wiring rather than on ontogenetic events affecting macroscopic structure. Neph2-knockout mice may provide a valuable rodent model for research on autism spectrum diseases and neurodevelopmental disorders.


Asunto(s)
Proteínas de la Membrana/genética , Proteínas de la Membrana/fisiología , Animales , Proteínas Portadoras/genética , Adhesión Celular/fisiología , Inmunoglobulinas/fisiología , Ratones , Ratones Noqueados , Neurogénesis , Neuronas/metabolismo , Sinapsis/metabolismo
10.
J Cell Biol ; 193(4): 633-42, 2011 May 16.
Artículo en Inglés | MEDLINE | ID: mdl-21555462

RESUMEN

The conserved Hippo signaling pathway regulates organ size in Drosophila melanogaster and mammals and has an essential role in tumor suppression and the control of cell proliferation. Recent studies identified activators of Hippo signaling, but antagonists of the pathway have remained largely elusive. In this paper, we show that NPHP4, a known cilia-associated protein that is mutated in the severe degenerative renal disease nephronophthisis, acts as a potent negative regulator of mammalian Hippo signaling. NPHP4 directly interacted with the kinase Lats1 and inhibited Lats1-mediated phosphorylation of the Yes-associated protein (YAP) and TAZ (transcriptional coactivator with PDZ-binding domain), leading to derepression of these protooncogenic transcriptional regulators. Moreover, NPHP4 induced release from 14-3-3 binding and nuclear translocation of YAP and TAZ, promoting TEA domain (TEAD)/TAZ/YAP-dependent transcriptional activity. Consistent with these data, knockdown of NPHP4 negatively affected cellular proliferation and TEAD/TAZ activity, essentially phenocopying loss of TAZ function. These data identify NPHP4 as a negative regulator of the Hippo pathway and suggest that NPHP4 regulates cell proliferation through its effects on Hippo signaling.


Asunto(s)
Neoplasias de la Mama/metabolismo , Proliferación Celular , Riñón/metabolismo , Proteínas/metabolismo , Transducción de Señal , Proteínas 14-3-3/metabolismo , Aciltransferasas , Animales , Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Proteínas de Ciclo Celular , Línea Celular Tumoral , Femenino , Genes Reporteros , Células HEK293 , Humanos , Inmunoprecipitación , Riñón/patología , Proteínas Nucleares/metabolismo , Fosforilación , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas/genética , Interferencia de ARN , Proteínas Recombinantes de Fusión/metabolismo , Factores de Transcripción/metabolismo , Transfección
11.
Biochemistry ; 45(45): 13429-37, 2006 Nov 14.
Artículo en Inglés | MEDLINE | ID: mdl-17087496

RESUMEN

The blind Drosophila mutant ninaD lacks the visual chromophore. Genetic evidence that the molecular basis is a defect in carotenoid uptake which causes vitamin A deficiency exists. The ninaD gene encodes a scavenger receptor that is significantly homologous in sequence with the mammalian scavenger receptors SR-BI (scavenger receptor class B type I) and CD36 (cluster determinant 36), yet NinaD has not been characterized in functional detail. Therefore, we established a Drosophila S2 cell culture system for biochemically characterizing the ninaD gene products. We show that the two splice variant isoforms encoded by ninaD exhibit different subcellular localizations. NinaD-I, the long protein variant, is localized at the plasma membrane, whereas the short variant, NinaD-II, is localized at intracellular membranes. Only NinaD-I could mediate the cellular uptake of carotenoids from micelles in this cell culture system. Carotenoid uptake was concentration-dependent and saturable. By in vivo analyses of different mutant and transgenic fly strains, we provide evidence of an essential role of NinaD-I in the absorption of dietary carotenoids to support visual chromophore synthesis. Moreover, our analyses suggest a role of NinaD-I in tocopherol metabolism. Even though Drosophila is a sterol auxotroph, we found no evidence of a contribution of NinaD-I to the uptake of these compounds. Together, our study establishes an evolutionarily conserved connection between class B scavenger receptors and the numerous functions of fat soluble vitamins in animal physiology.


Asunto(s)
Antígenos CD36/fisiología , Proteínas de Drosophila/fisiología , Receptores Inmunológicos/fisiología , Empalme Alternativo , Animales , Carotenoides/metabolismo , Membrana Celular/metabolismo , Células Cultivadas , Drosophila melanogaster/metabolismo , Membranas Intracelulares/metabolismo , alfa-Tocoferol/metabolismo
12.
Mol Cell Neurosci ; 30(3): 326-38, 2005 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-16154361

RESUMEN

Reggie/Flotillin proteins are upregulated after optic nerve dissection and evolutionary highly conserved components of lipid rafts. Whereas many biochemical and cell culture studies suggest an involvement in the assembly of multiprotein complexes at cell contact sites, not much is known about their biological in vivo functions. We therefore set out to study the expression pattern and the effects of loss- and gain-of-function in the Drosophila melanogaster model system. We found that in flies these proteins are mainly expressed in axons at the root of fiber tracts, in places where strong fasciculation is required, e.g. at the neck of the peduncle of the mushroom bodies and in the optic chiasms. Despite their evolutionary conservation which implies fundamental and important functions, a P-element-induced null mutant (KG00210) of reggie1/flotillin2 (reggie1/flo2) in D. melanogaster shows no apparent phenotypic defects. This was even more surprising as we show that in this reggie1/flo2 null mutant the paralogous Reggie2/Flo1 protein is unstable and degraded, while the transcript is still present. The requirement of Reggie1/Flo2 for Reggie2/Flo1 stabilization is confirmed by misexpression experiments. Reggie2/Flo1 can only be misexpressed when Reggie1/Flo2 is provided as well. Conversely, Reggie1/Flo2 immunoreactivity can be detected, when its transgene is misexpressed alone. Using appropriate Gal4 driver lines, misexpression of Reggie1/Flo2 alone or together with Reggie2/Flo1 in the eye imaginal disc results in a specific and severe mislocalization of cell adhesion molecules of the immunoglobulin superfamily (IgCAMs) (while DE-Cadherin is unaffected) and in differentiation defects pointing to impaired signaling. In the wing imaginal disc, global overexpression of Reggie/Flotillin proteins leads to a significant extension of the Wingless signal and severely disrupts normal wing development. Our data support the notion that Reggie/Flotillin proteins are implicated in signaling processes at cellular contact sites.


Asunto(s)
Proteínas de Drosophila/metabolismo , Drosophila/embriología , Ojo/embriología , Microdominios de Membrana/metabolismo , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Alas de Animales/embriología , Animales , Axones/metabolismo , Axones/ultraestructura , Moléculas de Adhesión Celular/metabolismo , Secuencia Conservada/genética , Drosophila/genética , Drosophila/metabolismo , Proteínas de Drosophila/genética , Proteínas de Drosophila/aislamiento & purificación , Evolución Molecular , Ojo/metabolismo , Anomalías del Ojo/genética , Anomalías del Ojo/metabolismo , Regulación del Desarrollo de la Expresión Génica/genética , Proteínas de la Membrana/aislamiento & purificación , Cuerpos Pedunculados/citología , Cuerpos Pedunculados/embriología , Cuerpos Pedunculados/metabolismo , Mutación/genética , Quiasma Óptico/citología , Quiasma Óptico/embriología , Quiasma Óptico/metabolismo , Procesamiento Proteico-Postraduccional/genética , Alas de Animales/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA