Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 301
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Cell ; 182(2): 404-416.e14, 2020 07 23.
Artículo en Inglés | MEDLINE | ID: mdl-32610081

RESUMEN

Problems arising during translation of mRNAs lead to ribosome stalling and collisions that trigger a series of quality control events. However, the global cellular response to ribosome collisions has not been explored. Here, we uncover a function for ribosome collisions in signal transduction. Using translation elongation inhibitors and general cellular stress conditions, including amino acid starvation and UV irradiation, we show that ribosome collisions activate the stress-activated protein kinase (SAPK) and GCN2-mediated stress response pathways. We show that the MAPKKK ZAK functions as the sentinel for ribosome collisions and is required for immediate early activation of both SAPK (p38/JNK) and GCN2 signaling pathways. Selective ribosome profiling and biochemistry demonstrate that although ZAK generally associates with elongating ribosomes on polysomal mRNAs, it specifically auto-phosphorylates on the minimal unit of colliding ribosomes, the disome. Together, these results provide molecular insights into how perturbation of translational homeostasis regulates cell fate.


Asunto(s)
Ribosomas/metabolismo , Estrés Fisiológico , Transportadoras de Casetes de Unión a ATP/metabolismo , Anisomicina/farmacología , Apoptosis/efectos de los fármacos , Daño del ADN/efectos de la radiación , Activación Enzimática , Humanos , Quinasas Quinasa Quinasa PAM/deficiencia , Quinasas Quinasa Quinasa PAM/genética , Quinasas Quinasa Quinasa PAM/metabolismo , Proteína Quinasa 14 Activada por Mitógenos/antagonistas & inhibidores , Proteína Quinasa 14 Activada por Mitógenos/metabolismo , Fosforilación , Polirribosomas/metabolismo , Isoformas de Proteínas/deficiencia , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/metabolismo , Interferencia de ARN , ARN Mensajero/metabolismo , ARN Interferente Pequeño/metabolismo , Transducción de Señal , Rayos Ultravioleta , eIF-2 Quinasa/metabolismo
2.
Nature ; 626(8001): 1042-1048, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-38418917

RESUMEN

The loss of the tail is among the most notable anatomical changes to have occurred along the evolutionary lineage leading to humans and to the 'anthropomorphous apes'1-3, with a proposed role in contributing to human bipedalism4-6. Yet, the genetic mechanism that facilitated tail-loss evolution in hominoids remains unknown. Here we present evidence that an individual insertion of an Alu element in the genome of the hominoid ancestor may have contributed to tail-loss evolution. We demonstrate that this Alu element-inserted into an intron of the TBXT gene7-9-pairs with a neighbouring ancestral Alu element encoded in the reverse genomic orientation and leads to a hominoid-specific alternative splicing event. To study the effect of this splicing event, we generated multiple mouse models that express both full-length and exon-skipped isoforms of Tbxt, mimicking the expression pattern of its hominoid orthologue TBXT. Mice expressing both Tbxt isoforms exhibit a complete absence of the tail or a shortened tail depending on the relative abundance of Tbxt isoforms expressed at the embryonic tail bud. These results support the notion that the exon-skipped transcript is sufficient to induce a tail-loss phenotype. Moreover, mice expressing the exon-skipped Tbxt isoform develop neural tube defects, a condition that affects approximately 1 in 1,000 neonates in humans10. Thus, tail-loss evolution may have been associated with an adaptive cost of the potential for neural tube defects, which continue to affect human health today.


Asunto(s)
Empalme Alternativo , Evolución Molecular , Hominidae , Proteínas de Dominio T Box , Cola (estructura animal) , Animales , Humanos , Ratones , Empalme Alternativo/genética , Elementos Alu/genética , Modelos Animales de Enfermedad , Genoma/genética , Hominidae/anatomía & histología , Hominidae/genética , Intrones/genética , Defectos del Tubo Neural/genética , Defectos del Tubo Neural/metabolismo , Fenotipo , Isoformas de Proteínas/deficiencia , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Proteínas de Dominio T Box/deficiencia , Proteínas de Dominio T Box/genética , Proteínas de Dominio T Box/metabolismo , Cola (estructura animal)/anatomía & histología , Cola (estructura animal)/embriología , Exones/genética
3.
Proc Natl Acad Sci U S A ; 118(20)2021 05 18.
Artículo en Inglés | MEDLINE | ID: mdl-33972431

RESUMEN

Febrile seizures (FSs) are the most common convulsion in infancy and childhood. Considering the limitations of current treatments, it is important to examine the mechanistic cause of FSs. Prompted by a genome-wide association study identifying TMEM16C (also known as ANO3) as a risk factor of FSs, we showed previously that loss of TMEM16C function causes hippocampal neuronal hyperexcitability [Feenstra et al., Nat. Genet. 46, 1274-1282 (2014)]. Our previous study further revealed a reduction in the number of warm-sensitive neurons that increase their action potential firing rate with rising temperature of the brain region harboring these hypothalamic neurons. Whereas central neuronal hyperexcitability has been implicated in FSs, it is unclear whether the maximal temperature reached during fever or the rate of body temperature rise affects FSs. Here we report that mutant rodent pups with TMEM16C eliminated from all or a subset of their central neurons serve as FS models with deficient thermoregulation. Tmem16c knockout (KO) rat pups at postnatal day 10 (P10) are more susceptible to hyperthermia-induced seizures. Moreover, they display a more rapid rise of body temperature upon heat exposure. In addition, conditional knockout (cKO) mouse pups (P11) with TMEM16C deletion from the brain display greater susceptibility of hyperthermia-induced seizures as well as deficiency in thermoregulation. We also found similar phenotypes in P11 cKO mouse pups with TMEM16C deletion from Ptgds-expressing cells, including temperature-sensitive neurons in the preoptic area (POA) of the anterior hypothalamus, the brain region that controls body temperature. These findings suggest that homeostatic thermoregulation plays an important role in FSs.


Asunto(s)
Regulación de la Temperatura Corporal/genética , Canales de Cloruro/genética , Fiebre/genética , Hipertermia/genética , Área Preóptica/metabolismo , Convulsiones Febriles/genética , Potenciales de Acción/fisiología , Animales , Animales Recién Nacidos , Temperatura Corporal/efectos de los fármacos , Temperatura Corporal/fisiología , Canales de Cloruro/deficiencia , Femenino , Fiebre/inducido químicamente , Fiebre/metabolismo , Fiebre/fisiopatología , Expresión Génica , Hipocampo/metabolismo , Hipocampo/fisiopatología , Hipertermia/metabolismo , Hipertermia/fisiopatología , Ácido Kaínico/administración & dosificación , Masculino , Ratones , Ratones Noqueados , Neuronas/metabolismo , Neuronas/patología , Área Preóptica/fisiopatología , Isoformas de Proteínas/deficiencia , Isoformas de Proteínas/genética , Ratas , Convulsiones Febriles/inducido químicamente , Convulsiones Febriles/metabolismo , Convulsiones Febriles/fisiopatología
4.
PLoS Genet ; 17(6): e1009603, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-34143769

RESUMEN

The inability to maintain a strictly regulated endo(lyso)somal acidic pH through the proton-pumping action of the vacuolar-ATPases (v-ATPases) has been associated with various human diseases including heritable connective tissue disorders. Autosomal recessive (AR) cutis laxa (CL) type 2C syndrome is associated with genetic defects in the ATP6V1E1 gene and is characterized by skin wrinkles or loose redundant skin folds with pleiotropic systemic manifestations. The underlying pathological mechanisms leading to the clinical presentations remain largely unknown. Here, we show that loss of atp6v1e1b in zebrafish leads to early mortality, associated with craniofacial dysmorphisms, vascular anomalies, cardiac dysfunction, N-glycosylation defects, hypotonia, and epidermal structural defects. These features are reminiscent of the phenotypic manifestations in ARCL type 2C patients. Our data demonstrates that loss of atp6v1e1b alters endo(lyso)somal protein levels, and interferes with non-canonical v-ATPase pathways in vivo. In order to gain further insights into the processes affected by loss of atp6v1e1b, we performed an untargeted analysis of the transcriptome, metabolome, and lipidome in early atp6v1e1b-deficient larvae. We report multiple affected pathways including but not limited to oxidative phosphorylation, sphingolipid, fatty acid, and energy metabolism together with profound defects on mitochondrial respiration. Taken together, our results identify complex pathobiological effects due to loss of atp6v1e1b in vivo.


Asunto(s)
Anomalías Múltiples/genética , Cutis Laxo/genética , Células Epiteliales/metabolismo , Piel/metabolismo , ATPasas de Translocación de Protón Vacuolares/genética , Proteínas de Pez Cebra/genética , Anomalías Múltiples/metabolismo , Anomalías Múltiples/patología , Animales , Cutis Laxo/metabolismo , Cutis Laxo/patología , Modelos Animales de Enfermedad , Endosomas/metabolismo , Endosomas/patología , Células Epiteliales/patología , Regulación de la Expresión Génica , Humanos , Larva/genética , Larva/crecimiento & desarrollo , Larva/metabolismo , Lipidómica , Longevidad/genética , Lisosomas/metabolismo , Lisosomas/patología , Metaboloma/genética , Mitocondrias/metabolismo , Mitocondrias/patología , Fosforilación Oxidativa , Isoformas de Proteínas/deficiencia , Isoformas de Proteínas/genética , Piel/patología , Síndrome , Transcriptoma , ATPasas de Translocación de Protón Vacuolares/deficiencia , Pez Cebra/embriología , Pez Cebra/genética , Pez Cebra/metabolismo , Proteínas de Pez Cebra/deficiencia
5.
FASEB J ; 35(2): e21224, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-33236473

RESUMEN

GABA is synthesized by glutamate decarboxylase (GAD), which has two isoforms, namely, GAD65 and GAD67, encoded by the Gad2 and Gad1 genes, respectively. GAD65-deficient (Gad2-/- ) mice exhibit a reduction in brain GABA content after 1 month of age and show spontaneous seizures in adulthood. Approximately 25% of Gad2-/- mice died by 6 months of age. Our Western blot analysis demonstrated that the protein expression ratio of GAD65 to GAD67 in the brain was greater in rats than in mice during postnatal development, suggesting that the contribution of each GAD isoform to GABA functions differs between these two species. To evaluate whether GAD65 deficiency causes different phenotypes between rats and mice, we generated Gad2-/- rats using TALEN genome editing technology. Western blot and immunohistochemical analyses with new antibodies demonstrated that the GAD65 protein was undetectable in the Gad2-/- rat brain. Gad2-/- pups exhibited spontaneous seizures and paroxysmal discharge in EEG at postnatal weeks 3-4. More than 80% of the Gad2-/- rats died at postnatal days (PNDs) 17-23. GABA content in Gad2-/- brains was significantly lower than those in Gad2+/- and Gad2+/+ brains at PND17-19. These results suggest that the low levels of brain GABA content in Gad2-/- rats may lead to epilepsy followed by premature death, and that Gad2-/- rats are more severely affected than Gad2-/- mice. Considering that the GAD65/GAD67 ratio in human brains is more similar to that in rat brains than in mouse brains, Gad2-/- rats would be useful for further investigating the roles of GAD65 in vivo.


Asunto(s)
Epilepsia/genética , Glutamato Descarboxilasa/genética , Animales , Encéfalo/metabolismo , Encéfalo/fisiopatología , Epilepsia/metabolismo , Glutamato Descarboxilasa/deficiencia , Glutamato Descarboxilasa/metabolismo , Isoformas de Proteínas/deficiencia , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Ratas , Ratas Long-Evans , Receptores de GABA/metabolismo , Potenciales Sinápticos , Ácido gamma-Aminobutírico/metabolismo
6.
Am J Physiol Cell Physiol ; 321(3): C607-C614, 2021 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-34378992

RESUMEN

Bovine milk exosomes (BMEs) are being explored in drug delivery despite their rapid elimination by macrophages. We aimed at identifying the BME transporter in murine bone marrow-derived macrophages (BMDMs). Fluorophore-labeled BMEs were used in transport studies in BMDMs from C57BL/6J and class A scavenger receptor type 1/2 (CASR-1/2) knockout mice and tissue accumulation in macrophage-depleted C57BL/6J mice. Parametric and nonparametric statistics tests for pairwise and multiple comparisons were used. Chemical inhibitors of phagocytosis by cytochalasin D led to a 69 ± 18% decrease in BME uptake compared with controls (P < 0.05), whereas inhibitors of endocytic pathways other than phagocytosis had a modest effect on uptake (P > 0.05). Inhibitors of class A scavenger receptors (CASRs) including CASR-1/2 caused a 70% decrease in BME uptake (P < 0.05). The uptake of BMEs by BMDMs from CASR-1/2 knockout mice was smaller by 58 ± 23% compared with wild-type controls (P < 0.05). Macrophage depletion by clodronate caused a more than 44% decrease in BME uptake in the spleen and lungs (P < 0.05), whereas the decrease observed in liver was not statistically significant. In conclusion, CASR-1/2 facilitates the uptake of BMEs in BMDMs and C57BL/6J mice.


Asunto(s)
Exosomas/metabolismo , Macrófagos/metabolismo , Leche/química , Receptores Depuradores de Clase A/genética , Animales , Bovinos , Ácido Clodrónico/farmacología , Citocalasina D/farmacología , Endocitosis/efectos de los fármacos , Exosomas/química , Femenino , Colorantes Fluorescentes/química , Expresión Génica , Hígado/efectos de los fármacos , Hígado/metabolismo , Pulmón/efectos de los fármacos , Pulmón/metabolismo , Macrófagos/citología , Macrófagos/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Fagocitosis/efectos de los fármacos , Isoformas de Proteínas/deficiencia , Isoformas de Proteínas/genética , Receptores Depuradores de Clase A/deficiencia , Bazo/efectos de los fármacos , Bazo/metabolismo , Coloración y Etiquetado/métodos
7.
RNA ; 25(1): 45-59, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30314980

RESUMEN

Cell migration requires a complicated network of structural and regulatory proteins. Changes in cellular motility can impact migration as a result of cell-type or developmental stage regulated expression of critical motility genes. Hnrnpab is a conserved RNA-binding protein found as two isoforms produced by alternative splicing. Its expression is enriched in the subventricular zone (SVZ) and the rostral migratory stream within the brain, suggesting possible support of the migration of neural progenitor cells in this region. Here we show that the migration of cells from the SVZ of developing Hnrnpab-/- mouse brains is impaired. An RNA-seq analysis to identify Hnrnpab-dependent cell motility genes led us to Eps8, and in agreement with the change in cell motility, we show that Eps8 is decreased in Hnrnpab-/- SVZ tissue. We scrutinized the motility of Hnrnpab-/- cells and confirmed that the decreases in both cell motility and Eps8 are restored by ectopically coexpressing both alternatively spliced Hnrnpab isoforms, therefore these variants are surprisingly nonredundant for cell motility. Our results support a model where both Hnrnpab isoforms work in concert to regulate Eps8 transcription in the mouse SVZ to promote the normal migration of neural cells during CNS development.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/genética , Ribonucleoproteína Heterogénea-Nuclear Grupo A-B/fisiología , Neuronas/fisiología , Empalme Alternativo , Animales , Línea Celular , Movimiento Celular/genética , Regulación del Desarrollo de la Expresión Génica , Ribonucleoproteína Heterogénea-Nuclear Grupo A-B/deficiencia , Ribonucleoproteína Heterogénea-Nuclear Grupo A-B/genética , Humanos , Ventrículos Laterales/citología , Ventrículos Laterales/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Modelos Neurológicos , Isoformas de Proteínas/deficiencia , Isoformas de Proteínas/genética , Isoformas de Proteínas/fisiología , Motivo de Reconocimiento de ARN , Análisis de Secuencia de ARN , Transcripción Genética
8.
J Immunol ; 203(8): 2141-2149, 2019 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-31541023

RESUMEN

Foxo3 acts as an important central regulator that integrates signaling pathways and coordinates cellular responses to environmental changes. Recent studies show the involvement of Foxo3 in osteoclastogenesis and rheumatoid arthritis, which prompted us to further investigate the FOXO3 locus. Several databases document FOXO3 isoform2, an N-terminal truncated mutation of the full-length FOXO3 However, the biological function of FOXO3 isoform2 is unclear. In this study, we established a conditional allele of Foxo3 in mice that deletes the full-length Foxo3 except isoform2, a close ortholog of the human FOXO3 isoform2. Expression of Foxo3 isoform2 specifically in macrophage/osteoclast lineage suppresses osteoclastogenesis and leads to the osteopetrotic phenotype in mice. Mechanistically, Foxo3 isoform2 enhances the expression of type I IFN response genes to RANKL stimulation and thus inhibits osteoclastogenesis via endogenous IFN-ß-mediated feedback inhibition. Our findings identify, to our knowledge, the first known biological function of Foxo3 isoform2 that acts as a novel osteoclastic inhibitor in bone remodeling.


Asunto(s)
Proteína Forkhead Box O3/metabolismo , Osteoclastos/citología , Osteoclastos/metabolismo , Animales , Diferenciación Celular/efectos de los fármacos , Células Cultivadas , Proteína Forkhead Box O3/deficiencia , Células HEK293 , Humanos , Interferón Tipo I/farmacología , Ratones , Ratones Endogámicos C57BL , Osteoclastos/efectos de los fármacos , Osteogénesis/efectos de los fármacos , Fenotipo , Isoformas de Proteínas/análisis , Isoformas de Proteínas/deficiencia , Isoformas de Proteínas/metabolismo , Células RAW 264.7
9.
Proc Natl Acad Sci U S A ; 115(45): E10768-E10777, 2018 11 06.
Artículo en Inglés | MEDLINE | ID: mdl-30348775

RESUMEN

Plant immune responses mediated by the hormone jasmonoyl-l-isoleucine (JA-Ile) are metabolically costly and often linked to reduced growth. Although it is known that JA-Ile activates defense responses by triggering the degradation of JASMONATE ZIM DOMAIN (JAZ) transcriptional repressor proteins, expansion of the JAZ gene family in vascular plants has hampered efforts to understand how this hormone impacts growth and other physiological tasks over the course of ontogeny. Here, we combined mutations within the 13-member Arabidopsis JAZ gene family to investigate the effects of chronic JAZ deficiency on growth, defense, and reproductive output. A higher-order mutant (jaz decuple, jazD) defective in 10 JAZ genes (JAZ1-7, -9, -10, and -13) exhibited robust resistance to insect herbivores and fungal pathogens, which was accompanied by slow vegetative growth and poor reproductive performance. Metabolic phenotypes of jazD discerned from global transcript and protein profiling were indicative of elevated carbon partitioning to amino acid-, protein-, and endoplasmic reticulum body-based defenses controlled by the JA-Ile and ethylene branches of immunity. Resource allocation to a strong defense sink in jazD leaves was associated with increased respiration and hallmarks of carbon starvation but no overt changes in photosynthetic rate. Depletion of the remaining JAZ repressors in jazD further exaggerated growth stunting, nearly abolished seed production and, under extreme conditions, caused spreading necrotic lesions and tissue death. Our results demonstrate that JAZ proteins promote growth and reproductive success at least in part by preventing catastrophic metabolic effects of an unrestrained immune response.


Asunto(s)
Proteínas de Arabidopsis/genética , Arabidopsis/genética , Ciclopentanos/metabolismo , Regulación de la Expresión Génica de las Plantas/inmunología , Aptitud Genética/inmunología , Isoleucina/análogos & derivados , Enfermedades de las Plantas/genética , Proteínas Represoras/genética , Animales , Arabidopsis/inmunología , Arabidopsis/microbiología , Arabidopsis/parasitología , Carbono/metabolismo , Resistencia a la Enfermedad/genética , Hongos/crecimiento & desarrollo , Hongos/patogenicidad , Perfilación de la Expresión Génica , Regulación del Desarrollo de la Expresión Génica , Insectos/patogenicidad , Insectos/fisiología , Isoleucina/metabolismo , Redes y Vías Metabólicas/genética , Redes y Vías Metabólicas/inmunología , Familia de Multigenes , Mutación , Enfermedades de las Plantas/inmunología , Enfermedades de las Plantas/microbiología , Enfermedades de las Plantas/parasitología , Inmunidad de la Planta/genética , Isoformas de Proteínas/deficiencia , Isoformas de Proteínas/genética , Proteínas Represoras/deficiencia , Reproducción/genética , Reproducción/inmunología , Transducción de Señal
10.
Int J Mol Sci ; 22(20)2021 Oct 12.
Artículo en Inglés | MEDLINE | ID: mdl-34681657

RESUMEN

BACKGROUND: The transient receptor potential ankyrin 1 (TRPA1) cation channels function as broadly-tuned sensors of noxious chemicals in many species. Recent studies identified four functional TRPA1 isoforms in Drosophila melanogaster (dTRPA1(A) to (D)), but their responses to non-electrophilic chemicals are yet to be fully characterized. METHODS: We determined the behavioral responses of adult flies to the mammalian TRPA1 non-electrophilic activators citronellal and menthol, and characterized the effects of these compounds on all four dTRPA1 channel isoforms using intracellular Ca2+ imaging and whole-cell patch-clamp recordings. RESULTS: Wild type flies avoided citronellal and menthol in an olfactory test and this behavior was reduced in dTrpA1 mutant flies. Both compounds activate all dTRPA1 isoforms in the heterologous expression system HEK293T, with the following sensitivity series: dTRPA1(C) = dTRPA1(D) > dTRPA1(A) ≫ dTRPA1(B) for citronellal and dTRPA1(A) > dTRPA1(D) > dTRPA1(C) > dTRPA1(B) for menthol. CONCLUSIONS: dTrpA1 was required for the normal avoidance of Drosophila melanogaster towards citronellal and menthol. All dTRPA1 isoforms are activated by both compounds, but the dTRPA1(B) is consistently the least sensitive. We discuss how these findings may guide further studies on the physiological roles and the structural bases of chemical sensitivity of TRPA1 channels.


Asunto(s)
Monoterpenos Acíclicos/farmacología , Aldehídos/farmacología , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/efectos de los fármacos , Mentol/farmacología , Canal Catiónico TRPA1/metabolismo , Animales , Animales Modificados Genéticamente/metabolismo , Calcio/metabolismo , Proteínas de Drosophila/deficiencia , Proteínas de Drosophila/genética , Drosophila melanogaster/metabolismo , Femenino , Células HEK293 , Humanos , Repelentes de Insectos/farmacología , Masculino , Técnicas de Placa-Clamp , Isoformas de Proteínas/deficiencia , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Canal Catiónico TRPA1/deficiencia , Canal Catiónico TRPA1/genética
11.
Int J Mol Sci ; 22(11)2021 May 30.
Artículo en Inglés | MEDLINE | ID: mdl-34070744

RESUMEN

The ClC-2 channel plays a critical role in maintaining ion homeostasis in the brain and the testis. Loss-of-function mutations in the ClC-2-encoding human CLCN2 gene are linked to the white matter disease leukodystrophy. Clcn2-deficient mice display neuronal myelin vacuolation and testicular degeneration. Leukodystrophy-causing ClC-2 mutant channels are associated with anomalous proteostasis manifesting enhanced endoplasmic reticulum (ER)-associated degradation. The molecular nature of the ER quality control system for ClC-2 protein remains elusive. In mouse testicular tissues and Leydig cells, we demonstrated that endogenous ClC-2 co-existed in the same protein complex with the molecular chaperones heat shock protein 90ß (Hsp90ß) and heat shock cognate protein (Hsc70), as well as the associated co-chaperones Hsp70/Hsp90 organizing protein (HOP), activator of Hsp90 ATPase homolog 1 (Aha1), and FK506-binding protein 8 (FKBP8). Further biochemical analyses revealed that the Hsp90ß-Hsc70 chaperone/co-chaperone system promoted mouse and human ClC-2 protein biogenesis. FKBP8 additionally facilitated membrane trafficking of ClC-2 channels. Interestingly, treatment with the Hsp90-targeting small molecule 17-allylamino-17-demethoxygeldanamycin (17-AAG) substantially boosted ClC-2 protein expression. Also, 17-AAG effectively increased both total and cell surface protein levels of leukodystrophy-causing loss-of-function ClC-2 mutant channels. Our findings highlight the therapeutic potential of 17-AAG in correcting anomalous ClC-2 proteostasis associated with leukodystrophy.


Asunto(s)
Encéfalo/metabolismo , Canales de Cloruro/genética , Células Intersticiales del Testículo/metabolismo , Neuronas/metabolismo , Enfermedad de Pelizaeus-Merzbacher/genética , Proteostasis/genética , Animales , Benzoquinonas/farmacología , Encéfalo/efectos de los fármacos , Encéfalo/patología , Células CHO , Canales de Cloruro CLC-2 , Canales de Cloruro/deficiencia , Cricetulus , Modelos Animales de Enfermedad , Degradación Asociada con el Retículo Endoplásmico/efectos de los fármacos , Regulación de la Expresión Génica , Células HEK293 , Proteínas HSP70 de Choque Térmico/genética , Proteínas HSP70 de Choque Térmico/metabolismo , Proteínas HSP90 de Choque Térmico/genética , Proteínas HSP90 de Choque Térmico/metabolismo , Humanos , Lactamas Macrocíclicas/farmacología , Células Intersticiales del Testículo/efectos de los fármacos , Células Intersticiales del Testículo/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Chaperonas Moleculares/genética , Chaperonas Moleculares/metabolismo , Neuronas/efectos de los fármacos , Neuronas/patología , Enfermedad de Pelizaeus-Merzbacher/tratamiento farmacológico , Enfermedad de Pelizaeus-Merzbacher/metabolismo , Enfermedad de Pelizaeus-Merzbacher/patología , Isoformas de Proteínas/deficiencia , Isoformas de Proteínas/genética , Transducción de Señal , Proteínas de Unión a Tacrolimus/genética , Proteínas de Unión a Tacrolimus/metabolismo
12.
Infect Immun ; 88(9)2020 08 19.
Artículo en Inglés | MEDLINE | ID: mdl-32631916

RESUMEN

Enteric pathogens exploit chemical and nutrient signaling to gauge their location within a host and control expression of traits important for infection. Ethanolamine-containing molecules are essential in host physiology and play important roles in intestinal processes. The transcription factor EutR is conserved in the Enterobacteriaceae and is required for ethanolamine sensing and metabolism. In enterohemorrhagic Escherichia coli (EHEC) O157:H7, EutR responds to ethanolamine to activate expression of traits required for host colonization and disease; however, the importance of EutR to EHEC intestinal infection has not been examined. Because EHEC does not naturally colonize or cause disease in mice, we employed the natural murine pathogen Citrobacter rodentium as a model of EHEC virulence to investigate the importance of EutR in vivo EHEC and C. rodentium possess the locus of enterocyte effacement (LEE), which is the canonical virulence trait of attaching and effacing pathogens. Our findings demonstrate that ethanolamine sensing and EutR-dependent regulation of the LEE are conserved in C. rodentium Moreover, during infection, EutR is required for maximal LEE expression, colonization, and transmission efficiency. These findings reveal that EutR not only is important for persistence during the primary host infection cycle but also is required for maintenance in a host population.


Asunto(s)
Citrobacter rodentium/genética , Infecciones por Enterobacteriaceae/microbiología , Escherichia coli Enterohemorrágica/genética , Proteínas de Escherichia coli/genética , Etanolamina/metabolismo , Regulación Bacteriana de la Expresión Génica , Fosfoproteínas/genética , Factores de Transcripción/genética , Animales , Citrobacter rodentium/patogenicidad , Recuento de Colonia Microbiana , Secuencia Conservada , Infecciones por Enterobacteriaceae/genética , Infecciones por Enterobacteriaceae/patología , Infecciones por Enterobacteriaceae/transmisión , Enterocitos/microbiología , Enterocitos/patología , Escherichia coli Enterohemorrágica/patogenicidad , Proteínas de Escherichia coli/metabolismo , Femenino , Interacciones Microbiota-Huesped/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Fosfoproteínas/metabolismo , Isoformas de Proteínas/deficiencia , Isoformas de Proteínas/genética , Transducción de Señal , Factores de Transcripción/deficiencia , Virulencia
13.
Cell Microbiol ; 21(8): e13033, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-31009148

RESUMEN

Vibrio cholerae produced-Cholix toxin (Cholix) is a cytotoxin that ADP-ribosylates eukaryotic elongation factor 2, inhibiting protein synthesis, and inducing apoptosis. Here, we identified prohibitin (PHB) 1 and 2 as novel Cholix-interacting membrane proteins in immortalised human hepatocytes and HepG2 cells by Cholix immunoprecipitation assays. The expression level of PHB1 was decreased by Cholix after a 12hr incubation. Cholix-induced poly (ADP-ribose) polymerase (PARP) cleavage was significantly enhanced in PHB (PHB1 or PHB2) knockdown cells. In contrast, transiently overexpressed PHB in hepatocytes attenuated Cholix-induced Bax/Bak conformational changes and PARP cleavage. In addition, Cholix-induced reactive oxygen species production and accumulation of fragmented mitochondria were enhanced in PHB-knockdown cells. Furthermore, Cholix induced activation of Rho-associated coiled coil-containing protein kinase 1 (ROCK1), which was enhanced in PHB-knockdown cells, followed by actin filament depolymerisation and accumulation of tubulin in the blebbing cells. Inhibition of ROCK1 by siRNA or its inhibitor suppressed Cholix-induced PARP cleavage and reactive oxygen species generation. Our findings identify PHB as a new protein that interacts with Cholix and is involved in Cholix-induced mitochondrial dysfunction and cytoskeletal rearrangement by ROCK1 activation during apoptosis.


Asunto(s)
Factores de Ribosilacion-ADP/química , Toxinas Bacterianas/química , Factor 2 Eucariótico de Iniciación/metabolismo , Interacciones Huésped-Patógeno/genética , Especies Reactivas de Oxígeno/metabolismo , Proteínas Represoras/genética , Vibrio cholerae/genética , ADP-Ribosilación , Factores de Ribosilacion-ADP/genética , Factores de Ribosilacion-ADP/metabolismo , Citoesqueleto de Actina/metabolismo , Citoesqueleto de Actina/microbiología , Citoesqueleto de Actina/ultraestructura , Secuencia de Aminoácidos , Apoptosis/genética , Toxinas Bacterianas/genética , Toxinas Bacterianas/metabolismo , Línea Celular Transformada , Factor 2 Eucariótico de Iniciación/genética , Regulación de la Expresión Génica , Células Hep G2 , Hepatocitos/metabolismo , Hepatocitos/microbiología , Hepatocitos/patología , Humanos , Mitocondrias/metabolismo , Mitocondrias/microbiología , Mitocondrias/ultraestructura , Prohibitinas , Unión Proteica , Isoformas de Proteínas/deficiencia , Isoformas de Proteínas/genética , Proteolisis , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Proteínas Represoras/deficiencia , Transducción de Señal , Tubulina (Proteína)/genética , Tubulina (Proteína)/metabolismo , Vibrio cholerae/metabolismo , Vibrio cholerae/patogenicidad , Virulencia , Proteína Destructora del Antagonista Homólogo bcl-2/genética , Proteína Destructora del Antagonista Homólogo bcl-2/metabolismo , Proteína X Asociada a bcl-2/genética , Proteína X Asociada a bcl-2/metabolismo , Quinasas Asociadas a rho/antagonistas & inhibidores , Quinasas Asociadas a rho/genética , Quinasas Asociadas a rho/metabolismo
14.
Nature ; 511(7510): 466-70, 2014 Jul 24.
Artículo en Inglés | MEDLINE | ID: mdl-24896188

RESUMEN

Because most neurons receive thousands of synaptic inputs, the neuronal membrane is a mosaic of specialized microdomains where neurotransmitter receptors cluster in register with the corresponding presynaptic neurotransmitter release sites. In many cases the coordinated differentiation of presynaptic and postsynaptic domains implicates trans-synaptic interactions between membrane-associated proteins such as neurexins and neuroligins. The Caenorhabditis elegans neuromuscular junction (NMJ) provides a genetically tractable system in which to analyse the segregation of neurotransmitter receptors, because muscle cells receive excitatory innervation from cholinergic neurons and inhibitory innervation from GABAergic neurons. Here we show that Ce-Punctin/madd-4 (ref. 5), the C. elegans orthologue of mammalian punctin-1 and punctin-2, encodes neurally secreted isoforms that specify the excitatory or inhibitory identity of postsynaptic NMJ domains. These proteins belong to the ADAMTS (a disintegrin and metalloprotease with thrombospondin repeats)-like family, a class of extracellular matrix proteins related to the ADAM proteases but devoid of proteolytic activity. Ce-Punctin deletion causes the redistribution of synaptic acetylcholine and GABAA (γ-aminobutyric acid type A) receptors into extrasynaptic clusters, whereas neuronal presynaptic boutons remain unaltered. Alternative promoters generate different Ce-Punctin isoforms with distinct functions. A short isoform is expressed by cholinergic and GABAergic motoneurons and localizes to excitatory and inhibitory NMJs, whereas long isoforms are expressed exclusively by cholinergic motoneurons and are confined to cholinergic NMJs. The differential expression of these isoforms controls the congruence between presynaptic and postsynaptic domains: specific disruption of the short isoform relocalizes GABAA receptors from GABAergic to cholinergic synapses, whereas expression of a long isoform in GABAergic neurons recruits acetylcholine receptors to GABAergic NMJs. These results identify Ce-Punctin as a previously unknown synaptic organizer and show that presynaptic and postsynaptic domain identities can be genetically uncoupled in vivo. Because human punctin-2 was identified as a candidate gene for schizophrenia, ADAMTS-like proteins may also control synapse organization in the mammalian central nervous system.


Asunto(s)
Proteínas de Caenorhabditis elegans/metabolismo , Caenorhabditis elegans/metabolismo , Neuronas Colinérgicas/metabolismo , Neuronas GABAérgicas/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Densidad Postsináptica/metabolismo , Proteínas ADAM/metabolismo , Acetilcolina/metabolismo , Animales , Proteínas de Caenorhabditis elegans/química , Proteínas de la Matriz Extracelular/metabolismo , Neuronas Motoras/metabolismo , Proteínas del Tejido Nervioso/química , Proteínas del Tejido Nervioso/deficiencia , Unión Neuromuscular , Isoformas de Proteínas/química , Isoformas de Proteínas/deficiencia , Isoformas de Proteínas/metabolismo , Receptores Colinérgicos/metabolismo , Receptores de GABA-A/metabolismo
15.
J Neurosci ; 38(44): 9579-9599, 2018 10 31.
Artículo en Inglés | MEDLINE | ID: mdl-30232223

RESUMEN

We created a neural-specific conditional murine glut3 (Slc2A3) deletion (glut3flox/flox/nestin-Cre+) to examine the effect of a lack of Glut3 on neurodevelopment. Compared with age-matched glut3flox/flox = WT and heterozygotes (glut3flox/+/nestin-Cre+), we found that a >90% reduction in male and female brain Glut3 occurred by postnatal day 15 (PN15) in glut3flox/flox/nestin-Cre+ This genetic manipulation caused a diminution in brain weight and cortical thickness at PN15, a reduced number of dendritic spines, and fewer ultrasonic vocalizations. Patch-clamp recordings of cortical pyramidal neurons revealed increased frequency of bicuculline-induced paroxysmal discharges as well as reduced latency, attesting to a functional synaptic and cortical hyperexcitability. Concomitant stunting with lower glucose concentrations despite increased milk intake shortened the lifespan, failing rescue by a ketogenic diet. This led to creating glut3flox/flox/CaMK2α-Cre+ mice lacking Glut3 in the adult male limbic system. These mice had normal lifespan, displayed reduced IPSCs in cortical pyramidal neurons, less anxiety/fear, and lowered spatial memory and motor abilities but heightened exploratory and social responses. These distinct postnatal and adult phenotypes, based upon whether glut3 gene is globally or restrictively absent, have implications for humans who carry copy number variations and present with neurodevelopmental disorders.SIGNIFICANCE STATEMENT Lack of the key brain-specific glucose transporter 3 gene found in neurons during early postnatal life results in significant stunting, a reduction in dendritic spines found on neuronal processes and brain size, heightened neuronal excitability, along with a shortened lifespan. When occurring in the adult and limited to the limbic system alone, lack of this gene in neurons reduces the fear of spatial exploration and socialization but does not affect the lifespan. These features are distinct heralding differences between postnatal and adult phenotypes based upon whether the same gene is globally or restrictively lacking. These findings have implications for humans who carry copy number variations pertinent to this gene and have been described to present with neurodevelopmental disorders.


Asunto(s)
Encéfalo/metabolismo , Conducta Exploratoria/fisiología , Eliminación de Gen , Transportador de Glucosa de Tipo 3/deficiencia , Transportador de Glucosa de Tipo 3/genética , Fenotipo , Factores de Edad , Animales , Animales Recién Nacidos , Encéfalo/patología , Espinas Dendríticas/genética , Espinas Dendríticas/metabolismo , Espinas Dendríticas/patología , Femenino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Embarazo , Isoformas de Proteínas/deficiencia , Isoformas de Proteínas/genética
16.
Can J Physiol Pharmacol ; 97(7): 623-630, 2019 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-30856349

RESUMEN

Structural dilation of cardiomyocytes (CMs) imposes a decline in cardiac performance that precipitates cardiac failure and sudden death. Since membrane proteins are implicated in dilated cardiomyopathy and heart failure, we evaluated the expression of the sarcolemmal membrane-associated protein (SLMAP) in dilated cardiomyopathy and its effect on CM contraction. We found that all 3 SLMAP isoforms (SLMAP-1, -2, and -3) are expressed in CMs and are downregulated in human dilated ventricles. Knockdown of SLMAPs in cultured CMs transduced with recombinant adeno-associated viral particles releasing SLMAP-shRNA precipitated reduced spontaneous contractile rate that was not fully recovered in SLMAP-depleted CMs challenged with isoproterenol (ISO), thus phenotypically mimicking heart failure performance. Interestingly, the overexpression of the SLMAP-3 full-length isoform induced a positive chronotropic effect in CMs that was more pronounced in response to ISO insult (vs. ISO-treated naïve CMs). Confocal live imaging showed that H9c2 cardiac myoblasts overexpressing SLMAP-3 exhibit a higher intracellular calcium transient peak when treated with ISO (vs. ISO-treated cells carrying a control adeno-associated viral particle). Proteomics revealed that SLMAP-3 interacts with the regulator of CM contraction, striatin. Collectively, our data demonstrate that SLMAP-3 is a novel regulator of CM contraction rate and their response to adrenergic stimuli. Loss of SLMAPs phenotypically mimics cardiac failure and crystallizes SLMAPs as predictive of dilated cardiomyopathy and heart failure.


Asunto(s)
Calcio/metabolismo , Cardiomiopatía Dilatada/metabolismo , Regulación hacia Abajo , Ventrículos Cardíacos/metabolismo , Espacio Intracelular/metabolismo , Proteínas de la Membrana/metabolismo , Miocitos Cardíacos/metabolismo , Animales , Cardiomiopatía Dilatada/tratamiento farmacológico , Cardiomiopatía Dilatada/patología , Cardiomiopatía Dilatada/fisiopatología , Línea Celular , Regulación hacia Abajo/efectos de los fármacos , Técnicas de Silenciamiento del Gen , Ventrículos Cardíacos/efectos de los fármacos , Humanos , Espacio Intracelular/efectos de los fármacos , Isoproterenol/farmacología , Proteínas de la Membrana/deficiencia , Proteínas de la Membrana/genética , Terapia Molecular Dirigida , Contracción Miocárdica/efectos de los fármacos , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/patología , Isoformas de Proteínas/deficiencia , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Ratas
17.
Proc Natl Acad Sci U S A ; 113(13): 3663-8, 2016 Mar 29.
Artículo en Inglés | MEDLINE | ID: mdl-26976581

RESUMEN

The clinical management of severe pain depends heavily on opioids acting through mu opioid receptors encoded by the Oprm1 gene, which undergoes extensive alternative splicing. In addition to generating a series of prototypic seven transmembrane domain (7TM) G protein-coupled receptors (GPCRs), Oprm1 also produces a set of truncated splice variants containing only six transmembrane domains (6TM) through which selected opioids such as IBNtxA (3'-iodobenzoyl-6ß-naltrexamide) mediate a potent analgesia without many undesirable effects. Although morphine analgesia is independent of these 6TM mu receptor isoforms, we now show that the selective loss of the 6TM variants in a knockout model eliminates the analgesic actions of delta and kappa opioids and of α2-adrenergic compounds, but not cannabinoid, neurotensin, or muscarinic drugs. These observations were confirmed by using antisense paradigms. Despite their role in analgesia, loss of the 6TM variants were not involved with delta opioid-induced seizure activity, aversion to the kappa drug U50, 488H, or α2-mediated hypolocomotion. These observations support the existence of parallel opioid and nonopioid pain modulatory systems and highlight the ability to dissociate unwanted delta, kappa1, and α2 actions from analgesia.


Asunto(s)
Receptores Opioides mu/genética , Receptores Opioides mu/fisiología , Empalme Alternativo , Analgesia , Analgésicos Opioides/farmacología , Animales , Femenino , Humanos , Masculino , Ratones , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Noqueados , Morfina/farmacología , Naltrexona/análogos & derivados , Naltrexona/farmacología , Manejo del Dolor , Dimensión del Dolor , Isoformas de Proteínas/deficiencia , Isoformas de Proteínas/genética , Isoformas de Proteínas/fisiología , Estructura Terciaria de Proteína , Receptores Opioides mu/deficiencia
18.
Cell Physiol Biochem ; 45(4): 1434-1443, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29466783

RESUMEN

BACKGROUND/AIMS: Fibroblast growth factor 2 (FGF2) plays a predominant role during angiogenesis in the adventitia and in atherosclerotic plaque. A dilemma exists, however, as to whether angiogenic stimulation by FGF2 for the prevention and treatment of atherogenesis is feasible. The aim of this study is to investigate the effect of the 18-kDa FGF-2 isoform on atherosclerosis progression in high-fat diet-fed apolipoprotein E knockout (ApoE-/-) mice. METHODS: We established a model of atherosclerosis using ApoE and 18-kDa FGF-2 gene double knockout mice. They were randomly divided into three groups depending on the duration of diet: 8 weeks, 12 weeks and 16 weeks. Then, we studied the morphology and inflammatory factor staining in the atherosclerosis plaque of these mice. RESULTS: Knockout of the 18-kDa FGF-2 isoform did not change the metabolic characteristics of the mice. Compared to the control group, knockout of the 18-kDa FGF-2 isoform significantly attenuated atherogenesis, reduced aortic plaques, reduced macrophage infiltration and suppressed oxidative stress in mice fed with a high fat diet at all-time points. CONCLUSIONS: 18-kDa FGF-2 aggravated the inflammatory reaction of atherosclerosis.


Asunto(s)
Aterosclerosis/patología , Factor 2 de Crecimiento de Fibroblastos/genética , Macrófagos/inmunología , Estrés Oxidativo , Animales , Aorta/metabolismo , Aorta/patología , Apolipoproteínas E/deficiencia , Apolipoproteínas E/genética , Aterosclerosis/metabolismo , Quimiocina CCL2/metabolismo , Dieta Alta en Grasa , Modelos Animales de Enfermedad , Factor 2 de Crecimiento de Fibroblastos/deficiencia , Genotipo , Lípidos/sangre , Macrófagos/patología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Peso Molecular , Miocardio/patología , NADPH Oxidasa 4/metabolismo , Isoformas de Proteínas/deficiencia , Isoformas de Proteínas/genética , Molécula 1 de Adhesión Celular Vascular/metabolismo
19.
Biochem Biophys Res Commun ; 496(2): 339-345, 2018 02 05.
Artículo en Inglés | MEDLINE | ID: mdl-29331378

RESUMEN

Sarcomeric protein turnover needs to be tightly balanced to assure proper assembly and renewal of sarcomeric units within muscle tissues. The mechanisms regulating these fundamental processes are only poorly understood, but of great clinical importance since many cardiac and skeletal muscle diseases are associated with defective sarcomeric organization. The SET- and MYND domain containing protein 1b (Smyd1b) is known to play a crucial role in myofibrillogenesis by functionally interacting with the myosin chaperones Unc45b and Hsp90α1. In zebrafish, Smyd1b, Unc45b and Hsp90α1 are part of the misfolded myosin response (MMR), a regulatory transcriptional response that is activated by disturbed myosin homeostasis. Genome duplication in zebrafish led to a second smyd1 gene, termed smyd1a. Morpholino- and CRISPR/Cas9-mediated knockdown of smyd1a led to significant perturbations in sarcomere structure resulting in decreased cardiac as well as skeletal muscle function. Similar to Smyd1b, we found Smyd1a to localize to the sarcomeric M-band in skeletal and cardiac muscles. Overexpression of smyd1a efficiently compensated for the loss of Smyd1b in flatline (fla) mutant zebrafish embryos, rescued the myopathic phenotype and suppressed the MMR in Smyd1b-deficient embryos, suggesting overlapping functions of both Smyd1 paralogs. Interestingly, Smyd1a is not transcriptionally activated in Smyd1b-deficient fla mutants, demonstrating lack of genetic compensation despite the functional redundancy of both zebrafish Smyd1 paralogs.


Asunto(s)
Regulación del Desarrollo de la Expresión Génica , N-Metiltransferasa de Histona-Lisina/genética , Músculo Esquelético/metabolismo , Miocitos Cardíacos/metabolismo , Miosinas/genética , Sarcómeros/metabolismo , Proteínas de Pez Cebra/genética , Pez Cebra/genética , Animales , Animales Modificados Genéticamente , Sistemas CRISPR-Cas , Embrión no Mamífero , Duplicación de Gen , Edición Génica , Genes Reporteros , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Proteínas HSP90 de Choque Térmico/genética , Proteínas HSP90 de Choque Térmico/metabolismo , N-Metiltransferasa de Histona-Lisina/antagonistas & inhibidores , N-Metiltransferasa de Histona-Lisina/deficiencia , Humanos , Chaperonas Moleculares/genética , Chaperonas Moleculares/metabolismo , Morfolinos/genética , Morfolinos/metabolismo , Proteínas Musculares , Músculo Esquelético/patología , Miocitos Cardíacos/patología , Miosinas/metabolismo , Pliegue de Proteína , Isoformas de Proteínas/deficiencia , Isoformas de Proteínas/genética , Sarcómeros/patología , Pez Cebra/crecimiento & desarrollo , Pez Cebra/metabolismo , Proteínas de Pez Cebra/antagonistas & inhibidores , Proteínas de Pez Cebra/deficiencia , Proteínas de Pez Cebra/metabolismo
20.
Cell Commun Signal ; 16(1): 35, 2018 06 28.
Artículo en Inglés | MEDLINE | ID: mdl-29954386

RESUMEN

BACKGROUND: Loss of PTEN is involved in tumor progression of several tumor entities including renal cell carcinoma (RCC). During the translation process PTEN generates a number of splice variants, including PTEN-Δ. We analyzed the impact of PTEN-Δ in RCC progression. METHODS: In specimens of RCC patients the expression of PTEN-Δ and PTEN was quantified. The PTEN expressing RCC cell line A498 and the PTEN deficient 786-O cell line were stably transfected with the PTEN-Δ or PTEN transcript. In Caki-1 cells that highly express PTEN-Δ, this isoform was knocked down by siRNA. Cell migration, adhesion, apoptosis and signaling pathways activities were consequently analyzed in vitro. RESULTS: Patients with a higher PTEN-Δ expression had a longer lymph node metastasis free and overall survival. In RCC specimens, the PTEN-Δ expression correlated with the PTEN expression. PTEN-Δ as well as PTEN induced a reduced migration when using extracellular matrix (ECM) compounds as chemotaxins. This effect was confirmed by knockdown of PTEN-Δ, inducing an enhanced migration. Likewise a decreased adhesion on these ECM components could be shown in PTEN-Δ and PTEN transfected cells. The apoptosis rate was slightly increased by PTEN-Δ. In a phospho-kinase array and Western blot analyses a consequently reduced activity of AKT, p38 and JNK could be shown. CONCLUSIONS: We could show that the PTEN splice variant PTEN-Δ acts similar to PTEN in a tumor suppressive manner, suggesting synergistic effects of the two isoforms. The impact of PTEN-Δ in context of tumor progression should thus be taken into account when generating new therapeutic options targeting PTEN signaling in RCC.


Asunto(s)
Carcinoma de Células Renales/metabolismo , Carcinoma de Células Renales/patología , Neoplasias Renales/metabolismo , Neoplasias Renales/patología , Fosfohidrolasa PTEN/metabolismo , Apoptosis , Carcinoma de Células Renales/genética , Adhesión Celular , Movimiento Celular , Progresión de la Enfermedad , Matriz Extracelular/metabolismo , Femenino , Regulación Neoplásica de la Expresión Génica , Silenciador del Gen , Humanos , Integrinas/metabolismo , Neoplasias Renales/genética , Masculino , Persona de Mediana Edad , Metástasis de la Neoplasia , Fosfohidrolasa PTEN/deficiencia , Fosfohidrolasa PTEN/genética , Isoformas de Proteínas/deficiencia , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , ARN Interferente Pequeño/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA