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










Base de datos
Intervalo de año de publicación
1.
MicroPubl Biol ; 20232023.
Artículo en Inglés | MEDLINE | ID: mdl-37908496

RESUMEN

The requirement for LRP5 and LRP6 to prevent ß-catenin degradation in the absence of the tumor suppressor APC is unclear because cell culture models have yielded conflicting results. We previously established that osteoblast-specific loss of APC causes ß-catenin accumulation and increased bone mass, while loss of both LRP5 and LRP6 reduces bone mass. We report here that the simultaneous loss of APC, LRP5, and LRP6 in osteoblasts in mice phenocopies the APC osteoblast-specific knockout. Thus, ß-catenin stabilization and increased bone mass after loss of APC in osteoblasts in vivo are not dependent on LRP5 and LRP6.

2.
Bone Res ; 11(1): 47, 2023 08 24.
Artículo en Inglés | MEDLINE | ID: mdl-37612291

RESUMEN

Proper regulation of Wnt signaling is critical for normal bone development and homeostasis. Mutations in several Wnt signaling components, which increase the activity of the pathway in the skeleton, cause high bone mass in human subjects and mouse models. Increased bone mass is often accompanied by severe headaches from increased intracranial pressure, which can lead to fatality and loss of vision or hearing due to the entrapment of cranial nerves. In addition, progressive forehead bossing and mandibular overgrowth occur in almost all subjects. Treatments that would provide symptomatic relief in these subjects are limited. Porcupine-mediated palmitoylation is necessary for Wnt secretion and binding to the frizzled receptor. Chemical inhibition of porcupine is a highly selective method of Wnt signaling inhibition. We treated three different mouse models of high bone mass caused by aberrant Wnt signaling, including homozygosity for loss-of-function in Sost, which models sclerosteosis, and two strains of mice carrying different point mutations in Lrp5 (equivalent to human G171V and A214V), at 3 months of age with porcupine inhibitors for 5-6 weeks. Treatment significantly reduced both trabecular and cortical bone mass in all three models. This demonstrates that porcupine inhibition is potentially therapeutic for symptomatic relief in subjects who suffer from these disorders and further establishes that the continued production of Wnts is necessary for sustaining high bone mass in these models.


Asunto(s)
Mutación con Ganancia de Función , Hiperostosis , Animales , Humanos , Ratones , Proteínas Adaptadoras Transductoras de Señales , Secreciones Corporales , Modelos Animales de Enfermedad , Hiperostosis/genética , Proteína-5 Relacionada con Receptor de Lipoproteína de Baja Densidad/genética , Mutación
3.
Development ; 150(3)2023 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-36789910

RESUMEN

Frizzled 2 (FZD2) is a transmembrane Wnt receptor. We previously identified a pathogenic human FZD2 variant in individuals with FZD2-associated autosomal dominant Robinow syndrome. The variant encoded a protein with a premature stop and loss of 17 amino acids, including a region of the consensus dishevelled-binding sequence. To model this variant, we used zygote microinjection and i-GONAD-based CRISPR/Cas9-mediated genome editing to generate a mouse allelic series. Embryos mosaic for humanized Fzd2W553* knock-in exhibited cleft palate and shortened limbs, consistent with patient phenotypes. We also generated two germline mouse alleles with small deletions: Fzd2D3 and Fzd2D4. Homozygotes for each allele exhibit a highly penetrant cleft palate phenotype, shortened limbs compared with wild type and perinatal lethality. Fzd2D4 craniofacial tissues indicated decreased canonical Wnt signaling. In utero treatment with IIIC3a (a DKK inhibitor) normalized the limb lengths in Fzd2D4 homozygotes. The in vivo replication represents an approach for further investigating the mechanism of FZD2 phenotypes and demonstrates the utility of CRISPR knock-in mice as a tool for investigating the pathogenicity of human genetic variants. We also present evidence for a potential therapeutic intervention.


Asunto(s)
Fisura del Paladar , Enanismo , Deformidades Congénitas de las Extremidades , Anomalías Urogenitales , Animales , Humanos , Ratones , Fisura del Paladar/genética , Enanismo/genética , Deformidades Congénitas de las Extremidades/genética , Anomalías Urogenitales/genética , Vía de Señalización Wnt/genética , Modelos Animales de Enfermedad , Receptores Frizzled/genética , Técnicas de Sustitución del Gen
4.
FEBS Open Bio ; 12(10): 1717-1728, 2022 10.
Artículo en Inglés | MEDLINE | ID: mdl-36062328

RESUMEN

We previously observed that genomic loss of galectin-3 (Gal-3; encoded by Lgals3) in mice has a significant protective effect on age-related bone loss. Gal-3 has both intracellular and extracellular functionality, and we wanted to assess whether the affect we observed in the Lgals3 knockout (KO) mice could be attributed to the ability of Gal-3 to bind glycoproteins. Mutation of a highly conserved arginine to a serine in human Gal-3 (LGALS3-R186S) blocks glycan binding and secretion. We generated mice with the equivalent mutation (Lgals3-R200S) and observed a subsequent reduction in Gal-3 secretion from mouse embryonic fibroblasts and in circulating blood. When examining bone structure in aged mice, we noticed some similarities to the Lgals3-KO mice and some differences. First, we observed greater bone mass in Lgals3-R200S mutant mice, as was previously observed in Lgals3-KO mice. Like Lgals3-KO mice, significantly increased trabecular bone mass was only observed in female Lgals3-R200S mice. These results suggest that the greater bone mass observed is driven by the loss of extracellular Gal-3 functionality. However, the results from our cortical bone expansion data showed a sex-dependent difference, with only male Lgals3-KO mice having an increased response, contrasting with our earlier study. These notable sex differences suggest a potential role for sex hormones, most likely androgen signaling, being involved. In summary, our results suggest that targeting extracellular Gal-3 function may be a suitable treatment for age-related loss of bone mass.


Asunto(s)
Hueso Esponjoso , Hueso Cortical , Galectina 3 , Andrógenos , Animales , Arginina , Hueso Esponjoso/crecimiento & desarrollo , Hueso Cortical/crecimiento & desarrollo , Femenino , Fibroblastos/metabolismo , Galectina 3/genética , Humanos , Masculino , Ratones , Ratones Noqueados , Mutación , Polisacáridos , Serina/genética
5.
Exp Eye Res ; 217: 108977, 2022 04.
Artículo en Inglés | MEDLINE | ID: mdl-35139333

RESUMEN

Loss-of-function mutations in the Wnt co-receptor, low-density lipoprotein receptor-related protein 5 (LRP5), result in familial exudative vitreoretinopathy (FEVR), osteoporosis-pseudoglioma syndrome (OPPG), and Norrie disease. CRISPR/Cas9 gene editing was used to produce rat strains deficient in Lrp5. The purpose of this study was to validate this rat model for studies of hypovascular, exudative retinopathies. The retinal vasculature of wildtype and Lrp5 knockout rats was stained with Giffonia simplifolia isolectin B4 and imaged by fluorescence microscopy. Effects on retinal structure were investigated by histology. The integrity of the blood-retina barrier was analyzed by measurement of permeability to Evans blue dye and staining for claudin-5. Retinas were imaged by fundus photography and SD-OCT, and electroretinograms were recorded. Lrp5 gene deletion led to sparse superficial retinal capillaries and loss of the deep and intermediate plexuses. Autofluorescent exudates were observed and are correlated with increased Evans blue permeability and absence of claudin-5 expression in superficial vessels. OCT images show pathology similar to OCT of humans with FEVR, and retinal thickness is reduced by 50% compared to wild-type rats. Histology and OCT reveal that photoreceptor and outer plexiform layers are absent. The retina failed to demonstrate an ERG response. CRISPR/Cas9 gene-editing produced a predictable rat Lrp5 knockout model with extensive defects in the retinal vascular and neural structure and function. This rat model should be useful for studies of exudative retinal vascular diseases involving the Wnt and norrin pathways.


Asunto(s)
Proteína-5 Relacionada con Receptor de Lipoproteína de Baja Densidad , Retina , Animales , Claudina-5/biosíntesis , Claudina-5/genética , Azul de Evans/farmacología , Vitreorretinopatías Exudativas Familiares/genética , Vitreorretinopatías Exudativas Familiares/metabolismo , Proteína-5 Relacionada con Receptor de Lipoproteína de Baja Densidad/genética , Proteína-5 Relacionada con Receptor de Lipoproteína de Baja Densidad/metabolismo , Mutación , Ratas , Retina/metabolismo , Relación Estructura-Actividad
6.
CRISPR J ; 3(4): 284-298, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32833527

RESUMEN

Humans carrying homozygous loss-of-function mutations in the Wnt co-receptor, low-density lipoprotein receptor-related protein 5 (LRP5), develop osteoporosis and a defective retinal vasculature known as familial exudative vitreoretinopathy (FEVR) due to disruption of the Wnt signaling pathway. The purpose of this study was to use CRISPR-Cas9-mediated gene editing to create strains of Lrp5-deficient rats and to determine whether knockout of Lrp5 resulted in phenotypes that model the bone and retina pathology in LRP5-deficient humans. Knockout of Lrp5 in rats produced low bone mass, decreased bone mineral density, and decreased bone size. The superficial retinal vasculature of Lrp5-deficient rats was sparse and disorganized, with extensive exudates and decreases in vascularized area, vessel length, and branch point density. This study showed that Lrp5 could be predictably knocked out in rats using CRISPR-Cas9, causing the expression of bone and retinal phenotypes that will be useful for studying the role of Wnt signaling in bone and retina development and for research on the treatment of osteoporosis and FEVR.


Asunto(s)
Huesos/metabolismo , Técnicas de Inactivación de Genes , Proteína-5 Relacionada con Receptor de Lipoproteína de Baja Densidad/genética , Vasos Retinianos/metabolismo , Animales , Huesos/fisiopatología , Femenino , Regulación de la Expresión Génica , Proteína-5 Relacionada con Receptor de Lipoproteína de Baja Densidad/metabolismo , Masculino , Mutación , Ratas , Vasos Retinianos/crecimiento & desarrollo , Vasos Retinianos/fisiopatología , Vía de Señalización Wnt
8.
PLoS Genet ; 16(5): e1008361, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-32463812

RESUMEN

Osteocalcin (OCN), the most abundant noncollagenous protein in the bone matrix, is reported to be a bone-derived endocrine hormone with wide-ranging effects on many aspects of physiology, including glucose metabolism and male fertility. Many of these observations were made using an OCN-deficient mouse allele (Osc-) in which the 2 OCN-encoding genes in mice, Bglap and Bglap2, were deleted in ES cells by homologous recombination. Here we describe mice with a new Bglap and Bglap2 double-knockout (dko) allele (Bglap/2p.Pro25fs17Ter) that was generated by CRISPR/Cas9-mediated gene editing. Mice homozygous for this new allele do not express full-length Bglap or Bglap2 mRNA and have no immunodetectable OCN in their serum. FTIR imaging of cortical bone in these homozygous knockout animals finds alterations in the collagen maturity and carbonate to phosphate ratio in the cortical bone, compared with wild-type littermates. However, µCT and 3-point bending tests do not find differences from wild-type littermates with respect to bone mass and strength. In contrast to the previously reported OCN-deficient mice with the Osc-allele, serum glucose levels and male fertility in the OCN-deficient mice with the Bglap/2pPro25fs17Ter allele did not have significant differences from wild-type littermates. We cannot explain the absence of endocrine effects in mice with this new knockout allele. Possible explanations include the effects of each mutated allele on the transcription of neighboring genes, or differences in genetic background and environment. So that our findings can be confirmed and extended by other interested investigators, we are donating this new Bglap and Bglap2 double-knockout strain to the Jackson Laboratories for academic distribution.


Asunto(s)
Sistema Endocrino/fisiología , Osteocalcina/genética , Animales , Densidad Ósea/genética , Huesos/metabolismo , Sistema Endocrino/metabolismo , Femenino , Fertilidad/genética , Resistencia a la Insulina/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Osteocalcina/deficiencia
9.
Bone Res ; 6: 17, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29844946

RESUMEN

Dysregulated Wnt signaling is associated with the pathogenesis of cancers, fibrosis, and vascular diseases. Inhibition of Wnt signaling has shown efficacy in various pre-clinical models of these disorders. One of the key challenges in developing targeted anti-cancer drugs is to balance efficacy with on-target toxicity. Given the crucial role Wnts play in the differentiation of osteoblasts and osteoclasts, acute inhibition of Wnt signaling is likely to affect bone homeostasis. In this study, we evaluated the skeletal effect of small molecule inhibitor of an o-acyl transferase porcupine (PORCN) that prevents Wnt signaling by blocking the secretion of all Wnts. Micro-computed tomography and histomorphometric evaluation revealed that the bones of mice treated with two structurally distinct PORCN inhibitors LGK974 and ETC-1922159 (ETC-159) had loss-of-bone volume and density within 4 weeks of exposure. This decreased bone mass was associated with a significant increase in adipocytes within the bone marrow. Notably, simultaneous administration of a clinically approved anti-resorptive, alendronate, a member of the bisphosphonate family, mitigated loss-of-bone mass seen upon ETC-159 treatment by regulating activity of osteoclasts and blocking accumulation of bone marrow adipocytes. Our results support the addition of bone protective agents when treating patients with PORCN inhibitors. Mitigation of bone toxicity can extend the therapeutic utility of Wnt pathway inhibitors.

10.
Bone ; 98: 68-78, 2017 05.
Artículo en Inglés | MEDLINE | ID: mdl-28384511

RESUMEN

Inactivating mutations that lead to loss of heterozygosity within the HRPT2/Cdc73 gene are directly linked to the development of primary hyperparathyroidism, parathyroid adenomas, and ossifying fibromas of the jaw (HPT-JT). The protein product of the Cdc73 gene, parafibromin, is a core member of the polymerase-associated factors (PAF) complex, which coordinates epigenetic modifiers and transcriptional machinery to control gene expression. We conditionally deleted Cdc73 within mesenchymal progenitors or within mature osteoblasts and osteocytes to determine the consequences of parafibromin loss within the mesenchymal lineage. Homozygous deletion of Cdc73 via the Dermo1-Cre driver resulted in embryos which lacked mesenchymal organ development of internal organs, including the heart and fetal liver. Immunohistochemical detection of cleaved caspase-3 revealed extensive apoptosis within the progenitor pools of developing organs. Unexpectedly, when Cdc73 was homozygously deleted within mature osteoblasts and osteocytes (via the Ocn-Cre driver), the mice had a normal life span but increased cortical and trabecular bone. OCN-Cre;Cdc73flox/flox bones displayed large cortical pores actively undergoing bone remodeling. Additionally the cortical bone of OCN-Cre;Cdc73flox/flox femurs contained osteocytes with marked amounts of cytoplasmic RNA and a high rate of apoptosis. Transcriptional analysis via RNA-seq within OCN-Cre;Cdc73flox/flox osteoblasts showed that loss of Cdc73 led to a derepression of osteoblast-specific genes, specifically those for collagen and other bone matrix proteins. These results aid in our understanding of the role parafibromin plays within transcriptional regulation, terminal differentiation, and bone homeostasis.


Asunto(s)
Remodelación Ósea/fisiología , Huesos/metabolismo , Osteoblastos/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Absorciometría de Fotón , Animales , Diferenciación Celular/fisiología , Citometría de Flujo , Inmunohistoquímica , Etiquetado Corte-Fin in Situ , Ratones , Ratones Noqueados , Ratones Mutantes , Osteogénesis , Transcriptoma , Microtomografía por Rayos X
11.
Dev Dyn ; 244(10): 1347-55, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26249818

RESUMEN

BACKGROUND: Canonical and noncanonical Wnt signaling pathways both play pivotal roles in bone development. Wntless/GPR177 is a chaperone protein that is required for secretion of all Wnt ligands. We previously showed that deletion of Wntless within mature osteoblasts severely impaired postnatal bone homeostasis. RESULTS: In this study, we systemically evaluated how deletion of Wntless in different stages of osteochondral differentiation affected embryonic bone development, by crossing Wntless (Wls)-flox/flox mice with strains expressing cre recombinase behind the following promoters: Osteocalcin, Collagen 2a1, or Dermo1. Ex vivo µCT and whole-mount skeletal staining were performed to examine skeletal mineralization. Histology and immunohistochemistry were used to evaluate cellular differentiation and alterations in Wnt signaling. In this work, we found that Wntless regulated chondrogenesis and osteogenesis through both canonical and noncanonical Wnt signaling. CONCLUSIONS: These findings provide more insight into the requirements of different Wnt-secretion cell types critical for skeletal development.


Asunto(s)
Huesos/embriología , Condrogénesis , Péptidos y Proteínas de Señalización Intracelular/fisiología , Osteogénesis , Receptores Acoplados a Proteínas G/fisiología , Proteínas Wnt/metabolismo , Vía de Señalización Wnt , Animales , Huesos/metabolismo , Calcificación Fisiológica , Condrocitos/fisiología , Humanos , Ligandos , Ratones Transgénicos , Osteoblastos/fisiología , Regiones Promotoras Genéticas , beta Catenina/metabolismo
12.
Hepatology ; 60(3): 964-76, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-24700412

RESUMEN

UNLABELLED: Liver-specific ß-catenin knockout (ß-Catenin-LKO) mice have revealed an essential role of ß-catenin in metabolic zonation where it regulates pericentral gene expression and in initiating liver regeneration (LR) after partial hepatectomy (PH), by regulating expression of Cyclin-D1. However, what regulates ß-catenin activity in these events remains an enigma. Here we investigate to what extent ß-catenin activation is Wnt-signaling-dependent and the potential cell source of Wnts. We studied liver-specific Lrp5/6 KO (Lrp-LKO) mice where Wnt-signaling was abolished in hepatocytes while the ß-catenin gene remained intact. Intriguingly, like ß-catenin-LKO mice, Lrp-LKO exhibited a defect in metabolic zonation observed as a lack of glutamine synthetase (GS), Cyp1a2, and Cyp2e1. Lrp-LKO also displayed a significant delay in initiation of LR due to the absence of ß-catenin-TCF4 association and lack of Cyclin-D1. To address the source of Wnt proteins in liver, we investigated conditional Wntless (Wls) KO mice, which lacked the ability to secrete Wnts from either liver epithelial cells (Wls-LKO), or macrophages including Kupffer cells (Wls-MKO), or endothelial cells (Wls-EKO). While Wls-EKO was embryonic lethal precluding further analysis in adult hepatic homeostasis and growth, Wls-LKO and Wls-MKO were viable but did not show any defect in hepatic zonation. Wls-LKO showed normal initiation of LR; however, Wls-MKO showed a significant but temporal deficit in LR that was associated with decreased ß-catenin-TCF4 association and diminished Cyclin-D1 expression. CONCLUSION: Wnt-signaling is the major upstream effector of ß-catenin activity in pericentral hepatocytes and during LR. Hepatocytes, cholangiocytes, or macrophages are not the source of Wnts in regulating hepatic zonation. However, Kupffer cells are a major contributing source of Wnt secretion necessary for ß-catenin activation during LR.


Asunto(s)
Regeneración Hepática/fisiología , Transducción de Señal/fisiología , Proteínas Wnt/fisiología , beta Catenina/fisiología , Uniones Adherentes/fisiología , Animales , Femenino , Regulación de la Expresión Génica , Hepatectomía , Macrófagos del Hígado/química , Macrófagos del Hígado/metabolismo , Macrófagos del Hígado/fisiología , Hígado/citología , Hígado/metabolismo , Hígado/fisiología , Masculino , Ratones , Ratones Noqueados , Transducción de Señal/genética , beta Catenina/deficiencia , beta Catenina/genética
13.
PLoS One ; 8(5): e63323, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23675479

RESUMEN

The canonical Wnt signaling pathway is critical for skeletal development and maintenance, but the precise roles of the individual Wnt co-receptors, Lrp5 and Lrp6, that enable Wnt signals to be transmitted in osteoblasts remain controversial. In these studies, we used Cre-loxP recombination, in which Cre-expression is driven by the human osteocalcin promoter, to determine the individual contributions of Lrp5 and Lrp6 in postnatal bone acquisition and osteoblast function. Mice selectively lacking either Lrp5 or Lrp6 in mature osteoblasts were born at the expected Mendelian frequency but demonstrated significant reductions in whole-body bone mineral density. Bone architecture measured by microCT revealed that Lrp6 mutant mice failed to accumulate normal amounts of trabecular bone. By contrast, Lrp5 mutants had normal trabecular bone volume at 8 weeks of age, but with age, these mice also exhibited trabecular bone loss. Both mutants also exhibited significant alterations in cortical bone structure. In vitro differentiation was impaired in both Lrp5 and Lrp6 null osteoblasts as indexed by alkaline phosphatase and Alizarin red staining, but the defect was more pronounced in Lrp6 mutant cells. Mice lacking both Wnt co-receptors developed severe osteopenia similar to that observed previously in mice lacking ß-catenin in osteoblasts. Likewise, calvarial cells doubly deficient for Lrp5 and Lrp6 failed to form osteoblasts when cultured in osteogenic media, but instead attained a chondrocyte-like phenotype. These results indicate that expression of both Lrp5 and Lrp6 are required within mature osteoblasts for normal postnatal bone development.


Asunto(s)
Huesos/metabolismo , Proteína-5 Relacionada con Receptor de Lipoproteína de Baja Densidad/metabolismo , Proteína-6 Relacionada a Receptor de Lipoproteína de Baja Densidad/metabolismo , Osteoblastos/metabolismo , Animales , Enfermedades Óseas Metabólicas/genética , Enfermedades Óseas Metabólicas/patología , Huesos/diagnóstico por imagen , Huesos/patología , Diferenciación Celular/genética , Proteína-5 Relacionada con Receptor de Lipoproteína de Baja Densidad/genética , Proteína-6 Relacionada a Receptor de Lipoproteína de Baja Densidad/genética , Masculino , Ratones , Ratones Transgénicos , Mutación , Osteoblastos/citología , Fenotipo , Radiografía
14.
Proc Natl Acad Sci U S A ; 109(33): E2197-204, 2012 Aug 14.
Artículo en Inglés | MEDLINE | ID: mdl-22745162

RESUMEN

Recent genome-wide association studies of individuals of Asian and European descent have found that SNPs located within the genomic region (1p31.3) encoding the Wntless (Wls)/Gpr177 protein are associated significantly with reduced bone mineral density. Wls/Gpr177 is a newly identified chaperone protein that specifically escorts Wnt ligands for secretion. Given the strong functional association between the Wnt signaling pathways and bone development and homeostasis, we generated osteoblast-specific Wls-deficient (Ocn-Cre;Wls-flox) mice. Homozygous conditional knockout animals were born at a normal Mendelian frequency. Whole-body dual-energy X-ray absorptiometry scanning revealed that bone-mass accrual was significantly inhibited in homozygotes as early as 20 d of age. These homozygotes had spontaneous fractures and a high frequency of premature lethality at around 2 mo of age. Microcomputed tomography analysis and histomorphometric data revealed a dramatic reduction of both trabecular and cortical bone mass in homozygous mutants. Bone formation in homozygotes was severely impaired, but no obvious phenotypic change was observed in mice heterozygous for the conditional deletion. In vitro studies showed that Wls-deficient osteoblasts had a defect in differentiation and mineralization, with significant reductions in the expression of key osteoblast differentiation regulators. In summary, these results reveal a surprising and crucial role of osteoblast-secreted Wnt ligands in bone-mass accrual.


Asunto(s)
Densidad Ósea , Huesos/metabolismo , Diferenciación Celular , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Osteoblastos/metabolismo , Animales , Matriz Ósea/metabolismo , Resorción Ósea/diagnóstico por imagen , Resorción Ósea/patología , Huesos/diagnóstico por imagen , Huesos/patología , Silenciador del Gen , Heterocigoto , Péptidos y Proteínas de Señalización Intracelular/deficiencia , Péptidos y Proteínas de Señalización Intracelular/genética , Masculino , Ratones , Tamaño de los Órganos , Especificidad de Órganos , Osteoblastos/patología , Receptores Acoplados a Proteínas G , Vía de Señalización Wnt , Microtomografía por Rayos X
15.
Mol Cancer Res ; 10(7): 958-68, 2012 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-22571869

RESUMEN

Malignant glioblastomas are characterized by their ability to infiltrate into normal brain. We previously reported that binding of the multifunctional cytokine TNF-like weak inducer of apoptosis (TWEAK) to its receptor fibroblast growth factor-inducible 14 (Fn14) induces glioblastoma cell invasion via Rac1 activation. Here, we show that Cdc42 plays an essential role in Fn14-mediated activation of Rac1. TWEAK-treated glioma cells display an increased activation of Cdc42, and depletion of Cdc42 using siRNA abolishes TWEAK-induced Rac1 activation and abrogates glioma cell migration and invasion. In contrast, Rac1 depletion does not affect Cdc42 activation by Fn14, showing that Cdc42 mediates TWEAK-stimulated Rac1 activation. Furthermore, we identified two guanine nucleotide exchange factors (GEF), Ect2 and Trio, involved in TWEAK-induced activation of Cdc42 and Rac1, respectively. Depletion of Ect2 abrogates both TWEAK-induced Cdc42 and Rac1 activation, as well as subsequent TWEAK-Fn14-directed glioma cell migration and invasion. In contrast, Trio depletion inhibits TWEAK-induced Rac1 activation but not TWEAK-induced Cdc42 activation. Finally, inappropriate expression of Fn14 or Ect2 in mouse astrocytes in vivo using an RCAS vector system for glial-specific gene transfer in G-tva transgenic mice induces astrocyte migration within the brain, corroborating the in vitro importance of the TWEAK-Fn14 signaling cascade in glioblastoma invasion. Our results suggest that the TWEAK-Fn14 signaling axis stimulates glioma cell migration and invasion through two GEF-GTPase signaling units, Ect2-Cdc42 and Trio-Rac1. Components of the Fn14-Rho GEF-Rho GTPase signaling pathway present innovative drug targets for glioma therapy.


Asunto(s)
Glioblastoma , Proteínas Proto-Oncogénicas , Receptores del Factor de Necrosis Tumoral , Proteína de Unión al GTP cdc42 , Animales , Astrocitoma/genética , Astrocitoma/metabolismo , Movimiento Celular/genética , Citocina TWEAK , Glioblastoma/genética , Glioblastoma/metabolismo , Factores de Intercambio de Guanina Nucleótido/metabolismo , Humanos , Ratones , Invasividad Neoplásica/genética , Neuropéptidos/metabolismo , Fosfoproteínas/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas/metabolismo , Receptores del Factor de Necrosis Tumoral/genética , Receptores del Factor de Necrosis Tumoral/metabolismo , Transducción de Señal , Receptor de TWEAK , Factores de Necrosis Tumoral/farmacología , Proteína de Unión al GTP cdc42/genética , Proteína de Unión al GTP cdc42/metabolismo , Proteínas de Unión al GTP rac/metabolismo , Proteína de Unión al GTP rac1
16.
PLoS One ; 7(3): e33676, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22438981

RESUMEN

The glucagon receptor (GCGR) is a member of the class B G protein-coupled receptor family. Activation of GCGR by glucagon leads to increased glucose production by the liver. Thus, glucagon is a key component of glucose homeostasis by counteracting the effect of insulin. In this report, we found that in addition to activation of the classic cAMP/protein kinase A (PKA) pathway, activation of GCGR also induced ß-catenin stabilization and activated ß-catenin-mediated transcription. Activation of ß-catenin signaling was PKA-dependent, consistent with previous reports on the parathyroid hormone receptor type 1 (PTH1R) and glucagon-like peptide 1 (GLP-1R) receptors. Since low-density-lipoprotein receptor-related protein 5 (Lrp5) is an essential co-receptor required for Wnt protein mediated ß-catenin signaling, we examined the role of Lrp5 in glucagon-induced ß-catenin signaling. Cotransfection with Lrp5 enhanced the glucagon-induced ß-catenin stabilization and TCF promoter-mediated transcription. Inhibiting Lrp5/6 function using Dickkopf-1(DKK1) or by expression of the Lrp5 extracellular domain blocked glucagon-induced ß-catenin signaling. Furthermore, we showed that Lrp5 physically interacted with GCGR by immunoprecipitation and bioluminescence resonance energy transfer assays. Together, these results reveal an unexpected crosstalk between glucagon and ß-catenin signaling, and may help to explain the metabolic phenotypes of Lrp5/6 mutations.


Asunto(s)
Receptores de Glucagón/metabolismo , beta Catenina/metabolismo , Animales , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Glucagón/agonistas , Glucagón/farmacología , Péptido 1 Similar al Glucagón/farmacología , Receptor del Péptido 1 Similar al Glucagón , Células HEK293 , Hepatocitos/efectos de los fármacos , Hepatocitos/metabolismo , Humanos , Proteína-5 Relacionada con Receptor de Lipoproteína de Baja Densidad/genética , Proteína-5 Relacionada con Receptor de Lipoproteína de Baja Densidad/metabolismo , Proteína-6 Relacionada a Receptor de Lipoproteína de Baja Densidad/genética , Proteína-6 Relacionada a Receptor de Lipoproteína de Baja Densidad/metabolismo , Ratones , Receptores de Glucagón/genética , Transducción de Señal/efectos de los fármacos , Transfección
17.
J Cell Biochem ; 113(1): 31-8, 2012 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-21866564

RESUMEN

Low-density lipoprotein receptor-related proteins 5 and 6 (Lrp5 and Lrp6) are co-receptors of Wnt ligands and play important roles in Wnt/ß-catenin signal transduction. Mice homozygous for a germline deletion of Lrp6 die at birth with several associated defects, while Lrp5-deficient mice are viable. Here, we conditionally deleted Lrp5 and/or Lrp6 in the mouse gut ((gut-/-)) by crossing mice carrying floxed alleles of Lrp5 and Lrp6 to a strain expressing Cre recombinase from the villin promoter (villin-Cre). The changes in morphology, differentiation, and Wnt signal transduction were validated using immunohistochemistry and other staining. Consistent with observations in mice carrying a homozygous germline deletion in Lrp5, intestinal development in Lrp5(gut-/-) mice was normal. In addition, mice homozygous for villin-Cre-induced deletion of Lrp6 (Lrp6(gut-/-)) were viable with apparently normal intestinal differentiation and function. However, mice homozygous for villin-Cre inactivated alleles of both genes (Lrp5(gut-/-) ; Lrp6(gut-/-)) died within 1 day of birth. Analysis of embryonic Lrp5(gut-/-); Lrp6(gut-/-) intestinal epithelium showed a progressive loss of cells, an absence of proliferation, and a premature differentiation of crypt stem/precursor cells; no notable change in differentiation was observed in the embryos lacking either gene alone. Further immunohistochemical studies showed that expression of the Wnt/ß-catenin target, cyclin D1, was specifically reduced in the intestinal epithelium of Lrp5(gut-/-); Lrp6(gut-/-) embryos. Our data demonstrate that Lrp5 and Lrp6 play redundant roles in intestinal epithelium development, and that Lrp5/6 might regulate intestinal stem/precursor cell maintenance by regulating Wnt/ß-catenin signaling.


Asunto(s)
Intestinos/embriología , Proteína-5 Relacionada con Receptor de Lipoproteína de Baja Densidad/metabolismo , Proteína-6 Relacionada a Receptor de Lipoproteína de Baja Densidad/metabolismo , Vía de Señalización Wnt/genética , Animales , Diferenciación Celular/genética , Proliferación Celular , Ciclina D1/metabolismo , Regulación del Desarrollo de la Expresión Génica , Proteína-5 Relacionada con Receptor de Lipoproteína de Baja Densidad/genética , Proteína-6 Relacionada a Receptor de Lipoproteína de Baja Densidad/genética , Ratones , Eliminación de Secuencia/genética
18.
Dev Biol ; 359(2): 222-9, 2011 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-21924256

RESUMEN

The role of Wnt signaling in osteoblastogenesis in the embryo remains to be fully established. Although ß-catenin, a multifunctional protein also mediating canonical Wnt signaling, is indispensable for embryonic osteoblast differentiation, the roles of the key Wnt co-receptors Lrp5 and Lrp6 are unclear. Indeed, global deletion of either Lrp5 or Lrp6 did not overtly affect osteoblast differentiation in the mouse embryo. Here, we generated mice lacking both receptors specifically in the embryonic mesenchyme and observed an absence of osteoblasts in the embryo. In addition, the double-deficient embryos developed supernumerary cartilage elements in the zeugopod, revealing an important role for mesenchymal Lrp5/6 signaling in limb patterning. Importantly, the phenotypes of the Lrp5/6 mutant closely resembled those of the ß-catenin-deficient embryos. These phenotypes are likely independent of any effect on the adherens junction, as deletion of α-catenin, another component of the complex, did not cause similar defects. Thus, Lrp5 and 6 redundantly control embryonic skeletal development, likely through ß-catenin signaling.


Asunto(s)
Huesos/metabolismo , Embrión de Mamíferos/metabolismo , Proteína-5 Relacionada con Receptor de Lipoproteína de Baja Densidad/genética , Proteína-6 Relacionada a Receptor de Lipoproteína de Baja Densidad/genética , Animales , Tipificación del Cuerpo/genética , Desarrollo Óseo/genética , Huesos/embriología , Cartílago/embriología , Cartílago/metabolismo , Embrión de Mamíferos/embriología , Extremidades/embriología , Femenino , Regulación del Desarrollo de la Expresión Génica , Hibridación in Situ , Masculino , Mesodermo/embriología , Mesodermo/metabolismo , Ratones , Ratones Noqueados , Osteogénesis/genética , Transducción de Señal/genética , Factores de Tiempo , beta Catenina/genética
19.
Cancers (Basel) ; 3(2): 2050-79, 2011 Apr 19.
Artículo en Inglés | MEDLINE | ID: mdl-24212796

RESUMEN

The ability of Wnt ligands to initiate a signaling cascade that results in cytoplasmic stabilization of, and nuclear localization of, ß-catenin underlies their ability to regulate progenitor cell differentiation. In this review, we will summarize the current knowledge of the mechanisms underlying Wnt/ß-catenin signaling and how the pathway regulates normal differentiation of stem cells in the intestine, mammary gland, and prostate. We will also discuss how dysregulation of the pathway is associated with putative cancer stem cells and the potential therapeutic implications of regulating Wnt signaling.

20.
Cancer Res ; 70(22): 9143-52, 2010 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-21062985

RESUMEN

Cross-talk between the canonical Wnt and mammalian target of rapamycin (mTOR) signaling pathways occurs at multiple levels in the cell and likely contributes to the oncogenic effects of these pathways in human cancer. To gain more insight into the interplay between Wnt and mTOR signaling in salivary gland tumorigenesis, we developed a mouse model in which both pathways are constitutively activated by the conditional inactivation of the Apc and Pten tumor suppressor genes. Loss of either Apc or Pten alone did not cause tumor development. However, deletion of both genes resulted in the formation of salivary gland tumors with 100% penetrance and short latency that showed a remarkable morphologic similarity to human acinic cell carcinoma. Treatment of tumor-bearing mice using the mTOR inhibitor rapamycin led to complete regression of tumors, indicating that tumor growth was dependent on continued mTOR signaling. Importantly, we found that human salivary gland acinic cell carcinomas also express markers of activated mTOR signaling. Together, these results suggest that aberrant activation of mTOR signaling plays a pivotal role in acinar cell neoplasia of the salivary gland. Because rapamycin analogues are approved for treating other types of human malignancies, our findings suggest that rapamycin therapy should be evaluated for treating patients with salivary gland acinic cell carcinoma.


Asunto(s)
Proteína de la Poliposis Adenomatosa del Colon/deficiencia , Carcinoma de Células Acinares/metabolismo , Fosfohidrolasa PTEN/deficiencia , Glándulas Salivales/metabolismo , Serina-Treonina Quinasas TOR/metabolismo , Proteína de la Poliposis Adenomatosa del Colon/genética , Animales , Antibióticos Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Carcinoma de Células Acinares/genética , Carcinoma de Células Acinares/patología , Femenino , Citometría de Flujo , Humanos , Inmunohistoquímica , Masculino , Ratones , Ratones de la Cepa 129 , Ratones Noqueados , Fosfohidrolasa PTEN/genética , Neoplasias de las Glándulas Salivales/genética , Neoplasias de las Glándulas Salivales/metabolismo , Neoplasias de las Glándulas Salivales/patología , Glándulas Salivales/efectos de los fármacos , Glándulas Salivales/patología , Transducción de Señal/efectos de los fármacos , Sirolimus/farmacología , Serina-Treonina Quinasas TOR/antagonistas & inhibidores , Carga Tumoral/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...