Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 53
Filtrar
1.
PLoS One ; 15(4): e0226661, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32240171

RESUMEN

CD47 is an immune checkpoint protein that downregulates both the innate and adaptive anti-tumor immune response via its counter receptor SIRPα. Biologics, including humanized CD47 monoclonal antibodies and decoy SIRPα receptors, that block the SIRPα-CD47 interaction, are currently being developed as cancer immunotherapy agents. However, adverse side effects and limited penetration of tumor tissue associated with their structure and large size may impede their clinical application. We recently developed a quantitative high throughput screening assay platform to identify small molecules that disrupt the binding of SIRPα and CD47 as an alternative approach to these protein-based therapeutics. Here, we report on the development and optimization of a cell-based binding assay to validate active small molecules from our biochemical screening effort. This assay has a low volume, high capacity homogenous format that relies on laser scanning cytometry (LSC) and associated techniques to enhance signal to noise measurement of cell surface binding. The LSC assay is specific, concentration dependent, and validated for the two major human SIRPα variants (V1 and V2), with results that parallel those of our biochemical data as well as published studies. We also utilized the LSC assay to confirm published studies showing that the inhibition of amino-terminal pyroglutamate formation on CD47 using the glutaminyl cyclase inhibitor SEN177 disrupts SIRPα binding. The SIRPα-CD47 interaction could be quantitatively measured in live and fixed tumor cells. Use of fixed cells reduces the burden of cell maintenance and provides stable cell standards to control for inter- and intra-assay variations. We also demonstrate the utility of the assay to characterize the activity of the first reported small molecule antagonists of the SIRPα-CD47 interaction. This assay will support the screening of thousands of compounds to identify or validate active small molecules as hits, develop structure activity relationships and assist in the optimization of hits to leads by a typical iterative medicinal chemistry campaign.


Asunto(s)
Inmunidad Adaptativa/efectos de los fármacos , Antígenos de Diferenciación/genética , Antígeno CD47/genética , Neoplasias/tratamiento farmacológico , Receptores Inmunológicos/genética , Bibliotecas de Moléculas Pequeñas/farmacología , Inmunidad Adaptativa/genética , Aminoaciltransferasas/antagonistas & inhibidores , Aminoaciltransferasas/química , Antígenos de Diferenciación/química , Antígeno CD47/química , Desarrollo de Medicamentos , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Ensayos Analíticos de Alto Rendimiento/métodos , Humanos , Inmunoterapia/métodos , Células Jurkat , Citometría de Barrido por Láser , Ligandos , Oncología Médica/tendencias , Neoplasias/genética , Neoplasias/inmunología , Neoplasias/patología , Fagocitosis/efectos de los fármacos , Mapas de Interacción de Proteínas/genética , Receptores Inmunológicos/química , Bibliotecas de Moléculas Pequeñas/química
2.
PLoS One ; 14(7): e0218897, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31276567

RESUMEN

CD47 is an immune checkpoint molecule that downregulates key aspects of both the innate and adaptive anti-tumor immune response via its counter receptor SIRPα, and it is expressed at high levels in a wide variety of tumor types. This has led to the development of biologics that inhibit SIRPα engagement including humanized CD47 antibodies and a soluble SIRPα decoy receptor that are currently undergoing clinical trials. Unfortunately, toxicological issues, including anemia related to on-target mechanisms, are barriers to their clinical advancement. Another potential issue with large biologics that bind CD47 is perturbation of CD47 signaling through its high-affinity interaction with the matricellular protein thrombospondin-1 (TSP1). One approach to avoid these shortcomings is to identify and develop small molecule molecular probes and pretherapeutic agents that would (1) selectively target SIRPα or TSP1 interactions with CD47, (2) provide a route to optimize pharmacokinetics, reduce on-target toxicity and maximize tissue penetration, and (3) allow more flexible routes of administration. As the first step toward this goal, we report the development of an automated quantitative high-throughput screening (qHTS) assay platform capable of screening large diverse drug-like chemical libraries to discover novel small molecules that inhibit CD47-SIRPα interaction. Using time-resolved Förster resonance energy transfer (TR-FRET) and bead-based luminescent oxygen channeling assay formats (AlphaScreen), we developed biochemical assays, optimized their performance, and individually tested them in small-molecule library screening. Based on performance and low false positive rate, the LANCE TR-FRET assay was employed in a ~90,000 compound library qHTS, while the AlphaScreen oxygen channeling assay served as a cross-validation orthogonal assay for follow-up characterization. With this multi-assay strategy, we successfully eliminated compounds that interfered with the assays and identified five compounds that inhibit the CD47-SIRPα interaction; these compounds will be further characterized and later disclosed. Importantly, our results validate the large library qHTS for antagonists of CD47-SIRPα interaction and suggest broad applicability of this approach to screen chemical libraries for other protein-protein interaction modulators.


Asunto(s)
Anticuerpos Monoclonales Humanizados/farmacología , Antígenos de Diferenciación/metabolismo , Antígeno CD47/metabolismo , Descubrimiento de Drogas/métodos , Ensayos Analíticos de Alto Rendimiento/métodos , Receptores Inmunológicos/metabolismo , Bibliotecas de Moléculas Pequeñas/farmacología , Animales , Antígenos de Diferenciación/química , Biotina/química , Biotina/metabolismo , Antígeno CD47/química , Antígeno CD47/inmunología , Humanos , Modelos Moleculares , Unión Proteica/efectos de los fármacos , Dominios Proteicos , Receptores Inmunológicos/química , Reproducibilidad de los Resultados , Transducción de Señal/efectos de los fármacos
3.
Mol Biol Cell ; 29(22): 2784-2799, 2018 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-30188763

RESUMEN

Mice that lack the epidermal growth factor receptor (EGFR) fail to develop a hair coat, but the mechanism responsible for this deficit is not completely understood. Here, we show that EGFR plays a critical role to attenuate wingless-type MMTV integration site family member (Wnt)/ß-catenin signaling during postnatal hair follicle development. Genetic ablation of EGFR in mice resulted in increased mitotic activity in matrix cells, apoptosis in hair follicles, and impaired differentiation of epithelial lineages that form hair. EGFR is activated in wild-type hair follicle stem cells marked with SOX9 or NFATc1 and is essential to restrain proliferation and support stem cell numbers and their quiescence. We observed elevated levels of Wnt4, 6, 7b, 10a, 10b, and 16 transcripts and hyperactivation of the ß-catenin pathway in EGFR knockout follicles. Using primary keratinocytes, we linked ligand-induced EGFR activation to suppression of nascent mRNA synthesis of Wnt genes. Overexpression of the Wnt antagonist sFRP1 in mice lacking EGFR demonstrated that elevated Wnts are a major cause for the hair follicle defects. Colocalization of transforming growth factor α and Wnts regulated by EGFR in stem cells and progeny indicates that EGFR autocrine loops control Wnts. Our findings define a novel mechanism that integrates EGFR and Wnt/ß-catenin pathways to coordinate the delicate balance between proliferation and differentiation during development.


Asunto(s)
Receptores ErbB/metabolismo , Folículo Piloso/crecimiento & desarrollo , Folículo Piloso/metabolismo , Vía de Señalización Wnt , Animales , Apoptosis/efectos de los fármacos , Recuento de Células , Diferenciación Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Daño del ADN , Eliminación de Gen , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Técnicas de Sustitución del Gen , Cabello/crecimiento & desarrollo , Folículo Piloso/citología , Ligandos , Ratones , Mitosis/efectos de los fármacos , Modelos Biológicos , Morfogénesis/efectos de los fármacos , Fenotipo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Piel/crecimiento & desarrollo , Transcripción Genética/efectos de los fármacos , Factor de Crecimiento Transformador alfa/farmacología , Vía de Señalización Wnt/efectos de los fármacos , Proteínas ras/metabolismo
4.
PLoS One ; 12(1): e0170903, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28125685

RESUMEN

Casein kinase 1 delta (CK1δ) is a conserved serine/threonine protein kinase that regulates diverse cellular processes. Mice lacking CK1δ have a perinatal lethal phenotype and typically weigh 30% less than their wild type littermates. However, the causes of death and small size are unknown. We observed cells with abnormally large nuclei in tissue from Csnk1d null embryos, and multiple centrosomes in mouse embryo fibroblasts (MEFs) deficient in CK1δ (MEFCsnk1d null). Results from γ-H2AX staining and the comet assay demonstrated significant DNA damage in MEFCsnk1d null cells. These cells often contain micronuclei, an indicator of genomic instability. Similarly, abrogation of CK1δ expression in control MEFs stimulated micronuclei formation after doxorubicin treatment, suggesting that CK1δ loss increases vulnerability to genotoxic stress. Cellular levels of total and activated checkpoint kinase 1 (Chk1), which functions in the DNA damage response and mitotic checkpoints, and its downstream effector, Cdc2/CDK1 kinase, were often decreased in MEFCsnk1d null cells as well as in control MEFs transfected with CK1δ siRNA. Hydroxyurea-induced Chk1 activation, as measured by Ser345 phosphorylation, and nuclear localization also were impaired in MEF cells following siRNA knockdown of CK1δ. Similar results were observed in the MCF7 human breast cancer cell line. The decreases in phosphorylated Chk1 were rescued by concomitant expression of siRNA-resistant CK1δ. Experiments with cycloheximide demonstrated that the stability of Chk1 protein was diminished in cells subjected to CK1δ knockdown. Together, these findings suggest that CK1δ contributes to the efficient repair of DNA damage and the proper functioning of mitotic checkpoints by maintaining appropriate levels of Chk1.


Asunto(s)
Quinasa de la Caseína I/genética , Quinasa 1 Reguladora del Ciclo Celular (Checkpoint 1)/genética , Daño del ADN , Regulación hacia Abajo , Inestabilidad Genómica , Animales , Quinasa de la Caseína I/metabolismo , Línea Celular Tumoral , Quinasa 1 Reguladora del Ciclo Celular (Checkpoint 1)/metabolismo , Fibroblastos/metabolismo , Humanos , Ratones , Ratones Noqueados , Mitosis/genética , Fosforilación
5.
Case Rep Ophthalmol ; 7(1): 163-6, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27099607

RESUMEN

Diabetic retinopathy worsens the prognosis of macular holes compared to those of idiopathic etiology. While spontaneous closure of idiopathic macular holes is a well-documented phenomenon, spontaneous closure of macular holes associated with proliferative diabetic retinopathy is rare. We report a case of spontaneous closure of a macular hole associated with proliferative diabetic retinopathy and persistent vitreomacular traction.

6.
Retin Cases Brief Rep ; 10(3): 205-7, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26510001

RESUMEN

PURPOSE: To describe a rare case of recurring central serous chorioretinopathy associated with retinitis pigmentosa successfully treated with oral acetazolamide. METHODS: A 17-year-old male with retinitis pigmentosa who developed four separate episodes of central serous chorioretinopathy over a 12-month period. RESULTS: After the patient's fourth recurrence, he was treated with daily oral acetazolamide, which resulted in resolution of submacular fluid. He has had no subsequent recurrences while being maintained on alternating and then biweekly doses of oral acetazolamide. CONCLUSION: Recurrent central serous chorioretinopathy associated with retinitis pigmentosa is a rare occurrence. The presented case demonstrates that oral acetazolamide successfully treated and may have delayed recurrent episodes of central serous chorioretinopathy in the patient with retinitis pigmentosa.


Asunto(s)
Acetazolamida/uso terapéutico , Inhibidores de Anhidrasa Carbónica/uso terapéutico , Coriorretinopatía Serosa Central/tratamiento farmacológico , Retinitis Pigmentosa/complicaciones , Administración Oral , Adolescente , Coriorretinopatía Serosa Central/complicaciones , Humanos , Masculino , Resultado del Tratamiento
7.
Cell Rep ; 10(2): 123-30, 2015 Jan 13.
Artículo en Inglés | MEDLINE | ID: mdl-25558062

RESUMEN

G protein-coupled receptor 124 (GPR124) is an orphan receptor in the adhesion family of GPCRs, and previous global or endothelial-specific disruption of Gpr124 in mice led to defective CNS angiogenesis and blood-brain barriergenesis. Similar developmental defects were observed following dual deletion of Wnt7a/Wnt7b or deletion of ß-catenin in endothelial cells, suggesting a possible relationship between GPR124 and canonical WNT signaling. Here, we show using in vitro reporter assays, mutation analysis, and genetic interaction studies in vivo that GPR124 functions as a WNT7A/WNT7B-specific costimulator of ß-catenin signaling in brain endothelium. WNT7-stimulated ß-catenin signaling was dependent upon GPR124's intracellular PDZ binding motif and a set of leucine-rich repeats in its extracellular domain. This study reveals a vital role for GPR124 in potentiation of WNT7-induced canonical ß-catenin signaling with important implications for understanding and manipulating CNS-specific angiogenesis and blood-brain barrier-genesis.


Asunto(s)
Receptores Acoplados a Proteínas G/metabolismo , Proteínas Wnt/metabolismo , Vía de Señalización Wnt , beta Catenina/metabolismo , Secuencias de Aminoácidos , Animales , Barrera Hematoencefálica/metabolismo , Encéfalo/citología , Encéfalo/metabolismo , Células Endoteliales/citología , Células Endoteliales/metabolismo , Células HEK293 , Humanos , Ratones , Ratones Noqueados , Ratones Transgénicos , Dominios PDZ , Proteínas Proto-Oncogénicas/metabolismo , Receptores Acoplados a Proteínas G/química , Receptores Acoplados a Proteínas G/deficiencia
8.
J Biol Chem ; 290(11): 6789-98, 2015 Mar 13.
Artículo en Inglés | MEDLINE | ID: mdl-25605717

RESUMEN

The seven-transmembrane-spanning receptors of the FZD1-10 class are bound and activated by the WNT family of lipoglycoproteins, thereby inducing a complex network of signaling pathways. However, the specificity of the interaction between mammalian WNT and FZD proteins and the subsequent signaling cascade downstream of the different WNT-FZD pairs have not been systematically addressed to date. In this study, we determined the binding affinities of various WNTs for different members of the FZD family by using bio-layer interferometry and characterized their functional selectivity in a cell system. Using purified WNTs, we show that different FZD cysteine-rich domains prefer to bind to distinct WNTs with fast on-rates and slow off-rates. In a 32D cell-based system engineered to overexpress FZD2, FZD4, or FZD5, we found that WNT-3A (but not WNT-4, -5A, or -9B) activated the WNT-ß-catenin pathway through FZD2/4/5 as measured by phosphorylation of LRP6 and ß-catenin stabilization. Surprisingly, different WNT-FZD pairs showed differential effects on phosphorylation of DVL2 and DVL3, revealing a previously unappreciated DVL isoform selectivity by different WNT-FZD pairs in 32D cells. In summary, we present extensive mapping of WNT-FZD cysteine-rich domain interactions complemented by analysis of WNT-FZD pair functionality in a unique cell system expressing individual FZD isoforms. Differential WNT-FZD binding and selective functional readouts suggest that endogenous WNT ligands evolved with an intrinsic natural bias toward different downstream signaling pathways, a phenomenon that could be of great importance in the design of FZD-targeting drugs.


Asunto(s)
Receptores Frizzled/metabolismo , Mapas de Interacción de Proteínas , Proteínas Wnt/metabolismo , Vía de Señalización Wnt , Animales , Línea Celular , Ratones , Fosforilación , Mapeo de Interacción de Proteínas , Isoformas de Proteínas/metabolismo , beta Catenina/metabolismo
9.
Mol Biol Cell ; 25(10): 1629-40, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24648492

RESUMEN

Inhibition of casein kinase 1 delta (CK1δ) blocks primary ciliogenesis in human telomerase reverse transcriptase immortalized retinal pigmented epithelial and mouse inner medullary collecting duct cells-3. Mouse embryonic fibroblasts (MEFs) and retinal cells from Csnk1d (CK1δ)-null mice also exhibit ciliogenesis defects. CK1δ catalytic activity and centrosomal localization signal (CLS) are required to rescue cilia formation in MEFs(Csnk1d null). Furthermore, expression of a truncated derivative containing the CLS displaces full-length CK1δ from the centrosome and decreases ciliary length in control MEFs, suggesting that centrosomal CK1δ has a role in ciliogenesis. CK1δ inhibition also alters pericentrosomal or ciliary distribution of several proteins involved in ciliary transport, including Ras-like in rat brain-11A, Ras-like in rat brain-8A, centrosomal protein of 290 kDa, pericentriolar material protein 1, and polycystin-2, as well as the Golgi distribution of its binding partner, A-kinase anchor protein 450 (AKAP450). As reported for AKAP450, CK1δ was required for microtubule nucleation at the Golgi and maintenance of Golgi integrity. Overexpression of an AKAP450 fragment containing the CK1δ-binding site inhibits Golgi-derived microtubule nucleation, Golgi distribution of intraflagellar transport protein 20 homologue, and ciliogenesis. Our results suggest that CK1δ mediates primary ciliogenesis by multiple mechanisms, one involving its centrosomal function and another dependent on its interaction with AKAP450 at the Golgi, where it is important for maintaining Golgi organization and polarized trafficking of multiple factors that mediate ciliary transport.


Asunto(s)
Proteínas de Anclaje a la Quinasa A/metabolismo , Quinasa Idelta de la Caseína/antagonistas & inhibidores , Centrosoma/metabolismo , Cilios/metabolismo , Aparato de Golgi/metabolismo , Proteínas Asociadas a Microtúbulos/metabolismo , Proteínas de Anclaje a la Quinasa A/biosíntesis , Animales , Antígenos de Neoplasias , Sitios de Unión , Proteínas Portadoras , Quinasa Idelta de la Caseína/genética , Proteínas de Ciclo Celular/metabolismo , Línea Celular , Proteínas del Citoesqueleto , Humanos , Ratones , Ratones Noqueados , Proteínas Asociadas a Microtúbulos/biosíntesis , Microtúbulos/metabolismo , Proteínas Nucleares/metabolismo , Transporte de Proteínas , Interferencia de ARN , ARN Interferente Pequeño , Retina/citología , Transducción de Señal/genética , Canales Catiónicos TRPP/metabolismo , Telomerasa/genética , Proteínas de Unión al GTP rab/metabolismo
10.
Hepatology ; 60(2): 576-87, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24492943

RESUMEN

UNLABELLED: Wnt signaling is important for cancer pathogenesis and is often up-regulated in hepatocellular carcinoma (HCC). Heparan sulfate proteoglycans (HSPGs) function as coreceptors or modulators of Wnt activation. Glypican-3 (GPC3) is an HSPG that is highly expressed in HCC, where it can attract Wnt proteins to the cell surface and promote cell proliferation. Thus, GPC3 has emerged as a candidate therapeutic target in liver cancer. While monoclonal antibodies to GPC3 are currently being evaluated in preclinical and clinical studies, none have shown an effect on Wnt signaling. Here, we first document the expression of Wnt3a, multiple Wnt receptors, and GPC3 in several HCC cell lines, and demonstrate that GPC3 enhanced the activity of Wnt3a/ß-catenin signaling in these cells. Then we report the identification of HS20, a human monoclonal antibody against GPC3, which preferentially recognized the heparan sulfate chains of GPC3, both the sulfated and nonsulfated portions. HS20 disrupted the interaction of Wnt3a and GPC3 and blocked Wnt3a/ß-catenin signaling. Moreover, HS20 inhibited Wnt3a-dependent cell proliferation in vitro and HCC xenograft growth in nude mice. In addition, HS20 had no detectable undesired toxicity in mice. Taken together, our results show that a monoclonal antibody primarily targeting the heparin sulfate chains of GPC3 inhibited Wnt/ß-catenin signaling in HCC cells and had potent antitumor activity in vivo. CONCLUSION: An antibody directed against the heparan sulfate of a proteoglycan shows efficacy in blocking Wnt signaling and HCC growth, suggesting a novel strategy for liver cancer therapy.


Asunto(s)
Anticuerpos Monoclonales/inmunología , Carcinoma Hepatocelular/inmunología , Glipicanos/inmunología , Heparitina Sulfato/inmunología , Neoplasias Hepáticas/inmunología , Vía de Señalización Wnt/inmunología , Animales , Anticuerpos Monoclonales/farmacología , Carcinoma Hepatocelular/tratamiento farmacológico , Técnicas de Visualización de Superficie Celular , Femenino , Células Hep G2 , Humanos , Neoplasias Hepáticas/tratamiento farmacológico , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Vía de Señalización Wnt/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto , beta Catenina/inmunología
11.
Cell Signal ; 26(1): 94-101, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24080158

RESUMEN

Wnt signaling regulates a variety of cellular processes during embryonic development and in the adult. Many of these activities are mediated by the Frizzled family of seven-pass transmembrane receptors, which bind Wnts via a conserved cysteine-rich domain (CRD). Secreted Frizzled-related proteins (sFRPs) contain an amino-terminal, Frizzled-like CRD and a carboxyl-terminal, heparin-binding netrin-like domain. Previous studies identified sFRPs as soluble Wnt antagonists that bind directly to Wnts and prevent their interaction with Frizzleds. However, subsequent observations suggested that sFRPs and Frizzleds form homodimers and heterodimers via their respective CRDs, and that sFRPs can stimulate signal transduction. Here, we present evidence that sFRP1 either inhibits or enhances signaling in the Wnt3a/ß-catenin pathway, depending on its concentration and the cellular context. Nanomolar concentrations of sFRP1 increased Wnt3a signaling, while higher concentrations blocked it in HEK293 cells expressing a SuperTopFlash reporter. sFRP1 primarily augmented Wnt3a/ß-catenin signaling in C57MG cells, but it behaved as an antagonist in L929 fibroblasts. sFRP1 enhanced reporter activity in L cells that were engineered to stably express Frizzled 5, though not Frizzled 2. This implied that the Frizzled expression pattern could determine the response to sFRP1. Similar results were obtained with sFRP2 in HEK293, C57MG and L cell reporter assays. CRDsFRP1 mimicked the potentiating effect of sFRP1 in multiple settings, contradicting initial expectations that this domain would inhibit Wnt signaling. Moreover, CRDsFRP1 showed little avidity for Wnt3a compared to sFRP1, implying that the mechanism for potentiation by CRDsFRP1 probably does not require an interaction with Wnt protein. Together, these findings demonstrate that sFRPs can either promote or suppress Wnt/ß-catenin signaling, depending on cellular context, concentration and most likely the expression pattern of Fzd receptors.


Asunto(s)
Glicoproteínas/metabolismo , Transducción de Señal , Proteína Wnt3A/metabolismo , beta Catenina/metabolismo , Animales , Receptores Frizzled/química , Receptores Frizzled/metabolismo , Células HEK293 , Humanos , Inmunoprecipitación , Péptidos y Proteínas de Señalización Intracelular , Ratones , Estructura Terciaria de Proteína , Ratas
13.
J Biol Chem ; 288(13): 9428-37, 2013 Mar 29.
Artículo en Inglés | MEDLINE | ID: mdl-23396967

RESUMEN

Dishevelled (Dvl) proteins are intracellular effectors of Wnt signaling that have essential roles in both canonical and noncanonical Wnt pathways. It has long been known that Wnts stimulate Dvl phosphorylation, but relatively little is known about its functional significance. We have previously reported that both Wnt3a and Wnt5a induce Dvl2 phosphorylation that is associated with an electrophoretic mobility shift and loss of recognition by monoclonal antibody 10B5. In the present study, we mapped the 10B5 epitope to a 16-amino acid segment of human Dvl2 (residues 594-609) that contains four Ser/Thr residues. Alanine substitution of these residues (P4m) eliminated the mobility shift induced by either Wnt3a or Wnt5a. The Dvl2 P4m mutant showed a modest increase in canonical Wnt/ß-catenin signaling activity relative to wild type. Consistent with this finding, Dvl2 4Pm preferentially localized to cytoplasmic puncta. In contrast to wild-type Dvl2, however, the P4m mutant was unable to rescue Wnt3a-dependent neurite outgrowth in TC-32 cells following suppression of endogenous Dvl2/3. Earlier work has implicated casein kinase 1δ/ε as responsible for the Dvl mobility shift, and a CK1δ in vitro kinase assay confirmed that Ser(594), Thr(595), and Ser(597) of Dvl2 are CK1 targets. Alanine substitution of these three residues was sufficient to abrogate the Wnt-dependent mobility shift. Thus, we have identified a cluster of Ser/Thr residues in the C-terminal domain of Dvl2 that are Wnt-induced phosphorylation (WIP) sites. Our results indicate that phosphorylation at the WIP sites reduces Dvl accumulation in puncta and attenuates ß-catenin signaling, whereas it enables noncanonical signaling that is required for neurite outgrowth.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/fisiología , Regulación de la Expresión Génica , Fosfoproteínas/fisiología , Proteínas Wnt/metabolismo , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Alanina/química , Animales , Medios de Cultivo Condicionados , Proteínas Dishevelled , Células HEK293 , Humanos , Mutación , Fosfoproteínas/metabolismo , Fosforilación , Proteínas Proto-Oncogénicas/metabolismo , ARN Interferente Pequeño/metabolismo , Ratas , Serina/química , Transducción de Señal , Treonina/química , Proteína Wnt-5a , Proteína Wnt3A/metabolismo
14.
J Biol Chem ; 288(13): 9438-46, 2013 Mar 29.
Artículo en Inglés | MEDLINE | ID: mdl-23396968

RESUMEN

Previously we reported that Wnt3a-dependent neurite outgrowth in Ewing sarcoma family tumor cell lines was mediated by Frizzled3, Dishevelled (Dvl), and c-Jun N-terminal kinase (Endo, Y., Beauchamp, E., Woods, D., Taylor, W. G., Toretsky, J. A., Uren, A., and Rubin, J. S. (2008) Mol. Cell. Biol. 28, 2368-2379). Subsequently, we observed that Dvl2/3 phosphorylation correlated with neurite outgrowth and that casein kinase 1δ, one of the enzymes that mediate Wnt3a-dependent Dvl phosphorylation, was required for neurite extension (Greer, Y. E., and Rubin, J. S. (2011) J. Cell Biol. 192, 993-1004). However, the functional relevance of Dvl phosphorylation in neurite outgrowth was not established. Dvl1 has been shown by others to be important for axon specification in hippocampal neurons via an interaction with atypical PKCζ, but the role of Dvl phosphorylation was not evaluated. Here we report that Ewing sarcoma family tumor cells express PKCι but not PKCζ. Wnt3a stimulated PKCι activation and caused a punctate distribution of pPKCι in the neurites and cytoplasm, with a particularly intense signal at the centrosome. Knockdown of PKCι expression with siRNA reagents blocked neurite formation in response to Wnt3a. Aurothiomalate, a specific inhibitor of PKCι/Par6 binding, also suppressed neurite extension. Wnt3a enhanced the co-immunoprecipitation of endogenous PKCι and Dvl2. Although FLAG-tagged wild-type Dvl2 immunoprecipitated with PKCι, a phosphorylation-deficient Dvl2 derivative did not. This derivative also was unable to rescue neurite outgrowth when endogenous Dvl2/3 was suppressed by siRNA (González-Sancho, J. M., Greer, Y. E., Abrahams, C. L., Takigawa, Y., Baljinnyam, B., Lee, K. H., Lee, K. S., Rubin, J. S., and Brown, A. M. (2013) J. Biol. Chem. 288, 9428-9437). Taken together, these results suggest that site-specific Dvl2 phosphorylation is required for Dvl2 association with PKCι. This interaction is likely to be one of the mechanisms essential for Wnt3a-dependent neurite outgrowth.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Neuritas/metabolismo , Fosfoproteínas/metabolismo , Proteína Quinasa C/fisiología , Proteína Wnt3A/metabolismo , Quinasa de la Caseína I/metabolismo , Proteínas Dishevelled , Células HEK293 , Hipocampo/metabolismo , Humanos , Isoenzimas/metabolismo , Microscopía Fluorescente/métodos , Modelos Biológicos , Neuronas/metabolismo , Fosforilación , Proteína Quinasa C/metabolismo , Estructura Terciaria de Proteína , ARN Interferente Pequeño/metabolismo , Transducción de Señal
15.
Cell Signal ; 25(1): 178-89, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23022962

RESUMEN

Cripto-1 is implicated in multiple cellular events, including cell proliferation, motility and angiogenesis, through the activation of an intricate network of signaling pathways. A crosstalk between Cripto-1 and the canonical Wnt/ß-catenin signaling pathway has been previously described. In fact, Cripto-1 is a downstream target gene of the canonical Wnt/ß-catenin signaling pathway in the embryo and in colon cancer cells and T-cell factor (Tcf)/lymphoid enhancer factor binding sites have been identified in the promoter and the first intronic region of the mouse and human Cripto-1 genes. We now demonstrate that Cripto-1 modulates signaling through the canonical Wnt/ß-catenin/Tcf pathway by binding to the Wnt co-receptors low-density lipoprotein receptor-related protein (LRP) 5 and LRP6, which facilitates Wnt3a binding to LRP5 and LRP6. Cripto-1 functionally enhances Wnt3a signaling through cytoplasmic stabilization of ß-catenin and elevated ß-catenin/Tcf transcriptional activation. Conversely, Wnt3a further increases Cripto-1 stimulation of migration, invasion and colony formation in soft agar of HC11 mouse mammary epithelial cells, indicating that Cripto-1 and the canonical Wnt/ß-catenin signaling co-operate in regulating motility and in vitro transformation of mammary epithelial cells.


Asunto(s)
Proteínas Ligadas a GPI/metabolismo , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Proteína-5 Relacionada con Receptor de Lipoproteína de Baja Densidad/metabolismo , Proteína-6 Relacionada a Receptor de Lipoproteína de Baja Densidad/metabolismo , Proteínas de Neoplasias/metabolismo , Vía de Señalización Wnt , Animales , Línea Celular , Movimiento Celular , Proteínas Ligadas a GPI/química , Células HEK293 , Humanos , Péptidos y Proteínas de Señalización Intercelular/química , Proteína-5 Relacionada con Receptor de Lipoproteína de Baja Densidad/genética , Proteína-6 Relacionada a Receptor de Lipoproteína de Baja Densidad/genética , Ratones , Proteínas de Neoplasias/química , Unión Proteica , Estructura Terciaria de Proteína , Proteína Smad2/metabolismo , Proteína smad3/metabolismo , Activación Transcripcional , Proteína Wnt3A/metabolismo , beta Catenina/metabolismo
16.
Invest Ophthalmol Vis Sci ; 53(11): 7043-51, 2012 Oct 09.
Artículo en Inglés | MEDLINE | ID: mdl-22956608

RESUMEN

PURPOSE: We previously discovered elevated levels of secreted frizzled-related protein 1 (sFRP1), the Wnt signaling pathway inhibitor, in the glaucomatous trabecular meshwork (GTM), and found that key canonical Wnt signaling pathway genes are expressed in the trabecular meshwork (TM). The purpose of our study was to determine whether a functional canonical Wnt signaling pathway exists in the human TM (HTM). METHODS: Western immunoblotting and/or immunofluorescent microscopy were used to study ß-catenin translocation as well as the actin cytoskeleton in transformed and primary HTM cells. A TCF/LEF luciferase assay was used to study functional canonical Wnt signaling, which was confirmed further by WNT3a-induced expression of a pathway target gene, AXIN2, via quantitative PCR. Intravitreal injection of an Ad5 adenovirus expressing Dickkopf-related protein-1 (DKK1) was used to study the in vivo effect of canonical Wnt signaling on IOP in mice. RESULTS: WNT3a induced ß-catenin translocation in the HTM, which was blocked by co-treatment with sFRP1. Similarly, WNT3a enhanced luciferase levels in TCF/LEF luciferase assays, which also were blocked by sFRP1. Furthermore, AXIN2 expression was elevated significantly by WNT3a. However, neither WNT3a nor sFRP1 affected actin cytoskeleton organization, which theoretically could be regulated by noncanonical Wnt signaling in HTM cells. Exogenous DKK1, a specific inhibitor for the canonical Wnt signaling pathway, or sFRP1 elevated mouse IOP to equivalent levels. CONCLUSIONS: There is a canonical Wnt signaling pathway in the TM, and this canonical Wnt pathway, but not the noncanonical Wnt signaling pathway, regulates IOP.


Asunto(s)
Malla Trabecular/metabolismo , Vía de Señalización Wnt/fisiología , Actinas/metabolismo , Adenoviridae/genética , Proteína Axina/genética , Western Blotting , Células Cultivadas , Densitometría , Electroforesis en Gel de Poliacrilamida , Expresión Génica , Humanos , Péptidos y Proteínas de Señalización Intercelular , Presión Intraocular/fisiología , Inyecciones Intravítreas , Microscopía Fluorescente , Transporte de Proteínas/efectos de los fármacos , Reacción en Cadena en Tiempo Real de la Polimerasa , Malla Trabecular/efectos de los fármacos , Proteína Wnt3A/farmacología , beta Catenina/metabolismo
17.
Stem Cells ; 30(5): 865-75, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-22290867

RESUMEN

Secreted Frizzled related proteins (sFRPs) are a family of proteins that modulate Wnt signaling, which in turn regulates multiple aspects of ventral midbrain (VM) and dopamine (DA) neuron development. However, it is not known which Wnt signaling branch and what aspects of midbrain DA neuron development are regulated by sFRPs. Here, we show that sFRP1 and sFRP2 activate the Wnt/planar-cell-polarity/Rac1 pathway in DA cells. In the developing VM, sFRP1 and sFRP2 are expressed at low levels, and sFRP1-/- or sFRP2-/- mice had no detectable phenotype. However, compound sFRP1-/-;sFRP2-/- mutants revealed a Wnt/PCP phenotype similar to that previously described for Wnt5a-/- mice. This included an anteroposterior shortening of the VM, a lateral expansion of the Shh domain and DA lineage markers (Lmx1a and Th), as well as an accumulation of Nurr1+ precursors in the VM. In vitro experiments showed that, while very high concentrations of SFRP1 had a negative effect on cell survival, low/medium concentrations of sFRP1 or sFRP2 promoted the DA differentiation of progenitors derived from primary VM cultures or mouse embryonic stem cells (ESCs), mimicking the effects of Wnt5a. We thus conclude that the main function of sFRP1 and sFRP2 is to enhance Wnt/PCP signaling in DA cells and to regulate Wnt/PCP-dependent functions in midbrain development. Moreover, we suggest that low-medium concentrations of sFRPs may be used to enhance the DA differentiation of ESCs and improve their therapeutic application.


Asunto(s)
Neuronas Dopaminérgicas/metabolismo , Células Madre Embrionarias/metabolismo , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Proteínas de la Membrana/metabolismo , Mesencéfalo/embriología , Proteínas del Tejido Nervioso/metabolismo , Animales , Neuronas Dopaminérgicas/citología , Relación Dosis-Respuesta a Droga , Células Madre Embrionarias/citología , Péptidos y Proteínas de Señalización Intercelular/genética , Péptidos y Proteínas de Señalización Intercelular/farmacología , Proteínas de la Membrana/genética , Proteínas de la Membrana/farmacología , Mesencéfalo/citología , Ratones , Ratones Noqueados , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/farmacología , Ratas , Ratas Sprague-Dawley , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología , Proteínas Wnt/genética , Proteínas Wnt/metabolismo
18.
PLoS One ; 7(1): e29455, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22238613

RESUMEN

R-spondins (Rspos) comprise a family of four secreted proteins that have important roles in cell proliferation, cell fate determination and organogenesis. Rspos typically exert their effects by potentiating the Wnt/ß-catenin signaling pathway. To systematically investigate the impact of Rspo/Wnt on gene expression, we performed a microarray analysis using C57MG mouse mammary epithelial cells treated with recombinant Rspo2 and/or Wnt3a. We observed the up- and down-regulation of several previously unidentified target genes, including ones that encode proteins involved in immune responses, effectors of other growth factor signaling pathways and transcription factors. Dozens of these changes were validated by quantitative real time RT-PCR. Time course experiments showed that Rspo2 typically had little or no effect on Wnt-dependent gene expression at 3 or 6 h, but enhanced expression at 24 h, consistent with biochemical data indicating that Rspo2 acts primarily to sustain rather than acutely increase Wnt pathway activation. Up-regulation of gene expression was inhibited by pre-treatment with Dickkopf1, a Wnt/ß-catenin pathway antagonist, and by siRNA knockdown of ß-catenin expression. While Dickkopf1 blocked Rspo2/Wnt3a-dependent down-regulation, a number of down-regulated genes were not affected by ß-catenin knockdown, suggesting that in these instances down-regulation was mediated by a ß-catenin-independent mechanism.


Asunto(s)
Células Epiteliales/efectos de los fármacos , Regulación de la Expresión Génica/efectos de los fármacos , Glándulas Mamarias Animales/efectos de los fármacos , Trombospondinas/farmacología , Proteína Wnt3A/farmacología , Animales , Línea Celular , Regulación hacia Abajo/efectos de los fármacos , Células Epiteliales/metabolismo , Femenino , Perfilación de la Expresión Génica , Glándulas Mamarias Animales/metabolismo , Ratones , Análisis por Micromatrices , ARN Interferente Pequeño/farmacología , Proteínas Recombinantes/farmacología , Trombospondinas/fisiología , Regulación hacia Arriba/efectos de los fármacos , Estudios de Validación como Asunto , Proteína Wnt3A/fisiología , beta Catenina/antagonistas & inhibidores , beta Catenina/genética
19.
Exp Eye Res ; 97(1): 130-6, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22248913

RESUMEN

Glaucoma is a leading cause of blindness worldwide. In primary open angle glaucoma (POAG) patients, impaired trabecular meshwork (TM) function results in elevated intraocular pressure (IOP), which is the primary risk factor of developing optic neuropathy. Our previous studies showed that Wnt signaling pathway components are expressed in the human TM (HTM), and the Wnt inhibitor, secreted frizzled-related protein 1 (SFRP1) is elevated in the glaucomatous TM (GTM). Elevated SFRP1 increased IOP in mice eyes and in perfusion cultured anterior segments of the human eye. However, the cause of elevated SFRP1 in the GTM remains unknown. Promoter methylation plays a key role in regulating SFRP1 expression in certain cancer cells. In light of this, we studied whether promoter methylation is also involved in SFRP1 differential expression in the TM. Two normal TM (NTM) and two GTM cell strains were cultured for an additional 7 days after they were confluent. RNA and genomic DNA (gDNA) were isolated simultaneously to compare SFRP1 expression levels by quantitative PCR (qPCR) and to determine SFRP1 promoter methylation status by bisulfite conversion and methylation-sensitive high resolution melting analysis (MS-HRM). To study whether DNA methylation inhibitors affect SFRP1 expression in TM cells, the four TM cell strains were treated with or without 2 µM 5-aza-2'-deoxycytidine (AZA-dC) for 4 days. RNA was isolated to compare SFRP1 expression by qPCR. In addition, a human cancer cell line, NCI-H460, was used as a positive control. We found that the two GTM cell strains had significantly higher expression levels of SFRP1 than the two NTM cell strains. However, the SFRP1 promoter of all four TM cell strains was unmethylated. In addition, AZA-dC treatment did not affect SFRP1 expression in any of the TM cell strains (n = 3, p > 0.05). In contrast, the hypermethylated SFRP1 promoter of NCI-H460 cells was partially demethylated by the same treatment. AZA-dC treatment also elevated SFRP1 expression by approximately two fold in NCI-H460 cells (n = 3, p < 0.01). Our data suggest that the differential expression of SFRP1 in HTM cells is not due to differential promoter methylation.


Asunto(s)
Metilación de ADN , Regulación de la Expresión Génica/fisiología , Glaucoma de Ángulo Abierto/genética , Péptidos y Proteínas de Señalización Intercelular/genética , Proteínas de la Membrana/genética , Regiones Promotoras Genéticas/genética , Malla Trabecular/metabolismo , Anciano , Anciano de 80 o más Años , Azacitidina/análogos & derivados , Azacitidina/farmacología , Células Cultivadas , Metilasas de Modificación del ADN , Cartilla de ADN/química , Decitabina , Inhibidores Enzimáticos/farmacología , Femenino , Glaucoma de Ángulo Abierto/metabolismo , Humanos , Masculino , Persona de Mediana Edad , Reacción en Cadena en Tiempo Real de la Polimerasa
20.
J Cell Physiol ; 227(5): 1960-71, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-21732367

RESUMEN

Rspo2 was identified as a novel common integration site (CIS) for the mouse mammary tumor virus (MMTV) in viral induced mouse mammary tumors. Here we show that Rspo2 modulates Wnt signaling in mouse mammary epithelial cells. Co-expression of both genes resulted in an intermediate growth phenotype on plastic and had minor effects on the growth-promoting properties of Wnt1 in soft agar. However, individual Rspo2 and Wnt1 HC11 transfectants as well as the double transfectant were tumorigenic in athymic nude mice, with tumors from each line having distinctive histological characteristics. Rspo2 and Rspo2/Wnt1 tumors contained many spindle cells, consistent with an epithelial-mesenchymal transformation (EMT) phenotype. When Rspo2 and Rspo2/Wnt1 tumor cells were transferred into naïve mice, they exhibited greater metastatic activity than cells derived from Wnt1 tumors. For comparison, C57MG/Wnt1/Rspo2 co-transfectants exhibited invasive properties in three-dimensional (3D) Matrigel cultures that were not seen with cells transfected only with Wnt1 or Rspo2. Use of Dickkopf-1, a specific antagonist of the Wnt/ß-catenin pathway, or short hairpin RNA targeting ß-catenin expression demonstrated that the invasive activity was not mediated by ß-catenin. Our results indicate that Rspo2 and Wnt1 have mutually distinct effects on mammary epithelial cell growth and these effects are context-dependent. While Rspo2 and Wnt1 act synergistically in the ß-catenin pathway, other mechanisms are responsible for the invasive properties of stable double transfectants observed in 3D Matrigel cultures.


Asunto(s)
Células Epiteliales/patología , Células Epiteliales/fisiología , Glándulas Mamarias Animales , Neoplasias Mamarias Experimentales/metabolismo , Neoplasias Mamarias Experimentales/patología , Trombospondinas/metabolismo , Animales , Línea Celular , Células Epiteliales/citología , Femenino , Proteína-1 Reguladora de Fusión/metabolismo , Perfilación de la Expresión Génica , Humanos , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Neoplasias Pulmonares/secundario , Glándulas Mamarias Animales/citología , Glándulas Mamarias Animales/metabolismo , Glándulas Mamarias Animales/patología , Virus del Tumor Mamario del Ratón/genética , Virus del Tumor Mamario del Ratón/metabolismo , Ratones , Ratones Desnudos , Invasividad Neoplásica/patología , Trasplante de Neoplasias , Trombospondinas/genética , Vía de Señalización Wnt/fisiología , Proteína Wnt1/genética , Proteína Wnt1/metabolismo , beta Catenina/genética , beta Catenina/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...