Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
1.
RNA Biol ; 14(2): 236-244, 2017 02.
Artículo en Inglés | MEDLINE | ID: mdl-27981895

RESUMEN

Alternative polyadenylation (APA) is a step in mRNA 3'-end processing that contributes to the complexity of the transcriptome by generating isoforms that differ in either their coding sequence or their 3'-untranslated regions (UTRs). The EPB41 genes, EPB41, EPB41L2, EPB41L3 and EPB41L1, encode an impressively complex array of structural adaptor proteins (designated 4.1R, 4.1G, 4.1B and 4.1N, respectively) by using alternative transcriptional promoters and tissue-specific alternative pre-mRNA splicing. The great variety of 4.1 proteins mainly results from 5'-end and internal processing of the EPB41 pre-mRNAs. Thus, 4.1 proteins can vary in their N-terminal extensions but all contain a highly homologous C-terminal domain (CTD). Here we study a new group of EPB41-related mRNAs that originate by APA and lack the exons encoding the CTD characteristic of prototypical 4.1 proteins, thereby encoding a new type of 4.1 protein. For the EPB41 gene, this type of processing was observed in all 11 human tissues analyzed. Comparative genomic analysis of EPB41 indicates that APA is conserved in various mammals. In addition, we show that APA also functions for the EPB41L2, EPB41L3 and EPB41L1 genes, but in a more restricted manner in the case of the latter 2 than it does for the EPB41 and EPB41L2 genes. Our study shows alternative polyadenylation to be an additional mechanism for the generation of 4.1 protein diversity in the already complex EPB41-related genes. Understanding the diversity of EPB41 RNA processing is essential for a full appreciation of the many 4.1 proteins expressed in normal and pathological tissues.


Asunto(s)
Empalme Alternativo , Proteínas del Citoesqueleto/genética , Regulación de la Expresión Génica , Proteínas de la Membrana/genética , Poliadenilación , Regiones no Traducidas 3' , Secuencia de Bases , Encéfalo/metabolismo , ADN Complementario , Exones , Humanos , Intrones , Neuropéptidos , Especificidad de Órganos/genética , ARN Mensajero/genética , Transcripción Genética
2.
Breast Cancer Res Treat ; 155(3): 431-40, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26860947

RESUMEN

The central role of HER2 as the disease driver and HER3 as its essential partner has made them rational targets for the treatment of HER2-amplifed breast cancers, and there is considerable interest in developing highly effective treatment regimens for this disease that consist of targeted therapies alone. Much of these efforts are focused on dual targeting approaches, particularly dual targeting of the HER2-HER3 tumor driver complex itself, or vertical combinations that target downstream PI3K or Akt in addition to HER2. There is also potential in lateral combinations based on evidence implicating cross-talk with other membrane receptor systems, particularly integrins, and such lateral combinations can potentially involve either HER2 or HER3. We established a preclinical model of targeting HER3 using doxycycline-inducible shRNA and determined the efficacy of a ß1 integrin inhibitor in combination with targeting HER3. We report that targeting HER3 and ß1 integrin provides a particularly effective combination therapy approach for HER2-amplified cancers, surpassing the combination of HER2 and ß1 integrin targeting, and evading some of the safety concerns associated with direct HER2-targeting. This further validates HER3 as a major hub mediating the tumorigenic functions of HER2 and identifies it as a high value target for lateral combination therapy strategies.


Asunto(s)
Neoplasias de la Mama/tratamiento farmacológico , Doxiciclina/administración & dosificación , Integrina beta1/genética , Receptor ErbB-2/genética , Receptor ErbB-3/genética , Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Resistencia a Antineoplásicos/genética , Femenino , Humanos , Integrina beta1/efectos de los fármacos , Terapia Molecular Dirigida , Fosfatidilinositol 3-Quinasas/genética , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/uso terapéutico , Receptor ErbB-2/antagonistas & inhibidores , Receptor ErbB-3/antagonistas & inhibidores , Transducción de Señal/efectos de los fármacos , Resultado del Tratamiento
3.
J Cell Sci ; 126(Pt 20): 4589-601, 2013 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-23943871

RESUMEN

The microtubule (MT) cytoskeleton is essential for many cellular processes, including cell polarity and migration. Cortical platforms, formed by a subset of MT plus-end-tracking proteins, such as CLASP2, and non-MT binding proteins such as LL5ß, attach distal ends of MTs to the cell cortex. However, the mechanisms involved in organizing these platforms have not yet been described in detail. Here we show that 4.1R, a FERM-domain-containing protein, interacts and colocalizes with cortical CLASP2 and is required for the correct number and dynamics of CLASP2 cortical platforms. Protein 4.1R also controls binding of CLASP2 to MTs at the cell edge by locally altering GSK3 activity. Furthermore, in 4.1R-knockdown cells MT plus-ends were maintained for longer in the vicinity of cell edges, but instead of being tethered to the cell cortex, MTs continued to grow, bending at cell margins and losing their radial distribution. Our results suggest a previously unidentified role for the scaffolding protein 4.1R in locally controlling CLASP2 behavior, CLASP2 cortical platform turnover and GSK3 activity, enabling correct MT organization and dynamics essential for cell polarity.


Asunto(s)
Proteínas del Citoesqueleto/metabolismo , Citoesqueleto/metabolismo , Proteínas de la Membrana/metabolismo , Proteínas Asociadas a Microtúbulos/metabolismo , Microtúbulos/metabolismo , Movimiento Celular/fisiología , Polaridad Celular/fisiología , Técnicas de Silenciamiento del Gen , Células HEK293 , Humanos , Proteínas Asociadas a Microtúbulos/genética , Unión Proteica , Estructura Terciaria de Proteína
4.
J Cell Sci ; 124(Pt 15): 2529-38, 2011 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-21750196

RESUMEN

In red blood cells, multifunctional protein 4.1R stabilizes the spectrin-actin network and anchors it to the plasma membrane. To contribute to the characterization of functional roles of 4.1R in nonerythroid cells, we have analyzed the participation of protein 4.1R in cell migration. The distribution of endogenous 4.1R is polarized towards the leading edge of migrating cells. Exogenous 4.1R isoforms containing a complete membrane-binding domain consistently localized to plasma membrane extensions enriched in F-actin. Silencing of 4.1R caused the loss of persistence of migration in subconfluent cells and of directional migration in cells moving into a wound. Coimmunoprecipitation and pull-down assays identified the scaffold protein IQGAP1 as a partner for protein 4.1R and showed that the 4.1R membrane-binding domain is involved in binding IQGAP1. Importantly, we show that protein 4.1R is necessary for the localization of IQGAP1 to the leading edge of cells migrating into a wound, whereas IQGAP1 is not required for protein 4.1R localization. Collectively, our results indicate a crucial role for protein 4.1R in cell migration and in the recruitment of the scaffold protein IQGAP1 to the cell front.


Asunto(s)
Movimiento Celular/fisiología , Proteínas del Citoesqueleto/metabolismo , Proteínas de la Membrana/metabolismo , Proteínas Activadoras de ras GTPasa/metabolismo , Animales , Western Blotting , Células COS , Línea Celular , Línea Celular Tumoral , Movimiento Celular/genética , Chlorocebus aethiops , Proteínas del Citoesqueleto/química , Proteínas del Citoesqueleto/genética , Técnica del Anticuerpo Fluorescente , Humanos , Inmunoprecipitación , Proteínas de la Membrana/química , Proteínas de la Membrana/genética , Unión Proteica , Estructura Terciaria de Proteína , Transporte de Proteínas/genética , Transporte de Proteínas/fisiología , Interferencia de ARN , Proteínas Activadoras de ras GTPasa/genética
5.
STAR Protoc ; 4(2): 102224, 2023 Apr 17.
Artículo en Inglés | MEDLINE | ID: mdl-37071532

RESUMEN

Current protocols measure antibody-dependent cellular cytotoxicity (ADCC) in vitro using peripheral blood mononuclear cells (PBMCs), but isolation and variability among donors limit the viability and reproducibility of this approach. Here, we present a standardized co-culture model system to quantify ADCC on human breast cancer cells. We describe steps to engineer a natural killer cell line that stably expresses FCγRIIIa (CD16), required to mediate ADCC. We then detail the steps for the cancer-immune co-culture setup, followed by cytotoxicity measurement and analysis.

6.
Nat Cancer ; 4(2): 240-256, 2023 02.
Artículo en Inglés | MEDLINE | ID: mdl-36759733

RESUMEN

BRAFV600E mutation confers a poor prognosis in metastatic colorectal cancer (CRC) despite combinatorial targeted therapies based on the latest understanding of signaling circuitry. To identify parallel resistance mechanisms induced by BRAF-MEK-EGFR co-targeting, we used a high-throughput kinase activity mapping platform. Here we show that SRC kinases are systematically activated in BRAFV600E CRC following targeted inhibition of BRAF ± EGFR and that coordinated targeting of SRC with BRAF ± EGFR increases treatment efficacy in vitro and in vivo. SRC drives resistance to BRAF ± EGFR targeted therapy independently of ERK signaling by inducing transcriptional reprogramming through ß-catenin (CTNNB1). The EGFR-independent compensatory activation of SRC kinases is mediated by an autocrine prostaglandin E2 loop that can be blocked with cyclooxygenase-2 (COX2) inhibitors. Co-targeting of COX2 with BRAF + EGFR promotes durable suppression of tumor growth in patient-derived tumor xenograft models. COX2 inhibition represents a drug-repurposing strategy to overcome therapeutic resistance in BRAFV600E CRC.


Asunto(s)
Neoplasias Colorrectales , Proteínas Proto-Oncogénicas B-raf , Humanos , Ciclooxigenasa 2/genética , Ciclooxigenasa 2/uso terapéutico , Proteínas Proto-Oncogénicas B-raf/genética , Proteínas Proto-Oncogénicas B-raf/metabolismo , Sistema de Señalización de MAP Quinasas , Neoplasias Colorrectales/tratamiento farmacológico , Neoplasias Colorrectales/genética , Neoplasias Colorrectales/patología , Receptores ErbB/genética , Familia-src Quinasas/genética , Familia-src Quinasas/uso terapéutico
7.
Cell Rep ; 38(5): 110285, 2022 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-35108526

RESUMEN

Surface-targeting biotherapeutic agents have been successful in treating HER2-amplified cancers through immunostimulation or chemodelivery but have failed to produce effective inhibitors of constitutive HER2-HER3 signaling. We report an extensive structure-function analysis of this tumor driver, revealing complete uncoupling of intracellular signaling and tumorigenic function from regulation or constraints from their extracellular domains (ECDs). The canonical HER3 ECD conformational changes and exposure of the dimerization interface are nonessential, and the entire ECDs of HER2 and HER3 are redundant for tumorigenic signaling. Restricting the proximation of partner ECDs with bulk and steric clash through extremely disruptive receptor engineering leaves tumorigenic signaling unperturbed. This is likely due to considerable conformational flexibilities across the span of these receptor molecules and substantial undulations in the plane of the plasma membrane, none of which had been foreseen as impediments to targeting strategies. The massive overexpression of HER2 functionally and physically uncouples intracellular signaling from extracellular constraints.


Asunto(s)
Anticuerpos Monoclonales Humanizados/farmacología , Neoplasias de la Mama/tratamiento farmacológico , Carcinogénesis/efectos de los fármacos , Receptor ErbB-2/efectos de los fármacos , Receptor ErbB-3/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Animales , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Humanos , Fosforilación/efectos de los fármacos , Receptor ErbB-2/metabolismo , Receptor ErbB-3/metabolismo , Trastuzumab/farmacología
8.
Cell Rep ; 38(5): 110291, 2022 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-35108525

RESUMEN

Effective inactivation of the HER2-HER3 tumor driver has remained elusive because of the challenging attributes of the pseudokinase HER3. We report a structure-function study of constitutive HER2-HER3 signaling to identify opportunities for targeting. The allosteric activation of the HER2 kinase domain (KD) by the HER3 KD is required for tumorigenic signaling and can potentially be targeted by allosteric inhibitors. ATP binding within the catalytically inactive HER3 KD provides structural rigidity that is important for signaling, but this is mimicked, not opposed, by small molecule ATP analogs, reported here in a bosutinib-bound crystal structure. Mutational disruption of ATP binding and molecular dynamics simulation of the apo KD of HER3 identify a conformational coupling of the ATP pocket with a hydrophobic AP-2 pocket, analogous to EGFR, that is critical for tumorigenic signaling and feasible for targeting. The value of these potential target sites is confirmed in tumor growth assays using gene replacement techniques.


Asunto(s)
Neoplasias de la Mama/metabolismo , Carcinogénesis/efectos de los fármacos , Receptor ErbB-2/metabolismo , Receptor ErbB-3/metabolismo , Compuestos de Anilina/farmacología , Neoplasias de la Mama/tratamiento farmacológico , Línea Celular Tumoral , Femenino , Humanos , Nitrilos/farmacología , Quinolinas/farmacología , Receptor ErbB-2/efectos de los fármacos , Transducción de Señal/fisiología
9.
Biochem Pharmacol ; 183: 114317, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33152346

RESUMEN

Coibamide A is a potent cancer cell toxin and one of a select group of natural products that inhibit protein entry into the secretory pathway via a direct inhibition of the Sec61 protein translocon. Many Sec61 client proteins are clinically relevant drug targets once trafficked to their final destination in or outside the cell, however the use of Sec61 inhibitors to block early biosynthesis of specific proteins is at a pre-clinical stage. In the present study we evaluated the action of coibamide A against human epidermal growth factor receptor (HER, ErbB) proteins in representative breast and lung cancer cell types. HERs were selected for this study as they represent a family of Sec61 clients that is frequently dysregulated in human cancers, including coibamide-sensitive cell types. Although coibamide A inhibits biogenesis of a broad range of Sec61 substrate proteins in a presumed substrate-nonselective manner, endogenous HER3 (ErbB-3) and EGFR (ErbB-1) proteins were more sensitive to coibamide A, and the related Sec61 inhibitor apratoxin A, than HER2 (ErbB-2). Despite this rank order of sensitivity (HER3 > EGFR > HER2), Sec61-dependent inhibition by coibamide A was sufficient to decrease cell surface expression of HER2. We report that coibamide A- or apratoxin A-mediated block of HER3 entry into the secretory pathway is unlikely to be mediated by the HER3 signal peptide alone. HER3 (G11L/S15L), that is fully resistant to the highly substrate-selective cotransin analogue CT8, was more resistant than wild-type HER3 but only at low coibamide A (3 nM) concentrations; HER3 (G11L/S15L) expression was inhibited by higher concentrations of either natural product. Time- and concentration-dependent decreases in HER protein expression induced a commensurate reduction in AKT/MAPK signaling in breast and lung cancer cell types and loss in cell viability. Coibamide A potentiated the cytotoxic efficacy of small molecule kinase inhibitors lapatinib and erlotinib in breast and lung cancer cell types, respectively. These data indicate that natural product modulators of Sec61 function have value as chemical probes to interrogate HER/ErbB signaling in treatment-resistant human cancers.


Asunto(s)
Depsipéptidos/administración & dosificación , Sistemas de Liberación de Medicamentos/métodos , Canales de Translocación SEC/antagonistas & inhibidores , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/fisiología , Relación Dosis-Respuesta a Droga , Receptores ErbB/antagonistas & inhibidores , Receptores ErbB/metabolismo , Células HEK293 , Humanos , Células MCF-7 , Canales de Translocación SEC/metabolismo
10.
Mol Cancer Res ; 19(6): 957-967, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-33727342

RESUMEN

The Src family kinases (SFK) are homologs of retroviral oncogenes, earning them the label of proto-oncogenes. Their functions are influenced by positive and negative regulatory tyrosine phosphorylation events and inhibitory and activating intramolecular and extramolecular interactions. This regulation is disrupted in their viral oncogene counterparts. However, in contrast to most other proto-oncogenes, the genetic alteration of these genes does not seem to occur in human tumors and how and whether their functions are altered in human cancers remain to be determined. To look for proteomic-level alterations, we took a more granular look at the activation states of SFKs based on their two known regulatory tyrosine phosphorylations, but found no significant differences in their activity states when comparing immortalized epithelial cells with cancer cells. SFKs are known to have other less well-studied phosphorylations, particularly within their unstructured N-terminal unique domains (UD), although their role in cancers has not been explored. In comparing panels of epithelial cells with cancer cells, we found a decrease in S17 phosphorylation in the UD of Src in cancer cells. Dephosphorylated S17 favors the dimerization of Src that is mediated through the UD and suggests increased Src dimerization in cancers. These data highlight the important role of the UD of Src and suggest that a deeper understanding of proteomic-level alterations of the unstructured UD of SFKs may provide considerable insights into how SFKs are deregulated in cancers. IMPLICATIONS: This work highlights the role of the N-terminal UD of Src kinases in regulating their signaling functions and possibly in their deregulation in human cancers.


Asunto(s)
Proteoma/metabolismo , Proteómica/métodos , Serina/metabolismo , Familia-src Quinasas/metabolismo , Sitios de Unión , Línea Celular , Línea Celular Tumoral , Activación Enzimática , Células Epiteliales/metabolismo , Humanos , Espectrometría de Masas/métodos , Microscopía Fluorescente/métodos , Mutación , Fosforilación , Unión Proteica , Serina/genética , Familia-src Quinasas/genética
11.
Nat Cell Biol ; 21(6): 778-790, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-31160710

RESUMEN

Phosphorylation networks intimately regulate mechanisms of response to therapies. Mapping the phospho-catalytic profile of kinases in cells or tissues remains a challenge. Here, we introduce a practical high-throughput system to measure the enzymatic activity of kinases using biological peptide targets as phospho-sensors to reveal kinase dependencies in tumour biopsies and cell lines. A 228-peptide screen was developed to detect the activity of >60 kinases, including ABLs, AKTs, CDKs and MAPKs. Focusing on BRAFV600E tumours, we found mechanisms of intrinsic resistance to BRAFV600E-targeted therapy in colorectal cancer, including targetable parallel activation of PDPK1 and PRKCA. Furthermore, mapping the phospho-catalytic signatures of melanoma specimens identifies RPS6KB1 and PIM1 as emerging druggable vulnerabilities predictive of poor outcome in BRAFV600E patients. The results show that therapeutic resistance can be caused by the concerted upregulation of interdependent pathways. Our kinase activity-mapping system is a versatile strategy that innovates the exploration of actionable kinases for precision medicine.


Asunto(s)
Proteínas Quinasas Dependientes de 3-Fosfoinosítido/genética , Neoplasias Colorrectales/tratamiento farmacológico , Melanoma/tratamiento farmacológico , Proteína Quinasa C-alfa/genética , Proteínas Proto-Oncogénicas c-pim-1/genética , Adulto , Anciano , Línea Celular Tumoral , Neoplasias Colorrectales/genética , Neoplasias Colorrectales/patología , Resistencia a Antineoplásicos/genética , Femenino , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Indoles/química , Estimación de Kaplan-Meier , Sistema de Señalización de MAP Quinasas/genética , Masculino , Melanoma/genética , Melanoma/patología , Persona de Mediana Edad , Péptidos/química , Péptidos/uso terapéutico , Fosforilación/efectos de los fármacos , Inhibidores de Proteínas Quinasas/uso terapéutico , Sulfonamidas/uso terapéutico
12.
Cell Rep ; 25(2): 449-463.e4, 2018 10 09.
Artículo en Inglés | MEDLINE | ID: mdl-30304684

RESUMEN

The mode of regulation of Src kinases has been elucidated by crystallographic studies identifying conserved structured protein modules involved in an orderly set of intramolecular associations and ligand interactions. Despite these detailed insights, much of the complex behavior and diversity in the Src family remains unexplained. A key missing piece is the function of the unstructured N-terminal region. We report here the function of the N-terminal region in binding within a hydrophobic pocket in the kinase domain of a dimerization partner. Dimerization substantially enhances autophosphorylation and phosphorylation of selected substrates, and interfering with dimerization is disruptive to these functions. Dimerization and Y419 phosphorylation are codependent events creating a bistable switch. Given the versatility inherent in this intrinsically disordered region, its multisite phosphorylations, and its divergence within the family, the unique domain likely functions as a central signaling hub overseeing much of the activities and unique functions of Src family kinases.


Asunto(s)
Proteínas Intrínsecamente Desordenadas/metabolismo , Multimerización de Proteína , Dominios Homologos src , Familia-src Quinasas/química , Familia-src Quinasas/metabolismo , Sitios de Unión , Células HEK293 , Humanos , Proteínas Intrínsecamente Desordenadas/química , Ligandos , Modelos Moleculares , Fosforilación , Unión Proteica , Conformación Proteica , Transducción de Señal
13.
Cancer Res ; 78(13): 3645-3658, 2018 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-29760043

RESUMEN

Current evidence suggests that HER2-driven tumorigenesis requires HER3. This is likely due to the unique ability of HER3 to activate PI3K/Akt pathway signaling, which is not directly accessible to HER2. By genetic elimination of HER3 or shRNA knockdown of HER3 in HER2-amplified cancer cells, we find residual HER2-driven activation of PI3K/Akt pathway signaling that is driven by HER2 through direct and indirect mechanisms. Indirect mechanisms involved second messenger pathways, including Ras or Grb2. Direct binding of HER2 to PI3K occurred through p-Tyr1139, which has a weak affinity for PI3K but becomes significant at very high expression and phosphorylation. Mutation of Y1139 impaired the tumorigenic competency of HER2. Total elimination of HER3 expression in HCC1569 HER2-amplified cancer cells significantly impaired tumorigenicity only transiently, overcome by subsequent increases in HER2 expression and phosphorylation with binding and activation of PI3K. In contrast to activation of oncogenes by mutation, activation by overexpression was quantitative in nature: weak intrinsic activities were strengthened by overexpression, with additional gains observed through further increases in expression. Collectively, these data show that progressive functional gains by HER2 can increase its repertoire of activities such as the activation of PI3K and overcome its dependency on HER3.Significance: The intrinsic ability of HER2 to activate PI3K correlates with increased HER2 expression and can supplant the dependency upon HER3 for growth in HER2-amplified cancers. Cancer Res; 78(13); 3645-58. ©2018 AACR.


Asunto(s)
Neoplasias de la Mama/patología , Carcinogénesis/patología , Receptor ErbB-2/metabolismo , Receptor ErbB-3/metabolismo , Animales , Neoplasias de la Mama/genética , Línea Celular Tumoral , Proliferación Celular , Femenino , Técnicas de Inactivación de Genes , Humanos , Ratones , Ratones Desnudos , Mutación , Fosfatidilinositol 3-Quinasas/metabolismo , Fosforilación , Proteínas Proto-Oncogénicas c-akt/metabolismo , Receptor ErbB-2/genética , Receptor ErbB-3/genética , Transducción de Señal , Ensayos Antitumor por Modelo de Xenoinjerto
15.
Oncotarget ; 6(38): 41123-33, 2015 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-26516700

RESUMEN

The clinical impact of HER2 inhibitors in the treatment of HER2-amplified breast cancers has been largely confined to chemotherapy combination regimens, since HER2 inhibitors appear to have very modest efficacies by themselves. This is due to the resilient nature of the functionally relevant HER2-HER3 tumor driver, bidirectionally linked with downstream PI3K/Akt pathway signaling, which can break through the inhibitory effects of most current HER2 or HER3 targeting therapies. A vertical combination approach targeting HER2 and a downstream pathway is a highly rational strategy for much more effective targeted therapy of this disease. However the importance of these downstream pathways in many human tissues and cells significant limits their usefulness as secondary targets by narrowing the therapeutic index of such combination therapies. The secondary target that can afford the highest potential for clinical translation is the one with the highest synergy against tumor cells in combination with HER2-inhibition, allowing the widest therapeutic index for clinical translation. We conducted a comparative analysis of such secondary targets in combination with the HER2 inhibitor lapatinib and find that the inhibition of mTor affords the highest degree of synergy. In further dissecting the individual roles of TORC1 and TORC2 complexes using pharmacologic and genetic tools, we find that it is specifically the inactivation of TORC2 that most synergistically enhances the efficacy of lapatinib. Although inhibitors that selectively target TORC2 are not currently available, these data make a compelling case for their development.


Asunto(s)
Complejos Multiproteicos/antagonistas & inhibidores , Inhibidores de Proteínas Quinasas/farmacología , Quinazolinas/farmacología , Receptor ErbB-2/antagonistas & inhibidores , Serina-Treonina Quinasas TOR/antagonistas & inhibidores , Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Western Blotting , Neoplasias de la Mama/genética , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Sinergismo Farmacológico , Humanos , Imidazoles/farmacología , Indoles/farmacología , Lapatinib , Diana Mecanicista del Complejo 1 de la Rapamicina , Diana Mecanicista del Complejo 2 de la Rapamicina , Complejos Multiproteicos/genética , Complejos Multiproteicos/metabolismo , Naftiridinas/farmacología , Proteínas Proto-Oncogénicas c-akt/antagonistas & inhibidores , Proteínas Proto-Oncogénicas c-akt/metabolismo , Purinas/farmacología , Quinolinas/farmacología , Interferencia de ARN , Receptor ErbB-2/metabolismo , Receptor ErbB-3/antagonistas & inhibidores , Receptor ErbB-3/metabolismo , Transducción de Señal/efectos de los fármacos , Sirolimus/farmacología , Serina-Treonina Quinasas TOR/genética , Serina-Treonina Quinasas TOR/metabolismo
17.
Mol Biol Cell ; 24(4): 483-94, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23264465

RESUMEN

The endothelium maintains a barrier between blood and tissue that becomes more permeable during inflammation. Membrane rafts are ordered assemblies of cholesterol, glycolipids, and proteins that modulate proinflammatory cell signaling and barrier function. In epithelial cells, the MAL family members MAL, MAL2, and myeloid-associated differentiation marker (MYADM) regulate the function and dynamics of ordered membrane domains. We analyzed the expression of these three proteins in human endothelial cells and found that only MYADM is expressed. MYADM was confined in ordered domains at the plasma membrane, where it partially colocalized with filamentous actin and cell-cell junctions. Small interfering RNA (siRNA)-mediated MYADM knockdown increased permeability, ICAM-1 expression, and leukocyte adhesion, all of which are features of an inflammatory response. Barrier function decrease in MYADM-silenced cells was dependent on ICAM-1 expression. Membrane domains and the underlying actin cytoskeleton can regulate each other and are connected by ezrin, radixin, and moesin (ERM) proteins. In endothelial cells, MYADM knockdown induced ERM activation. Triple-ERM knockdown partially inhibited ICAM-1 increase induced by MYADM siRNA. Importantly, ERM knockdown also reduced ICAM-1 expression in response to the proinflammatory cytokine tumor necrosis factor-α. MYADM therefore regulates the connection between the plasma membrane and the cortical cytoskeleton and so can control the endothelial inflammatory response.


Asunto(s)
Proteínas del Citoesqueleto/genética , Células Endoteliales de la Vena Umbilical Humana/efectos de los fármacos , Molécula 1 de Adhesión Intercelular/genética , Proteínas de la Membrana/genética , Proteínas de Microfilamentos/genética , Proteínas Proteolipídicas Asociadas a Mielina y Linfocito/genética , Animales , Transporte Biológico/efectos de los fármacos , Adhesión Celular/efectos de los fármacos , Proteínas del Citoesqueleto/metabolismo , Perros , Regulación de la Expresión Génica/efectos de los fármacos , Células HeLa , Células Endoteliales de la Vena Umbilical Humana/citología , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Humanos , Inflamación/genética , Inflamación/metabolismo , Molécula 1 de Adhesión Intercelular/metabolismo , Células de Riñón Canino Madin Darby , Microdominios de Membrana/química , Microdominios de Membrana/efectos de los fármacos , Microdominios de Membrana/metabolismo , Proteínas de la Membrana/metabolismo , Proteínas de Microfilamentos/metabolismo , Proteínas Proteolipídicas Asociadas a Mielina y Linfocito/antagonistas & inhibidores , Proteínas Proteolipídicas Asociadas a Mielina y Linfocito/metabolismo , ARN Interferente Pequeño/genética , Transducción de Señal/efectos de los fármacos , Factor de Necrosis Tumoral alfa/farmacología
18.
J Cell Biol ; 198(6): 1025-37, 2012 Sep 17.
Artículo en Inglés | MEDLINE | ID: mdl-22986496

RESUMEN

T cell antigen receptor-proximal signaling components, Rho-family GTPases, and formin proteins DIA1 and FMNL1 have been implicated in centrosome reorientation to the immunological synapse of T lymphocytes. However, the role of these molecules in the reorientation process is not yet defined. Here we find that a subset of microtubules became rapidly stabilized and that their α-tubulin subunit posttranslationally detyrosinated after engagement of the T cell receptor. Formation of stabilized, detyrosinated microtubules required the formin INF2, which was also found to be essential for centrosome reorientation, but it occurred independently of T cell receptor-induced massive tyrosine phosphorylation. The FH2 domain, which was mapped as the INF2 region involved in centrosome repositioning, was able to mediate the formation of stable, detyrosinated microtubules and to restore centrosome translocation in DIA1-, FMNL1-, Rac1-, and Cdc42-deficient cells. Further experiments indicated that microtubule stabilization was required for centrosome polarization. Our work identifies INF2 and stable, detyrosinated microtubules as central players in centrosome reorientation in T cells.


Asunto(s)
Centrosoma/metabolismo , Proteínas de Microfilamentos/metabolismo , Microtúbulos/metabolismo , Linfocitos T/metabolismo , Tirosina/metabolismo , Actinas/genética , Actinas/metabolismo , Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Línea Celular Tumoral , Proteínas del Citoesqueleto/genética , Proteínas del Citoesqueleto/metabolismo , Forminas , Reguladores de Proteínas de Unión al GTP/genética , Reguladores de Proteínas de Unión al GTP/metabolismo , Humanos , Células Jurkat , Proteínas de Microfilamentos/genética , Microtúbulos/genética , Fosforilación , Procesamiento Proteico-Postraduccional/genética , Estructura Terciaria de Proteína , Subunidades de Proteína/genética , Subunidades de Proteína/metabolismo , Transporte de Proteínas/genética , Receptores de Antígenos de Linfocitos T/genética , Receptores de Antígenos de Linfocitos T/metabolismo , Tubulina (Proteína)/genética , Tubulina (Proteína)/metabolismo , Tirosina/genética , Proteína de Unión al GTP rac1/genética , Proteína de Unión al GTP rac1/metabolismo
19.
Mol Biol Cell ; 22(8): 1252-62, 2011 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-21325632

RESUMEN

Membrane organization into condensed domains or rafts provides molecular platforms for selective recruitment of proteins. Cell migration is a general process that requires spatiotemporal targeting of Rac1 to membrane rafts. The protein machinery responsible for making rafts competent to recruit Rac1 remains elusive. Some members of the MAL family of proteins are involved in specialized processes dependent on this type of membrane. Because condensed membrane domains are a general feature of the plasma membrane of all mammalian cells, we hypothesized that MAL family members with ubiquitous expression and plasma membrane distribution could be involved in the organization of membranes for cell migration. We show that myeloid-associated differentiation marker (MYADM), a protein with unique features within the MAL family, colocalizes with Rac1 in membrane protrusions at the cell surface and distributes in condensed membranes. MYADM knockdown (KD) cells had altered membrane condensation and showed deficient incorporation of Rac1 to membrane raft fractions and, similar to Rac1 KD cells, exhibited reduced cell spreading and migration. Results of rescue-of-function experiments by expression of MYADM or active Rac1L61 in cells knocked down for Rac1 or MYADM, respectively, are consistent with the idea that MYADM and Rac1 act on parallel pathways that lead to similar functional outcomes.


Asunto(s)
Antígenos de Diferenciación/metabolismo , Movimiento Celular , Microdominios de Membrana/metabolismo , Proteínas/metabolismo , Proteína de Unión al GTP rac1/metabolismo , Animales , Antígenos de Diferenciación/genética , Adhesión Celular , Línea Celular Tumoral , Membrana Celular/metabolismo , Perros , Electroporación , Femenino , Expresión Génica , Silenciador del Gen , Haplorrinos , Humanos , Masculino , Proteínas Proteolipídicas Asociadas a Mielina y Linfocito , Filogenia , Proteínas/genética , ARN Interferente Pequeño/metabolismo , Transfección , Proteína de Unión al GTP rac1/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA