Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 20
Filtrar
2.
Cell ; 143(2): 201-11, 2010 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-20946980

RESUMEN

Signaling by ErbB receptors requires the activation of their cytoplasmic kinase domains, which is initiated by ligand binding to the receptor ectodomains. Cytoplasmic factors contributing to the activation are unknown. Here we identify members of the cytohesin protein family as such factors. Cytohesin inhibition decreased ErbB receptor autophosphorylation and signaling, whereas cytohesin overexpression stimulated receptor activation. Monitoring epidermal growth factor receptor (EGFR) conformation by anisotropy microscopy together with cell-free reconstitution of cytohesin-dependent receptor autophosphorylation indicate that cytohesins facilitate conformational rearrangements in the intracellular domains of dimerized receptors. Consistent with cytohesins playing a prominent role in ErbB receptor signaling, we found that cytohesin overexpression correlated with EGF signaling pathway activation in human lung adenocarcinomas. Chemical inhibition of cytohesins resulted in reduced proliferation of EGFR-dependent lung cancer cells in vitro and in vivo. Our results establish cytohesins as cytoplasmic conformational activators of ErbB receptors that are of pathophysiological relevance.


Asunto(s)
Adenocarcinoma/patología , Receptores ErbB/metabolismo , Factores de Intercambio de Guanina Nucleótido/metabolismo , Neoplasias Pulmonares/patología , Proteínas Tirosina Quinasas Receptoras/metabolismo , Adenocarcinoma/metabolismo , Animales , Dimerización , Proteínas Activadoras de GTPasa/antagonistas & inhibidores , Proteínas Activadoras de GTPasa/genética , Proteínas Activadoras de GTPasa/metabolismo , Técnicas de Silenciamiento del Gen , Factores de Intercambio de Guanina Nucleótido/antagonistas & inhibidores , Factores de Intercambio de Guanina Nucleótido/genética , Humanos , Neoplasias Pulmonares/metabolismo , Ratones , Trasplante de Neoplasias , Estructura Terciaria de Proteína , Transducción de Señal , Trasplante Heterólogo , Triazoles/farmacología
3.
Neurobiol Dis ; 132: 104543, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31351173

RESUMEN

α-Synuclein (αS) and tau have a lot in common. Dyshomeostasis and aggregation of both proteins are central in the pathogenesis of neurodegenerative diseases: Parkinson's disease, dementia with Lewy bodies, multi-system atrophy and other 'synucleinopathies' in the case of αS; Alzheimer's disease, frontotemporal dementia, progressive supranuclear palsy and other 'tauopathies' in the case of tau. The aggregated states of αS and tau are found to be (hyper)phosphorylated, but the relevance of the phosphorylation in health or disease is not well understood. Both tau and αS are typically characterized as 'intrinsically disordered' proteins, while both engage in transient interactions with cellular components, thereby undergoing structural changes and context-specific folding. αS transiently binds to (synaptic) vesicles forming a membrane-induced amphipathic helix; tau transiently interacts with microtubules forming an 'extended structure'. The regulation and exact nature of the interactions are not fully understood. Here we review recent and previous insights into the dynamic, transient nature of αS and tau with regard to the mode of interaction with their targets, the dwell-time while bound, and the cis and trans factors underlying the frequent switching between bound and unbound states. These aspects are intimately linked to hypotheses on how subtle changes in the transient behaviors may trigger the earliest steps in the pathogenesis of the respective brain diseases. Based on a deeper understanding of transient αS and tau conformations in the cellular context, new therapeutic strategies may emerge, and it may become clearer why existing approaches have failed or how they could be optimized.


Asunto(s)
Enfermedades Neurodegenerativas/genética , Enfermedades Neurodegenerativas/metabolismo , alfa-Sinucleína/genética , alfa-Sinucleína/metabolismo , Proteínas tau/genética , Proteínas tau/metabolismo , Animales , Encéfalo/metabolismo , Humanos , Enfermedades Neurodegenerativas/terapia , Agregación Patológica de Proteínas/genética , Agregación Patológica de Proteínas/metabolismo , Agregación Patológica de Proteínas/terapia , Pliegue de Proteína
4.
Genes Chromosomes Cancer ; 56(6): 460-471, 2017 06.
Artículo en Inglés | MEDLINE | ID: mdl-28177558

RESUMEN

TMEM16A, a Ca2+ -activated Cl- channel, contributes to tumor growth in breast cancer and head and neck squamous cell carcinoma (HNSCC). Here, we investigated whether TMEM16A influences the response to EGFR/HER family-targeting biological therapies. Inhibition of TMEM16A Cl- channel activity in breast cancer cells with HER2 amplification induced a loss of viability. Cells resistant to trastuzumab, a monoclonal antibody targeting HER2, showed an increase in TMEM16A expression and heightened sensitivity to Cl- channel inhibition. Treatment of HNSCC cells with cetuximab, a monoclonal antibody targeting EGFR, and simultaneous TMEM16A suppression led to a pronounced loss of viability. Biochemical analyses of cells subjected to TMEM16A inhibitors or expressing chloride-deficient forms of TMEM16A provide further evidence that TMEM16A channel function may play a role in regulating EGFR/HER2 signaling. These data demonstrate that TMEM16A regulates EGFR and HER2 in growth and survival pathways. Furthermore, in the absence of TMEM16A cotargeting, tumor cells may acquire resistance to EGFR/HER inhibitors. Finally, targeting TMEM16A improves response to biological therapies targeting EGFR/HER family members.


Asunto(s)
Neoplasias de la Mama/tratamiento farmacológico , Carcinoma de Células Escamosas/tratamiento farmacológico , Cetuximab/uso terapéutico , Canales de Cloruro/genética , Receptores ErbB/antagonistas & inhibidores , Neoplasias de Cabeza y Cuello/tratamiento farmacológico , Proteínas de Neoplasias/genética , Receptor ErbB-2/antagonistas & inhibidores , Trastuzumab/uso terapéutico , Animales , Anoctamina-1 , Neoplasias de la Mama/genética , Carcinoma de Células Escamosas/genética , Línea Celular Tumoral , Canales de Cloruro/inmunología , Cromosomas Humanos Par 11 , Femenino , Neoplasias de Cabeza y Cuello/genética , Humanos , Ratones , Ratones Desnudos , Proteínas de Neoplasias/inmunología , Receptor ErbB-2/genética , Receptor ErbB-2/inmunología , Carcinoma de Células Escamosas de Cabeza y Cuello
5.
Adv Exp Med Biol ; 966: 1-14, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28293832

RESUMEN

Multiple studies have described the high expression and amplification of Anoctamin 1 (ANO1) in various cancers, including, but not limited to breast cancer, head and neck cancer, gastrointestinal stromal tumors and glioblastoma. ANO1 has been demonstrated to be critical for tumor growth in breast and head and neck cancers through its regulation of EGFR signaling and pathway modulators like MAPK and protein kinase B. However, the discovery of ANO1 as a calcium activated chloride channel came as a surprise to the field and has given rise to many questions. How does a chloride channel promote oncogenesis? Is the chloride channel function of ANO1 important for its role in cancer? Does ANO1 exhibits chloride-independent functions in cancer cells? This review summarizes the current understanding of ANO1's function in cancer, provides a synopsis of the findings addressing the open questions in the field and gives an outlook on the promising future of ANO1 as a potential therapeutic target for the treatment of various cancers.


Asunto(s)
Anoctamina-1/metabolismo , Proliferación Celular , Proteínas de Neoplasias/metabolismo , Neoplasias/metabolismo , Transducción de Señal , Animales , Anoctamina-1/efectos de los fármacos , Anoctamina-1/genética , Antineoplásicos/uso terapéutico , Proliferación Celular/efectos de los fármacos , Transformación Celular Neoplásica/metabolismo , Transformación Celular Neoplásica/patología , Cloruros/metabolismo , Humanos , Terapia Molecular Dirigida , Proteínas de Neoplasias/efectos de los fármacos , Proteínas de Neoplasias/genética , Neoplasias/tratamiento farmacológico , Neoplasias/genética , Neoplasias/patología , Transducción de Señal/efectos de los fármacos , Carga Tumoral/efectos de los fármacos
6.
J Biol Chem ; 290(2): 889-903, 2015 Jan 09.
Artículo en Inglés | MEDLINE | ID: mdl-25425649

RESUMEN

The calcium-activated chloride channel ANO1 regulates multiple physiological processes. However, little is known about the mechanism of channel gating and regulation of ANO1 activity. Using a high-throughput, random mutagenesis-based variomics screen, we generated and functionally characterized ∼6000 ANO1 mutants and identified novel mutations that affected channel activity, intracellular trafficking, or localization of ANO1. Mutations such as S741T increased ANO1 calcium sensitivity and rendered ANO1 calcium gating voltage-independent, demonstrating a critical role of the re-entrant loop in coupling calcium and voltage sensitivity of ANO1 and hence in regulating ANO1 activation. Our data present the first unbiased and comprehensive study of the structure-function relationship of ANO1. The novel ANO1 mutants reported have diverse functional characteristics, providing new tools to study ANO1 function in biological systems, paving the path for a better understanding of the function of ANO1 and its role in health and diseases.


Asunto(s)
Canales de Cloruro/metabolismo , Canales Iónicos/metabolismo , Proteínas de Neoplasias/química , Proteínas de Neoplasias/metabolismo , Relación Estructura-Actividad , Animales , Anoctamina-1 , Células CHO , Canales de Cloruro/química , Canales de Cloruro/genética , Cricetulus , Células HEK293 , Humanos , Canales Iónicos/química , Canales Iónicos/genética , Mutagénesis Sitio-Dirigida , Proteínas de Neoplasias/genética , Conformación Proteica
7.
Proc Natl Acad Sci U S A ; 110(11): E1026-34, 2013 Mar 12.
Artículo en Inglés | MEDLINE | ID: mdl-23431153

RESUMEN

The calcium-activated chloride channel anoctamin 1 (ANO1) is located within the 11q13 amplicon, one of the most frequently amplified chromosomal regions in human cancer, but its functional role in tumorigenesis has remained unclear. The 11q13 region is amplified in ∼15% of breast cancers. Whether ANO1 is amplified in breast tumors, the extent to which gene amplification contributes to ANO1 overexpression, and whether overexpression of ANO1 is important for tumor maintenance have remained unknown. We have found that ANO1 is amplified and highly expressed in breast cancer cell lines and primary tumors. Amplification of ANO1 correlated with disease grade and poor prognosis. Knockdown of ANO1 in ANO1-amplified breast cancer cell lines and other cancers bearing 11q13 amplification inhibited proliferation, induced apoptosis, and reduced tumor growth in established cancer xenografts. Moreover, ANO1 chloride channel activity was important for cell viability. Mechanistically, ANO1 knockdown or pharmacological inhibition of its chloride-channel activity reduced EGF receptor (EGFR) and calmodulin-dependent protein kinase II (CAMKII) signaling, which subsequently attenuated AKT, v-src sarcoma viral oncogene homolog (SRC), and extracellular signal-regulated kinase (ERK) activation in vitro and in vivo. Our results highlight the involvement of the ANO1 chloride channel in tumor progression and provide insights into oncogenic signaling in human cancers with 11q13 amplification, thereby establishing ANO1 as a promising target for therapy in these highly prevalent tumor types.


Asunto(s)
Neoplasias de la Mama/metabolismo , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/metabolismo , Canales de Cloruro/metabolismo , Cromosomas Humanos Par 11/metabolismo , Amplificación de Genes , Proteínas de Neoplasias/metabolismo , Animales , Anoctamina-1 , Apoptosis/genética , Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/genética , Línea Celular Tumoral , Supervivencia Celular/genética , Canales de Cloruro/genética , Cromosomas Humanos Par 11/genética , Activación Enzimática/genética , Receptores ErbB/genética , Receptores ErbB/metabolismo , Femenino , Técnicas de Silenciamiento del Gen , Humanos , Ratones , Ratones Endogámicos NOD , Ratones SCID , Proteínas de Neoplasias/genética , Trasplante de Neoplasias , Transducción de Señal/genética , Trasplante Heterólogo
8.
J Biol Chem ; 289(16): 11029-11041, 2014 Apr 18.
Artículo en Inglés | MEDLINE | ID: mdl-24599954

RESUMEN

ANO1, a calcium-activated chloride channel, is highly expressed and amplified in human cancers and is a critical survival factor in these cancers. The ANO1 inhibitor CaCCinh-A01 decreases proliferation of ANO1-amplified cell lines; however, the mechanism of action remains elusive. We explored the mechanism behind the inhibitory effect of CaCCinh-A01 on cell proliferation using a combined experimental and in silico approach. We show that inhibition of ANO1 function is not sufficient to diminish proliferation of ANO1-dependent cancer cells. We report that CaCCinh-A01 reduces ANO1 protein levels by facilitating endoplasmic reticulum-associated, proteasomal turnover of ANO1. Washout of CaCCinh-A01 rescued ANO1 protein levels and resumed cell proliferation. Proliferation of newly derived CaCCinh-A01-resistant cell pools was not affected by CaCCinh-A01 as compared with the parental cells. Consistently, CaCCinh-A01 failed to reduce ANO1 protein levels in these cells, whereas ANO1 currents were still inhibited by CaCCinh-A01, indicating that CaCCinh-A01 inhibits cell proliferation by reducing ANO1 protein levels. Furthermore, we employed in silico methods to elucidate novel biological functions of ANO1 inhibitors. Specifically, we derived a pharmacophore model to describe inhibitors capable of promoting ANO1 degradation and report new inhibitors of ANO1-dependent cell proliferation. In summary, our data demonstrate that inhibition of the channel activity of ANO1 is not sufficient to inhibit ANO1-dependent cell proliferation, indicating that the role of ANO1 in cancer only partially depends on its function as a channel. Our results provide an impetus for gaining a deeper understanding of ANO1 modulation in cells and introduce a new targeting approach for antitumor therapy in ANO1-amplified cancers.


Asunto(s)
Antineoplásicos/química , Antineoplásicos/farmacología , Canales de Cloruro/antagonistas & inhibidores , Canales de Cloruro/metabolismo , Proteínas de Neoplasias/antagonistas & inhibidores , Proteínas de Neoplasias/metabolismo , Neoplasias/tratamiento farmacológico , Proteolisis/efectos de los fármacos , Anoctamina-1 , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Canales de Cloruro/genética , Sistemas de Liberación de Medicamentos , Humanos , Proteínas de Neoplasias/genética , Neoplasias/genética , Neoplasias/metabolismo , Neoplasias/patología
9.
J Am Chem Soc ; 133(21): 8372-9, 2011 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-21517092

RESUMEN

A homogeneous fluorescence resonance energy transfer (FRET) system for the real-time monitoring of exchange factor-catalyzed activation of a ras-like small GTPase is described. The underlying design is based on supramolecular template effects exerted by protein-protein interactions between the GTPase adenosine diphosphate ribosylation factor (ARF) and its effector protein GGA3. The GTPase is activated when bound to guanosine triphosphate (GTP) and switched off in its guanosine diphosphate (GDP)-bound state. Both states are accompanied by severe conformational changes that are recognized by GGA3, which only binds the GTPase "on" state. GDP-to-GTP exchange, i.e., GTPase activation, is catalyzed by the guanine nucleotide exchange factor cytohesin-2. When GGA3 and the GTPase ARF1 are labeled with thoroughly selected FRET probes, with simultaneous recording of the fluorescence of an internal tryptophan residue in ARF1, the conformational changes during the activation of the GTPase can be monitored in real time. We applied the FRET system to a multiplex format that allows the simultaneous identification and distinction of small-molecule inhibitors that interfere with the cytohesin-catalyzed ARF1 activation and/or with the interaction between activated ARF1-GTP and GGA3. By screening a library of potential cytohesin inhibitors, predicted by in silico modeling, we identified new inhibitors for the cytohesin-catalyzed GDP/GTP exchange on ARF1 and verified their increased potency in a cell proliferation assay.


Asunto(s)
Factor 1 de Ribosilacion-ADP/química , Proteínas Adaptadoras del Transporte Vesicular/química , GTP Fosfohidrolasas/química , Factor 1 de Ribosilacion-ADP/farmacología , Proteínas Adaptadoras del Transporte Vesicular/farmacología , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Transferencia Resonante de Energía de Fluorescencia , Proteínas Activadoras de GTPasa/química , Factores de Intercambio de Guanina Nucleótido/química , Guanosina Difosfato/metabolismo , Guanosina Trifosfato/metabolismo , Humanos , Unión Proteica , Triptófano/química
10.
Commun Biol ; 4(1): 1085, 2021 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-34526658

RESUMEN

We present two high-throughput compatible methods to detect the interaction of ectopically expressed (RT-Bind) or endogenously tagged (EndoBind) proteins of interest. Both approaches provide temporal evaluation of dimer formation over an extended duration. Using examples of the Nrf2-KEAP1 and the CRAF-KRAS-G12V interaction, we demonstrate that our method allows for the detection of signal for more than 2 days after substrate addition, allowing for continuous monitoring of endogenous protein-protein interactions in real time.


Asunto(s)
Ensayos Analíticos de Alto Rendimiento/métodos , Proteína 1 Asociada A ECH Tipo Kelch/química , Factor 2 Relacionado con NF-E2/química , Proteínas Proto-Oncogénicas p21(ras)/química , Células HEK293 , Humanos , Unión Proteica
11.
Clin Cancer Res ; 26(6): 1432-1448, 2020 03 15.
Artículo en Inglés | MEDLINE | ID: mdl-31699826

RESUMEN

PURPOSE: Despite the therapeutic success of existing HER2-targeted therapies, tumors invariably relapse. This study aimed at identifying new mechanisms responsible for HER2-targeted therapy resistance. EXPERIMENTAL DESIGN: We have used a platform of HER2-targeted therapy-resistant cell lines and primary cultures of healthy and tumor-associated fibroblasts (TAF) to identify new potential targets related to tumor escape from anti-HER2 therapies. RESULTS: We have shown that TAFs promote resistance to HER2-targeted therapies. TAFs produce and secrete high levels of FGF5, which induces FGFR2 activation in the surrounding breast cancer cells. FGFR2 transactivates HER2 via c-Src, leading to resistance to HER2-targeted therapies. In vivo, coinoculating nonresistant cell lines with TAFs results in more aggressive and resistant tumors. Resistant cells activate fibroblasts and secrete FGFR ligands, creating a positive feedback loop that fuels resistance. FGFR2 inhibition not only inhibits HER2 activation, but also induces apoptosis in cells resistant to HER2-targeted therapies. In vivo, inhibitors of FGFR2 reverse resistance and resensitize resistant cells to HER2-targeted therapies. In HER2 patients' samples, α-SMA, FGF5, and FGFR2 contribute to poor outcome and correlate with c-Src activation. Importantly, expression of FGF5 and phospho-HER2 correlated with a reduced pathologic complete response rate in patients with HER2-positive breast cancer treated with neoadjuvant trastuzumab, which highlights the significant role of TAFs/FGF5 in HER2 breast cancer progression and resistance. CONCLUSIONS: We have identified the TAF/FGF5/FGFR2/c-Src/HER2 axis as an escape pathway responsible for HER2-targeted therapy resistance in breast cancer, which can be reversed by FGFR inhibitors.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Neoplasias de la Mama/tratamiento farmacológico , Fibroblastos Asociados al Cáncer/patología , Resistencia a Antineoplásicos , Recurrencia Local de Neoplasia/tratamiento farmacológico , Receptor ErbB-2/antagonistas & inhibidores , Receptor Tipo 2 de Factor de Crecimiento de Fibroblastos/metabolismo , Animales , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Fibroblastos Asociados al Cáncer/metabolismo , Línea Celular Tumoral , Femenino , Humanos , Lapatinib/administración & dosificación , Ratones , Ratones Desnudos , Recurrencia Local de Neoplasia/metabolismo , Recurrencia Local de Neoplasia/patología , Receptor ErbB-2/metabolismo , Transducción de Señal , Tasa de Supervivencia , Trastuzumab/administración & dosificación , Ensayos Antitumor por Modelo de Xenoinjerto
12.
J Vis Exp ; (137)2018 07 10.
Artículo en Inglés | MEDLINE | ID: mdl-30059041

RESUMEN

N-methyl-D-aspartate (NMDA) receptors (NMDAR) are classified as ionotropic glutamate receptors and have critical roles in learning and memory. NMDAR malfunction, expressed as either over- or under-activity caused by mutations, altered expression, trafficking, or localization, can contribute to numerous diseases, especially in the central nervous system. Therefore, understanding the receptor's biology as well as facilitating the discovery of compounds and small molecules is crucial in ongoing efforts to combat neurological diseases. Current approaches to studying the receptor have limitations including low throughput, high cost, and the inability to study its functional abilities due to the necessary presence of channel blockers to prevent NMDAR-mediated excitotoxicity. Additionally, the existing assay systems are sensitive to stimulation by glutamate only and lack sensitivity to stimulation by glycine, the other co-ligand of the NMDAR. Here, we present the first plate-based assay with high-throughput power to study an NMDA receptor with sensitivity to both co-ligands, glutamate and D-serine/glycine. This approach allows the study of different NMDAR subunit compositions and allows functional studies of the receptor in glycine- and/or glutamate-sensitive modes. Additionally, the method does not require the presence of inhibitors during measurements. The effects of positive and negative allosteric modulators can be detected with this assay and the known pharmacology of NMDAR has been replicated in our system. This technique overcomes the limitations of existing methods and is cost-effective. We believe that this novel technique will accelerate the discovery of therapies for NMDAR-mediated pathologies.


Asunto(s)
Calcio/metabolismo , Ácido Glutámico/metabolismo , Glicina/metabolismo , Receptores de N-Metil-D-Aspartato/metabolismo , Serina/metabolismo , Células HEK293 , Humanos
13.
Sci Rep ; 7(1): 11608, 2017 09 14.
Artículo en Inglés | MEDLINE | ID: mdl-28912557

RESUMEN

N-methyl-D-aspartate-receptors (NMDARs) are ionotropic glutamate receptors that function in synaptic transmission, plasticity and cognition. Malfunction of NMDARs has been implicated in a variety of nervous system disorders, making them attractive therapeutic targets. Overexpression of functional NMDAR in non-neuronal cells results in cell death by excitotoxicity, hindering the development of cell-based assays for NMDAR drug discovery. Here we report a plate-based, high-throughput approach to study NMDAR function. Our assay enables the functional study of NMDARs with different subunit composition after activation by glycine/D-serine or glutamate and hence presents the first plate-based, high throughput assay that allows for the measurement of NMDAR function in glycine/D-serine and/or glutamate sensitive modes. This allows to investigate the effect of small molecule modulators on the activation of NMDARs at different concentrations or combinations of the co-ligands. The reported assay system faithfully replicates the pharmacology of the receptor in response to known agonists, antagonists, positive and negative allosteric modulators, as well as the receptor's sensitivity to magnesium and zinc. We believe that the ability to study the biology of NMDARs rapidly and in large scale screens will enable the identification of novel therapeutics whose discovery has otherwise been hindered by the limitations of existing cell based approaches.


Asunto(s)
Calcio/metabolismo , Ácido Glutámico/metabolismo , Glicina/metabolismo , Receptores de N-Metil-D-Aspartato/metabolismo , Serina/metabolismo , Expresión Génica , Células HEK293 , Humanos , Unión Proteica , Multimerización de Proteína , Receptores de N-Metil-D-Aspartato/antagonistas & inhibidores , Receptores de N-Metil-D-Aspartato/química , Receptores de N-Metil-D-Aspartato/genética , Proteínas Recombinantes
14.
PLoS One ; 11(9): e0163129, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27649498

RESUMEN

High throughput random mutagenesis is a powerful tool to identify which residues are important for the function of a protein, and gain insight into its structure-function relation. The human muscle nicotinic acetylcholine receptor was used to test whether this technique previously used for monomeric receptors can be applied to a pentameric ligand-gated ion channel. A mutant library for the α1 subunit of the channel was generated by error-prone PCR, and full length sequences of all 2816 mutants were retrieved using single molecule real time sequencing. Each α1 mutant was co-transfected with wildtype ß1, δ, and ε subunits, and the channel function characterized by an ion flux assay. To test whether the strategy could map the structure-function relation of this receptor, we attempted to identify mutations that conferred resistance to competitive antagonists. Mutant hits were defined as receptors that responded to the nicotinic agonist epibatidine, but were not inhibited by either α-bungarotoxin or tubocurarine. Eight α1 subunit mutant hits were identified, six of which contained mutations at position Y233 or V275 in the transmembrane domain. Three single point mutations (Y233N, Y233H, and V275M) were studied further, and found to enhance the potencies of five channel agonists tested. This suggests that the mutations made the channel resistant to the antagonists, not by impairing antagonist binding, but rather by producing a gain-of-function phenotype, e.g. increased agonist sensitivity. Our data show that random high throughput mutagenesis is applicable to multimeric proteins to discover novel functional mutants, and outlines the benefits of using single molecule real time sequencing with regards to quality control of the mutant library as well as downstream mutant data interpretation.


Asunto(s)
Secuenciación de Nucleótidos de Alto Rendimiento/métodos , Músculos/metabolismo , Mutagénesis , Receptores Nicotínicos/genética , Secuencia de Aminoácidos , Compuestos Bicíclicos Heterocíclicos con Puentes/farmacología , Bungarotoxinas/farmacología , Calcio/metabolismo , Células HEK293 , Humanos , Transporte Iónico/efectos de los fármacos , Mutación , Agonistas Nicotínicos/farmacología , Antagonistas Nicotínicos/farmacología , Piridinas/farmacología , Receptores Nicotínicos/metabolismo , Homología de Secuencia de Aminoácido , Tubocurarina/farmacología
15.
Oncotarget ; 6(11): 9173-88, 2015 Apr 20.
Artículo en Inglés | MEDLINE | ID: mdl-25823819

RESUMEN

The epidermal growth factor receptor (EGFR) contributes to the pathogenesis of head&neck squamous cell carcinoma (HNSCC). However, only a subset of HNSCC patients benefit from anti-EGFR targeted therapy. By performing an unbiased proteomics screen, we found that the calcium-activated chloride channel ANO1 interacts with EGFR and facilitates EGFR-signaling in HNSCC. Using structural mutants of EGFR and ANO1 we identified the trans/juxtamembrane domain of EGFR to be critical for the interaction with ANO1. Our results show that ANO1 and EGFR form a functional complex that jointly regulates HNSCC cell proliferation. Expression of ANO1 affected EGFR stability, while EGFR-signaling elevated ANO1 protein levels, establishing a functional and regulatory link between ANO1 and EGFR. Co-inhibition of EGFR and ANO1 had an additive effect on HNSCC cell proliferation, suggesting that co-targeting of ANO1 and EGFR could enhance the clinical potential of EGFR-targeted therapy in HNSCC and might circumvent the development of resistance to single agent therapy. HNSCC cell lines with amplification and high expression of ANO1 showed enhanced sensitivity to Gefitinib, suggesting ANO1 overexpression as a predictive marker for the response to EGFR-targeting agents in HNSCC therapy. Taken together, our results introduce ANO1 as a promising target and/or biomarker for EGFR-directed therapy in HNSCC.


Asunto(s)
Carcinoma de Células Escamosas/patología , Canales de Cloruro/metabolismo , Receptores ErbB/metabolismo , Neoplasias de Cabeza y Cuello/patología , Proteínas de Neoplasias/metabolismo , Inhibidores de Proteínas Quinasas/farmacología , Quinazolinas/farmacología , Anoctamina-1 , Línea Celular Tumoral , Proliferación Celular , Canales de Cloruro/genética , Receptores ErbB/antagonistas & inhibidores , Receptores ErbB/genética , Cadena Pesada de la Proteína-1 Reguladora de Fusión/genética , Gefitinib , Células HEK293 , Humanos , Complejos Multiproteicos/metabolismo , Mutación/genética , Proteínas de Neoplasias/genética , Estructura Terciaria de Proteína/genética , Interferencia de ARN , ARN Interferente Pequeño , Transducción de Señal , Carcinoma de Células Escamosas de Cabeza y Cuello
16.
PLoS One ; 9(6): e97973, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24886841

RESUMEN

The human prostacyclin receptor (hIP receptor) is a seven-transmembrane G protein-coupled receptor (GPCR) that plays a critical role in vascular smooth muscle relaxation and platelet aggregation. hIP receptor dysfunction has been implicated in numerous cardiovascular abnormalities, including myocardial infarction, hypertension, thrombosis and atherosclerosis. Genomic sequencing has discovered several genetic variations in the PTGIR gene coding for hIP receptor, however, its structure-function relationship has not been sufficiently explored. Here we set out to investigate the applicability of high throughput random mutagenesis to study the structure-function relationship of hIP receptor. While chemical mutagenesis was not suitable to generate a mutagenesis library with sufficient coverage, our data demonstrate error-prone PCR (epPCR) mediated mutagenesis as a valuable method for the unbiased screening of residues regulating hIP receptor function and expression. Here we describe the generation and functional characterization of an epPCR derived mutagenesis library compromising >4000 mutants of the hIP receptor. We introduce next generation sequencing as a useful tool to validate the quality of mutagenesis libraries by providing information about the coverage, mutation rate and mutational bias. We identified 18 mutants of the hIP receptor that were expressed at the cell surface, but demonstrated impaired receptor function. A total of 38 non-synonymous mutations were identified within the coding region of the hIP receptor, mapping to 36 distinct residues, including several mutations previously reported to affect the signaling of the hIP receptor. Thus, our data demonstrates epPCR mediated random mutagenesis as a valuable and practical method to study the structure-function relationship of GPCRs.


Asunto(s)
Aminoácidos/genética , Secuenciación de Nucleótidos de Alto Rendimiento , Mutagénesis/genética , Receptores de Prostaglandina/genética , Simulación por Computador , Células HEK293 , Humanos , Hidroxilamina , Mutación/genética , Tasa de Mutación , Reacción en Cadena de la Polimerasa , Receptores de Epoprostenol
17.
Clin Cancer Res ; 20(17): 4673-88, 2014 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-24919570

RESUMEN

PURPOSE: Tumor metastasis is the leading cause of death in patients with cancer. However, the mechanisms that underlie metastatic progression remain unclear. We examined TMEM16A (ANO1) expression as a key factor shifting tumors between growth and metastasis. EXPERIMENTAL DESIGN: We evaluated 26 pairs of primary and metastatic lymph node (LN) tissue from patients with squamous cell carcinoma of the head and neck (SCCHN) for differential expression of TMEM16A. In addition, we identified mechanisms by which TMEM16A expression influences tumor cell motility via proteomic screens of cell lines and in vivo mouse studies of metastasis. RESULTS: Compared with primary tumors, TMEM16A expression decreases in metastatic LNs of patients with SCCHN. Stable reduction of TMEM16A expression enhances cell motility and increases metastases while decreasing tumor proliferation in an orthotopic mouse model. Evaluation of human tumor tissues suggests an epigenetic mechanism for decreasing TMEM16A expression through promoter methylation that correlated with a transition between an epithelial and a mesenchymal phenotype. These effects of TMEM16A expression on tumor cell size and epithelial-to-mesenchymal transition (EMT) required the amino acid residue serine 970 (S970); however, mutation of S970 to alanine does not disrupt the proliferative advantages of TMEM16A overexpression. Furthermore, S970 mediates the association of TMEM16A with Radixin, an actin-scaffolding protein implicated in EMT. CONCLUSIONS: Together, our results identify TMEM16A, an eight transmembrane domain Ca2+-activated Cl- channel, as a primary driver of the "Grow" or "Go" model for cancer progression, in which TMEM16A expression acts to balance tumor proliferation and metastasis via its promoter methylation.


Asunto(s)
Carcinogénesis/genética , Carcinoma de Células Escamosas/genética , Proliferación Celular/genética , Canales de Cloruro/biosíntesis , Transición Epitelial-Mesenquimal/genética , Neoplasias de Cabeza y Cuello/genética , Proteínas de Neoplasias/biosíntesis , Animales , Anoctamina-1 , Carcinoma de Células Escamosas/patología , Línea Celular Tumoral , Movimiento Celular/genética , Canales de Cloruro/genética , Proteínas del Citoesqueleto/genética , Metilación de ADN/genética , Regulación Neoplásica de la Expresión Génica , Neoplasias de Cabeza y Cuello/patología , Humanos , Metástasis Linfática/genética , Proteínas de la Membrana/genética , Ratones , Proteínas de Neoplasias/genética , Carcinoma de Células Escamosas de Cabeza y Cuello , Ensayos Antitumor por Modelo de Xenoinjerto
18.
PLoS One ; 7(7): e41179, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22815959

RESUMEN

Epidermal growth factor receptor (EGFR) tyrosine kinase inhibitors (TKI), such as gefitinib, have been proven to efficiently inhibit the proliferation of a subset of non small-cell lung cancers (NSCLC). Unfortunately, the majority of NSCLC expressing wild type EGFR is primarily resistant to EGFR-TKI treatment. Here, we show that the proliferation of the gefitinib-resistant NSCLC cell lines H460 and A549 is reduced by the small molecule SecinH3 which indirectly attenuates EGFR activation by inhibition of cytohesins, a class of recently discovered cytoplasmic EGFR activators. SecinH3 and gefitinib showed a synergistic antiproliferative effect, which correlated with a profound inhibition of Akt activation and survivin expression. Treating mice bearing H460 xenografts with SecinH3 showed the antiproliferative and pro-apoptotic effect of SecinH3 in vivo. Our data suggest that targeting the EGFR indirectly by inhibiting its cytoplasmic activators, the cytohesins, has the potential to improve the treatment of primarily EGFR-TKI resistant lung cancers.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Resistencia a Antineoplásicos , Factores de Intercambio de Guanina Nucleótido/antagonistas & inhibidores , Neoplasias Pulmonares/tratamiento farmacológico , Quinazolinas/farmacología , Animales , Línea Celular Tumoral , Proliferación Celular , Citoplasma/metabolismo , Ensayos de Selección de Medicamentos Antitumorales/métodos , Receptores ErbB/metabolismo , Gefitinib , Regulación Neoplásica de la Expresión Génica , Humanos , Masculino , Ratones , Ratones Desnudos , Trasplante de Neoplasias , Receptor IGF Tipo 1/metabolismo , Transducción de Señal , Triazoles/farmacología
19.
ACS Chem Biol ; 5(9): 839-49, 2010 Sep 17.
Artículo en Inglés | MEDLINE | ID: mdl-20614894

RESUMEN

Virtual screening (VS) of chemical libraries formatted in silico provides an alternative to experimental high-throughput screening (HTS) for the identification of small molecule modulators of protein function. We have tailored a VS approach combining fingerprint similarity searching and support vector machine modeling toward the identification of small molecular probes for the study of cytohesins, a family of cytoplasmic regulator proteins with multiple cellular functions. A total of 40 new structurally diverse inhibitors were identified, and 26 of these compounds were more active than the primary VS template, a single known inhibitory chemotype, in at least one of three different assays (guanine nucleotide exchange, Drosophila insulin signaling, and human leukocyte cell adhesion). Moreover, these inhibitors displayed differential inhibitory profiles. Our findings demonstrate that, at least for the cytohesins, computational extrapolation from known active compounds was capable of identifying small molecular probes with highly diversified functional profiles.


Asunto(s)
Diseño de Fármacos , Factores de Intercambio de Guanina Nucleótido/antagonistas & inhibidores , Factores de Intercambio de Guanina Nucleótido/metabolismo , Bibliotecas de Moléculas Pequeñas/química , Bibliotecas de Moléculas Pequeñas/farmacología , Animales , Inteligencia Artificial , Adhesión Celular/efectos de los fármacos , Línea Celular , Drosophila/efectos de los fármacos , Drosophila/metabolismo , Proteínas de Drosophila/antagonistas & inhibidores , Proteínas de Drosophila/metabolismo , Humanos , Insulina/metabolismo , Leucocitos/citología , Leucocitos/efectos de los fármacos , Transducción de Señal/efectos de los fármacos
20.
RNA ; 12(9): 1650-60, 2006 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-16888322

RESUMEN

The beta-amyloid peptide (Abeta) is a major component of the Alzheimer's disease (AD)-associated senile plaques and is generated by sequential cleavage of the beta-amyloid precursor protein (APP) by beta-secretase and gamma-secretase. Since BACE1 initiates Abeta generation it represents a valuable target to interfere with Abeta production and treatment of AD. While the enzymatic activity of BACE1 resides in the extracellular domain, the protein also contains a short cytoplasmic tail (B1-CT). This domain serves as a binding site for at least two proteins, the copper chaperone for superoxide dismutase-1 (CCS), and the Golgi-localized, gamma-ear-containing, ADP ribosylation factor-binding (GGA1) protein, and contains a single phosphorylation site. However, the precise role of the B1-CT for the overall biological function of this protein is largely unknown. Functional studies focusing on the activity of this domain would strongly benefit from the availability of domain-specific inhibitors. Here we describe the isolation and characterization of RNA aptamers that selectively target the B1-CT. We show that these RNAs bind to authentic BACE1 and provide evidence that the binding site is restricted to the membrane-proximal half of the C terminus. Aptamer-binding specifically interferes with the recruitment of CCS, but still permits GGA1 association and casein kinase-dependent phosphorylation, consistent with selective binding site targeting within this short peptide. Because phosphorylation and GGA1 binding to B1-CT regulate BACE1 transport, these RNA inhibitors could be applied to investigate B1-CT activity without affecting the subcellular localization of BACE1.


Asunto(s)
Aptámeros de Nucleótidos/metabolismo , Citoplasma/química , Citoplasma/enzimología , Endopeptidasas/metabolismo , Proteínas/metabolismo , Factores de Ribosilacion-ADP/metabolismo , Proteínas Adaptadoras del Transporte Vesicular/metabolismo , Enfermedad de Alzheimer/enzimología , Enfermedad de Alzheimer/genética , Secuencias de Aminoácidos , Secuencia de Aminoácidos , Secretasas de la Proteína Precursora del Amiloide , Aptámeros de Nucleótidos/química , Aptámeros de Nucleótidos/genética , Aptámeros de Nucleótidos/farmacología , Ácido Aspártico Endopeptidasas , Secuencia de Bases , Sitios de Unión , Membrana Celular/enzimología , Membrana Celular/metabolismo , Endopeptidasas/química , Endopeptidasas/genética , Glutatión Transferasa/metabolismo , Técnicas In Vitro , Chaperonas Moleculares/metabolismo , Datos de Secuencia Molecular , Mutación , Conformación de Ácido Nucleico , Fosforilación , Estructura Terciaria de Proteína , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/metabolismo , Técnica SELEX de Producción de Aptámeros
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA