Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 33
Filtrar
1.
Biochem Biophys Res Commun ; 704: 149673, 2024 Apr 16.
Artículo en Inglés | MEDLINE | ID: mdl-38401305

RESUMEN

Epidermal growth factor receptor (EGFR)-mediated signal transduction controls cell growth and proliferation. The signaling pathway is regulated so that it is activated only by external EGF stimuli, but the mechanisms that prevent EGF-independent spontaneous activation of EGFR-mediated signaling are unknown. Here we report cholesterol depletion activates EGFR-mediated signaling without EGF. We applied automated single-molecule imaging to EGFR and characterized the lateral diffusion and cluster formation on cholesterol-depleted and cholesterol-supplemented membranes. In cells in which cholesterol was depleted by methyl-ß-cyclodextrin (MßCD) treatment, EGFR exhibited a reduction in lateral diffusion, an acceleration of cluster formation, and autophosphorylation without EGF. Concurrently, extracellular signal-regulated kinase (ERK), which is regulated by EGFR-mediated signaling, exhibited phosphorylation and nuclear translocation without EGF. These cholesterol depletion-induced changes were similar, albeit less efficient, to those that occurred with EGF stimulation in normal cells without MßCD, indicating the spontaneous activation of EGFR signaling. The exogenous supplementation of cholesterol suppressed the MßCD-induced spontaneous activation of EGFR and ERK nuclear translocation. Single-molecule imaging of EGFR in a large number of cells revealed cell-to-cell heterogeneity, with a sub-population showing a high ability for spontaneous activation. These results provide evidence that EGFR-mediated signaling is properly regulated by cholesterol metabolism to prevent uncontrolled spontaneous activation.


Asunto(s)
Factor de Crecimiento Epidérmico , Transducción de Señal , Factor de Crecimiento Epidérmico/farmacología , Factor de Crecimiento Epidérmico/metabolismo , Receptores ErbB/metabolismo , Fosforilación , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Colesterol/metabolismo
2.
Cell Struct Funct ; 48(2): 145-160, 2023 Aug 31.
Artículo en Inglés | MEDLINE | ID: mdl-37438131

RESUMEN

In eukaryotic motile cells, the active Ras (Ras-GTP)-enriched domain is generated in an asymmetric manner on the cell membrane through the excitable dynamics of an intracellular signaling network. This asymmetric Ras signaling regulates pseudopod formation for both spontaneous random migration and chemoattractant-induced directional migration. While membrane lipids, such as sphingomyelin and phosphatidylserine, contribute to Ras signaling in various cell types, whether they are involved in the Ras excitability for cell motility is unknown. Here we report that functional Ras excitability requires the normal metabolism of sphingomyelin for efficient cell motility and chemotaxis. The pharmacological blockade of sphingomyelin metabolism by an acid-sphingomyelinase inhibitor, fendiline, and other inhibitors suppressed the excitable generation of the stable Ras-GTP-enriched domain. The suppressed excitability failed to invoke enough basal motility to achieve directed migration under shallow chemoattractant gradients. The fendiline-induced defects in Ras excitability, motility and stimulation-elicited directionality were due to an accumulation of sphingomyelin on the membrane, which could be recovered by exogenous sphingomyelinase or phosphatidylserine without changing the expression of Ras. These results indicate a novel regulatory mechanism of the excitable system by membrane lipids, in which sphingomyelin metabolism provides a membrane environment to ensure Ras excitation for efficient cellular motility and chemotaxis.Key words: cell polarity, cell migration, Ras, excitability, sphingomyelin.


Asunto(s)
Quimiotaxis , Esfingomielinas , Quimiotaxis/fisiología , Esfingomielina Fosfodiesterasa/metabolismo , Fosfatidilserinas , Fendilina , Movimiento Celular , Factores Quimiotácticos , Guanosina Trifosfato
3.
J Biol Chem ; 298(3): 101630, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-35085554

RESUMEN

Cancer invasion and metastasis are the major causes of cancer patient mortality. Various growth factors, including hepatocyte growth factor (HGF), are known to promote cancer invasion and metastasis, but the regulatory mechanisms involved are not fully understood. Here, we show that HGF-promoted migration and invasion of breast cancer cells are regulated by CUB domain-containing protein 1 (CDCP1), a transmembrane activator of SRC kinase. In metastatic human breast cancer cell line MDA-MB-231, which highly expresses the HGF receptor MET and CDCP1, we show that CDCP1 knockdown attenuated HGF-induced MET activation, followed by suppression of lamellipodia formation and cell migration/invasion. In contrast, in the low invasive/nonmetastatic breast cancer cell line T47D, which had no detectable MET and CDCP1 expression, ectopic MET expression stimulated the HGF-dependent activation of invasive activity, and concomitant CDCP1 expression activated SRC and further promoted invasive activity. In these cells, CDCP1 expression dramatically activated HGF-induced membrane remodeling, which was accompanied by activation of the small GTPase Rac1. Analysis of guanine nucleotide exchange factors revealed that ARHGEF7 was specifically required for CDCP1-dependent induction of HGF-induced invasive ability. Furthermore, immunofluorescence staining demonstrated that CDCP1 coaccumulated with ARHGEF7. Finally, we confirmed that the CDCP1-SRC axis was also crucial for HGF and ARHGEF7-RAC1 signaling in MDA-MB-231 cells. Altogether, these results demonstrate that the CDCP1-SRC-ARHGEF7-RAC1 pathway plays an important role in the HGF-induced invasion of a subset of breast cancer cells.


Asunto(s)
Antígenos de Neoplasias , Neoplasias de la Mama , Factor de Crecimiento de Hepatocito , Factores de Intercambio de Guanina Nucleótido Rho , Familia-src Quinasas , Antígenos de Neoplasias/genética , Antígenos de Neoplasias/metabolismo , Neoplasias de la Mama/genética , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Moléculas de Adhesión Celular , Línea Celular Tumoral , Movimiento Celular/fisiología , Femenino , Factor de Crecimiento de Hepatocito/metabolismo , Humanos , Invasividad Neoplásica , Proteínas Proto-Oncogénicas c-met/genética , Proteínas Proto-Oncogénicas c-met/metabolismo , Factores de Intercambio de Guanina Nucleótido Rho/genética , Factores de Intercambio de Guanina Nucleótido Rho/metabolismo , Transducción de Señal , Familia-src Quinasas/genética , Familia-src Quinasas/metabolismo
4.
Nat Commun ; 13(1): 487, 2022 01 25.
Artículo en Inglés | MEDLINE | ID: mdl-35078997

RESUMEN

Signaling-biased ligands acting on G-protein-coupled receptors (GPCRs) differentially activate heterotrimeric G proteins and ß-arrestins. Although a wealth of structural knowledge about signaling bias at the GPCR level exists (preferential engagement of a specific transducer), little is known about the bias at the transducer level (different functions mediated by a single transducer), partly due to a poor understanding of GPCR kinase (GRK)-mediated GPCR phosphorylation. Here, we reveal a unique role of the Gq heterotrimer as a determinant for GRK-subtype selectivity that regulates subsequent ß-arrestin conformation and function. Using the angiotensin II (Ang II) type-1 receptor (AT1R), we show that ß-arrestin recruitment depends on both GRK2/3 and GRK5/6 upon binding of Ang II, but solely on GRK5/6 upon binding of the ß-arrestin-biased ligand TRV027. With pharmacological inhibition or genetic loss of Gq, GRK-subtype selectivity and ß-arrestin functionality by Ang II is shifted to those of TRV027. Single-molecule imaging identifies relocation of AT1R and GRK5, but not GRK2, to an immobile phase under the Gq-inactive, AT1R-stimulated conditions. These findings uncover a previously unappreciated Gq-regulated mechanism that encodes GRK-subtype selectivity and imparts distinct phosphorylation-barcodes directing downstream ß-arrestin functions.


Asunto(s)
Angiotensina II/farmacología , Quinasa 2 del Receptor Acoplado a Proteína-G/metabolismo , Quinasa 5 del Receptor Acoplado a Proteína-G/metabolismo , Subunidades alfa de la Proteína de Unión al GTP Gq-G11/metabolismo , Oligopéptidos/farmacología , Receptor de Angiotensina Tipo 1/metabolismo , beta-Arrestinas/metabolismo , Línea Celular , Humanos , Fosforilación , Transducción de Señal , Vasoconstrictores/farmacología
5.
Biophys J ; 121(3): 470-480, 2022 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-34958777

RESUMEN

Intercellular signals induce various cellular responses, including growth, proliferation, and differentiation, via the dynamic processes of signal transduction pathways. For cell fate decisions, ligand-binding induces the phosphorylation of ERBB receptors, which in turn activate downstream molecules. The ERBB family includes four subtypes, which diverged through two gene duplications from a common ancestor. Differences in the expression patterns of the subtypes have been reported between different organs in the human body. However, how these different expression properties influence the diverse phosphorylation levels of ERBB proteins is not well understood. Here we study the origin of the phosphorylation responses by experimental and mathematical analyses. The experimental measurements clarified that the phosphorylation levels heavily depend on the ERBB expression profiles. We developed a mathematical model consisting of the four subtypes as monomers, homodimers, and heterodimers and estimated the rate constants governing the phosphorylation responses from the experimental data. To understand the origin of the diversity, we analyzed the effects of the expression levels and reaction rates of the ERBB subtypes on the diversity. The difference in phosphorylation rates between ERBB subtypes showed a much greater contribution to the diversity than did the dimerization rates. This result implies that divergent evolution in phosphorylation reactions rather than in dimerization reactions after whole genome duplications was essential for increasing the diversity of the phosphorylation responses.


Asunto(s)
Receptor ErbB-2 , Transducción de Señal , Humanos , Fosforilación , Receptor ErbB-2/metabolismo
6.
Int J Mol Sci ; 22(16)2021 Aug 06.
Artículo en Inglés | MEDLINE | ID: mdl-34445178

RESUMEN

TRPV1 and TRPV4, members of the transient receptor potential vanilloid family, are multimodal ion channels activated by various stimuli, including temperature and chemicals. It has been demonstrated that TRPV channels function as tetramers; however, the dynamics of the diffusion, oligomerization, and endocytosis of these channels in living cells are unclear. Here we undertook single-molecule time-lapse imaging of TRPV1 and TRPV4 in HEK 293 cells. Differences were observed between TRPV1 and TRPV4 before and after agonist stimulation. In the resting state, TRPV4 was more likely to form higher-order oligomers within immobile membrane domains than TRPV1. TRPV1 became immobile after capsaicin stimulation, followed by its gradual endocytosis. In contrast, TRPV4 was rapidly internalized upon stimulation with GSK1016790A. The selective loss of immobile higher-order oligomers from the cell surface through endocytosis increased the proportion of the fast-diffusing state for both subtypes. With the increase in the fast state, the association rate constants of TRPV1 and TRPV4 increased, regenerating the higher-order oligomers. Our results provide a possible mechanism for the different rates of endocytosis of TRPV1 and TRPV4 based on the spatial organization of the higher-order structures of the two TRPV channels.


Asunto(s)
Canales Catiónicos TRPV/metabolismo , Difusión , Endocitosis , Células HEK293 , Humanos , Simulación de Dinámica Molecular , Imagen Individual de Molécula , Canales Catiónicos TRPV/análisis
7.
Angew Chem Int Ed Engl ; 60(42): 22745-22752, 2021 10 11.
Artículo en Inglés | MEDLINE | ID: mdl-34142433

RESUMEN

Designing synthetic surrogates of functional proteins is an important, albeit challenging, task in the field of chemistry. A strategy toward the design of synthetic agonists for growth factor or cytokine receptors that elicit a desired signal activity has been in high demand, as such ligands hold great promise as safer and more effective therapeutics. In the present study, we used a DNA aptamer as a building block and described the strategy-guided design of a synthetic receptor agonist with fine-tuned agonism. The developed synthetic partial agonist can regulate therapeutically relevant cellular activities by eliciting fine-tuned receptor signaling.


Asunto(s)
Aptámeros de Nucleótidos/metabolismo , Péptidos y Proteínas de Señalización Intercelular/agonistas , Receptores de Citocinas/agonistas , Células A549 , Aptámeros de Nucleótidos/química , Aptámeros de Nucleótidos/farmacología , Movimiento Celular/efectos de los fármacos , Dimerización , Factor de Crecimiento de Hepatocito/agonistas , Factor de Crecimiento de Hepatocito/metabolismo , Células Endoteliales de la Vena Umbilical Humana , Humanos , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Ligandos , Microscopía Fluorescente , Unión Proteica , Proteínas Proto-Oncogénicas c-met/agonistas , Proteínas Proto-Oncogénicas c-met/genética , Proteínas Proto-Oncogénicas c-met/metabolismo , Receptores de Citocinas/metabolismo , Transducción de Señal/efectos de los fármacos
8.
Adv Exp Med Biol ; 1310: 59-80, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33834432

RESUMEN

Cellular signaling is regulated by the spatiotemporal dynamics and kinetics of molecular behavior. To investigate the mechanisms at the molecular level, fluorescence single-molecule analysis is an effective method owing to the direct observation of individual molecules in situ in cells and the results in quantitative information about the behavior. The integration of machine learning into this analysis modality enables the acquisition of behavioral features at all time points of all molecules. As a case study, we described a hidden Markov model-based approach to infer the molecular states of mobility and clustering for epidermal growth factor receptor (EGFR) along a single-molecule trajectory. We reveal a scheme of the receptor signaling through the dynamic coupling of the mobility and clustering states under the influence of a local membrane structure. As the activation process progressed, EGFR generally converged to an immobile cluster. This state exhibited high affinity with a specific cytoplasmic protein, shown by two-color single-molecule analysis, and could be a platform for downstream signaling. The method was effective for elucidating the biophysical mechanisms of signaling regulation when comprehensive analysis is possible for a huge number and multiple molecular species in the signaling pathway. Thus, a fully automated system for single-molecule analysis, in which indispensable expertise was replicated using artificial intelligence, has been developed to enable in-cell large-scale analysis. This system opens new single-molecule approaches for pharmacological applications as well as the basic sciences.


Asunto(s)
Inteligencia Artificial , Imagen Individual de Molécula , Membrana Celular , Cinética , Transducción de Señal
9.
Biosystems ; 199: 104293, 2021 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-33221378

RESUMEN

Growth factors regulate cell fates, including their proliferation, differentiation, survival, and death, according to the cell type. Even when the response to a specific growth factor is deterministic for collective cell behavior, significant levels of fluctuation are often observed between single cells. Statistical analyses of single-cell responses provide insights into the mechanism of cell fate decisions but very little is known about the distributions of the internal states of cells responding to growth factors. Using multi-color immunofluorescent staining, we have here detected the phosphorylation of seven elements in the early response of the ERBB-RAS-MAPK system to two growth factors. Among these seven elements, five were analyzed simultaneously in distinct combinations in the same single cells. Although principle component analysis suggested cell-type and input specific phosphorylation patterns, cell-to-cell fluctuation was large. Mutual information analysis suggested that each cell type uses multitrack (bush-like) signal transduction pathways under conditions in which clear fate changes have been reported. The clustering of single-cell response patterns indicated that the fate change in a cell population correlates with the large entropy of the response, suggesting a bet-hedging strategy is used in decision making. A comparison of true and randomized datasets further indicated that this large variation is not produced by simple reaction noise, but is defined by the properties of the signal-processing network.


Asunto(s)
Péptidos y Proteínas de Señalización Intercelular/farmacología , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Transducción de Señal/efectos de los fármacos , Proteínas ras/metabolismo , Diferenciación Celular/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Receptores ErbB/metabolismo , Células HeLa , Humanos , Células MCF-7 , Microscopía Fluorescente , Modelos Biológicos , Fosforilación/efectos de los fármacos , Análisis de Componente Principal , Análisis de la Célula Individual/métodos
10.
Microscopy (Oxf) ; 69(2): 69-78, 2020 Apr 08.
Artículo en Inglés | MEDLINE | ID: mdl-32090254

RESUMEN

Single-molecule imaging analysis has been applied to study the dynamics and kinetics of molecular behaviors and interactions in living cells. In spite of its high potential as a technique to investigate the molecular mechanisms of cellular phenomena, single-molecule imaging analysis has not been extended to a large scale of molecules in cells due to the low measurement throughput as well as required expertise. To overcome these problems, we have automated the imaging processes by using computer operations, robotics and artificial intelligence (AI). AI is an ideal substitute for expertise to obtain high-quality images for quantitative analysis. Our automated in-cell single-molecule imaging system, AiSIS, could analyze 1600 cells in 1 day, which corresponds to ∼ 100-fold higher efficiency than manual analysis. The large-scale analysis revealed cell-to-cell heterogeneity in the molecular behavior, which had not been recognized in previous studies. An analysis of the receptor behavior and downstream signaling was accomplished within a significantly reduced time frame and revealed the detailed activation scheme of signal transduction, advancing cell biology research. Furthermore, by combining the high-throughput analysis with our previous finding that a receptor changes its behavioral dynamics depending on the presence of a ligand/agonist or inhibitor/antagonist, we show that AiSIS is applicable to comprehensive pharmacological analysis such as drug screening. This AI-aided automation has wide applications for single-molecule analysis.


Asunto(s)
Inteligencia Artificial , Imagen Individual de Molécula/métodos , Automatización , Evaluación Preclínica de Medicamentos/métodos , Citometría de Flujo/métodos , Farmacología/métodos
11.
Sci Signal ; 11(548)2018 09 18.
Artículo en Inglés | MEDLINE | ID: mdl-30228224

RESUMEN

G protein-coupled receptors (GPCRs) are major drug targets. Developing a method to measure the activities of GPCRs is essential for pharmacology and drug screening. However, it is difficult to measure the effects of a drug by monitoring the receptor on the cell surface; thus, changes in the concentrations of downstream signaling molecules, which depend on the signaling pathway selectivity of the receptor, are often used as an index of receptor activity. We show that single-molecule imaging analysis provides an alternative method for assessing the effects of ligands on GPCRs. Using total internal reflection fluorescence microscopy (TIRFM), we monitored the dynamics of the diffusion of metabotropic glutamate receptor 3 (mGluR3), a class C GPCR, under various ligand conditions. Our single-molecule tracking analysis demonstrated that increases and decreases in the average diffusion coefficient of mGluR3 quantitatively reflected the ligand-dependent inactivation and activation of receptors, respectively. Through experiments with inhibitors and dual-color single-molecule imaging analysis, we found that the diffusion of receptor molecules was altered by common physiological events associated with GPCRs, including G protein binding, and receptor accumulation in clathrin-coated pits. We also confirmed that agonist also decreased the average diffusion coefficient for class A and B GPCRs, demonstrating that this parameter is a good index for estimating ligand effects on many GPCRs regardless of their phylogenetic groups, the chemical properties of the ligands, or G protein-coupling selectivity.


Asunto(s)
Proteínas de Unión al GTP/metabolismo , Microscopía Fluorescente/métodos , Receptores Acoplados a Proteínas G/metabolismo , Transducción de Señal , Aminoácidos/metabolismo , Células HEK293 , Humanos , Ligandos , Toxina del Pertussis/metabolismo , Toxina del Pertussis/farmacología , Unión Proteica/efectos de los fármacos , Ensayo de Unión Radioligante/métodos , Receptores Acoplados a Proteínas G/análisis , Receptores Acoplados a Proteínas G/genética , Receptores de Glutamato Metabotrópico/análisis , Receptores de Glutamato Metabotrópico/genética , Receptores de Glutamato Metabotrópico/metabolismo , Xantenos/metabolismo
12.
Nat Commun ; 9(1): 3061, 2018 08 03.
Artículo en Inglés | MEDLINE | ID: mdl-30076305

RESUMEN

An automated single-molecule imaging system developed for live-cell analyses based on artificial intelligence-assisted microscopy is presented. All significant procedures, i.e., searching for cells suitable for observation, detecting in-focus positions, and performing image acquisition and single-molecule tracking, are fully automated, and numerous highly accurate, efficient, and reproducible single-molecule imaging experiments in living cells can be performed. Here, the apparatus is applied for single-molecule imaging and analysis of epidermal growth factor receptors (EGFRs) in 1600 cells in a 96-well plate within 1 day. Changes in the lateral mobility of EGFRs on the plasma membrane in response to various ligands and drug concentrations are clearly detected in individual cells, and several dynamic and pharmacological parameters are determined, including the diffusion coefficient, oligomer size, and half-maximal effective concentration (EC50). Automated single-molecule imaging for systematic cell signaling analyses is feasible and can be applied to single-molecule screening, thus extensively contributing to biological and pharmacological research.


Asunto(s)
Rastreo Celular/métodos , Imagen Individual de Molécula/métodos , Animales , Inteligencia Artificial , Línea Celular , Membrana Celular , Cricetulus , Relación Dosis-Respuesta a Droga , Colorantes Fluorescentes , Cinética , Modelos Biológicos
13.
J Phys Chem B ; 122(18): 4838-4843, 2018 05 10.
Artículo en Inglés | MEDLINE | ID: mdl-29668280

RESUMEN

Constitutively active mutants (CAMs) of G-protein-coupled receptors (GPCRs) cause various kinds of diseases. Rhodopsin, a light-absorbing GPCR in animal retinas, has retinal as an endogenous ligand; only very low levels of activation of G-protein can be obtained with the ligand-free opsin. However, the CAM of opsin activates G-protein much more efficiently than the wild type, but the mechanism underlying this remains unclear. The present work revisits the constitutive activity of rhodopsin from the standpoint of conformational dynamics. Single-molecule observation of the M257Y mutant of bovine rhodopsin demonstrated that the switch between active and inactive conformations frequently occurred in M257Y opsin, and frequent generation of the active state results in the population shift toward the active state, which accounts for the constitutive activity of M257Y opsin. Our findings demonstrate that the protein function has a direct connection with the structural dynamics.


Asunto(s)
Rodopsina/química , Rodopsina/metabolismo , Células HEK293 , Humanos , Modelos Moleculares , Mutación , Conformación Proteica , Rodopsina/genética
14.
J Mol Biol ; 430(9): 1386-1401, 2018 04 27.
Artículo en Inglés | MEDLINE | ID: mdl-29505756

RESUMEN

Cell signaling depends on spatiotemporally regulated molecular interactions. Although the movements of signaling proteins have been analyzed with various technologies, how spatial dynamics influence the molecular interactions that transduce signals is unclear. Here, we developed a single-molecule method to analyze the spatiotemporal coupling between motility, clustering, and signaling. The analysis was performed with the epidermal growth factor receptor (EGFR), which triggers signaling through its dimerization and phosphorylation after association with EGF. Our results show that the few EGFRs isolated in membrane subdomains were released by an EGF-dependent increase in their diffusion area, facilitating molecular associations and producing immobile clusters. Using a two-color single-molecule analysis, we found that the EGF-induced state transition alters the properties of the immobile clusters, allowing them to interact for extended periods with the cytoplasmic protein, GRB2. Our study reveals a novel correlation between this molecular interaction and its mesoscale dynamics, providing the initial signaling node.


Asunto(s)
Factor de Crecimiento Epidérmico/metabolismo , Proteína Adaptadora GRB2/metabolismo , Imagen Individual de Molécula/métodos , Animales , Células CHO , Análisis por Conglomerados , Cricetulus , Citoplasma/metabolismo , Receptores ErbB/química , Receptores ErbB/metabolismo , Técnica del Anticuerpo Fluorescente , Humanos , Transducción de Señal , Análisis Espacio-Temporal
15.
J Biol Chem ; 293(6): 2206-2218, 2018 02 09.
Artículo en Inglés | MEDLINE | ID: mdl-29233889

RESUMEN

Feedback control is a key mechanism in signal transduction, intimately involved in regulating the outcome of the cellular response. Here, we report a novel mechanism by which PHLDA1, Pleckstrin homology-like domain, family A, member 1, negatively regulates ErbB receptor signaling by inhibition of receptor oligomerization. We have found that the ErbB3 ligand, heregulin, induces PHILDA1 expression in MCF-7 cells. Transcriptionally-induced PHLDA1 protein directly binds to ErbB3, whereas knockdown of PHLDA1 increases complex formation between ErbB3 and ErbB2. To provide insight into the mechanism for our time-course and single-cell experimental observations, we performed a systematic computational search of network topologies of the mathematical models based on receptor dimer-tetramer formation in the ErbB activation processes. Our results indicate that only a model in which PHLDA1 inhibits formation of both dimers and tetramer can explain the experimental data. Predictions made from this model were further validated by single-molecule imaging experiments. Our studies suggest a unique regulatory feature of PHLDA1 to inhibit the ErbB receptor oligomerization process and thereby control the activity of receptor signaling network.


Asunto(s)
Receptor ErbB-3/metabolismo , Factores de Transcripción/metabolismo , Humanos , Células MCF-7 , Modelos Químicos , Neurregulina-1/metabolismo , Multimerización de Proteína , Transducción de Señal , Imagen Individual de Molécula , Análisis de la Célula Individual , Factores de Transcripción/fisiología , Transcripción Genética
16.
Biophys Rev ; 10(2): 317-326, 2018 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-29243093

RESUMEN

Single-molecule imaging (SMI) of proteins in operation has a history of intensive investigations over 20 years and is now widely used in various fields of biology and biotechnology. We review the recent advances in SMI of fluorescently-tagged proteins in structural biology, focusing on technical applicability of SMI to the measurements in living cells. Basic technologies and recent applications of SMI in structural biology are introduced. Distinct from other methods in structural biology, SMI directly observes single molecules and single-molecule events one-by-one, thus, explicitly analyzing the distribution of protein structures and the history of protein dynamics. It also allows one to detect single events of protein interaction. One unique feature of SMI is that it is applicable in complicated and heterogeneous environments, including living cells. The numbers, location, movements, interaction, oligomerization, and conformation of single-protein molecules have been determined using SMI in cellular systems.

18.
Cell Rep ; 16(8): 2156-2168, 2016 08 23.
Artículo en Inglés | MEDLINE | ID: mdl-27524610

RESUMEN

Cell polarity arises through the spatial segregation of polarity regulators. PAR proteins are polarity regulators that localize asymmetrically to two opposing cortical domains. However, it is unclear how the spatially segregated PAR proteins interact to maintain their mutually exclusive partitioning. Here, single-molecule detection analysis in Caenorhabditis elegans embryos reveals that cortical PAR-2 diffuses only short distances, and, as a result, most PAR-2 molecules associate and dissociate from the cortex without crossing into the opposing domain. Our results show that cortical PAR-2 asymmetry is maintained by the local exchange reactions that occur at the cortical-cytoplasmic boundary. Additionally, we demonstrate that local exchange reactions are sufficient to maintain cortical asymmetry in a parameter-free mathematical model. These findings suggest that anterior and posterior PAR proteins primarily interact through the cytoplasmic pool and not via cortical diffusion.


Asunto(s)
Proteínas de Caenorhabditis elegans/metabolismo , Caenorhabditis elegans/metabolismo , Citoplasma/metabolismo , Embrión no Mamífero/metabolismo , Modelos Estadísticos , Animales , Caenorhabditis elegans/citología , Caenorhabditis elegans/genética , Proteínas de Caenorhabditis elegans/genética , Compartimento Celular , Polaridad Celular , Citoplasma/ultraestructura , Embrión no Mamífero/citología , Regulación de la Expresión Génica , Cinética , Fosforilación , Transporte de Proteínas , Imagen Individual de Molécula
19.
Biophys J ; 107(10): 2221-9, 2014 Nov 18.
Artículo en Inglés | MEDLINE | ID: mdl-25418290

RESUMEN

Cellular differentiation proceeds along complicated pathways, even when it is induced by extracellular signaling molecules. One of the major reasons for this complexity is the highly multidimensional internal dynamics of cells, which sometimes causes apparently stochastic responses in individual cells to extracellular stimuli. Therefore, to understand cell differentiation, it is necessary to monitor the internal dynamics of cells at single-cell resolution. Here, we used a Raman and autofluorescence spectrum analysis of single cells to detect dynamic changes in intracellular molecular components. MCF-7 cells are a human cancer-derived cell line that can be induced to differentiate into mammary-gland-like cells with the addition of heregulin (HRG) to the culture medium. We measured the spectra in the cytoplasm of MCF-7 cells during 12 days of HRG stimulation. The Raman scattering spectrum, which was the major component of the signal, changed with time. A multicomponent analysis of the Raman spectrum revealed that the dynamics of the major components of the intracellular molecules, including proteins and lipids, changed cyclically along the differentiation pathway. The background autofluorescence signals of Raman scattering also provided information about the differentiation process. Using the total information from the Raman and autofluorescence spectra, we were able to visualize the pathway of cell differentiation in the multicomponent phase space.


Asunto(s)
Diferenciación Celular/efectos de los fármacos , Neurregulina-1/farmacología , Espectrometría Raman , Humanos , Células MCF-7 , Espectrometría de Fluorescencia
20.
Science ; 344(6185): 760-4, 2014 May 16.
Artículo en Inglés | MEDLINE | ID: mdl-24833394

RESUMEN

A switchlike response in nuclear factor-κB (NF-κB) activity implies the existence of a threshold in the NF-κB signaling module. We show that the CARD-containing MAGUK protein 1 (CARMA1, also called CARD11)-TAK1 (MAP3K7)-inhibitor of NF-κB (IκB) kinase-ß (IKKß) module is a switch mechanism for NF-κB activation in B cell receptor (BCR) signaling. Experimental and mathematical modeling analyses showed that IKK activity is regulated by positive feedback from IKKß to TAK1, generating a steep dose response to BCR stimulation. Mutation of the scaffolding protein CARMA1 at serine-578, an IKKß target, abrogated not only late TAK1 activity, but also the switchlike activation of NF-κB in single cells, suggesting that phosphorylation of this residue accounts for the feedback.


Asunto(s)
Proteínas Adaptadoras de Señalización CARD/metabolismo , Guanilato Ciclasa/metabolismo , Quinasa I-kappa B/metabolismo , Quinasas Quinasa Quinasa PAM/metabolismo , FN-kappa B/agonistas , Receptores de Antígenos de Linfocitos B/metabolismo , Animales , Linfocitos B/metabolismo , Proteínas Adaptadoras de Señalización CARD/genética , Línea Celular , Pollos , Retroalimentación Fisiológica , Guanilato Ciclasa/genética , Quinasas Quinasa Quinasa PAM/genética , Ratones , Ratones Noqueados , Mutación , Fosforilación , Receptores de Antígenos de Linfocitos B/genética , Serina/genética , Serina/metabolismo , Transducción de Señal
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA