Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Clin Immunol ; 264: 110258, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38762063

RESUMEN

Lymphocytes such as CD4+ T cells and B cells mainly infiltrate the salivary glands; however, the precise roles and targets of autoreactive T cells and autoantibodies in the pathogenesis of Sjögren's Syndrome (SS) remain unclear. This study was designed to clarify the role of autoreactive T cells and autoantibodies at the single-cell level involved in the development of sialadenitis. Infiltrated CD4+ T and B cells in the salivary glands of a mouse model resembling SS were single-cell-sorted, and their T cell receptor (TCR) and B cell receptor (BCR) sequences were analyzed. The predominant TCR and BCR clonotypes were reconstituted in vitro, and their pathogenicity was evaluated by transferring reconstituted TCR-expressing CD4+ T cells into Rag2-/- mice and administering recombinant IgG in vivo. The reconstitution of Th17 cells expressing TCR (#G) in Rag2-/- mice resulted in the infiltration of T cells into the salivary glands and development of sialadenitis, while an autoantibody (IgGr22) was observed to promote the proliferation of pathogenic T cells. IgGr22 specifically recognizes double-stranded RNA (dsRNA) and induces the activation of dendritic cells, thereby enhancing the expression of IFN signature and inflammatory genes. TCR#G recognizes antigens related to the gut microbiota. Antibiotic treatment severely reduces the activation of TCR#G-expressing Th17 cells and suppresses sialadenitis development. These data suggest that the anti-dsRNA antibodies and, TCR recognizing the gut microbiota involved in the development of sialadenitis like SS. Thus, our model provides a novel strategy for defining the roles of autoreactive TCR and autoantibodies in the development and pathogenesis of SS.


Asunto(s)
Autoanticuerpos , Receptores de Antígenos de Linfocitos T , Sialadenitis , Síndrome de Sjögren , Animales , Síndrome de Sjögren/inmunología , Sialadenitis/inmunología , Autoanticuerpos/inmunología , Ratones , Receptores de Antígenos de Linfocitos T/inmunología , Receptores de Antígenos de Linfocitos T/genética , Ratones Noqueados , Glándulas Salivales/inmunología , Ratones Endogámicos C57BL , Linfocitos T CD4-Positivos/inmunología , Modelos Animales de Enfermedad , Linfocitos B/inmunología , Células Th17/inmunología , Femenino , Receptores de Antígenos de Linfocitos B/inmunología , Proteínas de Unión al ADN/inmunología , Proteínas de Unión al ADN/genética
2.
PLoS Genet ; 18(6): e1010235, 2022 06.
Artículo en Inglés | MEDLINE | ID: mdl-35648786

RESUMEN

The transcription factor NF-κB, which plays an important role in cell fate determination, is involved in the activation of super-enhancers (SEs). However, the biological functions of the NF-κB SEs in gene control are not fully elucidated. We investigated the characteristics of NF-κB-mediated SE activity using fluorescence imaging of RelA, single-cell transcriptome and chromatin accessibility analyses in anti-IgM-stimulated B cells. The formation of cell stimulation-induced nuclear RelA foci was abolished in the presence of hexanediol, suggesting an underlying process of liquid-liquid phase separation. The gained SEs induced a switch-like expression and enhanced cell-to-cell variability in transcriptional response. These properties were correlated with the number of gained cis-regulatory interactions, while switch-like gene induction was associated with the number of NF-κB binding sites in SE. Our study suggests that NF-κB SEs have an important role in the transcriptional regulation of B cells possibly through liquid condensate formation consisting of macromolecular interactions.


Asunto(s)
FN-kappa B , Factor de Transcripción ReIA , Núcleo Celular/metabolismo , Regulación de la Expresión Génica , FN-kappa B/genética , FN-kappa B/metabolismo , Unión Proteica , Secuencias Reguladoras de Ácidos Nucleicos , Factor de Transcripción ReIA/genética , Activación Transcripcional
3.
Cell Rep ; 31(9): 107724, 2020 06 02.
Artículo en Inglés | MEDLINE | ID: mdl-32492432

RESUMEN

NF-κB is a transcription factor that activates super enhancers (SEs) and typical enhancers (TEs) and triggers threshold and graded gene expression, respectively. However, the mechanisms by which NF-κB selectively participates in these enhancers remain unclear. Here we show using mouse primary B lymphocytes that SE activity simultaneously associates with chromatin opening and enriched NF-κB binding, resulting in a higher fold change and threshold expression upon B cell receptor (BCR) activation. The higher fold change results from longer DNA, whereas the threshold response is explained by synergy in DNA-NF-κB binding and is supported by the coexistence of PU.1 and NF-κB in a SE before cell stimulation. This model indicates that the pre-existing NF-κB functions as a seed and triggers its processive binding upon BCR activation. Our mathematical modeling of the single-cell transcriptome reveals an additional role for SEs in divergent clonal responses in B cells.


Asunto(s)
Elementos de Facilitación Genéticos/genética , FN-kappa B/metabolismo , Animales , Linfocitos B/citología , Linfocitos B/metabolismo , Células Cultivadas , Cromatina/metabolismo , Regulación de la Expresión Génica , Histonas/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Modelos Biológicos , Unión Proteica , Proteínas Proto-Oncogénicas/metabolismo , Transactivadores/metabolismo , Factor de Transcripción ReIA/metabolismo
4.
Front Immunol ; 10: 1450, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31293597

RESUMEN

Granzyme A (GzmA), together with perforin, are well-known for their cytotoxic activity against tumor or virus-infected cells. In addition to this cytotoxic function, GzmA stimulates several immune cell types and induces inflammation in the absence of perforin, however, its effect on the dendritic cell (DC) is unknown. In the current study, we showed that recombinant GzmA induced the phenotypic maturation of plasmacytoid DCs (pDCs) and conventional DCs (cDCs), but not their apoptosis. Particularly, GzmA made pDCs more functional, thus leading to production of type I interferon (IFN) via the TLR9-MyD88 pathway. We also demonstrated that GzmA binds TLR9 and co-localizes with it in endosomes. When co-administered with antigen, GzmA acted as a powerful adjuvant for eliciting antigen-specific cytotoxic CD8+ T lymphocytes (CTLs) that protected mice from tumor challenge. The induction of CTL was completely abolished in XCR1+ DC-depleted mice, whereas it was reduced to less than half in pDC-depleted or IFN-α/ß receptor knockout mice. Thus, CTL cross-priming was dependent on XCR1+cDC and also type I IFN, which was produced by GzmA-activated pDCs. These results indicate that GzmA -stimulated pDCs enhance the cross-priming activity of cDCs in situ. We also showed that the adjuvant effect of GzmA is superior to CpG-ODN and LPS. Our findings highlight the ability of GzmA to bridge innate and adaptive immune responses via pDC help and suggest that GzmA may be useful as a vaccine adjuvant.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Células Dendríticas/inmunología , Granzimas/farmacología , Inmunidad Celular/efectos de los fármacos , Células Plasmáticas/inmunología , Animales , Linfocitos T CD8-positivos/citología , Células Dendríticas/citología , Granzimas/genética , Granzimas/inmunología , Ratones , Ratones Noqueados , Factor 88 de Diferenciación Mieloide/genética , Factor 88 de Diferenciación Mieloide/inmunología , Células Plasmáticas/citología , Receptor Toll-Like 9/genética , Receptor Toll-Like 9/inmunología
5.
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
6.
Sci Rep ; 6: 27706, 2016 06 09.
Artículo en Inglés | MEDLINE | ID: mdl-27277891

RESUMEN

Different dynamic behaviours of signalling activity can induce distinct biological responses in a variety of cells. However, the molecular mechanisms that determine the dynamics of kinase activities in immune cells are not well understood. In this study, we showed that the duration of both IκB kinase (IKK) and extracellular signal-regulated kinase (ERK) activities in B cell receptor (BCR)- and CD40-signalling pathways in B cells were regulated by transcriptional feedback loops. We conducted a time-course transcriptome analysis after BCR or CD40 stimulation and identified the following four candidate genes as feedback regulators for IKK and ERK: inhibitor of apoptosis protein (IAP), TNF alpha-induced protein 3, dual-specificity phosphatase 5, and sprouty homolog 2. Quantitative experiments and mathematical modelling suggested that IAP inhibition shortened the duration of IKK and ERK activity following both BCR and CD40 pathway stimulation, indicating a positive role for IAP in B cell signalling. Furthermore, transient kinase activities induced by IAP blockage reduced the levels of delayed expression genes. Together, our findings suggest that IKK and ERK activity durations can be fine-tuned by the coordinated regulation of positive and negative transcriptional feedback and that these network properties determine the biological output of B cells.


Asunto(s)
Linfocitos B/metabolismo , Ligando de CD40/farmacología , Receptores de Antígenos de Linfocitos B/metabolismo , Transducción de Señal , Animales , Línea Celular , Fosfatasas de Especificidad Dual/genética , Quinasas MAP Reguladas por Señal Extracelular/genética , Retroalimentación Fisiológica , Quinasa I-kappa B/genética , Proteínas Inhibidoras de la Apoptosis/genética , Péptidos y Proteínas de Señalización Intracelular/genética , Proteínas de la Membrana/genética , Ratones , Proteínas Serina-Treonina Quinasas , Proteínas Proto-Oncogénicas c-bcr/metabolismo , Proteína 3 Inducida por el Factor de Necrosis Tumoral alfa/genética
7.
NPJ Syst Biol Appl ; 2: 16024, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-28725478

RESUMEN

Transcription factor nuclear factor kappa B (NF-κB) shows cooperative switch-like activation followed by prolonged oscillatory nuclear translocation in response to extracellular stimuli. These dynamics are important for activation of the NF-κB transcriptional machinery, however, NF-κB activity regulated by coordinated actions of these dynamics has not been elucidated at the system level. Using a variety of B cells with artificially rewired NF-κB signaling networks, we show that oscillations and switch-like activation of NF-κB can be dissected and that, under some conditions, these two behaviors are separated upon antigen receptor activation. Comprehensive quantitative experiments and mathematical analysis showed that the functional role of switch activation in the NF-κB system is to overcome transient IKK (IκB kinase) activity to amplify nuclear translocation of NF-κB, thereby inducing the prolonged NF-κB oscillatory behavior necessary for target gene expression and B-cell activation.

8.
FEBS J ; 282(4): 613-29, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25491268

RESUMEN

Extracellular signal-regulated kinase (ERK) plays a central role in signal transduction networks and cell fate decisions. Sustained ERK activation induces cell differentiation, whereas transient ERK results in the proliferation of several types of cells. Sustained ERK activity stabilizes the proteins of early-response gene products. However, the effect of ERK activity duration on mRNA stability is unknown. We analyzed the quantitative relationship between the duration of four ERK activity kinetics and the mRNA expression profile in growth factor-treated cells. Time-course transcriptome analysis revealed that the cells with prolonged ERK activity generally showed sustained mRNA expression of late response genes but not early or mid genes. Selected late response genes decayed more rapidly in the presence of a specific ERK inhibitor than a general transcription inhibitor and the decay rate was not related to the number of AU-rich elements. Our results suggest that sustained ERK activity plays an important role in the lifespan of the mRNA encoded by late response genes, in addition to the previously demonstrated role in protein stabilization of early-response genes, including transcription factors regulating the transcription of mid and late genes. This double-positive regulation of ligand-induced genes, also termed feedforward regulation, is critical in cell fate decisions.


Asunto(s)
Receptores ErbB/metabolismo , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Estabilidad del ARN/genética , Butadienos/farmacología , Diferenciación Celular/efectos de los fármacos , Diferenciación Celular/genética , Diferenciación Celular/fisiología , Dactinomicina/farmacología , Receptores ErbB/genética , Quinasas MAP Reguladas por Señal Extracelular/genética , Humanos , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Sistema de Señalización de MAP Quinasas/genética , Sistema de Señalización de MAP Quinasas/fisiología , Células MCF-7 , Nitrilos/farmacología , Estabilidad del ARN/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Transducción de Señal/fisiología
10.
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
11.
J Biol Chem ; 286(1): 818-29, 2011 Jan 07.
Artículo en Inglés | MEDLINE | ID: mdl-21044952

RESUMEN

Quantitative phosphoproteome and transcriptome analysis of ligand-stimulated MCF-7 human breast cancer cells was performed to understand the mechanisms of tamoxifen resistance at a system level. Phosphoproteome data revealed that WT cells were more enriched with phospho-proteins than tamoxifen-resistant cells after stimulation with ligands. Surprisingly, decreased phosphorylation after ligand perturbation was more common than increased phosphorylation. In particular, 17ß-estradiol induced down-regulation in WT cells at a very high rate. 17ß-Estradiol and the ErbB ligand heregulin induced almost equal numbers of up-regulated phospho-proteins in WT cells. Pathway and motif activity analyses using transcriptome data additionally suggested that deregulated activation of GSK3ß (glycogen-synthase kinase 3ß) and MAPK1/3 signaling might be associated with altered activation of cAMP-responsive element-binding protein and AP-1 transcription factors in tamoxifen-resistant cells, and this hypothesis was validated by reporter assays. An examination of clinical samples revealed that inhibitory phosphorylation of GSK3ß at serine 9 was significantly lower in tamoxifen-treated breast cancer patients that eventually had relapses, implying that activation of GSK3ß may be associated with the tamoxifen-resistant phenotype. Thus, the combined phosphoproteome and transcriptome data set analyses revealed distinct signal transcription programs in tumor cells and provided a novel molecular target to understand tamoxifen resistance.


Asunto(s)
Neoplasias de la Mama/genética , Neoplasias de la Mama/metabolismo , Resistencia a Antineoplásicos/genética , Perfilación de la Expresión Génica/métodos , Fosfoproteínas/metabolismo , Proteoma/metabolismo , Tamoxifeno/farmacología , Neoplasias de la Mama/inmunología , Neoplasias de la Mama/patología , Línea Celular Tumoral , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/metabolismo , Resistencia a Antineoplásicos/efectos de los fármacos , Glucógeno Sintasa Quinasa 3/metabolismo , Glucógeno Sintasa Quinasa 3 beta , Humanos , Ligandos , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Fosfoproteínas/genética , Proteoma/genética , Transducción de Señal/efectos de los fármacos , Factor de Transcripción AP-1/metabolismo , Transcripción Genética , Resultado del Tratamiento
12.
Cell ; 141(5): 884-96, 2010 May 28.
Artículo en Inglés | MEDLINE | ID: mdl-20493519

RESUMEN

Activation of ErbB receptors by epidermal growth factor (EGF) or heregulin (HRG) determines distinct cell-fate decisions, although signals propagate through shared pathways. Using mathematical modeling and experimental approaches, we unravel how HRG and EGF generate distinct, all-or-none responses of the phosphorylated transcription factor c-Fos. In the cytosol, EGF induces transient and HRG induces sustained ERK activation. In the nucleus, however, ERK activity and c-fos mRNA expression are transient for both ligands. Knockdown of dual-specificity phosphatases extends HRG-stimulated nuclear ERK activation, but not c-fos mRNA expression, implying the existence of a HRG-induced repressor of c-fos transcription. Further experiments confirmed that this repressor is mainly induced by HRG, but not EGF, and requires new protein synthesis. We show how a spatially distributed, signaling-transcription cascade robustly discriminates between transient and sustained ERK activities at the c-Fos system level. The proposed control mechanisms are general and operate in different cell types, stimulated by various ligands.


Asunto(s)
Modelos Biológicos , Proteínas Proto-Oncogénicas c-fos/genética , Línea Celular Tumoral , Fosfatasas de Especificidad Dual/metabolismo , Receptores ErbB/metabolismo , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Humanos , Neurregulina-1/metabolismo , Estabilidad Proteica , Proteínas Proto-Oncogénicas c-fos/metabolismo , Transcripción Genética
13.
BMC Genomics ; 10: 545, 2009 Nov 20.
Artículo en Inglés | MEDLINE | ID: mdl-19925682

RESUMEN

BACKGROUND: Sharing a common ErbB/HER receptor signaling pathway, heregulin (HRG) induces differentiation of MCF-7 human breast cancer cells while epidermal growth factor (EGF) elicits proliferation. Although cell fates resulting from action of the aforementioned ligands completely different, the respective gene expression profiles in early transcription are qualitatively similar, suggesting that gene expression during late transcription, but not early transcription, may reflect ligand specificity. In this study, based on both the data from time-course quantitative real-time PCR on over 2,000 human transcription factors and microarray of all human genes, we identified a series of transcription factors which may control HRG-specific late transcription in MCF-7 cells. RESULTS: We predicted that four transcription factors including EGR4, FRA-1, FHL2, and DIPA should have responsibility of regulation in MCF-7 cell differentiation. Validation analysis suggested that one member of the activator protein 1 (AP-1) family, FOSL-1 (FRA-1 gene), appeared immediately following c-FOS expression, might be responsible for expression of transcription factor FHL2 through activation of the AP-1 complex. Furthermore, RNAi gene silencing of FOSL-1 and FHL2 resulted in increase of extracellular signal-regulated kinase (ERK) phosphorylation of which duration was sustained by HRG stimulation. CONCLUSION: Our analysis indicated that a time-dependent transcriptional regulatory network including c-FOS, FRA-1, and FHL2 is vital in controlling the ERK signaling pathway through a negative feedback loop for MCF-7 cell differentiation.


Asunto(s)
Diferenciación Celular/efectos de los fármacos , Regulación de la Expresión Génica/efectos de los fármacos , Factores de Transcripción/genética , Línea Celular Tumoral , Factor de Crecimiento Epidérmico/farmacología , Perfilación de la Expresión Génica , Humanos , Ligandos , Neurregulina-1/farmacología , Fenotipo , Reacción en Cadena de la Polimerasa , Interferencia de ARN , Reproducibilidad de los Resultados , Transducción de Señal/efectos de los fármacos , Factores de Tiempo , Factores de Transcripción/deficiencia , Factores de Transcripción/metabolismo
14.
FEBS J ; 276(18): 5239-51, 2009 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-19674104

RESUMEN

Controlled activation of epidermal growth factor receptor (EGFR) is systematically guaranteed at the molecular level; however, aberrant activation of EGFR is frequently found in cancer. Transcription induced by EGFR activation often involves the coordinated expression of genes that positively and negatively regulate the original signaling pathway; therefore, alterations in EGFR kinase activity may reflect changes in gene expression associated with the pathway. In the present study, we investigated transcriptional changes after EGF stimulation with or without the EGFR kinase inhibitor Iressa in H1299 human non-small-cell lung cancer cells [parental H1299, H1299 cells that overexpress wild-type EGFR (EGFR-WT) and mutant H1299 cells that overexpress EGFR where Leu858 is substituted with Arg (L858R)]. The results obtained clearly demonstrate differences in transcriptional activity in the absence or presence of EGFR kinase activity, with genes sharing the same molecular functions showing distinct expression dynamics. The results show the particular enrichment of EGFR/ErbB signaling-related genes in a differentially expressed gene set, and significant protein expression of MIG6/RALT(ERRFI1), an EGFR negative regulator, was confirmed in L858R. High MIG6 protein expression was correlated with basal EGFR phosphorylation and inversely correlated with EGF-induced extracellular signal-regulated protein kinase phosphorylation levels. Investigation of the NCI-60 cell lines showed that ERRFI1 expression was correlated with EGFR expression, regardless of tissue type. These results suggest that cells accumulate MIG6 as an inherent negative regulator to suppress excess EGFR activity when basal EGFR kinase activity is considerably high. Taking all the above together, an EGFR mutation can cause transcriptional changes to accommodate the activation potency of the original signaling pathway at the cellular level.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/fisiología , Receptores ErbB/fisiología , Proteínas Adaptadoras Transductoras de Señales/genética , Línea Celular Tumoral , Receptores ErbB/antagonistas & inhibidores , Receptores ErbB/genética , Perfilación de la Expresión Génica , Genes erbB , Humanos , Péptidos y Proteínas de Señalización Intracelular/fisiología , Mutación , Proteínas del Tejido Nervioso/fisiología , Transducción de Señal , Transcripción Genética , Proteínas Supresoras de Tumor
15.
Cancer Genomics Proteomics ; 5(3-4): 161-8, 2008.
Artículo en Inglés | MEDLINE | ID: mdl-18820370

RESUMEN

Cellular signal transduction pathways and gene expression are tightly regulated to accommodate changes in response to physiological environments. In the current study, molecules were identified that are activated as a result of intracellular signaling and immediately expressed as mRNA in MCF-7 breast cancer cells shortly after stimulation of ErbB receptor ligands, epidermal growth factor (EGF) or heregulin (HRG). For the identification of tyrosine-phosphorylated proteins and expressed genes, a SILAC (stable isotopic labeling using amino acids in cell culture) method and Affymetrix gene expression array system, respectively, were used. Unexpectedly, the overlapping of genes appeared in two experimental datasets was very low for HRG (43 hits in the proteome data, 1,655 in the transcriptome data, and 5 hits common to both datasets), while no overlapping gene was detected for EGF (15 hits in the proteome data, 211 hits in the transcriptome data, and no hits common to both datasets). The HRG overlapping genes included ERBB2, NEDD9, MAPK3, JUP and EPHA2. Biological pathway analysis indicated that HRG-stimulated molecular activation is significantly related to cancer pathways including bladder cancer, chronic myeloid leukemia and pancreatic cancer (p < 0.05). The proteome datasets of EGF and HRG contain molecules that are related to Axon guidance, ErbB signaling and VEGF signaling at a high rate.


Asunto(s)
Factor de Crecimiento Epidérmico/metabolismo , Neurregulina-1/metabolismo , Fosfoproteínas/metabolismo , Proteoma , ARN Mensajero/genética , Línea Celular Tumoral , Humanos , Ligandos , Fosfoproteínas/genética , Fosforilación
16.
PLoS One ; 3(3): e1803, 2008 Mar 19.
Artículo en Inglés | MEDLINE | ID: mdl-18350142

RESUMEN

Heregulin beta-1 (HRG) is an extracellular ligand that activates mitogen-activated protein kinase (MAPK) and phosphatidylinositol-3-OH kinase (PI3K)/Akt signaling pathways through ErbB receptors. MAPK and Akt have been shown to phosphorylate the estrogen receptor (ER) at Ser-118 and Ser-167, respectively, thereby mimicking the effects of estrogenic activity such as estrogen responsive element (ERE)-dependent transcription. In the current study, integrative analysis was performed using two tiling array platforms, comprising histone H3 lysine 9 (H3K9) acetylation and RNA mapping, together with array comparative genomic hybridization (CGH) analysis in an effort to identify HRG-regulated genes in ER-positive MCF-7 breast cancer cells. Through application of various threshold settings, 333 (326 up-regulated and 7 down-regulated) HRG-regulated genes were detected. Prediction of upstream transcription factors (TFs) and pathway analysis indicated that 21% of HRG-induced gene regulation may be controlled by the MAPK cascade, while only 0.6% of the gene expression is controlled by ERE. A comparison with previously reported estrogen (E2)-regulated gene expression data revealed that only 12 common genes were identified between the 333 HRG-regulated (3.6%) and 239 E2-regulated (5.0%) gene groups. However, with respect to enriched upstream TFs, 4 common TFs were identified in the 14 HRG-regulated (28.6%) and 13 E2-regulated (30.8%) gene groups. These results indicated that while E2 and HRG may induce common TFs, the regulatory mechanisms that govern HRG- and E2-induced gene expression differ.


Asunto(s)
Genoma Humano , Neurregulina-1/farmacología , Receptores de Estrógenos/fisiología , Transcripción Genética/fisiología , Línea Celular Tumoral , Humanos , Receptores de Estrógenos/química , Receptores de Estrógenos/metabolismo , Serina/metabolismo
17.
PLoS One ; 3(3): e1782, 2008 Mar 12.
Artículo en Inglés | MEDLINE | ID: mdl-18335053

RESUMEN

Ligand-induced homo- and hetero-dimer formation of ErbB receptors results in different biological outcomes irrespective of recruitment and activation of similar effector proteins. Earlier experimental research indicated that cells expressing both EGFR (epidermal growth factor receptor) and the ErbB4 receptor (E1/4 cells) induced E1/4 cell-specific B-Raf activation and higher extracellular signal-regulated kinase (ERK) activation, followed by cellular transformation, than cells solely expressing EGFR (E1 cells) in Chinese hamster ovary (CHO) cells. Since our experimental data revealed the presence of positive feedback by ERK on upstream pathways, it was estimated that the cross-talk/feedback pathway structure of the Raf-MEK-ERK cascade might affect ERK activation dynamics in our cell system. To uncover the regulatory mechanism concerning the ERK dynamics, we used topological models and performed parameter estimation for all candidate structures that possessed ERK-mediated positive feedback regulation of Raf. The structure that reliably reproduced a series of experimental data regarding signal amplitude and duration of the signaling molecules was selected as a solution. We found that the pathway structure is characterized by ERK-mediated positive feedback regulation of B-Raf and B-Raf-mediated negative regulation of Raf-1. Steady-state analysis of the estimated structure indicated that the amplitude of Ras activity might critically affect ERK activity through ERK-B-Raf positive feedback coordination with sustained B-Raf activation in E1/4 cells. However, Rap1 that positively regulates B-Raf activity might be less effective concerning ERK and B-Raf activity. Furthermore, we investigated how such Ras activity in E1/4 cells can be regulated by EGFR/ErbB4 heterodimer-mediated signaling. From a sensitivity analysis of the detailed upstream model for Ras activation, we concluded that Ras activation dynamics is dominated by heterodimer-mediated signaling coordination with a large initial speed of dimerization when the concentration of the ErbB4 receptor is considerably high. Such characteristics of the signaling cause the preferential binding of the Grb2-SOS complex to heterodimer-mediated signaling molecules.


Asunto(s)
Receptores ErbB/metabolismo , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Transducción de Señal , Animales , Western Blotting , Células CHO , Cricetinae , Cricetulus , Humanos , Cinética , Modelos Teóricos , Fosforilación , Receptor ErbB-4
18.
Mol Syst Biol ; 3: 144, 2007.
Artículo en Inglés | MEDLINE | ID: mdl-18004277

RESUMEN

Deregulation of ErbB signaling plays a key role in the progression of multiple human cancers. To help understand ErbB signaling quantitatively, in this work we combine traditional experiments with computational modeling, building a model that describes how stimulation of all four ErbB receptors with epidermal growth factor (EGF) and heregulin (HRG) leads to activation of two critical downstream proteins, extracellular-signal-regulated kinase (ERK) and Akt. Model analysis and experimental validation show that (i) ErbB2 overexpression, which occurs in approximately 25% of all breast cancers, transforms transient EGF-induced signaling into sustained signaling, (ii) HRG-induced ERK activity is much more robust to the ERK cascade inhibitor U0126 than EGF-induced ERK activity, and (iii) phosphoinositol-3 kinase is a major regulator of post-peak but not pre-peak EGF-induced ERK activity. Sensitivity analysis leads to the hypothesis that ERK activation is robust to parameter perturbation at high ligand doses, while Akt activation is not.


Asunto(s)
Modelos Biológicos , Proteínas Tirosina Quinasas Receptoras/metabolismo , Transducción de Señal , Androstadienos/farmacología , Butadienos/farmacología , Línea Celular Tumoral , Membrana Celular/efectos de los fármacos , Membrana Celular/enzimología , Dimerización , Activación Enzimática/efectos de los fármacos , Factor de Crecimiento Epidérmico/farmacología , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Retroalimentación Fisiológica/efectos de los fármacos , Humanos , Ligandos , Neurregulina-1/farmacología , Nitrilos/farmacología , Inhibidores de las Quinasa Fosfoinosítidos-3 , Fosforilación/efectos de los fármacos , Estructura Terciaria de Proteína , Proteínas Proto-Oncogénicas c-akt/metabolismo , Reproducibilidad de los Resultados , Transducción de Señal/efectos de los fármacos , Wortmanina
19.
J Biol Chem ; 282(6): 4045-56, 2007 Feb 09.
Artículo en Inglés | MEDLINE | ID: mdl-17142811

RESUMEN

ErbB receptor ligands, epidermal growth factor (EGF) and heregulin (HRG), induce dose-dependent transient and sustained intracellular signaling, proliferation, and differentiation of MCF-7 breast cancer cells, respectively. In an effort to delineate the ligand-specific cell determination mechanism, we investigated time course gene expressions induced by EGF and HRG that induce distinct cellular phenotypes in MCF-7 cells. To analyze independently the effects of ligand dosage and time for gene expression, we developed a statistical method for estimating the two effects. Our results indicated that signal transduction pathways convey quantitative properties of the dose-dependent activation of ErbB receptor to early transcription. The results also implied that moderate changes in the expression levels of a number of genes, not the predominant regulation of a few specific genes, might cooperatively work at the early stage of the transcription for determining cell fate. However, the EGF- and HRG-induced distinct signal durations resulted in the ligand-oriented biphasic induction of proteins after 20 min. The selected gene list and HRG-induced prolonged signaling suggested that transcriptional feedback to the intracellular signaling results in a graded to biphasic response in the cell determination process and that each ErbB receptor is inextricably responsible for the control of amplitude and duration of cellular biochemical reactions.


Asunto(s)
Diferenciación Celular/genética , Receptor ErbB-2/fisiología , Transducción de Señal/genética , Transcripción Genética , Línea Celular Tumoral , Proliferación Celular , Factor de Crecimiento Epidérmico/metabolismo , Factor de Crecimiento Epidérmico/fisiología , Retroalimentación Fisiológica , Humanos , Cinética , Ligandos , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Neurregulina-1/metabolismo , Neurregulina-1/fisiología , Fosforilación , Proteínas Proto-Oncogénicas c-akt/metabolismo , Proteínas Proto-Oncogénicas c-fos/biosíntesis , Proteínas Proto-Oncogénicas c-fos/genética , Receptor ErbB-2/metabolismo , Sistemas de Mensajero Secundario/genética
20.
Mol Cell Biochem ; 285(1-2): 165-71, 2006 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-16477370

RESUMEN

Expression of ErbB4 receptor is correlated with the incidence of non-metastatic types of human cancers, whereas the overexpression of other ErbB receptor families (ErbB1/EGFR, ErbB2 and ErbB3) is correlated to the formation of metastatic tumors. However, the molecular mechanism underlying this phenomenon has been unclear. Earlier, we demonstrated that okadaic acid (OA), an inhibitor of a serine/threonine phosphatase PP2A, stimulated the growth hormone-induced ERK phosphorylation in the wild type Chinese hamster ovary (CHO) cells and the cells expressing ErbB1 receptor, but suppressed ERK activation in CHO cells that express ErbB4 receptor. PP2A had been understood as a negative regulator of the growth hormone-stimulated signal transduction pathways, however, this observation suggested that expression of ErbB4 receptor reversed the regulation of PP2A in the ErbB4 signalling pathway. In this study, we found that OA suppressed phosphorylation of Shc at Tyr317, therefore it down-regulated ERK phosphorylation in the ErbB4 expressing CHO cells. Accordingly, basal PP2A contributed to the phosphorylation of Shc Tyr317 in ErbB4 expressing CHO cells, nevertheless it had been reported that PP2A negatively regulates Shc tyrosine phosphorylation in the EGF- or IGF-I-induced signalling pathways. By testing OA for human cancer cell lines that express different types of ErbB receptors, we found that ErbB4 receptor expression was accompanied with positive regulation of PP2A for phosphorylation of Shc Tyr317 and its downstream ERK phosphorylation in MCF-7 and SK-OV-3 cell lines, but not in LNCaP and PC-3 cells. Thus, PP2A regulates the ERK activity in a cell-specific manner, and it is speculated that distinct regulation of PP2A in the ErbB4 receptor signalling pathway may cause a difference in progression of cancer phenotypes.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Receptores ErbB/metabolismo , Fosfoproteínas Fosfatasas/metabolismo , Transducción de Señal , Animales , Progresión de la Enfermedad , Factor de Crecimiento Epidérmico/farmacología , Humanos , Ácido Ocadaico/farmacología , Fenotipo , Fosfoproteínas Fosfatasas/fisiología , Fosforilación/efectos de los fármacos , Receptor ErbB-4 , Proteínas Adaptadoras de la Señalización Shc , Proteína Transformadora 1 que Contiene Dominios de Homología 2 de Src , Células Tumorales Cultivadas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...