Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
1.
Tumour Biol ; 37(2): 2519-26, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26386720

RESUMEN

Lysophosphatidic acid (LPA) is a small glycerophospholipid ubiquitously present in tissues and plasma. It acts through receptors belonging to the G-protein-coupled receptor (GPCR) family, is involved in several biological processes, and is strongly implicated in different cancers. In this paper, we have investigated the effects of LPA on DNA synthesis and migration in a panel of pancreatic and colon cancer cells, with particular focus on the involvement of the epidermal growth factor (EGF) receptor (EGFR) in LPA-induced signaling. LPA stimulated DNA synthesis and/or migration in all the cell lines included in this study. In five of the six cell lines, LPA induced phosphorylation of the EGFR, and the effects on EGFR and Akt, and in some of the cells also ERK, were sensitive to the EGFR tyrosine kinase inhibitor gefitinib, strongly suggesting LPA-induced EGFR transactivation in these cells. In contrast, in one of the pancreatic carcinoma cell lines (Panc-1), we found no evidence of transactivation of the EGFR. In the pancreatic carcinoma cell lines where transactivation took place (BxPC3, AsPC1, HPAFII), gefitinib reduced LPA-induced DNA synthesis and/or migration. However, we also found evidence of transactivation in the two colon carcinoma cell lines (HT29, HCT116) although gefitinib did not inhibit LPA-induced DNA synthesis or migration in these cells. Taken together, the data indicate that in many gastrointestinal carcinoma cells, LPA uses EGFR transactivation as a mechanism when exerting such effects as stimulation of cell proliferation and migration, but EGFR-independent pathways may be involved instead of, or in concerted action with, the EGFR transactivation.


Asunto(s)
Movimiento Celular/efectos de los fármacos , Neoplasias Colorrectales/genética , ADN/efectos de los fármacos , Receptores ErbB/metabolismo , Lisofosfolípidos/farmacología , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/patología , Carcinoma/genética , Carcinoma/metabolismo , Carcinoma/patología , Línea Celular Tumoral , Movimiento Celular/genética , Proliferación Celular/efectos de los fármacos , Proliferación Celular/genética , Neoplasias Colorrectales/metabolismo , Neoplasias Colorrectales/patología , ADN/genética , Gefitinib , Células HCT116 , Células HT29 , Humanos , Lisofosfolípidos/fisiología , Neoplasias Pancreáticas/metabolismo , Fosforilación/efectos de los fármacos , Fosforilación/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , Quinazolinas/farmacología , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Activación Transcripcional/efectos de los fármacos , Activación Transcripcional/genética
2.
Mol Cancer ; 14: 14, 2015 Jan 27.
Artículo en Inglés | MEDLINE | ID: mdl-25623255

RESUMEN

BACKGROUND: B cell precursor acute lymphoblastic leukaemia (BCP-ALL) is the most common paediatric cancer. BCP-ALL blasts typically retain wild type p53, and are therefore assumed to rely on indirect measures to suppress transformation-induced p53 activity. We have recently demonstrated that the second messenger cyclic adenosine monophosphate (cAMP) through activation of protein kinase A (PKA) has the ability to inhibit DNA damage-induced p53 accumulation and thereby promote survival of the leukaemic blasts. Development of BCP-ALL in the bone marrow (BM) is supported by resident BM-derived mesenchymal stromal cells (MSCs). MSCs are known to produce prostaglandin E(2) (PGE(2)) which upon binding to its receptors is able to elicit a cAMP response in target cells. We hypothesized that PGE(2) produced by stromal cells in the BM microenvironment could stimulate cAMP production and PKA activation in BCP-ALL cells, thereby suppressing p53 accumulation and promoting survival of the malignant cells. METHODS: Primary BCP-ALL cells isolated from BM aspirates at diagnosis were cocultivated with BM-derived MSCs, and effects on DNA damage-induced p53 accumulation and cell death were monitored by SDS-PAGE/immunoblotting and flow cytometry-based methods, respectively. Effects of intervention of signalling along the PGE(2)-cAMP-PKA axis were assessed by inhibition of PGE(2) production or PKA activity. Statistical significance was tested by Wilcoxon signed-rank test or paired samples t test. RESULTS: We demonstrate that BM-derived MSCs produce PGE(2) and protect primary BCP-ALL cells from p53 accumulation and apoptotic cell death. The MSC-mediated protection of DNA damage-mediated cell death is reversible upon inhibition of PGE(2) synthesis or PKA activity. Furthermore our results indicate differences in the sensitivity to variations in p53 levels between common cytogenetic subgroups of BCP-ALL. CONCLUSIONS: Our findings support our hypothesis that BM-derived PGE(2), through activation of cAMP-PKA signalling in BCP-ALL blasts, can inhibit the tumour suppressive activity of wild type p53, thereby promoting leukaemogenesis and protecting against therapy-induced leukaemic cell death. These novel findings identify the PGE(2)-cAMP-PKA signalling pathway as a possible target for pharmacological intervention with potential relevance for treatment of BCP-ALL.


Asunto(s)
Daño del ADN , Dinoprostona/metabolismo , Células Madre Mesenquimatosas/metabolismo , Leucemia-Linfoma Linfoblástico de Células Precursoras B/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Muerte Celular , Línea Celular Tumoral , Técnicas de Cocultivo , Proteínas Quinasas Dependientes de AMP Cíclico/antagonistas & inhibidores , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Humanos , Modelos Biológicos , Transducción de Señal
3.
Cell Biol Int ; 39(10): 1177-84, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26146811

RESUMEN

The receptor tyrosine kinases EGFR and Met induce phosphorylation of the docking protein Gab1, and there is evidence that Gab1 may have a role in the signaling from these receptors. Studying hepatocytes, we previously found that although Gab1 mechanistically interacted in different ways with EGFR and Met, it was involved in mitogenic signaling induced by both EGF and HGF. It has been reported that in EGFR, Gab1 is required particularly at a low dose of EGF. Whether this also applies to HGF/Met signaling has not been investigated. We have studied the role of Gab1 in activation of the Akt and ERK pathways at low- and high-intensity stimulation with EGF and HGF in cultured hepatocytes. In cells where Gab1 was depleted by a specific Gab1-directed siRNA, the EGF-induced phosphorylation of ERK was lowered and HGF-induced phosphorylation of both ERK and Akt was substantially reduced. These effects were more marked at low-dose HGF stimulation. The inhibitory consequence of Gab1 depletion was particularly pronounced for HGF-induced Akt phosphorylation. The results suggest that Gab1 is an important signal amplifier for low-intensity stimulation by HGF.


Asunto(s)
Factor de Crecimiento de Hepatocito/fisiología , Hepatocitos/metabolismo , Fosfoproteínas/fisiología , Animales , Células Cultivadas , Hepatocitos/enzimología , Ratas Wistar , Transducción de Señal
4.
Biochim Biophys Acta ; 1833(12): 3286-3294, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24126105

RESUMEN

Grb2-associated binder (Gab) family proteins are docking molecules that can interact with receptor tyrosine kinases (RTKs) and cytokine receptors and bind several downstream signalling proteins. Studies in several cell types have shown that Gab1 may have a role in signalling mediated by the two RTKs epidermal growth factor (EGF) receptor (EGFR) and Met, the receptor for hepatocyte growth factor (HGF), but the involvement of Gab1 in EGFR and Met signalling has not been directly compared in the same cell. We have studied mechanisms of activation and role in mitogenic signalling of Gab1 in response to EGF and HGF in cultured rat hepatocytes. Gab1, but not Gab2, was expressed in the hepatocytes and was phosphorylated upon stimulation with EGF or HGF. Depletion of Gab1, using siRNA, decreased the ERK and Akt activation, cyclin D1 expression, and DNA synthesis in response to both EGF and HGF. Studies of mechanisms of recruitment to the receptors showed that HGF induced co-precipitation of Gab1 and Met while EGF induced binding of Gab1 to Grb2 but not to EGFR. Gab1 activation in response to both EGF and HGF was dependent on PI3K. While EGF activated Gab1 and Shc equally, within the same concentration range, HGF very potently and almost exclusively activated Gab1, having only a minimal effect on Shc. Collectively, our results strongly suggest that although Gab1 interacts differently with EGFR and Met, it is involved in mitogenic signalling mediated by both these growth factor receptors in hepatocytes.


Asunto(s)
Factor de Crecimiento Epidérmico/farmacología , Factor de Crecimiento de Hepatocito/farmacología , Hepatocitos/metabolismo , Mitógenos/farmacología , Fosfoproteínas/metabolismo , Transducción de Señal/efectos de los fármacos , Animales , Células Cultivadas , Ciclina D1/metabolismo , ADN/biosíntesis , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Hepatocitos/efectos de los fármacos , Hepatocitos/enzimología , Humanos , Masculino , Ratones , Fosfatidilinositol 3-Quinasas/metabolismo , Fosforilación/efectos de los fármacos , Proteínas Proto-Oncogénicas c-akt/metabolismo , Ratas , Ratas Wistar , Proteínas Adaptadoras de la Señalización Shc/metabolismo , Factores de Tiempo
5.
BMC Cancer ; 14: 432, 2014 Jun 13.
Artículo en Inglés | MEDLINE | ID: mdl-24928086

RESUMEN

BACKGROUND: Oral squamous cell carcinoma is an aggressive neoplasm with serious morbidity and mortality, which typically spreads through local invasive growth. Lysophosphatidic acid (LPA) is involved in a number of biological processes, and may have a role in cancer cell migration and invasiveness. LPA is present in most tissues and can activate cells through six different LPA receptors (LPAR1-6). Although LPA is predominantly promigratory, some of the receptors may have antimigratory effects in certain cells. The signalling mechanisms of LPA are not fully understood, and in oral carcinoma cells the specific receptors and pathways involved in LPA-stimulated migration are unknown. METHODS: The oral carcinoma cell lines E10, SCC-9, and D2 were investigated. Cell migration was studied in a scratch wound assay, and invasion was demonstrated in organotypic three dimensional co-cultures. Protein and mRNA expression of LPA receptors was studied with Western blotting and qRT-PCR. Activation of signalling proteins was examined with Western blotting and isoelectric focusing, and signalling mechanisms were further explored using pharmacological agents and siRNA directed at specific receptors and pathways. RESULTS: LPA stimulated cell migration in the two oral carcinoma cell lines E10 and SCC-9, but was slightly inhibitory in D2. The receptor expression profile and the effects of specific pharmacological antagonist and agonists indicated that LPA-stimulated cell migration was mediated through LPAR3 in E10 and SCC-9. Furthermore, in both these cell lines, the stimulation by LPA was dependent on PKC activity. However, while LPA induced transactivation of EGFR and the stimulated migration was blocked by EGFR inhibitors in E10 cells, LPA did not induce EGFR transactivation in SCC-9 cells. In D2 cells, LPA induced EGFR transactivation, but this was associated with slowing of a very high inherent migration rate in these cells. CONCLUSION: The results demonstrate LPA-stimulated migration in oral carcinoma cells through LPAR3, mediated further by PKC, which acts either in concert with or independently of EGFR transactivation.


Asunto(s)
Carcinoma de Células Escamosas/genética , Receptores ErbB/genética , Neoplasias de la Boca/genética , Receptores del Ácido Lisofosfatídico/biosíntesis , Carcinoma de Células Escamosas/patología , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Receptores ErbB/biosíntesis , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Lisofosfolípidos/administración & dosificación , Neoplasias de la Boca/patología , Proteína Quinasa C/genética , Receptores del Ácido Lisofosfatídico/genética , Transducción de Señal/efectos de los fármacos
6.
BMC Cancer ; 14: 413, 2014 Jun 09.
Artículo en Inglés | MEDLINE | ID: mdl-24912820

RESUMEN

BACKGROUND: Several studies have described an increased cyclooxygenase-2 (COX-2) expression in pancreatic cancer, but the role of COX-2 in tumour development and progression is not clear. The aim of the present study was to examine expression of COX-2 in cancer cells and stromal cells in pancreatic cancer specimens, and to explore the role of PGE2 in pancreatic stellate cell proliferation and collagen synthesis. METHODS: Immunohistochemistry and immunofluorescence was performed on slides from whole sections of tissue blocks using antibodies against COX-2 and α-smooth muscle actin (αSMA). Pancreatic stellate cells (PSC) were isolated from surgically resected tumour tissue by the outgrowth method. Cells were used between passages 4 and 8. Collagen synthesis was determined by [(3)H]-proline incorporation, or by enzyme immunoassay measurement of collagen C-peptide. DNA synthesis was measured by incorporation of [(3)H]-thymidine in DNA. Cyclic AMP (cAMP) was determined by radioimmunoassay. Collagen 1A1 mRNA was determined by RT-qPCR. RESULTS: Immunohistochemistry staining showed COX-2 in pancreatic carcinoma cells, but not in stromal cells. All tumours showed positive staining for αSMA in the fibrotic stroma. Cultured PSC expressed COX-2, which could be further induced by interleukin-1ß (IL-1ß), epidermal growth factor (EGF), thrombin, and PGE2, but not by transforming growth factor-ß1 (TGFß). Indirect coculture with the adenocarcinoma cell line BxPC-3, but not HPAFII or Panc-1, induced COX-2 expression in PSC. Treatment of PSC with PGE2 strongly stimulated cAMP accumulation, mediated by EP2 receptors, and also stimulated phosphorylation of extracellular signal-regulated kinase (ERK). Treatment of PSC with PGE2 or forskolin suppressed both TGFß-stimulated collagen synthesis and PDGF-stimulated DNA synthesis. CONCLUSIONS: The present results show that COX-2 is mainly produced in carcinoma cells and suggest that the cancer cells are the main source of PGE2 in pancreatic tumours. PGE2 exerts a suppressive effect on proliferation and fibrogenesis in pancreatic stellate cells. These effects of PGE2 are mediated by the cAMP pathway and suggest a role of EP2 receptors.


Asunto(s)
Colágeno/biosíntesis , Dinoprostona/farmacología , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/patología , Células Estrelladas Pancreáticas/efectos de los fármacos , Células Estrelladas Pancreáticas/metabolismo , Línea Celular Tumoral , Células Cultivadas , Técnicas de Cocultivo , Ciclooxigenasa 2/genética , Ciclooxigenasa 2/metabolismo , Replicación del ADN/efectos de los fármacos , Expresión Génica , Humanos , Inmunohistoquímica , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas
7.
Nat Commun ; 15(1): 3631, 2024 Apr 29.
Artículo en Inglés | MEDLINE | ID: mdl-38684731

RESUMEN

Idiopathic Parkinson's disease (iPD) is believed to have a heterogeneous pathophysiology, but molecular disease subtypes have not been identified. Here, we show that iPD can be stratified according to the severity of neuronal respiratory complex I (CI) deficiency, and identify two emerging disease subtypes with distinct molecular and clinical profiles. The CI deficient (CI-PD) subtype accounts for approximately a fourth of all cases, and is characterized by anatomically widespread neuronal CI deficiency, a distinct cell type-specific gene expression profile, increased load of neuronal mtDNA deletions, and a predilection for non-tremor dominant motor phenotypes. In contrast, the non-CI deficient (nCI-PD) subtype exhibits no evidence of mitochondrial impairment outside the dopaminergic substantia nigra and has a predilection for a tremor dominant phenotype. These findings constitute a step towards resolving the biological heterogeneity of iPD with implications for both mechanistic understanding and treatment strategies.


Asunto(s)
ADN Mitocondrial , Complejo I de Transporte de Electrón , Complejo I de Transporte de Electrón/deficiencia , Mitocondrias , Enfermedades Mitocondriales , Enfermedad de Parkinson , Enfermedad de Parkinson/genética , Enfermedad de Parkinson/metabolismo , Humanos , Complejo I de Transporte de Electrón/genética , Complejo I de Transporte de Electrón/metabolismo , Enfermedades Mitocondriales/genética , Enfermedades Mitocondriales/metabolismo , Masculino , ADN Mitocondrial/genética , Femenino , Mitocondrias/metabolismo , Mitocondrias/genética , Anciano , Sustancia Negra/metabolismo , Sustancia Negra/patología , Persona de Mediana Edad , Fenotipo , Neuronas/metabolismo
8.
Biochem Biophys Res Commun ; 421(2): 255-60, 2012 May 04.
Artículo en Inglés | MEDLINE | ID: mdl-22503980

RESUMEN

Prostaglandin E(2) (PGE(2)) enhances the mitogenic response to epidermal growth factor (EGF) in hepatocytes, but the underlying mechanisms are not clear. We previously observed that PGE(2) upregulates EGF-induced signalling in the MEK/ERK and PI3K/Akt pathways in hepatocytes. Other investigations have indicated that ErbB2 enhances the mitogenic effect of EGF in these cells. In the present study we found that treatment with PGE(2) increased ErbB2 and decreased ErbB3 expression at both the mRNA and protein level in cultured rat hepatocytes. Silencing of the ErbB2 expression with specific siRNA blocked the stimulation by PGE(2) and EGF of cyclin D1 expression and DNA synthesis. Both EGF and PGE(2) increased the expression of ERK and Akt, but while the effect of EGF was inhibited by ErbB2-directed siRNA, this did not affect the PGE(2)-induced upregulation of ERK and Akt. These data suggest that PGE(2) can enhance the mitogenic effect of EGF both by increasing ErbB2 expression and by ErbB2-independent mechanisms.


Asunto(s)
Dinoprostona/farmacología , Factor de Crecimiento Epidérmico/farmacología , Hepatocitos/efectos de los fármacos , Mitógenos/farmacología , Receptor ErbB-2/fisiología , Animales , Proliferación Celular , Células Cultivadas , Hepatocitos/citología , Hepatocitos/metabolismo , Sistema de Señalización de MAP Quinasas , Proteínas Proto-Oncogénicas c-akt/metabolismo , ARN Interferente Pequeño/genética , Ratas , Receptor ErbB-2/genética , Regulación hacia Arriba
9.
Biomolecules ; 12(6)2022 05 25.
Artículo en Inglés | MEDLINE | ID: mdl-35740871

RESUMEN

Idiopathic Parkinson's disease (iPD) is characterized by degeneration of the dopaminergic substantia nigra pars compacta (SNc), typically in the presence of Lewy pathology (LP) and mitochondrial respiratory complex I (CI) deficiency. LP is driven by α-synuclein aggregation, morphologically evolving from early punctate inclusions to Lewy bodies (LBs). The relationship between α-synuclein aggregation and CI deficiency in iPD is poorly understood. While studies in models suggest they are causally linked, observations in human SNc show that LBs preferentially occur in CI intact neurons. Since LBs are end-results of α-synuclein aggregation, we hypothesized that the relationship between LP and CI deficiency may be better reflected in neurons with early-stage α-synuclein pathology. Using quadruple immunofluorescence in SNc tissue from eight iPD subjects, we assessed the relationship between neuronal CI or CIV deficiency and early or late forms of LP. In agreement with previous findings, we did not observe CI-negative neurons with late LP. In contrast, early LP showed a significant predilection for CI-negative neurons (p = 6.3 × 10-5). CIV deficiency was not associated with LP. Our findings indicate that early α-syn aggregation is associated with CI deficiency in iPD, and suggest a double-hit mechanism, where neurons exhibiting both these pathologies are selectively lost.


Asunto(s)
Enfermedad de Parkinson , Complejo I de Transporte de Electrón/deficiencia , Humanos , Enfermedades Mitocondriales , Neuronas/metabolismo , Enfermedad de Parkinson/patología , Sustancia Negra/metabolismo , alfa-Sinucleína/metabolismo
10.
BMC Cancer ; 11: 421, 2011 Oct 02.
Artículo en Inglés | MEDLINE | ID: mdl-21961726

RESUMEN

BACKGROUND: Neurotensin has been found to promote colon carcinogenesis in rats and mice, and proliferation of human colon carcinoma cell lines, but the mechanisms involved are not clear. We have examined signalling pathways activated by neurotensin in colorectal and pancreatic carcinoma cells. METHODS: Colon carcinoma cell lines HCT116 and HT29 and pancreatic adenocarcinoma cell line Panc-1 were cultured and stimulated with neurotensin or epidermal growth factor (EGF). DNA synthesis was determined by incorporation of radiolabelled thymidine into DNA. Levels and phosphorylation of proteins in signalling pathways were assessed by Western blotting. RESULTS: Neurotensin stimulated the phosphorylation of both extracellular signal-regulated kinase (ERK) and Akt in all three cell lines, but apparently did so through different pathways. In Panc-1 cells, neurotensin-induced phosphorylation of ERK, but not Akt, was dependent on protein kinase C (PKC), whereas an inhibitor of the ß-isoform of phosphoinositide 3-kinase (PI3K), TGX221, abolished neurotensin-induced Akt phosphorylation in these cells, and there was no evidence of EGF receptor (EGFR) transactivation. In HT29 cells, in contrast, the EGFR tyrosine kinase inhibitor gefitinib blocked neurotensin-stimulated phosphorylation of both ERK and Akt, indicating transactivation of EGFR, independently of PKC. In HCT116 cells, neurotensin induced both a PKC-dependent phosphorylation of ERK and a metalloproteinase-mediated transactivation of EGFR that was associated with a gefitinib-sensitive phosphorylation of the downstream adaptor protein Shc. The activation of Akt was also inhibited by gefitinib, but only partly, suggesting a mechanism in addition to EGFR transactivation. Inhibition of PKC blocked neurotensin-induced DNA synthesis in HCT116 cells. CONCLUSIONS: While acting predominantly through PKC in Panc-1 cells and via EGFR transactivation in HT29 cells, neurotensin used both these pathways in HCT116 cells. In these cells, neurotensin-induced activation of ERK and stimulation of DNA synthesis was PKC-dependent, whereas activation of the PI3K/Akt pathway was mediated by stimulation of metalloproteinases and subsequent transactivation of the EGFR. Thus, the data show that the signalling mechanisms mediating the effects of neurotensin involve multiple pathways and are cell-dependent.


Asunto(s)
Neoplasias del Colon/metabolismo , Receptores ErbB/metabolismo , Neurotensina/farmacología , Proteína Quinasa C/metabolismo , Transducción de Señal , Animales , Calcio/metabolismo , Línea Celular Tumoral , Neoplasias del Colon/enzimología , Replicación del ADN/efectos de los fármacos , Activación Enzimática , Células HCT116 , Células HT29 , Humanos , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Proteína Oncogénica v-akt/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Fosforilación/efectos de los fármacos , Transducción de Señal/efectos de los fármacos
11.
Cell Physiol Biochem ; 25(4-5): 523-32, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-20332633

RESUMEN

BACKGROUND/AIMS: Liver regeneration factor 1 (LRF-1/ATF3) is an early response gene which is rapidly induced upon partial hepatectomy in rats, and by growth factors and G protein-coupled receptor (GPCR) agonists in cultured rat hepatocytes. The aim of the present study was to examine the mechanisms involved in induction of LRF-1/ATF3 by the GPCR agonist vasopressin. METHODS: Primary cultures of rat hepatocytes were treated with vasopressin, TPA, and the Ca2+-elevating agents thapsigargin and A23187. LRF-1/ATF3 mRNA and protein were measured by Northern blot analysis or RT-PCR and immunoblotting. Signalling pathways were examined by immunoblots and kinase assays. RESULTS: While elevation of intracellular calcium induced LRF-1/ATF3 expression, treatment with TPA did not. Inhibition of phospholipase C, protein kinase C, or pretreatment with calcium chelators did not affect vasopressin-induced expression of LRF-1/ATF3. Inhibition of each of the MAP kinases ERK1/2, JNK or p38 did not affect vasopressin-induced LRF-1/ATF3 expression. Combined inhibition of JNK and p38, and of ERK1/2 and either JNK or p38 suppressed vasopressin-induced expression of LRF-1/ATF3. CONCLUSION: Vasopressin induces LRF-1/ATF3 expression by mechanisms that differ from those activated by Ca2+-elevating agents. The results suggest that partly redundant, complex MAP kinase networks are involved in induction of LRF-1/ATF3 by vasopressin in hepatocytes.


Asunto(s)
Factor de Transcripción Activador 3/metabolismo , Fármacos Antidiuréticos/farmacología , Hepatocitos/metabolismo , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Vasopresinas/farmacología , Animales , Antracenos/farmacología , Células Cultivadas , Estrenos/farmacología , Proteínas Quinasas JNK Activadas por Mitógenos/antagonistas & inhibidores , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Masculino , Proteínas Quinasas Activadas por Mitógenos/antagonistas & inhibidores , Proteína Quinasa C/metabolismo , Pirrolidinonas/farmacología , Ratas , Ratas Wistar , Transducción de Señal , Acetato de Tetradecanoilforbol/farmacología , Fosfolipasas de Tipo C/metabolismo
12.
Thromb J ; 8(1): 1, 2010 Jan 27.
Artículo en Inglés | MEDLINE | ID: mdl-20181026

RESUMEN

BACKGROUND: The expression of pregnancy-associated plasma protein A (PAPP-A) was identified by immunohistochemistry (IHC) in culprit atherothrombotic plaque specimens harvested from patients admitted with ST-segment elevation myocardial infarction (STEMI). METHODS: The atherothrombotic samples were collected from a consecutive cohort consisting of 20 individuals admitted with STEMI to Stavanger University Hospital, Norway, from 2005-2006, presenting angiographically with an acute thrombotic occlusion of a coronary artery characterized by TIMI flow 0. The atherothrombotic plaques were obtained by aspiration thrombectomy during percutaneous coronary intervention within 12 hours from the onset of symptoms and prepared for IHC analysis. RESULTS: In the IHC analysis staining for PAPP-A occurred in the extracellular matrix of the plaques and no evidence of staining for PAPP-A was found in the thrombi. CONCLUSION: Our results indicate that in vivo PAPP-A is strongly expressed in atherothrombotic plaques harvested from patients admitted with STEMI, as documented by IHC. TRIAL REGISTRATION: biobankregisteret@fhi.no1846.

13.
J Cell Physiol ; 214(2): 371-80, 2008 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-17654493

RESUMEN

Prostaglandins (PGs) such as PGE2 enhance proliferation in many cells, apparently through several distinct mechanisms, including transactivation of the epidermal growth factor (EGF) receptor (EGFR) as well as EGFR-independent pathways. In this study we found that in primary cultures of rat hepatocytes PGE2 did not induce phosphorylation of the EGFR, and the EGFR tyrosine kinase blockers gefitinib and AG1478 did not affect PGE2-stimulated phosphorylation of ERK1/2. In contrast, PGE2 elicited EGFR phosphorylation and EGFR tyrosine kinase inhibitor-sensitive ERK phosphorylation in MH1C1 hepatoma cells. These findings suggest that PGE2 elicits EGFR transactivation in MH1C1 cells but not in hepatocytes. Treatment of the hepatocytes with PGE2 at 3 h after plating amplified the stimulatory effect on DNA synthesis of EGF administered at 24 h and advanced and augmented the cyclin D1 expression in response to EGF in hepatocytes. The pretreatment of the hepatocytes with PGE2 resulted in an increase in the magnitude of EGF-stimulated Akt phosphorylation and ERK1/2 phosphorylation and kinase activity, including an extended duration of the responses, particularly of ERK, to EGF in PGE2-treated cells. Pertussis toxin abolished the ability of PGE2 to enhance the Akt and ERK responses to EGF. The results suggest that in hepatocytes, unlike MH1C1 hepatoma cells, PGE2 does not transactivate the EGFR, but instead acts in synergism with EGF by modulating mitogenic mechanisms downstream of the EGFR. These effects seem to be at least in part G(i) protein-mediated and include upregulation of signaling in the PI3K/Akt and the Ras/ERK pathways.


Asunto(s)
Dinoprostona/farmacología , Factor de Crecimiento Epidérmico/farmacología , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Hepatocitos/efectos de los fármacos , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal/efectos de los fármacos , Animales , Carcinoma Hepatocelular/metabolismo , Carcinoma Hepatocelular/patología , Técnicas de Cultivo de Célula , Células Cultivadas , Medio de Cultivo Libre de Suero , Ciclina D , Ciclinas/metabolismo , ADN/biosíntesis , Dinoprostona/antagonistas & inhibidores , Activación Enzimática/efectos de los fármacos , Receptores ErbB/metabolismo , Quinasas MAP Reguladas por Señal Extracelular/análisis , Quinasas MAP Reguladas por Señal Extracelular/antagonistas & inhibidores , Gefitinib , Hepatocitos/metabolismo , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/patología , Masculino , Toxina del Pertussis/farmacología , Fosforilación/efectos de los fármacos , Inhibidores de Proteínas Quinasas/farmacología , Quinazolinas/farmacología , Ratas , Ratas Wistar , Tirfostinos/farmacología
14.
FEBS Lett ; 581(1): 15-20, 2007 Jan 09.
Artículo en Inglés | MEDLINE | ID: mdl-17174312

RESUMEN

Serotonin activates Ras and Ras-dependent ERK1/2 phosphorylation in HEK293 cells expressing G(s)-coupled 5-HT(4) or 5-HT(7) serotonin receptors through unknown mechanisms. Both Epac/Rap-dependent and -independent pathways for Ras-dependent ERK1/2 activation have been suggested. Epac overexpression or Epac-specific 8-CPT-2'-O-Me-cAMP did not cause ERK1/2 phosphorylation, despite Rap activation. The data did not support a role for PLCepsilon or DAG-dependent Ras GEFs of the Ras-GRP family in Ras-dependent ERK1/2 phosphorylation. However, serotonin stimulated phosphorylation of endogenous and recombinant Ras-GRF1, increased [Ca(2+)](i) and caused Ca(2+)- and calmodulin-dependent ERK1/2 phosphorylation. Different signalling pathways seem to be utilised by G(s)-coupled receptors in various isolates of HEK293 cells.


Asunto(s)
Señalización del Calcio/fisiología , Factores de Intercambio de Guanina Nucleótido/metabolismo , Sistema de Señalización de MAP Quinasas/fisiología , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Procesamiento Proteico-Postraduccional/fisiología , Receptores de Serotonina 5-HT4/metabolismo , Calcio/metabolismo , Señalización del Calcio/efectos de los fármacos , Calmodulina/metabolismo , Línea Celular , AMP Cíclico/análogos & derivados , AMP Cíclico/farmacología , Productos del Gen vpr/genética , Productos del Gen vpr/metabolismo , Factores de Intercambio de Guanina Nucleótido/antagonistas & inhibidores , Factores de Intercambio de Guanina Nucleótido/genética , Humanos , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Procesamiento Proteico-Postraduccional/efectos de los fármacos , Proteínas ras/metabolismo
15.
Toxicology ; 227(1-2): 105-16, 2006 Oct 03.
Artículo en Inglés | MEDLINE | ID: mdl-16963169

RESUMEN

Understanding how mineral particles trigger cellular responses is crucial in order to elucidate what characteristics determine their harmful effects. It is not clear whether cellular effects are triggered through the cell membrane or require particle uptake. However, studies with asbestos suggest that activation of the epidermal growth factor receptor (EGFR) may be important. We have previously reported that crystalline silica-induced interleukin (IL)-8 release from human lung epithelial cells (A549) was regulated through Src family kinases (SFKs) and the mitogen-activated protein kinases (MAPKs) p38 and extracellular signal-regulated kinase (ERK)-1 and -2. The present study shows that SFK and p38 phosphorylation increased almost immediately upon crystalline silica exposure, whereas ERK1/2 phosphorylation increased after 10 min of exposure. The p38 inhibitor SB202190 increased the silica-induced ERK1/2 phosphorylation suggesting that p38 activity may attenuate activation of ERK1/2. Scanning electron microscopy showed that some silica particles were phagocytosed between 1 and 4h of exposure, but that the majority remained bound by microvilli on the cell surface. The EGFR inhibitor AG1478 attenuated both silica-induced IL-8 release and phosphorylation of SFKs and ERK1/2. However, AG1478 also inhibited the respective background levels, and the EGFR was not phosphorylated at the onset of silica exposure. The results suggest that crystalline silica triggers p38 and SFK-ERK1/2 signaling through interactions with membrane components as both pathways were rapidly activated prior to particle internalization. However, the silica-induced up-regulation of IL-8 release through the SFK-ERK1/2 pathway does not appear to be initiated through activation of the EGFR, although basal EGFR activity may affect the magnitude of the responses.


Asunto(s)
Células Epiteliales/efectos de los fármacos , Receptores ErbB/metabolismo , Interleucina-8/metabolismo , Proteínas Quinasas/metabolismo , Dióxido de Silicio/toxicidad , Línea Celular , Células Epiteliales/metabolismo , Células Epiteliales/ultraestructura , Humanos , Tamaño de la Partícula , Fosforilación , Transducción de Señal/efectos de los fármacos
16.
Neoplasia ; 18(7): 447-56, 2016 07.
Artículo en Inglés | MEDLINE | ID: mdl-27435927

RESUMEN

Pancreatic ductal adenocarcinoma is characterized by a prominent fibroinflammatory stroma with both tumor-promoting and tumor-suppressive functions. The pancreatic stellate cell (PSC) is the major cellular stromal component and the main producer of extracellular matrix proteins, including collagens, which are degraded by metalloproteinases (MMPs). PSCs interact with cancer cells through various factors, including transforming growth factor (TGF)ß and interleukin (IL)-1α. The role of TGFß in the dual nature of tumor stroma, i.e., protumorigenic or tumor suppressive, is not clear. We aimed to investigate the roles of TGFß and IL-1α in the regulation of MMP profiles in PSCs and the subsequent effects on cancer cell migration. Human PSCs isolated from surgically resected specimens were cultured in the presence of pancreatic cancer cell lines, as well as IL-1α or TGFß. MMP production and activities in PSCs were quantified by gene array transcripts, mRNA measurements, fluorescence resonance energy transfer-based activity assay, and zymography. PSC-conditioned media and pancreatic cancer cells were included in a collagen matrix cell migration model. We found that production of IL-1α by pancreatic cancer cells induced alterations in MMP and tissue inhibitors of matrix metalloproteinase (TIMP) profiles and activities in PSCs, upregulated expression and activation of MMP1 and MMP3, and enhanced migration of pancreatic cancer cells in the collagen matrix model. TGFß counteracted the effects of IL-1α on PSCs, reestablished PSC MMP and TIMP profiles and activities, and inhibited migration of cancer cells. This suggests that tumor TGFß has a role as a suppressor of stromal promotion of tumor progression through alterations in PSC MMP profiles with subsequent inhibition of pancreatic cancer cell migration.


Asunto(s)
Carcinoma Ductal Pancreático/patología , Interleucina-1alfa/farmacología , Metaloproteinasa 1 de la Matriz/metabolismo , Metaloproteinasa 2 de la Matriz/metabolismo , Metaloproteinasa 3 de la Matriz/metabolismo , Neoplasias Pancreáticas/patología , Células Estrelladas Pancreáticas/metabolismo , Inhibidor Tisular de Metaloproteinasa-1/metabolismo , Inhibidor Tisular de Metaloproteinasa-2/metabolismo , Inhibidor Tisular de Metaloproteinasa-3/metabolismo , Factor de Crecimiento Transformador beta/farmacología , Línea Celular Tumoral , Movimiento Celular , Activación Enzimática/efectos de los fármacos , Transferencia Resonante de Energía de Fluorescencia , Humanos , Interleucina-1alfa/metabolismo , Conductos Pancreáticos/patología , Proteínas Supresoras de Tumor/metabolismo
17.
J Exp Clin Cancer Res ; 35(1): 122, 2016 07 29.
Artículo en Inglés | MEDLINE | ID: mdl-27473228

RESUMEN

BACKGROUND: The most abundant cells in the extensive desmoplastic stroma of pancreatic adenocarcinomas are the pancreatic stellate cells, which interact with the carcinoma cells and strongly influence the progression of the cancer. Tumor stroma interactions induced by IL-1α/IL-1R1 signaling have been shown to be involved in pancreatic cancer cell migration. TGFß and its receptors are overexpressed in pancreatic adenocarcinomas. We aimed at exploring TGFß and IL-1α signaling and cross-talk in the stellate cell cancer cell interactions regulating pancreatic adenocarcinoma cell migration. METHODS: Human pancreatic stellate cells were isolated from surgically resected pancreatic adenocarcinomas and cultured in the presence of TGFß or pancreatic adenocarcinoma cell lines. The effects of TGFß were blocked by inhibitors or amplified by silencing the endogenous inhibitor of SMAD signaling, SMAD7. Pancreatic stellate cell responses to IL-1α or to IL-1α-expressing pancreatic adenocarcinoma cells (BxPC-3) were characterized by their ability to stimulate migration of cancer cells in a 2D migration model. RESULTS: In pancreatic stellate cells, IL-1R1 expression was found to be down-regulated by TGFß and blocking of TGFß signaling re-established the expression. Endogenous inhibition of TGFß signaling by SMAD7 was found to correlate with the levels of IL-1R1, indicating a regulatory role of SMAD7 in IL-1R1 expression. Pancreatic stellate cells cultured in the presence of IL-1α or in co-cultures with BxPC-3 cells enhanced the migration of cancer cells. This effect was blocked after treatment of the pancreatic stellate cells with TGFß. Silencing of stellate cell expression of SMAD7 was found to suppress the levels of IL-1R1 and reduce the stimulatory effects of IL-1α, thus inhibiting the capacity of pancreatic stellate cells to induce cancer cell migration. CONCLUSIONS: TGFß signaling suppressed IL-1α mediated pancreatic stellate cell induced carcinoma cell migration. Depletion of SMAD7 upregulated the effects of TGFß and reduced the expression of IL-1R1, leading to inhibition of IL-1α induced stellate cell enhancement of carcinoma cell migration. SMAD7 might represent a target for inhibition of IL-1α induced tumor stroma interactions.


Asunto(s)
Interleucina-1alfa/metabolismo , Neoplasias Pancreáticas/metabolismo , Células Estrelladas Pancreáticas/citología , Proteína smad3/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Comunicación Celular , Línea Celular Tumoral , Movimiento Celular , Células Cultivadas , Técnicas de Cocultivo , Regulación Neoplásica de la Expresión Génica , Humanos , Interleucina-1alfa/genética , Neoplasias Pancreáticas/patología , Células Estrelladas Pancreáticas/metabolismo , Receptores Tipo I de Interleucina-1/genética , Receptores Tipo I de Interleucina-1/metabolismo , Transducción de Señal , Proteína smad3/genética , Proteína smad7/genética , Proteína smad7/metabolismo , Neoplasias Pancreáticas
18.
BMC Cell Biol ; 3: 5, 2002.
Artículo en Inglés | MEDLINE | ID: mdl-11914123

RESUMEN

BACKGROUND: Previous studies have shown that several agents that stimulate heptahelical G-protein coupled receptors activate the extracellular signal regulated kinases ERK1 (p44mapk) and ERK2 (p42mapk) in hepatocytes. The molecular pathways that convey their signals to ERK1/2 are only partially clarified. In the present study we have explored the role of Ca2+ and Ca2+-dependent steps leading to ERK1/2 activation induced by norepinephrine and prostaglandin (PG)F2alpha. RESULTS: Pretreatment of the cells with the Ca2+ chelators BAPTA-AM or EGTA, as well as the Ca2+ influx inhibitor gadolinium, resulted in a partial decrease of the ERK response. Furthermore, the calmodulin antagonists W-7, trifluoperazine, and J-8 markedly decreased ERK activation. Pretreatment with KN-93, an inhibitor of the multifunctional Ca2+/calmodulin-dependent protein kinase, had no effect on ERK activation. The Src kinase inhibitors PP1 and PP2 partially diminished the ERK responses elicited by both norepinephrine and PGF2alpha. CONCLUSION: The present data indicate that Ca2+ is involved in ERK activation induced by hormones acting on G protein-coupled receptors in hepatocytes, and suggest that calmodulin and Src kinases might play a role in these signaling pathways.


Asunto(s)
Calcio/fisiología , Calmodulina/fisiología , Dinoprost/farmacología , Hepatocitos/efectos de los fármacos , Hepatocitos/enzimología , Proteína Quinasa 1 Activada por Mitógenos/fisiología , Proteínas Quinasas Activadas por Mitógenos/fisiología , Norepinefrina/farmacología , Familia-src Quinasas/fisiología , Animales , Calcimicina/farmacología , Proteínas Quinasas Dependientes de Calcio-Calmodulina/antagonistas & inhibidores , Proteínas Quinasas Dependientes de Calcio-Calmodulina/fisiología , Calmodulina/antagonistas & inhibidores , Activación Enzimática/efectos de los fármacos , Activación Enzimática/fisiología , Ionóforos/farmacología , Sistema de Señalización de MAP Quinasas/fisiología , Masculino , Proteína Quinasa 3 Activada por Mitógenos , Ratas , Ratas Wistar , Familia-src Quinasas/antagonistas & inhibidores
19.
Eur J Pharmacol ; 481(1): 83-9, 2003 Nov 14.
Artículo en Inglés | MEDLINE | ID: mdl-14637179

RESUMEN

Carvedilol is a combined alpha(1)- and beta-adrenoceptor antagonist. The ability of carvedilol to antagonize functional effects mediated through myocardial alpha(1)-adrenoceptors has never been investigated. We tested the ability of carvedilol to antagonize the inotropic effect mediated by myocardial alpha(1)-adrenoceptors compared to the antagonism of beta-adrenoceptors. Papillary muscles from rat heart left ventricle were mounted in an organ bath and concentration-response experiments for the inotropic effects of separate alpha(1)- and beta-adrenoceptor stimulation were performed in the absence and presence of carvedilol. Carvedilol antagonized myocardial alpha(1)-adrenoceptors with an inhibition constant (K(i)) of 11.0+/-3.0 nmol/l and the functional experiments were supported by radioligand-binding studies. Corresponding functional studies on the response to beta-adrenoceptor stimulation revealed a K(i) of 1.2+/-0.35 nmol/l. Thus, carvedilol antagonizes the myocardial alpha(1)-adrenoceptors with a 9-fold lower potency than the beta-adrenoceptors. Antagonism of myocardial alpha(1)-adrenoceptor evoked effects may contribute to clinical effects of carvedilol.


Asunto(s)
Antagonistas de Receptores Adrenérgicos alfa 1 , Antagonistas Adrenérgicos alfa/farmacología , Carbazoles/farmacología , Contracción Miocárdica/efectos de los fármacos , Propanolaminas/farmacología , Agonistas alfa-Adrenérgicos/farmacología , Antagonistas Adrenérgicos beta/farmacología , Animales , Unión Competitiva/efectos de los fármacos , Carvedilol , Relación Dosis-Respuesta a Droga , Técnicas In Vitro , Masculino , Contracción Muscular/efectos de los fármacos , Norepinefrina/farmacología , Músculos Papilares/efectos de los fármacos , Músculos Papilares/fisiología , Fenilefrina/farmacología , Prazosina/metabolismo , Ensayo de Unión Radioligante , Ratas , Ratas Wistar , Receptores Adrenérgicos alfa 1/metabolismo , Receptores Adrenérgicos beta/efectos de los fármacos , Receptores Adrenérgicos beta/fisiología , Tritio
20.
Basic Clin Pharmacol Toxicol ; 95(6): 288-94, 2004 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-15569274

RESUMEN

Various cells and tissues contain high basal levels of inositol 1,4,5-trisphosphate, raising questions about the functional significance of inositol 1,4,5-trisphosphate in some tissues such as the heart. We used intact tissue and isolated cells from heart and liver of adult rats to examine if different fixation procedures might artificially elevate the level of inositol 1,4,5-trisphosphate. The basal level of inositol 1,4,5-trisphosphate in intact, freeze-clamped cardiac tissue from adult rats was 10 times higher than in isolated, non-frozen cardiomyocytes, while freeze-clamped liver contained approximately 4 times higher inositol 1,4,5-trisphosphate levels than isolated, non-frozen hepatocytes. Stimulation with norepinephrine induced a significant increase in the inositol 1,4,5-trisphosphate level in isolated cardiomyocytes, whereas no significant increase was observed in freeze-clamped cardiac tissue. Freezing of isolated cardiomyocytes or hepatocytes before extraction increased basal inositol 1,4,5-trisphosphate levels 3 times. In cellular homogenates prepared in the presence of EGTA and stored at 4 degrees , readdition of calcium resulted in a time-dependent increase in inositol 1,4,5-trisphosphate mass and a decrease in the mass of phosphatidylinositol 4,5-bisphosphate (PIP(2)). The reaction was essentially complete within 30 sec. in homogenates from cardiomyocytes, while PIP(2) hydrolysis was slower in hepatocyte homogenates. Perfusion of intact rat hearts with EGTA present during the last 2 min. of perfusion, followed by freeze-clamping, resulted in basal inositol 1,4,5-trisphosphate levels comparable to those in isolated cardiomyocytes, and norepinephrine stimulation increased inositol 1,4,5-trisphosphate mass by approximately 80%. The presence of EGTA did not significantly affect PIP(2) levels in perfused hearts. The results suggest that freezing or homogenization of intact tissue and isolated cells may result in Ca(2+)-dependent activation of phospholipase C, leading to high basal inositol 1,4,5-trisphosphate levels that may mask agonist-induced changes.


Asunto(s)
Criopreservación , Hepatocitos , Inositol 1,4,5-Trifosfato/metabolismo , Hígado , Miocardio , Miocitos Cardíacos , Animales , Artefactos , Calcio/metabolismo , Inositol 1,4,5-Trifosfato/aislamiento & purificación , Masculino , Ratas , Ratas Wistar , Fosfolipasas de Tipo C/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA