Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 72
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Odontology ; 111(3): 565-572, 2023 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-36396928

RESUMEN

In the present study, expression and localization of PLD2 were examined in mouse submandibular glands in situ in comparison with those of PLD1 previously reported by the present authors. In immunoblots, PLD2-expression was high at postnatal 0 week (P0W) and P2W, and at P4W decreased considerably with the decrease of the male gland more marked, and at P8W it was undetectable in the male gland, but remained faint in the female. In the male gland at P8W after castration at P6W, PLD2-expression reappeared faintly. In the female glands with daily injections of testosterone started at P6W, at P8W the expression was undetectable. In immunohistochemistry, PLD2 was localized throughout immature duct epithelia without distinct regional differences in its intensity at P0W and P2W. PLD2-localization was weak and diffuse in granular convoluted tubules at P4W and negligible there at P8W in the female gland, while it was at negligible levels in the tubules at P4W and at undetectable levels there at P8W in the male gland. The expression changes detected in immunoblots after castration or testosterone injection were duplicated in granular convoluted tubules in immunohistochemistry. The present findings suggest that PLD2 is dominantly involved in proliferation/differentiation/apoptosis of most immature ductal cells at early postnatal and at least perinatal stages, different from PLD1. This also indicates that the female-dominant sexual dimorphism of PLD2-expression occurs in synchrony of differentiation of granular convoluted tubules at puberty and at young adulthood, similar to PLD1 though at much lower levels.


Asunto(s)
Fosfolipasa D , Ratones , Animales , Masculino , Femenino , Fosfolipasa D/metabolismo , Glándula Submandibular/metabolismo , Caracteres Sexuales , Testosterona/farmacología , Testosterona/metabolismo , Diferenciación Celular
2.
J Anat ; 235(6): 1125-1136, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31402458

RESUMEN

To clarify the signal transduction mechanism in the differentiation and secretion of salivary glandular cells, the present study was attempted to examine in the submandibular gland (SMG) of mice, the expression and localization of phospholipase D1 (PLD1), one of the important effector molecules working in response to the activation of intramembranous receptors by first messengers. In immunoblotting analysis, the expression of PLD1 was high at postnatal 4 weeks (P4W) and decreased at P8W, and it was at negligible levels at newborn stage (P0W) and postnatal 2 weeks (P2W). The expression of PLD1 was greater in females, and it was suppressed by administration of testosterone to female mice. In immuno-light microscopy, immunoreactivity for PLD1 at P4W was moderate to intense, in the forms of dots and globules mainly in the apical domains of immature granular convoluted tubule (GCT)-cells localized largely in the proximal portion of the female GCT. By P8W, it decreased in intensity and remained weak to moderate along the apical plasmalemma of cells throughout the course of the female GCT, whereas it was faint throughout the GCT of the male SMG at P4W and negligible at P8W. In immuno-electron microscopy, immature GCT-cells characterized by electron-lucent granules were immunoreactive and the immunoreactive materials were deposited close to, but not within, those granules. Typical GCT cells, characterized by electron-dense granules, were immunonegative. No significant immunoreaction for PLD1 was seen in acini of SMGs of either sex at any time point examined. It is suggested that PLD1 is involved in the signaling for secretion of immature GCT cells and influences differentiation of these cells, probably through their own secretory substances.


Asunto(s)
Fosfolipasa D/metabolismo , Glándula Submandibular/metabolismo , Testosterona/farmacología , Animales , Diferenciación Celular/fisiología , Femenino , Masculino , Ratones , Factores Sexuales , Transducción de Señal/fisiología
3.
Anticancer Drugs ; 24(5): 473-83, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23426175

RESUMEN

Our previous study has shown that the activity and expression of sphingosine kinase (SPHK) regulated the sensitivity of human colon cancer cells to the chemotherapeutic oxaliplatin (L-OHP). In addition, the cancer stem cell marker CD44 increases cell resistance to anticancer drugs. Here, we use colon cancer cell lines to examine the relationship between SPHK1 activity and CD44 expression.CD44 expression was measured by western blotting and quantitative PCR in two human colon cancer cell lines: L-OHP-resistant RKO and L-OHP-sensitive HCT116. The regulation of CD44 by SPHK1 was examined by either blocking or overexpressing SPHK1 and by using an L-OHP-resistant HCT116 clone (HCT116-R).The levels of SPHK1, CD44, phosphorylated-Akt, and phosphorylated-extracellular signal-regulated kinase (ERK) were much higher in the RKO cells than in the HCT116 cells. The treatment of RKO cells with the SPHK inhibitor or SPHK1 silencing by RNA interference suppressed CD44 protein expression. SPHK1 and CD44 levels were much higher in HCT116-R cells compared with the parental HCT116 cells. Transfection of HCT116 cells with SPHK1 cDNA enhanced the expression of both CD44 and phosphorylated-ERK. The increase in the CD44 protein level was abolished by the inhibition of ERK phosphorylation. Treatment of RKO cells with the sphingosine-1-phosphate (S1P)2 receptor antagonist suppressed ERK phosphorylation and the expression of CD44 mRNA and protein. Exogenous stimulation with S1P increased ERK phosphorylation and CD44 protein expression in HCT116 cells, but treatment with an MEK inhibitor and S1P2 receptor antagonist blocked this effect.These findings indicate that SPHK1 and its product, S1P, contribute toward the regulation of CD44 protein expression through the ERK signaling pathway through S1P2 in human colon cancer cells.


Asunto(s)
Neoplasias del Colon/metabolismo , Resistencia a Antineoplásicos/efectos de los fármacos , Receptores de Hialuranos/metabolismo , Fosfotransferasas (Aceptor de Grupo Alcohol)/metabolismo , Línea Celular Tumoral , Neoplasias del Colon/patología , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Células HCT116/efectos de los fármacos , Humanos , Receptores de Hialuranos/genética , Lisofosfolípidos/metabolismo , Compuestos Organoplatinos/farmacología , Oxaliplatino , Fosforilación , Fosfotransferasas (Aceptor de Grupo Alcohol)/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal , Esfingosina/análogos & derivados , Esfingosina/metabolismo , Regulación hacia Arriba
4.
Biol Pharm Bull ; 36(7): 1204-7, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23811570

RESUMEN

FLJ00018, a heterotrimeric guanosine 5'-triphosphate (GTP)-binding protein (G protein) Gßγ subunit-activated guanine nucleotide exchange factor for Rho family small GTPases, regulates cellular responses, including cell morphological changes and gene transcriptional regulation, and targets the cellular membranes. FLJ00018 contains a Dbl homology (DH) domain in addition to a pleckstrin homology (PH) domain. Here we show that the PH domain of FLJ00018 is required for FLJ00018-induced, serum response element-dependent gene transcription. Although the PH domain of KIAA1415/P-Rex1, another Gßγ subunit-activated guanine nucleotide exchange factor for Rho family small GTPases, binds to phosphatidylinositol 3,4,5-triphosphate and phosphatidylinositol 3,4-bisphosphate, the PH domain of FLJ00018 binds to polyphosphoinositides including phosphatidylinositol 4,5-bisphosphate, and phosphatidic acid. These results suggest that FLJ00018 is targeted via its PH domain to cellular membranes.


Asunto(s)
Membrana Celular/metabolismo , Factores de Intercambio de Guanina Nucleótido/metabolismo , Lípidos de la Membrana/metabolismo , Fraccionamiento Celular , ADN Complementario/genética , Factores de Intercambio de Guanina Nucleótido/genética , Células HEK293 , Humanos , Luciferasas/genética , Plásmidos , Unión Proteica , Factores de Intercambio de Guanina Nucleótido Rho , Elemento de Respuesta al Suero/fisiología , Transcripción Genética
5.
Biochim Biophys Acta ; 1811(3): 119-28, 2011 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-21184844

RESUMEN

The role of sphingolipid metabolic pathway has been recognized in determining cellular fate. Although sphingolipid degradation has been extensively studied, gene expression of human sphingosine 1-phosphate lyase (SPL) catalyzing sphingosine 1-phosphate (S1P) remains to be determined. Among 5 human lung cancer cell lines examined, SPL protein levels paralleled the respective mRNA and enzyme activities. Between H1155 and H1299 cells used for further experiments, higher cellular S1P was observed in H1155 with higher SPL activity compared with H1299 with low SPL activity. GATA-4 has been reported to affect SPL transcription in Dictyostelium discoideum. GATA-4 was observed in H1155 but not in other cell lines. Overexpression of GATA-4 in H1299 increased SPL expression. However, promoter analysis of human SPL revealed that the most important region was located between -136bp and -88bp from the first exon, where 2 Sp1 sites exist but no GATA site. DNA pull-down assay of H1155 showed increased DNA binding of Sp1 and GATA-4 within this promoter region compared with H1299. Electrophoresis mobility shift assay (EMSA), chromatin immunoprecipitation (ChIP) assay, reporter assay using mutated binding motif, and mithramycin A, a specific Sp1 inhibitor, suggest the major role of Sp1 in SPL transcription and no direct binding of GATA-4 with this 5' promoter region. The collaborative role of GATA-4 was proved by showing coimmunoprecipitation of Sp1 and GATA-4 using GST-Sp1 and overexpressed GATA-4. Thus, high SPL transcription of H1155 cells was regulated by Sp1 and GATA-4/Sp1 complex formation, both of which bind to Sp1 sites of the 5'-SPL promoter.


Asunto(s)
Aldehído-Liasas/biosíntesis , Regulación Enzimológica de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Neoplasias Pulmonares/enzimología , Proteínas de Neoplasias/biosíntesis , Transcripción Genética , Aldehído-Liasas/genética , Línea Celular Tumoral , Dictyostelium/genética , Factor de Transcripción GATA4/genética , Factor de Transcripción GATA4/metabolismo , Expresión Génica , Humanos , Neoplasias Pulmonares/genética , Proteínas de Neoplasias/genética , Proteínas Protozoarias/biosíntesis , Proteínas Protozoarias/genética , Elementos de Respuesta , Factor de Transcripción Sp1/genética , Factor de Transcripción Sp1/metabolismo
6.
FASEB J ; 25(4): 1133-44, 2011 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-21163859

RESUMEN

Acid sphingomyelinase (ASM) regulates the homeostasis of sphingolipids, including ceramides and sphingosine-1-phosphate (S1P). Because sphingolipids regulate AKT activation, we investigated the role of ASM in hepatic glucose and lipid metabolism. Initially, we overexpressed ASM in the livers of wild-type and diabetic db/db mice by adenovirus vector (Ad5ASM). In these mice, glucose tolerance was improved, and glycogen and lipid accumulation in the liver were increased. Using primary cultured hepatocytes, we confirmed that ASM increased glucose uptake, glycogen deposition, and lipid accumulation through activation of AKT and glycogen synthase kinase-3ß. In addition, ASM induced up-regulation of glucose transporter 2 accompanied by suppression of AMP-activated protein kinase (AMPK) phosphorylation. Loss of sphingosine kinase-1 (SphK1) diminished ASM-mediated AKT phosphorylation, but exogenous S1P induced AKT activation in hepatocytes. In contrast, SphK1 deficiency did not affect AMPK activation. These results suggest that the SphK/S1P pathway is required for ASM-mediated AKT activation but not for AMPK inactivation. Finally, we found that treatment with high-dose glucose increased glycogen deposition and lipid accumulation in wild-type hepatocytes but not in ASM(-/-) cells. This result is consistent with glucose intolerance in ASM(-/-) mice. In conclusion, ASM modulates AKT activation and AMPK inactivation, thus regulating glucose and lipid metabolism in the liver.


Asunto(s)
Proteínas Quinasas Activadas por AMP/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Esfingomielina Fosfodiesterasa/fisiología , Animales , Glucemia/metabolismo , Glucosa/metabolismo , Transportador de Glucosa de Tipo 2/biosíntesis , Glucógeno/metabolismo , Hepatocitos/metabolismo , Metabolismo de los Lípidos/genética , Masculino , Ratones , Ratas
7.
J Cell Biochem ; 112(11): 3449-58, 2011 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-21769916

RESUMEN

Glial cell line-derived neurotrophic factor (GDNF) is important for the development and maintenance of dopamine neurons (Lin et al. [1993] Science 260: 1130-1132). GDNF is neuroprotective in animal models of Parkinson disease, where dopamine neurons show selective degeneration. We previously reported GDNF-induced SPHK1 gene expression in a neuroblastoma cell line, TGW (Murakami et al. [2007] J Neurochem 102: 1585-1594). In the present study, we focused on the regulatory mechanism of GAP43 (GDNF-induced neuronal phenotype) transcription to further elucidate physiological roles of GDNF-induced SPHK1 expression and activity. Stable wild-type (SPHK1-WT) but not dominant-negative SPHK1 (SPHK1-DN) overexpression increased both control- and GDNF-induced GAP43 expression. SPHK1-WT cells showed enhanced GDNF-induced sphingosine 1-phosphate (S1P) secretion compared with mock- and SPHK1-DN cells. Exogenous S1P also increased GAP43 expression. In TGW cells, PD98059, a MEK inhibitor, but not SB203580 (a p38 MAPK inhibitor) and LY294002 (a PI3K inhibitor) inhibited GDNF-induced GAP43 expression, suggesting the MEK/ERK pathway has a major role in GDNF-induced GAP43 transcription. A G-protein-coupled receptor inhibitor, pertussis toxin, and S1P(1) and S1P(3) receptor antagonists (VPC23019 and CAY10444) also inhibited ERK activation. Moreover, both S1P1 and S1P3 were serine-phosphorylated by GDNF, suggesting their activated states. C/EBPß transcription factor was induced by GDNF, and DNA pull-down and chromatin immunoprecipitation assays revealed the C/EBP binding site between -131 bp and -98 bp from the first exon of GAP43. Taken together, our results showed that in TGW cells, GDNF increased SPHK1 transcription, leading to the production and secretion of S1P. Through MEK/ERK pathway, S1P stimulates GAP43 transcription with increased binding of C/EBPß to the 5'-promoter.


Asunto(s)
Proteína GAP-43/genética , Factor Neurotrófico Derivado de la Línea Celular Glial/metabolismo , Lisofosfolípidos/metabolismo , Fosfotransferasas (Aceptor de Grupo Alcohol)/metabolismo , Esfingosina/análogos & derivados , Transcripción Genética , Secuencia de Bases , Western Blotting , Línea Celular Tumoral , Inmunoprecipitación de Cromatina , Clonación Molecular , Cartilla de ADN , Humanos , Reacción en Cadena de la Polimerasa , Regiones Promotoras Genéticas , Transducción de Señal , Esfingosina/metabolismo
8.
Biochem Biophys Res Commun ; 415(1): 168-73, 2011 Nov 11.
Artículo en Inglés | MEDLINE | ID: mdl-22033413

RESUMEN

Rho family GTPase-specific guanine nucleotide exchange factors of the Dbl family regulate a variety of cellular events including cytoskeletal arrangement, signal transduction, and gene expression through activation of Rho family GTPases. In this study, we show that hPEM-2 is a downstream effector of G(s) and G(q) signaling in Neuro-2a neuroblastoma cells. Co-expression with hPEM-2 and GTPase-deficient (constitutively active) mutants of Gαs (Gα(s)Q213L) or Gα(q) (Gα(q)Q209L), but not other GTPase-deficient mutants of Gα subunit and Gßγ subunits, activated serum response element (SRE)-dependent gene transcription, which is known to be induced by Rho family activation. Although a dominant negative mutant of Rac1 strongly blocks Gα(s)Q213L or Gα(q)Q209L/hPEM-2 activated SRE-dependent gene transcription, those of Cdc42 or RhoA are marginally affected. A PKA inhibitor, H-89, attenuated Gα(s)/hPEM-2-activated SRE-dependent gene transcription. And a dominant negative mutant of c-Src and an Src inhibitor attenuated Gα(q)Q209L/hPEM-2-activated SRE-dependent gene transcription. Experiments using hPEM-2 deletion mutants indicate that some regions of hPEM-2 play an important role in enhancing SRE activation by G(s) and G(q) signalings. These results reveal that G(s) and G(q) signalings regulate hPEM-2 functions through PKA and c-Src in Neuro-2a neuroblastoma cells, respectively.


Asunto(s)
Subunidades alfa de la Proteína de Unión al GTP Gq-G11/metabolismo , Subunidades alfa de la Proteína de Unión al GTP Gs/metabolismo , Factores de Intercambio de Guanina Nucleótido/metabolismo , Animales , Proteína Tirosina Quinasa CSK , Línea Celular Tumoral , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Factores de Intercambio de Guanina Nucleótido/genética , Humanos , Ratones , Proteínas Tirosina Quinasas/metabolismo , Factores de Intercambio de Guanina Nucleótido Rho , Transcripción Genética , Familia-src Quinasas
9.
Chem Res Toxicol ; 24(11): 1845-52, 2011 Nov 21.
Artículo en Inglés | MEDLINE | ID: mdl-21981408

RESUMEN

The antineoplastic efficacy of oxaliplatin, a widely used anticancer drug, is restricted by its adverse effects such as peripheral neuropathy. Infusing a combination of calcium gluconate and magnesium sulfate (Ca/Mg) suppresses the acute neurotoxic side effects of oxaliplatin, although the mechanism is unclear. To elucidate the molecular mechanisms of oxaliplatin-induced neurotoxicity and the effects of Ca/Mg against this toxicity, we examined the effect of Ca/Mg on oxaliplatin-induced inhibition of neurite outgrowth in PC12 cells, a commonly used neuronal cell model. Oxaliplatin and oxalate suppressed nerve growth factor (NGF)-induced neurite outgrowth and reduced the NGF-mediated increase in the intracellular calcium concentration [Ca(2+)](i). A calcium-chelating agent, BAPTA/AM, also exhibited similar inhibitory effects on neurite outgrowth and [Ca(2+)](i). The addition of Ca/Mg attenuated these inhibitions induced by oxaliplatin and oxalate. The NGF-induced upregulation of growth-associated protein-43 (GAP-43) was suppressed by oxaliplatin and oxalate. Oxaliplatin, but not oxalate, suppressed NGF-stimulated extracellular signal-regulated kinase activation, and this inhibition was not affected by Ca/Mg. Ca/Mg did not modify the oxaliplatin-induced loss of cell viability or apoptosis in PC12 or HCT-116 cells, a human colorectal cancer cell line. These results suggest that the inhibition of neurite outgrowth but not tumor cell death induced by oxaliplatin is partly associated with reductions in [Ca(2+)](i) and GAP-43 expression, and this inhibition was suppressed by the addition of Ca/Mg. Therefore, it may be assumed that Ca/Mg is useful for protecting against oxaliplatin-induced neurotoxicity without reducing the antitumor activity of oxaliplatin.


Asunto(s)
Gluconato de Calcio/uso terapéutico , Calcio/metabolismo , Sulfato de Magnesio/uso terapéutico , Neoplasias/metabolismo , Neuritas/efectos de los fármacos , Compuestos Organoplatinos/farmacología , Animales , Antineoplásicos/efectos adversos , Antineoplásicos/farmacología , Gluconato de Calcio/metabolismo , Gluconato de Calcio/farmacología , Muerte Celular/efectos de los fármacos , Diferenciación Celular/efectos de los fármacos , Quelantes/metabolismo , Quelantes/farmacología , Ácido Egtácico/análogos & derivados , Ácido Egtácico/metabolismo , Ácido Egtácico/farmacología , Quinasas MAP Reguladas por Señal Extracelular/genética , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Proteína GAP-43/genética , Proteína GAP-43/metabolismo , Expresión Génica , Células HCT116 , Humanos , Sulfato de Magnesio/metabolismo , Sulfato de Magnesio/farmacología , Neoplasias/tratamiento farmacológico , Neoplasias/patología , Factor de Crecimiento Nervioso/farmacología , Neuritas/metabolismo , Compuestos Organoplatinos/efectos adversos , Ácido Oxálico/metabolismo , Oxaliplatino , Células PC12 , Ratas , Transducción de Señal
10.
Neurochem Res ; 36(7): 1304-13, 2011 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-21369758

RESUMEN

Caspase plays an important role in apoptosis and physiological processes such as synaptic plasticity. However, the caspase substrate at the synapse is still unknown. Here we used an in vitro cleavage assay with a small-pool human brain cDNA library. We identified the presynaptic protein Caytaxin as a substrate of caspase-3 and caspase-7. Deficiency in Caytaxin causes Cayman ataxia, a disorder characterized by cerebellar dysfunction and mental retardation. Caytaxin cleavage in cerebellar granule neurons is dependent on caspase-3 activation. The cleavage site is upstream of the cellular retinal and the TRIO guanine exchange factor domain, producing a C-terminal fragment that may play an alternative role in inhibiting MEK2 signaling. Thus, we concluded that Caytaxin is a novel substrate of caspase-3 at the presynapse.


Asunto(s)
Caspasa 3/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Secuencia de Aminoácidos , Animales , Caspasa 7/metabolismo , Línea Celular , Humanos , MAP Quinasa Quinasa 2/fisiología , Ratones , Datos de Secuencia Molecular , Alineación de Secuencia , Transducción de Señal , Sinapsis/metabolismo
11.
Anticancer Drugs ; 22(5): 402-8, 2011 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-21317765

RESUMEN

The human aldo-keto reductase (AKR) 1B10 is suggested as a tumor marker in various solid tumors. Using colon cancer cells, we found that AKR1B10 was induced with acquisition of resistance to the anticancer drug mitomycin-c (MMC). In the resistant cells, treatment with an AKR1B10 inhibitor decreased their MMC tolerance. In the nonresistant cells, overexpression and silencing of AKR1B10 decreased and increased, respectively, susceptibility to cytotoxic effects of MMC and 4-hydroxy-2-nonenal, which was formed as a product of lipid peroxidation by MMC treatment. These results suggest a role of AKR1B10 in the development of MMC resistance, which may be mediated by its ability to detoxify cytotoxic aldehydes including 4-hydroxy-2-nonenal.


Asunto(s)
Aldehído Reductasa/metabolismo , Neoplasias del Colon/tratamiento farmacológico , Neoplasias del Colon/metabolismo , Mitomicina/farmacología , Especies Reactivas de Oxígeno/metabolismo , Aldehído Reductasa/antagonistas & inhibidores , Aldehído Reductasa/biosíntesis , Aldehído Reductasa/genética , Aldehídos/metabolismo , Aldehídos/farmacología , Aldo-Ceto Reductasas , Antibióticos Antineoplásicos/farmacología , Caspasa 3/metabolismo , Neoplasias del Colon/genética , Resistencia a Antineoplásicos , Sinergismo Farmacológico , Técnicas de Silenciamiento del Gen , Células HT29 , Humanos , Peroxidación de Lípido , ARN Mensajero/análisis , ARN Mensajero/genética , ARN Interferente Pequeño/administración & dosificación , ARN Interferente Pequeño/genética , Células Tumorales Cultivadas , Regulación hacia Arriba
13.
Biochim Biophys Acta ; 1789(11-12): 681-90, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-19698806

RESUMEN

Mg(2+)-dependent neutral SMases (NSMases) have emerged as prime candidates for stress-induced ceramide production. Among isoforms identified, previous reports have suggested the importance of NSMase2. However, its activation mechanism has not been precisely reported. Here, we analyzed the mechanism of NSMase2 gene expression by the anti-cancer drug, daunorubicin (DA). DA increased cellular ceramides (C16, C18 and C24) and NSMase activity of a human breast cancer cell line, MCF-7. DA remarkably increased the NSMase2 message and protein, whereas little change in NSMase1 and NSMase3 mRNAs and only a mild increase in acid SMase mRNA were observed. Overexpression and a knock down of NSMase2 indicated that NSMase2 played a role in DA-induced cell death. NSMase2 promoter analysis revealed that three Sp1 motifs located between -148 and -42bp upstream of the first exon were important in basic as well as in DA-induced promoter activity. Consistently, luciferase vectors containing three consensus Sp1-motifs but not its mutated form showed DA-induced transcriptional activation. DA-treated MCF-7 showed increased Sp3 protein. In SL2 cells lacking Sp family proteins, both Sp1 and Sp3 overexpression increased NSMase promoter activity. Increased binding of Sp family proteins by DA to three Sp1 motifs was shown by electrophoresis mobility shift and ChIP assays.


Asunto(s)
Daunorrubicina/farmacología , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Esfingomielina Fosfodiesterasa/genética , Animales , Antibióticos Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Sitios de Unión/genética , Western Blotting , Neoplasias de la Mama/enzimología , Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Línea Celular , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Ceramidas/metabolismo , Inmunoprecipitación de Cromatina , Ensayo de Cambio de Movilidad Electroforética , Humanos , Regiones Promotoras Genéticas/genética , Unión Proteica , ARN Interferente Pequeño/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factor de Transcripción Sp1/genética , Factor de Transcripción Sp1/metabolismo , Factor de Transcripción Sp3/genética , Factor de Transcripción Sp3/metabolismo , Esfingomielina Fosfodiesterasa/metabolismo , Transfección
14.
J Cell Biochem ; 109(2): 375-82, 2010 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-19950202

RESUMEN

A mouse cell line 3T3-L1 is differentiated into adipocytes when treated with an inducer cocktail (IDX) (insulin, dexametahsone, and a cAMP phosphodiesterase inhibitor of isobutyl-methylxanthine (IBMX)). Here, we report that PLD1, but not PLD2, mRNA and protein increased during the early differentiation process. Our analysis shows that IDX resulted in a sequential induction of C/EBPbeta, PLD1, and C/EBPalpha which is a key transcription factor of late adipocyte differentiation. Among the three inducers, IBMX + any other inducer induced mild adipocyte differentiation, whereas insulin + dexamethasone did not. IBMX increased PLD1 but not PLD2 mRNA. Forskolin, an adenylate cyclase activator, and dbcAMP also increased PLD1 mRNA, suggesting the cellular cAMP as the inducer of both adipocyte differentiation and PLD1 transcription. We focused on the regulatory mechanism of PLD1 transcription during this differentiation process. IDX or a combination of inducers including IBMX increased PLD1 promoter activity, which is consistent with mRNA analysis. Promoter analysis identified two adjacent C/EBP motifs located between -338 and -231 bp from the first exon as the IBMX responsive elements. Furthermore, overexpression of C/EBPbeta, but not C/EBPalpha, increased PLD1 mRNA and PLD1 5' promoter activity. EMSA and chromatin immunoprecipitation assay confirmed the direct binding of C/EBPbeta, but not C/EBPalpha, to these C/EBP motifs of PLD1 5' promoter. Our results show that PLD1 is a target gene of C/EBPbeta through the increased cellular cAMP during early adipocyte differentiation of 3T3-L1 cells.


Asunto(s)
1-Metil-3-Isobutilxantina/farmacología , Adipogénesis/genética , Proteína alfa Potenciadora de Unión a CCAAT/metabolismo , Proteína beta Potenciadora de Unión a CCAAT/metabolismo , AMP Cíclico/metabolismo , Fosfolipasa D/genética , Células 3T3-L1 , Adipocitos/citología , Adipocitos/metabolismo , Animales , Dexametasona/farmacología , Regulación de la Expresión Génica/efectos de los fármacos , Insulina/farmacología , Ratones , Inhibidores de Fosfodiesterasa/farmacología , Fosfolipasa D/metabolismo , Elementos de Respuesta
15.
J Neurochem ; 112(2): 511-20, 2010 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19903244

RESUMEN

Ceramide is the central lipid in the sphingolipid metabolism. Ceramide kinase (CERK) and its product, ceramide 1-phosphate, have been implicated in various cellular functions. However, the regulatory mechanism of CERK gene expression remains to be determined. Here, we examined CERK mRNA level during all-trans retinoic acid (ATRA)-induced differentiation of a human neuroblastoma cell line, SH-SY5Y. ATRA reduced CERK mRNA and protein levels. Over-expression and small interfering RNA (siRNA) of CERK revealed that CERK is inhibitory against ATRA-induced neuronal differentiation and cell growth arrest. ATRA inhibited the transcriptional activity of 5'-promoter of CERK. Truncation and mutation study suggests that ATRA-responsible region was mainly located in the tandem retinoic acid responsive elements (RARE) between -40 bp and the first exon. The electrophoresis mobility shift assay revealed that ATRA produced two retarded bands, which were erased by antibody against chicken ovalbumin upstream promoter transcription factor I (COUP-TFI), RARalpha, and RXRalpha, respectively. DNA pull-down assay confirmed increased binding of these transcription factors to RARE. Transient expression of RAR, RXR, and COUP-TFI and siRNA transfection of these genes revealed that COUP-TFI inhibited CERK mRNA. Furthermore, chromatin immunoprecipitation assay showed the recruitment of co-repressors as well as three transcription factors. These results suggest that COUP-TFI was the ATRA-responsive suppressive transcription factor of CERK gene transcription.


Asunto(s)
Factor de Transcripción COUP I/metabolismo , Fosfotransferasas (Aceptor de Grupo Alcohol)/metabolismo , Activación Transcripcional/efectos de los fármacos , Tretinoina/farmacología , Sitios de Unión/efectos de los fármacos , Factor de Transcripción COUP I/genética , Muerte Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Ensayo de Cambio de Movilidad Electroforética/métodos , Exones/efectos de los fármacos , Exones/fisiología , Humanos , Inmunoprecipitación/métodos , Datos de Secuencia Molecular , Neuritas/efectos de los fármacos , Neuroblastoma , Fosfotransferasas (Aceptor de Grupo Alcohol)/genética , Regiones Promotoras Genéticas/efectos de los fármacos , ARN Interferente Pequeño/farmacología , Receptores de Ácido Retinoico/metabolismo , Receptor alfa de Ácido Retinoico , Receptores X Retinoide/metabolismo , Transfección/métodos
16.
Nagoya J Med Sci ; 71(3-4): 127-36, 2009 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-19994725

RESUMEN

The underlying mechanisms of oncogene-induced phospholipase D (PLD) activation have not been fully elucidated. The effect of the mutated-ras on PLD mRNA was examined using colon cancer cell lines as well as mock- and mutated ras-transfected NIH3T3 cells. Ras-mutation and activation were correlated, and cells with enhanced ras-activation showed increased PLD1 mRNA and protein. Analysis of the 5' PLD1 promoter using a representative cell line, DLD-1 and also mutated ras-NIH3T3, showed one Sp1-site as the important ras-responsible motif. Spl inhibition with mithramycin A and Spl siRNA inhibited PLD1 protein expression and its promoter activity. Sp1 but not Sp3 protein level and increased Sp1-motif binding activity were correlated with ras activation. Furthermore, overexpression of Sp1 in drosophila SL2 cells lacking Sp family proteins increased PLD1 promoter activity. EMSA and chromatin immunoprecipitation assay confirmed the importance of Sp1 protein binding to the Sp1-motif in ras-induced PLD1 mRNA expression.


Asunto(s)
Genes ras , Mutación , Fosfolipasa D/genética , Factor de Transcripción Sp1/fisiología , Animales , Células CACO-2 , Regulación de la Expresión Génica , Humanos , Ratones , Regiones Promotoras Genéticas , ARN Mensajero/análisis
17.
J Immunol Res ; 2019: 3030268, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30838224

RESUMEN

IFN-γ is detected in chronic lesions of atopic dermatitis (AD); however, its specific role remains to be elucidated. An impaired stratum corneum barrier function is a hallmark of AD, and it is associated with a reduction in ceramides with long-chain fatty acids (FAs) in the stratum corneum. FA elongases, ELOVL1 and ELOVL4, are essential for the synthesis of these ceramides, together with ceramide synthase 3 (CerS3). We have previously shown that IFN-γ, but not other cytokines, induced the downregulation of these enzymes in cultured keratinocytes. Our aim was to investigate the in vivo role of IFN-γ in the lesional skin of AD by analyzing mouse dermatitis models. The local mRNA expression of IFN-γ increased in mite fecal antigen-induced AD-like dermatitis in NC/Nga mice but not in imiquimod-induced psoriasis-like dermatitis in BALB/c mice. The mRNA expression of ELOVL1 and ELOVL4 significantly decreased in AD-like dermatitis, whereas ELOVL1 increased in psoriasis-like dermatitis. The expression of CerS3 increased slightly in AD-like dermatitis, but it increased by 4.6-fold in psoriasis-like dermatitis. Consistently, the relative amount of ceramides with long-chain FAs decreased in AD-like dermatitis but not in psoriasis-like dermatitis. These results suggest that IFN-γ in the lesional skin may reduce ceramides with long-chain FAs by decreasing the expression of ELOVL. Thus, IFN-γ may contribute to the chronicity of AD by impairing barrier function.


Asunto(s)
Ceramidas/análisis , Dermatitis Atópica/inmunología , Interferón gamma/inmunología , Piel/inmunología , Acetiltransferasas/genética , Animales , Células Cultivadas , Citocinas/genética , Citocinas/inmunología , Dermatitis Atópica/inducido químicamente , Modelos Animales de Enfermedad , Proteínas del Ojo/genética , Elongasas de Ácidos Grasos , Ácidos Grasos/análisis , Femenino , Imiquimod , Interferón gamma/genética , Proteínas de la Membrana/genética , Ratones , Ratones Endogámicos BALB C , Psoriasis/inducido químicamente , Psoriasis/inmunología , Piel/patología , Esfingosina N-Aciltransferasa/genética
18.
J Neurochem ; 104(5): 1372-86, 2008 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-18004999

RESUMEN

The present study examined the role of phospholipase D2 (PLD2) in the regulation of depolarization-induced neurite outgrowth and the expression of growth-associated protein-43 (GAP-43) and synapsin I in rat pheochromocytoma (PC12) cells. Depolarization of PC12 cells with 50 mmol/L KCl increased neurite outgrowth and elevated mRNA and protein expression of GAP-43 and synapsin I. These increases were suppressed by inhibition of Ca2+-calmodulin-dependent protein kinase II (CaMKII), PLD, or mitogen-activated protein kinase kinase (MEK). Knockdown of PLD2 by small interfering RNA (siRNA) suppressed the depolarization-induced neurite outgrowth, and the increase in GAP-43 and synapsin I expression. Depolarization evoked a Ca2+ rise that activated various signaling enzymes and the cAMP response element-binding protein (CREB). Silencing CaMKIIdelta by siRNA blocked KCl-induced phosphorylation of proline-rich protein tyrosine kinase 2 (Pyk2), Src kinase, and extracellular signal-regulated kinase (ERK). Inhibition of Src or MEK abolished phosphorylation of ERK and CREB. Furthermore, phosphorylation of Pyk2, ERK, and CREB was suppressed by the PLD inhibitor, 1-butanol and transfection of PLD2 siRNA, whereas it was enhanced by over-expression of wild-type PLD2. Depolarization-induced PLD2 activation was suppressed by CaMKII and Src inhibitors, but not by MEK or protein kinase A inhibitors. These results suggest that the signaling pathway of depolarization-induced PLD2 activation was downstream of CaMKIIdelta and Src, and upstream of Pyk2(Y881) and ERK/CREB, but independent of the protein kinase A. This is the first demonstration that PLD2 activation is involved in GAP-43 and synapsin I expression during depolarization-induced neuronal differentiation in PC12 cells.


Asunto(s)
Diferenciación Celular/fisiología , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/fisiología , Fosfolipasa D/fisiología , Transducción de Señal/fisiología , Factores de Transcripción/fisiología , Animales , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/antagonistas & inhibidores , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/metabolismo , Diferenciación Celular/efectos de los fármacos , Diferenciación Celular/genética , Inhibidores Enzimáticos/farmacología , Proteína GAP-43/biosíntesis , Proteína GAP-43/genética , Neuritas/efectos de los fármacos , Neuritas/enzimología , Neuritas/metabolismo , Fármacos Neuromusculares Despolarizantes/farmacología , Células PC12 , Fosfolipasa D/antagonistas & inhibidores , Fosfolipasa D/biosíntesis , Fosfolipasa D/genética , Ratas , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Sinapsinas/biosíntesis , Sinapsinas/genética
19.
Anesth Analg ; 107(6): 1892-8, 2008 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-19020135

RESUMEN

BACKGROUND: Nicorandil, an adenosine triphosphate-sensitive potassium channel opener, is reported to have an antinociceptive effect by hyperpolarization through the K(+) channel. The activation of extracellular signal-regulated kinase (ERK), a family of mitogen-activated protein kinases, plays an important role in synaptic plasticity and noxious stimulation in the dorsal root ganglion, and spinal neurons have been reported to induce its activation. To understand the biological mechanisms of nicorandil, we examined the effects of nicorandil on muscarinic acetylcholine (ACh) receptor-mediated activation of ERK in a neuronal model cell, rat pheochromocytoma PC12 cells. METHODS: PC12 cells were stimulated with ACh in the presence or absence of nicorandil, and phosphorylation of ERK was examined by a Western blot analysis. We also examined the effects of nicorandil on the ERK activation induced by 4beta-phorbol 12-myristate 13-acetate, an activator of protein kinase C, or ionomycin, a calcium ionophore. Intracellular Ca(2+) increase was visualized in fluo-3-loaded PC12 cells using fluorescence microscopy. RESULTS: Nicorandil inhibited ACh-induced ERK activation in a concentration-dependent manner. The inhibition was abolished by glibenclamide, an adenosine triphosphate-sensitive potassium channel blocker. Nicorandil suppressed the ERK activation induced by ionomycin but not 4beta-phorbol 12-myristate 13-acetate. Pretreatment of PC12 cells with nicorandil reduced the intracellular Ca(2+) concentration stimulated by ACh. CONCLUSIONS: Nicorandil inhibits muscarinic activation of the ERK signaling pathway by reducing the intracellular Ca(2+) concentration.


Asunto(s)
Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Canales KATP/efectos de los fármacos , Nicorandil/farmacología , Receptores Muscarínicos/fisiología , Animales , Calcio/metabolismo , Activación Enzimática/efectos de los fármacos , Ionomicina/farmacología , Canales KATP/fisiología , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Células PC12 , Fosforilación , Ratas , Acetato de Tetradecanoilforbol/farmacología
20.
Mol Med Rep ; 18(6): 5151-5158, 2018 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-30320355

RESUMEN

Vemurafenib, a selective inhibitor of mutated BRAF, is used to treat late­stage melanoma. However, resistance to vemurafenib is urgently required as it can have fatal consequences. Fingolimod (FTY720), a sphingosine­1­phosphate receptor modulator, has been used for the treatment of several malignant neoplasms in clinical trials. The present study investigated the effects of FTY720 and vemurafenib combination treatment on cell death induction, and defined the molecular mechanisms in vemurafenib­resistant melanoma cells. The combination treatment with FTY720 and vemurafenib reduced cell viability, and the expression of apoptosis­associated cleaved poly (adenosine diphosphate­ribose) polymerase (PARP) was increased when compared with treatment with vemurafenib alone in WM­115 cells, a vemurafenib­resistant human melanoma cell line. In addition, the protein expression of phosphorylated extracellular signal­related kinase (ERK) in WM­115 cells was decreased by this combination treatment. Vemurafenib­resistant SK­Mel­28 cells (R­SK­Mel) were established by culturing SK­Mel­28 cells, which are the most sensitive to vemurafenib, in the presence of vemurafenib. Similar to WM­155 cells, the viability of R­SK­Mel cells was reduced and the expression of cleaved PARP was increased by the combination treatment with FTY720 and vemurafenib. In addition, the expression of phosphorylated ERK and Akt was also reduced by this treatment. These results suggested that FTY720 and vemurafenib synergistically induced cell death by downregulating proliferation and survival signalling pathways in vemurafenib­resistant melanoma cells.


Asunto(s)
Antineoplásicos/farmacología , Resistencia a Antineoplásicos/efectos de los fármacos , Clorhidrato de Fingolimod/farmacología , Vemurafenib/farmacología , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Sinergismo Farmacológico , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Humanos , Melanoma/genética , Melanoma/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA