Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
1.
Dev Med Child Neurol ; 63(4): 407-412, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33185287

RESUMEN

AIM: To examine pro- and anti-inflammatory cytokines in children with cerebral palsy (CP) at baseline and in response to endotoxin (lipopolysaccharide), and correlate outcomes compared with age-matched comparisons, to evaluate their ability to mount an immune response. METHOD: Serum cytokines were assessed in 12 children (eight males, four females; mean age 10y 1mo [SD 1y 8mo], 6-16y) with CP against 12 age-matched comparisons (eight males, four females; mean age 9y 1mo [SD 1y 1mo]). Pro- and anti-inflammatory cytokines (interleukin-1ß, interleukin-2, interleukin-6, interleukin-8, interleukin-10, interleukin-18, tumour necrosis factor [TNF]-α, TNF-ß, interferon-γ, granulocyte-macrophage colony-stimulating factor [GM-CSF], vascular endothelial growth factor [VEGF], erythropoietin, and interleukin-1 receptor antagonist) were measured at baseline and in response to in vitro simulation with lipopolysaccharide by multiplex enzyme-linked immunosorbent assay. RESULTS: Significantly higher erythropoietin was found at baseline in children with CP compared with the comparison group. There was a strong response to lipopolysaccharide for interleukin-8, VEGF, TNF-α, and GM-CSF in both children with CP and the comparison group; however, there was significant lipopolysaccharide hyporesponsiveness in children with CP compared with the comparison group for interleukin-1α, interleukin-1ß, interleukin-2, and interleukin-6. INTERPRETATION: Altered cytokine responses in children with CP compared with the comparison group demonstrate an altered inflammatory state that may contribute to ongoing sequelae and could be a target for therapy. WHAT THIS PAPER ADDS: Altered inflammatory responses persist in children with cerebral palsy (CP). Erythropoietin is elevated in children with CP compared with the comparison group. Children with CP have reduced interleukin-1α, interleukin-1ß, interleukin-2, and interleukin-6 inflammatory responses to lipopolysaccharide.


Asunto(s)
Parálisis Cerebral/sangre , Citocinas/sangre , Adolescente , Niño , Femenino , Factor Estimulante de Colonias de Granulocitos y Macrófagos/sangre , Humanos , Inflamación/sangre , Interleucinas/sangre , Masculino , Factor de Necrosis Tumoral alfa/sangre , Factor A de Crecimiento Endotelial Vascular/sangre
2.
Pediatr Res ; 81(5): 831-837, 2017 May.
Artículo en Inglés | MEDLINE | ID: mdl-28060792

RESUMEN

BACKGROUND: Male neonates display poorer disease prognosis and outcomes compared with females. Immune genes which exhibit higher expression in umbilical cord blood (UCB) of females may contribute to the female immune advantage during infection and inflammation. The aim of this study was to quantify expression of Toll-like receptor (TLR) 4 signaling genes encoded on the X-chromosome in UCB from term female vs. male neonates. METHODS: UCB samples were collected from term neonates (n = 26) born by elective Caesarean section and whole blood was collected from adults (n = 20). Leukocyte RNA was isolated and used in quantitative PCR reactions for IκB kinase γ (IKKγ), Bruton's tyrosine kinase (BTK), and IL-1 receptor associated kinase (IRAK)1. IRAK1 protein was analyzed by Western blot and confocal microscopy. RESULTS: In neonates there was no significant difference in the relative expression of IKKγ or BTK mRNA between genders. IRAK1 gene and protein expression was significantly higher in female vs. male UCB, with increased cytosolic IRAK1 expression also evident in female UCB mononuclear cells. Adults had higher expression of all three genes compared with neonates. CONCLUSION: Increased expression of IRAK1 could be responsible, in part, for sex-specific responses to infection and subsequent immune advantage in female neonates.


Asunto(s)
Cromosomas Humanos X , Quinasas Asociadas a Receptores de Interleucina-1/genética , Transducción de Señal/genética , Receptor Toll-Like 4/genética , Adulto , Agammaglobulinemia Tirosina Quinasa , Factores de Edad , Femenino , Edad Gestacional , Humanos , Quinasa I-kappa B/sangre , Quinasa I-kappa B/genética , Quinasas Asociadas a Receptores de Interleucina-1/sangre , Leucocitos/metabolismo , Masculino , Proteínas Tirosina Quinasas/sangre , Proteínas Tirosina Quinasas/genética , Proteínas Ribosómicas/sangre , Proteínas Ribosómicas/genética , Factores Sexuales , Nacimiento a Término , Receptor Toll-Like 4/sangre , Adulto Joven
3.
Mol Endocrinol ; 22(4): 881-92, 2008 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-18202152

RESUMEN

Aldosterone elicits rapid physiological responses in target tissues such as the distal nephron through the stimulation of cell signaling cascades. We identified protein kinase D (PKD1) as an early signaling response to aldosterone treatment in the M1-cortical collecting duct (M1-CCD) cell line. PKD1 activation was blocked by the PKC inhibitor chelerythrine chloride and by rottlerin, a specific inhibitor of PKCdelta. The activation of PKCdelta and PKCepsilon coincided with PKD1 activation and while a complex was formed between PKD1 and PKCepsilon after aldosterone treatment, there was a concurrent reduction in PKD1 association with PKCdelta. A stable PKD1 knockdown M1-CCD-derrived clone was developed in which PKD1 expression was 90% suppressed by gene silencing with a PKD1-specific siRNA. The effect of aldosterone treatment on the subcellular distribution of enhanced cyan fluorescent protein (eCFP)-tagged epithelial sodium channel (ENaC) subunits in wild type (WT) and PKD1 suppressed cells was examined using confocal microscopy. In an untreated confluent monolayer of M1-CCD cells, alpha, beta, and gamma ENaC subunits were evenly distributed throughout the cytoplasm of WT and PKD1-suppressed cells. After 2 min treatment, aldosterone stimulated the localization of each of the ENaC subunits to discrete regions within the cytoplasm of WT cells. The translocation of eCFP-ENaC subunits in WT cells was inhibited by rottlerin and the mineralocorticoid receptor (MR) antagonist spironolactone. No subcellular translocation of eCFP-ENaC subunits was observed in PKD1-suppressed cells treated with aldosterone. These data demonstrate the involvement of a novel MR/PKCdelta /PKD1 signaling cascade in the earliest ENaC subunit intracellular trafficking events that follow aldosterone treatment.


Asunto(s)
Aldosterona/farmacología , Canales Epiteliales de Sodio/metabolismo , Túbulos Renales Colectores/efectos de los fármacos , Proteína Quinasa C/metabolismo , Acetofenonas/farmacología , Animales , Benzofenantridinas/farmacología , Benzopiranos/farmacología , Transporte Biológico/efectos de los fármacos , Western Blotting , Línea Celular , Electroforesis en Gel de Poliacrilamida , Activación Enzimática/efectos de los fármacos , Canales Epiteliales de Sodio/genética , Inmunoprecipitación , Corteza Renal/citología , Corteza Renal/efectos de los fármacos , Corteza Renal/metabolismo , Túbulos Renales Colectores/citología , Túbulos Renales Colectores/metabolismo , Ratones , Microscopía Confocal , Proteína Quinasa C/genética , Interferencia de ARN , Espironolactona/farmacología
4.
J Steroid Biochem Mol Biol ; 108(3-5): 310-7, 2008 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-17951051

RESUMEN

Aldosterone is a crucial modulator of ion transport across high resistance epithelia and regulates whole body electrolyte balance through its effects on the kidney and colon. The net consequence of aldosterone release is to promote salt conservation. The genomic mechanism of aldosterone action is relatively well characterized and the role of the classical mineralocorticoid receptor as a ligand-dependent transcription factor is well established. The rapid effects of aldosterone on target tissues are less well understood and there is still controversy over the identity of the aldosterone non-genomic receptor. Greater understanding of the physiological consequences of aldosterone's rapid responses in the kidney and colon has been achieved through the identification of definite and putative membrane targets and their signaling regulators.


Asunto(s)
Aldosterona/fisiología , Colon/efectos de los fármacos , Riñón/efectos de los fármacos , Intercambiadores de Sodio-Hidrógeno/fisiología , Animales , Señalización del Calcio , Colon/fisiología , Canales Epiteliales de Sodio/fisiología , Humanos , Riñón/fisiología , Canales de Potasio/fisiología , Proteínas Quinasas/fisiología , ATPasas de Translocación de Protón/fisiología , Receptores de Mineralocorticoides/fisiología , ATPasa Intercambiadora de Sodio-Potasio/fisiología , Factores de Tiempo
5.
Steroids ; 73(9-10): 979-84, 2008 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-18308353

RESUMEN

Aldosterone is an important regulator of Na(+) and K(+) transport in the distal nephron modulating the surface expression of transporters through the action of the mineralocorticoid receptor as a ligand-dependent transcription factor. Aldosterone stimulates the rapid activation of protein kinase-based signalling cascades that modulate the genomic effects of the hormone. Evidence is accumulating about the multi-factorial regulation of the epithelial sodium channel (ENaC) by aldosterone. Recent published data suggests that the activation of a novel PKC/PKD signalling pathway through the c-Src-dependent trans-activation of epidermal growth factor receptor contributes to early ENaC trafficking in response to aldosterone.


Asunto(s)
Aldosterona/metabolismo , Cationes , Nefronas/metabolismo , Animales , Transporte Biológico , Comunicación Celular , Activación Enzimática , Factor de Crecimiento Epidérmico/metabolismo , Humanos , Transporte Iónico , Modelos Biológicos , Potasio/química , Proteínas Quinasas/metabolismo , Transducción de Señal , Activación Transcripcional
6.
J Steroid Biochem Mol Biol ; 107(3-5): 180-90, 2007.
Artículo en Inglés | MEDLINE | ID: mdl-17681751

RESUMEN

Aldosterone elicits physiological responses through the modulation of gene expression and by stimulating signaling processes. Here we investigated the activation pathway of protein kinase D1 (PKD1) by aldosterone in the murine M1 renal cortical collecting duct cell line. Aldosterone stimulated a rapid increase in PKD1 activity peaking at 2-5 min and at 30 min after treatment that was insensitive to inhibitors of transcription or translation. PKD1 was not activated by aldosterone in MR null NIH-3T3 fibroblasts or M1-CCD cells propagated without dexamethasone, which did not express MR. PKD1 activation was sensitive to the MR antagonists spironolactone and RU28318 but not to the glucocorticoid receptor antagonist RU486. Aldosterone activation of PKD1 was inhibited by the epidermal growth factor (EGFR) antagonist tyrphostin AG1478 and by the c-Src inhibitor PP2. Western blotting revealed EGFR phosphorylation following aldosterone treatment at the c-Src tyrosine kinase-specific residue Tyr845. The activation of c-Src was dependent on its interaction with HSP84, since HSP84 antagonist 17-AAG inhibited both the phosphorylation of EGFR in response to aldosterone by c-Src and also the subsequent activation of PKD1.


Asunto(s)
Aldosterona/fisiología , Receptores ErbB/genética , Riñón/metabolismo , Proteína Quinasa C/metabolismo , Receptores de Mineralocorticoides/genética , Activación Transcripcional/fisiología , Animales , Línea Celular , Activación Enzimática , Riñón/citología , Ratones , Antagonistas de Receptores de Mineralocorticoides , Células 3T3 NIH , Quinazolinas , Espironolactona/análogos & derivados , Espironolactona/farmacología , Tirfostinos/farmacología
7.
Nephron Physiol ; 107(1): p1-9, 2007.
Artículo en Inglés | MEDLINE | ID: mdl-17641544

RESUMEN

Rapid signalling responses stimulated by steroid hormones have been detected in various tissues including the nephron. The significance of these responses in modulating the physiological effects elicited by mineralocorticoids, glucocorticoids and the reproductive hormones in the kidney is now becoming more evident. This review outlines how rapid signalling responses stimulated by these hormones are coupled to the regulation of membrane transport targets that impact upon the reabsorptive and excretory functions of the kidney.


Asunto(s)
Hormonas/fisiología , Riñón/fisiología , Transducción de Señal/fisiología , Esteroides/fisiología , Aldosterona/fisiología , Animales , Glucocorticoides/metabolismo , Hormonas Esteroides Gonadales/metabolismo , Humanos , Riñón/metabolismo , Proteínas de Transporte de Membrana/metabolismo , Factores de Tiempo
8.
Cell Death Differ ; 24(11): 1975-1986, 2017 11.
Artículo en Inglés | MEDLINE | ID: mdl-28885616

RESUMEN

We have previously reported that myeloid differentiation primary response gene 88 (MyD88) is downregulated during all-trans retinoic acid (RA)-induced differentiation of pluripotent NTera2 human embryonal carcinoma cells (hECCs), whereas its maintained expression is associated with RA differentiation resistance in nullipotent 2102Ep hECCs. MyD88 is the main adapter for toll-like receptor (TLR) signalling, where it determines the secretion of chemokines and cytokines in response to pathogens. In this study, we report that loss of MyD88 is essential for RA-facilitated differentiation of hECCs. Functional analysis using a specific MyD88 peptide inhibitor (PepInh) demonstrated that high MyD88 expression in the self-renewal state inhibits the expression of a specific set of HOX genes. In NTera2 cells, MyD88 is downregulated during RA-induced differentiation, a mechanism that could be broadly replicated by MyD88 PepInh treatment of 2102Ep cells. Notably, MyD88 inhibition transitioned 2102Ep cells into a stable, self-renewing state that appears to be primed for differentiation upon addition of RA. At a molecular level, MyD88 inhibition combined with RA treatment upregulated HOX, RA signalling and TLR signalling genes. These events permit differentiation through a standard downregulation of Oct4-Sox2-Nanog mechanism. In line with its role in regulating secretion of specific proteins, conditioned media experiments demonstrated that differentiated (MyD88 low) NTera2 cell media was sufficient to differentiate NTera2 cells. Protein array analysis indicated that this was owing to secretion of factors known to regulate angiogenesis, neurogenesis and all three branches of TGF-ß Superfamily signalling. Collectively, these data offer new insights into RA controlled differentiation of pluripotent cells, with notable parallels to the ground state model of embryonic stem cell self-renewal. These data may provide insights to facilitate improved differentiation protocols for regenerative medicine and differentiation-therapies in cancer treatment.


Asunto(s)
Diferenciación Celular/efectos de los fármacos , Células Madre de Carcinoma Embrionario/patología , Factor 88 de Diferenciación Mieloide/metabolismo , Células Madre Pluripotentes/patología , Tretinoina/farmacología , Diferenciación Celular/genética , Autorrenovación de las Células/efectos de los fármacos , Autorrenovación de las Células/genética , Células Madre de Carcinoma Embrionario/efectos de los fármacos , Células Madre de Carcinoma Embrionario/metabolismo , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Humanos , Mesodermo/patología , Modelos Biológicos , Células Madre Pluripotentes/efectos de los fármacos , Células Madre Pluripotentes/metabolismo , Regulación hacia Arriba/efectos de los fármacos , Regulación hacia Arriba/genética
9.
Cancer Biol Ther ; 17(5): 526-42, 2016 05 03.
Artículo en Inglés | MEDLINE | ID: mdl-26828826

RESUMEN

Oncogenic mutations in BRAF are common in melanoma and thyroid carcinoma and drive constitutive activation of the MAPK pathway. Molecularly targeted therapies of this pathway improves survival compared to chemotherapy; however, responses tend to be short-lived as resistance invariably occursCell line models of melanoma and thyroid carcinoma, +/- BRAF(V600E) activating mutation, were treated with the MEK inhibitor PD0325901. Treated and naive samples were assayed for expression of key members of the MAPK pathway. Global microRNA expression profiling of naive and resistant cells was performed via next generation sequencingand indicated pluripotency pathways in resistance. Parental cell lines were progressed to holoclones to confirm the miRNA stemness profileMembers of the MIR302/373/374/520 family of embryonic stem cell specific cell cycle regulating (ESCC) microRNAs were identified as differentially expressed between resistant BRAF(V600E) melanoma and thyroid cell lines. Upregulated expression of gene and protein stemness markers, upregulated expression of MAPK pathway genes and downregulation of the ESCC MIR302 cluster in BRAF(V600E) melanoma indicated an increased stem-like phenotype in resistant BRAF(V600E) melanoma. Conversely, downregulated expression of gene and protein stemness markers, downregulated expression of MAPK pathway genes, upregulation of the ESCC MIR520 cluster, reeexpression of cell surface receptors, and induced differentiation-associated morphology in resistant BRAF(V600E) indicate a differentiated phenotype associated with MEK inhibitor resistance in BRAF(V600E) thyroid cellsThe differential patterns of resistance observed between BRAF(V600E) melanoma and thyroid cell lines may reflect tissue type or de novo differentiation, but could have significant impact on the response of primary and metastatic cells to MEK inhibitor treatment. This study provides a basis for the investigation of the cellular differentiation/self-renewal access and its role in resistance to MEK inhibition.


Asunto(s)
Melanoma/tratamiento farmacológico , Quinasas de Proteína Quinasa Activadas por Mitógenos/metabolismo , Neoplasias de la Tiroides/tratamiento farmacológico , Diferenciación Celular , Línea Celular Tumoral , Humanos , Células Madre Pluripotentes
10.
J Mol Endocrinol ; 56(3): 261-71, 2016 04.
Artículo en Inglés | MEDLINE | ID: mdl-26759391

RESUMEN

Metformin is the main drug of choice for treating type 2 diabetes, yet the therapeutic regimens and side effects of the compound are all undesirable and can lead to reduced compliance. The aim of this study was to elucidate the mechanism of action of two novel compounds which improved glucose handling and weight gain in mice on a high-fat diet. Wildtype C57Bl/6 male mice were fed on a high-fat diet and treated with novel, anti-diabetic compounds. Both compounds restored the glucose handling ability of these mice. At a cellular level, these compounds achieve this by inhibiting complex I activity in mitochondria, leading to AMP-activated protein kinase activation and subsequent increased glucose uptake by the cells, as measured in the mouse C2C12 muscle cell line. Based on the inhibition of NADH dehydrogenase (IC50 27µmolL(-1)), one of these compounds is close to a thousand fold more potent than metformin. There are no indications of off target effects. The compounds have the potential to have a greater anti-diabetic effect at a lower dose than metformin and may represent a new anti-diabetic compound class. The mechanism of action appears not to be as an insulin sensitizer but rather as an insulin substitute.


Asunto(s)
Dieta Alta en Grasa , Complejo I de Transporte de Electrón/antagonistas & inhibidores , Glucosa/metabolismo , Hipoglucemiantes/farmacología , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Piperazinas/farmacología , Tiofenos/farmacología , Proteínas Quinasas Activadas por AMP/metabolismo , Adenosina Trifosfato/metabolismo , Animales , Células CHO , Línea Celular , Cricetulus , Hipoglucemiantes/química , Masculino , Ratones , NAD/metabolismo , Consumo de Oxígeno , Ratas
11.
PLoS One ; 7(7): e40717, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22792399

RESUMEN

The IGROVCDDP cisplatin-resistant ovarian cancer cell line is also resistant to paclitaxel and models the resistance phenotype of relapsed ovarian cancer patients after first-line platinum/taxane chemotherapy. A TaqMan low-density array (TLDA) was used to characterise the expression of 380 genes associated with chemotherapy resistance in IGROVCDDP cells. Paclitaxel resistance in IGROVCDDP is mediated by gene and protein overexpression of P-glycoprotein and the protein is functionally active. Cisplatin resistance was not reversed by elacridar, confirming that cisplatin is not a P-glycoprotein substrate. Cisplatin resistance in IGROVCDDP is multifactorial and is mediated in part by the glutathione pathway and decreased accumulation of drug. Total cellular glutathione was not increased. However, the enzyme activity of GSR and GGT1 were up-regulated. The cellular localisation of copper transporter CTR1 changed from membrane associated in IGROV-1 to cytoplasmic in IGROVCDDP. This may mediate the previously reported accumulation defect. There was decreased expression of the sodium potassium pump (ATP1A), MRP1 and FBP which all have been previously associated with platinum accumulation defects in platinum-resistant cell lines. Cellular localisation of MRP1 was also altered in IGROVCDDP shifting basolaterally, compared to IGROV-1. BRCA1 was also up-regulated at the gene and protein level. The overexpression of P-glycoprotein in a resistant model developed with cisplatin is unusual. This demonstrates that P-glycoprotein can be up-regulated as a generalised stress response rather than as a specific response to a substrate. Mechanisms characterised in IGROVCDDP cells may be applicable to relapsed ovarian cancer patients treated with frontline platinum/taxane chemotherapy.


Asunto(s)
Subfamilia B de Transportador de Casetes de Unión a ATP/metabolismo , Antineoplásicos/farmacología , Cisplatino/farmacología , Neoplasias Ováricas/metabolismo , Paclitaxel/farmacología , Subfamilia B de Transportador de Casetes de Unión a ATP/genética , Biomarcadores/metabolismo , Proteínas de Transporte de Catión/metabolismo , Línea Celular Tumoral , Transportador de Cobre 1 , Resistencia a Antineoplásicos/genética , Femenino , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Genes BRCA1 , Glutatión/metabolismo , Humanos , Redes y Vías Metabólicas , Neoplasias Ováricas/genética
12.
PLoS One ; 6(10): e26125, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-22022533

RESUMEN

Thrombosis is common in ovarian cancer. However, the interaction of platelets with ovarian cancer cells has not been critically examined. To address this, we investigated platelet interactions in a range of ovarian cancer cell lines with different metastatic potentials [HIO-80, 59M, SK-OV-3, A2780, A2780cis]. Platelets adhered to ovarian cancer cells with the most significant adhesion to the 59M cell line. Ovarian cancer cells induced platelet activation [P-selectin expression] in a dose dependent manner, with the most significant activation seen in response to the 59M cell line. The platelet antagonists [cangrelor, MRS2179, and apyrase] inhibited 59M cell induced activation suggesting a P2Y12 and P2Y1 receptor mediated mechanism of platelet activation dependent on the release of ADP by 59M cells. A2780 and 59M cells potentiated PAR-1, PAR-4, and TxA2 receptor mediated platelet activation, but had no effect on ADP, epinephrine, or collagen induced activation. Analysis of gene expression changes in ovarian cancer cells following treatment with washed platelets or platelet releasate showed a subtle but valid upregulation of anti-apoptotic, anti-autophagy pro-angiogenic, pro-cell cycle and metabolic genes. Thus, ovarian cancer cells with different metastatic potential adhere and activate platelets differentially while both platelets and platelet releasate mediate pro-survival and pro-angiogenic signals in ovarian cancer cells.


Asunto(s)
Degranulación de la Célula , Neovascularización Patológica/patología , Neoplasias Ováricas/irrigación sanguínea , Neoplasias Ováricas/patología , Adhesividad Plaquetaria , Transducción de Señal , Adenosina Difosfato/análogos & derivados , Adenosina Difosfato/farmacología , Adenosina Monofosfato/análogos & derivados , Adenosina Monofosfato/farmacología , Adenosina Trifosfato/farmacología , Apirasa/farmacología , Ácido Araquidónico/farmacología , Degranulación de la Célula/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Sinergismo Farmacológico , Transición Epitelial-Mesenquimal/efectos de los fármacos , Transición Epitelial-Mesenquimal/genética , Femenino , Perfilación de la Expresión Génica , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Análisis de Secuencia por Matrices de Oligonucleótidos , Neoplasias Ováricas/genética , Fragmentos de Péptidos/farmacología , Adhesividad Plaquetaria/efectos de los fármacos , Receptores Purinérgicos P2Y1/metabolismo , Receptores Purinérgicos P2Y12/metabolismo , Receptores de Trombina/antagonistas & inhibidores , Receptores de Trombina/metabolismo , Reproducibilidad de los Resultados , Transducción de Señal/efectos de los fármacos
13.
J Steroid Biochem Mol Biol ; 118(1-2): 18-28, 2010 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19804826

RESUMEN

Aldosterone elicits transcriptional responses in target tissues and also rapidly stimulates the activation of protein kinase signalling cascades independently of de novo protein synthesis. Here we investigated aldosterone-induced cell proliferation and extra-cellular regulated kinase 1 and 2 (ERK1/2) mitogen activated protein (MAP) kinase signalling in the M1 cortical collecting duct cell line (M1-CCD). Aldosterone promoted the proliferative growth of M1-CCD cells, an effect that was protein kinase D1 (PKD1), PKCdelta and ERK1/2-dependent. Aldosterone induced the rapid activation of ERK1/2 with peaks of activation at 2 and 10 to 30 min after hormone treatment followed by sustained activation lasting beyond 120 min. M1-CCD cells suppressed in PKD1 expression exhibited only the early, transient peaks in ERK1/2 activation without the sustained phase. Aldosterone stimulated the physical association of PKD1 with ERK1/2 within 2 min of treatment. The mineralocorticoid receptor (MR) antagonist RU28318 inhibited the early and late phases of aldosterone-induced ERK1/2 activation, and also aldosterone-induced proliferative cell growth. Aldosterone induced the sub-cellular redistribution of ERK1/2 to the nuclei at 2 min and to cytoplasmic sites, proximal to the nuclei after 30 min. This sub-cellular distribution of ERK1/2 was inhibited in cells suppressed in the expression of PKD1.


Asunto(s)
Aldosterona/farmacología , Proliferación Celular/efectos de los fármacos , Células Epiteliales/metabolismo , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Corteza Renal/citología , Túbulos Renales Colectores/citología , Canales Catiónicos TRPP/metabolismo , Acetofenonas/farmacología , Transporte Activo de Núcleo Celular/efectos de los fármacos , Animales , Benzopiranos/farmacología , Línea Celular , Citoplasma/metabolismo , Activación Enzimática , Células Epiteliales/citología , Células Epiteliales/efectos de los fármacos , Receptores ErbB/antagonistas & inhibidores , Receptores ErbB/metabolismo , Quinasas MAP Reguladas por Señal Extracelular/antagonistas & inhibidores , Flavonoides/farmacología , Ratones , Antagonistas de Receptores de Mineralocorticoides/farmacología , Proteína Quinasa 1 Activada por Mitógenos/antagonistas & inhibidores , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/antagonistas & inhibidores , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Modelos Biológicos , Fosforilación/efectos de los fármacos , Unión Proteica/efectos de los fármacos , Proteína Quinasa C-delta/antagonistas & inhibidores , Proteína Quinasa C-delta/metabolismo , Inhibidores de Proteínas Quinasas/farmacología , Quinazolinas , Receptores de Mineralocorticoides/metabolismo , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología , Espironolactona/análogos & derivados , Espironolactona/farmacología , Canales Catiónicos TRPP/antagonistas & inhibidores , Canales Catiónicos TRPP/genética , Tirfostinos/farmacología
14.
Mol Cell Endocrinol ; 325(1-2): 8-17, 2010 Aug 30.
Artículo en Inglés | MEDLINE | ID: mdl-20434520

RESUMEN

Aldosterone treatment of M1-CCD cells stimulated an increase in epithelial Na(+) channel (ENaC) alpha-subunit expression that was mainly localized to the apical membrane. PKD1-suppressed cells constitutively expressed ENaCalpha at low abundance, with no increase after aldosterone treatment. In the PKD1-suppressed cells, ENaCalpha was mainly localized proximal to the basolateral surface of the epithelium both before and after aldosterone treatment. Apical membrane insertion of ENaCbeta in response to aldosterone treatment was also sensitive to PKD1 suppression as was the aldosterone-induced rise in the amiloride-sensitive, trans-epithelial current (I(TE)). The interaction of the mineralocorticoid receptor (MR) with specific elements in the promoters of aldosterone responsive genes is stabilized by ligand interaction and phosphorylation. PKD1 suppression inhibited aldosterone-induced SGK-1 expression. The nuclear localization of MR was also blocked by PKD1 suppression and MEK antagonism implicating both these kinases in MR nuclear stabilization. PKD1 thus modulates aldosterone-induced ENaC activity through the modulation of sub-cellular trafficking and the stabilization of MR nuclear localization.


Asunto(s)
Aldosterona/farmacología , Agonistas del Canal de Sodio Epitelial , Corteza Renal/metabolismo , Túbulos Renales Colectores/metabolismo , Proteína Quinasa C/fisiología , Amilorida/farmacología , Animales , Línea Celular , Canales Epiteliales de Sodio/metabolismo , Expresión Génica/efectos de los fármacos , Corteza Renal/efectos de los fármacos , Túbulos Renales Colectores/efectos de los fármacos , Ratones , Ratones Transgénicos , Proteína Quinasa C/antagonistas & inhibidores , Proteína Quinasa C/genética , Transporte de Proteínas/efectos de los fármacos , ARN Interferente Pequeño/farmacología , Receptores de Mineralocorticoides/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA