Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 47
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
J Endocrinol ; 216(2): 169-80, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23151358

RESUMEN

Obesity and diabetes are closely associated with hyperactivation of the hypothalamic-pituitary-adrenal (HPA) axis. In this study, the diet-induced obese C57BL/6 mouse was used to test the hypothesis that chronically elevated metabolic parameters associated with the development of obesity such as cholesterol and glucose can aggravate basal HPA axis activity. Because the lipocalin-type prostaglandin D(2) synthase (L-PGDS) knockout (KO) mouse is a model of accelerated insulin resistance, glucose intolerance, and obesity, it was further hypothesized that HPA activity would be greater in this model. Starting at 8 weeks of age, the L-PGDS KO and C57BL/6 mice were maintained on a low-fat or high-fat diet. After 20 or 37 weeks, fasting metabolic parameters and basal HPA axis hormones were measured and compared between genotypes. Correlation analyses were performed to identify associations between obesity-related chronic metabolic changes and changes in the basal activity of the HPA axis. Our results have identified strong positive correlations between total cholesterol, LDL-cholesterol, glucose, and HPA axis hormones that increase with age in the C57BL/6 mice. These data confirm that obesity-related elevations in cholesterol and glucose can heighten basal HPA activity. Additionally, the L-PGDS KO mice show early elevations in HPA activity with no age-related changes relative to the C57BL/6 mice.


Asunto(s)
Sistema Hipotálamo-Hipofisario/metabolismo , Resistencia a la Insulina/fisiología , Oxidorreductasas Intramoleculares/metabolismo , Lipocalinas/metabolismo , Obesidad/metabolismo , Sistema Hipófiso-Suprarrenal/metabolismo , Hormona Adrenocorticotrópica/sangre , Animales , Corticosterona/sangre , Genotipo , Hipercolesterolemia/genética , Hipercolesterolemia/metabolismo , Hiperglucemia/genética , Hiperglucemia/metabolismo , Resistencia a la Insulina/genética , Oxidorreductasas Intramoleculares/genética , Leptina/sangre , Lipocalinas/genética , Síndrome Metabólico/genética , Síndrome Metabólico/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Obesidad/genética
2.
Diabetes ; 61(12): 3139-47, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-22923471

RESUMEN

In this study, we define a new role for lipocalin prostaglandin D synthase (L-PGDS) in the control of metabolic fuel utilization by brown adipose tissue (BAT). We demonstrate that L-PGDS expression in BAT is positively correlated with BAT activity, upregulated by peroxisome proliferator-activated receptor γ coactivator 1α or 1ß and repressed by receptor-interacting protein 140. Under cold-acclimated conditions, mice lacking L-PGDS had elevated reliance on carbohydrate to provide fuel for thermogenesis and had increased expression of genes regulating glycolysis and de novo lipogenesis in BAT. These transcriptional differences were associated with increased lipid content in BAT and a BAT lipid composition enriched with de novo synthesized lipids. Consistent with the concept that lack of L-PGDS increases glucose utilization, mice lacking L-PGDS had improved glucose tolerance after high-fat feeding. The improved glucose tolerance appeared to be independent of changes in insulin sensitivity, as insulin levels during the glucose tolerance test and insulin, leptin, and adiponectin levels were unchanged. Moreover, L-PGDS knockout mice exhibited increased expression of genes involved in thermogenesis and increased norepinephrine-stimulated glucose uptake to BAT, suggesting that sympathetically mediated changes in glucose uptake may have improved glucose tolerance. Taken together, these results suggest that L-PGDS plays an important role in the regulation of glucose utilization in vivo.


Asunto(s)
Tejido Adiposo Pardo/metabolismo , Oxidorreductasas Intramoleculares/metabolismo , Lipocalinas/metabolismo , Animales , Composición Corporal/genética , Composición Corporal/fisiología , Línea Celular , Femenino , Oxidorreductasas Intramoleculares/genética , Lipocalinas/genética , Masculino , Ratones , Reacción en Cadena en Tiempo Real de la Polimerasa , Termogénesis/genética , Termogénesis/fisiología , Triglicéridos/metabolismo
3.
PLoS One ; 7(7): e39512, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22792179

RESUMEN

Mice lacking Peroxisome Proliferator-Activated Receptor γ2 (PPARγ2) have unexpectedly normal glucose tolerance and mild insulin resistance. Mice lacking PPARγ2 were found to have elevated levels of Lipocalin prostaglandin D synthase (L-PGDS) expression in BAT and subcutaneous white adipose tissue (WAT). To determine if induction of L-PGDS was compensating for a lack of PPARγ2, we crossed L-PGDS KO mice to PPARγ2 KO mice to generate Double Knock Out mice (DKO). Using DKO mice we demonstrated a requirement of L-PGDS for maintenance of subcutaneous WAT (scWAT) function. In scWAT, DKO mice had reduced expression of thermogenic genes, the de novo lipogenic program and the lipases ATGL and HSL. Despite the reduction in markers of lipolysis in scWAT, DKO mice had a normal metabolic rate and elevated serum FFA levels compared to L-PGDS KO alone. Analysis of intra-abdominal white adipose tissue (epididymal WAT) showed elevated expression of mRNA and protein markers of lipolysis in DKO mice, suggesting that DKO mice may become more reliant on intra-abdominal WAT to supply lipid for oxidation. This switch in depot utilisation from subcutaneous to epididymal white adipose tissue was associated with a worsening of whole organism metabolic function, with DKO mice being glucose intolerant, and having elevated serum triglyceride levels compared to any other genotype. Overall, L-PGDS and PPARγ2 coordinate to regulate carbohydrate and lipid metabolism.


Asunto(s)
Metabolismo de los Hidratos de Carbono , Oxidorreductasas Intramoleculares/metabolismo , Metabolismo de los Lípidos , Lipocalinas/metabolismo , PPAR gamma/metabolismo , Adipocitos/metabolismo , Tejido Adiposo Pardo/metabolismo , Tejido Adiposo Blanco/metabolismo , Animales , Biomarcadores/metabolismo , Metabolismo de los Hidratos de Carbono/genética , Eicosanoides/metabolismo , Regulación de la Expresión Génica , Resistencia a la Insulina/genética , Oxidorreductasas Intramoleculares/genética , Metabolismo de los Lípidos/genética , Lipocalinas/genética , Lipogénesis/genética , Hígado/metabolismo , Masculino , Ratones , Ratones Noqueados , PPAR gamma/genética , ARN Mensajero/genética , ARN Mensajero/metabolismo , Grasa Subcutánea/metabolismo
4.
Dev Dyn ; 240(10): 2335-43, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21887724

RESUMEN

In mammals, the Prostaglandin D(2) (PGD(2) ) signaling pathway is involved in male gonadal development, regulating Sox9 gene expression and SOX9 protein subcellular localization through lipocalin prostaglandin D synthase (L-Pgds) activity. Nevertheless, because L-Pgds is downstream of Sox9, its expression cannot explain the initial nuclear translocation of the SOX9 protein. Here, we show that another source of PGD(2) , hematopoietic-Pgds (H-Pgds) enzyme is expressed in somatic and germ cells of the embryonic gonad of both sexes, as early as embryonic day (E) 10.5, before the onset of L-Pgds expression. Inhibition of H-Pgds activity by the specific HQL-79 inhibitor leads to impaired nuclear translocation of SOX9 protein in E11.5 Sertoli cells. Furthermore, analysis of H-Pgds(-/-) male embryonic gonads confirms abnormal subcellular localization of SOX9 protein at the E11.5 early stage of mouse testicular differentiation suggesting a role for H-Pgds-produced PGD(2) in the initial nuclear translocation of SOX9.


Asunto(s)
Transporte Activo de Núcleo Celular/fisiología , Gónadas/embriología , Gónadas/metabolismo , Isomerasas/metabolismo , Factor de Transcripción SOX9/metabolismo , Animales , Núcleo Celular/metabolismo , Femenino , Gónadas/citología , Humanos , Oxidorreductasas Intramoleculares , Isomerasas/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Prostaglandina D2/metabolismo , Factor de Transcripción SOX9/genética , Células de Sertoli/citología , Células de Sertoli/metabolismo
5.
FEBS J ; 276(23): 7146-58, 2009 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-19878301

RESUMEN

Lipocalin-type prostaglandin D synthase (L-PGDS) is a multifunctional protein that produces prostaglandin D(2) and binds and transports various lipophilic substances after secretion into various body fluids as beta-trace. L-PGDS has been proposed to be a useful diagnostic marker for renal injury associated with diabetes or hypertension, because the urinary and plasma concentrations are increased in patients with these diseases. However, it remains unclear whether urinary L-PGDS is synthesized de novo in the kidney or taken up from the blood circulation. In crude extracts of monkey kidney and human urine, we found L-PGDS with its original N-terminal sequence starting from Ala23 after the signal sequence, and also its N-terminal-truncated products starting from Gln31 and Phe34. In situ hybridization and immunohistochemical staining with monoclonal antibody 5C11, which recognized the amino-terminal Ala23-Val28 loop of L-PGDS, revealed that both the mRNA and the intact form of L-PGDS were localized in the cells of Henle's loop and the glomeruli of the kidney, indicating that L-PGDS is synthesized de novo in these tissues. However, truncated forms of L-PGDS were found in the lysosomes of tubular cells, as visualized by immunostaining with 10A5, another monoclonal antibody, which recognized the three-turn alpha-helix between Arg156 and Thr173. These results suggest that L-PGDS is taken up by tubular cells and actively degraded within their lysosomes to produce the N-terminal-truncated form.


Asunto(s)
Oxidorreductasas Intramoleculares/biosíntesis , Riñón/enzimología , Lipocalinas/biosíntesis , Secuencia de Aminoácidos , Animales , Anticuerpos Monoclonales/inmunología , Haplorrinos , Humanos , Hibridación in Situ , Oxidorreductasas Intramoleculares/genética , Riñón/metabolismo , Lipocalinas/genética , Modelos Moleculares , Datos de Secuencia Molecular , Conformación Proteica , Ratas , Ratas Sprague-Dawley , Alineación de Secuencia
6.
PLoS One ; 4(11): e7737, 2009 Nov 09.
Artículo en Inglés | MEDLINE | ID: mdl-19898623

RESUMEN

BACKGROUND: THE SLEEP SEQUENCE: i) non-REM sleep, ii) REM sleep, and iii) wakefulness, is stable and widely preserved in mammals, but the underlying mechanisms are unknown. It has been shown that this sequence is disrupted by sudden REM sleep onset during active wakefulness (i.e., narcolepsy) in orexin-deficient mutant animals. Phospholipase C (PLC) mediates the signaling of numerous metabotropic receptors, including orexin receptors. Among the several PLC subtypes, the beta4 subtype is uniquely localized in the geniculate nucleus of thalamus which is hypothesized to have a critical role in the transition and maintenance of sleep stages. In fact, we have reported irregular theta wave frequency during REM sleep in PLC-beta4-deficient mutant (PLC-beta4-/-) mice. Daily behavioral phenotypes and metabotropic receptors involved have not been analyzed in detail in PLC-beta4-/- mice, however. METHODOLOGY/PRINCIPAL FINDINGS: Therefore, we analyzed 24-h sleep electroencephalogram in PLC-beta4-/- mice. PLC-beta4-/- mice exhibited normal non-REM sleep both during the day and nighttime. PLC-beta4-/- mice, however, exhibited increased REM sleep during the night, their active period. Also, their sleep was fragmented with unusual wake-to-REM sleep transitions, both during the day and nighttime. In addition, PLC-beta4-/- mice reduced ultradian body temperature rhythms and elevated body temperatures during the daytime, but had normal homeothermal response to acute shifts in ambient temperatures (22 degrees C-4 degrees C). Within the most likely brain areas to produce these behavioral phenotypes, we found that, not orexin, but group-1 metabotropic glutamate receptor (mGluR)-mediated Ca(2+) mobilization was significantly reduced in the dorsal lateral geniculate nucleus (LGNd) of PLC-beta4-/- mice. Voltage clamp recordings revealed that group-1 mGluR-mediated currents in LGNd relay neurons (inward in wild-type mice) were outward in PLC-beta4-/- mice. CONCLUSIONS/SIGNIFICANCE: These lines of evidence indicate that impaired LGNd relay, possibly mediated via group-1 mGluR, may underlie irregular sleep sequences and ultradian body temperature rhythms in PLC-beta4-/- mice.


Asunto(s)
Temperatura Corporal/fisiología , Fosfolipasa C beta/fisiología , Receptores de Glutamato Metabotrópico/metabolismo , Sueño/fisiología , Animales , Femenino , Regulación Enzimológica de la Expresión Génica , Heterocigoto , Masculino , Ratones , Ratones Endogámicos C57BL , Fenotipo , Fosfolipasa C beta/química , Sueño/genética , Sueño REM , Tálamo/metabolismo , Vigilia/fisiología
7.
Prostaglandins Other Lipid Mediat ; 90(3-4): 81-4, 2009 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-19800413

RESUMEN

We have recently found that prostaglandin (PG) D(2) stimulates food intake via DP(1) receptor. Here we show that complement C5a stimulates food intake by activating the orexigenic PGD(2) system. C5a (30-100 pmol/mouse), after intracerebroventricular administration, stimulated food intake in non-food-deprived mice. The orexigenic activity of C5a was blocked by co-administration of a DP(1) receptor antagonist, BWA868C. Central administration of C5a elevated the hypothalamic mRNA expression of COX-2 but not COX-1, and the food intake stimulation of C5a was inhibited by pretreatment with a COX-2 inhibitor, celecoxib, suggesting that C5a activates COX-2 upstream of the PGD(2)-DP(1) system. The orexigenic activity of C5a was also inhibited by an antagonist for neuropeptide Y (NPY) Y(1) receptor, which was activated downstream of the PGD(2)-DP(1) system. These results suggest that C5a stimulates food intake via a PGD(2)- and NPY-dependent mechanism. C5a is the first example of orexigenic peptides acting through the PGD(2)-NPY system in the central nervous system.


Asunto(s)
Complemento C5a/farmacología , Ingestión de Alimentos/efectos de los fármacos , Hidantoínas/farmacología , Neuropéptido Y/fisiología , Prostaglandina D2/fisiología , Animales , Ciclooxigenasa 1/genética , Ciclooxigenasa 1/metabolismo , Ciclooxigenasa 2/genética , Ciclooxigenasa 2/metabolismo , Ingestión de Alimentos/fisiología , Hipotálamo/efectos de los fármacos , Hipotálamo/enzimología , Masculino , Ratones , ARN Mensajero/biosíntesis , ARN Mensajero/genética , Receptores de Prostaglandina/antagonistas & inhibidores , Receptores de Prostaglandina/fisiología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
8.
Development ; 136(11): 1813-21, 2009 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-19429785

RESUMEN

Activation by the Y-encoded testis determining factor SRY and maintenance of expression of the Sox9 gene encoding the central transcription factor of Sertoli cell differentiation are key events in the mammalian sexual differentiation program. In the mouse XY gonad, SOX9 upregulates Fgf9, which initiates a Sox9/Fgf9 feedforward loop, and Sox9 expression is stimulated by the prostaglandin D2 (PGD2) producing lipocalin prostaglandin D synthase (L-PGDS, or PTDGS) enzyme, which accelerates commitment to the male pathway. In an attempt to decipher the genetic relationships between Sox9 and the L-Pgds/PGD2 pathway during mouse testicular organogenesis, we found that ablation of Sox9 at the onset or during the time window of expression in embryonic Sertoli cells abolished L-Pgds transcription. By contrast, L-Pgds(-/-) XY embryonic gonads displayed a reduced level of Sox9 transcript and aberrant SOX9 protein subcellular localization. In this study, we demonstrated genetically that the L-Pgds/PGD2 pathway acts as a second amplification loop of Sox9 expression. Moreover, examination of Fgf9(-/-) and L-Pgds(-/-) XY embryonic gonads demonstrated that the two Sox9 gene activity amplifying pathways work independently. These data suggest that, once activated and maintained by SOX9, production of testicular L-PGDS leads to the accumulation of PGD2, which in turn activates Sox9 transcription and nuclear translocation of SOX9. This mechanism participates together with FGF9 as an amplification system of Sox9 gene expression and activity during mammalian testicular organogenesis.


Asunto(s)
Factor 9 de Crecimiento de Fibroblastos/fisiología , Prostaglandina D2/metabolismo , Factor de Transcripción SOX9/metabolismo , Células de Sertoli/fisiología , Diferenciación Sexual/fisiología , Testículo/embriología , Transporte Activo de Núcleo Celular/fisiología , Animales , Núcleo Celular/metabolismo , Oxidorreductasas Intramoleculares/metabolismo , Lipocalinas/metabolismo , Masculino , Ratones , Mutación , Prostaglandina D2/genética , Factor de Transcripción SOX9/genética , Células de Sertoli/citología , Proteína de la Región Y Determinante del Sexo/metabolismo , Testículo/citología , Testículo/crecimiento & desarrollo
9.
J Clin Invest ; 119(6): 1477-88, 2009 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-19451694

RESUMEN

Lipocalin-type prostaglandin D synthase (L-PGDS), which was originally identified as an enzyme responsible for PGD2 biosynthesis in the brain, is highly expressed in the myocardium, including in cardiomyocytes. However, the factors that control expression of the gene encoding L-PGDS and the pathophysiologic role of L-PGDS in cardiomyocytes are poorly understood. In the present study, we demonstrate that glucocorticoids, which act as repressors of prostaglandin biosynthesis in most cell types, upregulated the expression of L-PGDS together with cytosolic calcium-dependent phospholipase A2 and COX2 via the glucocorticoid receptor (GR) in rat cardiomyocytes. Accordingly, PGD2 was the most prominently induced prostaglandin in vivo in mouse hearts and in vitro in cultured rat cardiomyocytes after exposure to GR-selective agonists. In isolated Langendorff-perfused mouse hearts, dexamethasone alleviated ischemia/reperfusion injury. This cardioprotective effect was completely abrogated by either pharmacologic inhibition of COX2 or disruption of the gene encoding L-PGDS. In in vivo ischemia/reperfusion experiments, dexamethasone reduced infarct size in wild-type mice. This cardioprotective effect of dexamethasone was markedly reduced in L-PGDS-deficient mice. In cultured rat cardiomyocytes, PGD2 protected against cell death induced by anoxia/reoxygenation via the D-type prostanoid receptor and the ERK1/2-mediated pathway. Taken together, these results suggest what we believe to be a novel interaction between glucocorticoid-GR signaling and the cardiomyocyte survival pathway mediated by the arachidonic acid cascade.


Asunto(s)
Glucocorticoides/farmacología , Corazón/efectos de los fármacos , Oxidorreductasas Intramoleculares/metabolismo , Lipocalinas/metabolismo , Daño por Reperfusión Miocárdica/metabolismo , Prostaglandina D2/biosíntesis , Animales , Células Cultivadas , Ciclooxigenasa 2/genética , Ciclooxigenasa 2/metabolismo , Activación Enzimática , Hipoxia/genética , Hipoxia/metabolismo , Ratones , Ratones Noqueados , Daño por Reperfusión Miocárdica/genética , Daño por Reperfusión Miocárdica/patología , Oxígeno/metabolismo , Ratas , Transducción de Señal
10.
Biochem Biophys Res Commun ; 378(4): 851-6, 2009 Jan 23.
Artículo en Inglés | MEDLINE | ID: mdl-19070593

RESUMEN

This study was designed to determine whether lipocalin type-prostaglandin D synthase (l-pgds) deficiency contributes to atherogenesis using gene knockout (KO) mice. A high-fat diet was given to 8-week-old C57BL/6 (wild type; WT), l-pgds KO (LKO), apolipoprotein E (apo E) KO (AKO) and l-pgds/apo E double KO (DKO) mice. The l-pgds deficient mice showed significantly increased body weight, which was accompanied by increased size of subcutaneous and visceral fat tissues. Fat deposition in the aortic wall induced by the high-fat diet was significantly increased in LKO mice compared with WT mice, although there was no significant difference between AKO and DKO mice. In LKO mice, atherosclerotic plaque in the aortic root was also increased and, furthermore, macrophage cellularity and the expression of pro-inflammatory cytokines such as interleukin-1beta and monocyte chemoattractant protein-1 were significant increased. In conclusion, l-pgds deficiency induces obesity and facilitates atherosclerosis, probably through the regulation of inflammatory responses.


Asunto(s)
Aterosclerosis/genética , Oxidorreductasas Intramoleculares/genética , Lipocalinas/genética , Obesidad/genética , Animales , Aorta/metabolismo , Aorta/patología , Apolipoproteínas E/genética , Aterosclerosis/patología , Quimiocina CCL2/biosíntesis , Grasas/metabolismo , Técnicas de Inactivación de Genes , Inflamación/genética , Interleucina-1beta/biosíntesis , Ratones , Ratones Noqueados , Obesidad/patología , Aumento de Peso/genética
11.
Hypertens Res ; 31(10): 1931-9, 2008 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-19015601

RESUMEN

Recent studies suggest that lipocalin-type prostaglandin (PG) D synthase (L-PGDS), which converts PGH2 to PGD2, is implicated in the pathogenesis of atherosclerosis. However, clinical evidence for the association between serum L-PGDS levels and atherosclerosis has not been reported. In this study, we measured the serum L-PGDS concentration using sandwich enzyme-linked immunosorbent assay (ELISA) and investigated the association with traditional cardiovascular risk factors and surrogate atherosclerotic indices, such as the maximum score of the intima-media complex thickness of the carotid artery (C-IMT(max)) and the brachial-ankle pulse wave velocity (ba-PWV), in 500 non-treated asymptomatic subjects. The serum concentration of L-PGDS was 0.56+/-0.01 (mean+/-SEM, range 0.25-1.27, median 0.54) mg/L. Serum L-PGDS levels increased with age and were higher in men than in women. Serum L-PGDS was higher in subjects with hypertension and increased with increasing numbers of the traditional atherosclerotic risk factors. When the subjects were divided into four groups according to the levels of serum L-PGDS, the age-adjusted values of C-IMT(max) and ba-PWV were significantly increased in subjects with higher serum L-PGDS levels (quartile 3 and quartile 4) compared to those in the lowest quartile (quartile 1), for both genders. Multiple regression analysis including risk factors revealed that serum L-PGDS was an independent determinant for ba-PWV (beta=0.130, p<0.001). Serum L-PGDS tended to associate with C-IMT(max) but was not statistically significant (beta=0.084, p=0.075). In conclusion, our results suggest that an increase in serum L-PGDS concentration is associated with the progression of atherosclerosis.


Asunto(s)
Enfermedades de las Arterias Carótidas/epidemiología , Enfermedades de las Arterias Carótidas/metabolismo , Hipertensión/epidemiología , Hipertensión/metabolismo , Oxidorreductasas Intramoleculares/sangre , Lipocalinas/sangre , Arterias/patología , Velocidad del Flujo Sanguíneo , Enfermedades de las Arterias Carótidas/diagnóstico por imagen , Ensayo de Inmunoadsorción Enzimática , Femenino , Humanos , Masculino , Persona de Mediana Edad , Flujo Pulsátil , Análisis de Regresión , Factores de Riesgo , Distribución por Sexo , Túnica Íntima/diagnóstico por imagen , Túnica Media/diagnóstico por imagen , Ultrasonografía
12.
FEBS Lett ; 582(5): 679-84, 2008 Mar 05.
Artículo en Inglés | MEDLINE | ID: mdl-18258196

RESUMEN

We found that prostaglandin (PG) D(2), the most abundant PG in the central nervous system, stimulates food intake after intracerebroventricular administration in mice. The orexigenic effect of PGD(2) was mimicked by a selective agonist for the DP(1) receptor among two receptor subtypes for PGD(2), and abolished by its antagonist. Central administration of an antagonist or antisense oligodeoxynucleotide for the DP(1) receptor remarkably decreased food intake, body weight and fat mass. Hypothalamic mRNA levels of lipocalin-type PGD synthase were up-regulated after fasting. The orexigenic activity of PGD(2) was also abolished by an antagonist for neuropeptide Y (NPY) Y(1) receptor. Taken together, PGD(2) may stimulate food intake through central DP(1) receptor coupled to the NPY system.


Asunto(s)
Sistema Nervioso Central/efectos de los fármacos , Conducta Alimentaria/efectos de los fármacos , Neuropéptido Y/metabolismo , Prostaglandina D2/farmacología , Animales , Depresores del Apetito/farmacología , Ciclooxigenasa 1/genética , Ciclooxigenasa 1/metabolismo , Ciclooxigenasa 2/genética , Ciclooxigenasa 2/metabolismo , Ayuno , Regulación de la Expresión Génica/efectos de los fármacos , Hipotálamo/citología , Hipotálamo/efectos de los fármacos , Hipotálamo/enzimología , Inyecciones Intraventriculares , Oxidorreductasas Intramoleculares/genética , Oxidorreductasas Intramoleculares/metabolismo , Lipocalinas/genética , Lipocalinas/metabolismo , Masculino , Ratones , Prostaglandina D2/administración & dosificación , Prostaglandina D2/metabolismo , Transporte de Proteínas/efectos de los fármacos , ARN Mensajero/genética , ARN Mensajero/metabolismo , Receptores de Prostaglandina/genética , Receptores de Prostaglandina/metabolismo , Receptores de Prostaglandina E/genética , Receptores de Prostaglandina E/metabolismo , Subtipo EP4 de Receptores de Prostaglandina E
13.
Arch Otolaryngol Head Neck Surg ; 133(7): 693-700, 2007 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-17638783

RESUMEN

OBJECTIVE: To clarify the relationship between prostaglandin D2 production and eosinophil accumulation. DESIGN: Screening and diagnostic tests. SUBJECTS: Nineteen patients with chronic rhinosinusitis. INTERVENTIONS: Nasal polyps were obtained from 19 patients at endoscopic sinus surgery. Eosinophils in nasal polyps were counted after hematoxylin-eosin staining and immunostaining with antibodies against 2 eosinophil markers-major basic protein and EG2. Hematopoietic prostaglandin D2 synthase (HPGDS) expression was examined by semiquantitative Western blot analysis and by immunohistochemical staining with anti-HPGDS antibody. RESULTS: Nasal polyps were divided into 3 groups by the degree of eosinophilic infiltration. Western blot analysis revealed that HPGDS was more intensely and frequently expressed in the group with high infiltration than in the groups with low or medium infiltration. Hematopoietic prostaglandin D2 synthase was immunohistochemically found in a subpopulation of EG2-positive eosinophils that had accumulated in the nasal polyps but not in the EG2-negative resting eosinophils. The ratio of HPGDS-positive eosinophils to EG2-positive eosinophils in the group with high eosinophil infiltration (mean+/-SD, 64.8%+/-19.2%) was twice that in the group with low eosinophil infiltration (30.5%+/-13.8%). CONCLUSION: Prostaglandin D2 was actively produced by an EG2 and HPGDS double-positive subpopulation of activated eosinophils that had infiltrated into nasal polyps.


Asunto(s)
Eosinófilos/enzimología , Oxidorreductasas Intramoleculares/metabolismo , Pólipos Nasales/metabolismo , Pólipos Nasales/patología , Adulto , Anciano , Western Blotting , Enfermedad Crónica , Proteínas en los Gránulos del Eosinófilo/análisis , Proteína Mayor Básica del Eosinófilo/análisis , Eosinófilos/inmunología , Eosinófilos/patología , Femenino , Humanos , Inmunohistoquímica , Lipocalinas , Masculino , Persona de Mediana Edad , Rinitis/metabolismo , Rinitis/patología , Sinusitis/metabolismo , Sinusitis/patología
14.
Cancer Res ; 67(3): 881-9, 2007 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-17283118

RESUMEN

Aspirin and other nonsteroidal anti-inflammatory drugs prevent some cases of colon cancer by inhibiting prostaglandin (PG) synthesis. PGE(2) promotes colon neoplasia, as shown by knockout mouse studies on enzymes and receptors in the PG cascade. A few experiments 20 to 30 years ago suggested that PGD(2) may suppress tumors, but a role for biosynthetic enzymes for PGD(2) in tumor development has not been studied. We report here that disruption of the gene for hematopoietic PGD synthase in Apc(Min/+) mice led to approximately 50% more intestinal adenomas compared with controls. Tumor size was not affected. By immunohistochemistry, we detected hematopoietic PGD synthase mainly in macrophages and monocytes of the gut mucosa. The mean number of tumors did not increase with knockout of the gene for the lipocalin type of the enzyme, which is not produced in the intestine. On the other hand, Apc(Min/+) mice with transgenic human hematopoietic PGD synthase tended to have 80% fewer intestinal adenomas. The transgene produced high mRNA levels (375-fold over endogenous). There was a suggestion of higher urinary excretion of 11beta-PGF(2alpha) and a lower excretion of a PGE(2) metabolite in transgenic mice, but differences (30-40%) were not statistically significant. The results support an interpretation that hematopoietic PGD synthase controls an inhibitory effect on intestinal tumors. Further studies will be needed to prove possible mechanisms, such as routing of PG production away from protumorigenic PGE(2) or inhibition of the nuclear factor-kappaB cascade by PGD(2) metabolites.


Asunto(s)
Poliposis Adenomatosa del Colon/enzimología , Sistema Hematopoyético/enzimología , Oxidorreductasas Intramoleculares/deficiencia , Oxidorreductasas Intramoleculares/metabolismo , Poliposis Adenomatosa del Colon/genética , Poliposis Adenomatosa del Colon/orina , Animales , Dinoprost/orina , Femenino , Oxidorreductasas Intramoleculares/biosíntesis , Oxidorreductasas Intramoleculares/genética , Lipocalinas , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Prostaglandina D2/biosíntesis , Prostaglandinas/orina
15.
Proc Natl Acad Sci U S A ; 103(47): 17949-54, 2006 Nov 21.
Artículo en Inglés | MEDLINE | ID: mdl-17093043

RESUMEN

Prostaglandin (PG) D2 has been proposed to be essential for the initiation and maintenance of the physiological sleep of rats because intracerebroventricular administration of selenium tetrachloride (SeCl4), a selective inhibitor of PGD synthase (PGDS), was shown to reduce promptly and effectively the amounts of sleep during the period of infusion. However, gene knockout (KO) mice of PGDS and prostaglandin D receptor (DP1R) showed essentially the same circadian profiles and daily amounts of sleep as wild-type (WT) mice, raising questions about the involvement of PGD2 in regulating physiological sleep. Here we examined the effect of SeCl4 on the sleep of WT and KO mice for PGDS and DP1R and that of a DP1R antagonist, ONO-4127Na, on the sleep of rats. The i.p. injection of SeCl4 into WT mice decreased the PGD2 content in the brain without affecting the amounts of PGE2 and PGF(2alpha). It inhibited sleep dose-dependently and immediately after the administration during the light period when mice normally sleep, increasing the wake time; and the treatment with this compound resulted in a distinct sleep rebound during the following dark period. The SeCl4-induced insomnia was observed in hematopoietic PGDS KO mice but not at all in lipocalin-type PGDS KO, hematopoietic and lipocalin-type PGDS double KO or DP1R KO mice. Furthermore, the DP1R antagonist ONO-4127Na reduced sleep of rats by 30% during infusion into the subarachnoid space under the rostral basal forebrain at 200 pmol/min. These results clearly show that the lipocalin-type PGDS/PGD2/DP1R system plays pivotal roles in the regulation of physiological sleep.


Asunto(s)
Cloruros/farmacología , Oxidorreductasas Intramoleculares/metabolismo , Isoenzimas/metabolismo , Prostaglandina D2/metabolismo , Receptores Inmunológicos/metabolismo , Receptores de Prostaglandina/metabolismo , Compuestos de Selenio/farmacología , Sueño/efectos de los fármacos , Animales , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Relación Dosis-Respuesta a Droga , Humanos , Oxidorreductasas Intramoleculares/antagonistas & inhibidores , Oxidorreductasas Intramoleculares/genética , Isoenzimas/antagonistas & inhibidores , Isoenzimas/genética , Lipocalinas , Ratones , Ratones Noqueados , Ratas , Receptores Inmunológicos/antagonistas & inhibidores , Receptores Inmunológicos/genética , Receptores de Prostaglandina/antagonistas & inhibidores , Receptores de Prostaglandina/genética
16.
Am J Rhinol ; 20(3): 342-8, 2006.
Artículo en Inglés | MEDLINE | ID: mdl-16871941

RESUMEN

BACKGROUND: Prostaglandin D2 (PGD2) is the major prostanoid produced in the acute phase of allergic reactions. However, its pathophysiological role in addition to the pathway of production in allergic rhinitis remains unclear. We sought to determine the expression of synthases and receptors for PGD2 in human nasal mucosa. These expressions were compared between allergic and nonallergic patients. METHODS: The expression and localization of hematopoietic-type (h)-PGD2 synthase (PGDS) and lipocalin-type (l)-PGDS were detected by immunohistochemistry. The expression of D prostanoid (DP) receptor and chemoattractant receptor-homologous molecule expressed on Th2 cells (CRTH2) was determined by quantitative real-time PCR. RESULTS: The h-PGDS but not l-PGDS was clearly expressed in nasal mucosa. The expression of h-PGDS in allergic patients was significantly higher than in control patients without mucosal hypertrophy. A variety of infiltrating cells including mast cells, eosinophils, macrophages, and lymphocytes as well as constitutive cells such as epithelial cells and fibroblasts expressed h-PGDS. The expression of both DP and CRTH2 was confirmed also. Although either the amount of DP or the amount of CRTH2 was not correlated with serum levels of IgE, the amount of CRTH2 but not DP was highly and significantly correlated with the number of eosinophils infiltrating into nasal musosa. CONCLUSION: These results suggest that PGD2 is released via the action of h-PGDS from various cells, and the expression of h-PGDS may be associated with the hypertrophic inflammation in the nose. In addition, ligation of PGD2 to CRTH2 appears to be selectively involved in eosinophil recruitment into the nose regardless of atopic status.


Asunto(s)
Oxidorreductasas Intramoleculares/metabolismo , Mucosa Nasal/metabolismo , Prostaglandina D2/metabolismo , Receptores de Prostaglandina/metabolismo , Rinitis Alérgica Perenne/metabolismo , Adulto , Eosinófilos/metabolismo , Femenino , Humanos , Inmunohistoquímica , Lipocalinas , Masculino , Persona de Mediana Edad , Receptores Inmunológicos/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Rinitis Alérgica Perenne/fisiopatología
17.
J Neurosci ; 26(16): 4383-93, 2006 Apr 19.
Artículo en Inglés | MEDLINE | ID: mdl-16624958

RESUMEN

Prostaglandin (PG) D2 is well known as a mediator of inflammation. Hematopoietic PGD synthase (HPGDS) is responsible for the production of PGD2 involved in inflammatory responses. Microglial activation and astrogliosis are commonly observed during neuroinflammation, including that which occurs during demyelination. Using the genetic demyelination mouse twitcher, a model of human Krabbe's disease, we discovered that activated microglia expressed HPGDS and activated astrocytes expressed the DP1 receptor for PGD2 in the brain of these mice. Cultured microglia actively produced PGD2 by the action of HPGDS. Cultured astrocytes expressed two types of PGD2 receptor, DP1 and DP2, and showed enhanced GFAP production after stimulation of either receptor with its respective agonist. These results suggest that PGD2 plays an important role in microglia/astrocyte interaction. We demonstrated that the blockade of the HPGDS/PGD2/DP signaling pathway using HPGDS- or DP1-null twitcher mice, and twitcher mice treated with an HPGDS inhibitor, HQL-79 (4-benzhydryloxy-1-[3-(1H-tetrazol-5-yl)-propyl]piperidine), resulted in remarkable suppression of astrogliosis and demyelination, as well as a reduction in twitching and spasticity. Furthermore, we found that the degree of oligodendroglial apoptosis was also reduced in HPGDS-null and HQL-79-treated twitcher mice. These results suggest that PGD2 is the key neuroinflammatory molecule that heightens the pathological response to demyelination in twitcher mice.


Asunto(s)
Astrocitos/metabolismo , Comunicación Celular , Enfermedades Desmielinizantes/metabolismo , Microglía/metabolismo , Prostaglandina D2/biosíntesis , Animales , Astrocitos/patología , Comunicación Celular/genética , Células Cultivadas , Enfermedades Desmielinizantes/genética , Enfermedades Desmielinizantes/patología , Leucodistrofia de Células Globoides/genética , Leucodistrofia de Células Globoides/metabolismo , Leucodistrofia de Células Globoides/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Mutantes Neurológicos , Ratones Transgénicos , Microglía/patología , Prostaglandina D2/genética
18.
Proc Natl Acad Sci U S A ; 103(13): 5179-84, 2006 Mar 28.
Artículo en Inglés | MEDLINE | ID: mdl-16547141

RESUMEN

Hematopoietic prostaglandin D(2) synthase (hPGD(2)S) metabolizes cyclooxygenase-derived prostaglandin (PG) H(2) to PGD(2), which is dehydrated to cyclopentenone PGs, including 15-deoxy-Delta(12,14)-PGJ(2) (15d-PGJ(2)). PGD(2) acts through two receptors (DP1 and DP2/CRTH2), whereas 15d-PGJ(2) can activate peroxisome proliferator-activated receptors or inhibit a range of proinflammatory signaling pathways, including NF-kappaB. Despite eliciting asthmatic and allergic reactions through the generation of PGD(2), it is not known what role hPGD(2)S plays in T helper (Th)1-driven adaptive immunity. To investigate this question, the severity and duration of a delayed type hypersensitivity reaction was examined in hPGD(2)S knockout and transgenic mice. Compared with their respective controls, knockouts displayed a more severe inflammatory response that failed to resolve, characterized histologically as persistent acute inflammation, whereas transgenic mice had little detectable inflammation. Lymphocytes isolated from inguinal lymph nodes of hPGD(2)S(-/-) animals showed hyperproliferation and increased IL-2 synthesis effects that were rescued by 15d-PGJ(2), but not PGD(2), working through either of its receptors. Crucially, 15d-PGJ(2) exerted its suppressive effects through the inhibition of NF-kappaB activation and not through peroxisome proliferator-activated receptor signaling. In contrast, lymph node cultures from transgenics proliferated more slowly and synthesized significantly less IL-2 than controls. Therefore, contrary to its role in driving Th2-like responses, this report shows that hPGD(2)S may act as an internal braking signal essential for bringing about the resolution of Th1-driven delayed type hypersensitivity reactions. Consequently, hPGD(2)S-derived cyclopentenone PGs may protect against inflammatory diseases, where T lymphocytes play a pathogenic role, as in rheumatoid arthritis, atopic eczema, and chronic rejection.


Asunto(s)
Hematopoyesis , Hipersensibilidad Tardía/enzimología , Hipersensibilidad Tardía/inmunología , Oxidorreductasas Intramoleculares/metabolismo , Animales , Antígenos/inmunología , Proliferación Celular , Extremidades/patología , Hipersensibilidad Tardía/genética , Inflamación/genética , Inflamación/inmunología , Inflamación/metabolismo , Inflamación/patología , Interleucina-2/biosíntesis , Oxidorreductasas Intramoleculares/deficiencia , Oxidorreductasas Intramoleculares/genética , Lipocalinas , Ganglios Linfáticos/metabolismo , Ganglios Linfáticos/patología , Ratones , Ratones Noqueados , FN-kappa B/metabolismo , Receptores Activados del Proliferador del Peroxisoma/metabolismo , Prostaglandinas/biosíntesis , Transducción de Señal
19.
Neurosci Res ; 55(1): 78-86, 2006 May.
Artículo en Inglés | MEDLINE | ID: mdl-16540195

RESUMEN

We found reduced locomotor activity (LA) under fasting in systemic carnitine-deficient juvenile visceral steatosis (jvs(-/-)) mice. When food was withdrawn at 8:00 a.m. (lights-off at 7:00 p.m., 12h/cycle), the nocturnal LA of jvs(-/-) mice was much less than the control (jvs(+/+) and jvs(+/-)) mice. LA recovered under carnitine or sucrose administration, but not under medium-chain triglyceride. In addition, fasted jvs(-/-) mice, without any energy supply, were activated by modafinil, a stimulator of the dopamine pathway. These results suggest that the reduced LA is not adequately explained by energy deficit. As the fasted jvs(-/-) mice showed lower body core temperature (BT), we examined the central nervous system regulating LA and BT. We found lower percentage of c-Fos positive orexin neurons in the lateral hypothalamus and reduced orexin-A concentration in the cerebrospinal fluid of fasted jvs(-/-) mice. Sleep analysis revealed that fasted jvs(-/-) mice had disruption of prolonged wakefulness, with a higher frequency of brief episodes of non-REM sleep during the dark period than fasted jvs(+/+) mice. These results strongly suggest that the reduced LA in fasted jvs(-/-) mice is related to the inhibition of orexin neuronal activity.


Asunto(s)
Carnitina/deficiencia , Ayuno/fisiología , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Actividad Motora/genética , Neuronas/fisiología , Neuropéptidos/metabolismo , Animales , Conducta Animal , Glucemia , Temperatura Corporal/efectos de los fármacos , Temperatura Corporal/fisiología , Carnitina/administración & dosificación , Electroencefalografía/métodos , Ácidos Grasos no Esterificados/sangre , Femenino , Glucosa/administración & dosificación , Inmunohistoquímica/métodos , Ratones , Ratones Noqueados , Neuronas/efectos de los fármacos , Orexinas , Polisomnografía/métodos , Proteínas Proto-Oncogénicas c-fos/metabolismo , Sueño REM/efectos de los fármacos , Sueño REM/fisiología , Sacarosa/administración & dosificación , Factores de Tiempo
20.
Nephrol Dial Transplant ; 21(4): 924-34, 2006 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-16384826

RESUMEN

BACKGROUND: Otsuka Long-Evans Tokushima Fatty (OLETF) rats genetically develop diabetes which is associated with hypertension. In preliminary studies, urinary excretions of L-PGDS (lipocaline-type prostaglandin D synthase) increase before diabetic nephropathy obviously develops, and this may predict progression of renal injury following diabetes. In the present study, we attempted to define whether urinary excretions of L-PGDS behave as the predictor of development of diabetic nephropathy in OLETF rats. METHODS: We investigated alterations of urinary L-PGDS excretions during the establishment of diabetes and assessed the relationship between the L-PGDS excretions and renal function in OLETF rats. Furthermore, we treated OLETF rats with troglitazone and analysed the effects on L-PGDS metabolisms. Urinary L-PGDS was measured by immunoenzyme assay and the occurrence of L-PGDS and its mRNA in the kidney was assessed by immunohistochemistry and a PCR method. RESULTS: Urinary excretions of L-PGDS were significantly higher in OLETF rats than non-diabetic Long-Evans Tokushima Otsuka (LETO) rats. The excretions age-dependently increased in OLETF and this increase appeared to be due to increased glomerular permeability to L-PGDS. Messenger RNA and antigenicity of L-PGDS were demonstrated in renal tissue; however, the de novo synthesis of L-PGDS mRNA seemingly contributed to urinary L-PGDS excretions much less than glomerular filtration. Multiple regression analysis revealed that urinary L-PGDS was determined by urinary protein excretions, and not by high blood pressure per se. Conversely, urinary proteinuria in the established diabetic nephropathy was predicted by urinary L-PGDS excretions in the early stage of diabetes. CONCLUSIONS: Urinary excretions of L-PGDS are likely to reflect the underlying increase in glomerular permeability. This property may be useful to predict forthcoming glomerular damage following diabetes in OLETF rats.


Asunto(s)
Nefropatías Diabéticas/orina , Oxidorreductasas Intramoleculares/orina , Proteinuria/orina , Insuficiencia Renal/orina , Animales , Cromanos/farmacología , Diabetes Mellitus Tipo 2/orina , Hipoglucemiantes/farmacología , Técnicas para Inmunoenzimas , Oxidorreductasas Intramoleculares/genética , Lipocalinas , ARN Mensajero/metabolismo , Ratas , Ratas Endogámicas OLETF , Tiazolidinedionas/farmacología , Troglitazona
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...