Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
1.
J Clin Invest ; 134(13)2024 May 14.
Artículo en Inglés | MEDLINE | ID: mdl-38743489

RESUMEN

Mutations in the cystic fibrosis transmembrane conductance regulator (CFTR) gene cause cystic fibrosis (CF), a multiorgan disease that is characterized by diverse metabolic defects. However, other than specific CFTR mutations, the factors that influence disease progression and severity remain poorly understood. Aberrant metabolite levels have been reported, but whether CFTR loss itself or secondary abnormalities (infection, inflammation, malnutrition, and various treatments) drive metabolic defects is uncertain. Here, we implemented comprehensive arteriovenous metabolomics in newborn CF pigs, and the results revealed CFTR as a bona fide regulator of metabolism. CFTR loss impaired metabolite exchange across organs, including disruption of lung uptake of fatty acids, yet enhancement of uptake of arachidonic acid, a precursor of proinflammatory cytokines. CFTR loss also impaired kidney reabsorption of amino acids and lactate and abolished renal glucose homeostasis. These and additional unexpected metabolic defects prior to disease manifestations reveal a fundamental role for CFTR in controlling multiorgan metabolism. Such discovery informs a basic understanding of CF, provides a foundation for future investigation, and has implications for developing therapies targeting only a single tissue.


Asunto(s)
Regulador de Conductancia de Transmembrana de Fibrosis Quística , Fibrosis Quística , Metabolómica , Animales , Regulador de Conductancia de Transmembrana de Fibrosis Quística/metabolismo , Regulador de Conductancia de Transmembrana de Fibrosis Quística/genética , Porcinos , Fibrosis Quística/metabolismo , Fibrosis Quística/patología , Fibrosis Quística/genética , Riñón/metabolismo , Pulmón/metabolismo , Pulmón/patología , Humanos , Glucosa/metabolismo , Ácido Araquidónico/metabolismo
2.
bioRxiv ; 2024 Apr 29.
Artículo en Inglés | MEDLINE | ID: mdl-38746329

RESUMEN

The liver acts as a master regulator of metabolic homeostasis in part by performing gluconeogenesis. This process is dysregulated in type 2 diabetes, leading to elevated hepatic glucose output. The parenchymal cells of the liver (hepatocytes) are heterogeneous, existing on an axis between the portal triad and the central vein, and perform distinct functions depending on location in the lobule. Here, using single cell analysis of hepatocytes across the liver lobule, we demonstrate that gluconeogenic gene expression ( Pck1 and G6pc ) is relatively low in the fed state and gradually increases first in the periportal hepatocytes during the initial fasting period. As the time of fasting progresses, pericentral hepatocyte gluconeogenic gene expression increases, and following entry into the starvation state, the pericentral hepatocytes show similar gluconeogenic gene expression to the periportal hepatocytes. Similarly, pyruvate-dependent gluconeogenic activity is approximately 10-fold higher in the periportal hepatocytes during the initial fasting state but only 1.5-fold higher in the starvation state. In parallel, starvation suppresses canonical beta-catenin signaling and modulates expression of pericentral and periportal glutamine synthetase and glutaminase, resulting in an enhanced pericentral glutamine-dependent gluconeogenesis. These findings demonstrate that hepatocyte gluconeogenic gene expression and gluconeogenic activity are highly spatially and temporally plastic across the liver lobule, underscoring the critical importance of using well-defined feeding and fasting conditions to define the basis of hepatic insulin resistance and glucose production.

3.
Med Sci Educ ; 31(6): 1895-1901, 2021 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-34513261

RESUMEN

OBJECTIVES: To assess student outcomes and experiences, as well as preceptor experiences, after emergently converting a preclinical medical school renal course to a remote setting during the COVID-19 pandemic. METHODS: First-year medical student examination scores and responses to Likert-scale questions on end-of-course evaluations from the 2018-2019 (traditional) and 2019-2020 (remote) academic years were compared. Free-text responses from students and preceptors were analyzed using a qualitative summative approach to extract major themes in perceptions of remote learning. RESULTS: Mean student scores on course examinations did not significantly differ between the traditional and remote settings (p = 0.23 and 0.84 respectively). Quantitative analysis of student evaluations revealed no significant difference across all items in mean Likert-scale responses. Student and preceptor free-text responses identified course leader engagement and responsiveness as essential to the success of remote-based learning. Optimal group size and online etiquette are areas that require attention. CONCLUSIONS: Despite rapid conversion of a preclinical medical school renal course to a remote-based format in the setting of the COVID-19 pandemic, student scores and evaluations remain positive and largely unchanged.

4.
Int J Mol Sci ; 19(1)2018 Jan 09.
Artículo en Inglés | MEDLINE | ID: mdl-29315239

RESUMEN

One of the main causes of hyperglycemia is inefficient or impaired glucose utilization by skeletal muscle, which can be exacerbated by chronic high caloric intake. Previously, we identified a natural compound, mangiferin (MGF) that improved glucose utilization in high fat diet (HFD)-induced insulin resistant mice. To further identify the molecular mechanisms of MGF action on glucose metabolism, we conducted targeted metabolomics and transcriptomics studies of glycolyic and mitochondrial bioenergetics pathways in skeletal muscle. These data revealed that MGF increased glycolytic metabolites that were further augmented as glycolysis proceeded from the early to the late steps. Consistent with an MGF-stimulation of glycolytic flux there was a concomitant increase in the expression of enzymes catalyzing glycolysis. MGF also increased important metabolites in the tricarboxylic acid (TCA) cycle, such as α-ketoglutarate and fumarate. Interestingly however, there was a reduction in succinate, a metabolite that also feeds into the electron transport chain to produce energy. MGF increased succinate clearance by enhancing the expression and activity of succinate dehydrogenase, leading to increased ATP production. At the transcriptional level, MGF induced mRNAs of mitochondrial genes and their transcriptional factors. Together, these data suggest that MGF upregulates mitochondrial oxidative capacity that likely drives the acceleration of glycolysis flux.


Asunto(s)
Metabolismo Energético/efectos de los fármacos , Glucólisis/efectos de los fármacos , Mitocondrias/efectos de los fármacos , Xantonas/farmacología , Animales , Línea Celular , Ciclo del Ácido Cítrico/efectos de los fármacos , ADN Mitocondrial/metabolismo , Dieta Alta en Grasa , Gliceraldehído-3-Fosfato Deshidrogenasas/genética , Gliceraldehído-3-Fosfato Deshidrogenasas/metabolismo , Metaboloma/efectos de los fármacos , Metabolómica , Ratones , Ratones Endogámicos C57BL , Mitocondrias/genética , Mitocondrias/metabolismo , Succinato Deshidrogenasa/genética , Succinato Deshidrogenasa/metabolismo
5.
Kidney Int ; 94(3): 438-439, 2018 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-33237006
6.
J Clin Invest ; 127(9): 3367-3374, 2017 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-28783044

RESUMEN

Thiazide diuretics are among the most widely used treatments for hypertension, but thiazide-induced hyponatremia (TIH), a clinically significant adverse effect, is poorly understood. Here, we have studied the phenotypic and genetic characteristics of patients hospitalized with TIH. In a cohort of 109 TIH patients, those with severe TIH displayed an extended phenotype of intravascular volume expansion, increased free water reabsorption, urinary prostaglandin E2 excretion, and reduced excretion of serum chloride, magnesium, zinc, and antidiuretic hormone. GWAS in a separate cohort of 48 TIH patients and 2,922 controls from the 1958 British birth cohort identified an additional 14 regions associated with TIH. We identified a suggestive association with a variant in SLCO2A1, which encodes a prostaglandin transporter in the distal nephron. Resequencing of SLCO2A1 revealed a nonsynonymous variant, rs34550074 (p.A396T), and association with this SNP was replicated in a second cohort of TIH cases. TIH patients with the p.A396T variant demonstrated increased urinary excretion of prostaglandin E2 and metabolites. Moreover, the SLCO2A1 phospho-mimic p.A396E showed loss of transporter function in vitro. These findings indicate that the phenotype of TIH involves a more extensive metabolic derangement than previously recognized. We propose one mechanism underlying TIH development in a subgroup of patients in which SLCO2A1 regulation is altered.


Asunto(s)
Hiponatremia/inducido químicamente , Inhibidores de los Simportadores del Cloruro de Sodio/efectos adversos , Tiazidas/efectos adversos , Anciano , Anciano de 80 o más Años , Acuaporina 1/genética , Acuaporina 2/genética , Estudios de Cohortes , Dinoprostona/metabolismo , Femenino , Estudio de Asociación del Genoma Completo , Humanos , Hiponatremia/genética , Masculino , Persona de Mediana Edad , Nefronas/metabolismo , Transportadores de Anión Orgánico/genética , Farmacogenética , Fenotipo , Polimorfismo de Nucleótido Simple , Prostaglandinas/metabolismo , Reino Unido , Agua/química
7.
J Allergy Clin Immunol Pract ; 5(4): 998-1007.e2, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28159558

RESUMEN

BACKGROUND: Aspirin desensitization followed by daily aspirin provides therapeutic benefits to patients with aspirin-exacerbated respiratory disease (AERD). It is not well understood how eicosanoid levels change during aspirin treatment. OBJECTIVE: To investigate associations between clinical outcomes of aspirin treatment and plasma eicosanoid levels in patients with AERD. METHODS: Thirty-nine patients with AERD were offered aspirin treatment (650 mg twice daily) for 4 weeks. Respiratory parameters and plasma levels of multiple eicosanoids were recorded at baseline and after 4 weeks of aspirin therapy using the Asthma Control Test and Rhinoconjunctivitis Quality of Life Questionnaire. Respiratory function was evaluated using the FEV1 and nasal inspiratory peak flow. RESULTS: After aspirin treatment, respiratory symptoms improved in 16 patients, worsened in 12 patients, and did not change in 4 patients. Seven patients were unable to complete the desensitization protocol. Patients with symptom improvement had higher baseline plasma 15-hydroxyeicosatetraenoic acid (15-HETE) levels than did patients with symptom worsening: 7006 pg/mL (interquartile range, 6056-8688 pg/mL) versus 4800 pg/mL (interquartile range, 4238-5575 pg/mL), P = .0005. Baseline 15-HETE plasma levels positively correlated with the change in Asthma Control Test score (r = 0.61; P = .001) and in FEV1 after 4 weeks of aspirin treatment (r = 0.49; P = .01). It inversely correlated with Rhinoconjunctivitis Quality of Life Questionnaire score (r = -0.58; P = .002). Black and Latino patients were more likely to have symptom worsening on aspirin or fail to complete the initial desensitization than white, non-Latino patients (P = .02). CONCLUSIONS: In patients with AERD, low baseline 15-HETE plasma levels and black or Latino ethnicity are associated with worsening of respiratory symptoms during aspirin treatment.


Asunto(s)
Aspirina/uso terapéutico , Asma Inducida por Aspirina/sangre , Asma Inducida por Aspirina/terapia , Inhibidores de la Ciclooxigenasa/uso terapéutico , Desensibilización Inmunológica , Ácidos Hidroxieicosatetraenoicos/sangre , Adulto , Asma Inducida por Aspirina/etnología , Asma Inducida por Aspirina/fisiopatología , Población Negra , Femenino , Volumen Espiratorio Forzado , Hispánicos o Latinos , Humanos , Masculino , Persona de Mediana Edad , Resultado del Tratamiento
8.
Trans Am Clin Climatol Assoc ; 126: 248-57, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26330684

RESUMEN

We discovered the prostaglandin transporter (PGT) and cloned the human cDNA and gene. PGT transports extracellular prostaglandins (PGs) into the cytoplasm for enzymatic inactivation. PGT knockout mice have elevated prostaglandin E2 (PGE2) and neonatal patent ductus arteriosus, which reflects PGT's control over PGE2 signaling at EP1/EP4 cell-surface receptors. Interestingly, rescued PGT knockout pups have a nearly normal phenotype, as do human PGT nulls. Given the benign phenotype of PGT genetic nulls, and because PGs are useful medicines, we have approached PGT as a drug target. Triazine library screening yielded a lead compound of inhibitory constant 50% (IC50) = 3.7 µM, which we developed into a better inhibitor of IC50 378 nM. Further structural improvements have yielded 26 rationally designed derivatives with IC50 < 100 nM. The therapeutic approach of increasing endogenous PGs by inhibiting PGT offers promise in diseases such as pulmonary hypertension and obesity.


Asunto(s)
Diseño de Fármacos , Eicosanoides/metabolismo , Terapia Molecular Dirigida , Transportadores de Anión Orgánico/antagonistas & inhibidores , Transducción de Señal/efectos de los fármacos , Triazinas/farmacología , Animales , Transporte Biológico , Perros , Genotipo , Humanos , Células de Riñón Canino Madin Darby , Ratones Transgénicos , Estructura Molecular , Transportadores de Anión Orgánico/genética , Transportadores de Anión Orgánico/metabolismo , Fenotipo , Ratas , Relación Estructura-Actividad , Transfección , Triazinas/química
9.
PLoS One ; 10(7): e0133615, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26230411

RESUMEN

Peripheral ischemia, resulting from diminished arterial flow and defective local vascularization, is one of the main causes of impaired wound healing in diabetes. Vasodilatory prostaglandins (PGs), including PGE2 and PGI2, regulate blood flow in peripheral tissues. PGs also stimulate angiogenesis by inducing vascular endothelial growth factor. However, PG levels are reduced in diabetes mainly due to enhanced degradation. We hypothesized that inhibition of the prostaglandin transporter (PGT) (SLCO2A1), which mediates the degradation of PGs, would increase blood flow and stimulate vascularization, thereby mitigating peripheral ischemia and accelerating wound healing in diabetes. Here we report that inhibiting PGT with intravenously injected PGT inhibitor, T26A, increased blood flow in ischemic hind limbs created in non-diabetic rats and streptozotocin induced diabetic rats. Systemic, or combined with topical, T26A accelerated closure of cutaneous wounds. Immunohistochemical examination revealed that inhibition of PGT enhanced vascularization (marked by larger numbers of vessels formed by CD34+ cells), and accelerated re-epithelialization of cutaneous wounds. In cultured primary human bone marrow CD34+ cells and human epidermal keratinocytes (HEKs) either inhibiting or silencing PGT increased migration in both cell lines. Thus PGT directly regulates mobilization of endothelial progenitor cells (EPCs) and HEKs, which could contribute to PGT-mediated vascularization and re-epithelialization. At the molecular level, systemic inhibition of PGT raised circulating PGE2. Taken together, our data demonstrate that PGT modulates arterial blood flow, mobilization of EPCs and HEKs, and vascularization and epithelialization in wound healing by regulating vasodilatory and pro-angiogenic PGs.


Asunto(s)
Diabetes Mellitus Experimental/metabolismo , Neovascularización Fisiológica/fisiología , Transportadores de Anión Orgánico/antagonistas & inhibidores , Prostaglandinas/metabolismo , Cicatrización de Heridas/fisiología , Animales , Antígenos CD34/metabolismo , Células de la Médula Ósea/efectos de los fármacos , Células de la Médula Ósea/metabolismo , Línea Celular , Células Progenitoras Endoteliales/efectos de los fármacos , Células Progenitoras Endoteliales/metabolismo , Miembro Posterior/irrigación sanguínea , Miembro Posterior/metabolismo , Humanos , Queratinocitos/efectos de los fármacos , Queratinocitos/metabolismo , Masculino , Neovascularización Fisiológica/efectos de los fármacos , Ratas , Ratas Sprague-Dawley , Repitelización/efectos de los fármacos , Repitelización/fisiología , Flujo Sanguíneo Regional/efectos de los fármacos , Piel/irrigación sanguínea , Piel/efectos de los fármacos , Piel/metabolismo , Estreptozocina/farmacología , Triazinas/farmacología , Cicatrización de Heridas/efectos de los fármacos , para-Aminobenzoatos/farmacología
10.
PLoS One ; 10(6): e0131735, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26121580

RESUMEN

Inhibiting the synthesis of endogenous prostaglandins with nonsteroidal anti-inflammatory drugs exacerbates arterial hypertension. We hypothesized that the converse, i.e., raising the level of endogenous prostaglandins, might have anti-hypertensive effects. To accomplish this, we focused on inhibiting the prostaglandin transporter PGT (SLCO2A1), which is the obligatory first step in the inactivation of several common PGs. We first examined the role of PGT in controlling arterial blood pressure blood pressure using anesthetized rats. The high-affinity PGT inhibitor T26A sensitized the ability of exogenous PGE2 to lower blood pressure, confirming both inhibition of PGT by T26A and the vasodepressor action of PGE2 T26A administered alone to anesthetized rats dose-dependently lowered blood pressure, and did so to a greater degree in spontaneously hypertensive rats than in Wistar-Kyoto control rats. In mice, T26A added chronically to the drinking water increased the urinary excretion and plasma concentration of PGE2 over several days, confirming that T26A is orally active in antagonizing PGT. T26A given orally to hypertensive mice normalized blood pressure. T26A increased urinary sodium excretion in mice and, when added to the medium bathing isolated mouse aortas, T26A increased the net release of PGE2 induced by arachidonic acid, inhibited serotonin-induced vasoconstriction, and potentiated vasodilation induced by exogenous PGE2. We conclude that pharmacologically inhibiting PGT-mediated prostaglandin metabolism lowers blood pressure, probably by prostaglandin-induced natriuresis and vasodilation. PGT is a novel therapeutic target for treating hypertension.


Asunto(s)
Presión Sanguínea/efectos de los fármacos , Hipertensión/metabolismo , Hipertensión/fisiopatología , Transportadores de Anión Orgánico/antagonistas & inhibidores , Prostaglandinas/metabolismo , Animales , Modelos Animales de Enfermedad , Hipertensión/tratamiento farmacológico , Ratones , Transportadores de Anión Orgánico/metabolismo , Ratas , Sodio/metabolismo , Sodio/orina , Tromboxanos/metabolismo , Triazinas/administración & dosificación , Triazinas/farmacología , Vasodilatación/efectos de los fármacos , para-Aminobenzoatos/administración & dosificación , para-Aminobenzoatos/farmacología
11.
Temperature (Austin) ; 2(3): 326-7, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-27227038
12.
Pharmacol Res Perspect ; 2(5): e00051, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25505603

RESUMEN

After synthesis and release from cells, prostaglandin E2 (PGE2) undergoes reuptake by the prostaglandin transporter (PGT), followed by cytoplasmic oxidation. Although genetic inactivation of PGT in mice and humans results in distinctive phenotypes, and although experiments in localized environments show that manipulating PGT alters downstream cellular events, a direct mechanistic link between PGT activity and PGE2 (EP) receptor activation has not been made. Toward this end, we created two reconstituted systems to examine the effect of PGT expression on PGE2 signaling via two of its receptors (EP1 and EP4). In human embryonic kidney cells engineered to express the EP1 receptor, exogenous PGE2 induced a dose-dependent increase in cytoplasmic Ca(2+). When PGT was expressed at the plasma membrane, the PGE2 dose-response curve was right-shifted, consistent with reduction in cell surface PGE2 availability; a potent PGT inhibitor acutely reversed this shift. When bradykinin was used to induce endogenous PGE2 release, PGT expression similarly induced a reduction in Ca(2+) responses. In separate experiments using Madin-Darby Canine Kidney cells engineered to express the PGE2 receptor EP4, bradykinin again induced autocrine PGE2 signaling, as judged by an abrupt increase in intracellular cAMP. As in the EP1 experiments, expression of PGT at the plasma membrane caused a reduction in bradykinin-induced cAMP accumulation. Pharmacological concentrations of exogenous PGE2 induced EP4 receptor desensitization, an effect that was mitigated by PGT. Thus, at an autocrine/paracrine level, plasma membrane PGT regulates PGE2 signaling by decreasing ligand availability at cell surface receptors.

13.
J Pharmacol Exp Ther ; 339(2): 633-41, 2011 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-21849625

RESUMEN

Prostaglandin E(2) (PGE(2)) triggers a vast array of biological signals and physiological events. The prostaglandin transporter (PGT) controls PGE(2) influx and is rate-limiting for PGE(2) metabolism and signaling termination. PGT global knockout mice die on postnatal day 1 from patent ductus arteriosus. A high-affinity PGT inhibitor would thus be a powerful tool for studying PGT function in adult animals. Moreover, such an inhibitor could be potentially developed into a therapeutic drug targeting PGT. Based on structure-activity relationship studies that built on recently identified inhibitors of PGT, we obtained N-(2-(2-(2-azidoethoxy)ethoxy)ethyl)-4-((4-((2-(2-(2-benzamidoethoxy)ethoxy)ethyl)amino)-6-((4-hydroxyphenyl)amino)-1,3,5-triazin-2-yl)amino)benzamide (T26A), a competitive inhibitor of PGT, with a K(i) of 378 nM. T26A seems to be highly selective for PGT, because it neither interacts with a PGT homolog in the organic anion transporter family nor affects PGE(2) synthesis. In Madin-Darby canine kidney cells stably transfected with PGT, T26A blocked PGE(2) metabolism, resulting in retention of PGE(2) in the extracellular compartment and the negligible appearance of PGE(2) metabolites in the intracellular compartment. Compared with vehicle, T26A injected intravenously into rats effectively doubled the amount of endogenous PGE(2) in the circulation and reduced the level of circulating endogenous PGE(2) metabolites to 50%. Intravenous T26A was also able to slow the metabolism of exogenously injected PGE(2). These results confirm that PGT directly regulates PGE(2) metabolism and demonstrate that a high-affinity inhibitor of PGT can effectively prevent PGE(2) metabolism and prolong the half-life of circulating PGE(2).


Asunto(s)
Dinoprostona/metabolismo , Conducto Arterioso Permeable/tratamiento farmacológico , Transportadores de Anión Orgánico/antagonistas & inhibidores , Triazinas/farmacología , para-Aminobenzoatos , Ácido 4-Aminobenzoico/química , Ácido 4-Aminobenzoico/metabolismo , Ácido 4-Aminobenzoico/farmacología , Animales , Transporte Biológico/efectos de los fármacos , Línea Celular , Grupos Control , Dinoprostona/sangre , Dinoprostona/química , Perros , Ensayos Analíticos de Alto Rendimiento , Concentración 50 Inhibidora , Oxidorreductasas Intramoleculares/metabolismo , Masculino , Ratones , Ratones Noqueados , Terapia Molecular Dirigida , Transportadores de Anión Orgánico/metabolismo , Prostaglandina-E Sintasas , Prostaglandina-Endoperóxido Sintasas/metabolismo , Ratas , Ratas Sprague-Dawley , Transducción de Señal , Relación Estructura-Actividad , Triazinas/química , Triazinas/metabolismo
14.
Biochem Biophys Res Commun ; 395(2): 168-72, 2010 Apr 30.
Artículo en Inglés | MEDLINE | ID: mdl-20346915

RESUMEN

Prostaglandin H(2) not only serves as the common precursor of all other PGs, but also directly triggers signals (e.g. platelet aggregation), depending on its location and translocation. The prostaglandin carrier PGT mediates the transport of several prostanoids, such as PGE(2), and PGF(2alpha). Here we used PGT in the plasma membrane as a model system to test the hypothesis that PGT also transports PGH(2). Using wild-type and PGT-expressing MDCK cells, we show that PGH(2) uptake is mediated both by simple diffusion and by PGT. The PGH(2) influx permeability coefficient for diffusion is (5.66+/-0.63)x10(-6)cm/s. The kinetic parameters of PGH(2) transport by PGT are K(m)=376+/-34nM and V(max)=210.2+/-11.4 fmol/mg protein/s. PGH(2) transport by PGT can be inhibited by excess PGE(2) or by a PGT inhibitor. We conclude that PGT may play a role in transporting PGH(2) across cellular membranes.


Asunto(s)
Membrana Celular/metabolismo , Transportadores de Anión Orgánico/metabolismo , Prostaglandina H2/metabolismo , Animales , Transporte Biológico , Perros , Humanos , Transportadores de Anión Orgánico/genética
15.
Circulation ; 121(4): 529-36, 2010 Feb 02.
Artículo en Inglés | MEDLINE | ID: mdl-20083684

RESUMEN

BACKGROUND: Prostaglandin E(2) (PGE(2)) plays a major role both in maintaining patency of the fetal ductus arteriosus and in closure of the ductus arteriosus after birth. The rate-limiting step in PGE(2) signal termination is PGE(2) uptake by the transporter PGT. METHODS AND RESULTS: To determine the role of PGT in ductus arteriosus closure, we used a gene-targeting strategy to produce mice in which PGT exon 1 was flanked by loxP sites. Successful targeting was obtained because neither mice hypomorphic at the PGT allele (PGT Neo/Neo) nor global PGT knockout mice (PGT(-/-)) exhibited PGT protein expression; moreover, embryonic fibroblasts isolated from targeted mice failed to exhibit carrier-mediated PGE(2) uptake. Although born in a normal mendelian ratio, no PGT(-/-) mice survived past postnatal day 1, and no PGT Neo/Neo mice survived past postnatal day 2. Necropsy revealed patent ductus arteriosus with normal intimal thickening but dilated cardiac chambers. Both PGT Neo/Neo and PGT(-/-) mice could be rescued through the postnatal period by giving the mother indomethacin before birth. Rescued mice grew normally and had no abnormalities by gross and microscopic postmortem analyses. In accordance with the known role of PGT in metabolizing PGE(2), rescued adult PGT(-/-) mice had lower plasma PGE(2) metabolite levels and higher urinary PGE(2) excretion rates than wild-type mice. CONCLUSIONS: PGT plays a critical role in closure of the ductus arteriosus after birth by ensuring a reduction in local and/or circulating PGE(2) concentrations.


Asunto(s)
Conducto Arterioso Permeable/etiología , Conducto Arterioso Permeable/metabolismo , Conducto Arterial/embriología , Conducto Arterial/metabolismo , Transportadores de Anión Orgánico/deficiencia , Animales , Fármacos Cardiovasculares/uso terapéutico , Células Cultivadas , Dinoprostona/metabolismo , Modelos Animales de Enfermedad , Conducto Arterial/patología , Conducto Arterioso Permeable/prevención & control , Femenino , Fibroblastos/metabolismo , Fibroblastos/patología , Indometacina/uso terapéutico , Pulmón/metabolismo , Pulmón/patología , Ratones , Ratones Noqueados , Transportadores de Anión Orgánico/genética , Transportadores de Anión Orgánico/metabolismo , Embarazo , Receptores de Prostaglandina E/metabolismo , Transducción de Señal/fisiología
16.
Am J Physiol Renal Physiol ; 295(3): F765-71, 2008 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-18579702

RESUMEN

Prostaglandin E(2) (PGE(2)) plays an important role in maintaining body fluid homeostasis by activating its receptors on the renal collecting duct (CD) to stimulate renal Na(+) and water excretion. The PG carrier prostaglandin transporter (PGT) is expressed on the CD apical membrane, where it mediates PG reuptake as part of the termination of autocrine PG signaling. Here we tested the hypothesis that dietary salt loading regulates PGT gene transcription in renal CDs. We placed green fluorescence protein (GFP) under control of 3.3 kb of the mouse PGT promoter and injected this construct into the pronuclei of fertilized FVB mouse eggs. Four of thirty-eight offspring were GFP positive by genotyping. We extensively characterized one (no. 29) PGT-GFP transgenic mouse line. On microscopic examination, GFP was expressed in CDs as determined by their expression of aquaporin-2. We fed mice a low (0.03% NaCl)-, normal (0.3% NaCl)-, or high-salt (3% NaCl) diet for 2 wk and quantified CD GFP expression. The average number of GFP-positive CD cells per microscopic section varied directly with dietary salt intake. Compared with mice on the control (0.3% sodium) diet, mice on a low-sodium (0.03%) diet had reduced numbers of GFP-positive cells (71% of control, P < 0.001), whereas mice on a high-sodium (3%) diet had increased numbers of GFP-positive cells (139% of control, P < 0.001). This increase in apparent CD PGT transcription resulted in a 51-55% increase (P < 0.001) in whole kidney PGT mRNA levels as determined by real-time PCR. The regulation of PG signal termination via reuptake represents a new pathway for controlling renal Na(+) balance.


Asunto(s)
Túbulos Renales Colectores/metabolismo , Transportadores de Anión Orgánico/metabolismo , Cloruro de Sodio Dietético/farmacología , Transcripción Genética/efectos de los fármacos , Animales , Cruzamiento , Desoxirribonucleasa BamHI/metabolismo , Desoxirribonucleasas de Localización Especificada Tipo II/metabolismo , Dosificación de Gen , Genes Reporteros , Genotipo , Proteínas Fluorescentes Verdes/genética , Ratones , Ratones Transgénicos , Regiones Promotoras Genéticas , Transgenes
18.
Am J Physiol Renal Physiol ; 290(3): F641-9, 2006 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-16263809

RESUMEN

During water deprivation, prostaglandin E(2) (PGE(2)), formed by renal medullary interstitial cells (RMICs), feedback inhibits the actions of antidiuretic hormone. Interstitial PGE(2) concentrations represent the net of both PGE(2) synthesis by cyclooxygenase (COX) and PGE(2) uptake by carriers such as PGT. We used cultured RMICs to examine the effects of hyperosmolarity on both PG synthesis and PG uptake in the same RMIC. RMICs expressed endogenous PGT as assessed by mRNA and immunoblotting. RMICs rapidly took up [(3)H]PGE(2) to a level 5- to 10-fold above background and with a characteristic time-dependent "overshoot." Inhibitory constants (K(i)) for various PGs and PGT inhibitors were similar between RMICs and the cloned rat PGT. Increasing extracellular hyperosmolarity to the range of 335-485 mosM increased the net release of PGE(2) by RMICs, an effect that was concentration dependent, maximal by 24 h, reversible, and associated with increased expression of COX-2. Over the same time period, there was decreased cell-surface activity of PGT due to internalization of the transporter. With continued exposure to hyperosmolarity over 7-10 days, PGE(2) release remained elevated, COX-2 returned to baseline, and PGT-mediated uptake became markedly reduced. Our findings suggest that hyperosmolarity induces coordinated changes in COX-2-mediated PGE(2) synthesis and PGT-mediated PGE(2) uptake in RMICs.


Asunto(s)
Fenómenos Fisiológicos Celulares , Dinoprostona/metabolismo , Médula Renal/citología , Médula Renal/fisiología , Transporte Biológico , Células Cultivadas , Dinoprostona/biosíntesis , Eicosanoides/metabolismo , Retroalimentación , Células HeLa , Humanos , Cinética , Vasopresinas/antagonistas & inhibidores
19.
J Pharmacol Exp Ther ; 316(3): 1346-50, 2006 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-16269530

RESUMEN

Prostaglandins (PGs) are involved in several major signaling pathways. Their effects are terminated when they are transported across cell membranes and oxidized intracellularly. The transport step of PG metabolism is carried out by the prostaglandin transporter (PGT). Inhibition of PGT would therefore be expected to change local or circulating concentrations of prostaglandins, and thus their biological effects. To develop PGT-specific inhibitors with high affinity, we designed a library of triazine compounds and screened 1842 small molecules by using Madin-Darby canine kidney cells stably expressing rat PGT. We found several effective PGT inhibitors. Among them, the most potent inhibitor had a Ki of 3.7 +/- 0.2 microM. These inhibitors allowed us to isolate the efflux process of PGE2 and to demonstrate that PGT does not transport PGE2 outwardly under physiological conditions.


Asunto(s)
Dinoprostona/metabolismo , Transportadores de Anión Orgánico/antagonistas & inhibidores , Células 3T3 , Animales , Transporte Biológico/efectos de los fármacos , Difusión , Perros , Ratones , Transportadores de Anión Orgánico/fisiología
20.
J Biol Chem ; 280(31): 28424-9, 2005 Aug 05.
Artículo en Inglés | MEDLINE | ID: mdl-15855165

RESUMEN

Prostaglandins mediate autacrine and paracrine signaling over short distances. We used the renal collecting duct as a model system to test the hypothesis that local control of prostaglandin signaling is achieved by expressing inactivation in the same cell as synthesis. Immunocytochemical studies demonstrated that renal collecting ducts in situ express the prostaglandin (PG) synthesis enzyme, cyclooxygenase-1 (COX-1), as well as both components of prostaglandin metabolic inactivation, i.e. the prostaglandin uptake carrier prostaglandin transporter (PGT) and the enzyme 15-hydroxyprostaglandin dehydrogenase. We characterized this system further using the collecting duct cell line Madin-Darby canine kidney (MDCK), which retains COX-2 and prostaglandin dehydrogenase expression but which has lost PGT expression. When we reintroduced PGT, it was correctly sorted to the apical membrane where it altered the sidedness of prostaglandin E2 (PGE2) release, a process we call "vectorial release via sided reuptake." Importantly, although COX-2 and prostaglandin dehydrogenase are expressed in the same MDCK cell, they must be compartmentalized because even in the presence of excess dehydrogenase newly synthesized PGE2 is released largely un-oxidized. However, when PGE2 undergoes first release and then PGT-mediated reuptake, significant oxidation takes place, suggesting that PGT imports PGE2 into the prostaglandin dehydrogenase compartment. Our data are consistent with a new model that offers significant new mechanisms for the fine control of eicosanoid signaling.


Asunto(s)
Túbulos Renales Colectores/fisiología , Prostaglandinas/fisiología , Animales , Línea Celular , Ciclooxigenasa 1 , Ciclooxigenasa 2 , Perros , Masculino , Proteínas de la Membrana , Oxidación-Reducción , Prostaglandina-Endoperóxido Sintasas/metabolismo , Prostaglandinas/biosíntesis , Ratas , Ratas Sprague-Dawley , Transducción de Señal
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...