Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Am J Physiol Renal Physiol ; 319(4): F712-F728, 2020 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-32893663

RESUMEN

Inhibitors of proximal tubular Na+-glucose cotransporter 2 (SGLT2) are natriuretic, and they lower blood pressure. There are reports that the activities of SGLT2 and Na+-H+ exchanger 3 (NHE3) are coordinated. If so, then part of the natriuretic response to an SGLT2 inhibitor is mediated by suppressing NHE3. To examine this further, we compared the effects of an SGLT2 inhibitor, empagliflozin, on urine composition and systolic blood pressure (SBP) in nondiabetic mice with tubule-specific NHE3 knockdown (NHE3-ko) and wild-type (WT) littermates. A single dose of empagliflozin, titrated to cause minimal glucosuria, increased urinary excretion of Na+ and bicarbonate and raised urine pH in WT mice but not in NHE3-ko mice. Chronic empagliflozin treatment tended to lower SBP despite higher renal renin mRNA expression and lowered the ratio of SBP to renin mRNA, indicating volume loss. This effect of empagliflozin depended on tubular NHE3. In diabetic Akita mice, chronic empagliflozin enhanced phosphorylation of NHE3 (S552/S605), changes previously linked to lesser NHE3-mediated reabsorption. Chronic empagliflozin also increased expression of genes involved with renal gluconeogenesis, bicarbonate regeneration, and ammonium formation. While this could reflect compensatory responses to acidification of proximal tubular cells resulting from reduced NHE3 activity, these effects were at least in part independent of tubular NHE3 and potentially indicated metabolic adaptations to urinary glucose loss. Moreover, empagliflozin increased luminal α-ketoglutarate, which may serve to stimulate compensatory distal NaCl reabsorption, while cogenerated and excreted ammonium balances urine losses of this "potential bicarbonate." The data implicate NHE3 as a determinant of the natriuretic effect of empagliflozin.


Asunto(s)
Compuestos de Bencidrilo/farmacología , Diabetes Mellitus/tratamiento farmacológico , Glucósidos/farmacología , Túbulos Renales Proximales/efectos de los fármacos , Natriuresis/efectos de los fármacos , Natriuréticos/farmacología , Inhibidores del Cotransportador de Sodio-Glucosa 2/farmacología , Transportador 2 de Sodio-Glucosa/metabolismo , Intercambiador 3 de Sodio-Hidrógeno/metabolismo , Equilibrio Ácido-Base/efectos de los fármacos , Animales , Glucemia/metabolismo , Presión Sanguínea/efectos de los fármacos , Diabetes Mellitus/metabolismo , Diabetes Mellitus/fisiopatología , Modelos Animales de Enfermedad , Glucosuria/metabolismo , Glucosuria/fisiopatología , Túbulos Renales Proximales/metabolismo , Túbulos Renales Proximales/fisiopatología , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Fosforilación , Intercambiador 3 de Sodio-Hidrógeno/deficiencia , Intercambiador 3 de Sodio-Hidrógeno/genética
2.
Nephron ; 144(8): 400-412, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32629454

RESUMEN

INTRODUCTION: Alström syndrome is a rare recessive genetic disease caused by mutations in ALMS1, which encodes a protein that is related to cilia function and intracellular endosome trafficking. The syndrome has been linked to impaired glucose metabolism and CKD. Polymorphisms in Alms1 have likewise been linked to CKD, but little is known about the modification of the phenotype by environmental factors. METHODS: To gain further insights, the fat aussie (foz) mouse strain, a genetic murine model of Alström syndrome, was exposed to a normal chow (NC) or to a Western diet (WD, 20% fat, 34% sucrose by weight, and 0.2% cholesterol) and renal outcomes were measured. RESULTS: Body weight and albuminuria were higher in foz than in wild-type (WT) mice on both diets but WD significantly increased the difference. Measurement of plasma creatinine and cystatin C indicated that glomerular filtration rate was preserved in foz versus WT independent of diet. Renal markers of injury, inflammation, and fibrosis were similar in both genotypes on NC but significantly greater in foz than in WT mice on WD. A glucose tolerance test performed in foz and WT mice on WD revealed similar basal blood glucose levels and subsequent blood glucose profiles. CONCLUSIONS: WD sensitizes a murine model of Alström syndrome to kidney injury, inflammation, and fibrosis, an effect that may not be solely due to effects on glucose metabolism. Polymorphisms in Alms1 may induce CKD in part by modulating the deleterious effects of high dietary fat and sucrose on kidney outcome.


Asunto(s)
Síndrome de Alstrom/complicaciones , Dieta Occidental/efectos adversos , Riñón/metabolismo , Riñón/patología , Nefritis/etiología , Animales , Glucemia/análisis , Proteínas de Ciclo Celular/genética , Cilios , Modelos Animales de Enfermedad , Fibrosis , Tasa de Filtración Glomerular , Glucosuria/etiología , Riñón/fisiopatología , Túbulos Renales/ultraestructura , Leptina/sangre , Masculino , Ratones , Nefritis/fisiopatología , Obesidad/etiología , Tamaño de los Órganos , Renina/genética , Renina/metabolismo
3.
Front Immunol ; 11: 1073, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32625203

RESUMEN

While B cells play a significant role in the onset of type-1 diabetes (T1D), little is know about their role in those early stages. Thus, to gain new insights into the role of B cells in T1D, we converted a physiological early pancreas-infiltrating B cell into a novel BCR mouse model using Somatic Cell Nuclear Transfer (SCNT). Strikingly, SCNT-derived B1411 model displayed neither developmental block nor anergy. Instead, B1411 underwent spontaneous germinal center reactions. Without T cell help, B1411-Rag1-/- was capable of forming peri-/intra-pancreatic lymph nodes, and undergoing class-switching. RNA-Seq analysis identified 93 differentially expressed genes in B1411 compared to WT B cells, including Irf7, Usp18, and Mda5 that had been linked to a potential viral etiology of T1D. We also found various members of the oligoadenylate synthase (OAS) family to be enriched in B1411, such as Oas1, which had recently also been linked to T1D. Strikingly, when challenged with glucose B1411-Rag1-/- mice displayed impaired glucose tolerance.


Asunto(s)
Autoinmunidad , Linfocitos B/inmunología , Estado Prediabético/etiología , Estado Prediabético/inmunología , Animales , Basidiomycota/genética , Basidiomycota/metabolismo , Señalización del Calcio/inmunología , Ensamble y Desensamble de Cromatina , Células Clonales/inmunología , Diabetes Mellitus Experimental/etiología , Diabetes Mellitus Experimental/genética , Diabetes Mellitus Experimental/inmunología , Femenino , Perfilación de la Expresión Génica , Prueba de Tolerancia a la Glucosa , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos DBA , Ratones Endogámicos NOD , Ratones Noqueados , Modelos Inmunológicos , Técnicas de Transferencia Nuclear , Estado Prediabético/genética , Receptores de Antígenos de Linfocitos B/genética , Receptores de Antígenos de Linfocitos B/inmunología
4.
Am J Physiol Renal Physiol ; 318(5): F1100-F1112, 2020 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-32116018

RESUMEN

In the early proximal tubule, Na+-glucose cotransporter 2 (SGLT2) mediates the bulk of renal glucose reabsorption. Gene deletion in mice (Sglt2-/-) was used to determine the role of SGLT2 in acute kidney injury induced by bilateral ischemia-reperfusion (IR). In Sglt2-/- and littermate wild-type mice, plasma creatinine increased similarly on day 1 after IR. This was associated with an equal increase in both genotypes in the urinary kidney injury molecule-1-to-creatinine ratio, a tubular injury marker, and similarly reduced urine osmolality and increased plasma osmolality, indicating impaired urine concentration. In both IR groups, FITC-sinistrin glomerular filtration rate was equally reduced on day 14, and plasma creatinine was similarly and incompletely restored on day 23. In Sglt2-/- mice subjected to IR, fractional urinary glucose excretion was increased on day 1 but reduced and associated with normal renal Na+-glucose cotransporter 1 (Sglt1) mRNA expression on day 23, suggesting temporary SGLT1 suppression. In wild-type mice subjected to IR, renal Sglt1 mRNA was likewise normal on day 23, whereas Sglt2 mRNA was reduced by 57%. In both genotypes, IR equally reduced urine osmolality and renal mRNA expression of the Na+-K+-2Cl- cotransporter and renin on day 23, suggesting thick ascending limb dysfunction, and similarly increased renal mRNA expression of markers of injury, inflammation, oxidative stress, and fibrosis (kidney injury molecule-1, neutrophil gelatinase-associated lipocalin, monocyte chemoattractant protein-1, transforming growth factor-ß1, NADPH oxidase-2, and collagen type 1). This was associated with equal increases in kidney histological damage scores and similar degree of capillary loss in both genotypes. The data indicate that genetic deletion of SGLT2 did not protect the kidneys in the initial injury phase or the subsequent recovery phase in a mouse model of IR-induced acute kidney injury.


Asunto(s)
Lesión Renal Aguda/metabolismo , Glucemia/metabolismo , Riñón/metabolismo , Daño por Reperfusión/metabolismo , Transportador 2 de Sodio-Glucosa/deficiencia , Lesión Renal Aguda/genética , Lesión Renal Aguda/patología , Lesión Renal Aguda/fisiopatología , Animales , Biomarcadores/sangre , Modelos Animales de Enfermedad , Tasa de Filtración Glomerular , Riñón/patología , Ratones Endogámicos C57BL , Ratones Noqueados , Eliminación Renal , Reabsorción Renal , Daño por Reperfusión/genética , Daño por Reperfusión/patología , Daño por Reperfusión/fisiopatología , Transportador 2 de Sodio-Glucosa/genética , Factores de Tiempo
5.
Am J Physiol Renal Physiol ; 317(2): F419-F434, 2019 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-31166707

RESUMEN

Na+/H+ exchanger isoform 3 (NHE3) contributes to Na+/bicarbonate reabsorption and ammonium secretion in early proximal tubules. To determine its role in the diabetic kidney, type 1 diabetic Akita mice with tubular NHE3 knockdown [Pax8-Cre; NHE3-knockout (KO) mice] were generated. NHE3-KO mice had higher urine pH, more bicarbonaturia, and compensating increases in renal mRNA expression for genes associated with generation of ammonium, bicarbonate, and glucose (phosphoenolpyruvate carboxykinase) in proximal tubules and H+ and ammonia secretion and glycolysis in distal tubules. This left blood pH and bicarbonate unaffected in nondiabetic and diabetic NHE3-KO versus wild-type mice but was associated with renal upregulation of proinflammatory markers. Higher renal phosphoenolpyruvate carboxykinase expression in NHE3-KO mice was associated with lower Na+-glucose cotransporter (SGLT)2 and higher SGLT1 expression, indicating a downward tubular shift in Na+ and glucose reabsorption. NHE3-KO was associated with lesser kidney weight and glomerular filtration rate (GFR) independent of diabetes and prevented diabetes-associated albuminuria. NHE3-KO, however, did not attenuate hyperglycemia or prevent diabetes from increasing kidney weight and GFR. Higher renal gluconeogenesis may explain similar hyperglycemia despite lower SGLT2 expression and higher glucosuria in diabetic NHE3-KO versus wild-type mice; stronger SGLT1 engagement could have affected kidney weight and GFR responses. Chronic kidney disease in humans is associated with reduced urinary excretion of metabolites of branched-chain amino acids and the tricarboxylic acid cycle, a pattern mimicked in diabetic wild-type mice. This pattern was reversed in nondiabetic NHE3-KO mice, possibly reflecting branched-chain amino acids use for ammoniagenesis and tricarboxylic acid cycle upregulation to support formation of ammonia, bicarbonate, and glucose in proximal tubule. NHE3-KO, however, did not prevent the diabetes-induced urinary downregulation in these metabolites.


Asunto(s)
Equilibrio Ácido-Base , Diabetes Mellitus Tipo 1/metabolismo , Nefropatías Diabéticas/metabolismo , Túbulos Renales/metabolismo , Reabsorción Renal , Intercambiador 3 de Sodio-Hidrógeno/deficiencia , Sodio/orina , Equilibrio Ácido-Base/genética , Aminoácidos de Cadena Ramificada/orina , Amoníaco/orina , Animales , Bicarbonatos/orina , Biomarcadores/orina , Glucemia/metabolismo , Diabetes Mellitus Tipo 1/genética , Diabetes Mellitus Tipo 1/fisiopatología , Diabetes Mellitus Tipo 1/orina , Nefropatías Diabéticas/genética , Nefropatías Diabéticas/fisiopatología , Nefropatías Diabéticas/orina , Modelos Animales de Enfermedad , Metabolismo Energético/genética , Regulación de la Expresión Génica , Técnicas de Silenciamiento del Gen , Concentración de Iones de Hidrógeno , Túbulos Renales/fisiopatología , Masculino , Metabolómica/métodos , Ratones Endogámicos C57BL , Ratones Noqueados , Transportador 1 de Sodio-Glucosa/genética , Transportador 1 de Sodio-Glucosa/metabolismo , Transportador 2 de Sodio-Glucosa/genética , Transportador 2 de Sodio-Glucosa/metabolismo , Intercambiador 3 de Sodio-Hidrógeno/genética
6.
Am J Physiol Renal Physiol ; 317(1): F207-F217, 2019 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-31091127

RESUMEN

Na+-glucose cotransporter (SGLT)1 mediates glucose reabsorption in late proximal tubules. SGLT1 also mediates macula densa (MD) sensing of an increase in luminal glucose, which increases nitric oxide (NO) synthase 1 (MD-NOS1)-mediated NO formation and potentially glomerular filtratrion rate (GFR). Here, the contribution of SGLT1 was tested by gene knockout (-/-) in type 1 diabetic Akita mice. A low-glucose diet was used to prevent intestinal malabsorption in Sglt1-/- mice and minimize the contribution of intestinal SGLT1. Hyperglycemia was modestly reduced in Sglt1-/- versus littermate wild-type Akita mice (480 vs. 550 mg/dl), associated with reduced diabetes-induced increases in GFR, kidney weight, glomerular size, and albuminuria. Blunted hyperfiltration was confirmed in streptozotocin-induced diabetic Sglt1-/- mice, associated with similar hyperglycemia versus wild-type mice (350 vs. 385 mg/dl). Absence of SGLT1 attenuated upregulation of MD-NOS1 protein expression in diabetic Akita mice and in response to SGLT2 inhibition in nondiabetic mice. During SGLT2 inhibition in Akita mice, Sglt1-/- mice had likewise reduced blood glucose (200 vs. 300 mg/dl), associated with lesser MD-NOS1 expression, GFR, kidney weight, glomerular size, and albuminuria. Absence of Sglt1 in Akita mice increased systolic blood pressure, associated with suppressed renal renin mRNA expression. This may reflect fluid retention due to blunted hyperfiltration. SGLT2 inhibition prevented the blood pressure increase in Sglt1-/- Akita mice, possibly due to additive glucosuric/diuretic effects. The data indicate that SGLT1 contributes to diabetic hyperfiltration and limits diabetic hypertension. Potential mechanisms include its role in glucose-driven upregulation of MD-NOS1 expression. This pathway may increase GFR to maintain volume balance when enhanced MD glucose delivery indicates upstream saturation of SGLTs and thus hyperreabsorption.


Asunto(s)
Glucemia/metabolismo , Diabetes Mellitus Experimental/enzimología , Diabetes Mellitus Tipo 1/enzimología , Nefropatías Diabéticas/enzimología , Tasa de Filtración Glomerular , Riñón/enzimología , Óxido Nítrico Sintasa de Tipo I/metabolismo , Transportador 1 de Sodio-Glucosa/deficiencia , Albuminuria/enzimología , Albuminuria/genética , Albuminuria/fisiopatología , Animales , Biomarcadores/sangre , Glucemia/efectos de los fármacos , Presión Sanguínea , Diabetes Mellitus Experimental/sangre , Diabetes Mellitus Experimental/tratamiento farmacológico , Diabetes Mellitus Experimental/fisiopatología , Diabetes Mellitus Tipo 1/sangre , Diabetes Mellitus Tipo 1/tratamiento farmacológico , Diabetes Mellitus Tipo 1/fisiopatología , Nefropatías Diabéticas/sangre , Nefropatías Diabéticas/tratamiento farmacológico , Nefropatías Diabéticas/fisiopatología , Tasa de Filtración Glomerular/efectos de los fármacos , Riñón/efectos de los fármacos , Riñón/fisiopatología , Ratones Endogámicos C57BL , Ratones Endogámicos DBA , Ratones Noqueados , Óxido Nítrico Sintasa de Tipo I/genética , Reabsorción Renal , Renina/sangre , Renina/genética , Transducción de Señal , Transportador 1 de Sodio-Glucosa/genética , Transportador 2 de Sodio-Glucosa , Inhibidores del Cotransportador de Sodio-Glucosa 2/farmacología , Regulación hacia Arriba
7.
Am J Physiol Renal Physiol ; 316(6): F1201-F1210, 2019 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-30995111

RESUMEN

Renal Na+-glucose cotransporter SGLT1 mediates glucose reabsorption in the late proximal tubule, a hypoxia-sensitive tubular segment that enters the outer medulla. Gene deletion in mice (Sglt1-/-) was used to determine the role of the cotransporter in acute kidney injury induced by ischemia-reperfusion (IR), including the initial injury and subsequent recovery phase. On days 1 and 16 after IR, absolute and fractional urinary glucose excretion remained greater in Sglt1-/- mice versus wild-type (WT) littermates, consistent with a sustained contribution of SGLT1 to tubular glucose reabsorption in WT mice. Absence of SGLT1 did not affect the initial kidney impairment versus WT mice, as indicated by similar increases on day 1 in plasma concentrations of creatinine and urinary excretion of the tubular injury marker kidney injury molecule-1 as well as a similar rise in plasma osmolality and fall in urine osmolality as indicators of impaired urine concentration. Recovery of kidney function on days 14/16, however, was improved in Sglt1-/- versus WT mice, as indicated by lower plasma creatinine, higher glomerula filtration rate (by FITC-sinistrin in awake mice), and more completely restored urine and plasma osmolality. This was associated with a reduced tubular injury score in the cortex and outer medulla, better preserved renal mRNA expression of tubular transporters (Sglt2 and Na+-K+-2Cl- cotransporter Nkcc2), and a lesser rise in renal mRNA expression of markers of injury, inflammation, and fibrosis [kidney injury molecule-1, chemokine (C-C motif) ligand 2, fibronectin 1, and collagen type I-α1] in Sglt1-/- versus WT mice. These results suggest that SGLT1 activity in the late proximal tubule may have deleterious effects during recovery of IR-induced acute kidney injury and identify SGLT1 as a potential therapeutic target.


Asunto(s)
Lesión Renal Aguda/metabolismo , Tasa de Filtración Glomerular , Glucosa/metabolismo , Túbulos Renales Proximales/metabolismo , Reabsorción Renal , Daño por Reperfusión/metabolismo , Transportador 1 de Sodio-Glucosa/deficiencia , Lesión Renal Aguda/genética , Lesión Renal Aguda/patología , Lesión Renal Aguda/fisiopatología , Animales , Modelos Animales de Enfermedad , Eliminación de Gen , Túbulos Renales Proximales/patología , Túbulos Renales Proximales/fisiopatología , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Recuperación de la Función , Daño por Reperfusión/genética , Daño por Reperfusión/patología , Daño por Reperfusión/fisiopatología , Transportador 1 de Sodio-Glucosa/genética , Transportador 2 de Sodio-Glucosa/genética , Transportador 2 de Sodio-Glucosa/metabolismo , Miembro 1 de la Familia de Transportadores de Soluto 12/genética , Miembro 1 de la Familia de Transportadores de Soluto 12/metabolismo , Factores de Tiempo
8.
Am J Physiol Renal Physiol ; 316(1): F173-F185, 2019 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-30427222

RESUMEN

Inhibitors of the Na+-glucose cotransporter SGLT2 enhance urinary glucose and urate excretion and lower plasma urate levels. The mechanisms remain unclear, but a role for enhanced glucose in the tubular fluid, which may interact with tubular urate transporters, such as the glucose transporter GLUT9 or the urate transporter URAT1, has been proposed. Studies were performed in nondiabetic mice treated with the SGLT2 inhibitor canagliflozin and in gene-targeted mice lacking the urate transporter Glut9 in the tubule or in mice with whole body knockout of Sglt2, Sglt1, or Urat1. Renal urate handling was assessed by analysis of urate in spontaneous plasma and urine samples and normalization to creatinine concentrations or by renal clearance studies with assessment of glomerular filtration rate by FITC-sinistrin. The experiments confirmed the contribution of URAT1 and GLUT9 to renal urate reabsorption, showing a greater contribution of the latter and additive effects. Genetic and pharmacological inhibition of SGLT2 enhanced fractional renal urate excretion (FE-urate), indicating that a direct effect of the SGLT2 inhibitor on urate transporters is not absolutely necessary. Consistent with a proposed role of increased luminal glucose delivery, the absence of Sglt1, which by itself had no effect on FE-urate, enhanced the glycosuric and uricosuric effects of the SGLT2 inhibitor. The SGLT2 inhibitor enhanced renal mRNA expression of Glut9 in wild-type mice, but tubular GLUT9 seemed dispensable for the increase in FE-urate in response to canagliflozin. First evidence is presented that URAT1 is required for the acute uricosuric effect of the SGLT2 inhibitor in mice.


Asunto(s)
Canagliflozina/farmacología , Proteínas Facilitadoras del Transporte de la Glucosa/metabolismo , Túbulos Renales Proximales/efectos de los fármacos , Transportadores de Anión Orgánico/metabolismo , Eliminación Renal/efectos de los fármacos , Inhibidores del Cotransportador de Sodio-Glucosa 2/farmacología , Transportador 2 de Sodio-Glucosa/efectos de los fármacos , Ácido Úrico/orina , Uricosúricos/farmacología , Animales , Genotipo , Proteínas Facilitadoras del Transporte de la Glucosa/deficiencia , Proteínas Facilitadoras del Transporte de la Glucosa/genética , Túbulos Renales Proximales/metabolismo , Ratones Endogámicos C57BL , Ratones Noqueados , Transportadores de Anión Orgánico/deficiencia , Transportadores de Anión Orgánico/genética , Fenotipo , Reabsorción Renal/efectos de los fármacos , Transportador 2 de Sodio-Glucosa/deficiencia , Transportador 2 de Sodio-Glucosa/genética , Transportador 2 de Sodio-Glucosa/metabolismo
10.
Am J Physiol Renal Physiol ; 315(2): F386-F394, 2018 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-29412698

RESUMEN

The sodium-glucose cotransporter SGLT2 inhibitor empagliflozin (plasma protein binding ~88%) may reach its target in the brush border of the early proximal tubule by glomerular filtration and tubular secretion. Here we determined whether empagliflozin is secreted by renal tubules in mice and whether genetic knockout of the basolateral organic anion transporter 3 ( Oat3-/-) affects its tubular secretion or glucosuric effect. Renal clearance studies in wild-type (WT) mice showed that tubular secretion accounted for 50-70% of empagliflozin urinary excretion. Immunostaining indicated that SGLT2 and OAT3 localization partially overlapped in proximal tubule S1 and S2 segments. Glucosuria in metabolic cage studies was reduced in Oat3-/- vs. WT mice for acute empagliflozin doses of 1, 3, and 10 mg/kg, whereas 30 mg/kg induced similar maximal glucosuria in both genotypes. Chronic application of empagliflozin (~25 mg·kg-1 ·day-1) in Oat3-/- mice was associated with lower urinary glucose-to-creatinine ratios despite maintaining slightly higher blood glucose levels than WT. On a whole kidney level, renal secretion of empagliflozin was largely unchanged in Oat3-/- mice. However, the absence of OAT3 attenuated the influence of empagliflozin on fractional glucose excretion; higher levels of plasma or filtered empagliflozin were needed to induce similar increases in fractional renal glucose excretion. We conclude that empagliflozin is excreted into the urine to similar extent by glomerular filtration and tubular secretion. The latter can occur largely independent of OAT3. However, OAT3 increases the glucosuric effect of empagliflozin, which may relate to the partial overlap of its localization with SGLT2 and thus OAT3-mediated tubular secretion of empagliflozin in the early proximal tubule.


Asunto(s)
Compuestos de Bencidrilo/farmacología , Glucemia/efectos de los fármacos , Glucósidos/farmacología , Glucosuria/metabolismo , Túbulos Renales Proximales/efectos de los fármacos , Transportadores de Anión Orgánico Sodio-Independiente/metabolismo , Eliminación Renal , Inhibidores del Cotransportador de Sodio-Glucosa 2/farmacología , Transportador 2 de Sodio-Glucosa/efectos de los fármacos , Animales , Compuestos de Bencidrilo/farmacocinética , Compuestos de Bencidrilo/orina , Glucemia/metabolismo , Tasa de Filtración Glomerular , Glucósidos/farmacocinética , Glucósidos/orina , Glucosuria/genética , Glucosuria/prevención & control , Túbulos Renales Proximales/metabolismo , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Proteína 1 de Transporte de Anión Orgánico/genética , Proteína 1 de Transporte de Anión Orgánico/metabolismo , Transportadores de Anión Orgánico Sodio-Independiente/deficiencia , Transportadores de Anión Orgánico Sodio-Independiente/genética , Transportador 2 de Sodio-Glucosa/metabolismo , Inhibidores del Cotransportador de Sodio-Glucosa 2/farmacocinética , Inhibidores del Cotransportador de Sodio-Glucosa 2/orina
11.
J Emerg Med ; 43(2): 308-15, 2012 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-21497476

RESUMEN

BACKGROUND: Acute ischemic stroke diagnosis and treatment are among the most challenging in Emergency Medicine. Perfusion computed tomography (CTP) can increase the sensitivity for detecting ischemic stroke and, especially with the addition of CT angiography (CTA), improve decision-making regarding thrombolytic therapy compared to non-contrast computed tomography (NCCT) alone. However, because acute stroke protocols do not generally include procedures for multimodal imaging, they are not commonly performed. In addition, there is concern that additional studies could delay or preclude therapy in patients otherwise eligible for thrombolytic therapy. OBJECTIVES: To demonstrate the feasibility of perfusion CTP and CTA in addition to NCCT of the brain in the emergency assessment of patients with acute ischemic stroke. METHODS: Starting January 2008, multimodal (CTP and CTA) imaging was added to NCCT in the Emergency Department (ED) initial assessment of patients with stroke of ≤ 5 h duration. Over the subsequent 9 months, we measured the time from ED arrival to imaging and to recombinant tissue plasminogen activator (rt-PA) treatment and compared these times to patients evaluated with CT alone. RESULTS: From January to October 2008, 95 patients had CTP and CTA studies in addition to NCCT for acute ischemic stroke. There were no differences between the average time to CT study or to rt-PA treatment between patients evaluated with multimodal CT imaging and patients assessed with NCCT alone. CONCLUSIONS: Combining CTP and CTA with NCCT is feasible and does not adversely increase the time to CT imaging or rt-PA treatment in patients with acute ischemic stroke.


Asunto(s)
Isquemia Encefálica/diagnóstico por imagen , Perfusión , Accidente Cerebrovascular/diagnóstico por imagen , Tomografía Computarizada por Rayos X/métodos , Isquemia Encefálica/tratamiento farmacológico , Angiografía Cerebral , Medios de Contraste , Estudios de Factibilidad , Femenino , Humanos , Masculino , Persona de Mediana Edad , Accidente Cerebrovascular/tratamiento farmacológico , Factores de Tiempo , Activador de Tejido Plasminógeno/administración & dosificación
12.
Arch Gen Psychiatry ; 62(4): 417-22, 2005 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-15809409

RESUMEN

BACKGROUND: Antidepressant medication prevents the return of depressive symptoms, but only as long as treatment is continued. OBJECTIVES: To determine whether cognitive therapy (CT) has an enduring effect and to compare this effect against the effect produced by continued antidepressant medication. DESIGN: Patients who responded to CT in a randomized controlled trial were withdrawn from treatment and compared during a 12-month period with medication responders who had been randomly assigned to either continuation medication or placebo withdrawal. Patients who survived the continuation phase without relapse were withdrawn from all treatment and observed across a subsequent 12-month naturalistic follow-up. SETTING: Outpatient clinics at the University of Pennsylvania and Vanderbilt University. PATIENTS: A total of 104 patients responded to treatment (57.8% of those initially assigned) and were enrolled in the subsequent continuation phase; patients were initially selected to represent those with moderate to severe depression. INTERVENTIONS: Patients withdrawn from CT were allowed no more than 3 booster sessions during continuation; patients assigned to continuation medication were kept at full dosage levels. MAIN OUTCOME MEASURES: Relapse was defined as a return, for at least 2 weeks, of symptoms sufficient to meet the criteria for major depression or Hamilton Depression Rating Scale scores of 14 or higher during the continuation phase. Recurrence was defined in a comparable fashion during the subsequent naturalistic follow-up. RESULTS: Patients withdrawn from CT were significantly less likely to relapse during continuation than patients withdrawn from medications (30.8% vs 76.2%; P = .004), and no more likely to relapse than patients who kept taking continuation medication (30.8% vs 47.2%; P = .20). There were also indications that the effect of CT extends to the prevention of recurrence. CONCLUSIONS: Cognitive therapy has an enduring effect that extends beyond the end of treatment. It seems to be as effective as keeping patients on medication.


Asunto(s)
Antidepresivos/uso terapéutico , Terapia Cognitivo-Conductual , Trastorno Depresivo/terapia , Adulto , Atención Ambulatoria , Antidepresivos/administración & dosificación , Trastorno Depresivo/tratamiento farmacológico , Trastorno Depresivo/psicología , Esquema de Medicación , Quimioterapia Combinada , Femenino , Estudios de Seguimiento , Humanos , Masculino , Encuestas Nutricionales , Paroxetina/uso terapéutico , Pacientes Desistentes del Tratamiento , Placebos , Escalas de Valoración Psiquiátrica , Prevención Secundaria , Índice de Severidad de la Enfermedad
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...