Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
AACE Clin Case Rep ; 8(1): 15-18, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35097195

RESUMEN

OBJECTIVE: We report a case of pituitary metastasis (PM) presenting with acute anterior and posterior pituitary dysfunction following a two-decade-long oncologic course marked by disease progression. CASE REPORT: An elderly woman with a history of stage IIA invasive ductal carcinoma of the breast presented with confusion. Her laboratory evaluation was significant for panhypopituitarism and central diabetes insipidus, and magnetic resonance imaging findings were suggestive of PM. She was treated with hormone replacement, resulting in the reversal of her metabolic and cognitive derangements. DISCUSSION: PM is a rare complication of advanced malignancy. Although several malignancies may spread to the pituitary, the most common are breast cancer in women and lung cancer in men. Unlike pituitary adenomas, which predominantly involve the anterior pituitary, PM has a predilection for the posterior lobe and infundibulum due to direct access via systemic circulation. The clinical presentation of PM depends on the size of the metastatic deposit and other structures involved in the vicinity of the sella. Magnetic resonance imaging with gadolinium is the gold standard for the evaluation of sellar masses. The diagnosis of PM involves a thorough history, physical examination, biochemical evaluation of the hypothalamic-pituitary axis, and imaging studies. CONCLUSION: Metastatic involvement of the pituitary is a rare condition seen in <2% of resected pituitary masses. The clinical presentation is heterogeneous and can include headache, visual impairment, and panhypopituitarism. Unfortunately, the presence of PM portends a poor prognosis, and the median survival rate after diagnosis is 6 to 13.6 months.

2.
Am J Physiol Renal Physiol ; 306(9): F1059-68, 2014 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-24598799

RESUMEN

Dietary potassium loading results in rapid kaliuresis, natriuresis, and diuresis associated with reduced phosphorylation (p) of the distal tubule Na(+)-Cl(-) cotransporter (NCC). Decreased NCC-p inhibits NCC-mediated Na(+) reabsorption and shifts Na(+) downstream for reabsorption by epithelial Na(+) channels (ENaC), which can drive K(+) secretion. Whether the signal is initiated by ingesting potassium or a rise in plasma K(+) concentration ([K(+)]) is not understood. We tested the hypothesis, in male rats, that an increase in plasma [K(+)] is sufficient to reduce NCC-p and drive kaliuresis. After an overnight fast, a single 3-h 2% potassium (2%K) containing meal increased plasma [K(+)] from 4.0 ± 0.1 to 5.2 ± 0.2 mM; increased urinary K(+), Na(+), and volume excretion; decreased NCC-p by 60%; and marginally reduced cortical Na(+)-K(+)-2Cl(-) cotransporter (NKCC) phosphorylation 25% (P = 0.055). When plasma [K(+)] was increased by tail vein infusion of KCl to 5.5 ± 0.1 mM over 3 h, significant kaliuresis and natriuresis ensued, NCC-p decreased by 60%, and STE20/SPS1-related proline alanine-rich kinase (SPAK) phosphorylation was marginally reduced 35% (P = 0.052). The following were unchanged at 3 h by either the potassium-rich meal or KCl infusion: Na(+)/H(+) exchanger 3 (NHE3), NHE3-p, NKCC, ENaC subunits, and renal outer medullary K(+) channel. In summary, raising plasma [K(+)] by intravenous infusion to a level equivalent to that observed after a single potassium-rich meal triggers renal kaliuretic and natriuretic responses, independent of K(+) ingestion, likely driven by decreased NCC-p and activity sufficient to shift sodium reabsorption downstream to where Na(+) reabsorption and flow drive K(+) secretion.


Asunto(s)
Hiperpotasemia/sangre , Riñón/metabolismo , Natriuresis , Potasio/sangre , Sodio/orina , Animales , Modelos Animales de Enfermedad , Canales Epiteliales de Sodio/metabolismo , Hiperpotasemia/inducido químicamente , Hiperpotasemia/fisiopatología , Hiperpotasemia/orina , Infusiones Intravenosas , Riñón/fisiopatología , Masculino , Fosforilación , Potasio/administración & dosificación , Potasio/orina , Canales de Potasio/metabolismo , Potasio en la Dieta/sangre , Potasio en la Dieta/orina , Proteínas Serina-Treonina Quinasas/metabolismo , Ratas , Ratas Sprague-Dawley , Sodio/sangre , Intercambiador 3 de Sodio-Hidrógeno , Intercambiadores de Sodio-Hidrógeno/metabolismo , Miembro 3 de la Familia de Transportadores de Soluto 12/metabolismo , Factores de Tiempo
3.
Am J Physiol Renal Physiol ; 305(4): F510-9, 2013 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-23720346

RESUMEN

During angiotensin II (ANG II)-dependent hypertension, ANG II stimulates, while hypertension inhibits, Na(+) transporter activity to balance Na(+) output to input. This study tests the hypothesis that ANG II infusion activates Na(+) transporters in the distal nephron while inhibiting transporters along the proximal nephron. Male Sprague-Dawley rats were infused with ANG II (400 ng·kg(-1)·min(-1)) or vehicle for 2 wk. Kidneys were dissected (cortex vs. medulla) or fixed for immunohistochemistry (IHC). ANG II increased mean arterial pressure by 40 mmHg, urine Na(+) by 1.67-fold, and urine volume by 3-fold, evidence for hypertension and pressure natriuresis. Na(+) transporters' abundance and activation [assessed by phosphorylation (-P) or proteolytic cleavage] were measured by immunoblot. During ANG II infusion Na(+)/H(+) exchanger 3 (NHE3) abundance decreased in both cortex and medulla; Na-K-2Cl cotransporter 2 (NKCC2) decreased in medullary thick ascending loop of Henle (TALH) and increased, along with NKCC2-P, in cortical TALH; Na-Cl cotransporter (NCC) and NCC-P increased in the distal convoluted tubule; and epithelial Na(+) channel subunits and their cleaved forms were increased in both cortex and medulla. Like NKCC2, STE20/SPS1-related proline alanine-rich kinase (SPAK) and SPAK-P were decreased in medulla and increased in cortex. By IHC, during ANG II NHE3 remained localized to proximal tubule microvilli at lower abundance, and the differential regulation of NKCC2 and NKCC2-P in cortex versus medulla was evident. In summary, ANG II infusion increases Na(+) transporter abundance and activation from cortical TALH to medullary collecting duct while the hypertension drives a natriuresis response evident as decreased Na(+) transporter abundance and activation from proximal tubule through medullary TALH.


Asunto(s)
Angiotensina II/farmacología , Presión Sanguínea/efectos de los fármacos , Canales Epiteliales de Sodio/metabolismo , Hipertensión/metabolismo , Riñón/metabolismo , Proteínas de Transporte de Membrana/metabolismo , Nefronas/metabolismo , Sodio/metabolismo , Animales , Canales Epiteliales de Sodio/efectos de los fármacos , Immunoblotting , Inmunohistoquímica , Masculino , Proteínas de Transporte de Membrana/efectos de los fármacos , Nefronas/efectos de los fármacos , Ratas , Ratas Sprague-Dawley
4.
Am J Physiol Cell Physiol ; 304(2): C147-63, 2013 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-23114965

RESUMEN

The renal distal tubule Na-Cl cotransporter (NCC) reabsorbs <10% of the filtered Na(+) but is a key control point for blood pressure regulation by angiotensin II (ANG II), angiotensin-converting enzyme inhibitors (ACEI), and thiazide diuretics. This study aimed to determine whether NCC phosphorylation (NCCp) was regulated by acute (20-30 min) treatment with the ACEI captopril (12 µg/min × 20 min) or by a sub-pressor dose of ANG II (20 ng·kg(-1)·min(-1)) in Inactin-anesthetized rats. By immuno-EM, NCCp was detected exclusively in or adjacent to apical plama membranes (APM) in controls and after ACEI or ANG II treatment, while NCC total was detected in both APM and subapical cytoplasmic vesicles (SCV) in all conditions. In renal homogenates, neither ACEI nor ANG II treatment altered NCCp abundance, assayed by immunoblot. However, by density gradient fractionation we identified a pool of low-density APM in which NCCp decreased 50% in response to captopril and was restored during ANG II infusion, and another pool of higher-density APM that responded reciprocally, indicative of regulated redistribution between two APM pools. In both pools, NCCp was preferentially localized to Triton-soluble membranes. Blue Native gel electrophoresis established that APM NCCp localized to ~700 kDa complexes (containing γ-adducin) while unphosphorylated NCC in intracellular membranes primarily localized to ~400 kDa complexes: there was no evidence for native monomeric or dimeric NCC or NCCp. In summary, this study demonstrates that phosphorylated NCC, localized to multimeric complexes in the APM, redistributes in a regulated manner within the APM in response to ACEI and ANG II.


Asunto(s)
Angiotensina II/farmacología , Inhibidores de la Enzima Convertidora de Angiotensina/farmacología , Túbulos Renales Distales/metabolismo , Simportadores del Cloruro de Sodio/metabolismo , Animales , Proteínas de Unión a Calmodulina/análisis , Captopril/farmacología , Túbulos Renales Distales/citología , Túbulos Renales Distales/efectos de los fármacos , Masculino , Fosforilación , Ratas , Ratas Sprague-Dawley , Inhibidores de los Simportadores del Cloruro de Sodio/farmacología
5.
Am J Physiol Renal Physiol ; 303(1): F92-104, 2012 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-22496411

RESUMEN

Dietary potassium (K(+)) restriction and hypokalemia have been reported to change the abundance of most renal Na(+) and K(+) transporters and aquaporin-2 isoform, but results have not been consistent. The aim of this study was to reexamine Na(+), K(+) and H(2)O transporters' pool size regulation in response to removing K(+) from a diet containing 0.74% NaCl, as well as from a diet containing 2% NaCl (as found in American diets) to blunt reducing total diet electrolytes. Sprague-Dawley rats (n = 5-6) were fed for 6 days with one of these diets: 2% KCl, 0.74% NaCl (2K1Na, control chow) compared with 0.03% KCl, 0.74% NaCl (0K1Na); or 2% KCl, 2%NaCl (2K2Na) compared with 0.03% KCl, 2% NaCl (0K2Na, Na(+) replete). In both 0K1Na and 0K2Na there were significant decreases in: 1) plasma [K(+)] (<2.5 mM); 2) urinary K(+) excretion (<5% of control); 3) urine osmolality and plasma [aldosterone], as well as 4) an increase in urine volume and medullary hypertrophy. The 0K2Na group had the lowest [aldosterone] (172.0 ± 17.4 pg/ml) and lower blood pressure (93.2 ± 4.9 vs. 112.0 ± 3.1 mmHg in 2K2Na). Transporter pool size regulation was determined by quantitative immunoblotting of renal cortex and medulla homogenates. The only differences measured in both 0K1Na and 0K2Na groups were a 20-30% decrease in cortical ß-ENaC, 30-40% increases in kidney-specific Ste20/SPS1-related proline/alanine-rich kinase, and a 40% increase in medullary sodium pump abundance. The following proteins were not significantly changed in both the 0 K groups: Na(+)/H(+) exchanger isoform 3; Na(+)-K(+)-Cl(-) cotransporter; Na(+)-Cl(-) cotransporter, oxidative stress response kinase-1; renal outer medullary K(+) channel; autosomal recessive hypercholesterolemia; c-Src, aquaporin 2 isoform; or renin. Thus, despite profound hypokalemia and renal K(+) conservation, we did not confirm many of the changes that were previously reported. We predict that changes in transporter distribution and activity are likely more important for conserving K(+) than changes in total abundance.


Asunto(s)
Canales Epiteliales de Sodio/metabolismo , Nefronas/metabolismo , Deficiencia de Potasio/metabolismo , Potasio en la Dieta/farmacología , Cloruro de Sodio Dietético/farmacología , Intercambiadores de Sodio-Hidrógeno/metabolismo , Simportadores de Cloruro de Sodio-Potasio/metabolismo , Animales , Masculino , Nefronas/efectos de los fármacos , Fosforilación/efectos de los fármacos , Ratas , Ratas Sprague-Dawley
6.
Am J Physiol Renal Physiol ; 296(4): F810-8, 2009 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-19144688

RESUMEN

When blood pressure (BP) is elevated above baseline, a pressure natriuresis-diuresis response ensues, critical to volume and BP homeostasis. Distal convoluted tubule Na(+)-Cl(-) cotransporter (NCC) is regulated by trafficking between the apical plasma membrane (APM) and subapical cytoplasmic vesicles (SCV). We aimed to determine whether NCC trafficking contributes to pressure diuresis by decreasing APM NCC or compensates for increased volume flow to the DCT by increasing APM NCC. BP was raised 50 mmHg (high BP) in rats by arterial constriction for 5 or 20-30 min, provoking a 10-fold diuresis at both times. Kidneys were excised, and NCC subcellular distribution was analyzed by 1) sorbitol density gradient fractionation and immunoblotting and 2) immunoelectron microscopy (immuno-EM). NCC distribution did not change after 5-min high BP. After 20-30 min of high BP, 20% of NCC redistributed from low-density, APM-enriched fractions to higher density, endosome-enriched fractions, and, by quantitative immuno-EM, pool size of APM NCC decreased 14% and SCV pool size increased. Because of the time lag of the response, we tested the hypothesis that internalization of NCC was secondary to the decrease in ANG II that accompanies high BP. Clamping ANG II at a nonpressor level by coinfusion of captopril (12 microg/min) and ANG II (20 ng.kg(-1).min(-1)) during 30-min high BP reduced diuresis to eightfold and prevented redistribution of NCC from APM- to SCV-enriched fractions. We conclude that DCT NCC may participate in pressure natriuresis-diuresis by retraction out of apical plasma membranes and that the retraction is, at least in part, driven by the fall in ANG II that accompanies acute hypertension.


Asunto(s)
Presión Sanguínea , Membrana Celular/metabolismo , Vesículas Citoplasmáticas/metabolismo , Diuresis , Hipertensión/metabolismo , Túbulos Renales Distales/metabolismo , Receptores de Droga/metabolismo , Simportadores del Cloruro de Sodio/metabolismo , Simportadores/metabolismo , Enfermedad Aguda , Angiotensina II/administración & dosificación , Inhibidores de la Enzima Convertidora de Angiotensina/administración & dosificación , Animales , Presión Sanguínea/efectos de los fármacos , Captopril/administración & dosificación , Fraccionamiento Celular , Membrana Celular/efectos de los fármacos , Membrana Celular/ultraestructura , Vesículas Citoplasmáticas/efectos de los fármacos , Vesículas Citoplasmáticas/ultraestructura , Modelos Animales de Enfermedad , Diuresis/efectos de los fármacos , Hipertensión/fisiopatología , Infusiones Intravenosas , Túbulos Renales Distales/efectos de los fármacos , Túbulos Renales Distales/ultraestructura , Masculino , Microscopía Inmunoelectrónica , Transporte de Proteínas , Ratas , Ratas Sprague-Dawley , Miembro 3 de la Familia de Transportadores de Soluto 12 , Factores de Tiempo
7.
Am J Physiol Cell Physiol ; 296(4): C900-10, 2009 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-19158399

RESUMEN

Hypertension provokes differential trafficking of the renal proximal tubule Na(+)/H(+) exchanger 3 (NHE3) to the base of the apical microvilli and Na(+)-P(i) cotransporter 2 (NaPi2) to endosomes. The resultant diuresis and natriuresis are key to blood pressure control. We tested the hypothesis that this differential trafficking of NHE3 vs. NaPi2 was associated with partitioning to distinct membrane domains. In anesthetized rats, arterial pressure was increased (104 +/- 2 to 142 +/- 4 mmHg, 15 min) by arterial constriction and urine output increased 23-fold. Renal membranes were fractionated by cold 1% Triton X-100 extraction then centrifugation through OptiPrep flotation gradients. In controls, 84 +/- 9% of NHE3 localized to flotillin-enriched lipid raft domains and 69 +/- 5% of NaPi2 localized to transferrin receptor-enriched nonrafts. MyosinVI and dipeptidyl peptidase IV, associated with NHE3 regulation, coenriched in lipid rafts with NHE3, while NHE regulatory factor-1 coenriched in nonrafts with NaPi2. Partitioning was not altered by hypertension. Detergent insoluble membranes were pelleted after detergent extraction. NHE3 detergent insolubility decreased as it redistributed from body (80 +/- 10% detergent insoluble) to base (75 +/- 3%) of the apical microvilli, while NaPi2 partitioned into more insoluble domains as it moved from the microvilli (45 +/- 7% detergent insoluble) to endosomes (82 +/- 1%). In conclusion, NHE3 and NaPi2, while both localized to apical microvilli, are segregated into domains: NHE3 to lipid rafts and NaPi2 to nonrafts. These domain properties may play a role in the distinct trafficking patterns observed during elevated pressures: NHE3 remains in rafts and settles to the base of the microvilli while NaPi2 is freely endocytosed.


Asunto(s)
Membrana Celular/metabolismo , Hipertensión/metabolismo , Túbulos Renales Proximales/metabolismo , Intercambiadores de Sodio-Hidrógeno/metabolismo , Proteínas Cotransportadoras de Sodio-Fosfato de Tipo II/metabolismo , Animales , Presión Sanguínea , Fraccionamiento Celular , Membrana Celular/enzimología , Dipeptidil Peptidasa 4/metabolismo , Modelos Animales de Enfermedad , Diuresis , Endocitosis , Endosomas/metabolismo , Hipertensión/fisiopatología , Túbulos Renales Proximales/enzimología , Masculino , Microdominios de Membrana/metabolismo , Microvellosidades/metabolismo , Cadenas Pesadas de Miosina/metabolismo , Natriuresis , Transporte de Proteínas , Ratas , Ratas Sprague-Dawley , Intercambiador 3 de Sodio-Hidrógeno
8.
Am J Physiol Cell Physiol ; 294(1): C126-35, 2008 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-18003746

RESUMEN

AMP-activated protein kinase (AMPK), activated by an increase in intracellular AMP-to-ATP ratio, stimulates pathways that can restore ATP levels. We tested the hypothesis that AMPK activation influences extracellular fluid (ECF) K(+) homeostasis. In conscious rats, AMPK was activated with 5-aminoimidazole-4-carboxamide-1-beta-d-ribofuranoside (AICAR) infusion: 38.4 mg x kg bolus then 4 mg x kg(-1) x min(-1) infusion. Plasma [K(+)] and [glucose] both dropped at 1 h of AICAR infusion and [K(+)] dropped to 3.3 +/- 0.04 mM by 3 h, linearly related to the increase in muscle AMPK phosphorylation. AICAR treatment did not increase urinary K(+) excretion. AICAR lowered [K(+)] whether plasma [K(+)] was chronically elevated or lowered. The K(+) infusion rate needed to maintain baseline plasma [K(+)] reached 15.7 +/- 1.3 micromol K(+) x kg(-1) x min(-1) between 120 and 180 min AICAR infusion. In mice expressing a dominant inhibitory form of AMPK in the muscle (Tg-KD1), baseline [K(+)] was not different from controls (4.2 +/- 0.1 mM), but the fall in plasma [K(+)] in response to AICAR (0.25 g/kg) was blunted: [K(+)] fell to 3.6 +/- 0.1 in controls and to 3.9 +/- 0.1 mM in Tg-KD1, suggesting that ECF K(+) redistributes, at least in part, to muscle ICF. In summary, these findings illustrate that activation of AMPK activity with AICAR provokes a significant fall in plasma [K(+)] and suggest a novel mechanism for redistributing K(+) from ECF to ICF.


Asunto(s)
Aminoimidazol Carboxamida/análogos & derivados , Hipoglucemiantes/farmacología , Complejos Multienzimáticos/metabolismo , Músculo Esquelético/efectos de los fármacos , Potasio/sangre , Proteínas Serina-Treonina Quinasas/metabolismo , Ribonucleótidos/farmacología , Proteínas Quinasas Activadas por AMP , Aminoimidazol Carboxamida/administración & dosificación , Aminoimidazol Carboxamida/farmacología , Animales , Glucemia/efectos de los fármacos , Regulación hacia Abajo , Activación Enzimática , Técnica de Clampeo de la Glucosa , Homeostasis , Hipoglucemiantes/administración & dosificación , Infusiones Intravenosas , Líquido Intracelular/efectos de los fármacos , Líquido Intracelular/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Complejos Multienzimáticos/genética , Músculo Esquelético/enzimología , Fosforilación , Potasio/orina , Proteínas Serina-Treonina Quinasas/genética , Ratas , Ratas Sprague-Dawley , Ribonucleótidos/administración & dosificación , ATPasa Intercambiadora de Sodio-Potasio/metabolismo , Factores de Tiempo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...