Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
AACE Clin Case Rep ; 8(1): 15-18, 2022.
Article in English | MEDLINE | ID: mdl-35097195

ABSTRACT

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.
Article in English | MEDLINE | ID: mdl-24598799

ABSTRACT

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.


Subject(s)
Hyperkalemia/blood , Kidney/metabolism , Natriuresis , Potassium/blood , Sodium/urine , Animals , Disease Models, Animal , Epithelial Sodium Channels/metabolism , Hyperkalemia/chemically induced , Hyperkalemia/physiopathology , Hyperkalemia/urine , Infusions, Intravenous , Kidney/physiopathology , Male , Phosphorylation , Potassium/administration & dosage , Potassium/urine , Potassium Channels/metabolism , Potassium, Dietary/blood , Potassium, Dietary/urine , Protein Serine-Threonine Kinases/metabolism , Rats , Rats, Sprague-Dawley , Sodium/blood , Sodium-Hydrogen Exchanger 3 , Sodium-Hydrogen Exchangers/metabolism , Solute Carrier Family 12, Member 3/metabolism , Time Factors
3.
Am J Physiol Renal Physiol ; 305(4): F510-9, 2013 Aug 15.
Article in English | MEDLINE | ID: mdl-23720346

ABSTRACT

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.


Subject(s)
Angiotensin II/pharmacology , Blood Pressure/drug effects , Epithelial Sodium Channels/metabolism , Hypertension/metabolism , Kidney/metabolism , Membrane Transport Proteins/metabolism , Nephrons/metabolism , Sodium/metabolism , Animals , Epithelial Sodium Channels/drug effects , Immunoblotting , Immunohistochemistry , Male , Membrane Transport Proteins/drug effects , Nephrons/drug effects , Rats , Rats, Sprague-Dawley
4.
Am J Physiol Cell Physiol ; 304(2): C147-63, 2013 Jan 15.
Article in English | MEDLINE | ID: mdl-23114965

ABSTRACT

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.


Subject(s)
Angiotensin II/pharmacology , Angiotensin-Converting Enzyme Inhibitors/pharmacology , Kidney Tubules, Distal/metabolism , Sodium Chloride Symporters/metabolism , Animals , Calmodulin-Binding Proteins/analysis , Captopril/pharmacology , Kidney Tubules, Distal/cytology , Kidney Tubules, Distal/drug effects , Male , Phosphorylation , Rats , Rats, Sprague-Dawley , Sodium Chloride Symporter Inhibitors/pharmacology
5.
Am J Physiol Renal Physiol ; 303(1): F92-104, 2012 Jul 01.
Article in English | MEDLINE | ID: mdl-22496411

ABSTRACT

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.


Subject(s)
Epithelial Sodium Channels/metabolism , Nephrons/metabolism , Potassium Deficiency/metabolism , Potassium, Dietary/pharmacology , Sodium Chloride, Dietary/pharmacology , Sodium-Hydrogen Exchangers/metabolism , Sodium-Potassium-Chloride Symporters/metabolism , Animals , Male , Nephrons/drug effects , Phosphorylation/drug effects , Rats , Rats, Sprague-Dawley
6.
Am J Physiol Renal Physiol ; 296(4): F810-8, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19144688

ABSTRACT

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.


Subject(s)
Blood Pressure , Cell Membrane/metabolism , Cytoplasmic Vesicles/metabolism , Diuresis , Hypertension/metabolism , Kidney Tubules, Distal/metabolism , Receptors, Drug/metabolism , Sodium Chloride Symporters/metabolism , Symporters/metabolism , Acute Disease , Angiotensin II/administration & dosage , Angiotensin-Converting Enzyme Inhibitors/administration & dosage , Animals , Blood Pressure/drug effects , Captopril/administration & dosage , Cell Fractionation , Cell Membrane/drug effects , Cell Membrane/ultrastructure , Cytoplasmic Vesicles/drug effects , Cytoplasmic Vesicles/ultrastructure , Disease Models, Animal , Diuresis/drug effects , Hypertension/physiopathology , Infusions, Intravenous , Kidney Tubules, Distal/drug effects , Kidney Tubules, Distal/ultrastructure , Male , Microscopy, Immunoelectron , Protein Transport , Rats , Rats, Sprague-Dawley , Solute Carrier Family 12, Member 3 , Time Factors
7.
Am J Physiol Cell Physiol ; 296(4): C900-10, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19158399

ABSTRACT

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.


Subject(s)
Cell Membrane/metabolism , Hypertension/metabolism , Kidney Tubules, Proximal/metabolism , Sodium-Hydrogen Exchangers/metabolism , Sodium-Phosphate Cotransporter Proteins, Type II/metabolism , Animals , Blood Pressure , Cell Fractionation , Cell Membrane/enzymology , Dipeptidyl Peptidase 4/metabolism , Disease Models, Animal , Diuresis , Endocytosis , Endosomes/metabolism , Hypertension/physiopathology , Kidney Tubules, Proximal/enzymology , Male , Membrane Microdomains/metabolism , Microvilli/metabolism , Myosin Heavy Chains/metabolism , Natriuresis , Protein Transport , Rats , Rats, Sprague-Dawley , Sodium-Hydrogen Exchanger 3
8.
Am J Physiol Cell Physiol ; 294(1): C126-35, 2008 Jan.
Article in English | MEDLINE | ID: mdl-18003746

ABSTRACT

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.


Subject(s)
Aminoimidazole Carboxamide/analogs & derivatives , Hypoglycemic Agents/pharmacology , Multienzyme Complexes/metabolism , Muscle, Skeletal/drug effects , Potassium/blood , Protein Serine-Threonine Kinases/metabolism , Ribonucleotides/pharmacology , AMP-Activated Protein Kinases , Aminoimidazole Carboxamide/administration & dosage , Aminoimidazole Carboxamide/pharmacology , Animals , Blood Glucose/drug effects , Down-Regulation , Enzyme Activation , Glucose Clamp Technique , Homeostasis , Hypoglycemic Agents/administration & dosage , Infusions, Intravenous , Intracellular Fluid/drug effects , Intracellular Fluid/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Multienzyme Complexes/genetics , Muscle, Skeletal/enzymology , Phosphorylation , Potassium/urine , Protein Serine-Threonine Kinases/genetics , Rats , Rats, Sprague-Dawley , Ribonucleotides/administration & dosage , Sodium-Potassium-Exchanging ATPase/metabolism , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...