Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 40
Filter
1.
Orthop Traumatol Surg Res ; 104(2): 267-271, 2018 04.
Article in English | MEDLINE | ID: mdl-29410212

ABSTRACT

BACKGROUND: Personal watercrafts (PWC) account for a disproportionate amount of water based injuries. Current literature suggests those with less PWC experience are more at risk for injury. Previous studies have not specifically evaluated the orthopedic implications of PWC usage or how various mechanisms of injury (MOI) contribute to different injury patterns. HYPOTHESIS: PWC injuries will frequently require orthopedic intervention. The presence of an orthopedic injury will result in increased injury severity score (ISS), hospital and intensive care unit (ICU) length of stay (LOS). Patients visiting our region will have less PWC experience and so are more prone to serious injuries. MATERIALS AND METHODS: Retrospective cohort study at a single Level 1 trauma center of admitted patients sustaining PWC injuries from 02/2004-03/2017. The following were studied: demographics, mechanism, season, ISS, hospital and ICU LOS, follow-up, fracture characteristics and management. RESULTS: Hundred and twenty-seven patients were admitted due to PWC injury, 66 (52.0%) sustained an orthopedic injury, totaling 103 fractures (48 [46.6%] lower extremity, 26 [25.2%] upper extremity, 14 [13.6%] vertebral, 11 [10.7%] pelvic ring and 4 [3.9%] acetabulum). The mean age of orthopedic patients was 29 years (range 8-62). Handle bar injuries were significantly associated with open fractures, (13 of 25 open fractures, 3 of which became infected). Injuries occurring during the winter were associated with a higher ISS, yet more injuries occurred in the summer. A patient being a "visitor" to the region did not influence ISS. The mean LOS was 12.6 days for orthopedic patients. Eighteen orthopedic patients (27.3%) required ICU admission and 36 (54.5%) patients required orthopedic surgery (mean 2.11 operations). DISCUSSION: A majority of PWC injuries resulted in extremity fractures with a moderate percentage requiring orthopedic surgery. Correlations between PWC experience and injury incidence can provide information for increased safety. LEVEL OF EVIDENCE: IV; retrospective.


Subject(s)
Fractures, Bone/epidemiology , Fractures, Bone/etiology , Sports Equipment/adverse effects , Water Sports/injuries , Adolescent , Adult , Child , Female , Florida/epidemiology , Fractures, Bone/surgery , Fractures, Open/etiology , Humans , Incidence , Injury Severity Score , Intensive Care Units , Length of Stay , Lower Extremity/injuries , Male , Middle Aged , Orthopedic Procedures/statistics & numerical data , Pelvic Bones/injuries , Retrospective Studies , Seasons , Ships , Spinal Fractures/epidemiology , Spinal Fractures/etiology , Upper Extremity/injuries , Young Adult
2.
Diabetes Obes Metab ; 17(2): 188-97, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25421015

ABSTRACT

AIMS: To compare the pharmacodynamic effects of the highest approved doses of the sodium glucose co-transporter 2 (SGLT2) inhibitors canagliflozin and dapagliflozin on urinary glucose excretion (UGE), renal threshold for glucose excretion (RTG ) and postprandial plasma glucose (PPG) excursion in healthy participants in a randomized, double-blind, two-period crossover study. METHODS: In each treatment period, participants (n = 54) received canagliflozin 300 mg or dapagliflozin 10 mg for 4 days (20 min before breakfast). A mixed-meal tolerance test (600 kcal; 75 g glucose) was performed at baseline and on day 4 of each treatment period to assess changes in incremental PPG (PPGΔAUC0-2 h ). We measured 24-h UGE and plasma glucose on day 4 to determine 24-h mean RTG . RESULTS: Canagliflozin 300 mg and dapagliflozin 10 mg had similar effects on UGE and RTG for 4 h after dosing, but canagliflozin was associated with higher UGE and greater RTG reductions for the remainder of the day. Mean 24-h UGE was ∼25% higher with canagliflozin than with dapagliflozin (51.4 vs. 40.8 g), and 24-h mean RTG was ∼0.4 mmol/l (7 mg/dl) lower with canagliflozin than with dapagliflozin (3.79 vs. 4.17 mmol/l; p < 0.0001). Dapagliflozin had no effect on PPG excursion; canagliflozin delayed and reduced PPG excursion (between-treatment difference in PPGΔAUC0-2 h from baseline expressed as a percentage of baseline mean, -10.2%; p = 0.0122). Canagliflozin and dapagliflozin were generally well tolerated. CONCLUSIONS: In healthy participants, canagliflozin 300 mg provided greater 24-h UGE, a lower RTG and smaller PPG excursions than dapagliflozin 10 mg.


Subject(s)
Benzhydryl Compounds/pharmacokinetics , Blood Glucose/drug effects , Glucose/metabolism , Glucosides/pharmacokinetics , Hypoglycemic Agents/pharmacokinetics , Kidney/metabolism , Thiophenes/pharmacokinetics , Adult , Aged , Benzhydryl Compounds/therapeutic use , Body Weight , Canagliflozin , Cross-Over Studies , Double-Blind Method , Drug Administration Schedule , Female , Glomerular Filtration Rate , Glucosides/therapeutic use , Humans , Hypoglycemic Agents/therapeutic use , Male , Middle Aged , Postprandial Period , Thiophenes/therapeutic use
3.
Diabetes Obes Metab ; 16(11): 1087-95, 2014 Nov.
Article in English | MEDLINE | ID: mdl-24939043

ABSTRACT

AIM: To evaluate the effects of canagliflozin on plasma volume, urinary glucose excretion (UGE), fasting plasma glucose (FPG), glycated haemoglobin (HbA1c) and additional measures of fluid/electrolyte balance in patients with type 2 diabetes on background therapy with metformin and angiotensin-converting enzyme inhibitors or angiotensin receptor blockers. METHODS: Patients (N = 36) were randomized (1:1) to receive canagliflozin 300 mg or placebo for 12 weeks. Pharmacodynamic parameters were assessed at baseline and at weeks 1 and 12. RESULTS: Increased 24-h UGE was seen in the canagliflozin group compared with a reduction in the placebo group at both week 1 (91.8 vs. -2.4 g) and week 12 (82.6 vs. -0.4 g). Canagliflozin also reduced both FPG and HbA1c. Reductions in body weight and blood pressure were observed at weeks 1 and 12. Canagliflozin decreased plasma volume compared with an increase with placebo at week 1 (-5.4 vs. 4.3%; p = 0.02), but this was largely attenuated at week 12 (4.6 vs. 5.8%; p = 0.76). A modest numerical increase in urine volume was observed with canagliflozin at week 1 that was attenuated at week 12; other measures of volume status (i.e. blood urea nitrogen, serum creatinine and haematocrit) remained modestly increased with canagliflozin at week 12. CONCLUSION: Canagliflozin provided sustained effects on UGE and FPG over 12 weeks and a transient reduction in plasma volume that was largely attenuated by week 12.


Subject(s)
Blood Pressure/drug effects , Diabetes Mellitus, Type 2/drug therapy , Diabetic Angiopathies/prevention & control , Glucosides/therapeutic use , Hypertension/prevention & control , Plasma Volume/drug effects , Sodium-Glucose Transporter 2 Inhibitors , Thiophenes/therapeutic use , Adult , Aged , Antihypertensive Agents , Blood Glucose/drug effects , Blood Glucose/metabolism , Body Weight/drug effects , Canagliflozin , Diabetes Mellitus, Type 2/blood , Diabetes Mellitus, Type 2/complications , Dose-Response Relationship, Drug , Drug Therapy, Combination , Fasting , Female , Glycated Hemoglobin/drug effects , Glycated Hemoglobin/metabolism , Humans , Hypoglycemic Agents/therapeutic use , Kidney/drug effects , Kidney/metabolism , Male , Metformin/therapeutic use , Middle Aged , Treatment Outcome , Water-Electrolyte Balance/drug effects , Weight Loss/drug effects
4.
Diabetes Obes Metab ; 16(11): 1055-64, 2014 Nov.
Article in English | MEDLINE | ID: mdl-24798870

ABSTRACT

AIMS: Macrophage recruitment through C-C motif chemokine receptor-2 (CCR2) into adipose tissue is believed to play a role in the development of insulin resistance and type 2 diabetes mellitus (T2DM). The objective of this Phase 2 proof-of-concept study was to evaluate the safety, tolerability, pharmacokinetics and pharmacodynamics of JNJ-41443532, an orally bioavailable CCR2 antagonist, in patients with T2DM. METHODS: This was a 4-week, double-blind, placebo-controlled, randomized, multicenter study. A total of 89 patients were randomized to receive either 250- or 1000-mg of JNJ-41443532 twice daily, 30-mg of pioglitazone once daily (reference arm), or placebo. The primary endpoint was change from baseline in 23-h weighted mean glucose (WMG); secondary endpoints included change from baseline in fasting plasma glucose (FPG), insulin resistance (Homeostatic Model Assessment [HOMA-IR]), insulin secretion (HOMA-%B) and body weight. RESULTS: Absorption of JNJ-41443532 into the systemic circulation occurred at a median tmax of 2 h, and the mean t½ was approximately 8 h for both doses; plasma systemic exposures increased slightly more than dose-proportionally. After 4 weeks, reductions in 23-h WMG and FPG were observed in all treatment groups compared with placebo and were significantly lower for 250-mg JNJ-41443532 and pioglitazone. HOMA-IR was lower for all treatment groups, but significantly lower only for pioglitazone. Conversely, HOMA-%B was increased for all groups, but significantly increased only for 250-mg JNJ-41443532. All groups, including placebo, had decreased body weight over time. There were no clinically significant findings during routine safety assessments and the incidence of treatment-emergent adverse events was similar across all groups. CONCLUSIONS: Administration of JNJ-41443532 resulted in modest improvement in glycaemic parameters compared with placebo, and was generally well tolerated in patients with T2DM.


Subject(s)
Blood Glucose/drug effects , Diabetes Mellitus, Type 2/drug therapy , Glycated Hemoglobin/drug effects , Hypoglycemic Agents/pharmacokinetics , Receptors, CCR2/antagonists & inhibitors , Thiazolidinediones/pharmacokinetics , Adult , Azetidines/administration & dosage , Azetidines/pharmacokinetics , Azetidines/pharmacology , Benzamides/administration & dosage , Benzamides/pharmacokinetics , Benzamides/pharmacology , Body Mass Index , Diabetes Mellitus, Type 2/epidemiology , Directive Counseling , Double-Blind Method , Drug Administration Schedule , Fasting , Female , Humans , Hypoglycemic Agents/administration & dosage , Hypoglycemic Agents/pharmacology , Male , Middle Aged , Pioglitazone , Thiazolidinediones/administration & dosage , Thiazolidinediones/pharmacology , Treatment Outcome , United States/epidemiology
5.
Diabetes Obes Metab ; 14(8): 709-16, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22340428

ABSTRACT

AIM: G-protein coupled receptor agonists are currently under investigation for their potential utility in patients with type 2 diabetes mellitus (T2DM). The objective was to determine the pharmacokinetics, pharmacodynamics, safety and tolerability of GPR119 agonist, JNJ-38431055 in T2DM subjects. METHODS: This was a randomized, double-blind, placebo- and positive-controled, single-dose cross-over study and a randomized, double-blind, placebo-controled multiple-dose parallel design study. The study was conducted at 4 US research centres. Two different experiments involving 25 and 32 different subjects were performed in male and female subjects, aged 25-60 years, mean body mass index between 22 and 39.9 kg/m2 who had T2DM diagnosed 6 months to 10 years before screening. JNJ-38431055 (100 and 500 mg) or sitagliptin (100 mg) as a single-dose or JNJ-38431055 (500 mg) once daily for 14 consecutive days were tested. Effects on stimulated plasma glucose, insulin, C-peptide and incretin concentrations were pre-specified outcomes. RESULTS: JNJ-38431055 was well tolerated and not associated with hypoglycaemia. Plasma systemic exposure of JNJ-38431055 increased as the dose increased, was approximately two-fold greater after multiple-dose administration, and attained steady-state after approximately 8 days. Compared with placebo, single-dose administration of oral JNJ-38431055 decreased glucose excursion during an oral glucose tolerance test, but multiple-dose administration did not alter 24-h weighted mean glucose. Multiple dosing of JNJ-38431055 increased post-meal total glucagon-like peptide 1 and gastric insulinotropic peptide concentrations compared to baseline. CONCLUSIONS: These studies provide evidence of limited glucose lowering and incretin activity for JNJ-38431055 in subjects with T2DM.


Subject(s)
Blood Glucose/metabolism , Diabetes Mellitus, Type 2/drug therapy , Dipeptidyl-Peptidase IV Inhibitors/pharmacology , Glucagon-Like Peptide 1/blood , Incretins/blood , Pyrazines/pharmacology , Receptors, G-Protein-Coupled/agonists , Triazoles/pharmacology , Administration, Oral , Adult , Blood Glucose/drug effects , Cross-Over Studies , Diabetes Mellitus, Type 2/blood , Dipeptidyl-Peptidase IV Inhibitors/administration & dosage , Double-Blind Method , Female , Glucagon-Like Peptide 1/drug effects , Glucose Tolerance Test , Humans , Male , Middle Aged , Pyrazines/administration & dosage , Sitagliptin Phosphate , Treatment Outcome , Triazoles/administration & dosage
6.
Clin Pharmacol Ther ; 90(5): 685-92, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21975348

ABSTRACT

The incidence of type 2 diabetes mellitus is increasing worldwide. Several G-protein-coupled receptor agonists are being studied for their efficacy as antidiabetes agents. JNJ-38431055 is a novel, potent, and orally available selective agonist of the glucose-dependent insulinotropic (GPR119) receptor. Double-blind, randomized, placebo-controlled studies were conducted to evaluate the safety, tolerability, pharmacokinetics, and pharmacodynamics of single oral doses of JNJ-38431055 (2.5-800 mg) in healthy male volunteers. The systemic exposure of JNJ-38431055 in plasma increased in proportion to the dose and was not influenced by coadministration of food. The terminal elimination half-life was ~13 h when administered as an oral suspension formulation. JNJ-38431055 was well tolerated and was not associated with hypoglycemia. As compared with placebo, single-dose oral JNJ-38431055 increased postmeal plasma glucagon-like peptide 1 (GLP-1), glucose-dependent insulinotropic peptide (GIP), and peptide YY (PYY) concentrations but did not significantly decrease glucose excursion or increase insulin secretion. However, in a graded glucose infusion study, JNJ-38431055 was shown to induce a higher insulin secretion rate (ISR) relative to placebo at elevated plasma glucose levels. These studies provide evidence for the potential efficacy of JNJ-38431055 as an antidiabetes agent in humans.


Subject(s)
Blood Glucose/drug effects , Hypoglycemic Agents/pharmacology , Receptors, G-Protein-Coupled/agonists , Administration, Oral , Adult , Double-Blind Method , Gastric Inhibitory Polypeptide/blood , Gastric Inhibitory Polypeptide/drug effects , Glucagon-Like Peptide 1/blood , Glucagon-Like Peptide 1/drug effects , Half-Life , Humans , Hypoglycemic Agents/adverse effects , Hypoglycemic Agents/pharmacokinetics , Insulin/metabolism , Insulin Secretion , Male , Middle Aged , Peptide YY/blood , Peptide YY/drug effects
7.
Diabetes Obes Metab ; 13(7): 669-72, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21457428

ABSTRACT

Canagliflozin, a potent, selective sodium glucose co-transporter 2 inhibitor in development for treatment of type 2 diabetes, lowers plasma glucose (PG) by lowering the renal threshold for glucose (RT(G) ) and increasing urinary glucose excretion (UGE). An ascending single oral-dose phase 1 study investigated safety, tolerability and pharmacodynamics of canagliflozin in healthy men (N = 63) randomized to receive canagliflozin (n = 48) or placebo (n = 15). Canagliflozin (10, 30, 100, 200, 400, 600 or 800 mg q.d. or 400 mg b.i.d.) was administered to eight cohorts (six subjects/cohort: canagliflozin; two subjects/cohort: placebo). Dose dependently, canagliflozin decreased calculated 24-h mean RT(G) with maximal reduction to approximately 60 mg/dl, and increased mean 24-h UGE. At doses >200 mg administered before breakfast, canagliflozin reduced postprandial PG and serum insulin excursions at that meal. Canagliflozin was generally well tolerated; most adverse events were mild and no hypoglycaemia was reported. These results support further study of canagliflozin.


Subject(s)
Diabetes Mellitus, Type 2/drug therapy , Glucosides/pharmacology , Glycosuria/metabolism , Hypoglycemic Agents/therapeutic use , Insulin/metabolism , Sodium-Glucose Transporter 2 Inhibitors , Thiophenes/pharmacology , Adolescent , Adult , Canagliflozin , Diabetes Mellitus, Type 2/metabolism , Dose-Response Relationship, Drug , Glucosides/therapeutic use , Glycosuria/chemically induced , Humans , Male , Middle Aged , Thiophenes/therapeutic use , Young Adult
8.
Eur J Clin Pharmacol ; 61(5-6): 341-6, 2005 Jul.
Article in English | MEDLINE | ID: mdl-15983826

ABSTRACT

OBJECTIVE: To examine the effect of aprepitant on the pharmacokinetics and pharmacodynamics of warfarin. Aprepitant is a neurokinin-1 (NK1)-receptor antagonist developed as an antiemetic for chemotherapy-induced nausea and vomiting. METHODS: This was a double-blind, placebo-controlled, randomized, two-period, parallel-group study. During period 1, warfarin was individually titrated to a stable prothrombin time (expressed as international normalized ratio, INR) from 1.3 to 1.8. Subsequently, the daily warfarin dose remained fixed for 10-12 days. During period 2, the warfarin dose was continued for 8 days, and on days 1-3 administered concomitantly with aprepitant (125 mg on day 1, and 80 mg on days 2 and 3) or placebo. At baseline (day -1 of period 2) and on day 3, warfarin pharmacokinetics was investigated. INR was monitored daily. During period 2, warfarin trough concentrations were determined daily. RESULTS: The study was completed by 22 healthy volunteers (20 men, 2 women). On day 3, steady-state pharmacokinetics of warfarin enantiomers after aprepitant did not change, as assessed by warfarin AUC(0-24 h) and C(max). However, compared with placebo, trough S(-) warfarin concentrations decreased on days 5-8 (maximum decrease 34% on day 8, P<0.01). The INR decreased after aprepitant with a mean maximum decrease on day 8 of 11% versus placebo (P=0.011). CONCLUSION: These data are consistent with a significant induction of CYP2C9 metabolism of S(-) warfarin by aprepitant. Subsequently, in patients on chronic warfarin therapy, the clotting status should be monitored closely during the 2-week period, particularly at 7-10 days, following initiation of the 3-day regimen of aprepitant with each chemotherapy cycle.


Subject(s)
Anticoagulants/pharmacokinetics , Antiemetics/pharmacology , Morpholines/pharmacology , Warfarin/pharmacokinetics , Anticoagulants/blood , Anticoagulants/pharmacology , Antiemetics/administration & dosage , Aprepitant , Area Under Curve , Aryl Hydrocarbon Hydroxylases/biosynthesis , Aryl Hydrocarbon Hydroxylases/metabolism , Cytochrome P-450 CYP2C9 , Double-Blind Method , Drug Interactions , Enzyme Induction , Female , Humans , International Normalized Ratio , Male , Metabolic Clearance Rate , Morpholines/administration & dosage , Prothrombin Time , Time Factors , Warfarin/blood , Warfarin/pharmacology
9.
J Cardiovasc Pharmacol ; 37(5): 502-11, 2001 May.
Article in English | MEDLINE | ID: mdl-11336101

ABSTRACT

Fluvastatin sodium (Lescol, Novartis Pharmaceutical Corp., East Hanover, NJ, U.S.A.), a potent 3-hydroxy-3-methylglutaryl coenzyme A (HMG Co-A) reductase inhibitor that limits cholesterol biosynthesis, is available as a 40-mg immediate-release formulation capsule. An extended-release formulation for once-daily administration has been developed for patients with primary hypercholesterolemia who may benefit from doses higher than 40 mg/day. This phase I study evaluated the safety, tolerability, and pharmacokinetics of a new fluvastatin extended-release formulation at doses ranging from 80-640 mg/day in 40 hypercholesterolemic patients. After a 2-week dietary stabilization phase, patients (Fredrickson type IIa/IIb), 18-55 years of age, were randomly assigned to four groups to receive oral fluvastatin extended-release (80, 160, 320, or 640 mg) or matching placebo once daily for 13 days. Fluvastatin extended-release was generally safe and well tolerated at doses of 80-320 mg/day. Within this dose range, linear pharmacokinetics was observed after single and multiple dosing. At 640 mg, fluvastatin extended-release was not well tolerated. Six of the seven actively treated patients at this dose experienced adverse events, including diarrhea, headache, and clinically relevant elevations in serum transaminase concentrations. In addition, nonlinear pharmacokinetics, possibly due to saturation of first-pass metabolism, was observed at this dose, causing higher than expected serum drug concentrations. Once-daily administration of fluvastatin extended-release at doses of 80-320 mg/day was generally safe and well tolerated in patients with primary hypercholesterolemia over a 13-day dosing period.


Subject(s)
Anticholesteremic Agents/administration & dosage , Anticholesteremic Agents/adverse effects , Fatty Acids, Monounsaturated/administration & dosage , Fatty Acids, Monounsaturated/adverse effects , Hypercholesterolemia/drug therapy , Indoles/administration & dosage , Indoles/adverse effects , Adolescent , Adult , Anticholesteremic Agents/pharmacokinetics , Area Under Curve , Delayed-Action Preparations , Dose-Response Relationship, Drug , Double-Blind Method , Drug Administration Schedule , Fatty Acids, Monounsaturated/pharmacokinetics , Female , Fluvastatin , Humans , Hypercholesterolemia/blood , Indoles/pharmacokinetics , Lipoproteins/blood , Liver Function Tests , Male , Middle Aged
10.
Diabetes ; 47(8): 1243-52, 1998 Aug.
Article in English | MEDLINE | ID: mdl-9703324

ABSTRACT

The insulin receptor (IR) is expressed by insulin-secreting beta-cells, but its cellular function is unknown. We transfected betaTC6-F7 beta-cells with cDNAs encoding either wild-type or mutant kinase-inactive (A/K1018) IRs, and by fluorescence-activated cell sorting generated polyclonal beta-cell lines that overexpressed each receptor type at levels two- to fourfold higher than the parental cells. Beta-cells overexpressing wild-type IRs had a proportional increase in insulin-stimulated tyrosine kinase activity; no change occurred in beta-cells expressing the mutant IR. We observed a threefold increase in cellular insulin content in beta-cells that overexpressed the wild-type IR, as determined by radioimmunoassay. This increase occurred despite a fivefold elevated rate of both basal and 10 mmol/l glucose-induced insulin secretion. Fractional insulin secretion (percentage of total cell insulin releasable at 10 mmol/l glucose) was unchanged in beta-cells overexpressing the wild-type IR compared with the parental beta-cell line. Insulin content and insulin secretion were unaffected by overexpression of kinase-inactive IRs. Steady-state insulin mRNA levels were elevated twofold in the beta-cells overexpressing the wild-type IR and unchanged in the beta-cells expressing the kinase-inactive receptor, as determined by Northern blot analysis. The rate of insulin mRNA degradation measured in the presence of 5 microg/ml actinomycin D was not significantly affected in either cell line. In the absence of glucose, the basal level of (pro)insulin biosynthesis in the beta-cells overexpressing the wild-type IR increased significantly (61%) compared with the beta-cells transfected with the kinase-inactive IR. These data indicate that IR signaling can regulate insulin gene transcription and can modulate the steady-state insulin content of beta-cells.


Subject(s)
Autocrine Communication/physiology , Gene Expression/physiology , Insulin/physiology , Islets of Langerhans/physiology , Receptor, Insulin/physiology , Signal Transduction/physiology , Animals , Cell Line , Insulin/genetics , Insulin/metabolism , Islets of Langerhans/metabolism , Mice , RNA, Messenger/metabolism , Receptor, Insulin/metabolism , Transfection/physiology
11.
Biochem J ; 318 ( Pt 2): 623-9, 1996 Sep 01.
Article in English | MEDLINE | ID: mdl-8809056

ABSTRACT

The physiological role of the beta-cell insulin receptor is unknown. To evaluate a candidate function, the insulin regulation of fluid-phase pinocytosis was investigated in a clonal insulinoma cell line (beta TC6-F7) and, for comparison, also in Chinese hamster ovary cells transfected with the human insulin receptor (CHO-T cells). In CHO-T cells, the net rate of fluid-phase pinocytosis was rapidly increased 3-4-fold over the basal rate by 100 nM insulin, with half-maximal stimulation at 2 nM insulin, as assayed by cellular uptake of horseradish peroxidase from the medium. Wortmannin, an inhibitor of phosphatidylinositol (PI)-3-kinase, blocked insulin-stimulated pinocytosis with an IC50 of 7.5 nM without affecting the basal rate of pinocytosis. In insulin-secreting beta TC6-F7 cells, the secretagogues glucose and carbachol (at maximally effective concentrations of 15 mM and 0.5 mM respectively) augmented fluid-phase pinocytosis 1.65-fold over the basal rate. Wortmannin also inhibited secretagogue-stimulated pinocytosis in these beta-cells with an IC50 of 7 nM but did not affect the basal rate of pinocytosis measured in the absence of secretagogues. Wortmannin did not influence either basal or secretagogue-induced insulin secretion. Although these beta TC6-F7 cells have cell-surface insulin receptors, adding exogenous insulin or insulin-like growth factor 1 did not affect their rate of fluid-phase pinocytosis, either in the absence or presence of secretagogues. From these observations, we conclude that: (1) in both insulin-secreting beta-cells and in conventional, insulin-responsive CHO-T cells, a common, wortmannin-sensitive reaction, which probably involves PI-3-kinase, regulates fluid-phase pinocytosis; (2) the insulin-receptor signal transduction pathway is dissociated from the regulation of fluid-phase pinocytosis in the insulin-secreting beta-cell line we studied; and (3) the enhancement of fluid-phase pinocytosis associated with secretagogue-induced insulin release in beta TC6-F7 cells is not attributable to autocrine activation of beta-cell surface insulin receptors.


Subject(s)
Insulin/metabolism , Insulin/pharmacology , Islets of Langerhans/physiology , Pinocytosis/physiology , Receptor, Insulin/physiology , Androstadienes/pharmacology , Animals , CHO Cells , Carbachol/pharmacology , Clone Cells , Cricetinae , Glucose/pharmacology , Humans , Insulin Antagonists/pharmacology , Insulin Secretion , Insulinoma , Islets of Langerhans/drug effects , Islets of Langerhans/metabolism , Kinetics , Pancreatic Neoplasms , Pinocytosis/drug effects , Receptor, Insulin/biosynthesis , Recombinant Proteins/metabolism , Transfection , Wortmannin
12.
Diabetes ; 45(7): 854-62, 1996 Jul.
Article in English | MEDLINE | ID: mdl-8666133

ABSTRACT

Glucose is the primary stimulus for insulin secretion by pancreatic beta-cells, and it triggers membrane depolarization and influx of extracellular Ca2+. Cholinergic agonists amplify insulin release by several pathways, including activation of phospholipase C, which hydrolyzes membrane polyphosphoinositides. A novel phospholipid, phosphatidylinositol 3,4,5- trisphosphate [PtdIns(3,4,5)P3], a product of phosphatidylinositol 3-kinase (PI 3-kinase), has recently been found in various cell types. We demonstrate by immunoblotting that PI 3-kinase is present in both cytosolic and membrane fractions of insulin-secreting beta-TC3 cells and in rat islets. The catalytic activity of PI 3-kinase in immunoprecipitates of islets and beta-TC3 cells was measured by the production of radioactive phosphatidylinositol 3-monophosphate from phosphatidylinositol (PtdIns) in the presence of [gamma-32P]ATP. Wortmannin, a fungal metabolite, dose dependently inhibited PI 3-kinase activity of both islets and beta-TC3 cells, with an IC50 of 1 nmol/l and a maximally effective concentration of 100 nmol/l, when it was added directly to the kinase assay. However, if intact islets were incubated with wortmannin and PI 3-kinase subsequently was determined in islet immunoprecipitates, approximately 50% inhibition of PI 3-kinase activity (but no inhibition of glucose- and carbachol-stimulated insulin secretion) from intact islets was obtained at wortmannin concentrations of 100 nmol/l. Wortmannin, at higher concentrations (1 and 10 micromol/l), inhibited glucose- and carbachol-induced insulin secretion of Intact rat islets by 58 and 92%, respectively. Wortmannin had no effect on the basal insulin release from rat islets. A similar dose curve of inhibition of glucose- and carbachol-induced insulin secretion by wortmannin was obtained when beta-TC3 cells were used. Cellular metabolism was, not changed by any wortmannin concentrations tested (0.01-10 micromol/l). Both basal cytosolic [Ca2+]i and carbamyl choline-induced increases of [Ca2]i were unaffected by wortmannin in the presence of 2.5 mmol/l Ca2+, while Ca2+ mobilization from intracellular stores was partially decreased by wortmannin. Together, these data suggest that wortmannin at concentrations that inhibit PI 3-kinase does not affect insulin secretion. PI 3-kinase is unlikely to have a major role in insulin secretion induced by glucose and carbachol.


Subject(s)
Androstadienes/pharmacology , Enzyme Inhibitors/pharmacology , Insulin/metabolism , Islets of Langerhans/metabolism , Phosphotransferases (Alcohol Group Acceptor)/antagonists & inhibitors , Animals , Calcium/metabolism , Calcium Chloride/pharmacology , Carbachol/pharmacology , Cell Line , Cell Membrane/metabolism , Cells, Cultured , Cytosol/metabolism , Dose-Response Relationship, Drug , Insulin Secretion , Islets of Langerhans/drug effects , Kinetics , Male , Phosphatidylinositol 3-Kinases , Rats , Rats, Sprague-Dawley , Wortmannin
13.
Diabetes ; 45(6): 711-7, 1996 Jun.
Article in English | MEDLINE | ID: mdl-8635642

ABSTRACT

The expression of insulin receptor mRNA was examined in rat pancreatic islet cells by single-cell reverse transcriptase (RT)-polymerase chain reaction (PCR). Single cells from disaggregated islets were individually isolated in a microcapillary pipet, and the beta-cells were identified by amplification of the mRNA for insulin. We found that in single beta-cells, the mRNA for the insulin receptor was also expressed. The fraction of single islet cells expressing both insulin receptor and insulin mRNAs corresponds closely to the fraction of beta-cells in the disaggregated islet cell preparation. These results indicate that normal beta-cells have the potential to express authentic insulin receptors. Immunohistochemical analysis was insufficiently sensitive for assaying insulin receptor protein; however, insulin receptor substrate 1 (IRS-1) was readily immunolocalized in islet beta-cells. Since IRS-1 links several cell surface receptors, including those for insulin and IGF-I, to distal signal transduction pathways, our observations indicate that hormonal regulation of islet beta-cells potentially involves the same signal transduction pathway that mediates insulin and growth factor signaling in peripheral insulin target tissue cell types.


Subject(s)
Islets of Langerhans/metabolism , Phosphoproteins/biosynthesis , Receptor, Insulin/biosynthesis , Transcription, Genetic , Animals , Base Sequence , Cells, Cultured , DNA Primers , Immunohistochemistry , Insulin Receptor Substrate Proteins , Islets of Langerhans/cytology , Male , Molecular Sequence Data , Polymerase Chain Reaction , RNA, Messenger/biosynthesis , Rats , Rats, Sprague-Dawley , Signal Transduction
15.
Diabetes ; 44(7): 802-9, 1995 Jul.
Article in English | MEDLINE | ID: mdl-7540574

ABSTRACT

In the beta TC3 insulin-secreting beta-cell line, glucose rapidly induces the tyrosine phosphorylation of the 97-kDa insulin receptor beta-subunit. Phosphorylation is transient, with fourfold stimulation by 2 min and subsequent dephosphorylation to basal levels by 10-15 min. Elevating the extracellular KCl concentration equipotently initiates receptor phosphorylation. Preventing insulin secretion with 1 mumol/l epinephrine or by removing extracellular Ca2+ blocks the effect. In the absence of glucose-induced secretion, exogenous insulin also stimulated insulin receptor autophosphorylation transiently and with an ED50 of 4 x 10(-9) mol/l. In addition, functional insulin-like growth factor I (IGF-I) receptors are also expressed by these beta-cells, as indicated by IGF-I-induced receptor tyrosine phosphorylation (ED50 = 5 x 10(-9) mol/l) and also by detection of hybrid insulin/IGF-I receptor autophosphorylation at 10(-7) mol/l IGF-I. Both glucose and insulin stimulate the tyrosine phosphorylation of the insulin receptor substrate (IRS) IRS-1 and increase by two- to fivefold the rapid association of IRS-1 with the 85-kDa alpha-subunit of the phosphatidylinositol-3-kinase, as determined by co-immunoprecipitation assays. These results demonstrate that in these beta-cells, glucose-induced insulin secretion activates the beta-cell surface insulin receptor tyrosine kinase and its intracellular signal transduction pathway, suggesting a new autocrine mechanism for the regulation of beta-cell function.


Subject(s)
Glucose/pharmacology , Insulin/metabolism , Insulin/pharmacology , Islets of Langerhans/metabolism , Receptor, Insulin/metabolism , Tyrosine/analogs & derivatives , Animals , Calcium/pharmacology , Cell Line , Dose-Response Relationship, Drug , Epinephrine/pharmacology , Insulin Receptor Substrate Proteins , Insulin Secretion , Insulin-Like Growth Factor I/pharmacology , Islets of Langerhans/drug effects , Kinetics , Phosphatidylinositol 3-Kinases , Phosphoproteins/metabolism , Phosphorylation , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Phosphotyrosine , Potassium Chloride/pharmacology , Receptor, Insulin/drug effects , Tyrosine/analysis , Tyrosine/metabolism
16.
J Clin Invest ; 90(5): 1839-49, 1992 Nov.
Article in English | MEDLINE | ID: mdl-1331176

ABSTRACT

Insulin rapidly stimulates tyrosine phosphorylation of a protein of approximately 185 kD in most cell types. This protein, termed insulin receptor substrate-1 (IRS-1), has been implicated in insulin signal transmission based on studies with insulin receptor mutants. In the present study we have examined the levels of IRS-1 and the phosphorylation state of insulin receptor and IRS-1 in liver and muscle after insulin stimulation in vivo in two rat models of insulin resistance, i.e., insulinopenic diabetes and fasting, and a mouse model of non-insulin-dependent diabetes mellitus (ob/ob) by immunoblotting with anti-peptide antibodies to IRS-1 and anti-phosphotyrosine antibodies. As previously described, there was an increase in insulin binding and a parallel increase in insulin-stimulated receptor phosphorylation in muscle of fasting and streptozotocin-induced (STZ) diabetic rats. There was also a modest increase in overall receptor phosphorylation in liver in these two models, but when normalized for the increase in binding, receptor phosphorylation was decreased, in liver and muscle of STZ diabetes and in liver of 72 h fasted rats. In the hyperinsulinemic ob/ob mouse there was a decrease in insulin binding and receptor phosphorylation in both liver and muscle. The tyrosyl phosphorylation of IRS-1 after insulin stimulation reflected an amplification of the receptor phosphorylation in liver and muscle of hypoinsulinemic animals (fasting and STZ diabetes) with a twofold increase, and showed a significant reduction (approximately 50%) in liver and muscle of ob/ob mouse. By contrast, the levels of IRS-1 protein showed a tissue specific regulation with a decreased level in muscle and an increased level in liver in hypoinsulinemic states of insulin resistance, and decreased levels in liver in the hyperinsulinemic ob/ob mouse. These data indicate that: (a) IRS-1 protein levels are differentially regulated in liver and muscle; (b) insulin levels may play a role in this differential regulation of IRS-1; (c) IRS-1 phosphorylation depends more on insulin receptor kinase activity than IRS-1 protein levels; and (d) reduced IRS-1 phosphorylation in liver and muscle may play a role in insulin-resistant states, especially of the ob/ob mice.


Subject(s)
Insulin Resistance , Liver/metabolism , Muscles/metabolism , Protein-Tyrosine Kinases/physiology , Proteins/metabolism , Receptor, Insulin/metabolism , Animals , CHO Cells , Cricetinae , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Type 2/metabolism , Disease Models, Animal , Fasting , Insulin/pharmacology , Male , Mice , Mice, Obese , Phosphorylation , Rats
17.
J Biol Chem ; 267(25): 17483-6, 1992 Sep 05.
Article in English | MEDLINE | ID: mdl-1381348

ABSTRACT

The major cytosolic substrate of the insulin receptor is a 185-kDa phosphoprotein (IRS-1) that contains multiple putative attachment sites for the p85 alpha regulatory subunit of phosphatidylinositol 3'-kinase (PI3K). To examine the possible interaction of pp185 with p85 alpha in vivo, we injected insulin or insulinomimetic agents (a combination of H2O2 and vanadate (H/V)) into the portal vein of anesthetized rats. IN this model system, H/V treatment and, to a lesser extent, injection of insulin resulted in rapid and sustained tyrosine phosphorylation of multiple cellular proteins, including pp185/IRS-1. The latter was found to undergo specific association with the p85 alpha regulatory subunit of PI3K but not with two other proteins that contain src homology domains. As p85 alpha was not detectably phosphorylated on tyrosine residues and did not appear to interact directly with the insulin receptor, we conclude that tyrosine phosphorylation of pp185 promotes its association with p85 alpha and the catalytic subunit of PI3K. The recruitment of the holoenzyme may also involve its enzymatic activation and thus constitute an important step in the transduction of insulin signals.


Subject(s)
Hydrogen Peroxide/pharmacology , Insulin/pharmacology , Liver/enzymology , Phosphoproteins/metabolism , Phosphotransferases/metabolism , Protein-Tyrosine Kinases/metabolism , Receptor, Insulin/metabolism , Vanadates/pharmacology , Animals , Blotting, Western , Enzyme Activation , Insulin Receptor Substrate Proteins , Macromolecular Substances , Male , Phosphatidylinositol 3-Kinases , Phosphoproteins/isolation & purification , Phosphorylation , Phosphotyrosine , Rats , Rats, Inbred Strains , Tyrosine/analogs & derivatives , Tyrosine/analysis
18.
J Biol Chem ; 267(6): 3946-51, 1992 Feb 25.
Article in English | MEDLINE | ID: mdl-1310987

ABSTRACT

Genistein, an isoflavone putative tyrosine kinase inhibitor, was used to investigate the coupling of insulin receptor tyrosine kinase activation to four metabolic effects of insulin in the isolated rat adipocyte. Genistein inhibited insulin-stimulated glucose oxidation in a concentration-dependent manner with an ID50 of 25 micrograms/ml and complete inhibition at 100 micrograms/ml. Genistein also prevented insulin's (10(-9) M) inhibition of isoproterenol-stimulated lipolysis with an ID50 of 15 micrograms/ml and a complete effect at 50 micrograms/ml. The effect of genistein (25 micrograms/ml) was not reversed by supraphysiological (10(-7) M) insulin levels. In contrast, genistein up to 100 micrograms/ml had no effect on insulin's (10(-9) M) stimulation of either pyruvate dehydrogenase or glycogen synthase activity. We determined whether genistein influenced insulin receptor beta-subunit autophosphorylation or tyrosine kinase substrate phosphorylation either in vivo or in vitro by anti-phosphotyrosine immunoblotting. Genistein at 100 micrograms/ml did not inhibit insulin's (10(-7) M) stimulation of insulin receptor tyrosine autophosphorylation or tyrosine phosphorylation of the cellular substrates pp185 and pp60. Also, genistein did not prevent insulin-stimulated autophosphorylation of partially purified human insulin receptors from NIH 3T3/HIR 3.5 cells or the phosphorylation of histones by the activated receptor tyrosine kinase. In control experiments using either NIH 3T3 fibroblasts or partially purified membranes from these cells, genistein did inhibit platelet-derived growth factor's stimulation of its receptor autophosphorylation. These findings indicate the following: (a) Genistein can inhibit certain responses to insulin without blocking insulin's stimulation of its receptor tyrosine autophosphorylation or of the receptor kinase substrate tyrosine phosphorylation. (b) In adipocytes genistein must block the stimulation of glucose oxidation and the antilipolytic effects of insulin at site(s) downstream from the insulin receptor tyrosine kinase. (c) The inhibitory effects of genistein on hormonal signal transduction cannot necessarily be attributed to inhibition of tyrosine kinase activity, unless specifically demonstrated.


Subject(s)
Adipose Tissue/drug effects , Insulin Antagonists/pharmacology , Insulin/pharmacology , Isoflavones/pharmacology , Protein-Tyrosine Kinases/antagonists & inhibitors , Receptor, Insulin/drug effects , Adipose Tissue/cytology , Animals , Blotting, Western , Cells, Cultured , Electrophoresis, Polyacrylamide Gel , Enzyme Activation , Genistein , Glucose/metabolism , Glycogen Synthase/metabolism , Isoproterenol/pharmacology , Lipolysis/drug effects , Male , Oxidation-Reduction , Phosphorylation , Protein-Tyrosine Kinases/metabolism , Pyruvate Dehydrogenase Complex/metabolism , Rats , Rats, Inbred Strains , Signal Transduction/drug effects
19.
Nature ; 352(6330): 73-7, 1991 Jul 04.
Article in English | MEDLINE | ID: mdl-1648180

ABSTRACT

Since the discovery of insulin nearly 70 years ago, there has been no problem more fundamental to diabetes research than understanding how insulin works at the cellular level. Insulin binds to the alpha subunit of the insulin receptor which activates the tyrosine kinase in the beta subunit, but the molecular events linking the receptor kinase to insulin-sensitive enzymes and transport processes are unknown. Our discovery that insulin stimulates tyrosine phosphorylation of a protein of relative molecular mass between 165,000 and 185,000, collectively called pp185, showed that the insulin receptor kinase has specific cellular substrates. The pp185 is a minor cytoplasmic phosphoprotein found in most cells and tissues; its phosphorylation is decreased in cells expressing mutant receptors defective in signalling. We have now cloned IRS-1, which encodes a component of the pp185 band. IRS-1 contains over ten potential tyrosine phosphorylation sites, six of which are in Tyr-Met-X-Met motifs. During insulin stimulation, the IRS-1 protein undergoes tyrosine phosphorylation and binds phosphatidylinositol 3-kinase, suggesting that IRS-1 acts as a multisite 'docking' protein to bind signal-transducing molecules containing Src-homology 2 and Src-homology-3 domains. Thus IRS-1 may link the insulin receptor kinase and enzymes regulating cellular growth and metabolism.


Subject(s)
Phosphoproteins/chemistry , Protein-Tyrosine Kinases/physiology , Signal Transduction , Animals , Base Sequence , Cricetinae , Insulin/pharmacology , Molecular Sequence Data , Phosphoproteins/genetics , Phosphoproteins/physiology , Phosphorylation , Rats , Receptor, Insulin , Substrate Specificity
20.
J Behav Med ; 14(3): 267-85, 1991 Jun.
Article in English | MEDLINE | ID: mdl-1714962

ABSTRACT

To assess the impact of a stressor, it is desirable to evaluate affected individuals' status both prior to and following a stressful event. Because of the difficulties inherent in prospective designs, investigators often ask people who have experienced an aversive event to evaluate their prestressor adjustment retrospectively. Do such retrospective evaluations provide a reasonable alternative to prospective assessment? To answer this question we compared retrospective and prospective data gathering procedures in the evaluation of sexual adjustment after prostate surgery. One hundred fifty-two married males who had undergone prostatectomy for benign prostatic enlargement completed a battery of measures which evaluated pre- and postsurgical sexual adjustment either prospectively (i.e., before and after surgery) or retrospectively (i.e., ratings made after surgery of both pre- and postsurgical adjustment). Retrospective assessment indicated considerable sexual deterioration pre- to postsurgery. In subjects tested prospectively, however, the results showed that surgery had little impact on sexual adjustment. Moreover, direct comparisons of retrospective and prospective methodologies reveal that discrepancies are due to differences in evaluations of presurgery status, with retrospective evaluation yielding more favorable ratings than prospective assessment. The results highlight a variety of biases which may affect self-ratings of pre- and post-stressor adaptation and show that discrepancies associated with the two methodologies have important implications for understanding the impact of a stressor on adjustment.


Subject(s)
Prostatectomy/psychology , Research Design , Sexual Behavior/psychology , Aged , Analysis of Variance , Bias , Humans , Life Change Events , Male , Mental Recall , Middle Aged , Personality Inventory , Prospective Studies , Prostatic Hyperplasia/surgery , Retrospective Studies , Risk Factors , Stress, Psychological/psychology
SELECTION OF CITATIONS
SEARCH DETAIL
...