Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
1.
Clin Transl Sci ; 10(1): 3-11, 2017 01.
Article in English | MEDLINE | ID: mdl-27860267

ABSTRACT

This phase I study assessed the safety, tolerability, pharmacokinetics, and pharmacodynamics of RN317 (PF-05335810), a specifically engineered, pH-sensitive, humanized proprotein convertase subtilisin kexin type 9 (PCSK9) monoclonal antibody, in hypercholesterolemic subjects (low-density lipoprotein cholesterol (LDL-C) ≥ 80 mg/dl) 18-70 years old receiving statin therapy. Subjects were randomized to: single-dose placebo, RN317 (subcutaneous (s.c.) 0.3, 1, 3, 6, or intravenous (i.v.) 1, 3, 6 mg/kg), or bococizumab (s.c. 1, 3, or i.v. 1 mg/kg); or multiple-dose RN317 (s.c. 300 mg every 28 days; three doses). Of 133 subjects randomized, 127 completed the study. RN317 demonstrated a longer half-life, greater exposure, and increased bioavailability vs. bococizumab. RN317 was well tolerated, with no subjects discontinuing because of treatment-related adverse events. RN317 lowered LDL-C by up to 52.5% (day 15) following a single s.c. dose of 3.0 mg/kg vs. a maximum of 70% with single-dose bococizumab s.c. 3.0 mg/kg. Multiple dosing of RN317 produced LDL-C reductions of ∼50%, sustained over an 85-day dosing interval.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Hydroxymethylglutaryl-CoA Reductase Inhibitors/therapeutic use , Hypercholesterolemia/drug therapy , PCSK9 Inhibitors , Protein Engineering , Antibodies, Monoclonal/adverse effects , Antibodies, Monoclonal/blood , Antibodies, Monoclonal/pharmacokinetics , Antibodies, Monoclonal, Humanized/pharmacokinetics , Antibodies, Monoclonal, Humanized/therapeutic use , Cholesterol, LDL/blood , Demography , Dose-Response Relationship, Drug , Drug Therapy, Combination , Female , Humans , Hydrogen-Ion Concentration , Hypercholesterolemia/blood , Injections, Intravenous , Injections, Subcutaneous , Male , Middle Aged , Proprotein Convertase 9/metabolism
2.
Am J Physiol Endocrinol Metab ; 278(2): E340-51, 2000 Feb.
Article in English | MEDLINE | ID: mdl-10662719

ABSTRACT

To study effects of Bcl-x(L) in the pancreatic beta-cell, two transgenic lines were produced using different forms of the rat insulin promoter. Bcl-x(L) expression in beta-cells was increased 2- to 3-fold in founder (Fd) 1 and over 10-fold in Fd 2 compared with littermate controls. After exposure to thapsigargin (10 microM for 48 h), losses of cell viability in islets of Fd 1 and Fd 2 Bcl-x(L) transgenic mice were significantly lower than in islets of wild-type mice. Unexpectedly, severe glucose intolerance was observed in Fd 2 but not Fd 1 Bcl-x(L) mice. Pancreatic insulin content and islet morphology were not different from control in either transgenic line. However, Fd 2 Bcl-x(L) islets had impaired insulin secretory and intracellular free Ca(2+) ([Ca(2+)](i)) responses to glucose and KCl. Furthermore, insulin and [Ca(2+)](i) responses to pyruvate methyl ester (PME) were similarly reduced as glucose in Fd 2 Bcl-x(L) islets. Consistent with a mitochondrial defect, glucose oxidation, but not glycolysis, was significantly lower in Fd 2 Bcl-x(L) islets than in wild-type islets. Glucose-, PME-, and alpha-ketoisocaproate-induced hyperpolarization of mitochondrial membrane potential, NAD(P)H, and ATP production were also significantly reduced in Fd 2 Bcl-x(L) islets. Thus, although Bcl-x(L) promotes beta-cell survival, high levels of expression of Bcl-x(L) result in reduced glucose-induced insulin secretion and hyperglycemia due to a defect in mitochondrial nutrient metabolism and signaling for insulin secretion.


Subject(s)
Apoptosis , Gene Expression , Insulin/metabolism , Islets of Langerhans/metabolism , Mitochondria/physiology , Proto-Oncogene Proteins c-bcl-2/genetics , Animals , Apoptosis/drug effects , Blood Glucose/metabolism , Calcium/metabolism , DNA Fragmentation/drug effects , Glucose/pharmacology , Glucose Tolerance Test , Immunohistochemistry , Insulin/analysis , Insulin Secretion , Islets of Langerhans/cytology , Islets of Langerhans/ultrastructure , Membrane Potentials/drug effects , Mice , Mice, Transgenic , Potassium Chloride/pharmacology , Proto-Oncogene Proteins c-bcl-2/analysis , Rats , Thapsigargin/pharmacology , bcl-X Protein
3.
Transplantation ; 68(3): 331-7, 1999 Aug 15.
Article in English | MEDLINE | ID: mdl-10459535

ABSTRACT

BACKGROUND: Insulin-dependent diabetes mellitus (IDDM) is the second most prevalent chronic illness of children. Investigation of the treatment of IDDM is hindered by the lack of a reproducible and easily maintained non-human primate model of this disorder. METHODS: We induced IDDM in 11 juvenile cynomolgus monkeys after a single (150 mg/kg) intravenous injection of streptozotocin (STZ). All diabetic monkeys were treated with insulin twice daily, based on a sliding scale. Subcutaneous vascular access ports were surgically placed in each monkey to facilitate serial blood sampling and drug administration. Allogeneic pancreatic islet cells from unrelated donors were subsequently transplanted into the mesenteric circulation of all STZ-treated monkeys. RESULTS: Mild, transient nausea and vomiting occurred in all animals after STZ injection; however, no additional signs of toxicity occurred. Within 36 hr, all monkeys required twice daily administration of exogenous insulin to maintain a non-ketotic state. Serum C-peptide levels decreased from >1.2 ng/ml before STZ, to between 0.0 and 0.9 ng/ml after STZ, confirming islet cell destruction. Animals were maintained in an insulin-dependent state for up to 147 days without any observable clinical complications. Subcutaneous vascular access port patency was maintained up to 136 days with a single incidence of local infection. Islet cell transplantation resulted in normoglycemia within 24 hr. Serum C-peptide levels increased (range: 2-8 ng/ml) for 6 - 8 days in immune competent animals, and for 39-98 days after transplant in immunosuppressed monkeys. CONCLUSIONS: IDDM can be consistently induced and safely treated in juvenile cynomolgus monkeys. Chronic vascular access can be maintained with minimal supervision and complications. This model is appropriate for studies investigating potential treatments for IDDM including islet cell transplantation.


Subject(s)
Diabetes Mellitus, Experimental/etiology , Diabetes Mellitus, Type 1/etiology , Animals , C-Peptide/blood , Catheterization , Child, Preschool , Chronic Disease , Diabetes Mellitus, Experimental/prevention & control , Disease Models, Animal , Femoral Vein , Glucose Tolerance Test , Humans , Immunosuppressive Agents/pharmacology , Insulin/therapeutic use , Insulin Infusion Systems , Islets of Langerhans Transplantation/methods , Kidney/pathology , Macaca fascicularis , Pancreas/pathology , Streptozocin , Vascular Patency
4.
Diabetes ; 48(5): 989-96, 1999 May.
Article in English | MEDLINE | ID: mdl-10331402

ABSTRACT

To determine whether loss of beta-cell mass and function in the NOD mouse occurs gradually, beginning after the onset of insulitis, or abruptly, just before the onset of overt diabetes, beta-cell mass and rates of beta-cell proliferation and insulin secretory responses from the perfused pancreas were measured in NOD and control NOD/Scid mice at 8-9, 13, and 18 weeks of age. Of the NOD mice, 11 and 70% had diabetes (fasting blood glucose >8.3 mmol/l) at 13 and 18 weeks of age, respectively. Beta-cell mass in 8-week-old NOD mice was 69% of control mice (P>0.05), but the rate of 5-bromo-2-deoxyuridine uptake was greater, suggesting a compensatory proliferative response to ongoing autoimmune beta-cell destruction. Despite an increase in the rate of beta-cell proliferation, beta-cell mass was significantly reduced by 42% in 13-week-old nondiabetic NOD mice and by 73% in 18-week-old diabetic NOD mice. Insulin secretory responses to glucose and arginine demonstrated reductions of similar magnitude. In 18-week-old diabetic NOD mice, insulin secretion was reduced to a greater degree than beta-cell mass, suggesting the presence of beta-cell dysfunction in addition to reduced mass. These results suggest that in the NOD mouse, beta-cell destruction begins soon after the onset of insulitis. Despite a compensatory beta-cell proliferative response, beta-cell mass progressively falls and is significantly reduced by 13 weeks despite normal blood glucose concentrations. Diabetes may be present when residual beta-cell mass represents 30% of control levels.


Subject(s)
Cell Division , Diabetes Mellitus, Type 1/pathology , Islets of Langerhans/pathology , Aging , Animals , Arginine/pharmacology , Blood Glucose/metabolism , Body Weight , Female , Glucose/pharmacology , Insulin/metabolism , Insulin Secretion , Islets of Langerhans/metabolism , Mice , Mice, Inbred NOD
5.
Diabetes ; 47(12): 1881-8, 1998 Dec.
Article in English | MEDLINE | ID: mdl-9836519

ABSTRACT

The present study was undertaken to test the hypothesis that exposure to high glucose concentrations enhances insulin secretion in pancreatic islets from glucokinase-deficient mice. Insulin secretion and intracellular calcium ([Ca2+]i) were measured as the glucose concentration was increased from 2 to 26 mmol/l in islets from heterozygous glucokinase (GK)-deficient mice (GK+/-) and their wild-type littermates (GK+/+). Results obtained in islets incubated in 11.6 or 30 mmol/l glucose for 48-96 h were compared. GK+/- islets that had been incubated in 30 mmol/l glucose showed improved although not normal insulin secretory and [Ca2+]i responses to the standard glucose challenge as well as an enhanced ability to sense small amplitude glucose oscillations. These effects were associated with increased glucokinase activity and protein. In contrast, exposure of GK+/+ islets to 30 mmol/l glucose increased their basal insulin secretion but reduced their incremental secretory responses to glucose and their ability to detect small amplitude glucose oscillations. Thus exposure of GK+/- islets to 30 mmol/l glucose for 48-96 h enhanced their ability to sense and respond to a glucose stimulus, whereas similar exposure of GK+/+ islets induced evidence of beta-cell dysfunction. These findings provide a mechanistic framework for understanding why glucokinase diabetes results in mild hyperglycemia that tends not to increase over time. In addition, the absence of one allele of the glucokinase gene appears to protect against glucose-induced beta-cell dysfunction (glucose toxicity).


Subject(s)
Glucokinase/genetics , Hyperglycemia/physiopathology , Insulin/metabolism , Animals , Blood Glucose/metabolism , Body Weight , Calcium/metabolism , Dose-Response Relationship, Drug , Glucokinase/drug effects , Glucokinase/metabolism , Glucose/administration & dosage , Glucose/pharmacology , Glucose Tolerance Test , Insulin Secretion , Islets of Langerhans/cytology , Islets of Langerhans/drug effects , Islets of Langerhans/metabolism , Mice , Mutation
6.
J Biol Chem ; 273(38): 24457-64, 1998 Sep 18.
Article in English | MEDLINE | ID: mdl-9733737

ABSTRACT

Mutations in the hepatocyte nuclear factor-1alpha (HNF-1alpha) gene cause maturity onset diabetes of the young type 3, a form of type 2 diabetes mellitus. In mice lacking the HNF-1alpha gene, insulin secretion and intracellular calcium ([Ca2+]i) responses were impaired following stimulation with nutrient secretagogues such as glucose and glyceraldehyde but normal with non-nutrient stimuli such as potassium chloride. Patch clamp recordings revealed ATP-sensitive K+ currents (KATP) in beta-cells that were insensitive to suppression by glucose but normally sensitive to ATP. Exposure to mitochondrial substrates suppressed KATP, elevated [Ca2+]i, and corrected the insulin secretion defect. NAD(P)H responses to glucose were substantially reduced, and inhibitors of glycolytic NADH generation reproduced the mutant phenotype in normal islets. Flux of glucose through glycolysis in islets from mutant mice was reduced, as a result of which ATP generation in response to glucose was impaired. We conclude that hepatocyte nuclear factor-1alpha diabetes results from defective beta-cell glycolytic signaling, which is potentially correctable using substrates that bypass the defect.


Subject(s)
Glycolysis , Insulin/metabolism , Islets of Langerhans/physiology , Nuclear Proteins , Transcription Factors/physiology , Adenosine Triphosphate/metabolism , Animals , Calcium/metabolism , DNA-Binding Proteins/physiology , Glucose/pharmacology , Glucose/physiology , Glyceraldehyde/pharmacology , Hepatocyte Nuclear Factor 1 , Hepatocyte Nuclear Factor 1-alpha , Hepatocyte Nuclear Factor 1-beta , In Vitro Techniques , Insulin Secretion , Islets of Langerhans/drug effects , Membrane Potentials/drug effects , Membrane Potentials/physiology , Mice , Mice, Knockout , Patch-Clamp Techniques , Potassium Channels/physiology , Potassium Chloride/pharmacology , Signal Transduction , Tolbutamide/pharmacology , Transcription Factors/deficiency , Transcription Factors/genetics
7.
J Clin Invest ; 101(10): 2215-22, 1998 May 15.
Article in English | MEDLINE | ID: mdl-9593777

ABSTRACT

Mutations in the gene for the transcription factor hepatocyte nuclear factor (HNF) 1alpha cause maturity-onset diabetes of the young (MODY) 3, a form of diabetes that results from defects in insulin secretion. Since the nature of these defects has not been defined, we compared insulin secretory function in heterozygous [HNF-1alpha (+/-)] or homozygous [HNF-1alpha (-/-)] mice with null mutations in the HNF-1alpha gene with their wild-type littermates [HNF-1alpha (+/+)]. Blood glucose concentrations were similar in HNF-1alpha (+/+) and (+/-) mice (7.8+/-0.2 and 7.9+/-0.3 mM), but were significantly higher in the HNF-1alpha (-/-) mice (13.1+/-0.7 mM, P < 0.001). Insulin secretory responses to glucose and arginine in the perfused pancreas and perifused islets from HNF-1alpha (-/-) mice were < 15% of the values in the other two groups and were associated with similar reductions in intracellular Ca2+ responses. These defects were not due to a decrease in glucokinase or insulin gene transcription. beta cell mass adjusted for body weight was not reduced in the (-/-) animals, although pancreatic insulin content adjusted for pancreas weight was slightly lower (0.06+/-0.01 vs. 0.10+/-0.01 microg/mg, P < 0.01) than in the (+/+) animals. In summary, a null mutation in the HNF-1alpha gene in homozygous mice leads to diabetes due to alterations in the pathways that regulate beta cell responses to secretagogues including glucose and arginine. These results provide further evidence in support of a key role for HNF-1alpha in the maintenance of normal beta cell function.


Subject(s)
DNA-Binding Proteins , Diabetes Mellitus, Type 2/physiopathology , Insulin/metabolism , Nuclear Proteins , Transcription Factors/physiology , Animals , Arginine/pharmacology , Blood Glucose/analysis , Body Weight , Calcium/analysis , Gene Expression Regulation/genetics , Glucose/pharmacology , Hepatocyte Nuclear Factor 1 , Hepatocyte Nuclear Factor 1-alpha , Hepatocyte Nuclear Factor 1-beta , Heterozygote , Homozygote , Immunohistochemistry , Insulin Secretion , Islets of Langerhans/chemistry , Islets of Langerhans/physiopathology , Mice , Mice, Knockout , Organ Size , Pancreas/pathology , Pancreas/physiopathology , RNA, Messenger/analysis , Transcription Factors/genetics
8.
Diabetes ; 47(3): 358-64, 1998 Mar.
Article in English | MEDLINE | ID: mdl-9519740

ABSTRACT

To define the mechanisms involved in the evolution of diabetes in the Zucker diabetic fatty (ZDF) rat, beta-cell mass and replication rates were determined by immunochemistry, point-counting morphometry, and 6-h 5-bromo-2'-deoxyuridine (BrdU) incorporation. The beta-cell mass in 5- to 7-week-old prediabetic ZDF rats (4.3 +/- 0.06 mg) was similar to age-matched insulin-resistant Zucker fatty (ZF) rats (3.7 +/- 0.05 mg) and greater than that in Zucker lean control (ZLC) rats (1.9 +/- 0.3, P < 0.05). At 12 weeks (after diabetes onset), beta-cell mass in the ZDF rats (8.1 +/- 1.7 mg) was significantly lower than the ZF rats (15.7 +/- 1.8 mg). The mass in the ZF rats was significantly greater than in the ZLC rats (4.3 +/- 0.8 mg, P < 0.05). The beta-cell proliferation rate (mean of both time points) was significantly greater in the ZDF rats (0.88 +/- 0.1%) compared with the ZF and ZLC rats (0.53 +/- 0.07%, 0.62 +/- 0.07%, respectively, P < 0.05), yet ZDF rats have a lower beta-cell mass than the ZF rats despite a higher proliferative rate. Morphological evidence of neogenesis and apoptosis is evident in the ZF and ZDF rats. In addition, even at 5-7 weeks a modest defect in insulin secretion per beta-cell unit was found by pancreas perfusion. These studies provide evidence that the expansion of beta-cell mass in response to insulin resistance and insulin secretory defects in diabetic ZDF rats is inadequate. This failure of beta-cell mass expansion in the ZDF rat does not appear to be from a reduction in the rate of beta-cell proliferation or neogenesis, suggesting an increased rate of cell death by apoptosis.


Subject(s)
Apoptosis/physiology , Diabetes Mellitus, Type 2/pathology , Insulin Resistance/physiology , Insulin/metabolism , Islets of Langerhans/metabolism , Islets of Langerhans/pathology , Animals , Cohort Studies , Disease Models, Animal , Dose-Response Relationship, Drug , Glucagon/analysis , Glucose/pharmacology , Glucose Intolerance/etiology , Glucose Tolerance Test , Immunohistochemistry , In Vitro Techniques , Insulin/analysis , Insulin Secretion , Islets of Langerhans/cytology , Male , Pancreatic Polypeptide/analysis , Perfusion , Rats , Rats, Zucker , Somatostatin/analysis , Time Factors
9.
J Immunol ; 159(11): 5187-91, 1997 Dec 01.
Article in English | MEDLINE | ID: mdl-9548454

ABSTRACT

Ag-specific T cell activation requires a CD28-mediated costimulatory interaction. This observation has suggested novel approaches to suppress donor-specific immunity, including the use of soluble CD28 antagonists, such as CTLA4Ig, which suppresses transplant rejection in small animal models. In this study, CTLA4Ig therapy was examined in a non-human primate model of allogeneic pancreatic islet transplantation. Two of five CTLA4Ig-treated monkeys showed prolonged graft survival, which correlated with donor-specific hyporesponsiveness in vitro. Humoral responses to the transplanted tissue were suppressed in all treated animals. These results suggest that CTLA4Ig is effective in suppressing both humoral and cellular immune responses in a non-human primate model of allogeneic transplantation.


Subject(s)
Antigens, Differentiation/pharmacology , Immunoconjugates , Islets of Langerhans Transplantation/immunology , Abatacept , Animals , Antigens, CD , CTLA-4 Antigen , Diabetes Mellitus, Experimental/surgery , Graft Survival , Immunoglobulin Fc Fragments/pharmacology , Isoantibodies/analysis , Lymphocyte Culture Test, Mixed , Macaca fascicularis , Recombinant Fusion Proteins/pharmacology , Transplantation, Homologous
10.
Diabetes ; 44(8): 947-53, 1995 Aug.
Article in English | MEDLINE | ID: mdl-7622001

ABSTRACT

To examine the kinetic steps in insulin's in vivo action, we have assessed the temporal relationship between arterial insulin, interstitial insulin, glucose disposal rate (GDR), and insulin receptor kinase (IRK) activity in muscle and between portal insulin, hepatic glucose production (HGP), and IRK activity in liver. Interstitial insulin, as measured by lymph-insulin concentration (muscle only), and IRK activity were used as independent methods to determine the arrival of insulin at its tissue site of action. Euglycemic clamps were conducted in seven mongrel dogs and consisted of an activation phase with a venous insulin infusion (7.2 nmol.kg-1.min-1, 100 min) and a deactivation phase. Liver and muscle biopsies were taken to assess IRK activity. Arterial, portal, and lymph insulin rose to 636 +/- 12, 558 +/- 18, and 402 +/- 24 pmol/l, respectively. GDR increased from 13.9 +/- 0.6 to 41.7 +/- 2.8, and HGP declined from 14.4 +/- 0.6 to 1.1 +/- 0.6 mumol.kg-1.min-1. Muscle and liver IRK activity increased significantly from 5.9 +/- 0.9 to 14.6 +/- 0.6 and 5.5 +/- 0.7 to 23.7 +/- 1.9 fmol P/fmol insulin receptor (IR), respectively. The time to half-maximum response (t1/2a) for stimulation of GDR (19.8 +/- 4.8 min) and suppression of HGP (21.5 +/- 3.7 min) were similar. The t1/2a for stimulation of GDR, muscle IRK, and rise in lymph insulin were not significantly different from one another and were all markedly greater than that for the approach to steady state of arterial insulin (2.3 +/- 1.2 min, P < 0.01).(ABSTRACT TRUNCATED AT 250 WORDS)


Subject(s)
Glucose/metabolism , Glycolysis , Insulin/metabolism , Insulin/pharmacology , Liver/metabolism , Muscle, Skeletal/metabolism , Receptor, Insulin/metabolism , Animals , Dogs , Glucose Clamp Technique , Glycolysis/drug effects , Insulin/blood , Kinetics , Lymph/metabolism , Male , Time Factors
12.
Article in English | MEDLINE | ID: mdl-7849918

ABSTRACT

A high-concentration 90% w/v perflubron (perfluorooctyl bromide [PFOB]) emulsion (Oxygent HT) is being evaluated as an oxygen carrier for use during surgery. This study was done to assess oxygen delivery by Oxygent HT during acute normovolemic hemodilution. Anesthetized mongrel dogs, instrumented with femoral and pulmonary artery catheters, were hemodiluted to a hematocrit of 25% with 3:1 (v/v) of Ringers-lactate (R-L). Dogs were then ventilated with 100% O2 and hemodiluted to a Hct approximately 11% with 1.5 (v/v) of colloid (autologous plasma and 5% albumin). Dogs then received either 3.3 mL/kg Oxygent HT (n = 5) or 3.3 mL/kg R-L (n = 4), and were monitored for 3 hours. Total oxygen delivery (DO2), blood oxygen content, cardiac output, mixed venous PO2, and mixed venous Hb saturation was higher in Oxygent HT treated dogs compared to the R-L controls. The percentage of total DO2 contributed by perflubron-dissolved oxygen was about 8-10% and accounted for 25-30% of total oxygen consumption (VO2). The percentage of VO2 contributed by Hb-carried oxygen was significantly higher in R-L controls (46 +/- 4%) than in the treated dogs (15 +/- 3%), indicating that the availability of the perflubron-dissolved oxygen allowed for a reserve of oxygen to remain available in the red blood cells.


Subject(s)
Blood Substitutes/therapeutic use , Fluorocarbons/therapeutic use , Hemodilution , Oxygen/administration & dosage , Animals , Dogs , Drug Carriers , Emulsions , Hydrocarbons, Brominated , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...