Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
Add more filters










Publication year range
1.
J Pharm Pharm Sci ; 27: 13074, 2024.
Article in English | MEDLINE | ID: mdl-38919469

ABSTRACT

Purpose: This study evaluated the safety and efficacy of two insulin regimens for inpatient hyperglycemia management: combination short-plus long-acting insulin (basal-bolus insulin regimen, BBIR) vs. short-acting insulin only (correctional insulin only regimen, CIOR). Methods: Chart reviews identified noncritically ill patients with pre-existing type 2 diabetes mellitus receiving insulin injections. Study participants (N = 138) were divided into BBIR (N = 104) and CIOR (N = 34) groups. Data for the entire duration of each patient's stay were analyzed. Results: The primary outcome of percent hyperglycemic days was higher in BBIR vs. CIOR (3.97 ± 0.33% vs. 1.22 ± 0.38%). The safety outcome of percent hypoglycemic events was not different between BBIR and CIOR (0.78 ± 0.22% vs. 0.53 ± 0.37%). Regarding secondary outcomes, the percentage of euglycemic days was lower in BBIR vs. CIOR (26.74 ± 2.97% vs. 40.98 ± 5.91%). Overall blood glucose (BG) and daily insulin dose were higher in BBIR vs. CIOR (231.43 ± 5.37 vs. 195.55 ± 6.25 mg/dL and 41.36 ± 3.07 vs. 5.02 ± 0.68 units, respectively). Insulin regimen-associated differences in hyperglycemia and daily insulin dose persisted after adjusting for covariates. Conclusion: Our observations linking BBIR to worse glycemic outcomes differ from those reported in the randomized controlled Rabbit 2 and Rabbit 2 Surgery trials. This discrepancy can be partly explained by the fact that BBIR patients displayed worse glycemic baselines. Also, there was no diabetes stewardship team to monitor BG and modify insulin therapy, which is relevant since achieving euglycemia in BBIR patients requires more dose adjustments. This study highlights challenges with standard inpatient glycemic management and calls for further research assessing the benefits of pharmacist-led diabetes stewardship.


Subject(s)
Diabetes Mellitus, Type 2 , Hospitals, Community , Hyperglycemia , Hypoglycemic Agents , Insulin , Humans , Diabetes Mellitus, Type 2/drug therapy , Male , Female , Hyperglycemia/drug therapy , Middle Aged , Insulin/administration & dosage , Insulin/therapeutic use , Hypoglycemic Agents/administration & dosage , Hypoglycemic Agents/therapeutic use , Aged , Retrospective Studies , Blood Glucose/drug effects
2.
Front Psychiatry ; 15: 1386263, 2024.
Article in English | MEDLINE | ID: mdl-38716117

ABSTRACT

Schizophrenia is a serious mental health disorder that confers one of the highest mortality rates of all psychiatric illnesses. Although the disorder's psychotic symptoms are treatable with conventional antipsychotics, they remain incurable. Moreover, medication adherence is poor, and individuals with schizophrenia choose to self-medicate with illicit substances, including cannabis. It is well-established that the delta-9-tetrahydrocannabinol (delta-9-THC) component of cannabis elicits psychotomimetic effects at high doses; worsens schizophrenia-related psychosis; commonly develops into cannabis use disorder in individuals with schizophrenia; and increases the risk of earlier-onset schizophrenia symptoms in those harboring genetic susceptibility. However, individuals with schizophrenia commonly use cannabis and cannabis derivatives such as cannabidiol (CBD). These products seem to alleviate psychotic symptoms and relieve adverse side effects of antipsychotic medications. Therefore, one notion that has gained traction is the potential utility of cannabis-derived cannabidiol (CBD) as adjunct treatment to reduce schizophrenia-associated psychosis and other symptoms. Currently, preclinical and clinical data remain inconclusive. The present review distinguishes the mechanisms underlying schizophrenia-associated vs. cannabis-induced psychosis; reviews the evidence for delta-9-THC-mediated exacerbation vs. CBD-mediated amelioration of schizophrenia-associated psychosis; and describes potential approaches for incorporating CBD into schizophrenia therapeutic regimen in a safe and efficacious manner.

3.
Front Endocrinol (Lausanne) ; 14: 1205490, 2023.
Article in English | MEDLINE | ID: mdl-37396171

ABSTRACT

Obesity adversely impacts millions of American adults by predisposing them to significant health risks and further complications. Obesity is differentiated into two groups: metabolically healthy and metabolically unhealthy. In contrast to metabolically healthy counterparts, obese individuals who are metabolically unhealthy display hallmark symptoms of metabolic syndrome (e.g., hypertension, dyslipidemia, hyperglycemia, abdominal obesity). Gastroesophageal reflux disease (GERD) commonly occurs in all obese populations, as do poor dietary habits. Proton-pump inhibitors (PPIs), due to their wide availability, are most often used to treat GERD-related heartburn and other symptoms. Here, we review the evidence on how poor diet as well as short- and long-term use of PPIs adversely affect the gastrointestinal microbiota to cause dysbiosis. Key components of dysbiosis-induced metabolically unhealthy obesity (MUO) associated with PPI use include "leaky gut," systemic low-grade inflammation, and reduced amounts of short-chain fatty acids (SCFAs) such as butyrate that promote metabolic health. The benefit of using probiotics to mitigate PPI-induced dysbiosis and MUO is also discussed.


Subject(s)
Gastroesophageal Reflux , Proton Pump Inhibitors , Adult , Humans , United States , Proton Pump Inhibitors/adverse effects , Dysbiosis/chemically induced , Dysbiosis/complications , Obesity/complications , Gastroesophageal Reflux/diagnosis , Inflammation
4.
Am J Health Syst Pharm ; 80(15): 974-983, 2023 07 21.
Article in English | MEDLINE | ID: mdl-37137335

ABSTRACT

PURPOSE: Pharmacists oversee parenteral drug preparation and administration in hospitals, clinics, infusion centers, and home infusion settings. Infusion-related phlebitis (IRP), the most common complication of intravenous infusion therapy, significantly impacts therapeutic outcomes, patient satisfaction, cost of care, and provider workload. Here we review the major etiologies of IRP and describe potential pharmacological and nonpharmacological interventions for preventing and managing the condition as well as for improving vascular access health in multiple-drug administration settings. SUMMARY: Many parenterally administered drugs cause phlebitis due to mechanical, chemical, or infectious etiologies. Pharmacists can recommend nonpharmacological strategies to mitigate phlebitis, including -judicious device selection and placement; adjustment of the drug concentration, flow rate, or formulation; infusion site rotation; and use of inline filters to minimize contaminant particulates. Pharmacological treatments for phlebitis include topical, local, and systemic anti-inflammatory and analgesic agents that can reduce symptom severity and prevent further treatment complications or delays. CONCLUSION: Pharmacists can contribute a unique perspective to interprofessional teams tasked with making policy and formulary decisions that minimize the negative impacts of IRP on drug delivery and patient outcomes.


Subject(s)
Pharmacists , Phlebitis , Humans , Infusions, Parenteral , Phlebitis/chemically induced , Phlebitis/prevention & control , Infusions, Intravenous , Administration, Intravenous
5.
Crit Care Nurs Clin North Am ; 31(1): 15-30, 2019 Mar.
Article in English | MEDLINE | ID: mdl-30736932

ABSTRACT

This update presents evidence for new antiplatelet therapies including modified P2Y12 inhibitors and a new class of thromboxane antagonists. Discussed are emerging data on established antihyperlipidemic medications that support an additional antiplatelet effect. Current information about the effectiveness of several bleeding reversal agents is discussed, and the concept of personalized antiplatelet therapy, wherein selection of an antiplatelet therapy is based on genetic factors or laboratory testing that predict response to therapy and risk of adverse effects. Finally, future drug targets are introduced and drug interactions that can be leveraged to design more effective and safe antiplatelet therapies are described.


Subject(s)
Cardiovascular Diseases/drug therapy , Platelet Aggregation Inhibitors/therapeutic use , Risk Reduction Behavior , Critical Care Nursing , Humans , Platelet Aggregation Inhibitors/pharmacokinetics
6.
Am J Physiol Regul Integr Comp Physiol ; 315(4): R595-R608, 2018 10 01.
Article in English | MEDLINE | ID: mdl-29949410

ABSTRACT

Long-acting glucagon-like peptide-1 (GLP-1) receptor (GLP-1R) agonists (GLP-1RA), such as exendin-4 (Ex4), promote weight loss. On the basis of a newly discovered interaction between GLP-1 and oleoylethanolamide (OEA), we tested whether OEA enhances GLP-1RA-mediated anorectic signaling and weight loss. We analyzed the effect of GLP-1+OEA and Ex4+OEA on canonical GLP-1R signaling and other proteins/pathways that contribute to the hypophagic action of GLP-1RA (AMPK, Akt, mTOR, and glycolysis). We demonstrate that OEA enhances canonical GLP-1R signaling when combined with GLP-1 but not with Ex4. GLP-1 and Ex4 promote phosphorylation of mTOR pathway components, but OEA does not enhance this effect. OEA synergistically enhanced GLP-1- and Ex4-stimulated glycolysis but did not augment the hypophagic action of GLP-1 or Ex4 in lean or diet-induced obese (DIO) mice. However, the combination of Ex4+OEA promoted greater weight loss in DIO mice than Ex4 or OEA alone during a 7-day treatment. This was due in part to transient hypophagia and increased energy expenditure, phenotypes also observed in Ex4-treated DIO mice. Thus, OEA augments specific GLP-1RA-stimulated signaling but appears to work in parallel with Ex4 to promote weight loss in DIO mice. Elucidating cooperative mechanisms underlying Ex4+OEA-mediated weight loss could, therefore, be leveraged toward more effective obesity therapies.


Subject(s)
Anti-Obesity Agents/pharmacology , Endocannabinoids/pharmacology , Exenatide/pharmacology , Glucagon-Like Peptide-1 Receptor/agonists , Incretins/pharmacology , Obesity/drug therapy , Oleic Acids/pharmacology , Weight Loss/drug effects , AMP-Activated Protein Kinases/metabolism , Animals , CHO Cells , Cricetulus , Diet, High-Fat , Disease Models, Animal , Drug Therapy, Combination , Feeding Behavior/drug effects , Glucagon-Like Peptide-1 Receptor/metabolism , Glycolysis/drug effects , Male , Mice, Inbred C57BL , Obesity/metabolism , Obesity/physiopathology , Obesity/psychology , Phosphorylation , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/drug effects , TOR Serine-Threonine Kinases/metabolism
7.
Am J Physiol Endocrinol Metab ; 313(6): E651-E662, 2017 12 01.
Article in English | MEDLINE | ID: mdl-28811293

ABSTRACT

Pharmacological activation of the glucagon-like peptide-1 receptor (GLP-1R) in the ventromedial hypothalamus (VMH) reduces food intake. Here, we assessed whether suppression of food intake by GLP-1R agonists (GLP-1RA) in this region is dependent on AMP-activated protein kinase (AMPK) and mammalian target of rapamycin (mTOR). We found that pharmacological inhibition of glycolysis, and thus activation of AMPK, in the VMH attenuates the anorectic effect of the GLP-1R agonist exendin-4 (Ex4), indicating that glucose metabolism and inhibition of AMPK are both required for this effect. Furthermore, we found that Ex4-mediated anorexia in the VMH involved mTOR but not acetyl-CoA carboxylase, two downstream targets of AMPK. We support this by showing that Ex4 activates mTOR signaling in the VMH and Chinese hamster ovary (CHO)-K1 cells. In contrast to the clear acute pharmacological impact of the these receptors on food intake, knockdown of the VMH Glp1r conferred no changes in energy balance in either chow- or high-fat-diet-fed mice, and the acute anorectic and glucose tolerance effects of peripherally dosed GLP-1RA were preserved. These results show that the VMH GLP-1R regulates food intake by engaging key nutrient sensors but is dispensable for the effects of GLP-1RA on nutrient homeostasis.


Subject(s)
Eating/physiology , Food , Glucagon-Like Peptide-1 Receptor/physiology , Sensation/physiology , Ventromedial Hypothalamic Nucleus/physiology , Acetyl-CoA Carboxylase/metabolism , Adenylate Kinase/metabolism , Animals , Body Composition/drug effects , CHO Cells , Cricetulus , Dose-Response Relationship, Drug , Eating/drug effects , Exenatide , Glucagon-Like Peptide-1 Receptor/agonists , Glucagon-Like Peptide-1 Receptor/metabolism , Glycolysis/drug effects , Homeostasis/physiology , Male , Mice , Mice, Inbred C57BL , Peptides/pharmacology , Sensation/drug effects , TOR Serine-Threonine Kinases/metabolism , Venoms/pharmacology , Ventromedial Hypothalamic Nucleus/metabolism
8.
Diabetes ; 66(2): 372-384, 2017 02.
Article in English | MEDLINE | ID: mdl-27908915

ABSTRACT

Pharmacological activation of the hypothalamic glucagon-like peptide 1 (GLP-1) receptor (GLP-1R) promotes weight loss and improves glucose tolerance. This demonstrates that the hypothalamic GLP-1R is sufficient but does not show whether it is necessary for the effects of exogenous GLP-1R agonists (GLP-1RA) or endogenous GLP-1 on these parameters. To address this, we crossed mice harboring floxed Glp1r alleles to mice expressing Nkx2.1-Cre to knock down Glp1r expression throughout the hypothalamus (GLP-1RKDΔNkx2.1cre). We also generated mice lacking Glp1r expression specifically in two GLP-1RA-responsive hypothalamic feeding nuclei/cell types, the paraventricular nucleus (GLP-1RKDΔSim1cre) and proopiomelanocortin neurons (GLP-1RKDΔPOMCcre). Chow-fed GLP-1RKDΔNkx2.1cre mice exhibited increased food intake and energy expenditure with no net effect on body weight. When fed a high-fat diet, these mice exhibited normal food intake but elevated energy expenditure, yielding reduced weight gain. None of these phenotypes were observed in GLP-1RKDΔSim1cre and GLP-1RKDΔPOMCcre mice. The acute anorectic and glucose tolerance effects of peripherally dosed GLP-1RA exendin-4 and liraglutide were preserved in all mouse lines. Chronic liraglutide treatment reduced body weight in chow-fed GLP-1RKDΔNkx2.1cre mice, but this effect was attenuated with high-fat diet feeding. In sum, classic homeostatic control regions are sufficient but not individually necessary for the effects of GLP-1RA on nutrient homeostasis.


Subject(s)
Eating/genetics , Energy Metabolism/genetics , Glucagon-Like Peptide-1 Receptor/genetics , Glucose/metabolism , Hypothalamus/metabolism , Animals , Body Composition , Diet, High-Fat , Eating/drug effects , Exenatide , Gene Knockdown Techniques , Glucagon-Like Peptide 1/pharmacology , Glucagon-Like Peptide-1 Receptor/metabolism , Glucose Tolerance Test , Homeostasis/genetics , Incretins/pharmacology , Liraglutide/pharmacology , Male , Mice , Neurons/metabolism , Paraventricular Hypothalamic Nucleus/metabolism , Peptides/pharmacology , Pro-Opiomelanocortin/metabolism , Venoms/pharmacology , Weight Gain/drug effects , Weight Gain/genetics
9.
Cell Metab ; 25(1): 152-165, 2017 01 10.
Article in English | MEDLINE | ID: mdl-27839908

ABSTRACT

Pharmacological inhibition of the dipeptidyl peptidase-4 (DPP4) enzyme potentiates incretin action and is widely used to treat type 2 diabetes. Nevertheless, the precise cells and tissues critical for incretin degradation and glucose homeostasis remain unknown. Here, we use mouse genetics and pharmacologic DPP4 inhibition to identify DPP4+ cell types essential for incretin action. Although enterocyte DPP4 accounted for substantial intestinal DPP4 activity, ablation of enterocyte DPP4 in Dpp4Gut-/- mice did not produce alterations in plasma DPP4 activity, incretin hormone levels, and glucose tolerance. In contrast, endothelial cell (EC)-derived DPP4 contributed substantially to levels of soluble plasma DPP4 activity, incretin degradation, and glucose control. Surprisingly, DPP4+ cells of bone marrow origin mediated the selective degradation of fasting GIP, but not GLP-1. Collectively, these findings identify distinct roles for DPP4 in the EC versus the bone marrow compartment for selective incretin degradation and DPP4i-mediated glucoregulation.


Subject(s)
Dipeptidyl Peptidase 4/metabolism , Gastric Inhibitory Polypeptide/metabolism , Glucagon-Like Peptide 1/metabolism , Glucose/metabolism , Homeostasis , Incretins/metabolism , Animals , Bone Marrow Transplantation , Diet, High-Fat , Dipeptidyl Peptidase 4/blood , Enteral Nutrition , Feeding Behavior/drug effects , Glucose Tolerance Test , Homeostasis/drug effects , Insulin Resistance , Intestinal Mucosa/drug effects , Intestinal Mucosa/metabolism , Intestines/drug effects , Intestines/enzymology , Male , Mice , Models, Biological , Sitagliptin Phosphate/pharmacology
10.
J Mol Cell Cardiol ; 76: 172-6, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25200599

ABSTRACT

The incretin hormone glucagon-like peptide-1 (Glp1) is cardioprotective in models of ischemia-reperfusion injury, myocardial infarction and gluco/lipotoxicity. Inflammation is a factor in these models, yet it is unknown whether Glp1 receptor (Glp1r) agonists are protective against cardiac inflammation. We tested the hypothesis that the Glp1r agonist Exendin-4 (Ex4) is cardioprotective in mice with cardiac-specific monocyte chemoattractant protein-1 overexpression. These MHC-MCP1 mice exhibit increased cardiac monocyte infiltration, endoplasmic reticulum (ER) stress, apoptosis, fibrosis and left ventricular dysfunction. Ex4 treatment for 8 weeks improved cardiac function and reduced monocyte infiltration, fibrosis and apoptosis in MHC-MCP1 mice. Ex4 enhanced expression of the ER chaperone glucose-regulated protein-78 (GRP78), decreased expression of the pro-apoptotic ER stress marker CCAAT/-enhancer-binding protein homologous protein (CHOP) and increased expression of the ER calcium regulator Sarco/Endoplasmic Reticulum Calcium ATPase-2a (SERCA2a). These findings suggest that the Glp1r is a viable target for treating cardiomyopathies associated with stimulation of pro-inflammatory factors.


Subject(s)
Cardiotonic Agents/pharmacology , Chemokine CCL2/metabolism , Myocytes, Cardiac/metabolism , Peptides/pharmacology , Venoms/pharmacology , Ventricular Dysfunction/drug therapy , Animals , Cells, Cultured , Chemokine CCL2/genetics , Drug Evaluation, Preclinical , Endoplasmic Reticulum Chaperone BiP , Exenatide , Gene Expression , Glucagon-Like Peptide-1 Receptor , Hypertrophy, Left Ventricular/drug therapy , Hypertrophy, Left Ventricular/metabolism , Hypertrophy, Left Ventricular/physiopathology , Inflammation Mediators/metabolism , Male , Mice, Transgenic , Receptors, Glucagon/agonists , Stroke Volume , Ventricular Dysfunction/metabolism , Ventricular Dysfunction/physiopathology
11.
Am J Physiol Endocrinol Metab ; 304(7): E677-85, 2013 Apr 01.
Article in English | MEDLINE | ID: mdl-23341495

ABSTRACT

Glucagon-like peptide-1 (GLP-1) suppresses food intake via activation of a central (i.e., brain) GLP-1 receptor (GLP-1R). Central AMP-activated protein kinase (AMPK) is a nutrient-sensitive regulator of food intake that is inhibited by anorectic signals. The anorectic effect elicited by hindbrain GLP-1R activation is attenuated by the AMPK stimulator AICAR. This suggests that central GLP-1R activation suppresses food intake via inhibition of central AMPK. The present studies examined the mechanism(s) by which central GLP-1R activation inhibits AMPK. Supporting previous findings, AICAR attenuated the anorectic effect elicited by intracerebroventricular (icv) administration of the GLP-1R agonist exendin-4 (Ex-4). We demonstrate that Ex-4 stimulates glycolysis and suppresses AMPK phosphorylation in a glucose-dependent manner in hypothalamic GT1-7 cells. This suggests that inhibition of AMPK and food intake by Ex-4 requires central glucose metabolism. Supporting this, the glycolytic inhibitor 2-deoxyglucose (2-DG) attenuated the anorectic effect of Ex-4. However, icv glucose did not enhance the suppression of food intake by Ex-4. AICAR had no effect on Ex-4-mediated reduction in locomotor activity. We also tested whether other carbohydrates affect the anorectic response to Ex-4. Intracerebroventricular pretreatment with the sucrose metabolite fructose, an AMPK activator, attenuated the anorectic effect of Ex-4. This potentially explains the increased food intake observed in sucrose-fed mice. In summary, we propose a model whereby activation of the central GLP-1R reduces food intake via glucose metabolism-dependent inhibition of central AMPK. We also suggest that fructose stimulates food intake by impairing central GLP-1R action. This has significant implications given the correlation between sugar consumption and obesity.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Anorexia/metabolism , Appetite Regulation/physiology , Fructose/metabolism , Glucose/metabolism , Hypothalamus/metabolism , Receptors, Glucagon/metabolism , AMP-Activated Protein Kinases/antagonists & inhibitors , AMP-Activated Protein Kinases/drug effects , Aminoimidazole Carboxamide/analogs & derivatives , Aminoimidazole Carboxamide/pharmacology , Animals , Antimetabolites/pharmacology , Appetite Regulation/drug effects , Cell Line , Deoxyglucose/pharmacology , Drinking/drug effects , Drinking/physiology , Eating/drug effects , Eating/physiology , Energy Metabolism/drug effects , Energy Metabolism/physiology , Exenatide , Fructose/administration & dosage , Glucagon-Like Peptide-1 Receptor , Hypoglycemic Agents/pharmacology , Hypothalamus/drug effects , Male , Mice , Mice, Knockout , Motor Activity/drug effects , Motor Activity/physiology , Neurons/cytology , Neurons/drug effects , Neurons/metabolism , Peptides/pharmacology , Receptors, Glucagon/drug effects , Receptors, Glucagon/genetics , Ribonucleotides/pharmacology , Signal Transduction/drug effects , Signal Transduction/physiology , Venoms/pharmacology
12.
Am J Physiol Cell Physiol ; 304(6): C508-18, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23302777

ABSTRACT

Hyperglycemia-induced cardiomyocyte apoptosis contributes to diabetic cardiomyopathy. Glucagon-like peptide-1 (Glp1) receptor (Glp1r) agonists improve cardiac function and survival in response to ischemia-reperfusion and myocardial infarction. The present studies assessed whether Glp1r activation exerts direct cardioprotective effects in response to hyperglycemia. Treatment with the Glp1r agonist Exendin-4 attenuated apoptosis in neonatal rat ventricular cardiomyocytes cultured in high (33 mM) glucose. This protective effect was mimicked by the cAMP inducer forskolin. The Exendin-4 protective effect was blocked by the Glp1r antagonist Exendin(9-39) or the PKA antagonist H-89. Exendin-4 also protected cardiomyocytes from hydrogen peroxide (H2O2)-induced cell death. Cardiomyocyte protection by Exendin-4 was not due to reduced reactive oxygen species levels. Instead, Exendin-4 treatment reduced endoplasmic reticulum (ER) stress, demonstrated by decreased expression of glucose-regulated protein-78 (GRP78) and CCAT/enhancer-binding homologous protein (CHOP). Reduced ER stress was not due to activation of the unfolded protein response, indicating that Exendin-4 directly prevents ER stress. Exendin-4 treatment selectively protected cardiomyocytes from thapsigargin- but not tunicamycin-induced death. This suggests that Exendin-4 attenuates thapsigargin-mediated inhibition of the sarco/endoplasmic reticulum Ca(2+) ATPase-2a (SERCA2a). High glucose attenuates SERCA2a function by reducing SERCA2a mRNA and protein levels, but Exendin-4 treatment prevented this reduction. Exendin-4 treatment also enhanced phosphorylation of the SERCA2a regulator phospholamban (PLN), which would be expected to stimulate SERCA2a activity. In sum, Glp1r activation attenuates high glucose-induced cardiomyocyte apoptosis in association with decreased ER stress and markers of enhanced SERCA2a activity. These findings identify a novel mechanism whereby Glp1-based therapies could be used as treatments for diabetic cardiomyopathy.


Subject(s)
Apoptosis/drug effects , Endoplasmic Reticulum Stress/drug effects , Myocytes, Cardiac/physiology , Peptides/pharmacology , Receptors, Glucagon/metabolism , Venoms/pharmacology , Animals , Calcium-Binding Proteins/metabolism , Cells, Cultured , Colforsin/pharmacology , Diabetic Cardiomyopathies/metabolism , Enzyme Activation , Exenatide , Glucagon-Like Peptide-1 Receptor , Glucose/metabolism , HSP70 Heat-Shock Proteins , Heat-Shock Proteins/metabolism , Hydrogen Peroxide/pharmacology , Hyperglycemia/metabolism , Hypoglycemic Agents/pharmacology , Isoquinolines/pharmacology , Membrane Proteins , Oxidative Stress/drug effects , Peptide Fragments/pharmacology , Phosphorylation/drug effects , Protein Kinase Inhibitors/pharmacology , RNA, Messenger/biosynthesis , Rats , Receptors, Glucagon/agonists , Receptors, Glucagon/antagonists & inhibitors , Sarcoplasmic Reticulum Calcium-Transporting ATPases/antagonists & inhibitors , Sarcoplasmic Reticulum Calcium-Transporting ATPases/genetics , Sarcoplasmic Reticulum Calcium-Transporting ATPases/metabolism , Sulfonamides/pharmacology , Thapsigargin/pharmacology , Transcription Factor CHOP/biosynthesis , Transcription Factor CHOP/metabolism , Tunicamycin/pharmacology , Unfolded Protein Response
13.
Hypertension ; 59(4): 869-76, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22371360

ABSTRACT

Cyclooxygenase (COX)-derived prostanoids have long been implicated in blood pressure (BP) regulation. Recently prostaglandin E(2) (PGE(2)) and its receptor EP(1) (EP(1)R) have emerged as key players in angiotensin II (Ang II)-dependent hypertension (HTN) and related end-organ damage. However, the enzymatic source of PGE(2,) that is, COX-1 or COX-2, and its site(s) of action are not known. The subfornical organ (SFO) is a key forebrain region that mediates systemic Ang II-dependent HTN via reactive oxygen species (ROS). We tested the hypothesis that cross-talk between PGE(2)/EP(1)R and ROS signaling in the SFO is required for Ang II HTN. Radiotelemetric assessment of blood pressure revealed that HTN induced by infusion of systemic "slow-pressor" doses of Ang II was abolished in mice with null mutations in EP(1)R or COX-1 but not COX-2. Slow-pressor Ang II-evoked HTN and ROS formation in the SFO were prevented when the EP(1)R antagonist SC-51089 was infused directly into brains of wild-type mice, and Ang-II-induced ROS production was blunted in cells dissociated from SFO of EP(1)R(-/-) and COX-1(-/-) but not COX-2(-/-) mice. In addition, slow-pressor Ang II infusion caused a ≈3-fold increase in PGE(2) levels in the SFO but not in other brain regions. Finally, genetic reconstitution of EP(1)R selectively in the SFO of EP(1)R-null mice was sufficient to rescue slow-pressor Ang II-elicited HTN and ROS formation in the SFO of this model. Thus, COX 1-derived PGE(2) signaling through EP(1)R in the SFO is required for the ROS-mediated HTN induced by systemic infusion of Ang II and suggests that EP(1)R in the SFO may provide a novel target for antihypertensive therapy.


Subject(s)
Angiotensin II/physiology , Cyclooxygenase 1/physiology , Dinoprostone/physiology , Hypertension/physiopathology , Receptors, Prostaglandin E, EP1 Subtype/physiology , Signal Transduction/physiology , Subfornical Organ/physiology , Angiotensin II/adverse effects , Angiotensin II/pharmacology , Animals , Cyclooxygenase 1/deficiency , Cyclooxygenase 1/genetics , Cyclooxygenase 2/deficiency , Cyclooxygenase 2/genetics , Cyclooxygenase 2/physiology , Disease Models, Animal , Hydrazines/pharmacology , Hypertension/prevention & control , Mice , Mice, Inbred C57BL , Mice, Knockout , Oxazepines/pharmacology , Reactive Oxygen Species/metabolism , Receptor Cross-Talk/physiology , Receptors, Prostaglandin E, EP1 Subtype/antagonists & inhibitors , Receptors, Prostaglandin E, EP1 Subtype/genetics , Signal Transduction/drug effects
14.
Am J Physiol Endocrinol Metab ; 302(3): E334-43, 2012 Feb 01.
Article in English | MEDLINE | ID: mdl-22094469

ABSTRACT

Glucagon-like peptide-1 (GLP-1) receptor knockout (Glp1r(-/-)) mice exhibit impaired hepatic insulin action. High fat (HF)-fed Glp1r(-/-) mice exhibit improved, rather than the expected impaired, hepatic insulin action. This is due to decreased lipogenic gene expression and triglyceride accumulation. The present studies overcome these secondary adaptations by acutely modulating GLP-1R action in HF-fed wild-type mice. The central GLP-1R was targeted given its role as a regulator of hepatic insulin action. We hypothesized that acute inhibition of the central GLP-1R impairs hepatic insulin action beyond the effects of HF feeding. We further hypothesized that activation of the central GLP-1R improves hepatic insulin action in HF-fed mice. Insulin action was assessed in conscious, unrestrained mice using the hyperinsulinemic euglycemic clamp. Mice received intracerebroventricular (icv) infusions of artificial cerebrospinal fluid, GLP-1, or the GLP-1R antagonist exendin-9 (Ex-9) during the clamp. Intracerebroventricular Ex-9 impaired the suppression of hepatic glucose production by insulin, whereas icv GLP-1 improved it. Neither treatment affected tissue glucose uptake. Intracerebroventricular GLP-1 enhanced activation of hepatic Akt and suppressed hypothalamic AMP-activated protein kinase. Central GLP-1R activation resulted in lower hepatic triglyceride levels but did not affect muscle, white adipose tissue, or plasma triglyceride levels during hyperinsulinemia. In response to oral but not intravenous glucose challenges, activation of the central GLP-1R improved glucose tolerance. This was associated with higher insulin levels. Inhibition of the central GLP-1R had no effect on oral or intravenous glucose tolerance. These results show that inhibition of the central GLP-1R deteriorates hepatic insulin action in HF-fed mice but does not affect whole body glucose homeostasis. Contrasting this, activation of the central GLP-1R improves glucose homeostasis in HF-fed mice by increasing insulin levels and enhancing hepatic insulin action.


Subject(s)
Glucagon-Like Peptide 1/metabolism , Hypothalamus/metabolism , Insulin Resistance , Insulin/metabolism , Liver/metabolism , Pancreas/metabolism , Receptors, Glucagon/metabolism , AMP-Activated Protein Kinases/metabolism , Animals , Diet, High-Fat/adverse effects , Glucagon-Like Peptide 1/administration & dosage , Glucagon-Like Peptide-1 Receptor , Glucose Clamp Technique , Glycogenolysis/drug effects , Hypothalamus/drug effects , Hypothalamus/enzymology , Infusions, Intraventricular , Insulin/blood , Insulin Secretion , Lipid Metabolism/drug effects , Liver/drug effects , Liver/enzymology , Male , Mice , Mice, Inbred C57BL , Nerve Tissue Proteins/agonists , Nerve Tissue Proteins/antagonists & inhibitors , Nerve Tissue Proteins/metabolism , Neurons/drug effects , Neurons/enzymology , Neurons/metabolism , Organ Specificity , Pancreas/drug effects , Proto-Oncogene Proteins c-akt/metabolism , Receptors, Glucagon/agonists , Receptors, Glucagon/antagonists & inhibitors , Signal Transduction/drug effects
15.
Kidney Int ; 79(3): 331-9, 2011 Feb.
Article in English | MEDLINE | ID: mdl-20944547

ABSTRACT

Preeclampsia is a major cause of maternal and neonatal morbidity and mortality. In mouse models, complement activation in the placenta is associated with abnormal placental development and miscarriage, and inhibiting complement prevents fetal injury. We mated two mouse strains, DBA/2 and CBA/J, expecting that the pregnancies might show features of preeclampsia and of immunologically mediated pregnancy loss. Along with placental dysfunction, these matings resulted in proteinuria, elevated BUN, fibrin deposition, and glomerular endotheliosis. We blocked placental complement activation throughout pregnancy by administering a single dose of the C3 inhibitor CR2-Crry given on day 5 of the pregnancy. This procedure specifically targets the sites of complement activation without inducing any systemic effects. Placental complement inhibition prevented oxidative stress and placental dysfunction, as well as proteinuria and renal pathologic features of preeclampsia. Thus, local blockade of complement activation at the maternal-fetal interface rescues preeclampsia in mice, and identifies new treatments. Hence, complement triggers a feed-forward cycle of placental damage, antiangiogenic factor production, and maternal vascular damage in patients.


Subject(s)
Complement Activation/drug effects , Kidney/drug effects , Placenta/drug effects , Pre-Eclampsia/drug therapy , Recombinant Fusion Proteins/administration & dosage , Animals , Blood Urea Nitrogen , Disease Models, Animal , Female , Fibrin/metabolism , Injections, Intravenous , Kidney/immunology , Kidney/metabolism , Kidney/pathology , Kidney/physiopathology , Mice , Mice, Inbred BALB C , Mice, Inbred CBA , Mice, Inbred DBA , Neovascularization, Physiologic/drug effects , Oxidative Stress/drug effects , Placenta/immunology , Placenta/metabolism , Placenta/physiopathology , Pre-Eclampsia/immunology , Pre-Eclampsia/metabolism , Pre-Eclampsia/physiopathology , Pregnancy , Proteinuria/immunology , Proteinuria/prevention & control , Vascular Endothelial Growth Factor A/administration & dosage , Vascular Endothelial Growth Factor Receptor-1/metabolism
16.
Hypertension ; 57(2): 289-97, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21173341

ABSTRACT

Renovascular hypertension in mice is characterized by an elevation in hypothalamic angiotensin II levels. The paraventricular nucleus (PVN) is a major cardioregulatory site implicated in the neurogenic component of renovascular hypertension. Increased superoxide (O(2)(-·)) production in the PVN is involved in angiotensin II-dependent neurocardiovascular diseases such as hypertension and heart failure. Here, we tested the hypothesis that excessive O(2)(-·) production and activation of the redox-regulated transcription factor activator protein-1 (AP-1) in PVN contributes to the development and maintenance of renovascular hypertension. Male C57BL/6 mice underwent implantation of radiotelemeters, bilateral PVN injections of an adenovirus (Ad) encoding superoxide dismutase (AdCuZnSOD) or a control gene (LacZ), and unilateral renal artery clipping (2-kidney, one-clip [2K1C]) or sham surgery. AP-1 activity was longitudinally monitored in vivo by bioluminescence imaging in 2K1C or sham mice that had undergone PVN-targeted microinjections of an Ad encoding the firefly luciferase (Luc) gene downstream of AP-1 response elements (AdAP-1Luc). 2K1C evoked chronic hypertension and an increase in O(2)(-·) production in the PVN. Viral delivery of CuZnSOD to the PVN not only prevented the elevation in O(2)(-·) but also abolished renovascular hypertension. 2K1C also caused a surge in AP-1 activity in the PVN, which paralleled the rise in O(2)(-·) production in this brain region, and this was prevented by treatment with AdCuZnSOD. Finally, Ad-mediated expression of a dominant-negative inhibitor of AP-1 activity in the PVN prevented 2K1C-evoked hypertension. These results implicate oxidant signaling and AP-1 transcriptional activity in the PVN as key mediators in the pathogenesis of renovascular hypertension.


Subject(s)
Hypertension, Renovascular/metabolism , Luminescent Measurements/methods , Paraventricular Hypothalamic Nucleus/metabolism , Transcription Factor AP-1/metabolism , Adenoviridae/genetics , Animals , Atrophy , Blood Pressure , Heart Rate , Hypertension, Renovascular/physiopathology , Immunohistochemistry , Kidney/metabolism , Kidney/pathology , Luciferases/genetics , Luciferases/metabolism , Male , Mice , Mice, Inbred C57BL , Organ Size , Oxidation-Reduction , Reactive Oxygen Species/metabolism , Superoxide Dismutase/genetics , Superoxide Dismutase/metabolism , Superoxides/metabolism , Transcription Factor AP-1/genetics
17.
Endocrinology ; 151(10): 4678-87, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20685876

ABSTRACT

Glucagon-like peptide-1 augments nutrient-stimulated insulin secretion. Chow-fed mice lacking the glucagon-like peptide-1 receptor (Glp1r) exhibit enhanced insulin-stimulated muscle glucose uptake but impaired suppression of endogenous glucose appearance (endoRa). This proposes a novel role for the Glp1r to regulate the balance of glucose disposal in muscle and liver by modulating insulin action. Whether this is maintained in an insulin-resistant state is unknown. The present studies tested the hypothesis that disruption of Glp1r expression overcomes high-fat (HF) diet-induced muscle insulin resistance and exacerbates HF diet-induced hepatic insulin resistance. Mice with a functional disruption of the Glp1r (Glp1r-/-) were compared with wild-type littermates (Glp1r+/+) after 12 wk on a regular chow diet or a HF diet. Arterial and venous catheters were implanted for sampling and infusions. Hyperinsulinemic-euglycemic clamps were performed on weight-matched male mice. [3-(3)H]glucose was used to determine glucose turnover, and 2[14C]deoxyglucose was used to measure the glucose metabolic index, an indicator of glucose uptake. Glp1r-/- mice exhibited increased glucose disappearance and muscle glucose metabolic index on either diet. This was associated with enhanced activation of muscle Akt and AMP-activated protein kinase and reduced muscle triglycerides in HF-fed Glp1r-/- mice. Chow-fed Glp1r-/- mice exhibited impaired suppression of endoRa and hepatic insulin signaling. In contrast, HF-fed Glp1r-/- mice exhibited improved suppression of endoRa and hepatic Akt activation. This was associated with decreased hepatic triglycerides and impaired activation of sterol regulatory element-binding protein-1. These results show that mice lacking the Glp1r are protected from HF diet-induced muscle and hepatic insulin resistance independent of effects on total fat mass.


Subject(s)
Diet, Atherogenic , Dietary Fats/adverse effects , Insulin Resistance/genetics , Receptors, Glucagon/genetics , Adiposity/drug effects , Adiposity/genetics , Adiposity/physiology , Animals , Female , Glucagon-Like Peptide-1 Receptor , Glucose/metabolism , Insulin/pharmacology , Liver/drug effects , Liver/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Muscle, Skeletal/drug effects , Muscle, Skeletal/metabolism , Sex Characteristics
18.
Circ Res ; 106(11): 1763-74, 2010 Jun 11.
Article in English | MEDLINE | ID: mdl-20413786

ABSTRACT

RATIONALE: Myocardial infarction (MI)-induced heart failure is characterized by central nervous system-driven sympathoexcitation and deteriorating cardiac function. The paraventricular nucleus (PVN) of the hypothalamus is a key regulator of sympathetic nerve activity and is implicated in heart failure. Redox signaling in the PVN and other central nervous system sites is a primary mechanism of neuro-cardiovascular regulation, and excessive oxidant production by activation of NADPH oxidases (Noxs) is implicated in some neuro-cardiovascular diseases. OBJECTIVE: We tested the hypothesis that Nox-mediated redox signaling in the PVN contributes to MI-induced sympathoexcitation and cardiac dysfunction in mice. METHODS AND RESULTS: Real-time PCR revealed that Nox4 was the most abundantly expressed Nox in PVN under basal conditions. Coronary arterial ligation (MI) caused a selective upregulation of this homolog compared to Nox1 and Nox2. Adenoviral gene transfer of Nox4 (AdsiNox4) to PVN (bilateral) attenuated MI-induced superoxide formation in this brain region (day 14) to the same level as that produced by PVN-targeted gene transfer of cytoplasmic superoxide dismutase (AdCu/ZnSOD). MI mice treated with AdsiNox4 or AdCu/ZnSOD in the PVN showed marked improvement in cardiac function as assessed by echocardiography and left ventricular hemodynamic analysis. This was accompanied by significantly diminished sympathetic outflow and apoptosis in the periinfarct region of the heart. CONCLUSIONS: These results suggest that MI causes dysregulation of Nox4-mediated redox signaling in the PVN, which leads to sympathetic overactivation and a decline in cardiac function. Targeted inhibition of oxidant signaling in the PVN could provide a novel treatment for MI-induced heart failure.


Subject(s)
Apoptosis , Gene Silencing , Heart Failure/enzymology , Heart/innervation , Myocardial Infarction/enzymology , Myocardium/pathology , NADPH Oxidases/metabolism , Paraventricular Hypothalamic Nucleus/enzymology , Sympathetic Nervous System/physiopathology , Adenoviridae/genetics , Animals , Catalase/genetics , Catalase/metabolism , Disease Models, Animal , Down-Regulation , Ganglionic Blockers/pharmacology , Gene Transfer Techniques , Genetic Therapy/methods , Genetic Vectors , Heart Failure/genetics , Heart Failure/pathology , Heart Failure/physiopathology , Heart Failure/prevention & control , Hemodynamics , Hydrogen Peroxide/metabolism , Mice , Mice, Inbred C57BL , Myocardial Infarction/genetics , Myocardial Infarction/pathology , Myocardial Infarction/physiopathology , Myocardial Infarction/therapy , NADPH Oxidase 4 , NADPH Oxidases/genetics , Norepinephrine/urine , Oxidation-Reduction , RNA Interference , Signal Transduction , Superoxide Dismutase/genetics , Superoxide Dismutase/metabolism , Superoxides/metabolism , Sympathetic Nervous System/drug effects , Sympathetic Nervous System/metabolism , Time Factors , Ventricular Function, Left
19.
Hypertension ; 54(5): 1106-14, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19805637

ABSTRACT

The renin-angiotensin system exerts a tremendous influence over fluid balance and arterial pressure. Angiotensin II (Ang-II), the effector peptide of the renin-angiotensin system, acts in the central nervous system to regulate neurohumoral outflow and thirst. Dysregulation of Ang-II signaling in the central nervous system is implicated in cardiovascular diseases; however, the mechanisms remain poorly understood. Recently we established that NADPH oxidase (Nox)-derived superoxide acting in the forebrain subfornical organ is critical in the physiological responses to central Ang-II. In addition, we have found that Nox2 and Nox4 are the most abundantly expressed Nox homologues within Ang-II-sensitive sites in the forebrain. To dissect out the functional importance and unique roles of these Nox enzymes in the pressor and dipsogenic effects of central Ang-II, we developed adenoviral vectors expressing small interfering RNA to selectively silence Nox2 or Nox4 expression in the subfornical organ. Our results demonstrate that both Nox2 and Nox4 are required for the full vasopressor effects of brain Ang-II but that only Nox2 is coupled to the Ang-II-induced water intake response. These studies establish the importance of both Nox2- and Nox4-containing NADPH oxidases in the actions of Ang-II in the central nervous system and are the first to reveal differential involvement of these Nox enzymes in the various physiological effects of central Ang-II.


Subject(s)
Angiotensin II/pharmacology , Brain Chemistry/genetics , NADPH Oxidases/metabolism , Renin-Angiotensin System/genetics , Adenoviridae/genetics , Animals , Disease Models, Animal , Drinking , Gene Expression Regulation , Genetic Vectors/genetics , Hypertension/physiopathology , Mice , Mice, Inbred C57BL , Myocytes, Cardiac/metabolism , NADPH Oxidases/genetics , Random Allocation , Rats , Rats, Sprague-Dawley , Reactive Oxygen Species/metabolism , Renin-Angiotensin System/drug effects , Reverse Transcriptase Polymerase Chain Reaction , Sensitivity and Specificity , Superoxides/metabolism
20.
Am J Physiol Regul Integr Comp Physiol ; 295(4): R1168-74, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18667715

ABSTRACT

Peripheral chemoreceptors located in the carotid bodies are the primary sensors of systemic hypoxia. Although the pattern of responses elicited by peripheral chemoreceptor activation is well established in rats, lambs, and rabbits, the cardiovascular responses to peripheral chemoreflex activation in conscious mice have not been delineated. Here we report that stimulation of peripheral chemoreceptors by potassium cyanide (KCN) in conscious mice elicits a unique biphasic response in blood pressure that is characterized by an initial and robust rise followed by a decrease in blood pressure, which is accompanied by a marked reduction in heart rate. The depressor and bradycardic responses to KCN were abolished by muscarinic receptor blockade with atropine, and the pressor response was abolished by alpha-adrenergic receptor blockade with prazosin, suggesting that vagal and sympathetic drive to the heart and sympathetic drive to the vasculature mediate these cardiovascular responses. These studies characterized the chemoreflex in conscious mice and established the reliability of using them for studying hypoxia-related diseases such as obstructive sleep apnea. In another series of experiments, two methods for analyzing baroreflex sensitivity were compared: the classical pharmacological approach using phenylephrine and sodium nitroprusside (i.e., the Oxford technique) or the sequence method for analyzing spontaneous baroreflex activity. Our findings indicate that both methods are reliable, and the sequence method certainly has its benefits as a predictive tool in the context of long-term noninvasive studies using telemetry. However, for absolute determination of baroreflex function, analysis of spontaneous baroreflex activity should be complemented by the classical pharmacological method.


Subject(s)
Baroreflex/physiology , Chemoreceptor Cells/physiology , Reflex/physiology , Telemetry/methods , Animals , Atropine/pharmacology , Baroreflex/drug effects , Blood Pressure/drug effects , Blood Pressure/physiology , Bradycardia/chemically induced , Cardiovascular Physiological Phenomena/drug effects , Consciousness , Heart Rate/drug effects , Heart Rate/physiology , Male , Mice , Mice, Inbred C57BL , Nitroprusside/pharmacology , Phenylephrine/pharmacology , Potassium Cyanide/pharmacology , Prazosin/pharmacology , Propranolol/pharmacology , Reflex/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...