Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
1.
PRiMER ; 7: 15, 2023.
Article in English | MEDLINE | ID: mdl-37465832

ABSTRACT

Objectives: This project analyzed the culture of safety quality improvement at the Family Medicine Center (FMC). The Agency for Healthcare Research and Quality (AHRQ) Culture of Safety Survey was used as a benchmark for internal and external comparison. Methods: The AHRQ Culture of Safety Survey was administered to health care staff in 2015, 2017, and 2019, respectively, at the Family Medicine Center. Baseline perceptions of safety and quality were established using the data from the AHRQ Culture of Safety Survey in 2015. We performed multiple large-scope quality improvement projects that focused on identified deficiencies. The changes in perception were monitored over time every 2 years. We analyzed the results using the Kruskal-Wallace test (P=.05). Results: The AHRQ Culture of Safety Survey showed statistically significant improvement in patient centeredness, effectiveness, timeliness, efficiency, equitableness, and overall patient safety from 2015 to 2019. Some inconsistencies were seen between different sections of responses, likely due to wording interpretations by the participants. Conclusion: Overall, the AHRQ Culture of Safety Survey is an effective way to help monitor employee perception of multiple domains that lead to a safe and effective clinical environment as compared to other practices across the country. Clinic-wide implementation of quality and patient care strategies resulted in significant improvements in nearly every category of the survey.

2.
JMIR Form Res ; 5(6): e28952, 2021 Jun 30.
Article in English | MEDLINE | ID: mdl-34255651

ABSTRACT

BACKGROUND: Screen ASSIST is a cessation trial offered to current smokers at the point of lung cancer screening. Because of the unique position of promoting a prevention behavior (smoking cessation) within the context of a detection behavior (lung cancer screening), this study employed prospect theory to design and formatively evaluate a targeted recruitment video prior to trial launch. OBJECTIVE: The aim of this study was to identify which message frames were most effective at promoting intent to participate in a smoking cessation study. METHODS: Participants were recruited from a proprietary opt-in online panel company and randomized to a 2 (benefits of quitting vs risks of continuing to smoke at the time of lung screening; BvR) × 2 (gains of participating vs losses of not participating in a cessation study; GvL) message design experiment (N=314). The primary outcome was self-assessed intent to participate in a smoking cessation study. Message effectiveness and lung cancer risk perception measures were also collected. Analysis of variance examined the main effect of the 2 message factors and a least absolute shrinkage and selection operator (LASSO) approach identified predictors of intent to participate in a multivariable model. A mediation analysis was conducted to determine the direct and indirect effects of message factors on intent to participate in a cessation study. RESULTS: A total of 296 participants completed the intervention. There were no significant differences in intent to participate in a smoking cessation study between message frames (P=.12 and P=.61). In the multivariable model, quit importance (P<.001), perceived message relevance (P<.001), and affective risk response (ie, worry about developing lung cancer; P<.001) were significant predictors of intent to participate. The benefits of quitting frame significantly increased affective risk response (Meanbenefits 2.60 vs Meanrisk 2.40; P=.03), which mediated the relationship between message frame and intent to participate (b=0.24; 95% CI 0.01-0.47; P=.03). CONCLUSIONS: This study provides theoretical and practical guidance on how to design and evaluate proactive recruitment messages for a cessation trial. Based on our findings, we conclude that heavy smokers are more responsive to recruitment messages that frame the benefits of quitting as it increased affective risk response, which predicted greater intention to participate in a smoking cessation study.

3.
J Neurosci ; 27(14): 3703-11, 2007 Apr 04.
Article in English | MEDLINE | ID: mdl-17409234

ABSTRACT

Despite the introduction of highly active antiretroviral therapy, dementia caused by human immunodeficiency virus-1 (HIV-1) infection remains a devastating and common neurological disorder. Although the mechanisms governing neurodegeneration during HIV-1 infection remain uncertain, the HIV-1 accessory protein, viral protein R (Vpr), has been proposed as a neurotoxic protein. Herein, we report that Vpr protein and transcript were present in the brains of HIV-infected persons. Moreover, soluble Vpr caused neuronal apoptosis, involving cytochrome c extravasation, p53 induction, and activation of caspase-9 while exerting a depressive effect on whole-cell currents in neurons (p < 0.05), which was inhibited by iberiotoxin. Vpr-activated glial cells secreted neurotoxins in a concentration-dependent manner (p < 0.001). Transgenic (Tg) mice expressing Vpr in brain monocytoid cells displayed the transgene principally in the basal ganglia (p < 0.05) and cerebral cortex (p < 0.01) compared with hindbrain expression. Vpr was released from cultured transgenic macrophages, which was cytotoxic to neurons and was blocked by anti-Vpr antibody (p < 0.05). Neuronal injury was observed in Tg animals compared with wild-type littermates, chiefly affecting GAD65 (p < 0.01) and vesicular acetylcholine transferase (p < 0.001) immunopositive neuronal populations in the basal ganglia. There was also a loss of subcortical synaptophysin (p < 0.001) immunoreactivity as well as an increase in activated caspase-3, which was accompanied by a hyperexcitable neurobehavioral phenotype (p < 0.05). Thus, HIV-1 Vpr caused neuronal death through convergent pathogenic mechanisms with ensuing in vivo neurodegeneration, yielding new insights into the mechanisms by which HIV-1 injures the nervous system.


Subject(s)
Apoptosis/physiology , Gene Products, vpr/physiology , HIV-1/physiology , Nerve Degeneration/metabolism , Neurons/metabolism , Animals , Cell Line, Tumor , Gene Products, vpr/biosynthesis , HIV-1/metabolism , Humans , Mice , Mice, Transgenic , Nerve Degeneration/pathology , Neurons/pathology , Rats , Rats, Sprague-Dawley , vpr Gene Products, Human Immunodeficiency Virus
4.
Am J Physiol Regul Integr Comp Physiol ; 292(5): R1872-80, 2007 May.
Article in English | MEDLINE | ID: mdl-17289819

ABSTRACT

Neuropeptide FF (NPFF) and neuropeptide VF (NPVF) are octapeptides belonging to the RFamide family of peptides that have been implicated in a wide variety of physiological functions in the brain, including central autonomic and neuroendocrine regulation. The effects of these peptides are mediated via NPFF1 and NPFF2 receptors that are abundantly expressed in the rat brain, including the hypothalamic paraventricular nucleus (PVN), an autonomic nucleus critical for the secretion of neurohormones and the regulation of sympathetic outflow. In this study, we examined, using whole cell patch-clamp recordings in the brain slice, the effects of NPFF and NPVF on inhibitory GABAergic synaptic input to parvocellular PVN neurons. Under voltage-clamp conditions, NPFF and NPVF reversibly and in a concentration-dependent manner reduced the evoked bicuculline-sensitive inhibitory postsynaptic currents (IPSCs) in parvocellular PVN neurons by 25 and 31%, respectively. RF9, a potent and selective NPFF receptor antagonist, blocked NPFF-induced reduction of IPSCs. Recordings of miniature IPSCs in these neurons following NPFF and NPVF applications showed a reduction in frequency but not amplitude, indicating a presynaptic locus of action for these peptides. Under current-clamp conditions, NPVF and NPFF caused depolarization (6-9 mV) of neurons that persisted in the presence of TTX but was abolished in the presence of bicuculline. Collectively, these data provide evidence for a disinhibitory role of NPFF and NPVF in the hypothalamic PVN via an attenuation of GABAergic inhibitory input to parvocellular neurons of this nucleus and explain the central autonomic effects of NPFF.


Subject(s)
GABA Antagonists/pharmacology , Neuropeptides/pharmacology , Oligopeptides/pharmacology , Paraventricular Hypothalamic Nucleus/cytology , Paraventricular Hypothalamic Nucleus/metabolism , gamma-Aminobutyric Acid/metabolism , Animals , Bicuculline/pharmacology , Cells, Cultured , Electrophysiology , Male , Narcotic Antagonists/pharmacology , Neurons/drug effects , Neurons/metabolism , Rats , Rats, Sprague-Dawley
5.
Peptides ; 27(5): 973-9, 2006 May.
Article in English | MEDLINE | ID: mdl-16517015

ABSTRACT

Neuropeptide FF (NPFF) is an octapeptide belonging to an extended family of RF amide peptides that have been implicated in a wide variety of physiological functions in the brain. NPFF and its receptors are abundantly expressed in the rat brain and spinal cord including the hypothalamic paraventricular nucleus (PVN), an autonomic nucleus critical for the secretion of neurohormones and the regulation of sympathetic outflow. In this study, we sought to examine the effects of NPFF on GABAergic inhibitory synaptic input to magnocellular neurosecretory cells (MNCs) of the PVN, which secrete the neurohormones, vasopressin and oxytocin from their terminals in the neurohypophysis. Whole cell patch clamp recordings under voltage clamp conditions were performed from PVN MNCs in the brain slice. Bicuculline-sensitive inhibitory postsynaptic currents (IPSCs) were isolated in the presence of glutamate receptor blockers. In tetrodotoxin, NPFF (5 microM) caused an increase in frequency, but not amplitude of miniature inhibitory postsynaptic currents (mIPSCs) in MNCs indicating a presynaptic locus of action for this peptide. Intracerebroventricular application of NPFF resulted in an activation of GABAergic neurons located adjacent to the PVN as revealed by immunohistochemistry for Fos protein and in situ hybridization for glutamic acid decarboxylase (GAD67) mRNA. Based on these observations we conclude that NPFF facilitates inhibitory input to MNCs of the PVN via GABAergic interneurons located in immediate vicinity of the nucleus. These findings provide a cellular and anatomic basis for the NPFF-induced inhibition of vasopressin release has been reported consequent to hypovolemia and hyperosmolar stimulation.


Subject(s)
Neurosecretory Systems/metabolism , Oligopeptides/physiology , Paraventricular Hypothalamic Nucleus/cytology , Animals , Bicuculline/pharmacology , Excitatory Postsynaptic Potentials/drug effects , Glutamate Decarboxylase/biosynthesis , Injections, Intraventricular , Isoenzymes/biosynthesis , Kynurenic Acid/pharmacology , Male , Neurosecretory Systems/cytology , Oligopeptides/pharmacology , Paraventricular Hypothalamic Nucleus/physiology , Patch-Clamp Techniques , Proto-Oncogene Proteins c-fos/biosynthesis , Rats , Rats, Sprague-Dawley , Receptors, GABA-A/drug effects , Receptors, GABA-A/physiology , Tetrodotoxin/pharmacology
6.
J Neurosci ; 26(2): 585-96, 2006 Jan 11.
Article in English | MEDLINE | ID: mdl-16407557

ABSTRACT

The insulin-like growth factor-II/mannose-6-phosphate (IGF-II/M6P) receptor is a single-pass transmembrane glycoprotein that plays an important role in the intracellular trafficking of lysosomal enzymes and endocytosis-mediated degradation of IGF-II. However, its role in signal transduction after IGF-II binding remains unclear. In the present study, we report that IGF-II/M6P receptor in the rat brain is coupled to a G-protein and that its activation by Leu27IGF-II, an analog that binds rather selectively to the IGF-II/M6P receptor, potentiates endogenous acetylcholine release from the rat hippocampal formation. This effect is mediated by a pertussis toxin (PTX)-sensitive GTP-binding protein and is dependent on protein kinase Calpha (PKCalpha)-induced phosphorylation of downstream substrates, myristoylated alanine-rich C kinase substrate, and growth associated protein-43. Additionally, treatment with Leu27IGF-II causes a reduction in whole-cell currents and depolarization of cholinergic basal forebrain neurons. This effect, which is blocked by an antibody against the IGF-II/M6P receptor, is also sensitive to PTX and is mediated via activation of a PKC-dependent pathway. These results together revealed for the first time that the single transmembrane domain IGF-II/M6P receptor expressed in the brain is G-protein coupled and is involved in the regulation of central cholinergic function via the activation of specific intracellular signaling cascades.


Subject(s)
Acetylcholine/metabolism , Hippocampus/metabolism , Nerve Tissue Proteins/physiology , Neurons/metabolism , Protein Kinase C-alpha/physiology , Receptor, IGF Type 2/physiology , Receptors, G-Protein-Coupled/physiology , Signal Transduction/physiology , Amino Acid Substitution , Animals , Binding, Competitive , Cholera Toxin/pharmacology , Delayed Rectifier Potassium Channels/antagonists & inhibitors , Delayed Rectifier Potassium Channels/physiology , GAP-43 Protein/physiology , GTP-Binding Protein alpha Subunit, Gi2/physiology , Guanosine 5'-O-(3-Thiotriphosphate)/metabolism , Guanylyl Imidodiphosphate/metabolism , Insulin/metabolism , Insulin/pharmacology , Insulin-Like Growth Factor I/pharmacology , Insulin-Like Growth Factor II/analogs & derivatives , Insulin-Like Growth Factor II/genetics , Insulin-Like Growth Factor II/pharmacology , Intracellular Signaling Peptides and Proteins/physiology , Isoproterenol/pharmacology , Male , Membrane Proteins/physiology , Myristoylated Alanine-Rich C Kinase Substrate , Nerve Tissue Proteins/immunology , Neurons/drug effects , Patch-Clamp Techniques , Peptides/pharmacology , Pertussis Toxin/pharmacology , Potassium Channel Blockers/pharmacology , Potassium Channels, Calcium-Activated/antagonists & inhibitors , Potassium Channels, Calcium-Activated/physiology , Rats , Rats, Sprague-Dawley , Receptor, IGF Type 2/immunology , Reverse Transcriptase Polymerase Chain Reaction
7.
Brain Cell Biol ; 35(2-3): 173-86, 2006 Jun.
Article in English | MEDLINE | ID: mdl-17957482

ABSTRACT

Beta-amyloid, a 39-43 amino acid peptide, may exert its biological effects via neuronal nicotinic acetylcholine receptors. Using the ratiometric dye, fura-2, we examined the effect of soluble beta-amyloid(1-42) on the concentration of intracellular Ca(2+) ([Ca(2+)](i)) in acutely dissociated rat basal forebrain neurons. Focal applications of nicotine (0.5-20 mM), evoked dose-dependent increases in intracellular [Ca(2+)](i) that were mediated by the entry of extracellular Ca(2+) via nicotinic acetylcholine receptors, and the release of intracellular Ca(2+) from stores. With repeated nicotine challenges, the nicotinic responses were potentiated by 98 +/- 12% (P < 0.05) while beta-amyloid(1-42)(100 nM) was present for approximately 5 min. This potentiation became larger during the subsequent washout of beta-amyloid(1-42), which was associated with a gradual rise in baseline [Ca(2+)](i). Application of beta-amyloid(1-42)by itself did not alter [Ca(2+)](i), and beta-amyloid(1-42)also had no significant effect on the response to repeated KCl challenges. Therefore, beta-amyloid(1-42) caused neither gross disturbance of cellular Ca(2+) homeostasis nor enhancement of voltage-gated Ca(2+) channels. Interestingly, beta-amyloid(1-42) transiently potentiated the response to repeated caffeine challenges, which was also associated with a transient rise in baseline [Ca(2+)](i). beta-amyloid(1-42) potentiation of nicotine-evoked rises in [Ca(2+)](i) was reversed by the SERCA pump inhibitor, thapsigargin, and the mitochondrial Na(+)/Ca(2+) exchanger inhibitor, CGP-37157. These results suggest that the dysregulation of [Ca(2+)](i) by beta-amyloid(1-42) during multiple challenges with nicotine or caffeine involved the sensitization or overfilling of intracellular stores that are maintained by SERCA pump and Ca(2+) efflux from the mitochondria.


Subject(s)
Amyloid beta-Peptides/metabolism , Basal Nucleus of Meynert/metabolism , Calcium Signaling/physiology , Calcium/metabolism , Neurons/metabolism , Receptors, Nicotinic/metabolism , Acetylcholine/metabolism , Action Potentials/drug effects , Action Potentials/physiology , Alzheimer Disease/metabolism , Alzheimer Disease/physiopathology , Amyloid beta-Peptides/pharmacology , Animals , Basal Nucleus of Meynert/drug effects , Caffeine/pharmacology , Calcium Signaling/drug effects , Cells, Cultured , Dose-Response Relationship, Drug , Drug Synergism , Excitatory Postsynaptic Potentials/drug effects , Excitatory Postsynaptic Potentials/physiology , Fura-2 , Intracellular Fluid/drug effects , Intracellular Fluid/metabolism , Male , Mitochondria/drug effects , Mitochondria/metabolism , Neurons/drug effects , Peptide Fragments/metabolism , Peptide Fragments/pharmacology , Rats , Rats, Sprague-Dawley , Receptors, Nicotinic/drug effects , Sarcoplasmic Reticulum Calcium-Transporting ATPases/drug effects , Sarcoplasmic Reticulum Calcium-Transporting ATPases/metabolism
8.
Eur J Neurosci ; 21(10): 2649-59, 2005 May.
Article in English | MEDLINE | ID: mdl-15926913

ABSTRACT

The deposition of beta-amyloid protein (A beta), a 39-43 amino acid peptide, in the brain and a loss of cholinergic neurons in the basal forebrain are pathological hallmarks of Alzheimer's disease (AD). Seaweeds consumed in Asia contain Fucoidan, a sulfated polysaccharide. Fucoidan has been known to exhibit various biological actions, such as an anti-inflammatory and antioxidant action. In this study, using whole-cell patch clamp recordings we examined the effects of Fucoidan on A beta-induced whole-cell currents in acutely dissociated rat basal forebrain neurons. We further investigated whether Fucoidan is capable of blocking A beta neurotoxicity in primary neuronal cultures. In dissociated cells, bath application of A beta(25-35) (1 microM) caused a reduction of the whole-cell currents by 16%. Fucoidan, in a dose-dependent manner, blocks the A beta(25-35) reduction of whole-cell currents. Exposure of A beta(25-35) (20 microM) or A beta(1-42) (20 microM) to rat cholinergic basal forebrain cultures for 48 h resulted in 40-60% neuronal death, which was significantly decreased by pretreatment of cultures with Fucoidan (0.1-1.0 microM). Fucoidan also attenuated A beta-induced down-regulation of phosphorylated protein kinase C. A beta(1-42)-induced generation of reactive oxygen species was blocked by prior exposure of cultures to Fucoidan. Furthermore, A beta activation of caspases 9 and 3, which are signaling pathways implicated in apoptotic cell death, is blocked by pretreatment of cultures with Fucoidan. These results show that Fucoidan is able to block A beta-induced reduction in whole-cell currents in basal forebrain neurons and has neuroprotective effects against A beta-induced neurotoxicity in basal forebrain neuronal cultures.


Subject(s)
Amyloid beta-Peptides/antagonists & inhibitors , Amyloid beta-Peptides/toxicity , Neurons/physiology , Neurotoxins/antagonists & inhibitors , Polysaccharides/pharmacology , Prosencephalon/physiology , Alzheimer Disease , Animals , Embryo, Mammalian , Female , Gestational Age , Humans , Membrane Potentials/drug effects , Membrane Potentials/physiology , Neurons/drug effects , Patch-Clamp Techniques , Pregnancy , Prosencephalon/drug effects , Prosencephalon/embryology , Rats , Rats, Sprague-Dawley , Reactive Oxygen Species/metabolism
9.
J Neurophysiol ; 89(6): 2923-30, 2003 Jun.
Article in English | MEDLINE | ID: mdl-12611974

ABSTRACT

Human amylin (hAmylin), a 37-amino acid pancreatic peptide, and amyloid beta protein (A beta), a 39-43 amino acid peptide, abundantly deposited in the brains of Alzheimer's patients, induce neurotoxicity in hippocampal and cortical cultures. Although the mechanism of this neurotoxicity is unknown, both peptides are capable of modulating ion channel function that may result in a disruption of cellular homeostasis. In this study, we examined the effects of hAmylin on whole cell currents in chemically identified neurons from the rat basal forebrain and the interactions of hAmylin-induced responses with those of A beta. Whole cell patch-clamp recordings were performed on enzymatically dissociated neurons of the diagonal band of Broca (DBB), a cholinergic basal forebrain nucleus. Bath application of hAmylin (1 nM to 5 microM) resulted in a dose-dependent reduction in whole cell currents in a voltage range between -30 and +30 mV. Single-cell RT-PCR analysis reveal that all DBB neurons responding to hAmylin or A beta were cholinergic. Using specific ion channel blockers, we identified hAmylin and A beta effects on whole cell currents to be mediated, in part, by calcium-dependent conductances. Human amylin also depressed the transient outward (IA) and the delayed rectifier (IK) potassium currents. The hAmylin effects on whole cell currents could be occluded by A beta and vice versa. Human amylin and A beta responses could be blocked with AC187 (50 nM to 1 microM), a specific antagonist for the amylin receptor. The present study indicates that hAmylin, like A beta, is capable of modulating ion channel function in cholinergic basal forebrain neurons. Furthermore, the two peptides may share a common mechanism of action. The ability of an amylin antagonist to block the responses evoked by hAmylin and A beta may provide a novel therapeutic approach for Alzheimer's disease.


Subject(s)
Amyloid beta-Peptides/physiology , Amyloid/physiology , Calcium Channels/drug effects , Cholinergic Fibers/drug effects , Neurons/drug effects , Potassium Channels/drug effects , Prosencephalon/drug effects , Alzheimer Disease/drug therapy , Amyloid/pharmacology , Amyloid beta-Peptides/pharmacology , Animals , Calcium Channel Blockers/pharmacology , Calcium Channels/physiology , Cell Culture Techniques , Cholinergic Fibers/physiology , Dose-Response Relationship, Drug , Islet Amyloid Polypeptide , Male , Neurons/physiology , Patch-Clamp Techniques , Peptide Fragments , Peptides/pharmacology , Potassium Channels/physiology , Prosencephalon/physiology , Rats , Rats, Sprague-Dawley , Receptors, Islet Amyloid Polypeptide , Receptors, Peptide/antagonists & inhibitors , Receptors, Peptide/physiology , Reverse Transcriptase Polymerase Chain Reaction
10.
J Neurophysiol ; 87(2): 696-704, 2002 Feb.
Article in English | MEDLINE | ID: mdl-11826038

ABSTRACT

Galanin, a 29-amino-acid neuropeptide, is generally viewed as an inhibitory neuromodulator in a variety of central systems. Galanin expression is upregulated in the cholinergic basal forebrain nuclei in Alzheimer's disease (AD) and is postulated to play an important role in memory and cognitive function. In this study, application of galanin to acutely dissociated rat neurons from the basal forebrain nucleus diagonal band of Broca (DBB), caused a decrease in whole cell voltage-activated currents in a majority of cells. Galanin reduces a suite of potassium currents, including calcium-activated potassium (I(C)), the delayed rectifier (I(K)), and transient outward potassium (I(A)) conductances, but not calcium or sodium currents. Under current-clamp conditions, application of galanin evoked an increase in excitability and a loss of accommodation in cholinergic DBB neurons. Using single-cell RT-PCR technique, we determined that galanin actions were specific to cholinergic, but not GABAergic DBB neurons The notion that galanin plays a deleterious role in AD is based, in part, on galanin hyperinnervation of cholinergic cells in the basal forebrain of AD patients, its ability to depress acetylcholine release and its inhibitory actions at other CNS sites. However, our results suggest that by virtue of its excitatory actions on cholinergic neurons, galanin may in fact play a compensatory role by augmenting the release of acetylcholine from remaining cholinergic basal forebrain neurons. This action might serve to delay the progression of AD pathology linked to a reduction in central cholinergic tone.


Subject(s)
Alzheimer Disease/metabolism , Cholinergic Fibers/drug effects , Cholinergic Fibers/metabolism , Diagonal Band of Broca/cytology , Galanin/pharmacology , Acetylcholine/metabolism , Action Potentials/drug effects , Action Potentials/physiology , Animals , Barium/pharmacokinetics , Choline O-Acetyltransferase/genetics , Large-Conductance Calcium-Activated Potassium Channels , Male , Patch-Clamp Techniques , Potassium/pharmacokinetics , Potassium Channel Blockers/pharmacology , Potassium Channels, Calcium-Activated/physiology , Rats , Rats, Sprague-Dawley , Reverse Transcriptase Polymerase Chain Reaction , Sodium/pharmacokinetics , Tetraethylammonium/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...