Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
2.
J Undergrad Neurosci Educ ; 16(3): A244-A251, 2018.
Article in English | MEDLINE | ID: mdl-30254539

ABSTRACT

The Faculty for Undergraduate Neuroscience (FUN) has mounted many summer workshops since its first in 1995 held at Davidson College. An important outcome of the 1995 workshop was the development of four "blueprints" to help guide institutions in developing and maintaining undergraduate programs in neuroscience. Since then, at approximately ten-year intervals, participants at the FUN workshops have revisited and amended the Blueprints to better reflect best practices in undergraduate neuroscience education, including adding a fifth blueprint in 2005. In 2017, participants at the FUN workshop held at Dominican University again conducted a review of the blueprints and amended each of the five. A significant change resulting from the 2017 discussions was revision of the neuroscience minor blueprint to reflect the evolution of this program type across institutions.

3.
Am J Physiol Cell Physiol ; 314(2): C233-C241, 2018 02 01.
Article in English | MEDLINE | ID: mdl-29141923

ABSTRACT

Pituitary adenylate cyclase activating polypeptide (PACAP, Adcyap1) activation of PAC1 receptors ( Adcyap1r1) significantly increases excitability of guinea pig cardiac neurons. This modulation of excitability is mediated in part by plasma membrane G protein-dependent activation of adenylyl cyclase and downstream signaling cascades. However, additional mechanisms responsible for the enhanced excitability are activated following internalization of the PAC1 receptor and endosomal signaling. Src family kinases play critical roles mediating endocytosis of many trophic factor and G protein-coupled receptors. The present study investigated whether Src family kinases also support the PACAP-induced PAC1 receptor internalization, phosphorylation of ERK, and enhanced neuronal excitability. Using human embryonic kidney cells stably expressing a green fluorescent protein-tagged PAC1 receptor, treatment with the Src family kinase inhibitor PP2 (10 µM) markedly reduced the PACAP-induced PAC1 receptor internalization, and in parallel, both PP2 and Src inhibitor 1 (Src-1, 2 µM) reduced ERK activation determined by Western blot analysis. In contrast, Src family kinase inhibitors did not eliminate a PACAP-induced rise in global calcium generated by inositol (1,4,5)-trisphosphate-induced release of calcium from endoplasmic reticulum stores. From confocal analysis of phosphorylated ERK immunostaining, PP2 treatment significantly attenuated PACAP activation of ERK in neurons within cardiac ganglia whole mount preparations. Intracellular recordings demonstrated that PP2 also significantly blunted a PACAP-induced increase in cardiac neuron excitability. These studies demonstrate Src-related kinase activity in PAC1 receptor internalization, activation of MEK/ERK signaling, and regulation of neuronal excitability. The present results provide further support for the importance of PAC1 receptor endosomal signaling as a key mechanism regulating cellular function.


Subject(s)
Endocytosis/drug effects , Extracellular Signal-Regulated MAP Kinases/metabolism , Heart/innervation , Neurons/drug effects , Pituitary Adenylate Cyclase-Activating Polypeptide/pharmacology , Protein Kinase Inhibitors/pharmacology , Pyrimidines/pharmacology , Receptors, Pituitary Adenylate Cyclase-Activating Polypeptide/agonists , src-Family Kinases/antagonists & inhibitors , Animals , Calcium Signaling/drug effects , Cell Line , Enzyme Activation , Female , Guinea Pigs , Humans , In Vitro Techniques , Male , Membrane Potentials/drug effects , Mitogen-Activated Protein Kinase Kinases/metabolism , Neurons/enzymology , Phosphorylation , Receptors, Pituitary Adenylate Cyclase-Activating Polypeptide/genetics , Receptors, Pituitary Adenylate Cyclase-Activating Polypeptide/metabolism , src-Family Kinases/metabolism
4.
Am J Physiol Cell Physiol ; 313(2): C219-C227, 2017 Aug 01.
Article in English | MEDLINE | ID: mdl-28592413

ABSTRACT

Forskolin, a selective activator of adenylyl cyclase (AC), commonly is used to establish actions of G protein-coupled receptors (GPCRs) that are initiated primarily through activation of AC/cAMP signaling pathways. In the present study, forskolin was used to evaluate the potential role of AC/cAMP, which is a major signaling mechanism for the pituitary adenylate cyclase-activating polypeptide (PACAP)-selective PAC1 receptor, in the regulation of guinea pig cardiac neuronal excitability. Forskolin (5-10 µM) increases excitability in ~60% of the cardiac neurons. The forskolin-mediated increase in excitability was considered related to cAMP regulation of a cyclic nucleotide gated channel or via protein kinase A (PKA)/ERK signaling, mechanisms that have been linked to PAC1 receptor activation. However, unlike PACAP mechanisms, forskolin enhancement of excitability was not significantly reduced by treatment with cesium to block currents through hyperpolarization-activated nonselective cation channels (Ih) or by treatment with PD98059 to block MEK/ERK signaling. In contrast, treatment with the clathrin inhibitor Pitstop2 or the dynamin inhibitor dynasore eliminated the forskolin-induced increase in excitability; treatments with the inactive Pitstop analog or PP2 treatment to inhibit Src-mediated endocytosis mechanisms were ineffective. The PKA inhibitor KT5702 significantly suppressed the forskolin-induced change in excitability; further, KT5702 and Pitstop2 reduced the forskolin-stimulated MEK/ERK activation in cardiac neurons. Collectively, the present results suggest that forskolin activation of AC/cAMP/PKA signaling leads to the recruitment of clathrin/dynamin-dependent endosomal transduction cascades, including MEK/ERK signaling, and that endosomal signaling is the critical mechanism underlying the forskolin-induced increase in cardiac neuron excitability.


Subject(s)
Adenylyl Cyclases/metabolism , Colforsin/administration & dosage , Heart/drug effects , Myocardium/metabolism , Neurons/drug effects , Animals , Carbazoles/administration & dosage , Clathrin/drug effects , Cyclic AMP/metabolism , Cyclic AMP-Dependent Protein Kinases/metabolism , Endosomes/drug effects , Endosomes/metabolism , Flavonoids/administration & dosage , Guinea Pigs , Humans , MAP Kinase Signaling System/drug effects , Myocardium/pathology , Neurons/metabolism , Neurons/pathology , Pituitary Adenylate Cyclase-Activating Polypeptide/metabolism , Pyrroles/administration & dosage , Receptors, G-Protein-Coupled/metabolism , Receptors, Pituitary Adenylate Cyclase-Activating Polypeptide/metabolism
5.
Am J Physiol Cell Physiol ; 311(4): C643-C651, 2016 Oct 01.
Article in English | MEDLINE | ID: mdl-27488668

ABSTRACT

Pituitary adenylate cyclase (PAC)-activating polypeptide (PACAP) peptides (Adcyap1) signaling at the selective PAC1 receptor (Adcyap1r1) participate in multiple homeostatic and stress-related responses, yet the cellular mechanisms underlying PACAP actions remain to be completely elucidated. PACAP/PAC1 receptor signaling increases excitability of neurons within the guinea pig cardiac ganglia, and as these neurons are readily accessible, this neuronal system is particularly amenable to study of PACAP modulation of ionic conductances. The present study investigated how PACAP activation of MEK/ERK signaling contributed to the peptide-induced increase in cardiac neuron excitability. Treatment with the MEK inhibitor PD 98059 blocked PACAP-stimulated phosphorylated ERK and, in parallel, suppressed the increase in cardiac neuron excitability. However, PD 98059 did not blunt the ability of PACAP to enhance two inward ionic currents, one flowing through hyperpolarization-activated nonselective cationic channels (Ih) and another flowing through low-voltage-activated calcium channels (IT), which support the peptide-induced increase in excitability. Thus a PACAP- and MEK/ERK-sensitive, voltage-dependent conductance(s), in addition to Ih and IT, modulates neuronal excitability. Despite prior work implicating PACAP downregulation of the KV4.2 potassium channel in modulation of excitability in other cells, treatment with the KV4.2 current blocker 4-aminopyridine did not replicate the PACAP-induced increase in excitability in cardiac neurons. However, cardiac neurons express the ERK target, the NaV1.7 sodium channel, and treatment with the selective NaV1.7 channel inhibitor PF-04856264 decreased the PACAP modulation of excitability. From these results, PACAP/PAC1 activation of MEK/ERK signaling may phosphorylate the NaV1.7 channel, enhancing sodium currents near the threshold, an action contributing to repetitive firing of the cardiac neurons exposed to PACAP.


Subject(s)
Action Potentials/physiology , Heart/physiology , MAP Kinase Signaling System/physiology , Neurons/metabolism , Neurons/physiology , Pituitary Adenylate Cyclase-Activating Polypeptide/metabolism , Signal Transduction/physiology , Animals , Calcium Channels/metabolism , Female , Guinea Pigs , Male , Myocardium/metabolism , NAV1.7 Voltage-Gated Sodium Channel/metabolism , Receptors, Pituitary Adenylate Cyclase-Activating Polypeptide/metabolism , Shal Potassium Channels/metabolism
7.
Am J Physiol Heart Circ Physiol ; 309(7): H1198-206, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26276818

ABSTRACT

This paper aims to determine whether chronic vagus nerve stimulation (VNS) mitigates myocardial infarction (MI)-induced remodeling of the intrinsic cardiac nervous system (ICNS), along with the cardiac tissue it regulates. Guinea pigs underwent VNS implantation on the right cervical vagus. Two weeks later, MI was produced by ligating the ventral descending coronary artery. VNS stimulation started 7 days post-MI (20 Hz, 0.9 ± 0.2 mA, 14 s on, 48 s off; VNS-MI, n = 7) and was compared with time-matched MI animals with sham VNS (MI n = 7) vs. untreated controls (n = 8). Echocardiograms were performed before and at 90 days post-MI. At termination, IC neuronal intracellular voltage recordings were obtained from whole-mount neuronal plexuses. MI increased left ventricular end systolic volume (LVESV) 30% (P = 0.027) and reduced LV ejection fraction (LVEF) 6.5% (P < 0.001) at 90 days post-MI compared with baseline. In the VNS-MI group, LVESV and LVEF did not differ from baseline. IC neurons showed depolarization of resting membrane potentials and increased input resistance in MI compared with VNS-MI and sham controls (P < 0.05). Neuronal excitability and sensitivity to norepinephrine increased in MI and VNS-MI groups compared with controls (P < 0.05). Synaptic efficacy, as determined by evoked responses to stimulating input axons, was reduced in VNS-MI compared with MI or controls (P < 0.05). VNS induced changes in myocytes, consistent with enhanced glycogenolysis, and blunted the MI-induced increase in the proapoptotic Bcl-2-associated X protein (P < 0.05). VNS mitigates MI-induced remodeling of the ICNS, correspondingly preserving ventricular function via both neural and cardiomyocyte-dependent actions.


Subject(s)
Autonomic Nervous System/physiopathology , Heart/innervation , Myocardial Infarction/physiopathology , Myocytes, Cardiac/metabolism , Neuronal Plasticity/physiology , Vagus Nerve Stimulation , Ventricular Dysfunction, Left/physiopathology , Animals , Evoked Potentials , Glycogenolysis , Guinea Pigs , Membrane Potentials , Norepinephrine/metabolism , Stroke Volume/physiology , Synaptic Transmission , Ventricular Function, Left , bcl-2-Associated X Protein/metabolism
8.
Am J Physiol Regul Integr Comp Physiol ; 309(2): R179-88, 2015 Jul 15.
Article in English | MEDLINE | ID: mdl-25947168

ABSTRACT

Neurohumoral remodeling is fundamental to the evolution of heart disease. This study examined the effects of chronic treatment with an ACE inhibitor (captopril, 3 mg·kg(-1)·day(-1)), AT1 receptor antagonist (losartan, 3 mg·kg(-1)·day(-1)), or AT2 receptor agonist (CGP42112A, 0.14 mg·kg(-1)·day(-1)) on remodeling of the guinea pig intrinsic cardiac plexus following chronic myocardial infarction (MI). MI was surgically induced and animals recovered for 6 or 7 wk, with or without drug treatment. Intracellular voltage recordings from whole mounts of the cardiac plexus were used to monitor changes in neuronal responses to norepinephrine (NE), muscarinic agonists (bethanechol), or ANG II. MI produced an increase in neuronal excitability with NE and a loss of sensitivity to ANG II. MI animals treated with captopril exhibited increased neuronal excitability with NE application, while MI animals treated with CGP42112A did not. Losartan treatment of MI animals did not alter excitability with NE compared with untreated MIs, but these animals did show an enhanced synaptic efficacy. This effect on synaptic function was likely due to presynaptic AT1 receptors, since ANG II was able to reduce output to nerve fiber stimulation in control animals, and this effect was prevented by inclusion of losartan in the bath solution. Analysis of AT receptor expression by Western blot showed a decrease in both AT1 and AT2 receptors with MI that was reversed by all three drug treatments. These data indicate that neuronal remodeling of the guinea pig cardiac plexus following MI is mediated, in part, by activation of both AT1 and AT2 receptors.


Subject(s)
Heart/innervation , Myocardial Infarction/metabolism , Presynaptic Terminals/metabolism , Receptor, Angiotensin, Type 1/metabolism , Receptor, Angiotensin, Type 2/metabolism , Receptors, Presynaptic/metabolism , Action Potentials , Angiotensin II/pharmacology , Angiotensin II Type 1 Receptor Blockers/pharmacology , Angiotensin II Type 2 Receptor Blockers/pharmacology , Animals , Disease Models, Animal , Electric Stimulation , Evoked Potentials , Guinea Pigs , Male , Myocardial Infarction/pathology , Myocardial Infarction/physiopathology , Norepinephrine/pharmacology , Presynaptic Terminals/drug effects , Receptor, Angiotensin, Type 1/drug effects , Receptor, Angiotensin, Type 2/drug effects , Receptors, Presynaptic/antagonists & inhibitors , Signal Transduction , Time Factors
9.
Auton Neurosci ; 181: 4-12, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24220238

ABSTRACT

Myocardial infarction (MI) is associated with remodeling of the heart and neurohumoral control systems. The objective of this study was to define time-dependent changes in intrinsic cardiac (IC) neuronal excitability, synaptic efficacy, and neurochemical modulation following MI. MI was produced in guinea pigs by ligation of the coronary artery and associated vein on the dorsal surface of the heart. Animals were recovered for 4, 7, 14, or 50 days. Intracellular voltage recordings were obtained in whole mounts of the cardiac neuronal plexus to determine passive and active neuronal properties of IC neurons. Immunohistochemical analysis demonstrated an immediate and persistent increase in the percentage of IC neurons immunoreactive for neuronal nitric oxide synthase. Examination of individual neuronal properties demonstrated that after hyperpolarizing potentials were significantly decreased in both amplitude and time course of recovery at 7 days post-MI. These parameters returned to control values by 50 days post-MI. Synaptic efficacy, as determined by the stimulation of axonal inputs, was enhanced at 7 days post-MI only. Neuronal excitability in absence of agonist challenge was unchanged following MI. Norepinephrine increased IC excitability to intracellular current injections, a response that was augmented post-MI. Angiotensin II potentiation of norepinephrine and bethanechol-induced excitability, evident in controls, was abolished post-MI. This study demonstrates that MI induces both persistent and transient changes in IC neuronal functions immediately following injury. Alterations in the IC neuronal network, which persist for weeks after the initial insult, may lead to alterations in autonomic signaling and cardiac control.


Subject(s)
Heart/innervation , Heart/physiopathology , Myocardial Infarction/physiopathology , Sympathetic Nervous System/physiopathology , Angiotensin II/pharmacology , Animals , Axons/drug effects , Axons/physiology , Bethanechol/pharmacology , Chronic Disease , Guinea Pigs , Heart/drug effects , In Vitro Techniques , Male , Membrane Potentials/drug effects , Membrane Potentials/physiology , Myocardial Infarction/drug therapy , Neurons/drug effects , Neurons/physiology , Nitric Oxide Synthase Type I/metabolism , Norepinephrine/pharmacology , Parasympathomimetics/pharmacology , Sympathetic Nervous System/drug effects , Sympathomimetics/pharmacology , Synaptic Transmission/drug effects , Synaptic Transmission/physiology , Time Factors , Vasoconstrictor Agents/pharmacology
10.
J Neurosci ; 33(10): 4614-22, 2013 Mar 06.
Article in English | MEDLINE | ID: mdl-23467377

ABSTRACT

After G-protein-coupled receptor activation and signaling at the plasma membrane, the receptor complex is often rapidly internalized via endocytic vesicles for trafficking into various intracellular compartments and pathways. The formation of signaling endosomes is recognized as a mechanism that produces sustained intracellular signals that may be distinct from those generated at the cell surface for cellular responses including growth, differentiation, and survival. Pituitary adenylate cyclase activating polypeptide (PACAP; Adcyap1) is a potent neurotransmitter/neurotrophic peptide and mediates its diverse cellular functions in part through internalization of its cognate G-protein-coupled PAC1 receptor (PAC1R; Adcyap1r1). In the present study, we examined whether PAC1R endocytosis participates in the regulation of neuronal excitability. Although PACAP increased excitability in 90% of guinea pig cardiac neurons, pretreatment with Pitstop 2 or dynasore to inhibit clathrin and dynamin I/II, respectively, suppressed the PACAP effect. Subsequent addition of inhibitor after the PACAP-induced increase in excitability developed gradually attenuated excitability with no changes in action potential properties. Likewise, the PACAP-induced increase in excitability was markedly decreased at ambient temperature. Receptor trafficking studies with GFP-PAC1 cell lines demonstrated the efficacy of Pitstop 2, dynasore, and low temperatures at suppressing PAC1R endocytosis. In contrast, brefeldin A pretreatments to disrupt Golgi vesicle trafficking did not blunt the PACAP effect, and PACAP/PAC1R signaling still increased neuronal cAMP production even with endocytic blockade. Our results demonstrate that PACAP/PAC1R complex endocytosis is a key step for the PACAP modulation of cardiac neuron excitability.


Subject(s)
Action Potentials/drug effects , Myocardium/cytology , Neurons/drug effects , Pituitary Adenylate Cyclase-Activating Polypeptide/pharmacology , Receptors, Pituitary Adenylate Cyclase-Activating Polypeptide/metabolism , Signal Transduction/drug effects , Action Potentials/genetics , Animals , Animals, Newborn , Barium Compounds/pharmacology , Biophysics , Brefeldin A/pharmacology , Cells, Cultured , Chlorides/pharmacology , Cyclic AMP/metabolism , Drug Administration Schedule , Electric Stimulation , Endosomes/drug effects , Enzyme Inhibitors/pharmacology , Female , Gene Expression Regulation/drug effects , Gene Expression Regulation/genetics , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Guinea Pigs , Humans , Hydrazones/pharmacology , In Vitro Techniques , Male , Neurons/physiology , Patch-Clamp Techniques , Rats , Receptors, Pituitary Adenylate Cyclase-Activating Polypeptide/genetics , Signal Transduction/physiology , Sulfonamides/pharmacology , Superior Cervical Ganglion/cytology , Temperature , Thiazolidines/pharmacology , Transfection
11.
Am J Physiol Regul Integr Comp Physiol ; 303(9): R950-8, 2012 Nov 01.
Article in English | MEDLINE | ID: mdl-22933026

ABSTRACT

Chronic heart disease induces remodeling of cardiac tissue and associated neuronal components. Treatment of chronic heart disease often involves pharmacological blockade of adrenergic receptors. This study examined the specific changes in neuronal sensitivity of guinea pig intrinsic cardiac neurons to autonomic modulators in animals with chronic cardiac disease, in the presence or absence of adrenergic blockage. Myocardial infarction (MI) was produced by ligature of the coronary artery and associated vein on the dorsal surface of the heart. Pressure overload (PO) was induced by a banding of the descending dorsal aorta (∼20% constriction). Animals were allowed to recover for 2 wk and then implanted with an osmotic pump (Alzet) containing either timolol (2 mg·kg(-1)·day(-1)) or vehicle, for a total of 6-7 wk of drug treatment. At termination, intracellular recordings from individual neurons in whole mounts of the cardiac plexus were used to assess changes in physiological responses. Timolol treatment did not inhibit the increased sensitivity to norepinephrine seen in both MI and PO animals, but it did inhibit the stimulatory effects of angiotensin II on the norepinephrine-induced increases in neuronal excitability. Timolol treatment also inhibited the increase in synaptically evoked action potentials observed in PO animals with stimulation of fiber tract bundles. These results demonstrate that ß-adrenergic blockade can inhibit specific aspects of remodeling within the intrinsic cardiac plexus. In addition, this effect was preferentially observed with active cardiac disease states, indicating that the ß-receptors were more influential on remodeling during dynamic disease progression.


Subject(s)
Adrenergic beta-Antagonists/pharmacology , Heart Diseases/physiopathology , Heart/innervation , Neurons/physiology , Receptors, Adrenergic, beta/drug effects , Receptors, Adrenergic, beta/physiology , Adrenergic Agonists/pharmacology , Angiotensin II/pharmacology , Animals , Cholinergic Agents/pharmacology , Chronic Disease , Disease Models, Animal , Evoked Potentials/drug effects , Evoked Potentials/physiology , Guinea Pigs , Male , Myocardial Infarction/physiopathology , Neurons/drug effects , Timolol/pharmacology
12.
J Undergrad Neurosci Educ ; 11(1): A27-37, 2012.
Article in English | MEDLINE | ID: mdl-23494749

ABSTRACT

There has been a growing emphasis on the use of core competencies to design and inform curricula. Based on our Faculty for Undergraduate Neuroscience workshop at Pomona we developed a set of neuroscience core competencies. Following the workshop, faculty members were asked to complete an online survey to determine which core competencies are considered most essential and the results are presented. Backward Design principles are then described and we discuss how core competencies, through a backward design process, can be used to design and assess an undergraduate neuroscience curriculum. Oberlin College is used as a case study to describe the use of core competencies to help develop learning objectives, activities, and assessment measures for an undergraduate neuroscience major.

13.
Am J Physiol Regul Integr Comp Physiol ; 301(5): R1391-9, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21865545

ABSTRACT

The intrinsic cardiac plexus represents a major peripheral integration site for neuronal, hormonal, and locally produced neuromodulators controlling efferent neuronal output to the heart. This study examined the interdependence of norepinephrine, muscarinic agonists, and ANG II, to modulate intrinsic cardiac neuronal activity. Intracellular voltage recordings from whole-mount preparations of the guinea pig cardiac plexus were used to determine changes in active and passive electrical properties of individual intrinsic cardiac neurons. Application of either adrenergic or muscarinic agonists induced changes in neuronal resting membrane potentials, decreased afterhyperpolarization duration of single action potentials, and increased neuronal excitability. Adrenergic responses were inhibited by removal of extracellular calcium ions, while muscarinic responses were inhibited by application of TEA. The adrenergic responses were heterogeneous, responding to a variety of receptor-specific agonists (phenylephrine, clonidine, dobutamine, and terbutaline), although α-receptor agonists produced the most frequent responses. Application of ANG II alone produced a significant increase in excitability, while application of ANG II in combination with either adrenergic or muscarinic agonists produced a much larger potentiation of excitability. The ANG II-induced modulation of firing was blocked by the angiotensin type 2 (AT(2)) receptor inhibitor PD 123319 and was mimicked by the AT(2) receptor agonist CGP-42112A. AT(1) receptor blockade with telmasartin did not alter neuronal responses to ANG II. These data demonstrate that ANG II potentiates both muscarinically and adrenergically mediated activation of intrinsic cardiac neurons, doing so primarily via AT(2) receptor-dependent mechanisms. These neurohumoral interactions may be fundamental to regulation of neuronal excitability within the intrinsic cardiac nervous system.


Subject(s)
Adrenergic Agonists/pharmacology , Heart/innervation , Muscarinic Agonists/pharmacology , Neurons/drug effects , Receptors, Adrenergic/drug effects , Receptors, Muscarinic/drug effects , Angiotensin II/metabolism , Angiotensin Receptor Antagonists/pharmacology , Animals , Calcium/metabolism , Electric Stimulation , Guinea Pigs , In Vitro Techniques , Male , Membrane Potentials , Neurons/metabolism , Potassium/metabolism , Potassium Channel Blockers/pharmacology , Receptor, Angiotensin, Type 1/drug effects , Receptor, Angiotensin, Type 1/metabolism , Receptor, Angiotensin, Type 2/drug effects , Receptor, Angiotensin, Type 2/metabolism , Receptors, Adrenergic/metabolism , Receptors, Muscarinic/metabolism , Time Factors
14.
J Undergrad Neurosci Educ ; 8(2): A101-7, 2010.
Article in English | MEDLINE | ID: mdl-23493671

ABSTRACT

A survey was presented to members of the Faculty for Undergraduate Neuroscience (FUN) to get a better idea of how neuroscience research and education is being delivered at the undergraduate level. A total of 155 individuals completed the survey, with 118 coming from faculty at traditional PUIs (primarily undergraduate institutions) and 37 from faculty at doctoral-granting institutions. The survey covered a number of different areas; including types of neuroscience programs, number of neuroscience faculty at the institution, average course loads, average number of research students, and external support for research. Results from this survey indicate that the structure of neuroscience programs vary among institutions. Course loads for faculty at PUIs averaged four to six courses per year and the total number of undergraduate students supervised in research per faculty member averaged five (± 2.8) students per year. Faculty show high success with external funding, both at PUIs and research universities. Faculty ranked FUN programs devoted to supporting both students and faculty development highly. The results of this survey provide data that can be used to determine future directions and priorities for FUN.

15.
Am J Physiol Regul Integr Comp Physiol ; 297(3): R859-66, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19605763

ABSTRACT

Chronic pressure overload (PO) is associated with cardiac hypertrophy and altered autonomic control of cardiac function, in which the latter may involve adaptations in central and/or peripheral cardiac neural control mechanisms. To evaluate the specific remodeling of the intrinsic cardiac nervous system following pressure overload, the descending thoracic aorta artery of the guinea pig was constricted approximately 20%, and the animals recovered for 9 wk. Thereafter, atrial neurons of the intrinsic cardiac plexus were isolated for electrophysiological and immunohistochemical analyses. Intracellular voltage recordings from intrinsic cardiac neurons demonstrated no significant changes in passive membrane properties or action potential depolarization compared with age-matched controls and sham-operated animals, but afterhyperpolarization duration was increased in PO animals. Neuronal excitability, as determined by the number of action potentials produced with depolarizing stimuli, was differentially increased in phasic neurons derived from PO animals in response to exogenously applied histamine compared with sham and age-matched controls. Conversely, pituitary adenylate cyclase-activating polypeptide-induced increases in intrinsic cardiac neuron evoked AP frequency were similar between control and PO animals. Immunohistochemical analysis demonstrated a twofold increase in the percentage of neurons immunoreactive for neuronal nitric oxide synthase in PO animals compared with control. The density of mast cells within the intrinsic cardiac plexus from PO animals was also increased twofold compared with preparations from control animals. These results indicate that congestive heart failure associated with chronic pressure overload induces a differential remodeling of intrinsic cardiac neurons and upregulation of neuronal responsiveness to specific neuromodulators.


Subject(s)
Cardiomegaly/physiopathology , Heart Atria/innervation , Heart Failure/physiopathology , Hypertension/complications , Parasympathetic Fibers, Postganglionic/physiopathology , Action Potentials , Animals , Aorta, Thoracic/surgery , Cardiomegaly/etiology , Cardiomegaly/pathology , Chronic Disease , Constriction , Disease Models, Animal , Evoked Potentials , Guinea Pigs , Heart Failure/etiology , Heart Failure/pathology , Histamine/metabolism , Hypertension/pathology , Hypertension/physiopathology , Male , Mast Cells/pathology , Nitric Oxide Synthase Type I/metabolism , Parasympathetic Fibers, Postganglionic/metabolism , Pituitary Adenylate Cyclase-Activating Polypeptide/metabolism , Time Factors
16.
Am J Physiol Regul Integr Comp Physiol ; 295(6): R1926-33, 2008 Dec.
Article in English | MEDLINE | ID: mdl-18832084

ABSTRACT

Chronic myocardial infarction (CMI) is associated with remodeling of the ventricle and evokes adaption in the cardiac neurohumoral control systems. To evaluate the remodeling of the intrinsic cardiac nervous system following myocardial infarction, the dorsal descending coronary artery was ligated in the guinea pig heart and the animals were allowed to recover for 7-9 wk. Thereafter, atrial neurons of the intrinsic cardiac plexus were isolated for electrophysiological and immunohistochemical analyses. Intracellular voltage recordings from intrinsic cardiac neurons demonstrated no significant changes in passive membrane properties or action potential configuration compared with age-matched controls and sham-operated animals. The intrinsic cardiac neurons from chronic infarcted hearts did demonstrate an increase in evoked action potential (AP) frequency (as determined by the number of APs produced with depolarizing stimuli) and an increase in responses to exogenously applied histamine compared with sham and age-matched controls. Conversely, pituitary adenylate cyclase-activating polypeptide (PACAP)-induced increases in intrinsic cardiac neuron-evoked AP frequency were similar between control and CMI animals. Immunohistochemical analysis demonstrated a threefold increase in percentage of neurons immunoreactive for neuronal nitric oxide synthase (NOS) in CMI animals compared with control and the additional expression of inducible NOS by some neurons, which was not evident in control animals. Finally, the density of mast cells within the intrinsic cardiac plexus was increased threefold in preparations from CMI animals. These results indicate that CMI induces a differential remodeling of intrinsic cardiac neurons and functional upregulation of neuronal responsiveness to specific neuromodulators.


Subject(s)
Autonomic Nervous System/physiopathology , Heart/innervation , Myocardial Infarction/physiopathology , Action Potentials , Animals , Autonomic Nervous System/enzymology , Autonomic Nervous System/pathology , Chronic Disease , Disease Models, Animal , Evoked Potentials , Guinea Pigs , Histamine/metabolism , Immunohistochemistry , Male , Mast Cells/pathology , Myocardial Infarction/pathology , Nitric Oxide Synthase Type I/metabolism , Nitric Oxide Synthase Type II/metabolism , Phenotype , Pituitary Adenylate Cyclase-Activating Polypeptide/metabolism , Time Factors
18.
Ann N Y Acad Sci ; 1070: 317-21, 2006 Jul.
Article in English | MEDLINE | ID: mdl-16888185

ABSTRACT

Pituitary adenylate cyclase-activating polypeptide (PACAP) effects on intracellular calcium ([Ca2+]i) and excitability have been studied in adult guinea pig intracardiac neurons. PACAP increased excitability, but did not elicit Ca2+ release from intracellular stores. Exposure to a Ca2+-deficient solution did not deplete [Ca2+]i stores but did eliminate the PACAP-induced increase in excitability. We postulate that Ca2+ influx is required for the PACAP-induced increase in excitability.


Subject(s)
Calcium/metabolism , Heart/drug effects , Ion Channels/metabolism , Myocardium/metabolism , Neurons/drug effects , Neurons/metabolism , Pituitary Adenylate Cyclase-Activating Polypeptide/pharmacology , Animals , Cytosol/drug effects , Cytosol/metabolism , Guinea Pigs , Ion Channel Gating
19.
J Neurophysiol ; 95(4): 2134-42, 2006 Apr.
Article in English | MEDLINE | ID: mdl-16371456

ABSTRACT

Mechanisms modulating the pituitary adenylate cyclase activating polypeptide (PACAP)-induced increase in excitability have been studied using dissociated guinea pig intrinsic cardiac neurons and intact ganglion preparations. Measurements of intracellular calcium (Ca2+) with the fluorescent Ca2+ indicator dye fluo-3 indicated that neither PACAP nor vasoactive intestinal polypeptide (VIP) at either 100 nM or 1 microM produced a discernible elevation of intracellular Ca2+ in dissociated intracardiac neurons. For neurons in ganglion whole mount preparations kept in control bath solution, local application of PACAP significantly increased excitability, as indicated by the number of action potentials generated by long depolarizing current pulses. However, in a Ca2+ -deficient solution in which external Ca2+ was replaced by Mg2+ or when cells were bathed in control solution containing 200 microM Cd2+, PACAP did not enhance action potential firing. In contrast, in a Ca2+ -deficient solution with Ca2+ replaced by strontium (Sr2+), PACAP increased excitability. PACAP increased excitability in cells treated with a combination of 20 microM ryanodine and 10 mM caffeine to interrupt release of Ca2+ from internal stores. Experiments using fluo-3 showed that ryanodine/caffeine pretreatment eliminated subsequent caffeine-induced Ca2+ release from intracellular stores, whereas exposure to the Ca2+ -deficient solution did not. In dissociated intracardiac neurons voltage clamped with the perforated patch recording technique, 100 nM PACAP decreased the voltage-dependent barium current (IBa). These results show that, in the guinea pig intracardiac neurons, the PACAP-induced increase in excitability apparently requires Ca2+ influx through Cd2+ -sensitive calcium permeable channels other than voltage-dependent Ca2+ channels, but not Ca2+ release from internal stores.


Subject(s)
Calcium/metabolism , Heart/innervation , Neurons/physiology , Pituitary Adenylate Cyclase-Activating Polypeptide/pharmacology , Action Potentials/drug effects , Animals , Barium/metabolism , Cadmium/pharmacology , Caffeine/pharmacology , Calcium Channels/drug effects , Calcium Channels/physiology , Electrophysiology , Female , Guinea Pigs , Magnesium/metabolism , Male , Neurons/chemistry , Neurons/drug effects , Patch-Clamp Techniques , Ryanodine/pharmacology , Strontium/metabolism , Vasoactive Intestinal Peptide/pharmacology
20.
Am J Physiol Regul Integr Comp Physiol ; 290(1): R241-50, 2006 Jan.
Article in English | MEDLINE | ID: mdl-16166202

ABSTRACT

Histamine, released from mast cells, can modulate the activity of intrinsic neurons in the guinea pig cardiac plexus. The present study examined the ionic mechanisms underlying the histamine-induced responses in these cells. Histamine evokes a small membrane depolarization and an increase in neuronal excitability. Using intracellular voltage recording from individual intracardiac neurons, we were able to demonstrate that removal of extracellular sodium reduced the membrane depolarization, whereas inhibition of K+ channels by 1 mM Ba2+, 2 mM Cs+, or 5 mM tetraethylammonium had no effect. The depolarization was also not inhibited by either 10 microM Gd3+ or a reduced Cl- solution. The histamine-induced increase in excitability was unaffected by K+ channel inhibitors; however, it was reduced by either blockage of voltage-gated Ca2+ channels with 200 microM Cd2+ or replacement of extracellular Ca2+ with Mg2+. Conversely, alterations in intracellular calcium with thapsigargin or caffeine did not inhibit the histamine-induced effects. However, in cells treated with both thapsigargin and caffeine to deplete internal calcium stores, the histamine-induced increase in excitability was decreased. Treatment with the phospholipase C inhibitor U73122 also prevented both the depolarization and the increase in excitability. From these data, we conclude that histamine, via activation of H1 receptors, activates phospholipase C, which results in 1) the opening of a nonspecific cation channel, such as a transient receptor potential channel 4 or 5; and 2) in combination with either the influx of Ca2+ through voltage-gated channels or the release of internal calcium stores leads to an increase in excitability.


Subject(s)
Heart/innervation , Histamine/pharmacology , Neurons/drug effects , Animals , Calcium/metabolism , Calcium/pharmacology , Estrenes/pharmacology , Female , Guinea Pigs , Male , Neurons/metabolism , Phosphodiesterase Inhibitors/pharmacology , Potassium Channel Blockers/pharmacology , Pyrrolidinones/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...