• Refine Query
  • Source
  • Publication year
  • to
  • Language
  • 1
  • Tagged with
  • 7
  • 7
  • 4
  • 4
  • 3
  • 2
  • 2
  • 2
  • 2
  • 2
  • 2
  • 2
  • 2
  • 2
  • 1
  • About
  • The Global ETD Search service is a free service for researchers to find electronic theses and dissertations. This service is provided by the Networked Digital Library of Theses and Dissertations.
    Our metadata is collected from universities around the world. If you manage a university/consortium/country archive and want to be added, details can be found on the NDLTD website.
1

Impairment of Baroreflex Control of Heart Rate and Structural Changes of Cardiac Ganglia in Conscious Streptozotocin (STZ)-Induced Diabetic Mice

Lin, Min, Ai, Jing, Harden, Scott W., Huang, Chenghui, Li, Lihua, Wurster, Robert D., Cheng, Zixi (Jack) 24 June 2010 (has links)
Baroreflex control of heart rate (HR) is impaired in human diabetes mellitus and in large experimental models. However, baroreflex impairment in diabetic mouse models and diabetes-induced remodeling of baroreflex circuitry are not well studied. We examined the impairment of baroreflex control of heart rate (HR) and assessed structural remodeling of cardiac ganglia in the streptozotocin (STZ)-induced diabetic mouse model. FVB mice were either injected with vehicle or STZ. Group 1: mice were anesthetized and the femoral artery and vein were catheterized at the 30th day after vehicle or STZ injection. On the second day after surgery, baroreflex-mediated HR responses to sodium nitroprusside (SNP) and phenylephrine (PE)-induced mean arterial blood pressure (MABP) changes were measured in conscious mice. Group 2: Fluoro-Gold was administered (i.p.) to label cardiac ganglia in each mouse at the 25th day after vehicle or STZ injection. After another five days, animals were perfused and cardiac ganglia were examined using confocal microscopy. Compared with control, we found in STZ mice: 1) the HR decreased, but MABP did not. 2) The PE-induced increases of MABP were decreased. 3) Baroreflex bradycardia was attenuated in the rapid MABP ascending phase but the steady-state ΔHR/ΔMABP was not different at all PE doses. 4) SNP-induced MABP decreases were not different. 5) Baroreflex tachycardia was attenuated. 6) The sizes of cardiac ganglia and ganglionic principal neurons were decreased. 7) The ratio of nucleus/cell body of cardiac ganglionic neurons was increased. We conclude that baroreflex control of HR is impaired in conscious STZ mice. In addition, diabetes may induce a significant structural remodeling of cardiac ganglia. Such an anatomical change of cardiac ganglia may provide new information for the understanding of diabetes-induced remodeling of the multiple components within the baroreflex circuitry.
2

Impairment of Baroreflex Control of Heart Rate in Conscious Transgenic Mice of Type 1 Diabetes (OVE26)

Lin, Min, Harden, Scott W., Li, Lihua, Wurster, Robert D., Cheng, Zixi J. 15 January 2010 (has links)
Baroreflex control of heart rate (HR) is impaired in human type 1 diabetes mellitus. The goal of this study is to use a transgenic mouse model of type 1 diabetes (OVE26) to assess the diabetes-induced baroreflex impairment in the conscious state. OVE26 transgenic mice (which develop hyperglycemia within the first three weeks after birth due to the specific damage of beta cells) and normal control mice (FVB) 5-6 months of age were anesthetized, and the left femoral artery and both veins were catheterized. On the second day after surgery, baroreflex-mediated HR responses to arterial blood pressure (ABP) changes that were induced by separate microinfusion of phenylephrine (PE) and sodium nitroprusside (SNP) at different doses (0.03-0.4 μg/min) were measured in the conscious state. Compared with FVB control, we found that in OVE26 diabetic mice 1) mean ABP (MABP) and HR were decreased (p < 0.05). 2) PE-induced MABP increases were comparable to those in FVB mice (p > 0.05). 3) Baroreflex-mediated bradycardia was attenuated (p < 0.05). 4) SNP-induced MABP decreases was reduced (p < 0.05). 5) Baroreflex-mediated tachycardia was attenuated (p < 0.05). Since baroreflex control of HR in conscious OVE26 mice is impaired in a similar fashion to human diabetes mellitus, we suggest that OVE26 mice may provide a useful model to study the neural mechanisms of diabetes-induced baroreflex impairment.
3

Diabetes Induces Neural Degeneration in Nucleus Ambiguus (NA) and Attenuates Heart Rate Control in OVE26 Mice

Yan, Binbin, Li, Lihua, Harden, Scott W., Epstein, Paul N., Wurster, Robert D., Cheng, Zixi (Jack) 01 November 2009 (has links)
Baroreflex sensitivity is impaired by diabetes mellitus. Previously, we found that diabetes induces a deficit of central mediation of baroreflex-mediated bradycardia. In this study, we assessed whether diabetes induces degeneration of the nucleus ambiguus (NA) and reduces heart rate (HR) responses to l-Glutamate (L-Glu) microinjection into the NA. FVB control and OVE26 diabetic mice (5-6 months) were anesthetized. Different doses of L-Glu (0.1-5 mM/l, 20 nl) were delivered into the left NA using a multi-channel injector. In other animals, the left vagus was electrically stimulated at 1-40 Hz (1 ms, 0.5 mA, 20 s). HR and mean arterial blood pressure (MAP) responses to L-Glu microinjections into the NA and to the electrical stimulation of the vagus were measured. The NA region was defined by tracer TMR-D injection into the ipsilateral nodose ganglion to retrogradely label vagal motoneurons in the NA. Brainstem slices at - 600, - 300, 0, + 300, and + 600 μm relative to the obex were processed using Nissl staining and the number of NA motoneurons was counted. Compared with FVB control, we found in OVE26 mice that: 1) HR responses to L-Glu injection into the NA at doses of 0.2-0.4 (mM/l, 20 nl) were attenuated (p < 0.05), but MAP responses were unchanged (p > 0.05). 2) HR responses to vagal stimulation were increased (p < 0.05). 3) The total number of NA (left and right) motoneurons was reduced (p < 0.05). Taken together, we concluded that diabetes reduces NA control of HR and induces degeneration of NA motoneurons. Degeneration of NA cardiac motoneurons may contribute to impairment of reflex-bradycardia in OVE26 diabetic mice.
4

Parasympathetic Control of the Heart. III. Neuropeptide Y-Immunoreactive Nerve Terminals Synapse on Three Populations of Negative Chronotropic Vagal Preganglionic Neurons

Gray, Alrich L., Johnson, Tannis A., Lauenstein, Jean Marie, Newton, Stephen S., Ardell, Jeffrey L., Massari, V. John 01 June 2004 (has links)
The vagal postganglionic control of cardiac rate is mediated by two intracardiac ganglia, i.e., the sinoatrial (SA) and posterior atrial (PA) ganglia. Nothing is known about the vagal preganglionic neurons (VPNs) that innervate the PA ganglion or about the neurochemical anatomy of central afferents that innervate these VPNs. These issues were examined using light microscopic retrograde labeling methods and dual-labeling electron microscopic histochemical and immunocytochemical methods. VPNs projecting to the PA ganglion are found in a narrow column exclusively in the ventrolateral nucleus ambiguus (NA-VL). These neurons are relatively large (37.6 ± 2.7 μm by 21.3 ± 3.4 μm) with abundant cytoplasm and intracellular organelles, rare somatic and dendritic spines, round uninvaginated nuclei, and myelinated axons. Previous physiological data indicated that microinjections of neuropeptide Y (NPY) into the NA-VL cause negative chronotropic effects. The present morphological data demonstrate that NPY-immunoreactive nerve terminals formed 18 ± 4% of the axodendritic or axosomatic synapses and close appositions on VPNs projecting to the PA ganglion. Three approximately equal populations of VPNs in the NA-VL were retrogradely labeled from the SA and PA ganglia. One population each projects to the SA ganglion, the PA ganglion, or to both the SA and PA ganglia. Therefore, there are both shared and independent pathways involved in the vagal preganglionic controls of cardiac rate. These data are consistent with the hypothesis that the central and peripheral parasympathetic controls of cardiac rate are coordinated by multiple potentially redundant and/or interacting pathways and mechanisms.
5

Amylin mediates brainstem control of heart rate in the diving reflex

Yang, Fan January 2012 (has links)
Amylin, or islet amyloid polypeptide is a 37-amino acid member of the calcitonin peptide family. Amylin role in the brainstem and its function in regulating heart rates is unknown. The diving reflex is a powerful autonomic reflex, however no neuropeptides have been described to modulate its function. In this thesis study, amylin expression in the brainstem involving pathways between the trigeminal ganglion and the nucleus ambiguus was visualized and characterized using immunohistochemistry. Its functional role in slowing heart rate and also its involvement in the diving reflex were elucidated using stereotaxic microinjection, whole-cel patch-clamp, and a rat diving model. Immunohistochemical and tract tracing studies in rats revealed amylin expression in trigeminal ganglion cells, which also contained vesicular glutamate transporter 2 positive. With respect to the brainstem, amylin containing fibers were discovered in spinal trigeminal tracts. These fibers curved dorsally toward choline acetyltransferase immunoreactive neurons of the nucleus ambiguus, suggesting that amylin may synapse to parasympathetic preganglionic neurons in the nucleus ambiguus. Microinjection of fluorogold to the nucleus ambiguus retrogradely labeled a population of trigeminal ganglion neurons; some of which also contained amylin. In urethane-anesthetized rats, stereotaxic microinjections of amylin to the nucleus ambiguus caused a dose-dependent bradycardia that was reversibly attenuated by microinjections of the selective amylin receptor antagonist, salmon calcitonin (8-32) (sCT (8-32)) or AC187, and abolished by bilateral vagotomy. In an anesthetized rat diving model, diving bradycardia was attenuated by glutamate receptor antagonists CNQX and AP5, and was further suppressed by AC187. Whole-cel patch-clamp recordings from cardiac preganglionic vagal neurons revealed that amylin depolarizes neurons while decreasing conductance. Amylin also resulted in a reduction in whole cell currents, consistent with the decrease in conductance. Amylin is also found to increase excitability of neurons. In the presence of TTX, spontaneous currents in cardiac preganglionic vagal neurons were observed to decrease in frequency in response to amylin while amplitude remained constant, signifying that amylin reduces presynaptic activity at cardiac preganglionic vagal neurons. Finally, evoked synaptic currents revealed that amylin decreases evoked currents, further demonstrating that amylin depolarization and increase in excitability of cardiac preganglionic vagal neurons is also associated with simultaneous inhibition of presynaptic transmission. Our study has demonstrated for the first time that the bradycardia elicited by the diving reflex is mediated by amylin from trigeminal ganglion cells projecting to cardiac preganglionic neurons in the nucleus ambiguus. Additionally, amylin results in the depolarization and increased excitability of cardiac preganglionic vagal neurons while inhibiting presynaptic transmission. / Pharmacology
6

Urotensin II-Immunoreactivity in the Brainstem and Spinal Cord of the Rat

Dun, S. L., Brailoiu, G. C., Yang, J., Chang, J. K., Dun, N. J. 01 June 2001 (has links)
The distribution of urotensin-II-immunoreactivity (irU-II) was studied in the rat brainstem and spinal cord with the use of an antiserum against the human urotensin II (U-II) peptide. A population of ventral horn neurons in the spinal cord, hypoglossal nucleus, dorsal motor nucleus of the vagus, facial motor nucleus, nucleus ambiguus, abducens nucleus and trigeminal motor nucleus exhibited irU-II of varying intensities. The number of irU-II motor neurons was higher in the lumbar segments as compared to that of cervical, thoracic and sacral segments. Double-labeling the sections with U-II- and choline acetyltransferase (ChAT)-antisera revealed that nearly all irU-II ventral horn and brainstem neurons were ChAT-positive. The result provides the first immunohistochemical evidence of the presence of irU-II in cholinergic motoneurons of the rat spinal cord and brainstem.
7

Origins of Cardiac Vagal Preganglionic Fibers: A Retrograde Transport Study

Stuesse, Sherry L. 18 March 1982 (has links)
The origin of cardiac preganglionic neurons in the rat was investigated using the retrograde transport of horseradish peroxidase (HRP). A single injection of HRP was made into the right myocardium in either a sinoatrial or mid-ventricular location. Labeled cells were found in the mid- and lower medulla primarily in and around the nucleus ambiguus (NA) 600-1800 μm above the obex. The dorsal motor nucleus of X (DMN) was sparsely labeled and a few cells were found in an intermediate zone near the level of the obex. Labeling was bilateral with slightly heavier labeling found ipsilateral to the injection site than contralateral to it. Following a unilateral vagotomy, labeled cells were only found ipsilateral to the intact vagus. Atrial and midventricular injections yielded similar results. Occasionally only 1- cells in the NA were labeled per section. Inspection of serial sections revealed that in these sparsely labeled rats, the HRP was often in the same location within the NA forming a column of cells within the nucleus. The columns sometimes extended at least 240 μm in the rostral-caudal direction. The columnar organization was most apparent in rats with few labeled cells presumably because it was obscured in nuclei that were heavily labeled. In a second group of rats, the right vagus was cut at the cervical level and dipped in HRP to determine the extent of the NA and DMN in rats. In these animals, heavier labeling was found in the DMN than in the NA. Cells in the DMN were filled from the upper spinal cord to its most rostral extent 1200 μm above the obex. Thus, although the DMN and NA send projections in the vagus nerve, those axons terminating in the myocardium primarily originate in the NA.

Page generated in 0.0421 seconds