Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 50
Filter
1.
Science ; 381(6655): 285-290, 2023 07 21.
Article in English | MEDLINE | ID: mdl-37471539

ABSTRACT

Disruption of the physiologic sleep-wake cycle and low melatonin levels frequently accompany cardiac disease, yet the underlying mechanism has remained enigmatic. Immunostaining of sympathetic axons in optically cleared pineal glands from humans and mice with cardiac disease revealed their substantial denervation compared with controls. Spatial, single-cell, nuclear, and bulk RNA sequencing traced this defect back to the superior cervical ganglia (SCG), which responded to cardiac disease with accumulation of inflammatory macrophages, fibrosis, and the selective loss of pineal gland-innervating neurons. Depletion of macrophages in the SCG prevented disease-associated denervation of the pineal gland and restored physiological melatonin secretion. Our data identify the mechanism by which diurnal rhythmicity in cardiac disease is disturbed and suggest a target for therapeutic intervention.


Subject(s)
Circadian Rhythm , Heart Diseases , Macrophages , Melatonin , Pineal Gland , Sleep Disorders, Circadian Rhythm , Superior Cervical Ganglion , Animals , Humans , Mice , Heart Diseases/physiopathology , Melatonin/metabolism , Pineal Gland/pathology , Pineal Gland/physiopathology , Sleep , Sleep Disorders, Circadian Rhythm/physiopathology , Superior Cervical Ganglion/pathology , Superior Cervical Ganglion/physiopathology , Macrophages/immunology , Fibrosis
2.
Auton Neurosci ; 224: 102641, 2020 03.
Article in English | MEDLINE | ID: mdl-32044642

ABSTRACT

Neurons in the superior cervical ganglia (SCG) are classified as rostral and caudal according to their regional locations. Although diverse phenotypes have been reported for these two subpopulations, differences in neuroplasticity, like long-term potentiation (LTP), have not been characterized. Here, we explored possible regional differences of LTP expression in rostral and caudal neurons of the SCG in control rats, Wistar and Wistar Kyoto (WKy), and in the spontaneously hypertensive rats (SHR) as a model of hypertension. We characterized the expression of gLTP evoked by a tetanic train (40 Hz, 3 s) in an in vitro SCG preparation. gLTP was recorded in rostral and caudal neurons at 8-weeks-old (wo) in Wistar rats, 6-wo and 12-wo in SHR and WKy rats. We found that gLTP was differentially expressed; gLTP was larger in caudal neurons in Wistar and adult WKy rats. In adult 12-wo hypertensive SHR, gLTP was expressed in caudal but not in rostral neurons. In contrast, in 6-wo pre-hypertensive SHR, gLTP was expressed in rostral but not in caudal neurons; while in 6-wo WKy, gLTP was expressed in caudal but not in rostral neurons. The lack of gLTP expression in caudal neurons of 6-wo SHR was not due to a GABAergic modulation because several GABA-A receptor antagonists failed to unmask gLTP. Data show that neuroplasticity, particularly gLTP expression, varied according to the ganglionic region. We propose that differential regional expression of gLTP may be correlated with selective innervation on different target organs.


Subject(s)
Ganglia, Sympathetic/drug effects , Long-Term Potentiation/drug effects , Neurons/metabolism , Superior Cervical Ganglion/metabolism , Animals , GABA-A Receptor Antagonists/pharmacology , Ganglia, Sympathetic/metabolism , Male , Neurons/drug effects , Rats, Inbred SHR , Rats, Wistar , Superior Cervical Ganglion/physiopathology
3.
Circ Res ; 126(6): 708-721, 2020 03 13.
Article in English | MEDLINE | ID: mdl-31928179

ABSTRACT

RATIONALE: Effector memory T lymphocytes (TEM cells) exacerbate hypertension in response to repeated hypertensive stimuli. These cells reside in the bone marrow for prolonged periods and can be reactivated on reexposure to the hypertensive stimulus. OBJECTIVE: Because hypertension is associated with increased sympathetic outflow to the bone marrow, we hypothesized that sympathetic nerves regulate accumulation and reactivation of bone marrow-residing hypertension-specific TEM cells. METHODS AND RESULTS: Using unilateral superior cervical ganglionectomy in wild-type C57BL/6 mice, we showed that sympathetic nerves create a bone marrow environment that supports residence of hypertension-specific CD8+ T cells. These cells, defined by their proliferative response on coculture with dendritic cells from Ang (angiotensin) II-infused mice, were reduced in denervated compared with innervated bone of Ang II-infused mice. Adoptively transferred CD8+ T cells from Ang II-infused mice preferentially homed to innervated compared with denervated bone. In contrast, ovalbumin responsive T cells from OT-I mice did not exhibit this preferential homing. Increasing superior cervical ganglion activity by activating Gq-coupled designer receptor exclusively activated by designer drug augmented CD8+ TEM bone marrow accumulation. Adoptive transfer studies using mice lacking ß2AR (ß2 adrenergic receptors) indicate that ß2AR in the bone marrow niche, rather than T-cell ß2AR is critical for TEM cell homing. Inhibition of global sympathetic outflow using Gi-coupled DREADD (designer receptor exclusively activated by designer drug) injected into the rostral ventrolateral medulla or treatment with a ß2AR antagonist reduced hypertension-specific CD8+ TEM cells in the bone marrow and reduced the hypertensive response to a subsequent response to low dose Ang II. CONCLUSIONS: Sympathetic nerves contribute to the homing and survival of hypertension-specific TEM cells in the bone marrow after they are formed in hypertension. Inhibition of sympathetic nerve activity and ß2AR blockade reduces these cells and prevents the blood pressure elevation and renal inflammation on reexposure to hypertension stimuli.


Subject(s)
Bone Marrow/innervation , CD8-Positive T-Lymphocytes/physiology , Cell Movement , Hypertension/physiopathology , Superior Cervical Ganglion/physiopathology , Adoptive Transfer , Adrenergic beta-2 Receptor Antagonists/pharmacology , Angiotensin II/pharmacology , Animals , Bone Marrow/immunology , CD8-Positive T-Lymphocytes/immunology , Denervation , Hypertension/immunology , Medulla Oblongata/drug effects , Medulla Oblongata/physiopathology , Mice , Mice, Inbred C57BL , Receptors, Adrenergic, beta-2/metabolism , Superior Cervical Ganglion/drug effects
4.
Brain Res ; 1729: 146599, 2020 02 15.
Article in English | MEDLINE | ID: mdl-31843626

ABSTRACT

Diabetic neuropathy is the most prevalent complication associated with diabetes mellitus (DM). The superior cervical ganglion (SCG) is an important sympathetic component of the autonomic nervous system. We investigated the changes in cellular electrophysiological properties and on Na+K+-ATPase activity of SCG neurons of rats with DM induced by streptozotocin (STZ). Three types of action potentials (AP) firing pattern were observed in response to a long (1 s) depolarizing pulse. Whilst some neurons fired a single AP (single firing phasic, SFP), others fired few APs (multiple firing phasic, MFP). A third type fired APs during more than 80% of the stimulus duration (tonic-like, TL). The occurrence of SFP, MFP and TL was 84.5, 13.8, and 1.7%, respectively. SFP and MFP differed significantly in their membrane input resistance (Rin). At the end of the 4th week of its time course, DM differently affected most types of neurons: DM induced depolarization of resting membrane potential (RMP), decreased AP amplitude in SFP, and decreased Rin in MFP. DM decreased spike after-hyperpolarization amplitude in MFP and the duration in SFP. Based on the RMP depolarization, we investigated the Na+K+-ATPase action and observed that DM caused a significant decrease in Na+K+-ATPase activity of SCG. In conclusion, we have demonstrated that DM affects several parameters of SCG physiology in a manner likely to have pathophysiological relevance.


Subject(s)
Action Potentials/physiology , Diabetic Neuropathies/physiopathology , Neurons/physiology , Superior Cervical Ganglion/physiopathology , Animals , Diabetes Mellitus, Experimental/physiopathology , Electrophysiological Phenomena , Female , Male , Rats , Rats, Wistar
5.
Cephalalgia ; 39(13): 1700-1709, 2019 Nov.
Article in English | MEDLINE | ID: mdl-31370669

ABSTRACT

PURPOSE: The pineal gland plays an important role in biological rhythms, circadian and circannual variations, which are key aspects in several headache disorders. OVERVIEW: Melatonin, the main pineal secreting hormone, has been extensively studied in primary and secondary headache disorders. Altered melatonin secretion occurs in many headache syndromes. Experimental data show pineal gland and melatonin both interfere in headache animal models, decreasing trigeminal activation. Melatonin has been shown to regulate CGRP and control its release. DISCUSSION: Melatonin has been used successfully as a treatment for migraine, cluster headaches and other headaches. There is a rationale for including the pineal gland as a relevant brain structure in the mechanisms of headache pathophysiology, and melatonin as a treatment option in primary headache.


Subject(s)
Headache/physiopathology , Pineal Gland/physiopathology , Adult , Animals , Calcitonin Gene-Related Peptide/physiology , Case-Control Studies , Child , Circadian Rhythm/physiology , Clinical Trials as Topic , Disease Models, Animal , Double-Blind Method , Headache/diagnostic imaging , Headache/drug therapy , Headache/pathology , Humans , Melatonin/physiology , Melatonin/therapeutic use , Oxidation-Reduction , Paraventricular Hypothalamic Nucleus/physiopathology , Pineal Gland/metabolism , Pineal Gland/pathology , Receptors, Melatonin/agonists , Receptors, Melatonin/physiology , Serotonin/metabolism , Superior Cervical Ganglion/physiopathology
6.
Turk Neurosurg ; 29(3): 434-439, 2019.
Article in English | MEDLINE | ID: mdl-30984987

ABSTRACT

AIM: To examine ischemic neurodegeneration of the ciliospinal center on permanent miosis following subarachnoid hemorrhage (SAH). MATERIAL AND METHODS: Nineteen rabbits were examined in this study. The animals were divided into three groups, as control (GI, n=5), sham (GII, n=5) and study group (GIII, n=9). Pupil diameters were measured after giving 0.5 mL physiological saline for sham and autologous arterial blood for the study group into the cervico-thoracic subarachnoid space. After three weeks of follow up, the cervico-thoracic cord and bilateral superior cervical sympathetic ganglia were removed. The pupil diameter values were compared with degenerated neuron volumes of sympathetic ganglia and degenerated neuron densities of thoracic sympathetic nuclei which were studied by stereological methods. RESULTS: The mean pupil diameter was 5180 ± 370 µm and the mean degenerated neuron density of the ciliospinal center was 4 ± 1/mm3 in animals of the control group (GI). These values were 9850 ± 610 εm, 10 ± 3/mm3 in sham (GII), and 7.010 ± 440 εm and 98 ± 21/mm3 in the study (GIII) groups. There was an inverse relationship between degenerated neuron density of the ciliospinal nuclei and pupil diameters. CONCLUSION: We showed and reported for the first time that ciliospinal sympathetic center ischemia-induced neurodegeneration may have been responsible for permanent miosis following SAH.


Subject(s)
Ischemia/diagnosis , Miosis/diagnosis , Subarachnoid Hemorrhage/diagnosis , Superior Cervical Ganglion/pathology , Animals , Disease Models, Animal , Ischemia/complications , Ischemia/physiopathology , Male , Miosis/etiology , Miosis/physiopathology , Nerve Degeneration/diagnosis , Nerve Degeneration/etiology , Nerve Degeneration/physiopathology , Pupil/physiology , Rabbits , Random Allocation , Subarachnoid Hemorrhage/complications , Subarachnoid Hemorrhage/physiopathology , Superior Cervical Ganglion/physiopathology
7.
Cardiovasc Res ; 114(2): 291-299, 2018 02 01.
Article in English | MEDLINE | ID: mdl-29186414

ABSTRACT

Aims: Cardiac inflammation has been suggested to be regulated by the sympathetic nervous system (SNS). However, due to the lack of methodology to surgically eliminate the myocardial SNS in mice, neuronal control of cardiac inflammation remains ill-defined. Here, we report a procedure for local cardiac sympathetic denervation in mice and tested its effect in a mouse model of heart failure post-myocardial infarction. Methods and results: Upon preparation of the carotid bifurcation, the right and the left superior cervical ganglia were localized and their pre- and postganglionic branches dissected before removal of the ganglion. Ganglionectomy led to an almost entire loss of myocardial sympathetic innervation in the left ventricular anterior wall. When applied at the time of myocardial infarction (MI), cardiac sympathetic denervation did not affect acute myocardial damage and infarct size. In contrast, cardiac sympathetic denervation significantly attenuated chronic consequences of MI, including myocardial inflammation, myocyte hypertrophy, and overall cardiac dysfunction. Conclusion: These data suggest a critical role for local sympathetic control of cardiac inflammation. Our model of myocardial sympathetic denervation in mice should prove useful to further dissect the molecular mechanisms underlying cardiac neural control.


Subject(s)
Ganglionectomy , Heart Failure/prevention & control , Heart Ventricles/innervation , Myocardial Infarction/complications , Myocarditis/prevention & control , Myocardium , Superior Cervical Ganglion/surgery , Animals , Heart Failure/immunology , Heart Failure/pathology , Heart Failure/physiopathology , Heart Ventricles/immunology , Heart Ventricles/metabolism , Heart Ventricles/pathology , Humans , Male , Mice, Inbred C57BL , Myocarditis/immunology , Myocarditis/pathology , Myocarditis/physiopathology , Myocardium/immunology , Myocardium/metabolism , Myocardium/pathology , Neuroimmunomodulation , Superior Cervical Ganglion/physiopathology , Ventricular Function, Left
8.
Turk Neurosurg ; 28(1): 48-55, 2018.
Article in English | MEDLINE | ID: mdl-27593832

ABSTRACT

AIM: To investigate the relationship between neuron density of the superior cervical sympathetic ganglia and pupil diameter in subarachnoid hemorrhage. MATERIAL AND METHODS: This study was conducted on 22 rabbits; 5 for the baseline control group, 5 for the SHAM group and 12 for the study group. Pupil diameters were measured via sunlight and ocular tomography on day 1 as the control values. Pupil diameters were re-measured after injecting 0.5 cc saline to the SHAM group, and autologous arterial blood into the cisterna magna of the study group. After 3 weeks, the brain, superior cervical sympathetic ganglia and ciliary ganglia were extracted with peripheral tissues bilaterally and examined histopathologically. Pupil diameters were compared with neuron densities of the sympathetic ganglia and ciliary ganglia which were examined using stereological methods. RESULTS: Baseline values were; normal pupil diameter 7.180±620 ?m and mean neuron density of the superior cervical sympathetic ganglia 6.321±510/mm3, degenerated neuron density of ciliary ganglia was 5±2/mm3 after histopathological examination in the control group. These values were measured as 6.850±578 ?m, 5.950±340/mm3 and 123±39/mm3 in the SHAM group and 9.910±840 ?m, 7.950±764/mm3 and 650±98/mm3 in the study group. A linear relationship was determined between neuron density of the superior cervical sympathetic ganglia and pupil diameters (p < 0.005). Degenerated ciliary ganglia neuron density had an inverse effect on pupil diameters in all groups (p < 0.0001). CONCLUSION: Highly degenerated neuron density of the ciliary ganglion is not responsible for pupil dilatation owing to parasympathetic pupilloconstrictor palsy, but high neuron density of the pupillodilatatory superior cervical sympathetic ganglia should be considered an important factor for pupil dilatation.


Subject(s)
Disease Models, Animal , Mydriasis/pathology , Pupil/physiology , Subarachnoid Hemorrhage/pathology , Superior Cervical Ganglion/pathology , Animals , Cisterna Magna/pathology , Cisterna Magna/physiopathology , Ganglia, Parasympathetic/pathology , Ganglia, Parasympathetic/physiopathology , Male , Mydriasis/physiopathology , Nerve Degeneration/pathology , Nerve Degeneration/physiopathology , Neurons/pathology , Neurons/physiology , Rabbits , Subarachnoid Hemorrhage/physiopathology , Superior Cervical Ganglion/physiopathology
10.
Ann Vasc Surg ; 34: 243-9, 2016 Jul.
Article in English | MEDLINE | ID: mdl-27116906

ABSTRACT

BACKGROUND: Hyperhidrosis is secondary to over activation of the sympathetic nervous system and surgical sympathectomy is the treatment of choice when other modalities have failed. This study investigated anatomic variation in the upper thoracic sympathetic chain and associated rami communicantes among cadaveric specimens. It considers the implications of these findings on surgical techniques to treat hyperhidrosis. METHODS: The upper 4 thoracic sympathetic ganglia, intercostal nerves, and connecting rami were dissected, measured and mapped in 40 sides of 20 adult human cadavers. Ganglia location was recorded. The incidence, orientation, and distance travelled by rami communicantes was compared across different ganglionic levels and between sides. RESULTS: The percentage of ganglia located below their associated intercostal space was 6.25% with stellate ganglions present in 70% of specimens and Kuntz fibers noted in 40%. There was a stepwise reduction in incidence of rami from superior to inferior placed ganglia. The number of rami identified across all levels was significantly greater on the right (P = 0.03). The horizontal distance between the sympathetic chain and union of the rami on the intercostal nerves was significantly greater on the right across all levels (P = 0.04). CONCLUSIONS: There was substantial variation in the rami communicantes across the upper 4 ganglia and between right and left sides. Consideration of this variation should be given when planning surgical sympathectomy for hyperhidrosis particularly to avoid symptom recurrence.


Subject(s)
Hyperhidrosis/surgery , Stellate Ganglion/abnormalities , Stellate Ganglion/surgery , Superior Cervical Ganglion/abnormalities , Superior Cervical Ganglion/surgery , Sympathectomy/methods , Anatomic Landmarks , Cadaver , Dissection , Female , Humans , Hyperhidrosis/diagnosis , Hyperhidrosis/physiopathology , Intercostal Nerves/anatomy & histology , Male , Stellate Ganglion/physiopathology , Superior Cervical Ganglion/physiopathology
11.
Hypertension ; 63(2): 309-15, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24324044

ABSTRACT

It has been recognized that the sympathetic nervous system is activated in pulmonary arterial hypertension (PAH), and abnormal sympathetic hyperactivity leads to worsening of PAH via endothelial dysfunction. The purpose of this study was to examine whether sympathetic ganglion block (SGB) can treat PAH by increasing the availability of nitric oxide (NO). PAH was induced in rats by 50 mg/kg of subcutaneous monocrotaline. After 2 weeks, daily injections of ropivacaine into the left superior cervical ganglion were repeated for 14 days (monocrotaline-SGB group). Monocrotaline group received sham SGB with saline, whereas control group received saline instead of monocrotaline. PAH was evident in monocrotaline group, with right ventricular systolic pressures (47±4 mm Hg) that were higher than those of controls (17±2 mm Hg), whereas SGB significantly attenuated monocrotaline-induced PAH (35±4 mm Hg). The right/left ventricular mass ratios exhibited similar changes to those seen with right ventricular pressures. Heart rate variability showed significantly higher sympathetic activity in the monocrotaline group. Microscopy revealed a higher proportion of muscular arteries with thicker medial walls in the monocrotaline group, which was attenuated by SGB. Monocrotaline induced arginase hyperactivity, which was in turn decreased by SGB-induced endothelial NO synthase activation. SGB restored monocrotaline-induced hypoactivity of superoxide dismutase. In conclusion, SGB could suppress PAH and the remodeling of pulmonary arteries via inactivation of arginase and reciprocal elevation of NO bioavailability, thus attenuating disproportionate hyperactivation of the sympathetic nervous system.


Subject(s)
Amides/pharmacology , Autonomic Nerve Block/methods , Ganglia, Sympathetic/drug effects , Hypertension, Pulmonary/drug therapy , Monocrotaline/pharmacology , Superior Cervical Ganglion/drug effects , Anesthetics, Local/pharmacology , Animals , Arginase/antagonists & inhibitors , Arginase/metabolism , Blood Pressure/drug effects , Blood Pressure/physiology , Cardiomegaly/drug therapy , Cardiomegaly/metabolism , Cardiomegaly/physiopathology , Ganglia, Sympathetic/metabolism , Ganglia, Sympathetic/physiopathology , Hypertension, Pulmonary/metabolism , Male , Nitric Oxide/metabolism , Nitric Oxide Synthase Type III/metabolism , Oxidative Stress/drug effects , Oxidative Stress/physiology , Rats , Rats, Sprague-Dawley , Ropivacaine , Superior Cervical Ganglion/metabolism , Superior Cervical Ganglion/physiopathology
12.
J Mol Neurosci ; 51(3): 763-70, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23832486

ABSTRACT

The majority of diabetics develop serious disorders of the autonomic nervous system; however, there is no clear understanding on the causes of these complications. In this study, we examined the effect of streptozocin (STZ)-induced diabetes on activity-dependent synaptic plasticity, associated levels of brain-derived neurotrophic factor (BDNF) and antioxidant biomarkers in the rat sympathetic superior cervical ganglion. Diabetes (STZ-induced) was achieved by a single intraperitoneal injection of streptozocin (55 mg/kg).Compound action potentials were recorded from isolated ganglia before (basal) and after repetitive stimulation, or trains of paired pulses to express ganglionic long-term potentiation (gLTP) or long-term depression (gLTD). The input/output curves of ganglia from STZ-treated animals showed a marked rightward shift along most stimulus intensities, compared to those of ganglia from control animals, indicating impaired basal synaptic transmission in ganglia from STZ-induced diabetic animals. Repetitive stimulation induced robust gLTP and gLTD in ganglia isolated from control animals; the same protocols failed to induce gLTP or gLTD in ganglia from STZ-induced diabetic animals, indicating impairment of activity-dependent synaptic plasticity in these animals. Molecular analysis revealed significant reduction in the levels of BDNF and the ratio of glutathione/oxidized glutathione. Additionally, the activity of glutathione peroxidase, glutathione reductase, catalase, and the levels of thiobarbituric acid-reactive substances were increased in ganglia from STZ-treated animals. In conclusion, impaired basal synaptic transmission and synaptic plasticity are associated with reduced BDNF and altered oxidative stress biomarkers in the sympathetic ganglia from STZ-induced diabetic animals, suggesting a possible correlation of these factors with the manifestations of STZ-induced diabetes in the peripheral nervous system.


Subject(s)
Brain-Derived Neurotrophic Factor/metabolism , Diabetes Mellitus, Experimental/metabolism , Long-Term Potentiation , Oxidative Stress , Superior Cervical Ganglion/metabolism , Action Potentials , Animals , Diabetes Mellitus, Experimental/physiopathology , Glutathione/metabolism , Male , Rats , Rats, Wistar , Superior Cervical Ganglion/physiopathology
13.
Eur Ann Otorhinolaryngol Head Neck Dis ; 130(5): 269-73, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23890788
14.
Brain ; 135(Pt 9): 2613-28, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22826602

ABSTRACT

Patients with small fibre neuropathy typically manifest pain in distal extremities and severe autonomic dysfunction. However, occasionally patients present with minimal autonomic symptoms. The basis for this phenotypic difference is not understood. Sodium channel Na(v)1.7, encoded by the SCN9A gene, is preferentially expressed in the peripheral nervous system within sensory dorsal root ganglion and sympathetic ganglion neurons and their small diameter peripheral axons. We recently reported missense substitutions in SCN9A that encode functional Na(v)1.7 variants in 28% of patients with biopsy-confirmed small fibre neuropathy. Two patients with biopsy-confirmed small fibre neuropathy manifested minimal autonomic dysfunction unlike the other six patients in this series, and both of these patients carry the Na(v)1.7/R185H variant, presenting the opportunity to compare variants associated with extreme ends of a spectrum from minimal to severe autonomic dysfunction. Herein, we show by voltage-clamp that R185H variant channels enhance resurgent currents within dorsal root ganglion neurons and show by current-clamp that R185H renders dorsal root ganglion neurons hyperexcitable. We also show that in contrast, R185H variant channels do not produce detectable changes when studied by voltage-clamp within sympathetic neurons of the superior cervical ganglion, and have no effect on the excitability of these cells. As a comparator, we studied the Na(v)1.7 variant I739V, identified in three patients with small fibre neuropathy characterized by severe autonomic dysfunction as well as neuropathic pain, and show that this variant impairs channel slow inactivation within both dorsal root ganglion and superior cervical ganglion neurons, and renders dorsal root ganglion neurons hyperexcitable and superior cervical ganglion neurons hypoexcitable. Thus, we show that R185H, from patients with minimal autonomic dysfunction, does not produce detectable changes in the properties of sympathetic ganglion neurons, while I739V, from patients with severe autonomic dysfunction, has a profound effect on excitability of sympathetic ganglion neurons.


Subject(s)
Autonomic Nervous System Diseases/genetics , Ganglia, Spinal/physiopathology , NAV1.7 Voltage-Gated Sodium Channel/genetics , Neuralgia/genetics , Neurons/physiology , Polyneuropathies/genetics , Superior Cervical Ganglion/physiopathology , Aged , Alleles , Autonomic Nervous System Diseases/diagnosis , Autonomic Nervous System Diseases/physiopathology , Female , Ganglia, Spinal/cytology , Gene Frequency , Genetic Variation , Genotype , Humans , Male , Middle Aged , Neuralgia/diagnosis , Neuralgia/physiopathology , Neurons/cytology , Polyneuropathies/diagnosis , Polyneuropathies/physiopathology , Superior Cervical Ganglion/cytology , Young Adult
15.
Curr Alzheimer Res ; 8(4): 377-84, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21453246

ABSTRACT

Basal synaptic transmission and activity-dependent synaptic plasticity were evaluated in superior cervical sympathetic ganglia (SCG) of amyloid-ß rat model of Alzheimer's disease (Aß rat) using electrophysiological and molecular techniques. Rats were administered Aß peptides (a mixture of 1:1 Aß1-40 and Aß1-42) by chronic intracerebroventricular infusion via 14-day mini-osmotic pumps (300 pmol/day). Control rats received Aß40-1 (inactive reverse peptide: 300 pmol/day). Ganglionic compound action potentials were recorded before (basal) and after repetitive stimulation. In isolated SCG, ganglionic long-term potentiation (gLTP) was generated by a brief train of stimuli (20Hz for 20s) and ganglionic long-term depression (gLTD) was produced with trains of paired pulses. The input/output (I/O) curves of ganglia from Aß rats showed a marked downward shift along all stimulus intensities, compared to those of ganglia from control animals, indicating impaired basal synaptic transmission. In addition, repetitive stimulation induced robust gLTP and gLTD in ganglia isolated from control animals, but, the same protocols failed to induce gLTP or gLTD in ganglia from Aß rats indicating impairment of activity-dependent synaptic plasticity in these animals. Western blotting of SCG homogenate from Aß rats revealed reduction in the ratio of phosphorylated-/total-CaMKII and in calcineurin protein levels. Although other mechanisms could be involved, these changes in signaling molecules could represent an important molecular mechanism linked to the failure to express synaptic plasticity in Aß rat ganglia. Results of the current study could explain some of the peripheral nervous system manifestations of Alzheimer's disease.


Subject(s)
Alzheimer Disease/chemically induced , Alzheimer Disease/pathology , Amyloid beta-Peptides/toxicity , Neuronal Plasticity/physiology , Superior Cervical Ganglion/pathology , Synaptic Transmission/physiology , Animals , Biophysics , Calcineurin/metabolism , Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Disease Models, Animal , Electric Stimulation/methods , Evoked Potentials/drug effects , Evoked Potentials/physiology , Gene Expression Regulation/drug effects , Long-Term Potentiation/drug effects , Long-Term Synaptic Depression/drug effects , Male , Neuronal Plasticity/drug effects , Rats , Rats, Wistar , Signal Transduction/drug effects , Superior Cervical Ganglion/physiopathology , Synaptic Transmission/drug effects
16.
Neurochem Int ; 56(3): 516-21, 2010 Feb.
Article in English | MEDLINE | ID: mdl-20036294

ABSTRACT

Axon degeneration is a common hallmark of many neurodegenerative diseases, and the underlying mechanism remains largely unknown. Lysosomes are involved in some neurodegenerative diseases, including Alzheimer's disease and Parkinson's disease. Whether lysosomes are involved in axon degeneration is yet to be elucidated. In this study, we found only about 10% lysosomes remained in axons of cultured superior cervical ganglia (SCGs) after transection for 4h when stained with LysoTracker. Furthermore, we found that lysosomal disruption occurred earlier than morphological changes and loss of mitochondrial membrane potential. In addition, the well-known axon-protective protein Wld(S) delayed injury-induced axon degeneration from both morphological changes and lysosomal disruption. Lysosomal inhibitors including chloroquine and ammonium chloride induced axon degeneration in cultured SCGs, and Wld(S) also slowed down the axon degeneration induced by lysosomal inhibitors. All these data suggest that lysosomal disruption is an early marker of axon degeneration, and inhibition of lysosome induces axon degeneration in a Wld(S)-protectable way. Thus, maintenance of normal lysosomal function might be an important approach to delay axon degeneration in neurodegenerative diseases.


Subject(s)
Axons/pathology , Lysosomes/pathology , Neurodegenerative Diseases/pathology , Neurodegenerative Diseases/physiopathology , Wallerian Degeneration/pathology , Wallerian Degeneration/physiopathology , Amines , Ammonium Chloride/toxicity , Animals , Animals, Newborn , Axons/metabolism , Cells, Cultured , Chloroquine/pharmacology , Chloroquine/toxicity , Coloring Agents , Lysosomes/metabolism , Membrane Potential, Mitochondrial/physiology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Mitochondria/metabolism , Mitochondria/pathology , Neurodegenerative Diseases/metabolism , Neuroprotective Agents/pharmacology , Rats , Rats, Sprague-Dawley , Superior Cervical Ganglion/metabolism , Superior Cervical Ganglion/pathology , Superior Cervical Ganglion/physiopathology , Time Factors , Wallerian Degeneration/chemically induced
17.
Neurochem Int ; 56(1): 101-6, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19778564

ABSTRACT

Axon degeneration is an active program of self-destruction observed in many physiological and pathological settings. There are three Nicotinamide mononucleotide adenylyl transferase (Nmnat, EC2.7.7.1) in mammals. Overexpression of Nmnat1 or Nmnat3 can delay axon degeneration, while the role of Nmnat2 in axon degeneration remains largely unknown. Here we found that Nmnat2 was specifically and highly expressed in brain compared with Nmnat1 and Nmnat3. Furthermore, we found brain Nmnat2 was correlated with Alzheimer's disease in APPswe/PS1dE9 transgenic mice. Nmnat2 delayed Wallerian degeneration in cultured superior cervical ganglia (SCGs) from morphological changes, microtubule destruction and neurofilament degradation, mutation of the conserved enzyme activity site in Nmnat2 disrupted its enzyme activity as well as the axon-protective function. Our results demonstrate that the brain-specific Nmnat2 delays injury-induced axon degeneration dependent on its NAD synthesis activity. These findings provide new clues to further study the molecular mechanisms of axon degeneration and the related neurodegenerative diseases.


Subject(s)
Axons/metabolism , NAD/biosynthesis , Nicotinamide-Nucleotide Adenylyltransferase/metabolism , Superior Cervical Ganglion/metabolism , Wallerian Degeneration/metabolism , Animals , Axons/pathology , Cell Death/physiology , Cell Line , Cells, Cultured , Cytoprotection/physiology , Humans , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microtubules/metabolism , Neurofilament Proteins/metabolism , Nicotinamide-Nucleotide Adenylyltransferase/genetics , Rats , Rats, Sprague-Dawley , Superior Cervical Ganglion/pathology , Superior Cervical Ganglion/physiopathology , Time Factors , Wallerian Degeneration/genetics , Wallerian Degeneration/physiopathology
18.
Neurobiol Aging ; 31(5): 805-12, 2010 May.
Article in English | MEDLINE | ID: mdl-18649973

ABSTRACT

Sustained increase in central sympathetic outflow to ganglia may provide the repeated high frequency presynaptic activity required for induction of long-term potentiation in sympathetic ganglia (gLTP), which is known to be involved in the manifestation of a neurogenic form of hypertension, namely stress-hypertension. Aging is often viewed as a progressive decline in physiological competence with a corresponding impaired ability to adapt to stressful stimuli. Old animals have exaggerated sympathetic activity as well as increased morbidity and mortality during prolonged exposure to stressful stimuli. Using the superior cervical ganglion (SCG) as a model for sympathetic ganglia, electrophysiological and biochemical evidence show that mildly hypertensive aged rats (22-month old) have expressed gLTP in vivo. This is suggested by a number of lines of evidence. Firstly, a shift in input/output (I/O) curve of ganglia from aged rats to the left side of I/O curve of ganglia from 6-month old (adult) rats indicating expression of gLTP. Secondly, failure of in vitro high frequency stimulation to induce gLTP in ganglia isolated from aged rats, which indicates occlusion due to saturation, which, in turn, suggests in vivo expression of gLTP in these ganglia. Thirdly, in vitro inhibition of basal ganglionic transmission by blockers of gLTP (5-HT(3) antagonists) is observed in ganglia isolated from aged rats, but not in those from adult rats. Finally, immunoblot analysis revealed that protein levels of signaling molecules such as calcium-calmodulin kinase II (CaMKII; phosphorylated and total), which normally increase during expression of LTP, are elevated in ganglia isolated from aged rats compared to those from adult ones. Protein levels of calcineurin, which dephosphorylates P-CaMKII, were reduced in ganglia isolated from aged rats, probably as a support mechanism to allow prolonged phosphorylation of CaMKII. Our findings suggest in vivo expression of gLTP in sympathetic ganglia of aged animals, which may contribute to the moderate hypertension often seen in aged subjects.


Subject(s)
Aging/physiology , Hypertension/physiopathology , Long-Term Potentiation/physiology , Superior Cervical Ganglion/physiopathology , Aging/metabolism , Animals , Blood Pressure/physiology , Blotting, Western , Calcineurin/metabolism , Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Electric Stimulation , Electrophysiology , Hypertension/metabolism , Long-Term Potentiation/drug effects , Male , Ondansetron/pharmacology , Phosphorylation/physiology , Rats , Receptors, Serotonin, 5-HT3/metabolism , Serotonin Antagonists/pharmacology , Signal Transduction/physiology , Superior Cervical Ganglion/drug effects , Superior Cervical Ganglion/metabolism
19.
Med Hypotheses ; 73(3): 410-3, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19409714

ABSTRACT

Cervical spinal cord stimulation (SCS) has been for many years hypothesized to be of use in treatment of cerebral vasospasm after subarachnoid hemorrhage. Experiments in animals and research in humans have demonstrated increase in cerebral blood flow (CBF), and different theories have been tried to explain these observations. Although there are many claims of circulatory improvements in these circumstances, no clinical application has yet been established. A complete understanding of physiological and anatomic correlation between CBF modulation and SCS remain unclear. We strongly believe that the main objective is not to treat vasospasm by increasing blood flow but to prevent vasoconstriction of the cerebral arteries by a functional sympathectomy. SCS may, at least theoretically, work in different ways at the same time: (1) preventing vasoconstriction of cerebral arteries by functional sympathectomy, acting at the lower cervical levels; and (2) increasing CBF through central pathways, perhaps involving brainstem connections, at the upper cervical levels. One of the practical implications of this hypothesis would be differential placement of cervical spinal cord stimulation electrodes in patients with subarachnoid hemorrhage depending on the timing of electrode insertion and presence or absence of vasospasm at the time of initial intervention.


Subject(s)
Brain/physiopathology , Cervical Vertebrae , Electric Stimulation Therapy/methods , Models, Neurological , Spinal Cord/physiopathology , Superior Cervical Ganglion/physiopathology , Sympathetic Nervous System/physiopathology , Vasospasm, Intracranial/prevention & control , Vasospasm, Intracranial/physiopathology , Humans
20.
Neurosci Res ; 63(2): 95-9, 2009 Feb.
Article in English | MEDLINE | ID: mdl-19038294

ABSTRACT

Previous work from this laboratory indicated that superior cervical ganglia from rats exposed to chronic psychosocial stress expressed ganglionic long-term potentiation (gLTP) in vivo. In the present study, we report additional pharmacological evidence indicating involvement of calmodulin and guanylyl cyclase in gLTP, and supporting the in vivo gLTP expression in ganglia from chronically stressed rats. Pretreatment with the calmodulin inhibitors W-7 (5 microM) or calmidazolium (5 microM) or with guanylyl cyclase inhibitor LY-83583 (5 microM) completely blocked HFS (20 Hz/20s)-induced gLTP in superior cervical ganglia isolated from normal rats. Along with that, inhibition of apparent basal ganglionic transmission by W-7 (5 microM), calmidazolium (5 microM) or LY-83583 (5 microM) is observed in ganglia isolated from chronically stressed rats, but not in those from control rats, indicating in vivo expression of gLTP in ganglia isolated from stressed rats. The present results confirm the involvement of both calmodulin and GC activities in gLTP, and indicate that ganglia from stressed rats may have expressed gLTP in vivo, which is known to precipitate hypertension in these animals.


Subject(s)
Enzyme Inhibitors/pharmacology , Imidazoles/pharmacology , Long-Term Potentiation/drug effects , Stress, Psychological/pathology , Sulfonamides/pharmacology , Superior Cervical Ganglion/drug effects , Aminoquinolines/pharmacology , Animals , Calmodulin/antagonists & inhibitors , Disease Models, Animal , Electric Stimulation/methods , Guanylate Cyclase/antagonists & inhibitors , Long-Term Potentiation/physiology , Male , Rats , Rats, Wistar , Superior Cervical Ganglion/physiopathology
SELECTION OF CITATIONS
SEARCH DETAIL
...