Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 89
Filter
1.
Psychophysiology ; 57(1): e13422, 2020 01.
Article in English | MEDLINE | ID: mdl-31206738

ABSTRACT

Overgeneralization (i.e., the transfer of fear to stimuli not related to an aversive event) is part of alterations in associative fear learning in mental disorders. In the present experimental study, we investigated whether this holds true for post-traumatic stress disorder (PTSD) related to childhood abuse. We expected that fear generalization under experimental conditions reflects generalization of aversive stimuli to different social domains in real life. Sixty-four women with PTSD after childhood abuse and 30 healthy participants (HC) underwent a differential fear conditioning and generalization paradigm. Online risk ratings, reaction time, and fear-potentiated startle served as dependent variables. Based on the subjectively assessed generalization of triggered intrusions across different domains of life, PTSD participants were split into two groups reporting low (low-GEN) and high (high-GEN) generalization. PTSD patients reported a higher expectation of an aversive event. During fear conditioning, they assessed the risk of danger related to a safety cue slower and showed a blunted fear-potentiated startle toward the danger cue. During generalization testing, reaction time increased in the high-GEN patients and decreased in the HC group with increasing similarity of a stimulus with the conditioned safety cue. Alterations of fear learning in PTSD suggest impaired defensive responses in case of a high threat probability. Moreover, our findings bridge the gap between the generalization of aversive cues during everyday life and laboratory-based experimental parameters: impairments in the processing of cues signaling safety generalize particularly in those patients who report a spreading of PTSD symptoms across different domains of everyday life.


Subject(s)
Adverse Childhood Experiences , Conditioning, Classical/physiology , Cues , Fear/physiology , Generalization, Psychological/physiology , Reflex, Startle/physiology , Stress Disorders, Post-Traumatic/physiopathology , Adult , Adult Survivors of Child Abuse , Female , Humans , Middle Aged , Stress Disorders, Post-Traumatic/etiology , Young Adult
2.
J Viral Hepat ; 23(7): 512-21, 2016 07.
Article in English | MEDLINE | ID: mdl-26891712

ABSTRACT

Hepatitis E virus (HEV) is the causative agent of hepatitis E in humans and a member of the genus Orthohepevirus in the family Hepeviridae. Infection usually leads to acute hepatitis that can become fulminant, particularly among pregnant women and in patients with preexisting liver disease, or may evolve to a chronic state, especially in immunosuppressed individuals. HEV has been shown to produce a range of extra-hepatic manifestations including aplastic anaemia, acute thyroiditis, glomerulonephritis as well as neurological disorders such as Guillain-Barré syndrome, neuralgic amyotrophy and encephalitis. The pathogenesis of these neurological injuries remains largely unknown, and it is also uncertain whether or not HEV can directly infect neuronal cells. In this study, we investigated whether HEV is capable of completing the viral life cycle in human neuronal-derived cell lines such as neuroepithelioma (SK-N-MC), desmoplastic cerebellar medulloblastoma (DAOY), glioblastoma multiforme (DBTRG), glioblastoma astrocytoma (U-373 MG) and oligodendrocytic (M03.13) cells. Following transfection of these cells with HEV Gaussia luciferase reporter virus, all tested cell lines supported HEV RNA replication. Furthermore, extra- and intracellular viral capsid was detected by an HEV antigen ELISA as a marker for virus assembly and release. Permissiveness for HEV cell entry could be demonstrated for the oligodendrocytic cell line M03.13. In conclusion, these results indicate that HEV tropism is not restricted to the liver and HEV can potentially complete the full viral life cycle in neuronal-derived tissues explaining neurologic disorders during HEV infection.


Subject(s)
Hepatitis E virus/physiology , Neuroglia/virology , Neurons/virology , Viral Tropism , Virus Replication , Cell Line , Humans
3.
Methods Inf Med ; 52(6): 514-21, 2013.
Article in English | MEDLINE | ID: mdl-23907324

ABSTRACT

OBJECTIVES: The OPTION scale ("observing patient involvement in decision making") assesses the extent to which clinicians involve patients in decisions across a range of situations in clinical practice. It so far just covers physician behavior. We intended to modify the scoring of the OPTION scale to incorporate active patient behavior in consultations. METHODS: Modification was done on scoring level, attempting a dyadic, relationship-centred approach in that high ratings can be evoked also by the behaviour of active patients. The German version of the OPTION scale was compared with a modified version by analysing video recordings of primary care consultations dealing with cardiovascular prevention. Fifteen general practitioners provided 40 videotaped consultations. Videos were analysed by two rater pairs and two experts in shared decision making (SDM). RESULTS: Reliability measures of the modified version were lower than those of the original scale. Significant associations of the dichotomised scale with the expert SDM rating as well as with physicians' expertise in SDM were only found for the modified OPTION scale. Receiver Operating Characteristic (ROC) analyses confirmed a valid differentiation between the presence of SDM (yes/no) on total score level, even though the cut-off point was quite low. Standard deviations of the single items in the modified version were higher compared to the original OPTION scale, while the means of total scores were similar. CONCLUSIONS: The original OPTION scale is physician-centered and neglects the activity and a possible self-involvement of the patient. Our modified instruction was able to capture the dyadic element partially. The development of a separate dyadic instrument might be more promising.


Subject(s)
Clinical Competence/statistics & numerical data , Cross-Cultural Comparison , Decision Making , Physician-Patient Relations , Referral and Consultation/statistics & numerical data , Adult , Cardiovascular Diseases/prevention & control , Clinical Competence/standards , Communication , Cross-Over Studies , Employee Performance Appraisal/statistics & numerical data , Female , Germany , Humans , Inservice Training , Male , Middle Aged , Observer Variation , Primary Health Care , Psychometrics/statistics & numerical data , Referral and Consultation/standards , Reproducibility of Results , Video Recording
4.
J Neuroendocrinol ; 25(7): 655-67, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23656518

ABSTRACT

Intranasal administration has been widely used to investigate the effects of the neuropeptides vasopressin and oxytocin on human behaviour and neurological disorders, although exactly what happens when these neuropeptides are administered intranasally is far from clear. In particular, it is not clear whether a physiological significant amount of peptide enters the brain to account for the observed effects. In the present study, we investigated whether the intranasal administration of vasopressin and oxytocin to rats induces the expression of the immediate-early gene product Fos in brain areas that are sensitive to centrally-administered peptide, whether it alters neuronal activity in the way that centrally-administered peptide does, and whether it affects behaviour in the ways that are expected from studies of centrally-administered peptide. We found that, whereas i.c.v. injection of very low doses of vasopressin or oxytocin increased Fos expression in several distinct brain regions, intranasal administration of large doses of the peptides had no significant effect. By contrast to the effects of vasopressin applied topically to the main olfactory bulb, we saw no changes in the electrical activity of olfactory bulb mitral cells after intranasal vasopressin administration. In addition, vasopressin given intranasally had no significant effects on social recognition or short-term recognition memory. Finally, intranasal infusions of vasopressin had no significant effects on the parameters monitored on the elevated plus maze, a rodent model of anxiety. Our data obtained in rats suggest that, after intranasal administration, significant amounts of vasopressin and oxytocin do not reach areas in the brain at levels sufficient to change immediate early gene expression, neural activity or behaviour in the ways described for central administration of the peptides.


Subject(s)
Behavior, Animal/drug effects , Gene Expression Regulation/drug effects , Genes, Immediate-Early , Vasopressins/administration & dosage , Administration, Intranasal , Animals , Male , Rats , Rats, Sprague-Dawley , Vasopressins/pharmacology
5.
J Neuroendocrinol ; 25(8): 711-8, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23656537

ABSTRACT

Adult male Brattleboro rats were used to investigate the impact of the congenital absence of vasopressin on the release pattern of oxytocin (OXT) within the hypothalamic supraoptic nucleus (SON) in response to a 10-min forced swimming session and osmotic stimulation. Both immunohistochemical and in situ hybridisation data suggest that vasopressin-deficient animals have more oxytocin-synthesising neurones in the SON than homozygous wild-type controls. Unexpectedly, both forced swimming and peripheral osmotic stimulation resulted in a blunted release profile of oxytocin within the SON of vasopressin-deficient rats compared to controls. A similar intranuclear OXT response to direct osmotic stimulation of the SON by retrodialysis with hypertonic Ringer's solution in both genotypes confirmed the capability of SON neurones to locally release oxytocin in vasopressin-deficient rats, indicating an altered processing of information originating from multisynaptic inputs rather than a deficit in release capacity. Taken together with data obtained in previous studies, the present findings provide evidence suggesting that autocrine and paracrine signalling of magnocellular neurones differs within the paraventricular nucleus and the SON. Thus, significant alterations in intra-SON oxytocin mRNA levels cannot easily be extrapolated to intranuclear release profiles and the local signal intensity of this neuropeptide after physiological stimulation.


Subject(s)
Hypothalamus, Anterior/metabolism , Neurons/metabolism , Oxytocin/biosynthesis , Animals , Hypothalamus, Anterior/cytology , Rats , Rats, Brattleboro , Swimming
6.
Amino Acids ; 44(3): 879-85, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23076252

ABSTRACT

Peptides of the corticotropin-releasing hormone/Urocortin (CRH/Ucn) family are known to suppress appetite primarily via CRH(2) receptors. In the rat hypothalamic supraoptic nucleus (SON), synthesis of both Ucn1 and CRH(2) receptors has been reported, yet little is known about the effects of Ucn1 in the SON on feeding behaviour. We first established the dose-related effects of Ucn1 injected into the SON on the feeding response in both freely fed and 24-h food-deprived rats. A conditioned taste avoidance paradigm was performed to investigate possible generalised effects of local Ucn1 treatment. Administration of Ucn1 into the SON at doses equal to or higher than 0.5 µg significantly decreased food intake in both freely fed and food-deprived rats. The Ucn1-mediated suppression of food intake was delayed in freely fed as compared to food-deprived animals. Conditioning for taste aversion to saccharine appeared at 0.5 and 1 µg of Ucn1. Both the early and the delayed onset of anorexia observed after intra-SON injection of Ucn1 under fasting and fed conditions, respectively, suggest the possible involvement of different CRH receptor subtypes in the two conditions, while the conditioned taste aversion seems to be responsible for the initial latency to eat the first meal in these animals.


Subject(s)
Appetite/drug effects , Eating/drug effects , Supraoptic Nucleus/drug effects , Urocortins/administration & dosage , Animals , Down-Regulation/drug effects , Feeding Behavior/drug effects , Food Deprivation , Hypothalamus/drug effects , Male , Rats , Rats, Wistar
7.
Amino Acids ; 35(1): 17-27, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18301960

ABSTRACT

Nitric oxide (NO) is a free radical gas synthesised from arginine and oxygen by enzymes of the family of the nitric oxide synthase. In particular, the neuronal nitric oxide synthase (nNOS) is highly expressed by cells of the hypothalamic paraventricular nucleus, where the sympatho-adrenal system, the hypothalamic-pituitary-adrenal axis and the hypothalamic-neurohypophyseal system originate. These structures are deputed to regulate the neuroendocrine stress response. In the past years, evidence has been accumulated to suggest that NO of nNOS origin plays a significant role in modulating the activity of the above mentioned systems under acute stressor exposure. The availability of nNOS knock-out mice allowed to investigate not only the physiological consequences of a constitutive lack of NO of nNOS origin at the hormonal and molecular level, but also to examine possible behavioural alterations. In this review, we shall discuss and confront the current trends of research in this area, especially focusing on the latest findings gained from genetically modified mice.


Subject(s)
Gene Expression Regulation, Enzymologic , Hypothalamo-Hypophyseal System/enzymology , Nitric Oxide Synthase Type I/metabolism , Paraventricular Hypothalamic Nucleus/enzymology , Pituitary-Adrenal System/enzymology , Stress, Physiological/enzymology , Animals , Behavior, Animal , Gene Expression Regulation, Enzymologic/genetics , Mice , Mice, Knockout , Nitric Oxide/biosynthesis , Nitric Oxide Synthase Type I/genetics , Stress, Physiological/genetics
8.
Stress ; 11(1): 42-51, 2008 Jan.
Article in English | MEDLINE | ID: mdl-17853069

ABSTRACT

The impact of a lifelong absence of the neuronal nitric oxide synthase (nNOS) in the neuroendocrine stress response was investigated in nNOS knockout (KO) and wild type (WT) mice under basal conditions and in response to forced swimming. In the hypothalamic paraventricular nucleus oxytocin and corticotropin-releasing-hormone mRNA levels did not differ between these genotypes under resting conditions, whereas vasopressin mRNA levels were significantly lower in nNOS KO than in WT animals. Also, in the adrenal glands basal levels of tyrosine hydroxylase protein, the rate-limiting enzyme for catecholamine biosynthesis, and of phenylethanolamine N-methyltransferase, which converts norepinephrine to epinephrine, were significantly reduced in nNOS KO mice. Plasma adrenocorticotropin, corticosterone, norepinephrine and epinephrine levels were similar in the KO and WT genotypes under resting conditions. In response to forced swimming, a similar increase in plasma adrenocorticotropin and corticosterone was observed in KO and WT animals. Stressor exposure triggered also an increased epinephrine release in WT animals, but did not significantly alter plasma epinephrine levels in KO mice. These data suggest that the chronic absence of nNOS reduces the capacity of epinephrine synthesising enzymes in the adrenal gland to respond to acute stressor exposure with an adequate epinephrine release.


Subject(s)
Adrenal Glands/enzymology , Arginine Vasopressin/metabolism , Catecholamines/biosynthesis , Nitric Oxide Synthase Type I/metabolism , Paraventricular Hypothalamic Nucleus/metabolism , Phenylethanolamine N-Methyltransferase/metabolism , Stress, Psychological/metabolism , Tyrosine 3-Monooxygenase/metabolism , Adrenocorticotropic Hormone/blood , Animals , Arginine Vasopressin/genetics , Blotting, Western , Catecholamines/blood , Corticosterone/metabolism , Corticotropin-Releasing Hormone , Down-Regulation , In Situ Hybridization , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Nitric Oxide Synthase Type I/deficiency , Nitric Oxide Synthase Type I/genetics , Oxytocin/metabolism , RNA, Messenger/metabolism
9.
Regul Pept ; 143(1-3): 34-8, 2007 Oct 04.
Article in English | MEDLINE | ID: mdl-17391780

ABSTRACT

Pregnancy is accompanied by an array of adaptive changes that play an important role in pre- and postnatal events. In rats, urocortin 1, a corticotropin-releasing factor-like peptide, is expressed mainly in the non-preganglionic Edinger-Westphal nucleus. We investigated the number of neurons immunoreactive for urocortin 1 at three different levels of the Edinger-Westphal nucleus in female rats by immunohistochemistry. The number of urocortin 1 immunoreactive cells was found to be decreased in pregnant rats compared to virgin rats. These results indicate that the hormonal status of the female rat affects urocortin 1 immunoreactive neurons in the non-preganglionic Edinger-Westphal nucleus and its signaling to target brain areas.


Subject(s)
Corticotropin-Releasing Hormone/metabolism , Mesencephalon/metabolism , Animals , Eating , Female , Immunohistochemistry , Male , Mesencephalon/cytology , Neurons/metabolism , Pregnancy , Rats , Rats, Wistar , Time Factors , Urocortins
10.
Nitric Oxide ; 16(1): 64-70, 2007 Feb.
Article in English | MEDLINE | ID: mdl-16769231

ABSTRACT

This study was undertaken to examine the importance of nitric oxide (NO) generated by the neural isoform of the nitric oxide synthase (nNOS) on the activity of the hypothalamic neurohypophyseal system in neural nitric oxide synthase knock-out (KO) and wild-type (WT) mice under basal conditions and in response to forced swimming. The intensity of the hybridisation signal for vasopressin (AVP) in the hypothalamic supraoptic nucleus (SON) was significantly higher in KO mice when compared with WT, whereas oxytocin (OXT) basal mRNA levels were similar in both groups. Although the basal peripheral release of AVP and OXT was equivalent in both genotypes, we observed in KO mice a significant drop of AVP and OXT plasma values 15 min after stressor onset and a robust increase in the OXT plasma concentration at 60 min. These findings suggest that in the male mouse, NO inhibits AVP gene transcription in magnocellular neurones of the SON and collaborates in maintaining constant AVP and OXT plasma levels following acute stressor exposure, exerting a bimodal regulatory action on OXT secretion. We conclude that NO is involved in the regulation of magnocellular neurones of the SON, and it is preferentially implicated in the attenuation of the peripheral release of OXT induced by acute stressor exposure.


Subject(s)
Gene Silencing , Nitric Oxide Synthase Type I/genetics , Oxytocin/blood , Swimming , Animals , Arginine Vasopressin/blood , Arginine Vasopressin/genetics , Base Sequence , DNA Primers , In Situ Hybridization , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Nitric Oxide Synthase Type I/physiology , Oxytocin/genetics , Oxytocin/metabolism , RNA, Messenger/genetics
11.
Cell Mol Life Sci ; 61(12): 1498-506, 2004 Jun.
Article in English | MEDLINE | ID: mdl-15197473

ABSTRACT

To identify neuronal substrates involved in NO/stress interactions we used Fos expression as a marker and examined the pattern of neuronal activation in response to swim stress in nNOS knock-out (nNOS-/-) and wild-type (WT) mice. Forced swimming enhanced Fos expression in WT and nNOS-/- mice in several brain regions, including cortical, limbic and hypothalamic regions. Differences in the Fos response between the two groups were observed in a limited set (6 out of 42) of these brain areas only: nNOS-/- mice displayed increased stressor-induced Fos expression in the medial amygdala, periventricular hypothalamic nucleus, supraoptic nucleus, CA1 field of the hippocampus, dentate gyrus and infralimbic cortex. No differences were observed in regions including the septum, central amygdala, periaqueductal grey and locus coeruleus. During forced swimming, nNOS-/- mice displayed reduced immobility duration, while no differences in general locomotor activity were observed between the groups in the home cage and during the open field test. The findings indicate that deletion of nNOS alters stress-coping ability during forced swimming and leads to an altered pattern of neuronal activation in response to this stressor in specific parts of the limbic system, hypothalamus and the medial prefrontal cortex.


Subject(s)
Brain/metabolism , Nitric Oxide Synthase/chemistry , Oncogene Proteins v-fos/biosynthesis , Animals , Brain/pathology , Corticosterone/blood , Immunohistochemistry , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , NADPH Dehydrogenase/metabolism , Neurons/metabolism , Nitric Oxide Synthase/antagonists & inhibitors , Nitric Oxide Synthase Type I , Swimming
12.
Amino Acids ; 26(1): 37-43, 2004 Feb.
Article in English | MEDLINE | ID: mdl-14752614

ABSTRACT

Neurons of the hypothalamo-neurohypophyseal system (HNS) are known to contain high amounts of neuronal nitric oxide (NO) synthase (nNOS). NO produced by those neurons is commonly supposed to be involved as modulator in the release of the two nonapeptides vasopressin (AVP) and oxytocin into the blood stream. Previous studies showed that forced swimming fails to increase the release of AVP into the blood stream while its secretion into the hypothalamus is triggered. We investigated here whether hypothalamically acting NO contributes to the control of the AVP release into blood under forced swimming conditions. Intracerebral microdialysis and in situ hybridization were employed to analyze the activity of the nitrergic system within the supraoptic nucleus (SON), the hypothalamic origin of the HNS. A 10-min forced swimming session failed to significantly alter the local NO release as indicated both by nitrite and, the main by-product of NO synthesis, citrulline levels in microdialysis samples collected from the SON. Microdialysis administration of NO directly into the SON increased the concentration of AVP in plasma samples collected during simultaneous forced swimming. In an additional experiment the effect of the defined stressor exposure on the concentration of mRNA coding for nNOS within the SON was investigated by in situ hybridization. Forced swimming increased the expression of nNOS mRNA at two and four hours after onset of the stressor compared to untreated controls. Taken together, our results imply that NO within the SON does not contribute to the regulation of the secretory activity of HNS neurons during acute forced swimming. Increased nNOS mRNA in the SON after forced swimming and the increase in AVP release in the presence of exogenous NO under forced swimming points to a possible role of NO in the regulation of the HNS under repeated stressor exposure.


Subject(s)
Nitrergic Neurons/metabolism , Nitric Oxide Synthase/metabolism , Nitric Oxide/blood , Supraoptic Nucleus/metabolism , Vasopressins/blood , Animals , Citrulline/blood , Citrulline/chemistry , Hypothalamo-Hypophyseal System/metabolism , Male , Oxytocin/blood , Oxytocin/metabolism , Physical Conditioning, Animal , Physical Exertion , Rats , Rats, Wistar , Supraoptic Nucleus/chemistry , Vasopressins/chemistry
13.
Anal Bioanal Chem ; 375(3): 460-4, 2003 Feb.
Article in English | MEDLINE | ID: mdl-12589514

ABSTRACT

Solid-phase micro extraction (SPME) and on-fiber derivatization followed by Gas Chromatography coupled with Flame Ionization Detection (GC-FID) or Selected Ion Monitoring Mass Spectrometry (GC-SIMMS) allows for simple yet sensitive quantification for the hexamethyldisilazane derivative of the beta-agonist clenbuterol. Using an 85- micro m polyacrylate fiber, the analysis method is optimized with respect to extraction time, derivatization time and temperature, and solution pH. In addition, the use of a rapid temperature ramping injection port allows for optimization of fiber desorption conditions. Under optimal conditions, the limits of detection for the hexamethyldisilazane derivative of clenbuterol are 1.1 ppb by FID and 0.20 ppb by SIMMS.


Subject(s)
Adrenergic beta-Agonists/analysis , Chromatography, Gas , Clenbuterol/analysis , Adrenergic beta-Agonists/chemistry , Clenbuterol/chemistry , Gas Chromatography-Mass Spectrometry , Methods , Sensitivity and Specificity , Solvents
14.
Acta Physiol Scand ; 176(2): 151-9, 2002 Oct.
Article in English | MEDLINE | ID: mdl-12354175

ABSTRACT

Clinical trials have demonstrated therapeutic benefit in inducing angiogenesis in chronic occlusive arterial disease. The route of application mostly used was the intramuscular injection of high dosages of plasmid. Therefore, a local perivascular application of low amounts of vascular endothelial growth factor (VEGF) plasmid was used in an interventional occlusion model, and the effect of VEGF on coronary and peripheral occlusions compared in the same animal model. Coronary and peripheral arteries were chronically occluded in Pietrain pigs using a non-surgical, interventional approach. Adventitial delivery of the DNA for VEGF was performed with a needle injection catheter. The DNA was applied as lipoplexes using the novel cationic liposomes DOCSPER. Optimized transfer conditions were used. Angiography, polymerase chain reaction (PCR), reverse transcriptase-polymerase chain reaction (RT-PCR) and immunohistochemistry were undertaken within a follow-up period of 6 months. Expression of the transfected VEGF gene was observed at 1 and 3 weeks following application. The DNA was detected up to 5 months following application. Around occluded coronary arteries, there was formation of new collaterals and arterial prolongation, whereas surrounding occluded peripheral arteries there was no collateralization but development of new arterial branches was seen. Results demonstrate that the response to VEGF is also sufficient, when minimal amounts of plasmid encoding for VEGF are applied locally into the perivasculature allowing for more safety of this therapy. Comparison of treatment of chronic coronary and peripheral arterial disease revealed differences in angiogenesis following VEGF application during a total follow-up period of almost 6 months which may be related to their different developmental origins. This may have important implications for developing future therapeutic strategies using VEGF in different vessels.


Subject(s)
Arterial Occlusive Diseases/therapy , Coronary Stenosis/therapy , Endothelial Growth Factors/administration & dosage , Intercellular Signaling Peptides and Proteins/administration & dosage , Lymphokines/administration & dosage , Neovascularization, Physiologic/drug effects , Animals , Arterial Occlusive Diseases/diagnostic imaging , DNA/administration & dosage , DNA/analysis , Endothelial Growth Factors/genetics , Genetic Therapy/methods , Genetic Vectors , Iliac Artery , Intercellular Signaling Peptides and Proteins/genetics , Lymphokines/genetics , Models, Biological , Neovascularization, Physiologic/genetics , Plasmids/administration & dosage , Plasmids/genetics , Radiography , Swine , Transfection , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors
15.
Eur J Neurosci ; 14(7): 1047-55, 2001 Oct.
Article in English | MEDLINE | ID: mdl-11683896

ABSTRACT

Previous experiments have shown that a 10-min forced swimming session triggers the release of vasopressin from somata and dendrites, but not axon terminals, of neurons of the hypothalamic-neurohypophysial system. To further investigate regulatory mechanisms underlying this dissociated release, we forced male Wistar rats to swim in warm (20 degrees C) water and monitored release of the potentially inhibitory amino acids gamma amino butyric acid (GABA) and taurine into the hypothalamic supraoptic nucleus using microdialysis. Forced swimming caused a significant increase in the release of taurine (up to 350%; P < 0.05 vs. prestress release), but not GABA. To reveal the physiological significance of centrally released taurine, the specific taurine antagonist 6-aminomethyl-3-methyl-4H-1,2,4-benzothiadiazine-1,1-dioxide was administered into the supraoptic nucleus via retrodialysis. Administration of this antagonist caused a significant increase in the release of vasopressin within the supraoptic nucleus and into the blood both under basal conditions and during stress (up to 800%; P < 0.05 vs. basal values), without affecting hypothalamic or plasma oxytocin. Local administration of the GABA(A) receptor antagonist bicuculline, in contrast, failed to influence vasopressin secretion at either time point. In a separate series of in vivo electrophysiological experiments, administration of the same dosage of the taurine antagonist into the supraoptic nucleus via microdialysis resulted in an increased electrical activity of identified vasopressinergic, but not oxytocinergic, neurons. Taken together our data demonstrate that taurine is released within the supraoptic nucleus during physical/emotional stress. Furthermore, at the level of the supraoptic nucleus, taurine inhibits not only the electrical activity of vasopressin neurons but also acts as an inhibitor of both central and peripheral vasopressin secretion during different physiological states.


Subject(s)
Neural Inhibition/physiology , Neurons/metabolism , Stress, Physiological/metabolism , Supraoptic Nucleus/metabolism , Taurine/metabolism , Vasopressins/metabolism , gamma-Aminobutyric Acid/metabolism , Action Potentials/drug effects , Action Potentials/physiology , Animals , Aspartic Acid/metabolism , Benzothiadiazines/pharmacology , Bicuculline/pharmacology , Consciousness/physiology , GABA Antagonists/pharmacology , GABA-A Receptor Antagonists , Glutamic Acid/metabolism , Male , Neural Inhibition/drug effects , Oxytocin/blood , Oxytocin/metabolism , Rats , Rats, Wistar , Receptors, GABA-A/metabolism , Stress, Physiological/physiopathology , Supraoptic Nucleus/drug effects , Swimming/physiology , Taurine/antagonists & inhibitors , Vasopressins/blood
16.
J Endovasc Ther ; 8(4): 401-7, 2001 Aug.
Article in English | MEDLINE | ID: mdl-11552732

ABSTRACT

PURPOSE: To determine the optimal endovascular approach to achieve long-term occlusion of large arteries, while preserving the integrity of periarterial tissue, in an animal model of ischemia. METHODS: Femoral artery occlusions were created in 16 pigs using detachable balloons, coils, or blinded stent-grafts. Feasibility, safety, primary and long-term success, and the degree of neovascularization were determined over a 6-month period by serial angiography and histological analyses. Four animals served as untreated controls. RESULTS: Overall primary success for all occlusion devices was 100%. The 6-month occlusion rate using detachable balloons or coils was 33% and 0%, respectively; however, all arteries occluded with blinded stent-grafts remained obstructed to the end of the study. There was no significant difference in capillary densities and collateralization of periarterial areas when occluded arteries were compared with nonoccluded controls in the same animal. No increase in collateralization was observed following endovascular arterial occlusion. CONCLUSIONS: Percutaneous insertion of blinded stent-grafts easily, safely, and reliably creates long-term arterial occlusion in pigs, which may make this a more appropriate model for studying the effects of angiogenic factors in vivo.


Subject(s)
Arterial Occlusive Diseases/surgery , Femoral Artery/surgery , Vascular Surgical Procedures , Anatomy, Cross-Sectional , Animals , Catheterization , Disease Models, Animal , Extremities/blood supply , Femoral Artery/anatomy & histology , Femoral Artery/diagnostic imaging , Follow-Up Studies , Radiography , Stents , Swine , Time , Time Factors , Vascular Surgical Procedures/instrumentation
18.
Eur J Neurosci ; 13(12): 2273-81, 2001 Jun.
Article in English | MEDLINE | ID: mdl-11454031

ABSTRACT

Previous studies have shown that a 10-min forced swimming session triggers the release of both vasopressin and oxytocin into the extracellular fluid of the hypothalamic paraventricular (PVN) and supraoptic nuclei (SON) in rats. At the same time oxytocin, but not vasopressin, was released from the axon terminals into the blood. Here we combined forced swimming with in situ hybridization to investigate whether (i) the stressor-induced release of vasopressin and oxytocin within the PVN originates from parvo- or magnocellular neurons of the nucleus, and (ii) central release with or without concomitant peripheral secretion is followed by changes in the synthesis of vasopressin and/or oxytocin. Adult male Wistar rats were killed 2, 4 or 8 h after a 10-min forced swimming session and their brains processed for in situ hybridization using 35S-labelled oligonucleotide probes. As measured on photo-emulsion-coated slides, cellular vasopressin mRNA concentration increased in magnocellular PVN neurons 2 and 4 h after swimming (P < 0.05). Similarly, oxytocin mRNA concentration was significantly increased in magnocellular neurons of the PVN at 2 and 8 h (P < 0.05). We failed to observe significant effects on vasopressin and oxytocin mRNA levels in the parvocellular PVN and in the SON. Taken together with results from previous studies, our data suggest that magnocellular neurons are the predominant source of vasopressin and oxytocin released within PVN in response to forced swimming. Furthermore, in the case of vasopressin, central release in the absence of peripheral secretion is followed by increased mRNA levels, implying a refill of depleted somato-dendritic vasopressin stores. Within the SON, however, mRNA levels are poor indicators of the secretory activity of magnocellular neurons during stress.


Subject(s)
Neurons/metabolism , Oxytocin/metabolism , Paraventricular Hypothalamic Nucleus/metabolism , Stress, Physiological/metabolism , Swimming/physiology , Vasopressins/metabolism , Animals , In Situ Hybridization , Male , Neurons/cytology , Oxytocin/genetics , Paraventricular Hypothalamic Nucleus/cytology , RNA, Messenger/metabolism , Rats , Rats, Wistar , Stress, Physiological/pathology , Stress, Physiological/physiopathology , Supraoptic Nucleus/cytology , Supraoptic Nucleus/metabolism , Time Factors , Vasopressins/genetics
19.
J Microencapsul ; 18(4): 491-506, 2001.
Article in English | MEDLINE | ID: mdl-11428678

ABSTRACT

Long-term benefits of coronary angioplasty remain limited by the treatment-induced renarrowing of arteries, termed restenosis. One of the mechanisms leading to restenosis is the proliferation of smooth muscle cells. Therefore, proliferating cells of the injured arterial wall, which can be selectively transduced by retroviruses, are potential targets for gene therapy strategies. A direct single-dose therapeutic application of retroviral vectors for inhibition of cell proliferation is normally limited by too low transduction efficiencies. Encapsulated retrovirus-producing cells release viral vectors from microcapsules, and may enhance the transduction efficiency by prolonged infection. Primary and immortal murine and porcine cells and murine retrovirus-producing cells were encapsulated in cellulose sulphate. Cell viability was monitored by analysing cell metabolism. Safety, stability, transfer efficiency and extent of restenosis using capsules were determined in a porcine restenosis model for local gene therapy using morphometry, histology, in situ beta-galactosidase assay and PCR. Encapsulation of cells did not impair cell viability. Capsules containing retrovirus-producing cells expressing the beta-galactosidase reporter gene were implanted into periarterial tissue or a pig model of restenosis. Three weeks following implantation, beta-galactosidase activity was detected in the pericapsular tissue with a transduction efficiency of approximately 1 in 500 cells. Adventitial implantation of vector-producing encapsulated cells for gene therapy may, therefore, facilitate successful targeting of proliferating vascular smooth muscle cells, and allow stable integration of therapeutic genes into surrounding cells. The encapsulation of vector-producing cells could represent a novel and feasible way to optimize local retroviral gene therapy.


Subject(s)
Cell Transplantation/methods , Cellulose/analogs & derivatives , Drug Compounding/methods , Gene Transfer Techniques , Retroviridae/genetics , 3T3 Cells , Animals , Base Sequence , Cell Division , Cells, Cultured , Coronary Restenosis/pathology , Coronary Restenosis/therapy , DNA Primers/genetics , Genes, Reporter , Genetic Vectors , Mice , Muscle, Smooth, Vascular/pathology , Swine , beta-Galactosidase/genetics
20.
Eur J Neurosci ; 13(6): 1254-6, 2001 Mar.
Article in English | MEDLINE | ID: mdl-11285023

ABSTRACT

A single social defeat in male rats has long lasting physiological and behavioural consequences, which are similar to those seen in depressive patients. In addition, the housing conditions after social defeat appear to be crucial for the development of depression-like symptoms. Because the dopaminergic system is thought to be altered in depressive illness, we investigated the impact of individual and group housing on the temporal development of changes of dopamine transporter (DAT) binding in male rats after a single social defeat. The number of striatal DAT binding sites was reduced in animals that remained isolated after being defeated. The isolation length after social defeat amplified this effect, indicating a temporal development of the changes on the striatal DAT. In animals which returned to the familiar group after social defeat the density of striatal DAT binding sites was not affected. We conclude that social isolation after a single defeat reduces the number of DAT binding sites. In contrast, a familiar environment after a single social defeat appears to prevent the stress-induced alterations on the dopaminergic system. This finding suggests that housing conditions are critical when investigating the central nervous effects of social defeat in male rats.


Subject(s)
Carrier Proteins/antagonists & inhibitors , Corpus Striatum/metabolism , Dominance-Subordination , Membrane Glycoproteins , Membrane Transport Proteins , Nerve Tissue Proteins , Social Isolation , Animals , Carrier Proteins/metabolism , Dopamine Plasma Membrane Transport Proteins , Male , Rats , Rats, Wistar
SELECTION OF CITATIONS
SEARCH DETAIL
...