Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 132
Filter
Add more filters

Country/Region as subject
Publication year range
1.
J Physiol ; 592(24): 5507-27, 2014 Dec 15.
Article in English | MEDLINE | ID: mdl-25362150

ABSTRACT

We examined the hypotheses that: (1) during incremental exercise and recovery following 4-6 days at high altitude (HA) global cerebral blood flow (gCBF) increases to preserve cerebral oxygen delivery (CDO2) in excess of that required by an increasing cerebral metabolic rate of oxygen ( CM RO2); (2) the trans-cerebral exchange of oxygen vs. carbohydrates (OCI; carbohydrates = glucose + ½lactate) would be similar during exercise and recovery at HA and sea level (SL). Global CBF, intra-cranial arterial blood velocities, extra-cranial blood flows, and arterial-jugular venous substrate differences were measured during progressive steady-state exercise (20, 40, 60, 80, 100% maximum workload (Wmax)) and through 30 min of recovery. Measurements (n = 8) were made at SL and following partial acclimatization to 5050 m. At HA, absolute Wmax was reduced by ∼50%. During submaximal exercise workloads (20-60% Wmax), despite an elevated absolute gCBF (∼20%, P < 0.05) the relative increases in gCBF were not different at HA and SL. In contrast, gCBF was elevated at HA compared with SL during 80 and 100% Wmax and recovery. Notwithstanding a maintained CDO2 and elevated absolute CM RO2 at HA compared with SL, the relative increase in CM RO2 was similar during 20-80% Wmax but half that of the SL response (i.e. 17 vs. 27%; P < 0.05 vs. SL) at 100% Wmax. The OCI was reduced at HA compared with SL during 20, 40, and 60% Wmax but comparable at 80 and 100% Wmax. At HA, OCI returned almost immediately to baseline values during recovery, whereas at SL it remained below baseline. In conclusion, the elevations in gCBF during exercise and recovery at HA serve to maintain CDO2. Despite adequate CDO2 at HA the brain appears to increase non-oxidative metabolism during exercise and recovery.


Subject(s)
Altitude , Brain/metabolism , Carbohydrate Metabolism , Cerebrovascular Circulation , Exercise , Oxygen Consumption , Adult , Brain/blood supply , Brain/physiology , Humans , Male
2.
Cereb Cortex ; 22(6): 1442-54, 2012 Jun.
Article in English | MEDLINE | ID: mdl-21878486

ABSTRACT

Psychological stress contributes to the development of clinical depression. This has prompted many preclinical studies to investigate the neurobiology of this relationship, however, the effects of stress on glia remain unclear. In this study, we wished to determine, first, how exposure to chronic psychological stress affects microglial activity within the prefrontal cortex (PFC) and, second, whether the observed changes were meaningfully related to corresponding changes in local neuronal activity and PFC-regulated behavior. Therefore, we examined markers of microglial activation, antigen presentation, apoptosis, and persistent neuronal activation within the PFC after exposure to repeated restraint stress. We also examined the effect of stress on spatial working memory, a PFC-dependent function. Finally, we tested the ability of a microglial activation inhibitor (minocycline) to alter the impact of chronic stress on all of these endpoints. Stressor exposure produced positively correlated increases in microglial and long-term neuronal activation in the PFC but not antigen presentation or apoptosis. As expected, it also impaired spatial working memory. Importantly, minocycline reduced the impact of stress on neuronal activation and working memory, as well as microglial activation. These results suggest a role for microglia in mediating the effects of stress on PFC neuronal function and PFC-regulated behavior.


Subject(s)
Maze Learning/physiology , Microglia/physiology , Prefrontal Cortex/physiology , Stress, Psychological/physiopathology , Animals , Chronic Disease , Male , Microglia/pathology , Prefrontal Cortex/pathology , Rats , Rats, Sprague-Dawley , Stress, Psychological/pathology
3.
J Physiol ; 590(14): 3261-75, 2012 Jul 15.
Article in English | MEDLINE | ID: mdl-22495584

ABSTRACT

Despite the importance of blood flow on brainstem control of respiratory and autonomic function, little is known about regional cerebral blood flow (CBF) during changes in arterial blood gases.We quantified: (1) anterior and posterior CBF and reactivity through a wide range of steady-state changes in the partial pressures of CO2 (PaCO2) and O2 (PaO2) in arterial blood, and (2) determined if the internal carotid artery (ICA) and vertebral artery (VA) change diameter through the same range.We used near-concurrent vascular ultrasound measures of flow through the ICA and VA, and blood velocity in their downstream arteries (the middle (MCA) and posterior (PCA) cerebral arteries). Part A (n =16) examined iso-oxic changes in PaCO2, consisting of three hypocapnic stages (PaCO2 =∼15, ∼20 and ∼30 mmHg) and four hypercapnic stages (PaCO2 =∼50, ∼55, ∼60 and ∼65 mmHg). In Part B (n =10), during isocapnia, PaO2 was decreased to ∼60, ∼44, and ∼35 mmHg and increased to ∼320 mmHg and ∼430 mmHg. Stages lasted ∼15 min. Intra-arterial pressure was measured continuously; arterial blood gases were sampled at the end of each stage. There were three principal findings. (1) Regional reactivity: the VA reactivity to hypocapnia was larger than the ICA, MCA and PCA; hypercapnic reactivity was similar.With profound hypoxia (35 mmHg) the relative increase in VA flow was 50% greater than the other vessels. (2) Neck vessel diameters: changes in diameter (∼25%) of the ICA was positively related to changes in PaCO2 (R2, 0.63±0.26; P<0.05); VA diameter was unaltered in response to changed PaCO2 but yielded a diameter increase of +9% with severe hypoxia. (3) Intra- vs. extra-cerebral measures: MCA and PCA blood velocities yielded smaller reactivities and estimates of flow than VA and ICA flow. The findings respectively indicate: (1) disparate blood flow regulation to the brainstem and cortex; (2) cerebrovascular resistance is not solely modulated at the level of the arteriolar pial vessels; and (3) transcranial Doppler ultrasound may underestimate measurements of CBF during extreme hypoxia and/or hypercapnia.


Subject(s)
Brain/blood supply , Hypercapnia/blood , Hypocapnia/blood , Hypoxia/blood , Adult , Blood Flow Velocity/physiology , Blood Gas Analysis , Carotid Artery, Internal/diagnostic imaging , Cerebral Arteries/diagnostic imaging , Female , Humans , Hypercapnia/diagnostic imaging , Hypocapnia/diagnostic imaging , Hypoxia/diagnostic imaging , Male , Regional Blood Flow/physiology , Ultrasonography, Doppler, Transcranial , Vasoconstriction/physiology , Vasodilation/physiology , Vertebral Artery/diagnostic imaging
4.
Cancer Gene Ther ; 13(8): 739-45, 2006 Aug.
Article in English | MEDLINE | ID: mdl-16543918

ABSTRACT

In this study, we investigated the in vitro and in vivo efficacy of Fas ligand (FasL) gene therapy for the treatment of head and neck cancer. Three head and neck squamous cell carcinoma (HNSCC) cell lines (SCC-1, SCC-12, and SCC-14a) were treated with the Fas agonist CH-11, a monoclonal antibody to the Fas receptor, or with a replication-incompetent adenovirus (AdGFPFasL) expressing a modified murine Fas ligand gene fused to green fluorescent protein (GFP). A replication-incompetent adenovirus containing the GFP gene alone was used as a control for viral transduction toxicity (AdGFP). Cell death was quantified using a tetrazolium-based (MTS) assay. Cells were analyzed by flow cytometry to determine the expression of adenoviral and Fas receptors on the surface of the cells. Our results showed that the head and neck cancer cell lines are resistant to cell death induction when treated with the anti-Fas monoclonal antibody CH-11. This resistance can be overcome with AdGFPFasL, which was able to induce cell death in all three cell lines. Apoptosis induction was demonstrated using Western blotting by evaluating poly(ADP-ribose) polymerase, and caspase 9 cleavages. In addition, intratumoral injections of AdGFPFasL into SCC-14a xenografts induced significant growth suppression of tumors, indicating that FasL gene therapy may provide a new efficient therapeutic modality for HNSCC that is worthy of a clinical trial.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Antibodies/therapeutic use , Carcinoma, Squamous Cell/therapy , Genetic Therapy/methods , Head and Neck Neoplasms/therapy , Membrane Glycoproteins/genetics , Tumor Necrosis Factors/genetics , Adenoviridae/genetics , Animals , Apoptosis/genetics , Bystander Effect , Carcinoma, Squamous Cell/genetics , Cell Line, Tumor , Fas Ligand Protein , Genetic Vectors , Green Fluorescent Proteins/genetics , Head and Neck Neoplasms/genetics , Humans , Membrane Glycoproteins/immunology , Mice , Tumor Necrosis Factors/immunology , Xenograft Model Antitumor Assays , fas Receptor/immunology
5.
Oral Oncol ; 42(9): 854-65, 2006 Oct.
Article in English | MEDLINE | ID: mdl-16798060

ABSTRACT

Despite many advances in surgical techniques, technology, radiation therapy, and chemotherapy, survival rates for head and neck cancer (HNCa) have not improved significantly in decades, with many patients being diagnosed at advanced disease stages. Adequate assessment of oral cavity malignancies is critical for appropriate planning of surgical, radiation, and chemotherapy treatment. Imaging modalities used to evaluate the oral cavity include plain radiography (panoramic radiography and intraoral radiography), nuclear medicine scintigraphy, ultrasound (US), computed tomography (CT), magnetic resonance imaging (MRI), and positron emission tomography (PET). This review describes these imaging techniques and their utility, primarily CT and MRI.


Subject(s)
Mouth Neoplasms/diagnosis , Humans , Magnetic Resonance Imaging , Radionuclide Imaging , Tomography, X-Ray Computed
6.
Neuropharmacology ; 41(5): 574-81, 2001 Oct.
Article in English | MEDLINE | ID: mdl-11587712

ABSTRACT

This study examined if brain pathways in morphine-dependent rats are activated by opioid withdrawal precipitated outside the central nervous system. Withdrawal precipitated with a peripherally acting quaternary opioid antagonist (naloxone methiodide) increased Fos expression but caused a more restricted pattern of neuronal activation than systemic withdrawal (precipitated with naloxone which enters the brain). There was no effect on locus coeruleus and significantly smaller increases in Fos neurons were produced in most other areas. However in the ventrolateral medulla (A1/C1 catecholamine neurons), nucleus of the solitary tract (A2/C2 catecholamine neurons), lateral parabrachial nucleus, supramamillary nucleus, bed nucleus of the stria terminalis, accumbens core and medial prefrontal cortex no differences in the withdrawal treatments were detected. We have shown that peripheral opioid withdrawal can affect central nervous system pathways.


Subject(s)
Brain/metabolism , Morphine , Narcotics , Peripheral Nervous System/metabolism , Substance Withdrawal Syndrome/metabolism , Animals , Brain/drug effects , Brain/pathology , Genes, fos/physiology , Male , Morphine/adverse effects , Naloxone/antagonists & inhibitors , Naloxone/pharmacology , Narcotic Antagonists/pharmacology , Narcotics/adverse effects , Neurons/drug effects , Neurons/metabolism , Neurons/pathology , Peripheral Nervous System/drug effects , Peripheral Nervous System/pathology , Rats , Rats, Wistar , Substance Withdrawal Syndrome/pathology
7.
J Neuroimmunol ; 152(1-2): 20-32, 2004 Jul.
Article in English | MEDLINE | ID: mdl-15223234

ABSTRACT

Using Fos immunolabelling as a marker of neuronal activation, we investigated the role of the parabrachial nucleus in generating central neuronal responses to the systemic administration of the proinflammatory cytokine interleukin-1beta (1 microg/kg, i.a.). Relative to intact animals, parabrachial nucleus lesions significantly reduced the number of Fos-positive cells observed in the central amygdala (CeA), the bed nucleus of the stria terminalis (BNST), and the ventrolateral medulla (VLM) after systemic interleukin-1beta. In a subsequent experiment in which animals received parabrachial-directed deposits of a retrograde tracer, it was found that many neurons located in the nucleus tractus solitarius (NTS) and the VLM neurons were both retrogradely labelled and Fos-positive after interleukin-1beta administration. These results suggest that the parabrachial nucleus plays a critical role in interleukin-1beta-induced Fos expression in CeA, BNST and VLM neurons and that neurons of the NTS and VLM may serve to trigger or at least influence changes in parabrachial nucleus activity that follows systemic interleukin-1beta administration.


Subject(s)
Interleukin-1/immunology , Neurons/metabolism , Pons/metabolism , Proto-Oncogene Proteins c-fos/metabolism , Animals , Brain/drug effects , Brain/immunology , Brain/metabolism , Ibotenic Acid/toxicity , Immunization , Interleukin-1/pharmacology , Male , Neurons/drug effects , Neurons/immunology , Pons/drug effects , Pons/injuries , Proto-Oncogene Proteins c-fos/drug effects , Rats , Rats, Wistar
8.
Neuroscience ; 58(4): 765-75, 1994 Feb.
Article in English | MEDLINE | ID: mdl-8190253

ABSTRACT

Noxious somatic stimuli elicit vasopressin secretion, an effect thought to result from activation of a facilitatory input from A1 catecholamine cells of the medulla oblongata. To better characterize the A1 cell response and effects on other neuroendocrine A1 projection targets, particularly within the paraventricular nucleus, we have now mapped c-fos expression in neurochemically identified catecholamine and neurosecretory cells following a noxious somatic stimulus. Unilateral hind paw pinch significantly increased c-fos expression in contralateral A1 cells whereas other brainstem catecholamine cell groups were unaffected. Expression of c-fos was also increased in the supraoptic nucleus, this effect being more pronounced for vasopressin than oxytocin neurosecretory cells and, as with A1 cells, primarily on the side contralateral to the stimulated paw. In contrast, the increase in the paraventricular nucleus was greater in oxytocin rather than in vasopressin cells. Additionally there was a significant rise in c-fos expression in medial parvocellular paraventricular nucleus cells of noxiously stimulated animals. Notably, the majority of tuberoinfundibular corticotropin-releasing factor cells are located in this medial parvocellular zone. These results are consistent with and expand on those previously reported from electrophysiological and anatomical studies. The finding of differing neurosecretory cell responses between supraoptic and paraventricular nuclei has interesting implications with regard to the afferent control of neurosecretory cell activity. For example, the substantially greater activation of supraoptic versus paraventricular nucleus vasopressin cells, despite being innervated by the same medullary noradrenergic cell group, raises the possibility of a differential input or differences in responsiveness. Furthermore, the activation of paraventricular nucleus parvocellular cells is consistent with suggestions that the A1 cell group provides an excitatory input to this population.


Subject(s)
Brain Stem/metabolism , Catecholamines/metabolism , Gene Expression/physiology , Genes, fos/physiology , Hypothalamus/metabolism , Neurosecretory Systems/metabolism , Animals , Brain Stem/cytology , Histocytochemistry , Male , Pain/physiopathology , Paraventricular Hypothalamic Nucleus/metabolism , Physical Stimulation , Proto-Oncogene Proteins c-fos/immunology , Proto-Oncogene Proteins c-fos/metabolism , Rats , Rats, Wistar , Supraoptic Nucleus/metabolism , Vasopressins/metabolism
9.
Neuroscience ; 94(1): 175-83, 1999.
Article in English | MEDLINE | ID: mdl-10613507

ABSTRACT

In the present study we examined the role of the central nucleus of the amygdala in hypothalamic-pituitary-adrenal axis responses to an immune challenge in the form of systemic administration of the proinflammatory cytokine interleukin-1beta (1 microg/kg). We found that bilateral ibotenic acid lesions of the central amygdala substantially reduced adrenocorticotropin hormone release and hypothalamic corticotropin-releasing factor and oxytocin cell c-fos expression responses to interleukin-1,8 suggesting a facilitatory role for this structure in the generation of hypothalamic-pituitary-adrenal axis responses to an immune challenge. Since only a small number of central amygdala cells project directly to the paraventricular nucleus, we then examined the effect of central amygdala lesions on the activity of other brain nuclei that might act as relay sites in the control of the hypothalamic-pituitary-adrenal axis function. We found that bilateral central amygdala lesions significantly reduced interleukin-1beta-induced c-fos expression in cells of the ventromedial and ventrolateral subdivisions of the bed nucleus of the stria terminalis and brainstem catecholamine cell groups of the nucleus tractus solitarius (A2 noradrenergic cells) and ventrolateral medulla (A1 noradrenergic and C1 adrenergic cells). These findings, in conjunction with previous evidence of bed nucleus of the stria terminalis and catecholamine cell group involvement in hypothalamic-pituitary-adrenal axis regulation, suggest that ventromedial and ventrolateral bed nucleus of the stria terminalis cells and medullary catecholamine cells might mediate the influence of the central amygdala on hypothalamic-pituitary-adrenal axis responses to an immune challenge. Thus these data establish that the central amygdala influences hypothalamic-pituitary-adrenal axis responses to a systemic immune challenge but indicate that it primarily acts by modulating the activity of other control mechanisms.


Subject(s)
Amygdala/physiology , Hypothalamo-Hypophyseal System/drug effects , Hypothalamo-Hypophyseal System/physiology , Interleukin-1/pharmacology , Pituitary-Adrenal System/drug effects , Pituitary-Adrenal System/physiology , Adrenocorticotropic Hormone/metabolism , Animals , Corticotropin-Releasing Hormone/metabolism , Denervation , Epinephrine/physiology , Excitatory Amino Acid Agonists , Hypothalamo-Hypophyseal System/cytology , Ibotenic Acid , Male , Neurons/chemistry , Neurons/metabolism , Norepinephrine/physiology , Oxytocin/metabolism , Paraventricular Hypothalamic Nucleus/cytology , Paraventricular Hypothalamic Nucleus/drug effects , Paraventricular Hypothalamic Nucleus/metabolism , Pituitary-Adrenal System/cytology , Proto-Oncogene Proteins c-fos/analysis , Rats , Rats, Wistar , Septal Nuclei/cytology , Septal Nuclei/drug effects , Septal Nuclei/physiology , Solitary Nucleus/chemistry , Solitary Nucleus/cytology , Solitary Nucleus/physiology
10.
Neuroscience ; 105(3): 707-19, 2001.
Article in English | MEDLINE | ID: mdl-11516835

ABSTRACT

Hypothalamic-pituitary-adrenal axis activation is a hallmark of the stress response. In the case of physical stressors, there is considerable evidence that medullary catecholamine neurones are critical to the activation of the paraventricular nucleus corticotropin-releasing factor cells that constitute the apex of the hypothalamic-pituitary-adrenal axis. In contrast, it has been thought that hypothalamic-pituitary-adrenal axis responses to emotional stressors do not involve brainstem neurones. To investigate this issue we have mapped patterns of restraint-induced neuronal c-fos expression in intact animals and in animals prepared with either paraventricular nucleus-directed injections of a retrograde tracer, lesions of paraventricular nucleus catecholamine terminals, or lesions of the medulla corresponding to the A1 or A2 noradrenergic cell groups. Restraint-induced patterns of neuronal activation within the medulla of intact animals were very similar to those previously reported in response to physical stressors, including the fact that most stressor-responsive, paraventricular nucleus-projecting cells were certainly catecholaminergic and probably noradrenergic. Despite this, the destruction of paraventricular nucleus catecholamine terminals with 6-hydroxydopamine did not alter corticotropin-releasing factor cell responses to restraint. However, animals with ibotenic acid lesions encompassing either the A1 or A2 noradrenergic cell groups displayed significantly suppressed corticotropin-releasing factor cell responses to restraint. Notably, these medullary lesions also suppressed neuronal responses in the medial amygdala, an area that is now considered critical to hypothalamic-pituitary-adrenal axis responses to emotional stressors and that is also known to display a significant increase in noradrenaline turnover during restraint. We conclude that medullary neurones influence corticotropin-releasing factor cell responses to emotional stressors via a multisynaptic pathway that may involve a noradrenergic input to the medial amygdala. These results overturn the idea that hypothalamic-pituitary-adrenal axis response to emotional stressors can occur independently of the brainstem.


Subject(s)
Amygdala/metabolism , Corticotropin-Releasing Hormone/metabolism , Hypothalamo-Hypophyseal System/metabolism , Medulla Oblongata/metabolism , Neurons/metabolism , Paraventricular Hypothalamic Nucleus/metabolism , Stress, Physiological/metabolism , Adrenocorticotropic Hormone/metabolism , Amygdala/cytology , Animals , Catecholamines/metabolism , Cell Count , Denervation , Gold Colloid/pharmacokinetics , Hypothalamo-Hypophyseal System/cytology , Immunohistochemistry , Male , Medulla Oblongata/cytology , Nerve Degeneration/chemically induced , Neural Pathways/cytology , Neural Pathways/metabolism , Neurons/cytology , Oxidopamine/pharmacology , Paraventricular Hypothalamic Nucleus/cytology , Presynaptic Terminals/drug effects , Presynaptic Terminals/metabolism , Proto-Oncogene Proteins c-fos/metabolism , Rats , Rats, Wistar , Restraint, Physical/adverse effects , Solitary Nucleus/cytology , Solitary Nucleus/metabolism , Stress, Physiological/physiopathology , Wheat Germ Agglutinins/pharmacokinetics
11.
Neuroscience ; 118(1): 189-203, 2003.
Article in English | MEDLINE | ID: mdl-12676149

ABSTRACT

Hypothalamic nuclei, particularly the paraventricular nuclei (PVN), are important brain sites responsible for central nervous system responses during an immune challenge. The brainstem catecholamine cells of the nucleus tractus solitarius (NTS) and ventrolateral medulla (VLM) have been shown to play critical roles in relaying systemic immune signals to the PVN. However, whilst it is well recognised that PVN divisions also innervate the NTS and VLM, it is not known whether descending PVN pathways can modulate the recruitment of brainstem cells during an immune challenge. Using systemic administration of the proinflammatory cytokine interleukin-1beta, in combination with Fos immunolabelling, we firstly investigated the effect of PVN lesions on NTS and VLM catecholamine and non-catecholamine cell responses. We found that ibotenic acid lesions of the PVN significantly reduced numbers of Fos-positive non-catecholamine, noradrenergic and adrenergic cells observable in the VLM and NTS after interleukin-1beta administration. We then investigated the origins of descending inputs to the VLM and NTS, activated by systemic interleukin-1beta, by mapping the distribution of Fos-positive retrogradely-labelled cells in divisions of the PVN after iontophoretically depositing choleratoxin-b subunit into the NTS or VLM one week prior to interleukin-1beta administration. We found that, after either NTS or VLM deposits, the majority of retrogradely-labelled Fos-positive cells activated by interleukin-1beta were localised in the medial and lateral parvocellular PVN divisions. Retrogradely-labelled Fos-positive cells were also observed in the NTS after VLM deposits, and in the VLM after NTS tracer deposits, suggesting reciprocal communication between these two nuclei after systemic interleukin-1beta. Thus the present study shows that the PVN has the capacity to modulate NTS and VLM responses after an immune challenge and that these may result from descending projections arising in the medial and lateral PVN divisions. These findings suggest that central nervous system responses to an immune challenge are likely to involve complex reciprocal connections between the PVN and the brainstem as well as between brainstem nuclei themselves.


Subject(s)
Autonomic Nervous System/physiology , Catecholamines/metabolism , Efferent Pathways/physiology , Inflammation/physiopathology , Medulla Oblongata/physiology , Neuroimmunomodulation/physiology , Paraventricular Hypothalamic Nucleus/physiology , Animals , Autonomic Nervous System/cytology , Cholera Toxin/metabolism , Denervation , Efferent Pathways/cytology , Immunohistochemistry , Inflammation Mediators/pharmacology , Interleukin-1/pharmacology , Male , Medulla Oblongata/cytology , Neurotoxins/pharmacology , Paraventricular Hypothalamic Nucleus/cytology , Proto-Oncogene Proteins c-fos/metabolism , Rats , Rats, Wistar , Reticular Formation/physiology , Solitary Nucleus/cytology , Solitary Nucleus/physiology
12.
Neuroscience ; 73(3): 637-42, 1996 Aug.
Article in English | MEDLINE | ID: mdl-8809784

ABSTRACT

It is now clear that ATP acts as a neurotransmitter in both the peripheral and central nervous systems. In the periphery, purinergic transmission has been best studied at certain sympathetic neuroeffector junctions where ATP, co-localized with noradrenaline, is used to elicit the primary post-junctional response. More recently, several groups have raised the possibility that central catecholaminergic neurons might use ATP in a similar fashion. Accordingly, we now present findings from immediate early gene expression and electrophysiological studies which indicate that ATP, acting through P2 purinoreceptors, is used as a transmitter by caudal brainstem noradrenergic neurons, the A1 group, in their interaction with vasopressinergic neurosecretory cells. Supraoptic nucleus vasopressin cell responses to moderate haemorrhage, known to be generated by the A1 projection, were suppressed by hypothalamic application of the P2 receptor antagonist suramin. However, suramin did not alter vasopressin cell responses to osmotic challenge or severely hypotensive haemorrhage, two stimuli known to excite vasopressin cells independently of the A1 projection. These data are consistent with an identity of action between the A1 input to vasopressin cells and the activation of ATP receptors on vasopressin cells. The use of ATP as a transmitter by other catecholamine neurons in the brain awaits further confirmation, but the present findings suggest that in certain instances the therapeutic manipulation of central catecholamine neuron output might best be achieved with pharmacological agents which target purinergic rather than adrenergic transmission.


Subject(s)
Adenosine Triphosphate/physiology , Adrenergic Fibers/physiology , Hemorrhage/physiopathology , Proto-Oncogene Proteins c-fos/metabolism , Suramin/pharmacology , Vasopressins/metabolism , Animals , Blood Pressure/drug effects , Hypothalamus/drug effects
13.
Neuroscience ; 39(3): 717-25, 1990.
Article in English | MEDLINE | ID: mdl-2097524

ABSTRACT

Antibodies to oxytocin and noradrenalin were utilized in an immunocytochemical study of the caudal ventrolateral medulla of the rat brainstem. Noradrenalin was visualized by using antibodies to noradrenalin and by means of a silver-gold intensification of diaminobenzidine, whereas oxytocin could be demonstrated in the same section by using the diaminobenzidine precipitate as a marker. At the light microscopic level, oxytocin fibers were densely distributed around the A1 cell bodies. At the ultrastructural level, oxytocin-containing fibers were seen to terminate synaptically onto noradrenalin-containing neurons. Previous studies have shown that electrical stimulation of A1 neurons selectively activates vasopressin-secreting neurons in the supraoptic nucleus. Therefore, separate electrophysiological studies were set up, in which we observed that oxytocin infusions (100-200 pg) into the A1 area enhanced the activity of 16 out of 19 putative vasopressin-secreting neurons and elicited no response from any of 10 oxytocin-secreting neurons. This finding suggests that some of the parvicellular neurons in the paraventricular nucleus of the hypothalamus, from which the A1 neurons derive their oxytocin innervation, can activate the A1 cell group via this peptidergic neurotransmitter. One of the consequences of A1 neuronal activation is enhanced firing of hypothalamic supraoptic (and paraventricular) vasopressin-secreting neurons, and a consequent rise in plasma vasopressin.


Subject(s)
Medulla Oblongata/ultrastructure , Norepinephrine/analysis , Oxytocin/analysis , Animals , Blood Pressure/drug effects , Dendrites/ultrastructure , Electric Stimulation , Histological Techniques , Immunohistochemistry , Male , Medulla Oblongata/physiology , Neurons/physiology , Norepinephrine/physiology , Oxytocin/pharmacology , Oxytocin/physiology , Pituitary Gland, Posterior/physiology , Rats , Rats, Inbred Strains , Supraoptic Nucleus/physiology , Synapses/physiology , Synapses/ultrastructure
14.
Int J Parasitol ; 31(10): 1093-100, 2001 Aug.
Article in English | MEDLINE | ID: mdl-11429173

ABSTRACT

These studies are focused on schistosomes derived from human infections not cured by three successive doses of praziquantel that also produced infections in mice that were significantly more difficult to cure than infections with control worms. Half (three of six) of these isolates retained their decreased response to praziquantel after multiple passages through the life-cycle in the absence of therapeutic pressure. Two of the isolates, including the one initially least sensitive to praziquantel; reverted, to a sensitivity not significantly different from controls. For example, the EE6 isolate initially required 680 mg/kg praziquantel to affect a 50% reduction in worm load in murine infections, but after only six passages through the life cycle over 5 years this was reduced to 113 mg/kg, not different from control infections. The stability of some of the isolates and the reversion of others indicates that the biological or genetic factors conferring decreased praziquantel response varies among the isolates. The three isolates that retained decreased sensitivity to praziquantel all showed compromises in reproductive fitness in the laboratory, expressed most frequently as a decreased cercarial production from snails infected with those isolates compared to controls. For example, the total cercarial production of snails infected with the EE10 isolate was only 57% that of controls. The reversion of some of the isolates to a praziquantel sensitive state and the decreased reproductive fitness of those that did not revert suggest that there is some biological cost associated with the relative praziquantel insensitivity of these worms, which could help limit the impact of such isolates in the field. Infections with the less sensitive isolates also produced significantly less circulating schistosomal antigen in mice, suggesting that a decrease in the host immune response elicited by these worms could be one of the factors contributing to the diminished praziquantel efficacy.


Subject(s)
Anthelmintics/pharmacology , Praziquantel/pharmacology , Schistosoma mansoni/drug effects , Schistosomiasis mansoni/drug therapy , Animals , Anthelmintics/therapeutic use , Antibodies, Monoclonal , Antigens, Protozoan/analysis , Drug Resistance , Feces/parasitology , Female , Humans , Immunohistochemistry , Intestines/parasitology , Liver/parasitology , Mice , Mice, Inbred BALB C , Parasite Egg Count , Praziquantel/therapeutic use , Reproduction , Schistosoma mansoni/isolation & purification , Schistosoma mansoni/physiology , Schistosomiasis mansoni/parasitology , Snails/parasitology
15.
Psychoneuroendocrinology ; 28(6): 715-32, 2003 Aug.
Article in English | MEDLINE | ID: mdl-12812860

ABSTRACT

Apomorphine is a dopamine receptor agonist that was recently licensed for the treatment of erectile dysfunction. However, although sexual activity can be stressful, there has been little investigation into whether treatments for erectile dysfunction affect stress responses. We have examined whether a single dose of apomorphine, sufficient to produce penile erections (50 microg/kg, i.a.), can alter basal or stress-induced plasma ACTH levels, or activity of central pathways thought to control the hypothalamic-pituitary-adrenal axis in rats. An immune challenge (interleukin-1 beta, 1 microg/kg, i.a.) was used as a physical stressor while sound stress (100 dB white noise, 30 min) was used as a psychological stressor. Intravascular administration of apomorphine had no effect on basal ACTH levels but did substantially increase the number of Fos-positive amygdala and nucleus tractus solitarius catecholamine cells. Administration of apomorphine prior to immune challenge augmented the normal ACTH response to this stressor at 90 min and there was a corresponding increase in the number of Fos-positive paraventricular nucleus corticotropin-releasing factor cells, paraventricular nucleus oxytocin cells and nucleus tractus solitarius catecholamine cells. However, apomorphine treatment did not alter ACTH or Fos responses to sound stress. These data suggest that erection-inducing levels of apomorphine interfere with hypothalamic-pituitary-adrenal axis inhibitory feedback mechanisms in response to a physical stressor, but have no effect on the response to a psychological stressor. Consequently, it is likely that apomorphine acts on a hypothalamic-pituitary-adrenal axis control pathway that is unique to physical stressors. A candidate for this site of action is the nucleus tractus solitarius catecholamine cell population and, in particular, A2 noradrenergic neurons.


Subject(s)
Apomorphine/pharmacology , Dopamine Agonists/pharmacology , Hypothalamo-Hypophyseal System/drug effects , Interleukin-1/physiology , Pituitary-Adrenal System/drug effects , Adrenocorticotropic Hormone/blood , Analysis of Variance , Animals , Apomorphine/therapeutic use , Dopamine Agonists/therapeutic use , Erectile Dysfunction/drug therapy , Immunohistochemistry , Interleukin-1/administration & dosage , Male , Neural Pathways/drug effects , Noise , Oncogene Proteins v-fos/drug effects , Rats , Rats, Wistar , Stress, Physiological/blood , Stress, Psychological/blood
16.
Prog Brain Res ; 81: 303-17, 1989.
Article in English | MEDLINE | ID: mdl-2694223

ABSTRACT

Activation of noradrenergic afferents arising from the A1 cell group of the caudal VLM excites neurosecretory AVP cells of both the supraoptic and paraventricular nuclei, thus stimulating the release of this potent vasoconstrictor into the circulation. Although this effect is mimicked by application of alpha 1-adrenoreceptor agonists to AVP cells, the excitatory effects of A1 afferents may not be mediated by activation of post-synaptic alpha 1-receptors. Evidence has also been obtained that the actions of A1 afferents are not dependent upon the release of excitatory amino acids or NPY, although the latter is co-stored with NA in A1 cells and potentiates the actions of low concentrations of NA on AVP cells. Although a projection to AVP and OXY neurosecretory cells from the A2 NA cell group of the NTS has been established, this projection does not appear to contribute directly to the control of SON AVP cell activity. Rather, NTS stimulation excites SON AVP cells via a relay projection through the A1 cell group. This pathway is likely to correspond to that involved in the stimulatory effects of haemorrhage and caval constriction on AVP secretion, although it is uncertain whether the effects of these particular stimuli are contingent upon unloading of arterial baroreceptors and atrial stretch receptors, as commonly presumed, or upon the activation of other receptors such as ventricular mechanoreceptors or chemoreceptors. On balance, current evidence suggests that the A1 projection is unlikely to be critically involved in mediating the effects of arterial baroreceptor, arterial chemoreceptor, or atrial stretch receptor activation on AVP cells.


Subject(s)
Arginine Vasopressin/metabolism , Medulla Oblongata/physiology , Norepinephrine/pharmacology , Paraventricular Hypothalamic Nucleus/drug effects , Supraoptic Nucleus/drug effects , Animals , Paraventricular Hypothalamic Nucleus/metabolism , Supraoptic Nucleus/metabolism
17.
J Neuroendocrinol ; 8(11): 819-24, 1996 Nov.
Article in English | MEDLINE | ID: mdl-8933358

ABSTRACT

Systemic administration of cholecystokinin (CCK) stimulates neurosecretory oxytocin (OT) and tuberoinfundibular corticotrophin releasing factor (CRF) cells of the hypothalamus. Data from previous studies suggest that A2 noradrengeric neurons of the dorsomedial medulla contribute to the OT cell response, but the role of other medullary catecholamine cells remains unclear. Using c-fos expression as a marker for cellular activity, we have found that CCK (100 micrograms/kg, i.p.) activates substantial populations of tyrosine hydroxylase and phenyl-N-methyl-transferase immunoreactive cells in the medulla, consistent with recruitment of overlapped noradrenergic and adrenergic cell populations in both the ventrolateral and dorsomedial medulla. In the ventrolateral medulla there was a particularly prominent activation of C1 adrenergic neurons at the level of the obex. To directly test the contribution of VLM catecholamine cells to hypothalamic neuroendocrine cell responses to CCK, animals were prepared with unilateral VLM lesions corresponding to those areas that had displayed the most marked response to CCK. VLM lesioned animals treated with CCK displayed a significant although small reduction in paraventricular nucleus (PVN) OT cell c-fos expression ipsilateral to the lesion, but no change in the responses of supraoptic nucleus OT cells or in cells of the medial parvocellular PVN, many of which are CRF cells. These findings indicate that VLM catecholamine cells make little contribution to hypothalamic neuroendocrine cell responses to CCK and thus serve to further highlight the role of dorsomedial catecholamine cells. However, it is now apparent that, in addition to A2 noradrenergic cells, CCK treatment also recruits C2 adrenergic cells of the dorsomedial medulla, many of which have previously been shown to project to the PVN.


Subject(s)
Catecholamines/physiology , Cholecystokinin/pharmacology , Medulla Oblongata/physiology , Neurosecretory Systems/drug effects , Animals , Corticotropin-Releasing Hormone/metabolism , Hypothalamus/drug effects , Immunohistochemistry , Male , Medulla Oblongata/cytology , Neurons/physiology , Oxytocin/metabolism , Phenylethanolamine N-Methyltransferase/metabolism , Proto-Oncogene Proteins c-fos/metabolism , Rats , Rats, Wistar , Tyrosine 3-Monooxygenase/metabolism
18.
J Neuroendocrinol ; 10(7): 519-28, 1998 Jul.
Article in English | MEDLINE | ID: mdl-9700679

ABSTRACT

Systemic administration of the cytokine IL-1 beta produces a significant release of ACTH into the plasma and activation of hypothalamic oxytocin (OT) and corticotropin releasing factor (CRF) cells. However, the mechanism(s) by which systemic IL-1 beta induces these responses is not clear. In the present study, we have investigated the proposal that catecholamine cells of the ventrolateral medulla (VLM) and nucleus of the solitary tract (NTS) can relay circulating IL-1 signals via a prostaglandin-dependent mechanism to effect the HPA axis responses in the rat. Intra-arterial administration of IL-1 beta (1 pg/kg) to otherwise untreated animals produced a prominent release of ACTH into the plasma, substantial c-fos expression in paraventricular medial parvocellular (mPVN) corticotropin releasing factor (CRF) cells, supraoptic (SON) and paraventricular nucleus (PVN) OT cells, area postrema cells, NTS and VLM catecholamine cells and cells of the central amygdala. Pretreatment with the prostaglandin synthesis inhibitor, indomethacin (10 mg/kg body weight ia) 15 min before IL-1 beta administration (1 pg/kg ia) significantly reduced plasma ACTH release and c-fos expression in PVN and SON OT cells and MPVN CRF cells, in addition, the area postrema, A1 and C1 catecholamine cell groups of the VLM and A2 and C2 catecholamine cell groups of the NTS, all exhibited concomitant reductions in c-fos expression. Conversely indomethacin administration did not alter the IL1 beta-induced expression of c-fos in the central amygdala. These data suggest that central pathways involved in the IL-1 beta-induced activation of the HPA axis and OT cells are, at least in part, dependent upon prostaglandin synthesis. It is proposed that neurons in the area postrema, NTS and VLM might mediate this IL-1 beta-induced activation of hypothalamic CRF and OT cells and release of ACTH into the plasma.


Subject(s)
Adrenal Glands/physiology , Hypothalamus/physiology , Indomethacin/pharmacology , Interleukin-1/pharmacology , Oxytocin/metabolism , Pituitary Gland/physiology , Adrenocorticotropic Hormone/metabolism , Amygdala/metabolism , Animals , Catecholamines/metabolism , Corticotropin-Releasing Hormone/metabolism , Cyclooxygenase Inhibitors/pharmacology , Gene Expression/drug effects , Genes, fos/genetics , Male , Medulla Oblongata/metabolism , Paraventricular Hypothalamic Nucleus/metabolism , Rats , Rats, Wistar , Supraoptic Nucleus/metabolism
19.
J Neuroendocrinol ; 12(8): 784-94, 2000 Aug.
Article in English | MEDLINE | ID: mdl-10929091

ABSTRACT

Oestrogen replacement therapy reportedly suppresses hypothalamic-pituitary-adrenal (HPA) axis responses to an emotional stressor in postmenopausal women. However, most studies in the rat suggest a facilitatory role for oestrogen in the control of HPA axis function. One explanation for this difference may be the regimen of oestrogen replacement: during oestrogen replacement therapy, oestrogen levels are low and constant whereas most animal studies examined the HPA axis response when oestrogen levels are rising. In the present study, we assessed HPA axis stress responses in mature ovariectomized rats after plasma oestrogen levels had been maintained at physiological levels for a prolonged period (25 or 100 pg/ml for 7 days). In the case of both an emotional stressor (noise) and a physical stressor (immune challenge by systemic interleukin-1beta administration), oestrogen replacement suppressed stress-related Fos-like immunolabelling, in hypothalamic neuroendocrine cells and plasma adrenocorticotropin hormone responses. From the present data, and past reports, it appears unlikely that these effects of oestrogen are due to a direct action on corticotropin-releasing factor or oxytocin cells. Therefore, to obtain some indication of oestrogen's possible site(s) of action, Fos-like immunolabelling was mapped in the amygdala and in brainstem catecholamine groups, which are neuronal populations demonstrating substantial evidence of involvement in the generation of HPA axis stress responses. In the amygdala, oestrogen replacement suppressed central nucleus responses to immune challenge, but not to noise. Amongst catecholamine cells, oestrogen replacement was more effective against responses to noise than immune challenge, suppressing A1 and A2 (noradrenergic) and C2 (adrenergic) responses to noise, but only A1 responses to immune challenge. These data suggest that, as in postmenopausal women on oestrogen replacement therapy, chronic low-level oestrogen replacement can suppress HPA axis stress responses in the rat. Moreover, oestrogen appears to exert effects at multiple sites within putative HPA axis control pathways, even though most of the relevant neuronal populations do not contain genomic receptors for this gonadal steroid and the pattern of oestrogen action differs for an emotional vs a physical stressor.


Subject(s)
Estrogen Replacement Therapy , Estrogens/pharmacology , Hypothalamo-Hypophyseal System/metabolism , Pituitary-Adrenal System/metabolism , Stress, Physiological/metabolism , Acoustic Stimulation , Adrenocorticotropic Hormone/blood , Amygdala/cytology , Amygdala/drug effects , Amygdala/metabolism , Animals , Brain Stem/cytology , Brain Stem/drug effects , Brain Stem/metabolism , Catecholamines/metabolism , Corticotropin-Releasing Hormone/metabolism , Estrogens/blood , Female , Hypothalamo-Hypophyseal System/drug effects , Hypothalamo-Hypophyseal System/immunology , Interleukin-1/immunology , Interleukin-1/pharmacology , Neurons/cytology , Neurons/drug effects , Neurons/metabolism , Ovariectomy , Oxytocin/metabolism , Paraventricular Hypothalamic Nucleus/drug effects , Paraventricular Hypothalamic Nucleus/metabolism , Pituitary-Adrenal System/drug effects , Pituitary-Adrenal System/immunology , Proto-Oncogene Proteins c-fos/metabolism , Rats , Supraoptic Nucleus/drug effects , Supraoptic Nucleus/metabolism , Time
20.
Ann N Y Acad Sci ; 897: 212-27, 1999.
Article in English | MEDLINE | ID: mdl-10676450

ABSTRACT

Nervous systems of helminths are highly peptidergic. Species in the phylum Nematoda (roundworms) possess at least 50 FMRFamide-related peptides (FaRPs), with more yet to be identified. To date, few non-FaRP neuropeptides have been identified in these organisms, though evidence suggests that other families are present. FaRPergic systems have important functions in nematode neuromuscular control. In contrast, species in the phylum Platyhelminthes (flatworms) apparently utilize fewer FaRPs than do nematodes; those species examined possess one or two FaRPs. Other neuropeptides, such as neuropeptide F (NPF), play key roles in flatworm physiology. Although progress has been made in the characterization of FaRP pharmacology in helminths, much remains to be learned. Most studies on nematodes have been done with Ascaris suum because of its large size. However, thanks to the Caenorhabditis elegans genome project, we know most about the FaRP complement of this free-living animal. That essentially all C. elegans FaRPs are active on at least one A. suum neuromuscular system argues for conservation of ligand-receptor recognition features among the Nematoda. Structure-activity studies on nematode FaRPs have revealed that structure-activity relationship (SAR) "rules" differ considerably among the FaRPs. Second messenger studies, along with experiments on ionic dependence and anatomical requirements for activity, reveal that FaRPs act through many different mechanisms. Platyhelminth FaRPs are myoexcitatory, and no evidence exists of multiple FaRP receptors in flatworms. Interestingly, there are examples of cross-phylum activity, with some nematode FaRPs being active on flatworm muscle. The extent to which other invertebrate FaRPs show cross-phylum activity remains to be determined. How FaRPergic nerves contribute to the control of behavior in helminths, and are integrated with non-neuropeptidergic systems, also remains to be elucidated.


Subject(s)
FMRFamide/analogs & derivatives , FMRFamide/pharmacology , Helminths/physiology , Amino Acid Sequence , Animals , FMRFamide/physiology , Helminths/drug effects , Nematoda/drug effects , Nematoda/physiology , Signal Transduction
SELECTION OF CITATIONS
SEARCH DETAIL