Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 84
Filter
Add more filters










Publication year range
1.
BMC Neurol ; 23(1): 227, 2023 Jun 12.
Article in English | MEDLINE | ID: mdl-37308838

ABSTRACT

Starting from the perspective of an immune-privileged site, our knowledge of the inflammatory processes within the central nervous system has increased rapidly over the last 30 years, leading to a rather puzzling picture today. Of particular interest is the emergence of disease- and injury-specific inflammatory responses within the brain, which may form the basis for future therapeutic approaches. To advance this important topic, we invite authors to contribute research and clinical papers to the Collection "Neuroinflammation and Brain Disease".


Subject(s)
Brain Diseases , Neuroinflammatory Diseases , Humans , Brain
2.
Clin Genet ; 91(1): 115-120, 2017 01.
Article in English | MEDLINE | ID: mdl-26891472

ABSTRACT

L1CAM gene mutations cause neurodevelopmental disorders collectively termed L1 syndrome. Insufficient information about L1CAM variants complicates clinical prognosis, genetic diagnosis and genetic counseling. We combined clinical data, in silico effect predictions and functional analysis of four L1CAM variants, p.I37N, p.T38M, p.M172I and p.D202Y, located to the two N-terminal Ig-like domains present in five families with symptoms of L1 syndrome. Software tools predicted destabilizing effects of p.I37N and p.D202Y but results for p.T38M and p.M172I were inconsistent. Cell surface expression of mutant proteins L1-T38M, L1-M172I and L1-D202Y was normal. Conversely, L1-I37N accumulated in the endoplasmic reticulum (ER) and showed temperature-sensitive protein maturation suggesting that p.I37N induces protein misfolding. L1CAM-mediated cell-cell aggregation was severely impaired by L1CAM variants p.I37N, p.M172I and p.D202Y but was preserved by the variant p.T38M. Our experimental data indicate that protein misfolding and accumulation in the ER affect function of the L1CAM variant p.I37N whereas the variants p.M172I and p.D202Y impair homophilic interaction at the cell surface.


Subject(s)
Genetic Diseases, X-Linked/genetics , Genetic Predisposition to Disease/genetics , Intellectual Disability/genetics , Mutation, Missense , Neural Cell Adhesion Molecule L1/genetics , Spastic Paraplegia, Hereditary/genetics , Amino Acid Sequence , Binding Sites/genetics , Cell Communication/genetics , Cell Membrane/metabolism , Endoplasmic Reticulum/metabolism , Family Health , Female , Genetic Diseases, X-Linked/diagnosis , Genetic Diseases, X-Linked/metabolism , HEK293 Cells , Humans , Immunoblotting , Immunoglobulin Domains/genetics , Intellectual Disability/diagnosis , Intellectual Disability/metabolism , Male , Microscopy, Confocal , Neural Cell Adhesion Molecule L1/metabolism , Pedigree , Sequence Homology, Amino Acid , Spastic Paraplegia, Hereditary/diagnosis , Spastic Paraplegia, Hereditary/metabolism
3.
Neuroscience ; 298: 302-17, 2015 Jul 09.
Article in English | MEDLINE | ID: mdl-25892702

ABSTRACT

L-3,4-Dihydroxyphenylalanine (L-DOPA) is the therapeutic gold standard in Parkinson's disease. However, long-term treatment is complicated by the induction of debilitating abnormal involuntary movements termed L-DOPA-induced dyskinesias (LIDs). Until today the underlying mechanisms of LID pathogenesis are not fully understood. The aim of this study was to reveal new factors, which may be involved in the induction of LID. We have focused on the expression of striatal tyrosine hydroxylase-positive (TH+) neurons, which are capable of producing either L-DOPA or dopamine (DA) in target areas of ventral midbrain DAergic neurons. To address this issue, a daily L-DOPA dose was administered over the course of 15 days to mice with unilateral 6-hydroxydopamine-induced lesions of the medial forebrain bundle and LIDs were evaluated. Remarkably, the number of striatal TH+ neurons strongly correlated with both induction and severity of LID as well as ΔFosB expression as an established molecular marker for LID. Furthermore, dyskinetic mice showed a marked augmentation of serotonergic fiber innervation in the striatum, enabling the decarboxylation of L-DOPA to DA. Axial, limb and orolingual dyskinesias were predominantly associated with TH+ neurons in the lateral striatum, whereas medially located TH+ neurons triggered locomotive rotations. In contrast, identified accumbal and cortical TH+ cells did not contribute to the generation of LID. Thus, striatal TH+ cells and serotonergic terminals may cooperatively synthesize DA and subsequently contribute to supraphysiological synaptic DA concentrations, an accepted cause in LID pathogenesis.


Subject(s)
Corpus Striatum/pathology , Dyskinesia, Drug-Induced/pathology , Functional Laterality/physiology , Neurons/metabolism , Tyrosine 3-Monooxygenase/metabolism , Amphetamine/pharmacology , Animals , Antiparkinson Agents/adverse effects , Disease Models, Animal , Dyskinesia, Drug-Induced/etiology , Levodopa/adverse effects , Male , Medial Forebrain Bundle/drug effects , Medial Forebrain Bundle/injuries , Mice , Mice, Inbred C57BL , Oxidopamine/toxicity , Parkinson Disease/drug therapy , Parkinson Disease/etiology , Phosphopyruvate Hydratase/metabolism , Proto-Oncogene Proteins c-fos/metabolism , Serotonin Plasma Membrane Transport Proteins/metabolism , Statistics, Nonparametric
4.
Diabetologia ; 56(5): 1047-56, 2013 May.
Article in English | MEDLINE | ID: mdl-23404442

ABSTRACT

AIMS/HYPOTHESIS: Imaging of beta cell mass (BCM) is a major challenge in diabetes research. The vesicular monoamine transporter 2 (VMAT2) is abundantly expressed in human beta cells. Radiolabelled analogues of tetrabenazine (TBZ; a low-molecular-weight, cell-permeant VMAT2-selective ligand) have been employed for pancreatic islet imaging in humans. Since reports on TBZ-based VMAT2 imaging in rodent pancreas have been fraught with confusion, we compared VMAT2 gene expression patterns in the mouse, rat, pig and human pancreas, to identify appropriate animal models with which to further validate and optimise TBZ imaging in humans. METHODS: We used a panel of highly sensitive VMAT2 antibodies developed against equivalently antigenic regions of the transporter from each species in combination with immunostaining for insulin and species-specific in situ hybridisation probes. Individual pancreatic islets were obtained by laser-capture microdissection and subjected to analysis of mRNA expression of VMAT2. RESULTS: The VMAT2 protein was not expressed in beta cells in the adult pancreas of common mouse or rat laboratory strains, in contrast to its expression in beta cells (but not other pancreatic endocrine cell types) in the pancreas of pigs and humans. VMAT2- and tyrosine hydroxylase co-positive (catecholaminergic) innervation was less abundant in humans than in rodents. VMAT2-positive mast cells were identified in the pancreas of all species. CONCLUSIONS/INTERPRETATION: Primates and pigs are suitable models for TBZ imaging of beta cells. Rodents, because of a complete lack of VMAT2 expression in the endocrine pancreas, are a 'null' model for assessing interference with BCM measurements by VMAT2-positive mast cells and sympathetic innervation in the pancreas.


Subject(s)
Insulin-Secreting Cells/metabolism , Pancreas/metabolism , Vesicular Monoamine Transport Proteins/metabolism , Aged , Animals , Female , Gene Expression Regulation , Humans , Insulin-Secreting Cells/cytology , Insulin-Secreting Cells/immunology , Ligands , Male , Mast Cells/cytology , Mast Cells/immunology , Mast Cells/metabolism , Mice , Middle Aged , Nerve Endings/metabolism , Pancreas/cytology , Pancreas/immunology , Pancreas/innervation , Radioligand Assay , Rats , Species Specificity , Sus scrofa , Sympathetic Nervous System/cytology , Sympathetic Nervous System/metabolism , Tetrabenazine/analogs & derivatives , Tetrabenazine/metabolism , Vesicular Monoamine Transport Proteins/genetics
5.
Nucleosides Nucleotides Nucleic Acids ; 29(4-6): 476-81, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20544541

ABSTRACT

An increasing body of evidence has suggested a role of pyrimidine nucleotides not only in metabolic pathways in the developing nervous system but also in adult brain functions. There is still little known about the cellular sources of pyrimidine synthesis and the distribution of enzymes involved in the biosynthetic pathway of pyrimidines in the central nervous system (CNS) of mammals. Therefore, we investigated the activity of dihydroorotate dehydrogenase (DHODH), the fourth enzyme in the pyrimidine de novo synthesis, by catalytic enzyme histochemistry in adult rat brain. Its distribution was confirmed by Western blot analysis of dissected brain regions and by immunohistochemical analysis of paraffin-embedded formalin fixed sections employing an affinity-purified highly specific antibody. Microscopic analysis of brain sections revealed the presence of enzymatically active DHODH in many CNS regions, albeit at different intensities. High levels of both DHODH activity and immunoreactivity were observed in the neocortex, hippocampus, spinal cord and choroid plexus; lower levels were seen in the cerebellum, and only marginal expression in brainstem. The prominent staining of neuronal cell bodies in these regions suggests a neuronal location of DHODH. Neuronally derived pyrimidine de novo synthesis in the rodent CNS, independent of exogeneous sources, would provide pyrimidines for dynamic processes of membrane assembly, rearrangement and neuronal plasticity, as well as supplying uridine nucleotides as neuronal signalling molecules.


Subject(s)
Central Nervous System/enzymology , Neurons/enzymology , Oxidoreductases Acting on CH-CH Group Donors/metabolism , Pyrimidines/biosynthesis , Animals , Blotting, Western , Dihydroorotate Dehydrogenase , Immunohistochemistry , Rats
6.
Regul Pept ; 165(1): 36-44, 2010 Nov 30.
Article in English | MEDLINE | ID: mdl-20005907

ABSTRACT

Immunoreactivity for both processed and unprocessed forms of chromogranin A (CGA) was examined, using an antibody recognizing the WE14 epitope, among terminal fields and cell bodies of anatomically defined GABAergic, glutamatergic, cholinergic, catecholaminergic, and peptidergic cell groups in the rodent central nervous system. CGA is ubiquitous within neuronal cell bodies, with no obvious anatomical or chemically-coded subdivision of the nervous system in which CGA is not expressed in most neurons. CGA expression is essentially absent from catecholaminergic terminal fields in the CNS, suggesting a relative paucity of large dense-core vesicles in CNS compared to peripheral catecholaminergic neurons. Extensive synaptic co-localization with classical transmitter markers is not observed even in areas such as amygdala, where CGA fibers are numerous, suggesting preferential segregation of CGA to peptidergic terminals in CNS. Localization of CGA in dendrites in some areas of CNS may indicate its involvement in regulation of dendritic release mechanisms. Finally, the ubiquitous presence of CGA in neuronal cell somata, especially pronounced in GABAergic neurons, suggests a second non-secretory vesicle-associated function for CGA in CNS. We propose that CGA may function in the CNS as a prohormone and granulogenic factor in some terminal fields, but also possesses as-yet unknown unique cellular functions within neuronal somata and dendrites.


Subject(s)
Central Nervous System/metabolism , Chromogranin A/metabolism , Neurons/metabolism , Animals , Immunohistochemistry , In Vitro Techniques , Male , Mice
7.
Neuroscience ; 156(2): 310-8, 2008 Oct 02.
Article in English | MEDLINE | ID: mdl-18722510

ABSTRACT

Classic neurotransmitter phenotypes are generally predetermined and develop as a consequence of target-independent lineage decisions. A unique mode of target-dependent phenotype instruction is the acquisition of the cholinergic phenotype in the peripheral sympathetic nervous system. A body of work suggests that the sweat gland plays an important role to determine the cholinergic phenotype at this target site. A key issue is whether neurons destined to innervate the sweat glands express cholinergic markers before or only after their terminals make target contact. We employed cholinergic-specific over-expression of the vesicular acetylcholine transporter (VAChT) in transgenic mice to overcome sensitivity limits in the detection of initial cholinergic sweat gland innervation. We found that VAChT immunoreactive nerve terminals were present around the sweat gland anlage already from the earliest postnatal stages on, coincident selectively at this sympathetic target with tyrosine hydroxylase-positive fibers. Our results provide a new mechanistic model for sympathetic neuron-target interaction during development, with initial selection by the target of pioneering nerve terminals expressing a cholinergic phenotype, and subsequent stabilization of this phenotype during development.


Subject(s)
Acetylcholine/metabolism , Neurons/metabolism , Norepinephrine/metabolism , Phenotype , Sweat Glands/innervation , Sympathetic Nervous System/cytology , Age Factors , Animals , Animals, Newborn , Choline O-Acetyltransferase/genetics , Embryo, Mammalian , Gene Expression Regulation, Developmental/physiology , Green Fluorescent Proteins/biosynthesis , Green Fluorescent Proteins/genetics , Mice , Mice, Transgenic , Sympathetic Nervous System/growth & development , Tyrosine 3-Monooxygenase/metabolism , Vesicular Acetylcholine Transport Proteins/genetics , Vesicular Acetylcholine Transport Proteins/metabolism
8.
Dev Biol (Basel) ; 131: 73-81, 2008.
Article in English | MEDLINE | ID: mdl-18634468

ABSTRACT

Rabies virus (RABV) infection is characterized by the rapid neuronal spread of RABV into the CNS before a protective immune response is raised. Therefore, a typical feature of RABV infection is the paucity of inflammatory reactions in the brain. Here we examined whether the induction of immunosuppressive neuropeptides, in particular CGRP, may contribute to the ability of RABV to evade immune responses. RABV infection of mice caused a strong induction of calcitonin gene-related peptide (CGRP) in neurons and fibres in the neocortex as well as in the dentate gyrus and CA1 region of the hippocampus although RABV did not infect neurons in which CGRP expression was upregulated. Neuropeptide Y (NPY) or vasoactive intestinal peptide (VIP) expressing neurons also were not infected by RABV. In contrast, somatostatin neurons were infected by RABV. There was evidence for an RABV-induced increase of VIP and somatostatin but not of NPY. To test how CGRP expression is related to TNFalpha-induced enhancement of CNS innate and adaptive immunity during RABV infection, we used recombinant RABVs that contained either an active (SPBN-TNFalpha(+)) or an inactive (SPBN-TNFalpha(-)) TNFalpha gene. As compared to SPBN-TNFalpha(-), infection with SPBN-TNFalpha(+) attenuated the induction of CGRP but simultaneously enhanced induction of the invariant chain of MHC II, microglial activation and T cell infiltration. In conclusion, distinct neuropeptidergic neurons in the brain are remarkably spared from RABV infection suggesting a pivotal role of neuropeptides during CNS virus infection. Given the inhibitory effect of CGRP on antigen presentation, we propose that the strong RABV-induced upregulation of CGRP in the brain may contribute to the mechanism by which RABV escapes immune detection. Targeting the expression of neuropeptides, in particular CGRP, that are induced during RABV infection may open a new avenue for therapeutic intervention in human rabies.


Subject(s)
Brain/metabolism , Central Nervous System Viral Diseases/immunology , Neuropeptides/metabolism , Rabies virus/immunology , Rabies/immunology , Animals , Brain/pathology , Brain/virology , Central Nervous System Viral Diseases/metabolism , Central Nervous System Viral Diseases/pathology , Female , Mice , Neuropeptides/genetics , Rabies/metabolism , Rabies/pathology , Up-Regulation
9.
Proc Natl Acad Sci U S A ; 105(28): 9775-80, 2008 Jul 15.
Article in English | MEDLINE | ID: mdl-18626010

ABSTRACT

Regulated intramembrane proteolysis by gamma-secretase cleaves proteins in their transmembrane domain and is involved in important signaling pathways. At least four different gamma-secretase complexes have been identified, but little is known about their biological role and specificity. Previous work has demonstrated the involvement of the Aph1A-gamma-secretase complex in Notch signaling, but no specific function could be assigned to Aph1B/C-gamma-secretase. We demonstrate here that the Aph1B/C-gamma-secretase complex is expressed in brain areas relevant to schizophrenia pathogenesis and that Aph1B/C deficiency causes pharmacological and behavioral abnormalities that can be reversed by antipsychotic drugs. At the molecular level we find accumulation of Nrg1 fragments in the brain of Aph1BC(-/-) mice. Our observations gain clinical relevance by the demonstration that a Val-to-Leu mutation in the Nrg1 transmembrane domain, associated with increased risk for schizophrenia, affects gamma-secretase cleavage of Nrg1. This finding suggests that dysregulation of intramembrane proteolysis of Nrg1 could increase risk for schizophrenia and related disorders.


Subject(s)
Amyloid Precursor Protein Secretases/deficiency , Antipsychotic Agents/pharmacology , Endopeptidases/deficiency , Gait Disorders, Neurologic/etiology , Neuregulin-1/metabolism , Protein Subunits/deficiency , Animals , Antipsychotic Agents/therapeutic use , Membrane Proteins , Mice , Mice, Knockout , Mutation, Missense/physiology , Schizophrenia/etiology
10.
J Neuroendocrinol ; 17(9): 570-6, 2005 Sep.
Article in English | MEDLINE | ID: mdl-16101895

ABSTRACT

Peptides participating in the hypothalamic control of feeding behaviour are also involved in the central autonomic control of gastrointestinal functions, such as secretion and motility. An anatomical interaction and functional relationship in the central nervous system between the feeding-related peptides neuropeptide Y and ghrelin is well documented. Furthermore, it has been shown that feeding-related peptides can influence digestive function via central corticotrophin-releasing factor (CRF) pathways. In the present study, we investigated the role of ghrelin in the central autonomic control of colonic motility. Furthermore, we addressed the hypothesis that ghrelin is involved in the hypothalamic control of colonic motor function, utilizing central neuropeptide Y receptors and hypothalamic CRF pathways. Ghrelin (0.03, 0.06 and 0.12 nmol) bilaterally microinjected into the paraventricular nucleus (PVN) induced a significant stimulation of colonic propulsion. In particular, the colonic transit time decreased from 312+/-7 min to 198+/-12 min. Microinjection of the neuropeptide Y1 receptor antagonist, BIBP-3226 (200 pmol), or the nonselective CRF receptor antagonist, astressin (30 pmol), into the PVN abolished the stimulatory effect of ghrelin injected into the PVN on colonic transit time, whereas pretreatment with the selective CRF2 receptor, antisauvagine-30 (28 pmol), failed to affect the effect of PVN-ghrelin injection on colonic propulsion. These results suggest that ghrelin can act as central modulator of gastrointestinal motor functions at the level of the PVN via neuropeptide Y1- and CRF1 receptor-dependent mechanisms.


Subject(s)
Colon/drug effects , Gastrointestinal Motility/drug effects , Hypothalamus/physiology , Neuropeptide Y/metabolism , Peptide Hormones/pharmacology , Receptors, Corticotropin-Releasing Hormone/metabolism , Animals , Arginine/administration & dosage , Arginine/analogs & derivatives , Arginine/pharmacology , Brain/anatomy & histology , Colon/innervation , Corticotropin-Releasing Hormone/pharmacology , Ghrelin , Hypothalamus/metabolism , Male , Microinjections , Neuropeptide Y/antagonists & inhibitors , Neuroprotective Agents/pharmacology , Paraventricular Hypothalamic Nucleus/drug effects , Paraventricular Hypothalamic Nucleus/metabolism , Peptide Fragments/administration & dosage , Peptide Fragments/pharmacology , Rats , Rats, Sprague-Dawley , Receptors, Corticotropin-Releasing Hormone/antagonists & inhibitors , Stimulation, Chemical
11.
Neurogastroenterol Motil ; 16(4): 489-96, 2004 Aug.
Article in English | MEDLINE | ID: mdl-15306004

ABSTRACT

Many neuropeptides participating in the hypothalamic control of feeding behaviour and satiety have been shown to be additionally involved in the autonomic control of gastrointestinal (GI) functions. Recently, the neuropeptide cocaine- and amphetamine-regulated transcript (CART) has been indicated to function as an anorectic substance in the brain. In the present study we examine the hypothesis that CART is involved in the modulation of GI motility. Colonic transit time was measured after peripheral and central injection of CART in fed and freely moving Sprague-Dawley rats. Intracerebroventricular injection of synthetic CART (55-102) (190 pmol and 1.9 nmol per 10 microL and saline controls) decreased the colonic transit time of conscious rats up to 46%. In contrast, i.p. injection of CART (55-102) (1.9 nmol and 19 nmol kg(-1) BW and saline controls) had no effect on colonic motility. Central administration of a CRF receptor antagonist (2.8 nmol) prior to central CART administration antagonized the CART-induced stimulation of colonic transit. Pretreatment with the peripherally acting cholinergic antagonist atropin methyl nitrate (0.1 mg kg(-1) i.p.) blocked the stimulatory CART effect on colonic motor function. The results suggest that CART acts in the central nervous system to modulate behavioural motor function via a central CRF receptor-dependent mechanism and peripheral cholinergic pathways.


Subject(s)
Cholinergic Fibers/drug effects , Colon/drug effects , Gastrointestinal Motility/drug effects , Peptide Fragments/pharmacology , Receptors, Corticotropin-Releasing Hormone/physiology , Animals , Colon/physiology , Consciousness/drug effects , Consciousness/physiology , Corticotropin-Releasing Hormone/pharmacology , Dose-Response Relationship, Drug , Eating/drug effects , Eating/physiology , Gastrointestinal Motility/physiology , Male , Nerve Tissue Proteins , Neural Pathways/drug effects , Neural Pathways/physiology , Rats , Rats, Sprague-Dawley , Receptors, Corticotropin-Releasing Hormone/antagonists & inhibitors
12.
Br J Pharmacol ; 140(3): 538-46, 2003 Oct.
Article in English | MEDLINE | ID: mdl-12970097

ABSTRACT

The administration of kappa-opioid receptor antagonists, nor-binaltorphimine (norBNI) and 5'-guanidinonaltrindole (GNTI) enhanced allodynia in rats and mice after sciatic nerve ligation. In order to understand the mechanism underlying this effect, we examined the possible involvement of the endogenous ligand of kappa-opioid receptor dynorphin. The experiments were carried out on male Wistar rats and on Albino-Swiss mice. The rats had been implanted with a catheter 7 days earlier in the subarachnoid space of the spinal cord. Intrathecal (i.t.) administrations in mice were made by lumbar puncture. The animals were i.t. injected with norBNI, GNTI (kappa-opioid receptor antagonists), dynorphin A1-17 antiserum (DYN A/S), ketamine (NMDA receptor antagonist) and their combinations. The nociceptive sensitivity was assessed using the mechanical (von Frey) and thermal allodynia tests on days 2-4 and 8-10 after the sciatic nerve ligation. Both antagonists, norBNI and GNTI, significantly enhanced mechanical and thermal allodynia in rats and mice with neuropathic pain. The potentiation of allodynia after the administration of norBNI or GNTI was inhibited by earlier administration of DYN A/S or by ketamine. Our results suggest that allodynia is mediated through nonopioid effect of the endogenous opioid peptide, dynorphin. The nonopioid action is potentiated by the blockade of kappa-opioid receptors, and corresponding to the elevation of prodynorphin mRNA level in neuropathic pain. Furthermore, the potentiation of allodynia after the administration of the above drugs appears to be mediated through the activation of NMDA receptors directly by dynorphin.


Subject(s)
Narcotic Antagonists/pharmacology , Pain Measurement/drug effects , Pain/metabolism , Receptors, Opioid, kappa/antagonists & inhibitors , Sciatic Nerve/physiology , Animals , Ligation , Male , Mice , Pain Measurement/methods , Rats , Rats, Wistar , Receptors, Opioid, kappa/biosynthesis , Sciatic Nerve/metabolism
13.
Anaesthesist ; 52(11): 1046-54, 2003 Nov.
Article in German | MEDLINE | ID: mdl-14992093

ABSTRACT

Ambulatory surgical care is intended to save healthcare expenditure from the economical viewpoint. From the patients point of view significant advantages as well as specific disadvantages of ambulatory surgery are known. The increase in the volume and complexity of procedures provided in an ambulatory setting are driven by improvements in anesthesia and surgical technique as well as by changes in financing and reimbursement. Therefore careful quality control and scientific evidence for the safety of increasingly used complex surgical procedures for higher risk patients is essential. Reducing the surgical trauma by minimally invasive surgical techniques and very good controllability by modern anesthesia concepts is making the management of the postoperative period crucial for successful ambulatory surgery. Most of the complications and common problems during the postoperative period, such as pain, nausea and vomiting, are not specific for ambulatory surgery, but management places an increasing burden of responsibility not only on general and specialised physicians, but also on other health professionals, patients, and family members.


Subject(s)
Ambulatory Surgical Procedures/trends , Aged , Ambulatory Surgical Procedures/economics , Anesthesia , Child , Humans , Pain, Postoperative/epidemiology , Pain, Postoperative/therapy , Patient Compliance , Risk Assessment
14.
Naunyn Schmiedebergs Arch Pharmacol ; 363(3): 288-94, 2001 Mar.
Article in English | MEDLINE | ID: mdl-11284443

ABSTRACT

After repeated administration of cocaine at intervals, sensitization phenomena can be observed, so that its behavioural effects are enhanced. Since this phenomenon is long-lasting, it was of interest to study which persistent alterations in the activity of dopaminergic neurones or of endogenous opioid systems downstream of dopaminergic synapses in the basal ganglia are involved in the sensitization. Cocaine (10 mg/kg i.p.) was administered to rats on days 1, 3, 5 and 7 and saline on days 2, 4 and 6 ("repeated cocaine"), or saline was injected on days 1-6 and cocaine on day 7 ("acute cocaine"), or saline was injected on days 1-7 ("saline group"). The "repeated cocaine" schedule led to a significant sensitization to the locomotor activation produced by cocaine on day 7 or on day 17, 10 days after the end of sensitization protocol. Microdialysis in the nucleus accumbens which was performed after administration of cocaine (10 mg/kg i.p.) on day 7, or after an administration of the same dose 10 days after the last administration of cocaine, respectively, revealed significant acute increases of extracellular dopamine to about 200% of basal values. These increases were similar in "acute cocaine" and in "repeated cocaine" animals both after 7 days and after 17 days. For in situ hybridization studies, rats were sacrificed on day 7, 4.5 h after the last cocaine or saline administration. The mRNA for tyrosine hydroxylase (TH) in substantia nigra + ventral tegmental area was significantly elevated to about 140% of saline controls both in the "repeated cocaine" and the "acute cocaine" group as compared with the "saline group". In contrast, there were no differences between the three groups in the mRNAs of preprodynorphin or preproenkephalin levels measured in the nucleus accumbens (core and shell). These results suggest that sensitization phenomena to cocaine are not necessarily connected with alterations in the dopaminergic activity in the mesolimbic system or in the transcription of precursors of endogenous opioid peptides which are located downstream of the dopaminergic synapses.


Subject(s)
Behavior, Animal/drug effects , Cocaine/pharmacology , Dynorphins/genetics , Enkephalins/genetics , Protein Precursors/genetics , RNA, Messenger/metabolism , Tyrosine Transaminase/genetics , Animals , Dopamine/metabolism , Dopamine/physiology , Dynorphins/biosynthesis , Enkephalins/biosynthesis , In Situ Hybridization , Locomotion/drug effects , Male , Microdialysis , Neurons/drug effects , Neurons/metabolism , Neurons/physiology , Nucleus Accumbens/drug effects , Nucleus Accumbens/enzymology , Nucleus Accumbens/metabolism , Protein Precursors/biosynthesis , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Rats , Rats, Wistar , Tyrosine Transaminase/biosynthesis
15.
J Endocrinol ; 169(1): 87-96, 2001 Apr.
Article in English | MEDLINE | ID: mdl-11250650

ABSTRACT

Cell-cell interactions are important regulatory elements in anterior pituitary (AP) physiology. As model systems to study pituitary cell-cell interactions, AP cells kept either as monolayers or as organotypic reaggregate cultures were analyzed by differential display PCR. We identified six cDNA fragments (osteopontin (Opn), connective tissue growth factor (CTGF), alpha(v)-integrin, cathepsin H, lysozyme and O-acetyl GD(3) ganglioside synthase) that showed elevated expression in monolayers compared with reaggregate cultures and the AP. The adenohypophyseal mRNA expression of Opn and CTGF, two secreted signaling substances, was studied in more detail. In situ hybridization histochemistry revealed that Opn mRNA expression is restricted to a subpopulation of gonadotropes whereas CTGF hybridization signals could not be ascribed to any known cell type. Opn transcript levels were downregulated in the APs of lactating rats and decreased when rats received s.c. injections of 17beta-estradiol for 5 days. The mRNA expression was higher in male than in female rats and increased after gonadectomy. CTGF transcript levels were higher in male compared with female rats and were increased in pregnant rats and in rats treated for 5 days with triiodothyronine or dexamethasone. These results indicate that Opn and CTGF may be of physiological importance as local communication factors in the AP.


Subject(s)
Growth Substances/metabolism , Immediate-Early Proteins/metabolism , Intercellular Signaling Peptides and Proteins , Pituitary Gland, Anterior/metabolism , Sialoglycoproteins/metabolism , Signal Transduction/physiology , Animals , Animals, Newborn , Blotting, Northern/methods , Cells, Cultured , Connective Tissue Growth Factor , Dexamethasone/pharmacology , Female , Growth Substances/genetics , Immediate-Early Proteins/genetics , In Situ Hybridization/methods , Lactation , Male , Models, Animal , Orchiectomy , Osteopontin , Ovariectomy , Polymerase Chain Reaction/methods , Pregnancy , RNA, Messenger/analysis , Rats , Rats, Sprague-Dawley , Sialoglycoproteins/genetics , Triiodothyronine/pharmacology
16.
J Neurosci ; 21(3): 798-811, 2001 Feb 01.
Article in English | MEDLINE | ID: mdl-11157066

ABSTRACT

To test the hypothesis of an involvement of tachykinins in destabilization and hyperexcitation of neuronal circuits, gliosis, and neuroinflammation during cerebral ischemia, we investigated cell-specific expressional changes of the genes encoding substance P (SP), neurokinin B (NKB), and the tachykinin/neurokinin receptors (NK1, NK2, and NK3) after middle cerebral artery occlusion (MCAO) in the rat. Our analysis by quantitative in situ hybridization, immunohistochemistry, and confocal microscopy was concentrated on cerebrocortical areas that survive primary infarction but undergo secondary damage. Here, SP-encoding preprotachykinin-A and NK1 mRNA levels and SP-like immunoreactivity were transiently increased in GABAergic interneurons at 2 d after MCAO. Coincidently, MCAO caused a marked expression of SP and NK1 in a subpopulation of glutamatergic pyramidal cells, and in some neurons SP and NK1 mRNAs were coinduced. Elevated levels of the NKB-encoding preprotachykinin-B mRNA and of NKB-like immunoreactivity at 2 and 7 d after MCAO were confined to GABAergic interneurons. In parallel, the expression of NK3 was markedly downregulated in pyramidal neurons. MCAO caused transient NK1 expression in activated cerebrovenular endothelium within and adjacent to the infarct. NK1 expression was absent from activated astroglia or microglia. The differential ischemia-induced plasticity of the tachykinin system in distinct inhibitory and excitatory cerebrocortical circuits suggests that it may be involved in the balance of endogenous neuroprotection and neurotoxicity by enhancing GABAergic inhibitory circuits or by facilitating glutamate-mediated hyperexcitability. The transient induction of NK1 in cerebrovenular endothelium may contribute to ischemia-induced edema and leukocyte diapedesis. Brain tachykinin receptors are proposed as potential drug targets in stroke.


Subject(s)
Brain Ischemia/metabolism , Cerebral Cortex/metabolism , Endothelium, Vascular/metabolism , Receptors, Tachykinin/biosynthesis , Tachykinins/biosynthesis , Animals , Brain Ischemia/genetics , Brain Ischemia/pathology , Cerebral Cortex/blood supply , Cerebral Cortex/pathology , Cerebrovascular Circulation , Gene Expression Regulation , Glutamic Acid/metabolism , Infarction, Middle Cerebral Artery/genetics , Infarction, Middle Cerebral Artery/metabolism , Infarction, Middle Cerebral Artery/pathology , Male , Neurokinin B/genetics , Neurokinin B/metabolism , Neuronal Plasticity , Protein Precursors/genetics , Protein Precursors/metabolism , RNA, Messenger/metabolism , Rats , Rats, Long-Evans , Receptors, Neurokinin-1/genetics , Receptors, Neurokinin-1/metabolism , Receptors, Neurokinin-3/genetics , Receptors, Neurokinin-3/metabolism , Receptors, Tachykinin/genetics , Substance P/genetics , Substance P/metabolism , Tachykinins/genetics , Tachykinins/metabolism , Venules/metabolism , Venules/pathology , gamma-Aminobutyric Acid/metabolism
17.
J Comp Neurol ; 428(2): 319-36, 2000 Dec 11.
Article in English | MEDLINE | ID: mdl-11064370

ABSTRACT

The distribution of the recently discovered thyrotropin-releasing hormone (TRH) receptor subtype TRH-R2 was studied in rat brain, pituitary, and spinal cord by in situ hybridization histochemistry and compared with the distribution patterns of the other elements of TRH signaling, namely TRH, TRH-R1, and the TRH-degrading ectoenzyme (TRH-DE). In contrast to the very restricted mRNA expression of TRH-R1 in the central nervous system, TRH-R2 mRNA was widely distributed with highest transcript levels throughout the thalamus, in the cerebral and cerebellar cortex, medial habenulae, medial geniculate nucleus, pontine nuclei, and throughout the reticular formation. In accordance with the well-known endocrine function of TRH, TRH-R1 is found predominantly expressed in hypothalamic regions. Expression of TRH-R1 in various brainstem nuclei and spinal cord motoneurons seems to be associated with the described effects of TRH on the vegetative and autonomic system as well as on the somatomotor system. Furthermore, the fully complementary expression of both receptor subtypes, even in regions where transcripts for both receptors were found (e.g., medial septum, lateral hypothalamus superior colliculi, substantia nigra, etc.), indicates that in discrete neuroanatomical pathways the two receptors serve highly specific functions for the transmission of TRH signals. Together with TRH-DE, the putative terminator of TRH actions that shows in various, but not all, brain areas, an overlapping mRNA distribution pattern with both receptors, the distribution of TRH-R2 mRNA seems to provide the anatomical basis for the described effects of TRH on higher cognitive functions as well as its effect on arousal, locomotor activity, and pain perception.


Subject(s)
Aminopeptidases/metabolism , Brain/metabolism , RNA, Messenger/metabolism , Receptors, Thyrotropin-Releasing Hormone/metabolism , Spinal Cord/metabolism , Thyrotropin-Releasing Hormone/metabolism , Animals , Central Nervous System/metabolism , Male , Pyrrolidonecarboxylic Acid/analogs & derivatives , Rats , Rats, Sprague-Dawley
18.
J Virol ; 74(17): 8018-27, 2000 Sep.
Article in English | MEDLINE | ID: mdl-10933711

ABSTRACT

The tissue tropism and spread of infection of the highly pathogenic avian influenza virus A/FPV/Rostock/34 (H7N1) (FPV) were analyzed in 11-day-old chicken embryos. As shown by in situ hybridization, the virus caused generalized infection that was strictly confined to endothelial cells in all organs. Studies with reassortants of FPV and the apathogenic avian strain A/chick/Germany/N/49 (H10N7) revealed that endotheliotropism was linked to FPV hemagglutinin (HA). To further analyze the factors determining endotheliotropism, the HA-activating protease furin was cloned from chicken tissue. Ubiquitous expression of furin and other proprotein convertases in the chick embryo indicated that proteolytic activation of HA was not responsible for restriction of infection to the endothelium. To determine the expression of virus receptors in embryonic tissues, histochemical analysis of alpha2,3- and alpha2,6-linked neuraminic acid was carried out by lectin-binding assays. These receptors were found on endothelial cells and on several epithelial cells, but not on tissues surrounding endothelia. Finally, we analyzed the polarity of virus maturation in endothelial cells. Studies on cultured human endothelial cells employing confocal laser scanning microscopy revealed that HA is specifically targeted to the apical surface of these cells, and electron microscopy of embryonic tissues showed that virus maturation occurs also at the luminar side. Taken together, these observations indicate that endotheliotropism of FPV in the chicken embryo is determined, on one hand, by the high cleavability of HA, which mediates virus entry into the vascular system, and, on the other hand, by restricted receptor expression and polar budding, which prevent spread of infection into tissues surrounding endothelia.


Subject(s)
Endothelium/virology , Influenza A virus/pathogenicity , Animals , Chick Embryo , Endothelium/metabolism , Endothelium/pathology , Furin , Hemagglutinin Glycoproteins, Influenza Virus/metabolism , Humans , In Situ Hybridization , Influenza A virus/ultrastructure , Microscopy, Confocal , Neuraminic Acids/analysis , Neuraminidase/metabolism , Organ Specificity , Proprotein Convertase 5 , Proprotein Convertases , Receptors, Virus/analysis , Serine Endopeptidases/metabolism , Subtilisins/metabolism
19.
J Immunol ; 164(10): 5446-52, 2000 May 15.
Article in English | MEDLINE | ID: mdl-10799911

ABSTRACT

Recent evidence suggests that the pathophysiology of neurodegenerative and inflammatory neurological diseases has a neuroimmunological component involving complement, an innate humoral immune defense system. The present study demonstrates the effects of experimentally induced global ischemia on the biosynthesis of C1q, the recognition subcomponent of the classical complement activation pathway, in the CNS. Using semiquantitative in situ hybridization, immunohistochemistry, and confocal laser scanning microscopy, a dramatic and widespread increase of C1q biosynthesis in rat brain microglia (but not in astrocytes or neurons) within 24 h after the ischemic insult was observed. A marked increase of C1q functional activity in cerebrospinal fluid taken 1, 24, and 72 h after the ischemic insult was determined by C1q-dependent hemolytic assay. In the light of the well-established role of complement and complement activation products in the initiation and maintenance of inflammation, the ischemia-induced increase of cerebral C1q biosynthesis and of C1q functional activity in the cerebrospinal fluid implies that the proinflammatory activities of locally produced complement are likely to contribute to the pathophysiology of cerebral ischemia. Pharmacological modulation of complement activation in the brain may be a therapeutic target in the treatment of stroke.


Subject(s)
Brain/immunology , Complement C1q/biosynthesis , Ischemic Attack, Transient/immunology , Microglia/immunology , Microglia/metabolism , Up-Regulation/immunology , Animals , Brain/pathology , Complement C1q/cerebrospinal fluid , Complement C1q/genetics , Digoxigenin , Immunohistochemistry , In Situ Hybridization , Ischemic Attack, Transient/cerebrospinal fluid , Ischemic Attack, Transient/pathology , Male , Microglia/pathology , RNA Probes , RNA, Complementary , Rats , Rats, Wistar , Sulfur Radioisotopes , Up-Regulation/genetics
20.
Neuroscience ; 96(4): 707-22, 2000.
Article in English | MEDLINE | ID: mdl-10727789

ABSTRACT

We examined the expression pattern of the vesicular acetylcholine transporter in the mouse nervous system, using rodent-specific riboprobes and antibodies, prior to comparing it with the distribution of vesicular acetylcholine transporter expressed from a human transgene in the mouse, using riboprobes and antibodies specific for human. Endogenous vesicular acetylcholine transporter expression was high in spinal and brainstem somatomotor neurons, vagal visceromotor neurons, and postganglionic parasympathetic neurons, moderate in basal forebrain and brainstem projection neurons and striatal interneurons, and low in intestinal intrinsic neurons. Vesicular acetylcholine transporter expression in intrinsic cortical neurons was restricted to the entorhinal cortex. The sequence of the mouse cholinergic gene locus to 5.1kb upstream of the start of transcription of the vesicular acetylcholine transporter gene was determined and compared with the corresponding region of the human gene. Cis-regulatory domains implicated previously in human or rat cholinergic gene regulation are highly conserved in mouse, indicating their probable relevance to the regulation of the mammalian cholinergic gene locus in vivo. Mouse lines were established containing a human transgene that included the vesicular acetylcholine transporter gene and sequences spanning 5kb upstream and 1.8kb downstream of the vesicular acetylcholine transporter open reading frame. In this transgene, the intact human vesicular acetylcholine transporter was able to act as its own reporter. This allowed elements within the vesicular acetylcholine transporter open reading frame itself, shown previously to affect transcription in vitro, to be assessed in vivo with antibodies and riboprobes that reliably distinguished between human and mouse vesicular acetylcholine transporters and their messenger RNAs. Expression of the human vesicular acetylcholine transporter was restricted to mouse cholinergic somatomotor neurons in the spinal cord and brainstem, but absent from other central and peripheral cholinergic neurons. The mouse appears to be an appropriate model for the study of the genetic regulation of the cholinergic gene locus, and the physiology and neurochemistry of the mammalian cholinergic nervous system, although differences exist in the distribution of cortical cholinergic neurons between the mouse and other mammals. The somatomotor neuron-specific expression pattern of the transgenic human vesicular acetylcholine transporter suggests a mosaic model for cholinergic gene locus regulation in separate subdivisions of the mammalian cholinergic nervous system.


Subject(s)
Acetylcholine/genetics , Acetylcholine/metabolism , Carrier Proteins/genetics , Carrier Proteins/metabolism , Cholinergic Fibers/metabolism , Gene Expression Regulation/physiology , Membrane Transport Proteins , Motor Neurons/metabolism , Vesicular Transport Proteins , Animals , Biological Transport/physiology , Brain Mapping , Central Nervous System/cytology , Central Nervous System/metabolism , Cholinergic Fibers/ultrastructure , Chromosome Mapping , Humans , Mice , Mice, Transgenic , Molecular Sequence Data , Motor Neurons/cytology , RNA, Messenger/metabolism , Synaptic Vesicles/genetics , Synaptic Vesicles/metabolism , Vesicular Acetylcholine Transport Proteins
SELECTION OF CITATIONS
SEARCH DETAIL
...