Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
Respir Physiol Neurobiol ; 188(1): 21-8, 2013 Aug 01.
Article in English | MEDLINE | ID: mdl-23648475

ABSTRACT

Perinatal inflammation is associated with respiratory morbidity. Immune modulation of brainstem respiratory control centers may provide a link for this pathobiology. We exposed 11-day old rats to intratracheal lipopolysaccharide (LPS, 0.5 µg/g) to test the hypothesis that intrapulmonary inflammation increases expression of the proinflammatory cytokine IL-1ß within respiratory-related brainstem regions. Intratracheal LPS resulted in a 32% increase in IL-1ß protein expression in the medulla oblongata. In situ hybridization showed increased intensity of IL-1ß mRNA but no change in neuronal numbers. Co-localization experiments showed that hypoglossal neurons express IL-1ß mRNA and immunostaining showed a 43% increase in IL-1ß protein-expressing cells after LPS exposure. LPS treatment also significantly increased microglial cell numbers though they did not express IL-1ß mRNA. LPS-induced brainstem expression of neuronal IL-1ß mRNA and protein may have implications for our understanding of the vulnerability of neonatal respiratory control in response to a peripheral proinflammatory stimulus.


Subject(s)
Brain Stem/metabolism , Gene Expression Regulation , Hypoglossal Nerve/metabolism , Interleukin-1beta/biosynthesis , Pneumonia/metabolism , Animals , Animals, Newborn , Brain Stem/drug effects , Hypoglossal Nerve/drug effects , Lipopolysaccharides/toxicity , Pneumonia/chemically induced , Rats , Rats, Sprague-Dawley
2.
Respir Res ; 13: 68, 2012 Aug 07.
Article in English | MEDLINE | ID: mdl-22870905

ABSTRACT

BACKGROUND: Hyperoxia is shown to impair airway relaxation via limiting L-arginine bioavailability to nitric oxide synthase (NOS) and reducing NO production as a consequence. L-arginine can also be synthesized by L-citrulline recycling. The role of L-citrulline supplementation was investigated in the reversing of hyperoxia-induced impaired relaxation of rat tracheal smooth muscle (TSM). METHODS: Electrical field stimulation (EFS, 2-20 V)-induced relaxation was measured under in vitro conditions in preconstricted tracheal preparations obtained from 12 day old rat pups exposed to room air or hyperoxia (>95% oxygen) for 7 days supplemented with L-citrulline or saline (in vitro or in vivo). The role of the L-citrulline/L-arginine cycle under basal conditions was studied by incubation of preparations in the presence of argininosuccinate synthase (ASS) inhibitor [α-methyl-D, L-aspartate, 1 mM] or argininosuccinate lyase inhibitor (ASL) succinate (1 mM) and/or NOS inhibitor [Nω-nitro-L-arginine methyl ester; 100 µM] with respect to the presence or absence of L-citrulline (2 mM). RESULTS: Hyperoxia impaired the EFS-induced relaxation of TSM as compared to room air control (p < 0.001; 0.5 ± 0.1% at 2 V to 50.6 ± 5.7% at 20 V in hyperoxic group: 0.7 ± 0.2 at 2 V to 80.0 ± 5.6% at 20 V in room air group). Inhibition of ASS or ASL, and L-citrulline supplementation did not affect relaxation responses under basal conditions. However, inhibition of NOS significantly reduced relaxation responses (p < 0.001), which were restored to control level by L-citrulline. L-citrulline supplementation in vivo and in vitro also reversed the hyperoxia-impaired relaxation. The differences were significant (p <0.001; 0.8 ± 0.3% at 2 V to 47.1 ± 4.1% at 20 V without L-citrulline; 0.9 ± 0.3% at 2 V to 68.2 ± 4.8% at 20 V with L-citrulline). Inhibition of ASS or ASL prevented this effect of L-citrulline. CONCLUSION: The results indicate the presence of an L-citrulline/L-arginine cycle in the airways of rat pups. L-citrulline recycling does not play a major role under basal conditions in airways, but it has an important role under conditions of substrate limitations to NOS as a source of L-arginine, and L-citrulline supplementation reverses the impaired relaxation of airways under hyperoxic conditions.


Subject(s)
Citrulline/administration & dosage , Dietary Supplements , Hyperoxia/drug therapy , Muscle Relaxation/drug effects , Trachea/drug effects , Animals , Animals, Newborn , Hyperoxia/physiopathology , Muscle Relaxation/physiology , Muscle, Smooth/drug effects , Muscle, Smooth/physiology , Organ Culture Techniques , Random Allocation , Rats , Rats, Sprague-Dawley , Trachea/physiology
3.
Neonatology ; 101(2): 106-15, 2012.
Article in English | MEDLINE | ID: mdl-21952491

ABSTRACT

BACKGROUND: Prolonged exposure of immature lungs to hyperoxia contributes to neonatal lung injury and airway hyperreactivity. We have previously demonstrated that neonatal exposure of rat pups to ≥95% O2 impairs airway relaxation due to disruption of nitric oxide (NO)-cyclic guanosine monophosphate (cGMP) signaling. OBJECTIVE: We now hypothesize that these impaired relaxation responses are secondary to hyperoxia-induced upregulation of arginase, which competes with NO synthase for L-arginine. METHODS: Rat pups were exposed to moderate neonatal hyperoxia (50% O2) or room air for 7 days from birth. In additional hyperoxic and room air groups, exogenous L-arginine (300 mg/kg/day i.p.) or arginase inhibitor (Nω-hydroxy-nor-arginine, 30 mg/kg/day i.p.) were administered daily. After 7 days, animals were anesthetized and sacrificed either for preparation of lung parenchymal strips or lung perfusion. RESULTS: In response to electrical field stimulation (EFS), bethanechol-preconstricted lung parenchymal strips from hyperoxic pups exhibited significantly reduced relaxation compared to room air controls. Supplementation of L-arginine or arginase blockade restored hyperoxia-induced impairment of relaxation. Expression of arginase I in airway epithelium was increased in response to hyperoxia but reduced by arginase blockade. Arginase activity was also significantly increased in hyperoxic lungs as compared to room air controls and reduced following arginase blockade. EFS-induced production of NO was decreased in hyperoxia-exposed airway smooth muscle and restored by arginase blockade. CONCLUSION: These data suggest that NO-cGMP signaling is disrupted in neonatal rat pups exposed to even moderate hyperoxia due to increased arginase activity and consequent decreased bioavailability of the substrate L-arginine. We speculate that supplementation of arginine and/or inhibition of arginase may be a useful therapeutic tool to prevent or treat neonatal lung injury.


Subject(s)
Arginase/physiology , Hyperoxia/physiopathology , Lung/enzymology , Lung/physiology , Muscle Relaxation/physiology , Animals , Animals, Newborn , Arginase/antagonists & inhibitors , Arginase/biosynthesis , Arginine/analogs & derivatives , Arginine/pharmacology , Bethanechol/pharmacology , Electric Stimulation , Hyperoxia/metabolism , Lung/cytology , Muscle, Smooth/drug effects , Muscle, Smooth/physiopathology , Nitric Oxide/biosynthesis , Parasympathomimetics/pharmacology , Rats , Rats, Sprague-Dawley , Signal Transduction/drug effects , Up-Regulation
4.
Am J Physiol Lung Cell Mol Physiol ; 301(1): L91-8, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21515660

ABSTRACT

Neurally derived tachykinins such as substance P (SP) play a key role in modulating airway contractility (especially with inflammation). Separately, the neurotrophin brain-derived neurotrophic factor (BDNF; potentially derived from nerves as well as airway smooth muscle; ASM) and its tropomyosin-related kinase receptor, TrkB, are involved in enhanced airway contractility. In this study, we hypothesized that neurokinins and neurotrophins are linked in enhancing intracellular Ca(2+) concentration ([Ca(2+)](i)) regulation in ASM. In rat ASM cells, 24 h exposure to 10 nM SP significantly increased BDNF and TrkB expression (P < 0.05). Furthermore, [Ca(2+)](i) responses to 1 µM ACh as well as BDNF (30 min) effects on [Ca(2+)](i) regulation were enhanced by prior SP exposure, largely via increased Ca(2+) influx (P < 0.05). The enhancing effect of SP on BDNF signaling was blunted by the neurokinin-2 receptor antagonist MEN-10376 (1 µM, P < 0.05) to a greater extent than the neurokinin-1 receptor antagonist RP-67580 (5 nM). Chelation of extracellular BDNF (chimeric TrkB-F(c); 1 µg/ml), as well as tyrosine kinase inhibition (100 nM K252a), substantially blunted SP effects (P < 0.05). Overnight (24 h) exposure of ASM cells to 50% oxygen increased BDNF and TrkB expression and potentiated both SP- and BDNF-induced enhancement of [Ca(2+)](i) (P < 0.05). These results suggest a novel interaction between SP and BDNF in regulating agonist-induced [Ca(2+)](i) regulation in ASM. The autocrine mechanism we present here represents a new area in the development of bronchoconstrictive reflex response and airway hyperreactive disorders.


Subject(s)
Brain-Derived Neurotrophic Factor/metabolism , Muscle, Smooth/metabolism , Respiratory System/metabolism , Substance P/metabolism , Acetylcholine/pharmacology , Animals , Brain-Derived Neurotrophic Factor/pharmacology , Calcium/metabolism , Hyperoxia/metabolism , Intracellular Space/drug effects , Intracellular Space/metabolism , Models, Biological , Muscle, Smooth/cytology , Myocytes, Smooth Muscle/drug effects , Myocytes, Smooth Muscle/metabolism , Protein Binding/drug effects , Rats , Rats, Sprague-Dawley , Receptor, trkB/metabolism , Receptors, Neurokinin-1/metabolism , Receptors, Neurokinin-2/metabolism , Substance P/pharmacology
5.
Am J Physiol Lung Cell Mol Physiol ; 295(2): L348-55, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18515408

ABSTRACT

Prolonged hyperoxic exposure contributes to neonatal lung injury, and airway hyperreactivity is characterized by enhanced contraction and impaired relaxation of airway smooth muscle. Our previous data demonstrate that hyperoxia in rat pups upregulates expression of brain-derived neurotrophic factor (BDNF) mRNA and protein, disrupts NO-cGMP signaling, and impairs cAMP production in airway smooth muscle. We hypothesized that BDNF-tyrosine kinase B (TrkB) signaling plays a functional role in airway hyperreactivity via upregulation of cholinergic mechanisms in hyperoxia-exposed lungs. Five-day-old rat pups were exposed to >or=95% oxygen or room air for 7 days and administered daily tyrosine kinase inhibitor K-252a (50 microg x kg(-1) x day(-1) i.p.) to block BDNF-TrkB signaling or vehicle. Lungs were removed for HPLC measurement of ACh or for in vitro force measurement of lung parenchymal strips. ACh content doubled in hyperoxic compared with room air-exposed lungs. K-252a treatment of hyperoxic pups restored ACh content to room air levels. Hyperoxia increased contraction and impaired relaxation of lung strips in response to incremental electrical field stimulation. K-252a administration to hyperoxic pups reversed this increase in contraction and decrease in relaxation. K-252a or TrkB-Fc was used to block the effect of exogenous BDNF in vitro. Both K-252a and TrkB-Fc blocked the effects of exogenous BDNF. Hyperoxia decreased cAMP and cGMP levels in lung strips, and blockade of BDNF-TrkB signaling restored cAMP but not cGMP to control levels. Therefore, hyperoxia-induced increase in activity of BDNF-TrkB receptor signaling appears to play a critical role in enhancing cholinergically mediated contractile responses of lung parenchyma.


Subject(s)
Brain-Derived Neurotrophic Factor/metabolism , Hyperoxia/metabolism , Lung/metabolism , Muscle Relaxation , Muscle, Smooth/metabolism , Acetylcholine/metabolism , Animals , Animals, Newborn , Carbazoles/pharmacology , Cyclic AMP/metabolism , Cyclic GMP/metabolism , Enzyme Inhibitors/pharmacology , Hyperoxia/pathology , Indole Alkaloids/pharmacology , Lung Injury , Muscle Relaxation/drug effects , Rats , Rats, Sprague-Dawley , Receptor, trkB/agonists , Receptor, trkB/antagonists & inhibitors , Receptor, trkB/metabolism , Signal Transduction/drug effects
6.
Am J Physiol Lung Cell Mol Physiol ; 293(4): L1029-36, 2007 Oct.
Article in English | MEDLINE | ID: mdl-17660329

ABSTRACT

Exposure of immature lungs to hyperoxia for prolonged periods contributes to neonatal lung injury and airway hyperreactivity. We studied the role of disrupted nitric oxide-guanosine 3',5'-cyclic monophosphate (NO-cGMP) signaling in impairing the relaxant responses of lung tissue from hyperoxia-exposed rat pups. Pups were exposed to >/=95% O(2) or room air for 7 days starting from days 1, 5, or 14. The animals were killed, lungs were removed, and 1-mm-thick lung parenchymal strips were prepared. Lung parenchymal strips of room air or hyperoxic pups were preconstricted using bethanechol and then graded electrical field stimulation (EFS) was applied to induce relaxation. EFS-induced relaxation of lung parenchymal strips was greater at 7 and 12 days than at 21 days in room air-exposed rat pups. Hyperoxic exposure significantly reduced relaxation at 7 and 12 days but not 21 days compared with room air exposure. NO synthase blockade with N(omega)-nitro-l-arginine methyl ester diminished relaxant responses in room air but not in hyperoxic pups at 12 days. After incubation with supplemental l-arginine, the relaxation response of hyperoxic strips was restored. cGMP, a key mediator of the NO signaling pathway, also decreased in strips from hyperoxic vs. room air pups and cGMP levels were restored after incubation with supplemental l-arginine. In addition, arginase activity was significantly increased in hyperoxic lung parenchymal strips compared with room air lung parenchymal strips. These data demonstrate disruption of NO-cGMP signaling in neonatal rat pups exposed to hyperoxia and show that bioavailability of the substrate l-arginine is implicated in the predisposition of this model to airway hyperreactivity.


Subject(s)
Animals, Newborn , Cyclic GMP/metabolism , Hyperoxia/physiopathology , Lung/physiopathology , Nitric Oxide/metabolism , Signal Transduction , Aging , Animals , Animals, Newborn/growth & development , Arginase/metabolism , Arginine/pharmacology , Electric Stimulation , Enzyme Inhibitors/pharmacology , Hyperoxia/metabolism , In Vitro Techniques , Lung/drug effects , Lung/metabolism , NG-Nitroarginine Methyl Ester/pharmacology , Nitric Oxide Synthase/antagonists & inhibitors , Rats , Rats, Sprague-Dawley
7.
Brain Res ; 1071(1): 42-53, 2006 Feb 03.
Article in English | MEDLINE | ID: mdl-16413509

ABSTRACT

During early development, adenosine contributes to the occurrence of respiratory depression and recurrent apneas. Recent physiological studies indicate that GABAergic mechanisms may be involved in this inhibitory action of adenosine, via their A(2A) receptors. In the present study, in situ hybridization with ribonucleotide probes for A(2A) receptor (A(2A)R) mRNA was combined with the immunolabeling technique for parvalbumin and transneuronal retrograde tracing method using green fluorescent protein expressing pseudorabies virus (GFP-PRV) to (1) characterize age-dependent changes in the expression of adenosine A(2A)Rs mRNA in brain stem regions where GABAergic neurons are located; (2) determine whether GABA-containing neurons express A(2A)R mRNA traits, and (3) identify whether bulbospinal GABAergic neurons projecting to phrenic nuclei contain A(2A)R mRNA. Results revealed expression of A(2A) receptors in regions of medulla oblongata containing GABAergic neurons, namely in the ventral aspect of the medulla, within the Bötzinger region and caudal to it, the gigantocellular reticular nucleus, midline neurons and the caudal ventrolateral medulla oblongata. Furthermore, a subpopulation of identified GABAergic neurons, projecting to the phrenic motor nuclei, possess A(2A)R mRNA. It is concluded that adenosine A(2A)Rs expressed by GABAergic neurons are likely to play a role in mediating adenosine-induced respiratory depression.


Subject(s)
Gene Expression Regulation, Developmental/physiology , Medulla Oblongata/cytology , Neurons/metabolism , Receptors, Adenosine A2/metabolism , gamma-Aminobutyric Acid/metabolism , Age Factors , Animals , Blotting, Northern/methods , Cell Count/methods , Diaphragm/innervation , Diaphragm/metabolism , Diaphragm/virology , Glutamate Decarboxylase/genetics , Glutamate Decarboxylase/metabolism , Green Fluorescent Proteins/metabolism , Herpesvirus 1, Suid/physiology , Immunohistochemistry/methods , In Situ Hybridization/methods , Medulla Oblongata/growth & development , Models, Neurological , Neural Pathways/metabolism , Neural Pathways/virology , Parvalbumins/genetics , Parvalbumins/metabolism , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Receptors, Adenosine A2/genetics , Reverse Transcriptase Polymerase Chain Reaction/methods , Statistics, Nonparametric
8.
Brain Res ; 1044(2): 133-43, 2005 May 24.
Article in English | MEDLINE | ID: mdl-15885212

ABSTRACT

Recent evidence indicates that brain-derived neurotrophic factor (BDNF) is present in neurons and may affect neurotransmitter release, cell excitability, and synaptic plasticity via activation of tyrosine kinase B (TrkB) receptors. However, whether airway-related vagal preganglionic neurons (AVPNs) produce BDNF and contain TrkB receptors is not known. Hence, in ferrets, we examined BDNF and TrkB receptor expression in identified AVPNs using in situ hybridization and immunohistochemistry. BDNF protein levels were measured within the rostral nucleus ambiguus (rNA) region by ELISA. We observed that the subpopulation of AVPNs, identified by neuroanatomical tract tracing, within the rNA region express BDNF mRNA, BDNF protein, as well as TrkB receptor. In addition, brain tissue from the rNA region contained measurable amounts of BDNF that were comparable to the hippocampal region of the brain. These data indicate, for the first time, that the BDNF-TrkB system is expressed by AVPNs and may play a significant role in regulating cholinergic outflow to the airways.


Subject(s)
Autonomic Fibers, Preganglionic/metabolism , Brain-Derived Neurotrophic Factor/metabolism , Neuronal Plasticity/physiology , Receptor, trkB/metabolism , Trachea/innervation , Vagus Nerve , Animals , Brain Stem/cytology , Brain Stem/metabolism , Brain-Derived Neurotrophic Factor/genetics , Cell Count , Cholera Toxin/metabolism , Enzyme-Linked Immunosorbent Assay/methods , Ferrets , Immunohistochemistry/methods , In Situ Hybridization/methods , Male , Neural Networks, Computer , Receptor, trkB/genetics , Trachea/metabolism
9.
Respir Physiol Neurobiol ; 147(1): 19-30, 2005 May 12.
Article in English | MEDLINE | ID: mdl-15848120

ABSTRACT

Recent in vitro data suggest that astrocytes may modulate respiration. To examine this question in vivo, we treated 5-day-old rat pups with methionine sulfoximine (MS), a compound that alters carbohydrate and glutamate metabolism in astrocytes, but not neurons. MS-treated pups displayed a reduced breathing frequency (f) in baseline conditions relative to saline-treated pups. Hypercapnia (5% CO(2)) increased f in both groups, but f still remained significantly lower in the MS-treated group. No differences between treatment groups in the responses to hypoxia (8% O(2)) were observed. Also, MS-treated rats showed an enhanced accumulation of glycogen in neurons of the facial nucleus, the nucleus ambiguus, and the hypoglossal nucleus, structures that regulate respiratory activity and airway patency. An altered transfer of nutrient molecules from astrocytes to neurons may underlie these effects of MS, although direct effects of MS upon neurons or upon peripheral structures that regulate respiration cannot be completely ruled out as an explanation.


Subject(s)
Astrocytes/drug effects , Brain/drug effects , Hypercapnia/physiopathology , Methionine Sulfoximine/toxicity , Respiration/drug effects , Animals , Animals, Newborn , Brain/metabolism , Brain/pathology , Glycogen/metabolism , In Situ Hybridization , Neurons/drug effects , Neurons/pathology , RNA, Messenger/analysis , Rats , Receptors, Neurokinin-1/drug effects , Receptors, Neurokinin-1/metabolism , Respiratory Function Tests
10.
J Alzheimers Dis ; 7(2): 159-71; discussion 173-80, 2005 Apr.
Article in English | MEDLINE | ID: mdl-15851854

ABSTRACT

Prion diseases are associated with the accumulation of a misfolded, protease resistant form of the prion protein, PrPres. In humans there are a variety of different prion related diseases that are sporadic, inherited, or acquired by infection. Gerstmann-Straussler-Sheinker syndrome (GSS) is an inherited prion disease in which PrPres accumulates as amorphous aggregates as well as in amyloid plaques. GSS has been associated with a variety of point mutations in the prion protein: 102, 105, 117, 131, 145, 187, 198, 202, 212, 217, and 232. The F198S mutation was discovered in a large Indiana kindred. Previous studies in vitro have shown that the 198 mutation results in structural instability of the prion protein. In the current study, we demonstrate in a cell model that the F198S mutant protein can be folded properly in a cellular context, but is unable to refold to a native state after denaturation. Further, the F198S mutation significantly affects glycosylation of the mutant protein.


Subject(s)
Gerstmann-Straussler-Scheinker Disease/genetics , Glycosylation , Point Mutation/genetics , PrPSc Proteins/metabolism , Prions/genetics , Amyloid/genetics , Binding Sites , Brain/metabolism , Cell Extracts , DNA Mutational Analysis , DNA Primers/genetics , Endopeptidase K/metabolism , Gerstmann-Straussler-Scheinker Disease/metabolism , Humans , Immunoprecipitation , Protein Conformation , Protein Folding , Protein Precursors/metabolism , Subcellular Fractions/metabolism
11.
J Alzheimers Dis ; 2(1): 27-35, 2000 Mar.
Article in English | MEDLINE | ID: mdl-12214108

ABSTRACT

The abnormal form of the prion protein has increased resistance to protease digestion and is insoluble in non-ionic detergents. The normal prion protein is modified by the non-obligatory addition of two N-linked glycans. One pathogenic mutation, Thr to Ala at residue 183 of the human prion protein, blocks addition of the first glycan to the Asp residue 181. This mutation has been reported to result in intracellular retention of the mutant protein and its acquisition of pathogenic properties, presumably due to the lack of the glycan. We report that the lack of the N-linked glycan at residue 181 is not responsible for the block in transport or the acquisition of pathogen-like properties, rather, the Thr to Ala mutation is itself the probable cause of the pathogenic phenotype.

SELECTION OF CITATIONS
SEARCH DETAIL
...