Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 47
Filter
Add more filters










Publication year range
1.
Mil Med ; 185(Suppl 1): 243-247, 2020 01 07.
Article in English | MEDLINE | ID: mdl-32074348

ABSTRACT

INTRODUCTION: Blast-induced mild traumatic brain injury was generated in a mouse model using a shock tube to investigate recovery and axonal injury from single blast. METHODS: A supersonic helium wave hit the head of anesthetized male young adult mice with a reflected pressure of 69 psi for 0.2 ms on Day 1. Subsequently, the mice were cardioperfused on Days 2, 5, or 12. The isolated brains were subjected to diffusion tensor imaging. Reduced fractional anisotropy (FA) indicated axonal injury. RESULTS: After single blast, FA showed a biphasic response in the corpus callosum with decrease on Days 2 and 12 and increase on Day 5. CONCLUSIONS: Blast-induced mild traumatic brain injury in a mouse model follows a biphasic FA response within 12 days after a single blast similar to that reported for human subjects.


Subject(s)
Anisotropy , Blast Injuries/complications , Brain Concussion/etiology , Animals , Blast Injuries/physiopathology , Brain Concussion/physiopathology , Diffusion Tensor Imaging/methods , Disease Models, Animal , Explosions/statistics & numerical data , Mice
2.
Shock ; 53(6): 744-753, 2020 06.
Article in English | MEDLINE | ID: mdl-31689268

ABSTRACT

Mild traumatic brain injury (TBI) is an important public health problem generated by closed head injury. This study is focused on the impact of blast-induced mild TBI on auditory trace and delay fear conditioning, models of declarative and non-declarative memory, respectively, and the correlation of conditioned freezing and fractional anisotropy, a measure of axonal state. A supersonic helium pressure wave was generated by a shock tube to blast 8-week-old male mice on Day 1 for 1.4 msec with an incident pressure of 16 psi, corresponding to a reflected pressure of 56.9 psi at the mouse head. On Day 3, the mice were subjected to auditory trace- or delay-fear conditioning. On Day 4, contextual freezing in the trained context, and precue and cued freezing in a novel context were determined. After cardiac perfusion on Day 5, ex vivo images were obtained with diffusion tensor imaging at 14.1 Tesla. We observed that delay fear conditioning prevented or reversed the decrease in fractional anisotropy in both the medial and lateral corpus callosum suggesting axonal stabilization of potentially behavioral therapeutic significance. Moderately strong and statistically significant Pearson correlations were found between fractional anisotropy and contextual freezing in the medial and lateral corpus callosum of blasted and sham-blasted delay- or trace-fear conditioned mice. Thus, contextual freezing is a neurobehavioral biomarker for axonal injury in mild TBI and is a reliable and high-throughput behavioral assay for the evaluation of potential therapeutics to treat mild TBI.


Subject(s)
Axons/pathology , Blast Injuries/pathology , Brain Concussion/pathology , Animals , Anisotropy , Biomarkers , Blast Injuries/diagnosis , Brain/diagnostic imaging , Brain/pathology , Brain Concussion/diagnosis , Conditioning, Classical , Diffusion Tensor Imaging , Disease Models, Animal , Fear , Freezing Reaction, Cataleptic , Male , Mice , Mice, Inbred C57BL
3.
Am J Physiol Heart Circ Physiol ; 307(5): H689-700, 2014 Sep 01.
Article in English | MEDLINE | ID: mdl-25015964

ABSTRACT

Urocortin 2 (Ucn2) is a cardioactive peptide exhibiting beneficial effects in normal and failing heart. In cardiomyocytes, it elicits cAMP- and Ca(2+)-dependent positive inotropic and lusitropic effects. We tested the hypothesis that, in addition, Ucn2 activates cardiac nitric oxide (NO) signaling and elucidated the underlying signaling pathways and mechanisms. In isolated rabbit ventricular myocytes, Ucn2 caused concentration- and time-dependent increases in phosphorylation of Akt (Ser473, Thr308), endothelial NO synthase (eNOS) (Ser1177), and ERK1/2 (Thr202/Tyr204). ERK1/2 phosphorylation, but not Akt and eNOS phosphorylation, was suppressed by inhibition of MEK1/2. Increased Akt phosphorylation resulted in increased Akt kinase activity and was mediated by corticotropin-releasing factor 2 (CRF2) receptors (astressin-2B sensitive). Inhibition of phosphatidylinositol 3-kinase (PI3K) diminished both Akt as well as eNOS phosphorylation mediated by Ucn2. Inhibition of protein kinase A (PKA) reduced Ucn2-induced phosphorylation of eNOS but did not affect the increase in phosphorylation of Akt. Conversely, direct receptor-independent elevation of cAMP via forskolin increased phosphorylation of eNOS but not of Akt. Ucn2 increased intracellular NO concentration ([NO]i), [cGMP], [cAMP], and cell shortening. Inhibition of eNOS suppressed the increases in [NO]i and cell shortening. When both PI3K-Akt and cAMP-PKA signaling were inhibited, the Ucn2-induced increases in [NO]i and cell shortening were attenuated. Thus, in rabbit ventricular myocytes, Ucn2 causes activation of cAMP-PKA, PI3K-Akt, and MEK1/2-ERK1/2 signaling. The MEK1/2-ERK1/2 pathway is not required for stimulation of NO signaling in these cells. The other two pathways, cAMP-PKA and PI3K-Akt, converge on eNOS phosphorylation at Ser1177 and result in pronounced and sustained cellular NO production with subsequent stimulation of cGMP signaling.


Subject(s)
Heart Ventricles/metabolism , Myocytes, Cardiac/metabolism , Nitric Oxide Synthase Type III/metabolism , Nitric Oxide/metabolism , Urocortins/metabolism , Animals , Cyclic AMP/metabolism , Cyclic AMP-Dependent Protein Kinases/metabolism , Cyclic GMP/metabolism , Heart Ventricles/cytology , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Phosphorylation , Proto-Oncogene Proteins c-akt/metabolism , Rabbits , Receptors, Corticotropin-Releasing Hormone/metabolism , Serine/metabolism , Signal Transduction
4.
Horm Behav ; 64(3): 519-26, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23845323

ABSTRACT

Corticotropin-releasing factor (CRF) plays an essential role in coordinating the autonomic, endocrine and behavioral responses to stressors. In this study, we investigated the role of CRF within the medial prefrontal cortex (mPFC) in modulating unconditioned defensive behaviors, by examining the effects of microinfusing cortagine a selective type-1 CRF receptor (CRF1) agonist, or acidic-astressin a preferential CRF1 antagonist, into the mPFC in male CD-1 mice exposed to a live predator (rat exposure test--RET). Cortagine microinfusions significantly reduced several indices of defense, including avoidance and freezing, suggesting a specific role for CRF1 within the infralimbic and prelimbic regions of the mPFC in modulating unconditioned behavioral responsivity to a predator. In contrast, microinfusions of acidic-astressin failed to alter defensive behaviors during predator exposure in the RET. Cortagine microinfusions also reduced Fos protein production in the medial, central and basomedial, but not basolateral subnuclei of the amygdala in mice exposed to the rat predatory threat stimulus. These results suggest that CRF1 activation within the mPFC attenuates predator-induced unconditioned anxiety-like defensive behaviors, likely via inhibition of specific amygdalar nuclei. Furthermore, the present findings suggest that the mPFC represents a unique neural region whereby activation of CRF1 produces behavioral effects that contrast with those elicited following systemic administration of CRF1 agonists.


Subject(s)
Amygdala/drug effects , Behavior, Animal/drug effects , Corticotropin-Releasing Hormone/pharmacology , Escape Reaction/drug effects , Oncogene Proteins v-fos/metabolism , Prefrontal Cortex/drug effects , Recombinant Fusion Proteins/pharmacology , Amygdala/cytology , Amygdala/metabolism , Animals , Cell Nucleus/drug effects , Cell Nucleus/metabolism , Dominance-Subordination , Food Chain , Infusions, Intraventricular , Male , Mice , Prefrontal Cortex/physiology , Rats , Rats, Long-Evans
5.
Br J Pharmacol ; 162(2): 544-56, 2011 Jan.
Article in English | MEDLINE | ID: mdl-20942811

ABSTRACT

BACKGROUND AND PURPOSE: Urocortin 2 is beneficial in heart failure, but the underlying cellular mechanisms are not completely understood. Here we have characterized the functional effects of urocortin 2 on mouse cardiomyocytes and elucidated the underlying signalling pathways and mechanisms. EXPERIMENTAL APPROACH: Mouse ventricular myocytes were field-stimulated at 0.5 Hz at room temperature. Fractional shortening and [Ca²(+)](i) transients were measured by an edge detection and epifluorescence system respectively. Western blots were carried out on myocyte extracts with antibodies against total phospholamban (PLN) and PLN phosphorylated at serine-16. KEY RESULTS: Urocortin 2 elicited time- and concentration-dependent positive inotropic and lusitropic effects (EC50 : 19 nM) that were abolished by antisauvagine-30 (10 nM, n= 6), a specific antagonist of corticotrophin releasing factor (CRF) CRF2 receptors. Urocortin 2 (100 nM) increased the amplitude and decreased the time constant of decay of the underlying [Ca²(+)](i) transients. Urocortin 2 also increased PLN phosphorylation at serine-16. H89 (2 µM) or KT5720 (1 µM), two inhibitors of protein kinase A (PKA), as well as KN93 (1 µM), an inhibitor of Ca²(+)/calmodulin-dependent protein kinase II (CaMKII), suppressed the urocortin 2 effects on shortening and [Ca²(+)](i) transients. In addition, urocortin 2 also elicited arrhythmogenic events consisting of extra cell shortenings and extra [Ca²(+)](i) increases in diastole. Urocortin 2-induced arrhythmogenic events were significantly reduced in cells pretreated with KT5720 or KN93. CONCLUSIONS AND IMPLICATIONS: Urocortin 2 enhanced contractility in mouse ventricular myocytes via activation of CRF2 receptors in a cAMP/PKA- and Ca²(+)/CaMKII-dependent manner. This enhancement was accompanied by Ca²(+)-dependent arrhythmogenic effects mediated by PKA and CaMKII.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Cyclic AMP-Dependent Protein Kinases/metabolism , Myocardial Contraction/drug effects , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/physiology , Urocortins/pharmacology , Aged , Aged, 80 and over , Animals , Arrhythmias, Cardiac/chemically induced , Arrhythmias, Cardiac/metabolism , Calcium/metabolism , Cardiotonic Agents/pharmacology , Cyclic AMP/metabolism , Female , Heart Atria/drug effects , Heart Atria/metabolism , Heart Ventricles , Humans , Male , Mice , Mice, Inbred C57BL , Middle Aged , Muscle Relaxation/drug effects , Myocytes, Cardiac/enzymology , Receptors, Corticotropin-Releasing Hormone/metabolism
6.
Horm Behav ; 58(2): 241-9, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20298695

ABSTRACT

Early-life stress produces an anxiogenic profile in adulthood, presumably by activating the otherwise quiescent hypothalamic-pituitary-adrenal (HPA) axis during the vulnerable 'stress hyporesponsive period'. While the long-term effects of such early-life manipulations have been extensively characterized, little is known of the short-term effects. Here, we compared the short-term effects of two durations of maternal separation stress and one unseparated group (US) on behavioral and physiological indices of the stress response in rat pups. Separations included 3h on each of 12days, from postnatal day (PND) 2 to 13 (MS2-13) and 3days of daily, 6-h separation from PND11-13 (MS11-13). On PND14 (Experiment 1), both MS2-13 and MS11-13 produced marked reductions in freezing toward an adult male conspecific along with reduced levels of glucocorticoid type 2 (GR) and CRF type-1 (CRF(1)) receptor mRNA in the hippocampus. Group MS2-13 but not MS11-13 produced deficits in stressor-induced corticosterone secretion, accompanied by reductions in body weight. Our results suggest that GR and/or CRF(1) levels, not solely the magnitude of corticosterone secretion, may be involved in the modulation of freezing. In a second experiment, we aimed to extend these findings by testing male and female separated and unseparated pups' unconditioned defensive behaviors to cat odor on PND26, and subsequent cue+context conditioning and extinction throughout postnatal days 27-32. Our results show that maternal separation produced reductions in unconditioned freezing on PND26, with MS2-13 showing stronger deficits than MS11-13. However, separation did not affect any other defensive behaviors. Furthermore, separated rats failed to show conditioned freezing, although they did avoid the no-odor block conditioned cue. There were no sex differences other than weight. We suggest that maternal separation may have produced these changes by disrupting normal development of hippocampal regions involved in olfactory-mediated freezing, not in mechanisms of learning and memory per se. These findings may have direct relevance for understanding the mechanisms by which early-life adverse experiences produce short-term and lasting psychopathologies.


Subject(s)
Behavior, Animal/physiology , Hippocampus/metabolism , Maternal Deprivation , Stress, Psychological/metabolism , Stress, Psychological/psychology , Aging , Animals , Animals, Newborn , Body Weight , Cats , Conditioning, Classical/physiology , Extinction, Psychological/physiology , Female , Freezing Reaction, Cataleptic , Male , Olfactory Perception/physiology , Rats , Rats, Long-Evans , Social Behavior , Time Factors
7.
Eur J Pharmacol ; 632(1-3): 1-6, 2010 Apr 25.
Article in English | MEDLINE | ID: mdl-20132811

ABSTRACT

Corticotropin-releasing factor (CRF), a 41 amino acid peptide, was discovered as a key signal in mediating neuroendocrine, autonomic, and behavioral responses to stress. It was revealed later that there exist additional CRF-like peptides, termed urocortins. The CRF receptor subtype 1 (CRF1 receptor) is predominant in the brain whereas subtype 2 (CRF2 receptor) is highly expressed in the brain and the heart. Both centrally and peripherally administered CRF and urocortins produce significant hemodynamic effects via activation of CRF receptors in the brain and the heart. CRF and urocortins are important neural and cardioactive hormones, and are potentially useful therapy for heart failure.


Subject(s)
Receptors, Corticotropin-Releasing Hormone/metabolism , Urocortins , Animals , Autonomic Nervous System/metabolism , Brain/metabolism , Corticotropin-Releasing Hormone/metabolism , Corticotropin-Releasing Hormone/physiology , Mice , Peptides/metabolism , Rats , Receptors, Corticotropin-Releasing Hormone/physiology
8.
Psychoneuroendocrinology ; 35(6): 887-95, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20036073

ABSTRACT

CRF receptor subtype 1 (CRF1), abundantly expressed in the central nervous system, has been implicated in defensive behavior in rodents. Pharmacological activation of CRF1 by peptidic agonists results in enhancement of anxiety-like behavior. However, receptor specificity of commonly used agonists was confounded by significant affinity to other receptors and widely used laboratory tests of experimental anxiety suffer from artificial aversive stimulation (e.g. electric shock), and limited measures of anxiety-like behavior. We used the recently developed, CRF1-selective agonist cortagine in a mouse model of defensive behaviors under semi-natural conditions, the rat exposure test (RET). Cortagine was injected bilaterally into the cerebral ventricles (i.c.v.) of male C57Bl/6J mice, 20min before exposure to a rat in specifically designed box that evokes a wide variety of defensive behaviors such as active/passive avoidance, freezing, risk assessment, and burying. Pre-injection of the CRF receptor antagonist acidic astressin was used to test for receptor specificity of the observed cortagine effects. A control experiment with no rat present was performed to test for baseline effects of cortagine in the exposure setup. Cortagine dose-dependently enhanced passive avoidance and freezing while burying was decreased. CRF receptor antagonism reliably blocked the effects of cortagine. Our results confirm previous findings of anxiogenic-like effects of cortagine, and demonstrate the usefulness of the RET in investigating differential pattering of drug-induced anxiety-like behavior in mice. In conclusion, our results suggest that CRF1 activation in forebrain areas promotes passive coping with the natural threat presented in the RET.


Subject(s)
Adaptation, Psychological/drug effects , Agonistic Behavior/drug effects , Corticotropin-Releasing Hormone/pharmacology , Receptors, Corticotropin-Releasing Hormone/agonists , Recombinant Fusion Proteins/pharmacology , Animals , Corticotropin-Releasing Hormone/administration & dosage , Dose-Response Relationship, Drug , Drug Interactions , Injections, Intraventricular , Male , Mice , Mice, Inbred C57BL , Peptide Fragments/pharmacology , Rats , Rats, Long-Evans , Receptors, Corticotropin-Releasing Hormone/antagonists & inhibitors , Recombinant Fusion Proteins/administration & dosage
9.
Neuropsychopharmacology ; 34(6): 1416-26, 2009 May.
Article in English | MEDLINE | ID: mdl-18843268

ABSTRACT

The neuropeptide corticotropin-releasing factor (CRF) plays a critical role in the proper functioning of the stress response system through its actions on its receptors, CRF receptor 1 (CRF1) and CRF receptor 2 (CRF2), located at multiple anatomical sites. Clinical data indicate that stress response dysfunctions, such as excessive CRF activity and hyperstimulation of CRF1, are present in a range of stress-related disorders, including depression and anxiety disorders. Our previous work along with that of other laboratories has demonstrated that mice deficient in CRF2 (CRF2-/-) display increased anxiety and depression-like behaviors. In this study, we found CRF2-/- mice display increased hippocampal levels of activated (phosphorylated) mitogen-activated protein kinase (MAP kinase)/ERK kinase (MEK), extracellular signal-regulated kinases 1 and 2 (ERK1/2), and ribosomal protein S6 kinases 1 (RSK1). These changes can be explained by overactive hippocampal CRF1, in view of the finding that the application of the nonselective CRF receptor antagonist [Glu(11,16)] astressin ([Glu(11,16)]Ast) into the dorsal hippocampus of mutant mice returned the levels of the phosphorylated proteins to baseline. Moreover, inhibition of the hippocampal MEK/ERK pathway with the specific MEK inhibitor U0126, decreased depression-like behaviors in the forced swim test and tail suspension test of CRF2-/- mice. Similarly, treatment with [Glu(11,16)]Ast reversed depression phenotype of CRF2-/- mice without affecting the phenotype of wild-type littermates. Our results support an involvement of CRF receptors in the development of depression, such that elevated hippocampal CRF1 activity, in the absence of CRF2, produces a depression-dominated phenotype through the activation of the MEK/ERK pathway.


Subject(s)
Depression/metabolism , Hippocampus/metabolism , MAP Kinase Signaling System/physiology , Receptors, Corticotropin-Releasing Hormone/metabolism , Animals , Butadienes/pharmacology , Corticotropin-Releasing Hormone/pharmacology , Depression/psychology , Enzyme Inhibitors/pharmacology , Gene Expression , Hippocampus/drug effects , MAP Kinase Kinase Kinases/antagonists & inhibitors , MAP Kinase Kinase Kinases/metabolism , MAP Kinase Signaling System/drug effects , Mice , Mice, Knockout , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Mitogen-Activated Protein Kinases/metabolism , Nitriles/pharmacology , Peptide Fragments/pharmacology , Phosphorylation , Receptors, Corticotropin-Releasing Hormone/antagonists & inhibitors , Receptors, Corticotropin-Releasing Hormone/genetics , Ribosomal Protein S6 Kinases, 90-kDa/metabolism
10.
J Neurochem ; 107(4): 1158-67, 2008 Nov.
Article in English | MEDLINE | ID: mdl-18823372

ABSTRACT

Previous studies have shown that tumor necrosis factor-alpha (TNF-alpha) induces neuroprotection against excitotoxic damage in primary cortical neurons via sustained nuclear factor-kappa B (NF-kappaB) activation. The transcription factor NF-kappaB can regulate the expression of small conductance calcium-activated potassium (K(Ca)) channels. These channels reduce neuronal excitability and as such may yield neuroprotection against neuronal overstimulation. In the present study we investigated whether TNF-alpha-mediated neuroprotective signaling is inducing changes in the expression of small conductance K(Ca) channels. Interestingly, the expression of K(Ca)2.2 channel was up-regulated by TNF-alpha treatment in a time-dependent manner whereas the expression of K(Ca)2.1 and K(Ca)2.3 channels was not altered. The increase in K(Ca)2.2 channel expression after TNF-alpha treatment was shown to be dependent on TNF-R2 and NF-kappaB activation. Furthermore, activation of small conductance K(Ca) channels by 6,7-dichloro-1H-indole-2,3-dione 3-oxime or cyclohexyl-[2-(3,5-dimethyl-pyrazol-1-yl)-6-methyl-pyrimidin-4-yl]-amine-induced neuroprotection against a glutamate challenge. Treatment with the small conductance K(Ca) channel blocker apamin or K(Ca)2.2 channel siRNA reverted the neuroprotective effect elicited by TNF-alpha. We conclude that treatment of primary cortical neurons with TNF-alpha leads to increased K(Ca)2.2 channel expression which renders neurons more resistant to excitotoxic cell death.


Subject(s)
Glutamic Acid/toxicity , NF-kappa B/metabolism , Neurotoxicity Syndromes/etiology , Neurotoxicity Syndromes/prevention & control , Small-Conductance Calcium-Activated Potassium Channels/metabolism , Tumor Necrosis Factor-alpha/metabolism , Up-Regulation/physiology , Animals , Cell Survival/drug effects , Cell Survival/genetics , Cells, Cultured , Cerebral Cortex/cytology , Embryo, Mammalian , Enzyme Inhibitors/pharmacology , Mice , Mice, Inbred C57BL , Mice, Knockout , Neurons/drug effects , Neuroprotective Agents/metabolism , Neuroprotective Agents/pharmacology , Nitriles/pharmacology , RNA, Small Interfering/pharmacology , Receptors, Tumor Necrosis Factor, Type I/deficiency , Receptors, Tumor Necrosis Factor, Type II/deficiency , Small-Conductance Calcium-Activated Potassium Channels/genetics , Subcellular Fractions/drug effects , Subcellular Fractions/metabolism , Sulfones/pharmacology , Time Factors , Tumor Necrosis Factor-alpha/pharmacology , Up-Regulation/drug effects , Up-Regulation/genetics
11.
J Neurochem ; 106(6): 2312-21, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18624921

ABSTRACT

Throughout the CNS, small conductance Ca(2+)-activated potassium (SK) channels modulate firing frequency and neuronal excitability. We have identified a novel, shorter isoform of standard SK2 (SK2-std) in mouse brain which we named SK2-sh. SK2-sh is alternatively spliced at exon 3 and therefore lacks 140 amino acids, which include transmembrane domains S3, S4 and S5, compared with SK2-std. Western blot analysis of mouse hippocampal tissue revealed a 47 kDa protein product as predicted for SK2-sh along with a 64 kDa band representing the standard SK2 isoform. Electrophysiological recordings from transiently expressed SK2-sh revealed no functional channel activity or interaction with SK2-std. With the help of real-time PCR, we found significantly higher expression levels of SK2-sh mRNA in cortical tissue from AD cases when compared with age-matched controls. A similar increase in SK2-sh expression was induced in cortical neurons from mice by cytokine exposure. Substantial clinical evidence suggests that excess cytokines are centrally involved in the pathogenesis of Alzheimer's disease. Thus, SK2-sh as a downstream target of cytokines, provide a promising target for additional investigation regarding potential therapeutic intervention.


Subject(s)
Alternative Splicing/genetics , Brain/metabolism , Cytokines/metabolism , Signal Transduction/physiology , Small-Conductance Calcium-Activated Potassium Channels/metabolism , Adult , Aged , Aged, 80 and over , Amino Acid Sequence , Animals , Calcium Signaling/drug effects , Calcium Signaling/physiology , Cells, Cultured , Cerebral Cortex/metabolism , Cytokines/pharmacology , Hippocampus/metabolism , Humans , Mice , Mice, Inbred C57BL , Molecular Sequence Data , Molecular Weight , Protein Isoforms/genetics , Protein Isoforms/isolation & purification , Protein Isoforms/metabolism , RNA, Messenger/metabolism , Rats , Small-Conductance Calcium-Activated Potassium Channels/genetics , Small-Conductance Calcium-Activated Potassium Channels/isolation & purification
12.
CNS Drug Rev ; 13(4): 423-43, 2007.
Article in English | MEDLINE | ID: mdl-18078427

ABSTRACT

Corticotropin-releasing factor (CRF) is a neuropeptide and mediating component of neuroendocrine, autonomic, and behavioral processes associated with the stress response. The two receptor subtypes identified in the mammalian brain, CRF receptor subtype 1 (CRF1) and CRF2, are suggested to differentially modulate these processes. Manipulation of these receptors with selective CRF compounds and transgenic models has revealed, in most studies, a clear potentiation of the stress response through central activation of CRF1. However, pharmacological activation of CRF restricted to CRF1 has been limited by the availability of selective peptidic compounds. Recently, a highly selective CRF1 agonist, cortagine, has been developed. It was synthesized from chimeric intermediate sequences of ovine CRF, sauvagine, and human/rat CRF into a highly soluble peptide with strong affinity for CRF1 (IC(50) < 5 nM) and a very low binding preference for CRF2 (IC(50) > 500 nM). Affinity for the CRF binding protein (IC(50) > 1,000 nM) can be abolished by the addition of a glutamate residue on position 21 of the cortagine peptide sequence. Cortagine has recently been tested in a variety of preclinical models of behavior including the elevated-plus-maze (EPM), forced swim test (FST), homecage, and rat exposure test (RET). Preliminary characterization in the EPM and FST suggested that this compound elicits anxiogenic and antidepressant-like effects, respectively. Additional testing in the homecage and RET, which targets various elements of behavior, directs to a more potent anxiogenic profile of cortagine. In this review, we discuss the behavioral findings and the tests used to measure these effects. Finally, we also discuss preliminary findings of autonomic activation obtained by central injection of cortagine that support CRF1 involvement in the modulation of heart rate and heart rate variability.


Subject(s)
Autonomic Nervous System/drug effects , Behavior, Animal/drug effects , Corticotropin-Releasing Hormone/pharmacology , Receptors, Corticotropin-Releasing Hormone/agonists , Recombinant Fusion Proteins/pharmacology , Animals , Humans
13.
Eur J Neurosci ; 25(11): 3385-97, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17553007

ABSTRACT

The objective of this study was to investigate the role of corticotropin-releasing factor receptors 1 (CRF(1)) and 2 (CRF(2)) in anxiety-like behavior and learning of C57BL/6J mice after exposure to a stressful stimulus. When C57BL/6J mice were exposed to immobilization (1 h) serving as stressful stimulus, context- and tone-dependent fear conditioning were impaired if the training followed immediately after immobilization. The stress-induced impairment of context-dependent fear conditioning was prevented by specific blockade of CRF(2) of the lateral septum (LS) with anti-sauvagine-30. Immobilization did not only affect conditioned fear, but also enhanced, through CRF(2) of the LS, anxiety-like behavior determined with the elevated plus maze. Recovery from stress-induced anxiety and impairment of context-dependent fear conditioning was observed after 1 h delay of training and required hippocampal CRF(1), as indicated by the finding that this recovery was prevented by blockade of intrahippocampal CRF(1). It was concluded that exposure to a stressor initially affected both anxiety-like behavior and contextual conditioned fear through septal CRF(2), while the later activation of hippocampal CRF(1) resulted in the return to baseline levels of both processes. Intraventricular injection of mouse urocortin 2, a CRF(2)-selective agonist, removed the stress-induced anxiety and learning impairment, but did not reduce the activation of the hypothalamic pituitary adrenal axis indicative of the hormonal stress response. We propose that the enhanced anxiety is the component of the stress response responsible for the memory deficit.


Subject(s)
Anxiety/etiology , Memory Disorders/etiology , Receptors, Corticotropin-Releasing Hormone/physiology , Stress, Physiological/complications , Adrenocorticotropic Hormone/metabolism , Amphibian Proteins , Animals , Antibodies/pharmacology , Anxiety/drug therapy , Anxiety/pathology , Autoradiography , Behavior, Animal , Conditioning, Classical/drug effects , Conditioning, Classical/physiology , Corticotropin-Releasing Hormone/pharmacology , Dose-Response Relationship, Drug , Fear , Immobilization/methods , Male , Maze Learning/drug effects , Memory Disorders/drug therapy , Memory Disorders/pathology , Mice , Mice, Inbred C57BL , Peptide Fragments/pharmacology , Peptide Hormones , Peptides/immunology , Receptors, Corticotropin-Releasing Hormone/agonists , Receptors, Corticotropin-Releasing Hormone/antagonists & inhibitors , Septal Nuclei/drug effects , Stress, Physiological/etiology , Time Factors , Urocortins
14.
Mol Cell Biochem ; 300(1-2): 9-17, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17396235

ABSTRACT

Small-conductance Ca(2+)-activated K(+) channels (SK) of the SK2 subtype are widely expressed in the central nervous system where they contribute to the control of neuronal excitability. Two SK2 isoforms, SK2-S and SK2-L, the latter representing an N-terminally extended protein of SK2-S, are expressed in similar patterns in the brain. However, our understanding of mechanisms by which the expression of SK2 is regulated is limited. We identified one functional glucocorticoid response element (GRE) at position -2248 bp and two functional nuclear factor-kappB (NF-kappaB) response elements at positions -1652 and -1586 bp in the SK2-S promoter. An increase in SK2-S promoter activity was observed in PC12 cells transiently transfected with a wild-type SK2-S promoter-luciferase reporter gene construct and treated with aldosterone or dexamethasone. The mineralocorticoid receptor (MR) antagonist spironolactone or the glucocorticoid receptor (GR) antagonist mifepristone fully inhibited aldosterone or dexamethasone activation of the SK2-S promoter, respectively. SK2-S promoter activity was also induced by the cell-permeable ceramide analog, N-acetylsphingosine (C2-ceramide). Antisense oligonucleotides directed to NF-kappaB p65 or p50 suppressed SK2-S transcription induced by C2-ceramide. Deletion studies showed that only the -1586 bp NF-kappaB binding site was necessary for maximum C2-ceramide response. Finally, we showed that activation of GRs but not of MRs repressed the NF-kappaB-mediated induction of SK2-S transcription. These findings suggest a possible transcriptional cross talk between GRs and NF-kappaB in the intronic promoter regulation of SK2-S channel gene transcription.


Subject(s)
Corticosterone/pharmacology , Gene Expression Regulation/drug effects , Introns/genetics , NF-kappa B/metabolism , Promoter Regions, Genetic/genetics , Small-Conductance Calcium-Activated Potassium Channels/genetics , Transcription, Genetic/drug effects , Animals , Binding Sites , Cloning, Molecular , Luciferases/metabolism , Mice , NF-kappa B p50 Subunit/metabolism , PC12 Cells , Rats , Receptors, Glucocorticoid/metabolism , Transcription Factor RelA/metabolism
15.
Cardiovasc Res ; 69(2): 402-11, 2006 Feb 01.
Article in English | MEDLINE | ID: mdl-16386238

ABSTRACT

OBJECTIVE: Urocortin II (UcnII), a peptide of the corticotropin-releasing factor (CRF) family, exerts profound actions on the cardiovascular system. Direct effects of UcnII on adult cardiomyocytes have not been evaluated before. Our aim was to characterize functional effects of UcnII on cardiomyocytes and to elucidate the underlying signaling pathway(s) and cellular mechanisms. METHODS: Rabbit ventricular cardiomyocytes were stimulated at 0.5 Hz (22-25 degrees C). Unloaded cell shortening (FS, edge detection), [Ca(2+)](i) transients (Fluo-4), and L-type Ca(2+) currents (I(Ca), whole-cell patch clamping) were measured. Sarcoplasmic reticulum (SR) Ca(2+) load was assessed by rapid application of caffeine (20 mmol/L). RESULTS: UcnII increased cell shortening and accelerated relaxation in a time- and concentration-dependent manner (EC(50): 10.7 nmol/L). The inotropic effect of UcnII was maximal at 100 nmol/L (35%+/-11% increase in FS, n=8, P<0.05). The inotropic and lusitropic actions of UcnII were largely eliminated by inhibition of CRF(2) receptors (10 nmol/L antisauvagine-30, n=5) or protein kinase A (PKA, 500 nmol/L H-89, n=5). UcnII increased [Ca(2+)](i) transient amplitude (by 63%+/-35%, n=7, P<0.05) and decreased the time constant for decay (from 800+/-63 to 218+/-27 ms, n=7, P<0.001). UcnII also increased SR Ca(2+) load (by 19%+/-7%, n=7, P<0.05) and fractional Ca(2+) release (from 57%+/-7% to 98%+/-2%, n=7, P<0.01). I(Ca) was augmented by 32.7%+/-10.0% (n=9, P<0.05) and the I(Ca)-V relationship was shifted by -15 mV during UcnII treatment. CONCLUSION: UcnII exerts positive inotropic and lusitropic effects in cardiomyocytes via CRF(2) receptor-mediated stimulation of PKA which augments I(Ca) and SR Ca(2+) load to increase SR Ca(2+) release and [Ca(2+)](i) transients.


Subject(s)
Corticotropin-Releasing Hormone/pharmacology , Cyclic AMP-Dependent Protein Kinases/metabolism , Myocytes, Cardiac/drug effects , Receptors, Corticotropin-Releasing Hormone/metabolism , Animals , Enzyme Activation , Heart Ventricles , Microscopy, Confocal , Myocardial Contraction/drug effects , Myocytes, Cardiac/metabolism , Patch-Clamp Techniques , Peptide Fragments/pharmacology , Rabbits , Stimulation, Chemical , Urocortins
16.
Mini Rev Med Chem ; 5(10): 953-60, 2005 Oct.
Article in English | MEDLINE | ID: mdl-16250837

ABSTRACT

The actions of the neuropeptide corticotropin-releasing factor (CRF) are modulated by a CRF binding protein (CRFBP). In view of the memory-enhancing effects of CRF, the release of endogenous CRF from CRFBP by CRFBP inhibitors has been suggested as a therapeutical strategy for the treatment of cognitive deficits. This mini-review will summarize recent advances in the field with a focus on the pharmaceutical potential of CRFBP inhibitors.


Subject(s)
Corticotropin-Releasing Hormone/metabolism , Receptors, Corticotropin-Releasing Hormone/metabolism , Amino Acid Sequence , Animals , Binding Sites , Carrier Proteins/antagonists & inhibitors , Carrier Proteins/chemistry , Cognition Disorders/drug therapy , Corticotropin-Releasing Hormone/chemistry , Humans , Ligands , Molecular Sequence Data , Receptors, Corticotropin-Releasing Hormone/chemistry , Urocortins
17.
FEBS Lett ; 579(20): 4259-64, 2005 Aug 15.
Article in English | MEDLINE | ID: mdl-16054139

ABSTRACT

Macrophages undergo apoptosis as a mechanism of regulating their activation and the inflammatory reaction. Macrophages express the Corticotropin-Releasing Factor Receptor-2 (CRFR2) the endogenous agonists of which, the urocortins, are also present at the site of inflammation. We have found that urocortins induced macrophage apoptosis in a dose- and time-dependent manner via CRFR2. In contrast to lipopolysaccharide (LPS)-induced apoptosis, the pro-apoptosis pathway activated by urocortins involved the pro-apoptotic Bax and Bad proteins and not nitric oxide, JNK and p38MAPK characteristic of LPS. In conclusion, our data suggest that endogenous CRFR2 ligands exert an anti-inflammatory effect via induction of macrophage apoptosis.


Subject(s)
Apoptosis , Corticotropin-Releasing Hormone/pharmacology , Macrophages/drug effects , Receptors, Corticotropin-Releasing Hormone/metabolism , Animals , Carrier Proteins/metabolism , Cells, Cultured , Inflammation/metabolism , JNK Mitogen-Activated Protein Kinases/metabolism , Lipopolysaccharides/pharmacology , Macrophages/metabolism , Mice , Polysaccharides, Bacterial/pharmacology , Proto-Oncogene Proteins c-bcl-2/metabolism , Receptors, Corticotropin-Releasing Hormone/antagonists & inhibitors , Signal Transduction , Urocortins , bcl-2-Associated X Protein , bcl-Associated Death Protein , p38 Mitogen-Activated Protein Kinases/metabolism
18.
Neurosci Biobehav Rev ; 29(8): 1361-73, 2005.
Article in English | MEDLINE | ID: mdl-16120463

ABSTRACT

Cholecystokinin (CCK) is a neurotransmitter in the brain closely related to anxiety. Of the two CCK receptor subtypes, CCK(2) receptors are most implicated in the control of anxiety-related behavior. CCK(2) receptor activation causes anxiogenic effects while the blockade of this receptor has anxiolytic effects. This review focuses on the molecular mechanisms of CCK(2) receptors underlying anxiety-related behaviors of PVG hooded and Spraque-Dawley (SD) rats in two anxiety models (elevated plus-maze [EPM] and cat exposure test). PVG hooded rats showed prolonged freezing behavior in the cat exposure test while SD rats showed very low levels of freezing. A CCK(2) receptor antagonist (LY225910) attenuated freezing behavior in PVG hooded rats while a CCK(2) receptor agonist (CCK-4) increased freezing behavior in SD rats. In contrast, the two strains behaved similarly on the EPM. CCK-4 caused a pronounced anxiogenic effect in PVG hooded rats but only a slight effect in SD rats. CCK(2) antagonists also showed more pronounced anxiolytic effects in PVG hooded rats than in SD rats. CCK(2) receptor expression was greater in PVG hooded than in SD rats in the cortex and hippocampus. Genetic studies also demonstrated four differences in the DNA sequence of the CCK(2) receptor gene between the two rat strains.


Subject(s)
Anxiety/physiopathology , Brain/metabolism , Receptor, Cholecystokinin B/physiology , Animals , Behavior, Animal , Brain/anatomy & histology , Brain/physiopathology , Cats , Disease Models, Animal , Gene Expression Regulation/physiology , Maze Learning/physiology , Mice , Mice, Knockout , Neurotransmitter Agents/metabolism , Rats , Receptor, Cholecystokinin B/deficiency , Receptor, Cholecystokinin B/genetics
19.
Neurosci Biobehav Rev ; 29(8): 1323-33, 2005.
Article in English | MEDLINE | ID: mdl-16099044

ABSTRACT

Corticotropin-releasing factor (CRF), a 41 amino acid peptide exhibits its actions through two pharmacologically distinct CRF receptor subtypes CRF(1) and CRF(2). Regulation of the relative contribution of the two CRF receptors to central CRF activity may be essential in coordinating physiological responses to stress. To facilitate the analysis of their differential involvement, we recently developed a CRF(1)-selective agonist cortagine by synthesis of chimeric peptides derived from human/rat CRF, ovine CRF, and sauvagine. Cortagine was analyzed in behavioral experiments using male wild type and CRF(2)-deficient C57BL/6J mice for its action on anxiety- and depression-like behaviors. In contrast to the current hypothesis that increased CRF(1) activity facilitates the expression of anxiety- and depression-like behavior, cortagine combines anxiogenic properties with antidepressant effects. In this article, we show that antidepressant effects are partially mediated by CRF(1) of the dorsal hippocampus. Possible pathways responsible for the paradoxical antidepressant effects observed after CRF(1) activation are discussed.


Subject(s)
Anxiety/metabolism , Depression/metabolism , Receptors, Corticotropin-Releasing Hormone/physiology , Animals , Behavior, Animal , Corticotropin-Releasing Hormone/pharmacology , Corticotropin-Releasing Hormone/physiology , Dose-Response Relationship, Drug , Drug Administration Routes , Hippocampus/drug effects , Hippocampus/physiopathology , Humans , Mice , Mice, Knockout , Receptors, Corticotropin-Releasing Hormone/agonists , Receptors, Corticotropin-Releasing Hormone/deficiency , Recombinant Fusion Proteins/pharmacology
20.
Learn Mem ; 12(2): 138-43, 2005.
Article in English | MEDLINE | ID: mdl-15805311

ABSTRACT

The effects of stress (restraint plus tail shock) on hippocampus-dependent trace eyeblink conditioning and hippocampal excitability were examined in C57BL/6 male mice. The results indicate that the stressor significantly increased the concentration of circulating corticosterone, the amount and rate of learning relative to nonstressed conditioned mice, and the excitability of CA1 hippocampal pyramidal neurons. Behaviorally, there was no effect of the stressor on control mice that received unpaired presentations of the tone and periorbital shock, i.e., neither stressed nor nonstressed control mice showed an increase in conditioned responding that was above baseline levels. Biophysically, the stressor significantly decreased the amplitude of the post-burst afterhyperpolarization (AHP) and decreased spike frequency accommodation relative to cells from nonstressed control mice. The effect was significant for mice that were stressed either 1 h or 24 h earlier. The results suggest that the stressor increases the excitability of hippocampal pyramidal neurons and that the mechanism underlying this increase may contribute to the more rapid acquisition of hippocampally dependent eyeblink conditioning.


Subject(s)
Conditioning, Classical/physiology , Conditioning, Eyelid/physiology , Pyramidal Cells/physiology , Stress, Psychological/physiopathology , Action Potentials/physiology , Acute Disease , Analysis of Variance , Animals , Association Learning/physiology , Corticosterone/blood , Disease Models, Animal , Hippocampus/cytology , Hippocampus/physiology , Long-Term Potentiation/physiology , Male , Mice , Stress, Psychological/blood
SELECTION OF CITATIONS
SEARCH DETAIL
...