sauvagine and Disease-Models--Animal

sauvagine has been researched along with Disease-Models--Animal* in 7 studies

Other Studies

7 other study(ies) available for sauvagine and Disease-Models--Animal

ArticleYear
Comparison of CRF-immunoreactive neurons distribution in mouse and rat brains and selective induction of Fos in rat hypothalamic CRF neurons by abdominal surgery.
    Brain research, 2011, Sep-30, Volume: 1415

    Mice and rats are widely used in stress-related behavioral studies while little is known about the distribution of the stress hormone, corticotropin-releasing factor (CRF) in the mouse brain. We developed and characterized a novel rat/mouse CRF polyclonal antibody (CURE ab 200101) that was used to detect and compare the brain distributions of CRF immunoreactivity in naïve and colchicine-treated rats and mice. We also assessed whether the visceral stressor of abdominal surgery activated brain CRF neurons using double labeling of Fos/CRF in naïve rats. CRF-ir neurons were visualized in the cortex, bed nucleus of the stria terminalis, central amygdala, hypothalamic paraventricular nucleus (PVN), Barrington's nucleus and dorsolateral tegmental area in naïve rats. CRF-immunoreactive (ir) neurons in the mouse brain were detected only after colchicine. The pattern shows fundamental similarity compared to the colchicine-treated rat brain, however, there were differences with a lesser distribution in both areas and density except in the lateral septum and external subnucleus of the lateral parabrachial nucleus which contained more CRF-ir neurons in mice, and CRF-ir neurons in the dorsal motor nucleus of the vagus were found only in mice. Abdominal surgery in naïve rats induced Fos-ir in 30% of total CRF-ir neurons in the PVN compared with control (anesthesia alone) while Fos was not co-localized with CRF in other brain nuclei. These data indicate that CRF-ir distribution in the brain displays similarity as well as distinct features in mice compared to rats that may underlie some differential stress responses. Abdominal surgery activates CRF-ir neurons selectively in the PVN of rats without colchicine treatment.

    Topics: Abdomen; Amphibian Proteins; Animals; Brain; Cell Count; Colchicine; Corticotropin-Releasing Hormone; Disease Models, Animal; Gene Expression Regulation; Mice; Neurons; Oncogene Proteins v-fos; Peptide Hormones; Postoperative Complications; Radioimmunoassay; Rats; Species Specificity; Stress, Psychological; Tubulin Modulators; Urotensins

2011
Amygdaloid pERK1/2 in corticotropin-releasing hormone overexpressing mice under basal and acute stress conditions.
    Neuroscience, 2009, Mar-17, Volume: 159, Issue:2

    Corticotropin-releasing hormone (CRH) coordinates neuroendocrine and behavioral adaptations to stress. Acute CRH administration in vivo activates extracellular signal-regulated kinase 1/2 (ERK1/2) in limbic brain areas, acting through the CRH receptor type 1 (CRH-R1). In the present study, we used CRH-COE-Cam mice that overexpress CRH in limbic-restricted areas, to analyze the effect of chronic CRH overexpression on ERK1/2 activation. By immunohistochemistry and confocal microscopy analysis we found that pERK1/2 levels in the basolateral amygdala (BLA) were similar in control and CRH overexpressing mice under basal conditions. Acute stress caused comparably increased levels of corticosterone in both control (CRH-COEcon-Cam) and CRH overexpressing (CRH-COEhom-Cam) animals. CRH-COEhom-Cam mice after stress showed reduced pERK1/2 immunoreactivity in the BLA compared to CRH-COEhom-Cam animals under basal conditions. Radioligand binding and in situ hybridization revealed higher density of CRH-R1 in the amygdala of CRH-COEhom mice under basal conditions compared to control littermates. A significant reduction of the receptor levels was observed in this area after acute stress, suggesting that stress may trigger CRH-R1 internalization/downregulation in these CRH overexpressing mice. Chronic CRH overexpression leads to reduced ERK1/2 activation in response to acute stress in the BLA.

    Topics: Amphibian Proteins; Amygdala; Animals; Autoradiography; Corticosterone; Corticotropin-Releasing Hormone; Disease Models, Animal; Gene Expression Regulation, Enzymologic; Iodine Isotopes; Male; Mice; Mice, Transgenic; Mitogen-Activated Protein Kinase 3; Peptide Hormones; Protein Binding; Radioimmunoassay; Receptors, Corticotropin-Releasing Hormone; Restraint, Physical; Stress, Psychological; Time Factors

2009
3-(4-Chloro-2-morpholin-4-yl-thiazol-5-yl)-8-(1-ethylpropyl)-2,6-dimethyl-imidazo[1,2-b]pyridazine: a novel brain-penetrant, orally available corticotropin-releasing factor receptor 1 antagonist with efficacy in animal models of alcoholism.
    The Journal of neuroscience : the official journal of the Society for Neuroscience, 2007, Mar-07, Volume: 27, Issue:10

    We describe a novel corticotropin-releasing factor receptor 1 (CRF1) antagonist with advantageous properties for clinical development, and its in vivo activity in preclinical alcoholism models. 3-(4-Chloro-2-morpholin-4-yl-thiazol-5-yl)-8-(1-ethylpropyl)-2,6-dimethyl-imidazo[1,2-b]pyridazine (MTIP) inhibited 125I-sauvagine binding to rat pituitary membranes and cloned human CRF1 with subnanomolar affinities, with no detectable activity at the CRF2 receptor or other common drug targets. After oral administration to rats, MTIP inhibited 125I-sauvagine binding to rat cerebellar membranes ex vivo with an ED50 of approximately 1.3 mg/kg and an oral bioavailability of 91.1%. Compared with R121919 (2,5-dimethyl-3-(6-dimethyl-4-methylpyridin-3-yl)-7-dipropylamino-pyrazolo[1,5-a]pyrimidine) and CP154526 (N-butyl-N-ethyl-4,9-dimethyl-7-(2,4,6-trimethylphenyl)-3,5,7-triazabicyclo[4.3.0]nona-2,4,8,10-tetraen-2-amine), MTIP had a markedly reduced volume of distribution and clearance. Neither open-field activity nor baseline exploration of an elevated plus-maze was affected by MTIP (1-10 mg/kg). In contrast, MTIP dose-dependently reversed anxiogenic effects of withdrawal from a 3 g/kg alcohol dose. Similarly, MTIP blocked excessive alcohol self-administration in Wistar rats with a history of dependence, and in a genetic model of high alcohol preference, the msP rat, at doses that had no effect in nondependent Wistar rats. Also, MTIP blocked reinstatement of stress-induced alcohol seeking both in postdependent and in genetically selected msP animals, again at doses that were ineffective in nondependent Wistar rats. Based on these findings, MTIP is a promising candidate for treatment of alcohol dependence.

    Topics: Administration, Oral; Alcohol Drinking; Alcoholism; Amphibian Proteins; Animals; Anxiety; Behavior, Animal; Brain; Cerebellum; Disease Models, Animal; Dose-Response Relationship, Drug; Ethanol; Male; Peptide Hormones; Peptides; Pituitary Gland; Pyridazines; Rats; Rats, Sprague-Dawley; Rats, Wistar; Receptors, Corticotropin-Releasing Hormone; Self Administration; Stress, Physiological; Substance Withdrawal Syndrome; Thiazoles

2007
A centrally acting, anxiolytic angiotensin II AT1 receptor antagonist prevents the isolation stress-induced decrease in cortical CRF1 receptor and benzodiazepine binding.
    Neuropsychopharmacology : official publication of the American College of Neuropsychopharmacology, 2006, Volume: 31, Issue:6

    Long-term pretreatment with an angiotensin II AT1 antagonist blocks angiotensin II effects in brain and peripheral organs and abolishes the sympathoadrenal and hypothalamic-pituitary-adrenal responses to isolation stress. We determined whether AT1 receptors were also important for the stress response of higher regulatory centers. We studied angiotensin II and corticotropin-releasing factor (CRF) receptors and benzodiazepine binding sites in brains of Wistar Hannover rats. Animals were pretreated for 13 days with vehicle or a central and peripheral AT1 antagonist (candesartan, 0.5 mg/kg/day) via osmotic minipumps followed by 24 h of isolation in metabolic cages, or kept grouped throughout the study (grouped controls). In another study, we determined the influence of a similar treatment with candesartan on performance in an elevated plus-maze. AT1 receptor blockade prevented the isolation-induced increase in brain AT1 receptors and decrease in AT2 binding in the locus coeruleus. AT1 receptor antagonism also prevented the increase in tyrosine hydroxylase mRNA in the locus coeruleus. Pretreatment with the AT1 receptor antagonist completely prevented the decrease in cortical CRF1 receptor and benzodiazepine binding produced by isolation stress. In addition, pretreatment with candesartan increased the time spent in and the number of entries to open arms of the elevated plus-maze, measure of decreased anxiety. Our results implicate a modulation of upstream neurotransmission processes regulating cortical CRF1 receptors and the GABA(A) complex as molecular mechanisms responsible for the anti-anxiety effect of centrally acting AT1 receptor antagonists. We propose that AT1 receptor antagonists can be considered as compounds with possible therapeutic anti-stress and anti-anxiety properties.

    Topics: Amphibian Proteins; Analysis of Variance; Angiotensin II; Angiotensin II Type 1 Receptor Blockers; Animals; Autoradiography; Behavior, Animal; Benzimidazoles; Benzodiazepines; Biphenyl Compounds; Cerebral Cortex; Disease Models, Animal; Flunitrazepam; GABA Modulators; In Situ Hybridization; Male; Maze Learning; Peptide Hormones; Peptides; Protein Binding; Pyrimidines; Pyrroles; Rats; Receptor, Angiotensin, Type 2; Receptors, Corticotropin-Releasing Hormone; RNA, Messenger; Social Isolation; Stress, Physiological; Tetrazoles; Tyrosine 3-Monooxygenase

2006
Discovery of corticotropin releasing factor 2 receptor selective sauvagine analogues for treatment of skeletal muscle atrophy.
    Journal of medicinal chemistry, 2005, Jan-13, Volume: 48, Issue:1

    The corticotropin release factor 2 receptor (CRF2R) has many biological activities including modulation of the stress response. Recently, we have demonstrated that CRF2R activation functions to prevent skeletal muscle wasting resulting from a variety of physiological stimuli. Thus we are interested in identifying CRF2R selective agonists with optimal pharmacological properties for use in treating muscle wasting diseases. Several CRF2R agonists are known including the frog peptide sauvagine (Svg), which display superior pharmacological properties compared to other CRF2R agonists. Unfortunately sauvagine is a nonselective CRFR agonist, thus making it of less utility due to side effects resulting from corticotropin release factor 1 receptor (CRF1R) activation. Because our initial modifications of Svg at position 11 improved CRF2R selectivity, we investigated the role of amino acids at positions 12 and 13 in Svg. We observed that phenylalanine, leucine, isoleucine, threonine, glutamine, histidine, and tyrosine at the 12th position were the strongest promoters of CRF2R selectivity whereas phenylalanine, glutamine, trytophane, tyrosine, valine, isoleucine, leucine, and 2-naphthylalanine were the preferred residues at the 13th position. Selective sauvagine peptides demonstrated improved antiatrophy effects in a mouse-casting model when compared to sauvagine itself. Thus, we demonstrate that the CRF2R selectivity can be improved by optimizing amino acids at positions 12 and 13 (all with proline at position 11) and that the selective sauvagine analogues demonstrate better in vivo efficacy than sauvagine itself.

    Topics: Amino Acid Substitution; Amphibian Proteins; Animals; Disease Models, Animal; Drug Evaluation, Preclinical; Mice; Muscular Disorders, Atrophic; Peptide Hormones; Peptides; Receptors, Corticotropin-Releasing Hormone; Structure-Activity Relationship; Tibia

2005
Receptor occupancy of nonpeptide corticotropin-releasing factor 1 antagonist DMP696: correlation with drug exposure and anxiolytic efficacy.
    The Journal of pharmacology and experimental therapeutics, 2003, Volume: 305, Issue:1

    4-(1,3-Dimethoxyprop-2-ylamine)-2,7-dimethyl-8-(2,4-dichlorophenyl)-pyrazolo[1,5-a]-1,3,5-triazine (DMP696) is a highly selective and potent, nonpeptide corticotropin-releasing factor 1 (CRF(1)) antagonist. In this study, we measured in vivo CRF(1) receptor occupancy of DMP696 by using ex vivo ligand binding and quantitative autoradiography and explored the relationship of receptor occupancy with plasma and brain exposure and behavioral efficacy. In vitro affinity (IC(50)) of DMP696 to brain CRF(1) receptors measured using the brain section binding autoradiography in this study is similar to that assessed using homogenized cell membrane assays previously. The ex vivo binding assay was validated by demonstrating that potential underestimation of receptor occupancy with this procedure could be minimized by identifying an appropriate in vitro incubation time (40 min) based upon the dissociation kinetics of DMP696. Orally administrated DMP696 dose dependently occupied CRF(1) receptors in the brain, with ~60% occupancy at 3 mg/kg. In the defensive withdrawal test of anxiety, this dose of DMP696 produced approximately 50% reduction in the exit latency. The time course of plasma and brain drug levels paralleled that of receptor occupancy, with peak exposure at 90 min after dosing. The plasma-free concentration of DMP696 corresponding to 50% CRF(1) receptor occupancy (in vivo IC(50), 1.22 nM) was similar to the in vitro IC(50) (~1.0 nM). Brain concentrations of DMP696 were over 150-fold higher than the plasma-free levels. In conclusion, doses of DMP696 occupying over 50% brain CRF(1) receptors are consistent with doses producing anxiolytic efficacy in the defense withdrawal test of anxiety, and the IC(50) value estimated in vivo based on plasma-free drug concentrations is consistent with the in vitro IC(50) value.

    Topics: Amphibian Proteins; Animals; Anxiety; Binding Sites; Corticotropin-Releasing Hormone; Disease Models, Animal; Drug Interactions; Iodine Radioisotopes; Male; Peptide Hormones; Peptides; Pyrazoles; Rats; Rats, Sprague-Dawley; Receptors, Corticotropin-Releasing Hormone; Statistics as Topic; Substance Withdrawal Syndrome; Time Factors; Triazines

2003
Use of selective mesenteric dilator peptides to examine the role of the intestine in experimental hemorrhagic shock.
    Circulatory shock, 1985, Volume: 17, Issue:3

    If mesenteric vasoconstriction sets in motion the chain of events that leads to shock, then the administration of a selective mesenteric vasodilator (which has no other known cardiovascular actions) should prevent, or at least modify, the hemodynamic events in a standardized shock preparation. We used the Wiggers model of experimental hemorrhagic shock in 20 pentobarbital-anesthetized dogs, giving half the dogs a selective mesenteric dilator peptide, sauvagine, to produce selective dilatation of the superior (cephalic) and inferior (caudal) mesenteric circulations. (Sauvagine does not dilate the coeliac vascular bed.) A third, sham-operated group of four dogs served as a time control. Sauvagine produced no observable beneficial effect in terms of hemodynamic measurements, intestinal oxygen kinetics, or intestinal histology. Since we took the precaution of delivering the peptide close-arterially (to ensure delivery during hypoperfusion), we conclude that mesenteric vasoconstriction per se is not the prime event in initiating shock. Since reduced cardiac output during hypovolemia led to diminished mesenteric flow (in spite of vasodilation), these experiments do not exclude the possibility of diminished intestinal blood flow being a central event in shock.

    Topics: Amphibian Proteins; Animals; Disease Models, Animal; Dogs; Hemodynamics; Intestines; Oxygen Consumption; Peptide Hormones; Peptides; Shock, Hemorrhagic; Splanchnic Circulation; Time Factors; Vasodilator Agents

1985